24.12.2012 Views

Annual Meeting Proceedings Part 1 - American Society of Clinical ...

Annual Meeting Proceedings Part 1 - American Society of Clinical ...

Annual Meeting Proceedings Part 1 - American Society of Clinical ...

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

Volume 30, Issue 15S, <strong>Part</strong> I <strong>of</strong> II May 20, 2012<br />

JOURNAL<br />

CLINICAL<br />

OF<br />

ONCOLOGY<br />

Official Journal <strong>of</strong> the<br />

<strong>American</strong> <strong>Society</strong> <strong>of</strong> <strong>Clinical</strong> Oncology<br />

2012 ASCO <strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong><br />

www.jco.org<br />

48th <strong>Annual</strong> <strong>Meeting</strong><br />

June 1-5, 2012<br />

McCormick Place<br />

Chicago, IL


48th<br />

<strong>Annual</strong> <strong>Meeting</strong> <strong>of</strong> the<br />

<strong>American</strong> <strong>Society</strong> <strong>of</strong> <strong>Clinical</strong> Oncology<br />

June 1–5, 2012<br />

Chicago, Illinois<br />

2012 <strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong> <strong>Part</strong> I<br />

(a supplement to the Journal <strong>of</strong> <strong>Clinical</strong> Oncology)<br />

Copyright 2012 <strong>American</strong> <strong>Society</strong> <strong>of</strong> <strong>Clinical</strong> Oncology


Editor: Michael A. Carducci, MD<br />

Managing Editor: Jennifer Giblin<br />

Editorial Coordinator: Lauren Burke<br />

Production Manager: Donna Dottellis<br />

Executive Editor: Lisa Greaves<br />

Requests for permission to reprint abstracts should be directed to Intellectual Property<br />

Rights Manager, <strong>American</strong> <strong>Society</strong> <strong>of</strong> <strong>Clinical</strong> Oncology, 2318 Mill Road, Suite 800,<br />

Alexandria, VA 22314. Tel: 571-483-1300; Fax: 571-366-9530; Email: permissions@asco.org.<br />

Editorial correspondence and production questions should be addressed to Managing Editor,<br />

<strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong>, <strong>American</strong> <strong>Society</strong> <strong>of</strong> <strong>Clinical</strong> Oncology, 2318 Mill Road, Suite<br />

800, Alexandria, VA 22314. Email: abstracts@asco.org.<br />

Copyright © 2012 <strong>American</strong> <strong>Society</strong> <strong>of</strong> <strong>Clinical</strong> Oncology. All rights reserved. No part <strong>of</strong> this<br />

publication may be reproduced or transmitted in any form or by any means, electronic or<br />

mechanical, including photocopy, recording, or any information storage and retrieval system,<br />

without written permission by the <strong>Society</strong>.<br />

The <strong>American</strong> <strong>Society</strong> <strong>of</strong> <strong>Clinical</strong> Oncology assumes no responsibility for errors or<br />

omissions in this document. The reader is advised to check the appropriate medical literature<br />

and the product information currently provided by the manufacturer <strong>of</strong> each drug to be<br />

administered to verify the dosage, the method and duration or administration, or<br />

contraindications. It is the responsibility <strong>of</strong> the treating physician or other health care<br />

pr<strong>of</strong>essional, relying on independent experience and knowledge <strong>of</strong> the patient, to determine<br />

drug, disease, and the best treatment for the patient.<br />

Abstract management and indexing provided by The Conference Exchange, Cumberland,<br />

RI. Composition services and print production provided by Cenveo Publisher Services,<br />

Richmond, VA.


Volume 30, Issue 15S Supplement to May 20, 2012<br />

JOURNAL OF<br />

CLINICAL<br />

ONCOLOGY<br />

CONTENTS<br />

Official Journal <strong>of</strong> the<br />

<strong>American</strong> <strong>Society</strong> <strong>of</strong> <strong>Clinical</strong> Oncology<br />

2012 ASCO ANNUAL MEETING PROCEEDINGS<br />

Special Award Lecture Abstracts ............................................................................................................. 1s<br />

Plenary (Abstracts LBA1 – 4) ....................................................................................................................... 5s<br />

Breast Cancer—HER2/ER<br />

Scheduled presentations (Abstracts 500 – TPS670) ........................................................................................ 7s<br />

Breast Cancer—Triple-Negative/Cytotoxics/Local Therapy<br />

Scheduled presentations (Abstracts LBA1000 – TPS1148) ............................................................................... 49s<br />

Cancer Prevention/Epidemiology<br />

Scheduled presentations (Abstracts 1500 – TPS1616) ....................................................................................... 86s<br />

Central Nervous System Tumors<br />

Scheduled presentations (Abstracts 2000 – TPS2107) ..................................................................................... 115s<br />

Developmental Therapeutics—<strong>Clinical</strong> Pharmacology and Immunotherapy<br />

Scheduled presentations (Abstracts 2500^ – TPS2622) .................................................................................. 142s<br />

Developmental Therapeutics—Experimental Therapeutics<br />

Scheduled presentations (Abstracts 3000 – TPS3118) ...................................................................................... 173s<br />

Gastrointestinal (Colorectal) Cancer<br />

Scheduled presentations (Abstracts LBA3500^ – TPS3643) .......................................................................... 203s<br />

Gastrointestinal (Noncolorectal) Cancer<br />

Scheduled presentations (Abstracts LBA4000 – TPS4153) ............................................................................. 239s<br />

Genitourinary Cancer<br />

Scheduled presentations (Abstracts 4500 – TPS4701) ..................................................................................... 277s<br />

continued on following page<br />

Journal <strong>of</strong> <strong>Clinical</strong> Oncology (ISSN 0732-183X) is published 36 times a year, three times monthly, by the <strong>American</strong> <strong>Society</strong> <strong>of</strong> <strong>Clinical</strong> Oncology, 2318 Mill Road, Suite 800,<br />

Alexandria, VA 22314. Periodicals postage is paid at Alexandria, VA, and at additional mailing <strong>of</strong>fices. Publication Mail Agreement Number 863289.<br />

Editorial correspondence should be addressed to Stephen A. Cannistra, MD, Journal <strong>of</strong> <strong>Clinical</strong> Oncology, 2318 Mill Road, Suite 800, Alexandria, VA 22314. Phone:<br />

703-797-1900; Fax: 703-684-8720. E-mail: jco@asco.org. Internet: www.jco.org.<br />

POSTMASTER: ASCO members should send changes <strong>of</strong> address to <strong>American</strong> <strong>Society</strong> <strong>of</strong> <strong>Clinical</strong> Oncology, 2318 Mill Road, Suite 800, Alexandria, VA 22314.<br />

Nonmembers should send changes <strong>of</strong> address to Journal <strong>of</strong> <strong>Clinical</strong> Oncology Customer Service, 2318 Mill Road, Suite 800, Alexandria, VA 22314.<br />

2012 annual subscription rates, effective September 1, 2011: United States and possessions: individual, $578 one year, $1,098 two years; single issue, $40. International: individual,<br />

$802 one year, $1,524 two years; single issue, $50. Institutions: bundled (print � online): Tier 1: $874 US, $1,209 Int’l; Tier 2: $1,019 US, $1,351 Int’l; Tier 3: $1,470 US, $1,792<br />

Int’l; Tier 4: contact JCO for quote. Institutions: online only, worldwide: Tier 1, $749; Tier 2: $873; Tier 3, $1,261; Tier 4: contact JCO for quote. See www.jco.org/ratecard for<br />

descriptions <strong>of</strong> each tier. Student and resident: United States and possessions: $289; all other countries, $401. To receive student/resident rate, orders must be accompanied by name<br />

<strong>of</strong> affiliated institution, date <strong>of</strong> term, and the signature <strong>of</strong> program/residency coordinator on institution letterhead. Orders will be billed at individual rate until pro<strong>of</strong> <strong>of</strong> status is<br />

received. Current prices are in effect for back volumes and back issues. Back issues sold in conjunction with a subscription rate are on a prorated basis. Subscriptions are accepted on a<br />

12-month basis. Prices are subject to change without notice. Single issues, both current and back, exist in limited quantities and are <strong>of</strong>fered for sale subject to availability. JCO Legacy<br />

Archive (electronic back issues from January 1983 through December 1998) is also available; please inquire.


Volume 30, Issue 15S Supplement to May 20, 2012<br />

...................................................................................................................................................<br />

Gynecologic Cancer<br />

Scheduled presentations (Abstracts LBA5000 – TPS5116) .............................................................................. 327s<br />

Head and Neck Cancer<br />

Scheduled presentations (Abstracts 5500 – TPS5602) .................................................................................... 356s<br />

Health Services Research<br />

Scheduled presentations (Abstracts 6000 – 6137) ............................................................................................ 382s<br />

Leukemia, Myelodysplasia, and Transplantation<br />

Scheduled presentations (Abstracts 6500^ – TPS6643) .................................................................................. 416s<br />

Lung Cancer—Non-small Cell Local-Regional/Small Cell/Other Thoracic Cancers<br />

Scheduled presentations (Abstracts 7000 – TPS7113) ...................................................................................... 452s<br />

Lung Cancer—Non-small Cell Metastatic<br />

Scheduled presentations (Abstracts LBA7500 – TPS7619) .............................................................................. 480s<br />

Lymphoma and Plasma Cell Disorders<br />

Scheduled presentations (Abstracts 8000 – TPS8118) ..................................................................................... 510s<br />

Melanoma/Skin Cancers<br />

Scheduled presentations (Abstracts LBA8500^ – TPS8606) .......................................................................... 540s<br />

Patient and Survivor Care<br />

Scheduled presentations (Abstracts 9000 – TPS9155) ..................................................................................... 567s<br />

Pediatric Oncology<br />

Scheduled presentations (Abstracts 9500 – TPS9597^) .................................................................................. 606s<br />

Sarcoma<br />

Scheduled presentations (Abstracts 10000 – TPS10103) .................................................................................. 630s<br />

Tumor Biology<br />

Scheduled presentations (Abstracts 10500 – TPS10635) ................................................................................. 656s<br />

Author Index ...................................................................................................................................................... 692s<br />

www.jco.org


<strong>American</strong> <strong>Society</strong> <strong>of</strong> <strong>Clinical</strong> Oncology<br />

48th <strong>Annual</strong> <strong>Meeting</strong><br />

2012 Abstracts<br />

Abstract Session Descriptions for Scheduled Presentations<br />

Oral Abstract Sessions<br />

Oral Abstract Sessions include didactic presentations <strong>of</strong> the abstracts <strong>of</strong> the highest scientific merit, as determined by<br />

the Scientific Program Committee. Experts in the field serve as Discussants to place the findings into perspective. The<br />

Plenary Session includes the abstracts selected by the Scientific Program Committee as having practice-changing<br />

findings.<br />

<strong>Clinical</strong> Science Symposia<br />

<strong>Clinical</strong> Science Symposia provide a forum for science in oncology, combining didactic lectures on a specific topic<br />

with the presentation <strong>of</strong> abstracts. Experts in the field classify studies on the basis <strong>of</strong> the strength <strong>of</strong> the evidence and<br />

critically discuss the conclusions in terms <strong>of</strong> their applicability to clinical practice.<br />

Poster Discussion Sessions<br />

Poster Discussion Sessions highlight selected abstracts <strong>of</strong> clinical research in poster format. The posters are grouped<br />

by topic and are on display for a specified time, followed by a discussion session in which experts provide<br />

commentary on the research findings.<br />

General Poster Sessions<br />

General Poster Sessions include selected abstracts <strong>of</strong> clinical research in poster format. The posters are grouped by<br />

topic and are on display for a specified time. Trials in Progress (TPS) abstract presentations are presented within a<br />

track’s general poster session.<br />

Publication-Only Abstracts<br />

Publication-only abstracts were selected to be published online in conjunction with the <strong>Annual</strong> <strong>Meeting</strong>, but not to be<br />

presented at the <strong>Meeting</strong>.<br />

All presented and publication-only abstracts are citable to this Journal <strong>of</strong> <strong>Clinical</strong> Oncology supplement. For citation<br />

examples, please see the Letter from the Editor.<br />

This publication contains abstracts selected by the ASCO Scientific Program Committee for presentation at the<br />

2012 <strong>Annual</strong> <strong>Meeting</strong>. Abstracts selected for electronic publication only are available in full-text versions<br />

online at abstract.asco.org and JCO.org. The type <strong>of</strong> session, the day, and the session start/end times are<br />

located to the right <strong>of</strong> the abstract number for scheduled presentations. To determine the location <strong>of</strong> the<br />

abstract session, refer to the <strong>Annual</strong> <strong>Meeting</strong> Program or the ePlanner, the online version <strong>of</strong> the <strong>Annual</strong><br />

<strong>Meeting</strong> Program, available at chicago2012.asco.org.<br />

Dates and times are subject to change.<br />

All modifications will be posted on ASCO.org.


Letter from the Editor<br />

The 2012 ASCO <strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong> <strong>Part</strong> I (a<br />

supplement to the Journal <strong>of</strong> <strong>Clinical</strong> Oncology) is<br />

an enduring record <strong>of</strong> the more than 2,700 abstracts<br />

selected by the ASCO Scientific Program Committee for<br />

presentation at the 48th <strong>Annual</strong> <strong>Meeting</strong> <strong>of</strong> the <strong>American</strong><br />

<strong>Society</strong> <strong>of</strong> <strong>Clinical</strong> Oncology, held June 1–5, in<br />

Chicago, Illinois. Accepted abstracts not presented at<br />

the meeting are available in full-text versions on<br />

ASCO.org and are included in the online supplement to<br />

the May 20 issue <strong>of</strong> Journal <strong>of</strong> <strong>Clinical</strong> Oncology at<br />

JCO.org.<br />

The majority <strong>of</strong> abstracts selected for presentation are<br />

included here in full and are categorized by scientific<br />

track. Abstracts are ordered numerically according to<br />

presentation type within a track. Presentation types<br />

include Oral Abstract Sessions, <strong>Clinical</strong> Science Symposia,<br />

Poster Discussion Sessions, and General Poster<br />

Sessions. Abstracts include the presenting author only.<br />

The full list <strong>of</strong> abstract authors and their disclosure<br />

information can be found online at abstract.asco.org.<br />

Certain abstracts are represented here by abstract title<br />

and presenting author only. These include <strong>Clinical</strong><br />

Review Abstracts (CRA), many <strong>of</strong> which will have important,<br />

and in some cases, immediate implications for<br />

patient care; Late-Breaking Abstracts (LBA), which<br />

feature results <strong>of</strong> phase III trials for which data were not<br />

available at the time <strong>of</strong> ASCO’s regular submission<br />

deadline; and Plenary Abstracts, which were selected by<br />

the Scientific Program Committee for their potential to<br />

have practice-changing results. The full-text versions <strong>of</strong><br />

abstracts within these three designations will be publically<br />

released on ASCO.org on the morning <strong>of</strong> their<br />

scheduled presentation at the <strong>Annual</strong> <strong>Meeting</strong> (abstracts<br />

scheduled for presentation on Tuesday will be<br />

released on Monday morning). These abstracts will also<br />

be included in the 2012 ASCO <strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong><br />

<strong>Part</strong> II, an online supplement to the June 20 issue <strong>of</strong><br />

Journal <strong>of</strong> <strong>Clinical</strong> Oncology on JCO.org. Print versions<br />

<strong>of</strong> these abstracts will be available onsite at the <strong>Annual</strong><br />

<strong>Meeting</strong> in the ASCO Daily News.<br />

All <strong>of</strong> the abstracts carry Journal <strong>of</strong> <strong>Clinical</strong> Oncology<br />

citations. The following are citation examples for print<br />

and electronic abstracts, respectively:<br />

J Clin Oncol 30:5s, 2012 (suppl; abstr LBA1)<br />

J Clin Oncol 30, 2012 (suppl; abstr e12000)<br />

Should you have any questions or comments about this<br />

publication, we encourage you to provide feedback by<br />

contacting us at abstracts@asco.org.<br />

Michael A. Carducci, MD<br />

Editor, 2012 ASCO <strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong>


JOURNAL <strong>of</strong> CLINICAL ONCOLOGY<br />

................................................................. Official Journal <strong>of</strong> the <strong>American</strong> <strong>Society</strong> <strong>of</strong> <strong>Clinical</strong> Oncology<br />

Journal <strong>of</strong> <strong>Clinical</strong> Oncology (ISSN 0732–183X) is published 36<br />

times a year, three times monthly, by the <strong>American</strong> <strong>Society</strong> <strong>of</strong><br />

<strong>Clinical</strong> Oncology, 2318 Mill Road, Suite 800, Alexandria, VA<br />

22314. Periodicals postage is paid at Alexandria, VA, and at<br />

additional mailing <strong>of</strong>fices.<br />

Postmaster<br />

Send all changes <strong>of</strong> address for Journal <strong>of</strong> <strong>Clinical</strong><br />

Oncology subscribers to:<br />

JCO Customer Service<br />

2318 Mill Road, Suite 800<br />

Alexandria, VA 22314<br />

Editorial Correspondence (manuscript-related inquiries):<br />

Stephen A. Cannistra, MD, Editor-in-Chief<br />

Journal <strong>of</strong> <strong>Clinical</strong> Oncology<br />

2318 Mill Road, Suite 800<br />

Alexandria, VA 22314<br />

Phone: 703-797-1900; Fax: 703-684-8720<br />

E-mail: jco@asco.org; Internet: www.jco.org<br />

<strong>American</strong> <strong>Society</strong> <strong>of</strong> <strong>Clinical</strong> Oncology (membership-related<br />

inquiries)<br />

ASCO Member Services<br />

2318 Mill Road, Suite 800<br />

Alexandria, VA 22314<br />

Phone: 703-299-0158; Toll-free: 888-282-2552<br />

Fax: 703-299-0255<br />

E-mail: membermail@asco.org; Internet: www.asco.org<br />

Hours: Monday-Friday, 8:30 a.m.-5:00 p.m. Eastern Time<br />

Customer Service, Subscriptions, and Changes <strong>of</strong> Address:<br />

JCO Customer Service<br />

2318 Mill Road, Suite 800<br />

Alexandria, VA 22314<br />

Phone: 703-519-1430; Toll-free: 888-273-3508; Fax: 703-518-8155<br />

E-mail: jcoservice@asco.org<br />

Internet orders/renewals: www.jco.org/subscriptions<br />

2012 SUBSCRIPTION RATES<br />

Individual Prices<br />

Domestic (US) Print � Online<br />

Individuals in training $289<br />

Individuals (1 year) $578<br />

International Print � Online<br />

Individuals in training $401<br />

Individuals (1 year) $802<br />

Institutional Prices<br />

Domestic (US) Print � Online Online Only<br />

Tier 1 $874 $749<br />

Tier 2 $1,019 $873<br />

Tier 3 $1,470 $1,261<br />

Tier 4 Call for quote Call for quote<br />

Institutional Prices<br />

International Print � Online Online Only<br />

Tier 1 $1,209 $749<br />

Tier 2 $1,351 $873<br />

Tier 3 $1,792 $1,261<br />

Tier 4 Call for quote Call for quote<br />

Orders and Payments<br />

P.O. Box 37211<br />

Baltimore, MD 21279-3211<br />

Important Tiers and Pricing Notes<br />

Additional rates along with tier descriptions are available online<br />

at www.jco.org/ratecard<br />

1. Prices are in effect from September 1, 2011, through August<br />

31, 2012. Prices are subject to change.<br />

2. Print-only subscriptions or additional print subscriptions are<br />

available for $730 in the US and $1,055 outside the US.<br />

3. Institutional online access, whether an online-only or bundled<br />

subscription, is for a single-site license, which allows an unlimited<br />

number <strong>of</strong> concurrent users from that site.<br />

4. For multisite licenses, please contact the appropriate agent<br />

for a quote.<br />

5. Subscribers outside the US, add $100 per print subscription<br />

for expedited delivery.<br />

6. Single-issue price: $40 US, $50 international.<br />

7. Prices quoted are in US dollars and payments must be made<br />

in US dollars.<br />

8. Except on Tier 5 orders, the publisher allows for a 5% discount<br />

to recognized subscription agents.<br />

Prices are subject to change without notice. Current prices are<br />

in effect for back volumes and back issues. Single issues, both<br />

current and back, exist in limited quantities and are <strong>of</strong>fered for sale<br />

subject to availability. Back issues sold in conjunction with a<br />

subscription are on a prorated basis.<br />

Advertising Sales<br />

The Walchli Tauber Group, Inc.<br />

225 Old Emmorton Road, Suite 201<br />

Bel Air, MD 21015<br />

Phone: 443-512-8899; Fax: 443-512-8909<br />

Internet: www.wt-group.com<br />

Business-to-Business Sales<br />

Rick Werdann<br />

Springer Healthcare, LLC<br />

233 Spring Street<br />

New York, NY 10013<br />

Phone: 212-460-1523; Mobile: 646-209-1840<br />

E-mail: rick.werdann@springer.com<br />

Internet: www.SpringerHealthcare.com<br />

LICENSES AND CONSORTIA<br />

USA, Canada, Europe, and India<br />

David Charles<br />

eLicensing<br />

92 Avenue du General de Gaulle<br />

78600 Maisons-Laffitte, France<br />

Phone/Fax: �33-1-39-12-29-29<br />

E-mail: dc.elicensing@orange.fr<br />

Japan<br />

USACO Corporation<br />

2-17-12 Higashi-Azuba Minato-ku<br />

Tokyo, Japan 106-0044<br />

Phone: �81-3-3505-3529; Fax: �81-3-3505-6284<br />

E-mail: import@usaco.co.jp; Internet: www.usaco.co.jp


JOURNAL <strong>of</strong> CLINICAL ONCOLOGY<br />

.................................................................... Official Journal <strong>of</strong> the <strong>American</strong> <strong>Society</strong> <strong>of</strong> <strong>Clinical</strong> Oncology<br />

China<br />

Charlesworth China<br />

Beijing Modern Palace Building, 12th Floor<br />

No. 20, Dongsanhuan Nanlu<br />

Chaoyang District<br />

Beijing 100022<br />

PR China<br />

Phone: �86-10-6779-1601; Fax: �86-10-6779-9806<br />

E-mail: sales@charlesworth.com.cn<br />

Internet: www.charlesworth.com.cn (in Mandarin)<br />

and www.charlesworth.com<br />

Indonesia, Malaysia, Philippines, Singapore, South Korea,<br />

Thailand, Taiwan, and Vietnam<br />

EBSCO EMpact<br />

5724 Highway 80 East<br />

Birmingham, AL 35242<br />

Phone: �1-205-980-6676<br />

Fax: �852-2575-8822<br />

E-mail: jmcdaniel@ebsco.com<br />

Central/South America, The Caribbean<br />

Accucoms (US), Inc.<br />

West Point Commons<br />

1816 West Point Pike, Suite 201<br />

Lansdale, PA 19446<br />

Phone: 215-395-5026<br />

Fax: 215-660-5042<br />

E-mail: anouk.snijders@accucoms.com<br />

Internet: www.accucoms.com<br />

Permissions Requests<br />

Licensing, Rights, and Permissions Division<br />

<strong>American</strong> <strong>Society</strong> <strong>of</strong> <strong>Clinical</strong> Oncology<br />

2318 Mill Road, Suite 800<br />

Alexandria, VA 22314<br />

Phone: 571-483-1722; Fax: 703-518-5094<br />

E-mail: permissions@asco.org<br />

Free Public Access<br />

Journal <strong>of</strong> <strong>Clinical</strong> Oncology (JCO) provides free online access<br />

to original research articles older than one year at<br />

www.jco.org. In addition, all ASCO Special Articles, Editorials,<br />

Comments and Controversies articles, the Art <strong>of</strong> Oncology<br />

series, and Correspondence articles are free immediately<br />

upon publication.<br />

Disclaimer<br />

The ideas and opinions expressed in JCO do not necessarily<br />

reflect those <strong>of</strong> the <strong>American</strong> <strong>Society</strong> <strong>of</strong> <strong>Clinical</strong> Oncology<br />

(ASCO). The mention <strong>of</strong> any product, service, or therapy in<br />

this publication or in any advertisement in this publication<br />

should not be construed as an endorsement <strong>of</strong> the products<br />

mentioned. It is the responsibility <strong>of</strong> the treating physician or<br />

other health care provider, relying on independent experience<br />

and knowledge <strong>of</strong> the patient, to determine drug dosages and<br />

the best treatment for the patient. Readers are advised to<br />

check the appropriate medical literature and the product<br />

information currently provided by the manufacturer <strong>of</strong> each<br />

drug to be administered to verify approved uses, the dosage,<br />

method, and duration <strong>of</strong> administration, or contraindications.<br />

Readers are also encouraged to contact the manufacturer<br />

with questions about the features or limitations <strong>of</strong> any<br />

products. ASCO assumes no responsibility for any injury or<br />

damage to persons or property arising out <strong>of</strong> or related to<br />

any use <strong>of</strong> the material contained in this publication or to any<br />

errors or omissions.<br />

Copyright<br />

Copyright © 2012 by <strong>American</strong> <strong>Society</strong> <strong>of</strong> <strong>Clinical</strong> Oncology<br />

unless otherwise indicated. All rights reserved. No part <strong>of</strong> this<br />

publication may be reproduced or transmitted in any form or<br />

by any means now or hereafter known, electronic or<br />

mechanical, including photocopy, recording, or any<br />

information storage and retrieval system, without permission<br />

in writing from the Publisher. Printed in the United States <strong>of</strong><br />

America.<br />

The appearance <strong>of</strong> the code at the bottom <strong>of</strong> the left column<br />

<strong>of</strong> the first page <strong>of</strong> an article in this journal indicates the<br />

copyright owner’s consent that copies <strong>of</strong> the article may be<br />

made for personal or internal use, or for the personal or internal<br />

use <strong>of</strong> specific clients, for those registered with the Copyright<br />

Clearance Center, Inc. (222 Rosewood Drive, Danvers, MA 01923;<br />

978-750-8400; www.copyright.com). This consent is given on the<br />

condition that the copier pay the stated per-copy fee for that<br />

article through the Copyright Clearance Center, Inc., for copying<br />

beyond that permitted by Sections 107 or 108 <strong>of</strong> the US<br />

Copyright Law. This consent does not extend to other kinds <strong>of</strong><br />

copying, such as copying for general distribution, for advertising<br />

or promotional purposes, for creating new collective works, or<br />

for resale. Absence <strong>of</strong> the code indicates that the material may<br />

not be processed through the Copyright Clearance Center, Inc.<br />

CPT © is a trademark <strong>of</strong> the <strong>American</strong> Medical Association.


ASCO Abstracts Policy<br />

Public Release <strong>of</strong> Abstracts<br />

ASCO has implemented changes to its abstract distribution to ensure simultaneous public release <strong>of</strong> important<br />

scientific information. Please note the new release schedule.<br />

The abstracts published in the 2012 ASCO <strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong> <strong>Part</strong> I, including those abstracts<br />

published but not presented at the <strong>Meeting</strong>, were publicly released by ASCO at 6:00 PM (EDT) on Wednesday,<br />

May 16, 2012. These abstracts are publicly available online at ASCO.org, the <strong>of</strong>ficial website <strong>of</strong> the <strong>Society</strong>.<br />

Plenary, Late-Breaking, and <strong>Clinical</strong> Review Abstracts (Newly Released Abstracts) will be publicly released<br />

according to the following schedule:<br />

● Newly Released Abstracts presented on Saturday, June 2, will be publicly released that day on ASCO.org at<br />

12:01 AM (EDT). These abstracts will also be available in the Saturday issue <strong>of</strong> ASCO Daily News in Section D.<br />

● Newly Released Abstracts presented on Sunday, June 3, will be publicly released that day on ASCO.org at<br />

12:01 AM (EDT). These abstracts will also be available in the Sunday issue <strong>of</strong> ASCO Daily News in Section D.<br />

● Newly Released Abstracts presented on Monday, June 4, and Tuesday, June 5, will be publicly released on<br />

Monday, June 4, on ASCO.org at 12:01 AM (EDT). These abstracts will also be available in the Monday issue <strong>of</strong><br />

ASCO Daily News in Section D.<br />

In the unlikely event that ASCO publicly releases an abstract in advance <strong>of</strong> the scheduled time, the release will be<br />

publicly announced on ASCO.org.<br />

Abstract Notice<br />

All abstracts presented at and published in conjunction with the <strong>Annual</strong> <strong>Meeting</strong> are included in online<br />

supplements to the Journal <strong>of</strong> <strong>Clinical</strong> Oncology. The abstracts released on May 16, 2012, are included in the May<br />

20 (Vol. 30, No. 15S) issue (2012 <strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong> <strong>Part</strong> I), and the Plenary, Late-Breaking, and <strong>Clinical</strong><br />

Review Abstracts, released on a daily basis during the <strong>Meeting</strong>, are included in the June 20 (Vol. 30, No. 18S)<br />

issue (2012 <strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong> <strong>Part</strong> II).<br />

Copyright 2012 <strong>American</strong> <strong>Society</strong> <strong>of</strong> <strong>Clinical</strong> Oncology


ASCO Conflict <strong>of</strong> Interest Policy and Exceptions<br />

In compliance with standards established by the ASCO Conflict <strong>of</strong> Interest Policy (J Clin Oncol. 2006;<br />

24[3]:519–521) and the Accreditation Council for Continuing Medical Information (ACCME), ASCO strives to<br />

promote balance, independence, objectivity, and scientific rigor through disclosure <strong>of</strong> financial and other interests,<br />

and identification and management <strong>of</strong> potential conflicts. According to the <strong>Society</strong>’s Conflict <strong>of</strong> Interest Policy, the<br />

following financial and other relationships must be disclosed: employment or leadership position, advisory role,<br />

stock ownership, honoraria, research funding, expert testimony, and other remuneration (J Clin Oncol.<br />

2006;24[3]:520).<br />

The ASCO Conflict <strong>of</strong> Interest Policy requirements apply to all abstract authors. Per the ASCO Conflict <strong>of</strong> Interest<br />

Policy Implementation Plan for CME Activities, all Oral Abstract Presenters will be subject to the same disclosure<br />

review and management strategies as faculty who participate in ASCO CME activities.<br />

For clinical trials that began accrual on or after April 29, 2004, the Conflict <strong>of</strong> Interest Policy places some<br />

restrictions on the financial relationships between a trial’s Principal Investigator (PI) and the trial’s company<br />

sponsor (J Clin Oncol. 2006;24[3]:521). If a PI holds any restricted relationships, his or her abstract may be<br />

ineligible for placement in the 2012 <strong>Annual</strong> <strong>Meeting</strong> unless the Ethics Committee grants an exception. Exceptions<br />

are generally not granted for PIs who have employment relationships with their trial’s company sponsor or stock<br />

in the company sponsor. Abstracts that receive exceptions will be subject to additional management strategies,<br />

including but not limited to additional peer review, advance slide review, and session audits. ASCO will collect<br />

information on accrual initiation date, financial relationships <strong>of</strong> the principal investigator, and National Institutes<br />

<strong>of</strong> Health (NIH) funding upon abstract submission. NIH-funded trials are exempt from the PI restrictions in the<br />

Conflict <strong>of</strong> Interest. Abstracts for which authors requested and have been granted an exception in accordance with<br />

ASCO’s Policy are designated with a caret symbol (^) in the <strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong>.<br />

For more information on the ASCO Conflict <strong>of</strong> Interest Policy, the Conflict <strong>of</strong> Interest Policy Implementation Plan<br />

for CME Activities, and the restrictions on PIs, please visit www.asco.org/rwi.


2012 ASCO <strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong> Special Awards<br />

1s<br />

ABSTRACTS<br />

The <strong>American</strong> <strong>Society</strong> <strong>of</strong> <strong>Clinical</strong> Oncology<br />

48th <strong>Annual</strong> <strong>Meeting</strong><br />

June 1–5, 2012<br />

McCormick Place<br />

Chicago, Illinois<br />

SPECIAL AWARD LECTURE ABSTRACTS<br />

David A. Karn<strong>of</strong>sky Memorial Award and Lecture<br />

Saturday, June 2, 9:30 AM<br />

Progress in chronic lymphocytic leukemia (CLL): A tribute to Dr. David A. Karn<strong>of</strong>sky.<br />

Kanti R. Rai, MB,BS; North Shore-Long Island Jewish Medical Center, New Hyde Park, NY<br />

My own journey in cancer research started in 1958 when I was a 26-year-old resident in pediatrics and was called<br />

upon one day to see and admit a 3-year-old named Laurie. Laurie, it turned out, had acute lymphoblastic<br />

leukemia (ALL).The attending hematologist helped me in making the diagnosis that night. I took care <strong>of</strong> Laurie<br />

not only that night but throughout her disease course. As, unfortunately, was the case with all children with acute<br />

leukemia in 1958, after whatever treatment we were able to give, Laurie died a few months later. The<br />

hematologist, Dr. Arthur Sawitsky, had watched how very involved I had become in Laurie’s care and how deeply<br />

her death affected me, and counseled me to pursue leukemia research and become a hematologist. After I<br />

completed my hematology fellowship, Dr. Sawitsky helped me join the research team <strong>of</strong> a world-class<br />

hematologist, Dr. Eugene P. Cronkite, at the Brookhaven National Laboratory. It was while working with Dr.<br />

Cronkite and seeing patients with all hematologic malignancies that I was intrigued by the wide spectrum <strong>of</strong> life<br />

expectancies <strong>of</strong> patients carrying the same diagnosis <strong>of</strong> chronic lymphocytic leukemia (CLL). David Karn<strong>of</strong>sky<br />

himself had already written about CLL in the 1950s. He, as well as several other hematologists <strong>of</strong> that era, had<br />

mentioned that CLL, in contrast with ALL, was not a very exciting disease, not much was happening with patients<br />

with CLL. Both Cronkite and Sawitsky encouraged me to study this disease. We then developed a relatively simple<br />

staging system, which was rapidly integrated in practice by clinicians, and it became possible to classify all CLL<br />

patients, on a prospective basis, into three distinct prognosis groups. It became possible to initiate therapeutic<br />

trials and ask research questions. Within two decades after clinical staging was developed, my colleague,<br />

Nicholas Chiorazzi, who is the leader <strong>of</strong> our CLL basic research team, demonstrated that one <strong>of</strong> the fundamental<br />

differences between the good prognosis and bad prognosis groups was that the former had somatic<br />

hypermutations in the IgVH genes while the latter did not. CLL has become a disease that, today, fascinates<br />

molecular biologists, immunologists, as well as clinicians. Promising new therapies targeting the B-cell receptor<br />

signaling pathways have become a reality. I feel certain that this progress, indeed, is in the tradition that Dr.<br />

Karn<strong>of</strong>sky started.<br />

Science <strong>of</strong> Oncology Award and Lecture<br />

Sunday, June 3, 1:00 PM<br />

Normalizing tumor microenvironment to treat cancer: Bench to bedside to biomarkers.<br />

Rakesh K. Jain, PhD; Harvard Medical School and Massachusetts General Hospital, Boston, MA<br />

For the past three decades our laboratory has focused on one challenge: How do we improve the delivery and<br />

efficacy <strong>of</strong> therapeutics in cancer patients? Working on the hypothesis that the abnormal tumor microenvironment<br />

fuels tumor progression and treatment resistance, we developed an array <strong>of</strong> sophisticated imaging<br />

technologies as well as mathematical and animal models to unravel the complex biology <strong>of</strong> the tumor<br />

microenvironment. Using these tools, we showed that the blood and lymphatic vessels as well as the extracellular<br />

matrix associated with tumors are abnormal. We further showed that these abnormalities generate a hostile tumor<br />

microenvironment (e.g., hypoxia, high interstitial fluid pressure). Our work also revealed how these abnormalities<br />

fuel malignant properties <strong>of</strong> a tumor as well as prevent treatments from reaching and attacking tumor cells. We<br />

next hypothesized that creative manipulation <strong>of</strong> the microenvironment should improve treatment outcomes.<br />

Toward this end, we discovered that “normalizing” the tumor vessels and matrix would allow cancer therapies to<br />

penetrate the mass and to function more effectively. Originally designed to destroy tumor vessels, we showed,<br />

first in mice and then in rectal and glioblastoma cancer patients, that antiangiogenic drugs could, paradoxically,


2s Special Awards<br />

also “normalize” them, creating a window <strong>of</strong> opportunity to attack the cancer more effectively. Moreover, tumor<br />

blood vessels’ normalization and improvement in tumor perfusion was associated with longer patient survival.<br />

These observations led to a number <strong>of</strong> clinical trials aimed at identifying biomarkers <strong>of</strong> response and resistance to<br />

antiangiogenic drugs. Our findings also opened doors to treating other vascular diseases, such as age-related wet<br />

macular degeneration, a leading cause <strong>of</strong> blindness, and neur<strong>of</strong>ibromatosis-2, which can lead to deafness. More<br />

recently, we discovered that drugs capable <strong>of</strong> normalizing the extracellular matrix improve the delivery and<br />

efficacy <strong>of</strong> molecular and nanomedicine. We are currently exploring this as a potential means <strong>of</strong> enhancing the<br />

delivery <strong>of</strong> chemotherapeutics to desmoplastic tumors (e.g., pancreatic ductal adenocarcinomas).<br />

ASCO–<strong>American</strong> Cancer <strong>Society</strong> Award and Lecture<br />

Saturday, June 2, 3:00 PM<br />

Reducing breast cancer risk: Progress toward resolution.<br />

Rowan T. Chlebowski, MD, PhD; Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center,<br />

Torrance, CA<br />

Randomized, full-scale clinical trials have 1) addressed lifestyle change in adjuvant breast cancer; 2) changed<br />

concepts regarding menopausal hormone therapy and breast cancer; and 3) identified new approaches to breast<br />

cancer risk reduction. The Women Intervention Nutrition Study (WINS) randomly assigned 2,437 women to<br />

standard therapy � a lifestyle intervention targeting fat intake. Intervention participants reduced fat intake, lost<br />

weight, and experienced a statistically significant increase in disease-free survival (HR 0.76, 95% CI<br />

0.60–0.98, p � 0.03). Long-term follow-up is underway and the European SUCCESS-C study will evaluate the<br />

emerging hypothesis that weight loss and increased physical activity will improve breast cancer outcome. The<br />

Women’s Health Initiative (WHI) placebo-controlled trials randomly assigned 16,000 women with no prior<br />

hysterectomy to estrogen plus progestin and 10,739 postmenopausal with prior hysterectomy to estrogen alone,<br />

and opposite breast cancer findings were seen. Estrogen plus progestin significantly increased breast cancer<br />

incidence, delayed diagnosis, and increased breast cancer mortality (mortality HR 1.96, 95% CI 1.00–4.05,<br />

p � .048). Estrogen alone significantly reduced breast cancer incidence and breast cancer mortality (mortality<br />

HR 0.37, 95% CI 0.13–0.91). The findings led to a worldwide reduction in hormone therapy use with substantial<br />

reduction in breast cancer incidence seen in many countries. In the MAP.3 randomized primary prevention trial,<br />

breast cancer incidence was significantly lower in the exemestane versus placebo group (HR 0.35, 95% CI<br />

0.18–0.70, p �0.002). Emerging studies evaluated additional agents for chemoprevention. In cohort analyses<br />

in 154,768 WHI study participants breast cancer incidence was lower in bisphosphonate users (HR 0.68, 95%<br />

CI 0.52–0.88, p � 0.01). In cohort analyses in 68,019 WHI clinical trial participants, women with diabetes<br />

using metformin had lower breast cancer incidence (HR 0.75, 95% CI 0.57–0.99, p � 0.04). These findings<br />

support consideration <strong>of</strong> combination regimens for breast cancer risk reduction with toxicity pr<strong>of</strong>iles favorable for<br />

board implementation.<br />

B. J. Kennedy Award and Lecture for Scientific Excellence in Geriatric Oncology<br />

Sunday, June 3, 9:45 AM<br />

Geriatric oncology: Realities and hopes around a patient’s history.<br />

Matti S. Aapro, MD; Clinique de Genolier, Genolier, Switzerland<br />

One <strong>of</strong> the significant milestones in the development <strong>of</strong> geriatric oncology was in 1988 when B. J. Kennedy<br />

commented in his Presidential Address at the ASCO <strong>Annual</strong> <strong>Meeting</strong> about the gaps in our knowledge in this<br />

important field. Recognition <strong>of</strong> the situation by a major society like ASCO boosted the efforts <strong>of</strong> the research<br />

community. The French GELA group demonstrated the feasibility <strong>of</strong> standard high-grade lymphoma chemotherapy<br />

in elderly patients, and CALGB showed its importance in adjuvant treatment <strong>of</strong> breast cancer (Aapro M.<br />

Launching the Journal <strong>of</strong> Geriatric Oncology: A historical milestone. J Geriatric Oncol. 2010;1:2–3). This<br />

presentation will address the many prejudices and sad realities <strong>of</strong> geriatric cancer patients, by following a slightly<br />

modified true patient history. We should recognize that there is a lot to do to overcome the obstacles that continue<br />

to mean that older individuals do not receive the best possible care. But evidence for treatments tailored to the<br />

real status <strong>of</strong> the patient and the true nature <strong>of</strong> the cancer is accumulating. The International <strong>Society</strong> for Geriatric<br />

Oncology has identified three key areas <strong>of</strong> priority in this field: education, clinical practice, and research<br />

(Extermann M, Aapro M, Audisio R, et al. Main priorities for the development <strong>of</strong> geriatric oncology: A worldwide<br />

expert perspective. J Geriatric Oncol. 2011;1:270–273). Education should be targeted at both the pr<strong>of</strong>essional<br />

and the population levels. In clinical practice, pilot models <strong>of</strong> multidisciplinary collaboration should be expanded<br />

first to key reference centers, and a two-step approach to screening and evaluation should be used to optimize<br />

resource use. In research, several strategies can render trials more relevant for older patients. These priorities are<br />

fully detailed in a monograph that can be viewed online at www.siog.org or ordered from siog@genolier.net.


Special Awards<br />

Pediatric Oncology Award and Lecture<br />

Friday, June 1, 2:45 PM<br />

Toward the total cure <strong>of</strong> childhood acute lymphoblastic leukemia with personalized therapy.<br />

Ching-Hon Pui, MD; St. Jude Children’s Research Hospital and the University <strong>of</strong> Tennessee Health Science<br />

Center, Memphis, TN<br />

The cure rate <strong>of</strong> approximately 90% achieved in childhood acute lymphoblastic leukemia (ALL) attests to the<br />

effectiveness <strong>of</strong> risk-directed therapy developed through well-designed clinical trials and the successful<br />

translation <strong>of</strong> laboratory discoveries. With optimal systemic and intrathecal therapy, central nervous system<br />

relapse can now be totally prevented without the need <strong>of</strong> prophylactic cranial irradiation. The results <strong>of</strong> allogeneic<br />

transplantation have also been improved by more precise HLA matching, killer immunoglobulin-like receptor<br />

genotyping, and better supportive care. Further improvements in treatment outcome and quality <strong>of</strong> life for these<br />

patients will almost certainly require continued dissection <strong>of</strong> ALL pathophysiology and the mechanisms <strong>of</strong> drug<br />

resistance as they relate to the genetic and epigenetic changes for individual patients. The high-risk leukemic cell<br />

genetic abnormalities alone can no longer be considered justification for stem cell transplantation, given the<br />

improved chemotherapy and the substantial heterogeneity among these patients in cooperating mutations,<br />

development stages <strong>of</strong> the transformed cells, and host pharmacogenetics. Recent advances in global genomic<br />

analyses have identified new recurrent somatic lesions in the leukemic cells that <strong>of</strong>fer not only additional<br />

prognostic information but also the opportunity to apply novel targeted therapy. Notably, the recently identified<br />

mutational spectrum in early T-cell precursor ALL—a subset <strong>of</strong> ALL with an extremely dire prognosis—<br />

recapitulates that <strong>of</strong> acute myeloid leukemia, while its global transcriptional pr<strong>of</strong>ile is similar to that <strong>of</strong> normal<br />

and myeloid leukemia hematopoietic stem cells, suggesting that additional myeloid-directed therapy might<br />

benefit patients with this leukemia variant. Finally, it may be possible to improve the outcome <strong>of</strong> “BCR-ABL1like”<br />

ALL, a new subtype <strong>of</strong> high-risk B-cell precursor ALL with an IZKF1 alteration, by targeting recently<br />

identified activation mutations with tyrosine kinase inhibitors. As the repertoire <strong>of</strong> targeted therapeutics<br />

including cell- and antibody-based treatment increases, we can look forward to a total cure <strong>of</strong> ALL with<br />

acceptable toxicity.<br />

3s


2012 ASCO <strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong> Plenary Session<br />

5s<br />

LBA1 Plenary Session, Sun, 1:00 PM-4:00 PM<br />

Primary results from EMILIA, a phase III study <strong>of</strong> trastuzumab emtansine<br />

(T-DM1) versus capecitabine (X) and lapatinib (L) in HER2-positive locally<br />

advanced or metastatic breast cancer (MBC) previously treated with<br />

trastuzumab (T) and a taxane. Presenting Author: Kimberly L. Blackwell,<br />

Duke University Medical Center, Durham, NC<br />

The full, final text <strong>of</strong> this abstract will be available at<br />

abstract.asco.org at 12:01 AM (EDT) on Sunday, June 3,<br />

2012, and in the <strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong> online<br />

supplement to the June 20, 2012, issue <strong>of</strong> Journal <strong>of</strong><br />

<strong>Clinical</strong> Oncology. Onsite at the <strong>Meeting</strong>, this abstract will<br />

be printed in the Sunday edition <strong>of</strong> ASCO Daily News.<br />

ABSTRACTS<br />

The <strong>American</strong> <strong>Society</strong> <strong>of</strong> <strong>Clinical</strong> Oncology<br />

48th <strong>Annual</strong> <strong>Meeting</strong><br />

June 1-5, 2012<br />

McCormick Place<br />

Chicago, Illinois<br />

2 Plenary Session, Sun, 1:00 PM-4:00 PM<br />

Long-term follow-up results <strong>of</strong> EORTC 26951: A randomized phase III<br />

study on adjuvant PCV chemotherapy in anaplastic oligodendroglial tumors<br />

(AOD). Presenting Author: Martin J. Van Den Bent, Erasmus University<br />

Medical Center, Daniel den Hoed Cancer Center, Rotterdam, Netherlands<br />

The full, final text <strong>of</strong> this abstract will be available at<br />

abstract.asco.org at 12:01 AM (EDT) on Sunday, June 3,<br />

2012, and in the <strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong> online<br />

supplement to the June 20, 2012, issue <strong>of</strong> Journal <strong>of</strong><br />

<strong>Clinical</strong> Oncology. Onsite at the <strong>Meeting</strong>, this abstract will<br />

be printed in the Sunday edition <strong>of</strong> ASCO Daily News.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


6s Plenary Session<br />

3 Oral Abstract Session, Sat, 3:00 PM-6:00 PM<br />

Bendamustine plus rituximab (B-R) versus CHOP plus rituximab (CHOP-R)<br />

as first-line treatment in patients with indolent and mantle cell lymphomas<br />

(MCL): Updated results from the StiL NHL1 study. Presenting Author:<br />

Mathias J. Rummel, Universitaetsklinik, Giessen, Germany<br />

The full, final text <strong>of</strong> this abstract will be available at<br />

abstract.asco.org at 12:01 AM (EDT) on Sunday, June 3,<br />

2012, and in the <strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong> online<br />

supplement to the June 20, 2012, issue <strong>of</strong> Journal <strong>of</strong><br />

<strong>Clinical</strong> Oncology. Onsite at the <strong>Meeting</strong>, this abstract will<br />

be printed in the Sunday edition <strong>of</strong> ASCO Daily News.<br />

4 Plenary Session, Sun, 1:00 PM-4:00 PM<br />

Intermittent (IAD) versus continuous androgen deprivation (CAD) in hormone<br />

sensitive metastatic prostate cancer (HSM1PC) patients (pts):<br />

Results <strong>of</strong> S9346 (INT-0162), an international phase III trial. Presenting<br />

Author: Maha Hussain, University <strong>of</strong> Michigan Comprehensive Cancer<br />

Center, Ann Arbor, MI<br />

The full, final text <strong>of</strong> this abstract will be available at<br />

abstract.asco.org at 12:01 AM (EDT) on Sunday, June 3,<br />

2012, and in the <strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong> online<br />

supplement to the June 20, 2012, issue <strong>of</strong> Journal <strong>of</strong><br />

<strong>Clinical</strong> Oncology. Onsite at the <strong>Meeting</strong>, this abstract will<br />

be printed in the Sunday edition <strong>of</strong> ASCO Daily News.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


500 Oral Abstract Session, Sun, 8:00 AM-11:00 AM<br />

Impact <strong>of</strong> concurrent (CON) and sequential (SEQ) radiotherapy (RT) with<br />

adjuvant aromatase inhibitors (AI) in early-stage breast cancer (EBC): NCIC<br />

CTG MA.27. Presenting Author: Abdullah Khalaf Altwairgi, NCIC <strong>Clinical</strong><br />

Trials Group, Queen’s University, Kingston, ON, Canada<br />

Background: Optimal timing <strong>of</strong> administration <strong>of</strong> adjuvant (adj) RT and AI in<br />

EBC is unknown. Methods: MA.27 was a phase III RCT <strong>of</strong> exemestane to<br />

anastrozole in postmenopausal women with hormone receptor positive EBC<br />

(Goss et al. Cancer Res. 70(24, Suppl):75s, 2010). The final trial database<br />

was used for this retrospective analysis. Median follow-up was 4.1 years.<br />

MA.27 patients received CON-AI [any overlap with AI; 4233 (57%)<br />

patients], SEQ-AI [RT preceded AI, no overlap with AI; 1010 (14%)<br />

patients] and No RT [AI only; 2128 (29%) patients]. Outcome measures for<br />

this analysis were: event free survival (EFS; time to locoregional or distant<br />

disease recurrence, new primary BC, or death from any cause), locoregional<br />

recurrence (LRFS), distant recurrence (DDFS) and overall survival (OS). RT<br />

groups were compared univariately (uni) with stratified log-rank tests, and<br />

multivariately (multi) with step-wise stratified Cox regression adjusted by<br />

stratification factors: nodal status, adj chemotherapy (chemo), celecoxib,<br />

aspirin, and trastuzumab. Results: 7371 eligible women received AI; were<br />

included in the analysis; and 71% (5243) received adj RT. CON-AI and<br />

SEQ-AI groups were comparable by median age (63 v 63), proportion T1<br />

tumours (75% v 75 %), and mastectomy rate (10% v 11%). The frequency<br />

<strong>of</strong> axillary dissection for CON-AI and SEQ-AI was 48% v 44%, proportion<br />

N0 was 73% v 69%, and proportion receiving adj chemo 29% v 41%.<br />

CON-AI had similar uni results to SEQ-AI: EFS, HR�0.86, p�0.20; LRFS,<br />

HR�0.82, p�0.51; DDFS, HR�0.92, p�0.59; and OS, HR�1.04,<br />

p�0.80. In multi analyses, CON-AI had better EFS than SEQ-AI patients<br />

[stratified HR <strong>of</strong> CON-AI to SEQ-AI 0.78 (0.66 – 0.91), p�0.001]; as well,<br />

age�70 (p�0.0001), ECOG PS�1 (p�0.0001), L-sided tumours<br />

(p�0.02), T2-T4 (p�0.0001), N2/N3 (p�0.0001), and no adj chemo<br />

(p�0.01) had significantly shorter EFS. There was no multi difference<br />

between CON-AI and SEQ-AI for LRFS (p�0.50), DDFS (p�0.72), or OS<br />

(p�0.85). Conclusions: Patients receiving CON-AI had significantly better<br />

EFS than SEQ-AI suggesting timing <strong>of</strong> administration <strong>of</strong> AI and RT may<br />

affect patient outcomes. Further research is necessary to confirm these<br />

findings.<br />

502 Oral Abstract Session, Sun, 8:00 AM-11:00 AM<br />

Adjuvant therapy with zoledronic acid (AZURE-BIG 01/04): The influence<br />

<strong>of</strong> menopausal status and age on treatment effects. Presenting Author:<br />

Helen Marshall, CTRU, Leeds, United Kingdom<br />

Background: The AZURE trial evaluated the use <strong>of</strong> zoledronic acid (ZOL) in<br />

addition to standard adjuvant therapy in patients with stage II/III breast<br />

cancer. Although analysis <strong>of</strong> the primary endpoint in the total study<br />

population showed no significant effects on disease outcomes, prespecified<br />

subgroup analyses identified significant benefit in both invasive<br />

disease-free survival (IDFS) and overall survival (OS) in women �5years<br />

postmenopause (Coleman et al. NESM. 2011;365:1396–1405). Here we<br />

evaluate the treatment modifying effects <strong>of</strong> both menopausal status and<br />

age. Methods: 3360 patients were randomized to receive standard (neo)<br />

adjuvant systemic therapy �/- ZOL 4mg iv every 3-4 weeks for 6 doses,<br />

then at reduced frequency to complete 5 years treatment. Results: ZOL had<br />

no overall effect on IDFS or OS at a median follow-up <strong>of</strong> 59 months.<br />

Pre-specified subgroup analyses showed significant heterogeneity <strong>of</strong> treatment<br />

effect (chi2 1 �7.91; p�.0049) on IDFS between those who were �5<br />

years post-menopause (n�1041; adjusted HR� 0.75; 95% CI 0.59-0.96,<br />

p�0.02) and all other (pre, peri and unknown status) menopausal groups<br />

(n�2318; adjusted HR� 1.15; 95% CI 0.97-1.36, p�0.11). The treatment<br />

interaction between the two menopausal groups was related to<br />

differences in first extra-skeletal IDFS events (chi2 1 � 14.00, p�0.0002),<br />

rather than first recurrence in bone (chi2 1 � 0.14, p�0.70). In women<br />

aged �40, a significant worsening in extra-skeletal IDFS events was seen<br />

(HR � 1.68; 95% CI � 1.14-2.47, p�.008). Although the ZOL effect on<br />

IDFS improved by age from a negative effect in younger patients (�40) to a<br />

beneficial effect for those aged 54 and over (chi2 1(trend) � 5.96,<br />

p�0.015, using Cox model analysis to test for an interaction effect<br />

between age, menopausal status and treatment for IDFS (using age 55 as a<br />

cut-<strong>of</strong>f) showed that the age effect is reversed by menopausal status, with<br />

this model providing a significantly better fit (chi2 2 � 22.8, p�.00001)<br />

than the aforementioned linear model. Conclusions: These results suggest<br />

menopausal status is more informative than age in the selection <strong>of</strong> patients<br />

for treatment with ZOL. In women aged �40, ZOL may increase extraskeletal<br />

recurrence.<br />

Breast Cancer—HER2/ER<br />

501 Oral Abstract Session, Sun, 8:00 AM-11:00 AM<br />

Effect <strong>of</strong> osteoporosis in postmenopausal breast cancer patients randomized<br />

to adjuvant exemestane or anastrozole: NCIC CTG MA.27. Presenting<br />

Author: Lois E. Shepherd, NCIC <strong>Clinical</strong> Trials Group, Queen’s University,<br />

Kingston, ON, Canada<br />

Background: NCIC CTG MA.27 compared adjuvant steroidal [exemestane<br />

(E)] and non-steroidal [anastrozole (A)] aromatase inhibitors (AIs) and<br />

showed neither to be superior in breast cancer outcomes. AIs are known to<br />

increase the risk <strong>of</strong> osteoporosis. We examined the effects <strong>of</strong> baseline or<br />

subsequent self reported osteoporosis on disease outcomes. Methods:<br />

MA.27 enrolled 7,576 women. Event free survival (EFS) was the primary<br />

endpoint. Distant disease-free-survival (DDFS) was a secondary outcome.<br />

MA.27 permitted bisphosphonates to prevent or treat osteopenia or<br />

osteoporosis unless prohibited by enrollment to one <strong>of</strong> two cohorts in a bone<br />

substudy. Osteoporosis was considered present for this analysis if reported<br />

at baseline or prior to relapse or breast cancer death. Multivariate stratified<br />

Cox regression was used to examine the effects <strong>of</strong> trial therapy, osteoporosis,<br />

baseline patient and tumour characteristics on EFS; DDFS; bone only<br />

relapse; bone concurrent with other relapse; and non-bone recurrence.<br />

Bone marrow recurrence (N�8) was excluded. Results: Osteoporosis was<br />

reported at baseline by 654 <strong>of</strong> 7576 (8.6%) women, and prior to relapse by<br />

an additional 661 women. EFS events occurred in 693/7576 (9.15%).<br />

Osteoporosis was significantly associated with better EFS [HR 0.81 (95%<br />

CI 0.66-0.99), p�0.04] with no difference in multivariate HR <strong>of</strong> E vs A:<br />

1.02, 95% CI 0.88-1.19, p�0.77. Women experienced 313 (4.1%) DDFS<br />

events. Osteoporosis was associated with better DDFS [HR 0.71 (95% CI<br />

0.51-0.98), p�0.04], adjusted HR <strong>of</strong> E to A: 0.94 (95% CI <strong>of</strong> 0.75-1.17),<br />

p�0.56. Both EFS and DDFS interactions <strong>of</strong> osteoporosis with trial therapy<br />

were not significant (p�0.05). Osteoporosis was not significantly associated<br />

with the site <strong>of</strong> relapse. Conclusions: Osteoporosis had a significant<br />

prognostic association with improved EFS and DDFS in women treated with<br />

AIs. We plan to investigate the use <strong>of</strong> bisphosphonates, raloxifene treatment<br />

prior to randomization, and markers <strong>of</strong> bone resorption with outcome.<br />

503 Oral Abstract Session, Sun, 8:00 AM-11:00 AM<br />

Whole genome sequencing to characterize luminal-type breast cancer.<br />

Presenting Author: Matthew James Ellis, Department <strong>of</strong> Internal Medicine,<br />

Division <strong>of</strong> Oncology, and Siteman Cancer Center, Washington University<br />

Medical Center, St. Louis, MO<br />

Background: To correlate clinical features <strong>of</strong> estrogen receptor positive<br />

breast cancer with somatic mutations, massively parallel sequencing<br />

(MPS) was applied to tumor and normal DNAs accrued from patients<br />

treated with neoadjuvant aromatase inhibitors (AI). Methods: MPS was<br />

applied to 77 baseline tumor biopsy samples from the preoperative<br />

letrozole trial (JACS 2009: 208, 906) and the Z1031 trial (JCO 2011: 29,<br />

2342) followed by targeted sequencing in another 240 trial samples.<br />

Standard statistical approaches were used to compare mutation status and<br />

clinical parameters and pathway-based correlation was used to assess<br />

interactions between signaling perturbations induced by gene mutations<br />

and response to neoadjuvant AI. Results: Eighteen genes were significantly<br />

mutated above background. Aside from PIK3CA mutations, the list is<br />

dominated by loss-<strong>of</strong>-function mutations in tumor suppressor genes. Five<br />

(RUNX1, CBFP, MYH9, MLL3 and SF3B1) have been previously linked to<br />

benign and malignant hematopoietic disorders. <strong>Clinical</strong> correlation revealed<br />

that TP53 mutation was associated with PAM50 LumB status,<br />

high-grade histology and high proliferation rates whereas loss-<strong>of</strong>-function<br />

mutations in MAP3K1 associate with PAM50 lumA status, low proliferation<br />

rates, and low grade histology. Mutations in GATA3 were associated with<br />

greater suppression <strong>of</strong> proliferation upon AI treatment suggesting mut-<br />

GATA3 may predict endocrine response. Notably, mutations in MAP3K1<br />

were more common in PIK3CA mutant cases, suggesting cooperation.<br />

Pathway analysis demonstrated that rare MAP2K4 mutations produce<br />

similar pathway perturbations as MAP3K1 mutation, a logical finding since<br />

MAP2K4 is a substrate for MAP3K1. Signaling network patterns driven by<br />

lncRNA MALAT1 mutations were associated with multiple poor clinical<br />

outcome features. Rare mutations in druggable kinases included two in the<br />

kinase domain <strong>of</strong> HER2. Conclusions: Tumor heterogeneity in luminal-type<br />

breast cancer is driven by specific patterns <strong>of</strong> somatic mutations, however<br />

most druggable or potentially prognostic mutations are infrequent. Prospective<br />

clinical trials based on these findings will require comprehensive<br />

genome sequencing approaches and large scale investigations.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.<br />

7s


8s Breast Cancer—HER2/ER<br />

504 Oral Abstract Session, Sun, 8:00 AM-11:00 AM<br />

Intrinsic subtype and its clinical significance in early node-negative breast<br />

cancer: Results from two randomized trials <strong>of</strong> adjuvant chemoendocrine<br />

therapy. Presenting Author: Ewan K. A. Millar, The Kinghorn Cancer<br />

Centre, Sydney, Australia<br />

Background: Gene expression pr<strong>of</strong>iling has identified four main intrinsic<br />

subtypes <strong>of</strong> breast cancer which have distinct clinical behaviours and<br />

responses to therapy. A simplified immunohistochemical (IHC) panel can<br />

provide meaningful discrimination and help guide patient management.<br />

Methods: Using tissue microarrays (TMA) created from two randomized<br />

trials <strong>of</strong> adjuvant chemoendocrine therapy in node negative early breast<br />

cancer (IBCSG VIII and IX,n� 1220) we assessed intrinsic subtype as<br />

follows: “luminal A” (LA): ER and/or PR present, Ki-67 low (�median:<br />

19%), HER2-; “Luminal B” (LB): ER and/or PR present, Ki-67<br />

high(��19% median) and/or HER2� (IHC 3� or FISH amplified); HER2<br />

enriched: ER and PR negative, HER2 positive (IHC 3� or FISH amplified),<br />

Triple Negative (TNP): ER, PR and HER2 negative. p53� and high cyclin<br />

D1 expression were then assessed to determine if they provided any further<br />

additive discriminatory value. Results: The standard definitions <strong>of</strong> intrinsic<br />

subtype demonstrated a significant difference in disease-free survival<br />

between the subtypes in both trials VIII and IX (p� 0.02), but this was not<br />

significant in multivariate analysis. The discrimination between luminal A<br />

and B tumors could not be improved upon by the addition <strong>of</strong> p53 or cyclin<br />

D1 expression. The standard definitions demonstrated that TNP and<br />

HER2-enriched tumors benefited from the addition <strong>of</strong> chemotherapy to<br />

endocrine therapy, but LA and LB did not. p53� was associated with a<br />

poorer prognosis in ER� luminal tumors but improved prognosis in ERnon-luminal<br />

tumors (Interaction p � 0.002). Conclusions: Intrinsic subtyping<br />

using a panel <strong>of</strong> ER, PR, HER2 and Ki67 provides meaningful<br />

prognostic information to help guide patient management in early breast<br />

cancer. Whilst it predicts benefit from the addition <strong>of</strong> chemotherapy in TNP<br />

and HER2-enriched tumors it did not in this series do so for LB. Although<br />

p53 status did not improve the definition <strong>of</strong> luminal tumors, p53�<br />

appeared to have a divergent effect on outcome dependent on ER status.<br />

LBA506 Oral Abstract Session, Sun, 8:00 AM-11:00 AM<br />

Evaluation <strong>of</strong> lapatinib as a component <strong>of</strong> neoadjuvant therapy for HER2�<br />

operable breast cancer: NSABP protocol B-41. Presenting Author: Andre<br />

Robidoux, National Surgical Adjuvant Breast and Bowel Project and Centre<br />

Hospitalier de l’Universite de Montreal, Montreal, QC, Canada<br />

The full, final text <strong>of</strong> this abstract will be available at<br />

abstract.asco.org at 12:01 AM (EDT) on Sunday, June 3,<br />

2012, and in the <strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong> online<br />

supplement to the June 20, 2012, issue <strong>of</strong> Journal <strong>of</strong><br />

<strong>Clinical</strong> Oncology. Onsite at the <strong>Meeting</strong>, this abstract will<br />

be printed in the Sunday edition <strong>of</strong> ASCO Daily News.<br />

505 Oral Abstract Session, Sun, 8:00 AM-11:00 AM<br />

13-gene signature to predict rapid development <strong>of</strong> brain metastases in<br />

patients with HER2-positive advanced breast cancer. Presenting Author:<br />

Renata Duchnowska, Military Institute <strong>of</strong> Medicine, Warsaw, Poland<br />

Background: Brain metastases (BM) <strong>of</strong> breast cancer constitute an important<br />

part <strong>of</strong> therapeutic failures and are associated with severe morbidity<br />

and mortality. The risk <strong>of</strong> BM is particularly high in HER2� advanced<br />

breast cancer pts. We earlier developed in this group a 13-gene signature<br />

strongly predicting for rapid development <strong>of</strong> BM (J Clin Oncol 2008; 26:<br />

45s). Now, we validated these results in an independent group <strong>of</strong> pts and<br />

on culture model system. Methods: Discovery group included 87 samples<br />

analyzed using cDNA synthesis, annealing, selection, extension, ligation<br />

and array hybridization (DASL). Independent validation group included 75<br />

samples analyzed using quantitative reverse-transcriptase PCR. 3D culture<br />

validation model system used immortal, non-tumorigenic human MCF10A<br />

breast epithelial cells with and without ectopic expression <strong>of</strong> HER-2 and<br />

RAD51, a DNA double strand break repair gene (one <strong>of</strong> the three genes <strong>of</strong><br />

this group overexpressed in 13-gene signature). The number and morphology<br />

<strong>of</strong> breast acini were scored using indirect immun<strong>of</strong>lourescence and<br />

confocal microscopy. Results: Median brain metastasis-free survival (BMFS)<br />

in the discovery group for ‘high’ vs. ‘low’ expression signature tumors was<br />

36 months and 66 months, respectively (P�0.0068), and in the validation<br />

group 54 and 86 months, respectively (P�0.032). Short BMFS was also<br />

associated with ER-negativity; BMFS in the cohort <strong>of</strong> ‘high’ 13-gene<br />

signature and ER- tumors vs. other 3 groups was 31 months and 66 months<br />

in discovery group, and 41 and 77 months in validation group (P�0.0001<br />

and P�0.02, respectively). Overexpression <strong>of</strong> RAD51 in MCF-10A breast<br />

cells altered their three-dimensional acinar morphology and increased the<br />

percentage <strong>of</strong> invasive structures by 6.5 fold, both in the presence and<br />

absence <strong>of</strong> HER2 overexpression. Conclusions: 13-gene signature and<br />

ER-negativity predict rapid development <strong>of</strong> BM in HER� advanced breast<br />

cancer pts. RAD51 may promote aggressiveness in breast epithelial cells.<br />

These data may be useful in the design <strong>of</strong> BM preventive trials and may<br />

prompt new treatment strategies.<br />

507 <strong>Clinical</strong> Science Symposium, Sat, 1:15 PM-2:45 PM<br />

Tumor PIK3CA mutations, lymphocyte infiltration, and recurrence-free<br />

survival (RFS) in early breast cancer (BC): Results from the FinHER trial.<br />

Presenting Author: Sherene Loi, Jules Bordet Institute, Brussels, Belgium<br />

Background: Tumor PIK3CA mutations (mts) and lymphocyte infiltration<br />

(LI) are prognostic in BC, but their importance is unknown in HER2positive<br />

(HER2�) BC treated with adjuvant trastuzumab (T). Methods: The<br />

FinHER trial randomized 1010 patients (pts) with pN� or high-risk pN- BC<br />

to 3 cycles <strong>of</strong> docetaxel (D) or vinorelbine, followed by 3 cycles <strong>of</strong> FEC. Pts<br />

with HER2� BC were further randomized to 9 weeks <strong>of</strong> T or no T (n�232).<br />

Pts treated with D and T had superior outcome in prior analyses. BC<br />

samples were subjected to somatic hotspot mt pr<strong>of</strong>iling (Sequenom) and<br />

quantification <strong>of</strong> percentage tumor LI using full-face H&E sections. RFS<br />

and interactions with T were explored with Kaplan-Meier and Cox regression<br />

analyses. Results: The median FU time was 62 months. 935 (92.6%) and<br />

687 (68.1%) tumors underwent LI and mt analysis, respectively. Correlation<br />

<strong>of</strong> LI assessment between 2 pathologists was 0.78 (p�0.001). 54 mts<br />

detected in 13 genes were evaluated. PIK3CA mts (exons 1,2,9,13,18,20)<br />

was present in 25.3% (n�174) and ERBB2 mt in 4.5% (n�31) BCs. Only<br />

1 AKT mt was found and none in KRAS, BRAF, NRAS or PTEN. In<br />

ER�/HER2-, HER2� and ER-/HER2- subtypes the prevalence <strong>of</strong> PIK3CA<br />

mts was 30.2%, 19.5% and 9.2%; ERBB2 mt 6.3%, 1.9% and 2.6%,<br />

respectively (both p�0.05). Neither PIK3CA nor ERBB2 mts were associated<br />

with RFS overall or within BC subgroups. No interaction was found for<br />

the benefit with T and presence <strong>of</strong> PIK3CA mt in HER2� BC (interaction<br />

p�0.17 T vs no T). Increasing LI was associated with favorable RFS in<br />

ER-/HER2- BC (continuous score adjusted p�0.032); 3-y RFS was 90%<br />

with extensive LI (�30% infiltrated) vs 66% with non-extensive LI<br />

(p�0.007). In HER2� BC, whilst there was no association with prognosis<br />

overall, there was a significant interaction with the benefit from T vs. no T<br />

and increasing LI (continuous score interaction p�0.042) with 3-yr RFS<br />

96% vs 78% for extensive and non-extensive LI treated with T respectively<br />

(p�0.014). Conclusions: We report 3 clinically relevant findings from this<br />

large clinical trial dataset: 1) PIK3CA mts were not prognostic; 2) LI is a<br />

strong prognostic factor in ER-/HER2- BC and 3) for the first time that<br />

extensive LI predicts benefit from adjuvant T.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


508 <strong>Clinical</strong> Science Symposium, Sat, 1:15 PM-2:45 PM<br />

A phase I/IB dose-escalation study <strong>of</strong> BEZ235 in combination with<br />

trastuzumab in patients with PI3-kinase or PTEN altered HER2� metastatic<br />

breast cancer. Presenting Author: Ian E. Krop, Dana-Farber Cancer<br />

Institute, Boston, MA<br />

Background: Alterations in the PI3K/AKT/mTOR pathway have been implicated<br />

in resistance to trastuzumab (T) in HER2� breast cancer. BEZ235, a<br />

potent oral dual PI3K/mTORC1/2 inhibitor, has demonstrated growth<br />

inhibition and apoptosis in HER2� breast cancer models, including those<br />

harboring PI3K pathway alterations, and with T resistance. In a Phase I<br />

study, BEZ235 was well tolerated as a single agent in pts with advanced<br />

solid tumors. The aim <strong>of</strong> this study was to determine the MTD <strong>of</strong> BEZ235 in<br />

combination with T in pts with T-resistant HER2� metastatic breast cancer<br />

(mBC) with alterations <strong>of</strong> the PI3K pathway. Methods: Pts with T-resistant<br />

HER2� mBC (i.e. disease progression during adjuvant therapy or metastatic<br />

disease on therapy with T) received oral BEZ235 daily, with weekly T<br />

(2 mg/kg). Pts were eligible for enrollment if a tumor sample was<br />

demonstrated to contain a molecular alteration <strong>of</strong> PIK3CA and/or PTEN.<br />

Dose escalation was guided by a Bayesian logistic regression model with<br />

overdose control. Results: As <strong>of</strong> 23 Sep 2011, 15 <strong>of</strong> the 19 enrolled pts<br />

were evaluable for dose escalation analysis. BEZ235 was evaluated at 3<br />

dose levels: (1) 400 mg/day (3 pts); (2) 600 mg/day (6 pts); (3) 800<br />

mg/day (10 pts), administered either in capsule form (400 mg) or in sachet<br />

form (600 mg and 800 mg). The MTD <strong>of</strong> BEZ235 in combination with T<br />

was estimated to be 600 mg/day. Observed DLTs were G3 nausea at 600<br />

mg/day (1 pt), and G3 nausea, G3 fatigue and G3 skin rash (1 pt each) at<br />

800 mg/day. The most frequent G3/4 adverse events (CTCAE v3.0)<br />

suspected to be related to study treatment were diarrhea (4 pts) and nausea<br />

(2 pts). No deaths related to study treatment occurred. 1 pt with lung and<br />

brain metastases had a partial response. 4 pts had disease stabilization for<br />

�4 cycles (16 weeks), including 1 pt with liver metastases, in whom<br />

BEZ235/T treatment resulted in disease stabilization for more than 21<br />

cycles (84 weeks). Conclusions: BEZ235in combination with T demonstrated<br />

an acceptable safety pr<strong>of</strong>ile in pts with HER2� mBC and PI3K<br />

pathway alterations. Following the Bayesian model recommendation, the<br />

MTD for BEZ235 in combination with T was estimated to be 600 mg/day.<br />

The safety expansion arm is ongoing at the MTD.<br />

510 <strong>Clinical</strong> Science Symposium, Sat, 1:15 PM-2:45 PM<br />

SU2C phase Ib study <strong>of</strong> pan-PI3K inhibitor BKM120 with letrozole in ER�/<br />

HER2- metastatic breast cancer (MBC). Presenting Author: Ingrid A. Mayer,<br />

Vanderbilt-Ingram Cancer Center, Nashville, TN<br />

Background: Mutations in PIK3CA, the gene encoding the p110a subunit <strong>of</strong><br />

PI3K, have been associated with antiestrogen resistance in ER� BC. In general,<br />

antiestrogen-resistant cancers retain ER and responsiveness to estradiol. This<br />

suggests that treatment <strong>of</strong> ER�/PI3K mutant BC should include PI3K inhibitors<br />

plus antiestrogens. Methods: We conducted a phase Ib trial <strong>of</strong> letrozole (2.5<br />

mg/d) with the pan-PI3K inhibitor BKM120 in post-menopausal patients (pts)<br />

with ER�/HER2 MBC. BKM120 (100 mg/d) was given continuously (Arm A) or<br />

intermittently (5 on/2 <strong>of</strong>f days; Arm B). Upon toxicity, BKM120 was reduced to<br />

80 and 60 mg/d. Treatment continued until unacceptable toxicity or disease<br />

progression. Disease was assessed every 2 months. FDG-PET was done at<br />

baseline and at 2 weeks in all pts in Arm A. Results: Fifty-one pts were accrued;<br />

49 had progressed on an AI previously. Median age was 56 years (range 34-77);<br />

95% had bone and 70% visceral metastases. Toxicities and outcomes are<br />

summarized in the table. The only DLT* in Arm A and B were transaminitis and<br />

depression, respectively; both at 100 mg. Over 50% <strong>of</strong> Arm A pts had �25%<br />

reduction in their peak SUV at the 2-week FDG-PET. Pts who did not exhibit a<br />

metabolic response by FDG-PET progressed rapidly on therapy. Three pts in Arm<br />

A had a PI3K mutation; one <strong>of</strong> them has had stable disease on treatment for �12<br />

months. Conclusions: The combination <strong>of</strong> letrozole/ BKM120 is safe in pts with<br />

AI-refractory ER�/HER2 MBC. Arm B is ongoing and will be updated at the<br />

meeting. All tumor biopsies will be analyzed for PI3K pathway alterations.<br />

FDG-PET at 2 weeks appears to be a useful pharmacodynamic biomarker <strong>of</strong> PI3K<br />

pathway inhibition in most patients. Its value in predicting treatment response<br />

remains to be determined.<br />

ArmA(N�20) Arm B (N�31)<br />

Grade (%)<br />

Grade (%)<br />

Toxicity<br />

1 2 3 Total 1 2 3 Total<br />

Hyperglycemia 50 10 10 70 16 3 0 19<br />

Nausea 55 10 0 65 12 0 0 12<br />

Fatigue 25 40 5 70 9 3 0 12<br />

Transaminitis 35 25 15* 75 16 6 6 16<br />

Diarrhea 40 10 0 50 19 0 0 19<br />

Anxiety 25 15 5 45 3 3 0 6<br />

Depression 15 35 5 55 3 6 3* 12<br />

Rash 30 0 5 35 3 0 0 3<br />

Outcomes Arm A (N�20) Arm B (N�31)<br />

CR 1 0<br />

PR 1 0<br />

SD >4 months 9 4<br />

PD 7 8<br />

Non-evaluable 2 5<br />

Still on treatment 1 17<br />

Median TTP (months) 4 (2-11) N/A<br />

Discontinued due to toxicity 6 3<br />

Treatment > 8 weeks without interruption 17 N/A<br />

Breast Cancer—HER2/ER<br />

509 <strong>Clinical</strong> Science Symposium, Sat, 1:15 PM-2:45 PM<br />

PI3K pathway (PI3Kp) dysregulation and response to pan-PI3K/AKT/mTOR/<br />

dual PI3K-mTOR inhibitors (PI3Kpi) in metastatic breast cancer (MBC)<br />

patients (pts). Presenting Author: Mafalda Oliveira, Breast Cancer Group,<br />

Vall d’Hebron University Hospital, Barcelona, Spain<br />

Background: The role <strong>of</strong> PI3Kp dysregulation as a predictor <strong>of</strong> sensitivity to<br />

PI3Kpi is unclear. We aimed to evaluate the efficacy <strong>of</strong> PI3Kpi in two<br />

cohorts <strong>of</strong> MBC pts with assessable PI3Kp status. Methods: MBC pts<br />

treated in �3rd line with PI3Kpi were reviewed. PI3Kp status: (a) No<br />

dysregulation: PIK3CA wt and PTEN normal; (b) PI3Kp dysregulation:<br />

PIK3CA mutation (PIK3CAmut) or PTEN low (HScore�50). Cohort A: pts<br />

treated with single agent PI3Kpi. Cohort B: pts treated with PI3Kpi in<br />

combination with hormonal therapy (HT), chemotherapy (CT) and/or<br />

trastuzumab (T). Results: Out <strong>of</strong> 232 MBC pts screened for PI3Kp<br />

alterations from Sep09 to Sep11, 32 were treated with PI3Kpi. Cohort A<br />

(n�17): HR�/HER2- 88%, HER2� 6%, triple negative 6%; median age<br />

43, median MBC lines 4 (2-9); PIK3CAmut in 10/17 (58.8%; 6 exon9, 4<br />

exon20), PTEN low 3/17 (17.6%), 1 pt both; PI3Kp dysregulation 12/17<br />

pts. Cohort B (n�15): HR�/HER2- 40%, HER2� 60%; median age 49,<br />

median MBC lines 4 (2-13); PIK3CAmut 3/13 assessable (23.1%; all<br />

exon20), PTEN low 6/15 (40%), 1 pt both; PI3Kp dysregulation 8/15 pts.<br />

Time to progression to PI3Kpi (TTP), overall survival from MBC diagnosis<br />

(OSMBC) and OS from PI3Kpi beginning (OSPI3Kpi), according to PIK3CA<br />

status and PI3Kp dysregulation, are shown. No differences were found<br />

according to PTEN status. Conclusions: These results suggest that the best<br />

outcomes with PI3Kpi in PIK3CAmut MBC pts occur when they are used in<br />

combination with HT/CT/T. Activity <strong>of</strong> non selective PI3Kpi used as single<br />

agents seems to be limited, making results from prospective trials with<br />

selective PI3K� inhibitors and PI3Kpi in combinations eagerly awaited.<br />

Months<br />

Median<br />

(95%CI)<br />

Cohort A<br />

n�17<br />

TTP<br />

OSMBC<br />

OSPI3Ki<br />

Cohort B<br />

n�15<br />

TTP<br />

OSMBC<br />

OSPI3Ki<br />

PIK3CA PI3Kp dysregulation<br />

WT Mut p No Yes p<br />

n�7 n�10 n�5 n�12<br />

7.4<br />

(3.5-11.2)<br />

95<br />

(0-206.9)<br />

NR<br />

1.4<br />

(0-3.6)<br />

47.1<br />

(12.4-81.9)<br />

8.5<br />

(2.8-14.2)<br />

0.026<br />

0.217<br />

0.048<br />

4.9<br />

(1.5-8.2)<br />

95<br />

(-)<br />

NR<br />

1.4<br />

(0-3.3)<br />

47.1<br />

(13.6-80.7)<br />

5.7<br />

(1.8-9.5)<br />

n�10 n�3 n�7 n�8<br />

3.6 (1.2-6.1)<br />

NR<br />

NR<br />

8.4 (-)<br />

NR<br />

NR<br />

0.053<br />

0.212<br />

0.223<br />

3.6 (0.7-6.6)<br />

55.7 (53.5-57.8)<br />

5.9 (2.6-9.2)<br />

3.7 (2.2-5.2)<br />

NR<br />

NR<br />

0.092<br />

0.097<br />

0.039<br />

0.451<br />

0.216<br />

0.306<br />

511 Poster Discussion Session (Board #1), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Randomized trial <strong>of</strong> marker-directed versus standard schedule zoledronic<br />

acid for bone metastases from breast cancer. Presenting Author: Robert<br />

Edward Coleman, CR-UK/YCR Sheffield Cancer Research Centre, Sheffield,<br />

United Kingdom<br />

Background: Zoledronic acid (ZOL) reduces skeletal morbidity associated<br />

with metastatic bone disease by 30-50%. Current recommendations are for<br />

indefinite administration <strong>of</strong> intravenous 3-4 weekly (w) ZOL 4mg. Over<br />

recent years it has become clear that the risk <strong>of</strong> skeletal morbidity is related<br />

to the rate <strong>of</strong> bone resorption. We have compared a marker directed<br />

schedule <strong>of</strong> ZOL (M-ZOL), using measurements <strong>of</strong> urinary n-telopeptide <strong>of</strong><br />

type I collagen (NTX), to a standard treatment schedule (S-ZOL) in patients<br />

with bone metastases from breast cancer. Methods: The primary endpoint<br />

was skeletal related events (SRE). A non-inferiority study was designed with<br />

80% power and (one-sided) 5% alpha to demonstrate that M-ZOL retained<br />

67% <strong>of</strong> the efficacy <strong>of</strong> S-ZOL. This required 1500 patients, assuming a<br />

SRE rate <strong>of</strong> 0.7/year. Following minimisation for known prognostic factors,<br />

patients were randomised to receive S-ZOL 3-4 w or M-ZOL (15-16w; 8-9 w<br />

or 3-4w if NTX levels were �50, 50-100, �100 nmol/mmol creatinine<br />

respectively), with the schedule adjusted according to NTX measured every<br />

16 weeks. The study duration was 24 months. Results: Due to lower than<br />

expected recruitment, the study closed in 2009 following recruitment <strong>of</strong><br />

289 patients. 90% <strong>of</strong> patients had received �4 cycles <strong>of</strong> ZOL or<br />

pamidronate prior to randomisation. The median number <strong>of</strong> ZOL infusions<br />

administered to S-ZOL patients was �2x that received on M-ZOL. 46<br />

(32%) S-ZOL and 55 (38%) M-ZOL patients experienced an SRE. The<br />

numbers <strong>of</strong> SRE were 94 and 138 in the S-ZOL and M-ZOL arms, with the<br />

excess in SRE being largely due to more patients on M-ZOL experiencing<br />

�2 SRE. Multivariate analysis adjusting for key minimisation factors and<br />

baseline NTX for all SRE showed a hazard ratio for M-ZOL vs. S-ZOL <strong>of</strong> 1.41<br />

(90%CI 0.98-2.02, p�.12). NTX levels were significantly higher at all time<br />

points in the M-ZOL treated patients. Osteonecrosis <strong>of</strong> the jaw was<br />

uncommon with 3 cases with S-ZOL and 1 with M-ZOL. Conclusions: The<br />

study is underpowered to demonstrate non-inferiority in SRE outcome<br />

between the treatment strategies. However, the results suggest that the<br />

adjustment <strong>of</strong> ZOL schedule based on NTX values alone may represent<br />

sub-optimal management.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.<br />

9s


10s Breast Cancer—HER2/ER<br />

512 Poster Discussion Session (Board #2), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Effects <strong>of</strong> everolimus (EVE) on disease progression in bone and bone<br />

markers (BM) in patients (pts) with bone metastases (mets). Presenting<br />

Author: Michael Gnant, Comprehensive Cancer Center, Medical University<br />

<strong>of</strong> Vienna, Department <strong>of</strong> Surgery, Vienna, Austria<br />

Background: BOLERO-2, a multinational, double-blind, placebo-controlled,<br />

phase III study in postmenopausal women with estrogen-receptor–positive<br />

breast cancer (BC) refractory to non-steroidal aromatase inhibitors (NSAIs),<br />

showed significant clinical benefits with the addition <strong>of</strong> EVE to exemestane<br />

(EXE) (Baselga J, et al. NEJM. 2011 Epub). As bone resorption is an<br />

important factor in BC mets, it is interesting to study bone-related effects <strong>of</strong><br />

EVE. In preclinical studies, mTOR inhibition was associated with decreased<br />

osteoclast survival and activity. Exploratory analyses in BOLERO-2<br />

evaluated the effect <strong>of</strong> EVE vs placebo (PBO) on BM levels and BC<br />

progression in bone in pts with bone mets at baseline. Methods: Eligible pts<br />

were treated with EXE (25 mg once daily) and randomized (2:1) to EVE (10<br />

mg once daily) or PBO. Bone turnover markers were exploratory endpoints<br />

analyzed at 6 and 12 wks after treatment initiation, and included<br />

bone-specific alkaline phosphatase, amino-terminal propeptide <strong>of</strong> type I<br />

collagen, and C-terminal cross-linking telopeptide <strong>of</strong> type I collagen.<br />

Progressive disease in bone (PDB) was defined as worsening <strong>of</strong> a preexisting<br />

bone lesion or a new bone lesion. Results: Baseline disease<br />

characteristics, including bone mets at baseline (n � 370, 76% EVE vs n �<br />

184, 77% PBO), were well balanced between arms (N � 724), and<br />

baseline bisphosphonate use was not (44% EVE vs 55% PBO). At 12.5 mo<br />

median follow-up, progression-free survival (primary endpoint), overall<br />

response rate, and clinical benefit rate (P � .0001, all) were significantly<br />

higher with EVE (n � 485) vs PBO (n � 239). BM levels at 6 and 12 wks<br />

increased vs baseline with PBO, but decreased with EVE. The cumulative<br />

incidence rate <strong>of</strong> BC PBD was lower for EVE vs PBO at day 60 (3.03% vs<br />

6.16%, respectively) and this trend was sustained beyond 6 months.<br />

Updated results will be presented. All bone-related adverse events reported<br />

were grade1/2 and occurred with similar frequency in EVE- (2.9%) and<br />

PBO- (3.8%) treated patients. Conclusions: Exploratory analyses from<br />

BOLERO-2 suggest that adding EVE has beneficial effects on bone turnover<br />

and BC progression in bone in pts receiving EXE therapy for NSAI-refractory<br />

BC.<br />

514 Poster Discussion Session (Board #4), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Impact <strong>of</strong> body mass index (BMI) on the efficacy <strong>of</strong> zoledronic acid in<br />

premenopausal patients with hormone receptor positive breast cancer: An<br />

analysis <strong>of</strong> the ABCSG-12 trial. Presenting Author: Georg Pfeiler, Department<br />

<strong>of</strong> Obstetrics and Gynecology, Medical University <strong>of</strong> Vienna, Vienna,<br />

Austria<br />

Background: Zoledronic acid (ZOL) improves disease outcome in premenopausal<br />

patients with breast cancer (BC), especially in those with low<br />

estrogen environment. Obesity is associated with increased estrogen serum<br />

levels, which might influence the efficacy <strong>of</strong> ZOL. We investigated the<br />

impact <strong>of</strong> BMI on the efficacy <strong>of</strong> ZOL in premenopausal patients with BC.<br />

Methods: ABCSG-12 examined the efficacy <strong>of</strong> ovarian suppression using<br />

goserelin (3.6 mg q28d SC) in combination with anastrozole (ANA) or<br />

tamoxifen (TAM) � ZOL 4 mg IV q6mo) in premenopausal women with<br />

endocrine-responsive BC. BMI was calculated using the prospectively<br />

collected data on patients’ height and weight at study entry. BMI definitions<br />

<strong>of</strong> the WHO were used: normal 18.5-24.9 kg/m2 , overweight: BMI<br />

25-29.9 kg/m2 , obese BMI � 30 kg/m2 ). Disease-free survival (DFS) and<br />

overall survival (OS) were calculated by Kaplan-Meier method and results<br />

were compared by using the log-rank test and Cox proportional hazard<br />

modeling. Results: Two thirds <strong>of</strong> the patients were normal weight (1,111)<br />

and one third was overweight (391) or obese (182). Normal weight patients<br />

treated with ZOL experienced the same benefit as overweight/obese<br />

patients treated with ZOL compared to patients without ZOL regarding<br />

disease free survival (HR 0.75, 95%CI 0.53 1.05 p � 0.09 and HR 0.71,<br />

95%CI 0.46 1.1, p�0.12, respectively) as well as overall survival (HR<br />

0.67, 95% CI 0.37 1.22, p�0.19 and HR 0.64, 95%CI 0.32 1.28,<br />

p�0.19, respectively). No difference regarding DFS and OS could be<br />

detected between normal weight and overweight/ obese patients treated<br />

with ZOL (HR 1.05, 95%CI 0.69 1.59, p�0.83 and HR 1.27, 95%CI 0.62<br />

2.61, p� 0.51). Comparing normal weight patients with ZOL to obese<br />

patients with ZOL, again no difference could be observed. Conclusions: The<br />

significant outcome benefit <strong>of</strong> adjuvant zoledronic acid in ABCSG-12 can<br />

be demonstrated both in normal weight and overweight/obese premenopausal<br />

patients with early breast cancer, indicating that the bone marrow<br />

microenvironment changing impact <strong>of</strong> adjuvant bisphopshonate treatment<br />

does not correlate with patients’ BMI.<br />

513 Poster Discussion Session (Board #3), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Adjuvant zoledronic acid (ZOL) in postmenopausal women with breast<br />

cancer and those rendered postmenopausal: Results <strong>of</strong> a meta-analysis.<br />

Presenting Author: Walter Gregory, <strong>Clinical</strong> Trials Research Unit, University<br />

<strong>of</strong> Leeds, Leeds, United Kingdom<br />

Background: A number <strong>of</strong> groups have now reported results <strong>of</strong> randomised<br />

trials <strong>of</strong> adjuvant zoledronic acid (ZOL) for women with early stage breast<br />

cancer. Following an interaction effect observed in the AZURE trial, where<br />

postmenopausal women appeared to benefit from ZOL, we performed a<br />

meta-analysis, from both publications and presented data, <strong>of</strong> the most<br />

recent results from these trials. We also looked at recently reported large<br />

studies <strong>of</strong> clodronate and ibandronate in older or postmenopausal women<br />

to further examine this hypothesis. Methods: DFS data from 8735 women in<br />

7 trials (AZURE, ABCSG-12, ZO-FAST, Z-FAST, EZO-FAST, NSABP-B34,<br />

GAIN) that included a randomization to ZOL vs. control or clodronate/<br />

ibandronate vs. control were examined. Median follow-up was 5 years. 14%<br />

<strong>of</strong> women experienced a DFS event. The ABCSG-12 study was included<br />

since, although it included only premenopausal women, they were all<br />

treated with goserelin, effectively rendering them postmenopausal, prior to<br />

and during treatment with ZOL. We analysed the subgroup <strong>of</strong> women aged<br />

�50 in the NSABP-B34 study in the absence <strong>of</strong> information on menopausal<br />

status, and included only postmenopausal women from the AZURE<br />

and GAIN trials. Sensitivity analyses confirmed that exclusion <strong>of</strong> some<br />

older, small, clodronate studies that lacked DFS data, or failed to report by<br />

age or menopausal status would be unlikely to alter these findings. Results:<br />

For the 5 ZOL studies alone, there was a very substantial and highly<br />

significant DFS benefit for ZOL, 2P�.0006, with a hazard ratio <strong>of</strong> 0.76,<br />

and therefore a DFS risk reduction <strong>of</strong> 24% (95% CI 11%-35%). The results<br />

were, in the main, extremely consistent, with the exception <strong>of</strong> the very small<br />

E-ZO-FAST study, which had only 30 events. The risk reduction lessened to<br />

18% (95% CI 8%-26%) when adding in the clodronate and ibandronate<br />

studies, but the result remained highly significant (2P�.00075).<br />

Conclusions: These meta-analysis results, including more than 8000<br />

women in a range <strong>of</strong> studies, now provide strong evidence that ZOL is an<br />

effective adjuvant treatment for postmenopausal women with early stage<br />

breast cancer, and that the treatment benefits are substantial.<br />

515 Poster Discussion Session (Board #5), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Progression <strong>of</strong> genomic signatures in local and metastatic estrogen receptorpositive<br />

(ER�) breast cancer: Relevance to palliative treatment. Presenting<br />

Author: William Fraser Symmans, University <strong>of</strong> Texas M. D. Anderson<br />

Cancer Center, Houston, TX<br />

Background: Biological progression <strong>of</strong> ER� breast cancer accelerates<br />

clinical progression and resistance to treatments. Methods: One laboratory<br />

used Affymetrix U133A gene expression microarrays to pr<strong>of</strong>ile 588 biopsy<br />

samples from patients with ER� breast cancer: 74 AJCC Stage I, 155<br />

Stage IIA, 105 Stage IIB, 127 Stage III, 127 Stage IV (27 at presentation,<br />

100 relapsed). We evaluated stage dependence <strong>of</strong> ER [ESR1, PGR,<br />

sensitivity to endocrine therapy (SET) index], proliferation [MKI67, AU-<br />

RKA, genomic grade index (GGI)], invasion [PLAU (uPA)], PI3-kinase<br />

(PIK3CA-GS), VEGF, genomic subtype [PAM50, 3-gene classifier (ESR1,<br />

ERBB2, AURKA)], and housekeeper control genes. Significance was<br />

evaluated through ordinal median regression (P � 0.002, for multiple<br />

testing) after adjusting for staging method (clinical or pathologic). Exploratory<br />

Cox regression analyses <strong>of</strong> progression-free survival (PFS) and overall<br />

survival (OS) were performed when treatment was hormonal therapy (HT,<br />

N�58) or chemotherapy (CT, N�27) after biopsy <strong>of</strong> metastatic ER� breast<br />

cancer (MBC). Results: Stage progression was associated with reduced SET<br />

index and increased proliferation (GGI, MKI67, AURKA) and metabolism<br />

(GAPDH). These changes occurred between Stages IIB and III, and Stages<br />

III and IV. Luminal B and proliferation subtypes were more prevalent in<br />

Stage IV and less in Stage I. Interestingly, invasion (PLAU) genes were<br />

lower in MBC. Only SET index demonstrated a significant interaction with<br />

treatment (HT or CT) for MBC (PFS: p�0.018). SET was predictive <strong>of</strong> PFS<br />

and OS following HT, as a continuous score (PFS: HR�0.69, 95%CI 0.49<br />

to 0.97, p�0.035; OS: HR�0.61, 95%CI 0.40 to 0.94, p�0.025) or<br />

dichotomized at median value (PFS: HR�0.43, 95%CI 0.24 to 0.76,<br />

p�0.003; OS: HR�0.37, 95%CI 0.18 to 0.77, p�0.006). Genomic<br />

subtype was prognostic for PFS irrespective <strong>of</strong> treatment type. High<br />

PIK3CA-GS expression predicted OS in the HT subset. Conclusions: Stage<br />

progression was associated with decreased ER-related transcription (SET)<br />

and increased proliferation, grade, higher risk subtype, and metabolism. In<br />

MBC samples, only SET index was predictive <strong>of</strong> PFS and OS with palliative<br />

hormonal therapy.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


516 Poster Discussion Session (Board #6), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Mammostrat as an immunohistochemical multigene assay for prediction <strong>of</strong><br />

early relapse risk in the TEAM pathology study. Presenting Author: John<br />

M. S. Bartlett, Ontario Institute for Cancer Research, Toronto, ON, Canada<br />

Background: Some postmenopausal patients with hormone sensitive early<br />

breast cancer remain at high risk <strong>of</strong> relapse despite endocrine therapy, and<br />

might benefit additionally from adjuvant chemotherapy. The challenge is to<br />

prospectively identify such patients. The Mammostrat test uses five<br />

immunohistochemical markers to stratify patients regarding recurrence<br />

risk, and may inform treatment decisions. We tested the efficacy <strong>of</strong> this<br />

panel in the TEAM trial. Methods: Pathology blocks from 4598 TEAM<br />

patients were collected and TMAs constructed. The cohort was 47% node<br />

positive and 36% were also treated with adjuvant chemotherapy. Triplicate<br />

0.6mm2 TMA cores were stained and positivity for p53, HTF9C, CEACAM5,<br />

NDRG1, SLC7A5 assessed. Cases were assigned a Mammostrat risk score,<br />

and distant relapse free (DRFS) and disease free survival (DFS) analysed.<br />

Results: In multivariate regression analyses, corrected for conventional<br />

clinicopathological markers, Mammostrat provided significant additional<br />

information on DRFS after endocrine therapy in ER positive node negative<br />

patients (N�1226) not receiving chemotherapy (p�0.004). Further analyses<br />

in all patients not exposed to chemotherapy, irrespective <strong>of</strong> nodal status<br />

(N�2559) and in the entire cohort (N�3837) showed Mammostrat scores<br />

provide additional information on DRFS in these groups (p�0.001 and<br />

p�0.0001 respectively; multivariate analyses). No differences were seen<br />

between the two endocrine treatment regimens. Conclusions: The Mammostrat<br />

score predicts DRFS for both exemestane and tamoxifenexemestane<br />

treated patients irrespective <strong>of</strong> nodal status and chemotherapy.<br />

The ability <strong>of</strong> this test to provide additional outcome data following<br />

treatment provides further evidence for its’ utility in risk stratification <strong>of</strong> ER<br />

positive postmenopausal breast cancer patients.<br />

518 Poster Discussion Session (Board #8), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Gene methylation in random FNA samples as biomarkers for breast cancer.<br />

Presenting Author: Vered Stearns, Sidney Kimmel Comprehensive Cancer<br />

Center at Johns Hopkins University, Baltimore, MD<br />

Background: Current methods to determine breast cancer risk are insufficiently<br />

sensitive to select women most likely to benefit from preventive<br />

strategies. We hypothesized that candidate gene promoter hypermethylation<br />

may provide an individualized risk pr<strong>of</strong>ile. We performed a prospective<br />

study to determine whether DNA cumulative methylation index (CMI)<br />

varies by menstrual phase or menopausal status, and to correlate CMI with<br />

established risk factors. Methods: We obtained random fine needle aspiration<br />

(rFNA) samples from healthy women age 35-60 and determined their<br />

menopausal and menstrual status, lifetime Gail risk, mammographic breast<br />

density, and cytologic atypia assessed as the Masood score. We evaluated<br />

CMI <strong>of</strong> 11 candidate genes in rFNA cells using the Quantitative Multiplex<br />

Methylation-Specific PCR (QM-MSP) technique. We used Wilcoxon test<br />

and ANOVA model to compare CMI across menopausal and menstrual<br />

(follicular, mid-cycle, luteal) categories, respectively. We used linear<br />

regression model to adjust for age and BMI. Methylation scores were<br />

log-transformed in the analysis. Results: We enrolled 390 women at the<br />

Avon Breast Centers at Johns Hopkins and Northwestern, the majority<br />

through the Love/Avon Army <strong>of</strong> Women, and 380 completed study<br />

procedures. Median age 50 (36-60), mean BMI 28 (18.7-50.8), 52% were<br />

postmenopausal. Mean life-time Gail risk 14.6 (5.6-54.1), mean percent<br />

mammographic density 19.6 (2.5-72.8), and mean Masood score (N�354)<br />

13.6 (7-18). QM-MSP analysis was completed on 229 samples. We did not<br />

observe differences in CMI among menopausal (P�0.4895) or menstrual<br />

categories (P�0.2333). There was no association between CMI and<br />

life-time Gail risk (P�0.706) or breast density (P�0.4116). We observed a<br />

significant correlation between CMI and Masood score (P�0.0167).<br />

Conclusions: CMI correlates with degree <strong>of</strong> cytologic atypia and is potentially<br />

a robust indicator <strong>of</strong> breast cancer risk since it does not vary with<br />

menstrual or menopausal status. Next, we will select genes that best reflect<br />

changes in the clinical parameters to create a gene methylation signature<br />

that will be validated in other studies and correlated with breast cancer risk.<br />

Breast Cancer—HER2/ER<br />

11s<br />

517 Poster Discussion Session (Board #7), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Validation <strong>of</strong> IHC4 algorithms for prediction <strong>of</strong> risk <strong>of</strong> recurrence in early<br />

breast cancer using both conventional and quantitative IHC approaches.<br />

Presenting Author: Jason Christiansen, HistoRx, Inc, Branford, CT<br />

Background: Hormone receptors, HER2 and Ki67 are residual risk markers<br />

in early breast cancer. Combining these markers into a unified algorithm<br />

(IHC4) provides information on residual recurrence risk <strong>of</strong> patients treated<br />

with hormone therapies. This study aimed to independently investigate the<br />

validity <strong>of</strong> the IHC4 algorithm for residual risk prediction using both<br />

conventional (DAB)-IHC and quantitative immun<strong>of</strong>luorescence (QIF-<br />

AQUA). Methods: The TEAM pathology study recruited �4500 samples<br />

from patients treated in the TEAM trial. TMAs were stained for ER, PgR,<br />

HER2 and Ki67 using QIF-AQUA technology or DAB-based immunohistochemistry<br />

(DAB-IHC). Central HER2 FISH was performed. Quantitative<br />

image analysis was used to generate expression scores that were normalized<br />

to produce “IHC4 algorithm” as well as novel algorithm scores.<br />

Algorithm scores were compared with disease recurrence in univariate and<br />

multivariate Cox Proportional Hazards models. Results: Both DAB-IHC and<br />

QIF-AQUA IHC4 continuous models were significant (P�0.0001) for<br />

prediction <strong>of</strong> disease recurrence with a continuous Hazard Ratio (HR) <strong>of</strong><br />

1.011 (1.010 – 1.013) for QIF-AQUA IHC4 versus 1.008 (1.007 – 1.010)<br />

for the DAB-IHC IHC4 model using the published IHC4 algorithm (Cuzick<br />

et al 2011). Binning continuous model scores (4 bins) by Kaplan-Meier<br />

survival analysis was used to graphically illustrate these effects. De novo<br />

models for both DAB-IHC and QIF-AQUA were also significantly (P�0.0001)<br />

predictive <strong>of</strong> residual risk in early breast cancer. Additionally, all 4 models<br />

were independent predictors <strong>of</strong> recurrence (P�0.0001) with other recognized<br />

clinical prognostic factors in multivariate analysis. Although results<br />

from DAB and QIF-AQUA were modestly correlated, the QIF-AQUA model<br />

showed enhanced prediction <strong>of</strong> recurrence in both Cox Proportional<br />

Hazards Modeling and C-index calculations. Conclusions: Either conventional<br />

DAB or QIF-AQUA methods <strong>of</strong> IHC provided evidence supporting the<br />

clinical utility <strong>of</strong> IHC4 algorithms in the context <strong>of</strong> the TEAM study. With<br />

careful standardization, either <strong>of</strong> these IHC4 assays should be considered<br />

for prediction <strong>of</strong> residual risk in early breast cancer.<br />

519 Poster Discussion Session (Board #9), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Dual effects <strong>of</strong> metformin on breast cancer proliferation in a randomized trial.<br />

Presenting Author: Andrea De Censi, E. O. Ospedali Galliera, Genoa, Italy<br />

Background: Metformin lowers breast cancer risk in observational studies in<br />

diabetics, but evidence for its clinical activity is scanty. We studied the<br />

metformin antiproliferative effect in a pre-surgical study. Since the antidiabetic<br />

effect <strong>of</strong> metformin is heterogeneous according to obesity and insulin resistance<br />

(IR), we also determined whether its antiproliferative effect was modified by risk<br />

biomarkers. Methods: After tumor biopsy, we randomly allocated 200 nondiabetic<br />

women with breast cancer to either metformin, 850 mg/bid (n�100) or<br />

placebo (n�100) for 4 wks. The primary endpoint was the post-pretreatment<br />

change in Ki-67 between arms. We explored effect modifications by STEPP and<br />

tested biomarker thresholds that showed an interaction with treatment on Ki-67.<br />

Results: Overall, median (IQR) Ki-67 was 19 (14-31) at baseline and 21 (14-32)<br />

after 4 wks and 18 (12-29) at baseline and 20 (13-31) after 4 wks in the<br />

metformin and placebo arm, respectively with mean increase <strong>of</strong> 4.0% (95%CI,<br />

-5.6 to 14.4) on metformin versus placebo. Multivariate analyses showed an<br />

increase <strong>of</strong> Ki-67 after 4 wks placebo in the following subgroups: HOMA� IR<br />

threshold, IGFBP3� highest quartile, IGFBP1� lowest quintile, IGFratio (IGF1/<br />

IGFBP3)� median, CRP� inflection point at STEPP analysis, HER2�ve tumors<br />

(versus –ve). Metformin blunted the increase <strong>of</strong> Ki-67 noted on placebo in these<br />

subgroups. Conclusions: Overall, metformin did not affect Ki-67 in most subjects<br />

with breast cancer. However, in exploratory analysis we identified subgroups <strong>of</strong><br />

patients where metformin showed antiproliferative effect. Further studies to a<br />

personalized approach are warranted with selection <strong>of</strong> study populations.<br />

Post-pretreatment median change in Ki-67 by treatment arm and biomarker<br />

thresholds.<br />

Risk biomarker threshold Placebo Metformin p interaction<br />

HOMA>2.8 0(n�29) 0 (n�24) 0.03<br />

HOMA4614ng/ml 0(n�27) 0 (n�23) 0.04<br />

IGFBP-32mg/L 2(n�25) 0 (n�37) 0.08<br />

CRP


12s Breast Cancer—HER2/ER<br />

520 Poster Discussion Session (Board #10), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Reduction in Ki-67 in benign breast tissue <strong>of</strong> high-risk premenopausal<br />

women with the SERM acolbifene. Presenting Author: Carol J. Fabian,<br />

University <strong>of</strong> Kansas Medical Center, Kansas City, KS<br />

Background: Selective Estrogen Receptor Modulators (SERMs) are approved<br />

for reduction <strong>of</strong> risk for breast cancer; however, uptake and use is<br />

limited. We conducted a pilot study <strong>of</strong> a 4th generation SERM to determine<br />

tolerability and effect on tissue biomarkers in healthy women at high risk<br />

for development <strong>of</strong> breast cancer. Methods: Premenopausal women at<br />

elevated risk for breast cancer were screened by random periareolar fine<br />

needle aspiration (RPFNA) performed during the follicular phase <strong>of</strong> the<br />

menstrual cycle. Women were eligible if breast epithelial cells exhibited<br />

evidence <strong>of</strong> cytologic hyperplasia with or without atypia, as well as Ki-67<br />

�2% by immunocytochemistry. Following 6-8 months <strong>of</strong> open-label<br />

acolbifene (20 mg/d), the RPFNA was repeated. The primary endpoint was<br />

modulation <strong>of</strong> the proportion <strong>of</strong> cells that expressed Ki-67. Body composition<br />

(DEXA), pelvic sonography, mammographic breast density, and serum<br />

levels <strong>of</strong> IGF-1/IGFBP3 and several bioavailable hormones were assessed<br />

pre and post intervention. Results: 76 women were screened by RPFNA,<br />

with 25 eligible and enrolled in the intervention over a 9 month period. All<br />

25 (7 on oral contraceptives) subjects completed the study, had a second<br />

RPFNA, and were evaluable. Median Ki-67 at baseline was 4.6% (range<br />

2.4 – 21.9%) and <strong>of</strong>f study 1.4% (range 0 – 6.6%); median change was a<br />

reduction <strong>of</strong> 3.0% (range -20.2% to �2.8%; decreased in 23, increased in<br />

2) or a relative reduction <strong>of</strong> 77%. The end-<strong>of</strong>-study Ki-67 was significantly<br />

less than baseline (p�0.001, 2-tailed Wilcoxon test). There were no<br />

statistically significant changes in cytomorphology over this short intervention<br />

period. There was a marginal effect on breast density (16 decreased; 8<br />

increased; p�0.067). Adverse events were minimal with greatest grade <strong>of</strong><br />

3 reported by 2 subjects, grade 2 by 7 subjects, and grade 1 by 11<br />

subjects. No serious adverse event was reported and no subject discontinued<br />

the study due to an AE. Conclusions: Based on preliminary evaluation<br />

showing favorable modulation <strong>of</strong> proliferation and minimal adverse events,<br />

further investigation <strong>of</strong> acolbifene, a fourth generation SERM, as a breast<br />

cancer chemoprevention agent for premenopausal women appears warranted.<br />

Supported by NO1-CN-35135.<br />

522 Poster Discussion Session (Board #12), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Mucin-1 protein and mRNA expression in breast cancer: Predictive value<br />

for therapy response and survival after neoadjuvant chemotherapy. Presenting<br />

Author: Bruno Valentin Sinn, Charité Universitätsmedizin Berlin,<br />

Berlin, Germany<br />

Background: Mucin-1 (MUC1) is expressed in normal epithelial cells and in<br />

breast cancer. Immunotherapeutic agents targeting MUC1 are currently<br />

tested in clinical trials. Methods: We evaluated the frequency <strong>of</strong> MUC1<br />

expression by immunohistochemistry (IHC; n � 691) and quantitative<br />

RT-PCR (qRT-PCR; n � 286) in formalin-fixed paraffin-embedded (FFPE)<br />

pre-treatment biopsies from patients <strong>of</strong> the GeparTrio trial (NCT 00544765).<br />

mRNA levels were analyzed as a continuous variable, a distribution-based<br />

cut-<strong>of</strong>f was chosen for IHC data. The predictive value for therapy response<br />

and survival after neoadjuvant anthracycline/taxane-based chemotherapy<br />

(CTX) was evaluated. Results: MUC1 was detectable by IHC in 95%.<br />

qRT-PCR covered a dynamic range <strong>of</strong> 3 orders <strong>of</strong> magnitude. IHC and<br />

mRNA data were correlated (p � 0.001). MUC1 was detected in higher<br />

quantities in HR� tumors (p � 0.001), the lowest levels were found in<br />

HR-/HER2- tumors (p � 0.001). High MUC1 protein and mRNA expression<br />

were associated with lower probability <strong>of</strong> pathologic complete response<br />

(pCR) in the overall population (p � 0.001) and in HR� (p � 0.004 and �<br />

0.001), HER2- (p � 0.001) and HR�/HER2- tumors (p � 0.001). In<br />

multivariable logistic regression, MUC1 was independently predictive (high<br />

MUC1 protein: odds ratio (OR) 0.464, 95% CI 0.300 – 0.719, p � 0.001;<br />

MUC1 mRNA (per 20-dCT unit): OR 0.673, CI 0.546 – 0.829, p � 0.001).<br />

MUC1 protein and mRNA expression were associated with longer patient<br />

survival in the overall population (p � 0.03 and � 0.001) and in HER2tumors<br />

(p � 0.005 and � 0.001). MUC1 mRNA was also prognostic in<br />

HR� (p � 0.001) and HR�/HER2- tumors (p � 0.001). In multivariable<br />

analysis, protein and mRNA expression were independently prognostic<br />

(high MUC1 protein: hazard ratio (HR) 0.388, CI 0.166 – 0.907, p �<br />

0.029; MUC1 mRNA per 20-dCT unit: HR 0.763, CI 0.595 – 0.909, p �<br />

0.005). Conclusions: MUC1 is frequently expressed in breast cancer and<br />

detectable by IHC and qRT-PCR from FFPE tissue. Despite its association<br />

with HR� status, it provides independent predictive information for<br />

therapy response and survival after neoadjuvant CTX. In clinical immunotherapy<br />

trials, MUC1 expression may serve as a predictive marker.<br />

521 Poster Discussion Session (Board #11), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Aspirin use and mortality in women with stage I-III breast cancer: A<br />

population-based study. Presenting Author: Thomas Ian Barron, Trinity<br />

College Dublin, Dublin, Ireland<br />

Background: Recent observational studies have associated aspirin (ASP)<br />

use with large reductions in breast cancer (BC) mortality. However, these<br />

studies have provided limited information on dose or duration <strong>of</strong> ASP use,<br />

key issues relevant to translation <strong>of</strong> the results into clinical practice.<br />

Methods: Linked National Cancer Registry Ireland and prescription refill<br />

data (General Medical Services Ireland, GMS) were used to identify women<br />

aged 50-80 with incident stage I-III BC (2001-2006). ASP use was defined<br />

as high or low by the median number <strong>of</strong> ASP days’ supply (85 days) in the<br />

90 days pre-diagnosis. Full capture <strong>of</strong> ASP use is expected as the GMS<br />

provides all medications, including ASP, without charge. Hazard ratios<br />

(HR) with 95% confidence intervals (CI) for ASP use and (i) all-cause and<br />

(ii) BC-specific mortality were estimated using Cox proportional hazards<br />

models adjusted for age, stage, grade, ER, PR, HER-2 status, comorbidity<br />

and other drug exposures. Analyses were stratified by tumor stage and nodal<br />

status. Results: 2714 women with stage I-III BC were identified (median<br />

follow-up � 3.3 years), <strong>of</strong> whom 642 (23.7%) used ASP in the 90 days pre<br />

diagnosis. High and low ASP exposure groups were strongly predictive <strong>of</strong><br />

post-diagnosis ASP exposure levels (High: mean post diagnosis exposure<br />

duration – 83% <strong>of</strong> follow-up; dose – 75mg/day in 91% <strong>of</strong> women. Low:<br />

mean post-diagnosis exposure duration – 58% <strong>of</strong> follow-up; dose –<br />

75mg/day in 80% <strong>of</strong> women). Women with any ASP use had a nonsignificant<br />

reduction in all-cause (HR 0.85 95%CI 0.68, 1.06) and<br />

BC-specific (HR 0.86 95%CI 0.65, 1.15) mortality, compared to ASP<br />

unexposed women. However, in the dose response analysis high ASP<br />

exposure was associated with a significant reduction in all-cause (HR 0.70<br />

95% CI 0.51, 0.95) and BC-specific (HR 0.63 95% CI 0.42, 0.96)<br />

mortality. No reduction was observed for low ASP exposure. The benefits <strong>of</strong><br />

ASP exposure were greater in early stage, node negative disease. Conclusions:<br />

Only high ASP exposure was associated with a significant reduction in all<br />

cause and BC-specific mortality. These findings may explain inconsistent<br />

results from previous studies. Our findings can inform future clinical trials.<br />

523 Poster Discussion Session (Board #13), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Impact <strong>of</strong> expression levels <strong>of</strong> mRNA HER2 and ESR1 on the pathologic<br />

complete remission (pCR) rate after neoadjuvant treatment with anthracycline-taxane<br />

containing chemotherapy in combination with trastuzumab in<br />

the GeparQuattro trial. Presenting Author: Jens Bodo Huober, University <strong>of</strong><br />

Tuebingen and Kantonsspital St. Gallen, St. Gallen, Switzerland<br />

Background: Recent data suggest that benefit from (neo)adjuvant trastuzumab<br />

might be related to expression <strong>of</strong> HER2 and estrogen receptor<br />

(ESR1). We investigated mRNA levels <strong>of</strong> HER2 and ESR1 in core biopsies<br />

in HER2 positive tumors <strong>of</strong> the GeparQuattro trial and compared it to<br />

response to neoadjuvant treatment. Methods: In the GeparQuattro trial 445<br />

HER2� pts were included based on local testing and received neoadjuvant<br />

trastuzumab and chemotherapy (either4xEC¡4 x docetaxel (D) or4xEC<br />

¡ 4 x D/capecitabine (C) or 4xEC¡4x D ¡4 X C). In 217 available<br />

pretherapeutic core biopsies HER2 levels were analysed by IHC, SISH and<br />

quantitative RT-PCR using predefined cut<strong>of</strong>fs; pCR was defined as ypT0is;<br />

ypN0. Results: Only 73% <strong>of</strong> the tumors (158 <strong>of</strong> 217) were centrally HER2<br />

positive (cHER2�) by IHC/SISH, while 59 tumors (27%) were centrally<br />

HER2 negative (cHER2-). As all pts had received neoadjuvant trastuzumab,<br />

this gave us the possibility to evaluate response <strong>of</strong> HER2- tumors to<br />

trastuzumab. The pCR rate <strong>of</strong> cHER2� tumors was significantly increased<br />

(46.8%, p�0.0005) compared to cHER2-, who had a pCR rate <strong>of</strong> only<br />

20.3%. Similar results were obtained if HER2 positivity was determined by<br />

RT-PCR (pCR rate 50% vs. 17.4%, p�0.0005). Assessment <strong>of</strong> HER2<br />

status by RT-PCR showed a concordance <strong>of</strong> 86.2% with the IHC/SISH<br />

status. In uni- and multivariate logistic regression analysis <strong>of</strong> cHER2�<br />

cases including continuous HER2 and ESR1 mRNA levels the HER2<br />

mRNA expression was significantly associated with prediction for a pCR in<br />

(univ.: OR 1.43, 95% CI 1.11-1.83, p�0.005; multiv.: OR 1.42, 95% CI<br />

1.11-1.83, p�0.006). In the cHER2� tumors, the pCR rate was similar<br />

for ESR1 mRNA positive (47.4%) and ESR1 negative tumors (46.3%).<br />

Conclusions: Only pts with cHER2� tumors independently <strong>of</strong> the method<br />

used have an additional benefit in terms <strong>of</strong> a pCR from adding trastuzumab<br />

to chemotherapy. In cHER2-negative patients the pCR rate is comparable<br />

to the pCR rate in the non trastuzumab treated population. Increasing<br />

HER2 mRNA levels were associated with a better response to trastuzumab<br />

based treatment in cHER2 � tumors.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


524 Poster Discussion Session (Board #14), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Vasomotor (VM) and musculoskeletal (MSK) symptoms and association<br />

with outcomes on extended adjuvant letrozole therapy: Analyses from NCIC<br />

CTG MA.17. Presenting Author: Pedro Emanuel Rubini Liedke, Massachusetts<br />

General Hospital, Avon International Breast Cancer Center, Boston,<br />

MA<br />

Background: Surrogate markers <strong>of</strong> clinical outcomes can potentially guide<br />

cancer therapy. New onset MSK symptoms and VM symptoms on aromatase<br />

inhibitors (AI) for hormone receptor positive (HR�) early breast cancer<br />

(EBC) have been investigated as markers <strong>of</strong> clinical outcomes, but results<br />

have been conflicting. MA.17 showed improved disease free survival (DFS)<br />

with letrozole (L) as extended adjuvant therapy after 5 years <strong>of</strong> tamoxifen<br />

(T). We performed this exploratory analysis <strong>of</strong> new onset symptoms and<br />

their association with clinical outcomes in the MA.17 trial. Methods:<br />

Patients (pts) with HR� EBC were randomized to receive L or placebo (P)<br />

for 5 years after 5 years <strong>of</strong> T. Symptoms were collected according to CTC v.<br />

2.0 at baseline, 1 month (mo), 6 mos, and every 12 mos thereafter.<br />

Analyses included pts with new symptoms <strong>of</strong> any grade who received<br />

therapy and excluded pts with baseline symptoms, EBC not HR�, and<br />

deaths/ relapses prior to each time point. Symptoms were categorized as<br />

VM - hot flashes/flushes, flushing, or sweating; and MSK - arthritis,<br />

arthralgia, myalgia, bone pain, or MSK other. Multivariate Cox Models<br />

adjusting for age, nodal status, duration <strong>of</strong> T and prior chemotherapy were<br />

used for analysis <strong>of</strong> DFS, distant DFS (DDFS), and overall survival (OS)<br />

assessed before unblinding. Hazard Ratios [HR] and 95% confidence<br />

intervals (CI) for patients with either VM or MSK symptoms are presented<br />

below for L using pts with no symptoms as controls. Results: Emergent VM<br />

symptoms were associated with improved DFS and DDFS on L (Table). No<br />

association was found for MSK symptoms at any time point. No association<br />

between either MSK or VM and OS was found at any time point.<br />

Conclusions: New VM symptoms initiating with extended letrozole were<br />

associated with improved outcomes. Similar findings have been seen with<br />

up-front AI therapy. Our results should be confirmed as they are <strong>of</strong> potential<br />

significance in guiding therapy.<br />

Month <strong>of</strong> assessment<br />

VM<br />

1<br />

HR (95% CI)<br />

6 12<br />

DFS 0.22 (0.05-0.92) 0.25 (0.07-0.85) 0.42 (0.15-1.18)<br />

DDFS<br />

MSK<br />

0.17 (0.02-1.26) 0.13 (0.02-0.99) 0.54 (0.16-1.81)<br />

DFS 2.33 (0.54-10.04) 0.70 (0.24-2.07) 0.68 (0.24-1.91)<br />

DDFS 1.91 (0.25-14.74) 0.57 (0.13-2.58) 0.45 (0.10-2.15)<br />

526 Poster Discussion Session (Board #16), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Common polymorphisms in the estrogen receptor-1 may determine risk <strong>of</strong><br />

hot flashes in early breast cancer patients using tamoxifen. Presenting<br />

Author: Vincent O. Dezentje, Department <strong>of</strong> <strong>Clinical</strong> Oncology, Leiden<br />

University Medical Center, Leiden, Netherlands<br />

Background: In breast cancer patients the occurrence <strong>of</strong> hot flashes as<br />

common side effect <strong>of</strong> tamoxifen therapy may be associated with effective<br />

estrogen receptor antagonism dependent on genetic variations <strong>of</strong> metabolic<br />

enzymes and the estrogen receptor. Methods: 742 early breast cancer<br />

patients who were randomized to receive tamoxifen, followed by exemestane<br />

after 2.5 to 3 years within the Tamoxifen Exemestane Adjuvant<br />

Multinational (TEAM) Trial were genotyped for 30 germ line genetic<br />

variants <strong>of</strong> 11 enzymes that are involved in the tamoxifen metabolism and<br />

the estrogen receptor 1 (ESR1). These genetic variants were related to the<br />

occurrence <strong>of</strong> hot flashes during the first year <strong>of</strong> tamoxifen use (primary<br />

aim) and during the complete tamoxifen treatment period (secondary aim).<br />

A multivariable logistic regression was used to adjust for age and adjuvant<br />

chemotherapy. Results: No genetic variant was associated with the occurrence<br />

<strong>of</strong> hot flashes during the first year. Higher age was related to a lower<br />

incidence <strong>of</strong> hot flashes in the first year (adjusted odds ratio 0.94, 95% CI<br />

0.92-0.96; p�0.001). The ESR1 PvuII XbaI CG haplotype (CG/CG vs<br />

CG/other � other/other: adjusted odds ratio 0.44, 95% CI 0.21-0.92;<br />

p�0.03), ESR1 PvuII XbaI TA haplotype (TA/TA � TA/other vs other/other:<br />

adjusted odds ratio 1.86, 95% CI 1.09-3.14; p�0.02) and age (adjusted<br />

odds ratio 0.94, 95% CI 0.92-0.97; p�0.001) were associated with the<br />

occurrence <strong>of</strong> hot flashes during the total tamoxifen treatment period. No<br />

association was found between the CYP2D6 predicted phenotype and hot<br />

flashes. Conclusions: Common polymorphisms in the estrogen receptor-1<br />

might help to predict the occurrence <strong>of</strong> hot flashes in breast cancer patients<br />

treated with adjuvant tamoxifen. If replicated, this may provide clinicians<br />

with a tool to <strong>of</strong>fer more personalized hormonal therapy.<br />

Breast Cancer—HER2/ER<br />

13s<br />

525 Poster Discussion Session (Board #15), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Genetic variants associated with toxicity-related discontinuation <strong>of</strong> adjuvant<br />

aromatase inhibitor (AI) therapy. Presenting Author: Norah Lynn<br />

Henry, University <strong>of</strong> Michigan Medical Center, Ann Arbor, MI<br />

Background: Discontinuation <strong>of</strong> adjuvant AI therapy due to intolerable<br />

symptoms occurs in up to 30% <strong>of</strong> patients. Predictors <strong>of</strong> which patients will<br />

be unable to tolerate these medications have not been defined. We<br />

hypothesized that inherited variants in candidate genes are associated with<br />

treatment discontinuation because <strong>of</strong> AI-associated toxicity. Methods: We<br />

prospectively evaluated reasons for treatment discontinuation in women<br />

with hormone receptor positive breast cancer initiating adjuvant AI through<br />

a multicenter, prospective, randomized clinical trial <strong>of</strong> exemestane (exe)<br />

versus letrozole (let). Using multiple genetic models, we evaluated potential<br />

associations between 136 single nucleotide polymorphisms (SNP) in<br />

24 candidate genes, selected a priori, primarily with roles in estrogen<br />

metabolism and signaling, and discontinuation <strong>of</strong> AI therapy because <strong>of</strong><br />

toxicity. To account for multiple comparisons, statistical significance was<br />

defined as p�0.0003. Results: Of the 467 enrolled patients who had<br />

available germ line DNA, 152 (33%) discontinued AI therapy because <strong>of</strong><br />

toxicity. After adjusting for multiple covariates, multivariable analyses<br />

revealed that two inherited genetic variants in ESR1, which encodes<br />

estrogen receptor (ER) alpha, and one in CYP19A1 were associated with<br />

increased risk <strong>of</strong> discontinuation <strong>of</strong> AI therapy because <strong>of</strong> toxicity (Table). A<br />

variant in NCOR1 (ER co-repressor) was associated with decreased risk <strong>of</strong><br />

discontinuation <strong>of</strong> letrozole because <strong>of</strong> toxicity. Conclusions: Variants in<br />

genes involved in estrogen metabolism and signaling may be associated<br />

with toxicity <strong>of</strong> AI therapy.<br />

Statistical model Gene SNP AI HR (95% CI) p value<br />

Recessive CYP19A1 rs28566535 Either 8.2 (2.7-24.9) �0.0002<br />

Exe 8.1 (1.6-41) 0.012<br />

Let 7.5 (1.5-38.1) 0.016<br />

ESR1 rs9322335 Either 4.2 (2-8.6) �0.0001<br />

Exe 10.5 (2.9-38.7) �0.0004<br />

Let 3.3 (1.2-9.1) 0.018<br />

ESR1 rs9322336 Either 3.7 (1.8-7.4) �0.0004<br />

Exe 7.2 (2.7-19.5) 0.0001<br />

Let 3 (1-8.8) 0.050<br />

Additive NCOR1 rs1065822 Either 0.7 (0.5-0.9) 0.015<br />

Exe 1.1 (0.7-1.6) 0.77<br />

Let 0.3 (0.2-0.6) �0.0002<br />

527 Poster Discussion Session (Board #17), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Impact <strong>of</strong> adjuvant trastuzumab on outcomes <strong>of</strong> HER2-positive breast<br />

cancer patients treated with HER2-targeted therapy in the metastatic<br />

setting. Presenting Author: Rashmi Krishna Murthy, University <strong>of</strong> Texas<br />

M. D. Anderson Cancer Center, Houston, TX<br />

Background: Trastuzumab (T) was approved for the adjuvant treatment <strong>of</strong><br />

women with early-stage, HER-2 overexpressing (HER2�) breast cancer in<br />

2006. There are limited data outlining the outcomes <strong>of</strong> patients with<br />

HER2� breast cancer who receive adjuvant T-based therapy and then<br />

receive T and/or lapatinib in the metastatic setting. Methods: We identified<br />

540 patients with HER2� breast cancer treated with T or lapatinib as part<br />

<strong>of</strong> their first-line treatment for metastatic disease from 01/1997 to<br />

11/2011. HER-2 positivity was assessed by immunohistochemistry (score,<br />

3�) or fluorescence in situ hybridization (HER2/CEP17 ratio � 2). We<br />

excluded 17 patients from this analysis because they were either lost to<br />

follow-up or received less than 2 cycles <strong>of</strong> therapy at the institution.<br />

Statistical analyses were performed using the chi-square test to compare<br />

proportions between groups and the Cox proportional hazards regression<br />

analysis to compare survival times and estimate the corresponding hazard<br />

ratio with 95% confidence interval. Results: Of the 523 patients eligible for<br />

analysis, 76 patients had received T in the adjuvant setting and 447 had<br />

not. In the group who did not receive adjuvant T, 48% (213/447) <strong>of</strong><br />

patients achieved a complete or partial response (CR/PR), whereas only<br />

13% (14/76) achieved a CR/PR in the adjuvant T group (P�.0001). After<br />

adjustment for age, disease-free interval, post-menopausal status, stage at<br />

presentation, ER/PR status, and nuclear grade, the odds ratio was 0.27 (CI<br />

0.13 - 0.56, p � 0.0004). Overall survival from first evidence <strong>of</strong> metastasis<br />

was significantly longer in the group who did not receive adjuvant T (39<br />

months vs. 24 months, HR � 1.8, 95% CI 1.3-2.4). For OS, the adjusted<br />

hazard ratio was 1.5 (CI 1.04 - 2.1, p � 0.029). Age, DFI and stage were<br />

also significant predictors <strong>of</strong> OS. Conclusions: Patients with HER2�<br />

metastatic breast cancer who were T naive, had a higher response rate<br />

(CR/PR) to front line HER2 targeted therapy and a longer OS compared to<br />

patients with metastatic HER2� breast cancer who received T in the<br />

adjuvant setting. These findings highlight the importance <strong>of</strong> recognizing a<br />

pre-treated population and calls for further research in this area.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


14s Breast Cancer—HER2/ER<br />

528 Poster Discussion Session (Board #18), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Results from a phase Ib study <strong>of</strong> trastuzumab emtansine (T-DM1),<br />

paclitaxel (T), and pertuzumab (P) in patients with HER2-positive metastatic<br />

breast cancer (MBC) previously treated with trastuzumab. Presenting<br />

Author: Shanu Modi, Memorial Sloan-Kettering Cancer Center, New York,<br />

NY<br />

Background: The antibody–drug conjugate T-DM1 has shown single-agent<br />

activity in phase II studies in patients (pts) with HER2–positive MBC.<br />

Preclinical data suggest synergy for T-DM1 combined with taxanes and with<br />

P. Methods: TDM4652g is a phase Ib, open-label, dose-escalation study<br />

evaluating the safety and tolerability <strong>of</strong> T-DM1 (qw and q3w) � T (qw) � P<br />

(q3w) in pts with HER2-positive MBC previously treated with trastuzumab.<br />

A3�3 dose-escalation scheme is used for T-DM1 � T to determine the<br />

maximum tolerated dose (MTD); P is added at this MTD. Initial restrictive<br />

dose-limiting toxicity (DLT) criteria were modified to establish a more<br />

clinically relevant MTD. Results: We report interim results with T-DM1 (qw<br />

and q3w) � T(�P) using modified DLT criteria. 24 pts have been enrolled<br />

in these cohorts; median age was 53 yrs (range, 23–69)*; median number<br />

<strong>of</strong> prior systemic therapies in MBC was 7 (range, 2–15)*. See table below.<br />

Conclusions: Data support combining T-DM1 � T � P at the MTD for future<br />

clinical trials. The MTD for weekly T-DM1 � T is 2.4 mg/kg � 80 mg/m2 qw; MTD for weekly T-DM1 � T � P is 2.4 mg/kg � 80 mg/m2 qw � 840<br />

mg LD, 420 mg q3w. Updated results will be presented, including the MTD<br />

for T-DM1 q3w � Tqw� P q3w, outcomes from pts with prolonged<br />

follow-up, and duration <strong>of</strong> response from an extension trial.<br />

Dosing † DLTs MTD Gr 3/4 AEs* SAEs* Best response*<br />

T-DM1 (1.6–2.4 mg/kg qw) �<br />

T (65–80 mg/m 2 qw)<br />

4 cohorts<br />

n�12<br />

T-DM1 (2.4 mg/kg qw) �<br />

T (80 mg/m 2 qw) �<br />

P (840 mg LD,<br />

420 mg q3w)<br />

1 cohort<br />

n�3<br />

T-DM1 (2.4-3.6 mg/kg q3w) �<br />

T (65-80 mg/m 2 qw)<br />

3 cohorts<br />

n�9<br />

None 2.4 mg/kg qw �<br />

T80mg/m 2 qw<br />

None 2.4 mg/kg qw �<br />

T80mg/m 2 qw �<br />

P (840 mg LD,<br />

mg q3w)<br />

G3 peripheral neuropathy,<br />

G3 neuropathy (n�2),<br />

G3 fatigue (n�2),<br />

G3 neutropenia (n�3),<br />

G3 thrombocytopenia,<br />

G3 sepsis, G3 ulcer<br />

TBD TBD G3 hypokalemia,<br />

G4 febrile neutropenia,<br />

G3 abdominal pain<br />

G3 sepsis, G3 ulcer<br />

SD�6<br />

PR�3<br />

PR<br />

unconfirmed�1<br />

NA�2<br />

NR NR NA�2<br />

G4 febrile neutropenia SD�3<br />

PR<br />

unconfirmed�1<br />

NA�4<br />

*Based on data available from 22 pts as <strong>of</strong> data cut<strong>of</strong>f: Jan 24, 2012; NR�none reported; NA�not available. † Pts receiving P<br />

added to T-DM1 q3w � T qw to be enrolled.<br />

530 Poster Discussion Session (Board #20), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Phase IB/II study <strong>of</strong> the HSP90 inhibitor AUY922, in combination with<br />

trastuzumab, in patients with HER2� advanced breast cancer. Presenting<br />

Author: Anthony Kong, Churchill Hospital, Oxford University Hospitals<br />

NHS Trust and University <strong>of</strong> Oxford, Oxford, United Kingdom<br />

Background: AUY922 is a novel HSP90 inhibitor that causes degradation <strong>of</strong><br />

oncogenic proteins. A Phase I study in advanced solid tumors gave a<br />

recommended Phase II dose (RP2D) <strong>of</strong> 70 mg/m2 . In the Phase II part <strong>of</strong><br />

the study, single-agent AUY922 showed preliminary activity in HER2�<br />

metastatic breast cancer (mBC) (Schröder et al ASCO 2011). This Phase<br />

Ib/II study assessed safety and efficacy <strong>of</strong> AUY922 � trastuzumab (T) in<br />

pts with T-refractory HER2� advanced/mBC. Methods: Pts with HER2� BC<br />

(ECOG PS �1) who progressed on 1–2 prior anti-HER2 regimens with at<br />

least 1 T-containing regimen, received weekly AUY922 (55 or 70 mg/m2 ,<br />

IV) � T (2 mg/kg) in a 28-day cycle. The Phase Ib primary objective was to<br />

define a MTD/RP2D <strong>of</strong> AUY922 combined with T using an adaptive<br />

Bayesian logistic regression model. The Phase II primary objective is to<br />

evaluate preliminary antitumor activity at this dose (ORR). Secondary<br />

objectives included evaluation <strong>of</strong> PK, safety and preliminary efficacy (PFS,<br />

OS). Results: In the Phase Ib part (11 pts) there was 1 DLT (Grade [Gr] 3<br />

diarrhea), and the full standard single dose <strong>of</strong> AUY922 was recommended<br />

for use in combination with T. As <strong>of</strong> 11th January 2012, 31 HER2� BC pts<br />

(median age 51 years [range 29–71]; 84% invasive ductal carcinoma;<br />

52% estrogen receptor-positive; 81% had received �2 prior antineoplastic<br />

regimens) received AUY922 at 55 mg/m2 (n�5) or 70 mg/m2 (n�26) � T.<br />

The most common study drug-related AEs (all Gr) were diarrhea (25 [81%]<br />

pts), visual AEs (e.g. vision blurred, visual impairment, photopsia) (25<br />

[81%] pts), and nausea (11 [35%] pts). Gr 3/4 AEs were rare; most<br />

frequently diarrhea and visual impairment (both 6%). Treatment was<br />

ongoing for 13 (37%) pts at the cut<strong>of</strong>f date. Preliminary Phase II best<br />

overall response data (RECIST, investigator assessed) was available for 22<br />

pts at AUY922 70 mg/m2 . Any partial responses were reported in 5/22<br />

(23%) pts. Conclusions: The data suggests AUY922 � T is well tolerated.<br />

Common AEs were considered manageable, and eye disorders were<br />

reversible with treatment interruption, dose reduction or discontinuation.<br />

The combination <strong>of</strong> AUY922 � T is active in pts with HER2� mBC who<br />

progressed on T-based therapy, and the regimen warrants further investigation.<br />

529 Poster Discussion Session (Board #19), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Phase Ib study <strong>of</strong> open-label AMG 386 plus paclitaxel (P) and trastuzumab<br />

(T) or capecitabine (C) and lapatinib (L) in patients (pts) with HER2�<br />

locally recurrent or metastatic breast cancer (MBC). Presenting Author:<br />

Hans Wildiers, Universitair Ziekenhuis Gasthuisberg, Department <strong>of</strong> General<br />

Medical Oncology, Leuven, Belgium<br />

Background: AMG 386, an investigational peptibody, reduces tumor angiogenesis<br />

by blocking interaction <strong>of</strong> angiopoietins 1 and 2 with the Tie2<br />

receptor. This interim analysis evaluates the tolerability and efficacy <strong>of</strong><br />

AMG 386 � P/T or C/L in HER2� MBC. Methods: In part 1, pts in cohorts<br />

A1 and A3 (no prior 1st-line MBC T or L ) received AMG 386 IV QW plus P<br />

80 mg/m2 IV QW and T 8 mg/kg loading dose, then 6 mg/kg Q3W; pts in<br />

cohorts B1 and B3 (history <strong>of</strong> failed 1st-line MBC T treatment; no prior L or<br />

C) received AMG 386 IV QW plus C 1000 mg/m2 PO Q12 hrs, days 1-14<br />

Q21D and L 1250 mg PO QD. A1 and B1 received AMG 386 at 10 mg/kg;<br />

A3 and B3 received AMG 386 at 30 mg/kg. In part 2, cohorts were<br />

expanded to n � 20 if � 1<strong>of</strong>6or�2 <strong>of</strong> 9 pts had dose-limiting toxicities<br />

(DLTs). Primary endpoints were adverse events (AEs) and DLTs; secondary<br />

endpoints included efficacy and pharmacokinetics (PK). Interim results<br />

from A1, A3, and B1 will be presented. Results: 46 pts were enrolled at<br />

interim analysis; all received � 1 dose <strong>of</strong> study treatment (A1, A3, B1; n �<br />

20, 6, 20). The median follow-up for A1, A3, and B1 was 39.6, 22.7, and<br />

31.3 wks. Across cohorts, there was 1 DLT in A1. AEs � 50% were<br />

peripheral edema, diarrhea, fatigue and alopecia in A1 and A3 combined,<br />

and diarrhea, nausea, palmar-plantar erythrodysaesthesia syndrome (PPES),<br />

and peripheral edema in B1. AEs grade �3 occurring in � 2 pts were<br />

peripheral neuropathy, peripheral sensory neuropathy, dyspnea (n � 5, 4,<br />

3, respectively, in A1 and A3 combined); PPES, diarrhea, and neutropenia<br />

(n � 6, 5, 3, respectively, in B1). Objective response rates were 80% in A1,<br />

50% in A3, and 50% in B1. Median (95% CI) progression-free survival was<br />

14.5 mo (6.8-20.5) in A1 and 10.1 mo (3.7-14.7) in B1. Median duration<br />

<strong>of</strong> response (DOR) was 12.6 mo (4.3-20.2) in A1 and 8.5 mo (4.1-not<br />

evaluable) in B1. PFS and DOR were not yet evaluable in A3. No changes<br />

were apparent in any PK parameters in these combinations relative to<br />

within study or historical monotherapy PK data. Conclusions: In this<br />

ongoing phase 1b study <strong>of</strong> pts with HER2� MBC, interim results suggest<br />

that adding AMG 386 to P/T or C/L is tolerable and has antitumor activity.<br />

Updated data will be presented.<br />

531 Poster Discussion Session (Board #21), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

A randomized phase II study (VEG108838) <strong>of</strong> lapatanib plus pazopanib<br />

(L�P) versus lapatanib (L) in patients with ErbB2� inflammatory breast<br />

cancer (IBC). Presenting Author: Massimo Crist<strong>of</strong>anilli, Fox Chase Cancer<br />

Center, Philadelphia, PA<br />

Background: ErbB2 amplification is frequently reported in IBC and there is<br />

evidence <strong>of</strong> positive association between ErbB2 and VEGF expression. We<br />

evaluated the combination <strong>of</strong> anti ErbB2 and VEGF therapy in ErbB2� IBC.<br />

Methods: We conducted a multicenter, randomized clinical trial for patients<br />

(pts) with relapsed ErbB2� IBC. Cohort 1: Pts stratified (prior trastuzumab;<br />

cutaneous disease only vs systemic) and randomized 1:1 to receive<br />

L 1500 mg � placebo or L 1500 mg � P 800 mg, QD. Due to high<br />

incidence <strong>of</strong> Grade 3/4 diarrhea in pts treated with L 1500 mg� P 800 mg<br />

in another study, Cohort 1 was closed after 76 pts randomized. Cohort 2<br />

(87 pts ): Pts were stratified (prior trastuzumab) and randomized 5:5:2 to<br />

receive L 1500 mg � placebo or L 1000 mg � P 400 mg (double-blind) or<br />

P 800 mg (open-label), respectively, QD. Treatment continued until PD,<br />

unacceptable toxicity or death. Primary endpoint was ORR. Secondary<br />

endpoints included PFS, OS, and safety. Results: Cohort 1: 76 pts were<br />

randomized and treated: L, n�38; L�P, n�38. ORR was 29% for the L<br />

arm, and 45% for the L�P arm. Median PFS was 16.1 and 14.3 wks,<br />

respectively, for the L and L�P arms. The most frequent Grade �3 AEs<br />

were diarrhea (0% vs 18%) vomiting (0% vs 8%), ALT increased (0% vs<br />

8%), neutropenia (3% vs 13%), and bilirubin increased (0% vs 5%). Dose<br />

reductions due to AE were 3% and 21% and dose interruptions due to AE<br />

were 11% and 55% in the L and L�P arms, respectively. Cohort 2: 88 pts<br />

were randomized (87 treated): L, n�36; P, n�14; L�P, n�38. The ORR<br />

was 47%, 31%, and 58% for the L, P, and L�P arms, respectively. Median<br />

PFS was 16.0, 11.4, and 16.0 wks for the L, P, and L�P arms,<br />

respectively. The most frequent Grade �3 AEs were ALT increased (0%,<br />

0%, 21%), AST increased (0%, 0%, 18%), diarrhea (3%, 8%, 8%), and<br />

fatigue (3%, 8%, 8%). Dose reductions due to AE occurred in 0%, 0%, and<br />

13% <strong>of</strong> pts and dose interruptions due to AE occurred in 22%, 23%, and<br />

39% <strong>of</strong> pts in the L, P, and L�P arms, respectively. Conclusions: This<br />

prospective, randomized study confirmed the clinical activity <strong>of</strong> lapatinib<br />

single agent in metastatic ErbB2� IBC. Furthermore, we demonstrated<br />

increased toxicity associated with the combination without a clinically<br />

meaningful improvement in efficacy.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


532 Poster Discussion Session (Board #22), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Cardiac safety in a phase II study <strong>of</strong> trastuzumab emtansine (T-DM1)<br />

following anthracycline-based chemotherapy as adjuvant or neoadjuvant<br />

therapy for early-stage HER2-positive breast cancer. Presenting Author:<br />

Chau T. Dang, Memorial Sloan-Kettering Cancer Center, New York, NY<br />

Background: T-DM1 has demonstrated clinical activity as a single agent in<br />

patients (pts) with previously untreated MBC. In a previous phase II<br />

randomized trial <strong>of</strong> T-DM1 vs trastuzumab � docetaxel, T-DM1 had no<br />

clinically significant cardiac events, no cases <strong>of</strong> post-baseline left ventricular<br />

ejection fraction (LVEF) �40%, and fewer grade �3 adverse events<br />

(AEs; Hurvitz, ESMO 2011). This phase II study assessed the clinical<br />

safety and feasibility <strong>of</strong> T-DM1 following anthracycline-based chemotherapy<br />

in the adjuvant or neoadjuvant setting for early-stage HER2positive<br />

breast cancer. Methods: TDM4874g (NCT01196052) is a phase II<br />

single-arm, open-label study <strong>of</strong> T-DM1 (3.6 mg/kg q3w IV; up to 17 cycles)<br />

following completion <strong>of</strong> doxorubicin/cyclophosphamide (AC; q2w or q3w<br />

for 4 cycles), or 5-fluorouracil/epirubicin (100 mg/m2 )/cyclophosphamide<br />

(FEC; q3w for 3-4 cycles) chemotherapy in pts with early-stage HER2positive<br />

breast cancer. Pre-chemotherapy LVEF by MUGA/ECHO �55%<br />

was required for enrollment. Co-primary endpoints are safety and rate <strong>of</strong><br />

pre-specified cardiac events following initiation <strong>of</strong> T-DM1 treatment. An<br />

interim analysis was planned for the first 60 pts evaluable for cardiac safety<br />

(received �1 T-DM1 dose). Results: For pts in the interim analysis (20<br />

received AC, 40 FEC), the most common all-grade T-DM1-related AEs were<br />

nausea (n�20), asthenia (n�17), and headache (n�17); 8 pts had grade<br />

3/4 T-DM1-related AEs (including 3 with grade 3 increased aspartate<br />

aminotransferase [AST] and/or alanine aminotransferase [ALT]). No deaths<br />

occurred. Two pts had AEs leading to T-DM1 discontinuation (grade 3 AST<br />

and grade 2 ALT increase; grade 3 thrombocytopenia). No pre-specified<br />

cardiac events occurred; no pts delayed or discontinued T-DM1 due to<br />

cardiac AEs; there were no reports <strong>of</strong> grade �2 left ventricular systolic<br />

dysfunction, heart failure, or LVEF �50%. Results will be updated with<br />

data from all 153 enrolled pts. Conclusions: T-DM1 following anthracyclinebased<br />

chemotherapy was not associated with cardiac toxicity in pts with<br />

early-stage HER2-positive breast cancer; this study continues without<br />

modification.<br />

534 Poster Discussion Session (Board #24), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

A phase I trial <strong>of</strong> the IGF-1R antibody IMC-A12 in combination with<br />

temsirolimus in patients with metastatic breast cancer. Presenting Author:<br />

Gini F. Fleming, The University <strong>of</strong> Chicago Medical Center, Chicago, IL<br />

Background: mTOR plays a critical role in promoting tumor cell growth. In<br />

preclinical studies, the anti-tumor activity <strong>of</strong> mTOR inhibitors is attenuated<br />

by feedback up-regulation <strong>of</strong> AKT mediated by IGF-1R. We designed a<br />

phase I trial to determine the maximum-tolerated dose (MTD), doselimiting<br />

toxicities (DLT) and pharmacodynamic effects <strong>of</strong> the IGF-1R<br />

antibody IMC-A12 in combination with temsirolimus (tem) in patients (pts)<br />

with metastatic breast cancer (MBC) where mTOR is frequently activated.<br />

Methods: A3�3 phase I design was chosen. Tem and IMC-A12 were<br />

administered IV days (d) 1, 8, 15, and 22 <strong>of</strong> a 4-week cycle in pts with MBC<br />

refractory to standard therapies. Tumor response was evaluated by RECIST.<br />

Adverse events (AE) were reported using CTC v3.0. Serum IGF 1 and<br />

C-peptide levels on d2 (24h post infusion) and d8 prior to drug infusion<br />

were compared to baseline (BL) using paired t-test. Results: Of 26 pts<br />

enrolled, 4 did not complete cycle 1 due to progression (3) or co-morbid<br />

condition (1). MTD was determined from remaining 22 pts aged 34-72<br />

(median 48) years with ECOG PS 0 (55%) or 1 (45%). 86% had ER�<br />

cancer. Median number <strong>of</strong> regimens for MBC was 4. Two DLTs at the<br />

starting DL (DL 1) necessitated dose de-escalation <strong>of</strong> tem to 20mg (DL-1),<br />

then to 15 mg (DL-2) which was tolerable (Table). Subsequent dose<br />

escalation <strong>of</strong> IMC-A12 led to DLTs in 0 <strong>of</strong> 6 in DL-2A and 2 <strong>of</strong> 3 pts in<br />

DL-2B. The MTD was defined as DL-2A. Other AEs included gr 1/2 fatigue,<br />

neutropenia, anemia, and hyperglycemia. No CR or PR, but 4 SD lasting �<br />

4 months were observed. At DL-2, -2A and -2B, serum IGF 1 levels were<br />

significantly elevated on d2 (p �0.002) and d8 compared to BL (p<br />

�0.001), but C-peptide levels were not found to differ from BL. Conclusions:<br />

The MTD for the combination <strong>of</strong> IMC-A12 and tem in pts with MBC is lower<br />

than that observed for single agents alone. A phase II study is ongoing in<br />

MBC. The study is supported in part by ASCO CDA, Komen Craft to CXM,<br />

N01-CM62205 and N01-CM-2011-00071.<br />

DL Tem (mg) IMC-A12 (mg/kg) No. DLT<br />

1 25 3 3 2: gr 2 mucositis with trt delay<br />

-1 20 3 1 1: gr 3 neutropenia and leukopenia<br />

-2 15 3 6 1: gr 3 mucositis<br />

-2A 15 4 6 0<br />

-2B 15 5 3 1: gr 2 thrombocytopenia with trt delay<br />

1: gr 3 neutropenia with trt delay<br />

Breast Cancer—HER2/ER<br />

15s<br />

533^ Poster Discussion Session (Board #23), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Cardiac tolerability <strong>of</strong> pertuzumab plus trastuzumab plus docetaxel in<br />

patients with HER2-positive metastatic breast cancer in the CLEOPATRA<br />

study. Presenting Author: Michael Ewer, University <strong>of</strong> Texas M. D.<br />

Anderson Cancer Center, Houston, TX<br />

Background: Addition <strong>of</strong> trastuzumab (T) to chemotherapy has transformed<br />

outcomes in pts with HER2-positive breast cancer. In pts who had received<br />

anthracyclines (A), T has been associated with cardiac dysfunction.<br />

Monitoring cardiac tolerability <strong>of</strong> anticancer drugs is important, including<br />

that <strong>of</strong> a new regimen combining T with a second anti-HER2 antibody that<br />

inhibits HER2 dimerization, pertuzumab (P). In CLEOPATRA, efficacy and<br />

safety <strong>of</strong> P�T�docetaxel (D) were studied in HER2-positive 1st-line MBC<br />

(Baselga 2012). Methods: CLEOPATRA, a randomized, double-blind,<br />

placebo-controlled, ph III trial, enrolled 808 pts; 804 were included in the<br />

safety population. A baseline (BL) LVEF �50%, no history <strong>of</strong> congestive<br />

heart failure, and no LVEF decline to �50% during/after prior T were<br />

required. Treatment was administered q3w until disease progression or<br />

unmanageable toxicity (P/placebo [Pla]: 840 mg, followed by 420 mg; T: 8<br />

mg/kg, followed by 6 mg/kg; D [�6 cycles recommended]: 75 mg/m2 ,<br />

escalating to 100 mg/m2 if tolerated). LVEF was assessed by ECHO or<br />

MUGA at BL, every 9 wks during treatment, at discontinuation, and up to 3<br />

yrs thereafter. Adverse events (AE) were monitored continuously and<br />

graded according to NCI-CTCAE v3.0. Results: The incidence <strong>of</strong> any cardiac<br />

disorder (grade �1) as assessed by the investigators was similar with<br />

Pla�T�D (16.4%) and P�T�D (14.5%). LVSD (grade �1) was the most<br />

frequent cardiac AE and more common with Pla�T�D. At the time <strong>of</strong> data<br />

cut<strong>of</strong>f for this analysis, 8/11 symptomatic LVSD events had resolved; none<br />

fatal. All pts who developed symptomatic LVSD had �1 potential cardiac<br />

risk factor (prior A, prior T, radiation, smoking, diabetes, hypertension,<br />

other). Compared to the overall pt population, only prior A and radiation<br />

were higher in pts who developed symptomatic LVSD. Conclusions: CLEO-<br />

PATRA provides evidence that P�T�D does not increase overall cardiac<br />

disorders, specifically symptomatic LVSD, compared to Pla�T�D.<br />

n (%) Pla�T�D(n�397) P�T�D(n�407)<br />

LVSD (grade >1)* 33 (8.3) 18 (4.4)<br />

Symptomatic LVSD (grade >3)* 7 (1.8) 4 (1.0)<br />

LVEF decline to 10% points from BL †<br />

25/379 (6.6) 15/393 (3.8)<br />

* NCI-CTCAE v3.0. † Pts with post-BL LVEF assessment.<br />

535 Poster Discussion Session (Board #25), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Activity <strong>of</strong> cabozantinib (XL184) in metastatic breast cancer (MBC):<br />

Results from a phase II randomized discontinuation trial (RDT). Presenting<br />

Author: Eric P. Winer, Dana-Farber Cancer Institute, Boston, MA<br />

Background: Dysregulation <strong>of</strong> MET and VEGF signaling has been implicated<br />

in breast cancer development and progression, including tumor invasion<br />

and dissemination. Cabozantinib (cabo) is an oral, potent inhibitor <strong>of</strong> MET<br />

and VEGFR2. A RDT evaluated activity and safety in 9 tumor types,<br />

including MBC. Methods: Eligible patients (pts) were required to have<br />

progressive measurable disease per RECIST. Pts received cabo at 100 mg<br />

qd over a 12 wk Lead-in stage. Tumor response (mRECIST) was assessed<br />

q6 wks. Treatment � wk 12 was based on response: pts with PR continued<br />

open-label cabo, pts with SD were randomized to cabo vs placebo, and pts<br />

with PD discontinued. Primary endpoint in the randomized discontinuation<br />

phase was progression-free survival (PFS). Results: Enrollment to this<br />

cohort is complete (n � 45); all pts are unblinded. Baseline characteristics:<br />

median age 56; invasive ductal 86%, invasive lobular 7%; ER� 93%,<br />

HER2� 18%, triple- 5%; visceral disease 91%; bone metastases 73%;<br />

median prior lines <strong>of</strong> therapy 3 (range 1-8), including 71% with prior<br />

anthracyclines. Median follow-up was 2.9 mos (range 0.1 -16). 21 pts<br />

(47%) completed Lead-in stage with only 9 randomized to continue cabo (n<br />

� 5) or placebo (n � 4). Median PFS from Study Day 1 was 4.1 mos. At wk<br />

12, objective response rate was 14% and disease control rate 48%. Tumor<br />

regression was observed in 25/39 pts (64%) with �1 post-baseline tumor<br />

assessment. 4/10 pts evaluable by bone scan had partial resolution <strong>of</strong> bone<br />

lesions. Of 12 pts receiving narcotics for bone pain, 5 pts reported<br />

improved pain and 2 pts had decreased narcotics use, per investigator.<br />

4/14 evaluable pts (29%) with bone metastases experienced �50%<br />

decline in serum NTx. Most common Grade 3/4 AEs were palmar-plantar<br />

erythrodyesthesia (13%) and fatigue (11%). One related Grade 5 AE <strong>of</strong><br />

respiratory compromise was reported during the Lead-in stage. Conclusions:<br />

Cabo demonstrated a 14% rate <strong>of</strong> objective tumor regression in heavily<br />

pretreated MBC pts. Observed effects on bone scan and pain are consistent<br />

with those seen in other malignancies. The safety pr<strong>of</strong>ile <strong>of</strong> cabo was<br />

comparable to that seen with other VEGFR TKIs.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


16s Breast Cancer—HER2/ER<br />

536 General Poster Session (Board #1A), Sat, 8:00 AM-12:00 PM<br />

Statin use and the development <strong>of</strong> bone metastasis in breast cancer<br />

patients. Presenting Author: Gentry T. King, Albert Einstein Medical<br />

Center, Philadelphia, PA<br />

Background: The Mevalonic Acid Pathway has been implicated in the<br />

promotion <strong>of</strong> a microenvironment suitable for establishment <strong>of</strong> bony<br />

metastasis from breast cancer. The statins, which act on this pathway, have<br />

been shown to have in-vitro anti-neoplastic activity against breast cancer.<br />

This study was designed to evaluate the association <strong>of</strong> statin use and<br />

development <strong>of</strong> bony metastasis in breast cancer patients. Methods:<br />

Medical records <strong>of</strong> patients treated for stage II-III breast cancer from 1999<br />

to 2010 were retrospectively reviewed. Statin use was defined as medication<br />

use for at least 3 months in patients with no evidence <strong>of</strong> disease after<br />

initial diagnosis and treatment. The primary outcome was development <strong>of</strong><br />

metastasis to bone. Secondary outcomes were overall survival, disease free<br />

survival and other sites <strong>of</strong> distant metastasis. Results: A total <strong>of</strong> 841<br />

patients were included in the study <strong>of</strong> which 223 used statins. Both<br />

unadjusted and multivariate analysis adjusted for age, race, grade, stage ,<br />

BRCA status, showed that patients on statins had a significantly lower<br />

incidence <strong>of</strong> metastasis to bone (OR 0.49, 95% CI 0.25-0.96, p�0.04).<br />

Adjusted analysis for other sites showed a trend towards decreased<br />

incidence <strong>of</strong> metastasis for statin users, but was not statistically significant<br />

(95% CI 0.39-1.08, p�0.10). Overall survival was increased in statin<br />

users with mean survival <strong>of</strong> 66.45 �/- 2.48 months versus non-users<br />

58.78 �/ - 1.41 months (p�0.05). Statin users had significantly longer<br />

disease free survival with a mean <strong>of</strong> 63.65 �/- 2.49 months versus 53.96<br />

�/- 1.42 months in non statin users (p�0.00). Conclusions: The use <strong>of</strong><br />

statin drugs in patients with breast cancer was significantly associated with<br />

decreased incidence <strong>of</strong> metastasis to bone, but not to other distant sites.<br />

The role <strong>of</strong> statins in chemoprevention <strong>of</strong> bone metastasis should be further<br />

explored.<br />

538 General Poster Session (Board #1C), Sat, 8:00 AM-12:00 PM<br />

Randomized trials <strong>of</strong> adjuvant tamoxifen versus tamoxifen and octreotide<br />

LAR in early-stage breast cancer: NCIC CTG MA.14 and NSABP B-29.<br />

Presenting Author: Judy-Anne W. Chapman, NCIC <strong>Clinical</strong> Trials Group,<br />

Queen’s University, Kingston, ON, Canada<br />

Background: MA.14 and B-29 examined whether the addition <strong>of</strong> Octreotide<br />

(SMS 201-995 pa LAR; OCT) to 5 years adjuvant tamoxifen (TAM)<br />

prolonged disease-free survival (DFS). Excessive gallbladder (GB) toxicity<br />

in B-29 led to DMC recommended stopping <strong>of</strong> OCT and MA.14 shortening<br />

<strong>of</strong> OCT to 2 years. Data for the 2 trials were pooled. Methods: Median<br />

follow-up <strong>of</strong> the 667 MA.14 patients was 9.8 years, and <strong>of</strong> the 893 B-29<br />

patients, 6.8 years for ITT analysis. The primary endpoint was DFS, defined<br />

as time from randomization to time <strong>of</strong> (local, regional or distant) breast<br />

cancer recurrence; any second primary cancer other than squamous or<br />

basal cell carcinoma <strong>of</strong> the skin, carcinoma in situ <strong>of</strong> the cervix, or lobular<br />

carcinoma in situ <strong>of</strong> the breast; or death. The primary test statistic was a<br />

pooled stratified log-rank test, with stratification within each trial by<br />

applicant stratification factors. Multivariate assessment was with Cox<br />

regression. Results: MA.14 patients were all postmenopausal with 97%<br />

�50 years <strong>of</strong> age while 62% <strong>of</strong> B-29 women were �50. 53% <strong>of</strong> MA.14<br />

patients were lymph node negative (LN-), while all <strong>of</strong> B-29 patients were<br />

LN-. 33% <strong>of</strong> MA.14 patients received adjuvant chemotherapy, 2% concurrently,<br />

while 53% <strong>of</strong> B-29 received concurrent chemotherapy. 90% <strong>of</strong><br />

MA.14 patients were hormone receptor positive while 100% <strong>of</strong> B-29 were<br />

positive. MA.14 patients experienced a 5 year DFS rate <strong>of</strong> 80% on TAM and<br />

76% on TAM�OCT, while B-29 patients had a 5 year rate <strong>of</strong> 88% for both<br />

arms. The pooled 5 year rates were 85% and 83%. Pooled univariate<br />

stratified Cox HR <strong>of</strong> TAM�OCT to TAM was 0.99 (95% CI 0.81-1.20;<br />

p-value� 0.69). The HR for MA.14 was 0.93 (0.72-1.19; p� 0.50) and for<br />

B-29 was 1.09 (0.80-1.50; p�0.59). Multivariate pooled HR <strong>of</strong> TAM�OCT<br />

to TAM was 0.98 (0.81-1.20; p�0.84). Patients who were older<br />

(p�0.0006), and had higher T stage (p�0.0001) with LN� (p�0.0008)<br />

had shorter DFS. Conclusions: OCT did not significantly improve DFS for<br />

women who were a broad spectrum <strong>of</strong> patients: pre- or post-menopausal,<br />

LN- or LN�, and received sequential, concurrent or no adjuvant chemotherapy.<br />

Neither study had the intended duration <strong>of</strong> OCT due to GB toxicity;<br />

thus, the results do not negate targeting the insulin - IGF-I receptor family.<br />

537 General Poster Session (Board #1B), Sat, 8:00 AM-12:00 PM<br />

ACE inhibitors and angiotensin receptor blockers use and breast cancer<br />

outcomes in patients treated with neoadjuvant systemic chemotherapy.<br />

Presenting Author: Young Kwang Chae, University <strong>of</strong> Texas M. D. Anderson<br />

Cancer Center, Houston, TX<br />

Background: There is a growing body <strong>of</strong> evidence to suggest that ACE<br />

inhibitors (ACEIs) and angiotensin receptor blockers (ARBs) might have<br />

anti-tumor properties. We investigated whether the use <strong>of</strong> ACEI/ARBs<br />

during chemotherapy affects the clinical outcomes <strong>of</strong> breast cancer<br />

patients receiving taxane and anthracycline-based neoadjuvant chemotherapy.<br />

Methods: We included in this analysis 1449 patients with<br />

diagnosis <strong>of</strong> invasive primary breast cancer diagnosed at the MD Anderson<br />

Cancer Center between 1995 and 2007 who underwent neoadjuvant<br />

chemotherapy. Of them, 160 (11%) patients were identified by review <strong>of</strong><br />

their medical record, as ACEI/ARBs users. We compared pathologic<br />

complete response (pCR) rates, relapse-free survival (RFS), diseasespecific<br />

survival (DSS) and overall survival (OS) between ACEI/ARB users<br />

and non-users. pCR was defined as no evidence <strong>of</strong> invasive carcinoma in<br />

the breast and axillary lymph nodes at the time <strong>of</strong> surgery. Descriptive<br />

statistics and Cox proportional hazards model were used in the analyses.<br />

Results: There was no difference in the pCR rates between ACEI/ARB users<br />

(16%) versus non-users (18.1%) (p�0.50). After adjustment for important<br />

demographic and clinical characteristics, no significant differences between<br />

ACEI/ARB users and nonusers were observed in RFS (HR�0.81;<br />

95% CI�0.54-1.21; P�0.30), DSS (HR�0.83; 95% CI�0.52-1.31;<br />

P�0.42), or OS (HR�0.91; 95% CI �0.61-1.37; P�0.66). In a subgroup<br />

analysis among ARB users, the 5-year RFS was 82% vs 71% in ACEI/ARB<br />

non-users (P�0.03). In the multivariable analysis, ARB use was also<br />

associated with a decreased risk <strong>of</strong> recurrence (HR�0.35; 95% CI�0.14-<br />

0.86; P�0.02). However, no statistically significant differences in DSS or<br />

OS were seen. Conclusions: No differences in pCR and survival outcomes<br />

were seen between ACEI/ARB users and non-users among breast cancer<br />

patients receiving neoadjuvant chemotherapy. ARB use may be associated<br />

with improved RFS. Further research is needed to validate this finding.<br />

539 General Poster Session (Board #1D), Sat, 8:00 AM-12:00 PM<br />

BOLERO-2: Health-related quality-<strong>of</strong>-life in metastatic breast cancer<br />

patients treated with everolimus and exemestane versus exemestane.<br />

Presenting Author: J. Thaddeus Beck, Highlands Oncology Group, Fayetteville,<br />

AR<br />

Background: BOLERO-2, a phase III study, randomized 724 patients with<br />

hormone-receptor–positive metastatic breast cancer, who had recurrence<br />

or progression on/after prior nonsteroidal aromatase inhibitor therapy, to<br />

everolimus (EVE) plus exemestane (EXE) or EXE and placebo. A preplanned<br />

12-month median time interim analysis demonstrated that EVE�EXE<br />

significantly improved progression-free survival (PFS) vs EXE�placebo but<br />

that EVE�EXE resulted in a higher rate <strong>of</strong> grade 3/4 toxicity. Per-protocol<br />

patient reported health-related quality-<strong>of</strong>-life (HRQoL) data are limited;<br />

here we report on additional post-hoc analyses <strong>of</strong> these outcomes. Methods:<br />

Using the EORTC–QLQ-C30 questionnaire, HRQoL was assessed at baseline<br />

and every 6 weeks thereafter until progression. The QLQ-C30 consists<br />

<strong>of</strong> 30 items combined into 15 subscales, including a Global Health Status<br />

(GHS) where higher scores (range 0 to 100) indicate better HRQoL. This<br />

analysis included a protocol-specified time to definitive deterioration (TTD)<br />

analysis at a 5% decrease in QoL relative to baseline, with no subsequent<br />

increase above this threshold. We report additional sensitivity analyses<br />

using 10-point minimally important difference (MID) decreases in QLQ-<br />

C30 score relative to baseline. Treatment arms were compared using a<br />

stratified logrank test and a Cox proportional hazards model adjusted for<br />

trial stratum (visceral metastases and previous hormone sensitivity), age,<br />

sex, race, baseline score and ECOG performance status, prognostic factors,<br />

and treatment history. Results: Baseline QLQ-C30 GHS scores were not<br />

statistically significantly different across treatment groups (64.7 vs 65.3;<br />

difference -0.7 [95% CI; -4.3, 3.0]). The median TTD in HRQoL was 7.0<br />

months (95% CI; 5.6, 8.3) for EVE�EXE vs 5.6 (95% CI; 4.2, 7.0) for EXE<br />

(P � .0792). Adjusted HR (0.80) approached significance (95% CI; 0.63,<br />

1.02). At the 10-point MID, median TTD for EVE�EXE was 9.7 months<br />

(95% CI; 8.3, 11.2) vs 8.4 months (95% CI; 6.3, 12.5) for EXE. Adjusted<br />

HR was 0.90 (95% CI; 0.69, 1.18). Conclusions: These additional analyses<br />

demonstrate that in addition to significantly improving PFS, EVE�EXE<br />

does not compromise HRQoL.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


540 General Poster Session (Board #1E), Sat, 8:00 AM-12:00 PM<br />

BOLERO-2: Everolimus with exemestane versus exemestane alone in Asian<br />

patients with HER2-negative, hormone receptor-positive breast cancer.<br />

Presenting Author: Shinzaburo Noguchi, Osaka University, Department <strong>of</strong><br />

Breast and Endocrine Surgery, Osaka, Japan<br />

Background: Estrogen-receptor–positive (ER�) breast tumors can become<br />

refractory to aromatase inhibitor (AI) therapy. The mTOR pathway plays a<br />

critical role in hormone-resistant advanced breast cancer (ABC). Accordingly,<br />

the addition <strong>of</strong> everolimus (EVE) to exemestane (EXE) was evaluated<br />

in an international, phase III study (BOLERO-2) in patients with ER� aBC<br />

refractory to letrozole or anastrozole. This report presents updated analyses<br />

from the population <strong>of</strong> Asian patients enrolled in this study. Methods:<br />

Eligible patients were randomized (2:1) to EXE (25 mg/day) with EVE (10<br />

mg/day) or with matching placebo. The primary endpoint was progressionfree<br />

survival (PFS). Secondary endpoints included overall survival, response<br />

rate, quality <strong>of</strong> life, and safety. Results: EVE � EXE significantly<br />

improved PFS versus EXE alone (HR � 0.44; 95% CI, 0.36-0.53; P �<br />

.0001) in the overall study population at median follow-up <strong>of</strong> 12.5 months.<br />

Of 143 patients <strong>of</strong> Asian origin (n � 106; 74% Japanese) enrolled in this<br />

study, 98 received EVE � EXE and 45 received EXE � placebo. At the time<br />

<strong>of</strong> database lock, 55 Asian patients (56%) in the combination arm had<br />

experienced disease progression compared with 34 patients (76%) in the<br />

EVE � placebo arm. Combination therapy reduced the risk <strong>of</strong> disease<br />

progression versus EXE alone by 44% among Asian patients (HR � 0.56;<br />

95% CI, 0.37-0.87; P � .05). Commonly reported adverse events in the<br />

combination arm were consistent with previous clinical studies <strong>of</strong> mTOR<br />

inhibitors and included stomatitis (80%), rash (49%), dysgeusia (31%).<br />

Adverse events that occurred more frequently in the Asian subset versus<br />

Caucasians included stomatitis (80% vs 54%) and rash (49% vs 37%),<br />

whereas dyspnea (8% vs 24%) and asthenia (1% vs 17%) occurred less<br />

frequently in the Asian subset versus Caucasians. Conclusions: The addition<br />

<strong>of</strong> EVE to EXE significantly prolonged PFS in Asian patients versus EXE<br />

alone. Adverse events were higher in the combination arm but generally<br />

manageable. Combining EVE with an AI is a safe and effective treatment<br />

option for Asian women with non-steroidal AI resistant/refractory ER� ABC.<br />

542 General Poster Session (Board #1G), Sat, 8:00 AM-12:00 PM<br />

Impact <strong>of</strong> the EndoPredict-clin score on risk stratification in ER-positive,<br />

HER2-negative breast cancer after considering clinical guidelines. Presenting<br />

Author: Martin Filipits, Institute <strong>of</strong> Cancer Research, Comprehensive<br />

Cancer Center, Medical University <strong>of</strong> Vienna, Vienna, Austria<br />

Background: Many ER-positive, HER2-negative breast cancer patients are<br />

treated by adjuvant chemotherapy according to current clinical guidelines.<br />

We retrospectively assessed whether the combined gene expression/<br />

clinicopathological EndoPredict-clin (EPclin) score improved the accuracy<br />

<strong>of</strong> risk classification in addition to considering clinical guidelines. Methods:<br />

Three clinical breast cancer guidelines (National Comprehensive Cancer<br />

Center Network (NCCN), German S3 and St. Gallen 2011), and the EPclin<br />

score - assessed by quantitative RT-PCR in formalin-fixed paraffinembedded<br />

tissue - were used to assign risk groups in 1,702 ER-positive,<br />

HER2-negative breast cancer patients from two randomized phase III trials<br />

(Austrian Breast and Colorectal Cancer Study Group 6 and 8) treated with<br />

endocrine therapy only. Results: Although all analyzed clinical guidelines<br />

identified a low-risk group with improved metastasis-free survival, the<br />

overwhelming majority <strong>of</strong> all patients (81-94%) were classified as intermediate<br />

/ high risk. In contrast to that, the EPclin classified only 37% <strong>of</strong> all<br />

patients as high risk and that stratification resulted in the best separation<br />

between low and high risk groups (p � 0.001, HR � 5.11 (3.48-7.51).<br />

Consequently, the majority <strong>of</strong> all patients deemed intermediate / high risk<br />

by the clinical guidelines was re-classified as low risk by the EPclin score.<br />

Kaplan Meier analyses demonstrated that the re-classified subgroups (47<br />

to 57% <strong>of</strong> all patients) had an excellent 10-year metastasis-free survival <strong>of</strong><br />

95% comparable to the clinical assigned low-risk groups although encompassing<br />

a higher proportion <strong>of</strong> the trial patients. Conclusions: The EPclin<br />

score predicted distant recurrence more accurately than all three clinical<br />

guidelines and is especially useful to reclassify patients considered as<br />

intermediate / high risk by the guidelines. The data suggests that the EPclin<br />

score provides clinically useful prognostic information beyond common<br />

clinical guidelines and can be used to accurately identify the clinically<br />

relevant group <strong>of</strong> patients who are adequately and sufficiently treated with<br />

adjuvant endocrine therapy alone.<br />

Breast Cancer—HER2/ER<br />

17s<br />

541 General Poster Session (Board #1F), Sat, 8:00 AM-12:00 PM<br />

Response pattern in 844 patients with infiltrating lobular carcinoma (ILC)<br />

<strong>of</strong> the breast after neoadjuvant chemotherapy. Presenting Author: Cristina<br />

Pirvulescu, Helios Klinikum Berlin-Buch, Berlin, Germany<br />

Background: Patients (pts) with infiltrating lobular carcinoma (ILC) have a<br />

different response to neoadjuvant chemotherapy compared to patients with<br />

non-lobular carcinoma (NLC). The aim <strong>of</strong> our analysis was to characterize<br />

correlates <strong>of</strong> the outcome impact <strong>of</strong> neoadjuvant chemotherapy in ILC and<br />

to compare those with NLC. Methods: Between 1998 and 2006; 6377<br />

breast cancer patients were enrolled to 7 randomized neoadjuvant chemotherapy<br />

trials in Germany. For 6205 (98.3%) pts the histological type was<br />

available. 844 (13.6%) were classified as ILC, 5361 (86.4%) were<br />

classified as NLC. Endpoints <strong>of</strong> the analyses were: pathological complete<br />

response (pCR) defined as no invasive and no in-situ residuals in the breast<br />

and the lymph nodes, type <strong>of</strong> surgery, disease-free (DFS), local recurrence<br />

(LRFS) and overall survival (OS). Results: ILC was associated with older age,<br />

larger tumor size, negative nodes, lower grading, and hormone receptor<br />

(HR) positivity (all p�0.0001). The majority <strong>of</strong> pts with ILC were HR<br />

positive (717 pts, 86.9%) and had G1-G2 tumors (603 pts, 78.6%).<br />

Patients with ILC had a significant lower pCR rate than pts with NLC (pCR<br />

6.04% vs. 16.4%, p�0.001). In the ILC group, pts with HR positive,<br />

triple-negative and HER2-positive tumors had a pCR rate <strong>of</strong> 5%, 18.3%<br />

and 9.2%, respectively. One <strong>of</strong> 35 pts with G1 and HR positive ILC had a<br />

pCR. Breast conservation therapy (BCT) in pts with pCR was 72.5% in the<br />

ILC group and 85.5% in the NLC group (p�0.0001). Predictors for pCR in<br />

the multivariate analysis were for ILC: patient age, grading and HR status.<br />

ILC pts had a significantly longer LRFS compared to NLC pts (with or<br />

without pCR after neoadjuvant chemotherapy, mean 112 months 95%CI<br />

[109.7-115.7] vs. 106 months 95%CI [104.4-107.4], p�0.002).<br />

Conclusions: In patients with ILC age, grading and HR status were<br />

predictive for pCR. Pts with ILC have a more favourable outcome after<br />

neoadjuvant chemotherapy despite a significantly lower pCR rate compared<br />

to pts with NLC.<br />

543 General Poster Session (Board #2A), Sat, 8:00 AM-12:00 PM<br />

Interpathologists discrepancies in Ki67 assessment in the PACS01 trial:<br />

An independent prognosis factor. Presenting Author: Frederique Madeleine<br />

Penault-Llorca, Centre Jean Perrin, Clermont-Ferrand, France<br />

Background: Several reports suggest an inter-observer variability in Ki67<br />

assessment. Nevertheless, there is no large study that evaluates the rate <strong>of</strong><br />

discrepancies, together with their impact. Methods: Ki67 expression was<br />

assessed on 663 samples from patients with ER-positive breast cancers<br />

included in the PACS01 trial (Roche, J Clin Oncol, 2006). Ki67 staining<br />

was done using MiB1 antibody (Dako, Copenhagen, Denmark, Dilution<br />

1:250). Prognostic and predictive values have been reported previously<br />

(Penault-Llorca, J Clin Oncol, 2009). A second central review was done by<br />

a senior breast pathologist from a French Cancer center. A discrepancy was<br />

defined as either a false positive or false negative result. Cut-<strong>of</strong>f for<br />

positivity was defined at 15% according to data from Cheang et al (JNCI,<br />

2009). Results: The rate <strong>of</strong> discrepancy was correlated with the percentage<br />

<strong>of</strong> stained tumor cells. A 10% discrepancy rate between the 2 pathologists<br />

was observed when the first pathologist reported �10% tumor cells<br />

stained. Same low rate <strong>of</strong> discrepancy (10%) was observed if more than<br />

30% cancer cells were stained according to the first assessment. At the<br />

opposite, discrepancy rates were 47%, 45%, 22%, 34% when the first<br />

pathologist reported 10-15%, 15-20%, 20-25% and 25-30% tumor cells<br />

stained. Overall, 36% <strong>of</strong> the patients presented a grade II tumor together<br />

with Ki67 �10% or �30%. We then evaluated the impact <strong>of</strong> discrepancy<br />

in terms <strong>of</strong> prognosis. Patients presenting a concordant result between the<br />

two pathologists showed a better outcome as compared to patients<br />

presenting a discrepancy, independently to the percentage <strong>of</strong> tumor<br />

stained (p�0.05, patients with concordant Ki67 ranged between 10-30%<br />

versus those with one reader �10% and the other one �10%-�30%).<br />

Conclusions: Discrepancy rates between pathologists are acceptable when<br />

Ki67 is either �10% or �30%. A Ki67 ranged between 10 and 30% could<br />

define a grey zone in which Ki67 should be reported with caution or be<br />

double checked by another pathologist. Survival analysis suggested that<br />

inter-observer discrepancies could act a prognostic factor. Such finding<br />

could reflect underlying intra-tumor heterogeneity.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


18s Breast Cancer—HER2/ER<br />

545 General Poster Session (Board #2C), Sat, 8:00 AM-12:00 PM<br />

A dendritic metagene that predicts prognosis and endocrine resistance in<br />

breast cancer. Presenting Author: Bianchini Giampaolo, Fondazione San<br />

Raffaele del Monte Tabor, Milan, Italy<br />

Background: High expression <strong>of</strong> dendritic-associated genes in estrogen<br />

receptor-positive (ER�), highly proliferative breast cancer is associated<br />

with good prognosis in untreated patients (pts) and poor response to<br />

neoadjuvant endocrine therapy (Dunbier SABCS 2010). We examined the<br />

prognostic and predictive value <strong>of</strong> a dendritic metagene (DM) signature in<br />

subgroups defined by proliferation and estrogen-related genes (Bianchini<br />

SABCS 2011). Methods: We evaluated Affymetrix HGU133A-based gene<br />

expression pr<strong>of</strong>iles from ER� untreated (n�511) and adjuvant tamoxifen<br />

(TAM)-treated pts (n�606). Three previously defined scores were assessed:<br />

MKS for proliferation (Bianchini, Cancer Res 2010); estrogenrelated<br />

score (ERS) adopted from Oncotype DX; and DM (Bianchini JCO<br />

2010). Median cut-<strong>of</strong>f points were used. Outcome was assessed according<br />

to distant relapse. Results: DM was significantly predictive for early (within<br />

5 years) relapse. In TAM-treated pts with low-MKS tumors, DM was not<br />

significantly prognostic. The table shows hazard ratios (HRs) for low vs.<br />

high-DM by ERS in untreated and TAM-treated pts with high-MKS. In<br />

low-DM, the 5-yrs distant-event free survival was 92.3% and 60.6% (HR<br />

6.58 (2.55-17.0); p�0.001) and in high-DM it was 80.7% and 79.8%<br />

(HR 1.06 (0.48-2.32); p�0.88), respectively in high and low-ERS<br />

respectively. The interaction between DM and ERS was significant only in<br />

TAM-treated tumors (p�0.003). In high-MKS/low-ERS tumors, DM retained<br />

significance over the 10-year period in multivariate analysis adjusting<br />

for clinical variables (HR� 2.27 (1.13-4.58); p�0.02). Conclusions:<br />

For high-MKS, untreated and TAM-treated tumors with low-ESR, high-DM<br />

is associated with a low risk <strong>of</strong> relapse, and for untreated tumors with<br />

high-ERS. These data suggest that high infiltration <strong>of</strong> tumor by dendritic<br />

cells may be a novel mechanism <strong>of</strong> endocrine resistance. DM could refine<br />

risk in poor prognosis ER� tumors and rationalize immuno-modulating<br />

strategies.<br />

Untreated/high MKS TAM-treated/high MKS<br />

>High-ERS No.<strong>of</strong> pts/events HR (CI 95%) p No.<strong>of</strong> pts/events HR (CI 95%) p<br />

Low<br />

(vs high-DM)<br />

116/21 2.9 (1.1-7.9) 0.039 127/15 0.4 (0.1-1.1) 0.067<br />

Low-ERS Low<br />

(vs high-DM)<br />

169/64 3.0 (1.8-5.0) �0.0001 175/46 2.3 (1.3-4.2) 0.007<br />

547 General Poster Session (Board #2E), Sat, 8:00 AM-12:00 PM<br />

Adjuvant capecitabine for invasive lobular/mixed early breast cancer (EBC):<br />

USON 01062 exploratory analyses. Presenting Author: Joyce<br />

O’Shaughnessy, Baylor Sammons Cancer Center, Texas Oncology, US<br />

Oncology, Dallas, TX<br />

Background: Randomized phase III USON 01062 trial determining if<br />

patients with EBC would benefit from addition <strong>of</strong> capecitabine to docetaxel<br />

after AC (AC-T vs AC-XT). AC-T: docetaxel 100mg/M2 IV; AC-XT: docetaxel<br />

75mg/M2 IV with capecitabine 825mg/M2 PO BID 14/7 days every 21days<br />

for 4 cycles. The primary endpoint, improvement in disease-free survival<br />

(DFS) at 5 years, was not met (HR�0.84, p�0.12) likely due to<br />

lower-than-expected event rate. The secondary endpoint, overall survival<br />

(OS), was improved with capecitabine (HR 0.68, p�0.01) (O’Shaughnessy,<br />

J. ASCO, 2011). Methods: Molecular analyses demonstrate that pleomorphic<br />

lobular (mixed lobular/ductal) carcinomas evolve from the same<br />

precursor and/or through the same genetic pathway as classical lobular<br />

cancers (Reis-Filho, J., J Path, 2005). We conducted exploratory analyses<br />

to evaluate the addition <strong>of</strong> adjuvant capecitabine in ductal vs lobular or<br />

lobular/ductal (mixed) EBC within USON 01062. Histology was classified<br />

according to local pathology assessment on patients’ primary cancers.<br />

Results: In ductal patients (n�2195), there was no difference in DFS<br />

(HR�0.92, p�0.48) and OS (HR�0.75, p�0.07) with AC-T vs AC-XT. In<br />

lobular/mixed patients (n�355), adding capecitabine improved DFS<br />

(HR�0.55, p�0.055) and OS (HR�0.38, p�0.04). There was no difference<br />

in DFS (HR�1.004, p�0.98) in the ER� ductal patients (n�1258)<br />

with the addition <strong>of</strong> capecitabine. Conclusions: Ductal and lobular cancers<br />

have distinct histologic and molecular characteristics; lobular cancers are<br />

generally less sensitive to chemotherapy (Crist<strong>of</strong>anilli, M. JCO, 2005). This<br />

exploratory analysis suggests that patients with lobular/mixed EBC may<br />

benefit from adjuvant capecitabine. This hypothesis requires evaluation in<br />

other adjuvant capecitabine trials.<br />

Hazard ratio by disease histology in patients with different treatment.<br />

End point Treatment<br />

Events in ductal<br />

n�2,195 (%)<br />

Events in lobular/mixed<br />

n�355 (%) HR P value<br />

DFS All patients 254 (11.6) 43 (12.1) 1.003 0.98<br />

AC-T 132 (12.0) 27 (15.3) 1.25 0.30<br />

AC-XT 122 (11.1) 16 (8.9) 0.75 0.28<br />

OS All patients 156 (7.1) 21 (5.9) 0.80 0.33<br />

AC-T 89 (8.1) 15 (8.5) 1.02 0.95<br />

AC-XT 67 (6.1) 6 (3.4) 0.52 0.13<br />

546 General Poster Session (Board #2D), Sat, 8:00 AM-12:00 PM<br />

A meta-analysis <strong>of</strong> receptor status discordance between primary breast<br />

cancer and metastases. Presenting Author: Gaetano Aurilio, European<br />

Institute <strong>of</strong> Oncology, Medical Oncology, Milan, Italy<br />

Background: There is an increasing awareness that biology <strong>of</strong> breast cancer<br />

may evolve over time. The discordance in estrogen (ER), progesterone<br />

(PgR) and HER2 receptor status between primary breast cancer and<br />

metastases is being intensively investigated and a large amount <strong>of</strong> data has<br />

been produced. However, results from different studies seem to be<br />

conflicting and heterogeneous. To highlight this issue, a meta-analysis <strong>of</strong><br />

published data was performed. Methods: A literature search was performed<br />

with Medline. All studies published from 1983 to 2011 comparing<br />

changes inER, PgR and/or HER2 status in patients with matched breast<br />

primary and recurrent tumors were included. We used random-effects<br />

models to estimate pooled discordance proportions. Results: We selected<br />

42 articles, mostly reporting retrospective studies. Twenty-eight, 20 and<br />

27 articles were focused on ER, PgR and HER2 changes, respectively. A<br />

total <strong>of</strong> 2806 tumors were evaluated for ER discordance, 1809 for PgR<br />

discordance and 2801 for HER2 discordance. The heterogeneity between<br />

study-specific discordance proportions was high (I2 �75%, p�0.0001) for<br />

ER, PgR and HER2. Pooled discordance proportions were 20% (95% CI:<br />

16-25%) for ER, 33% (95% CI: 28-38%) for PgR and 9% (95% CI:<br />

6-12%) for HER2. Pooled proportions <strong>of</strong> tumors shifting from positive to<br />

negative and from negative to positive were 24% and 12% for ER<br />

(p�0.0115), respectively. The same figures were 44% and 15% for PgR<br />

(p�0.0001), and 14% and 6% for HER2 (p�0.04). Conclusions: To our<br />

knowledge, this is the first meta-analysis addressing this topic. The findings<br />

strengthen the concept that changes in receptor expression may occur<br />

during the natural history <strong>of</strong> breast cancer and therefore clinical implications<br />

with possible impact on treatment choice cannot be excluded.<br />

However, the high heterogeneity observed in our analysis may explain the<br />

disagreement among oncologists on performing a reassessment <strong>of</strong> the<br />

biological features. In our opinion, only high-powered prospective and<br />

randomized trials could clarify the controversies in this field.<br />

548 General Poster Session (Board #2F), Sat, 8:00 AM-12:00 PM<br />

Bisphosphonates in the adjuvant setting <strong>of</strong> breast cancer therapy: Effect on<br />

survival—A systematic review and meta-analysis. Presenting Author: Liath<br />

Vidal, David<strong>of</strong>f Center, Rabin Medical Center, Petach Tikva, Israel<br />

Background: The role <strong>of</strong> bisphosphonates (BP) in the adjuvant setting in<br />

breast cancer has been evaluated in several studies, yielding inconsistent<br />

evidence. We performed a systematic review and meta-analysis <strong>of</strong> all<br />

randomized controlled trials (RCTs) that evaluate the effects <strong>of</strong> BP<br />

treatment on survival in patients with early breast cancer in the adjuvant<br />

setting. Methods: RCTs that compared BP therapy in addition to the<br />

standard adjuvant therapy (cytotoxic or hormonal) with standard adjuvant<br />

therapy only were identified by searching the Cochrane Library, LILACS,<br />

MEDLINE databases and conference proceedings (12.2011). Hazard<br />

ratios (HRs) <strong>of</strong> overall survival (OS), disease-free survival (DFS) and relative<br />

risks <strong>of</strong> adverse events were estimated and pooled. All statistical tests were<br />

two-sided. Results: Thirteen trials met the inclusion criteria., among which<br />

are the two recently published abstracts <strong>of</strong> large scale RCTs (NSABP-B34,<br />

GAIN) evaluating a total <strong>of</strong> 15,762 patients. Ten trials reported the OS<br />

outcome. Meta-analysis revealed no statistically significant benefit for BP<br />

(HR 0.89, 95% CI � 0.79 to 1.01). Nine trials reported the DFS outcome.<br />

Meta-analysis revealed no statistically significant better DFS for the<br />

intervention (HR 0.95 (0.80-1.11)). Six trials reported DFS stratified upon<br />

menopausal status. Postmenopausal patients who were treated with BP<br />

therapy had statistically significant better DFS than the control group (HR<br />

0.81(0.69-0.95)). In meta-regression, chemotherapy was negatively associated<br />

with HR <strong>of</strong> OS (coefficient, -0.23; standard error, 0.144). BP<br />

therapy resulted in less fractures in the intervention arm, but higher<br />

incidence <strong>of</strong> osteonecrosis <strong>of</strong> the jaw and pyrexia. Conclusions: Our<br />

meta-analysis indicates a positive effect for adjuvant BP on survival<br />

outcomes only in postmenopausal patients with breast cancer. Metaregression<br />

appraised the effect <strong>of</strong> confounders such as chemotherapy,<br />

showed a negative association between chemotherapy use and the effect <strong>of</strong><br />

bisphosphonates on survival. Further large scale RCTs are warranted to<br />

unravel the specific subgroups and adjuvant treatments that would benefit<br />

from the addition <strong>of</strong> BP in the adjuvant setting.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


549^ General Poster Session (Board #2G), Sat, 8:00 AM-12:00 PM<br />

A prospective clinical utility study <strong>of</strong> the impact <strong>of</strong> the 21-gene recurrence<br />

score assay (Oncotype DX) in estrogen receptor positive (ER�) node<br />

negative (pN0) breast cancer in academic Canadian centers. Presenting<br />

Author: James Ashley Davidson, British Columbia Cancer Agency (Fraser<br />

Valley Centre), Surrey, BC, Canada<br />

Background: The Oncotype DX 21-gene Recurrence Score assay (RS) can<br />

potentially predict the magnitude <strong>of</strong> chemotherapy benefit in patients with<br />

stage I-II, node-negative, ER� disease who will be treated with tamoxifen<br />

for 5 years. While use in the United States has grown significantly since its<br />

introduction, it is not yet routinely ordered by oncologists in most parts <strong>of</strong><br />

Canada. The primary purpose <strong>of</strong> this study was to measure the impact <strong>of</strong> the<br />

Oncotype DX test on the physician’s treatment recommendation in ER�<br />

pN0 breast cancer in British Columbia. Methods: After providing informed<br />

consent, patients and medical oncologists completed respective pre-RS<br />

questionnaires indicating their treatment preferences and level <strong>of</strong> confidence<br />

and a decisional conflict scale (patients only). At a subsequent visit,<br />

after the RS result was known and discussed, the patient and oncologist<br />

completed a second set <strong>of</strong> questionnaires. The proportion <strong>of</strong> physician<br />

treatment recommendations that changed from baseline to follow-up (post<br />

RS) were calculated with 95% confidence interval (CI). A prospective<br />

health economic (HE) analysis was also performed. Results: From May<br />

2010 to July 2011, two participating BCCA centres enrolled 156 patients.<br />

Of the 150 for whom successful RS assay results were obtained, physicians<br />

changed their chemotherapy recommendation in 45 cases (30%; 95% CI<br />

22.8-38.0%); either to add (10%; 95% CI 5.7-16.0%) or omit (20%; 95%<br />

CI 13.9-27.3%) adjuvant chemotherapy. As a secondary end-point, in 84<br />

cases (56%; 95% CI 47.7-64.1%) there was a change in either the<br />

planned chemo and/or endocrine therapy recommendation. There was an<br />

overall significant improvement in physician confidence post RS (p �<br />

0.001). Patient decisional conflict also significantly decreased following<br />

the RS assay (p � 0.001). The HE analysis is ongoing and will be presented<br />

separately. Conclusions: Within the context <strong>of</strong> a publicly funded health care<br />

system, the RS assay significantly affects adjuvant treatment recommendations<br />

in ER� node negative breast cancer, in addition to reducing patient<br />

decisional conflict.<br />

551 General Poster Session (Board #3B), Sat, 8:00 AM-12:00 PM<br />

Safety <strong>of</strong> everolimus for women over 65 years <strong>of</strong> age with advanced breast<br />

cancer (BC): 12.5-mo follow-up <strong>of</strong> BOLERO-2. Presenting Author: Kathleen<br />

I. Pritchard, Sunnybrook Odette Cancer Centre, University <strong>of</strong> Toronto,<br />

Toronto, ON, Canada<br />

Background: Postmenopausal women with estrogen-receptor–positive (ER� )<br />

BC who relapse/progress on a nonsteroidal aromatase inhibitor (NSAI) are<br />

usually treated with the steroidal AI exemestane (EXE); but there is no<br />

currently approved treatment for this indication. The BOLERO-2 trial<br />

showed that adding everolimus (EVE), an oral inhibitor <strong>of</strong> mammalian<br />

target <strong>of</strong> rapamycin (mTOR), to EXE significantly improved clinical benefit<br />

beyond that <strong>of</strong> EXE alone (Hortobagyi et al, SABCS 2011, Abstract S3-7).<br />

As many women with advanced BC are elderly, the tolerability pr<strong>of</strong>ile <strong>of</strong> EVE<br />

� EXE in this population is <strong>of</strong> interest. Methods: BOLERO-2 is a phase III,<br />

randomized, trial comparing EVE (10 mg once daily) vs placebo (PBO),<br />

both plus EXE (25 mg once daily) in postmenopausal women with advanced<br />

ER� BC progressing or recurring after NSAIs. Safety data with a focus on<br />

elderly patients are reported at 12.5 mo median follow-up. Results:<br />

Baseline disease characteristics, age, and prior cancer therapy were well<br />

balanced between treatment arms (N � 724). At 12.5 months’ median<br />

follow-up, the addition <strong>of</strong> EVE to EXE significantly improved progressionfree<br />

survival in patients �65 (HR � 0.37; P � .05) or �65 years <strong>of</strong> age (HR<br />

� 0.56; P � .05). Adverse events (AEs) <strong>of</strong> special interest (all grades)<br />

occurring more frequently with EVE vs PBO (overall study population)<br />

included stomatitis (66.6% vs 11.3%), infections (50.4% vs 25.2%), rash<br />

(44.0% vs 8.4%), pneumonitis (18.7% vs 0.4%), and hyperglycemia<br />

(15.4% vs 2.5%). Elderly EVE-treated patients (�65 years) had similar or<br />

marginally lower incidence <strong>of</strong> stomatitis (52.1%), rash (32.3%), pneumonitis<br />

(14.6%), and hyperglycemia (12.5%) compared with the overall<br />

population. Grade 3/4 AEs in patients �70 years <strong>of</strong> age (n � 161) reported<br />

only among patients receiving EVE (n � 118) included fatigue (10.2%),<br />

anemia (10.2%), hyperglycemia (8.5%), stomatitis (7.6%), dyspnea<br />

(6.8%), pneumonitis (5.1%), neutropenia (3.4%), and hypertension (3.4%).<br />

Conclusions: Adding EVE to EXE was well tolerated in the overall population<br />

and in elderly patients with advanced BC; grade 3/4 AEs were uncommon<br />

and manageable. Overall, AEs were consistent with the known safety pr<strong>of</strong>ile<br />

<strong>of</strong> EVE.<br />

Breast Cancer—HER2/ER<br />

19s<br />

550 General Poster Session (Board #3A), Sat, 8:00 AM-12:00 PM<br />

CYP2D6 genotype and adverse effects as indicators <strong>of</strong> plasma endoxifen in<br />

breast cancer patients taking tamoxifen. Presenting Author: Bavanthi<br />

Balakrishnar, Westmead Hospital, University <strong>of</strong> Sydney, Sydney, Australia<br />

Background: Tamoxifen is a prodrug. Its principal active metabolite endoxifen<br />

is a product <strong>of</strong> cytochrome P450 2D6 (CYP2D6) metabolism. The<br />

CYP2D6 gene is highly polymorphic with a number <strong>of</strong> relatively common<br />

reduced function alleles. The aim <strong>of</strong> this study was to determine whether<br />

plasma endoxifen levels were reflected by CYP2D6 genotype or adverse<br />

effects in individuals taking tamoxifen. Methods: Plasma endoxifen was<br />

measured by High Performance Liquid Chromatography / Mass Spectroscopy<br />

in 90 breast cancer patients taking 20mg tamoxifen per day. Ten<br />

CYP2D6 single nucleotide polymorphisms were assessed to designate four<br />

putative CYP2D6 functional categories: ultra-rapid (UM), extensive (EM),<br />

intermediate (IM) and poor (PM) metabolizers. CYP2D6 inhibitor use and<br />

adverse effects were documented. The study was part <strong>of</strong> an ongoing<br />

Australian trial <strong>of</strong> tamoxifen dose escalation. Results: There was marked<br />

variation in plasma endoxifen levels across the cohort (mean 27.6 nM, SD<br />

14.3). Endoxifen levels were significantly associated with metabolizer<br />

categories (p�0.001, r� -0.44), but were not distinctive between categories.<br />

For example, in the EM category (n�46) endoxifen levels ranged from<br />

3.8-72.2 nM (mean 32.6 nM) with levels in the lowest quartile (3.8-19.7<br />

nM) substantially overlapping the PM category (n�11); 6.1-24.7 nM.<br />

Consistent with an impact <strong>of</strong> non-CYP2D6 genotype related factors on<br />

endoxifen levels, endoxifen was significantly lower in 18 patients taking<br />

CYP2D6 inhibitor medications (p�0.005). There was no association<br />

between endoxifen levels and vasomotor symptoms or other adverse effects<br />

<strong>of</strong> tamoxifen. Conclusions: Endoxifen levels were highly variable in patients<br />

taking standard dose tamoxifen, and not predicted by CYP2D6 genotype or<br />

adverse effects. Therapeutic monitoring <strong>of</strong> endoxifen levels may be a useful<br />

approach to assess tamoxifen activity.<br />

552 General Poster Session (Board #3C), Sat, 8:00 AM-12:00 PM<br />

Prospective comparison <strong>of</strong> recurrence score and independent central<br />

pathology assessment <strong>of</strong> prognostic tools in early breast cancer (BC): Focus<br />

on HER2, ER, PR, Ki-67 results from the phase III WSG-Plan B trial.<br />

Presenting Author: Oleg Gluz, West German Study Group, Moenchengladbach,<br />

Germany<br />

Background: Use <strong>of</strong> multi-gene real-time PCR (RT-PCR) based assays e.g.<br />

Recurrence Score (RS) and single markers (grade, uPA/PAI-1, ER/PR,<br />

HER2, KI-67) is currently controversially discussed in early BC. Here, we<br />

present the final WSG-planB trial correlation analysis <strong>of</strong> risk assessment<br />

tools and first prospective comparison <strong>of</strong> independent central pathology<br />

IHC/FISH assessment and RT-PCR for single markers. Methods: Plan B trial<br />

(n�2,448 randomized for 6xTC vs. 4xEC-4xDOC in locally HER2- BC). RS<br />

has been used as selection criterion for cht omission in HR� BC (if RS�11<br />

in pN0 or pN1). uPA/PAI-1 was optionally obtained. Grade, ER/PR, HER2<br />

(IHC/FISH), Ki-67 were evaluated by the independent trial pathologist in<br />

all tumors. Results: From 04/09 to 11/11, 3196 patients have been<br />

recruited and 2448 randomized. RS distribution in 2551 HR� tumors:<br />

0-11 (18%), 12-25 (60%), �25 (22%). In 354 pN0-1 patients, cht was<br />

omitted based on low risk RS (88% compliance). Central grade for n�3038<br />

and IHC/FISH results are currently available in n�1476. Moderately<br />

significant correlations were only found between RS and both central grade<br />

(rs�0.313; p�0.001) as well as Ki-67 (rs�0.374; p�0.001) and a weak<br />

one for uPA/PAI-1, particularly due to poor correlations within the RS group<br />

�26. In 1476 locally HER2- cases, n�9 were found as 3� and/or FISH�<br />

by central analysis. In 6 HR�/HER2� cases, RS revealed 2 positive, 2<br />

equivocal and 2 negative results. In 7 cases positive for HER2 by RT-PCR<br />

central pathology revealed 4 negative results. 24 locally HR� cases are<br />

assessed as HR- in central pathology (2%). Among these, 6 were ER<br />

positive by RT-PCR. Final correlation analyses will be presented at the<br />

meeting. Conclusions: These first prospective data demonstrate that highrisk<br />

status according to RS is predictive <strong>of</strong> high risk by other factors, but the<br />

converse is not true. Regarding controversial HER2 and HR status by<br />

RT-PCR and IHC/FISH, we found few cases with false-negative or positive<br />

RT-PCR results in HER2- BC by local pathology. However, these discrepancies<br />

could potentially have a substantial impact on clinical patient<br />

management.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


20s Breast Cancer—HER2/ER<br />

553 General Poster Session (Board #3D), Sat, 8:00 AM-12:00 PM<br />

Cost-effectiveness <strong>of</strong> zoledronic acid (ZOL) in postmenopausal women with<br />

early breast cancer receiving adjuvant letrozole. Presenting Author: Mei<br />

Xue, Pharmerit North America, LLC, Bethesda, MD<br />

Background: In the ZO-FAST trial, postmenopausal women with early breast<br />

cancer and a bone mineral density (BMD) T-score � –2 and receiving<br />

adjuvant letrozole (2.5 mg/day) were randomized to either immediate ZOL<br />

(4 mg/6 months) treatment (upfront ZOL) or to the same therapy but only<br />

when BMD T-score decreased to � –2 or fracture occurrence (delayed<br />

ZOL). After 60 months, upfront ZOL increased both BMD and disease-free<br />

survival (P � .05) relative to delayed ZOL. The present analysis assessed,<br />

from a US payer perspective, the cost effectiveness <strong>of</strong> upfront ZOL vs<br />

delayed ZOL in this population. Methods: A Markov state-transition model<br />

was developed to estimate the lifetime costs and quality-adjusted life-years<br />

(QALYs) for a hypothetical cohort <strong>of</strong> postmenopausal women with early<br />

breast cancer receiving letrozole with upfront or delayed ZOL. Consistent<br />

with ZO-FAST, patients were 57 years <strong>of</strong> age and breast cancer recurrencefree<br />

at baseline. Patients could progress over time to Local Recurrence,<br />

Contra-lateral Tumor, Distant Recurrence, or Death. Transition probabilities<br />

were derived from ZO-FAST, supplemented with literature. Costs and<br />

utilities were literature based. All outcomes were discounted 3% per year.<br />

Results: Compared to delayed ZOL, upfront ZOL resulted in better overall<br />

survival, disease-free survival, and QALYs, but at a higher cost (Table). In<br />

the base case, the incremental cost/QALY gained with upfront vs delayed<br />

ZOL was $7,967. In � 95% <strong>of</strong> 1,000 probabilistic sensitivity analysis<br />

runs, upfront ZOL costs less than $38,376/QALY gained. Conclusions:<br />

Upfront ZOL may increase survival and QALY and, at a cost per QALY well<br />

under the $50,000/QALY threshold, is very cost-effective in this population.<br />

Life-years<br />

(95% CI)<br />

Upfront ZOL 20.90<br />

(20.15-21.68)<br />

Delayed ZOL 19.29<br />

(18.32-20.29)<br />

Difference 1.61<br />

(0.22-3.03)<br />

Disease-free<br />

life-years<br />

(95% CI)<br />

20.17<br />

(19.34-21.06)<br />

18.12<br />

(17.02-19.26)<br />

2.06<br />

(0.48-3.63)<br />

QALY<br />

(95% CI)<br />

14.36<br />

(13.58-15.02)<br />

13.43<br />

(12.58-14.20)<br />

0.93<br />

(1.12-1.75)<br />

Lifetime cost<br />

(95% CI)<br />

$86,594<br />

($62,904-$111,010)<br />

$79,159<br />

($53,964-$103,680)<br />

$7,436<br />

(–$2,050-$16,738)<br />

Incremental<br />

cost per QALY<br />

$7,967<br />

(95% upper CI<br />

limit � $38,376)<br />

555 General Poster Session (Board #3F), Sat, 8:00 AM-12:00 PM<br />

Obesity at diagnosis and breast cancer (BC) recurrence risk based on the<br />

21-gene assay recurrence score (RS). Presenting Author: Kimberly Ridolfi,<br />

Medical College <strong>of</strong> Wisconsin, Milwaukee, WI<br />

Background: Obesity at diagnosis has been associated with poorer overall survival<br />

in BC patients. The Oncotype Dx 21-gene assay RS has been shown to predict<br />

the risk <strong>of</strong> distant recurrence in estrogen receptor (ER) positive BC. This study<br />

aimed to evaluate if an association exists between body mass index (BMI) and<br />

RS. Methods: Retrospective chart review <strong>of</strong> patients (pts) with BC diagnosed<br />

2005-2010 and a 21-gene assay was performed. Risk factors including BMI,<br />

hormone use, and menopausal status were collected as well as tumor characteristics<br />

such as ER/PR/HER2 status, stage, and grade. Univariate analysis <strong>of</strong> the<br />

association between BMI level (�30 vs �30), and RS (�18 vs �18), stage (I vs<br />

II), and grade (1 vs 2 vs 3) was conducted using Chi square test. Correlation <strong>of</strong><br />

BMI with RS (0-100) was also assessed via Spearman’s correlation. All tests<br />

were at a two sided significance level <strong>of</strong> 0.05. Results: Of 495 pts with a RS<br />

value, 482 had a BMI within 6 months <strong>of</strong> diagnosis. Median BMI was 27.7<br />

kg/m2 (range 17-63). BMI for pts presenting with stage 2 disease is significantly<br />

higher (30�6.9) than those with stage 1 (29�7.8), p�0.04 (Wilcoxon rank sum<br />

test). No association was found between BMI level and stage, grade or RS. See<br />

Table. Mean RS was 18.8 vs 18.2 (BMI �30 vs �30), p�0.79 (Wilcoxon rank<br />

sum test). Conclusions: This is the largest study <strong>of</strong> the association between BMI<br />

and the RS. Obesity (BMI�30) has been associated with increased recurrence<br />

and death from BC. However, we found no association between BMI and RS. This<br />

suggests that poorer outcomes among obese ER� breast cancer patients may not<br />

be due to intrinsic tumor biology (as reflected in the RS), but to other non-tumor<br />

factors, whether higher circulating estrogen levels, reduced efficacy <strong>of</strong> aromatase<br />

inhibitors, other comorbidities, difficulty with chemotherapy or other<br />

unidentified factors. Further study is warranted.<br />

BC in pts with BMI 30.<br />

BMI 30 (N�184)<br />

N % N % p value*<br />

Grade Grade 1 99 33.2 56 30.4 0.44<br />

Grade 2 162 54.4 96 52.2<br />

Grade 3 34 11.4 28 15.2<br />

Unknown 3 1.0 4 2.2<br />

Stage I 213 71.5 123 66.9 0.14<br />

II 71 23.8 57 31.0<br />

Unknown 14 4.7 4 2.2<br />

Risk group RS�18 166 55.7 98 53.3 0.67<br />

RS�18 132 44.3 86 46.7<br />

*Chi-Square test, excluding unknown.<br />

554 General Poster Session (Board #3E), Sat, 8:00 AM-12:00 PM<br />

Chemotherapy-induced amenorrhea in premenopausal breast cancer patients<br />

treated with adjuvant anthracycline and taxane based chemotherapy<br />

within the SUCCESS study. Presenting Author: Julia Katharina Neugebauer,<br />

Department <strong>of</strong> Gynecology and Obstetrics, Ludwig-Maximilians-<br />

University, Munich, Germany<br />

Background: Chemotherapy induced amenorrhea (CIA) is <strong>of</strong>ten used as a<br />

surrogate marker for cytotoxic gonadal damage. The aim <strong>of</strong> this analysis was<br />

to identify predicitve factors for CIA in premenopausal breast cancer<br />

patients treated with adjuvant chemotherapy. Methods: The German multicenter,<br />

phase III SUCCESS trial compared 3 cycles <strong>of</strong> FEC q3w followed by<br />

3x docetaxel q3w vs. FEC followed by Doc�gemcitabine as adjuvant<br />

treatment in patients with node positive or high risk node negative primary<br />

breast cancer. Premenopausal patients with hormone receptor positive<br />

tumors underwent endocrine treatment with tamoxifen (Tam) for 5 years.<br />

Additional goserelin (GnRH) for 2 years was given in the following cases:<br />

Age �40 years at study entry, premenopausal hormone status or regular<br />

menses within 6 months after chemotherapy. We retrospectively investigated<br />

CIA rates <strong>of</strong> 1.189 initially premenopausal patients during diseasefree<br />

follow-up. Results: Median age at diagnosis was 44 years (range<br />

21-50). The median follow up time was 44 months (range 1-62). 791<br />

patients (66.5%) received endocrine treatment. Tam intake was reported<br />

in 370 patients, GnRH in 29 patients, Tam�GnRH in 392 patients. In 963<br />

patients (81.0%) CIA occurred for �3 months after chemotherapy. In<br />

22.7% <strong>of</strong> initially amenorrheic patients CIA was temporary. 192 patients<br />

(16.2%) had continuous menstrual bleeding. In multivariate analysis the<br />

risk <strong>of</strong> CIA was significantly higher in older patients, increasing by factor<br />

1.32 per life year (p�0.0001). Tam intake was also associated with<br />

increased CIA rates (p�0.0032). Patients with combined Tam�GnRH<br />

were more likely to resume menses compared to patients without endocrine<br />

therapy (p�0.0056). Factors like the cytotoxic regimen, tumor stage,<br />

grading, the patient�s BMI or the use <strong>of</strong> GnRH analogues as ovarian<br />

protectants during chemotherapy did not correlate with CIA rates.<br />

Conclusions: Age was the strongest predictor for CIA. Tam administered<br />

alone increased CIA rates. Opposite effects were observed for patients who<br />

received Tam�GnRH. The addition <strong>of</strong> gemcitabine to sequential anthracyline-taxane<br />

based chemotherapy did not influence CIA.<br />

556 General Poster Session (Board #3G), Sat, 8:00 AM-12:00 PM<br />

Everolimus (RAD) as treatment in breast cancer patients with bone<br />

metastases only: Results <strong>of</strong> the phase II RADAR study. Presenting Author:<br />

Nicolai Maass, University <strong>of</strong> Aachen, Aachen, Germany<br />

Background: RAD001 is an orally bioavailable rapamycin ester analogue,<br />

which acts by selectively inhibiting mTOR (mammalian target <strong>of</strong> rapamycin),<br />

a key player in downstream signaling <strong>of</strong> different pathways. In vitro,<br />

RAD stops formation and activity <strong>of</strong> osteoclasts. Treating progressive bone<br />

metastases in breast cancer (bc) with RAD seems reasonable. Methods: We<br />

evaluated RAD in a placebo-controlled, phase II, randomized discontinuation<br />

study in bc patients (pts) with bone metastases only. Pts were eligible<br />

if they had HER2-negative, hormone-receptor (HR)-positive or –negative<br />

bc, with a maximum <strong>of</strong> 2 previous lines <strong>of</strong> endocrine therapy (ET) and 1<br />

previous line <strong>of</strong> chemotherapy (CT). All pts received zoledronate and pts<br />

with HR-positive bc could receive ET. All pts started with RAD during a<br />

run-in phase <strong>of</strong> 8 weeks. Pts with stable disease were randomized to RAD or<br />

placebo; pts with response continued with RAD and pts with progression<br />

went <strong>of</strong>f study. Primary outcome was time to progression (TTP) in pts being<br />

stable on 8 weeks <strong>of</strong> RAD. It was assumed that placebo would obtain a<br />

median TTP <strong>of</strong> 8 weeks which would be increased to 16 weeks, thus<br />

requiring 76 randomized pts. It was expected that 70% <strong>of</strong> all pts would<br />

have stable disease after the run-in phase. Overall, 110 pts were needed.<br />

Due to slow recruitment and dysbalance between randomized and discontinued<br />

pts, recruitment stopped in 12/ 2010. Results: From 11/06 until<br />

12/10, 89 pts were enrolled. Median age was 59.5 years. 93% had<br />

HR-positive disease. 15% had prior chemotherapy; 58% had prior ET for<br />

metastases. 1/3 received concomitant ET. Three pts did not start therapy,<br />

41 discontinued during run-in phase, 32 due to progression. Six continued<br />

as responder. 39 pts with SD after run in phase were randomized to RAD or<br />

placebo. Twenty-seven stopped due to progression; 9 discontinued due to<br />

AE, 4 are still on treatment. 15 pts had 20 serious adverse events; 1<br />

hyperglycemia and one alveolitis. The TTP in patients with RAD was 8.5<br />

months vs. 2.9 months with placebo (HR: 0.559; 95% CI [0.284-1.10]<br />

p�0.092. Conclusions: Pts with bone metastases only had a longer TTP on<br />

RAD compared to pts on placebo. Overall 7/89 showed a sustained<br />

response on RAD � zoledronate � ET.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


557 General Poster Session (Board #3H), Sat, 8:00 AM-12:00 PM<br />

Predicting the Oncotype DX score: An inexpensive guesstimation. Presenting<br />

Author: Catherine Pesce, Memorial Sloan-Kettering Cancer Center,<br />

New York, NY<br />

Background: Oncotype DX Recurrence Score [RS] is used to predict the<br />

benefits <strong>of</strong> chemotherapy added to adjuvant hormone therapy in ER<br />

positive early stage breast cancer. It is also prognostic. However, its<br />

expense may be a concern in some health care systems and communities.<br />

In addition, it is labor intensive, requiring shipment <strong>of</strong> tissue samples to a<br />

single laboratory with an average 10-14 day turnaround time (in the US). A<br />

reliable and inexpensive estimator <strong>of</strong> the Oncotype DX risk score using<br />

readily available pathologic variables from tumor specimens could be<br />

useful. Methods: We reviewed our prospective database <strong>of</strong> patients with<br />

Oncotype DX results obtained over 2.5 years (1155 specimens with<br />

Oncotype DX scores, September 2008 – March 2011) and identified 766<br />

invasive ductal carcinomas with known ER status, PR status, histologic<br />

grade, and nuclear grade. Through linear regression analysis, we predicted<br />

recurrence score for each tumor using these four parameters. After<br />

categorizing according to the same risk classification as in Oncotype DX<br />

(low risk: RS�18, intermediate risk: RS 18-30, or high risk: RS�30) we<br />

compared our predicted recurrence score to actual Oncotype DX recurrence<br />

score. Results: Overall 69.7% <strong>of</strong> specimens were assigned the same risk<br />

category as with the Oncotype DX level. In the predicted low risk group,<br />

1.7% <strong>of</strong> patients were actually high risk. None <strong>of</strong> the predicted high risk<br />

patients had a low score. Conclusions: We found a strong correlation<br />

between scores estimated using our model and actual reported Recurrence<br />

Scores. If validated, our model could provide a clinically useful estimation<br />

<strong>of</strong> risk at lower cost.<br />

Predicted risk level Actual risk level<br />

Low risk Intermediate risk High risk Total<br />

Low risk 358<br />

97<br />

8 463<br />

(77.3%) (21.0%) (1.7%) (59.7%)<br />

Intermediate risk 95<br />

168<br />

34 297<br />

(32.0%) (56.6%) (11.5%) (38.3%)<br />

High risk 0<br />

1<br />

15 16<br />

(0%) (6.2%) (93.8%) (2.1%)<br />

Total 453<br />

266<br />

57 776<br />

(58.4%) (34.3%) (7.4%)<br />

559 General Poster Session (Board #4B), Sat, 8:00 AM-12:00 PM<br />

Everolimus for postmenopausal women with advanced breast cancer:<br />

Updated results <strong>of</strong> the BOLERO-2 phase III trial. Presenting Author:<br />

Martine Piccart, Institut Jules Bordet, Université Libre de Bruxelles,<br />

Brussels, Belgium<br />

Background: Current treatment options for postmenopausal patients with<br />

estrogen-receptor–positive (ER� ) breast cancer (BC) who relapse or progress<br />

on a nonsteroidal aromatase inhibitor (NSAI) are limited. The BO-<br />

LERO-2 trial supports the activity <strong>of</strong> everolimus (EVE; an oral mammalian<br />

target <strong>of</strong> rapamycin [mTOR] inhibitor) added to the steroidal aromatase<br />

inhibitor exemestane (EXE) to prolong progression-free survival (PFS) in<br />

this patient population. Long-term PFS and survival data are awaited.<br />

Methods: BOLERO-2 is a phase III double-blind, randomized, international<br />

trial comparing EVE (10 mg once daily) plus EXE (25 mg once daily) versus<br />

placebo (PBO) plus EXE in postmenopausal women with advanced ER� BC<br />

progressing or recurring after NSAIs (letrozole or anastrozole). Patients<br />

were randomized (2:1) to EVE � EXE or PBO � EXE. The primary endpoint<br />

was PFS by local investigator assessment. Main secondary endpoints<br />

included centrally assessed PFS, overall survival (OS), safety, bone<br />

turnover, and overall response rate (ORR). Results: Baseline disease<br />

characteristics including tumor burden and prior cancer therapy were well<br />

balanced between treatment arms (N � 724). Median PFS was doubled<br />

and response rates were consistently improved with EVE � EXE (n � 485)<br />

vs PBO � EXE (n � 239) in interim analyses. Median PFS by local<br />

assessment was ~3 mo with PBO � EXE vs 6.9 mo (hazard ratio [HR] �<br />

0.43; P � .0001) and 7.4 mo (HR � 0.44; P � .0001) with EVE � EXE at<br />

7.5 mo and 12.5 mo follow-up, respectively. Fewer deaths were reported<br />

with EVE � EXE (17.2%) vs PBO � EXE (22.7%) at 12.5 mo follow-up.<br />

Safety pr<strong>of</strong>iles were consistent with previous reports for mTOR inhibitors.<br />

PFS data including 528 events (protocol-specified final analysis), and<br />

updated OS and safety data will be presented. Conclusions: Adding EVE to<br />

EXE markedly prolonged PFS in patients with NSAI-refractory advanced<br />

ER� BC. There were fewer deaths among patients receiving EVE, and<br />

further follow-up will evaluate the effect <strong>of</strong> EVE on OS.<br />

Breast Cancer—HER2/ER<br />

21s<br />

558 General Poster Session (Board #4A), Sat, 8:00 AM-12:00 PM<br />

Aromatase inhibitors and cardiac outcomes in women undergoing cardiac<br />

angiography after early breast cancer. Presenting Author: Bostjan Seruga,<br />

Institute <strong>of</strong> Oncology Ljubljana, Ljubljana, Slovenia<br />

Background: Data show that in post-menopausal women with early breast<br />

cancer, longer use <strong>of</strong> aromatase inhibitor (AI) is associated with increased<br />

odds <strong>of</strong> ischemic heart disease. Here we explore the association between<br />

adjuvant AI use and cardiac disease in women undergoing cardiac<br />

angiography after a diagnosis <strong>of</strong> early breast cancer. Methods: We linked a<br />

database <strong>of</strong> 7,681 women who underwent cardiac angiography at the<br />

University <strong>Clinical</strong> Center <strong>of</strong> Ljubljana between December 2004 and<br />

November 2010 with the Cancer Registry for Slovenia. Women with early<br />

breast cancer that subsequently underwent cardiac angiography were<br />

identified. Information on cardiovascular risk factors was retrieved from the<br />

patients’ charts and from discharge letters after cardiac angiography. The<br />

endpoint <strong>of</strong> interest was a diagnosis <strong>of</strong> ischemic heart disease or left<br />

ventricular dysfunction (IHD-LVD) without evidence <strong>of</strong> valvular heart<br />

disease at the time <strong>of</strong> angiography. Conditional, logistic regression was<br />

used to test for associations between variables. Results: Among 117 eligible<br />

women 75% (n�88) were postmenopausal and 62% (n�73) had hormonal<br />

receptor positive (HR�) disease. Of these 42% (n�31) were treated with<br />

AI. Overall, 48% (n�56) <strong>of</strong> women were found to have IHD-LVD. In<br />

patients with HR� breast cancer, use <strong>of</strong> AIs was significantly associated<br />

with IHD-LVD as compared to tamoxifen alone (HR 2.50, 95% CI<br />

1.01-6.29, p�0.046). For each year <strong>of</strong> AI therapy, there was a trend for<br />

higher odds <strong>of</strong> IHD-LVD (OR: 1.25, 95% CI 0.95-1.67, p�0.116). This<br />

effect appeared independent <strong>of</strong> age, body mass index, baseline hypertension,<br />

hypercholesterolemia, diabetes and heart disease or prior anthracyclines<br />

exposure. Among all patients, use <strong>of</strong> anthracyclines and left-sided<br />

irradiation was associated with non-significant increases in IHD-LVD (HR<br />

2.37, 95% CI 0.89-6.09, p�0.45 and HR 1.28, 95% CI 0.69-2.40,<br />

p�0.44 respectively). Conclusions: Compared to tamoxifen, AIs are associated<br />

with a time dependent increase in IHD-LVD. This risk appears<br />

independent <strong>of</strong> other risk factors for heart disease. Anthracycline exposure<br />

and left breast or chest wall radiation showed non-significant associations<br />

with IHD-LVD in this small cohort.<br />

560 General Poster Session (Board #4C), Sat, 8:00 AM-12:00 PM<br />

The impact <strong>of</strong> the Oncotype Dx breast cancer assay in clinical practice:<br />

Systematic review and meta-analysis. Presenting Author: Josh John Carlson,<br />

University <strong>of</strong> Washington, Seattle, WA<br />

Background: Many studies have evaluated the Oncotype Dx Breast Cancer<br />

Assay (ODX) and its impact on adjuvant chemotherapy (ACT) treatment<br />

decisions. However, it can be difficult for clinicians and other stakeholders<br />

to interpret the collective findings <strong>of</strong> these studies, as they were conducted<br />

in diverse settings and have limited sample sizes. To address this issue, we<br />

conducted a systematic review and meta-analysis to synthesize ODX results<br />

and provide insights about the utility <strong>of</strong> ODX in actual clinical practice.<br />

Methods: We performed a systematic review <strong>of</strong> ODX studies using PubMed,<br />

Embase, ASCO, and SABCS. Studies were included if patients had ER �,<br />

node -, early stage breast cancer, reported use <strong>of</strong> ODX to inform actual ACT<br />

decisions, and reported outcomes <strong>of</strong> interest, including: 1) distribution <strong>of</strong><br />

ODX recurrence scores (RS); 2) impact <strong>of</strong> ODX on ACT recommendations;<br />

3) impact <strong>of</strong> ODX on ACT use; and 4) proportion <strong>of</strong> patients following the<br />

treatment suggested by the ODX RS. Results: A total <strong>of</strong> 28 studies met<br />

inclusion criteria. The distribution <strong>of</strong> RS categories was 48.8% low, 39.0%<br />

intermediate, and 12.2% high (21 studies, 4,156 patients). ODX changed<br />

the clinical-pathological ACT recommendation in 33.4% <strong>of</strong> patients (8<br />

studies, 1,437 patients). In patients receiving ODX, receipt <strong>of</strong> ACT was:<br />

28.2% overall, 5.8% low, 37.4% intermediate, and 83.4% high. High RS<br />

patients were significantly more likely to follow the treatment suggested by<br />

ODX vs. low RS patients RR: 1.07 (1.01–1.14). Conclusions: The pooled<br />

results for the impact <strong>of</strong> ODX on ACT recommendations are consistent with<br />

most individual studies to date. However, the proportion <strong>of</strong> intermediate RS<br />

results is nearly 2-fold higher than reported in the ODX development<br />

studies by Paik et al., which may have implications for the clinical utility<br />

and cost impacts <strong>of</strong> testing. Also, high RS vs. low RS patients were more<br />

likely to follow the ODX results, suggesting a tendency toward aggressive<br />

treatment despite a low ODX RS. Finally, there was a lack <strong>of</strong> studies on the<br />

impact <strong>of</strong> ODX on ACT use vs. standard approaches, suggesting additional<br />

studies are warranted.<br />

Count Mean % classified (95% CI)<br />

High RS 509 12.2% (10.5-14.0)<br />

Int RS 1,487 39.0% (36.3-41.7)<br />

Low RS 2,026 48.8% (45.2-52.3)<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


22s Breast Cancer—HER2/ER<br />

561 General Poster Session (Board #4D), Sat, 8:00 AM-12:00 PM<br />

Assessment <strong>of</strong> the prognostic and predictive utility <strong>of</strong> the breast cancer<br />

index (BCI): An NCIC CTG MA.14 study. Presenting Author: Dennis Sgroi,<br />

Massachusetts General Hospital, The Avon Foundation, Boston, MA<br />

Background: Breast Cancer Index (BCI), a continuous risk index, combines<br />

the ratio <strong>of</strong> HOXB13 to IL17BR (H/I) and the molecular grade index (MGI)<br />

(Jerevall et al., British J Cancer, 2011). Here, the prognostic and predictive<br />

performance <strong>of</strong> BCI for BC relapse in MA.14 trial was examined. Methods:<br />

MA.14 randomly assigned 667 hormone receptor positive (HR�) women to<br />

5 years <strong>of</strong> tamoxifen (TAM) �/- 2 years <strong>of</strong> octreotide LAR (TAM-OCT). A<br />

representative subgroup <strong>of</strong> 299 patients was pr<strong>of</strong>iled by RT-PCR for BCI.<br />

The primary objective was to determine the prognostic performance <strong>of</strong> BCI<br />

based on relapse-free survival (RFS) with median 9.8 years follow-up.<br />

Association <strong>of</strong> BCI was assessed with step-wise forward stratified Cox<br />

regression. Pre-defined categories <strong>of</strong> low (L), intermediate (I) and high (H)<br />

BCI risk groups were used to provide adjusted 5- and 10-year RFS. Results:<br />

292 <strong>of</strong> 299 patient samples passed internal analytical quality control. The<br />

292 patients contained 49% LN� patients and had 19.9% BC relapses.<br />

Both continuous and pre-specified BCI risk groups were significant multivariate<br />

factors (p�0.0001; p�0.007) with higher BCI associated with shorter<br />

RFS. Adjusted univariate hazard ratios and 95% CI were 2.53 (1.36 –<br />

4.71) for BCI-H vs -L and 1.28 (0.65 – 2.52) for BCI-I vs -L. With both LNand<br />

LN� included, BCI-L had 5- and 10-year RFS <strong>of</strong> 94.0% and 87.5%;<br />

-I, 91.8% and 83.9%; and -H, 81.5% and 74.7%. No significant<br />

difference in BCI’s ability to stratify patients into 3 risk groups was<br />

observed between LN-/no chemotherapy subgroup vs balance <strong>of</strong> MA.14<br />

patients (p�0.26). Higher pathologic T-stage was multivariately associated<br />

with shorter RFS (p�0.01). Interactions between trial therapy and BCI<br />

was not significant (p�0.40). Conclusions: This study confirmed the strong<br />

prognostic effect <strong>of</strong> BCI on breast cancer recurrence. BCI was prognostic in<br />

both LN- and LN� patients. The lower 10-year RFS in the BCI-L group than<br />

in our previous studies reflected the mixed LN-/LN� population examined.<br />

Like the parent MA.14 trial, BCI did not predict benefit <strong>of</strong> adding OCT to<br />

TAM therapy (Pritchard et al., J Clin Oncol, 2011). This retrospective study<br />

is an independent validation <strong>of</strong> the prognostic performance <strong>of</strong> BCI within a<br />

prospective trial.<br />

563 General Poster Session (Board #4F), Sat, 8:00 AM-12:00 PM<br />

Impact <strong>of</strong> exogenous female hormone use (EHU) on breast cancer (BC)<br />

recurrence as assessed by the 21-gene assay (Oncotype DX). Presenting Author:<br />

Wendy M. Ledesma, University <strong>of</strong> Wisconsin Carbone Cancer Center, Madison,<br />

WI<br />

Background: Observational studies suggest that EHU, such as oral contraceptive<br />

(OCPs) or postmenopausal hormone replacement therapy (HRT), increases the<br />

risk <strong>of</strong> developing BC. However, the prognosis <strong>of</strong> women taking EHU compared to<br />

nonusers remains controversial. The Oncotype Dx 21-gene assay RS predicts the<br />

distant recurrence risk for early stage, HR� BC. We assessed RS in patients (pts)<br />

on EHU at diagnosis (users) vs nonusers. Methods: Retrospective chart review<br />

was conducted on BC pts with a RS, diagnosed 2005–2010. Stage, grade,<br />

estrogen (ER), progesterone (PR) and HER2 receptor status were abstracted.<br />

Manual review <strong>of</strong> medications, as well as electronic data pull within �5yrs <strong>of</strong><br />

diagnosis, defined users as pts on EHU within 1 yr <strong>of</strong> diagnosis for �1 yr.<br />

Nonusers had been on EHU � 5 yrs before diagnosis or never. Comparisons<br />

between EHU users and nonusers were made using two-sample tests; chi-square<br />

or Fisher’s exact test for discrete data such as stage and ER status and t-test or<br />

Wilcoxon rank sum test for continuous data such as RS. All tests were at a<br />

two-sided significance level <strong>of</strong> 0.05. Results: 495 pts had RS. EHU was<br />

documentable for 475, 77 were using exogenous hormone at diagnosis. Type <strong>of</strong><br />

hormone use included systemic HRT (48%), OCPs (36%), other (4%), and<br />

unknown (12%). For users vs nonusers, mean RS was 17 vs 19, (p�0.007,<br />

t-test). See table for other predictors. Conclusions: Prior study shows21-gene<br />

assay predict distant recurrence risk for HR�BC. In our study, an association<br />

existed between EHU and lower RS, suggesting that BC developing on EHU may<br />

have less risk <strong>of</strong> recurrence. Prospective evaluation is warranted.<br />

Users at diagnosis versus nonusers.<br />

Nonusers N�398 Users N�77 P-value<br />

RS #<br />

19 (9.2) 17 (6.3) 0.007*<br />

Age #<br />

57 (10.3) 54 (9.4) 0.04*<br />

Stage I 280 (70%) 54 (70%) 0.8 ^<br />

II 105 (26%) 18 (23%)<br />

Grade<br />

Unknown<br />

1<br />

13 (3%)<br />

117 (29%)<br />

5 (6%)<br />

32 (42%) 0.013 ^<br />

2 217 (54%) 40 (52%)<br />

3 57 (14%) 3 (4%)<br />

ER<br />

Unknown<br />

�<br />

7 (2%)<br />

386 (97%)<br />

2 (3%)<br />

74 (96%) 1 $<br />

- 4 (1%) 0<br />

PR<br />

Unknown<br />

�<br />

8 (2%)<br />

340 (85%)<br />

3 (4%)<br />

70 (91%) 0.11 ^<br />

- 49 (12%) 4 (5%)<br />

HER2<br />

Unknown<br />

�<br />

9 (2%)<br />

4 (1%)<br />

3 (4%)<br />

1 (1%) 0.6 $<br />

- 368 (92%) 69 (90%)<br />

Unknown 26 (7%) 7 (9%)<br />

# Mean(SD);*t-test; ^ Chi-square test (excluding unknown); $ Fisher’s exact test (excludes<br />

unknown).<br />

562 General Poster Session (Board #4E), Sat, 8:00 AM-12:00 PM<br />

Association <strong>of</strong> the phosphorylation <strong>of</strong> the estrogen receptor at serine 118<br />

and 167 with prognosis in postmenopausal breast cancer patients. Presenting<br />

Author: Karin J. Beelen, Netherlands Cancer Institute, Amsterdam,<br />

Netherlands<br />

Background: The sensitivity <strong>of</strong> the estrogen receptor (ER�) to anti-estrogen<br />

therapy can be affected by phosphorylation events. In premenopausal<br />

breast cancer patients, phosphorylation <strong>of</strong> the ER� at serine 118<br />

(ER�S118-p) is predictive for benefit from adjuvant tamoxifen. Since<br />

ER�S118-p represents the common hallmark <strong>of</strong> different signaling cascades<br />

that differ in E2 dependency, the resulting effect on estrogen<br />

sensitivity may differ between pre- and postmenopausal patients. Phosphorylation<br />

<strong>of</strong> serine 167 (ER�S167-p) has been associated with favorable<br />

disease outcome, but whether ER�S167-p can predict tamoxifen sensitivity<br />

is currently unknown. We tested the predictive value <strong>of</strong> both ER�S118-p<br />

and ER�S167-p for benefit from adjuvant tamoxifen in postmenopausal<br />

breast cancer patients. Methods: We collected primary tumor blocks from<br />

563 ER� positive (stage I-III) postmenopausal patients who had been<br />

randomized between tamoxifen (1 to 3 years) vs. no adjuvant therapy (IKA<br />

trial). The median follow-up <strong>of</strong> patients without a recurrence event was 9.4<br />

years. Immunohistochemistry was performed on a TMA using monoclonal<br />

antibodies for ER�S118-p and ER�S167-p. The percentage <strong>of</strong> positive<br />

nuclei was scored and a score <strong>of</strong> � 10 % was considered as positive.<br />

Multivariate Cox models were used to assess hazard ratios (HRs) for<br />

recurrence free interval and the interaction between these phosphorylations<br />

and tamoxifen treatment. Results: We did not find a significant<br />

interaction between either ER�S118-p (p�0.99) or ER�S167-p (p�0.44)<br />

and tamoxifen, suggesting that the relative benefit from adjuvant tamoxifen<br />

in postmenopausal patients is not dependent on the presence <strong>of</strong> one <strong>of</strong><br />

these phosphorylations. Both tamoxifen treated patients as well as control<br />

patients had a better prognosis when their tumor was positive for ER�S118-p<br />

(adjusted HR 0.60 p�0.02) or ER�S167-p (adjusted HR 0.62, p�0.02)<br />

compared to patients whose tumor did not express these ER� phosphorylations.<br />

Conclusions: In postmenopausal patients ER�S118-p and ER�S167-p<br />

are both associated with better prognosis, but do not predict differential<br />

benefit from tamoxifen.<br />

564 General Poster Session (Board #4G), Sat, 8:00 AM-12:00 PM<br />

Effect <strong>of</strong> MDV3100, an androgen receptor signaling inhibitor, on tumor<br />

growth <strong>of</strong> estrogen and androgen receptor-positive (ER�/AR�) breast<br />

cancer xenografts. Presenting Author: Anthony D. Elias, University <strong>of</strong><br />

Colorado Anschutz Medical Campus, Aurora, CO<br />

Background: The androgen receptor (AR) is detected by immunohistochemistry<br />

in approximately 75% <strong>of</strong> all invasive breast cancer; with ~88% <strong>of</strong> ER�<br />

tumors expressing AR. Potent inhibition <strong>of</strong> AR activity could be a therapeutic<br />

strategy in ER�/AR� breast cancer. MDV3100 is an androgen receptor<br />

signaling inhibitor (ARSI), which inhibits AR activity via three mechanisms:<br />

inhibition <strong>of</strong> androgen binding to AR, inhibition <strong>of</strong> AR nuclear translocation,<br />

and inhibition <strong>of</strong> nuclear AR-DNA binding. MDV3100 has demonstrated<br />

an overall survival benefit in men with post-docetaxel prostate<br />

cancer. Methods: Two ER�/AR� breast cancer cell lines, MCF7 and BCK4<br />

(recently derived from a pleurocentesis), were used to assess the proliferative<br />

effect <strong>of</strong> dihydrotestosterone (DHT) and estradiol (E2). The efficacy <strong>of</strong><br />

MDV3100 at blocking DHT-mediated proliferation was compared to bicalutamide<br />

in ER�/AR� breast cancer cells. MCF7 xenograft studies were<br />

performed to determine if MDV3100 can affect DHT or E2 mediated<br />

proliferation in vivo. Results: Both bicalutamide and MDV3100 inhibited<br />

DHT-mediated proliferation <strong>of</strong> ER�/AR� cell lines. Although MDV3100<br />

binds AR very effectively and does not bind ER, it also uniquely inhibited<br />

E2-mediated proliferation, while bicalutamide slightly enhanced E2 mediated<br />

proliferation. Results presented here demonstrate that MDV3100<br />

inhibited E2-stimulated tumor growth <strong>of</strong> MCF7 mammary xenografts as<br />

effectively as tamoxifen. Conclusions: MDV3100 blocked both DHT- and<br />

E2-mediated growth <strong>of</strong> breast cancer cells, whereas bicalutamide enhanced<br />

E2-mediated proliferation. MDV3100 may have unique therapeutic<br />

utility in patients with AR� breast cancer, regardless <strong>of</strong> ER status.<br />

Funding: DOD Breast Cancer Program Idea Award BC074403, Avon<br />

Foundation for Women, and University <strong>of</strong> Colorado Cancer Center pilot<br />

project funds to JKR.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


565 General Poster Session (Board #4H), Sat, 8:00 AM-12:00 PM<br />

Prognostic factor Ki67 for breast cancer patients in each subgroup.<br />

Presenting Author: Naoki Niikura, Tokai University School <strong>of</strong> Medicine,<br />

Isehara Kanagawa, Japan<br />

Background: Immunohistochemical (IHC) Ki67 has described it as a<br />

prognostic and predictive marker for breast cancer. The St. Gallen<br />

Consensus <strong>Meeting</strong> determined that Ki67 labeling index is chiefly important<br />

for distinguishing between “Luminal A” and “Luminal B (HER2<br />

negative)” subtypes and is a predictive marker for chemotherapeutic<br />

efficacy. However, the high and low cut<strong>of</strong>f points remain controversial. Our<br />

objective is to compare survival in patients with low, intermediate, and high<br />

Ki67 levels in each subgroup. Methods: We retrospectively identified all the<br />

patients in the Tokai University breast cancer database for whom IHC Ki67<br />

data were available between January 1, 2000, and December 31, 2010.<br />

Ki67 was defined as low if �10% Ki67 was detected, as Intermediate if<br />

10–20% Ki67 was detected, and as high if �20% Ki67 was detected. To<br />

assess Ki67 levels and survival outcomes, survival curves were calculated<br />

using the Kaplan–Meier method and compared using the log-rank test.<br />

Results: We identified 1331 primary breast cancer patients without<br />

metastasis, <strong>of</strong> whom 686 received neoadjuvant or adjuvant chemotherapy.<br />

Patients with high Ki67 had poorer relapse-free survival (RFS) than<br />

patients with intermediate (p � 0.009) and low Ki67 (p � 0.001). Patients<br />

with intermediate Ki67 had poorer RFS than patients with low Ki67 (p �<br />

0.001). In ER-positive cases (n � 1059), patients with high and intermediate<br />

Ki67 had poorer RFS than patients with low Ki67 (p � 0.001 and p �<br />

0.002, respectively). In HER2-positive and ER-negative cases (n � 103),<br />

patients with high Ki67 had poorer RFS than patients with low Ki67 (p �<br />

0.002). In triple-negative cases (n � 164), patients with high Ki67 tended<br />

to have poorer RFS than patients with low Ki67 (p � 0.064). Conclusions:<br />

Our data demonstrated that low, intermediate, and high Ki67 levels may be<br />

used to differentiate prognosis in ER-positive cancer patients as well as<br />

HER2-positive and triple-negative cancer patients.<br />

567 General Poster Session (Board #5B), Sat, 8:00 AM-12:00 PM<br />

Characterization <strong>of</strong> the overall survival benefit in ENCORE 301, a randomized<br />

placebo-controlled phase II study <strong>of</strong> exemestane with and without<br />

entinostat in ER� postmenopausal women with metastatic breast cancer.<br />

Presenting Author: Pamela Klein, PMK Consulting, San Mateo, CA<br />

Background: Histone deacetylase inhibitors (HDACi) prevent the emergence<br />

<strong>of</strong> drug tolerant clones and sensitize cells to a variety <strong>of</strong> anti-cancer<br />

therapies. We previously reported ENCORE 301, a randomized placebo<br />

controlled phase 2 study comparing exemestane with (EE) and without (EP)<br />

the HDACi entinostat met its primary endpoint <strong>of</strong> prolonging progression<br />

free survival (PFS) (4.3 months vs 2.3 months) and extending overall<br />

survival (OS) benefit (26.9 vs 19.8 months). In order to characterize and<br />

better understand the OS benefit, exploratory analyses were conducted.<br />

Methods: Hazard ratios (HR) for treatment were estimated from the Cox<br />

proportional hazards model, with EP serving as the reference treatment in<br />

the calculations. Inferential comparisons between treatment groups were<br />

made using the log-rank test. Results: Analysis <strong>of</strong> OS across multiple<br />

subsets <strong>of</strong> interest demonstrated a consistent benefit in EE group versus<br />

EP. Sensitivity analysis <strong>of</strong> baseline characteristics potentially impacting<br />

OS did not identify any factors that influenced the effect <strong>of</strong> EE on OS.<br />

Examination <strong>of</strong> treatments received during follow-up after discontinuation<br />

<strong>of</strong> EE and EP demonstrated balance between treatment group for patients<br />

receiving chemotherapy (48% EE: 44% EP), hormone therapy (37% EE:<br />

35% EP), bisphosphonates (2% EE; 0% EP), radiation (6% EE; 5%EP),<br />

and surgery (0% EE: 2% EP). Compared to EP, OS was longer in EE group<br />

for patients whose first subsequent treatment was a hormone therapy (EE<br />

median not reached vs EP median 20.5 months; HR 0.65 [95% CI 0.26,<br />

1.58]) or chemotherapy (EE median 26.9 months vs EP median 17.6<br />

months; HR 0.51 [95% CI 0.25, 1.04]). Updated PFS and OS data will<br />

also be presented along with correlation analysis <strong>of</strong> PFS and OS. Conclusions:<br />

Analyses <strong>of</strong> baseline characteristics, subsequent treatment and subsets <strong>of</strong><br />

prognostic factors in ENCORE 301 did not identify any contributing factors<br />

that account for the extended OS benefit in the EE treatment group. Long<br />

lasting effects <strong>of</strong> entinostat on progenitor cells, emergence <strong>of</strong> drug tolerant<br />

cells and or priming to subsequent therapies cannot be ruled out.<br />

Breast Cancer—HER2/ER<br />

23s<br />

566 General Poster Session (Board #5A), Sat, 8:00 AM-12:00 PM<br />

Prognostic significance <strong>of</strong> PI3K pathway (PI3Kp) dysregulation in metastatic<br />

breast cancer (MBC) patients (pts). Presenting Author: Alejandro Navarro,<br />

Medical Oncology Department, Vall d’Hebron University Hospital, Barcelona,<br />

Spain<br />

Background: PI3Kp dysregulation represents a potential target for therapies that<br />

are currently being tested in clinical trials. This observational retrospective study<br />

aims to evaluate the prognostic implications <strong>of</strong> PI3Kp dysregulation in MBC.<br />

Methods: MBC pts with PI3Kp status assessment from Sep09 to Sep11 were<br />

reviewed. PIK3CA mutation status analyzed in paraffin-embedded tissue by DxS<br />

PI3K Mutation Test Kit or Sequenom MassARRAY. PTEN status determined by<br />

IHC. PI3Kp status: (a) No dysregulation: PIK3CA wt and PTEN normal; (b)<br />

PI3Kp dysregulation: PIK3CA mutation (PIK3CAmut) or PTEN low (HScore�50).<br />

Results: 232 MBC pts screened, median age 49.8 (22.9-83.1) and median MBC<br />

lines 4 (1-15). Distribution: HR�/HER2- 99 (43%), HER2� 52 (22%), triple<br />

negative 35 (15%), unclassified 46 (20%). Sites <strong>of</strong> metastasis: visceral 173<br />

(75%), only skin 10 (4%), only bone 49 (21%). PIK3CA status assessed in 174<br />

pts, 53 (22.8%) bearing a mutation (21 exon9, 32 exon20). PTEN status<br />

assessed in 229 pts, PTEN low 61 (26.6%). PI3Kp dysregulation in 103/185<br />

pts (55.6%). Time to progression to first line MBC treatment (TTP) and overall<br />

survival after MBC diagnosis (OS) are shown. Disease free survival (DFS) and<br />

distant-disease free survival (DDFS) in pts initially diagnosed with early breast<br />

cancer (n�193) has also been calculated. Conclusions: These results suggest<br />

that PI3Kp dysregulation, either by PIK3CA mutation or PTEN low, does not<br />

seem to have impact on disease recurrence, response to first line MBC treatment<br />

or overall MBC survival.<br />

PIK3CA PTEN PI3Kp dysregulation<br />

Median (95%CI) WT Mut Normal Low No Yes<br />

n�174 n�229 n�185<br />

n�121 n�53 n�168 n�61 n�82 n�103<br />

TTP (m) 12.0<br />

11.8<br />

13.0<br />

11.5<br />

12.3<br />

11.8<br />

(10.3-13.8) (9.6-14.0) (11.4-14.6) (7.4-15.6) (10.0-14.5) (9.1-14.5)<br />

p�0.780 p�0.891 p�0.516<br />

OS (y) 5.5<br />

5.0<br />

5.6<br />

5.1<br />

5.5<br />

5.0<br />

(4.1-6.9) (3.3-6.7) (4.9-6.3) (3.0-7.3) (3.9-7.1) (3.5-6.5)<br />

p�0.882 p� 0.460 p�0.443<br />

n�158 n�189 n�167<br />

n�107 n�51 n�132 n�57 n�69 n�98<br />

DFS (m) 45.0<br />

36.6<br />

38.6<br />

49.6<br />

36.0<br />

46.0<br />

(35.6-54.4) (28.6-44.6) (32.2-45.1) (42.6-56.5) (24.3-47.7) (38.5-53.4)<br />

p� 0.476 p�0.071 p�0.163<br />

DDFS (m) 51.0<br />

44.6<br />

46.3<br />

57.1<br />

40.0<br />

49.6<br />

(43.1-58.8) (32.5-56.7) (35.8-56.8) (44.3-69.9) (22.5-57.5) (39.7-59.4)<br />

p� 0.505 p� 0.289 p� 0.254<br />

568^ General Poster Session (Board #5C), Sat, 8:00 AM-12:00 PM<br />

Prospective study <strong>of</strong> the impact <strong>of</strong> using the 21-gene recurrence score<br />

assay on clinical decision making in women with estrogen receptorpositive,<br />

HER2-negative, early-stage breast cancer in France. Presenting<br />

Author: Joseph Gligorov, APHP Tenon APREC, CancerEst, University Paris<br />

VI, Paris, France<br />

Background: The 21-gene Oncotype DX Recurrence Score (RS) is a<br />

validated assay to help inform the appropriate treatment <strong>of</strong> estrogen<br />

receptor-positive (ER�), early stage breast cancer in the adjuvant setting.<br />

Treatment traditions regarding choice <strong>of</strong> adjuvant treatment vary significantly<br />

in different countries. This prospective multicenter study is the first<br />

to assess the impact <strong>of</strong> using the Oncotype DX assay in the French clinical<br />

setting. Methods: A total <strong>of</strong> 100 consecutive patients with ER�, HER2negative,<br />

node negative or pN1 (mi) breast cancer were enrolled. Overall<br />

treatment recommendation change, change from chemoendocrine to<br />

endocrine alone and change from endocrine alone to chemoendocrine<br />

treatment were recorded. Medical oncologists completed questionnaires<br />

regarding their confidence in their recommendation before and after<br />

knowing the patient’s RS. A preliminary analysis was conducted on the first<br />

92 evaluable patients with data available at the time <strong>of</strong> abstract submission.<br />

Final data will be presented at the meeting. Results: Prior to Oncotype<br />

DX 49% <strong>of</strong> patients were recommended chemoendocrine treatment and<br />

51% endocrine treatment alone. After having the RS, 26% were recommended<br />

chemoendocrine treatment and 74% endocrine treatment alone.<br />

The overall reduction in chemotherapy recommendation from 49% to 26%<br />

was significant (p�0.001). Of patients originally recommended chemoendocrine<br />

treatment, 58% were changed to endocrine treatment alone after<br />

having the RS. Of patients originally recommended endocrine treatment,<br />

11% were changed to chemoendocrine treatment after receiving the RS.<br />

There was a significant improvement in physician confidence in treatment<br />

recommendations (p�0.002) when using Oncotype DX. Conclusions: These<br />

are the first prospective data regarding the impact <strong>of</strong> using Oncotype DX in<br />

France. Using Oncotype DX was associated with a significant change in<br />

treatment decisions and an overall reduction in chemotherapy use. The<br />

data are consistent with those presented from Germany, Spain, the UK and<br />

the US.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


24s Breast Cancer—HER2/ER<br />

569 General Poster Session (Board #5D), Sat, 8:00 AM-12:00 PM<br />

Evaluation <strong>of</strong> Oncotype DX testing and subsequent patterns <strong>of</strong> care in<br />

patients (pts) with early-stage breast cancer (ESBC). Presenting Author:<br />

Meghan Fitzgerald, Cardinal Health, Specialty Solutions, Dublin, OH<br />

Background: Cardinal Health Specialty Solutions (CHSS) partners with<br />

payers to manage clinical care pathways in oncology and rheumatology.<br />

Observations from one such pathway with CareFirst BlueCross BlueShield<br />

revealed that pts with estrogen receptor-positive ESBC who underwent<br />

Oncotype DX testing and were found to have high-risk Recurrence Scores<br />

(RS) received less chemotherapy than expected. To better understand<br />

physician behavior as it relates to Oncotype DX testing, CHSS examined<br />

patterns <strong>of</strong> Oncotype DX use in a large private practice oncology group with<br />

a robust electronic medical record (EMR), Georgia Cancer Specialists<br />

(GCS). We aimed to determine the RS risk distribution among pts who<br />

received Oncotype DX testing and assess the patterns <strong>of</strong> care that followed.<br />

Methods: Using GCS EMR data from 2009 to 2011, the number <strong>of</strong> pts who<br />

were eligible to undergo Oncotype DX testing was compared to the number<br />

<strong>of</strong> pts who underwent testing. Pts were considered eligible for testing if they<br />

had node-negative, estrogen receptor-positive, HER2-negative ESBC. We<br />

determined the number <strong>of</strong> pts with a RS value in the low- (RS �18),<br />

intermediate- (RS 18-30), and high-risk (RS �31) groups along with the<br />

number <strong>of</strong> pts who subsequently received chemotherapy in each category.<br />

Results: Of the 1908 patients identified with ESBC, 788 were eligible for<br />

Oncotype DX testing and 288 (37%) underwent testing. Fifty percent <strong>of</strong> pts<br />

were in the low-, 34% in intermediate-, and 16% in the high-risk groups.<br />

Six percent <strong>of</strong> low-, 41% <strong>of</strong> intermediate-, and 83% <strong>of</strong> high-risk pts<br />

received chemotherapy. Seventeen percent <strong>of</strong> pts in the high-risk group did<br />

not receive chemotherapy. Conclusions: The use <strong>of</strong> chemotherapy in the<br />

low- and intermediate-risk groups was as expected based on adjuvant<br />

chemotherapy guidelines; however, an under utilization <strong>of</strong> chemotherapy<br />

was found in high-risk pts. There also appears to be a noteworthy underuse<br />

<strong>of</strong> Oncotype DX testing in eligible pts. Further data analysis is needed to<br />

understand the low use <strong>of</strong> Oncotype DX testing in eligible pts and to explain<br />

the lower than expected use <strong>of</strong> chemotherapy in high-risk pts.<br />

571 General Poster Session (Board #5F), Sat, 8:00 AM-12:00 PM<br />

Evaluation <strong>of</strong> variables that may impact chemotherapy (CT) administration<br />

after determination <strong>of</strong> Oncotype DX (ODX) recurrence score (RS). Presenting<br />

Author: Kristina E. Bowen, Georgia Cancer Specialists PC, Atlanta, GA<br />

Background: Cardinal Health Specialty Solutions (CHSS) partners with<br />

payers to manage clinical care pathways in oncology and rheumatology.<br />

Observations from one such pathway with CareFirst BlueCross Blue Shield<br />

revealed lower than expected use <strong>of</strong> CT in patients (pts) with estrogen<br />

receptor-positive early-stage breast cancer (ESBC) at high-risk for recurrence<br />

by ODX RS. To better understand physician behavior, CHSS examined<br />

patterns <strong>of</strong> care in a large private practice oncology group with a robust<br />

EMR, Georgia Cancer Specialists (GCS). Pt characteristics and physicianprescribing<br />

patterns were analyzed to determine variables that most<br />

impacted the physician’s decision to treat high-risk pts with CT. Methods:<br />

Using the GCS EMR from 2009 to 2011, we retrospectively identified pts<br />

with ESBC (stage I-II, node-negative, estrogen receptor-positive, HER2negative)<br />

who underwent ODX testing. We determined the number <strong>of</strong> pts<br />

with a RS value in the low- (RS �18), intermediate- (RS 18-30), and<br />

high-risk (RS �31) groups along with the number <strong>of</strong> pts who subsequently<br />

received CT in each category. The use <strong>of</strong> CT in high-risk pts was analyzed by<br />

pt age, tumor size, tumor grade, and physician prescribing patterns.<br />

Results: Of the 1908 pts identified with ESBC, 788 were eligible for ODX<br />

testing and 288 (37%) underwent testing. Fifty percent <strong>of</strong> pts were in the<br />

low-, 34% in intermediate-, and 16% in the high-risk groups. Six percent <strong>of</strong><br />

low-, 41% <strong>of</strong> intermediate-, and 83% <strong>of</strong> high-risk pts received CT. In<br />

high-risk pts analyzed by age, CT was used in 100% <strong>of</strong> pts � 45 years (y),<br />

86% in pts 46-55 y, 86% in pts 56-65 y, and 25% in pts �66 y. CT use in<br />

high-risk pts was 82% and 86% in pts with tumor sizes �2 cm and 2-5 cm,<br />

respectively; and, 100%, 60%, and 91% in pts with grade 1, 2, or 3<br />

tumors, respectively. Of the 41 physicians who were evaluated, 10 (24%)<br />

used ODX �15% <strong>of</strong> the time, 13 (32%) used ODX 15-40%, 10 used (24%)<br />

used ODX 41-60%, and 8 (20%) used ODX � 60% <strong>of</strong> the time. There were<br />

no significant differences in patterns <strong>of</strong> CT use. Conclusions: The less than<br />

expected use <strong>of</strong> CT in high-risk patients appears to be related to physician<br />

preference and pt age. Tumor size and grade did not appear to influence<br />

choice <strong>of</strong> therapy.<br />

570 General Poster Session (Board #5E), Sat, 8:00 AM-12:00 PM<br />

Treatment pattern by hormone receptors and HER2 status in patients with<br />

metastatic breast cancer in the United Kingdom, Germany, France, Spain,<br />

and Italy (EU-5): Results from a physician survey. Presenting Author: Mitch<br />

DeKoven, IMS Health, Alexandria, VA<br />

Background: The differences in country-specific treatment patterns across<br />

Europe for Stage IV metastatic breast cancer (mBC) patients have not been<br />

extensively studied. This study compared treatment choices, by biomarker<br />

status, between various lines <strong>of</strong> therapy (LOT) in clinical practice in the<br />

EU-5 countries among newly diagnosed stage IV mBC patients. Methods:<br />

The IMS LifeLink Oncology Analyzer (OA) database, based upon practicing<br />

oncologist surveys, was used to identify mBC patients aged �21 and<br />

surveyed between July 2008 – June 2010. The database encompasses over<br />

100,000 unique patients per year, treated by nearly 3,000 physicians,<br />

including nearly 60,000 patients and 800 physicians in the EU5. Results:<br />

A total <strong>of</strong> 152,311 mBC patients were included in the study. In Italy,<br />

30.8% <strong>of</strong> HER2�/HR� patients received chemotherapy following mBC<br />

diagnosis, as compared to only 8.6% in France. The majority <strong>of</strong> these<br />

patients in France received chemotherapy plus a HER2 targeted therapy<br />

(58.2%) as their first treatment. For HER2�/ HR- patients, chemotherapy<br />

plus a HER2 targeted therapy was provided to the majority <strong>of</strong> the patients in<br />

France (70.8%), with the UK providing this regimen to the fewest patients<br />

with this biomarker status (44.9%). Monotherapy hormonal regimens were<br />

given as a first LOT for HER2-/HR� patients (UK 64.7%, Spain 52.8%).<br />

Triple negative patients received chemotherapy (UK 84.1% as compared to<br />

France 57.3%) or chemotherapy plus bevacizumab (France 36.6% as<br />

compared to UK 0.9%) as a first treatment. Conclusions: Thisstudy<br />

confirmed that chemotherapy combined with HER2-targeted therapy was<br />

the most frequent initial treatment option for HER2� patients, while the<br />

vast majority <strong>of</strong> ER/PR� patients were initially treated by hormonal<br />

therapy, suggesting that a personalized medicine approach has been<br />

accepted in the EU-5. However, the use <strong>of</strong> HER2-targeted therapy and<br />

bevacizumab greatly varied; while they were most frequently used in<br />

France, they were least frequently opted for in the UK. Also, fewer<br />

treatment options existed for triple negative patients and patients with<br />

HER2� disease following trastuzumab treatment.<br />

572 General Poster Session (Board #5G), Sat, 8:00 AM-12:00 PM<br />

Synchronous bilateral breast cancer (SBBC): Concordance <strong>of</strong> receptor<br />

status between right and left breast. Presenting Author: Palak Desai, Rush<br />

University Medical Center, Chicago, IL<br />

Background: This analysis was prompted by a patient who presented with<br />

SBBC who had and ER�, HER2- cancer in one breast and an ER-, HER2�<br />

cancer in the other. Since both breasts have the same genetic background<br />

and environmental exposures, concordance should be virtually identical<br />

unless other factors are at play. Methods: SBBC was defined as bilateral<br />

cancer diagnosed concurrently or within 6 months <strong>of</strong> each other. Cases<br />

were identified retrospectively from patients seen at Rush University<br />

Medical Center. This analysis was limited to invasive SBCC (ISBBC).<br />

Estrogen receptor (ER) and Her2/neu (HER2) status were assessed according<br />

current ASCO/CAP guidelines. Correlation between ER and HER2<br />

status was determined using � statistic. Results: From January 1998 to<br />

December 2012, 33 cases <strong>of</strong> ISBBC were diagnosed. In 65% <strong>of</strong> cases<br />

synchronous disease was diagnosed concurrently. The average age at<br />

diagnosis was 56. The majority were multiparous and 79% post menopausal.<br />

About half <strong>of</strong> the patients (55%) had at least one first degree<br />

relative with a history <strong>of</strong> breast or ovarian cancer and 9% <strong>of</strong> the cohort had a<br />

deleterious mutation in BRCA1 or BRCA2. Infiltrating ductal was most<br />

common (74%) followed by infiltrating lobular (21%). 53% <strong>of</strong> breasts<br />

specimens also contained ductal carcinoma in situ. 68% <strong>of</strong> tumors were ER<br />

positive and 8% were HER2 positive. Concordance in ER status was seen in<br />

73% <strong>of</strong> cases (kappa coefficient 0.14) and in HER2 status was 64%<br />

(Kappa coefficient 0.35). Conclusions: Despite identical hereditary and<br />

environmental exposure, some patients develop ISBBC cancers with<br />

discordant receptor status. While this is the exception, it suggests that<br />

other factors influence important tumor characteristics.<br />

R breast � (n) R breast – (n) Total<br />

ER status<br />

L breast � 22 4 26<br />

L breast - 5 2 7<br />

Total 27 6 33<br />

HER2 status<br />

L breast � 1 2 3<br />

L breast - 1 28 29<br />

Total 2 30 32<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


573 General Poster Session (Board #5H), Sat, 8:00 AM-12:00 PM<br />

In situ quantitative measurement <strong>of</strong> mRNA to predict response to trastuzumab<br />

in a cohort <strong>of</strong> metastatic breast cancer patients. Presenting Author:<br />

Maria Vassilakopoulou, Yale University, New Haven, CT<br />

Background: Trastuzumab therapy is currently selected based on assessment<br />

<strong>of</strong> HER2 by either immunohistochemistry (IHC) for protein overexpression<br />

or fluorescence in situ hybridization (FISH) to detect gene<br />

amplification. We sought to determine the predictive value <strong>of</strong> in situ<br />

quantitative measurement <strong>of</strong> mRNA on a cohort <strong>of</strong> trastuzumab-treated<br />

metastatic breast cancer patients. Methods: A tissue microarray composed<br />

<strong>of</strong> ninety metastatic breast cancers treated with various chemotherapy<br />

regimens combined with trastuzumab was constructed. HER2 intracellular<br />

domain (ICD), HER2 extracellular domain (ECD) and HER2 mRNA were<br />

assessed using the AQUA method for quantitative immun<strong>of</strong>luorescence.<br />

For HER2 protein evaluation CB11was used to measure ICD and SP3 to<br />

measure ECD <strong>of</strong> the HER2 receptor. In addition, HER2 mRNA status was<br />

assessed using the RNAscope assay ERRB2 probe according to manufacturer’s<br />

protocol modified for detection with Cy-5 Tyramide. Cytokeratin was<br />

used in order to create the tumor mask and signal was quantified within the<br />

mask. Primary endpoints included time to progression (TTP) from trastuzumab<br />

initiation and overall survival (OS) times. Statistical analysis was<br />

performed using Kaplan-Meier analysis and the Cox proportional hazard<br />

model. Results: HER mRNA was tightly correlated with ICD HER2, as<br />

measured by CB11 (r2�0.35), but not with ECD HER2 as measured by<br />

SP3 (r2�0.14). Both ICD HER2 and HER2 mRNA were predictive <strong>of</strong><br />

response to trastuzumab, but ECD was not. Multivariate analysis including<br />

age, histological grade and hormone receptor status shows the ICD to be<br />

predictive <strong>of</strong> TTP (p�0.0377). HER2 mRNA levels were significant for<br />

prediction <strong>of</strong> TTP (p�0.0344) in multivariate analysis including age and<br />

histological grade. Conclusions: The expression <strong>of</strong> ICD, as detected by<br />

CB11 and HER2 mRNA levels were significantly associated with TTP in this<br />

trastuzumab-treated metastatic cohort. The ECD, as detected by SP3 is<br />

neither predictive <strong>of</strong> response, nor tightly correlated with HER2 mRNA. In<br />

situ assessment <strong>of</strong> HER2 mRNA has the potential to identify breast cancer<br />

patients who derive benefit from trastuzumab treatment.<br />

575 General Poster Session (Board #6B), Sat, 8:00 AM-12:00 PM<br />

Fertility preservation options and the young breast cancer patient: A survey.<br />

Presenting Author: Manuela Jacobsen Junqueira, Memorial Sloan-<br />

Kettering Cancer Center, New York, NY<br />

Background: Approximately 15% <strong>of</strong> breast cancers (BC) are diagnosed in<br />

reproductive aged women. Management <strong>of</strong> the disease in this age group<br />

frequently includes chemotherapy and hormonal therapy, which can both<br />

affect fertility. Considering that age at first delivery has been steadily<br />

increasing, young women may face BC before completion <strong>of</strong> childbearing.<br />

Methods: In this prospective study, women referred to our institution for<br />

surgical treatment <strong>of</strong> BC were asked, before their first visit, to fill out a<br />

questionnaire regarding their reproductive history and fertility preservation<br />

knowledge. Eligible patients included women between the ages <strong>of</strong> 18 and<br />

45, with a newly diagnosed BC, who had not yet started treatment. Results:<br />

Sixty women were eligible with a median age <strong>of</strong> 40 (range 20-45). 98% <strong>of</strong><br />

responders (59 out <strong>of</strong> 60) had been diagnosed within the previous 2<br />

months. 78% (47/60) had a college or post-graduate degree. 80% (48/60)<br />

had been pregnant before, while 86.5% (45/52) reported having had<br />

children. 81% (47/58) were premenopausal, and only 3 patients reported<br />

not having had periods for more than 1 year. 50% <strong>of</strong> responders (30/60)<br />

declared no interest in future childbearing, 25% were definitely interested,<br />

and 25% were undecided. However, only 9% (5/57) reported having<br />

received information on fertility preservation options before the survey.<br />

Women who have been pregnant were significantly less likely to consider<br />

fertility preservation options prior to treatment (egg/embryo/ovarian tissue<br />

cryopreservation [6% vs. 50%, p�0.001]), or after treatment (egg/embryo<br />

donation, surrogacy or adoption [6% vs. 58%, p�0.0001]). Conclusions:<br />

This pilot study was designed to gather information on reproductive health<br />

<strong>of</strong> newly diagnosed young BC patients and to assess their willingness to<br />

consider various fertility preservation options before or after treatment. Our<br />

study population consisted <strong>of</strong> mostly women who had been pregnant and<br />

had children. We found that 50% <strong>of</strong> the women were unsure or wanted<br />

future children. Yet, only 9% had received information on fertility options<br />

at diagnosis. This pilot study highlights the need for education and/or<br />

intervention in fertility preservation options for young women with breast<br />

cancer.<br />

Breast Cancer—HER2/ER<br />

25s<br />

574 General Poster Session (Board #6A), Sat, 8:00 AM-12:00 PM<br />

ER� mRNA expression in ER�-negative and triple-negative breast cancers.<br />

Presenting Author: Young Choi, Yale University School <strong>of</strong> Medicine,<br />

Bridgeport, CT<br />

Background: ER� is the main prognostic and therapeutic marker in breast<br />

cancer (BC). About 30% <strong>of</strong> BC cases are negative for ER (ER�-) and do not<br />

benefit from antiestrogen therapy (TAM). We aim to study ER-beta (ER�)<br />

expression in ER�- and triple negative (TN) cancers to explore alternate<br />

pathway <strong>of</strong> treatment in this cohort. Methods: We studied 67 ER�- BC cases<br />

including 44 TN together with 74 ER� �BC cases obtained from patients<br />

aged 29 to 97 years old between 2003 and 2010. The histology included<br />

110 intraductal, 12 medullary and 19 other types. 78 cases were grade 3,<br />

52 were grade 2, and 11 were grade 1. RNA was extracted from FFPE and<br />

mRNA levels <strong>of</strong> ER� is<strong>of</strong>orm and ER� were determined by real-time<br />

quantitative reverse transcription PCR. IHC stains were done on TMA the<br />

sections for ER�, PR, Her-2, Ki-67, CK5/6 and Cyclin D1. Results: ER�<br />

is<strong>of</strong>orms were highly expressed in ER�-, TN, basal-like and HER2 type BC<br />

cases. ER�2 was the major ER� variant expressed. Ki-67 proliferating cells<br />

(�20% nuclear staining) were mostly in ER�- rather than ER�� cases<br />

(69.0% vs. 31.0%) as were cyclin D1- cells (82.2% vs. 17.8%). On the<br />

other hand, in ER�� BC, ER� mRNA expression was consistently high and<br />

upregulated, and ER�, low and down regulated, and the ratio <strong>of</strong> ER�� to<br />

ER�� ranged from 3 to 100. ER�1, 2 and 5 were co-expressed with ER� in<br />

56%, 63%, and 30% <strong>of</strong> cases, respectively. Overall, ER� mRNA levels did<br />

not show any significant correlation with age, tumor size, lymph node status<br />

and histological grades. Conclusions: ER�-dependent proliferating tumor<br />

cells may render them more sensitive to TAM, and increase the effectiveness<br />

<strong>of</strong> TAM and its metabolites in ER�- and TN cases. Increased overall<br />

survival after adjuvant TAM ER�-BC may be directly related to ER�<br />

over-expression. ER� is<strong>of</strong>orm is potential selective therapeutic target in a<br />

sub-cohort <strong>of</strong> ER�- BC. Additionally, when ER� and ER� are co-expressed,<br />

ER� appears to play a distinct role from its action in ER�- BC.<br />

Types (# <strong>of</strong> cases) ER�1* ER�2* ER�5*<br />

ER�- (67) 39 42 15<br />

TN (44) 41 46 25<br />

HER2� (38) 66 82 37<br />

HER2 type (19) 74 85 47<br />

Basal-like type (11) 51 64 36<br />

Ki-67� /ER�- (27) 55 70 37<br />

Grade 3 (78) 41 46 31<br />

Grade 2 (52) 39 54 29<br />

* % mRNA expression.<br />

576 General Poster Session (Board #6C), Sat, 8:00 AM-12:00 PM<br />

Estrogen receptor (ER) signaling in normal, BRCA (B) 1 and B2 mutation<br />

associated, and ER-positive breast cancer (BC) mammary cells. Presenting<br />

Author: Elgene Lim, Dana-Farber Cancer Institute, Boston, MA<br />

Background: ER is expressed in �70% <strong>of</strong> sporadic BC and activates genes<br />

driving cell proliferation and tumorigenesis. B1 and B2 mutation associated<br />

BC have predominant triple negative and ER� phenotypes respectively,<br />

yet hormonal manipulation with oophorectomy reduces BC risk in<br />

both B1 and B2 carriers. We have previously performed genome-wide<br />

analysis <strong>of</strong> ER binding sites in MCF-7 BC cells, and identified distinct<br />

mechanisms <strong>of</strong> ER signaling. Here, we seek to elucidate differences in ER<br />

signaling between non-malignant (normal, B1 and B2) and ER� BC<br />

mammary cells. Methods: Breast tissue were obtained from patients<br />

undergoing reduction mammoplasties (normal), prophylactic mastectomies<br />

(B1 and B2 carriers) and surgical excision <strong>of</strong> ER� BC. After<br />

dissociation into a single-cell suspension, ER� EpCAM�CD49f- mature<br />

luminal (ML) and EpCAM�CD49f� luminal progenitor (LP) subpopulations<br />

were obtained by flow cytometry. Following estrogen stimulation, RNA<br />

was extracted for gene microarray analysis. ER chromatin immunoprecipitation<br />

and DNA sequencing (ChIP-seq) was performed and DNA libraries<br />

prepared and sequenced. Results: Triplicates <strong>of</strong>normal, prophylactic B1<br />

and B2, and ER� BC tissues were analyzed.Gene ontology analysis<br />

indicates different gene expression signatures between non-malignant<br />

tissue and MCF-7 BC cells following estrogen stimulation. Genes that<br />

promotes cell cycling and proliferation were downregulated in nonmalignant<br />

tissue, but were upregulated in BC cells (P � 10-5 ). Hierarchical<br />

clustering <strong>of</strong> the gene microarray by mutation status revealed distinct LP<br />

gene signatures in normal, B1 and B2 breast tissue, whilst the ML<br />

signatures were largely indistinguishable between the three tissue types.<br />

Conclusions: There are contrasting differences in ER signaling between<br />

normal mammary and BC cells, with estrogen having anti-proliferative<br />

effects in normal luminal cells compared to pro-proliferative effects in BC.<br />

Our data suggest that alterations in ER signaling play a major role in breast<br />

tumorigenesis. Differences in the LP population between B1 and B2 breast<br />

tissue may account for their distinct BC phenotypes.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


26s Breast Cancer—HER2/ER<br />

577 General Poster Session (Board #6D), Sat, 8:00 AM-12:00 PM<br />

Molecular subtyping using MammaPrint and BluePrint as an outcome<br />

predictor in 180 U.S. breast cancer (BC) patients. Presenting Author:<br />

Lisette Stork-Sloots, Agendia, Amsterdam, Netherlands<br />

Background: Combined use <strong>of</strong> MammaPrint and a molecular subtyping<br />

pr<strong>of</strong>ile (BluePrint) identifies disease subgroups with marked differences in<br />

long-term outcome and response to neo-adjuvant therapy (Glück<br />

SABCS2011). The aim <strong>of</strong> this study was to evaluate the prognostic value <strong>of</strong><br />

Molecular Subtyping using MammaPrint and BluePrint in women with early<br />

stage BC treated at US Institutions following National Comprehensive<br />

Cancer Network (NCCN) standard guidelines. Methods: Frozen tumor<br />

samples from 180 BC patients (TI-III, N0-Ib) median age 57 years at<br />

diagnosis (range 28-89) were suitable for hybridization on full genome<br />

array. MammaPrint and BluePrint Molecular Subtypes were determined<br />

and survival for Luminal A (MammaPrint Low Risk), Luminal B (MammaPrint<br />

High Risk), HER2-type and Basal-type patients was assessed.<br />

Patients were treated either with breast conserving therapy or mastectomy<br />

with axillary lymph node dissection between 1992 and 2005. The median<br />

follow-up is 12.7 years. 71% was ER positive and 20% Her2 positive by<br />

IHC/FISH. 58% received adjuvant endocrine therapy (ET) (excluding 13<br />

patients unknown treatment), 64% received adjuvant chemotherapy (CT)<br />

(excluding 12 patients unknown treatment) and 33% received both.<br />

Results: 61 (34%) Patients with MammaPrint Low Risk/Luminal-type<br />

(Luminal A) showed 5-year DFS <strong>of</strong> 97% (34% received CT and 69% ET)<br />

and 50 (28%) patients with MammaPrint High Risk/Luminal-type (Luminal<br />

B) had a 5-year DFS <strong>of</strong> 98% (60% received CT and 68% ET). Patients with<br />

BluePrint Basal-type tumors (46 (26%)) had a 5-year DFS <strong>of</strong> 80% (78%<br />

received CT); HER2-type (23 (13%)) had a 5-year DFS <strong>of</strong> 78% (87%<br />

received CT without HER-2 targeted therapy). Conclusions: In this retrospective<br />

study evaluating 180 US patients with early BC treated according to<br />

standard guidelines we showed how combining BluePrint with MammaPrint<br />

can detect molecularly defined subgroups <strong>of</strong> patients who are at<br />

high risk <strong>of</strong> recurrence (HER2 and Basal-type). Furthermore, we confirmed<br />

that molecularly defined Luminal type disease is associated with excellent<br />

disease-free survival. MammaPrint and BluePrint molecular and prognostic<br />

stratification should be prospectively evaluated for therapeutic selection.<br />

580 General Poster Session (Board #6G), Sat, 8:00 AM-12:00 PM<br />

Breast cancer in Sub-Saharan Africa: 1,000 patients with primary breast<br />

cancer in Addis Ababa followed for up to 5 years. Presenting Author: Eva<br />

Johanna Kantelhardt, Department <strong>of</strong> Gynecology, Martin Luther University,<br />

Halle/Wittenberg, Germany<br />

Background: There is little information on breast cancer (BC) patients (pts)<br />

receiving standardized treatment in Sub-Saharan Africa. This study evaluates<br />

pts presenting 2005-10 at the University Radiotherapy Center in<br />

Addis Ababa, the only institution in the country <strong>of</strong>fering standardized<br />

radiotherapy, systemic therapy and free endocrine treatment (ET) during<br />

that time. Methods: All pts with histologically verified BC were included.<br />

Ethical approval was obtained. Axios/AstraZenaca provided free ET. Therefore,<br />

the majority <strong>of</strong> pts underwent regular follow-up (FUP). We analyzed<br />

survival at 18 months by means <strong>of</strong> Kaplan-Meier survival analysis. We<br />

assumed right-censoring to be unrelated to the risk <strong>of</strong> metastasis. In a worst<br />

case sensitivity analysis, we considered all censored pts developing<br />

metastasis. Results: Pts with primary diagnosis between July 1st, 2005 and<br />

December 31st, 2010 were included (n�1303). The majority <strong>of</strong> pts were<br />

female (95.2%), most (52.3%) postmenopausal. Mean age was 44.1yrs<br />

(20-88yrs). Stages 1-4 presented in 3/19/53/25% respectively (36%<br />

unknown). Grade 2 tumors were seen in 434 out <strong>of</strong> 574 pts (58%).<br />

Estrogen receptor was pos. in 251 out <strong>of</strong> 381 pts (66%). Most M0-pts<br />

(n�942) underwent surgery (84%), received chemotherapy (59%), and<br />

received ET (63%). Median FUP was 18.4 months, 186 events (metastases)<br />

occurred. Metastasis-free survival (MFS) was 86%. Worst case<br />

analysis on censored observations revealed that MFS declined down to<br />

52%. Pts with early stage 1/2 showed a better MFS than pts with stage 3<br />

disease (93 to 77%). Surgery (no surgery 78% vs surgery 87%) and ET<br />

(79% vs 89%) improved MFS. The 5-year MFS for stage 1/2 was 78% and<br />

stage 3 was 38%. Conclusions: To our knowledge this is the first presentation<br />

<strong>of</strong> clinical features in 1300 pts with BC in Sub-Saharan Africa. Most<br />

pts in Addis Ababa (AA) are �45yrs and present at stage 3/4. Differences to<br />

5-year MFS from Europe stage 1/2 around 90% (AA 78%) and stage 3<br />

around 70% (AA 38%) are smaller in pts treated with surgery and ET. This<br />

data is consistent with overall survival in a treated pt cohort from Uganda<br />

stage 1/2: 74% and stage 3/4: 39% (n�285) (Gakwaya Brit J Cancer<br />

2008). Policies should focus on earlier presentation and access to care.<br />

579 General Poster Session (Board #6F), Sat, 8:00 AM-12:00 PM<br />

Complementary role <strong>of</strong> Ki67 index and 70-gene signature (MammaPrint)<br />

high-risk patients in the St Gallen risk group with uncertain chemotherapy<br />

suggestion. Presenting Author: Paolo Pronzato, Department <strong>of</strong> Medical<br />

Oncology, National Institute for Cancer Research, Genova, Italy<br />

Background: St Gallen (SG) Consensus Panel on adjuvant treatment<br />

recommends the use <strong>of</strong> proliferation markers and multigene assays when<br />

choosing the appropriate treatment in addition to traditional parameters.<br />

Ki67 predictive/prognostic value is widely accepted; we tested its role in<br />

increasing the accuracy <strong>of</strong> risk prediction among Mammaprint (MP)<br />

/High(HR) or /Low Risk (LR) breast cancer patients with a SG risk uncertain<br />

for chemotherapy selection. Methods: MP was determined (courtesy <strong>of</strong><br />

Agendia, Amsterdam) in 3 Italian Hospitals on 305 consecutive samples <strong>of</strong><br />

breast cancer patients. We focused on SG “Intermediate Risk” Group<br />

(HER2 neg, no VI and: ER�10�50%, or G2 or Ki67 �15�30% or N1a or<br />

T�2�5cm), where the indication for adjuvant CHT still remains uncertain.<br />

In MP/HR cases, a “low” Ki67 value (�15%) was used to eliminate CHT<br />

suggestion and viceversa an “intermediate” Ki67 value (�15�30%) in<br />

MP/LR cases. Results: Overall, 72/305 pts (26.6%) were in SG intermediate<br />

risk group with a median age <strong>of</strong> 64 years (26-98). Ki67 was non<br />

available in 4.1% <strong>of</strong> cases, MP rejected in 16.6%, HR was detected in 39<br />

(54.2%), whereas LR in 21 (29.2%). Ki67 resulted low in 23 MP/HR cases<br />

(59%), intermediate in 6 MP/LR cases (28.5%) and in 3 out 12<br />

MP/Rejected cases (25%). The overall concordance between MP and Ki67<br />

was 28/57 (49.1%), MP rejected excluded. Overall MP suggested 39/60<br />

CHT (65%), Ki67 29/69 (42%). Conclusions: The risk <strong>of</strong> relapse is a<br />

continous variable and MP evaluates it in a dichotomy (low vs high) wich<br />

could decrease the accuracy <strong>of</strong> the test. In the selected SG Group, the<br />

average risk <strong>of</strong> 5 yr relapse is around 20% (EBCTCG, Lancet 2011) and MP<br />

HR cases in our experience account for 55%. A low Ki67 value could help<br />

avoid more than 20% <strong>of</strong> cytotoxic treatments suggested by MP. In the same<br />

way, according to an ER value �50% (2 cases), an intermediate Ki67 value<br />

could suggest a more aggressive treatment in a further 13% <strong>of</strong> cases MP LR<br />

(6/21) or Rejected (3/12). MP probably overestimates the risk in 20% <strong>of</strong><br />

cases and underestimates it in further 20%. Ki67 could be usefull for a<br />

more personalized treatment in patients where the indication for adjuvant<br />

chemotherapy added to endocrine treatment is uncertain.<br />

581 General Poster Session (Board #6H), Sat, 8:00 AM-12:00 PM<br />

Assessment <strong>of</strong> method <strong>of</strong> breast cancer detection and Oncotype DX<br />

recurrence score. Presenting Author: Ciara Marie Kelly, Mater Misericordiae<br />

University Hospital, Dublin, Ireland<br />

Background: Data indicate that screen-detected (SD) cancers show more<br />

favourable pathological characteristics and have a better prognosis compared<br />

to symptomatic or interval cancers (SY). These findings can be partly<br />

explained by length time and lead time biases. OncotypeDX recurrence<br />

score (RS) provides prognostic information independent <strong>of</strong> traditional<br />

clinicopathological variables and provides a robust measure <strong>of</strong> tumour<br />

proliferation. The objective <strong>of</strong> this study was to determine whether the RS<br />

would vary depending on the method <strong>of</strong> detection (MOD) in patients with<br />

lymph node-negative, ER-positive, HER-2 negative breast cancer (BC).<br />

Methods: We included patients with ER-positive, lymph node-negative and<br />

HER2-negative BC who underwent OncotypeDX testing as part <strong>of</strong> routine<br />

care or a clinical trial. We retrospectively reviewed patient charts and<br />

extracted information on MOD (SD versus SY), clinicopathological variables<br />

and RS. Chi-squared test was used to test for differences between<br />

categorical variables and MOD. Analysis <strong>of</strong> variance (ANOVA) was used to<br />

investigate the relationship between RS and MOD and clinicopathological<br />

variables. Results: We identified 596 patients from 4 academic hospitals<br />

(Mater University Hospital n� 98 (16%), St Vincent’s Hospital n� 161<br />

(27%), Waterford Regional Hospital n� 27 (4%) and University <strong>of</strong> Texas,<br />

MD Anderson Cancer Center n� 310 (52%)). There were 359 (60 %) SDdetected<br />

and 237 (40%) SY-detected breast cancers. Clinicopathological<br />

variables were similar for age at diagnosis (56 versus 52 years), tumour size<br />

(16mm versus 18mm), grade (63% versus 61% grade 2; 15% versus 15%<br />

grade I; 21% versus 24% grade 3) and LVI (18% versus 18%) for SY<br />

compared to SD, respectively. The number <strong>of</strong> patients in each group with<br />

low (51% versus 50%), intermediate (41% each) or high (8% versus 9%)<br />

RS was similar in the SD compared to SY groups, respectively. There was a<br />

statistically significant association between RS and tumour grade<br />

(P��0.001) and lymphovascular invasion (P��0.001). We did not<br />

observe a statistically significant association between RS and MOD<br />

(P�0.086). Conclusions: In patients with clinically intermediate breast<br />

cancer sent for OncotypeDX testing we found no association between RS<br />

and MOD.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


582 General Poster Session (Board #7A), Sat, 8:00 AM-12:00 PM<br />

Relationship between body mass index (BMI) at diagnosis <strong>of</strong> ER� node negative<br />

breast cancer (BC) and Oncotype DX recurrence score. Presenting Author:<br />

Caroline A. Lohrisch, British Columbia Cancer Agency, Vancouver, BC, Canada<br />

Background: Inferior stage-adjusted survival for BC among obese (O) women has<br />

been reported. This may reflect differences in treatment planned for O women, in<br />

treatment received (due to toxicity), or in tumor biology arising in different<br />

cellular environments, such as high serum insulin levels, which are associated<br />

with a higher risk <strong>of</strong> recurrence. Methods: We examined whether overweight (BMI<br />

25-30) or obese (BMI �30) women had a higher Oncotype Dx Recurrence Score<br />

(RS) after ER�, node negative (NN) or N0i� BC than normal weight women.<br />

Included were all participants at the BC Cancer Agency in a RS clinical utility<br />

study in consecutive ER� NN, N0i� BC (n�156) at the BC Cancer Agency and<br />

in the TAILORx trial (n�56). Sixteen were excluded (n�5 withdrew consent; 1<br />

triple negative; 3 test failed; 2 her2�; 1 neoadjuvant treatment, n�4 height and<br />

weight missing). Results: A similar proportion <strong>of</strong> O patients had low, intermediate,<br />

and high RS tumors. Cancers with a high RS were more likely to be grade 3<br />

(55% <strong>of</strong> grade 3 tumours had high RS, 33% intermediate RS, 12% low RS) and<br />

fewer were strongly ER positive (69% <strong>of</strong> high RS versus 97% <strong>of</strong> intermediate and<br />

low RS). Conclusions: While this data does not support differences in tumor risk<br />

arising in O versus non obese environments in ER�, NN BC, examination <strong>of</strong> a<br />

larger data set may be more informative.<br />

All<br />

RS low<br />

(35, n (%)<br />

8 (4)<br />

2 (2)<br />

4 (6)<br />

2 (6)<br />

584 General Poster Session (Board #7C), Sat, 8:00 AM-12:00 PM<br />

Immunocytochemistry staining for ER and PR in circulating tumor cells as<br />

compared to primary tumor or metastatic biopsy. Presenting Author:<br />

Farideh Z. Bisch<strong>of</strong>f, Biocept Inc., San Diego, CA<br />

Background: Hormone receptor (estrogen receptor [ER] and progesterone<br />

receptor [PR]) status in all breast cancer patients is recommended for<br />

selection <strong>of</strong> treatment options. However, the analytical sensitivity <strong>of</strong><br />

immunohistochemistry (IHC) in detecting low levels <strong>of</strong> ER/PR is <strong>of</strong>ten poor<br />

and is likely due to methodological variation. Because biopsy is not <strong>of</strong>ten<br />

feasible in all patients with recurrent and/or metastatic breast disease,<br />

circulating tumor cells (CTCs) <strong>of</strong>fer an attractive alternative source <strong>of</strong> tumor<br />

tissue for determining ER/PR status and can be monitored more readily to<br />

enable a more effective course <strong>of</strong> treatment. Methods: Twenty mL <strong>of</strong><br />

peripheral blood was collected prospectively from 15 patients diagnosed<br />

with late stage metastatic/recurrent breast cancer. CTCs were isolated<br />

using the micr<strong>of</strong>luidic OncoCEE platform. A cocktail <strong>of</strong> antibodies was<br />

utilized for CTC capture and detection with an expanded anti-cytokeratin<br />

(CK) cocktail mixture and anti-CD45. ER/PR protein expression was<br />

assessed by immunocytochemistry (ICC) on the cells captured within the<br />

microchannels and compared to IHC performed on the primary tumor or<br />

metastatic biopsy. Results: CK�/CD45-/DAPI� cells were located and<br />

assessed for ER/PR ICC. Among the 15 cases, a high concordance (13/15;<br />

87%) in ER/PR status between IHC and ICC results was observed. Two<br />

cases were found to be discordant where one was positive by IHC and<br />

negative on the CTCs, and the other was negative by IHC and positive on the<br />

CTCs. However, both cases that were discordant had low numbers <strong>of</strong><br />

detected CTCs. Conclusions: There is significant heterogeneity between<br />

ER/PR protein expression in CTCs and primary tumor/metastatic biopsy,<br />

and this status may change over time due to therapy. ER/PR ICC on CTCs<br />

from peripheral blood using the OncoCEE platform is shown to be feasible<br />

with high concordance (87%) in ER/PR status between primary tumor/<br />

metastatic biopsy (by IHC) and CTCs (by ICC). The significance <strong>of</strong><br />

heterogeneity at the ER/PR protein level in CTCs ascertaining to the<br />

prognosis and predictive response to anti-estrogen therapy needs further<br />

evaluation in larger prospective clinical trials.<br />

Breast Cancer—HER2/ER<br />

27s<br />

583 General Poster Session (Board #7B), Sat, 8:00 AM-12:00 PM<br />

The role <strong>of</strong> Ki-67 in molecular breast cancer classification. Presenting<br />

Author: George Stathopoulos, Errikos Dunant Hospital and Oncology Clinic,<br />

Athens, Greece<br />

Background: The Ki-67 antigen was identified the involvement in early steps<br />

<strong>of</strong> polymerase I-dependent ribosomal RNA synthesis. Although it seems<br />

that the protein has an important function in cell division, its exact role is<br />

still obscure and there is little published work on its overall function. The<br />

aim <strong>of</strong> the present study is to evaluate the contribution <strong>of</strong> Ki-67 level in<br />

respect <strong>of</strong> tumor recurrence in molecular classified groups <strong>of</strong> breast cancer<br />

patients. Methods: Breast cancer tumor samples were examined for<br />

histological confirmation and for estrogen and progesterone receptors,<br />

c-erb-B2 expression, proliferation with Grade and Ki-67. Ki-67 was divided<br />

in percentage levels, up to 20 and higher than 20%. Immunohistochemistry<br />

and Fluorescence in situ hybridization is described for the results <strong>of</strong> ER,<br />

PR, c-erb-B2, Ki-67 biomarkers. Formaldehyde – fixed breast samples were<br />

paraffin wax embedded and processed for paraffin sections. The primary<br />

antibodies used were: The monoclinal antibody ID5 (M7047, Dakocytomation,<br />

Carpinteria, CA) for the detection <strong>of</strong> ER, the monoclonal anti-PR<br />

antibody 636 was used. For the detection <strong>of</strong> Ki-67 we used monoclonal<br />

mouse anti-human Ki-67 MIB-1. The patients molecular classification was<br />

Luminal A, Luminal B, Her-2 subtype and basal cell (triple negative).<br />

Results: 847 breast cancer patients were recruited. 291 were group as<br />

Luminal A, 228 as Luminal B, 221 Her-2 subtype and 107 triple negative.<br />

Follow-up was from 3 years to 15 years since diagnosis. It was found that in<br />

Luminal A patients, none had Ki-67 higher than 20% and the recurrence<br />

was in 10.65%. In Luminal B, the Ki-67 was higher than 20% in 61% <strong>of</strong><br />

the patients and recurrence 23.68%. In Her-2 subtype �20% Ki-67 was<br />

78.94%, recurrence 17.19%. In triple negative � 20% Ki-67 was in<br />

68.75% and recurrence in 29.90% <strong>of</strong> the patients. Conclusions: The data<br />

presented here indicate that Ki-67 level may be considered as one <strong>of</strong><br />

valuable biomarkers in breast cancer patients process and recurrence.<br />

585 General Poster Session (Board #7D), Sat, 8:00 AM-12:00 PM<br />

Correlation, comparison, and combined analysis <strong>of</strong> Ki-67 and histological<br />

grade for early luminal breast cancer. Presenting Author: Yoichi Naito,<br />

National Cancer Center Hospital East, Chiba, Japan<br />

Background: Both Ki-67 and histological grade are established prognostic<br />

factors in patients with early breast cancer. Recent international expert<br />

consensus employs Ki-67 to classify luminal breast cancer, and histological<br />

grade is described as an alternative. We investigated the prognostic<br />

value <strong>of</strong> Ki-67, histological grade, and their combination. Methods: Patients<br />

with early breast cancer treated with surgery between January 2000 and<br />

December 2008 were retrospectively reviewed. Inclusion criteria were as<br />

follows: estrogen receptor positive, HER2 negative, available for Ki-67 and<br />

histological grade. Clinicopathological data were retrieved from medical<br />

records. Ki-67 labeling index was investigated using MIB-1 antibody and<br />

subdivided into three categories: low 0-9%; intermediate 10-19%; high<br />

�20%. Histological grade was scored between 1 and 3. Disease-free<br />

survival (DFS) was defined as time from surgery to the first documented<br />

disease recurrence or death. Ki-67 categories and histological grade were<br />

analyzed respectively and then concomitantly for prognostic impact on<br />

DFS. Results: A total <strong>of</strong> 606 patients were included. Median age was 58 and<br />

28.1% were over 65 years. 65.4% were postmenopausal women. Progesterone<br />

receptor was positive in 84.0% <strong>of</strong> patients. 29.6% received neoadjuvant<br />

or adjuvant chemotherapy. Number <strong>of</strong> lymph node metastasis was 0 in<br />

70.0%, 1-3 in 21.3%, and �4 in 8.7% <strong>of</strong> patients. Ki-67 categories and<br />

histological grade showed weak but significantly correlation (Spearman’s<br />

rho � 0.385, p�0.001). High Ki-67 and histological grade 3 were<br />

correlated with worse prognosis to the same extent (HR 2.518, p � 0.002<br />

and HR 2.541, p � 0.048, respectively). For combined analysis, patients<br />

represented concomitant with high Ki-67 and histological grade 3 showed<br />

worse prognosis compared with those with Ki-67 low and histological grade<br />

1 (HR 3.137, p � 0.021). Conclusions: Ki-67 and histological grade were<br />

significantly but weakly correlated. Combined use <strong>of</strong> these two factors<br />

could better predict recurrence <strong>of</strong> early luminal breast cancer.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


28s Breast Cancer—HER2/ER<br />

586 General Poster Session (Board #7E), Sat, 8:00 AM-12:00 PM<br />

Relapse rate after adjuvant trastuzumab in the northeast <strong>of</strong> Italy: Preliminary<br />

data from a multicenter observational study in a clinical setting.<br />

Presenting Author: Giorgio Mustacchi, UOC Centro Oncologico, ASS1 Friuli<br />

Venezia Giulia/Università, Trieste, Italy<br />

Background: Relapse Rate (RR) after adjuvant trastuzumab (AT) reported in<br />

clinical trials is between 15 and 21%. Information on RR and clinical<br />

behaviour <strong>of</strong> HER2-POS metastatic disease after AT outside clinical trials<br />

is limited. At ASCO <strong>Meeting</strong> 2010 RR was reported 8% (Abs 1080) to 10%<br />

(Abs 684), with a Survival Post Progression (SPP) <strong>of</strong> 8.8 and 5.7 months,<br />

respectively. It is useful to increase informations about metastatic HER2-<br />

POS disease after AT, outside clinical trials. Methods: HER2-POS consecutive<br />

cases treated with AT in 6 Hospitals <strong>of</strong> North East Italy were<br />

anonymously collected and merged. The following data were analyzed:<br />

Stage, ER/PgR Status, Chemotherapy regimens (CHT) , follow up, RR,<br />

Disease Free Survival (DFS), Overall Survival (OS) and SPP. Results: The<br />

number <strong>of</strong> patients was 413, median age 55 (24-84). Anatomic Stage was I<br />

in 39.7%, II in 40,3%, III in 24.4%. ER/PgR neg cases were 164 (41.6%).<br />

Given CHT were Anthra-based 33.4% and Anthra-Taxane 54.5%. At a<br />

mean follow up <strong>of</strong> 35 months (7-115) RR was 10.8% (45 cases), with a<br />

mean DFS <strong>of</strong> 23.8 months (3.2-74). Among relapsed patients, 37/45<br />

(82.2%) had ER/PgR neg tumors. One patient died NED for CHF and 22 for<br />

metastatic disease, with a mean OS <strong>of</strong> 37.1 months (10.2-87.9). Mean<br />

SPP was 17.8 months. According to DFS �12 mos , �12�24 and �24,<br />

mean OS was 31.3, 31.7 and 56.12(p�0.03) months; SPP was 24.2,<br />

14.4 and 17.8 months, respectively. Conclusions: In our experience RR<br />

after AT is lower than in published Phase III clinical trials. ER/PgR neg<br />

status seems a negative prognostic factor (82.2% in the relapsed population<br />

vs 41.6% overall). OS is similar for patients relapsed within 24 months<br />

but SPP is 10 months longer in very early relapses. Relapse after 2 years is<br />

correlated with a long OS (nearly 5 years), with SPP shorter vs early relapse<br />

(17.8 vs 24.2 months). Our preliminary results suggest different disease<br />

behaviors in relapsed HER2-POS Breast Cancer after AT, with different<br />

DFS and SPP, suggesting a different effect <strong>of</strong> further therapies. Considering<br />

the small RR, the study target accrual is proceeding to collect up to<br />

more than 1000 AT treated patients, including post-progression treatments,<br />

not available at the moment.<br />

588 General Poster Session (Board #7G), Sat, 8:00 AM-12:00 PM<br />

A multicenter phase II trial <strong>of</strong> the LH-RH analogue and an aromatase<br />

inhibitor combination in premenopausal patients with advanced or recurrent<br />

breast cancer refractory to an LH-RH analogue with tamoxifen: JMTO<br />

BC08-01. Presenting Author: Reiki Nishimura, Kumamoto City Hospital,<br />

Kumamoto, Japan<br />

Background: The aim was to provide an endocrine therapy option against<br />

advanced (ABC) or recurrent breast cancer (RBC) in premenopausal<br />

women. We conducted an exploratory phase II trial in combination with an<br />

LH-RH analogue (LH-RHa) and an aromatase inhibitor (AI) to assess the<br />

efficacy and tolerability after failure <strong>of</strong> standard LH-RHa plus tamoxifen<br />

(TAM). Methods: Premenopausal patients (pts) with ER� and/or PgR� ABC<br />

or RBC refractory to LH-RHa � TAM were treated with LH-RHa (goserelin:<br />

GOS) and AI (anastrozole: ANA). The primary endpoint was an objective<br />

response rate (ORR). Secondary endpoints included progression-free survival<br />

(PFS), overall survival (OS), clinical benefit rate (CBR) based on<br />

RECIST, and safety assessed using CTCAE ver. 3.0. Pts with only bone<br />

legions were assessed using the criteria by Japanese Breast Cancer <strong>Society</strong><br />

(14th Ed.). Local assessment (CR, PR or long SD <strong>of</strong> 24 weeks or longer) was<br />

confirmed independently by two radiologists. Results: Between September<br />

2008 and November 2010, 37 pts were enrolled at 10 clinical institutions<br />

in Japan. Eleven had recurrence either during, or within one year after the<br />

end <strong>of</strong> adjuvant GOS � TAM (including GOS � TAM followed by only TAM).<br />

The disease progressed in 26 women during GOS � TAM. Mean age and<br />

BMI were 43.5 years and 22.2 kg/m2, respectively. Thirty-five pts (94.6%)<br />

were ER�, and 36 pts (97.3%) were HER2- (one with unknown HER2<br />

status). Non-endocrine treatment included chemotherapy (20 pts; 54%)<br />

and radiation therapy (13 pts; 35%). The viscera, s<strong>of</strong>t tissue, and bones<br />

were treated in 17, 15 and 14 pts, respectively. Pts with both measurable<br />

lesions and bone metastasis, measurable lesions only, and only bone<br />

metastasis were 21 (57%), 15 (41%) and 1 (2%), respectively. ORR was<br />

18.9% (95%CI: 8.0-35.2%, 1 CR and 6 PR cases), CBR 62.2% (23 pts,<br />

95%CI: 44.8-77.5%), and median PFS was 7.2 months. Eight pts<br />

(21.6%) had adverse events, but none resulted in treatment discontinuation.<br />

GOS � ANA was well tolerated. Conclusions: LH-RHa (GOS) � ANA<br />

can be a subsequent endocrine treatment for premenopausal pts with ABC<br />

or RBC after failure <strong>of</strong> GOS � TAM.<br />

587 General Poster Session (Board #7F), Sat, 8:00 AM-12:00 PM<br />

Endocrine therapy in early breast cancer: Encouraging observations in a<br />

nontrial setting. Presenting Author: Susan Faye Dent, The Ottawa Hospital<br />

Cancer Centre, Ottawa, ON, Canada<br />

Background: Poor adherence to adjuvant endocrine therapy (ET) in women<br />

with hormone receptor positive (HR �) early breast cancer (EBC) is well<br />

documented. Given the availability <strong>of</strong> multiple ET strategies [aromatase<br />

inhibitor (AI) upfront, tamoxifen (TAM) to AI, AI after 5 years TAM], we<br />

evaluated the delivery <strong>of</strong> ET in a non-trial setting. Methods: Postmenopausal<br />

women with HR � EBC treated with ET (that included an AI) at<br />

The Ottawa Hospital Cancer Center between 01/99 and 12/06 were<br />

included. Data was retrospectively collected and included: demographics,<br />

choice/duration <strong>of</strong> ET, adherence and toxicities. Results: In 626 patients<br />

(pts), median age 59 years (r: 30-92) with stage I (195 pts; 31 %), stage II<br />

(339 pts; 54 %) and stage III (88 pts; 14 %) EBC. Treatment strategies<br />

included: AI (s) only (251 pts; 40 %); TAM followed by AI (s) (323 pts;<br />

51.6 %); AI (s) followed by TAM (16 pts; 2.6 %); TAM-AI (s)-TAM (24 pts;<br />

3.8 %) and unknown (12 pts; 1.9 %). Prescribed AI therapy (upfront or<br />

sequential) was discontinued in 39 % <strong>of</strong> cases mainly due to toxicity (62.5<br />

%) after median <strong>of</strong> 6, 9, or 12 months (ms) on exemestane, anastrazole and<br />

letrozole respectively. Common toxicities included: arthralgias (40.6 %),<br />

hot flushes (34 %), fatigue (25 %), myalgias (24.7 %) and osteoporosis<br />

(22.5%). In cases (177) where AI therapy was discontinued due to toxicity:<br />

98 received no further ET, 29 TAM and 50 another AI. The majority <strong>of</strong> pts<br />

received at least 5 years <strong>of</strong> ET (79 %; median duration 64 ms; r 1-156 ms)<br />

and 376 pts (60 %) completed at least 5 years <strong>of</strong> AI therapy (median<br />

duration 59 ms; r: 1-124 ms). 9.9 % <strong>of</strong> pts are still receiving ET.<br />

Conclusions: This is one <strong>of</strong> the first non-clinical trial studies to demonstrate,<br />

that despite poor adherence to initial ET, the majority <strong>of</strong> women are able to<br />

complete five years <strong>of</strong> treatment (79 %). These results are encouraging and<br />

reflect the practical use <strong>of</strong> mutilple endorcrine strategies that have been<br />

shown to be efficacious in this pt population.<br />

589 General Poster Session (Board #7H), Sat, 8:00 AM-12:00 PM<br />

Multiple chemotherapy (CT) lines in metastatic breast cancer (MBC):<br />

Which survival benefit for women with hormone receptor (HR)-positive<br />

disease? Presenting Author: Raffaella Palumbo, Fondazione Maugeri-<br />

IRCCS, Pavia, Italy<br />

Background: Although the development <strong>of</strong> modern systemic therapies has<br />

clearly improved outcome <strong>of</strong> patients with MBC, the true impact <strong>of</strong> further<br />

CT on overall survival (OS) and QoL <strong>of</strong> these women is still debated. The aim<br />

<strong>of</strong> this study was to determine which benefit could be brought by successive<br />

CT lines in patients with HR-positive disease, aiming to identify factors<br />

affecting outcome and survival. Methods: This retrospective analysis<br />

included 980 women treated with CT for MBC at our Institution over a eight<br />

year period (July 2000-July 2008). With OS data updated in March 2010,<br />

the median follow-up was 146 months (range 48-198), OS and time to<br />

treatment failure (TTF) were calculated according to the Kaplan-Meyer<br />

method for each CT line. Cox proportional hazards model was used to<br />

identify factors that could influence TTF and OS. Results: Median OS<br />

evaluated from day 1 <strong>of</strong> each CT line decreased with the line number from<br />

34.8 months for first line to 8.2 months for 7 or more lines). Median TTF<br />

ranged from 9.2 months to 7.8 and 6.4 months for the first, second and<br />

third line, respectively, with no significant decrease observed beyond the<br />

third line (median 5.2 months, range 4.8-6.2). No statistically significant<br />

difference was found between HR-positive and HR-negative patients in<br />

terms <strong>of</strong> OS and TTF by each CT line. In univariate analysis factors<br />

positively linked to a longer duration <strong>of</strong> TTF for each CT line were positive<br />

hormonal receptor status, more than 3 hormonotherapy lines, absence <strong>of</strong><br />

liver metastasis, adjuvant CT exposure, response to CT for the metastatic<br />

disease; in the multivariate analysis the duration <strong>of</strong> TTF for each CT line<br />

was the only one factor with significant impact on survival benefit for<br />

subsequent treatments, in both HR-positive and negative populations<br />

(p�0.001). Conclusions: Our results support the benefit <strong>of</strong> multiple lines <strong>of</strong><br />

CT in a significant subset <strong>of</strong> women treated for MBC, since each CT line<br />

may contribute to a longer OS. Of interest, such a benefit was also observed<br />

for patients with HR-positive disease, although the number <strong>of</strong> hormonotherpy<br />

lines received did not significantly influence the outcome.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


590 General Poster Session (Board #8A), Sat, 8:00 AM-12:00 PM<br />

Docetaxel and cyclophosphamide as neoadjuvant chemotherapy in ER�<br />

HER2- breast cancer. Presenting Author: Hiroshi Yagata, St. Luke’s<br />

International Hospital, Tokyo, Japan<br />

Background: Recently, docetaxel plus cyclophosphamide (TC) has been<br />

established as a standard regimen for adjuvant chemotherapy in operable<br />

breast cancer. However, the efficacy <strong>of</strong> TC as neoadjuvant chemotherapy<br />

remains unclear, especially in hormone-responsive breast cancer. We<br />

prospectively evaluated the response to TC neoadjuvant chemotherapy in<br />

patients with ER� HER2- operable breast cancer. Methods: Eligible<br />

patients had ER� and HER2- invasive breast cancer that measured more<br />

than 2 cm in diameter or was node-positive clinically between March 2009<br />

and February 2011. Four cycles <strong>of</strong> TC (75 and 600 mg/m2 ) were<br />

administered intravenously every 3 weeks as neoadjuvant chemotherapy,<br />

followed by breast and axillary surgery. We investigated the clinical<br />

response on MRI and the pathological complete response (pCR) <strong>of</strong> primary<br />

invasive breast tumors. Results: We enrolled 43 patients in 45 lesions.<br />

Allred score <strong>of</strong> ER was 7 to 8 in 40 lesions, 6 in 3 lesions and 4 in 2 lesions.<br />

PgR was 7 to 8 in 31 lesions, 5 to 6 in 6 lesions, 3 to 4 in 5 lesions, and 0 in<br />

3 lesions. Nuclear grade was 1 in 31 lesions, 2 in 7 lesions, and 3 in 7<br />

lesions. MIB-1 index was median 22% (range, 6 to 71%) in 40 measurable<br />

lesions. <strong>Clinical</strong> response (CR and PR) was observed in 33 lesions (73%)<br />

and SD in 12 lesions without any lesions <strong>of</strong> clinical PD. pCR was achieved<br />

in only 1 lesions (ER 8, PgR 3, Nuclear grade 3, MIB-1 index 32.6%).<br />

Conclusions: Neoadjuvant TC produced a good clinical response in ER�<br />

HER2- operable breast cancer, while the pCR rate was very low, regardless<br />

<strong>of</strong> the characteristics <strong>of</strong> cancer cells. Therefore, pCR is not an appropriate<br />

prognostic factor in women with ER� HER2- operable breast cancer who<br />

receive neoadjuvant TC. Whether clinical response is related to survival<br />

should be addressed in future studies. The efficacy <strong>of</strong> neoadjuvant TC in<br />

ER� HER2- operable breast cancer is very limited, and more effective<br />

regimens are needed to achieve higher pCR rates.<br />

592 General Poster Session (Board #8C), Sat, 8:00 AM-12:00 PM<br />

ESR1 gene amplification and protein expression in 946 patients with<br />

resected breast cancer: A Hellenic Cooperative Oncology Group (HeCOG)<br />

translational research study. Presenting Author: George Pentheroudakis,<br />

Hellenic Cooperative Oncology Group, Athens, Greece<br />

Background: Discrepant data have been reported on the incidence <strong>of</strong> ESR1<br />

gene amplification in breast cancer, its correlation to clinicopathologic<br />

characteristics and its impact on prognosis. Methods: Formalin-fixed<br />

paraffin-embedded (FFPE) tumor tissue samples from 946 patients participating<br />

in two adjuvant chemotherapy phase III trials (HE10/97 and<br />

HE10/00) were centrally assessed in tissue microarrays by immunohistochemistry<br />

(IHC) for estrogen receptor (ER), progesterone receptor (PgR)<br />

and human epidermal growth factor receptor 2 (HER2). FISH was also<br />

performed for HER2, TOP2A and ESR1 using commercially available dual<br />

(for ESR1) and triple (for HER2 and TOP2A) hybridization probes. Results:<br />

The majority <strong>of</strong> patients had �T2 (69%), node-positive, ER-positive (�1%<br />

stained cells, 73%) tumors managed with resection, chemotherapy and<br />

hormonotherapy (76%). Only 38 tumors (4.0%) had ESR1 gene amplification<br />

(ESR1/CEN6 ratio �2) and 514 (54.3%) ESR1 gene gain (1�ratio�2).<br />

The number <strong>of</strong> ESR1 gene copies was 3-4 in 251 (26.5%) and<br />

5-10 in 42 (4.4%) <strong>of</strong> cases. HER2 and TOP2A gene amplification was seen<br />

in 234 (25.3%) and 101 (10.9%) <strong>of</strong> tumors, respectively. We studied the<br />

immunohistochemical expression <strong>of</strong> ER protein by evaluating the percentage<br />

<strong>of</strong> stained cells, the Allred score (0-2, 26.8%; 3-6, 62.5%; 7-8,<br />

10.7% <strong>of</strong> tumors) and the semiquantitative H-Score (50-100, 13.8%;<br />

101-200, 36.8%; 201-300, 15% <strong>of</strong> tumors). ESR1 gene amplification<br />

was significantly associated with taxane therapy, age �50, postmenopausal<br />

status, grade III-IV, absence <strong>of</strong> HER2 amplification and ER protein<br />

expression (p�0.05). At a median follow-up <strong>of</strong> 92 months, univariate Cox<br />

regression analysis showed that ER protein expression, but not ESR1 gene<br />

status, was a predictor <strong>of</strong> favorable outcome. In multivariate analysis,<br />

tumor size �5 cm, �4 involved nodes and negative/low ER protein<br />

expression by Allred score were independent adverse prognostic factors.<br />

Conclusions: Our data showed a rather low incidence <strong>of</strong> ESR1 gene<br />

amplification and failed to confirm its prognostic/predictive utility. ESR1<br />

mRNA expression data will be presented at the meeting.<br />

Breast Cancer—HER2/ER<br />

29s<br />

591 General Poster Session (Board #8B), Sat, 8:00 AM-12:00 PM<br />

A short-term neoadjuvant pilot study comparing exemestane and letrozole<br />

in postmenopausal women with estrogen receptor-positive HER2-negative<br />

breast cancer. Presenting Author: Daishu Miura, Toranomon Hospital,<br />

Tokyo, Japan<br />

Background: Current approaches <strong>of</strong> neoadjuvant endocrine therapy (NAET)<br />

are focusing on biomarker analysis <strong>of</strong> the diagnostic specimens. One <strong>of</strong> the<br />

important things in neoadjuvant treatment is to determine the in-vivo<br />

responsiveness to the agent. Little is known regarding biomarker changes in<br />

specific agents and the association with biomarker changes and treatment<br />

duration. A pilot study was conducted to investigate the differences <strong>of</strong><br />

biomarker changes in short-term NAET (letrozole vs. exemestane) and also<br />

the difference in treatment durations <strong>of</strong> each AI. Methods: Totally 120<br />

samples from 60 postmenopausal women with ER positive HER2 negative<br />

stage 1-3 breast cancers in NAET comparing letrozole (LET) and exemestane<br />

(EXE) were analyzed with pre- and post-treatment mRNA expression <strong>of</strong><br />

ER�,ER�, PR, HER2, Ki67, PIK3CA, and MAPK using QuantiGene Assay.<br />

Thirty-one patients took letrozole 2.5mg daily and 29 did exemestane<br />

25mg daily until the surgery (median duration 22 days). Statistical tests<br />

were two-sided. Results: <strong>Clinical</strong> characteristics were well balanced between<br />

the two treatment arms. Median % changes <strong>of</strong> Ki67 were -58%, ER�<br />

-44%, ER� �93%, and PR -69% in LET and Ki67 -59%, ER� -30%, ER�<br />

�135%, PR -72% in EXE. There were no significant differences <strong>of</strong> these<br />

marker changes between two agents. Dividing into 4 groups in treatment<br />

duration, 10-14 days (q1), 15-21 days (q2), 22-28 days (q3), and over 29<br />

days (q4), LET had 2 peaks in Ki67 decreases (-69% (q1), -28% (q2),<br />

-78% (q3), -59% (q4)), whereas EXE had a peak in q2 (-62%, -77%,<br />

-49%, -26%). Ki67 increasing cases including primary resistance and<br />

tachyphylaxis were 5 (16%) in LET (3 in q2 and 2 in q4) and 3 (10%) in<br />

EXE (one <strong>of</strong> each in q2-4). All 8 cases had increases <strong>of</strong> ER�, PIK3CA, and<br />

MAPK. Conclusions: Although both LET and EXE for short duration had<br />

significantly decreased Ki67 mRNA level irrespective <strong>of</strong> treatment duration,<br />

no significant differences was found between two agents. Tumor<br />

resistance with AIs may be associated with the other pathways.<br />

593 General Poster Session (Board #8D), Sat, 8:00 AM-12:00 PM<br />

Real-world aromatase inhibitor use and failure in women with metastatic<br />

ER�/HER2- breast cancer. Presenting Author: Pamela Landsman-<br />

Blumberg, Thomson Reuters, Washington, DC<br />

Background: Aromatase Inhibitors (AIs) have replaced tamoxifen as firstline<br />

therapy for postmenopausal women with metastatic, ER� breast<br />

cancer (BC). However, little information is available on the real-world use <strong>of</strong><br />

AIs. The objectives <strong>of</strong> this retrospective claims study were to compare<br />

demographic, clinical and treatment characteristics <strong>of</strong> postmenopausal<br />

women with metastatic ER�/HER2- BC treated with AIs and experiencing<br />

0, 1, 2 or � 3 AI failures (AIF). Methods: Women � 55 years old, newly<br />

diagnosed with metastatic BC (index) were identified in the 2006-2010<br />

Thomson Reuters MarketScan databases and followed until chemotherapy<br />

or 03/31/2011. ER�/HER2- disease was defined as any endocrine therapy<br />

(ET: tamoxifen, fulvestrant) or AI (anastrozole, letrozole, or exemestane)<br />

use and no trastuzumab or lapatinib use in the 6-month pre-or variable<br />

post-index periods. Those with any post-index AI use were retained for<br />

study. AIF post-index was defined as a switch to an alternative AI, ET or<br />

chemotherapy, or AI discontinuation with no further BC treatment. Patients<br />

were stratified by number <strong>of</strong> post-index AIF: 0, 1, 2 or � 3. Results: Among<br />

4,274 patients identified, 61% had � 1 AIF (1: 80%, 2: 15%, �3: 5%).<br />

There was no difference in pre-index AI use across AIF cohorts: 0, 1, 2, and<br />

�3 (54%, 52%, 48%, 56%; p�0.073). AIFs increased with Medicareeligibility<br />

(51%, 56%, 60%, 61%) and bone metastases at index (48%,<br />

53%, 62%, 63%). Among those with �1 AIF, median follow-up (FU)<br />

increased with each failure but there was no notable pattern to reason for<br />

FU end. Median FU <strong>of</strong> the 0 AIF cohort (408 days) fell between those <strong>of</strong> the<br />

1 and 2 AIF cohorts, 335 and 517 days. Anastrozole was the most common<br />

first line treatment for all cohorts except � 3 AIFs where letrozole was most<br />

common. Pre-index and first line fulvestrant use both increased with the<br />

number <strong>of</strong> post-index AIFs: 0.3%, 1.9%, 2.0%, 5.0% and 0.7%, 2.5%,<br />

4.0%, 14.9% respectively. There was no association between number <strong>of</strong><br />

AIFs and chemotherapy use (36%, 29%, 38%). Conclusions: Over 60% <strong>of</strong><br />

women with ER�/HER2- metastatic BC treated with AIs failed at least 1<br />

and 20% <strong>of</strong> those failed � 2. Surprisingly, increased rates <strong>of</strong> prior<br />

fulvestrant treatment appear associated with increasing numbers <strong>of</strong> AIF.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


30s Breast Cancer—HER2/ER<br />

594 General Poster Session (Board #8E), Sat, 8:00 AM-12:00 PM<br />

Limited efficacy and significant toxicities <strong>of</strong> lapatinib (Lap) plus chemotherapy<br />

as neoadjuvant therapy (NAC) for HER2-positive inflammatory<br />

breast cancer (IBC) patients (Pts). Presenting Author: Ricardo H. Alvarez,<br />

The Morgan Welch Inflammatory Breast Cancer Program and Clinic,<br />

University <strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: IBC is a rare disease but the most aggressive form <strong>of</strong> locally<br />

advanced breast cancer with a higher frequency <strong>of</strong> HER2 neu amplification<br />

(HER2�) compared to non-IBC. The EGFR pathways are also important<br />

therapeutic targets in IBC. (Zhang D, 2009). Lap is an oral reversible dual<br />

kinase inhibitor <strong>of</strong> epidermal growth factor receptor <strong>of</strong> EGFR and HER2.<br />

Our objective was to determine the efficacy <strong>of</strong> Lap in combination with<br />

paclitaxel as NAC in pts with previously untreated HER2� IBC. The primary<br />

end-point was to determine the rate <strong>of</strong> pathological complete response<br />

(pCR) defined as no residual invasive disease in the breast and the axillary<br />

lymph nodes or Residual Cancer Burden (RCB) <strong>of</strong> 0. (Symmans FW, 2008).<br />

The second endpoint was to assess general safety and cardiac toxicity <strong>of</strong><br />

this combination therapy. Methods: From October 2008 to May 2011, 15<br />

chemo-naïve pts were treated with Lap 1,250 mg/day as a single agent for 2<br />

weeks, followed by 12 weeks <strong>of</strong> paclitaxel (80 mg/m2 weekly) plus Lap<br />

(1,000 mg/day), and finally with 4 cycles <strong>of</strong> FEC-75 mg/m2 regimen plus<br />

Lap (1,000 mg/day) before surgery. Among 12 first pts enrolled, 6 pts had<br />

grade 3 diarrhea (CTCAC v3.0) and the protocol was amended to reduce the<br />

Lap dose to 750 mg/day. After modified radical mastectomy (MRM),<br />

patients received radiation therapy and one year <strong>of</strong> adjuvant trastuzumab.<br />

Results: Median age was 53.8 (range, 39 -70), performance status was 0<br />

(13 pts), 1 (2 pts), 4 pts had metastatic disease at diagnosis. 10 <strong>of</strong> 15 pts<br />

had MRM. One pt achieved pCR. Five pts did not complete the NAC due to<br />

grade 2 cardiomyopathy (1), liver dysfunction (2), diarrhea (1), and<br />

insurance denial after pt started treatment (1). The trial was stopped early<br />

due to severe toxicity (�15%) and lack <strong>of</strong> efficacy. Conclusions: Lap in<br />

combination with chemotherapy as NAC has a limited anti-tumor activity in<br />

pts with HER2� IBC with a pCR rate <strong>of</strong> 6.6%. Lap and paclitaxel was<br />

associated with severe diarrhea toxicity and required multiple dose reductions<br />

<strong>of</strong> Lap. With the recent promising results in HER2� NAC studies, the<br />

role <strong>of</strong> Lap should continue to be explored in combination with other<br />

anti-HER2 agents in IBC patients.<br />

596 General Poster Session (Board #8G), Sat, 8:00 AM-12:00 PM<br />

Adjuvant lapatinib in women with early-stage HER2-positive breast cancer<br />

(HER2� BC): Analysis <strong>of</strong> the hormone receptor-negative subgroup <strong>of</strong> the<br />

intent-to-treat (ITT) population <strong>of</strong> the TEACH trial. Presenting Author: Ian<br />

Edward Smith, The Royal Marsden Hospital, London, United Kingdom<br />

Background: TEACH is a randomized, double-blind, placebo (P)-controlled<br />

trial evaluating lapatinib (L) in reducing relapse risk in trastuzumab -naive<br />

patients (pts) previously treated with chemotherapy for HER2� BC. The<br />

primary results (presented at SABCS 2011) showed a hazard ratio (HR) for<br />

disease-free survival (DFS) in L arm compared with P <strong>of</strong> 0.83 (95%<br />

confidence interval [CI] 0.70-1.00; stratified log-rank 2-sided P�.053).<br />

The predefined hormone receptor negative subgroup was analyzed, which<br />

in contrast to the hormone receptor positive subgroup <strong>of</strong> the ITT population<br />

(N�3147) had a significantly improved DFS. Methods: 3161 HER2� BC<br />

pts (Stage I-IIIc) were randomized 1:1 to L daily or P for 1 year (yr). Primary<br />

endpoint was DFS in the ITT population. Central nervous system (CNS)<br />

recurrence rate was a secondary endpoint. A subgroup analysis by Cox<br />

Proportional Hazards Regression Models was performed for the hormone<br />

receptor negative pts from the ITT population. Results: 1288 pts (41%)<br />

comprised the hormone receptor negative subgroup <strong>of</strong> the ITT (L:639, and<br />

P:649). Median time from diagnosis to randomization was 2.4 (0.3-15)<br />

yrs, median follow-up was 4 yrs. Median age for this subgroup was 53<br />

(24-87) yrs. With 85 events in L arm and 128 in P arm, the HR for DFS for<br />

hormone receptor negative pts was 0.68 (95%CI 0.52-0.89) favoring L.<br />

Most hormone receptor negative subgroups showed benefit for L: pts up to<br />

1 yr from diagnosis (HR 0.64; 95%CI 0.41-0.99), node negative (HR 0.57;<br />

95%CI 0.35-0.92), premenopausal (HR 0.59, 95%CI 0.37-0.94), and<br />

those who received prior anthracycline chemotherapy without taxanes (HR<br />

0.63; 95%CI 0.43-0.92). Node positive patients had a trend to benefit<br />

from L (HR 0.74; 95%CI 0.53-1.03). CNS as one site <strong>of</strong> initial recurrence<br />

occurred in 1% in L (n�7) and P (n�8) arms. Conclusions: Lapatinib<br />

improved DFS in patients early or late in follow-up from diagnosis <strong>of</strong><br />

HER2�, hormone receptor negative BC. Similar subset analyses are<br />

important in other large adjuvant anti-HER2 therapy trials. Results might<br />

provide a better understanding <strong>of</strong> the subtypes <strong>of</strong> HER2� disease and help<br />

to further individualized therapy.<br />

595 General Poster Session (Board #8F), Sat, 8:00 AM-12:00 PM<br />

Neoadjuvant chemotherapy versus neoadjuvant hormonal therapy in postmenopausal<br />

women with ER-positive, HER2/neu negative locally advanced<br />

breast cancer. Presenting Author: Tadeu Frantz Ambros, Jackson Memorial<br />

Hospital, University <strong>of</strong> Miami, Miami, FL<br />

Background: Postmenopausal women with large ER�/HER-2 negative<br />

tumors frequently receive neoadjuvant chemotherapy (NC), but pathological<br />

complete response (pCR) rates are low. Neoadjuvant hormonal therapy<br />

(NH) may <strong>of</strong>fer benefit in this setting. Methods: Retrospective review <strong>of</strong><br />

medical records from University <strong>of</strong> Miami/Jackson Memorial Hospital from<br />

1998-2011. Primary outcomes: pCR (absence <strong>of</strong> invasive tumor in breast<br />

and lymph nodes at surgery), recurrence free survival (RFS) and tumor size<br />

reduction evaluated through comparison <strong>of</strong> palpable breast mass size at<br />

presentation with pathological tumor size in surgical specimen, and<br />

categorized as good response (GR) � 30% reduction or no response (NR) �<br />

30%. The Kaplan-Meier method and the log-rank test were used in the<br />

analysis <strong>of</strong> RFS. Results: Data from 151 post-menopausal women with<br />

ER�/HER-2 negative BC who received NC (57%) or NH (43%) was<br />

analyzed. Median follow-up among alive patients with no evidence <strong>of</strong><br />

disease was 5.4 years in NC and 2.9 years in NH. Mean age was higher in<br />

the NH group (63.3 vs 56.1, p�0.0001). There were no racial or ethnic<br />

differences between the groups. <strong>Clinical</strong> stage was comparable in NC and<br />

NH (IIA 5.8% vs 9.2%, IIB 25.6% vs 20%, IIIA 37.2% vs 29.2%, IIIB/IIIC<br />

31.4% vs 41.5%, p�0.775). Tumor histology was predominantly ductal in<br />

both groups (NC 85.7% and NH 78.5%, p�0.247). pCR was similar in NC<br />

and NH (4.7% vs 0%, p�0.078) along with RFS (median 8.5 yrs vs 6.0<br />

yrs, p�0.946). In the NC group, GR was significantly more frequent<br />

(77.9% vs 60%, p�0.017). Among patients in the NH group, having GR<br />

was predictive <strong>of</strong> longer RFS (5-year rate 83.7% vs 50.5%, p�0.014).<br />

Breast only pCR occurred at equivalent rates between NC and NH (9.3% vs<br />

3.1%, p�0.189) as did the absence <strong>of</strong> lymph node metastasis (29.1% vs<br />

26.2%, p�0.606). In the NH cohort 38.5% received no adjuvant chemotherapy.<br />

Conclusions: NH provides an effective alternative to NC and, if<br />

there is a GR, may preclude the need for chemotherapy in over one third <strong>of</strong><br />

postmenopausal women with large ER positive/HER-2 negative breast<br />

cancer.<br />

597^ General Poster Session (Board #8H), Sat, 8:00 AM-12:00 PM<br />

Adverse events with pertuzumab and trastuzumab: Evolution during treatment<br />

with and without docetaxel in CLEOPATRA. Presenting Author: José<br />

Baselga, Massachusetts General Hospital Cancer Center and Harvard<br />

Medical School, Boston, MA<br />

Background: In CLEOPATRA, the HER2-dimerization inhibitor pertuzumab<br />

(P) was combined with trastuzumab (T) and docetaxel (D) in HER2-positive<br />

1st-line MBC. P�T�D significantly improved efficacy compared with<br />

placebo (Pla)�T�D while having little effect on safety. Pts were recommended<br />

to receive �6 cycles <strong>of</strong> D but could discontinue D prior to Cycle 6<br />

due to poor tolerability or progressive disease (PD) or continue D beyond<br />

Cycle 6 at investigators’ discretion. Once D was discontinued, pts received<br />

Pla�TorP�T until PD. To understand the full safety pr<strong>of</strong>ile <strong>of</strong> the regimen,<br />

adverse events (AEs) occurring before and after D discontinuation were<br />

analyzed. Methods: Treatment was given q3w (Pla/P: 840 mg loading, then<br />

420 mg; T: 8 mg/kg loading, then 6 mg/kg; D: 75 mg/m2 , escalating to 100<br />

mg/m2 if tolerated; de-escalation by 25% allowed). AEs were graded<br />

according to NCI-CTCAE v3.0, monitored continuously during the treatment<br />

period, and their relationship to study drugs was assessed by<br />

investigators. Results: From 808 pts enrolled, 804 were analyzed in the<br />

safety population (pts who received �1 dose <strong>of</strong> study treatment). AEs that<br />

led to discontinuation <strong>of</strong> all study treatment were experienced by 5.3%<br />

(Pla�T�D) and 6.1% (P�T�D) <strong>of</strong> pts, whereas discontinuation <strong>of</strong> D alone<br />

due to AEs occurred in 23.2% (Pla�T�D) and 23.6% (P�T�D) <strong>of</strong> pts.<br />

Conclusions: Treatment in both arms was well tolerated. After discontinuation<br />

<strong>of</strong> D, there was a clear reduction in grade �3 AEs; however, the<br />

incidence <strong>of</strong> grade �3 diarrhea remained slightly elevated with P�T�D.<br />

The AE pr<strong>of</strong>ile <strong>of</strong> P�T is consistent with that seen in previous Phase II<br />

studies (Gianni Lancet Oncol 2012; Baselga JCO 2010). These data<br />

suggest that the combination <strong>of</strong> P�T may also be well tolerated in other<br />

breast cancer settings, such as in adjuvant therapy, which is currently<br />

under phase III study (APHINITY; NCT01358877).<br />

Grade >3 AEs, incidence >5%.<br />

Incidence during<br />

all study treatment<br />

Incidence after<br />

D discontinuation<br />

AE,n(%)<br />

Pla�T�D(n�397) P�T�D(n�407) Pla�T(n�255) P�T(n�298)<br />

Neutropenia 182 (45.8) 199 (48.9) 4 (1.6) 0 (0.0)<br />

Febrile neutropenia 30 (7.6) 56 (13.8) 0 (0.0) 0 (0.0)<br />

Leukopenia 58 (14.6) 50 (12.3) 1 (0.4) 0 (0.0)<br />

Diarrhea 20 (5.0) 32 (7.9) 0 (0.0) 3 (1.0)<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


598^ General Poster Session (Board #9A), Sat, 8:00 AM-12:00 PM<br />

Quality <strong>of</strong> life assessment in CLEOPATRA, a phase III study combining<br />

pertuzumab with trastuzumab and docetaxel in metastatic breast cancer.<br />

Presenting Author: Javier Cortes, Department <strong>of</strong> Oncology, Vall d’Hebron<br />

University Hospital, Barcelona, Spain<br />

Background: CLEOPATRA compared the efficacy and safety <strong>of</strong> the HER2<br />

dimerization inhibitor pertuzumab (P) plus trastuzumab (T) and docetaxel<br />

(D) with placebo (Pla)�T�D in HER2-positive 1st-line MBC. Pts in both<br />

arms received a median <strong>of</strong> 8 cycles <strong>of</strong> D; Pla/P�T was continued until<br />

progressive disease (PD). The safety pr<strong>of</strong>ile in both arms was similar with a<br />

substantial decrease in adverse events (AEs) once chemotherapy finished.<br />

The independently assessed progression-free survival was significantly<br />

improved with P�T�D compared with Pla�T�D; objective response and<br />

duration <strong>of</strong> response were also improved with P�T�D (Baselga NEJM<br />

2012). Here we report health-related quality <strong>of</strong> life (HRQoL) data from<br />

CLEOPATRA. Methods: Time to deterioration <strong>of</strong> HRQoL was evaluated using<br />

the FACT-B questionnaire and defined as decrease from baseline (BL) <strong>of</strong><br />

�5 points in the TOI-PFB subscale score. Female pts completed questionnaires<br />

every 3rd cycle <strong>of</strong> therapy within 3 days before each tumor<br />

assessment until independently determined PD. An exploratory analysis<br />

investigated time to deterioration in breast cancer symptoms and functions.<br />

Results: 56.7% (Pla�T�D) and 59.5% (P�T�D) <strong>of</strong> pts experienced<br />

deterioration <strong>of</strong> HRQoL during the study based on TOI-PFB. The median<br />

time to deterioration was 18.3 vs 18.4 wks (~6 cycles) (HR 0.97; P �<br />

.7161). At Cycle 6, the mean reduction in TOI-PFB score from BL was –3.5<br />

(Pla�T�D) vs –3.0 (P�T�D). At subsequent cycles, when most pts had<br />

discontinued D, mean reductions were smaller, suggesting that after an<br />

early decline patients’ scores improved slightly. Overall, mean changes<br />

were small in both arms. Compliance with completion <strong>of</strong> the FACT-B<br />

questionnaire was �75% beyond the 1st year in both arms. An exploratory<br />

analysis suggested that time to deterioration in BCS score, which measures<br />

symptoms and issues relevant in breast cancer, was delayed with P�T�D<br />

(18.3 vs 26.7 wks; HR 0.77; P � .0061). Conclusions: Combining P with<br />

T�D appears to have no detrimental effect on HRQoL. Results suggest that<br />

P�T�D is associated with a substantial delay in the time to deterioration in<br />

BCS score as would be expected given the improved efficacy and the low<br />

incidence <strong>of</strong> AEs once D is discontinued.<br />

600 General Poster Session (Board #9C), Sat, 8:00 AM-12:00 PM<br />

Trastuzumab-related cardiotoxicity among older breast cancer patients.<br />

Presenting Author: Mariana Chavez-Mac Gregor, University <strong>of</strong> Texas M. D.<br />

Anderson Cancer Center, Houston, TX<br />

Background: The use <strong>of</strong> trastuzumab in the adjuvant setting is associated<br />

with reduction in mortality and recurrence. Although trastuzumab is<br />

extremely well tolerated, its use is associated with congestive heart failure<br />

(CHF). The incidence <strong>of</strong> this complication in the general population<br />

remains largely unknown, especially in older patients. In this study we<br />

evaluated the rates and risk factors associated with trastuzumab-related<br />

CHF in a large cohort <strong>of</strong> older breast cancer patients. Methods: Breast<br />

cancer patients � 66 years with full Medicare coverage, diagnosed with<br />

stage I-III breast cancer between 2005-2007, and treated with chemotherapy<br />

were identified in the Surveillance, Epidemiology and End Results-<br />

Medicare (SEER-Medicare) database. Chemotherapy and trastuzumab use,<br />

comorbidities and CHF were identified using ICD-9 and HCPCS codes.<br />

Analyses included descriptive statistics, Cox proportional hazard model<br />

using trastuzumab as a time-dependent variable and logistic regression.<br />

Results: A total <strong>of</strong> 3,983 patients were included, 847 (21.7%) <strong>of</strong> them<br />

received trastuzumab, median age <strong>of</strong> the entire cohort was 71 years old.<br />

Among trastuzumab users the rate <strong>of</strong> CHF was 28.6% compared to 18.1%<br />

in non-trastuzumab users (p�.0001). After adjusting for demographic and<br />

clinical characteristics, comorbidities, and type <strong>of</strong> chemotherapy used,<br />

trastuzumab users were more likely to develop CHF than non-trastuzumab<br />

users (HR 1.74; 95% CI 1.47-2.06). Among trastuzumab users, older age<br />

did not further increase the risk, however history <strong>of</strong> coronary artery disease<br />

(OR 2.23; 95%CI 1.56-3.19), heart valve disease (OR 1.41; 95%CI<br />

1.41-2.88) and emphysema -as a proxy for smoking- (OR 2.03; 95% CI<br />

1.13-3.63) significantly increased the risk <strong>of</strong> CHF. There was a trend for<br />

increased risk <strong>of</strong> CHF associated with anthracycline use (OR 1.36; 95% CI<br />

0.93-2.00). Conclusions: In this large cohort <strong>of</strong> older breast cancer<br />

patients, the risk <strong>of</strong> CHF among trastuzumab users was 1.74 times higher<br />

than non-trastuzumab users, which is higher than what has been reported<br />

in younger clinical trial participants. Among older breast cancer patients<br />

treated with trastuzumab, cardiac comorbidities and emphysema may<br />

identify high risk patients.<br />

Breast Cancer—HER2/ER<br />

31s<br />

599 General Poster Session (Board #9B), Sat, 8:00 AM-12:00 PM<br />

Impact <strong>of</strong> hormone receptor (HR) status on clinicopathological features,<br />

patterns <strong>of</strong> recurrence, and clinical outcomes among patients (pts) with<br />

human epidermal growth factor receptor-2 positive (HER2) breast cancer<br />

(BC) in the National Comprehensive Cancer Network (NCCN). Presenting<br />

Author: Ines Maria Vaz Duarte Luis, Dana-Farber Cancer Institute, Boston,<br />

MA<br />

Background: According to gene expression pr<strong>of</strong>iling, HER2� BC is heterogeneous<br />

and appears to diverge by HR status. Methods: We evaluated 3394<br />

pts who presented to NCCN centers with stage I-III HER2� BC between<br />

2000-07. Pts were classified as HR� (ER� and/or PR�) and HR- (ER- and<br />

PR-). Chi-square, univariate logistic regression, log-rank test, and Cox<br />

hazard proportional regression were used for analysis. Results: Median<br />

follow-up was 51 months. 59% <strong>of</strong> patients had HR� and 41% HR- disease<br />

respectively. Pts with HR- BC were more likely to be postmenopausal and to<br />

present with higher stage and high grade disease (p�0.001). Most pts<br />

received adjuvant or neoadjuvant therapy; 44% received adjuvant trastuzumab.<br />

Recurrences were recorded for 458 pts. HR- patients were more<br />

likely to recur first in the central nervous system (CNS) (OR: 1.8, 95% CI:<br />

1.1, 2.9; p� 0.03) and less likely to recur in bone (OR: 0.5, 95% CI: 0.3,<br />

0.8; p�0.01). No differences in risk <strong>of</strong> lung or liver recurrence were<br />

observed. Combining first and subsequent sites <strong>of</strong> recurrence, the difference<br />

in CNS involvement was lost (p�0.107) but HR- were more likely to<br />

experience lung involvement (OR: 1.5, 95% CI: 1.0, 2.2; p� 0.05). After<br />

adjusting for age, year <strong>of</strong> diagnosis (y), race, stage, and grade, HR- had<br />

worse survival after initial BC diagnosis than HR� pts (Hazard Ratio <strong>of</strong><br />

death [HRd] 1.4, 95% CI: 1.14, 1.7; p�0.01). However, the risk <strong>of</strong> death<br />

was not proportional over time with HR- having significantly increased<br />

hazard in the first five years: HRd 0-2 y 1.9 [1.3, 2.9]; p� 0.01; HRd 2-5y<br />

1.5, [1.2, 2.0]; p�0.01; HRd 5� y, 0.8, [ 0.6, 1.2], p�0.29. Conclusions:<br />

Clinicopathological features, sites <strong>of</strong> recurrence, and risk <strong>of</strong> death over time<br />

for HER2� BC differed by HR status. This suggests that HR status in<br />

HER2� BC is clinically relevant. These differences should be further<br />

explored from a mechanistic and therapeutic standpoint.<br />

601 General Poster Session (Board #9D), Sat, 8:00 AM-12:00 PM<br />

Impact <strong>of</strong> adjuvant trastuzumab-based chemotherapy in T1ab nodenegative<br />

HER2 overexpressing breast carcinomas. Presenting Author:<br />

Jean-Sebastien Frenel, Institut de Cancerologie de l’Ouest/Site Rene<br />

Gauducheau, Saint-Herblain, France<br />

Background: HER2 overexpression has been recognized as a pejorative<br />

prognostic factor in node negative T1ab (T1abN0) breast tumors. Randomized<br />

clinical trials have shown benefit <strong>of</strong> adjuvant trastuzumab-based<br />

chemotherapy (ATBC) for node-positive and/or greater than 1 cm (T1c or<br />

more) HER2� breast carcinomas. There are no prospective efficacy data <strong>of</strong><br />

ATBC in T1abN0 HER2� tumors. Methods: We retrospectively evaluated<br />

276 cases <strong>of</strong> T1ab node-negative HER2� breast tumors in 8 French<br />

Comprehensive Cancer Centers. We assessed clinical, therapeutic features<br />

and outcome. We estimated the probability <strong>of</strong> disease-free survival (DFS),<br />

analyzed associations <strong>of</strong> ATBC, patient and tumor characteristics with DFS<br />

and prognosis factors using the log-rank test, multivariate analysis with<br />

logistic regression and Cox’s proportional hazards model. Results: Out <strong>of</strong> the<br />

276 T1abN0 cases, 129 (47%) received ATBC (ATBC�) and 123 (45%)<br />

were not treated by either trastuzumab or chemotherapy (ATBC-). Of these<br />

252 ATBC� or ATBC- patients, decision <strong>of</strong> ATBC was associated with date<br />

<strong>of</strong> diagnosis (before or after ASCO 2005 <strong>Annual</strong> <strong>Meeting</strong> when interim<br />

results from three trastuzumab adjuvant trials were reported) and with poor<br />

prognosis features: negative hormone receptors (HR-) status, Elston-Ellis<br />

high grade, tumor size � 5 mm and age. With a median follow-up <strong>of</strong> 44<br />

months 16 recurrences were observed (13 in the ATBC- group, 2 in the<br />

ATBC� and 1 with adjuvant chemotherapy alone). Nine recurrences were<br />

distant metastases. A survival benefit in ATBC� was observed with a 99%<br />

40-months DFS versus 93% for ATBC- (logrank p-test � 0.018). In an<br />

exploratory analysis, two factors were significantly associated with worst<br />

prognosis for ATBC- that were not observed for ATBC� : HR- status (98%<br />

40-months DFS for ATBC� patients versus 84% for ATBC- patients;<br />

logrank p-test � 0.0003) and presence <strong>of</strong> lymphovascular invasion (100%<br />

40-months DFS for ATBC� versus 73% in ATBC- cases; logrank p-test �<br />

0.003). Conclusions: In our seriesATBC is associated with a significant<br />

reduction <strong>of</strong> risk <strong>of</strong> recurrence <strong>of</strong> T1abN0 HER2� tumors. A clear DFS<br />

benefit <strong>of</strong> ATBC was observed in HR- tumors and/or in presence <strong>of</strong><br />

lymphovascular invasion.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


32s Breast Cancer—HER2/ER<br />

602 General Poster Session (Board #9E), Sat, 8:00 AM-12:00 PM<br />

Neoadjuvant bevacizumab (B) and trastuzumab (T) in combination with<br />

weekly paclitaxel (P) as neoadjuvant treatment in HER2-positive breast<br />

cancer: Results from a phase II trial (AVANTHER). Presenting Author:<br />

Maria Fernandez Abad, Centro Integral Oncológico Clara Campal, Madrid,<br />

Spain<br />

Background: B in combination with T has showed meaningful activity in<br />

patients (pts) with metastatic HER2-positive breast cancer. AVANTHER is<br />

a Phase II trial <strong>of</strong> preoperative systemic therapy combining B with T and P<br />

in a weekly regimen in HER2 positive breast cancer to assess safety and<br />

efficacy <strong>of</strong> the combination. Methods: Pts with centrally-confirmed HER2positive<br />

(IHC 3� or FISH positive) breast cancer (stage II or III including<br />

locally advanced) received neoadjuvant chemotherapy (NC) with weekly P<br />

(80mg/m2 /week) for 12 weeks in combination with weekly T (4mg/kg<br />

loading dose and 2 mg/kg maintenance) and B (15mg/kg every 3 weeks) for<br />

4 cycles. After surgery all pts received T (1 year) and liposomal doxorubicin<br />

plus cyclophosphamide every 3 weeks (4 cycles); primary endpoint was rate<br />

<strong>of</strong> pathological complete response (pCR) in breast and axilla. For all<br />

patients, a tissue sample at baseline as well as at surgery was collected for<br />

biomarker analyses. Results: A total <strong>of</strong> 44 pts have been enrolled. Median<br />

tumor size: 3.9 cm. Seven (19.4%) pts had stage IIA; 17 (47.2%) stage<br />

IIB; 8 (22.2%) stage IIIA and 4 (11.1%) stage IIIB. Twenty-one (58.3%)<br />

pts had both positive-hormonal receptors and 10 (27.8%) were hormone<br />

receptor negative. Eight (22.2%) pts had sentinel biopsy before NC, being<br />

negative in 6 (16.7%) cases. Data from surgery (only from 36 pts): pCR was<br />

achieved in 16 (44.4%) pts. Safety and tolerability were good, with rare<br />

adverse events <strong>of</strong> grade �3 [1 (2.8%) episode <strong>of</strong> severe hypertension].<br />

Conclusions: These data show that the combination <strong>of</strong> P with T and B<br />

without an anthracycline for 12 weeks is very effective as NC in HER2<br />

positive breast cancer pts with a high rate <strong>of</strong> pCR and minimal side effects.<br />

604 General Poster Session (Board #9G), Sat, 8:00 AM-12:00 PM<br />

Lapatinib in HER2� early breast cancer: Quality <strong>of</strong> life analysis. Presenting<br />

Author: Frances Boyle, Mater Hospital, North Sydney, Australia<br />

Background: TEACH, a randomized, double-blind, multi-center, phase III<br />

study evaluated the efficacy and safety <strong>of</strong> 12 months lapatinib (N�1571)<br />

versus placebo (N�1576) in women with HER2� EBC. Patients receiving<br />

lapatinib more <strong>of</strong>ten reported treatment-related AEs (87% vs 47%) in the<br />

Intent-to-treat (ITT) population. The comparison <strong>of</strong> recurrence-free survival<br />

was not significant (p�.053). This analysis focuses on QOL in the ITT.<br />

Methods: <strong>Part</strong>icipants on TEACH completed the Short Form-36 version 2<br />

(SF-36v2) at baseline and every 6 mo. for 24 mo. SF-36v2 captures<br />

physical, social, mental, emotional domains <strong>of</strong> patient perceptions; summary<br />

scores <strong>of</strong> physical and mental components are derived from domain<br />

scores using norm based scoring method. Changes in the domain and<br />

summary scores were compared between two groups using ANCOVAs.<br />

Missing post-baseline data were imputed using the last observation carried<br />

forward method. A clinically relevant change in scores was defined as a 3-5<br />

point change. Results: For randomised patients who completed the SF-36,<br />

QOL scores decreased relative to baseline for all domains and summary<br />

scores across all assessments and treatment arms, however the decreases<br />

were small and not clinically meaningful. There were no statistically<br />

significant (p� 0.05) or clinically meaningful differences between arms for<br />

the summary scores relative to baseline (Table). Conclusions: In HER2<br />

positive EBC one year treatment with lapatinib is associated with a<br />

significant increase in AEs but it does not have a significant detrimental<br />

impact on QOL when compared to placebo.<br />

N Changes in physical summary score Changes in mental summary score<br />

Lapatinib –<br />

Lapatinib –<br />

Post-baseline visit Lapatinib Placebo Lapatinib Placebo placebo (95% CI) Lapatinib Placebo placebo (95% CI)<br />

Mo. 6 1092 1283 -0.78 -0.44 -0.33 -2.34 -1.74 -0.61<br />

(-0.81, 0.14)<br />

(-1.28, 0.07)<br />

Mo. 12 1007 1204 -0.84 -0.54 -0.30 -2.74 -2.29 -0.45<br />

(-0.84, 0.23)<br />

(-1.16, 0.26)<br />

Mo.6FU 983 1074 -0.53 -0.87 0.34 -2.22 -1.87 -0.35<br />

(-0.25, 0.92)<br />

(-1.10, 0.40)<br />

Mo. 12 FU 940 988 -0.88 -0.82 -0.06 -3.08 -2.76 -0.31<br />

(-0.65, 0.53)<br />

(-1.11, 0.49)<br />

Mo. 18 FU 834 889 -0.61 -0.89 0.27 -2.38 -2.08 -0.29<br />

(-0.36, 0.91)<br />

(-1.17, 0.58)<br />

Mo. 24 FU 807 824 -0.99 -0.66 -0.33 -2.78 -3.05 0.28<br />

(-1.00, 0.34)<br />

(-0.61, 1.16)<br />

Abbreviation: FU, follow-up.<br />

603 General Poster Session (Board #9F), Sat, 8:00 AM-12:00 PM<br />

Quantitative HER2 levels and steroid receptor expression in primary breast<br />

cancers and in matched brain metastases. Presenting Author: Wojciech<br />

Biernat, Medical University <strong>of</strong> Gdansk, Gdansk, Poland<br />

Background: Breast cancer patients with HER2-positive tumors are at high<br />

risk for brain metastases. In the current study we examined expression <strong>of</strong><br />

ER, PR and HER2 in primary breast tumors and in matched brain<br />

metastases, as changes <strong>of</strong> their levels might reflect modes <strong>of</strong> escape from<br />

therapy. Methods: Fifty-three pairs <strong>of</strong> matched formalin-fixed paraffinembedded<br />

samples from primary breast cancers and brain metastases were<br />

assayed for ER and PR status by immuno-histochemistry, whereas HER2<br />

expression was quantified using the novel HERmark assay. Nuclear staining<br />

<strong>of</strong> ER and PR �10%, and relative fluorescence <strong>of</strong> HER2 �17.8/mm2 were<br />

considered as positive results. Results: HER2 levels in brain metastases<br />

were generally higher than in the primary tumors (p � 3e-6), with a median<br />

increase <strong>of</strong> 1.9-fold (range 0.08 to 199-fold). There were also substantial<br />

differences in ER and PR status between primary tumors and brain<br />

metastases. Loss <strong>of</strong> steroid receptor positivity in brain metastases was more<br />

frequent than its gain (ER: 46% vs. 26%; p � 0.16; PR: 57% vs. 23%; p �<br />

0.044). These changes resulted in a net increase in the number <strong>of</strong><br />

HER2-positive/ER-negative brain metastases, which more than doubled<br />

the proportion <strong>of</strong> primary breast tumors with this phenotype (26% vs. 11%,<br />

respectively; p � 0.08). Additionally, HER2 levels in the primary tumors<br />

significantly correlated with overall survival when stratified by ER status (p<br />

� 0.011). Conclusions: Brain metastases <strong>of</strong> breast cancer show significant<br />

changes in steroid receptor status and in quantitative HER2 levels<br />

compared to matched primary tumors. These data provide a rationale for<br />

future studies and may help in designing treatment strategies that target<br />

the most likely escape pathways <strong>of</strong> breast cancer.<br />

605 General Poster Session (Board #9H), Sat, 8:00 AM-12:00 PM<br />

A randomized phase III double-blinded placebo-controlled trial <strong>of</strong> first-line<br />

chemotherapy and trastuzumab with or without bevacizumab for patients with<br />

HER2/neu-overexpressing metastatic breast cancer (HER2� MBC): A trial <strong>of</strong> the<br />

Eastern Cooperative Oncology Group (E1105). Presenting Author: Carlos L.<br />

Arteaga, Vanderbilt University, Nashville, TN<br />

Background: Pre-clinical and nonrandomized clinical data support synergy <strong>of</strong> the<br />

combination <strong>of</strong> bevacizumab and trastuzumab. We conducted a randomized<br />

double-blinded phase III trial <strong>of</strong> chemotherapy/trastuzumab � bevacizumab to<br />

evaluate efficacy (PFS after treatment initiation) <strong>of</strong> the addition <strong>of</strong> bevacizumab<br />

in first-line treatment <strong>of</strong> HER2� MBC. Methods: Days 1, 8 and 15 paclitaxel �<br />

carboplatin were administered with trastuzumab and either placebo or bevacizumab<br />

10 mg/kg every 2 weeks for a total <strong>of</strong> 6 months. Antibody therapy was<br />

then continued without chemo every 3 weeks, until disease progression or<br />

unacceptable toxicity. Disease assessments were performed every 3 months.<br />

Results: Ninety-six <strong>of</strong> the planned 489 analyzable patients (pts) were accrued<br />

between 2007 and 2009, at which time the study terminated due to poor<br />

accrual. Seventy % <strong>of</strong> pts were post-menopausal, 60% had ER�/HER2� BC.<br />

Median age was 55 years (28-80). Outcomes and toxicities are summarized on<br />

the tables below. Median follow-up was 30 months. Conclusions: The combination<br />

<strong>of</strong> chemotherapy, trastuzumab, and bevacizumab did not significantly<br />

increased toxicity, and was overall safe and well tolerated. No significant<br />

differences in clinical benefit were observed between both treatment arms. Data<br />

on CTCs and Quality <strong>of</strong> Life will be presented at the meeting. This study was<br />

conducted by the Eastern Cooperative Oncology Group (Robert L. Comis, MD,<br />

Chair) and supported in part by Public Health Service Grants CA23318,<br />

CA66636, CA21115, CA32291, CA31946, CA49957, CA27525, CA14548,<br />

CA17145, CA32102, CA25224, CA49.<br />

Outcomes Placebo (%) N�48 Bev (%) N�48<br />

On treatment<br />

Reason for discontinuation<br />

Best response<br />

PFS (# events)<br />

Median PFS (months)<br />

OS (# deaths)<br />

Disease progression<br />

Toxicity<br />

Death<br />

Withdrawal<br />

Other<br />

CR<br />

PR<br />

SD<br />

PD<br />

Not evaluable/unknown<br />

7<br />

60<br />

15<br />

0<br />

4<br />

12<br />

10<br />

42<br />

27<br />

13<br />

8<br />

34<br />

11.1<br />

12<br />

9<br />

38<br />

21<br />

4<br />

4<br />

10<br />

10<br />

42<br />

19<br />

10<br />

18<br />

30<br />

12.2<br />

14<br />

NS<br />

HR (0.95 CI) 0.65<br />

(0.39-1.08) p�0.10<br />

HR (0.95 CI) 1.23<br />

(0.57-2.67) p�0.60<br />

Placebo N�47 Bev N�45<br />

Toxicity<br />

1,2 3 4<br />

Grade (%)<br />

5 1,2 3 4 5<br />

Cardiac troponin T<br />

Hypertension<br />

LV diastolic dysfunction<br />

LV systolic dysfunction<br />

Cardiomyopathy<br />

Wound<br />

Hemorrhage<br />

Proteinuria<br />

Renal failure<br />

Thrombosis<br />

Neutrophils<br />

Infection<br />

2<br />

2<br />

11<br />

2<br />

2<br />

4<br />

4<br />

2<br />

2<br />

6<br />

4<br />

7<br />

9<br />

2<br />

11<br />

7<br />

2<br />

3<br />

4<br />

2<br />

2<br />

2<br />

2<br />

7<br />

2<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


606 General Poster Session (Board #10A), Sat, 8:00 AM-12:00 PM<br />

A neoadjuvant, randomized, open-label phase II trial <strong>of</strong> afatinib (A) versus<br />

trastuzumab (T) versus lapatinib (L) in patients (pts) with locally advanced<br />

HER2-positive breast cancer (BC). Presenting Author: Mothaffar F. Rimawi,<br />

Baylor College <strong>of</strong> Medicine, Houston, TX<br />

Background: A is an oral, irreversible, ErbB Family Blocker. This Phase II<br />

study investigated A, L and T monotherapy in pts with locally advanced<br />

HER2 (ErbB2)-positive BC in a 6-wk preoperative treatment window.<br />

Methods: This international, multi-institutional, open-label, randomized,<br />

Phase II study included pts with HER2-positive locally advanced disease<br />

Stage IIIa–c and inflammatory BC with no evidence <strong>of</strong> distant metastatic<br />

disease, at least one tumor lesion �5 cm in diameter, and an ECOG score<br />

0–1. Pts agreed to fresh tumor biopsies for biomarker analyses at trial<br />

entry, and at 3 and 6 wks <strong>of</strong> treatment. Pts received 50 mg A once daily,<br />

1500 mg L once daily, or T 2 mg/kg weekly after a loading dose <strong>of</strong> 4 mg/kg<br />

for 6 wks. Results: 73 pts were screened and 29 pts randomized and<br />

treated* (10 pts A, 8 pts L, 11 pts T). All pts were female, median age 49<br />

(range 25–88) yrs, 28/29 pts had Stage III disease, 3/29 pts had<br />

inflammatory BC, median tumour size was 6.4 cm, and median time to<br />

treatment was 1.4 months (range 0–3.4). After 6 wks <strong>of</strong> treatment, OR was<br />

assessed using RECIST 1.0 (Table). Best objective response during<br />

treatment was 80%, 75%, and 36.4% in the pts treated with A, L, and T,<br />

respectively. Drug-related AEs assessed by CTCAE v.3.0 occurred in all pts<br />

treated with A, 75% <strong>of</strong> pts treated with L, and 45.5% <strong>of</strong> patients treated<br />

with T (largely GI and skin-related AEs for A and L, and GI and<br />

musculoskeletal AEs for T). Three pts treated with A and one pt with L had<br />

drug-related AEs <strong>of</strong> CTCAE Grade 3. Conclusions: Afatinib showed a high<br />

OR rate in this Phase II trial <strong>of</strong> a 6-wk preoperative treatment window and<br />

compared favorably to two approved agents in HER2-positive BC, albeit at a<br />

slightly higher rate <strong>of</strong> AEs. Further trials will be needed to identify the<br />

potential role <strong>of</strong> A as a single agent or in combination with other agents in<br />

HER2-positive BC.<br />

A L T<br />

<strong>Part</strong>ial response 7 (70.0)* 6 (75.0) 4 (36.4)<br />

Stable disease 2 (20.0) 1 (12.5) 6 (54.5)<br />

Progressive disease 0 (0.0) 1 (12.5) 1 (9.1)<br />

*One pt not included in this table withdrew from the study on Day 11;<br />

end-<strong>of</strong>-study tumor assessment showed partial response. Study closed early due<br />

to slow accrual.<br />

608 General Poster Session (Board #10C), Sat, 8:00 AM-12:00 PM<br />

Correlation <strong>of</strong> quantitative p95HER2 and HER2 protein expression with<br />

pathologic complete response (pCR) in HER2-positive breast cancer<br />

patients (pts) treated with neoadjuvant (NEO) trastuzumab-containing<br />

therapy. Presenting Author: Jose Caetano Villasboas, University <strong>of</strong> Miami<br />

Sylvester Comprehensive Cancer Center, Miami, FL<br />

Background: Elevated p95 [HER2-M611-CTF (carboxy-terminal-fragment)<br />

also known as p110 or p95HER2] expression has been correlated with poor<br />

outcomes in HER2� pts with metastatic breast cancer treated with<br />

trastuzumab (T); however, limited data have been presented on the<br />

correlation between p95 and pCR to T in the NEO setting, where p95 was<br />

measured by immunohistochemistry. In the current study, we sought to<br />

determine whether quantitative p95 and HER2 expression correlated with<br />

pCR in pts treated with T � chemotherapy in the NEO setting. Methods:<br />

HER2 expression (H2T) was quantified by HERmark in 47 breast tumors<br />

using formalin-fixed, paraffin-embedded sections. Tissue remained in 40<br />

cases to measure p95 by VeraTag and compare to a previously published<br />

cut<strong>of</strong>f (Clin Cancer Res 16:4226, 2010). pCR data were available for 45<br />

cases. pCR was defined as the absence <strong>of</strong> invasive disease in the breast.<br />

Results: The overall pCR rate was 46.7% (ER�: 14.3% vs. ER-: 75%;<br />

Wilcoxon rank p�0.0001) and was significantly associated with higher<br />

H2T levels (p�0.02). In ER- subjects (N�24), no difference in H2T levels<br />

was observed between pCR vs non-pCR groups [median H2T�111.5 (IQR<br />

63.4-162.2) vs 150.5 (IQR 43 – 226.2), respectively; p�0.721]. However,<br />

within the ER� group (N�21), H2T levels were significantly higher in<br />

the pCR group vs non-pCR group [median H2T�254 (IQR 181.5-584.5) vs<br />

37.3 (IQR 16.4-89); p�0.024]. Using multivariate logistic regression,<br />

increasing log(H2T) (p � 0.011), ER-negativity (p � 0.027) and low p95<br />

(p � 0.074) were found to correlate or trend with pCR. Conclusions: pCR<br />

was significantly associated with high H2T expression in ER� HER2�<br />

breast cancer pts who received NEO therapy with T � chemotherapy. A<br />

trend towards pCR was seen in tumors that had low p95. These data<br />

suggest that quantitative H2T and p95 may provide additional information<br />

on response to T-based regimens in breast cancer, particularly ER� breast<br />

cancer. Additional investigation into the possible relationship between<br />

quantitative levels <strong>of</strong> HER2 and p95 expression and T response in the NEO<br />

setting in larger cohorts is warranted.<br />

Breast Cancer—HER2/ER<br />

33s<br />

607 General Poster Session (Board #10B), Sat, 8:00 AM-12:00 PM<br />

Elevated pretreatment serum activin A and progression-free and overall<br />

survival in trastuzumab-treated metastatic breast cancer. Presenting Author:<br />

Allan Lipton, Penn State Hershey Medical Center, Hershey, PA<br />

Background: About half <strong>of</strong> HER2-positive metastatic breast cancer patients<br />

will respond t<strong>of</strong>irst-line trastuzumab-containing therapy, but most will<br />

progress within a year. Trastuzumab resistance remains a vexing clinical<br />

problem, and better predictive and prognostic biomarkers are needed.<br />

Activin A is a TGF-B superfamily member and regulates cell proliferation,<br />

apoptosis, differentiation, and immune response. Methods: Serum activin A<br />

was measured using ELISA (R&D Systems, Minneapolis, MN) in 60<br />

metastatic breast cancer patients before starting first-line trastuzumabcontaining<br />

therapy. Progression-free survival (PFS) and overall survival<br />

(OS) were analyzed using the Kaplan-Meier method and Cox modeling with<br />

both continuous and dichotomous (median) serum activin A analyses.<br />

Results: Pretreatment serum activin A levels averaged 2376 pg/ml, and had<br />

a median <strong>of</strong> 629 pg/ml and 25th and 75th quartile values <strong>of</strong> 406 and 1791<br />

pg/ml, respectively. Patients who were hormone receptor negative had<br />

significantly higher activin A levels (median 1287 vs 450 pg/ml, p�0.002).<br />

Higher serum activin A was significant on a continuous basis for predicting<br />

reduced PFS to first-line trastuzumab-containing therapy (p�0.003), and<br />

for predicting shorter OS (p�0.0001). When analyzed using a dichotomous<br />

(median) cutpoint, the elevated serum activin A cohort had a significantly<br />

reduced PFS (HR 2.79, p �0. 002) (median 6.6 mo vs. 31.1 mo) and OS<br />

(HR 5.24, p �0.0001) (median 19.6 mo vs. median not reached). In<br />

multivariate analysis for PFS with other covariates (age, line <strong>of</strong> therapy, CA<br />

15-3, and hormone receptor status), activin A was the only significant<br />

covariate (p�0.021). In multivariate analysis for OS, activin A (p�0.002)<br />

and CA 15-3 (p�0.03) remained significant as prognostic factors.<br />

Conclusions: Elevated pretreatment serum activin A predicts reduced PFS<br />

and overall survival in metastatic breast cancer patients treated with<br />

first-line trastuzumab-containing therapy. Activin A deserves further study<br />

as an adverse biomarker, and to select patients most likely to respond to<br />

activin A-targeted therapy.<br />

609 General Poster Session (Board #10D), Sat, 8:00 AM-12:00 PM<br />

Incidence and risk <strong>of</strong> central nervous system (CNS) metastases as a site <strong>of</strong><br />

first recurrence in patients (pts) with HER2-positive (HER2�) breast<br />

cancer treated with adjuvant trastuzumab (T). Presenting Author: Erin<br />

Macrae Olson, The Ohio State University Comprehensive Cancer Center,<br />

Stefanie Spielman Comprehensive Breast Center, Columbus, OH<br />

Background: T is the mainstay <strong>of</strong> adjuvant therapy in pts with HER2�<br />

breast cancer. CNS disease as the site <strong>of</strong> first relapse after exposure to<br />

adjuvant T has been reported, although the overall incidence and relative<br />

risk (RR) <strong>of</strong> this remains unclear. We performed an up-to-date metaanalysis<br />

to determine the risk <strong>of</strong> CNS metastases as the first site <strong>of</strong><br />

recurrence in pts with HER2� breast cancer who received adjuvant T.<br />

Methods: Pubmed databases were searched for articles from 1966 to 2011.<br />

Abstracts presented at the <strong>American</strong> <strong>Society</strong> <strong>of</strong> <strong>Clinical</strong> Oncology and the<br />

San Antonio Breast Cancer Symposium were also searched for relevant<br />

clinical trials. Eligible studies include randomized trials with adjuvant T<br />

administered for 1 year in pts with HER2� breast cancer who reported CNS<br />

metastases as first site <strong>of</strong> disease recurrence. Statistical analyses were<br />

conducted to calculate the summary incidence, RR, and 95% CIs using<br />

fixed effects inverse variance models. Results: A total <strong>of</strong> 9,020 pts were<br />

included. The incidence <strong>of</strong> CNS metastases as first site <strong>of</strong> disease<br />

recurrence in HER2� pts receiving adjuvant T was 2.56% (95% CI 2.07%<br />

to 3.01%) compared to 1.94% (95% CI: 1.54% to 2.38%) in HER2� pts<br />

who did not receive adjuvant T. The RR <strong>of</strong> CNS as first site <strong>of</strong> relapse in<br />

T-treated pts was 1.35 (95% CI 1.02 to 1.78, p�0.038) compared with<br />

control arms without T therapy. In subgroup analyses, there was no<br />

significant difference in CNS incidence or risk between pts treated with<br />

concurrent versus sequential T (p�0.29), weekly versus every 3 week T<br />

(p�0.56), and no difference in the T groups between studies due to<br />

median follow up time in years (p�0.68). No evidence <strong>of</strong> publication bias<br />

was observed (Q�1.78; P�0.62; I2 � 0.0%). Conclusions: This is the<br />

largest report to date demonstrating that adjuvant T is associated with an<br />

increased risk <strong>of</strong> CNS metastases as a site <strong>of</strong> first recurrence in HER2�<br />

breast cancer pts.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


34s Breast Cancer—HER2/ER<br />

610 General Poster Session (Board #10E), Sat, 8:00 AM-12:00 PM<br />

Phase I trial <strong>of</strong> pegylated liposomal doxorubicin and lapatinib in the<br />

treatment <strong>of</strong> metastatic breast cancer (MBC): Final results. Presenting<br />

Author: Mary E. Cianfrocca, Banner M. D. Anderson Cancer Center, Gilbert,<br />

AZ<br />

Background: Liposomal formulations including pegylated liposomal doxorubicin<br />

(PLD) were developed to improve the therapeutic index <strong>of</strong> anthracyclines<br />

(A). Lapatinib (L) is a selective, highly competitive inhibitor <strong>of</strong> ErbB1<br />

and ErbB2 tyrosine kinases. Conventional doxorubicin plus trastuzumab<br />

was effective but with unacceptable cardiac toxicity. PLD plus L may be<br />

effective with less cardiac risk. Methods: This is a phase I, dose-escalation<br />

trial <strong>of</strong> PLD 20, 30, 45 and 60 mg/m2 IV every 4 weeks (maximum <strong>of</strong> 8<br />

doses) and L, 1500 mg po daily until progression in patients (pts) with<br />

MBC. ErbB2 positivity was not required. Prior chemotherapy, endocrine<br />

therapy and trastuzumab were allowed. A subsequent amendment allowed<br />

prior L. Prior A use was limited to 240 mg/m2 <strong>of</strong> doxorubicin or 600 mg/m2 <strong>of</strong> epirubicin. Concomitant CYP3A4 inducers/ inhibitors were not allowed.<br />

A left ventricular ejection fraction (LVEF) <strong>of</strong> � 50% was required. The<br />

primary objective was to evaluate the safety (particularly cardiac), tolerability<br />

and feasibility <strong>of</strong> PLD and L. Results: 23 pts (PLD: 20 mg/m2 - 4 pts; 30<br />

mg/m2 - 3 pts; 45 mg/m2 – 13 pts; 60 mg/m2- 3 pts) have been treated;<br />

total <strong>of</strong> 73 treatment cycles. Dose-limiting toxicity (DLT) was not reached.<br />

One pt had an LVEF drop to � 50% after 4 cycles accompanied by a<br />

pericardial effusion due to progressive disease. Treatment-related grade<br />

III/IV adverse events included: 4 pts with hand-foot-syndrome (HFS), 2 pts<br />

each with leukopenia, infection, and skin changes, 1 pt each with pain,<br />

fatigue, diarrhea, mucositis, hypoalbuminemia, anemia, cough, pleural<br />

effusion, and edema. Grade 3 HFS occurred in 2 <strong>of</strong> 3 pts in the 60 mg/m2 cohort. Response data in 21 evaluable pts: 4 PR, 5 SD, and 12 PD.<br />

Preliminary pharmacokinetic (PK) analyses (7 pts) indicate L has no effect<br />

on PLD (45 mg/m2 ) concentrations, but L concentrations were approximately<br />

2-fold higher the day <strong>of</strong> PLD dosing. Conclusions: In 23 pts treated,<br />

PLD plus L was well tolerated with manageable toxicities and no treatmentrelated<br />

cardiac toxicity. DLT was not reached however grade 3 HFS<br />

occurred in 2 <strong>of</strong> 3 pts in the 60 mg/m2 cohort. Preliminary PK analyses<br />

demonstrate no effect <strong>of</strong> L on PLD, but an effect <strong>of</strong> PLD on L the day <strong>of</strong> PLD<br />

dosing.<br />

612 General Poster Session (Board #10G), Sat, 8:00 AM-12:00 PM<br />

Survival outcomes in HER2-positive invasive lobular breast carcinoma.<br />

Presenting Author: Carlos Hernando Barcenas, University <strong>of</strong> Texas M. D.<br />

Anderson Cancer Center, Houston, TX<br />

Background: HER2-positive (HER2�) invasive lobular breast carcinoma<br />

(ILC) is a rare entity and survival outcomes are not well characterized.<br />

Methods: We retrospectively searched for breast cancer patients treated at<br />

MD Anderson Cancer Center between 1992 and 2010 with a diagnosis <strong>of</strong><br />

early stage (I-III) HER2�ILC or with HER2� invasive mixed ductal/lobular<br />

carcinoma (MIX). With the purpose <strong>of</strong> comparing survival outcomes, we<br />

looked for patients with a diagnosis <strong>of</strong> HER2-negative [HER2(-)] ILC,<br />

HER2(-)MIX, and HER2� invasive ductal carcinomas (IDC). We obtained<br />

data on demographics, estrogen (ER) and progesterone (PR) receptor<br />

status, tumor grade, chemotherapy received and survival status. A multivariate<br />

analysis using a Cox proportional hazards regression was performed to<br />

compare disease-free survival (DFS) and overall survival (OS) between the<br />

following groups: 1) HER2�ILC vs. HER2(-)ILC; 2) HER2�MIX vs.<br />

HER2(-)MIX; 3) HER2�ILC vs. HER2�MIX; 4) HER2�ILC vs. HER2�IDC;<br />

5) HER2�MIX vs. HER2�IDC. Results: The number <strong>of</strong> patients identified<br />

for each group was: HER2�ILC (N� 67), HER2(-)ILC (N� 1260),<br />

HER2�MIX (N� 92), HER2(-)MIX (N� 932), HER2�IDC (N� 3080).<br />

Specifically for both the HER2�ILC and HER2�MIX groups, 24% patients<br />

were stage I, 43% stage II and 33% stage III. The median age at diagnosis<br />

was 53 years (y) in HER2�ILC, and 49 y in HER2�MIX. The ER/PR were<br />

both negative in 16% <strong>of</strong> HER2�ILC patients, and in 25% <strong>of</strong> HER2�MIX.<br />

While 80% <strong>of</strong> patients in these two groups received chemotherapy, 36% <strong>of</strong><br />

HER2�ILC patients received trastuzumab compared to 50% <strong>of</strong> HER2�MIX.<br />

The median follow up was 60 months in the HER2�ILC group and 53<br />

months in the HER2�MIX group. DFS and OS were not statistically<br />

different between all groups compared. However, the subgroup <strong>of</strong> ER/PR<br />

negative HER2�MIX patients had a better OS compared to ER/PR/HER2<br />

negative (triple negative) MIX patients (Hazard ratio � 0.4, 0.2 – 0.9, p�<br />

0.026). Conclusions: HER2�ILC and HER2�MIX are rare clinical entities<br />

and their survival outcomes may not differ from the more common<br />

HER2(-)ILC, HER2(-)MIX and HER2�IDC. ER/PR negative HER2�MIX<br />

may have an OS advantage over triple negative MIX tumors.<br />

611 General Poster Session (Board #10F), Sat, 8:00 AM-12:00 PM<br />

A phase I dose-escalation study evaluating weekly paclitaxel with neratinib and<br />

trastuzumab in women with metastatic HER2-positive breast cancer, NSABP<br />

FB-8. Presenting Author: Rachel Catherine Jankowitz, National Surgical Adjuvant<br />

Breast and Bowel Project and the University <strong>of</strong> Pittsburgh Medical Center,<br />

Magee-Womens Hospital, Pittsburgh, PA<br />

Background: Dual blockade <strong>of</strong> ErbB receptors combined with chemotherapy<br />

compared with single-agent blockade improves efficacy in HER2-positive disease.<br />

FB-8 is single arm, Phase I dose-escalation study to determine safety and<br />

tolerability <strong>of</strong> weekly paclitaxel (P), the irreversible tyrosine kinase inhibitor,<br />

neratinib (N) and trastuzumab (T) in women with metastatic, HER2-positive<br />

breast cancer (MBC). Methods: Women with measurable or non-measurable MBC<br />

(ECOG PS 0-2)received P (80 mg/m2 IV days 1, 8, 15 <strong>of</strong> 28-day cycle), T (4<br />

mg/kg IV, then 2 mg/kg IV weekly), and N (oral, daily). N was dose-escalated<br />

using 3�3 design at 120 mg, 160 mg, and 240 mg, dose-limiting toxicity (DLT)<br />

determined in cycle 1. All patients (pts) received prophylactic anti-diarrheals.<br />

Results: All pts were taxane and trastuzumab pre-treated, with mean <strong>of</strong> 4 prior<br />

regimens. At N�120 mg, there was 1 DLT, grade (gr) 3 diarrhea with<br />

dehydration. Three more pts were enrolled; none experienced DLT. At N�160<br />

mg, there were no DLTs. At N�240 mg, 2 <strong>of</strong> 3 pts had DLTs: one gr 3 diarrhea<br />

and dehydration, one gr 3 diarrhea, dehydration, and gr 3 mucositis. After<br />

de-escalation to N�160 mg, 3 more pts were added. If N�160 mg is well<br />

tolerated, a 200 mg cohort is planned. 12 pts completed cycle 1. Efficacy data<br />

are available on 10: CR 1, PR 3, SD 1 (8�months). One pt* had resolution <strong>of</strong><br />

non-target disease (skin and breast mass). 4 pts were <strong>of</strong>f-study before the first<br />

scan (DLT 3 and progressive disease 1) and are considered non-responders (NR).<br />

The most frequent grade 3-4 adverse event was diarrhea, 3 pts. Conclusions:<br />

Dual anti-HER blockade with N, T combined with P is tolerable and highly active<br />

in pts pre-treated with anti-HER2 agents and chemotherapy. This combination<br />

will be studied in a phase II neoadjuvant breast cancer trial (NSABP FB-7). We<br />

thank Pfizer, Inc and Puma Biotechnology, Inc for their support.<br />

Dose<br />

level (mg) Prior anthracycline Prior lapatinib Prior TDM1 Cycle<br />

no. Best response<br />

120 Yes Yes No 8� SD<br />

120 Yes Yes Yes 4 NR<br />

120 No No No 1 NR<br />

120 No No Yes 5� CR<br />

120 No No No 5� PR<br />

120 No Yes No 5� PR<br />

160 Yes Yes No 2 NR<br />

160 No Yes Yes 3� Non-measurable*<br />

160 No No No 3� PR<br />

240 No Yes No 1 NR<br />

240 No Yes Yes 2� Pending<br />

240 Yes Yes No 1 NR<br />

613 General Poster Session (Board #10H), Sat, 8:00 AM-12:00 PM<br />

Estrogen receptor (ER), Ki-67, p27Kip1 , and histologic grade as predictors<br />

<strong>of</strong> pathologic complete response (pCR) in patients with HER2-positive<br />

breast cancer treated with neoadjuvant chemotherapy (NAC) using fluorouracil,<br />

epirubicin, and cyclophosphamide (FEC), taxanes, and trastuzumab.<br />

Presenting Author: Sasagu Kurozumi, Saitama Cancer Center, Saitama,<br />

Japan<br />

Background: NAC with taxanes and FEC concurrently with trastuzumab is a<br />

potent regimen in patients with HER2-positive breast cancer (BC). Several<br />

studies revealed high pCR rates in BC patients treated with this regimen;<br />

however, predictive factors and a prognostic effect <strong>of</strong> pCR have been still<br />

unclear. In this study, we analyzed several factors including p27Kip1 (cyclin-dependent kinase inhibitor acting as tumor suppressor) for correlation<br />

with pCR. We also evaluated differences in recurrence-free survival<br />

(RFS) or overall survival (OS) between patients with pCR and non-pCR, and<br />

with positive and negative nodes after NAC. Methods: Our study included<br />

129 Japanese women with invasive, HER2-positive BC who received 12<br />

cycles <strong>of</strong> paclitaxel or 4 cycles <strong>of</strong> docetaxel followed by 4 cycles <strong>of</strong> FEC-75<br />

with concomitant trastuzumab for 24 weeks. We analyzed the correlation <strong>of</strong><br />

pCR (no invasive lesions in the breast) and nodal status after NAC with RFS<br />

and OS, and analyzed the baseline expressions <strong>of</strong> ER, Ki-67, and p27Kip1 ,<br />

and histological grade for correlation with pCR. Positive or high expression<br />

was defined by nuclear labeling index: ER �10%, p27Kip1 �75%, Ki-67<br />

�30%. Results: In 129 patients, pCR was found in 85 (66%). Patients with<br />

pCR after NAC had significantly better RFS than those without pCR<br />

(median follow-up: 41 months). Furthermore, patients with pathologically<br />

negative nodes after NAC had significantly better OS than those with<br />

pathologically positive nodes. Negative ER (79% vs. 40%), high Ki-67<br />

(72% vs. 47%), low p27Kip1 (71% vs. 50%), and histological grade 3 (70%<br />

vs. 39%) were significant predictors <strong>of</strong> pCR. Conclusions: In patients with<br />

HER2-positive BC, this regimen was effective achieving the high pCR rate.<br />

pCR and pathologically negative nodes after NAC were predictive <strong>of</strong> RFS<br />

and OS, respectively. The expressions <strong>of</strong> ER, Ki-67, and p27Kip1 , and<br />

histological grade at baseline were predictive <strong>of</strong> pCR. p27Kip1 , a new<br />

predictor <strong>of</strong> pCR after NAC needs to be further analyzed.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


614 General Poster Session (Board #11A), Sat, 8:00 AM-12:00 PM<br />

Quantitative HER2 measurement and PI3K mutation pr<strong>of</strong>ile in matched<br />

primary and metastatic breast cancer tissues. Presenting Author: Weidong<br />

Huang, Monogram Biosciences, South San Francisco, CA<br />

Background: HER2 status <strong>of</strong> primary breast cancer (PBC) is routinely used<br />

to determine systemic treatment for metastatic breast cancer (MBC)<br />

patients. Discordance rates <strong>of</strong> HER2 status between PBC and MBC range<br />

from 5.5% to 29% based on published meta-analyses. The clinical benefit<br />

<strong>of</strong> re-assessing HER2 in MBC tissues remains controversial. In this study,<br />

we measured quantitative HER2 expression in matched PBC and MBC<br />

tissues and correlated changes <strong>of</strong> HER2 with mutations in the catalytic<br />

domain <strong>of</strong> PI3 kinase(PIK3CA). Methods: Total HER2 protein expression<br />

(H2T) was quantified by the HERmark assay in 41 matched PBC and MBC<br />

formalin-fixed, paraffin-embedded specimens. PIK3CA mutation status in<br />

exons 9 (E545K and E542K) and 20 (H1047R) was determined using a<br />

validated pyrosequencing assay. Results: MBC samples included 5 lymph<br />

node, 13 viscera, 6 brain, and 17 s<strong>of</strong>t tissue lesions (N�41). 27 (66%)<br />

cases showed higher H2T in MBC than in matched PBC; and 14 (34%)<br />

cases had higher H2T in PBC than in matched MBC, indicating an overall<br />

increase <strong>of</strong> H2T in matched MBC lesions (fold change 0.25-17.57;<br />

p�0.005, paired Wilcoxon rank sum test). HER2 positive conversion<br />

(HERmark negative/equivocal in PBC, but positive in matched MBC) was<br />

found in 6 (15%) cases, while HER2 negative conversion (HERmark<br />

positive in PBC, but negative/equivocal in matched MBC) was seen in 2<br />

(5%) cases. HER2 status was unchanged in 33 (80%) cases. PIK3CA<br />

mutations were detected in 13 (32%) <strong>of</strong> PBC and 19 (46%) <strong>of</strong> MBC<br />

samples. Among the HER2 positive conversion cases, PIK3CA mutation<br />

was identified in 50% (3/6) PBC and 67% (4/6) MBC, compared to 0%<br />

(0/2, PBC or MBC) in the HER2 negative conversion cases. Among cases<br />

with unchanged HER2 status, PIK3CA mutation was observed in 30%<br />

(10/33) PBC and 42% (14/33) MBC. Conclusions: Quantitative HER2<br />

assessment revealed a 20% discordance in HER2 status between matched<br />

PBC and MBC tissues, with more frequent conversion from low HER2 in<br />

PBC to high HER2 in MBC. PIK3CA mutation was observed more frequently<br />

in patients who converted from HER2 negative PBC to HER2 positive MBC.<br />

These results suggest that re-assessment <strong>of</strong> biomarkers in MBC tissues may<br />

better inform the selection <strong>of</strong> therapeutic options for patients with MBC.<br />

616 General Poster Session (Board #11C), Sat, 8:00 AM-12:00 PM<br />

Can MRI accurately identify which patients with operable breast cancer will<br />

have a pathologic complete response after neoadjuvant therapy? Presenting<br />

Author: Carolyn Nessim, Sunnybrook Odette Cancer Centre, University <strong>of</strong><br />

Toronto, Toronto, ON, Canada<br />

Background: With the introduction <strong>of</strong> targeted therapy based on tumor<br />

subtypes, an increasing number <strong>of</strong> patients that receive neoadjuvant<br />

chemotherapy achieve a pathologic complete response (pCR). Previous<br />

studies have shown that the accuracy <strong>of</strong> MRI is poor at predicting the<br />

response to neoadjuvant chemotherapy in locally advanced and <strong>of</strong>ten<br />

non-resectable breast cancers, where the rate <strong>of</strong> pCR is low. The purpose <strong>of</strong><br />

this study is to evaluate MRI’s ability to predict a pCR in operable breast<br />

cancers after neoadjuvant therapy. Methods: All patients enrolled in the<br />

NSABP B-40, B-41, FB-5 and FB-6 protocols in a single tertiary care<br />

centre, that had an MRI done before and after neoadjuvant therapy were<br />

reviewed. A radiologist, blinded to the pathology results, interpreted the<br />

pre- and post- treatment MRI’s and made a prediction as to whether or not<br />

patients would have a pCR. In this study, a true negative was defined as a<br />

reading <strong>of</strong> a complete response on MRI that was confirmed as a pCR on final<br />

pathology. pCR was defined as having no residual invasive or in situ disease<br />

in the breast. Results: 129 women with a median age <strong>of</strong> 51 years were<br />

identified. 90% had invasive ductal carcinoma; 8% had invasive lobular.<br />

58% were ER�, 21% were triple negative and 21% were Her2�. 16% <strong>of</strong><br />

patients had a pCR. 25% <strong>of</strong> patients had no residual invasive cancer in the<br />

breast. pCR rates for ER� tumors was 5%, triple negative 37%, and Her2�<br />

26%. 19% <strong>of</strong> patients that had a pCR had a total mastectomy. The<br />

sensitivity and specificity <strong>of</strong> MRI for predicting residual disease were 88%<br />

and 52% respectively. The positive predictive value was 90% and the<br />

negative predictive value was 46% with an accuracy <strong>of</strong> 82%. Conclusions:<br />

MRI has limited value for determining which patients had a pCR after<br />

neoadjuvant chemotherapy, even in operable breast cancers. When residual<br />

disease is suspected on MRI, it is unlikely that a pCR has been<br />

achieved. Surgical excision following neoadjuvant therapy remains the gold<br />

standard to identify which patients have achieved a pCR. Other modalities<br />

will need to be used in order to accurately determine which patients would<br />

be eligible for studies evaluating non operative management following<br />

neoadjuvant therapy.<br />

Breast Cancer—HER2/ER<br />

35s<br />

615 General Poster Session (Board #11B), Sat, 8:00 AM-12:00 PM<br />

Long-term follow-up (FU) <strong>of</strong> patients with durable complete response<br />

(DCR) after chemotherapy (CT) and trastuzumab (T) for HER2-altered<br />

(HER2�) metastatic breast cancer (MBC). Presenting Author: Giuseppe<br />

Gullo, St Vincent’s University Hospital, Dublin, Ireland<br />

Background: Durable CT-induced complete response (DCR) <strong>of</strong> MBC is<br />

reported only anecdotally. The addition <strong>of</strong> monoclonal antibody T to<br />

conventional CT results in significant increase in response rates and<br />

prolonged survival for patients (pts) with HER2� MBC. Though CRs are<br />

reported in most phase II and III clinical trials <strong>of</strong> CT�T, there is lack <strong>of</strong> data<br />

about clinical features and long-term outcome <strong>of</strong> these pts. Methods: We<br />

retrospectively reviewed all pts with HER2� MBC treated at our Institutions<br />

with any CT�T and identified cases who achieved DCR. HER2<br />

positivity was defined as 3�score at immunohistochemistry and/or amplification<br />

at FISH test. DCR was defined as a CR according to RECIST 1.0<br />

criteria lasting �36 months. Results: We identified 13 pts with HER2�<br />

MBC who met criteria for DCR following CT�T. Their characteristics are:<br />

median age: 56yrs (range 30-65), stage at diagnosis: M0 54%/M1 46%,<br />

histology: ductal 84%/mixed ductal-lobular 8%/unknown 8%, tumour<br />

grade: G3 54%/G2 23%/unknown 23%, oestrogen receptors (ER): neg<br />

62%/pos 38%, sites <strong>of</strong> metastatic disease: liver only 54%/other visceral<br />

15%/bone and s<strong>of</strong>t tissues only 31%, chemotherapy regimen:<br />

taxane�carboplatin 54%/single-agent taxane 23%/docetaxel�lapatinib<br />

15%, capecitabine 8%. Metastatic disease was biopsy-proven in 54% <strong>of</strong><br />

pts. All pts received maintenance T. Median follow-up (FU) is 6.5 yrs (range<br />

3.0-11.6). Median duration <strong>of</strong> T was 59 months (17-121). At database<br />

cut-<strong>of</strong>f date 12 pts (92%) are alive. Five pts (38%) had disease relapse (3<br />

on maintenance T, 2 after T discontinuation), 2 <strong>of</strong> whom had radical<br />

surgery. In 8 pts (62%) disease has not relapsed (characteristics: all<br />

1st-line therapy, ER negative 75%, liver only distant disease 75%).<br />

Conclusions: This is the largest series to our knowledge analyzing long-term<br />

FU <strong>of</strong> HER2� MBC pts with DCR following CT�T. DCR pts appear to have<br />

prolonged survival and a substantial subgroup <strong>of</strong> them have no further<br />

disease relapse. These pts are usually treated in the 1st-line setting, have<br />

more frequently ER-negative disease and liver-only metastases. We are<br />

currently investigating the molecular pr<strong>of</strong>ile <strong>of</strong> this subset <strong>of</strong> pts.<br />

617 General Poster Session (Board #11D), Sat, 8:00 AM-12:00 PM<br />

Neuromedin U: A potential predictive biomarker for HER2-targeted drugs.<br />

Presenting Author: Sweta Rani, School <strong>of</strong> Pharmacy and Pharmaceutical<br />

Sciences and Molecular Therapeutics for Cancer Ireland, Trinity College<br />

Dublin, Dublin, Ireland<br />

Background: Not all HER2-positive breast cancer patients respond to<br />

HER2-targeted drugs and some, who initially respond, subsequently<br />

relapse. There is a need to identify biomarkers for improved patient<br />

selection. Here we aimed to identify gene expression changes associated<br />

with response to HER2-targeted drugs. Methods: To identify mRNA changes<br />

associated with resistance, whole genome microarrays were used to pr<strong>of</strong>ile<br />

conditioned medium (CM) from HER2-positive cell lines (SKBR3,<br />

HCC1954) and their lapatinib (L)-resistant variants (SKBR3-LR , HCC1954-<br />

LR ). Neuromedin U (NmU) over-expression, identified in this way, was<br />

confirmed in the L-resistant cells and corresponding CM by qPCR and<br />

ELISA. Pooled data from 21 published expression datasets (n�3489<br />

patients) was mined to relate NmU mRNA to tumour subtype and patients’<br />

outcome. NmU cDNA and siRNAs were used to over-express and knockdown<br />

expression <strong>of</strong> NmU, respectively, prior to assessing it’s association<br />

with response to L, Trastuzumab (T) and Neratinib (N). Results: Analysis <strong>of</strong><br />

the tumour data showed NmU expression to be particularly associated with<br />

poor outcome for patients with HER2-positive tumours (p�0.000005). In<br />

cell lines, we identified NmU mRNA and protein to be at significantly<br />

higher levels in L-resistant cells and corresponding CM, compared to that <strong>of</strong><br />

L-sensitive SKBR3 and HCC1954. This trend was observed for T-resistant<br />

and N-resistant SKBR3 cells (SKBR3T , SKBR3N ) and their CM, compared<br />

to sensitive parent SKBR3 cells and CM. NmU cDNA over-expression in<br />

SKBR3 and HCC1954 increased resistance to L, T and N. Knock-down <strong>of</strong><br />

NmU endogenous levels in SKBR3-LR and innately L-resistant MDA-MB-<br />

361 and T47D sensitised these cells to L, T and N. Further analysis <strong>of</strong> our<br />

NmU over-expressing and knock-down cells indicated NmU to also be<br />

associated with increased motility, invasion and anoikis resistance.<br />

Conclusions: NmU expression is prognostic <strong>of</strong> poor outcome in HER2positive<br />

breast tumours. In cell line models, NmU over-expression is<br />

associated with resistance to L, T and N. Taken together, we propose NmU<br />

as a possible prognostic and predictive biomarker for HER2-positive<br />

cancers, with potential also as a co-target to help circumvent resistance to<br />

these drugs. [SFI: 08/SRC/B1410]<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


36s Breast Cancer—HER2/ER<br />

618 General Poster Session (Board #11E), Sat, 8:00 AM-12:00 PM<br />

Use <strong>of</strong> the PRO Onc Assay (Prometheus) to measure HER2 overexpression/<br />

activation in circulating tumor cells (CTCs) in women with HER2-negative<br />

metastatic breast cancer (MBC): A Sarah Cannon Research Institute (SCRI)<br />

trial. Presenting Author: John D. Hainsworth, SCRI/Tennessee Oncology,<br />

PLLC, Nashville, TN<br />

Background: HER2- targeted therapy has markedly improved the treatment<br />

<strong>of</strong> women with HER2- positive breast cancer. FISH testing for HER2<br />

overexpression is the most reliable assay method; treatment decisions are<br />

usually based on HER2 expression <strong>of</strong> the breast primary tumor. Accurate<br />

determination <strong>of</strong> HER2 status by testing CTCs may improve patient<br />

management by allowing ongoing assessment <strong>of</strong> HER2 status during<br />

treatment and/or identifying additional patients (pts) for HER2- targeted<br />

therapy. Methods: The PRO Onc assay uses a multiplexed immunoassay<br />

format to provide expression and activation pr<strong>of</strong>iling <strong>of</strong> HER2 and other<br />

signal transduction molecules. The assay can detect overexpression and<br />

activation (phosphorylation) <strong>of</strong> HER2, with sensitivity at a single CTC level.<br />

We obtained blood specimens from 57 women with metastatic HER2negative<br />

breast cancer who were receiving standard chemotherapy. In these<br />

women, HER2 status had previously been determined by FISH (32) or 0 –<br />

1� IHC testing (25). All specimens were tested for the presence <strong>of</strong> CTCs;<br />

the PRO Onc assay was performed when CTCs were identified. The HER2<br />

overexpression cut<strong>of</strong>f (�3.0 CU) was based on the reference value (average<br />

� 4SD) determined by analyzing normal blood obtained from 42 healthy<br />

donors. Results: 31 <strong>of</strong> 57 pts (54%) had CTCs identified. 6 <strong>of</strong> 31 pts (19%)<br />

had HER2 overexpressed, and 4 pts (13%) had HER2 activation without<br />

overexpression. 7 <strong>of</strong> 10 pts with HER2 overexpression/activation by PRO<br />

Onc Assay were FISH- negative; 3 were IHC- negative. Since initial HER2<br />

determination, these 10 pts had received a median <strong>of</strong> 3 chemotherapy<br />

regimens (range1-5).Conclusions: When CTCs were present, the PRO Onc<br />

assay identified HER2 overexpression or activation in 31% <strong>of</strong> women with<br />

HER2- negative MBC. The therapeutic significance <strong>of</strong> this finding is<br />

unknown. Since the predetermined threshold <strong>of</strong> �10% HER2- positive<br />

assay results was exceeded, this trial will proceed to a planned second<br />

portion, in which women with HER2- negative MBC (by FISH testing) and<br />

HER2 overexpression or activation in CTCs will receive a trial <strong>of</strong> HER2targeted<br />

therapy.<br />

620 General Poster Session (Board #11G), Sat, 8:00 AM-12:00 PM<br />

Use <strong>of</strong> HER2 score correction for putative chromosome 17 (Chr-17)<br />

aneusomy to increase eligibility for anti-HER2 therapy. Presenting Author:<br />

Eugen C. Minca, Cleveland Clinic Foundation, Cleveland, OH<br />

Background: HER2 amplification or overexpression status directs therapy<br />

choice in breast carcinoma. HER2/Chr-17 ratio is commonly assessed by<br />

fluorescence in-situ hybridization (FISH) using a CEP17 centromeric<br />

reference probe and ASCO/CAP scoring criteria. However, �-centromeric<br />

reference probes may underestimate true HER2 status in cases with<br />

para-centromeric amplification. Here we present comprehensive algorithmic<br />

testing <strong>of</strong> an alternative Chr-17 reference locus for resolution <strong>of</strong><br />

putative CEP17-aneusomic cases in a consecutive series, within a single<br />

health system. Methods: 150 <strong>of</strong> 1256 consecutive breast carcinoma cases<br />

accessioned in 2011 within the Cleveland Clinic Health System displayed a<br />

mean CEP17 copy number greater than 3.0 by FISH (aneusomy). The<br />

patients were reflex-HER2 tested by FISH with a reference probe for the<br />

D17S122 locus (17p12) and a corrected HER2/Chr-17 ratio was calculated.<br />

Cases with equivocal HER2/D17S122 ratio (1.8-2.2) were further<br />

reflex tested for HER2 overexpression by immunohistochemistry (IHC).<br />

Results: Of 117 initially non-amplified cases by HER2/CEP17, 20 (17%)<br />

were revised to amplified and 18 (15.3%) to equivocal by HER2/D17S122.<br />

Of 3 initially equivocal cases, 1 was revised to amplified and 1 remained<br />

equivocal. Of the 19 equivocal cases by HER2/D17S122, 3 were revised to<br />

positive by IHC. Overall, for CEP17 aneusomic cases tested using this<br />

algorithmic approach, 24 <strong>of</strong> 120 (20%) patients with initial non-amplified<br />

or equivocal HER2 status became eligible for anti-HER2-based therapy,<br />

which was also considered in 10 equivocal cases with a HER2/D17S122<br />

ratio <strong>of</strong> 2.0 - 2.2. A significantly lower proportion <strong>of</strong> initially amplified cases<br />

was revised as non-amplified by HER2/D17S122 (1 <strong>of</strong> 30, 3.3%, p�0.05).<br />

Conclusions: Our data, collected within a single health system for a<br />

consecutive case series, underscores the clinical limitations <strong>of</strong> commonly<br />

used FISH probes for HER2 testing and demonstrates that algorithmic use<br />

<strong>of</strong> a non-centromeric Chr-17 reference probe alters HER2 status and<br />

increases eligibility for anti-HER2 based therapy in a significant proportion<br />

<strong>of</strong> patients.<br />

619 General Poster Session (Board #11F), Sat, 8:00 AM-12:00 PM<br />

Presentation and treatment <strong>of</strong> HER2-positive metastatic breast cancer patients<br />

already treated with adjuvant trastuzumab. Presenting Author: Davis Yuri<br />

Torrejon, Medical Oncology Department, Vall d’Hebron University Hospital,<br />

Barcelona, Spain<br />

Background: The introduction <strong>of</strong> trastuzumab in the clinical armamentarium has<br />

pr<strong>of</strong>oundly changed the natural history <strong>of</strong> HER2 positive breast cancer (BC).<br />

However, about 15% <strong>of</strong> patients with HER2 early BC treated with adjuvant<br />

trastuzumab continue to relapse. We aimed to analyze these patients with<br />

respect to clinical presentation and response to treatment. Methods: Data were<br />

retrieved from the institutional BC database <strong>of</strong> Vall d’Hebron. All the cases<br />

relapsing after exposure to adjuvant trastuzumab were analyzed. Change in<br />

expression <strong>of</strong> hormone-receptor (HR) and HER-2 status between primary tumour<br />

and corresponding local recurrence or distant metastasis was also evaluated<br />

Results: A total <strong>of</strong> 270 patients were identified. Twenty-six patients (9.6%)<br />

relapsed (Table). Overall median time from diagnosis to relapse was 27.1<br />

months (24.1-30.1) being 29.1 months (14.3-44.1) in HR positive and 23.1<br />

(9.9-36.2) in HR negative cases (p�0.037). Median time from last dose <strong>of</strong><br />

trastuzumab to relapse was 7.6 (2.7-12.7) months, being 10.6 (0-27.9) and 3<br />

months (0-7.6) in HR positive and negative cases, respectively (p�0.026).<br />

Sixteen patients have already progressed to first-line therapy with a median time<br />

to progression (TTP) <strong>of</strong> 7.4 months with no statistical difference among HR<br />

positive and HR negative (4.4 and 9.8 months, p�0.3). No difference was found<br />

in TTP to first line therapy among early (before 12 months) and delayed (after 12<br />

months) progression on adjuvant trastuzumab. Among the 17 cases with primary<br />

tumor and matched metastatic biopsy, HER-2 negativization was detected in 2<br />

cases; whereas a change in estrogen and progesterone receptors was seen in<br />

17.6% and 29.4% <strong>of</strong> cases, respectively. Conclusions: Patients with HER2�/HR<br />

negative treated with adjuvant trastuzumab seems to have a significantly shorter<br />

time to relapse compared with the HER2�/HR� tumors. In these patients<br />

biopsy <strong>of</strong> metastatic lesions might help to define the best treatment options.<br />

HER2�/HR� (n�160) HER2 �/HR- (n�110) Total (n�270)<br />

No % No % No %<br />

Recurrence 14 8.7 12 10.9 26 9.6<br />

Sites<br />

Locoregional 5 35.7 3 25 8 30.8<br />

Bone 3 21.4 7 58.3 10 38.5<br />

Liver 6 42.8 2 16.7 8 30.8<br />

Lung 3 21.4 2 16.7 5 19.2<br />

CNS 2 14.3 3 25 5 19.2<br />

Death 3 21.4 8 66.6 11 42.3<br />

621 General Poster Session (Board #11H), Sat, 8:00 AM-12:00 PM<br />

Susceptibility <strong>of</strong> HER2� breast cancer cells to poly (ADP-ribose) polymerase<br />

(PARP) inhibition independent <strong>of</strong> an inherent DNA repair defect.<br />

Presenting Author: Eddy Shih-Hsin Yang, University <strong>of</strong> Alabama at Birmingham,<br />

Birmingham, AL<br />

Background: HER2 overexpression in breast cancer confers increased tumor<br />

aggressiveness. Targeted therapy against HER2 has improved outcomes,<br />

but resistance and disease progression ultimately occurs, thus necessitating<br />

novel therapeutic strategies. PARP inhibitors target homologous recombination<br />

(HR) deficient tumors, such as the BRCA-associated breast and<br />

ovarian cancers. In this study, we report unexpected susceptibility <strong>of</strong><br />

HER2� breast cancer cells to PARP inhibition alone independent <strong>of</strong> an<br />

inherent HR deficiency. Methods: Cell viability was measured using colony<br />

formation and ATPLite assays. Tumor growth delay was assessed in vivo in<br />

mice bearing breast cancer xenografts. Proteins were detected by immunoblotting.<br />

HR was assayed using radiation (IR)-induced Rad51 foci or a<br />

GFP-based HR assay. NF�B activity was measured using a NF�B-driven<br />

luciferase assay. Results: Surprisingly, PARP inhibition with ABT-888<br />

alone reduced the colony forming ability and cell viability <strong>of</strong> the HER2�<br />

breast cancer cell lines BT474, SKBR3, MDA-MB361, and HCC1954<br />

(~70 – 99% reduction at 10�M). This susceptibility did not correlate with<br />

an inherent HR deficit. Interestingly, HER2 overexpression itself may be<br />

one mechanism <strong>of</strong> susceptibility to ABT-888 as evidenced by increased<br />

cellular cytotoxicity and in vivo tumor growth delay <strong>of</strong> MCF7 cells stably<br />

expressing HER2. Further dissection <strong>of</strong> the mechanism revealed that NF�B<br />

transcriptional activity was significantly inhibited by ABT-888 (�95%)<br />

which corresponded with reduced levels <strong>of</strong> phosphorylated p65 and total<br />

IKK�, and a concomitant increase in IkB�. Furthermore, overexpression <strong>of</strong><br />

p65 abrogated cellular sensitivity to ABT-888. Conclusions: HER2� breast<br />

cancer cells are highly susceptible to PARP inhibition despite being HR<br />

pr<strong>of</strong>icient. This may be, in part, due to inhibition <strong>of</strong> NF�B. Further<br />

investigation <strong>of</strong> whether the addition <strong>of</strong> PARP inhibition to HER2 targeted<br />

therapy will delay the onset <strong>of</strong> resistance to therapy is warranted to<br />

potentially improve outcomes in HER2� breast cancer patients.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


622 General Poster Session (Board #12A), Sat, 8:00 AM-12:00 PM<br />

ADAM17 as a novel therapeutic target for HER2-positive breast cancer.<br />

Presenting Author: Sumainizah Sukor, St Vincent’s University Hospital,<br />

Dublin, Ireland<br />

Background: Although the HER2 gene is amplifiend/overexpressed in<br />

15-20% <strong>of</strong> breast cancers, only a proportion <strong>of</strong> these patients benefit from<br />

anti-HER2 therapy. Clearly additional biomarkers and/or therapeutic targets<br />

are necessary to enhance efficacy and improve outcome in these<br />

patients. Here, we tested the hypothesis that inhibition <strong>of</strong> ADAM17mediated<br />

release <strong>of</strong> HER ligands may enhance response to trastuzumab<br />

and lapatinib. Methods: The ADAM17-selective inhibitor, PF-5480090<br />

(Pfizer) both alone and in combination with lapatinib, trastuzumab or 5FU<br />

was tested for potential growth inhibitory effects on a panel <strong>of</strong> 4 HER2positive<br />

breast cancer cell lines (BT474, MDA-MB-453, SKBR3 and<br />

JIMT-1). Results: Using the MTT viability assay, treatment with 1 �M TMI-2<br />

resulted in a significant reduction in cell proliferation compared to vehicle<br />

control in BT474 (p�0.01), MDA-MB-453 (p � 0.005), JIMT-1 (p�0.002)<br />

and SKBR3 cells (p � 0.005). Addition <strong>of</strong> PF-5480090 to lapatinib<br />

resulted in a significant reduction in cell growth compared to lapatinib<br />

alone (JIMT-1: p � 0.005; SKBR3: p � 0.005; MDA-MB-453: p �<br />

0.005), while addition <strong>of</strong> PF-5480090 to trastuzumab resulted in significant<br />

reduction in cell growth compared to trastuzumab alone (JIMT-1: p �<br />

0.005; SKBR3: p � 0.005; MDA-MB-453: p�0.001). Addition <strong>of</strong><br />

PF-5480090 to 5FU resulted in significant reduction in growth compared<br />

to 5FU alone (JIMT-1: p � 0.005; SKBR3: p � 0.005; MDA-MB-453:<br />

p�0.001). Consistent with its ability to block proliferation, addition <strong>of</strong><br />

PF-5480090 significantly reduced release <strong>of</strong> TGFalpha (BT474: p�0.003;<br />

JIMT1: p�0.023; SKBR3: p�0.036 and MDA-MB-453: p�0.014).<br />

Conclusions: Inhibition<strong>of</strong> ADAM17 resulted in growth inhibitory responses<br />

in HER2-positive breast cancer cell lines. Furthermore, addition <strong>of</strong> PF-<br />

5480090 to lapatinib, trastuzumab or 5FU resulted in significant growth<br />

inhibition compared to these agents alone. We propose that inhibition <strong>of</strong><br />

ADAM17 may be used in combination with existing treatments for<br />

HER2-positive breast cancer. Acknowledgement. The authors thank SFI<br />

(SRC award, 08/SRC/B1410 to MTCI) for funding this work.<br />

624 General Poster Session (Board #12C), Sat, 8:00 AM-12:00 PM<br />

Phase I/II trial <strong>of</strong> primary chemotherapy with nonpegylated liposomal<br />

doxorubicin, paclitaxel, and lapatinib in patients with HER2-positive,<br />

early-stage breast cancer. Presenting Author: Sherko Kuemmel, Kliniken<br />

Essen Mitte, Evang. Huyssens Stiftung/Knappschaft , Essen, Germany<br />

Background: Combinations <strong>of</strong> trastuzumab and anthracyclines in HER2positive<br />

breast cancer are highly active but associated with a high<br />

incidence <strong>of</strong> cardiotoxicity. The risk <strong>of</strong> cardiac damage can be significantly<br />

reduced through liposomal encapsulation <strong>of</strong> anthracyclines. This phase I/II<br />

study was initiated to evaluate the combination <strong>of</strong> non-pegylated liposomal<br />

doxorubicin (NPLD), paclitaxel and lapatinib as primary treatment for<br />

patients with early stage, HER2-positive primary breast cancer. Methods:<br />

Patients with newly diagnosed HER2-positive (IHC 3� or FISH�) early<br />

stage (T1c N1-2 or T2 N0-2) breast cancer were treated with NPLD<br />

(60mg/m2 ; day 1), paclitaxel (175mg/m2 , day 1) and lapatinib (750-1500<br />

mg orally daily) in 3-week intervals for up to 6 cycles. The primary<br />

endpoints were dose-limiting toxicities (DLT) and pathological complete<br />

response (pCR). Secondary endpoints include safety, incidence <strong>of</strong> cardiac<br />

events, and clinical response. Exploratory endpoints include molecular<br />

markers for sensitivity or resistance to chemotherapy and/or lapatinib<br />

evaluated. Results: A total <strong>of</strong> 84 patients have been included to date.No<br />

DLTs were observed and the maximum tolerated doses were NPLD<br />

60mg/m2 , paclitaxel 175mg/m2 and lapatinib 1500mg. Recommended<br />

phase 2 doses (P2D) were NPLD 60mg/m2 , paclitaxel 175mg/m2 and<br />

lapatinib 1250mg. The treatment was generally well tolerated and associated<br />

with toxicities that were consistent with the known side-effects <strong>of</strong> the<br />

individual agents. No cardiac event has been observed to date. Preliminary<br />

efficacy data confirm a pCR breast rate <strong>of</strong> 41.7% and pCR rate in breast<br />

and lymph nodes <strong>of</strong> 37.5%, in 24 evaluable patients treated at �P2D.<br />

Conclusions: The combination <strong>of</strong> NPLD, paclitaxel and lapatinib is well<br />

tolerated and has high antitumour activity in patients with HER2-positive<br />

primary breast cancer. The phase II part <strong>of</strong> the study is ongoing and<br />

updated results will be presented.<br />

Breast Cancer—HER2/ER<br />

37s<br />

623 General Poster Session (Board #12B), Sat, 8:00 AM-12:00 PM<br />

HER2 assessment and Ki-67 labeling index in a cohort <strong>of</strong> male breast<br />

cases: The Ich Network on Cancer Research (INCaRe) experience. Presenting<br />

Author: Giovanna Masci, Humanitas Cancer Center, IRCCS, Rozzano,<br />

Italy<br />

Background: The overall incidence <strong>of</strong> male breast cancers (MBC) is around<br />

1% <strong>of</strong> all breast cancers and is on the rise.Most <strong>of</strong> our current knowledge<br />

regarding its biology and treatment strategies has been extrapolated from<br />

its female counterpart. However, from literature data, it is more and more<br />

evident that MBC has biological differences compared with female breast<br />

cancer (FBC). While hormone receptors are more frequently positive in<br />

MBC than in FBC, HER-2 seems to be less expressed in MBC than in FBC,<br />

with data ranging from 0 to 18%; no data on Ki-67 have been so far<br />

reported. Methods: We retrospectively analyzed the immunohistochemical<br />

expression <strong>of</strong> hormone receptors status, HER-2 protein expression, and<br />

Ki-67 in 76 consecutive MBCs, treated within the Humanitas Institutes<br />

Network on Cancer Research (INCaRe). HER-2 determinations were carried<br />

out according to ASCO/ACP and NEQAS guidelines: cases with score 2� at<br />

IHC were further examined by fluorescent in situ hybridation (FISH).<br />

Results: From 2000 to 2011, we treated 76 male breast cases (age 25-87,<br />

median 64): 72 patients (94%) had ductal carcinoma and 4 had rare<br />

histotypes (2 papillary, 1 mucinous and 1 cribryform). Thirthy-two <strong>of</strong> 76<br />

patients (42%) had positive axillary lymph-nodes, while 6 (8%) were<br />

metastatic at diagnosis. Of these, estrogen receptor and progesterone<br />

receptor were positive in 96% and 93% patients respectively; HER-2,<br />

evaluable in 67 patients, was positive in 11 (16%). Ki-67 was evaluable in<br />

75 patients and was � 20% in 24 cases (32%), with 20/24 (26%) with<br />

Ki-67 � 30%. Grading was evaluable in 65 patients: G1 in 2 (3%), G2 in<br />

41(63%) and G3 in 22 (34%), respectively. Conclusions: In these series,<br />

MBC show different patterns from FBC, with some favorable aspects such<br />

as higher hormone receptor status and much lower HER-2 expression and<br />

some unfavorable features, such as higher Ki-67 values. Although further<br />

studies are needed to confirm these data, different treatment strategies<br />

would be suggested in MBC than its female counterpart.<br />

625 General Poster Session (Board #12D), Sat, 8:00 AM-12:00 PM<br />

An assessment <strong>of</strong> disease features and immune response in breast cancer<br />

patients that did not recur after receiving HER2 peptide, AE37 vaccine in a<br />

randomized phase II trial. Presenting Author: Diane F Hale, Brooke Army<br />

Medical Center, San Antonio, TX<br />

Background: In a phase I trial <strong>of</strong> AE37, the Ii-Key hybrid <strong>of</strong> HER2 derived<br />

peptide AE36 (776-790), administered with immunoadjuvant GMCSF<br />

demonstrated the vaccine to be safe and capable <strong>of</strong> stimulating CD4�helper T cells with HER2 specific anti-tumor activity. Here we present analysis <strong>of</strong><br />

immune markers and patient feature that may impact recurrence from an<br />

ongoing prospective, randomized, single-blinded Phase IIb trial <strong>of</strong><br />

AE37�GMCSF v GMCSF alone in adjuvant high risk breast cancer (BC)<br />

patients. Methods: After completion <strong>of</strong> standard therapy; disease-free, node<br />

positive or high risk node negative BC patients (pts) were randomized to<br />

receive either AE37�GMCSF or GMCSF in 6 monthly intradermal inoculations.<br />

Immunologic responses were measured using [ 3H]-thymidine incorporation<br />

assay (in vitro), delayed-type hypersensitivity (DTH) reactions (in<br />

vivo) and T regulatory cells (Tregs). Among those vaccinated recurrent pts<br />

(VR) are compared to non recurrent (VNR) pts. Results: We have vaccinated<br />

109 pts with 8.3% recurrence rate at 2 year median follow up. VR v VNR<br />

were younger (44 v 50 yo p�0.11), had higher grade (67% v 44%<br />

p�0.32), more ER/PR- (44% v 38% p�0.75), larger tumors (89% v 50%<br />

p�0.06), and node positive (89% v 70% p�0.37). No difference for HER2<br />

status (IHC 3�, 44% v 49% p�0.64). Both VR and VNR responded to<br />

vaccine though the mean DTH and proliferative stimulation index was<br />

approximately 10% less in VR pts (18 v 20 p�0.73; 1.96 v 2.2 p�0.77<br />

respectively). The most predictive measure was change in Tregs with VR pts<br />

less likely to decrease their Tregs levels (50% v 76% p�0.17) after<br />

vacination and more likely experience increased Tregs (17% v 8%<br />

p�0.48). A decrease in Tregs had an inverse trend towards recurrence<br />

(p�0.17). Conclusions: Preliminarily, it appears most pts immunologically<br />

respond to vaccine though slightly less for VR in most assays. The changes<br />

<strong>of</strong> Tregs appear to correlate best with disease recurrence. Whether this<br />

reflects an association with disease status or a failure <strong>of</strong> the vaccine is yet to<br />

be seen. These levels may become important in predicting risk for clinical<br />

recurrence in future vaccine trials.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


38s Breast Cancer—HER2/ER<br />

626 General Poster Session (Board #12E), Sat, 8:00 AM-12:00 PM<br />

Efficacy <strong>of</strong> INK128, an mTORC1/mTORC2 kinase inhibitor, in breast<br />

cancer models driven by HER2-PI3K-AKT-mTOR pathway. Presenting<br />

Author: Pradip De, Department <strong>of</strong> Medicine, Emory University, Atlanta, GA<br />

Background: Downstream <strong>of</strong> the PI3K-AKT pathway, the mTOR has been<br />

shown to be essential effector in promoting cell proliferation, survival,<br />

mRNA translation and tumor susceptibility. Aberrant activation <strong>of</strong> the<br />

PI3K-AKT-mTOR pathway occurs frequently in breast tumor (BT) and<br />

contributes to resistance to trastuzumab (T). Using a HER2 amplified BT<br />

model; we investigated the antitumor efficacy <strong>of</strong> the dual mTORC1/<br />

mTORC2 kinase inhibitor INK128 alone and in combination with T.<br />

Methods: The antiproliferative and HER2-mediated cellular signaling (pAKT,<br />

pP70S6K, pS6RP, p4EBP1 and p-ERK) effects <strong>of</strong> INK128 alone and in<br />

combination with T were evaluated in HER2 amplified T-sensitive (BT474),<br />

T-resistant (BT474HR), and HER2 amplified/PIK3CA mutated (HCC1954,<br />

UACC893) BT cell lines by MTT assay and Western blots. Athymic mice<br />

bearing BT474 and BT474HR xenograft tumors were treated with INK128<br />

and T (alone and in combination). Results: (1)INK128 exhibited excellent<br />

in vitro cell killing activity in MTT assay with IC50’s below 50nM. INK128<br />

was more potent when combined with T, (2) INK128 blocked mTORC1,<br />

mTORC2 and thus did not cause the mTORC2 activation <strong>of</strong> AKT observed<br />

with rapalogues, (3) inhibition <strong>of</strong> phosphorylation <strong>of</strong> AKT(S473), P70S6K,<br />

S6RP, and 4EBP1(T37/46, T70) was observed following INK128 treatment,<br />

and the combination <strong>of</strong> INK128 and T more effectively blocked the<br />

PI3K-AKT-mTOR pathway, (4) INK128 dose-dependently blocked 3D-ON-<br />

TOP clonogenic growth <strong>of</strong> HER2� cells. This effect was potentiated in the<br />

presence <strong>of</strong> T and (5) xenograft data show that the combination <strong>of</strong> INK128<br />

and T has strongly enhanced anti-tumor effect in both sensitive and<br />

resistant models, something that cannot be achieved by either monotherapy.<br />

Conclusions: Our data suggest that 1) therapeutic targeting <strong>of</strong> the<br />

PI3K-AKT-mTOR signaling should be effective in abrogating resistance to T<br />

therapy in HER2� BT, 2) INK128 mitigates the feedback activation <strong>of</strong> AKT<br />

caused by selective inhibition <strong>of</strong> mTORC1 and 3) targeting both the HER2<br />

and the mTORC1/2 signaling pathways is an attractive strategy to enhance<br />

the clinical activity <strong>of</strong> T therapy, as well as to prevent or delay the<br />

development <strong>of</strong> resistance.<br />

629 General Poster Session (Board #12H), Sat, 8:00 AM-12:00 PM<br />

Impact <strong>of</strong> PIK3CA mutations and p95HER2 expression on the outcome <strong>of</strong><br />

HER2-positive metastatic breast cancer patients treated with a trastuzumabbased<br />

therapy. Presenting Author: Andrea Fontana, Division <strong>of</strong> Medical<br />

Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Istituto Toscano<br />

Tumori, Pisa, Italy<br />

Background: Currently, no biomarkers <strong>of</strong> trastuzumab (T) clinical resistance<br />

have been validated. The aim <strong>of</strong> this pilot study was to evaluate the impact<br />

<strong>of</strong> PIK3CA mutations and p95HER2 (pHER2 truncated form) expression<br />

on the efficacy <strong>of</strong> a T based-therapy in a HER2-positive metastatic breast<br />

cancer (MBC) patients (pts). Methods: 107 HER2-positive MBC pts, treated<br />

in the last 10 years, were evaluated. Median age was 54 years (25-79);<br />

ECOG performance status was 0 in 56% <strong>of</strong> pts; all pts received several lines<br />

<strong>of</strong> treatment including T; biomarkers molecular analysis was performed in<br />

70 tumor specimens. The IHC expression <strong>of</strong> p95HER2 was evaluated by a<br />

monoclonal antibody that specifically recognizes only the HER2 external<br />

domain; the HER2 integrity was defined by the presence <strong>of</strong> a homogeneous<br />

membrane staining (moderate or intense) in at least 30% <strong>of</strong> the cells,<br />

otherwise the HER2 was defined as p95HER2 positive. PIK3CA mutations<br />

in exons 9 and 20 were detected by automated sequencing. The molecular<br />

data were correlated to Time to progression (TTP) <strong>of</strong> the first line treatment<br />

including T and the Overall Survival (OS) by using the Kaplan-Meir method<br />

and the log-rank-test. Results: p95HER2 positive pts and PIK3CA mutations<br />

in exon 9 or 20 were detected in 42% and 22% <strong>of</strong> tumor specimens,<br />

respectively. p95HER2 positive tumors showed a shorter TTP and OS that<br />

did not reach statistical significance; PIK3CA mutations correlated with a<br />

worse TTP (median 7,6 vs 11,3 months) and OS (median 20,1 vs 41,0<br />

months, p� 0,046). Conclusions: These preliminary results suggest a<br />

possible role <strong>of</strong> PIK3CA mutational status in predicting the outcome <strong>of</strong><br />

MBC pts treated with T.<br />

628 General Poster Session (Board #12G), Sat, 8:00 AM-12:00 PM<br />

Detection <strong>of</strong> trastuzumab (T) level in human serum using quartz crystal<br />

microbalance (QCM) piezo-immunosensors by immobilization <strong>of</strong> a HER2<br />

mimotope-derived synthetic peptide and its potential application in breast<br />

cancer. Presenting Author: Mohammad Muhsin Chisti, Oakland University<br />

William Beaumont School <strong>of</strong> Medicine, Royal Oak, MI<br />

Background: Varied clinico-pathological response to monoclonal antibodies<br />

like T has been reported either due to presence <strong>of</strong> antibodies, rapid<br />

clearance or low density <strong>of</strong> target receptor/antigens. This demands need for<br />

an assay to monitor serum T therapeutic levels to ensure appropriate<br />

dosage. Enzyme-linked immunosorbent assay (ELISA) is still the most<br />

widely used technique to detect T level in human serum which is expensive<br />

and time consuming. For the first time, we established a platform to detect<br />

T level by using a small (�2.2 kDa), inexpensive, highly stable HER2<br />

mimotope-derived synthetic peptide immobilized on the surface <strong>of</strong> a gold<br />

quartz electrode. Methods: HER2 mimotope was used as a substitute for the<br />

HER2 receptor protein in QCM assays to detect T level. The validation<br />

samples were prepared from the standard T solution in 10% human serum<br />

at three concentrations (10, 20 and 40 ug/ml). The changes in frequencies<br />

(�F) <strong>of</strong> sera from 3 female patients , 61, 32 and 44 years old , with ER/PR<br />

positive, HER2/neu positive metastatic breast cancer were obtained by<br />

calculating the differences between frequency shifts in pre and post T<br />

infusion.T level was calculated by equation, (�F �1.0022) ÷ 0.9997 �g /<br />

ml. Results: We showed that assay sensitivity was dependent upon the<br />

amino acids used to tether and link the peptide to the sensor surface and<br />

the buffers used. QCM assay was capable <strong>of</strong> detecting T serum level as low<br />

as 0.038 nM (linear operating range <strong>of</strong> 0.038–0.859 nM). T levels <strong>of</strong> 3<br />

patients were 43.34, 121.96 and 193.18 �g /ml corresponding to pre and<br />

post infusion �F <strong>of</strong> 3.33, 11.19 and 18.31 respectively. The time frame <strong>of</strong><br />

assay was 20-30 minutes. These results were in concordance with<br />

previously published results using ELISA. Conclusions: For the first time, we<br />

have established a low cost, highly sensitive, fast, synthetic peptide based<br />

QCM assay which could be used as a basis for developing a new generation<br />

<strong>of</strong> affinity-based Immunosensor assays to monitor serum levels <strong>of</strong> T and<br />

other monoclonal antibodies, helping physicians to determine the clinical<br />

efficacy <strong>of</strong> these drugs and ensuring appropriate dosages.<br />

630 General Poster Session (Board #13A), Sat, 8:00 AM-12:00 PM<br />

Impact <strong>of</strong> single/dual HER2 inhibition and chemotherapy (CT) backbone<br />

upon pathologic complete response (pCR) in patients receiving neoadjuvant<br />

CT for operable/locally advanced breast cancer (O/LABC): A treatmentinteraction<br />

analysis <strong>of</strong> randomized trials. Presenting Author: Jenny<br />

Furlanetto, Medical Oncology, University <strong>of</strong> Verona, Verona, Italy<br />

Background: Although the addition <strong>of</strong> trastuzumab to neoadjuvant CT for<br />

O/LABC have dramatically increased pCR, clinical research is moving<br />

forward to further improve the overall outcome. In this regard, the DUAL<br />

inhibition <strong>of</strong> the HER-2 pathway and the choice <strong>of</strong> the best CT backbone<br />

may represent an issue to be addressed. Methods: Phase II randomized/<br />

Phase III trials were considered. pCR events/rates were extracted from<br />

papers/presentation and cumulated (R[pCR]) according to a random-effect<br />

model; 95% confidence intervals (CI) were derived. A sensitivity analysis<br />

according to SINGLE/DUAL HER-2 inhibition and to administered CT<br />

(anthracyclines-taxanes: anthra-TAX; TAX alone) was accomplished, in<br />

order to test for interaction. Results: 7 trials (2155 pts) were gathered;<br />

1855 pts were enrolled in arms where anti-HER-2 targeted therapy was<br />

administered. HER-2 inhibition was obtained with trastuzumab, lapatinib<br />

and pertuzumab (alone or combined). Results according to HER-2 inhibition<br />

follow in the table below. With regard to CT, a significant interaction<br />

(p�0.0001) in favour <strong>of</strong> the addition <strong>of</strong> Anthra to TAX was found in the<br />

context <strong>of</strong> SINGLE HER-2 inhibition subgroup (R[pCR] 46.5%, 95% CI<br />

37-51, vs 26.9%, 95% CI 23-31), while no significant interaction was<br />

determined in the context <strong>of</strong> the DUAL subgroup (R[pCR] 49.0%, 95% CI<br />

26-72, vs 49.0%, 95% CI 43-55). A significant interaction (p�0.0001) in<br />

favour <strong>of</strong> the addition <strong>of</strong> Anthra to TAX was found for pts receiving<br />

trastuzumab (R[pCR] 45.5%, 95% CI 37-53, vs 26.5%, 95% CI 21-32) as<br />

well. Conclusions: Although biases in the pCR definition, the DUAL<br />

inhibition <strong>of</strong> HER-2 pathway may significantly increase pCR, regardless <strong>of</strong><br />

the CT backbone; if confirmed, this strategy allows to reduce toxicities by<br />

sparing Anthra. For pts receiving SINGLE HER-2 inhibition, Anthra-TAXbased<br />

CT should be still considered the best treatment option.<br />

Anthra-TAX TAX<br />

Single Dual Single Dual<br />

pCR (n°)/pts 259/591 139/390 136/506 127/259<br />

R[pCR] (95% CI) 46.5% (37-51) 49.0% (26-72) 26.9% (23-31) 49.0% (43-55)<br />

Interaction test p�0.67 p�0.0001<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


631 General Poster Session (Board #13B), Sat, 8:00 AM-12:00 PM<br />

Prognostic role <strong>of</strong> HER2 loss after neoadjuvant therapy in patients with<br />

HER2-positive operable breast cancer. Presenting Author: Valentina Guarneri,<br />

Department <strong>of</strong> Oncology, Hematology and Respiratory Diseases,<br />

Modena University Hospital, Modena, Italy<br />

Background: Emerging literature data are consistently showing that a<br />

change in HER2 status adversely affect the prognosis <strong>of</strong> breast cancer<br />

patients. Aim <strong>of</strong> the present analysis is to evaluate the prognostic impact <strong>of</strong><br />

HER2 loss in breast cancer patients with HER2 positive disease treated<br />

with neoadjuvant therapy with or without anti-HER2 agents. Methods: A<br />

total <strong>of</strong> 94 HER2 positive patients were identified from a prospectively<br />

maintained database. The first cohort (A) includes 40 patients treated with<br />

chemotherapy alone (enrolled before 2005). The second cohort (B)<br />

includes 54 patients treated with neoadjuvant chemotherapy in combination<br />

with anti-HER2 agents (trastuzumab and/or lapatinib). HER2 expression<br />

was evaluated by IHC or FISH on pre-treatment core biopsy and on<br />

surgical specimen after neoadjuvant therapy. Patients were considered as<br />

having HER2 positive disease in case <strong>of</strong> IHC 3� or FISH amplification.<br />

Results: No imbalance in terms <strong>of</strong> age, stage at diagnosis, tumor grade and<br />

expression <strong>of</strong> hormone receptor was observed in the two cohorts. In detail,<br />

67% and 61% <strong>of</strong> the patients have a co-expression <strong>of</strong> HER2 and hormone<br />

receptor in cohort A and B, respectively. The rate <strong>of</strong> breast conservation was<br />

significantly higher in cohort B (chemotherapy�anti-HER2 agents) as<br />

compared to cohort A (chemotherapy alone) (59% vs 38%, p�0.048).<br />

Similarly, the rate <strong>of</strong> pathologic complete response (pCR) was significantly<br />

higher in cohort B (42.6% vs 7.5% in cohort A, p�0.001). A change in<br />

HER2 expression from biopsy to post-therapy samples was observed in<br />

35% <strong>of</strong> the patients in cohort A vs 9% <strong>of</strong> the patients in cohort B (p�0.04).<br />

No patients achieving a pCR have recurred so far vs 25% <strong>of</strong> the patients<br />

with less than pCR (p�0.005). The rate <strong>of</strong> recurrence was significantly<br />

higher for patients experiencing a change in HER2 expression (47% vs<br />

15% in patients with no change, p�0.007). At 5 years, 53% <strong>of</strong> the<br />

patients with Her2 change and 75% <strong>of</strong> the patients without Her2 change<br />

were alive and free <strong>of</strong> recurrence (log rank test: p�0.03). Conclusions: The<br />

rate <strong>of</strong> HER2 loss was significantly higher in patients not receiving<br />

anti-HER2 agents as a part <strong>of</strong> the neoadjuvant therapy. In this series, the<br />

change in HER2 status has a negative prognostic impact.<br />

633 General Poster Session (Board #13D), Sat, 8:00 AM-12:00 PM<br />

Effect <strong>of</strong> acquired resistance to lapatinib in HER2-positive breast cancer<br />

cells on the Bcl-2 family members MCL-1 and BAX. Presenting Author:<br />

Alex J. Eustace, National Institute for Cellular Biotechnology, Dublin City<br />

University, Dublin, Ireland<br />

Background: Lapatinib (L) is approved for the treatment <strong>of</strong> trastuzumab (T)<br />

resistant HER2 positive metastatic breast cancer. Not all HER2 positive<br />

tumors respond to L and patients who initially respond frequently relapse,<br />

due to the development <strong>of</strong> resistance to L. Understanding the molecular<br />

changes associated with L resistance may lead to the identification <strong>of</strong><br />

targets to overcome resistance. Methods: SKBR3 cells were treated with<br />

either 250 nM L, or 125nM L and 5 �g/ml T twice weekly for 6 months to<br />

establish the resistant cell lines SKBR3-L and -TL. We measured by TUNEL<br />

assay the effect <strong>of</strong> L on apoptotic induction in SKBR3, -L and -TL cells.<br />

Gene array analysis <strong>of</strong> the SKBR3, -L and -TL cells identified differences in<br />

the expression <strong>of</strong> apoptosis-related genes, which were validated by immunoblotting.<br />

Finally using combinations <strong>of</strong> obatoclax (O) and L were tested in L<br />

resistant cells. Results: In SKBR3 cells, L (500 nM) induces apoptosis<br />

(15.8 � 2.0%) compared to untreated controls (4.6�2.7%), whilst in<br />

SKBR3-L and -TL cells L did not induce significant apoptosis compared to<br />

controls. Gene array analysis showed that BAX mRNA is down regulated 3.2<br />

fold in SKBR3-L cells compared to the parental cells. In SKBR3-TL cells,<br />

MCL-1 mRNA expression is increased 2.1 fold, whilst BAX mRNA is down<br />

regulated 2.1 fold compared to the parental cells. Immunoblotting confirmed<br />

that BAX protein expression was reduced in the SKBR3-L (1.5 fold,<br />

p�0.058) and significantly reduced in SKBR3-TL cells (2.0 fold, p�0.039)<br />

compared to SKBR3. MCL-1 protein expression was significantly increased<br />

in the SKBR3-L (1.6 fold, p�0.035) and -TL cells (2.3 fold, p�0.031)<br />

compared to SKBR3. Combining O (200 nM) and L (500 nM) in both<br />

SKBR3 and SKBR3-L cells produced greater growth inhibition than either<br />

drug on its own (SKBR3: 86.9�1.0% vs 73.5�3.1% for L (p�0.01) and<br />

44.7�7.7% for O, (p�0.01); SKBR3-L: 54.2� 8.6% vs 26.9�2.4% for L<br />

(p�0.027) and 33.8�10.7% for O (p�0.04)). Conclusions: Extended<br />

exposure to L in SKBR3-L and –TL cells alters the apoptotic response to L.<br />

O alone and in combination with L results in growth inhibition in L resistant<br />

cells.<br />

Breast Cancer—HER2/ER<br />

39s<br />

632 General Poster Session (Board #13C), Sat, 8:00 AM-12:00 PM<br />

Effect <strong>of</strong> neratinib (N) alone and in combination with trastuzumab (T) in<br />

HER2-positive breast cancer (BC) cell lines. Presenting Author: Alexandra<br />

Canonici, Molecular Therapeutics for Cancer Ireland, Dublin, Ireland<br />

Background: HER-2, a member <strong>of</strong> the transmembrane receptor tyrosine<br />

kinase ErbB family, is over-expressed in approximately 25% <strong>of</strong> BC. HER-2<br />

targeted therapies, in particular, T, a monoclonal antibody targeting<br />

HER-2, and lapatinib (L), a reversible HER-2 tyrosine kinase inhibitor, have<br />

been shown to significantly improve the prognosis for HER-2 positive BC<br />

patients. However, resistance to T and/or L is a significant clinical problem.<br />

The aim <strong>of</strong> this study is to assess the activity <strong>of</strong> N (HKI-272), an irreversible<br />

HER-2 tyrosine kinase inhibitor, in HER-2 overexpressing BC cell lines,<br />

including T and/or L resistant cells. Methods: Using proliferation assays, the<br />

effect <strong>of</strong> N was assessed alone and in combination with T in HER-2 positive<br />

BC cell lines, including T and/or L resistant cell lines. The effect <strong>of</strong> N on<br />

HER-2 and downstream signalling molecules, Erk and Akt, was determined<br />

by immunoblotting. Results: HER-2 positive BC cell lines, including T<br />

and/or L resistant cells, are sensitive to N alone with IC50 values<br />

(concentration which inhibits 50% <strong>of</strong> growth) ranging from 1 to 280 nM.<br />

The combination <strong>of</strong> N and T has additive effects in SkBR3 and BT474<br />

which are sensitive to T and also in SKBR3-Lwhich are resistant to L. In the<br />

cell lines HCC1954, HCC1954-L, MDA-MB-453, JIMT1 and SKBR3-HL<br />

which are resistant to T, combined treatment with T and N showed no<br />

enhancement compared to N alone. Finally, N decreased phosphorylation<br />

<strong>of</strong> HER-2, Erk and Akt in all cell lines tested. Conclusions: Our results<br />

suggest that N should be studied in patients with HER-2 positive BC,<br />

including patients with T and/or L resistant BC. We also demonstrate that N<br />

in combination with T may be more effective than either agent alone in T<br />

sensitive cells.<br />

634 General Poster Session (Board #13E), Sat, 8:00 AM-12:00 PM<br />

Evaluation <strong>of</strong> Ile655Val HER2 polymorphism associated with cardiac<br />

toxicity following the administration <strong>of</strong> trastuzumab in women with breast<br />

cancer. Presenting Author: Caroline Diorio, URESP, Centre de Recherche<br />

FRSQ du CHA Universitaire de Québec, Quebec City, QC, Canada<br />

Background: Trastuzumab is well tolerated without major side effects<br />

except for cardiac toxicity. Although a number <strong>of</strong> clinical parameters have<br />

been associated with trastuzumab-associated cardiac toxicity (TACT), there<br />

is some indication that genetic variation <strong>of</strong> the HER2 gene may play a toxic<br />

role in a population <strong>of</strong> metastatic breast cancer patients. However, this<br />

finding needs confirmation and we looked at a population <strong>of</strong> non-metastatic<br />

breast cancer. This study aimed to evaluate the association between<br />

cardiac toxicity and HER2 [Ile655Val] polymorphism in non-metastatic<br />

breast cancer patients treated with trastuzumab. Methods: The Ile655Val<br />

HER2 polymorphism was assessed in 73 women using TaqMan technology.<br />

For this study, the genotyping was performed using DNA extracted from<br />

normal breast tissue located at more than 1 cm <strong>of</strong> any other lesions. Charts<br />

review was used to collect information on TACT which was defined as any<br />

decline in LVEF � 10 % from the baseline to � 55 % or a decline in LVEF<br />

� 5%to�50 % (lower limit <strong>of</strong> normal). The Fisher exact test was used to<br />

evaluate the association between cardiac toxicity and HER2 polymorphism.<br />

Results: No deviation from the Hardy-Weinberg equilibrium has been<br />

observed for the allele and genotype frequencies. The distribution <strong>of</strong> HER2<br />

polymorphism was 3 Val/Val (4%), 18 Ile/Val (25%) and 52 Ile/Ile (71%).<br />

In this population, 19% (14/73) developed a cardiac toxicity. We found<br />

that 29% (6/21) <strong>of</strong> Ile/Val or Val/Val carriers compared to 15% (8/52) <strong>of</strong><br />

Ile/Ile carriers showed TACT, but this association did not reach statistical<br />

significance (P � 0.21). Conclusions: HER2 Ile655Val polymorphism may<br />

be an efficient marker <strong>of</strong> TACT considering this tendency with this small<br />

cohort <strong>of</strong> patients. Larger sample is needed to strengthen this conclusion,<br />

since this result may influence on prescribing decision for adjuvant<br />

chemotherapy and anti-HER2 therapy in HER2 positive patients.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


40s Breast Cancer—HER2/ER<br />

635 General Poster Session (Board #13F), Sat, 8:00 AM-12:00 PM<br />

Effect <strong>of</strong> heterogeneity <strong>of</strong> HER2 expression on brain metastases <strong>of</strong> breast<br />

cancer. Presenting Author: Mari Hosonaga, Division <strong>of</strong> Gene Regulation,<br />

Institute for Advanced Medical Research, Keio University, Tokyo, Japan<br />

Background: HER2-overexpressing or triple-negative [ER(-)/PR(-)/HER2(-)]<br />

breast cancers are associated with increased risk <strong>of</strong> brain metastases. The<br />

mechanisms leading to metastasis in each subtype are not well known.<br />

Methods: We introduced the wild-type HER2 gene into MDA-MB-231-luc-<br />

D3H2LN (231-Luc) cells, which are triple-negative breast cancer cells,<br />

and established HER2-expressing (63.2%) cells as 231-Luc-HER2 cells.<br />

We investigated the tumor formation following orthotopic inoculation and<br />

brain metastasis following intracardiac injection into nude mice. Metastasis<br />

was detected by bioluminescence imaging and confirmed in H&E<br />

staining and immunohistochemistry <strong>of</strong> vimentin and HER2 expressions.<br />

Flow cytometry analysis was used to detect the proportion <strong>of</strong> CD44�/CD24cells,<br />

a maker for stem-like breast cancer cells. Results: 231-Luc-HER2<br />

cells formed larger tumors in orthotopic xenograft models compared to<br />

231-Luc cells, however, no significant difference was observed in proliferation<br />

in vitro. Neither 231-Luc-HER2 nor 231-Luc metastasized in the brain<br />

from the breast after orthotopic inoculation. After intracardiac injections <strong>of</strong><br />

the 231-Luc-HER2 cells, brain metastasis developed (7/13 mice, 53.8%).<br />

Immunohistochemical analysis revealed that most metastasized cells<br />

expressed HER2, although we had injected a mixture <strong>of</strong> HER2-positive and<br />

HER2-negative cancer cells. Interestingly, administering Lapatinib, a dual<br />

EGFR and HER2 tyrosine kinase inhibitor, effectively prevented HER2positive<br />

cells to colonize the brain. However, the HER2-negative 231-Luc-<br />

HER2 cells developed into brain metastases. In fact, the 231-Luc cells,<br />

which are HER2-negative, also metastasized in the brain (10/16 mice,<br />

62.5%). Flow cytometry analysis <strong>of</strong> the 231-Luc-HER2 cells showed that<br />

HER2-positive cells decreased the population <strong>of</strong> CD44�/CD24- (HER2�/<br />

CD44�/CD24-: 86.8% and HER2-/CD44�/CD24-: 96.3%). Conclusions:<br />

The mechanism <strong>of</strong> brain metastases <strong>of</strong> HER2-positive breast cancer cells is<br />

different from that <strong>of</strong> HER2-negative breast cancer cells. It is therefore<br />

important to consider an additional therapeutic approach when dealing<br />

with HER2-negative cells in tumors having the heterogeneity <strong>of</strong> HER2<br />

expression.<br />

637 General Poster Session (Board #13H), Sat, 8:00 AM-12:00 PM<br />

The effects <strong>of</strong> lapatinib and neratinib on HER2 protein levels in breast<br />

cancer cell lines. Presenting Author: Denis Collins, National Institute for<br />

Cellular Biotechnology, Dublin City University, Dublin, Ireland<br />

Background: HER2, a member <strong>of</strong> the c-erbB receptor tyrosine kinase family,<br />

is over-expressed (HER2 gene amplification/IHC 3�, �30% cells) in<br />

approx. 25% <strong>of</strong> breast cancers. Pre-clinical studies have shown that the<br />

HER2-targeted small molecule tyrosine kinase inhibitor (TKI) lapatinib<br />

(LAP) can increase HER2 levels in cell line models and can potentiate<br />

trastuzumab-mediated antibody dependent cell-mediated cytotoxicity. To<br />

assess the potential effects <strong>of</strong> the next generation TKI neratinib (NER) on<br />

expression <strong>of</strong> HER2 in breast cancer, this study compares the effects <strong>of</strong><br />

LAP and NER on HER2 protein levels in the HER2-amplified SKBR3 and<br />

HER2-non-amplified T47D cell lines in vitro. Methods: SKBR3 and T47D<br />

were treated with LAP or NER (0.2, 1 and 2 �M) for 12, 24 and 48<br />

hours.HER2 protein levels were determined by ELISA, immunoblotting and<br />

high content analysis (HCA). HER2 protein was examined using two<br />

antibodies targeting the extracellular domain (ECD) and the intracellular<br />

domain (ICD) <strong>of</strong> HER2. pHER2, EGFR/pEGFR, MAPK/pMAPK and AKT/<br />

pAKT levels were determined by immunoblotting. Proliferation studies<br />

utilized an acid phosphatase-based assay. Results: ELISA analysis confirmedsignificantly<br />

lower HER2 expression in T47D (44 �/- 17 pg/�g)<br />

compared to SKBR3 (748 �/- 296 pg/�g), p � 0.015. NER proved a more<br />

potent inhibitor <strong>of</strong> pHER2 and pEGFR as analyzed by immunoblotting and<br />

in proliferation studies (NER IC50 - SKBR3 0.03 �/- 0.01 nM, T47D 199<br />

�/- 70 nM, LAP IC50 - SKBR3 20 �/- 1 nM, T47D 1.2 �/- 0.2 �M). LAP<br />

induced an increase in both ECD and ICD-containing HER2 protein levels<br />

in SKBR3 and T47D cells. No increase in HER-2 levels was observed with<br />

NER treatment, as determined by HCA and immunoblotting. EGFR protein<br />

levels increased in response to lapatinib in both cell lines. Conclusions: Our<br />

results suggest that LAP and NER have differing effects on HER2 protein<br />

levels in the models examined. NER provides greater inhibition <strong>of</strong> HER2<br />

signaling activity but does not increase HER2 protein levels as was<br />

observed with LAP. Further pre-clinical assessment <strong>of</strong> combinations <strong>of</strong> LAP<br />

and NER with HER2 and EGFR monoclonal antibody therapies is warranted<br />

in HER2-amplified, HER2-non-amplified and EGFR-expressing breast<br />

cancer models.<br />

636 General Poster Session (Board #13G), Sat, 8:00 AM-12:00 PM<br />

Adjuvant treatment <strong>of</strong> early-stage HER2� elderly breast cancer patients: A<br />

retrospective, multicenter French study. Presenting Author: Philippe Barthelemy,<br />

Hôpitaux Universitaires de Strasbourg, Strasbourg, France<br />

Background: Trastuzumab (T) is the standard <strong>of</strong> care for the adjuvant<br />

treatment <strong>of</strong> early stage, HER2� breast cancer (BC). However, few data are<br />

available for elderly HER2� breast cancer patients in this setting. In this<br />

current study, the patterns <strong>of</strong> care for elderly HER2� early stage BC in 7<br />

French cancer centres was evaluated. Methods: Medical records <strong>of</strong> all<br />

consecutive early stage HER2� BC patients over 70 years old treated<br />

between 2006 and 2011 among participating centres were retrospectively<br />

reviewed. Specific factors such as age, comorbidities, tumor stage, grade,<br />

ER/PR and HER2 status, treatment characteristics, follow-up and cardiotoxicity<br />

data were analysed. Results: One hundred and two patients were<br />

identified, median age 75.4 (range 70-95). Elderly patients presented<br />

mostly (57%) large tumors (pT �2), and positive lymph node involvement<br />

(n�61). Trastuzumab-based adjuvant treatment was administered in 62%<br />

<strong>of</strong> patients (n�63). 54% <strong>of</strong> patients (n�55) received adjuvant chemotherapy<br />

whereas five patients received neoadjuvant chemotherapy. Chemotherapy<br />

without T was administered in 2 additional patients. Anthracyclines<br />

(A)-Taxanes (Ta) combination-based chemotherapy was given in 27% <strong>of</strong><br />

patients (n�16), whereas 38% received a Ta-based chemotherapy (n�23),<br />

35% (n�19) an A-based chemotherapy. Five patients received singleagent<br />

T. Treatment delays for T were required in 37% <strong>of</strong> patients (n�23)<br />

among whom 15 and 8 permanently or temporarily stopped T, respectively.<br />

The most frequent reason for interrupting or delaying therapy was cardiotoxicity<br />

(n�12) as well as patients refusal (n�7). A � 10% decrease in LVEF<br />

was observed in 18/63 (29%) <strong>of</strong> patients, among whom T was stopped in<br />

12. After a median 33 months follow-up, the median progression-free<br />

survival was not reached in patients receiving T-based therapy. The 2 and<br />

3-year PFS rate were 94 and 89.5%, respectively. Conclusions: In routine<br />

practice only 62% <strong>of</strong> elderly early stage HER2� BC patients are treated<br />

with a neoadjuvant or adjuvant T-based regimen. However, less than 50%<br />

<strong>of</strong> all patients completed their therapy. A-based chemotherapy was<br />

administered in around 60% <strong>of</strong> treated patients, and could explain<br />

cardiotoxicity in this setting.<br />

638 General Poster Session (Board #14A), Sat, 8:00 AM-12:00 PM<br />

Lapatinib or trastuzumab? Which anti-HER2 treatment is more effective in<br />

the treatment <strong>of</strong> patients with HER2-positive breast cancer with brain<br />

metastases? An Anatolian <strong>Society</strong> <strong>of</strong> Medical Oncology Study. Presenting<br />

Author: Muhammet Ali Kaplan, Dicle Univercity Scholl <strong>of</strong> Medicine,<br />

Diyarbakir, Turkey<br />

Background: In the present study, we investigate that which treatment<br />

choice is more effective in the human epidermal growth factor receptor 2<br />

(HER2) positive breast cancer patients with brain metastases. Methods: Of<br />

405 metastatic breast cancer patients with brain metastases at referral<br />

centers in Turkey, 111 patients treated with lapatinib or trastuzumab after<br />

brain metastases eligible for the analyses were identified. Patients received<br />

both drugs consecutively or sequentially were excluded from the study.<br />

Results: Median age was 44 years (27-76) and 46 <strong>of</strong> the 111 patients<br />

(41.5%) had received lapatinib. Median time to development <strong>of</strong> brain<br />

metastases was 12.2 months (0-71). Sixteen patients (14.4%) had<br />

undergone surgery, 33 (29.7%) radiosurgery, and 108 (97.2%) whole<br />

brain radiation therapy (WBRT). Median overall survival (OS) after brain<br />

metastases was 15 months(95% confidence interval (CI): 12.3-17.6).<br />

Lapatinib usage was prolonged OS over trastuzumab alone (19.1 months vs<br />

12 months, p�0.039).Other parameters affecting the survival were Karn<strong>of</strong>sky<br />

performance score (KPS, �70), number <strong>of</strong> brain metastases (�3),<br />

extracranial metastases (�2), performed neurosurgery, and received radiosurgery.<br />

After correction for potential confounders, lapatinib therapy<br />

remained an independent positive predictor for survival [Odds ratio (OR),<br />

0.57; p�0.02). Conclusions: Although this retrospective multi-center study<br />

had several limitations, study results suggest that the usage <strong>of</strong> lapatinib<br />

after brain metastases prolonged survival compared to the usage <strong>of</strong><br />

trastuzumab. This result should be support with prospective studies.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


639 General Poster Session (Board #14B), Sat, 8:00 AM-12:00 PM<br />

Functional imaging <strong>of</strong> HER2-positive metastatic breast cancer using 64Cu DOTA-trastuzumab positron emission tomography (PET). Presenting Author:<br />

Joanne E. Mortimer, City <strong>of</strong> Hope Cancer Center/Beckman Research<br />

Institute, Duarte, CA<br />

Background: We are developing 64Cu-labeled trastuzumab (tras) as a PET<br />

imaging agent for women with HER2� beast cancer. To maximize image<br />

quality and minimize HER2 uptake in normal tissues, we pre-administered<br />

2 different doses <strong>of</strong> cold tras prior to injection <strong>of</strong> the imaging agent.<br />

Methods: Pts with advanced breast cancer were eligible if they had bx<br />

confirmed HER2 � disease, had not received anti-HER therapy for 6 mos.<br />

and had � 1 non-hepatic site <strong>of</strong> metastasis � 2 cm outside the biopsy site.<br />

All un-derwent staging FDG/PET and bone scan. Prior to injection <strong>of</strong><br />

64Cu-tras, pts. received 5 or 50 mg (2 and 4 pts) <strong>of</strong> cold tras. PET-CT was<br />

obtained at 21-25 h and 47-48 h over fields <strong>of</strong> view chosen in reference to<br />

18F-FDG scans. Uptake in organs and lesions were measured in terms <strong>of</strong><br />

body wt-normalized SUV. Lesions with intensity � adjacent tissue were<br />

considered positive on PET. The number <strong>of</strong> metastatic sites identified on<br />

64Cu-tras/PET was compared to the number on FDG/PET. Image quality<br />

was assessed according to differences between the 2 pretreatment doses.<br />

Results: Six pts have been imaged. Tras dose had a marked effect on liver<br />

uptake <strong>of</strong>64Cu [SUV � 23�3 (mean�sd) and 6.3�0.6 for 5 and 50 mg<br />

tras, respectively; P � 0.05]. 72 CT� lesions (32 bone, 28 nodal, 9 liver, 2<br />

breast, 1 lung) were included in D1 and/or D2 scans. Of these, 66 <strong>of</strong> 72<br />

(92%), 48 <strong>of</strong> 63 (76%) and 36 <strong>of</strong> 44 (82%) were detected on FDG, D1 and<br />

D2 scans, respectively. Lesion SUVmax was 9�5(n�37, range 3-23) on D1<br />

and 11�6 (n�27, range 1-29) on Day 2. 64Cu-tras-to-FDG SUVmax ratios<br />

were 1.0�0.8 (range 0.2-4.3) and 1.3�1.1 (range 0.2-3.5) for D 1 and 2.<br />

Lesion 64Cuuptake trendedliver � bone � nodes. Conclusions: 64Cu-tras/ PET visualizes HER2� lesions in bone, liver and, to a lesser degree, lymph<br />

nodes. Pre-administration <strong>of</strong> cold tras improves the quality <strong>of</strong> tras/PET,<br />

especially <strong>of</strong> intrahepatic lesions. In the next phase <strong>of</strong> study we will perform<br />

64Cu-tras/PET in pts with HER2 1� and 2� disease.<br />

641 General Poster Session (Board #14D), Sat, 8:00 AM-12:00 PM<br />

Outcome <strong>of</strong> HER2-positive breast cancer patients following metastatic<br />

relapse after adjuvant trastuzumab treatment since EMA regulatory approval.<br />

Presenting Author: Jean-Philippe Spano, Groupe Hospitalier Pitié<br />

Salpêtrière, AP-HP, Université Pierre et Marie Curie, Paris, France<br />

Background: Trastuzumab, a humanized monoclonal antibody against<br />

human epidermal growth factor receptor 2 (HER2) is approved in France<br />

since 2005 for HER2-positive breast cancer patients in the adjuvant<br />

setting. Efficacy <strong>of</strong> this treatment is clearly established but little is known<br />

related to patient outcome if they develop metastatic relapse after adjuvant<br />

trastuzumab. The aim <strong>of</strong> this study is to analyse these relapses with a<br />

special focus on efficacy <strong>of</strong> anti-HER2 treatment in this setting. Methods:<br />

We analysed 456 patients with HER2-positive early breast cancer treated<br />

with trastuzumab containing adjuvant therapy between 2006 and 2011 in<br />

two Departments <strong>of</strong> Medical Oncology from Université Pierre et Marie<br />

Curie, Paris, France. Data from patients with metastatic relapse were<br />

identified. Disease free survival (DFS) between end <strong>of</strong> trastuzumab and<br />

relapse, quality <strong>of</strong> response, and time to progression after anti-HER2 new<br />

treatment are reported. Results: Relapse rate was 3.3 % after adjuvant<br />

trastuzumab (15 pts, median age 53 years, ER� 40 %). DFS between end<br />

<strong>of</strong> trastuzumab and relapse was 244 days (95% CI: 91 - 397 days). One pt<br />

actually had a confirmed HER2-negative metastatic disease. The other 14<br />

pts were treated with anti-HER2 agents at relapse. On 12 evaluable pts, an<br />

objective response was observed in 8 pts, stabilisation in 2 pts and<br />

progression in 2 pts. Median time to progression after first line metastatic<br />

treatment was 222 days (95% CI: 114 - 330 days). Additional data about<br />

site <strong>of</strong> relapse and subsequent therapies are available. Conclusions:<br />

Relapse after adjuvant trastuzumab remains a rare event in this real life<br />

cohort <strong>of</strong> breast cancer pts. For these pts, we will present data <strong>of</strong> the<br />

patterns <strong>of</strong> relapse and response. These data may help us to determine if<br />

adjuvant trastuzumab has an impact on outcome <strong>of</strong> HER2 metastatic<br />

disease compared to data in trastuzumab naive metastatic HER2-positive<br />

breast cancer pts. We also plan to seek international collaboration to<br />

expand our analyses, as well as overall survival.<br />

Breast Cancer—HER2/ER<br />

41s<br />

640 General Poster Session (Board #14C), Sat, 8:00 AM-12:00 PM<br />

Trastuzumab treatment in patients with advanced breast cancer (ABC) confined<br />

to locoregional and skeletal sites: Results from a large noninterventional study.<br />

Presenting Author: Christian Jackisch, Klinikum Offenbach, Offenbach, Germany<br />

Background: In a prospective observation study routine trastuzumab (T) treatment<br />

in HER2� ABC was evaluated in a total <strong>of</strong> 1,687 patients (pts) since<br />

2001. This large number allows the subgroup analysis <strong>of</strong> patients with the<br />

involvement <strong>of</strong> specific sites. Methods: Information on extent <strong>of</strong> advanced<br />

disease was available in 1,674 pts (99%) with HER2 overexpressing ABC. Pts<br />

were either pre-treated (39%) or naïve to palliative chemotherapy (CT). The<br />

following subgroups and their characteristics were analysed: I: locally ABC only;<br />

II: bone metastases allowing for concomitant local tumour; III: other. Results:<br />

Locally advanced lesions were found to be the sole BC manifestation in 145 pts<br />

(8.7%), while bone metastases � locally ABC were found in 209 pts (12.5%).<br />

This leaves a comparative population <strong>of</strong> 1320 pts with metastases to other sites,<br />

predominantly visceral metastases. The table shows the baseline and treatment<br />

characteristics, as well as the long term outcome based on progression/death<br />

observed in 79%/56% <strong>of</strong> pts. Concurrent CT (74% <strong>of</strong> total group) predominantly<br />

consisted <strong>of</strong> a taxane (46%) or vinorelbin (24%), with no major differences<br />

between the groups. Conclusions: In this HER2� cohort, palliative pts with local<br />

lesions only show a comparatively high-risk pr<strong>of</strong>ile with respect to grading,<br />

hormone receptor status and adjuvant CT. Nevertheless, locally ABC only is a<br />

significant favourable prognostic factor in HER2� BC treated with T combinations,<br />

with a median survival <strong>of</strong> 4 to 5 years.<br />

ABC subgroups<br />

Parameter I II III<br />

Age [median, years] 57 59 59<br />

Age > 65 years [%] 27 26 28<br />

Grading G3 [%] 53 46 54<br />

Relapse-free interval [median, years] 2 2 2<br />

Hormone receptor positive [%] 51 71 60<br />

Local disease present [%] 100 28 30<br />

Adjuvant CT [%] 77 57 60<br />

Previous palliative CT [%] 26 28 42<br />

Previous palliative hormone therapy [%] 17 38 32<br />

> 1 year <strong>of</strong> T treatment [%] 54 58 47<br />

Concomitant hormone therapy [%] 32 44 31<br />

Complete remission [%] 43 12 13<br />

Progression-free survival [median, months] 23.5 18.7 10.3<br />

Progression-free survival rate at 2 years [%] 49 42 25<br />

Overall survival [median, months] 57.7 45.3 30.9<br />

642 General Poster Session (Board #14E), Sat, 8:00 AM-12:00 PM<br />

Estrogen receptor in HER2-positive early breast cancer: Two different diseases?<br />

Presenting Author: Eva Maria Ciruelos Gil, Medical Oncology Department,<br />

University Hospital 12 de Octubre, Madrid, Spain<br />

Background: HER2� breast cancer (BC) is a well characterized subtype <strong>of</strong> BC,<br />

due to the predictive value <strong>of</strong> HER2 overexpression for anti-HER2 targeted<br />

therapies. Nevertheless, around 50% <strong>of</strong> HER2� BC are ER� and clasiffied as<br />

luminal B/HER2�, but their biological and clinical behaviour may be different<br />

from HER2�/ER- tumors. Methods: We retrospectively reviewed 347 HER2�<br />

(Herceptest ��� or FISH�) early BC patients (see Table) diagnosed at our<br />

institution in 1997-2007, and were divided into two study groups: HER2�/ER�<br />

(group A) and HER2�/ER- (group B). ER� was defined if expressed in � 10%<br />

tumor cells. Results: See table below. Mean age: 54.7 y (44.6 – 65). Mean Ki 67<br />

was higher in B (37,2 vs 22,4%, p�0.0001). At the current FU, n¼ <strong>of</strong> events<br />

were insufficient to reach median DFS/OS. Mean DFS was 6.9 y (3.5 – 10.2);<br />

recurrent disease was higher (p 0.003) for B (54 pts, 43%) vs A (62 pts, 28%).<br />

5-year DFS estimates: 78.4 % (95% CI 72.3 – 83.3) and 62.3 % (95% CI 53 –<br />

70.27) for A and B, and 10-year DFS was 73.4% (95% CI 66.6 – 79) and 52.5%<br />

(95% CI 42.4 – 61.6) for A and B, respectively (p 0.0006). Most common<br />

recurrent sites were local (18) and bone (9) for HER2�/ER� and liver (8) and<br />

lung (8) for HER2�/ER-. Mean OS was 8.03y (5.4 – 10.8); 28 (12,6%) pts died<br />

in A, vs 26 (21%) in B (p 0.043). 5-year OS estimates: 93.9 % (95% CI 89.7 –<br />

96.4) and 87.6 % (95% CI 80.3 – 92.3) for A and B, and 10-year DFS was<br />

84.2% (95% CI 77.5 – 89.0) and 74.1% (95% CI 63.4 – 82.2) for A and B,<br />

respectively (p 0.01). Conclusions: ER expression in HER2� early BC defines<br />

two clinically distinct diseases with different long-term prognosis. These data<br />

may help to better individualize adjuvant therapies and future clinical trial<br />

designs.<br />

AN(%) BN(%)<br />

N 222 125<br />

T1<br />

104 (47)<br />

44 (35,2)<br />

T2<br />

94 (92,5)<br />

63 (50,4)<br />

T3<br />

9 (4)<br />

13 (10,4)<br />

T4<br />

14 (6)<br />

5 (4)<br />

Missing<br />

1<br />

N0<br />

120 (58,5)<br />

60 (51,2)<br />

N1<br />

46 (22,4)<br />

26 (22,2)<br />

N2<br />

28 (13,6)<br />

22 (18,8)<br />

N3<br />

11 (5,3)<br />

9 (7,7)<br />

Missing<br />

17<br />

8<br />

Neoadjuvant CT 50 (22,5) 30 (24)<br />

Adjuvant CT 126 (57) 88 (70,4)<br />

Type <strong>of</strong> CT<br />

44 (25)<br />

30 (25,4)<br />

No A, no T<br />

74 (42)<br />

57 (48,3)<br />

A, no T<br />

34 (19,3)<br />

27 (23)<br />

A and T<br />

Other<br />

24 (13,6)<br />

4 (3,3)<br />

Adjuvant RT 172 (77,4) 96 (77)<br />

Adjuvant trastuzumab 33 (14,8) 28 (22,4)<br />

Adjuvant HT<br />

213 (96)<br />

16 (13)<br />

Tam �/- LHRHa<br />

77 (36)<br />

8 (50)<br />

Aromatase inh.<br />

136 (64)<br />

8 (50)<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


42s Breast Cancer—HER2/ER<br />

643 General Poster Session (Board #14F), Sat, 8:00 AM-12:00 PM<br />

Correlation <strong>of</strong> TOP2A expression in HER2-positive breast cancer with<br />

pathologic response and clinical outcomes in patients undergoing neoadjuvant<br />

treatment based on anthracycline. Presenting Author: Elizabeth S.<br />

Santos, Hospital A.C. Camargo, São Paulo, Brazil<br />

Background: Factors predictive <strong>of</strong> response to anthracyclines can help<br />

selecting patients who really benefit from its use. TOP2A (a target <strong>of</strong><br />

anthracyclines) and HER2 genes are both amplified in 40% <strong>of</strong> breast<br />

cancers. Data from literature are conflicting regarding the potential <strong>of</strong><br />

TOP2A as a predictor <strong>of</strong> response to anthracyclines. Methods: We retrospectively<br />

analyzed data on the outcome <strong>of</strong> patients undergoing neoadjuvant<br />

anthracycline-based chemotherapy and evaluated nuclear TOP2A protein<br />

expression in breast cancer biopsies by immunohistochemistry (IHC) and<br />

reviewed the pathological responses after completion <strong>of</strong> neoadjuvant<br />

treatment. Results: From 2002 to 2011, data from 82 patients was<br />

reviewed. Forty nine patients had the paraffin blocks corresponding to the<br />

pre-treatment biopsy. All biopsies had some degree <strong>of</strong> TOP2A IHC<br />

expression. Patients were considered positive for TOP2A if the level <strong>of</strong><br />

expression in IHC was higher than 10%. This was found in 77.6% <strong>of</strong> cases.<br />

Forty percent <strong>of</strong> the patients achieved pathologic complete response in<br />

pos-treatment surgical specimens. Correlation was observed between<br />

TOP2A IHC expression and tumor pathological response (p0.02). It was<br />

observed a difference in pathological complete response in favour <strong>of</strong> the<br />

group positive for TOP2A (47.4% vs. 22.7%), although not significant<br />

(p�0.09). No difference in PFS and OS was observed between patients<br />

with complete response after median follow-up <strong>of</strong> 34 months. Conclusions:<br />

The expression <strong>of</strong> TOP2A seems to be a promising marker to predict<br />

pathological response to antracycline-based neoadjuvant chemotherapy in<br />

HER2 breast cancer; however these findings would be better evaluated in<br />

prospective trials.<br />

645 General Poster Session (Board #14H), Sat, 8:00 AM-12:00 PM<br />

Relationship between changes in left ventricular ejection and use <strong>of</strong><br />

angiotensin-converting enzyme inhibitors and/or betablockers during adjuvant<br />

trastuzumab chemotherapy in early breast cancer: Data from the real<br />

world. Presenting Author: Francesco Giotta, Medical Oncology Department,<br />

National Cancer Institute <strong>of</strong> Bari, Bari, Italy<br />

Background: Adjuvant trastuzumab chemotherapy (aTrastC) improves DFS<br />

<strong>of</strong> pts with breast cancer and overexpression <strong>of</strong> HER-2. However, cardiovascular<br />

complication (CV) such as heart failure (HF) or significant left<br />

ventricular ejection fraction (LVEF) reduction may appear especially in<br />

those pts at increased CV risk. Methods: 253 women treated with aTrastC<br />

for EBC in 7 italian oncologic centers during the period 2008-2009<br />

entered in a multicenter registry and were retrospectively studied in 4<br />

subgroups according to the treatment with ACEi and/or BB. Occurrence <strong>of</strong><br />

symptoms <strong>of</strong> HF and/or decrease in 10 points % <strong>of</strong> LVEF were recorded<br />

during the follow up. LVEF was measured at baseline and 3-6-9-12<br />

months. Results: Symptoms <strong>of</strong> HF occurred in 2% <strong>of</strong> pts who did not take<br />

either ACEi/ARB or BB. HF event-rate was similarly increased in pts<br />

receiving one or both medications, partially justified by the increased CV<br />

risk in these subgroups. Prevalence <strong>of</strong> decrease in LVEF � 10 points% was<br />

similar in all study subgroups. Trends in LVEF were characteristics for each<br />

study subgroup: at 3-month evaluation a significant decrease in LVEF was<br />

detected in ACEi/ARB and ACEi/ARB � BB group. Multiple logistic<br />

regression analysis showed that combined ACEi/ARB � BB therapy<br />

depended on history <strong>of</strong> hypertension (OR 36.7, CI 4.3-315.5) and<br />

reduction <strong>of</strong> LVEF from baseline to 3-month evaluation (OR 0.88, CI<br />

0.78-0.97) (best prediction – 3.5 points %, AUC 0.78, IC 0.65-0.91).<br />

Conversely, no association was found between changes in LVEF and<br />

ACEi/ARB or BB therapy alone. LVEF recovery from 3 to 12-month<br />

evaluation was inversely related to the changes in LVEF from baseline to<br />

3-month evaluation and did not depend on any pharmacological treatment.<br />

Conclusions: In clinical practice, a history <strong>of</strong> hypertension and changes in<br />

LVEF during the first 3 months <strong>of</strong> aTrastC for early breast cancer influence<br />

the use <strong>of</strong> ACEi/ARB and BB. This behaviour may explain the characteristic<br />

trend in LVEF showed in the subgroups <strong>of</strong> patient receiving or receiving not<br />

ACEi/ARB or BB. Subsequent recovery <strong>of</strong> LVEF does not seem to be related<br />

to any medical treatment.<br />

644 General Poster Session (Board #14G), Sat, 8:00 AM-12:00 PM<br />

Time to brain metastasis (TTBM) from initial diagnosis <strong>of</strong> distant metastasis<br />

in breast cancer: Prediction <strong>of</strong> TTBM according to breast cancer<br />

subtypes and treatment effect. Presenting Author: Yeon Hee Park, Division<br />

<strong>of</strong> Hematology-Oncology, Department <strong>of</strong> Medicine, Samsung Medical<br />

Center, Sungkyunkwan University School <strong>of</strong> Medicine, Seoul, South Korea<br />

Background: Brain metastasis (BM) from breast cancer (BC) is a growing<br />

problem. About 10-15% patients with metastatic breast cancer (MBC)<br />

developed BM, with a 1 year survival <strong>of</strong> 20%. Currently, one-third <strong>of</strong> MBC<br />

patients with either HER2�ve tumors or triple negative BC (TNBC) tumors<br />

develop brain metastases. We hypothesized that MBC patients may<br />

predispose to BM differently during the disease courses according to BC<br />

subtype and treatment. We analyzed TTBM from initial diagnosis <strong>of</strong> distant<br />

metastasis how BC subtypes and treatment affect on TTBM. Methods: We<br />

retrospectively investigated 189 consecutive patients who were diagnosed<br />

with BM from BC between 2000 and 2009 at Samsung Medical Center. We<br />

analyzed TTBM according to BC subtypes and treatment effect. Results:<br />

The median age <strong>of</strong> 189 BM patients from BC was 48 (range 26-87) years.<br />

The numbers <strong>of</strong> patients with hormone receptor (HR)�ve and HER2-ve,<br />

HER2�ve irrespective <strong>of</strong> HR status, and TNBC were 43 (22.8%), 88<br />

(46.6%), and 58 (30.7%), respectively. Median TTBM <strong>of</strong> all 189 patients<br />

was 10.4 (95% CI, 7.7-13.1) months. We analyzed TTBC into four groups<br />

considering BC subtypes and treatment; HR�ve/HER2-ve (n�43),<br />

HER2�ve with trastuzumab (T) (n�59), HER2�ve without T (n�29), and<br />

TNBC patients (n�58). The median TTBMs for each group were 17.7<br />

months, 13.8 months, 4.3 months, and 2.9 months, respectively<br />

(p�0.002). BM as an initial site <strong>of</strong> distant metastasis was much more<br />

common in HER2�ve without T and TNBC patients than in the other<br />

patients’ groups (40.2% vs 20.6%, p� 0.003). Conclusions: TTBMs were<br />

much shorter in patients with HER2�ve without T and TNBCs than in other<br />

BC patients. Different approaches for evaluation and therapeutic strategy<br />

for BM at the time <strong>of</strong> distant metastasis may be considered for these<br />

populations.<br />

TPS646 General Poster Session (Board #15A), Sat, 8:00 AM-12:00 PM<br />

An open-label randomized, multicenter, phase II study on neoadjuvant<br />

treatment with trastuzumab plus docetaxel versus trastuzumab plus<br />

docetaxel plus bevacizumab according to positron emission tomography<br />

(PET) value modification in patients with early stage HER2-positive breast<br />

cancer (AVATAXHER): Design description. Presenting Author: Alexandre<br />

Cochet, Department <strong>of</strong> Nuclear Medicine, Centre Georges-Francois Leclerc,<br />

Dijon, France<br />

Background: For patients with early HER2� breast cancer at diagnosis,<br />

addition <strong>of</strong> trastuzumab (T) to 6 cycles <strong>of</strong> preoperative docetaxel (D) can<br />

reach a pathological complete response (pCR) in ~50% <strong>of</strong> cases, and a high<br />

rate <strong>of</strong> conservative surgery. pCR can be predicted by changes <strong>of</strong> Fluorodeoxyglucose<br />

(FDG) tumor uptake evaluated by Positon Emission Tomography<br />

(PET) after one cycle <strong>of</strong> therapy. In order to increase this pCR rate, adding<br />

an antiangiogenic compound could be considered. Pre-clinical and phase<br />

I-II data support that the combination <strong>of</strong> bevacizumab (B) and T is<br />

synergistic and safe when patients are chemotherapy naïve. The neoadjuvant<br />

AVATAXHER trial (EUDRACT 2009-013410-26) investigates the<br />

potential increase <strong>of</strong> pCR rate by combining B with T and D for patients with<br />

HER2� breast cancer who are not predicted for pCR by FDG PET. Methods:<br />

In this multicenter, open-label, phase II trial, 2 phases are planned after a<br />

selection period: phase I: all patients receive two cycles <strong>of</strong> therapy<br />

combining T (8 mg/kg at the first cycle, then 6 mg/kg) and D (100 mg/m2 ).<br />

FDG PET is also performed within 7 days before cycle 1 (baseline) and less<br />

than 3 days before cycle 2 in order to calculate changes <strong>of</strong> the tumor FDG<br />

uptake between baseline and after cycle 1 (�SUV). Phase 2: if �SUV�70%,<br />

patients will continue to receive T and D for (cycles 3 to 6: D 100 mg/m2 �<br />

T 6 mg/kg); if �SUV�70%, patients are randomized 2:1 to arm A (cycles 3<br />

to 6 D 100 mg/m2 � T 6 mg/kg � B 15 mg/kg) or arm B ( cycles 3 to 6: D<br />

100 mg/m2 � T 6 mg/kg). The primary endpoint is pCR rate evaluated<br />

post-surgery 4 to 6 weeks after the last treatment <strong>of</strong> cycle 6. Enrolment<br />

began in May 2010 and 125 patients were to be recruited in 26 sites.<br />

According to the hypothesis that 60% <strong>of</strong> patients will have a �SUV�70%,<br />

it is presumed that 72 patients will be randomized. There are currently 107<br />

patients included (as <strong>of</strong> 06 January 2012 ), 95 <strong>of</strong> them reached the phase<br />

1; 52 <strong>of</strong> them (55%) showed a �SUV�70% and after randomization 34<br />

were included in arm A and 18 in arm B.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


TPS647 General Poster Session (Board #15B), Sat, 8:00 AM-12:00 PM<br />

LUX-breast 3: Randomized phase II study <strong>of</strong> afatinib alone or with<br />

vinorelbine versus investigator’s choice <strong>of</strong> treatment in patients (pts) with<br />

HER2-positive breast cancer (BC) with progressive brain metastases (BM)<br />

after trastuzumab or lapatinib-based therapy. Presenting Author: Heikki<br />

Joensuu, Department <strong>of</strong> Oncology, Helsinki University Central Hospital,<br />

Helsinki, Finland<br />

Background: Approximately 1/3 <strong>of</strong> pts with HER2-positive, advanced BC<br />

will develop BM with a poor prognosis. Efficacy <strong>of</strong> systemic therapies is<br />

limited since few drugs can readily cross the blood-brain barrier. BC BM<br />

represents a high unmet medical need for which novel targeted therapies<br />

are needed. Afatinib is a small molecule, irreversible ErbB Family Blocker<br />

that has shown preclinical activity in trastuzumab-resistant cell lines and<br />

HER2-positive tumor xenografts. Its clinical efficacy has been demonstrated<br />

in pts with heavily pre-treated, HER2-positive metastatic BC, who<br />

progressed after trastuzumab therapy, with partial responses in 10% and<br />

clinical benefit in 46% <strong>of</strong> pts. In a Phase I afatinib trial a sustained PR was<br />

observed in a NSCLC pt with BM, and in a phase II trial in NSCLC, pts with<br />

or without BM achieved comparable outcome on afatinib. The purpose <strong>of</strong><br />

this study (NCT01441596) is to investigate whether afatinib alone or in<br />

combination with vinorelbine has any effect on HER2-positive BC with BM<br />

after failure <strong>of</strong> prior trastuzumab or lapatinib. Methods: Key eligibility<br />

criteria: histologically confirmed HER2-positive BC; recurrent/progressive<br />

BM during/after prior trastuzumab or lapatinib therapy; �1 measurable BM<br />

(�10 mm on T1-weighted, gadolinium-enhanced MRI); extra-CNS lesions<br />

allowed; prior surgery, whole brain radiotherapy or stereotactic radiosurgery<br />

allowed; ECOG 0–2. Eligible pts are stratified by: ECOG score 0–1 vs 2; No<br />

<strong>of</strong> BM � 3vs�3; prior lapatinib therapy. Pts are randomized to (n �<br />

40/arm): Arm A: afatinib (40 mg/d oral); Arm B: afatinib (40 mg/d oral) �<br />

vinorelbine (25 mg/m2 / week); Arm C: investigator’s choice <strong>of</strong> medical<br />

treatment approved for metastatic BC. Primary endpoint: pt benefit at 12<br />

weeks (i.e. absence <strong>of</strong> CNS/extra CNS disease progression as per RECIST<br />

1.1) and no tumour-related worsening <strong>of</strong> neurological signs/symptoms or<br />

increase in steroid dosage. Secondary endpoints: PFS, OS, safety and<br />

afatinib concentration in plasma/cerebrospinal fluid in �12 pts. The study<br />

began in October 2011 and enrollment is ongoing.<br />

TPS649 General Poster Session (Board #15D), Sat, 8:00 AM-12:00 PM<br />

LUX-breast 1: Randomized, phase III trial <strong>of</strong> afatinib and vinorelbine versus<br />

trastuzumab and vinorelbine in patients with HER2-overexpressing metastatic<br />

breast cancer (MBC) failing one prior trastuzumab treatment.<br />

Presenting Author: Nadia Harbeck, University <strong>of</strong> Cologne, Cologne, Germany<br />

Background: Afatinib is an ErbB Family Blocker that irreversibly blocks<br />

signaling from all relevant ErbB Family dimers. Afatinib is being developed<br />

in EGFR (ErbB1)-driven (NSCLC/HNSCC) and HER2 (ErbB2)-driven (breast)<br />

malignancies. In trastuzumab-resistant, HER2-positive (SUM190) xenografts,<br />

afatinib showed antitumor activity which was superior to lapatinib<br />

and increased by addition <strong>of</strong> IV vinorelbine. Afatinib monotherapy also<br />

demonstrated clinical activity (progression-free survival [PFS] � 15.1 wk;<br />

objective response [OR] � 10%) in an open-label, single-arm, Phase II trial<br />

in patients with HER2-positive MBC after progression on trastuzumab.<br />

Methods: LUX-Breast 1 (NCT01125566) is a Phase III, open-label,<br />

multicenter trial evaluating the efficacy and safety <strong>of</strong> afatinib � vinorelbine<br />

vs. trastuzumab � vinorelbine in patients with HER2-overexpressing MBC<br />

who progressed on, or after one prior trastuzumab-based treatment regimen.<br />

Patients are randomized 2:1 to afatinib (40 mg/d oral) � vinorelbine<br />

(IV 25 mg/m2 /wk) or trastuzumab (IV 2 mg/kg/wk after 4 mg/kg loading<br />

dose) � vinorelbine (IV 25 mg/m2 /wk). Patients receive continuous<br />

treatment in the absence <strong>of</strong> disease progression or adverse events. Key<br />

eligibility criteria include histologically-confirmed HER2-positive BC, stage<br />

IV disease; no prior treatment with vinorelbine or HER2-targeted treatment<br />

other than trastuzumab; progression on one prior trastuzumab based<br />

regimen in either the adjuvant (or �12 months after trastuzumab completion)<br />

or first-line (or �6 months after trastuzumab completion) setting;<br />

prior anthracycline and/or taxane chemotherapy; ECOG score 0 or 1. The<br />

primary endpoint is PFS and secondary endpoints include OR, overall<br />

survival and safety. Serum and tissue biomarkers will be assessed on<br />

archival tissue. HER-receptor and HER-ligand reprogramming, putative<br />

resistance markers and EGFR response signature will be explored in fresh<br />

tissue biopsies. Enrollment began in June 2010 and is ongoing, targeting<br />

�240 sites with a recruitment target <strong>of</strong> 780 patients.<br />

Breast Cancer—HER2/ER<br />

43s<br />

TPS648 General Poster Session (Board #15C), Sat, 8:00 AM-12:00 PM<br />

BOLERO-1: A randomized, phase III, double-blind, placebo-controlled<br />

multicenter trial <strong>of</strong> everolimus in combination with trastuzumab and<br />

paclitaxel as first-line therapy in women with HER2-positive (HER2� ),<br />

locally advanced or metastatic breast cancer (BC). Presenting Author: Sara<br />

A. Hurvitz, UCLA Hematology, Oncology and Translational Research in<br />

Oncology (TRIO), Los Angeles, CA<br />

Background: Up to 25% <strong>of</strong> BCs overexpress human epidermal growth factor<br />

receptor 2 (HER2). Patients with HER2 � disease have a higher rate <strong>of</strong><br />

relapse and shorter overall survival (OS). Trastuzumab, a monoclonal<br />

antibody targeting HER2, is the standard <strong>of</strong> care and improves OS for<br />

HER2 � BC, but acquired resistance is common. The combination <strong>of</strong><br />

trastuzumab and paclitaxel has shown good tolerability and excellent<br />

objective response rates (ORR) in up to 84% <strong>of</strong> patients with HER2 �<br />

metastatic BC (MBC) (Gasparini G, et al. BCRT. 2007;101:355-65).<br />

Everolimus is an orally bioavailable inhibitor <strong>of</strong> mammalian target <strong>of</strong><br />

rapamycin (mTOR), a protein kinase central to multiple protein synthesis<br />

pathways and implicated in trastuzumab resistance. Everolimus-containing<br />

regimens have shown promising results in patients with ER � , HER2 –<br />

advanced BC in phase II/III trials; everolimus plus trastuzumab/paclitaxel<br />

or vinorelbine has shown encouraging ORR with an acceptable safety pr<strong>of</strong>ile<br />

in phase I/II trials in patients with HER2 � MBC. The present phase III study<br />

was undertaken to assess the effectiveness <strong>of</strong> adding everolimus to<br />

first-line standard therapy in HER2 � advanced BC. Methods: Women with<br />

HER2 � , locally advanced or metastatic BC who have received no prior<br />

systemic therapy (except endocrine) are eligible. Local disease must not be<br />

amenable to resection with curative intent. Women with a history <strong>of</strong> central<br />

nervous system metastasis are excluded. Patients are randomized 2:1<br />

(everolimus vs control) to receive standard therapy (paclitaxel and trastuzumab)<br />

plus everolimus (10 mg daily) or placebo. The primary endpoint is<br />

progression-free survival. Secondary endpoints include OS, ORR, and<br />

clinical benefit rate. Additional endpoints are safety, performance status,<br />

and biomarkers. This trial is sponsored by Novartis Pharmaceuticals and is<br />

registered (<strong>Clinical</strong>Trials.gov: NCT00876395). Enrollment began September<br />

2009, with a planned accrual <strong>of</strong> 717. The current accrual is 719, and<br />

the estimated primary completion date is October 2012.<br />

TPS650 General Poster Session (Board #15E), Sat, 8:00 AM-12:00 PM<br />

Open-label, phase II trial <strong>of</strong> afatinib, with or without vinorelbine (V), for the<br />

treatment <strong>of</strong> HER2-overexpressing inflammatory breast cancer (IBC).<br />

Presenting Author: Charles Swanton, Translational Cancer Therapeutics<br />

Lab, CR-UK, London, United Kingdom<br />

Background: IBC is an aggressive and rare form <strong>of</strong> BC (1–6% cases).<br />

Despite recent treatment advances, prognosis remains poor, with 3-yr<br />

survival rate <strong>of</strong> 40 vs 85% in non-IBC. IBC commonly lack ER/PR and more<br />

frequently harbour HER2 gene amplification (~40%), and EGFR overexpression<br />

(~30%) associated with poor prognosis. Recent data reported clinical<br />

efficacy <strong>of</strong> lapatinib, a reversible EGFR/HER2 inhibitor, in trastuzumab (T)<br />

naïve/resistant HER2 positive IBC. Afatinib is an irreversible ErbB Family<br />

Blocker with preclinical activity in T-resistant cell lines, HER2-positive<br />

tumor xenografts and HER2-negative SUM 149 xenograft model derived<br />

from an IBC cell line. Its clinical efficacy was shown in heavily pre-treated,<br />

HER2-positive metastatic BC pts who progressed after T, with PR in 10%<br />

and clinical benefit in 46% <strong>of</strong> pts. The purpose <strong>of</strong> this biomarker-driven<br />

study is to investigate efficacy and safety <strong>of</strong> afatinib alone and in<br />

combination with V upon progression on afatinib monotherapy and the<br />

genomic changes that occur through treatment in pts with T pretreated or<br />

naïve HER2-positive IBC. Methods: Pts are recruited in 2 parallel cohorts <strong>of</strong><br />

20 pts each: T-naïve and T failure. Key eligibility criteria: histologicallyconfirmed<br />

HER2-positive BC; investigator-confirmed IBC characterized by<br />

diffuse erythema and oedema (peau d’orange) while dermal lymphatic<br />

emboli or palpable mass are not necessary for diagnosis; no prior anti-<br />

HER2 therapy except T in the T failure cohort; no prior V; ECOG 0–2. All pts<br />

start afatinib monotherapy (40 mg/d oral) in 4-week cycles (<strong>Part</strong> A). Upon<br />

disease progression, pts may receive afatinib (40 mg/d) � V (IV 25<br />

mg/m2 /week) (<strong>Part</strong> B). Primary endpoint: <strong>Clinical</strong> Benefit Rate (CR, PR or<br />

SD) for �6 months. Secondary endpoints: objective response rate, PFS, OS<br />

and safety. Endpoints are assessed separately for <strong>Part</strong> A and <strong>Part</strong> B. Fresh<br />

biopsy and blood samples are taken prior to treatment and upon progression<br />

in <strong>Part</strong> A, to explore predictive markers <strong>of</strong> response/resistance to<br />

afatinib, to describe the IBC population which may benefit most, and for<br />

next generation sequencing and proteomic analysis. Enrollment ongoing<br />

since Aug 2011.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


44s Breast Cancer—HER2/ER<br />

TPS651 General Poster Session (Board #15F), Sat, 8:00 AM-12:00 PM<br />

LUX-breast 2: Phase II, open-label study <strong>of</strong> oral afatinib in HER2overexpressing<br />

metastatic breast cancer (MBC) patients (pts) who progressed<br />

on prior trastuzumab (T) and/or lapatanib (L). Presenting Author:<br />

Tamas Hickish, Royal Bournemouth Hospital, Bournemouth, United Kingdom<br />

Background: Management <strong>of</strong> HER2-overexpressing MBC has improved over<br />

the past decade. However, pts still develop resistance to currently available<br />

HER2-targeted therapies and novel effective treatments are increasingly<br />

required as dual targeted combinations are given in early treatment lines<br />

already. Current therapies focus on targeting HER2 and do not inhibit all<br />

relevant ErbB Family dimers. Afatinib is an oral, irreversible ErbB Family<br />

Blocker that inhibits signaling through activated EGFR (ErbB1), HER2<br />

(ErbB2) and ErbB4 receptors and transphosphorylation <strong>of</strong> ErbB3. Preclinical<br />

studies have demonstrated efficacy in T-sensitive and T-resistant<br />

human BC xenograft models dependent on ErbB signaling. Efficacy <strong>of</strong><br />

afatinib in a T-resistant SUM 190 xenograft model has been shown to be<br />

increased by addition <strong>of</strong> IV vinorelbine (V). Afatinib monotherapy has<br />

shown promising clinical benefit in 46% <strong>of</strong> HER2-overexpressing MBC pts<br />

who progressed on prior T, with 10% <strong>of</strong> pts achieving PR. Methods: This<br />

open-label Phase II trial (NCT01271725) investigates efficacy and safety<br />

<strong>of</strong> afatinib alone (40 mg/d) followed by afatinib ‘beyond progression’ plus<br />

chemotherapy in 120 pts with HER2-overexpressing MBC, who progressed<br />

on prior neoadjuvant and/or adjuvant T and/or L. Pts who progress on<br />

afatinib monotherapy receive afatinib plus either weekly paclitaxel (P) 80<br />

mg/m2 or V 25 mg/m2 . Eligible pts have confirmed HER2-overexpressing<br />

BC, stage IV disease measurable by RECIST 1.1, progressed on T and/or L<br />

therapy in either neoadjuvant and/or adjuvant setting, are eligible for<br />

retreatment with P or V and should not have been pretreated with P (�12<br />

months) or V, respectively. Exclusion criteria: inadequate cardiac, renal,<br />

hepatic and hematological function, pre-existing gastrointestinal dysfunction,<br />

rapidly progressing visceral disease, ILD and active brain metastases.<br />

The primary endpoint is objective response (OR) and secondary endpoints<br />

include best overall response, duration <strong>of</strong> OR and PFS; safety will be<br />

assessed separately for afatinib mono- and combination therapy. Patient<br />

enrollment began in May 2011 in ~35 sites and 5 countries.<br />

TPS653 General Poster Session (Board #15H), Sat, 8:00 AM-12:00 PM<br />

A combination <strong>of</strong> pertuzumab, trastuzumab, and vinorelbine for first-line<br />

treatment <strong>of</strong> patients with HER2-positive metastatic breast cancer: An<br />

open-label, two-cohort, phase II study (VELVET). Presenting Author: Edith<br />

A. Perez, Mayo Clinic, Jacksonville, FL<br />

Background: Pertuzumab (P) is a humanized monoclonal antibody directed<br />

against the dimerization domain <strong>of</strong> HER2: it prevents HER2 heterodimerization<br />

and thus activation <strong>of</strong> downstream signaling. Since P targets a<br />

different epitope than trastuzumab (H), a more comprehensive HER2<br />

blockade is achieved by combining the two agents. Data from CLEOPATRA<br />

showed improved efficacy for P and H plus docetaxel. The combination <strong>of</strong> P<br />

and H has not yet been assessed with other chemotherapy partners in the<br />

metastatic setting. H plus vinorelbine (V) has shown comparable efficacy to<br />

H plus docetaxel but with a superior safety pr<strong>of</strong>ile. VELVET will assess the<br />

overall response rate (ORR) <strong>of</strong> P with H�V in first-line patients (pts) with<br />

HER2-positive MBC. Co-administration <strong>of</strong> P and H within the same<br />

infusion bag will also be investigated as this could increase pt convenience<br />

by reducing administration and observation time. Methods: VELVET is a<br />

multicenter, open-label, two-cohort, Phase II trial. Pts with HER2-positive<br />

LABC or MBC not previously treated in the metastatic setting with<br />

non-hormonal anticancer therapy are eligible. Pts must have an LVEF<br />

�55% at baseline and an ECOG PS <strong>of</strong> 0 or 1. Study enrollment started in<br />

January 2012. A total <strong>of</strong> 210 pts will be included. Based on statistical<br />

assumptions, 95 pts must be evaluable per cohort, which assumes a<br />

withdrawal rate around 10%. Pts in Cohort 1 (the first 105 pts enrolled) will<br />

receive P and H sequentially and pts in Cohort 2 (the next 105 pts) will<br />

receive P and H in the same infusion bag at Cycle 2 onwards if drug<br />

administration in Cycle 1 was well tolerated. V will be given in both cohorts.<br />

Treatment duration is until disease progression or unacceptable toxicity.<br />

Study dose: P: 840 mg loading dose, 420 mg q3w (iv); H: 8 mg/kg loading<br />

dose, 6 mg/kg q3w (iv), and V: 25 mg/m2 Day 1 and 8 (first cycle) then<br />

30�35 mg/m2 Day 1 and 8 q3w (iv) (dose escalation at investigator’s<br />

discretion). The primary endpoint is ORR by independent assessment.<br />

Secondary endpoints include investigator assessment <strong>of</strong> ORR, time to<br />

response, duration <strong>of</strong> response, PFS, time to progression, overall survival,<br />

safety and tolerability, and QoL.<br />

TPS652 General Poster Session (Board #15G), Sat, 8:00 AM-12:00 PM<br />

A phase II trial <strong>of</strong> trabectedin (T) in patients with hormone receptorpositive,<br />

HER2-negative advanced breast cancer, according to xeroderma<br />

pigmentosum gene (XPG) expression. Presenting Author: Ahmad Awada,<br />

Institut Jules Bordet, Brussels, Belgium<br />

Background: Hormone receptor-positive, HER2-negative breast cancer (BC)<br />

is currently associated with 3-4 years overall survival in the metastatic<br />

setting and, after �2 relapses, therapeutic approaches are reduced. XPG<br />

expression is frequently modified in BC. T is a cytotoxic agent that forms a<br />

complex with the XPG, inducing cell apoptosis. As a single agent, T has<br />

shown anti-tumor activity in patients with poor prognosis BC, and a better<br />

response to T in BC patients with XPG RNA overexpression has been<br />

observed. Methods: This is an open-label, phase II study <strong>of</strong> T (1.3 mg/m2 in<br />

3-hour intravenous infusion every 3 weeks) in patients with hormone<br />

receptor-positive, HER2-negative advanced BC, according to their primary<br />

tumor’s XPG expression. Primary endpoint: to evaluate the efficacy <strong>of</strong> T in<br />

terms <strong>of</strong> progression free survival rate at 4 months (PFS4) according to the<br />

patient’s XPG expression. Secondary endpoints: Comparison <strong>of</strong> PFS,<br />

overall response rate, duration <strong>of</strong> response, overall survival and safety<br />

pr<strong>of</strong>ile in XPG-high and XPG-low patients. Assignment: BC patients who<br />

have previously received anthracyclins and/or taxanes and who progressed<br />

after 2-5 chemotherapy lines will be assigned according to their XPG<br />

expression from paraffin embedded tumor samples to stratum A (XPG-high<br />

[�3]) or to stratum B (XPG-low [�3]) (threshold was selected from median<br />

XPG expression values observed in a previous trial). Statistical methods: A<br />

two-stage design was chosen: at a first stage 20 patients will be enrolled in<br />

each stratum. A futility analysis (O’Brien Fleming boundary) based on the<br />

primary endpoint (PFS4) will be conducted once 40 evaluable patients<br />

have been recruited. If � 7 out <strong>of</strong> 20 patients achieve PFS4, recruitment<br />

will continue to a maximum sample size <strong>of</strong> 50 evaluable patients per<br />

stratum. If � 22 out <strong>of</strong> 50 patients achieve PFS4, T will be considered<br />

active in this group (alpha error: 0.025, power: 80%). To date, 35 patients<br />

(16 XPG-high and 15 XPG-low) have been enrolled from three countries<br />

and five centers. Recruitment is ongoing.<br />

TPS654^ General Poster Session (Board #16A), Sat, 8:00 AM-12:00 PM<br />

Pertuzumab in combination with trastuzumab plus an aromatase inhibitor<br />

in patients with hormone receptor-positive, HER2-positive metastatic<br />

breast cancer: A randomized phase II study (PERTAIN). Presenting Author:<br />

Mothaffar F. Rimawi, Baylor College <strong>of</strong> Medicine, Houston, TX<br />

Background: Resistance to endocrine therapy in patients (pts) with breast<br />

cancer remains a major clinical concern. Preclinical studies suggest that<br />

complete blockade <strong>of</strong> the hormone receptor (HR) combined with the<br />

inhibition <strong>of</strong> HER family members may be necessary to overcome resistance<br />

and improve clinical outcome in HR-positive and HER2-positive<br />

breast cancer (BC). The combination <strong>of</strong> pertuzumab (P) and trastuzumab<br />

(H) with docetaxel significantly improves patient outcome by blocking,<br />

more efficiently and completely, the HER signalling pathway. This benefit,<br />

however, may be smaller in HR-positive patients. PERTAIN is the first<br />

clinical trial to study whether a more potent blockade <strong>of</strong> the HER pathway<br />

with P and H in combination with endocrine therapy may restore or enhance<br />

endocrine sensitivity <strong>of</strong> HER2-positive BC and provide an effective treatment<br />

option in pts with HER2-positive and HR-positive MBC. Methods:<br />

PERTAIN is a multicenter, open-label, Phase II trial for post-menopausal<br />

women with HER2- and HR-positive BC, studying the efficacy <strong>of</strong> the<br />

combination <strong>of</strong> P plus H with an aromatase inhibitor (AI) as first-line<br />

therapy for MBC. Pts will be randomized 1:1 to Arm 1 (P: 840 mg loading<br />

dose, 420 mg q3w IV; H: 8 mg/kg loading dose, 6 mg/kg q3w IV; AI<br />

[anastrozole 1 mg or letrozole 2.5 mg qd po]) or Arm 2 (H � AI at same dose<br />

as Arm 1). Pts in either arm may also receive induction chemotherapy<br />

(docetaxel or paclitaxel) for up to 18 weeks at the investigator’s discretion.<br />

Study medication will be administered until disease progression or unacceptable<br />

toxicity. Pts must not have previously been treated with anti-<br />

HER2 agents except H and/or lapatinib in the (neo)adjuvant setting, and<br />

must have no CNS involvement or clinically significant cardiovascular<br />

disease. The primary endpoint is PFS, and secondary endpoints include<br />

overall survival, overall response rate, clinical benefit rate, duration <strong>of</strong><br />

response, time to response, safety and tolerability, and quality <strong>of</strong> life. The<br />

study opened in January 2012 and will recruit 250 pts. Analysis <strong>of</strong> the<br />

primary endpoint will be performed after 165 PFS events using the<br />

Kaplan-Meier approach.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


TPS655 General Poster Session (Board #16B), Sat, 8:00 AM-12:00 PM<br />

Docetaxel, carboplatin, trastuzumab, and bevacizumab (TCH�B) for earlystage<br />

HER2/neu(�) breast cancer and bone marrow micrometastases.<br />

Presenting Author: Joseph Baar, Seidman Cancer Center, Cleveland, OH<br />

Background: Bone marrow (BM) micrometastases confer a poor prognosis to<br />

early-stage breast cancer (BC) and HER-2/neu(�) BC is a high-risk<br />

phenotype. We have shown that BM-associated stromal cells support the<br />

survival <strong>of</strong> metastatic BC in microenvironmental niches and this activity is<br />

dependent on VEGF and CXCL-12. Therefore, the growth <strong>of</strong> nascent tumor<br />

cell clusters in BM might be impeded by disrupting stromal cells or their<br />

activating ligands, such as VEGF. There is also a positive association<br />

between HER-2/neu and VEGF expression in BC, thereby implicating VEGF<br />

in the aggressive phenotype <strong>of</strong> HER-2/neu overexpression. These observations<br />

support the use <strong>of</strong> combination therapies directed against both<br />

HER-2/neu and VEGF for the treatment <strong>of</strong> HER-2/neu(�) BC. We therefore<br />

hypothesize that an anti-VEGF agent (bevacizumab) given with docetaxel,<br />

carboplatin and trastuzumab (TCH) will eradicate the BM <strong>of</strong> HER-2/neu(�)<br />

BC micrometastases by disrupting stromal cell support within the micrometastatic<br />

BM niche. Methods: Subjects with AJCC Stage I-III HER2/<br />

neu(�) BC will undergo a single iliac-crest BM aspirate and biopsy. If the<br />

BM is positive for CK� micrometastases, up to 17 patients will receive<br />

TCH�B therapy (docetaxel at 75 mg/m2 IV and carboplatin at AUC 6 IV<br />

given every 3 weeks for 6 cycles; trastuzumab at 4 mg/kg IV loading dose,<br />

then 2mg/kg IV every week; bevacizumab at 15 mg/kg IV every 3 weeks).<br />

Four weeks after the 6th cycle <strong>of</strong> therapy, a repeat BM aspiration and biopsy<br />

will be performed to score for response <strong>of</strong> BC micrometastases to therapy.<br />

Patients with a complete clinical response in BM will continue with<br />

trastuzumab every 3 weeks at 6 mg/kg to a total <strong>of</strong> 1 year. Patients with<br />

persistent micrometastases will receive both trastuzumab and bevacizumab<br />

every 3 weeks to a total <strong>of</strong> 1 year <strong>of</strong> therapy. These patients will then<br />

have a repeat BM aspirate and biopsy at the end <strong>of</strong> the 1 year to score for<br />

final response. The primary objective <strong>of</strong> this clinical trial is to determine<br />

clinical response against BC micrometastases in BM. The second objective<br />

is to investigate the specific contribution <strong>of</strong> VEGF and CXCL-12 signaling to<br />

BM support <strong>of</strong> micrometastatic BC cells.<br />

TPS657 General Poster Session (Board #16D), Sat, 8:00 AM-12:00 PM<br />

A phase III clinical trial to compare trastuzumab (T) given concurrently with<br />

radiation therapy (RT) to RT alone for women with HER2� DCIS resected<br />

by lumpectomy (Lx): NSABP B-43. Presenting Author: Melody A. Cobleigh,<br />

National Surgical Adjuvant Breast and Bowel Project, Rush University<br />

Medical Center, Chicago, IL<br />

Background: Asignificant amount <strong>of</strong> DCIS is ER negative and/or overexpresses<br />

HER2. This provides an opportunity to test molecular therapy in<br />

DCIS. In xenograft models and cell lines, T boosts RT effectiveness. In<br />

T-treated HER2� patients, apoptosis occurs within 1 wk <strong>of</strong> single agent T<br />

use, with T found in ductal aspirates. Ample safety evidence for T exists. T<br />

given during whole breast irradiation (WBI) may improve results for<br />

Lx-resected HER2� DCIS. A trial to examine this question will enhance the<br />

understanding <strong>of</strong> breast tumor biology and the prevention <strong>of</strong> such tumors<br />

and could possibly extend breast-conserving surgery benefits for women<br />

with DCIS. Methods: After Lx for pure DCIS, each patient’s DCIS lesion is<br />

centrally tested for HER2 by IHC analysis. HER2 2� tumors undergo FISH<br />

analysis. HER2 3� or FISH� patients can be randomly assigned to 2 doses<br />

<strong>of</strong> T, 3 weeks apart during WBI or to WBI alone. Women �18 yrs. with a<br />

margin-clear Lx for pure DCIS, with ECOG status 0/1 who are and clinically<br />

or pathologically node negative are eligible. Centrally tested DCIS must be<br />

HER2 �. ER and/or PR status must be known before randomization.<br />

Primary aims are to determine if T decreases ipsilateral breast cancer<br />

recurrence, ipsilateral skin cancer recurrence, or ipsilateral DCIS. Secondary<br />

aims are to determine the benefit <strong>of</strong> T in preventing regional or distant<br />

recurrence and contralateral invasive breast cancer or DCIS. B-43 will<br />

determine if DFS, recurrence-free interval, and OS can be improved with<br />

the use <strong>of</strong> T. 2000 patients will be accrued over 7.9 yrs, with a definitive<br />

analysis <strong>of</strong> primary endpoints performed at163 ipsilateral breast cancer<br />

events (7.5 - 8 yrs. after protocol initiation) with an 80% power to detect a<br />

hazard reduction <strong>of</strong> 36%, from 1.73 ipsilateral breast cancer events per<br />

100 pt-yrs to 1.11 events per 100 pt-yrs. The 36% observed reduction in<br />

the hazard <strong>of</strong> IIBCR-SCR-DCIS on the T arm is based on a projection <strong>of</strong><br />

40% hazard reduction if the compliance were perfect, with a 10%<br />

noncompliance rate. As <strong>of</strong> 12-31-11, 763 patients have been randomized.<br />

NCT00769379 Grant support: PHS NCI-U10-CA-69651, -12027, and<br />

NCI P30-CA-14599 from the US NCI and Genentech, Inc.<br />

Breast Cancer—HER2/ER<br />

45s<br />

TPS656 General Poster Session (Board #16C), Sat, 8:00 AM-12:00 PM<br />

LCCC 1025: A phase II study evaluating the mTOR inhibitor everolimus<br />

with trastuzumab and vinorelbine to treat progressive HER2-positive breast<br />

cancer brain metastases. Presenting Author: Carey K. Anders, University <strong>of</strong><br />

North Carolina at Chapel Hill Lineberger Comprehensive Cancer Center,<br />

Chapel Hill, NC<br />

Background: HER2� breast cancer (BC) is an aggressive subset <strong>of</strong> BC with<br />

high rates <strong>of</strong> brain metastases (BM) and poor survival. A recent study<br />

illustrates activation <strong>of</strong> the PI3K/mTOR pathway in approximately twothirds<br />

<strong>of</strong> BCBM across all subtypes. Everolimus, a novel oral derivative <strong>of</strong><br />

rapamycin that selectively inhibits mTOR, is clinically-active in combination<br />

with trastuzumab (with or without chemotherapy) in advanced HER2�<br />

BC. Everolimus penetrates the blood brain barrier. We are evaluating the<br />

efficacy and safety <strong>of</strong> everolimus plus trastuzumab and vinorelbine in<br />

patients (pts) with HER2� BCBM. Methods: Pts with HER2� BCBM<br />

progressing following cranial radiation and measuring � 0.5cm are eligible<br />

for enrollment. Treatment with prior vinorelbine or mTOR inhibition is<br />

prohibited. Steroid treatment is allowed if stable or decreasing doses �7<br />

days prior to study entry. Pts with leptomeningeal disease are excluded. Pts<br />

receive everolimus (5mg orally daily), trastuzumab 2mg/kg and vinorelbine<br />

25mg/m2 (both IV days 1, 8, 15) <strong>of</strong> a 21 day cycle. Intra- and extracranial<br />

disease is assessed every 9 weeks by gadolinium-enhanced brain MRI and<br />

CT chest/abdomen/pelvis, respectively. The primary endpoint is intracranial<br />

response rate (RR) assessed by modified RECIST. Secondary objectives<br />

include extracranial RR (assessed by RECIST 1.1), intra- and<br />

extracranial time to progression, progression free survival, overall survival,<br />

quality <strong>of</strong> life, and correlative science endpoints. Statistical considerations:<br />

In this two stage design, a sample size <strong>of</strong> 28 evaluable pts (n�11 in<br />

Stage 1) has 80% power to detect a difference between the null (�5%<br />

intracranial RR) and alternative hypothesis (�5%) at a 0.05 significance<br />

level. If 1 pt has a CR, PR or SD for � 3 months in the 1st stage, 17<br />

additional pts will be enrolled. Assuming a 20% drop-out rate, 35 total pts<br />

will be enrolled. Correlative studies: Archival primary and/or metastatic<br />

tissues are required from all pts to evaluate intrinsic BC subtype, protein<br />

(p-AKT, pS6, PTEN) and gene expression to identify biomarkers that may<br />

be predictive <strong>of</strong> response or resistance to this novel combination<br />

(NCT01305941).<br />

TPS658 General Poster Session (Board #16E), Sat, 8:00 AM-12:00 PM<br />

HALT MBC: HER2 suppression with the addition <strong>of</strong> lapatinib to trastuzumab<br />

in HER2-positive metastatic breast cancer (LPT112515). Presenting<br />

Author: Nancy Lin, Dana-Farber Cancer Institute, Boston, MA<br />

Background: Dual blockade <strong>of</strong> HER2 with the combination <strong>of</strong> trastuzumab<br />

(T) and lapatinib (L) enhances antitumor activity in HER2-positive breast<br />

cancer (BC) preclinical models due to the complementary mechanisms <strong>of</strong><br />

action <strong>of</strong> the 2 agents. In patients (pts) with T-treated HER2-positive<br />

metastatic BC (MBC), treatment with the combination was associated with<br />

longer progression-free (PFS) and overall survival (OS) compared with L<br />

alone. In pts with stage II/III BC, preoperative treatment with the combination<br />

plus paclitaxel (P) resulted in significantly higher pathologic complete<br />

response rates compared with P combined with either agent alone. This<br />

evidence supports the concept <strong>of</strong> dual HER2 blockade as a treatment<br />

strategy for HER2-positive MBC. The present study is designed to evaluate<br />

whether the addition <strong>of</strong> L improves PFS among women with HER2-positive<br />

MBC receiving T as maintenance therapy. Methods: In this open-label,<br />

Phase III study, 280 pts will be stratified by line <strong>of</strong> treatment (first/second)<br />

and hormone receptor status (positive/negative), then randomized 1:1 to<br />

receive maintenance treatment with either L (1000 mg qd, continuously) in<br />

combination with T (6 mg/kg once every 3 weeks [q3w]) or T (6 mg/kg q3w)<br />

alone. Pts will receive study treatment until disease progression, death,<br />

discontinuation due to adverse events, or other reasons. The primary<br />

endpoint is PFS; secondary endpoints are OS, clinical benefit rate, and<br />

safety. Key eligibility criteria include pts with HER2-positive MBC who<br />

have completed 12 to 24 weeks <strong>of</strong> first- or second-line treatment with T<br />

plus chemotherapy with an objective response or stable disease at time <strong>of</strong><br />

chemotherapy discontinuation. Pts with stable brain metastases are<br />

eligible if entering the study on second-line treatment. Efficacy endpoints<br />

will be analyzed in the ITT population. A total <strong>of</strong> 193 PFS events is required<br />

to detect a 50% increase in median PFS (hazard ratio�0.67) for L plus T<br />

compared with T alone (median PFS 27 vs 18 weeks, respectively). The<br />

hypothesis will be tested using a 1-sided test with 80% power and a type I<br />

error <strong>of</strong> 0.025. The trial is currently open for accrual in the United States<br />

and Canada.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


46s Breast Cancer—HER2/ER<br />

TPS659 General Poster Session (Board #16F), Sat, 8:00 AM-12:00 PM<br />

WJOG6110B (ELTOP): Randomized phase II trial comparing trastuzumab<br />

plus capecitabine (HX) and lapatinib plus capecitabine (LX) in HER2positive<br />

metastatic breast cancer patients previously treated with trastuzumab<br />

and taxanes. Presenting Author: Toshimi Takano, Toranomon<br />

Hospital, Tokyo, Japan<br />

Background: In patients with HER2-positive metastatic breast cancer<br />

(MBC) previously treated with trastuzumab and taxanes, there are now two<br />

standard strategies: trastuzumab beyond progression or switch to lapatinib.<br />

A randomized trial comparing trastuzumab plus capecitabine (HX) and<br />

capecitabine alone (X) after first-line trastuzumab-based chemotherapy<br />

(GBG-26) showed that HX was superior to X in terms <strong>of</strong> time to progression<br />

(TTP). Another randomized trial comparing lapatinib plus capecitabine<br />

(LX) and X in patients previously treated with anthracyclines, taxanes, and<br />

trastuzumab (EGF100151) showed that LX was superior to X in terms <strong>of</strong><br />

TTP. To evaluate which strategy is better, we are conducting an open-label,<br />

randomized phase II trial comparing HX and LX. Methods: Primary endpoint<br />

is progression-free survival, and secondary endpoints are overall response<br />

rate, overall survival, proportion <strong>of</strong> patients progressing brain metastases as<br />

site <strong>of</strong> first progression, and safety. Major eligibility criteria include: (1)<br />

HER2-positive MBC, (2) previously treated with taxanes, (3) disease<br />

progression or distant relapse while receiving trastuzumab, (4) previously<br />

untreated with capecitabine, S-1, and anti-HER2 drugs other than trastuzumab,<br />

(5) previously treated with no more than two chemotherapy<br />

regimens for MBC, (6) no symptomatic brain metastases (asymptomatic<br />

brain metastases are allowed), and (7) baseline left ventricular ejection<br />

fraction �50%. Patients in the HX arm receive capecitabine 2,500<br />

mg/m2 /day on days 1 to 14 plus trastuzumab (8 mg/kg loading dose and<br />

6mg/kg thereafter) on day 1 every 3 weeks. Patients in the LX arm receive<br />

capecitabine 2,000 mg/m2 /day on days 1 to 14 plus lapatinib 1250<br />

mg/day on days 1 to 21 every 3weeks. Large-scale biomarker analyses are<br />

also performed to explore predictive factors <strong>of</strong> trastuzumab or lapatinib<br />

efficacy. We are investigating biomarkers related to HER family and other<br />

receptors, PI3K/Akt pathways, ligands, Fc�R, circulating tumor cells, and<br />

so on. This study has just begun and 7 <strong>of</strong> planned 170 patients have been<br />

enrolled.<br />

TPS661 General Poster Session (Board #16H), Sat, 8:00 AM-12:00 PM<br />

ALTERNATIVE (EGF114299): A study <strong>of</strong> lapatinib, trastuzumab, and<br />

endocrine therapy in patients who received neo-/adjuvant trastuzumab (IV)<br />

and endocrine therapy. Presenting Author: William Gradishar, Northwestern<br />

University, Chicago, IL<br />

Background: Approximately 50% <strong>of</strong> human epidermal growth factor receptor<br />

2 positive (HER2�) breast cancers are also hormone receptor-positive<br />

(HR�). For patients with HER2�/HR� disease, combining the aromatase<br />

inhibitor (AI) letrozole with the dual tyrosine kinase inhibitor lapatinib (L)<br />

has been shown to improve outcomes vs letrozole alone. Similar results<br />

were found for the combination <strong>of</strong> trastuzumab (T; a humanized monoclonal<br />

antibody targeting HER2) and anastrozole vs anastrozole alone. Finally,<br />

the combination <strong>of</strong> L and T has been shown to improve outcomes vs L alone.<br />

Methods: This phase III, randomized, open-label, multicenter trial (ALTER-<br />

NATIVE) will enroll postmenopausal female patients with HER2�/HR�<br />

metastatic breast cancer (MBC) who have not received prior treatment for<br />

MBC, have received neo/adjuvant T and endocrine therapy, and are not<br />

candidates for chemotherapy. Patients will be randomized 1:1:1 to 1 <strong>of</strong> 3<br />

treatment arms: L plus T plus an AI; T plus an AI; or L plus an AI. The AI can<br />

be letrozole, anastrozole, or exemestane, to be selected by the investigator.<br />

The primary objective <strong>of</strong> ALTERNATIVE is to examine the efficacy <strong>of</strong> L/T/AI<br />

compared with T/AI alone. The primary efficacy endpoint is overall survival<br />

(OS; time from randomization until death due to any cause) for L/T/AI vs<br />

T/AI. Secondary efficacy objectives include: comparisons <strong>of</strong> OS between<br />

T/AI and L/AI as well as between T/L/AI and L/AI, comparisons <strong>of</strong><br />

progression-free survival, overall response rate, time to response, duration<br />

<strong>of</strong> response, safety, and tolerability for all 3 treatment groups. The study is<br />

powered to detect a 42% reduction in risk <strong>of</strong> death (hazard ratio�0.70) in<br />

patients who receive L/T/AI (median 28.5 months) vs T/AI (median 20<br />

months) using a 1-sided test for superiority with ��0.025. The required<br />

number <strong>of</strong> total events to achieve a power <strong>of</strong> 80% is 249. Secondary<br />

comparisons are not powered and will be based on the ITT population. This<br />

study is currently recruiting, with a target <strong>of</strong> 525 patients. If its objectives<br />

are reached, it will provide a valuable, chemotherapy-free treatment option<br />

for HER2�/HR� MBC patients. <strong>Clinical</strong> trial registry number:<br />

NCT01160211.<br />

TPS660 General Poster Session (Board #16G), Sat, 8:00 AM-12:00 PM<br />

The PERSEPHONE trial: Duration <strong>of</strong> trastuzumab with chemotherapy in<br />

women with HER2-positive early breast cancer. Presenting Author: Helena<br />

Margaret Earl, Department <strong>of</strong> Oncology, University <strong>of</strong> Cambridge, and<br />

NIHR Cambridge Biomedical Research Centre, Cambridge, United Kingdom<br />

Background: Persephone is a phase III randomised controlled trial comparing<br />

six months <strong>of</strong> trastuzumab to the standard 12 month duration in<br />

patients with HER2 positive early breast cancer in respect <strong>of</strong> disease free<br />

survival, safety and cost-effectiveness. A Persephone sister study, the<br />

PHARE trial run by the National Institute for Cancer, successfully closed to<br />

recruitment in 2010. A prospective meta-analysis is planned once each<br />

trial has reported individually. Methods: A total <strong>of</strong> 4000 patients will be<br />

randomised into each <strong>of</strong> the two treatment groups. The power calculations<br />

assume that the disease-free survival (DFS) <strong>of</strong> the standard treatment <strong>of</strong> 12<br />

months trastuzumab will be 80% at 4 years. On this basis, with 5% 1-sided<br />

significance and 85% power, a trial randomising 2000 in each arm will<br />

have the ability to prove non-inferiority <strong>of</strong> the experimental arm defining<br />

non-inferiority as ‘no worse than 3%’ below the control arm 4 year DFS.<br />

Primary outcome is disease-free survival non-inferiority (equivalence) <strong>of</strong> 6<br />

months trastuzumab compared with 12 months in women with early breast<br />

cancer. Secondary outcomes are overall survival non-inferiority (equivalence);<br />

expected incremental cost effectiveness; cardiology function and<br />

analysis <strong>of</strong> predictive factors for development <strong>of</strong> cardiac damage. Two<br />

mandatory sub-studies are: Tumour block collection to discover molecular<br />

predictors <strong>of</strong> survival with respect to duration <strong>of</strong> trastuzumab treatment and<br />

blood sample collection, used to discover single nucleotide polymorphisms<br />

(SNPs) as genetic/pharmaco-genetic determinants <strong>of</strong> prognosis, toxicity<br />

and treatment outcome. A third optional sub-study is the quality <strong>of</strong> life<br />

questionnaires. Results: Persephone opened to recruitment in October<br />

2007. To date, 1847 patients (46%) <strong>of</strong> its total have been randomised<br />

from 147 UK sites. Recruitment is due to complete by December 2013 and<br />

the first planned interim analysis <strong>of</strong> the primary outcome will be mid-2016.<br />

The IDSMC last reviewed the trial in June 2011 and congratulated the Trial<br />

Management Group on the conduct <strong>of</strong> the trial and the quality <strong>of</strong> the data.<br />

No safety concerns were identified, and the IDSMC proposed that the trial<br />

continue to planned recruitment.<br />

TPS662 General Poster Session (Board #17A), Sat, 8:00 AM-12:00 PM<br />

HIT: A multicenter phase I-II study <strong>of</strong> trastuzumab administered by<br />

intrathecal injection for leptomeningeal meningitis <strong>of</strong> HER2� metastatic<br />

breast cancer. Presenting Author: Maya Gutierrez, Hôpital René Huguenin/<br />

Institut Curie, Saint-Cloud, France<br />

Background: Leptomeningeal metastases (LM) are commonly associated<br />

with breast cancer (BC), announcing short term prognosis that intrathecal<br />

(IT) chemotherapy poorly modifies. Incidence is particularly marked in<br />

HER2� tumours. With the better control <strong>of</strong> extra-cerebral disease and<br />

prolonged survival yielded by intra-venous (IV) trastuzumab (T), the main<br />

hypothesis is that intra-cerebral recurrences are not reached by this high<br />

molecular weight (148 kD) monoclonal antibody. Analyses performed in<br />

cerebrospinal fluid (CSF) following IV T have shown low T levels, suggesting<br />

that LM <strong>of</strong> HER2� BC would remain potentially sensitive to anti-HER2<br />

agents as long as they could by-pass the meningeal blood brain barrier.<br />

Intraventricular (via Ommaya port) or IT administration <strong>of</strong> T would allow<br />

keeping control on LM progression through high T therapeutic concentrations<br />

in CSF. This prospective trial is the first phase I-II study sponsored by<br />

Institut Curie to investigate the safety and efficacy <strong>of</strong> the IT administration<br />

<strong>of</strong> T for HER2� LM BC. Methods: Eligible patients must be 18�, have a<br />

diagnosis <strong>of</strong> LM either on CSF with HER2� cytology or on clinical/MRI<br />

pictures <strong>of</strong> meningitis, and normal end-organ functions. The phase I cohort<br />

investigates the maximum tolerated dose (MTD) and recommended dose <strong>of</strong><br />

T given weekly IT (or via Ommaya port) � hemisuccinate hydrocortisone 25<br />

mg, while aiming at yielding a T target-concentration in CSF close to the<br />

plasma one (30 �g/mL) obtained with standard IV schedule. The MTD is<br />

defined as the highest dose level (DL) tested with �2 dose-limiting toxicity<br />

(DLT, any grade �3 NCI CTCAE v4.0 attributed to T) observed in �6<br />

patients (3�3 Fibonacci dose escalation design, 4 DL 30-150 mg) with a<br />

maximum <strong>of</strong> 24 patients. The phase II cohort will investigate the efficacy <strong>of</strong><br />

the recommended dose (DLMTD-1) defined in cohort I on neurological<br />

progression-free survival at 2 months, in �19 patients. Secondary endpoints<br />

include CSF and MRI response, quality <strong>of</strong> life, FCGR3A influence,<br />

and comparative CSF/plasma pharmacokinetics. Since study activation in<br />

May 2011, 5 patients have been included at DL1, 3 being evaluable for<br />

DLT. DL2 is now open for accrual.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


TPS663 General Poster Session (Board #17B), Sat, 8:00 AM-12:00 PM<br />

A phase I study <strong>of</strong> MM-302, a HER2-targeted liposomal doxorubicin, in<br />

patients with advanced, HER2-positive (HER2�) breast cancer. Presenting<br />

Author: Pamela N. Munster, University <strong>of</strong> California, San Francisco Helen<br />

Diller Family Comprehensive Cancer Center, San Francisco, CA<br />

Background: Anthracyclines have been an effective backbone <strong>of</strong> breast<br />

cancer therapies for decades. However, cardiotoxicity issues associated<br />

with free anthracyclines have limited their effective use in the clinic and<br />

led to the exploration <strong>of</strong> anthracycline-free regimens, particularly with<br />

HER2-positive cancers that require treatment with another cardiotoxic<br />

agent, trastuzumab. While liposomal doxorubicin formulations have succeeded<br />

in reducing cardiotoxicity, they have failed to demonstrate clear-cut<br />

efficacy advantages and can involve other toxicities. To address the safety<br />

and efficacy limitations <strong>of</strong> currently available anthracyclines, we have<br />

designed a new liposomal formulation, MM-302, that targets doxorubicin<br />

to HER2-overexpressing tumor cells. Antibody fragments that bind to<br />

HER2 without blocking HER2-mediated signaling are coupled to the outer<br />

surface <strong>of</strong> pegylated liposomal doxorubicin. MM-302 specifically binds and<br />

enters tumor cells overexpressing HER2 with minimal uptake into normal<br />

cells such as cardiomyocytes which express low levels <strong>of</strong> HER2. This<br />

first-in-human phase I study evaluates the safety <strong>of</strong> MM-302 in patients<br />

and provides preliminary efficacy data in HER-2� advanced breast cancer<br />

(ABC). Methods: Patients aged � 18 years with histologically confirmed<br />

HER-2� advanced breast cancer that have progressed or recurred on<br />

standard therapy or for which no standard therapy exists who have adequate<br />

performance status, bone marrow reserve, and organ function, are eligible<br />

for the study. Following a standard 3 � 3 dose escalation design, the<br />

maximum tolerated dose (MTD) or maximum feasible dose (MFD) is<br />

determined and up to 25 additional patients with HER-2� ABC will be<br />

enrolled for a planned total <strong>of</strong> 40-49 patients. The primary endpoint is<br />

determination <strong>of</strong> the MTD/MFD. Secondary endpoints include determination<br />

<strong>of</strong> dose-limiting toxicity, adverse event(s), and pharmacokinetic and<br />

immunogenicity pr<strong>of</strong>iles <strong>of</strong> MM-302, as well as overall response and<br />

clinical benefit rates <strong>of</strong> MM-302. MM-302 is administered intravenously<br />

weekly in 4-week cycles. At the time <strong>of</strong> this submission, 8 patients have<br />

been enrolled in the dose escalation portion.<br />

TPS665 General Poster Session (Board #17D), Sat, 8:00 AM-12:00 PM<br />

OPTIMA prelim: Optimal personalized treatment <strong>of</strong> early breast cancer<br />

using multiparameter analysis: Preliminary study. Presenting Author: Rob<br />

Stein, University College London Hospitals, London, United Kingdom<br />

Background: “Multi-parameter” prognostic tests such as Oncotype DX are<br />

increasingly used to identify women with ER �ve HER2 -ve breast cancer<br />

treated with endocrine therapy who are unlikely to benefit meaningfully<br />

from adjuvant chemotherapy. The supporting evidence for predictive<br />

testing is retrospective and is strongest for women with negative nodes.<br />

Randomised trials to validate testing are in progress but more evidence is<br />

needed, especially for node positive disease. There is early evidence that<br />

other multi-parameter tests which are in development have a predictive<br />

utility. Methods: OPTIMA will assess the value <strong>of</strong> multi-parameter tests in a<br />

UK population. OPTIMA prelim, the feasibility phase, will recruit 300<br />

patients from May 2012 (with a 200 patient bridging extension to the main<br />

study). Eligible patients will have ER �ve HER2 -ve tumours with involved<br />

nodes (pN1-2). Patients will be randomised to the standard arm <strong>of</strong> both<br />

chemotherapy and endocrine therapy, or to the “test-directed treatment”<br />

arm in which patients will be assigned either the same chemotherapy and<br />

endocrine therapy or endocrine therapy only according to the result <strong>of</strong> an<br />

Oncotype DX test. OPTIMA prelim has 3 objectives. (1) To establish<br />

whether randomisation to test-directed treatment is acceptable to patients<br />

and clinicians. This will be done through qualitative research with in-depth<br />

interviews with OPTIMA researchers and by recording and analysing<br />

consultations when the trial is discussed with potential participants. (2)<br />

Alternative multi-parameter tests will be evaluated on all patients’ tumours<br />

and their performance will be compared to Oncotype DX using statistical<br />

and cost-effectiveness analysis to select a candidate test(s) appropriate for<br />

NHS use to be evaluated in the main trial. (3) The success <strong>of</strong> the main trial<br />

depends on efficient tumour sample collection and analysis to avoid<br />

treatment delays. The obstacles to this will be analysed in detail using a<br />

sample tracking database. The main study is an efficacy trial <strong>of</strong> 2-3 arms<br />

with the same design but uses tests selected in OPTIMA prelim. It will<br />

compare 5-year relapse free survival <strong>of</strong> test-directed vs. conventional<br />

treatment with a non-inferiority hypothesis.<br />

Breast Cancer—HER2/ER<br />

TPS664 General Poster Session (Board #17C), Sat, 8:00 AM-12:00 PM<br />

A phase I study <strong>of</strong> BKM120, a novel oral selective phosphatidylinositol-3-kinase<br />

(PI3K) inhibitor, in combination with fulvestrant in postmenopausal women with<br />

estrogen receptor positive metastatic breast cancer. Presenting Author: Gayathri<br />

Nagaraj, Washington University in St. Louis, St. Louis, MO<br />

Background: PI3K pathway activation plays a crucial role in mediating endocrine<br />

therapy resistance in estrogen receptor positive (ER�) breast cancer. We have<br />

shown previously that BKM120 in combination with fulvestrant induced synergistic<br />

apoptotic cell death in long-term estrogen deprived ER� breast cancer<br />

(LTED) cell line models, supporting the clinical investigation <strong>of</strong> this combination<br />

in ER� breast cancer. Methods: The study is composed <strong>of</strong> the dose escalation<br />

cohort (phase IA) and the expansion cohort (phase IB). In phase IA, a standard<br />

3�3 phase I design is employed to determine the maximum tolerated dose<br />

(MTD) <strong>of</strong> the combination <strong>of</strong> BKM120 and fulvestrant in patients (pts) with ER�<br />

metastatic breast cancer (MBC). In phase IB, an additional 10 pts will be<br />

enrolled at the MTD to further examine the toxicity pr<strong>of</strong>ile and preliminary<br />

efficacy <strong>of</strong> this combination. Steady state concentrations <strong>of</strong> BKM120 will be<br />

analyzed. Postmenopausal women with ER� MBC and measurable disease per<br />

RECIST are eligible. Pts who are currently taking fulvestrant without disease<br />

progression are eligible. There is no restriction on the number <strong>of</strong> prior lines <strong>of</strong><br />

systemic therapy for metastatic disease in phase IA but � 3 lines is required in<br />

phase IB. Treatment consists <strong>of</strong> fulvestrant 500 mg IM administered monthly on<br />

day (d) 1 <strong>of</strong> each 28-d cycle, following the loading dose <strong>of</strong> 500 mg on d1 and<br />

d15, and BKM120 orally daily on d1-28 <strong>of</strong> each cycle. The starting dose level<br />

(DL) is DL1 (Table). Correlative studies include assessing PI3K pathway<br />

abnormalities (loss <strong>of</strong> PTEN and PIK3CA mutation) on archival tumor specimen,<br />

and treatment induced inhibition <strong>of</strong> PI3K pathway activity (pAKT, pS6, Cyclin<br />

D1, subcellular localization <strong>of</strong> FOXO3a, phosphoproteomics), tumor cell proliferation<br />

(Ki67) and apoptosis (cleaved caspase 3 or TUNEL staining) on tumor<br />

biopsies collected at baseline and cycle 2 day 1 in consented patients.<br />

Enrollment to DL1 is complete and the study is currently enrolling pts to DL2.<br />

Dose levels for the dose escalation cohort (phase IA).<br />

DL<br />

BKM120 (mg)<br />

PO daily<br />

Dose<br />

Fulvestrant (mg)<br />

Day 1 <strong>of</strong> each cycle<br />

2 100 500<br />

1 80 500<br />

-1 60 500<br />

47s<br />

TPS666 General Poster Session (Board #17E), Sat, 8:00 AM-12:00 PM<br />

Randomized phase II open-label study <strong>of</strong> abiraterone acetate (AA) plus<br />

low-dose prednisone (P) with or without exemestane (E) in postmenopausal<br />

women with ER� metastatic breast cancer (MBC) progressing after<br />

letrozole or anastrozole therapy. Presenting Author: Joyce O’Shaughnessy,<br />

Texas Oncology-Baylor Charles A. Sammons Cancer Center and US Oncology,<br />

Dallas, TX<br />

Background: AA plus P treatment in men with metastatic castrationresistant<br />

prostate cancer has demonstrated a survival advantage over P<br />

alone. Circulating adrenal steroids including DHEA and DHEA-sulfate can<br />

stimulate proliferation <strong>of</strong> breast cancer cell lines in a low-estrogen<br />

environment. Thus, it is hypothesized that depletion <strong>of</strong> adrenal androgens<br />

as well as estrogens by AA, a potent CYP17 inhibitor, inhibits tumor growth<br />

by disruption <strong>of</strong> ER-dependent growth signaling. In a previously reported<br />

phase I study <strong>of</strong> AA in patients (pts) with breast cancer (Basu, ASCO 2011),<br />

2/21 pts who were ER� were on study for � 11 months; 1 had a confirmed<br />

PR and continued on treatment � 14 months. Mechanism-based adverse<br />

event <strong>of</strong> grade 3/4 hypokalemia occurred in 4 pts and was managed with<br />

potassium supplementation and low dose corticosteroids. Methods: In the<br />

current study, 300 postmenopausal women with ER� Her2- MBC progressing<br />

after letrozole or anastrozole are randomized to either AA 1000 mg � P<br />

5 mg daily or AA � P � E 25 mg daily vs E. Disease must have been<br />

sensitive to a non-steroidal aromatase inhibitor (AI) prior to study entry.<br />

Subjects are stratified by number <strong>of</strong> prior therapies and by AI use in<br />

adjuvant or metastatic setting. Central review <strong>of</strong> tissue is required prior to<br />

randomization. Gene expression pr<strong>of</strong>iling (GEP) (AR, ER, PR, Her2,<br />

CYP17, Ki-67, CYP 19, and 3-�-HSD) will be performed on FFPE archival<br />

tissue. Pre- and post-treatment circulating tumor cells and fresh tumor<br />

biopsies will also be obtained for GEP in a subset <strong>of</strong> pts. Primary endpoint is<br />

PFS. Secondary endpoints are OS, ORR, patient reported outcomes,<br />

changes in endocrine markers, and PK characterization <strong>of</strong> AA and E.<br />

Subjects randomized to E may crossover to AA � P at time <strong>of</strong> disease<br />

progression. One interim analysis is scheduled after 50% <strong>of</strong> PFS events<br />

have occurred. After review <strong>of</strong> interim data, the Data Review Committee will<br />

make recommendations regarding study continuation. 23pts from 45<br />

active sites have been randomized as <strong>of</strong> January 24,2012. (<strong>Clinical</strong>Trials.<br />

gov Identifier: NCT01381874)<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


48s Breast Cancer—HER2/ER<br />

TPS667 General Poster Session (Board #18A), Sat, 8:00 AM-12:00 PM<br />

ASTER 70s: Benefit <strong>of</strong> adjuvant chemotherapy for estrogen receptorpositive<br />

HER2-negative breast cancer in women over 70 according to<br />

genomic grade—A French GERICO/UCBG UNICANCER multicenter phase<br />

III trial. Presenting Author: Etienne Brain, Hôpital René Huguenin/Institut<br />

Curie, Saint-Cloud, France<br />

Background: The benefit <strong>of</strong> adjuvant chemotherapy (CT) is highly controversial<br />

for elderly breast cancer (BC) women presenting with an oestrogen<br />

receptor-positive (ER�) HER2-negative (HER2-) phenotype. Conversely to<br />

hormonal treatment (HT) that remains the cornerstone <strong>of</strong> adjuvant treatment<br />

for such luminal tumours, CT may severely decompensate comorbidities<br />

and alter quality <strong>of</strong> life in elderly patients. As disappointing as it is in<br />

drug development, elderly have been constantly excluded from trials<br />

evaluating new modern prognosis classifiers. This prospective multicentre<br />

trial funded by a French national grant (PHRC 2011) is the first phase III<br />

trial to investigate the impact on overall survival (OS) <strong>of</strong> adjuvant CT in<br />

elderly ER� HER2- BC patients selected with a modern prognosis classifier<br />

and taking into account competing risks for mortality (EudraCT 2011-<br />

004744-22). Methods: Following surgery, 2,000 women 70� with ER�<br />

HER2- BC (any pT/pN), will have a genomic grade (GG, derived from frozen<br />

MapQuantDx, Ipsogen) centrally assessed on formalin-fixed paraffinembedded<br />

samples. Only those with a high GG (estimation~700) will be<br />

randomized between HT alone vs CT followed by HT. CT regimen is left to<br />

the choice <strong>of</strong> investigators amongst 3 regimen <strong>of</strong> same duration [4 q3w<br />

cycles, docetaxel�cyclophosphamide, doxorubicin or non pegylated liposomal<br />

doxorubicin (Myocet)�cyclophosphamide, all with G-CSF], as well<br />

as HT (aromatase inhibitor�tamoxifen). Those with low GG or not included<br />

for other reasons (estimation~1,300) will be followed as an observational<br />

parallel cohort with HT alone. Sample size is based on 4-year OS as primary<br />

endpoint (87.5 vs 80%), bilateral ��0.05, ��0.20 and HR� 0.60.<br />

Secondary endpoints include assessment <strong>of</strong> competing risks for mortality,<br />

cost-effectiveness and Q-TWiST analysis, geriatric items (e.g. Lee’s 4-year<br />

mortality score and G8 screening tool), acceptability, quality <strong>of</strong> life<br />

(QLQ-C30 and specific elderly scale ELD15), and translational research on<br />

ageing/prognostic biomarkers and pharmacogenetic. The trial has been just<br />

opened to inclusion in February 2012.<br />

TPS669 General Poster Session (Board #18C), Sat, 8:00 AM-12:00 PM<br />

Aromatase inhibitors, arthralgias, and exercise in breast cancer survivors.<br />

Presenting Author: Melinda Irwin, Yale University, New Haven, CT<br />

Background: A substantial number <strong>of</strong> breast cancer survivors who are taking<br />

aromatase inhibitors (AIs) complain <strong>of</strong> side effects including arthralgias.<br />

Given the efficacy <strong>of</strong> AIs, it is essential that we identify approaches to<br />

mitigate arthralgias. We are conducting an NCI-funded trial is to examine<br />

the impact <strong>of</strong> a year-long exercise intervention in 180 breast cancer<br />

survivors taking an AI and experiencing at least mild arthralgias. Outcomes<br />

include severity <strong>of</strong> arthralgias, endocrine-related quality <strong>of</strong> life, % body fat,<br />

bone mass, and serum markers <strong>of</strong> inflammation. The purpose <strong>of</strong> this<br />

abstract is to present preliminary recruitment and adherence results.<br />

Methods: Women are being recruited via the Connecticut Tumor Registry<br />

and Rapid Case Ascertainment Shared Resource <strong>of</strong> Yale Cancer Center into<br />

the Hormones and Physical Exercise (HOPE) Study. <strong>Part</strong>icipants are<br />

randomly assigned to exercise (n � 90) or usual care (n � 90). The exercise<br />

intervention includes supervised resistance and unsupervised aerobic<br />

exercise sessions over 12 months. Data are collected at baseline, 3-, 6-, 9<br />

and 12-months. Results: Study recruitment began in April 2010 and will<br />

continue through December 2012. As <strong>of</strong> January 1, 2012, we have<br />

received 1605 names <strong>of</strong> potentially eligible women. Of these 1605, 777<br />

women are waiting to be screened (e.g., letter mailed to patient or on hold<br />

because recently diagnosed), 244 women could not be screened (e.g.,<br />

unable to contact by phone), and 584 women have completed a telephone<br />

screening. Of the 584 women screened, 24% were ineligible because <strong>of</strong><br />

discontinuing AIs because <strong>of</strong> arthralgias or chose not to take AIs primarily<br />

because <strong>of</strong> this potential side effect; another 36% were ineligible for<br />

various reasons; 24% were not interested; 11% were randomized (n � 65<br />

women); and 5% are undergoing baseline visits (n � 29). On average,<br />

women are 62 yrs old and have been taking an AI for 1.8 yrs. Twenty-three<br />

women have completed the trial, with high exercise adherence rates<br />

(attendance to supervised sessions � 82% ). Conclusions: A growing<br />

number <strong>of</strong> women are choosing to not take AIs or are discontinuing AIs<br />

because <strong>of</strong> side effects. Our study may prove beneficial for the growing<br />

number <strong>of</strong> women diagnosed with hormone receptor-positive breast cancer<br />

each year.<br />

TPS668 General Poster Session (Board #18B), Sat, 8:00 AM-12:00 PM<br />

MDV3100-08: A phase I open-label, dose-escalation study evaluating the<br />

safety, tolerability, and pharmacokinetics <strong>of</strong> MDV3100 in women with<br />

incurable breast cancer. Presenting Author: Anthony D. Elias, University <strong>of</strong><br />

Colorado Anschutz Medical Campus, Aurora, CO<br />

Background: MDV3100 is a novel, orally administered small molecule that<br />

directly binds the androgen receptor (AR) to potently inhibit AR-mediated<br />

signaling via three mechanisms: inhibition <strong>of</strong> androgen binding to AR;<br />

inhibition <strong>of</strong> AR nuclear translocation; and inhibition <strong>of</strong> nuclear AR<br />

association with DNA. MDV3100 is not known to agonize AR signaling, can<br />

be taken without prednisone, and demonstrated an overall survival benefit<br />

in post-docetaxel prostate cancer at 160 mg/day. AR expression is observed<br />

in ~70% <strong>of</strong> all breast cancer (BCa) regardless <strong>of</strong> histologic subtypes<br />

(estrogen receptor [ER] positive, HER2�, and triple negative [TN] disease).<br />

MDV3100 has demonstrated preclinical activity in AR�/TN and AR�/<br />

ER� BCa and could be a potential therapeutic strategy in patients with<br />

AR� BCa. Methods: This Phase 1 study has 2 stages: a standard 3�3<br />

dose-escalation stage (S1), evaluating safety, tolerability, and pharmacokinetics<br />

(PK) <strong>of</strong> MDV3100, starting at 80 mg MDV3100; and a doseexpansion<br />

stage (S2), evaluating the recommended Phase 2 dose in ~15<br />

women with AR� BCa. All patients must have received �2 chemotherapycontaining<br />

regimens for their incurable BCa, or �3 anti-HER2 containing<br />

regimens for their incurable HER2� BCa to be eligible; ER� patients must<br />

be post-menopausal. In S1, patients will receive their assigned dose <strong>of</strong><br />

MDV3100 on Day 1, and PK samples will be collected through Day 8. Daily<br />

dosing will begin on Day 8 and will continue until predefined discontinuation<br />

criterion is met. Dose-limiting toxicities will be recorded through Day<br />

35 and used to determine the Phase 2 dose. Extensive PK sampling is<br />

performed on Days 1 and 50 with predose PK samples collected throughout.<br />

In S2, patients with AR� BCa (defined as 10% or more tumor cells<br />

with nuclear AR expression) will take daily MDV3100 at the recommended<br />

Phase 2 dose; there is no single dose PK period. Tumor tissue submission is<br />

mandatory in S2. Safety and tolerability will be documented by frequent<br />

assessments <strong>of</strong> adverse events, vital signs, and laboratory assessments.<br />

Tumor assessments occur at 2 months then approximately every 3 months<br />

thereafter. Enrollment <strong>of</strong> 27 subjects is anticipated.<br />

TPS670 General Poster Session (Board #18D), Sat, 8:00 AM-12:00 PM<br />

Denosumab versus placebo as adjuvant treatment for women with earlystage<br />

breast cancer who are at high risk <strong>of</strong> disease recurrence (D-CARE): An<br />

international, randomized, double-blind, placebo-controlled phase III clinical<br />

trial. Presenting Author: Paul Edward Goss, Massachusetts General<br />

Hospital Cancer Center, Boston, MA<br />

Background: Bone is a common site <strong>of</strong> distant recurrence in women with<br />

early-stage breast cancer, and represents approximately 40% <strong>of</strong> all first<br />

recurrences. Tumor cells in bone release growth factors and cytokines that<br />

stimulate osteoclast-mediated bone resorption through the RANK ligand<br />

(RANKL) pathway. In preclinical studies, RANKL inhibition significantly<br />

delays skeletal tumor formation, reduces skeletal tumor burden, and<br />

prolongs survival <strong>of</strong> tumor-bearing mice. Denosumab is a fully human<br />

monoclonal antibody that binds to RANKL with high affinity and specificity.<br />

It is approved for the prevention <strong>of</strong> skeletal-related events in patients with<br />

established bone metastases from a variety <strong>of</strong> solid tumors. The purpose <strong>of</strong><br />

the D-CARE trial is to evaluate the ability <strong>of</strong> denosumab to prolong bone<br />

metastasis-free survival (BMFS) and disease-free survival (DFS) in the<br />

adjuvant breast cancer setting. Methods: Approximately 4,500 women with<br />

stage II or III breast cancer, at high risk for recurrence and with known<br />

hormone and HER-2 receptor status, are eligible. Standard-<strong>of</strong>-care adjuvant<br />

or neoadjuvant chemo-, endocrine, or HER-2 targeted therapy, alone<br />

or in combination must be planned. Exclusion criteria include: a prior<br />

history <strong>of</strong> breast cancer (except DCIS or LCIS) or distant metastasis, oral<br />

bisphosphonate (BP) use within 1 year <strong>of</strong> randomization or any intravenous<br />

BP use. Patients are randomized 1:1 to receive denosumab 120 mg or<br />

placebo subcutaneously monthly for 6 months, then every 3 months for a<br />

total <strong>of</strong> 5 years <strong>of</strong> treatment. All patients receive vitamin D (� 400 IU) and<br />

calcium (� 500 mg) supplements. Primary endpoint <strong>of</strong> this event-driven<br />

trial is BMFS. Secondary endpoints include DFS and overall survival.<br />

Safety, quality <strong>of</strong> life assessments, breast density, and biomarkers are<br />

additional endpoints. The trial, sponsored by Amgen Inc. and registered<br />

with the <strong>Clinical</strong>Trials.gov identifier NCT01077154, began enrolling patients<br />

in June 2010 and is expected to complete in October 2016. Paul<br />

Goss and Dianne Finkelstein supported in part by the Avon Foundation New<br />

York.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


LBA1000 Oral Abstract Session, Tue, 9:45 AM-12:45 PM<br />

NSABP B-38: Definitive analysis <strong>of</strong> a randomized adjuvant trial comparing<br />

dose-dense (DD) AC followed by paclitaxel (P) plus gemcitabine (G) with<br />

DD AC followed by P and with docetaxel, doxorubicin, and cyclophosphamide<br />

(TAC) in women with operable, node-positive breast cancer. Presenting<br />

Author: Sandra M. Swain, National Surgical Adjuvant Breast and Bowel<br />

Project and Washington Cancer Institute, MedStar Washington Hospital<br />

Center, Washington, DC<br />

The full, final text <strong>of</strong> this abstract will be available at<br />

abstract.asco.org at 12:01 AM (EDT) on Monday, June 4,<br />

2012, and in the <strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong> online<br />

supplement to the June 20, 2012, issue <strong>of</strong> Journal <strong>of</strong><br />

<strong>Clinical</strong> Oncology. Onsite at the <strong>Meeting</strong>, this abstract will<br />

be printed in the Monday edition <strong>of</strong> ASCO Daily News.<br />

CRA1002 Oral Abstract Session, Tue, 9:45 AM-12:45 PM<br />

CALGB 40502/NCCTG N063H: Randomized phase III trial <strong>of</strong> weekly<br />

paclitaxel (P) compared to weekly nanoparticle albumin bound nabpaclitaxel<br />

(NP) or ixabepilone (Ix) with or without bevacizumab (B) as<br />

first-line therapy for locally recurrent or metastatic breast cancer (MBC).<br />

Presenting Author: Hope S. Rugo, University <strong>of</strong> California, San Francisco<br />

Helen Diller Family Comprehensive Cancer Center, San Francisco, CA<br />

The full, final text <strong>of</strong> this abstract will be available at<br />

abstract.asco.org at 12:01 AM (EDT) on Monday, June 4,<br />

2012, and in the <strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong> online<br />

supplement to the June 20, 2012, issue <strong>of</strong> Journal <strong>of</strong><br />

<strong>Clinical</strong> Oncology. Onsite at the <strong>Meeting</strong>, this abstract will<br />

be printed in the Monday edition <strong>of</strong> ASCO Daily News.<br />

Breast Cancer—Triple-Negative/Cytotoxics/Local Therapy<br />

49s<br />

1001 Oral Abstract Session, Tue, 9:45 AM-12:45 PM<br />

Randomized phase III study <strong>of</strong> adjuvant chemotherapy for high-risk,<br />

node-negative breast cancer (BC) comparing FAC with FAC followed by<br />

weekly paclitaxel: First efficacy analysis <strong>of</strong> the GEICAM/2003-02 trial.<br />

Presenting Author: Miguel Martin, Hospital General Universitario Gregorio<br />

Marañón, Madrid, Spain<br />

Background: Adjuvant weekly paclitaxel (wP) sequential to anthracyclines<br />

improves the outcome <strong>of</strong> operable node-positive BC patients (pts) [Sparano<br />

NEJM 2008, Martin BCRT 2009]; however, most BC pts are currently<br />

node-negative at diagnosis. The role <strong>of</strong> wP in these pts is not well<br />

established yet. Methods: Pts aged 18-70, with T1-T3/N0 operable BC and<br />

at least one high-risk St Gallen 1998 criteria (size �2 cm, hormonereceptor<br />

[HR] negative, grade 2/3, age �35 years,) were eligible. HER2�<br />

pts were allowed, after 792 entered the trial, the study was amended to<br />

exclude them. Pts were stratified by site, menopausal status, nodal status<br />

diagnostic method (sentinel-node biopsy versus lymphadenectomy) and<br />

HR status and randomized to receive FAC x6 (500/50/500 mg/m2 every<br />

3w) or FAC x4¡wP x8 (paclitaxel 100 mg/m2 weekly). The primary<br />

endpoint was DFS. The trial was designed to detect an absolute 5-y DFS<br />

increase <strong>of</strong> 5% (80% FAC, 85% FAC¡wP); a sample size <strong>of</strong> 1812<br />

evaluable patients (906 per arm) was required to detect this difference<br />

(��0.05, �� 80%). Assuming a drop-out rate <strong>of</strong> 6%, 1929 pts were<br />

required. The first analysis <strong>of</strong> DFS was planned when a median follow-up <strong>of</strong><br />

5 years was reached. Results: Between September 2003 and October<br />

2008, 1925 pts (FAC 974, FAC¡wP 951) were randomized. Patient<br />

characteristics were well balanced between arms, median age was 50, 73%<br />

<strong>of</strong> pts were HR positive and 9% HER2 positive. 97% <strong>of</strong> pts with FAC and<br />

85% <strong>of</strong> pts with FAC¡wP completed all treatment as planned. The median<br />

dose intensity was 98% with FACx6, 99% with FACx4 and 98% with wP.<br />

The most frequent grade 3-4 toxicities (�3% in either arm) with FAC vs<br />

FAC¡wP were neutropenia (25% vs 22%) with 4% vs 3% <strong>of</strong> febrile<br />

neutropenia, fatigue (3% vs 8%), sensory neuropathy (0 vs 5%), and<br />

vomiting (4% in each arm). After a median follow-up <strong>of</strong> 5.3 years, the<br />

proportion <strong>of</strong> patients disease free is 93% and 90% with FAC¡wP and FAC<br />

(HR for relapse 0.732, 95% CI: 0.542 to 0.990; log-rank p-value�0.0423).<br />

Conclusions: For pts with high-risk node-negative BC, adjuvant FAC¡wP<br />

was associated with a small but significant improvement in DFS compared<br />

with FAC, with manageable toxicity.<br />

1003 Oral Abstract Session, Tue, 9:45 AM-12:45 PM<br />

A phase III, multicenter, randomized trial <strong>of</strong> maintenance versus observation<br />

after achieving clinical response in patients with metastatic breast<br />

cancer who received six cycles <strong>of</strong> gemcitabine plus paclitaxel as first-line<br />

chemotherapy (KCSG-BR 0702, NCT00561119). Presenting Author:<br />

Young-Hyuck Im, Samsung Medical Center, Seoul, South Korea<br />

Background: Chemotherapy provides a survival benefit in patients with<br />

metastatic breast cancer (MBC), but the optimal duration <strong>of</strong> chemotherapy<br />

remains controversial. Primary purpose <strong>of</strong> the study was to evaluate<br />

whether the maintenance chemotherapy with gemcitabine/paclitaxel (GP),<br />

which is one <strong>of</strong> the two regimens which showed a survival gain from a<br />

randomized trial, is superior to observation in terms <strong>of</strong> progression free<br />

survival (PFS) in responding patients with MBC after 6 cycles <strong>of</strong> GP as<br />

first-line treatment. Methods: This study is a prospective, randomized,<br />

multi-center, phase III study. Patients who achieved response (CR�PR�SD)<br />

following 6 cycles <strong>of</strong> GP chemotherapy (gemcitabine 1250 mg/m2 on day 1<br />

and 8 plus paclitaxel 175 mg/m2 on day 1 every 3 weeks) randomized to<br />

maintenance till progression or observation arm. The trial was conducted by<br />

the Korean Cancer Study Group (KCSG). Results: Among total 324 patients<br />

enrolled between 2007 and 2010 from 10 centers, 231 responding<br />

patients to were randomly assigned to maintenance chemotherapy (n�116)<br />

or observation (n�115). Median age was 49 (range 28-76). The numbers<br />

<strong>of</strong> hormone receptor (HR)�ve and HR-ve patients were 172 (74.5%) and<br />

59 (25.5%), respectively. The median No. <strong>of</strong> chemotherapy cycles in<br />

maintenance group was 12 (range 6-32). During median 33 months <strong>of</strong><br />

follow-up, median PFS was superior in maintenance than in observation<br />

(12.0 vs. 8.3 months, p�0.030). Patients � age 50 years (hazard ratio<br />

0.50, p�0.001) and HR-ve patients (hazard ratio 0.52, p�0.019)<br />

received more benefit from maintenance chemotherapy in terms <strong>of</strong> PFS.<br />

Median OS was superior in maintenance than in observation (36.8 vs 28.0<br />

months, p�0.047). Neurotoxicity (� grade 2) was more common in<br />

maintenance than in observation without statistical significance (41.7% vs<br />

33.3%, p�0.210). Serial assessment <strong>of</strong> Quality <strong>of</strong> Life (QoL) did not show<br />

any significant difference between two groups. Conclusions: Maintenance<br />

GP chemotherapy for responding patients with MBC showed clinical benefit<br />

in terms <strong>of</strong> PFS and OS without impairment <strong>of</strong> QoL.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


50s Breast Cancer—Triple-Negative/Cytotoxics/Local Therapy<br />

1004 Oral Abstract Session, Tue, 9:45 AM-12:45 PM<br />

RTOG 9804: A prospective randomized trial for “good risk” ductal<br />

carcinoma in situ (DCIS), comparing radiation (RT) to observation (OBS)<br />

Presenting Author: Beryl McCormick, Memorial Sloan-Kettering Cancer<br />

Center, New York, NY<br />

Background: Whole breast RT following conservation surgery (BCS) for low<br />

risk DCIS has remained controversial despite several large trials comparing<br />

RT to OBS, all showing significant benefit in local control with RT. RTOG<br />

9804 compares RT to OBS for mammographically detected disease, <strong>of</strong> low<br />

or intermediate nuclear grade, �2.5 cm size, and surgical margins � 3<br />

mm. Tamoxifen (TAM) use for 5 years was allowed but not required.<br />

Methods: The primary endpoint was ipsilateral breast local failure (LF). LF<br />

and contralateral breast failures (CBF) were estimated by the cumulative<br />

incidence method and treatment arms compared by log-rank test. Diseasefree<br />

(DFS) and overall survival (OS) were estimated by the Kaplan-Meier<br />

method and treatment arms compared by log-rank test. Patients were<br />

stratified by age, margin width, grade, TAM use, and primary size. With<br />

1790 patients, 80% power and using a 2-sided log rank test at 0.05, the<br />

study was designed to detect a reduction in 5-year local recurrence from<br />

6% to 3.5% with RT. Results: Accrual goals for the planned 1790 patients<br />

were not met; the study was closed early. From December 1999 to July<br />

2006, 636 women were randomized to receive 50 Gy in 5 weeks vs. OBS.<br />

43 women were ineligible on review and 8 withdrew consent. Median<br />

follow-up (F/U) time was 6.46 years. Mean age was 59; TAM was used in<br />

62% <strong>of</strong> women. There were 2 LF in the RT arm vs. 15 in the OBS arm: at 5<br />

years 0.4% RT vs. 3.2% OBS (p�0.0023, HR [95%CI] � 0.14 [0.03,<br />

0.61]). With limited events, LF is not correlated with size, grade, margin<br />

status, or age. The rate <strong>of</strong> CBF at 5 years was 3.0% for the RT arm vs. 1.9%<br />

for the OBS arm (p�0.42, HR [95%CI] � 1.46 [0.59, 3.62]) and does not<br />

appear to be influenced by TAM use (3.6 versus 2.7% TAM). The DFS and<br />

OS results were excellent. Rate <strong>of</strong> grade 1-2 toxicity was 76% in the RT arm<br />

vs. 30% in the OBS arm, and the rate <strong>of</strong> � 3 grade toxicities was 4% on<br />

both arms. Conclusions: In this “good risk” subset <strong>of</strong> DCIS, the LF rate was<br />

decreased significantly with the addition <strong>of</strong> RT. Longer follow-up is<br />

planned.<br />

1006 Oral Abstract Session, Tue, 9:45 AM-12:45 PM<br />

Targeting the androgen receptor (AR) in women with AR� ER-/PR- metastatic<br />

breast cancer (MBC). Presenting Author: Ayca Gucalp, Memorial Sloan-Kettering<br />

Cancer Center, New York, NY<br />

Background: Patients (pts) with ER-/PR- MBC do not benefit from endocrine<br />

treatment (tx). Doane et al (Oncogene 2006;25:3994) described a subset <strong>of</strong><br />

ER-/PR- BC with a gene expression pr<strong>of</strong>ile similar to ER� BC but characterized<br />

by AR expression and AR-dependent growth in vitro. Hence, we conducted a<br />

multicenter phase II trial (NCT00468715) <strong>of</strong> the AR antagonist, bicalutamide<br />

(B) for pts with AR� ER-/PR- MBC. Methods: Pts with ER-/PR- (IHC �10%) MBC<br />

were consented to AR testing, confirmed centrally at MSKCC. If AR� (DAKO IHC<br />

�10%), pts were eligible for tx if ECOG performance status (PS) �2 and normal<br />

organ function. No limit on prior tx except prior trastuzumab required if HER2�.<br />

Eligible AR� pts who consented to tx received B 150mg orally daily in 28-day<br />

cycles (C). Toxicity assessed q4wks, response q12wks. Primary endpoint �<br />

clinical benefit rate (CBR): CR � PR � stable disease (ds) �6mo (SD). B would<br />

be considered worthy <strong>of</strong> further study if �4/28 pts have clinical benefit. Results:<br />

As <strong>of</strong> 12/21/11,436 pts consented for AR testing. 24 were ineligible, 12 await<br />

testing. 47/400 tested were AR� (12%). 26 received tx with B (6 ineligible for<br />

tx, 15 are eligible for tx with B at progression (POD) if clinically appropriate and<br />

study slots remains). Two AR� pts treated with B were ER� and removed from<br />

study. Three have received tx �12wks. Treated pt characteristics (n�24):<br />

median (med) age 64 (41-83), PS 0 (0-1), HER2� 1, visceral metastases 17.<br />

Prior chemotherapy: neo/adjuvant 16; med # regimens for MBC 1 (0-8). Med C#<br />

3 (2-49�). Best response: (21 evaluable pts): CBR 19% (95% CI 5-42%),<br />

SD�6mo 4, CR/PR 0, SD�6mo 3, POD 14. Conclusions: ~12% <strong>of</strong> ER-/PR- MBC<br />

pts are AR�. For these pts, AR-inhibition with B is feasible, well tolerated, and<br />

has activity based on pre-specified criteria. This study has closed to accrual for<br />

AR testing as <strong>of</strong> 1/2012. These results <strong>of</strong> the primary endpoint are not expected<br />

to change by accrual <strong>of</strong> the 2 remaining pts. Supported by TBCRC/Breast Cancer<br />

Alliance/AstraZeneca.<br />

Tx-related toxicity (n�26).<br />

Grade (n)<br />

Toxicity<br />

1 2 3<br />

AST/ALT/ bilirubin 5 2 3<br />

Nausea 2 0 1<br />

Bone pain 6 2 0<br />

Anorexia 3 1 0<br />

Constipation 2 1 0<br />

Vaginal dryness 1 1 0<br />

Anemia 0 1 0<br />

Headache 0 1 0<br />

Hot flashes 6 0 0<br />

Fatigue 5 0 0<br />

Edema 5 0 0<br />

Diarrhea 3 0 0<br />

Anxiety 2 0 0<br />

Rash 2 0 0<br />

Dyspnea<br />

No grade 4/5 events<br />

2 0 0<br />

1005 Oral Abstract Session, Tue, 9:45 AM-12:45 PM<br />

Correlation between the DCIS score and traditional clinicopathologic<br />

features in the prospectively designed E5194 clinical validation study.<br />

Presenting Author: Sunil S. Badve, Indiana University School <strong>of</strong> Medicine,<br />

Indianapolis, IN<br />

Background: We previously reported that in the E5194 clinical trial patients<br />

with ductal carcinoma in situ (DCIS) treated with wide local excision (WLE)<br />

without radiation (RT), the DCIS Score was significantly associated with 10<br />

year risk <strong>of</strong> an ipsilateral breast event (IBE - recurrence <strong>of</strong> in situ or invasive<br />

carcinoma), whether evaluated as a continuous or categorical variable<br />

(P�0.02 for both). Here we evaluate correlation between DCIS Score and<br />

clinicopathologic (CP) features and if DCIS Score provides independent<br />

recurrence risk information. Methods: The study population included 327<br />

women with DCIS prospectively selected for treatment with WLE without<br />

RT, including low-intermediate grade tumors �2.5 cm or high-grade �1<br />

cm. CP variables included age, menopausal status, tamoxifen treatment<br />

(used in 29%) and expert centrally determined tumor size, grade, comedo<br />

necrosis, tumor type, and margin status. The association between DCIS<br />

Score and CP variables was examined by spearman rank correlation, and<br />

proportional hazards regression models were used to determine variables<br />

significantly associated with IBE. Results: Lesion size (p�0.009) and<br />

menopausal status (p�0.03) were significantly associated with IBE, while<br />

grade (p�0.69) and comedo necrosis (p�0.47) were not. DCIS Score was<br />

significantly associated with IBE after adjustment for CP features and<br />

tamoxifen use (p�0.02). DCIS Score was moderately correlated with grade<br />

(rs�0.46; 95% CI 0.37,0.54), percentage comedo necrosis (rs�0.49; CI<br />

0.41,0.57), and lesion size (rs�0.18; CI 0.08,0.29) but not other<br />

features. Exploratory analyses in all CP subgroups, including the multicomponent<br />

Van Nuys Prognostic Index, showed a wide range <strong>of</strong> DCIS Scores in<br />

each subgroup. Concordance <strong>of</strong> the grades among readers was low: local vs<br />

parent central, 68%; local vs central nuclear grade, 45%; parent central vs<br />

central nuclear grade, 37%. Conclusions: DCIS Score is moderately<br />

correlated with grade, comedo necrosis, and tumor size. DCIS Score<br />

provides recurrence risk information independent <strong>of</strong> these features and<br />

identifies subjects with DCIS who are at high risk for local invasive and<br />

in-situ local recurrence after WLE alone.<br />

1007 Oral Abstract Session, Tue, 9:45 AM-12:45 PM<br />

Use <strong>of</strong> microRNA (miR) expression pr<strong>of</strong>iling to identify distinct subclasses<br />

<strong>of</strong> triple-negative breast cancers (TNBC). Presenting Author: Charles L.<br />

Shapiro, Division <strong>of</strong> Medical Oncology and the Breast Program, Columbus,<br />

OH<br />

Background: TNBC is divided into basal and non-basal subclasses. To<br />

further subclassify TNBC we performed microRNA (miR) expression pr<strong>of</strong>iles<br />

and linked them to patient overall survival. Methods: During 1996-<br />

2005, 365 consecutive TNBC (phenotypically estrogen, progesterone and<br />

HER2 negative by immunohistochemistry [IHC]) were identified from the<br />

NCCN Breast Cancer Data Base/Tumor Registry at OSU Medical Center.<br />

One hundred fifty-eight (43%) formalin-fixed paraffin embedded (FFPE)<br />

breast cancer and 40 normal breast tissue blocks were available and tissue<br />

cores were obtained for RNA. RNA was isolated using the Ambion recoverall<br />

total nucleic acid isolation kit and the expression <strong>of</strong> ~700 miRs was<br />

assessed for each sample using the nanoString nCounter method. A<br />

consensus-clustering algorithm (ConsensusClusterPlus, Bioconductor www-<br />

.bioconductor.org) was used to identify subclasses <strong>of</strong> TNBC and Kaplan-<br />

Meier overall survival curves were compared using the log-rank test.<br />

Censoring occurred at the date <strong>of</strong> death from causes other than breast<br />

cancer or at time <strong>of</strong> the last known follow-up, whichever occurred first. The<br />

median follow-up was 67 mo. (range 4-171 mo.). Results: The median age<br />

was 52 yrs. (range 20-84 yrs.); 81% white and 9% African-<strong>American</strong>;<br />

stages I, II, and III were 31%, 54% and 15%, respectively; and most<br />

patients received adjuvant anthracycline-based regimens with (25%) or<br />

without taxanes (75%). The algorithm identified 5 distinct subclasses; 1<br />

clustering with normal breast miR expression whereas the other 4 each had<br />

a unique pattern <strong>of</strong> deregulated miRs. The median overall survivals were<br />

significantly different across the 5 cancer subclasses (log-rank p�0.028)<br />

(Table). Conclusions: miR expression pr<strong>of</strong>iling identifies and discriminates<br />

5 TNBC subclasses, which do not coincide with those identified as basal<br />

and non-basal by IHC. Molecular analyses are ongoing to associate the<br />

miR-based subclasses with specific clinical features or the expression <strong>of</strong><br />

specific pathways.<br />

Group N<br />

Median overall<br />

survival (mo.) Log-rank p<br />

1 12 46 0.028<br />

2 10 50<br />

3 51 66<br />

4 48 67<br />

5 37 82<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


1008 Oral Abstract Session, Tue, 9:45 AM-12:45 PM<br />

Quantitative hormone receptors, triple-negative breast cancer (TNBC), and<br />

molecular subtypes: A collaborative effort <strong>of</strong> the BIG-NCI NABCG. Presenting<br />

Author: Maggie Chon U. Cheang, British Columbia Cancer Agency,<br />

Vancouver, BC, Canada<br />

Background: Most TNBC trials focusing on biology <strong>of</strong> the basal-like subtype<br />

(BLBC) allow borderline (1-10% staining) estrogen receptor (ER) and<br />

progesterone receptor (PgR) expression by immunohistochemistry (IHC);<br />

however the optimal ER and PgR cut points to enrich for non-luminal<br />

subtypes has not been studied. In this study,we compared quantitative<br />

ER/PgR status with gene expression-based intrinsic subtype in order to<br />

determine if borderline cases should be included in TNBC trials. Methods:<br />

ER, PgR, and HER2 status was determined by central review <strong>of</strong> tumors<br />

collected from three phase III randomized trials: GEICAM 9906 (n�820),<br />

NCIC CTG MA.5 (n�476) and MA.12 (n�398). PAM50 intrinsic subtyping<br />

(BLBC, HER2-enriched, Luminal A, Luminal B and Normal-like) was<br />

performed using the qRT-PCR-based assay. Quantitative ER/PgR expression<br />

by IHC and subtype was tested using ANOVA and Fisher’s exact test.<br />

Results: Of 1,694 tumors, 15% were BLBC, 21% HER2-Enriched, 33%<br />

Luminal A, 25% Luminal B and 4% Normal-like. BLBC subtypes were<br />

significantly associated with low expression <strong>of</strong> ER and PgR (median �<br />

0.05%) compared to other subtypes (p � 0.001). The vast majority <strong>of</strong><br />

BLBC (96%) did not express any ER or PgR protein by IHC. BLBC<br />

represented 73% <strong>of</strong> TNBC (borderline cases not included) and significantly<br />

more than the additional TNBC with borderline ER/PgR (p � 0.001).<br />

Within borderline ER/PgR and HER2-negative cases only, 17% were BLBC<br />

and 46% were luminal subtypes (Table). Conclusions: BLBC rarely express<br />

ER or PgR by IHC. The majority <strong>of</strong> borderline TNBC (1-10% ER/PgR) are<br />

not BLBC; half <strong>of</strong> them are categorized as luminal categories that may be<br />

endocrine sensitive. TNBC trials seeking to target BLBC tumor biology<br />

should use the ASCO/CAP guidelines <strong>of</strong> 0% as the cut<strong>of</strong>fs for ER and PgR<br />

negativity.<br />

Intrinsic subtypes by PAM50<br />

Basal-like HER2-enriched Luminal A Luminal B Normal-like Total<br />

<strong>Clinical</strong> subtypes<br />

ER/PgR (0%)/HER2- 207(73%) 48 (17%) 6 (2%) 15 (5%) 7 (3%) 283<br />

ER/PgR (1-10%)/HER2- 8 (17%) 14 (29%) 10 (21%) 12 (25%) 4 (8%) 48<br />

ER/PgR 11-100%/HER2- 8 (0.7%) 103 (10%) 490 (49%) 351 (35%) 47 (5%) 999<br />

1010 Poster Discussion Session (Board #2), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

ABT-888 (veliparib) in combination with carboplatin in patients with stage<br />

IV BRCA-associated breast cancer. A California Cancer Consortium Trial.<br />

Presenting Author: George Somlo, City <strong>of</strong> Hope, Duarte, CA<br />

Background: Platinum and PARP inhibitors have both shown activity in<br />

BRCA-associated breast cancer (BC) patients (pts). We have conducted a<br />

phase I trial <strong>of</strong> carboplatin (Carb) and velapirib [V], a PARP inhibitor, to<br />

define dose limiting toxicities [(DLT) during cycle (C) 1] and the maximum<br />

tolerated dose (MTD). Methods: BRCA 1 or 2 carriers with stage IV BC were<br />

eligible. Carb starting at an AUC <strong>of</strong> 6 was given IV every 21 days (length <strong>of</strong><br />

planned C) and V was administered orally, BID at dose levels (L) L1 through<br />

L5 (highest L planned). Results: 22 pts (21 eligible/evaluable, 20 with<br />

measurable BC) carrying BRCA1 (10) or BRCA2 (11), or both (1) mutations<br />

were accrued. Median age: 45 years, (32-65); 68% <strong>of</strong> BCs were ER�, and<br />

10% were HER2�. In the table below are the schema, incidence <strong>of</strong> DLTs,<br />

and # <strong>of</strong> Cs on study. Toxicities: At L1, grade ¾ DLTs with C 1 were seen in<br />

2/6 evaluable pts (1 pt w/grade 3 hyponatremia, pleural effusion, and<br />

dehydration, and 1pt w/grade 4 thrombocytopenia [PLT]), leading to<br />

deescalation <strong>of</strong> carb (AUC 5) for pts treated at Ls 2-5. At L2, 1 <strong>of</strong> 6 pts had<br />

grade 4 PLT. There were no DLTs at Ls 3 and L4. L5 is currently being<br />

expanded to 6 pts (3 currently enrolled, 1 pt with grade 4 granulocytopenia<br />

(Gr) and grade PLT reached DLT). Non-DLT dose delays mostly due � grade<br />

2 Gr or PLT were needed at 60%, 53%, 53%, and 43% <strong>of</strong> Cs in pts treated<br />

on Ls 1-4. Response: In 12 eligible pts treated at Ls 1 and 2, 2 complete<br />

and 6 partial responders (67%) and a clinical benefit (CB) <strong>of</strong> 75% were<br />

seen. All pts at Ls 3-5 are still being treated, and in pts treated at Ls 3 and<br />

4, 2 unconfirmed PRs, and 4 cases <strong>of</strong> stable disease were seen, with L5 too<br />

early to assess. Conclusions: The combination <strong>of</strong> Carb at an AUC <strong>of</strong> 5 and<br />

daily V at doses 150 to 200 mg BID is feasible and the MTD is being<br />

defined. In preliminary analysis, response and CB rates are better than<br />

expected with the individual agents alone, providing justification to<br />

proceed with a planned phase II randomized single agent versus combination<br />

trial.<br />

Dose levels L1 L2 L3 L4 L5 (highest planned L)<br />

Carb AUC 6 5 5 5 5<br />

Vinmg 50 BID 50 BID 100 BID 150 BID 200 BID<br />

#<strong>of</strong>pts 7 (1 ineligible) 6 3 3 3<br />

(and 3 in screening)<br />

Evaluable pts 6 6 3 3 3<br />

DLTs 2 1 0 0 1<br />

# <strong>of</strong> Cs given 9 (1-15) 8.5� (1-11) 6� (6�) 3� (3� to 4�) 1� (1� to 2�)<br />

Breast Cancer—Triple-Negative/Cytotoxics/Local Therapy<br />

51s<br />

1009 Poster Discussion Session (Board #1), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Phase I, open-label study <strong>of</strong> olaparib plus cisplatin in patients with<br />

advanced solid tumors. Presenting Author: Judith Balmaña, Vall d’Hebron<br />

University Hospital, Barcelona, Spain<br />

Background: Olaparib (AZD2281) is an oral PARP inhibitor active in<br />

advanced ovarian and breast cancers. We conducted a multicenter,<br />

dose-finding study assessing safety/ tolerability <strong>of</strong> olaparib capsules plus<br />

cisplatin in patients (pts) with advanced solid tumors (NCT00782574), for<br />

potential use in the neoadjuvant setting. Methods: Pts received 21-day(d)<br />

cycles <strong>of</strong> olaparib, continuously (Cont) or intermittently (Int), plus cisplatin<br />

on d1 <strong>of</strong> each cycle.Each cohort recruited �3 evaluable pts with expansion<br />

to �6 pts if �1 had a dose-limiting toxicity. The last cohort was expanded<br />

to ensure �6 pts completed 4 treatment cycles. Pts who completed 6<br />

combined therapy cycles or who stopped cisplatin due to cisplatin-related<br />

toxicity could enter the monotherapy phase (up to 400 mg BID olaparib).<br />

Primary objective: safety/ tolerability <strong>of</strong> �4 combined cycles; secondary<br />

objectives: pharmacokinetics, antitumor activity. Results: 54 pts received<br />

treatment; pts had breast (n�42), ovarian (n�10), pancreatic (n�1) or<br />

peritoneal (n�1) cancer. Median number <strong>of</strong> prior regimens � 4 (1–13). C2,<br />

C4 and C6 enrolled �6 pts. Most common grade (G) 3/4 AEs: neutropenia<br />

(n�9; 16.7%), leukopenia (n�4; 7.4%), anemia (n�3; 5.6%), vomiting<br />

(n�3; 5.6%). In C1–C5, 3 pts had AEs leading to discontinuation: 1 in C2<br />

(thrombocytopenia); 2 in C3 (fatigue; complex migraine, dyspnea). In C6,<br />

all pts have ended combination phase and no G3/4 hematologic AEs were<br />

seen. Overall, 46% <strong>of</strong> pts had a dose reduction; 32% due to hematologic<br />

AEs. There were no drug-related deaths. 35 pts (65%) completed �4<br />

combined cycles (C6, n�8). 18/54 evaluable pts (33%) had an objective<br />

response (complete, n�1; partial, n�17); 23 (43%) achieved stable<br />

disease. 7 pts (13%) had durable responses on monotherapy for �1 year.<br />

PK and BRCA1/2 data will be presented. Conclusions: Hematologic AEs led<br />

to dose reductions � schedule changes <strong>of</strong> olaparib with cisplatin 75<br />

mg/m2 . Tolerability improved with cisplatin 60 mg/m2 . Antitumor activity<br />

was seen during combination and olaparib monotherapy phases, with some<br />

long responders.<br />

1 2 3<br />

Cohort (C)<br />

4 5 6<br />

Cisplatin, d1/21<br />

Dose (mg/m2 )<br />

Olaparib<br />

75 75 75 75 75 60<br />

Dose (mg BID) 50 100 200 100 50 50<br />

Schedule Cont Cont Cont Int<br />

Int<br />

Int<br />

d1–10/21 d1–5/21 d1–5/21<br />

n 3 13 6 14 6 12<br />

1011 Poster Discussion Session (Board #3), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

SOLTI NeoPARP: A phase II, randomized study <strong>of</strong> two schedules <strong>of</strong> iniparib<br />

plus paclitaxel and paclitaxel alone as neoadjuvant therapy in patients with<br />

triple-negative breast cancer (TNBC). Presenting Author: Antonio Llombart,<br />

Solti Group, Valencia, Spain<br />

Background: Iniparib is an anticancer agent with a mechanism <strong>of</strong> action still<br />

under investigation. A phase 2 randomized neoadjuvant study in patients<br />

(pts) with TNBC was designed to explore the activity and tolerability <strong>of</strong> two<br />

schedules <strong>of</strong> iniparib with weekly paclitaxel (PTX). Here we report the<br />

efficacy and safety results from a planned interim analysis (IA). Methods:<br />

The trial accrued a total <strong>of</strong> 141 pts in October 2011, <strong>of</strong> whom, 74 are<br />

included in this IA. All were chemo-naive, histologicallyconfirmed Stage<br />

II-IIIA TNBC (IIA 47%; IIB 35%; IIIA 16%) with a median age <strong>of</strong> 50 yr.<br />

Triple negative status was centrally confirmed [ER/PR �10%, HER2 IHC<br />

(0�,1�) or FISH negative]. Pts were randomized (1:1:1) to receive weekly<br />

PTX (80 mg/m2 ,IV,d1;N�25) alone or in combination with iniparib,<br />

either on a once weekly (QW) (11.2 mg/kg, IV, d 1; N�25) or twice weekly<br />

(BIW) (5.6 mg/kg, IV, d 1, 4; N�24) schedule. The total planned treatment<br />

duration was 12 wks. The IA endpoint is pathological complete response in<br />

the breast (pCR) as assessed by independent pathologists. Results: Two/2/3<br />

pts in the PTX/QW/BIW arms, respectively, discontinued due to progressive<br />

disease per RECIST. Another 3/2/2 pts, respectively, discontinued due to<br />

investigator decision or an adverse event (AE). Thirteen pts presented with<br />

Grade 3/4 Treatment Emergent AE: 3 pts in PTX arm (1 neutropenia, 1<br />

presyncope, 1 ALT elevation), 3 in QW arm (1 lymphopenia, 1 hyperkalemia,<br />

1 pulmonary embolism), and 8 in the BIW arm (1 febrile neutropenia,<br />

3 neutropenia, 1 aphonia, 1 syncope, 1 radius fracture and 1 vertigo).<br />

Laboratory Grade 3/4 neutropenia occurred in 4% <strong>of</strong> pts in PTX, 0% in QW<br />

and 21% <strong>of</strong> BIW arms, with 1/2/3 pts, respectively, requiring G-CSF usage.<br />

There were 4/7/6 pts in the PTX/QW/BIW arms with PTX dose modifications.<br />

Four pts (16%) in PTX arm, 4 pts (16%) in the QW arm and 6 pts (25%) in<br />

the BIW arm had confirmed pCR in the breast. Conclusions: In this IA<br />

population, the addition <strong>of</strong> iniparib regardless <strong>of</strong> the schedule to weekly<br />

PTX did not seem to add clinically significant toxicity. pCR rate in the<br />

breast is similar across treatment arms at this IA. NCT01204125.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


52s Breast Cancer—Triple-Negative/Cytotoxics/Local Therapy<br />

1012 Poster Discussion Session (Board #4), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

DNA repair metagene signature as a prognostic and predictive factor in<br />

molecular breast cancer subtypes. Presenting Author: Libero Santarpia,<br />

Istituto Toscano Tumori Hospital <strong>of</strong> Prato, Prato, Italy<br />

Background: We aimed to assess the prognostic and predictive role <strong>of</strong> DNA<br />

repair genes in breast cancer (bc) molecular subtypes. Methods: We<br />

evaluated Affymetrix gene expression pr<strong>of</strong>iling from untreated N- patients<br />

(N � 684), neoadjuvant treated tumors with taxanes- (N � 320),<br />

anthracyclines- (N � 211), and cisplatin- (N � 22) containing regimens.<br />

We assessed within 3 BC molecular subgroups (ER�/HER2-, HER2�, and<br />

ER-/HER2-) bimodality distribution, prognosis by association with distant<br />

relapse (N � 454, N � 105, and N � 125) and predictive value for<br />

likelihood <strong>of</strong> pathological complete response (pCR) (N � 208, N � 105,<br />

and N � 240). Moreover, we explored the function <strong>of</strong> relevant genes in BC<br />

cell lines. Results: Three genes (ERCC2, XRCC3, and RECQL4) showed<br />

bimodality in each bc subtype. Eight genes were associated with poor<br />

prognosis (including RECQL4) and 1 gene with good prognosis (ATM)[P�<br />

.0001] only in ER�/HER2- tumors. Our results suggest a subtype and<br />

treatment specific association with pCR although they did not satisfy<br />

stringent criteria for false discovery correction. In ER-/HER2- mismatch<br />

repair (MR) (MSH2 and MSH6) and MTMR15 genes were associated with<br />

response and resistance to taxane-containing regimens, respectively.<br />

TOP2A was the only gene associated with response to anthracycline but not<br />

taxanes in HER2� tumors. RECQL4 had a positive trend with higher pCR in<br />

both ER� and ER-/HER2- tumors. In in vitro studies we found that<br />

RECQL4 interacts with PARP1 and that the expression <strong>of</strong> these genes was<br />

correlated with sensitivity to chemotherapy and PARP inhibitors.<br />

Conclusions: We identified MR genes as potential predictive markers <strong>of</strong><br />

response to taxanes-based regimens in ER-/HER2-. A novel gene RECQL4<br />

showed bimodal distribution, prognostic value, and a trend for predictive<br />

association with response to taxanes-based chemotherapy, which was also<br />

confirmed by in vitro analysis. ATM deserves further evaluation as prognostic<br />

marker in ER�/HER2-.<br />

1014 Poster Discussion Session (Board #6), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Targeting XRCC1 (X-ray repair cross-complementing gene 1) deficiency in<br />

tumors for personalized cancer therapy. Presenting Author: Srinivasan<br />

Madhusudan, School <strong>of</strong> Molecular Medical Sciences, Nottingham University<br />

Hospitals, Nottingham, United Kingdom<br />

Background: XRCC1 is essential for DNA base excision repair, single strand<br />

break repair and nucleotide excision repair. XRCC1 deficiency promotes<br />

genomic instability and may increase cancer risk. Methods: We evaluated<br />

XRCC1 immunohistochemically in early stage breast (n�2046), ovarian<br />

(n�157), gastric (n�140), colorectal (n�250) and pancreaticobiliary<br />

cancers (n�240). Pre-clinically, we evaluated a panel <strong>of</strong> XRCC1 deficient<br />

and pr<strong>of</strong>icient Chinese hamster ovary and human cancer cell lines. Double<br />

strand break repair (DSB) inhibitors targeting ATM (KU55933), DNA-PKcs<br />

(NU7441) and ATR (NU6027) were evaluated for synthetic lethality and<br />

cisplatin alone or in combination with DSB inhibitors for chemopotentiation.<br />

Results: In breast cancer,XRCC1 loss (16%) was associated with<br />

higher grade (p�0.0001), loss <strong>of</strong> hormone receptors (p�0.0001), presence<br />

<strong>of</strong> triple negative (p�0.0001) and basal like phenotypes (p�0.001).<br />

Loss <strong>of</strong> XRCC1 was associated with a 2-fold increase in risk <strong>of</strong> death and<br />

metastasis (p�0.0001) and independently with poor outcome (p�0.0001).<br />

In ovarian cancer, XRCC1 was positive in 44% <strong>of</strong> tumour and was<br />

significantly associated with higher stage (p�0.001), clear/endometroid<br />

type (p�0.015) and sub-optimal debulking (p�0.004). XRCC1 positive<br />

tumours were more resistant to platinum chemotherapy (p�0.0001).<br />

XRCC1 positivity conferred a 2 fold increase <strong>of</strong> risk <strong>of</strong> death (p�0.002) and<br />

independently associated with poor survival (p�0.002). In gastric cancers,<br />

XRCC1 was positive in 37% <strong>of</strong> tumours. This was significantly associated<br />

with high stage disease (p�0.001) and poor survival (p�0.001). Preclinically,<br />

KU55933, NU7441 and NU6027 were synthetically lethal in<br />

XRCC1 deficient compared to pr<strong>of</strong>icient cells as evidenced by DSB<br />

accumulation, G2/M cell cycle arrest and apoptosis. XRCC1 deficient cells<br />

were hypersensitive to cisplatin which was enhanced by DSB repair<br />

inhibitors compared to in pr<strong>of</strong>icient cells. Conclusions: This is the largest<br />

study to confirm the clinical significance <strong>of</strong> XRCC1 expression in solid<br />

tumours. XRCC1 deficiency in human tumours may be suitable for<br />

synthetic lethality application and exploited for cisplatin chemotherapy<br />

potentiation.<br />

1013 Poster Discussion Session (Board #5), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

HAGE (DDX43) protein expression as an independent biomarker <strong>of</strong> poor<br />

clinical outcome <strong>of</strong> breast cancer (BC) and potential as a therapeutic target<br />

for ER-negative BC. Presenting Author: Stephen Chan, Nottingham University<br />

Hospital, Nottingham, United Kingdom<br />

Background: Recently, we have confirmed that HAGE is involved in<br />

promoting proliferation as assessed by increased thymidine incorporation<br />

and our preliminary results using shRNA to permanently knockdown HAGE<br />

expression also suggests the involvement <strong>of</strong> HAGE in tumor motility and<br />

metastasis. In this study we aimed to analyze the expression <strong>of</strong> HAGE in<br />

large well-characterized BC cohorts to determine its relationship with other<br />

clinico-pathological parameters and to investigate its prognostic value.<br />

Methods: HAGE protein expression was assessed in: a) 40 normal breast<br />

tissue (NBT), b) 60 invasive BCs and their matching NBT, c) BC cell lines,<br />

d) A series <strong>of</strong> 1650 consecutive cases <strong>of</strong> primary BC who treated with<br />

adjuvant CMF and/or endocrine therapies. Further validation was performed<br />

in 2 independent series <strong>of</strong> high risk ER- BC: a) 300 ER –BC who did<br />

not received any CT and b) 396 ER- BC treated with adjuvant anthracycline<br />

(ATC) based CT. Results: The NBT showed negative HAGE expression<br />

(HAGE-) throughout. HAGE overexpression (HAGE�) was observed in 10%<br />

<strong>of</strong> BC and was significantly associated with aggressive clinico-pathological<br />

features including: ER-, high grade and triple negative phenotypes.<br />

Moreover, HAGE� expression showed an adverse outcome with a 2-4 fold<br />

increase in the risk <strong>of</strong> death, recurrence and metastases (ps�0.00001)<br />

compared to HAGE-; ps�0.0001. Using a multivariate Cox regression<br />

model including ER status, grade, size and tumour stage, HAGE expression<br />

was confirmed as a powerful independent prognostic factor (p�0.0001).<br />

The poor clinical outcome <strong>of</strong> HAGE� was further confirmed in high risk<br />

(NPI�3.4) ER- patients who did not received any CT (p�0.0001). While,<br />

adjuvant CT either CMF or ATC had a positive impact on HAGE�/high risk<br />

ER- BC as HAGE� had a similar risk <strong>of</strong> death, recurrence and distant<br />

metastases to HAGE- expression. Conclusions: This is the first report which<br />

shows HAGE to be a potential predictor for poor prognosis in BC patients,<br />

and may be an attractive novel target for molecular and vaccine therapy for<br />

those patients. A prospective trial <strong>of</strong> adjuvant chemotherapy/vaccine to<br />

confirm this finding is warranted.<br />

1015 Poster Discussion Session (Board #7), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Cancer gene pr<strong>of</strong>ile <strong>of</strong> metastatic breast cancer. Presenting Author: Funda<br />

Meric-Bernstam, University <strong>of</strong> Texas M. D. Anderson Cancer Center,<br />

Houston, TX<br />

Background: There is great interest in using genomic information to guide<br />

therapy selection in cancer patients. The aim <strong>of</strong> this study was to determine<br />

the spectrum <strong>of</strong> genomic alterations identified in MBC patients, and<br />

evaluate the concordance <strong>of</strong> alterations between primary and recurrent<br />

tumors. Methods: We performed comprehensive pr<strong>of</strong>iling on formalin-fixed<br />

paraffin embedded samples from 42 patients with MBC using a targeted<br />

next generation sequencing (NGS) assay in a CLIA laboratory (Foundation<br />

Medicine). Genomic libraries were captured for 3,230 exons in 182 cancer<br />

related genes plus 37 introns from 14 genes <strong>of</strong>ten rearranged in cancer and<br />

sequenced to an average depth <strong>of</strong> 390X with 99% <strong>of</strong> bases covered<br />

�100X. In total 30 primary and 37 recurrent tumors were pr<strong>of</strong>iled,<br />

including 3 separate recurrences in 1 patient and matched primaryrecurrences<br />

in 22 patients. Point mutations, indels, copy number alterations<br />

and rearrangements were assessed. Alterations that are targetable<br />

with established or investigational therapeutics were considered “actionable”.<br />

Results: At least 1 genomic alteration was identified in all but 2<br />

breast samples (both primary tumors). Point mutations were identified in<br />

several cancer-related genes including PIK3CA, TP53, PTEN, CDH1,<br />

ARID1A, AKT1, NF1, FBXW7 and FGFR3. Amplification was observed in<br />

HER2; 11<strong>of</strong>12HER2 IHC positive samples were found to have HER2<br />

gains by NGS; in addition, a HER2 gain was identified by NGS in a HER2-<br />

(1� IHC) sample. Amplification <strong>of</strong> PIK3CA, IGF1R, FGFR2, AKT2, MDM2,<br />

and MCL1 plus a CDKN2A homozygous deletion were also identified. While<br />

the majority <strong>of</strong> known driver alterations (85%) were concordant in the<br />

matched pairs <strong>of</strong> primary and recurrent tumors, in 11 <strong>of</strong> 22 sets there was<br />

at least 1 discordant driver alteration, and these included both gains and<br />

losses <strong>of</strong> potential therapeutic targets. Overall 32 <strong>of</strong> 42 patients (76%) had<br />

an actionable genomic alteration. Conclusions: Genomic pr<strong>of</strong>iling <strong>of</strong> breast<br />

cancer samples reveals genomic alterations in most metastatic breast<br />

cancer patients. Over three quarters <strong>of</strong> patients have actionable findings,<br />

suggesting that genomic pr<strong>of</strong>iling may assist in individualized pathwaydirected<br />

therapy.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


1016 Poster Discussion Session (Board #8), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Tesetaxel: Activity <strong>of</strong> an oral taxane as first-line treatment in metastatic<br />

breast cancer. Presenting Author: Andrew David Seidman, Memorial<br />

Sloan-Kettering Cancer Center, New York, NY<br />

Background: Tesetaxel, unlike standard taxanes (docetaxel, paclitaxel), is<br />

not a substrate for Pgp, a major cause <strong>of</strong> taxane resistance in tumor models.<br />

In a DU4475 breast cancer xenograft that overexpresses Pgp, tesetaxel<br />

induced a 94% reduction in tumor size, markedly exceeding the activity <strong>of</strong><br />

docetaxel (46%) and paclitaxel (26%). Tesetaxel is associated with<br />

substantially less neuropathy preclinically than equi-myelotoxic doses <strong>of</strong><br />

docetaxel. In clinical studies to date, tesetaxel is not associated with<br />

hypersensitivity reactions (0% incidence in � 450 patients [pts]), thus<br />

eliminating the need for premedication and extended observation. In a prior<br />

study, tesetaxel (27-35 mg/m2 Q3 wks) achieved a 38% partial response<br />

(PR) rate as 2nd-line therapy in pts with metastatic breast cancer (MBC)<br />

who had progressed after multidrug anthracycline-containing regimens. To<br />

extend these data, we initiated a phase 2 study <strong>of</strong> tesetaxel as 1st-line therapy in women with MBC. Methods: Eligibility included MBC; HER2-;<br />

ECOG PS 0-1; and adequate organ function. Adjuvant chemotherapy<br />

(including taxanes) was allowed. Tesetaxel was administered orally without<br />

anti-allergic premedication at a starting dose <strong>of</strong> 27 mg/m2 once every 3<br />

wks. Overall response rate (ORR; RECIST) was the primary endpoint.<br />

Results: All 45 pts have been accrued. Median age is 58 y (range 36-80);<br />

median time from diagnosis, 4.0 y (range 0-21); triple negative status, 8<br />

pts at diagnosis, 14 at time <strong>of</strong> metastasis. Metastatic sites are lung (22<br />

pts), lymph nodes (22), liver (24), and bone (21). Prior treatment includes<br />

anti-estrogen therapy (32 pts), adjuvant chemotherapy (31), prior taxane<br />

(25), and radiotherapy (28). ORR in 24 pts evaluable for response is 50%<br />

(1 CR [4%], 11 PR [46%]); 5 responding pts had prior taxane therapy.<br />

Neutropenia is the most common � Grade 3 adverse event with 50% <strong>of</strong><br />

casesobservedafterdoseescalationto35mg/m2 ;febrileneutropeniaandGrade<br />

3 peripheral neuropathy occurred in 2 pts each. There were no hypersensitivity<br />

reactions. Conclusions: Tesetaxel is highly active in 1st-line MBC and<br />

overcomes multiple limitations <strong>of</strong> standard taxanes. Updated ORR and PFS<br />

for all pts will be presented. Weekly dosing will be evaluated in a new cohort<br />

<strong>of</strong> pts.<br />

1018 Poster Discussion Session (Board #10), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Everolimus with paclitaxel plus bevacizumab as first-line therapy for<br />

HER2-negative metastatic breast cancer (MBC): A randomized, doubleblind,<br />

placebo-controlled phase II trial <strong>of</strong> the Sarah Cannon Research<br />

Institute (SCRI). Presenting Author: Denise Aysel Yardley, SCRI/Tennessee<br />

Oncology, PLLC, Nashville, TN<br />

Background: Constitutive activation <strong>of</strong> mTOR and amplified PI3K/Akt/<br />

mTOR signaling are common in MBC, and increase as treatment-resistance<br />

is acquired. Everolimus (E), an mTOR inhibitor, has single agent activity,<br />

combines well with paclitaxel (P) and bevacizumab (B), and prolonged PFS<br />

when added to AI therapy in BOLERO-2. In this randomized phase II trial, E<br />

was added to P/B as first-line treatment <strong>of</strong> HER2-negative MBC. Methods:<br />

Women with untreated HER2-negative MBC were randomized (1:1) to P<br />

90mg/m2 IV (days 1, 8, and 15) and B10mg/kg IV (days 1 and 15) q28<br />

days with E 10mg PO (Arm 1) or placebo PO (Arm 2) daily. Response<br />

assessment was performed q8 weeks until progression or intolerable<br />

toxicity. Primary endpoint was PFS. Secondary endpoints: safety, overall<br />

response rate, response duration, overall survival. 110 pts allowed detection<br />

<strong>of</strong> improvement in median PFS from 11 to 16 months with 70% power.<br />

Results: Between 8/2009 and 6/2011, 112 pts were randomized (Arm<br />

1�55; Arm 2�57). Median age: 58 years (range: 25-79). 88% were ER�<br />

or PR�. Pts received a median 5 treatment cycles (range: �1- 26�); 18<br />

(16%) pts remain on treatment (Arm1, 9; Arm 2, 9). Median PFS were 8.8<br />

months (Arm 1) and 7.1 months (Arm 2) (95% CIs 7.4-9.6; 5.5-9.1<br />

months, p�0.79, HR 0.94). Complete responses were observed in 5%<br />

[Arm1, 4 (7%); Arm 2, 2 (4%)] with partial responses in 49% [Arm1, 29<br />

(53%); Arm 2, 26 (46%)]. Responses rates in taxane pretreated pts Arm 1<br />

22%, Arm 2 12%. Hematologic toxicity was similar in both arms; grade 3<br />

mucositis occurred in 13% <strong>of</strong> E pts. Dose reductions (47% vs 25%) and<br />

interruptions (42% vs 26%) were more frequent with E due to mucositis<br />

and rash. Treatment discontinuation rates were similar. Conclusions: The<br />

addition <strong>of</strong> E did not result in a significant improvement in the efficacy <strong>of</strong><br />

weekly paclitaxel/bevacizumab in the first-line treatment <strong>of</strong> HER2-negative<br />

MBC although response rates and median PFS were better with E. Possible<br />

explanation for these results may include lower dose intensity in the E arm,<br />

treatment <strong>of</strong> less resistant pts, or intrinsic differences in E activity when<br />

added to antiestrogen vs chemotherapy.<br />

Breast Cancer—Triple-Negative/Cytotoxics/Local Therapy<br />

1017 Poster Discussion Session (Board #9), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Final analysis <strong>of</strong> phase II study <strong>of</strong> EZN-2208 (PEG-SN38) in metastatic<br />

breast cancer (MBC). Presenting Author: Cynthia R. C. Osborne, Baylor<br />

Sammons Cancer Center, Texas Oncology PA, US Oncology, Dallas, TX<br />

Background: EZN-2208 is a water-soluble PEGylated conjugate <strong>of</strong> SN38.<br />

EZN-2208 results in prolonged exposure <strong>of</strong> tumors to SN38 via preferential<br />

accumulation <strong>of</strong> EZN-2208 in the tumor and prolonged release <strong>of</strong> SN38.<br />

These data represent the final analysis <strong>of</strong> our study evaluating EZN-2208 in<br />

MBC. Methods: EZN-2208 9 mg/m2 (SN38 equivalents) was delivered as a<br />

60-minute IV infusion, weekly for 3 wks in 4-wk cycles. The primary<br />

objective was to determine the overall response rate (RR) in female patients<br />

with metastatic breast cancer (MBC) who had received prior adjuvant or<br />

metastatic therapy with either 1) anthracycline and taxane (AT) or 2)<br />

anthracycline, taxane, and capecitabine (ATX). Secondary objectives included<br />

evaluation <strong>of</strong> RR based on tumor receptor status, duration <strong>of</strong><br />

response, progression-free survival (PFS), overall survival (OS), and safety.<br />

Results: Patients with MBC (n�164) were treated with a median (range) <strong>of</strong><br />

3.3 (0.3-22) cycles <strong>of</strong> EZN-2208. The objective response rate (RR) was<br />

20% for AT and 9% for ATX. The clinical benefit rate (CBR�%CR � %PR<br />

� %SD �6 months) was 41% and 27% in patients in the AT and ATX<br />

cohorts, respectively. The RR and CBR among ER� patients were 11%<br />

(10/91 pts) and 41.8% (38/91 pts). In patients who progressed during or<br />

within 30 days <strong>of</strong> prior platinum-containing regimens (Platinum Progressors),<br />

the CBR was 20% (8/40 pts). Among triple negative breast cancer<br />

(TNBC) patients, the RR and CBR were 22.5% (11/49 pts) and 36.7%<br />

(18/49 pts). For TNBC, Platinum Progressors, the CBR was 26.1% (6/23<br />

pts). Overall, most common reported drug-related adverse events were<br />

diarrhea, nausea and neutropenia. Conclusions: EZN-2208 has notable<br />

activity in patients with previously treated MBC and appears to be an active<br />

agent for treatment <strong>of</strong> TNBC. Patients with TNBC, who had been previously<br />

treated with a platinum-based regimen, also derive clinical benefit from<br />

EZN-2208. The safety pr<strong>of</strong>ile <strong>of</strong> EZN-2208 is acceptable with good<br />

tolerability in most patients. Further evaluation <strong>of</strong> EZN-2208 in MBC in<br />

general and TNBC in particular is warranted.<br />

1019 Poster Discussion Session (Board #11), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Metformin in early breast cancer (BC): A prospective, open-label, neoadjuvant<br />

“window <strong>of</strong> opportunity” study. Presenting Author: Saroj Niraula,<br />

Division <strong>of</strong> Medical Oncology and Hematology, Princess Margaret Hospital<br />

and University <strong>of</strong> Toronto, Toronto, ON, Canada<br />

Background: There is growing evidence that metformin may exert anticancer<br />

effects through indirect (insulin-mediated) or direct (insulinindependent)<br />

mechanisms. Here, we report final results <strong>of</strong> a neo-adjuvant<br />

“window-<strong>of</strong>-opportunity” study <strong>of</strong> metformin in women with operable BC.<br />

Methods: Newly diagnosed, untreated, non-diabetic BC patients received<br />

metformin 500 mg tid after diagnostic core-biopsy until definitive surgery(no<br />

other treatment). <strong>Clinical</strong> [weight, symptoms, European Organization<br />

for Research and Treatment <strong>of</strong> Cancer Quality <strong>of</strong> Life Questionnaire<br />

(EORTC-QLQ-C-30)] and biologic characteristics [insulin, glucose, homeostatic<br />

model assessment (HOMA), C-reactive protein (CRP), leptin] were<br />

compared pre- and post-metformin as were Terminal deoxynucleotidyl<br />

transferase-mediated dUTP nick end labeling (TUNEL, an apoptotic<br />

marker) and Ki67 (primary end-point) scored blinded by manual count <strong>of</strong><br />

positive nuclear-staining. The planned sample-size <strong>of</strong> 40 patients gave<br />

90% power to detect a 5.5 percentage point change in Ki67. Results:<br />

Thirty-nine patients were enrolled and mean age was 51 years; metformin<br />

was given for 18 days (median), range 13-40 days. Twenty patients had T1<br />

and 19 T2/T3 tumors; 16 tumors were grade III; 24 were N0; 32 ER/PR<br />

positive, 5 HER-2 positive. Grade 1-2 self-limiting diarrhea, anorexia and<br />

abdominal distention occurred in 50%, 41% and 32%. EORTC QLQ scores<br />

were stable in all function domains and overall scores. Main study<br />

outcomes are tabulated here. Conclusions: Short-term preoperative metformin<br />

was well-tolerated and resulted in clinical and cellular effects in<br />

keeping with beneficial anti-cancer effects as demonstrated by improved<br />

insulin resistance (HOMA), decreased proliferation (ki67) and increased<br />

apoptosis (TUNEL).<br />

Variable (units)<br />

Pre-metformin<br />

mean (SD)<br />

53s<br />

Mean change<br />

(SD) p change<br />

Weight (kg) 70.3 (12.3) -1.2 (1.4) �0.0001<br />

BMI (kg/m 2 ) 26.9 (4.6) -0.5 (0.5) �0.0001<br />

CRP (mg/L) 2.0 (2.7) -0.2 (2.6) 0.35<br />

Glucose (mmol/L) 5.30 (0.56 -0.14 (0.43) 0.045<br />

Insulin (pmol/L) 43.4 (23.9) -4.7 (18.1) 0.069<br />

HOMA 1.47 (0.95) -0.21 (0.75) 0.047<br />

Leptin (ng/ml) 16.5 (13.0) -1.3 (7.2) 0.15<br />

TUNEL 0.56 (0.58) 0.49 (1.00) 0.0037<br />

Ki67 36.5 (24.8) -3.0 (9.8) 0.016<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


54s Breast Cancer—Triple-Negative/Cytotoxics/Local Therapy<br />

1020 Poster Discussion Session (Board #12), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

The impact <strong>of</strong> genetic variability on severe toxicity <strong>of</strong> (neo-) adjuvant<br />

chemotherapy in breast cancer patients receiving 5-fluorouracil, epirubicin,<br />

and cycl<strong>of</strong>osfamide (FEC). Presenting Author: Christ<strong>of</strong> Vulsteke,<br />

Department <strong>of</strong> General Medical Oncology, University Hospitals Leuven,<br />

Leuven, Belgium<br />

Background: We assessed the impact <strong>of</strong> single nucleotide polymorphisms<br />

(SNP) <strong>of</strong> potential genes <strong>of</strong> interest in germline DNA on severe adverse<br />

events in breast cancer (bc) patients receiving (neo-) adjuvant FEC<br />

chemotherapy. Methods: Cases were retrospectively evaluated through<br />

electronic chart review for febrile neutropenia (primary endpoint), febrile<br />

neutropenia first cycle, prolonged grade 4 or deep (�100/�l) neutropenia,<br />

anemia grade 3-4, thrombocytopenia grade 3-4 and non-hematological<br />

grade 3-4 events. The panel <strong>of</strong> genes, genotyped using iPLEX technology on<br />

a MALDI-TOF based MassARRAY Compact Analyser (Sequenom Inc., CA,<br />

USA), included ABCBI, ABCC1, ABCC2, ABCG2, ALDH3A1, CYP2B6,<br />

CYP2C8, CYP2C9, CYP2C19, CYP3A4, CYP3A5, DPYD, FGFR4, GPX4,<br />

GSTA1, GSTP1, MTHFR, NQ01, TYMS, XPD/ERCC2, XRCC1, UGT1A1,<br />

UGT1A6 and UGT2B7. These genes are involved in the metabolization <strong>of</strong><br />

the studied chemotherapeutics. Because <strong>of</strong> multiple testing the false<br />

discovery rate (FDR) was calculated. Results: We identified 1089 patients<br />

treated between 2000-2010 with 3-6 cycles <strong>of</strong> FEC, for whom germline<br />

DNA was available.Homozygous (TT, 0.5%) and heterozygous (GT,11%)<br />

genotypes for rs4148350 in the Multi Drug Resistance Protein I (ABCC1/<br />

MRP1), compared to wild-type (GG, 88.5%), were associated with febrile<br />

neutropenia, febrile neutropenia in first cycle, prolonged grade 4 or deep<br />

neutropenia and thrombocytopenia (80 vs 25 vs 15.7%, 40 vs 17.6 vs<br />

9.5%, 100 vs 41.7 vs 33.8% and 20 vs 2.8 vs 0.34% respectively; p<br />

0.0006, 0.01, 0.002 and 0.008 FDR 0.03, 0.65, 0.06 and 0.2). Variant<br />

genotypes for rs45511401 (GT/TT, 12%) in ABCC1, compared to wild-type<br />

(GG, 88%), were associated with febrile neutropenia, febrile neutropenia in<br />

first cycle and thrombocytopenia (26.5 vs 15,8%, 17.1 vs 9.7% and 3.4 vs<br />

0.3%, respectively; p 0.007, 0.03 and 0.005, FDR 0.2, 0.75 and 0.2).<br />

Conclusions: Genetic variation in the ABCC1 gene was strongly associated<br />

with severe hematological toxicity <strong>of</strong> FEC. Other previously described SNP<br />

were not validated. This is the largest bc study in which the impact <strong>of</strong><br />

genetic variability on the adverse events <strong>of</strong> FEC chemotherapy was<br />

investigated.<br />

1022 Poster Discussion Session (Board #14), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Comparison <strong>of</strong> molecular (BluePrint�MammaPrint) and pathological subtypes<br />

for breast cancer among the first 800 patients from the EORTC 10041/BIG 3-04<br />

(MINDACT) trial. Presenting Author: Giuseppe Viale, European Institute <strong>of</strong><br />

Oncology, Milan, Italy<br />

Background: Biology has become the main driver <strong>of</strong> breast cancer therapy.<br />

Intrinsic biological subtypes by gene expression pr<strong>of</strong>iling have been identified.<br />

Pathology can be used to define surrogates <strong>of</strong> these subtypes but these are not<br />

always concordant, which may lead to different treatment plans. We investigated<br />

the concordance between BluePrint (BP) � MammaPrint (MP) (micro array<br />

based) breast cancer subtypes and pathological surrogates (based on ER, PR,<br />

HER2, and Ki67). Contrary to the Perou gene set (evolved into PAM50),<br />

BluePrint was trained using pathological data. Methods: Using available data<br />

(centrally assessed pathology and genomic) from the MINDACT pilot phase<br />

(Rutgers et al 2011) 621 tumors were analyzed. Two pathology classifications<br />

were used: one with 4 categories and one with 5 categories (Goldhirsch et al<br />

2011). Based on BP 3 subtypes are formed: Luminal, HER2 and Basal. The<br />

Luminal subtype is further split into Luminal A (MP low risk) and Luminal B (MP<br />

high risk). Results: See table. Conclusions: All pathological Basal cases are BP<br />

Basal, apart from 1 BP HER2 case. Of the BP Basal cases, 15 are not<br />

pathological Basal: 1 is Luminal A, 11 are Luminal B (<strong>of</strong> which 8 are IHC ER/PR<br />

borderline (�1% and � 10%)) and 3 are HER2. All pathological Luminal (A & B)<br />

that are BP HER2 are HER2- by TargetPrint. 25 <strong>of</strong> the 26 pathological HER2�<br />

that are BP Luminal A are ER�. Most discordant cases are seen within the<br />

Luminal subtype, indicating that Ki67 discriminates Luminal A vs. B differently<br />

than MammaPrint does. The observed subtype discrepancies reveal potential<br />

important impact for treatment-decision making. MINDACT will provide important<br />

information.<br />

4 category St Gallen 5 category<br />

Luminal A<br />

Er� and/or PR�<br />

HER2-<br />

Ki67 low<br />

Luminal B<br />

ER� and/or PR�<br />

HER2-<br />

Ki67 high<br />

HER2<br />

HER2�<br />

Basal<br />

ER-<br />

PR-<br />

HER2-<br />

Pathology BluePrint (BP) � MammaPrint (MP)<br />

Luminal A<br />

Idem<br />

Luminal B (HER2-)<br />

Idem<br />

Luminal B (HER2�)<br />

ER� and/or PR�<br />

HER2�<br />

Erb-B2<br />

ER-<br />

PR-<br />

HER2�<br />

Basal<br />

Idem<br />

Luminal A<br />

BP Luminal<br />

MP low risk<br />

Luminal B<br />

BP Luminal<br />

MP high risk<br />

HER2<br />

BP HER2<br />

263 19 4<br />

(all TP* HER2-)<br />

111 70 4<br />

(all TP* HER2-)<br />

25<br />

(All ER�)<br />

Basal<br />

BP Basal<br />

Total<br />

1 287<br />

11 196<br />

3 31 1 60<br />

1 0 13 2 16<br />

0 0 1<br />

(all TP* HER2-)<br />

61 62<br />

Total<br />

*TP � TargetPrint.<br />

400 92 55 76 621<br />

1021 Poster Discussion Session (Board #13), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

10-year update <strong>of</strong> E2197: Phase III doxorubicin/docetaxel (AT) versus<br />

doxorubicin/cyclophosphamide (AC) adjuvant treatment <strong>of</strong> LN� and highrisk<br />

LN- breast cancer and the comparison <strong>of</strong> the prognostic utility <strong>of</strong> the<br />

21-gene recurrence score (RS) with clinicopathologic features. Presenting<br />

Author: Joseph A. Sparano, Albert Einstein College <strong>of</strong> Medicine, Bronx, NY<br />

Background: At 5 years, AT did not improve disease free survival or overall<br />

survival and RS was a more accurate predictor <strong>of</strong> relapse than standard<br />

clinicopathologic characteristics for patients with hormone receptor (HR)<br />

positive tumors. Methods: A Phase III Intergroup trial tested adjuvant AT vs.<br />

AC. Women with 1-3 N � or N - and T-size � 1cm were randomized to 4<br />

cycles <strong>of</strong> AT (60 mg/m2 /60 mg/ m2 ) or AC (60 mg/m2 /600 mg/m2 )q3wkx<br />

4. Patients(pts) with ER � and/ or PR � tumors received tam for 5 yrs. Pts<br />

were stratified by nodal, HR (ER� PR�, ER�PR-, ER-PR�, ER-PR-,<br />

ER/PR unk) and menopausal status. The primary endpoint was DFS. A<br />

sample <strong>of</strong> 465 pts with HR � breast cancer with 0 to 3 positive axillary<br />

nodes who did (N �116) or did not have a recurrence had tumor tissue<br />

evaluated using the 21- gene assay. Grade and HR expression were<br />

evaluated locally and centrally. Results: 2952 pts were randomized<br />

between 7/30/98 and 1/21/00. 2883 were eligible and analyzable. Arms<br />

were balanced for age, HR, menopause, nodes, surgery, grade and T-size:<br />

median age 51; 64% ER �; 65% LN-; grade: 10% low, 38% int., 46%<br />

high; and median T-size - 2.0 cm. At a median follow-up <strong>of</strong> 11.5 years the<br />

DFS/OS results are shown in the table below. RS was a highly significant<br />

predictor <strong>of</strong> recurrence including node negative and node positive disease<br />

(P � .0001) and predicted recurrence more accurately than clinical<br />

variables. Conclusions: At 11.5 yrs. median follow-up, there remains no<br />

difference in DFS or OS, although there continue to be fewer events in the<br />

AT arm in the prespecified ER/PR negative subgroup. At 10 years, the RS<br />

continues to be a more accurate predictor <strong>of</strong> relapse than standard clinical<br />

features.<br />

Hazard ratio (HR)*,<br />

5-yr DFS<br />

Hazard ratio (HR)*,<br />

10-yr DFS<br />

95Cl, p- value** 5 yr AT AC 95Cl, p value** 10 yr AT AC<br />

Overall DFS 1.02 (0.86-1.22), 0.78 85% 85% 1.02 (0.88-1.18), 0.83 77% 77%<br />

ER� 0.89 (0.71-1.12), 0..32 87% 88% 0.91 (0.76-1.10), 0.34 78% 79%<br />

ER- 1.24 (0.95-1.62), 0.12 81% 77% 1.22 (0.96-1.56), 0.11 76% 71%<br />

OS 1.06 (0.85-1.31), 0.62 92% 91% 1.03 (0.86-1.23), 0.73 85% 84%<br />

*HR�1 favors AT; **based on log-rank test.<br />

1023 Poster Discussion Session (Board #15), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Prognostic and predictive impact <strong>of</strong> Ki-67 before and after neoadjuvant<br />

chemotherapy on PCR and survival: Results <strong>of</strong> the GeparTrio trial. Presenting<br />

Author: Gunter Von Minckwitz, German Breast Group, Neu-Isenburg,<br />

Germany<br />

Background: We previously reported as a result <strong>of</strong> the GeparTrio phase III<br />

trial that response-guided neoadjuvant chemotherapy (CT) with TACx8 or<br />

TAC/NX, compared to TACx6, can improve survival especially in hormonereceptor<br />

(HR)-positive tumors. As this benefit could not be predicted by<br />

pathological complete response (pCR), better surrogate response markers<br />

are warranted. Methods: 2072 patients with operable or locally advanced<br />

breast cancer were treated with 2 cycles TAC before interim response<br />

assessment. Responders were randomized to additional TACx4 or TACx6<br />

and non-responders to TACx4 or NXx4. We centrally measured Ki-67<br />

in1165 pre-CT core biopsies and in 676 post-CT surgical samples.<br />

Counting patients with a pCR as having 0% Ki-67, 757 pre-/ post-CT pairs<br />

were available. Ki-67 percentage levels were grouped to low (0-15%),<br />

moderate (15.01-35%), and high (35.01-100%) according to cut-point<br />

finding analysis in a training and validation cohort. Results: pCR rates were<br />

4.2%, 12.9%, and 29.0% in tumors with low, moderate, and high pre-CT<br />

Ki-67 levels (p�0.0001). Pre-CT Ki-67 levels significantly predicted<br />

disease-free survival (DFS) (log rank p�0.0001) overall, in the HR�<br />

(p�0.0001), but not in the HR- (p�0.5) subgroup. Post-CT Ki-67 levels<br />

correlated with DFS (p�0.0001). Patients with low post-CT Ki-67 levels<br />

showed comparable outcome to patients with pCR. Patients with increased<br />

Ki-67 levels from before to after CT showed an impaired outcome compared<br />

to patients with stable or decreased Ki-67 levels (p�0.0001). However,<br />

post-CT Ki-67 levels appeared to have more prognostic relevance than<br />

Ki-67 changes. Low post-CT Ki-67 levels were not more frequent after<br />

response-guided treatments (response-guided vs conventional: p�0.153;<br />

TACx6 vs TACx8: p�0.335; TACx6 vs TAC/NX: p�0.420). Similar negative<br />

results were found for HR� and HR- subgroups. Conclusions: Pre-CT Ki-67<br />

levels are predictive for pCR and prognostic for DFS. Post-CT Ki-67 levels<br />

and changes between pre-and post-CT-Ki-67 levels are prognostic for DFS.<br />

As neither could predict different treatment effects on DFS, Ki67 cannot<br />

replace pCR as a surrogate marker for outcome after neo-adjuvant CT.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


1024 Poster Discussion Session (Board #16), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

A gene expression signature <strong>of</strong> MEK pathway activation to predict survival<br />

in triple-negative breast cancer. Presenting Author: Justin M. Balko,<br />

Vanderbilt University, Nashville, TN<br />

Background: Tumor cell proliferation measured by Ki67 in the surgically<br />

removed tumor after neoadjuvant chemotherapy (NAC) has been shown to<br />

predict patient outcome in breast cancer. It is unclear from these studies if<br />

breast cancer subtype may account in part for the predictive ability <strong>of</strong> Ki67.<br />

Thus, we tested whether Ki67 score in the surgically-resected residual<br />

tumor (RT) after NAC predicted outcome in a cohort <strong>of</strong> triple negative<br />

breast cancer (TNBC). Gene expression pr<strong>of</strong>iling was performed to identify<br />

molecular subtype and test the association with gene modules indicative <strong>of</strong><br />

signaling pathway activation. Methods: Ki67 was scored in the RT <strong>of</strong> 89<br />

patients with stage II-III TNBC (ER/PR/HER2 negative by IHC at diagnosis)<br />

that had been treated with NAC. Expression levels for 450 genes were<br />

quantified by Nanostring. Ki67, node and menopause status, age, therapy<br />

type (� taxanes), molecular subtype, and gene expression scores were<br />

tested for association to RFS and OS using univariate and multivariate<br />

CoxPH models. Results: Ki67 score in the post-NAC RT demonstrated a<br />

wide range (1.5-77.7%; median: 36.2%). Twenty seven % <strong>of</strong> RTs were 3�<br />

HER2 by IHC. Molecular subtype in the RT was as follows: 64% Basal-like;<br />

20% HER2-enriched; 6% LumA; 6% LumB; 4% Normal-like. In univariate<br />

analyses to respectively predict RFS and OS, node status (p�0.005 and<br />

p�0.02), number <strong>of</strong> nodes (p�0.003 and p�0.001), and the score <strong>of</strong> a<br />

gene expression module <strong>of</strong> MEK pathway activation (p�0.04 and p�0.01,)<br />

were significant. Basal-like subtype approached significance for RFS<br />

(p�0.1) and OS (p�0.05). In multivariate analyses, node status (p�0.002<br />

for RFS and p�0.001 for OS) and MEK pathway activation score (p�0.04<br />

and p�0.01) were significant predictors. Conclusions: Ki67 in the RT after<br />

NAC was not predictive <strong>of</strong> outcome in a cohort <strong>of</strong> TNBC. However, a gene<br />

expression signature <strong>of</strong> activated MEK was negatively associated with<br />

outcome. These data are consistent with the reported preclinical activity <strong>of</strong><br />

MEK inhibitors against basal-like breast cancer cells and a possible role <strong>of</strong><br />

this signaling pathway in chemotherapy resistance. They also support deep<br />

sequencing studies to identify genetic alterations in the RAS/MEK/MAPK<br />

pathway in TNBC.<br />

1026 Poster Discussion Session (Board #18), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

A phase II study <strong>of</strong> preoperative (preop) bevacizumab (bev) followed by<br />

dose-dense (dd) doxorubicin (A)/cyclophosphamide (C)/paclitaxel (T) in<br />

combination with bev in HER2-negative operable breast cancer (BC).<br />

Presenting Author: Sara M. Tolaney, Dana-Farber Cancer Institute, Boston,<br />

MA<br />

Background: Two recent preop studies evaluating bev showed conflicting<br />

results, particularly in hormone receptor (HR)� BC. Identification <strong>of</strong><br />

predictive markers and their relationship to the pharmacodynamic effects<br />

<strong>of</strong> bev would facilitate the identification <strong>of</strong> BCs most likely to benefit from<br />

bev. To accomplish these goals, we conducted a unique preop trial with a<br />

run-in <strong>of</strong> single agent bev followed by ddACT with bev in two cohorts, one<br />

with HR�HER2– BC, and a smaller triple negative (TN) cohort. Methods:<br />

Pts with HR�, HER2– or TN BC were eligible if their tumor (T) was �1.5<br />

cm and high grade, or had axillary LN involvement; or if T�2.5cm and was<br />

low/intermediate grade. Treatment consisted <strong>of</strong> a single dose <strong>of</strong> bev 10<br />

mg/kg, followed two wks later by A 60 mg/m2 and C 600 mg/m2 with bev 10<br />

mg/kg q2 wks x 4, followed by T 175 mg/m2 with bev 10 mg/kg q2 wks x 3,<br />

followed by T 175 mg/m2 x1. Research core biopsies and interstitial fluid<br />

pressure (IFP) were assessed pre- and post- bev alone. Pathologic response<br />

was confirmed centrally and Miller-Payne (MP) was assessed. Results: 84<br />

pts with HR� and 20 pts with TN breast cancer were enrolled. Amongst<br />

HR� pts, 74 had surgical tissue centrally reviewed, and 6 (8%) had a pCR.<br />

Amongst TN pts, 18 pts had tissue centrally reviewed and 8 (44%) had a<br />

pCR. Grade was found to predict MP response in both HR� and TN pts<br />

(p�0.001). Several biomarkers were evaluated as predictors <strong>of</strong> response to<br />

bev. Baseline sVEGFR1 correlated with MP response to treatment among<br />

TN pts (p�0.015). Single-agent bev reduced the mean vascular density by<br />

18.5% (p�0.049) in HR� patients and the mean IFP in the overall cohort<br />

and HR� patients by 20 (p�0.020) and 24.5% (p�0.001), respectively.<br />

The reductions in IFP correlated with higher levels <strong>of</strong> sVEGFR2 (p�0.003).<br />

The IFP decreased � 50% in 24/65 pts and did not change in others. Gene<br />

expression pr<strong>of</strong>iling by PAM50 is underway. Conclusions: The addition <strong>of</strong><br />

bev to preop chemotherapy is well tolerated. Tumor grade appears to<br />

predict MP response in HR� and TN tumors, and sVEGFR1 may be a<br />

predictor <strong>of</strong> MP response to bev in TN tumors. Further work for biomarker<br />

predictors <strong>of</strong> response to bev is ongoing.<br />

Breast Cancer—Triple-Negative/Cytotoxics/Local Therapy<br />

55s<br />

1025 Poster Discussion Session (Board #17), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

NSABP FB-6: Phase II trial <strong>of</strong> weekly paclitaxel (WP) and pazopanib<br />

following doxorubicin and cyclophosphamide (AC) as neoadjuvant therapy<br />

for HER2-negative locally advanced breast cancer (LABC). Presenting<br />

Author: Antoinette R. Tan, National Surgical Adjuvant Breast and Bowel<br />

Project and The Cancer Institute <strong>of</strong> New Jersey, New Brunswick, NJ<br />

Background: Pazopanib is an oral, small molecule inhibitor <strong>of</strong> VEGFR-1, -2,<br />

and-3, PDGFR-�, and -�, and c-kit tyrosine kinases. The purpose <strong>of</strong> this<br />

trial was to determine the activity and safety pr<strong>of</strong>ile <strong>of</strong> pazopanib when<br />

added to neoadjuvant WP following AC in LABC. The primary endpoint was<br />

pathologic complete response in the breast and nodes (pCR-BN). Methods:<br />

Women with HER2-negative stage IIIA-IIIC breast cancer were treated with<br />

AC (60 mg/m2 /600 mg/m2 ) for 4 cycles every 3 weeks followed by WP 80<br />

mg/m2 on days 1, 8, and 15 every 28 days for 4 cycles concurrently with<br />

pazopanib 800 mg orally daily prior to surgery. Postoperatively, pazopanib<br />

was given for 6 months. The regimen would be considered active if �14<br />

responses (16% pCR rate in breast and nodes) were observed in 87<br />

evaluable patients. Patients were considered evaluable if they received at<br />

least 1 dose <strong>of</strong> pazopanib. Results: Between July 2009 and March 2011,<br />

101 pts (median age 51 yrs, range 30-71) were enrolled; 56% had stage<br />

IIIA, 34% stage IIIB, and 10% stage IIIC disease. 74 pts (73%) had<br />

ER-and/or PR-positive tumors and 27 pts (27%) were triple negative. 8<br />

patients did not begin pazopanib. The pCR-BN rate in evaluable patients<br />

for whom surgical information was known was 18% (16/89). The pCR-BN<br />

rate in ER positive disease was 9% (6/65) and was 42% (10/24) in TNBC.<br />

Toxicities observed with WP and pazopanib included diarrhea (gr 2/3,<br />

10%/5%), hand-foot syndrome (gr 2/3, 11%/1%), hypertension (gr 2/3,<br />

12%/3%), neuropathy (gr 2/3, 14%/1%), and neutropenia (gr 3/4, 25%/<br />

1%). Liver toxicity during WP and pazopanib included ALT (gr 2/3/4,<br />

13%/7%/1%), AST (gr 2/3, 7%/7%), and total bilirubin (gr 2, 2%).<br />

Conclusions: A regimen <strong>of</strong> WP and pazopanib following AC was active as<br />

neoadjuvant therapy in women with LABC and met the pre-specified criteria<br />

<strong>of</strong> interest. The activity in TNBC was notable. The toxicity pr<strong>of</strong>ile <strong>of</strong> WP and<br />

pazopanib was consistent with previous experience. Support: GlaxoSmith-<br />

Kline.<br />

1027 Poster Discussion Session (Board #19), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Association <strong>of</strong> a compact 13-gene VEGF signature with OS in E2100.<br />

Presenting Author: Scooter Willis, Scripps Research Institute, Jupiter, FL<br />

Background: E2100, an open-label, randomized, phase III trial, demonstrated<br />

a significant improvement in progression free survival and overall<br />

response rate with paclitaxel plus bevacizumab compared with paclitaxel<br />

alone as initial chemotherapy for patients with HER2-negative metastatic<br />

breast cancer. Genentech completed additional clinical trials and submitted<br />

these data to the FDA. On 18 Nov, 2011, the FDA Commissioner<br />

revoked the agency’s approval <strong>of</strong> bevacizumab for the breast cancer<br />

indication because <strong>of</strong> the lack <strong>of</strong> evidence <strong>of</strong> an improvement in overall<br />

survival or a clinical benefit to patients sufficient to outweigh the risks.<br />

However, the Commissioner “encouraged Genentech to consider additional<br />

studies to identify if there are select subgroups <strong>of</strong> women who might benefit<br />

from this drug”. Hu et al. (BMC Medicine 2009) published a compact 13<br />

gene VEGF-signature associated with distant metastases and poor outcomes.<br />

Supervised analyses comparing patients with distant metastases<br />

versus primary tumors or regional metastases showed that the distant<br />

metastases were distinct and distinguished by the lack <strong>of</strong> expression <strong>of</strong><br />

fibroblast/mesenchymal genes, and by the high expression <strong>of</strong> a 13 gene<br />

pr<strong>of</strong>ile that included VEGF, ANGPTL4, ADM and the monocarboxylic acid<br />

transporter SLC16A3. Methods: We have investigated the VEGF signature in<br />

silico on Illumina DASL analysis <strong>of</strong> 122 FFPE samples remaining from<br />

E2100. Results: PFS benefit is seen for pacli � bev vs pacli in both<br />

treatment arms with the low VEGF signature (HR 0.45 95% CI .27-.77 p<br />

.009 n 67) and with the high VEGF signature (HR 0.57 95% CI .32-1.0 p<br />

.015 n 55). However, OS benefit is only seen for pacli � bev vs pacli in the<br />

high VEGF group (HR 0.56 95% CI .30-1.05 p .02 n 52) and not in<br />

patients with the low VEGF signature (HR 1.12 95% CI .66-1.90 p .81 n<br />

67). Conclusions: Hence, this signature, which suggests that the response<br />

to hypoxia includes the ability to promote new blood and lymphatic vessel<br />

formation, shows great potential as a predictive biomarker <strong>of</strong> those patients<br />

to whom bevacizumab would convey an OS advantage benefit. We note with<br />

great caution that this exploratory analysis <strong>of</strong> trial subset is underpowered,<br />

hence, this compact VEGF signature is being pursued in other bevacizumab<br />

trial sets.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


56s Breast Cancer—Triple-Negative/Cytotoxics/Local Therapy<br />

1028 Poster Discussion Session (Board #20), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Are mastectomy rates really increasing? Experiences from a single institution<br />

and a population-based database. Presenting Author: Min Yi, University<br />

<strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: Recent studies have reported increased mastectomy rates for<br />

the treatment <strong>of</strong> early stage breast cancer during the last decade. The aims<br />

<strong>of</strong> this study were to examine trends in mastectomy rates at a single<br />

institution and in a population-based database and to compare differences<br />

between the two cohorts. Methods: Patients with stage 0-II breast cancer<br />

diagnosed from 2000 to 2008 were identified from our cancer center<br />

institutional database (CC cohort, n�8,915) and the Surveillance, Epidemiology<br />

and End Results database (SEER cohort, n�359,572). Patients<br />

without primary surgery or unknown surgery type were excluded. Mastectomy<br />

rates by the year <strong>of</strong> diagnosis were evaluated and multivariable<br />

logistic regression models were built to identify clinicopathologic factors<br />

that predicted mastectomy as the treatment choice. Results: The proportion<br />

<strong>of</strong> patients treated with mastectomy decreased from 44.5% to 37.8%<br />

between 2000 and 2005 in the CC cohort (P�0.003) and from 42.8% to<br />

36.6% in the SEER cohort (P�0.0001). Subsequently, the mastectomy<br />

rate increased to 48.6% in the CC cohort (P�0.0001) and to 40.1% in the<br />

SEER cohort by 2008 (P�0.0001). Multivariable analysis found that<br />

patients with younger age (�50), stage 0 or II cancer vs. stage I, high grade<br />

tumor, low median household income, and lobular histology were more<br />

likely to choose mastectomy in both the SEER and CC cohorts. In the CC<br />

cohort, patients with preoperative breast MRI were also more likely to<br />

undergo mastectomy. The percentages <strong>of</strong> patients receiving preoperative<br />

MRI and choosing prophylactic contralateral mastectomy increased each<br />

year in the CC cohort. The rate <strong>of</strong> preoperative breast MRI increased from<br />

4.7% in 2005 to 9.6% in 2008 (P�0.0001). Patients choosing prophylactic<br />

contralateral mastectomy increased from 8.4% in 2005 to 11.8% in<br />

2008 (P�0.06). Conclusions: Our study shows that there was a decrease in<br />

mastectomy rates from 2000 to 2005 and a subsequent increase in<br />

mastectomy rates from 2005-2008 in both the CC and SEER cohorts.<br />

Increased use <strong>of</strong> preoperative breast MRI and the decision to undergo<br />

contralateral prophylactic mastectomy likely contributed to the increased<br />

mastectomy rates in the CC cohort.<br />

1030 Poster Discussion Session (Board #22), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Short-term complications and use <strong>of</strong> breast brachytherapy in the national<br />

Medicare population in 2008-2009. Presenting Author: Carolyn J. Presley,<br />

Yale New Haven Hospital, New Haven, CT<br />

Background: Brachytherapy as an alternative to whole-breast irradiation<br />

(WBI) for early-stage breast cancer has disseminated into clinical practice;<br />

however, current national treatment patterns and associated complications<br />

remain unknown. Methods: We constructed a national sample <strong>of</strong> Medicare<br />

beneficiares aged 66 to 94 who underwent breast conserving surgery in<br />

2008-2009 and who were treated with brachytherapy or WBI. We used<br />

hospital referral regions to assess national treatment variation and an<br />

instrumental variable analysis to compare complication rates between<br />

treatment groups, adjusting for patient and clinical characteristics such as<br />

age, number <strong>of</strong> comorbidities, receipt <strong>of</strong> chemotherapy or screening<br />

mammogram, and type <strong>of</strong> radiation facility. We compared one-year overall,<br />

wound and skin, and deep tissue and bone complications between<br />

brachytherapy and WBI using specific procedure and diagnosis codes<br />

identified in Medicare claims. Results: Of the 29,648 women in our sample,<br />

4,671 (15.8%) received brachytherapy. The median percent <strong>of</strong> patients<br />

receiving brachytherapy varied substantially across hospital referral regions<br />

(interquartile range: 7.5%-23.3%). In the bivariate analysis, 34.3% <strong>of</strong><br />

women treated with brachytherapy had a complication compared to 27.3%<br />

<strong>of</strong> those who received WBI (P�0.001). After adjusting for patient and<br />

clinical characteristics, 35.3% (95% CI: 34.7, 35.8) <strong>of</strong> women treated<br />

with brachytherapy had a complication compared to 18.7% (95% CI:<br />

18.2,19.2) treated with WBI (average predicted difference: 16.5%, 95%<br />

CI: 15.8, 17.3, P�0.001). While brachytherapy was associated with a<br />

16.9% (95% CI: 10.0, 23.8, P�0.001) higher absolute percentage <strong>of</strong><br />

wound and skin complications compared to WBI, there was no difference in<br />

deep tissue and bone complications. Conclusions: Brachytherapy is commonly<br />

used among Medicare beneficiaries; in some regions nearly one in<br />

four women who underwent adjuvant radiation received brachytherapy.<br />

After one year, wound and skin complications were significantly more<br />

common among women who received brachytherapy compared to those<br />

receiving WBI, but there was no difference in deep tissue and bone<br />

complications.<br />

1029 Poster Discussion Session (Board #21), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Breast cancer multifocality-multicentricity and locoregional recurrance.<br />

Presenting Author: Xiudong Lei, University <strong>of</strong> Texas M. D. Anderson Cancer<br />

Center, Houston, TX<br />

Background: The impact <strong>of</strong> multifocality (MF) and multicentricity (MC) on<br />

locoregional (LR) control for invasive breast cancer, and the optimal local<br />

treatment strategy for these tumors, is unknown. In particular, there is<br />

disagreement in the literature regarding the use <strong>of</strong> Breast Conservation<br />

Therapy (BCT). We evaluated a large single institution cohort <strong>of</strong> MF and MC<br />

breast cancers to determine if they had inferior LR control rate when<br />

compared to their unifocal counterparts. Methods: MF and MC were defined<br />

pathologically as more than one lesion in the same quadrant and more than<br />

one lesion in separate quadrants, respectively. Patients were categorized by<br />

presence or absence <strong>of</strong> MF or MC disease and by the LR treatment modality<br />

received – BCT (n�256), mastectomy alone (n�466), or mastectomy plus<br />

post-mastectomy radiation therapy (n�184). 10 patients who underwent<br />

BCT for MC disease against physician advice were excluded. MF and MC<br />

tumors were analyzed both as a group and as separate entities. Kaplan-<br />

Meier product limit method was used to calculate 5-year LR control rate.<br />

Cox proportional hazards models were fit to determine independent<br />

associations <strong>of</strong> MF/MC disease with LR control. Results: Median follow up<br />

was 52 months. Out <strong>of</strong> 3722 patients with stage I-III disease who did not<br />

receive neoadjuvant chemotherapy, 906 (24%) had MF (n�673) or MC<br />

(n�233) disease. 5-year rate <strong>of</strong> LR control rate was 99% in the MF group,<br />

96% in the MC group, and 98% in the unifocal group, (P � 0.44). Subset<br />

analysis revealed no statistical difference in LR control regardless <strong>of</strong> the<br />

type <strong>of</strong> LR treatment, (P � 0.67 in the BCT group, P � 0.37 in the<br />

mastectomy alone group, and P � 0.29 in the mastectomy plus postmastectomy<br />

radiation therapy group). There were 21 in-breast recurrences<br />

after BCT (8.2%). After controlling for other risk factors, MF and MC did not<br />

have an independent impact on LR control rate. Conclusions: MF and MC<br />

disease are not independent risk factors for LR recurrence. Patients with<br />

MF and MC breast cancer had similar rates <strong>of</strong> LR control to their unifocal<br />

counterparts, regardless <strong>of</strong> LR treatment modality. Our data suggest that<br />

BCT is a safe option for patients with MF tumors and that MF or MC disease<br />

alone is not an indication for post-mastectomy radiation therapy.<br />

1031 Poster Discussion Session (Board #23), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Trends in the use <strong>of</strong> breast-conserving surgery and adjuvant radiation<br />

therapy in patients with DCIS: A U.S. population-based analysis from 1996<br />

to 2007. Presenting Author: Jennifer Nishimura, Case Western Reserve<br />

School <strong>of</strong> Medicine, Cleveland, OH<br />

Background: The treatment for patients with DCIS remains controversial.<br />

Current guidelines based upon best available evidence suggest that breast<br />

conserving surgery (BCS) followed by adjuvant radiation therapy (RT) result<br />

in acceptable local control and breast cancer specific survival. The purpose<br />

<strong>of</strong> this study was to analyze trends in patterns <strong>of</strong> care as well as identify<br />

factors associated with surgery type and use <strong>of</strong> adjuvant radiation therapy<br />

in a select cohort <strong>of</strong> patients enrolled into the SEER database. Methods: The<br />

study included females 18 years and older with focal DCIS and known<br />

tumor size <strong>of</strong> 5 cm or less diagnosed between 1996 and 2007. The<br />

Cochran-Armitage trend test was applied to identify trends in the use <strong>of</strong><br />

BCS and RT over time. Multivariate logistic regression analyses were used<br />

to determine factors associated with receiving BCS vs. mastectomy and<br />

BCS plus RT vs. BCS alone. Cox proportional hazard model was used to<br />

determine associations with breast cancer-specific mortality. Results: Of<br />

the 34,233 women with DCIS, 76.59% were treated with BCS. 66.36% <strong>of</strong><br />

BCS patients received adjuvant RT over the study period. The proportion <strong>of</strong><br />

women receiving BCS increased from 71.5% in 1996 to 76.9% in 2007<br />

(p�0.0001). Additionally, the proportion <strong>of</strong> women who underwent BCS<br />

and received adjuvant radiation therapy over the same time period<br />

increased from 55.3% to 69.7% (p�0.0001). Multivariate analysis<br />

demonstrated that year <strong>of</strong> diagnosis, race, marital status, geographic<br />

region, tumor size, tumor grade and comedo necrosis all were significantly<br />

associated with the use <strong>of</strong> adjuvant radiation therapy, but age was not. Cox<br />

proportional hazards models did not associate either surgery type or use <strong>of</strong><br />

adjuvant radiation in patients undergoing BCS with breast cancer-specific<br />

mortality. Conclusions: Based upon reporting within the SEER database,<br />

the proportion <strong>of</strong> DCIS patients undergoing BCS and the BCS patients<br />

receiving adjuvant radiation increased over the study time period. Surgery<br />

type and use <strong>of</strong> adjuvant radiation therapy in patients with BCS was not<br />

associated with decreased risk <strong>of</strong> breast-cancer specific death in this<br />

cohort.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


1032 Poster Discussion Session (Board #24), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Impact <strong>of</strong> surgery and radiation <strong>of</strong> the primary among women with de novo<br />

stage IV breast cancer. Presenting Author: Shaheenah S. Dawood, Dubai<br />

Hospital, Dubai, United Arab Emirates<br />

Background: The aim <strong>of</strong> this retrospective study was to determine the<br />

impact <strong>of</strong> surgery(S) and radiation(R) therapy to the primary tumor among<br />

patients (pts) with stage IV denovo breast cancer. Methods: The SEER<br />

registry was used to identify pts with denovo stageIV breast cancer<br />

diagnosed between 1988 and 2008. Pts were divided into 4 groups based<br />

on type <strong>of</strong> treatment to primary tumor: both S�R, S alone, R alone, or no<br />

treatment <strong>of</strong> primary (no S/R). Breast cancer specific survival (BCS) was<br />

calculated from the date <strong>of</strong> diagnosis <strong>of</strong> breast cancer to the date <strong>of</strong> death<br />

from breast cancer or last follow up. Survival outcomes were estimated by<br />

the Kaplan-Meier method, and Cox models were fit to determine the<br />

association between treatment <strong>of</strong> primary and survival after adjusting for<br />

potential confounders (e.g age, grade, hormone receptor and race). Results:<br />

25903 pts were identified; 4640 (17.9%) S�R, 6556 (25.3%) S, 4467<br />

(17.2%) R, and 10240 (39.5%) no S/R. 1183 (4.6%) had surgery to sites<br />

other than the primary. Median age was 63 years. Median follow-up was 14<br />

months. Median BCS was 23 months. Median BCS among pts who<br />

underwent S�R, S, R and no S/R was 36 months, 31 months, 18 months<br />

and 15 months respectively (p�0.0001). Among pts who underwent S�R,<br />

median BCS among pts who did and did not have surgery to sites other than<br />

primary was 50 months and 41 months respectively (p�0.029). Of the pts<br />

treated with S�R 10-year BCS was 18%. In the multivariable model<br />

compared to women who were in the no S/R group those who underwent S<br />

(HR� 0.59, 95%CI 0.55- 0.62,p�0.0001) and S�R (HR�0.51, 95%CI<br />

0.47-0.55,p�0.0001) had decreased risk <strong>of</strong> death from breast cancer and<br />

those who underwent R (HR�1.13, 95% CI 1.04-1.21, p�0.002) had an<br />

increased risk <strong>of</strong> death from breast cancer. Pts who had surgery to sites<br />

other than the primary tumor had decreased risk <strong>of</strong> death from breast<br />

cancer compared to those who did not (HR�0.80, 95%CI 0.72-<br />

0.89,p�0.0001). Conclusions: Our results indicate that S�R <strong>of</strong> the<br />

primary breast tumor among pts with denovo stage IV breast cancer maybe<br />

associated with a decreased risk <strong>of</strong> death from breast cancer. A select<br />

subgroup <strong>of</strong> pts who undergo S�R may also benefit from surgery to sites<br />

other than the primary which may afford them maximum survival advantage.<br />

1034 General Poster Session (Board #19A), Sat, 8:00 AM-12:00 PM<br />

Impact <strong>of</strong> adjuvant chemotherapy on recurrence-free survival in patients<br />

with pT1a/b hormone-negative and HER2-positive breast cancer. Presenting<br />

Author: Yazan Migdady, Memorial Hospital <strong>of</strong> Rhode Island, Pawtucket,<br />

RI<br />

Background: T1ab triple-negative (TN) or Her-2-positive (H2�) breast<br />

cancers are reported to pose relatively high risk <strong>of</strong> relapse, but benefits <strong>of</strong><br />

adjuvant chemotherapy are uncertain. We studied the impact <strong>of</strong> chemotherapy<br />

on recurrence-free survival in this group. Methods: Records <strong>of</strong> all<br />

consecutive cases diagnosed in Brown-affiliated centers in 2000 - 2010<br />

were reviewed. Factors influencing chemotherapy decision were studied<br />

with logistic regression, and recurrence-free interval (RFI) with a Cox<br />

proportional hazard model and Kaplan-Meier estimator. Results: Among<br />

1415 screened T1a/b N0 cases, 161 were eligible (57 TN; 104 HER2�),<br />

with a median age <strong>of</strong> 57 years; 66% tumors were T1b. 20% <strong>of</strong> patients<br />

underwent mastectomy, 76% received radiation and 30% hormonal<br />

therapy. Adjuvant chemotherapy was recommended in 53% <strong>of</strong> cases.<br />

Younger age (p�10-6 ), stage T1b (p�10-5 ), high grade (p�0.001),<br />

HER2�/ERPR- status (p�0.017) and diagnosis after 2006 (p�0.007)<br />

were significantly predictive <strong>of</strong> the medical oncology recommendation.<br />

There was a significant trend with decrease in anthracycline (p�0.001)<br />

and increase in taxane use (p�0.001). With a median follow up <strong>of</strong> 46<br />

months, the 5-year rate <strong>of</strong> relapse was 6.1% (95%CI 2.7-13.9%),<br />

somewhat higher in T1b tumors (8.1%) and without detectable difference<br />

in TN/HER2� subgroups. In a univariate analysis chemotherapy did not<br />

significantly impact the recurrence-free interval (HR�0.45; 95%CI 0.09-<br />

2.34; p�0.32), however there was a detectable benefit (p�0.02) for T1b<br />

tumors in a multivariable Cox model including age (p�0.02) and LVI<br />

(p�0.01). The histology, type <strong>of</strong> surgery and year <strong>of</strong> diagnosis were not<br />

significant. There were no relapses among ER/PR� patients who received<br />

hormonal therapy or HER2� patients who received trastuzumab.<br />

Conclusions: The risk <strong>of</strong> relapse in biologically aggressive T1ab breast<br />

cancers is very low with judicious use <strong>of</strong> adjuvant therapy. The benefit <strong>of</strong><br />

chemotherapy is likely restricted to the highest-risk patients with T1b<br />

tumors, lymphovascular invasion and younger age.<br />

Breast Cancer—Triple-Negative/Cytotoxics/Local Therapy<br />

57s<br />

1033 Poster Discussion Session (Board #25), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Utilization <strong>of</strong> post-lumpectomy radiation therapy in women 70 years <strong>of</strong> age<br />

or older: A report from the National Cancer Data Base. Presenting Author:<br />

Katharine Yao, NorthShore University HealthSystem, Evanston, IL<br />

Background: The Cancer and Leukemia Group B (CALGB) 9343 trial<br />

published in 2004 showed no overall survival benefit from radiation in<br />

patients �70 years old with estrogen receptor (ER) positive, pT1 tumors<br />

with the use <strong>of</strong> tamoxifen. We tested the hypothesis that the use <strong>of</strong><br />

radiotherapy decreased in this group <strong>of</strong> patients following publication <strong>of</strong> the<br />

trial, utilizing the National Cancer Data Base. Methods: 34,853 breast<br />

cancer patients 70 years or older with pT1N0/NX, ER positive tumors who<br />

underwent a lumpectomy between 2004 and 2007 were studied. Chisquare<br />

tests and logistic regression models were used to determine trends<br />

and factors related to the use <strong>of</strong> radiation. Results: The use <strong>of</strong> radiation<br />

decreased from 70.6% in 2004 to 66.4% in 2005, 66.6% in 2006, and<br />

67.2% in 2007 (p�0.001). The use <strong>of</strong> standard external beam radiation<br />

decreased from 58.8% in 2004 to 45.8% in 2007 while the use <strong>of</strong><br />

accelerated partial breast radiation using brachytherapy (APBI) increased<br />

from 4.5% to 10.0%, IMRT radiation from 3.1% to 5.3%, and 3D<br />

conformal radiation from 3.7% to 5.7% (p�0.001). Patients between the<br />

ages <strong>of</strong> 86� years old were less likely to undergo radiation than patients<br />

70-75 years old (OR�0.12, 95% CI: 0.11-0.13). Asian Pacific Islanders<br />

were more likely to undergo radiation than whites (OR�1.39, 95% CI:<br />

1.13-1.70). In community cancer programs, 67% patients received<br />

radiation, compared to 69.1% in comprehensive community programs and<br />

65.5% in academic programs (p�0.001). The use <strong>of</strong> radiation varied by<br />

facility location; 73.5% <strong>of</strong> facilities located in the Midwest radiated these<br />

patients as opposed to 62.6% in the South. In patients who had no nodes<br />

examined, 37% underwent radiation as opposed to 74% who did have<br />

nodes examined (p�0.001). Likewise, 79.4% <strong>of</strong> patients who received<br />

hormone therapy underwent radiation as opposed to 54.6% <strong>of</strong> patients who<br />

did not receive hormonal therapy (p�0.001). Conclusions: The use <strong>of</strong><br />

radiation therapy decreased only slightly and remained high in women with<br />

ER� stage I breast cancer over the age <strong>of</strong> 70, despite findings from the<br />

CALGB 9343 study. However, there was a large shift in radiation modality<br />

over the study period in the older patients.<br />

1035 General Poster Session (Board #19B), Sat, 8:00 AM-12:00 PM<br />

Pathologic complete response rates observed in women with locally<br />

advanced and inflammatory breast cancer receiving neoadjuvant carboplatin<br />

and paclitaxel. Presenting Author: Arvind Manohar Shinde, City <strong>of</strong><br />

Hope, Duarte, CA<br />

Background: Pathologic complete response (pCR) following neoadjuvant<br />

chemotherapy (NCT) is predictive <strong>of</strong> outcome in patients with locally<br />

advanced breast cancer (LABC). A non-anthracycline containing NCT<br />

regimen (Sikov et al. JCO 10/09) may reduce the risk <strong>of</strong> associated<br />

secondary hematologic malignancies and cardiac toxicity while yielding<br />

comparable pCR rates. Methods: A retrospective review <strong>of</strong> all LABC and<br />

inflammatory breast cancer (IBC) cases treated from 4/09 to 12/11 with a<br />

NCT regimen <strong>of</strong> carboplatin (AUC <strong>of</strong> 6, administered on day 1) and<br />

paclitaxel 80 mg/m2 (given weekly on a 21-28 day cycle) was conducted at<br />

the City <strong>of</strong> Hope Cancer Center (COHCC). Pts with HER2� (HER�) tumors<br />

received trastuzumab during the NCT treatment. All pCRs (pCR <strong>of</strong> primary<br />

only – �pCR1°�; pCR <strong>of</strong> primary and lymph nodes – �pCR-All�) were<br />

determined by a COHCC pathologist based on final surgical specimens.<br />

Results: 38 pts were identified, with 39 breast primaries; 18% had IBC,<br />

62% <strong>of</strong> LABCs/IBCs were hormone receptor positive (HR�), 46% <strong>of</strong> tumors<br />

were HER2�, and 26% were triple negative. Median age was 51 [27-63].<br />

All pts completed the planned number <strong>of</strong> cycles. Four pts required<br />

carboplatin dose reductions, 4 pts required dose reductions in paclitaxel, 3<br />

pts had paclitaxel changed to nab-paclitaxel, and 17 pts required G-CSF to<br />

complete their planned treatment. One pt receiving trastuzumab experienced<br />

asymptomatic LVEF decline below normal limits. Conclusions: A<br />

non-anthracycline-containing NCT regimen <strong>of</strong> carboplatin/paclitaxel was<br />

well tolerated and resulted in high pCRs when given to triple negative<br />

(HER2-/HR-) pts, and HER2� pts, especially with HER2�HR- subtypes.<br />

The findings warrant further studies <strong>of</strong> this regimen in a prospective<br />

randomized setting.<br />

HER2 status N pCR1° (%) pCR-All (%) HR status N pCR1° (%) pCR-All (%)<br />

HER2� BC 18 56 44 HR� 13 38 31<br />

HR- 5 100 80<br />

HER2- BC 21 38 29 HR� 11 9 0<br />

HR- 10 70 60<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


58s Breast Cancer—Triple-Negative/Cytotoxics/Local Therapy<br />

1036 General Poster Session (Board #19C), Sat, 8:00 AM-12:00 PM<br />

A phase II study <strong>of</strong> foretinib in triple-negative, recurrent/metastatic breast<br />

cancer: NCIC CTG trial IND.197 (NCT01147484). Presenting Author:<br />

Daniel Rayson, QEII Health Sciences Centre, Halifax, NS, Canada<br />

Background: Met, a receptor tyrosine kinase, is preferentially expressed in<br />

basal-like compared to luminal breast cancer. In murine models, overexpression<br />

<strong>of</strong> the oncogenic Met receptor transgene induces tumors with human<br />

basal gene expression characteristics supporting Met inhibition as a<br />

treatment strategy for triple negative (TN) breast cancer. Foretinib is an oral<br />

multi-kinase inhibitor <strong>of</strong> Met, RON, AXL, TIE-2 and VEGF receptors with<br />

anti-tumor activity in advanced HCC and papillary renal cell cancer.<br />

Methods: Patients (pts) with TN breast cancer and 0-1 prior regimens for<br />

metastatic disease received daily foretinib 60 mg po in a 2-stage single arm<br />

trial. Primary endpoints were objective response and early progression rates<br />

per RECIST 1.1. Tumor samples were centrally reviewed to confirm<br />

ER/PR/HER2 status and for correlative studies including Met, PTEN and<br />

EGFR expression. Stage 1 accrual required 23 response-evaluable Met<br />

unselected patients with accrual continuing if �/� 1 response or � 17<br />

early progressions (PD �� 8 weeks on study) were observed. Results:<br />

Accrual is 29 pts to date; 24 are eligible, 22 evaluable for toxicity and 15<br />

for response. Median age is 56 y (43-81), ECOG PS 0-1 in 23/24. Grade 3<br />

laboratory adverse events were: lymphopenia (9%), elevations in ALT (5%),<br />

GGT (5%) and INR (5%). Treatment-related non-hematologic toxicities<br />

included (all/grade 3-4) fatigue (64%/5%), nausea (55%/5%), diarrhea<br />

(41%/5%), hypertension (32%/14%), vomiting (27%/0%), anorexia (23%/<br />

5%) and rash (14%/0%). Three SAEs possibly related to foretinib included;<br />

asymptomatic pulmonary embolism, reversible CHF and pleural effusion<br />

with QTc prolongation. One PR (7%), 8 early PD (53%) and 6 SD (40%)<br />

have been observed to date with median SD duration <strong>of</strong> 5.4 months (range<br />

2.7-5.5). Preliminary correlative results (IHC): 5/8 (62.5%) evaluable Met<br />

positive cases had SD and 4/5 (80%) Met negative cases had PD as best<br />

response. Met IHC was negative in the pt with PR. Conclusions: Foretinib<br />

shows preliminary evidence <strong>of</strong> activity and tolerability in metastatic, TN<br />

breast cancer. Stage 2 <strong>of</strong> accrual will include 15 pts with pre-treatment<br />

biopsies <strong>of</strong> metastases and circulating tumor cell collection.<br />

1038 General Poster Session (Board #19E), Sat, 8:00 AM-12:00 PM<br />

Single institution experience with neoadjuvant chemotherapy for metaplastic<br />

breast cancer (MBC). Presenting Author: Anna Kaminsky, University <strong>of</strong><br />

Pittsburgh Medical Center (UPMC), Pittsburgh, PA<br />

Background: Metaplastic breast carcinoma (MBC) is a rare subtype that<br />

accounts for �1% <strong>of</strong> all breast carcinomas. MBC is frequently triple<br />

negative and neoadjuvant chemotherapy (NAC) is <strong>of</strong>ten used in triple<br />

negative breast cancer (TNBC). The objective <strong>of</strong> this analysis is to ascertain<br />

response rates <strong>of</strong> MBC to NAC as compared to non-metaplastic TNBC.<br />

Methods: We searched the Magee Women’s Cancer Center <strong>of</strong> UPMC<br />

IRB-approved neo-adjuvant treatment database which contains outcome<br />

data on 594 patients treated from 2004-2010. 116 patients with triple<br />

negative breast cancer (ER /PR negative or ER /PR weakly positive (H score<br />

<strong>of</strong> 10 or less) and HER2 negative or indeterminate (HER2 1� or 2�<br />

without amplification by FISH)), were identified. Nine <strong>of</strong> these TNBCs had<br />

metaplastic subtype and 2 groups were analyzed: metaplastic breast<br />

carcinoma (MBC) (N� 9) and non-metaplastic breast carcinoma (NMBC)<br />

(N � 107). Tumor volume reduction (TVR), pathologic complete response<br />

(pCR), recurrence and mortality were compared in both groups. Results:<br />

Mean follow up in MBC group was 43 months and no patients were lost to<br />

follow up. Mean tumor size on presentation in MBC group was 4.47 cm<br />

while in NMBC group it was 3.33 cm. pCR was noted in 0/9 MBC and<br />

43/107 NMBC cases (p � 0.0253). 6/9 patients had mastectomy, 2/9 had<br />

breast conserving surgery (BCS) and 1/9 patients did not have a surgery yet.<br />

Average TVR was 28% in MBC cases compared to 74% in NMBCs when<br />

cases with pCR were included (p � 0.0001) and 56% when cases with pCR<br />

were excluded (p � 0.0202). Follow up on 9 MBC cases revealed 1<br />

recurrence and subsequent death (11%). Follow up on 64 NMBC patients<br />

who failed to achieve pCR revealed 22 recurrences (34%) and 18 <strong>of</strong> them<br />

subsequently died (28%).Follow up on 43 NMBC cases that achieved pCR<br />

revealed 3 recurrences (7%) and 1 death (2%). Conclusions: MBC was<br />

characterized by larger size at baseline as compared to NMBC. There were<br />

no pCR’s seen in MBC, but some MBC did achieve response that allowed for<br />

breast conservation. Although the average tumor volume reduction was<br />

significantly less in MBC compared to NMBC, the NMBC that failed to<br />

achieve pCR fared much worse than MBC who did not achieve pCR.<br />

Therefore, the triple negative paradox is likely not applicable to MBC.<br />

1037 General Poster Session (Board #19D), Sat, 8:00 AM-12:00 PM<br />

Comprehensive investigation <strong>of</strong> adverse event (AE)-related costs in patients<br />

with metastatic breast cancer (MBC) treated with first- and second-line<br />

chemotherapies. Presenting Author: Sara A. Hurvitz, UCLA, Los Angeles,<br />

CA<br />

Background: MBC is incurable and managed with ongoing therapy. This<br />

study examined the incremental costs <strong>of</strong> chemotherapy-associated AEs in<br />

MBC. Methods: The PharMetrics Integrated Database (2000-2010) was<br />

used to identify MBC pts treated either 1st or 2nd line with a taxane (T)<br />

(paclitaxel or docetaxel) or capecitabine (C)-based regimen for �30 days<br />

(defined as a treatment episode (TE)). Incremental costs attributable to<br />

AEs were assessed by comparing costs incurred during TEs with and<br />

without AEs. AEs were identified using medical claims with a diagnosis for<br />

�1 event <strong>of</strong> interest (e.g., infections, fatigue, anemia, neutropenia). Pt<br />

characteristics were balanced between comparison groups (with and w/o<br />

AEs) using inverse probability weighting method. Incremental monthly<br />

costs due to AEs were estimated during the TEs and included the following<br />

cost components: inpt (IP), outpt (OP), emergency room (ER), other<br />

medical service, pharmacy costs (chemotherapy and other drugs), and total<br />

healthcare costs. Statistical comparisons were conducted using Wilcoxon<br />

tests. Results: 3,222 women (mean age�57) received aTorCas1stor<br />

2nd-line therapy for MBC. Of the 2,678 1st-line pts, 69.7% received T and<br />

30.3% with C; average monthly total costs ranged from $9,159 to<br />

$10,298. AEs were commonly seen in pts treated with 1st-line T and C<br />

(94.6% and 83.7%). On average, the total monthly incremental cost<br />

associated with AEs was 38% higher ($3,547) for T and 9% higher ($854)<br />

for C. IP and other drug costs accounted for a majority <strong>of</strong> these costs. Of<br />

1,084 2nd-line pts, 66% received T and 34% C, with average monthly total<br />

costs ranging from $5,950 to $12,979. 94.4% <strong>of</strong> T pts and 84% <strong>of</strong> C pts in<br />

the 2nd-line had an AE. The average total monthly incremental cost<br />

associated with AEs for T was $5,320 and $4,933 for C (69.5% and<br />

82.9% higher vs pts w/o AEs). Pharmacy costs accounted for a majority <strong>of</strong><br />

increased costs seen in pts with AEs treated with T; IP and OP accounted<br />

for a majority <strong>of</strong> these costs in pts treated with C. Conclusions: This is the 1st study assessing costs associated with AEs for tx <strong>of</strong> mBC. AEs are associated<br />

with a substantial economic burden that is mainly explained by increased<br />

IP, OP, and pharmacy costs.<br />

1039 General Poster Session (Board #19F), Sat, 8:00 AM-12:00 PM<br />

Association between vitamin D deficiency and breast cancer histology: A<br />

retrospective database review. Presenting Author: Ranjana S. Bhargava,<br />

University <strong>of</strong> Maryland Marlene and Stewart Greenebaum Cancer Center,<br />

Baltimore, MD<br />

Background: Vitamin D (Vit D) deficiency has been shown to be associated<br />

with a higher risk <strong>of</strong> developing breast cancer. Inverse association has also<br />

been shown between vit D level and tumor size. However, the association<br />

between vit D deficiency and breast cancer histology remains unclear.<br />

Preclinical data has suggested that vit D plays an essential role in the<br />

terminal differentiation <strong>of</strong> breast cells. Thus, we hypothesize that vit D<br />

deficiency would be associated with estrogen receptor-negative tumors,<br />

particularly triple-negative breast cancers (TNBC) which are associated<br />

with aggressive clinical course. Methods: We conducted a retrospective<br />

database review to obtain information including age, race, tumor histology,<br />

size, stage, and vit D levels in newly diagnosed breast cancer patients at<br />

University <strong>of</strong> Maryland Greenebaum Cancer Center. Results: 150 patients<br />

presented between July 2008 and August 2011 were included in this<br />

analysis. Average age at diagnosis was 57 (range 30-87), and 56% <strong>of</strong><br />

patients were African <strong>American</strong>. Overall, 80% <strong>of</strong> the patients were vit D<br />

deficient at diagnosis, with levels � 30 ng/ml. African-<strong>American</strong> patients<br />

were more likely to be severely vit D deficient with levels � 10 ng/ml<br />

compared to Caucasians (33% vs. 7.5%, p� 0.0002). Patients with TNBC<br />

were more likely to be vit D deficient at diagnosis compared to hormone<br />

receptor-positive patients (93% vs. 76%, p �0.015). These patients also<br />

had the lowest mean (18) and median (16) <strong>of</strong> vit D levels compared to all<br />

other patients. This difference is also statistically significant in multivariate<br />

analysis when adjusted for age, race, and stage (OR 3.96; p�0.04).<br />

Regardless <strong>of</strong> the ER/PR status, Her 2 negative patients were more vit D<br />

deficient compared to Her 2 positive patients (86% vs. 61%, p�0.001).<br />

Unlike previous studies, no correlation was seen between tumor size and vit<br />

D levels. Furthermore, there is also no association between stage, nodal<br />

involvement, or Ki67 and vit D level. Conclusions: Vit D deficiency is<br />

common among patients with newly-diagnosed breast cancer, particularly<br />

African <strong>American</strong> patients. Patients with TNBC have a significantly higher<br />

likelihood <strong>of</strong> being vit D deficient than patients with other histological<br />

subtypes.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


1040 General Poster Session (Board #19G), Sat, 8:00 AM-12:00 PM<br />

Frequency <strong>of</strong> TLE3 overexpression in breast carcinoma subtypes including<br />

a large cohort <strong>of</strong> triple-negative patients. Presenting Author: Gargi D. Basu,<br />

Caris Life Sciences, Phoenix, AZ<br />

Background: The taxanes are an important class <strong>of</strong> agents for the treatment<br />

<strong>of</strong> a broad range <strong>of</strong> malignancies including breast cancer. They improve<br />

survival in patients with early stage and metastatic breast cancer. Transducin-like<br />

enhancer <strong>of</strong> split 3 (TLE3) is a transcriptional repressor which<br />

influences growth and microtubule stability and its expression has been<br />

implicated in response to taxane therapy in breast cancer. We investigated<br />

the tumor expression <strong>of</strong> TLE3 in breast cancer patients, including a large<br />

cohort <strong>of</strong> the triple negative subtype. Methods: We analyzed TLE3 (M-201),<br />

ER(1D5), PR(PgR636) and HER2/neu(Polyclonal) expression by immunohistochemistry<br />

in 978 breast cancer patients. Immunoreactivity was<br />

assessed by scoring the percentage <strong>of</strong> cells stained in each field and by the<br />

intensity <strong>of</strong> staining. Results: To sub-classify the 978 breast cancer<br />

patients, we utilized hormone receptors (ER and PR) and HER2 expression/<br />

amplification. Overall, 36% <strong>of</strong> the total breast cancer patients were<br />

hormone receptor positive, 15% were HER2 positive and 49% were triple<br />

negative. The percentage <strong>of</strong> triple negative patients was higher in our<br />

cohort, given the fact that molecular pr<strong>of</strong>iling services are used more<br />

frequently for this subtype. A total <strong>of</strong> 477 patients were triple negative <strong>of</strong><br />

which 61% stained positive for TLE3 expression. Of the 150 HER2 positive<br />

patients, 73% stained positive for TLE3 expression as compared with 82%<br />

TLE3 positivity in the 351 hormone receptor positive patients. By pairwise<br />

comparison, the hormone receptor positive vs triple-negative subtype<br />

showed the highest statistical significance in ratios <strong>of</strong> TLE3 positives (p<br />

�2.5e-10). Conclusions: Our results show that TLE3 is over-expressed in<br />

the majority <strong>of</strong> HER2 positive and hormone receptor positive breast cancer<br />

patients. Interestingly, the frequency <strong>of</strong> over-expression <strong>of</strong> TLE3 was lowest<br />

in the triple negative subtype thereby making it more important to identify<br />

those patients in this group who are most likely to respond to taxanes prior<br />

to therapy. To our knowledge, this is the first study providing a comprehensive<br />

review <strong>of</strong> TLE expression in breast cancer subtypes.<br />

1042 General Poster Session (Board #20A), Sat, 8:00 AM-12:00 PM<br />

Preclinical evaluation <strong>of</strong> PARP inhibition in breast cancer: Comparative<br />

effectiveness <strong>of</strong> olaparib and iniparib. Presenting Author: Maura B. Cotter,<br />

School <strong>of</strong> Medicine and Medical Science, University College Dublin,<br />

Dublin, Ireland<br />

Background: The main function <strong>of</strong> PARP1 is repair <strong>of</strong> single-strand DNA.<br />

Phase I/II clinical trials have shown that the PARP inhibitor, olaparib has<br />

efficacy in BRCA1/2-related breast cancer. Due to the similarities between<br />

BRCA1/2-associated and triple negative breast cancer (TNBC), we hypothesise<br />

that TNBC may also be sensitive to PARP inhibition. In order to assess<br />

this we addressed the effects <strong>of</strong> 2 PARP/PARP-like inhibitors, on a panel <strong>of</strong><br />

breast cancer cell lines. Methods: PARP1 was measured by immunohistochemistry<br />

in 101 TNBC and 116 non-TN cancers. Comparative growth<br />

inhibitory capacity <strong>of</strong> olaparib and iniparib was evaluated using cell<br />

viability (MTT) and colony formation assays in 12 breast cancer cell lines<br />

(TN�7, non-TN�5). Results: Using immunohistochemistry, PARP1 staining<br />

was predominantly nuclear with some cytoplasmic staining. High<br />

staining intensity for PARP1 was found more frequently in ER-negative (p<br />

� 0.001), in high grade (p � 0.013) and in Ki67-positive ( p � 0.003)<br />

samples. Potentially important was the finding that high PARP1 staining<br />

intensity was detected more frequently in TN than non-TN samples (p �<br />

0.0001). IC50 concentrations across 12 cell lines ranged from 3.7-31 �M<br />

for olaparib and 13-70 �M for iniparib. No difference in sensitivity was<br />

observed between the TN and non-TN cell lines (by MTT). Olaparib also<br />

reduced the ability <strong>of</strong> cells to form colonies with IC50 values ranging from<br />

�0.01-2.5 �M. Addition <strong>of</strong> the CDKI inhibitor CDK1i (Calbiochem) to<br />

olaparib resulted in formation <strong>of</strong> significantly fewer colonies compared with<br />

either inhibitor alone, in a cell line dependent manner. Conclusions: Our<br />

results suggest that although PARP1 is expressed in the majority <strong>of</strong> breast<br />

cancer, significantly higher staining intensity was found in TN than non-TN<br />

samples. Furthermore, our work suggests that olaparib is a more potent<br />

inhibitor <strong>of</strong> the in vitro growth <strong>of</strong> breast cancer cells than iniparib.<br />

Combined inhibition <strong>of</strong> PARP1 with olaparib and CDK1 with CDK1i may be<br />

a way forward for the treatment <strong>of</strong> TNBC. Acknowledgement: The authors<br />

thank SFI (SRC award, 08/SRC/B1410 MTCI) for funding this work.<br />

Breast Cancer—Triple-Negative/Cytotoxics/Local Therapy<br />

59s<br />

1041 General Poster Session (Board #19H), Sat, 8:00 AM-12:00 PM<br />

Epigenetic aspects <strong>of</strong> triple-negative in patients with breast cancer.<br />

Presenting Author: Joaquina Martínez-Galan, Medical Oncology Department,<br />

Hospital Universitario Virgen de las Nieves, Granada, Spain<br />

Background: Identification <strong>of</strong> gene expression-based breast cancer subtypes<br />

is considered a critical means <strong>of</strong> prognostication. Genetic mutations<br />

along with epigenetic alterations contribute to gene-expression changes<br />

occurring in breast cancer. However, the reproducibility <strong>of</strong> differential DNA<br />

methylation discoveries for cancer and the relationship between DNA<br />

methylation and aberrant gene expression have not been systematically<br />

analysed. The present study was undertaken to dissect the breast cancer<br />

methylome and to deliver specific epigenotypes associated with particular<br />

breast cancer subtypes. Methods: By using Real Time QMSPCR SYBR green<br />

we analyzed DNA methylation in regulatory regions <strong>of</strong> 107 pts with breast<br />

cancer and analyzed association with prognostics factor in triple negative<br />

breast cancer and methylation promoter ESR1, APC, E-Cadherin, Rar B<br />

and 14-3-3 sigma. Results: We identified novel subtype-specific epigenotypes<br />

that clearly demonstrate the differences in the methylation pr<strong>of</strong>iles <strong>of</strong><br />

basal-like and human epidermal growth factor 2 (HER2)-overexpressing<br />

tumors. Of the cases, 37pts (40%) were Luminal A (LA), 32pts (33%)<br />

Luminal B (LB), 14pts (15%) Triple-negative (TN), and 9pts (10%)<br />

HER2�. DNA hypermethylation was highly inversely correlated with the<br />

down-regulation <strong>of</strong> gene expression. Methylation <strong>of</strong> this panel <strong>of</strong> promoter<br />

was found more frequently in triple negative and HER2 phenotype. ESR1<br />

was preferably associated with TN(80%) and HER2�(60%) subtype. With<br />

a median follow up <strong>of</strong> 6 years, we found worse overall survival (OS) with<br />

more frequent ESR1 methylation gene(p�0.05), Luminal A;ESR1 Methylation<br />

OS at 5 years 81% vs 93% when was ESR1 Unmethylation. Luminal<br />

B;ESR1 Methylation 86% SG at 5 years vs 92% in Unmethylation ESR1.<br />

Triple negative;ESR1 Methylation SG at 5 years 75% vs 80% in unmethylation<br />

ESR1. HER2;ESR1 Methylation SG at 5 years was 66.7% vs 75% in<br />

unmethylation ESR1. Conclusions: Our results provide evidence that<br />

well-defined DNA methylation pr<strong>of</strong>iles enable breast cancer subtype<br />

prediction and support the utilization <strong>of</strong> this biomarker for prognostication<br />

and therapeutic stratification <strong>of</strong> patients with breast cancer.<br />

1043 General Poster Session (Board #20B), Sat, 8:00 AM-12:00 PM<br />

Molecular characteristics and metastasis predictor genes <strong>of</strong> triple-negative<br />

breast cancer. Presenting Author: Wen-Hung Kuo, Department <strong>of</strong> Surgery,<br />

College <strong>of</strong> Medicine, National Taiwan University, Taipei, Taiwan<br />

Background: Triple-negative breast cancer(TNBC) is a subtype <strong>of</strong> breast<br />

cancer with aggressive tumor behavior and distinct disease etiology. Due to<br />

the lack <strong>of</strong> an effective targeted medicine, treatment options for triplenegative<br />

breast cancer are few and recurrence rates are high. Although<br />

various multi-gene prognostic markers have been proposed for the prediction<br />

<strong>of</strong> breast cancer outcome, most <strong>of</strong> them were proven clinically useful<br />

only for estrogen receptor-positive breast cancers. Reliable identification <strong>of</strong><br />

triple-negative patients with a favorable prognosis is not yet possible.<br />

Methods: Clinicopathological information and microarray data from 157<br />

invasive breast carcinomas were collected at National Taiwan University<br />

Hospital from 1995 to 2008. Gene expression data <strong>of</strong> 51 triple-negative<br />

and 106 luminal breast cancers were generated with oligonucleotide<br />

microarrays. A prognostic 45-gene signature for triple-negative breast<br />

cancer was identified using Student’s t test and receiver operating<br />

characteristic analysis. Results: Hierarchical clustering analysis revealed<br />

that the majority (94%) <strong>of</strong> triple-negative breast cancers were tightly<br />

clustered together carrying strong basal-like characteristics. A novel<br />

45-gene signature giving 98% predictive accuracy in distant metastasis<br />

recurrence was identified in our triple-negative patient cohort. External<br />

validation <strong>of</strong> the prognostic signature in an independent microarray dataset<br />

<strong>of</strong> 59 early-stage triple-negative patients also obtained statistical significance<br />

(hazard ratio 2.29, 95% CI 1.04-5.06, Cox P � 0.04), outperforming<br />

five other published breast cancer prognostic signatures. The prognostic<br />

signature was statistically predictive with the node-negative triple-negative<br />

patients in the validation cohort. Conclusions: The 45-gene prognostic<br />

signature identified in this study revealed that TGF-� signaling in immune/<br />

inflammatory regulation may be critically involved in distant metastatic<br />

invasion <strong>of</strong> TNBC. The 45-gene signature, if further validated, may be a<br />

clinically useful tool in risk assessment <strong>of</strong> metastasis recurrence for<br />

early-stage triple-negative patients.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


60s Breast Cancer—Triple-Negative/Cytotoxics/Local Therapy<br />

1044 General Poster Session (Board #20C), Sat, 8:00 AM-12:00 PM<br />

Association <strong>of</strong> body mass index and outcome in advanced breast cancer.<br />

Presenting Author: Claudia Andreetta, Department <strong>of</strong> Oncology, University<br />

Hospital <strong>of</strong> Udine, Udine, Italy<br />

Background: Obesity represents a well-known risk factor for the development<br />

<strong>of</strong> breast cancer and an adverse prognostic factor in early disease.<br />

Overweight is associated with reduced efficacy <strong>of</strong> aromatase inhibitors in<br />

adjuvant setting. Few data have been reported about the potential relationship<br />

<strong>of</strong> overweight and outcome in advanced breast cancer (ABC). Methods:<br />

We retrospectively evaluated body mass index (BMI) in a consecutive series<br />

<strong>of</strong> 400 ABC patients treated at our institution. BMI was calculated at<br />

baseline <strong>of</strong> diagnosis <strong>of</strong> ABC. We evaluated association <strong>of</strong> BMI and other<br />

prognostic and predictive markers with Progression Free Survival (PFS) and<br />

Overall Survival (OS). We evaluated PFS at first and subsequent lines <strong>of</strong><br />

chemotherapy (CT) and endocrine therapy (ET). Overweight patients were<br />

defined as having BMI � 25. Results: Overweight patients were 52%.<br />

Median age <strong>of</strong> the population was 58 years. Median OS was 33.7 months.<br />

Overall, 76% <strong>of</strong> patients presented with ER� and 17.7% with HER2�<br />

ABC.Overweight was associated with increased age at diagnosis, menopausal<br />

status and luminal B or triple negative immunophenotype. At<br />

multivariate analysis, BMI � 25 was associated with better PFS at first-line<br />

ET (HR� 0.68, 95% CI 0.46-0.99). BMI was not associated with OS.<br />

Conclusions: BMI at baseline does not seem to be an adverse prognostic<br />

factor for ABC patients. Overweight may be associated with better PFS in<br />

endocrine responsive ABC treated with ET, especially in first-line setting.<br />

The role <strong>of</strong> BMI in ABC deserves to be further investigated.<br />

1046 General Poster Session (Board #20E), Sat, 8:00 AM-12:00 PM<br />

Patterns in circulating microRNA in black (B) versus white (W) patients<br />

with triple-negative (TN) breast cancer (BC). Presenting Author: Iuliana<br />

Shapira, H<strong>of</strong>stra North Shore-LIJ School <strong>of</strong> Medicine, Lake Success, NY<br />

Background: Breast cancer is by far more common in W than in B women.<br />

Black women have more aggressive disease that occurs almost a decade<br />

earlier, it is usually triple negative and has a lower survival. Objectives to<br />

determine 1) if plasma miRNA expression differs between B and W women,<br />

and 2) if variation in miRs may explain the observed survival difference in<br />

TNBC in W compared to B women. Methods: We determined miRNA pr<strong>of</strong>iles<br />

in plasma collected before removal <strong>of</strong> breast tumors in three groups <strong>of</strong> W<br />

and B women: 1) normal controls (N), 2) TN and 2) ER/PR positive BC.<br />

Expression miRNA pr<strong>of</strong>iling <strong>of</strong> 754 miRNAs on the ABI Open Array<br />

detected 101miRNAs in plasma. We compared miRNA expression in<br />

cancer patients and race-matched controls. A moderated t-statistic through<br />

the R/Bioconductor ’limma’ was used to compare the mean response<br />

between subject factors <strong>of</strong> interest. Results with a p�0.05 were considered<br />

statistically significant. Characteristics: 32 patients were included in this<br />

analysis (mean age 50 years; range 31-68), 10 had stage III TNBC (5 B & 5<br />

W); 10 had stage III ER/PR�BC (5 B, 5 W); and 12 were controls (6 B, 6 W)<br />

Results: W TNBC patients overexpressed (15 fold higher than normal) 20<br />

miRs in plasma (let-7d, let7g, miR-103, -10a, 15a, -9, -99b, -181a, -18a,<br />

-502, -187, -365 and others), these miRs were not found in any <strong>of</strong> the B<br />

patients. Six microRNAs (such as miR-34a, -127 and others) were 15 fold<br />

higher than normal controls in B cancer patients, these miRs were not<br />

detected in W patients with TNBC. Four miRs in B and 8 miRs in W were not<br />

previously reported in association with breast cancer suggesting that they<br />

may be connected to the host response. Conclusions: The striking difference<br />

in the patterns <strong>of</strong> plasma miRNA expression between B and W<br />

patients may provide the key to the large difference in outcome between<br />

these groups when receiving similar treatments, the worse prognosis group<br />

may carry the miRs that promote metastasis. The seed-soil hypothesis may<br />

explain the better prognosis in W patients. W women may carry those<br />

miRNAs that support an “exaggerated” response to systemic treatment,<br />

while the B may have the miRs that favour metastasis.<br />

1045 General Poster Session (Board #20D), Sat, 8:00 AM-12:00 PM<br />

Neoadjuvant weekly nab-paclitaxel (wA), carboplatin (Cb) plus bevacizumab (B)<br />

with or without dose-dense doxorubicin-cyclophosphamide (ddAC) plus B in<br />

ER�/HER2-negative (HR�) and triple-negative (TN) breast cancer (BrCa): A<br />

BrUOG study. Presenting Author: Natalie Faye Sinclair, Department <strong>of</strong><br />

Medicine, Warren Alpert Medical School <strong>of</strong> Brown University, Providence,<br />

RI<br />

Background: High risk BrCa patients (pts) <strong>of</strong>ten receive weekly paclitaxel<br />

(wP) as well as ddAC. Switching to wA(Abraxane) or adding B or Cb may<br />

enhance its efficacy, especially in TN pts. Methods: Pts with clinical stage<br />

IIA-IIIC BrCa received wA 100 mg/m2 , Cb AUC 6 � B 15 mg/kg q3wks x 12<br />

wks only (cohort 1/Yale) or followed by ddAC � B x 4 (cohort 2/Brown).<br />

Endpoints: pathologic complete response (pCR) - absence <strong>of</strong> invasive BrCa<br />

in breast � axillary nodes, residual cancer burden (RCB), clinical CR/<br />

partial response (cCR/cPR), and toxicity. Correlative studies are being<br />

performed on biopsies obtained at baseline and after run-in doses <strong>of</strong> wA or<br />

B only. Post-op pts resume B for 34 wks; other systemic therapy, including<br />

ddAC in cohort 1, is at MD discretion. Results: 55 <strong>of</strong> 60 pts (median age 47,<br />

range 25-68; 31 HR�/29 TN) are evaluable for response (see table below).<br />

Median # doses wA 11,Cb 4, ddAC 4. Dose reductions: wA 25% for<br />

neutropenia (ANC), Cb 15% for thrombocytopenia (tcp). B 7% held for<br />

hypertension. Grade 3-4 toxicities (�5%): ANC 85%, tcp 35%, anemia<br />

25%. Serious adverse events during wA: 3 nausea/dehydration (N/D), 3<br />

infection w/o neutropenic fever (FN), 2 GI bleed; during ddAC: 6 (21%) FN<br />

despite G-CSF, 3 N/D. Conclusions: The combination <strong>of</strong> wA, q3wk Cb � B<br />

was well tolerated, with cCR�cPR 84%. However, overall pCR was only<br />

11% (27% in TN) after 12 wks <strong>of</strong> this regimen (cohort 1). Subsequent<br />

preop ddAC raised overall pCR to 54%, and 81% in TN, demonstrating that<br />

longer treatment duration or inclusion <strong>of</strong> anthracycline-based therapy<br />

improves responses. Results for cohort 2 compare favorably with those from<br />

I-SPY, GeparQuinto and NSABP B-40; the addition <strong>of</strong> Cb and/or B in TN is<br />

being evaluated in CALGB 40603.<br />

N<br />

cCR/cPR<br />

after wA<br />

cCR/cPR<br />

after ddAC pCR<br />

Total 55 13/33 (84%) 19 (35%) 29 (53%) 26<br />

HR � 28 5/17 (79%) 3 (11%) 7 (25%) 21<br />

TN 27 8/16 (89%) 16 (59%) 22 (82%) 5<br />

Cohort 1 27 4/18 (81%) 3 (11%) 10 (37%) 17<br />

HR� 16 2/10 (75%) 0 4 (25%) 12<br />

TN 11 2/8 (91%) 3 (27%) 6 (55%) 5<br />

Cohort 2 28 9/15 (86%) 13/11(92%)* 15 (54%) 18 (64%) 10<br />

HR � 12 3/7 (83%) 3/7 (91%) 2 (17%) 2 (17%) 10<br />

TN 16 6/8 (88%) 10/4 (93%) 13 (81%) 16 (100%) 0<br />

*2 pts not restaged after ddAC excluded.<br />

1047 General Poster Session (Board #20F), Sat, 8:00 AM-12:00 PM<br />

Molecular comparison <strong>of</strong> human triple-negative inflammatory breast cancer<br />

(TN-IBC) and human triple-negative noninflammatory breast cancer (TNnon-IBC).<br />

Presenting Author: Hiroko Masuda, Morgan Welch Inflammatory<br />

Breast Cancer Program and Clinic, University <strong>of</strong> Texas M. D. Anderson<br />

Cancer Center, Houston, TX<br />

Background: IBC has a poor prognosis because <strong>of</strong> its high rate <strong>of</strong> recurrence.<br />

There is an urgent need to define the biology <strong>of</strong> IBC to develop molecularbased<br />

targeted therapies for this disease. TNBC has a similar poor<br />

prognosis. Recently, Lehmann et al. (JCI, 2011) identified 7 subtypes <strong>of</strong><br />

TNBC: basal-like (BL) 1 and 2, immunomodulatory (IM), mesenchymal<br />

(M), mesenchymal stem-like (MSL), luminal androgen receptor (LAR), and<br />

“unknown.” In light <strong>of</strong> these findings, we hypothesized that the distribution<br />

<strong>of</strong> TNBC subtypes differs between TN-IBC and TN-non-IBC. Methods: We<br />

qualitatively reproduced the Lehmann et al. experiments using Affymetrix<br />

CEL files from the same datasets to ensure the reproducibility <strong>of</strong> their<br />

findings. We quantified all arrays using frozen robust multiarray analysis<br />

with a linear model adjustment to account for batches. Then validated the<br />

results in a TNBC cohort from the World IBC Consortium for which IBC<br />

status was known (41 cases <strong>of</strong> TN-IBC; 53 cases <strong>of</strong> TN-non-IBC). We used<br />

the Fisher exact test to determine associations between TNBC subtypes<br />

and IBC status. We used Kaplan-Meier curves and log-rank tests to<br />

compare clinical outcomes between TNBC subtypes. Results: We found the<br />

7 subtypes for both TN-IBC and TN-non-IBC. While the correspondence<br />

between our findings and those <strong>of</strong> Lehmann et al. was not perfect, there was<br />

a very significant correlation (P�2.2x10-16 ). We found no association<br />

between TNBC subtype and IBC status (P�.5023). As expected, we found<br />

that patients with IBC had significantly worse recurrence-free survival<br />

(RFS) than a comparison cohort <strong>of</strong> patients with advanced non-IBC that<br />

included not only patients with TNBC but also patients with ER-positive<br />

and HER2-amplified tumors (P�.0054). However, TNBC subtype did not<br />

predict RFS. IBC status was not a significant predictor <strong>of</strong> RFS or overall<br />

survival in the TNBC cohort. Conclusions: Both TN-IBC and TN-non-IBC are<br />

heterogeneous. TNBC subtypes are unrelated to IBC status. TN-IBC and<br />

TN-non-IBC have the same subtypes and clinical outcome.<br />

% IBC Non-IBC<br />

Relative M 9.7 13.2<br />

IM 7.3 22.6<br />

BL1 21.9 15.0<br />

BL2 7.3 3.7<br />

LAR 14.6 11.3<br />

Unk 19.5 18.8<br />

MSL 19.5 15.0<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.<br />

RCB<br />

0-1<br />

RCB<br />

2-3


1048 General Poster Session (Board #20G), Sat, 8:00 AM-12:00 PM<br />

Racial differences in outcomes <strong>of</strong> triple-negative breast cancer. Presenting<br />

Author: Jose Pacheco, Washington University, St. Louis, MO<br />

Background: The incidence and mortality <strong>of</strong> breast cancer can differ<br />

significantly among racial and ethnic groups. African <strong>American</strong> (AA) women<br />

have a lower incidence <strong>of</strong> breast cancer, but higher mortality compared to<br />

other racial groups. Triple negative breast cancer (TNBC: negative for the<br />

expression <strong>of</strong> estrogen receptor, progesterone receptor and HER2), which is<br />

an aggressive type <strong>of</strong> breast cancer, occurs more frequently in AA women.<br />

The few studies addressing whether racial differences exist in the outcomes<br />

<strong>of</strong> patients with TNBC have yielded inconsistent results. Methods: The<br />

Washington University Medical Oncology Database captures the clinical<br />

information for all new patients (pts) seen at the Breast Oncology Clinic.<br />

Most <strong>of</strong> these pts reside in the St. Louis metropolitan area. Using this<br />

database, we performed a retrospective analysis to examine the association<br />

<strong>of</strong> race with the clinical presentation and outcome <strong>of</strong> TNBC in this<br />

geographically defined patient population. Results: Between May 2006 and<br />

March 2011, 506 pts with TNBC were entered in the database. Analysis<br />

was done on 499 patients for whom follow up data is available. The median<br />

follow up (F/U) time was 24.5 months and the median age at diagnosis was<br />

53 (24 to 98) years. Thirty percent <strong>of</strong> pts were AA. Only 5% presented with<br />

stage IV at diagnosis and the majority <strong>of</strong> tumors (86%) were high grade.<br />

Neoadjuvant chemotherapy was administered in 151 pts, 22% <strong>of</strong> whom<br />

achieved a pathologic complete response (pCR). There was no significant<br />

difference between races in the age <strong>of</strong> diagnosis, F/U time, tumor stage,<br />

grade, frequency <strong>of</strong> receiving neo/adjuvant chemotherapy and pCR rate to<br />

neoadjuvant chemotherapy. There was no difference in disease free survival<br />

(DFS) and overall survival (OS) between AA and other racial groups by<br />

either univariate or multivariate analysis that took into account tumor<br />

stage, grade, patient age and menopausal status. The HR for OS was 1.154<br />

(CI 0.772 – 1.725, p value 0.4860) and for DFS it was 0.947 (CI 0.650 –<br />

1.380, p value 0.7764) in AA compared to other races. In the 92 pts who<br />

recurred, there was no racial difference in time from recurrence to death.<br />

Conclusions: Race does not significantly affect the clinical presentation or<br />

outcome <strong>of</strong> TNBC in the St. Louis metropolitan area.<br />

1050 General Poster Session (Board #21A), Sat, 8:00 AM-12:00 PM<br />

Evaluation <strong>of</strong> a multiparametric system able to predict nonsentinel lymph<br />

node status in breast cancer patients with a micrometastatic sentinel node<br />

assessed by the one step nucleic acidamplification (OSNA) assay. Presenting<br />

Author: Simonetta Buglioni, Regina Elena National Cancer Institute,<br />

Rome, Italy<br />

Background: Axillary lymph node dissection (ALND) may not be necessary in<br />

women with breast cancer (BC) who have micrometastasis in a sentinel<br />

lymph node (SLN), owing to the low risk <strong>of</strong> non-SLN (NSLN) involvement.<br />

In our Institute we validated and adopted the molecular diagnostic tool<br />

OSNA based on the quantitative measurement <strong>of</strong> Cytokeratin 19 (CK19)<br />

mRNA.The aims <strong>of</strong> our work in a subgroup <strong>of</strong> women with micrometastatic<br />

SLN, were: 1) to correlate the copy numbers <strong>of</strong> CK19 mRNA with the risk <strong>of</strong><br />

additional positive NSLNs; 2) to assess the relationships between the<br />

molecular subtype classification and the probability <strong>of</strong> a positive ALND; 3)<br />

to verify whether a combination <strong>of</strong> the new above mentioned parameters is<br />

able to identify a subgroup <strong>of</strong> patients with a micrometastatic SLN and a<br />

negligible risk <strong>of</strong> positive NSLNs in whom ALND may be avoided. Methods:<br />

The SLN lysates from 709 patients were analyzed by OSNA assay. We<br />

considered only patients with a micrometastatic SLN (copy numbers<br />

between 250 and 5000/�L) and the probability <strong>of</strong> having a positive ALND<br />

was calculated by the logistic regression model. This series <strong>of</strong> BC patients<br />

were divided into four main subtypes taking in account the BC classification<br />

as defined by a combination <strong>of</strong> estrogen, progesteron receptors and<br />

HER2 status. Results: OSNA positivity for micrometastasis was reported in<br />

91/709 cases (12,8%).The number <strong>of</strong> patients with positive ALND was 20<br />

(22%). The statistical analyses showed that the metastatic involvement <strong>of</strong><br />

NSLNs is associated with SLNs with a high copy numbers (�2000) <strong>of</strong><br />

CK19 mRNA together with HER2 subtype. Otherwise none <strong>of</strong> the luminal A<br />

patients with a positive SLN but presenting a copy number �1000, had a<br />

positive NSLNs. Conclusions: We showed that biologically-driven analyses<br />

may be able to build new models with higher performance in terms <strong>of</strong> breast<br />

cancer axillary status prediction after positive SLN biopsy for micrometastasis.<br />

The copy numbers <strong>of</strong> CK19 mRNA and the molecular subtypes are<br />

more advantageous than traditional parameters because they are not<br />

pathologist-dependent and therefore they are more reliable and reproducible.<br />

Breast Cancer—Triple-Negative/Cytotoxics/Local Therapy<br />

1049 General Poster Session (Board #20H), Sat, 8:00 AM-12:00 PM<br />

A prognostic model for predicting breast cancer (BC)-related survival in<br />

operable triple-negative (TN) patients (pts). Presenting Author: Elisabetta<br />

Munzone, European Institute <strong>of</strong> Oncology, Medical Oncology, Milan, Italy<br />

Background: TNBC represent a heterogeneous disease in terms <strong>of</strong> biology,<br />

prognosis, and treatment response. We propose a prognostic model to<br />

identify homogeneous subgroups <strong>of</strong> patients and tailor risk-adapted adjuvant<br />

therapies indications. Methods: We analyzed 1,049 pts operated in our<br />

institute from 1997 to 2007 for early TNBC. Pts who received neoadjuvant<br />

chemotherapy (CT), with T4 tumors or previous history <strong>of</strong> cancer were<br />

excluded. Death from BC was the primary endpoint <strong>of</strong> the study. We<br />

calculated an individual predicted risk using a multivariable Cox regression<br />

model, with age, tumor size, number <strong>of</strong> positive lymph nodes and Ki-67<br />

analyzed as continuous covariates, and tumor grade and perivascular<br />

invasion as categorical covariates. Results: Median age was 52 years, 562<br />

(53.4%) and 670 (65.1%) pts had a pT1 and pN0 TNBC, respectively.<br />

Median Ki-67 was 48%. Adjuvant CT regimens were distributed as follows:<br />

classical CMF 388 (37.0%), anthracycline containing regimens 455<br />

(43.4%), taxanes 12 (1.1%), other regimens 66 (6.3%) and no CT 128<br />

(12.2%). After a median follow-up <strong>of</strong> 6 years, 131 deaths from BC were<br />

observed (5-year cumulative incidence 11.9%). At multivariable analysis,<br />

age, tumor size, number <strong>of</strong> positive lymph nodes, Ki-67, tumor grade and<br />

perivascular invasion were associated with the risk <strong>of</strong> death and were<br />

included in the prognostic model. Its predictive accuracy was good (C-index<br />

0.73). We subsequently identified three homogeneous prognostic subgroups<br />

- low, medium and high-risk - according to the tertiles values <strong>of</strong> the<br />

predicted risk. The outcomes are shown in the table. Conclusions: We could<br />

identify homogeneous prognostic subgroups <strong>of</strong> TNBC pts according to<br />

clinical-pathological features. This prognostic model suggests that the use<br />

<strong>of</strong> CT in TN low-risk pts might be questionable. We are currently externally<br />

validating this model on a different series <strong>of</strong> pts.<br />

Risk subgroup Treatment No. (%)<br />

Deaths<br />

from BC<br />

61s<br />

5-year<br />

survival % P value<br />

Low-risk CT 262 (75.1) 15 95.5 0.72<br />

No CT 87 (24.9) 4 96.2<br />

Medium-risk CT 326 (93.1) 31 91.0 0.29<br />

No CT 24 (6.9) 4 81.6<br />

High-risk CT 334 (95.4) 70 78.8 0.03<br />

No CT 16 (4.6) 7 53.9<br />

1051 General Poster Session (Board #21B), Sat, 8:00 AM-12:00 PM<br />

Impact <strong>of</strong> biomarkers in predictivity <strong>of</strong> the efficacy and toxicity <strong>of</strong> a<br />

combination <strong>of</strong> panitumumab plus FEC 100 followed by docetaxel in a<br />

phase II neoadjuvant trial for triple-negative breast cancer (TNBC) patients.<br />

Presenting Author: Jean-Marc Nabholtz, Centre Jean Perrin, Clermont-<br />

Ferrand, France<br />

Background: We evaluated the combination <strong>of</strong> a standard chemotherapy<br />

with panitumumab as neoadjuvant therapy <strong>of</strong> operable TNBC. Complete<br />

pathologic response (pCR) was the primary endpoint, with toxicity and<br />

biologic ancillary studies as secondary endpoints. Methods: Sixty patients<br />

with stage II-IIIA disease were prospectively included in this multicentre<br />

pilot study. Systemic therapy (ST) consisted <strong>of</strong> 4 cycles <strong>of</strong> FEC 100<br />

(500/100/500 mg/m2 ) q.3 weeks followed by 4 cycles <strong>of</strong> T (100 mg/m2 )<br />

q.3 weeks, in combination with panitumumab (9 mg/kg) for 8 cycles q.3<br />

weeks. All patients underwent surgery at completion <strong>of</strong> ST. Paraffinembedded<br />

samples and frozen samples have been systematically realised<br />

before and after neaodjuvant treatment in order to evalutate the biological<br />

pr<strong>of</strong>ile <strong>of</strong> the tumor. Patients characteristics are : median age 50 ; median<br />

tumor size : 40 mm ; invasive ductal carcinoma : 96% ; Scarff-Bloom-<br />

Richardson Grade III : 70%, grade II : 30%, ki-67-positive : 100%,<br />

EGFR-positive : 78%, cytokeratine5-6-positive : 48% and p53-positive :<br />

59%. Pathological response showed a pCR according to Satal<strong>of</strong>f’s classification<br />

<strong>of</strong> 52.38% and according to Chevallier’s classification <strong>of</strong> 46.52%.<br />

Skin toxicity was the main side-effect : Cutaneous toxicity grade IV : 5%,<br />

grade III : 30%, grade II : 20%. Neutropenia grade IV : 27% ; febrile<br />

neutropenia : 5%. Infection : 0%. Hand-foot syndrome grade III : 3.3%.<br />

Ungueal toxicity grade III : 1.6%, grade II : 20%. Results: We have tested<br />

the predictive value <strong>of</strong> ki-67, EGFR, cytokeratine 5-6 and p53. Only ki-67<br />

is predictive <strong>of</strong> a pCR according to Chevallier’s classification (p�0.026),<br />

with a cut-<strong>of</strong>f <strong>of</strong> 40% <strong>of</strong> positive cells (ROC curve): 62% <strong>of</strong> pCR if ki-67�<br />

40% versus 23% if not (relative risk : 2.7). Low EGFR, high p53, and high<br />

cytokeratine 5-6 tended to be associated with poor reponse. No correlations<br />

were found between cutaneous toxicities and these biomarkers. The<br />

cutaneous toxicities were not predictive. Conclusions: HighKi-67 is predictive<br />

<strong>of</strong> more pCR. High EGFR, low p53 and low cytokeratine 5-6 tended to<br />

be associated with better response, but the data are not significant.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


62s Breast Cancer—Triple-Negative/Cytotoxics/Local Therapy<br />

1052 General Poster Session (Board #21C), Sat, 8:00 AM-12:00 PM<br />

Effect <strong>of</strong> therapeutic targeting <strong>of</strong> the oncogene EMP2 in triple-negative<br />

breast cancer on tumor load. Presenting Author: Madhuri Wadehra,<br />

DGSOM at UCLA, Los Angeles, CA<br />

Background: Understanding tumor induced angiogenesis is a challenging<br />

problem with important consequences for the diagnosis and treatment <strong>of</strong><br />

cancer. In this study we define a novel function for the tetraspan epithelial<br />

membrane protein-2 (EMP2) in the control <strong>of</strong> neoangiogenesis. EMP2 is an<br />

oncogene whose expression has been shown to correlate with tumor<br />

progression and survival in a number <strong>of</strong> human cancers including triple<br />

negative breast, ovarian, and endometrial tumors. In breast cancer, EMP2<br />

is highly expressed on the majority <strong>of</strong> invasive tumors examined, and in<br />

particular 70% <strong>of</strong> triple negative breast tumors showed surface expression<br />

<strong>of</strong> this marker. In this study, we examine the control <strong>of</strong> EMP2 on the tumor<br />

microenvironment and further characterize its therapeutic efficacy in<br />

breast cancer. Methods: A number <strong>of</strong> cancer cell lines were utilized both in<br />

vitro and in vivo to determine if EMP2 expression levels altered the tumor<br />

microenvironment. Cells were tested for their expression <strong>of</strong> pro-angiogenic<br />

proteins HIF-1a and VEGF-A as well as for their ability to induce endothelial<br />

cell migration and capillary like tube formation. Cells with modulated<br />

EMP2 levels were also tested in vivo for tumor formation and examined for<br />

histological changes in the tumor microenvironment. In addition, we<br />

recently constructed a fully human EMP2 IgG1 and determined its<br />

therapeutic efficacy in a number <strong>of</strong> triple negative breast cancer cells.<br />

Results: In vitro, upregulation <strong>of</strong> EMP2 promoted VEGF expression through<br />

a HIF-1a dependent pathway and resulted in successful capillary-like tube<br />

formation. In contrast, reduction <strong>of</strong> EMP2 correlated with reduced HIF-1a<br />

and VEGF expression with the net consequence <strong>of</strong> poorly vascularized<br />

tumors in vivo. Treatment <strong>of</strong> breast cancer cells with EMP2 IgG1 reduced<br />

tumor load with a significant improvement in survival. Conclusions: <strong>Clinical</strong>ly,<br />

EMP2 has been shown to correlate with poor survival, and these<br />

results suggest that this occurs in part through its control on tumor<br />

vasculature. In addition, we provide additional evidence on the potent<br />

cytotoxic effects <strong>of</strong> EMP2 treatment in vivo.<br />

1054 General Poster Session (Board #21E), Sat, 8:00 AM-12:00 PM<br />

Biomarkers <strong>of</strong> response to Akt inhibitor MK-2206 in breast cancer.<br />

Presenting Author: Takafumi Sangai, University <strong>of</strong> Texas M. D. Anderson<br />

Cancer Center, Houston, TX<br />

Background: Akt significantly contributes to cancer pathogenesis. PTEN, a<br />

negative regulator <strong>of</strong> PI3K/Akt signaling, is mutated or decreased, and<br />

PIK3CA is frequently mutated in multiple cancer lineages. We hypothesized<br />

that MK-2206, an allosteric Akt inhibitor, would inhibit Akt<br />

signaling thus blocking cancer cell growth, and PTEN and/or PIK3CA<br />

mutations may confer MK-2206 sensitivity in breast cancer. Methods: After<br />

determining sensitivity to MK-2206 in16 cell lines, the effect on Akt<br />

signaling was tested by reverse-phase protein array analysis and western<br />

blotting. The effect to cell cycle progression and cell death were analyzed<br />

by flow cytometry. Using RNA knockdown technique, the effect <strong>of</strong> PTEN,<br />

PIK3CA and Akt on MK2206 sensitivity was tested. Anti-tumor effect in<br />

vivo with or without paclitaxel was tested using PTEN-mutant ZR75-1<br />

breast cancer xenografts. Results: MK-2206 inhibited Akt signaling and<br />

cell cycle progression, and increased apoptosis in a dose-dependent<br />

manner in breast cancer cell lines. Cell lines with PTEN or PIK3CA<br />

mutations were more sensitive to MK-2206 (P�0.0337), however, a<br />

number <strong>of</strong> lines with PTEN/PIK3CA mutations were MK-2206-resistant.<br />

Small interfering RNA (siRNA) knockdown <strong>of</strong> PTEN in breast cancer cells<br />

increased Akt phosphorylation concordant with increased MK-2206 sensitivity.<br />

Stable transfection <strong>of</strong> PIK3CA E545K or H1047R mutant plasmids<br />

into normal-like MCF10A breast cells enhanced MK-2206 sensitivity. Cell<br />

lines that were less sensitive to MK-2206 had lower ratios <strong>of</strong> Akt1/Akt2 and<br />

had less growth inhibition with Akt siRNA knockdown. In ZR75-1 xenografts,<br />

MK-2206 treatment inhibited Akt signaling, cell proliferation, and<br />

tumor growth. In vitro, MK-2206 showed a synergistic interaction with<br />

paclitaxel in MK-2206-sensitive cell lines, and this combination had<br />

significantly greater antitumor efficacy than either agent alone in vivo.<br />

Conclusions: MK-2206 has antitumor activity alone and in combination<br />

with chemotherapy. This activity may be greater in tumors with PTEN loss<br />

or PIK3CA mutation, providing a strategy for patient enrichment in clinical<br />

trials. However, not all tumors with PIK3CA/PTEN aberrations are MK-2206sensitive,<br />

emphasizing the need for additional markers <strong>of</strong> response.<br />

1053 General Poster Session (Board #21D), Sat, 8:00 AM-12:00 PM<br />

<strong>Clinical</strong> characteristics and chemotherapy options <strong>of</strong> triple-negative breast<br />

cancer: Role <strong>of</strong> classical CMF regimen. Presenting Author: Min Hee Hur,<br />

Department <strong>of</strong> Surgery, Kwandong University College <strong>of</strong> Medicine, Cheil<br />

General Hospital, Seoul, South Korea<br />

Background: Triple-negative breast cancer is a high risk breast cancer that<br />

lacks the benefit <strong>of</strong> specific targeted therapy. We investigated the clinicopathologic<br />

characteristics between triple-negative breast cancers (TNBC)<br />

and non-TNBC. And we also analyzed the effect <strong>of</strong> chemotherapy options<br />

such as classical CMF regimen, anthracycline-based, or taxane-based<br />

chemotherapy. Methods: A total <strong>of</strong> 826 invasive breast cancer patients were<br />

evaluated from March 2003 to December 2008. We investigated them<br />

retrospectively, who had the median follow-up for 88 months. We examined<br />

the differences between TNBC compared with non-TNBC in relation to the<br />

clinicopathologic parameters, chemotherapy regimen, overall survival (OS).<br />

Results: 156 (18.9%) cases among 826 patients were triple negative breast<br />

cancers. There were significantly positive associations with younger age<br />

(below 35 years), large tumor (�2cm), high stage, poorly differentiated<br />

nuclear grade, poorly histologic grade in TNBC. Positive lymph node,<br />

lymphovascular invasion were not significantly different between TNBC and<br />

non- TNBC. A total <strong>of</strong> 677 patients were treated with chemotherapy. In<br />

TNBC patients, 142 (93.4%) patients were treated with chemotherapy<br />

more than in 535 (61.4%) <strong>of</strong> non- TNBC patients. The chemotherapy in<br />

TNBC patients was composed <strong>of</strong> classical CMF (47.9%), anthracyclinebased<br />

regimen (25.4%), taxane-based regimen (26.8%). 19 cases (12%)<br />

<strong>of</strong> TNBC experienced locoregional or systemic metastases. 48 (7.2%)<br />

patients <strong>of</strong> non- TNBC did local or systemic metastases. Patients with<br />

TNBC had worse 5-year OS than with non-TNT (95.7% vs 98.6%,<br />

p�0.01). Interestingly, patients treated with CMF regimen were better<br />

5-year OS than with anthracycline-based, or taxane-based regimen in<br />

TNBC (100% vs 96.9% vs 89.2%, p�0.001). There was no survival<br />

difference among chemotherapy regimens in non-TNBC patients.<br />

Conclusions: Patients with TNBC have poor prognosis compared with<br />

non-TNBC. Classical CMF regimen for TNBC patients may be more<br />

effective than anthracycline-based or taxane-based regimens.<br />

1055 General Poster Session (Board #21F), Sat, 8:00 AM-12:00 PM<br />

Preclinical evaluation <strong>of</strong> selective inhibitors <strong>of</strong> nuclear export (SINE) in<br />

basal-like breast cancer (BLBC). Presenting Author: Dilara McCauley,<br />

Karyopharm Therapeutics, Boston, MA<br />

Background: Basal-like breast cancers (BLBC) compose up to 15% <strong>of</strong> breast<br />

cancer (BC) and are usually triple negative characterized by lack <strong>of</strong> ER, PR,<br />

and HER-2 amplification. In addition, most BRCA1-associated BCs are<br />

BLBC and TNBC, expressing basal cytokeratins and EGFR. BLBC is<br />

characterized by an aggressive phenotype, high histological grade, and poor<br />

clinical outcomes: high recurrence and metastasis rates. CRM1 (XPO1) is<br />

the exclusive nuclear exporter <strong>of</strong> multiple Tumor Suppressor Proteins (TSP)<br />

including p53, p21, BRCA1&2, pRB, FOXO. CRM1 inhibition forces<br />

nuclear accumulation <strong>of</strong> TSPs, inducing apoptosis in cancer cells. KPT-<br />

SINE are novel, small molecule, irreversible inhibitors <strong>of</strong> CRM1 with potent<br />

anti cancer activity. Methods: The Cancer Genome Atlas (TCGA) and BC cell<br />

line databases were used for mRNA analyses. MTT assay was used to<br />

determine the cytotoxic effect <strong>of</strong> KPT-SINE (KPT-185 and KPT-330) on 44<br />

breast cell lines including luminal A, luminal B, HER2 positive, BLBC and<br />

TNBC cells. The effect <strong>of</strong> KPT-330 treatment on tumor growth was tested<br />

in vivo in the TNBC model MDA-MB-468 xenograft. Results: Analyses <strong>of</strong><br />

nuclear pore complex (NPC)-related mRNA levels (including CRM1)<br />

showed clear separation <strong>of</strong> BCs into high and low NPC expression. BLBC<br />

subtype was enriched with high NPC transcripts while luminal BC was<br />

enriched in low NPC levels (p�1.53e-20). High NPC levels had higher<br />

mutation levels in BRCA2 (cor�0.33, p�1.83e-8) and ABL1. NPC<br />

expression was inversely correlated with ER mRNA expression (cor�-0.58,<br />

p�1.37e-7). KPT-SINE showed potent cytotoxicity on �75% <strong>of</strong> the cell<br />

lines (IC50 values �1 �M). Only three <strong>of</strong> 24 TNBC cell lines displayed IC50 values �1.5 �M upon KPT-SINE treatment. Genomic analyses on all BC<br />

lines indicated that p53, PI3K/AKT and BRCA1 or 2 status did not affect<br />

cytotoxicity. In MDA-MB-468 xenograft, KPT-330 displayed efficacy in a<br />

dose-dependent manner inhibiting nearly 100% <strong>of</strong> tumor growth compared<br />

with vehicle treated animals, and was well tolerated. Conclusions: These<br />

data show that NPC/CRM1 mRNAs are overexpressed in BLBC/TNBC and<br />

that CRM-1 mediated nuclear export inhibition by SINE represents a<br />

potentially novel and well tolerated therapy for BLBC / TNBC.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


1056 General Poster Session (Board #21G), Sat, 8:00 AM-12:00 PM<br />

The role and significance <strong>of</strong> FoxM1 in invasive breast cancer. Presenting<br />

Author: Cha Kyong Yom, Department <strong>of</strong> Surgery, Seoul National University<br />

Bundang Hospital, Seoul National University College <strong>of</strong> Medicine, Seongnam,<br />

South Korea<br />

Background: The Forkhead Box protein M1 (FoxM1) is known to regulate a<br />

variety <strong>of</strong> biologic processes in mammalian cells including cell growth and<br />

survival, angiogenesis, DNA damage response, chemotherapeutic drug<br />

resistance, and cancer cell migration and invasion. We evaluate the role<br />

and significance <strong>of</strong> Fox M1 in primary breast cancer in vitro and analyzed<br />

the relation with FoxM1 expression and clinicopathologic features. Methods:<br />

Immunohistochemical staining was used for evaluation <strong>of</strong> cytoplasmic<br />

expression <strong>of</strong> FoxM1 with TMA <strong>of</strong> invasive breast cancer. In various breast<br />

cancer cell lines, we evaluated FoxM1 expression and treated docetaxel/<br />

cisplatin in combination with Siomycin A (FoxM1 inhibitor) for BT20 cell<br />

line. Results: From Nov 1995 to Jul 2007, in 84 patients with stage 1-3<br />

invasive breast cancer, FoxM1 expression was noted in 58.7%. Median<br />

follow-up duration was 85.1 months. Lymphovascular invasion was positively<br />

correlated with FoxM1 expression (p�0.040). In multivariate analysis,<br />

FoxM1 expression (p�0.005), HR negativity (p�0.002), high histologic<br />

grade (p�0.023), hign nuclear grade (p�0.045), lymphovascular invasiveness<br />

(p�0.017), and stage 3 cancer (p�0.015) matched poor disease-free<br />

survival. In vitro study, FoxM1 was expressed BT474, JIMT-1, BT20,<br />

HCC-1937, and MDA-MB-231 cell lines. The inhibition <strong>of</strong> FoxM1 had<br />

synergistic effect on cisplatin treatment, not docetaxel in BT20 cell.<br />

Conclusions: FoxM1 expression was noted in triple negative breast cancer<br />

cell lines and its inhibition had synergistically cytotoxic effect on BT20 cell<br />

line in combination with cisplatin. Although the further in vivo and clinical<br />

study should be needed to draw the solid conclusions, FoxM1 could be both<br />

a promising target <strong>of</strong> treatment for triple negative breast cancer and a<br />

independent prognostic factor.<br />

1058 General Poster Session (Board #22A), Sat, 8:00 AM-12:00 PM<br />

Next-generation sequencing mutational analysis <strong>of</strong> triple-negative breast<br />

cancer patients from matched FFPE and fresh frozen samples. Presenting<br />

Author: Mark Landers, AltheaDx, San Diego, CA<br />

Background: TNBC is an aggressive subtype <strong>of</strong> breast cancer accounting for<br />

10-15% <strong>of</strong> all cases. TNBC tumors (ER-/PR-/HER-) are more common in<br />

patients with BRCA mutations. BRCA mutations leading to homologous<br />

DNA repair deficient tumors enhance the efficacy <strong>of</strong> chemotherapy and<br />

PARP inhibitors. BRCA mutations have been identified in 20% <strong>of</strong> patients<br />

without family history. Identification <strong>of</strong> germline and somatic BRCA<br />

mutations in unselected patients could increase the number <strong>of</strong> patients<br />

who benefit from these therapies. Determining BRCA mutational status<br />

from FFPE and fresh frozen specimens may enable clinical studies in these<br />

patient populations.We describe the development <strong>of</strong> an NGS BRCA<br />

mutational assay compatible with FFPE and fresh frozen samples using<br />

tumor/adjacent normal matched tissues. Methods: Matched samples were<br />

purchased from Cureline. gDNA was isolated by lysis/column purification<br />

(Qiagen) and enriched for BRCA exons/flanking regions (Halogenomics<br />

Selector). Fragment libraries were constructed (Ion Torrent frag express)<br />

and prepared for sequencing by emPCR (Ion Torrent Template Xpress).<br />

Libraries were sequenced for 65 cycles (Ion Torrent PGM) yielding 2-4M<br />

reads/sample. Variants were called from tMAP aligned reads by GATK and<br />

VarScan. Overlapped exonic variants were filtered by p-value (�0.0001)<br />

from VarScan. Results: In the first patient set normalized average depth <strong>of</strong><br />

BRCA exon coverage was 64X and 72X per 150K reads in FFPE and fresh<br />

frozen tissues respectively covering 95-100% <strong>of</strong> target. hg19 alignment<br />

rates varied between 97-99% across all samples. Similar numbers <strong>of</strong><br />

variants were called in both FFPE (12) vs. fresh frozen (13) with a<br />

corresponding mean duplicate removed depth <strong>of</strong> coverage <strong>of</strong> 23.3X and<br />

42.4X at the called positions. 10/13 calls in fresh frozen overlapped with<br />

those in FFPE. A tumor specific somatic frameshift insertion in BRCA2 was<br />

detected in both FFPE and fresh frozen tissues. Conclusions: Results<br />

indicate that NGS mediated BRCA mutational analysis demonstrates<br />

equivalent utility in both FFPE and fresh frozen tumor samples although<br />

more sequencing reads are required to produce equivalent depth <strong>of</strong><br />

coverage starting from FFPE samples.<br />

Breast Cancer—Triple-Negative/Cytotoxics/Local Therapy<br />

63s<br />

1057 General Poster Session (Board #21H), Sat, 8:00 AM-12:00 PM<br />

Use <strong>of</strong> gene expression and alternative splicing signatures to discriminate<br />

breast cancer stem cells from fibroblasts. Presenting Author: David T.<br />

Weaver, Verastem Inc, Cambridge, MA<br />

Background: Tumors frequently contain cancer stem cells (CSCs) or<br />

tumor-initiating subpopulations, with an ability to self-renew and regenerate<br />

all cell types within the tumor. Basal-like breast cancers exhibit<br />

features <strong>of</strong> CSCs, including expression <strong>of</strong> surface markers, even though<br />

these cells are rare. Given the role <strong>of</strong> CSCs in the recurrence and spread <strong>of</strong><br />

cancer, there is an urgent need to develop new therapeutic agents that<br />

target CSCs. Development <strong>of</strong> CSC-targeted drugs will be greatly facilitated<br />

by biomarkers that can identify CSCs to aid in patient selection and<br />

determination <strong>of</strong> drug response. Defining the CSCs in tumors is complicated<br />

by the high mesenchymal nature <strong>of</strong> fibroblasts. Analysis <strong>of</strong> gene<br />

expression and alternative splicing patterns in CSCs that are not observed<br />

in fibroblasts may provide valuable new CSC-specific markers. Methods:<br />

Alternative splicing and gene expression microarray strategies were used to<br />

identify selected exons and differentially expressed genes between 10<br />

Basal human breast cancer cell lines and a combination <strong>of</strong> 12 Luminal and<br />

3 fibroblast cell lines. Q-PCR analysis was conducted to determine<br />

candidate CSC gene differential expression between Basal, Luminal, and<br />

Fibroblast cells lines. Results: Expression levels <strong>of</strong> 11 genes were higher<br />

and 24 genes were lower in the Basal cell lines versus Luminal or<br />

fibroblastic cell lines. Comparison <strong>of</strong> Basal cell lines to the Luminal/<br />

Fibroblast cell lines identified 36 cassette exons that were included, and<br />

26 that were excluded in Basal cell lines. Also, 19 genes were upregulated<br />

in Basal cell lines compared to the other groups as detected by Q-PCR.<br />

Interestingly, the 19-multigene model defined the Triple Negative Breast<br />

Cancer patients that were Likely to Recur under standard chemotherapy<br />

with a p � 1.90e-03 and AUC 0.723. Conclusions: Gene and exon marker<br />

sets distinguish CSC versus fibroblasts and may be instructive in identifying<br />

patients that recur early in Triple Negative Breast Cancer. The CSCassociated<br />

RNA signatures identified here will be further refined to develop<br />

new CSC-specific diagnostic markers to stratify breast cancer patients and<br />

monitor response to novel CSC-targeted therapies.<br />

1059 General Poster Session (Board #22B), Sat, 8:00 AM-12:00 PM<br />

A randomized phase II trial <strong>of</strong> doxorubicin plus pemetrexed followed by<br />

docetaxel versus doxorubicin plus cyclophosphamide followed by docetaxel<br />

as neoadjuvant chemotherapy (NACT) for early breast cancer: Three-year<br />

follow-up data. Presenting Author: Andreas Schneeweiss, University Hospital<br />

Heidelberg, Heidelberg, Germany<br />

Background: NACT for early breast cancer allows in vivo chemosensitivity<br />

testing. Primary results <strong>of</strong> this randomized, non-comparative 2-arm study<br />

have been published (Schneeweiss et al, Ann Oncol 2011). Here we<br />

provide 3-year follow-up data for disease-free survival (DFS) and safety.<br />

Methods: 257 patients (pts) with untreated operable T2–T4a–c N0–2 M0<br />

breast cancer were randomly assigned to receive either four cycles <strong>of</strong><br />

doxorubicin 60 mg/m² plus pemetrexed 500 mg/m² every 3 weeks (q3w)<br />

followed by four cycles <strong>of</strong> docetaxel 100 mg/m² q3w (AP-D; 135 pts), or<br />

four cycles <strong>of</strong> doxorubicin 60 mg/m² plus cyclophosphamide 600 mg/m²<br />

q3w followed by four cycles <strong>of</strong> docetaxel 100 mg/m² q3w (AC-D; 122 pts).<br />

Both arms were stratified according to hormone receptor (HR) status<br />

(estrogen and/or progesterone receptor positive vs both negative) and study<br />

center. Surgery was carried out within 2 months after last chemotherapy.<br />

Primary objective was pathological complete response (pCR) rate in the<br />

breast (ypT0/is). Secondary objectives included long-term efficacy and<br />

safety measures. DFS and adverse event data were collected from all<br />

patients for 3 years or until progression or death. The Kaplan-Meier (KM)<br />

analysis technique and Cox regression method were used as statistical<br />

measures. KM analyses were performed on HR-positive and -negative pts<br />

subgroups. Results: As reported earlier, pCR rates were 16.5% for AP-D and<br />

20.2% for AC-D. The 3-year DFS rate was 76% and 77% for AP-D and<br />

AC-D, respectively. Cox regression analysis for the overall enrolled population<br />

(regardless <strong>of</strong> treatment) revealed significantly longer DFS in HRpositive<br />

than in HR-negative pts (hazard ratio 0.35; 95% CI 0.22–0.58; p<br />

� 0.001). In HR-positive pts, the 3-year DFS rate was 88% (95% CI:<br />

81–95%) with AP-D and 83% (95% CI: 74–91%) with AC-D. In HRnegative<br />

pts, the 3-year DFS rate was 55% (95% CI: 40–70%) for AP-D<br />

and 68% (95% CI: 53–82%) for AC-D. The 3-year follow-up data did not<br />

reveal any changes in the safety pr<strong>of</strong>ile compared to the previously<br />

published results. Conclusions: The 3-year DFS rates <strong>of</strong> both NACTs are in<br />

line with published studies.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


64s Breast Cancer—Triple-Negative/Cytotoxics/Local Therapy<br />

1060 General Poster Session (Board #22C), Sat, 8:00 AM-12:00 PM<br />

Phase II study <strong>of</strong> a novel neoadjuvant chemotherapy (NAC) for breast<br />

cancer (BC). Presenting Author: Miguel E. Albino, Veterans Affairs Hospital,<br />

San Juan, PR<br />

Background: The goal <strong>of</strong> this NAC study was to improve the pathologic<br />

response <strong>of</strong> pts with localized BC. Methods: 51 pts with localized BC �1cm<br />

were treated with this novel regimen consisting first <strong>of</strong> docetaxel 75 mg/m 2 ,<br />

epirubicin 80 mg/m2 , and cyclophosphamide 500 mg/m2 (TEC) and PEG<br />

Filgrastim for 4 cycles. Pretreament PET scan was done and repeated after<br />

course 1. Following the 4th course, ER�/HER2- patients received 4<br />

additional TEC cycles if they achieved CR by MRI, or were switched to a non<br />

cross-resistant regimen (vinorelbine, bevacizumab, capecitabine) if they<br />

had � CR. HER2� pts were given docetaxel � trastuzumab for 4 additional<br />

cycles, or were switched to a different regimen if � PR. MD Anderson<br />

residual cancer burden (MDARCB) score was used to measure pathologic<br />

response and correlation with several prognostic factors was studied.<br />

Results: Median age � 53; 27 were postmenopausal; 42 had invasive<br />

ductal carcinoma, 5 invasive lobular; 12 triple negative, 11 HER2�, and<br />

28 ER� or PR�/HER2-. MDARCB was significantly better in HER-2� and<br />

triple negative tumors (table). ER� or PR�/Her2- pts had the least<br />

favorable MDARCB with none achieving 0 and only 21% attaining<br />

MDARCB�1. 59% <strong>of</strong> pts with � 5% SUV drop attained MDARCB score 0-1<br />

vs 13% with � 5% drop. %Ki-67 correlated well with MDARCB (table).<br />

Ki-67 correlated well with receptor status: 85% <strong>of</strong> Triple Neg or Her2� pts<br />

had Ki-67 �17 vs only 29% <strong>of</strong> ER� or PR�/Her2-. Conclusions: a) This<br />

novel NAC regimen leads to markedly favorable MDARCB scores in triple<br />

negative and Her-2� cases. b) Several factors may be useful in predicting<br />

response to chemotherapy, including receptor status, Ki-67, and PET scan<br />

response after 1st chemotherapy course. c) Ki-67 proliferative rate is<br />

closely correlated with receptor status. d) Early PET could be used to<br />

predict MDARCB. The challenge now is to improve response in ER� or<br />

PR�/Her2- pts.<br />

Factor<br />

MDARCB<br />

0<br />

MDARCB<br />

1<br />

MDARCB<br />

2<br />

MDARCB<br />

3 p N<br />

Triple neg or HER2� 18 (82%) 1 (5%) 2 (9%) 1 (5%) 22<br />

ER� or PR�/HER2- 0 6 (21%) 17 (61%) 5 (18%) 0.00001 28<br />

50*<br />

Ki-67 >17 13 (54%) 2 (8%) 8 (33%) 1 (4%) 24<br />

Ki-67 < 17 2 (9%) 5 (22%) 11 (48%) 5 (22%) .04 23<br />

% drop SUV > 5% 18 (44%) 6 (15%) 14 (34%) 3 (7%) 41<br />

% drop SUV < 5% 0 1 (13%) 4 (50%) 3 (38%) .04 8<br />

*One progressed in liver while on treatment and did not undergo surgery.<br />

1062 General Poster Session (Board #22E), Sat, 8:00 AM-12:00 PM<br />

Current patterns <strong>of</strong> chemotherapy and supportive care for early-stage breast<br />

cancer (ESBC). Presenting Author: Gary H. Lyman, Duke University,<br />

Durham, NC<br />

Background: ESBC is commonly treated with myelosuppressive chemotherapy,<br />

and high relative dose intensity (RDI) correlates with improved<br />

overall survival. A retrospective analysis <strong>of</strong> patients with ESBC treated from<br />

1997–2000 showed that 56% received an RDI � 85% (Lyman et al. JCO.<br />

2003;21:4524-4531). To determine current practice, we evaluated ESBC<br />

treatment patterns at 24 US community- and hospital-based oncology<br />

practices. Methods: Data were abstracted from medical records <strong>of</strong> 532<br />

patients with ESBC treated from January 2007–December 2009. Inclusion<br />

criteria included surgically resected ESBC (stage I-IIIA); � 18 years old;<br />

and completion <strong>of</strong> at least 1 standard chemotherapy cycle on an every 2 or<br />

3 week schedule. The primary endpoint was RDI over planned cycles. Other<br />

endpoints were incidence <strong>of</strong> dose delays � 7 days, dose reductions � 15%<br />

from standard, grade 3/4 neutropenia (SN), febrile neutropenia (FN),<br />

FN-related hospitalization, granulocyte-colony stimulating factor (G-CSF)<br />

use, and antimicrobial therapy. Descriptive statistics were generated for all<br />

endpoints. Results: In this study, mean (range) age was 55 (29–85) years.<br />

Relative to previously published results, chemotherapy regimens have<br />

shifted from mainly doxorubicin � cyclophosphamide (AC) (previously<br />

35%) to docetaxel � cyclophosphamide (TC; n � 221; 42%) and AC<br />

followed by paclitaxel (AC-T; n � 163; 31%). Mean RDI is now higher<br />

(93% for both TC and the most common AC-T schedule [dose dense AC-T;<br />

n � 84] vs 79% previously); the incidence <strong>of</strong> dose delays (16% vs 25%<br />

previously) and dose reductions (21% vs 37% previously) have decreased;<br />

and primary prophylactic use <strong>of</strong> G-CSF has increased (76% vs 3%<br />

previously). In this study, 40% <strong>of</strong> patients had SN, 3% had FN, 2% had an<br />

FN-related hospitalization, and 30% received antimicrobial therapy. These<br />

measures were not available in the previously published results. Conclusions:<br />

The observed changes between the two studies are noteworthy though<br />

inferential comparisons are limited by changes in treatments and other<br />

factors. RDI has improved over time, but 16% <strong>of</strong> patients in this study<br />

received an RDI � 85%. Further evaluation is needed to identify factors<br />

associated with lower RDI and determine outcomes for these patients.<br />

1061 General Poster Session (Board #22D), Sat, 8:00 AM-12:00 PM<br />

The first two lines <strong>of</strong> chemotherapy for anthracycline-naïve metastatic<br />

breast cancer: A comparative study <strong>of</strong> efficacy between anthracyclines and<br />

nonanthracyclines. Presenting Author: Wei-Wu Chen, National Taiwan<br />

University Hospital, Taipei, Taiwan<br />

Background: For anthracycline-naïve metastatic breast cancer (AN-MBC)<br />

patients, past evidence indicated that anthracyclines are beneficial in the<br />

first-two lines <strong>of</strong> palliative chemotherapy but with considerable toxicities.<br />

However, with the provision <strong>of</strong> newer chemotherapies, comparative studies<br />

addressing the efficacy between anthracyclines and non-anthracyclines in<br />

the first-two lines <strong>of</strong> palliative chemotherapy for AN-MBC were lacking.<br />

Methods: We collectedclinicopathological characteristics <strong>of</strong> AN-MBC patients<br />

who had received palliative chemotherapy in National Taiwan<br />

University Hospital between 2001 and 2006. Patients were classified as<br />

anthracycline or non-anthracycline group according to the first-two lines <strong>of</strong><br />

chemotherapy. Kaplan-Meier method and log-rank test were used for the<br />

estimation and comparison <strong>of</strong> both overall survival (OS) and time to<br />

treatment failure <strong>of</strong> the first-two lines (TTF2).Cox proportional hazard<br />

model was used for OS and TTF2. Best composite response rate (BCRR)<br />

were compared with logistic regression test. Results: A total <strong>of</strong> 109 (43.1%)<br />

patients in the anthracycline group and 144 (56.9%) patients in nonanthracycline<br />

group were analyzed. Between these two groups, the distributions<br />

<strong>of</strong> clinicopathological variables were generally similar and their<br />

median OS (33.3 vs 34.2 months, p � 0.179), TTF2 (13.3 vs 12.7<br />

months, p � 0.104), and BCRR (59.5 vs 61.1%, p � 0.81) were not<br />

significantly different. Subgroup analysis showed that patients in the<br />

anthracycline group had a trend toward better OS in the estrogen receptor<br />

(ER) negative/ human epidermal growth factor receptor type II (HER2)<br />

positive subtype (median OS 58.0 vs 31.2 months, p � 0.081). In<br />

multivariate analysis, patients in the anthracycline group had a trend<br />

toward better OS (HR 0.72, 95% CI 0.52 - 1.00, p � 0.052). However, the<br />

exclusion <strong>of</strong> ER-/Her2� subtype attenuated the impact <strong>of</strong> early anthracycline<br />

treatment on OS (HR 0.82, 95% CI 0.56 - 1.18, p � 0.28).<br />

Conclusions: Our study demonstrated that anthracyclines may not be<br />

mandatory in the first-two lines <strong>of</strong> palliative chemotherapy for AN-MBC but<br />

may be more beneficial to ER-/Her2� subtype patients.<br />

1063 General Poster Session (Board #22F), Sat, 8:00 AM-12:00 PM<br />

Chemotherapy <strong>of</strong> methioninase-synchronized S/G2 phase-blocked cancer<br />

cells identified by cell cycle-specific fluorescent reporters. Presenting<br />

Author: Shuya Yano, AntiCancer Inc., San Diego, CA<br />

Background: Cancer cells <strong>of</strong> all types have a generally elevated requirement<br />

for methionine compared to normal cells. This phenomenon is termed<br />

methionine-dependence and may be due to excessive methylation reactions<br />

in cancer cells, since methionine is the global source <strong>of</strong> cellular<br />

methyl groups (Biochim. Biophys. Acta, Reviews on Cancer 738, 49-87,<br />

1984). Deprivation <strong>of</strong> methionine selectively arrests cancer cells during<br />

late S-phase (Proc. Natl. Acad. Sci. USA 77, 7306-7310, 1980), where<br />

they are highly sensitive to chemotherapy drugs which damage DNA (J.<br />

Natl. Cancer Inst. 76, 629-639, 1986). Methods: Cancer cells, transformed<br />

to express different color fluorescent reporters during specific<br />

phases <strong>of</strong> the cell cycle (Cell 132, 487-498, 2008), were used to monitor<br />

the onset <strong>of</strong> the S/G2-phase block due to methionine deprivation effected<br />

by recombinant methioninase (rMETase). The S/G2-phase blocked cancer<br />

cells fluoresced yellow or green in contrast to cancer cells in G1 which<br />

fluoresced red. Cancer cells, including MKN45 stromal cancer and MCF-7<br />

breast cancer, synchronously blocked in S/G2-phase by rMETase, were<br />

identified by their yellow-green fluorescence and allowed to accumulate to<br />

the maximum extent. At the point <strong>of</strong> maximum yellow/green cells in the<br />

culture, the cells were administered chemotherapy drugs which interact<br />

with DNA or block DNA synthesis such as doxorubicin, cisplatin or<br />

5-fluorouracil. We termed this procedure color-coded chemotherapy (CCC).<br />

Results: CCC was highly effective against the cancer cells (90% cell kill). In<br />

contrast, treatment <strong>of</strong> cancer cells with drugs only, and without rMETaseeffected<br />

S/G2-phase synchrony, led to the majority <strong>of</strong> the cancer cell<br />

population being blocked in G1 phase (red fluorescent) where they were<br />

resistant to the drugs (40% cell kill). Conclusions: CCC, which identifies, by<br />

fluorescent color, when cancer cells are blocked in S/G2-phase by a unique<br />

cell-cycle-blocking agent, rMETase, demonstrates the potential <strong>of</strong> cellsynchronization-based<br />

chemotherapy.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


1064 General Poster Session (Board #22G), Sat, 8:00 AM-12:00 PM<br />

Low-dose, short-course sunitinib may normalize tumor vasculature and<br />

improve tumor blood flow to enhance chemotherapy efficacy in breast<br />

cancers. Presenting Author: Soo-Chin Lee, National University Cancer<br />

Institute, Singapore, Singapore, Singapore<br />

Background: Small molecule VEGFR inhibitors (VEGFR-I) have failed to<br />

improve outcome with chemotherapy in most solid tumors. Continuous<br />

administration <strong>of</strong> a potent VEGFR-I may destroy vasculature and impede<br />

drug delivery; in contrast, low-dose, short-course VEGFR-I before chemotherapy<br />

may normalize tumor vasculature and enhance drug delivery.<br />

Methods: We conducted a phase Ib followed by phase II randomized study<br />

in patients with measurable primary breast tumors using low-dose sunitinib<br />

(Su) for 1 week prior to doxorubicin/cyclophosphamide (AC) and measured<br />

tumor blood flow with DCE-MRI and microvessel density and pericyte<br />

recoverage with CD31 and �-smooth muscle actin (SMA) staining on tumor<br />

biopsies, at baseline, after 1 week <strong>of</strong> Su, and 2 weeks after cycle 1 AC.<br />

Patients in phase Ib received 12.5-25mg Su prior to AC; in phase II,<br />

patients were randomized to AC�/-12.5mg Su. Results: 21 patients have<br />

been enrolled, including 3 patients on 25mg Su�AC, 11 on 12.5mg<br />

Su�AC, and 7 on AC alone. After 1 week <strong>of</strong> 25mg Su, 2/3 patients had<br />

reduced tumor blood flow on DCE-MRI indicating anti-angiogenic effects.<br />

After 1 week <strong>of</strong> 12.5mg Su, significant increase in tumor fractional plasma<br />

volume (Vp) occurred (�28%�28%, p�0.025) suggesting increased<br />

perfusion, followed by decrease 2 weeks after AC (-33%�26%,p�0.035)<br />

corresponding to mean tumor size change <strong>of</strong> -17�15%, while no significant<br />

Vp changes occurred with AC alone (p�0.823); CD31 expression<br />

reduced (20.7 vs 15.7, p�0.173) while SMA/CD31 double staining<br />

increased (2.45 vs 7.34, p�0.046) indicating lower microvessel density<br />

and normalization <strong>of</strong> residual vasculature, which was not seen with AC<br />

alone (CD31, p�0.434; CD31/SMA, p�0.605). The main toxicity <strong>of</strong><br />

Su�AC and AC alone was febrile neutropenia (29% vs 43%). 14 patients<br />

had surgery with pathological complete response in 1/6 patients who<br />

received 12.5mg Su�AC. Conclusions: 25mg Su for 1 week can reduce<br />

tumor blood flow, suggesting that concomitant standard dose Su may<br />

compromise drug delivery. Low dose 12.5mg Su for 1 week is sufficient to<br />

normalize tumor vasculature and increase tumor fractional plasma volume,<br />

and may potentially improve drug delivery and treatment outcome.<br />

1066 General Poster Session (Board #23A), Sat, 8:00 AM-12:00 PM<br />

Early changes in angiogenesis and hypoxia following bevacizumab therapy<br />

in primary breast cancer. Presenting Author: Shaveta Mehta, University<br />

Department <strong>of</strong> Oncology, Oxford, United Kingdom<br />

Background: Bevacizumab (BV), a monoclonal antibody directed against<br />

vascular endothelial growth factor (VEGF), is an approved anti-angiogenic<br />

agent, but despite its widespread use, little is known about how tumours<br />

develop resistance to BV. We have conducted a window-<strong>of</strong>-opportunity<br />

study in which BV is administered as a short-term first-line treatment for<br />

primary breast cancer (PBC) patients, and assessed histological markers <strong>of</strong><br />

tumour angiogenesis and hypoxia before and after therapy. Methods: 47<br />

patients with locally advanced breast cancer were prospectively enrolled.<br />

Informed consent was obtained from all patients. A single infusion <strong>of</strong> BV<br />

(15 mg/kg) was given 2 weeks before starting neoadjuvant chemotherapy.<br />

Dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) and<br />

core biopsies for immunohistochemistry (IHC) analysis were performed<br />

immediately prior to and 2 weeks after BV. 36 patients with invasive ductal<br />

carcinoma and good quality MRI scans and core biopsies were included in<br />

this analysis. Wilcoxon rank test and Spearman’s correlation test were used<br />

for statistical analysis. Results: IHC revealed a significant upregulation <strong>of</strong><br />

carbonic anhydrase 9 (P � 0.04) and hypoxia inducible factor-1a (P �<br />

0.001) following BV therapy along with a significant reduction in plasmalemma<br />

vesicle associated protein (PLVAP) (p�0.01) and Ki67 (p� 0.006).<br />

DCE-MRI analysis revealed a significant reduction in vessel permeability<br />

and blood flow following BV, as measured by a decrease in the forward<br />

transfer constant Ktrans (P � 0.0001) and the reverse rate constant kep (P �<br />

0.0001). In addition, we found a significant negative correlation between<br />

CA9 levels and median Ktrans at baseline (Spearman’s rho � -0.46; P �<br />

0.01). Conclusions: Using combined DCE-MRI and IHC analysis, we found<br />

that PBC patients treated with single-agent BV showed a decrease in<br />

markers <strong>of</strong> tumour angiogenesis, together with a corresponding increase in<br />

tumour hypoxia. Our results suggest that tumour hypoxia may be a key<br />

mechanism governing the early onset <strong>of</strong> resistance to BV therapy, and argue<br />

for the use <strong>of</strong> suitable combination therapies to overcome the resistance<br />

and ultimately improve patient survival.<br />

Breast Cancer—Triple-Negative/Cytotoxics/Local Therapy<br />

65s<br />

1065 General Poster Session (Board #22H), Sat, 8:00 AM-12:00 PM<br />

Efficacy <strong>of</strong> low-dose capecitabine in metastatic breast cancer. Presenting<br />

Author: Caitlin Bertelsen, Keck School <strong>of</strong> Medicine <strong>of</strong> the University <strong>of</strong><br />

Southern California, Los Angeles, CA<br />

Background: The FDA-approved dose <strong>of</strong> capecitabine (cape) <strong>of</strong> 1250mg/m2 twice daily (BID) is associated with treatment-limiting toxicities. Published<br />

reports suggest that lower starting doses <strong>of</strong> cape can be as effective as the<br />

approved dose in treating metastatic breast cancer (MBC). We compared<br />

the efficacy <strong>of</strong> lower than previously published doses <strong>of</strong> cape with results <strong>of</strong><br />

registrational Phase III trials using the approved dose. Methods: We<br />

performed a retrospective analysis <strong>of</strong> patients treated at the University <strong>of</strong><br />

Southern California hospitals who received cape as the first, second, or<br />

third line <strong>of</strong> chemotherapy for MBC to determine the progression-free<br />

survival (PFS) associated with low starting doses. The primary endpoint was<br />

PFS among patients with measurable disease, and secondary aims were to<br />

analyze the relationships between PFS and various clinical characteristics<br />

for all patients. Results: Patients (n�84) received a median cape dose <strong>of</strong><br />

565 mg/m2 BID, <strong>of</strong>ten administered as a flat dose (not adjusted for body<br />

surface area) <strong>of</strong> 1000 mg BID. Median PFS among patients with measurable<br />

disease (n�62) was 4.1 months (95% confidence interval or CI �<br />

2.9-5.7), which was similar to the median PFS values <strong>of</strong> 4.4 months (95%<br />

CI � 4.14-5.42, n�480) (Sparano et al. JCO 2010;28:3256) and 4.2<br />

months (95% CI � 3.81-4.50, n�377) (Thomas et al. JCO 2008;25:<br />

5210) for single-agent cape reported in the major trials with similar<br />

eligibility criteria. Among all patients, PFS was shorter in measurable<br />

disease, triple negative and HER2� subtypes, and was similar in all lines <strong>of</strong><br />

therapy. Only two patients (2.4%) discontinued cape due to toxicity.<br />

Conclusions: These data support the efficacy <strong>of</strong> very low doses <strong>of</strong> cape for<br />

MBC. Prospective randomized controlled trials testing lower starting doses<br />

<strong>of</strong> cape are needed to optimize the benefit/risk ratio.<br />

Variable Median PFS (mo) Multivariate PFS OR p value<br />

Entire cohort 4.4<br />

Non-measurable; measurable 19.7; 4.1 Ref; 1.6 0.13<br />

ER or PR�/HER2-; triple negative; HER2� 8.5; 3.0; 6.0 Ref; 3.2; 2.5 0.011<br />

DFI � 0; >0-5yr 3.0; 4.4; 8.5 Ref; 0.42; 0.36 0.031<br />

No trastuzumab; trastuzumab in HER2� 4.1; 9.6 Ref; 0.31 0.079<br />

OR � odds ratio; DFI � disease free interval; Ref � reference; yr � year.<br />

1067 General Poster Session (Board #23B), Sat, 8:00 AM-12:00 PM<br />

Accuracy <strong>of</strong> MRI, mammography (MG), and 2D and 3D ultrasound<br />

(2DUS/3DUS) in determining the pathologic tumor response after neoadjuvant<br />

chemotherapy (NACT) in breast cancer patients. Presenting Author:<br />

Markus Hahn, University Hospital Tuebingen, Department <strong>of</strong> Gynecology<br />

and Obstetrics, Tuebingen, Germany<br />

Background: The pathological tumor response in patients with locally<br />

advanced breast cancer to NACT is essential for survival and for surgical<br />

strategies. Therapy monitoring based on German recommendations is<br />

routinely performed by clinical examination, MG and 2DUS. The clinical<br />

value <strong>of</strong> MRI and 3DUS has not been established yet. The aim <strong>of</strong> the study<br />

was to determine the accuracy, sensitivity, specificity, positive predictive<br />

value (PPV), negative predictive value (NPV) between the different imaging<br />

techniques in predicting postoperative histological tumor response after<br />

NACT. Methods: Patients with primary breast cancer (cT1-T4, cN0-1, M0)<br />

undergoing neoadjuvant chemotherapy between 2005 and 2010 were<br />

eligible for this prospective trial. The response was measured by MRI, MG,<br />

2DUS and 3DUS for complete or partial remission versus stable disease<br />

after the last cycle <strong>of</strong> treatment and compared with the final pathological<br />

response. Patients with progressive disease were excluded from the study.<br />

Statistical analysis was done by calculating the accuracy <strong>of</strong> each imaging<br />

technique and the size difference between imaging and histological tumor<br />

size. Sensitivity, specificity, PPV and NPV were calculated for complete or<br />

partial pathological response. The study was approved by the local ethic<br />

committee (BCD001 194/2004). Results: 103 patients with the mean age<br />

<strong>of</strong> 47.7 (range 24.5 – 71.4) years were evaluated. The accuracy was 0.680<br />

(95%CI: 0.580 -0.768) for MRI, 0.563 (95%CI: 0.453-0.669) for MG,<br />

0.724 (95%CI: 0.618 -0.815) for 2DUS and 0.710 (95%CI: 0.588-<br />

0.813) for 3DUS. Sensitivity, specificity, PPV and NPV were 78%, 47%,<br />

75% and 52% for MRI, 61%, 45%, 69% and 36% for MG, 93%, 23%,<br />

74% and 60% for 2DUS, 94%, 19%, 73% and 57% for 3DUS. The mean<br />

(standard deviation) size difference was -1.8 mm (14.8) on MRI, 1.5 mm<br />

(26.0) on MG, -9.1mm (19.1) on 2DUS and for the volume difference<br />

-6916mm3 (15831) on 3DUS. Conclusions: The data suggest that 2DUS is<br />

sufficient in predicting tumor response between NACT treatment. MRI and<br />

MG are more accurate the 2DUS in predicting the tumor size for surgical<br />

planning.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


66s Breast Cancer—Triple-Negative/Cytotoxics/Local Therapy<br />

1068 General Poster Session (Board #23C), Sat, 8:00 AM-12:00 PM<br />

Patient-reported pain and other symptoms as prognostic factors for overall<br />

survival (OS) in a phase III clinical trial <strong>of</strong> patients with advanced breast<br />

cancer. Presenting Author: Wei Shen, Eli Lilly and Company, Indianapolis,<br />

IN<br />

Background: The safety and efficacy <strong>of</strong> gemcitabine plus paclitaxel versus<br />

paclitaxel in advanced breast cancer after anthracycline-based adjuvant<br />

therapy has been reported previously (Albain et al. 2008). This post-hoc<br />

analysis evaluates the prognostic effect <strong>of</strong> baseline scores <strong>of</strong> the Brief Pain<br />

Inventory short form (BPI-SF) and the Rotterdam Symptom Checklist<br />

(RSCL) on OS. Methods: Patients completed theBPI-SF and the RSCL. BPI<br />

“worst pain” item and BPI interference subscale scores range from 0 (no<br />

pain or interference with daily living) to 10. Four RSCL subscales were<br />

transformed to 0-100, with 100 as best score. Univariate Cox models were<br />

used to determine the prognostic effect <strong>of</strong> each measure on OS. Multivariate<br />

Cox models were used to determine the prognostic effect <strong>of</strong> each<br />

measure in the presence <strong>of</strong> 11 demographic/clinical variables, including<br />

Karn<strong>of</strong>sky performance status, age, and estrogen and progesterone receptor<br />

status. Kaplan-Meier curves and log-rank tests were used to compare OS<br />

among patient groups categorized using clinically meaningful thresholds<br />

and the sample median <strong>of</strong> the BPI and RSCL scores. Results: Randomized<br />

patients were evaluable for this analysis (n�529). In the univariate<br />

analysis, significant prognostic effects on OS were observed for baseline<br />

scores <strong>of</strong> both BPI measures (worst pain and interference) (HRs, 1.07 for<br />

1-point increase; all p�0.0042) and three out <strong>of</strong> four RSCL subscales<br />

(activity level, physical distress, quality <strong>of</strong> life) (HRs, 0.86-0.91 for<br />

10-point increase; all p�0.0120). In the multivariate Cox models, BPI<br />

worst pain remained as a significant prognostic factor (p�0.0245), as did<br />

RSCL activity level (p�0.0004). The median OS for patients with BPI worst<br />

pain score 0 (no pain) was 23.8 months (mos) versus 17.9 mos and 14.8<br />

mos for scores 1-4 (mild) and 5-10 (moderate/severe) (log-rank p�0.0066).<br />

The median OS was 23.8 mos for patients with RSCL activity scores greater<br />

than or equal to the sample median (�95.2) versus 14.6 mos for patients<br />

with scores �95.2 (log-rank p�0.0001). Conclusions: This retrospective<br />

analysis resulted in strong evidence that BPI-SF and RSCL provide distinct<br />

prognostic information for OS.<br />

1070 General Poster Session (Board #23E), Sat, 8:00 AM-12:00 PM<br />

Phase I trial <strong>of</strong> ixabepilone and vorinostat in metastatic breast cancer.<br />

Presenting Author: Thehang H. Luu, City <strong>of</strong> Hope, Duarte, CA<br />

Background: VOR, an HDAC inhibitor, induces acetylation <strong>of</strong> tubulin, and<br />

decreases resistance by attenuating downstream activation <strong>of</strong> AKT, c-Raf<br />

and HER-2. Patients (pts) with MBC treated with VOR had a TTP <strong>of</strong> 8.5<br />

months (range 4-14) and stable disease in 29% (Luu et al., 2008). Our<br />

preclinical data showed synergistic effect <strong>of</strong> IXA and VOR in MDA-MB-231<br />

and MCF7. Methods: The primary aims were to: 1) define the maximum<br />

tolerated dose (MTD) based on dose limiting toxicities (DLT), and 2)<br />

describe the pharmacokinetics (PK) <strong>of</strong> 2 schedules <strong>of</strong> VOR and IXA.<br />

Secondary aims were to describe: 1) response rate (RR) and 2) clinical<br />

benefit rate (CBR). The study included: 1) pts with MBC; 2) ECOG PS 0-2;<br />

3) adequate marrow and organ functions; 4) no prior IXA or VOR; and 5) �<br />

grade (gr) 1 neuropathy. Stable brain metastasis were allowed. Pts were<br />

randomized to schedule A: VOR (QDx14) � IXA (D2) Q21D; or B: VOR<br />

(D1-7 and 15-21) � IXA (D2, 9, 16) Q28D. A modified toxicity probability<br />

interval design (target toxicity rate� 0.2 and equivalence range �/- 0.05)<br />

(Ji et al, 2010) determined dose escalation guidelines. PK were assessed<br />

with LC-MS/MS assays. Results: Among 37 pts randomized, 36 were<br />

evaluable [median age (55 yrs); median prior chemotherapy regimens (3);<br />

ER and/or PR � (64%); HER2 � (19%)]. In cohort A, 16 pts were treated<br />

(1 inevaluable). The MTD was: VOR 300mg (QDx14) � IXA (32mg/m2 D2)<br />

Q21D (dose level 1). DLT was experienced by 27% (4/15) pts [gr 4<br />

neutropenia, gr 3 fatigue/AST, hyponatremia and allergic reaction to IXA].<br />

In cohort B, 21 pts were treated (dose level 1, n�15; dose level 2, n�6).<br />

The MTD was VOR 300mg QD (D1-7 and 15-21) � IXA 16mg/m2 (D2, 9,<br />

16) Q28D (dose level 1), no DLTs were observed. Dose level 2 was closed<br />

with 50% (3/6) <strong>of</strong> pts experiencing DLTs [gr 3 neutropenia, hypertension,<br />

and hypokalemia]. Median cycles treated (cohort A: 6, cohort B: 4).<br />

Response in cohort A and B respectively was: 1 CR, 2 PR, 7 SD (RR: 20%,<br />

CBR: 67%) and 1 CR, 4 PR, 9 SD (RR: 33%, CBR: 93%). VOR and IXA PK<br />

were not influenced by the presence <strong>of</strong> the other drug, clearance values�<br />

220 L/h and 20 L/h/m2 , respectively. Conclusions: We established the MTD<br />

<strong>of</strong> VOR and IXA in pts with MBC. The combination demonstrated clinical<br />

activity in these heavily pretreated pts. Further studies are recommended.<br />

1069 General Poster Session (Board #23D), Sat, 8:00 AM-12:00 PM<br />

Cost-effectiveness analysis using a Markov model assessing the addition <strong>of</strong><br />

bevacizumab to paclitaxel in HER2-negative metastatic breast cancer<br />

patients. Presenting Author: Tamer Refaat, Northwestern University, Chicago,<br />

IL<br />

Background: Metastatic breast cancer (MBC) remains an incurable disease<br />

despite advances in treatment modalities. In 2008, Eastern Cooperative<br />

Oncology Group 2100 trial (E2100) results led to FDA approval for<br />

bevacizumab with paclitaxel in the initial treatment <strong>of</strong> HER2-negative<br />

MBC. The addition <strong>of</strong> bevacizumab to paclitaxel led to a gain <strong>of</strong> around 2.5<br />

months <strong>of</strong> progression-free survival (PFS), no significant benefit on overall<br />

survival (OS), and increased toxicity. In November 2011, the FDA <strong>of</strong>ficially<br />

revoked approval <strong>of</strong> bevacizumab for HER2-negative MBC. However, both<br />

the European Medicines Agency (EMEA) and NCCN still endorse bevacizumab<br />

for this indication. One <strong>of</strong> the greatest challenges facing healthcare<br />

worldwide is reconciling incremental clinical benefits with exponentially<br />

rising costs. This study aimed to assess the cost-effectiveness <strong>of</strong> bevacizumab<br />

with paclitaxel for HER2-negative MBC. Methods: A Markov decision<br />

tree using Data 3.5 (TreeAge S<strong>of</strong>tware, Inc.) was created to do decision and<br />

cost-effectiveness analyses <strong>of</strong> using bevacizumab in combination with<br />

paclitaxel versus paclitaxel alone as first-line chemotherapy in HER2negative<br />

MBC using efficacy and toxicity data from the E2100 study. Costs<br />

were obtained from the Center for Medicare Services Drug Payment Table<br />

and Physician Fee Schedule. The model was designed from the patient and<br />

payer perspectives and sensitivity analyses were run. Results: The marginal<br />

cost between paclitaxel alone versus bevacizumab and paclitaxel was 86K<br />

with a marginal efficacy <strong>of</strong> 0.369 quality-adjusted life-years and marginal<br />

cost effectiveness <strong>of</strong> 232,720.72 USD. The expected outcome value was<br />

1.86 for bevacizumab and paclitaxel and 1.67 for paclitaxel alone.<br />

However, the combination was not cost effective and only a marginal<br />

survival advantage that was not significant was observed. Conclusions: This<br />

study demonstrates that, despite a significant PFS advantage, the addition<br />

<strong>of</strong> bevacizumab to paclitaxel is not cost-effective for patients with HER2negative<br />

MBC. Such data could be informative to policymakers who<br />

consider the health economics and incremental cost-effectiveness <strong>of</strong><br />

medical therapies.<br />

1071 General Poster Session (Board #23F), Sat, 8:00 AM-12:00 PM<br />

Outcomes <strong>of</strong> 47 patients with human immunodeficiency virus infection<br />

treated for breast cancer: A 20-year experience. Presenting Author: Roberto<br />

Enrique Ochoa, University <strong>of</strong> Miami, Miami, FL<br />

Background: Patients (pts) with Human Immunodeficiency Virus (HIV) are<br />

living longer and non-AIDS defining malignancies have been increasingly<br />

reported in these patients. Methods: Retrospective review identified 47 pts<br />

with breast cancer (BC) and HIV who were seen at the University <strong>of</strong> Miami<br />

Sylvester Comprehensive Cancer Center/Jackson Memorial Hospital between<br />

January 1999 and June 2011. Results: Pt characteristics: 46<br />

female, 1 male, mean age 46 years (range 31-65). Race: African <strong>American</strong><br />

79%, Caucasian 21%. Ethnic: Hispanics 14%, non-Hispanics 86%.<br />

Premenopausal 68% postmenopausal 32%. Tumor characteristics: Stage:<br />

Tis 4%, Stage I: 6%,Stage II: 38%, Stage III: 38%, Stage IV: 9%. ER<br />

positive (50%) her-2 positive (15%), Triple negative (21%). HIV characteristics:<br />

36 pts with HIV before or concurrent with the diagnosis <strong>of</strong> BC. 6 pts<br />

diagnosed with HIV within 1 year <strong>of</strong> BC diagnosis. HIV dx date unavailable<br />

in 5 pts. 27% had AIDS. CD4 counts (in cells/�L)were: � 500 (23%);<br />

201-500 (37%), 51 – 200 (20%) � 50 (20%). 15 pts were diagnosed with<br />

BC in preHAART era. Of those dx with BC after 1996, 60% were on HAART.<br />

BC treatment: 43 pts had localized disease. 32 underwent modified radical<br />

mastectomy, 8 breast conservation and 3 pts refused surgery. 26 pts<br />

received curative or palliative chemotherapy. Complications <strong>of</strong> BC treatment:<br />

serious side effects were reported in 11 (42%) including neutropenic<br />

fever/sepsis (10 pts), ARDS (1 pt). Zoster infection was reported in 12% <strong>of</strong><br />

the pts. 3 patients developed rapidly progressive and fatal AIDS within 6<br />

months <strong>of</strong> completion <strong>of</strong> chemotherapy. Survival: See Table. Conclusions:<br />

BC in patients with HIV infection spans the spectrum <strong>of</strong> BC presentations.<br />

Hormonal therapy, surgery and radiation therapy were well tolerated.<br />

Infectious complications were common in patients treated with chemotherapy<br />

and routine use <strong>of</strong> growth factors and prophylactic acyclovir should<br />

be considered.<br />

Alive (% pts) Died BC (% deaths) Died HIV (% deaths)<br />

Tis 3 1 (50) 1 (50)<br />

Stage I 3 (100) 0 0<br />

Stage II 8 (50) 3 (38) 4 (50)<br />

Stage III 3 (28) 4 (67) 2 (33)<br />

Stage IV 0 4 (100) 0<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


1072 General Poster Session (Board #23G), Sat, 8:00 AM-12:00 PM<br />

Prognostic impact <strong>of</strong> weight change during chemotherapy. Presenting<br />

Author: Nikola S. Kasprowicz, Department <strong>of</strong> Gynecology and Obstetrics,<br />

Heinrich Heine University, Duesseldorf, Germany<br />

Background: Besides established prognostic factors such as tumor size or<br />

nodal status, individual host factors <strong>of</strong> the patient such as obesity, physical<br />

activity and diet seem to modulate the course <strong>of</strong> breast cancer (BC) as well.<br />

However, the specific impact <strong>of</strong> weight change during adjuvant chemotherapy<br />

remains unclear. The aim <strong>of</strong> this analysis was to evaluate the<br />

influence <strong>of</strong> weight change during chemotherapy on BC survival in a large,<br />

multi-center prospectively randomized trial. Methods: The ADEBAR trial<br />

compares two anthracycline based adjuvant chemotherapy regimen in<br />

patients (pts) with lymph node positive ( � 3 positive) early BC: 4 x<br />

epirubicin (E) 90 mg/m2 � cyclophosphamide (C) 600 mg/m2 q3w<br />

followed by 4 x docetaxel 100 mg/m2 q3w versus 6xE60mg/m2 � 5-FU<br />

500 mg/m2 d1�d8 and C 75 mg/m2 d1-14 q4w. Weight was measured<br />

before each cycle. The weight before the 1st and the 6th cycle was assessed.<br />

Significant weight change was defined as increase or decrease <strong>of</strong> � 5% <strong>of</strong><br />

the initial weight. Overall survival (OS), disease free survival (DFS), and BC<br />

specific survival (BCSS) were assessed by Kaplan-Meier analysis. Results:<br />

In total, 1502 pts were included in the study. 1177 <strong>of</strong> them completed 6<br />

cycles <strong>of</strong> chemotherapy. Out <strong>of</strong> the 350 pts (29.7%) who changed weight<br />

142 pts (12.1%) lost and 208 pts (17.7 %) gained weight. There was a<br />

significant correlation between weight change and menopausal status<br />

(p�0.0001), indicating that more premenopausal pts gained and postmenopausal<br />

pts lost weight. All other tumor characteristics were similarly<br />

distributed across the groups. Pts with weight change � 5% showed a<br />

significantly worse outcome with respect to OS (p � 0.0028) and BCSS (p<br />

� 0.0258). A difference in DFS was not observed (p � 0.1917). The<br />

difference in OS was limited to pts who lost weight (p � 0.0008), whereas<br />

pts with weight gain have no significant different OS (p � 0.1246) in<br />

comparison to pts with constant weight. Conclusions: Our results suggest<br />

that weight loss during anthracycline-based treatment <strong>of</strong> early stage BC is<br />

associated with poorer OS. While weight normalization has shown beneficial<br />

effects in lifestyle intervention trials, patients should be advised not to<br />

lose weight during chemotherapy.<br />

1074 General Poster Session (Board #24A), Sat, 8:00 AM-12:00 PM<br />

Impact <strong>of</strong> the delivery <strong>of</strong> adjuvant anthracycline-based nontaxane chemotherapy<br />

schedules on the outcome <strong>of</strong> breast cancer patients: Results from a<br />

retrospective database analysis. Presenting Author: Isabel Chirivella, Hospital<br />

Clinico Universitario, Valencia, Spain<br />

Background: A dose-response effect has previously been observed with CMF<br />

and anthracycline-based schedules. The objective <strong>of</strong> this post-hoc analysis<br />

was to give additional information on the impact <strong>of</strong> chemotherapy delivery<br />

on patients’ outcome. Methods: Selection criteria were to have early breast<br />

cancer (stage I-IIIA) from Jan 1980 to Dec 2000, to undergo surgery as<br />

primary treatment and to receive an anthracycline-based non-taxane<br />

schedule in the adjuvant setting. G-CSF support was not given to any<br />

patient. Chemotherapy delivery was assessed based on � 2 vs. � 2 delayed<br />

cycles, � 15 vs. �15 delayed days and � 85% vs. � 85% <strong>of</strong> the relative<br />

dose intensity (RDI) given. Patients’ outcomes were assessed based on 5and<br />

10-year disease-free survival (DFS) and overall survival (OS) rates.<br />

Results: 793 breast cancer patients with a median age <strong>of</strong> 51 years (21-79)<br />

were analyzed <strong>of</strong> whom 27% had stage IIIA and 23% had stage I disease.<br />

As shown in the table, 5- and 10-year rates <strong>of</strong> DFS and OS significantly<br />

improved when adjuvant chemotherapy was optimally delivered. Conclusions:<br />

The dose-response effect is a key factor that should be taken into account<br />

when an anthracycline-based non-taxane chemotherapy is administered in<br />

the adjuvant setting. Delays and/or reductions <strong>of</strong> chemotherapy should be<br />

avoided in order to achieve the maximal benefit for the patient. This study<br />

was partially funded by Amgen SA.<br />

At 5 years,<br />

% (95% CI) p value<br />

DFS<br />

Overall 79 (76-82) 66 (63-70)<br />

> 2 delayed cycles, n�324<br />

73 (68-78)<br />

� 0.001 56 (51-62)<br />

< 2 delayed cycles, n�469<br />

84 (80-87)<br />

74 (69-78)<br />

> 15 delayed days, n�233<br />

72 (66-78)<br />

� 0.001 55 (48-61)<br />

< 15 delayed days, n�559<br />

82 (79-86)<br />

72 (67-76)<br />

< 85% <strong>of</strong> RDI, n�93<br />

73 (64-82)<br />

0.066 54 (44-64)<br />

> 85% <strong>of</strong> RDI, n�697<br />

OS<br />

80 (77-83)<br />

68 (64-72)<br />

Overall 90 (89-93) 78 (74-81)<br />

> 2 delayed cycles, n�324<br />

87 (83-90)<br />

0.003 70 (65-75)<br />

< 2 delayed cycles, n�469<br />

93 (91-95)<br />

84 (80-88)<br />

> 15 delayed days, n�233<br />

86 (82-91)<br />

0.012 70 (64-76)<br />

< 15 delayed days, n�560<br />

92 (90-94)<br />

81 (78-85)<br />

85% <strong>of</strong> RDI, n�698<br />

92 (89-94)<br />

80 (76-83)<br />

Breast Cancer—Triple-Negative/Cytotoxics/Local Therapy<br />

At 10 years,<br />

% (95% CI) p value<br />

� 0.001<br />

� 0.001<br />

0.003<br />

� 0.001<br />

� 0.001<br />

0.002<br />

67s<br />

1073 General Poster Session (Board #23H), Sat, 8:00 AM-12:00 PM<br />

Phase III trial <strong>of</strong> first-line treatment with gemcitabine plus docetaxel versus<br />

gemcitabine plus paclitaxel in women with metastatic breast cancer<br />

(MBC): A comparison <strong>of</strong> different schedules and treatment. Presenting<br />

Author: Lucia Del Mastro, IRCCS AOU San Martino - IST, Genoa, Italy<br />

Background: Weekly (W) administration <strong>of</strong> Gemcitabine�Docetaxel (G�D)<br />

or Gemcitabine�Paclitaxel (G�P) at low doses may be associated with<br />

greater efficacy and/or lower toxicity compared to the standard 3-weekly<br />

(3W) schedule. Methods: Patients (pts) (360) with MBC, that relapsed after<br />

one adjuvant/neoadjuvant regimen containing an anthracycline (unless<br />

contraindicated) that was completed for at least 12 months, were to be<br />

randomized equally to a) D 75mg/m2 on Day1 � G 1000mg/m2 on Days1/8<br />

q3W ; b) P 175mg/m2 on Day1 � G 1250mg/m2 on Days1/8 q3W ; c) D<br />

30mg/m 2<br />

� G 800mg/m2 on Days1/8/15 qW; or d) P 80mg/m2 � G<br />

800mg/m2 on Days1/8/15 qW. Primary endpoint was time to progression<br />

(TTP). Secondary endpoints were overall survival (OS), overall response rate<br />

(ORR) and overall toxicity (T). Results: Due to slow accrual rate, a futility<br />

analysis was performed to evaluate the chance <strong>of</strong> observing a significant<br />

result in favour <strong>of</strong> the alternative hypothesis. The results from this led to<br />

early study termination. 241 pts [median age (range) 57.0(31-77) years]<br />

were enrolled and randomized to: 3W G�D 60(24.9%); 3W G�P<br />

64(26.6%); W G�D 58(24.1%); W G�P 59(24.5%), <strong>of</strong> which 18(30.0%),<br />

24(37.5%), 14(24.1%) and 19(32.2%) pts respectively completed the<br />

study protocol (i.e. received 6 cycles, extended up to 10 for partial/<br />

complete responders). Median TTP [months(95%CIs)] was 8.33 (6.19-<br />

10.16) in W and 7.51 (5.93-8.33) in 3W, with 86.3% <strong>of</strong> pts progressed in<br />

each schedule. OS did not significantly differ between treatments and<br />

schedules. ORR was comparable between D and P, while it was higher in W<br />

than in 3W (50.4% vs. 33.1%; odds ratio 0.44 [95%CI: 0.26-0.75],<br />

p�0.003). Grade 3/4 Ts were 69.2% and 71.9% <strong>of</strong> pts in D and P and<br />

showed a higher trend in 3W (75.2%) than in W (65.8%). Neutropenia was<br />

the most frequent grade 3/4 T (51.7% and 44.7% in D and P; 39.3% and<br />

56.5% in W and 3W). Conclusions: No substantial differences between<br />

treatments were observed in safety and efficacy. W might be associated<br />

with lower grade 3/4 Ts and possibly with a better tumour response<br />

compared to 3W. These results should be interpreted with caution due to<br />

early termination <strong>of</strong> the study.<br />

1075 General Poster Session (Board #24B), Sat, 8:00 AM-12:00 PM<br />

Cancer stem cell markers in locally advanced breast cancer. Presenting<br />

Author: Daniel G. Tiezzi, University <strong>of</strong> São Paulo, Ribeirão Preto, Brazil<br />

Background: the expressions <strong>of</strong> CD44/CD24, CXCR4 and ABCG2 have been<br />

reported as potential breast cancer stem-like cell (CSLC) markers. The<br />

association between the quantity <strong>of</strong> CSLCs and the response to neoadjuvant<br />

chemotherapy (NACT) remains unclear. Methods: we prospectively analyzed<br />

the expression <strong>of</strong> CD44/CD24, CXCR4 and ABCG2 in 20 patients with<br />

locally advanced or metastatic (LAMBC) invasive ductal carcinomas <strong>of</strong> the<br />

breast subjected to NACT. The mammosphere assay (Mammocult) was<br />

studied in 10 samples. Patients’ mean age was 55.6 � 8.2 yo. According to<br />

clinical stage (CS), 5 patients were IIb, 4 - IIIa, 8 - IIIb and 3 - IV. The mean<br />

clinical tumor diameter was 6.3 � 2.8cm. The ER, PgR and HER2 positive<br />

expression rates were 50%, 45% and 50%, respectively. Ten patients were<br />

treated with EC-T, eight were treated with EC-TH (HER2�) and two were<br />

treated with FEC75 combination as NACT. The median percentage CD44� /<br />

CD24- , CXCR4� , ABCG2� and ESA� cells within Lin- cells were determined<br />

by flow cytometry in fresh sampled tumors after tissue digestion. The<br />

relationship between flow cytometry analyses and clinical and pathological<br />

response to therapy was analyzed. Results: complete clinical response<br />

(cCR) and complete pathological response (pCR) was observed in 9 (45%)<br />

and 5 (25%) patients. We did not observe a significant association between<br />

pCR and ER, PgR or HER2 expression. We observed and association<br />

between the pCR with percentage <strong>of</strong> ABCG2� cells within the tumor and<br />

with the number <strong>of</strong> mammospheres. No correlation between pCR and<br />

CD44� /CD24- cell population within the tumor was observed. The median<br />

percentage <strong>of</strong> ESA� /Lin- /ABCG2� cells within the tumor in pCR patients<br />

was 0.6% and 3.5% in patients with no pCR (p� 0.02). The median<br />

number <strong>of</strong> sphere formation was 5/100 cells and 0.9/1000 cells in pCR<br />

and non-pCR patients, respectively (p� 0.02). Interestingly, there was a<br />

positive correlation between ABCG2 expression and the number <strong>of</strong> mammosfere<br />

formation (r� 0.66; p� 0.03). This correlation was not significant<br />

comparing to CD44�/CD24- cells or CXCR4. Conclusions: the percentage<br />

<strong>of</strong> ABCG2� cancer cells within the tumor and the number <strong>of</strong> mammosphere<br />

formation are predictive factors for pCR in LAMBC patients subjected to<br />

NACT. ABCG2 is a potential marker for CSLCs.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


68s Breast Cancer—Triple-Negative/Cytotoxics/Local Therapy<br />

1076 General Poster Session (Board #24C), Sat, 8:00 AM-12:00 PM<br />

EEG for evaluation <strong>of</strong> “chemobrain.” Presenting Author: Halle C. F. Moore,<br />

Cleveland Clinic, Cleveland, OH<br />

Background: Cognitive impairment is a poorly understood and worrisome<br />

potential complication <strong>of</strong> adjuvant chemotherapy (CT). We sought to<br />

evaluate electroencephalography (EEG) as a means to measure neurophysiologic<br />

function in women receiving CT for early breast cancer. Methods:<br />

Women planning to undergo CT for operable breast cancer and age-similar<br />

controls were evaluated at baseline, during CT and at 1 year with<br />

neurophysiologic assessments. Testing included a brief fatigue inventory<br />

(BFI), brief mental fatigue assessment (BMF), Processing Speed Index<br />

(PSI) derived from Digit Symbol Coding and Symbol Search subtests <strong>of</strong> the<br />

Wechsler Adult Intelligence Scale, and a sustained elbow flexion physical<br />

task (PT). EEG recordings were obtained at rest and after the cognitive and<br />

physical tasks. Data were analyzed using repeated measures <strong>of</strong> analysis <strong>of</strong><br />

variance. Results: Eight patient/control pairs completed baseline and<br />

on-treatment evaluations; 7 pairs also completed the 1 year assessment (1<br />

pair withdrawn due to a second malignancy). Subjective mental fatigue<br />

measured by BMF is similar for patients and controls at baseline but BMF<br />

scores increase significantly during CT for patients relative to controls<br />

(p�0.033), recovering to no difference at one year. Differences in PSI are<br />

not observed between patients and controls or at the different time points.<br />

BFI scores are greater in patients at all 3 time points but endurance on the<br />

PT is no different from controls. During chemotherapy EEG total spectrum<br />

amplitudes in patients are greater than in controls at rest (p�0.05) and<br />

following both the cognitive (p�0.001) and physical (p�0.001) tasks.<br />

EEG activity prior to chemotherapy and at one year is not different between<br />

patients and controls. For patients but not controls EEG readings after the<br />

cognitive task demonstrate greater amplitude than pre-task readings during<br />

the time <strong>of</strong> CT treatment only (p�0.012) with a similar trend seen for the<br />

physical task (p�0.06). Conclusions: Patient-perceived mental and physical<br />

fatigue during chemotherapy correspond to significant changes in EEG<br />

brain activity patterns but not to cognitive testing or physical endurance<br />

testing. EEG may <strong>of</strong>fer a sensitive means to measure alterations in brain<br />

function associated with CT.<br />

1078 General Poster Session (Board #24E), Sat, 8:00 AM-12:00 PM<br />

Comparing the outcome between multifocal, multicentric, and bilateral<br />

breast cancer and the impact <strong>of</strong> guideline-adherent adjuvant treatment: A<br />

retrospective multicenter cohort study <strong>of</strong> 5,308 patients. Presenting<br />

Author: Lukas Schwentner, Department <strong>of</strong> Gynecology and Obstetrics<br />

University Ulm, Ulm, Germany<br />

Background: Beside unifocal-unilateral (UU) breast cancer (BC) there are<br />

several subtypes including multifocal, multicentric and bilateral BC. This<br />

study tries to answer the following questions:(1) Does localization (multifocal/multicentric/bilateral)<br />

influence outcome concerning BC mortality? (2)<br />

Is there an impact <strong>of</strong> guideline-adherent adjuvant treatment in these BC<br />

subtypes? Methods: This German multi-center retrospective cohort study<br />

called BRENDA included 5277 patients obtained from 1992 until 2005.<br />

The definition <strong>of</strong> guideline adherence was based on the German national S3<br />

breast cancer guideline (2004). Results: 4085 (77.4%) were UU, 698<br />

(13.2%) multifocal, 282 (5.3%) multicentric and 212 (4.0%) bilateral<br />

BC. RFS in multifocal [p�0.003; HR�1.35 (95% CI: 1.11-1.65)],<br />

multicentric [p�0.001; HR�1.76 (95% CI: 1.31-2.34)] and bilateral<br />

[p�0.001; HR�2.28 (95% CI: 1.76-2.97)] BC was significantly lower<br />

compared to unilateral-unifocal BC. Concerning OAS we found only a<br />

borderline difference between UU and unilateral-multifocal [p�0.057;<br />

HR�1.22 (95% CI: 0.99-1.48)], but a significant difference between<br />

multicentric [p� 0.018; HR�1.42 (95% CI: 1.06-1.90)] resp. bilateral<br />

[p�0.001; HR�2.87 (95% CI: 2.21-3.74)] and UU-BC. There was a<br />

significant impact by guideline adherent adjuvant therapy [UU: p�0.001,<br />

HR�2.76,95%C.I.:2.25-3.38], [unilateral-multifocal: p�0.001,<br />

HR�2.04,95%C.I.:1.33-3.14], [unilateral-multicentric: p�0.020,<br />

HR�2.13,95%C.I.:1.13-4.01] and [bilateral: p�0.042,<br />

HR�2.10,95%C.I.:1.03-4.31]. After stratifying for 100% guideline adherent<br />

treatment and adjusting for age, tumor size, nodal status and grading<br />

there was no significant difference in RFS/OAS in patients with multifocal<br />

[p�0.282/p�0.610], multicentric [p�0.829/p�0.609] or bilateral BC<br />

[p�0.457/p�0.773] compared to patients with UU-BC. Conclusions:<br />

Patients with multicentric and bilateral BC have primarily a worse prognosis<br />

in terms <strong>of</strong> RFS and OAS. However if guideline adherent adjuvant treatment<br />

was applied it was no more possible to demonstrate significant differences<br />

in survival.<br />

1077 General Poster Session (Board #24D), Sat, 8:00 AM-12:00 PM<br />

Analysis according to prognostic factors in patients (pts) treated with first-line<br />

bevacizumab (BEV) combined with paclitaxel (PAC) for HER2-negative metastatic<br />

breast cancer (mBC) in a routine oncology practice study. Presenting<br />

Author: Iris Schrader, Gynäkolog.-onkolog. Gem.Praxis, Hannover, Germany<br />

Background: 1st-line BEV combined with weekly PAC significantly improves progression-free<br />

survival (PFS) and response rate (RR) vs PAC alone in HER2-negative<br />

mBC, as shown in E2100. We analyzed data from a German routine oncology<br />

practice study <strong>of</strong> 1st-line BEV–PAC according to prognostic factors. Methods: Pts<br />

who had received no prior chemotherapy for mBC received BEV–PAC according to<br />

the European label. Efficacy and safety were documented for up to 1 y (or until<br />

progression, death, or BEV discontinuation if earlier) with additional long-term<br />

follow-up. Efficacy was analyzed in clinically important subgroups. Results: Efficacy<br />

data were available for 818 pts. The median duration <strong>of</strong> follow-up was 11.4 mo. The<br />

composition <strong>of</strong> the pt population with respect to the subgroups below was generally<br />

similar to the population treated in E2100, except for a higher proportion <strong>of</strong> pts with<br />

visceral disease or metastases in �3 organs. RR was very similar across all<br />

subgroups analyzed. Differences in median PFS and OS were generally in line with<br />

the differing prognoses according to clinical characteristics. Conclusions: These data<br />

suggest that 1st-line BEV–PAC is typically associated with median PFS �9 moin<br />

the real-life setting, irrespective <strong>of</strong> baseline characteristics.<br />

PFS OS<br />

Subgroup N RR, % Median, mo p value Median, mo p value<br />

All<br />

Age, y<br />

818 62 9.4 – 20.8 –<br />

>65 262 57 9.2 0.83 20.6 0.99<br />

70 133 57 9.3 0.38 20.6 0.34<br />


1080 General Poster Session (Board #24G), Sat, 8:00 AM-12:00 PM<br />

Baseline comprehensive geriatric assessment to predict for toxicity <strong>of</strong><br />

single-agent chemotherapy in elderly metastatic breast cancer patients:<br />

Results from the OMEGA study <strong>of</strong> the Dutch Breast Cancer Trialists’ Group.<br />

Presenting Author: Marije Hamaker, Diakonessenhuis, Utrecht, Netherlands<br />

Background: A comprehensive geriatric assessment (CGA) systematically<br />

appraises the somatic, psychosocial and functional health status <strong>of</strong> elderly<br />

patients. If CGA can predict toxicity <strong>of</strong> chemotherapy in elderly cancer<br />

patients, this assessment could be useful in deciding on optimal treatment.<br />

In this analysis, we evaluated the association between frailty on CGA or<br />

Groningen Frailty Index (GFI), and grade 3/4 toxicity in metastatic breast<br />

cancer (MBC) patients treated with first-line chemotherapy. Methods: In the<br />

OMEGA study, MBC patients (� 65 years) were randomized between<br />

PEGdoxo 45mg/m2every 4 weeks or capecitabine 2000 mg/m2 on days<br />

1-14 every 3 weeks. Baseline geriatric assessment included functional<br />

status (ECOG performance status (PS), IADL), cognition (MMSE), mood<br />

(GDS), comorbidity (Charlson), polypharmacy and undernutrition (BMI)<br />

and GFI. Frailty on CGA was defined as one or more <strong>of</strong> the following:<br />

IADL�13, MMSE �23, GDS �5, BMI �20, �5 medications or Charlson<br />

�2. GFI score for frailty was �4. Results: In total, 78 patients were<br />

randomized (PEGdoxo 38, capecitabine 40), median age 75 years (range<br />

65-86). ECOG PS was 0-1 in 78% <strong>of</strong> patients and 2-3 in 22%. So far, 72<br />

patients were evaluable for toxicity and baseline CGA. Overall, 50 patients<br />

(70%) were frail on CGA, and 40 (55%) according to GFI. Grade 3/4<br />

toxicity related to chemotherapy was reported in 30 patients (42%) and<br />

50% <strong>of</strong> CGA-frail patients experienced grade 3/4 toxicity, compared to<br />

20% <strong>of</strong> CGA-fit patients (p�0.02). Grade 3/4 toxicity was experienced by<br />

44% <strong>of</strong> GFI-frail patients, compared to 38% <strong>of</strong> GFI-fit patients (p�0.39).<br />

After correcting for type <strong>of</strong> chemotherapy and age, toxicity was associated<br />

with CGA-frailty (odds ratio (OR) 4.00, 95% confidence interval (CI)<br />

1.77-13.59, p�0.03) while GFI-frailty was not associated with toxicity<br />

(OR 1.11, 95% CI 0.85-1.46, p�0.43). Conclusions: In this randomized<br />

study on first-line single-agent chemotherapy in elderly MBC patients,<br />

baseline CGA demonstrated a good predictive value for grade 3/4 toxicity <strong>of</strong><br />

chemotherapy but GFI did not.<br />

1082 General Poster Session (Board #25A), Sat, 8:00 AM-12:00 PM<br />

<strong>Part</strong>icipation in adjuvant clinical breast cancer trials: Is there a difference<br />

in survival compared to guideline adherent adjuvant treatment? A retrospective<br />

multicenter cohort study <strong>of</strong> 5,326 patients. Presenting Author: Achim<br />

Wöckel, Department <strong>of</strong> Gynecology and Obstetrics University Ulm, Ulm,<br />

Germany<br />

Background: <strong>Clinical</strong> trials (CT) usually compare a standard treatment<br />

regime versus innovative new substances or regimens. However participation<br />

in CT is available for only few patients and exclusion criteria is usually<br />

very strict. Therefore this study tries to answer the following questions: (1)<br />

Does participation in adjuvant CT improve survival in breast cancer (BC)?<br />

(2) Is there a difference in survival compared to guideline adherence and<br />

what is the role <strong>of</strong> the other treatments surrounding adjuvant breast cancer<br />

treatment? Methods: This German multi-center [17 participating hospitals<br />

all are certified as breast cancer centers] retrospective cohort study called<br />

BRENDA (BRENDA � quality <strong>of</strong> breast cancer care under evidence-based<br />

guidelines) included 5326 patients obtained from 1992 until 2005. The<br />

definition <strong>of</strong> guideline adherence was based on the German national S3<br />

guideline for diagnosis and treatment <strong>of</strong> breast cancer (2004). Results: 554<br />

(10,4%) patients participated adjuvant clinical trials and 4772 (89,6%)<br />

not. There was a trend towards a significantly impaired RFS [p�0.17;<br />

HR�0.87 (95% CI: 0.71-1.06)] and OAS [p�0.65; HR�0.84 (95% CI:<br />

0.65-1.09)] when comparing participants (PA) versus non-participants<br />

(NPA). When stratifying for guideline adherence (compared to PA-guideline<br />

conform in all adjuvant therapies) the outcome was not different in<br />

NPA-guideline adherent [RFS: p�0.13; HR�1.25 (95% CI: 0.94-1.67)]<br />

[OAS: p�0.83; HR�1.41 (95% CI: 0.96-2.06)]. However survival parameters<br />

were significantly impaired in non-guideline conform PA [RFS:<br />

p�0.005; HR�1.66 (95% CI: 1.16-2.38)] [OAS: p�0.01; HR�1.83<br />

(95% CI: 1.15-2.92)] and non-guideline conform NPA [RFS: p�0,001;<br />

HR�1.70 (95% CI: 1.27-2.26)] [OAS: p�0.002; HR�1.82 (95% CI:<br />

1.24-2.67)]. Conclusions: There is a strong association between guideline<br />

adherence in adjuvant treatment in BC and survival. PA in clinical trials<br />

trended to an improved survival, but only if guideline adherent treatment<br />

was applied. Patients who don�t have access to clinical seem to pr<strong>of</strong>it<br />

substantially <strong>of</strong> guideline adherent adjuvant treatment.<br />

Breast Cancer—Triple-Negative/Cytotoxics/Local Therapy<br />

69s<br />

1081 General Poster Session (Board #24H), Sat, 8:00 AM-12:00 PM<br />

Sequential treatment with epirubicin/cyclophosphamide, followed by docetaxel<br />

versus FEC120 in the adjuvant treatment <strong>of</strong> node-positive breast<br />

cancer patients: Final survival analysis <strong>of</strong> the German ADEBAR phase III<br />

study. Presenting Author: Wolfgang Janni, Universitätsklinikum Düsseldorf,<br />

Department <strong>of</strong> Gynecology and Obstetrics, Duesseldorf, Germany<br />

Background: Based on meta-analytic evidence, taxane containing adjuvant<br />

chemotherapy has been established as standard treatment in breast cancer<br />

(BC). However, in the MA-21 study, adriamycin-cyclophosphamide, followed<br />

by paclitaxel was significantly inferior FEC120. We prospectively<br />

compared a sequential epirubicin-docetaxel chemotherapy regimen to<br />

FEC120. Methods: The ADEBAR study was a multicenter phase III trial<br />

(n�1502) to evaluate whether pts with � 3 axillary lymph node metastases<br />

BC benefit from a sequential anthracycline-docetaxel regimen (E90C–D: 4<br />

cycles epirubicin [E] 90 mg/m2 plus cyclophosphamide [C] 600 mg/m2 q21d followed by 4 cycles docetaxel [D] 100mg/m2 q21d) compared to<br />

dose-intensive anthracycline-containing polychemotherapy (FE120C: 6<br />

cycles E 60 mg/m² d1�8, 5-FU 500mg/m² d1�8 and C 75 mg/m² d 1-14,<br />

q4w). The observation time (median – 95%CI) was 49.5 (47.4 – 51.3) m.<br />

Results: Treatment was stopped prematurely in 3.7% <strong>of</strong> the pts in the<br />

E90C–D arm and in 8.0% in the FE120C arm due to toxicity (p�0.0009).<br />

Antibiotic treatment was given in 10.4% (E90C–D) vs. 19.7% (FE120C), G-CSF support in 39.2% vs 61.4 % and erythropoietin stimulation in 8.7%<br />

vs. 20.0%, respectively (p�0.0001). Haematological toxicity (leucopenia,<br />

neutropenic fever, thrombocytopenia, anemia) was significantly higher in<br />

the FEC-arm. At the time <strong>of</strong> the current analysis, 369 events <strong>of</strong> recurrence,<br />

were observed: 166 events in the FE120C group and 193 in the E90C–D<br />

group. The unadjusted hazard ratio (HR) was 0.877 (95 percent confidence<br />

interval, 0.722 to 1.065; p�0.3819, log-rank test). Overall survival in the<br />

two groups was not significantly different: (131 deaths with FEC vs. 134<br />

with E90C–D (HR 0.996, 0.783-1.267, p�0.9691). Subgroup analyses,<br />

stratifying for tumor size, lymph node involvement, hormone receptor and<br />

HER2-neu status showed no significant difference between the two arms.<br />

Conclusions: Different toxicity pr<strong>of</strong>iles given, hematological toxicity in the<br />

FE120C group was more severe than in the E90C–D. In contrast to AC-P in<br />

earlier studies, EC-Doc provides a feasible and effective option to FEC120.<br />

1083 General Poster Session (Board #25B), Sat, 8:00 AM-12:00 PM<br />

Randomized phase II study <strong>of</strong> two doses <strong>of</strong> pixantrone in patients with<br />

metastatic breast cancer (N1031, Alliance). Presenting Author: Kostandinos<br />

Sideras, Mayo Clinic, Rochester, MN<br />

Background: Pixantrone (Pix) is a novel aza-anthracenedione with structural<br />

similarities to mitoxantrone and promising activity against non-Hodgkin’s<br />

lymphoma. Due to the lack <strong>of</strong> iron binding it is theorized to exhibit less<br />

cardiotoxicity than the anthracyclines. Methods: N1031 is a phase II RCT <strong>of</strong><br />

2 schedules <strong>of</strong> Pix, in pts with MBC. Group A pts received 180mg/m2 IV q3<br />

wks, and group B pts 85mg/m2 IV on days 1, 8, 15 q4 wks. Eligibility<br />

included prior exposure to anthracyclines and/or taxanes, and 1 to 3<br />

regiments in the metastatic setting (minimum <strong>of</strong> 2 if no prior adjuvant<br />

therapy given). Due to lack <strong>of</strong> long term cardiac safety data no more than 12<br />

cycles were allowed. Frequent cardiac imaging was performed per protocol.<br />

Primary endpoint was RR and secondary endpoints included PFS, OS,<br />

safety, and QOL. Planned sample size was 25 pts per group. Results: In<br />

total 46 pts were evaluable (23 per group), mean age 55.5 yrs (range<br />

38-79), 37% PS 0, 52% PS 1, and 11% PS 2. 80% <strong>of</strong> pts had prior<br />

exposure to doxorubicin, 72% had prior (neo)-adjuvant therapy, 76% were<br />

ER� and 57% received prior HT. Number <strong>of</strong> prior metastatic regiments<br />

was: 1 (28%), 2 (61%) and 3 (11%). Most common adverse events (%) <strong>of</strong><br />

any grade were: alopecia (74), anemia (74), fatigue (85), nausea (67), ANC<br />

decrease (87), and skin disorder (41). Grade 3-4 adverse events (%) at<br />

least possibly attributed to Pix and occurring in at least 2 pts were: ANC<br />

decrease (57), fatigue (9), increased AST (4%). One pt from each group<br />

(4%) had a grade 3 decrease in EF. There were no major differences<br />

between the two groups except for more oral mucositis in group A (35% vs<br />

4%). Median number <strong>of</strong> cycles was 3 in group A (range 1-12) and 2 in<br />

group B (range 1-8). There was only 1 confirmed tumor response per group<br />

(4%,95% CI: 0.1-22%) prompting early termination <strong>of</strong> the trial. The<br />

median PFS was 2.7 mo (95% CI: 1.8-3.8), and the median OS was 8.9 mo<br />

(95% CI: 7.5-N/A). Conclusions: Pixantrone has insufficient activity in<br />

patients with MBC exposed to prior anthracyclines and/or taxanes. Adverse<br />

events were similar to prior experience with Pix. There were no major<br />

differences between the 2 schedules <strong>of</strong> administration. There was no<br />

significant cardiac toxicity seen in this trial. Correlative studies are<br />

underway.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


70s Breast Cancer—Triple-Negative/Cytotoxics/Local Therapy<br />

1084 General Poster Session (Board #25C), Sat, 8:00 AM-12:00 PM<br />

Reduction <strong>of</strong> ovarian reserve in young early breast cancer patients: First<br />

data <strong>of</strong> a prospective cohort trial. Presenting Author: Lea Sanders, OB/GYN,<br />

University <strong>of</strong> Kiel, UKSH, Kiel, Germany<br />

Background: Breast cancer is the most common malignancy in premenopausal<br />

women. Sideeffects <strong>of</strong> chemotherapy are well known nevertheless<br />

the precise effects on ovarian function are inadequately studied by now.<br />

Premenopausal women undergoing chemotherapy are at risk for symptoms<br />

<strong>of</strong> sexual hormone deficiency and impaired fertility. Searching for predictive<br />

parameters <strong>of</strong> ovarian reserve after chemotherapy this prospective<br />

cohort study has been set up. Methods: 36 young patients with primary<br />

breast cancer have been included in this trial after written consent (April<br />

2010 to November 2011) and after the study was approved by the local<br />

review board. All women were premenopausal (� 46 years). They all<br />

received anthracycline based “A” neo- or adjuvant chemotherapy (as FEC)<br />

or combinations with taxanes “T” ( as TAC or FEC/Doc). Before and 6 and<br />

12 months after initiation <strong>of</strong> chemotherapy age and chemotherapy related<br />

changes in hormone (LH, FSH, E2 and Anti-Müllerian hormone) levels,<br />

antral follicel count and amenorrhea as parameters <strong>of</strong> endocrine function<br />

and fertility were assessed.The additional impact <strong>of</strong> parity, BMI and<br />

nicotine use on ovarian reserve was also evaluated. Results: There is a<br />

correlation <strong>of</strong> antral follicle count before and 1 year after chemotherapy and<br />

a negative correlation <strong>of</strong> age and follicle count before and after chemotherapy<br />

(n.s.). This analysis shows that patients receiving “T” compared to<br />

those with “A” have a significant increase <strong>of</strong> LH (p�0.025) and FSH<br />

(mean:24 vs.59 IU/l, p�0.021) between visit 1 and 3. The type <strong>of</strong><br />

chemotherapy has no influence on antral follicle count and AMH levels<br />

within the first 3 visits. BMI is negatively correlated with AMH at all time<br />

points (n.s.). BMI, nicotine abuse and age have no influence on the<br />

duration <strong>of</strong> amenorrhea, whereas patients with �T� showed 12 months <strong>of</strong><br />

amenorrhea instead <strong>of</strong> 9 months in patients with �A� (n.s.). Conclusions: Our<br />

study will contribute to a better understanding and prediction <strong>of</strong> ovarian<br />

reserve <strong>of</strong> young early breast cancer patients undergoing chemotherapy.<br />

The 12 months follow up data suggest to <strong>of</strong>fer fertility preserving measures<br />

before chemotherapy especially to patients planned for taxane containing<br />

chemotherapy.<br />

1086 General Poster Session (Board #26B), Sat, 8:00 AM-12:00 PM<br />

Amrubicin as second- or third-line treatment for patients with HER2negative<br />

metastatic breast cancer (MBC): A phase II trial <strong>of</strong> the Sarah<br />

Cannon Research Institute (SCRI). Presenting Author: John H. Barton,<br />

Tennessee Oncology, PLLC/Sarah Cannon Research Institute, Nashville,<br />

TN<br />

Background: Anthracyclines demonstrate significant activity in breast<br />

cancer, but the potential for cardiotoxicity is dose-limiting. Amrubicin is a<br />

novel anthracycline with broad-spectrum preclinical activity and low<br />

potential for cardiotoxicity. We present phase II results from a phase I/II<br />

trial <strong>of</strong> amrubicin as second/third- line therapy for HER2- negative MBC.<br />

Methods: Women with measurable HER2-negative MBC with 1 or 2 prior<br />

chemotherapy regimens for metastatic disease and normal LVEF were<br />

eligible. Prior anthracycline- containing adjuvant therapy was allowed.<br />

Amrubicin 110 mg/ m2 IV every 3 weeks was administered until disease<br />

progression or intolerable toxicity. Tumor assessments were performed<br />

every 6 weeks and LVEF assessments every 12 weeks. The primary<br />

endpoint was progression free survival (PFS); a median PFS � 4.5 months<br />

was considered a study result meriting further development <strong>of</strong> amrubicin.<br />

Results: 48 evaluable patients (pts) were treated from 1/2010 to 9/2011.<br />

Baseline characteristics included median age 57; 23% were triplenegative;<br />

33% had 2 prior chemotherapy regimens for MBC; 38% had<br />

anthracycline- containing adjuvant therapy. Median treatment duration<br />

was 6 weeks (2 cycles), range 1- 12� cycles. 8 pts (17%) had objective<br />

RECIST responses (1 CR, 7 PR); 5 <strong>of</strong> the 8 responders had received<br />

anthracycline-containing adjuvant therapy. 24 additional pts (50%) had<br />

stable disease at first reevaluation. The median PFS for all patients was 2.8<br />

months (95% CI 1.6- 4.0 months); median PFS was similar for pts with 1<br />

vs 2 previous regimens for MBC (95% CI 2.5 vs 4.0 months). 24% <strong>of</strong> pts<br />

were progression-free at 6 months. Neutropenia was the most common<br />

grade 3/4 toxicity (63%; 6% febrile neutropenia). No grade 3/4 nonhematologic<br />

toxicity occurred in � 5% pts. No cardiotoxicity occurred. Only<br />

1 pt discontinued amrubicin due to toxicity (grade 2 fatigue). Conclusions:<br />

Amrubicin had good tolerability, no cardiotoxicity and was active as a<br />

second/third-line treatment for HER2- negative MBC, including pts previously<br />

treated with adjuvant anthracyclines. The median PFS was comparable<br />

to other standard single agents in the MBC setting.<br />

1085 General Poster Session (Board #26A), Sat, 8:00 AM-12:00 PM<br />

Methallotionein expression and outcome in patients with metastatic breast<br />

cancer (MBC). Presenting Author: Jorge Arturo Rios-Perez, University <strong>of</strong><br />

Pittsburgh Cancer Institute, Pittsburgh, PA<br />

Background: Platinum-based agents are important components <strong>of</strong> therapy<br />

<strong>of</strong> metastatic breast cancer (MBC) and triple negative breast cancer. Their<br />

use can be limited by development <strong>of</strong> resistance. Metallothioneins (MT) are<br />

low molecular weight proteins believed to bind bivalent metal ions such as<br />

platinum and zinc. MT expression has been associated with decreased<br />

survival in breast cancer patients. A proposed mechanism confers resistance<br />

to platinum-based agents by their inactivation or limitation <strong>of</strong> their<br />

activity by MT binding. Methods: MT expression in 99 women with MBC<br />

(selected at random from our database <strong>of</strong> 800 women with MBC) was<br />

determined from primary breast cancer tissue (n�80) or metastatic tissue<br />

n�19). MT expression was determined by immunohistochemistry, and<br />

graded as negative, weak, moderate or strong. <strong>Clinical</strong> data was obtained<br />

through our database and supplemented by chart review. Overall survival<br />

from breast cancer diagnosis (OS), progression free survival for first<br />

metastastic regimen (PFS), and time from first metastasis to death or last<br />

update (metastatic survival, MS), were calculated through December 2011<br />

using the log rank test. Results: Consistent with prior studies, moderate to<br />

strong MT expression was associated with decreased 5-year OS (p�.03).<br />

There was no correlation between MT expression and PFS or MS in this<br />

cohort. Surprisingly, MT expression at any degree was strongly associated<br />

with better MS in patients with MBC that received carboplatin-based<br />

regimens in the first line (n�25, p�.0005) or at any line (n�41,<br />

p�.0437). Conclusions: Consistent with prior studies, MT expression was<br />

associated with decreased survival in patients with MBC. Surprisingly, MT<br />

expression was associated with longer MS in patients with MBC that<br />

received carboplatin. These findings are inconsistent with the hypothesis<br />

that MT expression causes chemoresistance to platinum based agents in<br />

patients with metastatic breast cancer. Further studies are needed to<br />

elucidate the mechanisms behind these findings.<br />

1087 General Poster Session (Board #26C), Sat, 8:00 AM-12:00 PM<br />

BCL2 protein in prediction <strong>of</strong> relapse in triple-negative breast cancer<br />

(TNBC) treated with adjuvant anthracycline-based chemotherapy. Presenting<br />

Author: Katerina Bouchalova, Laboratory <strong>of</strong> Experimental Medicine,<br />

Institute <strong>of</strong> Molecular and Translational Medicine, Palacky University,<br />

Olomouc, Czech Republic<br />

Background: Preclinical data show an association <strong>of</strong> BCL2 expression and<br />

resistance to anthracyclines. The absence <strong>of</strong> BCL2 expression in prechemotherapy<br />

samples is associated with a higher probability <strong>of</strong> pathological<br />

complete response to neoadjuvant doxorubicin-based chemotherapy. The<br />

aim <strong>of</strong> our research is identification <strong>of</strong> markers predicting sensitivity to<br />

adjuvant treatment in TNBC. Here we focus on BCL2 protein as a putative<br />

predictor <strong>of</strong> sensitivity to adjuvant anthracycline-based chemotherapy. The<br />

objective was to determine whether BCL2 expression predicts relapse in<br />

TNBC patients treated with anthracycline-based regimens. Methods: The<br />

study included 187 patients with TNBC, 178 <strong>of</strong> whom were treated with<br />

adjuvant chemotherapy (164 had anthracyclines). BCL2 analysis was<br />

performed using IHC, proportion score and intensity score were counted.<br />

The data were analysed with s<strong>of</strong>tware Statistica and R. Results: High BCL2<br />

expression predicts poor relapse free survival (RFS) in patients treated with<br />

adjuvant anthracycline-based regimens (logrank p�.035, hazard ratio, HR<br />

2.37, 95%CI 1.04-5.41). High BCL2 predicts trend to poor overall survival<br />

(OS) in patients treated with adjuvant anthracycline-based regimens<br />

(logrank p�0.075, HR 2.31, 95%CI 0.90-5.97). In univariate analysis <strong>of</strong><br />

anthracycline treated patients, stage (RFS p�.0004, OS p�.0005), size<br />

(RFS p�.003, OS p�.00009) and nodal status (RFS p�0.018, OS<br />

p�.028) were associated with outcome, as well. In multivariate analysis <strong>of</strong><br />

athracycline treated patients, BCL2, size and nodal status had an independent<br />

predictive significance for both RFS (p�.005, p�.056, p�.003)<br />

(logrank test, p�0.0004) and OS (p�.014, p�.006, p�.012) (logrank<br />

test p�0.0007). Conclusions: This study is the first to prove that high BCL2<br />

expression predicts poor outcome in TNBC treated with adjuvant anthracycline-based<br />

chemotherapy. BCL2 expression could facilitate decision<br />

making on adjuvant treatment in TNBC patients and its assessment should<br />

be included in standard diagnostics. In patients with high BCL2 expression<br />

other types <strong>of</strong> adjuvant treatment should be considered. Grants: IGA<br />

NS10286,IGA NS10357-3 and Biomedreg CZ.1.05/2.1.00/01.0030.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


1088 General Poster Session (Board #27A), Sat, 8:00 AM-12:00 PM<br />

Quantitative measures <strong>of</strong> FDG PET after neoadjuvant chemotherapy to<br />

predict breast cancer patient survival. Presenting Author: Erin-Siobhain R.<br />

Currin, University <strong>of</strong> Washington, Department <strong>of</strong> Medicine, Seattle, WA<br />

Background: In patients with locally advanced breast cancer (LABC),<br />

qualitative FDG positivity following neo-adjuvant chemotherapy (NC) has<br />

been shown to be inversely associated with survival (Emmering, Annals<br />

Oncol, 2008). We investigated quantitative measures <strong>of</strong> post-therapy FDG<br />

uptake, namely standardized uptake value (SUV) and glycolytic flux (Ki), as<br />

predictors <strong>of</strong> breast cancer survival. Methods: Forty-seven patients with<br />

LABC underwent dynamic FDG PET scans close to or at the end <strong>of</strong> NC and<br />

prior to surgical resection. Post-therapy FDG uptake at the primary tumor<br />

site was measured by mean SUV from 45-60 minutes after FDG injection,<br />

maximum SUV (SUVmax) from 50-55 minutes, and FDG glycolytic flux<br />

(Ki). Pathologic response (PR) was assessed for at the time <strong>of</strong> surgical<br />

resection. Cox proportional hazards models were used to estimate associations<br />

between log-transformed measures <strong>of</strong> post-therapy FDG uptake, PR<br />

and outcome. Results: Median SUVmax was 1.9 (0.9 – 9.2) and median Ki<br />

was 2.2 (0.02 – 47.7) mL/min/g. Median follow-up for relapse was 5.7<br />

years with 11 events and 6.3 years for survival with 10 deaths. PR was not<br />

significant for DFS (p � .39) or OS (p� .48). Post-therapy FDG uptake<br />

measures showed a statistically significant ability to predict survival.<br />

SUVmax predicted DFS (p�0.02) and OS (p�0.01). Ki was associated<br />

with DFS (p �0.01) and OS (p �0.01). PET measured hazard ratios were<br />

not attenuated in multivariate analysis controlling for known prognostic<br />

markers such as primary tumor PR and nodal status. However, multivariate<br />

survival models appeared highly influenced by one patient with the shortest<br />

survival time (1.3 years) and highest SUVmax and Ki. Without this patient,<br />

Ki remained a borderline independent predictor <strong>of</strong> DFS (p� .08) and OS<br />

(p� .07), but SUVmax was no longer significant for DFS (p� .32) or OS<br />

(p�0.26). Conclusions: Our analysis suggests that quantitative measures <strong>of</strong><br />

post-therapy FDG PET provide information beyond PR for predicting which<br />

LABC patients are at highest risk for relapse and death. This information<br />

may be useful in directing post-surgery treatment. Supported by NIH grants<br />

CA42045, CA138293, and CA148131.<br />

1090 General Poster Session (Board #27C), Sat, 8:00 AM-12:00 PM<br />

Circulating tumor cells in metastatic breast cancer: Are they a strong and<br />

independent predictor <strong>of</strong> poor progression-free and overall survival? Presenting<br />

Author: Markus Wallwiener, Department <strong>of</strong> Obstetrics and Gynaecology,<br />

University <strong>of</strong> Heidelberg, Heidelberg, Germany<br />

Background: Circulating tumor cells (CTCs) are detected in 30–60% <strong>of</strong><br />

patients with metastatic breast cancer (MBC). The aim <strong>of</strong> this prospective<br />

multi-center study was to evaluate the impact <strong>of</strong> CTCs on progression free<br />

survival (PFS) and overall survival (OS) in a large cohort <strong>of</strong> 486 patients<br />

with progressive metastatic disease. Methods: CTC levels were determined<br />

for 486 patients at nine German University Breast Cancer Centers between<br />

12/2007 and 06/2011. Samples <strong>of</strong> 7.5 ml blood were taken before<br />

initiation <strong>of</strong> a new line <strong>of</strong> therapy and CTCs were enumerated using the<br />

CellSearch System (Veridex LLC, Raritan, NJ, USA). CTC status (� 5 CTCs<br />

vs. � 5 CTCs per 7.5 ml blood) was assessed as a prognostic factor for PFS<br />

and OS using univariate (log-rank test) and multivariate (Cox regression<br />

model) statistical methods. Results: CTCs were detected in 205/486 (42%)<br />

patients. The median CTC count was 2 (range 0–6380) per 7.5 ml blood.<br />

The presence <strong>of</strong> � 5 CTCs/7.5 ml blood did not correlate with any <strong>of</strong> the<br />

established clinicopathological factors except estrogen receptor status (p<br />

� 0.038). PFS and OS were both significantly shorter in patients with � 5<br />

CTCs/7.5 ml than in those with � 5 CTCs/7.5 ml blood. PFS was 5.0 [95%<br />

CI 4.1–5.8] months vs.7.6 [95% CI 5.9–9.3] months, p � 0.001; and OS<br />

was 15.0 [95% CI 13.5–16.5] months vs. 18.3 [95% CI 17.4–19.2]<br />

months, p � 0.001. In the multivariate analysis considering all clinicopathological<br />

factors and the CTC status, independent predictors <strong>of</strong> reduced OS<br />

and PFS were site <strong>of</strong> metastasis (visceral vs. bone), number <strong>of</strong> metastatic<br />

sites (multiple sites vs. one site), and CTC status. Conclusions: The<br />

presence <strong>of</strong> � 5 CTCs/7.5 ml blood is a strong and independent predictor <strong>of</strong><br />

poor PFS and OS in patients with MBC.<br />

Breast Cancer—Triple-Negative/Cytotoxics/Local Therapy<br />

71s<br />

1089 General Poster Session (Board #27B), Sat, 8:00 AM-12:00 PM<br />

Evaluation <strong>of</strong> overall survival (OS), progression-free survival (PFS), or time<br />

to progression (TTP) in systematic review <strong>of</strong> randomized clinical trials<br />

(RCT) in patients with metastatic breast cancer (MBC). Presenting Author:<br />

Terence Mukonje, Medical College <strong>of</strong> Wisconsin, Milwaukee, WI<br />

Background: Limited data exists to establish if the introduction <strong>of</strong> newer<br />

agents has been associated with a trend towards an improved OS over time<br />

in women with MBC. Methods: Trough a computer-based systematic search<br />

<strong>of</strong> PubMed we identified RCT that treated patients with MBC published<br />

between 1980 and 2011. Trials that compared systemic chemotherapy for<br />

MBC, and reported a median OS were included. We excluded trials that use<br />

concomitant hormonal treatment, investigated only immunotherapy, or if<br />

they enrolled only responders to an initial regimen. The data abstracted<br />

included: year <strong>of</strong> publication, number <strong>of</strong> patients, regimens used, number<br />

<strong>of</strong> patient treated as 1st line vs. refractory patients, median PFS, TTP and<br />

OS. Linear regression analysis was used to establish trends. Results: An<br />

initial searched revealed 5485 publications, 134 RCT that enrolled<br />

38,090 patients fulfilled our entry criteria and were included. First line<br />

therapy was studied in 99 trials and 35 evaluated mostly refractory<br />

patients. The use <strong>of</strong> adjuvant therapy has increased substantially, trials<br />

reported in the 1980’s had a mean <strong>of</strong> 0.07% <strong>of</strong> patients having had<br />

adjuvant chemotherapy compare to a mean <strong>of</strong> 48% in the last decade.<br />

Overall survival has significantly improved; the slope <strong>of</strong> the fitted line for<br />

first line clinical trials was 0.39 (p�0.001), which indicates a 0.39-month<br />

increase in median survival time per year. In trials <strong>of</strong> subsequent lines <strong>of</strong><br />

therapy this slope was 0.19 (p�0.001). PFS or TTP was reported in 98<br />

trials, interestingly in contrast to OS, this has not significantly changed, the<br />

slope for the fitted line has remained almost flat in both first line (0.002,<br />

p�0.97) and refractory trials (0.01, P�0.81). Conclusions: Our study is<br />

the first <strong>of</strong> its kind conducted in trials <strong>of</strong> patients with MBC. It shows that<br />

progress has been made in the last 3 decades OS in women affected with<br />

MBC, however the lack <strong>of</strong> change in PFS or TTP appears to indicate that the<br />

increase in OS is driven by a combination <strong>of</strong> newer agents, more<br />

subsequent lines <strong>of</strong> therapy being <strong>of</strong>fered to patients and improved<br />

palliative care.<br />

1091 General Poster Session (Board #28A), Sat, 8:00 AM-12:00 PM<br />

Validation <strong>of</strong> NAD(P)H quinone oxidoreductase (NQO1) expression as a<br />

predictive factor for adjuvant chemotherapy benefit in patients with early<br />

breast cancer. Presenting Author: Monica Arnedos, Institut Gustave Roussy,<br />

Villejuif, France<br />

Background: NAD(P)H:quinone oxidoreductase-1 (NQO1) has important<br />

antioxidant functions by stabilizing p53 from proteasomal degradation.<br />

NQO1 knockdown is associated with higher susceptibility to oxidative<br />

stress. Polymorphisms suppressing NQO1 are predictors <strong>of</strong> poor survival in<br />

patients with breast cancer (BC) treated with anthracyclines. BC cell lines<br />

with impaired NQO1 function showed resistance to epirubicin chemotherapy<br />

(CT) independently <strong>of</strong> p53 status suggesting NQO1 as predictive<br />

factor for anthracycline benefit. In this study we hypothesized that lack <strong>of</strong><br />

NQO1 could predict resistance to anthracycline-containing CT in patients<br />

with early breast cancer (EBC). Methods: Patients were identified from two<br />

French multicentric trials that randomized patients with EBC to adjuvant<br />

anthracycline-based CT vs no CT between 1988 and 1995. NQO1 was<br />

determined in TMAs by automated quantitative assessment <strong>of</strong> immun<strong>of</strong>luorescence<br />

(On-Q-ity Inc, Waltham). Cut-<strong>of</strong>f for positivity was the median<br />

value <strong>of</strong> NQO1 expression. Univariate and multivariate Cox regression<br />

models were performed. Treatment effect was assessed on long term overall<br />

survival (OS). Results: NQO1 expression was assessed in 600 patients.<br />

75% were postmenopausal, had more <strong>of</strong>ten grade II (62%), LN-negative<br />

(58%) and ER-positive (67%) BC. Higher NQO1 expression was observed<br />

in postmenopausal (P�0.02) patients. No relation with other clinicopathological<br />

factors (grade, LN or ER) was observed. Effect <strong>of</strong> adjuvant CT on<br />

death rates was dependant on NQO1 level. Hazard ratio (HR) for treatment<br />

efficacy at the four quartiles <strong>of</strong> NQO1 were 1.07 (95%CI: 0.69-1.7), 0.87<br />

(0.64-1.19), 0.66 (0.45-0.97), 0.46 (0.22-0.95) (interaction test: 0.1).<br />

Interaction test was statistically significant (0.02) when NQO1 expression<br />

was considered as binary variable with median value taken as cut-<strong>of</strong>f.<br />

NQO1 remained predictive among ER� patients only. HR for OS was 0.61<br />

(0.36-1.02) and 1.25 (0.79-1.99) in patients with high and low NQO1<br />

respectively. Conclusions: This study adds to the existing data suggesting<br />

NQO1 expression as an independent predictor <strong>of</strong> efficacy for adjuvant<br />

anthracycline-containing CT.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


72s Breast Cancer—Triple-Negative/Cytotoxics/Local Therapy<br />

1093 General Poster Session (Board #28C), Sat, 8:00 AM-12:00 PM<br />

Analysis <strong>of</strong> pretreatment nonpharmacologic, pharmacologic factors, and<br />

yoga intervention on CINV outcomes in breast cancer patients undergoing<br />

adjuvant chemotherapy. Presenting Author: Malur R. Usharani, HCG<br />

Bangalore Institute <strong>of</strong> Oncology, Bangalore, India<br />

Background: Chemotherapy induced nausea and vomiting (CINV) is affected<br />

by both pretreatment patient factors, chemotherapy and antiemetic<br />

regimen and psychological interventions. In this study we evaluated the<br />

effects <strong>of</strong> mind body intervention such as yoga in modulating CINV<br />

outcomes controlled for the above factors. Methods: Chemotherapy naïve<br />

breast cancer patients with stage II and III disease participating in a<br />

randomized controlled trial comparing yoga (n�45) vs. supportive therapy<br />

(n�53) were assessed for CINV outcomes during adjuvant chemotherapy.<br />

Morrows Assessment <strong>of</strong> nausea and emesis was used to asses CINV<br />

symptoms including their frequency, severity and anticipatory nature. We<br />

developed a multiple regression analyses to test the role <strong>of</strong> intervention on<br />

CINV beyond that explained by the independent prognostic factors [age<br />

(�50/�50 years), stage <strong>of</strong> disease (II vs III), menopausal status (pre vs<br />

post), antiemetic regimen (5HT3 antagonists vs. antidopaminergics),<br />

administration <strong>of</strong> anxiolytics (yes/ no) and type <strong>of</strong> chemotherapy regimen<br />

(FAC vs. CMF)] that were included in model A. Model B includes these six<br />

variables plus intervention (yoga vs. supportive therapy) in predicting<br />

nausea and vomiting outcomes. Results: Intervention emerged as a primary<br />

predictor for nausea frequency (�� -0.38, p�0.002), intensity (�� -0.44,<br />

p�0.001 ), anticipatory nausea frequency (��-0.26 , p� 0.04) and<br />

intensity (��-0.38 , p�0.004 ). Age group emerged as a primary predictor<br />

for anticipatory vomiting frequency (��-0.39 , p�0.01 ) and secondary<br />

predictor for nausea frequency (��-0.41, p� 0.006). Administration <strong>of</strong><br />

anxiolytics emerged as a primary predictor for vomiting intensity (��-0.40,<br />

p� 0.001) and secondary predictor for anticipatory nausea frequency<br />

(��-0.26 , p� 0.05). Conclusions: Yoga intervention influences CINV<br />

outcomes when controlled for pretreatment and pharmacological factors<br />

during chemotherapy in breast cancer patients poorly controlled for nausea<br />

and vomiting.<br />

1095 General Poster Session (Board #29B), Sat, 8:00 AM-12:00 PM<br />

Management <strong>of</strong> antiangiogenics’ renovascular safety in breast cancer subgroup<br />

and intermediate results <strong>of</strong> the MARS study. Presenting Author: Catherine<br />

Daniel, Curie Institut, Paris, France<br />

Background: Hypertension (HTN) and proteinuria (Pu) are class-side-effects <strong>of</strong><br />

anti-VEGF drugs (AVD), related to the inhibition <strong>of</strong> the VEGF pathway. The MARS<br />

study has been conducted to assess the renovascular tolerance <strong>of</strong> AVD in the<br />

clinical setting. Methods: The MARS study is a multicentric prospective<br />

observational study <strong>of</strong> the renovascular safety <strong>of</strong> AVD in AVD-naive pts. 7 centres<br />

from 2009 to 2011. Data collected included: gender, age, serum creatinine<br />

(SCr), diabetes, HTN, hematuria (Hu) and Pu. This sub-group analysis presents<br />

the intermediate results for the 1st 155 pts with breast cancer (BC) receiving<br />

bevacizumab who completed the 1-year follow-up (f/u) (out <strong>of</strong> 337 BC pts in<br />

total). Results: Median age at inclusion: 62 years. Bone, visceral and cerebral<br />

metastasis frequencies were 74.2, 51.6 and 5.2%, respectively. Diabetes and<br />

HTN prevalences were 3.9% and 10.3%, respectively. Baseline renal assessment<br />

retrieved: Pu 14.2%, Hu 8.4%, aMDRD 98.2 ml/min/1.73m2 and 2 pts<br />

with aMDRD�60. The incidence <strong>of</strong> de novo Pu during f/u was 15% (Table).<br />

59.1% <strong>of</strong> pts with Pu at inclusion improved. Among pts with de novo Pu, 40.0%<br />

afterwards improved/normalized. No grade 3/4 Pu has been reported and no<br />

hematuria. 12.9% developed HTN. Moreover, renal function decreased by -3.2<br />

ml/min/1.73m2 /year and 4 pts had aMDRD�60 at the end <strong>of</strong> f/u. 32.2%<br />

increased their SCr: 27.1% grade 1, 4.5% grade 2, and 0.6% grade 3. All pts<br />

with grade 2-3 returned to normal or grade 1. No thrombotic micro-angiopathy<br />

(TMA) has been reported. Conclusions: These results show that 1) TMA remains<br />

rare, 2) Pu developed in 15% <strong>of</strong> pts, with no grade 3/4, 3) less than 13%<br />

developed HTN and 4) renal function was only slightly impaired with transient<br />

elevations in SCr. Furthermore, in case <strong>of</strong> a renovascular effect, investigators<br />

followed the recommendations from the French <strong>Society</strong> <strong>of</strong> Nephrology (Halimi<br />

JM. Nephrol Ther 2008) and no treatment withdrawal for unmanageable<br />

renovascular toxicity occurred.<br />

Renovascular effects Prevalence at inclusion (%) Incidence during f/u (%)<br />

Pu*<br />

Grade 1<br />

Grade 2<br />

Grade 3-4<br />

14.2<br />

12.3<br />

1.9<br />

0.0<br />

15.0<br />

12.8<br />

2.2<br />

0.0<br />

HTN 10.3 12.9<br />

Hu<br />

Traces/�<br />

8.4<br />

5.8<br />

0.0<br />

��<br />

0.7<br />

���<br />

1.9<br />

SCr increase*<br />

- 32.2<br />

Grade 1<br />

27.1<br />

Grade 2<br />

4.5<br />

Grade 3<br />

* NCI-CTC v4.03.<br />

0.6<br />

1094 General Poster Session (Board #29A), Sat, 8:00 AM-12:00 PM<br />

The relative value <strong>of</strong> anti-Müllerian hormone to predict premature menopause<br />

in patients receiving adjuvant chemotherapy for breast cancer:<br />

Results from the OPTION trial. Presenting Author: Robert C. F. Leonard,<br />

Imperial College London, London, United Kingdom<br />

Background: The OPTION trial in premenopausal women tested the ovarian<br />

protection effect <strong>of</strong> goserelin (G) given randomly before and during<br />

adjuvant chemotherapy for breast cancer. Methods: Using standard chemotherapy,<br />

women were randomised in 2 strata, under 40 yrs and over 40 yrs<br />

at diagnosis. 227 patients were recruited by end December 2009. 173 met<br />

the criteria for 1 year follow-up for this analysis; 140 patients <strong>of</strong> these had<br />

provided adequate data on menstrual bleeding; 87 patients were aged<br />

under 40 and 53 patients were aged over 40 at the time <strong>of</strong> chemotherapy.<br />

Cessation <strong>of</strong> menstruation during chemotherapy was defined as at least two<br />

consecutive cycles with no menstrual bleeding since the previous cycle and<br />

no return <strong>of</strong> menstrual bleeding prior to the final cycle <strong>of</strong> chemotherapy. Of<br />

those patients who had ceased periods during chemotherapy, those with no<br />

further menstrual bleeding at 12 months follow up were deemed to be<br />

menopausal. Patients were randomised to receive G or no G at start <strong>of</strong><br />

chemotherapy. Primary endpoint was recovery <strong>of</strong> menses at 12 months<br />

from start <strong>of</strong> chemotherapy. AMH was measured in 117 women pretreatment,<br />

and at 1 year after starting chemotherapy. Results: There were<br />

no differences in pretreatment AMH between control and goserelin-treated<br />

groups, thus further analyses were performed on all women grouped<br />

together. AMH was lower following chemotherapy (0.40�0.65 vs<br />

1.38�1.82ng/ml; mean�SD; P�0.001)). Pre-treatment AMH was a<br />

significant predictor <strong>of</strong> post-treatment amenorrhoea (P�0.001). By multivariate<br />

logistic regression analysis with age and AMH, age remained<br />

significant (P�0.003) whereas AMH did not (P�0.07). Grouping pretreatment<br />

and post-chemo AMH into quartiles showed that AMH became<br />

undetectable in 94% <strong>of</strong> women with lowest pre-treatment AMH vs 46.2%<br />

<strong>of</strong> women with the highest pretreatment AMH. We have previously<br />

demonstrated in a small cohort that pretreatment AMH can predict<br />

long-term (5 year) ovarian activity in women with breast cancer. Conclusions:<br />

The present data confirm the value <strong>of</strong> pretreatment AMH in assessing the<br />

likelihood <strong>of</strong> ongoing ovarian activity after chemotherapy for early breast<br />

cancer.<br />

1096 General Poster Session (Board #29C), Sat, 8:00 AM-12:00 PM<br />

Neoadjuvant epirubicin, gemcitabine, and docetaxel for primary breast<br />

cancer: Survival and prognostic factors in two consecutive neoadjuvant<br />

phase I/II trials. Presenting Author: Frederik Marme, Department <strong>of</strong><br />

Obstetrics and Gynecology, University Hospital Heidelberg, Heidelberg,<br />

Germany<br />

Background: We previously reported primary end points <strong>of</strong> two consecutive<br />

phase I/II trials which evaluated two different schedules <strong>of</strong> neoadjuvant<br />

gemcitabine (G), epirubicin (E) and docetaxel (Doc) for primary breast<br />

cancer. Here we report mature survival data and evaluate prognostic factors<br />

for disease-free (DFS) and overall survival (OS). Methods: 151 patients were<br />

treated in two phase I/II trials <strong>of</strong> G, E and Doc as neoadjuvant chemotherapy<br />

(NAC) for T2-4 N0-2 M0 PBC between Feb. ´02 and Dec. ´04.<br />

Patients were treated with six cycles <strong>of</strong> GEDoc (G 800mg/m2 day (d) 1�8,<br />

E 60-90mg/m2 d 1, Doc 60-75mg/m2 d 1 every three weeks) or five cycles<br />

<strong>of</strong> G 1250mg/m2 plus E 90-100mg/m2 every two weeks followed sequentially<br />

by four cycles <strong>of</strong> Doc 80-100mg/m2 every two weeks (GEsDoc).<br />

Pathologic complete response (pCR), clinical/pathological factors were<br />

correlated with DFS and OS. Results: There was no significant difference in<br />

DFS or OS between patients in the GEDoc and GEsDoc trial (DFS: Hazard<br />

ratio (HR) 1.13, p�0.67; OS: HR 1.06, p�0.88) with a 5-year DFS and OS<br />

<strong>of</strong> 72 vs 74% and 85 vs 86%, respectively. In an univariate analysis pCR<br />

unexpectedly was associated with a worse OS (HR 3.11; p� 0.007). HR for<br />

DFS showed a similar but non-significant trend (HR 1.78; p�0.1).<br />

Molecular subtypes (OS: HR [lum B] 3.17; [triple negative] 5.81; [HER2]<br />

11.5; p�0.002), negative estrogen receptor (ER) status (OS: HR 3.14;<br />

p�0.002) and Ki-67 �20% (OS: HR 5.41; p�0.001) were all significantly<br />

associated with DFS and OS. The recently published CPS-EG score<br />

(Mittendorf 2011) was also significantly correlated with OS (p�0.006) and<br />

DFS (p�0.0006). In a multivariate analysis high Ki-67 was the only<br />

significant predictor <strong>of</strong> OS (HR 10.4; p�0.0026) whereas molecular<br />

subtype (p�0.05) and Ki-67 (p�0.04) were significantly associated with<br />

DFS. Conclusions: These results raise caution on the reliability <strong>of</strong> pCR as a<br />

single surrogate marker for survival in trials with small sample sizes. Our<br />

results emphazise the role <strong>of</strong> additional factors, esp. Ki-67 and subtypes.<br />

Integrative scores based on clinical and pathologic stage as well as tumor<br />

biology, might be more reliable predictors <strong>of</strong> survival.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


1097 General Poster Session (Board #30A), Sat, 8:00 AM-12:00 PM<br />

Prevention <strong>of</strong> chemotherapy-induced damage to ovarian reserve by sphingosine-1-phosphate<br />

Presenting Author: Fang Li, Laboratory <strong>of</strong> Molecular<br />

Reproduction and Fertility Preservation, New York Medical College, Valhalla,<br />

NY<br />

Background: Chemotherapy agents such as cyclophosphamide (Cy) and<br />

doxorubicin (D) are known to compromise ovarian function. We have<br />

previously shown that these agents alter ovarian function by causing<br />

apoptotic death <strong>of</strong> primordial follicles and thereby causing diminishment <strong>of</strong><br />

ovarian reserve (Cancer Research 2007; Aging 2011). While assisted<br />

reproduction techniques exist to preserve fertility there has been no proven<br />

approach to pharmacologic preservation <strong>of</strong> ovarian function. Sphingosine-<br />

1-Phosphate (S1P) is a naturally occurring ceramide-induced death pathway<br />

inhibitor. Here we investigated whether S1P can prevent Cy or<br />

D-induced apoptotic follicle death in mouse ovaries. Methods: Eight wk old<br />

NOD mice (n�23) were treated with Cy (75 mg/kg), Cy�S1P (200 �M), D<br />

(10 mg/kg), D�S1P or vehicle only (Control). S1P was administered via<br />

continuous infusion using a mini-osmotic pump beginning 3 hours prior to<br />

single dose chemotherapy injection for 72 hours. Ovaries were removed<br />

72h later and serially sectioned, and stained with anti-caspase 3 (AC-3)<br />

antibody for the detection <strong>of</strong> apoptosis in primordial follicles. The ratio <strong>of</strong><br />

apoptotic to total follicles was expressed as percentage in each group.<br />

Results: Both Cy and D resulted in significant increase in apoptotic follicle<br />

death compared to controls (48.2�11.6 vs. 28.2�8.9, p�0.016 and<br />

46.4�5.4 vs. 24.8�5.2, p�0.004, respectively). Percentages <strong>of</strong> apoptotic<br />

follicles were similar between Cy and D-treated groups (48.2�11.6 vs.<br />

46.4�5.4, P�NS) indicating that these agents were equally gonadotoxic.<br />

S1P treatment resulted in a significant decrease in the percentage <strong>of</strong><br />

apoptotic follicles both in the Cy (25.6�3.5, P�0.011) and the D group<br />

(32.2�10.8, P� 0.013) compared to controls. In the S1P-treated groups,<br />

the percentages <strong>of</strong> apoptotic follicles were similar to those in untreated<br />

controls indicating that S1P completely blocked Cy and D-induced apoptotic<br />

follicle death. Conclusions: S1P can block apoptotic follicle death<br />

induced by highly cytotoxics agents. In addition, we showed that S1P can<br />

block follicle death by chemotherapy agents that act through different<br />

molecular mechanisms. If targeted delivery systems can be developed, S1P<br />

may hold significant promise in preserving fertility by pharmacological<br />

means.<br />

1099 General Poster Session (Board #30C), Sat, 8:00 AM-12:00 PM<br />

Phase II trial <strong>of</strong> sorafenib (S) and vinorelbine (V) in metastatic breast<br />

cancer (mBC) with pharmacokinetics (PK) analysis. Presenting Author:<br />

Cristiano Ferrario, Department <strong>of</strong> Oncology, McGill University, Montreal,<br />

QC, Canada<br />

Background: S inhibits pathways involved in cancer resistance to treatments<br />

(Raf, VEGFR, PDGFR). This phase 2 trial aimed to define tolerability<br />

and efficacy <strong>of</strong> full doses <strong>of</strong> S and V in mBC. Toxicity data were previously<br />

reported: frequent dose reductions were noted, for which we investigated a<br />

possible PK interaction. Indeed, the metabolism <strong>of</strong> both S and V depends<br />

on hepatic CYP3A isoenzymes. Methods: Patients with measurable (RE-<br />

CIST), HER2 negative mBC received first-line therapy with V (30 mg/m2 days 1, 8 every 21) � S (400 mg bid). After 8 cycles patients could be<br />

switched to S alone. For PK analysis 6 patients started S on day 4 <strong>of</strong> cycle<br />

1, to compare plasma levels <strong>of</strong> V, S and M2 (N-oxide active metabolite <strong>of</strong> S)<br />

when V and S were administered apart from each other versus concomitantly.<br />

Plasma samples were collected at time 0 (oral intake <strong>of</strong> S or right<br />

before V infusion), at completion <strong>of</strong> V infusion and after 0.5, 1, 2.5, 5, 7,<br />

24, 48 and 72 hours from time 0 (cycle 1 day 1 for V and day 21 for S�M2;<br />

cycle 2 day 1 for both). Samples were analyzed using validated LC-MS/MS<br />

assays. Results: 27 patients (median age 57, 35-71) received a median <strong>of</strong> 8<br />

cycles (1-28), with one patient still on treatment. With repeated cycles<br />

48% <strong>of</strong> patients required at least 1 dose reduction and 3 patients<br />

discontinued therapy for toxicity. 30% <strong>of</strong> patients had a partial response,<br />

85% had clinical benefit (including stable disease � 4 cycles). Median<br />

progression-free survival was 5.7 months (95% CI 4.4-7.6). Plasma levels<br />

<strong>of</strong> V were influenced by S, with a mean Cmax right after the infusion <strong>of</strong><br />

1301 ng/mL for V administered alone (cycle 1 day 1) versus 2039 ng/mL<br />

with concomitant S (cycle 2 day 1; paired t-test, p�0.004). Plasma levels<br />

<strong>of</strong> S and M2 showed a greater degree <strong>of</strong> interpatient variability, with no<br />

significant difference observed in the presence or absence <strong>of</strong> concomitant<br />

V. Conclusions: Combining S with V at full doses is feasible, but not devoid<br />

<strong>of</strong> toxicity. A PK interaction may contribute to the frequent dose reductions.<br />

A reasonable option in clinical practice is to start therapy at lower doses <strong>of</strong><br />

both agents, with a gradual dose increase if well tolerated. Promising<br />

efficacy <strong>of</strong> this combination is documented, with a very high rate <strong>of</strong> disease<br />

control.<br />

Breast Cancer—Triple-Negative/Cytotoxics/Local Therapy<br />

73s<br />

1098 General Poster Session (Board #30B), Sat, 8:00 AM-12:00 PM<br />

A study <strong>of</strong> sperm-associated antigen 5 (SPAG5) in predicting response to<br />

anthracycline (ATC)/platinum chemotherapies (CT) in breast (BC) and<br />

ovarian cancers (OVC). Presenting Author: Tarek M. A. Abdel-Fatah,<br />

Nottingham University City Hospital NHS Trust, Nottingham, United<br />

Kingdom<br />

Background: Recently TOP2A alteration was found to be a predictor for<br />

ATC-CT and our neural network analysis <strong>of</strong> BC gene expression array (GEA)<br />

data has revealed SPAG5 gene as a major hubs in both TOP2A and<br />

proliferation pathways. In this study the molecular and clinicopathological<br />

functions <strong>of</strong> SPAG5 was investigated in BC and OVC. Methods: (1) A series<br />

<strong>of</strong> 171 BC was evaluated for SPAG5 gene copy number (using aCGH) and<br />

mRNA expression (using GEA) which were validated in 5 independent<br />

databases. (2) The expression <strong>of</strong> SPAG5 protein was evaluated preclinically<br />

in BC and OVC cell lines and in both 40 normal breast tissues and<br />

a series <strong>of</strong> 1650 primary BC and was correlated to clinicopathological and<br />

other biomarkers. (3) The association between SPAG5 and response to CT<br />

was investigated in a) 350 ER negative BC treated with adjuvant ATC-CT, b)<br />

250 BC treated with neoadjuvant (NEO-A)-ATC-CT, and c) 200 primary<br />

OVC treated with cisplatinum based adjuvant CT. Results: (1) 5% and 15%<br />

<strong>of</strong> the 171 BC showed amplification and gain <strong>of</strong> SPAG5 locus, respectively,<br />

at 17q11.2. SPAG5 mRNA expression displayed a significant correlation<br />

with its copy number (p� 0.0001). (2) 30% and 20% <strong>of</strong> ovarian and BC<br />

respectively, showed overexpression <strong>of</strong> SPAG5 protein (�). In BC, SPAG5�<br />

at both mRNA and protein levels showed a significant association with<br />

aggressive phenotypes, high mitosis, ER-, high grade, p53 mutation and<br />

epithelial mesenchymal transition phenotypes (ps �0.0001). SPAG5<br />

mRNA (�) was statistically associated with poor survivals (p�0.0001). (3)<br />

In ER- BC treated with adjuvant ATC-CT, SPAG5 negative (-)had 7-times<br />

higher risk <strong>of</strong> progression compared with SPRAG� BC (p�0.0001).<br />

SPAG5� BC received NEO-A-ATC based CT achieved 38% pathological<br />

complete response (pCR) vs. 6% <strong>of</strong> SPAG5- (p�0.0001). After controlling<br />

to other predictors for pCR, SPAG5 was an independent predictor (HR; 2.4;<br />

p�0.001). Similarly, SPAG5- OVCs were resistant to platinum (p�0.001)<br />

and independently associated with poor survival (p�0.001). Conclusions:<br />

SPAG5 is an important novel gene implicated in the survival <strong>of</strong> BC and OVC<br />

cells and its protein expression is an independent predictor for anthracycline/<br />

cisplatinum CT.<br />

1100 General Poster Session (Board #31A), Sat, 8:00 AM-12:00 PM<br />

Receipt <strong>of</strong> locoregional therapy among young women with breast cancer.<br />

Presenting Author: Rachel A. Freedman, Dana-Farber Cancer Institute,<br />

Boston, MA<br />

Background: Although younger women with breast cancer have the most to<br />

gain from receipt <strong>of</strong> optimal care, few data are available regarding their<br />

receipt <strong>of</strong> locoregional breast cancer treatments. Methods: We identified<br />

318,083 women aged 18-64 who were diagnosed with invasive breast<br />

cancer at hospitals reporting to the National Cancer Data Base, a large<br />

national cancer registry, during 2004-2008. We used multivariable logistic<br />

regression to assess the association <strong>of</strong> patient age with mastectomy vs.<br />

breast-conserving surgery (BCS), radiation with BCS, and post-mastectomy<br />

radiation (PMRT) with varying indications, adjusting for patient and tumor<br />

characteristics, area-level socioeconomic status, and insurance. Results:<br />

Overall, 4% <strong>of</strong> women were aged �35 and 7% were aged 36-40. Women<br />

aged �35 were significantly more likely to have mastectomy than BCS<br />

compared with women aged 56-60 (57% vs. 35%, adjusted odds ratio [OR]<br />

1.97; 95% Confidence Interval [CI] 1.87-2.07) but were less likely to<br />

receive radiation if BCS was performed (69% vs. 80%, OR 0.77; 95% CI<br />

0.73-0.82). For those who underwent mastectomy, although overall rates<br />

<strong>of</strong> PMRT receipt were low, women aged �35 were more likely to receive<br />

postmastectomy radiation (PMRT) despite the presence or absence <strong>of</strong><br />

clinical indications for PMRT (OR 1.11; 95% CI 1.01-1.22 for strong<br />

indications, OR 1.71; 95% CI 1.53-1.91 for borderline indications, and<br />

OR 1.49; 95% CI 1.28-1.73 for no indications [all vs. ages 56-60]).<br />

Conclusions: Young women with breast cancer may not be receiving optimal<br />

locoregional therapy. We observed lower odds <strong>of</strong> radiation after BCS but<br />

higher odds <strong>of</strong> PMRT for young women regardless <strong>of</strong> indications for PMRT.<br />

Efforts are needed to further understand and improve the receipt <strong>of</strong><br />

appropriate adjuvant radiation therapy among young women to improve<br />

their disease-free and overall survival.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


74s Breast Cancer—Triple-Negative/Cytotoxics/Local Therapy<br />

1101 General Poster Session (Board #31B), Sat, 8:00 AM-12:00 PM<br />

Patterns <strong>of</strong> regional node irradiation therapy following breast-conserving<br />

surgery: Report from the National Cancer Data Base 2003-2007. Presenting<br />

Author: Ningqi Hou, University <strong>of</strong> Chicago, Chicago, IL<br />

Background: A recent randomized trial (MA.20) showed that regional nodal<br />

irradiation (RNI) in addition to breast irradiation in high risk node negative<br />

and 1-3 node positive patients undergoing breast-conserving therapy (BCT)<br />

reduced the risk <strong>of</strong> recurrence and improved disease-free survival. We<br />

investigated the trends <strong>of</strong> RNI use in the United States and related factors<br />

for the use <strong>of</strong> RNI using the National Cancer Data Base. Methods: This study<br />

includes 292,598 stage I-III breast cancer patients without neoadjuvant<br />

therapy who underwent BCT from 2003-2007. We investigated pathological,<br />

patient, and facility factors related to RNI use, by multivariable logistic<br />

regression, with odds ratio (OR) estimations. Results: The proportion <strong>of</strong><br />

radiotherapy use after BCT slightly declined from 78.6% in 2003 to 75.6%<br />

in 2007. The use <strong>of</strong> breast irradiation plus RNI decreased from 10.8% in<br />

2003 to 8.3% in 2007 (p�0.0001). The number <strong>of</strong> tumor positive lymph<br />

nodes strongly determined the use <strong>of</strong> additional RNI: 4.4% patients with<br />

negative nodes, 22.8% patients with 1-3 nodes, and 39.7% patients with<br />

4 or more nodes received breast irradiation plus RNI after undergoing BCT<br />

(p�0.0001). The proportion <strong>of</strong> patients undergoing RNI significantly<br />

decreased over the study period from 43.3% to 37.2% in the 4� node<br />

positive group, and from 23.6% to 22.0% in the 1-3 node positive group.<br />

At comprehensive community cancer centers, 25.5% patients with 1-3<br />

positive nodes were treated with breast irradiation plus RNI (vs. 23.2% in<br />

community cancer centers and 21.1% in academic/research cancer<br />

centers). Among node negative patients, 11.5% <strong>of</strong> those with tumor size<br />

greater than 5 cm received additional RNI, compared to 4.3% in patients<br />

with tumors less than 5cm (p�0.0001). Other significant factors related to<br />

RNI use included higher tumor grade, younger age, facility location, and<br />

facility volume. Conclusions: The use <strong>of</strong> RNI varies by number <strong>of</strong> tumor<br />

positive nodes and facility factors. Only 22.8% <strong>of</strong> patients with 1-3 positive<br />

nodes underwent RNI. Future studies are needed to determine if the use <strong>of</strong><br />

RNI will increase after publication <strong>of</strong> the MA.20 trial especially for the 1-3<br />

node positive group.<br />

1103 General Poster Session (Board #31D), Sat, 8:00 AM-12:00 PM<br />

SPIO-enhanced MR imaging for axillary staging to avoid sentinel node<br />

biopsy in patients with breast cancer. Presenting Author: Kazuyoshi<br />

Motomura, Department <strong>of</strong> Breast and Endocrine Surgery, Osaka Medical<br />

Center for Cancer and Cardiovascular Diseases, Osaka, Japan<br />

Background: We previously demonstrated the usefulness <strong>of</strong> SPIO-enhanced<br />

MR imaging for the detection <strong>of</strong> metastases in sentinel nodes localized by<br />

computed tomography (CT) lymphography (CT-LG) in patients with breast<br />

cancer (Ann Surg Oncol, 2011). These techniques have evolved and we<br />

report our most recent results <strong>of</strong> axillary staging using them. Methods:<br />

Previously unreported 87 consecutive patients with breast cancer and<br />

clinically negative nodes were enrolled in this study. Sentinel nodes<br />

identified by CT-LG were evaluated prospectively using SPIO-enhanced MR<br />

imaging. A node was considered non-metastatic if it showed a homogenous<br />

low signal intensity and metastatic if the entire node or a focal area did not<br />

show a low signal intensity on MR imaging. Sentinel nodes located by<br />

CT-LG were removed, and imaging results and histopathological findings<br />

were compared. Results: The mean patient age was 54.9 years (range,<br />

34-77). Sentinel nodes were identified by CT-LG and removed successfully<br />

in all patients. The mean number <strong>of</strong> sentinel nodes identified by CT-LG was<br />

1.16 (range, 1-2). Twenty <strong>of</strong> 22 patients with positive sentinel nodes<br />

definitively diagnosed by pathology demonstrated metastases on SPIOenhanced<br />

MR imaging. Fifty-eight <strong>of</strong> 65 patients with negative sentinel<br />

nodes definitively diagnosed by pathology were non-metastatic on imaging<br />

studies. The sensitivity, specificity and accuracy <strong>of</strong> MR imaging for the<br />

diagnosis <strong>of</strong> sentinel node metastases were 91%, 89%, and 90%,<br />

respectively. Two patients whose metastases were not detected had<br />

micrometastases. No adverse events were associated with either CT or MR<br />

imaging. Conclusions: SPIO-enhanced MR imaging provided accurate<br />

axillary staging, and therefore sentinel node biopsy may not be necessary<br />

for most patients with breast cancer.<br />

1102 General Poster Session (Board #31C), Sat, 8:00 AM-12:00 PM<br />

Primary tumor resection to improve survival and local disease control in<br />

stage IV inflammatory breast cancer. Presenting Author: Catherine Akay,<br />

University <strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: Inflammatory breast cancer (IBC) is a rare and aggressive form<br />

<strong>of</strong> breast cancer typically presenting with early metastasis. Optimal<br />

outcomes are achieved with multimodality treatment strategies in the<br />

non-metastatic setting. Data is limited, however, on the benefit <strong>of</strong> surgery<br />

in patients with metastatic IBC. We evaluated the effect <strong>of</strong> primary tumor<br />

resection on outcomes in patients with newly diagnosed stage IV IBC.<br />

Methods: We reviewed records <strong>of</strong> 172 patients with metastatic IBC treated<br />

at our institution from 1994 - 2009. All patients received systemic therapy<br />

with or without locoregional therapy (LRT). Patient demographics, receptor<br />

(ER) and HER2-neu status, grade, histology, presence <strong>of</strong> lymphovascular<br />

invasion, margin status, number <strong>of</strong> distant disease sites, pathologic<br />

response <strong>of</strong> primary tumor and clinical response to systemic therapy (CRS)<br />

at distant disease sites were recorded. Overall survival (OS), distant<br />

progression-free survival (DPFS), and chest/skin involvement at last follow-up<br />

were evaluated. Kaplan-Meier survival analyses, univariate (UV) and<br />

multivariate (MV) logistic regression models were used. Chest/skin involvement<br />

was compared between groups using Kruskal-Wallis test. Results:<br />

Seventy-nine (45%) patients underwent primary tumor resection. Average<br />

age was 51 (22-78). Median live-patient follow-up was 33 months. OS and<br />

DPFS were significantly better for patients who underwent LRT versus none<br />

(p�0.0001). Factors associated significantly for improved DPFS on MV<br />

analysis were ER and HER2-neu status (HR 0.61,0.60 p�0.02,0.05<br />

,respectively), LRT (HR .38, p�0.002) and CRS (HR 0.62, p�0.03). ER<br />

status (HR .45, p�0.001), LRT (HR .30, p�0.001) and CRS (HR 0.54,<br />

p�0.02) were significant predictors for higher OS on MV analysis. At last<br />

follow up, chest/skin involvement was moderate/severe in 11% <strong>of</strong> patients<br />

in LRT group versus 35% <strong>of</strong> patients in no LRT group (p�0.0001).<br />

Conclusions: This latest retrospective study demonstrates metastatic IBC<br />

patients who undergo LRT in addition to systemic therapy may have<br />

improved survival and local control outcomes. CRS may be used to guide<br />

LRT. A prospective randomized trial is needed to validate these findings.<br />

1104 General Poster Session (Board #31E), Sat, 8:00 AM-12:00 PM<br />

Breast cancer multifocality-multicentricity and survival outcomes. Presenting<br />

Author: Siobhan P. Lynch, University <strong>of</strong> Texas M. D. Anderson Cancer<br />

Center, Houston, TX<br />

Background: Studies have consistently shown a correlation between multifocal<br />

(MF) and multicentric (MC) breast cancers and the rate and extent <strong>of</strong><br />

lymph node metastases, but the literature is divided on whether there is a<br />

corresponding impact on survival outcomes. In the absence <strong>of</strong> compelling<br />

evidence to dictate otherwise, the convention according to current TNM<br />

staging guidelines has been to stage and treat MF and MC cancers<br />

according to the diameter <strong>of</strong> the largest lesions, without taking other foci <strong>of</strong><br />

disease into consideration. We evaluated a large single institution cohort <strong>of</strong><br />

MF and MC breast cancers to determine their frequency, clinicopathological<br />

characteristics and effect on survival outcomes. Methods: MF<br />

and MC were defined pathologically as more than one lesion in the same<br />

quadrant and more than one lesion in separate quadrants, respectively.<br />

Patients were categorized by presence or absence <strong>of</strong> MF or MC disease.<br />

Kaplan-Meier product limit method was used to calculate relapse-free<br />

survival (RFS), breast cancer-specific survival (BCSS) and overall survival<br />

(OS). Cox proportional hazards models were fit to determine independent<br />

associations <strong>of</strong> MF/MC disease with survival outcomes. Results: Out <strong>of</strong><br />

3924 patients, 942 (24%) had MF (n�695) or MC (n�247) disease. MF<br />

and MC disease was associated with higher T-stages (T2 26% vs. 21.6%;<br />

T3 7.4% vs. 2.3%, P�0.001), higher nuclear grade (grade 3 44% vs.<br />

38.2%, P�0.001), lymphovascular invasion (26.2% vs. 19.3%, P�0.001)<br />

and lymph node metastases (43.1% vs. 27.3%, P�0.001). After a median<br />

follow up <strong>of</strong> 51 months, MC but not MF breast cancers were associated with<br />

significantly worse 5-year RFS (90% vs. 95%, P�0.02) and BCSS (95%<br />

vs. 97%, P�0.01), and a trend towards worse 5-year OS (92% vs. 93%,<br />

P�0.08). After controlling for other risk factors, multifocality and multicentricity<br />

did not have an independent impact on RFS, BCSS or OS. This was<br />

true for the subset <strong>of</strong> T1N0 breast cancers as well. Conclusions: MF and MC<br />

breast cancers occurred in 24% <strong>of</strong> the cases and were associated with poor<br />

prognostic factors, but they were not independent predictors <strong>of</strong> worse<br />

survival outcomes. Our findings support the current TNM staging system <strong>of</strong><br />

using the diameter <strong>of</strong> the largest lesion to assign T-stage.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


1105 General Poster Session (Board #31F), Sat, 8:00 AM-12:00 PM<br />

Is there a need for sentinel lymph node biopsy in microinvasive breast<br />

cancer? Presenting Author: Nimmi S. Kapoor, John Wayne Cancer Institute,<br />

Santa Monica, CA<br />

Background: There is limited data on the long-term outcome <strong>of</strong> patients with<br />

microinvasive breast cancer. Moreover, predictors <strong>of</strong> lymph node involvement<br />

and the impact <strong>of</strong> multifocal microinvasion are not well understood.<br />

We examined the occurrence <strong>of</strong> nodal metastasis and the significance <strong>of</strong><br />

multifocality on disease recurrence. Methods: Patients with T1mic breast<br />

cancer, defined as tumors �1mm, surgically managed at our institute<br />

between 1995-2010 were identified. Specimen slides were independently<br />

reviewed. Multivariable analysis (MVA) was used to predict lymph node<br />

involvement and disease recurrence. Results: Fifty-two patients with T1mic<br />

breast cancers were identified. Median patient age was 53 (range 30-92),<br />

median size <strong>of</strong> in-situ disease was 3cm (range 0.1-12cm). Ten patients<br />

(19.2%) had multiple foci <strong>of</strong> microinvasion (range 2-7). The majority <strong>of</strong><br />

tumors were high-grade (76.9%). When the invasive tumor component was<br />

evaluated, 31 <strong>of</strong> 41 (73.8%) were ER positive, 40.5% were HER2�(15/<br />

37), and only one was ER-/PR-/HER2-. Twenty-nine patients (55.8%) had<br />

breast conserving surgery and 23 had mastectomies. Lymph nodes were<br />

assessed in 48 patients; there was 1 macrometastasis (2.1%), 4 micrometastases<br />

(8.3%) and 4 (8.3%) with isolated tumor cells. Seven <strong>of</strong> 9 patients<br />

with lymph node involvement underwent adjuvant chemotherapy. Univariable<br />

analysis showed that ER(-) invasive disease and high-grade DCIS<br />

tumors were more likely to have involved lymph nodes. On MVA, only<br />

negative ER status was a significant predictor <strong>of</strong> lymph node metastasis<br />

(p�0.02). At median follow-up <strong>of</strong> 83 months (range 6-172 months), 3<br />

patients (6.3%) had disease recurrence (1 local, 1 distant, 1 local and<br />

distant) at 8, 17, and 130 months from presentation. All patients with<br />

recurrence had negative lymph nodes and only one focus <strong>of</strong> microinvasion.<br />

No factors predicted disease recurrence. Conclusions: Microinvasive breast<br />

cancer clearly has the ability to metastasize and recur, but in this series<br />

only 2% <strong>of</strong> patients presented with nodal macrometastasis. The evaluation<br />

<strong>of</strong> lymph nodes in T1mic cancer is unnecessary in the majority <strong>of</strong> patients.<br />

In our cohort, neither lymph node status nor multifocal microinvasion<br />

predicts recurrence.<br />

1107 General Poster Session (Board #31H), Sat, 8:00 AM-12:00 PM<br />

Role <strong>of</strong> axillary ultrasound after neoadjuvant chemotherapy in women with<br />

node-positive breast cancer (T1-4, N1-2, M0) at initial diagnosis (ACOSOG<br />

Z1071). Presenting Author: Huong T. Le-Petross, University <strong>of</strong> Texas M. D.<br />

Anderson Cancer Center, Houston, TX<br />

Background: The role <strong>of</strong> axillary ultrasound (AUS) after neoadjuvant<br />

chemotherapy (NAC) to assess for residual nodal disease in patients<br />

presenting with node positive breast cancer remains unclear. ACOSOG<br />

Z1071 is a prospective multi-institutional trial evaluating sentinel node<br />

biopsy in patients with biopsy proven node positive breast cancer (T0-4,<br />

N1-2, M0) receiving NAC. Herein we report on the secondary objective<br />

evaluating the correlation <strong>of</strong> lymph node (LN) features on AUS with residual<br />

nodal disease. Methods: AUS images from diagnosis and after NAC were<br />

centrally reviewed for cortex size, LN size and LN morphology. Morphologic<br />

features were defined as: type I, no visible cortex, type II, � 3 mm<br />

hypoechoic cortex, type III, � 3mm hypoechoic cortex, type IV, generalized<br />

lobulated hypoechoic cortex, type V, focal hypoechoic cortical lobulation,<br />

and type VI, totally hypoechoic node with no hilum. Type I and II are<br />

considered normal. Results: Surgical and imaging data are available on 294<br />

patients. Median age was 50 years (range 23-93 years), mean initial tumor<br />

size 3cm (0 to 15cm) and clinical stage II in 64.5% and III in 35.5%. The<br />

maximum LN diameter decreased after NAC (mean 22mm pre-NAC to<br />

14mm post-NAC)(p�0.0001); however, there was no significant difference<br />

after NAC between the pathologically N� (13mm, range 5-46mm)<br />

and N0 cases (12mm, range 3-32mm)(p�0.13). LN cortical thickness<br />

correlated with residual nodal disease after NAC (p-value � 0.04). Using a<br />

cut<strong>of</strong>f point <strong>of</strong> cortical thickness <strong>of</strong> 3 mm, the sensitivity was 33%<br />

(48/145) and specificity 80% (66/82). AUS morphological features after<br />

NAC was associated with false negative rate 62%, false positive rate 28%,<br />

sensitivity 38%, and specificity 72%. Conclusions: AUS after NAC is useful<br />

to assess nodal response. Cortical thickness was the best predictor <strong>of</strong><br />

residual nodal metastasis. LN size and morphological features do not<br />

reliably exclude residual nodal metastasis in patients after NAC.<br />

Morphology At diagnosis<br />

After NAC<br />

node positive<br />

After NAC<br />

node negative<br />

Type I 0% 15.7% 22.0%<br />

Type II 0.3% 46.0% 50.5%<br />

Type III 2.4% 13.0% 13.8%<br />

Type IV 26.9% 7.6% 2.8%<br />

Type V 13.3% 7.6% 5.5%<br />

Type VI 57.1% 10.3% 5.5%<br />

Breast Cancer—Triple-Negative/Cytotoxics/Local Therapy<br />

75s<br />

1106 General Poster Session (Board #31G), Sat, 8:00 AM-12:00 PM<br />

Prognosis <strong>of</strong> early breast cancer patients treated with sentinel node biopsy:<br />

A prospective study from the Japanese society for sentinel node navigation<br />

surgery. Presenting Author: Shigeru Imoto, School <strong>of</strong> Medicine Kyorin<br />

University, Mitaka, Japan<br />

Background: Sentinel node biopsy (SNB) is a new standard <strong>of</strong> care for<br />

clinically node-negative breast cancer patients. Our society conducted a<br />

prospective study on SNB for early breast cancer patients from Jul 2004 to<br />

Oct 2005 (UMIN000006126). A preliminary result regarding with success<br />

rates <strong>of</strong> multiple mapping methods and the adverse events was presented<br />

at the 2007 ASCO. We observed patient’s outcome for 5 years. Methods: Of<br />

the 1,411 cases registered, the objects were 1,107 cases excluding cases<br />

with bilateral breast cancer, non-invasive breast cancer, past history <strong>of</strong><br />

other malignancy or failure <strong>of</strong> SNB and cases treated with primary<br />

chemotherapy or endocrine therapy. Adjuvant therapy and breast irradiation<br />

were decided by physician’s discretion and patient’s preference. To<br />

evaluate the risk <strong>of</strong> isolated tumor cells or micrometastases in sentinel<br />

lymph nodes (SLN), clinicopathological factors were analyzed using the<br />

Cox regression model. Results: After a median follow-up <strong>of</strong> 62 months,<br />

there were 85 recurrences and 14 deaths. 5-year Kaplan-Meire estimates<br />

for disease-free survival and overall survival (OS) were 92.6% and 97.5%<br />

in 848 cases with pN0(sn), 96.2% and 100% in 26 with pN0(i�)(sn),<br />

89.3% and 95.3% in 68 with pN1mi(sn) and 82.8% and 92.0% in 165<br />

with pN1(sn) or greater nodal metastases. No axillary lymph node dissection<br />

(ALND) was performed in 809 cases (95.4%), 18 (69.2%), 38<br />

(55.9%), and 24 (14.5%), respectively. Regional node recurrence was<br />

found in 8 cases (0.9%), 0 (0%), 2 (2.9%) and 2 (1.2%), respectively.<br />

Univariate analysis showed that pN1(sn) or greater nodal metastases,<br />

pT2-4, nuclear grade 3, lymphovascular invasion, negative hormone<br />

receptor status, SNB followed by ALND, chemotherapy therapy were<br />

significant risk factors for OS. However, from multivariate analysis, nuclear<br />

grade 3, lymphovascular invasion and SNB followed by ALND were<br />

independent unfavorable prognostic factors (hazard ratio: 3.21, 2.61 and<br />

3.93). Conclusions: Although a non-randomized prospective study, isolated<br />

tumor cells and micrometastases in SLN did not affect patient’s survival.<br />

ALND should be omitted for early breast patients with those metastases.<br />

1108 General Poster Session (Board #32A), Sat, 8:00 AM-12:00 PM<br />

Post-neoadjuvant chemotherapy sentinel node biopsy and axillary sampling<br />

for node negative (N0) axilla. Presenting Author: Vani Parmar, Tata<br />

Memorial Hospital, Mumbai, India<br />

Background: Post neoadjuvant chemotherapy (NACT) sentinel node biopsy<br />

(SNB) is not a standard <strong>of</strong> care due to the wide variability in false negative<br />

rate (FNR), varying from 5.7% to 33%. In operable breast cancer (OBC),<br />

FNR <strong>of</strong> less than 10% is acceptable. We attempted to find out the reliability<br />

<strong>of</strong> low axillary sampling(LAS), with dissection limited below the first<br />

intercostobrachial nerve, to correctly identify the node negative axilla in the<br />

post NACT clinically node negative (N0) patients. Methods: Women with<br />

large operable (LOBC) and locally advanced breast cancer (LABC), post-<br />

NACT clinically N0, underwent concomitant blue dye-colloid guided SNB<br />

and LAS. The identification rate, FNR, and negative predictive value (NPV)<br />

<strong>of</strong> both procedures were compared. Results: Post-NACT 209 eligible women<br />

underwent combined LAS and SNB procedure. At presentation, the tumors<br />

were large (median 5.0 cm) with 70% clinically palpable nodes. All<br />

patients received 4 cycles <strong>of</strong> neo-adjuvant anthracycline-based chemotherapy<br />

and were clinically node negative after chemotherapy. SNB was<br />

defined as blue and/or hot node plus palpable node(s). A blue or hot node<br />

(median 2 nodes) was identified in 93.8%, and median <strong>of</strong> 5 sentinel nodes<br />

were removed. The false negative rate <strong>of</strong> SNB was 15.3% (95% CI<br />

8.7%-25.3%). The LAS technique comparatively had nodal yield in 98.5%<br />

with median 8 nodes removed; and FNR 8.5% (95% CI, 4.2%-16.6%,<br />

p�0.19). Comparative NPV for LAS and SNB were 94.6% and 91.8%<br />

respectively. Conclusions: Axillary sampling results for FNR and NPV are<br />

similar if not superior to SNB and could be a reliable method <strong>of</strong> axillary<br />

nodal evaluation in advanced breast cancers following neo-adjuvant chemotherapy.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


76s Breast Cancer—Triple-Negative/Cytotoxics/Local Therapy<br />

1109 General Poster Session (Board #32B), Sat, 8:00 AM-12:00 PM<br />

Accuracy <strong>of</strong> breast MRI in predicting pathologic tumor size. Presenting<br />

Author: Kimberly Anne Caprio, Yale University, New Haven, CT<br />

Background: MRI use as a preoperative planning tool is increasing in women<br />

with breast cancer, yet the correlation between MRI and pathologic size <strong>of</strong><br />

cancers is unclear. The purpose <strong>of</strong> this study was to determine the accuracy<br />

<strong>of</strong> MRI in predicting pathologic tumor size, and factors that affect this<br />

correlation. Methods: Clinicopathologic and imaging data from 84 patients<br />

diagnosed with invasive or in situ breast cancer from September 2010 to<br />

October 2011 who had preoperative MRI were reviewed. 12 patients who<br />

had neoadjuvant chemotherapy were excluded. MRI detected 147 lesions<br />

in the remaining 72 patients. Concordance between MRI and pathology<br />

size was determined using Spearman rho coefficients, and factors affecting<br />

the accuracy <strong>of</strong> MRI in predicting tumor size within �/- 0.5 cm were<br />

determined. Results: There was a modest correlation between MRI and<br />

pathology size for all MRI detected lesions (benign or malignant) with a<br />

Spearman coefficient <strong>of</strong> 0.53. Of the 147 MRI detected lesions, 45<br />

(30.6%) had pathologic and MRI size correlating within �/- 0.5 cm; 76<br />

(51.7%) were overestimated (�0.5cm) by MRI, and 26 (17.7%) were<br />

underestimated (�0.5cm). 101 (68.7%) <strong>of</strong> the 147 lesions were found to<br />

be malignant (either with invasive disease or DCIS). In this subgroup, 35<br />

lesions (34.7%) had an MRI size within �/- 0.5 cm <strong>of</strong> the pathologic size;<br />

40 (39.6%) were overestimated by MRI and 26 (25.7%) were underestimated.<br />

Patient age, tumor histology, LVI and grade did not predict<br />

concordance between pathologic and MRI size. However, small MRI lesion<br />

size more accurately correlated with pathologic tumor size. While 51.1% <strong>of</strong><br />

tumors that had concordant MRI and pathologic findings within 0.5 cm<br />

were �1 cm on MRI, no tumor found to be � 5 cm on MRI was within<br />

�/-0.5 cm on final pathology (p�0.001). Conclusions: MRI accurately<br />

predicts pathologic tumor size only when the size <strong>of</strong> the lesion on MRI is �1<br />

cm.<br />

1112 General Poster Session (Board #32E), Sat, 8:00 AM-12:00 PM<br />

Comparison <strong>of</strong> long-term results <strong>of</strong> endoscopic video-assisted breast<br />

surgery (VABS) between transaxillary retromammary approach (TARM) and<br />

periareolar approach. Presenting Author: Koji Yamashita, Nippon Medical<br />

School, Tokyo, Japan<br />

Background: The breast conserving surgery (BCS) and the sentinel node<br />

(SN) biopsy became to be recognized as the standard treatment for early<br />

breast cancers. We have reported about cosmetic effectiveness and lower<br />

infestation <strong>of</strong> the video-assisted breast surgery (VABS) for the breast<br />

diseases. We devised the trans-axillary retro-mammary (TRAM) approach <strong>of</strong><br />

VABS. It needs only one skin incision in the axilla and can treat any tumor<br />

<strong>of</strong> the breast without making any injuries on the breast skin. We evaluated<br />

the aesthetic results and the curability <strong>of</strong> this surgical method. Methods: We<br />

have performed VABS on 300 patients since December, 2001. The newly<br />

devised TARM was performed on 120 patients <strong>of</strong> early breast cancer, stage<br />

I and II. After endoscopic SN biopsy, we elongated the axillary skin incision<br />

to 2.5 cm. We dissected major pectoral muscle fascia to detach retromammary<br />

tissue behind the tumor. We cut the mammary gland with clear<br />

surgical margin, and removed it through the axillary port. The breast<br />

reconstruction was made by filling absorbable oxydized cellulose. The<br />

postoperative aesthetic results were evaluated by ABNSW. Results: BCS<br />

was performed on 286 patients (26 after preoperative chemotherapy) and<br />

skin-sparing mastectomy on 14. There was no serious complication after<br />

surgery. Surgical margin was minimally positive in 2. The original shapes <strong>of</strong><br />

the breast were preserved well. The follow-up is 126 months at maximum<br />

and 74 months on average. There is 3 locoregional recurrences and 14<br />

distant metastases. 5-year survival rate is 97.3%. With regard to TARM,<br />

The skin incision only in the axilla made better looks and shapes <strong>of</strong> the<br />

breast. It could be applied for tumors in any area <strong>of</strong> the breast without<br />

tumor nipple extension. The reconstruction with oxidized cellulose needs<br />

no excessive detachment <strong>of</strong> the skin beyond the surgical margin. The<br />

postoperative esthetic results were excellent and better. The sensory<br />

disturbance was minimal. All patients expressed great satisfaction.<br />

Conclusions: VABS can be considered as a good surgical procedure<br />

concerning locoregional control and esthetics. TARM is better on the<br />

patients without tumor nipple extension.<br />

1111 General Poster Session (Board #32D), Sat, 8:00 AM-12:00 PM<br />

Magnetic resonance imaging (MRI) evaluation <strong>of</strong> pathologic complete<br />

response (pCR) in different breast cancer subtypes after neoadjuvant<br />

chemotherapy (NAC). Presenting Author: Lucia Gonzalez-Cortijo, Hospital<br />

Universitario Quiron Madrid, Madrid, Spain<br />

Background: MRI is being used to address treatment response to NAC in<br />

breast cancer patients. However, its ability to predict pCR in histologically<br />

different tumors remains unclear. We tried to investigate the usefulness <strong>of</strong><br />

MRI in evaluation <strong>of</strong> pCR in different breast cancer subtypes after<br />

treatment with NAC. Methods: Serial MRI studies were acquired before,<br />

during and after NAC in 75 evaluable patients. MRI interpretation included<br />

lesion size, morphology and dynamic enhanced evaluation imaging with<br />

initial and late enhancement. On the basis <strong>of</strong> the final MRI, response was<br />

determined to be a clinically complete response (CCR) when no residual<br />

tumor and no late enhancement were found. By using inmunohistochemistry<br />

and fluorescence in situ hybridization (FISH) for human epidermal<br />

growth factor receptor 2 (HER2/neu) amplification, tumors were divided<br />

into three subtypes: triple negative, HER2 positive, and estrogen receptor<br />

(ER) positive/HER2 negative. Every patient received chemotherapy with<br />

taxanes and anthracyclines and HER2 positive tumors were treated with<br />

trastuzumab. All patients received surgery. pCR was defined as no residual<br />

invasive tumor in the surgical specimen. Ductal carcinoma in situ residual<br />

disease was considered pCR. Results: 22 <strong>of</strong> 75 patients (29%) achieved a<br />

CCR on the final MRI. Of 22 patients with CCR all 22 (100%) were<br />

confirmed pathologically. 19 were pathologic complete responses and 3<br />

showed in situ microscopic residual disease. 12 (55%) were HER2 positive<br />

tumors, 4 (18%) were triple negative tumors and 6 (27%) were ER<br />

positive/HER2 negative tumors. The negative predictive value <strong>of</strong> MRI for<br />

predicting pCR after NAC was 100%. Conclusions: Absence <strong>of</strong> both residual<br />

tumor and late enhancement in MRI predict pCR with high accuracy in<br />

triple negative, HER2 positive and ER positive/HER2 negative breast<br />

cancer after NAC.<br />

1113 General Poster Session (Board #32F), Sat, 8:00 AM-12:00 PM<br />

The impact <strong>of</strong> primary surgery on stage IV breast cancer. Presenting Author:<br />

Ella Harris, Breastcheck, Dublin, Ireland<br />

Background: The role <strong>of</strong> primary surgery in metastatic breast cancer is<br />

unclear. Here in we have performed metaanalysis on available data to<br />

assess the role <strong>of</strong> surgery on oncological outcome in patients with stage IV<br />

breast cancer. Methods: A comprehensive search for published trials that<br />

examined outcome following removal <strong>of</strong> primary disease in stage IV breast<br />

cancer was performed using MEDLINE and cross referencing available<br />

data. Reviews <strong>of</strong> each study were conducted, and data were extracted.<br />

Primary outcome was overall survival related to surgical removal <strong>of</strong> primary<br />

disease. Results: We identified 15 relevant studies <strong>of</strong> which 10 were<br />

appropriate for analysis. Data was available on 28,693 patients with stage<br />

IV disease, <strong>of</strong> whom 52.8% underwent removal <strong>of</strong> the primary carcinoma.<br />

Patients undergoing primary surgery in this setting were more likely to be<br />

alive at 3 years 40% vs. 22% (OR 2.32 CI 2.08-2.6, p�0.01 (surgery vs.<br />

no surgery)). Analysis <strong>of</strong> subgroups for selection to surgery or not, favoured<br />

smaller tumours, fewer comorbidities, fewer metastases (p�0.01). There<br />

was no difference between the two groups in location <strong>of</strong> metastases, grade<br />

<strong>of</strong> tumour or receptor status. Conclusions: Patients undergoing removal <strong>of</strong><br />

primary carcinoma in the setting <strong>of</strong> stage IV breast cancer appear to have an<br />

improved overall survival. However the available data suggest that these<br />

surgical patients probably have better prognosis stage IV disease than those<br />

patients not undergoing surgery.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


1114 General Poster Session (Board #32G), Sat, 8:00 AM-12:00 PM<br />

Prognostic impact <strong>of</strong> local therapy <strong>of</strong> the primary tumor in metastatic breast<br />

cancer. Presenting Author: Isabelle Katrin Himsl, Ludwig Maximilians<br />

University Munich, Munich, Germany<br />

Background: MBC is an incurable disease and the treatment aims are<br />

palliative. It is not known whether the difference in OS is the result <strong>of</strong> a<br />

selection bias or caused by dissemination <strong>of</strong> tumor stem cells in pat.<br />

without surgery. Methods: To identify the impact <strong>of</strong> surgical therapy <strong>of</strong> the<br />

primary tumor, a mono-institutional retrospective review from 1990-2006<br />

was done in primary MBC pts. Results: We identified 269 pts. with primary<br />

MBC, 63 <strong>of</strong> whom had received no surgical local treatment. Mean follow up<br />

is 65 m for pts., observed mortality 87%. Location <strong>of</strong> metastases were bone<br />

only (36%), visceral or s<strong>of</strong>t tissue (one organ only, 19%), multiple organs<br />

(40%) and including CNS metastases (5%). 50% had G3 tumors, 25%<br />

negative receptor status, 7% non-resectable local disease and 57%<br />

symptomatic metastases. In univariate analysis, pat. without local treatment<br />

had a median OS <strong>of</strong> 14.4m, pts. with local therapy 28.1m (p�0.001).<br />

Pts. not receiving local treatment were significantly more likely to have<br />

multiorgan or CNS involvement (p� 0.001), symptoms at diagnosis<br />

(p�0.009), non-resectable tumor (p�0.001) and were more likely to die<br />

within the first 30d after diagnosis (p� 0.001). In multivariate analysis,<br />

local treatment had no significant impact on OS. The only significant<br />

variables were: number <strong>of</strong> involved organs, symptoms at diagnosis, receptor<br />

status, grading, and size <strong>of</strong> the local tumor. The effect <strong>of</strong> local treatment on<br />

OS was not homogenous across subgroups. Local treatment was a significant<br />

factor in tumors with only one involved organ or asymptomatic disease.<br />

In all other groups, local treatment did not result in an OS benefit.<br />

Conclusions: Our cohort showed significantly improved OS in univariate<br />

analysis if the breast primary tumor had been removed in metastatic<br />

disease. Yet, the decision for local treatment was biased by the extent and<br />

presentation <strong>of</strong> metastatic disease. Pts. with more advanced MBC seem not<br />

to benefit from removal <strong>of</strong> the primary tumor. However, we see significant<br />

influence in pts. with limited and asymptomatic MBC. The potential<br />

dissemination <strong>of</strong> tumor stem cells from the breast primary in metastatic but<br />

locally untreated disease may only influence prognosis in pts. with limited<br />

disease.<br />

1116 General Poster Session (Board #33A), Sat, 8:00 AM-12:00 PM<br />

Postmastectomy radiotherapy for patients with one to three positive lymph<br />

nodes: Utilization and benefit. Presenting Author: Dezheng Huo, The<br />

University <strong>of</strong> Chicago, Chicago, IL<br />

Background: The use <strong>of</strong> postmastectomy radiotherapy (PMRT) for patients<br />

with pT1-2pN1 tumors is controversial and ASCO guidelines indicate that<br />

there is insufficient evidence to make recommendation. We hypothesized<br />

that the use <strong>of</strong> PMRT in this patient group was low and has minimal impact<br />

on survival. Methods: The study includes 83,742 invasive breast cancer<br />

patients from the National Cancer Data Base who underwent mastectomy<br />

with pT1-2 and pN1 disease from 1998-2007. Neoadjuvant cases were<br />

excluded. We investigated factors related to PMRT use using cross tables<br />

and logistic regression. Survival analysis was conducted using Cox models<br />

in patients diagnosed from 1998-2002, with a median follow-up <strong>of</strong> 5.5<br />

years. Results: The proportion <strong>of</strong> N1 patients undergoing PMRT remained<br />

stable from 1998 to 2007, at approximately 20%. PMRT use increased<br />

with larger tumor size (15.4% in T1N1 and 24.4% in T2N1), and with<br />

increasing positive lymph nodes (14.6%, 23.7%, and 35.2% for patients<br />

with one, two, or three positive nodes, respectively). Age was significantly<br />

inversely correlated with PMRT use: the proportion <strong>of</strong> patients receiving<br />

PMRT was 31.3% for age �40 years and 8.2% for 80� years (p�0.001).<br />

Asians are more likely to receive PMRT (25.5%), compared to other races<br />

(20.3% white, 20.7% black, and 20.6% Hispanic; p�0.001). PMRT also<br />

varied considerably by facility location, the highest in the Northeast at<br />

31.3%, and the lowest in the South at 15.8% (p�0.001). There was only<br />

minor difference in PMRT use between different types <strong>of</strong> cancer centers.<br />

Insurance status, income and education level were not associated with<br />

PMRT use. After adjusting for prognostic factors in the Cox models, PMRT<br />

use was associated with a reduced mortality (hazard ratio�0.87, 95% CI:<br />

0.81-0.93; p�0.001). The multivariable-adjusted 5-year death rate was<br />

16.1% in patients receiving PMRT and 18.1% in patients not receiving<br />

PMRT. For pT1N1 tumors the absolute benefit was 1.3% compared to<br />

2.7% for pT2N1 tumors. Conclusions: PMRT use varies with facility and<br />

clinicopathologic factors, but not socioeconomic factors. The risks <strong>of</strong><br />

radiation need to be weighed against the 2% absolute survival benefit when<br />

deciding on whether to use PMRT for pT1-2N1 patients.<br />

Breast Cancer—Triple-Negative/Cytotoxics/Local Therapy<br />

77s<br />

1115 General Poster Session (Board #32H), Sat, 8:00 AM-12:00 PM<br />

Role <strong>of</strong> breast surgery in T1-T3 breast cancer patients with synchronous<br />

bone metastases. Presenting Author: Edoardo Botteri, European Institute<br />

<strong>of</strong> Oncology, Division <strong>of</strong> Epidemiology and Biostatistics, Milan, Italy<br />

Background: The role <strong>of</strong> breast surgery in advanced breast cancer (ABC) is<br />

controversial. The main potential advantage <strong>of</strong> removing the primary tumor<br />

is to eliminate the source <strong>of</strong> further metastatic spread. While previous<br />

studies addressed the question in very heterogeneous populations (e.g.<br />

patients with any local and distant extension), we have focused on a<br />

homogeneous series <strong>of</strong> ABC patients. Methods: From our institutional<br />

Tumor Registry we selected 191 consecutive women diagnosed between<br />

2000 and 2008 with locally operable (T1-T3) ABC, synchronous bone<br />

metastases and no other distant sites involved. The progression free<br />

survival (PFS) was calculated from diagnosis to the date <strong>of</strong> progression,<br />

defined as either a new site <strong>of</strong> metastatic disease or clinical/radiographic<br />

evidence <strong>of</strong> increasing tumor burden at a previously known bone metastatic<br />

site. Results: Median age was 51 years and 92% <strong>of</strong> the women had an<br />

endocrine-responsive tumor. One-hundred and thirty patients out <strong>of</strong> 191<br />

(68%) underwent surgery at the time <strong>of</strong> diagnosis, while 61 (32%) did not.<br />

Twenty-six <strong>of</strong> the operated patients (20%) had previously undergone<br />

neoadjuvant chemotherapy; 15 (12%) had positive or undetermined<br />

surgical margins. Operated and non-operated patients were similar with<br />

respect to age, tumor size, nodal involvement, estrogen and progesterone<br />

receptor status, HER2 overexpression and Ki-67, but differed in terms <strong>of</strong><br />

number <strong>of</strong> bone metastatic sites: a single metastasis was detected in 34<br />

(26%) operated and 7 (11%) non-operated cases (P�0.02). First-line<br />

treatment strategies with endocrine therapy, chemotherapy and Trastuzumab<br />

were similarly distributed between the two groups. The 5-year PFS<br />

was 22.0% and 10.4% in operated and non-operated patients, respectively.<br />

The multi-adjusted hazard ratio was 0.62 (95% confidence interval<br />

0.39-0.98) in favor <strong>of</strong> surgery. The exclusion <strong>of</strong> the patients who had<br />

received neoadjuvant chemotherapy and patients with positive or undetermined<br />

surgical margins did not alter the results. Conclusions: In this large<br />

and homogeneous series <strong>of</strong> ABC patients with synchronous bone metastases,<br />

the role <strong>of</strong> breast surgery had a favorable impact on the progression <strong>of</strong><br />

the disease, indicating a potential survival benefit.<br />

1117 General Poster Session (Board #33B), Sat, 8:00 AM-12:00 PM<br />

Patient factors and satisfaction in the choice <strong>of</strong> contralateral prophylactic<br />

mastectomy. Presenting Author: Amanda Kathleen Arrington, City <strong>of</strong> Hope,<br />

Duarte, CA<br />

Background: The percentage <strong>of</strong> women undergoing contralateral prophylactic<br />

mastectomy (CPM) has more than doubled in recent years. The<br />

underlying reasons patients choose CPM have not been fully evaluated. Our<br />

objective was to survey patients who have undergone a unilateral mastectomy<br />

with or without CPM to identify reasons surrounding their decisions.<br />

Methods: After obtaining IRB approval, a 30-question cross-sectional<br />

validated survey was mailed to 691 patients who underwent mastectomy<br />

from 1972 to 2011 and are receiving treatment or surveillance at City <strong>of</strong><br />

Hope. The questionnaire queried the factors behind the choice <strong>of</strong> surgery<br />

for each patient. Demographic questions were included and patient charts<br />

were also reviewed. Results: The overall response rate was 53% (N�368).<br />

Patients were classified into those who underwent mastectomy with CPM<br />

(N�139, 38%) and those who underwent mastectomy without CPM<br />

(no-CPM) (N�229, 62%). Of returned surveys, the median age was 50;<br />

24% <strong>of</strong> patients reported a family history <strong>of</strong> breast cancer (42% CPM vs.<br />

13% no-CPM, p�0.0001) and 80% <strong>of</strong> patients had education beyond the<br />

high school level (87% CPM vs. 77% no-CPM, p�0.013). PM patients<br />

reported being “very concerned” about breast cancer more <strong>of</strong>ten than<br />

no-CPM patients (46% vs. 34%, p�0.033).The primary reasons for CPM<br />

were: concern <strong>of</strong> recurrence (55%), cosmetic symmetry (27%), physician<br />

recommendation (17%), and unclear pre-operative imaging (9%). When<br />

questioned about regrets, the top response was decreased sensation<br />

(26%). Although 81% <strong>of</strong> CPM patients were “very satisfied” with their<br />

decision, 32% <strong>of</strong> no-CPM patients reported the same level <strong>of</strong> satisfaction<br />

with their decision (p�0.0001). For no-CPM patients, the primary reasons<br />

for the choice <strong>of</strong> no-CPM was physician advice and “monitoring is<br />

sufficient”; with 18% <strong>of</strong> the responders still considering a CPM. Conclusions:<br />

Patients’ perceived risk <strong>of</strong> contralateral breast cancer is the primary reason<br />

for CPM. CPM patients tend to be more satisfied with their decision<br />

compared to no-CPM patients. This may be related to the active decisionmaking<br />

thought processes and education necessary to choose CPM. Further<br />

patient education is warranted to minimize the risk <strong>of</strong> regret in making this<br />

decision.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


78s Breast Cancer—Triple-Negative/Cytotoxics/Local Therapy<br />

1118 General Poster Session (Board #33C), Sat, 8:00 AM-12:00 PM<br />

Predictors <strong>of</strong> recurrence in postmastectomy patients with one to three<br />

positive lymph nodes. Presenting Author: Tracy-Ann Moo, New York<br />

Presbyterian Hospital, Weill Cornell Medical Center, New York, NY<br />

Background: Although the role <strong>of</strong> post mastectomy radiation therapy<br />

(PMRT) is well established in women with � 4 positive axillary lymph nodes<br />

(ALN), its indications in patients with 1-3 positive ALN is controversial. A<br />

recent large meta-analysis suggested a survival benefit in patients with 1-3<br />

positive ALN who received PMRT. However, because recurrence rates in<br />

this group are low, identifying a subgroup <strong>of</strong> patients at higher risk for<br />

locoregional recurrence (LRR) could aid decision-making about PMRT.<br />

Methods: From an institutional database, 1,333 breast cancer patients who<br />

underwent mastectomy between 1996 and 2006 and had 1-3 positive ALN<br />

were identified. Among these, T3/T4 tumors and those who received PMRT<br />

were excluded. 926 patients were analyzed. The Kaplan-Meier method and<br />

Cox regression was used to explore clinicopathologic features that predicted<br />

LRR, distant metastases (DM), and recurrence-free survival (RFR).<br />

Results: Median follow-up was 7yrs. LRR occurred in 49 patients and DM in<br />

126 patients. LRR and/or DM occurred in 146. On univariate analysis,<br />

factors significantly affecting LRR recurrence were increasing tumor size<br />

(p�0.04), age �50 (p�0.003), histologic grade (p�0.03), nuclear grade<br />

(p�0.008), lymphovascular invasion (LVI) (p�0.0001), and macroscopic<br />

ALN metastases (p�0.02). On multivariate analysis, age �50 (p�0.0012)<br />

and the presence <strong>of</strong> LVI (p�0.0001) predicted a higher LRR; increasing<br />

tumor size (p�0.0005), age �50 (p�0.04), higher histologic grade<br />

(p�0.01), number <strong>of</strong> positive ALN (p�0.04), LVI (p�0.02), macroscopic<br />

ALN metastases (p�0.02), and no chemotherapy (p�0.02) predicted a<br />

significantly lower RFR. Conclusions: Pts with T1-2 tumors and 1-3 positive<br />

ALN are at low risk <strong>of</strong> isolated LRR; however, patients �50 and with LVI<br />

may have additional risk that warrants consideration <strong>of</strong> PMRT.<br />

1120 General Poster Session (Board #33E), Sat, 8:00 AM-12:00 PM<br />

Feasibility <strong>of</strong> sentinel node biopsy in patients with locally advanced breast<br />

cancer after neoadjuvant therapy: A pilot study. Presenting Author: Ariadna<br />

Tibau Martorell, Hospital de la Santa Creu i Sant Pau, Medical Oncology<br />

Department, Barcelona, Spain<br />

Background: Sentinel lymph node biopsy (SLNB) is a widely used staging<br />

method for patients with early breast cancer. Neoadjuvant Therapy (NT)<br />

modifies the anatomical conditions in the breast and axilla, and thus<br />

reliability <strong>of</strong> SLNB after NT remains controversial. The aim <strong>of</strong> this study is<br />

to prospectively evaluate the feasibility and accuracy <strong>of</strong> this procedure in<br />

this particular group <strong>of</strong> patients. Methods: Between December 2007-2011,<br />

69 patients (mean age 56 years) with locally advanced breast cancer<br />

(LABC) were prospectively studied. Patients were T1-4, N0-1, M0. Prior to<br />

surgery, 61 patients received chemotherapy (CT) (adryamicin/cyclophosphamide<br />

followed by docetaxel) and 8 patients endocrine therapy (ET). Thirty<br />

nine patients were initially node-negative (cN0) and 30 patients had<br />

clinical/ultrasound node-positive confirmed by cytology (cN1) at presentation.<br />

All patients were clinical and ultrasound node-negative after NT. The<br />

study contained two groups <strong>of</strong> patients: group A (validation) included the<br />

first 29, associated with an axillary lymph node dissection (ALND) after NT,<br />

in order to validate the study, and group B included the last 40, only<br />

associated with an ALND when SLNB was positive or not found. Results:<br />

Whole SLNB identification rate was 89.9%, and no significant differences<br />

were found between patients initially cN0 (92%; 36/39) and initially cN1<br />

(87%; 26/30). Four <strong>of</strong> 7 patients in whom SLNB was not found had<br />

residual nodal metastasis after NT (3 <strong>of</strong> them were initially cN1). Sentinel<br />

lymph nodes were successfully identified in 87% (7/8) <strong>of</strong> patients after ET<br />

and in 90% (55/61) <strong>of</strong> patients after CT. There was one false negative (FN)<br />

case after CT in group A (9% <strong>of</strong> overall false negative rate, initially cN0) and<br />

there were no FN cases after ET. Positive SLNB were higher in initially cN1<br />

group (53%; 16/30) than in initially cN0 group (18%; 7/39). Conclusions:<br />

SLNB after NT (CT or ET) is safe and feasible in patients with LABC, not<br />

only in initially cN0 but also in initially cN1. It accurately predicts the<br />

status <strong>of</strong> the axilla and avoids unnecessary ALND.<br />

1119 General Poster Session (Board #33D), Sat, 8:00 AM-12:00 PM<br />

Radi<strong>of</strong>requency ablation (RFA) <strong>of</strong> breast cancer: A multicenter retrospective<br />

analysis. Presenting Author: Toshikazu Ito, Rinku General Medical<br />

Center, Izumisano, Japan<br />

Background: Local ablative therapy <strong>of</strong> breast cancer represents the next<br />

frontier in the minimally invasive breast-conservation treatment. We<br />

performed a retrospective study <strong>of</strong> ultrasound-guided percutaneous radi<strong>of</strong>requency<br />

ablation (RFA) <strong>of</strong> breast cancers to determine safety and<br />

complication related to this treatment. Methods: Four hundred and ninetyseven<br />

patients with core biopsy proven breast carcinoma in 10 institutions<br />

<strong>of</strong> non-surgical ablaton study group underwent RFA without surgical<br />

excision were enrolled in this study. Results: Mean patient age was 54 years<br />

(range 22 - 92 years). Mean tumor size was 1.6 cm. Four hundred and<br />

twenty-five tumors ( 86 %) were � 2 cm. The median follow-up period was<br />

50 months (range 3–92months). The mean required for ablation was 19<br />

minutes (range, 4- 72 minutes), and the average temperature <strong>of</strong> the tumor<br />

after ablation was 91 degrees Celsius. The local recurrence rate after RFA<br />

was higher in tumors <strong>of</strong> negative estrogen receptor (8 <strong>of</strong> 78, 10%) than in<br />

tumors <strong>of</strong> positive estrogen receptor (17 <strong>of</strong> 437, 4%; p�0.05), and was<br />

higher in tumors <strong>of</strong> positive HER2/neu than in tumors <strong>of</strong> negative<br />

HER2/neu (14.9% vs. 3.2%; p�0.01). The local recurrence rate after RFA<br />

was higher in tumors <strong>of</strong> positive node than in tumors <strong>of</strong> negative node<br />

(9.8% vs. 3.6%), and was higher in tumors without irradiation than in<br />

tumors with irradiation (18.2% vs. 3.2%; p�0.001). The local recurrence<br />

rate after RFA was higher in tumors <strong>of</strong> � 2 cm (13 <strong>of</strong> 72, 18%) than in<br />

tumors <strong>of</strong> � 2 cm (11 <strong>of</strong> 425, 3%; p�0.001). RFA-relating adverse events<br />

were observed in 17 patients <strong>of</strong> local pain, 14 patients <strong>of</strong> skin burn and 4<br />

patients <strong>of</strong> retraction <strong>of</strong> nipple. Conclusions: RFA is considered to be a safe<br />

and promising minimally invasive treatment <strong>of</strong> small breast cancer � 2cm<br />

in diameter. Further studies are necessary to optimize the technique and<br />

evaluate its future role as local therapy for breast cancer.<br />

1121 General Poster Session (Board #33F), Sat, 8:00 AM-12:00 PM<br />

Can primary tumor markers <strong>of</strong> cancer-initiating cells predict lymph node<br />

positivity in breast cancer patients? Presenting Author: Anees B. Chagpar,<br />

Yale University, New Haven, CT<br />

Background: Cancer initiating cells, characterized by ALDH1 positivity<br />

and/or colocalization <strong>of</strong> ALDH1 and CD44, have been shown to be<br />

associated with poor prognosis in breast cancer patients. The prognostic<br />

value <strong>of</strong> these tumor markers with respect to prediction <strong>of</strong> lymph node (LN)<br />

status remains unclear. Methods: Tissue microarrays from a cohort <strong>of</strong> 223<br />

breast cancer patients diagnosed between 2003 and 2007 were evaluated<br />

using the AQUA method for quantitative immun<strong>of</strong>luorescence for CD44<br />

and ALDH1. These data, along with other clinicopathologic data, were<br />

correlated with LN positivity. Results: The median patient age <strong>of</strong> the cohort<br />

was 56 (range; 26-89), with a median tumor size <strong>of</strong> 1.5 cm. 72 (32.0%)<br />

patients were LN positive. The median number <strong>of</strong> LNs excised was 3 (range;<br />

1-27). Of the LN positive patients, the median number <strong>of</strong> positive LNs was<br />

1.5 (range; 1-24). Levels <strong>of</strong> CD44, ALDH1, and ALDH1 colocalizing with<br />

CD44 did not correlate with number <strong>of</strong> positive LNs (Spearman rho<br />

coefficients: -0.042, 0.131, and 0.058, respectively), nor overall LN<br />

status. Tumor size and lymphovascular invasion (LVI) were the only factors<br />

found to be significantly correlated with LN status. Conclusions: While<br />

ALDH1 colocalized with CD44 has been found to be associated with poor<br />

prognosis in breast cancer patients, these markers do not predict LN status.<br />

Given that the only factors that reliably predict LN status are tumor size and<br />

LVI, further work is required to find primary tumor markers that may predict<br />

LN status in order to spare patients axillary surgery.<br />

Factor LN - LN � P value<br />

Median total CD44 46.6 41.6 0.499<br />

Median total ALDH1 390.6 442.4 0.193<br />

Median ALDH1 colocalizing with CD44 413.6 454.8 0.562<br />

Median patient age 53 52 0.083<br />

Median tumor size 1.25 2.40 �0.001<br />

LVI� 15.2% 46.7% �0.001<br />

ER� 78.7% 85.7% 0.332<br />

PR� 69.9% 73.0% 0.738<br />

HER2-neu� 12.8% 13.2% 1.000<br />

High-tumor grade 23.1% 27.0% 0.092<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


1122 General Poster Session (Board #33G), Sat, 8:00 AM-12:00 PM<br />

Association <strong>of</strong> pathologic complete response following neoadjuvant chemotherapy<br />

with survival among young women with breast cancer. Presenting<br />

Author: Rachel Adams Greenup, Massachusetts General Hospital, Boston,<br />

MA<br />

Background: Neoadjuvant chemotherapy is increasingly being used in the<br />

treatment <strong>of</strong> breast cancer, yet data on efficacy and significance <strong>of</strong><br />

pathologic complete response (pCR) is limited among young women. We<br />

sought to determine whether timing <strong>of</strong> chemotherapy impacted diseasefree<br />

(DFS) or overall survival (OS), and whether pCR is associated with<br />

improved prognosis among young women with breast cancer. Methods: We<br />

performed an IRB-approved review <strong>of</strong> women �40 years old who received<br />

treatment for stage I-III breast cancer during 1996-2008 at our institution.<br />

DFS and OS were determined through use <strong>of</strong> state tumor registry, death<br />

certificate data, and Social Security Master Death Index. Tumor biology was<br />

categorized as hormone receptor positive (HR�), HER-2�, or triple<br />

negative (TN) breast cancer. pCR was defined as lack <strong>of</strong> invasive cancer in<br />

the breast and axilla on final pathologic review. Cox regression analyses<br />

were conducted to evaluate the hazard ratios (HRs) <strong>of</strong> the association<br />

between chemotherapy and outcomes. Results: 370 women �40 years old<br />

(median age � 36.5, range: 22-40) were treated with systemic therapy for<br />

stage I-III breast cancer. 54.7% <strong>of</strong> tumors were HR�, 20.9% were<br />

HER-2�, and 24.4% were TN. After adjusting for stage, there was no<br />

difference in DFS or OS among women who received neoadjuvant versus<br />

adjuvant chemotherapy (p�0.6 and 0.5 respectively). pCR following<br />

neoadjuvant chemotherapy was higher among HER-2� (50%) and TN<br />

(28.6%) tumors when compared to HR� tumors (17.6%). Among women<br />

who received neoadjuvant chemotherapy, 10-year DFS and OS rates were<br />

significantly higher when pCR was achieved when compared to lack <strong>of</strong> pCR<br />

(HR�0.20, p value�0.01 and HR�0.13, p�0.05). pCR with neoadjuvant<br />

chemotherapy trended towards higher 10-year DFS and OS when compared<br />

to women who received adjuvant chemotherapy (HR�0.30, p value�0.08<br />

and HR�0.20, p�0.1). Conclusions: pCR after neo-adjuvant chemotherapy<br />

is associated with improved disease-free and overall survival in<br />

young women with breast cancer. Pathologic complete response may be a<br />

valuable surrogate marker for survival, and aid in the evaluation <strong>of</strong><br />

therapeutic efficacy in young breast cancer patients.<br />

1124 General Poster Session (Board #34A), Sat, 8:00 AM-12:00 PM<br />

<strong>Clinical</strong> findings and outcomes from MRI staging <strong>of</strong> breast cancers in<br />

women. Presenting Author: Akshara Raghavendra, University <strong>of</strong> Southern<br />

California Norris Comprehensive Cancer Center, Los Angeles, CA<br />

Background: For patients diagnosed with breast cancer, case series have<br />

shown that staging MRI can detect occult breast cancers in 1-10% <strong>of</strong><br />

cases. Prevalence and risk factors in underserved populations remain<br />

unclear. Methods: We performed a retrospective analysis <strong>of</strong> all patients,<br />

newly diagnosed, with breast cancer who had a preoperative staging MRI<br />

seen at Norris Comprehensive Cancer Center and LAC �USC, that cares for<br />

an underserved and minority population, from 2006 to 2011. Demographic,<br />

clinicopathologic and imaging data were obtained through a review<br />

<strong>of</strong> electronic records. Non index lesions were defined as those not known to<br />

be malignant, not presenting with clinical, mammographic or ultrasound<br />

findings, in a different quadrant and given an MRI BIRADS score <strong>of</strong> 4 or 5.<br />

Results: A total <strong>of</strong> 718 patients were analyzed and 148 patients (21%) had<br />

a total <strong>of</strong> 187 non index lesions; 63% were ipsilateral, 26% contralateral<br />

and 11% bilateral. As initial evaluation <strong>of</strong> non-index ipsilateral lesions, 71<br />

(38%) had biopsy, 24 (13%) had excision and 34 (18%) had mastectomy.<br />

For contralateral non-index lesions, 41 (22%) had contralateral biopsy, 6<br />

(3%) had excision and 11(6%) had mastectomy. Among all non index<br />

lesions, 111 (59%) were benign, 14 (7%) DCIS and 62 (33%) invasive<br />

cancer. Occult ipsilateral cancer was detected in 50 (6.9%) <strong>of</strong> patients and<br />

contralateral in 10 (1.4%) and bilateral in 6 (0.8%). Conclusions: The<br />

occult cancer detection rate with staging MRI was in this 9.2% <strong>of</strong> this<br />

diverse population. No clear risk factors were identified, with detailed<br />

factors, including BRCA status to be updated and reanalyzed.<br />

Variable Total series Non index lesions Occult cancers<br />

Total 718 148 66<br />

Hispanic (304/631) 48% (63/125) 50% (18/55) 33%<br />

African <strong>American</strong> (50/631) 8% (10/125) 8% (3/55) 5%<br />

Caucasian (193/631) 31% (36/125) 29% (25/55) 45%<br />

Asian (66/631) 10% (11/125) 9% (6/55) 11%<br />

Others (17/631) 3% (4/125) 3% (3/55) 5%<br />

Age: 50 (422/718) 59% (82/148) 55% (45/66) 68%<br />

Hormone receptor�/HER2– (288/513) 56% (56/94) 60% (25/39) 64%<br />

HER2� (105/478) 22% (24/92) 26% (10/39) 26%<br />

Triple negative (75/569) 13% (10/108) 9% (3/43) 7%<br />

Breast Cancer—Triple-Negative/Cytotoxics/Local Therapy<br />

79s<br />

1123 General Poster Session (Board #33H), Sat, 8:00 AM-12:00 PM<br />

Long-term rates <strong>of</strong> breast cancer in a population <strong>of</strong> women with ductal<br />

carcinoma in situ treated by breast-conserving surgery. Presenting Author:<br />

Eileen Rakovitch, Institute for <strong>Clinical</strong> Evaluative Sciences, Sunnybrook<br />

Health Sciences Centre, Toronto, ON, Canada<br />

Background: Ductal Carcinoma in Situ (DCIS) is a non-invasive form <strong>of</strong><br />

breast cancer which is <strong>of</strong>ten treated by breast-conserving surgery. The<br />

addition <strong>of</strong> radiotherapy to surgery has been shown to reduce the risk <strong>of</strong><br />

local recurrence (LR), but use <strong>of</strong> radiotherapy varies. It is not known to what<br />

extent women with DCIS are at risk for recurrent cancer due to the omission<br />

<strong>of</strong> radiation therapy. We studied a large provincial cohort <strong>of</strong> women with<br />

DCIS who were treated with breast-conserving surgery for factors which<br />

predict local recurrence and estimate the impact <strong>of</strong> radiotherapy on local<br />

recurrence and long-term rates <strong>of</strong> breast preservation. Methods: All women<br />

diagnosed with DCIS in Ontario from 1994 to 2003 were identified.<br />

Treatments and outcomes were identified through administrative databases<br />

and validated by chart review. Women treated with breast-conserving<br />

surgery, alone or with radiotherapy, were included. Survival analyses were<br />

used to study local recurrence (DCIS or invasive) in relation to patient<br />

characteristics, tumour characteristics and treatment. Results: The cohort<br />

included 3975 women who were treated with breast-conserving therapy; <strong>of</strong><br />

these, 1949 (49%) received radiation. At 10 years median follow-up, 736<br />

developed LR(19%). LR developed in 259 <strong>of</strong> 1949 women who received<br />

radiotherapy (13%) and in 477 <strong>of</strong> 2026 women who did not (24%;<br />

p�0.001). The differences were significant for both invasive LR (7% vs.<br />

14%; p�0.001) and DCIS recurrence (6% vs.9%; p�0.001). The 10-year<br />

cumulative rate <strong>of</strong> mastectomy was 13% for women who received radiotherapy<br />

compared to 17% for those who did not (p�0.01).We estimate that<br />

29% (N�214) <strong>of</strong> all local recurrences diagnosed in Ontario in women<br />

treated for DCIS between 1994 and 2003 would be prevented if all<br />

patients received radiotherapy. Conclusions: The omission <strong>of</strong> radiation<br />

therapy after breast-conserving surgery in women with DCIS resulted in a<br />

substantial number <strong>of</strong> local recurrences that might have been avoided and<br />

lower rates <strong>of</strong> breast preservation. Improvements in guidelines that facilitate<br />

the selection <strong>of</strong> women in whom radiotherapy can be avoided are<br />

needed.<br />

1125 General Poster Session (Board #34B), Sat, 8:00 AM-12:00 PM<br />

Overuse <strong>of</strong> sentinel lymph node biopsy with breast conserving surgery for<br />

clinical DCIS. Presenting Author: Natalie Beckman Jones, The Ohio State<br />

University Medical Center, Columbus, OH<br />

Background: The National Comprehensive Cancer Network (NCCN) guidelines<br />

recommend against sentinel lymph node biopsy (SLNB) for ductal<br />

carcinoma in-situ (DCIS) treated with breast conservation surgery (BCS).<br />

SLNB is appropriate with mastectomy because it precludes subsequent<br />

SLNB if invasive cancer is identified. However, SLNB is commonly<br />

performed with BCS for DCIS. We hypothesize SLNB use in the setting <strong>of</strong><br />

BCS for DCIS varies and may be over used in some cancer centers. Methods:<br />

We examined 6,070 cases with initial biopsy showing DCIS presenting to<br />

13 institutions participating in the NCCN Breast Outcomes Database from<br />

1998-2009. Receipt <strong>of</strong> SLNB was defined as SLNB performed at any point<br />

in primary treatment for those with a final diagnosis <strong>of</strong> DCIS or at the first<br />

surgical procedure for those upstaged to invasive cancer. Characteristics <strong>of</strong><br />

patients who did and did not have SLNB were compared using Chi-square<br />

tests. Logistic models adjusting for clinical and pathologic variables were<br />

performed to assess factors associated with use <strong>of</strong> SLNB. Results: Of 3,725<br />

treated with BCS, 778 (20.9%) had SLNB. Among 2,345 treated with<br />

mastectomy, 1,484 (63.3%) had SLNB. Within BCS, patients presenting<br />

with clinical symptoms (vs. screening detected) were more likely to have<br />

SLNB (p�0.0006, OR: 1.76; 95% CI 1.31-2.36). For both groups,<br />

presence <strong>of</strong> comedo necrosis, year <strong>of</strong> diagnosis, and treating institution<br />

were predictors <strong>of</strong> SLNB (p�0.0001). 1,171 (19.3%) were upstaged from<br />

DCIS at initial biopsy to invasive cancer on final pathology. 212 (18.1%<br />

invasive cancer group) had positive nodes. Use <strong>of</strong> SLNB increased over<br />

time from 1998-2009 in mastectomy group. Among BCS group, SNLB use<br />

decreased over the first half <strong>of</strong> the study period and then remained stable at<br />

approximately 15% across all centers. Conclusions: Although use <strong>of</strong> SNLB<br />

has decreased over time, a substantial percentage <strong>of</strong> patients undergoing<br />

BCS for DCIS receive SNLB. Practices vary considerably across centers.<br />

SLNB can be performed as a second procedure for those treated with BCS<br />

and identified with invasive cancer, thereby avoiding unnecessary risk <strong>of</strong><br />

significant morbidity. Breast programs should review their practices to<br />

curtail the use <strong>of</strong> unnecessary surgery for women with DCIS.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


80s Breast Cancer—Triple-Negative/Cytotoxics/Local Therapy<br />

1126 General Poster Session (Board #34C), Sat, 8:00 AM-12:00 PM<br />

Axillary lymph node status in breast cancer staging: What patient and tumor<br />

factors affect the accuracy <strong>of</strong> ultrasound-guided fine needle aspiration?<br />

Presenting Author: Celin Chacko, Montefiore Medical Center, Bronx, NY<br />

Background: The purpose <strong>of</strong> the study is to evaluate the accuracy <strong>of</strong><br />

ultrasound-guided fine needle aspiration (FNA) <strong>of</strong> axillary lymph nodes(ALNs)<br />

in patients with breast cancer and to determine factors that<br />

influence accuracy <strong>of</strong> ultrasound-guided FNA. Methods: Retrospective<br />

review <strong>of</strong> patients with breast cancer who had FNA <strong>of</strong> ALNs as well as<br />

sentinel lymph node excision or complete axillary dissection. Patients<br />

treated with neoadjuvant chemotherapy were excluded. 55 axillary FNAs in<br />

54 patients were included in the final analysis. Pathology reports were<br />

reviewed for size <strong>of</strong> the primary tumor, FNA results, number <strong>of</strong> positive<br />

ALNs, and greatest tumor size in ALNs. FNA was performed if a suspicious<br />

lymph node was identified. Surgical sentinel lymph node biopsy or full<br />

axillary dissection were the reference standard. Micrometastases (� 0.2<br />

mm) and isolated tumor cells in the lymph node were included in the<br />

negative group. Atypical and nondiagnostic FNA results were considered<br />

negative cytologic results. Significance was analyzed using the Mann-<br />

Whitney test. Results: Size <strong>of</strong> the primary cancer ranged from 0.3 mm to<br />

8.5 cm. The sensitivity <strong>of</strong> FNA was 73%, with positive predictive value <strong>of</strong><br />

97% and negative predictive value <strong>of</strong> 52%. The NPV <strong>of</strong> FNA for primary<br />

tumors �1 cm, 1.1-2, 2.1-5 and �5 cm is 100%, 36%, 50% and 66%<br />

respectively. Correlation <strong>of</strong> primary tumor size with sensitivity <strong>of</strong> FNA was<br />

not statistically significant. The sensitivity <strong>of</strong> FNA for lymph nodes with<br />

metastatic deposit � 5mm, 6-10mm, 11-15mm, 16-20mm, and 21mm�<br />

is 0%, 57%, 59%, 89%, and 100%, which is statistically significant (p �<br />

0.007). The number <strong>of</strong> positive ALNs at axillary dissection is not correlated<br />

to the sensitivity <strong>of</strong> FNA. The sensitivity <strong>of</strong> FNA for 1-3, 4-9 and 10�<br />

positive ALNs is 78%, 64% and 80%. Conclusions: Our findings indicate<br />

that FNA <strong>of</strong> suspicious axillary lymph nodes is valuable even in small<br />

tumors, which differs from the literature. The overall negative predictive<br />

value <strong>of</strong> FNA is 52%, so sentinel lymph node biopsy is essential after<br />

negative FNA. Sensitivity <strong>of</strong> FNA increases with the size <strong>of</strong> the metastatic<br />

deposit in the lymph node, but is not correlated to the number <strong>of</strong> positive<br />

ALNs found at dissection.<br />

1128 General Poster Session (Board #34E), Sat, 8:00 AM-12:00 PM<br />

Phase I/II trial <strong>of</strong> partial breast irradiation (PBI) with various concurrent<br />

chemotherapy regimens. Presenting Author: Lana Maria De Souza Lawrence,<br />

The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University,<br />

Baltimore, MD<br />

Background: Potential benefits <strong>of</strong> concurrent chemo-radiation include: shorter<br />

duration <strong>of</strong> treatment, smaller interval between surgery and adjuvant therapies,<br />

and synergistic effects. We have previously shown that PBI with concurrent dose<br />

dense doxorubicin and cyclophosphamide (ddAC) is well tolerated (Zellars JCO<br />

2009). We performed a follow-up feasibility trial <strong>of</strong> PBI delivered concurrently<br />

with other various chemotherapy regimens. Methods: Women with T1-2, N0-1<br />

breast cancer s/p lumpectomy with �2 mm margins were eligible. Chemotherapy<br />

regimen was at the discretion <strong>of</strong> the medical oncologist (Table). PBI (40.5 Gy in<br />

15 daily 2.7 Gy fractions) was delivered within the first 2 cycles <strong>of</strong> chemotherapy.<br />

Primary endpoints were hematologic and non-hematologic toxicities<br />

graded according to Common Terminology Criteria for Adverse Events manual (v.<br />

3.0). Results: Thirty-four patients enrolled with median f/u <strong>of</strong> 19.2 mos. (4.0 -<br />

38.6). Mean tumor size was 1.8 cm (�/- 0.7 cm), 71% pN0, 68% HR �, 18%<br />

Her2 �. All patients completed concurrent chemo-radiation. Three patients had<br />

a 3-8 day delay in chemotherapy (1 grade 2 thrombocytopenia; 1 Grade 2 liver<br />

enzymes; 1 T desensitization). There was 1 local (DCIS) and no regional/distant<br />

recurrences or deaths. Toxicity: 2 grade 4 neutropenia (ddAC, TCarboH); 1 grade<br />

3 neutropenia (post-AC paclitaxel); 1 Grade 3 hyponatremia and DVT (TC); 1<br />

syncope (TAC). None had � grade 2 radiation dermatitis. Conclusions: PBI and<br />

concurrent chemotherapy is associated with minimal toxicity and appears to be<br />

well tolerated. These results deserve further investigation. Funded by The Breast<br />

Cancer Research Foundation.<br />

Regimen Patients N�34 Dose/schedule Schedule No. <strong>of</strong> cycles<br />

Standard AC<br />

then P<br />

Dose dense<br />

AC then P<br />

2 A 60 mg/m 2<br />

C 600 mg/m 2<br />

P80mg/m 2<br />

12 A 60 mg/m 2<br />

C 600 mg/m 2<br />

G-CSF<br />

P80mg/m 2<br />

TAC 1 T 75 mg/m 2<br />

A50mg/m 2<br />

C 500 mg/m 2<br />

TC* 16 T 75 mg/m 2<br />

C 600 mg/m 2<br />

TCarboH 3 T 75 mg/m 2<br />

Carbo AUC6<br />

C#1 H 8mg/kg, then 6mg/kg<br />

Q 3 wks<br />

Qwk<br />

Q 2 wks<br />

Qwk<br />

4<br />

12<br />

4<br />

12<br />

Q 3 wks 6<br />

Q 3 wks 4-6<br />

Q 3 wks 6<br />

Abbreviations: C, cyclophosphamide; P, paclitaxel; T, docetaxel (Taxotere); A, doxorubicin<br />

(Adriamycin); Carbo, carboplatin; H, trastuzumab (Herceptin); G-CSF, granulocyte colony<br />

stimulating factor.<br />

*One patient had 6 cycles <strong>of</strong> TC, all others had 4 cycles.<br />

1127 General Poster Session (Board #34D), Sat, 8:00 AM-12:00 PM<br />

Breast cancer surgery during pregnancy. Presenting Author: Rakesh Surapaneni,<br />

Cooper University Hospital, Camden, NJ<br />

Background: There is limited literature on breast surgery during pregnancy.<br />

We present prospective registry data on 88 breast cancer patients who<br />

underwent breast cancer surgery during pregnancy. Methods: The Cancer<br />

and Pregnancy Registry is a voluntary international registry that prospectively<br />

collects the clinical course, treatment, and disease outcome <strong>of</strong><br />

women diagnosed with cancer during pregnancy and the perinatal and<br />

neonatal outcomes <strong>of</strong> their children. Results: We identified 88 patients who<br />

were diagnosed with breast cancer and had surgery while pregnant. 59<br />

patients (67%) underwent Mastectomy while29 patients (32%) underwent<br />

breast conserving surgery (BCS). Out <strong>of</strong> 43 patients who underwent BCS as<br />

their first surgery 13 patients (30.23%) required subsequent mastectomy<br />

during pregnancy. 15 patients (34.88%) from the BCS group and 4<br />

patients (8.69%) from the Mastectomy group had positive margins. There<br />

was no significant difference between patients who underwent mastectomy<br />

vs BCS based on Age (34.67 vs 34.72 P: 0.97), gestational age at surgery<br />

(14.05 vs 16.06 P: 0.23) or ER positivity (47.5% vs 46.4% P: 0.93). 2<br />

patients had neo-adjuvant chemotherapy. 17 patients (19.31%) had<br />

sentinel lymph node biopsy. 37 patients (42%) had a pregnancy complication.<br />

There was no difference in the rate <strong>of</strong> complication based on<br />

mastectomy vs BCS(45.8% vs 34.5% P: 0.31). There was only 1 patient<br />

(from mastectomy group) that delivered within 2 weeks <strong>of</strong> surgery. Of the<br />

17 patients (19.3%) with spontaneous preterm delivery, there was no<br />

difference between Mastectomy and BCS group (22% vs 13.2% P: 0.41).<br />

Of the 25 patients (28.4%) with birth complications, there was no<br />

significant difference between mastectomy vs BCS (30.5% vs 24.1% P:<br />

0.53). There was also no difference in mean birth weight between the<br />

groups (2598 grams vs 2672.3 grams P: 0.57). Conclusions: The data<br />

supports the safety <strong>of</strong> breast cancer surgery during pregnancy. In addition,<br />

there were no identified adverse effects in patients who underwent BCS as<br />

opposed to mastectomy. Of note, only 19% <strong>of</strong> patients underwent sentinel<br />

node biopsy which is considered the standard <strong>of</strong> care in early breast cancer<br />

patients regardless <strong>of</strong> pregnancy status.<br />

1129 General Poster Session (Board #34F), Sat, 8:00 AM-12:00 PM<br />

18F-fluorodeoxyglucose (FDG) positron emission tomography (PET) as<br />

early imaging biomarker <strong>of</strong> axillary sentinel lymph node (SLN) status in<br />

locally advanced node-positive breast cancer (NPBC) patients (pts) receiving<br />

neoadjuvant chemotherapy (NACT). Presenting Author: Yanina Dockx,<br />

Antwerp University Hospital, Edegem, Belgium<br />

Background: Use <strong>of</strong> FDG-PET in the early evaluation <strong>of</strong> NACT for NPBC is<br />

currently under investigation. The aim <strong>of</strong> this study is to assess FDG-PET in<br />

NPBC pts receiving NACT in order to identify a subset <strong>of</strong> pts that can be<br />

spared axillary lymph node dissection (ALND) in case <strong>of</strong> response. Methods:<br />

All pts (period 2009-2011) had [cT2 (�3cm)-T4, pN1-3], and no prior BC<br />

treatment. All pts received 8 cycles (cy) <strong>of</strong> NACT �/- trastuzumab.<br />

FDG-PET was performed at baseline and after 2 cy. After NACT, mastectomy<br />

with SLN and ALND was peformed. As primary endpoint, the change<br />

in maximum standardized uptake value (SUVmax) in the primary tumor (PT)<br />

and ALNs was compared with pathological response (Jonckheere-Terpstra<br />

test). Logistic regression was used to determine the predictive value <strong>of</strong> a<br />

50% reduction in SUVmax on pathological response and SLN status.<br />

Results: Forty-one pts were evaluable for the primary endpoint and 31 pts<br />

had successful SN procedures. The median age was 49.8 years (range<br />

27-75). Overall, 9.8% (4/41; 95% CI 2.7 – 23.1) <strong>of</strong> pts had progressive<br />

disease (PD), 7.3% (3/41 ; 95% CI 1.5 – 19.9 ) stable disease (SD),<br />

53.7% (22/41; 95% CI 37.4 – 69.3) partial response (PR), and 21.9%<br />

(9/41; 95% CI 10.6 – 37.6) a complete pathological response (pCR). A<br />

linear trend existed between pCR and higher decreases in SUVmax <strong>of</strong> the PT<br />

site (p�0.008), but not with lymph node SUVmax (p�0.294). The odds <strong>of</strong><br />

achieving a pCR, increased significantly when SUVmax <strong>of</strong> the PT decreased<br />

with �50% (OR 10.7; 95% CI 1.2 - 98.0; p�0.03), but not lymph node<br />

SUVmax (OR 4.75; 95% CI 0.82 - 27.5; p�0.08). Achieving a true negative<br />

SLN status was more likely with �50% reductions in PT SUVmax (OR 41.2;<br />

95% CI 4.0 - 421.9; p� 0.002). A reduction <strong>of</strong> �50% in nodal SUVmax was not predictive <strong>of</strong> negative SLN status (OR 3.33; 95% CI 0.66 - 16.8;<br />

p�0.1). Conclusions: Early changes in PT metabolism imaged with FDG-<br />

PET after only 2 cy <strong>of</strong> NACT is a promising biomarker in the management <strong>of</strong><br />

the axilla in NPBC. Molecular imaging <strong>of</strong> PT biology and to a lesser extent <strong>of</strong><br />

axillary lymph nodes using FDG-PET warrants further study.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


1130 General Poster Session (Board #34G), Sat, 8:00 AM-12:00 PM<br />

Evaluation <strong>of</strong> the stage IB designation <strong>of</strong> the 7th edition <strong>of</strong> the AJCC<br />

Staging System. Presenting Author: Nikolaos Andreas Dallas, University <strong>of</strong><br />

Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: Recent data from large cooperative group trials have questioned<br />

the relevance <strong>of</strong> small volume metastases identified in the sentinel<br />

lymph nodes (SLN) <strong>of</strong> early stage breast cancer patients. The 7th ed. <strong>of</strong> the<br />

AJCC staging system differentiates node negative patients (stage IA) from<br />

those with micrometastases (stage IB) or macrometastases (stage II or III).<br />

This study was undertaken to determine the utility <strong>of</strong> the stage IB<br />

designation. Methods: Review <strong>of</strong> a prospectively maintained database<br />

identified 3474 patients who underwent SLN biopsy between 1993 and<br />

2007. Clinicopathologic and outcomes data were recorded and patients<br />

staged according to the 7th ed. AJCC system. Recurrence-free (RFS),<br />

disease-specific (DSS) and overall survival (OS) were determined using the<br />

Kaplan-Meier method and compared using the log-rank test. Results: AJCC<br />

stage distribution included: 2246 (65%) stage IA, 207 (6%) stage IB, 685<br />

(20%) stage IIA, 209 (6%) stage IIB, and 127 (3%) stage III. For patients<br />

with stage IB disease, SLN micrometastasis was identified by H&E in 173<br />

(84%) and immunohistochemistry (IHC) in 34 (16%); suggesting that 16%<br />

would have been staged IA if enhanced evaluation had not been performed.<br />

Median follow-up was 6.1 yrs (range 0-17.2). The 5-yr RFS,DSS and OS<br />

rates for patients with stage IB disease were 98.0%, 99.5%, and 95.9%<br />

respectively, which did not differ significantly from patients with stage IA<br />

disease who had 5-year RFS,DSS and OS rates <strong>of</strong> (97.5%, p�.9), (98.8%,<br />

p�.7) and (96.2%, p�.8). When all stage I patients (IA and IB) were<br />

evaluated by ER status or grade (grade 1 vs 2 vs 3), these biologic factors<br />

were able to significantly discriminate patients with respect to RFS, DSS<br />

and OS. Conclusions: Differentiating patients with micrometastases from<br />

node negative patients does not stratify patients with respect to survival.<br />

Biologic factors (ER status and grade) are better discriminants <strong>of</strong> survival<br />

than the presence <strong>of</strong> small volume nodal metastases in patients with early<br />

stage disease. These data do not support routine use <strong>of</strong> IHC or alterations in<br />

adjuvant therapy decisions based on identification <strong>of</strong> SLN micrometastases.<br />

1132 General Poster Session (Board #35A), Sat, 8:00 AM-12:00 PM<br />

Breast cancer in older women. Presenting Author: David B. Pearlstone,<br />

Hackensack University Medical Center, Hackensack, NJ<br />

Background: Recent guidelines recommend minimizing breast cancer<br />

screening for women over the age <strong>of</strong> 75. This study examines the<br />

presentation and outcome <strong>of</strong> older patients with breast cancer to determine<br />

if increasing screening among older patients may result in better outcomes.<br />

Methods: A prospective database at Hackensack University Medical Center<br />

was queried for all patients with breast cancer diagnosed between 1/1/<br />

2006 and 1/1/2011. Numerical values compared by Student’s t -test;<br />

categorical values by Fisher exact test. Median time to events determined<br />

by Kaplan-Meier; outcomes compared by log-rank test. Results: 2200<br />

patients were identified (� 75 years, n�335, � 75 years, n�1865).<br />

Among the older cohort, mean age was 81 �/- 4 years (range: 75 – 101);<br />

mean tumor size was 2.3 �/- 0.1 cm (range: .1 – 12). Most tumors were<br />

invasive and localized (in-situ: 12.7%, localized: 57.3%, node positive:<br />

24.0%, metastatic: 5.8%) and most commonly ductal histology (ductal:<br />

77.0%; lobular: 16.6%, mixed:6.2%). Only 10.0% <strong>of</strong> patients with<br />

invasive disease and 29.2% with positive nodes received systemic therapy;<br />

90.2% underwent surgical resection. Disease-free and overall 5 year<br />

survival was 54.9% and 62.9% (in-situ: 82.2%; localized: 67.1%; node<br />

positive: 57%; metastatic 22%). Older patients had larger tumors (2.30<br />

�/- .11 v. 1.91 �/- .04 cm; p�0.001), more locally advanced (T4) disease<br />

(6.7% v. 1.7%, p�.001), more invasive disease (89% v. 84%, p�.01).<br />

Older patients had less positive family history (21.7% v. 37.3%, p�.0001).<br />

There was no difference between the groups in race, presentation with<br />

stage IV disease, tumor grade, lymphovascular invasion, histologic subtype,<br />

triple negativity, or nodal positivity. Overall and disease free five year<br />

survival were significantly worse for older patients (OS: 90.2% v. 63.9%,<br />

p� .0001; DFS: 80.9% v. 54.9%, p�.0001). Conclusions: Breast cancer<br />

among older women leads to worse outcomes compared to younger<br />

patients, regardless <strong>of</strong> histology, invasiveness <strong>of</strong> disease, hormonal status,<br />

nodal status or tumor grade. Older women present with more invasive<br />

disease, larger tumors and more locally advanced disease, despite having<br />

less family history. This implies that continued screening beyond age 75<br />

may lead to less advanced presentation and better outcomes.<br />

Breast Cancer—Triple-Negative/Cytotoxics/Local Therapy<br />

81s<br />

1131 General Poster Session (Board #34H), Sat, 8:00 AM-12:00 PM<br />

Prospective outcomes trial <strong>of</strong> immediate, implant breast reconstruction<br />

using acellular dermal matrix (POBRAD Trial): Pilot series results. Presenting<br />

Author: David Westbroek, Kings Health<strong>Part</strong>ners, London, United<br />

Kingdom<br />

Background: Acellular dermal matrices (ADM) are biological meshes <strong>of</strong><br />

dermal origin (allo- or xenografts) stripped <strong>of</strong> all cellular components<br />

leaving a structurally intact and immunologically inert extracellular matrix.<br />

They are currently in use as adjunctive subdermal scaffolding during<br />

implant breast reconstruction to optimise aesthetic outcome. Despite<br />

current use in clinical practice there is no prospective data on their clinical<br />

efficacy, associated complication rates and cost-benefit analysis. These are<br />

the endpoints <strong>of</strong> our pilot cohort series, intended to provide baseline event<br />

rates to help power a multi-centre prospective, phase II, outcomes cohort<br />

study. Methods: From July 2011 to January 2012 and with our institution<br />

review board’s approval - 20 consecutive patients undergoing immediate,<br />

implant breast reconstruction were prospectively accrued to the study.<br />

Primary endpoints: 30-day complication rates; Secondary endpoints:<br />

cosmetic outcome and cost-benefit analysis. Indications for mastectomy<br />

included therapeutic intent and/or risk reduction. The ADM used in all<br />

cases was SurgiMend PRS (TEI Bioscience Inc. Boston, MA). Results:<br />

Preliminary 30 day complication rates are in keeping with peer reviewed<br />

estimates for implant based breast reconstruction. Sub-set analysis <strong>of</strong><br />

cosmetic outcome and cost-benefit ratio’s are currently pending. In<br />

overview, ADM’s appear to deliver the aesthetic benefit <strong>of</strong> autologousimplant<br />

cover (eg. latissimus dorsi myocutaneous flap) without the downside<br />

<strong>of</strong> donorsite morbidity and prolonged in-patient admission. Conclusions:<br />

This is to our knowledge the only prospectively accrued dataset seeking to<br />

critically evaluate the clinical efficacy, cost-benefit value and enhanced<br />

aesthetic utillity <strong>of</strong> ADMs (SurgiMend PRS) in the immediate breast<br />

reconstruction setting. The early results provide new evidence in support <strong>of</strong><br />

the use ADMs in breast reconstruction and validate our intention to extend<br />

the study to a multicentre trial.<br />

1133 General Poster Session (Board #35B), Sat, 8:00 AM-12:00 PM<br />

Scoring system for prediction <strong>of</strong> having three or more involved axillary<br />

nodes for breast cancer. Presenting Author: Soo Kyung Ahn, Department <strong>of</strong><br />

Surgery, Seoul National University Hospital, Seoul, South Korea<br />

Background: Completion axillary lymph node dissection (ALND) is currently<br />

the standard <strong>of</strong> care in the event <strong>of</strong> a positive sentinel lymph node biopsy<br />

(SLNB). However, result from Z0011 indicate that women with a one or two<br />

involved axillary nodes and clinical T1-T2 tumors undergoing lumpectomy<br />

with radiation therapy followed by systemic therapy do not benefit from<br />

completion <strong>of</strong> ALND in terms <strong>of</strong> survival. The purpose <strong>of</strong> this study was to<br />

define possible predictors <strong>of</strong> having three or more involved axillary node to<br />

provide information for surgeons making decision about sparing intraoperative<br />

frozen section analysis <strong>of</strong> sentinel lymph node and completion ALND.<br />

Methods: We reviewed the records <strong>of</strong> 1215 patients with clinical T1-T2<br />

invasive breast cancer. None <strong>of</strong> these patients were in situ cancer on initial<br />

gun biopsy nor received neoadjuvant chemotherapy. Factors associated<br />

with having three or more involved axillary nodes were evaluated by<br />

univariate and multivariate logistic regression analysis. Results: Among<br />

1215 patients, 321 patients had three or more positive nodes. On a<br />

multivariate analysis, having three or more positive nodes was associated<br />

with primary tumor size by breast US, axillary LN grade according to<br />

cortical thickness by US, presence <strong>of</strong> axillary LN enlargement on chest CT<br />

and age. A scoring system to predict the probability <strong>of</strong> having three or more<br />

nodes based on patients’ data and preoperative image findings was<br />

developed from the multivariate logistic regression model. The area under<br />

the ROC curve was 0.827 (95% CI: 0.793-0.860), and negative predictive<br />

value was 90.2% for a score �2.7. Similar findings were observed for a<br />

validation dataset <strong>of</strong> 505 patients. Conclusions: Patients with a low<br />

probability <strong>of</strong> having three or more positive nodes can be identified from<br />

preoperative image finding. The scoring system developed will be helpful to<br />

surgeons making decision about sparing intraoperative frozen section<br />

analysis <strong>of</strong> sentinel lymph node and completion ALND.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


82s Breast Cancer—Triple-Negative/Cytotoxics/Local Therapy<br />

TPS1134 General Poster Session (Board #35C), Sat, 8:00 AM-12:00 PM<br />

Phase II neoadjuvant trial with carboplatin and eribulin mesylate in<br />

patients with triple-negative breast cancer. Presenting Author: Sara E.<br />

Barnato, Northwestern University, Chicago, IL<br />

Background: Several neoadjuvant trials have been conducted in triple<br />

negative breast cancer (TNBC) with platinum agents with pathologic<br />

complete response (pCR) ranging from 16%-32%. Eribulin mesylate, a<br />

nontaxane microtubule dynamics inhibitor, has clinical activity as monotherapy<br />

in breast cancer and other solid tumors. A recent phase I trial found<br />

the combination <strong>of</strong> eribulin mesylate with carboplatin was well tolerated<br />

and showed activity in advanced solid tumors. The recommended dose for<br />

future trials was eribulin mesylate 1.1 mg/m2 and carboplatin AUC6. We<br />

proposed a neoadjuvant phase II trial with the combination <strong>of</strong> carboplatin<br />

and eribulin in patients with TNBC. Methods: This is a non-randomized,<br />

open-label, multi-center, phase II clinical trial <strong>of</strong> eribulin and carboplatin<br />

enrolling histologically-confirmed TNBC patients. Our primary endpoint is<br />

to determine the pCR in TNBC patients treated with the combination <strong>of</strong><br />

carboplatin and eribulin. Secondary endpoints include determination <strong>of</strong><br />

the clinical response rate, toxicity evaluation and measurement <strong>of</strong> stem cell<br />

and TLE3 as a biomarker <strong>of</strong> response to eribulin therapy. To obtain an alpha<br />

<strong>of</strong> 0.10 and a power <strong>of</strong> 0.90, a sample size <strong>of</strong> 30 patients is required to<br />

detect a pCR rate ��30%. 10 <strong>of</strong> the planned 30 patients have been<br />

enrolled to date. Treatment will be given every 3 weeks for a total <strong>of</strong> 4 cycles<br />

<strong>of</strong> therapy. There will be an initial safety run-in to evaluate the appropriate<br />

dose <strong>of</strong> eribulin in this population. The first 10 patients will receive eribulin<br />

at 1.4 mg/m2 (intravenously over 2-5 minutes) followed by carboplatin<br />

AUC�6 (intravenously over 30 minutes). After the 10th patient has been<br />

enrolled, the study will be temporarily suspended pending review; toxicity<br />

will be assessed for these first 10 patients (cycle 1 only) to assess whether<br />

this dose <strong>of</strong> eribulin will be used for the remaining patients or if a reduction<br />

to a dose <strong>of</strong> 1.1 mg/m2 will be required. Definitive surgery will be performed<br />

3-8 weeks after completion <strong>of</strong> therapy, which will conclude the duration <strong>of</strong><br />

the study. <strong>Clinical</strong> Trial Registry Number NCT01372579.<br />

TPS1136 General Poster Session (Board #35E), Sat, 8:00 AM-12:00 PM<br />

ANZ1001 SORBET: Study <strong>of</strong> estrogen receptor beta and efficacy <strong>of</strong><br />

tamoxifen, a single arm, phase II study <strong>of</strong> the efficacy <strong>of</strong> tamoxifen in<br />

triple-negative but estrogen receptor beta-positive metastatic breast cancer.<br />

Presenting Author: Kelly-Anne Phillips, Peter MacCallum Cancer<br />

Centre, Melbourne, Australia<br />

Background: Targeted therapies are needed for triple negative breast cancer<br />

(BC). ER� is expressed in at least 20% <strong>of</strong> triple negative BCs. ER� binds<br />

estrogen and tamoxifen with a similar affinity to ER�. ER�has 5 is<strong>of</strong>orms<br />

but only ER�1 is fully functional. ER� expression has been shown to be<br />

significantly associated with improved distant disease free survival and<br />

better overall survival in tamoxifen treated ER� negative patients in<br />

retrospective studies. This “pro<strong>of</strong> <strong>of</strong> principle” study will determine the<br />

efficacy <strong>of</strong> tamoxifen in patients with triple negative but ER� positive<br />

metastatic BC. Methods: This single arm phase II study, being conducted by<br />

the Australia and New Zealand Breast Cancer Trials Group, has a Simon’s 2<br />

stage optimal design. The primary end-point is objective response rate<br />

(complete and partial responses). Progression free survival and clinical<br />

benefit rate will also be assessed. Eligibility criteria include histologically or<br />

cytologically confirmed metastatic triple negative BC (ER and PR absent,<br />

HER2 ISH negative or IHC 0 or 1) and measureable disease as per RECIST<br />

1.1. Consenting patients undergo central ER� testing and confirmation <strong>of</strong><br />

triple negative status on a metastatic biopsy sample. ER� positive patients<br />

(ER�1 nuclear staining with Allred score �4) are <strong>of</strong>fered trial participation.<br />

To date 12 potentially eligible patients have been screened for ER�; 4 had<br />

Allred score �4 (although 2 <strong>of</strong> these subsequently proved to be ineligible<br />

for the trial), 7 had Allred scores �4 and 1 result is pending. Consenting<br />

patients receive tamoxifen 20mg per oral daily until disease progression,<br />

unacceptable toxicity or withdrawal <strong>of</strong> consent. If there are �2 responses in<br />

the first stage <strong>of</strong> 28 patients, an additional 38 patients will be accrued.<br />

Tamoxifen will be considered worthy <strong>of</strong> further research if there are �6<br />

responses in the total 66 patients recruited. Current accrual is 1.<br />

Registered on ANZCTR (12610000506099).<br />

TPS1135 General Poster Session (Board #35D), Sat, 8:00 AM-12:00 PM<br />

A phase III, randomized trial <strong>of</strong> docetaxel plus carboplatin (TP) versus<br />

epirubicin plus cyclophosphamide followed by docetaxel (EC-T) as adjuvant<br />

treatment for triple-negative, early-stage breast cancer in Chinese<br />

patients. Presenting Author: Peng Yuan, Cancer Hospital, Chinese Academy<br />

<strong>of</strong> Medical Sciences, Beijing, China<br />

Background: Triple-negative [estrogen receptor (ER)-/progesterone receptor<br />

(PR)-/HER2-] breast cancer (TNBC) accounts for about 15% <strong>of</strong> all breast<br />

cancers and is associated with very poor prognosis. A combination <strong>of</strong><br />

anthracycline and taxanes represents the most commonly used adjuvant<br />

chemotherapy for TNBC patients. BRCA1, a protein responsible for repair<br />

upon DNA damage, is <strong>of</strong>ten dysfunctional in TNBC. As a result, TNBC is<br />

<strong>of</strong>ten sensitive to DNA-damaging agents (e.g., cisplatin and carboplatin). A<br />

phase II study <strong>of</strong> docetaxel plus carboplatin as neoadjuvant treatment for<br />

TNBC achieved promising response rate (Chang, CANCER, 2010). Encouraged<br />

by this important finding, we are currently conducting a phase III trial<br />

to examine docetaxel plus carboplatin as adjuvant chemotherapy in<br />

patients with early-stage TNBC (registration number at www.<strong>Clinical</strong>Trials.<br />

gov: NCT 01150513). Methods: All participants received radical mastectomy<br />

or breast-conserving surgery for pT1-3N0-2 breast cancer. All cancers<br />

were negative for ER, PR, HER2/Neu. All participants had adequate organ<br />

function and performance status. The age ranged from 18 to 70 years.<br />

Patients randomly received a TP regimen (docetaxel 100 mg/m2 plus<br />

carboplatin AUC�6 on day 1, 21 days a cycle for 6 cycles) or a EC-T<br />

regimen (epirubicin 90 mg/m2 plus cyclophosphamide 600 mg/m2 on d1,<br />

21 days a cycle for 4 cycles; followed by docetaxel 100 mg/m2 on d1, 21<br />

days a cycle for 4 cycles). Adjuvant radiotherapy was permitted in both<br />

arms. The primary endpoint is disease-free survival (DFS). Secondary<br />

endpoints included adverse event and quality <strong>of</strong> life (QoL). The study<br />

planned to recruit 500 subjects over a period <strong>of</strong> 5 years, with 5-year<br />

follow-up (once every 6 months). Target hazard ratio is 0.86 (5 year DFS <strong>of</strong><br />

74.0% vs. 71.0%). The estimated power is 0.80; 1-sided type 1 error was<br />

set at 0.05). The study was activated in June 2010 with enrollment <strong>of</strong> 110<br />

subjects.<br />

TPS1137 General Poster Session (Board #35F), Sat, 8:00 AM-12:00 PM<br />

Neoadjuvant bevacizumab with weekly nab-paclitaxel plus carboplatin<br />

followed by doxorubicin plus cyclophosphamide (AC) for triple-negative<br />

breast cancer. Presenting Author: Jessica N. Snider, University <strong>of</strong> Tennessee<br />

Health Science Center, Memphis, TN<br />

Background: Triple negative breast cancer (TNBC) predominantly clusters<br />

with “basal like” subtype on genomic pr<strong>of</strong>iling. Over-expression <strong>of</strong> Secreted<br />

Protein Acidic and Rich in Cysteine (SPARC) has been observed in basal<br />

like breast cancer (Charaffe-Jaufrett et al. Oncogene 2006; 2273-84).<br />

Endothelial transcytosis <strong>of</strong> nab- paclitaxel occurs via albumin- gp60<br />

receptor-caveolin 1 interaction. SPARC entraps the albumin resulting in<br />

higher intratumoral accumulation, which may explain the increased efficacy<br />

<strong>of</strong> nab-paclitaxel (Desai et al. Translational Oncology 2009; 59-63).<br />

Exploiting this mechanism and the dysfunctional BRCA mediated DNA<br />

repair in basal tumors, we hypothesize that nab-paclitaxel � the DNA<br />

damaging drug carboplatin would produce high response rates in TNBC.<br />

Adding bevacizumab may enhance efficacy by blocking angiogenesis. In<br />

TNBC, pathologic complete remission (pCR) to neo-adjuvant therapy<br />

correlates with better disease-free survival (DFS). We hypothesize that high<br />

pCR rates can be achieved for patients with TNBC with this combination<br />

translating to an improved DFS than seen historically. Methods: Patients<br />

with palpable and operable TNBC � 2 cm are eligible for this single stage<br />

phase II trial. pCR (defined as the absence <strong>of</strong> invasive tumor cells) in the<br />

breast is the primary end point, while pCR in the breast � axillary nodes<br />

and pCR � near pCR (residual tumor � 5mm) in the breast are secondary<br />

end points. 57 evaluable patients are needed, assuming a pCR rate <strong>of</strong> 25%<br />

vs. 40% for the null and alternate hypotheses respectively. 34 patients<br />

have been accrued to date. Patients receive carboplatin AUC 6 day 1 and<br />

nab-paclitaxel 100mg/m2 days 1, 8 and 15 <strong>of</strong> a 28 day cycle for 4 cycles<br />

and then dose dense AC for 4 cycles. Bevacizumab is given at 10mg/kg Q 2<br />

weeks with chemotherapy for the first 6 cycles. Surgery and radiation are<br />

per institutional standards. Bevacizumab is continued postoperatively to<br />

complete 1 year <strong>of</strong> treatment. A core biopsy for collection <strong>of</strong> fresh tumor<br />

tissue is required prior to the start <strong>of</strong> study treatment. Blood is collected at<br />

baseline, and after 4 and 8 cycles for biomarker analysis. Gene expression<br />

pr<strong>of</strong>iling will be undertaken for correlation with response.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


TPS1138 General Poster Session (Board #35G), Sat, 8:00 AM-12:00 PM<br />

ABT-888 (veliparib) in combination with weekly carboplatin and paclitaxel<br />

in advanced solid tumors. Presenting Author: Jorge Arturo Rios-Perez,<br />

University <strong>of</strong> Pittsburgh Cancer Institute, Pittsburgh, PA<br />

Background: The combination <strong>of</strong> paclitaxel and carboplatin is widely used<br />

for the treatment <strong>of</strong> patients with advanced solid tumors <strong>of</strong> diverse<br />

histologies. In breast cancer patients, a weekly regimen <strong>of</strong> paclitaxel has<br />

shown greater efficacy with comparable safety, when compared to everythree-weeks<br />

dosing (ECOG 1199). ABT-888 (veliparib) is an oral inhibitor<br />

<strong>of</strong> poly-ADP-ribose polymerase (PARP). Inhibition <strong>of</strong> PARP has been shown<br />

in preclinical studies to potentiate the effect <strong>of</strong> cytotoxic agents which<br />

induce DNA damage, such as platinum agents. The preclinical synergy <strong>of</strong><br />

carboplatin with veliparib and the efficacy <strong>of</strong> the combination <strong>of</strong> paclitaxel<br />

with carboplatin supports exploration <strong>of</strong> this triplet regimen. Methods: This<br />

3�3 phase I trial will seek to determine the maximum tolerated dose (MTD)<br />

<strong>of</strong> the combination <strong>of</strong> carboplatin (AUC 2), paclitaxel (80 mg/m2 ), and<br />

veliparib in patients with advanced solid tumors. Veliparib will be escalated<br />

beginning at 50 mg PO BID to a maximum <strong>of</strong> 200mg PO BID. Treatment<br />

will be given on a weekly basis over a 21-day cycle. There will be an<br />

expansion cohort <strong>of</strong> 6-12 patients with triple negative breast cancer at the<br />

maximum tolerated dose. This group <strong>of</strong> patients will undergo mandatory<br />

pre- and post-cycle 1 tumor biopsies. Secondary aims <strong>of</strong> the study include<br />

safety and toxicity <strong>of</strong> the combination, its pharmacokinetic and pharmacodynamic<br />

effects, documentation <strong>of</strong> any anti-tumor response, and assessment<br />

<strong>of</strong> the characteristics <strong>of</strong> the tumor specimens obtained in the<br />

expansion cohort that may contribute to efficacy. The latter will include<br />

whole genome microarray analysis to evaluate expression <strong>of</strong> genes involved<br />

in DNA repair pathways. Currently, the recommended phase II dose has not<br />

been determined and enrollment is ongoing on the last planned dose level<br />

(veliparib 200 mg BID).<br />

TPS1140^ General Poster Session (Board #36A), Sat, 8:00 AM-12:00 PM<br />

Phase III open-label, randomized, multicenter study <strong>of</strong> NKTR-102 versus<br />

treatment <strong>of</strong> physician’s choice (TPC) in patients (pts) with locally<br />

recurrent or metastatic breast cancer (MBC) previously treated with an<br />

anthracycline, a taxane, and capecitabine (ATC). Presenting Author: Edith<br />

A. Perez, Mayo Clinic, Jacksonville, FL<br />

Background: NKTR-102 is a topoisomerase I inhibitor-polymer conjugate<br />

that hydrolyzes to provide continuous exposure to SN-38. A phase 2 trial <strong>of</strong><br />

single-agent NKTR-102 was conducted in pts with 3rd-line MBC; 2<br />

schedules (q14d; q21d) investigated a dose <strong>of</strong> 145 mg/m2. ORR was 29%<br />

(including 3% CR) with the prior ATC subset demonstrating an ORR <strong>of</strong><br />

31%. Dosing q21d was better tolerated; in this arm, median PFS and OS<br />

equaled 5.3m and 13.1m, respectively. Trial Design: Pts will be randomized<br />

1:1 to receive single-agent NKTR-102 or TPC in an open-label,<br />

randomized, multicenter phase 3 study in pts with advanced breast cancer.<br />

Key Entry Criteria: Adult females, with ECOG 0 or 1 with adequate liver,<br />

kidney and marrow function. All pts must have received prior therapy with<br />

ATC (these drugs can be administered in the neo/adjuvant or locally<br />

advanced/metastatic setting). Prior A is not mandated if contraindicated.<br />

Prior toxicities must have resolved to � Grade 1 (except sensory neuropathy<br />

� Grade 2; complete resolution <strong>of</strong> prior diarrhea). Pts with brain metastases<br />

may be eligible, if lesions are stable for prior 3 weeks without steroids.<br />

Methods: Primary efficacy endpoint is OS. Secondary endpoints include:<br />

ORR by RECIST v1.1, clinical benefit rate (ORR�SD � 6 months), PFS<br />

and QoL. NKTR-102 is given IV at 145 mg/m2 over 90-min every 21 days<br />

without premedications. Pts randomized to TPC receive 1 <strong>of</strong> the following:<br />

eribulin, ixabepilone, vinorelbine, gemcitabine, paclitaxel, docetaxel or<br />

nab-paclitaxel (the agent must be available at the treating institution). Pts<br />

are stratified by region, prior eribulin and receptor status (TNBC, Her2� or<br />

Other). Target Accrual: ~840 pts will be required for sufficient events to<br />

occur in the planned follow-up time; OS will be compared using a two-sided<br />

log-rank test; 1 interim analysis will occur when 50% <strong>of</strong> the deaths are<br />

reported. PK sampling is performed in a subset <strong>of</strong> pts. CTCs (isolated by<br />

Apocell ApoStream technology) are serially assessed for potential predictive<br />

markers <strong>of</strong> response and toxicity. Enrollment is expected to remain<br />

open until late 2013.<br />

Breast Cancer—Triple-Negative/Cytotoxics/Local Therapy<br />

83s<br />

TPS1139 General Poster Session (Board #35H), Sat, 8:00 AM-12:00 PM<br />

Q-CROC-03: A prospective biopsy driven clinical trial to study the mechanisms<br />

<strong>of</strong> resistance to chemotherapy in triple-negative breast cancer<br />

patients. Presenting Author: Adriana Aguilar-Mahecha, Segal Cancer Center/<br />

Jewish General Hospital, McGill University, Montreal, QC, Canada<br />

Background: Resistance to chemotherapy or targeted agents is the cause <strong>of</strong><br />

death in most patients dying <strong>of</strong> breast cancer and one <strong>of</strong> the major<br />

challenges presently faced by oncologists. In triple negative breast cancers<br />

(TNBCs), drug resistance emerges quicker than in other breast cancer<br />

subtypes and contributes to the poor prognosis seen in these patients. The<br />

lack <strong>of</strong> targeted therapies to treat TNBC highlights the important need to<br />

better understand the molecular mechanisms contributing to chemotherapy<br />

resistance in order to develop new therapeutic strategies. However,<br />

the difficulty in obtaining tissue samples from drug resistant tumors has<br />

been one <strong>of</strong> the limiting factors in this field <strong>of</strong> study. Methods: We have<br />

designed a prospective phase II clinical trial where paired biopsies are<br />

collected from chemotherapy resistant TNBCs (NCT01276899). Four<br />

needle core biopsies are collected before the initiation <strong>of</strong> treatment and 2<br />

weeks before surgery or at the time <strong>of</strong> progression in the neoadjuvant and<br />

metastatic settings respectively. Metastatic sites eligible for biopsy include<br />

liver, lung, skin and lymph nodes. This study is presently recruiting at 5<br />

major health centers in Quebec and will soon open in the USA. We have<br />

currently enrolled 13 patients in the neoadjuvant setting and 2 metastatic<br />

patients. Major challenges in patient enrolment will be discussed. We have<br />

standardized the methods <strong>of</strong> collection and processing <strong>of</strong> tissue and blood<br />

specimens to ensure their molecular integrity and compatibility with<br />

different genomic and proteomic molecular platforms. Analysis <strong>of</strong> tumor<br />

cellularity has been incorporated into our quality control and we have<br />

optimized the extraction <strong>of</strong> nucleic acids to obtain high yields and optimal<br />

quality. Paired biopsies will undergo Next Gen Sequencing, flow sorted<br />

aCGH analysis, gene expression and miRNA pr<strong>of</strong>iling as well as phosphoproteomic<br />

pr<strong>of</strong>iling using reverse phase protein arrays. Collection <strong>of</strong> clinical<br />

data will allow molecular pr<strong>of</strong>iling data to be linked to clinical response<br />

data so as to determine DNA, RNA and protein factors correlated with<br />

tumor resistance to chemotherapy.<br />

TPS1141 General Poster Session (Board #36B), Sat, 8:00 AM-12:00 PM<br />

Randomized controlled trial comparing zoledronic acid plus chemotherapy<br />

with chemotherapy alone as a neoadjuvant treatment in patients with<br />

HER2-negative primary breast cancer. Presenting Author: Norio Kohno,<br />

Department <strong>of</strong> Breast Oncology, Tokyo Medical University Hospital, Tokyo,<br />

Japan<br />

Background: Zoledronic acid (ZOL) is a nitrogen-containing bisphosphonate,<br />

and it induces osteoclast apoptosis and inhibits bone resorption by<br />

inhibiting the mevalonate pathway. ZOL has also been found to have<br />

antitumor effects that include an angiogenesis inhibiting action, an<br />

inhibitory effect on tumor cell adhesion to and invasion <strong>of</strong> the extracellular<br />

matrix, and activation <strong>of</strong> a gdT cells. In addition, it has been found to have a<br />

synergistic apoptosis-inducing effect when used in combination with<br />

antitumor drugs. In this study we will investigate the pCR rate when ZOL is<br />

added to anthracycline followed by taxane to treat T2 and T3 breast cancer<br />

patients. Methods: Women with resectable invasive StageIIA-IIIB (T � a3<br />

cm or T � a2 cm and lymph node positive) breast cancer who are<br />

HER-2-negative, between 20 and 70 years <strong>of</strong> age, and ECOG PS 0-1 are<br />

eligible. Patients with distant metastasis, patients who have had received<br />

chemotherapy, hormone therapy, or radiotherapy for breast cancer, patients<br />

with serious complications, such as heart disease or an infection,<br />

patients with a complicating dental or jaw infection or traumatic condition<br />

<strong>of</strong> the teeth, and patients with a history <strong>of</strong> treatment with a bisphosphonate<br />

within the previous 12 months are excluded. A total <strong>of</strong> 4 courses <strong>of</strong> FEC100<br />

are administered every 3 weeks followed by weekly paclitaxel for 12<br />

courses. ZOL 4mg is administered every 3-4 weeks a total <strong>of</strong> 7 times.<br />

Patients are randomized 1:1 to chemotherapy � ZOL group or chemotherapy<br />

alone group, according to the presence or absence <strong>of</strong> lymph node<br />

metastasis, estrogen receptor (ER) status, and their menopausal status.<br />

The primary endpoint is pCR. Secondary endpoints are tumor response<br />

rate, the breast-conserving surgery ratio, and disease-free survival (DFS).<br />

We calculated the sample size on the basis <strong>of</strong> a pCR rate <strong>of</strong> 18% in the<br />

chemotherapy alone group and 35% in the chemotherapy � ZOL group, at<br />

a one-sided significance rate <strong>of</strong> 5%, and test power <strong>of</strong> 80%, and as a result<br />

we will target a patient sample size <strong>of</strong> 180 patients. 162 <strong>of</strong> planned 180<br />

patients have been enrolled as <strong>of</strong> January 24, 2012.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


84s Breast Cancer—Triple-Negative/Cytotoxics/Local Therapy<br />

TPS1142 General Poster Session (Board #36C), Sat, 8:00 AM-12:00 PM<br />

NSABP B-47: A phase III trial <strong>of</strong> adjuvant therapy comparing chemotherapy<br />

alone (six cycles <strong>of</strong> docetaxel plus cyclophosphamide or four cycles<br />

<strong>of</strong> doxorubicin plus cyclophosphamide followed by weekly paclitaxel) to<br />

chemotherapy plus trastuzumab in women with node-positive or high-risk,<br />

node-negative, HER2-low invasive breast cancer. Presenting Author: Louis<br />

Fehrenbacher, National Surgical Adjuvant Breast and Bowel Project and<br />

Kaiser Permanente Northern California, Vallejo, CA<br />

Background: Adjuvant studies utilizing trastuzumab in early HER2� breast<br />

cancer demonstrated a large reduction in recurrence and death. Postenrollment<br />

central testing showed HER2 non-amplified participants derived<br />

similar benefit. Methods: Selection <strong>of</strong> one <strong>of</strong> the two chemotherapy<br />

regimens is by physician choice: The non-anthracycline regimen is TC<br />

(docetaxel 75 mg/m2, cyclophosphamide 600 mg/m2) administered IV<br />

every 3 weeks for 6 cycles; the anthracycline regimen is AC followed by WP<br />

(doxorubicin 60 mg/m2 and cyclophosphamide 600 mg/m2 administered<br />

IV either every 3 weeks or every 2 weeks [per investigator discretion] for 4<br />

cycles followed by paclitaxel 80 mg/m2 IV weekly for 12 doses). Patients<br />

are randomly assigned to receive chemotherapy with or without trastuzumab<br />

therapy. For patients receiving the TC chemotherapy regimen,<br />

trastuzumab is given every 3 weeks during and following chemotherapy<br />

until 1 year after the first trastuzumab dose (8 mg/kg loading dose; 6 mg/kg<br />

for the remaining doses). For patients receiving the AC followed by WP<br />

chemotherapy regimen, trastuzumab begins with the first dose <strong>of</strong> weekly<br />

paclitaxel and will be given weekly for 12 doses (4 mg/kg loading dose; 2<br />

mg/kg for the remaining weekly doses). Following completion <strong>of</strong> WP,<br />

trastuzumab therapy continues with 6 mg/kg doses given every 3 weeks for<br />

a total <strong>of</strong> 1 year. Eligibility: Eligibility includes: node positive or high risk<br />

node negative female breast cancer patients; HER2 IHC 1� or 2� scores,<br />

but non amplified by FISH Statistical Design: The primary aim is to<br />

determine whether the addition <strong>of</strong> trastuzumab to chemotherapy improves<br />

invasive disease-free survival (IDFS). 3260 patients will be enrolled to<br />

provide statistical power <strong>of</strong> 0.9 to detect a 33% reduction in the hazard rate<br />

<strong>of</strong> IDFS using a one-sided alpha level <strong>of</strong> 0.025. Progress: Protocol was<br />

activated in January 2011. As <strong>of</strong> January 27, 2012, 486 <strong>of</strong> 3260 patients<br />

have been enrolled. Supported by NCI U10-12027, -37377, 69651,<br />

69974, and Genentech, Inc.<br />

TPS1144 General Poster Session (Board #36E), Sat, 8:00 AM-12:00 PM<br />

ARTemis: Randomized trial with neoadjuvant chemotherapy for patients<br />

with early breast cancer. Presenting Author: Helena Margaret Earl, Department<br />

<strong>of</strong> Oncology, University <strong>of</strong> Cambridge, and NIHR Cambridge Biomedical<br />

Research Centre, Cambridge, United Kingdom<br />

Background: Bevacizumab is a new humanised monoclonal antibody which<br />

targets vascular endothelial growth factor (VEGF) and thereby the neoangiogenic<br />

process in cancer. Bevacizumab has shown promising anti-tumour<br />

effect when given concurrently with taxane-based chemotherapy in breast<br />

cancer. However two neoadjuvant breast cancer trials have produced<br />

conflicting results. The NSABP-B40 study showed significant benefit for<br />

bevacizumab only in the ER�ve patients (pathological complete response<br />

(pCR) in ER�ve population 15.2% vs 23.3%, p�0.008). Whereas the<br />

GEPARQUINTO trial reported significant benefit only in the triple negative<br />

patients (pCR 27.8% vs 36.4% p�0.021). Methods: ARTemis is a phase<br />

III randomised trial to determine whether the addition to neo-adjuvant<br />

chemotherapy <strong>of</strong> bevacizumab is more effective than standard chemotherapy<br />

alone in patients with HER2-negative early breast cancer. A total <strong>of</strong><br />

400 patients will be randomised into each <strong>of</strong> the two treatment arms which<br />

will allow an absolute difference in the pCR rates in excess <strong>of</strong> 10% to be<br />

detected at the 5% (2-sided) level <strong>of</strong> significance with an 85% power.<br />

Primary outcome is pathological complete response rates after neoadjuvant<br />

chemotherapy, i.e. no residual invasive carcinoma in the breast,<br />

and no evidence <strong>of</strong> metastatic disease within the lymph nodes. Secondary<br />

outcomes are disease free survival, overall survival, complete pathological<br />

response rates in breast alone, radiological response after 3 and 6 cycles <strong>of</strong><br />

chemotherapy and rate <strong>of</strong> breast conservation and toxicities. Results:<br />

Recruitment into ARTemis began in April 2009 and as <strong>of</strong> January 2012<br />

had recruited 469 (59%). ARTemis is due to complete recruitment by Dec<br />

2012 and the first planned interim analysis <strong>of</strong> the primary outcome will be<br />

Dec 2013. Conclusion: The IDSMC reviewed the trial in June 2011 and<br />

recommended continuation <strong>of</strong> recruitment to this important trial given<br />

there were no safety concerns, and results from the other neoadjuvant<br />

studies (NSABP-B40 and GEPARQUINTO) are conflicting.<br />

TPS1143 General Poster Session (Board #36D), Sat, 8:00 AM-12:00 PM<br />

IBL2001: Phase (ph) I/II study <strong>of</strong> a novel dose-dense (dd) schedule <strong>of</strong><br />

indibulin for the treatment <strong>of</strong> metastastic breast cancer. Presenting Author:<br />

Tiffany A. Traina, Memorial Sloan-Kettering Cancer Center, New York, NY<br />

Background: Indibulin (Zybulin, ZIO-301) is a new, synthetic agent that<br />

inhibits tumor cell growth at the G2/M phase through destabilization <strong>of</strong><br />

microtubule dynamics. It binds tubulin at a different site than taxanes and<br />

vinca alkaloids. Indibulin does not interact with acetylated (neuronal)<br />

tubulins and has not exhibited the neurotoxicity associated with other<br />

tubulin binders. Indibulin has potent antitumor activity in human cancer<br />

cell lines, including multidrug-, taxane-, and vinblastine-resistant lines.<br />

Norton-Simon modeling based on cell line data suggested that dd administration<br />

could optimize efficacy while limiting toxicity. Methods: Eligible are<br />

patients (pts) with metastatic or unresectable locally advanced breast<br />

cancer, measurable or non-measurable disease, and any number <strong>of</strong> prior<br />

therapies. The objective <strong>of</strong> the Ph I portion is to determine the maximum<br />

tolerated dose (MTD) <strong>of</strong> indibulin when given in a dd fashion (5 days<br />

treatment, 9 days rest (14 total) using standard 3�3 dose escalation<br />

schema. Ph II will seek to estimate the proportion <strong>of</strong> pts progression free at<br />

4 months when treated with indibulin at the dose determined in the Ph I.<br />

Secondary endpoints are overall response rate, proportion <strong>of</strong> subjects with<br />

stable disease �6 months and toxicity pr<strong>of</strong>ile <strong>of</strong> indibulin. Optimal Simon<br />

two-stage design with an unpromising rate <strong>of</strong> pts who have not progressed<br />

at four months as 20% and a promising rate <strong>of</strong> 40% with a type I error <strong>of</strong><br />

5% and power <strong>of</strong> 80% will be used. 13 pts will initially enter the 1st stage <strong>of</strong><br />

the ph II portion. If there are 3 or fewer pts who do not experience disease<br />

progression at 4 months in the first stage <strong>of</strong> ph II, the study will be<br />

terminated and declared to have a negative result. If 4 or more pts do not<br />

progress at 4 months, enrollment in the ph II <strong>of</strong> the study will be extended<br />

to 43 pts. A total <strong>of</strong> 45 pts will be enrolled to allow for a 5% drop out rate to<br />

account for pts who may not be evaluable for the primary endpoint due to<br />

toxicity or study withdrawal rather than progression <strong>of</strong> disease or death prior<br />

to 4 months. If �13 out <strong>of</strong> 43 pts are progression-free at 4 months, the<br />

study will be considered to have a positive result and indibulin would be<br />

considered worthy <strong>of</strong> further testing in this disease.<br />

TPS1145 General Poster Session (Board #37A), Sat, 8:00 AM-12:00 PM<br />

A phase II, single-arm, feasibility study <strong>of</strong> dose-dense doxorubicin and<br />

cyclophosphamide (AC) followed by eribulin mesylate for the adjuvant<br />

treatment <strong>of</strong> early-stage breast cancer (EBC). Presenting Author: Tiffany A.<br />

Traina, Memorial Sloan-Kettering Cancer Center, New York, NY<br />

Background: Randomized trials have confirmed the benefit <strong>of</strong> the combination<br />

<strong>of</strong> an anthracycline (A) and cyclophosphamide (C) for the adjuvant<br />

treatment <strong>of</strong> EBC. The addition <strong>of</strong> taxane therapy to AC therapy has further<br />

improved survival. Despite the improvement in adjuvant therapies for BC,<br />

new approaches for improving outcomes are <strong>of</strong> significant importance and<br />

may involve developing improved combination regimens. Eribulin mesylate<br />

has demonstrated antitumor activity and significant improvement in overall<br />

survival (OS) in patients with heavily pretreated locally advanced or<br />

metastatic breast cancer (MBC) and may improve outcomes in EBC as well.<br />

Methods: This study will determine the feasibility <strong>of</strong> eribulin as adjuvant<br />

therapy following dose-dense AC for HER2 normal EBC. A completion rate<br />

<strong>of</strong> �80% was set as a threshold for feasibility as established adjuvant<br />

regimens have shown feasibility rates ranging from ~65% for trastuzumab<br />

to �80%. This is a phase 2, single-center, feasibility study <strong>of</strong> dose-dense<br />

adjuvant chemotherapy in patients with EBC. Dose-dense AC (Doxorubicin<br />

60 mg/m2 IV plus C 600 mg/m2 IV) on day 1 <strong>of</strong> every 14-day cycle is given<br />

for 4 cycles, followed by 4 cycles <strong>of</strong> eribulin mesylate at 1.4 mg/m2 over<br />

2-5 minutes IV on days 1 and 8 every 21 days. Growth factors are given on<br />

day 2 <strong>of</strong> AC cycles and only for neutropenia events with eribulin treatment.<br />

Feasibility is determined by the ability to complete the eribulin portion <strong>of</strong><br />

the regimen without a dose delay or reduction. Exploratory objectives<br />

include efficacy endpoints <strong>of</strong> 3-year disease-free survival (DFS) and OS.<br />

Patients have histologically confirmed HER2 normal stage I-III invasive<br />

disease and adequate bone marrow, liver, and renal function. Thirty two <strong>of</strong><br />

approximately 80 planned patients are currently enrolled. Feasibility rates<br />

will be calculated with growth factor support (ie, the successful use <strong>of</strong><br />

growth factor support following a neutropenia event is not considered a<br />

dose delay)and without growth factor support (ie, where need for growth<br />

factors is considered a delay). Secondary endpoints include DFS and OS<br />

and will be estimated by Kaplan-Meier method.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


TPS1146 General Poster Session (Board #37B), Sat, 8:00 AM-12:00 PM<br />

DETECT III: A multicenter, randomized, phase III study to compare<br />

standard therapy alone versus standard therapy plus lapatinib in patients<br />

(pts) with initially HER2-negative metastatic breast cancer but with<br />

HER2-positive circulating tumor cells (CTC). Presenting Author: Carsten<br />

Hagenbeck, Department <strong>of</strong> Gynecology and Obstetrics, Heinrich Heine<br />

University, Duesseldorf, Germany<br />

Background: HER2 status may change over the course <strong>of</strong> disease in breast<br />

cancer pts. Approx. 20-30% <strong>of</strong> pts with initially HER2-negative breast<br />

cancer have HER2-positive metastasis (Zidan et al. 2005; Tewes et al.<br />

2009). Determining HER2 status on CTC is one option to re-evaluate HER2<br />

status at the time metastasis is diagnosed. Currently it is unclear if<br />

HER2-targeted therapy based on the assessment <strong>of</strong> HER2 status <strong>of</strong> CTC<br />

reveals a clinical benefit. Methods: This is a randomized, open-label, two<br />

arm phase III study to investigate the clinical efficacy <strong>of</strong> lapatinib, as a<br />

HER2-targeted therapy in initially HER2-negative metastatic breast cancer<br />

pts with HER2-positive CTC at the time <strong>of</strong> distant disease. As only half <strong>of</strong><br />

the pts with HER2-negative metastatic breast cancer show CTC-positivity<br />

and <strong>of</strong> those approx. 32% will exhibit HER2-positive CTC (Fehm et al.<br />

2010), screening <strong>of</strong> about 1420 pts is required to enroll 228 pts. Main<br />

inclusion criteria: metastatic breast cancer with HER2-negative primary<br />

tumor tissue and/or biopsies from metastatic sites or locoregional recurrences,<br />

evidence <strong>of</strong> �1 HER2-positive CTC and �1 measurable metastatic<br />

lesion according to RECIST. Eligible pts will be randomized 1:1 to receive<br />

standard treatment vs. standard treatment plus lapatinib. Standard chemoor<br />

endocrine therapy must be approved in combination with lapatinib or<br />

been investigated in prior clinical trials. Primary endpoint is progression<br />

free survival. Secondary endpoints include overall response rate, clinical<br />

benefit rate, overall survival and dynamic <strong>of</strong> CTC. The DETECT III trial is<br />

one <strong>of</strong> the first trials where treatment is based on phenotypic characteristics<br />

<strong>of</strong> CTC. If this trial succeeds in proving efficacy <strong>of</strong> lapatinib in pts with<br />

initially HER2-negative metastatic breast cancer but HER2-positive CTC,<br />

this will establish a new strategy in the treatment <strong>of</strong> metastatic breast<br />

cancer.<br />

TPS1148 General Poster Session (Board #37D), Sat, 8:00 AM-12:00 PM<br />

Phase II study <strong>of</strong> the multikinase inhibitor dovitinib (TKI258) or placebo in<br />

combination with fulvestrant in postmenopausal, endocrine resistant<br />

HER2-/HR� breast cancer. Presenting Author: Fabrice Andre, Institut<br />

Gustave Roussy, Villejuif, France<br />

Background: Overcoming endocrine resistance is a critical goal in the<br />

treatment <strong>of</strong> hormone receptor�positive (HR�) breast cancer. Molecular<br />

mechanisms associated with endocrine resistance include adaptive “crosstalk”<br />

between the estrogen receptor and the fibroblast growth factor<br />

receptor (FGFR). Up to 8% <strong>of</strong> HR�/HER2- breast cancer patients (pts)<br />

have amplification <strong>of</strong> the FGFR1 gene, which is associated with resistance<br />

to endocrine therapy but can be overcome via FGFR1 inhibition in<br />

preclinical models. Dovitinib is a potent FGF, VEGF, and PDGF receptor<br />

tyrosine kinase inhibitor that demonstrated antitumor activity in heavily<br />

pretreated breast cancer pts with FGF pathway amplification (FGFR1,<br />

FGFR2, or ligand FGF3; Andre et al, ASCO 2011). Dovitinib may reverse<br />

resistance to endocrine therapy related to FGF-pathway amplification and<br />

is studied here to determine if it can improve outcomes when combined<br />

with fulvestrant. Methods: Postmenopausal HER2-/HR� locally advanced<br />

or metastatic breast cancer pts (N»150) progressing within 12 months <strong>of</strong><br />

completion <strong>of</strong> adjuvant endocrine therapy or after � 1 prior endocrine<br />

therapy in the advanced setting will be enrolled in this multicenter,<br />

randomized, double blind, placebo controlled, phase II trial. Pts will<br />

prospectively undergo molecular screening to enrich for FGF-amplification<br />

(FGFR1, FGFR2, orFGF3 amplification by qPCR; 45 amplified and 30<br />

non-amplified pts per arm). Pts will be randomized 1:1 to receive<br />

fulvestrant (500 mg q4w [with an additional dose 2 wks after the initial<br />

dose]) in combination with oral dovitinib (500 mg, 5 days on/2 days <strong>of</strong>f) or<br />

placebo until disease progression, unacceptable toxicity, or death. The<br />

primary endpoint is progression-free survival, with tumor assessments<br />

performed q8w. Secondary endpoints include overall response rate per<br />

RECIST v1.1, duration <strong>of</strong> response, overall survival, ECOG performance<br />

status and patient reported outcome scores over time, and safety. The<br />

pharmacodynamic effect <strong>of</strong> dovitinib on FGFR-associated angiogenic<br />

pathways in tumor specimens and potential predictive biomarkers <strong>of</strong><br />

response to dovitinib will be explored.<br />

Breast Cancer—Triple-Negative/Cytotoxics/Local Therapy<br />

85s<br />

TPS1147 General Poster Session (Board #37C), Sat, 8:00 AM-12:00 PM<br />

A multicenter prospective study <strong>of</strong> image-guided radi<strong>of</strong>requency ablation<br />

for small breast carcinomas. Presenting Author: Takayuki Kinoshita,<br />

National Cancer Center Hospital, Tokyo, Japan<br />

Background: As the management <strong>of</strong> breast carcinoma evolves toward less<br />

invasive treatments, the next step is the possibility <strong>of</strong> removing the primary<br />

tumor without surgery. The most promising noninvasive ablation technique<br />

is radi<strong>of</strong>requency ablation (RFA), which can effectively kill tumor cells with<br />

a low complication rate. Our preliminary studies <strong>of</strong> RFA followed by<br />

standard surgical resection have indicated that this technique is effective<br />

for surgical ablation <strong>of</strong> small (� 2cm) breast tumors without extensive<br />

intraductal components (EIC). Methods: To determine if RFA is oncologically<br />

and cosmetically appropriate for the local treatment <strong>of</strong> primary breast<br />

carcinoma, this multi-center prospective study used RFA as the sole local<br />

treatment <strong>of</strong> breast tumors � 1.5cm in size on ultrasound and MRI.<br />

Exclusion criteria include receiving <strong>of</strong> preoperative chemotherapy, or the<br />

presence <strong>of</strong> invasive lobular carcinoma or invasive ductal carcinoma with<br />

suspicious EIC. After confirmation that the standard baseline core biopsy<br />

for diagnosis and measurement <strong>of</strong> tumors markers (ER, PgR, HER-2/neu<br />

expression and the presence <strong>of</strong> the Ki-67 proliferative marker) have been<br />

obtained, consent will be obtained and the patient scheduled RFA. All<br />

patients received adjuvant radiation therapy. The use and choice <strong>of</strong><br />

systemic therapy will be based on the information from the baseline core<br />

biopsy and imaging studies. The first primary endpoints <strong>of</strong> this study is<br />

successful tumor ablation, as evidenced by negative findings on vacuumassisted<br />

or core biopsies and imaging studies after RFA. The second<br />

primary endpoints is the incidence <strong>of</strong> procedure related adverse events.<br />

Forty patients with small tumors that are clearly identifiable and measurable<br />

by ultrasound and MRI were enrolled. The response to ablation was<br />

evaluated with both vacuum-assisted or core biopsies and imaging studies<br />

every 3 months during the first year. The long-term outcomes were assessed<br />

using quality <strong>of</strong> life measurement scales and imaging studies every 6<br />

months thereafter through year 5.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


86s Cancer Prevention/Epidemiology<br />

1500 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

A phase III prevention trial <strong>of</strong> low-dose tamoxifen in HRT users: The HOT<br />

trial. Presenting Author: Bernardo Bonanni, European Institute <strong>of</strong> Oncology,<br />

Milan, Italy<br />

Background: Primary prevention trials have shown that tamoxifen (T) lowers<br />

ER� breast cancer (BC) incidence, but adverse events are a barrier to its<br />

broad use. The Italian Tamoxifen Prevention Trial showed a positive<br />

risk/benefit ratio in the subgroup <strong>of</strong> hormone replacement therapy (HRT)<br />

users in the T arm, with fewer BC and no cardiovascular disease (CVD)<br />

excess. In a dose-ranging study, T 5 mg/day showed a favorable biomarker<br />

modulation in HRT users. We conducted a multicentre, phase III, BC<br />

prevention trial in current or de novo HRT users, randomized to either T 5<br />

mg/day or placebo (P) for 5 yrs, with 5yrs follow-up. Methods: The study was<br />

powered to detect a 40% decrease <strong>of</strong> BC on T from an annual rate <strong>of</strong><br />

4/1000 on P with 20% dropout and 5y recruitment. All analyses were<br />

performed on ITT basis. The cumulative incidence <strong>of</strong> events was determined<br />

using Kaplan-Meier methods. The two treatment groups were<br />

compared by using the log-rank test. Hazard ratios (HRs) and 95%<br />

confidence intervals were obtained using Cox proportional regression<br />

model. All P values were two-sided. Results: Due to the WHI-HRT trial<br />

results, recruitment was lower than anticipated, with a total <strong>of</strong> 1884<br />

subjects being randomized (946 P and 938 T arm, respectively). 79% were<br />

�50 years old at study entry. 66% were normal weight, 21% were<br />

hysterectomized; 80% were already on HRT at baseline, 53% were on<br />

trasdermal route and 28% had 5y-Gail risk�1.5%. At 7.7y mean follow-up,<br />

43 BC were diagnosed, 24 (annual rate 4.1/1000) on P and 19 (3.3/1000)<br />

on T (HR 0.80, 95%CI 0.44-1.46). The efficacy <strong>of</strong> T was greater in<br />

luminal-A type (HR�0.32, 95% CI, 012-086). CVD events were rare<br />

(including 1 DVT on T), with no statistical difference between arms. A HR <strong>of</strong><br />

4.74 (95% CI, 1.96-11.5) was observed for benign endometrial polyps,<br />

but no increase <strong>of</strong> uterine cancers (3 on P vs 1 on T). Menopausal<br />

symptoms were more frequent on T, whereas headache was less frequent on<br />

T. Conclusions: The combination <strong>of</strong> low-dose T and HRT is safe and provides<br />

a promising way to retain the benefits while reducing the risks <strong>of</strong> either<br />

agent. While the insufficient power <strong>of</strong> our study prevents firmer conclusions,<br />

it supports further studies <strong>of</strong> low-dose T in the prevention setting.<br />

1502 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

BMI, long-term changes in BMI, and risk <strong>of</strong> cancer mortality in a large<br />

cohort study. Presenting Author: Nilo<strong>of</strong>ar Taghizadeh, Department <strong>of</strong><br />

Epidemiology, University Medical Center Groningen, Groningen, Netherlands<br />

Background: There are indications <strong>of</strong> an association between Body Mass<br />

Index (BMI) and risk <strong>of</strong> different cancer types. There is dispute whether this<br />

association differs between males and females. Methods: We studied the<br />

association <strong>of</strong> BMI at the first survey with risk <strong>of</strong> mortality from the most<br />

common types <strong>of</strong> cancer (lung, colorectal, breast and prostate cancer) in a<br />

large general population-based cohort study (Vlagtwedde-Vlaardingen,<br />

1965-1990) with follow-up on mortality status until 2009. Additionally,<br />

we assessed this association based on tertiles <strong>of</strong> the annual change in BMI<br />

(defined as the difference between BMI at last survey and first survey<br />

divided by the time between last and first survey). We used 3 categories <strong>of</strong><br />

BMI (� 25 kg/m2 , 25-30 kg/m2 , and � 30 kg/m2 ) and changes in BMI<br />

(� 0.02 kg/m2 /yr, 0.02-0.2 kg/m2 /yr, and � 0.2 kg/m2 /yr) in the analyses.<br />

The multivariate Cox regression model was adjusted for age, smoking,<br />

gender. Analyses were additionally stratified by gender and smoking.<br />

Results: Among all 8645 subjects, 1194 died due to cancer (lung cancer:<br />

275; colorectal cancer: 134; breast cancer: 117; prostate cancer: 83).<br />

Mortality from all types <strong>of</strong> cancer was significantly increased in subjects<br />

with BMI � 30 kg/m2 (HR (95 % CI)) � 1.22 (1.00-1.48)), especially in<br />

females (1.38 (1.06-1.81)) and in never smokers (1.39 (1.02-1.90)).<br />

Prostate cancer mortality was significantly increased in males with BMI<br />

25-30 kg/m2 (2.04 (1.90-3.83)) and � 30 kg/m2 (2.61 (1.02-6.67)). This<br />

association between prostate cancer mortality and BMI was higher in<br />

smokers. Lung cancer mortality risk was decreased in subjects with BMI<br />

25-30 kg/m2 (0.71 (0.54-0.93)) and � 30 kg/m2 (0.82 (0.50-1.32)),<br />

especially in males, in smokers, and in smoking males. There were no<br />

significant associations between BMI and colorectal or breast cancer<br />

mortality nor between change in BMI and mortality from all analyzed types<br />

<strong>of</strong> cancer. Conclusions: We show that an increase in BMI is associated with<br />

an increased risk <strong>of</strong> mortality from all types <strong>of</strong> cancer in females and with<br />

an increased mortality risk from prostate cancer in males but with a<br />

decreased lung cancer mortality risk, especially in males. More research is<br />

needed into the biological mechanisms that link BMI to cancer.<br />

1501 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Estrogen plus progestin (E�P) and breast cancer incidence and mortality.<br />

Presenting Author: Rowan T. Chlebowski, Los Angeles Biomedical Research<br />

Institute at Harbor-UCLA Medical Center, Torrance, CA<br />

Background: In the WHI clinical trial, E�P increased both breast cancer<br />

incidence and breast cancer mortality (JAMA 2010;304:1684). In contrast,<br />

breast cancers associated with E�P use in most observational studies<br />

have a more favorable prognosis. To address differences, a cohort <strong>of</strong> WHI<br />

Observational Study participants with characteristics similar to the WHI<br />

clinical trial was identified to examine E�P association with invasive breast<br />

cancer incidence and outcome. Methods: 41,449 postmenopausal women<br />

with no prior hysterectomy and mammogram negative for breast cancer � 2<br />

years before who either were not hormone users (25,328) or were using<br />

E�P (16,121) were identified. Breast cancers were verified by centralized<br />

medical record review. Adjusted Cox proportional hazard regression was<br />

used to calculate hazard ratios (HRs) with 95% confidence intervals (CI).<br />

Additional analyses adjusted for breast cancer screening, censoring participants<br />

for incidence analyses who had a � 2 year interval without a<br />

mammogram. Results: After a mean (SD) follow-up 11.3 (3.1) years, 2,236<br />

breast cancers were diagnosed. Breast cancer incidence was higher in E�P<br />

users (0.60% vs 0.42%, annualized rate, respectively: HR 1.55, 95% CI<br />

1.41-1.70, P�0.001). Screening adjusted analyses had stronger breast<br />

cancer association (0.63% vs 0.39%, HR 1.72, 95% CI 1.54-1.93;<br />

P�0.001). Survival following breast cancer, measured from diagnosis<br />

date, was similar in E�P users and non-users (HR 0.95, 95% CI<br />

0.74-1.23). Breast cancer mortality, analyzed from cohort entry date, are<br />

shown in the table. Conclusions: E�P use is associated with increased<br />

breast cancer incidence. As breast cancer prognosis following diagnosis on<br />

E�P is similar to that <strong>of</strong> nonusers, the higher incidence with E�P leads to<br />

increased breast cancer mortality.<br />

Deaths from breast cancer HR (95% CI) P values<br />

Screening adjusted*<br />

No 1.32 (0.90-1.93) 0.15<br />

Yes 1.54 (1.00-2.39) 0.052<br />

Deaths after breast cancer diagnosis<br />

Screening adjusted *<br />

No 1.65 (1.29-2.12) �0.001<br />

Yes 2.21 (1.66-2.94) �0.001<br />

* Censored for breast cancer diagnosis when time w/o mammogram � 2 yrs.<br />

1503 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Long-term therapy with thiazolidinediones and the risk <strong>of</strong> bladder cancer: A<br />

cohort study. Presenting Author: Ronac Mamtani, Perelman School <strong>of</strong><br />

Medicine at the University <strong>of</strong> Pennsylvania, Philadelphia, PA<br />

Background: The US Food and Drug Administration and other regulatory<br />

agencies recently warned prescribers that use <strong>of</strong> pioglitazone, a thiazolidinedione<br />

(TZD), may increase the risk <strong>of</strong> bladder cancer. France and Germany<br />

removed the drug from their markets, although the rest <strong>of</strong> Europe did not.<br />

However, no information was available about the risk from alternative TZDs.<br />

We aimed to compare bladder cancer risk over time with use <strong>of</strong> TZDs<br />

relative to sulfonylureas (SUs), and between pioglitazone and rosiglitazone.<br />

Methods: We conducted a cohort study <strong>of</strong> patients with type 2 diabetes who<br />

initiated treatment with a TZD or SU using The Health Improvement<br />

Network (2000-2010), a UK general practitioner medical record database.<br />

Incident cancers within the database were identified. We computed hazard<br />

ratios (HRs) <strong>of</strong> bladder cancer for TZDs in comparison to the reference<br />

group <strong>of</strong> SU users. Results: There were 60 incident diagnoses <strong>of</strong> bladder<br />

cancer in the TZD cohort (n�18,459) and 137 in the SU cohort<br />

(n�41,396). There was no significant difference in bladder cancer risk<br />

between the cohorts (HR 0·93, [95% CI 0·68-1·29]), but most use was<br />

short-term use. In contrast, the risk <strong>of</strong> bladder cancer increased with<br />

increasing time since initiation <strong>of</strong> TZD versus SU therapy (HRs 1·15, 1·40,<br />

and 1·72 for 3-4, 4-5, and � 5 years, respectively; P-trend�0·033).<br />

Bladder cancer risk also increased with increasing time since initiation <strong>of</strong><br />

pioglitazone (P-trend�0·001) and rosiglitazone (P-trend�0·006). Direct<br />

comparison <strong>of</strong> pioglitazone to rosiglitazone did not demonstrate significant<br />

differences in cancer risk with increasing time since initiation (Ptrend�0·12)<br />

or duration <strong>of</strong> therapy (P-trend�0·75). Conclusions: Longterm<br />

TZD therapy is associated with an increased risk <strong>of</strong> bladder cancer,<br />

which appears to be a class effect.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


1504 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Dexrazoxane exposure and risk <strong>of</strong> secondary acute myeloid leukemia in<br />

pediatric cancer patients. Presenting Author: Dana Marie Walker, Children’s<br />

Hospital <strong>of</strong> Philadelphia, Philadelphia, PA<br />

Background: Dexrazoxane (DXZ) is an effective cardioprotectant in children<br />

with leukemia and solid tumors. However, the potential risk <strong>of</strong> secondary<br />

acute myeloid leukemia (AML) limits DXZ use. We compared the incidence<br />

<strong>of</strong> secondary AML in pediatric cancer patients with and without DXZ<br />

exposure to estimate the risk <strong>of</strong> DXZ-associated secondary AML. Methods:<br />

We conducted a retrospective cohort study <strong>of</strong> anthracycline exposed<br />

pediatric cancer patients (excluding de novo AML) hospitalized from<br />

January 1999 to March 2011 in 43 freestanding children’s hospitals in the<br />

Pediatric Health Information System (PHIS) database. Secondary AML was<br />

defined as an ICD9 code for AML that occurred at least 90 days after first<br />

anthracycline exposure. Proportions <strong>of</strong> patients with secondary AML were<br />

compared between patients with and without an exposure to DXZ after the<br />

initial anthracycline exposure. Results: During the study period <strong>of</strong> interest,<br />

15,532 pediatric cancer patients were exposed to anthracycline, <strong>of</strong> which<br />

1404 (9.04%) subsequently received DXZ. Secondary AML was identified<br />

in 235 (1.51%) patients. Patients � 10 years <strong>of</strong> age, black patients,<br />

patients in New England and the Midatlantic regions, and those with bone<br />

tumors were significantly more likely to have received DXZ. The unadjusted<br />

incidence <strong>of</strong> secondary AML in the DXZ exposed and unexposed patients<br />

did not differ significantly (1.21% v. 1.54%, p�0.3308). After adjusting<br />

for these variations in demographics, the incidence <strong>of</strong> secondary AML still<br />

did not significantly differ between DXZ exposed and unexposed patients<br />

(p�0.6224). Conclusions: Our findings suggest that DXZ exposure does not<br />

lead to an increased risk <strong>of</strong> secondary AML in children. These data are<br />

important given the conflicting results from individual clinical trials about<br />

the risk <strong>of</strong> DXZ-associated secondary AML. While subject to well-known<br />

limitations <strong>of</strong> administrative database analyses, these data represent the<br />

largest cohort <strong>of</strong> DXZ exposed patients with available data on the occurrence<br />

<strong>of</strong> secondary AML. Additional multivariate analyses <strong>of</strong> chemotherapeutic<br />

covariates and time to secondary AML are ongoing.<br />

1506 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Sensitivity <strong>of</strong> clinical BRCA1 testing compared with linkage analysis.<br />

Presenting Author: Payal D. Shah, University <strong>of</strong> Pennsylvania, Philadelphia,<br />

PA<br />

Background: Specific clinical interventions in BRCA mutation carriers<br />

reduces the risk <strong>of</strong> breast and ovarian cancers and may improve survival;<br />

thus,identification <strong>of</strong> mutation carriers is important. The sensitivity <strong>of</strong><br />

current BRCA mutation testing is unclear as a “gold standard” test is<br />

lacking. We assessed the BRCA1 mutation detection rate with current<br />

comprehensive clinical testing using linkage analysis as the comparator.<br />

Methods: 26 families with linkage analyses results available were included<br />

in this analysis. BRCAPRO, BOADICEA, and other risk estimation models<br />

were applied. Maximum-likelihood linkage analyses were performed to<br />

compute two-point and multipoint LOD scores using previously described<br />

BRCA1 –linked genetic markers; scores were classified as linked, not<br />

linked, or suggestive. At least one individual from each family underwent<br />

comprehensive testing. Results: Of 26 families analyzed, 9 demonstrated<br />

linkage and 4 demonstrated suggestive linkage to BRCA1. Of these 13<br />

families, 12 were found to have BRCA1 mutations: a detection rate <strong>of</strong><br />

92.3%. In 3 <strong>of</strong> these 12 families, genetic mutation testing performed prior<br />

to large genomic rearrangement (LGR) testing was negative, demonstrating<br />

an improved detection rate with LGR testing which in this series was 23%.<br />

The 12 families with higher LOD scores and positive mutation testing had<br />

high mean prior probabilities by all models. Families which were not linked<br />

and who tested negative for mutations had lower mean prior probabilities.<br />

In one family, disease demonstrated linkage with a two-point LOD score <strong>of</strong><br />

1.59 and multipoint LOD <strong>of</strong> 0.85 and all risk estimation models yielded<br />

high prior probabilities. Despite this, mutation testing was negative by full<br />

sequencing and MLPA analysis. Conclusions: When evaluated in a sample<br />

<strong>of</strong> families from high-risk clinics, current comprehensive testing compares<br />

well to linkage data. The inclusion <strong>of</strong> LGR testing has improved the<br />

mutation detection capability <strong>of</strong> clinical testing; however, some mutations<br />

may still be missed. Negative mutation testing results should be interpreted<br />

in the context <strong>of</strong> family history and prior probability during<br />

counseling. Despite recent advances, further improvements in genetic<br />

testing are warranted.<br />

Cancer Prevention/Epidemiology<br />

87s<br />

CRA1505 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Quality <strong>of</strong> cancer family history and referral for genetic counseling and<br />

testing among oncology practices: A pilot test <strong>of</strong> quality measures as part <strong>of</strong><br />

the ASCO Quality Oncology Practice Initiative (QOPI). Presenting Author:<br />

Marie Wood, University <strong>of</strong> Vermont, Burlington, VT<br />

The full, final text <strong>of</strong> this abstract will be available at<br />

abstract.asco.org at 12:01 AM (EDT) on Monday, June 4,<br />

2012, and in the <strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong> online<br />

supplement to the June 20, 2012, issue <strong>of</strong> Journal <strong>of</strong><br />

<strong>Clinical</strong> Oncology. Onsite at the <strong>Meeting</strong>, this abstract will<br />

be printed in the Monday edition <strong>of</strong> ASCO Daily News.<br />

1507 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

The APC I1307K polymorphism as a significant risk factor for CRC in<br />

average-risk Ashkenazi Jews. Presenting Author: Ben Boursi, Sheba Medical<br />

Center, Ramat Gan, Israel<br />

Background: The I1307K adenomatous polyposis coli gene variant, prevalent<br />

among Ashkenazi Jews, may increase risk for colorectal neoplasia. We<br />

studied the clinical importance <strong>of</strong> screening for this polymorphism in 3305<br />

Israelis undergoing colonoscopic assessment. Methods: Blood samples and<br />

risk factor information were collected from individuals undergoing colonoscopic<br />

examination at our medical center. Germline genetic analysis for the<br />

APC I1307K variant was performed using real-time PCR for DNA extracted<br />

from peripheral mononuclear cells. Results: The overall prevalence <strong>of</strong> the<br />

I1307K polymorphism was 8.0% (10.1% among Ashkenazi while only<br />

2.7% among Sephardic Jews, p�0.001). The overall adjusted odds ratio<br />

(OR) for CR neoplasia among carriers was 1.51(95% CI, 1.16 –1.98).<br />

Among average risk Ashkenazi Jews, the OR was 1.76 (95% CI 1.26-2.45).<br />

On the contrary, among Sephardi subjects the OR was 0.996 (95% CI,<br />

0.51-1.93). A multiplicative interaction was identified between Ashkenazi<br />

ethnicity and APC I1307K carrier status (PINTERACTION�0.055). The<br />

histopathological features <strong>of</strong> adenomas and cancers did not differ between<br />

carriers and non-carriers. Conclusions: The APC I1307K gene variant is an<br />

important risk factor for CRC in average risk Ashkenazi jews and should be<br />

considered for screening in this population.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


88s Cancer Prevention/Epidemiology<br />

1508 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Use <strong>of</strong> PTEN protein dosage to predict for underlying germ-line PTEN<br />

mutations among patients presenting with thyroid cancer and Cowden-like<br />

phenotypes. Presenting Author: Joanne Y. Y. Ngeow, Genomic Medicine<br />

Institute, Cleveland Clinic, Cleveland, OH<br />

Background: Thyroid cancer is a major component <strong>of</strong> Cowden Syndrome<br />

(CS), a heritable syndrome characterized also by breast, uterine and kidney<br />

cancers. CS patients with an underlying germline PTEN mutation have a<br />

70-fold increased risk <strong>of</strong> developing epithelial thyroid cancer. In contrast,<br />

�1% <strong>of</strong> sporadic epithelial thyroid cancer harbor a germline PTEN<br />

mutation. Thus, cost-efficient markers capable <strong>of</strong> shortlisting thyroid<br />

cancers for CS genetic testing would be clinically useful. Here, we analyzed<br />

the relationship <strong>of</strong> patient PTEN protein levels in predicting the presence <strong>of</strong><br />

germline PTEN mutations. Methods: We conducted a 5-year, multi-center<br />

prospective study <strong>of</strong> 2792 CS and CS-like patients, all <strong>of</strong> whom had<br />

comprehensive PTEN analysis done. Analysis <strong>of</strong> PTEN and downstream<br />

proteins by immunoblotting was performed on total protein lysates from<br />

patient-derived lymphoblast lines. We compared PTEN protein expression<br />

levels between patients with pathogenic PTEN mutations (PTENmut� ), and<br />

those with variants <strong>of</strong> unknown significance (VUS) or wildtype PTEN. PTEN<br />

immunohistochemistry (IHC) was performed on available thyroid samples.<br />

Results: Of 2792 CS/CS-like patients, 722 had thyroid cancer. PTEN<br />

germline pathogenic mutations were present in 36/722 (5%) patients.<br />

PTEN mut�<br />

cases presented at an earlier age (35 vs 42 years; p�0.02). PTEN<br />

frameshift and truncation mutations accounted for 55% <strong>of</strong> mutations. 95%<br />

(19/20) <strong>of</strong> PTENmut� patients had blood-PTEN protein levels in the lowest<br />

quartile as compared to 27% (90/330) <strong>of</strong> PTEN wildtype/PTEN VUS<br />

patients (p�0.001). Low blood-PTEN levels predicted for PTENmut� cases<br />

with a 99.6 % NPV (95%CI 97.7- 99.9) and a positive test likelihood ratio<br />

<strong>of</strong> 3.5 (95%CI 2.8-4.3). Affected thyroid tissues (n�22) from PTENmut� cases showed loss <strong>of</strong> PTEN protein by IHC, correlating with patient blood<br />

PTEN levels (r2�0.30, p�0.016). Conclusions: Our study shows that<br />

PTEN protein dosage could serve as a molecular correlate predicting for<br />

germline PTEN mutation in CS and CS-like presentations <strong>of</strong> thyroid cancer.<br />

The clinical utility <strong>of</strong> PTEN IHC in identifying thyroid cancer patients for<br />

germline PTEN testing should be further explored.<br />

1510 <strong>Clinical</strong> Science Symposium, Sat, 8:00 AM-9:30 AM<br />

Meta-analysis <strong>of</strong> cancer risk among users <strong>of</strong> insulin glargine. Presenting<br />

Author: Alice Koechlin, International Prevention Research Institute, Lyon,<br />

France<br />

Background: The association between diabetes, its risk factors and treatments,<br />

and cancer risk and death is now high on the clinical and research<br />

agenda. Methods: All data regarding cancer risk and use <strong>of</strong> insulin glargine<br />

has been assembled and meta-analyses performed using state-<strong>of</strong>-the-art<br />

statistical methodology. Glargine is the most studied insulin in this regard.<br />

A random effects model was employed with tests for heterogeneity (I2 ) and<br />

publication bias. These meta-analyses are based on reports from epidemiological<br />

studies involving a total <strong>of</strong> 907,008 diabetic subjects and 2,597,602<br />

person-years <strong>of</strong> observation. Results: Based on independent estimates from<br />

14 studies, the Summary Relative Risk (SRR) for all forms <strong>of</strong> cancer was<br />

(SRR�0.90, 95% CI (0.82, 0.98)) and for breast cancer SRR�1.14 (95%<br />

CI (1.00, 1.29)). For new users <strong>of</strong> glargine, from 7 studies, the SRR for<br />

breast cancer was SRR�1.20 (95% CI (0.90, 1.58)). Based on independent<br />

estimates for 9 studies, for colorectal cancer the SRR was 0.73 (95%<br />

CI (0.59, 0.91)) and for prostate cancer SRR�1.16 (95% CI (1.03,<br />

1.30)). Overall, the risk <strong>of</strong> developing cancer among users <strong>of</strong> insulin<br />

glargine is reduced compared to the risk <strong>of</strong> users <strong>of</strong> other insulins.<br />

Similarly, the risk <strong>of</strong> colorectal cancer is reduced among users <strong>of</strong> glargine.<br />

While above unity, the risks <strong>of</strong> breast cancer and prostate cancer are<br />

increased marginally. Potential limitations to this meta-analysis include<br />

that the comparison group was not the same in all studies but this could<br />

also be seen as a strength. This is not likely to invalidate the findings <strong>of</strong> this<br />

analysis nor would the fact that different adjustments were made in the<br />

individual studies. Conclusions: The current evidence gives no support to<br />

the hypothesis that insulin glargine is associated with an increased risk <strong>of</strong><br />

cancer as compared to other insulins and should give reassurance to<br />

physicians and their patients. Given the short exposure time possible to<br />

glargine (less than 5 years maximum), it is not biologically plausible to have<br />

a causal link to common forms <strong>of</strong> cancer.<br />

1509 <strong>Clinical</strong> Science Symposium, Sat, 8:00 AM-9:30 AM<br />

Diabetes, related factors, and breast cancer risk. Presenting Author: Peter<br />

Boyle, International Prevention Research Institute, Lyon, France<br />

Background: Diabetes and breast cancer are both common conditions in<br />

women and may share common risk factors. Methods: To shed clarification<br />

on the potential association between diabetes, related factors and breast<br />

cancer risk, a comprehensive literature review and formal meta-analysis<br />

was carried out, planned, conducted and reported following PRISMA<br />

guidelines regarding meta-analysis <strong>of</strong> observational studies. Variables<br />

studies in relation to breast cancer risk were adiposity, physical activity,<br />

glycaemic load, glycaemic index, diabetes, IGF-1, fasting glucose, fasting<br />

insulin and C-peptide, adiponectin, metformin and glargine use. The<br />

Summary Relative Risk (SRR) and the corresponding 95% Confidence<br />

Interval (CI) was calculated. Results: For breast cancer at all ages, the<br />

calculated risks were as follows: diabetes (SRR�1.23 95% CI (1.12,<br />

1.34); physical activity (SRR�0.88, 95% CI (0.85, 0.92)); glycaemic load<br />

(SRR�1.05, 95% CI (1.00, 1.10)); glycaemic index (SRR�1.05, 95% CI<br />

(1.00, 1.09)); fasting glucose (SRR�1.14, 95% CI (0.94, 1.37)); serum<br />

insulin (SRR�1.26, 95% CI (0.86, 1.84)); c-peptide (SRR�1.29, 95% CI<br />

(0.91, 1.82)); adiponectin (SRR�1.16, 95% CI (0.93, 1.46)); metformin<br />

(SRR�0.97, 95% CI (0.82, 1.16)); and glargine (SRR�1.10, 95% CI<br />

(0.94, 1.30)). An increase <strong>of</strong> 5 units in Body Mass Index (a weight increase<br />

if 14.5 kg in a person 1.70 metres tall) was associated in post-menopausal<br />

breast cancer (SRR�1.12, 95% CI (1.08, 1.16)) but not at premenopausal<br />

ages (SRR�0.83, 95% CI (0.72, 0.95)). Serum insulin was<br />

associated with breast cancer at post-menopausal ages but not at premenopausal<br />

ages whereas with c-peptide there was a significant association<br />

at pre-menopausal ages but not post-menopausal. For IGF-1, Hodge’s<br />

Standardised Mean Difference (HSMD) was calculated and there was no<br />

significant association with breast cancer at all ages (HSMD�-0.026, 95%<br />

CI (-0.031, 0.084)), at post-menopausal ages (HSMD�0.007, 95% CI<br />

(-0.068, 0.082)) or at pre-menopausal ages (HSMD�-0.065, 95% CI<br />

(-0.009, 0.140)). Conclusions: Key risk factors for breast cancer appear to<br />

be adiposity and physical activity which are both related to the risk <strong>of</strong><br />

developing diabetes.<br />

1511 <strong>Clinical</strong> Science Symposium, Sat, 8:00 AM-9:30 AM<br />

Changes in glucose, insulin, and insulin resistance from presurgery to post<br />

adjuvant treatment for breast cancer. Presenting Author: Emer M. Guinan,<br />

Trinity College Dublin, Dublin, Ireland<br />

Background: Elevated fasting insulin levels at breast cancer diagnosis are<br />

associated with increased risk <strong>of</strong> recurrence and reduced survival. (Goodwin<br />

PJ, et al. JCO. 2012;30:164-171) Weight gain related to adjuvant<br />

treatment coupled with sedentary activity habits may further alter insulinrelated<br />

measures for breast cancer survivors. We present results from a<br />

prospective longitudinal study examining changes in insulin-related markers<br />

from pre breast cancer surgery to post adjuvant treatment. Methods:<br />

Subjects were recruited from St. James’s hospital, Dublin, Ireland at the<br />

time <strong>of</strong> breast cancer diagnosis (stage I-III). Fasting pre-surgery blood<br />

samples were drawn to measure insulin, glucose and glycosylated haemoglobin<br />

A1c (HBA1C). Insulin resistance was calculated using the homeostatic<br />

model assessment (HOMA). Subjects with no evidence <strong>of</strong> active<br />

cancer completed the same measurements 2-5 years later. Paired sample<br />

t-tests were used to measure changes from pre-surgery to post-adjuvant<br />

treatment. Statistical significance was set at P � .05. Results: Forty-four<br />

subjects (n � 22 postmenopausal) completed both measurements (mean<br />

� SD age 54.1�8.6 years). Mean time since diagnosis to follow-up<br />

measures was 3.4�0.8 years. Adjuvant therapy included chemotherapy<br />

(n�37), radiotherapy (n�36), biological therapy (n�8) and concurrent<br />

hormonal therapy (n�37). There was a statistically significant increase in<br />

insulin (mean � SD change 2.8�4.1mU/L, P�.000), insulin resistance<br />

(0.54�1.0, P�.001) and HBA1C (0.1�0.2 %, P� .003). Fasting glucose<br />

levels did not change (P � .06). Conclusions: Results indicate that patients<br />

with breast cancer experience significant increases in insulin, insulin<br />

resistance and HBA1C from pre-surgery to post adjuvant treatment.<br />

Elevated post-treatment insulin levels may further contribute to risk <strong>of</strong><br />

breast cancer recurrence and mortality, while also predisposing to the<br />

development <strong>of</strong> cardiovascular disease and type 2 diabetes mellitus.<br />

Exercise and diet interventions are warranted in this population to improve<br />

markers <strong>of</strong> insulin resistance as a surrogate for improving breast cancer and<br />

overall health outcomes.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


1512 Poster Discussion Session (Board #1), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Routine universal screening for Lynch syndrome in colorectal cancer<br />

patients in the community setting. Presenting Author: Jeffrey M. Goldberg,<br />

Norton Cancer Institute, Louisville, KY<br />

Background: Identification <strong>of</strong> colorectal cancer (CRC) cases that are due to<br />

Lynch Syndrome (LS) is important to prevent secondary malignancies or<br />

development <strong>of</strong> additional malignancies among relatives. Utilization <strong>of</strong><br />

clinical criteria to select CRC patients for laboratory testing fails to identify<br />

a significant proportion <strong>of</strong> patients whose CRC is due to LS. Therefore,<br />

universal screening for LS among CRC patients has been advocated by the<br />

Evaluation <strong>of</strong> Genomic Applications in Practice and Prevention Working<br />

Group. However, the feasibility <strong>of</strong> routine universal screening has not been<br />

evaluated in a community-based, non-research setting. We implemented a<br />

universal screening protocol for LS as part <strong>of</strong> routine care <strong>of</strong> CRC at Norton<br />

Healthcare, a multi-institutional community-based healthcare organization.<br />

Methods: Beginning in October 2009, all resected CRC tumors<br />

automatically underwent immunohistochemistry analysis (IHC) for Lynch<br />

Syndrome-associated mismatch repair gene (MMR) expression. Selected<br />

patients who underexpressed MLH1 also underwent testing for the BRAF<br />

V600E mutation. Patients who had a BRAF V600E mutation were considered<br />

to have sporadic CRC. All other patients with abnormal IHC were<br />

automatically referred for genetic counseling. We retrospectively reviewed<br />

the outcomes <strong>of</strong> genetic counseling in all patients referred through this<br />

universal screening protocol. Results: Between October 2009 and December<br />

2011, 388 patients underwent resection <strong>of</strong> CRC, all <strong>of</strong> whom had IHC<br />

for MMR expression. Seventy patients were identified as having abnormal<br />

IHC. Sixty-two had MLH1/PMS2 underexpression, <strong>of</strong> which 18 underwent<br />

BRAF testing and 13 had a BRAF V600E mutation. Eight had MSH2/MSH6<br />

underexpression. Nine patients have been confirmed to have LS, 31 have<br />

been found not to have LS, and 19 are in the process <strong>of</strong> evaluation. Seven<br />

patients declined genetic counseling, and 4 died before they could be<br />

adequately evaluated. Conclusions: Universal screening for CRC due to LS,<br />

using IHC with select BRAF V600E mutation testing and automated<br />

referral for genetic counseling, is feasible in the community setting.<br />

Funded in part with federal funds: NCI, NIH, Contract No.<br />

HHSN261200800001.<br />

1514 Poster Discussion Session (Board #3), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Use <strong>of</strong> clinical history (CH) for Lynch syndrome (LS) risk assessment: An<br />

economic decision analysis. Presenting Author: Sarmad Sadeghi, Cleveland<br />

Clinic Taussig Cancer Institute, Cleveland, OH<br />

Background: Previous studies comparing screening strategies (STs) for LS<br />

concluded that immunohistochemistry (IHC)-based STs are most cost effective.<br />

However, these analyses generally exclude clinical history (CH)-based criteria<br />

due to concerns for reliability, e.g. Amsterdam (Ams), Bethesda (Beth), MMRpro<br />

(Mpro), MMRpredict (Mpre) and PREMM (PRE). We conducted a comprehensive<br />

comparison <strong>of</strong> all STs. Methods: Using assumptions from published reports we<br />

performed a cost effectiveness analysis (CEA) with a societal viewpoint using<br />

TreeAge s<strong>of</strong>tware. 20 STs for proband (Pd) and general population (GP)<br />

screening in a cohort <strong>of</strong> 100,000 assumed a 3% LS prevalence (Prev) in Pd and<br />

0.23% in GP, 5 first degree relatives (FDRs) per LS diagnosis (Dx), 50% LS Prev<br />

in FDRs, and 90% genetic testing (GT) compliance in Pds and GP and 52% in<br />

FDRs. We used the number <strong>of</strong> LS Dxs as effectiveness measure (EM). Sensitivity,<br />

accuracy, and incremental cost effectiveness ratios (ICERs) were calculated.<br />

Results: The ST <strong>of</strong> Mpro followed by GT is highly sensitive and has an accuracy <strong>of</strong><br />

.997. Assuming 1-1.4 life years saved per Dx based on prior studies, this<br />

strategy is also cost effective. Strategies with greater sensitivity were dominated<br />

or not cost effective. Table shows results in order <strong>of</strong> increasing cost per Dx.<br />

Conclusions: This comprehensive comparison suggests that Mpro is an appropriate<br />

first step in screening for LS in Pds, and its routine use may be considered as<br />

a quality measure in the management <strong>of</strong> colorectal cancer. IHC � BRAF, GT<br />

should be reserved for Pds where CH is incomplete.<br />

ST Sensitivity LS in Pds LS in FDRs ICER<br />

No screening 0 0 0 -<br />

Ams, IHC, GT .17 493 641 -<br />

Ams, GT .2 594 772 -<br />

Mpre, IHC, GT .55 1,546 2,010 -<br />

Mpro, IHC, GT .72 1,994 2,593 $4.9K<br />

Beth, IHC, GT .66 1,838 2,389 -<br />

Mpre, GT .67 1,863 2,422 -<br />

PRE, IHC, GT .73 2,017 2,622 -<br />

Mpro, GT .88 2,403 3,124 $21.6K<br />

Beth, GT .8 2,214 2,878 -<br />

PRE, GT .89 2,430 3,159 $513K<br />

IHC � BRAF, GT .82 2,241 2,913 -<br />

IHC, GT .82 2,241 2,913 -<br />

MSI, GT .84 2,295 2,983 -<br />

PRE GP 4, GT age > 35 .89 110 144 -<br />

PRE GP 3, GT age > 30 .89 120 156 -<br />

PRE GP 2, GT age > 25 .89 129 168 -<br />

PRE GP 1, GT age > 20 .89 138 179 -<br />

MSI � IHC, GT .89 2,430 3,159 -<br />

MSI � IHC � BRAF, GT .89 2,430 3,159 -<br />

Up-front GT<br />

- � dominated.<br />

1 2,700 3,510 $1,053K<br />

Cancer Prevention/Epidemiology<br />

89s<br />

1513 Poster Discussion Session (Board #2), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

An alternative approach to identify women at risk for colorectal cancer.<br />

Presenting Author: Amanda S. Bruegl, University <strong>of</strong> Texas M. D. Anderson<br />

Cancer Center, Houston, TX<br />

Background: Hereditary colorectal cancer (CRC) is preventable; however,<br />

identification <strong>of</strong> individuals at sufficiently high risk to warrant heightened<br />

surveillance is difficult. Lynch Syndrome (LS) is an inherited cancer<br />

syndrome due to germline mutation in a DNA mismatch repair gene. For<br />

women with LS, the lifetime risk <strong>of</strong> endometrial cancer (EC) is 64% and<br />

CRC is 54%. Fifty percent <strong>of</strong> women with LS will present with EC or ovarian<br />

cancer prior to CRC. Therefore, women with LS associated EC represent an<br />

ideal group for CRC prevention. The optimal method to identify women with<br />

LS associated EC is not known. The purpose <strong>of</strong> this study was to determine<br />

the utility <strong>of</strong> Amsterdam II and <strong>Society</strong> <strong>of</strong> Gynecologic Oncology (SGO)<br />

Criteria (modified Bethesda criteria that use EC as the sentinel cancer) in<br />

identifying women with LS associated EC. Our ultimate goal is to identify<br />

women at increased risk <strong>of</strong> CRC. Methods: Immunohistochemistry (IHC) for<br />

DNA mismatch repair proteins and MLH1 methylation analyses were used<br />

to identify LS associated EC among 388 women. EC was designated as LS<br />

if there was loss <strong>of</strong> mismatch repair protein expression. Absence <strong>of</strong> MLH1<br />

methylation was required to confirm LS in tumors with MLH1 protein loss.<br />

Results: Fifty-nine (15.2%) <strong>of</strong> the EC patients tested had LS. These<br />

patients are summarized in the table. Conclusions: <strong>Clinical</strong> criteria to detect<br />

LS identify 17/59 (29%) - 44/59 (74%) <strong>of</strong> women who present with EC<br />

first. EC with MSH2 loss is most likely to occur in younger women and<br />

women with positive family history <strong>of</strong> EC and CRC, features classically<br />

associated with LS. In general, the MSH6 mutation is associated with older<br />

age at diagnosis and fewer familial CRCs, however, we found a large<br />

number <strong>of</strong> MLH1 (50%) and PMS2 (86%) cases diagnosed at greater than<br />

50 years with no family history <strong>of</strong> CRC. Our data suggest that classic<br />

clinical screening criteria are inadequate to detect patients with LS who<br />

present with EC, potentially missing up to 25% <strong>of</strong> these patients.<br />

Gene MLH1 MSH2 MSH6 PMS2 Total<br />

Total number 14 27 11 7 59<br />

Median age at diagnosis (range) 52 44 56 66 50<br />

(42-79) (33-81) (31-76) (45-87) (31-87)<br />

Diagnosis at greater than 50 years 7 8 9 6 30<br />

FH CRC 3 16 4 3 26<br />

Amsterdam criteria 3 13 0 1 17<br />

SGO criteria 11 22 7 4 44<br />

1515 Poster Discussion Session (Board #4), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Next-generation sequencing to identify candidate gene for hereditary mixed<br />

polyposis syndrome. Presenting Author: Michele Caroline Gornick, University<br />

<strong>of</strong> Michigan, Ann Arbor, MI<br />

Background: Rare monogenic disorders are <strong>of</strong>ten due to mutations that are<br />

highly penetrant, extremely rare, and strongly disrupt normal biology.<br />

Hereditary mixed polyposis syndrome (HMPS, OMIM ID %601228), is<br />

characterized by a mixture <strong>of</strong> atypical juvenile polyps, hyperplasic polyps,<br />

sessile serrated adenomas and an increased risk <strong>of</strong> colorectal cancer.<br />

Polyps appear to be inherited in an autosomal dominant fashion. The<br />

putative susceptibility locus initially mapped to 15q13-14, however, the<br />

genetic basis <strong>of</strong> this syndrome is not well understood, and no mutation or<br />

associated variants have been identified. Methods: Germline DNA from four<br />

individuals from a family with clinically and pathologically confirmed<br />

HMPS was subjected to massively parallel sequencing (Illumina GAII).<br />

Affected individuals had an average <strong>of</strong> 30x coverage <strong>of</strong> their exome and 10x<br />

coverage <strong>of</strong> their genome. Sequence calls were filtered using the criteria <strong>of</strong><br />

� 4x coverage, quality score over depth � 50, depth <strong>of</strong> coverage � 360,<br />

allele balance � 0.75, number or MAPQ zero reads at locus � 4. Common<br />

variants were filtered out by excluding variants found in dbSNP (build131),<br />

1000 Genomes Project, the Exome Variant Server or 80 unaffected<br />

controls sequenced by hybrid capture and whole exome sequencing.<br />

Polyphen2 and SIFT were used to predict pathogenicity <strong>of</strong> the novel, shared<br />

variants, and validated by Sanger sequencing. Expression levels <strong>of</strong> novel<br />

variants were examined in available tumor samples using qRT-PCR.<br />

Results: From the 32 previously unidentified nonsense, missense or splice<br />

site variants shared by the family members whose whole genomes were<br />

sequenced, only 5 (4 missense, 1 nonsense) were predicted to be<br />

damaging, leading to a small subset <strong>of</strong> novel candidate genes including<br />

ZNF426, which may be responsible for HMPS within this family.<br />

Conclusions: HMPS extremely difficult to accurately diagnosis and the<br />

genetic basis is unknown. Using next-generation sequencing we were able<br />

to detect previously unidentified low frequency allelic variants including a<br />

novel candidate locus.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


90s Cancer Prevention/Epidemiology<br />

1516 Poster Discussion Session (Board #5), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

PTEN germline mutations in patients first tested for other hereditary cancer<br />

syndromes: Would use <strong>of</strong> risk assessment tools have reduced health care<br />

costs? Presenting Author: Jessica Mester, Genomic Medicine Institute,<br />

Cleveland Clinic, Cleveland, OH<br />

Background: PTEN Hamartoma Tumor Syndrome (PHTS) includes patients<br />

with Cowden (CS) or Bannayan-Riley-Ruvalcaba syndrome (BRRS) with<br />

germline PTEN mutation. Breast, colon, endometrial cancer and GI<br />

polyposis are associated, creating overlap with Hereditary Breast and<br />

Ovarian Cancer (HBOC), Lynch (LS) and adenomatous polyposis syndromes<br />

(APS). We reviewed our PHTS patient series to find how <strong>of</strong>ten testing<br />

criteria for these syndromes were met or testing was pursued before PTEN.<br />

Methods: Patients were prospectively recruited by relaxed International<br />

Cowden Consortium criteria or presence <strong>of</strong> known germline PTEN mutation;<br />

mutations were identified by mutation scanning or MLPA analysis and<br />

confirmed by sequencing or quantitative PCR, respectively. Families were<br />

excluded if diagnosed with CS or BRRS before 1998, family history was<br />

unavailable, or proband was �18 yrs. Pedigree, genetic testing reports, and<br />

medical records were reviewed to determine if patients met HBOC testing<br />

criteria, Amsterdam II or Bethesda 2004 criteria, or had adenomatous<br />

polyps. Risk assessment was conducted for BRCA1/2 via the BRCAPRO,<br />

Myriad II, and Penn II models; for MLH1/MSH2/MSH6 via the PREMM1,2,6<br />

and MMRPro models; and for PTEN via the Cleveland Clinic (CC) PTEN<br />

Risk Calculator. Results: 115 PTEN mutation-positive adult probands were<br />

identified among 3,405 patients. 53 (46.1%) met testing criteria for HBOC<br />

or LS and 34 (29.6%) underwent previous testing for HBOC, LS or APS.<br />

Average risk for BRCA1/2 and MLH1/MSH2/MSH6 mutations in tested<br />

patients were 7.5-16.9% and 33.1-41.4%, respectively; whereas PTEN<br />

mutation risks were 35.9% and 79.7%. No patient tested for APS (average<br />

PTEN mutation risk 73%) had �5 adenomas. An estimated $89,700 was<br />

spent on negative analyses for HBOC, LS and APS among patients<br />

ultimately found to have germline PTEN mutations. Conclusions: PHTS<br />

should be part <strong>of</strong> the differential diagnosis for patients referred for question<br />

<strong>of</strong> HBOC, LS or APS. Pathology review is key for patients with polyposis.<br />

Using the CC PTEN risk calculator enables clinicians to understand when<br />

PTEN mutation risk is high, allowing testing to proceed in a step-wise and<br />

cost-saving manner.<br />

1519 Poster Discussion Session (Board #8), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Pathologic findings at risk-reducing salpingo-oophorectomy among women<br />

at increased ovarian cancer risk: Results from GOG-199. Presenting<br />

Author: Phuong L. Mai, National Cancer Institute, Rockville, MD<br />

Background: Although risk-reducing salpingo-oophorectomy (RRSO) is a<br />

standard management option for women with BRCA1/2 mutations, the lack<br />

<strong>of</strong> large, prospective cohort studies makes estimating the prevalence <strong>of</strong><br />

cancer at RRSO problematic. Methods: GOG-199 is a large, nonrandomized<br />

multi-center trial which enrolled women at high-risk (due to<br />

BRCA mutations or strong family history) <strong>of</strong> ovarian cancer, comparing<br />

surgery at enrollment with serial transvaginal ultrasound and CA-125<br />

screening. RRSO specimens were processed according to a standardized<br />

tissue processing protocol including 2-3mm sectioning <strong>of</strong> both ovaries and<br />

tubes. Results: 2,605 participants were accrued to GOG-199. Of the 1 030<br />

enrolled in the baseline RRSO cohort, 28 were ineligible and 36 declined<br />

surgery after enrollment, resulting in 966 baseline RRSO. Pathology review<br />

demonstrated 4 tubal intraepithelial carcinoma and 20 serous pelvic<br />

cancers, <strong>of</strong> which 12 were identified only microscopically. Among the 20<br />

serous cancers, the predominant or exclusive site <strong>of</strong> involvement was ovary<br />

in 10, fallopian tube in 5, and peritoneum in 5 cases. In addition, 6<br />

endometrial cancers (among the 515 undergoing concomitant hysterectomy)<br />

and 3 adenocarcinomas suggestive <strong>of</strong> metastasis were identified. The<br />

serous pelvic cancer prevalence was: entire cohort�2.1% (20/966), all<br />

BRCA mutation carriers�3.2 (18/558), BRCA1 mutation carriers�3.7%<br />

(12/325), BRCA2 mutation carriers�2.6% (6/231), and mutationnegative�0.5%<br />

(2/402). Compared to those without cancer, women with<br />

serous pelvic cancer were older at surgery (p� .001), and more <strong>of</strong>ten<br />

menopausal (vs pre-menopausal, p� .002), nulliparous (vs parous, p�.04)<br />

and never users <strong>of</strong> tamoxifen (vs ever users, p� .04). Serous pelvic cancers<br />

were more frequent in BRCA mutation carriers (vs no mutation, p� .004),<br />

and among carriers, more common in those with BRCA1 mutations (vs<br />

BRCA2 mutation, p� .02). Conclusions: The prevalence <strong>of</strong> serous pelvic<br />

cancers in this cohort was 3.2% among carriers vs 0.5% among the<br />

mutation-negative but with a strong family history. Our data will be useful<br />

when counseling women at increased ovarian cancer risk who are contemplating<br />

risk-reducing surgery.<br />

1518 Poster Discussion Session (Board #7), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Risk-reducing salpingo-oophorectomy and prophylactic mastectomy among<br />

BRCA mutation “previvors.” Presenting Author: Laura L. Holman, University<br />

<strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: We prospectively evaluated the timing and uptake <strong>of</strong> riskreducing<br />

surgery in a cohort <strong>of</strong> female BRCA mutation carriers that have no<br />

personal cancer history (“previvors”). Methods: Patients at high risk <strong>of</strong><br />

breast and ovarian cancer were enrolled between 2007 and 2011 and<br />

followed in a high-risk ovarian cancer screening clinic. Women were <strong>of</strong>fered<br />

risk-reducing salpingo-oophorectomy (RRSO) and/or prophylactic mastectomy<br />

(PM) per guidelines. Their clinical data were recorded and analyzed<br />

using descriptive statistics. Results: Of 260 BRCA mutation carriers<br />

enrolled, 73 have no personal history <strong>of</strong> cancer and are “previvors.”<br />

Patients have been followed for a median <strong>of</strong> 26.5 months (1-50 months).<br />

The median age is 38 years, 81.1% are white, 16.2% are Ashkenazi<br />

Jewish, and 79.7% are premenopausal. BRCA1 carriers account for 43.2%<br />

<strong>of</strong> participants and 55.4% have a BRCA2 mutation. The majority <strong>of</strong><br />

patients (77.6%) presented for ovarian cancer screening �1 year after<br />

their BRCA testing. In all, 60.8% <strong>of</strong> women underwent prophylactic<br />

surgery: 28.4% chose RRSO, 18.9% chose PM, and 13.5% chose both<br />

procedures. Postmenopausal women were more likely to choose RRSO,<br />

while uptake for both procedures was common for premenopausal women<br />

(Table, p�0.04). RRSO was also more likely in parous than nulliparous<br />

premenopausal women (35.2% vs 9% p�0.001). PM was not associated<br />

with parity (p�0.79). Of women that had both surgeries, 20% had them<br />

concurrently and 20% had PM first. Of the 60% that underwent RRSO first,<br />

all had their second surgery within 14 months. Conclusions: BRCA mutation<br />

“previvors” have a high overall uptake <strong>of</strong> prophylactic surgery. Premenopausal<br />

women are more likely to choose PM than postmenopausal women;<br />

reasons for this are unclear. “Previvors” that choose RRSO and PM typically<br />

have both surgeries within a fairly short timeframe. With the growing<br />

population <strong>of</strong> “previvors” in the US, further study <strong>of</strong> patient preferences<br />

regarding preventative surgery and long-term consequences is needed.<br />

Surgery choices <strong>of</strong> pre- versus postmenopausal women.<br />

None RRSO PM RRSO�PM<br />

Premenopausal 35.6% 23.7% 22% 16.9%<br />

Postmenopausal 20% 46.7% 0% 6.7%<br />

1520 Poster Discussion Session (Board #9), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

A multi-institution study <strong>of</strong> the accuracy <strong>of</strong> BRCAPRO in predicting<br />

BRCA1/BRCA2 mutations in women with ovarian cancer. Presenting<br />

Author: Molly S. Daniels, University <strong>of</strong> Texas M. D. Anderson Cancer<br />

Center, Houston, TX<br />

Background: 10-15% <strong>of</strong> ovarian cancer patients have a germline BRCA1 or<br />

BRCA2 mutation, with significant management implications for both<br />

patients and relatives. Genetic testing decisions are guided in part by the<br />

estimated likelihood <strong>of</strong> identifying a mutation. The BRCAPRO model uses<br />

personal and family history <strong>of</strong> breast and ovarian cancer to calculate the<br />

likelihood <strong>of</strong> a BRCA1/2 mutation. This study’s purpose was to assess the<br />

ability <strong>of</strong> BRCAPRO to accurately determine this likelihood. Methods:<br />

BRCAPRO scores were calculated using CancerGene v5.1 for 589 ovarian<br />

cancer patients referred for genetic counseling at three institutions. The<br />

study population was divided into quintiles by BRCAPRO score, with<br />

cutpoints chosen such that each quintile represented 20% <strong>of</strong> the sample.<br />

Chi-square goodness-<strong>of</strong>-fit test was used to compare observed BRCA1/2<br />

mutations to the number predicted. ANOVA models were used to assess<br />

factors impacting BRCAPRO accuracy. Results: 180/589 (31%) ovarian<br />

cancer patients tested positive for a BRCA1/2 mutation. At BRCAPRO<br />

scores under 40%, more mutations were observed than expected (93<br />

observed vs. 34.1 expected, p�0.001). If patients with BRCAPRO scores<br />

�10% had not been <strong>of</strong>fered genetic testing, almost one-third <strong>of</strong> mutations<br />

(51/180, 28%) would have been missed. Multivariate analysis demonstrated<br />

that BRCAPRO underestimated risk for high grade serous ovarian<br />

cancers but overestimated risk for other histologies (p�0.0001), underestimation<br />

increased as age at diagnosis decreased (p�0.02), and model<br />

performance varied by institution (p�0.02). Conclusions: Ovarian cancer<br />

patients classified as low risk by BRCAPRO are more likely to test positive<br />

than predicted, therefore the BRCAPRO prediction could falsely reassure<br />

patients considering genetic testing. BRCAPRO performance could be<br />

improved by incorporating factors such as ovarian cancer histology.<br />

Alternatively, given the high prevalence <strong>of</strong> BRCA1/2 mutations in high<br />

grade serous ovarian cancer and the apparent limitations <strong>of</strong> using family<br />

history to predict mutation probability, BRCA1/2 genetic testing could be<br />

<strong>of</strong>fered to high grade serous ovarian cancer patients regardless <strong>of</strong> family<br />

history.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


1521 Poster Discussion Session (Board #10), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Alcohol and breast cancer risk in postmenopausal women: The PLCO<br />

experience. Presenting Author: Roni Falk, Division <strong>of</strong> Cancer Epidemiology<br />

and Genetics, National Cancer Institute, Rockville, MD<br />

Background: Moderate alcohol consumption is an established breast cancer<br />

(BC) risk factor. Invasive BC is a heterogeneous disease comprised <strong>of</strong><br />

several histological subtypes with distinct biological features that suggest<br />

etiologic differences. Several studies show the alcohol link may be stronger<br />

for estrogen receptor-positive tumors (ER�/PR� and ER�/PR-) and for<br />

lobular BC. Few have evaluated the role <strong>of</strong> alcohol consumption by BC<br />

histology and hormone receptor status combined. Methods: We evaluated<br />

the risk <strong>of</strong> BC by baseline alcohol consumption in the Prostate, Lung,<br />

Colorectal and Ovarian Cancer Screening Trial cohort (54649 participants,<br />

1786 incident invasive breast cancers) by histology and hormone receptor<br />

status. Histologic subtype-specific associations were evaluated using a<br />

recently developed Cox proportional hazards model to calculate multivariate<br />

hazards ratios (HR) and 95% confidence intervals (CI). Results: 1444<br />

ductal (81%), 233 lobular (13%) and 109 mixed ductal lobular (6%) cases<br />

were identified. The majority were ER�/PR� (ductal: 959 ER�/PR�, 136<br />

ER�/PR-, 229 ER-/PR-; lobular: 179 ER�/PR�,37ER�/PR-, 6 ER-/PR-;<br />

mixed ductal/lobular: 88 ER�/PR�, 6ER�/PR-, 5 ER-/PR-). For ductal<br />

and lobular BC, risks for alcohol consumption were modestly elevated;<br />

compared to never drinkers, women consuming 7-14 drinks/week had a<br />

40% increase in HR in both histologic subtypes. Risks for mixed ductal<br />

lobular cancer were significantly higher, particularly for women consuming<br />

7-14 drinks per week (HR�3.0; 95% CI�1.5, 6.1). For all histologies<br />

combined, alcohol risks were confined to ER�/PR� cancers with HR�1. 6<br />

(95% CI�1.3, 2.1) for women consuming 7-14 drinks/ week; p<br />

trend�0.0005. Similarly, for each histologic subtype, the risk for alcohol<br />

consumption was limited to ER�/PR� tumors, and notably, very few cases<br />

with pure lobular or mixed ductal lobular cancer were either ER�/PR- or<br />

ER-/PR-. Conclusions: Moderate alcohol consumption is associated with<br />

risk <strong>of</strong> ER�/PR� BC in each <strong>of</strong> the predominant histologic subtypes, but<br />

not with hormone receptor-negative cancer. Unlike other studies, we did<br />

not find a link between alcohol consumption and ER�/PR- BC.<br />

1523 Poster Discussion Session (Board #12), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Genetic polymorphisms <strong>of</strong> the OPG gene associated with breast cancer.<br />

Presenting Author: Gunter Assmann, University Medical School <strong>of</strong> Saarland<br />

and Jose Carreras Research Center, Homburg, Germany<br />

Background: The receptor activator <strong>of</strong> NfkappaB (RANK) and osteoprotegerin<br />

(OPG) cascade system have been reported to play a role in the<br />

pathogenesis <strong>of</strong> breast cancer (BC). Genetic variations in the genes coding<br />

for RANK, RANK ligand (RANKL), and OPG are supposed to play a role in<br />

the susceptibility <strong>of</strong> breast cancer. Methods: In the present case-control<br />

study genomic DNA was obtained from 307 BC patients (age: median 56)<br />

and 396 healthy female controls (healthy blood donors, HC; age median:<br />

45). We studied six single nucleotide polymorphisms (SNPs) in the genes<br />

coding for RANK (2 SNPs, rs1805034, rs35211496), OPG (2 SNPs,<br />

rs3102735, rs2073618), and RANKL (2 SNPs, rs9533156, rs2277438)<br />

using TaqMan assay-guided PCR for the respective SNPs. The genotype<br />

and allelic frequencies comparing BC with HC were analyzed with �2 test<br />

for 2x3 and 2x2 tables, respectively. Results: The genotype distributions as<br />

well as the allelic frequencies <strong>of</strong> the SNP rs3102735 in the OPG gene<br />

differed significantly (p�0.006 and p�0.019, respectively) between BC<br />

patients and HC; the genotypes containing the minor allele were more<br />

frequent in BC patients (table). In the OPG SNP rs2073618 the minor<br />

allele C was significantly less frequent in BC patients compared to HC<br />

(43.8 vs. 49.7%; OR: 0.788; p�0.031). In addition, BC patients carried<br />

less frequently the homozygous genotype <strong>of</strong> the minor allele compared to<br />

major allele (18.6 vs. 23.9%, p�0.083). No significant risk was detected<br />

for the other SNPs investigated in this study. Conclusions: This is the first<br />

study reporting a significant association <strong>of</strong> the SNP rs3102735 in the OPG<br />

gene with the susceptibility to develop BC in the Caucasian population. The<br />

minor allele C <strong>of</strong> OPG SNP rs2073618, however, seems to be protective<br />

against BC disease. The impact <strong>of</strong> the OPG SNP rs3102735 (location:<br />

5�near region, chromosome 8q24) and <strong>of</strong> the OPG SNP rs2073618 (a<br />

missense SNP, leading to amino acid exchanges Leu3Asn) on the pathogenesis<br />

<strong>of</strong> BC are unclear.<br />

SNP<br />

Allele /<br />

genotypes<br />

Breast<br />

cancer<br />

Healthy<br />

females<br />

OR<br />

(95% CI) p value<br />

OPG rs3102735 n�614 (%) n�784 (%)<br />

Allele minor C 113 (18.4%) 102 (13.0%) 1.508 0.006<br />

Major T 501 (81.6%) 682 (87.0%) (1.127-2.018)<br />

n�307 (%) n�392 (%)<br />

Genotype CC 10 (3.3%) 5 (1.3%) 0.019<br />

CT 93 (30.3%) 92 (23.5%)<br />

TT 204 (66.4%) 295 (75.3%)<br />

Cancer Prevention/Epidemiology<br />

91s<br />

1522 Poster Discussion Session (Board #11), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Contribution <strong>of</strong> TP53 p.R337H mutation to breast cancer prevalence in<br />

Brazil. Presenting Author: Patricia Ashton-Prolla, UFRGS/HCPA, Porto<br />

Alegre, Brazil<br />

Background: The exact contribution <strong>of</strong> TP53 germline mutations, associated<br />

with Li Fraumeni Syndrome, to the overall burden <strong>of</strong> cancer is still only<br />

partially known. Studies in Southern and Southeastern Brazil have shown<br />

that a specific germline mutation at codon 337 (c.1010G�A; p.R337H),<br />

has incomplete penetrance and may be present in a significant number <strong>of</strong><br />

subjects (estimated frequency at the populational level <strong>of</strong> 1:300 individuals).<br />

In an exploratory approach, the aim <strong>of</strong> the study is to assess the<br />

frequency <strong>of</strong> the p.R337H mutation in women from different Brazilian<br />

regions, diagnosed with breast cancer (BC) before 46 and after 55 years <strong>of</strong><br />

age, and unselected for family history <strong>of</strong> cancer. Methods: Formalin-fixed<br />

paraffin-embedded (FFPE) non-tumoral tissue (lymph nodes or normal<br />

breast) <strong>of</strong> women diagnosed with BC between 2000 and 2010 in 3<br />

pathology laboratories from the Brazilian cities <strong>of</strong> Porto Alegre, São Paulo<br />

and Barretos were obtained retrospectively and analyzed after anonimization.<br />

Genomic DNA was isolated with standard methods and genotyping<br />

performed in duplicates by qPCR (TaqMan assay). Confirmation <strong>of</strong> all<br />

mutation-positive and a sample <strong>of</strong> mutation-negative cases were done by<br />

TP53 exon 10 sequencing or by a second independent qPCR analysis.<br />

Results: Analysis <strong>of</strong> 515 BC-affected women identified the p.R337H<br />

mutation in the germline <strong>of</strong> 70 (8,6%) cases: 49/403 (12,1%) diagnosed<br />

before 46 years and 21/412 (5,1%) diagnosed after 55 years. BC occurred<br />

earlier in p.R337H mutation carriers than in non-carriers (p�0.001).<br />

Conclusions: Preliminary analysis in a sample <strong>of</strong> women with BC indicates<br />

that p.R337H founder mutation is present in a high proportion <strong>of</strong> cases,<br />

especially those diagnosed at a young age. Regional genetic background<br />

and recruitment strategies may account for the different mutation frequencies<br />

observed in the centers <strong>of</strong> study. The occurrence <strong>of</strong> this mutation at<br />

such a high frequency in a particular geographic region has important<br />

implications for disease management and cancer risk counseling for these<br />

patients and families. This mutation likely contributes to a significant<br />

proportion <strong>of</strong> the health burden associated with BC in Brazil. Financial<br />

support: FIPE-HCPA, CAPES, FAPERGS and Glaxo Smith Kline.<br />

1524 Poster Discussion Session (Board #13), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Breast cancer intrinsic subtypes by PAM50 in older women. Presenting<br />

Author: Emily Oldham Jenkins, University <strong>of</strong> North Carolina Lineberger<br />

Comprehensive Cancer Center, Chapel Hill, NC<br />

Background: Breast cancer (BC) incidence dramatically increases with age<br />

and the number <strong>of</strong> older BC patients (pts) in the U.S. is rising. Although<br />

immunohistochemical (IHC) data confirm that the incidence <strong>of</strong> luminal BC<br />

increases with age while the incidence <strong>of</strong> triple negative (TN) BC decreases,<br />

age-specific data on the frequency <strong>of</strong> BC subtypes defined by gene<br />

expression is limited. We characterized the incidence <strong>of</strong> BC intrinsic<br />

subtypes using gene microarrays according to age. Methods: Data from<br />

2,150 pts were pooled from publicly available microarray datasets to<br />

determine the incidence <strong>of</strong> PAM50 breast cancer intrinsic subtypes<br />

(Luminal A [LumA], Luminal B [LumB], HER2-enriched [Her2E], Basallike<br />

[Basal] and Normal-like [Norm]) by age. Adjuvant treatment data<br />

(none, chemotherapy, endocrine therapy or both) were available for 1,741<br />

samples. Relapse-free (RFS) and overall survival (OS) differences were<br />

determined using the Kaplan-Meier method. Results: PAM50 intrinsic<br />

subtypes by age are tabulated below. The incidence <strong>of</strong> luminal (A, B, A�B)<br />

tumors increased with age (p�0.01, p�0.0001, p�0.0001, respectively),<br />

while the percentage <strong>of</strong> basal tumors decreased (p�0.0001). Basal tumors<br />

represented 13% <strong>of</strong> pts aged � 70 years (yrs); <strong>of</strong> the 70% with available<br />

IHC data, 74% were TNBC. Age as a continuous variable was not associated<br />

with RFS (p � 0.37). Subtype alone (n�2031), and after controlling for<br />

treatment (n�1645), was significantly associated with RFS (both<br />

p�0.0001). The addition <strong>of</strong> age to a multivariate analysis added no<br />

prognostic information. Conclusions: Though more favorable subtypes<br />

increase with age, older BC pts still have a substantial percentage <strong>of</strong><br />

high-risk subtypes. Age alone was not a significant factor in outcome.<br />

Tumor biology as defined by intrinsic subtype is an important clinical<br />

predictor.<br />

PAM50 intrinsic subtypes by age.<br />

Age (total n�2150)


92s Cancer Prevention/Epidemiology<br />

1525 Poster Discussion Session (Board #14), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Cost-effectiveness <strong>of</strong> the 21-gene recurrence score assay in the setting <strong>of</strong><br />

multifactorial decision making for chemotherapy in early-stage breast<br />

cancer. Presenting Author: Shelby D. Reed, Duke <strong>Clinical</strong> Research<br />

Institute, Durham, NC<br />

Background: The objective <strong>of</strong> this study was to incorporate evidence from<br />

two recently-published studies to reevaluate the cost-effectiveness <strong>of</strong> the<br />

21-gene Recurrence Score (RS) assay (Oncotype DX) in the context <strong>of</strong><br />

multifactorial decision making to guide the use <strong>of</strong> chemotherapy for<br />

node-negative, estrogen receptor–positive breast cancer in the United<br />

States from the societal and healthcare system perspectives. Methods: We<br />

developed a decision-analytic model to first cross-classify hypothetical<br />

patients by clinicopathologic characteristics according to the Adjuvant!<br />

using risk groups and RS risk groups. We generated estimates <strong>of</strong> long-term<br />

costs, survival, and quality-adjusted survival for the RS-guided and<br />

non–RS-guided strategies using a probabilistic state transition model. In<br />

addition to costs for the 21-gene assay, we assigned attributable costs for<br />

chemotherapy, hormonal therapy, monitoring for disease recurrence, and<br />

distant recurrence. For the societal perspective, we also considered<br />

incremental patient time costs. Costs and survival were discounted at 3%<br />

annually. Results: With the RS-guided strategy, 40.4% <strong>of</strong> patients were<br />

expected to receive chemotherapy relative to 47.3% in the non–RS-guided<br />

strategy. Targeted use <strong>of</strong> chemotherapy in the RS-guided strategy was<br />

expected to increase survival by 0.19 years (95% CI, 0.09 to 0.32) and<br />

0.16 QALYs (95% CI, 0.08 to 0.28). Lifetime direct medical costs were<br />

expected to be $2692 (95% CI, 1546 to 3821) higher with the RS-guided<br />

strategy. The incremental cost-effectiveness ratios (ICERs) were $14,059<br />

per life-year saved (95% CI, $6840-$28,912) and $16,677 per QALY<br />

(95% CI, $7613-$37,219). When incorporating lower indirect costs <strong>of</strong><br />

$950 per patient, the ICERs were $9095 per life-year saved (95% CI,<br />

dominant-$23,397) and $10,788 per QALY (95% CI, $6840-$30,265).<br />

In probabilistic sensitivity analysis, more than 99% <strong>of</strong> the ICERs were less<br />

than $50,000 per life-year saved and per QALY. Conclusions: Our updated<br />

cost-effectiveness estimates are supportive <strong>of</strong> the economic value <strong>of</strong> the<br />

21-gene RS assay in the setting <strong>of</strong> node-negative, estrogen receptor–<br />

positive breast cancer.<br />

1527 Poster Discussion Session (Board #16), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Associations between allergies and risk <strong>of</strong> hematologic malignancies:<br />

Results from the Vitamins and Lifestyle (VITAL) cohort study. Presenting<br />

Author: Mazyar Shadman, University <strong>of</strong> Washington, Seattle, WA<br />

Background: Several case-control studies have suggested a reduced risk <strong>of</strong><br />

hematologic malignancies (HMs) for individuals with allergies, but results<br />

have been inconsistent, and prospective cohort studies have not confirmed<br />

this association. Herein, we used the large prospective VITAL study to<br />

examine this association. Methods: 64,847 men and women, aged 50-76,<br />

completed a baseline questionnaire in 2000-2002 and reported on their<br />

current allergies and history <strong>of</strong> physician-diagnosed asthma. Individuals<br />

with prior cancer other than non-melanoma skin cancer were excluded from<br />

this analysis. Incident HMs were identified through December 2009 by<br />

linkage to the SEER registry; those with a diagnosis <strong>of</strong> a non-HM during<br />

follow-up were censored at the time <strong>of</strong> that diagnosis. Multivariable Cox<br />

proportional hazards models were built with adjustment for sex, race/<br />

ethnicity, education, smoking, self-rated health, physical activity, history<br />

<strong>of</strong> anemia in the year before baseline, vegetable use and family history <strong>of</strong><br />

leukemia or lymphoma. Results: Our analysis included 64,839 participants,<br />

among whom 681 (1.03%) incident HMs were found (MDS [69],<br />

AML (41], myeloproliferative disorders [60], mature B-cell neoplasms<br />

[262], chronic lymphocytic leukemia [107], plasma cell disorders [83],<br />

Hodgkin lymphoma [23], T-cell/NK-cell neoplasms [22], and others [14]).<br />

In multivariable analyses, a history <strong>of</strong> any allergy was associated with<br />

increased risk <strong>of</strong> HM (HR�1.21; 95% CI 1.02-1.43, p�0.02). This<br />

association was seen for current allergies to plants/grass/trees (HR 1.27<br />

[1.06-1.52], p�0.001) but not for allergies to mold/dust, animals, insects,<br />

food, or medications. We did not find an association between a history <strong>of</strong><br />

asthma and incident HMs (HR�0.95 [0.72-1.26]). Conclusions: Our study<br />

indicates a moderately increased risk <strong>of</strong> HMs inindividuals with a history <strong>of</strong><br />

allergy, especially to plant, grass or trees. While no causality can be<br />

inferred, this may suggest a possible carcinogenic role for chronic stimulation<br />

<strong>of</strong> the immune system. However, further mechanistic investigations<br />

focusing on the biochemical markers <strong>of</strong> immune system as well as on<br />

possible gene effect modifiers are needed.<br />

1526 Poster Discussion Session (Board #15), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Racial disparities in treatment patterns and clinical outcomes in patients (pts)<br />

with HER2� metastatic breast cancer (MBC). Presenting Author: Adam Brufsky,<br />

University <strong>of</strong> Pittsburgh School <strong>of</strong> Medicine, Pittsburgh, PA<br />

Background: Data examining prognosis and treatment outcomes for black pts<br />

with HER2� MBC are limited. Methods: registHER is a large, observational<br />

cohort <strong>of</strong> pts (N�1,001) with HER2� MBC diagnosed w/in 6 mos <strong>of</strong> enrollment<br />

and followed until death, disenrollment, or 6/09 (median follow-up 27 mos).<br />

Demographics, treatment patterns, and clinical outcomes were described for<br />

black (n�126) and white pts (n�793). Progression Free Survival (PFS) and<br />

Overall Survival (OS) were examined. Multivariate analyses adjusted for baseline<br />

and treatment factors. Results: Black pts were more likely to be obese (BMI �<br />

30), have diabetes, and a history <strong>of</strong> cardiovascular disease (CVD) than white pts<br />

(Table). Black pts were less likely to have estrogen receptor (ER)/progesterone<br />

receptor (PR)� disease and more likely to present with stage IV MBC. In<br />

trastuzumab (T)-treated pts, incidence <strong>of</strong> cardiac safety events (grade �3) was<br />

higher in black (13/119 [10.9%]) than white pts (59/746 [7.9%]). Unadjusted<br />

median OS (mos) was significantly lower (blacks: 27.1, 95%CI 23.2-32.1;<br />

whites: 37.3, 95%CI 34.6-41.1) and median PFS (mos) was lower (blacks: 7.0,<br />

95%CI 5.7-9.7; whites: 10.2, 95%CI 9.3-11.2) in black than white pts. The<br />

adjusted OS hazard ratio (HR) for black vs. white was 1.32 (95%CI 1.04, 1.69);<br />

the adjusted PFS HR was 1.31 (95% CI 1.07, 1.61). Conclusions: These<br />

population-based data show poorer prognostic factors and independently worse<br />

clinical outcomes in black vs. white pts, and represent the largest database to<br />

date with black pts with HER2� MBC. Further research is needed to explore the<br />

basis for the differences noted in this hypothesis-generating analysis.<br />

Demographic and clinical characteristics <strong>of</strong> black and white pts.<br />

Black<br />

White<br />

Variable (%)<br />

(n�126)<br />

(n�793)<br />

Age (y), median (min, max)<br />

BMI (kg/m<br />

50 (20,90) 54 (22,92)<br />

2 )<br />

-<br />

-<br />

30<br />

50.8<br />

32.4<br />

ER/PR status<br />

-<br />

-<br />

ER� or PR�<br />

42.1<br />

54.7<br />

ER- and PR-<br />

54.8<br />

41.5<br />

Unknown<br />

3.2<br />

3.8<br />

Stage, initial diagnosis<br />

-<br />

-<br />

IV<br />

31.0<br />

26.6<br />

I-III, MBC 12 mos<br />

50.8<br />

61.0<br />

Baseline CVD 30.2 15.8<br />

Diabetes 13.5 6.5<br />

T-based 1st-line regimens<br />

(n�102)<br />

(n�670)<br />

C only<br />

77.5<br />

65.4<br />

HT only<br />

4.9<br />

6.3<br />

C and HT<br />

11.8<br />

20.6<br />

T alone<br />

5.9<br />

7.8<br />

Abbreviations: BMI, body mass index; C, chemotherapy; HT, hormone therapy.<br />

1529 Poster Discussion Session (Board #18), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Outcomes from an electronic medical record (EMR)-based standardized<br />

tobacco assessment and cessation program in a NCI-designated comprehensive<br />

cancer center. Presenting Author: Graham Walter Warren, Roswell Park<br />

Cancer Institute, Buffalo, NY<br />

Background: Tobacco assessment and cessation is advocated by ASCO and<br />

national clinical oncology guidelines, but there is little information on large<br />

scale clinically efficient models to assess tobacco use and provide<br />

cessation in a structured evidence based manner. Automation through the<br />

electronic medical record (EMR) could reduce subjective interpretation by<br />

clinicians and assist in increasing data tracking accuracy to enhance<br />

meaningful use initiatives. Methods: A standard set <strong>of</strong> evidence based<br />

tobacco assessment questions were incorporated into an annotated fixedvariable<br />

response system in the EMR delivered by nursing at initial consult<br />

and at follow-up. A logic based EMR referral system was developed to<br />

determine patient eligibility for mandatory automated referral to a dedicated<br />

tobacco cessation service. An evidence based institutional clinical<br />

cessation program was developed to provide cessation support to referred<br />

patients. The evidence based screening and referral algorithms will be<br />

presented. Results: Over 13 months, 677 patients were referred and 529<br />

patients were successfully contacted to date for cessation support. In the<br />

529 patients, 21 (3.9%) were inappropriate referrals (never smokers or<br />

long term former smokers), 48 patients (9.1%) did not want to enroll, but<br />

wanted to discuss cessation at a later date. Notably, only 18 patients<br />

(3.4%) refused any intervention. In a total <strong>of</strong> 415 patients enrolled in the<br />

cessation program, 104 patients (25.1%) were thinking about quitting<br />

(contemplation), 134 patients (32.3%) were preparing to quit, 169<br />

patients (40.7%) were quitting (action phase), and 8 patients (1.9%) have<br />

relapsed. Tobacco assessments and automated referrals through the EMR<br />

took a median <strong>of</strong> 4 minutes to complete. Conclusions: A large volume <strong>of</strong><br />

patients were screened and referred to a dedicated cessation program with<br />

low patient refusal for intervention without impeding physician workflow.<br />

These data suggest that this nursing driven EMR based assessment is a<br />

highly efficient clinical model for tobacco assessment and cessation.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


1530 Poster Discussion Session (Board #19), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Potential risk <strong>of</strong> asbestos exposure among Japanese general population:<br />

Japanese general screening study for asbestos-related diseases (JGSARD).<br />

Presenting Author: Nobuhiko Seki, Teikyo University School <strong>of</strong> Medicine,<br />

Tokyo, Japan<br />

Background: The number <strong>of</strong> patients with pleural mesothelioma and lung<br />

cancer associated with asbestos exposure has recently been increasing in<br />

Japan. The aim <strong>of</strong> this study was to evaluate the results <strong>of</strong> screening for<br />

asbestos-related diseases in a group <strong>of</strong> Japanese general population.<br />

Methods: This prospective study was approved by the institutional review<br />

board. From 2006 to 2008, 9810 subjects (5283 men and 4527 women;<br />

mean age, 57 years) underwent chest radiography and low-dose CT<br />

examinations in 26 institutions in Japan. Among them, 6286 (64.1%)<br />

subjects underwent subsequent CT examinations after 2 years <strong>of</strong> interval.<br />

<strong>Clinical</strong> information such as histories <strong>of</strong> smoking and asbestos exposure<br />

was reviewed. Images were interpreted independently by 15 experienced<br />

pulmonologists or chest radiologists. Results: The history <strong>of</strong> asbestos<br />

exposure was definitely present in 1253 (12.8%) subjects, possibly<br />

present in 2058 (21.0%), and absent in 6499 (66.2%). On chest<br />

radiograph, pleural plaque and thickening were seen in 61 (0.6%) and 65<br />

(0.6%) subjects, respectively. On low-dose CT, pleural plaque and thickening<br />

were identified in 264 (2.7%) and 245 (2.5%) subjects, respectively,<br />

and non-calcified pulmonary nodule/mass was seen in 1003 (10.2%).<br />

Furthermore, lung cancer was identified in 29 (0.3%) subjects. The history<br />

<strong>of</strong> asbestos exposure was not confirmed in 77 out <strong>of</strong> 264 subjects (29.2%)<br />

having pleural plaques on low-dose CT. Based on the logistic regression<br />

analysis, pleural plaque on low-dose CT was significantly correlated with<br />

male, age more than 60 years, smoking, and a history <strong>of</strong> asbestos exposure.<br />

Especially, total residential period in asbestos factory area as well as<br />

asbestos exposure work period showed significantly increased relative risk<br />

every 10 years. Similarly, lung cancer was significantly correlated with age<br />

more than 60 years, a history <strong>of</strong> asbestos exposure, and presence <strong>of</strong> pleural<br />

plaques. Conclusions: Our results indicate the presence <strong>of</strong> pleural plaques<br />

on low-dose CT among Japanese general population is closely associated<br />

with potential risk <strong>of</strong> asbestos exposure. However, about 30% <strong>of</strong> such<br />

subjects are not aware <strong>of</strong> a history <strong>of</strong> asbestos exposure.<br />

1533 Poster Discussion Session (Board #22), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

<strong>Clinical</strong> characteristics and natural history <strong>of</strong> patients with squamous cell<br />

lung carcinoma with FGFR1 amplification. Presenting Author: Marc Bos,<br />

Lung Cancer Group Cologne, Department I <strong>of</strong> Internal Medicine and Center<br />

for Integrated Oncology, University Hospital Cologne, Cologne, Germany<br />

Background: FGFR1 amplifications have been described as a promising<br />

oncogenic target in squamous cell lung cancer. Here we aimed at<br />

describing the clinical characteristics and natural history <strong>of</strong> FGFR1amplified<br />

squamous cell lung cancer patients. Methods: From 01/2011 to 01/2012<br />

we screened 553 squamous cell lung cancer patients in our Network for<br />

Molecular Screening <strong>of</strong> Lung Cancer for the presence <strong>of</strong> FGFR1 amplifications<br />

by FISH analysis in accordance with the local ethics committee.<br />

FGFR1 was defined as amplified if the ratio <strong>of</strong> FGFR1 copies to centromeric<br />

copies was above 2 or if more than 50% <strong>of</strong> tumor cells showed 5 copies or if<br />

more than 15% <strong>of</strong> tumor cells demonstrated clusters <strong>of</strong> FGFR1. <strong>Clinical</strong><br />

data were collected by extracting information from medical records, the<br />

local cancer registry and by questioning treating physicians and patients.<br />

Results: FGFR1FISH analysis could be performed in 95% <strong>of</strong> the cases and<br />

was amplified in 16%. Of the amplified cases 75 % were male and 25%<br />

female without significant enrichment for male or female. The median age<br />

<strong>of</strong> the patients at diagnosis was 67 yrs (range 46 - 82). Stage at<br />

presentation was: 16% I; 17.3% II; 26.7% IIIa, 40% IIIb/IV. 97,3% <strong>of</strong> the<br />

patients were former or active smokers with a median <strong>of</strong> 40 pack years. The<br />

median progression free survival <strong>of</strong> patients with stage IIIb/IV disease was<br />

11 months (95% CI 8-14; n�14). The median overall survival was not yet<br />

reached after a median follow-up time <strong>of</strong> 14 months (95% CI 11 - 17;<br />

n�24). We further screened for co-existing genetic lesions such as<br />

mutations in EGFR, BRAF, KRAS, PIK3CA as well as translocations <strong>of</strong> ALK<br />

and amplifications <strong>of</strong> ERBB2. Two patients demonstrated co-occurring<br />

PIK3CA mutations and one a BRAFmutation. Conclusions: Screening for<br />

FGFR1 is feasible under routine clinical conditions. By implementation <strong>of</strong> a<br />

regional molecular screening network the ability to screen for FGFR1<br />

amplification was successfully expanded to non-academic centers and<br />

private practices. FGFR1 amplifications in squamous cell cancer <strong>of</strong> the<br />

lung are frequent (16%) and associated with smoking history. Screening for<br />

FGFR1 might pave the way for the application <strong>of</strong> new FGFR1 directed<br />

targeted drugs in squamous cell lung cancer.<br />

Cancer Prevention/Epidemiology<br />

93s<br />

1531 Poster Discussion Session (Board #20), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Never-smoking women with lung cancer from the Spanish WORLD07<br />

database. Presenting Author: Dolores Isla, Hospital Lozano Blesa, Zaragoza,<br />

Spain<br />

Background: Gender differences in lung cancer (LC) have been reported,<br />

but with many unresolved issues yet. Tobacco causes the majority <strong>of</strong><br />

women lung cancer (WLC), although the rate <strong>of</strong> never-smoking WLC is<br />

higher than in men. Several factors may play etiologic roles, and an<br />

in-depth understanding is needed. Methods: WORLD07 is a Spanish<br />

prospective, multicenter, epidemiologic female-specific LC database sponsored<br />

by ICAPEM, a pr<strong>of</strong>essional association committed with WLC research.<br />

In order to improve the knowledge on never-smoking WLC,<br />

information has been extracted from WORLD07 database. Results: From<br />

October/2007 to October/2011, 1371 newly diagnosed WLC were included<br />

in an e-database from 32 centers, 539 (39.3%) never-smoking.<br />

Patient (p) characteristics: median age 71.1 years(y); median age <strong>of</strong><br />

menarche 13y.; motherhood 91.2% (median 2.3 children, median age at<br />

first child 26.4y); oral contraceptive use 11.9%; postmenopausal 88.9%<br />

(median age <strong>of</strong> menopause 49y); HRT 5.2%; second-hand smokers 40%<br />

(work-exposure 17.1%, home-exposure 88.8%); obesity 16.3%; familiar<br />

history <strong>of</strong> cancer 39.9% (LC 29.8%); previous history <strong>of</strong> cancer 13%<br />

(breast/lung/cervix: 41.4/5.7/2.9%); current LC histology(%): adenocarcinoma/SqCC/LCC/SCLC:<br />

83.4/6.2/5.5/3.9; EGFR mut� (268 p analyzed):<br />

55.5% (exon 19/20/21(%): 61.1/7.4/36.9); TNM NSCLC I/II/III/IV(%):<br />

14/3.3/19.8/60.3. Treatment: EGFR-TKI in p EGFR mut�, stage IV(1st-/ 2nd-line)(%): 51.7/15.4; stage IV NSCLC (1st-line/2nd-line): platinumbased<br />

CT 42.5%, EGFR-TKI 33.5%, combinations with bevacizumab<br />

2.9% / EGFR-TKI 15.8%. Overall survival: median 27 months(m), 1/2y(%)<br />

74.8/55.2; stage IV NSCLC: median 20.5m, 1/2-y(%) 67/46;<br />

EGFR-mut� p: median 27.3m, 1/2-y(%) 75/54.3. Conclusions: According<br />

to our e-database, WLC showed high rates <strong>of</strong> never-smokers (39.3%), and<br />

<strong>of</strong> relatives diagnosed with malignant tumours (39.9%, �1/3 LC). Adenocarcinoma<br />

was the most frequent histology (76.1%), and more than half <strong>of</strong><br />

the cases analyzed harboured EGFR mutations. Although 40% were<br />

second-hand smokers, further investigations are warranted. Survival outcomes<br />

remain satisfactory, as expected from this selected subgroup <strong>of</strong> p.<br />

Additional epidemiologic and treatment data will be presented.<br />

1534 Poster Discussion Session (Board #23), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Molecular epidemiological prospective study <strong>of</strong> EGFR mutations from<br />

Asian patients (pts) with advanced lung adenocarcinoma (PIONEER).<br />

Presenting Author: Pan-Chyr Yang, National Taiwan University Hospital,<br />

College <strong>of</strong> Medicine, Taipei, Taiwan<br />

Background: The epidemiological, multicenter prospective study<br />

(NCT01185314; PIONEER) assessed EGFR mutation (M) rates in pts from<br />

Asia with newly diagnosed advanced lung adenocarcinoma (ADC). Influence<br />

<strong>of</strong> demographic and clinical factors on EGFR M rates was investigated.<br />

Methods: Pts were aged �20 years, with treatment naïve stage<br />

IIIB/IV lung ADC. Tumor sample (biopsy, surgical specimen, cytology)<br />

EGFR M status (primary endpoint; positive [M�], negative [M-], undetermined<br />

[MU]) was determined using Scorpion ARMS (Therascreen EGFR<br />

RGQ kit). EGFR M frequency was calculated and compared between<br />

demographic/clinical factor subgroups (chi-square/Fisher’s exact test).<br />

Factors with p�0.05 in the univariate analysis were further analyzed by<br />

multivariate logistic regression at 1% significance level to take the large<br />

dataset into consideration. Results: Of 1482 pts from 7 Asian countries,<br />

43.4% were female, mean age was 60 years (range 17-94), and 52.6%<br />

were never-smokers. EGFR M status was evaluated in 1450 patients (32<br />

[2.2%] were MU): 746 (51.4%) were M�, 704 (48.6%) were M-. Country,<br />

gender, ethnicity, smoking status, pack years, metastasis, (all p�0.001)<br />

and disease stage (p�0.009) correlated significantly with EGFR M status.<br />

EGFR M� rate by country: Vietnam 64.2% (77/120), Taiwan 62.1%<br />

(108/174), Thailand 53.8% (63/117), Philippines 52.3% (34/65), China<br />

50.2% (372/741), Hong Kong 47.2% (76/161), India 22.2% (16/72).<br />

EGFR M� rates were 61.1% in females, 44.0% in males, 60.7% in never<br />

smokers, and 31.4% in heavy smokers (�50 pack years). EGFR M� rates<br />

were 37.5% in male regular smokers (113/301), 34.8% in female regular<br />

smokers (8/23), 56.5% in male never smokers (104/184) and 62.0% in<br />

female never smokers (358/577). Ethnic group (p�0.001) and pack years<br />

(p�0.001) were identified as important factors by logistic regression;<br />

gender was not significant when adjusted for smoking status. Conclusions:<br />

The overall EGFR M� rate in unselected ADC was 51.4%. Ethnicity and<br />

pack years were found to have a statistically significant association with<br />

EGFR M rates. There was no association between gender and EGFR M rates<br />

when adjusted for smoking status.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


94s Cancer Prevention/Epidemiology<br />

1535 Poster Discussion Session (Board #24), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Genetic sequence variant (GSV) in TERT-CLPTM1L (5p15.33 locus) and<br />

survival in platinum-treated stage-IV non-small cell lung cancer (NSCLC).<br />

Presenting Author: Abul Kalam Azad, Ontario Cancer Institute, Princess<br />

Margaret Hospital, Toronto, ON, Canada<br />

Background: Lung cancer is a leading cause <strong>of</strong> cancer-related mortality in<br />

North America. Besides tobacco smoking, inherited genetic factors can<br />

also influence the development <strong>of</strong> lung cancer. These genetic factors may<br />

lead to biologically distinct subsets <strong>of</strong> cancers that have different outcomes.<br />

We evaluated whether GSVs associated with lung cancer risk are<br />

also associated with overall survival (OS) and progress free survival (PFS) in<br />

platinum-treated stage-IV NSCLC patients. We chose this subset <strong>of</strong><br />

patients with lung cancer because they are uniformly treated and followed<br />

up at our institute (Princess Margaret Hospital). Methods: A total <strong>of</strong> 32<br />

candidate GSVs in 21 genes previously reported to be associated with lung<br />

cancer risk were genotyped in 188 platinum-treated NSCLC stage-IV<br />

patients. Illumina Custom GoldenGate Genotyping Panel was used for the<br />

genotyping assay platform. Multivariate Cox proportional hazard models<br />

adjusted for the potential clinical prognostic factors were generated for OS<br />

and PFS. Results: Median age is 60 yr (range: 31-81); 73% with<br />

adenocarcinoma. Median follow-up time is 27 mo; median OS is 16 mo and<br />

PFS is 8 mo. The top significant GSV was rs4975616 (g.1315660G�A) in<br />

telomerase reverse transcriptase (TERT) and cleft lip and palate transmembrane<br />

1-like (CLPTM1L) gene on chromosome 5p15.33. The adjusted<br />

hazard ratio (aHR) for OS was 0.76 (95% CI: 0.58-0.99; p�0.04) and for<br />

PFS aHR was 0.70 (95% CI: 0.55-0.90; p�0.005) for each protective<br />

allele, respectively. This GSV has previously been associated with lung<br />

cancer risk in genome-wide association study (PMID: 19654303).<br />

Conclusions: The TERT-CLPTM1L:rs4975616 GSV is not only a risk factor<br />

for lung cancer but also is associated with survival in patients with stage-IV<br />

NSCLC treated with platinum based chemotherapy. GSVs may be able to<br />

define subsets <strong>of</strong> patients with different risk and prognosis <strong>of</strong> NSCLC.<br />

Future studies should evaluate whether this GSV is predictive or prognostic.<br />

1537 General Poster Session (Board #1A), Sat, 1:15 PM-5:15 PM<br />

Correlation <strong>of</strong> breast cancer risk in European BRCA2 mutation carriers with<br />

birth cohort. Presenting Author: Muy-Kheng Maria Tea, Medical University<br />

<strong>of</strong> Vienna, Department <strong>of</strong> OB/GYN, Division <strong>of</strong> Senology, Vienna, Austria<br />

Background: Mutations in the Breast Cancer Gene 1 (BRCA1) and Breast<br />

Cancer Gene 2 (BRCA2) lead to an elevated risk <strong>of</strong> developing breast (BC)<br />

and ovarian cancer (OC). However, risk estimates vary, depending on the<br />

study population. Furthermore, there are indications that birth cohort can<br />

influence the cancer risk. We investigated the risks for BC and OC<br />

associated with BRCA2 mutations in a cohort <strong>of</strong> female mutation carriers <strong>of</strong><br />

a genetically heterogeneous central European population who were identified<br />

by molecular genetic testing in our institute. Methods: This study<br />

included 171 Caucasian women from Austria who underwent genetic<br />

counseling and where molecular genetic analysis identified a mutation in<br />

the BRCA2 gene at the Medical University <strong>of</strong> Vienna, Division <strong>of</strong> Senology,<br />

in Austria. A total <strong>of</strong> 57 healthy and 114 affected BRCA2-carriers were<br />

detected. The risk was estimated using the product limit method. The log<br />

rank test was used to compare different strata. Results: The risk <strong>of</strong><br />

developing cancer to age 70 was found to be 85% for BC (95% CI<br />

77–93%) and 31% for OC (95% CI 16–46%) in our BRCA2 study<br />

population. Female BRCA2-carriers born in 1958 or later were at a<br />

significantly higher risk <strong>of</strong> developing BC (p�0.001; 88% vs. 46% to age<br />

40) but not <strong>of</strong> OC (p is not significant; 0% vs. 2% to age 40) compared to<br />

mutation carriers born earlier. Conclusions: We conclude that female<br />

BRCA2 mutation carriers should also be counseled about their cohortdependent<br />

cancer risk, especially for breast cancer. Further research about<br />

variables that may affect cancer risk like lifestyle-related factors should be<br />

considered.<br />

1536 Poster Discussion Session (Board #25), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Second malignant neoplasms (SMN) among 18,627 testicular cancer<br />

survivors (TCS) after chemotherapy (CHEM) or surgery (SURG). Presenting<br />

Author: Chunkit Fung, James P. Wilmot Cancer Center, University <strong>of</strong><br />

Rochester School <strong>of</strong> Medicine and Dentistry, Rochester, NY<br />

Background: Increased risks <strong>of</strong> SMN after radiotherapy (RT) for testicular<br />

cancer (TC) are well established. Few population-based studies, however,<br />

have focused on SMN risk among a contemporary group <strong>of</strong> TCS managed<br />

initially with non-RT approaches, including CHEM. Methods: Standardized<br />

incidence ratios (SIR) <strong>of</strong> SMN stratified by site and time since TC diagnosis<br />

were calculated for 18,627 TCS reported to the SEER program (1980-<br />

2008) who initially had CHEM (n�8,058) or SURG (n�10,569) alone<br />

without RT, with each cohort accruing 65,398 and 92,681 person-years<br />

(PY) <strong>of</strong> follow-up respectively. Results: After CHEM, significantly increased<br />

risks <strong>of</strong> solid cancers (n�154; SIR 1.3; 95% CI 1.1-1.5; absolute excess<br />

risk (AER) 5.4 per 10,000 PY) and leukemias (n�18, SIR 3.9; 95% CI<br />

2.3-6.1; AER 2.0) were observed. Solid cancer risk remained elevated for<br />

� 20 yrs, whereas excess leukemias were concentrated within 10 years<br />

after diagnosis. SIRs for solid cancers during the �1, 1-4, 5-9, 10-14,<br />

15-19, and 20� yr periods were 2.0 (95% CI 1.03-3.5), 1.4 (95% CI<br />

0.97-2.0), 0.8 (95% CI 0.5-1.2), 1.3 (95% CI 0.9-1.8), 1.6 (95% CI<br />

1.05-2.2), and 1.5 (95% CI 0.95-2.2) respectively (P-trend 0.5) whereas<br />

SIRs for leukemia were 3.2, 9.9 (P�0.05), 2.6, and 2.3 respectively, with<br />

no cases reported in the latter 2 intervals. Median latencies to the<br />

development <strong>of</strong> solid cancers and leukemia were 12.5 yr (0.1-28) and 2.5<br />

yr (0.1-14) respectively. Increased site-specific risks were apparent for<br />

cancers <strong>of</strong> liver (SIR 1.9; 95% CI 0.6-4.4); pancreas (SIR 2.1; 95% CI<br />

0.8-4.6); s<strong>of</strong>t tissue (SIR 4.9; 95% CI 2.1-9.7); bladder (SIR 1.9; 95% CI<br />

1.02-3.3); kidney (SIR 2.6; 95% CI 1.4-4.3); brain/CNS (SIR 1.8; 95% CI<br />

0.7-3.7), and thyroid (SIR 3.9; 95% CI 2.1-6.6). Secondary leukemias<br />

included 16 non-lymphocytic and 2 lymphocytic leukemias. In contrast,<br />

among TCS managed initially with SURG alone, no excess solid cancers<br />

were observed (n�198; SIR 1.0; 95% CI 0.8-1.1), albeit an increased risk<br />

<strong>of</strong> leukemia (n�15; SIR 1.9; 95% CI 1.1-3.2, AER 0.8) was seen.<br />

Conclusions: Future analytic studies should further evaluate the sitespecific<br />

risks <strong>of</strong> SMN after modern CHEM for TC and the baseline risk<br />

among patients managed with non-cytotoxic approaches.<br />

1538 General Poster Session (Board #1B), Sat, 1:15 PM-5:15 PM<br />

Identification <strong>of</strong> new susceptibility genes in familial breast cancer by<br />

exome sequencing. Presenting Author: Henry T. Lynch, Creighton University,<br />

Omaha, NE<br />

Background: Familial breast cancer was described in the early 1970s with<br />

its syndromy confirmed by the discovery <strong>of</strong> BRCA germline mutations in the<br />

1990s. BRCA mutations account for less than 10% <strong>of</strong> affected families.<br />

Efforts made in the past two decades have resulted in limited results in<br />

identifying additional susceptibility genes for familial breast cancer.<br />

Methods: Using exome sequencing, we analyzed eight members in a<br />

BRCA1-, BRCA2-, p53- and PTEN-negative breast cancer family; five with<br />

breast cancer and three unaffected. Mutation candidates were idenified by<br />

bioinformatics analysis, experimentally validated by Sanger sequencing<br />

and their damaging effect was predicted by the SIFT program. Validated<br />

mutations were also tested in 42 additional breast cancer samples from<br />

BRCA-negative breast cancer families. Results: We identified 55 nonsynonymous<br />

germline mutations affecting 49 genes in multiple members<br />

<strong>of</strong> this family, 20 <strong>of</strong> 22 selected mutations predicted to cause damaging<br />

effects were validated. As an exemplar, two mutations in the MYST4 gene<br />

were detected at the C terminal (p.D1516Y and p.R1577C), validated, and<br />

predicted to cause damaging consequences. MYST4 is a histone acetyltransferase<br />

(Champagne, N. et al. JBC. 274, 28528-28536,1999). It contains<br />

a C2HC-type finger and a PHD-type zinc finger. Its N-terminal is involved in<br />

transcriptional repression while its C-terminal is involved in transcriptional<br />

activation and interacts with important transcriptional regulators RUNX1<br />

and RUNX2. MYST4 is involved in DNA replication, transcriptional regulation,<br />

and epigenetic modification <strong>of</strong> chromatin structure. A translocation<br />

t(10;16)(q22;p13) between CREBBP and MYST4 was identified in acute<br />

myeloid leukemia. The 20 validated germline mutations were tested by<br />

Sanger sequencing in 42 additional breast cancer cases from 26 BRCAnegative<br />

families. None <strong>of</strong> the 20 mutations were detected. Conclusions:<br />

Results show that remaining unknown susceptibility genes are likely<br />

family-specific and can be detected in each affected family using genome<br />

sequencing approaches. These results may be the first reported in an exome<br />

sequencing study <strong>of</strong> BRCA-negative breast cancer families.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


1539 General Poster Session (Board #1C), Sat, 1:15 PM-5:15 PM<br />

Evaluation <strong>of</strong> genes associated with undescended testes as predictors <strong>of</strong><br />

TGCT susceptibility. Presenting Author: Ariela Marshall, Hospital <strong>of</strong> the<br />

University <strong>of</strong> Pennsylvania, Philadelphia, PA<br />

Background: Testicular germ cell tumors (TGCTs) are highly heritable.<br />

Cryptorchidism (undescended testes; UDT) is a strong risk factor for TGCT,<br />

with increased risk to both the ipsilateral and contralateral testes. However,<br />

recent genome-wide association studies (GWAS) identifying genetic variants<br />

associated with TGCT susceptibility have not found differences in<br />

genotype carriage between TGCT patients with and without UDT. We<br />

investigated the role <strong>of</strong> potential risk variants for UDT as risk factors for<br />

TGCT. Methods: 1300 SNPs in and around 25 gene regions with published<br />

associations with UDT were evaluated in 474 TGCT cases (45 with UDT)<br />

and 919 controls. Genotype information was available from the Affymetrix<br />

Genome-Wide Human SNP Array 6.0. Statistical analysis was performed<br />

using PLINK, and statistical significance was assessed by Fisher’s Exact<br />

test. Results: Comparing TGCT cases with and without UDT, variants in the<br />

region <strong>of</strong> four genes (EPHB2, ESR1, SEMA3C, TGFBR3) were suggestively<br />

associated with UDT. When TGCT cases with UDT were compared with<br />

unaffected controls, the associations all met the required level <strong>of</strong> significance<br />

(p � 4x10-5 ). Only variation at ESR1 approached significance<br />

(P�0.0004) when TGCT cases and unaffected controls were compared.<br />

Conclusions: We identified variants at three genetic loci - SEMA3C,<br />

TGFBR3 and EPHB2 - that were significantly associated with UDT, but not<br />

with TGCT. The association <strong>of</strong> variation in EPHB2 and SEMA3C with UDT<br />

are novel findings. While associated with UDT, variation at ESR1 is also<br />

potentially associated with TGCT risk. These data continue to suggest that<br />

genetic risk factors for UDT are largely independent <strong>of</strong> those for TGCT.<br />

Thus, screening for TGCT could be targeted in a UDT population to those<br />

with genetic risk factors. Further studies should be done to investigate the<br />

genetic link between UDT and TGCT.<br />

1541 General Poster Session (Board #1E), Sat, 1:15 PM-5:15 PM<br />

<strong>Clinical</strong> characteristics <strong>of</strong> patients with malignant pleural mesothelioma (MPM)<br />

harboring somatic BAP1 mutations. Presenting Author: Marjorie Glass Zauderer,<br />

Memorial Sloan-Kettering Cancer Center, New York, NY<br />

Background: Efforts to elucidate tumorigenic mutations in MPM are essential to<br />

advance therapy. We reported a 23% incidence <strong>of</strong> somatic mutations in<br />

BRCA1-associated protein-1 (BAP1) in 53 patients with MPM (Bott et al. Nat<br />

Genet 2011). Germline BAP1 mutations were also reported in two families with a<br />

predisposition to mesothelioma and uveal melanoma (Testa et al. Nat Genet<br />

2011). While BAP1 somatic mutations are more common in poor prognosis uveal<br />

melanoma (84% class 2, 4% class 1; Harbour et al. Science 2010), the<br />

significance <strong>of</strong> these mutations in MPM is unknown. Therefore, we analyzed the<br />

clinical characteristics <strong>of</strong> patients with somatic BAP1 mutations in order to<br />

describe this newly identified subpopulation. Methods: We reviewed the charts <strong>of</strong><br />

121 patients with tumors tested for somatic BAP1 mutations. Results: Patient<br />

characteristics are in the Table. Twenty percent harbored somatic BAP1<br />

mutations. Other than the percent <strong>of</strong> current or former smokers (75% BAP1<br />

mutations, 42% BAP1 wild-type, p�0.006), no other clinical feature was<br />

significantly different among those with and without BAP1 mutations. Among 53<br />

samples analyzed for NF2 mutation and p16 deletion, no correlation was seen<br />

with BAP1 mutation. Conclusions: Somatic BAP1 mutations occur in about 20%<br />

<strong>of</strong> MPM tumors. Aside from smoking history, no other differences in clinical<br />

characteristics or outcomes were noted in the BAP1 mutated cases. Similar<br />

efforts are needed to describe the features <strong>of</strong> germline mutations in order to<br />

define this new cancer predisposition syndrome. We are planning a prospective<br />

trial to further evaluate the prevalence <strong>of</strong> germline BAP1 mutations, and we are<br />

also exploring the therapeutic implications.<br />

BAP1 mutants<br />

(N�24)<br />

BAP1 wild-type<br />

(N�97) p value<br />

Gender (M � male) M: 79% M: 68% 0.33<br />

Median age at diagnosis 65 63 0.31<br />

Histology<br />

E: 71% E: 77% 0.28<br />

E � epithelial<br />

M: 25% M: 12%<br />

M � mixed<br />

S � sarcomatoid<br />

S: 4% S: 10%<br />

Stage I: 13%<br />

I: 5% 0.43<br />

II: 25% II: 27%<br />

III: 33% III: 45%<br />

IV: 29% IV: 22%<br />

Known asbestos exposure 54% 46% 1<br />

Smoking (former or current) 75% 42% 0.006<br />

Surgery<br />

EPP: 54% EPP: 55% 1<br />

EPP � extrapleural pneumonectomy Other: 38% Other: 35%<br />

None: 8% None: 10%<br />

Median overall survival from<br />

diagnosis (months)<br />

14.8 15.3 0.78<br />

Cancer Prevention/Epidemiology<br />

95s<br />

1540 General Poster Session (Board #1D), Sat, 1:15 PM-5:15 PM<br />

MEN1 gene mutations with different phenotypic presentations in two<br />

families: Is it a time to grade MEN1 mutations as high-risk and low-risk?<br />

Presenting Author: Alexander L. Shifrin, Jersey Shore University Medical<br />

Center, Neptune, NJ<br />

Background: MEN1 syndrome results from MEN1 gene mutations and is<br />

characterized by primary hyperparathyroidism (PHPT), pancreatic endocrine<br />

tumors (PET), pituitary adenomas, thymic and other tumors. About<br />

600 different mutations have been reported on Human Gene Mutation<br />

Database and 30 <strong>of</strong> them presented as Familial Isolated Hyperparathyroidism<br />

(FIHP); however, no genotype-phenotype correlations have been<br />

identified. Methods: We characterized 2 independent families with newly<br />

diagnosed MEN1. Mutation analyses were performed in Dept <strong>of</strong> Genetics<br />

Yale University. Diagnostic tests, surgical treatment were implemented.<br />

Results: Family 1 has 21 members. 5 presented with early onset <strong>of</strong> PHPT<br />

and no other tumors. 3 ½ parathyroidectomies were performed. A proband<br />

had direct sequencing that showed a mutation in Exon 7, consisting <strong>of</strong><br />

single base insertion in codon 319 (TAC to TTAC)(c1065_1066insT).<br />

There were no deaths in the family. Family 2 has 20 members. 7 were<br />

tested and showed a mutation in Exon 2, consisting <strong>of</strong> single base deletion<br />

in codon 103 (CTG to _TG)(c417delC). One 1st generation member died<br />

from PET at age 79, three 3rd generation members had multifocal<br />

metatstatic malignant gastrinomas (ages 32, 39, 41). The proband had<br />

total pancreatectomy at age 32. Five members had PHPT, angi<strong>of</strong>ibromas.<br />

Three 2nd generation members had thymic carcinomas (ages 20, 54, 60).<br />

Conclusions: Phenotypic presentation <strong>of</strong> MEN1 syndrome may vary with the<br />

same mutation suggesting a lack <strong>of</strong> genotype-phenotype correlation and<br />

the effect <strong>of</strong> modifying factors. On the other hand, these two families—one<br />

with a mutation predicted to truncate the menin protein close to its<br />

terminus and the other with a truncating mutation early in the coding<br />

sequence—have dramatically different severity <strong>of</strong> the syndrome. Our<br />

findings suggest that the exon 7 mutation has a milder effect on protein<br />

function resulting in FIHP. The management may be heavily influenced by<br />

the predicted clinical phenotype, we are proposing the development <strong>of</strong> a<br />

grading system for MEN1 gene mutations as low-risk and high-risk and to<br />

attempt to identify other modifying genetic and environmental factors to<br />

improve screening and timing for surgery.<br />

1542 General Poster Session (Board #1F), Sat, 1:15 PM-5:15 PM<br />

Genetic testing referral, uptake, and results in a program for women age 40<br />

or younger with breast cancer. Presenting Author: Asma Ali, Sunnybrook<br />

Health Sciences Centre, University <strong>of</strong> Toronto, Toronto, ON, Canada<br />

Background: PYNK: Breast Cancer Program for Young Woman is a novel<br />

program started in 2008 at our center to optimize management and<br />

promote research for women � age 40 newly diagnosed with breast cancer.<br />

<strong>Clinical</strong> and epidemiological data including cancer family history (FH) is<br />

prospectively collected on each consenting patient. As Toronto’s population<br />

is uniquely multiethnic we sought to determine BRCA testing eligibility,<br />

uptake and results for PYNK patients. Methods: Of the 145 consecutive<br />

patients, data were available for 109, <strong>of</strong> whom 2 had testing prior to<br />

diagnosis. Our provincial BRCA testing criteria are age � 35 at diagnosis;<br />

suspicious FH; or Ashkenazi Jewish (AJ) and age � 50 at diagnosis. Results:<br />

Of the 107 previously untested patients, 40 were � 35 at diagnosis. In 5 <strong>of</strong><br />

the other 67 testing eligibility could not be assessed. 66 <strong>of</strong> the 102 (65%)<br />

were eligible and <strong>of</strong> those 65 (98%) were <strong>of</strong>fered referral for counseling.<br />

One declined counseling, 9 were not yet seen, 2 declined testing, and 53<br />

were tested. Test results are available for 47 as follows: 30 (64%) no<br />

mutation, 4 (8%) variant <strong>of</strong> uncertain significance (VUS), 7(15%) BRCA1<br />

mutations and 6 (13%) BRCA2 mutations including 1 <strong>of</strong> the 4 AJ women.<br />

Ethnicity <strong>of</strong> the other 12 mutation carriers was: 1 Hispanic, 2 European, 2<br />

African, 4 Asians, 1 mix, 1 unknown and 1 not recorded. Two (15%) <strong>of</strong> the<br />

mutation carriers had no FH <strong>of</strong> breast or ovarian cancer. Four additional<br />

women opted for counseling and testing despite ineligibility and none had<br />

mutations. Conclusions: A specialized program for young women facilitates<br />

appropriate referral for genetic testing and encourages high testing uptake,<br />

which is important given the high prevalence <strong>of</strong> mutations (28% <strong>of</strong> tested<br />

women). Further research is necessary to assess the psychological and<br />

management impact <strong>of</strong> having a VUS on young women compared to their<br />

older counterparts.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


96s Cancer Prevention/Epidemiology<br />

1543 General Poster Session (Board #1G), Sat, 1:15 PM-5:15 PM<br />

Breast cancer in Irish families with Lynch syndrome. Presenting Author:<br />

Emmet Jordan, Mater Misericordiae University Hospital, Dublin, Ireland<br />

Background: Breast cancer is not among the recognised malignant manifestations<br />

<strong>of</strong> Lynch Syndrome which include colorectal, endometrial, gastric,<br />

ovarian and upper urinary tract tumours. In this study we report the<br />

prevalence <strong>of</strong> breast cancer in Irish Lynch Syndrome families and determine<br />

immunohistochemical (IHC) expression <strong>of</strong> mismatch repair proteins<br />

(MMR) in available breast cancer tissue. Methods: Breast cancer prevalence<br />

was determined among Lynch Syndrome kindreds from two institutions in<br />

Ireland. One kindred was omitted due to a biallelic MMR and BRCA1<br />

mutation.The clinicopathological data that was collected on breast cancer<br />

cases included age <strong>of</strong> onset, morphology, and hormone receptor status, and<br />

a genotype phenotype correlation was investigated. Immunohistochemical<br />

staining was performed for MLH1, MSH2, MSH6, and PMS2 on all<br />

available breast cancer tissue from affected individuals. Results: The<br />

distribution <strong>of</strong> MMR mutations seen in sixteen pedigrees were as follows;<br />

MLH1 (n�5), MSH2 (7), MSH6 (3), PMS2 (1). Sixty cases <strong>of</strong> colorectal<br />

cancer and 14 cases <strong>of</strong> endometrial cancer were seen. Seven breast<br />

cancers (5 invasive ductal and 2 invasive lobular cancers) and 1 case <strong>of</strong><br />

ductal carcinoma in situ were reported in 7 pedigrees. This compared with<br />

4 cases <strong>of</strong> prostate cancer. Of the 7 LS kindreds containing breast cancer,<br />

6 MSH2 mutations and 1 MSH6 mutations were identified. Median age <strong>of</strong><br />

breast cancer diagnosis was 49 years (range 38-57). Hormone receptor<br />

status is available on 3 breast cancer cases at time <strong>of</strong> abstract submission;<br />

all were ER positive and HER 2 negative. All cases had grade 2 or 3<br />

tumours. 5 samples were available for IHC evaluation. 3 out <strong>of</strong> 5 cases<br />

showed loss <strong>of</strong> MMR expression, all showed loss <strong>of</strong> MSH2 and MSH6<br />

expression. One <strong>of</strong> the two cases with normal IHC expression in breast<br />

tissue belonged to a kindred where 3 siblings with colorectal cancer and<br />

documented deleterious mutations demonstrated no IHC loss. Conclusions:<br />

Breast cancer occurred at an early age and was more common than prostate<br />

cancer in Irish Lynch Syndrome pedigrees. All identified breast cancer were<br />

in kindreds with MSH2 or MSH6 mutations. Enhanced breast cancer<br />

screening may be warranted in certain Lynch Syndrome kindreds.<br />

1545 General Poster Session (Board #2B), Sat, 1:15 PM-5:15 PM<br />

BRCA carrier status as an independent prognostic factor associated with<br />

earlier biochemical relapse in local prostate cancer. Presenting Author:<br />

Elena Castro, The Institute <strong>of</strong> Cancer Research and The Royal Marsden<br />

NHS Foundation Trust, London, United Kingdom<br />

Background: Biochemical relapse after local treatment for prostate cancer<br />

(PCa) indicates recurrent disease and is associated with shorter survival.<br />

Germline BRCA mutations are associated with worse PCa outcomes. BRCA<br />

carriers are currently treated with the same protocols used for non-carriers.<br />

We analyzed biochemical-progression free survival (bPFS) after conventional<br />

treatment for localized PCa in a cohort <strong>of</strong> BRCA patients (pts).<br />

Methods: In this retrospective case-control study, each BRCA carrier (9<br />

BRCA1 and 34 BRCA2) treated with radical prostatectomy (RP) or external<br />

beam radiotherapy (RT) was matched with 3 non-carriers (NC) by age at<br />

diagnosis (�5 yrs), TNM stage, Gleason score, presenting PSA, local<br />

treatment , androgen-deprivation therapy (ADT) and year <strong>of</strong> treatment (�3<br />

yrs). All NC were screened for BRCA1 and BRCA2 mutations. Biochemical<br />

failure was reviewed according to ASTRO and NCCN criteria. The Kaplan-<br />

Meier method and a multivariate Cox regression model adjusted by<br />

matching factors were employed. Results: 172 pts were included. Median<br />

follow-up was 76 months (ms). Median age at diagnosis was 58 yrs<br />

(43-75). Tumour stages were I 11%, IIA 19%, IIB 28%, III 28%, IV 14%.<br />

80 pts received RT (18 BRCA2, 5BRCA1, 57NC) and 85% also received<br />

ADT (70% for �6 months). 92 pts underwent RP (16 BRCA2, 4 BRCA1<br />

and 72 NC), and 9% <strong>of</strong> them received ADT (�6months). Overall, median<br />

bPFS was 71ms. For those treated with RT, median bPFS was 65ms in NC<br />

vs 39ms in BRCA carriers (p�0.023). bPFS was not affected by ADT<br />

duration. Median bPFS after RP was 65ms in BRCA carriers. No difference<br />

was observed in 3yrs-bPFS between BRCA carriers and NC (73% vs 76%).<br />

The adjusted MVA confirmed the independent prognostic value <strong>of</strong> tumour<br />

stage (p�0.004) and BRCA status (p�0.032) for bPFS. Among BRCA<br />

carriers, the risk was greater when the analysis was limited to BRCA2 pts<br />

(p�0.013, HR 2.1,.95%CI 1.2-3.7). Conclusions: Our results suggest that<br />

BRCA carriers with PCa have worse local disease control than NC when<br />

treated with RT, regardless <strong>of</strong> ADT duration. No differences in bPFS were<br />

observed in pts treated with RP after �6 yrs median follow-up. These<br />

results may have implications for tailoring clinical management for these<br />

patients.<br />

1544 General Poster Session (Board #2A), Sat, 1:15 PM-5:15 PM<br />

Cost sharing and hereditary cancer risk: Predictors <strong>of</strong> willingness-to-pay for<br />

genetic testing. Presenting Author: Jennifer Madeline Matro, Fox Chase<br />

Cancer Center, Philadelphia, PA<br />

Background: The increasing availability <strong>of</strong> genetic testing (GT) in cancer<br />

(CA) care has been paralleled by increasing cost-sharing practices by<br />

payors that are intended to reduce overuse <strong>of</strong> health care services. Patients<br />

referred for hereditary CA risk assessment may be subject to financial<br />

deterrents such as high out-<strong>of</strong>-pocket (OOP) costs. Little is known about<br />

what factors may influence high-risk patients’ willingness-to-pay (WTP) for<br />

GT. Methods: The Gastrointestinal (GI) Tumor Risk Assessment Registry<br />

includes individuals referred for evaluation <strong>of</strong> genetic CA risk based on<br />

personal and/or family hx. At enrollment, participants complete a survey<br />

collecting detailed demographic, CA hx, and psychosocial items related to<br />

CA risk. Baseline data on 406 participants were available; 21 who did not<br />

respond to WTP items were excluded. WTP items included intention for: GT<br />

only if paid by insurance; GT even if paid by self; and amount WTP (7 levels,<br />

$25 to $2000). Multivariable models examined predictors <strong>of</strong> WTP (self vs<br />

only if paid by insurance) (model 1, logistic), and predictors <strong>of</strong> amount WTP<br />

(model 2, ordinal logistic, 5 levels). All statistical tests are two-sided<br />

(��0.05). Results: Most participants were women (73%), white (92%) and<br />

aged 45-64 (58%). 51% had � 4 yr college degree; 22% had household<br />

income �$75,000; 42% had hx <strong>of</strong> GI CA; 56% had � 11st degree relative<br />

(FDR) with colorectal CA (CRC). Overall, 82 (21.3%) were willing to have<br />

GT only if paid by insurance, and 303 (78.7%) were WTP OOP. Of those<br />

WTP, 271 (89%) stated an amount. Independent predictors <strong>of</strong> WTP (model<br />

1) were: 1) expectation <strong>of</strong> positive result, 2) confidence to control CA risk,<br />

3) fewer perceived barriers to CRC screening and 4) benefit <strong>of</strong> guiding<br />

screening (all p�0.05). Subjects WTP a higher amount (model 2) were<br />

male, more educated, and had greater CA worry, fewer FDR with CRC (all<br />

p�0.05), and more positive attitudes toward GT (p�0.01). Conclusions:<br />

Patients who are WTP some OOP costs for GT anticipate benefits to control<br />

<strong>of</strong> CA risk afforded by GT. Women and less educated patients may face<br />

greater barriers from high co-pays. Identifying patient-level factors associated<br />

with WTP for genetic services is increasingly important as GT is<br />

integrated into routine CA care.<br />

1546 General Poster Session (Board #2C), Sat, 1:15 PM-5:15 PM<br />

Comparison <strong>of</strong> BRCA1/2-positive and -negative women diagnosed with<br />

metachronous breast and ovarian cancers. Presenting Author: Amanda C.<br />

Brandt, University <strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: After a primary breast cancer (BC) or ovarian cancer (OC)<br />

diagnosis, women have a risk to develop a second primary cancer,<br />

significantly more so in women with a BRCA gene mutation. The objective<br />

was to evaluate characteristics <strong>of</strong> BRCA positive and negative women<br />

diagnosed with metachronous BC and OC. Methods: Women with metachronous<br />

BC and OC were identified at the University <strong>of</strong> Texas MD Anderson<br />

Cancer Center. BRCA test results, age at diagnosis <strong>of</strong> both BC and OC, first<br />

degree relatives (FDR) with BC and/or OC, total relatives affected, and<br />

histopathologic tumor features were obtained from a prospectively maintained<br />

research database. Univariate and logistical regression analyses<br />

were performed. Results: A total <strong>of</strong> 64 women with metachronous primary<br />

cancers underwent BRCA genetic testing. Of the 45 BRCA positive women,<br />

36 (80%) were diagnosed with BC (mean 40 years) prior to OC and 9 (20%)<br />

were diagnosed with OC first (mean 47 years). Of the 19 BRCA negative<br />

women, 10 (53%) were diagnosed with BC first (mean 63 years), and 9<br />

(47%) diagnosed with OC first (mean 48 years). Women diagnosed with BC<br />

first were more likely to be BRCA positive than women diagnosed with OC<br />

first (p�0.03). Patients with a FDR with BC were more likely BRCA positive<br />

than women without family history (p�0.0062). Patients with PR negative<br />

BR (p�0.0199) or triple negative (TN) BC (p�0.0139) were more likely to<br />

test BRCA positive. High grade ovarian carcinomas approached significance<br />

(p�0.053) in BRCA positive when compared to BRCA negative<br />

cohort. Ethnicity, total number <strong>of</strong> affected relatives with BC or OC, ER<br />

status, Her2 status, and nuclear grade were not statistically significant<br />

between the two groups. Conclusions: Women with OC preceding their BC<br />

with lack <strong>of</strong> family history <strong>of</strong> BC and OC are significantly less likely to test<br />

BRCA positive. BC is <strong>of</strong>ten the sentinel cancer in women with a BRCA<br />

mutation compared to BRCA negative women with metachronous BC and<br />

OC. This should be considered during the risk assessment to more<br />

accurately estimate the chance <strong>of</strong> identifying a BRCA mutation in women<br />

with metachronous BC and OC.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


1547 General Poster Session (Board #2D), Sat, 1:15 PM-5:15 PM<br />

Features <strong>of</strong> familiality in a cohort <strong>of</strong> patients (pts) with sarcoma. Presenting<br />

Author: Georgios Lypas, Dana-Farber Cancer Institute, Boston, MA<br />

Background: Sarcomas occur in a number <strong>of</strong> cancer predisposition syndromes,<br />

yet genetic evaluation is rarely recommended for sarcoma pts. We<br />

broadly reviewed the family cancer histories <strong>of</strong> unselected pts in a tertiary<br />

center sarcoma clinic to explore the potential relevance <strong>of</strong> genetic assessment.<br />

Methods: Between 2005 and 2008, 397 pts (median age 52 years)<br />

with documented sarcoma who consented to research use <strong>of</strong> medical<br />

records and tissues also completed a family history questionnaire. Patient<br />

and family histories were reviewed for features suggesting hereditary risk<br />

(e.g. early age at diagnosis or familial clustering <strong>of</strong> certain associated<br />

diagnoses). Results: A family history <strong>of</strong> malignancy was noted in 301 <strong>of</strong> 397<br />

pts (76%). Seventy-six pts (19%) were diagnosed with 99 additional<br />

malignancies, most frequently breast cancer (n�23), prostate cancer (12),<br />

melanoma (12), thyroid cancer (6) or a second sarcoma (10). Sarcoma was<br />

diagnosed prior to or synchronously (within 6 months) with another<br />

malignancy in 20 pts and metachronously in 56 pts. In 18 pts, sarcoma<br />

arose within a prior radiation field. Thirty-seven pts (9%) had a sarcoma<br />

subtype associated with a recognized syndrome: desmoid fibromatosis<br />

(n�16), malignant peripheral nerve sheath tumor (11), PEComa/<br />

angiomyolipoma (6), retinoblastoma (3), and GIST with paraganglioma (1).<br />

Based on additional features, the associated syndromes were confirmed in<br />

6 <strong>of</strong> them (1.5% <strong>of</strong> the cohort): familial adenomatous polyposis (n�1),<br />

neur<strong>of</strong>ibromatosis type 1 (2), familial retinoblastoma (2), and Carney-<br />

Stratakis dyad (1). Thirty-three pts (8% <strong>of</strong> the cohort, or 16% <strong>of</strong> pts age<br />

�50) fulfilled TP53 testing criteria (Chompret, Birch or classical Li-<br />

Fraumeni Syndrome [LFS] criteria). Another 147 pts (37% <strong>of</strong> the entire<br />

cohort) did not fulfill LFS testing criteria, but were diagnosed with sarcoma<br />

before age 50 and had at least one first or second degree relative with a<br />

diagnosis <strong>of</strong> malignancy before age 50. Conclusions: Sarcomas are rare and<br />

diverse cancers. Given the potential association with cancer predisposition<br />

syndromes, genetic evaluation should be strongly considered in sarcoma<br />

pts based on age at diagnosis, histology, and family cancer history.<br />

1549 General Poster Session (Board #2F), Sat, 1:15 PM-5:15 PM<br />

Predictors <strong>of</strong> contralateral prophylactic mastectomy among patients with<br />

ductal carcinoma in situ (DCIS) who underwent evaluation for BRCA<br />

genetic testing. Presenting Author: Nisreen Elsayegh, University <strong>of</strong> Texas<br />

M. D. Anderson Cancer Center, Houston, TX<br />

Background: Patients with DCIS are at increased risk for developing<br />

contralateral breast cancer (CBC). Therefore, an increasing number <strong>of</strong><br />

women with DCIS are electing for contralateral prophylactic mastectomy<br />

(CPM). In a previous study involving 2072 women with DCIS, 13.5% chose<br />

CPM. In this study, we aimed to evaluate factors associated with CPM in<br />

patients with DCIS who underwent genetic counseling for BRCA. Methods:<br />

165 women with pure DCIS, who underwent genetic counseling, were<br />

included in the study. Patients’ characteristics were obtained from a<br />

prospectively maintained research database at UT M.D. Anderson Cancer<br />

Center. Univariate and multivaraite logistic regression analysis were used to<br />

determine predictive factors associated with CPM. Patients’ characteristics<br />

included age, marital and educational status, tumor markers, nuclear<br />

grade, family history with breast (BC) and ovarian cancer (OC), race,<br />

Ashkenazi Jewish ancestry, and BRCA genetic test results. Results: Out <strong>of</strong><br />

165 patients, 17(10.3%) were found to have a BRCA deleterious mutation.<br />

44(26.7%) underwent CPM. Younger patients (median � 45 yr) were more<br />

likely to elect for CPM than older patients (p� 0.0098). Patients who<br />

tested positive for a BRCA mutation were more likely to elect for CPM than<br />

those who tested negative or were not tested (p� 0.0001). Patients who<br />

had a family history <strong>of</strong> OC (15 (57.7%) were more likely to choose CPM<br />

than those who did not (p� 0.0004). These three factors remained<br />

significant in the multivariate model (p �0.008). Marital and educational<br />

status, tumor markers, nuclear grade, and family history <strong>of</strong> breast cancer<br />

were not significant predictors <strong>of</strong> CPM. Conclusions: The rate <strong>of</strong> CPM in<br />

patients with DCIS is high. Factors associated with increased likelihood <strong>of</strong><br />

undergoing CPM include family history <strong>of</strong> OC, age, and BRCA positivity.<br />

Further studies are needed to evaluate patients perception <strong>of</strong> CBC risk, and<br />

if this may play a role in the high number <strong>of</strong> CPM.<br />

Cancer Prevention/Epidemiology<br />

97s<br />

1548 General Poster Session (Board #2E), Sat, 1:15 PM-5:15 PM<br />

Analysis <strong>of</strong> mutations in formalin-fixed paraffin-embedded (FFPE) tumor<br />

samples from two phase II clinical trials using a Sequenom-based approach.<br />

Presenting Author: Maria Orr, AstraZeneca, Macclesfield, United<br />

Kingdom<br />

Background: Analysis <strong>of</strong> tumour samples for biomarker discovery, personalised<br />

health care and disease stratification is becoming increasingly<br />

important. Methods to detect somatic mutations are <strong>of</strong>ten limited by the<br />

requirement for a significant amount <strong>of</strong> input DNA, low sensitivity or low<br />

sample throughput. The aim <strong>of</strong> this work was to obtain a comprehensive<br />

mutation pr<strong>of</strong>ile <strong>of</strong> tumour samples taken from patients enrolled on two<br />

Phase II clinical trials. A Sequenom based approach was used to determine<br />

mutation pr<strong>of</strong>iles for the samples so that these could be correlated with<br />

response at some future date. Methods: After successfully piloting the<br />

Sequenom MassArray 4 platform on formalin fixed paraffin embedded<br />

(FFPE) tumour samples and tumour cell line admixes, we ran an exploratory<br />

analysis <strong>of</strong> 177 melanoma and 230 non small cell lung cancer (NSCLC)<br />

FFPE samples collected from two phase II clinical trials. DNA was extracted<br />

from the FFPE samples using the QIAmp DNA FFPE Tissue Kit (Qiagen).<br />

We designed a panel <strong>of</strong> 86 assays in 14 genes covering approximately<br />

160� mutations. Mutations were chosen based on mutation frequency and<br />

biological significance in these tumour types. Results: Using this approach<br />

we were able to detect concomitant mutation pr<strong>of</strong>iles in a significant<br />

percentage <strong>of</strong> samples using only approximately 20uL <strong>of</strong> FFPE DNA (500<br />

copies/�L average). Mutation status concordance was over 97% in BRAF<br />

and KRAS when compared to data generated via other approaches<br />

(sequencing, ARMS). Conclusions: Using Sequenom to screen tumour<br />

material, collected during clinical trials, is a feasible, rapid and comprehensive<br />

approach. Such an approach can be used to identify mutation pr<strong>of</strong>iles<br />

which correlate with patient response, potentially identify further patient<br />

stratification and help generate new hypotheses for further exploration.<br />

1550 General Poster Session (Board #2G), Sat, 1:15 PM-5:15 PM<br />

Pr<strong>of</strong>iling uptake <strong>of</strong> single site testing (SST) for familial cancer (CA) risk in an<br />

administrative genetic testing database. Presenting Author: Michael J. Hall, Fox<br />

Chase Cancer Center, Philadelphia, PA<br />

Background: Among the anticipated social and cost-saving benefits to identifying<br />

a mutation in a CA predisposition gene in an individual is the subsequent<br />

opportunity to perform SST in close relatives to confirm risk status and to guide<br />

risk-reducing interventions. Despite ample research into uptake <strong>of</strong> primary<br />

genetic testing for Lynch syndrome (LS) and hereditary breast ovarian syndrome<br />

(HBOS) among moderate- and high-risk individuals, few studies have focused on<br />

the resultant cascade <strong>of</strong> SST that is predicted to follow in families. Methods:<br />

Administrative data on 2,631 consecutive single-site mutation tests performed<br />

over a 3-month interval (12/1/10-2/28/11) were obtained from a commercial<br />

genetic testing database maintained by Myriad Genetic Laboratories. 14<br />

subjects (29 tests) were excluded due to multiple entries. Demographic,<br />

personal/family history (hx), and provider data are collected on a Test Requisition<br />

Form included in kits. All statistical tests are 2-sided (��0.05). Results: Women<br />

had SST more <strong>of</strong>ten than men (HBOS 82.8%; LS 65.5%). Mean age at time <strong>of</strong><br />

SST was 42.7 yrs, and was higher for HBOS than LS (p�0.01). Among subjects<br />

with hx <strong>of</strong> CA, mean age at CA diagnosis (dx) was similar (HBOS 48.6; LS 48.7<br />

yrs). Subjects reported CA in �1 relatives (range 1-16) with a mean age at dx<br />

40.1 yrs. Compared to HBOS, those tested for LS were more likely to report CA in<br />

a 1st degree relative (81.3 vs 74.4%, p�0.001) and a lower age <strong>of</strong> youngest CA<br />

dx in the family (p�0.001). Non-European ancestry was more common for<br />

HBOS SST than LS (30.0 vs 24.8%) (p�0.05). SST positive test rate was no<br />

different by syndrome or ancestry, but was higher for men (47.5 vs 42.0%,<br />

p�0.02). Conclusions: Uptake <strong>of</strong> commercial SST in high-risk families is uneven<br />

by age, sex, ancestry and CA syndrome. While between-syndrome variability in<br />

CA risks may explain some differences, more research is needed to understand<br />

barriers to uptake <strong>of</strong> SST among at-risk individuals if CA prevention goals are to<br />

be realized.<br />

Characteristics <strong>of</strong> 2,602 subjects undergoing SST for familial CA risk.<br />

%<br />

Sex F 79.5<br />

M 20.5<br />

Cancer Hx Y 19.9<br />

N 71.6<br />

Unreported 8.5<br />

Ancestry European 70.5<br />

Other 29.5<br />

Syndrome (genes) HBOS (BRCA1/2) 82.7<br />

LS (MLH1/MSH2/MSH6) 13.1<br />

Other 4.2<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


98s Cancer Prevention/Epidemiology<br />

1551 General Poster Session (Board #3A), Sat, 1:15 PM-5:15 PM<br />

NAD(P)H: Quinone oxidoreductase 1 (NQO1) C609T polymorphism and<br />

lung cancer risk: A meta-analysis. Presenting Author: Yuqing Lou, Department<br />

<strong>of</strong> Pulmonary, Shanghai Chest Hospital, Jiaotong University, Shanghai,<br />

PR China, Shanghai, China<br />

Background: No clear consensus has been reached on the NAD(P)H:<br />

quinone oxidoreductase 1 (NQO1) gene C609T polymorphism and lung<br />

cancer risk. We performed a meta-analysis to summarize the possible<br />

association. Methods: We conducted a computer retrieval <strong>of</strong> PUBMED and<br />

EMBASE databases prior to January 2012. References <strong>of</strong> retrieved articles<br />

were also screened. According to the inclusion criteria, 24 articles (31<br />

studies) were finally included. The fixed-effects model and the randomeffects<br />

model were applied for dichotomous outcomes to combine the<br />

results <strong>of</strong> the individual studies. Results: There was no statistical association<br />

between C609T polymorphism and lung cancer risk in overall, East<br />

Asians, African-<strong>American</strong>s or Hispanics. In Caucasians, significant association<br />

was found in allele comparison model (T vs. C) (P�0.04, OR�1.09,<br />

95%CI [1.00–1.19], Pheterogeneity�0.24, fixed-effects model). In Hawaiians,<br />

significant association could be found in dominant genetic model<br />

((TT�CT) vs. CC) (P�0.04, OR�0.52, 95%CI [0.28–0.96], fixed-effects<br />

model). In the subgroup <strong>of</strong> squamous cell carcinoma, a borderline<br />

significance could be found in the dominant genetic model ((TT�CT) vs.<br />

CC) (P�0.05, OR�1.20, 95%CI [1.00–1.43], Pheterogeneity�0.65, fixed-effects model). Significant association could also be found in<br />

allele comparison (T vs. C) (P�0.03, OR�1.21, 95%CI [1.01–1.44],<br />

Pheterogeneity�0.68, fixed-effects model). In the subgroup <strong>of</strong> small cell lung<br />

cancer risk, significant association were found in allele comparison (T vs. C)<br />

(P�0.03, OR�1.68, 95%CI [1.05–2.68], Pheterogeneity�0.10, randomeffects<br />

model) and in the homozygote comparison (TT vs. CC) (P�0.02,<br />

OR�2.79, 95%CI[1.14–6.85], Pheterogeneity�0.72, fixed-effects model).<br />

No association was observed in adenocarcinoma subgroup. Conclusions:<br />

NQO1 C609T polymorphism might associate with lung cancer risk in<br />

Caucasians and Hawaiians. NQO1 C609T polymorphism might also associate<br />

with squamous cell carcinoma and small cell lung cancer risk.<br />

1553 General Poster Session (Board #3C), Sat, 1:15 PM-5:15 PM<br />

Why do breast cancer programs fail to refer patients to genetic counseling<br />

upon obtaining family history? Presenting Author: Julia R. Trosman, Center<br />

for Business Models in Healthcare, Chicago, IL<br />

Background: Women with personal or family history suggestive <strong>of</strong> susceptibility<br />

to hereditary breast or ovarian cancer should be referred to genetic<br />

counseling (US Preventive Services Task Force, Ann Intern Med. 2005).<br />

Our goal is to examine whether this guideline is followed by breast cancer<br />

programs providing screening and treatment in a large urban area; and if<br />

not followed, why. Funded by Susan G. Komen for the Cure. Methods: Using<br />

the framework approach <strong>of</strong> qualitative research, we conducted interviews<br />

with 130 providers <strong>of</strong> breast cancer screening and treatment at 26<br />

institutions in Chicago (18 community, 4 academic and 4 public hospitals).<br />

We interviewed radiologists, mammography technologists, nurses,<br />

surgeons, oncologists, internists, and patient navigators. Interviews were<br />

transcribed and coded; theme analysis was conducted; simple frequencies<br />

and Fisher’s exact test were calculated. Results: While all 26 programs<br />

collect patient personal and family history, only one program has both a<br />

protocol for referral to genetic counseling and a genetic counseling service.<br />

All six interviewees from that program (6/130, 5%) report referring<br />

appropriate patients to genetic counseling, compared to none from other<br />

programs (p�0.0001). 90% <strong>of</strong> interviewees (118/130) did not perceive<br />

any role in raising patient awareness or referring them to genetic counseling.<br />

Among the 124 interviewees not referring to genetic counseling, 51<br />

(41%) have genetic counseling available, but only 12% (6/51) <strong>of</strong> them view<br />

referring or making patients aware <strong>of</strong> genetic services as their responsibility;<br />

while none <strong>of</strong> the interviewees without genetic counseling services view<br />

this their responsibility (p�0.0001). None <strong>of</strong> the interviewees noted<br />

reimbursement <strong>of</strong> genetic counseling as a barrier to referral. Conclusions:<br />

The lack <strong>of</strong> accountability by care providers is a barrier to referring patients<br />

with personal or family history <strong>of</strong> breast or ovarian cancer to genetic<br />

counseling, even when the service is available. A comprehensive approach<br />

addressing access to genetic counseling, adoption <strong>of</strong> referral protocols and<br />

clear assignment <strong>of</strong> referral responsibilities is needed to ensure that women<br />

appropriately receive genetic assessment.<br />

1552 General Poster Session (Board #3B), Sat, 1:15 PM-5:15 PM<br />

Ethnic difference <strong>of</strong> driver mutation frequencies and correlations between<br />

driver mutations and histologic subtypes in lung adenocarcinoma. Presenting<br />

Author: Yuki Yamane, Division <strong>of</strong> Thoracic Oncology, National Cancer<br />

Center Hospital East, Kashiwa, Chiba, Japan<br />

Background: The frequencies <strong>of</strong> known driver mutation in lung adenocarcinoma<br />

from patients in the United States have been reported by the NCI’s<br />

Lung Cancer Mutation Consortium (LCMC), indicating driver mutations<br />

were detected in 54% (280/516) <strong>of</strong> tumors. In this report, mutations<br />

found: EGFR 17%, KRAS 22%, HER2 0.6%, PIK3CA 1.2%, BRAF 2%,<br />

MET amplification 0.6%, MAP2K1 0.4%, NRAS 0.4%, AKT 0%, ALK<br />

rearrangements 7%. However little is known about ethnic difference <strong>of</strong><br />

driver mutation frequencies and correlations between driver mutations and<br />

histological subtypes in lung adenocarcinoma. Methods: Known driver<br />

mutations in tumors from 97 Japanese patients with lung adenocarcinoma<br />

who underwent surgical resection between 1999 and 2003 in National<br />

Cancer Center Hospital East were analyzed by next-generation sequencing<br />

and confirmed by Sanger sequencing. Correlations between driver mutations<br />

and histological subtypes were also assessed. Results: Driver mutations<br />

were detected in 72% <strong>of</strong> tumors. Mutations found: EGFR 57%,<br />

KRAS9%, HER2 2%, PIK3CA 2%, BRAF 1%, MET amplification 1%,<br />

MAP2K1 0%, NRAS 0%, AKT 0%. Due to the limitation <strong>of</strong> rearrangement<br />

detection by exon-sequencing, ALK rearrangements were not analyzed.<br />

Compared with the report by LCMC, the frequency <strong>of</strong> EGFR mutations was<br />

high and that <strong>of</strong> KRAS mutations was low in the present study. All<br />

mutations were mutually exclusive. The number <strong>of</strong> predominant histological<br />

subtypes <strong>of</strong> tumors harbored EGFR mutations were papillary 28, acinar<br />

3, solid 5, lepidic 19. That with KRAS mutations showed papillary 2, acinar<br />

2, solid 2, lepidic 3, and HER2 mutations showed papillary 1 and acinar 1.<br />

Two tumors harbored PIK3CA mutations showed both histological acinar<br />

pattern. Each <strong>of</strong> BRAF mutation and MET amplification showed lepidic and<br />

papillary pattern, respectively. Conclusions: It was suggested that there<br />

should be ethnic difference <strong>of</strong> driver mutation frequencies in lung adenocarcinoma<br />

between Asian and non-Asian patients, although the details <strong>of</strong><br />

ethnic distribution included in LCMC study has not been opened. In<br />

addition, each driver mutations did not correspond to specific histological<br />

subtypes <strong>of</strong> lung adenocarcinoma.<br />

1554 General Poster Session (Board #3D), Sat, 1:15 PM-5:15 PM<br />

Breast cancer prognosis in BRCA1/2 mutation carriers: A case control<br />

study. Presenting Author: Grazia Arpino, Medical Oncology, Endocrinology<br />

and Oncology Department, University Federico II <strong>of</strong> Napoli, Napoli, Italy<br />

Background: We evaluated the clinical impact <strong>of</strong> germ-line BRCA1/2<br />

mutations and variants <strong>of</strong> unknown clinical significance (VUS) for BRCA1/2<br />

in patients (pts) with early breast cancer (BC). Methods: Twenty-eight<br />

BRCA-positive (BRCA�) BC pts with germ-line BRCA1 /2 mutations and<br />

16 VUS BC pts were selected from our database and matched (1:3) with<br />

154 nonhereditary BC controls (sporadic controls, SC, defined by no<br />

associated personal history <strong>of</strong> breast cancer and no family history <strong>of</strong> breast<br />

and ovarian cancer or an uninformative BRCA mutation test) for stage,<br />

histologic subtype, age, and year <strong>of</strong> diagnosis. <strong>Clinical</strong> characteristics,<br />

recurrence (rec) pattern, disease free survival (DFS) and overall survival<br />

(OS) were analyzed. Results: Compared with VUS and SC, BRCA� pts were<br />

less likely to express estrogen receptor (64% vs. 89% vs. 54% respectively<br />

p�.005) and progesterone receptor (64% vs. 86% vs. 59% respectively<br />

p�.005) but more likely to be triple negative (0 vs. 3.4% vs 47.4%<br />

respectively p�.005). Compared with VUS and SC, BRCA� pts were more<br />

likely treated by radical mastectomy (37.5% vs. 26.4% vs. 59.3% %<br />

respectively p�.005). Pattern <strong>of</strong> rec was also different. Compared with<br />

VUS and SC, BRAC� pts developed more second tumors (11% vs. 6.3% vs.<br />

1.9% respectively p�.0001) but less local or distant rec (31% vs. 2.6 vs.<br />

0% for local rec and 12% vs. 16% vs. 11% for distant rec respectively<br />

p�.0001). Controlateral BC was more frequent in VUS compared to the<br />

BRAC� and SC pts (12% vs. 7% vs. 1% respectively, p�.0001). At a<br />

median follow up <strong>of</strong> 88 months, at univariate analysis, BRAC� but not VUS<br />

pts had worse OS compared to SC (p�.006). No difference in DFS was<br />

observed for VUS or BRAC� when compared to SC pts. After adjustment for<br />

age, stage, grade, nodal status, hormone receptors, adjuvant therapy and<br />

year <strong>of</strong> diagnosis, BRCA� pts continued to have and increased risk <strong>of</strong> death<br />

compared to SC (HR 5.9, 95% CI 1.9-18.1, p�.002). Most <strong>of</strong> the deaths<br />

observed in BRAC� pts were not cancer related. Conclusions: Despite<br />

decrease incidence <strong>of</strong> local or distant recs, BRAC� pts seems to be more<br />

likely to die compared to SC. Development <strong>of</strong> second cancers and unknown<br />

effects <strong>of</strong> BRCA1/2 mutations on nonneoplastic diseases that cause death<br />

may account for this findings.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


1555 General Poster Session (Board #3E), Sat, 1:15 PM-5:15 PM<br />

Variants <strong>of</strong> uncertain significance in BRCA testing: Impact on risk perception,<br />

worry, prevention, and counseling. Presenting Author: Tracy Graham,<br />

Sunnybrook Hospital, Toronto, ON, Canada<br />

Background: Genetic testing for BRCA1 and BRCA2 is an instrumental tool<br />

in clinical decision-making. Variants <strong>of</strong> uncertain significance (VUS) can<br />

occur in up to 25% <strong>of</strong> individuals and pose clinical challenges. We<br />

compared i) result recall ii) risk perception iii) worry and iv) risk modifying<br />

behaviours in individuals with VUS, a pathogenic mutation (PM) or an<br />

uninformative result (UR). Care provider attitudes to VUS were also<br />

studied. Methods: A questionnaire was mailed to women testing positive for<br />

a VUS, PM or UR. Items included demographics, time from disclosure,<br />

result recall and cancer worry using the validated Trask score. Likelihood <strong>of</strong><br />

risk modifying behaviours or intensified screening was evaluated by 5 point<br />

Likert scale. 9 regional genetic counselors and 11 referring physicians were<br />

surveyed on VUS management. Responses were evaluated by chi square<br />

and ANOVA. Results: All groups had similar age, marital status and<br />

education. Failure to correctly identify a result as uninformative occurred in<br />

32% <strong>of</strong> VUS; 90% had low economic status. 87% (VUS) and 44% (PMC)<br />

had personal history <strong>of</strong> breast cancer. Perceived risk increased after<br />

disclosure in 29% (VUS), 70%(PM) and 10% (UR) (p�0.001). Mean<br />

Trask scores were 7.6 (VUS) and 9.8 (PMC) (p�0.006, t-test). In affected<br />

patients, mastectomy was 22% (VUS) and 55% (PMC) (p�0.01), oophorectomy<br />

39% (VUS) and 85% (PMCs) (p�0.001) with75% and 95%<br />

intensified screening 75% in unaffected VUS and PM (p�0.16). 3% (VUS)<br />

and 6% (PMC) agreed with chemoprophylaxis (p�NS). Genetic counselors<br />

unanimously reported low comprehension in VUS vs PM. VUS was<br />

disclosed only 75% <strong>of</strong> the time with poor comprehension, perceived anxiety<br />

and negative family history as reasons. Referring physicians recommended<br />

predictive testing for relatives <strong>of</strong> VUS carriers (100%). Conclusions: Nearly<br />

1/3 <strong>of</strong> patients fail to recall their VUS as uninformative. Perceived risk was<br />

increased after VUS disclosure but lower than PMC. Worry scores showed<br />

impact on daily functioning. Uptake <strong>of</strong> prophylactic surgery was lower in<br />

affected VUS vs PMC carriers with similar rates <strong>of</strong> intensified screening if<br />

unaffected. Patients with low economic status or anxiety are at particular<br />

risk <strong>of</strong> incomplete counseling.<br />

1557 General Poster Session (Board #3G), Sat, 1:15 PM-5:15 PM<br />

A comparison <strong>of</strong> simple single-item measures and the NCI Common<br />

Toxicity Criteria version 3.0 measure <strong>of</strong> peripheral neuropathy. Presenting<br />

Author: Suneetha Puttabasavaiah, Mayo Clinic, Rochester, MN<br />

Background: Peripheral neuropathy (PN), a common and intrusive side<br />

effect <strong>of</strong> chemotherapy in clinical trials, has been assessed via the<br />

clinician-rated common terminology criteria for adverse events (CTCAE)<br />

and/or patient-reported outcome (PRO) measures (Morton, ASCO, 2005).<br />

We assessed the relative sensitivity <strong>of</strong> CTCAE and PRO measures for<br />

evaluating interventions for preventing and ameliorating CIPN. Methods:<br />

Data from285 patients in two prevention trials (N05C3, N04C7) and 432<br />

patients from three intervention trials (N00C3, N01C3, N06CA) were<br />

analyzed separately. CTCAE version 3.0 item for neuropathy was compared<br />

to 2 NCCTG numerical analogue CIPN items, 6 EORTC CIPN20 individual<br />

items, 2 McGill Pain Questionnaire items, and one Brief Pain Inventory<br />

item. The sample size provided over 80% power to detect a true Spearman<br />

rank correlation <strong>of</strong> 0.15 assessed the relationship between CTCAE and PRO<br />

measures. Results: Results from both the prevention and treatment trials<br />

were similar in terms <strong>of</strong> the relationship between the CTCAE and PRO<br />

measures. No correlation coefficients between the CTCAE and any <strong>of</strong> the<br />

PRO items at baseline were above 0.35, with the majority around 0.2.<br />

Higher scores for the numbness items were observed than for the CTCAE in<br />

47% <strong>of</strong> the patients at baseline and 39% during treatment in the treatment<br />

trials. Many patients had severe numbness PRO scores but mild CTCAE<br />

scores (12%) at baseline and (8%) during treatment in the treatment trials.<br />

Results for the numbness and tingling PRO measures were redundant. The<br />

correlations <strong>of</strong> the CTCAE with pain were weak, all below 0.35 at baseline<br />

with differences as much as 29 points on average on a 0-100 transformed<br />

scale. Conclusions: The CTCAE and PRO measures <strong>of</strong> PN measure different<br />

constructs. The CTCAE includes both functional and ADL/motor aspects<br />

which may or may not be related to the singular constructs <strong>of</strong> pain,<br />

numbness, tingling, or adjectival characteristics <strong>of</strong> pain assessed by the<br />

PRO measures. The two measures cannot be used in place <strong>of</strong> each other.<br />

Cancer Prevention/Epidemiology<br />

99s<br />

1556 General Poster Session (Board #3F), Sat, 1:15 PM-5:15 PM<br />

Differential effects <strong>of</strong> light at night and shift rotation pattern on the<br />

circadian system in night workers. Presenting Author: Pedram Razavi,<br />

Department <strong>of</strong> Medicine, University <strong>of</strong> Southern California, Los Angeles,<br />

CA<br />

Background: Light at night as in shift work suppresses nocturnal secretion<br />

<strong>of</strong> melatonin, a pineal hormone with oncostatic properties. Several studies<br />

have associated night shift work with higher risk <strong>of</strong> cancer, leading WHO in<br />

2007 to classify rotating night shift work as “probably carcinogenic”. We<br />

conducted one <strong>of</strong> the most comprehensive studies, to date, to evaluate the<br />

effects <strong>of</strong> light and night shift work on melatonin measurements in the<br />

field. Methods: Study participants were 130 active nurses (84 current<br />

rotating night shift workers and 46 day shift workers) participating in<br />

NHS2. Each nurse wore a head-mounted light- and accelerometer for a<br />

3-day study period, during which each spontaneous urine was collected for<br />

repeated urinary 6-sulfatoxymelatonin (melatonin) measurements. In addition,<br />

nurses were asked to fill out paper questionnaires and diaries. We<br />

used mixed models to evaluate the influence <strong>of</strong> light, activity and night shift<br />

work on urinary melatonin level adjusting, for age, lifestyle, and occupational<br />

history. We log-transformed main variables and report geometric<br />

means (GM [standard deviation]). Results: Greater levels <strong>of</strong> light were<br />

associated with lower melatonin (P � 0.0001), independent <strong>of</strong> activity<br />

level. An increase in light intensity from 10 to 100 lux was associated with<br />

a 12% decrease in geometric mean <strong>of</strong> melatonin level; however, this<br />

inverse association was only significant at night (Ptrend � 0.01). At night,<br />

each hour increase in exposure to � 20 lux light lowered melatonin level by<br />

5.7% (Ptrend � 0.0001). A single night shift affected the circadian system<br />

by lowering melatonin peak by 22% (day shift: GM � 17.57 [2.73]; night<br />

shift: GM � 13.64 [2.54]) and induced a phase shift (PS) <strong>of</strong> 0.9 hours,<br />

-changes that reset to normal by the next day. Two consecutive night shifts<br />

had a similar effect as a single shift. However, the effect was worse after<br />

three consecutive night shifts (GM � 10.11 [2.77]; PS � 2.2 hours).<br />

Conclusions: We found significant inverse associations <strong>of</strong> intensity and<br />

duration <strong>of</strong> exposure to light at night with urinary melatonin, independent<br />

<strong>of</strong> activity level. Three consecutive night shifts affected the circadian<br />

system more strongly than two consecutive, or a single night shift.<br />

1558 General Poster Session (Board #3H), Sat, 1:15 PM-5:15 PM<br />

Association <strong>of</strong> CYP19A1 gene variants with differences in disease progression<br />

in breast cancer patients from different racial/ethnic backgrounds.<br />

Presenting Author: Reina Villareal, University <strong>of</strong> New Mexico and New<br />

Mexico VA Health Care System, Albuquerque, NM<br />

Background: Polymorphisms in the aromatase (CYP19A1) gene result in<br />

differences in the risk for breast cancer and response to treatment. We<br />

hypothesize that allele frequencies in the CYP19A1 gene vary according to<br />

race and may result in differences in progression <strong>of</strong> breast cancer among<br />

women from different racial backgrounds. The objectives <strong>of</strong> this study are:<br />

1) to determine the allele/genotype frequencies in the CYP19A1 gene<br />

among women with breast cancer from different racial backgrounds, and,<br />

2) to determine the association between disease progression and CYP19A1<br />

gene variants. Methods: <strong>Clinical</strong> data and stored DNA from 327 patients<br />

participating in the Expanded Breast Cancer Registry (EBCR) program at<br />

the University <strong>of</strong> New Mexico were analyzed. These patients were followed-up<br />

for a period <strong>of</strong> 1 to 6 years. Comprehensive genotyping for<br />

CYP19A1 gene single nucleotide polymorphisms (SNPs) was performed<br />

using microarray (Illumina). Results: Data from 164 non-Hispanic white<br />

and 119 Hispanic women were analyzed. Four SNPs (rs1259269,<br />

rs17703883, rs16964211, rs28757101) were associated with differences<br />

in genotype/allele frequencies between the 2 racial groups. Furthermore,<br />

3 SNPs (rs4646, rs17647478, and rs6493486) were associated<br />

with differences in disease progression. The rare allele (G) for the<br />

rs17647478 (G/T) is associated with poor progression compared those<br />

without the allele (41.7% vs. 17.1%, p�0.04). Similarly the minor (A)<br />

allele for the rs4646 (A/C) and the rs6493486 (A/G) is also associated with<br />

a greater chance <strong>of</strong> worsening disease (23.2% vs. 12.4%, p�0.02 and<br />

23.4% vs. 12.4%, p�0.02, respectively). A significant percentage <strong>of</strong><br />

women carrying the A allele for both rs4646 and rs6493486 and the T<br />

allele for the rs17647478 have more advanced disease at the time <strong>of</strong><br />

presentation. None <strong>of</strong> the SNPs analyzed result in racial differences in<br />

breast cancer progression. Conclusions: Polymorphisms in the CYP19A1<br />

gene influence overall disease progression in breast cancer patients but<br />

have no impact on the racial differences <strong>of</strong> disease behavior. Women<br />

carrying the risk alleles present at a more advanced stage and are also at<br />

greater risk <strong>of</strong> progression.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


100s Cancer Prevention/Epidemiology<br />

1559 General Poster Session (Board #4A), Sat, 1:15 PM-5:15 PM<br />

Public perception <strong>of</strong> cancer risk. Presenting Author: Lisa Burns, University<br />

College Cork, Cork, Ireland<br />

Background: The public’s knowledge <strong>of</strong> cancer risk factors has rarely been<br />

studied in Ireland. An understanding <strong>of</strong> this can help inform cancer<br />

prevention programs. Methods: An online surveywas used to assess the<br />

public’s perception <strong>of</strong> cancer risk. Results: 525 people completed the<br />

survey. Mean age was 40yrs (range:18-74), 82% were female and 36% had<br />

college degrees. 81% were concerned about developing cancer, however<br />

20% believed if cancer was in their family there was nothing they could do<br />

about personal cancer risk. 20% did not know that cancer risk increased<br />

with age, 27% believed that �50% <strong>of</strong> cancers are inherited, and 54%<br />

believed 10-20% <strong>of</strong> cancers are inherited. The top 5 risk factors listed by<br />

respondents were: smoking 85%, diet 74%, alcohol 44%, genetics 38%,<br />

and environment 31%. Only 32% were aware that obesity is a risk factor for<br />

cancer and 33% did not think the location <strong>of</strong> fat was important for cancer<br />

prevention. When given a list <strong>of</strong> potential behaviours relevant to cancer risk<br />

33% believed wearing a tight bra and 49% believed a blow to the breast<br />

could increase cancer risk. 87% believed genetics ‘strongly’ increased risk,<br />

85% stress, and 86% believed cell phones increased risk. 12% believed<br />

‘luck’ was important in avoiding cancer, 35% thought ‘detox’ diets and<br />

61% believed organic food reduced risk. Only 33% agreed with the<br />

statement that ‘frozen vegetables/fruit are as good as fresh’, 40% were<br />

unaware <strong>of</strong> the link between red meat and cancer. The following foods were<br />

thought to increase risk: cheese (29%), soy (9%), milk chocolate (30%),<br />

red wine (25%), and eggs (11%). Aerosol use (71%), cleaning agents<br />

(73%), smoking (99%), cooking methods (68%), processed meat (86%),<br />

food irradiation (77%), and genetically modified foods (81%) were believed<br />

to increased risk. The majority were aware that berries, green tea, garlic,<br />

brassica vegetables and physical activity <strong>of</strong> 30 minutes a day can reduce<br />

cancer risk. Conclusions: There is a sizable portion <strong>of</strong> the population who<br />

are misinformed about cancer risk. Most are aware <strong>of</strong> classic risk factors<br />

(e.g. smoking, poor diet). Many overestimate risk attributable to genetics,<br />

environment, stress, and underestimate age, obesity and sunlight. One in 5<br />

believes lifetime risk <strong>of</strong> cancer is non-modifiable.<br />

1561 General Poster Session (Board #4C), Sat, 1:15 PM-5:15 PM<br />

<strong>Clinical</strong> and genetic predictors <strong>of</strong> weight gain in patients diagnosed with<br />

breast cancer. Presenting Author: Sangeetha M. Reddy, Northwestern<br />

University Feinberg School <strong>of</strong> Medicine, Chicago, IL<br />

Background: Obesity and weight gain in breast cancer patients has been<br />

associated with decreased quality <strong>of</strong> life, decreased response to chemotherapy,<br />

increased cancer recurrence, and higher all-cause mortality. Our<br />

study was designed to identify factors that contribute to this weight gain.<br />

Methods: Chart review was conducted on 565 breast cancer patients to<br />

obtain weights and BMIs over an 18 month period from diagnosis, tumor<br />

characteristics (ER/PR/Her2 status, grade, presence <strong>of</strong> LN metastases,<br />

stage), demographics (age, race), clinical factors (menopausal status), and<br />

treatment regimens (chemotherapy, hormone therapy, radiation). Blood<br />

samples were genotyped for polymorphisms in FTO (fat mass and obesityassociated<br />

protein) and the adiponectin pathway, two pathways found to be<br />

associated with obesity and breast cancer risk. Results: See table. For<br />

genetic analysis, one statistically significant epistatic and three gene x<br />

environmental interactions were detected for adiponectin SNPs: rs822396d<br />

x BMI at diagnosis (effect size 8.65), rs2232853a x age (2.75),<br />

rs1501299a x BMI at diagnosis (3.63), and rs266729d x rs7539542d<br />

(6.40). Conclusions: We have identified multiple clinical and genetic<br />

variables that are likely predictors <strong>of</strong> weight gain in breast cancer patients.<br />

We are conducting a prospective study <strong>of</strong> 200 breast cancer patients to<br />

validate the findings <strong>of</strong> this retrospective study. By identifying a high risk<br />

patient population, we hope to target them for aggressive lifestyle interventions<br />

to prevent weight gain and thereby improve their mortality and<br />

morbidity.<br />

OR for statistically significant clinical variables.<br />

Univariate analysis<br />

Multivariate<br />

Variable<br />

6 mo 1 yr 18 mo analysis<br />

Chemotherapy 0.75** 0.88** 0.88**<br />

Hormone therapy 1.28** 1.18** 1.18** 1.20**<br />

Stage (3 or 4) 0.78** 0.86** 0.87** 0.85**<br />

Postmenopausal status at diagnosis 0.87* 0.91**<br />

LN metastases 0.84** 0.91* 0.91**<br />

ER status 1.37** 1.18** 1.18** 1.22**<br />

PR status 1.23** 1.11* 1.13**<br />

Grade (2 or 3) 0.80* 0.87* 0.89**<br />

African <strong>American</strong> vs. white 0.88* 0.90**<br />

Hispanic vs. white 1.43* 1.17*<br />

Asian vs. white<br />

*p � 0.05<br />

** p � 0.01<br />

0.79* 0.87*<br />

1560 General Poster Session (Board #4B), Sat, 1:15 PM-5:15 PM<br />

The effects <strong>of</strong> BR-DIM (BioResponse 3, 3’-Diindolylmethane) administered<br />

pre-prostatectomy on the androgen receptor (AR). Presenting Author:<br />

Elisabeth I. Heath, Karmanos Cancer Institute, Wayne State University,<br />

Detroit, MI<br />

Background: Consumption <strong>of</strong> cruciferous vegetables is associated with<br />

decreased risk <strong>of</strong> prostate cancer (PCa). 3,3’-diindolylmethane (DIM), an in<br />

vivo active compound formed after consumption <strong>of</strong> cruciferous vegetables,<br />

down-regulates the AR, and causes its nuclear exclusion which results in in<br />

vitro growth arrest and apoptosis <strong>of</strong> PCa cells. We conducted a biomarker<br />

trial evaluating BR-DIM in pre-prostatectomy patients with the primary<br />

objective <strong>of</strong> measuring DIM levels in prostate tissue and in plasma.<br />

Methods: Patients with organ-confined PCa who were candidates for surgery<br />

were treated with BR-DIM at a dose <strong>of</strong> 225 mg orally twice daily for a<br />

minimum <strong>of</strong> 14 days. Patients did not receive any androgen deprivation<br />

therapy. Patients received their last dose <strong>of</strong> BR-DIM the day before surgery.<br />

Blood samples for DIM levels were collected pre-treatment and just prior to<br />

surgery. DIM concentration was measured in the plasma and in prostate<br />

tissue specimens using a validated high-performance liquid chromatographic<br />

method with tandem mass spectrometric detection. AR was<br />

evaluated by immunohistochemistry (IHC). Results: 36 patients were<br />

treated at 2 institutions. The 26 evaluable patients had median age <strong>of</strong> 60<br />

years (range 41 - 76), 14 were Caucasian, and 10 were African <strong>American</strong>.<br />

Reasons for inevaluability included change in surgery date (n�4), inadequate<br />

BR-DIM treatment (2), withdrawal from study (2), canceled surgery<br />

(1), and other (1). Toxicity was minimal; only two patients with grade 3<br />

headache. Post dosing, DIM was found at a mean trough level <strong>of</strong> 10.2 ng/ml<br />

(range, 0.5 to 24.7) and 12.3 ng/gm <strong>of</strong> tissue (range, 0.0 to 26.8) in<br />

plasma and in prostate tissue, respectively. PSA levels had a slightly<br />

downward trend after BR-DIM treatment. In 18 <strong>of</strong> 20 evaluable PCa<br />

specimens, IHC showed nuclear exclusion <strong>of</strong> AR. Conclusions: BR-DIM was<br />

well tolerated, and DIM was detected in both plasma and prostate tissues at<br />

~12 h post dosing. Nuclear exclusion <strong>of</strong> AR was found in 90% <strong>of</strong> PCa<br />

specimens post BR-DIM dosing, suggesting in vivo inactivation <strong>of</strong> AR<br />

activity. PSA levels were slightly reduced overall. Accrual is ongoing and<br />

nearly complete. Additional studies with BR-DIM in PCa are warranted.<br />

1562 General Poster Session (Board #4D), Sat, 1:15 PM-5:15 PM<br />

Breast cancer risk reduction among patients with DCIS. Presenting Author:<br />

Melanie R. Palomares, City <strong>of</strong> Hope, Duarte, CA<br />

Background: The incidence <strong>of</strong> ductal carcinoma in situ (DCIS) has dramatically<br />

increased with widespread mammographic screening. Although risk <strong>of</strong><br />

recurrence <strong>of</strong> DCIS is low, it is associated with a higher risk for subsequent<br />

contralateral breast cancer (CBC), for which preventive measures are<br />

available. We evaluated the uptake <strong>of</strong> surgical and pharmacologic interventions<br />

to reduce CBC risk at our institution and investigated factors that may<br />

influence treatment choices for DCIS. Methods: City <strong>of</strong> Hope (COH) DCIS<br />

patients were identified using two sources, the Circulating Breast Tumor<br />

Marker (BrTM) Registry and the National Comprehensive Cancer Network<br />

(NCCN) database. Datasets were linked together, and treatment variables<br />

were cross-tabulated with patient and tumor characteristics. Results: Of<br />

782 patients with breast malignancy diagnosed since 1997, 370 were<br />

excluded due to concurrent or prior invasive disease, 8 due to suspected<br />

misclassification based on therapies received, and 4 due to treatment on<br />

protocol. Of the remaining pure DCIS patients, treatment choices were<br />

recorded for 289. Of those, 40 (14%) chose bilateral risk reduction<br />

mastectomy (BRRM), 82 (28%) unilateral mastectomy, 165 (57%) lumpectomy,<br />

and 2 had no surgery. Hormonal therapy (HT) was recorded for 215<br />

individuals who did not pursue BRRM: 124 (57%) took tamoxifen, 3 <strong>of</strong><br />

whom switched to raloxifene, 5 (2%) started with raloxifene, and 7 (3%)<br />

took an unspecified hormonal agent, for a total HT uptake <strong>of</strong> 55%. This<br />

included 8 <strong>of</strong> 29 women with ER-negative DCIS who chose HT for CBC risk<br />

reduction. Younger age at diagnosis was associated with BRRM (24% <strong>of</strong><br />

women diagnosed before age 50, 10% <strong>of</strong> those diagnosed 50-64, and 5%<br />

<strong>of</strong> women 65� had BRRM, p�0.001) and HT (59% <strong>of</strong> women �65 chose<br />

HT compared to 40% <strong>of</strong> women 65�,p�0.009). Within ethnic minorities,<br />

more Asian women chose BRRM (22% vs 7% <strong>of</strong> other minorities, p�0.08).<br />

Interestingly, fewer high grade DCIS women opted for HT (39% vs 55% for<br />

low to intermediate grade, p�0.06). Conclusions: Young women tend to<br />

pursue surgical prophylaxis. Among women who keep their breasts, HT<br />

uptake was high across all age and ethnic groups, except for those older<br />

than 65 at diagnosis. It is unclear if this is due to patient choice or reflects<br />

age bias in physician recommendation.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


1563 General Poster Session (Board #4E), Sat, 1:15 PM-5:15 PM<br />

Risk perception and knowledge <strong>of</strong> breast and colon cancers in a diverse<br />

population. Presenting Author: Katherine Lang, Virginia Commonwealth<br />

University, Richmond, VA<br />

Background: The accuracy <strong>of</strong> cancer risk perception has implications for<br />

preventive behaviors. Studies show that despite receiving personalized<br />

breast and colon cancer risk information, people continue to overestimate<br />

their numeric risks. It is still not fully understood why this occurs. Breast<br />

cancer, especially, is highly visible in the media so an individual may hear<br />

more about breast cancer, increasing general knowledge but potentially<br />

inflating risk perceptions. This study explores relationships between<br />

general knowledge and numeric risk perceptions for breast cancer (BC) and<br />

colon cancer (CC) among women. We hypothesize that general knowledge<br />

<strong>of</strong> BC will be high relative to CC, but risk perception for BC will be less<br />

accurate. Methods: Data was obtained from the first 369 (final N�490)<br />

patients recruited for the Kin Fact study from a Women’s Health Clinic. Kin<br />

Fact is a randomized controlled trial examining effects <strong>of</strong> an intervention to<br />

increase cancer risk communication in families. Women complete baseline<br />

surveys including knowledge and numeric risk perception measures for BC<br />

and CC. We use CA Gene s<strong>of</strong>tware to calculate actual lifetime risk for BC<br />

and CC. Correlations, t-tests and linear regressions were used for the<br />

analysis. Results: Women averaged 33 years old, and 58% were African<br />

<strong>American</strong>. Average lifetime risk was 3% for CC and 11% for BC. Women<br />

overestimated their numeric risk for both BC and CC, but the mean<br />

overestimation for BC (24%) was significantly larger than for CC (19%)<br />

(p�0.001). Average scores for BC knowledge were also significantly higher<br />

than for CC (p�0.001). Compared to knowledge about CC, women who had<br />

greater knowledge <strong>of</strong> BC also were more inaccurate in terms <strong>of</strong> their<br />

perceived numeric risk (r� -0.131, p�0.016). This finding remained<br />

significant controlling for age, race and genetic literacy. Conclusions:<br />

Results endorse an apparently paradoxical effect that compared with CC,<br />

women with increased knowledge <strong>of</strong> BC have less accurate risk perception<br />

for BC. Inaccuracies in perceived risk can affect psychosocial well-being<br />

and adherence to screening and prevention recommendations. Findings<br />

reveal a need for increasing knowledge about cancer without adversely<br />

impacting the accuracy <strong>of</strong> risk perceptions.<br />

1565 General Poster Session (Board #4G), Sat, 1:15 PM-5:15 PM<br />

Factors and trends in cancer screening in the United States from 2004 to<br />

2010. Presenting Author: Thanyanan Reungwetwattana, Mayo Clinic,<br />

Rochester, MN<br />

Background: Understanding the prevalence <strong>of</strong> cancer screening in the US<br />

and the factors associated with its accessibility is important for public<br />

health promotion. Methods: The 2004 and 2010 Behavioral Risk Factor<br />

Surveillance Systems were used to ascertain cancer screening rates among<br />

populations indicated for each test by age, gender, and the <strong>American</strong><br />

Cancer <strong>Society</strong> recommendation for cancer screenings [fecal occult blood<br />

test (FOBT) or endoscopy for colorectal cancer (CRC) screening, digital<br />

rectal examination (DRE) or prostate specific antigen (PSA) for prostate<br />

cancer screening, clinical breast examination (CBE) or mammogram for<br />

breast cancer screening, and Papanicolaou (Pap) test for cervical cancer<br />

screening]. Results: Over this period, CRC and breast cancer screening rates<br />

significantly increased (15.9%, 13.9%) while prostate and cervical cancer<br />

screening rates significantly decreased (1.2%, 5.2%). Race/ethnicity<br />

might be an influence in CRC and cervical cancer screening accessibility.<br />

Prostate cancer screening accessibility might be influenced by education<br />

and income. The older-aged populations (70-79, �79) had high prevalence<br />

<strong>of</strong> CRC, prostate and breast cancer screenings even though there is<br />

insufficient evidence for the benefits and harms <strong>of</strong> screenings in the<br />

older-aged group. Conclusions: The disparities in age, race/ethnicity, health<br />

insurance, education, employment, and income for the accession to cancer<br />

screening <strong>of</strong> the US population have decreased since 2004. The trajectory<br />

<strong>of</strong> increasing rates <strong>of</strong> CRC and breast cancer screenings should be<br />

maintained. To reverse the trend, the causes <strong>of</strong> the decreased rate <strong>of</strong><br />

cervical cancer screening and the high rates <strong>of</strong> screenings in older-aged<br />

populations should, however, be further explored.<br />

Proportion <strong>of</strong> cancer screenings in year 2004 and 2010 (weighted %).<br />

Screenings 2004 2010 Absolute difference P value<br />

CRC<br />

FOBT<br />

17.52 18.25<br />

�0.73 0.001<br />

Sigmoidoscope/colonoscope 49.66 59.90 �10.24 0.000<br />

Total<br />

49.74 65.62 �15.88 0.000<br />

Prostate cancer<br />

DRE<br />

64.35 59.89<br />

-4.46 0.000<br />

PSA<br />

65.87 66.71<br />

�0.84 0.080<br />

Total<br />

77.40 76.24<br />

-1.16 0.008<br />

Breast cancer<br />

CBE<br />

63.26 82.99 �19.73 0.000<br />

Mammogram<br />

74.06 74.88<br />

�0.82 0.004<br />

Total<br />

74.06 87.94 �13.88 0.000<br />

Cervical cancer<br />

Pap 76.06 70.83 -5.23 0.000<br />

Cancer Prevention/Epidemiology<br />

101s<br />

1564 General Poster Session (Board #4F), Sat, 1:15 PM-5:15 PM<br />

First report on screening an asymptomatic population for cancer: The yield<br />

<strong>of</strong> an integrated cancer prevention center. Presenting Author: Tal Sella,<br />

Sheba Medical Center, Ramat Gan, Israel<br />

Background: Cancer is a leading cause <strong>of</strong> mortality worldwide. Screening is<br />

a key strategy for reducing cancer morbidity and mortality. We aimed to<br />

evaluate the utility <strong>of</strong> cancer screening in an asymptomatic population at<br />

an integrated cancer prevention center. Methods: One-thousand consecutive<br />

asymptomatic, apparently healthy adults, aged 20-80 years, were<br />

screened for early detection <strong>of</strong> 11 common cancers by routine screening<br />

tests. Results: Malignant and benign lesions were found in 2.4% and 7.1%<br />

<strong>of</strong> the screenees, respectively. The most common malignant lesions were in<br />

the gastrointestinal tract and breast followed by gynecological and skin.<br />

The compliance rate for the different screening procedures was considerably<br />

higher than the general Israeli population – 78% compared to 60% for<br />

mammography (p�0.001) and 39% compared to 16% for colonoscopy<br />

(p�0.001). Advanced age, family history and certain lifestyle parameters<br />

were associated with increased risk for cancer. Moreover, polymorphisms in<br />

the APC and CD24 genes indicated high cancer risk. When two <strong>of</strong> the<br />

polymorphisms existed in an individual, the risk for a neoplastic lesion was<br />

extremely high (OR 2.3 [95% CI 0.94-5.9]). Conclusions: A significant<br />

number <strong>of</strong> neoplastic lesions were diagnosed at an early stage. Polymorphisms<br />

in the APC and CD24 genes may identify individuals at an<br />

increased risk for cancer. Cancer may be diagnosed at an early stage using<br />

the screening facilities <strong>of</strong> a multidisciplinary outpatient clinic.<br />

1566 General Poster Session (Board #4H), Sat, 1:15 PM-5:15 PM<br />

Sun exposure pr<strong>of</strong>ile in the French population: Results <strong>of</strong> the EDIFICE<br />

melanoma survey. Presenting Author: Bruno Sassolas, CHRU Brest, Brest,<br />

France<br />

Background: The main environmental risk factor for melanoma is related to<br />

UV-exposure. Our objective was to evaluate in a survey the pr<strong>of</strong>ile <strong>of</strong> sun<br />

exposition in the French population as well as the level <strong>of</strong> knowledge about<br />

UV risk and UV protection. Methods: “Edifice Melanoma” a nationwide<br />

observational survey was conducted in France via phone interviews among a<br />

representative sample <strong>of</strong> 1502 subjects aged � 18, using the quota<br />

method. The survey took place shortly after the summer (September 28th to<br />

October 20th , 2011). Results: From the total population <strong>of</strong> 1502 subjects,<br />

330 (22%) denied any sun exposition. Among those declaring being<br />

exposed to the sun (1172), 62% affirmed using a photoprotection method<br />

(clothes, sun protective cream). The opportunities for sun exposure arose<br />

during holidays (85%), sport (79%) or pr<strong>of</strong>essional activities (23%), with<br />

an annual average exposure <strong>of</strong> 113 days. 902 subjects (77%) declared<br />

avoiding sun exposure between 12 and 4 PM. The main sociodemographic<br />

characteristics <strong>of</strong> subjects exposed to the sun, compared to non-exposed<br />

ones are as followed (p�0,05): men, aged �40, with no children or with<br />

children aged �15, from high social and occupational group, with at least a<br />

2-years university degree. The analysis <strong>of</strong> sociodemographic characteristics<br />

<strong>of</strong> subjects declaring to use sun protective measures are (p�0,05): women<br />

from high social and occupational group, with at least a 2-years university<br />

degree, with high income levels. We explored additional risk behavior in the<br />

population studied. Subjects who declared having sun exposure were more<br />

likely (p�0,05) to be : smokers, alcohol users and/or alcoholic, tanning bed<br />

users before the age <strong>of</strong> 30, and have a high number <strong>of</strong> nevi (�50). On the<br />

other hand, prior history <strong>of</strong> cancer was associated with a non-exposure<br />

behavior. Conclusions: These data provide important new information about<br />

sun behavior in the French population and might give important clues in<br />

the field <strong>of</strong> public health and education to increase the effectiveness <strong>of</strong><br />

melanoma primary prevention campaigns. A better knowledge <strong>of</strong> the target<br />

to improve would provide better results for public health benefit.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


102s Cancer Prevention/Epidemiology<br />

1567 General Poster Session (Board #5A), Sat, 1:15 PM-5:15 PM<br />

Cancer screening: Source <strong>of</strong> information and level <strong>of</strong> trust. Presenting<br />

Author: Jean F. Morere, Oncology Department, Hôpital Avicenne, Bobigny,<br />

France<br />

Background: Providing public information is critical for cancer control.<br />

EDIFICE surveys were conducted to provide a better understanding <strong>of</strong> the<br />

participation <strong>of</strong> the French population in cancer screening programs<br />

(colorectal, breast) or individual cancer screening (prostate). To evaluate<br />

sources <strong>of</strong> information in the general population and their level <strong>of</strong> trust,<br />

specific questions were addressed in the EDIFICE 3 survey. Methods: This<br />

third nationwide observational study, EDIFICE 3, was conducted by phone<br />

interviews among a representative sample <strong>of</strong> 1603 subjects aged between<br />

40 and 75 years old, using the quota method. Questions on the source <strong>of</strong><br />

information were: Concerning cancer screening, what are all your means <strong>of</strong><br />

information? What level <strong>of</strong> trust (quoted from 1 to 10) do you have on the<br />

following media <strong>of</strong> information; your own general practitioner, physicians in<br />

general, patient associations, national health insurance system, health<br />

authorities, your close circle, radio and television, lay press, internet.<br />

Results: In the overall population, the most frequently quoted means <strong>of</strong><br />

information for cancer screening were: radio and television (61%), general<br />

practitioners (52%), lay press (33%), institutional letters (19%) and<br />

internet (18%). People under 50 years old named significantly more <strong>of</strong>ten<br />

radio and television (66% vs 58%) and internet (24% vs 15%), while older<br />

people above 50 years old (target population for screening programs) more<br />

frequently named institutional letters (26% vs 7%). In a trust scale from 0<br />

to 10, the best level <strong>of</strong> trust was achieved by general practitioners (8.2),<br />

patient associations (6.8), national health insurance system (6.6) and<br />

health authorities (6.2). In contrast, radio and television (5.7), lay press<br />

(5.2) and internet (4.6) were mistrusted. Overall, women and younger<br />

subjects were more trustful. Conclusions: Radio and television are the most<br />

frequently quoted source <strong>of</strong> information but paradoxically they are not<br />

highly trusted. The general practitioner appears to be the best option<br />

(trusted and used) for providing cancer screening information. As institutional<br />

letters were frequently quoted and highly trusted, they might be<br />

useful for informing the population even before the age <strong>of</strong> recommended<br />

screening.<br />

1569 General Poster Session (Board #5C), Sat, 1:15 PM-5:15 PM<br />

Patient perceptions <strong>of</strong> stool-based DNA testing across racial groups.<br />

Presenting Author: Lily Huang, Case Western Reserve University, Cleveland,<br />

OH<br />

Background: Despite the widespread acceptance <strong>of</strong> colonoscopy as the gold<br />

standard for Colorectal Cancer (CRC) screening, compliance rates with<br />

recommended CRC screening lag far behind screening for breast and<br />

cervical cancers. Patients <strong>of</strong>ten delay or forgo screening colonoscopy due to<br />

associated discomfort, inconvenience, anxiety, and high cost. In addition,<br />

recent data have casted doubt on its effectiveness, especially in detecting<br />

right-sided colon neoplasia. Stool-based DNA (sDNA) testing is an emerging<br />

CRC screening tool that is convenient, noninvasive, and effective in<br />

detecting both right and left sided colon cancer. The goal <strong>of</strong> this study was<br />

to evaluate patient attitudes towards sDNA testing, identify preferences in<br />

CRC screening tools, and assess racial differences in these attitudes and<br />

preferences. Methods: In a colonoscopy-based case-control study, 100<br />

patients <strong>of</strong> average CRC risk were asked to complete (1) sDNA testing<br />

(LabCorp ColoSure test), (2) screening colonoscopy, and (3) a patient<br />

satisfaction survey. Results: Eighty patients completed the study, including<br />

55 (69%) White, 22 (27%) African <strong>American</strong>, and 3 (4%) Others. Overall,<br />

patients rated sDNA testing favorably in all preparation and test related<br />

features, except for perceived accuracy. Patients reporting a preferred<br />

screening method favored sDNA testing (78%) to FOBT (12%) or colonoscopy<br />

(10%), and 83% <strong>of</strong> all patients reported that they are likely to repeat<br />

sDNA testing. Non-Caucasians rated sDNA testing less favorably than<br />

Caucasians – reporting more discomfort (ANOVA, p�0.004), more anxiety<br />

(ANOVA, p�0.001), and less suitability (Chi-Squared, p�0.0035). Of<br />

those patients reporting a preferred screening test, 82% <strong>of</strong> Caucasians and<br />

64% <strong>of</strong> non-Caucasians selected sDNA testing. Conclusions: Although<br />

sDNA testing was ranked less favorably by non-Caucasians, it was well<br />

received overall as the preferred CRC screening test, as compared to FOBT<br />

and colonoscopy. The differences between racial groups may reflect the<br />

generally lower uptake <strong>of</strong> CRC screening among non-Caucasians, regardless<br />

<strong>of</strong> the test. In conclusion, sDNA testing may be an effective means to<br />

increase CRC screening in African <strong>American</strong>s, hence preventing CRC and<br />

closing the racial disparity gap.<br />

1568 General Poster Session (Board #5B), Sat, 1:15 PM-5:15 PM<br />

Cancer screening in France: 3rd edition <strong>of</strong> the EDIFICE survey. Presenting<br />

Author: Jérôme Viguier, CHRU Trousseau, Tours, France<br />

Background: The EDIFICE survey program started in 2005 and was aimed at<br />

providing a better understanding <strong>of</strong> the participation <strong>of</strong> the French<br />

population in cancer screening programs and assess the evolution over<br />

time. The EDIFICE 3 survey was conducted in 2011, following EDIFICE 1<br />

(2005) and EDIFICE 2 (2008), and focused on colorectal, breast and<br />

prostate cancer. Methods: This third nationwide observational study,<br />

EDIFICE 3, was conducted by phone interviews among a representative<br />

sample <strong>of</strong> 1603 subjects aged between 40 and 75 years, using the quota<br />

method. The analysis focused on the target population <strong>of</strong> the national<br />

screening programs for breast and colorectal cancer (50-74 years). The<br />

same population was analysed for prostate cancer screening behaviours.<br />

Results: For breast cancer, the rate <strong>of</strong> women attending at least one<br />

screening test was 93%/94%/95% in 2005/2008/2011 respectively. A<br />

mammography had been performed as recommended within the last two<br />

years for 75%/83%/83% among them. We observed an increase in timing<br />

compliance between 2005 and 2011, significant for women aged 65-74.<br />

For colorectal cancer, the rate <strong>of</strong> subjects attending at least one screening<br />

test was 25%/38%/59%. A fecal test or colonoscopy had been performed<br />

according to the recommended timing for NA/30%/51% among them.<br />

Colorectal cancer screening has increased significantly in all age groups,<br />

especially between 65 and 69 years, and for both genders. For prostate<br />

cancer, the rate <strong>of</strong> men having performed at least one screening test (PSA<br />

and/or rectal examination) was 36%/49%/50%.This rate have significantly<br />

decreased in men aged 50-59 between 2008 and 2011(44% vs 37%,<br />

p�0.05). Conclusions: For National Programs, the attendance rate remains<br />

high for breast cancer screening and is improving for colorectal cancer<br />

screening. However, the European guideline objective rate <strong>of</strong> participation<br />

for colorectal cancer screening has not yet been reached. Despite the<br />

absence <strong>of</strong> recommendations, prostate cancer screening is frequently<br />

carried out and stable overall.<br />

1570 General Poster Session (Board #5D), Sat, 1:15 PM-5:15 PM<br />

On-line breath analysis <strong>of</strong> volatile organic compounds as a method for<br />

colorectal cancer detection. Presenting Author: Francisco Zambrana Tevar,<br />

Department <strong>of</strong> Medical Oncology, Hospital Infanta S<strong>of</strong>ía, Madrid, Spain<br />

Background: Analysis <strong>of</strong> exhaled volatile organic compounds (VOCs) in<br />

breath is an emerging approach for cancer diagnosis, but little is known<br />

about its potential use as a biomarker for colorectal cancer (CRC). We<br />

investigated whether a combination <strong>of</strong> VOCs could distinct CRC patients<br />

from healthy volunteers. Methods: In a pilot study, we prospectively<br />

analyzed breath exhalations <strong>of</strong> 38 CRC patient and 43 healthy controls all<br />

scheduled for colonoscopy, older than 50 in the average-risk category. The<br />

samples were ionized and analyzed using a Secondary ElectroSpray<br />

Ionization (SESI) coupled with a Time-<strong>of</strong>-Flight Mass Spectrometer (SESI-<br />

MS). After a minimum <strong>of</strong> 2 hours fasting, volunteers deeply exhaled into the<br />

system. Each test requires three s<strong>of</strong>t exhalations and takes less than ten<br />

minutes. No breath condensate or collection are required and VOCs masses<br />

are detected in real time, also allowing for a spirometric pr<strong>of</strong>ile to be<br />

analyzed along with the VOCs. A new sampling system precludes ambient<br />

air from entering the system, so background contamination is reduced by<br />

an overall factor <strong>of</strong> ten. Potential confounding variables from the patient or<br />

the environment that could interfere with results were analyzed. Results:<br />

255 VOCs, with masses ranging from 30 to 431 Dalton have been identified<br />

in the exhaled breath. Using a classification technique based on the ROC<br />

curve for each VOC, a set <strong>of</strong> 9 biomarkers discriminating the presence <strong>of</strong><br />

CRC from healthy volunteers was obtained, showing an average recognition<br />

rate <strong>of</strong> 81.94%, a sensitivity <strong>of</strong> 87.04% and specificity <strong>of</strong> 76.85%.<br />

Conclusions: A combination <strong>of</strong> cualitative and cuantitative analysis <strong>of</strong> VOCs<br />

in the exhaled breath could be a powerful diagnostic tool for average-risk<br />

CRC population. These results should be taken with precaution, as many<br />

endogenous or exogenous contaminants could interfere as confounding<br />

variables. On-line analysis with SESI-MS is less time-consuming and<br />

doesn’t need sample preparation. We are recruiting in a new pilot study<br />

including breath cleaning procedures and spirometric analysis incorporated<br />

into the postprocessing algorithms, to better control for confounding<br />

variables.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


1571 General Poster Session (Board #5E), Sat, 1:15 PM-5:15 PM<br />

The effect <strong>of</strong> family history on adherence to breast cancer screening<br />

guidelines. Presenting Author: Lauren Marie Hibler, Yale University, New<br />

Haven, CT<br />

Background: A family history <strong>of</strong> breast cancer increases the risk <strong>of</strong><br />

developing this disease. We sought to determine the effect <strong>of</strong> a family<br />

history <strong>of</strong> a first degree relative with breast cancer (FDFHx) on adherence to<br />

mammography (MMG) and clinical breast examination (CBE) guidelines.<br />

Methods: The National Health Interview Survey (NHIS), conducted annually<br />

by the Centers for Disease Control, is designed to be representative <strong>of</strong> the<br />

US population. The 2010 NHIS data was used to evaluate the effect <strong>of</strong><br />

FDFHx on the likelihood that women aged � 30 had MMG and/or CBE<br />

within the past year. Results: Of the 12,320 women aged � 30, 1276<br />

(10.7%) had a FDFHx; 1263 (99.0%) <strong>of</strong> female breast cancer and 13<br />

(1.0%) <strong>of</strong> male breast cancer. Overall, 4573 (38.4%) <strong>of</strong> women reported<br />

having had MMG within the past year; 5377 (46.3%) had CBE within the<br />

past year. FDFHx was associated with use <strong>of</strong> MMG and CBE within the past<br />

year (58.0% vs. 36.0%, p�0.001, and 57.5% vs. 45.0%, p�0.001,<br />

respectively). Women with a FDFHx <strong>of</strong> male breast cancer were no more<br />

likely to have MMG (72.5% vs. 57.9%, p�0.313) nor CBE (66.8% vs.<br />

57.4%, p�0.518) within the past year than those with a FDFHx <strong>of</strong> female<br />

breast cancer. On multivariate analysis controlling for age, race, personal<br />

history <strong>of</strong> breast cancer (PHx), income, insurance, education and region,<br />

FDFHx was associated with a higher likelihood <strong>of</strong> having MMG (OR�1.75;<br />

95% CI: 1.48-2.06, p�0.001) and CBE (OR�1.48; 95% CI: 1.26-1.74,<br />

p�0.001) within the past year. Region was not associated with adherence<br />

to either MMG or CBE screening guidelines. Conclusions: FDFHx, along with<br />

age, race, PHx, income, insurance, and education, is an independent<br />

predictor <strong>of</strong> adherence to screening MMG and CBE guidelines. Still, only<br />

58% <strong>of</strong> women with a FDFHx had MMG and CBE in the past year. Further<br />

efforts are warranted to improve screening in this population at increased<br />

risk.<br />

Multivariate analysis.<br />

Factor<br />

MMG<br />

p value<br />

CBE<br />

p value<br />

FHx �0.001 �0.001<br />

Age �0.001 �0.001<br />

Race 0.018 �0.001<br />

PHx �0.001 �0.001<br />

Income �0.001 �0.001<br />

Insurance �0.001 �0.001<br />

Education 0.005 �0.001<br />

Region 0.700 0.196<br />

1573 General Poster Session (Board #5G), Sat, 1:15 PM-5:15 PM<br />

Esophageal cancer incidence gender disparity. Presenting Author: Luckson Noe<br />

Mathieu, Sidney Kimmel Comphrehensive Cancer Center at Johns Hopkins<br />

Hospital, Baltimore, MD<br />

Background: Over the past 30 years, esophageal cancer incidence has been<br />

increasing more rapidly than any other solid neoplasm in the Western world.<br />

Globally, there is a large male predominance in both esophageal squamous and<br />

adenocarcinoma. The reasons for this gender difference and the possible role <strong>of</strong><br />

estrogen are unclear. The objective <strong>of</strong> this study is to determine if estrogen<br />

exposure is consistent with the male predominance observed in esophageal<br />

cancer incidence. Methods: A database assessment <strong>of</strong> esophageal cancer<br />

incidence rates (age-adjusted) from 1975 to 2008 was conducted in the<br />

National SEER 9 Database and the Maryland Cancer Registry. Rates by gender<br />

and age were calculated. Gender-specific incidence rate ratios were compared<br />

across age groups. <strong>Annual</strong> percentage change (APC) was compared for each<br />

gender-age group. Results: In both national and state, male and female, adeno<br />

and squamous cell histologies, incidence rates increase with patient age. SEER<br />

data trends revealed the 50-64 aged female cohort as the only gender-age cohort<br />

APC decreasing between 1975-2008. Furthermore, a significant increase in<br />

incidence for females is observed in the 65� age groups; whereas among males,<br />

the rates are rising in all age groups. Conclusions: Using age as a proxy for<br />

estrogen exposure, our findings suggest a hormonal component in the declining<br />

male: female esophageal cancer incidence rate ratios with increasing age and<br />

confirm gender differences in incidence long observed in esophageal cancer.<br />

Histological features may be predictive <strong>of</strong> a lower rate ratio. (Bodelon et al.<br />

2011) Estrogen exposure may play a protective role in esophageal cancer which<br />

dissipates with time after age 60-69. Our data suggest possible future roles <strong>of</strong><br />

estrogen as a chemopreventive agent in esophageal cancer (Barone et al. 2011).<br />

Esophageal cancer annual percentage change, 1975-2008, SEER 9.<br />

Age 50-64 Age 65-74 Age 75�<br />

Males �1.2 �1.4 �1.9<br />

Females -1.5 �0.3 �0.7<br />

The M: F incidence ratio decreases with age in the nation (SEER) and in Maryland.<br />

Male to female incidence ratio, SEER, 1975-2008 and MD Registry 1992-<br />

2008.<br />

Age 40-49 Age 50-59 Age 60-69 Age 70-79 Age 80-89<br />

SEER 9.0 7.6 7.9 5.7 4.3<br />

MD 6.0 6.0 5.4 4.2 3.3<br />

Cancer Prevention/Epidemiology<br />

103s<br />

1572 General Poster Session (Board #5F), Sat, 1:15 PM-5:15 PM<br />

ABO blood group and the risk <strong>of</strong> breast cancer: Multicenter, case-control,<br />

observational study. Presenting Author: Yuksel Urun, Department <strong>of</strong><br />

Medical Oncology, Ankara University School <strong>of</strong> Medicine, Ankara, Turkey<br />

Background: The role <strong>of</strong> genetic factors in the development <strong>of</strong> cancer is<br />

widely accepted. ABO blood type is an inherited characteristic and previous<br />

studies have observed an association between ABO blood group and risk <strong>of</strong><br />

certain malignancies, including pancreatic and gastric cancer. The data on<br />

the role <strong>of</strong> ABO blood group and Rh factor in breast cancer is inconclusive.<br />

Methods: All patients who had breast cancer (BC) and treated between<br />

2000-2010 at the Departments <strong>of</strong> Medical Oncology <strong>of</strong> both Ankara and<br />

Hacettepe Universities (Ankara, Turkey) with defined ABO blood type and<br />

Rh factor were included in our retrospective reviews <strong>of</strong> tumor registry<br />

records. A group <strong>of</strong> volunteer healthy women donors <strong>of</strong> Turkish Red<br />

Crescent between 2004-2011 were identified as a control group, without<br />

any matching factors. The relationship <strong>of</strong> ABO blood types and Rh factor<br />

with various prognostic factors such as age at diagnosis, menopausal<br />

status, family history <strong>of</strong> breast cancer, and ER/PR/HER2 status were<br />

evaluated from 1740 BC patients. We compared the distributions <strong>of</strong> ABO<br />

blood types, Rh factors among 1740 patients and 204,553 healthy<br />

controls. Among BC patients, differences between each <strong>of</strong> aforementioned<br />

ABO blood groups and Rh factors with respect to various prognostic factors<br />

were explored, respectively. Results: Overall distributions <strong>of</strong> ABO blood<br />

groups as well as Rh factor were comparable between patients (44% A, 8%<br />

AB, 16% B, 32% O, 88% Rh�) and controls (41% A, 8% AB, 16% B, 35%<br />

O, 87% Rh�). However, there were statistically significant differences<br />

between patients and controls with respect to A vs. nonA (p�0.019) and<br />

marginal significance (p�0.051) for O vs. nonO. Among patients, there<br />

were statistically significant differences between A and nonA with respect<br />

to HER2 (p�0.0421), M stage (p�0.0447), T stage (p�0.0020). Only T<br />

stage (p�0.0337) were significantly different between O vs nonO. Grade<br />

(p�0.0227) and M stage (p�0.0107) were significantly different between<br />

Rh factors. Conclusions: In our study sample, ABO blood type was<br />

statistically significantly associated with breast cancer. Additional studies<br />

are necessary to determine the mechanisms by which ABO blood type may<br />

influence the risk <strong>of</strong> breast cancer.<br />

1574 General Poster Session (Board #5H), Sat, 1:15 PM-5:15 PM<br />

Study KBP-2010-CPHG: Characteristics and management <strong>of</strong> 7,051 new<br />

cases <strong>of</strong> lung cancer managed in French general hospitals in 2010.<br />

Presenting Author: Chrystele Locher, Saint-Faron Hospital, Meaux, France<br />

Background: An initial epidemiologic study was performed in 2000 by the<br />

French College <strong>of</strong> General Hospital Respiratory Physicians (Study KBP-<br />

2000-CPHG). Over the last 10 years, lung cancer management changed:<br />

new drugs such as targeted therapies appeared; new diagnostic techniques<br />

such as exploration for genetic mutations in the tumour have been<br />

developed; a new TNM classification has been drawn up. The aims <strong>of</strong> this<br />

study were to describe patient characteristics, first-line management, 1, 4<br />

and 5-year survival rates and to compare the results with those <strong>of</strong> Study<br />

KBP-2000-CPHG. Methods: A prospective multi-centre study included all<br />

patients �18 years presenting with a new case <strong>of</strong> primary lung cancer,<br />

histologically or cytologically diagnosed between 1 January and 31 December<br />

2010 and managed by one <strong>of</strong> the participating centers. A standardised<br />

form was completed for each patient. A steering committee checked the<br />

exhaustivity <strong>of</strong> data’s collection. Results: 7,610 patients from 119 general<br />

hospitals were included between 1 January and 31 December 2010. The<br />

main patient characteristics were: mean age 65.5 years (�/-11.3); 24.3%<br />

female; 10.9% non-smokers, 39.9% ex-smokers, 49.2% current smokers;<br />

68.9% performance status 0 and 1; 9.1% <strong>of</strong> patients had lost �10 kg<br />

within the previous 3 months. The main tumour characteristics were:<br />

13.7% small-cell lung cancer; 46.2% adenocarcinoma, 26.8% squamouscell<br />

carcinoma; EGFR mutation, explored in 30.5% <strong>of</strong> cases, were found in<br />

10.5% <strong>of</strong> cases; 16.4% stage IA to IIB, 13.4% stage IIIA, 10.2% stage<br />

IIIB and 60.0% stage IV. First-line treatments were: curative surgery,<br />

16.6%; chemotherapy, 63.4%; radiotherapy alone, 17.8%; combined<br />

radio-chemotherapy, 8.8%; and supportive care, 11.1%. Targeted therapy<br />

was used in 6.6% <strong>of</strong> patients treated by chemotherapy. Conclusions: In 10<br />

years, characteristics <strong>of</strong> lung cancer patients changed with an significantly<br />

increase <strong>of</strong> women, non-smokers, adenocarcinoma histology and stage IV at<br />

diagnosis.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


104s Cancer Prevention/Epidemiology<br />

1575 General Poster Session (Board #6A), Sat, 1:15 PM-5:15 PM<br />

Hepatitis B reactivation risk in patients with solid tumors receiving<br />

single-agent capecitabine versus doxorubicin-based chemotherapy. Presenting<br />

Author: Winnie Hui Yee Ling, National University Cancer Institute,<br />

Singapore<br />

Background: Reactivation <strong>of</strong> hepatitis B virus (HBV) in cancer patients on<br />

chemotherapy is a challenge especially in Asia where HBV infection is<br />

endemic. Risk <strong>of</strong> HBV reactivation differs according to cancer type,<br />

chemotherapy regimen, and concomitant use <strong>of</strong> steroids. The United<br />

States Centre for Disease Control and Prevention recommends universal<br />

screening for chronic HBV infection for all patients before chemotherapy<br />

although screening for low-risk chemotherapy regimens may not be cost<br />

effective. Methods: We sought to assess and compare HBV reactivation risk<br />

and effectiveness <strong>of</strong> prophylactic anti-viral therapy in preventing HBV<br />

reactivation in patients receiving a ‘high-risk’ (doxorubicin-based) chemotherapy<br />

regimen for which prophylactic anti-viral therapy for hepatitis B<br />

carriers is generally accepted versus a ‘low-risk’ (single agent capecitabine)<br />

chemotherapy regimen for solid tumours at a tertiary cancer centre. The<br />

electronic medical records <strong>of</strong> eligible patients were reviewed between<br />

January 2007 and December 2010. Results: A total <strong>of</strong> 708 patients were<br />

identified, including 434 and 274 who received doxorubicin-based chemotherapy<br />

and single agent capecitabine respectively. HBV screening rate was<br />

42.6% (51% for doxorubicin-based chemotherapy, 30% for single agent<br />

capecitabine, p�0.0001). 15/302 (5%) screened patients were found to<br />

be hepatitis B carriers (6 on doxorubicin, 9 on capecitabine). Overall,<br />

3/708 patients (0.4%) developed HBV reactivation (all from the unscreened<br />

doxorubicin group, 3/214 (1.4%) compared to 0/192 (0%)<br />

unscreened patients in the capecitabine group, p�0.0001). All 15<br />

identified hepatitis B carriers received prophylactic anti-viral therapy (14<br />

received lamuvidine, 1 had entacavir) and none had HBV reactivation.<br />

Conclusions: Not all chemotherapy regimens are associated with risk <strong>of</strong> HBV<br />

reactivation. Routine hepatitis B screening and prophylaxis for low-risk<br />

chemotherapy regimens such as single agent capecitabine may add<br />

morbidity and may not be cost effective.<br />

1577 General Poster Session (Board #6C), Sat, 1:15 PM-5:15 PM<br />

Assessing a prognostic model for predicting VTE occurrence in cancer<br />

patients. Presenting Author: Deesha Sarma, Princeton University, Princeton,<br />

NJ<br />

Background: Venous thromboembolism (VTE) poses a significant health risk<br />

to cancer patients and is one <strong>of</strong> the leading causes <strong>of</strong> death among this<br />

population. The most effective way to prevent VTE and reduce its<br />

prominence as a public health burden is by identifying high-risk patients<br />

and administering prophylactic measures. In 2008, Khorana et al. developed<br />

a model that classified patients by risk based on clinical factors.<br />

Methods: We conducted a retrospective study to test this model’s efficacy,<br />

on 150 patients with cancer receiving chemotherapy at an outpatient<br />

oncology clinic between January 1 and August 1, 2011. We aggregated<br />

data and assigned points based on the five factors in the Khorana model:<br />

site <strong>of</strong> cancer with 2 points for very high-risk site and 1 point for high-risk<br />

site, 1 point each for leukocyte counts more than 11 x 109 /L, platelet<br />

counts greater than 350 X 109 /L, hemoglobin levels less than 100 g/L<br />

and/or the use <strong>of</strong> erythropoiesis-stimulating agents, and BMI greater than<br />

35 kg/m2 (Khorana et al., Blood 2008). Based on this scoring system,<br />

patients with 0 points were grouped into the low-risk category, those with<br />

1-2 points were considered intermediate-risk, and those with 3-4 points<br />

were classified as high-risk. Results: As shown in the table, VTE incidence<br />

for the low-risk group was 1.9%, intermediate-risk group was 3.9%, and<br />

high-risk group was 9.1%. Conclusions: High-risk patients were about 4.5<br />

times more likely to develop a VTE than low risk patients. These results<br />

provide valuable insight in determining which patients might benefit from<br />

prophylaxis and in motivating the design <strong>of</strong> prospective clinical trials that<br />

assess the VTE predictive model in various ambulatory cancer settings.<br />

Risk level<br />

Number <strong>of</strong><br />

patients<br />

Number <strong>of</strong><br />

patients with VTE<br />

VTE incidence<br />

(%)<br />

Low risk (score�0) 52 1 1.9<br />

Intermediate risk (score�1-2) 76 3 3.9<br />

High risk (score�3�) 22 2 9.1<br />

Total: 150 6 4<br />

1576 General Poster Session (Board #6B), Sat, 1:15 PM-5:15 PM<br />

Predictors <strong>of</strong> pediatric and adult fibrosarcoma survival: Analyses <strong>of</strong> the<br />

Surveillance, Epidemiology, and End Results (SEER) program, 1973-<br />

2008. Presenting Author: Simona Ognjanovic, Biothera, Eagan, MN<br />

Background: Fibrosarcoma (FS) is the second most common group <strong>of</strong> s<strong>of</strong>t<br />

tissue sarcomas in children, however, most FS are diagnosed in adults. The<br />

goal <strong>of</strong> this study was to compare clinical features and outcomes <strong>of</strong> children<br />

and adults diagnosed with FS and identify predictors <strong>of</strong> survival. Methods:<br />

We have analyzed data from 569 children (� 19 years) and 5,508 adults<br />

(�19 years) diagnosed between 1973 and 2008 with FS (ICCC category<br />

IXb) available in the SEER database. Survival estimates were determined<br />

using survival time with end point death from any cause. Results: Comparison<br />

<strong>of</strong> two diagnostic periods, 1973-1989 vs. 1990-2008 showed that<br />

5-year survival rates have improved in adults (48.4% vs 60.9%, P�0.0001),<br />

but not in children (72.3% vs. 73.9%, P�0.16). Children with FS had<br />

significantly better outcome compared to adults (5-year survival rates were<br />

73.4% � 2%, and 56.7% � 0.7%, respectively, P�0.0001). Sites with<br />

survival rates higher than the median 5-year survival were grouped as<br />

favorable (extremities, head and neck, and genitourinary site); trunk,<br />

abdomen, and other sites were grouped as unfavorable. Cox proportional<br />

hazard regression model identified diagnosis in childhood, fibromyxosarcoma<br />

histologic subtype (ICD-O3 code 8811/3), favorable site, and<br />

localized stage as positive predictors <strong>of</strong> survival. Compared to fibromyxosarcoma,<br />

other histologic types had hazard ratio (HR) <strong>of</strong> 11.6 (95% CI�<br />

1.6-83.2, P�0.01) in children, and HR <strong>of</strong> 1.8 (95%CI�1.5-2.1,<br />

P�0.0001) in adults. Diagnosis at unfavorable site was associated with<br />

HR�1.9 (95%CI�1.4-2.6) in children and HR�1.4 (95%CI�1.3-1.5) in<br />

adults. Compared to localized, distant disease had HR�4.2 in children and<br />

HR�5.1 in adults, both highly significant (P�0.0001). Conclusions:<br />

Adults diagnosed with FS had worse survival than children. Predictors <strong>of</strong><br />

good outcome, including histiologic subtype, primary site, and stage, were<br />

valid in both children and adults.<br />

1579 General Poster Session (Board #6E), Sat, 1:15 PM-5:15 PM<br />

Incidence <strong>of</strong> bone metastases among children with cancer in Denmark. Presenting<br />

Author: Rohini Khorana Hernandez, Center for Observational Research,<br />

Amgen Inc, Thousand Oaks, CA<br />

Background: There are no published population-based studies on the incidence<br />

<strong>of</strong> bone metastases (BM) among children with cancer. The current literature is<br />

limited to small case series or case reports. Methods: We used the Danish Cancer<br />

Registry (DCR) to identify children �18 years old diagnosed with cancer<br />

between 1/1/1994 and 12/31/2009. Patients were followed from cancer<br />

diagnosis to BM, emigration, death, or end <strong>of</strong> study (1/1/2011). DCR data were<br />

linked to (1) Danish Civil Registration System to obtain information on death and<br />

emigration, and (2) Danish National Registry <strong>of</strong> Patients to identify ICD-10<br />

codes for BM. We estimated incidence rates (IRs) <strong>of</strong> BM and mortality rates<br />

overall and stratified by gender, calendar year, age, and primary tumor type.<br />

Results: During the study period, 2,652 children were identified with a first-time<br />

diagnosis <strong>of</strong> cancer, <strong>of</strong> whom 35 (1.3%) developed BM (mean follow-up <strong>of</strong> 7.0<br />

years). The IR <strong>of</strong> BM was 1.9 per 1,000 person-years (95% CI: 1.4 – 2.6); the<br />

highest rates occurred in children aged 12 – 17 years and among those with<br />

osteosarcoma (Table). Twenty-one (60%) children with BM died during followup,<br />

yielding a mortality rate <strong>of</strong> 192 per 1,000 person-years (95% CI: 125 –<br />

295). The median time from cancer diagnosis to BM was 221 days and from BM<br />

to death was 283 days. Conclusions: This study represents the first comprehensive<br />

examination <strong>of</strong> BM in children and reveals that BM is a rare event, with<br />

median survival <strong>of</strong> less than one year from diagnosis.<br />

IR <strong>of</strong> BM among children 0-17 years diagnosed with cancer in Denmark,<br />

1994-2009.<br />

Characteristic N<br />

N<br />

with BM<br />

IR <strong>of</strong> BM<br />

per 1,000 person-years<br />

(95% CI)<br />

Overall<br />

Gender<br />

Female<br />

Male<br />

Age at cancer diagnosis<br />

0–27days<br />

28 days – 23 months<br />

2 – 11 years<br />

12 – 17 years<br />

Calendar year <strong>of</strong> cancer diagnosis<br />

1994 – 1997<br />

1998 – 2001<br />

2002 – 2005<br />

2006 – 2009<br />

Tumor type<br />

Leukemia<br />

Lymphoma<br />

Brain / other CNS tumors<br />

Neuroblastoma<br />

Nephroblastoma<br />

Osteosarcoma<br />

Other tumors<br />

NA � Not Applicable.<br />

2,652<br />

1,163<br />

1,489<br />

38<br />

392<br />

1,314<br />

908<br />

642<br />

649<br />

691<br />

670<br />

827<br />

156<br />

378<br />

20<br />

115<br />

154<br />

1,002<br />

35<br />

14<br />

21<br />

0<br />

1<br />

18<br />

16<br />

3<br />

13<br />

8<br />

11<br />

1<br />

0<br />

1<br />

1<br />

1<br />

12<br />

19<br />

1.9 (1.4 - 2.6)<br />

1.7 (1.0 - 2.9)<br />

2.0 (1.3 - 3.1)<br />

NA<br />

0.37 (0.05 - 2.6)<br />

1.9 (1.2 - 3.1)<br />

2.6 (1.6 - 4.2)<br />

0.43 (0.14 - 1.3)<br />

2.3 (1.3 - 4.0)<br />

2.0 (1.0 - 4.0)<br />

6.0 (3.3 - 11)<br />

0.17 (0.02 - 1.2)<br />

NA<br />

0.45 (0.06 - 3.2)<br />

8.7 (1.2 - 62)<br />

1.0 (0.15 - 7.4)<br />

14 (8.0 - 25)<br />

2.4 (1.5 - 3.8)<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


1580 General Poster Session (Board #6F), Sat, 1:15 PM-5:15 PM<br />

Compliance <strong>of</strong> recommendations with French clinical practice in the management<br />

<strong>of</strong> thromboembolism in patients with cancer: The CARMEN study.<br />

Presenting Author: Marie-Antoinette Sevestre, Service de Medecine Vasculaire,<br />

Amiens, France<br />

Background: Long-term treatment with low-molecular-weight heparin (LMWH) is<br />

recommended for treatment <strong>of</strong> venous thromboembolism (VTE) in cancer<br />

patients. Few data are available on compliance in this population. Our study<br />

measured whether the management <strong>of</strong> VTE in patients with cancer was<br />

consistent with French recommendations. Methods: Compliance with recommendations<br />

(CR�) was analysed according to malignancy and VTE from a 500patient<br />

cross-sectional observational study run between May and October 2010.<br />

CR� was defined as the compliance to initial 10-day treatment followed by<br />

long-term LMWH for at least 3 months, avoiding LMWH in patients with renal<br />

insufficiency (SRI). All inpatients with a diagnosis <strong>of</strong> cancer and VTE <strong>of</strong> less than<br />

6 months were included in the study. Results: Of 500 patients included in 47<br />

centers, 242 (49%) were male, 81 (18%) had local (T�), 83 (18%) had<br />

loco-regional (N�) and 287 (64%) had metastatic malignancies. Malignancies<br />

were gastro-intestinal (25%), gynaecologic (23%), pulmonary (21%), haematological<br />

(14%), urologic (10%) or other (8%). Twelve patients had SRI. Overall,<br />

treatment was CR� in 289/500 patients (58% [95% CI 53%-62%]). Out <strong>of</strong> 12<br />

patients with SRI only 3 (25%) were treated long-term with vitamin K<br />

antagonists (VKA), as usually recommended. Tumour site influenced CR�<br />

(p�0.02). Treatment for haematological malignancy was poorly compliant with<br />

recommendations (32%) while patients with lung malignancy had the best<br />

compliance (68%). TNM stage and VTE location had no influence on treatment<br />

compliance. Conclusions: In French practice, treatment <strong>of</strong> cancer-related VTE is<br />

CR� in 58% <strong>of</strong> cases. TNM stage and VTE location do not influence compliance<br />

which remains insufficient, especially in patients with haematological malignancy.<br />

Characteristics N (%) CR� (%)*<br />

Primary tumor Gastrointestinal 92 (18) 56<br />

Gynaecologic 52 (10) 65<br />

Haematological 67 (13) 37<br />

Pancreas 32 (6) 50<br />

Lung 106 (21) 68<br />

Breast 63 (13) 57<br />

Urologic 48 (10) 65<br />

Other 40 (8) 72<br />

TNM stage T� 81 (18) 63<br />

N� 83 (18) 59<br />

M� 287 (64) 61<br />

VTE Pulmonary embolism 149 (30) 60<br />

Deep vein thrombosis 315 (63) 61<br />

Superficial thrombosis 16 (3) 25<br />

Other 18 (4) NA<br />

*Rate <strong>of</strong> compliance with recommendations.<br />

1582 General Poster Session (Board #6H), Sat, 1:15 PM-5:15 PM<br />

Age, race/ethnicity, and the ER/PR/HER2 subtypes in breast cancer.<br />

Presenting Author: Carol Parise, Sutter Institute for Medical Research,<br />

Sacramento, CA<br />

Background: The association <strong>of</strong> age and ER/PR/HER2 subtype in African-<br />

<strong>American</strong> (AA) and Hispanic women with breast cancer has been reported.<br />

This study examines the association <strong>of</strong> age and subtype among 12<br />

self-reported race/ethnicity categories. Methods: Using the California Cancer<br />

Registry 2000-2010, we examined 136,175 cases <strong>of</strong> first primary<br />

female invasive breast cancer. Race/ethnicity was stratified into 12<br />

categories. The distribution <strong>of</strong> age and race/ethnicity among the 8<br />

ER/PR/HER2 subtypes was examined. Age was stratified into �40, 40-69,<br />

and 70� years. For each age strata, odds ratios (OR) and 95% confidence<br />

intervals (CI) were computed to assess the association <strong>of</strong> race/ethnicity with<br />

ER-/PR-/HER2�, ER-/PR-/HER2- (TN), and ER�/PR�/HER2� (TP) when<br />

compared with the ER�/PR�/HER2- subtype. Results: The % <strong>of</strong> each race<br />

was: White 66.8; AA 6.0; Hispanic 15.9; Pacific Islander (PI) 0.3;<br />

Southeast Asian (SEA) 2.5; Indian Continent (IC) 0.7; <strong>American</strong> Indian;<br />

0.2; Chinese 2.5; Japanese 1.2; Filipino 3.1; Korean 0.7; Hispanic/nonwhite<br />

0.3. All AA women had increased ORs <strong>of</strong> the ER-/PR-/HER2�, TN<br />

and TP. Hispanic women mimicked the AA population except for young<br />

women with TP. Chinese (OR�0.53; 95%CI�0.37-0.76) Japanese<br />

(OR�0.51; 95%CI�0.28-0.96), and Filipino (OR�0.45; 95%CI�0.31-<br />

0.67) women �40 were less likely to have TN subtype. For women 40-69,<br />

SEA, Chinese, Filipino, and Korean women had increased ORs for TP, and<br />

ER-/PR-/HER2�. The Japanese had increased ORs for the TP (OR�1.23;<br />

95%CI�1.00-1.49) but decreased odds for the TN (OR�0.72;<br />

95%CI�0.57-0.90). IC women had increased odds for the TN (OR�1.38;<br />

95%CI�1.10-1.72). Korean women 70� were more likely to have the TP<br />

and ER-/PR-/HER2�. SEA had increased ORs for ER-/PR-/HER2� and TN,<br />

IC only had increased ORs for TN (OR�2.03; 95%CI�1.26-3.27).<br />

Chinese and Filipino women only had increased ORs for the ER-/PR-/<br />

HER2�. Conclusions: AA and Hispanic women <strong>of</strong> all ages are at increased<br />

risk <strong>of</strong> the TN, and ER-/PR-/HER2� subtypes. AA women <strong>of</strong> all ages and<br />

Hispanic women over age 40 are at increased risk <strong>of</strong> the TP subtype. The<br />

diversity <strong>of</strong> Asian women with regard to breast cancer necessitates<br />

accurately defining this population.<br />

Cancer Prevention/Epidemiology<br />

105s<br />

1581 General Poster Session (Board #6G), Sat, 1:15 PM-5:15 PM<br />

Castration resistance and high-risk disease among nonmetastatic (M0)<br />

prostate cancer (PC) patients on androgen deprivation therapy (ADT).<br />

Presenting Author: Melissa Pirolli, SDI Health, Plymouth <strong>Meeting</strong>, PA<br />

Background: Prostate-specific antigen (PSA) is a well-known PC biomarker.<br />

In the M0 setting, rising PSAs despite ADT is an indication <strong>of</strong> the<br />

development <strong>of</strong> castration-resistant prostate cancer (CRPC). In this disease<br />

state, men are at risk for developing bone metastasis (BM), which is<br />

associated with significant morbidity and may negatively affect survival.<br />

Using real-world data, we explored PSA-based criteria to identify patients<br />

who develop CRPC while on ADT and the subsets that may be at increased<br />

risk <strong>of</strong> BM. Methods: We used the Oncology Services Comprehensive<br />

Electronic Records (OSCER) database, which includes electronic medical<br />

record (EMR) data on cancer patients from 328 urology and oncology<br />

clinics in the US. Eligible patients were adult men with M0 PC with �1 PSA<br />

recorded between 3/1/2010 and 2/28/2011 and currently receiving ADT<br />

(gonadotropin-releasing hormone agonists or bilateral orchiectomy) for �6<br />

months (mos). We defined CRPC as two sequential PSA rises while on ADT<br />

and high risk for BM as any PSA �8 ng/mL or PSA doubling time (DT) �10<br />

mos, as described by Smith MR et al, Lancet 2012. We explored subsets <strong>of</strong><br />

CRPC patients who may be at even higher risk <strong>of</strong> BM using PSA thresholds<br />

(�8 ng/mL and �20 ng/mL) and DT (�4, 6, 8, and 10 mos). Results: Of<br />

1,818 men with M0 PC receiving ADT �6 mos, 36% (N�646) met the<br />

CRPC definition, <strong>of</strong> whom 80% (N�517) had PSA �8 ng/mL and/or PSA<br />

DT �10 mos (high risk). PSA DT alone explained 63% (44% / 70%) to<br />

93% (65% / 70%) <strong>of</strong> subgroup eligibility (Table), and emerged as a main<br />

driver in defining increased risk <strong>of</strong> BM for CRPC subsets. Conclusions: In<br />

this analysis <strong>of</strong> EMR data, over one-third <strong>of</strong> men with M0 PC on ADT met<br />

criteria for CRPC, and most CRPC patients (80%) may be considered at<br />

high risk for BM. Requiring �3 PSAs to define CRPC may be a limitation;<br />

however, because PSAs are closely monitored in patients on ADT, these<br />

definitions <strong>of</strong> CRPC and high risk may be useful in practice. These data<br />

suggest that PSA DT may be a more clinically meaningful measure <strong>of</strong><br />

defining CRPC subsets than absolute PSA thresholds.<br />

CRPC subsets PSA >8 ng/mL PSA >20 ng/mL No PSA threshold (PSA < 8 ng/mL)<br />

DT


106s Cancer Prevention/Epidemiology<br />

1584 General Poster Session (Board #7B), Sat, 1:15 PM-5:15 PM<br />

Impact <strong>of</strong> response shift on time to quality-<strong>of</strong>-life scores deterioration in<br />

patients with breast cancer. Presenting Author: Zeinab Hamidou, Biostatistics<br />

and Epidemiology Unit, Medical Information Department, Centre<br />

Georges-François Leclerc and EA4184, College <strong>of</strong> Medicine, Dijon, France<br />

Background: Time to quality <strong>of</strong> life (QoL) score deterioration (TD) is a<br />

method <strong>of</strong> longitudinal QoL data analysis that has been proposed for breast<br />

cancer (BC) patients (Hamidou et al Oncologist 2011). As for RECIST<br />

criteria, the optimal definitions dealing with reference should be explored.<br />

This study aims to study the impact <strong>of</strong> changes in internal standards (CIS)<br />

<strong>of</strong> response-shift (RS) and the influence <strong>of</strong> baseline QoL expectancies on<br />

TD. Methods: A prospective multicenter study including all women hospitalized<br />

for a primary BC was conducted. The EORTC-QLQ-C30 and BR-23<br />

questionnaires were used to assess the QoL at baseline, at the end <strong>of</strong> 1st hospitalization, and 3 and 6 months after. CIS was investigated by the<br />

then-test method. QoL expectancy was assessed at baseline using Likert<br />

scale. Deterioration was defined as a decrease in QoL scores reaching at<br />

least the mean difference identified as minimal clinically important<br />

difference (MCID) using Jaeschke’s transition question. Sensitivity analyses<br />

were done using the then-test score as reference score, and considering<br />

5 and 10 points as MCID. TD was estimated using Kaplan-Meier method.<br />

Cox regression analyses were used to identify factors influencing TD.<br />

Results: From February 2006 to February 2008, 381 women were included.<br />

For role functioning dimension, the median TD increased from 3.2<br />

months [95% CI: 3.1-3.36] to 4.76 months [3.3-6.2] when adjusting on<br />

CIS. For body image when adjusting on CIS, sentinel lymph node biopsy<br />

became significantly associated with longer TD (HR: 0.64[0.43-0.94]) as<br />

compared to axillary lymph node dissection, radiotherapy to a shorter TD<br />

(HR: 0.63[0.42-0.95] and the type <strong>of</strong> surgery had no effect on TD. For<br />

global health, cognitive and social functioning dimensions, patients expecting<br />

deterioration in their QoL had a significantly shorter TD. For fatigue and<br />

breast symptom scales, patients expecting no change had a significantly<br />

shorter TD, as compared to patients expecting an improvement. Sensitivity<br />

analyses using a MDCS <strong>of</strong> 5 or 10 confirmed these results. Conclusions: Our<br />

results suggest that it would be more accurate to take into account CIS<br />

component <strong>of</strong> RS as well as QoL expectancies to estimate TD <strong>of</strong> QoL scores<br />

in patient with BC.<br />

1586 General Poster Session (Board #7D), Sat, 1:15 PM-5:15 PM<br />

Outcomes <strong>of</strong> male breast cancer in the United States: A SEER database<br />

analysis from 1973 to 2008. Presenting Author: Akm Mosharraf Hossain,<br />

Ellis Fischel Cancer Center, Missouri University School <strong>of</strong> Medicine,<br />

Columbia, MO<br />

Background: Male breast cancer (MBC) is rare compared to female breast<br />

cancer (FBC). There has been no systemic study to examine the epidemiology<br />

<strong>of</strong> MBC in USA in the past decade. Methods: We examined 6,157 cases<br />

<strong>of</strong> MBC reported in SEER database from 1973-2008 (released 2011). We<br />

performed rate, frequency and survival sessions to examine age, sex, stage,<br />

grade, treatment modalities and survival using the SEER*Stat S<strong>of</strong>tware<br />

7.0.5. These variables were compared with that <strong>of</strong> 877,885 FBC cases<br />

reported in SEER 1973-2008. Results: The incidence rate was 1.3 (MBC)<br />

and 124 (FBC) per 100,000, respectively. Median age <strong>of</strong> diagnosis was<br />

higher in MBC compared to FBC (72 vs. 61 yrs. P�0.001). 18% <strong>of</strong> MBC<br />

were seen at age less than 60 years compared to 32% in FBC (p�0.001).<br />

FBC tend to be more hormone receptor positive (74% vs. 61%; p�0.001).<br />

There were 5,145 Caucasians (WH), 701 African <strong>American</strong>s (AA), 258<br />

Hispanics and 42 Asian patients. Stage III and IV, Grade III and IV<br />

histologies were more common in MBC. Staging and Grading did not differ<br />

among the different ethnicities except for Grade IV histology in AA 19%,<br />

Asians 20% when compared to WH 13% (p�0.001). Treatment modalities<br />

included, surgery (MBC 78% vs. FBC 93%; p�0.001), chemotherapy<br />

(MBC 82% vs. FBC 43%; p�0.001) and radiation therapy (MBC 19% vs.<br />

36% FBC; p�0.001). Treatment modalities were similar among WH and<br />

AA. Survival was higher in FBC compared to MBC (table) but improved over<br />

time in MBC. AA and Asians had lower 1-5 year survival compared to WH.<br />

Conclusions: MBC presents at a later age, higher stage and grade,<br />

predominantly requiring chemotherapy with an overall poor survival compared<br />

to FBC. But 1-5 year survival improved over time in MBC. This can be<br />

explained by early diagnosis, better treatment and other factors. Further<br />

studies should be performed in this regard.<br />

Comparison <strong>of</strong> survival between male and female breast cancer patients<br />

reported in SEER database from 1973 to 2008.<br />

Variable Male (N�6,157) Female (N�877,885) p value<br />

1 year (%) 91.3 (91.6 – 93) 96.1 � 0.001<br />

2 years (%) 85.9 (84.9 – 86.9) 90.1 � 0.001<br />

3 years (%) 79.9 (78.7 – 81.1) 85.1 � 0.001<br />

4 years (%) 73.5 (72.2 – 74.8) 80.7 � 0.001<br />

5 years (%) 67.9 (66.6 – 69.3) 76.7 � 0.001<br />

1585 General Poster Session (Board #7C), Sat, 1:15 PM-5:15 PM<br />

An exploratory study to determine if BRCA1 and BRCA2 mutation carriers<br />

have higher risk <strong>of</strong> cardiac toxicity. Presenting Author: Roohi Ismail-Khan,<br />

H. Lee M<strong>of</strong>fitt Cancer Center & Research Institute, Tampa, FL<br />

Background: Anthracycline related cardiac toxicity (CT) is a concern in<br />

treating women with breast cancer. The prevalence <strong>of</strong> heart failure (HF)<br />

affects 2% <strong>of</strong> the population, less so in women. Patients receiving<br />

anthracycline based therapy (ABT) have a dose-dependent risk <strong>of</strong> reduction<br />

in ejection fraction. Recent work by Dr. Verma suggests that BRCAdeficient<br />

mice manifest increased levels <strong>of</strong> cardiac failure. We sought to<br />

explore the risk for CT and evaluate the association between ABT and HF in<br />

female BRCA mutation carriers. Methods: An online survey was developed<br />

to collect information about breast cancer treatment (including HF) in<br />

BRCA mutation carriers through the national BRCA patient advocacy<br />

organization FORCE via their 2011 conference and their website as well as<br />

the M<strong>of</strong>fitt-based Inherited Cancer Registry (ICARE). The prevalence <strong>of</strong> CT<br />

and HF was calculated in both BRCA 1 and 2 breast cancer patients and<br />

compared to general population risks. Data from those that received ABT<br />

was compared to published HF rates from ABT. Results: Our sample<br />

included 227 BRCA1 carriers and 164 BRCA2 carriers in whom 6.4%<br />

reported cardiac toxicity (i.e., either HF and/or CT). This included similar<br />

proportions in BRCA1 vs BRCA2 carriers (i.e., 6.6% and 6.1%, respectively).<br />

These proportions are significantly higher than the published rate <strong>of</strong><br />

2% (all p-values � 0.001). Specifically regarding ABT, 112 mutation<br />

carriers had doxorubicin (Adriamycin) for treatment <strong>of</strong> whom 8% reported<br />

HF, similar to the 11 who had Epirubicin (11 patients), <strong>of</strong> whom 9%<br />

reported HF. Conclusions: Our data suggests that BRCA mutation carriers<br />

may have an increased risk <strong>of</strong> CT compared to the general population. In<br />

particular, women with BRCA mutations treated with ABT also appear to<br />

have a higher risk <strong>of</strong> developing CT and/or HF. This exploratory study<br />

provides the basis upon which larger retrospective and prospective studies<br />

are currently being planned. The high percentage <strong>of</strong> CT observed in this<br />

study requires confirmation as they could inform recommendation for<br />

cardiac screening and review <strong>of</strong> the current standard for ABT use in this<br />

population.<br />

1587 General Poster Session (Board #7E), Sat, 1:15 PM-5:15 PM<br />

Evaluation <strong>of</strong> perioperative therapy for stage II and III rectal cancer in<br />

California, 1994-2009. Presenting Author: Myung Mi Cho, Loma Linda<br />

University School <strong>of</strong> Public Health, Department <strong>of</strong> Epidemiology and<br />

Biostatistics, Loma Linda, CA<br />

Background: Surgical treatment <strong>of</strong> stage II and III rectal cancer changed<br />

little during the past two decades, while ancillary therapies have undergone<br />

two evolutionary changes. We sought to evaluate changes in chemoradiation<br />

(chemoRT) practices for patients receiving radical operations for stage<br />

II and III rectal cancer in California, 1994-2009. Methods: We extracted<br />

age, sex, race/ethnicity (R/E), socioeconomic status (SES) (demographic<br />

variables) and chemoRT variables for stage II and III rectal cancer cases in<br />

California. We used unconditional logistic regression to compute independent<br />

odds ratios <strong>of</strong> receiving preoperative chemoRT (PreOP Tx) to postoperative<br />

chemoRT (PostOP Tx) during Early (1994-1999), Middle (2000-<br />

2003), and Late (2004-2009) time-periods using the late time-period as<br />

the referent group. Results: Logistic regression analyses assessing timeperiod<br />

and demographic variables reveal trends and odds ratios with 95%<br />

confidence intervals (OR; CI) for chemoRT variables. Our findings showed a<br />

stepwise increase in the odds <strong>of</strong> PreOP Tx to PostOP Tx across<br />

Early, Middle, to Late time-periods (OREarly/Late�0.11; 0.10, 0.13,<br />

ORMiddle/Late�0.36; 0.32, 0.40). Age �40 and 40-49, independently<br />

predicted higher odds <strong>of</strong> PreOP Tx than PostOP Tx compared to age 50-74,<br />

while the findings for subjects age 75� were similar to those seen in the<br />

age 50-74 group (OR�40/50-74�1.63; 1.30, 2.05, OR40-49/50-74�1.18; 1.03, 1.36, OR75�/50-74�1.04; 0.90, 1.18). Females showed lower odds<br />

<strong>of</strong> PreOP Tx to PostOP Tx than males (ORFemale/Male�0.85; 0.77, 0.93).<br />

Odds <strong>of</strong> PreOP Tx to PostOP Tx were significantly lower among Asian/Other<br />

(A/O) and Hispanic relative to Non-Hispanic White (NHW), while findings<br />

were similar to NHW for Non-Hispanic Black (NHB) (ORA/O/NHW�0.85; 0.74, 0.98, ORHispanic/NHW�0.84; 0.73, 0.96, ORNHB/NHW�0.92; 0.73,<br />

1.15). SES did not predict odds <strong>of</strong> PreOP Tx versus PostOP Tx. Conclusions:<br />

These findings depict independent increases in PreOP Tx across the three<br />

time-periods among stage II and III rectal cancer patients receiving radical<br />

operations. Further analyses will evaluate survival characteristics predicted<br />

by changes in perioperative therapies for stage II and III rectal cancer<br />

patients.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


1588 General Poster Session (Board #7F), Sat, 1:15 PM-5:15 PM<br />

Second primary gynecologic neoplasms among breast cancer survivors.<br />

Presenting Author: Maria Pilar Barretina Ginesta, Institut Catala d’Oncologia<br />

Hospital Universitari Josep Trueta, Girona, Spain<br />

Background: Excess <strong>of</strong> risk <strong>of</strong> several second primary malignancies after<br />

treatment for breast cancer (BC) has been reported. This risk can be related<br />

with shared risk factors, cancer susceptibility genes or prior treatments<br />

received. Hormonal factors and hormone therapy are known to be risk<br />

factors for both BC and some gynecological malignancies. The aim <strong>of</strong> this<br />

study was to asses gynecological cancer risk as a second neoplasm among<br />

BC patients in our population. Methods: Patients diagnosed with invasive<br />

BC (CIE10: C50.0-C50.9) and registered in the Girona Cancer Registry<br />

from 1980 to 2006 were included in our study. We analyzed their<br />

incidence <strong>of</strong> second gynaecological malignancies except for contralateral<br />

BC. Standardized Incidence Ratios (SIR) and absolute excess <strong>of</strong> risk (AER)<br />

<strong>of</strong> these patients compared to general population were calculated. Results:<br />

6.209 patients were diagnosed <strong>of</strong> invasive BC in this period, with median<br />

age at diagnosis 62 years, median follow-up 4 years. 84 <strong>of</strong> them developed<br />

a second malignancy <strong>of</strong> gynaecological origin (SIR 1,98 CI 95% 1,59-<br />

2,44; AER 104,01/100.000 person-year). We observed 4 uterine cervix<br />

(SIR 0.64 IC95% 0,20-1,54), 3 vulvar-vaginal (SIR 0,96 IC95% 0,24-<br />

2,60), 13 ovarian (SIR 1,13 IC95% 0,63-1,89) and 63 uterine corpus<br />

neoplasms (UC), as well as 1 genital female tract neoplasm unspecified.<br />

High and statistically significant SIR was observed for gynecological<br />

malignancies in general and specifically for UC(SIR 3.14 IC 95% 2,44-<br />

4,00). With this finding, histology <strong>of</strong> UC cases was reviewed. 12 out <strong>of</strong> 63<br />

were malignant histologies, not otherwise specified. Among the remaining<br />

51, there were 8 type II (1 clear cell and 7 serous adenocarcinoma,<br />

15.7%), 34 type I (endometrioid adenocarcinoma, 66.7%) and 6 carcinosarcomas<br />

(11.8%). There were also 2 adenosarcomas (3.9%) and 1<br />

mucinous adenocarcinoma (1.9%). Conclusions: Women with previous BC<br />

have an elevated risk <strong>of</strong> developing a second primary gynecological<br />

malignancy compared with general population, particularly for UC. These<br />

patients should be followed up for its early detection. We detected a slight<br />

increase in unfavourable uterine carcinoma histologies respect to the<br />

general population. Further investigation <strong>of</strong> this finding is warranted.<br />

1590 General Poster Session (Board #7H), Sat, 1:15 PM-5:15 PM<br />

Renal impairment after chemotherapy in lung (LC), colorectal (CRC), and<br />

breast cancer (BC) patients (pts) from the Henry Ford Health System<br />

(HFHS) tumor registry. Presenting Author: Laura Chu, Genentech, South<br />

San Francisco, CA<br />

Background: Renal impairment is a common comorbidity in cancer pts. It<br />

can delay excretion and alter metabolism <strong>of</strong> anticancer drugs leading to<br />

further renal toxicity. The objective <strong>of</strong> this study was to determine the<br />

proportion <strong>of</strong> pts with renal impairment, defined as the presence <strong>of</strong><br />

proteinuria (PR), acute kidney injury (AKI), or chronic kidney disease<br />

(CKD), after chemotherapy (chemo) initiation. Methods: LC, CRC, and BC<br />

pts newly diagnosed between 2000 and 2007 (regardless <strong>of</strong> prior renal<br />

status) were identified using the HFHS tumor registry, with follow-up<br />

through Mar 2009. AKI was defined based on RIFLE (severity categories<br />

only) using lab data within 1 to 7 days after chemo initiation. CKD was<br />

defined based on NKF-KDOQI criteria and lab data up to 1 year after chemo<br />

initiation. Proteinuria was defined as protein/creatinine �30 mg/g within 3<br />

months after chemo initiation. Descriptive statistics include proportions<br />

stratified by renal impairment status (yes, no) at cancer diagnosis. Results:<br />

We identified 1,896 LC, 1,088 CRC, 1,611 BC chemo treated pts. Median<br />

age for all pts was 66 years. For LC and CRC, 56% and 51% were men,<br />

respectively. The proportion <strong>of</strong> pts with renal impairment after chemo by<br />

tumor type was the following: LC (AKI�23.4%; CKD�48.2%; PR�0.4%);<br />

CRC (AKI�20.3%; CKD�55.7%; PR�0.1%); BC (AKI�7.8%;<br />

CKD�63.9%; PR�0.6%). Pts with pre-existing renal impairment at<br />

cancer diagnosis were more likely to have impairment after chemo (Table).<br />

Conclusions: In LC, CRC, and BC pts, the proportion <strong>of</strong> pts with renal<br />

impairment after chemo initiation was high. Extent <strong>of</strong> renal impairment,<br />

especially AKI, appears to be associated with tumor type. Older age, as in<br />

this HFHS cohort, is associated with higher prevalence <strong>of</strong> CKD. Renal<br />

function should be closely monitored, particularly among pts with preexisting<br />

renal impairment, and preventive measures implemented to<br />

minimize further toxicity after treatment.<br />

Cancer Prevention/Epidemiology<br />

107s<br />

1589 General Poster Session (Board #7G), Sat, 1:15 PM-5:15 PM<br />

The influence <strong>of</strong> prognostic factors on metastatic breast cancer survival<br />

over time. Presenting Author: Margaret Quinn Rosenzweig, University<br />

<strong>of</strong> Pittsburgh School <strong>of</strong> Nursing, Pittsburgh, PA<br />

Background: Recent evidence suggests that survival in metastatic breast<br />

cancer is slowly improving associated with the use <strong>of</strong> better adjuvant and<br />

metastatic chemotherapeutic and targeted agents. Patient and clinical<br />

factors such as age, estrogen status, non-white race, Her 2 status, disease<br />

free interval and sites <strong>of</strong> metastatic breast cancer involvement indicate<br />

worse clinical outcome after recurrence. This analysis focused on the<br />

influence <strong>of</strong> these factors on metastatic breast cancer survival over time.<br />

Methods: Subjects were women with metastatic breast cancer from one<br />

large urban practice, <strong>of</strong> the University <strong>of</strong> Pittsburgh Cancer Institute Breast<br />

Cancer Program followed from 1999 through December, 2008. Patients<br />

were dichotomized into two time categories: A) 1999 through 2004 and B)<br />

2005 through 2008. Outliers <strong>of</strong> long term survivors (n �72) with survival<br />

extending beyond 6 years were excluded. Log rank tests were conducted for<br />

assessing the relationship between prognostic factors and survival. Results:<br />

Cohorts included patients diagnosed with metastatic breast cancer in 1999<br />

through 2004, (n�284) and 2005 through 2008, (n�332). They were<br />

followed up to December, 2011. Median survival improved over time<br />

(p�0.053). Estrogen negativity remained significant for worse survival<br />

across both time periods (p�0.0001). Age, presence <strong>of</strong> brain metastasis<br />

and Her 2 status were not significant for influence on survival at either time<br />

interval. Shorter disease free interval (p� 0.02), higher number <strong>of</strong><br />

metastatic sites (p�.001) and presence <strong>of</strong> visceral metastasis at diagnosis<br />

(p�0.003) became significant for worse survival in the 2005-2008<br />

intervals but had not been in the earlier time period. African <strong>American</strong> race<br />

was highly significant (�0.001) for worse survival in 1999-2004 but lost<br />

significance in 2005 through 2008 with dramatic survival increase<br />

(median survival - 12.5 months to 35 months). Conclusions: It is important<br />

for clinicians to clarify the prognostic features associated with worse<br />

outcomes in metastatic breast cancer. With newly emergent therapies and<br />

sensitivity toward specific patient factors these features evolve over time.<br />

1591 General Poster Session (Board #8A), Sat, 1:15 PM-5:15 PM<br />

Treatment patterns and comorbidities in a population-based cohort <strong>of</strong><br />

glioblastoma patients diagnosed 1997-2008. Presenting Author: Susan A.<br />

Oliveria, EpiSource, Newton, MA<br />

Background: Glioblastoma multiforme (GBM) is one <strong>of</strong> the most common,<br />

most malignant and rapidly progressive forms <strong>of</strong> brain cancer. Published<br />

reports <strong>of</strong> GBM in non-trial populations are limited. The purpose <strong>of</strong> this<br />

study was to assemble a population-based cohort <strong>of</strong> GBM patients with<br />

comprehensive clinical and demographic information and to define treatment<br />

patterns and outcomes in this population. Methods: GBM patients<br />

served by Henry Ford Health System (HFHS), a major GBM referral center,<br />

were identified using ICD-O and histology codes from the HFHS tumor<br />

registry. Eligible patients were newly diagnosed with GBM between 1997<br />

and 2008. Comprehensive, population-based data were compiled using<br />

tumor registry data (including histology and mortality) with linkages to<br />

pharmacy data (including infusion), outpatient and inpatient encounter<br />

data, and laboratory results. Results: Overall, 812 GBM patients were<br />

identified; mean age was 54 years (standard deviation 15.8). The cohort<br />

was 63% male and 84% white. Among the most common post-diagnosis<br />

comorbidities/complications, 14% <strong>of</strong> patients had venous thromboembolism<br />

(95% confidence interval [CI] 12-17), 16% had seizures (95% CI<br />

14-19), and 6% had central nervous system hemorrhage (95% CI 4-7).<br />

Arterial thromboembolism occurred in 3% <strong>of</strong> patients (95% CI 2-4), while<br />

7% experienced infections such as candidiasis, herpes simplex, and<br />

pneumocystis pneumonia (95% CI 5-9) and 2% experienced wound<br />

dehiscence (95% CI 1-3). 94% (95% CI 93-96) <strong>of</strong> the cohort received<br />

some form <strong>of</strong> treatment with 81% undergoing surgery (95% CI 78-84),<br />

75% receiving radiation therapy (95% CI 72-78), and 67% receiving<br />

chemotherapy (95% CI 64-70). Median overall survival after diagnosis was<br />

1.4 years (95% CI 1.3-1.5) among all patients, and 1.6 years (95% CI<br />

1.4-2.0) among those diagnosed 2005-2008. Conclusions: This study<br />

presents a well-characterized population-based cohort <strong>of</strong> GBM patients.<br />

Understanding treatment patterns and comorbidities outside the clinical<br />

trial setting is important in informing and evaluating real-world treatment<br />

decisions. Additional results exploring time trends in systemic cancer<br />

therapy and Kaplan-Meier survival curves will be presented.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


108s Cancer Prevention/Epidemiology<br />

1592 General Poster Session (Board #8B), Sat, 1:15 PM-5:15 PM<br />

Secondary breast cancer (s-BC) after hematopoietic cell transplantation<br />

(HCT) for leukemia: Role <strong>of</strong> total body irradiation (TBI). Presenting Author:<br />

Can-Lan Sun, City <strong>of</strong> Hope, Duarte, CA<br />

Background: s-BC after conventional chest radiation for histologically<br />

distinct cancer is well-recognized, and has resulted in radiologic screening<br />

recommendations by <strong>American</strong> Cancer <strong>Society</strong>. However, risk <strong>of</strong> s-BC after<br />

TBI is not well-defined. Methods: We determined the risk <strong>of</strong> s-BC in 648<br />

consecutive females transplanted for leukemia (ALL [19%], AML/MDS<br />

[57%], CML [24%]) between 1976 and 2008 with a post-HCT survival <strong>of</strong><br />

�2 years. The study was restricted to leukemia to understand the role <strong>of</strong><br />

TBI without the influence <strong>of</strong> pre-HCT chest radiation. Near-complete<br />

ascertainment <strong>of</strong> s-BC was assured by combining institutional follow-up<br />

with California Cancer Registry data. Results: Median age at HCT was 38<br />

years (range, 0-71); 14% had received autologous HCT, 64% related and<br />

22% unrelated donor HCT; 70% had received TBI for myeloablative<br />

conditioning. After 5,675 person-years <strong>of</strong> observation, 16 patients developed<br />

s-BC (14 after TBI). Median latency from HCT to s-BC was 16y<br />

(2-27). Median age at s-BC was 50y (29-66). Cumulative incidence <strong>of</strong> s-BC<br />

was 6% at 20y after HCT, contributed to largely by TBI (s-BC incidence<br />

after TBI: 5.7%). Among TBI-exposed, 8.6% were projected to develop<br />

s-BC by age 65y (1.6% among non-TBI-exposed, p�0.03). TBI-exposed<br />

patients were 2.4 times more likely to develop s-BC compared with general<br />

population (p�0.001); non-TBI exposed were not at increased risk<br />

(SIR�0.9, p�0.9). Multivariate analysis (adjusted for age at HCT, follow-up<br />

and primary diagnosis) revealed a 4.7-fold (p�0.068) increased<br />

risk <strong>of</strong> s-BC among TBI-exposed. Conclusions: Risk <strong>of</strong> s-BC after TBI as the<br />

sole source <strong>of</strong> radiation to breast is increased. Nearly 9% <strong>of</strong> those exposed<br />

to TBI will develop s-BC by age 65y. Identification <strong>of</strong> vulnerable subpopulations<br />

among the TBI-exposed patients is underway to create targeted<br />

screening strategies for early detection <strong>of</strong> s-BC.<br />

1594 General Poster Session (Board #8D), Sat, 1:15 PM-5:15 PM<br />

Association <strong>of</strong> a history <strong>of</strong> autoimmunity or hypersensitivity with overall<br />

survival in breast cancer. Presenting Author: Hakan Lars Olsson, Departments<br />

<strong>of</strong> Oncology and Cancer Epidemiology, Lund University, Lund,<br />

Sweden<br />

Background: Antibody therapy <strong>of</strong> malignant melanoma with ipilimumab is<br />

associated with the development <strong>of</strong> an autoimmune disease. The aim was<br />

to investigate if preexisting autoimmune disorders or hypersensitivities,<br />

similar to the side effects <strong>of</strong> immunotherapy in malignant melanoma, gave<br />

a better overall survival for breast cancer (BC) patients, compared with<br />

patients without these disorders. Methods: A consecutive clinical material<br />

consisting <strong>of</strong> 1,705 breast cancer patients diagnosed between 1980 and<br />

2010 was used. The patients were grouped according to preexisting<br />

autoimmune disease or hypersensitivities. The remaining BC patients were<br />

used as a reference group. All analyses were adjusted for age at diagnosis,<br />

T, N, M-status <strong>of</strong> the tumor, and ever-use <strong>of</strong> HRT simultaneously. A p value<br />

<strong>of</strong> less than 0.05 was considered significant. Results: One hundred and<br />

twenty-five (7.3%) patients had a history <strong>of</strong> autoimmunity and 72 (4.2%)<br />

patients had a history <strong>of</strong> hypersensitivity prior to BC diagnosis. Our main<br />

finding was that BC patients with a preexisting autoimmune disease or<br />

hypersensitivities with ER-negative tumors had a longer overall survival<br />

compared to patients without, HR 0.55, 95% CI 0.31-0.97. The risk was<br />

specifically low in BC patients with hypersensitivity and ER-negative<br />

tumors, HR 0.39, 95% CI 0.16-0.96. The risk was nonsignificantly lower<br />

in the autoimmune group, HR 0.78, 95% CI 0.37-1.61. Stratifying both on<br />

ER-status and menopausal status, similar results were seen in premenopausal<br />

BC patients with hypersensitivity. Postmenopausal BC patients with<br />

an ER-negative tumor and an autoimmune disease had a longer nonsignificant<br />

overall survival, HR 0.3, 95% CI 0.07-1.26. Conclusions: BC patients<br />

with an ER-negative tumor and/or diagnosed before menopause had a<br />

longer overall survival when previously diagnosed with hypersensitivity. For<br />

BC patients with an autoimmune disease, the prognostic benefit was seen<br />

when diagnosed postmenopausally with an ER-negative BC. Preexisting or<br />

induced autoimmunity or hypersensitivity may prolong life in breast cancer<br />

especially in young patients and those with ER-negative tumors.<br />

1593 General Poster Session (Board #8C), Sat, 1:15 PM-5:15 PM<br />

Effect <strong>of</strong> sunlight exposure on survival in patients with lymphoma: Results<br />

from the prospective Singapore Lymphoma Study. Presenting Author: Yin<br />

Leng Lee, National Cancer Center, Singapore<br />

Background: Recently, several studies evaluating the effect <strong>of</strong> ultraviolet<br />

light exposure on the risk <strong>of</strong> developing lymphoma have yielded conflicting<br />

results, in part due to seasonal and latitude variation. Further, most <strong>of</strong><br />

these studies also lack long term follow up and could not systemically<br />

address the influence <strong>of</strong> sunlight exposure on survival. In this prospective,<br />

epidemiological study, we explored the relationship between past sunlight<br />

exposure and survival in lymphoma patients in tropical Singapore, where<br />

sunlight exposure is constant throughout the year. Methods: Singapore<br />

Lymphoma Study was established in 2005. A total <strong>of</strong> 384 patients with<br />

histologically confirmed lymphoma were followed from date <strong>of</strong> diagnosis<br />

through November 2011. Interviewers were trained to conduct in-person<br />

interviews to collect detailed information, including every occupational<br />

period that lasted at least 1 year and number <strong>of</strong> hours worked outdoor from<br />

9am to 5pm using a structured questionnaire. Descriptive, multivariate and<br />

survival analysis (Kaplan Meier; age, gender, race and education level<br />

adjusted) were performed. Results: Among all lymphoma subtypes, the<br />

most prevalent was diffuse large B cell lymphoma, 47.7%, followed by<br />

Hodgkin lymphoma, 14.6%, follicular lymphoma, 12.2% and T cell<br />

lymphoma, 6.5%. 97.1% had black or brown eye color and 49.6% had fair<br />

skin. Overall survival <strong>of</strong> lymphoma and NHL subtype decreased significantly<br />

with increasing lifetime occupational hours (ptrend�0.01 and<br />

0.03). Chinese was associated with longer survival with increasing daily<br />

sunlight exposure during childhood and adolescent years, whether at<br />

school or outdoor leisure (ptrend�0.04) and there were no evidence for<br />

non-Chinese (Malays or Indians). Patients with lighter eye color results in<br />

shorter survival (HR�2.91, 95% CI: 1.15-7.34, p�0.02). Conclusions: We<br />

found leisure sunlight exposure during childhood and adolescent years to<br />

be beneficial to the survival <strong>of</strong> Chinese lymphoma patients and increasing<br />

lifetime cumulative occupational exposure is associated with increasing<br />

risk <strong>of</strong> death. This data contributes to the growing research on the effects <strong>of</strong><br />

sunlight exposure on lymphoma patients.<br />

1595 General Poster Session (Board #8E), Sat, 1:15 PM-5:15 PM<br />

The evolution <strong>of</strong> breast cancer screening in the Medicare population:<br />

<strong>Clinical</strong> and economic implications. Presenting Author: Brigid K. Killelea,<br />

Yale University School <strong>of</strong> Medicine, Yale Comprehensive Cancer Center,<br />

New Haven, CT<br />

Background: Breast screening has evolved as newer approaches to mammography,<br />

ultrasound, and MRI have diffused into clinical practice. The use <strong>of</strong><br />

these technologies and their impact on screening-related costs and<br />

outcomes remain undefined, particularly among older women. Methods:<br />

Using the Surveillance Epidemiology and End Results – Medicare linked<br />

database, we identified women aged 66 and older without a diagnosis <strong>of</strong><br />

breast cancer. We constructed two cohorts (2001 vs. 2006) and followed<br />

each for two years. We assessed changes in imaging technology, screeningrelated<br />

costs (defined as costs for screening and subsequent imaging and<br />

testing, adjusted to 2009 USD), and stage at diagnosis between the two<br />

cohorts. Results: There were 136,845 women in the 2001-2002 (earlier)<br />

cohort and 137,733 in the 2006-2007 (later) cohort. The mean age was<br />

76.9 and 77.2 respectively, (p�.001). The proportion <strong>of</strong> women receiving<br />

any screening mammogram was 42.5% in the earlier cohort and 43.4% in<br />

the later cohort, (p�.001). The use <strong>of</strong> digital mammography for screening<br />

increased from 2.2% to 15.0%, (p�.001). The use <strong>of</strong> any computer aided<br />

detection (CAD) increased from 3.2% to 29.3% (p�.001). MRI use<br />

increased from 0.03% to 0.2%, and ultrasound use from 4.0% to 4.5% (p<br />

�.001 for both). Average screening-related cost increased 31%, from<br />

$101 to $132 (p�.001). There was no significant difference in early stage<br />

at diagnosis over time (58.1% <strong>of</strong> women were in situ/stage I in early period<br />

vs. 57.2% in later period, p�.65). Conclusions: The use <strong>of</strong> digital<br />

mammography and CAD increased substantially between 2001 and 2007,<br />

contributing to a 31% increase in screening-related costs for women in the<br />

Medicare program. The increased cost <strong>of</strong> screening and downstream testing<br />

must be evaluated in context <strong>of</strong> an absence <strong>of</strong> benefit in terms <strong>of</strong> stage at<br />

diagnosis.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


1596 General Poster Session (Board #8F), Sat, 1:15 PM-5:15 PM<br />

ABO blood group and the risk <strong>of</strong> lung cancer: Multicenter, case-control,<br />

observational study. Presenting Author: Gungor Utkan, Ankara University<br />

School <strong>of</strong> Medicine, Ankara, Turkey<br />

Background: The role <strong>of</strong> genetic factors in the development <strong>of</strong> cancer is<br />

widely accepted. Previous studies have observed an association between<br />

ABO blood group and risk <strong>of</strong> certain malignancies, including pancreatic<br />

and gastric cancer. The data on the role <strong>of</strong> ABO blood group and Rh factor<br />

in lung cancer is limited. Methods: All patients who had Lung cancer (LC)<br />

and treated between 2000-2011 at the involved centers with defined<br />

ABO/Rh were included in our retrospective reviews <strong>of</strong> tumor registry<br />

records. A group <strong>of</strong> volunteer healthy blood donors <strong>of</strong> Turkish Red Crescent<br />

between 2004 and 2011 were identified as a control group. The relationship<br />

<strong>of</strong> ABO/Rh with clinical features such as age at diagnosis, histological<br />

subtype and sex were evaluated. We compared the distributions <strong>of</strong> ABO/Rh<br />

among 1954 patients and 3,022,883 controls. Among LC patients,<br />

differences between each <strong>of</strong> aforementioned ABO/Rh groups with respect<br />

to various clinical features were explored, respectively. Results: Of these<br />

patients the median age was 62 (range: 17-90). The 84% <strong>of</strong> patients were<br />

male. Overall distributions <strong>of</strong> ABO blood groups as well as Rh factor were<br />

statistically different between patients (43.6% A, 8.3% AB, 17.3% B,<br />

30.8% O, and 86.3% Rh�) and controls (42.2% A, 7.6% AB, 16.3% B,<br />

33.9% O, and 87.7% Rh�) (p�0,03). In addition, there were statistically<br />

significant differences between patients and controls with respect to O vs.<br />

nonO (p�0.004) and marginal significance for A vs. nonA (p�0,065), B vs.<br />

nonB (p�0,076), and Rh� vs. Rh- (p�0,057). Among patients, there<br />

weren’t statistically significant differences between blood group with<br />

respect to sex and age. However there was statistically significant difference<br />

between blood group with respect to histology (p�0,001). Although<br />

the distribution <strong>of</strong> A and O were similar according to histology, patients with<br />

squamous histology had antigen B more frequently than other histological<br />

sub types (p�0,009). Conclusions: In the study populations, ABO blood<br />

type was statistically significantly associated with the LC and having blood<br />

type other than O increases the risk <strong>of</strong> LC. Further studies are necessary to<br />

define the mechanisms by which ABO blood type may influence breast<br />

cancer risk.<br />

1598 General Poster Session (Board #8H), Sat, 1:15 PM-5:15 PM<br />

Analyzing excess mortality from cancer among individuals with mental<br />

illness. Presenting Author: Jackson S. Musuuza, Department <strong>of</strong> Population<br />

Health Sciences, University <strong>of</strong> Wisconsin School <strong>of</strong> Medicine and Public<br />

Health, Madison, WI<br />

Background: Previous studies have documented higher cancer-related<br />

mortality in individuals with mental illness, compared to their healthier<br />

counterparts. The aim <strong>of</strong> this study was to identify the anatomic cancer<br />

sites for which mortality is higher in individuals with mental illness.<br />

Methods: We used a linked dataset comprised <strong>of</strong> death certificate data for<br />

the state <strong>of</strong> Ohio for the years—2004-2007 and data from the publicly<br />

funded mental health system in Ohio. Decedents with mental illness were<br />

those identified concomitantly in both data sets. We used age-adjusted<br />

standardized mortality ratios (SMRs) in race- and sex-specific strata to<br />

estimate excess deaths for each <strong>of</strong> the anatomic cancer sites. Results:<br />

Overall, there was excess mortality from cancer associated with having<br />

mental illness in Non-Black men and women (SMR: 1.14 (95% confidence<br />

interval [CI] � 1.05-1.22), and 1.13 (95% CI� 1.05-1.20, respectively),<br />

but lower mortality in Black men, likely due to premature mortality from<br />

other causes (0.71 (95% CI� 0.61-0.83). In Black women, we observed<br />

no excess mortality from cancer that was associated with having mental<br />

illness. There was excess mortality from hepatobiliary cancer in Non-Black<br />

men (SMR� 1.65; 95% CI �1.15-2.29), but not in any other sex- and<br />

race- stratum. In addition, we observed excess mortality from lung and<br />

laryngeal cancers among Non-Black men and women (SMRs for lung<br />

cancer <strong>of</strong> 1.28 (95% CI� 1.13-1.45), and 1.45 (95% CI� 1.29-1.62),<br />

respectively; and for laryngeal cancer (2.01 (95% CI� 1.19-3.18 for men,<br />

and 2.47 (95% CI� 1.08-4.89) for women). Black women experienced<br />

excess mortality from cancer <strong>of</strong> the kidney and renal pelvis (SMR� 2.91;<br />

95% CI �1.53-5.06), while Non-Black women experienced excess mortality<br />

from urinary bladder cancers (SMR�1.89; 95% CI�1.15- 2.92).<br />

Conclusions: Compared to the general population in Ohio, individuals with<br />

mental illness experienced excess mortality from certain cancers. In part,<br />

this excess mortality may be mediated by a higher prevalence <strong>of</strong> smoking or<br />

substance abuse in individuals with mental illness. Further investigation is<br />

needed to explain the observed racial variation in cancer mortality, and to<br />

examine mortality for specific cancers based on severity <strong>of</strong> mental illness.<br />

Cancer Prevention/Epidemiology<br />

109s<br />

1597 General Poster Session (Board #8G), Sat, 1:15 PM-5:15 PM<br />

Association <strong>of</strong> p53 Arg72Pro polymorphism and HPV status with the<br />

initiation, progression, and development <strong>of</strong> cervical cancer (CC): A metaanalysis.<br />

Presenting Author: Steven Habbous, Princess Margaret Hospital,<br />

Toronto, ON, Canada<br />

Background: CC develops through progression from normal cervical epithelium<br />

through squamous intra-epithelial lesions (SILs) to cancer. CC is<br />

classically associated with oncogenic human papillomavirus (HPV). Yet,<br />

not all CC patients demonstrate HPV infection at diagnosis. HPV� and<br />

HPV– subsets <strong>of</strong> CC patients may thus represent distinct entities. HPV<br />

binds to P53, promoting its degradation; the Arg variant <strong>of</strong> p53 Arg72Pro<br />

binds more ardently to HPV than the Pro variant. Although there have been<br />

meta-analyses <strong>of</strong> Arg72Pro in CC risk, none has evaluated the relationship<br />

between Arg72Pro and HPV status together. We hypothesize that p53<br />

Arg72Pro modifies aspects <strong>of</strong> the carcinogenic process in HPV� (but not<br />

HPV-) subsets <strong>of</strong> CC. Methods: Pubmed/Embase databases identified 1494<br />

potential studies; 51 were eligible and included. Separate analyses were<br />

performed to evaluate CC risk (CC vs normal), initiation <strong>of</strong> cancer (SIL vs<br />

normal), and progression towards cancer (CC vs SIL), by HPV status. Pooled<br />

odds ratios (pOR) were generated using RevMan 5.1 s<strong>of</strong>tware. Results: p53<br />

Arg72Pro was not associated with CC risk in either HPV� or HPV- patient<br />

subsets (pORs 0.92; 95%CI, 0.6-1.3 and pORs 1.07; 95%CI, 0.7-1.7,<br />

respectively). Similarly, there was no association with the initiation <strong>of</strong><br />

disease (SIL vs normal): pORs 0.96; 95%CI, 0.7-1.3 (HPV� subset);<br />

1.11; 95%CI, 0.9-1.4 (HPV- subset). There was no association with<br />

disease progression from SIL to CC in the HPV- subset (pOR, 0.98; 95%CI,<br />

0.7-1.4). However, in the HPV� subset, comparing the progression from<br />

SIL through to CC, there was strong and significant association: pOR, 1.37;<br />

95%CI, 1.2-1.6 (p�4.0x10E-4), with no evidence <strong>of</strong> heterogeneity (I2 �<br />

0%) or bias (by funnel plot). Sensitivity analyses demonstrated similarly<br />

significant results, even after taking into account differing quality <strong>of</strong><br />

methodological designs and type <strong>of</strong> biological sample analyzed (tumour vs.<br />

blood) <strong>of</strong> the underlying studies. Associations were mainly restricted to<br />

Caucasian studies. Conclusions: In this meta-analysis, the Arg variant <strong>of</strong><br />

p53 Arg72Pro was associated with progression <strong>of</strong> SIL to CC only in HPV�<br />

subsets, but not to disease initiation or risk <strong>of</strong> CC.<br />

1599 General Poster Session (Board #9A), Sat, 1:15 PM-5:15 PM<br />

Analysis <strong>of</strong> KRAS and BRAF mutant colorectal cancers in a multiracial<br />

population. Presenting Author: Jared David Acoba, University <strong>of</strong> Hawaii<br />

Cancer Center, Honolulu, HI<br />

Background: We previously demonstrated racial disparities in colorectal<br />

cancer (CRC) survival despite adjustments for tumor and socioeconomic<br />

factors. Molecular differences in breast and lung tumors contribute to<br />

racial survival inequality, yet in CRC, molecular tumor characteristics have<br />

not been studied extensively in a racially diverse cohort. We performed a<br />

comprehensive evaluation <strong>of</strong> KRAS and BRAF mutations in a multiracial<br />

population to determine the prevalence in CRC and the degree to which<br />

these molecular traits impact survival. Methods: A retrospective cohort<br />

study <strong>of</strong> patients diagnosed with CRC between 2008 and 2011 from<br />

hospital tumor registries was performed. The prevalence <strong>of</strong> KRAS and<br />

BRAF mutations was determined for the study population and individual<br />

racial groups. Multivariable Cox proportional hazards regression models for<br />

survival were built for KRAS and BRAF mutation status while adjusting for<br />

age at diagnosis, race, and stage <strong>of</strong> disease. Results: Of 706 patients<br />

diagnosed with CRC, KRAS mutational analysis was performed on 148<br />

subjects. 14% <strong>of</strong> subjects were white (W), 64% Asian (A), and 21% Native<br />

Hawaiian/Pacific Islander (NH). KRAS mutation was identified in 48<br />

subjects (32%). The prevalence <strong>of</strong> mutant tumors among racial groups was<br />

W 33%, A 36%, and NH 30%. Analysis for KRAS G13D mutations revealed<br />

a prevalence <strong>of</strong> W 11%, A 9%, and NH 7%. When compared to published<br />

datasets <strong>of</strong> predominantly white patients, our multiracial cohort had a<br />

significantly higher rate <strong>of</strong> KRAS G13D mutant tumors, p�0.039. Of 74<br />

subjects tested for the BRAF mutation, two mutant tumors were detected<br />

(3%). The prevalence <strong>of</strong> the BRAF mutation by race was 10% W, 3% A, and<br />

0% NH (p�0.18). BRAF and KRAS G13D mutations were adverse<br />

prognostic factors in a multivariate analysis, although the odds ratios failed<br />

to meet statistical significance. Conclusions: The prevalence <strong>of</strong> BRAF and<br />

KRAS mutations in this multiracial cohort is similar to what has been<br />

previously reported. However the rates <strong>of</strong> KRAS G13D and BRAF mutant<br />

tumors in our cohort are higher than prior reports. Furthermore, KRAS<br />

G13D which has been postulated to be a favorable prognostic factor for<br />

CRC, may adversely impact survival <strong>of</strong> minority patients.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


110s Cancer Prevention/Epidemiology<br />

1600^ General Poster Session (Board #9B), Sat, 1:15 PM-5:15 PM<br />

Correlation <strong>of</strong> high body mass index and circulating tumor cell positivity in<br />

patients with early-stage breast cancer. Presenting Author: Uta Ortmann,<br />

Heinrich Heine University , Duesseldorf, Germany<br />

Background: The prognostic relevance <strong>of</strong> both body mass index (BMI) and<br />

circulating tumor cells (CTC) has been confirmed in different trials for<br />

patients with early breast cancer. This analysis evaluates the correlation<br />

between high BMI and CTC positivity as risk factors for reduced disease free<br />

and overall survival. Methods: Data <strong>of</strong> 3658 patients <strong>of</strong> the SUCCESS A trial<br />

have been analyzed. CTC count and BMI were documented before (N �<br />

2026) and after (N � 1504) chemotherapy. Within this trial patients with<br />

early breast cancer were randomized to two chemotherapy regimens and<br />

received either 3 cycles <strong>of</strong> fluorouracil, epirubicin and cyclophosphamide<br />

followed by 3 cycles <strong>of</strong> docetaxel (FE100C-Doc) or 3 cycles <strong>of</strong> fluorouracil,<br />

epirubicin and cyclophosphamide followed by 3 cycles <strong>of</strong> docetaxel and<br />

gemcitabine(FE100C-DG). In addition, patients were randomized to zoledronic<br />

acid either for 2 or 5 years. CTC were analyzed using the CellSearch<br />

System (Veridex, USA). Different groups <strong>of</strong> bodyweight were classified<br />

according to the WHO’s international definition: Underweight (BMI � 18,5<br />

kg/m2 ), normal range (BMI � 18,5- � 25), overweight (BMI �25- � 30),<br />

obesity (BMI � 30). Correlation between CTC count and BMI was analyzed<br />

using frequency-table methods. Results: At study entry 24 (1.2%) patients<br />

were underweight, 952 (47%) patients were normal weight, 658 (32.5%)<br />

patients were overweight and 392 (19.4%) patients were obese. Before the<br />

start <strong>of</strong> chemotherapy, CTC were detected in 435 (21.5%) patients. We did<br />

not find a correlation between CTC positivity and BMI (p�0.94). After<br />

chemotherapy CTC were detected in 330 (16,3%) patients. Again, there<br />

was no statistically significant correlation between BMI and CTC positivity<br />

(p�0.86). In particular, CTC positivity was not observed more frequently in<br />

obese patients neither before (p�0.70) nor after chemotherapy (p�0.95)<br />

compared to patients with a BMI � 30 kg/m2 . Conclusions: As compared to<br />

patients with normal BMI, there was no significant difference in the<br />

prevalence <strong>of</strong> CTC in underweight, overweight and obese patients, respectively,<br />

neither before nor after chemotherapy. The risk factors obesity and<br />

prevalence <strong>of</strong> CTC seem to be independent prognostic factors.<br />

1602 General Poster Session (Board #9D), Sat, 1:15 PM-5:15 PM<br />

Correlation <strong>of</strong> BMI with tumor stage in early breast cancer patients (pts):<br />

Pooled analysis <strong>of</strong> the German SUCCESS A, B, and C trials. Presenting<br />

Author: Carola Anna Melcher, Department <strong>of</strong> Gynecology and Obstetric,<br />

Heinrich Heine University, Duesseldorf, Germany<br />

Background: Independent from known prognostic factors, e.g., tumor size<br />

and nodal status, obesity is a risk factor for poor disease free, distant<br />

disease free, and overall survival in breast cancer. The aim <strong>of</strong> this analysis<br />

was to examine the correlation <strong>of</strong> the body mass index (BMI) with tumor<br />

characteristics in early breast cancer. Methods: We analyzed the data <strong>of</strong><br />

7,997 pts with early, node positive or high risk node negative primary<br />

breast cancer treated with adjuvant taxan-based chemotherapy within the<br />

German multicenter phase III SUCCESS A, B, or C trials. The pts’ tumor<br />

stage at primary diagnosis was classified according to the UICC tumor-nodemetastasis<br />

(TNM) classification. Additionally, the tumor’s hormonereceptor<br />

status and HER2/neu status were determined. Before enrollment<br />

into the study each patient was grouped according to the WHO global<br />

database on BMI. Contingency table methods were used to analyze the<br />

correlation <strong>of</strong> BMI and tumor characteristics. Results: Among the 7,997 pts<br />

100 (1.3%) pts were underweight, 3,556 (44.5%) pts were normal weight,<br />

2,569 (32.1%) pts were overweight and 1,772 (22.2%) were obese. Of all<br />

pts 4,508 pts (56.4%) suffered from a pT2-4 tumor, 4830 (60.4%)<br />

showed lymph node involvement (pN1-3) and 7509 (93.9%) had G2-3<br />

tumors. 5839 pts (73.0%) showed positivity for ER or PR and 935 (11.7%)<br />

for HER2/neu. Overweight and obese pts had significantly larger tumors<br />

compared to pts with normal BMI (p�0.0001; p�0.0001). Furthermore,<br />

overweight and obesity were associated with a significantly higher rate <strong>of</strong><br />

lymph node involvement (p�0.0001; p�0.0003) respectively. In contrast<br />

neither grading, tumor histology, ER/PR-status nor HER2/neu-overexpression<br />

were correlated with BMI. Conclusions: These data are the first to show<br />

in a large number <strong>of</strong> pts that both obese and overweight women suffering<br />

from primary breast cancer have significantly larger tumors and more <strong>of</strong>ten<br />

positive axillary lymph nodes. As there are no differences in tumor biology,<br />

the advanced tumor stage might be due to more difficult and delayed<br />

detection <strong>of</strong> breast cancer and lymph node lesions in these women.<br />

1601 General Poster Session (Board #9C), Sat, 1:15 PM-5:15 PM<br />

Evolution and landscape <strong>of</strong> clinical trials for breast cancer patients.<br />

Presenting Author: Semih Dogan, Institut Gustave Roussy, Villejuif, France<br />

Background: Recent advances in cancer research are expected to have<br />

dramatically changed the way clinical trials are being done. In an effort to<br />

outline new trends <strong>of</strong> clinical research in the breast cancer area, we have<br />

evaluated the evolution <strong>of</strong> clinical trials started between 2006 and 2011.<br />

Methods: 622 clinical trials, started during the first semester 2006, the<br />

second semester 2008 and the first semester 2011, were analyzed. The<br />

data source was <strong>Clinical</strong>trials.gov. 30 items were included in the database.<br />

Results: The overall number <strong>of</strong> patients included in prospective clinical<br />

trials increased over these periods (n�67,820, n�91,429, n�98,417).<br />

However, when large epidemiological cohorts are excluded, a significant<br />

fall in the number <strong>of</strong> patients is seen (n�67,820, n�44,554, n�37,417).<br />

The absolute number <strong>of</strong> therapeutic trials also decreased during this time<br />

(n�93, n�99, n�78), mainly related to the dramatic fall in the number <strong>of</strong><br />

trials testing conventional treatments (n�40, n�24, n�20). In the<br />

meantime, the number <strong>of</strong> trials testing targeted agents remained similar<br />

(n�46, n�67, n�50). Interestingly, in the same time, the number <strong>of</strong> trials<br />

testing a targeted agent in a population defined by a biomarker increased<br />

(n�1, n�6, n�14). At the opposite, prospective studies aimed at<br />

validating biomarkers did not increase, and remained only driven by large<br />

consortiums. As illustration, when we exclude RxPONDER trial, the<br />

biomarker-validation studies opened in 2011 planned to include only 2<br />

493 patients, representing 2% <strong>of</strong> the breast cancer research field.<br />

Prospective trials testing social sciences and supportive care (n� 70,<br />

n�72, n�70) tend to become as important as therapeutic trials. Finally,<br />

large epidemiological cohorts opened in 2011 will represent more than<br />

60% <strong>of</strong> the patients included in clinical trials. Conclusions: The part <strong>of</strong><br />

cohorts and social sciences in breast cancer research is increasing and<br />

represents now the majority <strong>of</strong> the clinical research activity. When the<br />

analysis focuses on therapeutic trials, the breast cancer field recently<br />

shifted to targeted agents and early data suggest that therapeutic research<br />

is increasingly incorporating companion biomarker. Analysis on all 2,762<br />

clinical trials done between 2006 and 2011 will be presented.<br />

1603 General Poster Session (Board #9E), Sat, 1:15 PM-5:15 PM<br />

Effect <strong>of</strong> gender on age-specific effects for ulcerated malignant melanomas.<br />

Presenting Author: Blakely S. Richardson, University Hospitals Case<br />

Medical Center/Case Western Reserve University School <strong>of</strong> Medicine,<br />

Cleveland, OH<br />

Background: Gender, age at diagnosis, and ulceration are all independent<br />

prognostic factors for melanoma. The purpose <strong>of</strong> this study was to further<br />

examine the interactions among gender, age, and melanoma ulceration.<br />

Methods: Using the NCI’s SEER 17 Registries Database (2004-2008), we<br />

assessed ulceration status among White men and women with malignant<br />

melanoma, stratified by younger (ages 10-39 years) and older (ages 40-84<br />

years) ages at diagnosis as well as by tumor depth. The analysis was<br />

restricted to melanomas common to both age groups, i.e. superficial<br />

spreading, nodular, and unclassified melanomas. There were 2905 older<br />

men, 301 younger men, 1624 older women and 276 younger women that<br />

fit our criteria. Relative risks were expressed as incidence rate ratios (IRR)<br />

with 95% confidence intervals (CI). Results: At every tumor depth, IRRs for<br />

ulcerated melanomas were significantly higher for older than younger<br />

persons (IRR�1.0). The trend, however, was different in men as compared<br />

to women. In men, the IRR (older to younger) rose continuously with<br />

increasing depth [�1.0mm: IRR 7.81 (CI 6.18-9.98), 1.01mm-�2.0mm:<br />

IRR 9.71 (CI 7.55-12.68), 2.01mm-�4.0mm: IRR 11.04 (CI 8.78-<br />

14.06); and �4.01mm: IRR 12.26 (CI 9.60-15.87)]. On the other hand,<br />

in women, IRR (older to younger) was stable for all depths �4.0mm then<br />

nearly doubled for melanomas �4.01mm [�4.01mm: IRR 8.00 (CI<br />

5.85-11.20)]. The IRR trend analysis was statistically significant for men<br />

(p � 0.01) but not for women (p � 0.34). Conclusions: Our study confirms<br />

that older age at diagnosis is associated with higher incidence rates <strong>of</strong><br />

ulcerated malignant melanomas. This large-scale population-based analysis<br />

also shows an effect modification by gender and tumor thickness,<br />

suggesting different biologic behavior in melanoma in younger and older<br />

men and women. Future studies should further investigate tumor biology<br />

differences in these populations and the interactions among gender, age at<br />

diagnosis, and ulceration in melanoma.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


1604 General Poster Session (Board #9F), Sat, 1:15 PM-5:15 PM<br />

Long-term survival and prognostic factors in neuroendocrine tumors:<br />

Results from a large multicenter study. Presenting Author: Antongiulio<br />

Faggiano, Department <strong>of</strong> Molecular and <strong>Clinical</strong> Endocrinology and Oncology,<br />

Federico II University, Naples, Italy<br />

Background: The clinical behavior <strong>of</strong> neuroendocrine tumors (NET) is highly<br />

variable, but the only data available in literature on large series are mainly<br />

based on retrospective register data collected in 3-5 decades, focusing on<br />

short-term follow-up. These are clearly insufficient due to the frequent<br />

evolution in classification criteria, diagnostic techniques and the scarcity <strong>of</strong><br />

clinical data. Methods: Data from a total <strong>of</strong> 1659 NET patient were<br />

collected from 12 Italian referral centers. A specific s<strong>of</strong>tware was developed<br />

for the study. Only patients with histological diagnosis revised<br />

according to the WHO Classification in active long term follow-up in the<br />

period 1990-2010 were included in the study. Patients with incomplete<br />

diagnostic work-up, without clinical data or lost at follow-up were excluded.<br />

Cumulative survival were analyzed according to site <strong>of</strong> origin, histotype,<br />

grading, ki67 score, secreting pattern (functioning or not functioning) and<br />

staging. Results: At 2 yr cumulative survival do not differ between<br />

pancreatic (pNET 94.9%), gastrointestinal (GINET 94.4%) and lung NET<br />

(TNET 94.6%), functioning (93.5 %) and non functioning (93.9%), while<br />

was significantly different between T1N0M0 (98%), T1N1M0 (92.9%),<br />

T1N1M1 (90.2%) and grading (G1 98%, G2 96.8%, G3 84.1%). At 5, 10<br />

and 15 yr, cumulative survival were respectively 90.9, 77.1, 62.7% for<br />

pNET, 88.7, 83.5, 62.7% for GINET 90.3, 80.2, 67.9% for TNET. 88.4,<br />

77.0, 59.0% in functioning and 89.8, 79.1, 64.6% in not functioning.<br />

96.1, 89.5 and 77.0% for T1N0M0, 88.2, 83.5, 70.4 for T1N1M0 and<br />

82.0, 55.8 and 43.9 for T1N1M1, 93.1, 82.3, 75.4% for G1, 87.8, 64.8,<br />

48.6% for G2, 81.4, 75.2, 40.1% for G3. Conclusions: Brief (2y) and very<br />

long term (15y) survival do not significantly differ between different site <strong>of</strong><br />

NET origin. Functioning tumors have the same survival rate at 2 y but lower<br />

at 15 y. Staging and grading appear as the most significant prognostic<br />

factors particularly at 10 and 15 y. Long-term cumulative survival in NET<br />

results significantly higher than in historical series, probably due to the<br />

anticipation <strong>of</strong> the diagnosis and the availability <strong>of</strong> new therapeutic<br />

strategies.<br />

1606 General Poster Session (Board #9H), Sat, 1:15 PM-5:15 PM<br />

Breast cancer survival: Is the Asian population homogenous? Presenting<br />

Author: Vincent Caggiano, Sutter Institute for Medical Research, Sacramento,<br />

CA<br />

Background: Study <strong>of</strong> the ER-/PR-/HER2- (TN) subtype has generated<br />

interest in race/ethnicity with respect to breast cancer. Poor survival <strong>of</strong><br />

African-<strong>American</strong> (AA) women with breast cancer has been reported. Less<br />

is known about the Asian population. This study examines survival <strong>of</strong> four<br />

ER/PR/HER2 subtypes among 10 self-reported race/ethnicity categories<br />

Methods: Using the California Cancer Registry 2000-2010, we examined<br />

136,175 cases first primary female invasive breast cancer. For stages 1<br />

and 2, Kaplan-Meier survival and the Log-Rank test were computed by<br />

stage and race for the ER�/PR�/HER2-, ER-/PR-/HER2�, ER�/PR�/<br />

HER2� (TP) and TN subtypes. Results: When the Asian population is<br />

combined into the category Asian/Pacific Islander (API): ER�/PR�/<br />

HER2-: AAs had worse 5-year survival than whites in stages 1 and 2 but<br />

Hispanics and APIs had better survival in both stages; ER-/PR-/HER2�:<br />

AAs and Hispanics were no different from whites in stage 1 but APIs had<br />

better 5-year survival (0.96 vs 0.89, p�0.001). For stage 2, there were no<br />

differences in survival among the races. ER�/PR�/HER2�: AAs had worse<br />

and APIs had better survival in both stages; Hispanics had better survival<br />

only in stage 1 (0.93 vs 0.92, p�0.03) and were no different from whites in<br />

stage 2. ER-/PR-HER2-: AAs and Hispanics were no different from whites<br />

in stage 1 but APIs had better survival (0.93 vs 0.88, p� .003); AAs had<br />

worse survival than whites in stage 2 (0.71 vs 0.75, p� 0.04) Hispanics<br />

were no different, and APIs had better survival (0.81, p � 0.001). When<br />

the Asian population was expanded into 7 categories, Pacific Islanders and<br />

Koreans were no different than whites in stages 1 or 2 for all four subtypes<br />

examined. For Stage 1, Southeast Asians had better survival for all<br />

subtypes except for the TN whereas Filipinos had better survival for all<br />

subtypes except for TP. For the ER�/PR�/HER2- subtype, women from the<br />

Indian Continent had better survival in stage 1 (0.98 vs 0.92, p�0.003)<br />

and in stage 2 (0.94 vs 0.88, p�.001) and better survival for stage 1 <strong>of</strong> the<br />

TN subtype (1.00, vs 0.88, p �0.04). Conclusions: There is heterogeneity<br />

among the Asian population, but regardless <strong>of</strong> how categorized, Asians<br />

have either the same or better 5-year survival than whites for all subtypes in<br />

stages 1 and 2.<br />

Cancer Prevention/Epidemiology<br />

111s<br />

1605 General Poster Session (Board #9G), Sat, 1:15 PM-5:15 PM<br />

Survival <strong>of</strong> elderly Medicare patients after standard chemotherapy for<br />

advanced lung and gastrointestinal cancers in the real world. Presenting<br />

Author: Elizabeth B. Lamont, Massachusetts General Hospital Cancer<br />

Center, Boston, MA<br />

Background: Elderly cancer patients are under-represented on clinical trials<br />

that determine standards <strong>of</strong> treatment, thus survival <strong>of</strong> such patients<br />

following standard treatment in the real world is not known. We describe<br />

unadjusted survivals for site, stage, and treatment-specific cohorts <strong>of</strong><br />

elderly Medicare cancer patients who were treated in usual care settings.<br />

Methods: From SEER-Medicare data, we identified elderly Medicare patients<br />

with advanced lung or GI cancers who received specific standard<br />

chemotherapy regimens within six months <strong>of</strong> diagnosis. Of the 108,386<br />

patients with the cancer sites and stages <strong>of</strong> interest, 39% (42,570/<br />

108,386) received some form <strong>of</strong> chemotherapy within six months <strong>of</strong><br />

diagnosis. Only 32% (13,689/42,570) received one <strong>of</strong> the specific<br />

standard regimens we studied. Results: Median survival times and interquartile<br />

ranges (IQRs) varied according to cancer site, stage, and treatment. For<br />

598 stage IV CRC patients on FOLFOX, the median survival was 19.4 mos<br />

(IQR 9.3-43.0); this is comparable to PRIME Study patients (19.7 mos).<br />

For 130 stage IV CRC patients on FOLFIRI, the median survival was 16.1<br />

mos (IQR 5.6-30.0), which is lower than BICC-C Study patients (23.1<br />

mos). For 3,815 advanced pancreatic cancer patients on gemcitabine, the<br />

median survival was 4.3 mos (IQR 2.0-8.7), which is slightly lower than<br />

CALGB 80303 patients (5.8 mos). For 8,040 stage IV NSC lung cancer<br />

patients on carboplatin and paclitaxel, the median survival was 7.0 mos<br />

(IQR 3.2-14.2); this is comparable to ECOG 1594 patients (7.8 mos). For<br />

1,104 extensive stage SCLC patients on cisplatin and VP16, the median<br />

survival was 8.5 mos (IQR 4.7-12.9), which is slightly lower than CALGB<br />

9732 patients (10.0 mos). Conclusions: The observed survival <strong>of</strong> cancer<br />

site, stage, and treatment-specific cohorts <strong>of</strong> elderly Medicare patients with<br />

advanced lung or GI cancers who were treated in real world settings with<br />

standard chemotherapy was, in general, similar to that <strong>of</strong> patients treated<br />

with nominally identical regimens on trials. Further analyses should explore<br />

the shorter survival for elderly Medicare patients with stage IV CRC treated<br />

with FOLFIRI in the usual care setting compared to patients treated on<br />

trials.<br />

1607 General Poster Session (Board #10A), Sat, 1:15 PM-5:15 PM<br />

Impact <strong>of</strong> obesity and overweight in the prognosis <strong>of</strong> women diagnosed with<br />

non metastatic breast cancer in a Mexican cohort. Presenting Author: José<br />

Luis Aguilar, Instituto Nacional de Cancerología, Mexico City, Mexico<br />

Background: Mexico positions right up at the top with U.S. in worldwide<br />

rankings <strong>of</strong> the most obese countries. In addition, breast cancer (BrCa) is<br />

the main type <strong>of</strong> cancer among women in this country. Studies have shown<br />

inconsistent results regarding obesity as a prognostic factor for worse<br />

outcome. Methods: Our aim is to identify if overweight and obesity confer<br />

poor prognosis in non-metastasic BrCa patients (pts). We identified 1799<br />

Hispanic women with newly diagnosed BrCa who attended the National<br />

Cancer Institute in Mexico from 2004-2008 and compared clinical and<br />

pathological features and overall survival (OS) between pts with a body<br />

mass index (BMI) � or � than 25. Results: The median age at diagnosis was<br />

51 years. A BMI�25 was found in 71% <strong>of</strong> pts. Postmenopausal women<br />

comprised 52%, and had a greater proportion <strong>of</strong> cases with a BMI�25 than<br />

premenopausal pts (75% vs. 67%, p�0.0001). Pts with BMI�25 presented<br />

with more advanced TNM stages and nodal involvement than their<br />

counterparts (73% vs. 67%, p�0.005 and 76% vs. 71%, p�0.017;<br />

respectively). Overall prevalence <strong>of</strong> hormone-receptor (HR), triple-negative<br />

(TN) and HER2 positive disease was 62%, 23%, and 27%, respectively.<br />

Differences according to receptor status between pre and postmenopausal<br />

pts and BMI are shown in table. There was no difference in disease-free<br />

survival and OS according to overweight and obesity in the overall<br />

population, but when menopausal status was considered, premenopausal<br />

pts with BMI�25 had a worse OS compared to pts with BMI�25 (HR 1.6,<br />

p�0.037). This difference was not seen in the postmenopausal group.<br />

Conclusions: Obesity may influence BrCa outcomes via several hormonal<br />

and inflammatory mechanisms. In this study, overweight and obesity confer<br />

a poor prognosis in premenopausal patients, possibly related to excess<br />

estrogen availability and higher prevalence <strong>of</strong> TN BrCa. Therefore, overweight<br />

and obesity deserve additional attention to assess possible causal<br />

relationships that potentially could be modified to improve outcomes in<br />

premenopausal patients.<br />

Premenopausal Postmenopausal<br />

25 (%) p 25 (%) p<br />

HR - 34 41 0.028 45 34 0.003<br />

� 66 59 55 66<br />

TN - 79 73 0.043 76 81 0.079<br />

� 22 27 24 1<br />

HER2 - 71 76 0.094 65 73 0.010<br />

� 29 24 35 27<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


112s Cancer Prevention/Epidemiology<br />

1608 General Poster Session (Board #10B), Sat, 1:15 PM-5:15 PM<br />

<strong>American</strong> BRCA outcomes and utilization <strong>of</strong> testing (ABOUT) study.<br />

Presenting Author: Rebecca Sutphen, Epidemiology Center, University <strong>of</strong><br />

South Florida Morsani College <strong>of</strong> Medicine, Tampa, FL<br />

Background: An estimated 100,000 individuals currently undergo genetic<br />

testing for hereditary susceptibility to breast and ovarian cancer annually in<br />

the U.S., yet little is known about their characteristics, testing experience<br />

or outcomes. Research in this high-risk group has been limited to patients<br />

recruited at academic medical centers where case ascertainment, health<br />

services delivery, decision-making and, quite likely, outcomes are different<br />

from those in the community setting where the majority <strong>of</strong> individuals<br />

currently receive healthcare services. Methods: Eligible subjects include<br />

10,000 consecutive individuals requesting BRCA testing through the<br />

nation’s third-largest health insurer, Aetna, beginning in December, 2011.<br />

De-identified data are analyzed from each test request form submitted by<br />

the ordering provider. Each eligible subject is mailed a study packet<br />

inviting them to complete a questionnaire (by mail, online or telephone)<br />

designed to investigate informational and healthcare services, test results,<br />

knowledge, risk perception and medical intentions. Results: Of 442<br />

subjects contacted during the first two weeks <strong>of</strong> accrual, 143 (32%) have<br />

completed the questionnaire to date. Similar to Aetna member demographics,<br />

7% are African-<strong>American</strong> and 7% report Hispanic ethnicity. Based on<br />

Chi-Square tests, there were no differences between respondents and<br />

non-respondents with regard to age (51% under age 50), race, ethnicity or<br />

personal history <strong>of</strong> cancer (58%). Among respondents, deleterious mutations<br />

were identified in 8%. Testing for a known familial mutation was<br />

performed in 8%. Among women with breast cancer, 13% were undergoing<br />

testing prior to initial surgical treatment. Updated results from 3000<br />

eligible subjects will be presented. Conclusions: This innovative, academicindustry<br />

collaboration enables an unprecedented investigation <strong>of</strong> significant<br />

issues surrounding individuals at increased risk for hereditary breast<br />

and ovarian cancer and undergoing genetic testing in the U.S. The results<br />

will guide the development and dissemination <strong>of</strong> more targeted decisionsupport<br />

tools and strategies to improve medical outcomes for such<br />

individuals.<br />

1610 General Poster Session (Board #10D), Sat, 1:15 PM-5:15 PM<br />

Health and quality <strong>of</strong> life (QOL) issues in four groups <strong>of</strong> lung cancer<br />

patients: Low-dose computed tomography (LDCT) screened, chest x-ray<br />

detected, incidentally found, and routinely diagnosed. Presenting Author:<br />

Ping Yang, Mayo Clinic, Rochester, MN<br />

Background: Low dose computed tomography (LDCT) scans have reduced<br />

lung cancer deaths by 20.3% in high risk populations, although there is an<br />

unknown balance between the benefits and harms <strong>of</strong> LDCT scans as a<br />

screening tool. Our purpose was to compare health-related QOL issues<br />

among lung cancer patients who were initially detected by LDCT scans; 4<br />

comparison groups included: lung cancer diagnosed by a screening chest<br />

X-ray, as an incidental finding from procedures taken for other medical<br />

reasons, or based on symptoms indicative for lung cancer and routinely<br />

diagnosed, and individuals who were LDCT screened but found no lung<br />

cancer (controls who participated in Mayo’s lung cancer CT screening trial).<br />

Methods: A total <strong>of</strong> 1,658 lung cancer patients (cared at Mayo Clinic) in the<br />

4 groups (37, 151, 389, and 1081 respectively) and 488 controls were<br />

compared on following patient-reported outcomes (collected via validated<br />

tools): overall QOL, four symptoms (cough, pain, dyspnea, fatigue), mental/<br />

physical/ emotional/ social/ spiritual QOL, and other concerns (e.g., family/<br />

friends/ financial/ legal). A clinically significant deficit was defined as at<br />

least 10-points in difference (or �50 points) on a 0-100 scale. The rates <strong>of</strong><br />

deficits were compared via Fisher’s exact tests and average QOL values via<br />

Kruskal-Wallis tests. Results: Overall QOL and individual symptoms were<br />

significantly worse (p�0.05) in all lung cancer groups than in controls,<br />

except for pain. LDCT-screened patients reported the greatest deficit<br />

among the 4 lung cancer groups in physical (41%), emotional (24%),<br />

social (38%), and spiritual QOL (24%); whereas chest X-ray detected<br />

patients had the least deficit in overall QOL (22%) and pain (32%). All 4<br />

lung cancer groups experienced much worse fatigue (52-64%) than the<br />

controls (32%). Conclusions: Our preliminary results suggest that LDCTscreening<br />

detected lung cancer patients reported a different QOL pr<strong>of</strong>ile<br />

from other lung cancer patients and non-lung cancer controls. The clinical<br />

course, smoking behavior, and QOL related health issues associated with<br />

LDCT screening for lung cancer warrant thorough investigation.<br />

1609 General Poster Session (Board #10C), Sat, 1:15 PM-5:15 PM<br />

Association between age, gender, residential region, and prevalence <strong>of</strong><br />

subependymal giant cell astrocytoma among patients with tuberous sclerosis<br />

complex: A U.S. Healthcare Claims Database study. Presenting Author:<br />

Michael Kohrman, University <strong>of</strong> Chicago, Chicago, IL<br />

Background: Subependymal giant-cell astrocytomas (SEGA) are slow growing<br />

non-cancerous tumors that can occur in patients with tuberous sclerosis<br />

complex (TSC). The objectives <strong>of</strong> this study were to examine SEGA<br />

prevalence among TSC patients in a real world setting, and to ascertain the<br />

relationships between SEGA prevalence and age groups, genders, or<br />

residential regions. Methods: We conducted a retrospective cohort study<br />

based on a large US commercial healthcare claims database. Patients with<br />

a TSC claim between 2000 and 2009 and with 12-month continuous<br />

enrollment after their first TSC claim were included into the study. SEGA<br />

was identified based on ICD-9CM codes in inpatient or outpatient claims.<br />

Patients’ genders, residential regions and ages <strong>of</strong> their first TSC claim and<br />

/or their first SEGA claim (if they had SEGA) were extracted from enrollment<br />

files respectively. SEGA prevalence was assessed with subgroup analyses <strong>of</strong><br />

genders, age groups <strong>of</strong> first TSC claim, and residential regions. Two-sample<br />

t tests and Chi-square tests were used to explore the relationships between<br />

SEGA prevalence and age groups, genders, or residential regions. Results:<br />

The study had 2,767 patients with a mean age <strong>of</strong> 28.5 years on their first<br />

observed TSC claim, and 55.3% were female. Among these TSC patients,<br />

7.7% had SEGA with variations by age groups <strong>of</strong> their first TSC claim (5.9%<br />

for age�1 year, 12.6% for age 1-5 years, 11.0% for age 6-10 years, 11.0%<br />

for age 11-18 years, 7.6% for age 19-25 years, and 5.1% for age 26 years<br />

or more), by genders (8.7% for males vs. 6.9% for females), and by<br />

residential regions (6.0%-11.0%). The associations between SEGA prevalence<br />

and age groups, genders or residential regions were all statistically<br />

significant with p�0.05. Conclusions: In a real world setting, and by the<br />

end <strong>of</strong> the first post-TSC year, 7.7% TSC patients had an observed SEGA<br />

diagnosis. This prevalence varied by age groups, genders, residential<br />

regions. Further research is needed to explore the factors that results into<br />

these variations.<br />

1611 General Poster Session (Board #10E), Sat, 1:15 PM-5:15 PM<br />

A retrospective analysis <strong>of</strong> all non-AIDS defining cancers (NADC): A subset<br />

analysis <strong>of</strong> the County Hospital AIDS Malignancy Project (CHAMP).<br />

Presenting Author: Shweta Gupta, John H. Stroger Jr. Hospital <strong>of</strong> Cook<br />

County, Chicago, IL<br />

Background: County Hospital (CCH) with its HIV clinic, the CORE Center<br />

(CC) is the largest provider for HIV patients (pts) in Chicago, treating over<br />

5,500 HIV pts yearly. There is paucity <strong>of</strong> data on characteristics <strong>of</strong> HIV�<br />

cancers (ca) in the inner city. The CHAMP cohort is a retrospective study <strong>of</strong><br />

all HIV associated cancers at CC and CCH over past 14 years (yrs). We<br />

analyzed all <strong>of</strong> the NADC from this cohort. Methods: All HIV pts with NADC<br />

were identified from the CHAMP cohort and retrospectively reviewed for<br />

HIV and cancer characteristics, overall survival (OS), and pt demographics.<br />

Statistics: Survival data was analyzed using Kaplan-Meier analysis and Cox<br />

Proportional Hazards model. Results: Of 438 pts identified, 157 were<br />

NADC representing 21 ca. The average (ave) age was 48 yrs (range 44-57),<br />

with prostate ca having highest age presentation. Over the past 10 yrs, the<br />

number <strong>of</strong> NADC has risen from 10 to over 20 each yr. Unlike historical<br />

controls (HC) where lung ca is most common, anal ca (21%) was most<br />

frequent followed by lung ca (17%). Prostate, head and neck (HNSCC),<br />

liver, and colorectal ca were seen in 9, 9, 8, and 7% respectively. 65% <strong>of</strong><br />

pts were African <strong>American</strong>s (AA) and 18% Caucasians. 78% <strong>of</strong> all NADC<br />

were men. 45% <strong>of</strong> anal ca present with stage IIIa/b disease, moderately to<br />

poorly differentiated ca in 48%, with a median OS <strong>of</strong> 34 mo. CD4 count did<br />

not alter OS but stage predicted better outcomes. 86% lung ca presented<br />

as stage III/IV disease with ave CD4 count 204. Histologically, 36% were<br />

SCC, 28% adenosquamous and 20% adenocarcinoma. OS was 5.5 mo and<br />

did not change by histology, CD4, or age. 68% HNSCC present with stage<br />

IVa/b but no IVc. Ave age was 48 yrs with an OS <strong>of</strong> 18mo. 50% were<br />

oropharyngeal compared to 22% in HC. Conclusions: Based on data by<br />

Sheilds et. al, CCH treats just over 1% <strong>of</strong> the country’s NADC population.<br />

We demonstrate a higher incidence <strong>of</strong> NADC over time, dominated by a<br />

younger, AA and male population. Each ca presents with advanced stage<br />

45-86% and poorly differentiated tumors ranging from 15-30%. The OS <strong>of</strong><br />

each cancer is consistent with HC with exception <strong>of</strong> HNSCC. As HIV pts age<br />

becoming prone to cancers <strong>of</strong> elderly, education and screening <strong>of</strong> inner city<br />

HIV pts will help improve cancer rates.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


TPS1612 General Poster Session (Board #10F), Sat, 1:15 PM-5:15 PM<br />

The MIRACLE trial: A randomized, controlled trial comparing green tea<br />

extract versus placebo for the prevention <strong>of</strong> metachronous colon adenomas<br />

in a screening population. Presenting Author: Thomas Ettrich, Department<br />

<strong>of</strong> Internal Medicine I, Martin-Luther-University Halle-Wittenberg, Halle,<br />

Germany<br />

Background: Prevention <strong>of</strong> colorectal cancer is a major health care issue.<br />

After polypectomy there is an increased risk <strong>of</strong> polyp recurrence and various<br />

means <strong>of</strong> chemoprevention have been tried to prevent this. NSAIDs have<br />

been shown to be effective but confer side effects such as gastrointestinal<br />

bleeding. Nutraceuticals such as polyphenols from tea plants have demonstrated<br />

remarkable therapeutic and preventive effects in molecular, epidemiological<br />

and some clinical trials. However, their value in preventing<br />

colorectal polyps has not been demonstrated in a large, randomized trial.<br />

The beneficial safety pr<strong>of</strong>ile <strong>of</strong> decaffeinated green tea extract and<br />

accumulating evidence <strong>of</strong> its cancer preventive potential justify and<br />

require, in our view, a validation <strong>of</strong> this compound for the nutriprevention <strong>of</strong><br />

colorectal adenoma. Good accessibility and low costs might render this<br />

neutraceutical a top candidate for wider use as nutritional supplement in<br />

colon cancer prevention. Methods: Randomized, double blinded, placebocontrolled,<br />

multicenter trial. After a one month run-in period with verum,<br />

2534 patients (age: 50-80 years) who have undergone polypectomy within<br />

the last 6 months will be randomized to receive either decaffeinated green<br />

tea extract (containing 150 mg epigallocatechin gallate (EGCG) two times<br />

daily) or placebo over a period <strong>of</strong> three years. 2028 patients are expected to<br />

complete the whole study course. Primary outcome: Incidence <strong>of</strong> metachronous<br />

colorectal adenomas (tubulovillous, tubular, villous, serrated lesions)<br />

at the 3 year follow-up colonoscopy Secondary outcomes: Occurrences,<br />

number, localization, size and histological subtypes <strong>of</strong> adenomas, frequency<br />

<strong>of</strong> colorectal carcinoma. In addition, genetic and biochemical<br />

biomarkers in blood samples and genetic alterations (e.g. K-ras, B-raf,<br />

specific microRNAs) in tissue samples <strong>of</strong> adenomas will be analyzed<br />

(biobanking subprojects). Additionally, nutrikinetics and nutrigenetics <strong>of</strong><br />

EGCG and other catechins will be assessed in healthy volunteers. Patient<br />

recruitment has started in November 2011. We expect the last patient out<br />

in fall 2017. (Trial identifier: NCT01360320)<br />

TPS1614 General Poster Session (Board #11B), Sat, 1:15 PM-5:15 PM<br />

Trial <strong>of</strong> exercise in ovarian cancer survivors. Presenting Author: Melinda<br />

Irwin, Yale University, New Haven, CT<br />

Background: Physical activity (PA) has been associated with improved QOL<br />

and survival in breast and colorectal cancer survivors, yet no study has<br />

examined the effect <strong>of</strong> exercise on ovarian cancer outcomes. The purpose <strong>of</strong><br />

our NCI-funded trial is to examine, in 230 women diagnosed with Stage I-IV<br />

ovarian cancer, the impact <strong>of</strong> exercise vs. attention control on: QOL, body<br />

composition, and serum markers associated with ovarian cancer. This<br />

abstract presents preliminary recruitment and adherence results. Methods:<br />

Women are being recruited via Connecticut Tumor Registry and Yale Cancer<br />

Center into the Women’s Activity and Lifestyle Study in Connecticut<br />

(WALC). We are also recruiting patients nationally via self-referral and<br />

collaboration with medical oncologists at DFCI and Summa Akron City<br />

Hospital. Women are randomly assigned to exercise (n � 115) or attention<br />

control (n � 115). Both groups receive weekly telephone calls for six<br />

months from a certified health educator to discuss ovarian cancer health<br />

topics. In addition, the exercise group receives PA counseling to participate<br />

in 150 min/wk <strong>of</strong> aerobic exercise (primarily brisk walking) for six months.<br />

Results: Study recruitment began in March, 2010 and will be ongoing<br />

through December, 2013. As <strong>of</strong> December 31, 2011, we have identified<br />

556 potentially eligible women. Of these, 145 are recently diagnosed and<br />

are awaiting screening, 115 were unable to be contacted, and 296 have<br />

completed the telephone screening. Of the 270, 46% were ineligible (e.g.,<br />

already exercising, physical limitations); 30% were not interested, 24% (n<br />

� 66) have been randomized, and 9% (n � 24) are undergoing baseline<br />

visits. On average, women are 62 years old, diagnosed 1.8 years in the past,<br />

with primarily stage III disease. Thirty-three patients have completed the<br />

trial, with high adherence among women randomized to exercise (average<br />

141 min/wk <strong>of</strong> exercise reported and 75% <strong>of</strong> exercisers participating in at<br />

least 150 min/wk <strong>of</strong> exercise). Conclusions: Women are interested in and<br />

able to participate in exercise after an ovarian cancer diagnosis. Our trial<br />

could suggest a unique and important role for exercise in ovarian cancer<br />

care given that physical and functional aspects <strong>of</strong> QOL are <strong>of</strong>ten the most<br />

compromised in ovarian cancer patients.<br />

Cancer Prevention/Epidemiology<br />

TPS1613 General Poster Session (Board #11A), Sat, 1:15 PM-5:15 PM<br />

Mitigation <strong>of</strong> iatrogenic risk in young women Hodgkin’s survivors, acceptability<br />

<strong>of</strong> a structured information process. Presenting Author: François<br />

Eisinger, Institut Paoli Calmettes, Marseille, France<br />

Background: Long term follow up <strong>of</strong> cancer survivors uncovers iatrogenic<br />

risk. Higher risk for second cancers should be mainly observed for cancer<br />

with a high rate <strong>of</strong> therapeutic efficacy, using aggressive therapeutic and for<br />

cancers occurring in young people. Hodgkin lymphoma fits all these<br />

criteria. Consistent data about higher risk for breast cancer in these women<br />

had been published. However, risk management is not yet <strong>of</strong>ten carried out.<br />

Methods: We plan to carry out a national program aiming both at <strong>of</strong>fering<br />

counselling and screening protocol and to assess key parameters allowing<br />

to better estimate risk, and <strong>of</strong>fering acceptable and effective risk management<br />

option. Our National program will start in January 2013. We are now<br />

in a pilot phase in a single medical institution in which we <strong>of</strong>fered women<br />

previously affected with an Hodgkin lymphoma to have access to risk<br />

analysis and information and if they agree to a specific breast cancer<br />

screening program. We present some <strong>of</strong> our psychosocial questionnaire<br />

analysis based on 27 women (aged 19-50 years old) fulfilled at the end <strong>of</strong><br />

the information consultation and one month latter. Results: There was few<br />

or no negative assessment: no one stated that they express regrets to have<br />

been informed, nor they better ignore some delivered information, no one<br />

feel that risk management description is unsatisfying and no one will advice<br />

an other women against that kind <strong>of</strong> consultation. Only 4 out 27 i.e. 15%<br />

stated that there was in delivered information a source for anxiety. However<br />

positive assessment was only fairly high and not stable after one month.<br />

Information process to inform women survivors from Hodgkin lymphoma <strong>of</strong><br />

their higher risk <strong>of</strong> being affected with a breast cancer and the current<br />

recommendation for earlier breast cancer screening reach a high level <strong>of</strong><br />

satisfaction at the initial stage <strong>of</strong> the process. However it could decrease<br />

with time and thus, need to be monitored.<br />

At the end <strong>of</strong><br />

the consult<br />

113s<br />

One month<br />

later<br />

No induced anxiety 11/27 (41%) 8/27 (30%)<br />

No regrets 17/27 (63%) 13/27 (48%)<br />

Nothing better to ignore 18/27 (67%) 13/27 (48%)<br />

Risk management clear and fine 17/27 (63%) 11/27 (41%)<br />

Advise other women to undergone such consult 16/27 (59%) 12/27 (44%)<br />

TPS1615 General Poster Session (Board #11C), Sat, 1:15 PM-5:15 PM<br />

Statin polyp prevention trial in patients with resected colon cancer: NSABP<br />

protocol P-5. Presenting Author: Bruce M. Boman, National Surgical<br />

Adjuvant Breast and Bowel Project and Helen F. Graham Cancer Center,<br />

Newark, DE<br />

Background: HMG-CoA reductase inhibitors (statins) are effective lipidlowering<br />

agents that are used for treating hyperlipidemia. Results from cell<br />

culture, animal experiments, and epidemiologic studies indicate that<br />

statins may be active chemopreventive agents against colorectal cancer<br />

(CRC). However, the clinical studies come largely from retrospective,<br />

observational studies originally designed to investigate lipid-lowering or<br />

cardiovascular endpoints rather than tumor endpoints. Methods: NSABP<br />

P-5 is a randomized, prospective placebo-controlled, double-blind study <strong>of</strong><br />

rosuvastatin (10 mg per day x 5 years) or placebo for the prevention <strong>of</strong><br />

adenomatous polyps <strong>of</strong> the colon or rectum, metachronous colorectal<br />

carcinoma, and colon cancer recurrence. Additional secondary endpoints<br />

include the size, number and features <strong>of</strong> the colorectal adenomas as well as<br />

health-related quality <strong>of</strong> life and self-reported symptoms. Selected Eligibility:<br />

(1) Patients who have undergone complete resection <strong>of</strong> AJCC Stage I or<br />

II colon cancer within 1 year before randomization. (2) Colonoscopy within<br />

180 days before randomization. Selected Ineligibility: (1) Hyperlipidemia,<br />

(2) Familial Adenomatous Polyposis, (3) Lynch Syndrome. Sample Size:<br />

1,740 patients will be entered, and as <strong>of</strong> January 23, 2012, 163 (9.37%)<br />

have been randomized. The study is open at NSABP and CTSU sites in<br />

North America. Funded by NCI PHS grants U10-CA-37377 and U10-CA-<br />

6974, with additional funding from AstraZeneca Pharmaceuticals, LP.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


114s Cancer Prevention/Epidemiology<br />

TPS1616 General Poster Session (Board #11D), Sat, 1:15 PM-5:15 PM<br />

O6-benzylguanine (BG) and temozolomide (TMZ) therapy <strong>of</strong> glioblastoma<br />

multiforme (GBM) with infusion <strong>of</strong> autologous lentiviral transduced<br />

P140KMGMT� hematopoietic progenitors to protect hematopoiesis: A<br />

phase I study. Presenting Author: Andrew E. Sloan, University Hospital<br />

Case Medical Center, Cleveland, OH<br />

Background: The administration <strong>of</strong> radiation therapy (RT) and concomitant<br />

and adjuvant temozolomide (TMZ) for the treatment <strong>of</strong> newly-diagnosed<br />

glioblastoma (GBM) is associated with modestly prolonged survival. However,<br />

the benefit <strong>of</strong> concurrent therapy is restricted to a subgroup <strong>of</strong><br />

patients whose tumor exhibits methylation <strong>of</strong> the promoter for methylguanine-DNA-methyltransferase<br />

(MGMT). Promoter methylation silences the<br />

expression <strong>of</strong> DNA alkyltransferase (AGT), the product <strong>of</strong> the MGMT gene<br />

which functions to repair alkylated DNA. Unfortunately, the majority <strong>of</strong><br />

GBMs have non-methylated MGMT promoters and do not appear to benefit<br />

from adding TMZ to radiotherapy. Two potential strategies for targeting<br />

MGMT-induced chemoprotection in gliomas include depletion <strong>of</strong> tumor<br />

MGMT, and dose escalation <strong>of</strong> TMZ accomplished by minimizing TMZinduced<br />

bone marrow toxicity. In this study, we propose to combine the<br />

irreversible MGMT inhibitor O6-benzylguanine (BG) with infusion <strong>of</strong> autologous<br />

hematopoetic stem cells (HSCs) transduced with lentiviral P140K-<br />

MGMT, an MGMT mutant which is highly resistant to inactivation by BG,<br />

with the intent to reduce or prevent the myelosuppressive effects <strong>of</strong> TMZ.<br />

The primary objective is to assess the safety and feasibility <strong>of</strong> combining<br />

these two strategies in patients with newly-diagnosed GBM. Methods: This<br />

is a three cohort, Phase I study for patients with newly resected supratentorial<br />

GBM. There will be at least four evaluable patients per cohort. Cohort 1<br />

will receive standard RT and concomitant TMZ prior to infusion <strong>of</strong><br />

autologous lentiviral P140K-MGMT transduced HSCs, followed by adjuvant<br />

TMZ. Cohort 2 will receive an induction dose <strong>of</strong> TMZ�BG followed by<br />

infusion <strong>of</strong> autologous LV-P140K-MGMT HSCs prior to concomitant RT<br />

�TMZ and adjuvant TMZ. Cohort 3 will include intra-patient dose escalation<br />

<strong>of</strong> TMZ in patients who exhibit detectable levels <strong>of</strong> P140K-MGMT<br />

marked cells in vivo, using the design (i.e. cohort 1 or 2) proven to be safest<br />

and most effective. The study is open and has accrued the first patient.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


2000 Oral Abstract Session, Sun, 8:00 AM-11:00 AM<br />

MGMT promoter methylation as a predictive biomarker for response to<br />

radiotherapy versus chemotherapy in malignant astrocytomas in the elderly:<br />

The NOA-08 trial. Presenting Author: Wolfgang Wick, University<br />

Hospital Heidelberg, Heidelberg, Germany<br />

Background: In a few years more than half <strong>of</strong> the patients with glioblastoma<br />

will be older than 65 years <strong>of</strong> age and thus be classified as elderly. The<br />

current standard <strong>of</strong> care in elderly patients with glioblastoma (GB) or<br />

anapestic astrocytoma (AA) is resection or biopsy followed by involved-field<br />

radiotherapy (RT). The role <strong>of</strong> primary chemotherapy is poorly defined. The<br />

NOA-08 trial compared efficacy and safety <strong>of</strong> RT to temozolomide (TMZ) in<br />

patients with newly diagnosed AA or GB. Methods: Patients (N�412; 39<br />

AA, 373 GB) � 65 years with a Karn<strong>of</strong>sky performance score � 60 were<br />

randomized to receive RT or TMZ. The primary endpoint was overall survival<br />

(OS). The trial sought to demonstrate the non-inferiority <strong>of</strong> TMZ compared<br />

with RT. Results: Patient characteristics in the intention-to-treat population<br />

[N�373 (178 patients RT, 195 patients TMZ)] were balanced. All<br />

histologic diagnoses (11% AA and 89% GB) were centrally confirmed.<br />

Median OS [HR�1.09 (95% CI: 0.84-1.42)] and event-free survival (EFS)<br />

[hazard ratio (HR)�1.15 (0.92-1.43)] <strong>of</strong> TMZ versus RT did not differ<br />

between both arms. Non-inferiority <strong>of</strong> TMZ compared with RT was significant<br />

(p�0.05). Extent <strong>of</strong> resection, but not age or diagnosis <strong>of</strong> AA were<br />

associated with prolonged EFS and OS. DNA repair protein O6-methylgua nine DNA-methyltransferase (MGMT) promoter methylation in tumor tissue<br />

was associated with prolonged OS [HR�0.67 (0.38-1.29)]. Patients with<br />

MGMT promoter methylation had longer EFS when treated with TMZ (8.4<br />

months [5.5-11.7] versus RT (4.6 [4.2-5] months) whereas patients<br />

without MGMT promoter methylation had longer EFS when treated with RT<br />

(4.6 [3.7-6.3] versus 3.3 [3-3.5] months). This effect persisted for OS.<br />

Conclusions: NOA-08 demonstrates the non-inferiority <strong>of</strong> TMZ compared<br />

with RT in the treatment <strong>of</strong> elderly patients with malignant astrocytoma.<br />

MGMT promoter methylation is a strong predictive biomarker for the choice<br />

between RT and TMZ.<br />

2002 Oral Abstract Session, Sun, 8:00 AM-11:00 AM<br />

The NF�B pathway as a key mediator <strong>of</strong> the glioblastoma mesenchymal<br />

subtype in glioblastoma stem cells and human tumors. Presenting Author:<br />

Brian D. Vaillant, The Methodist Hospital, Houston, TX<br />

Background: The mesenchymal (MES) subtype <strong>of</strong> glioblastoma (GBM) is<br />

associated with worse prognosis and increased treatment resistance.<br />

Several transcription factors driving the MES phenotype have been identified,<br />

including STAT3, CEBP-�, and WWTR1/TAZ. However, the key<br />

signaling pathways driving this transcriptional program remain unknown.<br />

Methods: Expression pr<strong>of</strong>iling analyses was performed on multiple GBM<br />

stem cell (GSC) lines derived from individual human tumors. Three large<br />

GBM tumor gene expression datasets (TCGA, Rembrandt, Erasmus) were<br />

also used in the analyses to identify pathways activated in MES tumors and<br />

GSCs. Intracranial GSC xenograft models were used for in vivo validation.<br />

Results: We found that GSCs with a MES phenotype also demonstrated<br />

upregulation <strong>of</strong> a gene cassette associated with NF�B activation. Analysis<br />

<strong>of</strong> multiple large expression data sets also demonstrated a tight correlation<br />

between the MES phenotype and NF�B activation in human tumors. To<br />

determine the effect <strong>of</strong> NF�B activation in GSCs, we stimulated NF�B by<br />

addition <strong>of</strong> the inflammatory cytokine TNF�. This was accompanied by<br />

increased expression <strong>of</strong> the MES transcription factors (STAT3, CEBP-�,<br />

TAZ), and MES markers including CD44. Conversely, blockade <strong>of</strong> NF�B<br />

signaling in GSCs by I�B-� was sufficient to prevent TNF-induced MES<br />

transition. Investigation <strong>of</strong> potential source <strong>of</strong> NF�B activation suggested a<br />

role <strong>of</strong> microglia. Indeed, addition <strong>of</strong> supernatant from activated microglia<br />

was sufficient to activate NF�B and MES transition in GSCs that could be<br />

blocked by I�B-�. To test the role <strong>of</strong> microglia and NF�B activation on<br />

treatment resistance in vivo, treatment with minocycline, an inhibitor <strong>of</strong><br />

microglia activation, led to a reduction <strong>of</strong> tumor grade and down-regulation<br />

<strong>of</strong> MES markers in intracranial GSC xenografts. Conclusions: NF�B appears<br />

to play an important role in induction <strong>of</strong> the MES phenotype in GBM and<br />

GSCs. Furthermore, activation <strong>of</strong> microglia is a potential source <strong>of</strong> NF�B<br />

activation in these tumors. These data suggest that blockade <strong>of</strong> NF�B<br />

and/or inhibition <strong>of</strong> microglia activation may be attractive therapeutic<br />

approaches for downregulating MES transition and overcoming treatment<br />

resistance in GBM.<br />

Central Nervous System Tumors<br />

2001 Oral Abstract Session, Sun, 8:00 AM-11:00 AM<br />

A revised RTOG recursive partitioning analysis (RPA) model for glioblastoma<br />

based upon multiplatform biomarker pr<strong>of</strong>iles. Presenting Author:<br />

Arnab Chakravarti, Arthur G. James Cancer Center, The Ohio State<br />

University, Columbus, OH<br />

Background: The Radiation Therapy Oncology Group (RTOG) recursive<br />

partitioning analysis (RPA) model, which relies on clinical variables, has<br />

been used worldwide to establish distinct prognostic classes <strong>of</strong> patients<br />

(pts) with malignant glioma as well as eligibility criteria for clinical trials. In<br />

the present study, we have updated the RPA to include additional<br />

molecular variables, specifically for glioblastoma (GBM) patients treated in<br />

the temozolomide (TMZ)-era, to make the model more relevant, contemporary,<br />

and discriminatory. Methods: The dataset utilized was from RTOG<br />

0525, a phase III study examining radiation (RT) with concurrent TMZ,<br />

followed by adjuvant standard dose vs. dose-dense TMZ in pts with<br />

newly-diagnosed GBM. 162 pts from RTOG 0525 had available tissues for<br />

pr<strong>of</strong>iling <strong>of</strong> key signaling molecules using the AQUA platform. Results:<br />

pAKT, c-met, and MGMT protein were each found to be significantly<br />

associated with adverse outcome on multivariate analysis. These variables<br />

were combined with clinical and genetic biomarkers (e.g., MGMT promoter<br />

methylation, IDH1 mutation, mRNA pr<strong>of</strong>iling) previously found to be <strong>of</strong><br />

significance (MCP model, ASCO, 2011) to generate an even more robust,<br />

discriminatory RTOG RPA model. The explained variation for these three<br />

classification models was found to be 41.7 (Current RPA), 19 (MCP), and<br />

14.9% (<strong>Clinical</strong> RPA), respectively, with higher values indicating better<br />

separation <strong>of</strong> prognostic groups (see table). Conclusions: The current RTOG<br />

RPA classification model, based upon incorporation <strong>of</strong> multi-platform<br />

biomarker analysis, holds promise for RT�TMZ-treated GBM patients;<br />

further validation <strong>of</strong> this model is planned. Financial Support: NCI grants<br />

U10 CA21661, U10 CA37422, U24 CA114734, 1RC2CA148190,<br />

1RC2CA148190, RO1CA108633, and BTFC grant.<br />

<strong>Clinical</strong> RPA Current RPA<br />

Model class<br />

Median<br />

survival<br />

(months)<br />

Lower 95%><br />

confidence<br />

limit (CL)<br />

Upper<br />

95% CL<br />

No.<br />

cases<br />

(%)<br />

Model<br />

class<br />

Median<br />

survival Lower<br />

(months) 95% CL Upper<br />

95% CL<br />

115s<br />

No.<br />

cases<br />

(%)<br />

III 33.3 16.8 -na- 32 (19.6) III -na- 23.5 -na- 17 (10.5)<br />

IV 16.3 13.4 19.9 89 (55.9) IV 22.4 15.5 33.3 33 (20.4)<br />

V 13.1 7.9 16.6 41 (25.3) V 15.7 12.9 17.0 101 (62.4)<br />

VI 5.2 2.2 13.6 11 (6.8)<br />

Abbreviation: na, not reached.<br />

2003 Oral Abstract Session, Sun, 8:00 AM-11:00 AM<br />

Randomized phase II 8-arm factorial study <strong>of</strong> adjuvant dose-dense (dd)<br />

temozolomide (TMZ) with permutations <strong>of</strong> thalidomide (Thal), isotretinoin<br />

(CRA), and/or celecoxib (Cel) for newly diagnosed glioblastoma (GBM).<br />

Presenting Author: Mark R. Gilbert, University <strong>of</strong> Texas M. D. Anderson<br />

Cancer Center, Houston, TX<br />

Background: Concurrent radiation and TMZ followed by 6-12 months <strong>of</strong><br />

adjuvant TMZ (d 1-5 <strong>of</strong> a 28d cycle) is the current standard <strong>of</strong> care for<br />

patients with newly diagnosed GBM. The addition <strong>of</strong> cytostatic or signal<br />

transduction agents may enhance efficacy without a significant increase in<br />

toxicity. A phase I trial (Neuro-oncology 2009) established the safety <strong>of</strong><br />

ddTMZ with 2 or 3 <strong>of</strong> the cytostatic agents. Methods: This randomized<br />

phase II study was conducted by the Brain Tumor Trials Collaborative<br />

(BTTC) and the MDACC CCOP. The primary objectives: determine if<br />

specific cytostatic agents added to ddTMZ alters outcomes (PFS, OS) and<br />

compare triplet with doublet therapy. Eligibility criteria: centrally confirmed<br />

newly diagnosed GBM, age �18, KPS�60, stable or improved after<br />

chemoradiation (pseudoprogression allowed), adequate hematologic, renal<br />

and hepatic function. Pts were randomly assigned to 12 treatment cycles<br />

(28 d/cycle) in 8 arms: ddTMZ alone (150 mg/m2/day, 7-d on, 7-d <strong>of</strong>f) or<br />

TMZ-containing doublet, triplet and quadruplet combinations with Thal,<br />

CRA, or Cel. Results: The study enrolled 155 eligible patients from<br />

11/2005 to 9/2010 to the 8 arms <strong>of</strong> the factorial design. Median age was<br />

53 (18-84) and median KPS, 90 (60-100). Compared with TMZ alone, the<br />

TMZ�CRA doublet had worse PFS (10.5, 6.5 mo; p�0.043) and OS<br />

(21.2, 11.7 mo; p�0.037). Trends were also seen for worse outcome (PFS,<br />

OS) for CRA-containing regimens, improved OS for Cel containing arms and<br />

no impact <strong>of</strong> Thal. A strong trend for OS improvement was seen for triplet<br />

compared with doublet regimens (20.1, 17.0 mo; p�0.15), but no<br />

difference for PFS. Treatment was well tolerated with expected high rates <strong>of</strong><br />

grade 3/4 lymphopenia, and overall a modest toxicity rate. Conclusions: The<br />

results indicate that the addition <strong>of</strong> CRA to ddTMZ may be detrimental in<br />

patients with newly diagnosed GBM. This study demonstrated the utility <strong>of</strong><br />

the factorial design in efficiently testing drug combinations, the impact <strong>of</strong><br />

individual agents in these combinations as well as doublet vs. triplet<br />

regimens and supports its utility in testing combinations <strong>of</strong> targeted agents.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


116s Central Nervous System Tumors<br />

2004 Oral Abstract Session, Sun, 8:00 AM-11:00 AM<br />

Relationship between overall survival and progression-free survival for<br />

recent NCCTG glioblastoma multiforme trials. Presenting Author: Wenting<br />

Wu, Mayo Clinic, Rochester, MN<br />

Background: With the approval <strong>of</strong> novel agents for glioblastoma (GBM)<br />

treatment, reliable historical outcome data for the common phase II<br />

endpoint <strong>of</strong> progression free survival (PFS) is not available. In particular,<br />

the association between progression and overall survival (OS) has not been<br />

evaluated after temozolomide (TMZ) became the standard therapy for<br />

newly diagnosed GBM. Methods: Datasets from 5 TMZ-including NCCTG<br />

clinical trials in newly diagnosed GBM (n�325, 2005-2011, 1 phase I<br />

only and 4 phase I and single arm phase II trials) were pooled and analyzed<br />

to evaluate the individual level correlation between PFS and OS using<br />

correlation coefficient, landmark analyses, and time-dependent variable<br />

analyses. A historical control group was constructed by pooling datasets<br />

from 12 pre-TMZ era NCCTG clinical trials in the same patient population<br />

(n�1359, 1980-2004). Each <strong>of</strong> the 5 newer trials was compared to the<br />

control group to estimate the treatment effect (hazard ratio) on PFS and<br />

OS, which were analyzed to evaluate the trial level correlation between PFS<br />

and OS using correlation coefficient and weighted linear regression.<br />

Results: The estimated correlation coefficient between PFS and OS at<br />

individual patient level is 0.75 (95%CI, 0.7 to 0.8). The estimated hazard<br />

ratios <strong>of</strong> death in the landmark analysis at 4 and 6 months are 2.15<br />

(95%CI, 1.47 to 3.15) and 2.17 (95%CI, 1.57 to 3.12), respectively, and<br />

the estimated hazard ratio <strong>of</strong> death is 10.1 (95%CI, 6.6 to 15.4) when<br />

progression is treated as a time-dependent variable. For trial level correlation<br />

(against the historical control group), the estimated correlation<br />

coefficient between HRos and HRpfs is 0.51 (95%CI, -0.67 to 0.96), with<br />

moderate prediction: the predicted HRos is very close to the observed HRos in 4 <strong>of</strong> 5 cases, however the predicted HRos is significantly different from 1<br />

in only 2 out <strong>of</strong> 5 cases due to small sample size in other 3 cases.<br />

Conclusions: PFS and OS are highly associated at the individual patient<br />

level but only moderately at the trial level, the latter possibly due to small<br />

sample size in some <strong>of</strong> the trials. Additional validation in other trials could<br />

support use <strong>of</strong> PFS as a primary endpoint for randomized phase II trials in<br />

GBM.<br />

2006 Oral Abstract Session, Sun, 8:00 AM-11:00 AM<br />

Consolidation reduced dose whole brain radiotherapy (rdWBRT) following<br />

methotrexate, rituximab, procarbazine, vincristine, cytarabine (R-MPV-A)<br />

for newly diagnosed primary CNS lymphoma (PCNSL): Final results and<br />

long-term outcome. Presenting Author: Richard Charles Curry, Memorial<br />

Sloan-Kettering Cancer Center, New York, NY<br />

Background: After promising early results in the pilot phase (N� 30)<br />

reported by Shah 2007, we report on final results <strong>of</strong> a multicenter phase II<br />

study (N�52) in newly diagnosed PCNSL combining R-MPV-A and rdW-<br />

BRT, expanded to address long-term disease control and cognitive outcomes<br />

in pts who actually received rdWBRT. Methods: Pts received 5 cycles<br />

<strong>of</strong> R-MPV (MTX dose: 3.5g/m2). Those with partial response (PR) received<br />

additional 2 cycles. Pts with a complete response (CR) after 5-7 cycles<br />

received rdWBRT (23.4 Gy), otherwise standard WBRT was <strong>of</strong>fered (45<br />

Gy). Consolidation cytarabine was given to all pts. Primary end-point was<br />

2-y progression-free survival (PFS) in pts receiving rdWBRT (n�30 pts in<br />

CR required). Exploratory end-points included comprehensive neuropsychological<br />

testing, white matter changes (WMC) analysis (Fazekas scale) and<br />

apparent diffusion coefficient (ADC) on MRI. Results: Accrual was completed<br />

(N�52; 22 women); median (med) age� 60 (30-79); med KPS�<br />

70 (50-100). In total, 31 (59%) pts were assessable for the primary<br />

endpoint (achieved a CR after induction and received rdWBRT). The 2-y<br />

PFS for this group was 78% (95% ci: 64%- 92%); med PFS� 7.7 y; the<br />

med OS was not reached (med follow-up� 6y); 3y-OS� 88% (ci 70-95);<br />

5y-OS� 81% (ci 62-91). Cognitive testing showed improvement in<br />

executive function (P � 0.01) and verbal memory (P � 0.05) following<br />

induction chemotherapy; follow-up scores remained stable across the<br />

various domains. Minimal WMC developed in long term survivors: 36% <strong>of</strong><br />

pts showed no change in Fazekas’ scores, 64% <strong>of</strong> pts developed scores 1 or<br />

2 and no pt showed scores 3-6. The intent-to-treat (n�52) med PFS was<br />

3.3y; med OS� 6.6 y. Differences in ADC values did not predict response<br />

(p�0.15), PFS (p�0.41) or OS (p�0.48). Conclusions: Consolidation<br />

rdWBRT is a highly effective and safe treatment for newly diagnosed<br />

PCNSL. Long term follow-up showed robust PFS and OS, comparable or<br />

superior to full dose WBRT, with excellent cognitive outcomes and no<br />

significant WMC over time. An RTOG randomized trial has been initiated<br />

comparing R-MPV-A with vs without rdWBRT.<br />

2005 Oral Abstract Session, Sun, 8:00 AM-11:00 AM<br />

Bayesian adaptive randomized trial design for patients with recurrent<br />

glioblastoma. Presenting Author: Brian Michael Alexander, Dana-Farber<br />

Cancer Institute/Brigham and Women’s Cancer Center, Boston, MA<br />

Background: Bayesian-based trial design has the ability to utilize accumulating<br />

data in real time to alter the course <strong>of</strong> the trial, thereby enabling<br />

dynamic allocation to experimental arms and earlier dropping <strong>of</strong> ineffective<br />

arms. This flexibility results in a potentially more efficient trial framework<br />

by increasing the probability <strong>of</strong> enrollment to arms that show evidence <strong>of</strong><br />

efficacy. In this study we considered a hypothetical scenario in which<br />

patients who have been previously treated on several separate experimental<br />

protocols at the Dana-Farber/Harvard Cancer Center and the University <strong>of</strong><br />

California, Los Angeles were instead enrolled in a single multi-arm protocol<br />

utilizing a Bayesian adaptively randomized trial design. The purpose was to<br />

determine whether similar scientific results could have been accomplished<br />

more efficiently. Methods: We generated an adaptive randomization procedure<br />

that was retrospectively applied to primary patient data from four<br />

separate phase II clinical trials in patients with recurrent glioblastoma. We<br />

then compared AR to more conventional trial designs using realistic<br />

hypothetical scenarios consistent with survival data reported in the literature.<br />

Our primary endpoint is the number <strong>of</strong> patients needed to achieve a<br />

desired statistical power. Results: If our phase II trials had been a single<br />

multi-arm AR trial, bevacizumab would have been identified as an<br />

efficacious therapy, and 30 fewer patients would have been needed<br />

compared to a multi-arm balanced randomized (BR) design. More generally,<br />

Bayesian AR trial design for patients with glioblastoma would result in<br />

trials with fewer overall patients with no loss in statistical power, and in<br />

more patients randomized to effective treatment arms. For a trial with a<br />

control arm, two ineffective arms and one effective arm with hazard ratio<br />

0.6, a median <strong>of</strong> 47 patients would be randomized to the effective arm<br />

compared with 35 in a BR design. Conclusions: Given the desire for control<br />

arms in phase II trials, an increasing number <strong>of</strong> experimental therapeutics<br />

for patients with glioblastoma, and a relatively short time for events,<br />

Bayesian adaptive designs are attractive for clinical trials in glioblastoma.<br />

2007 Oral Abstract Session, Sun, 8:00 AM-11:00 AM<br />

Elderly patients with primary CNS lymphoma: Results from the G-PCNSL-<br />

SG-1 trial. Presenting Author: Patrick Roth, University Hospital Zurich,<br />

Zurich, Switzerland<br />

Background: Age is the most important therapy-independent prognostic<br />

factor in patients with primary central nervous system lymphoma (PCNSL).<br />

Here we aimed at providing an analysis <strong>of</strong> the impact <strong>of</strong> higher age on<br />

response to therapy, toxicity, and survival in the largest PCNSL trial ever<br />

performed to date. Methods: Response to therapy, toxicity and survival <strong>of</strong><br />

PCNSL patients enrolled in the G-PCNSL-SG-1 trial evaluating the role <strong>of</strong><br />

radiotherapy after high-dose methotrexate (HD-MTX)-based chemotherapy<br />

were monitored. Subjects aged 70 or more were compared to younger<br />

patients. Results: Of all eligible patients (n�526), 126 (24%) were aged<br />

70 or more. In the per protocol population, 66 <strong>of</strong> 318 patients (21%) were<br />

at least 70 years old. Among the eligible patients, the rate <strong>of</strong> complete and<br />

partial responses (CR�PR) to HD-MTX-based chemotherapy was 44% in<br />

the elderly compared to 57% in the younger patients (p�0.016). A higher<br />

rate <strong>of</strong> grade III/IV leukopenia was observed in the elderly (34% versus<br />

21%, p�0.007). Also, death on therapy was more frequent (18% versus<br />

11%; p�0.027) in these patients. In contrast, there was no other major<br />

age-dependent toxicity. Survival analyses revealed shorter progression-free<br />

survival (PFS) (4.0 versus 7.7 months, p�0.014) and overall survival (OS)<br />

(12.5 versus 26.2 months, p�0.001) in the elderly population. The PFS <strong>of</strong><br />

CR patients was 35.0 months in younger patients compared to 16.1 in the<br />

elderly (p�0.024). Salvage therapy was used less commonly in elderly<br />

patients. When salvage WBRT was applied in patients who had failed on<br />

HD-MTX-based chemotherapy, there was no association between age and<br />

survival (p�0.633). Conclusions: Elderly PCNSL patients have a lower<br />

response rate and higher mortality on HD-MTX-based chemotherapy. Their<br />

PFS is shorter and they receive less salvage therapy which may contribute<br />

to the poor prognosis.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


2008 Oral Abstract Session, Sun, 8:00 AM-11:00 AM<br />

Rituximab, methotrexate (MTX), procarbazine, and vincristine (R-MPV)<br />

followed by consolidation high-dose chemotherapy (HDC) and autologous<br />

stem-cell transplant (ASCT) for newly diagnosed primary CNS lymphoma<br />

(PCNSL). Presenting Author: Antonio Marcilio Padula Omuro, Memorial<br />

Sloan-Kettering Cancer Center, New York, NY<br />

Background: In our previous study in newly diagnosed PCNSL, induction<br />

chemotherapy with MTX and cytarabine followed by consolidation HDC<br />

(carmustine, etoposide, cytarabine, melphalan [BEAM]) with ASCT without<br />

radiotherapy resulted in only 50% <strong>of</strong> pts transplanted, reflecting low<br />

efficacy <strong>of</strong> induction chemotherapy, and short intent-to-treat (ITT) median<br />

PFS (�6m). In this phase II trial, we sought to optimize this strategy by<br />

utilizing a more effective induction regimen (R-MPV) and a more aggressive<br />

HDC regimen (Soussain et al). Methods: Pts received 5-7 cycles <strong>of</strong> R-MPV<br />

(MTX: 3.5g/m2) and if a partial or complete response was achieved, HDC<br />

with thiothepa, cyclophosphamide and busulfan was given, followed by<br />

ASCT and no radiotherapy. The primary endpoint was ITT 1 year event-free<br />

survival (promising: 75%, non-promising: 50%; 90% power, significance�0.05).<br />

Follow-up included comprehensive neuropsychological evaluation.<br />

Results: Accrual has been completed (N�32 pts, median age 57<br />

[range 23-67], median KPS�80). Following R-MPV, 17 pts achieved a CR,<br />

13 pts a PR and two pts progressed. A total <strong>of</strong> 25 (78%) pts were<br />

transplanted; the reasons for not receiving transplant were progressive<br />

disease (N�2), poor performance status/ physician’s decision (N� 2),<br />

mobilization failure (N�1) and consent withdrawn (N� 2). One pt who<br />

withdrew consent relapsed and received HDCASCT for salvage. Two (8%)<br />

pts died from early complications <strong>of</strong> ASCT (Stevens-Johnson: one, sepsis:<br />

one) and one pt experienced a fatal late colitis <strong>of</strong> unknown etiology. In the<br />

ITT population, the median EFS and OS have not been reached after a<br />

median follow-up <strong>of</strong> 22 months. The 1 year EFS was 78% (95%CI 58-90)<br />

and the 2y OS was 76% (95% CI 54-89). No pt has developed delayed<br />

neurotoxicity. Conclusions: R-MPV induction regimen resulted in improved<br />

response rates, allowing 78% <strong>of</strong> pts to receive HDC-ASCT. Although more<br />

toxic, this regimen resulted in excellent disease control and survival in the<br />

ITT population, far exceeding the efficacy <strong>of</strong> our previous transplant study.<br />

The primary endpoint was met, warranting further investigation.<br />

2009 <strong>Clinical</strong> Science Symposium, Sat, 1:15 PM-2:45 PM<br />

Effects <strong>of</strong> cediranib, a VEGF signaling inhibitor, in combination with<br />

chemoradiation on tumor blood flow and survival in newly diagnosed<br />

glioblastoma. Presenting Author: Elizabeth Robins Gerstner, Massachusetts<br />

General Hospital, Boston, MA<br />

Background: Anti-angiogenic therapy is hypothesized to synergize with<br />

radiation and chemotherapy by improving tumor blood flow. We evaluated<br />

the tolerability, efficacy and potential mechanism <strong>of</strong> action <strong>of</strong> radiation,<br />

temozolomide, and cediranib in newly diagnosed glioblastoma patients.<br />

Methods: Newly diagnosed glioblastoma patients were treated with radiation,<br />

temozolomide, and cediranib followed by monthly temozolomide for 6<br />

cycles and daily cediranib until tumor progression or toxicity as part <strong>of</strong> an<br />

IRB-approved, Phase Ib/II clinical trial. MRI scans including measurement<br />

<strong>of</strong> cerebral blood flow were performed at baseline, weekly during the 6<br />

weeks <strong>of</strong> chemoradiation and then monthly. Radiographic response was<br />

determined by RANO criteria. Results: Six patients were enrolled in the<br />

phase Ib part <strong>of</strong> the study with cediranib 30 mg daily in combination with<br />

temozolomide and radiation. No dose-limiting toxicities were identified.<br />

Forty patients were enrolled in the phase II part <strong>of</strong> the study. Among the<br />

entire cohort <strong>of</strong> 46 patients, median age was 57 (range 35-74), median<br />

KPS was 90% (60-100), 36 patients underwent a subtotal resection and<br />

10 underwent biopsy. 26/30 patients taking corticosteroids were able to<br />

taper corticosteroids during chemoradiation. Off study reasons included<br />

toxicity (14), disease progression (18), and patient preference (2). Five<br />

patients remain on study without disease progression and 20 patients have<br />

died. Median duration on study was 158 days. Median progression free<br />

survival was 288 days (95%CI 240,�) and median overall survival was 786<br />

days (95%CI 411 ,�). Best radiographic response in patients who completed<br />

chemoradiation was CR in 2 patients, PR in 20 patients, and SD in<br />

15 patients. Patients with increased tumor perfusion during chemoradiation<br />

survived nearly 1 year longer (mean OS�611 days) than patients with<br />

decreased perfusion (mean OS�269 days). Conclusions: Cediranib was<br />

well tolerated and led to improved PFS and OS compared to historical<br />

controls, particularly in those with improved perfusion. This combination is<br />

being evaluated in an ongoing randomized trial (RTOG 0837).<br />

Central Nervous System Tumors<br />

117s<br />

2008b Oral Abstract Session, Sun, 8:00 AM-11:00 AM<br />

Chemotherapy plus radiotherapy (CT-RT) versus RT alone for patients with<br />

anaplastic oligodendroglioma: Long-term results <strong>of</strong> the RTOG 9402 phase<br />

III study. Presenting Author: J. Gregory Cairncross, University <strong>of</strong> Calgary,<br />

Calgary, AB, Canada<br />

Background: Anaplastic oligodendrogliomas, pure (AO) and mixed (AOA),<br />

are chemosensitive tumors, especially if co-deleted for chromosomes 1p<br />

and 19q, but whether addition <strong>of</strong> CT to RT prolongs overall survival (OS), is<br />

unknown. Methods: In the RTOG 9402 Phase III trial, patients (pts) with<br />

AO/AOA were randomly assigned to PCV [procarbazine, CCNU (lomustine)<br />

and vincristine] followed by immediate RT vs. immediate RT alone. Early<br />

analysis showed no OS benefit for the PCV�RT group but combined therapy<br />

was associated with a longer progression-free survival (PFS). It also showed<br />

that the finding <strong>of</strong> 1p/19q co-deletion was associated with a longer OS<br />

independent <strong>of</strong> treatment. The current analysis has a median follow up <strong>of</strong><br />

11.3 years (yrs). Results: Two hundred ninety-one patients were randomized,<br />

148 to PCV�RT and 143 to RT. PCV�RT was associated with longer<br />

PFS [2.5 vs. 1.7 yrs, hazard ratio (HR) 0.68, 95% confidence interval (CI)<br />

(0.53, 0.88), P � 0.003] and the 1p/19q co-deletion with a longer Median<br />

Survival Time (MST) [8.7 vs. 2.7 yrs, HR 0.41, 95% CI (0.30, 0.55), P �<br />

0.001]. For the entire cohort, there was no difference in MST by treatment<br />

[4.6 yrs for PCV�RT vs. 4.7 yrs for RT, HR 0.79, 95% CI (0.60, 1.04), P �<br />

0.1]. However, patients with 1p/19q co-deleted tumors lived much longer<br />

after PCV�RT (n � 59) than after RT (n � 67) [14.7 vs. 7.3 yrs, HR 0.59,<br />

95% CI (0.37, 0.95), P � 0.03]. There was no difference in MST by<br />

treatment in pts without the 1p/19q co-deletion [n�137; 2.6 vs. 2.7 yrs,<br />

HR 0.85, 95% CI (0.58, 1.23), P � 0.39]. Re-operation rates upon<br />

progression were similar between treatment arms in co-deleted pts (43%,<br />

PCV�RT vs. 54%, RT) but salvage CT rates were higher in the RT arm [57%<br />

vs. 81% (P � 0.04)]. Conclusions: PCV followed by immediate RT was a<br />

highly effective therapy for patients with 1p/19q co-deleted AO/AOA. In<br />

this setting, 1p/19q co-deletion was both prognostic and predictive, and<br />

the early PFS benefit in co-deleted cases was a harbinger <strong>of</strong> their longer OS.<br />

[This work was supported by RTOG grants U10 CA21661 and U10<br />

CA32115, NCCTG grant U10 CA25224, ECOG grants CA17145 and<br />

CA21115, SWOG grant CA32102, and CCOP grant U10 CA37422 from<br />

the National Cancer Institute (NCI)]<br />

2010 <strong>Clinical</strong> Science Symposium, Sat, 1:15 PM-2:45 PM<br />

Effect <strong>of</strong> antiangiogenic therapy on tumor-associated macrophages in<br />

recurrent glioblastoma. Presenting Author: Christine Lu-Emerson, Massachusetts<br />

General Hospital, Boston, MA<br />

Background: Antiangiogenic therapy is associated with increased radiographic<br />

responses in glioblastoma (GBM), but tumors invariably recur.<br />

Tumor associated macrophages (TAMs) have been proposed as a mechanism<br />

<strong>of</strong> resistance to anti-angiogenic therapy in preclinical models. To<br />

examine the role <strong>of</strong> TAMs in recurrent GBM, we analyzed autopsy<br />

specimens from patients with or without history <strong>of</strong> antiangiogenic therapy.<br />

Methods: We compared autopsy brain specimens from 17 recurrent GBM<br />

patients who received anti-angiogenic treatment and chemoradiation<br />

(AAT�) to 7 patients who received chemotherapy and/or radiotherapy<br />

without anti-angiogenic therapy, or no treatment (AAT-). TAMs were<br />

morphologically and phenotypically identified with flow cytometry and<br />

immunohistochemistry (IHC) with CD68, CD11b, CD14, and CD163<br />

markers. All specimens were obtained from the Department <strong>of</strong> Pathology at<br />

Massachusetts General Hospital and clinical information gained through<br />

review <strong>of</strong> the patients’ records. Results: Using flow cytometry, we observed<br />

an increase in CD11b�CD14� cells in the AAT� patients compared to<br />

AAT- patients. Using IHC analysis, we observed a significant increase in<br />

CD68� macrophages in the tumor bulk (p�0.01) and infiltrative areas<br />

(p�0.05) in AAT� versus AAT- patients. We also observed a significant<br />

increase in CD11b� myeloid cells in the tumor bulk (p�0.01) and a<br />

significant increase in CD163� cells in the infiltrative areas (p�0.05) in<br />

the AAT� group. Finally, we noted a trend toward an increase in CD163�<br />

cells in the tumor bulk (p�0.087) in the AAT� versus the AAT- patients.<br />

Conclusions: Patients with recurrent GBM after antiangiogenic therapy<br />

showed a significant increase in CD68� TAMs and in CD11b� cells in the<br />

tumor bulk. Additionally, antiangiogenic treatment induced an increase in<br />

CD68� and CD163� TAMs in the infiltrative region. These data indicate<br />

that TAMs may participate in escape from antiangiogenic therapy and may<br />

represent a future therapeutic target in recurrent GBM.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


118s Central Nervous System Tumors<br />

2011 <strong>Clinical</strong> Science Symposium, Sat, 1:15 PM-2:45 PM<br />

Modulation <strong>of</strong> antiangiogenic resistance: Targeting the JAK/STAT3 pathway<br />

in glioblastoma. Presenting Author: John Frederick De Groot, University<br />

<strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: Determining the mechanism <strong>of</strong> treatment failure <strong>of</strong> antiangiogenic<br />

therapies would provide insight into either potential combination<br />

approaches or salvage therapies. Methods: Tumor and monocyte expression<br />

<strong>of</strong> phosphorylated-signal transducer and activator <strong>of</strong> transcription 3 (p-<br />

STAT3) was compared between recurrent glioblastoma patients treated<br />

with either conventional chemotherapy or bevacizumab (BEV), and secondarily<br />

validated in murine model systems <strong>of</strong> intracerebral glioma treated with<br />

either BEV or cediranib. The JAK/STAT3 inhibitor AZD1480 alone and in<br />

combination with cediranib was evaluated in immunocompetent mice<br />

bearing intracerebral GL261 xenografts to evaluate therapeutic synergy.<br />

Results: Human glioblastoma patients failing BEV have an increase in<br />

intratumoral p-STAT3- expression with a mean number <strong>of</strong> p-STAT3<br />

expressing cells <strong>of</strong> 36.5 � 8.6% (n�7) compared to a mean <strong>of</strong> 20.2 � 4%<br />

(n�5) in patients never receiving antiangiogenic therapy. In a separate<br />

cohort <strong>of</strong> recurrent glioblastoma patients, the up-regulation <strong>of</strong> p-STAT3<br />

was detected in monocytes within 24 hours after initial BEV administration.<br />

In intracranial xenograft models, both BEV and cediranib extended<br />

median animal survival time, but during treatment failure BEV increased<br />

the mean percentage <strong>of</strong> p-STAT3-expressing cells to 24.6 � 6.4%<br />

(P�0.0011), and cediranib increased this to 16.2 � 1.8% (P�0.0125)<br />

relative to 9.0 � 4.4% in controls. Administration <strong>of</strong> the JAK/STAT3<br />

inhibitor AZD1480 alone and in combination with cediranib reduced tumor<br />

hypoxia and infiltration <strong>of</strong> VEGF inhibitor-induced p-STAT3 macrophages.<br />

The combination reduced tumor volume by 80% compared to untreated<br />

controls, and by 65% compared to cediranib or AZD1480 treatment alone.<br />

Microvascular density and tumor de-differentiation were reduced, indicating<br />

that up-regulation <strong>of</strong> the STAT3 pathway can mediate resistance to<br />

antiangiogenic therapy. Conclusions: Cumulatively, these data suggest that<br />

a prominent mechanism <strong>of</strong> failure <strong>of</strong> anti-VEGF therapy in malignant<br />

glioma patients involves up-regulation <strong>of</strong> the STAT3 pathway, and resistance<br />

to antiangiogenic therapy can be modulated with inhibitors <strong>of</strong> the<br />

JAK/STAT pathway.<br />

2013 Poster Discussion Session (Board #1), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Phase I safety and pharmacokinetic (PK) study <strong>of</strong> veliparib in combination<br />

with whole brain radiation therapy (WBRT) in patients (pts) with brain<br />

metastases. Presenting Author: Minesh P. Mehta, Northwestern University,<br />

Chicago, IL<br />

Background: Veliparib is an oral PARP-1 and -2 inhibitor that enhances the<br />

antitumor activity <strong>of</strong> DNA damaging agents including radiation therapy in<br />

vivo. In pre-clinical models, veliparib crosses the blood-brain barrier. This<br />

ongoing phase I dose-escalation study evaluates the safety, PK, and<br />

provides preliminary antitumor activity <strong>of</strong> veliparib in combination with<br />

WBRT in pts with brain metastases. Methods: Pts with brain metastases<br />

from non-CNS primary solid malignancy, adequate organ function, RPA<br />

Class 2, and KPS �70 were treated with WBRT (37.5 Gy in 15 fractions or<br />

30 Gy in 10 fractions) QD with veliparib BID with every fraction <strong>of</strong> WBRT in<br />

escalating doses <strong>of</strong> 10, 20, 30, 50, 100, 150, and 200 mg; the final WBRT<br />

fraction was followed by 1 extra day <strong>of</strong> veliparib. Safety, PK, and tumor<br />

response by RECIST were assessed. Results: At the time <strong>of</strong> reporting 59 pts<br />

(M/F, 21/38; median age 57 y) had been treated. Baseline KPS was 70,<br />

80, 90, and 100 in 6.8, 32.2, 40.7, and 20.3% pts, respectively; primary<br />

tumor types were breast (n�20), NSCLC (n�20), melanoma (n�9),<br />

colorectal (n�2), and others (n�8); 71.2% pts had multiple lesions; and<br />

18.6% had prior brain SRS. Grade 3/4 treatment-emergent adverse events<br />

(TEAEs; �5%) were fatigue (6.8%), anemia (5.1%), hyponatraemia<br />

(5.1%), and thrombocytopenia (5.1%); other TEAEs (�20%) were fatigue<br />

(57.6%), headache (42.4%), nausea (40.7%), alopecia (28.8%), vomiting<br />

(22%), radiation skin reactions (22%), and decreased appetite (22%). PK<br />

<strong>of</strong> veliparib were approximately dose-proportional, with oral clearance <strong>of</strong><br />

21.6 � 14.2 L/h (mean � SD, n�45), minimal drug accumulation at day<br />

15, and no significant effect <strong>of</strong> food on bioavailability. Tumor response was<br />

evaluable in 48 pts. Best tumor response and median survival were 37.5%<br />

and 10 months (m) for NSCLC, and 52.9% and 12.5 m for breast cancer<br />

(excluding pts with leptomeningeal disease). Conclusions: Addition <strong>of</strong><br />

veliparib up to 200 mg BID was well tolerated with concurrent WBRT and<br />

dose escalation ongoing. The PK <strong>of</strong> veliparib was dose proportional with no<br />

food effect. Preliminary antitumor activity is encouraging and informative<br />

for the design <strong>of</strong> more definitive trials.<br />

2012 <strong>Clinical</strong> Science Symposium, Sat, 1:15 PM-2:45 PM<br />

Effects <strong>of</strong> cediranib (VEGF signaling inhibitor) on edema in newly diagnosed<br />

glioblastoma patients during initial chemoradiation. Presenting<br />

Author: Pavlina Polaskova, Martinos Center for Biomedical Imaging,<br />

Massachusetts General Hospital, Charlestown, MA<br />

Background: A significant benefit <strong>of</strong> antiangiogenic therapy is control <strong>of</strong><br />

brain edema. We evaluated the impact <strong>of</strong> adding cediranib to standard<br />

chemoradiation (CRT) on peritumoral edema in patients with newly<br />

diagnosed glioblastoma(GBM) during the initial 6 weeks <strong>of</strong> CRT. Methods:<br />

Two cohorts <strong>of</strong> patients were enrolled in two clinical trials. The control<br />

group (N�13) received radiation for 6 weeks plus temozolomide. The<br />

cediranib (CED) group received standard CRT plus daily cediranib (N�34).<br />

MRIs were performed at baseline and weekly during CRT. Volumes <strong>of</strong><br />

interest (VOIs) were drawn outlining the enhancing tumor on T1-weighted<br />

post contrast images and the abnormal FLAIR hyperintensity. ADC (apparent<br />

diffusion coefficient) maps were calculated from diffusion-weighted<br />

images and histograms <strong>of</strong> the distribution <strong>of</strong> ADC values created for each<br />

visit using the baseline FLAIR VOI to characterize the peritumoraledema.<br />

Patients were on stable or decreasing doses <strong>of</strong> steroids. Results: In the CED<br />

group, T1 and FLAIR VOI decreased during CRT vs controls where T1 VOI<br />

did not change and FLAIR VOI increased. By the end <strong>of</strong> CRT, the mode <strong>of</strong><br />

the ADC histogram in the CED group shifted to the left while the mode <strong>of</strong><br />

the controls shifted to the right. The skewness, a measure <strong>of</strong> asymmetry <strong>of</strong><br />

the distribution, increased in the CED group and decreased in controls.<br />

Conclusions: In patients with newly diagnosed GBM treated with CRT and<br />

cediranib, tumor volume decreased on T1 and FLAIR images whereas the<br />

FLAIR volume significantly increased in the control group suggesting<br />

increased edema. The shift <strong>of</strong> mode to the right and decreasing skewness in<br />

controls (indicating an increase in the proportion <strong>of</strong> very high ADC values)<br />

suggests that adding cediranibprevented the development <strong>of</strong> edema and<br />

contributed to the resolution <strong>of</strong> existing edema. Preventing the edema by<br />

adding anti-VEGF treatment may improve the tolerability <strong>of</strong> CRT for GBM<br />

patients.<br />

T1 VOI<br />

(ml)<br />

FLAIR VOI<br />

(ml)<br />

ADC histogram mode<br />

(x10 -4 �m 2 /ms)<br />

ADC histogram<br />

skewness<br />

Baseline 6 wks Baseline 6 wks Baseline 6 wks Baseline 6 wks<br />

CED 18.0 11.5 36.0 32.9 10.8 10.1 2.64 2.97<br />

Control 14.8 14.5 25.9 57.1* 9.5 11.8* 2.62 2.42<br />

* Significant difference between CED and controls (p � .05).<br />

2014 Poster Discussion Session (Board #2), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

FIP200 and Rb1 expression in CNS metastasis from breast cancer (CNS<br />

met): Potential predictors <strong>of</strong> patient outcome. Presenting Author: Nooshin<br />

Hashemi Sadraei, Cleveland Clinic Foundation, Cleveland, OH<br />

Background: Patients with CNS met have a poor outcome with significant<br />

variation in overall survival. No marker to predict survival exists. Rb1 is a<br />

negative regulator <strong>of</strong> the cell cycle, and FiP200 is an upstream signaling<br />

node that is distributed between the nucleus and cytoplasm. Nuclear<br />

FIP200 inhibits cell proliferation by promoting expression <strong>of</strong> Rb1, whereas<br />

cytoplasmic FIP200 promotes cell survival through autophagy. Previously<br />

FIP200 deletion/mutation has been reported in 20% <strong>of</strong> primary invasive<br />

breast cancer [BR ca] patients. FIP200 cellular localization and genetic<br />

alterations have not been examined in CNS met. Methods: A retrospective<br />

analysis <strong>of</strong> BR ca and CNS met was performed based on tissue availability<br />

from 2 institutions. FIP200 and Rb1 expression and localization was<br />

evaluated by immunohistochemistry. Genetic alterations were evaluated by<br />

DNA array analysis. Results: 80 patients (42 BR ca and 38 CNS met) were<br />

identified. Overall CNS met samples had significantly higher levels <strong>of</strong><br />

cytoplasmic FIP200 as compared to BR ca (52% vs 24% respectively,<br />

p�0.0002) and increased expression <strong>of</strong> FIP200 around areas <strong>of</strong> hypoxia/<br />

necrosis, consistent with increased autophagy. There was also a significant<br />

increase in expression <strong>of</strong> nuclear FIP200 and Rb1 in the CNS met<br />

compared to BR ca (p�0.0007 and p�0.0055 respectively). Median<br />

survival from development <strong>of</strong> CNS met was longer in the group with high<br />

levels <strong>of</strong> nuclear Rb1; 14 (3-27) vs 20 (12-33) months (low expression �<br />

20% vs high � 20%, p�0.1). The same finding was observed with FIP200,<br />

longer survival was observed with high nuclear expression; 16 (7-33) vs 19<br />

(0.4-27) months (low expression �20% vs high � 20%, p�0.2). Our DNA<br />

analysis for copy number variation and LOH in CNS met revealed no<br />

deletion in Rb1 or FIP200. Conclusions: The pattern <strong>of</strong> expression <strong>of</strong> Rb1<br />

and FIP200 in CNS met is different from BR ca. Notably, cytoplasmic<br />

FIP200 which promotes cell survival is highly expressed in CNS mets vs BR<br />

ca. This suggests metastatic breast cancer cells utilize autophagy as a<br />

survival mechanism. In addition, there is a trend to better survival <strong>of</strong> those<br />

patients with CNS met who express high nuclear FIP200 and Rb1, both <strong>of</strong><br />

which have anti-proliferative roles in the nucleus.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


2015 Poster Discussion Session (Board #3), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Intraventricular (IVe) topotecan for women with neoplastic meningitis (NM)<br />

associated with �responsive� malignancies. Presenting Author: Kurt A.<br />

Jaeckle, Mayo Clinic, Jacksonville, FL<br />

Background: A prior study <strong>of</strong> intra-CSF topotecan (TOPO) for unselected pts<br />

w/ NM reported PFS 6 mo <strong>of</strong> 19%, and OS <strong>of</strong> 15 wks (Groves, NeuroOncol<br />

2008;10:208). We postulated that greater activity might occur in pts w/<br />

malignancies considered sensitive to topoisomerase inhibitors. Methods:<br />

We reviewed outcome <strong>of</strong> women with NM and adenocarcinoma <strong>of</strong> the<br />

breast, ovary or lung receiving IVe TOPO (0.4 mg 2x/wk x 4wk, Q wk x 4, Q<br />

2wk x 2, Q mo x 3, Q 2mo x 3, and Q 3mo x 4) until progression (PROG) or<br />

adverse events (AE). All had baseline CSF cytology, and MRI <strong>of</strong> brain and<br />

spine. CSF cytology was obtained at each treatment (Rx), and brain/spine<br />

MRI Q 3mo. Neuro-specific PROG was defined as recurrent � CSF cytology;<br />

PROG <strong>of</strong> NM on MRI; all-cause neurologic worsening; pt refusal; or death.<br />

PFS/OS were measured from 1st TOPO Rx. All pts signed consent; the study<br />

was IRB approved. Results: 17 women (breast -12; lung-3; ovary - 2) were<br />

treated via Ommaya reservoirs; 7 (41%) had VP shunts w/ valves, adjusted<br />

for Rx. Median (med) age was 53 (41-79), and KPS 70 (50-90). At<br />

presentation, 11(65%) had � CSF cytology and 14 (82%) had NM on MRI<br />

[brain-11 (65%); spine-10 (59%)]. 15 (88%) had no prior intrathecal Rx.<br />

13 pts (76%) received focal RT for CNS disease, and 8 (47%), chemotherapy<br />

for extracranial disease. Med.number <strong>of</strong> Rx were 13/pt (range,<br />

3-50); med. duration <strong>of</strong> TOPO Rx was 14 wks (range, 1-109). Med.<br />

neuro-specific PFS was 13 wks; PFS6 was 41%, and PFS12, 29 %. Med.<br />

OS was 33 wks (range, 5-180), w/ 4 alive at 13�, 30�, 33�, and 180�<br />

wks. 4 pts (24%) lived � 95 wks. Of 11 w/ baseline � CSF cytology, 7<br />

(64%) cleared CSF <strong>of</strong> malignant cells (med. duration clear � 47 wk (range,<br />

9-104). AE included arachnoiditis (18%), leukoencephalopathy (18%),<br />

and Ommaya infections (12%). Rx was stopped for neuro PROG (29%);<br />

systemic PROG (23%); refusal (18%); AE (12%); or persistent negative<br />

CSF (6%); 12% are still on Rx. Conclusions: Promising activity <strong>of</strong> IVe TOPO<br />

was observed in women with NM from breast, lung and ovarian cancer. PFS<br />

6 and 12, OS and cytologic response were twice that noted in prior studies<br />

<strong>of</strong> NM pts w/ unselected malignancies. This data supports our plan for a<br />

phase II study targeting this select cohort.<br />

2017 Poster Discussion Session (Board #5), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Modeling radiation dose to circulating lymphocytes during brain tumor<br />

treatment: Effects <strong>of</strong> target volume, dose rate, and treatment technique.<br />

Presenting Author: Susannah G. Yovino, Johns Hopkins University School<br />

<strong>of</strong> Medicine, Baltimore, MD<br />

Background: Severe treatment-related lymphopenia (TRL) occurs in 40% <strong>of</strong><br />

glioblastoma patients despite minimal radiation (RT) doses to bone marrow<br />

or nodal sites. In glioblastoma, TRL is associated with decreased survival.<br />

To explain the lymphopenia, we sought to estimate radiation doses received<br />

by circulating lymphocytes during partial brain RT. Methods: An in-house<br />

computer program linked to treatment planning s<strong>of</strong>tware was used to<br />

calculate the mean radiation dose to circulating blood (DCB) and the<br />

fraction <strong>of</strong> blood receiving �0.5 Gy. The model also studied the impact <strong>of</strong><br />

different target volumes (PTV), dose rates (DR), and delivery techniques<br />

(IMRT, 3D-CRT). Results: The mean DCB for a 60-Gy course (8-cm<br />

diameter PTV, dose rate 600 MU/minute) was 2.2 Gy. With this the entire<br />

blood pool receives a lymphotoxic dose <strong>of</strong> �0.5 Gy. DCB is correlated with<br />

fraction number, PTV size, and DR. Regardless <strong>of</strong> dose rate or delivery<br />

technique, the percent <strong>of</strong> circulating blood receiving �0.5 Gy approached<br />

100% as the number <strong>of</strong> fractions increased. Changing dose rate had<br />

minimal effects on mean DCB (3.1Gy for 300 MU/min vs 2.2 Gy for 1200<br />

MU/min). Smaller PTV size reduced the percent <strong>of</strong> blood receiving �0.5 Gy<br />

(15% for 2-cm diameter PTV vs 100% for 8-cm PTV). Conclusions:<br />

Standard RT for brain tumors delivers a lymphotoxic radiation dose to<br />

circulating blood. Altering dose rate may initially affect DCB, but advantages<br />

disappear over the course <strong>of</strong> 30 fractions. Marked reductions in target<br />

size appear to be the best way to avoid radiation injury to normal circulating<br />

lymphocytes. Other novel approaches are needed to limit radiation exposure<br />

to circulating lymphocytes given evidence associating lymphopenia<br />

with poorer outcomes in cancer patients.<br />

Central Nervous System Tumors<br />

119s<br />

2016 Poster Discussion Session (Board #4), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Addition <strong>of</strong> upfront whole-brain radiotherapy to surgery or stereotactic<br />

radiosurgery versus surgery or stereotactic radiosurgery alone for treatment<br />

<strong>of</strong> brain metastases: A systematic review and meta-analysis. Presenting<br />

Author: Yu Yang Soon, National University Cancer Institute, Singapore<br />

Background: The benefits <strong>of</strong> adding upfront whole-brain radiotherapy<br />

(WBRT) to surgery or stereotactic radiosurgery (SRS) when compared to<br />

surgery or SRS alone for treatment <strong>of</strong> brain metastases are unclear. We<br />

performed a systematic review and meta-analysis <strong>of</strong> published randomized<br />

controlled trials (RCT) to determine the efficacy and safety <strong>of</strong> additional<br />

upfront WBRT. Methods: We searched MEDLINE, EMBASE, CENTRAL<br />

from date <strong>of</strong> inception and annual meeting proceedings <strong>of</strong> ASCO and<br />

ASTRO from 1999 to September 2011 for RCTs comparing surgery or SRS<br />

plus WBRT with surgery or SRS alone for treatment <strong>of</strong> brain metastases.<br />

The primary outcome was overall survival (OS). Secondary outcomes<br />

include progression free survival (PFS), local and distant intracranial<br />

disease progression, neurocognitive function (NF), quality <strong>of</strong> life (Qol) and<br />

neurological toxicity. Hazard ratios (HR), confidence intervals (CI), p values<br />

(p) were estimated with random effects models using Revman 5.1. Results:<br />

We found five RCTs including 663 patients with one to four brain<br />

metastases. Adding upfront WBRT decreased the one-year incidence <strong>of</strong> any<br />

intracranial disease progression from 73-76% to 22 - 47% but did not<br />

improve OS (HR 1.11, 95%CI 0.83 - 1.48, p � 0.47) and PFS (HR 0.76,<br />

95%CI 0.53 - 1.10, p � 0.14). Subgroup analyses showed that the effects<br />

on overall survival are similar regardless <strong>of</strong> types <strong>of</strong> focal therapy used,<br />

number <strong>of</strong> brain metastases, dose and sequence <strong>of</strong> WBRT. The effects <strong>of</strong><br />

upfront WBRT on NF, Qol and neurological toxicity were variable.<br />

Conclusions: Adding upfront WBRT to surgery or SRS significantly decreased<br />

any intracranial disease progression at one year but did not improve<br />

overall and progression free survival and produced variable effects on<br />

neurocognitive function, quality <strong>of</strong> life and neurological toxicity when<br />

compared with surgery or SRS alone. Future research should focus on<br />

developing more effective approaches to characterize and ameliorate the<br />

potential neurological toxicity <strong>of</strong> WBRT.<br />

2018 Poster Discussion Session (Board #6), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Phase II trial <strong>of</strong> sunitinib as adjuvant therapy after stereotactic radiosurgery<br />

(SRS) in patients with 1-3 newly diagnosed brain metastases. Presenting<br />

Author: David M. Peereboom, Cleveland Clinic, Cleveland, OH<br />

Background: For patients with 1-3 brain metastases, standard therapy after<br />

SRS is adjuvant whole brain radiotherapy (WBRT). SRS without WBRT<br />

carries a higher rate <strong>of</strong> brain relapse. Due to concerns about neurologic<br />

sequelae <strong>of</strong> WBRT, however, many patients and physicians opt to defer<br />

WBRT until the time <strong>of</strong> central nervous system (CNS) progression. This trial<br />

used sunitinib as an alternative to WBRT for post-SRS adjuvant therapy.<br />

Sunitinib inhibits vascular endothelial growth factor signaling, and we<br />

hypothesized that it would prevent growth <strong>of</strong> microscopic brain metastases<br />

presumed to be present. The objective was to use adjuvant sunitinib after<br />

SRS to prevent the emergence <strong>of</strong> new or progressive disease in the brain or<br />

at the site <strong>of</strong> SRS and to preserve neurocognitive function. Methods:<br />

Eligible patients had 1-3 newly diagnosed brain metastases, RTOG RPA<br />

class 1-2, and started sunitinib � 1 month after SRS and baseline<br />

neuropsychological testing (NPT). Patients with controlled systemic disease<br />

were allowed to continue chemotherapy for their primary disease<br />

according to a list <strong>of</strong> published regimens (therapy � sunitinib) included in<br />

the protocol. Patients received sunitinib 37.5 or 50 mg/d days 1-28 every<br />

42 days until CNS progression. NPT and MRIs were obtained every 2<br />

cycles. The primary endpoint was the rate <strong>of</strong> CNS progression at 6 months<br />

(PFS6) after SRS. Results: Fourteen patients enrolled. The median age was<br />

59 (range 46-80). Main histologies: lung 36%, breast 21%, melanoma<br />

14%. Toxicities: Grade 4: neutropenia [ANC] (1 pt); Grade 3: fatigue (5),<br />

ANC (2), rash (1). Dose reduction due to toxicity: 1 pt (to 37.5 mg/d). The<br />

CNS PFS6 and PFS12 were 50% � 13% and 43% � 13%, respectively.<br />

The median PFS was 6.6 months (95% C.I. 1.5-19). NPT results will be<br />

reported at the meeting. Conclusions: Sunitinib after SRS for 1-3 brain<br />

metastases was well tolerated with a PFS6 <strong>of</strong> 50%. The use <strong>of</strong> novel agents<br />

to prevent progressive brain metastasis after SRS requires the incorporation<br />

<strong>of</strong> chemotherapy regimens to control the patient’s primary disease.<br />

Future trials should continue to explore the paradigm <strong>of</strong> secondary<br />

chemoprevention <strong>of</strong> brain metastases after definitive local therapy (surgery<br />

or SRS).<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


120s Central Nervous System Tumors<br />

2019 Poster Discussion Session (Board #7), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

IDH1 status and survival benefit from surgical resection <strong>of</strong> enhancing and<br />

nonenhancing tumor in malignant astrocytomas. Presenting Author: Daniel<br />

P. Cahill, Massachusetts General Hospital, Boston, MA<br />

Background: The value <strong>of</strong> maximal safe resection for malignant astrocytic<br />

gliomas (AA, WHO Grade III anaplastic astrocytoma and GBM, WHO Grade<br />

IV glioblastoma) has sometimes been controversial, because <strong>of</strong> confounding<br />

between measures <strong>of</strong> surgical resection and other prognostic factors.<br />

IDH1 gene mutations are associated with improved survival in glioma<br />

patients, and are thought to identify tumors with a distinct molecular<br />

evolutionary origin. We sought to determine the prognostic impact <strong>of</strong><br />

surgical resection on survival after controlling for IDH1 status in malignant<br />

astrocytomas. Methods: <strong>Clinical</strong> parameters including preoperative and<br />

postoperative MRI-based tumor volume were recorded prospectively on<br />

407 malignant astrocytoma patients – AA (n�157) and GBM (n�250).<br />

IDH1 status was assessed by sequencing and R132H-specific immunohistochemistry.<br />

Results: The measures <strong>of</strong> surgical resection associated with<br />

longer survival differed between IDH1 wild-type and mutant tumors. In<br />

multivariate analyses <strong>of</strong> IDH1 wild-type tumors (controlling for age,<br />

Karn<strong>of</strong>sky performance score, tumor location, and tumor grade), residual<br />

postoperative enhancement was associated with a median survival <strong>of</strong> 9.9<br />

mo vs. 17.4 mo with no enhancement (HR�1.73, 95% CI, 1.19-2.52,<br />

p�.004). Residual non-enhancing disease, however, was not associated<br />

with survival (scored as continuous volumetric cc, 95% CI 0.99-1.01,<br />

p�.608). These results are consistent with prior studies <strong>of</strong> GBM, which are<br />

largely IDH1 wild-type lesions (Lacroix et al., J Neurosurg 95:190-8,<br />

2001). In contrast, in IDH1 mutant tumors, both residual enhancing<br />

(HR�7.93, 95%CI 1.14-55.22, p�.037) and non-enhancing (HR�1.03,<br />

95% CI 1.01-1.05, p�.005) postoperative tumor burden were associated<br />

with worse survival. Conclusions: These data suggest surgical resection in<br />

malignant astrocytic gliomas may be individualized based on IDH1 genotype.<br />

IDH1 mutant tumors have a better baseline overall prognosis,<br />

therefore more aggressive surgery and tolerance <strong>of</strong> temporary peri-operative<br />

neurologic deficits can be weighed in an attempt to gain the additional<br />

survival benefit that appears to be associated with reducing non-enhancing<br />

tumor burden.<br />

2021 Poster Discussion Session (Board #9), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

MET and ALK in glioblastoma multiforme (GBM): Comparison <strong>of</strong> IHC and<br />

FISH. Presenting Author: Kimary Kulig, National Comprehensive Cancer<br />

Network, Fort Washington, PA<br />

Background: GBM, the most common and lethal malignant primary brain<br />

tumor in adults, has an overall one-year life expectancy <strong>of</strong> 33.7%.<br />

Personalized medicine on the basis <strong>of</strong> predictive biomarkers remains<br />

elusive in GBM; however a recent case report showed clinical improvement<br />

after treatment with a mesenchymal-epithelial transition (MET) inhibitor.<br />

The authors <strong>of</strong> that report ascribe clinical response to the MET inhibitor,<br />

but the drug is also an inhibitor <strong>of</strong> anaplastic lymphoma kinase (ALK). We<br />

explored the co-expression, gain, or amplification <strong>of</strong> both MET and ALK by<br />

IHC and FISH in GBM. Methods: A convenience sample <strong>of</strong> 56 available<br />

tumors from gross-resected GBM cases within the Duke Brain Tumor tissue<br />

bank was stained for MET using clone 8F11 (Leica) and for ALK using clone<br />

5A4 (Novocastra). FISH employed the LSI ALK (2p23) breakpointspanning<br />

dual-color DNA probe and the LSI D7S486 (MET)/CEP 7 dual<br />

color DNA probe (Vysis). Results: Of 56 GBM cases, MET was expressed in<br />

69.6% and ALK in 17.9% by IHC. By FISH, gain or amplification was found<br />

in 100% <strong>of</strong> cases for MET and in 48.2% for ALK. Co-expression <strong>of</strong> MET and<br />

ALK by IHC was 14.3% and 48.2% by FISH. Co-expression by either IHC or<br />

FISH was also 48.2%. OS estimation for this cohort is premature, with<br />

77% <strong>of</strong> patients still alive. Conclusions: MET and ALK are (co-)expressed in<br />

a significant proportion <strong>of</strong> GBM. IHC detection <strong>of</strong> MET and ALK did not<br />

correlate well with FISH results, suggesting that gene gain or amplification<br />

does not necessarily lead to abnormal MET and ALK protein expression or<br />

that IHC methods or antibodies used can be optimized. Prior studies<br />

reported MET amplification in GBM as low as 4%. Our study indicates that<br />

MET gain/amplification and protein expression may be much higher, having<br />

implications for MET-targeted therapy. Additionally, co-expression <strong>of</strong> ALK<br />

and MET has implications for dual inhibitors <strong>of</strong> these signaling pathways as<br />

well as resistance mechanisms. Our continued work will explore significance<br />

<strong>of</strong> MET and ALK as predictors <strong>of</strong> therapeutic response, with careful<br />

control for clinically relevant patient and disease characteristics.<br />

Biomarker IHC �<br />

FISH �<br />

(gain or amplification) IHC- FISH-<br />

MET 39 56 17 0<br />

ALK 10 27 46 29<br />

MET � ALK 8 27 15 0<br />

2020 Poster Discussion Session (Board #8), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Genomic characterization <strong>of</strong> meningiomas. Presenting Author: Priscilla<br />

Kaliopi Brastianos, Dana-Farber Cancer Institute, Boston, MA<br />

Background: Understanding the genetic alterations in cancer has lead to<br />

groundbreaking discoveries in targeted therapies. Meningiomas are among<br />

the most common primary brain tumors, with approximately 18,000 new<br />

cases diagnosed annually. Though certain genes have been associated with<br />

the development <strong>of</strong> meningiomas, most notably the tumor suppressor gene<br />

neur<strong>of</strong>ibromatosis 2 (NF2), the genetic changes that drive meningiomas<br />

remain poorly understood. Our objective was to comprehensively characterize<br />

the somatic genetic alterations <strong>of</strong> meningiomas to gain insight into the<br />

molecular pathways that drive this disease. Methods: Fresh frozen specimens<br />

and paired blood were collected from 16 consented patients. DNA<br />

was extracted from regions <strong>of</strong> high tumor purity determined by evaluation <strong>of</strong><br />

H&E slides. Whole-genome sequencing from 10 tumor-normal pairs and<br />

whole-exome sequencing from 6 tumor-normal pairs was carried out. We<br />

performed an unbiased screen for point mutations, insertions-deletions,<br />

rearrangements and copy-number changes across the exomes and genomes.<br />

Recurrent (potential driver) events were then analyzed with<br />

additional algorithms for statistical significance. Results: Alterations in the<br />

NF2 gene were present in 9 <strong>of</strong> 16 patients. Multiple novel rearrangements<br />

and recurrent non-NF2 mutations were also identified in the cohort.<br />

Massive genomic rearrangement termed chromothripsis was observed in<br />

chromosome 1 in one sample, which has never previously been described in<br />

meningiomas, and represents a potentially new mechanism <strong>of</strong> malignant<br />

transformation in this tumor type. Conclusions: While NF2 mutations<br />

appear to drive a majority <strong>of</strong> these tumors, our analysis has uncovered<br />

additional potential driver genes in meningiomas, particularly in those<br />

tumors negative for NF2 alterations. To our knowledge, this is the first study<br />

to comprehensively characterize the totality <strong>of</strong> somatic genetic alterations<br />

in meningiomas, and brings us closer to the development <strong>of</strong> new therapeutic<br />

targets for this disease.<br />

2022 Poster Discussion Session (Board #10), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Pilocytic astrocytomas in adults: A retrospective study <strong>of</strong> 125 patients.<br />

Presenting Author: Brett James Theeler, University <strong>of</strong> Texas M. D. Anderson<br />

Cancer Center, Houston, TX<br />

Background: Pilocytic astrocytomas (PAs) are rare primary brain tumors in<br />

adults. The natural history is poorly defined with the largest published<br />

series including only 30-40 patients. To better define clinical characteristics<br />

<strong>of</strong> adult PAs, we present an analysis <strong>of</strong> adult PAs at MD Anderson<br />

Cancer Center. Methods: We conducted an IRB-approved, retrospective<br />

chart review <strong>of</strong> patients �18 years diagnosed with PAs from 1990-2011 at<br />

our institution. Results: We identified 125 patients with a median age <strong>of</strong> 29<br />

(range 18-72y), female to male ratio <strong>of</strong> 1.7:1, and median follow-up <strong>of</strong> 62<br />

months. The most common anatomic locations were brainstem 22%,<br />

cerebellum 14%, intraventricular 14%, and cortical/lobar 14% (36% other<br />

locations). 21% <strong>of</strong> cases had an initial pathologic diagnosis discordant with<br />

neuropathologic review at our institution. Comorbidities included neur<strong>of</strong>ibromatosis<br />

type 1 (5 pts) and a history <strong>of</strong> asthma or autoimmune disease<br />

(14%). 38 patients had a gross total (GTR), 55 a sub-total resection (STR)<br />

and the remainder a biopsy. 62 patients were treated with radiotherapy<br />

(RT): 45 as adjuvant (adj), 17 at recurrence; 2 received RT in both adj and<br />

recurrent settings. 73 (58%) patients were stable after initial treatment<br />

(surgery �/- adj RT). Median progression free survivals (mPFS) based on<br />

upfront treatment as follows: GTR only (n�30) - not been reached, STR<br />

only (n�32) 226.6 months, GTR plus adj RT (n�8) 13.2 months, STR plus<br />

adj RT (n�23) is 52.4 months (p�0.026, logrank test for trend). At last<br />

follow-up, 77% <strong>of</strong> all patients were stable and 13% were deceased.<br />

Conclusions: This is the largest series <strong>of</strong> adult PAs reported to date.<br />

Extracerebellar location, slight female predominance, and history <strong>of</strong><br />

allergic/autoimmune disease were associations found in this cohort.<br />

Neuropathologic review is essential to avoid misdiagnosis. A significant<br />

subset <strong>of</strong> adult PAs behave more aggressively than expected for a grade I<br />

tumor with over 40% <strong>of</strong> patients experiencing recurrence after initial<br />

treatment. Adj RT following GTR or STR did not improve PFS, and a trend<br />

towards worse PFS with adj RT was observed although these analyzed<br />

subgroups were small. These results may help provide a framework for<br />

prospective studies in this tumor type.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


2023 Poster Discussion Session (Board #11), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Recurrent genetic alterations in primary central nervous system lymphoma<br />

<strong>of</strong> immunocompetent patients. Presenting Author: Alberto Gonzalez, APHP,<br />

UPMC, CRICM, UMR975, Hopital Pitié-Salpêtrière, Paris, France<br />

Background: Little is known about the molecular pathogenesis <strong>of</strong> primary<br />

central nervous system lymphoma (PCNSL) in immunocompetent patients.<br />

Our objective was to identify the genetic changes involved in PCNSL<br />

oncogenesis and evaluate their clinical relevance. Methods: Twenty nine<br />

and four newly diagnosed, HIV-negative PCNSL patients were investigated<br />

using high-resolution single nucleotide polymorphism (SNPa) arrays (Infinium<br />

Illumina Human 610-Quad SNP array-Illumina; validated by realtime<br />

quantitative polymerase chain reaction) and whole-exome sequencing<br />

respectively. Molecular results were correlated with prognosis. Results: All<br />

PCNSLs were diffuse large B-cell lymphomas, and the patients received<br />

high-dose methotrexate-based polychemotherapy without radiotherapy as<br />

an initial treatment.SNPa analysis revealed recurrent large and focal<br />

chromosome imbalances that target candidate genes in PCNSL oncogenesis.<br />

The most frequent genomic changes were (i) 6p21.32 loss (79%),<br />

corresponding to the HLA locus; (ii) 6q loss (27-37%); (iii) CDKN2A<br />

homozygous deletions (45%); (iv) 12q12-q22 (27%); (v) chromosome<br />

7q21 and 7q31 gains (20%). Sequencing <strong>of</strong> matched tumor and blood<br />

DNA samples identified novel somatic mutations in MYD88 (L265P hot<br />

spot mutation) and TBL1XR1 in 38% and 14% <strong>of</strong> the cases, respectively.<br />

The correlation <strong>of</strong> genetic abnormalities with clinical outcomes using<br />

multivariate analysis showed that 6q22 loss (p�0.006 and p�0.01), and<br />

CDKN2A homozygous deletion (p�0.02 and p�0.01) were significantly<br />

associated with shorter progression free survival and overall survival.<br />

Conclusions: Our study identified novel genetic alterations in PCNSL, such<br />

as MYD88 and TBL1XR1 somatic mutations, which would both contribute<br />

to the constitutive activation <strong>of</strong> the NFkB signaling pathway and represent<br />

potential promising targets for future therapeutic strategies.<br />

2025 Poster Discussion Session (Board #13), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Impact <strong>of</strong> adjuvant anti-VEGF therapy on treatment-related pseudoprogression<br />

in patients with newly diagnosed glioblastoma receiving chemoradiation<br />

with or without anti-VEGF therapy. Presenting Author: Marco C. Pinho,<br />

Martinos Center for Biomedical Imaging, Massachusetts General Hopital,<br />

Boston, MA<br />

Background: Chemoradiation (CRT) can significantly modify the appearance<br />

<strong>of</strong> glioblastoma (GBM) on MRI, especially within the 3-month window<br />

after CRT. Pseudoprogression (PsP) is an increasingly recognized phenomenon<br />

in this scenario, and is thought to be secondary to increased<br />

permeability <strong>of</strong> the tumor vessels from irradiation, possibly enhanced by<br />

temozolomide (TMZ). We sought to determine if the addition <strong>of</strong> a VEGF<br />

signaling inhibitor (cediranib) during and after CRT had an impact on the<br />

frequency <strong>of</strong> PsP, by comparing two groups <strong>of</strong> patients with newly<br />

diagnosed GBMs. Methods: Two groups <strong>of</strong> patients enrolled in IRBapproved<br />

trials underwent serial MRIs at baseline, weekly during CRT and<br />

monthly thereafter. The control group received radiation plus TMZ, while<br />

the other group (CED) also received cediranib until progression (which was<br />

defined by RANO criteria) or toxicity. Patients that progressed up to 3<br />

months after CRT were considered to have apparent progression (AP) and<br />

kept on treatment. Lesions that subsequently stabilized/regressed were<br />

classified as PsP, while those that maintained growth despite treatment<br />

were classified as true early progression (EP). Results: Forty patients were<br />

enrolled in CED and 11 as controls. Ten patients from CED and three<br />

controls were excluded due to treatment discontinuation before 3 months<br />

(drug-related toxicity and enrollment in post-CRT trials respectively). Three<br />

patients in CED (6%) had AP (all confirmed as EP) and no patients fulfilled<br />

criteria for PsP. Six patients from control (54%) had AP; 3 (27%) were<br />

eventually classified as EP and 3 (27%) as PsP. The frequency <strong>of</strong> PsP was<br />

significantly higher in the control group (p�0.0086). Conclusions: The<br />

addition <strong>of</strong> a VEGF inhibitor as part <strong>of</strong> adjuvant treatment in this cohort <strong>of</strong><br />

glioblastoma patients prevented the development <strong>of</strong> early treatmentrelated<br />

effects (PsP). Suppressing the edema and mass effect that<br />

accompanies PsP may improve the tolerability <strong>of</strong> treatment. Additionally,<br />

this effect may be exploited as a strategy to make chemoradiation feasible<br />

in patients with large, unresectable tumors and/or poor baseline performance<br />

status.<br />

Central Nervous System Tumors<br />

121s<br />

2024 Poster Discussion Session (Board #12), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Blood alterations preceding clinical manifestation <strong>of</strong> glioblastoma. Presenting<br />

Author: Aysegul Ilhan-Mutlu, Medical University <strong>of</strong> Vienna, Vienna,<br />

Austria<br />

Background: Glioblastoma is the most common and aggressive primary<br />

brain tumour in adults. There is lack <strong>of</strong> knowledge on biochemical blood<br />

alterations prior to clinical diagnosis <strong>of</strong> glioblastoma. We had the rare<br />

opportunity to investigate selected blood markers from plasma samples<br />

taken 12, 42 and 72 months before tumor manifestation in a glioblastoma<br />

patient. This index patient was enrolled in the longitudinal population<br />

based Vienna Transdanube Aging Study (VITA), which prospectively collects<br />

blood plasma from 600 participants at baseline (age 75), 30 and 60<br />

months thereafter. Methods: We determined plasma levels <strong>of</strong> S100B,<br />

neuropeptide Y (NPY), secretagogin (SCGN), microRNA-21, microRNAlet7,<br />

microRNA-128, and microRNA-342-3p in all three blood samples<br />

from our index patient and in consecutive blood samples from five male and<br />

five female controls from the VITA cohort. These proteins and microRNAs<br />

were previously shown to be relevant for the pathobiology <strong>of</strong> glioblastomas.<br />

None <strong>of</strong> the controls had a malignant or neurological disease. Protein<br />

markers were analysed using commercially available ELISAs and microR-<br />

NAs were quantified using RT-qPCR. Results: Compared to baseline values,<br />

we found a significant increase <strong>of</strong> microRNA-21 (up to 4-fold) and<br />

microRNA-let7 (up to 7-fold) levels in the blood samples <strong>of</strong> our index<br />

patient, whereas control samples showed almost stable levels <strong>of</strong> these<br />

markers. There was no significant difference in S100B, NPY, SCGN,<br />

microRNA-128, and microRNA-342-3p plasma levels between the index<br />

patient and controls. Conclusions: Increases <strong>of</strong> microRNA-21 and microRNAlet7<br />

plasma levels may be associated with pre-clinical development <strong>of</strong><br />

glioblastoma.<br />

2026 Poster Discussion Session (Board #14), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

A single-institution phase II trial <strong>of</strong> radiation (RT), temozolomide (TMZ),<br />

erlotinib, and bevacizumab for initial treatment <strong>of</strong> glioblastoma (GBM).<br />

Presenting Author: Jennifer Leigh Clarke, University <strong>of</strong> California, San<br />

Francisco, San Francisco, CA<br />

Background: Standard treatment for GBM includes surgery followed by RT<br />

and TMZ, rarely a curative treatment. Both the EGFR and VEGF pathways<br />

are frequently overactive in GBM; we previously added erlotinib, an EGFR<br />

inhibitor, to RT and TMZ, with modest improvement in survival. The present<br />

study combined bevacizumab, a VEGF inhibitor, and erlotinib with RT and<br />

TMZ, with the goal <strong>of</strong> improving overall survival (OS). Methods: Treatment<br />

consisted <strong>of</strong> fractionated RT to 60 Gy with daily TMZ at 75 mg/m2/d and<br />

erlotinib 150-200 mg/d (or 500-600 mg/d for patients on enzymeinducing<br />

antiepileptic drugs [EIAEDs]). Bevacizumab was given at 10<br />

mg/kg every 2 wks, starting � 4 wks after surgery. After RT, adjuvant TMZ<br />

was given at 200 mg/m2/d x 5d per 28d cycle, with unchanged erlotinib<br />

and bevacizumab doses. Treatment was continued until progression or for<br />

12 mo, with an option to continue for up to 24 mo. OS and progression-free<br />

survival (PFS) from initial diagnosis were compared against institutional<br />

historical controls (recent prior up-front clinical trials including RT and<br />

TMZ). Results: 59 pts were enrolled; 12 were receiving EIAEDs. Median age<br />

was 54 yrs; median KPS was 90. 33% underwent gross total resection and<br />

53% subtotal resection. 16 pts had tumors with methylated MGMT<br />

(mMGMT), 26 with unmethylated MGMT (umMGMT), and 17 with unknown<br />

status. The most frequent related grade 3/4 AEs were lymphopenia<br />

(68%) thrombocytopenia (24%), neutropenia (10%), diarrhea (14%),<br />

weight loss (14%), and fatigue (12%). 1 pt died <strong>of</strong> aspergillosis. Median<br />

OS and PFS were 19.8 mo and 13.5 mo, respectively, vs. 18 mo and 8.6<br />

mo for the historical control. The hazard ratio (adj for age, KPS, and extent<br />

<strong>of</strong> surgery) for PFS was 0.7 (95% CI 0.49-0.99, p�0.04). Median OS in<br />

pts with mMGMT was 20.9 mo, vs. 17.5 mo in pts with umMGMT; median<br />

PFS was 14 mo vs. 12.4 mo, respectively. Conclusions: The combination <strong>of</strong><br />

bevacizumab, erlotinib, TMZ, and RT showed improved PFS but not OS vs.<br />

historical controls. Though the numbers were small, PFS and OS for pts<br />

with umMGMT were not significantly different from pts with mMGMT,<br />

suggesting that this combination may be more effective than standard<br />

therapy alone for pts with unMGMT tumors.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


122s Central Nervous System Tumors<br />

2027 Poster Discussion Session (Board #15), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Vorinostat, temozolomide, and bevacizumab for patients with recurrent<br />

glioblastoma: A phase I/II trial. Presenting Author: Katherine B. Peters,<br />

Duke University Medical Center, Durham, NC<br />

Background: Prognosis for recurrent glioblastoma (GBM) remains poor with<br />

median survival between 3 to 6 months. Use <strong>of</strong> anti-vascular endothelial<br />

growth factor inhibitor, bevacizumab (BEV), has advanced this area <strong>of</strong><br />

research, but continued studies have focused on whether the addition <strong>of</strong><br />

other chemotherapies can improve efficacy in recurrent GBM. Vorinostat, a<br />

small molecule inhibitor <strong>of</strong> histone deactylase (HDAC), has anti-tumor<br />

activity directly through HDAC inhibition and indirectly by promoting<br />

anti-angiogenesis. Its good oral bioavailability and favorable toxicity pr<strong>of</strong>ile<br />

make it a promising additive agent to standard therapy. Thus, we evaluated<br />

the efficacy <strong>of</strong> vorinostat in combination with BEV and daily temozolomide<br />

(TMZ) in recurrent GBM. Methods: This was a phase I/II open-label, single<br />

arm study in recurrent GBM patients. Primary endpoint was 6-month<br />

progression free survival (PFS). Secondary endpoints were safety/<br />

tolerability, radiographic response, PFS, and overall survival <strong>of</strong> recurrent<br />

GBM patients treated with BEV plus daily TMZ and vorinostat. Chief<br />

eligibility criteria included age � 18 years, KPS � 70, time interval � four<br />

weeks from previous treatment, and maximum <strong>of</strong> 2 prior progressions.<br />

Dosing regimen was as follows: BEV 10 mg/kg IV every two weeks, TMZ 50<br />

mg/m2 po daily, and vorinostat 400 mg po for 7 days on then 7 days <strong>of</strong>f in a<br />

28 day cycle. Results: 46 recurrent GBM patients were enrolled with 42 <strong>of</strong><br />

those patients receiving a vorinostat dose <strong>of</strong> 400 mg. Most common grade 2<br />

and above toxicities were leukopenia (36%), neutropenia (29%), fatigue<br />

(24%), and thrombocytopenia (19%). Serious toxicities included 4 grade 4<br />

toxicities (grade 4 hyperglycemia, pulmonary embolism, bowel perforation<br />

and intracranial hematoma) and 1 patient expired on day <strong>of</strong> informed<br />

consent due to pulmonary embolism (grade 5). With a median follow-up <strong>of</strong><br />

11.3 months (95% CI: 10.1, 13.1 months), the 6-month PFS was 52.4%<br />

(95% CI: 36.4%, 66.1%). Best radiographic responses were 2 complete<br />

responses, 17 partial responses, 20 stable responses, and 1 radiographic<br />

progression. Conclusions: In summary, combination <strong>of</strong> BEV, daily TMZ, and<br />

vorinostat has promising efficacy on recurrent GBM with reasonable<br />

toxicity/safety.<br />

2029^ Poster Discussion Session (Board #17), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Bevacizumab versus bevacizumab plus vorinostat in adults with recurrent<br />

malignant glioma: Results <strong>of</strong> a phase I part <strong>of</strong> a phase I/II trial. Presenting<br />

Author: Jing Wu, University <strong>of</strong> North Carolina at Chapel Hill, Lineberger<br />

Comprehensive Cancer Center, Department <strong>of</strong> Neurosurgery, Chapel Hill,<br />

NC<br />

Background: Antiangiogenic therapy using bevacizumab (Bev) has shown<br />

promising activity against recurrent glioblastoma (GBM). However, most<br />

patients have disease progression after striking but transient responses.<br />

Salvage therapies have been uniformly ineffective, suggesting development<br />

<strong>of</strong> resistance mechanisms including upregulation <strong>of</strong> other proangiogenic<br />

factors and increased activity <strong>of</strong> hypoxia inducible factors (HIF)-1a.<br />

Vorinostat, a histone deacetylase (HDAC) inhibitor has single agent activity<br />

against recurrent GBM and downregulates HIF-1a and other proangiogenic<br />

and invasive factors. We hypothesized that HDAC inhibition combined with<br />

Bev would result in improved clinical outcome. We report the results <strong>of</strong> the<br />

Phase I portion <strong>of</strong> the study preceding the initiation <strong>of</strong> the 2-arm adaptive<br />

randomized Phase II study. Methods: Adults with recurrent malignant<br />

glioma, KPS � 60, normal hepatic, renal and marrow organ function and no<br />

prior exposure to Bev or Vorinostat were enrolled to the combination therapy<br />

after the confirmation <strong>of</strong> recurrent GBM. A conventional 3�3 Phase I<br />

design was used to determine the maximum tolerated dose (MTD) and the<br />

toxicity pr<strong>of</strong>ile <strong>of</strong> the combination <strong>of</strong> Bev and Vorinostat. The starting dose<br />

was Bev at 10mg/kg administered on days 1 and 15 intravenously and<br />

Vorinostat 400 mg/day orally on days 1 to 7, and days 15 to 21 with each<br />

cycle being 28 days. Results: A total <strong>of</strong> 6 patients were enrolled and all 6<br />

patients were evaluable. Three patients were enrolled in the first cohort at<br />

the starting dose <strong>of</strong> the combination and completed the first cycle. One<br />

patient experienced a grade 3 ALT elevation and grade 3 hyperglycemia,<br />

which were designated as possibly related to vorinostat and constituting a<br />

dose-limiting toxicity (DLT). No grade 4 toxicities were noted. The cohort<br />

was expanded by 3 more patients with none <strong>of</strong> these patients experienced a<br />

DLT in the first cycle. The starting dose level <strong>of</strong> Bev and vorinostat was<br />

declared the Phase II dose. Conclusions: Combination <strong>of</strong> Bev (10 mg/kg q 2<br />

weeks) and <strong>of</strong> vorinostat (400 mg on days 1 to 7 and 15 to 21) has tolerable<br />

toxicity pr<strong>of</strong>ile. This will be followed by a multicenter Bayesian adaptive<br />

randomized Phase II study.<br />

2028 Poster Discussion Session (Board #16), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Bevacizumab for recurrent WHO grade III anaplastic glioma (AG). Presenting<br />

Author: Anna Maria Delios, Memorial Sloan-Kettering Cancer Center,<br />

New York, NY<br />

Background: Salvage treatment with bevacizumab (BEV) has been increasingly<br />

used in grade III AG, but limited data are available, with conflicting<br />

results reported. Because <strong>of</strong> differences in molecular characteristics,<br />

angiogenesis mechanisms, growth rate and chemosensitivity, results observed<br />

in glioblastoma (GBM) treated with BEV may not apply to AG, and<br />

may differ between anaplastic astrocytomas (AA) and oligodendrogliomas<br />

(AO). Methods: IRB approved retrospective review <strong>of</strong> all pts with recurrent<br />

WHO grade III AG treated with BEV at MSKCC. Response and progression<br />

were determined by RANO criteria. Results: BEV was given to 39 pts with<br />

recurrent grade III AG (AA: 26; AO: 10; anaplastic oligoastrocytoma [AOA]:<br />

3); median (med) age: 49 (range 20-75); med KPS: 80 (60-100). MGMT<br />

promoter methylation was determined in 17 pts (methylated 8, unmethylated<br />

9). Amongst AO/AOA, 1p/19q co-deletion was present in 6 and absent<br />

in 5. IDH1/ IDH2 mutations were present in 3/5 tested tumors. Med time<br />

from diagnosis <strong>of</strong> AG to BEV treatment was 11.5m (3 – 112.5). The med<br />

number <strong>of</strong> previous recurrences was 1 (range 1-4). BEV was given as single<br />

agent to 16 pts and combined with a cytotoxic agent in 23. Objective<br />

response rate (ORR) was 41% (complete response: 5; partial: 11). The med<br />

progression-free survival (PFS) was 4.8m (6-m PFS: 35% [95% CI<br />

20-50]). The med overall survival (OS) was 11.5 m (1y-OS: 45% [95% CI<br />

29-61]). In pts achieving ORR, med OS was 13 m vs 4minptswith<br />

stable/progressive disease (P�0.02). Pts at first recurrence fared better<br />

than pts with more than one recurrence (med PFS: 6 vs 3.4 months,<br />

P�0.04). AO/AOA tended to fare worse than AA (med PFS 3 vs 5.5 m,<br />

P�0.07). There were no differences in PFS (P�0.8) or OS (P�0.4)<br />

between single agent vs BEV � cytotoxic agent. Toxicity included one grade<br />

4 brain hemorrhage. Conclusions: In this relatively large series, BEV was<br />

associated with higher ORR and PFS as compared to historical controls,<br />

although improvements in those endpoints were <strong>of</strong> a lower magnitude than<br />

in GBM. While ORR predicted OS, improvements in OS were not apparent;<br />

results in AO were particularly disappointing. The use <strong>of</strong> BEV in this<br />

population requires reappraisal in a randomized study with adequate<br />

stratification for histology and number <strong>of</strong> previous recurrences.<br />

2030 Poster Discussion Session (Board #18), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Differences in outcome due to bevacizumab (BEV) discontinuation versus<br />

BEV failure in adults with glioblastoma. Presenting Author: Mark Daniel<br />

Anderson, University <strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: BEV is approved for use in recurrent glioblastoma. Patients<br />

(pts) who benefit from BEV therapy are <strong>of</strong>ten treated until tumor progression<br />

but fail to respond to salvage therapy suggesting that BEV may alter<br />

tumor biology. In a subset <strong>of</strong> pts who benefit from BEV, treatment is<br />

discontinued for reasons other than disease progression; the characteristics<br />

and outcomes <strong>of</strong> this subset are poorly defined. Methods: In this IRB<br />

approved retrospective study, our neuro-oncology longitudinal database<br />

was screened for pts treated with BEV for � 6 months (mo) between<br />

2005-2010 and 18 pts were identified in whom BEV was discontinued for<br />

reasons other than disease progression (BEV-D group). A cohort <strong>of</strong> 72 pts<br />

who received BEV until treatment failure due to progression was used as<br />

comparator (BEV-F group). Results: In the BEV-D group, 5 pts completed a<br />

planned treatment course and 13 stopped BEV due to toxicity; in this<br />

group, progression free survival at 12 mo (PFS12) was 83.3% (95% CI,<br />

56.8-94.3) and median time to progression (TTP) 27.6 mo. Median TTP<br />

after BEV discontinuation was 7.0 mo. In contrast, in the BEV-F group,<br />

PFS12 was 24.6% (95% CI, 13.9-36.2) and median PFS 9.7 mo. Length<br />

<strong>of</strong> BEV therapy was not significantly different between the groups with a<br />

median time to discontinuation <strong>of</strong> 10.2 and 12 mo. In 12/18 pts in the<br />

BEV-D group who subsequently had tumor recurrence a predominantly<br />

local pattern <strong>of</strong> progression was seen unlike those in the BEV-F group who<br />

had more infiltrative or distant failures. Salvage therapy yielded a PFS-6 <strong>of</strong><br />

28.6% (95% CI, 4.1-61.2) with a median PFS <strong>of</strong> 17.1 wk compared with<br />

6.8% (95% CI, 1.2-19.8) and 9 wk in the comparator group. Conclusions:<br />

Among pts who benefit from BEV therapy, the BEV-D group did not<br />

experience an immediate progression suggesting continued benefit after<br />

BEV cessation. This cohort also had a less invasive pattern <strong>of</strong> recurrence<br />

and a possibly improved response to salvage therapy compared with BEV-F<br />

group. Our results suggest that planned cessation <strong>of</strong> BEV therapy could<br />

potentially change patterns <strong>of</strong> progression and response to subsequent<br />

therapy. This strategy warrants further evaluation in prospective studies<br />

given the absence <strong>of</strong> effective salvage therapy after BEV failure.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


2031 Poster Discussion Session (Board #19), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

NCCTG N057K phase II trial <strong>of</strong> everolimus, temozolomide, and radiotherapy<br />

in patients with newly diagnosed glioblastoma: A North Central<br />

Cancer Treatment Group trial. Presenting Author: Daniel Ma, Mayo Clinic,<br />

Rochester, MN<br />

Background: The mammalian target <strong>of</strong> rapamycin (mTOR) functions within<br />

the PI3K/Akt pathway as a critical modulator <strong>of</strong> cell survival. Preclinical<br />

studies in GBM indicate that the combination <strong>of</strong> mTOR inhibitors, such as<br />

everolimus (RAD001), with either radiation therapy (RT) or temozolomide<br />

(TMZ) provide increased tumor cell killing. Methods: Newly diagnosed GBM<br />

pts were eligible for the study. RAD001 was dosed orally at 70 mg/week<br />

weekly, starting 1 week prior to RT/TMZ, and continued throughout<br />

RT/TMZ, adjuvant TMZ and then until progression. This was a single arm<br />

phase II design powered to detect a true overall survival at 12 months<br />

(OS12) <strong>of</strong> 73% (vs 58% in historical controls). Secondary endpoints were<br />

toxicity, response rate, and time to progression (TTP). A subgroup <strong>of</strong><br />

patients with measurable residual disease were eligible for the PET imaging<br />

component <strong>of</strong> the study, consisting <strong>of</strong> an 18FLT-PET/CT scan performed<br />

before and after the initial two doses <strong>of</strong> RAD001 but before the first dose <strong>of</strong><br />

RT or TMZ. Results: 103 patients were accrued to phase II <strong>of</strong> which 100<br />

were evaluable. The median age was 60.5 years (23-81), median ECOG PS<br />

was 1, 46 patients had GTR, 33 STR, and the remainder had biopsy at<br />

diagnosis. Treatment tolerance was acceptable: 17% patient had at least<br />

one grade 3 hematologic toxicity; 14% had at least one grade 4 hematologic<br />

toxicity, 42% had at least one grade 3 non-heme toxicity, while 12%<br />

had at least one grade 4 non-heme toxicity. No increased incidence <strong>of</strong><br />

infectious complications was observed and there were no treatment related<br />

deaths. Median PFS was 5.3 months (1.3-21.4), with 22 patients<br />

progression free. Mature OS data will be available at the meeting. MGMT<br />

methylation status analysis is ongoing. Of the pts who had evaluable<br />

FLT-PET data, three <strong>of</strong> eight (37.5%) had a drop in SUVmax �25% after<br />

two treatments <strong>of</strong> RAD001. Conclusions: RAD001 with standard <strong>of</strong> care<br />

chemoradiation had moderate toxicity. Serial 18FLT-PET was feasible for<br />

evaluating drug-induced changes in tumor proliferation following RAD001.<br />

Final outcome data and association <strong>of</strong> MGMT status with outcome will be<br />

reported at the meeting.<br />

2033 Poster Discussion Session (Board #21), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Final results <strong>of</strong> a randomized phase III trial <strong>of</strong> nimotuzumab for the<br />

treatment <strong>of</strong> newly diagnosed glioblastoma in addition to standard radiation<br />

and chemotherapy with temozolomide versus standard radiation and<br />

temoziolamide. Presenting Author: Manfred Westphal, Department <strong>of</strong><br />

Neurosurgery, UKE Hamburg, Hamburg, Germany<br />

Background: The receptor for epidermal growth factor (EGF-R) has consistently<br />

been found expressed or overexpressed in human malignant glioma<br />

disease. Therapeutic targeting <strong>of</strong> the EGF-R has shown mixed responses<br />

which appear to be restricted by specific features <strong>of</strong> the tumor cells.Nimotuzumab<br />

binds preferentially to highly over expressing cells, a phase III trial<br />

was initiated to test efficacy in glioma. Methods: Nimotuzumab was tested<br />

in an open label, randomized, multicenter Phase III trial in patients with<br />

histologically confirmed, newly diagnosed glioblastoma. 12 consecutive<br />

weekly infusions <strong>of</strong> 400mg during standard radiotherapy with Temozolomide<br />

followed by biweekly infusions <strong>of</strong> 400mg arm A was randomized<br />

against standard radio chemotherapy alone arm B. Primary endpoint was<br />

PFS as determined by centralized neuro-imaging review. OS as secondary<br />

endpoint with quality <strong>of</strong> life, safety as additional parameters.MGMT and<br />

EGF-R were assessed where sufficient materials could be retrieved as well<br />

as tumor hypoxia. Results: Between 2007 and 2010, 149 patients were<br />

randomized and 142 were available for analysis. Stratification according to<br />

resection status resulted in 40 pts in the treatment arm and 41 in the<br />

control arm with residual tumor and 31 vs 30 pts without residual tumor.<br />

PFS: 12 month was 25.5% arm A vs 20.3% in arm B (p � 0.78) and OS:<br />

679 days vs 596 days. For non - methylated MGMT the difference was most<br />

notable: OS arm A 28 pats: 19.6 months vs arm B 28 pats 15.0 months<br />

EGF-R amplification, gave no clear signal, neither was there a statistically<br />

significant treatment effect between with or without residual tumor. Tumor<br />

hypoxia does not appear to have predictive value in the overall group but<br />

possibly in subgroups.AEs and SAEs did not reveal a new specific toxicity<br />

pr<strong>of</strong>ile beyond the known side effects from glioma treatment with current<br />

standard. Conclusions: Nimotuzumab shows a clear trend towards efficacy<br />

in MGMT non-methylated glioblastoma patients. Safety pr<strong>of</strong>ile and indications<br />

for subgroup efficacy potentially justify focused evaluation <strong>of</strong> antibody<br />

therapy against glioblastoma.<br />

Central Nervous System Tumors<br />

123s<br />

2032 Poster Discussion Session (Board #20), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Phase I/randomized phase II trial <strong>of</strong> either dasatinib or placebo combined<br />

with standard chemoradiotherapy for newly diagnosed glioblastoma multiforme<br />

(GBM): Final results <strong>of</strong> phase I study. Presenting Author: Nadia N.<br />

Laack, Mayo Clinic, Rochester, MN<br />

Background: Dasatinib is a potent oral ATP competitive multi-targeted<br />

kinase inhibitor <strong>of</strong> multiple members <strong>of</strong> the Src kinase family, known to be<br />

involved in gliomagenesis and invasion. N0877 is a phase I/randomized<br />

phase II trial evaluating the combination <strong>of</strong> dasatinib, radiation (RT) and<br />

temozolomide (TMZ) in newly diagnosed GBM. The phase I portion has<br />

been completed and is the focus <strong>of</strong> this report. Methods: A cohorts-<strong>of</strong>-3<br />

design was used to assess the safety <strong>of</strong> dasatinib in combination with RT<br />

and concomitant TMZ, and establish the phase II dose <strong>of</strong> the combination.<br />

Dasatinib was given orally for 42 days, beginning with the first day <strong>of</strong> RT<br />

(total dose 60 Gy) and first dose <strong>of</strong> TMZ (75 mg/m2/d). A 24 - 42 day rest<br />

(cycle 2) followed the RT/TMZ/dasatinib. Patients (pts) were observed for<br />

DLT to the end <strong>of</strong> cycle 2. Patients then received 6 cycles (28 day cycles) <strong>of</strong><br />

dasatinib (once daily) and TMZ (days 1-5). At the completion <strong>of</strong> 6 cycles <strong>of</strong><br />

TMZ � dasatinib, pts stay on dasatinib only (28 day cycles) until<br />

progressive disease. Results: Phase I component is complete with 13<br />

patients (3 at dose level 0, 3 at dose level 0-A, 7 at dose level 1). One<br />

patient in dose level 1 had to be replaced due to the development <strong>of</strong><br />

unrelated medical issues. One DLT (grade 4 pancytopenia) was observed in<br />

1 patient at dose level 0 (50mg bid) and one DLT (grade 3 rash) was<br />

observed in 1 patient at dose level 1 (150mg qd). MTD <strong>of</strong> dasatinib was<br />

determined to be 150mg daily. Most common adverse events throughout<br />

the entire study period were hematologic with the most common toxicity<br />

being lymphopenia (grade 3 in 69% <strong>of</strong> patients, grade 4 in 8%). Other<br />

toxicities attributed to treatment and occurring in � 10% <strong>of</strong> patients<br />

included anemia (31%), neutropenia (15%), and fatigue(15%). Best<br />

response was stable disease in 10 patients, progressive disease in 1<br />

patient, and not evaluable in 2. Conclusions: MTD for dasatinib in<br />

combination with TMZ and RT in newly diagnosed GBM patients is 150mg<br />

daily. Toxicity was primarily hematologic with minimal non-hematologic<br />

events. This dose is currently being used in the ongoing placebo-controlled,<br />

randomized phase II trial.<br />

2034 Poster Discussion Session (Board #22), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

APG101_CD_002: A phase II, randomized, open-label, multicenter study<br />

<strong>of</strong> weekly APG101 plus reirradiation versus reirradiation in the treatment <strong>of</strong><br />

patients with recurrent glioblastoma. Presenting Author: Martin Bendszus,<br />

Department <strong>of</strong> Neuroradiology, University Hospital Heidelberg, Heidelberg,<br />

Germany<br />

Background: Preclinical data indicate antiinvasive activity <strong>of</strong> APG101, an<br />

intravenous CD95 ligand-binding fusion protein, as well as synergistic<br />

activity together with radiotherapy in glioblastoma. Methods: Patients with<br />

recurrent glioblastoma after prior standard radiochemotherapy with temozolomide<br />

(� 1 second-line chemotherapy) were considered for re-irradiation<br />

provided a tumor diameter 1-4 cm and time since the end radiotherapy � 8<br />

months. Patients were randomized 1:2 between radiotherapy (36 Gy; 5<br />

times 2 Gy per week) or radiotherapy plus APG101 at 400 mg weekly flat<br />

dose to be continued until progression. Radiotherapy plans were centrally<br />

evaluated. Primary endpoint was 6-months progression free survival (PFS-<br />

6). MRIs were performed every 6-weeks and centrally read. Sample size <strong>of</strong><br />

the investigational treatment arm according to a two-stage design <strong>of</strong> Simon<br />

required 55 patients. A control arm <strong>of</strong> 28 patients was implemented to<br />

validate the assumptions on PFS-6 in a cohort <strong>of</strong> patients treated with<br />

reirradiation alone. Results: Between 12/09 and 09/11, 84 pts in 25<br />

centers were randomized and treated, preliminary data <strong>of</strong> the current report<br />

are available on 71 patients (49 APG01 � irradiation, 22 irradiation<br />

alone). Median age was 57 years, median KPS 90%. The maximal tumor<br />

diameter was � 2-5 cm in 34 patients and � 2.5 cm in 37 patients. No<br />

SUSARs have to be reported. Nine patients achieved PFS-6 in the APG101<br />

arm and none in the radiotherapy arm. Conclusions: APG101_CD_002 is<br />

the first trial evaluating CD95-mediated pathway inhibition as a therapeutic<br />

strategy. This trial is also the prospective trial on reirradiation in<br />

glioblastoma. Side effects <strong>of</strong> the combination were minimal, and treatment<br />

could be delivered as planned. The experimental arm met the primary<br />

endpoint. The approach to block rather than stimulate the CD95 system is<br />

breaking a paradigm. Our data suggest that CD95 inhibition by APG101<br />

should be evaluated in the management <strong>of</strong> newly diagnosed glioblastoma in<br />

combination with standard radiochemotherapy. Further molecular data and<br />

updated results will be presented.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


124s Central Nervous System Tumors<br />

2035 Poster Discussion Session (Board #23), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

A phase II trial evaluating tumor penetration <strong>of</strong> bortezomib in patients with<br />

recurrent malignant gliomas treated prior to surgery and then treated with<br />

bortezomib and temozolomide postoperatively. Presenting Author: Jeffrey<br />

J. Raizer, Northwestern University, Feinberg School <strong>of</strong> Medicine, Chicago,<br />

IL<br />

Background: NF-Kappa B is one <strong>of</strong> the mechanisms <strong>of</strong> resistance for<br />

malignant gliomas. A few trials have assessed bortezomib in the treatment<br />

<strong>of</strong> malignant gliomas with limited activity. This may in part be due to<br />

limitations in dose escalation from peripheral neuropathy. We performed a<br />

phase II trial with the goal <strong>of</strong> measuring bortezomib in tumor tissue and also<br />

its effects on NF-Kappa B. Methods: Patients felt to be surgical candidates<br />

were enrolled after signing an IRB approved consent. They were treated<br />

with bortezomib 1.7 mg/m2 IV on day 1, 4 and 8 and then had surgery on<br />

day 8 or 9. Approximately, 14 days post operatively, patients were started<br />

on temozolomide 75 mg/m2 PO on days 1-7 and 14-21; on day 7 and day<br />

21 they received bortezomib 1.7 mg/m2 (1 cycle was 1 month). Treatment<br />

continued until progression. If �1 patient had a PFS at 6 months the trial<br />

would be stopped. Results: 10 patients were enrolled (8 M and 2 F). Median<br />

age and KPS were 50 (42-64) and 90% (70-90). All but 1 patient went to<br />

surgery, one had an intracranial hemorrhage. The median number <strong>of</strong> cycles<br />

post operatively was 2 (1-4), with two patients discontinuing for wound<br />

infection (post 3 cycles, not restarted due to prolonged delays) and<br />

meningitis (post 2 cycles then withdrew from trial). Six patients are<br />

deceased. Trial was stopped as no patient had a PFS-6. PK analysis<br />

revealed measurable drug levels in tumor tissue. Conclusions: Post operative<br />

treatment with temozolomide and Bortezomib did not have any activity.<br />

Bortezomib can achieve measurable drug levels in tumor but may not be<br />

sufficient for anti-tumor activity. Tissue will be assessed for bortezomib<br />

effects on NF-Kappa B.<br />

2037 Poster Discussion Session (Board #25), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Phase II trial <strong>of</strong> dose-dense temozolomide as initial treatment for progressive<br />

low-grade oligodendroglial tumors: A multicentric study <strong>of</strong> the Italian<br />

Association <strong>of</strong> Neuro-Oncology (AINO). Presenting Author: Roberta Ruda,<br />

Division <strong>of</strong> Neuro-Oncology, University Hospital San Giovanni Battista,<br />

Turin, Italy<br />

Background: Standard temozolomide has been shown to be active in<br />

progressive low grade gliomas after surgery, whereas few data are available<br />

on the impact <strong>of</strong> dose dense regimens. Thus, we developed a phase II single<br />

arm multicenter study to evaluate the efficacy and toxicity <strong>of</strong> a regimen <strong>of</strong><br />

dose dense temozolomide in progressive low grade oligodendroglial tumors.<br />

Methods: The inclusion criteria <strong>of</strong> the study were as follows: 1)biopsyproven<br />

supratentorial WHO grade II oligodendroglioma and oligoastrocytoma;<br />

2)progressive disease, clinically (epileptic seizures)or radiologically;<br />

3)measurable disease on MRI (at least 1 cm); 4)age �18 years; 5)Karn<strong>of</strong>sky<br />

Performance Status �70. Temozolomide was administered at<br />

150mg/m2 1 week on/1 week <strong>of</strong>f up to a maximum <strong>of</strong> 18 cycles or<br />

unacceptable toxicity. The primary end-point was response rate (RR)<br />

according to RANO criteria, whereas secondary end-points were clinical<br />

benefit in terms <strong>of</strong> reduction <strong>of</strong> epileptic seizures �50%, progression-free<br />

survival (PFS), quality <strong>of</strong> life and toxicity. Results: From January 2005 until<br />

December 2010 60 patients (median age 39 and median KPS 80) have<br />

been accrued and are now evaluable for response. Response rates on<br />

T2/FLAIR images were as follows: CR 0/60, PR 21/60 (35%), MR 14/60<br />

(23%), SD 21/60 (35%) and PD 4/60 (7%). The clinical benefit was<br />

significant in 29/34 patients (85%). As for toxicity 5/60 (8%) patients<br />

stopped treatment for lymphopenia grade IV, whereas 11/31 patients<br />

(35%) were switched to the standard regimen <strong>of</strong> temozolomide. PET with<br />

methionine was added to MRI in 17 patients: in 10/17 (59%) a disappearance<br />

or a significant reduction <strong>of</strong> uptake was observed, being the reduction<br />

<strong>of</strong> seizures better correlated with the response on PET rather than that on<br />

MRI. 1p/19q codeletion was not associated with either the response or the<br />

clinical benefit, whereas the analysis <strong>of</strong> MGMT methylation and IDH1<br />

mutations are ongoing. Thirty-nine (65%) patients are still free <strong>of</strong> tumor<br />

progression. Conclusions: Dose-dense TMZ seems to be active, especially in<br />

terms <strong>of</strong> clinical benefit, but the myelotoxicity could be a concern.<br />

2036 Poster Discussion Session (Board #24), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Lenalidomide and irinotecan in adults with recurrent malignant gliomas:<br />

Phase I results <strong>of</strong> the phase I/II trial. Presenting Author: Vinay K. Puduvalli,<br />

University <strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: Patients with recurrent malignant gliomas have limited treatment<br />

options. We previously reported promising results using 6 month<br />

progression free survival [PFS6] endpoint combining irinotecan and thalidomide<br />

compared to historical controls. In this study, we tested the<br />

tolerability and efficacy <strong>of</strong> Irinotecan combined with Lenalidomide, a more<br />

potent thalidomide analogue with unique antiangiogenic and immunomodulatory<br />

properties. Methods: This phase I portion <strong>of</strong> the phase I/II study<br />

aimed to determine the maximum tolerated doses (MTD) <strong>of</strong> irinotecan and<br />

lenalidomide. Eligible patients were adults (��18 y) with recurrent WHO<br />

Grade 3 or 4 gliomas who were not on EIAED, had failed prior radiation<br />

therapy, had a KPS��60, had adequate marrow, renal and hepatic organ<br />

function, no major medical illnesses and no other concurrent malignancies.<br />

Each cycle was designated as 4 weeks in duration. Results: Two <strong>of</strong> the 4<br />

patients enrolled at the starting dose level <strong>of</strong> Lenalidomide 10 mg/day on<br />

days 1-21 and irinotecan 200 mg/m2 q2 weeks developed rash as dose<br />

limiting toxicity (DLT). The trial was restarted with no change in irinotecan<br />

dose but with a lowered Lenalidomide dose <strong>of</strong> 7.5 mg/day for cycle 1 with<br />

escalation to 10 mg/day for cycle 2 and beyond. Of 3 eligible patients<br />

enrolled, no DLTs were noted even after cycle 2. The dose was hence<br />

escalated to Lenalidomide 10 mg/day with unchanged irinotecan dose.<br />

Only 1/6 patients experienced a DLT (pulmonary embolism); however, it<br />

was noted that 4/6 patients required dose reduction <strong>of</strong> irinotecan to 150<br />

mg/m2 after cycle 1. One patient died during cycle 2 <strong>of</strong> unknown causes<br />

(autopsy declined) without reports <strong>of</strong> preceding toxicities. Non DLT<br />

toxicities included neutropenia, leukopenia, hypokalemia, diarrhea, fatigue<br />

and nausea/vomiting. Lenalidomide pharmacokinetic data, obtained by<br />

serial blood draws initially after one dose <strong>of</strong> the drug on day 0 and<br />

subsequently after irinotecan and lenalidomide dose on day 1 to examine<br />

drug interactions, will be presented. Conclusions: Based on these results,<br />

the maximum tolerated dose was Lenalidomide 10 mg/day on days 1-21<br />

and irinotecan 150 mg/m2 q 2 weeks every 28 days which will be used in<br />

the phase II study that will open shortly.<br />

2038 General Poster Session (Board #12A), Sat, 1:15 PM-5:15 PM<br />

Does pilocytic astrocytoma (PA) in adults have a different prognosis than<br />

PA in children? Presenting Author: Daniel Fernandes Marques, Instituto do<br />

Cancer do Estado de São Paulo, Universidade de São Paulo, São Paulo,<br />

Brazil<br />

Background: Pilocytic astrocytoma (PA) accounts for up to 25% <strong>of</strong> all brain<br />

tumors in children. A 75% progression-free survival rate at 5 years and<br />

overall survival rates <strong>of</strong> 95.8% at 10 years have been reported after a<br />

complete resection. However, the incidence in adults is low and the<br />

outcome less well characterized. We conducted a retrospective analysis <strong>of</strong><br />

adult PAs identified in order to review the clinical characteristics and<br />

outcome <strong>of</strong> this neoplasm in this age group. Methods: A descriptive,<br />

cross-sectional study was undertaken in patients over 16 y.o., diagnosed<br />

with PA between January 1990 and December 2010 at our center. <strong>Clinical</strong><br />

characteristics, date and extent <strong>of</strong> surgery, tumor location, postoperative<br />

treatment, complications and recurrences were collected from hospital<br />

records. Results: From May, 1990 to December, 2010, 18 eligible adult<br />

patients (pts) with the diagnosis <strong>of</strong> PA were identified. Median age was<br />

25,5 (16-52) years, 72,2% female and 83,3% had an ECOG performance<br />

status � 2. The most frequent tumor sites were the cerebellum (44,4%),<br />

followed by the cerebral hemispheres (38%). All patients underwent<br />

surgery as primary treatment, 72% had a complete macroscopic resection<br />

(CMR) and 28% had a partial resection (PR). Two patients received<br />

postoperative radiotherapy: one following a PR and the other after tumor<br />

relapse following a CMR. Two patients relapsed five months and 44 months<br />

after initial complete gross resection. Salvage surgery achieved CMR in<br />

both. With a median follow-up <strong>of</strong> 89 months, all patients are alive, except<br />

for one who died in the postoperative period due to fungal meningitis. Of<br />

note, none <strong>of</strong> 5 pts undergoing a partial resection progressed and 3 are alive<br />

for more than 10 years. Conclusions: As it occurs in the pediatric<br />

population, PA in adults seems to carry a similarly favorable prognosis. It is<br />

conceivable that after initial surgical resection a watch and wait type <strong>of</strong><br />

approach is appropriate, even following partial resection. The role <strong>of</strong><br />

upfront radiotherapy is uncertain and it should probably be left for<br />

progressive tumors.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


2039 General Poster Session (Board #12B), Sat, 1:15 PM-5:15 PM<br />

Use <strong>of</strong> a non-coplanar half-beam block on the lower spinal field to decrease<br />

the maximum bowel and cumulative dose in craniospinal irradiation.<br />

Presenting Author: Madeera Kathpal, University <strong>of</strong> Medicine and Dentistry<br />

<strong>of</strong> New Jersey, New Brunswick, NJ<br />

Background: To develop and compare a non-coplanar half beam block<br />

technique that can be used in both prone and supine treatment positions<br />

with conventional beam matching for craniospinal irradiation (CSI) in order<br />

to decrease the maximum cumulative dose and dose to the bowel, while<br />

maintaining the therapeutic dose to the spinal axis. Methods: Ten treatment<br />

plans from five patients who underwent CSI were analyzed. The bowel was<br />

contoured en bloc for each patient on their simulation cat scan. Two<br />

different geometric techniques for each patient were planned and analyzed.<br />

The first technique consisted <strong>of</strong> the conventional method for CSI<br />

utilizing two coplanar beams to cover the entire spinal axis. The other<br />

technique used a non-coplanar half beam block on the lower spinal beam to<br />

exactly match the upper spinal beam’s divergence. Four “featherings”<br />

between the two spinal beams for each technique were still necessary to<br />

minimize under and overdosing which occur at abutting beam fields.<br />

Maximum doses for the plan and the bowel were compared between the two<br />

techniques on the same patient. Results: The maximum bowel dose was<br />

decreased between 10 to 35 percent when the non-coplanar half beam<br />

block was used. The maximum doses for the conventional technique were 5<br />

to 35 percent higher than the plans using a non-coplanar half beam block.<br />

The homogeneity <strong>of</strong> the dose to the spinal axis was not altered with the use<br />

<strong>of</strong> the non-coplanar half beam block. Conclusions: Use <strong>of</strong> a non-coplanar<br />

half beam block to match the two spinal fields in craniospinal irradiation<br />

significantly reduces the maximum dose to the bowel and <strong>of</strong> the entire plan<br />

possibly resulting in reduced gastrointestinal toxicity while maintaining<br />

therapeutic dose to the spinal axis.<br />

2041 General Poster Session (Board #12D), Sat, 1:15 PM-5:15 PM<br />

Toxicity pr<strong>of</strong>ile <strong>of</strong> temozolomide in the treatment <strong>of</strong> 300 malignant glioma<br />

patients in Korea. Presenting Author: Chae-yong Kim, Department <strong>of</strong><br />

Neurosurgery, Seoul National University College <strong>of</strong> Medicine, Seoul National<br />

University Bundang Hospital, Seoul, South Korea<br />

Background: Toxicity <strong>of</strong> temozolomide itself has been rarely reported in the<br />

field <strong>of</strong> neuro-oncology. In an attempt to explore the toxicity pr<strong>of</strong>iles <strong>of</strong><br />

temozolomide we investigated the records <strong>of</strong> 300 glioma patients in two<br />

institutions in Korea. Methods: We reviewed the records <strong>of</strong> 300 glioma<br />

patients who were treated with temozolomide between Jan 2004 and May<br />

2010 at two hospitals <strong>of</strong> our university at two medical centers in Seoul<br />

National University, Korea. The age range <strong>of</strong> patients was 17 ~ 84 years old<br />

with its median <strong>of</strong> 49 years old. Their pathologies were glioblastoma,<br />

anaplastic astrocytoma, anaplastic oligodendroglioma, anaplastic oligoastrocytoma,<br />

gliomastosis cerebri, gliosarcoma and others. Temozolomide<br />

was used as only concurrent manner with radiotherapy in 46 patients, as<br />

both concurrent and adjuvant manner in 93 patients, and as only adjuvant<br />

or palliative manner in 161 patients. We classified the side effects by<br />

Common Terminology Criteria for Adverse Events version 3.0(CTCAE).<br />

Results: We found 603 events <strong>of</strong> side effects <strong>of</strong> temozolomide. Nonhematolgic<br />

toxicities were most common (n�482). Especially, nausea(n�135),<br />

vomiting(n�110), and anorexia(n�40) were common among all kinds <strong>of</strong><br />

toxicities. Hematologic adverse events(n�121), for example, thrombocytopenia(n�43),<br />

anemia(n�33), and neutropenia(n�20) were the second<br />

common toxicities. Some patients suffered from fatigue(n�31). None <strong>of</strong><br />

our patient presented fatal Pneumocystis jiroveci pneumonitis. In our<br />

series, 512 cases(84.9%) <strong>of</strong> toxicity were grade 1 or 2, and 91<br />

cases(15.1%) were grade 3 or 4. Grade 3 or 4 toxicity were 140<br />

cases(15.1%). Only 27 cases(2.9%) presented grade 4 toxicity. There was<br />

no mortality due to temozolomide. Only 4 patients (1.3%) presented<br />

leukopenia, which is different pattern compared with the incidence<br />

reported in prior studies from western countries. Conclusions: Temozolomide<br />

had many kinds <strong>of</strong> toxicities. However, most <strong>of</strong> the toxicity was<br />

tolerable. This pr<strong>of</strong>ile could be used to prevent toxic reactions more<br />

effectively and improve the patient’s quality <strong>of</strong> life. Furthermore, we could<br />

use these lessons for education <strong>of</strong> patients during the treatment with<br />

temozolomide.<br />

Central Nervous System Tumors<br />

125s<br />

2040 General Poster Session (Board #12C), Sat, 1:15 PM-5:15 PM<br />

Relationship <strong>of</strong> cognitive function, quality <strong>of</strong> life (QOL), and neuroimaging<br />

in primary CNS lymphoma (PCNSL) survivors. Presenting Author: Nancy<br />

Diane Doolittle, Oregon Health & Science University, Portland, OR<br />

Background: Delayed treatment-related neurotoxicity in PCNSL is a significant<br />

problem since improved treatments have increased survival. The study<br />

purpose is to describe and correlate neuropsychologic (NP), QOL and<br />

neuroimaging outcomes as neurotoxicity indicators. Methods: Four centers<br />

in Germany and U.S. prospectively evaluated PCNSL patients (pts) in<br />

complete remission (CR) for 2 yrs or more, treated as shown in the Table.<br />

NP tests evaluated attention/executive function, verbal memory, motor<br />

skills, and QOL (Correa, Ann Oncol 2007). Brain MRI was obtained; the<br />

size <strong>of</strong> T2 abnormalities was determined using two perpendicular linear<br />

measurements where hyperintensities were largest and the sum <strong>of</strong> the<br />

measurements was calculated. Differences in total T2 among treatments<br />

were compared using analysis <strong>of</strong> variance; correlations between total T2<br />

and NP or QOL results were assessed using Pearson’s correlation coefficient<br />

(r). Results: From Feb 2009 to Feb 2011, 80 pts were evaluated (43<br />

male; median age, 59; median KPS, 80). Median follow-up from diagnosis<br />

to evaluation was 5.5 yrs. Total T2 abnormalities were significantly<br />

different among treatments (p � 0.0006). The mean area <strong>of</strong> total T2 in pts<br />

treated with WBRT was significantly higher and more than twice the mean<br />

<strong>of</strong> any <strong>of</strong> the other 3 treatments. Total T2 abnormalities were negatively<br />

associated with NP results ie. attention/executive function, r � -0.38 (p �<br />

0.0006), verbal memory, r � -0.23 (p � 0.042), motor skills, r � -0.28 (p<br />

� 0.016), composite score, r � -0.34 (p � 0.002); and functional/global<br />

QOL (higher total T2 associated with lower QOL). Conclusions: This large<br />

PCNSL series in long-term (LT) CR reveals higher total T2 abnormalities in<br />

pts treated with WBRT, which are associated with poorer cognitive<br />

performance and lower QOL at LT follow-up. Enhanced chemotherapy<br />

results in exciting LT survival and function.<br />

Treatment No. pts Follow-up (median, yrs)<br />

High-dose methotrexate (HD MTX)-based<br />

32 4.5<br />

chemotherapy (CHT) alone<br />

MTX-based intra-arterial CHT with blood-brain<br />

25 12.3<br />

barrier disruption alone<br />

HD MTX-based CHT f/b HDT/autologous stem cell<br />

8 4.5<br />

transplantation (ASCT) alone<br />

HD MTX-based CHT (with/without HDT/ASCT) and WBRT 15 5.6<br />

2042 General Poster Session (Board #12E), Sat, 1:15 PM-5:15 PM<br />

The oral transforming growth factor-beta (TGF-ß) receptor I kinase inhibitor<br />

LY2157299 plus lomustine in patients with treatment-refractory malignant<br />

glioma: The first human dose study. Presenting Author: Analia Azaro,<br />

Medical Oncology, Vall d’Hebron, Barcelona, Spain<br />

Background: Activated TGF-b signaling has been associated with poor<br />

survival in several tumors, including glioma. TGF-b inhibitors are expected<br />

to improve outcome. <strong>Part</strong> B <strong>of</strong> this Phase I trial assessed safety, pharmacokinetics,<br />

pharmacodynamics, and antitumor activity <strong>of</strong> the intermittent<br />

treatment with LY2157299 in combination with lomustine. Methods: After<br />

evaluating safety <strong>of</strong> LY2157299 monotherapy in part A <strong>of</strong> the study, 2<br />

cohorts <strong>of</strong> patients were treated with LY2157299 at 160 or 300 mg/day<br />

with intermittent dosing (each cycle�14 days on followed by 14 days <strong>of</strong>f)<br />

in combination with lomustine at standard dose (100 to 130 mg/m2 every 6<br />

weeks) for �2 cycles. Toxicity was assessed using the Common Terminology<br />

Criteria for Adverse Events, version 3. Results: Twenty-six patients with<br />

glioma (18 WHO Grade IV; 5 Grade III; 3 unspecified grade) were treated<br />

with LY2157299 and lomustine (160 mg/day, n�15; 300 mg/day, n�11).<br />

There were no dose-limiting toxicities. No clinically meaningful cardiotoxicities<br />

were observed. Twenty-one SAEs occurred in 10 patients, all<br />

considered to be related to lomustine and none to LY2157299. Of the 26<br />

patients, 7 had thrombocytopenia (27%), and recovery around weeks 4 to 6<br />

was not impacted by the second cycle <strong>of</strong> LY2157299. Two patients taking<br />

160 mg/day were treated for �6 cycles, with 1 <strong>of</strong> the 2 patients showing an<br />

unconfirmed partial response. At the 300 mg/day dose, no responses were<br />

observed; 2 patients were treated for �6 cycles. Co-administration <strong>of</strong><br />

lomustine did not alter LY2157299 exposure. Observed exposure <strong>of</strong><br />

LY2157299 increased with dose escalation between the 2 cohorts. On Day<br />

7, the variability estimates (coefficients <strong>of</strong> variation) <strong>of</strong> exposure and<br />

maximum concentration were slightly higher in presence <strong>of</strong> lomustine<br />

(58%) when compared with LY2157299 alone (47%) and then again<br />

reduced on Day 14 (53%). Pharmacodynamic results will be reported at a<br />

later date. Conclusions: The 14 days on/14 days <strong>of</strong>f treatment with<br />

LY2157299 did not increase the known lomustine toxicity. Given the<br />

overall safety pr<strong>of</strong>ile and antitumor effect, LY2157299 is being investigated<br />

in Phase II studies.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


126s Central Nervous System Tumors<br />

2043 General Poster Session (Board #12F), Sat, 1:15 PM-5:15 PM<br />

Expression <strong>of</strong> CXCR7, CXCR4, CXCR3 and their chemokine ligands in<br />

glioblastoma. Presenting Author: Robert D. Berahovich, ChemoCentryx,<br />

Inc., Mountain View, CA<br />

Background: The chemokine receptors CXCR4 and CXCR7, and their shared<br />

ligand CXCL12/SDF-1, have been implicated in multiple aspects <strong>of</strong><br />

tumorigenesis, including tumor cell proliferation, angiogenesis, vasculogenesis,<br />

and metastasis. Recently, a role for CXCR4 and CXCR7 has been<br />

postulated in the control <strong>of</strong> glioblastoma growth and vasculogenesis. We set<br />

out to define the expression patterns <strong>of</strong> these proteins on primary<br />

glioblastoma tumors and associated vasculature. Methods: First, a tumor<br />

microarray containing grades 1, 3, and 4 glioma samples were analyzed by<br />

immunohistochemical techniques for the expression <strong>of</strong> CXCR7. Second,<br />

two glioblastoma microarrays were analyzed by the same technique for<br />

CXCR7, CXCR4, and their shared chemokine ligand CXCL12. The same<br />

microarrays were also probed for the expression <strong>of</strong> the additional CXCR7<br />

ligand, CXCL11/ITAC, as well as the chemokine receptor CXCR3, which is<br />

also a receptor for CXCL11. Tissues were scored in a blinded fashion by a<br />

certified neuropathologist for both the intensity <strong>of</strong> signal and location <strong>of</strong><br />

signal for all proteins tested. Results: CXCR7 expression was largely<br />

dependent on tumor grade, as the receptor was usually absent on most<br />

grade 1 gliomas but was moderately-to-highly expressed in the majority <strong>of</strong><br />

anaplastic astrocytomas and glioblastomas. CXCR7 expression on tumorassociated<br />

vasculature was seen in all samples and its expression level also<br />

increased with glioma grade. In contrast to CXCR7, CXCR4 expression in<br />

glioblastoma was usually limited to the glioma cells, with only infrequent<br />

expression on the vasculature. Conversely, CXCL12 expression in glioblastoma<br />

was usually limited to the vasculature. CXCR3 was detected in a<br />

subset <strong>of</strong> glioblastomas, and only on the glioma cells. The CXCR7 and<br />

CXCR3 ligand CXCL11 was, like CXCR7, expressed on both the glioma cells<br />

and vasculature. Conclusions: These results, in light <strong>of</strong> prior evidence for<br />

CXCR7 in tumor growth, suggest a positive role for CXCR7 in the<br />

development <strong>of</strong> glioblastoma. Because CXCR7 colocalizes with its ligands,<br />

CXCL12 and CXCL11, as well as their other receptors, CXCR4 and CXCR3,<br />

the function <strong>of</strong> CXCR7 in glioblastoma may lie in the regulation <strong>of</strong> both the<br />

CXCR4/CXCL12 and CXCR3/CXCL11 axes.<br />

2045 General Poster Session (Board #12H), Sat, 1:15 PM-5:15 PM<br />

A phase II multicentric study <strong>of</strong> sunitinib administered as upfront therapy<br />

in glioblastoma: A study by the GEINO group. Presenting Author: Carmen<br />

Balana, Institut Catala d’Oncologia Badalona, Barcelona, Spain<br />

Background: Sunitinib is a multitargeted tyrosine kinase inhibitor that has<br />

direct antitumor and antiangiogenesis activity through targeting VEGFR<br />

1-2, PDGFR �-�, c-kit, bFGF, (CSF-1), FLT3 and RET. Experimental<br />

studies in GB mice showed angiogenic activity, prolonged survival and<br />

synergy with irradiation Methods: Eligible pts were �75 years with GB not<br />

amenable to resection, PS�2, MMS �25. Treatment was sunitinib<br />

37.5mg daily for 8 w before radiotherapy, during radiotherapy (60Gy, 6 w)<br />

and after radiotherapy until disease progression. Primary endpoint was<br />

overall response (OR) (RANO criteria) after 8w<strong>of</strong>sunitinib treatment<br />

before radiotherapy. Secondary endpoints were % pts free <strong>of</strong> neurological<br />

deterioration before radiotherapy, % pts who completed radiotherapy, PFS,<br />

OS and 1-year survival. We used a Simon 2-stage design (12 ¡20) based<br />

on OR to calculate the number <strong>of</strong> patients needed to detect at least 10% <strong>of</strong><br />

responses with � error <strong>of</strong> 0.05 and � error <strong>of</strong> 0.10. Twelve patients were<br />

included in the first phase and one response was needed to continue study<br />

accrual. Results: 12 pts were enrolled: 7 males, median age 65 years,<br />

PS�1: 8pts, median MMS 26.5. Neurological deficit: 5pts, DXM treatment<br />

10/12, non-EIDS 3/12. Treatment: 5 pts completed 8w<strong>of</strong>therapy, 2pts<br />

completed 7 w, 4 pts � 6w. Toxicity G3 and 4: diarrhea (1pt), asthenia<br />

(2pts), skin and EDDPP (2), mucositis (2), fatal CNS hemorrhage (1),<br />

hypercholesterolemia and hypertriglycemia (1). OR was 1/12 SD, 11/12 P;<br />

radiological response was SD in 5 pts but 4 pts progressed neurologically.<br />

Only 2 pts completed radiotherapy plus sunitinib. The only SD received<br />

treatment for 18 w. Median PFS was 7.5w (CI 95% 5.8-9.2), OS 16w (CI<br />

95% 0.5-23.7), 1year OS: 0%. Conclusions: the trial was closed after the<br />

first stage due to lack <strong>of</strong> response. Sunitinib is an inactive agent in<br />

glioblastoma.<br />

2044 General Poster Session (Board #12G), Sat, 1:15 PM-5:15 PM<br />

Use <strong>of</strong> tumor-targeting salmonella typhimurium to eradicate human glioma<br />

in an orthotopic model in nude mice. Presenting Author: Masashi Momiyama,<br />

AntiCancer Inc., San Diego, CA<br />

Background: Prognosis remains poor for glioma and therefore requires new<br />

approaches. We have previously developed a genetically-engineered strain<br />

<strong>of</strong> Salmonella typhimurium, A1-R, that targets tumors without overt toxicity<br />

in mouse models (Proc. Natl. Acad. Sci. USA 102, 755-760, 2005; Cancer<br />

Research 66, 7647-7652, 2006; Proc. Natl. Acad. Sci. USA 104,<br />

10170-10174, 2007; Expert Opinion on Drug Discovery 7, 73-83, 2012).<br />

In the present study, we demonstrated that Salmonella typhimurium A1-R<br />

can inhibit and eradicate human glioma in an orthotopic nude mouse<br />

model. Methods: S. typhimuirum A1-R was administered by injection<br />

through a craniotomy open-window or intravenously in nude mice. To<br />

establish the model, 2�105 U87 human glioma cells, expressing red<br />

fluorescent protein (RFP), were injected stereotactically into the mouse<br />

brain through the craniotomy open window. Two weeks after glioma-cell<br />

implantation, mice were treated with S. typhimurium A1-R (2�107 CFU /<br />

200 �l intravenous injection [i.v.] or 1�106 CFU / 1 �l intracranial<br />

injection [i.c.]) once a week for 3 weeks. Results: Brain tumors were<br />

observed by fluorescence imaging through the craniotomy open window<br />

over time. S. typhimurium A1-R, administered i.c., inhibited brain tumor<br />

growth 7.6-fold compared with untreated mice (p � 0.009) and improved<br />

survival 73% (p � 0.001). Two <strong>of</strong> ten mice had their tumors eradicated.<br />

Intravenous administration <strong>of</strong> S. typhimurium A1-R was not effective.<br />

Conclusions: The results <strong>of</strong> the present study demonstrate that bacterial<br />

therapy <strong>of</strong> glioma is a novel, effective and safe treatment strategy in a<br />

highly treatment-resistance cancer in an orthothopic model, a first step<br />

toward clinical development.<br />

2046 General Poster Session (Board #13A), Sat, 1:15 PM-5:15 PM<br />

Neuraxis imaging in leptomeningeal metastasis: A retrospective case<br />

series. Presenting Author: Marc C. Chamberlain, Fred Hutchinson Cancer<br />

Research Center, Seattle Cancer Care Alliance, Seattle, WA<br />

Background: (and Objective) Leptomeningeal metastasis (LM) is a central<br />

nervous system metastatic complication <strong>of</strong> cancer that affects the entire<br />

neuraxis. Quantify imaging (brain and spine MRI and radio-isotope cerebrospinal<br />

fluid [CSF] flow study) abnormalities in a retrospective case series <strong>of</strong><br />

patients with LM. Methods: 240 adult patients with LM (125 non-brain<br />

solid tumor patients with positive CSF cytology; 40 non-brain solid tumor<br />

patients with negative CSF cytology; 50 lymphoma and 40 leukemia<br />

patients with positive CSF flow cytometry) underwent prior to treatment<br />

brain and entire spine MRI and radio-isotope CSF flow studies (FS). Results:<br />

Neuraxis MRI in pathologically defined patients was more <strong>of</strong>ten normal in<br />

hematologic tumors (80-84%) compared to solid tumors (60%). Similarly,<br />

FS was more <strong>of</strong>ten normal in hematologic tumors (90-92%) compared to<br />

solid tumors (72-75%). However, neuraxis MRI and FS abnormalities (i.e.<br />

CSF flow obstruction; nodular subarachnoid or parenchymal disease;<br />

hydrocephalus) altered therapy by requiring CSF diversion, site specific<br />

radiotherapy, systemic chemotherapy or recommending no further therapy<br />

in one third <strong>of</strong> CSF cytology positive solid tumors and 15% <strong>of</strong> CSF flow<br />

cytometry positive hematologic tumors. Conclusions: Notwithstanding less<br />

frequent imaging abnormalities in hematologic tumors, similar to solid<br />

tumors imaging abnormalities frequently result in treatment alteration for<br />

patients with LM. Consequently, neuraxis imaging is recommended in both<br />

tumor categories in patients being considered for LM-directed and in<br />

particular intra-CSF chemotherapy treatment.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


2047 General Poster Session (Board #13B), Sat, 1:15 PM-5:15 PM<br />

The effect <strong>of</strong> the addition <strong>of</strong> chemotherapy to radiotherapy on cognitive<br />

function in patients with low-grade glioma: Secondary analysis <strong>of</strong> RTOG<br />

98-02. Presenting Author: Roshan Sudhir Prabhu, Winship Cancer Institute,<br />

Emory University, Atlanta, GA<br />

Background: The addition <strong>of</strong> PCV chemotherapy to radiotherapy (RT) for<br />

patients with WHO grade II glioma improves progression free survival (PFS)<br />

and overall survival (OS), for patients surviving at least 2 years (Shaw, J Clin<br />

Oncol 26: 2008). The effect <strong>of</strong> therapy intensification on cognitive function<br />

(CF) remains a concern in this population with substantial long term<br />

survival. Methods: 251patients with WHO grade II glioma and age � 40<br />

with any extent <strong>of</strong> resection, or age � 40 with subtotal resection/biopsy<br />

were randomized to RT (54Gy) or RT � PCV. 111 patients with age � 40<br />

and gross total resection were observed. CF was assessed by mini-mental<br />

status exam (MMSE)at baseline and years 1, 3, and 5 for patients without<br />

progression. Change in MMSE score from baseline <strong>of</strong> � 3 points was<br />

considered clinically significant. Results: Overall, very few patients experienced<br />

significant decline in MMSE score, with a median follow-up time <strong>of</strong><br />

9.7 years for alive patients. There were no significant differences in the<br />

proportion <strong>of</strong> patients experiencing MMSE decline between study arms at<br />

any time point. The table below summarizes MMSE change from baseline<br />

over time. Neither baseline MMSE score nor change in MMSE at year 1<br />

significantly predicted for OS or PFS, but there was a trend towards worse<br />

OS for patients with MMSE loss <strong>of</strong> � 2 points [HR 1.73, 95% CI (0.86,<br />

3.47), p�0.12]. Conclusions: The MMSE is a relatively insensitive tool that<br />

has not been validated in patients receiving cranial RT, and subtle changes<br />

in CF may have been missed. However, the addition <strong>of</strong> PCV to RT for low<br />

grade glioma did not result in significantly higher rates <strong>of</strong> MMSE decline<br />

than RT alone or observation. There was a trend towards an MMSE decline<br />

<strong>of</strong> � 2 points at year 1 predicting for worse OS.<br />

Observation (%) RT (%) RT � PCV (%)<br />

Significant MMSE decline<br />

Year 1 0 / 62 (0) 5 / 74 (6.8) 2 / 51 (3.9)<br />

Year 3 0 / 44 (0) 1 / 48 (2.1) 0 / 43 (0)<br />

Year 5<br />

No MMSE change<br />

0/27(0) 0/22(0) 2/25(8)<br />

Year 1 60 / 62 (96.8) 66 / 74 (89.2) 44 / 51 (86.3)<br />

Year 3 44 / 44 (100) 45 / 48 (93.8) 38 / 43 (88.4)<br />

Year 5<br />

Significant MMSE gain<br />

27 / 27 (100) 21 / 22 (95.5) 20 / 25 (80)<br />

Year 1 2 / 62 (3.2) 3 / 74 (4.1) 5 / 51 (9.8)<br />

Year 3 0 / 44 (0) 2 / 48 (4.2) 5 / 43 (11.6)<br />

Year 5 0 / 27 (0) 1 / 22 (4.5) 3 / 25 (12)<br />

2049 General Poster Session (Board #13D), Sat, 1:15 PM-5:15 PM<br />

Predictive role <strong>of</strong> MGMT status in recurrent glioblastoma (GBM) patients<br />

(PTS) treated with antiangiogenic drug (AD) plus temozolomide (TMZ) or<br />

CPT-11. Presenting Author: Giuseppe Lombardi, Medical Oncology 1,<br />

Istituto Oncologico Veneto-IRCCS, Padua, Italy<br />

Background: Methylation and silencing <strong>of</strong> MGMT promoter is a favourable<br />

predictive factor in PTS with GBM treated with single alkylating agent such<br />

as TMZ. Yet, MGMT is an important resistance determinant to CPT-11<br />

activity. AD can be administered in second line treatment as single agent or<br />

in combination to cytotoxic drugs. We analyzed the predictive role <strong>of</strong> MGMT<br />

status in PTS treated with AD plus TMZ or CPT-11 as second line<br />

treatment. Methods: Retrospectively, 55 PTS were analyzed: 36 (65%) with<br />

unmethlyated MGMT, 19 (35%) with methylated MGMT; 3 (5%) PTS with<br />

IDH1 mutations. 17 (31%) PTS performed a reoperation before the second<br />

line treatment and MGMT status was changed in 2 (18%) PTS, IDH1 status<br />

was unchanged in all PTS. 32 PTS were treated with sorafenib 800mg/die<br />

plus TMZ 40mg/m2/die, 23 with bevacizumab 10mg/Kg plus irinotecan<br />

every two weeks. Tumor response was evaluated by clinician assessment<br />

and by MRI according to RANO criteria every two months or when clinically<br />

indicated. Results: Among all PTS, median PFS was 2.7 months (95%CI<br />

1.5-3.5), median OS from start <strong>of</strong> AD was 7.3 months (95%CI 6.02-8.5),<br />

6-month PFS was 32%; no significant differences were observed and<br />

MGMT status was balanced between the two AD groups (p�0.05).<br />

Analyzing MGMT status at first surgery, according to univariate analyses<br />

there were no significant differences in terms <strong>of</strong> PFS (3.5ms vs 2.1ms;<br />

p�0.2), OS (6.5ms vs 7.2ms; p�0.1) and 6-PFS (HR�0.2, CI95%<br />

0.05-1.07) between PTS with unmethylated and methylated MGMT,<br />

respectively. On multivariate analysis, adjusted for performance status, age<br />

and cytotoxic drug, MGMT status was not statistically significant in terms <strong>of</strong><br />

PFS (p�0.8) and OS (p�0.1). Yet, no significant differences emerged with<br />

MGMT status at second surgery as well as analyzing the two AD groups,<br />

separately. Conclusions: MGMT status might not be a predictive factor in<br />

recurrent GBM patients treated with AD plus TMZ or CPT-11 as second line<br />

treatment, although MGMT status may change in some PTS between first<br />

and second surgery. In conclusion, the outcome <strong>of</strong> patients with recurrent<br />

GBM receiving AD plus TMZ or CPT-11 is not significantly influenced by<br />

MGMT methylation status.<br />

Central Nervous System Tumors<br />

127s<br />

2048 General Poster Session (Board #13C), Sat, 1:15 PM-5:15 PM<br />

First results <strong>of</strong> radiotherapy after hyperbaric oxygenation with temozolomide<br />

for high-grade gliomas. Presenting Author: David Hamid Khelif,<br />

Service de Radiothérapie Groupe Hospitalier sud Reunion, Saint Pierre,<br />

Reunion<br />

Background: The outcome <strong>of</strong> patients with high-grade gliomas remains poor<br />

despite modern therapeutic arsenal. The objective <strong>of</strong> this study was to<br />

assess the feasibility and efficacy <strong>of</strong> radiotherapy (RT) given immediately<br />

after hyperbaric oxygenation (HBO) with Stupp protocol. Methods: All<br />

patients with histologically confirmed high-grade gliomas from 2008 till<br />

2011 coveraged at GHSR hospital were enrolled. Patients underwent<br />

Stupp protocol consisting in 60 Gy RT with a daily dose <strong>of</strong> 2 Gy for 5 days<br />

per week administrated immediately after HBO. Temozolomide (TMZ) was<br />

administrated at the daily dose 75 mg/m², 7 days per week followed by 6<br />

cycles <strong>of</strong> TMZ 150 to 200 mg/m² for 5 days each 28 day-cycle. HBO<br />

schedule was approximately 15 min <strong>of</strong> compression with air, 60 min <strong>of</strong><br />

100% oxygen inhalation and 10 min <strong>of</strong> decompression with oxygen.<br />

Results: A total <strong>of</strong> 21 patients were diagnosed and histologically confirmed<br />

high grade gliomas. Five were excluded because <strong>of</strong> HBO contraindications.<br />

Twelve patients (75%) had undergone debulking surgery. The time interval<br />

from completion <strong>of</strong> decompression to start <strong>of</strong> irradiation was less than 15<br />

minutes (mean 14.25, range 8-18). Twelve patients (75%) received the<br />

complete dose <strong>of</strong> RT and TMZ. HBO was completed for 7 (43.75%). Five<br />

(31.25%) were temporarily stopped and 4 (25%) were definitively stopped.<br />

One patient (6.25%) suffered from pancytopenia with grade 4 thrombopenia<br />

and grade 3 neutropenia. The median follow up was 46.95 weeks<br />

(range 5.7-108.3). Nine patients (56.25%) are still alive. Four (25%) have<br />

a survival more than 16 months. Conclusions: HBO with Stupp protocol is<br />

feasible and promising but requiring a large multicentric study.<br />

2050 General Poster Session (Board #13E), Sat, 1:15 PM-5:15 PM<br />

Phase II trial <strong>of</strong> sunitinib malate and lomustine in patients with temozolomide<br />

refractory recurrent low-grade and anaplastic gliomas. Presenting<br />

Author: Johnny Duerinck, UZ Brussel, Brussels, Belgium<br />

Background: Recurrent low-grade and anaplastic gliomas eventually transform<br />

to a higher grade that is characterized by neo-angiogenesis. Sunitinib<br />

inhibits multiple tyrosine kinase receptors (including VEGFR, PDGFR and<br />

c-Kit) but has no meaningful activity as a mono-therapy for recurrent<br />

glioblastoma when given at a daily dose <strong>of</strong> 37,5 mg (Neyns et al. J<br />

Neurooncol. 2011). We investigated sunitinib in combination with lomustine<br />

(CCNU) for the treatment <strong>of</strong> patients (pts) with temozolomide (TMZ)<br />

refractory recurrent low-grade or anaplastic glioma. Methods: Pts received a<br />

daily dose <strong>of</strong> 25 mg sunitinib for 28 consecutive days followed by a 14 days<br />

treatment-free interval. CCNU was administered as a single dose (80<br />

mg/m²) on day 14 <strong>of</strong> a 6w cycle. T1 � Gd and T2/FLAIR weighted MRI<br />

images were obtained q6 wks. 18FET-PET was performed at baseline and<br />

reassessed in responding pts. Results: 13 pts were enrolled (mean age 40<br />

[range 33-49]; M/F 8/5; KPS 80-90/70-60: 8/5 pts). All pts had PD<br />

following surgery, RT and TMZ. In 7 pts, treatment was initiated at 2nd<br />

recurrence, in 4 pts at 3rd recurrence and in 2 pts at the 4th recurrence.<br />

Most frequent AEs where fatigue (gr 2: n� 3; gr 3: n� 1), thrombopenia (gr<br />

2: n� 1gr3:n�3 gr4: n� 1), neutropenia (gr 2: n� 2; gr3: n� 2; gr4: n�<br />

2) and lymphopenia (gr 2: n�1; gr 3: n�2; gr 4: n�1). In 5/13 pts CCNU<br />

had to be discontinued because <strong>of</strong> AEs. Treatment with sunitinib was<br />

continued in these cases without reoccurrence <strong>of</strong> AEs. BOR according to<br />

RANO criteria: 1 CR, 1 PR (not-confirmed) and 2 SD (DCR: 4/13� 31%).<br />

In the patient with CR, FET-PET indicated a complete metabolic response.<br />

After a mean FU <strong>of</strong> 7 mths (range 2 -19), 5 pts are alive. The 6-month PFS<br />

is 23% (3/13 pts); median PFS is 1,8 mths (95%CI 1.0 - 2,7). A durable<br />

disease control was obtained in 3 pts (TTP respectively 14.8, 11.8� and<br />

19.2� mths). Conclusions: in this heavily pretreated population with<br />

recurrent low-grade and anaplastic glioma the combination <strong>of</strong> sunitinib and<br />

CCNU is associated with acceptable toxicity and <strong>of</strong>fers a durable progressionfree<br />

survival in a subgroup <strong>of</strong> pts. Molecular predictive factors identifying<br />

the sensitive population would be needed to justify further clinical<br />

investigation <strong>of</strong> this combination.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


128s Central Nervous System Tumors<br />

2051 General Poster Session (Board #13F), Sat, 1:15 PM-5:15 PM<br />

Phase II study <strong>of</strong> PX-866 in recurrent glioblastoma. Presenting Author:<br />

Marshall W. Pitz, CancerCare Manitoba, University <strong>of</strong> Manitoba, Winnipeg,<br />

MB, Canada<br />

Background: Glioblastoma (GBM) is the most aggressive malignancy <strong>of</strong> the<br />

central nervous system. The majority <strong>of</strong> GBM have genetic changes that<br />

increase the activity <strong>of</strong> the phosphatidylinositol-3-OH kinase (PI3K) signal<br />

transduction pathway, critical for cell motility, proliferation, and survival.<br />

We present the interim results <strong>of</strong> PX-866, an oral PI3K inhibitor, in patients<br />

(pts) with recurrent GBM. Methods: Pts with histologically confirmed GBM<br />

at first recurrence after treatment with chemoradiation and adjuvant<br />

temozolomide are given PX-866 8 mg daily on this single-arm phase II<br />

study. MRI and clinical exam are done every 8 weeks to determine<br />

treatment response. The trial has a 2-stage design with dual endpoints <strong>of</strong><br />

objective response and early progression (within 8 weeks). In Stage I, 15<br />

pts are evaluated and if 0 responses and 10 or more early progressions are<br />

seen, enrolment will stop. Otherwise, Stage II will enrol another 15 pts for<br />

efficacy analysis. Tumour tissue is collected for analysis <strong>of</strong> potential<br />

markers <strong>of</strong> PI3K inhibitory activity (PTEN, EGFRvIII, PIK3CA mutations).<br />

Results: Seventeen pts have been enroled to date: 14 evaluable for response<br />

and 15 for toxicity. Median age was 54 years (range 35-70), with 7 females<br />

and 10 males. No pts had received treatment for recurrent GBM, and<br />

median time between initial diagnosis and study enrolment was 300 days<br />

(range: 113-447 days). Pts have received a median <strong>of</strong> one 8-week cycle <strong>of</strong><br />

PX-866 (range: 1-4). Twelve pts have discontinued therapy, 9 due to<br />

disease progression and 3 due to grade 3/4 liver enzyme abnormalities.<br />

Other adverse effects have included fatigue (10 pts/1 grade 3), diarrhea (6<br />

pts/3 grade 3), nausea (7 pts/0 grade 3), vomiting (6 pts/0 grade 3),<br />

lymphopenia (14 pts/3 grade 3). Stage I response data are premature; it is<br />

not yet known if the trial will continue to Stage II. Archival tissue is<br />

available on all patients and is undergoing analysis. Conclusions: This is one<br />

<strong>of</strong> the first trials <strong>of</strong> a PI3K inhibitor in pts with recurrent GBM. PX-866 has<br />

been relatively well tolerated. Stage I response data are premature; while it<br />

is not yet known if the criteria will be met to continue to Stage II, prolonged<br />

SD has been observed in some pts. The correlative biomarker assays<br />

underway will be important to understand this observation.<br />

2053 General Poster Session (Board #13H), Sat, 1:15 PM-5:15 PM<br />

Correlation <strong>of</strong> large brain edema with favorable prognosis in patients with<br />

single brain metastases. Presenting Author: Matthias Preusser, Department<br />

<strong>of</strong> Medicine I and Comprehensive Cancer Center CNS Tumours Unit,<br />

Medical University <strong>of</strong> Vienna, Austria, Vienna, Austria<br />

Background: The sub-cohort <strong>of</strong> brain metastases (BM) patients (pts) with<br />

single BM has relatively favorable survival times, but novel prognostic<br />

factors are needed to individualize patient management. Methods: We<br />

retrospectively evaluated pre-operative magnetic resonance images in<br />

patients, who underwent neurosurgical resection <strong>of</strong> a single BM. The<br />

localization and largest diameter <strong>of</strong> the BM was recorded. The extent <strong>of</strong><br />

brain edema (BE) was graded on contrast-enhanced T1, T2 and fluidattenuated<br />

inversion recovery (FLAIR) images as follows: BE �1cm, BE �1<br />

cm without affection <strong>of</strong> the contralateral hemisphere, BE �1cm with<br />

affection <strong>of</strong> the contralateral hemisphere. Further, immunohistochemically<br />

assessed expression <strong>of</strong> hypoxia-induced factor 1 alpha (HIF1a) was<br />

analyzed in each BM tissue specimen. Results: 159 pts were studied (81<br />

male, 78 female; age range 24 to 80 years, median 58). The primary<br />

tumors were lung carcinoma in 74, breast cancer in 26, kidney cancer in<br />

13, colorectal cancer in 11, melanoma in 9 and other tumor types in 26<br />

cases. At univariate analysis, we found a significant positive correlation <strong>of</strong><br />

overall survival time (OS) with young age, Karn<strong>of</strong>sky index �80, nonmelanoma<br />

histology, and, surprisingly, large BE (p�0.05, log-rank test). At<br />

multivariate analysis, pts age, histological diagnosis and extent <strong>of</strong> BE<br />

remained independent prognostic parameters (Cox regression model,<br />

p�0.05). We found a significant positive correlation <strong>of</strong> extent <strong>of</strong> BE with<br />

expression <strong>of</strong> HIF1a (Mann-Whitney-U test, p�0.003). Conclusions: Large<br />

BE positively and independently correlates with favorable prognosis in pts<br />

operated for single BM. Peritumoral BE correlates with HIF1a expression,<br />

indicating a possible role <strong>of</strong> tumor hypoxia in its formation.<br />

Parameter n<br />

Median<br />

OS/days<br />

(95% CI)<br />

p value<br />

(univariate)<br />

p value<br />

(multivariate)<br />

Age � 60 years 90 435 (334-536) 0.02 0.014<br />

�60 years 69 263(121-405)<br />

Karn<strong>of</strong>sky index �80 29 179 (37-321) 0.047 0.242<br />

�80 110 443(344-542)<br />

Histological diagnosis Non-melanoma 150 423 (329-517) �0.001 �0.001<br />

Melanoma 9 130 (95-165)<br />

Extent <strong>of</strong> BE �1cm 26 209(137-281) 0.004 0.008<br />

�1cm, only ipsilateral<br />

hemisphere<br />

105 435 (334-536)<br />

�1cm, ipsi- and<br />

contralateral hemisphere<br />

28 575 (235-915)<br />

2052 General Poster Session (Board #13G), Sat, 1:15 PM-5:15 PM<br />

Concurrent intrathecal methotrexate and liposomal cytarabine for the<br />

treatment <strong>of</strong> leptomeningeal metastasis from solid tumors. Presenting<br />

Author: Brian J. Scott, University <strong>of</strong> California, San Francisco, San<br />

Francisco, CA<br />

Background: Leptomeningeal metastasis (LM) from solid tumors is typically<br />

a late manifestation <strong>of</strong> disease with a median survival <strong>of</strong> weeks to a few<br />

months. Treatment is palliative, with no widely accepted standard <strong>of</strong> care.<br />

Options include intrathecal (IT) or systemic chemotherapy, radiation<br />

therapy or ventriculoperitoneal shunting. Randomized trials comparing<br />

single agent IT methotrexate to liposomal cytarabine have shown similar<br />

efficacy and tolerability. There is limited data, however, on the use <strong>of</strong><br />

combination IT chemotherapy in solid tumor LM. Methods: We conducted a<br />

retrospective cohort study <strong>of</strong> 19 subjects treated for LM from solid tumors<br />

at a single institution. In addition to therapies directed at active solid tumor<br />

sites, each subject received IT liposomal cytarabine plus IT methotrexate<br />

injections every two weeks. Survival data and treatment-related toxicities<br />

were determined by systematic chart review. Results: LM was diagnosed by<br />

CSF cytology in 12/19 (63%), while the remainder were diagnosed by<br />

clinical and MRI findings. The most common cancer types were breast<br />

7(37%), glioblastoma 6(32%) and lung 3(16%). The majority 18(95%)<br />

had active systemic or parenchymal brain disease at the time <strong>of</strong> treatment,<br />

requiring systemic chemotherapy 18(95%) or radiation therapy 13(68%).<br />

The median number <strong>of</strong> IT treatments was 4(range 1-9). Treatment was<br />

interrupted due to toxicity in 3(17%), while 7(37%) experienced � CTCAE<br />

grade III toxicities, most commonly meningitis 3(16%). Treatment was<br />

stopped in 7/19(37%) following complete cytologic response 6/11(55%)<br />

or radiographic clearance 1/7(14%). The median overall survival was 96<br />

days(n�6; range 29-158), median time to neurologic progression was 46<br />

days (n�9; range 6-101) and the most common cause <strong>of</strong> death was<br />

progression <strong>of</strong> systemic disease 4(67%). Conclusions: Combination IT<br />

chemotherapy was reasonably well-tolerated, even in a population also<br />

receiving chemotherapy for progressive systemic disease. IT-related adverse<br />

events occurred at rates similar to previously reported single agent<br />

trials. Prospective evaluation is necessary to determine whether there is a<br />

survival benefit compared to single agent IT chemotherapy.<br />

2054 General Poster Session (Board #14A), Sat, 1:15 PM-5:15 PM<br />

Management <strong>of</strong> primary intraocular lymphoma (PIOL): Results from the<br />

prospective German PIOL registry (PIOL-R). Presenting Author: Kristoph<br />

Jahnke, Hematology and Oncology, Charité Campus Benjamin Franklin,<br />

Berlin, Germany<br />

Background: Primary intraocular lymphoma (PIOL) is a very rare disorder,<br />

and the optimal treatment is yet to be defined. Here, we report clinical<br />

characteristics and outcome <strong>of</strong> patients with PIOL enrolled in the prospective<br />

German PIOL registry (PIOL-R). Methods: Patient data in this prospective,<br />

non-interventional multicenter study were compiled by standardized<br />

questionnaires sent to Ophthalmology and Hematology/Oncology centers in<br />

Germany. All histologically or cytologically confirmed immunocompetent<br />

PIOL patients were eligible. Results: Fifteen patients (8 female, median age<br />

66 years, median Karn<strong>of</strong>sky performance status 90%) from 4 centers were<br />

included between August 2008 and January 2012. Median follow-up was 7<br />

months. Median time from first occurrence <strong>of</strong> symptoms to PIOL diagnosis<br />

was 5 (range, 1-11) months. All PIOL were <strong>of</strong> diffuse large B-cell histology.<br />

Eight patients had concomitant (n�3), prior (n�2) and/or subsequent<br />

(n�4) parenchymal brain involvement. First-line treatment included methotrexate<br />

(MTX)-, rituximab- and/or ifosfamide-based systemic chemotherapy<br />

in 12 (including high-dose chemotherapy [HDCT] with autologous stem cell<br />

transplantation [ASCT] in 2), intraocular (i.o.) chemotherapy with MTX<br />

(n�2) and/or rituximab (n�6) in 7, and ocular radiation in one patient(s).<br />

Two patients received prophylactic intrathecal (i.th.) treatment with MTX<br />

and none had whole-brain irradiation. The PIOL remission rate was 100%<br />

(complete remission in 11/14 evaluable patients and partial remission in 3<br />

patients). Four patients relapsed in the brain after 5, 6, 21 and 23 months<br />

and received salvage treatment with MTX and/or ifosfamide (n�3) and<br />

HDCT with ASCT (n�1). No ocular or systemic relapses were observed.<br />

Median progression-free survival was 23 (95% confidence interval, 19-26)<br />

months. All patients are alive. Conclusions: Although the awareness <strong>of</strong> PIOL<br />

and diagnostic possibilities have improved over the past 2 decades, time<br />

from symptom presentation to diagnosis seems to remain at previously<br />

reported levels. There appears to be a shift from local ocular treatments and<br />

radiation to systemic therapy as compared to anecdotal data with promising<br />

response and survival rates.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


2055 General Poster Session (Board #14B), Sat, 1:15 PM-5:15 PM<br />

Effect <strong>of</strong> the integrin inhibitor cilengitide on TGF-beta signaling. Presenting<br />

Author: Michael Weller, University Hospital Zurich, Zurich, Switzerland<br />

Background: Glioblastoma is the most malignant form <strong>of</strong> intrinsic brain<br />

tumors. One <strong>of</strong> its characteristic features, angiogenesis, is mediated by the<br />

expression <strong>of</strong> integrins �v�3 and �v�5 on tumor-associated vasculature.<br />

Cilengitide is a synthetic peptide which inhibits ligand binding to the �v�3<br />

and �v�5 integrins. The addition <strong>of</strong> cilengitide to the standard treatment<br />

regimen <strong>of</strong> radiochemotherapy in patients with newly diagnosed glioblastoma<br />

demonstrated promising results without significant toxicity which led<br />

to the initiation <strong>of</strong> a randomized phase registration III trial. Besides other<br />

functions, av-integrins can be involved in the activation <strong>of</strong> the cytokine<br />

transforming growth factor (TGF)-� which contributes to the malignant<br />

phenotype <strong>of</strong> glioma cells. Methods: Integrin expression was examined by<br />

immunohistochemistry and flow cytometry. The effects <strong>of</strong> cilengitide on<br />

TGF-� signaling in glioma cells were investigated by PCR, immunoblot and<br />

ELISA. Further assessments included reporter assays and glioma xenografts<br />

in nude mice. Results: The target integrins <strong>of</strong> cilengitide are<br />

expressed in glioblastoma blood vessels and tumor cells. Exposure to<br />

cilengitide results in detachment and reduced phosphorylation <strong>of</strong> Smad2<br />

in most glioma cell lines, including stem-like glioma cells. Furthermore,<br />

exposure <strong>of</strong> glioma cells to cilengitide is associated with reduced TGF-�mediated<br />

reporter gene activity. Smad2 phosphorylation, but not attachment,<br />

can be rescued by co-exposure to recombinant TGF-�. Cilengitide<br />

results in decreased TGF-�1 and TGF-�2 mRNA and protein expression and<br />

loss <strong>of</strong> transcriptional activity <strong>of</strong> the TGF-� promoter. Blocking antibodies<br />

to integrins �v, �v�3 or�v�5, or silencing <strong>of</strong> integrins �v, �3, �5 or�8 by<br />

RNA interference mimics the effects <strong>of</strong> cilengitide on TGF-� expression.<br />

Intracranial LN-308 glioma xenografts in nude mice display reduced<br />

pSmad2 levels in response to cilengitide. Conclusions: This study demonstrate<br />

a novel mechanism which may in part mediate anti-glioblastoma<br />

activity <strong>of</strong> cilengitide. These anti-TGF-� properties <strong>of</strong> cilengitide might be<br />

exploited in future clinical trials.<br />

2057 General Poster Session (Board #14D), Sat, 1:15 PM-5:15 PM<br />

Natural history <strong>of</strong> glioblastoma in the modern era: Longitudinal results from<br />

a large prospective Italian register. Presenting Author: Alba Ariela Brandes,<br />

Medical Oncology Department, Bellaria-Maggiore Hospital, Azienda USL <strong>of</strong><br />

Bologna, Bologna, Italy<br />

Background: The role <strong>of</strong> temozolomide (TMZ) concurrent with and adjuvant<br />

to radiotherapy (RT) in the treatment <strong>of</strong> glioblastoma (GBM) has been<br />

demonstrated by the EORTC 22981/26981-NCIC CE.3 (EORTC/NCIC)<br />

randomized trial, and has been widely accepted as the standard treatment.<br />

The impact <strong>of</strong> RT/TMZ in the general patient population was assessed in<br />

the context <strong>of</strong> the Registry <strong>of</strong> the Project <strong>of</strong> Emilia-Romagna Region in<br />

Neuro-Oncology (PERNO), that represents the first italian prospectic<br />

observational population-based study in neuro-oncology. Methods: Approvals<br />

from local Ethical Committees were obtained by 8 participating centres.<br />

Patients (pts) who met the following inclusion criteria were evaluated: age<br />

�18 years; PS 0-3; histologically confirmed GBM, no previous or concomitant<br />

non glial tumoral disease, resident in Emilia Romagna region. The data<br />

were prospectively collected. Results: From January 2009 to January 2011,<br />

194 GBM pts were enrolled. The median age was 62.5, with 26% <strong>of</strong> pts<br />

over 70 years. After surgery pts received RT/TMZ (73%), RT alone (19%),<br />

TMZ alone (5%) or no further treatment (3%); 22% were included in<br />

clinical trials. Median overall survival (OS) was 12.9 months. Pts �70<br />

years received RT/TMZ in 85% <strong>of</strong> cases. In this group <strong>of</strong> pts mOS was 17<br />

months (95%CI: 15.4–18.6). Interestingly, pts �70 years included in<br />

experimental clinical trials showed a significant OS improvement (p�0.04).<br />

In multivariate analysis, only extent <strong>of</strong> surgery (p�0.047), KPS (p�0.01)<br />

and RT/TMZ (p�0.001) were associated with OS in pts �70 years.<br />

Conclusions: Our population data reproduces the beneficial effect <strong>of</strong><br />

RT/TMZ from the EORTC/NCIC randomized trial, confirming how this<br />

successful approach as been widely incorporated in daily practice. Interestingly,<br />

our data suggest that the survival <strong>of</strong> GBM pts treated with RT/TMZ<br />

could be greater than patients treated with RT/TMZ in the EORTC/NCIC<br />

trial at the beginning <strong>of</strong> this century.<br />

Central Nervous System Tumors<br />

129s<br />

2056 General Poster Session (Board #14C), Sat, 1:15 PM-5:15 PM<br />

Impaired expression <strong>of</strong> ketone metabolizing enzymes in malignant gliomas:<br />

Implication for ketogenic diet therapy. Presenting Author: Howard T.<br />

Chang, Michigan State University, East Lansing, MI<br />

Background: Malignant glial cells may have altered expression <strong>of</strong> mitochondrial<br />

enzymes that are key for deriving energy from ketones. The novel<br />

strategy <strong>of</strong> treating malignant gliomas with a ketogenic diet has been tested<br />

in animal models with promising results. To evaluate the applicability <strong>of</strong><br />

these findings to human patients we tested the hypothesis that high grade<br />

human gliomas may lack or have decreased expression <strong>of</strong> mitochondrial<br />

enzymes that are important for metabolizing ketones. Methods: We evaluated<br />

the expression <strong>of</strong> 2 key mitochondrial enzymes involved in ketone<br />

metabolism in 22 patients (17 males and 5 females, mean age 55,range<br />

30-85) with either glioblastoma (GBM, WHO grade IV) or anaplastic glioma<br />

(WHO grade III) (Table). Immunohistochemistry reactions were performed<br />

on formalin-fixed paraffin-embedded sections from brain biopsies, using<br />

antibodies raised against succinyl CoA: 3-oxoacid CoA transferase (OXCT1)<br />

and 3-hydroxybutyrate dehydrogenase 1 (BDH1). Immunoreactivities were<br />

graded using a semi-quantitative scale based on the percentage <strong>of</strong> positive<br />

cells: negative (NEG) � 5%; intermediate (INT) 5-20%; and positive (POS)<br />

more than 20%. Focal non-neoplastic “normal” brain tissue present within<br />

the specimens expressed both enzymes and served as an internal positive<br />

control. Results: Either absent or marked decreased expression <strong>of</strong> OXCT1<br />

and BDH1 were observed in most GBM and anaplastic glioma. Concordance<br />

for absent or marked decrease expression <strong>of</strong> both OXCT1 and BDH1<br />

was observed in 14 <strong>of</strong> the 17 (82%) GBM’s, and 1 <strong>of</strong> 5 (20%) anaplastic<br />

gliomas. Conclusions: These data support the rationale for a pilot clinical<br />

study investigating the therapeutic utility <strong>of</strong> a ketogenic diet in selected<br />

glioma patients. Malignant gliomas with diminished expression <strong>of</strong> key<br />

enzymes (e.g., OXCT1, BDH1) may benefit from the ketogenic diet therapy,<br />

whereas those with positive expression <strong>of</strong> these enzymes likely will not.<br />

OXCT1 BDH1<br />

N NEG INT POS NEG INT POS<br />

GBM 17 10 4 3 11 6 0<br />

Anaplastic glioma 5 0 1 4 1 3 1<br />

2058 General Poster Session (Board #14E), Sat, 1:15 PM-5:15 PM<br />

Quality <strong>of</strong> life in patients with malignant brain tumors. Presenting Author:<br />

Ali Choucair, Norton Neuroscience Institute, Louisville, KY<br />

Background: A positive quality <strong>of</strong> life (QOL) serves as an important outcome<br />

priority in patients with malignant brain tumors whose survival is limited by<br />

tumor resistance to treatment. The FDA and ASCO have designated QOL as<br />

an outcome secondary in importance to survival. We conducted a prospective<br />

longitudinal collection <strong>of</strong> QOL measures (EORTC QLQ-C30, EORTC<br />

QLQ-BN20, and Hospital Anxiety and Depression Scale [HADS]) coupled<br />

with clinical standards <strong>of</strong> care (brain MRI and clinical history and physical<br />

examinations) in patients with newly diagnosed primary brain tumors<br />

(PBT). We also examined the QOL measures over time as related to tumor<br />

progression. Methods: Under an IRB-approved protocol, we reviewed the<br />

validated QOL measures and standard clinical measures from patients who<br />

were newly diagnosed with PBT. The patients were followed every 3 months<br />

over 2 years. No proxies were allowed. A total <strong>of</strong> 52 patients were enrolled<br />

in the study. Survey responses were compared between two tumor groups <strong>of</strong><br />

patients: patients with tumor recurrence and/or died and patients without a<br />

recurrence (n�26 each group). A total <strong>of</strong> 17 patients succumbed to a<br />

tumor-related death. Statistical analysis utilizing non-parametric t-tests<br />

and Wilcoxon sign tests assessed QOL responses. Results: Significant group<br />

differences were noted in the questions <strong>of</strong> the QOL measures surveys with<br />

more negative responses in the recurrence group. The questions with<br />

significant group differences on the EORTC QLQ-C30 ranged from symptoms<br />

<strong>of</strong> dyspnea (p�.05) and difficulty sleeping (p�.05) as well as a poor<br />

QOL (p�.005) and pain interfering with daily activities (p�.05). Significant<br />

group differences <strong>of</strong> questions on the EORTC QLQ-BN20 included<br />

weakness <strong>of</strong> the legs (p�.05), coordination difficulties (p�.005), and<br />

unsteady gait (p�.05). Significant group differences <strong>of</strong> questions on the<br />

HADS reflected a patient who is �slowed down� (p�.01) and �frightened�<br />

(p�.05). Conclusions: Our analysis <strong>of</strong> questions answered by patients with<br />

newly diagnosed PBT may shed light on the impact <strong>of</strong> cancer and its<br />

treatment and potential recurrence on a patient’s QOL. Further research is<br />

warranted to incorporate QOL measurements into improvement <strong>of</strong> patient<br />

care and their use in clinical trials and as a prognostic indicator.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


130s Central Nervous System Tumors<br />

2060 General Poster Session (Board #14G), Sat, 1:15 PM-5:15 PM<br />

Tolerance and feasibility <strong>of</strong> chemotherapy by procarbazine, lomustine, and<br />

vincristine (PCV) for oligodendroglial anaplastic gliomas (OAG). Presenting<br />

Author: Marine Davos, University Hospital Timone, Marseille, France<br />

Background: Chemosensitivity <strong>of</strong> OAG has been documented although their<br />

role as part <strong>of</strong> adjuvant treatment combined to radiotherapy is debated.<br />

Beside temozolomide still under investigation, several studies have underlined<br />

activity <strong>of</strong> PCV regimen in this setting. However, significant toxicity<br />

has been described and dose intensity and schedule duration are not<br />

clearly reported. Methods: This monocentric retrospective analysis included<br />

all pts with OAG treated with at least 1 cycle <strong>of</strong> PVC (lomustine 110mg/m2<br />

every 6 weeks) at our institution from August 2007 to August 2011<br />

(pharmacy charter). Toxicities data were collected from clinical observations<br />

and blood investigations. Results: 40 pts were identified (M/F: 23/17).<br />

Median age was 45y (20 – 72). At the time <strong>of</strong> analyses, 32 pts were still<br />

alive. Histologies were oligodendroglioma (73%) or oligoastrocytoma (27%).<br />

Codeletion 1p19q was observed in 30 % <strong>of</strong> pts. PCV was administered in<br />

first or second line <strong>of</strong> chemotherapy for 60 % and 40 % <strong>of</strong> pts respectively.<br />

Seventy five percent and 52.5% <strong>of</strong> pts completed at least <strong>of</strong> 4 and 6 cycles<br />

respectively. Discontinuation <strong>of</strong> PCV occurred in 45% <strong>of</strong> pts for toxicity<br />

(10%), progression (30%), or other reason (5%). Because <strong>of</strong> toxicity<br />

38/196 (19%) PCV cycles were delayed. Dose was adapted for 73% <strong>of</strong> pts<br />

and decreased under 60% <strong>of</strong> theoretical dose in 30% <strong>of</strong> pts at cycle 4 and<br />

in 57% <strong>of</strong> pts at cycle 6 (table1). Grade III or IV toxicity occurred in 28% <strong>of</strong><br />

pts and only grade I-II in 50%. Neuropathies occurred in 43 % <strong>of</strong> pts.<br />

Conclusions: Despite activity <strong>of</strong> PCV regimen, significant toxicity is associated<br />

to this schedule, that impact feasibility and compliance. PVC<br />

schedule should be redefined taking into account dose intensity applied in<br />

toxicity observed.<br />

Cycles<br />

Cumulative number (nb)<br />

<strong>of</strong> pts under PCV<br />

Nb <strong>of</strong> pts per dose<br />

% <strong>of</strong> theoretical dose <strong>of</strong> lomustine<br />

90–100% 70–80% 50–60% < 50 %<br />

Total nb <strong>of</strong><br />

pts per cycle<br />

1 40 (100%) 23 (57.5%) 13 (32.5%) 3 (7.5%) 1 (2.5%) 6 (15%)<br />

2 34 (85%) 18 (53%) 11 (32%) 4 (12%) 1 (3%) 2 (5%)<br />

3 32 (80%) 12 (38%) 12 (38%) 6 (18%) 2 (6%) 2 (5%)<br />

4 30 (75%) 10 (33%) 11 (37%) 6 (20%) 3 (10%) 3 (7.5%)<br />

5 27 (32.5%) 8 (30%) 8 (30%) 6 (22%) 5 (18%) 6 (15%)<br />

6 21 (52.5%) 4 (19%) 5 (24%) 7 (33%) 5 (24%) 21 (52.5%)<br />

2062 General Poster Session (Board #15A), Sat, 1:15 PM-5:15 PM<br />

The association <strong>of</strong> pretreatment neutrophil to lymphocyte ratio with overall<br />

survival in patients with glioblastoma multiforme (GBM). Presenting<br />

Author: Richard Martin Bambury, Department <strong>of</strong> Medical Oncology, Cork<br />

University Hospital, Cork, Ireland<br />

Background: GBM is the most common and aggressive primary brain tumor.<br />

The neutrophil to lymphocyte ratio (NLR) gives a measure <strong>of</strong> systemic<br />

inflammatory response and lymphopenia, both <strong>of</strong> which are poor prognostic<br />

factors in many malignancies. No published study has assessed the<br />

prognostic impact <strong>of</strong> NLR in GBM. Methods: Patients treated for GBM at our<br />

regional referral centre with assessable complete blood count at first<br />

presentation (prior to corticosteroid therapy or surgery) were identified.<br />

Medical notes were reviewed to extract demographic and treatment data.<br />

Survival curves were estimated via Kaplan-Meier method and compared via<br />

log-rank method. Multivariate analysis was performed via Cox proportional<br />

hazards regression modelling. Results: A total <strong>of</strong> 86 patients were identified,<br />

<strong>of</strong> which 65(76%) were male. Median age at diagnosis was 58 years<br />

(range: 18–76). At the time <strong>of</strong> analysis all patients still alive had � 2 years<br />

follow-up. Median overall survival (OS) was 9.3 months (range: 1-82). 57%<br />

completed the standard adjuvant Stupp protocol and 43% discontinued<br />

early due to disease progression or treatment toxicity. Median OS was 11.2<br />

months in patients with NLR�4 and 7.5 months in patients with NLR�4<br />

(HR 0.59, p�.04). Other significant prognostic factors based on univariate<br />

analysis were consistent with published data (Table). After correcting for<br />

known prognostic factors NLR remained a significant predictor <strong>of</strong> survival<br />

(Table). Conclusions: Recent advances in immunotherapy have highlighted<br />

the importance <strong>of</strong> the immune system in the treatment and prognosis <strong>of</strong><br />

cancer patients. Many GBM patients are on corticosteroids for a significant<br />

proportion <strong>of</strong> their disease course which may abrogate the effects <strong>of</strong> host<br />

immunity on outcome. Nevertheless, we have shown that NLR at diagnosis<br />

is an independent predictor <strong>of</strong> survival in GBM patients. Investigation <strong>of</strong><br />

therapies which harness the immune response are warranted in this<br />

disease.<br />

Univariate Multivariate<br />

Hazard ratio p value p value<br />

NLR (4) 0.59 0.01 0.01<br />

Age (60) 0.47 �0.01 0.01<br />

Surgery (debulking vs biopsy) 0.60 0.01 0.01<br />

Stupp protocol (complete vs incomplete) 0.21 �0.01 �0.01<br />

2061 General Poster Session (Board #14H), Sat, 1:15 PM-5:15 PM<br />

Anaplastic gliomas at first recurrence: Outcome analysis. Presenting<br />

Author: Enrico Franceschi, Medical Oncology Department, Bellaria-<br />

Maggiore Hospital, Azienda USL <strong>of</strong> Bologna, Bologna, Italy<br />

Background: Although anaplastic gliomas show a more favorable response<br />

to postsurgical therapy than glioblastoma, when recurs, salvage therapies<br />

<strong>of</strong>fer only temporary benefit. Moreover, due to the rarity <strong>of</strong> these entities, no<br />

study exists to evaluate the clinical outcomes <strong>of</strong> anaplastic gliomas at first<br />

recurrence. Methods: A retrospective analysis was made using a database<br />

on 249 anaplastic glioma patients (pts) followed prospectively between<br />

01/2000 and 12/2011. Eligibility criteria: age �18 years; PS 0-2;<br />

histological diagnosis at first surgery <strong>of</strong> anaplastic astrocytoma (AA),<br />

anaplastic oligodendroglioma (AO), and anaplastic oligoastrocytoma (AOA);<br />

first recurrence after postsurgical treatments. Results: 163 pts (recurrent<br />

AA:52%, AOA:23%, AO: 25%, mean age: 45 years, range: 21–74) were<br />

enrolled. 1p deletion and MGMT methylation were found in 23% and 56%<br />

<strong>of</strong> evaluable specimens, respectively. At time progression, second surgery<br />

was performed in 51 pts (31%). Chemotherapy was delivered in 128 pts:<br />

temozolomide in 96 pts (75%), PCV regimen in 11 pts (9%), and other<br />

cytotoxic treatments in 21 pts (16%). Median PFS was 7.1 months (95%<br />

CI: 5.8–8.5), being PFS-6 57.8% (95%CI: 49.8%–65.8%), mOS: 18.3<br />

months (95% CI: 13.9–22.8) and OS-12: 63% (95%CI: 55.4%-70.6%).<br />

1p status was not significantly associated with PFS and OS measured from<br />

first recurrence to second progression or death, respectively (p�0.12 and<br />

p�0.08). In the Cox proportional hazard model, PFS at recurrence was<br />

significantly correlated with OS measured at the first disease recurrence<br />

(p�0.00001). Conclusions: our data represent the largest series that<br />

evaluated the outcome <strong>of</strong> anaplastic gliomas at first recurrence. In this<br />

setting, PFS should be considered as a worthy endpoint, being significantly<br />

correlated with OS.<br />

2063 General Poster Session (Board #15B), Sat, 1:15 PM-5:15 PM<br />

Illness intrusiveness and subjective well-being in patients with glioblastoma<br />

multiforme. Presenting Author: Linda Coate, Princess Margaret<br />

Hospital, Toronto, ON, Canada<br />

Background: Glioblastoma multiforme (GBM) is the most common adult<br />

CNS malignancy. The quality-<strong>of</strong>-life (QOL) impact <strong>of</strong> its neurological<br />

sequelae and poor prognosis is poorly understood. In this study we<br />

examined relations between disease severity and mood in GBM patients.<br />

Methods: GBM patients (n�73) completed validated questionnaires examining<br />

depression (CESD), positive affect (ABS), illness intrusiveness (II),<br />

and health-related QOL (EORTC-QLQ30, BN-20). Median age was 53 years<br />

(range 26-75), median time since diagnosis was 1.1 years (range 0.1-<br />

12.4). 88% were on temozolomide. Questionnaire scores were compared to<br />

normative data from GI, GU, Breast, Head and Neck, Lymphoma, and Lung<br />

cancer groups using t-tests. Hierarchical multiple regression analyses<br />

tested the impact <strong>of</strong> disease severity indicators (ECOG; Symptoms, derived<br />

from QLQ30, BN20) and potential moderators on mood and whether II<br />

mediates those effects. Results: GBM patients reported less positive affect,<br />

more depression and greater II than other cancer patients (p�.05).<br />

Increase in symptoms correlated with greater II (��.59; 95% CI [.31, .87],<br />

p�.0001) and depression (��.37; 95% CI [.20, .55], p�.0001) and less<br />

positive affect (��-.04; 95% CI [-.08, -.01], p�.02). Surprisingly, higher<br />

ECOG (i.e., poorer performance status) was associated with less II<br />

(��-3.58; 95%CI [-7.06, -.10], p�.04) and depression (��-2.20; 95%CI<br />

[-4.21, -.19], p�.03). II partially mediated the relations between disease<br />

severity and mood, evidenced by the change in the disease severity<br />

coefficient when II was added to the models (mean change measured by<br />

bootstrap sampling: CESD�.23, 95% CI [.06, .43]; ABS�.21, 95%CI<br />

[.02, .44]). The occurrence <strong>of</strong> other stressful life events was associated<br />

with II (��2.73; 95% CI [.43, 5.02], p�.02), but there was no evidence <strong>of</strong><br />

a moderating effect on this or any other relationship (p�.05). Conclusions:<br />

GBM patients are more distressed than other cancer patients. GBMinduced<br />

lifestyle disruptions partially mediate the association between<br />

disease severity and subjective well-being. Efforts to engage patients in<br />

valued activities and interests, despite the constraints, can help to preserve<br />

QOL.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


2064 General Poster Session (Board #15C), Sat, 1:15 PM-5:15 PM<br />

Impact <strong>of</strong> duration <strong>of</strong> bevacizumab (Bev) treatment in the prognosis <strong>of</strong><br />

adults with recurrent malignant gliomas. Presenting Author: Mohamed Ali<br />

Hamza, University <strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: Bev is the standard treatment for patients with recurrent<br />

glioblastoma (GB) but is also used in treating recurrent anaplastic gliomas<br />

(AG). Differences in outcome between these groups and optimal duration <strong>of</strong><br />

treatment with Bev in pts with recurrent malignant gliomas are not well<br />

defined. We examined the relationship between the duration <strong>of</strong> Bev<br />

treatment and the outcome in pts with GB and AG. Methods: In this<br />

retrospective chart and data review derived from our longitudinal database,<br />

we identified pts with recurrent AG and GB who were treated with Bev alone<br />

or Bev-containing regimens between 2005 and 2009; the data was<br />

analyzed to determine the overall survival (OS) and the progression free<br />

survival (PFS). Results: A total <strong>of</strong> 261 patients with recurrent malignant<br />

gliomas (196 with GB and 65 with AG) were identified. There was no<br />

significant difference between the median length <strong>of</strong> treatment between AG<br />

and GB (5.81�0.66 months vs. 6.77�0.52 months, p�0.32). PFS6 was<br />

34.2% (95% CI, 27.8-41.3) for patients with GB and 44.2% (95% CI,<br />

32.5-56.7) for patients with AG. Patients with GB who were treated �6<br />

months had a significantly higher OS (29.13 months vs. 20.16 months, p�<br />

0.001) compared to those treated �6 months, and a significantly higher<br />

PFS compared to those treated �6months (11.33 months vs. 3.7 months,<br />

p�0.0001). For patients with AG, although treatment �6 months had a<br />

significantly higher PFS (13.93 months vs. 3.53 months, p�0.0001), OS<br />

was not significantly different (months 38.6 vs. 52.5 months, p�0.6)<br />

compared with those treated �6 months. Conclusions: Length <strong>of</strong> treatment<br />

�6 mo with Bev or Bev-containing regimen was associated with improved<br />

PFS in both AG and GB but only the GB subgroup showed improved OS.<br />

These results suggest equivocal survival benefit in patients with AG with<br />

longer duration <strong>of</strong> bevacizumab treatment, which requires further study in<br />

prospective trials.<br />

2066 General Poster Session (Board #15E), Sat, 1:15 PM-5:15 PM<br />

A preclinical study on the combined treatment <strong>of</strong> nimotuzumab and<br />

sirolimus in glioblastoma. Presenting Author: Chee Kian Tham, National<br />

Cancer Centre, Singapore<br />

Background: The human epidermal growth factor receptor (EGFR) is an<br />

ideal therapeutic target for inhibiting glioblastoma (GBM) growth as the<br />

signaling pathway is highly dysregulated in GBM. However, trials so far with<br />

EGFR inhibitors have not shown clinically meaningful improvement in<br />

response rates and survival. One <strong>of</strong> the postulated mechanisms <strong>of</strong> resistance<br />

is the constitutive activation <strong>of</strong> the downstream, PI3K/AKT/mTOR<br />

pathway, independent <strong>of</strong> the upstream EGFR activation status. Hence, the<br />

dual blockage <strong>of</strong> the pathways with an anti-EGFR agent and mTOR inhibitor<br />

is postulated to have synergistic anti-tumour effects. We aim to investigate<br />

the anti-tumour effect <strong>of</strong> combined nimotuzumab (anti-EGFR monoclonal<br />

antibody) and sirolimus (mTOR inhibitor) in a GBM preclinical model.<br />

Methods: Primary human GBM cells were derived from surgical GBM<br />

specimens. The endogenous expressions <strong>of</strong> glial-fibrillary acidic proteins<br />

and EGFR were determined by IHC staining and Western Blot analysis. Cell<br />

viability assays were then carried out in these GBM cells and immortalized<br />

human normal astrocytes (iHNA) using a range <strong>of</strong> concentrations <strong>of</strong><br />

nimotuzumab alone, sirolimus alone or combined treatment. Results: GBM<br />

cells treated with nimotuzumab showed a dose- dependent cell kill and the<br />

optimal concentration was determined to be 2 �g/ml. Treatment <strong>of</strong> the<br />

GBM cells with sirolimus showed that the drug was capable <strong>of</strong> inducing cell<br />

death at varying levels and the optimal level was determined to be 0.1 mM.<br />

Combined treatment with nimotuzumab (2�g/ml) and rapamycin (0.1 mM)<br />

showed a dose dependent cell kill in GBM cells which was not observed in<br />

iNHA cells. The combined treatment resulted in only 10% <strong>of</strong> residual<br />

gliomas at 24 h post treatment. Single treatment with rapamycin has no<br />

cytotoxic effect whereas treatment with nimotuzumab alone exerts a<br />

cytotoxicity effect <strong>of</strong> 33%. Taken together, we observed an additive effect<br />

<strong>of</strong> cell kill when rapamycin is used together with nimotuzumab in human<br />

glioma cells. Conclusions: In this study, combined treatment <strong>of</strong> nimotuzumab<br />

and sirolimus resulted in a greater cytocidal effects in GBM cells<br />

than either agent alone. We will further examine this combination regimen<br />

in a subsequent phase I clinical trial.<br />

Central Nervous System Tumors<br />

131s<br />

2065 General Poster Session (Board #15D), Sat, 1:15 PM-5:15 PM<br />

Rising incidence <strong>of</strong> glioblastoma and meningioma in the United States:<br />

Projections through 2050. Presenting Author: Derek Richard Johnson,<br />

Mayo Clinic, Rochester, MN<br />

Background: In the absence <strong>of</strong> proven environmental or behavioral risk<br />

factors, patient age and sex remain the most important predictors <strong>of</strong><br />

primary brain tumor risk. In coming years, an increasing incidence <strong>of</strong> brain<br />

tumors in the United States can be anticipated based on shifts in the<br />

demographic structure <strong>of</strong> the population. This study provides estimates <strong>of</strong><br />

glioblastoma and meningioma incidence through 2050. Methods: Groupspecific<br />

glioblastoma and meningioma incidence rates based on age<br />

(eleven categories) and sex were calculated from National Cancer Institute<br />

(NCI) Surveillance, Epidemiology, and End Results (SEER) data for the<br />

period 2004-2008. United States Census projections based on data from<br />

the 2000 census were used to compute the size <strong>of</strong> the twenty-two<br />

demographic subpopulations <strong>of</strong> interest in 2010, 2020, 2030, 2040, and<br />

2050. The estimated number <strong>of</strong> new glioblastoma and meningioma<br />

diagnoses for each <strong>of</strong> these years was calculated from the defined<br />

incidence rates and population size. Results: Crude annual tumor incidence<br />

rates were 3.13 per 100,000 persons for glioblastoma and 6.81 per<br />

100,000 persons for meningioma. While the overall size <strong>of</strong> the population<br />

is expected to increase only 42% between 2010 and 2050, the number <strong>of</strong><br />

<strong>American</strong>s over 65 years <strong>of</strong> age, the group at highest risk <strong>of</strong> glioblastoma<br />

and meningioma, is expected to increase by 120%. We estimate that the<br />

number <strong>of</strong> new glioblastoma diagnoses will rise from 10,688 in 2010 to<br />

18,466 in 2050, a 72% increase. Likewise, the number <strong>of</strong> new meningioma<br />

diagnoses will rise from 22,946 to 40,680 over the same period, a<br />

77% increase. Conclusions: The number <strong>of</strong> new glioblastoma and meningioma<br />

diagnoses will increase substantially in the future. This analysis, which<br />

assumes a fixed incidence rate, may underestimate the true magnitude <strong>of</strong><br />

the coming change given reports suggesting that incidence rates <strong>of</strong><br />

glioblastoma and meningioma are rising over time.<br />

2067 General Poster Session (Board #15F), Sat, 1:15 PM-5:15 PM<br />

Bevacizumab failure in glioblastomas. Presenting Author: Ghazaleh Tabatabai,<br />

Department <strong>of</strong> Neurology, University Hospital Zurich, Zurich, Switzerland<br />

Background: Bevacizumab has been approved for the treatment <strong>of</strong> recurrent<br />

glioblastoma in various countries. The phase III registration trial assessing<br />

the addition <strong>of</strong> bevacizumab to standard chemoradiotherapy in newly<br />

diagnosed glioblastomas has completed recruitment. Salvage treatment <strong>of</strong><br />

glioblastoma patients after bevacizumab failure, however, is commonly not<br />

successful. In the present study, we aimed at characterizing histological<br />

and molecular characteristics associated with glioma recurrence after<br />

bevacizumab. Methods: Paired tissue samples from primary and recurrent<br />

tumors obtained from 12 patients with glioblastoma (n�11) or anaplastic<br />

astrocytoma (n�1) before and after bevacizumab treatment, and 14<br />

non-bevacizumab-treated patients as a reference group, were analyzed for<br />

histological features as well as molecular markers. <strong>Clinical</strong> records and<br />

magnetic resonance images were reviewed. Results: Histologically, recurrent<br />

tumors after bevacizumab showed more commonly a decreased blood<br />

vessel density and reduced glomeroloid microvascular proliferations when<br />

compared to recurrent tumors after conventional radiochemotherapy. In<br />

contrast, tumor cellularity and proliferation rate were similar. Distant<br />

relapse was more common in bevacizumab-treated patients. Molecular<br />

genetic studies <strong>of</strong> the respective tissue specimens are currently under way.<br />

Conclusions: Histological and molecular analyses <strong>of</strong> tumor tissue samples<br />

from glioma patients before and after bevacizumab treatment, combined<br />

with thorough clinical and radiological correlates, may provide hints for<br />

therapy escape mechanisms that can then be validated in preclinical<br />

models.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


132s Central Nervous System Tumors<br />

2069 General Poster Session (Board #15H), Sat, 1:15 PM-5:15 PM<br />

Effect <strong>of</strong> electrochemotherapy with IV bleomycin on complete response in a<br />

preclinical brain tumor study. Presenting Author: Birgit Agerholm-Larsen,<br />

Department <strong>of</strong> Oncology, C*EDGE at Glostrup Research Institute, Glostrup,<br />

Denmark<br />

Background: Electrochemotherapy (ECT) describes enhanced chemotherapeutic<br />

drug uptake after cell membranes have been made transiently<br />

permeable due to application <strong>of</strong> an electric field to the tumor. ECT is<br />

routinely used to treat skin metastases, with high response rates after<br />

once-only treatment. We have developed an electrode for use in the brain,<br />

both a rodent model and an electrode for clinical use. The aim <strong>of</strong> the<br />

present study was to evaluate ECT using intravenous (iv) bleomycin in a rat<br />

brain tumor model. Methods: Sprague Dawley male rats (7-11 week old)<br />

were inoculated with rat glial cell derived tumor cells (N32) through a burr<br />

hole in the skull. When tumors appeared on MRI, animals were allocated to<br />

ECT (iv bleomycin and local electric pulses), iv bleomycin only, iv<br />

bleomycin with placement <strong>of</strong> electrodes (no pulses), or no treatment.<br />

Bleomycin was injected iv in the tail vein (600�L, 3 IU/�L) and electrode<br />

deployment was made through the burr hole into the brain tissue.<br />

Electrochemotherapy parameters were 32 pulses, 100V, 100�s, 1Hz.<br />

Tumor size was determined based on contrast enhanced area from MR<br />

scans. Kaplan-Meyer events were defined as termination due to extensive<br />

tumor progression prior to end <strong>of</strong> the three independent experiments<br />

performed. Immuno-histo-pathology was performed after termination to<br />

verify MRI findings. Results: In 88% <strong>of</strong> the animals (14 <strong>of</strong> 16) treated with<br />

electrochemotherapy we found complete response (no tumor), validated by<br />

MRI and immuno-histo-pathology, whereas bleomycin only, bleomycin and<br />

electrodes, and no treatment showed progression in 11 out <strong>of</strong> 13 control<br />

animals. A Kaplan-Meier plot showed a pronounced improved survival for<br />

the ECT group as compared to controls within 3 weeks from treatment<br />

(p�0.001). Treatment was well tolerated. Conclusions: The present data<br />

suggest that electrochemotherapy with iv injection <strong>of</strong> bleomycin is a new<br />

promising treatment for brain tumors. In a clinical study using iv bleomycin<br />

and electrochemotherapy for brain metastases (www.clinicaltrials.gov,<br />

ID:NCT01322100) the first patient has just been treated successfully with<br />

ECT, indicating feasibility <strong>of</strong> the approach in the clinical setting.<br />

2071 General Poster Session (Board #16B), Sat, 1:15 PM-5:15 PM<br />

Health-related quality <strong>of</strong> life (HRQOL) in patients with glioblastoma (GBM)<br />

and their caregivers in the end-<strong>of</strong>-life phase: A retrospective study.<br />

Presenting Author: Birgit Flechl, Medical University <strong>of</strong> Vienna, Vienna,<br />

Austria<br />

Background: Glioblastoma multiforme (GBM) still harbours an inevitably<br />

fatal prognosis. The specially course <strong>of</strong> this disease poses unique challenges<br />

in care provision to the relatives. We still lack data about the<br />

end-<strong>of</strong>-life phase <strong>of</strong> GBM patients to improve counseling and supporting<br />

GBM patients and their proxies. Methods: In this retrospecitve study we<br />

included 52 caregivers <strong>of</strong> deceased GBM patients treated in two hospitals<br />

in Vienna, Austria. We used a specially developed questionnaire by the<br />

Medical University <strong>of</strong> Amsterdam to explore and document the last three<br />

months <strong>of</strong> living <strong>of</strong> GBM patients. Results: Most <strong>of</strong> the included caregivers<br />

were the partners <strong>of</strong> the patients (88%) and two thirds were female. The<br />

most common symptom in GBM patients was fatigue (87%), followed by<br />

reduced consciousness (81%) and aphasia (77%). 22% <strong>of</strong> the patients<br />

were bedbound during their last three months increasing to 80% in the last<br />

week <strong>of</strong> life. 30% <strong>of</strong> the caregivers told that they felt incompletely informed<br />

for their task and about the illness <strong>of</strong> their loved one. They stated the quality<br />

<strong>of</strong> life (QOL) <strong>of</strong> the patients with 2.2 and their own with 2.8 on a scale <strong>of</strong> 1<br />

to 7 whereas 7 displays the best possible answer. The majority <strong>of</strong> the<br />

patients (46%) died in hospitals and 38% at home, which was the most<br />

<strong>of</strong>ten expressed wish for place <strong>of</strong> death (45%)by patients. Regarding the<br />

caregivers´ symptoms, sadness (90%), fear (69%), burnout (60%), less<br />

interest in others (54%) and irritation (42%) were the leading ones and did<br />

not differ significantly in-between the places <strong>of</strong> death. Conclusions: The<br />

end-<strong>of</strong>-life phase <strong>of</strong> GBM patients is different from that <strong>of</strong> patients dying<br />

from other cancers. Most alarmingly, one thirds <strong>of</strong> their caregivers feels<br />

poorly informed. About two thirds <strong>of</strong> the caregivers fell overstrained and<br />

stresses thereby the urgent need for support and dedicated educational<br />

programs.<br />

2070 General Poster Session (Board #16A), Sat, 1:15 PM-5:15 PM<br />

Prospective follow-up <strong>of</strong> a cohort <strong>of</strong> 112 patients with leptomeningeal<br />

metastases <strong>of</strong> breast cancer recruited from 2007 to 2011: Prognostic<br />

factors. Presenting Author: Fahed Zairi, CHRU, Lille, France<br />

Background: Around 5% <strong>of</strong> patients with breast cancers (BC) will develop<br />

leptomeningeal metastasis (LM). The incidence may increase. Methods: We<br />

reported the description and outcome <strong>of</strong> 112 consecutive BC patients<br />

diagnosed with LM in our institution from 2007 to 2011. Correlations<br />

between characteristics and overall survival (OS) were analyzed using usual<br />

statistical methods (Kaplan Meier, Log-rank, Cox model). Results: BC were<br />

invasive ductal carcinoma in 69.7%. Estrogen and progesterone receptors<br />

were detected in respectively 61.6 and 44.6%. HER2 expression was<br />

observed in 26%. 23% were triple negative. Median time between BC<br />

diagnosis and LM diagnosis was 44 months. At LM diagnosis, median age<br />

was 54 and median Performance Status (PS) was 2. CSF cytology and<br />

cerebrospinal MRI were positive in respectively 72,5% and 87%. 103<br />

(92%) LM patients received IT liposomal cytarabine as 1st line <strong>of</strong><br />

treatment (ventricular device in 47%). IT therapy could be associated with<br />

systemic treatment in 58% <strong>of</strong> the cases and cerebral radiotherapy for LM in<br />

14% <strong>of</strong> the cases. <strong>Clinical</strong> response after 1st line treatment was observed<br />

in 57%, CSF response in 30,5%, MRI response in 62,5%. 24 patients<br />

received a 2nd line <strong>of</strong> IT thiotepa, 6 a 3rd line <strong>of</strong> IT methotrexate. The more<br />

significant prognostic factors (p�0,0001) were initial PS, associated<br />

systemic treatment and triple negative BC status. Other significant predictors<br />

<strong>of</strong> OS were thiotepa as 2nd line treatment (p�0,0004), intracranial<br />

hypertension at LM diagnosis (p�0,019), associated cerebral radiotherapy<br />

(p�0,02), progesterone receptor status (p�0,04). Median OS <strong>of</strong> the 103<br />

treated patients was 3,8 months (4,75 months for 0-2 PS and 1,6 months<br />

for 3-4 PS patients). Conclusions: Median OS was consistent those <strong>of</strong> other<br />

recent cohorts <strong>of</strong> BC LM. Our results confirm the role <strong>of</strong> a very early<br />

diagnosis, before the degradation <strong>of</strong> the general status. The association<br />

with systemic treatment or cerebral radiotherapy is indicated when possible.<br />

2072 General Poster Session (Board #16C), Sat, 1:15 PM-5:15 PM<br />

Pediatric glioblastoma: Results with adjuvant chemoradiation using temozolomide.<br />

Presenting Author: Nikhil Purushottam Joshi, All India Institute<br />

<strong>of</strong> Medical Sciences, New Delhi, India<br />

Background: Paediatric glioblastoma patients are underrepresented in<br />

major trials for this disease. The value <strong>of</strong> concurrent and adjuvant<br />

temozolomide is not known in this subset <strong>of</strong> patients. Methods: We<br />

retrospectively analysed our database between 2004 and 2010. All<br />

patients were treated with maximally safe surgical resection. This was<br />

followed by a uniform treatment schedule <strong>of</strong> post-operative radiation<br />

according to RTOG guidelines with concurrent daily temozolomide at 75<br />

mg per meter square. The radiation dose was 60 Gy in 30 fractions planned<br />

by 3 dimensional conformal radiotherapy. 4 weeks later, adjuvant temozolomide<br />

was started at 150 mg per meter square, day 1 to 5 every 28 days<br />

and escalated to 200 mg per meter square if well tolerated. Log-rank test<br />

was used to compare survival distribution. The data was analysed using<br />

SPSS 16. Results: 23 patients were analysed. The median age was 11 years<br />

(range: 5 to 19 years). 15 males and 8 females were noted. 4 patients had<br />

seizures at presentation while 19 did not. 13 patients underwent a gross<br />

total resection while 10 underwent a subtotal resection. 5 patients received<br />

only concurrent temozolomide while 18 received concurrent and adjuvant<br />

temozolomide. The median number <strong>of</strong> adjuvant temozolomide cycles were<br />

6 (range 2 to 12). The median follow up was 11.9 months (range: 1.6 to<br />

30.1 months). 6 patients were dead and 17 were alive at the time <strong>of</strong><br />

analysis. The median overall survival was not reached. The median survival<br />

for patients receiving only concurrent temozolomide was 13 months while<br />

that for patients receiving concurrent and adjuvant temozolomide was not<br />

reached and the difference was statistically significant (p�0.0009). The<br />

overall survival was not statistically different for age less or more than 10<br />

years, gender, and type <strong>of</strong> surgery or the number <strong>of</strong> adjuvant cycles <strong>of</strong><br />

temozolomide (less or more than 6). Conclusions: Temozolomide may have<br />

a role in paediatric Glioblastoma. Both concurrent and adjuvant temozolomide<br />

seem to be important for superior overall survival in this group <strong>of</strong><br />

patients.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


2073 General Poster Session (Board #16D), Sat, 1:15 PM-5:15 PM<br />

Bevacizumab use and central nervous system (CNS) hemorrhage. Presenting<br />

Author: Nathalie LeTarte, Centre Hospitalier de l’université de Montreal<br />

(CHUM), Montreal, QC, Canada<br />

Background: Bevacizumab is widely used and may cause life-threatening<br />

bleeding, usually at sites <strong>of</strong> disease involvement such as the CNS. We<br />

attempted to identify clinical characteristics associated with CNS hemorrhage<br />

in a broad population. Methods: We did a retrospective review <strong>of</strong> the<br />

FDA Medwatch database <strong>of</strong> adverse events reported with bevacizumab from<br />

11/1997 to 5/2009. Results: We searched the database for keywords<br />

bleeding, hemorrhage, cerebral, intracranial, subarachnoid, cerebellar,<br />

hemorrhagic stroke, and brain. 17,466 reports were included in the<br />

database: 154 described CNS hemorrhage in 99 patients, and 1041<br />

reports described non-CNS bleeds. For CNS bleeds, cerebral hemorrhage<br />

was the most frequent event reported (n�39), followed by intracranial<br />

hemorrhage (n�20) and subarachnoid hemorrhage (n�18). Median age<br />

was 62 years; 54% <strong>of</strong> patients were female. Primary cancer was colorectal<br />

(42%), glioma (13%), and breast cancer (10%). Patients received a<br />

median <strong>of</strong> three (1-36) doses <strong>of</strong> bevacizumab prior to the bleed. 54% <strong>of</strong><br />

patients received myelosuppressive chemotherapy, and 30% had documented<br />

history <strong>of</strong> hypertension. Sixteen patients with CNS hemorrhage<br />

were reported to have CNS metastases. For 70 patients, information on<br />

CNS involvement was not reported. Death was reported as a complication <strong>of</strong><br />

hemorrhage in 48% <strong>of</strong> patients. Nine patients with brain metastases died<br />

and CNS hemorrhage was the cause <strong>of</strong> death in seven. Six patients with<br />

glioma died, and CNS hemorrhage was the cause in three. One patient with<br />

brain metastases, and one patient with glioma also experienced a non-CNS<br />

bleed. Five patients in each group had received heparin, warfarin or<br />

NSAIDs. Low platelet counts were reported in 3 patients with CNS<br />

metastases and 2 patients with glioma. The most common factor associated<br />

with CNS hemorrhage was medication predisposing to bleeding,<br />

followed by thrombocytopenia. Hypertension, a risk factor for CNS hemorrhage,<br />

was reported in 4 patients with brain metastases, and 2 patients with<br />

glioma. Conclusions: In this database, 154 <strong>of</strong> 1195 reports <strong>of</strong> bleeding<br />

associated with bevacizumab described a CNS bleed. Although CNS bleeds<br />

were not common, they were the reported cause <strong>of</strong> death in more than half<br />

<strong>of</strong> the cases.<br />

2075 General Poster Session (Board #16F), Sat, 1:15 PM-5:15 PM<br />

Phase II trial <strong>of</strong> bevacizumab and fotemustine in recurrent grade III<br />

gliomas. Presenting Author: Riccardo S<strong>of</strong>fietti, Division <strong>of</strong> Neuro-Oncology,<br />

University Hospital San Giovanni Battista, Turin, Italy<br />

Background: Bevacizumab (BV) has shown activity in recurrent grade III<br />

gliomas, and few data are available on the combination <strong>of</strong> BV and<br />

nitrosoureas. Fotemustine (FTM) is a nitrosourea with elevated lipophilic<br />

properties. Methods: In this phase II study patients with grade III gliomas<br />

recurrent after surgery, radiation therapy and concomitant/adjuvant temozolomide<br />

were eligible. The treatment consisted <strong>of</strong> an induction phase with<br />

BV at 10mg/kg intravenously on day 1 and 15 and FTM at 75mg/m2<br />

intravenously on day 1 and 8, followed after a 3 week interval by a<br />

maintenance phase with BV 10mg/kg and FTM 75mg/m2 every 3 weeks<br />

until tumor progression or unacceptable toxicity. Patients had undergone<br />

clinical and MRI assessment 1 month after the start <strong>of</strong> treatment and<br />

thereafter every 2 months. The primary endpoint was progression-free<br />

survival at 6 months (PFS-6), whereas secondary end-points were response<br />

rate (RR), based on RANO criteria, progression-free survival (PFS), overall<br />

survival (OS), and safety. Results: From May 2008 to September 2011, 32<br />

patients (males 23, females 9, median age 46) were enrolled. PFS-6, PFS<br />

-12 and median PFS were 31%, 7% and 5 months (range: 1-43�),<br />

respectively. OS-6, OS-12 and median OS were 78%, 37.8% and 8.6<br />

months (3.5-43�), respectively. Response rates were as follows: 4 CR<br />

(12.5%), 12 PR (37.5%), 14 SD (44%) and 2 PD (6%). A significant<br />

neurological improvement was observed in 48% <strong>of</strong> symptomatic patients,<br />

being steroids reduced or withdrawn in 83% <strong>of</strong> patients. 29/32 (91%)<br />

patients have progressed and patterns <strong>of</strong> tumor progression were local in 20<br />

(69%), multicentric in previous multicentrinc tumors in 5 (17%), gliomatosis-like<br />

in 3 (10%) and local plus leptomeningeal seeding in 1 (4%). 23/29<br />

patients had salvage treatment after failure. Toxicities after BV were as<br />

expected; 5 patients interrupted FTM for grade III-IV myelotoxicity. We did<br />

not observe any additional toxicity from the combination. Conclusions: The<br />

combination <strong>of</strong> bevacizumab and fotemustine in grade III gliomas recurrent<br />

after standard treatment seems to have some activity.<br />

Central Nervous System Tumors<br />

133s<br />

2074^ General Poster Session (Board #16E), Sat, 1:15 PM-5:15 PM<br />

Phase II study to evaluate the efficacy and safety <strong>of</strong> rilotumumab and<br />

bevacizumab (BEV) in subjects with recurrent malignant glioma (MG).<br />

Presenting Author: Mary Lou Affronti, Duke University Medical Center,<br />

Durham, NC<br />

Background: Prognosis <strong>of</strong> recurrent MG remains poor with a median survival<br />

<strong>of</strong> 3-9 months. Few evidence based treatments are available and response<br />

rates have been � 20% with 6 month progression–free survival (PFS) <strong>of</strong> �<br />

9-16%. Poor prognosis is linked to the MG’s ability to induce vascular<br />

proliferation and invasion. MG’s have an abundance <strong>of</strong> neo-vascularization<br />

and high levels <strong>of</strong> vascular endothelial growth factor (VEGF). BEV, an<br />

antibody to VEGF, is the most active agent tested for recurrent MG with an<br />

overall response rate <strong>of</strong> 21.2%. Rilotumumab is a human monoclonal<br />

antibody that blocks human hepatocyte growth factor (HGF) and its<br />

receptor c-Met. HGF/c-Met signaling plays a role in MG tumorigenicity<br />

inducing angiogenesis and expression <strong>of</strong> additional angiogenic autocrine<br />

factors such as VEGF. The novel combination <strong>of</strong> rilotumumab (C-Met<br />

inhibitor) with an anti-VEGF drug (BEV) to block vascular invasion and<br />

tumor proliferation may demonstrate synergistic tumor growth inhibition.<br />

Methods: A two-stage design is used to assess radiographic response rate<br />

with � 3 responses required for the first stage. Secondary endpoints were<br />

PFS, overall survival (OS) and toxicity. Subjects with recurrent BEV-naïve<br />

MGs received rilotumumab at 20 mg/kg in combination with BEV at 10<br />

mg/kg every two weeks � 28 days after the last surgical procedure. A brain<br />

MRI was obtained after each 6-week cycle. Results: 15 <strong>of</strong> the 19 patients<br />

required for the first stage have been accrued. 10(67%) were male and �<br />

50 years. 73% received � 2 prior regimens and 46% had � 2 prior<br />

progressions. Best tumor response included 1(7%) complete response, 2<br />

(13%) partial responses, 8 (53%) stable disease, 1 (7%) progressive<br />

disease with 20% (3/15) non-evaluable due to early termination secondary<br />

to toxicity. Median PFS was 3.9 months (95% CI: 1, 7.1) with a 6-month<br />

PFS <strong>of</strong> 25.4% (95% CI: 6.4, 50.5) and 6-month OS <strong>of</strong> 68.3% (95% CI:<br />

34, 87.1). Toxicities are listed below. Maximum average weight increase<br />

over all cycles was 6.3 % (SD � 6.09). Conclusions: Rilotumumab in<br />

combination with BEV is an active regimen in MG. Toxicity will need to be<br />

closely monitored.<br />

Toxicity Grade 3 Grade 4 Grade 5<br />

Prolonged QTc interval 1 (7%)<br />

Hypotension 1 (7%)<br />

Hypokalemia 1 (7%)<br />

DVT/PE 1 (7%) 2 (13%) 1 (7%)<br />

2076 General Poster Session (Board #16G), Sat, 1:15 PM-5:15 PM<br />

Adult low-grade gliomas: The Cleveland Clinic experience. Presenting<br />

Author: Lizbeth Robles Irizarry, Cleveland Clinic, Cleveland, OH<br />

Background: Low-grade gliomas account for 10-20% <strong>of</strong> all primary brain<br />

tumors. There is limited data on specific prognostic factors for patients with<br />

low-grade gliomas. Methods: After obtaining IRB approval, the Cleveland<br />

Clinic Brain Tumor and Neuro-Oncology Center’s database was used to<br />

identify patients with histologically confirmed grade 2 glioma at the time <strong>of</strong><br />

diagnosis. Multivariable analysis was conducted with use <strong>of</strong> a Cox proportional<br />

hazards model and a stepwise selection algorithm that used p�0.10<br />

as the criteria for entry and p�0.05 as retention in the model to identify<br />

independent predictors <strong>of</strong> survival. Results: Chart records <strong>of</strong> 478 patients<br />

(54% <strong>of</strong> whom were men) diagnosed between 1991 and 2010 were<br />

included for analysis. The median age at presentation was 41 years (range,<br />

18-83 years). 42% <strong>of</strong> patients had biopsy only, 27% had gross total<br />

resection, 6% had near total resection, and 25% had subtotal resection.<br />

22% <strong>of</strong> patients were initially treated with radiation, 30% with adjuvant<br />

chemotherapy, and 7% with concurrent chemotherapy. Median progression<br />

free survival and median overall survival (OS) were 5.1 years and 12.8<br />

years, respectively. On multivariate analysis, independent <strong>of</strong> additional<br />

therapy, five factors were identified as independent predictors <strong>of</strong> OS: age at<br />

diagnosis (p�0.002), Karn<strong>of</strong>sky performance status (KPS, p�0.0001),<br />

histology (astrocytoma vs. other, p�0.001), hemispheric location (left or<br />

bilateral involvement vs. right, p�0.02) and gender (male vs. female,<br />

p�0.0003). Conclusions: Older age, male gender, poor performance<br />

status, astrocytic histology, and left hemispheric or bilateral involvement<br />

are associated with higher mortality.<br />

Risk factors for mortality: Multivariable Cox proportional hazards analysis.<br />

Factor1 Hazard ratio (95% C.I.) p2 KPS (55 vs. 50-55 vs.


134s Central Nervous System Tumors<br />

2077 General Poster Session (Board #16H), Sat, 1:15 PM-5:15 PM<br />

Utility <strong>of</strong> iron oxide nanoparticles in the radiographic diagnosis <strong>of</strong> CNS<br />

lymphoma and inflammatory diseases. Presenting Author: Brian T. Farrell,<br />

Oregon Health & Science University, Portland, OR<br />

Background: Ferumoxytol, an ultrasmall superparamagnetic iron oxide<br />

nanoparticle (USPIO), is approved for intravenous treatment <strong>of</strong> iron<br />

deficiency in chronic renal failure. The package insert warns <strong>of</strong> potential<br />

alterations <strong>of</strong> magnetic resonance imaging (MRI). Our initial animal studies<br />

provided evidence <strong>of</strong> USPIO uptake by macrophages and lymphoreticular<br />

cells suggesting its potential in CNS lymphoma (CNSL), post transplant<br />

lymphoproliferative disease, or CNS inflammation (i.e. multiple sclerosis).<br />

The mechanism <strong>of</strong> USPIO enhancement differs from gadolinium-based<br />

contrast agents (GBCA), which optimally image areas <strong>of</strong> increased vascular<br />

permeability. Methods: USPIO was administered as an MRI contrast agent<br />

to 20 patients with presumptive or de facto diagnosis <strong>of</strong> CNSL or CNS<br />

inflammation. Seventeen <strong>of</strong> 20 patients underwent both USPIO- and<br />

gadolinium-based MRI. Three patients received only USPIO due to risk <strong>of</strong><br />

nephrogenic systemic fibrosis (NSF) with GBCA exposure. In the pilot<br />

phase <strong>of</strong> the study, patients were given the USPIO ferumoxtran-10 and<br />

subsequent patients were given a successor USPIO (ferumoxytol) which<br />

has improved pharmaceutical properties. All patients were monitored for<br />

adverse reactions. MRI scans were subsequently reviewed by a neuroradiologist.<br />

Results: No significant toxicities were seen. Thirteen <strong>of</strong> 17 patients<br />

who received both contrast agents showed equal numbers <strong>of</strong> enhancing<br />

lesions on postcontrast images. USPIO-based contrast revealed additional<br />

areas <strong>of</strong> enhancement in three <strong>of</strong> 17 patients that were not visible when<br />

GBCA was administered. In three patients more lesions were seen with<br />

GBCA. Examples <strong>of</strong> ferumoxytol utility include 1) improved targeting for<br />

surgical biopsy; 2) avoidance <strong>of</strong> NSF; 3) demonstrating enhancement<br />

related to macrophages within lesions; 4) correlating cerebral blood volume<br />

and CNS inflammation; 5) demonstrating pitfalls such as prolonged<br />

imaging changes (3 months) in one case due to intensive inflammation,<br />

which can simulate hemorrhage. Conclusions: These studies provide data<br />

about the utility and value <strong>of</strong> USPIO-based contrast imaging in CNSL and<br />

inflammatory CNS lesions that support a dual role for these agents beyond<br />

iron replacement.<br />

2079 General Poster Session (Board #17B), Sat, 1:15 PM-5:15 PM<br />

Tegwondo-CCNU: A novel combination <strong>of</strong> protracted, near-continuous<br />

temozolomide and CCNU with activity in recurrent malignant gliomas.<br />

Presenting Author: Herwig Matthias Strik, University <strong>of</strong> Marburg, Marburg,<br />

Germany<br />

Background: Recent data have been raising doubt if dose-dense temozolomide<br />

is more efficient against malignant gliomas than with conventional<br />

dosing. We report here updated results <strong>of</strong> combined near-continuous<br />

temozolomide, aimed at depleting anti-alkylating MGMT, with once weekly<br />

application <strong>of</strong> low-dose lomustine (CCNU) at first or second relapse <strong>of</strong><br />

malignant gliomas. Methods: 25 consecutive patients with recurrent<br />

malignant gliomas (12 anaplastic gliomas WHO III, 13 glioblastomas or<br />

gliosarkomas WHO IV) were treated: 15 males (60%), 10 females (40%);<br />

mean age at start <strong>of</strong> chemotherapy was 52 (20-78) years; Eight patients<br />

were treated at first, 15 at second and two at third recurrence. All patients<br />

were pretreated with temozolomide. Four patients received fractionated,<br />

stereotactically guided re-irradiation (FSRT). The treatment regimen consisted<br />

<strong>of</strong> near-continuous temozolomide 50mg/m2 day 1-5/7 and low-dose<br />

CCNU 40mg absolute dose day 6/7. In cases <strong>of</strong> bone-marrow depression,<br />

temozolomide was reduced in steps <strong>of</strong> 20mg total dose or – in more severe<br />

cases – chemotherapy was interrupted until normalization <strong>of</strong> blood counts.<br />

Results: In total, 89 cycles (months) <strong>of</strong> chemotherapy were applied. The<br />

combination was well tolerated in terms <strong>of</strong> nausea and fatigue. Blood<br />

counts usually decreased continuously, enabling a gradual dose adaptation.<br />

Hematological WHO grade 3�4 toxicity occurred in 5/25 patients<br />

(20%), two <strong>of</strong> them were symptomatic. Three patients had prolonged<br />

elevation <strong>of</strong> liver enzymes. Best responses after � 3 months (23 patients)<br />

were: 2 complete and 1 partial remissions (13%, ), 13 stable diseases<br />

(57%), and 7 progressive diseases (30%). Progression-free survival at six<br />

months from start <strong>of</strong> chemotherapy <strong>of</strong> the 16 patients treated � 6 months<br />

or deceased (PFS 6) was 37%, median overall survival 6.3 months.<br />

Conclusions: Although some hematotoxicity was observed, the regimen<br />

presented here is well tolerated and safe if carefully controlled. The<br />

objective responses and considerable rate <strong>of</strong> stable diseases indicate that<br />

this combination is active in malignant gliomas after pretreatment with<br />

temozolomide alone. The results have to be controlled in a prospective trial.<br />

2078 General Poster Session (Board #17A), Sat, 1:15 PM-5:15 PM<br />

MRI-based vessel function maps for high-resolution differentiation between<br />

tumor and nontumoral tissues in brain tumor patients. Presenting<br />

Author: Leor Zach, Sheba Medical Center, Ramat-Gan, Israel<br />

Background: Changes in enhancement seen in post treatment brain MRIs in<br />

primary and secondary brain malignancies <strong>of</strong>ten mimic tumor progression<br />

(pseudoprogression, radiation necrosis). Conventional MRI cannot differentiate<br />

tumor progression from treatment related effects resulting in suboptimal<br />

patient management. Methods: The application <strong>of</strong> delayed contrast<br />

extravasation MRI for depicting unique vessels characteristics with high<br />

resolution and high sensitivity to subtle BBB disruption is demonstrated in<br />

17 GBM and 15 brain metastases patients undergoing standard chemoradiation<br />

and radiosurgery respectively. Results: 2 primary vessel function<br />

populations were defined: a slow population where contrast clearance from<br />

the tissue was slower than accumulation, and a fast population where<br />

clearance was faster than accumulation. Ten stereotactic biopsies acquired<br />

from GBM patients showed complete correlation with the maps, confirming<br />

the discrimination between fast population regions, reflecting morphological<br />

active tumor and slow regions reflecting necrosis/treatment-induced<br />

changes. Typical fast population vessel morphology consisted <strong>of</strong> proliferating<br />

endothelial cells, dilated lumen, peri vascular fibrosis and glumeroloid<br />

vessels, while slow regions consisted <strong>of</strong> necrotic vessels, in agreement with<br />

the observed MRI vessel function. The fast component volume 3 weeks post<br />

treatment was significantly correlated with the fast volume 4 (r2�0.84, p�0.0005) and 6 months (r2�0.84, p�0.01) later, suggesting early<br />

prediction <strong>of</strong> response. Brain metastases histology showed similar results:<br />

5 brain metastases with a fast population component were confirmed<br />

histologically to contain morphologically active tumor. One metastasis<br />

consisting <strong>of</strong> the slow population only significantly decreased in volume in<br />

the following MRIs. Conclusions: These results demonstrate the feasibility<br />

<strong>of</strong> applying our delayed contrast extravasation high resolution function<br />

vessel maps for improving patient management by clearly depicting tumor<br />

and non-tumoral tissues in patients with primary and secondary brain<br />

malignancies undergoing standard chemoradiation or radiosurgery.<br />

2080 General Poster Session (Board #17C), Sat, 1:15 PM-5:15 PM<br />

Retrospective analysis <strong>of</strong> glioblastoma patients treated with bevacizumab<br />

who presented with multifocal disease at diagnosis. Presenting Author:<br />

Jethro Lisien Hu, Cedars-Sinai Medical Center, Los Angeles, CA<br />

Background: Glioblastoma patients who present with multifocal disease<br />

have an extremely poor prognosis – a recent analysis at our institution<br />

demonstrated a median survival <strong>of</strong> 6 months, versus 11 months for a cohort<br />

<strong>of</strong> unifocal glioblastoma patients matched for age, extent <strong>of</strong> resection, and<br />

KPS. Bevacizumab is frequently used to treat patients with recurrent or<br />

progressive glioblastoma. However, its benefit in the setting <strong>of</strong> multifocal<br />

disease is unproven, particularly in light <strong>of</strong> the concern that antiangiogenic<br />

treatment may promote a more invasive, multifocal, and potentially<br />

treatment-resistant phenotype. Methods: Between 2004 to 2010, 368<br />

patients were diagnosed with glioblastoma at our institution. We identified<br />

46 patients with multifocal disease on initial presentation, and retrospectively<br />

reviewed their clinical course and treatment history. Kaplan-Meier<br />

estimates and Wilcoxon tests were used to assess survival distribution.<br />

Results: Of the 46 patients with multifocal disease, 12 were treated with<br />

bevacizumab (7 upfront and 5 at first recurrence). All 12 bevacizumabtreated<br />

patients received external beam radiation therapy, and 11 received<br />

temozolomide. In the bevacizumab-treated cohort, median age at the time<br />

<strong>of</strong> surgery was 59 years, and median KPS was 80. For the 34 patients with<br />

multifocal disease who did not receive bevacizumab, median age was 69<br />

years, and median KPS was 75. Median survival for the bevacizumabtreated<br />

patients was 12.9 months, with 12-month survival <strong>of</strong> 58.3%. By<br />

comparison, median survival for patients with multifocal disease who did<br />

not receive bevacizumab was 5.8 months, with 12-month survival <strong>of</strong><br />

20.6%. The difference in survival between the two groups was statistically<br />

significant, with p-value <strong>of</strong> 0.045 by the Wilcoxon test. Conclusions: This<br />

single-institution retrospective analysis suggests that bevacizumab treatment<br />

is associated with a survival benefit for glioblastoma patients who<br />

present with multifocal disease. One can infer that the clinical benefit <strong>of</strong><br />

bevacizumab in this setting outweighs any potential concerns regarding the<br />

ability <strong>of</strong> bevacizumab to promote an invasive, treatment-resistant tumor<br />

phenotype.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


2081 General Poster Session (Board #17D), Sat, 1:15 PM-5:15 PM<br />

The impact <strong>of</strong> bevacizumab on the occurrence and recurrence <strong>of</strong> intracranial<br />

brain metastases in non-small cell lung cancer (NSCLC) patients.<br />

Presenting Author: Mohamed E. Salem, Karmanos Cancer Institute, Wayne<br />

State University School <strong>of</strong> Medicine, Dertoit, MI<br />

Background: Patients (pts) with brain metastases (BM) have a poor<br />

prognosis and limited treatment options. Approximately 40-50% <strong>of</strong> pts<br />

with NSCLC will develop BM during the course <strong>of</strong> their disease. A better<br />

understanding <strong>of</strong> the pathobiology <strong>of</strong> BM and the development <strong>of</strong> targeted<br />

agents hold promise for improved prophylaxis and therapy <strong>of</strong> BM which<br />

could have significant impact on disease progression and pt’s survival.<br />

Vascular Endothelial Growth Factor has been implicated in setting up a<br />

metastatic niche allowing cells to seed and grow in the brain parenchyma.<br />

Inhibition <strong>of</strong> this signaling has been studied and shown to be effective in<br />

reducing BM in animal models. Methods: The objective <strong>of</strong> this retrospective<br />

study was to determine whether bevacizumab decreases the incidence and<br />

or the recurrence <strong>of</strong> BM in pts with NSCLC. We retrospectively identified<br />

NSCLC pts with and without BM treated with bevacizumab. The primary<br />

endpoint was the proportion <strong>of</strong> patients who developed BM on or after<br />

bevacizumab therapy. Secondary endpoints were rate <strong>of</strong> local or regional<br />

recurrence after stereotactic radiosurgery (SRS). Occurrence <strong>of</strong> BM was<br />

evaluated by retrospectively reviewing MRI and or CT scans. Results: A total<br />

<strong>of</strong> 30 pts with stage IV non-squamous NSCLC who had no BM at diagnosis<br />

treated with bevacizumab (Non-BM group, NBMG) and 85 pts with<br />

non-squamous NSCLC who had BM and underwent SRS (BM group, BMG)<br />

were studied (46%M; median age 60 years (range 32-84). In the NBMG,<br />

28% <strong>of</strong> pts developed BM. Median time to development <strong>of</strong> BM was 10.5<br />

months. In the BMG, 4 pts were treated with bevacizumab following SRS.<br />

Of these 4 pts, 3 pts (75%) had no recurrence while 1 pt (25%) developed<br />

local then regional recurrence at 7 and 9 months, respectively. Of the<br />

remaining 81 pts who underwent SRS but never received bevacizumab,<br />

27% <strong>of</strong> pts had local recurrence and 41% had regional recurrence at<br />

median time <strong>of</strong> 4 and 4.5 months, respectively. Conclusions: Bevacizumab<br />

may have a role in the treatment <strong>of</strong> NSCLC in regards to development and<br />

recurrence <strong>of</strong> brain metastases. These results support the need for further<br />

prospective investigation in a larger patient population with matched<br />

controls.<br />

2083 General Poster Session (Board #17F), Sat, 1:15 PM-5:15 PM<br />

Plasma levels <strong>of</strong> IL-8 and g-CSF in high-grade gliomas treated with<br />

bevacizumab. Presenting Author: Marica Eoli, Fondazione IRCCS Istituto<br />

Neurologico Carlo Besta, Milan, Italy<br />

Background: Bevacizumab, an anti-VEGF antibody, has shown significant<br />

activity in high grade gliomas (HGG). However, tumor recurrence inevitably<br />

occurs. Methods: We treated 63 recurrent HGG patients with poor prognostic<br />

factors with bevacizumab (10 mg/kg) and irinotecan (125 or 340<br />

mg/m2 ) every 2 weeks, and investigated IL-12, IL-13, IL-17, FGF basic,<br />

G-CSF, MIP-1b, PDGF-beta, plasma levels before starting treatment and<br />

every 8 weeks by Bioplex. Ten age- and sex-matched healthy controls were<br />

used for comparison. Results: After a median follow-up <strong>of</strong> 27 weeks,<br />

median OS and PFS were 33 and 18 weeks, respectively. PFS at 6 and 12<br />

months were 32% and 12%, respectively. OS at 6 months was 60%.<br />

Toxicity was mild. Baseline higher amounts <strong>of</strong> IL-13 (48�174 pg/ml vs<br />

3.44�0.9 pg/ml, p�0.0001) and lower amounts <strong>of</strong> MIP-1b (35.3�20.9<br />

pg/ml vs 67.2�18.8 pg/ml, p�0.0002), PDGF-beta (1585.5�1585<br />

pg/ml vs 7098�1585 pg/ml, p�0.0001) and VEGF (27�39.8 pg/ml vs<br />

54.5�32 pg/ml, p�0.001) were detected in patients than in healthy<br />

controls. In a cohort <strong>of</strong> 15 non-responders (patients who progressed 8<br />

weeks after treatment onset), baseline IL-8 (15.7�10.8 pg/ml vs 10.9�9.4<br />

pg/ml, p�0.03) and G-CSF (113.3�54 pg/ml vs 84.9�59.2 pg/ml,<br />

p�0.03) were significantly higher than in patients responding to treatment.<br />

In the same cohort no significant reduction <strong>of</strong> VEGF and other<br />

cytokines was observed after 8 weeks <strong>of</strong> treatment, while a decrease <strong>of</strong><br />

plasma VEGF was observed in the remaining patients (26�32 pg/ml vs<br />

13.3�28.5 pg/ml, p�0.001). Furthermore, in a cohort <strong>of</strong> 22 long-term<br />

responders (patients who progressed after more than 18 weeks <strong>of</strong> treatment),<br />

levels <strong>of</strong> VEGF decreased after 8 weeks <strong>of</strong> treatment when compared<br />

to baseline, whereas no difference was observed in baseline levels<br />

(23.9�22.6 pg/ml vs 9.8�9.4 pg/ml, p�0.001). Conclusions: Data<br />

suggest that high levels <strong>of</strong> IL-8 and G-CSF at baseline associated with a<br />

lack <strong>of</strong> VEGF decrease after 8 weeks <strong>of</strong> treatment identify patients who are<br />

resistant to bevacizumab. This hypothesis should be tested in a large<br />

number <strong>of</strong> patients.<br />

Central Nervous System Tumors<br />

135s<br />

2082 General Poster Session (Board #17E), Sat, 1:15 PM-5:15 PM<br />

Relationship between IDH1 mutation status and magnetic resonance<br />

imaging features in WHO grade II and III oligodendroglial tumors. Presenting<br />

Author: Frederique Toulgoat, Neuroradiologie CHU Nantes, Nantes,<br />

France<br />

Background: In gliomas, relationship between radiological characteristics<br />

and several biomarkers was the subject <strong>of</strong> numerous publications. Mutations<br />

in the isocitrate dehydrogenase 1 (IDH1) gene have been identified<br />

recently to play a key role in these tumors occuring in up to 75% <strong>of</strong><br />

low-grade diffuse (WHO grade II) and anaplastic (WHO grade III) astrocytic,<br />

oligodendroglial and mixed oligodendroglial neoplasms. However, the<br />

correlation with magnetic resonance imaging (MRI) features has been little<br />

studied. Methods: Patients treated for WHO grade II and III oligodendroglial<br />

tumors between 2005 and 2011 were retrospectively identified. Each case<br />

has been reviewed by the same neuropathologist. IDH1 and IDH2 mutations<br />

were available. Preoperative MRI, including T1 weighted, T2 weighted,<br />

T1 contrast enhanced, FLAIR, T2* weighted, diffusion weighted (ADC<br />

ratio), perfusion weighted (CBV ratio) and MR spectroscopy, were analyzed<br />

by two radiologists blinded from molecular data. Logistic regression<br />

analysis and Fisher’s test were used to develop predictive models <strong>of</strong> genetic<br />

pr<strong>of</strong>ile from imaging. Results: Sixty eight patients, WHO grade II (n� 37)<br />

and grade III (n�31) patients were identified. Mean age at diagnosis was<br />

46 years; ratio male/female was 40/28. IDH1 mutations were identified in<br />

42 patients (62 %), IDH2 in 4 patients (6 %). Analysis <strong>of</strong> tumor location,<br />

size, borders, morphological aspect, and signal did not shown any significant<br />

difference between IDH1 mutated group and IDH1 non mutated group<br />

neither in grade II nor in grade III oligodendroglial tumors. In the same way,<br />

MR spectroscopy (Choline/NAA ratio and detection <strong>of</strong> lipid and lactate) was<br />

not relevant to discern the two groups. As well, ADC ratio (1,5 versus 1,4;<br />

p�0,35) and CBV ratio (3,4 versus 4,2; p� 0,46) did not reveal any<br />

difference between mutated group and non mutated group. Conclusions: In<br />

our study, IDH1 mutations were not correlated with MRI features available<br />

during routine MRI. Nevertheless, recent studies suggest the ability <strong>of</strong> MR<br />

spectroscopy to detect 2-hydroxyglutarate as an MRI marker <strong>of</strong> IDH1<br />

mutated tumors, which encourage carrying on research in molecular<br />

imaging.<br />

2084 General Poster Session (Board #17G), Sat, 1:15 PM-5:15 PM<br />

The final report <strong>of</strong> a phase I trial <strong>of</strong> surgical resection with biodegradable<br />

carmustine (BCNU) wafer placement followed by vaccination with<br />

dendritic cells pulsed with tumor lysate for patients with glioblastoma.<br />

Presenting Author: Jeremy Rudnick, Neuro-Oncology Program, Cedars-<br />

Sinai Medical Center, Los Angeles, CA<br />

Background: Our prior immunotherapy trials demonstrated efficacy in<br />

generating a tumor specific immune response in malignant glioma and the<br />

potential for high tumor-specific toxicity and sustained tumoricidal activity.<br />

Immunotherapy may synergize with chemotherapy and biodegradable<br />

carmustine (BCNU) wafers and have a modest impact to extend overall<br />

survival. We exploited this synergistic effect to maintain a cytotoxic<br />

environment around the tumor milieu, and this is a presentation <strong>of</strong> the final<br />

results <strong>of</strong> our clinical trial. Methods: Patients with glioblastoma were<br />

eligible after maximal resection with biodegradable carmustine (BCNU)<br />

wafer placement. Screening leukapheresis was used to isolate mononuclear<br />

cells which were differentiated into dendritic cells, pulsed with tumor<br />

lysate, and then 3 intradermal vaccines administered at 2-week intervals.<br />

Patients continued systemic chemotherapy after vaccine or at progression.<br />

Results: Twenty three patient with glioblastoma received therapy including<br />

8 with newly diagnosed disease (35%) and 15 with recurrent disease<br />

(65%) were evaluable. Immune response data is available for 20/23<br />

patients although survival data is present for all. One grade 3 SAE <strong>of</strong> fever<br />

and chills was noted otherwise therapy was well tolerated. Within the newly<br />

diagnosed GBM cohort the median overall survival (OS) was 25.5 months<br />

(15,31�), and within the recurrent GBM cohort, the median OS was 16<br />

months (8,23�). Among the recurrent GBM an increase <strong>of</strong> �1.5 X baseline<br />

interferon gamma production post vaccination was associated with a<br />

prolonged median OS 22 months (8,40) in 4/12 patients versus 17 months<br />

(9,27) in 8/12 patients. Conclusions: We were able to generate an immune<br />

response in 25% <strong>of</strong> patients which is lower than what we have seen in<br />

previous trials and suggests a limited synergy with local control. However,<br />

within the recurrent GBM cohort we did find prolonged survival in both<br />

groups with an increased survival noted in immune responders demonstrating<br />

the potential for this therapy.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


136s Central Nervous System Tumors<br />

2085 General Poster Session (Board #17H), Sat, 1:15 PM-5:15 PM<br />

The role <strong>of</strong> fluorescence-guided surgical resection in the combined treatment<br />

<strong>of</strong> malignant glioma. Presenting Author: Carmine Maria Carapella,<br />

Neurosurgery, Cancer Institute Regina Elena, Roma, Italy<br />

Background: Malignant glioma represents a relevant therapeutic issue and<br />

the value <strong>of</strong> extensive surgical resection remains debated; recent evidence<br />

suggests that radical removal is associated with better survival. An<br />

interesting tool for identifying tumor tissue and increasing the extent <strong>of</strong><br />

surgery is represented by fluorescence-guided resection, taking advantage<br />

<strong>of</strong> metabolic and structural changes induced by a natural precursor <strong>of</strong> heme<br />

biosynthetic pathway, 5-amino-levulinic acid (ALA). Methods: The present<br />

experience is related to 32 patients affected by malignant glioma (18 newly<br />

diagnosed: 16 glioblastoma (GBM), 2 anaplastic oligodendroglioma; and<br />

14 recurrent GBM) eligible for fluorescence-guided resection, operated on<br />

in our Institute since fall <strong>of</strong> 2009. All patients underwent preoperative and<br />

early postoperative MRI, showing contrast enhancing lesions. An oral dose<br />

<strong>of</strong> 20 mg 5-ALA /kg bw was administered to each patient. Microsurgical<br />

resection was performed by an operating microscope enabled to visualize<br />

the fluorescence. All the patients, as first line treatment, have been<br />

submitted to radiotherapy and chemotherapy; second and in some cases<br />

third line treatments were utilized in recurrent cases. The patients<br />

follow-up ranged from 2 years to 6 months. Results: In more than 90% <strong>of</strong><br />

patients tumor tissue showed intraoperative red fluorescence; mainly in<br />

recurrent GBM, when MRI documented heterogeneous lesions with enhancing<br />

areas mixed with non enhancing gliotic scars, fluorescence-guided<br />

surgery allowed a better definition <strong>of</strong> active tumor, with net margins from<br />

perilesional “healthy” brain. Early postoperative MRI confirmed gross total<br />

resection without contrast enhancement in 80 % <strong>of</strong> patients. In the present<br />

experience the procedure did not determine any relevant additional<br />

neurological deficit. Considering overall survival <strong>of</strong> recurrent patients we<br />

obtained a median extension <strong>of</strong> at least 9.0 months (3 – 16� months).<br />

Conclusions: Fluorescence-guided surgery improves tumor detection and<br />

allows extended resection <strong>of</strong> malignant glioma, without any relevant impact<br />

on neurological status, resulting helpful mainly in the recurrent setting with<br />

a consistent effect on overall survival.<br />

2087 General Poster Session (Board #18B), Sat, 1:15 PM-5:15 PM<br />

Correlation <strong>of</strong> survival with tumor antigen expression in patients with newly<br />

diagnosed glioblastoma receiving a multi-epitope pulsed dendritic cell<br />

vaccine. Presenting Author: Surasak Phuphanich, Neuro-Oncology Program,<br />

Cedars-Sinai Medical Center, Los Angeles, CA<br />

Background: This study evaluated the safety and immune responses to an<br />

autologous dendritic cell vaccine pulsed with class I peptides from tumor<br />

associated antigens (TAA) expressed on gliomas and overexpressed in their<br />

cancer stem cell population (ICT-107). These TAA epitodes AIM-2. TRP-2,<br />

HER2 and AIM-2, are also overexpressed on glioblastoma multiforme<br />

(GBM) cancer stem cells. Methods: TAA epitopes included HER2, TRP-2,<br />

gp100, MAGE-1, IL13R�2, and AIM-2. HLA-A1 and/or HLA-A2 positive<br />

glioblastoma (GBM) patients with gross total resection were eligible.<br />

Mononuclear cells from leukapheresis were differentiated into dendritic<br />

cells, pulsed with TAA peptides, and administered intradermally three<br />

times at two-week intervals in the axilla region after a standard treatment<br />

with concurrent temozolomide (TMZ) and radiation therapy for newly<br />

diagnosed (ND-GBM). Results: Twenty-one patients were enrolled with 17<br />

ND-GBM and three recurrent GBM patients and one brainstem glioma.<br />

Immune response data on 15 newly diagnosed patients showed 33%<br />

responders. TAA expression by qRT-PCR showed all patient tumors<br />

expressed at least three TAA with 75% expressing all six. Correlations <strong>of</strong><br />

increased PFS and quantitative expression <strong>of</strong> MAGE1, AIM-2, gp100 and<br />

HER2 were observed. A decrease or absence <strong>of</strong> CD133 expression was seen<br />

in five patients who underwent a second resection. As <strong>of</strong> February 1, 2012,<br />

six <strong>of</strong> 16 ND-GBM patients showed no evidence <strong>of</strong> tumor recurrence<br />

(44-63 months). Median progressive free survival (PFS) in newly diagnosed<br />

patients was 16.9 months with a two-year PFS rate was 43.8%<br />

(95%CI,19.8-66.0 ). The median overall survival rate (OS) was 38.4<br />

months and a two-year OS was 80.3% (95%CI,58.6-96.7). Conclusions:<br />

Expression <strong>of</strong> four ICT-107 targeted antigens in the pre-vaccine tumors<br />

correlated with prolonged overall survival and PFS in ND-GBM patients.<br />

The goal <strong>of</strong> targeting tumor antigens highly expressed on glioblastoma<br />

cancer stem cells is supported by the observation <strong>of</strong> decreased or absent<br />

CD133 expression in the recurrent areas <strong>of</strong> gadolinium-enhanced tumor.<br />

2086 General Poster Session (Board #18A), Sat, 1:15 PM-5:15 PM<br />

An investigator-initiated monocentric phase I dose escalation trial <strong>of</strong><br />

temsirolimus and irinotecan (TEMIR) in patients with relapsing glioblastoma<br />

multiforme (reGBM). Presenting Author: Max E. Scheulen, Department<br />

<strong>of</strong> Medical Oncology, West German Cancer Center, University <strong>of</strong><br />

Duisburg-Essen, University Hospital Essen, Essen, Germany<br />

Background: Deregulation <strong>of</strong> the PI3K/AKT pathway has been preclinically<br />

involved in the pathophysiology <strong>of</strong> GBM. The mammalian target <strong>of</strong><br />

rapamycin (mTOR) is an important downstream mediator <strong>of</strong> PI3K/AKT<br />

signalling. The mTOR-inhibitor temsirolimus (Tem) achieves significant<br />

drug levels in brain. Thus, Tem may be effectively combined with irinotecan<br />

(Iri) which has shown promising results in combination with bevacizumab<br />

in patients (pts) with reGBM refractory to temozolomide (Tmz) in phase II.<br />

Exposure to Tem as well as Iri is substantially reduced in pts receiving<br />

CYP3A4 enzyme-inducing anticonvulsants (CYP3A4-Ind). Methods: TEMIR<br />

is a phase I trial <strong>of</strong> escalating doses <strong>of</strong> Tem in combination with Iri in pts<br />

with reGBM refractory to Tmz depending on the cotreatment with CYP3A4-<br />

Ind. In pts without CYP3A4-Ind dose escalation <strong>of</strong> Tem was performed<br />

according to the “3�3 protocol” from 15 via 20 to 25 mg iv wkly together<br />

with 85 mg/m2 Iri iv wkly. In pts with CYP3A4-Ind the doses <strong>of</strong> Tem were<br />

30, 40 and 50 mg iv wkly together with 170 mg/m2 Iri iv wkly, respectively.<br />

The determination <strong>of</strong> the pharmacokinetics (PK) <strong>of</strong> Tem and its influence<br />

on the PK <strong>of</strong> Iri and its active metabolite SN38 was performed by validated<br />

RP-HPLC methods with fluorescence detection. Results: Up to now, 14 pts<br />

[4 female and 10 male, median age 47 yrs (range 41-65 yrs), 9 without and<br />

5 with CYP3A4-Ind] have been treated. All 9 pts without CYP3A4-Ind (3<br />

pts on 15, 20 and 25 mg Tem each) are evaluable for safety with only mild<br />

toxicity [highest CTC-grade �2 (gr) per pt]: diarrhea 4 (gr2) and 1 (gr3),<br />

neutropenia 1 (gr3), thrombopenia 1 (gr2), anemia 1 (gr2), pneumonia 2<br />

(gr2), rash 1 (gr2). PK parameters <strong>of</strong> Iri and SN38 were in agreement with<br />

previously published studies and there was no significant effect <strong>of</strong> 15 to 25<br />

mg Tem combined with 85 mg/m2 Iri on the PK <strong>of</strong> Iri and SN38. In 7 pts<br />

evaluable for efficacy there was no remission (RECIST) and median time to<br />

symptomatic and/or MRT progression was 10 wks (7, 9�, 10, 10�, 11,<br />

15, 20 wks). Conclusions: The combination <strong>of</strong> Tem in standard dose <strong>of</strong> 25<br />

mg iv wkly with 85 mg/m2 Iri wkly is safe in pts with reGBM without<br />

CYP3A4-Ind. Tem has no significant effect on the PK <strong>of</strong> Iri and SN38.<br />

2089 General Poster Session (Board #18D), Sat, 1:15 PM-5:15 PM<br />

High-dose chemotherapy (HDC) followed by autologous stem cell transplant<br />

(ASCT) for recurrent/progressive CNS lymphoma. Presenting Author:<br />

Mary Roberta Welch, Memorial Sloan-Kettering Cancer Center, New York,<br />

NY<br />

Background: In two reports by Soussain et al, promising efficacy was<br />

observed in recurrent primary CNS lymphoma with induction cytarabine/<br />

VP-16 (CYVE) followed by HDC-ASCT (busulfan, thiotepa and cyclophosphamide<br />

[BTC]), but significant toxicity, mainly from CYVE, has limited<br />

widespread use. We report our experience with HDC-ASCT with alternative<br />

induction regimens. Methods: Retrospective review <strong>of</strong> pts with recurrent/<br />

refractory non-Hodgkin lymphoma (NHL) with CNS involvement treated<br />

with HDC-ASCT (2000-present). Results: Seventeen pts met inclusion<br />

criteria: med age� 58 (41-65); 9 were women; med KPS prior to<br />

transplant� 90 (range 70-100). At initial presentation, 10 had primary<br />

CNS lymphoma (ocular: 1); 7 had systemic NHL without CNS involvement;<br />

1 had both systemic and CNS disease. Pts had been heavily pre-treated.<br />

Among those with PCNSL, high dose MTX was used in all pts and WBRT in<br />

4. Two pts had received a previous HDC-ASCT. Among systemic NHL pts,<br />

various regimens were used, mostly R-CHOP(4), but also R-EPOCH (1),<br />

CVP (1), ICE (1) and CODOX-M (1). At CNS recurrence, pts received various<br />

induction regimens prior to HD-ASCT: high-dose methotrexate (MTX)based<br />

chemotherapy (N� 13), cytarabine-based regimens (N�2), and<br />

other (N� 2). All pts achieved a CR or near CR prior to HDC-ASCT.<br />

Harvesting was obtained with G-CSF alone in 9 pts; 8 required plerixafor.<br />

Two pts failed mobilization N�15 received HDC-ASCT. The HDC consisted<br />

<strong>of</strong> BTC (N�13); 1 received BEAM and 1 received reduced intensity<br />

fludarabine, melphalan and alemtuzumab. Eight pts experienced a grade<br />

III or IV toxicity – most commonly fatigue, febrile neutropenia, and<br />

infection. One previously transplanted pt died from sepsis. With a med<br />

follow-up <strong>of</strong> 11 months, post-transplant med-PFS has not been reached.<br />

The 12m PFS was 92% (95% CI 56-98). Because no patient has<br />

progressed, the OS was identical to PFS. Conclusions: HDC-ASCT was a<br />

highly effective salvage approach in this population <strong>of</strong> recurrent/refractory<br />

CNS lymphoma. To reduce the risk <strong>of</strong> harvesting failure at the time <strong>of</strong><br />

recurrence, harvesting stem cells at the time <strong>of</strong> initial treatment could be<br />

considered in pts with high risk for relapse.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


2090^ General Poster Session (Board #18E), Sat, 1:15 PM-5:15 PM<br />

The addition <strong>of</strong> bevacizumab to temozolomide and radiation therapy<br />

followed by bevacizumab, temozolomide, and oral topotecan for newly<br />

diagnosed glioblastoma multiforme (GBM). Presenting Author: Henry S.<br />

Friedman, Duke University Medical Center, Durham, NC<br />

Background: The prognosis for newly-diagnosed GBM is dismal. The<br />

addition <strong>of</strong> temozolomide to radiation therapy improved the median overall<br />

survival (OS) to 14.6 months (mos). GBM’s have the highest levels <strong>of</strong><br />

vascular endothelial growth factor (VEGF). Hypoxia inducing factor-1 alpha<br />

(HIF-1 alpha) is an important regulator <strong>of</strong> VEGF, and topotecan may inhibit<br />

HIF-1 alpha. Methods: We performed a phase II trial in newly diagnosed<br />

GBM by adding bevacizumab and topotecan to standard therapy. 80 newly<br />

diagnosed GBM patients were enrolled between January 2010 and January<br />

2011. Patients received standard radiation therapy and temozolomide.<br />

Bevacizumab at 10 mg/kg every 14 days was added a minimum <strong>of</strong> 4 weeks<br />

post-op. Two weeks after radiation therapy was completed, 12 monthly<br />

cycles <strong>of</strong> temozolomide at 150 mg/m2 /d days 1-5, oral topotecan at 1.5<br />

mg/m2 for patients not on an enzyme inducing anti-epileptic drug (EIAED)<br />

and 2.0 mg/m2 for patients on an EIAED days 2-6, and bevacizumab at 10<br />

mg/kg on days 1 and 15. Results: The addition <strong>of</strong> bevacizumab to standard<br />

radiation therapy and daily temozolomide was safe. Of the 80 patients, 76<br />

completed radiation therapy. Four patients did not complete radiation, two<br />

with clinical decline, one each with a bone flap infection, and a pulmonary<br />

embolus. Fifteen patients came <strong>of</strong>f study for toxicity, five with recurrent<br />

grade IV thrombocytopenia, three with grade III fatigue, two each with<br />

grade 2 CNS hemorrhage, and wound dehiscence requiring surgery and one<br />

each with GI perforation, pulmonary embolism and an aortic thrombus. Of<br />

the 80 patients, 56 have progressed and 37 have died. The median<br />

progression-free survival (PFS) and OS are 11.1 mos (95% CI: 9.4-13.6)<br />

and 17.2 mos (95% CI: 15.2) at a median follow-up <strong>of</strong> 18.4 months. The<br />

two year OS is 45.3%. Conclusions: The addition <strong>of</strong> bevacizumab to<br />

temozolomide and radiation followed by temozolomide, bevacizumab and<br />

oral topotecan is tolerable. The median PFS and OS are encouraging. The<br />

randomized phase III trials with bevacizumab for newly diagnosed GBM<br />

patients are essential.<br />

2092 General Poster Session (Board #18G), Sat, 1:15 PM-5:15 PM<br />

Primary central nervous system lymphoma: The Cleveland Clinic experience.<br />

Presenting Author: Manmeet Singh Ahluwalia, Cleveland Clinic,<br />

Cleveland, OH<br />

Background: Primary central nervous system lymphoma (PCNSL) is an<br />

uncommon variant <strong>of</strong> extranodal non-Hodgkin lymphoma that involves the<br />

brain, leptomeninges, eyes, or spinal cord without evidence <strong>of</strong> systemic<br />

disease. Untreated PCNSL has a rapidly fatal course, with survival <strong>of</strong><br />

approximately 1.5 months from the time <strong>of</strong> diagnosis. In this study, we<br />

sought to describe the demographics, diagnoses, management, and outcomes<br />

<strong>of</strong> patients with PCNSL at a single institution. Methods: After<br />

obtaining IRB approval, the Cleveland Clinic Brain Tumor and Neuro-<br />

Oncology Center’s database was used to identify patients with histologically<br />

proven PCNSL at the Cleveland Clinic between 1986 and 2010. Data were<br />

subjected univariate and multivariate analysis followed by recursive partitioning<br />

analysis in order to generate a prognostic model. Results: 153<br />

patients were diagnosed with PCNSL with a median age <strong>of</strong> 61 and<br />

Karn<strong>of</strong>sky performance status (KPS) <strong>of</strong> 70. The progression-free survival<br />

(PFS) was 9.3 months; the overall survival (OS) was 27 months. The<br />

treatment regimen included methotrexate-based chemotherapy (MTX) or a<br />

combination <strong>of</strong> methotrexate-based chemotherapy and whole brain radiation<br />

therapy (WBRT). Patients treated with the MTX regimen had an OS <strong>of</strong><br />

65 months; those treated with the MTX � WBRT regimen had an OS <strong>of</strong> 74<br />

months. The Cox proportional hazards regression identified age and KPS as<br />

the only prognostic indicators to OS and PFS. Recursive partitioning<br />

analysis categorized the patients into three groups according to these<br />

prognostic factors. Patients with KPS � 70 had a favorable outcome<br />

compared to patients with KPS � 70. This held true especially for patients<br />

age 60 and younger, whose median OS was 100 months. Conclusions: The<br />

survival and prognostic indicators approximate those reported previously<br />

and provide independent validation for a simple yet powerful prognostic<br />

model that uses age and KPS to predict survival.<br />

PFS and OS <strong>of</strong> patients in different groups based on age and performance<br />

status.<br />

Outcome PFS (months) OS (months)<br />

All patients 9.3 27<br />

Group 1 17 100<br />

Group 2 18 41<br />

Group 3 2.3 6.0<br />

Group 1: age � 60 years and KPS � 70; group 2: age � 60 years and KPS � 70;<br />

group 3: KPS � 70 (p � 0.0001).<br />

Central Nervous System Tumors<br />

137s<br />

2091 General Poster Session (Board #18F), Sat, 1:15 PM-5:15 PM<br />

Comparison <strong>of</strong> fractionated and single session radiosurgery for radiation<br />

resistant brain metastases. Presenting Author: Eric Karl Oermann, University<br />

<strong>of</strong> North Carolina at Chapel Hill, Chapel Hill, NC<br />

Background: Melanoma and renal cell carcinoma are commonly called<br />

radiation resistant tumors due to the decreased response rate to traditional<br />

radiation (1.8-2 Gy /Fraction). Large brain metastases from radioresistant<br />

primaries are clinical challenges. This study examines the impact <strong>of</strong><br />

fractionation on the treatment <strong>of</strong> intracranial metastases from radiation<br />

resistant tumors. Methods: Patients with a primary diagnosis <strong>of</strong> melanoma<br />

or renal cell carcinoma and intracranial metastases who completed<br />

treatment at The University <strong>of</strong> North Carolina with frameless robotic<br />

radiosurgery between 2007-2011 were retrospectively analyzed. Patients<br />

were treated with either single or 3-5 fractions depending on volume. The<br />

study’s primary endpoints were overall survival (OS) and local control (LC),<br />

and its secondary endpoint was patient steroid requirements. Outcomes<br />

data and pre-treatment variables were analyzed for significance using<br />

appropriate non-parametric tests. Results: 25 patients were included in the<br />

single fraction arm and 13 patients in the multi-fraction arm, and both had<br />

equivalent pre-treatment characteristics with the exception <strong>of</strong> tumor<br />

volume which was larger in the multi-fraction arm (p�0.01). At a median<br />

follow-up <strong>of</strong> 4.7 months (range, 1.8-11.6), the median OS for all patients<br />

was 6.2 months. One year survival was 35.1% and 5 patients (13%) had<br />

local failures at a median time to local failure <strong>of</strong> 5.5 months. Patients in the<br />

multi-fraction arm failed at a higher rate (10.5%) than patients in the<br />

single fraction arm (2.6%) (p�0.04), but there was no difference in OS<br />

(p�0.34). There was no difference between pre-treatment and posttreatment<br />

steroid requirements between the two arms (p�0.351).<br />

Conclusions: Fractionation is intended to facilitate radiosurgery in large<br />

tumors by limiting high doses <strong>of</strong> radiation to a large portion <strong>of</strong> normal brain.<br />

In our study <strong>of</strong> radioresistant intracranial metastases, large tumors receiving<br />

multi-fraction radiosurgery had decreased local control with no difference<br />

in toxicity or OS. These results suggest a need for either dose<br />

escalation, or combination therapy designed to reduce tumor size (resection<br />

or aspiration) in order to facilitate single fraction treatment.<br />

2093 General Poster Session (Board #18H), Sat, 1:15 PM-5:15 PM<br />

Low-grade gliomas in older patients. Presenting Author: Adewale Alade<br />

Fawole, Fairview Hospital, Cleveland, OH<br />

Background: Low grade gliomas account for 10-20% <strong>of</strong> all primary brain<br />

tumors. The outcomes <strong>of</strong> older patients with low-grade glioma (LGG) are not<br />

well known. Methods: After obtaining IRB approval, the Cleveland Clinic<br />

Brain Tumor and Neuro-Oncology Center’s database was used to identify<br />

older patients defined as those �55 years <strong>of</strong> age with histologically<br />

confirmed grade 2 glioma at the time <strong>of</strong> diagnosis. Multivariable analysis<br />

was conducted with use <strong>of</strong> a Cox proportional hazards model and a stepwise<br />

selection algorithm that used p�.10 as the criteria for entry and p�0.05 as<br />

retention in the model to identify independent predictors <strong>of</strong> survival.<br />

Results: Chart records <strong>of</strong> 61 patients diagnosed between 1991 and 2010<br />

were included for final analysis. In contrast to patients �55 years, older<br />

patients are more likely to be male (p�.06), have poorer performance<br />

status at presentation (p�.0001), more comorbid conditions (p�.0001),<br />

present with more neurologic symptoms (p�.0001), have a prior history <strong>of</strong><br />

cancer (p�.0001). They are more likely to have astrocytic histology<br />

(p�.008) and present with multifocal tumors more frequently (p�.001).<br />

Older patients received radiation (RT) upfront more frequently (p�.07) and<br />

undergo gross total resection less frequently (p�.003). Median Progression<br />

free survival (PFS) and median overall survival (OS) was 1.9 and 4.3 years,<br />

respectively in these patients. On univariate analysis, patients with poor<br />

performance status have worse PFS (p�.01) and OS (p�.003). Patients<br />

with memory impairment have worse PFS (p�.009) and OS (p�.0001).<br />

Patients treated with adjuvant chemotherapy had better OS (p�.01).<br />

Tumor related characteristics associated with poor survival included, �pure�<br />

or mixed astrocytoma histology (OS, p�.06), multifocal tumors (OS,<br />

p�.08), left-sided tumors (p�.07). In multivariable analysis, performance<br />

status was the only independent predictor <strong>of</strong> either PFS or OS. Conclusions:<br />

Older patients with low grade gliomas have less favorable outcomes as<br />

compared to younger patients. They have more comorbid conditions and<br />

symptoms at presentation. Use <strong>of</strong> chemotherapy in this patient group was<br />

associated with improved survival. Performance status was the only<br />

independent predictor <strong>of</strong> either PFS or OS.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


138s Central Nervous System Tumors<br />

2094^ General Poster Session (Board #19A), Sat, 1:15 PM-5:15 PM<br />

Safety and efficacy <strong>of</strong> the addition <strong>of</strong> bevacizumab to temozolomide and<br />

radiation therapy followed by bevacizumab, temozolomide, and irinotecan<br />

for newly diagnosed glioblastoma multiforme. Presenting Author: James J.<br />

Vredenburgh, Duke University Medical Center, Durham, NC<br />

Background: Glioblastoma (GBM) has a very poor prognosis, and the<br />

majority <strong>of</strong> patients die within 2 years <strong>of</strong> diagnosis. GBMs have high<br />

concentrations <strong>of</strong> vascular endothelial growth factor (VEGF), and the more<br />

VEGF, the worse the prognosis. Bevacizumab is a humanized antibody to<br />

VEGF and is active in recurrent GBMs. The study aims to improve the<br />

survival <strong>of</strong> newly diagnosed GBM patients by incorporating an antiangiogenic<br />

agent with radiation and temozolomide, and adding a topoisomerase<br />

I inhibitor, and an anti-angiogenic agent to temozolomide postradiation<br />

therapy. Methods: Patients received standard radiation and<br />

temozolomide at 75 mg/m2 /day, with bevacizumab at 10 mg/kg every 14<br />

days beginning a minimum <strong>of</strong> 28 days post-operatively. Following the<br />

completion <strong>of</strong> radiation therapy, patients received 6-12 cycles <strong>of</strong> bevacizumab,<br />

temozolomide and irinotecan. Each cycle was 28 days. Bevacizumab<br />

was given at a dose <strong>of</strong> 10 mg/kg days 1 and 15, temozolomide 200<br />

mg/m2 days 1-5 and irinotecan on days 1 and 15 at 125 mg/m2 for patients<br />

not on an enzyme inducing anti-epileptic (EIAED) and 340 mg/m2 for<br />

patients on an EIAED. The statistical design was a goal <strong>of</strong> 60% overall<br />

survival at 16 months. Results: 125 patients were enrolled between 8/07<br />

and 3/09. All the patients have completed therapy. Nine patients had<br />

thromboembolic complications (DVT or PE). Two patients had wound<br />

dehiscence, one bowel perforation, one secondary AML and two pneumocystis<br />

carinii pneumonias (PCP). Seventeen had grade 4 hematologic toxicity<br />

requiring dose decrements. There were 4 toxic deaths, one each with a<br />

myocardial infarction PCP, PE and sepsis. At a median follow-up <strong>of</strong> 40<br />

mos, the median overall survival was 20.9 mos, the median progressionfree<br />

survival was 13.8 mos and the 2-year overall survival was 42.4%.<br />

Conclusions: Adding bevacizumab to temozolomide and radiation therapy<br />

followed by bevacizumab, temozolomide with irinotecan is tolerable and<br />

efficacious.<br />

2096 General Poster Session (Board #19C), Sat, 1:15 PM-5:15 PM<br />

Prognostic nomogram in elderly patients with glioblastoma. Presenting<br />

Author: Mital Patel, Cleveland Clinic, Cleveland, OH<br />

Background: Glioblastoma (GBM) is the most common malignant primary<br />

brain tumor. More than half the cases occur in patients aged �65 years;<br />

however, there is limited data on specific prognostic factors in these<br />

patients and there is a lack <strong>of</strong> easy to use nomograms in elderly GBM<br />

patients to provide them prognostic information. Methods: The Cleveland<br />

Clinic Brain Tumor and Neuro-Oncology Center’s database was used to<br />

identify elderly patients with GBM ( �65 years at the time <strong>of</strong> diagnosis).<br />

Multivariable analysis was conducted to identify independent predictors <strong>of</strong><br />

survival using a Cox proportional hazards model and a stepwise selection<br />

algorithm with p�.10 as criteria for entry and retention. �Points� were<br />

assigned to each <strong>of</strong> these poor prognostic features and prognostic groups<br />

were formed based on the total number <strong>of</strong> calculated points. Results: 512<br />

GBM patients with a median age <strong>of</strong> 73 years (range 65-96, 54% male) were<br />

included in the final analysis. On multivariable analysis six factors were<br />

identified as having an independent impact on overall survival after<br />

controlling for non-surgical treatment related factors like chemotherapy<br />

and radiation: age at diagnosis (p�.0001), surgery (p� .0001), multifocal<br />

disease (p�.0008), seizure at presentation (p�.02), hypertension (p�.05)<br />

and Karn<strong>of</strong>sky Performance Status (p�.0001). Prognostic groups were<br />

developed as summarized in the table. 28% <strong>of</strong> the patients had the most<br />

favorable pr<strong>of</strong>ile which was associated with 56% one year survival and a<br />

13.2 months median survival. In contrast, almost all patients in the least<br />

favorable group (27%) died within one year. Conclusions: This is an easy to<br />

use nomogram in elderly GBM patients to provide them prognostic<br />

information.<br />

Prognostic groups in elderly patients (> 65 years) with glioblastoma.<br />

Prognostic group No. <strong>of</strong> “points” 1 N (%)<br />

1-yr survival<br />

(%)<br />

Median survival<br />

(months)<br />

Favorable 0-4 123 (28%) 56% 13.2<br />

Intermediate 5-8 196 (45%) 19% 6.5<br />

Unfavorable �8 117 (27%) 1% 2.8<br />

1 Lack <strong>of</strong> seizure, HTN � 1 point; multiple tumors, age 70-79, KPS 70-80 � 2<br />

points; biopsy only � 3 points; age �80, KPS �70 � 4 points.<br />

2095^ General Poster Session (Board #19B), Sat, 1:15 PM-5:15 PM<br />

Phase II study <strong>of</strong> bevacizumab plus irinotecan and carboplatin for recurrent<br />

WHO grade 3 malignant gliomas with no prior bevacizumab failure.<br />

Presenting Author: Annick Desjardins, Duke University Medical Center,<br />

Durham, NC<br />

Background: No standard treatment exists for recurrent WHO grade 3<br />

malignant glioma patients. Bevacizumab (BV) with irinotecan has significant<br />

anti-tumor activity for recurrent glioblastoma. Carboplatin has antiglioma<br />

activity and can potentiate the cytotoxicity <strong>of</strong> irinotecan. We<br />

evaluated the progression-free survival (PFS) <strong>of</strong> BV in combination to<br />

irinotecan and carboplatin in recurrent WHO grade 3 malignant gliomas, as<br />

well as its safety. Methods: Adult patients with measurable recurrent WHO<br />

grade 3 malignant glioma, �12 weeks after radiation therapy, �4 weeks<br />

after chemotherapy, with adequate organ function, KPS �70%, no prior<br />

failure or grade �3 toxicity to the agents, and no contraindications to BV<br />

were eligible for study. Patients received BV at 10 mg/kg with irinotecan on<br />

days 1 and 15 <strong>of</strong> a 28-day cycle. The dose <strong>of</strong> irinotecan was 125 mg/m2 for<br />

patients not on enzyme inducing anti-epileptics (EIAEDs) and 340 mg/m2<br />

for patients on EIAEDs. All patients received carboplatin at an AUC <strong>of</strong> 4 on<br />

day 1 <strong>of</strong> each cycle. MRIs were obtained every 8 weeks. Results: As<br />

planned, 39 WHO grade III malignant glioma patients were enrolled on<br />

study. Median age was 47 (range, 26-71). At a median follow up <strong>of</strong> 14<br />

months, the 6-month PFS is 69% and the median PFS is 11 months. A<br />

median <strong>of</strong> 8 cycles were given. Seven patients completed the planned<br />

course <strong>of</strong> treatment (12 cycles) with hypometabolic PET scan and nine<br />

patients remain on study. Fifteen patients came <strong>of</strong>f study due to disease<br />

progression and eight due to toxicity. As <strong>of</strong> 1/25/2012, 22 patients are still<br />

alive and 17 have died. Grade 3-4 toxicities included: neutropenia (grade<br />

3, n�12; grade 4, n� 1), thrombocytopenia (grade 3, n�6; grade 4, n�4),<br />

nausea (grade 3, n�7), diarrhea (grade 3, n�2), deep venous thrombosis<br />

(grade 3, n�2), febrile neutropenia (grade 3, n�1; grade 4, n�1), fatigue<br />

(grade 3, n�8; grade 4, n�1), cerebral infarction (grade 4, n�3),<br />

intracranial hemorrhage (grade 4, n�1), posterior reversible encephalopathy<br />

syndrome (grade 3, n�1). Conclusions: The combination <strong>of</strong> bevacizumab,<br />

irinotecan and carboplatin for WHO grade 3 malignant glioma<br />

patients is effective and with no more than expected toxicity.<br />

2097 General Poster Session (Board #19D), Sat, 1:15 PM-5:15 PM<br />

Poor prognostic factors <strong>of</strong> post-CNS recurrence survival in breast cancer<br />

patients. Presenting Author: Carlos Castaneda Altamirano, Instituto Nacional<br />

de Enfermedades Neoplásicas, Lima, Peru<br />

Background: Survival after the onset <strong>of</strong> metastases in the central nervous<br />

system is very short. However, some variables could indicate subsets <strong>of</strong><br />

worse prognosis. Our aim was to determine the value <strong>of</strong> clinicopathological<br />

characteristics and prognostic scores in the post-SNC recurrence survival.<br />

Methods: We evaluated a retrospective cohort <strong>of</strong> 2597 breast cancer<br />

patients treated at the Instituto Nacional de Enfermedades Neoplasicas<br />

(Lima-Peru) between 2000-2005. Clinicopathological data was retrieved,<br />

RPA and GPA brain metastases prognostic scores were constructed and<br />

phenotypes were categorized according to the IHC expression in<br />

[HR�,HER2-], [Any HR, HER2�] and Triple Negative. Survival was<br />

calculated according to the Kaplan Meier methodology and cases were<br />

stratified by variables evaluated. The log-rank or Breslow tests were used<br />

when appropriate and multivariate analysis was done by the cox regression.<br />

AP�0.05 was considered statistically significant. Results: One hundred<br />

and fifty seven cases developed CNS metastasis, from which 23 developed<br />

leptomeningeal metastases. The post recurrence CNS survival was 0.405<br />

years. There were not differences according to phenotype (P�0.102),<br />

histological grade (P�0.647), number <strong>of</strong> brain metastases (P�0.695) and<br />

metastases volume (P�0.155). We found statistic differences in regard to<br />

leptomeningeal carcinomatosis (present, 0.249ys vs absent 0.436ys;<br />

P�0.033); CSF infiltration (present, 0.115ys vs absent, 1.044ys;<br />

P�0.022); status <strong>of</strong> primary tumor (controlled, 0.501ys vs uncontrolled,<br />

0.263ys; P�0.001); ECOG performance status (�2, 0.504ys vs �2,<br />

0.288ys; P�0.030); and time from BC diagnosis to SNC metastases (�8<br />

moths, 0.115 vs �8 months, 0.425ys; P�0.023). Cox regression identifies<br />

to CSF infiltration as statistically significant (HR�9.77; P�0.025). In<br />

regard to Prognostic scores, we found differences when cases were<br />

stratified according to RPA score (Class I, 0.564ys vs Class II, 0.455ys vs<br />

Class III, 0.288ys; P�0.049) and GPA score (0-1, 0.26ys vs 1.5-3,<br />

0.455ys vs 3.5-5, 0,564; 0.048). Conclusions: RPA and GPA scores are<br />

more accurate to identify poor survival subsets in this group <strong>of</strong> patients than<br />

other tumor features (phenotype or histology).<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


2098 General Poster Session (Board #19E), Sat, 1:15 PM-5:15 PM<br />

Effects <strong>of</strong> lazaroid U-74389G liposomes in a glioblastoma mouse model.<br />

Presenting Author: Fady Ibrahim, University <strong>of</strong> British Columbia, Vancouver,<br />

BC, Canada<br />

Background: Lazaroid U-74389G (LAZ) is a 21-aminosteroid that has<br />

radioprotective effects against radiation-induced lipid peroxidation. Also in<br />

vitro antiproliferative effects have been reported against glioblastoma cell<br />

lines. The aim <strong>of</strong> this study was to use a liposomal formulation to evaluate<br />

LAZ radioprotective and antiproliferative effects in a glioblastoma mouse<br />

model. Methods: LAZ PEGylated liposomes (Lipo G) were developed at the<br />

University <strong>of</strong> Houston, College <strong>of</strong> Pharmacy. Glioblastoma cell line U87expressing<br />

firefly luciferase reporter gene (100,000 cells in 2 �L) was<br />

injected intracranially in each male SCID hairless outbred mouse. There<br />

were 4 treatment groups (n�8-9, each): brain model (M) without treatment<br />

(control), radiation 2Gy weekly (M�R), Lipo G at 5 mg/kg dose IP twice per<br />

week (M�L) and radiation with Lipo G (M�R�L). Treatment lasted three<br />

weeks. Tumor size was monitored using non-invasive bioluminescence<br />

imaging (BLI), in each mouse. Mice were sacrificed after 3 weeks. Brain<br />

was harvested for immunohistochemistry examinations. Lipid peroxidation<br />

<strong>of</strong> brain tissues was quantified by measuring malondialdehyde (MDA) as a<br />

surrogate biomarker. Survival was evaluated using Kaplan Meier analysis at<br />

P� 0.05. Results: The relative BLI intensity was 4002.03�1737.67,<br />

2034�737.72, 1387.36�684.53 and 2498.89�2521.32 % for M,<br />

M�R, M�L and M�R�L, respectively. The tumor size <strong>of</strong> the M�L group<br />

was reduced by 65% compared to control . For the radiation treated groups<br />

(M�R and M�R�L), there was no significant difference in tumor size<br />

compared to control group. MDA brain concentration in M�L and M�R�L<br />

groups was significantly less than in M�R group (8.27�0.78 and<br />

10.37�3.30 �M/gm vs. 23.09� 3.79 �M/gm). The survival mean was<br />

22.67, 25.33, 25.22 and 27.13 days for M, M�R, M�R�L and M�L<br />

groups, respectively. Mean survival <strong>of</strong> LAZ treated groups (M�L and<br />

M�R�L) was significantly longer than that <strong>of</strong> the control group Conclusions:<br />

LAZ liposomal formulations administered at 5 mg/kg dose reduced tumor<br />

growth by 65%. LAZ also protected brain tissue from radiation-induced<br />

lipid peroxidation by reducing MDA concentration by 50%. These provocative<br />

data warrant further investigation <strong>of</strong> LAZ as a radiation protectant and<br />

chemotherapeutic agent.<br />

2100 General Poster Session (Board #19G), Sat, 1:15 PM-5:15 PM<br />

Anaplastic oligodendroglial tumors: The Cleveland Clinic experience.<br />

Presenting Author: Neda Hashemi-Sadraei, Cleveland Clinic, Cleveland,<br />

OH<br />

Background: Anaplastic oligodendroglial tumors include both anaplastic<br />

oligodendroglioma (AO), and anaplastic oligoastrocytoma (AOA), histological<br />

categories <strong>of</strong> WHO grade 3 gliomas. There is limited data on specific<br />

prognostic factors for patients with these tumors. Methods: After obtaining<br />

IRB approval, the Cleveland Clinic Brain Tumor and Neuro-Oncology<br />

Center’s database was used to identify patients with histologically confirmed<br />

AO and AOA at the time <strong>of</strong> diagnosis. Multivariable analysis was<br />

conducted with use <strong>of</strong> a Cox proportional hazards model and a stepwise<br />

selection algorithm that used p�0.05 both as the criteria for entry and<br />

retention in the model to identify independent predictors <strong>of</strong> survival.<br />

Results: Chart records <strong>of</strong> 139 patients, 52% <strong>of</strong> whom were male, diagnosed<br />

between 1992 and 2009 were included for analysis. Median age at<br />

presentation was 47 years (range, 18-83 years). 22% <strong>of</strong> patients had<br />

biopsy only, 35% had gross total resection, 43% had near total resection or<br />

subtotal resection. Following surgery, 30% <strong>of</strong> patients were treated with<br />

chemotherapy (CT) alone, 14% were treated with radiotherapy (RT) and<br />

47% received both CT and RT. Median progression free survival and<br />

median overall survival (OS) were 29.0 and 58.7 months respectively. On<br />

multivariate analysis, four factors were identified as independent predictors<br />

<strong>of</strong> OS: age at diagnosis (�50 vs. �50, p�0.004), Hypothyroidism<br />

(p�0.009), multifocal disease (p�0.005) and 1p 19q co-deletion<br />

(p�0.001). Choice <strong>of</strong> initial therapy did not impact survival in this cohort<br />

<strong>of</strong> patients. Conclusions: Older age, hypothyroidism and multifocal disease<br />

were associated with higher mortality. 1p, 19q co-deletion was associated<br />

with lower mortality.<br />

Risk factors for mortality: Multivariable Cox proportional hazards analysis.<br />

Factor Hazard ratio (95% C.I.) p2 Age at diagnosis, years<br />

(>50 vs.


140s Central Nervous System Tumors<br />

2102 General Poster Session (Board #20A), Sat, 1:15 PM-5:15 PM<br />

Correlation between CD4 lymphocytes count and the opportunistic infections<br />

in treated glioblastoma. Presenting Author: Marie Josee Begin,<br />

University <strong>of</strong> Montreal, Montreal, QC, Canada<br />

Background: Less than 200 CD4 lymphocytopenia happens frequently with<br />

temozolomide treatment <strong>of</strong> glioblastoma. This immunosuppression is<br />

associated with many opportunistic infections and antibiotic prophylaxis is<br />

given in some centers to prevent Pneumocystis pneumonia. Methods: We<br />

analyzed the association between documented culture-proved infections<br />

and CD4 lymphocytes level in 140 patients treated with temozolomide in<br />

first-line glioblastoma in Notre-Dame Hospital from 2006 to 2009.<br />

Demographic and treatment data were collected and analyzed with Kaplan-<br />

Meier survival plots and Log Rank tests. Results: The cohort <strong>of</strong> infected<br />

patients was represented by 31 patients who developed 49 culture-proved<br />

infections: 21 urinary origin, 8 pulmonary (1 Pneumocytis and 2 Aspergillosis),<br />

5 brain, 5 bacteremia and 2 other. The infected cohort (n�31) had<br />

similar demographs, but also similar outcome compared to our control non<br />

infected glioblastoma treated patients cohort (n�109) with a 2 year<br />

survival <strong>of</strong> 40.8% vs 50.2% (p�). The median CD4 lymphocytes level was<br />

260, but clearly infections were associated with less than 200 CD4<br />

lymphocytes. During their infection, 20 patients had a CD4 level less than<br />

200, compared with 11 patients with more than 200 CD4 lymphocytes<br />

count. Conclusions: CD4 lymphocytopenia less than 200 is associated with<br />

increase in number <strong>of</strong> documented culture-proven infections in patients<br />

treated with temozolomide, but the survival is not altered by the infection.<br />

TPS2104 General Poster Session (Board #20C), Sat, 1:15 PM-5:15 PM<br />

A phase III randomized controlled trial <strong>of</strong> short-course radiotherapy with or<br />

without concomitant and adjuvant temozolomide in elderly patients with<br />

glioblastoma (NCIC CTG CE.6, EORTC 26062-22061, TROG 08.02,<br />

NCT00482677). Presenting Author: James R. Perry, Sunnybrook Health<br />

Sciences Centre, Toronto, ON, Canada<br />

Background: The EORTC (26981-22981)/NCIC CTG (CE.3) RCT in newly<br />

diagnosed GBM found improved survival with concomitant and adjuvant<br />

temozolomide (TMZ) added to radiotherapy (RT). Study pts were 18-71<br />

(median 56) years; however a sub-group analysis noted a trend <strong>of</strong><br />

decreasing benefit from the addition <strong>of</strong> TMZ with increasing age, such that<br />

for age 65-71, the hazard ratio <strong>of</strong> 0.8 did not reach statistical significance<br />

(p�0.340). Recent RCTs in elderly GBM found improved survival with RT<br />

compared to supportive care alone and detected non-inferiority <strong>of</strong> 40 Gy/15<br />

vs. a 60 Gy/30 RT regimen. Based upon these results short-course<br />

hyp<strong>of</strong>ractionated RT is <strong>of</strong>ten recommended for elderly pts. However,<br />

whether the addition <strong>of</strong> TMZ to RT confers a survival advantage in elderly<br />

pts remains unanswered. Methods: Patients �65 yrs <strong>of</strong> age with histologically<br />

confirmed newly diagnosed glioblastoma, ECOG 0-2, are randomized<br />

1:1 to receive 40Gy/15 RT vs. 40Gy/15 RT with 3 weeks <strong>of</strong> concomitant<br />

temozolomide plus monthly adjuvant TMZ until progression or 12 cycles.<br />

Stratification is by centre, age (65-70, 71-75, or 76�), ECOG 0,1 vs 2, and<br />

biopsy vs resection. For 90% power to detect a 25% reduction in the<br />

primary outcome <strong>of</strong> overall survival (increased MST from 6 to 8 months)<br />

between arms, using a two-sided 5% alpha, a minimum <strong>of</strong> 520 deaths<br />

must be observed prior to analysis; total sample size is 560 patients. The<br />

trial is open in Canada (NCIC CTG), Europe (EORTC), Australia and New<br />

Zealand (TROG), and Japan. As <strong>of</strong> Jan 25, 2012, 361 (65%) <strong>of</strong> the target<br />

560 pts were randomized (147 Canada, 144 Europe, 64 Australasia, 6<br />

Japan). Median age <strong>of</strong> randomized patients is 73 (65-88) years. A planned<br />

futility analysis after 120 events by the independent DSMB resulted in a<br />

recommendation that the trial continue.Accrual is expected to be complete<br />

in 2013. A comprehensive molecular companion analysis, including<br />

MGMT promoter methylation, is planned.<br />

TPS2103 General Poster Session (Board #20B), Sat, 1:15 PM-5:15 PM<br />

REACT: A phase II study <strong>of</strong> rindopepimut (CDX-110) plus bevacizumab<br />

(BV) in relapsed glioblastoma (GB). Presenting Author: David A. Reardon,<br />

Dana-Farber Cancer Institute, Boston, MA<br />

Background: EGFRvIII is a constitutively active tumorigenic deletion mutation<br />

<strong>of</strong> EGFR. It is expressed in ~30% <strong>of</strong> primary GB where it is linked to<br />

poor long-term survival (Pelloski 2007). The investigational vaccine rindopepimut<br />

consists <strong>of</strong> the unique EGFRvIII peptide sequence conjugated to<br />

keyhole limpet hemocyanin (KLH), delivered intradermally (500ug with<br />

150ug GM-CSF as an adjuvant). Remarkably consistent and promising<br />

results across 3 phase II studies in newly diagnosed, resected EGFRvIII�<br />

GB (Lai 2011) represent a statistically significant improvement over a<br />

historical control cohort matched for major eligibility criteria (median<br />

overall survival [OS] � 24.4 - 24.6 vs. 15.2 months from diagnosis [m] and<br />

median progression-free survival [PFS] � 12.3 - 15.3 vs. 6.4 m). ACT IV, a<br />

phase III trial in this population, is ongoing. The immunosuppressive<br />

influence <strong>of</strong> residual/advanced GB presents a challenge to activation <strong>of</strong><br />

efficacious antitumor immune responses. Anecdotal evidence (compassionate<br />

use cases, Sampson 2008) suggests that rindopepimut may induce<br />

specific immune responses and regression in multifocal and bulky residual<br />

tumors. Rindopepimut with BV, which inhibits VEGF and its immunosuppressive<br />

properties (including impaired maturation <strong>of</strong> dendritic cells and<br />

disruption <strong>of</strong> tumoral T cell infiltration [Johnson 2007, Shrimali 2010])<br />

may further optimize EGFRvIII-specific immune response and antitumor<br />

activity. Methods: ReACT is a Phase II study <strong>of</strong> rindopepimut plus BV in<br />

patients (pts) with 1st or 2nd relapse <strong>of</strong> EGFRvIII� GB. BV-naïve pts will be<br />

enrolled to Group 1 (n�70: randomized 1:1 to BV plus either rindopepimut/<br />

GM-CSF or control injection [low-dose KLH]) while BV-refractory patients<br />

will enter Group 2 (n�25: to receive BV plus open-label rindopepimut/GM-<br />

CSF). Concurrent with BV (10 mg/kg, q 2 wks), blinded treatment or<br />

open-label vaccine is given in priming phase (days 1, 15 and 29), then<br />

monthly until PD. Tumor response is assessed every 8 weeks, and patients<br />

are followed for survival after PD. Objectives are PFS at 6 months (primary),<br />

objective response rate, PFS, OS, safety, immunogenicity and elimination<br />

<strong>of</strong> EGFRvIII. ReACT opened to accrual in December 2011 (NCT01498328).<br />

TPS2105 General Poster Session (Board #20D), Sat, 1:15 PM-5:15 PM<br />

Bevacizumab plus radiotherapy for elderly patients with glioblastoma<br />

(ARTE). Presenting Author: Ghazaleh Tabatabai, Department <strong>of</strong> Neurology,<br />

University Hospital Zurich, Zurich, Switzerland<br />

Background: The standard <strong>of</strong> care for patients with newly diagnosed<br />

glioblastomas after surgical resection is radiotherapy with concomitant and<br />

adjuvant temozolomide chemotherapy. Yet, inclusion into the pivotal trial<br />

was limited to patients up to the age <strong>of</strong> 70, and subgroup analyses<br />

suggested that older patients did not gain benefit from combined modality<br />

treatment. Further, the efficacy <strong>of</strong> radiotherapy has been confirmed in a<br />

randomized trial comparing best supportive care versus radiotherapy alone.<br />

Of note, hyp<strong>of</strong>ractionated radiotherapy is equieffective in patients aged<br />

65-70 years and more. Two randomized trials in elderly patients (NOA-08,<br />

Nordic Trial) indicated smilar efficacy <strong>of</strong> primary temozolomide chemotherapy<br />

alone compared with radiotherapy alone. Combined radiochemotherapy<br />

is compared with radiotherapy alone in an ongoing NCIC-CTG<br />

EORTC TROG Japanese group trial 26062-22061. Thus, radiotherapy<br />

alone is the standard <strong>of</strong> care for newly diagnosed glioblastoma <strong>of</strong> elderly<br />

patients. These clinical data justify the exploration <strong>of</strong> new, temozolomidefree<br />

first-line treatment strategies in elderly patients. Methods: The ARTE<br />

trial will therefore investigate bevacizumab when added to a short course <strong>of</strong><br />

radiotherapy and bevacizumab maintenance therapy until progression. The<br />

translational research program shall include blood and urine biomarker<br />

analysis and FET-PET in addition to MRI for monitoring the course <strong>of</strong> the<br />

disease. This trial is an explorative randomized, non-comparative phase II<br />

trial aiming at recruiting 60 patients in Switzerland. Elderly patients (� 65)<br />

will be randomized 2:1 either to the experimental arm (bevacizumab plus<br />

radiotherapy) or the standard arm (radiotherapy). The primary endpoint is<br />

median overall survival. Secondary endpoints include overall survival at 6<br />

months, overall survival at 12 months, median progression-free survival,<br />

progression-free survival at 6 months, median time to treatment failure.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


TPS2106 General Poster Session (Board #20E), Sat, 1:15 PM-5:15 PM<br />

Phase III trial <strong>of</strong> tumor-treating fields (TTFields) together with temozolomide<br />

compared with temozolomide (TMZ) alone in patients with newly<br />

diagnosed glioblastoma multiforme (NCT00916409). Presenting Author:<br />

Santosh Kesari, University <strong>of</strong> California, San Diego, San Diego, CA<br />

Background: TTFields therapy is a novel, non-invasive, anti-mitotic treatment,<br />

inhibiting tumor growth by interfering with the metaphase to<br />

anaphase transition during mitosis (Lee et al., 2011). TTF therapy was<br />

recently approved by the FDA for recurrent GBM following a prospective,<br />

randomized trial. Preclinical studies have shown TTFields and TMZ to have<br />

an additive inhibitory effect on glioma cell proliferation. The design for the<br />

current study was supported by results from a pilot study with TTFields<br />

therapy and TMZ in patients with newly diagnosed GBM demonstrating<br />

favorable safety, tolerability and promising efficacy (Kirson et al., 2009).<br />

Methods: The hypothesis <strong>of</strong> the current study is that the addition <strong>of</strong><br />

TTFields therapy to maintenance TMZ will increase progression free<br />

survival (PFS) <strong>of</strong> newly diagnosed GBM patients. Secondary endpoints<br />

include overall survival (OS), radiological response, quality <strong>of</strong> life and<br />

safety. Eligible patients will be randomized (2:1) to continuous TTFields<br />

therapy (200 kHz) in combination with maintenance TMZ versus maintenance<br />

TMZ alone. Maintenance TMZ will be administered as per the Stupp<br />

Protocol for 6 courses (Stupp et al., 2005). TTFields therapy may be<br />

administered for up to 24 months. Patients will be stratified based on:<br />

extent <strong>of</strong> resection and MGMT methylation status. Eligibility criteria<br />

include: pathological diagnosis <strong>of</strong> GBM; Karn<strong>of</strong>sky � 70; previous chemoradiotherapy<br />

per Stupp Protocol; no progressive disease at baseline.<br />

Patients with infratentorial tumor or implanted electronic devices are<br />

excluded. The sample size <strong>of</strong> 700 patients will have 80% power to detect a<br />

2 month (9 vs 7 months) improvement in PFS using log-rank test with an<br />

overall 5% 2-sided type I error. The trial is also powered to detect a 4.5<br />

month increase in median OS in NovoTTF-100A/TMZ patients. Patients<br />

will be followed clinically monthly and with bimonthly MRI to assess tumor<br />

progression and additional endpoints. 221 patients have been enrolled at<br />

the time <strong>of</strong> abstract submission. The DMC last reviewed the trial in October<br />

2010 and recommended that the trial continue as designed.<br />

Central Nervous System Tumors<br />

141s<br />

TPS2107 General Poster Session (Board #20F), Sat, 1:15 PM-5:15 PM<br />

A randomized, double-blind, controlled phase IIb study <strong>of</strong> the safety and<br />

efficacy <strong>of</strong> ICT-107 in newly diagnosed patients with glioblastoma multiforme<br />

following resection and chemoradiation. Presenting Author: Elma S.<br />

Hawkins, Immunocellular Therapeutics, Ltd., Woodland Hills, CA<br />

Background: Tumor stem cells have been correlated with recurrence and<br />

clinical outcome in glioblastoma multiforme (GBM). ICT-107 is an autologous<br />

vaccine consisting <strong>of</strong> the patient’s dendritic cells pulsed with 6<br />

synthetic peptide CTL epitopes targeting the GBM tumor and tumor-stem<br />

cell associated antigens MAGE-1, HER-2, AIM-2, TRP-2, gp100 and<br />

IL-13R�2. Phase I results showed a good safety pr<strong>of</strong>ile and interesting<br />

clinical potential (ASCO, 2010, abs#2097 and ASCO, 2011, abs#2042)<br />

in 16 newly diagnosed GBM patients with a median progression-free<br />

survival (PFS) <strong>of</strong> 16.9 months (measured from surgery) and a median<br />

overall survival (OS) <strong>of</strong> 38.4 months. Methods: In this Phase II study eligible<br />

patients have newly diagnosed GBM and complete surgical resection or<br />

minimum residual tumor � 1cm3 , are HLA-A1 and/or HLA-A2 positive,<br />

older than 18, have Karn<strong>of</strong>sky Performance Score (KPS) <strong>of</strong> � 70% and<br />

have adequate hematologic and chemistry parameters. Patients with a<br />

serious immune or autoimmune disorder or other systemic disease are<br />

excluded. Patients undergo apheresis to isolate peripheral blood mononuclear<br />

cells (PBMCs) to be used for preparation <strong>of</strong> study treatment (ICT-107<br />

and Control). Pre-study treatment consists <strong>of</strong> 6 weeks <strong>of</strong> concurrent<br />

temozolomide (TMZ) and radiation. After stratification by site and age,<br />

patients are randomized 2:1 to receive either ICT-107 or its matching<br />

control (autologous, unpulsed dendritic cells). Patients then receive<br />

induction ICT-107 or control once a week for four weeks. All patients<br />

subsequently receive maintenance TMZ for 5 days per month for 12<br />

months. Booster vaccinations occur at Cycles 1, 3, 6 and 10, and every six<br />

months thereafter. The primary endpoint is OS and secondary endpoints<br />

include PFS, rates <strong>of</strong> OS and PFS at 6 months after surgery and every 3<br />

months thereafter, safety and tolerability <strong>of</strong> ICT-107, immune response to<br />

ICT-107 and predictors <strong>of</strong> response. 120 patients have been enrolled in<br />

this ongoing trial. It is expected that approximately 200 patients will be<br />

enrolled for screening with the intention to randomize at least 102 patients.<br />

The trial significance is alpha�0.025 one-sided.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


142s Developmental Therapeutics—<strong>Clinical</strong> Pharmacology and Immunotherapy<br />

2500^ Oral Abstract Session, Mon, 3:00 PM-6:00 PM<br />

Pharmacodynamic assessment <strong>of</strong> INCB024360, an inhibitor <strong>of</strong> indoleamine<br />

2,3-dioxygenase 1 (IDO1), in advanced cancer patients. Presenting<br />

Author: Robert Charles Newton, Incyte Corporation, Wilmington, DE<br />

Background: The ineffectiveness <strong>of</strong> the immune system to control tumor<br />

growth is, in part, a result <strong>of</strong> immunosuppression imposed by negative<br />

regulatory mechanisms. Indoleamine 2,3-dioxygenase 1 (IDO1), an enzyme<br />

that catabolizes the first and rate-limiting step in the degradation <strong>of</strong><br />

the essential amino acid tryptophan (Trp) to kynurenine (Kyn), has been<br />

shown preclinically to play an important role in tumor-mediated immunosuppression.<br />

In cancer patients (pts), elevated IDO1 levels are associated<br />

with poor prognosis and shortened survival in a number <strong>of</strong> tumor types.<br />

Here we describe the pharmacodynamic (PD) assessment <strong>of</strong> INCB024360,<br />

a novel inhibitor <strong>of</strong> IDO1. Methods: Plasma samples were obtained from<br />

consented pts in study INCB 24360-101, a phase I dose-escalation study<br />

in patients with advanced malignancies. Trp and Kyn levels in plasma were<br />

determined by LC/MS/MS. IDO1 activity in activated peripheral blood cells<br />

was also monitored. Results: Using anti-IDO1 specific antisera and archived<br />

tumor samples, we found IDO1 expression in various human tumors,<br />

including ovarian, colorectal, breast and prostate. Consistent with this<br />

result, higher Kyn/Trp ratios (1.5-3.4 fold above healthy volunteers) were<br />

detected in archived plasma samples from pts, indicative <strong>of</strong> higher IDO1<br />

activity in cancer pts. To date, 23 pts have been treated with the selective<br />

IDO1 inhibitor INCB024360. When plasma samples from patients were<br />

collected pre- and post-INCB024360 treatment, significant dosedependent<br />

reductions in plasma Kyn/Trp ratios and Kyn levels were<br />

detected. As an additional biomarker measurement, whole blood samples<br />

collected from pts at various times after dosing were stimulated ex vivo with<br />

interferon-� and lipopolysaccharide to increase IDO1 activity and also<br />

showed dose-dependent decreases in IDO activity. With the current dose<br />

regimens and assays we have successfully achieved sustained inhibition <strong>of</strong><br />

�90% at a well tolerated dose <strong>of</strong> INCB024360. Conclusions: This is the<br />

first demonstration <strong>of</strong> PD activity <strong>of</strong> an IDO1-specific inhibitor in cancer<br />

pts. Our study also confirms that IDO1 is frequently activated in cancer pts.<br />

The methods described will be used to establish a phase II dose.<br />

2502 Oral Abstract Session, Mon, 3:00 PM-6:00 PM<br />

Isolation <strong>of</strong> human anti-MICA antibody from cancer patients responding to<br />

immunotherapies. Presenting Author: Kenneth F. May, Dana-Farber Cancer<br />

Institute, Boston, MA<br />

Background: Immunotherapy is a promising modality for the treatment <strong>of</strong><br />

cancer, and elucidating the mechanism <strong>of</strong> action is crucial to guiding<br />

patient selection and developing future immunotherapeutic strategies.<br />

Engagement <strong>of</strong> the immune molecule NKG2D by the cancer antigen MICA<br />

is important for immune surveillance <strong>of</strong> many cancers. Tumors can evade<br />

immune surveillance by shedding cell surface MICA, which leads to<br />

dampening <strong>of</strong> anti-tumor immunity. Investigations by our laboratory revealed<br />

that patients responding to immunotherapy can mount antibody<br />

responses targeting MICA, which permits re-engagement <strong>of</strong> immunity.<br />

Methods: Patients with advanced cancer treated at our institution with<br />

immunotherapies (tumor cell vaccination, ipilimumab) were identified,<br />

including several with long-term clinical remissions. Plasma was analyzed<br />

for MICA reactivity to three distinct MICA alleles, and rare memory B cells<br />

reactive to MICA were isolated using tetramerized MICA protein. Immunoglobulin<br />

heavy and light chain variable domains were sequenced with single<br />

cell RT-PCR to generate recombinant monoclonal antibody. Results: Plasma<br />

from patients treated with immunotherapy revealed considerable interpatient<br />

variation in reactivity to MICA. Patients with marked plasma<br />

reactivity (including several with sustained clinical remissions) were<br />

selected for isolation <strong>of</strong> rare memory B cells reactive to MICA. From these<br />

cells, we have generated a recombinant fully-human MICA-reactive monoclonal<br />

antibody that exhibits binding to a variety <strong>of</strong> MICA alleles. Conclusions:<br />

We have developed novel methods to analyze anti-MICA antibody responses<br />

and isolate rare memory B cells from cancer patients who gained significant<br />

clinical benefit from immunotherapy. This has facilitated the generation <strong>of</strong><br />

fully human recombinant anti-MICA monoclonal antibody. To our knowledge,<br />

this is the first example <strong>of</strong> a specific antibody reactive to a cancer<br />

antigen isolated from a cancer patient responding to immunotherapy. As<br />

these antibody responses likely played a role in tumor destruction, these<br />

results inform the development <strong>of</strong> new antibody-based immunotherapies<br />

targeting the NKG2D-MICA pathway.<br />

2501 Oral Abstract Session, Mon, 3:00 PM-6:00 PM<br />

A phase I study <strong>of</strong> 1-methyl-D-tryptophan in patients with advanced<br />

malignancies. Presenting Author: Hatem Hussein Soliman, H. Lee M<strong>of</strong>fitt<br />

Cancer Center & Research Institute, Tampa, FL<br />

Background: The indoleamine 2, 3 dioxygenase pathway (IDO) can create<br />

immune suppression and unresponsiveness to tumor antigens in tumorbearing<br />

hosts. 1-methyl-D-tryptophan (1-MT) is an oral inhibitor <strong>of</strong> the IDO<br />

pathway, currently in development as an immune adjuvant for anti-tumor<br />

vaccines and chemotherapy. The primary goal <strong>of</strong> this trial is to determine<br />

the MTD and toxicity for oral 1-MT. Secondary endpoints include response<br />

rates, PK, serum tryptophan catabolites, circulating T-reg cells, and C<br />

reactive protein (CRP) levels. Methods: This phase I study utilizes a 3�3<br />

design comprised <strong>of</strong> ten dose levels (200, 300,400,600,800,mg QD, 600,<br />

800,1200,1600,2000mg BID). Inclusion criteria: patients with measurable<br />

metastatic solid malignancy, age �18, life expectancy �4 months,<br />

and adequate organ/marrow function. Exclusion criteria: chemotherapy in<br />

the past 3 weeks, untreated brain metastases, active autoimmune disease,<br />

or malabsorptive GI disease. Continuous toxicity monitoring early stopping<br />

rules were used. Results: At submission, 48 out <strong>of</strong> the planned 50 patients<br />

were accrued. MTD was not reached at 2000mg BID. At 200mg QD dose, 3<br />

patients previously treated with active immunotherapy (ipilimumab, n�2,<br />

CD40-mAb, n�1) developed an autoimmune hypophysitis. Since IDO is<br />

closely linked to both CTLA-4 and CD40/CD40L pathways this was<br />

considered on-target effect. Subsequently, patients with prior immunotherapy<br />

were excluded and no additional treatment related G3-5 adverse<br />

events were observed. Five patients showed SD�6 months (2 melanoma, 2<br />

sarcoma 1 colon) as well as some mixed responses were seen in some<br />

patients including regression <strong>of</strong> a liver visceral metastases. 1-MT plasma<br />

PK AUC and Cmax were proportional with dose. Cmax (~40 �M at 2000mg<br />

BID) is at 2.9 hours and the t1/2 is at 10 hours. Elevations in CRP levels<br />

and declines in T-reg cell counts were seen across multiple dose levels.<br />

Additional correlative data analysis is ongoing. Conclusions: The treatment<br />

was well tolerated and orally bioavailable. Biologic activity in terms <strong>of</strong><br />

prolonged disease stabilization, induction <strong>of</strong> hypophysitis, and changes in<br />

T-reg levels and CRP were observed. Trials combining 1-MT with docetaxel<br />

and a dendritic cell vaccine are ongoing.<br />

2503^ Oral Abstract Session, Mon, 3:00 PM-6:00 PM<br />

Durability <strong>of</strong> complete remission by moxetumomab pasudotox (HA22 or<br />

CAT-8015) assessed by clone-specific real-time quantitative PCR (RQ-<br />

PCR). Presenting Author: Robert J. Kreitman, Laboratory <strong>of</strong> Molecular<br />

Biology, NCI, NIH, Bethesda, MD<br />

Background: The anti-CD22 recombinant immunotoxin moxetumomab<br />

pasudotox, also known as HA22 or CAT-8015, was recently reported in<br />

phase I testing to achieve complete remissions (CRs) in 13 (46%) <strong>of</strong> 28<br />

patients with relapsed/refractory hairy cell leukemia (HCL); 3 <strong>of</strong> 13<br />

patients have relapsed. Methods: To complete this trial, 20 additional<br />

patients received the highest dose level (50 �g/Kg every other day x 3<br />

doses); none <strong>of</strong> the 48 HCL patients had dose-limiting toxicity (DLT).<br />

Results: Of the first 42 patients with �6 mo <strong>of</strong> follow up <strong>of</strong>f-treatment, 23<br />

(55%) had CRs, with an overall response rate <strong>of</strong> 88%. Of the 23 CRs, 21<br />

were evaluable for minimal residual disease (MRD) using flow cytometry <strong>of</strong><br />

blood and immunohistochemistry <strong>of</strong> the bone marrow biopsy, and 17<br />

(81%) were negative. Of these 17 patients, 11 (65%) were negative by<br />

bone marrow aspirate (BMA) flow cytometry. PCR using consensus primers<br />

for the heavy chain immunoglobulin (IgH) rearrangement was less specific<br />

than flow cytometry <strong>of</strong> blood, since IgH rearrangements <strong>of</strong> normal B cells,<br />

which recovered rapidly after immunotoxin treatment, were also amplified.<br />

For better MRD detection in blood, patient IgH sequences were cloned and<br />

sequence specific primers and probes designed for real-time quantitative<br />

PCR (RQ-PCR). RQ-PCR <strong>of</strong> blood was negative in 6 (100%) <strong>of</strong> 6 patients<br />

achieving flow-negativity in both blood and BMA and positive in 3 (100%)<br />

<strong>of</strong> 3 patients flow-negative in blood but not BMA (p�0.01). No relapses<br />

from CR have been observed in 10 patients who became RQ-PCR-negative<br />

in blood or flow-negative in BMA, with 5-38 (median 11) mo <strong>of</strong> follow-up.<br />

Conclusions: We conclude that clone-specific RQ-PCR is the most sensitive<br />

blood test for MRD in our HCL patients after moxetumomab pasudotox, and<br />

could be used to assess the possibility <strong>of</strong> long-term molecular remissions.<br />

We believe these results, including durable CRs without DLT, support a<br />

pivotal trial in which moxetumomab pasudotox is compared with alternative<br />

therapy. Note: this summary contains investigator reported data. This study<br />

was funded by MedImmune, LLC, and supported by NCI’s Intramural<br />

Research Program and the Hairy Cell Leukemia Research Foundation.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


Developmental Therapeutics—<strong>Clinical</strong> Pharmacology and Immunotherapy<br />

2504 Oral Abstract Session, Mon, 3:00 PM-6:00 PM<br />

A phase I study <strong>of</strong> EpCAM/CD3-bispecific antibody (MT110) in patients<br />

with advanced solid tumors. Presenting Author: Walter M. Fiedler, Medical<br />

Clinic II, University Medical Center Hamburg-Eppendorf, Center <strong>of</strong> Oncology<br />

(Hubertus Wald Tumorzentrum), Hamburg, Germany<br />

Background: MT110 is a bispecific T cell engaging antibody construct<br />

(BiTE) that directs CD3 expressing T cells to kill target cells that express<br />

EpCAM, which is widely found on solid tumors. Methods: Safety, tolerability,<br />

pharmacokinetics and anti-tumor activity <strong>of</strong> MT110 in patients (pts)<br />

with advanced solid tumors were studied using a 3�3 design. Continuous<br />

IV infusions for at least 28 days <strong>of</strong> doses ranging from 1 to 48 �g/d have<br />

been tested. Stepwise intra-patient dose escalations within cycle 1 or at<br />

start <strong>of</strong> cycle 2 have been explored. Results: 51 pts (6 gastric cancers, 32<br />

CRC, 2 SCLC, 5 NSCLC, 3 HRPC, 1 ovarian cancer) have been treated in<br />

12 dose cohorts. The maximally administered dose includes day 1 dose <strong>of</strong> 3<br />

�g/d escalated up to 48 �g/d. The maximum tolerable dose (MTD) has not<br />

yet been established. Diarrhea and elevations in liver function tests have<br />

been reported as dose limiting toxicity (DLT) for individual patients.<br />

Transaminase elevations were fully reversible and mitigated by dexamethasone<br />

at initiation or escalation <strong>of</strong> dose. Other adverse events <strong>of</strong> grade 3 or<br />

higher included transient lymphopenia, increase in lipase or amylase,<br />

nausea, vomiting, and hypoalbuminaemia. No CNS safety signal was<br />

observed. The mean serum half-life <strong>of</strong> MT110 was 4.5 hours. Mean steady<br />

state serum concentrations <strong>of</strong> MT110 were linear with dose across<br />

treatment cycles. At 48 �g/d, the mean Css was 3.2 ng/mL. This compares<br />

to preclinical EC90 <strong>of</strong> 1.35 – 3.85 ng/ml in CRC cell lines. Sixteen pts who<br />

received a dose <strong>of</strong> � 24 �g/d were evaluable for anti-tumor response. Best<br />

response <strong>of</strong> SD was reported in 38%; median duration was 155 days (95%<br />

CI 92; 161). Anti-tumor activity was demonstrated on biopsy <strong>of</strong> liver<br />

metastases in one patient. Following at least 1 week <strong>of</strong> MT110 treatment at<br />

48 �g/d, a decrease in circulating tumor cells was observed with a median<br />

nadir 74% below baseline. Conclusions: The use <strong>of</strong> stepwise dose escalation<br />

and concomitant dexamethasone resulted in improved tolerability. A<br />

dose <strong>of</strong> 48 �g/d appears tolerable. Tumor biopsy evaluation on treatment<br />

and pharmacodynamic evidence <strong>of</strong> anti-tumor activity are encouraging.<br />

Additional dose cohorts will seek to identify the recommended phase II<br />

dose and further evidence <strong>of</strong> anti-tumor activity.<br />

2506 Oral Abstract Session, Mon, 3:00 PM-6:00 PM<br />

WT1-targeted dendritic cell vaccination as a postremission treatment to<br />

prevent or delay relapse in acute myeloid leukemia. Presenting Author: Zwi<br />

N. Berneman, Antwerp University Hospital, Edegem, Belgium<br />

Background: Vaccination with tumor antigen-loaded dendritic cells (DC)<br />

holds promise for the adjuvant treatment <strong>of</strong> cancer. Methods: In a phase I/II<br />

trial, we investigated the effect <strong>of</strong> autologous DC vaccination in 17 patients<br />

with acute myeloid leukemia (AML) in remission but at very high risk <strong>of</strong> full<br />

relapse. Wilms’ tumor 1 protein (WT1) was chosen as immunotherapeutic<br />

target and introduced into the DC by mRNA electroporation. We are<br />

continuing a phase II trial, which is still recruiting. Results: Two out <strong>of</strong> 3<br />

patients, who were in partial remission with chemotherapy-refractory<br />

disease, were brought into complete remission following 4 biweekly<br />

intradermal injections <strong>of</strong> WT1 mRNA-electroporated DC. In those 2<br />

patients as well as in 6 other patients who were in complete remission but<br />

who had molecularly demonstrable residual disease, there was a return to<br />

normal <strong>of</strong> the AML-associated WT1 mRNA tumor marker following DC<br />

vaccination, compatible with reaching clinical and molecular remission in<br />

8/17 patients. Among the 8 responders, there have been 2 relapses and 2<br />

deaths. Of the 9 non-responders, 8 have relapsed and 7 have died. Of the 2<br />

patients in partial remission who were brought into complete remission by<br />

DC vaccination, 1 has died following relapse. Median overall survival was 6<br />

months in non-responders and 52 months in responders (p�0.0007).<br />

Median relapse-free survival was 3 months in non-responders as compared<br />

to 47 months in responders (p�0.0001). <strong>Clinical</strong> responses overall were<br />

correlated with elevated levels <strong>of</strong> activated natural killer (NK) cells<br />

post-vaccination. Long-term clinical responses, lasting for at least 3 years,<br />

were significantly correlated with an increase in polyepitope WT1-specific<br />

tetramer� CD8� T-cell frequencies. Conclusions: DC-based immunotherapy<br />

elicits both innate (NK) and adaptive (T cells) cellular responses<br />

correlated with clinical benefit. WT1 mRNA-transfected DC emerge as a<br />

feasible and effective strategy to control residual disease in AML, in<br />

particular as a post-remission treatment to prevent full relapse.<br />

143s<br />

2505 Oral Abstract Session, Mon, 3:00 PM-6:00 PM<br />

Anti-CD3 immunotoxin to induce remissions in cutaneous T-cell lymphoma<br />

patients. Presenting Author: Arthur E. Frankel, Scott and White Hospital,<br />

Temple, TX<br />

Background: The anti-CD3 immunotoxin, AdmDT390bisFv(UCHT1), composed<br />

<strong>of</strong> the catalytic and translocation domains <strong>of</strong> diphtheria toxin<br />

(DT390) fused to two anti-CD3 sFvs showed selective cytotoxicity to normal<br />

and malignant human T cells in vitro and in vivo and was prepared for a<br />

phase I clinical study (Woo, Protein Exp Purif 58, 1, 2008). FDA approval<br />

(BB IND#100712), and IRB approval was obtained. Methods: Nineteen T<br />

cell lymphoma patients were screened, and 12 patients treated. Median<br />

age was 62 years (range, 39-84 years) with 6 males and 6 females. Disease<br />

was previously treated with one (6), two (3), three (1) or four (2) systemic<br />

therapies. One patient had stage IV PTCL and the other patients had CTCL<br />

(stage IB in 5, IIB in 4, IIIB in 1, and IV in 1). Six patients were treated with<br />

2.5�g/kg x 8; five patients were treated with 5�g/kgx5-8andonepatient<br />

treated with 7.5�g/kg x 8. Results: Drug-related toxicities were mild in<br />

11/12 patients and were not dose dependent with transient fevers, chills,<br />

lymphopenia, transaminasemia, CMV and/or EBV viremia, and hypoalbuminemia.<br />

One 69 year old male with pre-existing heart failure treated at the<br />

5�g/kg x 5 suffered severe vascular leak syndrome and heart failure.<br />

Immunotoxin Cmax was 1 – 60ng/mL, and mean half-life was 42min. Four<br />

patients had pre-treatment anti-immunotoxin titers �10�g/mL, and the<br />

others had lower pre-treatment antibody levels. All evaluable patients<br />

except two had increases <strong>of</strong> antibody titer between day 12 to 30 <strong>of</strong> 4 –<br />

2000-fold. Among 10 evaluable patients, there were 2 complete remissions<br />

(CRs) <strong>of</strong> 17 and 41� months durations and 4 partial remissions (PRs)<br />

<strong>of</strong> 1� -32�months. Conclusions: The immunotoxin has exciting clinical<br />

efficacy in this heavily pretreated group <strong>of</strong> patients. Dose escalation is<br />

proceeding.<br />

2507 Oral Abstract Session, Mon, 3:00 PM-6:00 PM<br />

Phase IB study on combined intradermal (ID) and intravenous (IV)<br />

administration <strong>of</strong> autologous mRNA electroporated dendritic cells (DC) as a<br />

single-agent cellular immunotherapy or combined with ipilimumab. Presenting<br />

Author: Bart Neyns, UZ Brussel, Brussels, Belgium<br />

Background: Autologous monocyte-derived DC electroporated with synthetic<br />

mRNA encoding CD40 ligand, a constitutively active TLR4, and<br />

CD70 (TriMix-DC) have superior T-cell stimulatory capacity. In a pilot<br />

clinical trial, ID administration <strong>of</strong> TriMixDC-MEL (a mixture <strong>of</strong> TriMix-DC<br />

co-electroporated with mRNA encoding a fusion <strong>of</strong> DC.LAMP and 1 <strong>of</strong> 4<br />

melanoma associated antigens) was immunogenic but resulted in limited<br />

anti-tumor activity in patients (pts) with advanced melanoma. Ipilimumab<br />

(IPI) is a CTLA-4 blocking mAb with established activity in pts with<br />

advanced melanoma. Methods: TriMixDC-MEL by the IV and ID-route was<br />

investigated as a single-agent or in combination with IPI (10 mg/kg q3wks<br />

x4, allowing for maintenance with IPI-alone q12wks in pts PFS at �24<br />

wks). Ratio <strong>of</strong> ID/IV administered DC: Cohort-1: 20.106 /4.106 DC [2pts],<br />

-2: 12.106 /12.106 DC [3pts], -3: 4.106 /20.106 [6pts], and -4: 0/24.106 DC [4pts]; DC were administered 4x q2w, and a 5th administration on w16.<br />

in cohort-5, DC (4.106-ID/20.106-IV) were first administered alone and 2w<br />

thereafter in combination with IPI for a total <strong>of</strong> 4 administrations [6pts].<br />

Results: Local skin reactions (gr1-2) were observed in all pts receiving DC<br />

ID, flu-like symptoms (� gr2) were observed in 12/21 pts, post IV-infusion<br />

chills (gr2) in 7/21 pts. Cytokines release (including a �2-fold rise in the<br />

serum levels <strong>of</strong> IL-1RA, IL-6, IL-8, IL-17, G-CSF, IFN-g, MIP-1a, MIP-1b,<br />

TNF-a) was documented during chills. Best objective tumor response: 2x<br />

PR � 2x CR (by RECIST) out <strong>of</strong> 15 pts (27%) treated with DC-only (ongoing<br />

after 10�, 14�, 19�, and 20� mths) and 3 PR out <strong>of</strong> 6 pts (all stage<br />

IV-M1c) treated with DC�IPI (ongoing after 5�,5�,6�mths). Conclusions:<br />

ID/IV-administration <strong>of</strong> TriMixDC-MEL as a single-agent cellular immunotherapy<br />

or combined with IPI is associated with distinct but manageable<br />

side-effects and has clinical activity against pretreated advanced melanoma.<br />

Activity compares favorably with TriMixDC-MEL by ID-administrationonly.<br />

Combined ID/IV administration <strong>of</strong> TriMixDC-MEL with IPI is currently<br />

under further evaluation in a phase II trial.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


144s Developmental Therapeutics—<strong>Clinical</strong> Pharmacology and Immunotherapy<br />

2508 Oral Abstract Session, Mon, 3:00 PM-6:00 PM<br />

From bench to bedside: The use <strong>of</strong> the li-Key technology to improve helper<br />

peptides for clinical use in cancer vaccines. Presenting Author: Timothy J.<br />

Vreeland, San Antonio Military Medical Center, Ft. Sam Houston, TX<br />

Background: Work involving peptide vaccines has shown that peptides<br />

containing MHC Class II epitopes, which elicit CD4� T cell responses, may<br />

play a role in potentiating an immune response. The Ii-Key peptide (amino<br />

acids 77-80 <strong>of</strong> the immune-regulatory Ii protein), when covalently linked to<br />

an MHC Class II epitope, can induce conformational change in the epitope<br />

binding groove, increasing CD4� T cell stimulation up to 250 fold. Here we<br />

present an update <strong>of</strong> results from clinical trials evaluating this novel<br />

technology in an adjuvant breast cancer vaccine targeting HER2/neu.<br />

Methods: We reviewed our trials investigating AE37, a hybrid peptide<br />

created by the addition <strong>of</strong> the Ii-Key peptide (LRMK) to AE36 (GVGSPYVS-<br />

RLLGICL), an MHC Class II-binding peptide from the intracellular domain<br />

<strong>of</strong> the HER2 protein. We have completed a phase I study and are currently<br />

conducting a randomized phase II trial <strong>of</strong> the AE37 peptide � GM-CSF in<br />

the adjuvant treatment <strong>of</strong> disease-free breast cancer patients with any level<br />

<strong>of</strong> HER2 expression (IHC 1-3� or FISH�1.2). Results: Phase I data showed<br />

the vaccine to be safe and effective in raising anti-HER2 immunity.<br />

Importantly, even the cohort <strong>of</strong> patients given AE37 without GM-CSF<br />

showed significant increases in both in vivo and in vitro immune responses.<br />

To date, we have enrolled 201 patients to our phase II trial (Vaccine<br />

(VG)�103, Control (CG)�98). Toxicity has been minimal (99% <strong>of</strong> local<br />

and systemic toxicities grade �2). VG patients have shown significant<br />

increases in both in vivo and in vitro responses to both AE36 and AE37,<br />

with consistently stronger responses to the AE37 hybrid peptide than the<br />

native AE36. With a median f/u <strong>of</strong> 22 months, Kaplan Meier projections<br />

estimate recurrence rates <strong>of</strong> 10.3% in the VG compared to 18% in the CG;<br />

a 43% risk reduction. Conclusions: The AE37 peptide vaccine appears to be<br />

effective in eliciting a strong immune response and possibly preventing<br />

breast cancer recurrence. These results provide an important pro<strong>of</strong> <strong>of</strong><br />

concept and suggest that additional studies evaluating Ii-Key hybrid<br />

peptide vaccines are warranted, whether in the field <strong>of</strong> immunotherapy or<br />

more traditional vaccines.<br />

2510 <strong>Clinical</strong> Science Symposium, Sat, 1:15 PM-2:45 PM<br />

PD-1/PD-L1 pathway as a target for cancer immunotherapy: Safety and<br />

clinical activity <strong>of</strong> BMS-936559, an anti-PD-L1 antibody, in patients with<br />

solid tumors. Presenting Author: Scott S. Tykodi, Fred Hutchinson Cancer<br />

Research Center, Seattle, WA<br />

Background: Blocking the interaction between programmed death-1 (PD-<br />

1), a co-inhibitory receptor expressed by activated T cells, and its ligand<br />

PD-L1 may enhance T-cell function. Here we describe the safety and<br />

activity from the first clinical study <strong>of</strong> BMS-936559 in patients (pts) with<br />

advanced solid tumors, further validating the importance <strong>of</strong> the PD-1/<br />

PD-L1 pathway as a target for cancer therapy. Methods: BMS-936559 was<br />

given Q2 wk IV in a 6-wk cycle at doses <strong>of</strong> 0.3�10.0 mg/kg during dose<br />

escalation and/or cohort expansion; pts could receive up to 16 cycles (48<br />

doses). Results: As <strong>of</strong> August 1, 2011, 162 pts with melanoma (MEL,<br />

n�53), non-small-cell lung (NSCLC, n�50), colorectal (n�18), renal cell<br />

(RCC, n�17), ovarian (n�17), and pancreatic (n�7) cancer were treated.<br />

Median therapy duration was 11 wks (max 99 wks). Common related<br />

adverse events (rAEs; �5% pts) included fatigue, diarrhea, infusion<br />

reaction, arthralgia, rash, and pruritus. The incidence <strong>of</strong> grade 3-4 rAEs<br />

was 8.6%. AEs <strong>of</strong> special interest included hypothyroidism, hepatitis,<br />

sarcoidosis, endophthalmitis, and myasthenia gravis; there were no drugrelated<br />

deaths. <strong>Clinical</strong> activity was observed in MEL, RCC, and NSCLC.<br />

Objective responses (ORs; RECIST 1.0) were observed in heavily pretreated<br />

pts. ORs were durable; among 16 pts with ORs, 7 had responses lasting �1<br />

yr. Two other pts had ongoing ORs with durations <strong>of</strong> 2.3 and 8.5 mo at data<br />

lock. Several pts had prolonged stable disease. Some pts had a persistent<br />

reduction in overall tumor burden in the presence <strong>of</strong> new lesions but were<br />

not categorized as responders. In NSCLC, ORs were observed irrespective<br />

<strong>of</strong> histology. Conclusions: BMS-936559 was active and generally well<br />

tolerated in pts with NSCLC, RCC, and MEL. In conjunction with data from<br />

anti-PD-1/BMS-936558 trials, this study concurrently validates the importance<br />

<strong>of</strong> the PD-1/PD-L1 pathway for cancer immunotherapy and supports<br />

further clinical development <strong>of</strong> anti-PD-1/PD-L1 directed therapy.<br />

Dose<br />

(mg/kg)<br />

MEL RCC NSCLC<br />

N a OR uPR b RR c (%) N a OR uPR b RR c (%) N a OR uPR b RR c (%)<br />

1 18 1 1 11 0 - - - 11 0 0 0<br />

3 17 5 1 35 0 - - - 13 1 0 8<br />

10 17 3 0 18 17 2 1 18 25 4 3 28<br />

a response evaluable pts<br />

b unconfirmed PR<br />

c response rate ([OR � uPR] � N)<br />

CRA2509 <strong>Clinical</strong> Science Symposium, Sat, 1:15 PM-2:45 PM<br />

Anti-PD-1 (BMS-936558, MDX-1106) in patients with advanced solid tumors:<br />

<strong>Clinical</strong> activity, safety, and a potential biomarker for response.<br />

Presenting Author: Suzanne Louise Topalian, The Johns Hopkins University<br />

School <strong>of</strong> Medicine, Baltimore, MD<br />

The full, final text <strong>of</strong> this abstract will be available at<br />

abstract.asco.org at 12:01 AM (EDT) on Saturday, June 2,<br />

2012, and in the <strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong> online<br />

supplement to the June 20, 2012, issue <strong>of</strong> Journal <strong>of</strong><br />

<strong>Clinical</strong> Oncology. Onsite at the <strong>Meeting</strong>, this abstract will<br />

be printed in the Saturday edition <strong>of</strong> ASCO Daily News.<br />

2511 <strong>Clinical</strong> Science Symposium, Sat, 1:15 PM-2:45 PM<br />

Costimulatory effect <strong>of</strong> agonistic 4-1BB antibody on proliferation and<br />

effector phenotype <strong>of</strong> tumor-infiltrating lymphocytes in melanoma. Presenting<br />

Author: Amod Sarnaik, H. Lee M<strong>of</strong>fitt Cancer Canter & Research<br />

Institute, Tampa, FL<br />

Background: Adoptive cell therapy with tumor-infiltrating lymphocytes (TIL)<br />

is a promising form <strong>of</strong> immunotherapy with a 20-30% durable response<br />

rate. Its widespread implementation is limited by the relatively long time<br />

needed to establish TIL in vitro and the lack <strong>of</strong> effective tumor reactivity.<br />

Our objective was to incorporate co-stimulation with 4-1BB agonism to<br />

optimize TIL growth preparation time and tumor reactivity to improve<br />

outcomes. Methods: 7 metastatic melanoma patients were consented to an<br />

IRB-approved tissue procurement protocol. For each specimen, TIL were<br />

propagated in vitro from 4 melanoma tumor fragments per condition with<br />

standard 6000 IU/mL <strong>of</strong> IL-2 and either 10 ug/mL <strong>of</strong> 4-1BB agonistic<br />

antibody (BMS-663513) or its isotype control. After 3 weeks, TIL were<br />

counted and phenotyped using flow cytometry. TIL were re-stimulated with<br />

HLA-matched melanomas and assessed for interferon-� (IFN�) release by<br />

ELISA. Statistical comparisons involved the 2-tailed, paired t test using<br />

GraphPad Prism s<strong>of</strong>tware. Results: 4-1BB treatment yielded a median<br />

4.3x10 7 TIL (range 1.1x10 7 -8.4x10 7 ) compared to a median 2.3x10 7<br />

cells in the control (range 1.5x10 6 -5.3x10 7 ,p� 0.009). This is clinically<br />

significant as the initial minimum target number for TIL generation is<br />

3x10 7 . On phenotypic analysis compared to control, 4-1BB treatment<br />

resulted in a 1.5X higher CD8 � cell numbers (p�0.02), a 6X lower CD4 �<br />

cell numbers (p�0.02), and 5.5X lower CD4 � CD25 � FoxP3 � Treg cell<br />

numbers (p�0.09). A paired sample was re-stimulated in triplicate with<br />

HLA-matched tumor target, and ELISA <strong>of</strong> the supernatant treated with<br />

4-1BB yielded 1.6 fold higher IFN� (p�0.05). A paired sample was then<br />

assayed for known markers <strong>of</strong> T cell cytolytic activity: EOMES and perforin.<br />

4-1BB treatment resulted in 34.5% EOMES and perforin double positive<br />

TIL compared to 18.8% in the control group. Conclusions: 4-1BB agonism<br />

is associated with enhanced kinetics <strong>of</strong> TIL proliferation, increased yield <strong>of</strong><br />

CD8 � TIL, lower Treg numbers, increased markers <strong>of</strong> cytolysis, and<br />

enhanced IFN� release upon tumor re-stimulation. 4-1BB agonism may be<br />

a useful adjuvant in the generation <strong>of</strong> TIL in future clinical trials.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


Developmental Therapeutics—<strong>Clinical</strong> Pharmacology and Immunotherapy<br />

2512 <strong>Clinical</strong> Science Symposium, Sat, 1:15 PM-2:45 PM<br />

Phase I study <strong>of</strong> MK-3475 (anti-PD-1 monoclonal antibody) in patients<br />

with advanced solid tumors. Presenting Author: Amita Patnaik, South<br />

Texas Accelerated Research Therapeutics, LLC, San Antonio, TX<br />

Background: Programmed death-1 (PD-1) is an inhibitory T-cell coreceptor that<br />

may lead to suppression <strong>of</strong> antitumor immunity. MK-3475 is a humanized<br />

monoclonal IgG4 antibody against PD-1. Preclinically, MK-3475 has shown<br />

antitumor activity in multiple tumor types. This first-in-human phase I trial<br />

explored safety, PK, PD, and antitumor activity <strong>of</strong> MK-3475. Methods: An<br />

open-label, dose escalation study was conducted in patients with advanced<br />

malignancy refractory to standard therapy. Cohorts <strong>of</strong> 3-6 patients were<br />

enrolled (3�3 design) at escalating IV doses <strong>of</strong> 1, 3, and 10 mg/kg. Following<br />

an initial dose and 28-day Cycle 1, patients were allowed to subsequently<br />

receive multiple doses given every 2 wks. Radiographic assessment was<br />

conducted every 8 wks using RECIST 1.1 guidelines. Results: Nine patients, 3<br />

at each dose level, completed the dose-limiting toxicity (DLT) period (28 d).<br />

Patients had non–small cell lung cancer (NSCLC, n�3), rectal cancer (n�2),<br />

melanoma (MEL, n�2), sarcoma (n�1), or carcinoid (n�1). To date, a total <strong>of</strong><br />

63 doses were administered (median 7/patient; max 12) without DLT.<br />

Drug-related adverse events (AEs) across all doses included Grade 1 fatigue<br />

(n�3), nausea (n�2), diarrhea (n�1), dysgeusia (n�1), breast pain (n�1),<br />

and pruritus (n�1). One drug-related Grade 2 AE <strong>of</strong> pruritus was reported. No<br />

drug-related AEs � Grade 3 were observed. PK data are shown in the table.<br />

Based on RECIST, 1 patient with MEL on therapy �6 mths had a partial<br />

response, and preliminary evidence <strong>of</strong> tumor size reduction (stable disease)<br />

was observed in 3 additional patients with advanced cancer. Conclusions:<br />

MK-3475 was well-tolerated without DLT across 3 tested dose levels.<br />

Evidence <strong>of</strong> antitumor activity was observed. Enrollment continues to obtain<br />

additional safety, PK, and efficacy data; updated data will be presented at the<br />

meeting.<br />

Mean (CV%) PK parameter values <strong>of</strong> MK-3475 following single IV dose <strong>of</strong><br />

1, 3, or 10 mg/kg in cycle 1.<br />

Dose (mg/kg) N Cmax (·g/mL) AUC (0-28day) (�g·day/mL) a t1/2 (day)<br />

1 4 6.8 (23) 163 (20) b 15.1 (41) b<br />

3 3 109 (26) 990 (23) 21.7 (11)<br />

10 2 337 (8) 2,640 (30) 13.6 (28)<br />

aPK sampling up to 28 days following first IV administration; therefore t1/2 not<br />

fully characterized.<br />

bN�3 due to subject discontinuation.<br />

2515 Poster Discussion Session (Board #3), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Radiotherapy plus the anti-EGFR mAb nimotuzumab or placebo for the<br />

treatment <strong>of</strong> high-grade glioma patients. Presenting Author: Maria Teresa<br />

Solomon, Calixto García Hospital, Havana, Cuba<br />

Background: Despite remarkable advances in multimodal therapy, high<br />

grade glioma (HGG) patients still face a poor prognosis. EGFR is well<br />

validated as a primary contributor <strong>of</strong> HGG initiation and progression.<br />

Nimotuzumab is a humanized monoclonal antibody (mAbs) that recognizes<br />

the EGFR extracellular domain. While it has similar preclinical and clinical<br />

activity when compared to other anti-EGFR mAbs, it does not induce skin<br />

toxicity or hypomagnesemia. Methods: A randomized, double blind, multicentric<br />

clinical trial was conducted in 70 anaplastic astrocytoma (AA) and<br />

glioblastoma multiforme (GBM) patients that received radiotherapy (RT)<br />

plus nimotuzumab or placebo. Patients received 6 weekly doses <strong>of</strong><br />

nimotuzumab or placebo together with radiotherapy. Treatment was maintained<br />

every 3 weeks, until completing 1 year <strong>of</strong> treatment. GBM patients<br />

did not receive temozolomide since the drug cannot be sold to Cuba. The<br />

objectives <strong>of</strong> this study were to assess the overall survival, progression free<br />

survival (PFS), response rate, immunogenicity and safety in both treatment<br />

groups. Results: Seventy patients were included in the study: 41 AA and 29<br />

GBM. The median cumulative dose was 3600 mg <strong>of</strong> nimotuzumab and the<br />

median antibody number <strong>of</strong> doses was 16. The combination <strong>of</strong> nimotuzumab<br />

and radiotherapy was very safe. The most prevalent related adverse<br />

events included grade 1-2 nausea, fever, tremors and anorexia. There was<br />

no increasing toxicity with repeated drug exposure. No anti-idyotipic<br />

response was detected. The mean and median survival time for subjects<br />

treated with nimotuzumab and RT was 31.06 and 17.76 months while the<br />

mean and median survival time for controls was 21.07 and 12.63 months,<br />

respectively. For the evaluable patients <strong>of</strong> the AA stratum, the median<br />

survival time was 44.56 months (active drug) vs. 14.6 months (control).<br />

For the evaluable patients in the GBM cohort, the median survival time was<br />

16.06 months (nimotuzumab arm) vs. 8.36 months (placebo arm). Median<br />

PFS was 18.23 vs. 6.25 months. Conclusions: In this randomized trial,<br />

nimotuzumab continues showing an excellent safety pr<strong>of</strong>ile and positive<br />

efficacy results in patients with high grade glioma in combination with<br />

irradiation.<br />

145s<br />

2514 Poster Discussion Session (Board #2), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Effect <strong>of</strong> stimulation <strong>of</strong> natural killer cells with an anti-CD137 mAb on the<br />

efficacy <strong>of</strong> trastuzumab, cetuximab, and rituximab. Presenting Author:<br />

Holbrook Edwin Kohrt, Stanford Cancer Center, Stanford, CA<br />

Background: Antibody-dependent cell-mediated cytotoxicity (ADCC), mediated<br />

by natural killer (NK) cells, plays an important role in the efficacy <strong>of</strong><br />

monoclonal antibodies (mAb)s. CD137 is a costimulatory molecule expressed<br />

on immune cells following activation, including NK cells. We<br />

hypothesize that as the antitumor efficacy <strong>of</strong> mAbs is due to ADCC, their<br />

activity can be enhanced by stimulation <strong>of</strong> NK cells with an anti-CD137<br />

agonistic mAb. Methods: Upregulation <strong>of</strong> CD137 on NK cells was assessed<br />

using CD20�lymphoma, HER2�breast, and EGFR�head and neck cell<br />

lines and primary patient samples. NK cell degranulation, cytokine release<br />

and cytotoxicity were assessed by CD107a mobilization, IFN-� secretion,<br />

and chromium release. Mechanism <strong>of</strong> synergy was explored by cell<br />

depletion in an immune competent mouse model. Xenotransplanted<br />

models were used to demonstrate anti-tumor activity and sufficiency <strong>of</strong> an<br />

innate immune response. Results: NK cells in human primary patient<br />

samples do not express CD137 at baseline, however CD137 is highly<br />

upregulated when encountering mAb-coated tumor cells. MAb-induced NK<br />

cell degranulation and cytotoxicity are enhanced by anti-CD137 agonistic<br />

mAb. In a murine lymphoma model, anti-CD137 mAb significantly enhances<br />

anti-tumor activity <strong>of</strong> anti-CD20 mAb leading to complete tumor<br />

resolution and prolonged survival. NK cell depletion completely abrogates<br />

the therapeutic effect. In seven xenotransplant models, sequential administration<br />

<strong>of</strong> rituximab, trastuzumab or cetuximab plus anti-CD137 mAb<br />

provided superior reduction in tumor burden and prolonged overall survival.<br />

In a phase 0 biomarker study, level <strong>of</strong> CD137 expression on circulating and<br />

intratumoral NK cells was influenced by disease burden, prior treatment,<br />

Fc�RIII polymorphism, and time since mAb therapy. Conclusions: Our<br />

results demonstrate the synergy <strong>of</strong> anti-CD137 mAb and a tumor-targeting<br />

mAb by stimulation <strong>of</strong> mAb-activated NK cells with anti-CD137 mAb to<br />

enhance ADCC. These results support a novel, sequential antibody approach<br />

against CD20�B cell, HER2�breast, and EGFR�head and neck<br />

malignancies by targeting first the tumor and then the host immune system.<br />

2516 Poster Discussion Session (Board #4), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Safety <strong>of</strong> the antimyostatin monoclonal antibody LY2495655 in healthy<br />

subjects and patients with advanced cancer. Presenting Author: Gayle S.<br />

Jameson, Virginia G. Piper Cancer Center/Translational Genomics Research<br />

Institute, Scottsdale, AZ<br />

Background: Skeletal muscle wasting (cachexia) is a prevalent and not<br />

readily managed condition in advanced cancer patients. LY2495655 is a<br />

humanized monoclonal antibody to myostatin, which has demonstrated<br />

positive effects on cachexia measures in animal models. We present phase<br />

I trial data on use <strong>of</strong> LY2495655 in healthy volunteers (Study 1) and<br />

interim data from an ongoing phase I study in patients with advanced<br />

cancer (Study 2). Methods: Study 1 was a randomized, placebo-controlled,<br />

blinded, single-dose, parallel, dose-escalation study evaluating the safety<br />

and tolerability <strong>of</strong> IV or SC LY2495655 (0.7 mg-700 mg). Study 2 is an<br />

ongoing nonrandomized, open-label study evaluating the safety and pharmacokinetics<br />

(PKs) <strong>of</strong> LY2495655 in patients with advanced cancer not<br />

receiving chemotherapy. Dose cohorts (2 mg-700 mg, �3 patients per<br />

cohort) were to be treated until the maximum tolerated dose (MTD) was<br />

met, or the highest dose (700 mg) cohort was completed. Final locked data<br />

from Study 1 and interim data from the dose escalation phase <strong>of</strong> Study 2<br />

were used in the analyses. Results: In Study 1, 64 healthy volunteers were<br />

enrolled (48 LY2495655, 16 placebo). In Study 2, 22 patients had<br />

received treatment with LY2495655 at the time <strong>of</strong> the analysis. In both<br />

studies, all doses <strong>of</strong> LY2495655 were well tolerated (no DLTs were<br />

observed and MTD was not reached), and nonlinear PKs were observed<br />

(most evident in lower dose levels). In Study 1, thigh muscle volume<br />

generally increased with LY2495655. In Study 2, increased muscle<br />

volume was observed only at 21-mg and 70-mg doses. Consistent increases<br />

in hand grip strength and improvements in functional tests were observed<br />

at doses �21 mg. Conclusions: There were no unusual safety concerns in<br />

healthy subjects or cancer patients. PK results were consistent between the<br />

2 studies. Increases in muscle volume were observed in both studies, with<br />

concomitant improvement in functional measures. However, there is no<br />

clear trend in dose-dependent efficacy, possibly due to extremely small<br />

sample sizes and patient heterogeneity. Enrollment in Study 2 continues<br />

with dose expansion cohorts. A Phase 2 study is ongoing in pancreatic<br />

cancer patients.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


146s Developmental Therapeutics—<strong>Clinical</strong> Pharmacology and Immunotherapy<br />

2517 Poster Discussion Session (Board #5), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

A phase I safety study <strong>of</strong> NPC-1C: A novel, therapeutic antibody to treat<br />

pancreas and colorectal cancers. Presenting Author: Philip M. Arlen,<br />

Neogenix Oncology, Rockville, MD<br />

Background: NPC-1C (NEO-101; Ensituximab) is a chimeric monoclonal<br />

antibody being developed as a novel biological treatment for pancreatic and<br />

colorectal cancers. This antibody was selected from a panel <strong>of</strong> hybridomas<br />

generated from mice immunized with semi-purified membrane-associated<br />

proteins derived from biologically screened, pooled human allogeneic colon<br />

cancer tissues. The NPC-1C target appears to be a variant <strong>of</strong> MUC5AC that<br />

is expressed specifically by human colon and pancreatic tumors with<br />

minimal cross-reactivity to normal GI tract tissues. Methods: A phase I open<br />

label, multi-center dose escalation clinical trial with NPC-1C has completed<br />

accrual <strong>of</strong> subjects with advanced pancreatic and colorectal cancer<br />

refractory to standard therapy. The primary objectives are to determine<br />

safety and tolerability <strong>of</strong> escalating doses <strong>of</strong> NPC-1C and to assess<br />

pharmacokinetics (PK) and select immune responses to the antibody at<br />

each dose level. Secondary objectives are to evaluate evidence <strong>of</strong> clinical<br />

benefit and to explore immunologic correlates Results: Three cohorts <strong>of</strong><br />

subjects have been treated with NPC-1C at escalating dose levels <strong>of</strong> 1, 1.5,<br />

and 2 mg/kg IV every two weeks. All 15 subjects (10 colorectal, 5<br />

pancreatic) have enrolled on study (5/15 non-evaluable). No DLT was noted<br />

at the 1mg/kg dose level with 2 <strong>of</strong> 3 patients showing stable disease at<br />

completion <strong>of</strong> Course 1. At the 2 mg/kg dose level, one patient demonstrated<br />

stable disease after course 1. At escalating rates <strong>of</strong> infusion, 3<br />

subjects experienced mild to moderate hemolysis that resolved without<br />

intervention. All 6 patients who have received 1.5 mg/kg at a constant rate<br />

<strong>of</strong> infusion have been evaluated, with no dose limiting toxicity. One <strong>of</strong> 5<br />

patients re-staged thus far in the 1.5 mg/kg cohort showed stable disease<br />

on scans after completing the first course <strong>of</strong> treatment (4 doses), and<br />

continued to receive additional doses. Preliminary data show no drug<br />

induced antibody responses in treated subjects. PK, HAMA and cytokine<br />

evaluation is ongoing. Conclusions: NPC-1C is tolerated well at the current<br />

dose level <strong>of</strong> 1.5 mg/kg every 2 weeks without producing any DLT We plan<br />

to continue accrual in the Phase 2A to evaluate clinical responses and<br />

additional safety data.<br />

2519 Poster Discussion Session (Board #7), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Assessment <strong>of</strong> a novel immunological biomarker SNP panel from a phase<br />

III trial <strong>of</strong> Bacillus Calmette-Guérin (BCG) adjuvant treatment in stage III<br />

melanoma patients. Presenting Author: Connie Ging Ting Chiu, Department<br />

<strong>of</strong> Molecular Oncology and Division <strong>of</strong> Surgical Oncology, John Wayne<br />

Cancer Institute, Santa Monica, CA<br />

Background: Melanoma is an immunogenic cancer, whereby immunotherapy<br />

in stage III patients has shown some success. However, there are no<br />

effective immune biomarker tests to identify patients most likely to benefit<br />

from immunotherapy. BCG (Bacillus Calmette-Guérin) therapy is a form <strong>of</strong><br />

immunomodulation. The objective was to assess the predictive clinical<br />

effect <strong>of</strong> a single nucleotide polymorphism(SNP) immune biomarker panel<br />

in stage III resected melanoma patients treated with BCG. Methods:<br />

MMAIT-III was a phase III prospective randomized international multicenter<br />

trial <strong>of</strong> BCG�melanoma vaccine vs BCG�placebo after complete<br />

resection <strong>of</strong> stage III melanoma patients (NIH #NCT00052130). 120/292<br />

patients with palpable disease treated with resection and BCG�placebo<br />

had lymphocytes(PBL) from USA sites available for analysis. Endpoints<br />

were overall survival(OS) and disease-free survival(DFS), with a 10-yr<br />

follow-up. PBL DNA was assessed by MassARRAY MALDI-TOF for 28 SNPs<br />

associated with macrophage/monocyte-related immune response pathways<br />

to BCG/tuberculosis. A pilot study(n�34) from phase II BCG trial confirmed<br />

presence <strong>of</strong> the SNPs. A logistic regression determined a SNP score,<br />

and a cut<strong>of</strong>f was identified by ROC and used as a predictor in a Cox<br />

proportional hazard model in a verification study(n�120). Results: 9 SNPs<br />

in 6 genes had prognostic value: NRAMP1 and CD14, 18, 195, 209, 282.<br />

The 9-SNP panel distinguished patients in 2 survival groups(OS median<br />

4.9-yrs vs 1.5-yrs, p�0.0008), AUC�0.77. SNP biomarker positivity<br />

demonstrates significant association with 10-yr OS (59.7% vs 15.7%; HR<br />

1.97, CI 1.11-3.50, p�0.018) and DFS (47.2% vs 12.3%; HR 2.35, CI<br />

1.43-3.92, p�0.0007). In Cox model, the SNP panel was a significant<br />

predictor <strong>of</strong> OS(HR 2.69, CI 1.56-9.00, p�0.0052) and DFS(HR 2.33, CI<br />

1.49-5.16, p�0.0003) independent <strong>of</strong> known melanoma prognostic<br />

factors. Conclusions: The 9-SNP panel identified patients with an exceptionally<br />

favourable disease outcome, and may represent a stratifying predictive<br />

SNP panel for identifying patients that are inherently responsive to BCG<br />

therapy and potentially other immunomodulating agents in melanoma.<br />

2518 Poster Discussion Session (Board #6), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Myeloid-derived suppressor cell quantity prior to treatment with ipilimumab<br />

at 10mg/kg to predict for overall survival in patients with metastatic<br />

melanoma. Presenting Author: Shigehisa Kitano, Memorial Sloan-<br />

Kettering Cancer Center, New York, NY<br />

Background: Ipilimumab, an antibody that blocks the function <strong>of</strong> the<br />

immune inhibitory molecule cytotoxic T lymphocyte antigen 4 (CTLA-4),<br />

significantly prolongs survival in patients with metastatic melanoma.<br />

Approximately 30% <strong>of</strong> patients derive clinical benefit from therapy.<br />

Defining biomarkers <strong>of</strong> response to ipilimumab therapy would enable<br />

selection <strong>of</strong> patients more likely to respond and is relevant for both<br />

practicing clinicians and for clinical trial design. We performed a pilot<br />

correlative study evaluating myeloid derived suppressor cells (MDSC), a<br />

population <strong>of</strong> immune suppressive monocytic cells, as a biomarker <strong>of</strong><br />

clinical outcome. Methods: Peripheral blood from 26 patients with stage IV<br />

melanoma treated with ipilimumab 10mg/kg every 3 weeks for 4 doses at<br />

our center, as part <strong>of</strong> an expanded access program (BMS CA184-045) was<br />

assessed for MDSC quantity (%CD14� ,HLA-DRlow/- cells) pre-treatment, at<br />

week 7, week 12, and week 24 by flow cytometry. MDSC ability to inhibit T<br />

cell proliferation was tested using an in vitro suppression assay. Results: We<br />

found that lower MDSC quantity pre-treatment predicted for improved<br />

overall survival (Hazard ratio 1.07 (1.03, 1.11) p�0.002) and trended<br />

toward associating with clinical benefit measured at week 24 imaging<br />

(p�0.09). This effect was independent <strong>of</strong> pre-treatment or week 7 absolute<br />

lymphocyte counts (ALC) and pre-treatment LDH when evaluated in a<br />

multivariate model with ALC and MDSC quantity HR 1.10; 95% CI 1.04,<br />

1.17 p�0.0006 and LDH and MDSC quantity HR 1.06; 95% CI 1.01,<br />

1.11 p � 0.013. Furthermore, a general trend <strong>of</strong> increasing MDSC number<br />

by week 24 from the pre-treatment baseline was associated with patients<br />

that did not achieve clinical benefit. MDSC suppressed peripheral blood T<br />

cell proliferation as measured by CFSE dilution in response to anti-CD3<br />

antibody stimulation. Conclusions: Pre-treatment MDSC quantity may<br />

predict clinical response following ipilimumab therapy. Further studies<br />

evaluating MDSC as a biomarker <strong>of</strong> ipilimumab therapy are warranted both<br />

retrospectively and prospectively in a broader group <strong>of</strong> patients.<br />

2520^ Poster Discussion Session (Board #8), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Investigating genes and pathways that play a role in immune responses<br />

mediated by anti-CTLA-4 therapy. Presenting Author: Jianjun Gao, University<br />

<strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: Blockade <strong>of</strong> the inhibitory T cell molecule CTLA-4 with a<br />

monoclonal antibody has led to enhanced anti-tumor immune responses<br />

and clinical benefit. Ipilimumab, an anti-CTLA-4 antibody (BMS), was<br />

recently FDA-approved for the treatment <strong>of</strong> metastatic melanoma. Only a<br />

subset <strong>of</strong> patients benefit from anti-CTLA-4. In order to identify genes and<br />

pathways that are induced by anti-CTLA-4, which may be used in future<br />

studies for potential correlation with clinical outcomes or provide additional<br />

targets for therapy, we purified and analyzed CD4 T cells from<br />

patients treated with anti-CTLA-4 for changes in gene expression pr<strong>of</strong>ile.<br />

We also obtained tumor tissues from treated patients for similar studies.<br />

Methods: On an IRB-approved Phase Ia pre-surgical clinical trial, 6 patients<br />

with localized bladder cancer were treated with two doses <strong>of</strong> Ipilimumab at<br />

10 mg/kg at weeks 1 and 4. Blood was collected pre-therapy and<br />

post-therapy (3 weeks after each dose). CD4 T cells were enriched from<br />

peripheral blood by using the CD4 T cell isolation kit from Miltenyi Biotec<br />

(Auburn, CA). Total RNA was isolated from purified CD4 T cells using<br />

Qiagen RNeasy kit for Affymetrix microarray analyses. Microarray data were<br />

then analyzed using Ingenuity iReport (Redwood City, CA). RT-PCR, RPPA<br />

and Western blot were used to confirm significant changes in genes or<br />

pathways identified in microarray analyses. Results: Ipilimumab treatment<br />

resulted in modulation <strong>of</strong> differentially expressed genes (DEGs). After dose<br />

#1, Ipilimumab only modulated 16 DEGs �2-fold (p�0.05) in CD4 T cells.<br />

After two doses <strong>of</strong> treatment, Ipilimumab significantly changed expression<br />

<strong>of</strong> a total <strong>of</strong> 100 DEGs. Further pathway analyses indicated that Ipilimumab<br />

induced a variety <strong>of</strong> pathways involved in cell cycle control, cell proliferation,<br />

apoptosis, and immune modulation. Specifically, these pathways<br />

include PI3K/AKT, MAP/ERK, IFN/JAK-STAT, granzyme, and protein<br />

ubiquination. Conclusions: Ipilimumab treatment results in modulation <strong>of</strong><br />

multiple genes and pathways, which likely play important roles in antitumor<br />

immune responses and need to be considered for future optimization<br />

<strong>of</strong> anti-CTLA-4 therapy and design <strong>of</strong> combination immunotherapy strategies.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


Developmental Therapeutics—<strong>Clinical</strong> Pharmacology and Immunotherapy<br />

2521 Poster Discussion Session (Board #9), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

The immunological impact <strong>of</strong> the RAF inhibitor BMS908662: Preclinical<br />

and early clinical experience in combination with CTLA-4 blockade.<br />

Presenting Author: Margaret Callahan, Memorial Sloan-Kettering Cancer<br />

Center, New York, NY<br />

Background: Two new approaches to treat advanced melanoma have<br />

transformed the standard <strong>of</strong> care: the CTLA-4 blocking antibody, ipilimumab,<br />

and the targeted inhibitor <strong>of</strong> mutated BRAF, vemurafenib. These<br />

agents are mechanistically unique and combination therapy is a promising<br />

next step. We evaluated the combination <strong>of</strong> BMS908662 (662), a pan RAF<br />

inhibitor, with CTLA-4 blockade in preclinical studies and report first-inhuman<br />

clinical experience with this combination. Methods: 1) We tested<br />

the impact <strong>of</strong> 662 on T cells in vitro, using cultured human T cells, and in<br />

vivo, using OT-1 transgenic mice. T cell activation and MAPK pathway<br />

signaling were assessed in parallel. 2) Preclinical studies measuring the<br />

anti-tumor activity <strong>of</strong> combination therapy were performed in CT-26 and<br />

SA1N tumor models. 3) Three pts with BRAF mutant stage IV melanoma<br />

were treated at MSKCC on CA206005, an IRB-approved protocol, receiving<br />

ipilimumab (3 mg/kg) and 662 (25 mg bid) (NCT01245556). Two pts<br />

consented to an IRB-approved protocol permitting immune monitoring.<br />

Results: 1) In vitro studies demonstrate that 662 can potentiate T cell<br />

activation after stimulation. This corresponds with increased MAPK pathway<br />

signaling, consistent with paradoxical activation <strong>of</strong> the MAPK pathway<br />

in wild type cells, a class effect <strong>of</strong> RAF inhibitors. In vivo, enhanced<br />

expansion <strong>of</strong> OT-1 cells after ovalbumin challenge was seen in mice treated<br />

with 662. T cell expansion was greatest in mice treated with a combination<br />

<strong>of</strong> CTLA-4 blockade and 662 (p�0.05). 2) Both preclinical models<br />

demonstrate superior anti-tumor activity with combination therapy compared<br />

to monotherapy (p�0.05). 3) All pts treated on protocol CA206005<br />

tolerated combination therapy. New keratoacanthomas and SCCs, likely<br />

related to 662, were identified. One pt has an ongoing response at 10 mos<br />

(-85%), one had stable disease for 24 wks (-19%) and a third had disease<br />

progression. Enhanced MAPK signaling in PBMCs after treatment with 662<br />

was detected ex vivo. Conclusions: RAF inhibitors may potentiate T cell<br />

activation in vitro and in vivo, <strong>of</strong>fering one explanation for the enhanced<br />

anti-tumor activity seen in combination with CTLA-4 blockade in preclinical<br />

models.<br />

2523 Poster Discussion Session (Board #11), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Cyclin-A1 expression in acute myeloid leukemia stem cells and its<br />

representation as an immunogenic antigen that can be targeted by<br />

cytotoxic T cells. Presenting Author: Sebastian Ochsenreither, Department<br />

<strong>of</strong> Hematology and Medical Oncology, Charité, CBF, Berlin, Germany<br />

Background: Targeted T-cell therapy represents a more specific and less<br />

toxic alternative to allogeneic stem cell transplantation for providing a<br />

cytotoxic anti-leukemic response to eliminate the leukemic stem cell (LSC)<br />

compartment in acute myeloid leukemia (AML). The aim <strong>of</strong> this study was<br />

to identify a leukemia-associated antigen that is both immunogenic and<br />

exhibits selective high expression in AML LSCs. Methods: Expression<br />

microarrays <strong>of</strong> leukemic and normal cells were used to identify suitable<br />

candidate genes. Cyclin-A1 appeared to be differentially expressed, and<br />

was further analyzed by quantitative RT-PCR, intracellular FACS staining,<br />

and immun<strong>of</strong>luorescence microscopy. T-cell clones specific for cyclin-A1<br />

were generated by stimulation in vitro with an overlapping peptide library,<br />

followed by cloning <strong>of</strong> responding cells. Specificity <strong>of</strong> the clones was<br />

documented by intracellular IFNg and tetramer staining. Cytotoxicity was<br />

determined by chromium release and caspase-3 cleavage assays. Results:<br />

To identify target candidates with a suitable expression pattern, microarray<br />

data from LSCs, hematopoietic cells and healthy tissues were compared.<br />

Cyclin-A1 was found to be selectively expressed in LSCs <strong>of</strong> �50% <strong>of</strong> AML<br />

patients, but minimally expressed in normal tissues with exception <strong>of</strong><br />

testis. Using dendritic cells and monocytes pulsed with a cyclin-A1 peptide<br />

library, T-cells were generated against eight cyclin-A1 oligopeptides. Two<br />

HLA A2-restricted epitopes were further characterized, and specific T-cell<br />

clones recognized both peptide-pulsed target cells and the HLA A2-positive<br />

AML line THP-1. Furthermore, cyclin-A1-specific CD8 T-cells lysed primary<br />

AML cells. Conclusions: Cyclin-A1, known to be important in meiosis,<br />

is expressed in AML LSCs and testis and can be targeted by T-cells. Thus,<br />

cyclin-A1 is the first prototypic LSC-associated leukemia-testis-antigen.<br />

Cyclin-A1 already has been shown to be leukemogenic in mice and to<br />

promote proliferation and survival in AML blasts. This pro-oncogenic<br />

activity, together with high expression levels and a multitude <strong>of</strong> immunogenic<br />

epitopes, make it a promising target for T-cell based therapy<br />

approaches.<br />

147s<br />

2522 Poster Discussion Session (Board #10), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Immune responses and association with clinical outcome <strong>of</strong> advanced<br />

colorectal cancer patients treated with the multi-peptide vaccine IMA910.<br />

Presenting Author: Sabrina Kuttruff, Immatics Biotechnologies GmbH,<br />

Tuebingen, Germany<br />

Background: IMA910 is a novel multi-peptide cancer vaccine consisting <strong>of</strong><br />

10 HLA class I and 3class II tumor-associated peptides (TUMAPs), which<br />

were selected based on natural presentation on colorectal tumors by the<br />

XPRESIDENT antigen discovery platform. Methods: 92 HLA-A*02� advanced<br />

colorectal cancer (aCRC) patients (pts) with stable or responding<br />

disease after 12 weeks <strong>of</strong> first-line oxaliplatin-based therapy were enrolled<br />

in this phase I/II trial. After pre-treatment with cyclophosphamide (300<br />

mg/m2 ), patients were immunized intradermally with IMA910 plus the<br />

immune modulator GM-CSF without (cohort 1; n�66) or with (cohort 2;<br />

n�26) topically applied imiquimod, another immune modulator acting via<br />

toll-like receptor 7 on antigen presenting cells. T-cell responses to<br />

individual IMA910 peptides were analyzed by HLA multimer and intracellular<br />

cytokine staining (ICS) assays. Results: IMA910 elicited immune<br />

responses towards multiple class I (43%) and class II TUMAPs (65%).<br />

34% <strong>of</strong> pts responded to multiple class I and class II TUMAPs. Pts that<br />

received imiquimod were more <strong>of</strong>ten multi-peptide class I responders in the<br />

ICS assay (p�0.016) and showed an approx. 2x higher frequency <strong>of</strong> T-cell<br />

response (p�0.12). Responses to multiple class I and class II TUMAPs<br />

were associated with higher disease control rate at all time points (all<br />

p�0.02), increased time to progression (p�0.006), progression-free<br />

survival (p�0.009) and OS (p�0.088, HR�0.53). Baseline characteristics<br />

<strong>of</strong> multi vs. non-multi responders were overall well comparable. In a<br />

prospectively defined, blinded matched-pair analysis with patients in arm C<br />

<strong>of</strong> the COIN trial, multi-peptide responder patients showed a prolonged<br />

survival vs. corresponding COIN patients (p�0.04), while the non-multi<br />

responder patients had comparable survival. Conclusions: IMA910 is<br />

immunogenic. Imiquimod increases the quality <strong>of</strong> immune responses.<br />

Responses to multiple TUMAPs are associated with better clinical outcome.<br />

Several observations indicate that this association is not a reflection<br />

<strong>of</strong> better prognosis <strong>of</strong> the immunologically responding subset <strong>of</strong> patients.<br />

These results suggest further development <strong>of</strong> IMA910.<br />

2524 Poster Discussion Session (Board #12), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Adjuvant dendritic cell (DC)-based vaccine therapy <strong>of</strong> melanoma patients.<br />

Presenting Author: Natalia N. Petenko, N. N. Blokhin Cancer Research<br />

Center, Russian Academy <strong>of</strong> Medical Sciences, Moscow, Russia<br />

Background: Earlier DC vaccine therapy demonstrated clinical benefit in<br />

some patients (pts) with advanced melanoma although the effect was seen<br />

only in pts with minimal tumor burden (ASCO2007 abstract No: 3077). As<br />

there is no effective treatment for high risk resected melanoma we<br />

conducted an exploratory trial to assess the effectiveness <strong>of</strong> DC vaccine in<br />

stage III and IV melanoma pts after radical surgery in comparison with<br />

observation. Methods: 108 stage III and IV melanoma pts were enrolled in<br />

two comparative arms. The arms were well balanced in respect with<br />

demographic and prognostic factors. The vaccine was based on mature<br />

autologous monocyte-derived DC primed with autologous tumor lysate,<br />

administered intradermally every 2-6 weeks until the disease progression.<br />

Disease free survival (DFS) and overall survival (OS) were evaluated with<br />

Kaplan-Meier method and compared between the two arms using the log<br />

rank test. Safety <strong>of</strong> DC vaccine was also monitored. Results: The vaccine<br />

arm included 56 pts (stage III – 46, stage IV – 10 pts) who were surgically<br />

rendered free <strong>of</strong> disease and treated with DC vaccine. 52 pts were enrolled<br />

in the control arm (stage III – 47, stage IV – 5 pts), they were treated<br />

surgically and observed afterwards. At a median follow-up <strong>of</strong> 22 months,<br />

the HR (DC vs observation) for DFS was 0.45 (p � 0.05; 95%CI<br />

0.29-0.69) and for OS was 0.71 (p�0.23; 95%CI 0.40-1.25). 60% <strong>of</strong> pts<br />

in the DC arm remained alive at this time point. Risk reduction significantly<br />

correlated with the strong delayed type hypersensitivity reaction induced by<br />

vaccine in 31 (55%) out <strong>of</strong> 56 vaccinated pts. The vaccine was safe and<br />

well tolerated although vitiligo was registered in 4 cases which was<br />

associated with more durable time to progression and OS. Conclusions: The<br />

immunotherapy with DC vaccine is safe and significantly improves DFS<br />

compared with observation in the adjuvant treatment <strong>of</strong> stage III and IV<br />

melanoma.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


148s Developmental Therapeutics—<strong>Clinical</strong> Pharmacology and Immunotherapy<br />

2525 Poster Discussion Session (Board #13), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

CD40 ligand/interferon-� matured DC immunization with gp100 antigen<br />

HLA class I A *0201 restricted peptides in patients with newly diagnosed<br />

metastatic melanoma. Presenting Author: Gerald P. Linette, Washington<br />

University, St. Louis, MO<br />

Background: CD40L/IFN-� matured Dendritic Cells (DCs) produce IL-12<br />

and are potent antigen-presenting cells for naïve resting T cells. We sought<br />

to determine the magnitude and kinetics <strong>of</strong> CD8� T cell growth in patients<br />

receiving autologous CD40L/IFN-� matured DC and identify biomarkers<br />

associated with clinical outcome. Methods: A phase I clinical trial<br />

(NCT00683670) incorporating CD40L/IFN-� for the ex vivo maturation <strong>of</strong><br />

autologous DCs pulsed with three well characterized gp100 melanoma<br />

antigen derived peptides (G154, G209-2M, G280-9V) was initiated with<br />

enrollment from 2008-11 at a single center. HLA-A*0201� individuals<br />

with treatment naïve metastatic melanoma were immunized every 3 weeks<br />

by intravenous infusion for six doses after a single dose <strong>of</strong> cyclophosphamide<br />

(300 mg/m2 iv). CT imaging was performed at baseline, week 9<br />

and 18 for clinical assessment using RECIST. Responding patients were<br />

eligible for maintenance doses every 2-4 months. PBMC were taken weekly<br />

for immune monitoring by tetramer analysis and functional assays. DC<br />

preparations were characterized to assess for biomarkers <strong>of</strong> response.<br />

Results: 10 patients were screened. Among the 7 treated patients, there<br />

were 3 confirmed responses (independently verified), including one durable<br />

CR �3 years and 2 PR. Three patients had rapid disease progression<br />

and received only 3 doses. Four patients (1 CR, 2 PR, 1 PD) received 6 or<br />

more vaccine doses. No SAEs were noted. There was no correlation between<br />

tumor volume and response. Using pre-specified immune response criteria,<br />

6 (86%) treated patients developed CD8� T cell immunity to all three<br />

peptides as assessed by tetramer analysis. The vaccine-induced T cells<br />

from all 6 individuals were polyfunctional and killed gp100�, HLA-A2�<br />

human melanoma targets in a standard 51Cr release assay. IL-12 production<br />

by DCs correlated with TTP (p�0.0198, likelihood ratio test) but not<br />

OS (p�0.08). Conclusions: Weekly immune monitoring reveals the rapid<br />

onset <strong>of</strong> CD8� T cell immunity against gp100 among the responder<br />

patients. This is the first DC vaccine clinical trial in melanoma to<br />

demonstrate a correlation <strong>of</strong> IL-12 production and TTP.<br />

2527 Poster Discussion Session (Board #15), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

EGF-based cancer vaccine for advanced NSCLC: Results from a phase III<br />

trial. Presenting Author: Tania Crombet Ramos, Center <strong>of</strong> Molecular<br />

Immunology, Havana, Cuba<br />

Background: The prognosis <strong>of</strong> patients with advanced non small cell lung<br />

cancer (NSCLC) remains dismal. Epidermal Growth Factor Receptor<br />

(EGFR) is overexpressed in epithelial tumors and its role in the development<br />

<strong>of</strong> NSCLC is widely proven. The EGF vaccine is a therapeutic cancer<br />

vaccine composed by recombinant Epidermal Growth Factor (EGF) conjugated<br />

to a carrier protein, P64K from Neisseria Meningitides and Montanide,<br />

as adjuvant. The vaccine is intended to induce antibodies against<br />

self EGF that would block EGF-EGFR interaction. Methods: A multicentric,<br />

randomized Phase III trial was designed to assess the efficacy, immunogenicity<br />

and safety <strong>of</strong> the EGF cancer vaccine in advanced NSCLC patients.<br />

Patients older than 18 years with histology or cytology proven NSCLC at<br />

stage IIIB and IV were enrolled in the trial. All patients received no less than<br />

4 platinum-based cycles and achieved at least stable disease, before<br />

entering the trial. A low-dose <strong>of</strong> cyclophosphamide was administered 3<br />

days before the first immunization. Then, patients received 4 quarterly<br />

immunizations followed by monthly re-immunizations. Control patients<br />

received best supportive care. Results: In total, 405 patients bearing stage<br />

IIIB/IV NSCLC were recruited in 21 sites. The vaccine was very well<br />

tolerated. The most frequent adverse events consisted in grade 1/2<br />

injection site pain, fever, headache, vomiting and chills. The vaccine was<br />

immunogenic. Antibody titers against EGF significantly increased with<br />

vaccination and on the contrary, EGF concentration in sera showed a fast<br />

reduction after immunization. There was an inverse correlation between the<br />

anti-EGF antibody titers and the EGF concentration in sera. Baseline EGF<br />

concentration was a worse prognostic factor for the control patients and a<br />

predictive factor for vaccinated subjects. The overall survival was significantly<br />

better for vaccinated patients as compared to controls. There was a<br />

direct correlation between the antibody titers and survival. Conclusions: The<br />

EGF cancer vaccine was very well tolerated and significantly increased<br />

overall survival <strong>of</strong> the vaccinated patients. Baseline EGF concentration<br />

predicted survival benefit. The EGF vaccine is a new therapeutic option for<br />

advanced NSCLC patients.<br />

2526 Poster Discussion Session (Board #14), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Interim analysis <strong>of</strong> a phase II randomized clinical trial <strong>of</strong> samrium-153<br />

(Sm-153) with or without PSA-TRICOM vaccine in metastatic castrationresistant<br />

prostate cancer after docetaxel. Presenting Author: Christopher<br />

Ryan Heery, Medical Oncology Branch, National Cancer Institute, National<br />

Institutes <strong>of</strong> Health, Bethesda, MD<br />

Background: A prior randomized, placebo-controlled, multi-center phase 2<br />

trial <strong>of</strong> PSA-TRICOM (PROSTVAC) demonstrated an overall survival benefit.<br />

Sm-153 is a radiopharmaceutical that targets osteoblastic bone lesions.<br />

Preclinical data indicated that Sm-153 could alter tumor phenotype,<br />

causing upregulation <strong>of</strong> Fas, MHC Class I, and tumor-associated antigens,<br />

making tumor cells more amenable to immune-mediated killing. Methods:<br />

This is a phase 2 multi-center trial design intended to randomize 68<br />

patients (pts) to Sm-153 with or without PROSTVAC. Eligibility included<br />

castrate resistant prostate cancer bone metastases, no visceral disease,<br />

prior docetaxel, ECOG �2, and normal organ function. Sm-153 was given<br />

at 1mCi/kg IV on day 8 and then every 12 weeks. PROSTVAC was given on<br />

days 1, 15, 29, then every 4 weeks. The 1° endpoint is a comparison <strong>of</strong><br />

progression-free survival at 4 months (mo) utilizing PCCWG, but not PSA<br />

criteria. 68 patients will provide 80% power to detect a difference <strong>of</strong> 15%<br />

vs. 40% without progression at 4 mo with a one-tailed alpha � 0.10<br />

assuming Fisher’s exact test comparing these fractions as the primary<br />

method <strong>of</strong> analysis. 2° endpoints are OS, ORR, PSA changes, immunologic,<br />

toxicity, and palliation. Reported here is the result <strong>of</strong> a pre-specified<br />

interim analysis, which required �20% conditional power to detect 15%<br />

vs. 40% without progression at 4 months for the trial to continue. Results:<br />

Of 37 enrolled pts, 3 were not evaluable for PFS. PFS and PSA findings are<br />

found below. Hematologic toxicities (anemia, thrombocytopenia, neutropenia,<br />

or lymphocytopenia) are most common, with grade 3 or 4 thrombocytopenia<br />

occurring in 22% and 26% <strong>of</strong> treatment cycles on Arms A and B,<br />

respectively. The conditional power for the comparison <strong>of</strong> fractions without<br />

progression at 4 mo is 77%. Conclusions: This interim analysis suggests the<br />

combination <strong>of</strong> PROSTVAC and Sm-153 is well tolerated with similar<br />

toxicity pr<strong>of</strong>ile to Sm-153 alone. The early indication <strong>of</strong> improved TTP<br />

warrants continued study accrual.<br />

Sm-153<br />

(A)<br />

Sm-153 � PROSTVAC<br />

(B)<br />

PFS<br />

At4mo 2/17 (11.8%) 5/17 (29.4%)<br />

Median PFS days 60 117<br />

Pt # confirmed PSA decline<br />

> 30% 0 4<br />

> 50% 0 2<br />

2528 Poster Discussion Session (Board #16), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Association <strong>of</strong> idiotype vaccine-induced T-cell response with improved<br />

survival and time-to-next treatment (TTNT) in untreated mantle cell<br />

lymphoma (MCL). Presenting Author: Kieron Dunleavy, Metabolism Branch,<br />

National Cancer Institute, National Institutes <strong>of</strong> Health, Bethesda, MD<br />

Background: Murine models show Id-vaccines induce antitumor responses,<br />

possibly through Th1/Tc1 cytokines. We report an 11-year follow-up <strong>of</strong><br />

Id-vaccine following DA-EPOCH-Rituximab in 26 untreated MCL patients.<br />

Methods: DA-EPOCH-R was administered q3 weeks � 6, followed 12 weeks<br />

later by 5 cycles <strong>of</strong> Id-vaccine. Id protein was made by hybridoma<br />

technology, conjugated to keyhole limpet hemocyanin (KLH) and administered<br />

with GM-CSF. Pre- and post-vaccine immune responses (IR) were<br />

tested in parallel: anti-Id and anti-KLH humoral responses (ELISA);<br />

anti-KLH cellular responses (intracellular cytokine assay); and anti-tumor<br />

cellular responses (cytokine induction, IFN� ELISPOT). Results: Characteristics:<br />

median age 57 (r 22-73), blastoid variant 15%, and MIPI (low-65%;<br />

intermediate-16%; high-19%). With 122 mo median follow-up (r 111-<br />

132), the median PFS is 24 mo and OS is 104 mo. MIPI was associated<br />

with OS (p�0.0002); median OS: low (not reached), intermediate (84 mo)<br />

and high (44 mo). We found no association between OS and anti-KLH IR,<br />

anti-Id humoral response, IFN� ELISPOT, or antitumor TNF� or IFN�<br />

responses. Normalized antitumor T-cell GM-CSF response (median �4.3<br />

vs. �4.3) was associated with OS <strong>of</strong> 79 mo vs. not reached, respectively<br />

(p�0.015 (unadj.) and p�0.045 (adj.)). MIPI (p�0.02) and GM-CSF<br />

(p�0.057) were independently associated with OS. TTNT (based on<br />

disease activity), correlated with antitumor GM-CSF response (p�0.018)<br />

but not MIPI and was independent <strong>of</strong> MIPI (GM-CSF; p�0.041). Correlation<br />

<strong>of</strong> pre-and post-treatment GM-CSF production suggested a priming<br />

effect. Tumor proliferation by GEP did not correlate with OS (n�14).<br />

Conclusions: Antitumor GM-CSF response was significantly associated with<br />

OS and TTNT, suggesting antitumor cellular immune response significantly<br />

delayed tumor growth. Antitumor GM-CSF response may serve as a<br />

surrogate biomarker for vaccine efficacy. Our results are consistent with<br />

recent data that T-cell GM-CSF production is required for breaking<br />

tolerance against self-antigens. Id vaccines may prolong survival <strong>of</strong> MCL<br />

following rituximab-based chemotherapy and should be further evaluated.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


Developmental Therapeutics—<strong>Clinical</strong> Pharmacology and Immunotherapy<br />

2529 Poster Discussion Session (Board #17), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Safety and long-term maintenance <strong>of</strong> anti-HER2 immunity following<br />

booster inoculations <strong>of</strong> the E75 breast cancer vaccine. Presenting Author:<br />

Raetasha Sheavette Dabney, Brooke Army Medical Center, San Antonio, TX<br />

Background: We have completed accrual and are in the follow up portion <strong>of</strong><br />

phase I/II clinical trials evaluating the E75 HER2 peptide vaccine. E75 has<br />

been proven safe, capable <strong>of</strong> stimulating HER2 immunity, and effective in<br />

decreasing breast cancer recurrence rates. During the conduct <strong>of</strong> this trial,<br />

it was noted that E75-specific immunity waned after the Primary Vaccine<br />

Series (PVS) which corresponded with late recurrences. To maintain<br />

long-term immunity, a voluntary booster program was started. Here we<br />

present analysis <strong>of</strong> the booster inoculations. Methods: The trial enrolled<br />

node-positive or high-risk, node-negative breast cancer patients (pts) with<br />

tumors expressing any level <strong>of</strong> HER2 (IHC 1-3�). HLA-A2/A3� pts<br />

comprised the vaccine group (VG), HLA-A2/A3- pts were followed as the<br />

control group (CG). The VG received 4-6 monthly inoculations <strong>of</strong> E75�GM-<br />

CSF. Volunteer booster program pts (BG) received inoculations every 6<br />

months after the PVS. Pts were monitored for toxicities, in vivo responses by<br />

local reactions (LR) and DTH, and in vitro responses measured by<br />

enumeration <strong>of</strong> E75 specific cytotoxic T lymphocytes. Results: 53 pts<br />

received at least 1 booster, 34 received 2, 24 three, 20 four, 12 five, and 8<br />

at least 6. 24% <strong>of</strong> pts had no local toxicity, 73% Grade 1 (G1), 3% G2.<br />

74% had no systemic toxicity, 35% G1, 1% G2. LRs increased significantly<br />

from the initial vaccine (R1) during PVS to each booster (B) (R1: 59.5�3.1<br />

v B1: 89.2�3.3, p�0.001; v B2: 95.15�5, p�0.001; v B3: 86.63�5.5,<br />

p�0.001; v B4: 83.26�4.6, p��0.001; v B5: 80.67�6.7, p�0.006; v<br />

B6: 78.75�9.4, p�0.04). Dimer values increased from the end <strong>of</strong> PVS to<br />

each post-booster value (pre B1:1.29�0.25 v post B1: 1.46�0.38; post<br />

B2: 1.41�0.4; post B3: 1.84�0.35; post B4: 2.23�0.4; post B5:<br />

1.94�0.31; post B6: 2.73�0.09, p�0.02). At median 60 months, the<br />

recurrence rate for BG was 3.8% vs 18.9% in the CG (p�0.01).<br />

Conclusions: Booster inoculations are well-tolerated and appear to assist in<br />

the maintenance <strong>of</strong> long term peptide-specific immunity. Boosted pts have<br />

improved recurrence rates. Based on the success <strong>of</strong> this program, we have<br />

incorporated the practice <strong>of</strong> booster inoculations in our current cancer<br />

vaccine trials.<br />

2531 Poster Discussion Session (Board #19), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Response to epidermal growth factor vaccine in patients with metastatic<br />

non-small cell lung cancer (NSCLC) after progressing to first-line therapy.<br />

Presenting Author: Diego Venegas, Universidad Peruana Cayetano Heredia,<br />

Lima, Peru<br />

Background: A direct correlation between anti–epidermal growth factor<br />

(EGF)antibody titers and survival was demonstrated in vaccinated patients<br />

with novel NSCLC advanced in Phase II studies. We show the results <strong>of</strong><br />

treatment with anti-EGF vaccine in a cohort <strong>of</strong> patients with metastatic<br />

NSCLC after progressing to first line therapy. We evaluated immunogenicity,<br />

safety, treatment response and effect on survival. Methods: 12 patients<br />

with metastatic NSCLC after progressing to first-line therapy received anti<br />

EGF-vaccine alone or in combination with chemotherapy. Results: From<br />

October 2009 until August 2011, 12 patients started treatment with anti<br />

EGF vaccine; mean age 56.5 (42-79 y); 66.7% male; ECOG 0 and 1:<br />

41.7% and 58,3% respectively. Adenocarcinoma (50%), bronchioloalveolar<br />

(33.3%), adenosquamous (16.7%). Metastatic sites: lung (41.7%),<br />

pleura (25%), CNS (16.7%), Kidney (8.3%). In addition to chemotherapy<br />

previous used: radiotherapy (50%), surgery (41.7%), erlotinib (41.7%),<br />

bevacizumab (25%). The 50% patients received vaccine alone. The 83.3%<br />

<strong>of</strong> patients had titers 1/4000 sera dilutions or more (good responders).<br />

According to RECIST 1.1: CR: 8.3%, PR: 16.7%, SD: 41.7%, PD 25%.<br />

Median overall survival was 18.8 months (95% CI: 13.3- 24.4 m). Median<br />

progression-free survival was 7.3 months (95% CI: 6.4 -8.2 m). We found<br />

no statistically significant differences in OS and PFS when comparing<br />

vaccine alone or combined (p � 0.181 and p � 0.801). 75% <strong>of</strong> patients<br />

had adverse effect: more frequently were: 42.4% application site pain,<br />

15.1% fever and 10.38% chills, none <strong>of</strong> them serious. Conclusions:<br />

Vaccination anti EGF in patients with metastatic NSCLC after progressing<br />

to first line, alone or in combination, was safe and provoked an increase in<br />

anti-EGF antibody titers, produced clinical benefit, improved overall<br />

survival and progression free survival.<br />

2530 Poster Discussion Session (Board #18), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Phase I clinical trial <strong>of</strong> a genetically modified and oncolytic vaccinia virus<br />

GL-ONC1 with green fluorescent protein imaging. Presenting Author: Jesus<br />

Corral Jaime, Royal Marsden Hospital and Institute <strong>of</strong> Cancer Research,<br />

Sutton, United Kingdom<br />

Background: GL-ONC1 is a genetically engineered vaccinia virus attenuated<br />

by insertion <strong>of</strong> the RUC-GFP (Renilla luciferase and Aequorea green<br />

fluorescent protein fusion gene), beta-galactosidase (lacZ) and betaglucuronidase<br />

(gusA) reporter genes into the F14.5L, J2R (thymidine<br />

kinase) and A56R (hemaglutinin) loci. A phase I clinical trial <strong>of</strong> iv GL-ONC1<br />

was pursued to evaluate safety, tolerability, tumour delivery, neutralizing<br />

antibody development and anti-tumour activity. Methods: GL-ONC1 was<br />

administered to patients with advanced solid tumours at escalating doses<br />

(1�105 ,1�106 ,1�107 ,1�108 ,1�109 ,3�109 plaque-forming units<br />

(pfu) on day 1; 1.667�107 and 1.667�108 pfu on day 1-3 <strong>of</strong> a 28-day<br />

cycle) using a 3�3 dose escalation design. Green fluorescent protein (GFP)<br />

imaging was performed on skin rash and mucosal tumour lesions at<br />

baseline and after each cycle. Optional tumour biopsies were obtained for<br />

pharmacodynamic and viral delivery evaluation. Results: 27 patients (21<br />

males, median age 60 years) were treated. One <strong>of</strong> six patients at the 1x109 pfu dose level developed a dose-limiting, grade 3 rise in aspartate<br />

transaminase levels after a single infusion. Other reported adverse events<br />

(grade 1/2) included pyrexia (21), musculoskeletal pain (9), fatigue (8),<br />

nausea (7), and vomiting (4). Two patients developed skin rash during the<br />

first week <strong>of</strong> treatment, which appeared green by GFP and were positive to<br />

viral plaque assay (VPA). VPA <strong>of</strong> blood, urine, stool and sputum were<br />

negative for viral shedding in all but one patient who had positive shedding<br />

for 11 days. Increased neutralizing antibody titres were detected in all<br />

tested patients apart from one. Best response by RECIST was stable<br />

disease at 48 weeks (1), 24 weeks (3) and 8-12 weeks (5). A patient with<br />

squamous cell carcinoma <strong>of</strong> the tongue had a biopsy after 4 cycles which<br />

showed positive IHC staining <strong>of</strong> vaccinia virus. This is the first time that a<br />

virus is shown to be delivered to solid tumour after iv delivery. Conclusions:<br />

GL-ONC1 administered iv is well tolerated, with documented colonisation<br />

in patient tumour, and preliminary evidence <strong>of</strong> anti-tumour activity.<br />

2532 Poster Discussion Session (Board #20), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Association <strong>of</strong> breast cancer in men with exposure to 5-� reductase inhibitors: A<br />

RADAR report. Presenting Author: Steven M. Belknap, Department <strong>of</strong> Dermatology,<br />

Northwestern University Feinberg School <strong>of</strong> Medicine, Chicago, IL<br />

Background: Breast cancers in men (BCM) account for �1% <strong>of</strong> all breast<br />

cancers. Dihydrotestosterone (DHT) inhibits proliferation <strong>of</strong> normal and neoplastic<br />

mammary tissue and constrains the effect <strong>of</strong> estrogens. Finasteride (F) and<br />

dutasteride (D) are 5-� reductase inhibitors (5-�RIs) that reduce systemic and<br />

local dihydrotestosterone and cause gynecomastia in 1–3% <strong>of</strong> men. The package<br />

inserts for F and D state, “the relationship between long-term use <strong>of</strong> (finasteride/<br />

dutasteride) and male breast neoplasia is currently unknown.” F and D are<br />

marketed for treatment <strong>of</strong> symptomatic benign-prostatic hyperplasia. F is<br />

marketed for treatment <strong>of</strong> androgenetic alopecia. Methods: To detect disproportionality<br />

in the FDA MedWatch dataset, we calculated the empiric Bayes<br />

geometric mean (EBGM) for association <strong>of</strong> BCM with F or D. We also calculated<br />

the attributable risk <strong>of</strong> BCM exposed to F or D among men at an urban academic<br />

hospital (Northwestern Memorial Hospital) and at a rural healthcare system<br />

(Marshfield Clinic). Results: In the MedWatch dataset, we identified 33 reports <strong>of</strong><br />

F-associated BCM and 5 reports <strong>of</strong> D-associated BCM. For F–associated BCM,<br />

the EBGM was 58.95 (95% CI 24.47-81.76; p�0.0001). For D-associated<br />

BCM, the EBGM was 15.79 (95% CI 4.57-35.49; p�0.0001). The mean age<br />

for BCM after 5-�RI exposure was 70�11 years; 11/38 (29%) had gynecomastia.<br />

There were 38 cases <strong>of</strong> BCM associated with 5-�RI in the combined<br />

Northwestern and Marshfield cohort (see table below). Conclusions: We found a<br />

highly significant association between BCM and 5-�RI exposure in each <strong>of</strong> 6<br />

separate analyses (3 sources X 2 drugs), with an estimated 1 extra BCM per 564<br />

men exposed to 5-�RIs. We now plan to assess BRCA status and other risk<br />

factors. Given that 5-�RIs are marketed for control <strong>of</strong> lower urinary tract<br />

symptoms or for cosmetic purposes, it is not immediately obvious that use <strong>of</strong><br />

finasteride or dutasteride for their labeled indications would provide any net<br />

benefit.<br />

Risk <strong>of</strong> breast cancer in men with 5-�RI exposure.<br />

Breast cancer<br />

Risk/<br />

Yes No Total 1,000<br />

5-�RI exposure 38 17,161 17,199 2.21<br />

No 5-�RI exposure 576 1,319,183 1,319,759 0.436<br />

Total 614 1,336,344<br />

95% CI<br />

1,336,958<br />

Attributable risk per 1.77 (1.07–2.48)<br />

1,000 men<br />

Risk ratio 5.1 (3.6–7.0)<br />

Number needed to harm 564<br />

Chi-square 116.3<br />

p value p � 0.00001<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.<br />

149s


150s Developmental Therapeutics—<strong>Clinical</strong> Pharmacology and Immunotherapy<br />

2533 Poster Discussion Session (Board #21), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Effect <strong>of</strong> age on the pharmacokinetics <strong>of</strong> busulfan (Bu): An alliance study.<br />

Presenting Author: Jan Hendrik Beumer, University <strong>of</strong> Pittsburgh Cancer<br />

Institute, Pittsburgh, PA<br />

Background: Older acute myeloid leukemia (AML) and myelodysplastic<br />

syndrome (MDS) patients (pts) have been excluded from myeloablative<br />

regimens including agents such as busulfan because <strong>of</strong> the potential for<br />

non-disease related morbidity and mortality. Busulfan, <strong>of</strong>ten used as part <strong>of</strong><br />

myeloablative regimens, is cleared in part by glutathione–S-transferase and<br />

CYP3A4. We hypothesized that busulfan clearance (BuCL) in the elderly<br />

(� 60 years) would be reduced compared to that in young patients<br />

(� 60 years), thus potentially explaining differences in tolerability.<br />

Methods: AML and MDS pts in three CALGB bone marrow transplant studies<br />

across a wide age range (17-74) were treated with a conditioning regimen<br />

using IV busulfan, dosed at 12.8 mg/m2 in CALGB studies 10503 and<br />

19808 and 6.4 mg/m2 in CALGB 100103. Blood samples were collected<br />

for determination <strong>of</strong> busulfan plasma pharmacokinetics (PK). Plasma<br />

busulfan was quantitated by LC-MS. BuCL was determined using noncompartmental<br />

modeling, and normalized to actual (ABW), ideal (IBW) and<br />

corrected (CBW) body weight (kg). Differences between age groups were<br />

examined using the Wilcoxon rank sum test. Results: A total <strong>of</strong> 185 pts were<br />

accrued and signed consent, <strong>of</strong> which 174 provided useable PK data.<br />

Twenty-nine pts �60 years old (median age 66.3) had a significantly higher<br />

mean BuCL vs those � 60 years old (median age 49.8); BuCL 263 vs 186<br />

mL/min, p�0.0002; BuCL/ABW 3.22 vs 2.34 mL/min/kg, p�0.0001;<br />

BuCL/IBW 3.63 vs 2.91 mL/min/kg, p�0.0035; BuCL/CBW 3.50 vs 2.70<br />

mL/min/kg, p�0.0005. Inter-patient variability in clearance (CV%) was up<br />

to 47% in the elderly and up to 48% in young patients. Phenytoin use, a<br />

potential confounder, did not affect BuCL, regardless <strong>of</strong> weight normalization<br />

(p�0.74). Conclusions: Contrary to our hypothesis, BuCL is significantly<br />

higher in pts �60 years old compared to younger patients and does<br />

not explain the previously reported increase in Bu toxicity observed in the<br />

elderly.<br />

2535 Poster Discussion Session (Board #23), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Exposure-response (E-R) analysis <strong>of</strong> rilotumumab (R, AMG 102) plus<br />

epirubicin/cisplatin/capecitabine (ECX) in patients (pts) with locally advanced<br />

or metastatic gastric or esophagogastric junction (G/EGJ) cancer.<br />

Presenting Author: Min Zhu, Amgen Inc., Thousand Oaks, CA<br />

Background: R is an investigational, fully human monoclonal antibody to<br />

hepatocyte growth factor/scatter factor (HGF/SF), the only known MET<br />

receptor ligand. In a double-blind, placebo-controlled, phase 2 trial <strong>of</strong> 121<br />

pts with G/EGJ cancer, R (7.5 or 15 mg/kg) � ECX showed trends for<br />

improved overall survival (OS) and progression-free survival (PFS) compared<br />

to ECX alone (placebo, P). Methods: E-R analyses were performed<br />

using pharmacokinetic (PK), biomarker, efficacy, and safety data. A<br />

population PK model was used to evaluate covariate effects (eg, demographics,<br />

ECX, and MET status) on R PK and predict individual R exposure<br />

(trough steady-state Cmin [C]). The effect <strong>of</strong> C on OS/PFS was evaluated<br />

with Kaplan-Meier estimates and Cox proportional hazards models. Covariate<br />

effects for OS/PFS were evaluated with multivariate analysis. The<br />

relationships between adverse events <strong>of</strong> interest (AEs), lab values, and C<br />

were analyzed with descriptive statistics and linear regression models.<br />

Results: R showed linear PKs that were unaffected by ECX or MET levels.<br />

Pts who received R � ECX were divided into low C (C L ) and high C (CH )<br />

groups by the median C (94 �g/mL). Median PFS was 4.2, 4.5, and 6.9<br />

months in the P, CL , and CH groups; median OS was 8.9, 9.5, and 13.3<br />

months. Improved OS/PFS in CH and improved PFS in CH and CL groups (vs<br />

P) were seen after adjusting for baseline covariates. Pts with tumors that<br />

expressed high MET levels (METHigh : � 50% cells positive) showed greater<br />

improvement in OS in the CH vs CL group. No improvement in OS was seen<br />

in METLow pts (CH and CL groups) compared to placebo (see Table). No<br />

relationship was seen between R exposure and AEs or lab values.<br />

Conclusions: Higher R exposure was associated with improved OS/PFS in<br />

METHigh pts without increased toxicity, supporting further study <strong>of</strong> R � ECX<br />

in G/EGJ cancer.<br />

C group N<br />

Median OS -<br />

mo (80% CI)<br />

Adjusted HR<br />

(95% CI) p value<br />

METHigh P 11 5.7 (4.5,10.4) - -<br />

CL 14 9.3 (8.0, 11.1) 0.50 (0.18, 1.39) 0.183<br />

CH 13 13.4 (13.3, NE) 0.12 (0.03, 0.45) 0.002<br />

METLow P 17 NE (8.5, NE) - -<br />

CL 15 8.6 (4.9, 9.9) 2.39 (0.90, 6.33) 0.081<br />

CH 20 11.6 (7.8, 12.5) 1.52 (0.59, 3.95) 0.389<br />

NE: Not estimable.<br />

2534 Poster Discussion Session (Board #22), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

A first-in-human, phase I safety and pharmacokinetic study <strong>of</strong> genz-<br />

644282, a non-camptothecin topoisomerase I inhibitor, in patients with<br />

advanced solid tumors. Presenting Author: Hyo S. Han, H. Lee M<strong>of</strong>fitt<br />

Cancer Center & Research Institute, Tampa, FL<br />

Background: The approved topoisomerase I (TopI) inhibitors (irinotecan and<br />

topotecan) are camptothecin derivatives. The lactone ring structure <strong>of</strong><br />

camptothecins negatively impacts their clinical potential.Genz-644282 is<br />

a novel non-camptothecin that induces TopI-DNA cleavage complexes at<br />

similar as well as unique genomic positions; that are more persistent. This<br />

study was designed using a pharmacokinetic-pharmacodynamic (PK-PD)<br />

model to predict the maximum tolerated dose (MTD). Methods: A PK-PD<br />

relationship <strong>of</strong> Genz-644282 to inhibit tumor growth was derived (Simeoni,<br />

Can Res 2004). Using an accelerated titration design, cohorts <strong>of</strong> 1, 3 or 6<br />

patients received 3 weekly doses <strong>of</strong> Genz-644282 on a 28 day schedule<br />

starting with 0.5 mg/m2 . After MTD on the 28 day schedule was exceeded,<br />

21 day schedule was initiated. Results: 49 patients (N�44 data available:<br />

26M :18F) were dosed. Tumor types were colorectal (15), breast (5), small<br />

cell lung (SCLC 3), non-small cell lung (4), others (17). As predicted from<br />

the PK-PD model 8 cohorts were evaluated on the 28 day schedule before<br />

defining the MTD. The MTD was 8 mg/m2 and 9 mg/m2 for the 28 day and<br />

21 day schedules, respectively. Dose limiting toxicities that determined the<br />

MTD were: gr 4 thrombocytopenia (n�2); gr 2 thrombocytopenia (n�1)<br />

and gr 4 neutropenia (n�1) both <strong>of</strong> which resulted in �72h delays in<br />

initiating cycle 2. Treatment emergent adverse events (�10%) were<br />

nausea (45%), fatigue (39%), anorexia (32%), anemia (27%), vomiting<br />

(23%), thrombocytopenia (18%), diarrhea (16%), dehydration (16%),<br />

hyperglycemia (16%), cough (16%), dyspnea (14%), depression (11%)<br />

and hyponatremia (11%). Efficacy data suggest 2 responders with SCLC (1<br />

minor response;1 complete response), and 2 patients (breast, gastric) with<br />

stable disease (� 6 months). PK data show a Genz-644282 half-life <strong>of</strong><br />

approximately 50 h, a linear dose-exposure relationship, and no accumulation<br />

in between doses. Conclusions: Genz-644282 is a non-camptothecin<br />

TopI inhibitor in phase I development with ongoing expansion phase. The<br />

emerging pk, efficacy and safety data are suggestive <strong>of</strong> a distinct clinical<br />

pr<strong>of</strong>ile <strong>of</strong> Genz-644282 from the camptothecins.<br />

2536 Poster Discussion Session (Board #24), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Pharmacological interactions <strong>of</strong> TKIs with the P-gp drug transport protein.<br />

Presenting Author: Sandra Roche, NICB, Dublin City University, Dublin,<br />

Ireland<br />

Background: Tyrosine Kinase Inhibitors (TKIs) can interact with drug<br />

transport proteins. P-gp is a transporter with two important roles in cancer<br />

drug therapy. If overexpressed in tumour cells it can cause drug resistance.<br />

However, P-gp, expressed in tissues as part <strong>of</strong> normal drug clearance<br />

mechanisms, is also involved in termination <strong>of</strong> drug action. Hence,<br />

TKI-mediated interactions with P-gp have significant therapeutic consequences.<br />

Methods: P-gp over-expressing cancer cell lines were used to<br />

determine the inhibitor or substrate status <strong>of</strong> tyrosine kinase inhibitors<br />

(erlotinib, gefitinib, lapatinib, dasatinb, neratinib, afatinib and pazopanib).<br />

Cell proliferation assays in combination with a potent P-gp inhibitor, or<br />

P-gp substrate were also employed. Findings were augmented using<br />

LC-MS-based quantitation <strong>of</strong> cellular levels <strong>of</strong> target drugs. Results: We<br />

summarise our findings <strong>of</strong> four distinct interactions with P-gp among<br />

various TKIs. Some agents have little interaction at conventional doses;<br />

others can act as P-gp inhibitors without being substrates; substrates<br />

without being inhibitors or substrates which also prevent the actions <strong>of</strong> the<br />

transporter.Each<strong>of</strong> the investigated TKIs has a distinct relationship with<br />

P-gp. As examples, lapatinib is an inhibitor but not a substrate, dasatinib is<br />

a substrate but not an inhibitor, while pazopanib has little interaction with<br />

P-gp. Other agents also have an effect on or are affected by P-gp to varying<br />

amounts with some <strong>of</strong> these interactions likely to be suprapharmacological.<br />

Conclusions: P-gp protein has important roles both in resistance and drug<br />

toxicology, hence, a clear understanding <strong>of</strong> the interaction <strong>of</strong> emerging<br />

drugs with this transporter is vital. Agents which are inhibitors <strong>of</strong> P-gp may<br />

have applications in drug resistance circumvention but may also greatly<br />

exacerbate the toxicity <strong>of</strong> concurrently administered P-gp substrate cytotoxics;<br />

conversely the activity <strong>of</strong> P-gp substrate TKIs may be reduced by<br />

tumour overexpression <strong>of</strong> the transporter. Hence in vitro screening <strong>of</strong><br />

TKI-transporter interactions may identify putative TKI resistance mechanisms,<br />

help guide the development <strong>of</strong> combination schedule trials and/or<br />

reducing unwanted treatment side effects.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


Developmental Therapeutics—<strong>Clinical</strong> Pharmacology and Immunotherapy<br />

2537^ Poster Discussion Session (Board #25), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Pharmacogenetic study in metastatic colorectal cancer (CRC) patients<br />

receiving third-line panitumumab-irinotecan: A GERCOR ancillary study.<br />

Presenting Author: Marie-Christine Etienne-Grimaldi, Centre Antoine-<br />

Lacassagne, Nice, France<br />

Background: Test the predictive value <strong>of</strong> gene polymorphisms potentially<br />

linked to panitumumab and irinotecan pharmacodynamics. Methods: 45<br />

patients with metastatic CRC refractory to standard therapy were enrolled<br />

in this ancillary pharmacogenetic study as part <strong>of</strong> a phase II trial that<br />

included 63 patients. Inclusion required wild-type KRAS tumor status<br />

(codons 12-13, analyses performed in each center). After inclusion, central<br />

KRAS re-assessment was performed (codons 12-13-59-61) and 9 patients<br />

over 45 were found to carry a KRAS mutation. Patients received panitumumab<br />

(6 mg/kg, day 1) associated with irinotecan (180 mg/m2 , day 1),<br />

Q2W until disease progression or unacceptable toxicity. Analyzed polymorphisms<br />

on blood DNA were : EGFR (CA repeats in intron 1, -216G�T,<br />

-191C�A), EGF (61A�G), CCND1 (870A�G), UGT1A1 (*28). Results:<br />

Cutaneous toxicity imputable to panitumumab i.e. folliculitis and paronychia<br />

was not linked to EGFR, EGF or CCND1 polymorphisms. Diarrhea<br />

(12 grade 1, 13 grade 2, 8 grade 3) was linked to CCND1 polymorphism:<br />

22% grade 2-3 in AA patients vs 63% in AG�GG patients (p � 0.007). As<br />

expected, neutropenia (6 grade 1, 4 grade 2, 5 grade 3) was more frequent<br />

in patients bearing the *28 allele <strong>of</strong> UGT1A1 gene (33% grade 2-3 vs 5%<br />

in *1/*1, p � 0.020). In these 45 patients, response rate, progression-free<br />

survival and overall survival (OS) were only significantly related to reassessed<br />

KRAS status. Of note, a tendency for a longer OS was observed in<br />

patients bearing the *28 allele <strong>of</strong> UGT1A1 gene (p � 0.091, analysis<br />

adjusted for KRAS). Interestingly, the frequency <strong>of</strong> KRAS mutation was<br />

higher in patients with long CA repeats in EGFR gene intron 1: 54.5% i.e. 6<br />

mutated patients among the 11 patients with both alleles �17 vs 10.5%<br />

i.e. 2 mutated patients among the 19 patients with intermediary alleles vs.<br />

6.7% i.e. 1 mutated patient among the 15 patients with both alleles �17<br />

(p � 0.004). Conclusions: Present data indicate that A870 allele <strong>of</strong> CCND1<br />

gene (40% AA patients) may protect from diarrhea induced by panitumumab-irinotecan.<br />

Also, these results report an original relationship<br />

between EGFR polymorphism in intron 1 and KRAS mutation status that<br />

merits further confirmation.<br />

2539 General Poster Session (Board #1B), Mon, 8:00 AM-12:00 PM<br />

Phase Ib study <strong>of</strong> plitidepsin (APL) with gemcitabine (GEM) in refractory<br />

solid tumors and lymphoma patients. Presenting Author: Mark N. Stein,<br />

Cancer Institute <strong>of</strong> New Jersey, New Brunswick, NJ<br />

Background: Combination chemotherapy (CT) regimens may improve the<br />

outcome <strong>of</strong> patients with advanced metastatic disease. APL (a marinederived<br />

cyclodepsipeptide) is a cytotoxic agent that induces apoptosis by<br />

JNK-pathway activation and increased oxidative stress; it has also shown<br />

anti-angiogenic properties in vitro. GEM has a different mechanism <strong>of</strong><br />

action and toxicity pr<strong>of</strong>ile from APL, making this combination appropriate<br />

for clinical testing. Moreover, APL has been shown pre-clinically to<br />

potentiate the anti-tumor effect <strong>of</strong> GEM. Methods: Patients with ECOG-PS<br />

� 1 received escalating doses <strong>of</strong> APL/GEM (1.8/750, 1.8/1000, 2.4/1000<br />

and 3.0/1000 mg/m2 ) on days 1, 8 and 15 every four weeks. The primary<br />

objective was to determine <strong>of</strong> the maximum tolerated dose (MTD) and the<br />

recommended dose (RD) for APL/GEM. Secondary objectives included<br />

characterization <strong>of</strong> safety pr<strong>of</strong>ile, pharmacokinetics (PK) and anti-tumor<br />

activity <strong>of</strong> APL/GEM. Results: 34 patients were included and 33 treated (23<br />

solid tumors and 11 lymphomas) with a median age <strong>of</strong> 59 years (r: 28–86),<br />

median prior CT lines 3 (r:1-7) and a median <strong>of</strong> 2 cycles (r:1-12). Relative<br />

dose intensity was 66.7% (r: 37.7-85.7%) and 80.9% (r: 56.1-100.6%)<br />

for APL and GEM, respectively. The MTD was 3.0/1000 mg/m2 ,as2/6<br />

patients experienced dose limiting toxicities (DLTs) [grade (G) 4 thrombocytopenia<br />

and 2 missed doses due to G2 ALT/AST]. The RD was 2.4/1000<br />

mg/m2 , with no patients experiencing DLTs. Adverse events (AEs) were<br />

mainly G 1-2 anemia, thrombocytopenia and ALT/AST increase. Six<br />

patients with solid tumors had stable disease (SD) � 3 months, one patient<br />

with NK T-cell lymphoma had a complete response, a patient with Hodgkin<br />

disease (HD) had a partial response and three other patients with HD had<br />

SD � 3 months with tumor decrease at the RD. No relevant drug-drug<br />

interaction was observed for the dose levels tested. Conclusions: APL/GEM<br />

at the RD can be safely combined. Objective responses were observed in<br />

patients with HD and NK T-Cell lymphoma. Further studies <strong>of</strong> this<br />

combination in lymphoma are warranted.<br />

151s<br />

2538 General Poster Session (Board #1A), Mon, 8:00 AM-12:00 PM<br />

A mismatch between methods and objectives in phase I drug development:<br />

Statistical limitations and personalized medicine—A California Cancer<br />

Consortium (CCC) study. Presenting Author: Paul Henry Frankel, City <strong>of</strong><br />

Hope, Duarte, CA<br />

Background: Patient variation in drug response and toxicity impacts all<br />

phases <strong>of</strong> drug development. While detailed sample-size calculations and<br />

analysis based on statistical methodology are critical for addressing<br />

variation in late stage trials, phase I studies pose unique and largely<br />

unsolved challenges related to variability. Changes in patient selection<br />

during the study, small sample-sizes and large patient variation in toxicity<br />

are exactly the kind <strong>of</strong> problems that statistical methods cannot address in<br />

small, isolated phase I studies, regardless <strong>of</strong> apparent mathematical rigor.<br />

We have documented these problems via a physician survey. Methods: A<br />

10-question anonymous survey was sent to 670 oncologists at National<br />

Comprehensive Cancer Network (NCCN) and CCC institutions via an online<br />

survey. 19 % (126/670) <strong>of</strong> the oncologists polled responded. 78/126<br />

(62%) specialized in Medical Oncology. The number <strong>of</strong> years in practice<br />

varied from 2-45 yrs with a median <strong>of</strong> 17 yrs. Results: a) 66% <strong>of</strong> all<br />

respondents stated that non-DLT toxicities on one dose level would impact<br />

their patient selection on the following dose level, conflicting with the<br />

assumption <strong>of</strong> random patient selection implicit in simulations used in<br />

evaluating statistical designs. b) Only 13% stated a desire to target a<br />

non-heme toxicity as high as 20% grade 3, while 87% desired a 10% or<br />

less grade 3 rate; c) More than half the respondents would prefer not to<br />

escalate if 3/3 patients experienced grade 2 LFTs; d) 82% <strong>of</strong> the<br />

respondents thought the appropriate target toxicity differed for patients<br />

depending on their potential for becoming a surgical candidate, furthering<br />

the need for personalized dosing. Conclusions: Statistical methods in phase<br />

I trials are unable to address many <strong>of</strong> the salient features <strong>of</strong> phase I study<br />

conduct and investigator goals. We will present several approaches we have<br />

initiated to address these limitations, and present future plans to help<br />

produce a more reliable estimate <strong>of</strong> a recommended dose. Supported in<br />

part by NCI grants U01CA062505, N01-CM-62209, and NCCN data<br />

collection assistance.<br />

2540 General Poster Session (Board #1C), Mon, 8:00 AM-12:00 PM<br />

Prediction <strong>of</strong> early death among patients (pts) enrolled in phase I trials:<br />

Development and validation <strong>of</strong> a new model based on platelet count and<br />

albumin level. Presenting Author: Anne Ploquin, Centre Oscar Lambret,<br />

Lille, France<br />

Background: Selecting pts with “sufficient life expectancy” for phase I<br />

oncology trials remains challenging. The Royal Marsden Model (RMM;<br />

Arkenau, JCO 2009) identified high-risk pts as those with �2 <strong>of</strong>: albumin<br />

(ALB) �35g/l; LDH �ULN; more than 2 metastatic sites. The RMM was<br />

assessed in 1845 pts treated in phase I trials by members <strong>of</strong> the European<br />

New Drug Development Network (ENDDN). The present study aimed to<br />

develop an alternative prognostic model using a different methodology and<br />

to compare its performance with the RMM. Methods: The primary endpoint<br />

was 90-day mortality rate (90-DMR). The new model was developed from<br />

the ENDDN database using CHAID, an exploratory non-parametric data<br />

analysis method evaluating the relationship between a dependent variable<br />

(90-DMR) and possible predictive variables through a decision-tree analysis.<br />

The ROC characteristics and calibration <strong>of</strong> both methods were then<br />

validated in the independent EORTC Database (n�341 pts). Results: The<br />

CHAID method identified low and high-risk groups with 90-DMR <strong>of</strong> 9.5% vs<br />

37.5%. High-risk pts had ALB�33g/L or ALB�33g/L but platelet count<br />

�400.000/mm3 . Applying both models to the validation dataset; the rates<br />

<strong>of</strong> correctly-classified pts were 0.86 [CI-95% 0.82-0.90] vs. 0.67 [CI-95%<br />

0.60-0.74], with the CHAID model and RMM respectively. The CHAID<br />

model had higher specificity (0.90 vs. 0.65), but lower sensitivity (0.36 vs.<br />

0.93) than the RMM. Discriminative slopes were similar for the CHAID<br />

model and RMM (19.4% and 19.3%, respectively). The negative predictive<br />

value (NPV; correct identification <strong>of</strong> pts surviving 90 days) was similar for<br />

the CHAID model and RMM (0.94 [0.91-0.97] and 0.99 [0.94-1.00]<br />

respectively). Calibration, assessed by the Brier score, was slightly better<br />

with the CHAID model (0.001 and 0.098, respectively). Conclusions: In<br />

selecting pts for phase I trials arguably an important criterion is NPV; the<br />

CHAID model and RMM provided a similar high level <strong>of</strong> NPV but the CHAID<br />

model gave a better rate <strong>of</strong> correctly-classified patients. Both models<br />

improved the screening process and reduce the attrition rate in phase I<br />

trials.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


152s Developmental Therapeutics—<strong>Clinical</strong> Pharmacology and Immunotherapy<br />

2541 General Poster Session (Board #1D), Mon, 8:00 AM-12:00 PM<br />

Relationship <strong>of</strong> variability in tumor measurement and response assessment.<br />

Presenting Author: Binsheng Zhao, Department <strong>of</strong> Radiology, Columbia<br />

University Medical Center, New York, NY<br />

Background: RECIST is widely used to evaluate anti-cancer therapy efficacy.<br />

This study explored variability in reporting tumor change and<br />

response to therapy due to both target lesion selection and measurement.<br />

Methods: 2256 measurements were performed in CT scans <strong>of</strong> chest,<br />

abdomen and pelvis from 30 patients retrospectively taken from a multicenter<br />

Phase II/III colorectal clinical trial. Using RECIST, three radiologists<br />

interpreted baseline, 6-wk and 12-wk scans in the following manner: (1)<br />

Radiologists independently selected and measured target lesions, (2) one<br />

radiologist’s target lesions were re-measured by the other two and (3) one<br />

radiologist re-measured the same scans in the above manner at an interval<br />

<strong>of</strong> greater than a month to prevent memory recall. Measurement variability<br />

in total tumor burden (TTB) on relative changes at 6-wk and 12-wk from<br />

baseline was analyzed for inter- and intra-reader target lesion selection,<br />

inter- and intra-reader measurement using Bland-Altman method, and<br />

agreements on RECIST categorical responses were assessed by kappa<br />

coefficient. Results: When the same target lesions were used, inter- and<br />

intra-reader variability in TTB on relative changes at 6-wk and 12-wk from<br />

baseline were similar; all had 95% limits <strong>of</strong> agreement within (-15%,<br />

15%). Kappa coefficients for RECIST were 0.74 (6-wk) and 0.87 (12-wk)<br />

for inter-reader and 0.64 (6-wk) and 0.88 (12-wk) for intra-reader to report<br />

responses. When radiologists independently selected and measured target<br />

lesions, variability in relative changes was within (-17%, 16%) at 6-wk and<br />

(-24%, 23%) at 12-wk for inter-reader and (-16%, 16%) at 6-wk and<br />

(-14%, 18%) at 12-wk for intra-reader interpretations. Kappa coefficients<br />

were 0.66 (6-wk) and 0.75 (12-wk) for inter-reader and 0.63 (6-wk) and<br />

0.80 (12-wk) for intra-reader to report responses. Conclusions: Differences<br />

exist in measuring tumor change. The magnitude <strong>of</strong> change in categorical<br />

RECIST response is quantifiable. The largest differences are when radiologists<br />

independently select and measure target lesions, the smallest when<br />

one radiologist repeats measurements on identical target lesions. The<br />

variability may impact the reporting <strong>of</strong> categorical responses and trial<br />

results, especially in a single arm study.<br />

2543 General Poster Session (Board #1F), Mon, 8:00 AM-12:00 PM<br />

<strong>Clinical</strong> importance <strong>of</strong> including new and nontarget lesion assessment <strong>of</strong><br />

disease progression (PD) to predict overall survival (OS): Implications for<br />

randomized phase II study design. Presenting Author: William Leonard<br />

Mietlowski, Novartis Oncology, Florham Park, NJ<br />

Background: Fridlyand (2011) retrospectively compared PFS vs. change in<br />

tumor burden as a primary endpoint in phase II non-small cell lung cancer<br />

(NSCLC) trials to inform phase III decision making and found the use <strong>of</strong><br />

PFS was superior. Since the classic tumor burden model only uses<br />

measurements <strong>of</strong> target lesions, we investigated whether the model could<br />

be strengthened by incorporating new and non-target lesion progression.<br />

The ability to use a strong tumor burden model has the benefit <strong>of</strong> potentially<br />

earlier decision making and considerable timeline savings. Methods: We<br />

analyzed five phase III trials <strong>of</strong> combination chemotherapy � targeted<br />

therapies with an OS primary endpoint: 1st, 2nd line NSCLC (ATTRACT-1,<br />

-2), 1st, 2nd line colorectal carcinoma (CONFIRM-1, -2), and 2nd line<br />

ovarian cancer (EPO906A2303). We applied Cox’s proportional hazards<br />

model to OS using the covariates <strong>of</strong> baseline tumor burden, 1st tumor<br />

assessment percentage change from baseline, new lesions, and non-target<br />

PD. Results: See table. Conclusions: We show that predictive models for OS<br />

should consider new and non-target lesions for PD, as well as target lesion<br />

tumor burden, findings independently corroborated by Suzuki (2011). We<br />

propose a longitudinal rank-based randomized phase II design, ranking a<br />

patient’s risk <strong>of</strong> death, differentially weighting PDs by type and time <strong>of</strong> PD,<br />

and percentage change in tumor burden. This may be more informative for<br />

phase II decision making for phase III trials based on OS, than PFS which<br />

only uses time <strong>of</strong> PD. Further studies with other tumor types and treatment<br />

modalities are warranted.<br />

Study<br />

Baseline<br />

tumor burden*<br />

Estimated hazard ratio (Wald test p value)<br />

% change<br />

from baseline* New lesion<br />

Non-target<br />

lesion PD<br />

ATTRACT-1 1.36 (p�0.001) 1.37 (p�0.001) 3.37 (p�0.001) 1.95 (p�0.001)<br />

ATTRACT-2 1.21 (p�0.001) 1.49 (p�0.001) 3.91 (p�0.001) 1.19 (p�0.332)<br />

EPO906A2303 1.09 (p�0.053) 1.12 (p�0.046) 2.20 (p�0.001) 1.67 (p�0.001)<br />

CONFIRM-1 1.31 (p�0.001) 1.38 (p�0.001) 3.02 (p�0.001) 1.40 (p�0.087)<br />

CONFIRM-2 1.41 (p�0. 001) 1.47 (p�0.001) 2.54 (p�0.001) 1.81 (p�0.001)<br />

* Unit <strong>of</strong> measurement is one standard deviation.<br />

2542 General Poster Session (Board #1E), Mon, 8:00 AM-12:00 PM<br />

A multicenter phase II neoadjuvant trial <strong>of</strong> bevacizumab combined with<br />

docetaxel plus carboplatin in the treatment <strong>of</strong> triple-negative breast cancer:<br />

Korean Cancer Study Group (KCSG-BR 0905, NCT 01208480). Presenting<br />

Author: Hye Ryun Kim, Division <strong>of</strong> Medical Oncology, Department <strong>of</strong><br />

Internal Medicine, Yonsei University College <strong>of</strong> Medicine, Seoul, South<br />

Korea<br />

Background: Triple negative breast cancer (TNBC) is dismal disease that is<br />

ineligible for endocrine or anti-HER2 therapy. We have conducted a phase<br />

II neoadjuvant trial to evaluate efficacy and tolerability <strong>of</strong> bevacizumab<br />

combined with docetaxel plus carboplatin in TNBC. Methods: Women with a<br />

histologically confirmed stage II or III TNBC were eligible. Hormone<br />

receptors and HER2 negativity were confirmed by immunohistochemistry<br />

and/or fluorescence in situ hybridization. Patients received 6 cycles <strong>of</strong><br />

docetaxel 75 mg/m2 , carboplatin AUC 5, and bevacizumab 15 mg/kg on<br />

day1 every 21 days. Bevacizumab was omitted in the last cycle to avoid<br />

wound complication. The primary endpoint was pathologic complete<br />

response (pCR) rate in both breast and axillary lymph node and secondary<br />

endpoints were clinical response rate, toxicity pr<strong>of</strong>iles, and breast conserving<br />

surgery (BCS) rate. Results: Forty-five TNBC patients were recruited<br />

from 7 institutes in the Korean Cancer Study Group (KCSG) between<br />

October 2010 and August 2011. The median age <strong>of</strong> the patients was 45<br />

years (30-72 years). The T1, T2, and T3 were 4.4 %, 68.9 %, and 26.7 %,<br />

respectively and axillary lymph node was positive in 80% by breast MRI.<br />

Among 45 patients, 44 patients completed 6 cycles <strong>of</strong> therapy followed by<br />

surgery and one patient was withdrawn due to patient’s refusal <strong>of</strong> further<br />

therapy after 3rd cycle. The pCR rate was 42.2 % (19/45) and clinical<br />

response rate was 95.5 % (43/45) (CR, n� 6; PR, n�37; SD, n�1; not<br />

evaluable, n�1) based on RECIST criteria 1.1. BCS was undertaken in<br />

77.7 % (35/45). The grade 3 or 4 adverse events were neutropenia (38),<br />

febrile neutropenia (4), vomiting (3), nausea (2), anemia (1), thrombocytopenia<br />

(1), stomatitis (1), gastrointestinal bleeding (1), and increased ALT<br />

(1). Only one patient experienced delayed wound healing after breast<br />

surgery. Conclusions: Bevacizumab in combination with docetaxel and<br />

carboplatin as neoadjuvant treatment provided an encouraging pCR rate<br />

(42.2 %) and a negligible wound healing problem after surgery. The<br />

adverse events were manageable.<br />

2544 General Poster Session (Board #1G), Mon, 8:00 AM-12:00 PM<br />

How to run survival-endpoint phase II trials with 25% to 40% fewer<br />

patients. Presenting Author: Michael J. Schell, H. Lee M<strong>of</strong>fitt Cancer<br />

Center & Research Institute, Tampa, FL<br />

Background: New molecular information, whether arising from expression<br />

pr<strong>of</strong>iling, identification <strong>of</strong> key mutations, or proteomic analyses is making<br />

the personalization <strong>of</strong> therapy increasingly feasible. This leads to the<br />

fragmentation <strong>of</strong> common disease entities into multiple molecular subtypes.<br />

Having sufficient numbers <strong>of</strong> patients to demonstrate the effectiveness<br />

<strong>of</strong> novel targeted therapies for each subtype has therefore become<br />

challenging, essentially impeding progress. Methods: Use <strong>of</strong> sample size<br />

determination s<strong>of</strong>tware allows for comparison <strong>of</strong> phase II trials with a target<br />

survival proportion at a given time T based upon Kaplan-Meier versus<br />

exponential survival methods. Results: The use <strong>of</strong> exponential survival<br />

fitting methods, in lieu <strong>of</strong> the currently implemented trial designs, to single<br />

arm phase II studies can routinely reduce patient numbers by 25-40%<br />

(Table) without compromising the statistical power. The biggest advantage<br />

is that survival information both before and after time T reduces the<br />

variability for the exponential fit estimate. This approach will lead to the<br />

saving <strong>of</strong> financial and patient resources for researchers. Moreover, the<br />

time saved will accelerate the approval <strong>of</strong> agents making the proposed trial<br />

designs ethically attractive. However, this approach is only applicable if the<br />

exponential distribution assumption behind the approach is valid.<br />

Conclusions: The exponential survival fitting method should replace the<br />

current standard approach in all instances unless the exponential distribution<br />

assumption is known to be false. Detailed examples and analysis will<br />

be presented.<br />

Percent savings obtained by using the exponential fit approach.<br />

(T,T*)<br />

HR (1,1) (2,2) (3,3) (4,4) (1,1.5) (1,2) (2,3) (2,4)<br />

2.0 5 15 25 36 30 41 31 38<br />

1.83 8 15 26 40 31 42 30 36<br />

1.67 9 12 28 38 32 42 28 33<br />

1.50 6 16 23 38 30 40 30 36<br />

Avg. 7 14 26 38 31 41 30 36<br />

T is scaled so T�1 is the median survival time; T* is the time through which the<br />

exponential fit is used; HR is the ratio <strong>of</strong> the alternative to null hypothesis<br />

median survival times.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


Developmental Therapeutics—<strong>Clinical</strong> Pharmacology and Immunotherapy<br />

2545 General Poster Session (Board #2A), Mon, 8:00 AM-12:00 PM<br />

Feasibility and safety <strong>of</strong> sequential research-related tumor core biopsies in<br />

clinical trials with anti-angiogenic and targeted therapies. Presenting<br />

Author: Jung-min Lee, Medical Oncology Branch, Center for Cancer<br />

Research, National Cancer Institute, Bethesda, MD<br />

Background: There has been increasing interest in serial/paired research<br />

biopsies (bx) in studies <strong>of</strong> molecularly targeted therapies. Definition <strong>of</strong><br />

optimal target tissue and patient characteristics for safe and feasible<br />

research tumor bx in the era <strong>of</strong> anti-angiogenic and targeted therapies is<br />

needed. Methods: IRB-approved review <strong>of</strong> retrospective clinical data<br />

including patient demographics, pre-and post-procedure and recovery<br />

notes, and radiographic bx images. The cohort consisted <strong>of</strong> 142 pts<br />

scheduled for serial bx enrolled in phase 1 (5) and phase 2 (1) studies.<br />

Tumor core bx were obtained percutaneously under local anesthesia using a<br />

16 or 18g needles under US or CT visualization by interventional radiologists.<br />

Results: Paired tumor core bx were collected in 96 pts (68%) and<br />

baseline only in 42 pts (32%). Bx were aborted in 4 pts due to cystic mass<br />

(3) and safety (1). 132 pts were female and 10 pts were male, median age<br />

56 yrs (27-78), median BMI 25.8 (14.4-46.2), ECOG PS 0-2, and normal<br />

end-organ function. All pts had recurrent metastatic cancers (gynecologic<br />

121, breast 6, sarcoma 5, melanoma 4, colon 2, adrenal 1, mesothelioma<br />

1, RCC 1, papillary thyroid ca 1). 67 pts (51%) were on bevacizumab<br />

(bev)-based therapy at the time <strong>of</strong> their 2nd /3rd bx (bev � sorafenib; bev �<br />

dasatinib); others received gefitinib (27), imatinib (23), vandetinib (9), or<br />

olaparib � carboplatin (16). Median tumor size was 2.7cm (1-14.5cm). Bx<br />

sites were abd/pelvic masses 42, liver 40, lymph nodes 42, abd/chest wall<br />

12, lung 4 (3 pleural, 1 parenchymal), and psoas muscle 1. Minor<br />

complications were observed in four pts (3%; uncomplicated liver subcapsular<br />

hematoma [1]; vasovagal syncope responding to fluids [2]; uncomplicated<br />

pneumothorax without intervention [1]) and there were no major<br />

complications. The median number <strong>of</strong> tumor cores was 3 (1-6) at the<br />

baseline, 2nd (during or at the end <strong>of</strong> the first cycle) and 3rd time point<br />

(during cycle 2). Conclusions: Core needle bx <strong>of</strong> deep internal lesions such<br />

as intra-abdominal masses and retroperitoneal lymph nodes can be<br />

obtained percutaneously under imaging guidance. Sequential tumor bx are<br />

feasible and safe for pts on anti-angiogenic and chemotherapeutic agents.<br />

2547 General Poster Session (Board #2C), Mon, 8:00 AM-12:00 PM<br />

Predictive model for survival and toxicity in early-phase trials in hematology.<br />

Presenting Author: Rahima Jamal, University <strong>of</strong> Montreal, Montreal,<br />

QC, Canada<br />

Background: Strict criteria are used to limit toxicity for patients (pts)<br />

enrolled in phase I/II clinical trials, but the ability to survive beyond 12<br />

weeks, a common inclusion criterion, is subjective and error-prone. A<br />

prognostic score with 3 variables was designed and validated as an<br />

independent predictor <strong>of</strong> OS in pts with solid tumors included in phase I<br />

trials (Arkenau, JCO 2009). We examined the ability <strong>of</strong> objective measures<br />

to predict OS and risk <strong>of</strong> grade �3 toxicity in pts with hematologic<br />

malignancies enrolled in early-phase trials. Methods: A retrospective<br />

analysis was conducted on 290 pts in Phase I (79 pts) and II (211 pts)<br />

trials from the NCIC - <strong>Clinical</strong> Trials Group and our institution from 1995 to<br />

2009. Only pts with survival data up to 90 days were included. Univariate<br />

model (UVA) was used to identify factors significantly associated with OS. A<br />

Cox proportional hazards model (MVA) included all factors identified from<br />

the UVA. Six prognostic factors were identified in the MVA, and each<br />

assigned a score <strong>of</strong> 0 or 1. Pts were categorized as high or low risk based on<br />

the total scores that they received, �3 were assigned to the high risk group,<br />

and a �2 to the low risk group. Kaplan-Meier method was used to generate<br />

the survival distribution for the high and low risk groups. Multivariate<br />

logistic regression was used to identify the risk factors for grade �3 toxicity.<br />

Results: The overall median survival was 238 days (95% CI 237 to 331),<br />

and 27% <strong>of</strong> pts had grade � 3 toxicity. In the MVA, albumin (alb), alkaline<br />

phosphatase (ALP), lactate dehydrogenase (LDH), lymphocytes, platelets<br />

(plts) and diagnosis were significantly associated with OS. Pts in the low<br />

risk group had a median survival <strong>of</strong> 10.6 months, relative to 2.4 months<br />

among pts in the high risk group. Survival at 90 days was 80% in lower risk<br />

vs. 36% in the high risk group. In addition, the MVA for toxicity showed that<br />

alb, ALP, LDH, plts and performance status were significantly associated<br />

with grade �3 toxicity. Conclusions: Our model predicts OS, 90 day survival<br />

and risk <strong>of</strong> grade � 3 toxicity among pts with hematologic malignancies<br />

entered in Phase I/II trials. Future work will include a linear predictor score<br />

based on MVA to refine the prediction model. The model will also be<br />

validated using another dataset.<br />

153s<br />

2546 General Poster Session (Board #2B), Mon, 8:00 AM-12:00 PM<br />

Phase I immunotherapy trial using glioblastoma apoptotic body-pulsed<br />

dendritic cells. Presenting Author: Christopher L. Moertel, University <strong>of</strong><br />

Minnesota, Minneapolis, MN<br />

Background: We recently reported that tumor cell vaccines cultured in 5%<br />

oxygen (O2) had enhanced the immunogenicity relative to those grown in<br />

standard atmospheric O2. In order to develop an “<strong>of</strong>f-the-shelf” source <strong>of</strong><br />

whole cell antigen we screened a panel <strong>of</strong> primary glioblastoma (GBM) cell<br />

lines, leading to identification <strong>of</strong> (GBM6) as an ideal candidate. GBM6 was<br />

extensively characterized and shown to express glioma-associated antigens<br />

(IL13Ra2, Sox2, Epha2, etc.). A Phase I clinical trial was initiated to<br />

evaluate the safety and feasibility <strong>of</strong> a vaccine consisting <strong>of</strong> dendritic cells<br />

(DC) pulsed with apoptotic bodies from GBM6 grown in 5% O2. Methods:<br />

Patients ranging from 3 to 71 years with recurrent GBM (n�6) or<br />

ependymoma (n�1) were enrolled. Monocytes were collected via apheresis,<br />

matured into DC and pulsed with apoptotic bodies derived from GBM6.<br />

The first three patients received escalating doses <strong>of</strong> DC (5x106 , 10x106 ,<br />

and 15x106 ), the remainder received 15 x 106 DC. Pulsed dendritic<br />

cellswere injectedsubcutaneously into the supra-scapular region. Imiquimod<br />

cream was applied at the injection site just prior to vaccination and<br />

24 hours later. The vaccine schedule dictated administration every 2 weeks<br />

for 8 weeks (total <strong>of</strong> 5 doses) then monthly to progression or a total <strong>of</strong> 52<br />

weeks. Prior to each vaccination, patients met eligibility parameters and<br />

had no progression on MRI. Patients were imaged monthly and blood was<br />

drawn to evaluate toxicity and immune response. Results: No vaccinerelated<br />

toxicity has been reported. Time to progression ranges from 6.5<br />

weeks for the patient treated at the first dose level to 35 weeks for one<br />

patient receiving 15 x 106 DC. The latter patient experienced a partial<br />

response at 20 weeks. Two patients have stable disease at 18.5 and 28<br />

weeks, respectively. One patient was not evaluable. Flow cytometric<br />

analysis demonstrated expansion <strong>of</strong> central memory T cells amidst declining<br />

effector memory cells following vaccination in three patients at a dose<br />

<strong>of</strong> 15x106 DC. Conclusions: Apoptotic body-pulsed DC vaccination was well<br />

tolerated and preliminarily demonstrated clinical activity but a minimum <strong>of</strong><br />

15x106 DC was required for a modulation <strong>of</strong> central memory T cells.<br />

2548 General Poster Session (Board #2D), Mon, 8:00 AM-12:00 PM<br />

Difference between duration <strong>of</strong> treatment (DOT) and progression-free<br />

survival (PFS) as a marker <strong>of</strong> unbalanced censoring. Presenting Author:<br />

Laleh Amiri-Kordestani, National Cancer Institute, National Institutes <strong>of</strong><br />

Health, Bethesda, MD<br />

Background: In the conduct <strong>of</strong> randomized trials Kaplan and Meier<br />

envisioned rates <strong>of</strong> censoring as similar between arms, providing accurate<br />

assessment <strong>of</strong> clinical trial results. Censoring is used when patients<br />

withdraw consent, leave study due to toxicity, or reach data cut-<strong>of</strong>f without<br />

disease progression or death. Censoring can lead to erroneous conclusions<br />

as it can be either beneficial or detrimental to the arm under study. Such<br />

censoring can also explain how a statistically valid difference in PFS<br />

“disappears” when overall survival (OS) is examined. We hypothesized that<br />

censoring, especially that due to toxicity, would lead to a discrepancy<br />

between DOT and PFS since two different patient populations would be<br />

scored. Methods: We reviewed all phase III randomized studies <strong>of</strong> drugs<br />

approved by FDA since 2005 for pts with metastatic solid tumors, looking<br />

for DOT and PFS. We used standard statistical analyses using SAS. Results:<br />

We identified 55 Phase III studies conducted with abiraterone, axitinib,<br />

bevacizumab, cabazitaxel, cetuximab, eribulin, erlotinib, everolimus, ipilimumab,<br />

ixabepilone, lapatinib, panitumumab, pazopanib, sorafenib,<br />

sunitinib, temsirolimus and vandetinib. DOT was not provided in 27%.<br />

Forty-four comparisons (88 arms) were included in the analysis. The<br />

median PFS, DOT, delta PFS (difference in PFS between experimental and<br />

control arms) and delta DOT were: 161, 126, 51 and 36 days, respectively.<br />

The slopes <strong>of</strong> PFS vs DOT and delta PFS vs delta DOT were 1.16 and 1.03,<br />

respectively close to the ideal <strong>of</strong> 1.0. Five trials fell above the 90% CI<br />

boundary with delta PFS/delta DOT <strong>of</strong> 3 to 36, including two everolimus<br />

studies (PNET and breast cancer) two sunitinb studies (RCC and PNET)<br />

and one bevacizumab study (E2100, breast cancer). Conclusions: PFS and<br />

DOT as well as delta PFS and delta DOT should be concordant. The most<br />

likely explanation for a discordance between these values is toxicity-driven<br />

censoring and its occurrence raises concerns regarding the degree <strong>of</strong><br />

efficacy. A greater utilization <strong>of</strong> “Time to Treatment Failure”, an endpoint<br />

that includes toxicity in its definition would be valuable in oncology trials,<br />

particularly those with high levels <strong>of</strong> toxicities.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


154s Developmental Therapeutics—<strong>Clinical</strong> Pharmacology and Immunotherapy<br />

2549 General Poster Session (Board #2E), Mon, 8:00 AM-12:00 PM<br />

Recommendation <strong>of</strong> dose banding <strong>of</strong> cytotoxics according to pharmacokinetic<br />

criteria. Presenting Author: Etienne Chatelut, Institut Claudius<br />

Regaud, Toulouse, France<br />

Background: Dose-banding has been recently suggested in order to optimize<br />

chemotherapy preparation. Ranges (or bands) <strong>of</strong> body surface area (BSA)<br />

are predefined. The individual dose <strong>of</strong> a particular patient is calculated<br />

according to a single BSA value per band, usually the mid-point <strong>of</strong> the BSA<br />

band in which the actual BSA <strong>of</strong> the patient lies. Thanks to this simple<br />

procedure, chemotherapy provision can be rationalized and chemotherapies<br />

can be prepared in advance for drugs with sufficient long-term drug<br />

stability. The primary purpose <strong>of</strong> dose-banding is to reduce patient waiting<br />

time and improve capacity planning <strong>of</strong> the pharmacy production, but<br />

additional benefits can also be found, such as reduced potential for<br />

medication errors, reduced drug wastage, and prospective quality control <strong>of</strong><br />

preparations. The objective <strong>of</strong> this analysis was to compare dose-banding to<br />

individual BSA-dosing (current practice) according to pharmacokinetic<br />

(PK) criteria. Methods: Dose-banding was defined according to three bands<br />

<strong>of</strong> BSA: BSA�1.7m², 1.7m²�BSA�1.9m², and BSA�1.9m² for which the<br />

values <strong>of</strong> 1.55m², 1.80m², and 2.05m² were allocated, respectively. By<br />

using individual actual values <strong>of</strong> clearance <strong>of</strong> six drugs (cisplatin, docetaxel,<br />

paclitaxel, doxorubicin, topotecan, and irinotecan) from a total <strong>of</strong><br />

1,206 adult cancer patients, the AUCs corresponding to the two dosing<br />

methods were compared to a target value <strong>of</strong> AUC for each drug. Results:<br />

Over all 6 drugs, by using dose-banding the percent change <strong>of</strong> individual<br />

dose in comparison with BSA dosing ranged between -14% and �22%. In<br />

terms <strong>of</strong> capacity to attain the target AUC, there was no significant<br />

difference in precision when using dose-banding as compared to BSAdosing<br />

for all drugs except paclitaxel (precision <strong>of</strong> 23.2% versus 21.8%,<br />

respectively). For all drugs including paclitaxel, distributions <strong>of</strong> AUC values<br />

were very similar with both dosing methods. Conclusions: For these 6 drugs<br />

and maybe others, dose-banding may be implemented without any risk <strong>of</strong><br />

increasing interindividual plasma exposure. Dose-banding would make it<br />

possible to anticipate chemotherapy preparation and analytical control<br />

without any delay for the patients.<br />

2551 General Poster Session (Board #2G), Mon, 8:00 AM-12:00 PM<br />

Thymidylate synthase genotype specific dosing <strong>of</strong> capecitabine: Pro<strong>of</strong> <strong>of</strong><br />

concept phase I study. Presenting Author: Ross A. Soo, National University<br />

Cancer Institute, Singapore<br />

Background: Thymidylate synthase (TYMS) is the target <strong>of</strong> fluoropyrimidines<br />

(FP). TYMS 3R3R is the predominant genotype in East Asian (EA) patients<br />

and is associated with increased TYMS expression, reduced FP related<br />

toxicity and relative FP resistance. Dose finding studies for FP were<br />

developed in Caucasians, a population that typically harbors TYMS 2R2R<br />

and 2R3R genotypes. We hypothesize the recommended phase II dose<br />

(RP2D) <strong>of</strong> capecitabine is higher in 3R3R and similar for 2R allele carriers<br />

compared to the FDA approved dose. Objectives 1) To determine the<br />

maximal tolerated dose (MTD) and RP2D <strong>of</strong> capecitabine in EA patients<br />

with advanced stage malignancy 2) To determine the safety and toxicity <strong>of</strong><br />

this regimen and 3) To perform plasma pharmacokinetics (PK) <strong>of</strong> capecitabine<br />

and its metabolites. Methods: EA patients with advanced stage<br />

cancer were prospectively allocated into two cohorts: Group A (3R3R) and<br />

Group B (2R3R, 2R2R). In each cohort, dose escalation followed a<br />

standard phase I 3�3 design. Additional patients were treated at the R2P2<br />

dose level (DL). Initial dose was 1250/m2 bd for 14 days q3w (DL 1) with<br />

125 mg/m2 bd increments subsequently. Pharmacokinetics (PK) <strong>of</strong> capecitabine<br />

and its metabolites were performed using a LC-MS/MS method.<br />

Results: 23 patients (Group A�18; group B�5) received 94 cycles (median<br />

2.5, range 1-8). Median age was 58 (range 34-74) years. Median<br />

turnaround time for TYMS genotyping was 1 day. In group A, grade 3-4<br />

DLTs were seen in 3 patients: diarhoea, neutropenic fever, hand-foot<br />

syndrome, and mucositis. MTD was at DL 4 (1625mg/m2 bd) and the RP2D<br />

was 1500mg/m2 bd (DL 3). DL 3 was expanded to n�9. At DL 3, day 1<br />

mean (� SD) capecitabine and 5FU Cmax was 12.3 � 9.9 and 0.91 � 0.73<br />

�g/mL, respectively and AUC0 – twas 9.84 � 5.53 and 1.21 � 0.54<br />

h*�g/mL, respectively. Compared with DL1, Cmax for capecitabine and 5FU<br />

was 2 and 2.02 fold higher and AUC 0-t was 1.49 and 1.59 fold higher at<br />

DL3. Accrual in group B was ceased at DL2 due to lack <strong>of</strong> patient<br />

enrolment; no DLT was seen. By ROC analysis, day 1 5FU AUC0–t<strong>of</strong> 1.74<br />

h*�g/mL predicted for DLT (p�0.022, sensitivity 100%, specificity 90%).<br />

Conclusions: In EA patients with TSER 3R3R, the RP2D was 1500 mg/m2 bd. The D1 5FU AUC0–tat RP2D was 59% more than the FDA approved<br />

dose (DL1).<br />

2550 General Poster Session (Board #2F), Mon, 8:00 AM-12:00 PM<br />

Phase I study <strong>of</strong> weekly oral docetaxel (ModraDoc001) plus ritonavir in<br />

patients with advanced solid tumors. Presenting Author: Serena Marchetti,<br />

Netherlands Cancer Institute, Amsterdam, Netherlands<br />

Background: ModraDoc001 is a novel oral formulation containing docetaxel<br />

as a solid dispersion. Oral administration <strong>of</strong> docetaxel is feasible in<br />

combination with the CYP3A4 inhibitor ritonavir. Objectives were to<br />

determine the safety, maximum tolerated dose (MTD) and pharmacokinetics<br />

(PK) <strong>of</strong> weekly oral docetaxel (as ModraDoc001) in combination with<br />

ritonavir. Methods: Patients with advanced solid tumors, WHO PS � 2, no<br />

concomitant use <strong>of</strong> MDR or CYP3A modulating drugs, adequate bone<br />

marrow (ANC � 1.5x109 /L), liver (bilirubin � 1.5xULN, ALAT/ASAT �<br />

2.5xULN) and renal function (creatinine � 1.5xULN or clearance � 50<br />

ml/min) were eligible. Docetaxel (ModraDoc001 10mg capsule) and<br />

ritonavir (Norvir 100mg) were simultaneously given once weekly in a ‘3�3<br />

cohort’ dose escalation design. MTD was defined as the highest dose<br />

resulting in �1/6 probability <strong>of</strong> causing a dose limiting toxicity in the first 4<br />

weeks <strong>of</strong> treatment. This cohort was expanded with 6 patients. PK was<br />

determined on days 1 and 8. Results: 40 patients (25 male, 35 evaluable<br />

for safety) were enrolled in 6 dose levels (30/100, 40/100, 60/100,<br />

80/100, 60/200 and 80/200 mg docetaxel/ritonavir). Common treatment<br />

related adverse events in the 4 highest dose levels were diarrhea (68%),<br />

nausea (62%) and fatigue (62%), mostly CTC grade 1-2 (80%, 95% and<br />

73% respectively). Five patients experienced a DLT (grade 3 diarrhea (4),<br />

elevated ASAT/ALAT (1), grade 4 dehydration and grade 3 mucositis (1),<br />

grade 3 fatigue (2)). The MTD was 60 mg/200 mg docetaxel/ritonavir.<br />

<strong>Part</strong>ial remission was seen in 4 patients (CUP, NSCLC, gastric and mamma<br />

ca) and sustained stable disease in 15 patients (6x NSCLC). Both drugs<br />

were rapidly absorbed after oral administration. Mean Tmax was 2.0 hours<br />

(CV�73%) for docetaxel. Cmax and AUC <strong>of</strong> docetaxel increased less than<br />

proportionally with dose to 162 ng/ml (CV� 67%) and 1615 ng/ml*hr<br />

(CV� 81%), respectively, in 15 patients at the MTD. Conclusions: At the<br />

MTD (once weekly 60 mg docetaxel and 200 mg ritonavir) ModraDoc001 is<br />

safe, well tolerated and shows encouraging antitumor activity. The exposure<br />

<strong>of</strong> docetaxel with this regimen is comparable to weekly intravenous<br />

administration <strong>of</strong> 35 mg/m2 docetaxel. Phase II studies in solid tumors are<br />

planned.<br />

2552 General Poster Session (Board #3A), Mon, 8:00 AM-12:00 PM<br />

A phase Ib dose-escalation study <strong>of</strong> eribulin mesylate in combination with<br />

capecitabine in patients with advanced/metastatic cancer. Presenting<br />

Author: Muhammad Yaser Nasim, Medical Oncology, The Beatson West <strong>of</strong><br />

Scotland Cancer Centre, Glasgow, United Kingdom<br />

Background: Eribulin mesylate is a microtubule dynamics inhibitor approved by<br />

FDA for patients (pts) with metastatic breast cancer after treatment with at<br />

least two prior chemotherapeutic regimens. This Phase Ib, open-label doseescalation<br />

study determined the maximum tolerated dose (MTD) <strong>of</strong> eribulin in<br />

combination with capecitabine. Methods: Pts with advanced solid malignancies<br />

refractory to standard therapies, adequate organ function and ECOG<br />

performance status �2 received eribulin mesylate (2–5-min IV) by Schedule 1<br />

(1.2, 1.6 or 2.0 mg/m2 on Day 1) or Schedule 2 (0.7, 1.1 or 1.4 mg/m2 on<br />

Days 1 and 8), in combination with twice-daily oral capecitabine 1000 mg/m2 Days 1-14 every 21 days. The MTD was defined as the highest dose in each<br />

schedule where �1/6 pts experienced dose-limiting toxicity (DLT). Safety and<br />

pharmacokinetics (PK) were assessed. Results: Of the 34 pts recruited, 19<br />

(53% male; median age 62 years; 42% ECOG 0, 58% ECOG 1) and 15 (33%<br />

male; median age 61 years; 33% ECOG 0, 60% ECOG 1, 7% ECOG 2) were<br />

enrolled in Schedules 1 and 2, respectively. Most common tumor types were<br />

large intestine (20.6%), lung/bronchus (17.7%) and breast (14.7%). DLTs are<br />

shown in the table; there were no unexpected toxicities with the combination.<br />

The MTD for eribulin mesylate was 1.6 and 1.4 mg/m2 for Schedules 1 and 2,<br />

respectively, in combination with capecitabine 1000 mg/m2 twice-daily.<br />

Eribulin PK were dose proportional and independent <strong>of</strong> schedule or capecitabine<br />

co-administration. Combination with eribulin had no effect on the<br />

disposition <strong>of</strong> capecitabine and its metabolites. Although sample size was<br />

small, preliminary signs <strong>of</strong> efficacy were observed. Conclusions: The combination<br />

<strong>of</strong> eribulin and capecitabine was well tolerated with no unexpected safety<br />

findings. Schedule 2 MTD (1.4 mg/m2 Days 1 and 8) delivered a higher dose<br />

intensity <strong>of</strong> eribulin than Schedule 1 and was selected for evaluation in an<br />

ongoing Phase II breast cancer study.<br />

Summary <strong>of</strong> DLTs.<br />

Schedule<br />

Dose<br />

mg/m2 DLT<br />

n (%) DLT DLT grade<br />

1(N�19) 1.2 1 (5.3) Febrile neutropenia 3<br />

1.6 1 (5.3) Neutropenia 4<br />

2.0 2 (10.5) 1x Fatigue 3<br />

1x Lethargy 3<br />

2(N�15) 0.7 0 (0) NA NA<br />

1.1 1 (6.7) Neutropenic sepsis 4<br />

1.4 1 (6.7) Neutropenia 3<br />

NA, not applicable<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


Developmental Therapeutics—<strong>Clinical</strong> Pharmacology and Immunotherapy<br />

2553 General Poster Session (Board #3B), Mon, 8:00 AM-12:00 PM<br />

Phase I pharmacokinetic (PK) and pharmacodynamic (PD) study <strong>of</strong><br />

intravenous dimethane sulfonate (DMS612, NSC 281612) in advanced<br />

malignancies. Presenting Author: Susan Elaine Bates, Center for Cancer<br />

Research, National Cancer Institute, Bethesda, MD<br />

Background: DMS612 is a dimethane sulfonate compound that was<br />

identified as preferentially cytotoxic to renal cell carcinoma (RCC) cell lines<br />

in a chemical screen <strong>of</strong> the NCI-60 panel. DMS612 has bifunctional<br />

alkylating activity in vitro. Objectives <strong>of</strong> this first-in-human phase I study<br />

included determining the dose-limiting toxicity (DLT), maximum tolerated<br />

dose (MTD), recommended phase 2 dose (RP2D), PK and PD <strong>of</strong> DMS612<br />

administered by 10 minute intravenous infusion on day 1, 8 and 15 <strong>of</strong> a 28<br />

day cycle. Methods: Eligibility criteria included adults with advanced solid<br />

malignancies or lymphoma with ECOG performance status 0-2, life<br />

expectancy � 3 months and adequate organ and marrow function. Patients<br />

were enrolled using a standard “3�3” dose escalation scheme. Plasma PK<br />

<strong>of</strong> DMS612 and metabolites was assessed by LC-MS/MS. DNA damage PD<br />

was assessed by �-H2AX immun<strong>of</strong>luorescence. Results: 35 subjects were<br />

enrolled (22 male, 13 female) with median age 59 years (41-75). Tumor<br />

types included colorectal (8), RCC (4), cervix (2), and urothelial (2). Doses<br />

administered were 1.5, 3, 5, 7, 9 and 12 mg/m2 . The MTD was determined<br />

to be 9 mg/m2 , with only one DLT <strong>of</strong> grade 4 thrombocytopenia in 12<br />

subjects enrolled. The maximum administered dose <strong>of</strong> 12 mg/m2 was<br />

considered to be intolerable after 1 <strong>of</strong> 3 subjects had grade 4 neutropenia<br />

and 1 had prolonged grade 3 thrombocytopenia. Prolonged thrombocytopenia<br />

in later cycles was observed in other subjects, including one patient<br />

naïve to prior cytotoxic chemotherapy. One subject with RCC had a<br />

confirmed partial response at 7 mg/m2 . DMS612 was rapidly converted into<br />

carboxy, chloroethyl and hydroxyethyl analogues and their glucuronides,<br />

some <strong>of</strong> which retained alkylating activity in vitro. Dose-dependent pharmacodynamic<br />

evidence <strong>of</strong> DNA damage induced by DMS612 in vivo was<br />

observed by �-H2AX immun<strong>of</strong>luorescance in both peripheral blood lymphocytes<br />

and plucked scalp hairs. Conclusions: The MTD <strong>of</strong> DMS12 administered<br />

by intravenous infusion on day 1, 8 and 15 <strong>of</strong> a 28-day cycle was 9<br />

mg/m2 . Pre-clinical and clinical observations suggest that further study <strong>of</strong><br />

DMS612 in RCC is warranted.<br />

2555 General Poster Session (Board #3D), Mon, 8:00 AM-12:00 PM<br />

Phase I development <strong>of</strong> a weekly dosing schedule for the oral taxane<br />

tesetaxel. Presenting Author: Amy Lang, South Texas Accelerated Research<br />

Therapeutics, LLC, San Antonio, TX<br />

Background: Tesetaxel (TST) has anticancer activity in patients (pts) with<br />

metastatic breast cancer (MBC) and gastric cancer when administered<br />

orally once every 3 weeks (Q3W). This drug is not a Pgp substrate, does not<br />

cause hypersensitivity reactions, and may be associated with less neurotoxicity.<br />

The maximally tolerated dose (MTD) on the Q3W schedule is 27<br />

mg/m2 , and neutropenia is dose-limiting. Since taxane activity may be<br />

schedule-dependent, we conducted a weekly, dose-ranging, and PK evaluation<br />

<strong>of</strong> TST. Methods: Eligibility: advanced solid tumors; ECOG PS 0-2;<br />

adequate organ function. TST was given once weekly for 3 weeks in a<br />

28-day cycle, beginning at a total flat dose <strong>of</strong> 25 mg/cycle with progressive<br />

increases in total dose up to 75 mg/cycle. Subsequently, dosing was<br />

converted to a weight-based regimen at weekly doses <strong>of</strong> 12.5, 15, and 17.5<br />

mg/m2 (i.e., total per cycle dose up to 52.5 mg/m2 ). Serial plasma samples<br />

were assayed by HPLC. Results: 26 pts were treated in 8 dose cohorts. The<br />

MTD was 15 mg/m2 /wk (total cycle dose, 45 mg/m2 ). With repeated cycles,<br />

constitutional symptoms (fatigue and anorexia) rather than neutropenia<br />

proved dose-limiting at 17.5 mg/m2 /wk. Only 1 pt (at 12.5 mg/m2 /wk)<br />

developed grade 3 neutropenia; no other episodes <strong>of</strong> Grade 3-4 myelotoxicity<br />

were observed. One pt with MBC (who had progressed after 2 prior<br />

taxane regimens) achieved a PR; 1 pt with prostate cancer has maintained<br />

prolonged reduction <strong>of</strong> PSA (� 57%). PK analyses showed low but<br />

progressive increases in trough TST concentrations (0.4–4.6 nmol/mL) 7<br />

days after each succeeding dose, which was consistent with the prolonged<br />

T½ <strong>of</strong> this drug (~180 hrs). There was no substantial drug accumulation<br />

over multiple cycles. Conclusions: Weekly oral TST is safe, provides<br />

long-term low-level drug exposure, and increases the total delivered dose<br />

over 12 wks by 25%. Interestingly, this regimen is not associated with<br />

significant myelosuppression. Thus, weekly TST dosing provides a highly<br />

convenient, “dose-dense”, taxane regimen that is not associated with<br />

neutropenia, which should facilitate its integration into multiple chemotherapy<br />

regimens. An ongoing study is evaluating efficacy <strong>of</strong> weekly TST<br />

dosing in MBC.<br />

155s<br />

2554 General Poster Session (Board #3C), Mon, 8:00 AM-12:00 PM<br />

Phase I and pharmacokinetic (PK)/pharmacodynamic (PD) study <strong>of</strong><br />

LY2334737, an oral gemcitabine prodrug, in patients (pts) with advanced<br />

solid tumors. Presenting Author: Sandrine J. Faivre, Department <strong>of</strong> Medical<br />

Oncology, Beaujon University Hospital, Clichy, France<br />

Background: LY2334737 is a valproic acid-bound carboxylesterasesubstrate<br />

prodrug <strong>of</strong> gemcitabine that can be given orally. The primary<br />

objective was to define the recommended phase II doses and schedules<br />

(sch) <strong>of</strong> administration <strong>of</strong> LY2334737 in 28-day cycles. Methods: Eligible<br />

pts (ECOG�2) had adequate hematologic, renal, and hepatic functions. In<br />

sch-A, LY2334737 was given every other day for 21 days. In sch-B,<br />

LY2334737 was given daily every other week. Dose escalation (3�3) was<br />

based on CTCAE V3 toxicities. Secondary objectives were PKs (LY2334737,<br />

dFdC, dFdU), PD biomarkers (dFdC DNA incorporation, Cytokeratin 18 -<br />

M30), and antitumor activity. Results: 57 pts (38 males, 19 females,<br />

median age 60 yrs) were treated using 7 dose-levels (40–100 mg/day). Pts<br />

received a median <strong>of</strong> 2 cycles. Dose limiting toxicities (DLTs) at cycle 1<br />

(any dose level) consisted <strong>of</strong> diarrhea (2pt), AST elevation (1pt), lower<br />

extremity edema (1pt), QTC prolongation (1pt), and general deterioration<br />

(1pt). A total <strong>of</strong> 2/7pts treated with 100mg in sch-A and 1/4pts treated with<br />

90mg in sch-B had DLTs. During dose escalation most frequent toxicities<br />

were 1) Sch-A (28 pts): nausea (10pts), pyrexia (9pts), vomiting (8pts),<br />

mucositis (6pts), anorexia (6pts), ALT elevation (5pts), asthenia (5pts),<br />

anemia (5pts), fatigue (4pts), chills (4pts), and lymphopenia (4pts). 2)<br />

Sch-B (29 pts): pyrexia (13pts), nausea (8pts), diarrhea (6pts), vomiting<br />

(5pts), chills (6pts), anemia (5pts), asthenia (6pts), AST elevation (6pts),<br />

ALT elevation (5pts), and fatigue (4pts). Nine pts had stable disease (3pts<br />

completed �6 cycles). PKs showed a dose-proportional increase in<br />

exposure <strong>of</strong> LY2334737 and dFdC with no accumulation after repeated<br />

dosing. dFdC is detected in the DNA <strong>of</strong> peripheral blood mononuclear cells<br />

(increasing amount with dose and over time after repeated cycle). In sch-A,<br />

we observed a significant increase in cytokeratin 18 M30 relative to<br />

baseline. Conclusions: LY2334737 displayed linear PK and acceptable<br />

safety pr<strong>of</strong>ile in both schedules. The dose <strong>of</strong> 90mg given every other day is<br />

the recommended dose for phase II trials with LY2334737 (dose expansion<br />

is ongoing).<br />

2556 General Poster Session (Board #3E), Mon, 8:00 AM-12:00 PM<br />

Effect <strong>of</strong> dexrazoxane on doxorubicin-induced testicular toxicity. Presenting<br />

Author: Irit Ben-Aharon, David<strong>of</strong>f Center, Rabin Medical Center, Petach<br />

Tikva, Israel<br />

Background: Seminal advances in anti-cancer therapy result in growing<br />

numbers <strong>of</strong> young male cancer survivors for whom treatment-induced<br />

infertility represents a major late-term concern. Doxorubicin (DXR) has<br />

been previously shown to exert toxic effect on the testicular germinal<br />

epithelium. Based upon the cardioprotective traits <strong>of</strong> dexrazoxane (DEX),<br />

we aimed to study its potential effect to reduce DXR-induced testicular<br />

toxicity. Methods: Male mice were injected intraperitoneally with 5mg/kg<br />

DXR or 100mg/kg DEX or the combination <strong>of</strong> both and scarified at one<br />

month post treatment. Saline-injected mice served as controls. Testes were<br />

excised, weighed and further processed. For oxidative stress determination<br />

glutathione assay was performed on testes’ lysates and P38 protein levels<br />

were determined by western blot analysis. Bax levels were used to assess<br />

apoptosis. Immunohistochemistry and confocal microscopy were used to<br />

study the effect <strong>of</strong> DXR, DEX and their combination, on the testis histology<br />

as well as on the spermatogonial reserve. Results: One month after DEX and<br />

DXR treatment, a striking decline in testicular weight was observed<br />

(decrease by 60% compared with control values; decrease <strong>of</strong> 54% in<br />

DXR-only treated mice; p�0.05). DEX prevented DXR-induced oxidative<br />

stress. However, DEX enhanced DXR-induced apoptosis within the testes<br />

and furthermore, the combination depleted the spermatogonial reserve one<br />

month after treatment. Conclusions: DEX activity in the testis may differ<br />

from its activity in cardiomyocytes. Adding DEX to DXR may exacerbate<br />

DXR-induced testicular toxicity.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


156s Developmental Therapeutics—<strong>Clinical</strong> Pharmacology and Immunotherapy<br />

2557 General Poster Session (Board #3F), Mon, 8:00 AM-12:00 PM<br />

First-in-human phase I and pharmacokinetic study <strong>of</strong> DTS-108 in patients<br />

with advanced carcinomas. Presenting Author: Romain Coriat, Cochin<br />

Teaching Hospital, AP-HP, Paris Descartes University, Paris, France<br />

Background: DTS-108 is a soluble pro-drug <strong>of</strong> SN38, the active metabolite<br />

<strong>of</strong> irinotecan, where the SN38 moiety is covalently linked to a 20AA<br />

peptide through a specific cross-linker that releases SN38 by esterase bond<br />

cleavage. DTS-108 was designed to achieve therapeutic levels <strong>of</strong> SN38,<br />

while reducing diarrhea. Methods: This trial identified the pharmacokinetics,<br />

the adverse event (AE) pr<strong>of</strong>ile (NCI-CTCv3 criteria), the dose limiting<br />

toxicities (DLT) at cycle 1, the maximum tolerated dose (MTD), and the<br />

recommended phase II dose (RD) <strong>of</strong> intravenous DTS-108 (1-2 hours)<br />

every two weeks (q2w) in patients (pts) with advanced/metastatic carcinomas,<br />

ECOG:0-1, and adequate hepatic, marrow, and kidney functions.<br />

Results: Forty-Two pts received DTS-108 across 14 dosing cohorts (range:<br />

3-416 mg/m2). All grade AEs were asthenia (43%), nausea (43%),<br />

diarrhea (41%), vomiting (33%), anemia (31%), rash (21%), anorexia<br />

(21%), alopecia (19%), abdominal pain (19%), and neutropenia (19%).<br />

Grade 3 diarrhea was only reported in two pts with either partial bowel<br />

obstruction or recent history <strong>of</strong> a total colectomy. Neutropenia was the dose<br />

limiting toxicity <strong>of</strong> DTS-108. Neutropenia started to be observed at doses<br />

�56 mg/m2. At 416 mg/m2, 3/6 pts had grade 4 neutropenia. Infusion<br />

related reactions (itching urticaria) were observed with increasing frequency<br />

and severity at doses �75 mg/m2 and required antihistamine<br />

premedications. At 313 mg/m2 and 416 mg/m2, infusion skin reactions<br />

were 2/6 pts and 6/6 pts, respectively. Fluorescence HPLC was initially<br />

used to quantify DTS-108 and its metabolites (SN-38 and SN-38G).<br />

Sample stability analysis <strong>of</strong> this method showed that SN38 values were<br />

inaccurate as DTS-108 degraded forming ex-vivo SN-38 during the blood<br />

collection, plasma storage, and/or sample extraction. New processes and<br />

analytical LC/MS/MS methods for testing SN-38 were implemented. At the<br />

dose <strong>of</strong> 313 mg/m2, mean DTS-108, SN38, and SN38G AUCINF (CV%)<br />

were 439293 (24%), 1992 (34%), and 4538 (46%) h*ng/mL. Tumor<br />

stabilization (RECIST) was observed in 9 pts and confirmed in 6 pts.<br />

Conclusions: DTS induced neutropenia and infusion skin reactions but no<br />

dose-limiting diarrhea. The MTD was 416 mg/m2 and the RD was 313<br />

mg/m2 q2w.<br />

2559 General Poster Session (Board #3H), Mon, 8:00 AM-12:00 PM<br />

Phase II trial <strong>of</strong> a GM-CSF-producing and CD40L-expressing cell line combined<br />

with allogeneic tumor antigen as a novel vaccine for metastatic lung<br />

adenocarcinoma. Presenting Author: Ben C. Creelan, H. Lee M<strong>of</strong>fitt Cancer<br />

Center & Research Institute, Tampa, FL<br />

Background: We created a vaccine in which irradiated allogeneic lung<br />

adenocarcinoma cells are combined with a bystander K562 cell line<br />

transfected with hCD40L and hGM-CSF. By recruiting and activating<br />

dendritic cells, we hypothesized the vaccine would induce tumor regression<br />

in metastatic lung adenocarcinoma. Methods: Intradermal vaccine was<br />

given every 14 days x3, followed by monthly x3. Cyclophosphamide (300<br />

mg/m2 IV) was administered before 1st and 4th vaccines to deplete<br />

regulatory T-cells. All-trans retinoic acid was given (150/mg/m2/day) after<br />

1st and 4th vaccines to enhance dendritic differentiation. Peripheral blood<br />

mononuclear cells (PBMCs) were collected at baseline and after each<br />

vaccination. T-cell activation pr<strong>of</strong>iles were analyzed by ELISpot assay and<br />

tested by generalized Wilcoxon for correlation to survival. Results: 24<br />

participants were accrued at a single center from 10/2006 to 6/2008, with<br />

median age 64 and median <strong>of</strong> 3 previous lines <strong>of</strong> chemotherapy prior to<br />

entry. 20 were former smokers and 4 had brain metastases. A total <strong>of</strong> 101<br />

vaccines were administered. Common toxicities <strong>of</strong> any grade were joint pain<br />

(79%) and fatigue (75%). Significant adverse events included a grade 3<br />

hypotension and a grade 3 acute respiratory distress. No confirmed<br />

complete or partial radiologic responses were observed. Median overall<br />

survival (OS) was 8.0 mo (95% CI 3.5 – 12.5) and median time-toprogression<br />

was 2.4 mo (95% CI 0.3 – 4.6). Presence <strong>of</strong> HLA-A2 conferred<br />

reduced risk <strong>of</strong> progression (HR 0.37, 95% CI 0.14 -0.89, p�0.02) and<br />

trend to improved OS (HR 0.59, p � 0.06). Of 14 participants with<br />

evaluable PBMCs, 5 demonstrated sustained tumor peptide-specific T-cell<br />

activation after vaccination. Ex vivo peptide immune response correlated<br />

with improved OS compared to non-responders (23 vs. 7 mo, HR 0.48, p �<br />

0.04). Conclusions: Vaccine administration was feasible and tolerable in a<br />

heavily pretreated population <strong>of</strong> metastatic lung cancer. These data<br />

suggest the vaccine has clinical activity in the subset with peptide-induced<br />

T-cell immune responses and warrants further investigation. A randomized<br />

trial <strong>of</strong> the vaccine is currently in development.<br />

2558 General Poster Session (Board #3G), Mon, 8:00 AM-12:00 PM<br />

Human papillomavirus type 16 (HPV16) E6/E7-specific cytotoxic T lymphocytes<br />

(CTL) for immunotherapy <strong>of</strong> HPV-associated cancer (Ca). Presenting<br />

Author: Carlos Almeida Ramos, Baylor College <strong>of</strong> Medicine, Houston, TX<br />

Background: Vaccines prevent HPV-associated Ca, but their benefits in<br />

established Ca are disappointing: although these tumors express viral E6<br />

and E7 antigens (Ags) immune responses are limited even after vaccination,<br />

likely due to negative environmental cues that block tumor recognition<br />

and T cell (TC) activation in vivo. We postulated that ex vivo stimulation <strong>of</strong><br />

TCs in an immunologically favorable milieu would allow us to reactivate<br />

tumor-directed CTLs from Ca patients and benefit the recipients on<br />

reinfusion. Methods: We studied 68 patients with HPV� Ca. To detect<br />

HPV16 E6/E7-specific TCs (HPV-TCs) in blood, we measured the IFN�<br />

ELISpot responses <strong>of</strong> TCs stimulated by monocyte-derived dendritic cells<br />

(DCs) loaded with pepmixes (peptide libraries <strong>of</strong> 15-mers overlapping by<br />

11 aa) spanning E6/E7. Because HPV-TCs from these patients may be<br />

anergized by their tumors, potent Ag presenting strategies might be<br />

required for reactivation, and thus we stimulated these cells in the<br />

presence <strong>of</strong> IL-6, -7, -12 and -15, as we have shown that these can induce<br />

responses to poorly immunogenic Ags. Results: We successfully reactivated<br />

HPV-TCs from 8/16 cervical and 33/52 oropharyngeal Ca patients. We<br />

could expand these HPV-TCs to clinically useful numbers by using patient<br />

B-cell blasts (BBs) as APCs. Stimulation <strong>of</strong> DC-stimulated HPV-TCs by<br />

E6/E7 pepmix-loaded BBs further expanded (3.8 � 1.5�/round) HPV-TC<br />

lines, which phenotypically are almost exclusively composed <strong>of</strong> TCs (98 �<br />

3% CD3�), with a variable proportion <strong>of</strong> mostly effector memory (CD45RA–,<br />

CD45RO�, CD62L– and CCR7–) CD4� and CD8� cells (37 � 28% and<br />

49 � 27%, respectively). The viral/tumor associated epitopes recognized<br />

mapped to E6 aa 49-71, 77-91 and 125-143, and E7 aa 1-19 and 73-87.<br />

The expanded cells from patients killed E6/E7� targets (specific lysis up to<br />

45-61% vs. 0-8% in controls, 40:1 E:T ratio). Conclusions: We have<br />

developed a system that allows the robust generation <strong>of</strong> HPV-directed CTLs<br />

from patients with HPV16� Ca, which recognize specific epitopes in<br />

tumor-associated Ags. Our lines have the potential to be used for adoptive<br />

cellular immunotherapy <strong>of</strong> HPV� Ca.<br />

2560 General Poster Session (Board #4A), Mon, 8:00 AM-12:00 PM<br />

Where to look for sentinel node in breast cancer? Presenting Author: Anton<br />

J. Scharl, Klinikum St Marien, Amberg, Germany<br />

Background: It has been observed, that the caudal Axilla on the border to<br />

pectoralis muscle is predicive for the sentinel node. The sono-morphology<br />

<strong>of</strong> lymph nodes has been the subject <strong>of</strong> multiple publications, usually<br />

dealing with malignant melanoma. In the context <strong>of</strong> sentinel lymph node<br />

biopsy (SLNB) in breast cancer patients, the following study examines the<br />

feasibility <strong>of</strong> the sonographic differentiation <strong>of</strong> the Sentinel lymph node<br />

(SLN) from neighboring non-SLNs and whether sentinel-ultrasound-needle<br />

localization (SUN) is a useful addition or alternative to current methods <strong>of</strong><br />

“lymphatic mapping”. Methods: During a prospective study performed from<br />

1/2003 to 9/2005 including 404 breast cancer patients (Tis-T4), the<br />

SLNB was performed using patent blue�/- 99Tc-Nanocoll. In addition to<br />

and independent <strong>of</strong> this method, the axilla was sonographically examined<br />

for “reactive” lymph nodes n�180 pt. (Siemens Elegra 7.5 MHz). The<br />

“reactivity” <strong>of</strong> the nodes was quantified using an index , which allowed the<br />

comparison <strong>of</strong> adjacent nodes. The most “reactive” lymph node in the<br />

caudal axilla was identified as the “Ultrasound-Sentinel-Node”(US-SLN)<br />

and has been marked with a wire. Results: In 180 patients the SLN was<br />

localized using the standard methods as well as (SUN). The was no<br />

difference in detection rates <strong>of</strong> US-SLN and the standard methods in<br />

tumor-free nodes(SLN-). However, for patients with axillary metastases<br />

(SLN�) SUN provided superior detection rate (99,1%). The false-negativerate<br />

was reduced from 10,7 % to 1,3%. This was attributed to the<br />

embolization <strong>of</strong> lymph vessels afferent to the metastasized (SLN�) node<br />

causing a bypass <strong>of</strong> the “lymphatic mapping” and inhibiting detection.<br />

Conclusions: The SUN–Method is comparable to “lymphatic mapping” in<br />

tumor free nodes (SLN -). If SLN is metastasized (SLN�) - SUN is superior<br />

to the standard methods in sensitivity and specificity (80%) and the<br />

false-negative-rate can be reduced. Systematic axilla sonography is an<br />

effective method for the SLN-Localisation, and <strong>of</strong>fers an excellent method<br />

for quality control during SLNB.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


Developmental Therapeutics—<strong>Clinical</strong> Pharmacology and Immunotherapy<br />

2561 General Poster Session (Board #4B), Mon, 8:00 AM-12:00 PM<br />

A randomized pilot phase I study <strong>of</strong> modified carcinoembryonic antigen<br />

(CEA) peptide (CAP1-6D)/montanide/GM-CSF-vaccine (CEA-vac) in patients<br />

(pts) with pancreatic adenocarcinoma (PC). Presenting Author:<br />

Daniel M. Geynisman, University <strong>of</strong> Chicago, Chicago, IL<br />

Background: CEA is expressed in �90% <strong>of</strong> PC and may be an appropriate<br />

immunotherapy target. CEA is poorly immunogenic due to immune tolerance;<br />

CAP1-6D, an altered peptide ligand can help bypass tolerance. We<br />

conducted a pilot randomized phase I trial in PC pts to determine the<br />

appropriate CEA-vac peptide dose required to induce an optimal cytotoxic T<br />

lymphocyte (CTL) response. Methods: Eligible pts, PS 0-1, expressed<br />

HLA-A2 and had CEA-expressing, histologically-confirmed, previouslytreated<br />

PC. Randomization: 10 �g (arm A), 100 �g (arm B) or 1000 �g<br />

(arm C). CEA-vac was given Q 2 weeks until disease progression (amended<br />

4/09 to a 24 dose maximum). Results: From 2/06-9/09 66 pts were<br />

screened for HLA-A2; 19 pts randomized to Arms A/B/C 5/8/6 are evaluable<br />

for toxicity; 14 pts (5/5/4) who received at least 3 doses <strong>of</strong> CEA-vac are<br />

evaluable for the primary immunologic endpoint. Median age: 60 (range<br />

27-86), female: 68%, PS 0: 58%. Disease stage: metastatic (M) 74%,<br />

locally advanced (LA) 5%, resected (R) 21%. Cycles: median 4 (range<br />

1-81). Mean CTL response by ELISPOT (spots per 104 CD8� cells, Arm<br />

A/B/C): 37/126/248. (A vs. C, p�0.037). CTL responses developed in<br />

20%/60%/100% <strong>of</strong> pts in Arms A/B/C. 1 LA pt in Arm C had a complete<br />

radiologic response, a strong CTL response, and remains alive at 71 mo. 1<br />

M pt in Arm B had SD for 11 mo, a strong CTL response, and is alive at 39<br />

mo. Toxicity: no grade 3/4 toxicities, 58% grade 1/2 skin toxicity.<br />

Conclusions: CTL response was dose-dependent in this randomized phase I<br />

trial; the 1 mg peptide dose elicited the most robust CTL response. <strong>Clinical</strong><br />

activity was demonstrated at the higher doses. Further evaluation <strong>of</strong> 1 mg<br />

CEA-vac with stronger adjuvants, combined with agents to overcome<br />

immune inhibitory pathways, may be warranted in PC pts. Supported by<br />

UCCCSG P30CA14599.<br />

2563 General Poster Session (Board #4D), Mon, 8:00 AM-12:00 PM<br />

Evaluation <strong>of</strong> immune activation following neoadjuvant sipuleucel-T in<br />

subjects with localized prostate cancer. Presenting Author: Nadeem A.<br />

Sheikh, Dendreon Corporation, Seattle, WA<br />

Background: Sipuleucel-T is an FDA-approved autologous cellular immunotherapy<br />

for men with asymptomatic or minimally symptomatic metastatic<br />

castrate resistant prostate cancer (mCRPC). NeoACT (Study P07-1) was<br />

undertaken to investigate neoadjuvant sipuleucel-T treatment in subjects<br />

with localized prostate cancer. Methods: In this open-label, phase 2 study<br />

(NCT00715104), subjects with localized prostate cancer received 3<br />

infusions <strong>of</strong> sipuleucel-T at approximately 2-week intervals, beginning 6–7<br />

weeks prior to radical prostatectomy (RP). Following RP, subjects were<br />

randomized 1:1 to receive / not receive a sipuleucel-T booster infusion 12<br />

weeks post-RP. Cellular composition, antigen presenting cell (APC) activation,<br />

cytokines, and T and B cell activation were pr<strong>of</strong>iled before and after<br />

each culture with PA2024, the fusion protein containing prostatic acid<br />

phosphatase used to generate sipuleucel-T. Results: Of the 42 enrolled<br />

subjects (median age: 61 years; 98% Caucasian), 38 received all 3<br />

infusions <strong>of</strong> sipuleucel-T, and 15 subjects received a booster infusion.<br />

Consistent with sipuleucel-T in mCRPC, CD54 upregulation (APC activation)<br />

was greater at the second and third infusions relative to the first<br />

(p�0.001). The expression <strong>of</strong> early T cell activation markers (CD134,<br />

CD137, CD278 and CD279) were increased in pre-culture cells obtained<br />

after the first infusion, and further increased after culture. Activated<br />

mature B cells (CD20�CD27�IgD�CD86�) increased following culture<br />

in all 3 products (p�0.01); memory B cells (CD20�CD27�IgD-CD86�)<br />

were progressively increased following the first infusion (p�0.05 third vs.<br />

first product). TNF-�, IFN-�, IL-2 were secreted at higher levels during<br />

culture <strong>of</strong> the second and third products (all p�0.001). The observed<br />

increases in CD54 upregulation, early T cell activation markers, and<br />

memory and activated mature B cells were maintained at booster treatment.<br />

Conclusions: Neoadjuvant sipuleucel-T resulted in robust immune<br />

system activation that was consistent with boosting <strong>of</strong> an immune response<br />

primed with the first infusion. Immune activation was maintained at the<br />

booster infusion 3 months following initial sipuleucel-T treatment.<br />

157s<br />

2562 General Poster Session (Board #4C), Mon, 8:00 AM-12:00 PM<br />

Lymphoid and myeloid biomarkers for clinical outcome <strong>of</strong> combined<br />

immunotherapy with granulocyte-macrophage colony-stimulating factortranduced<br />

allogeneic prostate cancer cells (GVAX) and ipilimumab in<br />

castration-resistant prostate cancer patients. Presenting Author: Alfons<br />

J. M. van den Eertwegh, VU University Medical Center Department <strong>of</strong><br />

Medical Oncology, Amsterdam, Netherlands<br />

Background: In a phase-I dose escalation trial in patients with castrationresistant<br />

prostate cancer we showed that GVAX and ipilimumab had an<br />

acceptable safety pr<strong>of</strong>ile. Moreover, we observed tumor responses and<br />

prolonged survival as compared to the Halabi predicted overall survival<br />

(OS). However, ipilimumab can also lead to severe immune-related adverse<br />

events. To avoid unnecessary exposure to this risk, it is essential to identify<br />

biomarkers that correlate with clinical activity. Methods: Patients had<br />

castration-resistant prostate cancer and were chemotherapy-naïve. They<br />

received bi-weekly GVAX for a 24 week period combined with monthly<br />

intravenous administrations <strong>of</strong> ipilimumab. Each cohort <strong>of</strong> 3 patients<br />

received an escalating dose <strong>of</strong> ipilimumab at 0·3, 1·0, 3·0 or 5·0 mg/kg. In<br />

an expansion cohort 16 patients were treated with GVAX and 3·0 mg/kg<br />

ipilimumab. Flowcytometric monitoring <strong>of</strong> lymphoid and myeloid subsets<br />

in blood were performed. Results: We observed a significantly prolonged OS<br />

for patients with high pre-treatment frequencies <strong>of</strong> CD4�CTLA-4�,<br />

CD4�PD-1�, or differentiated CD8� T cells, or low pre-treatment frequencies<br />

<strong>of</strong> differentiated CD4� T cells or CD4�CD25hiFoxP3� regulatory T<br />

cells. In contrast, increased frequencies <strong>of</strong> granulocytic Myeloid-Derived<br />

Suppressor Cells (MDSC) and high pre-treatment frequencies <strong>of</strong> monocytic<br />

CD14�HLA-DRlo/- MDSC were associated with reduced OS. Treatmentinduced<br />

CD4� T cell differentiation and CD4� and CD8� T cell activation<br />

was associated with clinical benefit. Moreover, treatment-induced activation<br />

<strong>of</strong> CD1c� conventional Dendritic Cells (cDC) and 6-sulfo LacNAc�<br />

inflammatory DC were associated with significantly prolonged OS.<br />

Conclusions: Together these data provide an immune pr<strong>of</strong>ile to predict<br />

clinical outcome. Importantly, cluster analysis revealed pre-treatment,<br />

CRPC-associated expression <strong>of</strong> CTLA-4� by CD4� T cells to be a dominant<br />

predictor for OS after GVAX/ipilimumab. This potentially biomarker for<br />

patient selection should be validated in patients treated with ipilimumab.<br />

2564 General Poster Session (Board #4E), Mon, 8:00 AM-12:00 PM<br />

Neoadjuvant sipuleucel-T in localized prostate cancer: Effects on immune<br />

cells within the prostate tumor microenvironment. Presenting Author:<br />

Lawrence Fong, University <strong>of</strong> California, San Francisco, San Francisco, CA<br />

Background: Sipuleucel-T is an FDA-approved autologous cellular immunotherapy<br />

for patients with asymptomatic or minimally symptomatic metastatic<br />

castrate resistant prostate cancer (mCRPC). To date, studies <strong>of</strong><br />

sipuleucel-T in patients with mCRPC have studied immune response in<br />

peripheral blood. The effects <strong>of</strong> sipuleucel-T on prostate tumors are<br />

unknown. Methods: NeoACT (P07-1; NCT00715104) is an open-label,<br />

phase 2 study <strong>of</strong> patients with localized prostate cancer who received<br />

sipuleucel-T prior to radical prostatectomy (RP) to examine the immunologic<br />

effects <strong>of</strong> treatment on prostate tissue. Patients received 3 infusions<br />

<strong>of</strong> sipuleucel-T at approximately 2-week intervals, beginning 6-7 weeks<br />

prior to RP. The primary endpoint was the change in the frequency <strong>of</strong><br />

lymphocytes between prostate biopsies (pre-treatment) and RP tissue<br />

(post-treatment), as assessed by immunohistochemistry (IHC). Results: The<br />

median age <strong>of</strong> the 42 enrolled patients was 61 years, and all had an ECOG<br />

performance status <strong>of</strong> 0. Thirty-eight patients received all 3 pre-RP<br />

sipuleucel-T infusions. To date, tissue IHC analysis has been completed on<br />

32 patients. Treatment-related AEs were manageable and transient.<br />

Sipuleucel-T did not appear to impact surgery, as judged by operative<br />

complications, procedure time, and estimated blood loss. Frequent events<br />

that occurred �1 day after infusion (�10% <strong>of</strong> patients) were fatigue,<br />

headache, and myalgia. Significant increases (�3 fold) in CD3� and<br />

CD4� T cell populations were observed at the tumor interface (where<br />

benign and malignant glands interface), compared with the pre-treatment<br />

biopsy, benign RP tissue, and tumor RP tissue (ANOVA post hoc Newman-<br />

Keuls test: p�0.0001 for each comparison). FoxP3� CD4� T cells were<br />

also increased (p�0.0005) at the tumor interface, but represented a small<br />

fraction <strong>of</strong> the observed CD4� T cells. Conclusions: Neoadjuvant sipuleucel-T<br />

treatment is associated with an increased frequency <strong>of</strong> T cells in<br />

prostate cancer tissue at the interface <strong>of</strong> the benign and malignant glands.<br />

These data suggest that sipuleucel-T can modulate the presence <strong>of</strong><br />

lymphocytes at the prostate tumor site. Work is ongoing to more fully<br />

characterize the immune response.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


158s Developmental Therapeutics—<strong>Clinical</strong> Pharmacology and Immunotherapy<br />

2565 General Poster Session (Board #4F), Mon, 8:00 AM-12:00 PM<br />

IFN-� to improve immunotherapy for melanoma. Presenting Author: Marco<br />

Donia, Center for Cancer Immune Therapy, Department <strong>of</strong> Hematology,<br />

Copenhagen University Hospital Herlev, Herlev, Denmark<br />

Background: Adoptive cell therapy (ACT) with tumor infiltrating lymphocytes<br />

(TILs) holds the promise to change the long-term prognosis <strong>of</strong><br />

patients with metastatic melanoma. In this study we have explored<br />

strategies to improve the efficacy <strong>of</strong> cell products for infusion through<br />

tumor immune-sensitization. Methods: We examined whether objective<br />

responses in our pilot ACT trial (NCT937625) were associated with the<br />

number <strong>of</strong> tumor-reactive T cells infused. Subsequently, we assessed the<br />

ability <strong>of</strong> immune-sensitizing agents to modulate the constitutive response<br />

<strong>of</strong> 12 clinical grade TIL products to matched autologous short-term<br />

cultured melanoma cell lines. Results: Our data showed that a high absolute<br />

number <strong>of</strong> infused tumor-reactive T cells is critical to achieve an objective<br />

response. In addition, a defective population <strong>of</strong> tumor-specific CD8� and<br />

CD4� T cells unable to exert antitumor effector functions ex vivo was<br />

detected in most TIL products. However, an antitumor specific response<br />

could be restored by pretreatment <strong>of</strong> the autologous tumor cells with low<br />

dose IFN-�. Of note, IFN-� treatment was invariably associated with<br />

increased cancer immunogenicity as demonstrated by up-regulation <strong>of</strong><br />

MHC molecules. Conclusions: Combination strategies represent the next<br />

frontier <strong>of</strong> cancer immunotherapy. Although previous trials in melanoma<br />

showed that IFN-� does not mediate clinical benefit when used as<br />

monotherapy, our findings show that its ability to increase tumor cell<br />

immunogenicity may enhance the efficacy <strong>of</strong> current immune-based<br />

therapies such as ACT through improved activation <strong>of</strong> pre-existing but<br />

unresponsive tumor specific lymphocytes. Therefore, additional studies on<br />

the possibility <strong>of</strong> a sequential combination <strong>of</strong> low dose IFN-� and ACT are<br />

highly warranted.<br />

2567 General Poster Session (Board #4H), Mon, 8:00 AM-12:00 PM<br />

Human pharmacokinetic (PK) characterization <strong>of</strong> the novel dual-action<br />

anti-HER3/EGFR antibody MEHD7945A (MEHD) in patients with refractory/<br />

recurrent epithelial tumors. Presenting Author: Manuel Hidalgo, START-<br />

Madrid, Centro Integral Oncológico Clara Campal, Madrid, Spain<br />

Background: MEHD is a novel dual-action human IgG1 antibody that blocks<br />

ligand binding to HER3 and EGFR, and elicits antibody-dependent<br />

cell-mediated cytotoxicity (ADCC). MEHD demonstrates single-agent activity<br />

in a broad panel <strong>of</strong> tumor models, including models resistant to<br />

anti-HER3 or anti-EGFR treatment alone. The objective <strong>of</strong> this analysis was<br />

to characterize the PK <strong>of</strong> MEHD associated with body weight (BW)-based<br />

dosing used in a phase I study in patients with epithelial tumors and to<br />

evaluate the potential for using fixed dosing in future studies. Methods:<br />

Preliminary non-compartmental and population PK analyses were performed<br />

using patient data from the dose-escalation stage [1, 4, 10, 15, 22,<br />

and 30 mg/kg every two weeks (q2w)] and expansion stage (14 mg/kg q2w)<br />

<strong>of</strong> the phase I study. Patient demographic data and other relevant clinical<br />

covariates were evaluated in the population analysis. PK simulation <strong>of</strong><br />

1000 subjects with a log-normal BW distribution was performed to<br />

compare the inter-individual variability <strong>of</strong> MEHD exposure following fixed<br />

or BW-based dosing. Results: As expected,MEHD exhibited nonlinear PK.<br />

In the noncompartmenal analysis, the apparent clearance (CL) decreased<br />

in a dose-dependent fashion (about 40 to 9.9 mL/day/kg from 1 to 30<br />

mg/kg) and approached linearity at doses �10 mg/kg (q2w). In the<br />

population analysis, the PK pr<strong>of</strong>ile <strong>of</strong> MEHD was well described by a two<br />

compartment model with linear and nonlinear clearance. The targetmediated<br />

clearance was consistent with that <strong>of</strong> anti-EGFR antibodies. The<br />

nonspecific CL and central volume <strong>of</strong> distribution (V1) values were<br />

approximately 6 mL/day/kg and 52.4 mL/kg, respectively. BW had a<br />

moderate effect on V1, but not on CL. PK simulations suggest that,<br />

compared with BW-based dosing, fixed dosing would result in less<br />

inter-individual variability in MEHD exposure. Both 1100 mg q2w or 1650<br />

mg q3w <strong>of</strong> MEHD achieve the targeted therapeutic exposure. Conclusions:<br />

The dual-action antibody MEHD demonstrated PK consistent with anti-<br />

EGFR antibodies. Fixed dosing <strong>of</strong> MEHD on an every 2 or 3 week schedule<br />

is supported.<br />

2566 General Poster Session (Board #4G), Mon, 8:00 AM-12:00 PM<br />

A first-in-human phase Ib study to evaluate the MEK inhibitor GDC-0973,<br />

combined with the pan-PI3K inhibitor GDC-0941, in patients with advanced<br />

solid tumors. Presenting Author: Patricia LoRusso, Karmanos<br />

Cancer Institute, Detroit, MI<br />

Background: Both RAS/RAF/MEK and PI3K/Akt signaling pathways are<br />

deregulated in many tumor types. Targeting both pathways may be more<br />

efficacious than targeting either pathway alone. In preclinical models,<br />

concurrent administration <strong>of</strong> GDC-0973, a potent, selective, MEK1/2<br />

inhibitor and GDC-0941, a potent class I PI3K inhibitor, shows improved<br />

efficacy compared to either agent alone dosed continuously or intermittently.<br />

Methods: A phase Ib dose-escalation study with 3�3 design was<br />

initiated in patients (pts) with advanced solid tumors to evaluate the safety<br />

and pharmacokinetics (PK) <strong>of</strong> oral dosing <strong>of</strong> GDC-0973 and GDC-0941.<br />

Pts received: concurrent GDC-0973 � GDC-0941 once daily (qd) on a 21<br />

day on/7 day <strong>of</strong>f (21/7) schedule; intermittent GDC-0973 on Days 1, 4, 8,<br />

11, 15, 18 <strong>of</strong> a 28 day cycle � GDC-0941 qd on a 21/7 schedule (MEK<br />

int); or GDC-0973 � GDC-0941 qd on a 7 day on /7 day <strong>of</strong>f schedule (7/7).<br />

Starting doses were 20 mg GDC-0973 � 80 mg GDC-0941 (21/7), 100 mg<br />

GDC-0973 � 130 mg GDC-0941 (MEK int); 40 mg GDC-0973 � 130 mg<br />

GDC-0941 (7/7). Serial plasma PK samples, FDG-PET, and CT scans were<br />

obtained. Results: 78 pts have enrolled. DLTs were G3 lipase (n�1), G4<br />

CPK elevation (n�1). Compared to the 21/7 MTD <strong>of</strong> 40 mg GDC-0973 �<br />

100 mg GDC-0941, higher doses <strong>of</strong> GDC-0973 � GDC-0941 were<br />

tolerated on the MEK int schedule. Overall, adverse events related to the<br />

study drug combination in � 20% pts were diarrhea, rash, nausea, fatigue,<br />

vomiting, decreased appetite, dysgeusia, and elevated CPK. Preliminary<br />

analysis indicated PK <strong>of</strong> GDC-0973 and GDC-0941 are not altered when<br />

dosed in combination. Of 46 evaluable pts, 26 had an FDG-PET partial<br />

metabolic response (� 20% decrease in mean SUVmax from baseline) at �1<br />

time points. <strong>Part</strong>ial responses were observed in 3 pts (mBRAF melanoma,<br />

mBRAF pancreatic ca, mKRAS endometrioid ca); 5 pts had stable disease<br />

� 5 months. Conclusions: Combination dosing <strong>of</strong> GDC-0973 and GDC-<br />

0941 is generally well tolerated, with toxicities similar to those observed in<br />

single agent GDC-0973 and GDC-0941 phase 1 trials. There are early signs<br />

<strong>of</strong> anti-tumor activity. Dose escalation on MEK int and 7/7 schedules<br />

continues and updated data will be presented.<br />

2568 General Poster Session (Board #5A), Mon, 8:00 AM-12:00 PM<br />

A phase I study <strong>of</strong> MEHD7945A (MEHD), a first-in-class HER3/EGFR<br />

dual-action antibody, in patients (pts) with refractory/recurrent epithelial<br />

tumors: Expansion cohorts. Presenting Author: Andres Cervantes-Ruiperez,<br />

Department <strong>of</strong> Hematology and Medical Oncology, INCLIVA, University <strong>of</strong><br />

Valencia, Valencia, Spain<br />

Background: Dysregulated human epidermal growth factor receptor tyrosine<br />

kinase (HER RTK) signaling is an important driver <strong>of</strong> tumor growth,<br />

metastasis, and survival. Extensive co-expression and heterodimerization<br />

suggest that simultaneous blockade <strong>of</strong> multiple HER RTKs may be more<br />

effective than blockade <strong>of</strong> a single RTK. MEHD is a novel dual-action<br />

human IgG1 antibody. Each antigen-binding fragment blocks ligand<br />

binding to HER3 or EGFR, and intended to inhibit signaling from all major<br />

ligand-dependent HER dimers. MEHD has single-agent activity in multiple<br />

tumor models including models resistant to anti-HER3 or anti-EGFR.<br />

Methods: This Phase I study evaluated safety, tolerability, pharmacokinetics<br />

(PK), and pharmacodynamic (PD) activity <strong>of</strong> intravenous MEHD every 2<br />

weeks (q2w). Tumor PD assessments were pre- and post-dose FDG-PET<br />

and IHC for pS6, pPRAS40 or pERK in tumor biopsies. Tumor CT<br />

assessments were performed q8w. No dose-limiting toxicity (DLT) was<br />

observed in six 3�3 cohorts from 1-30 mg/kg (n�30). We report results<br />

from 4 tumor-specific cohorts receiving 14 mg/kg MEHD (recommended<br />

Phase II dose). Results: As <strong>of</strong> 21 Nov 2011, 36 pts (CRC�12, NSCLC�9,<br />

SCCHN�10, pancreatic�5), median age 62.5 (35–87), all PS 0-1,<br />

median # prior regimens 3.5 (1-8), received a median <strong>of</strong> 4 doses (1-9) <strong>of</strong><br />

MEHD; 18 pts remain on study. PK data are consistent with human<br />

anti-EGFR antibodies. No related Grade (G) �3 adverse events (AEs) have<br />

been observed. Common related G1/2 AEs included rash/dermatitis (53%),<br />

diarrhea (36%), fatigue (22%), paronychia (19%), dry skin, nausea, and<br />

decreased appetite (all 17%), asthenia and stomatitis/oral pain (all 14%).<br />

Related AEs � 24 h after first infusion included G1/2 headache (42%),<br />

fever (33%), and chills (17%), and decreased in intensity and frequency<br />

with later infusions. Early PD data indicate target inhibition in 8/36 pts<br />

(SCCHN�2, NSCLC�2, CRC�4), and best response by RECIST includes 2<br />

PR (both SCCHN), 6 pts with SD � 8 weeks (NSCLC�2, CRC�4), 7/8<br />

previously treated with EGFR inhibitors. Conclusions: MEHD at 14 mg/kg<br />

q2w has an encouraging safety pr<strong>of</strong>ile and evidence <strong>of</strong> anti-tumor activity.<br />

Phase II studies are planned.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


Developmental Therapeutics—<strong>Clinical</strong> Pharmacology and Immunotherapy<br />

2569 General Poster Session (Board #5B), Mon, 8:00 AM-12:00 PM<br />

Patient-specific immunotherapy utilizing putative tumor stem cells in<br />

patients with metastatic melanoma: A pooled analysis comparing tumor<br />

cell and dendritic cell vaccines in two phase II trials and a randomized<br />

phase II trial. Presenting Author: Robert Owen Dillman, Hoag Institute for<br />

Research and Education and Hoag Family Cancer Institute, Newport<br />

Beach, CA<br />

Background: Metastatic melanoma is seldom cured, even in patients who<br />

achieve a complete remission, because new sites <strong>of</strong> disease emerge. Autologous,<br />

proliferating, self-renewing tumor cells (putative tumor stem cells and/or<br />

early progenitor cells), are critical to establishment <strong>of</strong> new depots <strong>of</strong> metastatic<br />

cancer, and may be excellent sources <strong>of</strong> antigen for vaccines. These trials<br />

addressed the impact on survival from immunizing with antigens from such<br />

cells. Methods: Data was pooled from 3 successive phase II trials, all <strong>of</strong> which<br />

included patients with documented metastatic melanoma, who were treated in<br />

protocols that utilized antigens from cell cultures <strong>of</strong> autologous tumor cells.<br />

S.C. injections were given weekly for 3 weeks, then monthly for 5 months.<br />

1992-2000: 74 patients were injected with irradiated tumor cells (TC).<br />

2000-2006: 54 patients were injected with autologous dendritic cells (DC)<br />

that had been co-cultured with irradiated autologous tumor cells (NCI-V01-<br />

1646). 2007-2011: in a randomized phase II trial, 24 patients were injected<br />

with TC, and 18 with DC (NCT00436930). Results: The table summarizes<br />

overall survival (OS) in each trial. In the pooled analysis there were 98 TC and<br />

72 DC patients. Characteristics were similar in terms <strong>of</strong> age (51, 52), male<br />

gender (62%, 62%), no evidence <strong>of</strong> disease at the time <strong>of</strong> treatment (46%,<br />

47%), and presence <strong>of</strong> M1c visceral disease at the time <strong>of</strong> treatment (13%,<br />

14%). OS was longer in patients treated with DC (median 63.1 vs 20.2<br />

months, 5-year OS 51% vs 26%, p�0.0002 Mantle-Cox log-rank test). The<br />

difference in OS in the randomized trial is also significant (p�0.007).<br />

Conclusions: Patient-specific DC vaccines primed with antigens from autologous<br />

proliferating, self-renewing tumor cells are associated with encouraging<br />

long-term survival rates, and are superior to patient-specific TC vaccines in<br />

populations <strong>of</strong> patients who have been diagnosed with metastatic melanoma.<br />

Vaccine # patients # deaths Median OS 2-yr OS 5-yr OS<br />

TC 74 60 20.3 mos 45% 28%<br />

DC 54 31 58.4 mos 72% 50%<br />

TC 24 16 15.9 mos 31% ---<br />

DC 18 5 Not reached 72% ---<br />

2571 General Poster Session (Board #5D), Mon, 8:00 AM-12:00 PM<br />

Correlation <strong>of</strong> interferon-g (IFN-�) response with survival in a phase II<br />

hyperacute (HAL) immunotherapy trial for non-small cell lung cancer<br />

(NSCLC). Presenting Author: John Charles Morris, University <strong>of</strong> Cincinnati,<br />

Cincinnati, OH<br />

Background: Lung cancer is the leading cause <strong>of</strong> cancer-related mortality.<br />

Patients progressing after initial systemic therapy have a poor prognosis.<br />

Thus, new therapies are needed. HAL immunotherapy exploits the hyperacute<br />

rejection <strong>of</strong> a xenotransplant as defense mechanism to initiate<br />

anti-tumor immune response by administering genetically modified allogeneic<br />

tumor cells expressing the �Gal moieties on their cell surface. HAL<br />

immunotherapy utilizes �Gal epitopes and natural human antibodies (Ab)<br />

against these targets as the proposed mechanism for anti-tumor immunity.<br />

Methods: We completed a phase II study <strong>of</strong> HAL immunotherapy in patients<br />

with metastatic or recurrent NSCLC, age �18, ECOG PS �2, �2 prior<br />

systemic therapies. Trial objectives were to determine response rate, safety<br />

and immunological responses. Patients received 300 x 106 HAL cells every<br />

2 weeks for 8 doses. Response was determined using RECIST and adverse<br />

events were assessed using CTCAE v3. Immune responses were assessed<br />

by changes in serum anti-�Gal titers and IFN-� response. Results: Twentyeight<br />

patients were treated. Eight (29%) demonstrated stable disease (SD)<br />

�16 wks including one patient that initially progressed and later regressed,<br />

surviving over 40 months. Median overall survival (OS) was 11.3 months<br />

(95% CI 3.8-21.9). Post vaccination, all patients responded by increasing<br />

anti-�Gal Ab levels (2-100 fold) and 61% (11/18) <strong>of</strong> tested patients<br />

showed increases in IFN-� levels (by ELISPOT). Interestingly, patients with<br />

increased IFN-� responses after vaccination (�10-500 fold-increase<br />

compared to baseline) had a median survival <strong>of</strong> 21.9 months compared to<br />

5.5 months in patients with lesser IFN-� responses after vaccination<br />

(p�0.001). In addition, patients with higher IFN-� responses showed<br />

reactivity to a NSCLC cell line that was not a component <strong>of</strong> HAL, suggesting<br />

cross-priming to shared tumor antigens. Conclusions: HAL immunotherapy<br />

is safe and feasible. The median survival compared favorably to that<br />

reported in patients receiving 2nd line chemotherapy for relapsed or<br />

advanced NSCLC. The correlation between IFN-� response and survival is<br />

highly encouraging.<br />

2570 General Poster Session (Board #5C), Mon, 8:00 AM-12:00 PM<br />

Effect <strong>of</strong> matrix metalloproteinase-2 on antitumor immunity. Presenting<br />

Author: Emmanuelle Godefroy, New York University, New York, NY<br />

Background: Cancer cells, including melanoma, can be highly resistant to<br />

traditional treatments such as chemotherapy and radiotherapy. As a result,<br />

a lot <strong>of</strong> effort has been put into developing immune therapies to treat<br />

cancer patients. The main barrier to design effective immunotherapies is<br />

the immunosuppression induced by the tumor. Matrix metalloproteinase-2<br />

(MMP-2) is over-expressed in most cancers including melanoma, and its<br />

expression is associated with increased dissemination and poorer survival/<br />

prognosis. Here, we uncovered an immunosuppressive role <strong>of</strong> MMP-2 in<br />

inducing ineffective/detrimental TH2 immune responses and its underlying<br />

mechanisms. Methods: Human dendritic cells (DCs) were cultured in the<br />

presence <strong>of</strong> MMP-2 or various TLR agonists. DCs responses were monitored<br />

using immunostaining, ELISA or cytometric bead array methods. Autologous<br />

CD4� T cells were stimulated using these conditioned tumorassociated<br />

antigen (TAA)-pulsed DCs. TAA-specific CD4� T cells were<br />

cloned and characterized using the same techniques as for DCs. Results:<br />

Herewe showed that MMP-2-conditioned human DCs primed naïve CD4� T<br />

cells into an inflammatory TH2 phenotype, i.e. mainly secreting TNF�, IL-4<br />

and IL-13 and expressing GATA3. Accordingly, we detected MMP-2specific<br />

CD4� T cells displaying the same inflammatory TH2 pr<strong>of</strong>ile in<br />

tumor-infiltrating lymphocytes from melanoma patients. We revealed the<br />

underlying mechanisms: on the one hand, MMP-2 was found to degrade the<br />

type-I IFN receptor IFNAR1, thereby preventing STAT1 phosphorylation,<br />

which is necessary for IL-12 production. On the other hand, MMP-2<br />

triggers the Toll-like receptor (TLR)-2 on DCs, which leads to NF-kB<br />

activation and OX40L expression. Both lack <strong>of</strong> IL-12 and over-expression <strong>of</strong><br />

OX40L were found responsible for skewing T cell responses towards a<br />

detrimental TH2 phenotype. Conclusions: MMP-2, therefore, acts as an<br />

endogenous TH2 �conditioner� and may underlie the prevalence <strong>of</strong> detrimental<br />

TH2 responses in cancer. Our findings also described the responsible<br />

MMP-2-dependent mechanisms, which open the way to novel therapeutic<br />

strategies and/or targets to skew anti-tumor CD4� T cell responses towards<br />

a more efficient anti-tumor TH1 phenotype to treat cancer patients.<br />

2572 General Poster Session (Board #5E), Mon, 8:00 AM-12:00 PM<br />

Does the addition <strong>of</strong> molecular targeted therapy to standard treatments<br />

lead to better or worse outcomes overall? A systematic review <strong>of</strong> EGFRtargeted<br />

therapies used in combination with standard treatments. Presenting<br />

Author: Jennifer Rauw, University <strong>of</strong> Toronto, Toronto, ON, Canada<br />

Background: Combining novel targeted therapies with standard treatment is<br />

a common strategy in drug development. The primary aim <strong>of</strong> this systematic<br />

review was to provide a summary <strong>of</strong> the outcomes from studies combining<br />

EGFR targeted therapy (ETT) with standard treatments. Methods: A PubMed<br />

(1975-Jan, 2012) and ASCO (2005-2011) abstract database search was<br />

performed. Eligible studies were RCTs that compared standard therapies<br />

(chemotherapy, radiation therapy, or hormonal therapy) to standard therapy<br />

plus an ETT. Efficacy outcomes: overall survival (OS), progression free<br />

survival (PFS), objective response rate (ORR) time to progression (TTP),<br />

clinical benefit rate (CBR), and toxicity (total and grade 3�) were recorded.<br />

If any toxicity was significantly more frequent in one arm it was coded as<br />

such. Results: 128 studies (60 manuscripts, 68 abstracts) met criteria.<br />

Median study enrolment was 230 patients, with breast, lung, and colorectal<br />

cancer as the main tumor sites. ETT, cetuximab (41%), trastuzumab<br />

(19%), gefitinib (13%), erlotinib (13%), and lapatinib (16%), was combined<br />

with platinums, 5FU, taxanes, nucleoside analogs; hormonal therapy<br />

and/or radiation therapy. Refer to the table for efficacy results. Most<br />

frequent toxicities (common and grade 3�) in the combined arm were rash,<br />

diarrhea, hematological toxicity and fatigue. Conclusions: Despite enthusiasm<br />

for combining standard treatments with targeted therapies, we have<br />

demonstrated mixed efficacy results, with marginal benefit and worse<br />

toxicity. Quality <strong>of</strong> life (Q <strong>of</strong> L) was rarely reported. A similar analysis is<br />

underway for VEGFR, and mTor inhibitors, but raises the question <strong>of</strong> how<br />

best to approach the question <strong>of</strong> combination therapy.<br />

Outcome<br />

Not<br />

reported<br />

Significantly<br />

worse in<br />

combined arm<br />

No significant<br />

difference<br />

between arms<br />

159s<br />

Significantly<br />

better in the<br />

combined arm<br />

OS 60 (51%) 1 (0.8%) 39 (31%) 23 (18%)<br />

DFS 66 (52%) 1 (0.8%) 29 (23%) 32 (25%)<br />

TTP 106 (83%) 0 10 (8%) 12 (9%)<br />

CBR 108 (84%) 0 13 (10%) 7 (6%)<br />

ORR 73 (57%) 0 29 (23%) 26 (20%)<br />

Toxicity 58 (45%) 47 (36%) 22 (17%) 1 (0.8%)<br />

Q<strong>of</strong>L 110 (86%) 0 12 (9%) 6 (5%)<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


160s Developmental Therapeutics—<strong>Clinical</strong> Pharmacology and Immunotherapy<br />

2573^ General Poster Session (Board #5F), Mon, 8:00 AM-12:00 PM<br />

Messenger RNA vaccination and B-cell responses in NSCLC patients.<br />

Presenting Author: Martin Sebastian, Medizinische Klinik II Hämatologie/<br />

Onkologie, Rheumatologie, Infektiologie, HIV Klinikum der J.W. Goethe-<br />

Universität Frankfurt, Frankfurt am Main, Germany<br />

Background: Vaccination with mRNA is a novel technology in cancer<br />

immunotherapy. CV9201 consists <strong>of</strong> self-adjuvanted mRNA molecules<br />

(RNActive) coding for five non small cell lung cancer (NSCLC)-associated<br />

tumor antigens (MAGE-C1, MAGE-C2, NY-ESO-1, BIRC5, 5T4). We report<br />

final results <strong>of</strong> a phase I/IIa trial <strong>of</strong> CV9201 in patients (pts.) with NSCLC.<br />

Methods: Pts. with stage IIIB/IV NSCLC with a response or stable disease<br />

after first-line chemotherapy or chemoradiation were eligible. Cohorts <strong>of</strong> 3<br />

pts. were treated at three dose levels (400�g, 800�g and 1600�g<br />

CV9201) and observed for DLTs to select the highest tolerated dose for<br />

phase IIa. Primary endpoint was safety and tolerability; secondary endpoints<br />

were immune response, clinical efficacy and survival. Pts. received<br />

up to five vaccinations <strong>of</strong> CV9201 within 15 weeks. Antigen-specific<br />

immune responses against each <strong>of</strong> the 5 antigens were measured at<br />

baseline, and two weeks after the 3rd and 5th vaccination. Frequency <strong>of</strong><br />

lymphocyte subsets and expression <strong>of</strong> activation and maturation markers<br />

were measured and retrospectively correlated with immunological and<br />

clinical parameters. Results: 46 pts. were included (9 phase I; 37 phase<br />

IIa); No DLTs occurred, and the 1600 �g dose was investigated in phase<br />

IIa. The most frequent related adverse events were mild to moderate<br />

injection site reactions and flu-like symptoms. 3 patients (7%) had<br />

potentially related grade 3 AEs (fatigue, injection site granuloma, asthma<br />

attack) and no related SAEs were reported. There were no objective<br />

responses. Data on PFS and survival will be presented. Antigen specific<br />

immune responses against at least one antigen were induced in 65% <strong>of</strong> pts.<br />

(39% cellular and 49% humoral). Consistently, a significant �2 fold<br />

increase <strong>of</strong> pre germinal center B cells (pGCB) was observed in 61% <strong>of</strong> pts.<br />

This increase <strong>of</strong> pGCB correlated significantly (p�0.0028) with increase <strong>of</strong><br />

total CD4 effector T cells. Frequency <strong>of</strong> CD 4 T Reg cells did not increase<br />

during treatment. Conclusions: Vaccination with CV9201 has a favorable<br />

safety pr<strong>of</strong>ile and induces T and B cell responses against all included<br />

antigens. Vaccine-induced increase <strong>of</strong> pGCB is a new finding and might be<br />

used as a biomarker in cancer immunotherapy.<br />

2575 General Poster Session (Board #5H), Mon, 8:00 AM-12:00 PM<br />

Bispecific Fynomer-antibody fusion proteins targeting two epitopes on<br />

HER2. Presenting Author: Frederic Mourlane, Covagen AG, Zurich-<br />

Schlieren, Switzerland<br />

Background: Fynomers are small binding proteins (7 kDa) derived from the<br />

SH3 domain <strong>of</strong> Fyn kinase. They can be engineered to yield specific and<br />

high-affinity binding domains to target proteins <strong>of</strong> interest. In the past, we<br />

have isolated Fynomers with excellent physico-chemical properties binding<br />

to 16 different antigens. One important application <strong>of</strong> the Fynomer<br />

technology represents the genetic fusion <strong>of</strong> a Fynomer to an antibody to<br />

provide bispecific fusion proteins. Methods: Using phage display technology<br />

we have isolated Fynomers binding to tumor-associated antigens. After<br />

genetic fusion <strong>of</strong> these Fynomers to antibodies <strong>of</strong> interest the resulting<br />

bispecific proteins were evaluated in vitro and in vivo for their antitumoral<br />

activity. Results: The fusion <strong>of</strong> Fynomers to the antibodies <strong>of</strong> interest did not<br />

alter the favorable biophysical properties <strong>of</strong> the antibodies. First, the<br />

Fynomer-antibody fusion proteins could be purified with the same high<br />

yields from the supernatant <strong>of</strong> transiently transfected CHO cells as the<br />

unmodified antibodies (in the range <strong>of</strong> 100 mg/L). Second, the purified<br />

fusion proteins were monomeric and showed no signs <strong>of</strong> aggregation even<br />

after four months <strong>of</strong> storage at 4 °C or -20 °C in PBS as determined by size<br />

exclusion chromatography. Third, the Fc-mediated effector functions <strong>of</strong><br />

antibodies were not altered by their fusion with Fynomers. Antibodydependent<br />

cellular cytotoxicity (ADCC) was maintained, and the affinity to<br />

the neonatal Fc-receptor (FcRn) was similar as observed for the unmodified<br />

antibody. In addition, the bispecific Fynomer-antibody fusion proteins<br />

demonstrated excellent growth inhibition <strong>of</strong> tumor cells in vitro and high<br />

efficacy in vivo in tumor xenograft mouse models. Conclusions: These<br />

encouraging preclinical results indicate that the bispecific Fynomerantibody<br />

fusions have highly promising properties with a great potential for<br />

further preclinical and clinical development.<br />

2574 General Poster Session (Board #5G), Mon, 8:00 AM-12:00 PM<br />

Adoptive cell therapy for melanoma patients using tumor-infiltrating<br />

lymphocytes, lymphodepleting chemotherapy, and low-dose interleukin-2.<br />

Presenting Author: Eva Ellebaek, Department <strong>of</strong> Oncology and Center for<br />

Cancer ImmuneTherapy, Department <strong>of</strong> Hematology, Copenhagen University<br />

Hospital Herlev, Herlev, Denmark<br />

Background: Adoptive T cell therapy is based on isolation <strong>of</strong> tumor<br />

infiltrating lymphocytes (TIL) from autologous tumor, in vitro activation and<br />

expansion, and the reinfusion <strong>of</strong> these cells into a lymphodepleted patient.<br />

Together with high-dose interleukin (IL)-2 this treatment has in a few other<br />

countries successively been given to patients with advanced malignant<br />

melanoma and an impressive 50% response rate has been observed. Here<br />

we report the experience from a Danish Translational Research Center<br />

using low-dose subcutaneous IL-2 injections. Methods: A pilot trial including<br />

patients with metastatic melanoma, PS �1, age �70, measurable and<br />

progressive disease, and no CNS involvement. Six patients were treated<br />

with lymphodepleting chemotherapy, TIL infusion, and 14 days <strong>of</strong> low-dose<br />

IL-2, 2 MIU/day. Results: Low-dose IL-2 considerably decreased the<br />

toxicity <strong>of</strong> the treatment with no grade 3-4 events related to this drug. Two<br />

<strong>of</strong> the 6 treated patients have an ongoing complete response (30� and 7�<br />

months), 2 patients had stable disease (4.5 and 5 months) and 2 patients<br />

progressed shortly after treatment. Five patients went through surgery but<br />

were not treated because <strong>of</strong> rapid disease progression (2), development <strong>of</strong><br />

brain metastases (2) or the inability to grow cells (1). Tumor-reactivity <strong>of</strong><br />

the infused cells and peripheral monocytes before and after therapy were<br />

analyzed. High tumor responses in the infusion products were correlated to<br />

clinical response and also an induction <strong>of</strong> tumor reactive T cells were seen<br />

in the peripheral blood for 1 patient in complete response. Conclusions:<br />

Complete and durable responses are induced after treatment with adoptive<br />

cell transfer in combination with low-dose IL-2 questioning the need for<br />

high and toxic doses <strong>of</strong> IL-2.<br />

2576 General Poster Session (Board #6A), Mon, 8:00 AM-12:00 PM<br />

A phase I study <strong>of</strong> the safety and pharmacodynamic effects <strong>of</strong> everolimus in<br />

combination with lenalidomide in patients with advanced solid malignancies.<br />

Presenting Author: Ta<strong>of</strong>eek Kunle Owonikoko, Winship Cancer Institute,<br />

Emory University, Atlanta, GA<br />

Background: Everolimus (E), an mTOR inhibitor, and Lenalidomide (L) are<br />

both potent anti cancer agents with immunomodulatory effects. Preclinical<br />

evidence <strong>of</strong> enhanced cytotoxicity <strong>of</strong> the combination <strong>of</strong> an mTOR inhibitor<br />

and lenalidomide provided the rationale for this phase I study. Methods:<br />

Patients with advanced solid malignancies, ECOG performance status (PS)<br />

0-2 and adequate organ function were eligible. Using standard 3�3 dose<br />

escalation schema, patients were treated in cohorts <strong>of</strong> three with increasing<br />

doses <strong>of</strong> E (5mg, 10mg) and L (10, 15, 20, 25mg) on day 1-28 <strong>of</strong> a 28-day<br />

cycle. Treatment cycles were repeated until disease progression by RECIST<br />

criteria or intolerable toxicity. Pharmacodynamic (PD) effects <strong>of</strong> treatment<br />

on circulating B and T lymphocytes subsets were assessed by multiparameter<br />

flow cytometry at baseline and after 2 cycles. Results: We enrolled 21<br />

patients (thyroid-5, salivary gland-5, colon-4, sarcoma-2, and others-5);<br />

median age-58 (29-74); male-13; ECOG PS <strong>of</strong> 0, 1, 2 (3/17/1). Salient<br />

grade 3/4 toxicities included rash (67%), anemia (19%) and vomiting (5%)<br />

without dose limiting toxicities. The median number <strong>of</strong> completed cycles<br />

was3(1-�15) and best response in 14 <strong>of</strong> 18 evaluable patients was<br />

stable disease (range 2- �14 months); median PFS was 116 days (14-<br />

�498). The RP2D <strong>of</strong> E and L was defined as 10mg and 25mg once daily,<br />

respectively. PD endpoint analysis after 2 cycles <strong>of</strong> treatment showed a<br />

significant increase in activated cytotoxic T cell subset (CD8�ICOS1�) in<br />

patients with salivary or thyroid cancers compared to other tumor types<br />

(�2051.0 vs. �394 vs. -1687.4 respectively; p�0.0303) and a trend for<br />

an increase in patients with non-progressing tumors (�661.7 vs. -384.1;<br />

p�0.0988). Other significant correlations were: Changes in total B-cells<br />

(CD3-CD19�) with PFS; NK cells with age (p�0.0211) and activated T<br />

cells (CD3�CD69�; CD3�CD62L-; p�0.01) with gender. Conclusions:<br />

The combination <strong>of</strong> E and L is well tolerated at the recommended single<br />

agent doses and showed promising efficacy in neuroendocrine and salivary<br />

adenoidcystic carcinoma. Modulation <strong>of</strong> activated T cell subsets<br />

(CD8�ICOS1�) correlates with efficacy <strong>of</strong> these agents.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


Developmental Therapeutics—<strong>Clinical</strong> Pharmacology and Immunotherapy<br />

2577 General Poster Session (Board #6B), Mon, 8:00 AM-12:00 PM<br />

Vaccine therapy with tumor-specific mutated ras peptides and IL-2,<br />

GM-CSF, or both in adult patients with solid tumors. Presenting Author:<br />

Osama E. Rahma, National Institutes <strong>of</strong> Health/National Cancer Institute,<br />

Bethesda, MD<br />

Background: We have shown in previous trials that vaccination with mutant<br />

ras peptides corresponding to the tumor mutation is feasible and can<br />

induce specific immune responses against the mutant proteins. Immune<br />

adjuvants are used to enhance the effect <strong>of</strong> antigen vaccines. Here, we<br />

tested the ras vaccine in combination with Interleukin-2 (IL-2), GM-CSF, or<br />

the combination <strong>of</strong> both in patients with advanced malignancies. Methods:<br />

We treated 53 patients with advanced cancers (38 CRC, 11 pancreatic, 3<br />

NSCLC and 1 cholangiocarcinoma) with 5000�g <strong>of</strong> the corresponding<br />

mutant ras peptide given SQ along with IL-2 (Arm A), GM-CSF (Arm B) or<br />

both (Arm C). IL-2 was given SQ at 6.0 million IU/m2/day starting day 5, 5<br />

days/week for 2 weeks. GM-CSF was given SQ in a dose <strong>of</strong> 100 �g/day one<br />

day prior to each vaccination for 4 days. Vaccines were repeated every 4<br />

weeks for a maximum <strong>of</strong> 15 cycles or until disease progression. Results: The<br />

median PFS and OS for the full cohort was 3.6, and 16.9, respectively.<br />

Median PFS was 3.9, 3.2 and 3.6 months for arm A, B, C, respectively.<br />

Median OS was 17.3, 20.8 and 9.1 months, respectively. There was no<br />

difference in PFS or OS between the three arms (p� 0.74 and 0.99,<br />

respectively). In a subgroup analysis, patients with advanced CRC had a<br />

median PFS and OS <strong>of</strong> 3.5 and 14.2 months, respectively. Most adverse<br />

events were grade I-II toxicities and resolved spontaneously. Conclusions:<br />

Two important conclusions came out <strong>of</strong> this trial: 1) no difference was<br />

found in clinical response between the three arms; and 2) although patients<br />

with CRC in the subgroup analysis progressed earlier compared to historical<br />

control (3.5 vs. 7.3 months), the time from progression to death “post<br />

progression survival, PPS” was notably longer (10.3 vs. 5.6 months). This<br />

provides further evidence to the emerging understanding <strong>of</strong> the difference<br />

in clinical trial monitoring between immunotherapy and other types <strong>of</strong><br />

therapy. Accordingly, removing patients with standard CTEP definition <strong>of</strong><br />

progressive disease may adversely affect the clinical outcome <strong>of</strong> vaccine<br />

therapy. We believe that future clinical trial design strategies in cancer<br />

vaccine should be modified to allow patients to continue therapy despite <strong>of</strong><br />

disease progression.<br />

2579 General Poster Session (Board #6D), Mon, 8:00 AM-12:00 PM<br />

Targeting hyaluronan (HA) in tumor stroma: A phase I study to evaluate the<br />

safety, pharmacokinetics (PK), and pharmacodynamics (PD) <strong>of</strong> pegylated<br />

hyaluronidase (PEGPH20) in patients with solid tumors. Presenting Author:<br />

Mitesh J. Borad, Mayo Clinic, Scottsdale, AZ<br />

Background: HA, a glycosaminoglycan, accumulates in ~25% <strong>of</strong> solid<br />

tumors and is associated with poor prognosis. Tumors that accumulate HA<br />

develop high interstitial fluid pressure (IFP) and become resistant to<br />

chemotherapy. In animal models, PEGPH20 reduced tumor-associated HA<br />

and increased efficacy <strong>of</strong> chemotherapy. Methods: This ongoing phase 1<br />

study was designed to assess safety, tolerability, PK, PD, and the maximum<br />

tolerated dose (MTD) <strong>of</strong> PEGPH20 as a single agent in patients with<br />

treatment-refractory solid tumors. IV PEGPH20 was administered in a<br />

twice weekly or a once weekly dosing schedule with oral dexamethasone<br />

(dex) pre-and post dose. Biomarkers included tumor histochemistry to<br />

detect HA depletion, plasma HA catabolites, and DCE-MRI or FDG-PET.<br />

Results: Fifteen patients have been enrolled into the study. Dose administration<br />

ranged from 0.5 �g/kg to 5 �g/kg. Musculoskeletal events (MSEs) were<br />

the most common and dose limiting toxicities. To date, up to 3 �g/kg twice<br />

weekly IV infusions <strong>of</strong> PEGPH20 were well tolerated with manageable<br />

Grade 1 MSEs in most patients. MTD has not been reached. Plasma<br />

concentrations <strong>of</strong> PEGPH20 showed dose proportional exposure with a<br />

terminal half life <strong>of</strong> ~2 days. Enzymatic activity <strong>of</strong> PEGPH20 was reflected<br />

by dose-dependent HA catabolites in plasma. Pre- and post-dose tumor<br />

biopsies from 2 colon patients demonstrated 23-60% reduction in tumor<br />

HA after 4 weeks <strong>of</strong> dosing. Serial DCE-MRI and FDG-PET demonstrated<br />

increased tumor perfusion (Ktrans ) and decreased metabolic activity 8 and<br />

24hrs post-PEGPH20 in one patient each. Doses <strong>of</strong> �3 �g/kg were<br />

tolerated with intermittent Grade 1-2 MSE. Enrollment at dose levels � 3<br />

�g/kg is ongoing. Conclusions: Repeated IV PEGPH20 infusion resulted in<br />

dose-dependent systemic exposure and is tolerated with concomitant dex<br />

treatment. HA catabolites in plasma demonstrated the in vivo enzymatic<br />

activity <strong>of</strong> PEGPH20. Reduced HA in tumor biopsies and serial DCE-MRI<br />

and FDG-PET images confirmed PD activity. Increased tumor perfusion by<br />

PEGPH20 may enhance drug delivery to tumor. Combination studies <strong>of</strong><br />

PEGPH20 with cytotoxic chemotherapy are planned.<br />

161s<br />

2578 General Poster Session (Board #6C), Mon, 8:00 AM-12:00 PM<br />

An alternative clinical trial design for early cancer vaccine development to<br />

phase 3�3 design. Presenting Author: Emily Gammoh, National Institutes<br />

<strong>of</strong> Health, Bethesda, MD<br />

Background: Traditional phase I, “3�3 dose escalation” design, is conducted<br />

to identify the MTD and in some cases the optimal biologic dose.<br />

Given their unique mechanism <strong>of</strong> action and the pr<strong>of</strong>ile <strong>of</strong> their clinical<br />

outcome, this design may not apply to cancer vaccines. The therapeutic<br />

cancer vaccine FDA guidance calls for an alternative early development<br />

design. Nevertheless, whether an alternative design should be based on<br />

“dose escalation” is still an opened question. Methods: We analyzed the<br />

toxicity pr<strong>of</strong>ile in 241 phase 1, 1/2 and pilot therapeutic cancer vaccine<br />

trials conducted between 1990 and 2011. Results: Sixty-two grade 3/4<br />

vaccine related systemic toxicities were reported in 4952 treated patients<br />

(1.25 events/100 patients). Interestingly, only 2 out <strong>of</strong> 127 trials that used<br />

dose escalation reported vaccine related DLTs, both trials used bacterial<br />

vectors. Furthermore, correlation <strong>of</strong> immunological response with dose<br />

level showed no consistent trend. Conclusions: Our analysis suggests that in<br />

cancer vaccines neither toxicity nor cellular immune response correlates<br />

with dose levels. Accordingly, dose escalation is not suitable for most<br />

cancer vaccine studies. Here, we propose a two-step alternative design for<br />

early development <strong>of</strong> cancer vaccines. The first step is to determine and<br />

confirm the minimum Immune-Active Dose (IAD). If a vaccine class has<br />

been used in humans, IAD dose is chosen based on previous experience if<br />

the class is non-toxic (eg. Peptide), otherwise, a traditional dose escalation<br />

will be used. For a vaccine class that has not been tested or has<br />

undetermined toxicity we recommend “One Patient Escalation Design”<br />

(OPSD): one patient is treated per tested dose until an immune response is<br />

induced. To confirm this activity, an expanded cohort <strong>of</strong> 7 patients will be<br />

tested until demonstrating an additional response. This will then be used in<br />

phase II combination therapy trial. Alternatively, IAD can be directly tested<br />

in combination with an immune modulator in a phase II clinical trial using a<br />

two-stage design. The first stage <strong>of</strong> the phase 2 trial can be set at 4-5<br />

patients for a target response rate <strong>of</strong> over 50%. If no response is seen, then<br />

the immune modulator will be escalated in the second stage.<br />

2580 General Poster Session (Board #6E), Mon, 8:00 AM-12:00 PM<br />

Phase I safety, pharmacokinetic (PK), and pharmacodynamic (PD) trial <strong>of</strong><br />

NGR-hTNF given at high doses in patients with refractory solid tumors.<br />

Presenting Author: Paolo Andrea Zucali, Department <strong>of</strong> Oncology, Humanitas<br />

Cancer Center, Rozzano, Italy<br />

Background: NGR-hTNF exploits the asparagine-glycine-arginine (NGR)<br />

peptide for selectively targeting tumor necrosis factor (TNF) to a CD13<br />

overexpressed by tumor vasculature. Maximum tolerated dose (MTD) <strong>of</strong><br />

NGR-hTNF was previously established at 45 �g/m2 , when given as 1-h<br />

infusion every 3 weeks (q3w), with dose limiting toxicities (DLT) being<br />

grade 3 infusion-related reactions (IRRs). We explored further dose<br />

escalation by prolonging infusion time (2-h) and using premedication<br />

(paracetamol). Methods: DLTs were defined as drug-related grade 3/4<br />

adverse events (AEs). PK and soluble TNF receptors (sR1-sR2) were tested<br />

in 46 pts. The volume transfer coefficient (Ktrans ) and initial area under<br />

gadolinium concentration (IAUGC) were assessed before and 2 hours after<br />

dosing by dynamic contrast-enhanced magnetic resonance imaging (DCE-<br />

MRI) in 37 pts. Results: 12 dose levels (DLs) from 60 to 325 �g/m² q3w<br />

were evaluated. 48 pts (PS 0/1: 21/27; M/F: 37/11; median age: 61 years)<br />

received a total <strong>of</strong> 117 cycles (range 1-6). Prior regimens ranged from 1 to<br />

7 (median 3). No DLT occurred and MTD was not reached. Study-emergent<br />

grade 3 and 4 AEs were reported in 12 (25%) and 5 (10%) pts,<br />

respectively. Grade 1/2 IRRs included chills (58%) and pyrexia (56%).<br />

Both Cmax (p�.0001) and AUC (p�.0001) increased with dose. Posttreatment<br />

peaks <strong>of</strong> sR2 were higher than sR1 (9.6 v 4.9 ng/mL; p�.0001).<br />

However, changes in sRs did not differ across DLs, with a plateau in<br />

shedding kinetics. By DCE-MRI assessment, median pre- and post-first<br />

cycle values declined from 0.15 to 0.09 min-1 for Ktrans (p�.02) and from<br />

10.2 to 7.2 mM/L/sec for IAUGC (p �.0005). Over treatment, 28 pts<br />

(76%) showed decreases in IAUGC (-47%, p�.0001) and Ktrans (-59%;<br />

p�.0001) that were inversely correlated with baseline Ktrans values<br />

(p�.0001) and NGR-hTNF Cmax (p�.03). Of 41 evaluable pts, 12 (29%)<br />

had stable disease for a median time <strong>of</strong> 2.9 months. Survival at 1 year was<br />

34%, with improved survival observed in pts with lower sTNF-R2 levels<br />

(p�.01) and greater Ktrans reductions (p�.05) after 1st cycle. Conclusions:<br />

NGR-hTNF can be safely escalated at doses higher than MTD and induces<br />

low shedding <strong>of</strong> receptors and early antivascular effects.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


162s Developmental Therapeutics—<strong>Clinical</strong> Pharmacology and Immunotherapy<br />

2581 General Poster Session (Board #6F), Mon, 8:00 AM-12:00 PM<br />

A phase II, single arm clinical trial involving an alternative cancer treatment<br />

psorinum therapy in patients with unresectable metastatic colorectal<br />

carcinoma. Presenting Author: Aradeep Chatterjee, Critical Cancer Management<br />

Research Centre and Clinic, Kolkata, India<br />

Background: We prospectively studied the clinical efficacy <strong>of</strong> an alternative<br />

cancer treatment “psorinum therapy” in treating unresectable metastatic<br />

colorectal carcinoma (UMCRC). Methods: Our study was phase-II, open<br />

level, single arm and single stage. <strong>Part</strong>icipants eligibility criteria included<br />

(1) histopathology confirmation <strong>of</strong> the malignancy (2) metastatic and<br />

unresectable disease status (3) No prior conventional cancer treatments<br />

(4) Karn<strong>of</strong>sky performance status between 40 – 70%. The primary outcome<br />

measures <strong>of</strong> the study were (1) to assess the radiological tumor response<br />

rate (using CT scanning procedure and following the RECIST criteria); (2) to<br />

assess how many participants survived at least 1 yr, 2 yrs, 3 yrs, 4 yrs and<br />

finally, after 5 yrs <strong>of</strong> the study. The secondary outcome measure was to<br />

assess the side effects <strong>of</strong> the investigational anti- cancer drug (psorinum) if<br />

any. Psorinum (an alcoholic extract <strong>of</strong> scabies slough and pus cells) was<br />

administered orally at a dose <strong>of</strong> 0.02ml / kg body weight / day as a single<br />

dose on an empty stomach for a complete course duration <strong>of</strong> 2 yrs to all the<br />

participants along with allopathic and homeopathic supportive cares.<br />

Results: 105 participants included in the final analysis at the end <strong>of</strong> the<br />

study. According to the RECIST criteria, complete response occurred in 12<br />

(11.43%) cases and partial response occurred in 38 (36.19%) cases. 76<br />

(72.38%) <strong>of</strong> them survived at least 1 yr, 58 (55.24%) survived at least 2<br />

yrs, 51 (48.57%) survived at least 3 yrs, 43 (40.95%) survived at least 4<br />

yrs and 37 (35.24%) <strong>of</strong> them survived at least 5 yrs. These participants<br />

didn’t receive conventional or any other investigational cancer treatments.<br />

Conclusions: The results <strong>of</strong> the study show clinical efficacy <strong>of</strong> psorinum<br />

therapy in treating patients with UMCRC. The investigational drug psorinum<br />

is non- toxic. Randomized controlled clinical trial should be conducted<br />

for further investigation <strong>of</strong> this alternative cancer treatment in<br />

treating unresectable metastatic colorectal carcinoma to integrate it into<br />

the mainstream <strong>of</strong> oncology treatments.<br />

2583 General Poster Session (Board #6H), Mon, 8:00 AM-12:00 PM<br />

Phase I/II, two-part, open-label, multiple-dose, dose-escalation study <strong>of</strong><br />

siltuximab in patients with solid tumors. Presenting Author: Eric Angevin,<br />

Institut de Cancérologie Gustave Roussy, Villejuif, France<br />

Background: Siltuximab (S) is a chimeric mAb with high affinity for soluble<br />

IL-6. This Ph 1/2 study evaluated safety, efficacy and PK <strong>of</strong> escalating,<br />

multiple IV doses <strong>of</strong> S from a new cell line in patients (pts) with advanced<br />

refractory solid tumors. Methods: In Ph 1, S was given to 20 pts with<br />

advanced solid tumors at escalating dose levels (2.8 or 5.5 mg/kg q2w or<br />

11 or 15 mg/kg q3w) and to 24 pts with taxane and platinum resistant<br />

ovarian cancer (OVC) or KRAS mutant (KRASmt) tumors at 15 mg/kg q3w.<br />

In Ph 2, 17 OVC pts and 23 KRASmt pts received the Ph 1 selected dose <strong>of</strong><br />

15 mg/kg q3w. The Ph 2 primary efficacy endpoint was clinical benefit rate<br />

(CBR: CR � PR � SD <strong>of</strong> � 6 wk). Results: In Ph 1-2, 84 pts (35 colorectal,<br />

29 OVC, 9 pancreatic, 11 other) pretreated with a median <strong>of</strong> 4 prior tx<br />

regimens received a median <strong>of</strong> 3 (range 1, 45) cycles. 1 DLT occurred at<br />

5.5 mg/kg; MTD was not reached. 62% pts had AEs gr � 3 in Ph 1-2, most<br />

common were hepatic function abnormalities (15%), PS deterioration<br />

(12%), fatigue (11%). 10% pts had possibly S-related AEs gr � 3; only<br />

neutropenia (4%) was reported in �1 pt. 42% pts had SAEs, most were<br />

related to underlying disease, 2% were possibly S-related. 18 deaths<br />

occurred on study, 17 due to PD, 1 due to AE. No pt had antibodies to S.<br />

The PK pr<strong>of</strong>ile was bi-exponential with a t1/2 ranging 15 � 20 d. PK pr<strong>of</strong>ile<br />

<strong>of</strong> S from the new Chinese hamster ovary cell line appears similar to the<br />

previous murine Sp2/0 myeloma cell line. CBR in Ph 2 was 5%. ORR by<br />

RECIST was 26% SD in Ph 1, 9% SD in Ph 2. 1 OVC pt had CA 125<br />

response. In Ph 2 the median PFS was ~2moinboth cohorts; the median<br />

OS was 4.2 mo in KRAMSmt and 11 mo in OVC. Hb increased by a max <strong>of</strong><br />

�10 g/L in 73% pts given 15 mg/kg in Ph 2, with 94% showing a median<br />

decrease <strong>of</strong> 68% at d 8 after dose 1 in hepcidin, a regulator <strong>of</strong> iron<br />

homeostasis. CRP was suppressed in all pts during tx, with median<br />

decrease 93% at 11 mg/kg and 88 � 93% at 15 mg/kg as early as d 8 after<br />

dose 1. Other biomarkers will be presented. Conclusions: S appears to be<br />

well tolerated; doses <strong>of</strong> 11 � 15 mg/kg q3-4w can be used in future<br />

studies. No CR/PR was observed, although some pts with advanced solid<br />

tumors had durable SD and CRP suppression. Improvement in Hb was<br />

observed in a large proportion <strong>of</strong> pts, indicating a role <strong>of</strong> IL-6 in anemia that<br />

needs to be further explored.<br />

2582 General Poster Session (Board #6G), Mon, 8:00 AM-12:00 PM<br />

Randomized phase II trial <strong>of</strong> multipeptide vaccination with or without a<br />

single pre-vaccine dose <strong>of</strong> denileukin diftitox in advanced melanoma.<br />

Presenting Author: Thomas Gajewski, The University <strong>of</strong> Chicago, Chicago,<br />

IL<br />

Background: The efficacy <strong>of</strong> immunotherapy approaches such as vaccines<br />

in melanoma appears limited, in part, due to immune suppressive mechanisms<br />

in the tumor microenvironment. One <strong>of</strong> these mechanisms is the<br />

presence <strong>of</strong> CD4�CD25�FoxP3� regulatory T cells (Tregs). It has been<br />

hypothesized that strategies to reduce Treg numbers should improve the<br />

efficacy <strong>of</strong> melanoma vaccines. Methods: The study design was to randomize<br />

24 HLA-A2� patients to receive vaccine alone or denileukin diftitox (18<br />

mcg/kg i.v.) as a single dose 4 days prior to the first vaccination. The<br />

vaccine formulation consisted <strong>of</strong> 4 melanoma antigen peptides (derived<br />

from MelanA, gp100, MAGE3, and NA17A) emulsified in Montanide and<br />

GM-CSF, administered i.d./s.c. every 2 weeks. The primary endpoints were<br />

assessment <strong>of</strong> depletion <strong>of</strong> Tregs from the peripheral blood, and measurement<br />

<strong>of</strong> antigen-specific CD8� T cell responses by ELISPOT. Secondary<br />

endpoints included clinical response and analyses <strong>of</strong> the tumor microenvironment<br />

for changes in Treg infiltration. Results: The treatment was well<br />

tolerated. Enrollment was halted at 16 patients when it was clear that<br />

immune response endpoints would not be reached. There was no significant<br />

decrease in Treg numbers, nor was there an increase in vaccineinduced<br />

T cell responses, when patients received denileukin diftitox prior<br />

to vaccination. In patients with biopsiable tumors, there was no decrease in<br />

Tregs in metastatic lesions post-versus-pre-treatment. Of the 9 patients<br />

who showed clinical benefit (1 PR, 8 SD), 4 <strong>of</strong> them received denileukin<br />

diftitox and 5 did not. Estimated time to progression was 146 days in the<br />

group that received denileukin diftitox and 131 days in the group that did<br />

not. Conclusions: Denileukin diftitox given as a single dose prior to<br />

vaccination was not sufficient to deplete Tregs or improve the potency <strong>of</strong><br />

this melanoma vaccine. Alternative strategies to reduce Tregs, such as<br />

multiple doses <strong>of</strong> denileukin diftitox or anti-CD25 mAbs, should be<br />

considered.<br />

2584 General Poster Session (Board #7A), Mon, 8:00 AM-12:00 PM<br />

Association <strong>of</strong> humoral antitumor response with clinical benefit in catumaxomab-treated<br />

malignant ascites patients: Results from a phase IIIb study.<br />

Presenting Author: Peter Ruf, TRION Research GmbH, Martinsried, Germany<br />

Background: The bispecific antibody catumaxomab (anti-EpCAM x anti-<br />

CD3) which is approved for the treatment <strong>of</strong> malignant ascites (MA)<br />

patients (pts) in the EU elicits a humoral immune response against<br />

tumor-associated antigens. Here we present follow up data analyzing the<br />

influence <strong>of</strong> this antibody response on puncture free (PFS) and overall<br />

survival (OS). Methods: In the course <strong>of</strong> the phase IIIb study CASIMAS, MA<br />

pts with EpCAM positive tumors were treated with/without prednisolone<br />

premedication by 4 infusions <strong>of</strong> 10, 20, 50 and 150 �g <strong>of</strong> catumaxomab<br />

over 11 days (d). EpCAM and HER2-specific antibody responses in plasma<br />

samples <strong>of</strong> 23 pts were measured by ELISA. Additionally, samples <strong>of</strong> 5 pts<br />

receiving a second treatment cycle were analyzed. Pts with significant<br />

increase (� 2) or de novo development <strong>of</strong> anti-tumor antibodies within 38 d<br />

after treatment start were defined as responders (R). Non responders (NR)<br />

showed no humoral response by d 38 or died before. PFS and OS <strong>of</strong> R and<br />

NR were compared by Kaplan Meier analysis. Results: 11 <strong>of</strong> 23 pts (48%)<br />

showed an anti-EpCAM response and 14 <strong>of</strong> 23 pts (61%) developed<br />

HER2-specific antibodies. 6 pts (26%) reacted positive against both<br />

antigens, 4 pts (17%) had neither response. In contrast to the EpCAM R<br />

group where 73% <strong>of</strong> pts displayed detectable antibodies prior to therapy,<br />

antibodies against HER2 developed de novo. Of note, 4 <strong>of</strong> 5 individuals<br />

who received a second treatment cycle after recurring MA demonstrated an<br />

anti-HER2 Ig booster reaction which was stronger and faster in comparison<br />

to pts who received only one treatment cycle (median values at d 18: 65 vs.<br />

11 ng/ml). Most important, there was an almost 6 fold increase in median<br />

PFS between the HER2 R and NR group (68 vs 12 d, p � 0.044 (log-rank).<br />

There was also a trend towards improved OS for pts showing either<br />

anti-HER2 and/or anti-EpCAM responses (n�19) in comparison to pts with<br />

no response at all (n�4; 143 vs. 67 d, median, p � 0.054). Conclusions:<br />

The results indicate that active anti-tumor immunization triggered by<br />

catumaxomab treatment in MA pts is associated with improved clinical<br />

outcome. Further investigations <strong>of</strong> these vaccine-like effects with higher<br />

pts numbers are warranted.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


Developmental Therapeutics—<strong>Clinical</strong> Pharmacology and Immunotherapy<br />

2585 General Poster Session (Board #7B), Mon, 8:00 AM-12:00 PM<br />

Effect <strong>of</strong> the vaccine Ad5 [E1-, E2b-]-CEA(6D) on CEA-directed CMI<br />

responses in patients with advanced CEA-expressing malignancies in a<br />

phase I/II clinical trial. Presenting Author: Michael Morse, Duke University<br />

Medical Center, Durham, NC<br />

Background: Adenovirus (Ad) vectors are promising platforms for use as<br />

cancer vaccines because <strong>of</strong> their substantial immunogenicity; however,<br />

Ad-specific neutralizing antibodies, present due to natural infection, limit<br />

their use. We created a new vector containing additional deletions <strong>of</strong> the<br />

E2b gene and observed that this Ad5 [E1-, E2b-] vector, engineered to<br />

express carcinoembryonic antigen (CEA(6D)), could induce antitumor<br />

immune responses in mice despite pre-existing Ad5 immunity. The<br />

purpose <strong>of</strong> the current translational study was to evaluate the immunologic<br />

effect <strong>of</strong> increasing doses <strong>of</strong> Ad5 [E1-, E2b-]-CEA(6D) in mice and then to<br />

extended these pre-clinical observations into a phase I/II study. Methods:<br />

Ad5 naïve female C57Bl/6 mice, 4-6 weeks old, were immunized subcutaneously<br />

3 times at 2 week intervals with doses <strong>of</strong> 1.4X107 viral particles<br />

(vp) up to1.4X109 vp <strong>of</strong> Ad5 [E1-, E2b-]-CEA(6D). Two weeks after the last<br />

immunization, splenocytes were harvested for immune determinations.<br />

Cohorts <strong>of</strong> patients (n�25 total) with advanced colorectal cancer, refractory<br />

to prior therapies, received escalating doses <strong>of</strong> Ad5 [E1-, E2B-]-<br />

CEA(6D) (109 to 1011 vp) subcutaneously every 3 weeks for 3<br />

immunizations.CEA-specific cell mediated immunity was measured by<br />

ELISPOT. Results: Increasing doses <strong>of</strong> Ad5 [E1-, E2b-]-CEA(6D) in mice<br />

induced increasing CEA-specifc CMI responses. Similarly, patients who<br />

received the highest dose <strong>of</strong> Ad5 [E1-, E2b-]-CEA(6D) exhibited the<br />

highest levels <strong>of</strong> CEA-specific CMI responses. The induction <strong>of</strong> CEAspecific<br />

CMI responses increased over the course <strong>of</strong> the 3 injections despite<br />

the presence <strong>of</strong> pre-existing Ad5 immunity in the majority (75%) <strong>of</strong><br />

patients. There were no drug related grade 3/4 toxicities.Two patients with<br />

stable disease remained so during the study. All other patients experienced<br />

progressive disease; however, 1-year survival was 54%. Conclusions:<br />

Multiple homologous doses <strong>of</strong> Ad5 [E1-, E2b-]-CEA(6D) could break<br />

tolerance and generate CEA-specific CMI responses in the setting <strong>of</strong> Ad<br />

specific immunity in colorectal cancer patients.<br />

2587 General Poster Session (Board #7D), Mon, 8:00 AM-12:00 PM<br />

Development <strong>of</strong> an assay based on SOMAmer affinity reagents that detects<br />

drug-unbound serum EGFR in the presence <strong>of</strong> cetuximab and panitumumab.<br />

Presenting Author: Noh Jin Park, Quest Diagnostics, San Juan<br />

Capistrano, CA<br />

Background: EGFR is an oncogene product whose extracellular domain<br />

(ECD) can be either up-regulated or down-regulated in serum <strong>of</strong> patients<br />

with various cancers. In colon cancer patients with positive tissue EGFR<br />

expression, 2 EGFR ECD-targeting monoclonal antibody drugs, cetuximab<br />

(Erbitux) and panitumumab (Vectibix), are widely used chemotherapeutic<br />

agents. Patient responses to these drugs vary, and the mechanism <strong>of</strong> this<br />

variability remains unelucidated. Methods: A Slow Off-Rate Modified<br />

Aptamer (SOMAmer) targeting the EGFR ECD was selected via Systematic<br />

Evolution <strong>of</strong> Ligands by Exponential Enrichment (SELEX). Using this<br />

SOMAmer as a capturing reagent and based on published studies ( Gold et<br />

al. PLoS ONE; Dec 7, 2010:10.1371/journal.pone.0015004), we developed<br />

a quantitative serum EGFR assay to reliably quantify EGFR in serum.<br />

Results: Recovery tests using various amounts <strong>of</strong> purified EGFR spiked into<br />

serum demonstrated a full level <strong>of</strong> EGFR. Intra and inter assay variability<br />

were tested and showed minimum variability. The detection range is 0.95<br />

ng/ml to 600 ng/ml. Interestingly, when serum samples from patients<br />

taking cetuximab or panitumumab at the time <strong>of</strong> blood collection were<br />

tested, we observed markedly lower levels <strong>of</strong> EGFR-captured SOMAmer.<br />

ELISA assays from 2 different vendors showed normal to high levels <strong>of</strong><br />

EGFR in these samples. We further showed that pretreatment <strong>of</strong> normal<br />

serum with either cetuximab or panitumumab can dose-dependently<br />

reduce the EGFR SOMAmer signal. The data suggest that our EGFR<br />

SOMAmer assay detects serum EGFR molecules that are unbound by<br />

cetuximab or panitumumab. Some treated patient samples had more<br />

drastic reductions in circulating serum EGFR than others. Conclusions:<br />

Various assay development parameters such as accuracy, detection range,<br />

and intra and inter assay variability showed that a SOMAmer-based assay<br />

detecting serum EGFR can be used in a clinical setting. Our data suggest<br />

that this assay can accurately measure drug-unbound EGFR in patients,<br />

which may serve as a surrogate drug efficacy indicator, and this may help<br />

physicians to adjust the drug dosage. Further studies will be necessary to<br />

investigate this possibility.<br />

163s<br />

2586 General Poster Session (Board #7C), Mon, 8:00 AM-12:00 PM<br />

Effect <strong>of</strong> modulation <strong>of</strong> the hostile tumor microenvironment through<br />

adoptive transfer <strong>of</strong> IL-12 expressing MUC-16 targeted T cells on ovarian<br />

tumors in vivo. Presenting Author: Alena A. Chekmasova, Memorial<br />

Sloan-Kettering Cancer Center, New York, NY<br />

Background: T cells may be genetically modified to recognize tumor<br />

associated antigens (TAAs) through the introduction <strong>of</strong> genes encoding<br />

artificial T cell receptors termed chimeric antigen receptors (CARs).<br />

MUC16 (CA125) is an antigen over-expressed on ovarian carcinomas and a<br />

serum marker for the diagnosis <strong>of</strong> ovarian cancer. We have previously<br />

demonstrated enhanced antitumor efficacy <strong>of</strong> CAR � T cells further modified<br />

to secrete IL-12. We therefore tested whether MUC-16 targeted T cells<br />

further modified to express IL-12 would exhibit an enhanced antitumor<br />

efficacy in a syngeneic immunocompetent tumor model <strong>of</strong> ovarian cancer.<br />

Methods: We have constructed SFG retroviral vectors encoding the second<br />

(4H11m28mz) generation CARs as well as IL-12 modified CAR<br />

(4H11m28mz/mIL12) targeted to the retained extra-cellular domain <strong>of</strong><br />

MUC16, termed MUC-CD. We demonstrated an antitumor efficacy <strong>of</strong> these<br />

T cells in a syngeneic tumor model using the C57BL6 (B6) mice<br />

intraperitoneally (i.p.) injected with ID8(MUC-CD) tumor cells. Results: In<br />

our studies treatment <strong>of</strong> mice bearing established ID8(MUC-CD) ovarian<br />

tumor with MUC-CD specific T cells expressing IL-12 gene, in contrast to T<br />

cells targeted to MUC-CD alone, fully eradicate highly advanced intraperitoneal<br />

ovarian tumors. Significantly, we found that mice treated with<br />

4H11m28mz/mIL12 T cells had increased number <strong>of</strong> modified T cells in<br />

the peritoneum at day 4 and 7 with increased recruitment <strong>of</strong> endogenous T<br />

cells to the site <strong>of</strong> the tumor when compared to controls and mice treated<br />

with 4H11m28mz T cells. The observed antitumor effect did not required<br />

prior lymphodepletion and was well tolerated in treated mice. Conclusions:<br />

CAR modified T cells targeted to the MUC-16 antigen efficiently eradicate<br />

orthotopic ovarian cancer in syngeneic immunocompetent mice with<br />

markedly enhanced antitumor efficacy seen in those mice treated with<br />

CAR � T cells further modified to secrete IL-12. These data support future<br />

clinical trials utilizing adoptive T cell therapy in patients with relapsed<br />

ovarian cancer.<br />

2588 General Poster Session (Board #7E), Mon, 8:00 AM-12:00 PM<br />

Effect <strong>of</strong> the novel therapeutic cancer vaccine formulation DPX-0907 on<br />

multifunctional T-cell responses in ovarian, breast, and prostate cancer<br />

patients. Presenting Author: Neil Lorne Berinstein, Odette Cancer Centre,<br />

Toronto, ON, Canada<br />

Background: To increase the efficacy <strong>of</strong> peptide cancer vaccines, we<br />

developed a novel vaccine platform called DepoVax, an adjuvanted waterfree<br />

depot formulation with the ability to generate enhanced immune<br />

responses. Naturally processed HLA-A2 restricted peptides that are selectively<br />

presented by breast, ovarian and prostate cancer cell lines, but not by<br />

normal HLA-A2� cells, were used as antigens with a proprietary adjuvant<br />

and a T helper peptide epitope to create a therapeutic cancer vaccine,<br />

DPX-0907. Methods: A phase I clinical study was designed to examine the<br />

safety and immune activating potential <strong>of</strong> DPX-0907 in advanced stage<br />

breast, ovarian and prostate cancer patients. A total <strong>of</strong> 23 late stage cancer<br />

patients were recruited and were divided into two dose volume cohorts in a<br />

three immunization clinical protocol. Results: DPX-0907 proved to be safe<br />

with no serious adverse effects related to the vaccine reported. Of those<br />

evaluable for immunogenicity, all breast cancer patients (3/3), most <strong>of</strong><br />

ovarian (5/6) and one third <strong>of</strong> prostate (3/9) cancer patients demonstrated<br />

immune response to one or more <strong>of</strong> seven antigenic peptides, resulting in a<br />

61% response rate. Immune responses correlated with achievement <strong>of</strong> CR,<br />

PR or SD to last treatment. No difference in immune response rate or<br />

magnitude was seen between the two dose groups. DPX-0907 displayed<br />

strong immune induction potential, with 73% <strong>of</strong> immune responders<br />

showing responses with just one dose <strong>of</strong> vaccine. In 83% <strong>of</strong> responders,<br />

immune responses were detected at �2 time points post vaccination and<br />

64% had a persistent immune response at one month post last vaccination.<br />

Immune monitoring showed peptide-specific CD8 T cells, and these T cells<br />

were able to secrete multiple Th1 cytokines indicating their multifunctionality,<br />

a feature attributable to cells that mediate protective responses.<br />

Conclusions: These data support the ability <strong>of</strong> DPX-0907 to elicit Th1<br />

dominated, specific immunity and support the rationale for further testing<br />

in immunologically competent cancer patients. The novel DepoVax formulation<br />

may promote multifunctional memory responses with peptides from<br />

other tumor associated antigens.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


164s Developmental Therapeutics—<strong>Clinical</strong> Pharmacology and Immunotherapy<br />

2589 General Poster Session (Board #7F), Mon, 8:00 AM-12:00 PM<br />

Phase I/II study <strong>of</strong> resiquimod as an immunologic adjuvant for NY-ESO-1<br />

protein vaccination in patients with melanoma. Presenting Author: Rachel<br />

Lubong Sabado, New York University Cancer Institute, New York, NY<br />

Background: The TLR 7/8 agonist, Resiquimod has been shown to induce<br />

local activation <strong>of</strong> immune cells, production <strong>of</strong> cytokines, and antigenpresentation<br />

by dendritic cells, features desirable for cancer vaccine<br />

adjuvants. In this study, we evaluated the safety and immunogenicity <strong>of</strong><br />

vaccination with NY-ESO-1 protein emulsified in Montanide ISA-51 VG<br />

when given with or without Resiquimod in patients with surgically resected<br />

stage IIB-IV melanoma patients. Methods: This is a two-part study design.<br />

<strong>Part</strong> I represents an open-label dose-escalation with Resiquimod using 2<br />

cohorts treated with 100ug NY-ESO-1 protein emulsified in 1.25mL<br />

Montanide (day1) followed by topical application <strong>of</strong> 1000mg <strong>of</strong> the 0.2%<br />

Resiquimod gel on days 1 and 3 for cohort-1 (N�3) or days 1, 3, and 5 for<br />

cohort-2 (N�3). The cycles were repeated every 3 weeks, total <strong>of</strong> 4 cycles.<br />

<strong>Part</strong> II <strong>of</strong> the study is blinded. Patients were randomized to receive 100ug<br />

NY-ESO-1 protein emulsified in 1.25mL Montanide (day1) followed by<br />

topical application <strong>of</strong> placebo gel (Arm-A; N�8) or 1000mg <strong>of</strong> 0.2%<br />

Resiquimod gel (Arm-B; N�12) using the dosing regimen established in<br />

<strong>Part</strong> I. Blood samples were collected at baseline, one week after each cycle<br />

<strong>of</strong> vaccination, and at follow-up visit for the assessment <strong>of</strong> NY-ESO-1specific<br />

humoral and cellular immune responses. Results: Enrollment has<br />

been completed. 25/26 patients received all 4 vaccinations. The treatment<br />

was generally well-tolerated, with no grade 4 adverse events or studyrelated<br />

deaths. The most common toxicities were mild to moderate and<br />

included local injection site reactions (granuloma, pruritus, induration) and<br />

systemic flu-like symptoms. One patient experienced a grade 3 syncopal<br />

episode that was deemed unrelated to the drug. Another patient experienced<br />

a grade 3 injection site necrosis that was possibly related to the study<br />

drug and was removed from the study prior to receiving the 4th vaccine.<br />

Conclusions: This study demonstrates the safety <strong>of</strong> Resiquimod as an<br />

adjuvant for NY-ESO-1 protein vaccination. The study will remain blinded<br />

until all immune monitoring assays have been completed. An updated<br />

abstract will be submitted once the study is unblinded.<br />

2591 General Poster Session (Board #7H), Mon, 8:00 AM-12:00 PM<br />

Cellular function <strong>of</strong> the mononuclear phagocyte system (MPS) as a<br />

phenotypic probe for pharmacokinetics (PK) and pharmacodynamics (PD)<br />

<strong>of</strong> PEGylated liposomal doxorubicin (PLD) in patients with recurrent<br />

ovarian cancer. Presenting Author: Whitney Paige Caron, University <strong>of</strong><br />

North Carolina at Chapel Hill, Chapel Hill, NC<br />

Background: A phenotypic probe is a test that can be administered to a<br />

patient as a potential indicator <strong>of</strong> a drug’s PK/PD which can then be used to<br />

individualize therapy. Since nanoparticles are cleared via the MPS,<br />

including blood monocytes (MO), we hypothesize that circulating MO could<br />

potentially play a major role in, and be a surrogate marker <strong>of</strong> nanoparticle<br />

clearance (CL). Furthermore, we postulate that the ability to measure MO<br />

functional activity in blood samples can be used to predict CL and<br />

subsequently PD endpoints such as progression free survival (PFS) and<br />

hand-foot syndrome (HFS), in patients (pt). Methods: PLD 30/40 mg/m2 IV<br />

x 1 with or without carboplatin (AUC � 5 mg/mL/min) was administered to<br />

pts (n�10) with recurrent ovarian cancer. Serial PK samples were obtained<br />

at time 0 to day 28 post dose. Plasma was processed to measure<br />

encapsulated and released doxorubicin using solid phase separation and<br />

HPLC. Data was analyzed with WinNonlin to obtain PK parameters. MO and<br />

granulocyte phagocytic function (uptake <strong>of</strong> FITC-labeled opsonized E. Coli)<br />

and oxidative burst activity (intracellular oxidation <strong>of</strong> dihydrorhodamine<br />

123) were assessed via flow cytometry at 0, 48, 96 h, and on day 28. For<br />

both assays, changes in mean fluorescence intensity <strong>of</strong> the gated MO<br />

population, following incubation <strong>of</strong> whole blood with the appropriate<br />

substrate, served as an index <strong>of</strong> activity. Pts were followed from the first<br />

dose <strong>of</strong> PLD until time <strong>of</strong> disease progression. Results: There was a direct<br />

linear relationship between encapsulated doxorubicin CL and both phagocytosis<br />

(R2 � 0.87) and oxidative burst activity (R2 � 0.64) in blood MO.<br />

Similarly, phagocytosis (R2 � 0.77) and oxidative burst probes correlated<br />

with PFS (R2 � 0.67) in the 4 pts who progressed while on PLD. Oxidative<br />

burst also correlated with degree <strong>of</strong> HFS (R2 � 0.56). There was no<br />

relationship between phagocytosis and oxidative burst in granulocytes and<br />

PK/PD <strong>of</strong> PLD. Conclusions: These findings indicate that probes <strong>of</strong> MPS<br />

function predict PLD CL, HFS and PFS and thus may be useful for<br />

individualizing PLD therapy in ovarian cancer and other malignancies.<br />

2590 General Poster Session (Board #7G), Mon, 8:00 AM-12:00 PM<br />

A phase I study <strong>of</strong> lapatinib (LPT) and cetuximab (CTX) in patients with<br />

CTX-sensitive solid tumors. Presenting Author: John F. Deeken, Lombardi<br />

Comprehensive Cancer Center, Georgetown University, Washington, DC<br />

Background: Preclinical research has shown that one mechanism <strong>of</strong><br />

acquired resistance to CTX is via EGFR-ErbB2 heterodimerization, which<br />

can reactivate oncogenic pathways. LPT is a dual EGFR and ErbB2<br />

intracellular tyrosine kinase inhibitor. A phase I translational clinical study<br />

was performed to determine the maximum tolerated dose (MTD), dose<br />

limiting toxicities (DLTs), and clinical activity <strong>of</strong> CTX and LPT in patients<br />

with EGFR-driven solid tumor malignancies that can be treated with CTX.<br />

Methods: Patients (Pts) were enrolled in a 3�3 dose escalation trial. Prior<br />

CTX therapy was allowed. CTX was given at 400mg/m2 on Cycle 1, Day 1<br />

(C1D1), then 250 mg/m2 weekly. LPT dose levels (DL) were (1) 750mg, (2)<br />

1000mg, and (3) 1250mg orally daily. Rash management included daily<br />

sunblock, steroid cream, and doxycycline. Cycle length was 21 days, and<br />

patients were assessed for toxicity through the end <strong>of</strong> C2, and for efficacy<br />

after every 2 cycles. Fresh tumor biopsies were obtained at baseline and at<br />

the end <strong>of</strong> C1 to compare EGFR and ErbB2 expression levels and EGFR<br />

related pathway activation. DNA from blood samples was analyzed for<br />

pharmacogenetic (PGx) variations and correlations with toxicity and pharmacokinetics<br />

(PK). Results: Between October, 2010 to January 2012, 13<br />

pts were enrolled, with colon (4), lung (3), head and neck (3), and anal<br />

cancers (3); 10 were evaluable for toxicity. Treatment-related toxicities <strong>of</strong><br />

any grade included: rash (67%), diarrhea (42%), fatigue (33%), nausea/<br />

vomiting (17%), and dehydration (8%). DLTs included G3 rash in 1 <strong>of</strong> 6 pts<br />

on DL1, and G3 diarrhea despite optimal therapy in 1 <strong>of</strong> 4 pts on DL2.<br />

Enrollment to DL2 continues. Of 7 pts evaluable for response, 1 had an<br />

uPR, 3 had SD, including 1 with SD <strong>of</strong> �4 cycles, and 3 had DP. Both<br />

patients with uPR and prolonged SD were treated on DL1. Tumor<br />

EGFR-ErbB2 and EGFR pathway phosphorylation analyses and PGx results<br />

will be presented. Conclusions: The combination <strong>of</strong> CTX and LPT is well<br />

tolerated with expected toxicities. Efficacy was seen even on DL 1. Phase II<br />

clinical studies in CTX-sensitive diseases such as colon and head and neck<br />

cancer are planned.<br />

2592 General Poster Session (Board #8A), Mon, 8:00 AM-12:00 PM<br />

Phase I study <strong>of</strong> docetaxel injection concentrate for nano-dispersion<br />

(DICN), a novel polysorbate 80-free formulation <strong>of</strong> docetaxel. Presenting<br />

Author: Minish Mahendra Jain, KEM Hospital and Research Center, Pune,<br />

India<br />

Background: A polysorbate 80-free formulation <strong>of</strong> docetaxel may preclude<br />

the need for dexamethasone pre-medication and may also reduce toxic<br />

effects associated with polysorbate 80. DICN is a novel polysorbate 80-free<br />

formulation <strong>of</strong> docetaxel stabilized with lipid and polymer using NanotectonTM<br />

technology. We studied safety, tolerability, and the pharmacokinetics<br />

(PK) <strong>of</strong> DICN in patients with advanced solid malignancies. Methods: Entry<br />

criteria included: age 18-65 years, histologically/cytologically confirmed<br />

advanced malignancy, performance status � ECOG 2, estimated survival �<br />

12 weeks, and adequate organ function. A standard phase I 3�3 dose<br />

escalation schema was employed with an increase <strong>of</strong> 12.5 to 50% over<br />

previous DICN dose level as per safety pr<strong>of</strong>ile. The infusion was 60 min for<br />

1 cycle and major objectives were to determine maximum tolerated dose<br />

(MTD), and PK and safety pr<strong>of</strong>iles. Premedication to prevent hypersensitivity<br />

was not administered to patients receiving DICN. Three patients were<br />

treated with docetaxel 75 mg/m2 to gather PK data. Plasma was analyzed<br />

for docetaxel level using a validated assay. Results: Twenty-seven patients<br />

treated with DICN had a mean age <strong>of</strong> 48.8 yrs (range 29-65); 21 were<br />

females; and entered with metastatic breast cancer (MBC; n�14), nonsmall<br />

cell lung carcinoma (NSCLC; n�6), ovarian (n�2), and other (n�5).<br />

Doses (mg/m2 ) studied were 60 (n�7), 75 (n�5), 100 (n�3), 125 (n�3),<br />

150 (n�6), and 170 (n�3). Despite lack <strong>of</strong> dexamethasone premedication,<br />

no patient receiving DICN reported a hypersensitivity reaction. Two<br />

DLTs (febrile neutropenia) were reported at DICN 170 mg/m2 . DICN PK<br />

(AUC AUC 0-24, 0-�, and Cmax) increased in a dose proportionate manner from<br />

60 to 170 mg/m2 . Compared with docetaxel 75 mg/m2 ,Cmax and AUC0-24 <strong>of</strong> DICN 75 mg/m2 was 1.4 and 1.2 times higher, respectively, and 1.9 and<br />

1.8 times higher, respectively for DICN 150 mg/m2 . The median Tmax <strong>of</strong><br />

DICN 75 mg/m2 and docetaxel 75 mg/m2 were 1.00 and 0.517 hours,<br />

respectively. Conclusions: In this study,DICN demonstrated acceptable<br />

tolerability and a favorable PK pr<strong>of</strong>ile. A 150 mg/m2 is the recommended<br />

phase II dose for DICN.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


Developmental Therapeutics—<strong>Clinical</strong> Pharmacology and Immunotherapy<br />

2593 General Poster Session (Board #8B), Mon, 8:00 AM-12:00 PM<br />

Phase I and pharmacokinetic (PK) study <strong>of</strong> pazopanib (P) in combination<br />

with two schedules <strong>of</strong> ifosfamide (I) in patients (pts) with advanced solid<br />

tumors (STs). Presenting Author: Paul Hamberg, Erasmus University<br />

Medical Center, Daniel den Hoed Cancer Center, Rotterdam, Netherlands<br />

Background: P is a tyrosine kinase inhibitor <strong>of</strong> PDGFR, VEGFR and KIT. This<br />

study aimed to determine the recommended phase II dose (RP2D) <strong>of</strong> P<br />

combined with I. To assess the impact <strong>of</strong> scheduling on tolerability, two I<br />

schedules were explored. Methods: Pts with progressive ST, no standard<br />

therapy available, PS 0-1 and good organ function were eligible. Pts<br />

received daily P with 3-weekly I, 9 g/m2 /cycle, either continuously (Iciv) or<br />

as 3 hrs boluses (Ibolus) for 3 days. P was escalated in serial cohorts <strong>of</strong> 3-9<br />

pts per I-schedule. If in 3-6 pts dose-limiting toxicity (DLT) <strong>of</strong> 33.3%<br />

within 1st cycle was seen 3 more pts at that dose level (DL) were enrolled.<br />

RP2D was exceeded if in � 1/3 pts, � 2/6 pts or � 3/9 pts DLT occurred. P<br />

was maximized at 1000 mg/day. At RP2D, an expansion cohort <strong>of</strong> 6-9 pts<br />

was studied to confirm safety. PK samples <strong>of</strong> P and I were collected in all<br />

pts. Sampling allowed intra-individual comparison <strong>of</strong> I-PK with or without P<br />

and vice versa. Results: 47 pts were studied. For Iciv (25 pts), RP2D was<br />

the maximum dose <strong>of</strong> P (1000 mg), higher than the single agent RP2D. At<br />

RP2D 2 DLTs in 15 pts occurred: 1 febrile neutropenia (FN); 1 encephalopathy.<br />

With Ibolus (22 pts), P 400 mg with or without GCSF was not tolerated<br />

(5 DLTs in 10 pts: 3*FN, 1 encephalopathy; 1 proteinuria). At RP2D (P<br />

200 mg � Ibolus � GCSF) 3 DLTs in 12 pts (FN; encephalopathy, renal<br />

insufficiency) were observed. Toxicity occurring in �30% <strong>of</strong> pts in Iciv (all<br />

grades/grade 3-4,%) were neutropenia (92/92) anemia (92/8); thrombocytopenia<br />

(35/12), elevated ASAT, ALAT, alkaline phosphatase (73/4; 73/0;<br />

62/2), nausea (58/0), vomiting (73/0), diarrhea (54/0) and hypertension<br />

(31/12). 4/25 pts in Iciv evaluable for response had PR (2 sarcoma, 2<br />

ovary) and 10 prolonged (�3 mths) SD. In Ibolus, 6/20 pts had PR (2<br />

urothelial, 1 sarcoma, 1 ovary, 1 CUP, 1 mesothelioma) and 3/20<br />

prolonged SD. Preliminary PK analysis showed no effect <strong>of</strong> P on I-exposure,<br />

neither <strong>of</strong> Ibolus or Iciv on P. Conclusions: P is tolerated at a higher dose<br />

combined with Iciv compared to Ibolus (1000 vs 200 mg). PK cannot<br />

explain this difference. This study underlines the impact <strong>of</strong> scheduling on<br />

the tolerability <strong>of</strong> VEGFR-TKI and cytotoxic drug combinations.<br />

2595 General Poster Session (Board #8D), Mon, 8:00 AM-12:00 PM<br />

Pharmacokinetic modeling and simulation (PK M&S) supported dose<br />

escalation <strong>of</strong> PF-03446962, a monoclonal antibody (mAb) against activin<br />

receptor like kinase 1, in patients with solid tumors. Presenting Author:<br />

Erjian Wang, Pfizer Inc., San Diego, CA<br />

Background: PF-03446962 is a first in class mAb. The PK <strong>of</strong> mAbs is<br />

complex, as their clearance (CL) can be linear or non-linear, besides<br />

extremely slow. Caution must be exercised when designing mAbs’ dose<br />

escalation schemes. Along with safety and activity evaluation, we used PK<br />

M&S in real time to ensure that subsequent recommended doses did not<br />

expose too many patients to subtherapeutic levels while preserving safety.<br />

Methods: The model incorporated both Michaelis-Menten approximation<br />

and linear components. A prespecified target exposure was defined by<br />

preclinically projected therapeutic and toxic concentrations. The PK M&S<br />

results were used to make dose escalation decisions. Results: Eight dose<br />

levels (0.5, 1, 2, 3, 4.5, 6.75, 10, and 15 mg/kg) were studied in 44<br />

patients. CL <strong>of</strong> PF-03446962 exhibited nonlinear characteristics at low<br />

doses and linear at high doses. CL was saturated at dose levels <strong>of</strong> 3 mg/kg<br />

and above. Mean half-life was 2.6 days for the first dose level and 12-15<br />

days after CL saturation. The dose ratio <strong>of</strong> 15 to 0.5 mg/kg was 30, while its<br />

corresponding Cmax and AUC ratios were 46 and 97, respectively.<br />

However, the PK M&S accurately predicted exposure following each dose.<br />

95% <strong>of</strong> the observed drug concentrations across all dose levels were within<br />

the predicted values. Concentrations at the 4.5 mg/kg dose exceeded the<br />

predicted efficacious level over the dosing period. None <strong>of</strong> 44 patients in<br />

the study were exposed to the toxic level derived from a study in monkeys.<br />

PK data also implied a biologically active dose at 2 mg/kg dose and above,<br />

confirmed by observing target toxicities (ie, telangiectasia) and clinical<br />

tumor responses. One patient with PR was at the 2 mg/kg dose and 2<br />

patients with PR were at the 10 mg/kg. The PK M&S well predicted the drug<br />

exposure following multiple doses in patients, indicating that patients did<br />

not develop antidrug immunogenicity. Conclusions: The PK M&S based<br />

approach ensured that the majority <strong>of</strong> patients were treated at or near the<br />

therapeutic dose. It can be readily implemented in the conduct <strong>of</strong> dose<br />

escalation trials <strong>of</strong> antitumor mAbs, while minimizing risk to patients and<br />

maximizing efficiency.<br />

165s<br />

2594 General Poster Session (Board #8C), Mon, 8:00 AM-12:00 PM<br />

Modeling the combined efficacy <strong>of</strong> rilotumumab (R; AMG 102) plus<br />

epirubicin/cisplatin/capecitabine (ECX) for the treatment <strong>of</strong> locally advanced<br />

or metastatic gastric or esophagogastric junction (G/EGJ) cancer.<br />

Presenting Author: Sameer Doshi, Amgen Inc., Thousand Oaks, CA<br />

Background: R is an investigational, fully human monoclonal antibody to<br />

hepatocyte growth factor/scatter factor (HGF/SF) that inhibits signaling<br />

though the MET receptor. In a double-blind, placebo-controlled, phase 2<br />

trial <strong>of</strong> 121 patients with G/EGJ cancer, R (7.5 or 15 mg/kg) � ECX showed<br />

trends for improved progression-free survival (PFS) and overall survival<br />

compared to ECX alone. The combined effects <strong>of</strong> R and ECX on tumor<br />

reduction and PFS were modeled using the phase 2 data. Methods: A<br />

population pharmacokinetic (PK) model was used to estimate R PK<br />

parameters and individual R concentrations (C) over time. A tumor dynamic<br />

model was developed to characterize the combined effects <strong>of</strong> R with ECX on<br />

tumor growth and cell death and to evaluate the impact <strong>of</strong> baseline patient<br />

characteristics and lab values on model parameters. A discrete timesurvival<br />

model was developed with PFS and C data to evaluate the effect <strong>of</strong><br />

R, ECX, and the interaction between R and ECX on PFS within a given time<br />

interval. Results: R exhibited linear PKs with an estimated mean clearance<br />

<strong>of</strong> 0.216 L/d/70 kg. The tumor dynamic model suggested that R and ECX<br />

worked jointly to reduce tumor size from baseline via inhibition <strong>of</strong> the tumor<br />

growth rate constant (Kgrowth) and stimulation <strong>of</strong> the death rate <strong>of</strong> tumor<br />

cells (Kdeath). Assuming the maximal inhibition <strong>of</strong> Kgrowth is 100%, the<br />

estimated mean C that provided 50% maximal Kgrowth inhibition (EC50) was<br />

6.71 �g/mL. The mean (SD) estimated effect <strong>of</strong> 7.5 and 15 mg/kg R on<br />

K was 90.1% (3.8%) and 95.5% (1.9%) inhibition, respectively. None<br />

growth<br />

<strong>of</strong> the tested baseline factors appeared to significantly affect tumor<br />

reduction by R � ECX. In the discrete time-survival model, the R and ECX<br />

combination significantly improved PFS, and the model suggests that the<br />

magnitude <strong>of</strong> the effect <strong>of</strong> one treatment was dependent on the other.<br />

Conclusions: R and ECX appeared to work in combination to decrease tumor<br />

size and to improve PFS.<br />

2596 General Poster Session (Board #8E), Mon, 8:00 AM-12:00 PM<br />

Individual PK-guided sunitinib dosing: A feasibility study in patients with<br />

advanced solid tumors. Presenting Author: Nienke A.G. Lankheet, Slotervaart<br />

Hospital, Amsterdam, Netherlands<br />

Background: Sunitinib treatment shows a clear dose-efficacy relationship.<br />

However, due to large inter-individual variations in plasma exposure, target<br />

total trough levels (� 50 ng/mL) are frequently not reached with the current<br />

dosing schedule. Therefore, a prospective Phase I study was performed to<br />

determine the safety and feasibility <strong>of</strong> PK-guided dosing <strong>of</strong> sunitinib.<br />

Methods: Thirty patients with solid tumors with a potential benefit from<br />

sunitinib treatment were included. Patients were treated continuously with<br />

sunitinib 37.5 mg once daily. At day 15 and 29 <strong>of</strong> sunitinib treatment,<br />

plasma trough levels <strong>of</strong> sunitinib and its active metabolite (N-desethyl<br />

sunitinib) were measured. If the total trough level (TTL) was � 50 ng/mL<br />

and the patient did not show any � grade 3 toxicity (CTCAE 4.02), the daily<br />

sunitinib dose was increased by 12.5 mg. If the patient suffered from<br />

� grade 3 toxicity, the sunitinib dose was lowered by 12.5 mg. After 8<br />

weeks a final TTL evaluation was performed. Results: All 30 patients started<br />

treatment and in 18 patients all 3 TTLs were measured at the time <strong>of</strong> this<br />

analysis. Given TTL � 50 ng/mL and � grade 3 toxicity, a first sunitinib<br />

dose increase could be applied in 10 patients and a second dose increase<br />

in 2 patients at day 15 and 29, respectively. At day 15, day 29 and 8 weeks<br />

<strong>of</strong> treatment, mean TTLs were 49.7 ng/mL (coefficient <strong>of</strong> variation (CV)<br />

27.7%), 62.4 ng/mL (CV 35.5%) and 56.7 ng/mL (CV 47.8%), respectively.<br />

At day 15, using the sunitinib standard dose <strong>of</strong> 37.5 mg, target TTLs<br />

were reached in 8 patients (44%). At 8 weeks, the mean sunitinib daily<br />

dose intensity was increased to 40.3 mg (standard deviation (SD) � 11.0).<br />

At the end <strong>of</strong> the 8 week study period, 5 out <strong>of</strong> 18 patients (28%) were still<br />

at an increased sunitinib dose level without additional side effects. In these<br />

patients the mean daily sunitinib dose was 55.0 mg (SD � 6.8) and their<br />

final TTLs were all � 50 ng/mL (mean: 70.8 ng/mL (CV 29.8%)). Using<br />

PK-guided sunitinib dosing, target TTLs were reached in 10 out <strong>of</strong> 18<br />

patients (56%) at the final TTL evaluation, compared to 44% at day 15<br />

before any dose adjustment. Conclusions: Individual PK guided dosing is<br />

feasible and enables a significant increase in sunitinib dose intensity<br />

without causing additional toxicity.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


166s Developmental Therapeutics—<strong>Clinical</strong> Pharmacology and Immunotherapy<br />

2597 General Poster Session (Board #8F), Mon, 8:00 AM-12:00 PM<br />

<strong>Clinical</strong> pharmacologic considerations for the phase II/III dose/regimen <strong>of</strong><br />

the investigational Aurora A kinase (AAK) inhibitor MLN8237 (alisertib):<br />

Pharmacokinetics (PK), pharmacodynamics (PD), and exposure-safety<br />

relationships. Presenting Author: Karthik Venkatakrishnan, <strong>Clinical</strong> Pharmacology,<br />

Millennium Pharmaceuticals, Inc., Cambridge, MA<br />

Background: MLN8237, an oral selective AAK inhibitor, is primarily<br />

metabolized by multiple glucuronidation enzymes including the polymorphic<br />

UGT1A1. Phase 1 studies included comprehensive PK and PD<br />

sampling. We report integrated PK, PD, and PK-safety analyses in support<br />

<strong>of</strong> dose/regimen selection for phase 2/3 studies. Methods: Phase 1 studies<br />

in adults with advanced cancers evaluated dosing on d 1-7 in 21-d cycles or<br />

d 1-21 in 35-d cycles. Data from 294 patients in 4 phase 1 and 2 phase 2<br />

studies contributed to population PK modeling. PD endpoints included<br />

mitotic index (MI) in skin and chromosome alignment and spindle<br />

bipolarity (CA/SB) in mitotic tumor cells. Logistic regression analyses<br />

evaluated relationships between MLN8237 PK parameters and DLTs<br />

(N�86) or CNS adverse events (AEs; n�134) in 2 phase 1 studies. Results:<br />

MLN8237 displayed dose-linear PK (5-200 mg/d), described by a 2-compartment<br />

model with first order absorption. Covariate analyses did not<br />

reveal significant effects <strong>of</strong> age, body size, sex, UGT1A1 genotype, or<br />

creatinine clearance (�30 mL/min). Exposure-related increases in skin MI<br />

and decreases in CA/SB in tumor mitotic cells confirmed AAK inhibition by<br />

MLN8237. At the MTD <strong>of</strong> 50 mg BID (d 1-7 dosing) geometric mean<br />

steady-state exposures (48,200 nM.hr) were comparable to those associated<br />

with �50% CA/SB reductions in mitotic tumor cells (57,300 nM.hr).<br />

Exposures at the 21-d MTD (QD dosing) were lower, favoring 50 mg BID (d<br />

1-7 dosing) for further development. At 50 mg BID (d 1-7 dosing) logistic<br />

regression relating MLN8237 AUC to DLT rate estimated a DLT probability<br />

<strong>of</strong> 8% (95% CI 3-20%). Similar analyses identified Cmax rather than AUC as<br />

the predictor <strong>of</strong> CNS AEs, supporting BID dosing in adults to reduce peak<br />

concentrations while preserving total systemic exposure. Conclusions:<br />

MLN8237 exhibits dose-linear PK independent <strong>of</strong> age, body size, mild or<br />

moderate renal impairment, or UGT1A1*28 polymorphism. Exposures<br />

achieved at or near 50 mg BID are expected to result in tumor AAK<br />

inhibition, supporting a pharmacologically active dose range for future<br />

clinical development.<br />

2599 General Poster Session (Board #8H), Mon, 8:00 AM-12:00 PM<br />

Effects <strong>of</strong> metformin and sulfonylureas on overall and colorectal cancerspecific<br />

mortality. Presenting Author: Susan Spillane, Trinity College<br />

Dublin, Dublin, Ireland<br />

Background: Preclinical studies have suggested a role for metformin in the<br />

treatment <strong>of</strong> colorectal cancer (CRC). Associations between metformin<br />

versus sulfonylurea exposure and mortality (all-cause and colorectal cancer<br />

specific) are assessed in this population-based study <strong>of</strong> patients with a<br />

diagnosis <strong>of</strong> stage I-IV CRC. Methods: National Cancer Registry Ireland<br />

records were linked to prescription claims data and used to identify a cohort<br />

<strong>of</strong> patients with incident TNM stage I-IV CRC diagnosed 2001-2006. From<br />

this cohort, 2 patient groups were identified and compared for outcomes -<br />

those who received a prescription for metformin �/- a sulfonylurea (MET) or<br />

a prescription for sulfonylurea alone (SUL) in the 90 days pre CRC<br />

diagnosis. Adjusted hazard ratios (HR) with 95% confidence intervals (CI)<br />

were estimated using Cox proportional hazards models adjusted for age,<br />

sex, stage, grade, site, comorbidities, year <strong>of</strong> diagnosis, and insulin, aspirin<br />

or statin exposure. Analyses were repeated stratifying by stage and site.<br />

Results: 5,617 patients with stage I-IV CRC were identified, <strong>of</strong> whom 369<br />

received a prescription for metformin or a sulfonylurea in the 90 days pre<br />

diagnosis (median follow-up 1.6 years; MET: n�257; SUL: n�112). In<br />

adjusted analyses metformin exposure was associated with a 28% lower<br />

risk <strong>of</strong> all-cause mortality relative to sulfonylurea exposure (HR 0.72, 95%<br />

CI 0.53-0.98) and a non-significant 24% reduction in CRC-specific<br />

mortality (HR 0.76, 95% CI 0.52-1.13). In analyses stratified by site, in<br />

colon cancer, metformin exposure was associated with a significant<br />

one-third reduction in all-cause mortality (HR 0.66, 95% CI 0.46-0.95)<br />

and a non-significant reduction in site-specific mortality (HR 0.64, 95% CI<br />

0.40-1.02). No mortality benefit was observed for rectal cancer. The<br />

association between metformin exposure and reduced mortality was strongest<br />

for stage I/II disease (all-cause mortality: HR 0.56, 95% CI 0.32-<br />

0.98; CRC-specific mortality: HR 0.48, 95% CI 0.21-1.11). Conclusions:<br />

Pre-diagnosis metformin exposure in CRC patients was associated with a<br />

significant reduction in mortality relative to sulfonylurea exposure. This<br />

benefit was greatest in patients with colon cancer and early stage disease.<br />

2598 General Poster Session (Board #8G), Mon, 8:00 AM-12:00 PM<br />

Docetaxel pharmacogenetics: The influence <strong>of</strong> RXR� and HNF4� genetic<br />

variations on docetaxel disposition in Asian nasopharyngeal carcinoma<br />

patients. Presenting Author: Sin Chi Chew, Division <strong>of</strong> Medical Sciences,<br />

National Cancer Centre, Singapore<br />

Background: The transactivations <strong>of</strong> the metabolism enzymes (CYP3A4/5)<br />

and efflux transporters (ABCB1/ABCC2) involved in docetaxel disposition<br />

are regulated by the orphan nuclear receptors such as PXR, CAR, RXR� and<br />

HNF4�. This study aimed to explore the associations between the genetic<br />

variations present in the genes encoding the orphan nuclear receptors,<br />

RXR� and HNF4�, on docetaxel disposition. Methods: The DNAs from<br />

healthy Chinese, Malay and Indian subjects (n�56 each) were screened for<br />

RXR� and HNF4� SNPs in exons and intronic/exonic boundaries by direct<br />

sequencing. The high frequency SNPs (�1%) were pr<strong>of</strong>iled in a cohort <strong>of</strong><br />

local nasopharyngeal cancer patients (n�54). Genotypic-phenotypic correlations<br />

were conducted using Mann-Whitney U-test and Kruskal-Wallis<br />

test. Results: Eighty-eight and sixty-nine SNPs were identified from the<br />

healthy screening <strong>of</strong> RXR� and HNF4�, respectively, across 3 populations.<br />

A total <strong>of</strong> 30 and 35 high frequency SNPs in RXR� and HNF4�,<br />

respectively, were pr<strong>of</strong>iled in the patients. Six RXR� SNPs [IVS2�33G�A<br />

(rs2234753), IVS7�70A�G (rs1536475),*846G�A (rs4240711),<br />

*�4458G�A (rs3132291), *�4768C�A (rs4842196) and *�4988A�G<br />

(rs4842198)] and 6 HNF4� SNPs [-728A�C (rs1800963), IVS2-<br />

278A�G (rs55934816), IVS6�141A�G (rs6103731), IVS7-88T�C<br />

(rs2273618), IVS9–145T�C (rs3746574) and IVS9-67C�G<br />

(rs3746575)] were significantly associated with higher clearance and<br />

lower AUC0-� and/or Cmax <strong>of</strong> docetaxel (P�0.05). Conversely, HNF4� SNP<br />

[IVS9�354G�T (rs3818247)] was associated with lower clearance and<br />

higher AUC0-� and Cmax <strong>of</strong> docetaxel (p�0.05). A high linkage pattern was<br />

observed among the abovementioned RXR� SNPs except for IVS2�33G�A<br />

(rs2234753) (D’�0.74). Similarly, HNF4� SNPs were found to be highly<br />

linked, except for -728A�C (rs1800963) (D’�0.62). Conclusions: The<br />

results highlight the contributions <strong>of</strong> RXR� and HNF4� pharmacogenetics<br />

in influencing the inter-individual variability in docetaxel disposition in<br />

Asian nasopharyngeal patients.<br />

2600 General Poster Session (Board #9A), Mon, 8:00 AM-12:00 PM<br />

Antibody-dependent cell-mediated cytotoxicity (ADCC) evolution under<br />

treatment by cetuximab and links with treatment outcome in colorectal<br />

cancer (CRC) patients. Presenting Author: Marco Carlo Merlano, St. Croce<br />

General Hospital, Cuneo, Italy<br />

Background: ADCC plays a role in antitumor activity <strong>of</strong> IgG1 mAb by<br />

inducing immune cell-mediated lysis <strong>of</strong> tumor cells. We evaluated ADCC<br />

ability before and under cetuximab-based treatment and examined its<br />

impact on treatment outcome. Methods: 29 patients (17 men, 12 women,<br />

median age 72, range 51-84) with metastatic wild-type KRAS CRC treated<br />

with chemotherapy (irinotecan or Folfiri) plus cetuximab were prospectively<br />

enrolled. ADCC ex-vivo was measured before starting treatment and every 2<br />

months during treatment (1 to 7 measurements/patient, 12 patients with<br />

�2 measurements). 400 000 purified Natural Killer cells (CD3- CD56�)<br />

from patients were incubated with 10 000 target cells (CAL166 cancer cell<br />

line expressing EGFR) and 10 �g/ml cetuximab (triplicates). Cytotoxicity<br />

was measured by the LDH-release assay. ADCC was expressed as the % <strong>of</strong><br />

lyzed target cells. Gene polymorphisms <strong>of</strong> Fc� receptors FCGR2a<br />

(131Arg�His) and FCGR3a (158Phe�Val) were analyzed (Allelic Discrimination<br />

assay). Results: The feasibility rate <strong>of</strong> ADCC measurement was 88%.<br />

ADCC basal values ranged between 30% and 100% (mean 62%, median<br />

66%). Basal ADCC was not influenced by patient gender. A tendency for an<br />

increased ADCC basal value was observed in younger patients: median was<br />

76% in the 6 patients � 60 vs 56% in the 23 patients over 60 years-old (p<br />

� 0.031). FCGR2A and FCGR3A gene polymorphisms were not linked to<br />

basal ADCC. The evolution <strong>of</strong> individual ADCC ability before treatment and<br />

2 months later revealed a significant drop in ADCC following treatment<br />

initiation (intra-patient comparison, n�18, p�0.006), with an absolute<br />

median drop <strong>of</strong> 19%. This decrease was not sustained over time and<br />

intra-patient comparison between basal value and 4-month measurement<br />

was not significant. 25 patients were assessable for survival (11 deaths).<br />

Basal ADCC values were not related to survival. Conclusions: A new<br />

generation <strong>of</strong> mAb is currently being developed with the aim to amplify<br />

ADCC. Present data illustrate the feasibility <strong>of</strong> ADCC measurement in<br />

mAb-treated patients and reveal an initial drop in ADCC under treatment<br />

that may reflect the variable chemotherapy-induced impact on host<br />

immunity.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


Developmental Therapeutics—<strong>Clinical</strong> Pharmacology and Immunotherapy<br />

2601 General Poster Session (Board #9B), Mon, 8:00 AM-12:00 PM<br />

Biomarkers <strong>of</strong> vincristine neuropathy in Kenyan children. Presenting<br />

Author: Jodi L. Skiles, Riley Hospital for Children, Indiana University<br />

School <strong>of</strong> Medicine, Indianapolis, IN<br />

Background: In a U.S. population, cytochrome P450 (CYP) 3A5 highexpressers<br />

metabolize vincristine (VCR) more efficiently than lowexpressers<br />

and vincristine-induced peripheral neuropathy (VIPN) is less<br />

common in African-<strong>American</strong>s than Caucasians. We test the hypothesis<br />

that more children in Kenya are CYP3A5 high-expressers compared to U.S.<br />

children and as such experience less VIPN. Methods: This study was<br />

conducted in Kenyan children with cancer (n�78) being treated with VCR<br />

at Moi University AMPATH Oncology Institute in partnership with Indiana<br />

University. Saliva Oragene kits for DNA extraction and genotyping were<br />

obtained. Whole blood was collected via finger stick on Whatman protein<br />

saver dried blood spot cards for analysis <strong>of</strong> VCR concentrations. VIPN was<br />

assessed prospectively using two neuropathy assessment tools (Modified<br />

Total Neuropathy Score (TNS) and National Cancer Institute Common<br />

Terminology Criteria for Adverse Events (CTCAE), version 4.0). Results:<br />

Compared to 14% in the U.S. sample, 94% <strong>of</strong> Kenyan subjects are<br />

CYP3A5 high-expressers (p�2.2x10-16 ). Kenyan children have significantly<br />

lower VCR plasma concentrations (11.15ng/ml/mg � 1.051) compared<br />

to US (primarily Caucasian) children (13.26ng/ml/mg � 2.897) one<br />

hour following the administration <strong>of</strong> VCR (p�0.011). By TNS VIPN<br />

assessment, 71% <strong>of</strong> Kenyan children developed VIPN compared to 100%<br />

<strong>of</strong> U.S. children (p�8.8x10-7 ). CYP3A5 high-expresser genotype is associated<br />

with lower TNS VIPN severity scores than low-expresser genotypes<br />

(p�0.006). TNS VIPN assessments show that height is positively correlated<br />

with severity <strong>of</strong> VIPN (p�0.025). CTCAE VIPN assessments also<br />

showed a positive correlation with height (p�0.036), but did not show any<br />

association with CYP3A5 genotype. Conclusions: Kenyan children are more<br />

likely to be CYP3A5 high-expressers than U.S. children and as such may<br />

metabolize VCR more efficiently. Supporting these data is that Kenyan<br />

children experience significantly less VIPN than U.S. children. CYP3A5<br />

genotype and height are independent predictors <strong>of</strong> VIPN in Kenyan children<br />

with cancer. The CTCAE may lack sufficient sensitivity to detect VIPN for<br />

use in future studies aimed at optimizing VCR dosing strategies.<br />

2603 General Poster Session (Board #9D), Mon, 8:00 AM-12:00 PM<br />

Dovitinib (TKI258): Exploration <strong>of</strong> pharmacokinetics and pharmacodynamics<br />

(PK/PD) for dose and regimen consideration. Presenting Author: Samira<br />

M. Garonzik, Novartis Pharmaceuticals, East Habover, NJ<br />

Background: Dovitinib is a potent oral inhibitor <strong>of</strong> Receptor Tyrosine<br />

Kinases with activity against FGFR, VEGFR and PDGFR. The objectives <strong>of</strong><br />

this analysis are to describe the PK/PD <strong>of</strong> dovitinib to characterize the<br />

differences between two different dosing regimens, 400 mg once daily (qd)<br />

or 500 mg 5 days on, 2 days <strong>of</strong>f (int) in terms <strong>of</strong> exposure and biomarker<br />

response. Methods: PK/PD data from 127 patients receiving dovitinib,<br />

100-600 mg int and 50 – 600 mg qd were available. Duration <strong>of</strong> therapy<br />

was 15 to 859 days. Plasma concentration-time data � sparse biomarker<br />

(BM) data were available after the first dose and at steady state (SS). Data<br />

was analyzed using non-linear mixed effects modeling to characterize<br />

nonlinearities in PK as well as BM response. VEGF, sVEGFR2 and PDGF<br />

were described by indirect response models and FGF23 was described<br />

using a mechanism based model to characterize acute drop, rebound,<br />

tolerance and increase to a new SS over time. Results: Raw data revealed<br />

prolonged absorption, linear clearance after the first dose, but dose<br />

dependent 2 in clearance at SS (leading to over-proportional accumulation),<br />

and apparent linear clearance at doses below 400 mg qd. 2 exposure<br />

for doses up to 400 mg at SS compared to 1st dose implied auto-induction.<br />

The PK was well described by a 1-compartment (CMT) model with 3 transit<br />

CMTs to characterize absorption lag. Auto-induction and over-proportional<br />

accumulation at SS were modeled as depending on cumulative exposure<br />

through a time-dependent process with half-life <strong>of</strong> 18 h. In our study,<br />

median (10th –90th percentile) Clearance was 30 (17 – 80) L/h on Day 1,<br />

and 96 (51 – 185) L/h at SS. The model predicted 36%1in VEGF, 18% 2<br />

in sVEGFR2, 82% and 57% 1in PDGF and FGF23 respectively at SS, for<br />

median exposure at a dose <strong>of</strong> 400 mg qd. Simulations <strong>of</strong> PK and biomarker<br />

responses were performed for different dosing regiments to assess relative<br />

safety and target effects. Conclusions: The PK/PD model suggests 400 mg<br />

qd may lead to over-proportional accumulation <strong>of</strong> dovitinib in �5% <strong>of</strong><br />

subjects while int dosing minimizes this. 500 mg int may be a more<br />

tolerable regimen while maintaining adequate PD response, and is being<br />

used in the pivotal phase III study for dovitinib in patients with mRCC.<br />

167s<br />

2602 General Poster Session (Board #9C), Mon, 8:00 AM-12:00 PM<br />

Is cigarette smoke associated with increased catabolism <strong>of</strong> gemcitabine in<br />

patients with advanced solid tumors? Presenting Author: Meaghan Working<br />

O’Malley, University <strong>of</strong> Michigan Comprehensive Cancer Center, Ann Arbor,<br />

MI<br />

Background: Cigarette smoking can accelerate chemotherapy metabolism<br />

and result in lower plasma concentrations <strong>of</strong> the chemotherapeutic agent.<br />

Reduced drug levels may lead to undertreatment in smokers and, conversely,<br />

increased treatment-related neutropenia in nonsmokers. Methods:<br />

An IRB-approved retrospective chart review was performed on 151 patients<br />

with solid tumor malignancies who received gemcitabine alone or in<br />

combination with oral chemotherapy agents from July 1, 2009 to June 30,<br />

2011 at the University <strong>of</strong> Michigan. Using logistic regression, we compared<br />

toxicity, including neutropenia, and smoking history measured in packyears<br />

in smokers vs. nonsmokers to ascertain whether cigarette smoking is<br />

an independent factor predictive <strong>of</strong> toxicity to gemcitabine. Results: Tumor<br />

types included breast (9.3%), lung (4.6%), pancreatobiliary (70.9%), or<br />

other/unknown primary (15.2%). Most patients had advanced disease<br />

(stage III-IV; 78.1%); specifically, <strong>of</strong> the pancreatobiliary cohort (PB;<br />

n�107), 77.6% patients had stage III-IV disease. Within the PB cohort,<br />

most patients were “ever” smokers as compared to “never” smokers<br />

(60.7% vs. 39.3%). Logistic regression <strong>of</strong> this cohort showed that current<br />

smokers had decreased CTC-AE grade 3-4 neutropenia vs. never smokers<br />

(OR 0.667; 95% CI [0.156-2.859]). This effect was more pronounced with<br />

higher pack-year history: smokers with �50 pack-years had even less<br />

neutropenia as compared to never smokers (OR 0.333; 95% CI [0.065-<br />

1.699]). Further statistical analysis <strong>of</strong> subgroups was not performed due to<br />

small sample size. Conclusions: Smokers with pancreatobiliary malignancies<br />

receiving gemcitabine had less treatment-related neutropenia as<br />

compared to never smokers, a finding that was more pronounced as<br />

pack-years increased. Decreased toxicity, including neutropenia, may be<br />

due to increased metabolism and drug clearance as a result <strong>of</strong> smoking.<br />

This may lead to potential undertreatment <strong>of</strong> smokers and, conversely,<br />

increased treatment-related toxicity in never smokers. A prospective<br />

clinical trial is needed to further elucidate this correlation, and is currently<br />

being designed.<br />

2604 General Poster Session (Board #9E), Mon, 8:00 AM-12:00 PM<br />

A randomized, double-blind, placebo-controlled study to assess the efficacy<br />

and toxicity <strong>of</strong> subcutaneous ketamine in the management <strong>of</strong> cancer<br />

pain. Presenting Author: David C. Currow, Flinders University, Adelaide,<br />

Australia<br />

Background: The dissociative anaesthetic ketamine is widely used for<br />

cancer related pain. A Cochrane review concluded that insufficient evidence<br />

was available to support its use in this setting. Methods: This phase<br />

III, multisite, double-blind, dose escalation, placebo, randomised controlled<br />

study aimed to determine whether ketamine, delivered subcutaneously<br />

over three to five days is more effective than placebo, when used in<br />

conjunction with adjuvant therapy in the management <strong>of</strong> chronic uncontrolled<br />

cancer pain. Ketamine would be considered to be <strong>of</strong> net benefit if it<br />

provided a reduction in average pain scores by �2/10 points from baseline,<br />

with limited breakthrough analgesia and acceptable toxicity. Results: For<br />

the 185 participants, there was no significant difference between the<br />

proportion <strong>of</strong> positive outcomes (0.04 (-0.10, 0.18) p�0.55) in the<br />

placebo and intervention arms (response rates 27% (25/92) and 31%<br />

(29/93)). Pain type (nociceptive versus neuropathic) was not a predictor <strong>of</strong><br />

response. There was almost twice the incidence <strong>of</strong> adverse events worse<br />

than baseline in the ketamine group after day 1 (IRR � 1.95 (1.46, 2.61),<br />

p�0.001) and throughout the study. Those receiving ketamine were more<br />

likely to experience a more severe grade <strong>of</strong> adverse event/day (OR�1.09<br />

(1.00, 1.18), p�0.039). The number needed to treat for one additional<br />

patient to get a positive outcome from ketamine was 25 (6, �). The number<br />

needed to harm, because <strong>of</strong> toxicity-related withdrawal was 6 (4, 13).<br />

Conclusions: Ketamine does not have net clinical benefit when used as an<br />

adjunct to opioids and standard co-analgesics in cancer pain.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


168s Developmental Therapeutics—<strong>Clinical</strong> Pharmacology and Immunotherapy<br />

2605 General Poster Session (Board #9F), Mon, 8:00 AM-12:00 PM<br />

Vectorized gene therapy <strong>of</strong> liver tumors: Safety and pro<strong>of</strong> <strong>of</strong> concept <strong>of</strong><br />

TG4023 (MVA-FCU1)/5-FC combination. Presenting Author: Faress Husseini,<br />

Hôpitaux Civils de Colmar, Colmar, France<br />

Background: TG4023 is a non-integrative and non-propagative Modified<br />

Vaccinia virus Ankara (MVA) expressing a chimeric yeast transgene (FCU1)<br />

coding for cytosine deaminase and uracil phosphoribosyl transferase that<br />

transform the pro-drug 5-flucytosine (5-FC) respectively into 5-FU and<br />

5-FUMP in infected cells; these derivatives then diffuse and kill additional<br />

tumor cells (bystander effect). Methods: This phase I study determined<br />

safety and Maximal Tolerated Dose (MTD) <strong>of</strong> a single intra-tumor injection<br />

<strong>of</strong> TG4023 combined with a 2-week dosing period <strong>of</strong> 5-FC 200 mg/kg/day.<br />

Plasma and tumor 5-FC (PK) and 5-FU (PD) concentrations were measured.<br />

Other assessments were tumor response, changes in tumor markers<br />

and immune response. Patient requirements were � 1 injectable primary or<br />

metastatic unresectable liver tumor <strong>of</strong> 2-5 cm, no treatment option left,<br />

ECOG PS � 2. Consenting patients were allocated to cohorts according to a<br />

3�3 dose-escalating design driven by Dose-Limiting Toxicities (DLTs) and<br />

Data Safety Monitoring Board recommendations. Results: Among 16<br />

enrolled patients (13 colorectal, 1 pancreatic and 1 liver cancers, 1 cancer<br />

<strong>of</strong> unknown primary) 6 were injected in one tumor with 107 plaque forming<br />

units (pfu) TG4023, 3 with 108 pfu and 7 with 4.108 pfu, using a multiport<br />

needle and ultrasound imaging guidance. 5-FC was given IV the first days<br />

then orally. One DLT (grade 3 transient increase in AST/ALT) was recorded<br />

at 4.108 pfu; other severe adverse events, diarrhea, hypertension, alkaline<br />

phosphatase increase were related to 5-FC; most frequent AEs were<br />

transient fever, asthenia, site injection pain, nausea/vomiting. 5-FU concentrations<br />

in tumor biopsies at Day 8 were 56�30 ng/g and 1.9� 2.6 ng/mL<br />

in plasma. 11/16 injected, 12/23 non-injected target tumors were stable<br />

and 10/16 patients progressed at week 6. One patient had a 3-fold<br />

decrease in CEA and CA 19.9. Conclusions: This study showed that<br />

vectorized gene therapy <strong>of</strong> liver tumors with TG4023/5-FC combination is<br />

feasible and safe, as supported by a high therapeutic index; MTD for<br />

TG4023 was 4.108 pfu for one injected tumor. A pro<strong>of</strong> <strong>of</strong> concept <strong>of</strong> the in<br />

vivo conversion <strong>of</strong> the pro-drug 5-FC into 5-FU was obtained.<br />

2607 General Poster Session (Board #9H), Mon, 8:00 AM-12:00 PM<br />

Impact <strong>of</strong> body surface area on efficacy <strong>of</strong> gefitinib in patients with<br />

non-small cell lung cancer harboring activating epidermal growth factor<br />

receptor mutation. Presenting Author: Eiki Ichihara, Department <strong>of</strong> Respiratory<br />

Medicine, Okayama University Hospital, Okayama, Japan<br />

Background: Gefitinib is an essential drug for the treatment <strong>of</strong> non-small<br />

cell lung cancer (NSCLC) with epidermal growth factor receptor (EGFR)<br />

mutation. Its approved dosage is fixed at 250 mg/body/day without any<br />

adjustment by physical size. On the other hand, most cytotoxic agents are<br />

administered with body surface area (BSA) based dose-adjustment to<br />

reduce the pharmacokinetic inter-patient variability. However, the impact<br />

<strong>of</strong> BSA on efficacy <strong>of</strong> gefitinib has not been fully evaluated. We here tried to<br />

clarify this issue using our retrospective cohort. Methods: We reviewed the<br />

medical records <strong>of</strong> consecutive advanced NSCLC patients harboring EGFR<br />

mutation who underwent gefitinib monotherapy at Okayama University<br />

Hospital from October 2002, when gefitinib was approved in Japan, to<br />

October 2011. Results: A total <strong>of</strong> 101 patients with EGFR-mutant tumors<br />

who received gefitinib (250 mg/body/day) were included in this study. The<br />

median progression free survival (PFS) <strong>of</strong> patients with high BSA (�1.5 m2 :<br />

the median value in this cohort) was significantly worse than that <strong>of</strong><br />

patients with low BSA (�1.5 m2 ) (10.4 vs. 18.0 months, p � 0.019<br />

log-rank test). The multivariate analysis also showed a significant impact <strong>of</strong><br />

BSA on PFS; (hazard ratio <strong>of</strong> 2.34 with 95% confidence interval <strong>of</strong> 1.78 -<br />

2.89, p � 0.002). By contrast, such significant association between BSA<br />

and PFS was not observed in cytotoxic chemotherapy (5.4 months vs. 3.7<br />

months, p � 0.49, log lank test). Conclusions: BSA affected PFS in the<br />

gefitinib monotherapy, potentially suggesting the need for appraisal <strong>of</strong><br />

BSA-based dose adjustment even in this molecular-target therapy.<br />

2606 General Poster Session (Board #9G), Mon, 8:00 AM-12:00 PM<br />

The role <strong>of</strong> phase I clinical trials in advanced malignant melanoma:<br />

Retrospective analysis <strong>of</strong> CTEP-sponsored trials 1995-2011. Presenting<br />

Author: Jason John Luke, Dana-Farber Cancer Institute, Boston, MA<br />

Background: Standard chemotherapy for melanoma is DTIC (RR ~10%).<br />

Many physicians do not refer to phase I due to perceived limited clinical<br />

benefit (CB�CR�PR�SD) and increased toxicity. To understand the<br />

actual experience <strong>of</strong> melanoma patients (pts) in phase I trials, we analyzed<br />

the outcomes <strong>of</strong> melanoma pts treated on CTEP phase 1 trials (1995-<br />

2011) and compared them to DTIC. Methods: We queried the CTMS <strong>of</strong><br />

CTEP for phase I trials in which advanced melanoma pts were treated.<br />

Trials were separated into targeted (T), chemo (C) and immunotherapy (I).<br />

Pt characteristics, response and toxicity data were collected. Chemotherapy<br />

included chemo with targeted or immunotherapy. Toxicity was drug<br />

related if attributed possibly, probably or definitely to drug. Fisher’s Exact<br />

Test (2-sided p) was used to compare groups. DTIC data was pooled from 6<br />

modern phase III clinical trials (1999-2011). Results: 937 pts (M595:<br />

F342) participated in 148 trials (T: 68, C: 53, I: 27). Characteristics<br />

included (median) Age: 51.5 yrs; ECOG status: 0; Prior therapies: 2<br />

(majority receiving prior DTIC); LDH: 206 and albumin: 4.1. Response and<br />

toxicity data are shown in the Table. Targeted therapy was associated with<br />

lower RR (p�.01), immuno with lower CB rate (p�.001) and chemo with<br />

higher incidence <strong>of</strong> G4 toxicity (p�.001) relative to the other groups.<br />

Comparing phase 1 to DTIC, RR and CB were not clinically different (phase<br />

I: 6.3% and 26.8% vs. DTIC: 8.8% and 27.9%) but G3-4 toxicity was<br />

significantly higher (54% vs. 28%) in phase I (p�.0001). Conclusions:<br />

Melanoma pts in prior CTEP phase I trials, a majority DTIC pre-treated, had<br />

similar therapeutic benefit but more toxicity compared to DTIC naïve pts in<br />

modern clinical trials.<br />

All (937) Targeted (182) Chemo (186) Immuno (569)<br />

Response Total % 95% CI Total % 95% CI Total % 95% CI Total % 95% CI<br />

CR � PR 59 6.3 4.8-8.1 4 2.2 0.6-5.5 14 7.5 4.2-12.3 41 7.2 5.2-9.7<br />

SD 192 20.5 59 32.4 53 28.5 80 14.0<br />

CB 251 26.8 24.0-29.8 63 34.6 27.7-42.0 67 36.0 29.1-43.4 121 21.2 18.0-24.9<br />

Not evaluated 61 6.5 15 8.2 16 8.6 30 5.3<br />

PD<br />

Toxicity<br />

625 66.7 104 57.1 103 55.4 418 73.3<br />

G3 G4 G3 G4 G3 G4 G3 G4<br />

% 42.2 12.1 42.3 9.3 46.2 23.7 40.8 9.1<br />

95% CI 39.0-45.4 10.0-14.3 35.0-49.8 5.5-14.5 38.9-53.7 17.8-30.4 36.7-44.9 6.9-11.8<br />

2608 General Poster Session (Board #10A), Mon, 8:00 AM-12:00 PM<br />

Multivariate screening <strong>of</strong> prognostic factors to identify a novel, simple, and<br />

objective marker to optimize patient selection and predict survival benefit<br />

in early-phase trials. Presenting Author: Chara Stavraka, Imperial College<br />

London, London, United Kingdom<br />

Background: Several prognostic indices have been devised to optimize<br />

patient selection for phase I trials. However, there is no consensus as to the<br />

optimal score and none qualifies as a marker <strong>of</strong> response. We developed a<br />

simple score through a multivariate screening <strong>of</strong> individual variables and<br />

compared its performance with existing prognostic scores including the<br />

Royal Marsden, Nijmegen and Prince Margaret Hospital scores. Methods:<br />

We retrospectively analyzed characteristics and outcomes <strong>of</strong> 120 referrals<br />

to our phase I center (2007 - 2011). Independent predictors for overall<br />

survival (OS) were identified from univariate (Kaplan Meier) and multivariate<br />

(Cox regression) analyses and used to create the Hammersmith Hospital<br />

score (HS). This was compared with the other indices for predicting<br />

progression free survival (PFS), OS and 90 day mortality (90 DM).<br />

Multivariate logistic regression and ROC curves were used to estimate 90<br />

DM and c-index was used to estimate the prognostic ability <strong>of</strong> the different<br />

indices. Changes in HS following treatment (�HS) were calculated at 6<br />

weeks RECIST in a subset <strong>of</strong> patients receiving targeted therapies (n� 50).<br />

Results: Median age was 62 years (range: 28 – 80); median OS 4.3 months<br />

(range: 0.2 – 39); 34% male; 25% PS�1. Multivariate screening identified<br />

albumin �35 g/L, lactate dehydrogenase (LDH) �450 U/L and sodium<br />

�135mmol/L as the strongest independent predictors <strong>of</strong> OS (p�0.05).<br />

These were entered into a 3-point score (HS) that classifies patients as<br />

being at high risk (score 2-3) vs. low risk (score 0-1) <strong>of</strong> worse OS (HR� 5.8,<br />

p�0.001), PFS (HR� 2.7, p� 0.04) and 90 DM (OR� 5.9, p� 0.001). All<br />

scores predicted for OS on univariate analysis (p�0.05). On multivariate<br />

analysis HS performed best to predict OS (HR� 3.9, p�0.001 C-index<br />

score� 0.71), PFS (HR� 2.6 p� 0.04) and 90 DM with area under the<br />

ROC curve 0.71. �HS independently predicted for OS (p�0.001), with<br />

worsening <strong>of</strong> the score reflecting poorer OS. Prospective validation is<br />

ongoing. Conclusions: HS is a simple score that outperforms other prognostic<br />

indices. This can be used to select individuals for future studies as well<br />

as an additional marker <strong>of</strong> response.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


2609 General Poster Session (Board #10B), Mon, 8:00 AM-12:00 PM<br />

Should patients with extrapulmonary small cell carcinoma receive prophylactic<br />

cranial irradiation? An Irish experience. Presenting Author: Jarushka<br />

Naidoo, Waterford Regional Hospital, Waterford, Ireland<br />

Background: Extrapulmonary small cell carcinoma (EPSCC) is a rare<br />

disease. Management is based on small cell lung carcinoma, and experience<br />

<strong>of</strong> disease biology on single institution studies. Prophylactic cranial<br />

irradiation (PCI) is not routinely administered in EPSCC. This study<br />

investigates the role <strong>of</strong> PCI in EPSCC, by analyzing the incidence,<br />

treatment and survival <strong>of</strong> patients with brain metastases in a national<br />

cohort. Secondary aims were to investigate disease biology and epidemiology.<br />

Methods: An audit was undertaken <strong>of</strong> patients diagnosed with primary<br />

EPSCC from the National Cancer Registry <strong>of</strong> Ireland, between 1995-2007.<br />

The number <strong>of</strong> patients who developed brain metastases, received cranial<br />

radiotherapy, and their survival data, were documented. Patient and<br />

disease characteristics, treatment, and survival data stratified by stage and<br />

primary site, were tabulated. Results: 280 patients were found. 141<br />

(50.4%) patients were male, and 139 (49.6%) were female. 186 (66.4%)<br />

patients had extensive stage disease, 65 (23.2%) had limited stage<br />

disease, and in 29 (10.3%) patients the stage was not known. 17 patients<br />

(5.4%) developed brain metastases, 11 <strong>of</strong> which (64.7%) received cranial<br />

irradiation. These patients had a median progression-free survival (PFS) <strong>of</strong><br />

5.2 months, and a median overall survival (OS) <strong>of</strong> 10.2 months. The<br />

commonest primary sites were the oesophagus (n�43, 15.4%), cervix uteri<br />

(n�17, 6.0%), bladder (n�13, 4.6%) and prostate (n�10, 3.6%). There<br />

were 315 events <strong>of</strong> distant metastasis, and 21 local recurrence events. The<br />

commonest metastatic sites were the liver, 110 (34.9%), lymph nodes 58<br />

(20.7%), lung 38 (12.1%), and bone 36 (12.8%). The median PFS and OS<br />

for limited stage EPSCC was 8.8 months and 14.9 months, and 1.5 months<br />

and 2.3 months for extensive stage EPSCC. Conclusions: Brain metastases<br />

were uncommon in this cohort (5.4%), and patients lived longer, suggesting<br />

a low rate <strong>of</strong> mortality from brain metastases. PCI is thus probably not<br />

warranted in EPSCC. Prospective data are necessary to support PCI in<br />

EPSCC. This is one <strong>of</strong> the largest datasets on EPSCC, providing insights<br />

into disease biology.<br />

2611 General Poster Session (Board #10D), Mon, 8:00 AM-12:00 PM<br />

Evaluation <strong>of</strong> utility <strong>of</strong> pharmacokinetic studies in phase I combination<br />

trials. Presenting Author: Kehua Wu, The University <strong>of</strong> Chicago, Chicago, IL<br />

Background: There are many phase I clinical combination trials including<br />

drug-drug interaction (DDI) studies, but very few <strong>of</strong> them report positive<br />

results. We hypothesized that the utility <strong>of</strong> DDI studies is low in the absence<br />

<strong>of</strong> a mechanistic hypothesis. Methods: We retrospectively reviewed 119<br />

phase I (2 drug) combination studies published in 2007-2011. Results:<br />

Only 23 (19%) studies had a positive rationale, either inhibition/induction<br />

<strong>of</strong> a drug metabolizing enzyme or transporter, co-substrates for the same<br />

enzyme or transporter, potential for end-organ toxicity, or protein- binding.<br />

Only 9 (8%) studies demonstrated a statistically significant effect DDI,<br />

based on AUC <strong>of</strong> parent drug or active metabolite. There was a strong<br />

association between lack <strong>of</strong> rationale and a lack <strong>of</strong> interaction, as only 2%<br />

<strong>of</strong> studies without a rationale demonstrated a DDI, compared to 30% <strong>of</strong><br />

studies with a rationale (Fisher’s exact test, p�0.0004) (Table). Conclusions:<br />

DDI studies should not be routinely performed as part <strong>of</strong> phase I clinical<br />

trials.<br />

Preclinical<br />

rationale<br />

Developmental Therapeutics—<strong>Clinical</strong> Pharmacology and Immunotherapy<br />

Number <strong>of</strong><br />

papers with (�) DDI<br />

Number <strong>of</strong><br />

papers with (�) DDI<br />

(�) 94 2<br />

(�) 16 7<br />

169s<br />

2610 General Poster Session (Board #10C), Mon, 8:00 AM-12:00 PM<br />

Phase I/II dose-finding study <strong>of</strong> crizotinib (CRIZ) in combination with<br />

erlotinib (E) in patients (pts) with advanced non-small cell lung cancer<br />

(NSCLC). Presenting Author: Sai-Hong Ignatius Ou, Chao Family Comprehensive<br />

Cancer Center, Orange, CA<br />

Background: c Met expression is common in NSCLC tumors and has been<br />

implicated in the development <strong>of</strong> resistance to EGFR inhibitors. CRIZ is an<br />

ALK and MET/HGF receptor tyrosine kinase inhibitor (TKI). In pre-clinical<br />

studies, combining CRIZ with an EGFR inhibitor enhanced anti-tumor<br />

activity in NSCLC cell lines that were either sensitive or resistant to EGFR<br />

inhibition. The phase I portion <strong>of</strong> a phase I/II study (A8081002;<br />

NCT00965731) investigated the combination <strong>of</strong> E (EGFR TKI) and CRIZ in<br />

pts with advanced NSCLC. Methods: Pts had advanced NSCLC, 1 or 2 prior<br />

chemotherapy regimens, and no previous MET-directed therapy. Endpoints<br />

included maximum tolerated dose (MTD) determination, safety, and<br />

pharmacokinetics (PK). Pts received CRIZ 150 or 200 mg BID combined<br />

with E 100 mg QD (150/100 and 200/100, respectively). Results:<br />

Twenty-five pts have been treated to date. Median duration <strong>of</strong> combination<br />

therapy in 150/100 (n�18) was 6.6 weeks (0.1–25.3); for 200/100 (n�7)<br />

was 6.9 weeks (3.0–64.7). Five pts had dose-limiting toxicities (grade [G]<br />

2 and unable to receive at least 80% <strong>of</strong> the planned dose or �G3), all <strong>of</strong><br />

which resolved: 3 pts at 150/100, G2 vomiting, G2 esophagitis and<br />

dysphagia, G3 diarrhea and dehydration; and at 200/100, G3 dry eye (1 pt)<br />

and G3 esophagitis (1 pt). Most pts (92%) experienced treatment-related<br />

adverse events (TRAEs), mainly <strong>of</strong> G1 or 2 severity. Common TRAEs were<br />

diarrhea (72%), rash (56%) and fatigue (44%). Six pts discontinued<br />

therapy due to TRAEs (150/100: G3 diarrhea, G3 dehydration, and G2 rash<br />

in 1 pt, G2 asthenia, G2 vomiting, G2 esophagitis, n�1 each; 200/100: G3<br />

dry eyes, G1 esophagitis, n�1 each). One partial response (200/100;<br />

duration 61 weeks) and 9 stable diseases (n�7 150/100, n�2 200/100;<br />

duration 7–54 weeks) were observed overall. Co-administration <strong>of</strong> both<br />

doses <strong>of</strong> CRIZ with E increased E AUC by 1.8 fold, while CRIZ PK<br />

parameters appeared to be unaffected. Plasma exposure to E 100 mg QD<br />

with CRIZ was comparable to that <strong>of</strong> 150 mg QD from historical data.<br />

Conclusions: E plus CRIZ at the MTD was well tolerated, with no unexpected<br />

AEs, and showed signs <strong>of</strong> activity in a pre-treated population. E 100 mg QD<br />

plus CRIZ 150 mg BID was defined as MTD.<br />

2612 General Poster Session (Board #10E), Mon, 8:00 AM-12:00 PM<br />

Effect <strong>of</strong> baseline QTc interval prolongation on oncology clinical trial<br />

enrollment. Presenting Author: Carolina Moreno, H. Lee M<strong>of</strong>fitt Cancer<br />

Center & Research Institute, Tampa, FL<br />

Background: An increasing number <strong>of</strong> clinical trials in oncology require<br />

normal or near-normal intervals <strong>of</strong> electrocardiographic corrected QT (QTc)<br />

as an eligibility criterion. However the normal QTc reference range<br />

(conventionally defined as �450ms) has been developed in broad populations<br />

<strong>of</strong> patients, and it is unclear whether average QTc intervals are higher<br />

in adult cancer patients. Methods: All electrocardiograms (ECGs) performed<br />

for any indication at the M<strong>of</strong>fitt Cancer Center during a one-week period<br />

were reviewed. The QTc interval for each patient was calculated according<br />

to the Bazett and Fridericia formulas. Mean and median QTc intervals were<br />

calculated along with standard deviations. The percentage <strong>of</strong> QTc intervals<br />

above 450ms, 470ms, and 500ms were calculated and graded accordingly<br />

to the Common Terminology Criteria for Adverse Events v4.0 (CTCAE v4.0).<br />

The effects <strong>of</strong> gender on the QTc interval were also evaluated. Results: 257<br />

patients underwent ECGs over a one-week period. A total <strong>of</strong> 90 patients<br />

(35%) had abnormally prolonged QTc. 50 patients (20%) had QTc<br />

prolongation �450 (� grade 1), 32 (12%) had QTc prolongation � 470<br />

(�grade 2) and 8 patients (3%) had QTc prolongation �500ms (�grade 3)<br />

using the Bazett formula. The corresponding numbers according to the<br />

Fridericia formula were 23 (9%), 13 (5%) and 2 (1%) . The median QTc<br />

interval was 441 (Bazett) and 422 (Fridericia) with a standard deviation <strong>of</strong><br />

30 and 28 respectively. Women had a mean QTc <strong>of</strong> 438 (Bazett) versus<br />

443 for men. Conclusions: A very large percentage <strong>of</strong> patients seen at a<br />

comprehensive cancer center have QTc intervals above the normal reference<br />

range on routine ECGs. The growth in number <strong>of</strong> trials requiring a<br />

normal QTc interval as an eligibility criteria suggests that an ever-larger<br />

number <strong>of</strong> patients are excluded from trial participation. Regulatory<br />

agencies may need to reconsider eligibility criteria that substantially<br />

restrict trial enrollment and that accrue patients who are not reflective <strong>of</strong><br />

the general patient population.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


170s Developmental Therapeutics—<strong>Clinical</strong> Pharmacology and Immunotherapy<br />

TPS2613 General Poster Session (Board #10F), Mon, 8:00 AM-12:00 PM<br />

An exploratory study to investigate the immunomodulatory activity <strong>of</strong><br />

BMS-936558 in patients with metastatic clear-cell renal cell carcinoma.<br />

Presenting Author: Charles G. Drake, Sidney Kimmel Comprehensive<br />

Cancer Center at Johns Hopkins University, Baltimore, MD<br />

Background: With the enhanced understanding <strong>of</strong> the immune response to a<br />

tumor, new immunotherapeutic agents may expand treatment options for<br />

metastatic renal cell carcinoma (mRCC). The programmed death-1 (PD-1)<br />

receptor is upregulated by activated lymphocytes, and upon binding to its<br />

ligands, inhibits lymphocyte function. In a phase 1 study <strong>of</strong> the anti-PD-1<br />

monoclonal antibody BMS-936558, encouraging antitumor activity has<br />

been observed in patients with previously treated mRCC. Responses<br />

occurred in extensive mRCC and were durable in many patients. Here we<br />

describe an exploratory study to investigate the immune-regulatory activity<br />

<strong>of</strong> various doses <strong>of</strong> BMS-936558 in patients with metastatic clear-cell<br />

RCC. Methods: Eighty patients with metastatic clear-cell RCC will be<br />

enrolled in this open-label, parallel group, randomized study. Patients who<br />

have received prior treatment with �3 therapies, including �1 antiangiogenic<br />

therapy, will be enrolled in Arms 1-3 (~20 patients per arm); 20<br />

treatment-naïve patients will be enrolled in Arm 4. BMS-936558 will be<br />

administered every 3 weeks until disease progression or unacceptable<br />

toxicity at 0.3 mg/kg (Arm 1), 2 mg/kg (Arm 2), and 10 mg/kg (Arms 3 and<br />

4). Patients in the 0.3-mg/kg group may escalate to 2 mg/kg if not<br />

responding. Tumor assessments will occur every 6 weeks for the first 12<br />

months then every 12 weeks for the remainder <strong>of</strong> the study. The primary<br />

objective is to evaluate the effects <strong>of</strong> BMS-936558 on the frequency <strong>of</strong><br />

circulating immune cell subsets, levels <strong>of</strong> soluble factors associated with<br />

immune responses, and tumor infiltrating lymphocyte populations (based<br />

on pre-treatment and week 5 paired biopsies). Secondary objectives are to<br />

assess the safety, tolerability, antitumor activity, and immunogenicity <strong>of</strong><br />

BMS-936558. Exploratory objectives include the association <strong>of</strong> clinical<br />

activity and safety with selected biomarkers in peripheral blood and tumor<br />

microenvironment and the characterization <strong>of</strong> the pharmacokinetics <strong>of</strong><br />

BMS-936558. As <strong>of</strong> February 1, 2012, 30 patients were enrolled.<br />

TPS2615 General Poster Session (Board #10H), Mon, 8:00 AM-12:00 PM<br />

A phase I study <strong>of</strong> BMS-936558 in combination with gemcitabine/<br />

cisplatin, pemetrexed/cisplatin, or carboplatin/paclitaxel in patients with<br />

treatment-naive, stage IIIB/IV non-small-cell lung cancer. Presenting<br />

Author: Scott N. Gettinger, Yale University School <strong>of</strong> Medicine, New Haven,<br />

CT<br />

Background: Lung cancer is the leading cause <strong>of</strong> cancer-related mortality<br />

worldwide and non-small-cell lung cancer (NSCLC) accounts for ~85% <strong>of</strong><br />

all cases. Platinum-based chemotherapy alone or in combination with<br />

bevacizumab is considered first-line standard <strong>of</strong> care for advanced NSCLC.<br />

Despite such therapy, most patients (pts) will experience disease progression<br />

within 6 months and die from their disease within 1 year. Programmed<br />

death-1 (PD-1) is a coinhibitory receptor that negatively regulates T-cell<br />

activation; PD-1 expression by tumor infiltrating lymphocytes is associated<br />

with poor prognosis in NSCLC. BMS-936558 is a fully human anti-PD-1<br />

monoclonal antibody that blocks ligand interaction with PD-L1 and PD-L2.<br />

In a phase 1 trial, BMS-936558 had antitumor activity at doses <strong>of</strong> 1-10<br />

mg/kg in pts with advanced tumors, and impressive tumor responses in<br />

heavily pre-treated pts with NSCLC. Here we describe a phase 1 study<br />

planned to evaluate the safety and tolerability <strong>of</strong> BMS-936558 in combination<br />

with 3 standard, platinum-based doublet chemotherapy regimens in<br />

pts with NSCLC. Methods: Treatment-naïve pts with stage IIIB/IV NSCLC<br />

will be enrolled. Treatment arms will include: A) gemcitabine 1250 mg/M2 on day 1 (D1) and day 8 (D8)/cisplatin 75 mg/M2 D1/BMS-936558 D1; B)<br />

pemetrexed 500 mg/M2 D1/cisplatin 75 mg/M2 D1/BMS-936558 D1; and<br />

C) carboplatin AUC 6 D1/paclitaxel 200 mg/M2 D1/BMS-936558 D1. The<br />

starting dose <strong>of</strong> BMS-936558 will be 10 mg/kg for each treatment arm. A<br />

decision to de-escalate to a lower dose (2 or 0.3 mg/kg) will be guided by<br />

the number <strong>of</strong> dose-limiting toxicities reported per treatment arm. Chemotherapy<br />

will be given for 4 cycles (1 cycle � 3 weeks). BMS-936558 will be<br />

administered every 3 weeks until disease progression, intolerable toxicity,<br />

or withdrawal <strong>of</strong> consent. The primary objectives are to assess safety and<br />

tolerability by measuring the frequency <strong>of</strong> adverse events (AEs), serious<br />

AEs, and laboratory abnormalities; secondary objectives are to determine<br />

the overall response rate and disease control rates based on RECIST 1.1. As<br />

<strong>of</strong> February 1, 2012, 2 patients were enrolled.<br />

TPS2614 General Poster Session (Board #10G), Mon, 8:00 AM-12:00 PM<br />

A phase I study <strong>of</strong> BMS-936558 plus sunitinib or pazopanib in patients<br />

with metastatic renal cell carcinoma. Presenting Author: Hans J. Hammers,<br />

Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University,<br />

Baltimore, MD<br />

Background: Standard treatments for metastatic renal cell carcinoma<br />

(mRCC) block the vascular endothelial growth factor pathway (eg, sunitinib,<br />

pazopanib) or mammalian target <strong>of</strong> rapamycin pathway (e.g., temsirolimus,<br />

everolimus). However, most patients (pts) develop resistance and complete<br />

responses are rare. Upregulation <strong>of</strong> programmed death-1 (PD-1) in tumor<br />

infiltrating lymphoctyes, and its ligand PD-L1 in tumors, is associated with<br />

more aggressive disease and poor prognosis. Blocking the PD-1/PD-L1<br />

interaction is a novel immunotherapeutic approach for mRCC. In preliminary<br />

results <strong>of</strong> a phase I trial, the anti-PD-1 monoclonal antibody BMS-<br />

936558 had antitumor activity in pts with advanced tumors, including<br />

objective responses (ORs) in 6 <strong>of</strong> 18 pts with mRCC. Here we describe a<br />

phase I study planned to evaluate the safety, tolerability, and maximum<br />

tolerated dose (MTD) <strong>of</strong> BMS-936558, when combined with sunitinib or<br />

pazopanib in pts with mRCC. Methods: This open-label study will have two<br />

parallel treatment arms (BMS-936558 plus sunitinib and BMS-936558<br />

plus pazopanib) conducted in two parts (dose escalation and dose expansion).<br />

Pts must have received �1 prior systemic therapy to be eligible for<br />

dose escalation. Only treatment-naïve pts will be eligible for dose expansion.<br />

Up to 36 pts (18 per arm) will be treated in the dose-escalation phase.<br />

After determining the MTD <strong>of</strong> BMS-936558, treatment-naïve pts will be<br />

enrolled to expansion cohorts allowing 24 pts to be treated at the MTD <strong>of</strong><br />

each arm. Each treatment cycle will be 6 weeks, with BMS-936558 dosed<br />

on Days 1 and 22 and sunitinib or pazopanib given according to product<br />

label. Adverse events will be graded according to NCI CTCAE v4.0. Disease<br />

will be assessed every 6 weeks for the first four assessments and then every<br />

12 weeks until disease progression. Pts will be treated until unacceptable<br />

toxicity, disease progression, or withdrawal <strong>of</strong> consent. The safety pr<strong>of</strong>ile in<br />

pts treated at the MTD will be used to determine the recommended phase II<br />

study dose <strong>of</strong> BMS-936558 in each combination arm. Secondary objectives<br />

will include OR rate and duration <strong>of</strong> response based on RECIST 1.1.<br />

Exploratory analyses will investigate predictive biomarkers <strong>of</strong> BMS-<br />

936558.<br />

TPS2616 General Poster Session (Board #11A), Mon, 8:00 AM-12:00 PM<br />

A phase Ib/II study testing the safety and efficacy <strong>of</strong> combined inhibition <strong>of</strong><br />

the AKT/PI3K and AR signaling pathways in castration-resistant prostate<br />

cancer: GDC-0068 or GDC-0980 with abiraterone acetate versus abiraterone<br />

acetate. Presenting Author: Roel Peter Funke, Genentech, South<br />

San Francisco, CA<br />

Background: Loss <strong>of</strong> PTEN, leading to activation <strong>of</strong> the AKT/PI3K pathway is<br />

frequent and associated with poor prognosis in prostate cancer. In addition,<br />

AR and AKT/PI3K cross-regulate by reciprocal feedback and combined<br />

inhibition <strong>of</strong> both pathways resulted in improved preclinical efficacy. This<br />

study is designed to evaluate the effect <strong>of</strong> combined inhibition <strong>of</strong> AR<br />

pathway (with abiraterone) and AKT/PI3K pathway (with either GDC-0068<br />

or GDC-0980). GDC-0068 is a potent, selective ATP-competitive inhibitor<br />

<strong>of</strong> AKT1, 2, and 3. Preclinical studies showed that cell and tumor models<br />

with PTEN loss are more likely to be sensitive to GDC-0068. GDC-0068<br />

was generally well tolerated in phase I and a MTD <strong>of</strong> 600 mg daily was<br />

identified. GDC-0980 is a potent pan-inhibitor <strong>of</strong> Class I PI3K and inhibits<br />

wild-type and mutated p110� is<strong>of</strong>orms, as well as mTOR kinase. The<br />

recommended phase II dose (RP2D) for single-agent GDC-0980 is 40 mg<br />

daily. Methods: This study will enroll CRPC patients previously treated with<br />

docetaxel. In phase Ib, the RP2D will be determined separately for<br />

GDC-0068 and GDC-0980 in combination with abiraterone 1000 mg qd<br />

and prednisone 5mg bid. In phase II, patients will be randomized 1:1:1 to<br />

receive GDC-0068 � abiraterone, GDC-0980 � abiraterone, or placebo �<br />

abiraterone. The primary endpoint <strong>of</strong> phase II is PFS measured by PCWG2<br />

in all patients and in patients with PTEN loss. Secondary endpoints include<br />

OS, PSA response rate, ORR, safety, Pharmacokinetics and biomarker<br />

analyses. The effect <strong>of</strong> each treatment on the number <strong>of</strong> circulating tumor<br />

cells will be assessed. Primary and secondary analyses will include all<br />

randomized patients and will be conducted according to assigned treatment<br />

arm. Kaplan-Meier methodology will be used to estimate median PFS<br />

for each arm. Up to 24 patients are planned to be enrolled in phase Ib; 240<br />

patients (80 per arm) are planned for phase II. This study is open for<br />

accrual; to date 2 patients have been enrolled in phase Ib.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


Developmental Therapeutics—<strong>Clinical</strong> Pharmacology and Immunotherapy<br />

TPS2617 General Poster Session (Board #11B), Mon, 8:00 AM-12:00 PM<br />

First-in-human phase I trial <strong>of</strong> NHS-IL12 in advanced solid tumors.<br />

Presenting Author: Joseph W. Kim, Laboratory <strong>of</strong> Tumor Immunology and<br />

Biology, Medical Oncology Branch, National Cancer Institute, National<br />

Institutes <strong>of</strong> Health, Bethesda, MD<br />

Background: NHS-IL12 is an antibody-cytokine conjugate consisting <strong>of</strong> two<br />

heterodimers <strong>of</strong> interleukin (IL)-12, fused to a human monoclonal antibody<br />

specific for DNA-histone H1 complex exposed in tumor necrosis. IL12 is a<br />

proinflammatory cytokine, produced by activated phagocytes and dendritic<br />

cells, that plays a critical role in regulating the transition from innate to<br />

adaptive immunity. Early clinical trials <strong>of</strong> IL12 reported clinical responses<br />

in metastatic renal cell carcinoma, Kaposi sarcoma (50–71% response<br />

rate), T-cell lymphoma (56%), and non-Hodgkin’s lymphoma (21%).<br />

However, systemic toxicity limited further clinical development. NHS-IL12<br />

is designed to reduce toxicity associated with systemic administration <strong>of</strong><br />

IL12 by selectively targeting delivery to necrotic areas <strong>of</strong> tumors. A<br />

fluorescence imaging study <strong>of</strong> NHS-murine (mu)-IL12 in mice with<br />

syngeneic lung carcinoma demonstrated effective targeting by NHSmuIL12.<br />

Immunohistochemistry studies further confirmed effective targeting<br />

<strong>of</strong> tumors by NHS-muIL12. In another study in canines with<br />

spontaneously occurring solid tumors, 2 <strong>of</strong> 11 subjects achieved a partial<br />

response with a single dose. Methods: This is a phase I dose-escalation<br />

study designed to determine the maximum tolerated dose <strong>of</strong> NHS-IL12 and<br />

to define the biologically optimal dose and schedule based on immunologic<br />

(pharmacodynamic) analysis <strong>of</strong> cytokines such as interferon-g, and IL-10,<br />

at multiple times points from 4 hours up to 2 weeks following the<br />

administration <strong>of</strong> NHS-IL12. Eligible patients (pts) include those with<br />

evaluable or measurable solid tumors who have progressive disease on at<br />

least one prior line <strong>of</strong> therapy. Pts with a condition associated with<br />

significant necrosis <strong>of</strong> non-tumor bearing tissue, (e.g., ulcerative colitis),<br />

are excluded. Modified immune-related response criteria are used to assess<br />

response, the major features <strong>of</strong> these criteria being (a) imaging confirmation<br />

<strong>of</strong> both progression and response at least 4 weeks after initial imaging,<br />

and (b) no consideration <strong>of</strong> new lesions unless the total measurable tumor<br />

burden meets the criteria for progressive disease. This trial opened to<br />

accrual in Nov. 2011 and may enroll up to 78 pts.<br />

TPS2619 General Poster Session (Board #11D), Mon, 8:00 AM-12:00 PM<br />

A phase I clinical trial <strong>of</strong> autologous, anti-CD19 gene targeted T cells for<br />

adults with B cell acute lymphoblastic leukemia (B-ALL). Presenting<br />

Author: Marco L. Davila, Memorial Sloan-Kettering Cancer Center, New<br />

York, NY<br />

Background: Complete remission (CR) rates for B-ALL in adults is high<br />

(�80%), but overall survival remains low (approximately 30%). Patients<br />

with relapsed disease have a very poor prognosis with a median survival<br />

measured in months. Therefore, there is a dire need for novel therapies for<br />

adults with relapsed or minimal residual B-ALL. To this end we have<br />

developed a novel T cell therapy for B cell malignancies using patient<br />

derived T cells genetically modified to express the 19-28z chimeric antigen<br />

receptor (CAR), an artificial T cell receptor specific to the CD19 antigen<br />

expressed on most B cell tumors. Human T cells retrovirally modified to<br />

express the 19-28z CAR successfully targets and eradicates B cell tumors<br />

in vitro and systemic human B cell cancers in mice. We have recently<br />

published the results <strong>of</strong> our initial Phase I clinical trial experience<br />

(Brentjens, Rivière et al., Blood 2011) utilizing this technology in adults<br />

with chronic lymphocytic leukemia. These results suggest clinical activity<br />

and demonstrate a persistence <strong>of</strong> 19-28z� T cells that is inversely<br />

associated with tumor burden. These studies suggest that 19-28z� T cells<br />

may be particularly effective in relapsed B-ALL because these tumors<br />

retain a degree <strong>of</strong> chemotherapy sensitivity, which will allow the infusion <strong>of</strong><br />

T cells after salvage chemotherapy when patients have low tumor burdens.<br />

To our knowledge, this is the first clinical trial using autologous CD19<br />

targeted T cells in adults with B-ALL. Methods: We are enrolling patients to<br />

a Phase I T cell dose escalation trial (NCT01044069). Adults with CD19�<br />

B-ALL classified as a CR, relapsed, or refractory are eligible for enrollment.<br />

After patients are enrolled they are leukapheresed to obtain peripheral<br />

blood T cells, which are then retrovirally transduced to express the 19-28z<br />

CAR and expanded in our GMP gene transfer facility. Enrolled patients with<br />

relapsed or refractory B-ALL undergo a cycle <strong>of</strong> re-induction chemotherapy<br />

and conditioning cyclophosphamide followed by infusion <strong>of</strong> autologous<br />

19-28z� T cells. To date, 11 patients have been enrolled and 2 patients<br />

have been infused with 19-28z� T cells.<br />

171s<br />

TPS2618 General Poster Session (Board #11C), Mon, 8:00 AM-12:00 PM<br />

Safety, pharmacokinetics, and antitumor activity <strong>of</strong> MEDI-573, an investigational<br />

monoclonal antibody that targets IGF-I and IGF-II, in adult<br />

patients with advanced solid tumors. Presenting Author: Paul Haluska,<br />

Mayo Clinic, Rochester, MN<br />

Background: MEDI-573 is an investigational dual-targeting human antibody<br />

that neutralizes IGF-I/-II ligands and inhibits IGF-1R and insulin<br />

receptor-A (IR-A) signaling pathways. By sparing IR-B and its hybrid<br />

receptors, MEDI-573 may achieve antitumor activity without perturbing<br />

glucose homeostasis. Objectives <strong>of</strong> this phase 1 dose-escalation and<br />

expansion study are to evaluate safety and determine the maximum<br />

tolerated dose (MTD), optimal biologically effective dose, pharmacokinetics,<br />

pharmacodynamics, and immunogenicity <strong>of</strong> MEDI-573 in adult patients<br />

with solid tumors. Methods: Patients with advanced solid tumors,<br />

Karn<strong>of</strong>sky status �60, and adequate organ function were treated with<br />

escalating doses <strong>of</strong> MEDI-573 IV once weekly (0.5 to 15 mg/kg) or every 3<br />

weeks (30 or 45 mg/kg q3w) using a 3�3 design. Three patients per dose<br />

level were evaluated for safety. A biomarker-rich dose-expansion arm tested<br />

weekly 5 or 15 mg/kg MEDI-573 in patients with advanced bladder cancer.<br />

Results: Preliminary data are presented for 43 patients (25 M, 18 F),<br />

median age 64 years. No dose-limiting toxicities or serious toxicity patterns<br />

occurred at any dose level. Drug-related adverse events occurring in �10%<br />

<strong>of</strong> patients were fatigue (28%), decreased appetite (23%), nausea (16%),<br />

diarrhea (14%), and anemia (12%); the majority were � grade 2.<br />

Perturbations in insulin and growth hormone were not observed. <strong>Clinical</strong>ly<br />

significant changes in serum glucose were rare. The MTD was not reached<br />

after completely enrolling patients through the highest dose, 45 mg/kg IV<br />

q3w. Serum exposure <strong>of</strong> MEDI-573 increased with increasing dose. Full<br />

suppression <strong>of</strong> IGF-I/-II was achieved with weekly and q3w dosing. No<br />

neutralizing antibodies against MEDI-573 were detected. Of 43 patients, 8<br />

(19%) had stable disease �12 weeks. The longest exposure was 21 months<br />

in a patient with well-differentiated liposarcoma. Conclusions: MEDI-573<br />

demonstrated an acceptable safety pr<strong>of</strong>ile at all doses tested. No doselimiting<br />

toxicity or serious toxicities, including those related to glucose<br />

homeostasis, were detected. These preliminary results support further<br />

clinical development.<br />

TPS2620 General Poster Session (Board #11E), Mon, 8:00 AM-12:00 PM<br />

A phase I study <strong>of</strong> 1-methyl-D-tryptophan in combination with docetaxel in<br />

metastatic solid tumors. Presenting Author: Erica Jackson, University <strong>of</strong><br />

South Florida Morsani College <strong>of</strong> Medicine, Tampa, FL<br />

Background: The indoleamine 2, 3 dioxygenase pathway (IDO) can create<br />

immune suppression and unresponsiveness to tumor antigens in tumorbearing<br />

hosts. 1-methyl-D-tryptophan (1-MT), an oral inhibitor <strong>of</strong> the IDO<br />

pathway, showed favorable toxicity pr<strong>of</strong>ile and biologic activity in prior<br />

studies. Remarkably, in prior animal models (MMTV-neu mice), 1-MT in<br />

combination with chemotherapy produced 30% greater tumor regressions.<br />

Based on this data, a phase I trial was initiated to study the synergism <strong>of</strong><br />

1-MT with docetaxel. The primary goal <strong>of</strong> this trial is to determine the MTD<br />

and toxicity for the combination <strong>of</strong> docetaxel and oral 1-MT. A secondary<br />

endpoint will be to determine the PK data and the overall response rate.<br />

Methods: This phase I study utilizes a 3�3 design comprised <strong>of</strong> five dose<br />

levels. Dose levels 1-4 will evaluate docetaxel 60mg/m2 IV d1 q3wks plus<br />

1-MT at 300mg, 600mg, 1000mg, and 2000mg PO BID d1-21 respectively.<br />

Dose level 5 is docetaxel 75mg/m2 IV d1 q3wks � 1-MT 2000mg<br />

PO BID d1-21. Eligibility for this study includes patients with measurable<br />

metastatic solid malignancy, no prior docetaxel for metastatic disease, age<br />

�18, life expectancy �4 months, and adequate organ/marrow function.<br />

Patients will be excluded if they meet any <strong>of</strong> the following criteria:<br />

chemotherapy within the past 3 weeks, untreated brain metastases, active<br />

autoimmune disease, or GI disease causing malabsorption. In addition, any<br />

patients who have received prior immunotherapy such as ipilimumab are<br />

excluded. Treatment will continue until disease progression, unacceptable<br />

toxicity, or patient/physician discretion. Accrual to dose level 3 is complete<br />

and dose level 4 accrual is underway. The PK <strong>of</strong> 1-MT and docetaxel will<br />

also be characterized, using an HPLC assay. PK measurements for 1-MT<br />

are drawn on C1D1 after a single dose <strong>of</strong> 1-MT is given and then on C1D8<br />

after the morning dose <strong>of</strong> 1-MT is given. (Drawn at 0,1,2,4,8,12,24, and<br />

48 hours) Because IDO is hypothesized to cause regulatory T cell<br />

expansion, circulating Tregs will be quantified utilizing flow cytometry for<br />

CD4�CD25� FoxP3� cells. (NCT01191216)<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


172s Developmental Therapeutics—<strong>Clinical</strong> Pharmacology and Immunotherapy<br />

TPS2621 General Poster Session (Board #11F), Mon, 8:00 AM-12:00 PM<br />

Safety, pharmacokinetics, and antitumor activity <strong>of</strong> MEDI3617, a selective<br />

angiopoietin inhibitor, in adult patients with advanced solid tumors.<br />

Presenting Author: Ronald B. Natale, Cedars-Sinai Outpatient Cancer<br />

Center, Los Angeles, CA<br />

Background: MEDI3617 is an investigational monoclonal antibody that<br />

inhibits angiogenesis by preventing the interaction <strong>of</strong> angiopoietin-1 and<br />

angiopoietin-2 (Ang2) ligands with the Tie2 receptor. Methods: This is an<br />

ongoing phase 1 dose-escalation and dose-expansion study in patients with<br />

advanced solid tumors, Karn<strong>of</strong>sky performance status �70, and adequate<br />

organ function. Patients were treated with escalating IV doses <strong>of</strong> MEDI3617<br />

on day 1 <strong>of</strong> each 21 day cycle in cohorts <strong>of</strong> 3-6 patients. The objectives are<br />

to evaluate the safety pr<strong>of</strong>ile and determine the maximum tolerated dose,<br />

dose-limiting toxicities (DLT), pharmacokinetics, pharmacodynamics, and<br />

clinical activity <strong>of</strong> MEDI3617. Results: Preliminary data are presented for<br />

21 patients (14M/7F), median age 65 yrs, evaluable for DLT. One DLT<br />

occurred: grade 4 neutropenia lasting more than 7 days. The most<br />

frequently reported drug-related adverse events (% <strong>of</strong> patients) were<br />

fatigue (24%), diarrhea (19%), nausea (19%), dysgeusia (14%), and<br />

headache (14%), all <strong>of</strong> which were � grade 2. All monotherapy dose levels<br />

have completely enrolled patients without exceeding the maximum tolerated<br />

dose. The exposure <strong>of</strong> MEDI3617 after the first dose showed a more<br />

than dose-proportional increase. Suppression <strong>of</strong> free Ang2 was dosedependent.<br />

Maximum accumulation <strong>of</strong> total Ang2 was observed at the<br />

lowest administered dose. No anti-MEDI3617 neutralizing antibodies were<br />

detected. Of the 21 patients, 4 continue on treatment (maximum <strong>of</strong> 6�<br />

months in a patient with carcinoid tumor <strong>of</strong> the jejunum) as <strong>of</strong> the data<br />

cut-<strong>of</strong>f date. Of the 17 patients for whom response data are available, 5 had<br />

stabilization <strong>of</strong> disease <strong>of</strong> 12 weeks or greater. Conclusions: Preliminary<br />

safety and activity signals warrant continued evaluation <strong>of</strong> MEDI3617. This<br />

study was funded by MedImmune, LLC.<br />

TPS2622 General Poster Session (Board #11G), Mon, 8:00 AM-12:00 PM<br />

<strong>Clinical</strong> pharmacokinetics (PK) <strong>of</strong> BMS-936558, a fully human anti-PD-1<br />

monoclonal antibody. Presenting Author: Shruti Agrawal, Bristol-Myers<br />

Squibb, Princeton, NJ<br />

Background: BMS-936558 is a fully human IgG4 monoclonal antibody<br />

targeted against human Programed Death-1 (PD1) receptor on activated T<br />

and B lymphocytes. Blocking <strong>of</strong> PD-1 prevents these activated cells from<br />

becoming anergic, thereby maintaining anti-tumor immunologic activity.<br />

Methods: The PK <strong>of</strong> BMS-936558 was characterized with data from a<br />

single ascending dose (SAD) and a multiple ascending dose (MAD) study in<br />

subjects with advanced solid malignancies. Subjects received a single IV<br />

infusion <strong>of</strong> 0.3 to 10 mg/kg BMS-936558 in the SAD study (MDX-1106-01<br />

Study) and 0.1 to 10 mg/kg BMS-936558 every two weeks in the MAD<br />

study (MDX-1106-03/CA209003 Study). The PK <strong>of</strong> BMS-936558 was<br />

characterized by non-compartmental analysis <strong>of</strong> intensively sampled PK<br />

data from a SAD study, as well as by population PK analysis <strong>of</strong> available<br />

intensive and sparse PK data from the SAD and MAD studies, respectively.<br />

Results: The PK <strong>of</strong> BMS-936558 is linear in the studied dose range with<br />

dose-proportional increase in Cmax and AUC with low to moderate<br />

(20-44%) inter-subject variability. Geometric mean clearance ranged from<br />

0.13 - 0.19 mL/h/kg, whereas mean volume <strong>of</strong> distribution ranged from 83<br />

-113 mL/kg. The range <strong>of</strong> mean terminal elimination half-life <strong>of</strong> BMS-<br />

936558 is 17 to 25 days which is consistent with half-life <strong>of</strong> endogenous<br />

IgG4. BMS-936558 PK was adequately described by a linear twocompartment<br />

model, and there was no evidence <strong>of</strong> a contribution <strong>of</strong> target<br />

mediated drug disposition to drug elimination within tested dose range.<br />

Body weight and gender are potentially clinically significant covariate for<br />

both clearance and volume <strong>of</strong> distribution (� 20% effect); and baseline<br />

CRP, LDH, and albumin are potentially clinically significant covariates for<br />

CL. The body weight normalized dosing employed produces trough concentrations<br />

that are approximately constant over a wide range <strong>of</strong> body weights.<br />

Conclusions: The pharmacokinetics <strong>of</strong> BMS-936558 is dose-proportional<br />

and body weight normalized dosing is appropriate to ensure consistent<br />

exposure over a wide range <strong>of</strong> body weights.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


3000 Oral Abstract Session, Sun, 9:45 AM-12:45 PM<br />

Phase I clinical and pharmacologic study <strong>of</strong> the focal adhesion kinase<br />

(FAK) inhibitor GSK2256098 in pts with advanced solid tumors. Presenting<br />

Author: Jean-Charles Soria, Institut Gustave Roussy, INSERM U981,<br />

Villejuif, France<br />

Background: FAK has an important role in cancer invasion and metastasis.<br />

FAK and phospho-FAK (pFAK) are increased in advanced cancer and are<br />

correlated with poor prognosis. GSK2256098 is a potent and specific small<br />

molecule inhibitor <strong>of</strong> FAK. Methods: This is a first-time in cancer pts, dose<br />

escalation study (NCT01138033) to identify the maximum tolerated dose<br />

(MTD), safety, pharmacokinetics (PK), pharmacodynamics (PD), and<br />

clinical activity. <strong>Part</strong> 1 used a modified accelerated titration procedure<br />

switching to standard 3�3 design based on toxicity during the first 21 days<br />

<strong>of</strong> treatment. Pts were treated with GSK2256098 orally twice daily (BID)<br />

with food. Safety was established in cohorts <strong>of</strong> 1-6 pts. After MTD<br />

determination, <strong>Part</strong>s 2 and 3 were opened to further evaluate safety, PK<br />

and PD. Serial PK samples were obtained over 24 h on Days 1 and 15. Preand<br />

post-dose skin, hair and tumor tissue biopsies were analyzed for pFAK.<br />

Results: 57 pts with advanced solid tumors have been treated at escalating<br />

doses ranging from 80 mg to 1500 mg BID: median age�58.8 (range<br />

21-84). The most common tumor types include: mesothelioma (23), ovary<br />

(7), colon (3), kidney (3), and pancreas (3). MTD was determined to be<br />

1000 mg BID. DLTs were Gr 2 proteinuria (1000 mg), Gr 2 nausea,<br />

vomiting, fatigue (1250 mg) and Gr 3 asthenia (1500 mg). Most frequent<br />

toxicities were nausea (32 pts, 56%), diarrhea (31 pts, 54%), vomiting (25<br />

pts, 44%), decreased appetite (18 pts 32%) and asthenia (11 pts, 19%).<br />

The majority <strong>of</strong> toxicities were Gr 1-2; Gr 3 drug-related events included<br />

hypertriglyceridemia (n�2), hyperlipasemia (2), asthenia (1), increased<br />

amylase (1), and loss <strong>of</strong> consciousness (1). Few dose reductions and<br />

interruptions have occurred. Minor responses/stable disease (SD) were<br />

observed in pts with mesothelioma (-12%, 27 wks), melanoma (-26%, 54<br />

wks), and naso/pharyngeal cancer (-31% target lesion, 30 wks) and SD in<br />

renal cell (48� wks). In preliminary analysis <strong>of</strong> single-dose PK, exposure<br />

generally increased in a dose-proportional manner, and the geometric mean<br />

t½ was 4.2 h. Accumulation was not observed with repeat dosing.<br />

Conclusions: GSK2256098 is well tolerated with evidence <strong>of</strong> clinical<br />

activity. PK supports BID dosing.<br />

3002 Oral Abstract Session, Sun, 9:45 AM-12:45 PM<br />

Phase I study <strong>of</strong> OPB51602, a small molecule inhibitor <strong>of</strong> STAT3<br />

phosphorylation, in patients with refractory solid malignancies. Presenting<br />

Author: Boon C. Goh, Cancer Science Institute <strong>of</strong> Singapore, Singapore<br />

Background: STAT3 is constitutively activated by growth signaling pathways<br />

in many malignancies; in many cell lines inhibition <strong>of</strong> STAT3 leads to cell<br />

death. OPB51602 is a small molecule inhibitor <strong>of</strong> STAT3 phosphorylation<br />

(Tyr705) and activation. Methods: A phase I study <strong>of</strong> OPB51602 administered<br />

for 2 weeks every 3 weeks was initiated to determine the maximum<br />

tolerable dose (MTD), evaluate pharmacokinetics (PK), and assess pharmacodynamics<br />

effects on STAT3 in peripheral blood mononuclear cells<br />

(PBMCs). The starting dose was 2mg/day, and dose escalations to 5mg/d,<br />

10mg/d, 20mg/d, were planned. Single dose PK was done on the first day <strong>of</strong><br />

administration for 4 days. Dose escalation was based on the “3�3” design,<br />

MTD was defined as the dose with at least 2/6 dose limiting toxicities<br />

(DLTs) in the first cycle and toxicities were graded by NCICTCv4.0. Results:<br />

32 patients (pt) were treated at doses <strong>of</strong> 2mg/d (n�7), 4mg/d (n�18),<br />

5mg/d (n�7). The main toxicities observed included nausea/vomiting,<br />

diarrhea, peripheral neuropathy and fatigue. 5 mg/d was the MTD, where<br />

cycle 1 DLTs <strong>of</strong> grade 3 diarrhea/dehydration and hyponatremia occurred in<br />

1 patient respectively. One pt developed grade 3 peripheral neuropathy at<br />

4mg/d cohort. PK showed maximal drug levels 2-3 hours after administration,<br />

bi-exponential decay, with mean oral clearance <strong>of</strong> 316.5 �/- 638.9<br />

L/h and long terminal mean half-life <strong>of</strong> 61.8 �/- 15.9 h on day 17. STAT3<br />

phosphorylation in PBMCs assessed in 6 pts receiving 4mg/d was reduced<br />

from 67.4 �/- 17.4% at baseline to 53.0 �/- 18.1% (p�0.001) after<br />

administration on day 1. Interestingly, reduction in tumor metabolism by<br />

PET CT on day 15 was observed in 4/8 pts receiving 4mg/d. A pt with<br />

heavily pretreated adenocarcinoma <strong>of</strong> lung at 5mg/d dose had partial<br />

response; another pt with metastatic endometrial cancer at 4mg/d dose<br />

experienced stable disease for 6 cycles. Conclusions: OPB51602 has an<br />

MTD <strong>of</strong> 5mg/d on this schedule, demonstrates inhibition <strong>of</strong> STAT3<br />

phosphorylation, and evidence <strong>of</strong> clinical activity. Further pro<strong>of</strong> <strong>of</strong> concept<br />

studies <strong>of</strong> OPB51602 are warranted.<br />

Developmental Therapeutics—Experimental Therapeutics<br />

173s<br />

3001 Oral Abstract Session, Sun, 9:45 AM-12:45 PM<br />

A first-in-human phase I study <strong>of</strong> the oral p38 MAPK inhibitor LY2228820<br />

dimesylate in patients with advanced cancer. Presenting Author: Matthew<br />

P. Goetz, Mayo Clinic , Rochester, MN<br />

Background: p38 MAPK regulates production <strong>of</strong> cytokines by the tumor<br />

microenvironment and its activation enables cancer cells to survive in the<br />

presence <strong>of</strong> oncogenic stress, radiation, chemotherapy, and targeted<br />

therapies. LY2228820 is a selective small-molecule inhibitor <strong>of</strong> p38<br />

MAPK and preclinical studies demonstrate antitumor activity as a single<br />

agent and in combination with standard agents. We performed a phase I<br />

study to determine the maximum tolerated dose (MTD) and dose-limiting<br />

toxicity (DLT) <strong>of</strong> LY2228820 and to characterize its pharmacokinetics and<br />

pharmacodynamics. Methods: Dose escalation was performed in a 3�3<br />

design. LY2228820 was taken orally every 12 hours on days 1-14 <strong>of</strong> a<br />

28-day cycle. Results: 54 patients received either capsules at 8 dose levels<br />

(10, 20, 40, 65, 90, 120, 160, and 200mg) or tablets at 5 dose levels<br />

(160, 200, 300, 420, and 560mg). For both formulations, Cmax and AUC<br />

increased in a dose-dependent manner. LY2228820 inhibited p38 MAPK<br />

induced phosphorylation <strong>of</strong> MAPKAP-K2 in peripheral blood with dosedependent<br />

maximum inhibition from 10 to 70% across the dose range<br />

10-200mg. The most common drug-related adverse events included<br />

fatigue, nausea, rash, constipation, vomiting, and pruritus. 1 patient<br />

(200mg) had DLT <strong>of</strong> erythema multiforme (Gr3) and 2 patients (560mg)<br />

had DLT <strong>of</strong> ataxia (Gr3) and dizziness (Gr2), respectively. Although the<br />

MTD was 420mg, the frequency <strong>of</strong> Gr1/2 adverse events (mainly rash,<br />

dizziness, and tremor) and observation <strong>of</strong> clinical activity at lower dose<br />

levels led to a recommended dose <strong>of</strong> 300mg (mean AUC0-24 � 11.7ughr/ml<br />

at steady state). Early clinical activity has been observed in ovary,<br />

breast, and kidney cancers. One patient with metastatic clear cell carcinoma<br />

<strong>of</strong> the kidney refractory to sorafenib, sunitinib, and temsirolimus had<br />

confirmed near partial response (29% decrease) after 8 cycles and remains<br />

on therapy. 15 patients (28%) achieved best overall response <strong>of</strong> stable<br />

disease, which in 12 patients (22%) was prolonged (�4 cycles). Conclusions:<br />

LY2228820 demonstrates acceptable pharmacokinetics, safety, and early<br />

clinical activity as a single agent in advanced cancer. A phase II study for<br />

patients with ovary cancer is planned.<br />

3003 Oral Abstract Session, Sun, 9:45 AM-12:45 PM<br />

A phase lb, open-label, multicenter, dose-escalation study <strong>of</strong> the oral<br />

pan-PI3K inhibitor BKM120 in combination with the oral MEK1/2 inhibitor<br />

GSK1120212 in patients (pts) with selected advanced solid tumors.<br />

Presenting Author: Philippe Bedard, Division <strong>of</strong> Medical Oncology &<br />

Hematology, Princess Margaret Hospital, Department <strong>of</strong> Medicine, University<br />

<strong>of</strong> Toronto, Toronto, ON, Canada<br />

Background: MAPK and PI3K/AKT signaling pathways regulate proliferation,<br />

differentiation and cell death in human cancers. Known interaction<br />

between the 2 pathways provides the rationale for combining both<br />

inhibitors in a phase I study. Methods: The objective is to determine the<br />

maximum tolerated dose (MTD) and/or recommended phase II dose (RP2D)<br />

for oral, daily administered, BKM120 � GSK1120212, mainly in pts with<br />

tumors with RAS/RAF mutations (mt). A Bayesian logistic regression model<br />

with overdose control guides the dose escalation <strong>of</strong> the treatment. Secondary<br />

objectives include safety, tolerability, PK and efficacy. Results: As <strong>of</strong><br />

22.09.11, 49 pts were treated with BKM120 � GSK1120212 as follows:<br />

30mg � 0.5mg, 60mg � 0.5mg, 60mg � 1.0mg, 60mg � 1.5mg, 60mg<br />

� 2.0mg, 70mg � 1.5mg, 80mg � 1.0mg, 80mg � 1.5mg. 6 pts had<br />

dose-limiting toxicities (DLTs); all were reversible. Grade 3 DLTs were: 3 x<br />

stomatitis, 1 x dysphagia, 1 x LVEF decrease, 1xCKincrease, 1 x nausea,<br />

1 x anorexia, 1 x decreased oral intake. MTD and/or RP2D for the<br />

combination have not been reached. Most common adverse events (AEs)<br />

(�25%), all grades and causality, were dermatitis acneiform, diarrhea<br />

(51% each); nausea (41%); vomiting (37%); rash (33%); asthenia (31%);<br />

CK increase, decreased appetite, pyrexia or stomatitis (29% each) and<br />

hyperglycemia (27%). There were 4 on-treatment deaths unrelated to<br />

treatment. AEs led to treatment discontinuation, 17 pts (35%) and<br />

interruptions/dose reductions, 25 pts (51%). Apparent steady-states <strong>of</strong><br />

BKM120 and GSK1120212 were reached by day 28. Plasma concentrations<br />

<strong>of</strong> BKM120 in combination with GSK1120212 were lower than for<br />

monotherapy. Exposure to GSK1120212 with BKM120 was similar to that<br />

observed in monotherapy studies. 3 confirmed partial responses have been<br />

observed in pts with KRAS mt ovarian cancer; 2 lasting �9 months. 2<br />

patients with BRAF mt melanoma, who had previously progressed on BRAF<br />

inhibitors, had stable disease, for 1 <strong>of</strong> whom treatment is still ongoing in<br />

cycle 6. Conclusions: BKM120 and GSK1120212 can be safely combined.<br />

Signs <strong>of</strong> clinical activity have been seen in pts with RAS/RAF mt tumors.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


174s Developmental Therapeutics—Experimental Therapeutics<br />

3004 Oral Abstract Session, Sun, 9:45 AM-12:45 PM<br />

First-in-human phase I trial <strong>of</strong> the dual mTORC1 and mTORC2 inhibitor<br />

AZD2014 in solid tumors. Presenting Author: Udai Banerji, Drug Development<br />

Unit, The Institute <strong>of</strong> Cancer Research/The Royal Marsden Hospital,<br />

Sutton, United Kingdom<br />

Background: AZD2014 is a potent, dual mTORC1/mTORC2 inhibitor with<br />

clear activity in in vivo and in vitro experimental models. Methods: This<br />

2-part study consisted <strong>of</strong> �rolling six� dose escalation (<strong>Part</strong> A) and<br />

expansion (<strong>Part</strong> B) phases. <strong>Part</strong> A: 3–6 pts per cohort received an oral<br />

solution <strong>of</strong> AZD2014 BD starting at 50 mg. A further 6 pts were treated in<br />

<strong>Part</strong> A below the MTD to study changes in pharmacodynamic (PD)<br />

biomarkers. <strong>Part</strong> B: additional pts were dosed at the MTD, including a<br />

group <strong>of</strong> ER�/PR� or HER2� patients with breast cancer. Primary<br />

endpoint: safety and tolerability; secondary endpoints: pharmacokinetics<br />

(PK), PD and efficacy. Biomarkers assessed: mTORC1: pS6 (S235/236)<br />

and p4EBP1 (T37/46); mTORC2: pAKT (S473). Results: 50 pts have been<br />

enroled in this ongoing study and interim data are reported: <strong>Part</strong> A, n�23<br />

(25 mg, n�6; 50 mg, n�8; 70 mg, n�5; 100 mg, n�4; all BD); <strong>Part</strong> B,<br />

n�27. The MTD was 50 mg BD. DLTs were seen at both 100 mg (Gr 2 and<br />

3 lethargy/fatigue, n�4/4) and 70 mg (Gr 3 mucositis, Gr 2 lethargy,<br />

n�2/4); no DLTs were seen at 25 mg or 50 mg. The most common AEs in<br />

order <strong>of</strong> incidence were fatigue, stomatitis, decreased appetite, nausea and<br />

diarrhea. Seven SAEs (nausea, vomiting, lethargy, abdominal pain, mucositis)<br />

reported by 3 pts were considered ‘possibly related’ to the study drug by<br />

investigators. AZD2014 is rapidly absorbed following single and BD<br />

multiple doses with a short t1/2 <strong>of</strong> ~3 h. At 50 mg (n�32), preliminary data<br />

show geometric mean AUCss�7.4 �g.h/ml and Cmax ss�1.7 �g/ml. One pt<br />

with acinar pancreatic cancer had a RECIST partial response. pAKT and<br />

p4EBP1 reductions were observed between 2–8 h in platelet-rich plasma<br />

and peripheral blood mononuclear cells respectively. Target modulation in<br />

paired tumor biopsies was seen at the MTD. Reduction in the phosphorylation<br />

<strong>of</strong> S6 and 4EBP1 was evident in 8/10 and 3/9 paired biopsies<br />

respectively. pAKT was reduced in 3/6 evaluable paired biopsies. As<br />

opposed to rapalogs, pAKT was not upregulated in any <strong>of</strong> the evaluable<br />

post-treatment biopsies. Conclusions: The MTD for AZD2014 is 50 mg BD.<br />

Further clinical evaluation <strong>of</strong> AZD2014 is now warranted based on the<br />

safety, PK and pro<strong>of</strong>-<strong>of</strong>-mechanism PD data, as well as its preliminary<br />

clinical activity. Updated results will be presented.<br />

3006^ Oral Abstract Session, Sun, 9:45 AM-12:45 PM<br />

Phase I trial <strong>of</strong> an oral TORC1/TORC2 inhibitor (CC-223) in advanced solid<br />

and hematologic cancers. Presenting Author: Kent C. Shih, Sarah Cannon<br />

Research Institute, Nashville, TN<br />

Background: The mammalian target <strong>of</strong> rapamycin (mTOR) signaling pathway<br />

is frequently activated in cancer. The mTOR kinase exists in two<br />

multi-protein complexes, TORC1 and TORC2, which drive key cellular<br />

metabolic and proliferative functions. TORC1-selective inhibitors can<br />

induce feedback upregulation <strong>of</strong> TORC2 and treatment resistance. CC-223<br />

is an oral, potent, selective, ATP-competitive inhibitor <strong>of</strong> both TORC1 and<br />

TORC2, selected to address this escape mechanism. Methods: Subjects<br />

with advanced solid and hematologic cancers were enrolled using an<br />

accelerated (1�5) dose escalation design. CC-223 was administered<br />

orally once daily (QD) in 28 day cycles until disease progression. Safety,<br />

pharmacokinetic, pharmacodynamic and clinical endpoints were evaluated.<br />

Results: 28 subjects were treated across 5 dose levels: 7.5 (n�1), 15<br />

(n�2), 30 (n�9), 45 (n�7) and 60 mg (n�8). The most common<br />

(� 20%) related adverse events (all grades) were fatigue (64%), nausea<br />

(50%), hyperglycemia and diarrhea (43% each), mucositis (39%), anorexia<br />

and vomiting (32% each) and rash (29%). Dose-limiting toxicity (all<br />

grade 3) occurred in 4 subjects: hyperglycemia (30 mg), rash (45 mg),<br />

fatigue (60mg), and mucositis (60 mg). The maximum tolerated dose<br />

(MTD) was 45 mg QD. Dose proportional exposure was observed with a<br />

terminal half life <strong>of</strong> 4 to 8 hrs (mean steady state Cmax 485 ng/mL, AUC0-24 2371 ngxhr/mL at 45 mg). Inhibition <strong>of</strong> TORC1 (pS6, p4EBP1) and<br />

TORC2 (pAKT) biomarkers in blood cells was characterized to be exposuredependent<br />

and described by an Emax model. Near maximal inhibition <strong>of</strong><br />

both TORC1 and TORC2 biomarkers was achieved at the peak concentrations<br />

<strong>of</strong> 30 or 45mg QD. Target inhibition was predicted to last 8 to 20<br />

hours at 45mg QD. Tumor responses included: 1 partial response lasting 9<br />

months (ER� breast) and 7 subjects with stable disease (SD) lasting 8�<br />

weeks (range 8 to 23.3). Conclusions: CC-223 was well tolerated with<br />

toxicities comparable to other drugs in this class. Evidence <strong>of</strong> TORC1/<br />

TORC2 pathway inhibition was observed as well as preliminary signals <strong>of</strong><br />

anti-tumor activity, including one durable PR. Expansion cohorts in<br />

selected hematologic and solid tumors will evaluate CC-223 at the MTD <strong>of</strong><br />

45 mg QD.<br />

3005 Oral Abstract Session, Sun, 9:45 AM-12:45 PM<br />

First-in-human phase I study <strong>of</strong> LY2780301, an oral P70S6K/AKT<br />

inhibitor, in patients with refractory solid tumors. Presenting Author:<br />

Emiliano Calvo, START Madrid, Centro Integral Oncológico Clara Campal,<br />

Madrid, Spain<br />

Background: LY2780301is an oral potent, highly selective ATP competitive<br />

inhibitor against p70S6 kinase and AKT. We report on safety, pharmacokinetics<br />

(PK), pharmacodynamics (PD), and anti-tumor activity from the<br />

first-in-men study <strong>of</strong> LY2780301 in solid tumors. Methods: Eligible pts<br />

(ECOG�1) had relapsed solid tumors, adequate hematologic, renal, and<br />

hepatic functions. Using a predictive pre-clinical PK/PD model and<br />

non-clinical toxicology data, a total daily dose <strong>of</strong> 100-1000 mg, was<br />

selected. LY2780301 was given either once or twice daily for 28-day cycle<br />

until disease progression. Dose escalation (3�3 design) was based on<br />

CTCAE V4 toxicities. The primary objective was to define the recommended<br />

phase 2 dose <strong>of</strong> LY2780301. Secondary objectives were PK, PD (pS6 in<br />

skin), and preliminary antitumor activity. Results: 32 patients were treated<br />

at dose levels from 100mg – 500mg total daily dose. LY2780301 was well<br />

tolerated at all dose levels No dose-limiting toxicities were observed in<br />

cycle 1. Common toxicities potentially related to study therapy included:<br />

constipation (7pts), fatigue (5pts), nausea (4pts), diarrhea (3pts), and<br />

vomiting (3pts). Grade 3/4 toxicities potentially related to study therapy<br />

were: anemia (3pts), increased ALT/AST (1pt), and increased GGT (1pt).<br />

Average number <strong>of</strong> cycles received across all dose levels was 2. There were<br />

no partial or complete responses; best response observed was prolonged<br />

stable disease for 6.5 cycles. Plasma exposures <strong>of</strong> LY278030 exceeded<br />

predicted efficacious dose in all pts (AUC 0-24H �25000 ng*hr/mL).<br />

Increase in exposure with dose was observed (QD or BID) with significant<br />

inter-pt variability. The exposure increase was less than linear with dose.<br />

Preliminary PK parameters are CL/F 4.5L/h and t1/2 <strong>of</strong> 24 hours. pS6<br />

assessment in skin showed inhibition in � 50% <strong>of</strong> pts receiving 400-500<br />

mg per day. Positive correlation with the exposure <strong>of</strong> the didesmethyl<br />

metabolite was observed. Conclusions: LY2780301displayed favorable<br />

safety pr<strong>of</strong>ile, and high PK exposures in the range <strong>of</strong> efficacious dose. The<br />

recommended dose will be defined in the range between 300 and 500 mg.<br />

Further exploration <strong>of</strong> PK/PD effect in mesothelioma is ongoing.<br />

3007 Oral Abstract Session, Sun, 9:45 AM-12:45 PM<br />

First-in-human phase I study <strong>of</strong> the ALK inhibitor LDK378 in advanced<br />

solid tumors. Presenting Author: Ranee Mehra, Fox Chase Cancer Center,<br />

Philadelphia, PA<br />

Background: LDK378 is a novel, potent and selective small molecule<br />

anaplastic lymphoma kinase (ALK) inhibitor (IC50 0.00015 �M), that does<br />

not inhibit c-MET (IC50 3.2 �M). Tumor regression has been observed in<br />

ALK-driven NSCLC xenografts. A first-in-human, Phase I study is being<br />

conducted to determine the MTD and safety pr<strong>of</strong>ile in patients (pts) with<br />

tumors with ALK rearrangement, amplification or mutation. Other objectives<br />

were safety, PK and antitumor activity in pts with ALK-driven NSCLC,<br />

both naïve to ALK inhibitors and relapsed following previous ALK inhibitor<br />

treatment, and other ALK-positive cancers. Methods: Adult pts with<br />

advanced malignancies harboring a genetic alteration in ALK who progressed<br />

on standard therapy or for whom there was no effective therapy,<br />

were given once daily oral LDK378 on a continuous 21-day schedule. Dose<br />

escalation, starting at 50 mg/day, was guided by a Bayesian logistic<br />

regression model (BLRM) to determine the MTD. Results: At a January 5th 2012 cut<strong>of</strong>f,31 pts (primary site: lung 26 pts; breast 3 pts; other 2 pts;<br />

median age 52 years; 87% ECOG PS 0/1) were enrolled and received<br />

LDK378 at doses <strong>of</strong> 50–750 mg/day. Two dose limiting toxicities, Grade<br />

(Gr) 3 alanine aminotransferase elevation (1 pt), and Gr 3 hypophosphatemia<br />

(1 pt) occurred in 8 pts at a 400 mg dose level. BLRM allowed<br />

dose escalation, and there were no DLTs in 4 pts at 500 mg. Median<br />

duration <strong>of</strong> treatment with LDK378 was 7 weeks (range �1–22�). At the<br />

cut<strong>of</strong>f date, 13 (42%) pts discontinued treatment: 1 (3%) due to adverse<br />

events (AEs), and 12 (39%) due to disease progression; 18 (58%) pts were<br />

still on treatment. The most frequent AEs (all Gr) were nausea (45%),<br />

vomiting (36%), and diarrhea (29%). The most frequent Gr 3/4 AEs were<br />

diarrhea and dyspnea (2 pts [7%] each). At doses �400 mg steady state<br />

exposures exceeded efficacious exposures in xenograft models. Of 16 pts<br />

with available response data (RECIST, per investigator) there were 4/6<br />

responses in crizotinib (CRZ)-treated pts, and 2/10 responses in CRZ-naïve<br />

pts, <strong>of</strong> whom 7 were treated below 400 mg. All responses were in NSCLC.<br />

Conclusions: Daily oral LDK378 is well tolerated up to 500 mg/day, and<br />

escalation continues at 750 mg/day. Preliminary responses have been seen<br />

in both CRZ-naïve and CRZ-relapsed pts.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


3008 Oral Abstract Session, Sun, 9:45 AM-12:45 PM<br />

A first-in-human phase I study <strong>of</strong> the oral Notch inhibitor LY900009 in<br />

patients with advanced cancer. Presenting Author: Shubham Pant, Sarah<br />

Cannon Research Institute/University <strong>of</strong> Oklahoma Health Sciences Center,<br />

Oklahoma City, OK<br />

Background: Notch signaling plays a critical role during stem cell selfrenewal<br />

and is deregulated in multiple human cancers. The Notch pathway<br />

may be activated inappropriately by receptor mutation and overexpression<br />

as well as aberrant signals from the tumor microenvironment. LY900009 is<br />

a selective small-molecule inhibitor <strong>of</strong> gamma secretase, the enzyme that<br />

cleaves and thereby activates Notch receptors. Methods: Dose escalation<br />

was performed in cohorts <strong>of</strong> 3 patients (pts) using a modified continual<br />

reassessment method. LY900009 was taken orally thrice weekly (every<br />

MWF) during a 28-day cycle. Safety, pharmacokinetic, pharmacodynamic,<br />

and clinical endpoints were evaluated. Results: 22 patients received<br />

LY900009 across 6 dose levels: 2mg (3pts), 4mg (4pts), 8mg (3pts),<br />

15mg (3pts), 30mg (6pts), and 60mg (3pts). The most common treatment<br />

emergent adverse events possibly related to LY900009 across all grades<br />

included diarrhea (27%), vomiting (23%), nausea (18%), fatigue (23%),<br />

anorexia (23%), hypophosphatemia (14%), and rash (18%). Dose-limiting<br />

toxicities <strong>of</strong> fatigue/N/V (G3) and diarrhea (G3) were seen in 2 patients,<br />

respectively, treated at 60mg. The maximum tolerated dose (MTD) was<br />

tentatively identified at 30mg. After a single dose, mean Cmax increased<br />

from 4 to 158 ng/ml and mean AUC0-t(last) increased from 14 to 1160<br />

ng-hr/ml. Both Cmax and AUC0-t(last) increased in a dose-dependent<br />

manner. Elimination half-life <strong>of</strong> LY900009 was approximately 2-3 hrs.<br />

LY900009 inhibited plasma levels <strong>of</strong> amyloid-� peptide (a downstream<br />

product <strong>of</strong> gamma secretase) in a dose-dependent manner with 80-90%<br />

inhibition observed in the 30 and 60mg cohorts. In the 15mg cohort, one<br />

patient had colonic biopsy that showed markedly increased glandular<br />

mucin consistent with pharmacologic inhibition <strong>of</strong> the Notch pathway. Two<br />

patients (10%) with leiomyosarcoma and ovarian cancer received 4 cycles<br />

<strong>of</strong> therapy. Conclusions: LY900009 demonstrates acceptable safety and<br />

pharmacokinetics in patients with advanced cancer. Pharmacodynamic<br />

endpoints show pathway inhibition at tolerable doses. One more cohort at<br />

45mg is ongoing to refine the MTD and will be followed by an expansion<br />

cohort for patients with ovarian cancer.<br />

3010 Poster Discussion Session (Board #2), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Phase I/II study <strong>of</strong> the investigational aurora A kinase (AAK) inhibitor<br />

MLN8237 (alisertib) in patients (pts) with non-small cell lung cancer<br />

(NSCLC), small cell lung cancer (SCLC), breast cancer (BrC), head/neck<br />

cancer (H&N), and gastroesophageal (GE) adenocarcinoma: Preliminary<br />

phase II results. Presenting Author: Peter Lee, Tower Cancer Research<br />

Foundation, Beverly Hills, CA<br />

Background: AAK is a key mitotic regulator that is amplified or overexpressed<br />

in multiple tumor types. MLN8237 is an investigational oral,<br />

selective AAK inhibitor being studied in both hematologic (phase III) and<br />

nonhematologic malignancies. In the phase 1 part <strong>of</strong> this study, MLN8237<br />

was generally well tolerated in pts with solid tumors. The maximum<br />

tolerated dose was 50 mg BID for 7d � 2 wks rest in 21-d cycles; this dose<br />

was recommended for phase II evaluation. Here we report preliminary data<br />

from the 5-arm phase 2 part (NCT01045421). Methods: 20 pts each with<br />

one <strong>of</strong> 5 tumor types in the relapsed/refractory setting were enrolled in<br />

phase II for efficacy evaluation: NSCLC (inc. 8–10 pts with mainly<br />

squamous-cell histology), SCLC (inc. 8–12 chemotherapy refractory pts),<br />

BrC (inc. 8–10 pts with a basal-like intrinsic subtype, and 8–10 HER2�<br />

pts), H&N (inc. 8–12 pts with tonsillar or base-<strong>of</strong>-tongue disease), or GE<br />

adenocarcinoma. Pts were aged �18 y, and had ECOG PS 0–1, measurable<br />

disease by RECIST, and �2 prior cytotoxic chemotherapy regimens<br />

(�4 in BrC). Oral MLN8237 was administered as an enteric-coated tablet<br />

at 50 mg BID for 7d � 2 wks rest in 21-d cycles. The primary endpoint was<br />

overall response rate. Response was determined by RECIST v1.1. Each arm<br />

with �2 partial responses (PR) is being expanded with 25 additional pts.<br />

Results: In 100 efficacy-evaluable pts, median age was 59 y (range 33–88)<br />

and 89% were caucasian. Pts received a median <strong>of</strong> 3 cycles (range 1–9).<br />

PRs were observed in 11 pts: H&N, SCLC (each n�3), BrC, GE adenocarcinoma<br />

(each n�2), and NSCLC (n�1). 45 pts had a best response <strong>of</strong> stable<br />

disease. In the safety population (n�196), most frequent grade �3<br />

drug-related AEs were neutropenia (37%), leukopenia (12%), fatigue,<br />

anemia, stomatitis, and thrombocytopenia (each 6%). Updated data will be<br />

presented. Conclusions: Preliminary phase II data suggest that oral<br />

MLN8237 is tolerable and has antineoplastic activity in a range <strong>of</strong> solid<br />

tumors studied. This study is currently ongoing to further evaluate<br />

MLN8237 in the expansion cohorts.<br />

Developmental Therapeutics—Experimental Therapeutics<br />

175s<br />

3009 Poster Discussion Session (Board #1), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

First-in-human study <strong>of</strong> AMG 900, an oral pan-Aurora kinase inhibitor, in<br />

adult patients (pts) with advanced solid tumors. Presenting Author:<br />

Michael Anthony Carducci, The Johns Hopkins University School <strong>of</strong><br />

Medicine and Sidney Kimmel Comprehensive Cancer, Baltimore, MD<br />

Background: Aurora kinases A, B, and, C play essential roles in regulating<br />

cell division. Overexpression <strong>of</strong> aurora A and B in human tumors is<br />

associated with high proliferation rates and poor prognosis. AMG 900 is an<br />

investigational, orally administered, highly selective inhibitor <strong>of</strong> aurora A,<br />

B, and C that demonstrated activity in drug-resistant cell lines and human<br />

tumor xenografts. This study evaluated the safety, tolerability, pharmacokinetics<br />

(PK), and pharmacodynamics <strong>of</strong> AMG 900. Methods: Key eligibility<br />

criteria: � 18 years old, advanced solid tumors, measurable disease, ECOG<br />

� 2, and adequate organ function. AMG 900 was administered orally daily<br />

for 4 consecutive days (D) every 2 weeks (Q2W). In the dose-escalation<br />

phase, the starting dose was 1 mg and was escalated by 100% in<br />

subsequent cohorts (1 pt/cohort) until dose limiting toxicity (DLT) or grade<br />

2 (G2) AMG 900-related toxicity occurred in the first 28 D. Dose escalation<br />

continued in a standard 3�3 design at � 25% if DLT occurred or � 50% if<br />

G2 related toxicity occurred. The maximum tolerated dose (MTD) was<br />

determined without prophylactic G-CSF support. Results: As <strong>of</strong> OCT 2011,<br />

19 pts (1 pt at 1, 2, 4, and 8 mg; 3 pts at 16 mg; and 6 pts at 24 and 30<br />

mg) had received � 1 dose <strong>of</strong> AMG 900. Demographics were 58% women,<br />

median age 60 (32-77) years, and ECOG 0/1, 63%/37%. There were 4<br />

DLTs: G4 neutropenia � 7 days at 24 mg (n�1) and 30 mg (n�1); G4<br />

neutropenia � 7 days � G4 thrombocytopenia at 30 mg (n�1); and febrile<br />

neutropenia (FN) at 30 mg (n�1). MTD without G-CSF support was 24 mg.<br />

89% <strong>of</strong> pts had treatment-related adverse events (AEs). G�3 treatmentrelated<br />

AEs were neutropenia, 8 (42%); leukopenia, 4 (21%); anemia, 2<br />

(11%); thrombocytopenia, 1 (5%); and FN, 1 (5%). Preliminary PK showed<br />

no obvious departures from dose-proportionality with a half-life <strong>of</strong> ~16 h.<br />

Tumor-response data were available for 17 pts: stable disease, 13 pts<br />

(range 0.4 to 43.7 wks); progressive disease, 4 pts. One pt (16 mg cohort)<br />

with recurrent ovarian cancer had 16% tumor shrinkage and 45% decrease<br />

in CA-125 (SD � 6 months). Conclusions: AMG 900 administered at 24 mg<br />

daily for 4D Q2W is the recommended dose for phase 2 trials. Dose<br />

escalation is now ongoing to determine the MTD with prophylactic G-CSF.<br />

3011 Poster Discussion Session (Board #3), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

A phase I study <strong>of</strong> selective cyclin dependent kinase inhibitor P1446A-05<br />

administered on an intermittent schedule in patients with advanced<br />

refractory tumors. Presenting Author: Sudeep Gupta, Tata Memorial<br />

Centre, Mumbai, India<br />

Background: Cyclin-dependent kinases (Cdks) have emerged as important<br />

targets in anticancer drug development. P1446A-05 is a potent and<br />

specific inhibitor <strong>of</strong> Cdk4-D1 (IC50-0.09�M), Cdk1-B (IC50-0.025�M), and Cdk9-T (IC50-0.022�M). This study was designed to determine the<br />

maximum tolerated dose (MTD), dose limiting toxicity (DLT), safety pr<strong>of</strong>ile,<br />

pharmacokinetics, and antitumor activity <strong>of</strong> orally administered P1446A-05<br />

in patients with advanced refractory tumors. Methods: This study was<br />

conducted at 5 centers in India. P1446A-05 was administered in escalating<br />

doses across 5 cohorts <strong>of</strong> eligible pts starting with a dose <strong>of</strong> 75 mg once<br />

a day (OD) for 14 days in a 21 day cycle utilizing a modified Fibonacci<br />

scheme for dose escalation. 10 pts positive for cyclin D1 expression in the<br />

tumor were enrolled at MTD. Treatment was continued until unacceptable<br />

toxicity or disease progression. For pharmacokinetic analysis, blood samples<br />

were collected on days 1 and 13/14 <strong>of</strong> cycle 1 at multiple time points. In<br />

consenting pts, skin and tumor tissue biopsies were collected on days 1<br />

(pre-dose), 8 and 15 <strong>of</strong> cycle 1. Results: A total <strong>of</strong> 29 patients were dosed.<br />

Two DLTs (abdominal pain and acute renal failure) were reported at 850<br />

mg/d. The MTD was 600 mg/d and diarrhea was reported as 1 DLT at this<br />

dose level. Six SAEs including one death related to study drug were<br />

reported. Pharmacokinetic analysis demonstrated a median half-life <strong>of</strong> 16<br />

to 26 h and linear increase in exposure at steady state across tested doses.<br />

Plasma concentration at 300 mg/d dose level crossed efficacy exposure <strong>of</strong><br />

100mg/kg <strong>of</strong> five day dosing in SCID mice. The recommended phase II dose<br />

is 600 mg OD on this schedule. Stable disease for 4 to 6 cycles was<br />

reported in 5 pts. Of them, one patient each with breast cancer, spindle cell<br />

sarcoma in neck, and nasopharyngeal carcinoma had cyclin D1 over<br />

expression. No objective responses were observed in this group <strong>of</strong> heavily<br />

pretreated patients. Conclusions: The safety pr<strong>of</strong>ile <strong>of</strong> P1446A-05 is<br />

considered acceptable. On this dosing schedule, the MTD is determined as<br />

600 mg/day. Further testing <strong>of</strong> P1446A-05 in phase II studies is planned.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


176s Developmental Therapeutics—Experimental Therapeutics<br />

3012 Poster Discussion Session (Board #4), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

A first-in-human phase I study <strong>of</strong> oral pan-CDK inhibitor BAY 1000394 in<br />

patients with advanced solid tumors: Dose escalation with an intermittent 3<br />

days on/4 days <strong>of</strong>f schedule. Presenting Author: Rastislav Bahleda, Institut<br />

Gustave Roussy, Villejuif, France<br />

Background: BAY 1000394 (BAY) is an oral pan-CDK inhibitor targeting<br />

CDKs 1,2,4, 7 and 9 in the low nanomolar range. A phase I dose escalation<br />

study was initiated to determine the maximum tolerated dose (MTD),<br />

pharmacokinetics (PK), and pharmacodynamics (PD) in patients (pts) with<br />

advanced solid tumors. Methods: BAY was administered twice daily in a 3<br />

days on / 4 days <strong>of</strong>f schedule (cycle length 21 days, 3�3 design). PK was<br />

evaluated on cycle 1 day 1 and day 10. Response rate was assessed<br />

according to RECIST 1.1. PD markers included CK18 fragments in plasma.<br />

Results: As <strong>of</strong> Jan 08 2011, 34 pts were treated at doses <strong>of</strong> 0.6 (3 pts), 1.2<br />

(4), 2.4 (3), 4.8 (3), 9.6 (3), 19.2 (6) mg per day as oral solution and at<br />

doses <strong>of</strong> 10 (4), 15 (6) and 20 (2) mg per day as tablet. Tumor types<br />

included 10 colorectal, 4 mesothelioma and 20 others. Cohort 9 (20 mg<br />

tablet) is ongoing. Frequent CTCAEv4 grade 1/2 drug related AEs occurring<br />

in more than 25% <strong>of</strong> patients up to cohort 8 were asthenia, diarrhea,<br />

nausea, vomiting and anorexia. DLTs (grade 3, 1 pt each) were hyponatremia,<br />

aphtous stomatitis at 19.2 mg solution and arterial thrombosis at 15<br />

mg tablet. Aphthous stomatitis (20%) has not been observed with the<br />

tablet formulation. Other grade 3 related AEs were asthenia in 2 and<br />

nausea and vomiting in one pt each. Nausea and vomiting on treatment<br />

days were observed despite antiemetic treatment (aprepitant �/- setron).<br />

PK was dose proportional up to 9.6 mg, T1/2 was 10 hours, and relative<br />

bioavailability <strong>of</strong> tablet formulation was excellent; major metabolite levels<br />

were low (�10%). Levels <strong>of</strong> CK18 fragments did not correlate with dose or<br />

tumor response. Stable disease (SD) lasting for 2-4 months was observed in<br />

9 patients, among others in 4 <strong>of</strong> 4 mesothelioma and 2 <strong>of</strong> 2 ovarian pts.<br />

One additional pt with cholangiocarcinoma has ongoing SD lasting for 5<br />

months. One <strong>of</strong> the ovarian pts had a significant decline <strong>of</strong> CA125 lasting<br />

for 3 months. Conclusions: The tablet formulation <strong>of</strong> BAY 1000394 was<br />

better tolerated than oral solution. So far, doses up to a 15 mg per day with<br />

concomitant antiemetic treatment showed an acceptable tolerability. SD<br />

was observed in 10 <strong>of</strong> 25 heavily pretreated pts across cohorts3–8.<br />

3014 Poster Discussion Session (Board #6), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Molecular pr<strong>of</strong>iling <strong>of</strong> patients (pts) with colorectal cancer (CRC) and<br />

matched targeted therapy (MTA) in phase I clinical trials. Presenting<br />

Author: Rodrigo Dienstmann, Molecular Therapeutics Research Unit, Vall<br />

d’Hebron University Hospital, Barcelona, Spain<br />

Background: Molecular prescreening and biomarker enrichment strategies<br />

in Phase 1 trials with targeted therapies are anticipated to improve the<br />

outcomes <strong>of</strong> affected pts. Methods: As part <strong>of</strong> our personalized oncology<br />

program, tumors from pts with advanced chemorefractory CRC were<br />

analyzed for specific molecular aberrations (KRAS/ BRAF/ PIK3CA mutations<br />

[mut], PTEN and pMET expression) at the Vall d’Hebron Molecular<br />

Pathology and Genomics Labs. Those whose tumors were found to have a<br />

dysregulation were <strong>of</strong>fered a Phase 1 trial with MTA. Results: During 2010<br />

and 2011, tumor molecular analysis was performed in 254 pts: KRAS mut<br />

(80/254, 31.5%), BRAF mut (24/196, 12.2%), PIK3CA mut (15/114,<br />

13.2%), KRAS�PIK3CA mut (9/114, 7.9%), PTEN low (97/183, 53.0% -<br />

HSCORE�50; 45/183, 24.6% - PTEN null), KRAS mut � PTEN low<br />

(38/138, 20.8%), pMET high (38/64, 59.4% with HSCORE�30). In total,<br />

68 pts (median, age 63 yrs; prior therapies 3), received 82 different<br />

matched therapies. Type <strong>of</strong> MTA: PI3K pathway inh (if PIK3CA mut, n�10;<br />

or PTEN low, n�32), MEK�PI3K pathway inh (if KRAS mut, n�10; or<br />

BRAF mut, n�1), second-generation anti-EGFR mAbs (if KRAS wild-type,<br />

n�11), anti-HGF mAb (if pMET high, n�10), mTOR inh � anti-IGFR-1R<br />

mAb (if PTEN low, n�5) and BRAF inh (if BRAF mut, n�3). Median time to<br />

treatment failure (TTF) with MTA was 7.9 weeks (CI95%:7.6-8.1) vs. 16.3<br />

weeks (CI95%:13.9-18.7) for their prior systemic antitumor therapy<br />

(p�0.001). If prior therapy non-standard (according to NCCN guidelines,<br />

n�39), TTF with MTA 7.9 weeks (CI95%:6.8-8.9) vs. TTF with prior<br />

therapy 8.7 weeks (CI95%:7.3-10.1). If an approved standard regimen<br />

(n�43), TTF with MTA 7.9 weeks (CI95%:7.6-8.1) vs. TTF with prior<br />

therapy 21.9 weeks (CI95%:15.0-28.7). <strong>Part</strong>ial response was seen in one<br />

pt (1.2%, PI3K inh with PIK3CA mut) and stable disease � 16 weeks in 10<br />

cases (12.2%). <strong>Clinical</strong> benefit, defined as a TTF ratio � 1.3 (TTF on MTA/<br />

TTF on prior therapy), was seen with 15.9% <strong>of</strong> the therapies (13/82).<br />

Conclusions: Preliminary results suggest that matching chemorefractory<br />

CRC patients with targeted agents in early clinical trials based on the<br />

current molecular pr<strong>of</strong>ile does not result in longer TTF compared to their<br />

prior therapy.<br />

3013 Poster Discussion Session (Board #5), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

A phase I and pharmacokinetic (PK) study <strong>of</strong> continuous daily administration<br />

<strong>of</strong> P1446A-05, a potent and specific oral Cdk4 inhibitor. Presenting<br />

Author: Desiree Hao, Tom Baker Cancer Centre, Calgary, AB, Canada<br />

Background: P1446A-05 is a novel oral inhibitor <strong>of</strong> Cdk4-D1, Cdk1-B, and<br />

Cdk9-T, and has been shown to inhibit tumor growth both in vitro and in vivo.<br />

Pharmacodynamic studies demonstrate that activation <strong>of</strong> Cdk1 reappears<br />

within 48 hours after P1446A-05 is withdrawn, suggesting the need for<br />

prolonged administration hence, we sought to evaluate the feasibility, safety<br />

and tolerability <strong>of</strong> a continuous daily schedule <strong>of</strong> P1446A-05 in patients (pts)<br />

with advanced malignancies. Methods: P1446A-05 was given at escalating<br />

doses <strong>of</strong> 75, 150, 250, 350 and 500mg. Samples were collected for PK at<br />

multiple time points over 24 hours on cycle 1 day 1 and 15, as well as at single<br />

time points on cycle 1 day 8, 22 and cycle 2 day 1. Results: Thirty-nine pts<br />

(median age�63 years, 51% male, 51% ECOG PS�1) collectively received<br />

more than 100 cycles <strong>of</strong> P1446A-05. The majority <strong>of</strong> drug-related toxicities<br />

were �Grade 2, the most common <strong>of</strong> which were diarrhea (n�54), nausea/<br />

vomiting (n�27/17), fatigue (n�22) and anorexia (n�16). Two pts developed<br />

study-drug related diarrhea with hypokalemia/elevated creatinine and died<br />

during cycle 1. Dose-limiting toxicities (DLT) at 500mg (Table) led to<br />

subsequent de-escalation and expansion <strong>of</strong> the 350mg cohort. A total <strong>of</strong> 24<br />

pts were treated at 350mg; only one patient experienced dose-limiting<br />

diarrhea. PK data are summarized below. Accumulation ratios across dose<br />

levels suggest moderate accumulation with continuous dosing. Nine pts<br />

achieved stable disease (SD) for at least 2 cycles. One pt with alveolar s<strong>of</strong>t<br />

tissue sarcoma, whose disease was progressing at enrollment, remains on<br />

treatment with SD after 11 cycles. Conclusions: The recommend phase II dose<br />

<strong>of</strong> P1446A-05 is 350mg. Further phase II studies at this dose will be<br />

conducted with potential enrichment strategies.<br />

DLTs and PK data.<br />

Dose<br />

level<br />

(mg)<br />

No. <strong>of</strong><br />

pts in<br />

cohort<br />

No. <strong>of</strong><br />

pts with<br />

DLT<br />

DLT event<br />

description<br />

Tmax (h) Cmax (ng/mL) AUC0-inf (ng.h/mL)<br />

Accumulation<br />

Day 1 Day 15 Day 1 Day 15 Day 1 Day 15 ratio<br />

75 3 0 4 2 107 195 3,033 24,893 2.22<br />

150 3 0 6 6 255 748 14,753 164,073 3.45<br />

250 3 0 4 4 446 1,027 15,208 54,503 3.07<br />

350 3�9�12 1 Diarrhea 4 4 512 992 16,700 38,099 2.30<br />

500 6 3 Elevated INR<br />

Sudden death<br />

Diarrhea<br />

6 2 1,040 2,239 38,125 76,779 2.43<br />

3015 Poster Discussion Session (Board #7), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Phase I study <strong>of</strong> BPM 31510 in advanced solid tumors: Updated analysis <strong>of</strong><br />

a novel treatment with promising activity. Presenting Author: Andrew<br />

Eugene Hendifar, Sarcoma Oncology Center, Santa Monica, CA<br />

Background: BPM 31510 is a novel small molecule that targets the<br />

metabolic machinery <strong>of</strong> the cancer microenvironment to reverse the aerobic<br />

glycolytic phenotype <strong>of</strong> cancer cells. Effector downstream signaling results<br />

in re-capitulation <strong>of</strong> BCL-2 mediated apoptosis and disruption in tumor<br />

vasculature by modulation <strong>of</strong> VEGF. (NR Narain et al., <strong>Proceedings</strong> <strong>of</strong> AACR<br />

<strong>Meeting</strong> Abstracts 2011). Methods: A standard 3�3 phase I, doseescalation<br />

study design was used in patients with advanced solid tumors<br />

refractory to standard treatment. Primary objectives were establishment <strong>of</strong><br />

the maximum tolerated dose (MTD) and safety/pharmacokinetic (PK)<br />

correlates. Secondary objectives included exploratory pharmacodynamics<br />

(PD) and preliminary efficacy (RECIST-1.1) <strong>of</strong> BPM 31510 in sequential<br />

cohorts <strong>of</strong> 3 to 6 pts. Results: At time <strong>of</strong> submission, 34 patients with<br />

advanced cancer who had failed multiple chemotherapeutic regimens had<br />

been enrolled in 7 dose cohorts (ranging from 5.6 mg/kg to 78.2 mg/kg).<br />

Patients received a median <strong>of</strong> 2 cycles (1-7). 2 patients have had grade 3<br />

elevation in PT/INR, otherwise there have been no grade 3/4 treatment<br />

related toxicities to date. The pharmacokinetics <strong>of</strong> BPM 31510 are linear<br />

and there were no sex differences in the parameters normalized by dose and<br />

body surface area. Tmax and Cmax are associated with the end <strong>of</strong> the<br />

infusion. The values for t1/2 ranged from 2.18 to 13.3 hr, with little or no<br />

dependence <strong>of</strong> t1/2 on dose. Objective tumor responses have been noted at<br />

the dose <strong>of</strong> 58.6mg/kg with 1 partial response (myxoid liposarcoma) and 1<br />

minor response (pleomorphic sarcoma). Six patients (19%) have had<br />

disease stabilization (� 4 months). Conclusions: Interim data from this<br />

phase I study indicate that BPM 31510 is well tolerated with no dose<br />

limiting toxicities to date. A partial response and minor response were<br />

observed and correlates with dose escalation. Taken together, there is<br />

strong rationale for further clinical development <strong>of</strong> this compound as an<br />

anti-cancer agent.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


3016 Poster Discussion Session (Board #8), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

A first-in-human phase I study <strong>of</strong> MORAb-004 (M4), a humanized<br />

monoclonal antibody recognizing endosialin (TEM-1), in patients with solid<br />

tumors. Presenting Author: Luis A. Diaz, Sidney Kimmel Comprehensive<br />

Cancer Center at Johns Hopkins University, Baltimore, MD<br />

Background: Endosialin (TEM-1) is a membrane glycoprotein on the cell<br />

surface <strong>of</strong> activated mesenchymal cells (e.g. pericytes, fibroblasts and<br />

mesenchymal tumor cells) and on a subset <strong>of</strong> human cancers. It is involved<br />

in the development <strong>of</strong> tumor vasculature, stromal/tumor organization and<br />

PDGFRb signaling. M4 is a humanized IgG monoclonal antibody which<br />

targets TEM-1. Preclinical studies suggest that M4 may have single agent<br />

activity via inhibition <strong>of</strong> the tumor microenvironment and by direct effects<br />

on tumors expressing this target. Methods: A phase I dose escalation study<br />

in patients (pts) with advanced solid tumors was conducted to evaluate the<br />

safety, pharmacokinetic pr<strong>of</strong>ile and preliminary efficacy <strong>of</strong> M4 administered<br />

intravenously on a weekly schedule; final results are presented.<br />

Results: 36 pts refractory to therapy were treated at 10 dose levels (0.0625<br />

to 16 mg/kg administered i.v. on a weekly schedule). Drug-related adverse<br />

events (AE) observed were primarily infusion toxicity (grade 1 and 2 fever,<br />

chills, headache, myalgia). The MTD was determined to be 12 mg/kg. DLT<br />

<strong>of</strong> grade 3 vomiting was observed at 16 mg/kg. Preliminary pharmacokinetic<br />

(PK) analyses demonstrate M4 accumulation beginning at 4 mg/kg,<br />

and that elimination mechanisms are saturable beginning at 0.25 mg/kg.<br />

Exposure (AUC) increases in a greater than dose-proportional manner, with<br />

the apparent t1/2 increasing proportionally with dose. Tumor shrinkage<br />

(Minor/mixed tumor responses, mR) were seen in four pts (s<strong>of</strong>t tissue<br />

sarcoma (2), hepatocellular carcinoma, pancreatic neuroendocrine tumor).<br />

Each mR was associated with prolonged disease stabilization in these pts<br />

(6-14 months). A cohort (n�4) <strong>of</strong> pts with refractory colorectal carcinoma<br />

demonstrated disease stabilization (15-24 weeks). Analysis <strong>of</strong> archival<br />

tumor revealed TEM-1 stromal staining in all cases with tumor cell<br />

expression noted on a subset <strong>of</strong> tumors <strong>of</strong> mesenchymal origin. Conclusions:<br />

In this phase I study, M4 was tolerated up to 12 mg/kg (MTD) and early<br />

signs <strong>of</strong> potential activity were observed. Expanded cohorts <strong>of</strong> 1-12 mg/kg<br />

are ongoing to further define an optimal dose.<br />

3018^ Poster Discussion Session (Board #10), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Phase I trial <strong>of</strong> the polo-like kinase (Plk) inhibitor volasertib (BI 6727) combined<br />

with cisplatin or carboplatin in patients with advanced solid tumors. Presenting<br />

Author: Herlinde Dumez, Department <strong>of</strong> General Medical Oncology, University<br />

Hospitals Leuven, Leuven, Belgium<br />

Background: Volasertib (V) is a first in class, selective and potent cell cycle kinase<br />

inhibitor that induces mitotic arrest and apoptosis by targeting Plk. This phase I<br />

study evaluates dose-limiting toxicities (DLT), maximum tolerated dose (MTD),<br />

safety and pharmacokinetics (PK) <strong>of</strong> V combined with cisplatin (Cis) or<br />

carboplatin (Ca). Methods: Sequential cohorts <strong>of</strong> patients (pts) received a single<br />

2h infusion <strong>of</strong> V with Cis (arm A) or Ca (arm B) every 3 wks. Cis and Ca were given<br />

for up to 6 cycles (Cy); V was continued until progression or intolerance. MTD was<br />

the highest dose at which �1/6 pts experienced a DLT in Cy 1. MTD cohorts were<br />

expanded to 12 DLT-evaluable pts to further characterize safety. Results: As <strong>of</strong><br />

January 11 2012, 61 pts (arm A: 30; arm B: 31) were treated. Pt characteristics<br />

were (arm A/B): median age 55/58 yrs, male 16/18 pts, ECOG PS 0: 13/14 pts,<br />

PS 1: 17/17 pts. Tumors included (pts): non-small cell lung cancer (15);<br />

sarcoma (8); colorectal cancer (6); melanoma (4); urothelial cancer (4); other<br />

(24). Pts received V � Cis for a median [range] <strong>of</strong> 3.5 Cy [1-6], V � Ca for 2 Cy<br />

[1-6] and V for 3.5 Cy [1-20] in arm A and 2 Cy [1-14] in B. PK analyses are<br />

ongoing. MTD was reached at V 300 mg � Cis 100 mg/m2 and V 300 mg � Ca<br />

AUC 6. Five partial responses (PR) were seen. 15/30 pts in arm A and 12/31 in B<br />

achieved stable disease (SD) or PR. PR or SD for �6 Cy was observed in 6/30 pts<br />

and 5/31 pts in arms A and B, respectively. Conclusions: In this phase I study, V<br />

in combination with Cis or Ca at full single-agent doses was well tolerated.<br />

Furthermore, several objective responses and cases <strong>of</strong> sustained SD were<br />

observed in heavily pretreated pts with advanced solid tumors.<br />

Dose levels N <strong>of</strong> pts DLTs in Cy 1 (n <strong>of</strong> pts)<br />

Arm A:<br />

V (mg)/Cis<br />

(mg/m 2 )<br />

Arm B:<br />

V (mg)/Ca<br />

(AUC)<br />

*MTD<br />

Developmental Therapeutics—Experimental Therapeutics<br />

A1: 100/60 3 -<br />

A2: 100/75 3 -<br />

A3: 200/75 3 -<br />

A4: 300/75 3 -<br />

A5: 300/100* 6�6 Increased serum creatinine (1) �<br />

fatigue (1), neutropenia (1)<br />

A6: 350/75 6 Increased alanine transaminase (1),<br />

neutropenia (1)<br />

B1: 100/4 3 -<br />

B2: 100/5 3 -<br />

B3: 200/5 3 -<br />

B4: 300/5 6 Thrombocytopenia with neutropenia (1)<br />

B5: 300/6* 6�7 Thrombocytopenia (1) � (1)<br />

B6: 350/5 3 Thrombocytopenia (1),<br />

neutropenia with thrombocytopenia,<br />

febrile neutropenia, fatigue,<br />

nausea and anorexia (1)<br />

177s<br />

3017 Poster Discussion Session (Board #9), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Phase I study <strong>of</strong> oral rigosertib in patients with advanced solid tumors.<br />

Presenting Author: Daniel W. Bowles, Division <strong>of</strong> Medical Oncology,<br />

University <strong>of</strong> Colorado Denver, Aurora, CO<br />

Background: Rigosertib (ON 01910.Na) is a new multi-targeted inhibitor <strong>of</strong><br />

several kinases, including polo-like kinase 1 and PI-3 kinase, and is in<br />

advanced trials for myelodysplastic syndrome and pancreatic cancer as an<br />

IV agent. An oral formulation with good oral bioavailability has entered<br />

phase 1 in patients (pts) with advanced solid tumors to determine the dose<br />

limiting toxicities (DLT), recommended phase II dose, pharmacokinetic<br />

(PK) pr<strong>of</strong>iles, and to document any antitumor activity <strong>of</strong> this compound.<br />

Methods: Pts with histologically confirmed solid tumors refractory to<br />

standard therapy were given escalating doses <strong>of</strong> rigosertib (70, 140, 280,<br />

560, 700 mg) twice daily continuously. Doses were escalated until<br />

intolerable grade 2 or grade 3/4 toxicities, at which point the previous dose<br />

level was expanded to define the MTD. All pts were assessed for safety, PK,<br />

PD and response. Urinary PK assessments were also performed at the MTD.<br />

Results: 25 pts (median age� 59; median ECOG PS� 1) received a median<br />

<strong>of</strong> 6 weeks <strong>of</strong> therapy at 5 dose levels: (70 mg n�3; 140 mg n�2 ; 280 mg<br />

n�3; 560 mg n�10; 700 mg n�7). The DLT was dysuria at 700 mg and<br />

led to expansion at 560 mg bid. There were no DLTs in the expansion<br />

cohort. Grade 2/3 toxicities related to rigosertib included dysuria (5/1),<br />

cystitis (4/0), urinary frequency (3/0), hematuria (2/1), abdominal pain<br />

(2/0), pelvic pain (1/1), nausea (1/0), distention/bloating (1/0) and<br />

hyponatremia (0/1). Improvements in dysuria were seen with oral hydration<br />

and sodium bicarbonate. A confirmed PR occurred in 1 pt (HN squamous<br />

cell ca, 42� weeks), and SD was observed in 2 pts with ovarian cancer (56<br />

weeks, 28 weeks), and 1 pt each with pancreatic neuroendocrine (40<br />

weeks), carcinoid (32 weeks), nasopharyngeal (24 weeks) and renal cell<br />

(32� weeks) tumors. Preliminary PK data reveal plasma rigosertib levels<br />

above the predicted pharmacodynamically active levels. Conclusions: The<br />

MTD <strong>of</strong> oral rigosertib administered twice daily continuously is 560mg.<br />

Dysuria is the dose limiting and most common toxicity and can be managed<br />

with oral hydration and sodium bicarbonate. Antitumor activity has been<br />

observed. Final safety and efficacy results, plasma and urinary PK relationships,<br />

and mutational analyses from archival tissue will be presented.<br />

3019 Poster Discussion Session (Board #11), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Phase I study <strong>of</strong> intravenous PI3K inhibitor BAY 80-6946: Activity in<br />

patients (pts) with advanced solid tumors and non-Hodgkin lymphoma<br />

treated in MTD expansion cohorts. Presenting Author: Michael T. Lotze,<br />

Hillman Cancer Center G27A, Pittsburgh , PA<br />

Background: BAY 80-6946 (BAY) is a potent and highly selective reversible<br />

pan-Class I PI3K inhibitor, previously reported to be tolerated as a 1-hr<br />

infusion at a dose <strong>of</strong> 0.8 mg/kg on days 1, 8 and 15 every 28 days (MTD).<br />

Additional pts were treated in MTD expansion cohorts to assess safety, PK,<br />

biomarkers and clinical benefit in selected tumor types, as well as safety in<br />

Type 2 diabetics. Methods: To date, 23 nondiabetic pts with solid tumors<br />

and 5 with follicular lymphoma (FL) received BAY at the MTD, until disease<br />

progression or unacceptable toxicity. Tumor types were selected for high<br />

frequency <strong>of</strong> PIK3CA mutation, including breast cancer (BC; 16), endometrial<br />

(3), gastric (2), GU transitional cell (1) and ovarian clear cell (1).<br />

<strong>Part</strong>ial enrichment for PIK3CA mutation was achieved by analysis <strong>of</strong><br />

plasma DNA. 3 diabetic pts have been enrolled, at starting dose <strong>of</strong> 0.4<br />

mg/kg. PK was done after the 1st and 3rd doses. FDG-PET/CT scans were<br />

done at baseline and 48 hrs after the 1st dose for pharmacodynamic<br />

assessment. Results: Safety and tolerability assessments confirmed MTD.<br />

There were no 1st cycle DLTs. Almost all nondiabetic pts had acute Grade<br />

2/3 hyperglycemia (HG) following each dose; at least 10 <strong>of</strong> them received<br />

insulin for 1-3 days post dose. Hypertension (HTN) lasting � 24 hrs was<br />

common in pts with preexisting HTN, and manageable. 2 FL pts developed<br />

interstitial pneumonitis (IP) after cycles 2 and 3, both responsive to<br />

steroids. Diabetic pts tolerated 0.4 mg/kg. Tumor SUVmax consistently fell<br />

at 48 hrs. 3 <strong>of</strong> 4 FL pts had partial response (PR) after 2 cycles, with 2<br />

confirmed PR pts on BAY for 10� and 8� mos. 2 BC pts showed PR , 1<br />

confirmed. PIK3CA mutation (n�7) does not appear to correlate with<br />

response. Average T1/2 was 36 hrs. Observation <strong>of</strong> high Cmax in very obese<br />

pts led to recommended maximum dose <strong>of</strong> 65 mg. Conclusions: BAY<br />

induced PRs in pts with BC and FL. The acute toxicities <strong>of</strong> HG in most pts<br />

and HTN in some are manageable, and IP has been limited to 2 lymphoma<br />

pts and is responsive to steroids. The observed clinical activity <strong>of</strong> BAY,<br />

along with its acceptable safety pr<strong>of</strong>ile, provide a rationale for the ongoing<br />

development <strong>of</strong> BAY in combination with cytotoxic and targeted agents.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


178s Developmental Therapeutics—Experimental Therapeutics<br />

3020 Poster Discussion Session (Board #12), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

A phase I, single-institution, open-label, dose escalation trial with an<br />

expansion cohort evaluating the safety and tolerability <strong>of</strong> AZD6244 and<br />

IMC-A12 in subjects with advanced solid malignancies. Presenting Author:<br />

Shabina Roohi Ahmed, The Johns Hopkins University School <strong>of</strong> Medicine,<br />

Baltimore, MD<br />

Background: Significant crosstalk exists between the PI3K and Raf/MEK/<br />

ERK pathways and treatment <strong>of</strong> select cell lines with a MEK inhibitor and<br />

an IGF1-R inhibitor has been shown to cause greater inhibition <strong>of</strong> growth<br />

than either agent alone. IMC-A12 (I) is a recombinant human monoclonal<br />

antibody directed IGFI-R; it blocks interaction between IGF-1R and its<br />

ligands, IGF-I and -II. AZD6244 (A) is a highly selective, non-competitive<br />

MEK ½ inhibitor. Methods: The study is a phase I, dose-escalation with a<br />

standard 3�3 design. Eligible patients had advanced solid tumor, good<br />

end organ function and performance status with exclusion for poorly<br />

controlled diabetes or growth hormone abnormalities. <strong>Part</strong> 1: open label,<br />

dose escalation. Pts were treated with A at 50 mg BID, then 75 mg BID; I at<br />

12mg/kg q2wk held constant. DLTs were defined as grade (G) 3 or 4<br />

toxicities during cycle 1. <strong>Part</strong> 2: expansion cohort <strong>of</strong> 15 pts for correlative<br />

endpoints and PK studies; serial tumor biopsies will be evaluated for<br />

changes in signaling in the IGF pathway, including p-ERK, p-Akt and RAS.<br />

Mutational analysis <strong>of</strong> RAS and RAF will be done to correlate with PD<br />

endpoints and disease response. Results: <strong>Part</strong> 1: 16 patients accrued with<br />

6 treated at DL1 and 10 treated at DL2. Multiple tumor types were<br />

represented, including 4 colon, 2 thyroid, 2 pancreatic, and 1 breast. At 75<br />

mg <strong>of</strong> A, there was 1 DLT <strong>of</strong> visual changes. G3 AEs included nausea/<br />

vomiting (n�2, 12%), visual changes/retinopathy (n�2, 12%), MRSA skin<br />

infection (n�1, 6%), (n�1, 6%), hyperglycemia (n�1, 6%), and stroke<br />

(n�1, 6%). Other common AEs included acneform rash (n�10, 63%),<br />

nausea/vomiting (n�6, 37%), anorexia (n�4, 25%), and diarrhea (n�3,<br />

19%). Of 10 evaluable patients in <strong>Part</strong> 1, 1 had a partial response and 4<br />

patients had stable disease (40%). Sustained responses <strong>of</strong> 10 and 14<br />

months were seen in the DTC patients. 3 patients are currently enrolled in<br />

the expansion cohort. The RP2D is 50 mg <strong>of</strong> A, due to the visual changes<br />

seen with 75 mg, and 12mg/kg <strong>of</strong> I. Conclusions: A at 50 mg QD combines<br />

safely with I 12mg/kg. An expansion cohort is underway to determine if this<br />

combination warrants further investigation.<br />

3022 Poster Discussion Session (Board #14), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

A pharmacokinetic (PK) pharmacodynamic (PD) driven first-in-human<br />

study <strong>of</strong> the oral class I PI3K inhibitor CH5132799, in patients with<br />

advanced solid tumors. Presenting Author: Aurelius Gabriel Omlin, Section<br />

<strong>of</strong> Medicine, The Institute <strong>of</strong> Cancer Research, Sutton, United Kingdom,<br />

and Drug Development Unit, The Royal Marsden NHS Foundation Trust,<br />

Sutton, United Kingdom<br />

Background: The phosphatidylinositol 3-kinase (PI3K) pathway is a promising<br />

target in cancer. CH5132799 is a novel PI3K inhibitor, selectively<br />

inhibiting class I PI3Ks (�, �, � and �) with no inhibition <strong>of</strong> class II and III<br />

or mTOR, and with potent antitumor activity in preclinical studies (Tanaka<br />

et al, Clin Cancer Res; 17; 3272-81, 2011). First-in-human study<br />

objectives were determination <strong>of</strong> maximum tolerated dose (MTD), safety/<br />

tolerability, PK/PD and clinical activity (RECIST). Methods: A3�3 dose<br />

escalation design was used. The initial dosing schedule <strong>of</strong><br />

CH5132799(schedule A) was once a day (QD). Due to a relatively short<br />

half-life, a twice a day (BID) schedule (schedule B) was introduced. PK<br />

pr<strong>of</strong>iles were studied over 72 hours. PD analyses included quantification <strong>of</strong><br />

various phosphoproteins in platelet rich plasma (PRP). Tumor assessments<br />

were performed at baseline and cycle 3 day 1 (C3D1) and FDG-PET scans<br />

at baseline, C1D8 and C3D1. Results: 29 patients (pts) with a variety <strong>of</strong><br />

solid tumors have been treated (A 23 pts, B 6 pts, the most common tumors<br />

were breast, oesophageal, colorectal and ovarian). The starting doses were<br />

2 mg (A) and 48 mg (B). The current doses being explored are 96 mg (A)<br />

and 72 mg (B). The most frequently reported drug-related AEs were Grade<br />

1/2 diarrhea, nausea, fatigue, anorexia and anemia. 1 DLT (Grade 3<br />

elevated LFT) was observed in a hepatocellular carcinoma pt at 48 mg BID.<br />

MTD has not yet been determined. The preliminary mean �SD Cmax and<br />

AUC at 96 mg QD were 202�129 ng/ml and 1407�935 ng·hr/ml<br />

respectively, which is consistent with an efficacious exposure based on<br />

preclinical models. Some patients achieved the expected exposure at over<br />

32 mg. From single dose <strong>of</strong> 48mg there was a reduction <strong>of</strong> phosphorylation<br />

<strong>of</strong> AKT in PRP after treatment, consistent with pathway modulation. A<br />

patient with clear cell ovarian cancer and a PIK3CA mutation treated at 48<br />

mg BID showed �50% decrease in SUV on a PET scan at C1D8 and a 75%<br />

decrease in CA-125 at C2D1. 5 pts exhibited SD (�8 weeks). Conclusions:<br />

CH5132799 is well tolerated either QD �96 mg or BID �48 mg.<br />

Dose-escalation continues and updated safety/efficacy/PK/PD data will be<br />

presented.<br />

3021 Poster Discussion Session (Board #13), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

A phase Ib study <strong>of</strong> the Akt inhibitor GDC-0068 with docetaxel (D) or<br />

mFOLFOX-6 (F) in patients (pts) with advanced solid tumors. Presenting<br />

Author: Cristina Saura, Medical Oncology Department, Vall d’Hebron<br />

University Hospital, Barcelona, Spain<br />

Background: Activation <strong>of</strong> the Akt pathway is observed in multiple tumors<br />

and may contribute to chemoresistance. GDC-0068 is an ATP-competitive<br />

small molecule inhibitor <strong>of</strong> all three is<strong>of</strong>orms <strong>of</strong> Akt; in a phase Ia study, it<br />

was well tolerated with maximum tolerated dose (MTD) <strong>of</strong> 600 mg daily (21<br />

days on/7days <strong>of</strong>f) and pharmacodynamic down-regulation <strong>of</strong> Akt signaling<br />

in tumors at doses �100 mg. In vitro, GDC-0068 shows synergism with<br />

cytotoxic agents. This phase Ib study defines the dose limiting toxicities<br />

(DLT), MTD, safety and pharmacokinetics (PK) <strong>of</strong> GDC0068 in combination<br />

with D and F. Methods: Using a 3�3 designeligible patients (pt) with<br />

advanced/metastatic solid tumors were treated with either D, 75 mg/m2 day<br />

1 and GDC-0068 daily on days 2-15 <strong>of</strong> a 21 day cycle (Arm A); or F, bolus<br />

5FU 400mg/m2 , leucovorin 400 mg/m2 , oxaliplatin 85 mg/m2 all day 1,<br />

and infusional 5FU 2400mg/m2 for 46 hours and GDC-0068 daily on days<br />

1-7 <strong>of</strong> a 14 day cycle (Arm B). PK sampling was performed in Cycles 1 and<br />

2. Results: 23 pts have enrolled; Arm A (GDC-0068, mg): 100 (n�3), 200<br />

(n�4), and 400 (n�5); Arm B:100 (n�6) and 200 (n�5). Median prior<br />

therapies � 3. GDC-0068-related adverse events in � 10% <strong>of</strong> pts were<br />

diarrhea, nausea, vomiting, fatigue, and decreased appetite. All GDC-0068related<br />

AEs were grade 1 or 2, except one grade 4 neutropenia in Arm A. No<br />

DLTs have been reported to date. Preliminary data show no alteration in the<br />

PK <strong>of</strong> GDC-0068, D or F compared to phase Ia or historical data. Two<br />

heavily pretreated pts with cervical and PTEN-loss colon cancers treated in<br />

Arm B demonstrated both RECIST partial response and tumor marker<br />

decrease by first CT evaluation. Conclusions: The combination <strong>of</strong> GDC-<br />

0068 with D or F is well-tolerated and shows early signs <strong>of</strong> anti-tumor<br />

activity. Dose-escalations continue.<br />

3023 Poster Discussion Session (Board #15), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

A five-arm, open-label, phase I/lb study to assess safety and tolerability <strong>of</strong><br />

the oral MEK1/MEK2 inhibitor trametinib (GSK1120212) in combination<br />

with chemotherapy or erlotinib in patients with advanced solid tumors.<br />

Presenting Author: Carlos Becerra, Internal Medicine, Sammons Cancer<br />

Center, Dallas, TX<br />

Background: Trametinib, an oral MEK1/MEK2 inhibitor, has demonstrated<br />

single-agent clinical activity. In vitro studies <strong>of</strong> trametinib plus docetaxel<br />

(doc), pemetrexed (pem) and erlotinib (erl) showed enhanced growth<br />

inhibition <strong>of</strong> lung cancer cell lines with and without RAS/RAF mutations.<br />

Trametinib�doc significantly increased apoptosis compared with either<br />

agent alone. Methods: This is a two-part, five-arm, phase I/Ib, open-label<br />

study to evaluate the safety and tolerability <strong>of</strong> trametinib plus doc, erl, pem,<br />

pem�carboplatin (pem�carbo), or nab-paclitaxel (nab-pac)<br />

(NCT01192165). <strong>Part</strong> I is dose escalation in patients (pts) with advanced<br />

solid tumors; part II is dose expansion in pts with lung and pancreatic<br />

cancers. A 3�3 dose-escalation design was used to determine the<br />

maximum tolerated dose (MTD) and the recommended phase II regimen<br />

(RP2R) for each combination. Dose-limiting toxicities (DLTs) were determined<br />

during the first treatment cycle (21 days). Trametinib was started at<br />

0.5 mg/day; chemotherapy was given at full recommended doses. Erl was<br />

escalated from 50 mg/day. Pharmacokinetic (PK) samples were collected<br />

pre-, and 1, 2, 3 and 6 hours post-dose. Results: As <strong>of</strong> January 2012, 80 pts<br />

have been enrolled across all arms except trametinib�nab-pac. Preliminary<br />

exposure results <strong>of</strong> trametinib�doc, erl, or pem suggest no PK<br />

drug-drug interaction. The predominant DLT for trametinib�doc without<br />

growth factors (MTD � 0.5 mg�60 mg/m2 ) was neutropenia. When<br />

administered with growth factors, trametinib�doc has been given up to 1.5<br />

mg�75 mg/m2 with no DLTs. The predominant DLTs for trametinib�erl<br />

(MTD � 1 mg�100 mg) were diarrhea and mucositis and for<br />

trametinib�pem (MTD � 1.5 mg�500 mg/m2 ) were mucositis and febrile<br />

neutropenia. The MTD for trametinib�pem�carbo has not yet been<br />

determined. To date there are 5 PRs in the trametinib�doc group and 2<br />

PRs in the trametinib�pem group. An NSCLC expansion cohort for<br />

trametinib�pem is enrolling. Conclusions: Trametinib�doc and<br />

trametinib�pem have shown acceptable tolerability and initial evidence <strong>of</strong><br />

clinical activity.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


3024 Poster Discussion Session (Board #16), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

PX-866 and docetaxel in patients with advanced solid tumors. Presenting<br />

Author: Antonio Jimeno, Division <strong>of</strong> Medical Oncology, University <strong>of</strong><br />

Colorado Denver, Aurora, CO<br />

Background: PX-866, an irreversible pan-is<strong>of</strong>orm inhibitor <strong>of</strong> Class 1 PI-3K<br />

has additive to synergistic effects when combined with docetaxel in<br />

xenograft models <strong>of</strong> NSCLC and SCCHN. A phase I/II study <strong>of</strong> PX-866 and<br />

docetaxel was initiated to further evaluate this combination. Enrollment in<br />

phase I is complete, and the randomized, controlled phase II portion is now<br />

enrolling patients with either NSCLC or SCCHN. Phase I safety and<br />

pharmacokinetics were previously described; the recommended phase II<br />

dose <strong>of</strong> PX-866 was 8 mg daily, the same as the single agent MTD (Jimeno<br />

A, et al. AACR-NCI-EORTC, 2011). Updated phase I antitumor and<br />

biomarker results are presented here. Methods: Phase 1 consisted <strong>of</strong> dose<br />

escalation <strong>of</strong> PX-866 at 4, 6, or 8 mg po qd in combination with docetaxel<br />

75 mg/m2 IV once every 21 days (d). Patients had advanced solid tumors<br />

for which docetaxel was compendia listed. Tumor restaging was performed<br />

every 2 cycles. Archived tumor biopsies were collected for assessment <strong>of</strong><br />

potential biomarkers <strong>of</strong> response, including PIK3CA and KRAS mutations<br />

and PTEN expression. Results: 43 pts were enrolled: NSCLC (n�6),<br />

prostate (n�5), ovarian (n�5), SCCHN (n�3), and pancreatic (n�3) were<br />

the most common tumor types. Median time on study (TOS) was 81 d<br />

(5-361), with 9 pts still on study. 16 pts received � 6 cycles (6-17),<br />

including 3 pts with NSCLC, and 4 pts with ovarian cancer. Biomarker data<br />

are available for 20 evaluable pts. Median days on study by mutational<br />

status was: PIK3CA/KRAS WT (n�13): 91 d (28-286); PIK3CA-MUT<br />

(n�5): 183 d (64-342); KRAS-MUT (n�3): 141 d (125-361); and<br />

PIK3CA/KRAS-MUT (n�2): 96 d (86-105). A trend toward longer TOS was<br />

observed in pts with PIK3CA-MUT vs PIK3CA/KRAS-WT (p�0.14). Assessment<br />

<strong>of</strong> PTEN is ongoing. Best response in 32 evaluable pts was 2 PR<br />

(6%), 22 SD (69%), and 8 PD (25%). The PRs were in NSCLC and ovarian<br />

cancer (both PIK3CA/KRAS WT). 8 other pts had �15% tumor shrinkage,<br />

including NSCLC (n�2). Conclusions: PX-866 with docetaxel was associated<br />

with a disease control rate <strong>of</strong> 75%, with 50% <strong>of</strong> evaluable pts<br />

demonstrating SD or better for � 6 cycles. Based on available data, a trend<br />

for a longer TOS was seen with PIK3CA-MUT pts. This relationship will be<br />

further evaluated in phase II.<br />

3026 Poster Discussion Session (Board #18), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

A phase I study <strong>of</strong> Debio 0932, an oral HSP90 inhibitor, in patients with solid<br />

tumors. Presenting Author: Nicolas Isambert, Centre Georges François Leclerc,<br />

Dijon, France<br />

Background: Debio 0932 is an oral second-generation heat shock protein 90<br />

(HSP90) inhibitor that has shown extended tumor retention, blood-brain-barrier<br />

penetration, and promising anti-tumor activity both as monotherapy and combination<br />

against a broad range <strong>of</strong> tumors in pre-clinical models. Here we report the<br />

results <strong>of</strong> the dose escalation part <strong>of</strong> a phase I study in patients with advanced<br />

solid tumors or lymphoma (NCT01168752). Methods: This was an open-label,<br />

non-randomized, 3 � 3 dose-escalation study to determine the maximum<br />

tolerated dose (MTD) <strong>of</strong> Debio 0932 when given QD or Q2D during the first 30<br />

days <strong>of</strong> treatment in patients with advanced solid tumours or lymphoma resistant<br />

to standard therapy. The starting dose in both treatment groups was 50mg.<br />

Doses were increased according to an algorithm based on observed toxicity and<br />

dose limiting toxicities (DLT). Tumor assessments were performed every 8<br />

weeks. Results: Patient characteristics and results are summarized below. DLTs<br />

occurred at 1600mg in both dose groups. Adverse events (AE) were mostly<br />

CTCAE grade 1 or 2, with no apparent dose relationship. No ocular or cardiac<br />

toxicity was observed. The main reason for treatment withdrawal was progressive<br />

disease. Investigator-reported cases <strong>of</strong> SD and PR were observed. Conclusions:<br />

Debio 0932 mono-therapy was generally well tolerated and showed promising<br />

signs <strong>of</strong> efficacy in patients with advanced solid tumors. The recommended<br />

phase II dose, established at 1000mg QD, will be tested in an additional 30<br />

patients in an ongoing expansion study. A phase I-II study <strong>of</strong> Debio 0932 in<br />

combination with standard <strong>of</strong> care in the first- and second-line treatment <strong>of</strong><br />

NSCLC is planned.<br />

Q2D dosing<br />

N � 23<br />

QD dosing<br />

N � 28<br />

Mean age (range) (yrs) 58 (39-72) 55 (27-74)<br />

Gender M / F % 48/52 68/32<br />

Predominant cancer types % Colorectal 26<br />

Colorectal 29<br />

Lung 17<br />

Pancreas 13<br />

Lung 18<br />

>3 previous treatment regimens % 83 86<br />

Days on treatment (range) 69 (1 – 223) 66 (6 – 252)<br />

DLTs / dose Febrile neutropnia / 1600mg Diarrhea / 1600mg<br />

Asthenia / 1600mg<br />

Most frequent AEs %<br />

.<br />

.<br />

Constipation<br />

44<br />

36<br />

Diarrhea<br />

26<br />

61<br />

Nausea<br />

39<br />

46<br />

Vomiting<br />

39<br />

43<br />

Asthenia<br />

61<br />

71<br />

Decreased appetite<br />

35<br />

50<br />

Best overall tumor response<br />

.<br />

.<br />

PR %<br />

4<br />

0<br />

SD %<br />

17<br />

29<br />

PD %<br />

57<br />

61<br />

Not evaluable %<br />

13<br />

3<br />

Not assessed %<br />

9<br />

7<br />

Developmental Therapeutics—Experimental Therapeutics<br />

179s<br />

3025 Poster Discussion Session (Board #17), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

A first-in-human phase I study to evaluate the fully human monoclonal<br />

antibody OMP-59R5 (anti-Notch2/3) administered intravenously to patients<br />

with advanced solid tumors. Presenting Author: Anthony W. Tolcher,<br />

South Texas Accelerated Research Therapeutics, LLC, San Antonio, TX<br />

Background: The Notch pathway plays a central role in embryonic development,<br />

the regulation <strong>of</strong> stem and progenitor cells, and is implicated<br />

centrally in many human cancers. OMP-59R5 is a fully human IgG2<br />

originally identified by binding to Notch2. It inhibits the signaling <strong>of</strong> both<br />

Notch2 and Notch3 receptors. Mouse xenograft studies using minimallypassaged,<br />

patient-derived xenografts show that OMP-59R5 impedes tumor<br />

growth and eliminates cancer stem cells (CSCs) in many tumor types.<br />

OMP-59R5 modulates the expression <strong>of</strong> stromal genes and genes associated<br />

with the function <strong>of</strong> tumor vascular pericytes. As such, OMP-59R5 is a<br />

novel anti-cancer agent that inhibits tumor growth through direct actions on<br />

tumor cells, including CSCs, and effects the stroma and vasculature.<br />

Methods: A phase I dose escalation study (3�3 design) was initiated in<br />

solid tumor patients. OMP-59R5 was administered to study safety, pharmacokinetics<br />

(PK), pharmacodynamics (PD), preliminary efficacy, and to<br />

determine the maximum tolerated dose (MTD). Results: Twenty-four patients<br />

have been enrolled in 5 dose-escalation cohorts at doses <strong>of</strong> 0.5, 1,<br />

2.5, and 5mg/kg administered weekly (QW) and 5mg/kg administered every<br />

other week (QOW). The most frequently reported drug-related adverse<br />

events were: mild to moderate diarrhea, fatigue, nausea, vomiting, decreased<br />

appetite, and constipation. Diarrhea was dose related and occurred<br />

at doses �2.5mg/kg weekly and appears less pronounced with every other<br />

week dosing. Two dose-limiting toxicities (grade 3 diarrhea and grade 3<br />

hypokalemia) occurred at 5mg/kg QW and 2.5mg/kg was established as an<br />

MTD for QW dosing. A QOW dosing schedule is currently under investigation.<br />

The PK <strong>of</strong> OMP-59R5 was characterized by fast and dose-dependent<br />

clearance. Two patients (Kaposi’s sarcoma at 5mg/kg and adenoid cystic<br />

carcinoma at 2.5mg/kg) had prolonged stable disease for �110 days. PD<br />

analyses for Notch pathway modulation are ongoing. Conclusions: OMP-<br />

59R5 is generally well tolerated. An MTD <strong>of</strong> 2.5mg/kg QW has been<br />

established and a QOW schedule is currently under study. Updated results<br />

will be presented.<br />

3027 Poster Discussion Session (Board #19), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Rational molecularly targeted combinations: A parallel-arm phase I trial <strong>of</strong><br />

the humanized anti-IGF-1R antibody dalotuzumab (D) in combination with<br />

the allosteric AKT inhibitor MK-2206 or the gamma secretase inhibitor<br />

MK-0752, in patients with advanced solid tumors. Presenting Author:<br />

Irene Brana, Princess Margaret Hospital, Toronto, ON, Canada<br />

Background: Two rational strategies to interrogate the IGF-1R pathway were<br />

investigated in this parallel-arm phase I trial: 1) vertical inhibition with D<br />

and MK-2206 to potentiate PI3K pathway targeting, 2) horizontal crosstalk<br />

inhibition with D and MK-0752 to exert effects against cellular proliferation,<br />

angiogenesis and stem cell propagation. Methods: Patients (pts) with<br />

advanced solid tumors and no prior exposure to agents <strong>of</strong> the same targets<br />

were eligible. Dose escalation was based on a modified toxicity probability<br />

interval method (Ji et al, 2010). Arm A is comprised <strong>of</strong> fixed D dose <strong>of</strong> 10<br />

mg/m2 IV weekly plus MK-2206 at escalating doses <strong>of</strong> 90-200 mg PO<br />

weekly. Arm B is comprised <strong>of</strong> fixed MK-0752 dose <strong>of</strong> 1800 mg PO weekly<br />

plus D at escalating doses <strong>of</strong> 7.5-10 mg/m2 IV weekly. Each cycle � 4<br />

weeks in both arms. Primary objectives were to determine DLT and RP2D <strong>of</strong><br />

these combinations. Results: 30 patients were enrolled in the dose<br />

escalation phase (18 in Arm A and 12 in Arm B) with: median age � 56<br />

(range 36-74); M:F � 13:17; ECOG 0:1 � 12:18; primary diagnosis �<br />

colorectal (CRC) (9), NSCLC (3), others (17); prior metastatic regimens �<br />

3 (range 1-4). In Arm A, 1/6 DLT was observed with MK-2206 at 135 mg<br />

(G3 serum sickness-like reaction); 2/3 DLTs with MK-2206 at 200 mg (G4<br />

leukopenia/neutropenia; G3 rash). No other G4 or worse adverse events<br />

(AEs) were observed. RP2D <strong>of</strong> Arm A � D 10mg/m2 IV weekly and<br />

MK-2206 150 mg PO weekly. Other common AEs in Arm A included:<br />

fatigue, rash and nausea. In Arm B, 2/6 DLTs were observed with D at 10<br />

mg/m2 (G3 rash; G3 nausea/vomiting). RP2D <strong>of</strong> Arm B � D 10mg/m2 IV<br />

weekly and MK-0752 at 1800 mg PO weekly based on Ji et al. Other<br />

common AEs in Arm B included: anorexia, nausea and fatigue. No PR was<br />

observed and SD � 4 months occurred in 4 pts (all in Arm A). PK analyses<br />

are ongoing. Conclusions: Targeted combinations <strong>of</strong> D with MK-2206 or<br />

with MK-0752 were both tolerable. Expansion cohorts based on biomarker<br />

selection are underway, specifically ovarian cancer with high IGF1 expression/low<br />

RAS score for Arm A; and KRAS wild-type CRC with high IGF1/low<br />

IGF2 expression for Arm B.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


180s Developmental Therapeutics—Experimental Therapeutics<br />

3028 Poster Discussion Session (Board #20), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

First-in-human phase I study: Results <strong>of</strong> a second-generation nonansamycin<br />

heat shock protein 90 (HSP90) inhibitor AT13387 in refractory<br />

solid tumors. Presenting Author: Daruka Mahadevan, Arizona Cancer<br />

Center, Tucson, AZ<br />

Background: AT13387 is a second-generation potent, novel non-ansamycin<br />

HSP90 inhibitor (IC50 0.71 nM). AT13387 has good tissue distribution,<br />

long tumor t1/2 resulting in extended knockdown <strong>of</strong> client proteins in cells<br />

and animal models <strong>of</strong> gastric, prostate and melanoma. Methods: AT13387<br />

was administered as a 1-hr IV infusion twice weekly on Days 1, 4, 8, 11, 15<br />

and 18 (<strong>Part</strong> A) and then subsequently, weekly on Days 1, 8 and 15 (<strong>Part</strong> B)<br />

<strong>of</strong> a 28-day cycle in a standard 3�3 dose-escalation design. The primary<br />

endpoint was to determine the MTD; secondary endpoints included PK, PD,<br />

safety and tolerability. Results: As <strong>of</strong> 1 Dec 2011, 53 patients received<br />

1–12 cycles <strong>of</strong> AT13387 (median 2). In <strong>Part</strong> A, AT13387 was evaluated at<br />

5 doses (10, 20, 40, 80, 120 mg/m2 ). In <strong>Part</strong> B, 4 additional doses (150,<br />

220, 260 and 310 mg/m2 ) were explored. Common treatment related<br />

toxicities (� 10%) included transient and reversible GI disturbance<br />

(vomiting, nausea, dry mouth, diarrhea, abdominal pain), fatigue, local<br />

infusion site irritation and systemic infusion-related symptoms including<br />

chills, rash, itch, cardiovascular (tachycardia, bradycardia, hypertension,<br />

hypotension), and visual changes (diplopia, transient flashes, delayed<br />

light/dark accommodation, blurred vision). Severity <strong>of</strong> infusion-related<br />

symptoms, GI toxicities, and fatigue at 310 mg/m2 prevented further dose<br />

escalation. A dose <strong>of</strong> 260 mg/m2 has been identified as the once weekly<br />

MTD and the study is currently accruing at this dose. Biological evidence <strong>of</strong><br />

HSP90 inhibition, demonstrated by an increase in HSP70 in PBMCs, was<br />

detected at all doses and exhibited evidence <strong>of</strong> dose dependence. PK<br />

demonstrated dose proportionality, without significant accumulation. One<br />

durable RECIST PR (8 months) in an imatinib relapsed metastatic GIST<br />

patient with c-kit mutations in exons 11 and 17 was observed. Three SD �<br />

6 months (follicular cell thyroid carcinoma, metastatic uveal melanoma,<br />

GIST) were also observed. Conclusions: The MTD <strong>of</strong> AT13387, administered<br />

as a once weekly IV, is 260 mg/m2 . All drug-related toxicities were<br />

generally reversible. Single-agent activity was observed. Dose expansion at<br />

MTD and phase II studies are ongoing.<br />

3030 Poster Discussion Session (Board #22), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Phase I dose-finding study <strong>of</strong> golvatinib (E7050), a c-Met and Eph receptor<br />

targeted multi-kinase inhibitor, administered orally QD to patients with<br />

advanced solid tumors. Presenting Author: Gennaro Daniele, Royal Marsden<br />

Hospital and Institute <strong>of</strong> Cancer Research, Sutton, United Kingdom<br />

Background: Golvatinib is a highly potent, small molecule ATP-competitive<br />

inhibitor <strong>of</strong> the c-Met receptor tyrosine kinase and multiple members <strong>of</strong> the<br />

Eph receptor family as well as c-Kit and Ron, based on isolated kinase<br />

assays. Golvatinib showed preclinical evidence <strong>of</strong> anti-tumor activity. This<br />

phase 1 study was performed to determine the MTD, safety, PK, PD and<br />

preliminary activity <strong>of</strong> golvatinib. Methods: Patients (pts) with advanced<br />

solid tumors, ECOG PS 0-1, � 18 years (yrs) and adequate organ function<br />

were eligible. Golvatinib was administered orally, once daily (QD), continuously.<br />

Blood samples for PK and PD analysis were collected at multiple<br />

time-points. Mandatory tumor biopsies for PD analysis were taken pre and<br />

post Cycle 1 in an expanded MTD cohort. Results: 34 pts (M/F: 21/13;<br />

median age 63.5yrs [range 32-78]) were treated at 6 dose levels: 100,<br />

200, 270, 360, 450 and 400 mg. Tumor types were colorectal (n�15),<br />

lung (n�4), renal (n�4), esophageal (n�2), melanoma (n�2) and others<br />

(n�7). Three DLTs were observed: Gr3 GGT and alkaline phosphatase<br />

(n�1; 200mg) and repeated Gr2 (n�1) and Gr3 (n�1) fatigue, both at<br />

450mg. The MTD was determined to be 400 mg QD. Frequently occurring<br />

adverse events ([AEs]; all grades) were fatigue: 68% (Gr3: 15%), diarrhea:<br />

65%, nausea: 62%, vomiting: 53%, decreased appetite: 47% (Gr3: 9%),<br />

ALT increase: 38% and AST increase: 23%. No Gr4 AEs were observed.<br />

Best response was stable disease in 6 pts lasting �84 days. PK showed<br />

high variability and plasma concentration increased with dose. The Cmax was reached within a median time <strong>of</strong> 4 hours. Plasma PD analysis showed<br />

an increase in soluble c-Met and Ang 2 levels after golvatinib treatment.<br />

Tumor PD analysis in 5 pts at 400 mg demonstrated a baseline elevated<br />

MET gene copy number, with c-Met overexpression and post treatment<br />

decline in phospho(p)-c-Met expression in 1 pt; post-treatment decline in<br />

p-c-Met in a 2nd pt, and post-treatment decline in p-ERK in a 3rd pt.<br />

Conclusions: Golvatinib at an MTD <strong>of</strong> 400 mg QD has manageable toxicity.<br />

Preliminary PD analysis demonstrated evidence <strong>of</strong> c-Met target modulation.<br />

Further evaluation will continue in phase 2 combination studies.<br />

3029 Poster Discussion Session (Board #21), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

<strong>Clinical</strong> pharmacokinetics <strong>of</strong> the Smac-mimetic birinapant (TL32711) as a<br />

single agent and in combination with multiple chemotherapy regimens.<br />

Presenting Author: Gerald J. Fetterly, Roswell Park Cancer Institute,<br />

Buffalo, NY<br />

Background: Birinapant is a novel small molecule Smac-mimetic that<br />

targets members <strong>of</strong> the inhibitor <strong>of</strong> apoptosis proteins (cIAP1, cIAP2 and<br />

XIAP) involved in the blockade <strong>of</strong> apoptosis. A population PK model was<br />

developed to characterize the interpatient variability in birinapant PK and<br />

to evaluate the effect <strong>of</strong> multiple combination regimens on birinapant<br />

disposition and safety. Methods: Birinapant was administered alone or in<br />

combination to 114 patients (55M/59F; 89% Caucasian) with advanced<br />

malignancies. Birinapant was administrated by a 30 min IV infusion QW<br />

alone (30 pats), or approx. 30 min. after chemotherapy with irinotecan (19<br />

pats), docetaxel (20 pats), gemcitabine (17 pats), liposomal doxorubicin<br />

(13 pats), or paclitaxel/carboplatin (15 pats). Birinapant dose levels ranged<br />

from 0.18 to 35 mg/m2 . Population PK modeling was performed to<br />

investigate the effect <strong>of</strong> the following patient covariates: [BW (38.5-127.5<br />

kg), age (27.5-86.0 yrs), CrCL (36.4-219.2 ml/min), ALT (6-121 IU/L),<br />

and TBIL (0.1-1.7 mg/dL)]. Results: A 3-compartment PK model described<br />

the time course <strong>of</strong> birinapant disposition with predicted values for T1/2,<br />

CL, and Vd <strong>of</strong> 40 h, 21 L/h and 10.2 L, respectively. Birinapant displayed<br />

linear PK across the dose range with no significant accumulation in plasma<br />

following weekly dosing. Goodness <strong>of</strong> fit plots supported the model fit, with<br />

residual variability <strong>of</strong> 23%. The PK <strong>of</strong> birinapant remained unchanged<br />

when combined with irinotecan, docetaxel, gemcitabine and liposomal<br />

doxorubicin. Concomitant administration with paclitaxel/carboplatin resulted<br />

in a 2-fold increase in birinapant AUC possibly due to reduced<br />

OATP1B3 mediated tissue uptake. Conclusions: These data show that<br />

birinapant possesses an excellent PK pr<strong>of</strong>ile with dose proportional<br />

kinetics, a long terminal half-life for target coverage, low/moderate interpatient<br />

variability in CL and no significant accumulation following weekly<br />

dosing. Importantly, the PK <strong>of</strong> birinapant remained unchanged when<br />

combined with multiple chemotherapy regimens and the increased exposure<br />

with paclitaxel/carboplatin was not associated with any change in<br />

birinapant tolerability.<br />

3031 Poster Discussion Session (Board #23), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Pro<strong>of</strong> <strong>of</strong> mechanism (POM) in the first-in-human trial <strong>of</strong> two novel<br />

indenoisoquinoline, non-camptothecin topoisomerase I (TOP1) inhibitors.<br />

Presenting Author: James H. Doroshow, Division <strong>of</strong> Cancer Treatment and<br />

Diagnosis, National Cancer Institute, Bethesda, MD<br />

Background: Indenoisoquinolines LMP 400 and LMP 776 form stable<br />

DNA-top1 cleavage complexes, have a preference for unique DNA cleavage<br />

sites, and are not substrates for ABC transporters compared to camptothecins.<br />

We conducted a randomized phase I trial <strong>of</strong> LMP 400 and LMP 776<br />

in patients (pts) with refractory tumors to establish safety and MTD <strong>of</strong> LMP<br />

400 and LMP 776 administered IV daily x5d,q28d; determine PK;<br />

evaluate TOP1 and �H2AX levels in tumor biopsies (bx); and compare<br />

pharmacodynamic responses <strong>of</strong> LMP400 and LMP776. Methods: Pts had<br />

refractory malignancies; � 18 yrs old; KPS � 70%; adequate organ<br />

function. DLT: drug-related gr � 3 non-hem or gr 4 hem toxicities during<br />

C1. Pts assigned to receive either LMP 400 or LMP 776. Dose level (DL) in<br />

mg/m2 /day for LMP 776: 1, 2, 3, 4.5 mg; LMP 400: 2.5, 5, 10, 20, 40,<br />

80. Accelerated titration design with one pt per dose level (DL), 100% dose<br />

escalation until one DLT or 2 gr 2 toxicities, then 3�3 design. Tumor bx<br />

and circulating tumor cells (CTCs) were obtained at baseline and C1D3.<br />

Results: Twenty pts accrued to date [LMP 400 (11 pts); LMP 776 (9 pts)];<br />

M/F 7/13; median age 59 (range 32-71 yrs); diagnosis (# <strong>of</strong> pts): colorectal<br />

(10), vaginal adenocarcinoma (1), head and neck (2), bladder (1),<br />

melanoma (1), pancreas (1), thyroid (1), small cell lung (1), sarcoma (1),<br />

lymphoma (1). DLTs: LMP 400- gr 4 myelosuppression, gr 3 fatigue at DL<br />

6; DL 5 expanded to establish MTD; LMP 776 (DL2)- gr 4 hypercalcemia,<br />

no other DLTs, dose escalation continuing. One pt with irinotecanrefractory<br />

colon cancer had shrinkage <strong>of</strong> lung nodules post one cycle <strong>of</strong><br />

LMP 400. Relative to baseline, 5 <strong>of</strong> 6 tumor bx demonstrated 20-50%<br />

reduction in TOP1 levels (LMP 776); an increase in �H2AX foci in tumor<br />

was observed in 2/6 LMP 776 and 1/1 LMP 400 pts. An increase in �H2AX<br />

was also observed in hair bulbs (3/3 pts) and CTCs (9/20 pts). PK for LMP<br />

400 and LMP 776: mean T1/2 48 and 36 h; Cl 1.5 and 2.0 L/h/m2 ;Vc30<br />

and 31 L/m2 , respectively. Conclusions: POM, as assessed by reduction in<br />

TOP 1 levels and increase in �H2AX in tumor and CTCs, and preliminary<br />

evidence <strong>of</strong> activity, has been demonstrated in this first-in-human trial <strong>of</strong><br />

indenoisoquinoline-class <strong>of</strong> TOP1 inhibitors. Accrual is ongoing.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


3032 Poster Discussion Session (Board #24), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Phase Ib safety trial <strong>of</strong> CVX-060, an intravenous humanized monoclonal<br />

CovX body inhibiting angiopoietin 2 (Ang-2), with sunitinib. Presenting<br />

Author: Lee S. Rosen, UCLA School <strong>of</strong> Medicine, Santa Monica, CA<br />

Background: CVX-060 (CVX) is a humanized monoclonal antibody fused to<br />

two Ang2 binding peptides. The recommended phase 2 dose (RP2D) is15<br />

mg/kg/wk by intravenous (IV) infusion. Sunitinib (Sun), a kinase with many<br />

targets including VEGFR1-3, is dosed 50 mg/day orally x 4/6 wks. Methods:<br />

Patients (pts) with solid tumors were to receive CVX at doses <strong>of</strong> 6, 12, or 15<br />

mg/kg/wk with Sun 50 mg/day x 4/6 wks in successive cohorts. Standard<br />

definitions <strong>of</strong> dose limiting toxicities (DLTs) were used. Serum PK was<br />

assessed by ELISA. Results: Thirty-four pts were treated at 5 dose levels<br />

(DL) –see table. The median age was 62 years (29-76), ECOG performance<br />

status 0-1. The safety <strong>of</strong> CVX plus lower Sun dose was established in DL 1.1<br />

due to DLTs at the initial cohort. The safety <strong>of</strong> full dose Sun was further<br />

established in 1.2 before further escalating the CVX dose. Overall, DLTs<br />

related to CVX and Sun included hemorrhage (DL 1.0), asymptomatic<br />

proteinuria (DL 1.2), ischemic colitis/lower GI bleed/recurrent lower GI<br />

bleed (DL 3.0) and abdominal pain/microperforation on CT (DL 3.0). The<br />

most common adverse events (AEs) related to either agent were fatigue<br />

(68%), diarrhea (50%), nausea (47%), hypertension (35%), and dysgeusia<br />

(32%). The majority <strong>of</strong> AEs were Gr 1 to 2. There were no PK interactions<br />

between CVX and Sun. Total serum Ang2 (bound plus free) increased with<br />

CVX treatment. Of the 34 patients, 24 (71%) remained on study � 8 weeks<br />

and 2 patients continue on treatment � 9 months. Anti-drug antibodies<br />

were seen in 16 patients at low titers without effects on PK or safety.<br />

Conclusions: In pts with advanced disease, CVX is able to be combined at<br />

the RP2D with Sun at its labeled dose without exacerbation <strong>of</strong> Sunassociated<br />

toxicities. Combination trials <strong>of</strong> CVX and other VEGF inhibitors<br />

are planned.<br />

Dose levels evaluated.<br />

Dose level CVX-060 mg/kg/wk Sunitinib mg/d x 4/6 wks Pts<br />

1.0 6.0 50.0 6<br />

1.1 6.0 37.5 3<br />

1.2 6.0 50.0 6<br />

2.0 12.0 50.0 3<br />

3.0 15.0 50.0 16<br />

3034 General Poster Session (Board #11H), Mon, 8:00 AM-12:00 PM<br />

A phase Ib dose-escalation study <strong>of</strong> TRC105 in combination with bevacizumab<br />

for advanced solid tumors. Presenting Author: David S. Mendelson,<br />

Pinnacle Oncology Hematology, Scottsdale, AZ<br />

Background: CD105 (endoglin) is an endothelial cell membrane receptor<br />

highly expressed on angiogenic tumor vessels that is essential for angiogenesis<br />

and upregulated by hypoxia and VEGF inhibition. TRC105 is a<br />

human/mouse chimeric anti-CD105 monoclonal antibody that completed<br />

phase I testing and is being studied in multiple phase II trials. Methods: Pts<br />

with advanced solid tumors (non-CNS), ECOG PS 0-1, and normal organ<br />

function were treated with escalating doses <strong>of</strong> intravenously administered<br />

TRC105 plus bevacizumab (BEV) at 15 mg/kg q3wk or 10 mg/kg q2wk and<br />

assessed for safety, PK, and response. Results: Twelve pts were treated (3<br />

with TRC105 at 3 mg/kg q1wk � BEV at 15 mg/kg q3wk, 5 with TRC105 at<br />

6 mg/kg q1wk � BEV at 15 mg/kg q3wk, and 4 with TRC105 at 6 mg/kg<br />

q1wk � BEV at 10 mg/kg q2wk), and 10 have completed the DLT<br />

evaluation period. TRC105 at 6 mg/kg q1wk � BEV at 15 mg/kg q3wk<br />

resulted in grade 1-3 headaches with sinus congestion following the cycle 1<br />

day 1 (C1D1) dose in 5 <strong>of</strong> 5 pts that improved with continued dosing.<br />

Treatment with BEV 10 mg/kg q2wk and sequential dosing with Bev<br />

starting on C1D1 and TRC105 on C1D8 resulted in decreased frequency<br />

and severity <strong>of</strong> headaches, allowing dose escalation to continue. TRC105<br />

serum concentrations that saturate CD105 binding sites (�200 ng/mL)<br />

were achieved continuously at 6 mg/kg q1wk. At 3 mg/kg q1wk TRC105 �<br />

15 mg/kg q3wk BEV, one pt with BEV-resistant ovarian cancer had stable<br />

disease for 6 cycles and one pt with refractory hepatocellular cancer who<br />

failed sorafenib and sunitinib had an AFP decrease from 1578 at baseline<br />

to 179 at the end <strong>of</strong> cycle 3. Two <strong>of</strong> three evaluable pts at 6 mg/kg q1wk<br />

TRC105 � 15 mg/kg q3wk BEV with VEGF-refractory tumors (HCC and<br />

CRC) are ongoing in C4 with stable disease. Dose escalation continues and<br />

TRC105 dose levels <strong>of</strong> 8 and 10 mg/kg are planned. Conclusions: The<br />

combination <strong>of</strong> TRC105 and BEV was tolerated with early evidence <strong>of</strong><br />

activity in VEGF-refractory pts by sequential administration during initial<br />

dosing in C1.<br />

Developmental Therapeutics—Experimental Therapeutics<br />

181s<br />

3033 Poster Discussion Session (Board #25), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

NKP-1339: Maximum tolerated dose defined for first-in-human GRP78<br />

targeted agent. Presenting Author: Dana Shelton Thompson, Sarah Cannon<br />

Research Institute/Tennessee Oncology, PLLC, Nashville, TN<br />

Background: NKP-1339 is a first-in-class small molecule anti-cancer<br />

compound that down-regulates GRP78, a key regulator <strong>of</strong> misfolded<br />

protein processing and tumor survival/anti-apoptosis. GRP78 up-regulation<br />

occurs in many tumors, and is associated with intrinsic and chemotherapyinduced<br />

resistance. Preclinically, single agent NKP-1339 demonstrates<br />

activity against multiple tumor types, including chemoresistant lines. This<br />

phase I trial evaluates the safety, tolerability, maximum tolerated dose<br />

(MTD), pharmacokinetics, and pharmacodynamics (PD) <strong>of</strong> NKP-1339.<br />

Methods: Patients (pts) with advanced solid tumors, adequate organ<br />

function, ECOG 0-1 are enrolled. NKP-1339 is infused on day 1, 8, and 15<br />

<strong>of</strong> 28 day cycles. Single pt cohorts are enrolled until grade 2 toxicity, then<br />

adjusted to standard 3�3 design. Doses from 20-780 mg/m2 are evaluated.<br />

PD samples for plasma GRP78 are collected. At MTD, an expanded<br />

cohort <strong>of</strong> 25 pts is enrolled. Results: 34 pts are enrolled for dose escalation:<br />

30 evaluable for dose determination; 4 replaced due to tumor progression<br />

in cycle 1. Demographics: 19M/15F; median age 62 yrs (range 28 to 79).<br />

Tumor types: colorectal (CRC, 10), non-small cell lung (NSCLC, 8);<br />

neuroendocrine (NET, 5); head and neck (H&N, 3); and other cancer (8).<br />

Dose limiting toxicity <strong>of</strong> grade (gr) 2-3 nausea, with gr 2 creatinine in 1 pt,<br />

occurred at 780 mg/m2 ; MTD is 625 mg/m2 . Gr 1 fever/chills is observed<br />

above 420 mg/m2 , but is prevented by steroid premedication. The most<br />

common drug-related events are gr 1 nausea, gr 1-2 vomiting, and gr 1-2<br />

fatigue. No lab abnormalities were noted except reversible creatinine<br />

elevation in 4 pts: 3 pt with gr 1; 1 pt with gr 2 secondary to DLT. <strong>Part</strong>ial<br />

response was in 1 pt (NET); stable disease in 7 pts, including NET (2),<br />

NSCLC (2), CRC (1), sarcoma (1), and unknown primary (1). Therapy<br />

duration 14�-88� weeks. Soluble GRP78 is detected in the plasma <strong>of</strong><br />

patients at baseline. Conclusions: NKP-1339is well tolerated with manageable<br />

side effects. Single agent activity is noted in multiple tumors including<br />

NET. Results from the expanded cohort and pre/post-therapy PD will be<br />

presented. Phase II single agent NKP-1339 and phase I NKP-1339<br />

combination trials are planned.<br />

3035 General Poster Session (Board #12A), Mon, 8:00 AM-12:00 PM<br />

The effect <strong>of</strong> bevacizumab on targeting <strong>of</strong> anti-epidermal growth factor<br />

receptor and anti-insulin-like growth factor 1 receptor antibodies. Presenting<br />

Author: Sandra Heskamp, Department <strong>of</strong> Medical Oncology, Radboud<br />

University Nijmegen Medical Centre, Nijmegen, Netherlands<br />

Background: Bevacizumab and cetuximab are approved antibodies for<br />

treatment <strong>of</strong> patients with metastasized colorectal cancer. However, the<br />

combination <strong>of</strong> bevacizumab and cetuximab does not improve progression<br />

free survival (Tol et al. NEJM 2009). This may be explained by the<br />

disruption <strong>of</strong> tumor vascularity by bevacizumab, thereby reducing targeting<br />

<strong>of</strong> other antibodies to the tumor. The aim <strong>of</strong> this study was to determine the<br />

effect <strong>of</strong> bevacizumab on the targeting <strong>of</strong> anti-EGFR and IGF-1R antibodies<br />

in tumors with SPECT/CT imaging. Methods: Mice with subcutaneous EGFR<br />

and IGF-1R-expressing SUM149 xenografts were injected intraperitoneally<br />

with a single dose <strong>of</strong> bevacizumab (10 mg/kg). After four days, mice<br />

received an intravenous injection <strong>of</strong> 17 MBq 111In-labeled cetuximab, an<br />

anti-EGFR antibody, or R1507, an anti-IGF-1R antibody. A control group<br />

was injected with labeled hLL2 anti-CD22, an irrelevant IgG . Three days<br />

after injection, SPECT/CT images were acquired and mice were dissected<br />

for ex vivo biodistribution. Tumors were analyzed immunohistochemically<br />

to determine vascular density (CD34), EGFR and IGF-1R expression.<br />

Results: SPECT imaging revealed that bevacizumab treatment reduced<br />

targeting <strong>of</strong> anti-EGFR and anti-IGF-1R antibodies by 42% and 35%,<br />

respectively. Ex vivo biodistribution showed that uptake <strong>of</strong> 111In-cetuximab in untreated tumors was 35.2 � 1.6 %ID/g, compared with 19.7 � 5.3<br />

%ID/g for bevacizumab treated tumors (p � 0.009). A similar effect was<br />

observed for 111In-R1507 (control: 26.7 � 2.8 %ID/g, bevacizumab:18.9<br />

� 2.8 %ID/g, p �0.009). No significant differences in tumor uptake were<br />

observed in mice that received the irrelevant IgG. Immunohistochemical<br />

analysis showed that vascular density decreased with 40%, while EGFR<br />

and IGF-1R expression was unaltered. Conclusions: Bevacizumab treatment<br />

can significantly reduce targeting <strong>of</strong> other antibodies to tumors. This<br />

underlines the importance <strong>of</strong> timing and sequencing <strong>of</strong> bevacizumab<br />

therapy in combination with other antibodies.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


182s Developmental Therapeutics—Experimental Therapeutics<br />

3036 General Poster Session (Board #12B), Mon, 8:00 AM-12:00 PM<br />

Response <strong>of</strong> nonenhancing regions in glioblastoma to VEGF-signaling<br />

inhibitor cediranib correlates with tumor infiltration. Presenting Author:<br />

Summer A. Fakhro, Martinos Center for Biomedical Imaging, Massachusetts<br />

General Hopital, Charlestown, MA<br />

Background: Antiangiogenic therapy has limited direct antitumor effect in<br />

glioblastoma (GBM). Mainly its benefits may derive from anti-permeability<br />

and anti edema effects. In GBMs treated with cediranib we found that<br />

regions on MRI that are suggestive <strong>of</strong> being vasogenic edema, according to<br />

Diffusion Tensor Imaging (DTI) data, experience a greater decrease in<br />

volume than non-enhancing regions suggestive <strong>of</strong> being tumor infiltrated.<br />

Methods: Thirty-four newly diagnosed GBM patients were scanned on<br />

3Tesla Siemens at two pre-therapy time points (2 days apart). Patients were<br />

treated with cediranib, standard radiation and temozolomide. Due to<br />

technical difficulties, 4 patients were left out <strong>of</strong> the analysis. Diffusion<br />

images were acquired with TR�7500ms, TE�84ms and b-values 0 and<br />

700s/mm2 in 42 directions. A board certified radiologist determined the<br />

extent <strong>of</strong> abnormality on FLAIR and T1-weighted post contrast images.<br />

Non-enhancing regions (NER) were determined by subtracting the T1<br />

enhancing area and any necrosis from the FLAIR abnormality. Volume <strong>of</strong><br />

NERs was observed at baseline and day 47 <strong>of</strong> treatment. Mean Apparent<br />

Diffusion Coefficient (ADC) and mean Fractional Anisotropy (FA) <strong>of</strong> the<br />

NER were used to represent extent <strong>of</strong> tumor burden. Tumor Infiltration<br />

Index (TII), a measure based on comparing DTI metrics in metastatic<br />

tumors to GBM, was also derived. Twelve patients with metastatic cancer to<br />

the brain who underwent the same imaging and analysis pretreatment were<br />

used. High FA and low ADC and TII represent areas with more tumor<br />

burden. Results: There was a significant correlation between mean FA in the<br />

NERs at baseline and the percent volume reduction <strong>of</strong> the NER at day 47<br />

(rho�0.45, p�0.02). Similarly, ADC and TII were correlated with percent<br />

volume reduction in the NER. Conclusions: DTI metrics have been used in<br />

GBMs to measure tumor infiltration. We found volume reduction <strong>of</strong> NERs to<br />

be inversely correlated to tumor burden. Thus, glioma patients with greater<br />

vasogenic edema may benefit from a different treatment paradigm than<br />

patients with lower vasogenic edema.<br />

% Volume reduction <strong>of</strong> NER at day 47 vs<br />

ADC mean FA mean TII mean<br />

Spearman correlation (rho) -0.33 0.45 -0.44<br />

p-value 0.083 0.015 0.017<br />

3038 General Poster Session (Board #12D), Mon, 8:00 AM-12:00 PM<br />

Phase I study <strong>of</strong> safety and pharmacokinetics <strong>of</strong> fruquintinib, a selective<br />

inhibitor <strong>of</strong> VEGF receptor-1, -2, and -3 tyrosine kinases in patients with<br />

advanced solid tumors. Presenting Author: Jin Li, Fudan University<br />

Shanghai Cancer Center, Shanghai, China<br />

Background: Fruquintinib is a novel oral small molecule compound discovered<br />

and developed by Hutchison MediPharma that selectively inhibits<br />

vascular endothelial growth factor receptors (VEGFR) 1, 2, and 3 and has<br />

demonstrated potent inhibitory effects on multiple human tumor xenografts.<br />

This first-in-human study is conducted to assess the maximum<br />

tolerated dose (MTD) and dose-limiting toxicity (DLT), safety and tolerability,<br />

pharmacokinetics (PK), and preliminary anti-tumor activity <strong>of</strong> fruquintinib.<br />

Methods: This phase I study used the 3�3 design for dose-escalation<br />

<strong>of</strong> fruquintinib given once daily (QD) in 28-day cycles in patients with solid<br />

tumors who had failed standard therapies. Endpoints included safety, PK,<br />

and preliminary efficacy measurements. Results: To date 16 patients were<br />

enrolled in 5 dose cohorts <strong>of</strong> 1-6 mg QD, with age 42-69 yr, 44% male,<br />

ECOG 0-1 and all heavily pretreated. Tumor types included 9 colorectal, 3<br />

lung, 3 breast, and 2 gastric (one patient with bi-primary carcinoma <strong>of</strong><br />

breast and colon). The most common adverse events included hypertension<br />

(50%), proteinuria (43.8%), hand-foot syndrome (HFS 43.8%), diarrhea<br />

(31.2%), and hoarseness (25%). Grade 3/4 AEs were diarrhea (25%), HFS<br />

(12.5%), thrombocytopenia (6.2%), and hyperbilirubinemia (6.2%). Three<br />

DLTs were observed: 2 grade 3 HFS both at 6mg and 1 grade 3<br />

hyperbilirubinemia at 4mg. PK analysis showed good and rapid absorption<br />

followed by slow terminal elimination with a half-life <strong>of</strong> approximately 37<br />

hours which was consistent across all dose groups. Both Cmax and AUC<br />

exhibited good dose proportionality over the studied dose range with low<br />

inter-patient variability following single and multiple doses. Among 8<br />

evaluable patients, 2 (1 colorectal and 1 gastric) had confirmed partial<br />

response for more than 4 months; 5 had stable disease, including 3 SDs <strong>of</strong><br />

3-8 months. Conclusions: Fruquintinib was well tolerated at doses up to 4<br />

mg QD to date and demonstrated excellent pharmacokinetic properties.<br />

Encouraging clinical activity was observed. Further clinical studies are<br />

warranted and under planning.<br />

3037 General Poster Session (Board #12C), Mon, 8:00 AM-12:00 PM<br />

Phase I study <strong>of</strong> vandetinib in combination with gemcitabine and oxaliplatin<br />

in advanced solid malignancies. Presenting Author: Peng Wang,<br />

University <strong>of</strong> Pittsburgh Cancer Institute, Pittsburgh, PA<br />

Background: Vandentinib (VAN) is a tyrosine kinase inhibitor with activity<br />

against vascular endothelial growth factor receptor-2 (VEGFR-2) and 3<br />

(VEGFR-3) as well as epidermal growth factor receptor (EGFR) and RET<br />

(‘rearranged during transfection’). Combinations <strong>of</strong> VEGFR kinase inhibitors<br />

and cytotoxic agents have been studied with interest, but have been<br />

associated with increased toxicity. This phase I trial evaluated the safety,<br />

tolerability and maximum tolerated dose <strong>of</strong> VAN in combination with<br />

gemcitabine (GEM) and oxaliplatin (OX). Methods: Subjects with advanced<br />

solid malignancy, � 2 prior regimens with adequate hematologic, hepatic<br />

and renal function were eligible. GEM and OX were administered intravenously<br />

at 1000 mg/m2 and 85 mg/m2 respectively every 14 days. VAN was<br />

administered orally at two dose levels: level 1 (200 mg) and level 2 (300<br />

mg) once daily continuously. After a maximum <strong>of</strong> 6 cycles <strong>of</strong> GEMOX,<br />

non-progressing patients were allowed to continue VAN until progression or<br />

intolerance. A dose level was determined to be tolerable if �2 DLTs out <strong>of</strong> 6<br />

evaluable subjects were observed. Response was assessed by restaging<br />

scan after every 2 cycles. Results: A total <strong>of</strong> 20 subjects were enrolled in the<br />

study; 15 male, 5 female. Median age 62 years (47-68). Nine subjects<br />

were enrolled on dose level 1 and 11 subjects on dose level 2. One subject<br />

in dose level 1 experienced a deep venous thrombosis in cycle 1 deemed a<br />

DLT. Thrombocytopenia (worst-grade 3) as well as diarrhea and rash<br />

(worst-grade 2) were observed in several patients, without dose-limiting<br />

toxicity in cycle 1. Reversible posterior leukoencephalopathy was observed<br />

in 1 subject following abrupt self-discontinuation <strong>of</strong> an antihypertensive<br />

mediation. The recommended phase 2 dose was established as GEM<br />

(1000mg/m2 ) OX (85mg/m2 ) and VAN (300mg). One subject with refractory<br />

small cell lung cancer had a confirmed partial response, and 10<br />

subjects (pancreas-7, bladder-2 and biliary-1) achieved stable disease<br />

�12 weeks. Conclusions: VAN in combination with GEM/OX was well<br />

tolerated at full doses <strong>of</strong> each <strong>of</strong> the 3 agents. Preliminary anti-tumor<br />

activity seen in refractory solid tumors supports further evaluation <strong>of</strong> this<br />

regimen.<br />

3039 General Poster Session (Board #12E), Mon, 8:00 AM-12:00 PM<br />

MGCD265, a multitargeted oral tyrosine kinase receptor inhibitor <strong>of</strong> Met<br />

and VEGFR: Dose-escalation phase I study. Presenting Author: Christian K.<br />

Kollmannsberger, British Columbia Cancer Agency, Vancouver, BC, Canada<br />

Background: MGCD265 is a multikinase inhibitor, with nanomolar IC50 against Met, VEGFR 1, 2, and 3, Tie-2, and Ron. This spectrum may confer<br />

greater anti-tumor activity than inhibiting either target alone. MGCD265<br />

has broad anti-tumor effects in preclinical models. Methods: Patients (pts)<br />

with advanced malignancies were enrolled in this Phase I, open-label, dose<br />

escalating study using the classic 3�3 study design. MGCD265 was<br />

administered every day over a 21-day cycle. The aim <strong>of</strong> this study is to<br />

determine the safety pr<strong>of</strong>ile including the maximum tolerated dose and the<br />

dose limiting toxicities (DLTs) <strong>of</strong> MGCD265. The pharmacokinetic (PK),<br />

pharmacodynamic (PD) pr<strong>of</strong>iles and the anti-tumor activity <strong>of</strong> MGCD265<br />

were also evaluated. Results: As <strong>of</strong> January 10, 2012, 56 patients were<br />

enrolled (M/F: 37/19; ECOG 0/1/2: 16/38/2; median age: 61 years old).<br />

MGCD265 was dose escalated from 24 mg/m2 QD to 235 mg/m2 BID with a<br />

favorable safety pr<strong>of</strong>ile. The DLTs (n�1 for each event) captured were:<br />

grade 2 hypertension (per protocol’s definition), grade 3 lipase elevation,<br />

grade 3 fatigue and grade 3 pituitary hemorrhage, all occurring at daily<br />

doses � 250 mg/m2 . Additional � grade 3 drug-related adverse events<br />

(observed beyond Cycle 1) were diarrhea, fatigue, elevated lipase and<br />

elevated alkaline phosphatase (n�1 for each event). Two schedules <strong>of</strong><br />

MGCD265 were tested sequentially: once (QD) and twice daily (BID),<br />

respectively. Increasing the dose frequency to BID increased the exposure<br />

<strong>of</strong> MGCD265 by ~ 2 fold at steady state. To date, the exposure (Cmax) attained at steady state with the BID schedule was ~20 fold higher than the<br />

IC50 for Met and VEGFR inhibition and was also above the plasma<br />

exposures associated with efficacy in the Met-sensitive xenograft model<br />

(MKN45). Six patients achieved SD for 6 cycles or more (up to 12 cycles).<br />

Conclusions: The safety pr<strong>of</strong>ile <strong>of</strong> MGCD265 continues to be favorable in<br />

this Phase I dose escalating study. While no evidence <strong>of</strong> objective<br />

responses have been noted to date, several pts have had prolonged SD.<br />

Dose escalation is ongoing.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


3040 General Poster Session (Board #12F), Mon, 8:00 AM-12:00 PM<br />

First-in-human phase I study <strong>of</strong> a selective VEGFR/FGFR dual inhibitor<br />

sulfatinib in patients with advanced solid tumors. Presenting Author:<br />

Jian-Ming Xu, Cancer Center, 307 Hospital, Academy <strong>of</strong> Military Medical<br />

Science, Beijing, China<br />

Background: Sulfatinib is a highly selective oral small molecule tyrosine<br />

dual inhibitor <strong>of</strong> vascular endothelial growth factor receptors (VEGFR) and<br />

fibroblast growth factor receptors (FGFR). It demonstrated potent in vivo<br />

inhibitory effects on a variety <strong>of</strong> human tumor xenografts. Methods: This<br />

phase I dose-escalation study were to determine the maximum tolerated<br />

dose (MTD), dose-limiting toxicities (DLTs), pharmacokinetics (PK) pr<strong>of</strong>ile,<br />

and preliminary antitumor activity, and to assess potential pharmacodynamics<br />

(PD) markers <strong>of</strong> sulfantinib when given orally in continuous cycles <strong>of</strong> 28<br />

days, until disease progression or unacceptable toxicity. Results: Forty one<br />

patients (pts) have been enrolled and treated with doses <strong>of</strong> 50-300mg once<br />

or twice daily. Pts had a median age <strong>of</strong> 50 (36-70) yrs, with 63% male and<br />

93% gastrointestinal tumors. Common adverse events (AEs) included<br />

fatigue, hypertension, vomiting, nausea, diarrhea, electrolyte disorder and<br />

elevated AST/ALT, mostly grade (G) 1/2. Three DLTs including a G3<br />

coagulation disorders, upper gastrointestinal bleeding and impaired liver<br />

function were observed in the 50 and 265mg qd and 150mg bid cohorts,<br />

respectively. MTD has not been reached and the dose escalation is ongoing.<br />

Among 28 evaluable pts, 10 (36%) had stable disease (SD) for 8 weeks or<br />

longer, including liver and neuroendocrine cancer, GIST and solidpseudopapillary<br />

tumor <strong>of</strong> pancreas. A patient with GIST had SD <strong>of</strong> 9<br />

months at 265mg qd. There was a significant decrease in soluble VEGFR2<br />

level and increase in FGF23 level after 4-week treatment comparing to the<br />

baseline at 265 mg or higher daily doses, indicating inhibition <strong>of</strong> both<br />

VEGFR2 and FGFR. PK analyses showed that sulfatinib was rapidly<br />

absorbed with Tmax 1.3~2.8h and half-life <strong>of</strong> 15.3~19.1h. Both Cmax and<br />

AUC displayed dose-proportional increases and no obvious accumulation<br />

occurred at day 28. Conclusions: Sulfatinib was well tolerated at doses up to<br />

300 mg per day and demonstrated preliminary anti-tumor activity, especially<br />

in liver cancer and GIST. PD marker analysis indicates dual inhibition<br />

<strong>of</strong> VEGFR and FGFR. PK data suggests good dose proportionality in<br />

exposure without marked drug accumulation.<br />

3042^ General Poster Session (Board #12H), Mon, 8:00 AM-12:00 PM<br />

Dose-specific clearance <strong>of</strong> TRC105 (anti-CD105 antibody) in advanced<br />

solid tumor patients. Presenting Author: Shawn D. Spencer, SAIC-<br />

Frederick, National Cancer Institute at Frederick, Bethesda, MD<br />

Background: CD105 is an endothelial cell membrane receptor highly<br />

expressed on angiogenic tumor vessels. TRC105 is an anti-CD105 monoclonal<br />

antibody that inhibits angiogenesis and tumor growth by endothelial<br />

cell growth inhibition, ADCC and apoptosis. Methods: Patients with advanced<br />

solid tumors and normal organ function were treated with escalating<br />

doses <strong>of</strong> intravenously administered TRC105 and assessed for safety and<br />

pharmacokinetics (PK). PK parameters for determining dose-linearity were<br />

estimated following single doses on in 16 patients at 3, 10 and 15 mg/kg<br />

and correlated with safety. Results: TRC105 was tolerated at 10 mg/kg<br />

weekly and 15 mg/kg every 2 weeks, but hyproliferative anemia was<br />

dose-limiting at 15 mg/kg weekly and was associated with TRC105<br />

accumulation in serum from target saturation. Preliminary evidence <strong>of</strong><br />

activity was observed with 21 <strong>of</strong> 45 evaluable patients progression-free at 2<br />

months, including two ongoing responders. The AUC-single dose relationship<br />

<strong>of</strong> TRC105 revealed supra-proportionality in serum exposure at 15<br />

mg/kg compared to 3 and 10 mg/kg. Dose proportionality using a power<br />

model revealed a nonlinear pharmacokinetic process (log[AUC] �<br />

0.125*Dose�2.67, r2 � 0.92, ANOVA p�0.0035). The steady state<br />

volume <strong>of</strong> distribution was low (� 5.40 L/70kg) indicating TRC105 was<br />

confined to the vasculature with a low capacity target (i.e., low relative<br />

abundance) making it susceptible to saturation. The post-infusion Cmax<br />

however was linearly related to dose (r � 0.85) which suggested the<br />

nonlinearity in clearance was attributed to target-mediated disposition. The<br />

nonlinear disposition in the presence <strong>of</strong> low distribution indicated TRC105<br />

bound avidly to endothelial cells. Concentrations expected to saturate<br />

CD105 binding were achieved continuously at 15 mg/kg dosed every 2<br />

weeks and 10 mg/kg/wk. TRC105 accumulated at 15 mg/kg/wk as the<br />

accumulation factor at 56 days was 1.77-fold over single doses on C1D1.<br />

Conclusions: TRC105 clearance decreased above the MTD resulting in drug<br />

accumulation and hypoproliferative anemia with weekly dosing. The<br />

nonlinearity in clearance was attributed to saturating target-mediated<br />

disposition, consistent with binding to proliferating endothelial cells.<br />

Developmental Therapeutics—Experimental Therapeutics<br />

183s<br />

3041 General Poster Session (Board #12G), Mon, 8:00 AM-12:00 PM<br />

Phase I, first-in-human trial <strong>of</strong> an oral VEGFR tyrosine kinase inhibitor (TKI)<br />

x-82 in patients (pts) with advanced solid tumors. Presenting Author:<br />

Kathleen N. Moore, Sarah Cannon Research Institute/University <strong>of</strong> Oklahoma<br />

Health Sciences Center, Oklahoma City, OK<br />

Background: VEGFR TKIs have shown benefit in the treatment <strong>of</strong> various<br />

tumor types. Side effects <strong>of</strong> the TKIs have affected the duration <strong>of</strong> therapy<br />

pts can tolerate, as well as the combinability with chemotherapy. X-82 is a<br />

highly potent VEGFR/PDGFR TKI with a smaller volume <strong>of</strong> distribution and<br />

limited tissue accumulation designed to minimize side effects while<br />

maintaining target effect. Methods: Pts with advanced solid tumors were<br />

enrolled using an accelerated titration scheme followed by 3�3 dose<br />

escalation design. X-82 was administered orally once daily (QD) or twice<br />

daily (BID) every 28 days. Safety, pharmacokinetic, clinical endpoints and<br />

blood-based biomarkers were evaluated. Results: 16 pts were treated across<br />

8 dose levels: 20 (n�1), 40 (n�1), 80 (n�1), 160 (n�1), 300 (n�2), 400<br />

mg QD (n�3), 140 mg BID (n�3) and 200 mg BID (n�4). The most<br />

common related adverse events (AEs) by pt were fatigue (4 G1, 1 G2),<br />

nausea (3 G1, 1 G2), diarrhea (3 G1), anorexia (1 G1, 1 G2), and vomiting<br />

(2 G1). G2 hypertension, resolved with treatment, was experienced by 1<br />

patient. No � G3 related AEs have been reported. Dose proportional<br />

exposure was observed with doses up to 160 mg QD, and BID dosing<br />

cohorts were added. At 160 mg QD X-82 has plasma concentrations �100<br />

ng/mL for at least 12 hours, the desired exposure predicted preclinically.<br />

Preliminary blood biomarker data (n�8) shows �25% decrease in VEGFR1<br />

in 62% <strong>of</strong> pts and in VEGFR2 in 25% <strong>of</strong> pts, including pts at 40 and 80 mg<br />

cohorts. Efficacy has also been noted, including 1 confirmed complete<br />

response lasting 24� weeks (pancreatic adenocarcinoma), and 6 pts with<br />

stable disease lasting 8� weeks (range 12.1�, 38.3� weeks), including<br />

the pt at 20 mg with SCLC (25.1 weeks). Conclusions: X-82 is a<br />

well-tolerated VEGFR/PDFGR TKI designed to minimize side effects while<br />

inhibiting target receptors. No MTD has yet been defined, though preliminary<br />

biomarker data are encouraging and clinical efficacy has been noted.<br />

3043 General Poster Session (Board #13A), Mon, 8:00 AM-12:00 PM<br />

A phase I study <strong>of</strong> TRC105 (Anti-CD105 [endoglin] antibody) in metastatic<br />

castration resistant prostate cancer (mCRPC). Presenting Author: Fatima<br />

H. Karzai, Medical Oncology Branch, National Cancer Institute, National<br />

Institutes <strong>of</strong> Health, Bethesda, MD<br />

Background: Pre�clinical and clinical evidence demonstrates an important<br />

role for angiogenesis in mCRPC biology. CD105 (endoglin) is a transmembrane<br />

protein expressed on the surface <strong>of</strong> proliferating vascular endothelial<br />

cells. The expression <strong>of</strong> CD105 is required for the formation <strong>of</strong> new blood<br />

vessels. TRC105 is a human/murine chimeric IgG1 kappa monoclonal<br />

antibody that binds to human CD105 (endoglin). It inhibits angiogenesis<br />

and tumor growth through inhibition <strong>of</strong> endothelial cell proliferation,<br />

antibody-dependent cellular cytotoxicity, and induction <strong>of</strong> apoptosis. The<br />

primary objective is to evaluate safety and identify the maximum tolerable<br />

dose (MTD) <strong>of</strong> TRC105. Secondary objectives include the assessment <strong>of</strong><br />

TRC105 pharmacokinetics, PSA response rate, evaluation <strong>of</strong> progression<br />

free survival (PFS), overall response rate (ORR) and overall survival (OS).<br />

Methods: Patients with an ECOG performance status (PS) � 2, progressive<br />

mCRPC and either chemotherapy-naïve or post-docetaxel treatment were<br />

eligible. Six cohorts <strong>of</strong> patients, on escalating dose levels, receive TRC105<br />

intravenously at doses <strong>of</strong> 1, 3, 10, 15, or 20 mg/kg IV every 2 weeks<br />

(cohorts 1, 2, 3, 5, and 6) or 10 mg/kg IV weekly (cohort 4) on a 4 week<br />

cycle. Response is assessed with imaging studies every 2 months for the<br />

first four months and then every 3 months thereafter. Results: Sixteen<br />

patients are enrolled in cohorts 1-5. Median age is 65 (range 48-87),<br />

median ECOG PS is 1 (range 0�2), median Gleason score is 8 (range<br />

6�10), median on�study PSA is 147.5 (range 0.1-3373), and median<br />

number <strong>of</strong> prior (non-hormonal) therapies is 3 (range 0�6). Median time on<br />

study is 16 weeks (range 8-28 weeks). One patient experienced a dose<br />

limiting toxicity (grade 4 vasovagal episode) in cohort 5. PSA declines were<br />

seen in 6 patients ranging from 20% to 57% from baseline. Ten out <strong>of</strong> 12<br />

patients with measurable s<strong>of</strong>t tissue disease achieved stable disease for at<br />

least two cycles. Conclusions: TRC105 is tolerated up to 15 mg/kg every two<br />

weeks with early evidence <strong>of</strong> clinical activity in mCRPC. An additional<br />

cohort (6), with dosage <strong>of</strong> 20 mg/kg, is currently under investigation.<br />

Accrual is ongoing to evaluate ORR, PFS, and OS in the phase II portion <strong>of</strong><br />

this study.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


184s Developmental Therapeutics—Experimental Therapeutics<br />

3044 General Poster Session (Board #13B), Mon, 8:00 AM-12:00 PM<br />

SORAVE: Phase I study for the treatment <strong>of</strong> relapsed solid tumors with the<br />

combination <strong>of</strong> sorafenib and everolimus. Presenting Author: Lucia Nogova,<br />

Lung Cancer Group Cologne, Center for Integrated Oncology, University<br />

Hospital Cologne, Cologne, Germany<br />

Background: Dual inhibition <strong>of</strong> signaling pathways interfering with angiogenesis<br />

and cell proliferation may increase anti-tumor efficacy. We evaluated<br />

the combination <strong>of</strong> the VEGFR inhibitor sorafenib (S) and the mTOR<br />

inhibitor everolimus (E) and used FDG-PET to assess pharmacodynamic<br />

(PD) activity <strong>of</strong> E. Methods: Patients with relapsed solid tumors were treated<br />

with escalating doses <strong>of</strong> E 2.5-10.0mg/d p.o. in a 14 days monotherapy run<br />

in phase followed by combination therapy with a fixed dose <strong>of</strong> S 400mg bid<br />

from day 15. The primary aim was to define a safe and feasible combination<br />

treatment regimen for a subsequent phase II trial. DLT was defined as any<br />

drug related toxicity <strong>of</strong> CTC IV° or toxicity requiring hospitalization or<br />

interruption <strong>of</strong> therapy for more than 2 weeks within the first 29 days <strong>of</strong><br />

treatment. Pharmacokinetic (PK) analyses were performed on day 5, 14<br />

and 29 combined with explorative PD assessment <strong>of</strong> E by FDG-PET on days<br />

1, 5 and 14 <strong>of</strong> treatment. Results: Nineteen patients were treated with the<br />

combination <strong>of</strong> E and S. The DLT was not reached according to protocol<br />

definition. However, at a dose level <strong>of</strong> E 10mg/d p.o. the following adverse<br />

events (AE) occurred in the non-DLT interval: Pneumonia III°, treatment<br />

delay due to leucopoenia/thrombopenia III° and sudden cardiac death<br />

probably due to arrhythmia. Based on these observations, the dose level <strong>of</strong><br />

7.5mg/day E p.o. in combination with 400mg S bid was defined as the<br />

MTD. The steady state <strong>of</strong> E alone was reached after 5 days <strong>of</strong> treatment and<br />

did not change until day 14. However, on day 29 everolimus plasma<br />

concentrations (AUC and Cmax) showed a significant 30 % reduction when<br />

co-administered with S. A metabolic response <strong>of</strong> the hottest lesion in PET<br />

on day 5 defined as � 80% <strong>of</strong> baseline was seen in 4 <strong>of</strong> 15 evaluable<br />

patients. Median PFS and OS were 4.2 and 5.4 months, respectively. A<br />

patient with neuroendocrine carcinoma reached the longest PFS <strong>of</strong> 16.6<br />

months. Conclusions: Treatment <strong>of</strong> patients with relapsed solid tumors with<br />

a combination <strong>of</strong> E 7.5mg/day and 400mg S bid is safe and feasible. The<br />

extension phase for confirmation <strong>of</strong> safety data, further PK and PD<br />

modeling and preliminary exploration <strong>of</strong> efficacy in KRAS mutant cancer<br />

patients will be performed at this dose level.<br />

3046 General Poster Session (Board #13D), Mon, 8:00 AM-12:00 PM<br />

A dose-escalation phase I study <strong>of</strong> oral pan-CDK inhibitor BAY 1000394 in<br />

patients with advanced solid tumors: Dose escalation with an intermittent<br />

28 days on/14 days <strong>of</strong>f schedule. Presenting Author: Juneko E. Grilley-<br />

Olson, University <strong>of</strong> North Carolina at Chapel Hill, Chapel Hill, NC<br />

Background: BAY 1000394 (BAY) is an oral pan-CDK inhibitor targeting<br />

CDKs 1, 2, 4, 7, and 9 in the low nanomolar range. A phase I dose<br />

escalation multicenter study was initiated to determine the maximum<br />

tolerated dose (MTD), pharmacokinetics (PK), and pharmacodynamics<br />

(PD) in patients (pts) with advanced solid tumors. Methods: BAY was<br />

administered twice daily as an oral solution on a 28 days on / 14 days <strong>of</strong>f<br />

schedule (cycle length 42 days, 3�3 design). PK was evaluated on cycle 1<br />

day 1, 2, and 15. Response rate was assessed according to RECIST 1.1.<br />

Results: Ten pts were treated at doses <strong>of</strong> 0.6 (4 pts) and 1.0 (6 pts) mg per<br />

day. Tumor types included 3 non-small cell lung, 2 colorectal, 2 melanoma<br />

and 3 others. CTCAEv4 grade 1/2 drug related adverse events (AEs)<br />

occurring in more than 3 patients were nausea (5 pts), hot flashes (4),<br />

vomiting, diarrhea, dyspepsia, and fatigue (3). The median interval<br />

between start <strong>of</strong> treatment and occurrence <strong>of</strong> hot flashes was 23 days.<br />

Drug-related grade 3 AEs were hyponatremia (2 pts) and edema, lymphopenia,<br />

myalgia, fatigue, and hypokalemia in 1 pt each. Hyponatremia in one<br />

and hypokalemia in another patient were dose limiting toxicities in cohort<br />

2. Enrollment has been stopped. BAY PK was dose proportional and T1/2 was 13 hours; major metabolite levels were low. One pt with metastatic<br />

malignant melanoma pretreated with interferon, talimogene laherparepvec,<br />

and ipilimumab (best prior response: progressive disease) achieved stable<br />

disease (SD) lasting for 5 months. Three pts had SD lasting for 2.5 - 3<br />

months (thyroid, colorectal, squamous esophageal). Conclusions: BAY<br />

administered for 28 days on / 14 days <strong>of</strong>f demonstrated a limited<br />

tolerability. This is in contrast to the ongoing 3 days on / 4 days <strong>of</strong>f trial<br />

which is currently at a dose level <strong>of</strong> 20 mg per day. Hot flashes were an<br />

infrequent AE in the other dosing schedule but occurred in 4 <strong>of</strong> the 10 pts<br />

presented here. The long interval between start <strong>of</strong> treatment and occurrence<br />

<strong>of</strong> hot flashes suggests that the 28 days <strong>of</strong> continuous treatment<br />

contributed to the lower tolerability observed in this trial. The 3 days on / 4<br />

days <strong>of</strong>f schedule will be used in the further clinical development <strong>of</strong> BAY.<br />

3045 General Poster Session (Board #13C), Mon, 8:00 AM-12:00 PM<br />

Phase I dose-escalation study <strong>of</strong> AZD7762 alone and in combination with<br />

gemcitabine in Japanese patients with advanced solid tumors. Presenting<br />

Author: Takashi Seto, National Kyushu Cancer Center, Fukuoka, Japan<br />

Background: AZD7762, a potent Chk1/Chk2 inhibitor, has been shown to<br />

enhance the antitumor activity <strong>of</strong> gemcitabine in xenograft models (Zablud<strong>of</strong>f<br />

SD et al. Mol Cancer Ther 2008;7:2955–66). Methods: This open-label<br />

dose-escalation study evaluated the safety, pharmacokinetics (PK), and<br />

preliminary efficacy (RECIST) <strong>of</strong> AZD7762 alone and in combination with<br />

gemcitabine in Japanese patients (pts) with advanced solid tumors<br />

(NCT00937664). Pts received AZD7762 iv alone on days 1 and 8 <strong>of</strong> a<br />

14-day cycle (Cycle 0), followed by AZD7762 plus gemcitabine 1000<br />

mg/m2 on days 1 and 8 <strong>of</strong> 21-day cycles, in sequential ascending AZD7762<br />

dose cohorts. Results: 20 pts (mean age 60 years) received AZD7762 at<br />

doses <strong>of</strong> 6 (n�3), 9 (3), 21 (6), and 30 mg (8). The most common primary<br />

tumor site was lung (n�14). All pts had received �1 prior chemotherapy<br />

and 18 had metastatic disease. Dose-limiting toxicities (DLTs) occurred in<br />

two <strong>of</strong> six evaluable pts (both 30 mg cohort): one, grade 3 (CTCAE, v3.0)<br />

elevated troponin T (Cycle 0; AZD7762 monotherapy); one, neutropenia,<br />

thrombocytopenia, and elevated AST and ALT (Cycle 1; combination<br />

therapy). Thus, the 30 mg dose was regarded as non-tolerable. DLTs<br />

resolved following treatment discontinuation. The most frequently reported<br />

adverse events (AEs) in Cycle 0 (AZD7762 monotherapy) were bradycardia<br />

(50%), hypertension (25%) and fatigue (15%). Overall, the most common<br />

AEs were bradycardia (55%), neutropenia (45%), and hypertension,<br />

fatigue, and rash (30% each). AEs grade �3 were reported in 11 pts, the<br />

most common being neutropenia (45%) and leukopenia (25%). No pt died<br />

due to an AE. AZD7762 exposure (Cmax, AUC) increased in an approximately<br />

linear manner. Gemcitabine did not appear to affect AZD7762 PK.<br />

Arithmetic mean t½ and geometric mean CL <strong>of</strong> AZD7762 across the dose<br />

groups were 16.1–19.4 h and 22.0–32.7 L/h, respectively during the<br />

monotherapy cycle, and 15.6–18.3 h and 21.1–24.4 L/h, respectively in<br />

combination with gemcitabine. There were no objective responses; five pts<br />

(all lung cancer) had stable disease. Conclusions: The maximum tolerated<br />

dose <strong>of</strong> AZD7762 in combinationwith gemcitabine 1000 mg/m2 was<br />

determined as 21 mg in Japanese pts.<br />

3047 General Poster Session (Board #13E), Mon, 8:00 AM-12:00 PM<br />

<strong>Clinical</strong> and correlative science results in a phase II study <strong>of</strong> UCN-01 in<br />

combination with irinotecan in recurrent triple-negative breast cancer<br />

(TNBC). Presenting Author: Cynthia X. Ma, Washington University School<br />

<strong>of</strong> Medicine, St. Louis, MO<br />

Background: Chk1 inhibitors enhance chemotherapy efficacy by inducing<br />

“mitotic catastrophe” in p53 deficient TNBC preclinical models. Irinotecan<br />

(I) combined with UCN-01, a nonselective Chk1 inhibitor, showed<br />

promising activity in TNBC in our phase I study. The primary objective <strong>of</strong><br />

the phase II trial was to determine the efficacy and toxicity. Correlatives<br />

included assessing tumor molecular subtype, TP53, PTEN and pathways<br />

targeted by UCN-01. Methods: Pts with measurable, metastatic (met)<br />

TNBC, prior anthracycline (A) and taxane (T), received I (100-125 mg/m2 IV on days (d) 1, 8, 15, 22) and UCN-01 (70 mg/m2 IV on d2 and 35 mg/m2 on d23 and later doses) on a 42-d cycle (C). Archival tumors and serial<br />

peripheral blood mononuclear cells (PBMC) and optional tumor biopsies<br />

were collected. Results: Twenty five pts were enrolled. All had prior A and T.<br />

The median no. <strong>of</strong> prior regimens for met disease was 3 (range 1-4).<br />

Toxicities included neutropenia, diarrhea, nausea, vomiting, and hyperglycemia.<br />

Best responses included 1 PR, 8 SD (range 2.3-8.6 mos) for a<br />

clinical benefit rate (CR�PR�SD�6 mos) <strong>of</strong> 3/25 (12%), 95% CI (3,<br />

31%). The median PFS and OS were 2.3 and 11.3 mos, respectively. pS6<br />

was examined since UCN-01 inhibits PDK1. pS6 was reduced in PBMC<br />

24h post UCN-01, but close to baseline by d8. Immunostain <strong>of</strong> cleaved<br />

caspase 3 (CC3), pHistone H3 (pHH3), �H2AX, and pS6 were done on<br />

serial biopsies from 4 pts with adequate biopsy materials. In all cases, pS6<br />

was reduced 24h post UCN-01. Results for other markers were variable.<br />

One case with TP53 deletion showed an induction <strong>of</strong> CC3, with an increase<br />

in pHH3 and �H2AX, suggesting abrogation <strong>of</strong> cell cycle arrest and<br />

enhanced DNA damage. Among 15 with sufficient specimen for analysis,<br />

most were basal-like (basal 10, basal/HER2-E 1, HER2-E 2, Luminal B 2)<br />

by PAM50, low in PTEN level (11) and carried mutations in TP53 (8).<br />

Median OS was 5.5 (95% CI: 2, 11.3) mos in TP53 mutant and 20.3 (95%<br />

CI: 2.9, - ) mos in wild type populations (p�0.004). Conclusions: This<br />

regimen had limited activity in TNBC. Despite the long half-life, drug<br />

activity is not detectable by d8 based on PBMC analysis. Our data indicates<br />

that future trials in TNBC should consider p53 status.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


3048 General Poster Session (Board #13F), Mon, 8:00 AM-12:00 PM<br />

Safety and efficacy results from two randomized expansions <strong>of</strong> a phase I<br />

study <strong>of</strong> a tablet formulation <strong>of</strong> the PARP inhibitor, olaparib, in ovarian and<br />

breast cancer patients with BRCA1/2 mutations. Presenting Author: L<br />

Rhoda Molife, Royal Marsden Hospital and Institute <strong>of</strong> Cancer Research,<br />

Surrey, United Kingdom<br />

Background: We previously reported the comparative bioavailability <strong>of</strong> the<br />

capsule (CAP) formulation <strong>of</strong> olaparib and the more convenient new tablet<br />

(TAB) formulation (Molife et al ASCO 2010). We subsequently performed<br />

two separate dose expansions (DE1 and DE2) to explore comparative safety<br />

and efficacy <strong>of</strong> the TAB (NCT00777582). Methods: Patients with breast or<br />

ovarian cancer and BRCA1/2 mutations, ECOG PS 0–2 and adequate organ<br />

function were randomized to receive: DE1: 200 TAB or 400 CAP; DE2: 300<br />

TAB, 400 TAB or 400 CAP (all mg BID). Endpoints included safety,<br />

pharmacokinetics and exploratory analysis <strong>of</strong> efficacy (change in tumor size<br />

at 8 weeks by RECIST 1.0). Groups were compared using analysis <strong>of</strong><br />

covariance, including baseline tumor size and treatment as covariates;<br />

results are presented using least square (LS) means. Results: 24 patients<br />

(ovarian n�15, breast n�9) entered DE1 and 53 patients (ovarian n�38,<br />

breast n�15) entered DE2. Baseline characteristics including age, BRCA<br />

mutation status and tumor histology were balanced. The table shows key<br />

toxicity-related information and change in tumor size at 8 weeks by cohort.<br />

Conclusions: These data suggest a dose-response effect for tolerability and,<br />

possibly, efficacy with the new TAB between 200 and 400 mg BID. Further<br />

studies will require dosing according to patient tolerability within this dose<br />

range.<br />

DE1 DE2<br />

200 TAB 400 CAP 300 TAB 400 TAB 400 CAP<br />

(n�13) (n�11) (n�18) (n�16) (n�18)<br />

G3/4 AEs, n (%)<br />

Nausea 0 0 0 2 (13%) 0<br />

Fatigue 0 0 3 (17%) 1 (6%) 0<br />

Diarrhea 0 0 2 (11%) 0 0<br />

Anemia (Hb changes)<br />

Other, n (%)<br />

0 0 4 (22%) 4 (25%) 1 (6%)<br />

Blood transfusions<br />

Dose modifications, n (%)<br />

0 0 5 (28%) 7 (44%) 2 (11%)<br />

Dose reductions 2 (15%) 1 (9%) 4 (22%) 10 (63%) 3 (17%)<br />

Drug discontinuations due to AEs<br />

Change in tumor size, 8 weeks<br />

0 0 0 0 0<br />

LS, mean (%) 4.5 -12.8 -6.8 -28.6 -27.8<br />

Difference 17.3 21.0 -0.8<br />

95% CI -11.8, 46.3 1.3, 40.7 -20.8, 19.3<br />

3050 General Poster Session (Board #13H), Mon, 8:00 AM-12:00 PM<br />

Developing rational drug combination strategies for PARP inhibitors.<br />

Presenting Author: Farah Rehman, The Institute <strong>of</strong> Cancer Research,<br />

London, United Kingdom<br />

Background: The recent clinical development <strong>of</strong> Poly (ADP-ribose) polymerase<br />

(PARP) inhibitors has provided formal pro<strong>of</strong>-<strong>of</strong>-concept that synthetic<br />

lethal approaches can be used to treat cancer. The promise <strong>of</strong> PARP<br />

inhibitors in BRCA mutant tumours is apparent but questions regarding<br />

their wider clinical use remain. To date, a number <strong>of</strong> clinical trials have<br />

been designed using PARP inhibitors as single agents as well as in<br />

combination with current standards <strong>of</strong> care. However, there is a paucity <strong>of</strong><br />

pre-clinical information about PARP inhibitor/drug combinations and<br />

empirical clinical trials <strong>of</strong> standard <strong>of</strong> care plus PARP inhibitors may not<br />

represent the most effective approach to dissect this. We aim to identify<br />

effective drug combination strategies that could be used clinically in<br />

selected patient groups. Methods: Using a combination <strong>of</strong> high and medium<br />

throughput techniques and established methodologies for determining<br />

synergy, additivity and antagonism, we have characterised the effects <strong>of</strong><br />

combining PARP inhibitors with a diverse panel <strong>of</strong> over 3,000 compounds<br />

in a range <strong>of</strong> tumour cell models including isogenic matched cell line pairs<br />

to determine the impact <strong>of</strong> genetic background on drug combination<br />

efficacy. We have integrated this data with functional pr<strong>of</strong>iling data to<br />

identify genetic backgrounds where these combinations may be most<br />

effective. Results: Results to date suggest promising combinations <strong>of</strong> PARP<br />

inhibitors with temozolomide, CHK1 inhibitors, ATM inhibitors and other<br />

agents. Some <strong>of</strong> these combinations show specificity for genetic backgrounds<br />

such as PTEN mutations. Conclusions: Screening <strong>of</strong> drug libraries<br />

has identified promising PARP inhibitor drug combinations and demonstrates<br />

how some drug combinations show specificity in particular genetic<br />

backgrounds. This raises the possibility <strong>of</strong> widening the therapeutic index<br />

in patients with these genetic backgrounds and treating a wider group <strong>of</strong><br />

patients with PARP inhibitors.<br />

Developmental Therapeutics—Experimental Therapeutics<br />

185s<br />

3049 General Poster Session (Board #13G), Mon, 8:00 AM-12:00 PM<br />

A phase I study <strong>of</strong> veliparib (ABT-888) in combination with carboplatin and<br />

paclitaxel in advanced solid malignancies. Presenting Author: Leonard<br />

Joseph Appleman, University <strong>of</strong> Pittsburgh Cancer Institute, Pittsburgh, PA<br />

Background: Veliparib (ABT-888, NSC 737664) is an orally available<br />

inhibitor <strong>of</strong> poly(ADP-ribose) polymerase (PARP)-1 and -2: enzymes that<br />

recruit base excision repair machinery to single-stranded DNA breaks.<br />

Expression <strong>of</strong> PARP-1 may be increased in cancer cells and confer<br />

resistance to DNA-damaging agents. The objectives <strong>of</strong> this phase I study<br />

included determination <strong>of</strong> the recommended phase 2 dose (RP2D),<br />

maximum tolerated dose (MTD), dose limiting toxicity (DLT) and pharmacokinetics<br />

(PK) <strong>of</strong> veliparib in combination with paclitaxel (P) and carboplatin<br />

(C). Methods: Eligibility criteria included advanced solid tumors, � 3 prior<br />

chemotherapy regimens for advanced disease, ECOG performance status<br />

0-2. Veliparib was given PO BID on days 1-7 <strong>of</strong> each 21 day cycle, and P<br />

and C were administered on day 3. In dose levels 1-7, veliparib was not<br />

given during cycle 1 to serve as intra-patient control for toxicity and PK<br />

assessment, and DLT was evaluated during cycle 2. A standard “3�3”<br />

dose escalation was utilized starting at veliparib 20 mg BID, P 150 mg/m2,<br />

C AUC 5. Plasma concentrations <strong>of</strong> veliparib, P and C were determined by<br />

LC-MS/MS and AAS during cycle 1 and 2. Results: To date, 68 patients<br />

have been enrolled. Tumor types included lung (15), breast (14), melanoma<br />

(10), squamous cell <strong>of</strong> head/neck (7), and urothelial (5). Toxicities<br />

observed were expected with C plus P chemotherapy, including neutropenia,<br />

thrombocytopenia, peripheral neuropathy. DLTs were seen in 2 out <strong>of</strong> 7<br />

evaluable patients at the maximum administered dose: veliparib 120 mg<br />

BID, P 200 mg/m2 , C AUC 6, (febrile neutropenia, hyponatremia).<br />

Veliparib 80 mg, P 200 mg/m2 , C AUC 6 was well tolerated with 1 out <strong>of</strong> 9<br />

DLT (febrile neutropenia). Median number <strong>of</strong> cycles was 5 (1-17). <strong>Part</strong>ial<br />

response was seen in 11 (Lung-2, breast-2, melanoma-2, urothelial-2,<br />

head and neck, gastric, unknown primary) and complete response in 1<br />

patient with breast cancer and 1 patient with urothelial cancer. Stable<br />

disease was observed in 35 patients. Veliparib did not affect the PK<br />

disposition <strong>of</strong> P or C. Conclusions: Veliparib in combination with P and C<br />

was well-tolerated with a safety pr<strong>of</strong>ile similar to P and C alone. Promising<br />

anti-tumor activity was observed in several tumor types.<br />

3051 General Poster Session (Board #14A), Mon, 8:00 AM-12:00 PM<br />

Phase I study to determine the bioavailability and tolerability <strong>of</strong> a tablet<br />

formulation <strong>of</strong> the PARP inhibitor olaparib in patients with advanced solid<br />

tumors: Dose-escalation phase. Presenting Author: Avinash Gupta, Oxford<br />

University Hospitals NHS Trust, Oxford, United Kingdom<br />

Background: We previously reported the comparative bioavailability <strong>of</strong> the<br />

olaparib tablet (TAB) up to 200 mg BID, with the initial capsule formulation;<br />

gmean AUC0–Tfollowing 200 mg BID TAB was ~20% lower than 400<br />

mg BID capsule (CAP; Molife et al ASCO 2010). Olaparib 200 mg BID TAB<br />

had an acceptable tolerability pr<strong>of</strong>ile suggesting further dose escalation<br />

might be justified. Methods: Study NCT00777582 was amended to include<br />

a dose-escalation phase, to define the maximum tolerated dose (MTD) and<br />

safety pr<strong>of</strong>ile <strong>of</strong> the TAB, and 2 expansion phases to compare safety and<br />

efficacy <strong>of</strong> TAB doses vs 400 mg BID CAP. Expansion phase data are<br />

reported separately. Here, we report preliminary data from the doseescalation<br />

phase where patients (pts) with advanced solid tumors, ECOG<br />

PS 0–2 and adequate organ function were assigned to treatment with<br />

increasing olaparib BID TAB doses in 28-day continuous dosing cycles.<br />

Pharmacokinetic (PK) sampling was performed on days 1, 8, 15, 29, 57.<br />

Results: 30 pts (M:F, 3:27; median age 54 yrs [range 19–70]) were<br />

enrolled in the dose-escalation phase and received treatment at 5 dose<br />

levels: 250, 300, 350, 400 and 450 mg (each dose level, n�6). Overall,<br />

the most common AEs were nausea (80%), fatigue (73%) and diarrhea<br />

(36%). The majority <strong>of</strong> AEs were mild to moderate (CTC grade [G] 1/2).<br />

Hematologic toxicity in terms <strong>of</strong>, mostly mild, anemia, neutropenia and<br />

thrombocytopenia, appeared to increase at doses <strong>of</strong> 300 mg BID or higher.<br />

Two pts had dose-limiting toxicities at 450 mg (G3 thrombocytopenia and<br />

G3 anemia); the TAB MTD was 400 mg BID. Exposure increased proportionally<br />

with increasing dose: mean exposure after 400 mg BID was double that<br />

following the previously reported 200 mg BID dose. Following 300 and 400<br />

mg doses, gmean Cmax ss, AUC0–Tand Cmin ss matched or exceeded the 400<br />

mg BID CAP dose. Conclusions: The MTD <strong>of</strong> olaparib TAB was 400 mg BID.<br />

Based on PK analysis, the 300 and 400 mg BID doses were selected for<br />

evaluation in the MTD expansion phase to further define safety and<br />

preliminary efficacy.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


186s Developmental Therapeutics—Experimental Therapeutics<br />

3052 General Poster Session (Board #14B), Mon, 8:00 AM-12:00 PM<br />

Phase I dose-escalation study to evaluate the safety, pharmacokinetics,<br />

and pharmacodynamics <strong>of</strong> CEP-9722 (a PARP1-2 inhibitor) as singleagent<br />

and in combination with temozolomide in patients with advanced<br />

solid tumors (NCT00920595). Presenting Author: Mario Campone, Institut<br />

de Cancérologie de l’Ouest - René Gauducheau, Centre de Recherche en<br />

Cancérologie, Saint Herblain-Nantes, France<br />

Background: Poly-ADP-ribose (PAR) polymerases (PARPs) are essential in<br />

cellular processing <strong>of</strong> DNA damage via the base excision repair pathway.<br />

CEP-9722, an orally available PARP1-2 inhibitor, demonstrated singleagent<br />

antitumor activity and synergy with temozolomide (TMZ) in xenograft<br />

models. This phase I study evaluated the pharmacokinetics, pharmacodynamics,<br />

and the maximum tolerated dose (MTD) <strong>of</strong> CEP-9722 in monotherapy<br />

and combination with TMZ. Methods: Adult patients with advanced<br />

solid tumors were enrolled in cohorts <strong>of</strong> 3-6 patients to receive a 14-day<br />

cycle <strong>of</strong> CEP-9722 (days 1-5), followed by 28-day cycles <strong>of</strong> CEP-9722<br />

(days 1-5) plus TMZ (150 mg/m2 days 1-5). The dose <strong>of</strong> CEP-9722 was<br />

150 mg/day in the first cohort and was escalated depending on the<br />

occurrence <strong>of</strong> dose-limiting toxicities (DLTs) during cycles 1 and 2. The<br />

safety, pharmacokinetics, and pharmacodynamics (PAR levels in peripheral<br />

blood mononuclear cells) <strong>of</strong> CEP-9722 were analyzed. Results: Overall,<br />

26 patients (18F,8 M) 18 to 71 years <strong>of</strong> age were enrolled in 5 cohorts <strong>of</strong><br />

3-9 patients and treated with CEP-9722 at 150-1000 mg/day. The MTD <strong>of</strong><br />

CEP-9722 in combination with TMZ (150 mg/m2 ) was reached at 1000<br />

mg/day. The recommended dose was 750 mg/day. A total <strong>of</strong> 9 patients were<br />

treated at the recommended dose. DLTs were observed in 2 patients during<br />

cycle 1 (1 grade 3 myositis at 750 mg/day and 1 grade 3 asthenia at 1000<br />

mg/day) and 2 patients during cycle 2 (1 grade 3 asthenia at 300 mg/day<br />

and 1 persistent grade 2 weight loss at 1000 mg/day). Overall during this<br />

study, 4 grade 3 treatment-related adverse events were observed. The<br />

pharmacokinetics showed high intra- and inter-patient variability at all<br />

doses. The pharmacodynamic analysis demonstrated PARP inhibition at all<br />

doses, but the high inter- and intra-patient variability prevented any<br />

conclusion regarding a dose / PARP inhibition relationship. Conclusions:<br />

TheMTD <strong>of</strong> CEP-9722 when administered with TMZ was 1000 mg days 1-5<br />

and the combination CEP-9722/TMZ was well tolerated. In addition, a<br />

clear signal <strong>of</strong> PARP inhibition was demonstrated.<br />

3054 General Poster Session (Board #14D), Mon, 8:00 AM-12:00 PM<br />

A phase I study <strong>of</strong> chronically dosed, single-agent veliparib (ABT-888) in<br />

patients (pts) with either BRCA 1/2-mutated cancer (BRCA�), platinumrefractory<br />

ovarian cancer, or basal-like breast cancer (BRCA-wt). Presenting<br />

Author: Shannon Leigh Huggins-Puhalla, University <strong>of</strong> Pittsburgh<br />

Cancer Institute, Magee-Womens Hospital, Pittsburgh, PA<br />

Background: Veliparib (ABT-888) is an oral, potent inhibitor <strong>of</strong> PARP 1/2.<br />

Preclinically, PARP inhibitors have activity in tumors with defective<br />

homologous recombination (HR), particularly those that are BRCA�.<br />

Reduced levels <strong>of</strong> BRCA expression have been observed in ovarian cancer<br />

and basal-like breast cancer, which share genotypic and phenotypic<br />

similarities with BRCA� cancers. We postulated that these tumors types<br />

may be similarly sensitive to single-agent PARP inhibition. This study<br />

sought to establish the maximum tolerated dose (MTD), dose limiting<br />

toxicities (DLT), pharmacokinetic and pharmocodynamic properties, and<br />

preliminary efficacy <strong>of</strong> chronically-dosed veliparib. Methods: A3�3 dose<br />

escalation phase I trial was performed. Nine dose levels (DL) were planned,<br />

and dose escalation started at 50 mg BID to a maximum <strong>of</strong> 500 mg BID.<br />

Veliparib was administered orally continuously on a 28 day cycle. Results:<br />

63 pts have been enrolled to date. Thirty-eight were BRCA� (20 ovary, 12<br />

breast, 2 pancreas, and one each - prostate, peritoneal, fallopian tube,<br />

endometrial); 25 BRCA-wt. (21 breast, 4 ovarian). DLTs occurred at the<br />

following dose levels: BRCA�: gr. 2 thrombocytopenia at 50 mg BID;<br />

BRCA�: gr.3 Nausea/vomiting at 400 mg BID; BRCA-wt: gr 2 seizure at<br />

400 mg BID. The MTD has not been determined and 500 mg BID is<br />

presently enrolling. Notable toxicities have included low-grade fatigue and<br />

nausea. PK was linear and non-saturable with t½<strong>of</strong>5h.Thenumber <strong>of</strong><br />

cycles administered ranged from 1- 15, median 2. In BRCA� pts, there<br />

were 2 partial responses (breast, ovarian) and 10 pts had evidence <strong>of</strong><br />

prolonged SD � 4 months. In BRCA-wt pts, there was 1 PR (breast) and 7<br />

pts with SD� 4 months. Correlative studies, including assessment <strong>of</strong> PAR<br />

inhibition and BRCA methylation status, are ongoing. Conclusions: Veliparib<br />

is tolerable on a continuous oral dosing schedule with evidence <strong>of</strong><br />

anti-tumor activity seen in BRCA� and BRCA-wt tumors. A mandatory<br />

biopsy expansion cohort is planned at the recommended phase II dose,<br />

which will allow further insights regarding efficacy and mechanisms <strong>of</strong><br />

resistance to PARP inhibition.<br />

3053 General Poster Session (Board #14C), Mon, 8:00 AM-12:00 PM<br />

Phase I study <strong>of</strong> sequential sapacitabine and seliciclib in patients with<br />

advanced solid tumors. Presenting Author: Ge<strong>of</strong>frey Shapiro, Dana-Farber<br />

Cancer Institute, Boston, MA<br />

Background: Sapacitabine is an orally administered nucleoside analogue;<br />

the active metabolite CNDAC generates ss DNA breaks that are converted to<br />

ds DNA breaks (DSB) during subsequent replication, resulting in cell death<br />

if unrepaired. CNDAC-induced DSB repair is dependent on homologous<br />

recombination (HR). Seliciclib is a potent orally bioavailable inhibitor <strong>of</strong><br />

CDK2, 7 and 9, and sensitizes cells to CNDAC by decreasing DSB repair via<br />

compromise <strong>of</strong> HR protein activation and transcriptional inhibition <strong>of</strong> HR<br />

components. This phase I study evaluates sequential sapacitabine and<br />

seliciclib. Methods: Dose escalation was conducted in patients with<br />

incurable solid tumors and adequate organ function with sapacitabine<br />

b.i.d. x 7 consecutive days (d1-7), seliciclib b.i.d. x 3 consecutive days<br />

(d8-10) followed by 11 days <strong>of</strong> rest. At least 3 patients were evaluated per<br />

dose level. MTD was the highest dose level at which less than one-third <strong>of</strong> at<br />

least 6 patients experienced cycle 1 DLT. Skin biopsies were obtained to<br />

assess DNA damage following sapacitabine (d8 vs pre-treatment) and<br />

further augmentation <strong>of</strong> DNA damage after seliciclib (d11 vs d8). Results:<br />

27 patients were treated. The MTD and RP2D is sapacitabine 50 mg<br />

b.i.d./seliciclib 1200 mg b.i.d. DLTs were reversible transaminase elevations<br />

and neutropenia. The most frequent adverse events (all cycles,<br />

regardless <strong>of</strong> causality) included anorexia, fatigue, abdominal pain, dizziness,<br />

nausea, anemia, neutropenia, creatinine elevation, hyperglycemia,<br />

hyperbilirubinemia, hypophosphatemia, hypokalemia and hypomagnesemia,<br />

the majority mild to moderate in intensity. Skin biopsies showed a<br />

2.3-fold increase in H2AX staining post-sapacitabine (n�16; p�0.007)<br />

and a further 0.58-fold increase post-seliciclib (n�12; p�0.069). Two<br />

confirmed PRs occurred in patients with pancreatic and breast cancer, both<br />

BRCA mutation carriers. SD as best response �/� 12 weeks was observed<br />

in 6 additional patients, including one BRCA mutation carrier with ovarian<br />

cancer (ongoing at 24 weeks). Conclusions: Sequential sapacitabine and<br />

seliciclib is safe with preliminary antitumor activity. BRCA mutation carrier<br />

status may be a potential biomarker for response across multiple tumor<br />

types.<br />

3055 General Poster Session (Board #14E), Mon, 8:00 AM-12:00 PM<br />

Using proton MRSI to predict response to vorinostat treatment in recurrent<br />

GBM. Presenting Author: Hyunsuk Shim, Emory University, Atlanta, GA<br />

Background: A major impediment to the development <strong>of</strong> new therapies for<br />

glioblastoma (GBM) is a lack <strong>of</strong> biomarkers indicating response. Epigenetic<br />

modifications are now recognized as a frequent occurrence in the early<br />

phases <strong>of</strong> tumorigenesis, playing a central role in tumor development.<br />

Epigenetic alterations differ significantly from genetic modifications in that<br />

they may be reversed by ‘‘epigenetic drugs’’ such as histone deacetylase<br />

inhibitors (HDACis). As a promising new modality for cancer therapy, the<br />

first generation <strong>of</strong> HDACi is currently being tested in phase I/II clinical<br />

trials. Methods: GBM alterations from therapy with HDACis, such as<br />

vorinostat (SAHA), include tumor redifferentiation/cytostasis rather than<br />

tumor size reduction limits the utility <strong>of</strong> traditional imaging methods such<br />

as MRI. Magnetic resonance spectroscopic imaging (MRSI) quantitates<br />

various metabolite levels in tumor and normal brain, allowing characterization<br />

<strong>of</strong> metabolic processes in live tissue. Results: In our preclinical model,<br />

MRS detected metabolic response to SAHA after only 3 days <strong>of</strong> treatment:<br />

reduced alanine and lactate and elevated myo-inositol, N-acetyl aspartate<br />

and creatine; each returning toward normal brain levels. This led to our<br />

clinical study <strong>of</strong> MRSI to evaluate the metabolic response <strong>of</strong> recurrent<br />

GBMs to SAHA � temozolomide. After only 7 days <strong>of</strong> SAHA treatment,<br />

MRSI can distinguish metabolic responders (normalization/restoration <strong>of</strong><br />

tumor metabolites towards normal brain-like metabolism) from nonresponders<br />

(no significant change in tumor metabolites). Our initial cohort<br />

(n�6) consists <strong>of</strong> 3 responders and 3 non-responders with highly significant<br />

differences in their change in metabolite levels (p � 0.001).<br />

Conclusions: Our results provide exciting insights into the mechanisms by<br />

which HDACi exerts its effect on GBMs. Tumor cells have increased<br />

biosynthetic needs requiring reprogramming <strong>of</strong> cellular metabolism. This<br />

creates increased energy demands, making tumor cells even more vulnerable<br />

to interventions targeting their metabolism. HDACi may induce<br />

redifferentiation in tumors by targeting tumor metabolism. Thus, MRSI<br />

provides a novel modality to predict response to HDACi-containing combination<br />

therapy in GBM.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


3056 General Poster Session (Board #14F), Mon, 8:00 AM-12:00 PM<br />

A phase I/II study to evaluate the ability <strong>of</strong> decitabine and panobinostat to<br />

improve temozolomide chemosensitivity in metastatic melanoma. Presenting<br />

Author: Chang Xia, Hematology, Oncology, and Blood & Marrow<br />

Transplantation, UIHC, Iowa City, IA<br />

Background: Epigenetic gene regulation is likely a contributing mechanism<br />

<strong>of</strong> cancer initiation and progression. Emerging evidence indicates that<br />

epigenetics may also play a key role in the development <strong>of</strong> chemoresistance<br />

in melanoma. We proposed combining the histone deacetylase inhibitor<br />

panobinostat (PT) and the demethylator decitabine (D) to overcome the<br />

development <strong>of</strong> epigenetic mediated temozolomide (TMZ) resistance in<br />

metastatic melanoma. Methods: Eligible patients must be �18, stage IV<br />

melanoma, and naïve or previously treated, with good performance status<br />

(ECOG � 2) and normal organ functions. Patients with previous exposure to<br />

TMZ were allowed on study. The study includes the dose escalation <strong>of</strong> D<br />

and PT followed by expansion cohorts. D (0.1mg/kg SQ, 0.2mg/kg SQ) on<br />

days 1, 3, 5, 8, 10,12, PT (10mg PO, 20mg PO, 30mg PO) Q96h starting<br />

day 8, and TMZ 150mg/m2 PO daily on days 9-13 <strong>of</strong> each 42 day cycle.<br />

TMZ was increased to 200mg/m2 in the absence <strong>of</strong> grade 2 thrombocytopenia.<br />

Primary endpoints <strong>of</strong> phase I study are to determine the toxicity, safety<br />

and maximum tolerated dose (MTD) <strong>of</strong> the D, PT and TMZ combination.<br />

Results: To date, the phase I portion <strong>of</strong> this trial is completed. We report on<br />

the safety data for this combination. 17 patients received treatment (1st cohort: 5; 2nd cohort: 4; 3rd cohort: 4; 4th cohort: 4 ). M:F 11:6. Median<br />

age: 56 (32-77); Median ECOG PS: 1; 82% <strong>of</strong> the patients received at least<br />

one cycle (n�14). Median number <strong>of</strong> cycles given: 2 (0-6). To date, no<br />

DLTs have occurred. The MTD was not reached. The only grade (G) 4<br />

adverse event (AE) is neutropenia on a patient in cohort 3 and it resolved<br />

within 3 days. G3 AEs included lymphopenia (n�4, 23%), anemia (n�2,<br />

12%), leukopenia (n�2, 12%) and fatigue (n�2, 12%). Common G2<br />

toxicities were leukopenia (n�5, 30%), neutropenia (n�4, 23%), nausea<br />

(n�4, 23%) and lymphopenia (n�3, 18%). Conclusions: The combination<br />

<strong>of</strong> D, PT, and TMZ appears to be safe and well-tolerated. The recommended<br />

dose for the phase II study remains to be determined.<br />

3058 General Poster Session (Board #14H), Mon, 8:00 AM-12:00 PM<br />

A phase I trial <strong>of</strong> the histone deacetylase inhibitor panobinostat (LBH589)<br />

and epirubicin in patients with solid tumor malignancies. Presenting<br />

Author: Amy Patricia Moore, UCSF, San Francisco, CA<br />

Background: Preclinical and clinical data suggest pre-exposure <strong>of</strong> cancer<br />

cells to a histone deacetylase inhibitor (HDACi) potentiates topoisomerase<br />

inhibitors. HDACi-induced histone acetylation and chromatin modulation<br />

facilitates DNA access and target recruitment for topo II inhibitors. In vitro<br />

data further suggest effective inhibition <strong>of</strong> HDAC2 is necessary for<br />

enhanced epirubicin-induced apoptosis. Methods: This phase I trial explores<br />

the safety, tolerability, and maximum tolerated dose (MTD) <strong>of</strong><br />

escalating doses <strong>of</strong> panobinostat given orally on days 1, 3, and 5 followed<br />

by epirubicin administered intravenously at 75 mg/m2 on day 5 in 21-day<br />

cycles. Histone acetylation and HDAC2 expression are evaluated in preand<br />

post-treatment peripheral blood mononuclear cells (PBMCs) in all<br />

patients and in tumor cells <strong>of</strong> 16 patients treated at the MTD. Results: 36<br />

patients have enrolled [10M/26F, median age 47 years (22-80)] in 5<br />

panobinostat cohorts: 20, 30, 40, 50, 60 mg. Tumor types include<br />

melanoma (n�6), breast (n�6), sarcoma (n�16), ovarian (n�2), lung<br />

(n�2), and one each <strong>of</strong> neuroblastoma, pancreatic, testicular, and colon<br />

cancer. Prior to enrollment, patients received a median <strong>of</strong> 3 (0-8) prior<br />

chemotherapy regimens and 40% had anthracyclines. Dose-limiting toxicities<br />

(DLTs) included 1/3 grade 3 fatigue and 1/3 grade 4 thrombocytopenia<br />

at 60 mg <strong>of</strong> panobinostat, 1/6 patient experienced grade 3 atrial fibrillation<br />

at 50 mg, defining 50 mg panobinostat as the MTD. Non-dose–limiting<br />

grade 3/4 hematological toxicities include neutropenia (n�19, 53%),<br />

febrile neutropenia (n�6, 17%), thrombocytopenia (n�6, 17%), and<br />

anemia (n�4, 11%). Of 34 evaluable patients, 5 had partial responses and<br />

14 had stable disease in anthracycline-refractory sarcomas (4) and<br />

Her2neu positive breast cancer (2), and small cell lung cancer. Correlative<br />

studies demonstrate increased H4 acetylation in PBMCs on day 3 and 5<br />

suggesting sufficient histone deacetylase inhibition. Conclusions: Sequencespecific<br />

combination <strong>of</strong> panobinostat and epirubicin shows early activity<br />

without potentiating epirubicin toxicity. Dose expansion in anthracyclinepretreated<br />

sarcoma patients is ongoing.<br />

Developmental Therapeutics—Experimental Therapeutics<br />

187s<br />

3057 General Poster Session (Board #14G), Mon, 8:00 AM-12:00 PM<br />

Testing targeted demethylation to overcome resistance to epidermal growth<br />

factor receptor (EGFR) blocking agents in wild-type (wt) KRAS metastatic<br />

colorectal cancer patients (mCRC). Presenting Author: Ignacio Garrido-<br />

Laguna, Division <strong>of</strong> Oncology and Center for Investigational Therapeutics at<br />

Huntsman Cancer Institute at the University <strong>of</strong> Utah, Salt Lake City, UT<br />

Background: Panitumumab, a monoclonal antibody against EGFR, led to<br />

improved progression-free survival (PFS) in patients with (wt)KRAS mCRC.<br />

However, the benefit <strong>of</strong> panitumumab is limited to a not yet identified small<br />

subset <strong>of</strong> (wt)KRAS patients. The CpG island methylator phenotype<br />

(CIMP-high) is present in 15-20% <strong>of</strong> CRC patients. Epigenetic silencing<br />

through methylation <strong>of</strong> PTEN and/or genes in the EGFR pathway might play<br />

a role in resistance to therapy. We assessed whether decitabine (a<br />

hypomethylating agent) reverted resistance to anti-EGFR therapies. Methods:<br />

Patients (n�19) with (wt)KRAS mCRC previously treated with anti-EGFR<br />

therapies were enrolled. Patients were treated with decitabine 45mg/kg IV<br />

on day (d)1 and d15 and panitumumab 6mg/kg IV on d8 and d22 q28days.<br />

Peripheral blood, skin biopsies and buccal smear samples were collected<br />

on d1, d8, d15 and d22 to assess methylation changes in PTEN, RASSF1A<br />

and SOCS-1 in response to therapy. Tumors were analyzed for PIK3CA<br />

mutations using PCR-based DNA sequencing <strong>of</strong> exon 9 and 20 by PCR as<br />

well as for PTEN by immunohistochemistry. Results: Median age was 53<br />

(range 31-72). Eleven (58%) patients were female; 15 (79%) had ECOG<br />

PS 1; 3 (16%) had ECOG PS 2; and 1 had ECOG PS 0. Median number <strong>of</strong><br />

previous therapies was 4. The most common toxicities were rash (68%),<br />

hypomagnesemia (26%) and neutropenia (10%) (all grade 1-2). The most<br />

common grade 3-4 toxicities were neutropenic fever (5%) and hypomagnesemia<br />

(5%). Of 19 (wt)KRAS mCRC patients previously treated with<br />

anti-EGFR therapies, 2 (11%) had a partial response (PR) (-50% and<br />

-30%, respectively) and 3 (16%) had stable disease (SD) �4 months (7.8,<br />

5.9 and 4.2 months). Both responders were (wt)PIK3CA and (wt)PTEN.<br />

<strong>Clinical</strong> benefit rate (PR�SD) was 27%. Median PFS was 60 days (95% CI<br />

45.4-74.5), similar to median PFS after last anti-EGFR therapy, 61d (95%<br />

CI 50.6-71.3). Conclusions: Panitumumab � decitabine is an active<br />

combination in a subset <strong>of</strong> patients with mCRC previously treated with<br />

anti-EGFR therapies (2 PRs in this population). Efficacy assessment in an<br />

anti-EGFR therapy naïve population is warranted. Methylation studies are<br />

ongoing.<br />

3059 General Poster Session (Board #15A), Mon, 8:00 AM-12:00 PM<br />

Phase Ib study <strong>of</strong> CNTO 888 (anti-CCL 2) in combination with chemotherapies<br />

for treatment <strong>of</strong> patients with solid tumors. Presenting Author: Antonio<br />

Calles, START-Madrid, Centro Integral Oncológico Clara Campal, Madrid,<br />

Spain<br />

Background: CC-chemokine ligand 2 (CCL2) is expressed in various malignancies,<br />

implicated in angiogenesis, proliferation and metastasis. CNTO888<br />

is a hIgG1� mAb with selective high CCL2 binding affinity and showed<br />

preclinical antitumor activity. Methods: Primary objectives were safety and<br />

a recommended dose <strong>of</strong> CNTO 888 in combination with chemotherapy:<br />

Arm1-15mg/kg CNTO 888 � docetaxel 75 mg/m2 Q3W; Arm 2-15<br />

mg/kg CNTO 888 � gemcitabine 1000 mg/m2 on D1 and 8 Q3W; Arm 3 -<br />

15 mg/kg CNTO 888 � paclitaxel 175 mg/m2 and carboplatin AUC6 Q3W;<br />

Arm4-10mg/kg CNTO 888 Q2W � pegylated doxorubicin 50 mg/m2 Q4W. Two (Arms 1, 2, 3) or 4 (Arm 4) prior chemotherapies for advanced<br />

disease were allowed. Pharmacokinetic (PK) pr<strong>of</strong>ile <strong>of</strong> CNTO 888 and<br />

chemotherapies, free and total CCL2, CTC/CEC count, and uNTX were<br />

evaluated. Adverse events (AE) and dose limiting toxicities (DLT) were<br />

evaluated after 6 subjects completed 1 cycle. Results: 53 subjects<br />

(22F/31M) were treated: Arm 1 n�15, Arm 2 n�12, Arm 3 n�12, Arm 4<br />

n�14. Two DLTs occurred: a grade 4 febrile neutropenia (Arm 1) and a<br />

grade 3 neutropenia (Arm 2). All 4 combinations were found to be safe and<br />

tolerable and continued enrolment to a minimum <strong>of</strong> 12 subjects evaluable<br />

for safety and PK. Best overall response were partial response (PR) (1) and<br />

stable disease (18; 2 � 4 months). One subject with pancreatic adenocarcinoma<br />

achieved PR, allowing for surgical resection. Most frequently<br />

reported (� 20%) related serious AEs are: Arm 1: Neutropenia, Stomatitis,<br />

Fatigue, Febrile neutropenia, Alopecia. Arm 2: Anemia. Arm 3: Thrombocytopenia,<br />

Alopecia, Neutropenia, Asthenia, Fatigue, Anemia, Arthralgia,<br />

Nausea, Vomiting. Arm 4: Stomatitis, Rash, Fatigue, Nausea, Anemia,<br />

Neutropenia. CNTO 888 did not affect the chemotherapy PK pr<strong>of</strong>ile;<br />

similarly, chemotherapy did not affect the CNTO 888 PK pr<strong>of</strong>ile. No<br />

subjects (n�38) tested positive for antibodies to CNTO 888. Free CCL2<br />

declined 2 hrs post treatment and returned to baseline levels by 48 hrs and<br />

continued to increase with further administrations in all treatment arms.<br />

There were no consistent changes with other biomarkers. Conclusions:<br />

CNTO 888 in combination with standard chemotherapy was well tolerated<br />

but few objective responses were seen.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


188s Developmental Therapeutics—Experimental Therapeutics<br />

3060 General Poster Session (Board #15B), Mon, 8:00 AM-12:00 PM<br />

A phase I study <strong>of</strong> EP-100, a luteinizing hormone releasing hormone<br />

(LHRH) ligand conjugated to a synthetic cytolytic peptide in patients with<br />

advanced refractory LHRH- receptor (R)-expressing tumors. Presenting<br />

Author: Ramesh K. Ramanathan, Virginia G. Piper Cancer Center at<br />

Scottsdale Healthcare/TGen, Scottsdale, AZ<br />

Background: EP-100 is a synthetic 28 amino acid LHRH peptide conjugated<br />

to an 18 aa cytolytic peptide. Selective targeting in LHRH-R<br />

expressing cells occurs via direct binding to the extracellular membrane<br />

receptor followed by disruption <strong>of</strong> the membrane by the cytolytic peptide<br />

portion. A first in human, phase I study was performed. Methods: Eligible<br />

pts had adequate organ function and LHRH-R tumor expression, as<br />

determined by IHC. EP-100 was given IV (30-60 min) weekly x3for<br />

cohorts 1- 6 (n�20 at 0.6 - 5.2 mg/m2 ) and then twice weekly x3for<br />

cohorts 7- 11 (n�18 at 7.8 - 40 mg/m2 ) with a week break. The<br />

pharmacokinetic pr<strong>of</strong>ile <strong>of</strong> EP-100 and antibody production against EP-<br />

100 was determined by a validated HPLC/MS and ELISA respectively.<br />

Potential activity <strong>of</strong> EP-100 on the pituitary gonadotropes was determined<br />

by plasma LH and FSH. Results: We screened 97 pts, archival tumor tissue<br />

obtained from 81 pts; 53 (65%) were positive for LHRH-R, <strong>of</strong> which 37<br />

were enrolled (female 76%) and treated. The LHRH-R expression rate in<br />

breast was 81% (N�21) and in ovarian cancer 56% (n�18). Most pts had<br />

breast (n� 16, 42%) ovary (n�7,18%), colon (n�4, 11%) or uterine<br />

(n�3, 8%) cancer. Pts were treated in a 3 � 3 standard dose escalation<br />

scheme. Doses were escalated 100% in the absence <strong>of</strong> a grade 2 drug<br />

related toxicity. MTD was not reached at the highest dose level <strong>of</strong> 40<br />

mg/m2 . Only one DLT occurred, a grade 2 increase in ALT/AST. The only<br />

other drug-related toxicity was a self limited infusion-related skin reaction<br />

(grade 1-2) in 10 pts. EP-100 was rapidly cleared from circulation, mean<br />

half life ranged from 7.1 � 3.8 min to 15.9 � 3.6 min. Best response was<br />

stable disease for �12 weeks in 5 pts. In 3 pts rapid, sustained decreases<br />

in LH/FSH levels were noted. Antibody production against EP-100 was<br />

absent in all pts. Conclusions: The study has completed accrual, the phase<br />

II dose is 30-40 mg/m2 twice a week x 3 weeks.EP-100is safe, well<br />

tolerated and not antigenic/immunogenic. Preclinical studies indicated<br />

synergy with paclitaxel and doxorubicin. A phase II study <strong>of</strong> EP-100 plus<br />

paclitaxel in ovarian cancer is planned.<br />

3062 General Poster Session (Board #15D), Mon, 8:00 AM-12:00 PM<br />

Open-label extension study <strong>of</strong> the RNAi therapeutic ALN-VSP02 in cancer<br />

patients responding to therapy. Presenting Author: Maria Alsina, Molecular<br />

Therapeutics Research Unit, Vall d’Hebron University Hospital, Barcelona,<br />

Spain<br />

Background: ALN-VSP02 is an RNA interference (RNAi) therapeutic comprised<br />

<strong>of</strong> lipid nanoparticle-formulated small interfering RNAs targeting<br />

vascular endothelial growth factor (VEGF)-A and kinesin spindle protein<br />

(KSP). In a phase 1 trial, ALN-VSP02 administered as an iv infusion q2 wks<br />

was well-tolerated and showed evidence <strong>of</strong> anti-VEGF pharmacology and<br />

antitumor activity. Methods: Patients treated on the phase I trial with stable<br />

disease (SD) or better after 4 months (8 doses) were eligible to continue on<br />

an extension study until disease progression. Main objectives included<br />

continued evaluation <strong>of</strong> safety/tolerability and assessment <strong>of</strong> disease<br />

response. Results: Seven <strong>of</strong> 37 patients (18.9%) evaluable for response<br />

went onto the extension study, including 1 <strong>of</strong> 7 (14.2%) at 0.4 mg/kg, 2 <strong>of</strong><br />

5 (40%) at 0.7 mg/kg, and 4 <strong>of</strong> 11 (36.3%) at 1.0 mg/kg. All had<br />

progressed after one or more prior therapies. Tumor types included head<br />

and neck squamous cell carcinoma, angiosarcoma, endometrial cancer,<br />

renal cell carcinoma (RCC, N�2), and pancreatic neuroendocrine tumor<br />

(PNET, N�2). At the time <strong>of</strong> enrollment, 6 had SD and one (endometrial<br />

cancer with multiple liver metastases) had an unconfirmed partial response<br />

(PR). The average length <strong>of</strong> time on treatment (including phase I and<br />

extension studies) was 9.5 months (range 5-19). As <strong>of</strong> January 2012, 3<br />

patients remain on study, including the endometrial cancer patient with an<br />

ongoing PR who has had �80% tumor regression after 19 months <strong>of</strong><br />

treatment at 0.7 mg/kg and two patients with RCC and PNET with<br />

continued SD after nearly 1 year <strong>of</strong> treatment at 1.0 mg/kg. The other<br />

patients with RCC and PNET at 1.0 mg/kg with SD came <strong>of</strong>f after 8.5 and<br />

5.5 months, respectively, for adverse events that included fatigue or<br />

elevated alkaline phosphatase. A decrease in spleen volume, likely an<br />

on-target effect and not associated with any adverse events, occurred to a<br />

greater degree on the extension study than on the phase I trial and was most<br />

pronounced in patients receiving � 12 doses. Conclusions: ALN-VSP02 has<br />

preliminary activity against endometrial cancer, RCC and PNET and a<br />

favorable safety pr<strong>of</strong>ile that permits chronic dosing. Phase II trials are<br />

warranted in these and other VEGF-overexpressing tumors.<br />

3061 General Poster Session (Board #15C), Mon, 8:00 AM-12:00 PM<br />

Improved sorafenib activity on hepatocellular carcinoma in Notch3 silenced<br />

in vivo and in vitro models. Presenting Author: Michele Baglioni,<br />

Department <strong>of</strong> <strong>Clinical</strong> Medicine, University <strong>of</strong> Bologna, and Center for<br />

Applied Biomedical Research (CRBA), S. Orsola-Malpighi Hospital, Bologna,<br />

Italy<br />

Background: Sorafenib is the only approved drug for the treatment <strong>of</strong> the<br />

advanced stage <strong>of</strong> HCC. Although its efficacy has been proven with<br />

randomized clinical trials, the clinical benefit seen in overall survival for<br />

advanced HCC patients treated with sorafenib could be improved. New<br />

molecules or combination <strong>of</strong> novel targeted agents to improve sorafenib<br />

efficacy for advanced stage HCC patients are needed. Notch genes are a<br />

family <strong>of</strong> receptors involved in many cell fate regulations, their expression<br />

has been found altered in many tumors, including HCC. The aim <strong>of</strong> this<br />

study was to study the combination <strong>of</strong> sorafenib and Notch3 signaling<br />

inhibition to improve sorafenib’s therapeutic effect. Methods: HepG2 and<br />

Huh7 cellular models were used for Notch3 stable silencing by retroviral<br />

introduction <strong>of</strong> specific interfering RNAs Xenograft models in both Notch3<br />

stable shRNA cell lines have been developed using NOD/SCID mice.<br />

Animals bearing tumors were treated with 60 mg/kg <strong>of</strong> sorafenib for 21<br />

consecutive days. Notch3, p21 and pGSK3�Ser9 protein expression were<br />

also analyzed in 20 human HCCs. Results: Notch3 silencing (shN3) in<br />

HuH7 and HepG2 cell lines treated with sorafenib showed an increase in<br />

cell death (3.8 to 5 fold increase) when shN3 cell lines were compared with<br />

their relative controls (GL2). A difference in tumor growth was observed<br />

between GL2 negative control vs shN3 xenografts in both HepG2 and Huh7<br />

after 21 days <strong>of</strong> sorafenib treatment. Two tailed student’s t test (shN3 vs<br />

GL2) P�0,04 and P�0,01 for Huh7 and HepG2 respectively. Molecular<br />

investigations have shown the involvement <strong>of</strong> p21 and GSK3� in the<br />

enhanced response to sorafenib in Notch 3 silenced models. A significant<br />

inverse correlation between Notch3 and pGSK3�Ser9 proteins accumulation<br />

(Spearman �� -0.666) (p � 0.01) and a direct correlation between<br />

Notch3 and p21 proteins expression (Spearman �� 0.681) (p � 0.01) was<br />

found in HCC samples obtained from patients. Conclusion: Our preclincal<br />

findings outline the effect <strong>of</strong> combined Notch3 inhibition and sorafenib.<br />

The molecular mechanisms responsible for sorafenib induced cell death<br />

associated with Notch3 silencing relays on inhibition <strong>of</strong> p21/CDKN1A and<br />

GSK3�Ser9 . This study is supported by a grant from Bayer Healthcare, Italy.<br />

3063 General Poster Session (Board #15E), Mon, 8:00 AM-12:00 PM<br />

Evaluation <strong>of</strong> toxicities <strong>of</strong> target therapy phase I and II trials on glioblastoma<br />

multiforme patients treated by curative radiotherapy: A metaanalysis.<br />

Presenting Author: Marcos Antonio Santos, Institut Gustave<br />

Roussy, Villejuif, France<br />

Background: An important limitation <strong>of</strong> the combination <strong>of</strong> target therapies<br />

(TT) to radiation (RT) is its potentially high level <strong>of</strong> toxicity. The objective <strong>of</strong><br />

this study was to describe and quantify the drug related acute adverse<br />

events presented by patients taking part in phase I and/or phase II trials on<br />

TT combined to curative RT for the treatment <strong>of</strong> glioblastoma multiforme<br />

(GBM). Methods: A meta-analysis <strong>of</strong> summary data including all phase I, I/II<br />

and II trials published between 2000 and 2011 on newly diagnosed GBM<br />

patients treated by TT with RT was performed. Pooled incidence rates (IR)<br />

<strong>of</strong> all and specific toxicities were estimated using a generalized linear<br />

mixed model with fixed and random study effects. The pooled median <strong>of</strong><br />

progression-free (PFS) and overall survivals (OS) were also considered.<br />

These results were compared to those obtained by patients receiving<br />

standard treatment (ST). Results: Twenty-one trials (8 phases I, 3 phases<br />

I/II and 10 phases II), testing 9 different drugs, have been selected,<br />

including 915 patients. The median follow-up varied from 1.4 to 34.0<br />

months. The estimated pooled IR <strong>of</strong> all toxicities was 68.6 [53.5-88.0] per<br />

1000 person-months <strong>of</strong> follow-up, compared to 20.7 [17.6-24.1] on ST<br />

(p�0.001). The pooled IRs <strong>of</strong> thrombocytopenia, neutropenia or leucopenia,<br />

fatigue, gastrointestinal events and treatment-related deaths are 11.2<br />

[7.5-16.9], 9.0 [6.1-13.4], 6.4 [4.1-10.0], 5.5 [2.9-10.2] and 3.2<br />

[1.6-6.4], compared to 4.1 [2.8-5.8], 4.9 [3.5-6.6], 4.6 [3.2-6.4], 0.8<br />

[0.3-1.6] and 0.1 [0.0-0.6], respectively, on ST. Median survival times are<br />

7.7 [6.1-9.8] and 13.8 months [11.9-16.1] on TT studies, and 6.9<br />

[5.8-8.2] and 14.6 [13.2-16.8], on ST, for PFS and OS, respectively,<br />

without statistically significant difference (p�0.39 and 0.55). The heterogeneity<br />

among specific toxicities and survival endpoints may be partially<br />

explained by the study design and/or the intake or not <strong>of</strong> temozolomide.<br />

Conclusions: The use <strong>of</strong> TT combined to RT represented a significative<br />

increase in severe adverse events in patients with GBM compared to ST,<br />

whereas no significant difference was observed in survival endpoints.<br />

Grants from French Cancer League.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


3064 General Poster Session (Board #15F), Mon, 8:00 AM-12:00 PM<br />

Microarray genomic expression (MAGE)-based therapy for heavily pretreated<br />

patients with metastatic solid tumors. Presenting Author: Joseba<br />

Rebollo, Plataforma de Oncología, USP-Hospital San Jaime, Torrevieja,<br />

Spain<br />

Background: Oligonucleotide-MAGE assay to identify tumor targets and<br />

potential active drugs based on molecular pr<strong>of</strong>iling in chemotherapy<br />

resistant/refractory (R/R) patients with metastatic solid tumors. A potential<br />

clinical benefit has been observed by Von H<strong>of</strong>f et al (JCO 2010;20:4877-<br />

83). Methods: Patients (pts) with R/R metastatic solid tumors were<br />

included in the study. Pathology confirmed fresh-frozen biopsies were<br />

obtained and molecular pr<strong>of</strong>iling oligonucleotide-MAGE (Agilent Whole<br />

Human Genome 4x44K) results, confirmed whenever possible by IHC, were<br />

used to identify potential therapy targets. Results were discussed and<br />

<strong>of</strong>fered to pts and the response evaluated using RECIST criteria. OS and<br />

PFS were measured from the beginning <strong>of</strong> therapy. All pts signed informed<br />

consent. Results: From August 2010 we performed 70 procedures in 69<br />

pts. No complications derived from the biopsy were observed. Thirty pts did<br />

not follow therapeutic recommendations and 9 pts are too early to evaluate.<br />

Thirty procedures from 29 pts are clinically evaluable. Ten pts were male<br />

and 19 female, with a median (M) age <strong>of</strong> 57 years (range 41-70). Seven pts<br />

had breast cancer, 5 NSCLC, 5 pancreas, 4 CRC, 2 ovary and one each <strong>of</strong><br />

gastric, parotid gland, biliary tree, head and neck, esophagus and gallbladder<br />

cancers, respectively. The M previous treatments were 3 (range 1 to<br />

�6). Oligonucleotide-MAGE assay confirmed resistance in 82% <strong>of</strong> drugs<br />

previously used. AM<strong>of</strong>3conventional (range 0-5) and 2 experimental<br />

(range 0-4) potentially active drugs were identified. <strong>Part</strong>ial response was<br />

observed in 7 (26.9%), stable disease �2 months in 12 (46.1%) and<br />

progression in 7 (26.9%) pts. Four pts were not evaluable. MPFS was 4<br />

months (95%CI: 1.815-6.185), MOS was 6 months (95%CI: 3.4-8.6) and<br />

1-year projected OS was 21%. Conclusions: We confirmed that it is possible<br />

to identify new potentially active drugs in heavily pretreated solid tumor pts<br />

with oligonucleotide-MAGE assay. Therapy based on these drugs showed to<br />

be active in one fourth <strong>of</strong> treated pts. These assays might be also integrated<br />

in early tumor stages to aid in the selection <strong>of</strong> chemotherapy drugs, and for<br />

this reason deserve further studies. Updated results will be presented.<br />

3066 General Poster Session (Board #15H), Mon, 8:00 AM-12:00 PM<br />

Feasibility to obtain a chemogram in circulating tumorigenic cells to guide<br />

further treatments in refractory solid tumors. Presenting Author: Antonio<br />

Cubillo, START-Madrid, Centro Integral Oncológico Clara Campal, Madrid,<br />

Spain<br />

Background: Circulating tumorigenic cells (CTCs) may be responsible for<br />

tumor progression, represent a new source <strong>of</strong> information <strong>of</strong> tumor biology,<br />

avoiding unnecessary biopsies. Gene-expression analysis (GEA) <strong>of</strong> CTCs<br />

could guide anti-cancer therapies in patients with refractory solid tumors.<br />

Methods: Pts �18y ECOG PS 0-2 with advanced refractory solid tumors<br />

were recruited. Twelve ml blood samples were collected from each pt and<br />

processed for isolation <strong>of</strong> CTCs using a novel cell adhesion assay (Vita-Cap,<br />

Vitatex Inc., Stony Brook, NY) following the manufacturer protocol. Total<br />

RNA was extracted with RNeasy Mini Kit (Qiagen Inc.) and GEA was<br />

performed with Affymetrix GeneChip Human Genome U133 Plus 2.0 array.<br />

The CEL file obtained for each pt sample was used in a gene-set expression<br />

analysis (GSEA) to determine the enrichment pr<strong>of</strong>ile <strong>of</strong> the pt sample to a<br />

pre-determined set <strong>of</strong> 12 PGx-modeled drugs created from the GI50 data <strong>of</strong><br />

the NCI-60 cell lines. The drug with the highest Normalized Enrichment<br />

Score (NES) obtained from the GSEA was selected as the most sensitive.<br />

The NES for each drug was standardized and rank-ordered according to<br />

Sensitive/Intermediate/Resistant categories to generate a nominal chemosensitivity<br />

pr<strong>of</strong>ile for the pt. This generates a comprehensible report called<br />

chemogram (ChG). We assessed preliminary efficacy in pts treated based<br />

on ChG. Results: ChG was obtained in 74/75 pts (57% male/43% female,<br />

median age 57y [20-81]) Tumor’s type was GI 67% (CRC n�18;<br />

pancreatic n�18; gastric n�6; cholangiocarcinoma n�7), NSCLC 14%,<br />

breast 8%, sarcoma 8%, others 3%. Median <strong>of</strong> 2 previous lines <strong>of</strong> therapy<br />

[range 1-12]. ECOG PS was 1 in 56%. Average RNA/pt 2.4 ng/�l (95% CI<br />

1.5-3.4) Median time to obtain the report from blood extraction was 4 wks<br />

[range 2-7] Pts treated after the ChG were 37 (49%), <strong>of</strong> them 25 (69%)<br />

based on ChG recommended drug. In patients treated with ChG sensitive<br />

drugs, PFSratio � 1.3 (PFS on therapy after ChG/PFS on prior therapy) was<br />

24%, and 3 partial responses (12%) were observed. Conclusions: It is<br />

feasible to generate a sensitivity drug pr<strong>of</strong>iling on CTCs which shows<br />

preliminary activity and merits further studies.<br />

Developmental Therapeutics—Experimental Therapeutics<br />

189s<br />

3065 General Poster Session (Board #15G), Mon, 8:00 AM-12:00 PM<br />

High concordance for MammaPrint 70 genes by RNA next generation<br />

sequencing. Presenting Author: Lorenza Mittempergher, Netherlands Cancer<br />

Institute (NKI-AVL), Amsterdam, Netherlands<br />

Background: The development <strong>of</strong> new biomarkers <strong>of</strong>ten requires fresh<br />

frozen (FF) samples. Recently we showed that microarray gene expression<br />

data generated from FFPE material are comparable to data extracted from<br />

the FF counterpart, including known signatures such as the 70-gene<br />

prognosis signature (Mittempergher L et al., 2011). As described by Luo et<br />

al (2010) RNA pr<strong>of</strong>iling using next generation sequencing (RNA-Seq) is<br />

now applicable to archival FFPE specimens. Methods: Technical performance<br />

and the comparison between the RNA-Seq 70-gene read-out and<br />

the MammaPrint test (Glas et al., 2006) is evaluated in a series <strong>of</strong> 15<br />

patients (11/15 with matched FFPE/FF material). RNA-Seq was carried out<br />

using minor adjustments <strong>of</strong> the Illumina TruSeq RNA preparation method.<br />

RNA sequencing libraries were prepared starting from 100ng <strong>of</strong> total RNA.<br />

Next, the DSN (Duplex-Specific Nuclease) normalization process was used<br />

to remove ribosomal RNA and other abundant transcripts (Luo et al, 2010).<br />

The libraries were paired-end sequenced on the Illumina HiSeq 2000<br />

instrument with multiplexing <strong>of</strong> 4 libraries per lane. The resulting sequences<br />

were mapped to the human reference genome (build 37) using<br />

TopHat 1.3.1(Trapnell et al., 2009). The HTSeq-count tool was used to<br />

generate the total number <strong>of</strong> uniquely mapped reads for each gene. Results:<br />

Between 14% and 45% <strong>of</strong> the total number <strong>of</strong> reads were assigned to<br />

protein-coding genes. The minimum coverage per 1000bp <strong>of</strong> CDS was 38<br />

reads. The 70 MammaPrint genes were successfully mapped to the<br />

RNA-Seq transcripts. We calculated the Pearson correlation coefficient<br />

between the centroids <strong>of</strong> the original good prognosis template (van’t Veer et<br />

al., 2002) and the 70-gene read count determined by RNA-Seq <strong>of</strong> each<br />

sample. Predictions based on the 70-gene RNA-Seq data showed a high<br />

agreement with the actual MammaPrint test predictions (�90%), irrespective<br />

<strong>of</strong> whether the RNA-seq was performed on FF or FFPE tissue.<br />

Conclusions: New generation RNA-sequencing is a feasible technology to<br />

assess diagnostic signatures.<br />

3067 General Poster Session (Board #16A), Mon, 8:00 AM-12:00 PM<br />

ME-143, a novel inhibitor <strong>of</strong> tumor-specific NADH oxidase (tNOX): Results<br />

from a first-in-human phase I study. Presenting Author: Carla D. Kurkjian,<br />

University <strong>of</strong> Oklahoma Health Sciences Center, Oklahoma City, OK<br />

Background: ME-143, a second generation is<strong>of</strong>lavone-derived compound,<br />

specifically binds tNOX, shifting the ceramide-S1P equilibrium and resulting<br />

in prompt apoptosis via pleotrophic caspase activation. A first generation<br />

compound, phenoxodiol (PXD), showed promising phase II activity<br />

with cisplatin in ovarian cancer. ME-143 is broadly active against human<br />

cancers in both in vitro and in vivo models, with IC50’s 1-2 logs lower than<br />

PXD. Toxicology studies up to 140mg/kg (human dose equivalent ~23mg/<br />

kg) showed no STD level. The only significant findings were dose dependent<br />

hypospermia and testicular atrophy in rats. We report clinical and PK<br />

results from the first-in-human phase I study. Methods: A 3�3 dose<br />

escalation design was used with 4 dose cohorts: 2.5, 5, 10, and 20mg/kg<br />

IV over 30 minutes weekly times 3, followed by a 1 week break, and then<br />

continuous weekly dosing in patients with advanced solid tumors. Dense<br />

PK sampling was performed at 0, 5, 10, 20, 30, 60, 90, 120, 180, 240,<br />

300, 360 minutes, and 24 hours post-infusion day 1 and 15 <strong>of</strong> the first<br />

treatment cycle. Results: To date, 9 patients have been enrolled, 3 in each<br />

<strong>of</strong> the first 3 cohorts. Median time on treatment is 56 days (range 6 to 62).<br />

Five patients have discontinued protocol therapy, all due to PD. No DLT’s<br />

have been observed. Related AE’s include grade 1: myalgia (1), anorexia<br />

(1), fatigue (2), headache (1), diarrhea (1), vomiting (1) and grade 2:<br />

fatigue (1). Preliminary PK analyses in the first 2 cohorts is shown in table<br />

below. Conclusions: ME-143 appears to be well tolerated when administered<br />

IV. Preliminary PK data suggest that drug levels achieve target<br />

concentration extrapolated from pre-clinical studies (AUC0-t ~10mg*hr/<br />

mL) and exceed levels obtained with IV PXD in the phase II study (AUC0-t ~2mg*hr/mL) . Updated clinical data from all planned dose cohorts,<br />

including the final pharmacokinetic analysis and planned phase II dose,<br />

will be presented.<br />

Day 1 Day 15<br />

Dose<br />

2.5mg/kg 5.0mg/kg 2.5mg/kg 5.0mg/kg<br />

Cmax (mg/mL)<br />

T1/2 (hr)<br />

AUC0-t (mg*hr/mL)<br />

CL (L/hr)<br />

2.18<br />

4.12<br />

2.93<br />

69.73<br />

8.65<br />

6.95<br />

9.92<br />

37.30<br />

3.55<br />

4.36<br />

3.53<br />

56.18<br />

5.31<br />

5.55<br />

7.29<br />

41.52<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


190s Developmental Therapeutics—Experimental Therapeutics<br />

3068 General Poster Session (Board #16B), Mon, 8:00 AM-12:00 PM<br />

Integrated next-generation sequencing and patient-derived xenografts to<br />

personalized cancer treatment. Presenting Author: Elena Garralda, <strong>Clinical</strong><br />

Research Programme. Spanish National Cancer Research Center (CNIO),<br />

Madrid, Spain<br />

Background: The knowledge <strong>of</strong> actionable somatic genomic alterations<br />

present in each tumor is making possible the era <strong>of</strong> personalized cancer<br />

treatment. Methods: Using massively parallel sequencing we performed<br />

whole exome sequencing analysis <strong>of</strong> tumor and matched normal blood<br />

samples <strong>of</strong> 8 patients (2 pancreatic adenocarcinoma, 1 neuroendocrine<br />

tumor, 1 glioblastoma, 1 uveal melanoma, 1 colon cancer) to identify<br />

putatively actionable tumor specific genomic alterations. We used 2 in<br />

silico methods (Polyphen and SIFT) to estimate the functional significance<br />

<strong>of</strong> a given confirmed mutation. Primary xenografts (PDX), generated by<br />

direct engraftment <strong>of</strong> tumor samples from the patients into immunocompromised<br />

mice, were used as an in vivo platform that provided the opportunity<br />

to test proposed personalized medicine strategies. Results: At this time<br />

exome sequencing analyses have been performed for 5 patients (1 patient<br />

died prematurely, 1 tumor sample was insufficient, 1 result is pending).<br />

More than 30 million bases <strong>of</strong> target DNA were analyzed in the tumor and<br />

normal samples in every case, with at least 70 distinct reads at each base.<br />

Tumor specific mutations (Muts) and copy number variations (CNVs) were<br />

identified: 5 Muts in the neuroendocrine tumor; 62Muts/6CNVs and<br />

38Muts/10CNVs in the pancreatic tumors; 63Muts/23CNVs in the glioblastoma;<br />

5 Muts in the melanoma. All samples pr<strong>of</strong>iled contained actionable<br />

alterations with the most relevant mutations affecting NF1, PTPN11,<br />

EPHA3, CDKN2A, FAS (glioblastoma); PI3KCA, ARID1A, ARID1B, DDR2,<br />

SMAD4, TP53, KRAS, PTCHD3 (pancreatic); CREB3L3, ITPR2 (neuroendocrine);<br />

GNA11, TAOK3 (melanoma). PDX from the pancreatic cancer<br />

patient was treated with a PI3K inhibitor and dasatinib, reported to be<br />

effective in discodin domain receptor 2 (DDR2) mutant cancer with no<br />

effect. Accordingly treatment <strong>of</strong> that patient with dasatinib was not<br />

effective and the level <strong>of</strong> this mutation in the tumor was observed to be low.<br />

Conclusions: Detection <strong>of</strong> actionable tumor-specific genomic alterations in<br />

the clinical setting is feasible. In silico methods and primary xenografts can<br />

help in the challenge <strong>of</strong> linking confirmed mutations to protein function<br />

and ultimately to clinical utility.<br />

3070 General Poster Session (Board #16D), Mon, 8:00 AM-12:00 PM<br />

OM-RCA-01, an FGFR1 specific humanized antibody for the treatment <strong>of</strong><br />

renal cell carcinoma (RCC). Presenting Author: Ilya Tsimafeyeu, Kidney<br />

Cancer Research Bureau, Moscow, Russia<br />

Background: Fibroblast growth factor (FGF) receptor 1 (FGFR1) is a<br />

potential therapeutic target for the treatment <strong>of</strong> metastatic RCC. We<br />

investigated the preclinical activity <strong>of</strong> OM-RCA-01, a novel therapeutic<br />

humanized anti-FGFR1 antibody with high affinity (Kd <strong>of</strong> 1.59 nM), in<br />

RCC. Methods: To assess the effect <strong>of</strong> anti-FGFR1 antibody on FGFmediated<br />

signaling, the human renal carcinoma Caki-1 FGFR1-expressing<br />

cells were dosed with OM-RCA-01 at 100, 10, and 1 mcg/ml. Control wells<br />

were left untreated. Three hours after dosing, bFGF was added at a<br />

concentration <strong>of</strong> 50 ng/ml. Additional control wells were treated with<br />

OM-RCA-01 without FGF-stimulation. Cell growth inhibition was determined<br />

using Promega’s Cell Titer-Glo assay. CR female NCr nu/nu mice<br />

were set up with 1 mm3 Caki-1 tumor fragments sc in flank. Tumor sizes<br />

were measured in a blind fashion twice a week with a vernier caliper. Mice<br />

with established tumors were randomly divided into vehicle, non-specific<br />

IgG or OM-RCA-01 groups per 10 animals in group. Endpoint was<br />

significant differences in tumor growth delay. Results: In vitro study showed<br />

that bFGF increased proliferation <strong>of</strong> the human FGFR1-expressing renal<br />

carcinoma cells (p�0.011). OM-RCA-01 antibody significantly inhibits<br />

FGF-triggered cell proliferation in comparison with control. In vivo, the<br />

tumors in untreated mice or mice treated with non-specific IgG continued<br />

their aggressive growth to reach the size <strong>of</strong> 2000 cm3, at which point the<br />

mice were killed. In contrast, treatment with OM-RCA-01 not only significant<br />

arrested further growth <strong>of</strong> the tumors (p�0.006) but also demonstrated<br />

differences in tumor volume compared with vehicle already on Day<br />

13. A similar anti-tumor activity <strong>of</strong> OM-RCA-01 was observed when the<br />

antibody was given in low (1 mg/kg) or high (10 mg/kg) doses (p�0.917).<br />

Administration <strong>of</strong> 10 mg/kg antibody for up to 35 days resulted in minimal<br />

body weight loss and no observations <strong>of</strong> gross toxicity were made.<br />

Conclusions: Targeting FGFR1 blocks FGF/FGFR1 pathway in RCC. Monoclonal<br />

antibody OM-RCA-01 has significant early anti-tumor efficacy in<br />

Caki-1 xenograft model. Isolated blocking <strong>of</strong> FGFR1 by low-dose antibody<br />

could be safe and effective.<br />

3069 General Poster Session (Board #16C), Mon, 8:00 AM-12:00 PM<br />

CD30 expression in nonlymphomatous malignancies. Presenting Author:<br />

Jeff Porter Sharman, Willamette Valley Cancer Institute and Research<br />

Center, Eugene, OR<br />

Background: CD30 is commonly expressed in Hodgkin lymphoma (HL),<br />

anaplastic large cell lymphoma (ALCL), and testicular embryonal carcinoma.<br />

Expression <strong>of</strong> CD30 in other solid tumors and non-lymphomatous<br />

malignancies has been reported but not investigated systematically. CD30<br />

is the target <strong>of</strong> brentuximab vedotin (Adcetris), an antibody drug conjugate<br />

(ADC) that is approved for the treatment <strong>of</strong> patients with relapsed HL and<br />

systemic ALCL after failure <strong>of</strong> other therapies. A study was initiated to<br />

determine the incidence <strong>of</strong> CD30 expression in non-lymphomatous malignancies<br />

and to identify patients who may be candidates for treatment with<br />

brentuximab vedotin. Methods: Patients with non-lymphomatous malignancies<br />

were eligible for screening if they were relapsed or refractory to<br />

previous therapy or had no effective treatment options available. Archived<br />

tissue from solid tumors was tested for CD30 expression by immunohistochemistry<br />

(IHC); fresh bone marrow or blood samples from multiple<br />

myeloma or leukemia patients were tested by flow cytometry. Patients were<br />

considered CD30 positive and eligible for a companion treatment protocol<br />

with brentuximab vedotin if �10% <strong>of</strong> malignant cells stained positive by<br />

IHC or �20% by flow cytometry. Results: At this interim analysis, a total <strong>of</strong><br />

875 patients have been tested for CD30 expression: 95% had solid tumors,<br />

3% had leukemia, and 2% had multiple myeloma. Twenty-two patients<br />

(2.5%) were CD30 positive, including 7 <strong>of</strong> 94 patients with ovarian cancer<br />

(7%), 5 <strong>of</strong> 20 with melanoma (25%), 2 <strong>of</strong> 5 with mesothelioma (40%), 1 <strong>of</strong><br />

4 with skin squamous cell carcinoma (25%), 2 <strong>of</strong> 41 with triple negative<br />

breast cancer (5%), 1 <strong>of</strong> 37 with pancreatic cancer (3%), 1 <strong>of</strong> 26 with<br />

small cell lung cancer (4%), and 1 <strong>of</strong> 3 with anal cancer (33%), and thyroid<br />

carcinoma (33%). One patient was identified with CD30-positive mast cell<br />

leukemia. In positive patients, the percent <strong>of</strong> CD30-positive malignant<br />

cells varied between 10 and 80%. Conclusions: CD30 expression was<br />

observed in multiple types <strong>of</strong> non-lymphomatous malignancies, thereby<br />

identifying additional populations who may be candidates for treatment<br />

with a CD30-targeted ADC, such as brentuximab vedotin. A companion<br />

clinical trial with brentuximab vedotin is currently ongoing.<br />

3071 General Poster Session (Board #16E), Mon, 8:00 AM-12:00 PM<br />

<strong>Clinical</strong> significance <strong>of</strong> PRL-3 genomic amplification/expression pr<strong>of</strong>iles<br />

and preclinical study <strong>of</strong> PRL-3 inhibitor in human gastrointestinal cancers.<br />

Presenting Author: Keishi Yamashita, Kitasato University School <strong>of</strong> Medicine,<br />

Sagamihara, Japan<br />

Background: PRL (phosphatase <strong>of</strong> regenerating liver) is a tyrosine phosphatase<br />

that dephosphorylates molecules involved in cancer invasion and<br />

metastasis and subsequently activates either PI3 kinase or Src pathway.<br />

PRL-3 was initially identified as a gene that is amplified in metastasis <strong>of</strong><br />

colorectal cancer. We are thinking much <strong>of</strong> clinical potential <strong>of</strong> PRL-3<br />

inhibitor as a molecular targeted therapy in GI cancer. Methods: Immunohistochemistry<br />

and genomic status were assessed for PRL-3 in esophageal<br />

squamous cell carcinoma (ESCC)(n�88), gastric adenocarcinoma (n�173),<br />

and colorectal adenocarcinoma (n�107).RNA interference was used to<br />

determine its specific functional roles in GI cancers. Preclinical study using<br />

PRL-3 inhibitors (1-4-bromo-2-benzylidene rhodanine) was performed in 4<br />

ESCC and 4 gastric cancer cell lines that were intensely expressed for<br />

PRL-3, or TE5 which showed basal level expression. Results: (1) PRL-3<br />

expression is associated with lymph node metastasis in primary ESCC<br />

(p�0.0012), gastric adenocarcinoma (p�0.0001), and colorectal carcinoma<br />

(p�0.0001). In the primary tumors with lymph node metastasis, its<br />

expression was recognized in 96, 80, 86%, respectively. (2) If restricted to<br />

patients with PRL-3 expression, genomic amplification was found exclusively<br />

in stage IV ESCC (30%), and stage III/IV gastric cancer (40%). In<br />

colorectal cancer, PRL-3 genomic amplification is found in 35% <strong>of</strong> stage<br />

III/IV, but only in 10% <strong>of</strong> stage II (p�0.002). (3) PRL-3 RNA interference<br />

robustly suppressed metastatic cancer phenotypes. (4) PRL-3 inhibitor<br />

suppressed phenotypes as well in a dose dependent manner (0, 1, 10, and<br />

50 mM). Interestingly, 10 mM <strong>of</strong> the inhibitor could not suppress muscle<br />

cell (C2C12) that endogenously express PRL-3, but 50 mM suppressed it.<br />

Conclusions: PRL-3 inhibitor has a great clinical potential to suppress<br />

tumors with metastatic ability, that was represented by lymph node<br />

metastasis or recurrent disease <strong>of</strong> GI cancers.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


3072 General Poster Session (Board #16F), Mon, 8:00 AM-12:00 PM<br />

Effect <strong>of</strong> MDV3100, a novel androgen receptor signaling inhibitor, on cell<br />

proliferation and tumor size in an apocrine breast cancer xenograft model.<br />

Presenting Author: Sebastián Bernales, Medivation, Inc., San Francisco,<br />

CA<br />

Background: MDV3100, a new androgen receptor (AR) signaling inhibitor,<br />

has demonstrated therapeutic effects in men with post-docetaxel prostate<br />

cancer (AFFIRM). AR is also expressed in the majority <strong>of</strong> breast cancers.<br />

Molecular pr<strong>of</strong>iling <strong>of</strong> estrogen receptor negative (ER-) breast cancer<br />

classifies this disease into basal and molecular apocrine subtypes. Apocrine<br />

breast cancer represents ~50% <strong>of</strong> ER- tumors and is characterized by<br />

a steroid response gene signature that includes AR expression. Here we<br />

report the effects <strong>of</strong> MDV3100 on the growth <strong>of</strong> the ER-/AR� breast cancer<br />

cell line MDA-MB-453 in tissue culture and in an in vivo xenograft mouse<br />

model. Methods: MDA-MB-453 cells were maintained in 5% complete or<br />

5% charcoal-stripped serum supplemented with dihydrotestosterone (CSS-<br />

DHT). The DHT induced nuclear translocation <strong>of</strong> AR was determined by<br />

subcellular fractionation followed by western blot. AR target gene expression<br />

was measured by qPCR. To generate a mouse xenograft model, 5-6 wk<br />

old female NOD-SCID-IL2Rgc–/– mice were injected ortotopically with<br />

6x106 MDA-MB-453 cells into the fourth inguinal gland and were administered<br />

exogenous DHT (12.5 mg in a 60 day release pellet) or control pellets.<br />

When tumor size reached 100 mm3 , mice were treated with 10 mg/kg/day<br />

MDV3100 or vehicle (0.5% methocel solution) by oral gavage for 33 days.<br />

Results: Viability <strong>of</strong> MDA-MB-453 cells treated with 10 uM MDV3100 was<br />

reduced by 10% after 3 days and 27% after 6 days. Treatment with 25 uM<br />

MDV3100 reduced cell viability by 27% after 3 days and 36% after 6 days.<br />

Similar results were obtained when cells maintained in CSS-DHT were<br />

treated with MDV3100. Treatment with MDV3100 blocked DHT-induced<br />

nuclear translocation and concordantly reduced the expression <strong>of</strong> ARtargets<br />

fatty acid synthase, prolactin receptor, and gross cystic disease<br />

protein-15. In the xenograft model, MDV3100 inhibited tumor growth by<br />

78% as compared to tumors treated with vehicle. Conclusions: MDV3100<br />

reduced cell proliferation in apocrine cell lines. MDV3100 also suppressed<br />

exogenous DHT-induced growth <strong>of</strong> ER-/AR� tumors in vivo. These results<br />

support the evaluation <strong>of</strong> MDV3100 as a treatment for apocrine tumors.<br />

3074 General Poster Session (Board #16H), Mon, 8:00 AM-12:00 PM<br />

Targeted therapies for the treatment <strong>of</strong> non-small cell lung cancer: Bull’s<br />

eye or missed target? Presenting Author: Shakun M. Malik, U.S. Food and<br />

Drug Administration, Silver Spring, MD<br />

Background: Targeted drugs approved for advanced non-small cell lung<br />

cancer (NSCLC) include bevacizumab [(monoclonal antibody directed<br />

against vascular endothelial growth factor-A (VEGF-A)], erlotinib [(an<br />

epidermal growth factor receptor kinase (EGFR) inhibitor)] and crizotinib<br />

[(an anaplastic lymphoma kinase (ALK) inhibitor)]. The development<br />

programs for erlotinib and bevacizumab did not prospectively employ in<br />

vitro diagnostic tests to select patients (pts). Post-hoc exploratory analyses<br />

for bevacizumab based on VEGF-A levels have not identified subgroups with<br />

differential treatment effects: however for erlotinib, pts with activating<br />

EGFR mutations had greater improvement in overall response rates (ORR)<br />

and progression-free survival. Crizotinib is the first targeted therapy for<br />

NSCLC in which pts were selected using an analytically validated investigational<br />

test for ALK translocations. Crizotinib received accelerated approval<br />

on August 26, 2011 for the treatment <strong>of</strong> pts with locally advanced or<br />

metastatic ALK-positive NSCLC as detected by an FDA-approved test (Vysis<br />

ALK Break Apart FISH Probe Kit) that was approved by FDA on the same<br />

day. Methods: NDA file for crizotinib was examined. Results: Two single arm<br />

trials assessing efficacy <strong>of</strong> crizotinib monotherapy in ALK positive pts<br />

showed ORR <strong>of</strong> 61% and 50%. At FDA’s request, a cohort <strong>of</strong> 23 pts that<br />

tested negative for the ALK translocation evaluated showed ORR <strong>of</strong> 26%.<br />

Two additional pts had a single assessment and, if confirmed, the response<br />

rate would be 37% in this “marker negative” subgroup. Further exploration<br />

<strong>of</strong> this subgroup is a postmarketing commitment. Conclusions: Evaluation<br />

<strong>of</strong> target status (positive or negative) in all pts provides optimal information<br />

to guide clinical practice. As observed with crizotinib, responses may be<br />

observed in the “marker negative” subgroup. This unexpected finding may<br />

reflect the drug’s activity via alternate pathways, selection <strong>of</strong> a suboptimal<br />

biomarker for assessing ALK activation, or selection <strong>of</strong> a suboptimal<br />

cut-point for positive/negative. Accurate diagnostic tests will facilitate drug<br />

development with possible greater observed treatment effect and potentially<br />

smaller clinical trials.<br />

Developmental Therapeutics—Experimental Therapeutics<br />

3073 General Poster Session (Board #16G), Mon, 8:00 AM-12:00 PM<br />

Inhibition <strong>of</strong> autophagy: Phase I safety, tolerability, and pharmacokinetics<br />

(PK) <strong>of</strong> hydroxychloroquine (H) in combination with vorinostat (V) in<br />

patients with advanced solid tumors. Presenting Author: Devalingam<br />

Mahalingam, Institute for Drug Development, Cancer Therapy and Research<br />

Center, University <strong>of</strong> Texas Health Science Center at San Antonio,<br />

San Antonio, TX<br />

Background: The histone deacetylase (HDAC) inhibitor V has pleiotropic<br />

effects, and induces both apoptosis and autophagy. As autophagy can<br />

promote cancer cell survival, we hypothesized that inhibiting autophagy<br />

would enhance the anti-cancer activity <strong>of</strong> V. Our pre-clinical studies<br />

showed that inhibition <strong>of</strong> autophagy using H significantly increased the<br />

pro-apoptotic effects <strong>of</strong> V. The primary objective <strong>of</strong> this NCI-funded study<br />

was to determine the maximum tolerated dose (MTD) <strong>of</strong> H in combination<br />

with V in patients (pts) with advanced solid tumors. Methods: Pts with ECOG<br />

PS 0-2 and adequate organ function received PO V 300-400 mg QD and H<br />

400-1000 mg QD in 21d cycles. 3�3 dose escalation. Dose limiting<br />

toxicities (DLTs) � non-hem toxicity � grade (Gr) 3 or hem toxicity � Gr 4<br />

in the first 2 cycles. PBMCs were analyzed for autophagic changes at<br />

baseline, week 1 � 6. PKs done on cycle 1.Tumor biopsies were performed<br />

at baseline and post-cycle 2 on 3 pts at MTD. Results: 31 pts enrolled, 27<br />

evaluable for DLT. No DLT was observed in cohort 1 (300 mg V/400 mg H)<br />

or cohort 2 (400 mg V/400 mg H). In cohort 3 (400 mg V/600 mg H) 1/6<br />

pts experienced a DLT <strong>of</strong> Gr 3 fatigue. Cohort 4 (400 mg V/800 mg H) 3/6<br />

pts had DLT <strong>of</strong> Gr 3 fatigue, and 1 had a Gr 2 seizure. Expansion <strong>of</strong> cohort 3<br />

resulted in no further DLTs. 10 pts got the MTD. Treatment-related<br />

toxicities were mostly Gr 1- 2: nausea/vomiting (10 pts), diarrhea (7),<br />

fatigue (5), anorexia (4), weight loss (3), anemia (4), elevated creatinine<br />

(4) and thrombocytopenia (2). Gr 3 toxicities were fatigue (4), anemia,<br />

thrombocytopenia and neutropenia (1 each). No drug-related deaths or Gr<br />

4 toxicities were noted. One pt (RCC) had a confirmed PR (cohort 2 active<br />

on cycle 32) and 2 pts with CRC had prolonged SD (�4 cycles). PK analysis<br />

indicated dose-proportional exposure for H. Changes from baseline in<br />

biomarkers <strong>of</strong> autophagy and HDAC inhibition were observed in PBMC and<br />

primary tumors with therapy. Conclusions: Recommended dose is V 400<br />

and H 600 mg QD. Dose-dependent fatigue was observed with the<br />

combination, but well tolerated at the MTD without any further increase in<br />

toxicities compared to V alone. Analyses <strong>of</strong> PK-PD interactions are<br />

underway.<br />

3075 General Poster Session (Board #17A), Mon, 8:00 AM-12:00 PM<br />

Translational assessment <strong>of</strong> the efficacy <strong>of</strong> CPI-613 against pancreatic<br />

cancer in animal models versus patients with stage IV disease. Presenting<br />

Author: Avi S. Retter, Eastchester Center for Cancer Care, Bronx, NY<br />

Background: CPI-613 is a novel agent that selectively targets the altered<br />

mitochondrial enzymes <strong>of</strong> tumor cells, causing apoptosis, necrosis, and<br />

autophagia. Results assessing clinical efficacy <strong>of</strong> CPI-613 translated from<br />

animal tumor xenograft models to patients with stage IV pancreatic cancer<br />

are presented. Methods: Efficacy <strong>of</strong> CPI-613 was tested in mice with<br />

pancreatic tumor xenografts generated by inoculation <strong>of</strong> BxPC-3 human<br />

pancreatic tumor cells. The safety and efficacy <strong>of</strong> CPI-613 (70-320<br />

mg/m2 ), when used in combination with gemcitabine (1,000 mg/m2 ), was<br />

assessed in patients with stage IV pancreatic cancer. Results: In the animal<br />

pancreatic tumor xenograft model (n�10/grp), CPI-613 (25 mg/kg, IV, 1x<br />

weekly for 4 weeks) suppressed tumor growth by ~100%, when compared<br />

to vehicle. The positive control, gemcitabine (50 mg/kg, IV, 1x weekly for 4<br />

weeks), suppressed tumor growth by only ~50%. Median overall survival in<br />

tumor-bearing mice treated with CPI-613 was ~240 days, which was<br />

significantly longer than those with gemcitabine or vehicle treatments (~65<br />

and ~50 days, respectively). In 6 humans with stage IV pancreatic cancer<br />

(Table), the CPI-613�gemcitabine combination was well-tolerated. In<br />

those without prior chemotherapy before participating in the clinical trial<br />

(first three patients in the table), the CPI-613�gemcitabine combination<br />

prolonged survival that correlated with the dose <strong>of</strong> CPI-613. Conclusions:<br />

CPI-613 exhibits efficacy against pancreatic cancer in animal models,<br />

which is translational to patients with stage IV disease.<br />

Patients with stage IV pancreatic cancer.<br />

ID# Sex Age Race/ethnicity<br />

Chemotherapy prior to<br />

study participation<br />

CPI-613 dose a<br />

(mg/m 2 )<br />

191s<br />

Survival b<br />

(months)<br />

1-106 M 52 Hispanic None 70 4.1<br />

1-110 F 65 Caucasian None 150 7.5<br />

1-113 F 77 African <strong>American</strong> None 190 �22 (still alive)<br />

1-107 M 63 Caucasian Gemcitabine 105 9.8<br />

1-111 F 76 African <strong>American</strong> 5-FU; gemcitabine;<br />

FOLFOX � transferrinlaced<br />

oxaliplatin<br />

150 11.3<br />

2-107 M 65 Caucasian Gemcitabine � erlotinib;<br />

Xenoda � oxaliplatin<br />

320 �10<br />

Abbreviations: f � female; ID � identification; M � male; mo � month. a CPI-613 was given in<br />

combination with gemcitabine (1,000 mg/m 2 ). b Survival was measured since the start <strong>of</strong> first<br />

chemotherapy the patient received.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


192s Developmental Therapeutics—Experimental Therapeutics<br />

3076 General Poster Session (Board #17B), Mon, 8:00 AM-12:00 PM<br />

A phase I study <strong>of</strong> HM781-36B, a novel pan-HER inhibitor, in patients (pts)<br />

with advanced solid tumors. Presenting Author: Tae Min Kim, Department<br />

<strong>of</strong> Internal Medicine, Seoul National University Hospital, Seoul, South<br />

Korea<br />

Background: HM781-36B is a pan-HER tyrosine kinase inhibitor, which<br />

showed a potent activity against the gefitinib- or erlotinib-resistant, EGFR<br />

L858R/T790M double mutant cells. A phase I study was conducted to<br />

determine the MTD, pharmacokinetics, and antitumor activity. Methods:<br />

Eligible pts had advanced malignancies refractory to standard therapies.<br />

Standard 3�3 scheme was used in the dose escalation part, and additional<br />

12 pts were enrolled in the expansion cohort <strong>of</strong> molecular enrichment.<br />

Results: In dose-escalation part, 43 pts (median age: 55 yrs (range 25-82),<br />

M:F�25:18, ECOG PS 0/1/2/3: 23/17/2/1, median prior chemotherapy: 4)<br />

were treated. DLTs were G3 diarrheas in 5 pts, one at 12 mg, 16 mg, 24<br />

mg, and two at 32 mg. The MTD was determined as 24mg. The most<br />

common drug-related adverse events were diarrhea, stomatitis, rash,<br />

pruritus, and anorexia. Among 41 evaluable pts, 4 pts achieved PR (1<br />

unconfirmed, duration <strong>of</strong> response: 11.9 mo, 7.07 mo�, 4.5 mo�), and<br />

19 pts had SD. Two <strong>of</strong> 4 PR pts were Her2-positive breast cancer pts. The<br />

median duration <strong>of</strong> treatment in pts with PR or SD was 3.87 (2.47- 15.17)<br />

months. In the dose range <strong>of</strong> 0.5 to 24 mg, it showed linear pharmacokinetics<br />

proportional to dose-escalation, relatively short half-life, and little<br />

accumulation. Additional 12 pts in the expansion cohort are under<br />

treatment at 24 mg (6 pts: EGFR-mutant NSCLC, 3 pts: Her2-positive<br />

gastric cancer, 2 pts: Her2-positive breast cancer, 1 pt: rectal cancer).<br />

Conclusions: HM781-36B was safe and well tolerable in advanced solid<br />

tumors. Preliminary evidence <strong>of</strong> anticancer activity has been observed.<br />

Updated data will be presented at the meeting.<br />

3078 General Poster Session (Board #17D), Mon, 8:00 AM-12:00 PM<br />

Phase I pharmacokinetic study <strong>of</strong> dasatinib (BMS-354825) in patients<br />

with advanced malignancies and varying levels <strong>of</strong> liver dysfunction: S0711,<br />

a SWOG early therapeutics committee study. Presenting Author: John<br />

Sarantopoulos, Institute for Drug Development, Cancer Therapy and<br />

Research Center, University <strong>of</strong> Texas Health Science Center San Antonio,<br />

San Antonio, TX<br />

Background: Dasatinib (D) is a first in class Src kinase inhibitor, and inhibits<br />

BCR-Abl, c-Kit, PDGFR-beta, EPHA2 and Src family kinases including Src,<br />

Lck, Yes, Fyn at nanomolar concentrations. Initially FDA approved for use<br />

in imatinib resistant CML. It is a small molecule targeted therapy<br />

hepatically metabolized primarily by CYP3A4. We conducted a phase I<br />

study to determine maximum tolerated dose (MTD) and pharmacokinetics<br />

(PK) <strong>of</strong> D in patients (pts) with liver dysfunction (LD). Methods: Pts with<br />

advanced solid tumors or lymphoma, Zubrod �2, no baseline ascites or<br />

pleural effusions, adequate renal and bone marrow function, received PO D<br />

daily. Cycles q28 days. Pts stratified into 4 LD groups: normal, mild,<br />

moderate, severe, using Child-Pugh classification (CPC). Data also collected<br />

for NCI ODWG Organ Dysfunction Working GroupCriteria. D dose was<br />

escalated in sequential cohorts <strong>of</strong> pts within each LD category. Blood<br />

analysis for D concentrations were determined during cycle 1 using a<br />

validated LC-MC/MS assay. Study objectives included characterizing safety,<br />

tolerability, PK, identifying the MTD and obtaining preliminary evidence <strong>of</strong><br />

efficacy. Results: 54 registered pts, 51 pts received 51 cycles <strong>of</strong> D at doses<br />

starting at 100 mg in mild LD (50-140 mg). Median age 60, male 55%,<br />

Zubrod 1 70%. CRC 27%. Groups: normal-17, mild-20, moderate-13,<br />

severe-1 pt(s). Related AEs include fatigue 35%, diarrhea 27%, anemia<br />

27%, nausea 25%, vomiting 21%, lymphopenia 13%, rash 13%, pleural<br />

effusion 8%. 1 DLT <strong>of</strong> increased CK in a pt in moderate LD with past history<br />

<strong>of</strong> similar episode with previous sorafenib. Previous linear PK disposition,<br />

and no accumulation. No apparent PK differences between normal and<br />

mild groups. Cycle 1 Day 1 D 140 mg Normal Group: Cmax 129 ng/mL. Mild<br />

Group 140mg: Cmax 157 ng/mL. Prolonged disease stabilization (�4<br />

cycles) in 6 pts, 3 CRC (4,5,8); 1 Pancreas, HCC, Bladder (4,5,6).<br />

Conclusions: Recommended dose for Dasatinib given PO QD for pts with<br />

mild, moderate, or severe LD using clinical criteria with CPC and no<br />

baseline ascites, are 140 mg, 70 mg, insufficient pts, respectively. Dose<br />

adjustment not necessary in pts with mild LD.<br />

3077 General Poster Session (Board #17C), Mon, 8:00 AM-12:00 PM<br />

Tumor drug distribution and target engagement <strong>of</strong> MLN9708, an investigational<br />

proteasome inhibitor, in patients with advanced solid tumors.<br />

Presenting Author: Alessandra Di Bacco, Translational Medicine, Millennium<br />

Pharmaceuticals, Inc., Cambridge, MA<br />

Background: MLN9708 is a potent investigational proteasome inhibitor,<br />

which upon intravenous (IV) administration immediately hydrolyzes to the<br />

active form MLN2238. MLN9708 is currently being evaluated in a phase 1<br />

trial in solid tumors (NCT00830869). This trial has a dose-escalation arm<br />

and five expansion cohorts: non-small cell lung cancer (NSCLC), s<strong>of</strong>t tissue<br />

sarcoma, head and neck cancer, prostate cancer, and a tumor biopsy cohort<br />

<strong>of</strong> mixed histology. The purpose <strong>of</strong> the tumor biopsy cohort was to obtain<br />

pre- and post-dose biopsies to determine drug distribution and target<br />

engagement in post-dose tumor samples. The latter was measured by the<br />

increase in levels <strong>of</strong> ATF-3, a marker <strong>of</strong> unfolded protein response/<br />

endoplasmic reticulum stress, which is upregulated in response to proteasome<br />

inhibition. Methods: The tumor biopsy cohort included 20 patients<br />

dosed at the maximum tolerated dose who consented to core needle<br />

biopsies during screening and after either the first or second dose <strong>of</strong><br />

MLN9708 (IV 1.76 mg/m2 ; 4–20 hours post-dose). Tumor biopsies were<br />

individually weighed, homogenized, and analyzed for the presence <strong>of</strong><br />

MLN2238 using a quantified LC/MS/MS methodology. ATF-3 levels in<br />

tumors were determined by an immunohistochemical assay (IHC) on six<br />

sections for each tumor biopsy. Tumor area was identified using Aperio<br />

Genie, a machine learning program for pattern recognition, and the<br />

percentage <strong>of</strong> ATF-3 positive area in the tumor was measured. Results:<br />

Biopsies from 20 patients were collected for assessment <strong>of</strong> drug distribution<br />

and target engagement. Ten patients with paired pre- and post-dose<br />

biopsies <strong>of</strong> sufficient size were considered evaluable for PK analysis;<br />

MLN2238 was present in all 10 (100%) post-dose biopsies analyzed.<br />

Tumor pairs from 7 patients passed quality control by H&E staining for<br />

tumor content and were evaluable for ATF-3 IHC. Six <strong>of</strong> 7 paired samples<br />

(86%) showed a statistically significant (p�0.05) increase in post-dose<br />

ATF-3 levels. Conclusions: Overall, emerging data from MLN9708 phase 1<br />

solid tumor analysis show that MLN2238 is present in tumors and<br />

demonstrates target engagement upon inhibition <strong>of</strong> the proteasome in<br />

tumor tissue biopsies.<br />

3079 General Poster Session (Board #17E), Mon, 8:00 AM-12:00 PM<br />

Phase I dose-finding study <strong>of</strong> golvatinib (E7050), a c-Met and Eph receptor<br />

targeted multi-kinase inhibitor, administered orally BID to patients with<br />

advanced solid tumors. Presenting Author: Toshihiko Doi, National Cancer<br />

Center Hospital East, Kashiwa, Japan<br />

Background: Golvatinib is a highly potent, small molecule ATP-competitive<br />

inhibitor <strong>of</strong> the c-Met receptor tyrosine kinase and multiple members <strong>of</strong> the<br />

Eph receptor family as well as c-Kit and Ron, based on isolated kinase<br />

assays. Golvatinib showed antiproliferative activity in human cancer<br />

xenograft models, warranting further investigation in the clinical setting.<br />

Methods: Japanese patients (pts) with advanced solid tumors that had<br />

progressed after approved therapy received oral golvatinib at escalating<br />

doses <strong>of</strong> 50 to 100, 200 and 300 mg administered twice daily (BID) in<br />

conventional 3 pt cohorts. The primary objective was to determine the MTD<br />

<strong>of</strong> golvatinib when administered continually BID. The secondary objectives<br />

were to evaluate the safety, PK, PD biomarkers, and efficacy. Results: 16pts<br />

were enrolled (8 M, 8 F; med age 59 years; ECOG 0-1). Tumor types were<br />

colorectal (5), gastric (4) and others (7). One DLT (Gr 3 ALT increase) and 1<br />

equivalent DLT (Gr 2 vomiting, nausea and anorexia) appeared in 2 pts in<br />

the 300 mg BID cohort and the dose escalation was terminated. The MTD<br />

was determined to be 200 mg BID. Frequently observed adverse drug<br />

reactions were proteinuria [50.0% (8/16)], ALT increased [43.8% (7/16)],<br />

AST increased, nausea, vomiting [37.5% (6/16)], and blood alkaline<br />

phosphatase increased, decreased appetite, diarrhea, leukopenia [31.3%<br />

(5/16)], which were all mild to moderate in severity with � Gr 2. No related<br />

severe adverse event was observed. Responses included 4 stable diseases<br />

(84-113 days) in 3 pts with gastric cancer and 1 patient with NSCLC, 11<br />

progressive diseases, and 1 non evaluable. The golvatinib plasma concentration<br />

after first-dose reached Cmax at a median time <strong>of</strong> 2 to 4 hours, and<br />

declined bi-exponentially with a t½ <strong>of</strong> approximately 40 hours. Cmax and<br />

AUC0-inf increased with dose. Analysis <strong>of</strong> blood PD biomarker showed a<br />

significant increase in soluble c-Met levels after golvatinib treatment.<br />

Conclusions: The MTD <strong>of</strong> golvatinib was achieved at 200 mg BID. Golvatinib<br />

was well tolerated with manageable toxicities at dose levels up to and<br />

including the MTD. 4 <strong>of</strong> 16 pts showed stable disease after golvatinib<br />

treatment.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


3080 General Poster Session (Board #17F), Mon, 8:00 AM-12:00 PM<br />

A phase I study <strong>of</strong> ombrabulin (O) combined with bevacizumab (B) in<br />

patients with advanced solid tumors (NCT01193595). Presenting Author:<br />

Gianluca Del Conte, Fondazione IRCCS Istituto Nazionale dei Tumori,<br />

Milan, Italy<br />

Background: O is a vascular-disrupting agent derived from combretastatin<br />

A4-phosphate that induces rapid but transient tumor vascular shutdown. In<br />

experimental models, the combination <strong>of</strong> O with VEGF-blockade induced<br />

more regressions than O alone. This phase I study was performed to<br />

determine the maximum tolerated dose (MTD), and assess the overall<br />

safety, pharmacokinetics (PK), pharmacodynamics (PD) and preliminary<br />

antitumor activity <strong>of</strong> O plus B. Trial is funded by san<strong>of</strong>i. Methods: Patients<br />

(pt) with advanced solid tumors, ECOG PS �1, and adequate organ<br />

function were eligible. Increasing doses <strong>of</strong> O (mg/m²) was administered<br />

intravenously (IV) on day (d) 1; B (5 or 10mg/kg) was administered IV on<br />

d2, in 21d cycles (C). A Bayesian design was applied to determine dose<br />

escalation steps and MTD. PK sampling was performed in C1 and C2. PD<br />

sampling for circulating endothelial cells (CEC) and progenitors (CEP) were<br />

performed throughout. Dynamic contrast-enhanced ultrasound (DCE-US)<br />

to assess tumor perfusion was performed before and after C1 and C2<br />

dosing. Results: 25 pts (M:F 6:19; median age 49 years [range 27-75])<br />

have been treated at 8 dose levels <strong>of</strong> O/B: 8/5 (3), 11.5/5 (4), 11.5/10 (3),<br />

15.5/10 (3), 20/10 (3), 25/10 (3); 35/10 (3) and 42/10 (3). Ovarian<br />

cancer was the most frequent tumor type (n�11). Median Cs received was<br />

3 (1 to 14). No DLTs were observed during C1. Drug-related grade (g) 3-4<br />

adverse events (AE) included hypertension in 2 pts (8/5; 11.5/5) and ileal<br />

perforation in 1 pt with peritoneal carcinomatosis at laparotomy (11.5/10).<br />

A case <strong>of</strong> duodenal perforation was unlikely related (25/10). Two pts with<br />

ovarian cancer (11.5/5; 20/10) had confirmed partial responses (4.4<br />

months (mo) and 7� mo). 13/23 (57%) pts with � 1 tumor assessment<br />

had stable disease (n�8; median 4.3 mo [1.8-9.5]; 5 ongoing). Data from<br />

cohorts 1 and 2 demonstrated no evidence <strong>of</strong> a PK interaction.17/25<br />

patients demonstrated a peak in CEC, and 7/9 patients a peak in CEP. 4/7<br />

DCE-US evaluable pts had �50% drop in tumor perfusion (AUC) at Day 8<br />

<strong>of</strong> C1. Conclusions: O combined with B is well tolerated with early evidence<br />

<strong>of</strong> clinical activity. The MTD has not been reached (current dose level<br />

50/10); cohorts <strong>of</strong> O combined with B at 15 mg/kg will be tested.<br />

3082 General Poster Session (Board #17H), Mon, 8:00 AM-12:00 PM<br />

A phase I study <strong>of</strong> R04929097, an oral gamma secretase inhibitor, in<br />

combination with gemcitabine in patients with advanced solid tumors<br />

(PHL-078/CTEP 8575). Presenting Author: Suzanne Richter, Princess<br />

Margaret Hospital, Toronto, ON, Canada<br />

Background: RO4929097 (RO) is an oral inhibitor <strong>of</strong> �-secretase that<br />

disrupts Notch signaling. Gemcitabine (GEM) is active against many solid<br />

tumors with a favorable toxicity pr<strong>of</strong>ile suited to combination. The primary<br />

objective <strong>of</strong> this trial is to establish the recommended phase II dose (RP2D)<br />

<strong>of</strong> RO in combination with GEM; secondary objectives include the evaluation<br />

<strong>of</strong> safety, tolerability, pharmacokinetics (PK), biomarkers <strong>of</strong> Notch<br />

signaling and preliminary anti-tumor activity. Methods: Patients (pts) with<br />

advanced solid tumors were enrolled in escalating RO dose levels (DL) as<br />

follows: DL1 20mg, DL2 30mg; DL3 45mg and DL4 90mg using a 3�3<br />

design. Treatment with RO was administered once daily on days (d) 1-3,<br />

8-10, 15-17, 22-24 and GEM 1000mg/m2 on d1, 8, and 15 in 28d cycles<br />

(c). Dose limiting toxicities (DLTs) during c1 were defined as CTCAE v4<br />

grade (g) 3 non-hematological or g4 hematological toxicities, failure to start<br />

c2 within �14 days, or failure to receive �75% doses <strong>of</strong> RO or d8 GEM in<br />

c1. Serial plasma samples for RO PK were collected on d1 and 10. Results:<br />

As <strong>of</strong> January 2012, 15 pts (median age 55) have been treated. DLTs were:<br />

DL1 0/3, DL2 1/7 (g3 ALT), DL3 0/3, DL4 2/2 (g3 AST/ALT and failure to<br />

receive ³75% doses); all were reversible. One death (perforated viscus)<br />

related to disease progression was observed. Most common g1/2 toxicities<br />

were nausea (9), vomiting (6), fatigue (5), hypophosphatemia (5), transaminitis<br />

(3) hypomagnasemia (2) and maculopapular rash (2). G3 hypophosphatemia<br />

(1) was observed beyond C1. RO PKs demonstrated comparable<br />

exposures at 30mg and 45mg (Table). Best response (RECIST 1.1) was<br />

stable disease � 4 months in 3 pts (pancreas, tracheal, breast CA).<br />

Conclusions: RO and GEM can be safely combined. RO levels achieved<br />

exceeded the AUC0-24 for efficacy in preclinical models using daily dosing.<br />

The maximum tolerated dose was exceeded at 90mg RO. Dose expansion at<br />

45mg RO is ongoing to confirm the RP2D.<br />

DL n<br />

Cmax ng/ml<br />

(SD) D1<br />

Cmax ng/ml<br />

(SD) D10<br />

AUC 0-24<br />

(ng*hr/mlx10 4 )<br />

(SD) D1<br />

AUC 0-24<br />

(ng*hr/mlx10 4 )<br />

(SD) D10<br />

1 3 552 (100) 615 (382) 6.8 (1.0) 9.9 (7.2)<br />

2 7 833 (352) 1221 (315) 12.6 (4.8) 19.2 (5.9)<br />

3 3 986 (153) 1315 (332) 12.8 (1.2) 16.5 (8.2)<br />

Developmental Therapeutics—Experimental Therapeutics<br />

3081 General Poster Session (Board #17G), Mon, 8:00 AM-12:00 PM<br />

A phase I study to assess oral bioavailability <strong>of</strong> a novel oral s<strong>of</strong>t gelatin<br />

capsule formulation <strong>of</strong> rigosertib (ON 01910.Na) under fasted and fed<br />

conditions in patients with myelodysplastic syndromes. Presenting Author:<br />

Azra Raza, Columbia Presbyterian Medical Center, New York, NY<br />

Background: Rigosertib (ON 01910.Na) is a novel multikinase inhibitor,<br />

with selective cytotoxic effects on tumor cells without impact on normal<br />

cells. Rigosertib, administered as a 3-day continuous infusion, is now<br />

undergoing phase 3 evaluation in higher risk MDS patients refractory to<br />

hypomethylating agents. Here, we report the results <strong>of</strong> the effect <strong>of</strong> food on<br />

the absolute bioavailability <strong>of</strong> a novel rigosertib oral formulation (s<strong>of</strong>t<br />

gelatin capsule) in MDS patients. Methods: This was a single-dose,<br />

three-treatment, three-period sequential design for studying the effects <strong>of</strong><br />

food on the bioavailability <strong>of</strong> an immediate-release s<strong>of</strong>t gelatin capsule<br />

formulation. The following dosing groups were tested in 12 patients: IV<br />

Dose 800 mg/m2 over 24 hours and oral dose 560 mg (2 x 280 mg<br />

capsules) under fasting and fed conditions. The oral dose was the<br />

recommended phase 2 dose, as reported previously (R.S. Komrokji et al.,<br />

Oral Formulation <strong>of</strong> Rigosertib (ON 01910.Na) in Patients with Myelodysplastic<br />

Syndrome (MDS) – Phase I Study Results. Blood 2011, 118:<br />

Abstract #3797). Plasma samples were collected pre-dose, and over 32<br />

hours (IV dose) or 8 hours (oral dose) after dose initiation. Rigosertib<br />

plasma levels were analyzed by a validated LC/MS/MS method. Pharmacokinetic<br />

parameters were estimated by noncompartmental analysis (WinNonlinÒ).<br />

Results: Rigosertib pharmacokinetic parameters are presented in the<br />

table below. The results <strong>of</strong> the present study demonstrate good oral<br />

bioavailability under fasting condition.Oral administration <strong>of</strong> rigosertib<br />

after a meal decreased Cmax and AUC by 77% and 61%, respectively,<br />

compared to fasting conditions. Conclusions: The results <strong>of</strong> this study<br />

support the potential for oral delivery <strong>of</strong> rigosertib, which could become a<br />

preferred therapy over a 3-day continuous intravenous infusion.<br />

Parameter<br />

800 mg/m 2 IV<br />

(24-hr infusion)<br />

Dosing group<br />

560 mg oral<br />

(fasting)<br />

193s<br />

560 mg oral<br />

(fed)<br />

C max (�g/ml) 3.14 � 1.13 2.42 � 1.26 0.56 � 0.31<br />

AUC (�g-hr/ml) 52.7 � 19.2 6.89 � 3.98 2.71 � 1.51<br />

T max (hr) 2.91 � 1.30 1.00 � 0.45 2.82 � 1.15<br />

t 1/2 (hr) 3.25 � 0.97 2.79 � 1.23 2.61 � 0.93<br />

Bioavailability (%) N/A 34.9 � 17.6 13.8 � 6.04<br />

3083 General Poster Session (Board #18A), Mon, 8:00 AM-12:00 PM<br />

A phase I dose-escalation study <strong>of</strong> the HSP90 inhibitor AUY922 and<br />

erlotinib for patients with EGFR-mutant lung cancer with acquired resistance<br />

(AR) to EGFR tyrosine kinase inhibitors (EGFR TKIs). Presenting<br />

Author: Melissa Lynne Johnson, Feinberg School <strong>of</strong> Medicine, Northwestern<br />

University, The Robert H. Lurie Comprehensive Cancer Center, Chicago,<br />

IL<br />

Background: AUY922 is a highly potent, non-geldanamycin analog, HSP90<br />

inhibitor that degrades mutated EGFR and MET. Preclinical studies<br />

demonstrate that HSP90 inhibitors have anti-tumor activity in EGFR<br />

mutant, EGFR TKI-sensitive and TKI-resistant lung cancer xenograft<br />

models. To prevent disease flare after stopping EGFR TKIs in patients (pts)<br />

with EGFR mutated lung cancer, erlotinib (E) is <strong>of</strong>ten continued with<br />

subsequent lines <strong>of</strong> treatment for AR. This phase I study will determine the<br />

maximally tolerated dose (MTD) <strong>of</strong> AUY922 and E for pts with mutated<br />

EGFR and RECIST progression on EGFR TKIs. Methods: All pts have EGFR<br />

mutant lung cancer, with development <strong>of</strong> AR (per Jackman, JCO 2010)<br />

after treatment with an EGFR TKI. Pts underwent repeat tumor biopsies<br />

after development <strong>of</strong> AR and prior to study entry. Pts receive AUY922 IV<br />

weekly and E oral daily in 28-day cycles, with dose escalation using<br />

standard 3�3 design. Pharmacokinetics, serial ophthalmology evaluations<br />

and weekly assessment for toxicity are required. Tumor tissue from<br />

re-biopsy at study entry is analyzed for EGFR T790M and MET. Results:<br />

Since April 2011, 11 pts have been enrolled, in 4 <strong>of</strong> 5 planned cohorts<br />

(AUY922 25 mg/m2 � E 75 mg; AUY922 25 mg/m2� E 150 mg; AUY922<br />

37.5 mg/m2� E 150 mg; AUY922 55 mg/m2�E 150 mg). Prior to<br />

developing AR, pts were treated with EGFR TKIs for a median <strong>of</strong> 9 months<br />

(range 8-32). Of 9 pts with tissue analyzed thus far, 6 had EGFR T790M.<br />

Pts have received a median <strong>of</strong> two cycles (range 1-7), and 5 remain on<br />

study. There have been no dose-limiting toxicities. Adverse events reported<br />

in � 20% <strong>of</strong> pts were diarrhea, nausea, vomiting, fatigue, and rash, all<br />

graded 1 or 2. No cardiovascular, renal or hepatic toxicities have been<br />

observed. Three pts have reported “flashing lights” and 2 pts transient<br />

night blindness, all grade 1. Dose escalation continues. No partial<br />

responses have been seen in 6 evaluable pts, 4 patients have confirmed<br />

stable disease at 8 wks. Conclusions: AUY922 and E is a well-tolerated<br />

combination at dose levels up to AUY922 55 mg/m2� E 150 mg. Further<br />

dose-escalation, MTD, and correlation with markers <strong>of</strong> AR will be reported.<br />

Supported by Novartis, Inc.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


194s Developmental Therapeutics—Experimental Therapeutics<br />

3084 General Poster Session (Board #18B), Mon, 8:00 AM-12:00 PM<br />

Phase I and pharmacokinetic study <strong>of</strong> farletuzumab in solid tumors.<br />

Presenting Author: Yasutsuna Sasaki, Saitama Medical University International<br />

Medical Center, Saitama, Japan<br />

Background: Farletuzumab (F) is a humanized monoclonal antibody against<br />

folate receptor � (FRA). Expression <strong>of</strong> FRA is reported to be 90% and 40%<br />

in ovarian cancer (OC) and gastric cancer (GC), respectively. The purpose <strong>of</strong><br />

the study is to assess tolerability, pharmacokinetic pr<strong>of</strong>ile and preliminary<br />

antitumor effect in solid tumors. Methods: Patients with OC or FRA<br />

expressing solid tumor who are resistant to standard treatments are eligible<br />

in the study. After pharmacokinetic (PK) run-in, F was administered by IV<br />

injection repeating every week until disease progression. PK pr<strong>of</strong>ile was<br />

analyzed on day1 to day 10 in PK run-in and on day 22 to 29 in cycle 1.<br />

Dose limiting toxicities (DLTs) were defined as G4 hematological and G3/4<br />

non-hematological toxicities by NCI-CTCAE. Dose escalation was planed in<br />

4 cohorts (50, 100, 200 and 400mg/m2 ). Results: Sixteen patients (14 OC<br />

and 2GC) received F infusion. Neither DLTs nor G3/4 toxicities were<br />

reported in all cohorts. As adverse events, G1/2 infusion reaction (44%),<br />

headache (44%), cytokine release syndrome (38%), nausea (31%) and<br />

appetite loss (31%) were observed and medically managed. AUC and<br />

Cmax.were increased dose-dependently and linear PK pr<strong>of</strong>iles were identified<br />

with median clearance between 6.02 and 10.54 mL/hr/m2 in each<br />

cohort. No tumor shrinkage was recorded, but long-term disease stabilization<br />

for 22� mos. and 10� mos. were observed in one patient with clear<br />

cell OC (100mg/m2 ) and one patient with GC (400 mg/m2 ), respectively.<br />

No cumulative toxicities were observed in these 2 patients. Conclusions:<br />

The toxicities <strong>of</strong> F in Japanese patients were manageable with similar PK<br />

pr<strong>of</strong>ile as compared with the US population (Konner et al. Clin Cancer Res<br />

2010: 16; 5288-95). Long term disease stabilization is observed in<br />

subpopulation <strong>of</strong> clear cell OC and GC.<br />

3086 General Poster Session (Board #18D), Mon, 8:00 AM-12:00 PM<br />

Associating retinal drug exposure and retention with the ocular toxicity<br />

pr<strong>of</strong>iles <strong>of</strong> Hsp90 inhibitors. Presenting Author: Dan Zhou, Synta Pharmaceuticals<br />

Corporation, Lexington, MA<br />

Background: In clinical trials certain Hsp90 inhibitors, including AUY922,<br />

SNX-5422, and 17-DMAG have caused visual symptoms suggesting retinal<br />

dysfunction; others, including ganetespib and 17-AAG, have not. Previous<br />

animal toxicology experiments have suggested that retinal changes may be<br />

linked to photoreceptor degeneration or cell death. Here histopathologic<br />

changes and/or exposure pr<strong>of</strong>iles <strong>of</strong> Hsp90 inhibitors with or without<br />

clinical reports <strong>of</strong> ocular toxicity were evaluated to understand the observed<br />

differences in toxicity pr<strong>of</strong>ile between agents in this class. Methods: We<br />

reviewed visual symptoms among subjects administered ganetespib, a<br />

potent Hsp90 inhibitor currently in phase II trials; evaluated retinal<br />

morphology in rats and cyno monkeys given ganetespib; and compared<br />

TUNEL-positive photoreceptors and drug exposure pr<strong>of</strong>iles in the retina <strong>of</strong><br />

rats treated with AUY922, 17-DMAG, and 17-AAG. Studies <strong>of</strong> ganetespib’s<br />

retinal pharmacokinetics and photoreceptor toxicity in rats are ongoing.<br />

Results: AUY922 and 17-DMAG induce visual disorders in the clinic. Both<br />

drugs induced marked photoreceptor cell death (increased TUNEL-positive<br />

cells) in rats with a high retina/plasma (R/P) exposure ratio, and a slow<br />

elimination rate (at 6 h post-dose, over 54% <strong>of</strong> AUY922 present at 30 min<br />

remained in the retina). In contrast, and consistent with an absence <strong>of</strong><br />

clinical visual changes, 17-AAG at the maximum tolerated dose did not<br />

elicit photoreceptor injury. Further, retinal elimination was rapid (at 6 h<br />

post-dose, 94% <strong>of</strong> 17-AAG had been eliminated from the retina, resulting<br />

in a low R/P ratio). Ganetespib given by 1-hour IV infusion on days 1 and 15<br />

<strong>of</strong> four 21-day cycles did not cause histopathological changes in the retina<br />

<strong>of</strong> rats or cynos. This finding is consistent with very low rate (~3%) <strong>of</strong><br />

drug-related visual symptoms observed among over 300 subjects to date<br />

that received ganetespib in the clinic. Conclusions: Unlike AUY922 and<br />

17-DMAG, ganetespib is not associated with ocular toxicity in humans,<br />

primates, or rodents. The findings suggest that Hsp90 inhibitors may elicit<br />

visual disorders when associated with high retina/plasma exposure ratio<br />

and lower retinal elimination rate.<br />

3085 General Poster Session (Board #18C), Mon, 8:00 AM-12:00 PM<br />

Phase IB study <strong>of</strong> an all-oral chemotherapy regimen, tesetaxel plus<br />

capecitabine, in patients with advanced solid tumors. Presenting Author:<br />

Michael Gary Martin, The West Clinic, Memphis, TN<br />

Background: Tesetaxel is a novel oral taxane that is not a substrate for<br />

P-glycoprotein, does not cause hypersensitivity, and may cause less<br />

neurotoxicity. The anticancer activity <strong>of</strong> tesetaxel exceeds other taxanes in<br />

various cell lines and xenografts. Phase II studies showed overall response<br />

rates [ORRs] <strong>of</strong> 50% and 38% in patients (pts) treated for 1st- and 2nd-line breast cancer, respectively, and 20% in pts with 2nd-line gastric cancer.<br />

Neutropenia was dose-limiting, and the maximally tolerated dose (MTD)<br />

was 27 mg/m2 once every 3 weeks. Capecitabine, an oral fluoropyrimidine,<br />

is commonly used for treatment <strong>of</strong> breast and gastric cancer. While the<br />

approved capecitabine dose is 2500 mg/m2 /day x 14 days, most pts are<br />

unable to sustain treatment at this dose due to hand-foot syndrome (HFS).<br />

A prior PK study suggested that short courses <strong>of</strong> tesetaxel could be<br />

combined with capecitabine at the recommended MTDs <strong>of</strong> both drugs with<br />

non-overlapping adverse events (AEs) (Cancer Chemother Pharmacol.<br />

68:1565, 2011). We conducted a phase IB study <strong>of</strong> the tesetaxel/<br />

capecitabine regimen in pts using lower doses <strong>of</strong> capecitabine to confirm a<br />

tolerable combination dose for extended treatment. Methods: Eligibility<br />

included: solid tumor treated with � 1 line <strong>of</strong> chemotherapy; no prior<br />

taxane exposure; adequate organ function; ECOG PS � 1. Pts received<br />

tesetaxel 27 mg/m2 on Day 1 and capecitabine 1500-2000 mg/m2 /day (in<br />

divided doses on Days 1-14) every 3 weeks. Results: To date, 10 pts have<br />

been treated. AEs were largely Grade 1-2 (83%) and consistent with those<br />

for each drug as a single agent. Grade 3-4 neutropenia occurred in 3 pts<br />

(30%) and HFS in 4 (40%). Tesetaxel dose reduction was required in 1 pt<br />

for neutropenia; capecitabine dose reduction was required in 3 pts (30%)<br />

(2 for hand-foot syndrome, 1 for diarrhea). Comprehensive toxicity data will<br />

be presented. Conclusions: This combination <strong>of</strong> all-oral chemotherapy is<br />

generally well-tolerated, although lower than maximally approved doses <strong>of</strong><br />

capecitabine are required, similar to other regimens that employ this agent.<br />

AEs are largely non-overlapping, which suggests this combination may be<br />

clinically useful in pts with advanced breast and gastric cancer.<br />

3087 General Poster Session (Board #18E), Mon, 8:00 AM-12:00 PM<br />

Phase l study assessing a two-consecutive-day (QD x 2) dosing schedule <strong>of</strong><br />

the HSP90 inhibitor, AT13387, in patients with advanced solid tumors.<br />

Presenting Author: Khanh Do, National Cancer Institute, Bethesda, MD<br />

Background: AT13387, a synthetic, non-ansamycin, small molecule inhibitor<br />

<strong>of</strong> HSP90 (Kd 0.71 nM), binds to the N-terminal ATP-binding site<br />

resulting in down-regulation <strong>of</strong> key oncoproteins (HER2 and ERK). In<br />

xenograft models AT13387 produced tumor growth inhibition on a QD x 2<br />

dosing schedule. Objectives: To determine toxicity, MTD, PK, and pharmacodynamic<br />

(PD) effect <strong>of</strong> AT13387 given IV QD x 2, 3 weeks out <strong>of</strong> 4.<br />

Methods: Adult patients (pts) with advanced cancers progressing following<br />

standard therapy; ECOG � 2; adequate organ function. One pt cohorts for<br />

first three dose levels (DLs, 20, 40, 80 mg/m2 ), planned 3�3 design at DL<br />

4 (120 mg/m2 ). PK evaluations on C1D1 and C1D15. Mandatory tumor<br />

biopsies in expansion cohort at MTD for HER2� tumors (baseline, C1D17)<br />

to evaluate HSP70 and HSP27 mRNA by RT-PCR; and client protein by<br />

IHC and immunoassay. HSP70 in PBMCs as a PD marker (pre-dose, D2,<br />

D15, D16) assessed by Western blot; serum markers <strong>of</strong> cell death, M30<br />

and M65 (baseline and pre-dose C1D16), assessed by ELISA. Results: 12<br />

pts treated up to DL 5 (160 mg/m2). Median age, 59; median # prior<br />

therapies, 3; diagnosis (# pts): colorectal (7); esophageal (2), liposarcoma<br />

(1); head and neck (1); synovial sarcoma (1). Drug-related toxicities (gr �<br />

2; # pts) in 9 evaluable pts: fatigue (5), rash (2), nausea (5), diarrhea (7),<br />

QTc prolongation (2), thrombocytopenia (5), AST (3) and ALT (2). Vision<br />

disorder (2 pts): gr 1, reversible, no objective findings on eye exams. DLT (1<br />

pt, with liver metastases): DL 5, gr 3 AST/ALT. PK was dose proportional;<br />

T1/2 8 hrs. In one paired tumor biopsy (DL 3) an increase in HSP70 (� 1.5<br />

fold) and HSP27 (2 fold) mRNA and a decrease in pERK was observed.<br />

M30 and M65 increased in 5/6 pts (p-value n.s.). Induction <strong>of</strong> HSP70<br />

(1.48 - 5.01 fold) in PBMCs was noted at all time points following first dose<br />

(� DL 2). 3/9 pts had SD after 2 cycles. Conclusions: Preliminary data<br />

indicate that AT13387 QD x2iswell tolerated and produces a sustained<br />

induction <strong>of</strong> HSP70 in PBMCs. Accrual is ongoing to establish the MTD and<br />

obtain further tumor biopsies for assessment <strong>of</strong> drug effect. Optional<br />

Indium-labeled trastuzumab scan will be used to evaluate drug effect on<br />

HER2 levels in pts with HER2 expressing tumors.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


3088^ General Poster Session (Board #18F), Mon, 8:00 AM-12:00 PM<br />

A phase I study <strong>of</strong> dovitinib (TKI258) in Japanese patients with advanced<br />

solid tumors. Presenting Author: Hiroya Takiuchi, Osaka Medical College,<br />

Osaka, Japan<br />

Background: Dovitinib is a tyrosine kinase inhibitor with demonstrated<br />

inhibitory activity against FGFRs, VEGFRs, and PDGFRs in vivo. Based on<br />

responses observed in renal cell carcinoma, breast cancer, AML, melanoma,<br />

and multiple myeloma in clinical studies in the West, we investigated<br />

dovitinib in Japanese patients (pts). Methods: This multicenter phase<br />

I study determined the maximum tolerated dose (MTD) <strong>of</strong> dovitinib based<br />

on the occurrence <strong>of</strong> dose-limiting toxicity (DLT) in Japanese pts with<br />

advanced solid tumors. Following a 2-day pharmacokinetic (PK) run-in<br />

period, dovitinib was administered orally once daily on a 5-days-on/2-days<strong>of</strong>f<br />

schedule in 28-day cycles until disease progression or withdrawal. The<br />

planned dose range was 100-600 mg/day. A 2-parameter Bayesian logistic<br />

regression model based on the principle <strong>of</strong> escalation with overdose control<br />

was used to estimate the MTD. Results: In total, 28 pts received dovitinib:<br />

100 mg (n � 3), 200 mg (n � 3), 300 mg (n � 7), 400 mg (n � 9), and<br />

500 mg (n � 6). The median age was 58.5 years (range, 30-76); 16 <strong>of</strong> 28<br />

pts (57%) were male. All pts had stage IV disease, with an ECOG<br />

performance status <strong>of</strong> 0 or 1. Pts completed a median <strong>of</strong> 3 cycles. One pt is<br />

currently ongoing in the study (peritoneal adenocarcinoma, 400-mg cohort,<br />

cycle 19), 23 discontinued due to disease progression, and 4 discontinued<br />

due to adverse events (AEs). All DLTs were grade 3: anorexia (n � 1; 300<br />

mg), nausea/vomiting (n � 1; 400 mg), liver function disorder (n � 1; 400<br />

mg), and increased alanine transaminase (n � 1; 500 mg). The most<br />

common grade 3/4 AEs (occurring in �10% <strong>of</strong> pts) suspected to be related<br />

to study drug were lymphopenia (18%), neutropenia (14%), abnormal<br />

hepatic function (14%), decreased white blood cell count (14%), decreased<br />

appetite (14%), and hypertension (14%). Best responses were<br />

confirmed partial response in 1 pt (4%; peritoneal adenocarcinoma,<br />

400-mg cohort), stable disease in 9 pts (32%), and progressive disease in<br />

10 pts (36%). No treatment-related deaths have been reported. Safety and<br />

PK parameters were comparable to those <strong>of</strong> non-Japanese pts in the global<br />

study. Conclusions: The study has completed enrollment. Dovitinib was<br />

found to be tolerable at doses up to 500 mg, which was declared as the<br />

MTD in Japanese pts.<br />

3090 General Poster Session (Board #18H), Mon, 8:00 AM-12:00 PM<br />

Preclinical activity <strong>of</strong> the Hsp90 inhibitor, ganetespib, in ALK- and<br />

ROS1-driven cancers. Presenting Author: David A. Proia, Synta Pharmaceuticals<br />

Corporation, Lexington, MA<br />

Background: Ganetespib is a potent inhibitor <strong>of</strong> heat shock protein 90<br />

(Hsp90) currently being studied in a broad range <strong>of</strong> clinical trials. Phase II<br />

results with ganetespib demonstrated an encouraging objective response<br />

rate <strong>of</strong> 50% in non-small cell lung carcinoma (NSCLC) patients whose<br />

tumors contained ALK translocations and had progressed on previous<br />

treatments. To further understand the clinical potential <strong>of</strong> ganetespib in<br />

ALK-driven cancers, we evaluated its activity in (1) crizotinib-sensitive and<br />

-resistant cancer cells harboring ALK fusions; (2) cells expressing amplified<br />

ALK or ROS1 translocations (a kinase structurally similar to ALK); and (3)<br />

in combination with crizotinib. Methods: H3122 NSCLC cells, which<br />

express EML4-ALK, were treated with ganetespib, crizotinib or the combination,<br />

and cell viability and signaling cascades were assessed. Similar<br />

experiments were done in cells with amplified, constitutively active ALK<br />

(NB-39-nu) or ROS1 kinase fusions (HCC-78, U118MG). To generate a<br />

model <strong>of</strong> crizotinib resistance, NPM-ALK-expressing BaF3 cells were<br />

exposed to various crizotinib concentrations. Fifteen different ALK kinase<br />

domain substitutions were identified; clonal NPM-ALK/BaF3 cells were<br />

made for each resistance mutation and assayed for sensitivity to ganetespib.<br />

Results: Ganetespib was 50-fold more potent than crizotinib in killing<br />

H3122 cells, and when combined together at sub-lethal doses, displayed<br />

strong synergistic anticancer activity. Ganetespib showed similar potency<br />

in other cells driven by constitutively active ALK or ROS1 kinase fusions,<br />

due to abrogation <strong>of</strong> their oncogenic kinase activity. All 15 <strong>of</strong> the<br />

crizotinib-resistant NPM-ALK/BaF3 mutant clones were highly sensitive to<br />

ganetespib (IC50 values ranging from 14-23 nM), including the 7<br />

mutations reported to date in patients with ALK-driven tumors. Conclusions:<br />

Ganetespib effectively inhibits the activity <strong>of</strong> ALK and ROS1, kinases<br />

associated with several tumor types, resulting in marked single agent<br />

anticancer activity in cells driven by them. Importantly, ganetespib retains<br />

its potency irrespective <strong>of</strong> the mutational status <strong>of</strong> ALK. The strong synergy<br />

observed between ganetespib and crizotinib warrants further study.<br />

Developmental Therapeutics—Experimental Therapeutics<br />

195s<br />

3089^ General Poster Session (Board #18G), Mon, 8:00 AM-12:00 PM<br />

Endoxifen for breast cancer: Multiple-dose, dose-escalation study characterizing<br />

pharmacokinetics and safety in metastatic breast cancer patients.<br />

Presenting Author: Ateeq Ahmad, Jina Pharmaceuticals, Inc., Libertyville,<br />

IL<br />

Background: Endoxifen is an active metabolite <strong>of</strong> tamoxifen, a drug used in<br />

the treatment <strong>of</strong> breast cancer. To be clinically effective, tamoxifen must<br />

be converted to endoxifen by CYP2D6. Direct administration <strong>of</strong> endoxifen<br />

would not be subject to pharmacogenetic variations or drug-drug interactions.<br />

Our preclinical studies (Breast Cancer Treat 122, 579-584, 2010)<br />

have validated the concept <strong>of</strong> using endoxifen for the treatment <strong>of</strong> breast<br />

cancer. In human (Clin. Pharmacol. Ther. 88, 814-817, 2010), the single<br />

oral doses tested up to 4 mg <strong>of</strong> endoxifen were safe, well tolerated and<br />

bioavailable. Methods: A multiple-dose escalating study was conducted in 3<br />

cohorts and each cohort had 6 patients (18 metastatic breast cancer<br />

patients). Endoxifen at 3 dose levels (2, 4, or 8 mg) was given once daily for<br />

28 days. Routine laboratory tests, vital signs and electrocardiograms were<br />

measured throughout the study. Blood samples for PK analysis were<br />

collected after 28 days post dose. Endoxifen in plasma samples was<br />

determined using LC-MS/MS. Results: Endoxifen was found to be safe up to<br />

8.0 mg. At steady state, it displays dose-proportional PK with respect to<br />

Cmax and AUC ( see Table below). Conclusions: Multiple daily endoxifen<br />

doses <strong>of</strong> 4.0-8.0 mg resulted in endoxifen exposures that would be<br />

sufficient for effective therapy. The favorable safety and multiple-dose PK<br />

pr<strong>of</strong>ile <strong>of</strong> endoxifen warrants further evaluation <strong>of</strong> safety and efficacy <strong>of</strong><br />

endoxifen in breast cancer patients.<br />

Pharmacokinetics parameters <strong>of</strong> endoxifen in metastatic breast cancer<br />

patients (n�5) after multiple-dose <strong>of</strong> 2, 4 or 8 mg <strong>of</strong> endoxifen tablets.<br />

Parameters (units) 2mg<br />

Mean � SD<br />

4mg 8mg<br />

Tmax (h)*<br />

Cmin ss (ng/mL)<br />

Cmax ss (ng/mL)<br />

AUCtau (ng.h/mL)<br />

PTF (%)<br />

5 (3.0-12.0)<br />

15.7�6.4<br />

24.5 �7.3<br />

445.3 �146.2<br />

50.8�19.0<br />

5 (4.0-6.0)<br />

44.0�11.6<br />

75.9 �18.5<br />

1363.3�396.3<br />

56.5 �17.9<br />

5 (4.0-6.0)<br />

80.4�26.7<br />

134.1�32.1<br />

2322.6�619.8<br />

57.6 �14.6<br />

Cav ss (ng/mL) 18.6 �6.1 56.8 �16.5 96.8�25.8<br />

*Data presented as median (range)<br />

3091 General Poster Session (Board #19A), Mon, 8:00 AM-12:00 PM<br />

Therapy <strong>of</strong> human solid tumor xenografts with CD74-targeted milatuzumab-<br />

SN-38 immunoconjugates. Presenting Author: David M. Goldenberg,<br />

Center for Molecular Medicine and Immunology, Morris Plains, NJ<br />

Background: CD74 is commonly considered to be an antigen present in<br />

hematopoietic cancers, but it is also expressed in a number <strong>of</strong> solid tumors.<br />

The humanized anti-CD74 antibody, milatuzumab (hLL1), was previously<br />

shown to have exquisite internalization properties, making it an attractive<br />

carrier for drug delivery. In this study, we evaluated hLL1 conjugates <strong>of</strong> a<br />

potent topo I inhibitor, SN-38, for treating solid tumors. Methods: Two<br />

hLL1-SN-38 conjugates, possessing either a pH-sensitive linker (‘CL2A’<br />

form) or a cathepsin-B-cleavable linker (�CL2E� form), were examined. In<br />

vitro analyses were carried out in the A-375 human melanoma cell line, and<br />

therapy experiments were performed in female athymic nude mice bearing<br />

established s.c. A-375 or s.c. human pancreatic adenocarcinoma (Capan-1)<br />

xenografts using specified doses (i.p.) at a twice weekly � 4 wks<br />

schedule; animals were sacrificed when tumor volumes (TVs) reached 1<br />

cm3 . Results: Both A-375 and Capan-1 cell lines tested positive for CD74<br />

expression by IHC. In vitro, both SN-38 derivatives liberated free drug at<br />

the same rate when exposed to cathepsin-B at pH 5, while serum stabilities<br />

for CL2A and CL2E forms <strong>of</strong> the conjugates were ~1dand�10 d,<br />

respectively. In the A-375 melanoma cell line, IC50 for the hLL1-SN-38<br />

conjugates <strong>of</strong> CL2A and CL2E forms were 5 nM and 34 nM, respectively. In<br />

the aggressive A-375 s.c. model <strong>of</strong> melanoma, in nude mice (n �8; TV:<br />

0.23 � 0.06 cm3 ) treated with 12.5 mg/kg protein dose (total 1.7 mg/kg<br />

SN-38 eq.) <strong>of</strong> specific or non-specific SN-38 conjugates <strong>of</strong> CL2A and CL2E<br />

forms, only hLL1-CL2A-SN-38 conjugate was efficacious (Log-rank:<br />

P�0.009 vs. all controls), with a median survival time <strong>of</strong> 28 d vs. 10.5 d for<br />

untreated. Likewise, in mice bearing Capan-1 xenografts (n � 8-10;<br />

TV�0.27 � 0.05 cm3 ), two low doses <strong>of</strong> hLL1-CL2A-SN-38 (12.5 or 5<br />

mg/kg) significantly improved survival in comparison to saline control mice<br />

(p�0.035), whereas the CL2E form and a non-specific control demonstrated<br />

no efficacy. Conclusions: These results indicate the therapeutic<br />

potential <strong>of</strong> hLL1-CL2A-SN-38 conjugate and the importance <strong>of</strong> linker<br />

chemistry, and encourage additional testing in other CD74-positive solid<br />

cancers.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


196s Developmental Therapeutics—Experimental Therapeutics<br />

3092 General Poster Session (Board #19B), Mon, 8:00 AM-12:00 PM<br />

BI 836845, a fully human IGF ligand neutralizing antibody, to improve the<br />

efficacy <strong>of</strong> rapamycin by blocking rapamycin-induced AKT activation.<br />

Presenting Author: Paul J. Adam, Boehringer Ingelheim RCV GmbH & Co.<br />

KG, Vienna, Austria<br />

Background: Analogs <strong>of</strong> rapamycin (rapalogs) targeting mammalian target<br />

<strong>of</strong> rapamycin complex 1 (mTORC1) have shown clinical activity in several<br />

cancers. Nonetheless, preclinical and clinical data suggest that there may<br />

be intrinsic resistance to rapalogs through a feedback loop which activates<br />

upstream signaling when mTORC1 is blocked. BI 836845 is a fully human<br />

antibody, currently in phase I clinical trials, which potently neutralizes both<br />

IGF-1 and IGF-2. We tested whether BI 836845 is able to improve the<br />

efficacy <strong>of</strong> rapamycin by inhibiting upstream signaling in preclinical<br />

models. Methods: Cancer cell lines were pr<strong>of</strong>iled in vitro and in vivo for<br />

sensitivity to BI 836845 and rapamycin, alone or in combination. Mitogenic<br />

signaling was examined by measuring levels <strong>of</strong> phosphorylated AKT<br />

(pAKT) using Western blot analysis. IGF bioactivity was determined using a<br />

cellular IGF-1R phosphorylation ELISA. Results: The combination <strong>of</strong> BI<br />

836845 and rapamycin was more effective than either agent alone at<br />

inhibiting the proliferation <strong>of</strong> Ewing’s sarcoma cells cultured in vitro as well<br />

as in a nude mouse xenograft model in vivo. Analysis <strong>of</strong> cell signaling<br />

upstream <strong>of</strong> mTOR demonstrated that treatment with rapamycin alone<br />

resulted in elevated pAKT, indicating feedback loop activation. BI 836845<br />

treatment alone or in combination with rapamycin inhibited AKT phosphorylation,<br />

demonstrating that the rapamycin-induced increase in pAKT was<br />

due to elevated IGF bioactivity. Consistent with this we demonstrated that<br />

rapamycin increased IGF bioactivity in mice and that this could be<br />

inhibited by BI 836845. We extended these studies to include other cancer<br />

cell lines and pr<strong>of</strong>iled the correlation between improved efficacy <strong>of</strong> the<br />

combination with BI 836845 inhibition <strong>of</strong> rapamycin-induced feedback. A<br />

correlation has been observed for cancer cells derived from several<br />

indications. Conclusions: Rapamycin treatment increases AKT activation<br />

via elevated IGF ligand bioactivity. This effect can be inhibited by BI<br />

836845, thus explaining the improved pre-clinical efficacy seen when both<br />

agents are combined. These data provide a rationale for the clinical<br />

combination <strong>of</strong> rapalogs and BI 836845.<br />

3094 General Poster Session (Board #19D), Mon, 8:00 AM-12:00 PM<br />

A phase I and pharmacokinetic study <strong>of</strong> multiple schedules <strong>of</strong> ganetespib<br />

(STA-9090), a heat shock protein 90 inhibitor, in combination with<br />

docetaxel for subjects with advanced solid tumor malignancies. Presenting<br />

Author: John S. Kauh, Winship Cancer Institute, Emory University, Atlanta,<br />

GA<br />

Background: Ganetespib is a next-generation Hsp90 inhibitor unrelated to<br />

the first-generation ansamycin class <strong>of</strong> Hsp90 inhibitors and has superior<br />

activity to these agents in preclinical studies. Ganetespib is well tolerated<br />

with promising antitumor activity. Based on synergy between ganetespib<br />

(G) and docetaxel (D), a phase I study evaluating preclinical dosing models<br />

was performed. Methods: Patients (pts) with advanced solid tumor malignancies<br />

and ECOG performance status (PS) 0-2 were eligible. Sequential<br />

cohorts <strong>of</strong> pts were treated (3�3 design) with increasing doses <strong>of</strong> D (day 1)<br />

and G (days 1, 8) (A) Q 21 days. Following MTD determination, safety and<br />

exploratory cohorts <strong>of</strong> D day 1 with G days 1, 15 (B); or G days 1, 4, 15 (C)<br />

were completed. PK sampling was performed during schedule A. The<br />

primary endpoint was determination <strong>of</strong> optimal doses and schedule <strong>of</strong> the<br />

two agents for combination therapy. Results: Twenty-seven patients were<br />

enrolled in schedules A (n�12), B (n�8), and C (n�7). Median age-64<br />

(44-75); 11-M, 16-F; ECOG PS: 0 (n�5),1(n�21), 2 (n�1). Most pts had<br />

NSCLC (n�9), others were head/neck (n�4), and SCLC (n�3). The defined<br />

MTD was level 2 (D-75 mg/m2, G-150 mg/m2), as 2 <strong>of</strong> 4 pts at level 3<br />

(D-75 mg/m2, G-200 mg/m2) had DLTs (febrile neutropenia and one g4<br />

neutropenia <strong>of</strong> � 7 days), requiring expansion <strong>of</strong> level 2. The median<br />

number <strong>of</strong> cycles received is 2 (1-11), with 3 pts in schedule C still on<br />

study. Among 22 pts evaluable for response, 1 had a PR (head/neck), 12<br />

had SD following 6 weeks evaluation, 10 pts for 12 weeks and 6 pts for 18<br />

weeks. Common AEs included neutropenia, diarrhea, anemia, fatigue,<br />

nausea, and febrile neutropenia. Prophylactic filgrastim/pegfilgrastim was<br />

not used at any time. PK data indicates similar G exposure alone compared<br />

to G administered prior to D. No accumulation was observed following<br />

once-weekly dosing, consistent with studies <strong>of</strong> G alone. Conclusions: The<br />

combination is well tolerated at the recommended doses <strong>of</strong> D 75 mg/m2<br />

and G 150 mg/m2. Promising anti-cancer activity was noted, and a<br />

randomized phase 2b/3 study with D day 1 and G days 1, 15 regimen is<br />

ongoing in advanced NSCLC (NCT01348126).<br />

3093 General Poster Session (Board #19C), Mon, 8:00 AM-12:00 PM<br />

Phase I dose-finding and pharmacokinetic study <strong>of</strong> a combination <strong>of</strong><br />

elisidepsin (E) and erlotinib (T) in patients (pts) with advanced solid<br />

tumors. Presenting Author: Sanjay Goel, Montefiore Einstein Cancer<br />

Center, Bronx, NY<br />

Background: E is a new marine compound that has shown synergism with T<br />

in vitro even in T-resistant non-small-cell lung cancer (NSCLC) cell lines.<br />

Based on these findings, a phase I clinical trial was undertaken to identify<br />

the maximum tolerated dose (MTD) and recommended dose (RD) <strong>of</strong> E (3-h<br />

iv, days 1, 8 and 15) followed by T (once daily) in 3-week cycles. Secondary<br />

objectives were evaluation <strong>of</strong> safety and feasibility, PK and preliminary<br />

efficacy results. Methods: Patients (pts) with advanced solid tumors with no<br />

standard therapeutic option were recruited. Cohorts <strong>of</strong> 3-6 pts were<br />

included at each dose level (DL), with escalating doses <strong>of</strong> E in increments<br />

<strong>of</strong> 25-50% according to toxicities, and 2 different T doses (100 and 150<br />

mg/day). Results: Thirty pts (median age, 57 years; 19 females) were<br />

evaluable. Main tumor types included NSCLC, colorectal, melanoma, and<br />

ovarian cancer. Six DLs were assessed. Starting DL was E 0.33 mg � T<br />

100mg. One dose-limiting toxicity (DLT) out <strong>of</strong> 6 pts (grade 3 bilirubin<br />

increase) was observed at DL3 (E 0.75 mg � T 150 mg). Another DLT (dose<br />

omissions due to ALT increase) was found at DL6 (E 2.25 mg � T 100 mg).<br />

Frequent toxicities were diarrhea (53%), nausea (23%), vomiting (33%),<br />

rash (47%), pruritus (43%), dry skin (27%) and acneiform dermatitis<br />

(17%). Grade 3/4 toxicities included diarrhea (2 pts), rash (2 pts) and<br />

pruritus (1 pts). Main biochemical abnormalities were ALT (grade 3/4 in 4<br />

pts) and total bilirubin increase (grade 3 in 2 pts). No severe hematological<br />

abnormalities were observed. Most toxicities were related to T; therefore, T<br />

dose was reduced to 100 mg/day. No PK interaction between E and T was<br />

observed. No objective responses were reported. Six pts attained stable<br />

disease �3 months (3 NSCLC; 1 ovarian cancer, 1 colorectal cancer, 1<br />

invasive thymoma). Conclusions: E � T was a manageable combination;<br />

however, the difficulty <strong>of</strong> combining E with the standard dose <strong>of</strong> T (150mg)<br />

and the lack <strong>of</strong> activity made this combination unattractive for further<br />

development in the current schedule. Possibly, other schedules may<br />

demonstrate more efficacy.<br />

3095 General Poster Session (Board #19E), Mon, 8:00 AM-12:00 PM<br />

A phase I trial <strong>of</strong> the mTOR inhibitor temsirolimus (TEM) in combination<br />

with capecitabine (CAP) in patients with advanced malignancies. Presenting<br />

Author: Michael J. Pishvaian, Lombardi Comprehensive Cancer Center,<br />

Georgetown University, Washington, DC<br />

Background: The mammalian target <strong>of</strong> rapamycin (mTOR) signaling pathway<br />

is critical for cell growth and proliferation. mTOR antagonists, such as<br />

TEM, have proven anti-cancer efficacy. Pre-clinical models and early phase<br />

clinical trials have demonstrated increased efficacy when an mTOR<br />

antagonist is combined with cytotoxic chemotherapy. Methods: Patients<br />

(pts) with advanced malignancies and adequate hepatic and renal function<br />

were eligible for enrollment. An alternating dose escalation <strong>of</strong> TEM and CAP<br />

was employed in separate q2week and q3week arms. Planned dose levels<br />

were: TEM 15 and 25mg IV weekly; and CAP 500, 750, 1000, and 1250<br />

mg/m2 orally twice daily (BID). Restaging occurred every 4 cycles for the<br />

q2week arm, and every 3 cycles for the q3week arm. PK samples assessed<br />

serum levels <strong>of</strong> sirolimus at Day 0, 8, and 15 <strong>of</strong> the q2week arm. Archived<br />

tumor specimens were assayed by immunohistochemistry (IHC) for expression<br />

<strong>of</strong> phospho-AKT, phospo-4EBP1, phoshpo-p70S6, and PTEN. Results:<br />

Thirty-two pts were enrolled, 30 <strong>of</strong> whom were evaluable for toxicity, <strong>of</strong><br />

which 18 were male, age range 30-72 years, 25 with colorectal cancer. Pts<br />

had received an average <strong>of</strong> 4 prior lines <strong>of</strong> therapy. The most common<br />

adverse events (AEs) were mucositis and fatigue. The most common grade<br />

3/4 AEs were fatigue (n�4), diarrhea (n�2), and hypophosphatemia<br />

(n�2). In 16/30 pts, the dose <strong>of</strong> CAP was reduced. There were two DLTs,<br />

both hypophosphatemia in the q3week arm (TEM�25mg and<br />

CAP�1000mg/m2 ). For the q2week arm, the recommended phase II dose<br />

(RP2D) was TEM 25 mg � CAP 1000mg/m2 . For the q3week arm, the<br />

RP2D was TEM 25mg � CAP 750mg/m2 . Twenty-five pts were evaluable<br />

for response (18 for OS). There were no PRs or CRs, but 14/25 pts (56%)<br />

had SD, with 4/24 (16%) having prolonged SD <strong>of</strong> �6 months. Median TTP<br />

and OS were 3 and 7 months, respectively. Five pts are still on study. PK<br />

assessment <strong>of</strong> serum sirolimus levels, and the results <strong>of</strong> the IHC on<br />

archived tumor samples will be presented. Conclusions: The combination <strong>of</strong><br />

TEM and CAP is safe on both a q2week and a q3week schedule. The<br />

combination demonstrated promising evidence <strong>of</strong> disease control in this<br />

highly refractory population and should be tested in disease-specific phase<br />

II trials.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


3096 General Poster Session (Board #19F), Mon, 8:00 AM-12:00 PM<br />

Phase I study <strong>of</strong> sirolimus plus gemcitabine in patients with advanced solid<br />

tumors: A Spanish Group for Research on Sarcomas (GEIS) study. Presenting<br />

Author: Juan Martin Liberal, Instituto Catalan Oncologia, Barcelona,<br />

Spain<br />

Background: In preclinical studies, combination <strong>of</strong> sirolimus with gemcitabine<br />

enhances apoptosis in vitro and increases anti-tumor efficacy in vivo.<br />

Methods: Patients with advanced solid tumors, age 18-70 years, no prior<br />

mTOR inhibitor or gemcitabine, ECOG PS 0-1, and adequate hematological,<br />

renal and hepatic function, were enrolled in this phase I study to assess<br />

safety, tolerability, pharmacokinetics (PK), and to identify the dose limiting<br />

toxicity (DLT), maximum tolerated dose (MTD) and recommended dose<br />

(RD) <strong>of</strong> the combination <strong>of</strong> sirolimus and gemcitabine. A 3�3 dose<br />

escalation design with cohorts <strong>of</strong> 3-6 patients was used. Sirolimus was<br />

given po continuously. Gemcitabine was given iv 10mg/m2 /minute on days<br />

1 and 8 every 3 weeks. Dose levels 1, 2 and 3 corresponded to sirolimus 2,<br />

2 and 5mg/24h plus gemcitabine 800, 1000 and 1000mg/m2 respectively.<br />

After observing DLTs at higher dose level and poorer mTOR signaling<br />

inhibition at lower doses, a new cohort <strong>of</strong> sirolimus 5mg/24h plus<br />

gemcitabine 800 mg/m2 was added. Skin biopsies pre and post treatment<br />

were performed to assess the inhibition <strong>of</strong> mTOR pathway. Results: 19<br />

patients were enrolled: median age 51 years (36-70); gender 12M, 7F.<br />

Median number <strong>of</strong> cycles was 4. Patients were treated at 4 dose levels, the<br />

MTD was reached at level 3 and the RD was: sirolimus 5mg/24h and<br />

gemcitabine 800mg/m2 . 3 DLTs were observed, 1 at dose level 2 and 2 at<br />

dose level 3: transaminitis grade 3, thrombocytopenia grade 3 and<br />

thrombocytopenia grade 4. Other toxicities grade 1-2 included anemia,<br />

neutropenia, asthenia, mucositis and high cholesterol levels. 2 patients<br />

achieved partial response (1 uterine cervix cancer and 1 colon cancer).<br />

Immunohistochemistry <strong>of</strong> pS6 in skin biopsies showed significative inhibition<br />

<strong>of</strong> mTOR pathway at RD. PK parameters estimated were in agreement<br />

with those previously reported in the literature. No influence <strong>of</strong> sirolimus<br />

administration on gemcitabine clearance was found. Conclusions: Combination<br />

<strong>of</strong> sirolimus and gemcitabine is feasible and safe, allowing administration<br />

<strong>of</strong> active doses <strong>of</strong> both agents and achieving mTOR signaling<br />

inhibition. A phase II study to assess the activity <strong>of</strong> this combination in<br />

sarcomas is ongoing.<br />

3098 General Poster Session (Board #19H), Mon, 8:00 AM-12:00 PM<br />

A phase Ib pro<strong>of</strong>-<strong>of</strong>-concept study <strong>of</strong> LBH589 (LBH) and everolimus (EVE)<br />

in advanced solid tumors enriched for Epstein-Barr virus (EBV)-related<br />

cancers. Presenting Author: Daniel Shao-Weng Tan, National Cancer<br />

Centre, Singapore, Singapore<br />

Background: Histone deacetylase and mTOR inhibition circumvent critical<br />

EBV-oncogenic pathways with preclinical studies demonstrating lytic<br />

induction in EBV infected cells, as well as immunomodulatory and<br />

antiangiogenic effects. Methods: Patients (Pt) with advanced solid tumors<br />

enriched for EBV–related cancers were enrolled to determine safety,<br />

tolerability, maximum tolerated dose (MTD), pharmacokinetics (PK), pharmacodynamics<br />

and preliminary antitumor activity. LBH was instituted<br />

7-days prior to combination treatment. NPC pt received antiviral prophylaxis<br />

<strong>of</strong> either acyclovir (Ac) or valaciclovir (Vc). Serum EBV DNA levels<br />

(EBNA-1) were measured weekly and plasma cytokines pr<strong>of</strong>iled using a<br />

31-plex Luminex panel. Results: 15 pt have been treated (M:F 13:2,<br />

median age 50, R: 38-63) 10 NPC, 5 non-NPC (colon, RCC, breast,<br />

gastric, sarcoma) at 3 dose levels – LBH (3x/wk)-EVE (daily):10-2.5, 10-5,<br />

15-5. Two dose limiting toxicities <strong>of</strong> G4 (grade) thrombocytopenia were<br />

observed at LBH15-RAD5. Significant adverse events (AE) (G�3) were<br />

dysphagia (1) and thrombocytopenia (3). Common AEs (G1/2) included<br />

fatigue (80%), anorexia (60%), mucositis (53%), xerostomia (26%),<br />

dysphagia (26%). Two minor responses were seen (1 NPC, 1 breast) and 2<br />

pt (1 NPC, 1 RCC) had prolonged stable disease (�16 weeks). Modulation<br />

<strong>of</strong> EBV DNA titres was seen only in NPC pt, with median fold-change from<br />

baseline <strong>of</strong> 10.9 (0.05-174). Pt on Ac prophylaxis (n�5) had significant<br />

increases in DNA titres (9-174 fold), while those on Vc (n�4) were less<br />

pronounced (0.05-11 fold, p�0.03), with one pt (with minor response)<br />

having persistent decline in EBV titres. In a limited pt subset (n�9, 30<br />

timepoints), plasma cytokine pr<strong>of</strong>iles were consistent with a T-cell response,<br />

specifically, elevated levels <strong>of</strong> FLT3L, IFN-gamma, IL-13 and<br />

IL-17. PK and PBMC target modulation studies are being analysed.<br />

Conclusions: LBH-EVE results in induction <strong>of</strong> EBV DNA titres with an<br />

associated host T cell response. MTD is LBH10-EVE5 in Asian pt, majority<br />

<strong>of</strong> whom had NPC. A pre-planned expansion cohort that incorporates<br />

multi-parametric functional imaging exploring two schedules <strong>of</strong> LBH in<br />

combination with EVE5 is ongoing.<br />

Developmental Therapeutics—Experimental Therapeutics<br />

197s<br />

3097 General Poster Session (Board #19G), Mon, 8:00 AM-12:00 PM<br />

The PI3K/mTOR inhibitor BEZ235 given twice daily for the treatment <strong>of</strong><br />

patients (pts) with advanced solid tumors. Presenting Author: Hendrik-<br />

Tobias Arkenau, Sarah Cannon Research Institute, London, United Kingdom<br />

Background: The PI3 kinase (PI3K) pathway is the most deregulated<br />

pathway in cancers and is an attractive target for antitumor therapy.<br />

BEZ235 is an oral highly specific and selective inhibitor <strong>of</strong> the PI3K and<br />

TORC1/2. The MTD (1200 mg) and toxicity pr<strong>of</strong>ile <strong>of</strong> BEZ235 once daily<br />

dosing has been established. This study was designed to evaluate twice<br />

daily dosing <strong>of</strong> BEZ235 and its effect on treatment tolerability, PK, PD, and<br />

preliminary efficacy. Methods: Pts with advanced disease were enrolled in a<br />

3�3 dose escalation schedule starting at 200 mg PO BID in 28 day cycles.<br />

For intrapatient PK comparison the first week pts received the total dose in<br />

a QD schedule. DLT assessment was in Cycle 1. Efficacy evaluations were<br />

every 2 cycles, and PK and PD were assessed. Results: 12 pts were enrolled<br />

at the following dose levels: 200 mg (n�3), 400 mg (n�3), 600 mg (n�3),<br />

and 600 mg (BID only, no QD lead-in) (n�3). No G4 AEs were reported. G3<br />

related AEs were mucositis (n�2), AST/ALT elevation (n�2), anorexia<br />

(n�1) and diarrhea (n�1). The most common related G1/2 adverse events<br />

(AEs) were anorexia (n�6), diarrhea (n�3), fatigue (n�2), and headache<br />

(n�2). DLTs <strong>of</strong> G3 mucositis were observed in 2 patients at the 600 mg<br />

BID dose level with 1 week 1200 mg QD lead-in, which were attributed to<br />

the QD lead-in dosing during the first week. 3 pts then enrolled at 600 mg<br />

BID without lead in and had no further DLT. PK shows consistent increase<br />

in PK parameters with dose level. Of 10 evaluable, 3 pts had stable disease<br />

(13� to 21� weeks, 2 colorectal, 1 endometrial). Of 9 evaluable, 4 pts at<br />

various dose levels had decreased PET SUV uptake by greater than 25%.<br />

Conclusions: BEZ235 is tolerable at a dose <strong>of</strong> 600 mg BID, with less toxicity<br />

than has been seen with equivalent QD dosing. Preliminary signs <strong>of</strong> clinical<br />

and PD activity are noted.<br />

3099 General Poster Session (Board #20A), Mon, 8:00 AM-12:00 PM<br />

Meta-analysis on the relationship between everolimus exposure and safety<br />

and efficacy. Presenting Author: Dalila B. Sellami, Oncology <strong>Clinical</strong><br />

Development, Novartis Pharmaceuticals Corporation, Florham Park, NJ<br />

Background: Data on everolimus exposure–safety and response relationships<br />

are available from individual studies. We performed a meta-analysis<br />

to assess these relationships using everolimus predose minimum blood<br />

concentration (Cmin). Methods: Individual patient data from 5 phase 2 or 3<br />

oncology studies in which steady-state predose pharmacokinetic samples<br />

were taken from patients administered everolimus monotherapy 10 mg/day<br />

were pooled. In a Cox regression, the times to first grade �3 select adverse<br />

events (AEs) associated with everolimus and first progression-free survival<br />

(PFS) event were related to log-transformed instant or time-averaged Cmin as time-varying covariates stratified by study. Probability <strong>of</strong> tumor size<br />

reduction according to RECIST (yes vs no) was assessed using a generalized<br />

linear mixed model fitted with GEE method with log-transformed instant or<br />

time-averaged Cmin as covariates and treatment arm as fixed effect. Results:<br />

945 patients were evaluable for efficacy, 938 for safety. Patient characteristics<br />

were relatively well balanced across studies. Regardless <strong>of</strong> whether<br />

instant or time-averaged Cmin was used, a 2-fold increase in everolimus<br />

exposure increased the risk <strong>of</strong> grade �3 pulmonary, metabolic, and<br />

stomatitis events (Table); no increased risk <strong>of</strong> other grade �3 AEs was<br />

observed. A 2-fold increase in everolimus exposure increased the likelihood<br />

<strong>of</strong> tumor size reduction; there was a trend <strong>of</strong> reduced risk <strong>of</strong> a PFS event<br />

(Table). Conclusions: A 2-fold increase in everolimus exposure is associated<br />

with a higher probability <strong>of</strong> tumor size reduction and increased risk <strong>of</strong><br />

high-grade pulmonary, metabolic, and stomatitis AEs associated with<br />

everolimus.<br />

Risk and odds ratios associated with 2-fold everolimus exposure increase.<br />

Tumor size<br />

Grade >3 AEs, risk ratio (95% CI)<br />

PFS event,<br />

risk ratio<br />

reduction,<br />

odds ratio<br />

Cmin Pulmonary Metabolic Stomatitis (95% CI) (95% CI)<br />

Time-averaged 1.93 1.30 1.49 0.90 1.40<br />

(1.12-3.34) (1.02-1.65) (1.05-2.10) (0.69-1.18) (1.23-1.60)<br />

Instant 2.15 1.37 1.44 0.88 1.43<br />

(1.17-3.92) (1.06-1.77) (1.01-2.05) (0.68-1.14) (1.25-1.63)<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


198s Developmental Therapeutics—Experimental Therapeutics<br />

3100 General Poster Session (Board #20B), Mon, 8:00 AM-12:00 PM<br />

A phase I study <strong>of</strong> S-222611 an oral reversible dual inhibitor <strong>of</strong> EGFR and<br />

HER2, in patients with solid tumors. Presenting Author: Richard D. Baird,<br />

University <strong>of</strong> Cambridge, Department <strong>of</strong> Oncology, Cambridge, United<br />

Kingdom<br />

Background: S-222611 is a novel oral, potent, reversible tyrosine kinase<br />

inhibitor with antiproliferative activity in human tumor cell lines expressing<br />

EGFR and/or HER2 in vitro and in mouse xenograft models. We conducted<br />

the first study in patients (pts) with solid tumors expressing EGFR or HER2.<br />

Methods: Daily oral doses <strong>of</strong> S-222611 were escalated in successive<br />

cohorts from 100 mg to a maximum 1600 mg, using a 3 � 3 design. Full<br />

plasma pharmacokinetic (PK) pr<strong>of</strong>iles were obtained in all pts for 7 days<br />

after a single dose on Day 1 and for 24 h following 21 days <strong>of</strong> once daily<br />

dosing (Cycle 1). Trough PK samples were drawn weekly during the first 3<br />

weeks <strong>of</strong> daily dosing. A CT scan was performed at baseline and every 8<br />

weeks post-dose to assess response (RECIST). Results: Of 23 treated pts<br />

(16 male) aged 24-77y, 19 completed Cycle 1. One dose-limiting toxicity<br />

(DLT) has been seen to date, a rash at 1200 mg, which resolved on<br />

interruption and dose reduction. Other adverse events related to drug,<br />

diarrhea, rash, and nausea, were mild (grade 1 or 2) but more frequent at<br />

higher doses. Tumor responses have been seen over a wide range <strong>of</strong> doses.<br />

One complete clinical response was observed at 1200 mg in a pt with<br />

HER2 positive breast carcinoma previously treated with lapatinib and<br />

trastuzumab. Two pts showed confirmed partial responses (one with EGFR<br />

positive renal cell carcinoma at 200 mg daily and one with EGFR and HER2<br />

positive esophageal carcinoma at 400 mg daily); 2 pts showed unconfirmed<br />

partial responses and 3 pts (with vaginal, gastric and pancreatic adenoCa)<br />

showed stable disease for �6 months (mo); 2 pts have been treated for<br />

�12 mo. Plasma Cmax, AUC and steady state concentrations increased with<br />

dose up to 1200 mg, exceeding those eliciting maximal responses in mice.<br />

The plasma t½ is �24 h. Conclusions: S-222611 was generally tolerated<br />

well in doses up to 1200 mg, with only one DLT; rash, diarrhea and nausea<br />

have been readily manageable. PK data support once daily dosing to<br />

achieve highly effective concentrations. A substantial proportion <strong>of</strong> patients<br />

with HER2 or EGFR expressing tumors have shown partial responses<br />

or stable disease (�6 mo) and one patient has shown a complete clinical<br />

response.<br />

3103 General Poster Session (Board #20E), Mon, 8:00 AM-12:00 PM<br />

A phase I study <strong>of</strong> selumetinib (AZD6244/ARRY-142866) in combination<br />

with cetuximab (cet) in refractory solid tumors and KRAS mutant colorectal<br />

cancer (CRC). Presenting Author: Dustin A. Deming, University <strong>of</strong> Wisconsin<br />

Carbone Cancer Center, Madison, WI<br />

Background: KRAS mutations have been recognized as clinically important<br />

predictors <strong>of</strong> resistance to EGFR-directed therapies in CRC. Oncogenic<br />

activation <strong>of</strong> the RAS/RAF/MEK/ERK signaling cascade mediates proliferation<br />

independent <strong>of</strong> growth factor receptor signaling. We hypothesized that<br />

targeting MEK with selumetinib could overcome resistance to cet in KRAS<br />

mutant CRC. A phase I study (NCT01287130) was undertaken to determine<br />

the tolerability, and pharmacokinetic pr<strong>of</strong>iles <strong>of</strong> the combination <strong>of</strong><br />

selumetinib and cet, with an expanded cohort in KRAS mutant CRC at the<br />

MTD dose to evaluate preliminary anti-tumor activity. Methods: In the dose<br />

escalation portion, patients (pts) with advanced solid tumors received fixed<br />

dose cet with escalating doses <strong>of</strong> selumetinib in cohorts <strong>of</strong> 3-6 pts. In the<br />

expansion cohort, 14 pts with KRAS mutant CRC were enrolled at the MTD<br />

level. Results: 15 pts (9 M, 6 F), average age <strong>of</strong> 60 (41-73) years were<br />

treated at 3 dose levels in the dose escalation cohort and 14 pts were<br />

treated in the expansion cohort. Pts had the following tumor types: CRC<br />

73%, NSCLC 13%, and H&N 13%, and had received a median <strong>of</strong> 4 (1-8)<br />

prior lines <strong>of</strong> therapy. 33% (only CRC) had prior EGFR-directed therapies.<br />

ECOG PS 0 (40%), 1 (53%), 2 (7%). 13 <strong>of</strong> 15 pts were evaluable for<br />

tolerability and response. One DLT for grade 4 hypomagnesemia occurred,<br />

and no other grade 4 toxicities were seen. Grade 3 (20%) toxicities<br />

included; rash, hyponatremia, and headache. The most common cycle 1<br />

grade 1 and 2 adverse events included acneiform rash (100%), fatigue<br />

(54%), nausea/vomiting, (54%), diarrhea (54%), dry skin (46%), fever<br />

(23%), and hypomagnesemia (15%). Most pts (60%) required no dose<br />

modifications. The MTD was established at selumetinib 75 mg PO BID and<br />

cet 250 mg/m2 weekly following a 400 mg/m2load. Best response included<br />

2 PR in pts with CRC and SD in 4 pts (1 SCC <strong>of</strong> the tonsil, 1 NSCLC, and 2<br />

CRC). Conclusions: The combination <strong>of</strong> selumetinib and cet is well<br />

tolerated, and preliminary anti-tumor activity was observed in multiple pts.<br />

Results <strong>of</strong> the KRAS mutant CRC expansion cohort will be presented.<br />

3101 General Poster Session (Board #20C), Mon, 8:00 AM-12:00 PM<br />

A phase I study <strong>of</strong> the gamma-secretase inhibitor RO4929097 and<br />

capecitabine in refractory solid tumors. Presenting Author: Noelle K.<br />

LoConte, University <strong>of</strong> Wisconsin Carbone Cancer Center, Madison, WI<br />

Background: RO4929097 is a oral inhibitor <strong>of</strong> gamma-secretase, which<br />

results in Notch signaling inhibition. Prior work has demonstrated that<br />

Notch-signaling inhibition enhances chemosensitivity <strong>of</strong> colon cancer<br />

cells. This study sought to combine RO4929097 with capecitabine (cape)<br />

and determine maximum tolerated dose (MTD), toxicities and efficacy.<br />

Preclinical and prior phase I work demonstrated possible autoinduction <strong>of</strong><br />

RO4929097, so pharmacokinetic (PK) evaluation <strong>of</strong> RO4929097 and<br />

cape was planned. Methods: Adult patients with refractory solid tumors<br />

were eligible and received RO4929097 and cape at a fixed dose <strong>of</strong> 1000<br />

mg/m2 BID with escalating doses <strong>of</strong> RO4929097 on a 21-day cycle<br />

according to a 3�3 design. Cape was administered for 14 days and the<br />

RO49029097 once daily, 3 days per week. RO4929097 plasma concentrations<br />

were evaluated by LC/MS/MS on days 3 and 10 <strong>of</strong> cycle 1, and PK<br />

parameters analyzed with WinNonLin version 5.2. Results: 4 dose levels<br />

have been completed (20, 30, 45 and 68 mg); 18 <strong>of</strong> 19 patients are<br />

evaluable for toxicity and PK data is available for 11 patients. One DLT has<br />

been observed (intolerable grade 2 fatigue) at 68 mg. There have been 2<br />

confirmed partial responses: fluoropyrimide-refractory colon cancer (for 12<br />

cycles) and cervical cancer (for 6 cycles). The half-life, Cmax and AUC <strong>of</strong><br />

RO4929097 all significantly decreased on day 10 compared to day 3, with<br />

an increase in clearance for all doses. The half-life was significantly shorter<br />

in dose level 3 (p�0.0012) and dose level 4 (p�0.0126) compared to<br />

dose level 1. Conclusions: RO4929097 plus cape was well tolerated.<br />

Activity was seen in cervical and colon cancer. Autoinduction <strong>of</strong> RO4929097<br />

was seen both with increasing cycle number and increasing dose. However,<br />

all plasma concentrations <strong>of</strong> RO4929097 were above those needed for<br />

Notch inhibition (1.9 ng/mL based on prior studies). Dose level 1 (cape<br />

1000 mg/m2 BID and RO4929097 20 mg daily) is the recommended<br />

phase II dose.<br />

N�11<br />

Ratio day 10/day 3<br />

Mean (95% CI) P value<br />

Half-life (hr) 0.62 (0.44 – 0.80) � 0.001<br />

Cmax/dose 0.63 (0.44 – 0.82) � 0.001<br />

AUC/dose 0.50 (0.12 – 2.57) 0.016<br />

Vd (mL) 1.88 (1.19 – 2.57) � 0.001<br />

Cl (mL/hr) 3.69 (2.13 – 5.25) � 0.001<br />

3104 General Poster Session (Board #20F), Mon, 8:00 AM-12:00 PM<br />

Phase I trial to assess the safety and pharmacokinetics <strong>of</strong> afatinib and<br />

weekly vinorelbine in patients with advanced solid tumors. Presenting<br />

Author: Antoine Hollebecque, Institut Gustave Roussy, Villejuif, France<br />

Background: Afatinib (A) is an oral, irreversible ErbB Family Blocker with<br />

activity in a wide range <strong>of</strong> tumor cell lines dependent on ErbB signaling.<br />

Vinorelbine (VNR) interferes with tubulin polymerization and spindle<br />

formation during metaphase. Additive or supra-additive activity <strong>of</strong> A or<br />

EGFR TKI with VNR has been demonstrated in preclinical models. Methods:<br />

This dose-escalation Phase I study established the safety pr<strong>of</strong>ile, MTD and<br />

PK <strong>of</strong> A with i.v. and p.o. VNR using a modified 3�3 design. Eligible<br />

patients (pts) were �18 years with refractory advanced or metastatic<br />

tumors, an ECOG PS 0–1, and adequate organ and bone marrow function.<br />

VNR i.v. (25 mg/m2 )/oral (60 mg/m2 with escalation to 80 mg/m2 after 3<br />

weeks) was administered weekly on Days 1, 8, 15 and 22 with escalating<br />

oral daily doses <strong>of</strong> A 20 mg, 40 mg and 50 mg in 28-day treatment courses.<br />

Results: The study treated 55 pts: 30 pts for MTD determination and 25 pts<br />

in the PK expansion cohort (24 M/31 F), median age 54 years (range<br />

34–72). 28/27 pts were included in the VNR i.v./p.o. cohorts, respectively.<br />

Patients had NSCLC, breast, pancreatic (n�13/5/3), head and neck,<br />

stomach or colorectal cancer (n�2 each group); 28 pts had other cancers.<br />

At 20 mg A, no DLTs occurred in the VNR i.v./p.o. cohorts; whereas, at 50<br />

mg A, 4/6 pts and 3/5 pts experienced DLTs. At 40 mg A and VNR i.v./p.o.,<br />

1/6 pts included for MTD determination experienced DLT, as did 7/13 pts<br />

and 2/12 pts in the VNR i.v./p.o. PK expansion cohorts, respectively. Main<br />

DLTs were diarrhea and febrile neutropenia. Median treatment duration<br />

(snapshot date 9 Dec 2011) across all dose groups was 80 days (range<br />

10–687), with 98 and 58 days in the MTD cohorts <strong>of</strong> VNR i.v./p.o.,<br />

respectively. Related AEs observed in most patients were diarrhea, asthenia,<br />

nausea, vomiting, neutropenia, decreased appetite, mucositis and<br />

rash. Five pts had a PR, confirmed in 2 pts. Preliminary PK analyses<br />

suggest no drug–drug interaction between A and i.v. VNR. Conclusions:<br />

Afatinib in combination with weekly i.v./p.o. VNR is tolerated, with<br />

manageable, reversible, target-related side effects and promising signs <strong>of</strong><br />

clinical activity in heavily pretreated patients. The MTD for A for both<br />

combinations is 40 mg daily.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


3105 General Poster Session (Board #20G), Mon, 8:00 AM-12:00 PM<br />

A phase Ib trial <strong>of</strong> FOLFIRINOX plus saridegib, an oral hedgehog (Hh)<br />

inhibitor, in pts with advanced pancreatic cancer (PDAC). Presenting<br />

Author: Andrew H. Ko, University <strong>of</strong> California, San Francisco Comprehensive<br />

Cancer Center, San Francisco, CA<br />

Background: FOLFIRINOX has emerged as the optimal 1st-line treatment<br />

option for pts with advanced PDAC and good performance status; whether it<br />

can serve as the backbone upon which to add targeted agents in clinical<br />

trial design remains uncertain. The goal <strong>of</strong> this multicenter phase Ib study<br />

is to evaluate FOLFIRINOX in combination with saridegib, a novel oral<br />

agent that inhibits the Hh signaling pathway. In preclinical models <strong>of</strong><br />

PDAC, saridegib increases chemotherapy delivery by depleting peritumoral<br />

stroma and increasing vascularity. Methods: Pts with previously untreated<br />

metastatic or locally advanced PDAC and ECOG PS 0-1 were eligible.<br />

Treatment consists <strong>of</strong> once-daily saridegib with concurrent administration<br />

<strong>of</strong> biweekly FOLFIRINOX (omitting the 5-FU bolus). A 3�3 dose escalation<br />

design was used (see dose levels below). Prophylactic WBC growth factor<br />

support is mandated. DLT definitions include ALT/AST �10x ULN, grade 4<br />

plts or ANC �5 d, or grade 3-4 nonheme toxicity. CT scans are obtained<br />

every 4 cycles. Limited PK analyses are performed. Results: Seven pts have<br />

been enrolled at the first 2 dose levels. Grade 1-2 AEs include GI (N/V/D),<br />

dehydration, fatigue, and LFT abnormalities. There was one DLT (grade 3<br />

ALT elevation) at DL2. Other serious toxicities seen include grade 3 nausea<br />

(DL1) and grade 3 diarrhea (DL2). Tumor shrinkage has been observed in<br />

all 4 pts at DL1, ranging from 17-54%, with 2 unconfirmed PRs. Final MTD<br />

determination and updated safety and efficacy data will be presented at the<br />

meeting. Conclusions: A modified FOLFIRINOX regimen can be safely<br />

administered in combination with novel agents in clinical trials <strong>of</strong> PDAC.<br />

While saridegib was not beneficial when added to gemcitabine in a separate<br />

randomized phase II study, early evidence <strong>of</strong> significant responses on the<br />

current trial suggests that a more intensive chemotherapy platform may<br />

represent a preferable strategy in PDAC trial design.<br />

Dose level<br />

5-FU bolus<br />

(mg/m2)<br />

5-FU infusion<br />

(mg/m2 x 46 hrs)<br />

LV<br />

(mg/m2) Oxaliplatin Irinotecan<br />

(mg/m2) (mg/m2)<br />

IPI-926<br />

(mg/day)<br />

-1 -- 1600 400 50 120 130<br />

1* -- 1920 400 65 150 130<br />

2 -- 2400 400 85 180 130<br />

3 -- 2400 400 85 180 160<br />

*Starting dose level.<br />

3107^ General Poster Session (Board #21A), Mon, 8:00 AM-12:00 PM<br />

First-in-human phase I dose-escalation study <strong>of</strong> the oral selective C-met<br />

inhibitor EMD 1204831 in patients with advanced solid tumors. Presenting<br />

Author: Hesham M. Amin, Department <strong>of</strong> Hematopathology, University<br />

<strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: The cell surface receptor tyrosine kinase c-Met and its ligand,<br />

the hepatocyte growth factor, are implicated in tumor cell migration,<br />

invasion, survival, and proliferation. EMD 1204831 is a novel potent and<br />

highly selective reversible, ATP-competitive small molecule c-Met inhibitor.<br />

Methods: This is a phase I, first-in-human clinical trial with escalating<br />

doses <strong>of</strong> EMD 1204831 (NCT01110083). The primary objective was to<br />

determine the maximum tolerated dose (MTD). Secondary objectives<br />

included evaluation <strong>of</strong> safety, pharmacokinetics (PK), pharmacodynamics<br />

(Pd), and preliminary anti-tumor activity. Eligible patients had advanced<br />

solid tumors not amenable to standard therapies. Following a classical 3�3<br />

dose-escalation scheme, successive cohorts <strong>of</strong> patients were treated with<br />

twice daily (BID) oral EMD 1204831 in 21-day cycles. Pd markers were<br />

evaluated in paired tumor biopsies (phospho-c-Met). Results: Until 31<br />

December 2011, 30 patients were enrolled and treated. The dose was<br />

escalated in successive cohorts starting from 50 mg BID up to 1400 mg<br />

BID. After first (single) administration, median Cmax and AUC0–12 values<br />

increased with dose. At higher doses, a decrease in exposure <strong>of</strong> EMD<br />

1204831 was noted after multiple dosing, potentially caused by autoinduction<br />

<strong>of</strong> the compound’s metabolism. Further dose escalation was discontinued,<br />

and no further patients were enrolled. One dose-limiting toxicity (DLT)<br />

<strong>of</strong> grade (G) 3 pancreatitis, considered as a serious adverse event (AE), was<br />

observed at 400 mg BID. No other DLTs or treatment-related serious AEs<br />

were observed. The remaining treatment-related AEs <strong>of</strong> G2 or higher<br />

included G3 and G2 lipase elevation (n�1 for each grade), G2 upper<br />

abdominal pain (n�2), G2 gastroesophageal reflux disease (n�2), and G2<br />

constipation (n�1). Twenty-five patients (83%) had no drug-related<br />

toxicity greater than G1. Of 29 patients evaluable for anti-tumor activity, 3<br />

had stable disease lasting for at least 4 months. Conclusions: Due to<br />

potential autoinduction <strong>of</strong> the compound’s metabolism, dose escalation<br />

was discontinued before an MTD was reached. Final safety, PK, and clinical<br />

tumor response results will be presented.<br />

Developmental Therapeutics—Experimental Therapeutics<br />

199s<br />

3106 General Poster Session (Board #20H), Mon, 8:00 AM-12:00 PM<br />

NRAS mutations in patients with advanced cancers treated with targetbased<br />

therapies in early-phase clinical trials. Presenting Author: Vinu<br />

Madhusudanannair, Department <strong>of</strong> Investigational Cancer Therapeutics<br />

(Phase I Program), University <strong>of</strong> Texas M. D. Anderson Cancer Center,<br />

Houston, TX<br />

Background: NRAS mutations in cancers increase MAPK signaling. It is<br />

plausible that NRAS mutations may be associated with sensitivity to drugs<br />

targeting the MAPK pathway. Methods: Tumors from 689 patients with<br />

advanced cancers referred to the <strong>Clinical</strong> Center for Targeted Therapy<br />

(phase I program) were screened in a CLIA-certified laboratory for NRAS<br />

mutations and if feasible, for PIK3CA, KRAS, BRAF mutations and PTEN<br />

aberrations. Whenever possible, patients with NRAS mutations were<br />

treated with agents targeting the RAF-MEK pathway. Results: Of the 689<br />

patients, 58 (8%) had NRAS mutations. NRAS mutations were most<br />

prevalent in melanoma (26/170, 25%), thyroid (5/26, 19%), endometrial<br />

(2/27, 7%), non-small cell lung (1/31, 3%), ovarian (1/60, 2%) and<br />

colorectal cancers (1/74, 1%). NRAS mutations were not seen in any <strong>of</strong> the<br />

tested head and neck squamous cell cancers (n � 44); sarcomas (n � 41)<br />

and breast cancers (n � 25). None (0/62, 0%) <strong>of</strong> the patients tested for<br />

KRAS had simultaneous NRAS and KRAS mutations, and only 2/42 (5%)<br />

tested for BRAF had simultaneous NRAS and BRAF mutations. Of 58<br />

patients with NRAS mutations, 14 (24%) were enrolled in trials that<br />

included MEK axis inhibitors. Of these 14 patients, 13 had melanoma and<br />

1 had ovarian cancer and 4/14 (29%) demonstrated tumor shrinkage<br />

�10% from baseline. In comparison, only 22/187 (12%) patients with<br />

wild-type/unknown NRAS, KRAS and BRAF treated on the same trials<br />

demonstrated tumor shrinkage �10% (p�0.086). Conclusions: NRAS<br />

mutations are diagnosed in variety <strong>of</strong> advanced cancers. Patients with<br />

NRAS mutations demonstrated a trend to better antitumor activity with<br />

MEK axis inhibitors compared to patients without NRAS, KRAS or BRAF<br />

mutations. Further studies are warranted to delineate the role <strong>of</strong> NRAS<br />

mutations in patients treated with MEK axis inhibitors.<br />

3108 General Poster Session (Board #21B), Mon, 8:00 AM-12:00 PM<br />

Phase I trials <strong>of</strong> PS-341 (bortezomib, B) in combination with topotecan (T)<br />

in advanced solid tumors: A potential pharmacokinetic (PK) interaction.<br />

Presenting Author: Robert Morgan, City <strong>of</strong> Hope, Duarte, CA<br />

Background: Preclinical data has demonstrated that B when combined with<br />

camptothecins enhances apoptotic effects which may be independent <strong>of</strong><br />

NF-�B status and dependent on the sequence <strong>of</strong> exposure to the proteasome<br />

inhibitor. Other data suggests PK interactions <strong>of</strong> B with some<br />

chemotherapeutic agents. It is hypothesized that B will act synergistically<br />

with T and enhance the apoptotic effects <strong>of</strong> T. Conflicting data exists<br />

regarding schedule dependent synergistic cytotoxicity <strong>of</strong> the combination.<br />

This study examined both sequences <strong>of</strong> administration and determined the<br />

PK <strong>of</strong> each sequence. Methods: A standard 3 � 3 schedule was used in both<br />

sequences (S). The dose <strong>of</strong> B was fixed with escalating doses <strong>of</strong> T. S1: B 1.3<br />

mg/m2 on days 1, 4, 8, and 11.T2to3.5mg/m2 on days 1 and 8 was given<br />

6 h before B. S2: B 1.3 mg/m2 on days 1, 4, 8, 11 followed by T2to3<br />

mg/m2 given 6 h after B. Cycle length was q28 days. PK sampling for both<br />

sequences was performed. Results: 54 pts (S1: 27, S2: 27) received a<br />

median <strong>of</strong> 2 cycles (range 1-7) <strong>of</strong> therapy. Pts were heavily pre-treated. For<br />

S1, the maximum tolerated dose (MTD) <strong>of</strong> B and T were 1.3 and 3 mg/m2 respectively. For S2, the MTD <strong>of</strong> B and T were 1.3 and 2.5 mg/m2 respectively. Dose-limiting toxicities (2 pt in each sequence) were gr 3<br />

thrombocytopenia in S1, and gr 4 thrombocytopenia in S2. Stable disease<br />

was seen in 6/27 pts in S1, and 6/27 in S2. PK results for S2 show no<br />

significant difference between the day 1 and day 8 PKs. However, the mean<br />

AUC (454�150 mg/Lxhr) and Cmax (123�51 mg/L) at the MTD <strong>of</strong> 2.5<br />

mg/m2 reached on this trial is similar to those reported with single agent<br />

doses <strong>of</strong> 4 mg/m2 given over 30 minutes weekly x 3. As a result, the<br />

estimated topotecan clearance (11.3�5 L/hr) when given following bortezomib<br />

is approximately one half <strong>of</strong> the reported clearance for the single<br />

agent. (Curtis et al, J Clin Pharmacol, 50:268, 2010). Conclusions: The<br />

tolerability and toxicity <strong>of</strong> both sequences was similar. MTD differs with S2<br />

having a lower tolerated dose <strong>of</strong> T. PK results suggest a possible drug-drug<br />

interaction with decreased clearance <strong>of</strong> T with S2. PK results for S1 are<br />

pending. Supported by CCSG CA62505.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


200s Developmental Therapeutics—Experimental Therapeutics<br />

TPS3109 General Poster Session (Board #21C), Mon, 8:00 AM-12:00 PM<br />

Vorinostat to restore sensitivity to aromatase inhibitor therapy in metastatic<br />

breast cancer: A phase II clinical trial with ER imaging correlates.<br />

Presenting Author: Hannah M. Linden, University <strong>of</strong> Washington, Seattle,<br />

WA<br />

Background: Endocrine refractory,Indolent, s<strong>of</strong>t tissue, and bone dominant<br />

metastatic breast cancer is a clinical problem, for which alternatives to<br />

chemotherapy are needed, Histone deacetylace inhibitors (HDACi) have<br />

shown pre-clinical promise in estrogen receptor-modulation and restoring<br />

sensitivity to endocrine manipulation, suggesting potential clinical benefit.<br />

HDACi restore ER expression in ER-negative cells which are then sensitive<br />

to AI treatment in xenografts (Sabnis 2011) and are toxic to ER-positive<br />

cell lines (Huang 2000). Vorinostat is an FDA approved HDACi for CTCL,<br />

and could have a beneficial role in restoring ER-signaling in endocrineresistant<br />

tumors. We hypothesized that HDACi therapy could restore<br />

sensitivity to aromatase inhibitor (AI) therapy, in patients with prior<br />

progression on AI, and allow continued endocrine therapy. Recent data<br />

suggest clinical benefit <strong>of</strong> Tamoxifen and vorinostat given continuously<br />

(Munster 2011) as well as entinostat and an AI (Yardley 2011). Our prior<br />

work has shown the feasibility <strong>of</strong> monitoring regional ER expression and<br />

ER-estrogen binding in vivo using [F-18]fluoroestradiol (FES) PET imaging<br />

(Linden 2011) providing a biomarker to interrogate the molecular target.<br />

Methods: Patients with metastatic breast cancer with prior clinical benefit<br />

from endocrine manipulation who progressed on AI therapy are eligible.<br />

Patients must be <strong>of</strong>f ER blocking therapies, and functionally postmenopausal.<br />

Baseline FES and FDG PET are performed and patients receive<br />

investigational vorinostat treatment at 400 mg po BID for 2 weeks followed<br />

by serial FES and FDG PET to assess the impact <strong>of</strong> vorinostat on ER<br />

expression and tumor metabolism. Patients are retreated on their prior AI as<br />

a single agent for 6 weeks, followed by paired FES and FDG and clinical<br />

assessment. Patients with clinical benefit may continue on treatment: 2<br />

weeks <strong>of</strong> vorinostat followed by 6 weeks <strong>of</strong> AI in 8-week cycles until<br />

progressive disease or toxicity. We have enrolled 4 <strong>of</strong> a planned 20 patients.<br />

TPS3111 General Poster Session (Board #21E), Mon, 8:00 AM-12:00 PM<br />

A phase I and pharmacologic study <strong>of</strong> MM-111, a bispecific HER2/HER3<br />

antibody fusion protein, in combination with multiple treatment regimens<br />

in patients with advanced HER2-positive solid tumors. Presenting Author:<br />

Donald A. Richards, Texas Oncology-Tyler, Tyler, TX<br />

Background: The HER2/HER3 oncogenic heterodimer is a potent HER<br />

receptor pairing with respect to strength <strong>of</strong> interaction, receptor tyrosine<br />

phosphorylation, and downstream signaling through mitogen activated<br />

protein kinase and phosphoinositide-3 kinase pathways. MM-111 is a novel<br />

bispecific antibody fusion protein that inhibits ligand activated HER3<br />

signaling through abrogation <strong>of</strong> ligand binding specifically in HER2<br />

overexpressing tumors. In preclinical models <strong>of</strong> HER2� solid tumors,<br />

MM-111 inhibits ligand-induced HER3 phosphorylation, cell cycle progression,<br />

and tumor growth. HER2 is a well-established target <strong>of</strong> anticancer<br />

agents such as trastuzumab and lapatinib, specifically in breast and gastric<br />

tumors. Preclinical data demonstrate that MM-111 potentiates the antitumor<br />

activity <strong>of</strong> both trastuzumab and lapatinib and this phase I study seeks<br />

to determine if MM-111 can be safely used in combination with various<br />

standard <strong>of</strong> care HER2 targeting regimens, namely; (1) trastuzumab,<br />

cisplatin and, capecitabine, (2) trastuzumab and lapatinib, and (3)<br />

trastuzumab and paclitaxel. Methods: This is a multi-arm phase I, open<br />

label, multicenter, dose-escalation study <strong>of</strong> MM-111 in an add-on design<br />

that evaluates the safety, pharmacokinetics (PK), and anti-tumor activity <strong>of</strong><br />

MM-111 in combination. Each arm is designed to run as a separate phase I<br />

study to address safety and tolerability <strong>of</strong> each combination. For eligibility,<br />

patients are required to have documented advanced HER2-positive cancer,<br />

with adequate bone marrow, hepato-renal and cardiac function. The dose<br />

escalation for each arm utilizes a standard “3 � 3” design with MM-111<br />

dosed initially at a moderate dose <strong>of</strong> 10 mg/kg and escalated up to 20<br />

mg/kg to identify a phase II dose in combination with each regimen. Once<br />

the maximum tolerated dose or phase II dose is identified, expansion<br />

cohorts will accrue to further evaluate these combinations. The flexibility <strong>of</strong><br />

the design allows additional combinations arms to be added. Key exploratory<br />

analyses will include an evaluation <strong>of</strong> safety, PK, and efficacy as<br />

evidenced by best overall response and duration <strong>of</strong> response.<br />

TPS3110 General Poster Session (Board #21D), Mon, 8:00 AM-12:00 PM<br />

Design <strong>of</strong> a phase I clinical trial with PNT2258, a novel DNA interference<br />

oligonucleotide (DNAi), in patients with advanced solid tumors. Presenting<br />

Author: Drew Warren Rasco, South Texas Accelerated Research Therapeutics,<br />

LLC, San Antonio, TX<br />

Background: With the knowledge that Bcl-2 facilitates drug resistance and<br />

cell survival, a DNA interference (DNAi) strategy was applied to silence<br />

Bcl-2 in cancer cells and promote apoptosis. DNAi differs from cytoplasmic<br />

mRNA targeting (antisense, RNAi, and miRNA targets) as it targets<br />

genomic DNA, blocking transcription. PNT100, a first in class DNAi, is a<br />

novel single-stranded 24-base unmodified DNA designed to bind to an<br />

upstream region <strong>of</strong> the Bcl-2 promoter. The drug product (PNT2258) is<br />

PNT100 encapsulated in a specialized pH tunable liposome and is being<br />

assessed for safety and tolerability in a phase I trial. PNT2258 avoids the<br />

toxicities associated with modified oligonucleotides and double-stranded<br />

RNAs; since the liposome formulation is anionic and contains no surface<br />

spacers, vehicle toxicities are minimal. Xenograft experiments demonstrated<br />

marked single agent activity in a diffuse large cell lymphoma, and<br />

therapy potentiation when combined with either rituximab in Daudi-<br />

Burkitt’s Lymphoma or docetaxel in A375 melanoma. Methods: An openlabel,<br />

single-arm, first-in-man phase I dose-escalation study <strong>of</strong> PNT2258<br />

in patients with advanced solid tumors was designed to evaluate safety,<br />

tolerability, dose-limiting toxicities, pharmacokinetics, and pharmacodynamics<br />

<strong>of</strong> PNT2258 to recommend a dose for phase II studies. In this<br />

phase I study, pharmacodynamic effects <strong>of</strong> PNT2258 will be evaluated<br />

through analyses <strong>of</strong> soluble serum and plasma markers and peripheral<br />

blood mononuclear cells. Patients will receive PNT2258 as an intravenous<br />

infusion over 2 hours once daily for 5 consecutive days (days 1-5) <strong>of</strong> each<br />

21-day treatment cycle (3 weeks). The starting dose <strong>of</strong> 1 mg/m2 with<br />

PNT2258 administered to one patient per cohort and dose-escalation will<br />

proceed by dose-doubling in each successive cohort until a dose level <strong>of</strong> 64<br />

mg/m2 is attained, provided no dose-limiting toxicities are observed in<br />

cycle 1. Thereafter, dose escalations shall proceed at 33% increments <strong>of</strong><br />

the previous cohort dose-level to 85, 113, and 150 mg/m2 with expansions<br />

<strong>of</strong> up to six patients per cohort as needed. The ten planned dose cohorts<br />

have been completed with all patients enrolled.<br />

TPS3112 General Poster Session (Board #21F), Mon, 8:00 AM-12:00 PM<br />

A phase I trial <strong>of</strong> riluzole and sorafenib in patients with advanced solid<br />

tumors and melanoma. Presenting Author: Janice M. Mehnert, Cancer<br />

Institute <strong>of</strong> New Jersey, New Brunswick, NJ<br />

Background: Metabotropic glutamate receptor 1 (GRM1) has been identified<br />

as a potential therapeutic target in melanoma. Over 60% <strong>of</strong> human<br />

melanomas express this cell surface glutamate receptor and excitation <strong>of</strong><br />

GRM1 results in the activation <strong>of</strong> MAPK and PI3K/AKT pathways. Riluzole,<br />

an oral GRM1 blocking agent, results in growth arrest <strong>of</strong> melanoma cells in<br />

vitro and in vivo. We previously reported that administration <strong>of</strong> riluzole to<br />

melanoma patients suppressed activity <strong>of</strong> the PI3K/AKT and MAPK<br />

pathways in paired tumor samples (Yip Clin Cancer Res 2009). In<br />

preclinical studies, the efficacy <strong>of</strong> riluzole was attenuated in melanoma<br />

cells harboring BRAFV600E mutations, but sorafenib, a RAF kinase<br />

inhibitor, enhanced the effect <strong>of</strong> riluzole on these cells. The combination <strong>of</strong><br />

riluzole and sorafenib was additive or synergistic in both BRAF mutant and<br />

BRAF wildtype melanoma cells in vitro and in BRAF wildtype cells in a<br />

xenograft model (Lee HJ Clin Cancer Res 2011). We thus designed a phase<br />

I trial to test the combination <strong>of</strong> riluzole with sorafenib in patients with solid<br />

tumors and advanced melanoma. Methods: The primary objective <strong>of</strong> this<br />

trial is identification <strong>of</strong> the maximum tolerated dose (MTD). An expansion<br />

cohort at the MTD is planned for patients with advanced melanoma to<br />

examine the correlation <strong>of</strong> clinical or radiographic response with signaling<br />

through the MAPK and PI3K/AKT pathways and with GRM1 receptor status<br />

<strong>of</strong> individual tumors. Eligible patients must have advanced solid tumors<br />

(phase I) or stage III unresectable/stage IV melanoma with biopsiable tumor<br />

(expansion cohort) and ECOG PS � 2. Riluzole will be administered at 100<br />

mg twice daily combined with sorafenib beginning at 200 mg daily and<br />

escalating in subsequent cohorts at 200 mg increments. Correlative<br />

studies: Tumors will be assessed for BRAF and NRAS mutational status.<br />

Pretreatment tumor blocks will be examined for GRM1 receptor status by<br />

immunohistochemistry. Pre and post treatment levels <strong>of</strong> pERK and pAKT<br />

will be measured in paired tumor samples to assess effects <strong>of</strong> treatment on<br />

MAPK and PI3K signaling. Limited sampling pharmacokinetic studies will<br />

be performed. Progress: Accrual to three cohorts is complete without DLT.<br />

Accrual to the final planned cohort is in progress.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


TPS3113 General Poster Session (Board #21G), Mon, 8:00 AM-12:00 PM<br />

First-in-human phase I clinical trial <strong>of</strong> QBI-139, a human ribonuclease<br />

variant, in solid tumors. Presenting Author: Laura E. Strong, Quintessence<br />

Biosciences, Inc., Madison, WI<br />

Background: RNA has been recognized as a drug target for cancer therapy,<br />

as evidenced by the ongoing clinical trials <strong>of</strong> RNAi and antisense therapies.<br />

An alternative approach that circumvents the delivery and stability issues <strong>of</strong><br />

RNAi and antisense is to harness the activity <strong>of</strong> naturally occurring enzymes<br />

that degrade RNA. Variants <strong>of</strong> human ribonucleases (RNase) have been<br />

generated with diminished binding to their natural inhibitor inside cells,<br />

which allows the new proteins to kill cancer cells. QBI-139, a variant that<br />

retains 95% sequence identity to a naturally occurring RNase, has<br />

demonstrated efficacy as both a single agent and in combination against<br />

multiple tumor types in in vivo models <strong>of</strong> human cancer. A first in human<br />

phase I trial was designed and initiated for QBI-139. Methods: Since<br />

QBI-139 showed efficacy against multiple cancers in model systems, a first<br />

in human phase I trial was designed for patients with advanced solid<br />

tumors (NCT00818831). Patients receive QBI-139 by intravenous infusion<br />

once weekly for a cycle <strong>of</strong> three weeks. In the absence <strong>of</strong> disease<br />

progression or unacceptable toxicity, treatment can continue on the twenty<br />

one day cycle. The trial is a standard 3�3 design with cohorts <strong>of</strong> three to six<br />

patients receiving escalating doses <strong>of</strong> QBI-139 until the maximum tolerated<br />

dose (MTD) and/or recommended phase II dose is determined. The<br />

inclusion/exclusion criteria are typical for the patient population. Forty<br />

three patients have been treated to date (January 2012) without identification<br />

<strong>of</strong> dose limiting toxicity. The starting dose in the clinical trial was 3<br />

mg/m2 while the most recently completed cohort was treated with a dose <strong>of</strong><br />

50.4 mg/m2 . Dose escalation is ongoing. The primary outcome is to<br />

evaluate the toxicity and tolerability <strong>of</strong> QBI-139 in patients with advanced<br />

refractory solid tumors. This information will allow for identification <strong>of</strong> the<br />

maximum tolerated dose. <strong>Clinical</strong> exposure levels are being monitored by<br />

measuring the pharmacokinetics <strong>of</strong> QBI-139. Tumor response to QBI-139<br />

will be measured using RECIST criteria. Since QBI-139 demonstrated<br />

broad efficacy in model systems, another outcome <strong>of</strong> the phase I trial may<br />

be identification <strong>of</strong> the indications for the next stage <strong>of</strong> clinical development.<br />

TPS3115 General Poster Session (Board #22A), Mon, 8:00 AM-12:00 PM<br />

BARIS: A phase I trial to evaluate the safety and tolerability <strong>of</strong> combined<br />

BIBF 1120 and RAD001 in solid tumors and to determine the maximum<br />

tolerated dose (MTD) <strong>of</strong> the combination. Presenting Author: Matthias<br />

Scheffler, Lung Cancer Group Cologne, Center for Integrated Oncology,<br />

University Hospital Cologne, Cologne, Germany<br />

Background: Simultaneious inhibition <strong>of</strong> several signallingpathways involved<br />

in angiogenesis as well as in tumor cell growth regulation by kinase<br />

inhibitor combination therapy may increase therapeutic efficacy. Here we<br />

evaluate the combination <strong>of</strong> the mTOR-inhibitor RAD001 (everolimus) and<br />

the triple kinase (FGFR, VEGFR, PDGFR) inhibitor BIBF 1120in a phase I<br />

trial in advanced solid tumors.In addition we use DCE-MRI for early<br />

identification <strong>of</strong> patients with benefit from BIBF 1120. Methods: This is an<br />

open-label, monocentric phase I trial with three dosage arms in a classical<br />

�3�3� design: Patients in arm A receive 5 mg <strong>of</strong> RAD001 and 2 x 150 mg<br />

BIBF 1120, in arm B 10 mg RAD001 and 2 x 150 mg BIBF 1120 will be<br />

administered, whereas in arm C, 10 mg <strong>of</strong> RAD001 and 2 x 200 mg BIBF<br />

1120 will be given. There is no interindividual dose escalation, and the<br />

enrollment <strong>of</strong> the patients will be performed sequentially. Eligible are all<br />

patients with relapsed or refractory advanced/metastatic solid tumors<br />

(UICC stage IV) and an ECOG performance state <strong>of</strong> 0-1 for whom no further<br />

standard therapies are available and who have predefined adequate organ<br />

functions. All patients will start with a 2-week run-in phase <strong>of</strong> 2 x 200 mg<br />

BIBF 1120. Dynamic contrast-enhanced magnetic resonance imaging<br />

(DCE-MRI) scans will be performed at baseline staging, on day 3 and day<br />

14. On day 14, there will also be 12 hours-pharmacokinetic (PK)<br />

assessment. Combination therapy within the forementioned dosage arms<br />

starts on day 15. After two weeks <strong>of</strong> combination therapy, on day 29, a<br />

DCE-MRI scan and 12-hours PK will be performed. Restaging for the<br />

evaluation <strong>of</strong> the efficacy will be performed on day 57. The safety <strong>of</strong> this<br />

combination will be assessed throughout the complete therapy phase using<br />

CTC-AE V4.0, with predefined dose-limiting toxicities (DLTs) being assessed<br />

until day 42. Patients who experience clinical benefit (i.e., response<br />

or stable disease) on day 57 with adequate tolerability <strong>of</strong> the combination<br />

will further receive the medication, as long as the benefit lasts. The trial is<br />

registered under NCT01349296 (clinicaltrials.gov). Recruitment in arm A<br />

has started in February 2012.<br />

Developmental Therapeutics—Experimental Therapeutics<br />

201s<br />

TPS3114 General Poster Session (Board #21H), Mon, 8:00 AM-12:00 PM<br />

Phase Ia/b study <strong>of</strong> combination carboplatin, paclitaxel, and ridaforolimus<br />

in patients with solid, endometrial, and ovarian cancers. Presenting Author:<br />

Hye Sook Chon, H. Lee M<strong>of</strong>fitt Cancer Center & Research Institute, Tampa,<br />

FL<br />

Background: mTOR is a member <strong>of</strong> the PI3K family. Ridaforolimus (RIDA) is<br />

a mTOR inhibitor that has demonstrated tolerability as intravenous (IV) and<br />

oral formulations. PI3K/AKT/mTOR signalling is associated with resistance<br />

to taxanes. In cancer (ca) cells, inhibition <strong>of</strong> mTOR signalling counteracts<br />

AKT-mediated resistance to drugs inhibiting tubulin. Paclitaxel and carboplatin<br />

(PC) have broad activity including endometrial, ovarian, and many<br />

cancers. mTOR inhibition with PC has synergistic and additive effects in<br />

solid tumors. A common defect in endometrioid ca is mutation <strong>of</strong> PTEN,<br />

causing activation <strong>of</strong> the PI3K/AKT/mTOR pathway. RIDA improved PFS<br />

over hormonal therapy <strong>of</strong> metastatic endometrial cancer in a randomized<br />

phase II study (Oza AM, et al. J Clin Oncol. 2011;29 [suppl; abstr 5009]).<br />

In 35 patients in a phase II study <strong>of</strong> RIDA, there were 7.7% partial response<br />

(PR) and 58% with stable disease with median duration 6.6 months<br />

(Mackay H et al. J Clin Oncol. 2011;29 [suppl; abstr 5013]). Ovarian ca<br />

ultimately develops a phenotype resistant to taxanes that may be overcome<br />

with mTOR inhibition. RIDA may therefore potentiate the response <strong>of</strong><br />

endometrial, ovarian, and other solid cancers to PC. Methods: Patients (pts)<br />

with advanced solid ca and measureable disease and up to 3 prior therapies<br />

received P (175mg/m2 IV) and C (AUC 5 to 6 mg/ml/min IV) on day (D)1 <strong>of</strong><br />

each 3-week cycle. RIDA in escalating cohorts <strong>of</strong> 10-40 mg is administered<br />

orally daily for 5 days per week (D1-5, D 8-12, D 15-19) in phase IA<br />

cohorts. Samples for pharmacokinetics <strong>of</strong> RIDA and P and pharmacodynamics<br />

(PD) are collected. More than 1 anticipated DLT <strong>of</strong> thrombocytopenia or<br />

neutropenia in the latter part <strong>of</strong> the cycle shifts the escalation <strong>of</strong> RIDA to an<br />

alternate schedule (D1-5, D 8-12). Escalation continues in a 3X3 design<br />

until 40 mg/day <strong>of</strong> RIDA or MTD. Seven pts have been enrolled; 5 ovarian, 1<br />

endometrial, 1 urethral ca. The second cohort <strong>of</strong> an alternate schedule has<br />

been filled and evaluation continues. Further characterization <strong>of</strong> tolerability,<br />

efficacy, and PD are planned at the MTD in the phase IB expansion<br />

cohorts <strong>of</strong> 15 ovarian/15 endometrial cancers.<br />

TPS3116 General Poster Session (Board #22B), Mon, 8:00 AM-12:00 PM<br />

A dose escalation, single arm, phase Ib/II combination study <strong>of</strong> BEZ235<br />

with everolimus to determine the safety, pharmacodynamics, and pharmacokinetics<br />

in subjects with advanced solid malignancies including glioblastoma<br />

multiforme. Presenting Author: Mohamad Adham Salkeni, University<br />

<strong>of</strong> Cincinnati Cancer Institute, Cincinnati, OH<br />

Background: Downregulation <strong>of</strong> a number <strong>of</strong> signaling pathways, including<br />

the mammalian target <strong>of</strong> rapamycin (mTOR) pathway, has been demonstrated<br />

to be efficacious in a large range <strong>of</strong> solid tumors such as breast,<br />

colon, endometrial, glial and hepatocellular carcinoma (HCC). However, we<br />

find that rapamycins lead to suppression <strong>of</strong> a negative feedback loop from<br />

S6 Kinase 1 (S6K1) to Protein Kinase B (PKB), leading to hyperactivation<br />

<strong>of</strong> PKB. In pre-clinical studies using a mouse model <strong>of</strong> carcinogen-induced<br />

HCC, we have demonstrated that combining BEZ235 (a potent and highly<br />

selective reversible ATP site competitive inhibitor <strong>of</strong> PI3K and mTOR) with<br />

everolimus (an allosteric inhibitor <strong>of</strong> mTOR) synergizes to inhibit tumor<br />

growth. BEZ235, an orally administered agent, has demonstrated preliminary<br />

antitumor activity in a first-in-human phase I study. The current study<br />

will evaluate this combination in patients with a variety <strong>of</strong> solid malignancies<br />

that includes glioblastoma multiforme (GBM). Methods: This study is<br />

divided into a phase 1b portion designed to determine safety <strong>of</strong> increasing<br />

doses <strong>of</strong> the combination, with extensive pharmacokinetics, pharmacodynamics<br />

and pharmacogenomics analysis; and a phase 2 portion that<br />

includes both solid tumors and GBM based on predominance <strong>of</strong> the mTOR<br />

and PI3K deregulation in these tumors, to determine preliminary antitumor<br />

activity and the recommended dose for phase 2 studies. We will also<br />

integrate biomarker assessment for gene expression products <strong>of</strong> the mTOR<br />

downstream pathway such as eukaryotic initiation factor 4E binding protein<br />

(4EBP1) and S6 kinase (S6K). The phase 1b portion has started accruing.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


202s Developmental Therapeutics—Experimental Therapeutics<br />

TPS3117 General Poster Session (Board #22C), Mon, 8:00 AM-12:00 PM<br />

A phase I study determining the safety and tolerability <strong>of</strong> combination<br />

therapy with pazopanib, a VEGFR/PDGFR/raf inhibitor, and GSK1120212,<br />

a MEK inhibitor, in advanced solid tumors enriched with patients with<br />

advanced differentiated thyroid cancer. Presenting Author: Shabina Roohi<br />

Ahmed, The Johns Hopkins University School <strong>of</strong> Medicine, Baltimore, MD<br />

Background: Mutations <strong>of</strong> the RAS/RAF/MEK/ERK signaling pathway,<br />

especially RAS, BRAF and IGF-I/II receptor genes play a critical role in the<br />

development <strong>of</strong> many different types <strong>of</strong> cancers, including breast, colorectal,<br />

melanoma, NSCLC, and thyroid. Differentiated thyroid cancer (DTC) is<br />

the most common endocrine malignancy, but there is currently no approved<br />

therapy for advanced radioiodine-resistant disease. In DTC xenograft<br />

models, vertical inhibition <strong>of</strong> this pathway, achieved by targeting the<br />

vascular endothelial growth factor receptor (VEGFR) and MEK, has shown<br />

greater clinical efficacy than with either agent alone. Pazopanib (P) is a<br />

small molecule tyrosine kinase inhibitor that selectively inhibits VEGFR1-3,<br />

PDGFR-�, PDGFR-b, c-kit, and FGFR 1-3. Phase II data in advanced DTC<br />

from Bible, et al, indicate a 49% response rate with a PFS <strong>of</strong> 11.7 months<br />

in a patients with PD within 6 months. GSK1120212 (G) is a potent, highly<br />

selective, allosteric inhibitor <strong>of</strong> MEK1/2. In a phase I/II study, Infante et al.<br />

reported an ORR <strong>of</strong> 81% in BRAF mutant melanoma patients when G was<br />

combined with a RAF inhibitor. Methods: A phase I, open label, dose<br />

escalation trial with P�G is currently accruing pts with advanced malignancies.<br />

The study is a standard 3�3 design with an expansion cohort <strong>of</strong> 25 pts<br />

with advanced DTC for correlative endpoints and PK studies. Pts will be<br />

treated with P at 400 mg QD, 600 mg QD and 800 mg QD, with G held<br />

constant at 1 mg QD; G will then be escalated to 1.5 mg QD and 2 mg QD.<br />

Eligibility includes advanced solid tumor, good end organ function and<br />

performance status, and PD within 6 months in the expansion cohort. The<br />

primary objective is to determine the safety and tolerability <strong>of</strong> the<br />

combination and determine the MTD. Secondary objectives include assessing<br />

preliminary efficacy as defined by RECIST criteria and PFS, and<br />

exploration <strong>of</strong> PK/PD endpoints. Serial tumor biopsies will be evaluated for<br />

changes in signaling through p-ERK. RAS and RAF mutation sites will be<br />

sequenced in order to correlate with PD endpoints and disease response.<br />

Currently, DL1 has accrued with no DLTs.<br />

TPS3118 General Poster Session (Board #22D), Mon, 8:00 AM-12:00 PM<br />

Phase lb combination trial <strong>of</strong> a MEK inhibitor, pimasertib (MSC1936369B),<br />

and a PI3K/mTOR inhibitor, SAR245409, in patients with locally advanced<br />

or metastatic solid tumors. Presenting Author: Jeffrey R. Infante,<br />

Sarah Cannon Research Institute, Nashville, TN<br />

Background: The PI3K/mTOR and MAPK signaling pathways are frequently<br />

aberrantly activated in tumors and interact to promote growth and resistance<br />

to treatment. Simultaneous inhibition <strong>of</strong> both pathways may enhance<br />

antitumor activity. This trial investigates the combination <strong>of</strong> pimasertib, a<br />

highly selective MEK1/2 inhibitor, and SAR245409, a dual PI3K/mTOR<br />

inhibitor (<strong>Clinical</strong>Trials.gov NCT01390818). Maximum tolerated dose<br />

(MTD) when administered once daily (qd) as a single agent to solid tumor<br />

patients (pts) is 90 mg for both compounds. Methods: Pts with advanced<br />

solid tumors characterized by frequent alterations <strong>of</strong> the MAPK or PI3K/<br />

mTOR pathways (pancreatic, thyroid, colorectal, non-small cell lung,<br />

endometrial, renal, breast, ovarian and melanoma) and/or with identified<br />

alterations in genes activating these pathways, adequate performance<br />

status and organ function, and no retinal disease are eligible for entry. The<br />

primary objective <strong>of</strong> the trial is to determine the MTD <strong>of</strong> the combination<br />

therapy. Secondary objectives include: safety, pharmacokinetics (PK),<br />

pharmacodynamics (PD), biomarker identification and antitumor efficacy<br />

(response rate via RECIST v1.1). Both compounds are dosed together qd<br />

continuously in 21 day cycles. The study uses a classical 3 � 3 design, with<br />

modified Fibonacci (decreasing increments) dose escalation based on<br />

occurrence <strong>of</strong> dose-limiting toxicities (DLTs). In parallel with dose escalation,<br />

more pts may be enrolled in already tested lower dose level (DL)<br />

cohorts to further evaluate PK, PD, safety and antitumor activity. After MTD<br />

confirmation, additional pts may be enrolled in up to 4 disease-specific<br />

cohorts based on scientific rationale, and observed preclinical and clinical<br />

activity signals. As <strong>of</strong> January 25th 2012, 20 pts have been treated. No<br />

DLTs have been reported.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


LBA3500^ Oral Abstract Session, Sun, 9:45 AM-12:45 PM<br />

Bevacizumab (Bev) with or without erlotinib as maintenance therapy,<br />

following induction first-line chemotherapy plus Bev, in patients with<br />

metastatic colorectal cancer (mCRC): Efficacy and safety results <strong>of</strong> the<br />

international GERCOR DREAM phase III trial. Presenting Author: Christophe<br />

Tournigand, Hôpital Saint-Antoine, Paris, France<br />

The full, final text <strong>of</strong> this abstract will be available at<br />

abstract.asco.org at 12:01 AM (EDT) on Sunday, June 3,<br />

2012, and in the <strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong> online<br />

supplement to the June 20, 2012, issue <strong>of</strong> Journal <strong>of</strong><br />

<strong>Clinical</strong> Oncology. Onsite at the <strong>Meeting</strong>, this abstract will<br />

be printed in the Sunday edition <strong>of</strong> ASCO Daily News.<br />

3502 Oral Abstract Session, Sun, 9:45 AM-12:45 PM<br />

Phase III CORRECT trial <strong>of</strong> regorafenib in metastatic colorectal cancer (mCRC).<br />

Presenting Author: Eric Van Cutsem, Digestive Oncology Unit, Leuven Cancer<br />

Institute, University Hospital Gasthuisberg, Leuven, Belgium<br />

Background: Regorafenib (REG) is an oral multi-kinase inhibitor. The CORRECT<br />

trial was conducted to evaluate REG in patients (pts) with mCRC who had<br />

progressed after all approved standard therapies. Methods: Enrollment criteria<br />

included documented mCRC and progression during or �3 months after last<br />

standard therapy. Pts were randomized 2:1 to receive best supportive care plus<br />

either REG (160 mg od po, 3 wks on/1 wk <strong>of</strong>f) or placebo (PL). The primary<br />

endpoint was overall survival (OS). Secondary endpoints included progressionfree<br />

survival (PFS), overall response rate, disease control rate, safety and quality<br />

<strong>of</strong> life (QoL). Efficacy analyses across prespecified subgroups were evaluated<br />

using univariate Cox regression. Results: 760 pts were randomized (REG: 505;<br />

PL: 255). The OS primary endpoint was met at a preplanned interim analysis. OS<br />

and PFS were significantly improved in REG arm compared to PL arm: hazard<br />

ratio (HR) for OS 0.77 (95% CI 0.64-0.94, 1-sided p�0.0052), median OS 6.4<br />

vs 5.0 mos; HR for PFS 0.49 (95% CI 0.42-0.58, 1-sided p�0.000001),<br />

median PFS 1.9 vs 1.7 mos. Comparable OS and PFS benefits were observed in<br />

exploratory subgroup analyses by region, age, time from diagnosis <strong>of</strong> mCRC to<br />

randomization, prior lines <strong>of</strong> treatment, and KRAS status (shown in table). The<br />

most common grade 3� AEs related to REG were hand-foot skin reaction<br />

(16.6%), fatigue (9.6%), hypertension (7.2%), diarrhea (7.2%) and rash/<br />

desquamation (5.8%). QoL data will be presented. Conclusions: REG demonstrated<br />

statistically significant improvement in OS and PFS over PL, as well as<br />

comparable efficacy benefits across pt subgroups analyzed.<br />

OS PFS<br />

Subgroups N HR 95% CI HR 95% CI<br />

Region*<br />

North America, Western EU, 632 0.77 0.62-0.95 0.50 0.42-0.60<br />

Israel and Australia<br />

Asia 104 0.79 0.43-1.46 0.43 0.28-0.68<br />

Age<br />

< 65 yrs 475 0.72 0.56-0.91 0.42 0.34-0.51<br />

> 65 yrs 285 0.86 0.61-1.19 0.65 0.50-0.86<br />

Time from Dx <strong>of</strong> mCRC to randomization<br />

< 18 mo 140 0.82 0.53-1.25 0.58 0.41-0.84<br />

> 18 mo 620 0.76 0.61-0.95 0.48 0.40-0.58<br />

No. <strong>of</strong> prior treatment lines on or after metastatic disease<br />

< 3 395 0.79 0.60-1.04 0.53 0.43-0.67<br />

> 3 365 0.75 0.56-0.99 0.47 0.37-0.59<br />

KRAS ^<br />

WT 299 0.65 0.48-0.90 0.48 0.36-0.62<br />

MUT 430 0.87 0.67-1.12 0.53 0.43-0.65<br />

* 24 pts from other regions not shown. ^ Historical record.<br />

Gastrointestinal (Colorectal) Cancer<br />

203s<br />

LBA3501 Oral Abstract Session, Sun, 9:45 AM-12:45 PM<br />

Results <strong>of</strong> the X-PECT study: A phase III randomized double-blind<br />

placebo-controlled study <strong>of</strong> perifosine plus capecitabine (P-CAP) versus<br />

placebo plus capecitabine (CAP) in patients (pts) with refractory metastatic<br />

colorectal cancer (mCRC). Presenting Author: Johanna C. Bendell, SCRI/<br />

Tennessee Oncology, Nashville, TN<br />

The full, final text <strong>of</strong> this abstract will be available at<br />

abstract.asco.org at 12:01 AM (EDT) on Sunday, June 3,<br />

2012, and in the <strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong> online<br />

supplement to the June 20, 2012, issue <strong>of</strong> Journal <strong>of</strong><br />

<strong>Clinical</strong> Oncology. Onsite at the <strong>Meeting</strong>, this abstract will<br />

be printed in the Sunday edition <strong>of</strong> ASCO Daily News.<br />

CRA3503 Oral Abstract Session, Sun, 9:45 AM-12:45 PM<br />

Bevacizumab (BEV) plus chemotherapy (CT) continued beyond first progression<br />

in patients with metastatic colorectal cancer (mCRC) previously<br />

treated with BEV plus CT: Results <strong>of</strong> a randomized phase III intergroup<br />

study (TML study). Presenting Author: Dirk Arnold, Hubertus Wald Tumor<br />

Center, University Cancer Center Hamburg (UCCH), Hamburg, Germany<br />

The full, final text <strong>of</strong> this abstract will be available at<br />

abstract.asco.org at 12:01 AM (EDT) on Sunday, June 3,<br />

2012, and in the <strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong> online<br />

supplement to the June 20, 2012, issue <strong>of</strong> Journal <strong>of</strong><br />

<strong>Clinical</strong> Oncology. Onsite at the <strong>Meeting</strong>, this abstract will<br />

be printed in the Sunday edition <strong>of</strong> ASCO Daily News.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


204s Gastrointestinal (Colorectal) Cancer<br />

3504 Oral Abstract Session, Sun, 9:45 AM-12:45 PM<br />

Final analysis <strong>of</strong> the phase III randomized trial <strong>of</strong> cetuximab (CET) plus<br />

either brivanib alaninate (BRIV) or placebo in patients (pts) with chemotherapy<br />

refractory, K-RAS wild-type (WT), metastatic colorectal carcinoma<br />

(mCRC): The NCIC <strong>Clinical</strong> Trials Group and AGITG CO.20 trial. Presenting<br />

Author: Lillian L. Siu, Princess Margaret Hospital and University <strong>of</strong> Toronto,<br />

Toronto, ON, Canada<br />

Background: The anti-EGFR monoclonal antibody CET has improved survival<br />

in pts with chemotherapy refractory, K-RAS WT mCRC. BRIV is a<br />

potent inhibitor <strong>of</strong> multiple receptor tyrosine kinases including both VEGFR<br />

and FGFR. The combination <strong>of</strong> CET and BRIV targets tumor growth and<br />

angiogenesis and demonstrated encouraging activity in an early phase<br />

clinical trial. Methods: Pts with mCRC previously treated with combination<br />

chemotherapy were randomized 1:1 to receive CET 400 mg/m2 IV loading<br />

dose followed by weekly maintenance <strong>of</strong> 250 mg/m2 plus either BRIV 800<br />

mg PO daily (Arm A) or placebo (Arm B). Pts may have had 1 prior<br />

anti-VEGF, but no prior anti-EGFR therapy. Primary endpoint was overall<br />

survival (OS). Results: From 02/2008 to 02/2011, 750 pts were randomized<br />

(376 in Arm A and 374 in Arm B). Demographics: median age�64<br />

(range 27-88); male�64%; ECOG 0:1:2 (%)�32:58:10; �3 prior chemotherapy<br />

regimens�92%; prior anti-VEGF therapy�41%; K-RAS WT�97%.<br />

Primary analysis was conducted per protocol after 536 deaths were<br />

observed, with median OS <strong>of</strong> 8.8 months in Arm A and 8.1 months in Arm<br />

B, hazard ratio (HR)�0.88; 95% CI�0.74 to 1.03; p�0.12. Median<br />

progression-free survival (PFS) was 5.0 months in Arm A and 3.4 months in<br />

Arm B, HR�0.72; 95% CI�0.62 to 0.84; p�0.0001. Incidence <strong>of</strong> any<br />

�grade 3 adverse event (AE) was 78% in Arm A and 53% in Arm B. Time to<br />

deterioration <strong>of</strong> physical function was shorter and global quality <strong>of</strong> life<br />

scores were lower in Arm A vs Arm B. Planned subgroup analyses revealed<br />

no statistically difference in treatment effects on OS based on pre-specified<br />

factors <strong>of</strong> age, gender, ECOG and race. Likewise, no difference was<br />

detected based on exploratory subgroup analyses <strong>of</strong> LDH and prior<br />

anti-VEGF therapy. Conclusions: Despite positive effects on PFS, the<br />

combination <strong>of</strong> CET�BRIV did not significantly improve OS in pts with<br />

chemotherapy refractory, K-RAS WT mCRC. Final updated results based on<br />

20-25% additional events for a total <strong>of</strong> nearly 700 deaths, as well as<br />

further exploratory subgroup analyses, will be presented.<br />

3506 Oral Abstract Session, Sun, 9:45 AM-12:45 PM<br />

FOLFOX4 (12 cycles) versus sequential dose-dense FOLFOX7 (6 cycles)<br />

followed by FOLFIRI (6 cycles) in patients with initially resectable<br />

metastatic colorectal cancer: A GERCOR randomized phase III study<br />

(MIROX). Presenting Author: Mohamed Hebbar, Medical Oncology Unit -<br />

Hôpital Huriez, Lille, France<br />

Background: Perioperative FOLFOX4 is the standard chemotherapy (CT) in<br />

patients with resectable metastases from colorectal cancer (CRC) (Nordlinger<br />

et al, Lancet 2008). To increase curability and reduce the risk <strong>of</strong><br />

severe neuropathy, we evaluated the sequential administration <strong>of</strong> a<br />

dose-dense oxaliplatin-based regimen followed by an irinotecan-based<br />

regimen. Methods: Patients with CRC and initially resectable or resected<br />

metastases were randomized between arm A (control): FOLFOX4 (12<br />

cycles; oxaliplatin 85 mg/m²) or arm B (investigational): FOLFOX7 (6<br />

cycles; oxaliplatin 130 mg/m²) followed by FOLFIRI (6 cycles; irinotecan<br />

180 mg/m²). CT was either perioperative or postoperative. Stratification<br />

criteria were: perioperative vs. postoperative CT, surgery alone vs. radi<strong>of</strong>requency<br />

ablation �/- surgery, Blumgart’s prognostic score (0-1 vs. 2-3 vs.<br />

4-5). Only one metastatic site was allowed, but the number <strong>of</strong> metastases<br />

per organ was not limited. The primary endpoint was disease-free survival<br />

(DFS). Results: From May 2004 to June 2010, 284 patients were<br />

randomized (142 in each arm). Baseline patient characteristics were<br />

similar in both arms. Liver was the main metastatic site. There was only one<br />

metastasis in 70 (49.3%) patients in arm A and 69 (48.6%) patients in<br />

arm B. CT was administered perioperatively in 168 (59.1%) patients, and<br />

postoperatively in 116 (40.9%) patients. In case <strong>of</strong> perioperative CT,<br />

R0-R1 resection was achieved in 58/85 (68.2%) patients in arm A and<br />

67/83 (80.7%) patients in arm B. Grade 3 neuropathy occurred in 34<br />

(23.9%) and 28 (20.0%) patients in arm A and B, respectively. Grade 3/4<br />

thrombocytopenia and gastrointestinal toxicities (nausea, vomiting, diarrhea)<br />

were more frequent in arm B. Median follow-up was 50.4 months<br />

[95% CI: 45.6-54.4]. Median DFS were 22.4 months [95% CI: 17.9-36.2]<br />

in arm A and 23.0 months [95% CI: 19.7-35.6] in arm B (HR�0.97 [95%<br />

CI: 0.72-1.31]; p�.856). Conclusions: FOLFOX7 followed by FOLFIRI is<br />

not superior to the standard FOLFOX4 chemotherapy in patients with<br />

resectable metastatic colorectal cancer.<br />

3505 Oral Abstract Session, Sun, 9:45 AM-12:45 PM<br />

Effects <strong>of</strong> prior bevacizumab (B) use on outcomes from the VELOUR study:<br />

A phase III study <strong>of</strong> aflibercept (Afl) and FOLFIRI in patients (pts) with<br />

metastatic colorectal cancer (mCRC) after failure <strong>of</strong> an oxaliplatin regimen.<br />

Presenting Author: Carmen Joseph Allegra, University <strong>of</strong> Florida/Shands,<br />

Gainesville, FL<br />

Background: Aflibercept (Afl; also known as VEGF Trap) is a recombinant<br />

human fusion protein that acts as a decoy receptor and prevents the<br />

interaction <strong>of</strong> vascular endothelial growth factor (VEGF)-A, VEGF-B, and<br />

placental growth factor (PlGF) with their receptors. In the phase III<br />

VELOUR study, Afl � FOLFIRI improved overall survival (OS) compared<br />

with FOLFIRI � placebo (pbo) in mCRC (ECCO 2011, abstract 6LBA). We<br />

report outcomes from a pre-specified subgroup analysis by prior B use.<br />

Methods: Pts with mCRC and progression during or after oxaliplatin were<br />

randomized 1:1 to receive either FOLFIRI � pbo or FOLFIRI � Afl 4 mg/kg<br />

IV Q2W with stratification by ECOG performance score (PS, 0v1v2)and<br />

prior B. OS and progression-free survival (PFS) in the prior B-treated pts are<br />

reported as median estimate and hazard ratios (HRs); 95.34% CI for OS<br />

and 95% CI for PFS. Results: Of the 1226 pts in the overall study, 187 in<br />

the pbo and 186 in the Afl group were stratified to prior B. The 2 arms were<br />

well balanced: median age 60 yrs; male 58%; PS 0-1 97%; and 55% �1<br />

metastatic organ. Although not powered for survival, Afl produced a<br />

consistent trend towards prolonged OS and PFS, regardless <strong>of</strong> prior B use,<br />

with no evidence <strong>of</strong> interaction (OS, P�0.7231; PFS, P�0.6954). The<br />

incidence <strong>of</strong> treatment-emergent adverse events in the Afl arm was similar<br />

in pts with prior B (100%) to those without (98.9%), with a similar<br />

incidence <strong>of</strong> grade 3/4 events (82.5% and 83.9%, respectively).<br />

Conclusions: Results <strong>of</strong> this pre-specified subgroup analysis indicate that<br />

adding Afl to FOLFIRI resulted in a consistent trend <strong>of</strong> increased OS and<br />

PFS, regardless <strong>of</strong> prior B use. Prior treatment with B did not appear to<br />

impact the safety pr<strong>of</strong>ile <strong>of</strong> Afl.<br />

Pbo Afl HR<br />

Overall survival<br />

All pts<br />

12.1<br />

13.5<br />

0.817<br />

(N�1,226) (11.07-13.11) (12.52-14.95) (0.713-0.937)<br />

No prior B (N�853) 12.4<br />

13.9<br />

0.788<br />

mo<br />

(11.17-13.54) (12.72-15.64) (0.669-0.927)<br />

Prior B (N�373)<br />

11.7<br />

12.5<br />

0.862<br />

mo<br />

(9.82-13.77) (10.78-15.51) (0.673-1.104)<br />

Progression-free survival<br />

No prior B<br />

5.4<br />

6.9<br />

0.797<br />

mo<br />

(4.53-5.68) (6.37-7.20) (0.679-0.936)<br />

Prior B<br />

3.9<br />

6.7<br />

0.661<br />

mo<br />

(3.02-4.30) (5.75-8.21) (0.512-0.852)<br />

3507 Oral Abstract Session, Sun, 9:45 AM-12:45 PM<br />

Impact on survival <strong>of</strong> primary tumor resection in patients with colorectal<br />

cancer and unresectable metastasis: Pooled analysis <strong>of</strong> individual patients’<br />

data from four randomized trials. Presenting Author: Matthieu Faron,<br />

Institut Gustave Roussy, Villejuif, France<br />

Background: In patients with colorectal cancer (CRC) and unresectable<br />

metastasis, the prognostic impact <strong>of</strong> primary tumor resection still remains a<br />

matter <strong>of</strong> debates. The goal <strong>of</strong> this study was to estimate, after adjustment<br />

for prognostic factors, the effect <strong>of</strong> primary tumor resection on survival.<br />

Methods: Individual patients’ data <strong>of</strong> the 1155 patients with metastatic<br />

CRC included in 4 first-line chemotherapy trials (FFCD 9601, FFCD<br />

2000-05, ACCORD 13 and ML 16987) where retrieved. Patients were<br />

eligible for this study if they had synchronous metastasis judged unresectable.<br />

Primary endpoint was overall survival (OS), secondary endpoint was<br />

progression free survival (PFS). A Cox proportional hazard model stratified<br />

on the trial was used to estimate the impact on survival. Results: 810<br />

patients beginning first-line chemotherapy with either fluoropyrimidine<br />

alone, oxaliplatin, irinotecan and/or bevacizumab were eligible. Patients<br />

with a history <strong>of</strong> resection (n � 478 (59%)), as compared to those without<br />

(n � 332 (41%)), were more likely to have colonic primary (p � 0.0001),<br />

lower carcino embryonic antigen (CEA) (p � 0.0001) or alkaline phosphatase<br />

(ALP) level (p�0.04) and normal white blood cell count (WBC) (p �<br />

0.0001). In the univariate analysis, stratified on the trial, primary tumor<br />

resection was associated with a better OS (Hazard Ratio HR: 0.73<br />

[0.63-0.84]; p � 0.0001) and PFS (HR : 0.73 [0.63-0.84]; p � 0.0001).<br />

Multivariate analysis, adjusted for primary tumor location, CEA, ALP and<br />

WBC levels, OMS performance status and number <strong>of</strong> metastatic sites<br />

confirmed that primary tumor resection was an independent predictor <strong>of</strong><br />

better OS (HR : 0.63 [0.53-0.75] ; p � 0.0001), and PFS (HR : 0.82<br />

[0.70-0.95] ; p � 0.0007). Significant interactions were found between<br />

resection and CEA level (p�0.02) and resection and primary tumor<br />

location (p�0.01) for OS (not for PFS) with a lower impact <strong>of</strong> resection with<br />

higher CEA levels or a colonic primary. Conclusions: This study confirmed<br />

the independent prognostic value on survival <strong>of</strong> primary tumor resection in<br />

patients with unresectable metastases <strong>of</strong> CRC.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


3508 Oral Abstract Session, Sun, 9:45 AM-12:45 PM<br />

EORTC liver metastases intergroup randomized phase III study 40983:<br />

Long-term survival results. Presenting Author: Bernard Nordlinger, Hopital<br />

Ambroise Paré, Boulogne, France<br />

Background: This study evaluates the benefit <strong>of</strong> combining peri-operative<br />

chemotherapy and surgery for patients with liver only metastases from<br />

colorectal cancer (LM) deemed resectable on pre-operative imaging. The<br />

PFS results, the primary endpoint, were reported (ASCO 2007). We now<br />

report on the secondary endpoint overall survival (OS) results, after a<br />

median follow-up <strong>of</strong> 8.5 years. Methods: Between September 2000 and<br />

July 2004, 364 patients (pts) with up to 4 LM were randomized between<br />

peri-operative FOLFOX4 (oxaliplatin 85mg/m² and LV5FU2), 6 cycles<br />

before and 6 cycles after surgery, (CT arm), and surgery alone (S arm). OS<br />

was compared between arms using a 2-sided non-stratified log-rank test at<br />

the 0.05 level <strong>of</strong> significance (intent-to-treat analysis). Results: 182 pts<br />

were randomized in each arm (CT arm and S arm) <strong>of</strong> which 171 were<br />

eligible (CT and S) and 152 underwent resection (CT and S). In the CT arm,<br />

171 received preop CT and 115 received postop CT. At a median follow-up<br />

<strong>of</strong> 8.5 years, 221 deaths were reported (61 % <strong>of</strong> all randomized pts).<br />

Causes <strong>of</strong> death in CT arm were cancer relapse (85 pts), complication <strong>of</strong><br />

surgery (2 pts), other causes (16 pts) and unknown cause (4 pts), and in S<br />

arm, cancer relapse (99 pts), complication <strong>of</strong> surgery (2 pts), other causes<br />

(11 pts) and unknown cause (2 pts). Observed median OS was higher in CT<br />

arm: 61 months (CT) vs. 54 months (S) in all randomized pts and 64<br />

months (CT) vs. 55 months (S) in eligible pts. The 5 year OS rate was<br />

51.2% (CT) vs. 47.8% (S) (all randomized) and 52.4% (CT) vs. 48.3% (S)<br />

(eligible pts). There was no significant difference in OS between arms<br />

(HR�0.88, 95% CI 0.68-1.14, p�0.34) (all randomized). After progression,<br />

second line treatment with chemotherapy was given in 59 % (CT) and<br />

77% (S) <strong>of</strong> the pts with cancer progression and repeat surgery in 46% (CT)<br />

and 40% (S), respectively. Conclusions: Peri-operative chemotherapy with<br />

FOLFOX4 improves PFS but does not significantly improve OS over surgery<br />

alone. A potential benefit may have been diluted by second treatment lines<br />

and by more deaths unrelated to cancer in the CT arm.<br />

3510 Poster Discussion Session (Board #2), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Validation <strong>of</strong> a genomic classifier (ColoPrint) for predicting outcome in the<br />

T3-MSS subgroup <strong>of</strong> stage II colon cancer patients. Presenting Author:<br />

Ramon Salazar, Institut Catala d’Oncologia, Barcelona, Spain<br />

Background: Adjuvant therapy for stage II patients is recommended for<br />

patients with high risk features, especially with T4 tumors. Adjuvant<br />

therapy is not indicated for patients with MSI-H status who are considered<br />

<strong>of</strong> being at low risk <strong>of</strong> disease relapse. However, this leaves the majority <strong>of</strong><br />

patients with an undetermined risk. ColoPrint is an 18-gene expression<br />

classifier that identifies early-stage colon cancer patients at higher risk <strong>of</strong><br />

disease relapse. Methods: ColoPrint was developed using whole genome<br />

expression data and was validated in public datasets (n�322) and<br />

independent patient cohorts from 5 European hospitals. Tissue specimen,<br />

clinical parameters, MSI-status and follow-up data (median follow-up 70<br />

months) for patients were available and the ColoPrint index was determined<br />

using validated diagnostic arrays. Uni-and multivariate analysis was performed<br />

on the pooled stage II patient set (n�320) and the subset <strong>of</strong><br />

patients who were T3/ MSS (n�227). Results: In the analysis <strong>of</strong> all stage II<br />

patients, ColoPrint classified two-third <strong>of</strong> stage II patients as being at lower<br />

risk. The 3-year Relapse-Free-Survial (RFS) RFS was 91% for Low Risk and<br />

74% for patients at higher risk with a HR <strong>of</strong> 2.9 (p�0.001). Clinicopathological<br />

parameters from the ASCO recommendations (T4, perforation, �12<br />

LN assessed, and/ or high grade) or NCCN guidelines (ASCO factors plus<br />

angio-lymphatic invasion) did not predict a differential outcome for high<br />

risk patients (p� 0.20). In the subgroup <strong>of</strong> patients with T3 and MSS<br />

phenotype, ColoPrint classified 61% <strong>of</strong> patients at lower risk with a 3-year<br />

RFS <strong>of</strong> 91% (86-96%) and 39% <strong>of</strong> patients at higher risk with a 3-year RFS<br />

<strong>of</strong> 73% (63-83%) (p�0.002). No clinical parameter was significantly<br />

prognostic in this subgroup. Conclusions: ColoPrint combined with established<br />

clinicopathological factors and MSI, significantly improves prognostic<br />

accuracy, thereby facilitating the identification <strong>of</strong> patients at higher risk<br />

who might be considered for additional treatment.<br />

Gastrointestinal (Colorectal) Cancer<br />

3509 Poster Discussion Session (Board #1), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Validation <strong>of</strong> two gene-expression risk scores in a large colon cancer cohort<br />

and contribution to an improved prognostic method. Presenting Author:<br />

Arnaud Roth, University Hospital Geneva, Geneva, Switzerland<br />

Background: Prognosis prediction for resected primary colon cancer is<br />

currently based on the tumor, nodes, metastasis (TNM) staging system.<br />

Gene expression based risk scores have been proposed, but need to be<br />

validated and integrated with clinical and TNM variables. We performed an<br />

independent assessment <strong>of</strong> the individual recurrence signatures developed<br />

for commercial use by Veridex and Genomic Health. Methods: The Veridex<br />

(aVDS) and Genomic Health (aGHS) risk scores were applied to existing<br />

gene expression data <strong>of</strong> 580 stage III and 108 stage II tumors from the<br />

PETACC-3 trial. Association <strong>of</strong> the scores with relapse-free (RFS) and<br />

overall survival (OS) <strong>of</strong> the patients was assessed singularly, and in a<br />

combination with TNM and other clinico-pathological variables, by univariate<br />

and multivariate Cox regression models and logrank methods. Results:<br />

Both risk scores were significantly associated with RFS in univariate and<br />

multivariate models (Table) for the stage III cohort. The scores contributed<br />

different and additive prognostic information, and reached highest effect<br />

sizes (approximate p-value 0.001 and HR per interquartile range 1.4 each)<br />

in a combined model (Table) that includes both scores as well as T-stage,<br />

N-stage and MSI status as significant factors. Analysis in the stage II cohort<br />

gave similar effect estimates. Conclusions: This study confirms in an<br />

independent colon cancer patient cohort that gene expression based risk<br />

scores improve current prognostic models. The best prognostic model was<br />

obtained by using clinico-pathological variables and both gene-expression<br />

risk-scores.<br />

aGHS aVDS<br />

RFS OS RFS OS<br />

Model HR (95% CI) p value HR (95% CI) p value HR (95% CI) p value HR (95% CI) p value<br />

Univariate 1.32(1.10 - 1.58) 0.0023 1.35(1.10 - 1.65) 0.0043 1.25(1.05 - 1.50) 0.0130 1.22(0.99 - 1.50) 0.0571<br />

Multivariate 1.24(1.04 - 1.49) 0.0190 1.27(1.03 - 1.58) 0.0255 1.28(1.05 - 1.55) 0.0130 1.25(1.00 - 1.56) 0.0500<br />

Combined<br />

(aGHS<br />

� aVDS)<br />

1.36(1.13 - 1.65) 0.0014 1.40(1.11 - 1.75) 0.0037 1.41(1.15 - 1.73) 0.0011 1.38(1.09 - 1.75) 0.0074<br />

Hazard ratios for 1 interquartile range variation.<br />

205s<br />

3511 Poster Discussion Session (Board #3), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Identification and validation <strong>of</strong> gene expression subtypes in a large set <strong>of</strong><br />

colorectal cancer samples. Presenting Author: Eva Budinska, Swiss Institute<br />

<strong>of</strong> Bioinformatics, Lausanne, Switzerland<br />

Background: From a clinical perspective colorectal cancer (CRC) is a<br />

heterogeneous disease whose biological background is insufficiently understood.<br />

In order to adapt targeted treatment to biologically different<br />

categories <strong>of</strong> CRC patients, in depth understanding <strong>of</strong> the molecular<br />

mechanisms involved in CRC heterogeneity is urgently needed. Methods:<br />

Consensus cluster analysis <strong>of</strong> 1113 stage II-III CRC gene expression<br />

pr<strong>of</strong>iles from PETACC3 and 4 public datasets defined a set <strong>of</strong> subtypes<br />

which were confirmed in an independent set <strong>of</strong> 720 samples. The similarity<br />

between tumors was based on a set <strong>of</strong> 54 meta-genes. This approach<br />

improves the robustness to measurement noise and gives equal chances to<br />

each biological process to be accounted for in the subtype definition. We<br />

tested the association <strong>of</strong> the subtypes with clinical variables, molecular<br />

markers and patient survival. Results: We identified and validated 5 major<br />

subtypes A-E, with different levels <strong>of</strong> expression in 6 main molecular<br />

processes: epithelial-mesenchymal transition (EMT), immune response,<br />

colon crypt differentiation, proliferation, Wnt signaling and chromosome<br />

20q. Significant differences in survival and an enrichment for markers such<br />

as MSI, BRAF mutation, site or p53 status were found between the<br />

subtypes. In addition, the new subtype classification uncovered heterogeneity<br />

within groups defined by these commonly used markers. Survival<br />

analysis showed significant prognostic value for meta-genes connected to<br />

EMT, proliferation and differentiation. Identical data processing and<br />

clustering applied to the validation set on the same meta-genes resulted in<br />

subtypes with almost identical expression patterns. Conclusions: We<br />

identified and validated robust molecular subtypes in the largest set <strong>of</strong><br />

stage II-III CRC samples as a combination <strong>of</strong> multiple molecular processes,<br />

that complement current disease stratification based on clinico-pathological<br />

variables and molecular markers. The biological relevance <strong>of</strong> these<br />

subtypes was reflected in significant differences in survival. These insights<br />

open new perspectives for improving prognostic models and rationalize the<br />

prediction <strong>of</strong> tumor-specific drug sensitivity.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


206s Gastrointestinal (Colorectal) Cancer<br />

3512 Poster Discussion Session (Board #4), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Validation <strong>of</strong> the 12-gene colon cancer recurrence score (RS) in NSABP<br />

C07 as a predictor <strong>of</strong> recurrence in stage II and III colon cancer patients<br />

treated with 5FU/LV (FU) and 5FU/LV�oxaliplatin (FU�Ox). Presenting<br />

Author: Michael O’Connell, National Surgical Adjuvant Breast and Bowel<br />

Project, Pittsburgh, PA<br />

Background: Standardized clinical tools which accurately quantify recurrence<br />

risk are needed for optimal adjuvant treatment <strong>of</strong> colon cancer. The<br />

12-gene RS has been validated in stage II colon cancer pts from QUASAR<br />

and CALGB 9581. We conducted a large prospectively-designed clinical<br />

validation study <strong>of</strong> RS, w/ pre-specified endpoints, methods, and analysis<br />

plan, in stage II and III colon cancer pts randomized to FU or FU�Ox in<br />

NSABP C-07. Methods: 50% <strong>of</strong> C-07 pts w/ tissue were randomly selected,<br />

stratified on stage (AJCC 6th ) and recurrence. Gene expression was<br />

quantitated by RT-PCR on 25 �m manually microdissected fixed colon<br />

tumor tissue. Data were analyzed by Cox regression controlling for stage and<br />

treatment (TRT). Results: RT-PCR was successful in 892/921 pts (97%):<br />

449 FU, 443 FU � Ox; 264 st II, 409 st IIIA/B, 219 st IIIC. The primary<br />

endpoint was met: RS predicted recurrence (HR/25 units�1.96, 95% CI<br />

1.50-2.55 p�.001). RS also predicted disease-free survival (p�.001) and<br />

overall survival (p�.001). RS predicted recurrence (p�.001) independent<br />

<strong>of</strong> T and N stage, MMR, nodes examined, grade, and TRT. Predefined high<br />

RS group (26% <strong>of</strong> pts) had higher recurrence risk than low RS group (39%<br />

<strong>of</strong> pts): HR�2.11, p�.001. Cox model 5 yr recurrence risk (95%CI) in FU<br />

treated pts by RS group (low, int, high): st II 9% (6-13%), 13% (8-17%),<br />

18% (12-25%); st IIIA/B 21% (16-26%), 29% (24-34%), 38% (30-<br />

46%); st IIIC 40% (32-48%), 51% (43-59%), 64% (55-74%). RS did not<br />

have significant interaction w/ stage (p�0.90) or age (p�0.76). Relative<br />

benefit <strong>of</strong> Ox was similar across range <strong>of</strong> RS (interaction p�0.48);<br />

accordingly, in Cox model and Kaplan-Meier analyses, absolute benefit <strong>of</strong><br />

Ox increased w/ higher RS. Conclusions: RS predicts recurrence risk in<br />

stage II and III colon cancer, capturing underlying biology and providing<br />

risk information beyond conventional factors. RS is not predictive <strong>of</strong><br />

relative benefit <strong>of</strong> Ox added to adjuvant FU but enables better discrimination<br />

<strong>of</strong> absolute Ox benefit as a function <strong>of</strong> risk. Incorporating RS into the<br />

clinical context may better inform adjuvant therapy decisions for pts w/<br />

stage III as well as stage II colon cancer.<br />

3514 Poster Discussion Session (Board #6), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Prognostic impact <strong>of</strong> BRAF and KRAS mutations and their relationship to<br />

DNA mismatch repair (MMR) status in 2,686 stage III colon cancer<br />

patients (pts) treated in a phase III study <strong>of</strong> adjuvant FOLFOX with or<br />

without cetuximab: NCCTG N0147. Presenting Author: Frank A. Sinicrope,<br />

Mayo Clinic, Rochester, MN<br />

Background: Activating mutations in the BRAF oncogene (mutBRAF) are<br />

associated with deficient MMR (dMMR) in sporadic colon cancers and are<br />

mutually exclusive with KRAS mutations (mutKRAS). We evaluated BRAF<br />

and KRAS in relation to MMR and disease-free survival (DFS) in stage III<br />

pts treated with adjuvant FOLFOX �/-cetuximab; study was amended<br />

mid-way to require prospective KRAS testing (Alberts SR, ASCO 2010).<br />

Methods: Extracted DNA from archival tumors was analyzed for BRAF exon<br />

15 (V600E) and KRAS exon 2 mutations by allele specific RT-PCR. Tumors<br />

with loss <strong>of</strong> any MMR protein (MLH1, MSH2, MSH6) were categorized as<br />

dMMR vs pr<strong>of</strong>icient (pMMR). DFS was censored at 4 yrs; median follow-up<br />

was 3.1 yrs. Results: Among 2,686 pts, 12% (314/2580) <strong>of</strong> tumors were<br />

dMMR; 28% (716/2579) had mutKRAS, and 14% (346/2515) had<br />

mutBRAF <strong>of</strong> which 43% (150/346) were dMMR. mutBRAF was associated<br />

with high grade, proximal site, T3-4, �4 LNs, and dMMR (all p�0.002);<br />

mutKRAS with low grade, proximal, �3 LNs, and pMMR (all p�0.02).<br />

Among dMMR tumors, mutKRAS was more frequent in distal vs proximal<br />

cancers (28% vs. 8%, p�0.0004). Both mutKRAS (HR�1.42, p�0.0001)<br />

and mutBRAF (HR�1.34, p�0.02) were associated with worse DFS and<br />

remained significant after adjustment for treatment, MMR, grade, site, T<br />

stage and LNs. mutBRAF (p�0.003) and mutKRAS (p�0.003) were each<br />

prognostic for DFS in pMMR, but not dMMR tumors (KRAS, p�0.79;<br />

BRAF, p�0.60). While MMR was not significant univariately, proximal<br />

dMMR tumors had favorable DFS [HR 0.8, 95% CI (0.6-1)] and distal<br />

dMMR tumors had poor DFS [HR 2 (1.1-3.4)] after adjusting for KRAS,<br />

BRAF, grade, T stage and LNs (pinteraction�0.02). pMMR/mutBRAF<br />

(p�0.027) and pMMR/mutKRAS (p�0.001) had worse DFS vs pMMR/<br />

bothWT. Conclusions: mutBRAF and mutKRAS were significantly associated<br />

with worse DFS independent <strong>of</strong> treatment, covariates, and each other,<br />

with the DFS impact restricted to pMMR tumors. Prognostic impact <strong>of</strong><br />

MMR was dependent upon tumor site after adjusting for covariates.<br />

Support: NIH Grant CA 25224, NCI K05CA 142885, Bristol-Myers<br />

Squibb, ImClone, San<strong>of</strong>i-Aventis, and Pfizer.<br />

3513 Poster Discussion Session (Board #5), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

The prognostic importance <strong>of</strong> miRNA-21 in stage II colon cancer: A<br />

population-based study. Presenting Author: Sanne Kjaer-Frifeldt, Department<br />

<strong>of</strong> Oncology, Vejle Hospital, and Danish Colorectal Cancer Group<br />

South, Vejle, Denmark<br />

Background: Adjuvant chemotherapy for stage II colon cancer patients is<br />

still controversial and the debate on which patients should be considered as<br />

high risk patients is still ongoing. The decision is based on clinical and<br />

pathological markers <strong>of</strong> risk, which are inadequately informative in most <strong>of</strong><br />

the patients, and better methods are highly needed. The aim <strong>of</strong> the present<br />

study was to investigate the possible prognostic importance <strong>of</strong> miRNA-21,<br />

quantified by in situ hybridization (ISH), in a unique, large populationbased<br />

cohort <strong>of</strong> patients treated for stage II colon cancer patients. Methods:<br />

The study included all patients diagnosed with stage II colon cancer in<br />

Denmark in the year 2003 (711 patients), representing a full population <strong>of</strong><br />

five million people. Patients receiving adjuvant chemotherapy were excluded<br />

(N�15). One paraffin-embedded tissue block was obtained from<br />

each patient. A 6�m-thick section was processed for formazan-based<br />

chromogenic miR-21 ISH analysis and counter stained with nuclear red.<br />

The blue miR-21 ISH signal was assessed by image analysis to obtain two<br />

quantitative expression estimates: the total blue area (TB) and the ratio <strong>of</strong><br />

TB with the nuclear density (TBR). Results: The miRNA-21 signal was<br />

predominantly observed in fibroblast-like cells located in the stromal<br />

compartment <strong>of</strong> the tumors. Patients expressing high levels <strong>of</strong> miRNA-21<br />

(high mean TBR) had significantly inferior cancer specific survival (CSS):<br />

HR � 1.26 (95% CI; 1.15-1.60), p �0.001. In the COX regression<br />

analysis (including; gender, T-category, malignancy grade, localization,<br />

tumor perforation, tumor fixation, number <strong>of</strong> lymph nodes and MSI status),<br />

mean TBR was found to be an independent predictive marker <strong>of</strong> poor CSS,<br />

HR � 1.41 (95%CI; 1.19-1.67, p� 0.001). The same applied to TB.<br />

Conclusions: The present study shows that increasing miRNA-21 expression<br />

level is significantly correlated to decreasing CSS. Analyses <strong>of</strong><br />

miRNA-21 should be considered as a potential adjunct in the selection <strong>of</strong><br />

high risk stage II patients.<br />

3515 Poster Discussion Session (Board #7), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

BRAF, PIK3CA, and PTEN status and benefit from cetuximab (CET) in the<br />

treatment <strong>of</strong> advanced colorectal cancer (CRC): Results from NCIC CTG/AGITG<br />

CO.17. Presenting Author: Derek J. Jonker, The Ottawa Hospital Research<br />

Institute, Ottawa, ON, Canada<br />

Background: CET, a monoclonal antibody targeting the epidermal growth factor<br />

receptor, improves overall survival (OS) and progression free survival (PFS) in<br />

patients (pts) with KRAS wild-type (WT) chemotherapy refractory CRC. BRAF<br />

and PIK3CA mutation status, and PTEN expression levels may further predict<br />

benefit from CET therapy. Methods: Available colorectal tumour samples were<br />

analyzed from a phase III trial <strong>of</strong> CET plus best supportive care (BSC) vs BSC<br />

alone (NEJM 2007; 357(20)). BRAF and PIK3CA mutations (MUT) identified in<br />

tumour-derived DNA using a high resolution melting analysis to identify<br />

amplicons with mutations were confirmed by sequencing. PTEN expression by<br />

immunohistochemistry (IHC) was performed on tissue microarrays constructed<br />

from available tumour blocks. For each biomarker, prognostic (treatment<br />

independent) effects were assessed in patients on the BSC alone arm. Predictive<br />

effects (benefit from CET) on OS and PFS among all patients and those in the<br />

KRAS wild-type subset were examined using a Cox model with tests for<br />

treatment-biomarker interaction, adjusting for covariates. Results: Of 401 pts<br />

assessed for BRAF status (70% <strong>of</strong> CO17 population), 13(3%) had mutations. Of<br />

407 pts assessed for PIK3CA status (71% <strong>of</strong> CO17 population), 61(15%) had<br />

mutations. Of 205 pts assessed for PTEN (36% <strong>of</strong> CO17 population), 148(72%)<br />

were negative for IHC expression. No biomarker was prognostic for OS or PFS,<br />

and none were predictive <strong>of</strong> benefit from CET, either in the whole study<br />

population or the KRAS WT subset. Conclusions: In chemotherapy-refractory<br />

CRC, neither PIK3CA mutation status nor PTEN expression were predictive <strong>of</strong><br />

benefit from CET therapy. BRAF mutations are uncommon in this setting. Larger<br />

sample sizes would be required to determine if BRAF status is predictive for CET<br />

benefit.<br />

Prognostic analysis in BSC patients<br />

adj HR (MUT vs WT), [for PTEN, negative vs present]<br />

Biomarker<br />

OS PFS<br />

BRAF 1.47, p�0.41 1.52, p�0.37<br />

PI3KCA 1.11, p�0.65 1.10, p�0.66<br />

PTEN 1.13, p�0.70 0.99, p�0.98<br />

Predictive analysis in KRAS WT subset<br />

adj HR (CET�BSC vs BSC), interaction p value<br />

OS PFS<br />

BRAF 1.39, p�0.69 1.18, p�0.84<br />

PI3KCA 0.79, p�0.63 0.73, p�0.50<br />

PTEN 0.75, p�0.61 2.47, p�0.08<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


3516 Poster Discussion Session (Board #8), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Epiregulin (EREG) and amphiregulin (AREG) gene expression to predict<br />

response to cetuximab therapy in combination with oxaliplatin (Ox) and<br />

5FU in first-line treatment <strong>of</strong> advanced colorectal cancer (aCRC). Presenting<br />

Author: Richard A. Adams, School <strong>of</strong> Medicine, Cardiff University,<br />

Cardiff, United Kingdom<br />

Background: Previous data suggest the EGF ligands EREG/AREG may<br />

predict outcome <strong>of</strong> KRAS wt patients (pts) in the chemo-refractory setting<br />

but has not been previously reported from first line randomised trials.<br />

Methods: FFPE samples from primary tumors <strong>of</strong> pts in Arms A&B <strong>of</strong> the<br />

COIN trial <strong>of</strong> Ox fluoropyrimidine (Fp) �/- cet were analysed for EGFR IHC,<br />

KRAS/NRAS/BRAF mutation and EREG/AREG expression by RT-PCR.<br />

Ligand levels were assessed against baseline data, prognostic markers as<br />

uni/multivariate analyses and as predictive markers in wild type (wt) and<br />

mutant (mt) cohorts and separately by Fp backbone [capecitabine (CapOx)<br />

or 5FU (FOLFOX)]. Tests for interaction were performed with EREG/AREG<br />

continuous, using Flexible Parametric survival analysis. Optimal cut<strong>of</strong>fs for<br />

predictive effects were defined using point at which 95% CI first excluded<br />

zero. Results: 952/1630 (57%) <strong>of</strong> pts were evaluable for all parameters.<br />

EREG/AREG were highly correlated Pearson’s rho (�) � 0.74; p�0.0001.<br />

High EREG/AREG levels were associated with KRAS wt (p�0.005) and<br />

with primary tumor in left colon/rectum, presence <strong>of</strong> liver metastases and<br />

high CEA (p�0.05). In the control arm, high EREG/AREG conferred a<br />

better prognosis among KRAS wt pts in multivariate. EREG superseded<br />

AREG in a combined model. High EREG predicted for OS benefit in KRAS<br />

wt pts treated with FOLFOX �cet, with optimal cut-<strong>of</strong>f 80th centile. OS HR<br />

for �cet �80th centile �0.33, 95% CI 0.14-0.78, p�0.011; �80%<br />

centile HR�0.99, 95% CI 0.67-1.47, p�0.96; interaction p�0.059;<br />

�50th centile HR�0.66, 95% CI 0.40-1.09, p�0.11; �50% centile<br />

HR�1.09, 95% CI 0.66-1.81, p�0.73; interaction p�0.17. Similar<br />

results were seen for PFS, with optimal cut-<strong>of</strong>f 50th centile. There was no<br />

predictive effect for pts treated with CapOx. Conclusions: The data suggest a<br />

prognostic effect <strong>of</strong> EREG/AREG in aCRC. The original hypothesis, that<br />

KRAS wt patients with high EREG expression have improved outcome with<br />

cet, seems to be limited to patients treated with FOLFOX in the first-line<br />

setting.<br />

3518 Poster Discussion Session (Board #10), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Prospective evaluation <strong>of</strong> candidate SNPs <strong>of</strong> VEGF/VEGFR pathway in<br />

metastatic colorectal cancer (mCRC) patients (pts) treated with first-line<br />

FOLFIRI plus bevacizumab (BV). Presenting Author: Chiara Cremolini,<br />

U.O. Oncologia Medica 2, Azienda Ospedaliero-Universitaria Pisana, Istituto<br />

Toscano Tumori, Pisa, Italy<br />

Background: Despite several attempts, no predictors <strong>of</strong> benefit from BV<br />

have been so far identified. We reported the association <strong>of</strong> VEGF rs833061<br />

T/T to a worse outcome in mCRC pts treated with first-line FOLFIRI�BV,<br />

but not in a historical cohort treated with FOLFIRI. Recent post-hoc<br />

analyses on randomized trials suggested the potential prognostic and/or<br />

predictive role <strong>of</strong> other VEGF and VEGFR1/2 SNPs. Methods: Moving from<br />

our retrospective finding, we designed a prospective validation trial in<br />

mCRC pts treated with first-line FOLFIRI� BV to detect a HR for PFS <strong>of</strong> 1.7<br />

for VEGF rs833061 T/T compared to C- variants. With two-sided a�0.05<br />

and b�0.20, 199 events were required. Accrual was faster than expected<br />

and in the meanwhile promising results about other SNPs were reported<br />

and we therefore included a confirmatory analysis <strong>of</strong> VEGF rs699946 A/G,<br />

VEGFR1 rs9582036 A/C and rs7993418 A/G, VEGFR2 rs11133360 C/T,<br />

rs12505758 C/T and rs2305948 C/T and EPAS rs4145836 A/G SNPs.<br />

Germ-line DNA was extracted from peripheral blood. SNPs were analyzed<br />

by PCR and sequencing. Results: Four-hundred-twenty-four pts were<br />

included. At a median follow up <strong>of</strong> 24 months, median PFS was 10.5<br />

months. At the univariate analysis, no differences in PFS according to<br />

VEGF rs833061 C/T variants were observed (p�0.38). Among analyzed<br />

SNPs, only VEGFR2 rs12505758 C- variants (n�118) were associated to<br />

shorter PFS compared to TT (n�306) (HR: 1.40 [95%CI: 1.07-1.84],<br />

p�0.015). In the multivariate model, this association retained significance<br />

(HR: 1.402 [95%CI:1.079-1.822], p�0.012), that was lost by applying<br />

multiple testing correction. Conclusions: This prospective experience failed<br />

to validate the hypothesized predictive impact <strong>of</strong> VEGF rs833061 variants.<br />

Also other previous retrospective findings on different candidate SNPs were<br />

not confirmed. Only VEGFR2 rs12505758 variants, whose prognostic<br />

rather than predictive impact was previously reported, correlated with PFS.<br />

We suggest that future studies on biomarkers <strong>of</strong> benefit from BV should<br />

look at the complexity <strong>of</strong> tumoral angiogenesis at different levels and not<br />

only from the genetic perspective.<br />

Gastrointestinal (Colorectal) Cancer<br />

207s<br />

3517 Poster Discussion Session (Board #9), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

A novel interaction <strong>of</strong> genotype, gender, and adjuvant treatment in survival<br />

after resection <strong>of</strong> stage III colon cancer: Results <strong>of</strong> CALGB 89803.<br />

Presenting Author: Robert S. Warren, University <strong>of</strong> California, San Francisco,<br />

San Francisco, CA<br />

Background: The p53 tumor suppressor is frequently mutated in colon<br />

cancer, but the influence <strong>of</strong> such mutations on survival is remains<br />

undefined. We investigated whether domain-specific mutations in p53 are<br />

predictive <strong>of</strong> survival in stage III colon cancer. Methods: p53 was evaluated<br />

in an intergroup trial (CALGB 89803) <strong>of</strong> patients with stage III colon cancer<br />

who were randomized to receive adjuvant 5-fluorouracil/leucovorin (5FU/<br />

LV) or 5FU/LV with irinotecan (IFL) Tissue was collected to allow correlation<br />

<strong>of</strong> molecular markers with outcomes. p53 was genotyped in 607<br />

patient tumors. Results: p53 mutations were identified in 274 tumors,<br />

divided ~ equally between zinc binding and non-zinc binding regions <strong>of</strong> the<br />

DNA binding domain. Overall, p53 status was not predictive <strong>of</strong> benefit from<br />

either adjuvant regimen. Unexpectedly, the 5 year overall survival (OS) <strong>of</strong><br />

women with tumors harboring non-zinc binding mutations treated with<br />

5FU/LV was 97% compared to OS <strong>of</strong> 72% for women with p53 wild-type<br />

(wt) tumors (p �0.004). Adding irinotecan to 5FU/LV negated this survival<br />

benefit (5 year OS <strong>of</strong> 81% vs. 72%). Conversely, 5 year OS <strong>of</strong> women<br />

harboring tumors with zinc binding mutations who received 5FU/LV was<br />

50% compared to 72% for women with p53 wt tumors (p�0.04). Adding<br />

irinotecan to 5FU/LV reversed the poor survival <strong>of</strong> women with tumors<br />

harboring zinc binding mutations and improved 5 year OS (50% vs. 73%;<br />

p�0.1). No difference in OS was observed for men on either treatment arm<br />

or when genotype was considered. Conclusions: CALGB 89803 demonstrated<br />

a lack <strong>of</strong> survival benefit for stage III colon cancer patients when<br />

irinotecan was added to 5FU/LV (IFL). We now show that in the setting <strong>of</strong> a<br />

large clinical trial, refined stratification <strong>of</strong> women, based upon domainspecific<br />

mutations <strong>of</strong> p53 identifies subsets <strong>of</strong> patients likely to benefit<br />

from, or respond poorly to, adjuvant 5FU/LV. The interaction <strong>of</strong> p53<br />

genotype, gender, and adjuvant therapy regimen has the potential to be<br />

paradigm changing in the treatment <strong>of</strong> colon cancer, and possibly other<br />

malignancies. These data, if validated, suggest that evaluation <strong>of</strong> p53<br />

genotype and gender may guide clinicians to make rational choices <strong>of</strong><br />

adjuvant therapy.<br />

3519^ Poster Discussion Session (Board #11), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Ligand expression <strong>of</strong> the EGFR ligands amphiregulin, epiregulin, and<br />

amplification <strong>of</strong> the EGFR gene to predict for treatment efficacy in KRAS<br />

wild-type mCRC patients treated with cetuximab plus CAPIRI and CAPOX:<br />

Analysis <strong>of</strong> the randomized AIO CRC-0104 trial. Presenting Author:<br />

Sebastian Stintzing, Department <strong>of</strong> Hematology and Oncology, Klinikum<br />

Grosshadern and Comprehensive Cancer Center, LMU Munich, Munich,<br />

Germany<br />

Background: We investigated the expression <strong>of</strong> the EGFR ligands amphiregulin<br />

(AREG) and epiregulin (EREG) as well as the amplification <strong>of</strong> the<br />

EGFR-gene in tumor specimens <strong>of</strong> mCRC patients (pts) treated first-line<br />

with anti-EGFR targeted cetuximab together with CAPOX or CAPIRI.<br />

Expression levels were correlated with overall response rate (ORR), progression<br />

free survival (PFS) and overall survival (OS) to determine their<br />

relationship with effectiveness in this setting. Methods: A total <strong>of</strong> 185<br />

mCRC pts were randomized to cetuximab (400mg/m² day 1, followed by<br />

250mg/m² weekly) plus CAPIRI (irinotecan 200mg/m², day 1; capecitabine<br />

800mg/m² twice daily days 1-14, every 3 weeks; 20% dose reduction <strong>of</strong><br />

both agents for pts older than 65 years) or plus CAPOX (oxaliplatin<br />

130mg/m² day 1; capecitabine 1000mg/m² twice daily days 1-14, every<br />

three weeks). The primary study endpoint was ORR. KRAS mutational<br />

status did not correlate with treatment outcome. The cut-<strong>of</strong>fs for EGFRamplification<br />

using FISH, AREG and EREG levels determined by RT-qPCR<br />

were calculated using ROC analysis for ORR. Results: Within the subgroup<br />

<strong>of</strong> KRAS wildtype tumors, analysis <strong>of</strong> EREG- and AREG-expression was<br />

possible in 99 pts and <strong>of</strong> EGFR-amplification in 63 pts. Higher AREG levels<br />

correlated significantly with higher ORR (83% vs 46%, p�0.006, OR<br />

0.31), longer PFS (9.6mo vs 4.9, p�0.001, HR 0.35) and longer OS<br />

(39.9mo vs 17.2mo, p�0.001, HR 0.36). Higher EREG levels showed a<br />

significant correlation with ORR (74% vs 47%, p�0.036, OR 0.54), longer<br />

PFS (7.9mo vs 4.9mo, p�0.026, HR 0.57) and OS (33.0mo vs 20.2mo,<br />

p�0.041, HR 0.57). EGFR-amplification correlated significantly with<br />

higher ORR (71% vs 33%, p�0.004, OR 0.49), longer PFS (8.4mo vs<br />

4.6mo, p�0.004, HR 0.50) and longer OS (30.5mo vs 15.2mo, p�0.001,<br />

HR 0.44). Conclusions: In the treatment setting <strong>of</strong> cetuximab combined<br />

with CAIPIRI or CAPOX, AREG, EREG and EGFR-amplification predicted<br />

treatment efficacy. Within the subgroup <strong>of</strong> pts with KRAS wildtype tumors,<br />

EGFR-FISH and AREG expression have the strongest relationship with<br />

treatment efficacy.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


208s Gastrointestinal (Colorectal) Cancer<br />

3520 Poster Discussion Session (Board #12), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Effect <strong>of</strong> low-frequency KRAS mutations on the response to anti-EGFR<br />

therapy in metastatic colorectal cancer. Presenting Author: David Tougeron,<br />

Department <strong>of</strong> Gastroenterology, Poitiers University Hospital, Poitiers,<br />

France<br />

Background: Monoclonal antibodies against the epidermal growth factor<br />

receptor (EGFR) are standard components <strong>of</strong> treatment algorithms in<br />

metastatic colorectal cancer (mCRC). It is already well established that<br />

only patients with wild-type KRAS tumors benefit from treatment with an<br />

anti-EGFR agent. Pyrosequencing is now used for a precise determination<br />

<strong>of</strong> KRAS mutation burden and a conservative cut<strong>of</strong>f <strong>of</strong> 10% was defined as<br />

the lower limit <strong>of</strong> quantification for the assignment <strong>of</strong> mutation status. Up<br />

to now, the impact <strong>of</strong> low-signal KRAS mutations below 10% on the<br />

response to anti-EGFR therapy in mCRC has not been evaluated. Methods:<br />

All consecutive patients treated by anti-EGFR for a mCRC between January<br />

2006 and June 2011 have been retrospectively analyzed by pyrosequencing<br />

using the therascreen KRAS Pyro Kit (Qiagen). All patients were defined<br />

wild-type (WT) for KRAS status using direct sequencing. The PFS data were<br />

plotted as Kaplan–Meier curve and compared by the log-rank test. Results:<br />

A total <strong>of</strong> 141 patients treated by anti-EGFR for a mCRC were included in<br />

the study. Mean age was 64.1 � 14.8 years and a majority <strong>of</strong> patients had<br />

synchronous metastases (68.6%). Patients benefited from anti-EGFR in<br />

first-line chemotherapy (30.7%), second-line (22.9%), third-line (35%) or<br />

later (11.4%). Majority <strong>of</strong> patients benefited from anti-EGFR combined<br />

with cytotoxic chemotherapy (91.4%), mostly irinotecan (78.6%). Using<br />

pyrosequencing, 117 tumors had a KRAS WT status and 24 tumors had<br />

low-signal mutation, between 2 and 10% (KRAS lowMT). Response rate<br />

according RECIST criteria were 13.6% versus 35.4% partial response,<br />

13.6% versus 37.2% stabilization and 72.7% versus 27.1% progression<br />

(p�0.01), for KRAS lowMT versus KRAS WT respectively. The PFS was<br />

respectively 2.6 � 0.2 months for KRAS lowMT versus 6.0 � 0.5 months<br />

for KRAS WT (p�0.024). Overall survival was not different between the two<br />

groups (27.4 months versus 33.0 months, p�0.4). Conclusions: Patients<br />

with tumors harboring KRAS lowMT benefit less <strong>of</strong> anti-EGFR therapy than<br />

patients with tumor harboring KRAS WT. While these results invite to<br />

consider low-signal tumors as positives, generalization awaits a large<br />

prospective trial.<br />

3522 Poster Discussion Session (Board #14), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Impact <strong>of</strong> age and medical comorbidity (MC) on adjuvant treatment<br />

outcomes for stage III colon cancer (CC): A pooled analysis <strong>of</strong> individual<br />

patient data from four randomized controlled trials. Presenting Author:<br />

Daniel G. Haller, Abramson Cancer Center at the University <strong>of</strong> Pennsylvania,<br />

Philadelphia, PA<br />

Background: Studies show significantly improved disease-free and overall<br />

survival (DFS, OS) for oxaliplatin (Ox)-based vs. leucovorin/5-fluorouracil<br />

(LV/5-FU) adjuvant therapy in CC, with conflicting reports <strong>of</strong> Ox benefit in<br />

patients � 70 years old. The impact <strong>of</strong> MC on Ox benefit has not been<br />

assessed. We assessed the impact <strong>of</strong> age and MC on adjuvant treatment<br />

outcomes for stage III CC. Methods: N � 4,819 patients from NSABP C-08,<br />

XELOXA, X-ACT, and AVANT were analyzed by Ox therapy (XELOX/FOLFOX)<br />

vs. LV/5-FU, MC, and age; patients treated with bevacizumab were<br />

excluded. Endpoints were DFS (primary), OS, and safety. MC was assessed<br />

(except NSABP C-08) by adapted Charlson Comorbidity and NCI Combined<br />

Indices (CCI, NCI): Low (� 1) vs. high (� 1). Hazardratios (HR) were<br />

estimated by Cox regression analyses. Multivariate Cox regression analyses<br />

(MVA) tested for independent effects <strong>of</strong> age and MC on Ox benefit,<br />

controlling for gender, T, and N stage. Results: Patient demographics, MC,<br />

and disease characteristics (except lymph nodes examined) were well<br />

balanced across groups. Median follow-up was shorter in NSABP C-08 and<br />

AVANT (36 and 50 months) vs. XELOXA and X-ACT (83 and 74 months).<br />

MVA-confirmed DFS/OS benefit was consistently shown for XELOX/FOLFOX<br />

vs. LV/5-FU, regardless <strong>of</strong> age or MC. Grade 3/4 serious adverse event (AE)<br />

rates were comparable across cohorts and CCI scores, and higher in<br />

patients aged � 70. Grade 3/4 AEs <strong>of</strong> interest, including peripheral sensory<br />

neuropathy, were comparable across ages and CCI scores, and higher with<br />

XELOX/FOLFOX. Conclusions: Ox benefit is modestly attenuated in patients<br />

aged � 70; however, significant benefit is observed regardless <strong>of</strong> age or MC<br />

in this analysis. Our results further support XELOX or FOLFOX as standard<br />

options for the adjuvant management <strong>of</strong> stage III CC in all age groups.<br />

Cox regression DFS HR OS HR 95% CI P<br />

CCI � 1 0.59 0.59 0.46–0.760.44–0.78 � .0001 .0003<br />

� 1 0.69 0.65 0.61–0.780.56–0.75 � .0001� .0001<br />

NCI � 1 0.58 0.46–0.730.42–0.74 � .0001� .0001<br />

0.56<br />

� 1 0.70 0.66 0.62–0.790.57–0.76 � .0001� .0001<br />

Age � 70 0.77 0.78 0.62–0.950.61–0.99 .014 .045<br />

� 70 0.68 0.62 0.61–0.760.54–0.72 � .0001� .0001<br />

3521 Poster Discussion Session (Board #13), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

MicroRNA signature to predict sensitivity to anti-EGFR monoclonal antibodies<br />

in metastatic colorectal cancer (mCRC). Presenting Author: Federico<br />

Cappuzzo, Istituto Toscano Tumori-Ospedale-Civile-Livorno, Livorno, Italy<br />

Background: MicroRNAs (MiRNAs) are post-transcriptional regulators that<br />

bind to complementary sequences on target messenger RNA transcripts,<br />

usually resulting in gene silencing. Microarray technology is a powerful<br />

high-throughput tool capable <strong>of</strong> monitoring the expression <strong>of</strong> thousands <strong>of</strong><br />

small noncoding RNAs at once. In the present study we aimed to<br />

investigate whether MiRNA signature was predictive for sensitivity to<br />

anti-EGFR monoclonal antibodies in mCRC. Methods: A total <strong>of</strong> 183 mCRC<br />

from two independent cohorts (cohort 1: 74 cases; validation cohort: 109<br />

cases) treated with cetuximab/panitumumab either alone (n�19) or in<br />

combination with chemotherapy (n�164) were included onto the study.<br />

MiRNA arrays were analysed using Agilent’s miRNA platform. Results:<br />

MiRNA array analyses identified the cluster miR-99a/Let7c/miR-125b<br />

located on 21p11.1 as associated with different outcome to anti-EGFR<br />

therapies. In the first cohort, individuals with high signature (n�25,<br />

33.8%) had a significantly longer progression free survival (PFS, 6.1 versus<br />

2.3 months, p�0.01, HR�0.42) and overall survival (OS, 29.8 versus 7.0<br />

months, p�0.04, HR�0.39) than patients with low signature (n�25,<br />

33.8%). Similar results were observed in the validation cohort. Patients<br />

with high signature (n�60, 32.8%) had a significantly longer PFS and<br />

longer OS than individuals with low signature (PFS 8.2 versus 4.2 months,<br />

p�0.04, HR�0.52; OS 12.8 versus 6.8 months, p�0.09, HR�0.65). To<br />

further assess the potential confounding effect <strong>of</strong> KRAS and BRAF<br />

mutations, we analyzed the outcome <strong>of</strong> patients with high and low signature<br />

in the 75 cases with KRAS or BRAF mutation and in 90 cases KRAS and<br />

BRAF wild-type. In the wild-type population, high signature patients<br />

(n�29, 32.2%) had a significantly longer PFS (8.8 versus 4 months,<br />

p�0.002, HR:0.46) and longer OS (17.7 versus 10 months, p�0.015,<br />

HR�0.62) than low signature individuals, with no difference in the KRAS<br />

or BRAF mutated patients. Conclusions: MiR-99a/Let7c/miR-125b signature<br />

is useful for improving selection <strong>of</strong> KRAS/BRAF wild-type mCRC<br />

patients candidate for anti-EGFR strategies.<br />

3523 Poster Discussion Session (Board #15), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Effect <strong>of</strong> oxaliplatin-based adjuvant therapy on post-relapse survival (PRS)<br />

in patients with stage III colon cancer: A pooled analysis <strong>of</strong> individual<br />

patient data from four randomized controlled trials. Presenting Author:<br />

Christopher Twelves, University <strong>of</strong> Leeds and St. James’s University<br />

Hospital, Leeds, United Kingdom<br />

Background: Oxaliplatin-based adjuvant therapy is the standard <strong>of</strong> care for<br />

stage III colon cancer; however, its impact on PRS in these patients is<br />

unclear. We therefore compared PRS in trials <strong>of</strong> patients with stage III<br />

colon cancer treated with oxaliplatin plus capecitabine (XELOX) or 5-flourouracil<br />

(5-FU; FOLFOX) vs. leucovorin/5-FU (LV/5-FU). Methods: Individual<br />

patientdata (N � 4,819) from NSABP C-08, XELOXA, X-ACT, and AVANT<br />

were pooled and analyzed by XELOX/FOLFOX vs. LV/5-FU; patients treated<br />

with bevacizumab were excluded. Hazard ratios (HR) were estimated by<br />

Cox regression analyses and multivariate Cox regression analyses controlled<br />

for age, gender, T, and N stage. Post-relapse treatment data were collected<br />

when available. Results: Patient demographics and disease characteristics<br />

(except lymph nodes examined) were well balanced across analytic groups.<br />

Median follow-up was shorter in NSABP C-08 and AVANT (36 and 50<br />

months) than in XELOXA and X-ACT (83 and 74 months). PRS was very<br />

similar for XELOX/FOLFOX and LV/5-FU (HR 0.94, 95% CI, 0.82–1.07; P<br />

� .33). Multivariate analyses supported these findings, but showed that<br />

PRS was associated with younger age and lower N stage at diagnosis after<br />

controlling for gender and T stage. PRS was also comparable for capecitabine<br />

or XELOX vs. LV/5-FU or FOLFOX (N � 5,819, HR 1.07, 95% CI,<br />

0.95–1.20; P � .26). Post-relapse therapies were comparable across the<br />

two cohorts. Conclusions: Adjuvant chemotherapy regimen did not impact<br />

on PRS in patients with stage III colon cancer; however, both N stage and<br />

age demonstrated independent effects on PRS. Studies have demonstrated<br />

significantly improved disease-free and overall survival for oxaliplatinbased<br />

therapy, and our data show that survival is not compromised by<br />

worsened PRS at subsequent relapse.<br />

Multivariate analysis PRS HR 95% CI P<br />

Randomized treatment:<br />

0.92 0.81–1.05 .23<br />

XELOX/FOLFOX vs. LV/5-FU<br />

Female vs. male 1.02 0.90–1.16 .71<br />

< 70 vs. > 70 0.70 0.60–0.81 � .0001<br />

T1–2 vs. T3–4 0.82 0.61–1.10 .19<br />

N1 vs. N2 0.73 0.64–0.83 � .0001<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


3524 Poster Discussion Session (Board #16), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Randomized phase III study <strong>of</strong> adjuvant chemotherapy with oral uracil and<br />

tegafur plus leucovorin versus intravenous fluorouracil and lev<strong>of</strong>olinate in<br />

patients (pts) with stage III colon cancer (CC): Final results <strong>of</strong> Japan<br />

<strong>Clinical</strong> Oncology Group study (JCOG0205). Presenting Author: Yasuhiro<br />

Shimada, National Cancer Center Hospital, Tokyo, Japan<br />

Background: NSABP C-06 reported the non-inferiority <strong>of</strong> oral adjuvant<br />

uracil and tegafur plus leucovorin (UFT/LV) to weekly fluorouracil and<br />

folinate (5-FU/LV) in disease-free survival (DFS) in stage II/III CC. Although<br />

adjuvant FOLFOX for stage III is standard care in US or EU, its toxicity and<br />

cost is major problem. This is the first report <strong>of</strong> JCOG0205, which<br />

compared UFT/LV to standard 5-FU/lev<strong>of</strong>oloinate (l-LV) in stage III CC.<br />

Methods: Pts were randomized to 3 courses <strong>of</strong> 5-FU/l-LV (5-FU 500 mg/m2 ,<br />

l-LV 250 mg/m2 , on days 1, 8, 15, 22, 29, 36, q8w), or 5 courses <strong>of</strong><br />

UFT/LV (UFT 300 mg/m2 /day, LV 75 mg/day, on days 1–28, q5w). Primary<br />

endpoint was DFS. Sample size was 1,100, determined with one-sided<br />

alpha <strong>of</strong> 0.05, power <strong>of</strong> 0.78, and non-inferiority margin <strong>of</strong> hazard ratio<br />

(HR) <strong>of</strong> 1.27. Owing to interim analyses, the multiplicity-adjusted alpha<br />

and confidence coefficient <strong>of</strong> CI in the final analysis were 0.0433 and<br />

91.3%. Results: Between Feb 2003 and Nov 2006, 1,101 pts (1,092<br />

eligible) were randomized to 5-FU/l-LV (n�550) or UFT/LV (n�551).<br />

Median follow-up was 72.0 months, median age: 61, colon/rectum:<br />

67%/33%, number <strong>of</strong> positive nodes �3/4�: 73%/27%, stage IIIa/IIIb:<br />

75%/25%. The HR <strong>of</strong> DFS was 1.02 (91.3% CI, 0.84–1.23) and<br />

non-inferiority <strong>of</strong> UFT/LV was demonstrated (P�0.0236). Incidences <strong>of</strong><br />

G3/4 toxicities were 8.4% neutropenia in 5-FU/l-LV and 8.7% ALT<br />

elevation in UFT/LV. In the 2 arms (5-FU/l-LV, UFT/LV), diarrhea (9.6%,<br />

8.5%) and anorexia (4.0%, 3.7%) were similar. G3/4 diarrhea was<br />

one-third less and G3/4 ALT elevation was three times more common than<br />

those in C-06. No treatment-related death was reported. Conclusions:<br />

Adjuvant UFT/LV is demonstrated to be non-inferior to standard 5-FU/l-LV<br />

in DFS. Five-year OS (87.5%) is favorable to 69.6% in C-06, possibly due<br />

to D3 dissection and stage migration. UFT/LV should be an oral treatment<br />

option for stage III CC.<br />

5-FU/l-LV UFT/LV Overall<br />

N 546 546 1,092<br />

3Y DFS (%) 79.3 77.8 78.6<br />

5Y DFS (%) 74.3 73.6 73.9<br />

5Y OS (%) 88.4 87.5 87.9<br />

N 544 540 1,084<br />

G3/4 neutropenia (%) 8.4 1.5 5.0<br />

G3/4 ALT (%) 0.7 8.7 4.7<br />

G3/4 diarrhea (%) 9.6 8.5 9.0<br />

3526 Poster Discussion Session (Board #18), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Smoking status and prognosis in patients with stage III colon cancer: A<br />

correlative analysis <strong>of</strong> NCCTG phase III trial N0147. Presenting Author:<br />

Amanda I. Phipps, Fred Hutchinson Cancer Research Center, Seattle, WA<br />

Background: Prior observational studies have suggested that smoking is<br />

associated with poorer overall survival after colon cancer (CC) diagnosis.<br />

Using data from N0147, a phase III randomized adjuvant trial <strong>of</strong> patients<br />

with stage III CC, we assessed the relationship between smoking status and<br />

cancer outcomes (disease-free survival (DFS) and time to recurrence<br />

(TTR)), accounting for possible heterogeneity by patient and tumor characteristics.<br />

Methods: Patients completed a risk factor questionnaire at<br />

baseline prior to randomization to FOLFOX or FOLFOX�cetuximab<br />

(N�1968). Information was collected on smoking and other lifestyle<br />

factors, including alcohol consumption, body mass index (BMI), and<br />

physical activity. Multivariate Cox models assessed the association between<br />

smoking history and the primary trial outcome <strong>of</strong> DFS, as well TTR,<br />

adjusting for other lifestyle and clinical factors. The median follow-up was<br />

3.5 years. Results: Overall, 52% <strong>of</strong> patients were former or current smokers.<br />

Compared to never smokers, ever smokers experienced significantly shorter<br />

DFS [3-year DFS: 70% vs 74%, hazard ratio (HR)�1.21, 95% confidence<br />

interval (CI): 1.02-1.42]. This association persisted after adjusting for age,<br />

sex, tumor subsite, number <strong>of</strong> nodes involved, T-stage, mismatch repair<br />

deficiency, BRAF mutation status, performance score, physical activity,<br />

BMI, and alcohol consumption (HR�1.23, 95% CI: 1.02-1.49). There was<br />

significant interaction in this association by BRAF mutation status (p�0.02):<br />

smoking was associated with shorter DFS in BRAF wildtype CC patients<br />

(HR�1.25, 95% CI: 1.04-1.51) but not in BRAF mutant CC patients<br />

(HR�0.72, 95% CI: 0.47-1.10). Patterns <strong>of</strong> association with smoking,<br />

overall and by BRAF status, were identical in analyses <strong>of</strong> TTR. Conclusions:<br />

Overall, smoking was significantly associated with shorter DFS and TTR in<br />

CC patients. These adverse relationships were most evident in patients with<br />

BRAF wildtype CC.<br />

Gastrointestinal (Colorectal) Cancer<br />

3525 Poster Discussion Session (Board #17), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Comparative evaluation <strong>of</strong> capecitabine or infusional leucovorin/5-fluorouracil<br />

(LV/5-FU) with or without oxaliplatin (Ox) for stage III colon cancer (CC): A<br />

pooled analysis <strong>of</strong> individual patient data from four randomized controlled trials.<br />

Presenting Author: Weijing Sun, Abramson Cancer Center at the University <strong>of</strong><br />

Pennsylvania, Philadelphia, PA<br />

Background: A fluoropyrimidine � Ox is the standard <strong>of</strong> care for stage III CC but a<br />

fluoropyrimidine alone is recommended for selected patients in several practice<br />

guidelines. We assessed efficacy and safety <strong>of</strong> adjuvant capecitabine � Ox vs.<br />

LV/5-FU � Ox across a population <strong>of</strong> stage III CC patients enrolled in four trials.<br />

Methods: N � 5,819 patients from NSABP C-08, XELOXA, X-ACT, and AVANT<br />

were pooled and analyzed; bevacizumab-treated patients were excluded. Endpoints<br />

were disease-free survival (DFS, primary), relapse-free survival (RFS),<br />

overall survival (OS), and safety. Multivariate Cox regression analyses (MVA)<br />

controlled for age, gender, T, and N stage. Results: Patient demographics and<br />

disease characteristics (except lymph nodes examined) were well balanced<br />

across groups. The number <strong>of</strong> patients receiving capecitabine and LV/5-FU were<br />

1,942 and 3,877, respectively. Median follow-up was shorter in NSABP C-08<br />

and AVANT (36 and 50 months) vs. XELOXA and X-ACT (83 and 74 months).<br />

Five-year DFS was 62.8% for capecitabine � Ox and LV/5-FU � Ox. The<br />

capecitabine by Ox interaction was significant for OS with a trend for DFS and<br />

RFS; likely due to improved outcomes with capecitabine alone and similar<br />

outcomes for capecitabine or LV/5-FU � Ox. Serious adverse event (AE) rates<br />

were similar for LV/5-FU- and capecitabine-based therapy (16% vs. 20%,<br />

respectively). Overall, treatment-related grade 3/4 AEs were more common with<br />

LV/5-FU (59% vs. 47%). Treatment-related grade 3/4 AEs <strong>of</strong> interest included<br />

peripheral sensory neuropathy (5% vs. � 1%), diarrhea (12% vs. 15%), febrile<br />

neutropenia (2% vs. � 1%), and hand–foot syndrome (� 1% vs. 12%).<br />

Conclusions: Adjuvant capecitabine � Ox and LV/5-FU � Ox show comparableefficacy<br />

benefits for the treatment <strong>of</strong> stage III CC; further supporting capecitabine<br />

or LV/5-FU-based regimens as standard options for the adjuvant therapy <strong>of</strong> stage<br />

III CC.<br />

Capecitabine � Ox vs.<br />

LV/5-FU � Ox<br />

Univariate MVA<br />

Cox regression HR<br />

95% CI P<br />

209s<br />

Ox interaction HR<br />

95% CI P<br />

DFS 1.01 .86 1.14 .17<br />

0.92–1.10 0.95–1.37<br />

RFS 1.01 .86 1.15 .15<br />

0.92–1.11 0.95–1.39<br />

OS 1.02 .65 1.33 .01<br />

0.92–1.14 1.06–1.67<br />

3527 Poster Discussion Session (Board #19), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Adjuvant chemotherapy (AC) use and outcomes in stage II colon cancer<br />

(CC) with and without poor prognostic features. Presenting Author: Aalok<br />

Kumar, British Columbia Cancer Agency, Vancouver, BC, Canada<br />

Background: AC is frequently considered inpatients (pts) with �high risk�<br />

stage II CC, defined by the presence <strong>of</strong> �/�1 poor prognostic features, such<br />

as obstruction or perforation, T4 stage, �12 lymph nodes retrieved,<br />

positive margins, and lymphovascular or perineural invasion. However,<br />

survival benefits associated with AC use in high risk pts remain largely<br />

unproven. Our aims were to 1) examine patterns <strong>of</strong> AC use in stage II CC<br />

and 2) explore the relationship between AC use and survival in high vs low<br />

risk pts. Methods: All pts diagnosed with stage II CC in British Columbia<br />

from 1999 to 2008 and evaluated at any 1 <strong>of</strong> 5 regional centers were<br />

reviewed. Kaplan-Meier and Cox regression methods were used to correlate<br />

a) high vs low risk status and b) receipt <strong>of</strong> AC with relapse-free (RFS),<br />

disease specific (DSS) and overall survival (OS). Results: We identified<br />

1,697 stage II CC pts: 1,236 (73%) high risk and 461 (27%) low risk<br />

among whom 363 (29%) and 61 (13%) received AC, respectively.<br />

Individuals with high risk features who received AC were younger (median<br />

62 vs 72 years, p�0.001) and had better performance status (ECOG 0/1<br />

47% vs 34%%, p�0.02). In the high risk group, AC was associated with<br />

improved 5-year OS, but not with RFS or DSS (Table). After adjusting for<br />

confounders, an OS advantage from AC persisted for high risk pts (HR<br />

0.67, 95CI 0.52-0.86, p�0.002), but no significant RFS or DSS benefits<br />

were seen (HR 0.76, 95CI 0.58-0.99, p�0.05 and HR 0.75, 95CI<br />

0.55-1.02, p�0.11, respectively). Subgroup analyses showed that individuals<br />

with T4 lesions had improved RFS (HR 0.63, 95CI 0.42-0.95,<br />

p�0.03), DSS (HR 0.59, 95CI 0.37-0.93, p�0.02), and OS (HR 0.50,<br />

95CI 0.33-0.77, p�0.002). Conclusions: In this population-based cohort<br />

<strong>of</strong> stage II CC, AC was associated with an OS advantage in high risk pts,<br />

likely due to pt selection. RFS and DSS benefits were mainly seen in T4<br />

lesions, suggesting a limited role for AC in pts deemed high risk based on<br />

clinical and pathological factors. Risk stratification based on molecular<br />

testing should be further explored.<br />

Survival in high vs low risk stage II CC.<br />

3-year RFS % p value 5-year DSS % p value 5-year OS % p value<br />

High risk<br />

AC 78 0.98 80 0.83 75 �0.01<br />

No AC 80 79 68<br />

Low risk<br />

AC 87 0.18 93 0.78 87 0.26<br />

No AC 93 93 85<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


210s Gastrointestinal (Colorectal) Cancer<br />

3528 Poster Discussion Session (Board #20), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

BRAF V600 mutant colorectal cancer (CRC) expansion cohort from the<br />

phase I/II clinical trial <strong>of</strong> BRAF inhibitor dabrafenib (GSK2118436) plus<br />

MEK inhibitor trametinib (GSK1120212). Presenting Author: Ryan Bruce<br />

Corcoran, Massachusetts General Hospital Cancer Center, Boston, MA<br />

Background: BRAF V600 mutations occur in 10-15% <strong>of</strong> CRC and may<br />

predict lack <strong>of</strong> response to standard chemotherapy and anti-EGFR treatment.<br />

There have been no significant recent changes in the treatment <strong>of</strong><br />

CRC. Novel therapeutic strategies for BRAF mutant CRC are critically<br />

needed. To collect preliminary efficacy data, an expansion cohort <strong>of</strong> 25<br />

BRAF mutant CRC patients (pts) was included in part 2 <strong>of</strong> a phase I/II study<br />

evaluating the combination <strong>of</strong> dabrafenib (BRAFi) and trametinib (MEKi).<br />

Methods: Eligible pts had BRAF V600 mutant CRC, measurable disease per<br />

RECIST 1.1, and previous treatment with anti-cancer therapies. Pts were<br />

treated with established dose <strong>of</strong> dabrafenib (150mg BID) and trametinib<br />

(2mg QD). Results: Data for 23 pts are available at data cut-<strong>of</strong>f: median age<br />

54 years, 74% female, ECOG performance status 0 (48%) or 1 (52%).<br />

39% had received prior EGFR inhibitor treatment. 74% had received � 2<br />

prior chemotherapy regimens; 48% had � 1 biologic therapy. 48% had<br />

� 3 metastatic sites. Three <strong>of</strong> the 23 pts were non-evaluable at data cut-<strong>of</strong>f<br />

as they had not reached the first restaging assessment. Among the 20<br />

evaluable pts, 1 (5%) achieved partial response and 10 (50%) stable<br />

disease. Minor responses (�10% to �30% tumor shrinkage) were observed<br />

in 4/10 patients (40%) with stable disease. The most frequent<br />

adverse events observed in all <strong>Part</strong> 2 patients who received the same dosing<br />

combination (n�75) included pyrexia (65%), fatigue (47%), nausea<br />

(44%), chills (41%), vomiting (31%), constipation (28%), diarrhea (24%)<br />

and rash (21%). Conclusions: Dabrafenib at 150 mg BID combines safely<br />

with trametinib 2 mg QD in this BRAF mutant CRC cohort. Based on the<br />

preliminary anti-tumor activity seen, further investigation is warranted in<br />

BRAF mutant CRC pts. Updated safety and efficacy data and an analysis <strong>of</strong><br />

the relationship between efficacy and potential predictive biomarkers,<br />

including microsatellite status, PTEN loss, PIK3CA mutation, and total and<br />

phosphorylated EFGR levels, will be presented.<br />

3530 Poster Discussion Session (Board #22), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Overall survival (OS) <strong>of</strong> advanced colorectal cancer (aCRC) patients (pts)<br />

treated with the multipeptide vaccine IMA910: Results <strong>of</strong> a matched-pair<br />

analysis with arm C pts from COIN. Presenting Author: Tim S. Maughan,<br />

Gray Institute for Radiation Oncology and Biology, University <strong>of</strong> Oxford,<br />

Oxford, United Kingdom<br />

Background: IMA910 is a novel cancer vaccine consisting <strong>of</strong> 10 class I and<br />

3 class II tumor-associated peptides (TUMAPs), naturally presented on<br />

HLA molecules <strong>of</strong> CRC. Vaccine-induced immune responses are associated<br />

with prolonged OS <strong>of</strong> renal cell carcinoma pts treated with IMA901, a<br />

multi-peptide vaccine identified by the same antigen discovery platform.<br />

Methods: 92 HLA-A*02� aCRC pts with stable or responding disease after<br />

12 weeks <strong>of</strong> first-line oxaliplatin-based therapy were enrolled in this phase<br />

I/II trial. After immunomodulation with cyclophosphamide (300 mg/m2 )to<br />

reduce regulatory T cells, pts were immunized intradermally (up to 16<br />

vaccinations) with IMA910 in combination with GM-CSF without (cohort 1;<br />

n�66) or with (cohort 2; n�26) topically applied imiquimod. Safety and<br />

PFS data was recently presented [Mayer et al., ASCO-GI 2012]. OS <strong>of</strong><br />

IMA910 pts was compared to matched pts from arm C (intermittent<br />

chemotherapy) <strong>of</strong> the COIN trial [Adams et al., Lancet Oncology 2011].<br />

Matching was performed in a prospectively defined, fully blinded fashion<br />

based on a propensity score involving all available prognostic factors. T-cell<br />

responses to individual IMA910 peptides were analyzed by HLA multimer<br />

and intracellular cytokine assays. Results: At a median follow up <strong>of</strong> 1.7 yrs,<br />

OS <strong>of</strong> IMA910 vaccinated pts was significantly longer in comparison to 1:1<br />

matched HLA-A*02� COIN pts (HR 0.675, 95% CI 0.458-0.995,<br />

p�0.047; 1 yr OS: 69% vs 55%; 2 yr OS: 40% vs 24%). Multi-TUMAP<br />

responders (�2 CD8� and �2 CD4� vaccine-induced T cells) had a<br />

significantly longer OS (HR 0.45, p�0.04) compared to matched COIN pts<br />

while non-multi-TUMAP responders showed similar OS compared to<br />

matched COIN pts (HR 0.76, p�0.22). Conclusions: OS <strong>of</strong> IMA910 pts was<br />

significantly longer in comparison to matched COIN pts with separation <strong>of</strong><br />

survival curves at ~9 months and increasing OS difference over time.<br />

Multi-TUMAP immune responders had a significantly longer OS compared<br />

to non-multi-TUMAP responders and compared to matched COIN pts.<br />

These clinical and immune response data strongly suggest clinical activity<br />

<strong>of</strong> IMA910 in 1st line CRC pts and warrant further development.<br />

3529 Poster Discussion Session (Board #21), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Pharmacodynamic biomarker-driven trial <strong>of</strong> MK-2206, an AKT inhibitor, with<br />

AZD6244 (selumetinib), a MEK inhibitor, in patients with advanced colorectal<br />

carcinoma (CRC). Presenting Author: Giovanna Speranza, National Cancer<br />

Institute, Bethesda, MD<br />

Background: The RAF/MEK/ERK and PI3K/AKT signaling pathways are commonly<br />

deregulated in CRC. Each can serve as a compensatory mechanism<br />

mediating resistance to single pathway blockade. Combined inhibition with<br />

MK-2206 and selumetinib (AZD6244) could enhance antitumor activity.<br />

Tolerable doses for the combination in solid tumors (ASCO 2011, abstr 3004)<br />

are less than single agent MTDs. We conducted a biomarker driven trial <strong>of</strong><br />

MK-2206 and selumetinib at the established combination doses to determine<br />

extent <strong>of</strong> pERK and pAKT inhibition in tumor biopsies (bx), using new, clinically<br />

validated immunoassays for pERK and pAKT. Methods: Patients (pts) with<br />

advanced CRC, refractory to standard therapy, ECOG � 2, adequate organ<br />

function, and disease amenable to bx were treated with oral MK-2206, 90 mg<br />

QW and selumetinib, 75 mg daily, stratified for KRAS mutation status (� or WT).<br />

For each strata, pAKT and pERK levels from paired tumor bx, baseline and 3-6<br />

hrs post-dose in 3 pts each on C1D1 or C1D22, were measured using a<br />

quantitative, chemiluminescence immunoassay (dilution linearity R2 � 0.985<br />

pERK, 0.995 pAKT; % CV at 0 � 6). 70% inhibition was considered biologically<br />

significant based on levels observed in preclinical models that demonstrated<br />

antitumor activity. Results: 11 pts enrolled to date (KRAS �, 6 pts; WT, 5 pts).<br />

2/9 pts had SD after 2 cycles. Gr 1/ 2 toxicities: rash, reversible retinal<br />

detachment, liver abnormalities, hypoalbuminemia, lymphopenia. Target protein<br />

inhibition data are available for 8 paired bx samples (Table). Conclusions:<br />

Although 4/8 pts demonstrated biologically significant inhibition in one marker,<br />

at the MTD for this combination no pt had � 70% inhibition <strong>of</strong> both targets. If<br />

repeated dosing does not produce the desired inhibition <strong>of</strong> pERK and pAKT, we<br />

will conclude that the dose reductions for each agent necessitated by the toxicity<br />

<strong>of</strong> the agents used in combination preclude the possibility <strong>of</strong> providing adequate<br />

dual pathway inhibition. Loss <strong>of</strong> PTEN and DNA mutation analyses (KRAS,<br />

BRAF, PIK3Ca, AKT) are planned.<br />

% Inhibition <strong>of</strong> phosphorylation<br />

pt pERK pAKT<br />

C1D1<br />

KRAS � 1 46 15<br />

2 �77 4<br />

3 61 23<br />

C1D1<br />

KRAS WT 4 53 19<br />

5 �70 34<br />

6 48 78<br />

C1D22<br />

KRAS � 7 77 20<br />

8 0 60<br />

3531 Poster Discussion Session (Board #23), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Molecular analysis <strong>of</strong> the randomized phase II/III study <strong>of</strong> the anti-IGF-1R<br />

antibody dalotuzumab (MK-0646) in combination with cetuximab (Cx) and<br />

irinotecan (Ir) in the treatment <strong>of</strong> chemorefractory KRAS wild-type metastatic<br />

colorectal cancer (mCRC). Presenting Author: David J. Watkins, The<br />

Royal Marsden Hospital, London and Surrey, United Kingdom<br />

Background: Previously reported results <strong>of</strong> this randomized study demonstrated<br />

that the addition <strong>of</strong> dalotuzumab (Dz) worsened the PFS and OS <strong>of</strong><br />

chem<strong>of</strong>ractory mCRC patients receiving Cx and Ir (Watkins et al; ASCO<br />

2011). Comprehensive molecular analysis has been undertaken retrospectively<br />

to identify possible predictors <strong>of</strong> Cx resistance and Dz response.<br />

Methods: Quantitative RT-PCR for IGF-1, IGF-2, immunohistochemistry for<br />

IGF-1R and microarray expression pr<strong>of</strong>iling was conducted on archival<br />

tumor tissue. All patients had received Cx and Ir with either placebo<br />

(n�107), weekly Dz (n�112), or 2 weekly Dz (n�110). Results: Data from<br />

292 and 206 patients was successfully obtained by RT-PCR or microarray<br />

respectively. Within the placebo arm, high IGF-1 expression was found to<br />

be associated with resistance to Cx (IGF-1-/IGF-1�; PFS 6.7/3.7 months,<br />

OS 15.5/9.6 months). High IGF-1 expression was associated with benefit<br />

from the addition <strong>of</strong> weekly Dz (placebo/weekly Dz; PFS 3.7/5.7 months,<br />

OS 9.6/18.2 months). By contrast the addition <strong>of</strong> Dz was not effective in<br />

tumors with high IGF-2 expression (placebo/weekly Dz; PFS 8.4/2.7<br />

months). Microarray analysis revealed distinct populations that differentially<br />

correlated with Cx and Dz response. An epithelial phenotype appeared<br />

more associated with Cx response, whereas a mesenchymal phenotype<br />

more associated with Dz response. Rectal cancers showed greater association<br />

with increased IGF-1 expression, EMT gene signature and Dz response.<br />

Conclusions: These data support IGF-1 and IGF-2 as potential biomarkers<br />

for response to Dz therapy and high IGF-1 as a marker <strong>of</strong> resistance to Cx<br />

therapy. Based on these data Dz is being further evaluated in a molecularly<br />

selected population <strong>of</strong> mCRC.<br />

Weekly Dz vs placebo arm<br />

Hazard ratio (95% CI)<br />

Difference<br />

in response rate%<br />

All patients<br />

n�191<br />

Colorectal<br />

high IGF-2<br />

n�25<br />

Colorectal<br />

high IGF-1<br />

n�48<br />

Rectal<br />

high IGF-1<br />

n�23<br />

- 7.4% - 13.3% � 14.6% � 24.6%<br />

PFS 1.30 (0.95, 1.78) 2.94 (1.09, 7.9) 0.59 (0.28, 1.24) 0.35 (0.10, 1.19)<br />

OS 1.37 (0.96, 1.95) 3.48 (0.97, 12.54) 0.67 (0.31, 1.45) 0.49 (0.15, 1.56)<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


3532 Poster Discussion Session (Board #24), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Randomized phase II open-label study <strong>of</strong> mFOLFOX6 in combination with<br />

linifanib or bevacizumab for metastatic colorectal cancer. Presenting<br />

Author: Bert H. O’Neil, University <strong>of</strong> North Carolina at Chapel Hill, Chapel<br />

Hill, NC<br />

Background: Linifanib is a potent and selective inhibitor <strong>of</strong> VEGF/PDGF<br />

receptors. This trial assessed the efficacy and safety <strong>of</strong> mFOLFOX6 in<br />

combination with linifanib or bevacizumab as second-line treatment for<br />

metastatic colorectal cancer (mCRC). Methods: Patients (pts) with measurable<br />

mCRC refractory to 1 prior regimen and ECOG PS 0–1, stratified by<br />

prior bevacizumab treatment and radiotherapy, were randomized to receive<br />

mFOLFOX6 with bevacizumab 10 mg/kg on day (d) 1 <strong>of</strong> 14-d cycle (Arm A),<br />

mFOLFOX6 with daily linifanib 7.5 mg (Arm B), or mFOLFOX6 with daily<br />

linifanib 12.5 mg (Arm C). The primary endpoint was progression-free<br />

survival (PFS). Severity <strong>of</strong> adverse events (AEs) was graded using NCI-<br />

CTCAE v3.0. Results: 148 pts were randomized at 45 sites in 14 countries.<br />

32 pts (21.6%) had received prior bevacizumab. PFS and response data<br />

are shown below (Table). Median survival (OS) was not reached at median<br />

follow up 7.6 months. Palmar-plantar erythrodysesthesia (PPE) was the<br />

only Grade 3/4 AE significantly higher on linifanib (high dose, 16.3%) vs.<br />

bevacizumab (0%). Rate <strong>of</strong> any Grade 3� AE was significantly higher on<br />

linifanib vs. bevacizumab.Hypertension rates were 41.7% (Arm A), 40.0%<br />

(Arm B), and 36.7% (Arm C). AEs dose-related to linifanib were constipation,<br />

proctalgia, stomatitis, fatigue, weight decrease, decreased appetite,<br />

and PPE. Conclusions: The addition <strong>of</strong> linifanib to mFOLFOX6, compared to<br />

mFOLFOX6 � bevacizumab, did not provide a PFS advantage for mCRC.<br />

OS results will be updated for conference presentation.<br />

ArmAN�49 Arm B N�50 ArmCN�49<br />

Median PFS, m [95% CI] 9.0 [6.7, --] 6.6 [5.5, 9.2] 7.7 [6.3, 9.3]<br />

HR 1.45, P�0.186* HR 1.26, P�0.466*<br />

Confirmed response rate, % 34.7 24.0 22.4<br />

Best response rate, % 38.8 32.0 34.7<br />

Best % change in tumor size, median –23.1 –19.5 –22.2<br />

* Stratified log-rank test two-sided P-value compared with mFOLFOX6 � bevacizumab.<br />

3534 General Poster Session (Board #22E), Mon, 8:00 AM-12:00 PM<br />

Analysis <strong>of</strong> PTEN in patients with advanced colorectal cancer (CRC)<br />

receiving capecitabine alone or in combination with bevacizumab with or<br />

without mitomycin C in the phase III AGITG MAX trial. Presenting Author:<br />

Timothy Jay Price, The Queen Elizabeth Hospital, Adelaide, Australia<br />

Background: There is an urgent need for potential biomarkers for anti-VEGF<br />

therapies. The tumour suppressor gene PTEN is thought to have a potential<br />

role as a biomarker for anti-EGFR therapy in CRC, but there is also evidence<br />

that decreased levels <strong>of</strong> PTEN leads to increased expression <strong>of</strong> VEGF<br />

suggesting a potential relationship to outcome with anti-VEGF therapy*.<br />

The prognostic value <strong>of</strong> PTEN also remains controversial. Methods: Patients<br />

enrolled in the Phase III MAX trial <strong>of</strong> capecitabine (C) �/- bevacizumab (B)<br />

(�/- mitomycin C (M)) with available tissue were analysed for PTEN<br />

expression (loss v no loss) as assessed using a Taqman copy number assay<br />

to measure copy number variation at the PTEN locus. PTEN status was<br />

correlated with progression-free survival (PFS) and overall survival (OS)<br />

outcomes. The predictive value <strong>of</strong> PTEN status for bevacizumab efficacy<br />

was also examined. Results: Of the original 471 patients enrolled in the<br />

trial, tissue from 302 (64.1%) patients were analysed. Baseline characteristics<br />

<strong>of</strong> those with and without tissue were comparable. PTEN loss was<br />

observed in 38.7% <strong>of</strong> patients. There was no relationship between PTEN<br />

loss and KRAS or BRAF mutation. Patients with PTEN loss were more likely<br />

to have rectal primary (p�0.01) and less likely to have lung metastasis<br />

(p�0.03). By using the comparison <strong>of</strong> CvCB�CBM, PTEN status was not<br />

significantly predictive <strong>of</strong> the effectiveness <strong>of</strong> B for PFS or OS (Table).<br />

PTEN status is also not prognostic for PFS or OS in multivariate analyses<br />

adjusting for other baseline factors (Table). Conclusions: PTEN status did<br />

not significantly predict different benefit with anti-VEGF therapy. PTEN<br />

status was also not prognostic for survival in advanced colorectal cancer.<br />

P value<br />

CvsCB�CBM Loss No loss (for interaction)<br />

PFS HR 0.51<br />

HR 0.72<br />

P � .26<br />

0.33 - 0.79 0.52-0.98<br />

OS HR 0.75<br />

HR 1.00<br />

0.47-1.19 0.70-1.43 P � .35<br />

Prognosis Loss vs no loss P value<br />

PFS HR 0.90<br />

P�.42<br />

0.70 - 1.16<br />

OS HR 1.04<br />

P�.76<br />

0.79 - 1.38<br />

Gastrointestinal (Colorectal) Cancer<br />

3533 Poster Discussion Session (Board #25), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Use <strong>of</strong> next-generation sequencing (NGS) to detect a novel ALK fusion and<br />

a high frequency <strong>of</strong> other actionable alterations in colorectal cancer (CRC).<br />

Presenting Author: Jeffrey S. Ross, Albany Medical College, Albany, NY<br />

Background: We hypothesized that NGS could identify genomic alterations<br />

that guide selection <strong>of</strong> targeted therapies and discover novel drug targets<br />

for primary and metastatic CRC. Methods: DNA was extracted and sequenced<br />

from 4 X 10 � FFPE sections from 40 (32 primary and 8<br />

metastatic) CRCs obtained from 2004-10 (52% male; 48% female; mean<br />

age 60 years; 10% Stages I/II; 40% Stage III; 40% Stage IV; 10% Stage<br />

unknown) using a targeted NGS assay in a CLIA laboratory (Foundation<br />

Medicine). 2574 exons <strong>of</strong> 145 cancer-related genes plus 37 introns from<br />

14 genes <strong>of</strong>ten rearranged in cancer were fully sequenced for point<br />

mutations, insertions/deletions, copy number alterations (CNAs) and select<br />

gene fusions. Results: NGS revealed 125 genomic alterations in 39/40<br />

tumors (mean 3.1, range1-7) in 21 genes, including 80 base substitutions<br />

(64%), 39 insertions/deletions (31%), 4 CNAs (3%) and 2 gene fusions<br />

(1.6%). TP53 and APC were altered in 80% (32/40) and 67.5% (27/40) <strong>of</strong><br />

CRCs respectively, with both mutated more frequently than reported in<br />

COSMIC. Alterations associated with potential sensitivity to targeted<br />

therapies were identified in 21 (52.5%) <strong>of</strong> CRCs including: 10 KRAS<br />

(TK/MEK inhibitors), 6 BRAF (BRAF inhibitors), 5 FBXW7 (mTOR inhibitors);<br />

2 PIK3CA (PI3K/mTOR inhibitors); 2 BRCA2 (PARP inhibitors); 2<br />

GNAS (MEK/ERK inhibitors) and 1 CDK8 (CDK inhibitors). In 1 CRC, a 5.2<br />

Mb tandem duplication generated a novel C2orf44-ALK gene fusion<br />

starting at the canonical exon 20 recombination site previously reported for<br />

the majority <strong>of</strong> ALK gene fusions. cDNA sequencing identified an 90-fold<br />

increase in 3’ ALK expression, suggesting the C2orf44-ALK fusion results<br />

in ALK kinase domain overexpression and contains the same intracellular<br />

domain as other ALK fusions including the ALK inhibitor sensitive<br />

EML4-ALK. Conclusions: NGS <strong>of</strong> broad, cancer-related gene content from<br />

FFPE CRC samples uncovered an unexpectedly high frequency <strong>of</strong> genomic<br />

alterations, many <strong>of</strong> which may be clinically actionable by informing<br />

treatment decisions, including a novel ALK gene fusion. This previously<br />

unrecognized subset <strong>of</strong> CRC patients may be candidates for clinical trials <strong>of</strong><br />

crizotinib or other ALK inhibitors.<br />

3535 General Poster Session (Board #22F), Mon, 8:00 AM-12:00 PM<br />

Final results <strong>of</strong> study 20050181: A randomized phase III study <strong>of</strong> FOLFIRI<br />

with our without panitumumab (pmab) for the second-line treatment (tx) <strong>of</strong><br />

metastatic colorectal cancer (mCRC). Presenting Author: Alberto F. Sobrero,<br />

Ospedale San Martino, Medical Oncology, Genova, Italy<br />

Background: The primary analysis <strong>of</strong> study 20050181 showed that in<br />

patients (pts) with wild-type (WT) KRAS mCRC, pmab plus FOLFIRI given<br />

as second-line therapy significantly improved progression-free survival<br />

(PFS) vs FOLFIRI alone. Here, we report on a prespecified descriptive<br />

analysis planned for 30 months (mos) after the last pt was enrolled.<br />

Methods: Pts with mCRC, ECOG 0-2, who had one prior fluoropyrimidinebased<br />

chemotherapy regimen were randomized 1:1 to pmab 6.0 mg/kg<br />

Q2W�FOLFIRI (Arm 1) vs FOLFIRI (Arm 2). Co-primary endpoints were OS<br />

and PFS (central assessment). Secondary endpoints included objective<br />

response rate (ORR) and safety. Tumor KRAS status was determined by a<br />

blinded central lab. Results: 1186 pts were randomised and received tx:<br />

591 in Arm 1, 595 In Arm 2. 1083 pts (91%) had KRAS results. Adverse<br />

event rates were consistent with the primary analysis. Results are shown<br />

below. Conclusions: In pts with WT KRAS mCRC receiving pmab�FOLFIRI<br />

vs FOLFIRI, PFS and ORR were significantly improved, and there was a<br />

trend toward improved OS. Post-progression anti-EGFR tx may have<br />

attenuated any significant difference in OS between tx arms. In pts with MT<br />

KRAS mCRC, no difference in efficacy was observed between tx arms.<br />

KRAS testing is critical to select appropriate pts for tx with pmab.<br />

WT KRAS<br />

(n�597)<br />

PFS<br />

Median, mos<br />

(95%CI)<br />

Arm 1<br />

Pmab�FOLFIRI<br />

(n�303)<br />

Arm 2<br />

FOLFIRI<br />

(n�294)<br />

HR or OR<br />

(95%CI) p value<br />

6.7 (5.8–7.4) 4.9 (3.8–5.5) HR�0.82<br />

(0.69–0.97)<br />

HR�0.73a (0.60–0.88)<br />

14.5 (13.0–16.1) 12.5 (11.2–14.2) HR�0.92<br />

(0.78–1.10)<br />

OS<br />

Median, mos<br />

(95% CI)<br />

ORRb 36 (31–42) [107/297] 10 (7–14) [28/286] OR�5.50<br />

% (95%CI), [x/n]<br />

(3.32–8.87)<br />

MT KRAS<br />

(n�486)<br />

(n�238) (n�248)<br />

PFS<br />

Median, mos (95%CI)<br />

5.3 (4.2–5.7) 5.4 (4.0–5.6) HR�0.95<br />

(0.78–1.14)<br />

OS<br />

Median, mos (95% CI)<br />

ORR<br />

11.8 (10.4–13.3) 11.1 (10.3–12.4) HR�0.93<br />

(0.77–1.13)<br />

b<br />

13 (9–18) [31/232] 15 (11–20) [35/237] OR�0.93<br />

% (95%CI), [x/n]<br />

Pts receiving post-study<br />

anti-EGFR tx, n (%)<br />

(0.53–1.63)<br />

WT KRAS 38 (12.5) 101 (34.4)<br />

MT KRAS 21 (8.8) 79 (31.9)<br />

211s<br />

0.023<br />

0.001 a<br />

0.366<br />

�0.0001<br />

0.561<br />

0.482<br />

0.885<br />

aOn tx (including deaths occurring �60 days after last tumor assessment); bPts with baseline measurable<br />

disease (modified RECIST).<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


212s Gastrointestinal (Colorectal) Cancer<br />

3536 General Poster Session (Board #22G), Mon, 8:00 AM-12:00 PM<br />

Do we need adjuvant therapy in rectal cancer with complete pathologic<br />

response (ypT0N0) after induction chemoradiation and laparoscopic mesorectal<br />

excision? Presenting Author: Verónica Pereira, Department <strong>of</strong> Medical<br />

Oncology, Hospital Clínic de Barcelona, Barcelona, Spain<br />

Background: Neoadjuvant chemo-radiotherapy (CRT) is the standard <strong>of</strong> care<br />

for patients (pts) with u�T3 by endoscopic ultrasound (EUS) rectal cancer.<br />

Although pts with complete pathological response (ypT0N0) fare well in<br />

multiple series, there is uncertainty <strong>of</strong> whether it’s due to the induction<br />

(CRT), due to the adjuvant chemotherapy (ACT) or due to the combination<br />

<strong>of</strong> both therapies. We have evaluated long-term outcomes in CRT-treated<br />

pts. Those with ypT0N0, were not treated with ACT. Methods: Pts with<br />

u�T3 rectal cancer, received neoadjuvant chemotherapy (225mg/m2/day<br />

5-fluorouracil (FU)) in continuous infusion (CI) per 5 weeks (wks) and<br />

concomitant radiotherapy (45 Gy). Laparoscopic surgery (LAP) was planned<br />

after an interval <strong>of</strong> 5-8 wks. Pts achieving ypT0N0 were no treated with<br />

ACT. Pts with ypT�1 or N1 were treated with 3 gr/m2 FU in 48 hour CI and<br />

LV 200 mg/m2 every 2 wks x 6 cycles. Results: From November 2000 to<br />

November 2008, a cohort <strong>of</strong> 173 pts were treated with induction CRT and<br />

167 pts underwent total mesorectal excision (LAP, n�158, open surgery<br />

n�9). Complete pathological response was achieved in 26/167 pts<br />

(15.5%). After a median follow-up <strong>of</strong> 58.3 months, pts with ypT0N0 have a<br />

5-year disease-free survival and overall survival rate <strong>of</strong> 96% (95% CI 76 to<br />

99%) and 100% (95% CI not estimable) respectively. Conclusions: Using<br />

these results, a clinical trial comparing observation versus adjuvant therapy<br />

in ypT0N0 after standard CRT, would need to enroll 3088 pts to show a HR<br />

<strong>of</strong> 0.75 in favor <strong>of</strong> ACT after 5 years <strong>of</strong> follow-up (alpha�.05, beta�.2). In<br />

case that the true DFS lied in the lower bound <strong>of</strong> the 95% CI, 636 patients<br />

would be needed under the same assumptions. These results do not<br />

support the administration <strong>of</strong> ACT to ypT0N0 patients.<br />

3538 General Poster Session (Board #23A), Mon, 8:00 AM-12:00 PM<br />

Maintenance therapy with biweekly cetuximab (C) in the first-line treatment<br />

<strong>of</strong> metastatic colorectal cancer (mCRC): The NORDIC 7.5 study<br />

(NCT00660582), by the Nordic Colorectal Cancer Biomodulation Group.<br />

Presenting Author: Per Pfeiffer, Department <strong>of</strong> Oncology, Odense University<br />

Hospital, Odense, Denmark<br />

Background: NORDIC 7.5 is a multi-center phase II trial <strong>of</strong> Nordic FLOX<br />

plus biweekly C followed by maintenance C in first-line treatment <strong>of</strong> mCRC,<br />

KRASwild-type (KRASwt). The aim was to complement the data <strong>of</strong> the<br />

NORDIC VII trial, arm C (Tveit et al, ASCO GI 2011). Specifically, the goal<br />

was to substantiate efficacy and safety <strong>of</strong> biweekly C as maintenance<br />

therapy and to assess time to failure <strong>of</strong> strategy (TFS) as an end-point in<br />

maintenance therapy studies. Methods: Patients had KRASwt, measurable,<br />

non-resectable mCRC; no prior chemotherapy for metastases; WHO PS 0-2<br />

and good organ function. Patients received 8 courses <strong>of</strong> FLOX (bolus<br />

5FU/folinic acid days 1 � 2 with oxaliplatin day 1 every 2 weeks) plus<br />

biweekly C (500 mg/m2 every 2 weeks) for 16 weeks followed by biweekly C<br />

as maintenance therapy until progression (PD). After at least 2 months <strong>of</strong><br />

maintenance therapy, patients restarted FLOX plus biweekly C at the time<br />

<strong>of</strong> RECIST PD, and continued until a second RECIST PD. The primary<br />

endpoint was response rate (RR). Eighty-six patients were planned to<br />

confirm a response rate <strong>of</strong> 60%. While awaiting the results <strong>of</strong> Nordic VII, we<br />

amended the protocol to include 150 patients. Secondary endpoints<br />

included PFS, OS, resection rate, and safety. TFS was an explorative<br />

endpoint. Results: From July 2008 to September 2010, 152 KRASwt<br />

patients were included in 11 Nordic centers. Results were updated<br />

December 2011. Median age 64 years; 94% PS 0-1. RR 62%, median PFS<br />

8.0 months, median OS 23.2 months, median TFS 11.9 months. Ten<br />

patients (15%) had R0-resection <strong>of</strong> metastasis. FLOX � C was reintroduced<br />

in 47 <strong>of</strong> 85 patients (55%) who were candidates for retreatment<br />

and was started median 18 days after PD. Median duration <strong>of</strong><br />

re-introduction was 5 months (1-29), 9 patients (20%) had response.<br />

Elevated baseline platelets and neutr<strong>of</strong>iles were associated with poor OS.<br />

The most common grade 3/4 non-hematologic AEs in were diarrea (9%),<br />

skin rash (9%), infection without neutropenia (7%), and fatigue (7%)<br />

Conclusions: Maintenance therapy with biweekly cetuximab is an encouraging<br />

strategy with efficacy and toxicity comparable to weekly cetuximab.<br />

3537 General Poster Session (Board #22H), Mon, 8:00 AM-12:00 PM<br />

Patterns <strong>of</strong> chemotherapy (CT) use in a U.S.-wide population-based cohort<br />

<strong>of</strong> patients (pts) with metastatic colorectal cancer (mCRC). Presenting<br />

Author: Thomas Adam Abrams, Dana-Farber Cancer Institute, Boston, MA<br />

Background: Few population studies have examined treatment regimens<br />

and duration in mCRC since the introduction <strong>of</strong> biologic therapies in 2004.<br />

Methods: We assessed 4,877 consecutive mCRC pts who received CT<br />

between Jan 2004 and Mar 2011 at academic, private, and community<br />

hospital-based practices participating in a US-wide CT order entry (COE)<br />

system that captures patient demographics, stage and treatment data.<br />

Multivariate analyses <strong>of</strong> prospectively recorded patient and provider characteristics<br />

identified significant predictors <strong>of</strong> specific therapeutic approaches.<br />

Results: Among all pts, median age was 64; 55% male; (ECOG<br />

PS 0/1/2�) (56%/33%/11%). In first-line CT, 17% <strong>of</strong> pts received<br />

fluoropyrimidine (FU) only, while 16% received FU and irinotecan and<br />

65% received FU and oxaliplatin. Older pts, those with poorer ECOG PS,<br />

and those treated in private practices were significantly more likely to<br />

receive FU only (all p �0.05). 53% <strong>of</strong> all pts received treatment in line 2,<br />

28% in line 3, and 13% in line 4. Mean duration <strong>of</strong> treatment was 170 days<br />

(line 1), 139 (line 2), 135 (line 3) and 126 (line 4). Bevacizumab (bev) was<br />

administered to 65% <strong>of</strong> pts at some point in their treatment. Among the<br />

51% who received bev in line 1, 33% continued bev beyond progression<br />

(BBP) in line 2. Pts treated at academic centers, pts who received longer<br />

duration <strong>of</strong> line 1 therapy, and pts whose providers had greater CRC pt<br />

volume were significantly more likely to continue BBP. Overall, 23% <strong>of</strong> pts<br />

received cetuximab (cet) (4% <strong>of</strong> all line 1 pts, 17% <strong>of</strong> line 2, 31% <strong>of</strong> line 3,<br />

and 29% <strong>of</strong> line 4). Mean duration <strong>of</strong> cet use was 168 days (line 1), 125<br />

(line 2), 133 (line 3) and 129 (line 4). Pts treated at academic centers, pts<br />

who received longer duration <strong>of</strong> line 1 therapy, and pts treated in the West<br />

US were significantly more likely to ever receive cet. Cet use decreased by<br />

18% following FDA label change restricting use to KRAS wt pts in Jun<br />

2009 (p �0.001). Of all pts, 6% received panitumumab at some point. Of<br />

those, 39% had previously received cet. Conclusions: This populationbased<br />

study provides insight into treatment patterns <strong>of</strong> mCRC in the US.<br />

Usage <strong>of</strong> biologic agents varies significantly according to patient, practice,<br />

and provider characteristics.<br />

3539 General Poster Session (Board #23B), Mon, 8:00 AM-12:00 PM<br />

Phase I study <strong>of</strong> EMD 525797 (DI17E6), an antibody targeting ���<br />

integrins, in combination with cetuximab and irinotecan, as a second-line<br />

treatment for patients with k-ras wild-type metastatic colorectal cancer.<br />

Presenting Author: Elena Elez, Vall d’Hebron University Hospital, Barcelona,<br />

Spain<br />

Background: EMD 525797 is a humanized monoclonal antibody that<br />

specifically targets all �� integrins, including ���1, ���3, ���5, ���6,<br />

and ���8. In colorectal cancer (CRC), ���3 is highly expressed on<br />

tumor-associated vessels, and ���5 is strongly expressed on tumor cells,<br />

vessels, and stromal cells. Overexpression <strong>of</strong> ���6 is associated with a<br />

significant reduction in median overall survival. Methods: Safety, tolerability,<br />

and anti-tumor activity <strong>of</strong> escalating doses <strong>of</strong> EMD525797 in combination<br />

with irinotecan and cetuximab were assessed in patients (pts) with<br />

metastatic CRC who had failed first-line fluoropyrimidine/oxaliplatincontaining<br />

chemotherapy. Pts received IV infusions over 1 hour <strong>of</strong> 250,<br />

500, 750, and 1,000 mg EMD 525797 every 2 weeks in combination with<br />

irinotecan at 180 mg/m2 IV every 2 weeks and cetuximab at 400 mg/m2 on<br />

Cycle 1, Day 1 and then 250 mg/m2 IV weekly. A standard 3�3 trial design<br />

was used. Three pts were enrolled at each dose level and if no dose limiting<br />

toxicities (DLTs) were observed, pts were enrolled at the next dose level. An<br />

additional 3 pts were added if 1 <strong>of</strong> the initial 3 pts in the cohort experienced<br />

a DLT. Results: A total <strong>of</strong> 16 pts received treatment.No DLTs were observed<br />

at any dose <strong>of</strong> EMD 525797 in combination with irinotecan and cetuximab.<br />

Serious adverse events related to EMD 525797 included Grade 3 hypokalemia<br />

and tachyarrhythmia. Dosing was well tolerated over treatment intervals<br />

as long as 18 months. Objective responses included 1 partial response<br />

(PR) at 250 mg; 1 complete response and 1 PR at 750 mg; and 2 PRs at<br />

1,000 mg. Two pts had prolonged stable disease lasting longer than 11<br />

weeks at 750 mg. Anti-tumor activity was observed in pts who had failed<br />

anti-VEGF therapy in combination with first-line chemotherapy. Conclusions:<br />

The combination <strong>of</strong> EMD 525797 with irinotecan and cetuximab was well<br />

tolerated and has anti-tumor activity. Further evaluation <strong>of</strong> this regimen is<br />

warranted and is underway in an open-label, randomized phase II study <strong>of</strong><br />

two doses <strong>of</strong> EMD 525797 (500 and 1,000 mg) combined with irinotecan<br />

and cetuximab that are compared with standard irinotecan and cetuximab.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


3540 General Poster Session (Board #23C), Mon, 8:00 AM-12:00 PM<br />

Prospective study <strong>of</strong> EGFR intron 1 CA tandem repeats to predict factor<br />

benefit from cetuximab and irinotecan. Presenting Author: Carlotta Antoniotti,<br />

U.O. Oncologia Medica 2, Azienda Ospedaliero-Universitaria Pisana,<br />

Istituto Toscano Tumori, Pisa, Italy<br />

Background: Retrospective experiences have investigated the potential<br />

influence <strong>of</strong> EGFR intron 1 CA repeats on the efficacy <strong>of</strong> cetuximabcontaining<br />

treatments. Different series, adopting different criteria to define<br />

short (S) and long (L) variants, have provided contrasting results. Methods:<br />

We designed a prospective confirmatory study, to detect a HR for PFS <strong>of</strong><br />

1.75 for L- compared to SS genotypes in a population <strong>of</strong> KRAS and BRAF<br />

wild-type irinotecan-resistant mCRC pts treated with cetuximab and<br />

irinotecan. Estimating a prevalence <strong>of</strong> 60% <strong>of</strong> the SS variant and setting a<br />

two-sided alfa�0.05 with a power <strong>of</strong> 80%, 104 events were required. We<br />

defined S and L allelic variants those presenting � and �20 CA repeats,<br />

respectively. EGFR (CA) n repeat polymorphism was assessed following a<br />

5’-end [�-33P] ATP-labeled PCR protocol. Results: One-hundred-fifteen pts<br />

were included. At a median follow up <strong>of</strong> 21.9 months, PFS and OS were 5.2<br />

and 13.4 months, respectively. Thirty-three (29%) out <strong>of</strong> 114 evaluable<br />

pts achieved response. EGFR (CA) n repeat genotype was L- and SS in 45<br />

(40%) and 68 (60%) out <strong>of</strong> 113 evaluable cases. No differences in PFS or<br />

OS were observed between L- and SS genotypes (median PFS: 4.4 vs. 5.3<br />

months, HR: 1.00 [95%CI: 0.67-1.51], p�0.991; median OS: 11.3 vs.<br />

14.2 months, HR: 1.30 [95%CI: 0.80-2.22], p�0.261). Ten (22%) out <strong>of</strong><br />

45 L- pts achieved response compared to 22 (33%) out <strong>of</strong> 67 SS pts<br />

(Fisher’s Exact test: p�0.617). Other exploratory analyses adopting<br />

different cut-<strong>of</strong>f values reported in literature led to similar results.<br />

Conclusions: This prospective study, including a clinically homogenous and<br />

molecularly selected population, does not confirm any predictive or<br />

prognostic effect for EGFR (CA) n repeat allelic variants with respect to the<br />

efficacy <strong>of</strong> cetuximab and irinotecan in advanced lines <strong>of</strong> treatment. The<br />

present experience strengthens the need <strong>of</strong> prospectively challenging<br />

retrospective findings, as an essential step on biomarkers’ way toward<br />

clinical practice.<br />

3542 General Poster Session (Board #23E), Mon, 8:00 AM-12:00 PM<br />

Utilization and outcomes <strong>of</strong> primary tumor surgery for stage IV colon cancer<br />

in the United States: A population-based study. Presenting Author: Yvonne<br />

Sada, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX<br />

Background: Stage IV colon cancer treatment may include resection <strong>of</strong> the<br />

primary tumor. Current use <strong>of</strong> primary tumor surgery (PTS) in clinical<br />

practice is unknown. This study examined utilization and determinants <strong>of</strong><br />

PTS and evaluated its effect on survival. Methods: Using national Surveillance,<br />

Epidemiology, and End Results registry data, stage IV colon cancer<br />

patients diagnosed from 1998-2008 were identified. Data on demographics,<br />

PTS, and tumor features were collected. Temporal changes in receipt <strong>of</strong><br />

PTS were examined over 3 periods (1998-2000, 2001-2004, 2005-<br />

2008). Multiple logistic regression was used to identify significant determinants<br />

<strong>of</strong> PTS. 1- and 3-year cancer-specific survival was calculated in PTS<br />

and non-PTS patients. Cox proportional hazards models examined the<br />

effect <strong>of</strong> PTS on mortality risk. Results: 16,029 patients were identified.<br />

Median age was 69 (IQR: 57-78), and 50% were male. Approximately 67%<br />

<strong>of</strong> patients received PTS. Receipt <strong>of</strong> PTS significantly declined from 72%<br />

in 1998-2000 to 68% in 2001-2004, and 63% in 2005-2008 (p�0.01).<br />

Results from the logistic regression analysis showed that patients who were<br />

younger, white, married, had right sided cancer and higher tumor grade<br />

were more likely to receive PTS (all p�0.01). The 1- and 3-year survival<br />

was higher in patients who received PTS compared with those who did not<br />

(1-year: 55% (95% CI: 54-56) vs. 24% (95% CI: 23-26); 3-year: 19%<br />

(95% CI: 19-20) vs. 4% (95%CI: 3.4-4.9)). Adjusted for demographics<br />

and tumor features, risk <strong>of</strong> mortality was 54% (HR�0.46; 95% CI:<br />

0.44-0.48) lower in patients who received PTS than those without PTS.<br />

Recent year <strong>of</strong> diagnosis (HR�0.88; 95% CI: 0.75-0.80) and being<br />

married (HR�0.90, 95% CI: 0.86-0.95) were associated with lower<br />

mortality. Older age (HR�1.48; 95% CI: 1.39-1.56), black race (HR�1.09;<br />

95% CI: 1.03-1.15), right sided cancer (HR�1.21; 95% CI: 1.17-1.26),<br />

and poorly differentiated tumors (HR� 1.62; 95% CI: 1.46-1.80) were<br />

associated with increased mortality. Conclusions: PTS utilization for stage<br />

IV colon cancer has significantly declined, yet survival was higher in<br />

patients who received PTS. However, these findings are limited by the<br />

absence <strong>of</strong> co-morbidity and chemotherapy data.<br />

Gastrointestinal (Colorectal) Cancer<br />

213s<br />

3541 General Poster Session (Board #23D), Mon, 8:00 AM-12:00 PM<br />

Capecitabine (cape)-associated hand-foot skin reaction (HFS) as a clinical<br />

predictor <strong>of</strong> improved survival in patients (pts) with colorectal cancer.<br />

Presenting Author: Ralf H<strong>of</strong>heinz, Day Treatment Centre at the Interdisciplinary<br />

Tumour Centre Mannheim, Universitätsmedizin Mannheim, Mannheim,<br />

Germany<br />

Background: HFS is frequently observed during treatment with cape. Post<br />

hoc analyses <strong>of</strong> the X-ACT trial suggested that HFS may be associated with<br />

improved prognosis in cape recipients in stage III colon cancer. In the<br />

present analysis we evaluated the potential association <strong>of</strong> HFS and survival<br />

in pts with metastatic colorectal cancer and locally advanced rectal cancer.<br />

Methods: Pts receiving cape within AIO KRK-0104 and Mannheim rectal<br />

cancer trials were analyzed. HFS was graded according to NCI-CTC. Time to<br />

first occurrence <strong>of</strong> HFS was described per cycle and HFS developing during<br />

cycles 1 and 2 was defined as “early HFS”. Baseline characteristics in the<br />

groups <strong>of</strong> pts with or without HFS were compared using standard tests.<br />

Toxicities observed in both groups were compared with Fisher’s exact test.<br />

Progression-free (PFS) or disease-free (DFS) and overall survival (OS) data<br />

from both trials were pooled and the HFS group was compared to the<br />

non-HFS using Kaplan-Meier analysis. Results: A total <strong>of</strong> 374 pts are<br />

included, <strong>of</strong> whom 29.3% developed HFS. Of these, 70.9% had “early<br />

HFS”. Baseline characteristics were comparable between the HFS and the<br />

non-HFS groups concerning age, gender, ECOG status and UICC stage. On<br />

multivariate analysis none <strong>of</strong> these factors had influence on the occurrence<br />

<strong>of</strong> HFS. The percentage <strong>of</strong> all-grade hematological toxicities did not differ<br />

between both groups. Contrarily, patients in the HFS group had a<br />

significantly higher rate <strong>of</strong> all grade (but not grade 3-4) diarrhea, stomatitis/<br />

mucositis and fatigue (p�0.01 resp.). Moreover, pts in the HFS group had<br />

improved PFS/DFS (29.0 vs. 11.4 months; p�0.015, HR 0.69) and OS<br />

(75.8 vs. 41.0 months; p�0.001, HR�0.56). Within the HFS group the<br />

PFS/DFS and OS were comparable between pts in the “early” and the “late<br />

HFS” groups. Conclusions: This analysis provides further evidence for the<br />

association <strong>of</strong> HFS and survival in patients with colorectal cancer. Baseline<br />

characteristics and the time <strong>of</strong> occurrence <strong>of</strong> HFS had no impact on<br />

survival. Patients developing HFS had a higher probability <strong>of</strong> developing<br />

any-grade gastrointestinal toxicity and fatigue while no correlation with<br />

hematological toxicity was noticed.<br />

3543 General Poster Session (Board #23F), Mon, 8:00 AM-12:00 PM<br />

Phase Ib study <strong>of</strong> dulanermin combined with FOLFIRI (with or without<br />

bevacizumab [BV]) in previously treated patients (Pts) with metastatic<br />

colorectal cancer (mCRC). Presenting Author: Saifuddin M. Kasubhai,<br />

Northwest Medical Specialties, Tacoma, WA<br />

Background: Dulanermin is a recombinant soluble human Apo2 ligand/TNFrelated<br />

apoptosis-inducing ligand (Apo2L/TRAIL) that activates apoptotic<br />

pathways by binding to the pro-apoptotic death receptors DR4 and 5. This<br />

is the final update <strong>of</strong> the study assessing the safety <strong>of</strong> dulanermin<br />

combined with FOLFIRI (� BV) in previously treated mCRC pts. Methods:<br />

This was a multicenter, open-label, dose-escalation and cohort expansion<br />

study. Dulanermin was administered intravenously in 14-day cycles on<br />

Days 1, 2 and 3 at 9 mg/kg or 18 mg/kg, with FOLFIRI (� BV in pts not<br />

previously treated with BV) on Day 1. Dose-limiting toxicity (DLT) was<br />

assessed through 2 cycles (28 days). Adverse events (AE) were recorded<br />

through all cycles. Response was assessed with RECIST (v1.0). Results: A<br />

total <strong>of</strong> 27 pts received 1-48 cycles (median 10) <strong>of</strong> dulanermin with<br />

FOLFIRI (1 pt received BV). No DLTs were reported and no relationship<br />

between dulanermin dose level and AE incidence or severity was detected.<br />

The most frequent AEs reported as dulanermin-related were fatigue (37%),<br />

anemia (19%), diarrhea, nausea, stomatitis, and weight decreased (15%<br />

each). The most frequent Grade �3 AEs reported as dulanermin-related<br />

were fatigue (15%), anemia, febrile neutropenia and vomiting (7% each).<br />

Serious AEs reported as dulanermin-related included: vomiting, dehydration,<br />

and febrile neutropenia (1 pt each). Some <strong>of</strong> these events were<br />

reported as related to dulanermin and other study drugs. One pt died on<br />

study due to disease progression. Other reasons for discontinuation <strong>of</strong> study<br />

treatment included disease progression (21 pts, 78%), and physician’s<br />

decision and pt’s decision (2 pts each, 7%). One pt is still receiving<br />

treatment. Among the 27 pts, best responses included 6 (22%) partial<br />

responses (5 confirmed, 1 unconfirmed), 17 (63%) stable disease, 3<br />

(11%) progressive disease and 1 (4%) pt with no on-study tumor assessment.<br />

Conclusions: The addition <strong>of</strong> dulanermin to FOLFIRI (� BV) was well<br />

tolerated in previously treated mCRC pts. AEs were similar to those<br />

previously reported for the chemotherapy alone. Tumor responses were<br />

consistent with treatment response to FOLFIRI in this patient population.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


214s Gastrointestinal (Colorectal) Cancer<br />

3544 General Poster Session (Board #23G), Mon, 8:00 AM-12:00 PM<br />

Effect <strong>of</strong> bevacizumab on rate <strong>of</strong> oxaliplatin-induced thrombocytopenia and<br />

splenomegaly. Presenting Author: Kanwal Pratap Singh Raghav, University<br />

<strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: Oxaliplatin-based chemotherapy can cause hepatic sinusoidal<br />

injury (HSI) leading to portal hypertension. This results in splenic sequestration<br />

<strong>of</strong> platelets and thrombocytopenia. Evidence suggests that bevacizumab<br />

may protect against HSI, although the clinical impact <strong>of</strong> this has not<br />

been well examined. The purpose <strong>of</strong> our study was to evaluate rate <strong>of</strong><br />

thrombocytopenia and splenomegaly as clinical end-points indicative <strong>of</strong><br />

bevacizumab mediated modulation <strong>of</strong> oxaliplatin-induced hepatic toxicity.<br />

Methods: We performed a retrospective review <strong>of</strong> metastatic colorectal<br />

cancer patients treated at M D Anderson Cancer Center from 1/2003 to<br />

1/2010 with � 3 months <strong>of</strong> frontline fluoropyrimidine and oxaliplatin<br />

(FOLFOX) with or without bevacizumab. Patients with prior liver disease,<br />

liver surgery or absence <strong>of</strong> spleen were excluded. Splenic volumes pre,<br />

during and post FOLFOX therapy were determined with CT scans and<br />

correlated with platelet counts and treatment. Results: A total <strong>of</strong> 184<br />

patients [Bev cohort: FOLFOX/Bev (n �138); Non-Bev cohort: FOLFOX<br />

alone (n � 46)] were identified. Baseline characteristics (age, gender,<br />

spleen size and platelet counts) were similar in both groups. Median<br />

number <strong>of</strong> oxaliplatin cycles for each cohort was 9.00 (p � 0.9). The<br />

median total oxaliplatin dose did not differ between the two cohorts (p �<br />

0.9). Development <strong>of</strong> splenomegaly (volume increase � 30%) was more<br />

common in the Non-Bev cohort (48% vs. 32%; p � 0.013). The median<br />

time to develop splenomegaly was significantly longer in the Bev cohort<br />

(7.6 vs. 5.5 months; p � 0.023). Splenomegaly correlated with a higher<br />

rate <strong>of</strong> thrombocytopenia (defined as platelets � 150K) at 3 months (40%<br />

vs. 16%, p � 0.0005) and beyond (during 3 to 6 months <strong>of</strong> treatment, p �<br />

0.0001). Correspondingly, bevacizumab therapy was associated with lower<br />

rates <strong>of</strong> thrombocytopenia at 3 months (24% in Bev cohort vs. 43% in<br />

Non-Bev cohort, p � 0.006) and beyond (p � 0.003). Conclusions: In our<br />

cohort, addition <strong>of</strong> bevacizumab to oxaliplatin chemotherapy reduces the<br />

occurrence <strong>of</strong> splenic enlargement, which correlates with a decreased rate<br />

<strong>of</strong> thrombocytopenia. Further studies to understand the potential hepatoprotective<br />

mechanism <strong>of</strong> anti-VEGF therapy are needed.<br />

3546 General Poster Session (Board #24A), Mon, 8:00 AM-12:00 PM<br />

KSR1 gene polymorphism in mCRC patients treated with first-line FOLFIRI<br />

and bevacizumab. Presenting Author: Marta Schirripa, U.O. Oncologia<br />

Medica 2, Azienda Ospedaliero-Universitaria Pisana, Istituto Toscano<br />

Tumori, Pisa, Italy<br />

Background: Kinase suppressor <strong>of</strong> Ras (KSR) 1 is a scaffolding protein<br />

regulating the Raf/Mek/ERK cascade. Preclinical data showed that KSR1<br />

could increase estrogen receptor transcriptional activity after exposure to<br />

estrogens. Recent retrospective analyses suggested a possible role for<br />

KSR1 rs2241906 C/T polymorphism in predicting the outcome <strong>of</strong> patients<br />

with mCRC. Methods: MCRC patients receiving first-line<br />

FOLFIRI�bevacizumab prospectively enrolled in a translational research<br />

program were selected on the basis <strong>of</strong> KRAS and BRAF mutational status<br />

availability. KSR1 rs2241906 polymorphism was analyzed on DNA extracted<br />

from peripheral blood by means <strong>of</strong> PCR and direct sequencing.<br />

Taking into account the connection between estrogen pathway and KSR1,<br />

subgroup analyses according to gender were pre-planned. Results: 287<br />

patients were included. Main patients’ characteristics were the following:<br />

M/F�175/112; median age�62 (range 26-79); ECOG-PS 0/1-2�240/47;<br />

synchronous/metachronous disease�213/74; Köhne score (low/intermediate/high/data<br />

missing)�122/129/22/14; KRAS-BRAF mutational status<br />

(wt-wt/mut-wt/wt-mut)� 123/146/18. In the overall KRAS-BRAF wt population,<br />

KSR1 polymorphism did not affect the outcome. The analysis<br />

performed according to gender showed that in the KRAS-BRAF wt population<br />

females with KSR1 rs2241906 T/- achieved a significantly better PFS<br />

(median 15.9 mos) in comparison to C/C variant carriers (median 8.8 mos)<br />

(HR�0.44 [95%CI 0.21-0.91], p�0.010); meanwhile males with T/showed<br />

a worse PFS in comparison to those carrying the C/C variant<br />

(HR�1.92 [95%CI 1.05-3.53], p�0.021). These results were significant<br />

also in a multivariate model and a significant interaction <strong>of</strong> gender with PFS<br />

according to KSR1 allelic variants was demonstrated (p�0.004).<br />

Conclusions: This prospectively conceived pharmacogenetic study confims<br />

the role <strong>of</strong> KSR1 polymorphisms in affecting the outcome <strong>of</strong> mCRC<br />

KRAS-BRAF wt patients. In particular, these results indirectly indicate that<br />

estrogenic stimulation could affect the proliferation <strong>of</strong> KRAS-BRAF wt CRC<br />

cells through an interaction with KSR1. Preclinical investigations to further<br />

explore this preliminary hypothesis are ongoing.<br />

3545 General Poster Session (Board #23H), Mon, 8:00 AM-12:00 PM<br />

Comparative effectiveness <strong>of</strong> sphincter-sparing surgery (SSS) versus abdomino-perineal<br />

resection (APR) in rectal cancer: Patient-reported outcomes<br />

(PROs) from NSABP R-04. Presenting Author: Patricia A. Ganz,<br />

University <strong>of</strong> California, Los Angeles Jonsson Comprehensive Cancer<br />

Center, Los Angeles, CA<br />

Background: R-04 is a trial <strong>of</strong> pre-surgical RT and either capecitabine or<br />

5-FU with or without oxaliplatin in patients (Pts) with resectable rectal<br />

cancer. PROs were measured before treatment, post-RT, and 1 yr post-op.<br />

We compare PROs at 1 yr by type <strong>of</strong> surgery with hypothesis that APR Pts<br />

would have worse quality <strong>of</strong> life (QOL). Methods: Pts completed the FACT-C<br />

and EORTC-CR38 at all times. Baseline and 1 yr were compared within<br />

groups (SSS and APR) with paired t-test, and between groups at 1 yr, with<br />

adjustment for covariates <strong>of</strong> age, gender, clinical stage, baseline score, and<br />

surgery intent in a general linear model. These secondary/exploratory<br />

hypotheses were significant if p � 0.05. Results: 1,608Pts were randomized<br />

and 1405 completed baseline QOL form after consent, prior to<br />

treatment. 1,003 completed QOL form 1 yr post-op: 6 were ineligible, 10<br />

did not have surgery, leaving 987 Pts. 615 had SSS and 372 had APR.<br />

66.6% were male, 61.5% stage II, and almost all had post-surgical<br />

adjuvant chemotherapy. SSS Pts were significantly younger (60.3% vs.<br />

53.5% � 59 yr, p�0.04). FACT-C total and subscale scores were not<br />

significantly different by surgery type at 1 yr, with only minimal decline<br />

from baseline in both groups. For SSS Pts, EORTC-CR38 scores significantly<br />

worsened for body image, sexual function, sexual enjoyment (all p<br />

�0.0001), while future perspective improved (p�0.0001) at 1 yr from<br />

baseline. All symptom subscales except weight loss and micturition showed<br />

worsened symptoms (GI tract, chemo side effects, defecation problems,<br />

sexual problems) all p � 0.0001 at 1 yr from baseline. Compared to APR<br />

Pts, SSS had less severe functional problems and symptoms, especially in<br />

sexual enjoyment, weight loss, GI symptoms, and body image, all significant.<br />

Conclusions: Contrary to prediction, there were no significant differences<br />

in FACT-C scores for SSS vs. APR. Symptoms and functional<br />

problems were detected by the EORTC-CR38, reflecting expected differences.<br />

The largest differences between the surgical treatments were for GI<br />

symptoms and body image. Information from these PROs may be useful in<br />

counseling Pts anticipating surgery for rectal cancer.<br />

3547 General Poster Session (Board #24B), Mon, 8:00 AM-12:00 PM<br />

First European phase II trial <strong>of</strong> intravenous (iv) cetuximab (Cet) and hepatic<br />

artery infusion (HAI) <strong>of</strong> irinotecan, 5-fluorouracil and oxaliplatin in patients<br />

with unresectable liver metastases from wt KRAS colorectal cancer (CRC)<br />

after systemic therapy failure (OPTILIV, NCT00852228). Presenting<br />

Author: Francis Levi, Medical Oncology Department, INSERM U776, Paul<br />

Brousse Hospital, Villejuif, France<br />

Background: HAI <strong>of</strong> chronomodulated (Chrono) irinotecan (I), 5-Fluorouracil<br />

(F) and oxaliplatin (O), or flat O combined with iv F-leucovorin allowed<br />

secondary metastases resections and prolonged survival in patients (pts)<br />

with CRC liver metastases despite prior chemotherapy failure (Bouchahda,<br />

Cancer 2009; Goere, Ann Surg 2010). Purpose: To prospectively evaluate<br />

safety and efficacy <strong>of</strong> combining iv Cet with HAI <strong>of</strong> IFO in pts with CRC liver<br />

metastases. Methods: This Phase II trial involved previously treated pts with<br />

unresectable CRC liver metastases. Pts received iv Cet (500 mg/m²) and<br />

Chrono or conventional HAI <strong>of</strong> I (180 mg/m²), F (2800 mg/m²), and O (85<br />

mg/m²) q2 weeks. Liver metastases were resected if adequately downstaged.<br />

Results: Planned accrual <strong>of</strong> 60 pts was reached on 01/24/2012. 3<br />

pts were not treated, 9 are ongoing. 48 consecutive treated pts (18F, 30 M;<br />

aged 32-76 years) are fully assessed and monitored. They had PS 0-1<br />

(98%), bilobar liver lesions (69%), a median <strong>of</strong> 8 metastases (1-50; largest<br />

diameter, 57 mm – 15-172). Prior chemotherapy involved one (43%), or<br />

two or three (57%) lines. A median <strong>of</strong> 5 protocol courses (1-13) was given.<br />

Main grade 3-4 toxicities per pt were neutropenia (40%), abdominal pain<br />

(15%), fatigue (15%), nausea (15%), diarrhea (13%) and sensory neuropathy<br />

(4%). Objective response rate was 44% [30-58], with 6% radiological<br />

complete responses. Disease control rate was 85%. Secondary liver surgery<br />

was performed in 15 pts (31.3%).One pt with 25 metastases (1-6 cm) in all<br />

liver segments had pathologic complete response in 24 lesions removed<br />

through three-stage hepatectomy. She currently has an <strong>of</strong>f treatment<br />

disease-free survival <strong>of</strong> 17� months.With a follow up <strong>of</strong> 4 to 47 months,<br />

median progression-free survival is 14.2 months [9.7-18.8], 1- and 2-year<br />

survival rates are 92.8% and 59.2% respectively. Conclusions: OPTILIV<br />

<strong>of</strong>fers a safe and unusually effective treatment option for patients with CRC<br />

liver metastases after failure <strong>of</strong> systemic chemotherapy within a coordinated<br />

medico-surgical strategy.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


3548 General Poster Session (Board #24C), Mon, 8:00 AM-12:00 PM<br />

Plasma concentrations <strong>of</strong> YKL-40 in chemo-naive patients with metastatic<br />

colorectal cancer treated with FLOX with or without cetuximab: Results<br />

from the NORDIC VII study. Presenting Author: Line Schmidt Tarpgaard,<br />

Department <strong>of</strong> Oncology, Odense University Hospital, Odense, Denmark<br />

Background: KRAS status is presently the best biomarker to select patients<br />

with metastatic colorectal cancer (mCRC) for therapy with EGFR inhibition<br />

even though not confirmed in all phase III studies. In the NORDIC VII study<br />

a survival benefit <strong>of</strong> adding cetuximab to the Nordic FLOX regimen could<br />

not be confirmed. Identification <strong>of</strong> new predictive and prognostic biomarkers<br />

is essential. Plasma concentration <strong>of</strong> YKL-40 is emerging as a new<br />

biomarker in patients with cancer and inflammatory diseases. We tested<br />

the hypothesis that high plasma YKL-40 associates with short progression<br />

free survival (PFS) and short overall survival (OS) in patients included in the<br />

NORDIC VII Study. Methods: 566 patients with mCRC were randomized to:<br />

A) Nordic FLOX; B) FLOX � cetuximab; and C) FLOX for 16 weeks �<br />

cetuximab continuously. Plasma samples were available from 510 patients<br />

(90%). Pretreatment plasma YKL-40 was determined by ELISA (Quidel),<br />

and the plasma YKL-40 concentration was dichotomized according to the<br />

age-corrected 95% YKL-40 level in 3130 healthy subjects. Results: Plasma<br />

YKL-40 was elevated in 204 patients (40%) and the median plasma<br />

YKL-40 was higher in the study group compared to healthy subjects (120<br />

mg/l vs. 40 mg/l, p�0.001). Elevated plasma YKL-40 was associated with<br />

short PFS (HR�1.25, 95% CI 1.04-1.51, p�0.02). This relationship was<br />

demonstrated only in patients treated with FLOX chemotherapy alone<br />

(HR�1.42, 1.02-1.98, p�0.04). High plasma YKL-40 was associated<br />

with short OS in all patients (HR�1.55, 1.25-1.92, p�0.001) and in the<br />

different treatment groups (A: HR�1.54, 1.05-2.26, p�0.03; B:<br />

HR�1.40, 0.97-2.01, p�0.07; C: 1.79, 1.23-2.60, p�0.002). Multivariate<br />

analysis (YKL-40, performance status, number <strong>of</strong> metastatic sites)<br />

demonstrated that elevated plasma YKL-40 was an independent biomarker<br />

<strong>of</strong> short OS (HR�1.46, 1.17-1.81, p�0.001). Conclusions: Plasma<br />

YKL-40 may be a new prognostic biomarker in patients with mCRC treated<br />

with 1st line FLOX chemotherapy, with or without cetuximab. The predictive<br />

value <strong>of</strong> plasma YKL-40 is not yet clarified.<br />

3550 General Poster Session (Board #24E), Mon, 8:00 AM-12:00 PM<br />

Adjuvant chemotherapy (ACT) in stage II colon cancer (CC) in patients with<br />

Lynch syndrome. Presenting Author: Karsten Schulmann, Ruhr University<br />

Bochum, Knappschaftskrankenhaus Med. Dpt., Bochum, Germany<br />

Background: Previous studies showed conflicting results regarding the value<br />

<strong>of</strong> ACT in MSI-H CC. A recent study reported differential benefits from<br />

5-FU-based ACT comparing suspected sporadic vs suspected hereditary<br />

MSI-H CC. We sought to evaluate the prognostic impact <strong>of</strong> ACT in a large<br />

cohort <strong>of</strong> Lynch syndrome (LS) patients (pts) with stage II CC. Methods: To<br />

minimize selection bias diagnoses �2 years prior to registration in the<br />

database <strong>of</strong> the German HNPCC consortium were excluded. 278 patients<br />

(61% male, mean age 42.9y, 13% stage IIB, 51% with MMR gene<br />

mutation) were eligible. Overall Survival (OS), CC-specific Survival (CSS),<br />

and Disease Free Survival (DFS) were analyzed using Kaplan-Meier and Cox<br />

Regression analyses. Results: 5y OS, CSS and DFS were 95%, 95% and<br />

93% respectively. Right-sided CC was independently associated with lower<br />

DFS in stage II and IIA. Increasing age was associated with lower OS, CSS<br />

and DFS in stage IIA, however we observed only trends in the multivariate<br />

analysis. Surgery alone (without ACT) was associated with a slightly lower<br />

OS in stage IIA (univariate HR 3,659; 95% CI 0,81-16,5; P�0.092); but<br />

not with lower DFS and CSS. Prognosis was not different comparing<br />

FOLFOX vs. 5-FU-based ACT. Conclusions: Our data suggest that LS pts<br />

with stage II CC do not benefit from ACT. FOLFOX was not superior to<br />

5-FU-based ACT. If our results are confirmed, LS pts with stage IIA CC<br />

should not receive ACT. The group <strong>of</strong> stage IIB CC was too small to make<br />

definite conclusions.<br />

OS<br />

Univariate<br />

CSS DFS<br />

HR 95% CI HR 95% CI HR 95% CI<br />

Stage II<br />

(n�278)<br />

Age Per 10y 1,034 0,993-1,08 1,034 0,993-1,08 1,039 0,997-1,08 &<br />

Localization Right<br />

Left 1,58 0,64-3,90 1,58 0,64-3,890 2,78 1,10-6,98 *<br />

T-stage T3<br />

T4 2,12 0,69-6,51 2,12 0,69-6,51 2,66 0,85-8,37 &&<br />

ACT No<br />

Yes 1,37 0,50-3,70 1,37 0,50-3,70 0,99 0,35-2,78<br />

ACT 5-FU<br />

FOLFOX<br />

0,04 0-887 0,04 0-887 1,82 0,33-9,93<br />

Stage IIA<br />

(n�242)<br />

Age Per 10y 1,049 1,000-1,100 ** 1,067 1,000-1,139 *** 1,052 1,003-1,104 §<br />

Localization Right<br />

Left 1,474 0,526-4,131 2,730 0,760-9,805 3,039 1,069-8,683 #<br />

ACT No<br />

Yes 3,659 0,808-16,562 ## 3,974 0,477-33,070 1,566 0,415-5,908<br />

ACT 5-FU<br />

FOLFOX 0,046 0.000-.... 0,045 0.000-.... 1,871 0,169-20,668<br />

*p�0.03; ** p�0.051; *** p� 0.052; § p�0.039; # p� 0.037; ## p�0.092; & p�0.07; && p�0.081.<br />

Gastrointestinal (Colorectal) Cancer<br />

215s<br />

3549 General Poster Session (Board #24D), Mon, 8:00 AM-12:00 PM<br />

Primary side <strong>of</strong> origin affects the outcome <strong>of</strong> mCRC patients treated with<br />

first-line FOLFIRI plus bevacizumab independently <strong>of</strong> BRAF status and<br />

mucinous histology. Presenting Author: Fotios Loupakis, University <strong>of</strong><br />

Southern California Norris Comprehensive Cancer Center, Los Angeles, CA<br />

Background: Several studies tried to address the question <strong>of</strong> whether<br />

primary site <strong>of</strong> CRC could affect the outcome. Those analyses were limited<br />

by small sample size numbers and/or high degree <strong>of</strong> heterogeneity in stage<br />

and received treatments and/or limited information regarding molecular<br />

and pathologic features. Mucinous histology and BRAF mutations are more<br />

frequent in right-sided tumors and are both associated with worse outcome<br />

in the metastic setting. Methods: the analysis was conducted on a database<br />

<strong>of</strong> 455 mCRC prospectively enrolled in a pharmacogenetic translational<br />

study. Patients (pts) were treated with FOLFIRI/bevacizumab. Pts were<br />

excluded if data regarding mucinous histology and/or BRAF mutational<br />

status were lacking or if affected by synchronous right and left-sided<br />

tumors. Results: 200 pts were identified and included in the study.<br />

Mucinous histology, BRAF mutation and right-side primary were found in<br />

35 (17.5%), 14 (7%) and 56 (28%) patients respectively. Right-side<br />

tumors were more frequently BRAF mutated (p�0.001) and the association<br />

was still significant when adjusting for mucinous histology (p�0.001).<br />

There was a trend in the association between mucinous and side (p�0.082).<br />

At a multivariate analysis, pts with right-side primary were at higher risk <strong>of</strong><br />

progression (HR�1.88 [95%CI: 1.25-2.84], p�0.0026) and death<br />

(HR�2.07 [95%CI: 1.23-3.49], p�0.006). At a prespecified analysis in<br />

the subgroup <strong>of</strong> non-mucinous and BRAF wild-type tumors, pts with<br />

right-side primary achieved median PFS and OS <strong>of</strong> 10.0 and 28.9 mos<br />

compared to 13.0 and 47.6 mos <strong>of</strong> left-side (PFS, right vs left: HR�1.87<br />

[95%CI: 1.19-2.91], p�0.003; OS: HR�1.91 [95%CI: 1.07-3.39],<br />

p�0.022). This was again confirmed in a multivariate model. Conclusions:<br />

These data underline a possible strong impact <strong>of</strong> side on the outcome <strong>of</strong><br />

mCRC treated with first-line FOLFIRI plus bevacizumab. To give a biological<br />

explanation for these results, a GWAS on germinal DNA and geneexpression<br />

analyses on tumors are ongoing. In order to clarify the prognostic<br />

vs predictive role to antiangiogenic treatments <strong>of</strong> this feature analyses from<br />

2 phase III randomized trials have been planned.<br />

3551 General Poster Session (Board #24F), Mon, 8:00 AM-12:00 PM<br />

An 18-gene blood-based IVD test for colorectal cancer early detection with<br />

high sensitivity and specificity. Presenting Author: Ye Xu, Fudan University<br />

Cancer Hospital, Shanghai, China<br />

Background: Colorectal cancer (CRC) is <strong>of</strong>ten curable particularly diagnosed<br />

at early stages. Different screening strategies are in place in various<br />

counties. However, the compliance rate with current CRC screening<br />

recommendations remains poor. A simple blood-based IVD test would<br />

represent an interesting alternative. We previously reported a 100-gene<br />

signature for early detection <strong>of</strong> CRC using microarray technology. Based on<br />

this result, we have developed a new blood test with real-time PCR<br />

technology. Methods: 144 CRC cases and 153 Controls were enrolled. A<br />

total <strong>of</strong> 52 genes were selected as candidate markers to be transferred from<br />

microarray to real-time PCR. The gene expression pr<strong>of</strong>iles from 100 CRC<br />

cases and 100 Controls were used as a training set for signature discovery.<br />

Signature identification process was conducted by Minimum Redundancy<br />

Maximum Relevance (mRMR) feature selection method and Support Vector<br />

Machine (SVM) classification algorithm under the Leave-One-Out Cross<br />

Validation (LOOCV) framework. The optimal size <strong>of</strong> signature was determined<br />

by a stepwise inclusion selection procedure which started with one<br />

gene and repeatedly added genes one by one until all genes were included.<br />

The performance <strong>of</strong> each N-gene signature by the LOOCV was estimated<br />

and used to determine the best size <strong>of</strong> signature. Results: The gene<br />

expression pr<strong>of</strong>iles measured by microarray and real-time PCR technology<br />

were highly comparable and resulted in a significant correlation in term <strong>of</strong><br />

fold change ratio between CRC cases and Controls (Pearson’s Correlation<br />

Coefficient � 0.95). An 18-gene signature was identified and validated in<br />

an independent validation set with 44 CRC cases and 53 Controls. The<br />

performance <strong>of</strong> 18-gene signature in the validation set reached 88.7%<br />

accuracy (95%CI: 0.80-0.94), 88.6% sensitivity (95%CI: 0.75-0.96),<br />

and 88.7% specificity (95%CI: 0.76-0.95). Conclusions: Our new results<br />

demonstrated the feasibility <strong>of</strong> technology transfer from microarray to<br />

real-time PCR. The blood test could be <strong>of</strong>fered to individuals who are<br />

unwilling or unable to undergo colonoscopy to increase the CRC screening<br />

compliance rate. Meanwhile, its high specificity could also help to avoid<br />

unnecessary colonoscopies.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


216s Gastrointestinal (Colorectal) Cancer<br />

3552 General Poster Session (Board #24G), Mon, 8:00 AM-12:00 PM<br />

Phase Ib study <strong>of</strong> dulanermin combined with first-line FOLFOX plus<br />

bevacizumab (BV) in patients (Pts) with metastatic colorectal cancer<br />

(mCRC). Presenting Author: Mark Kozl<strong>of</strong>f, Ingalls Hospital and University<br />

<strong>of</strong> Chicago, Harvey, IL<br />

Background: Dulanermin is a recombinant soluble human Apo2 ligand/TNFrelated<br />

apoptosis-inducing ligand (Apo2L/TRAIL) that activates apoptotic<br />

pathways by binding to the pro-apoptotic death receptors DR4 and 5. This<br />

study assessed the safety <strong>of</strong> dulanermin combined with FOLFOX�BV as<br />

first-line therapy in mCRC pts. Methods: This was a multicenter, open-label<br />

dose-escalation and cohort expansion study with treatment until disease<br />

progression or treatment intolerance. Dulanermin was administered intravenously<br />

in 14-day cycles on Days 1, 2 and 3 at 4.5 mg/kg or 9 mg/kg, with<br />

FOLFOX, and BV 5 mg/kg on Day 1. Dose-limiting toxicity (DLT) was<br />

assessed through 2 cycles (Days 1-28). Adverse events (AE) were recorded<br />

through all cycles. Response was assessed with RECIST (v1.0). Results: A<br />

total <strong>of</strong> 23 pts received 2-33 cycles (median 15) <strong>of</strong> dulanermin with<br />

FOLFOX�BV. No DLTs were reported. The most frequent AEs reported as<br />

dulanermin-related were fatigue (26%), neutropenia and nausea (22%<br />

each), and alanine aminotransferase increased and diarrhea (17% each).<br />

The most frequent Grade �3 AEs reported as dulanermin-related were<br />

neutropenia (13%) and infusion-related reaction (9%). Some <strong>of</strong> these<br />

events were reported as related to dulanermin and other study drugs. Three<br />

pts (13%) discontinued study treatment due to serious AEs: infusionrelated<br />

reaction (2 pts, 9%; both reported as dulanermin-related) and<br />

pulmonary embolism (1 pt, 4%). Other reasons for discontinuation included<br />

disease progression (13 pts, 57%), physician’s decision and pt’s<br />

decision (2 pts each, 9%), and non-compliance (1 pt, 4%). Two pts are still<br />

receiving treatment. Among the 23 patients, best responses included 13<br />

(57%) partial responses (9 confirmed and 4 unconfirmed), 7 (30%) stable<br />

disease and 3 (13%) progressive disease. Conclusions: Addition <strong>of</strong> dulanermin<br />

to first-line FOLFOX�BV was well tolerated overall in these mCRC pts.<br />

AEs were similar to those previously reported for FOLFOX�BV alone. Tumor<br />

responses suggest no adverse interactions between dulanermin and the<br />

standard first-line treatment evaluated in this study.<br />

3554 General Poster Session (Board #25A), Mon, 8:00 AM-12:00 PM<br />

Prognostic impact <strong>of</strong> miR-146 polymorphism in patients with resected<br />

colorectal cancer. Presenting Author: Jong Gwang Kim, Kyungpook National<br />

University Hospital, Daegu, South Korea<br />

Background: MicroRNAs (miRNAs) are small noncoding RNAs with regulatory<br />

functions as tumor suppressors and oncogenes. The rs2910164 is a C<br />

to G polymorphism located within the sequence <strong>of</strong> miR-164a precursor,<br />

which leads to a change from a C:U pair to a G:U mismatch in its stem<br />

region. Recent evidence suggested that the rs2910164 SNP in miR-146a<br />

was associated with development <strong>of</strong> familial breast and ovarian cancers,<br />

and prostatic cancer. The aim <strong>of</strong> this study was to investigate the<br />

association between this genetic variant and prognosis <strong>of</strong> colorectal cancer<br />

(CRC) operated curatively. Methods: A total <strong>of</strong> 343 CRC patients underwent<br />

curative surgery were consecutively enrolled between 2004 and 2006.<br />

DNA was extracted from fresh frozen normal tissue and miR-146 polymorphism<br />

was genotyped by polymerase chain reaction-restriction fragment<br />

length polymorphism (PCR-RFLP). Results: With a median follow up <strong>of</strong><br />

42.3 months, the combined GG�CG genotype demonstrated a better<br />

survival outcome compared with the CC genotype in a Kaplan-Meier<br />

survival analysis. Multivariate analysis showed that the G allele <strong>of</strong> miR-146<br />

was associated with better progression-free and disease-specific survival as<br />

a dominant model adjusted to age, sex, histologic grade and stage<br />

[HR�0.54 and 0.49; p � 0.018 and 0.025, respectively]. Moreover, the<br />

tumors containing the G allele were histologically associated with more<br />

prominent lymphovascular invasion. Conclusions: Our results suggest that<br />

miR-146 polymorphism is possible prognostic marker in operated CRC<br />

patients in Korean.<br />

3553 General Poster Session (Board #24H), Mon, 8:00 AM-12:00 PM<br />

Influence <strong>of</strong> BRAF mutations and RAC1b/RAC1 mRNA expression ratio on<br />

outcome in patients with metastatic colorectal cancer (mCRC) treated with<br />

first-line chemotherapy. Presenting Author: Virginia Alonso-Espinaco, Hospital<br />

Clínic, Barcelona, Spain<br />

Background: Metastatic colorectal cancer (mCRC) patients (pts), with BRAF<br />

tumor mutation (V600E) present poor overall survival (OS). RAC1b, a RAC1<br />

spliced variant, is overexpressed in CRC, and impairs apoptosis by<br />

activation <strong>of</strong> nuclear-factor-KB. Because RAC1b and BRAF (V600E) are<br />

significantly associated in CRC (Matos et al, 2008), we evaluated if<br />

RAC1b/RAC1 mRNA expression ratio (RNA ER) was an independent<br />

prognostic factor in mCRC. Methods: We analyzed tumor samples from 186<br />

mCRC pts, treated in first-line therapy with FOLFOX or CAPOX in three<br />

Spanish hospitals. We examined BRAF (V600E) mutational status by<br />

allelic discrimination, RAC1b/RAC1 expression ratio by mRNA RT-PCR<br />

(quantitative real time polymerase chain reaction) and mismatch repair<br />

(MMR) deficiency by microsatellite instability (MSI) analysis. We assessed<br />

whether these biomarkers were independently predictive <strong>of</strong> response rate<br />

(RR), progression free survival (PFS) or OS. Results: 88% <strong>of</strong> samples were<br />

informative for BRAF, 75% for MMR status and 85% for RAC1b/RAC1<br />

(RNA ER). BRAF (V600E) was found in 7%, MSI-H in 5% and high<br />

�0.9-fold RAC1b/RAC1 (RNA ER) in 20% <strong>of</strong> pts. Five <strong>of</strong> 11 pts (46%) with<br />

BRAF mutation had high RAC1b/RAC1 (RNA ER) compared with 25/144<br />

(18%) without BRAF mutation (p�0.036). All pts with BRAF mutation<br />

were MMR and none <strong>of</strong> the MSI-H pts, showed high RAC1b/RAC1 (RNA<br />

ER). Patients with low RAC1b/RAC1 (RNA ER) and BRAF WT had higher<br />

response rate (68% vs 43%; p�0.035). Response rate were not different<br />

according BRAF mutation (64% WT vs 63% mutant) (p�NS). The<br />

multivariate regression analysis identified ECOG PS (HR: 4.2 (CI 1.4-12.7)<br />

as a significant variable for PFS and LDH levels (HR: 2.26 (CI 1.3-3.8), PS<br />

(HR: 3.85 (CI 1.5-9.8) and high RAC1b/RAC1 (RNA ER) (HR: 2.6 (CI<br />

1.1-5.9) for OS in WT BRAF pts. Conclusions: High RAC1b/RAC1 (RNA ER)<br />

constitutes a marker <strong>of</strong> poor OS and a potential marker <strong>of</strong> acquired<br />

chemo-resistance in WT BRAF mCRC pts treated with first-line oxaliplatinbased<br />

therapy.<br />

3555 General Poster Session (Board #25B), Mon, 8:00 AM-12:00 PM<br />

Phase II study <strong>of</strong> panitumumab (P) in combination with FOLFOXIRI as<br />

first-line treatment <strong>of</strong> metastatic colorectal cancer (mCRC): Activity in<br />

molecularly selected patients (pts). Presenting Author: Sara Lonardi,<br />

Oncologia Medica 1, Istituto Oncologico Veneto - IRCCS, Padova, Italy<br />

Background: GONO-FOLFOXIRI demonstrated higher activity and efficacy<br />

compared to FOLFIRI. P with oxaliplatin- or irinotecan-based doublets is<br />

feasible and associated with improved activity in KRAS codon 12-13<br />

wild-type pts. BRAF and other RAS rare mutations have been suggested as<br />

additional potential biomarkers for anti-EGFR agents. Methods: Pts with<br />

untreated unresectable mCRC and wild-type BRAF-RAS genes were enrolled<br />

in this GONO multicenter phase II trial <strong>of</strong> biweekly P 6 mg/kg d1 with<br />

a modified FOLFOXIRI regimen (IRI 150 mg/m2 d1, OXA 85 mg/m2 d1,<br />

l-LV 200 mg/m2 d1 and 5FU 3000 mg/m2 48-h continuous infusion d1,<br />

reduced to 2400 mg/m2 due to grade 3-4 toxicity in 2 <strong>of</strong> first 3 pts<br />

enrolled). Primary end-point was response rate (RR, RECIST Criteria).<br />

Based on a two stage Simon’s Minimax design (p0�60%, p1�80%;<br />

a�0.05, � �0.2) at least 26 responses on 36 evaluable pts should be<br />

observed to satisfy the primary end-point. Results: 37 out <strong>of</strong> 87 screened<br />

pts were enrolled (M/F, 57/43%; median age 63 years, range 33-72; ECOG<br />

PS 0/1-2, 76/24%; primary colon/rectum, 70/30%; primary on site, 42%;<br />

sites <strong>of</strong> disease single/multiple, 54/46%; liver only mts, 35%). Among the<br />

first 3 pts treated with 5FU 3000 mg/m2 , 2 experienced SAEs (1 grade 4<br />

diarrhea and neutropenia; 1 grade 3 diarrhea). Grade 3-4 toxicities<br />

observed among the 34 pts treated at the amended dose were: neutropenia<br />

53% (2 febrile neutropenia); diarrhea 32%; stomatitis 15%; neurotoxicity<br />

(grade 2-3) 30%; cutaneous rash 24%. Delays or dose reductions were<br />

needed only in 9% and 10% <strong>of</strong> the total 310 cycles, respectively. One SAE<br />

(febrile neutropenia and sepsis) resulting in pt death occurred after<br />

amendment. 32 partial responses, 4 disease stabilizations and 1 progression<br />

were observed, with a RR <strong>of</strong> 86% (95% CI: 75-97%). 9 pts underwent<br />

local procedures on metastases, achieving an R0 resection in 8 and a<br />

pathologic complete response in 3 <strong>of</strong> them. At a median follow-up <strong>of</strong> 7.4<br />

months mPFS has not been reached. Conclusions: In molecularly selected<br />

unresectable pts adding P to FOLFOXIRI resulted in high activity and<br />

interesting resection rate. The regimen appears feasible. Further follow-up<br />

is needed to assess long-term outcome.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


3556 General Poster Session (Board #25C), Mon, 8:00 AM-12:00 PM<br />

Exploratory analysis <strong>of</strong> adjuvant chemotherapy benefits after preoperative<br />

chemoradiotherapy and radical resection for rectal cancer. Presenting<br />

Author: George J. Chang, University <strong>of</strong> Texas M. D. Anderson Cancer<br />

Center, Houston, TX<br />

Background: Evidence regarding adjuvant chemotherapy (CTx) for patients<br />

with rectal cancer after preoperative chemoradiotherapy (CXRT) is limited.<br />

The aim <strong>of</strong> study was to explore the relationship between CXRT response<br />

and adjuvant CTx for patients with rectal cancer treated by CXRT and<br />

radical resection. Methods: We performed a retrospective consecutive<br />

cohort study <strong>of</strong> patients with locally advanced (cStage II-III by EUS, CT, or<br />

MRI) rectal carcinoma treated with preoperative CXRT and radical resection,<br />

1993 to 2008. To explore the late effects, we performed a landmark<br />

analysis <strong>of</strong> patients alive without recurrence at 24 months. The duration<br />

free from recurrence (FFR) was compared among patients with complete<br />

(CR, ypT0N0), intermediate (IR, ypT1-2N0), or poor (PR, ypT3-4 or N�)<br />

response according to adjuvant CTx use and type using multivariate Cox<br />

regression. Results: A total <strong>of</strong> 725 patients met criteria and were evaluated.<br />

Median follow-up was 69 months (IQR: 39-104 months). 611 patients<br />

received adjuvant CTx: 447 with fluoropyrimidine only (FP), and 139 also<br />

received oxaliplatin (Ox). Although receipt <strong>of</strong> adjuvant chemotherapy was<br />

not associated with 5-year FFR overall, (HR: 0.91, 95% CI: 0.58-1.43),<br />

adjuvant therapy did appear to suggest a benefit in the IR group (HR, 0.49;<br />

95%CI, 0.21-1.19). 2-year landmark analysis showed that adjuvant CTx<br />

was associated with a significant improvement in FFR among the IR<br />

patients only (HR, 0.28; 95% CI, 0.08-1.00, p�0.04). Among PR patients<br />

FP alone did not improve FFR (HR, 1.22; 95% CI, 0.7-2.12) but the<br />

addition <strong>of</strong> Ox was associated with a non-significant reversal in the<br />

direction <strong>of</strong> the effect (HR, 0.53; 95% CI, 0.16-1.79). Conclusions: In this<br />

exploratory analysis, the addition <strong>of</strong> adjuvant FP CTx appeared to favorably<br />

affect the duration FFR only for patients with rectal cancer and intermediate<br />

response to CXRT followed by radical resection. These data support the<br />

investigation <strong>of</strong> the role for adjuvant fluoropyrimidine therapy among<br />

patients with CR or IR and the addition <strong>of</strong> oxaliplatin for PR patients.<br />

3558 General Poster Session (Board #26B), Mon, 8:00 AM-12:00 PM<br />

Phase II study <strong>of</strong> combination therapy with S-1 and cetuximab in patients<br />

with KRAS wild-type unresectable colorectal cancer who had previously<br />

received irinotecan, oxaliplatin, and fluoropyrimidines (KSCC0901). Presenting<br />

Author: Kazuma Kobayashi, Department <strong>of</strong> Medical Oncology,<br />

Kochi Health Sciences Center, Kochi, Japan<br />

Background: Anti-epidermal growth factor receptor (anti-EGFR) antibodies<br />

alone or in combination with irinotecan (Iri) can be considered standard<br />

third-line therapy for KRAS wild-type (wKRAS) unresectable colorectal<br />

cancer (UNCRC). However, some UNCRC patients (pts) cannot tolerate<br />

Iri-containing therapy. S-1, an oral fluorouracil (FU) derivative, enhances<br />

the anti-tumor effect by inhibiting dihydropyrimidine dehydrogenase activity<br />

and reducing digestive toxicity. Combination therapy with cetuximab<br />

(C-mab) may restore 5-FU resistance in 5-FU–resistant CCs. Therefore, we<br />

examined the efficacy <strong>of</strong> S-1�C-mab therapy in wKRAS UNCRC pts, who<br />

had previously received Iri, oxaliplatin (OX), and FUs. Methods: The study<br />

design was multicenter, single-arm, open-label phase II study. The major<br />

inclusion criteria were written informed consent; histologically proven CRC<br />

and clinically proven UNCRC; presence <strong>of</strong> measurable lesions; previous<br />

therapy with Iri, OX, and 5-FU; documented progressive disease after<br />

5-FU–based chemotherapy; wKRAS tumors; age � 20 years; performance<br />

status (PS) 0–1; and adequate organ function. The treatment protocol was<br />

as follows: weekly durable intravenous (DIV) C-mab administration at 400<br />

mg/m2 (day 1) and 250 mg/m2 /week (except day 1) and oral administration<br />

<strong>of</strong> 80 mg/m2 /day S-1 on days 1–28 <strong>of</strong> each 42-day cycle. The primary<br />

endpoint was progression-free survival (PFS). A sample size <strong>of</strong> 39 was<br />

planned for a threshold PFS <strong>of</strong> 3.5 months and expected value <strong>of</strong> 6.0<br />

months, with one-sided alpha <strong>of</strong> 0.05 and beta <strong>of</strong> approximately 0.2.<br />

Results: One patient was ineligible; 38 pts (PS 0/1, 32/6; 1/2/�3 prior<br />

chemotherapy regimens, 4/23/11) were enrolled from 10/2009 to 12/<br />

2010. The median PFS (central review) was 5.5 months (90% CI: 4.4 –<br />

5.7); median overall survival (OS), 13.1 months; and the best ORR,<br />

36.8%. The most common grade 3–4 adverse events were neutrophils,<br />

hypokalemia, rash, and dry skin. Conclusions: This study showed that<br />

S-1�C-mab may be a promising and well-tolerated treatment choice <strong>of</strong><br />

wKRAS UNCRC, who had previously heavily treated by Iri, OX and FUs.<br />

Gastrointestinal (Colorectal) Cancer<br />

3557 General Poster Session (Board #26A), Mon, 8:00 AM-12:00 PM<br />

Post-progression survival (PPS) according to treatment line, type, and time<br />

in phase III trials in advanced colorectal cancer (ACC). Presenting Author:<br />

Marc E. Buyse, International Drug Development Institute, Louvain-la-<br />

Neuve, Belgium<br />

Background: Since post-progression therapy influences overall survival<br />

(OS), PPS is <strong>of</strong> interest as a determinant <strong>of</strong> OS. We quantified the<br />

relationship between several median times to event in ACC: progressionfree<br />

survival (PFS), PPS and OS. We looked for factors with an impact on<br />

these times. Methods: We searched PubMed for phase III trials published<br />

between January 1998 and December 2010 in 12 leading journals. PPS is<br />

the difference between median OS and median PFS or time to tumor<br />

progression. The statistics <strong>of</strong> interest (median times [PFS, OS and PPS]<br />

and the ratio <strong>of</strong> PPS/OS) were compared using t-tests. Results: We retrieved<br />

70 trials that yielded 71 comparisons and 156 arms (one trial was factorial)<br />

involving 38,504 patients. Trials from the period 2005-10 enrolled more<br />

patients than trials from the period 1998-2004 (medians <strong>of</strong> 246 vs 174<br />

per arm; P�.026). PPS could be calculated for 60 trials and 135 arms<br />

(Table). Treatment type did not have an impact on any statistic. Treatment<br />

line had a significant impact on all times and on the ratio <strong>of</strong> PPS/OS,<br />

although the difference on the latter statistic was small. Treatment period<br />

had a significant impact on all times but not on the ratio <strong>of</strong> PPS/OS.<br />

Conclusions: None <strong>of</strong> the factors examined have a major impact on the ratio<br />

<strong>of</strong> PPS/OS. This suggests that in ACC, gains in PFS are translated into<br />

proportional gains in OS, one <strong>of</strong> the conditions required for PFS to be<br />

considered a surrogate for OS.<br />

PFS (months) PPS (months) OS (months) PPS / OS (%)<br />

Trial type N Average* p Average* p Average p Average* P<br />

Overall 135 6.3 - 9.0 - 15.3 - 59 -<br />

Chemotherapy only<br />

97 6.1 .30 9.0 .64 15.1 .43 59 .82<br />

At least one targeted agent 38 6.6<br />

9.3<br />

15.9<br />

59<br />

1st line only<br />

108 6.8 �.001 9.6 �.001 16.4 �.001 58 .004<br />

2nd / 3rd line<br />

27 4.0<br />

7.0<br />

11.0<br />

63<br />

Published 1998-2004<br />

59 5.2 �.001 7.8 �.001 13.0 �.001 60 .51<br />

Published 2005-2010 76 7.1<br />

10.0<br />

17.1<br />

58<br />

1st line, 1998-2004<br />

51 5.5 �.001 8.2 �.001 13.7 �.001 59 .16<br />

1st line, 2005-2010 57 8.0<br />

10.8<br />

18.8<br />

57<br />

2nd/3rd line, 1998-2004 8 3.1 .033 5.2 .016 8.3 .007 62 .74<br />

2nd / 3rd line, 2005-2010 19 4.4<br />

7.7<br />

12.1<br />

63<br />

N, Number <strong>of</strong> treatment arms; * Average <strong>of</strong> median values for treatment arms.<br />

217s<br />

3560 General Poster Session (Board #27A), Mon, 8:00 AM-12:00 PM<br />

Impact <strong>of</strong> body mass index (BMI) and weight changes on recurrence and survival<br />

in stage III colon cancer (CC). Presenting Author: Joanna Vergidis, British<br />

Columbia Cancer Agency, Vancouver, BC, Canada<br />

Background: Research suggests that physical activity can lower the risk <strong>of</strong> relapse<br />

and mortality from CC. It is unclear if such potential benefits are mediated<br />

through an effect on weight. Our aims were to 1) evaluate the impact <strong>of</strong><br />

pre-treatment BMI on recurrence and survival in early stage CC, 2) examine how<br />

weight gains and losses from baseline may affect outcomes, and 3) explore if the<br />

effects <strong>of</strong> different body compositions are modified by adjuvant therapy (AT).<br />

Methods: Patients (pts) diagnosed with stage III CC from 2006 to 2008,<br />

evaluated at any 1 <strong>of</strong> 5 cancer centers in British Columbia, and who had their<br />

weights measured serially throughout their AT period were reviewed. Using Cox<br />

proportional hazards models, we compared outcomes among a) BMI�25<br />

[normal] vs. 25-30 [overweight] vs. �30 [obese] and b) weight increase or<br />

decrease <strong>of</strong> � vs. �/�5% and � vs. �/�10%, while controlling for confounders.<br />

Results: A total <strong>of</strong> 821 pts were included: median age was 65 years, 50% were<br />

men, and 83% were ECOG 0/1. At baseline, 43, 37, and 20% were normal,<br />

overweight, and obese, respectively. Compared to pts with normal BMI, those<br />

who were overweight or obese did not show an increased risk <strong>of</strong> recurrence (HR<br />

0.81, 95%CI 0.42-1.58 and HR 1.09, 95%CI 0.48-2.51, respectively, p<br />

trend�0.74) or death (HR 0.54, 95%CI 0.26-1.13 and HR 0.62, 95%CI<br />

0.22-1.74, respectively, p trend�0.24). A number <strong>of</strong> pts experienced weight<br />

gain or loss during their AT period, but none <strong>of</strong> these weight changes affected<br />

outcomes (Table). Receipt <strong>of</strong> AT did not modify the effects <strong>of</strong> BMI or weight<br />

change. In Cox models, advanced age and poor performance status were the<br />

main factors that consistently correlated with an increased risk <strong>of</strong> recurrence and<br />

death (p�0.05). Conclusions: Neither baseline BMI nor short-term weight<br />

changes during AT appear to affect outcomes in stage III CC. Physical activity<br />

likely exert its benefit on relapse and survival by influencing long-term weight<br />

changes or acting via other alternative mechanisms.<br />

Recurrence Death<br />

% <strong>of</strong> Cohort HR p value HR p value<br />

Weight gain<br />

/�5% 50.4 1.04 0.90<br />

/�10%<br />

Weight loss<br />

18.7 0.75 0.98<br />

/�5% 12.4 0.98 1.42<br />

/�10% 4.5 0.41 0.83<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


218s Gastrointestinal (Colorectal) Cancer<br />

3561 General Poster Session (Board #27B), Mon, 8:00 AM-12:00 PM<br />

Relationship <strong>of</strong> Src activity and prior oxaliplatin on outcomes after<br />

hepatectomy for metastatic colorectal cancer. Presenting Author: Van<br />

Karlyle Morris, University <strong>of</strong> Texas M. D. Anderson Cancer Center, Houston,<br />

TX<br />

Background: The nonreceptor tyrosine kinase Src regulates pathways<br />

critical to tumor proliferation, chemoresistance, and epithelial-tomesenchymal<br />

transition. In vitro, Src is activated after acute oxaliplatin<br />

exposure and in acquired oxaliplatin resistance, but not after 5-FU alone.<br />

Activation <strong>of</strong> Src and its substrate FAK in metastatic colorectal cancer<br />

treated with oxaliplatin has not been studied in human specimens.<br />

Methods: Samples from 170 hepatic resections from two cohorts <strong>of</strong> patients<br />

with metastatic colorectal cancer were examined by IHC for expression <strong>of</strong><br />

activated Src (pSrc) and FAK (total and pFAK). In the first cohort (n�50),<br />

tissue was collected at consecutive hepatic resections before and after<br />

oxaliplatin. Patients in the second cohort (n�120) were compared based<br />

on whether or not oxaliplatin was administered after resection. IHC was<br />

graded semi-quantitatively, 0 to 4 based on intensity (first cohort), and by<br />

automated image analysis (second cohort). Results: In the first cohort,<br />

pFAK expression increased after oxaliplatin exposure (mean IHC score<br />

2.04 vs. 1.18, p�0.01). In the second cohort, Src activation was<br />

correlated with pFAK expression (p�0.01). Patients pretreated with<br />

oxaliplatin demonstrated increased expression <strong>of</strong> activated FAK (p�0.02)<br />

compared to 5-FU alone or irinotecan regimens. There was a weak<br />

association between total Src expression and the number <strong>of</strong> oxaliplatin<br />

cycles (p�0.06). Among patients in the second cohort, five-year relapsefree<br />

survival was inversely related to levels <strong>of</strong> pFAK (21.1%, 16.5%, and<br />

7.4% for low, medium, and high levels <strong>of</strong> pFAK, respectively; p�0.02) and<br />

<strong>of</strong> pSrc (19.6%, 13.6%, and 8.2% for low, medium, and high levels <strong>of</strong><br />

pSrc, respectively; p� 0.01). Conclusions: Patients treated with neoadjuvant<br />

oxaliplatin demonstrated increased Src signaling in liver metastases, a<br />

finding associated with worse relapse-free survival. These results are<br />

consistent with prior in vitro studies correlating oxaliplatin exposure with<br />

Src pathway activation and support the idea that inhibition <strong>of</strong> Src, when<br />

used in combination with platinum chemotherapy, warrants further investigation<br />

in patients with metastatic colorectal cancer.<br />

3563 General Poster Session (Board #28A), Mon, 8:00 AM-12:00 PM<br />

Evaluation <strong>of</strong> FDG-PET in pretreatment staging in locally advanced rectal<br />

cancer: A prospective study. Presenting Author: Jose G. Guillem, Memorial<br />

Sloan-Kettering Cancer Center, New York, NY<br />

Background: The utility <strong>of</strong> adding 18F-Fluorodeoxyglucose-Positron Emission<br />

Tomography (PET) to the pre-treatment staging <strong>of</strong> patients with locally<br />

advanced rectal cancer has not been well evaluated. Methods: Following<br />

completion <strong>of</strong> standard work-up, including physical examination, endorectal<br />

ultrasound, CT <strong>of</strong> the abdomen and pelvis, and chest CT or chest x-ray,<br />

150 patients with primary, resectable, biopsy-proven rectal adenocarcinoma<br />

requiring neoadjuvant chemoradiation were enrolled in a prospective,<br />

single-stage, phase II study evaluating the role <strong>of</strong> PET. All patients<br />

underwent a dedicated whole-body PET prior to initiation <strong>of</strong> neoadjuvant<br />

chemoradiation. The primary endpoint <strong>of</strong> this analysis was to determine the<br />

accuracy <strong>of</strong> PET in detecting extrapelvic metastatic disease in primary<br />

rectal cancer patients considered operable on the basis <strong>of</strong> currentlystandard<br />

work-up. Results: Among the 117 eligible patients found to be<br />

stage II or III by standard staging techniques, none (0%) were found by PET<br />

to have extrapelvic metastatic disease. PET yielded a false positive result in<br />

3 patients (2.6%). PET yielded a false-negative result in 3 patients who<br />

were subsequently diagnosed with metastatic disease: 2 pre-operatively<br />

(liver, lung) and 1 intra-operatively (obturator lymph node). In 1 patient,<br />

PET did identify a previously unappreciated obturator lymph node metastasis;<br />

left pelvic sidewall dissection at the time <strong>of</strong> rectal resection confirmed<br />

metastatic adenocarcinoma in one obturator lymph node. Conclusions: Of<br />

the 117 rectal cancer patients evaluated and found to have locoregional<br />

disease by standard examination and imaging techniques, none had<br />

extrapelvic metastatic disease accurately identified by PET. PET did not<br />

improve the accuracy <strong>of</strong> pre-treatment staging. As such, consistent with<br />

current NCCN guidelines, PET does not have a role in pre-treatment staging<br />

<strong>of</strong> locally advanced rectal cancer.<br />

3562 General Poster Session (Board #27C), Mon, 8:00 AM-12:00 PM<br />

Chemotherapy plus cetuximab in patients with liver-limited or non-liver-limited<br />

KRAS wild-type colorectal metastases: A pooled analysis <strong>of</strong> the CRYSTAL and<br />

OPUS studies. Presenting Author: Claus-Henning Kohne, Onkologie Klinikum<br />

Oldenburg, Oldenburg, Germany<br />

Background: In the CRYSTAL and OPUS studies, adding cetuximab (cet) to<br />

first-line chemotherapy (CT) improved clinical benefit in patients (pts) with<br />

KRAS wild-type (wt) metastatic colorectal cancer. In a pooled analysis <strong>of</strong> these<br />

trials the benefit <strong>of</strong> treatment according to whether pts had liver-limited disease<br />

(LLD) or non-LLD was analyzed. Methods: Cox‘s proportional hazards model for<br />

overall survival (OS) and progression-free survival (PFS) or logistic regression<br />

model for best overall response and R0 resection were used on individual pt data,<br />

stratified by study. Likelihood ratio tests were used to explore interactions.<br />

Results: Adding cet to CT significantly improved PFS and overall response rate<br />

(ORR) in LLD pts, and OS, PFS and ORR in non-LLD pts and increased R0<br />

resection rates (table). No treatment-by-study interactions were found. Treatment<br />

effects did not vary significantly by LLD status (PFS p�0.60, OS p�0.68,<br />

ORR p�0.0737, R0 resection p�0.71). However LLD vs non-LLD pts had<br />

improved outcome in each treatment arm: PFS, CT hazard ratio, HR�0.74,<br />

p�0.0910; CT � cet HR�0.66, p�0.0309; OS, CT HR�0.70, p�0.0091; CT<br />

� cet HR�0.74, p�0.0388; ORR, CT odds ratio�1.20, p�0.47, CT � cet odds<br />

ratio�2.32, p�0.0015; R0 resection, CT odds ratio�3.15, p�0.0496, CT �<br />

cet odds ratio�3.82, p�0.0018. Kaplan Meier survival plots will be shown.<br />

Conclusions: The OS benefit from adding cet to CT is more pronounced in<br />

non-LLD pts, thus strengthening the value <strong>of</strong> cet in palliative treatment.LLD<br />

status is associated with improved prognosis and may be predictive for response<br />

in pts receiving CT � cet, facilitating potentially curative resection. More pts<br />

(with R0 resection and longer follow-up) may be needed to confirm an OS benefit<br />

from CT � cet in LLD pts.<br />

CT<br />

(n�95)<br />

KRAS wt pts<br />

LLD Non-LLD<br />

CT� cet<br />

(n�93)<br />

CT<br />

(n�352)<br />

CT � cet<br />

(n�305)<br />

OS<br />

Median, mo 27.0 27.0 17.3 22.0<br />

HR 0.81 0.76<br />

p<br />

PFS<br />

0.25 0.0023<br />

Median, mo 9.2 11.9 7.4 9.4<br />

HR 0.53 0.68<br />

p<br />

Response<br />

0.0095 0.0004<br />

ORR% 43.2 72.0 37.2 52.8<br />

Odds ratio 3.51 1.88<br />

p<br />

R0 resection<br />

�0.0001 �0.0001<br />

Rate, % 5.3 11.8 1.7 3.3<br />

Odds ratio 2.38 1.97<br />

p 0.1121 0.1870<br />

Data (except for R0 resection) are adjusted for differences in baseline characteristics<br />

3564 General Poster Session (Board #28B), Mon, 8:00 AM-12:00 PM<br />

Association study <strong>of</strong> the let-7 microRNA-binding site polymorphism in<br />

3’-untranslated region (UTR) <strong>of</strong> the KRAS gene in stage III colon cancers<br />

from adjuvant trial NCCTG N0147. Presenting Author: Dan Sha, Mayo<br />

Clinic, Rochester, MN<br />

Background: Lethal-7 (let-7) microRNA has been shown to inhibit colon<br />

cancer cell proliferation by inducing KRAS downregulation after binding to<br />

specific sites in the 3’- UTR. Recent studies identified a KRAS 3’-UTR<br />

polymorphism in the let-7 complimentary site (LCS6) that has been shown<br />

to weaken let-7 binding and alter KRAS expression. Carriers <strong>of</strong> the LCS6<br />

G-allele showed worse survival in metastatic colorectal cancer from prior<br />

studies <strong>of</strong> limited sample size. In the current study, we evaluated the LCS6<br />

variant in 2,834 Stage III colon cancer patients and its association with<br />

disease-free survival (DFS), KRAS mutation status, and other clinicopathological<br />

features. Methods: In the NCCTG phase III trial (N0147), the LCS6<br />

variant was assayed in germline DNA extracted from whole blood using<br />

RT-PCR. Extracted tumor DNA was analyzed for KRAS exon 2 mutations.<br />

Chi-squared and two-sample t test were used to compare baseline factors<br />

and KRAS mutation status between patients defined by LCS6 variant<br />

status. Log-rank tests and multivariate Cox models assessed the unadjusted<br />

and adjusted associations between LCS6 status and DFS, respectively.<br />

Results: We identified 432 (15.2%) patients heterozygous or<br />

homozygous for the LCS6 G-allele and 2402 (84.8%) patients homozygous<br />

for the LCS6 T-allele. G-allele carriers were significantly more frequent in<br />

Caucasians than other races (Chi-Squared Test, p�0.0001). No associations<br />

were found between the LCS6 genotype and T stage, positive lymph<br />

node number, tumor differentiation, or primary tumor location (all p�0.2).<br />

Moreover, the LCS6 genotype was not associated with DFS (hazard ratio �<br />

0.93, p�0.49) even after combining LCS6 genotype with KRAS mutation<br />

status. Conclusions: We report the largest association study investigating<br />

the LCS6 polymorphism and colon cancer outcome. There is no significant<br />

evidence that the germline LCS6 genotype was associated with DFS in<br />

stage III colon cancer patients. Additional studies are needed to determine<br />

if LCS6 genotype differs between tumor and germline DNA which may<br />

further clarify its prognostic value.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


3565 General Poster Session (Board #28C), Mon, 8:00 AM-12:00 PM<br />

Bevacizumab (BEV) plus capecitabine as maintenance therapy after initial<br />

treatment with BEV plus XELOX in previously untreated patients (pts) with<br />

metastatic colorectal cancer (mCRC): Mature data from STOP and GO, a<br />

phase III, randomized, multicenter study. Presenting Author: Suayib<br />

Yalcin, Hacettepe University Medicine Faculty, Ankara, Turkey<br />

Background: Colorectal cancer is one <strong>of</strong> the most frequent malignancies,<br />

second after breast cancer in women and third after lung cancer and<br />

prostate cancer in men. The aim <strong>of</strong> this study was to evaluate and compare<br />

the progression-free survival (PFS) between two arms: Arm A is a combination<br />

<strong>of</strong> BEV � XELOX; Arm B is a combination <strong>of</strong> BEV � XELOX for 6 cycles<br />

followed by maintenance BEV � capecitabine as first-line therapy in<br />

mCRC. Methods: BEV (7.5 mg/kg) � XELOX (capecitabine 1000 mg/m2 bid<br />

d1–14 � oxaliplatin 130 mg/m2 d1 q3w) were administered until progression<br />

(Arm A) or 6 cycles <strong>of</strong> BEV � XELOX followed by BEV � capecitabine<br />

were administered until progression (Arm B). PFS was the primary<br />

endpoint; secondary endpoints included overall survival (OS), objective<br />

response rate (ORR), and safety. A sample size <strong>of</strong> 118 pts was required to<br />

detect with 80% power an increase <strong>of</strong> 1.5 months in median PFS between<br />

two arms with a standard deviation <strong>of</strong> 3.9 months and significance level <strong>of</strong><br />

0.05 (10% drop-out rate). Results: A total <strong>of</strong> 123 pts were randomized.<br />

Demographic characteristics were balanced between the arms. Median<br />

treatment period was 7.5 (range 0.5–13.9) and 8.1 (range 0.1–20.7)<br />

months in Arms A and B, respectively. There was a statistically significant<br />

difference in median PFS between arms, although there was no significant<br />

difference in ORR and OS (see table). Tolerability was acceptable in both<br />

arms with the following grade 3/4 adverse events (AEs): Arm A 48.4%; Arm<br />

B 34.4% (p�0.116). Grade 3/4 diarrhoea occurred in 9.7% vs. 3.3%,<br />

weakness in 8.1% vs. 8.2%, hand-foot syndrome in 3.2% vs. 1.6%, and<br />

neuropathy in 4.8% vs. 3.3% <strong>of</strong> pts in Arms A and B, respectively.<br />

Conclusions: These findings suggest that maintenance therapy with BEV �<br />

capecitabine following induction with 6 cycles <strong>of</strong> BEV � XELOX may be<br />

superior to continuous BEV � XELOX until progression inpts with previously<br />

untreated mCRC.<br />

Arm A<br />

(n�62)<br />

Arm B<br />

(n�61) p value<br />

Efficacy<br />

Median PFS, months<br />

8.3<br />

11.0<br />

0.002<br />

(95% CI)<br />

(7.1–9.5) (9.1–12.9)<br />

Median OS, months 20.2 23.8 0.1<br />

ORR, % 58.9 66.7 0.861<br />

3567 General Poster Session (Board #29B), Mon, 8:00 AM-12:00 PM<br />

Implications <strong>of</strong> generating genetic test results for colon cancer in the<br />

international, population-based colon cancer family registry. Presenting<br />

Author: Louise A. Keogh, The University <strong>of</strong> Melbourne, Melbourne, Australia<br />

Background: The ability to genotype large numbers <strong>of</strong> people rapidly and<br />

inexpensively for research purposes highlights the need to develop guidelines<br />

for providing medically-relevant research results - including unanticipated<br />

findings - to study participants. The Colon Cancer Family Registry<br />

(C-CFR) is the oldest and largest international colon cancer populationbased<br />

registry; its experience managing genetic research findings can <strong>of</strong>fer<br />

guidance to clinicians and researchers. The C-CFR has enrolled 10,019<br />

cases with colon cancer and 24,708 family members in six registries in the<br />

US, Canada, Australia, and New Zealand. Deleterious (“high risk”) germline<br />

mutations have been identified in DNA mismatch repair (MMR) genes<br />

(MLH1, MSH2, MSH6, PMS2) and the MutYH gene. The aims <strong>of</strong> this<br />

presentation are to: (1) report the uptake <strong>of</strong> genetic test results by C-CFR<br />

participants; (2) systematically compare disclosure protocols and barriers<br />

to uptake by registry; (3) make recommendations to guide clinicians and<br />

researchers. Methods: Uptake <strong>of</strong> genetic test results was calculated from<br />

data collected by the C-CFR; key investigators (KIs) from each registry<br />

completed a survey about disclosure decision-making; KIs also took part in<br />

discussions to generate recommendations. Results: Registry-wide molecular<br />

testing has identified deleterious MMR germline mutations for at least<br />

one member <strong>of</strong> 424 families (4%) and 48 biallelic MutYH gene carriers.<br />

Uptake <strong>of</strong> test results ranged from 56-86% (n� 1542) across registries.<br />

Barriers to disclosure include: (1) lack <strong>of</strong> pre-existing notification protocols;<br />

(2) logistics <strong>of</strong> re-consent; (3) limited involvement <strong>of</strong> genetic<br />

counselors at some registries; (4) in the US, the requirement that genetic<br />

testing be performed in a CLIA approved laboratory; (5) IRBs declining<br />

approval; and (6) budget constraints. Conclusions: Based on our international<br />

registry’s findings we recommend that researchers generating genetic<br />

information establish plans for disclosure at the outset; obtain<br />

subject consent a priori; consider subject knowledge and disclosure<br />

preferences; provide guidance and budget for clinical follow-up; and<br />

involve genetic counselors.<br />

Gastrointestinal (Colorectal) Cancer<br />

219s<br />

3566 General Poster Session (Board #29A), Mon, 8:00 AM-12:00 PM<br />

Prognostic significance <strong>of</strong> lymph node retrieval in localized rectal cancer<br />

(RC): A registry analysis <strong>of</strong> 8,964 patients (pts). Presenting Author:<br />

Amandeep Gill, University <strong>of</strong> California, Davis, Sacramento, CA<br />

Background: In contrast to colon cancer, the prognostic implications <strong>of</strong><br />

reduced lymph node retrieval in RC are unclear. We investigated the<br />

association between the total number <strong>of</strong> lymph nodes examined in early<br />

stage RC and disease specific survival (DSS) in two cohorts: 1) pts who<br />

underwent surgery and adjuvant (ADJ) chemoradiation (chemoXRT), and 2)<br />

those who received neoadjuvant (NEO) chemoXRT. Methods: Using the<br />

California Cancer Registry, we identified 8,946 pts with stage I – III RC<br />

diagnosed from 2000 to 2007. Of these, 4,790 underwent tri-modality<br />

therapy: 2,946 patients (61.5%) had NEO chemoXRT and 1,844 patients<br />

(38.5%) had ADJ chemoXRT. Multivariate Cox proportional hazards models<br />

were constructed for DSS, adjusting for age, sex, race, socioeconomic<br />

status, T-stage and lymph node number. DSS was compared between NEO<br />

and ADJ cohorts within separate subgroups <strong>of</strong> pathologic node positive and<br />

node negative RC. Results: Although there was no difference in overall DSS<br />

between the NEO and ADJ cohorts, the median number <strong>of</strong> lymph nodes<br />

examined was reduced in patients who had undergone NEO chemoXRT (8<br />

vs. 11, p�0.0001). For all pts treated with tri-modality therapy, advancing<br />

age and higher T-stage were associated with significantly reduced DSS.<br />

Positive lymph nodes were associated with worse DSS regardless <strong>of</strong> the<br />

timing <strong>of</strong> therapy, although the effect was stronger for residual lymph nodes<br />

in the NEO cohort (HR 2.8 vs. 1.8 in ADJ cohort, p�0.001). For node<br />

negative pts in the ADJ cohort, increased lymph node retrieval was<br />

associated with improved DSS (HR per node 0.952, p�0.017); however,<br />

no association between lymph nodes examined and DSS was seen in the<br />

NEO cohort (p�0.282). Conclusions: Residual positive lymph nodes in pts<br />

treated with NEO chemoXRT are more strongly associated with poorer DSS<br />

than in pts treated with surgery and ADJ chemoXRT. NEO chemoXRT<br />

dissociates the connection between lymph node retrieval and survival in<br />

RC. This finding highlights a key difference between colon and RC and<br />

underscores the need for a different interpretation <strong>of</strong> the pathologic<br />

findings after NEO therapy.<br />

3568 General Poster Session (Board #29C), Mon, 8:00 AM-12:00 PM<br />

Association <strong>of</strong> microsatellite instability with prognosis in the pathologic N2<br />

colon cancer patients treated with adjuvant FOLFOX chemotherapy. Presenting<br />

Author: Jeong Eun Kim, Asan Medical Center, Seoul, South Korea<br />

Background: The impact <strong>of</strong> microsatellite instability (MSI) on survival has<br />

been rarely explored, with controversy, in stage III colon cancer treated with<br />

adjuvant 5-flurouracil-oxaliplatin combination (FOLFOX) chemotherapy.<br />

We evaluated association between MSI and survival in this population.<br />

Methods: We analyzed 312 patients (pts) with curatively resected stage III<br />

colon cancer treated with adjuvant FOLFOX chemotherapy. We determined<br />

MSI status using polymerase chain reaction amplification as high levels <strong>of</strong><br />

MSI (MSI-H, � 2 unstable markers), low levels <strong>of</strong> MSI (MSI-L, 1 unstable<br />

marker), and microsatellite stable (MSS, no unstable marker). Results: Of<br />

312 pts, 8.3% showed MSI-H and they were younger (median age, 51.5 vs.<br />

58 years, p�0.018). MSI-H tumors were more commonly located ascending<br />

colon (46.2% vs. 23.1%, p�0.002) and had poorly differentiated<br />

features (30.8% vs. 5.2%, p�0.0001). After the median follow-up <strong>of</strong> 30.9<br />

months, 3-year relapse-free (RFS) and overall survival (OS) rates were<br />

76.1% and 91.7%, respectively. In univariate analysis, pathologic N2<br />

(pN2) was associated with reduced RFS (p�0.0001) and OS (p�0.002),<br />

while MSI status did not affect either RFS (p�0.254) or OS (p�0.961). In<br />

patients with pN2 tumors, however, MSI-H was associated with better<br />

survival compared with MSS/MSI-L; the RFS and OS in pts with pN2/MSI-H<br />

were comparable to those in pts with pN1 tumors (table). Conclusions: MSI<br />

status alone did not affect survival in our population. However, pts with<br />

pN2/MSI-H showed favorable survival outcomes comparable to those with<br />

pN1, and MSS/MSI-L/pN2 was associated with worst survival, implicating<br />

that MSI-H modifies the inherent worse prognosis <strong>of</strong> pN2 tumors.<br />

pN1 pN2<br />

MSS/MSI-L MSI-H MSS/MSI-L MSI-H p value<br />

3yr RFS (%) 86.4 88.0 58.9 85.7 �0.0001<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


220s Gastrointestinal (Colorectal) Cancer<br />

3569 General Poster Session (Board #30A), Mon, 8:00 AM-12:00 PM<br />

Short-term clinical outcomes from a randomized controlled trial to evaluate<br />

laparoscopic versus open complete mesocolic excision for stage II,III<br />

colorectal cancer (CRC): Japan <strong>Clinical</strong> Oncology Group study JCOG0404<br />

(NCT00147134). Presenting Author: Yusuke Nishizawa, National Cancer<br />

Center Hospital East, Kashiwa, Japan<br />

Background: The benefits <strong>of</strong> laparoscopic surgery (LAP) in comparison with<br />

open surgery (OP) have been suggested; however, the long-term survival <strong>of</strong><br />

LAP for advanced CRC requiring complete mesocolic excision is still<br />

unclear. We conducted a study to confirm the non-inferiority <strong>of</strong> LAP to OP<br />

in terms <strong>of</strong> overall survival (OS)with less frequent post-operative morbidity.<br />

Short-term outcomes including post-operative complications are presented<br />

here. Methods: Only accredited surgeons from 30 Japanese institutions<br />

participated. Eligibility criteria included histologically proven CRC; tumor<br />

located in the cecum, ascending, sigmoid or rectosigmoid colon; T3 or<br />

deeper lesion without involvement <strong>of</strong> other organs; N0–2 and M0; tumor<br />

size ��8 cm; patient age 20-75 years. Patients were randomized<br />

preoperatively.Patients with pathological stage III received adjuvant chemotherapy<br />

with fluorouracil plus leucovorin. The primary endpoint is OS. and<br />

the planned sample size was 1050. Results: A total <strong>of</strong> 1057 patients were<br />

randomized (OP: 528, LAP: 529) between October 2004 and March 2009.<br />

Conversion to OP was needed for 29 (5.4%) patients in LAP arm (technical<br />

conversion; 2.3%, indicated conversion; 2.8%, complicated conversion;<br />

0.4%). Patients assigned to LAP had less blood loss compared with those<br />

assigned to OP (median 30 ml vs 85 ml, p�0.001), although LAP lasted<br />

52 minutes longer than did OP (p�0.001). Radicality <strong>of</strong> resection as<br />

assessed by number <strong>of</strong> resected lymph nodes did not differ between two<br />

groups. LAP was associated with earlier recovery <strong>of</strong> bowel function<br />

(p�0.001), and with a shorter hospital stay (p�0.001) compared with OP.<br />

Morbidity and mortality untill discharge did not differ between two groups,<br />

except for less wound-related complications in LAP (p�0.007). Conclusions:<br />

Laparoscopic complete mesocolic excision for stage II,III colorectal cancer<br />

can be performed safely and short-term clinical benefits was demonstrated.<br />

If the non-inferiority <strong>of</strong> LAP in OS is demonstrated in the primary analysis<br />

planned in 2014 , LAP will be the new standard procedure for CRC.<br />

3571 General Poster Session (Board #30C), Mon, 8:00 AM-12:00 PM<br />

A randomized phase II study <strong>of</strong> capecitabine-based chemoradiation with or<br />

without bevacizumab in resectable locally advanced rectal cancer. Presenting<br />

Author: Mercedes Martinez Villacampa, Instituto Catalán de Oncología,<br />

Hospital Durans i Reynals, Hospitalet de Llobregat, Spain<br />

Background: The addition <strong>of</strong> bevacizumab (BEV) to capecitabine (CAP)based<br />

chemoradiation (CRT) has shown encouraging efficacy in locally<br />

advanced rectal cancer (LARC), in nonrandomized studies. This randomized<br />

phase II study investigated the effect <strong>of</strong> adding BEV to preoperative<br />

CAP-based CRT in patients (pts), with LARC. Methods: The primary end<br />

point was pathologic complete response (pCR). A two-stage design was<br />

used. Assuming a minimum pCR rate <strong>of</strong> at least 15% in one <strong>of</strong> the arms, a<br />

difference between the two arms <strong>of</strong> 10%, and accepting a probability <strong>of</strong><br />

correct selection <strong>of</strong> 87%, 41 pts per arm were needed. Patients with LARC<br />

(Stages II-III assessed by MRI) and ECOG PS �2 were randomized to<br />

concurrent radiotherapy 45Gy/25f/5 weeks � CAP (825mg/m²/bid) � BEV<br />

every 2 weeks (5 mg/kg for 3 doses) (arm A) or the same schedule without<br />

BEV (arm B). Surgery was scheduled 6-8 weeks after completing CRT.<br />

Results: 90 pts were randomized (arm A/B: 44/46). Patient’s characteristics<br />

were well balanced between both arms: male 61%, median age 62<br />

years, median distance from anal verge 7 cm, T3 79%, N� 87%. 40<br />

(91%)/43 (93%) <strong>of</strong> pts (arm A/B) finalized the planned CRT � surgery<br />

treatment. Overall grade 3-4 toxicity rates were 18 % and 13% (arm A/B,<br />

p�0.50); no grade 3-4 hematological toxicity was reported. Postoperative<br />

complications were 19(43%)/17(37%)(arm A/B). Efficacy data on patients<br />

who actually underwent surgery are reported in the table. Conclusions: The<br />

addition <strong>of</strong> BEV to CAP-based preoperative CRT has shown to be feasible<br />

and safe in the local control <strong>of</strong> LARC. No differences in pCR were observed<br />

and longer follow-up is needed to assess the impact on survival endpoints.<br />

Arm A: RT�CAP�BVZ Arm B: RT�CAP<br />

Surgery performed 43 (98%) 46 (100%) p:0.49<br />

pCR (ypT0N0) 7 (16%) 5 (11%) p: 0.54<br />

R0 resection rates 42 (95.45%) 44 (96%) p:1.0<br />

Downstaging (lower<br />

26 (59%) 18 (39%) p: 0.0429<br />

pT compared with<br />

the pretreatment cT)<br />

Sphincter saving surgery 27 (61%) 31 (67%) p: 0.66<br />

3570 General Poster Session (Board #30B), Mon, 8:00 AM-12:00 PM<br />

Phase II study <strong>of</strong> dacarbazine for metastatic colorectal cancer with<br />

determined MGMT status. Presenting Author: Alessio Amatu, Ospedale<br />

Niguarda Ca’ Granda, Milan, Italy<br />

Background: O6-methylguanine-DNA-methyltransferase (MGMT) is a DNA<br />

repair protein that removes mutagenic and cytotoxic adducts from O6- guanine in DNA. Roughly 40% <strong>of</strong> colorectal cancers (CRCs) display MGMT<br />

deficiency due to promoter hypermethylation leading to silencing <strong>of</strong> the<br />

gene. Alkylating agents exert their antitumor activity by DNA methylation at<br />

the O6 –guanine site, inducing base pair mismatch; therefore, activity <strong>of</strong><br />

these compounds may be enhanced in CRCs lacking MGMT (Esteller,<br />

2004; Pegg, 1990). Despite anecdotal reports about activity <strong>of</strong> dacarbazine<br />

in CRC (Shacham-Shmueli, 2011) no clinical trials assessed monotherapy<br />

with dacarbazine for metastatic CRC. Methods: We conducted a<br />

phase II study <strong>of</strong> dacarbazine in CRCs who had failed standard therapies<br />

(oxaliplatin, irinotecan, fluoropyrimidines and cetuximab or panitumumab<br />

if KRAS WT), PS�0-1, with adequate organ function, and measurable<br />

disease as per RECIST criteria assessed by CT scans at baseline and every 8<br />

weeks. All patients had tumor tissue assessed for MGMT as promoter<br />

hypermethylation. Patients received dacarbazine 250 mg/m2 i.v. d1-4,<br />

q21 until PD or untolerable toxicity (with a maximum <strong>of</strong> 6 cycles in case <strong>of</strong><br />

SD). We employed a Simon, two-stage design to determinate if the ORR <strong>of</strong><br />

dacarbazine treatment would be � 10%. Secondary endpoints were<br />

association <strong>of</strong> response/resistance with loss <strong>of</strong> expression <strong>of</strong> MGMT, PFS<br />

and disease control rate. Results: In the first stage, from June 2011 to<br />

November 2011 we enrolled 40 pts (male 68%, median age 67 y [range<br />

29-81], PS�0 50%, KRAS mut 55%). Pts received a median <strong>of</strong> 3 cycles<br />

[range 1-11]. Toxicities included (All Grades/Grade 3-4 %): fatigue (58/5),<br />

constipation (38/0), nausea/vomiting (35/0), anemia (20/2.5), platelet<br />

count decrease (10/5). 36 pts were evaluable for objective response.<br />

Overall, 2 pts (5%) achieved PR and 5 pts (12.5%) had SD. All pts with PR<br />

had tumors with MGMT promoter hypermethylation. Conclusions: Having<br />

exceeded the boundaries <strong>of</strong> futility (at least 2 objective responses in the<br />

first stage), the trial proceeds to the second stage, enrolling 28 more<br />

patients. Analysis <strong>of</strong> MGMT promoter hypermethylation and association<br />

with sensitivity/resistance is performed at the end <strong>of</strong> the second stage.<br />

3572 General Poster Session (Board #31A), Mon, 8:00 AM-12:00 PM<br />

Analysis <strong>of</strong> plasma biomarkers potentially associated with antiangiogenic<br />

resistance in NCIC CTG/AGITG CO.20: A phase III randomized trial <strong>of</strong><br />

cetuximab (CET) plus either brivanib alaninate (BRIV) or placebo in<br />

patients (pts) with chemotherapy refractory, k-RAS wild-type (WT), metastatic<br />

colorectal carcinoma (mCRC). Presenting Author: Jeremy David<br />

Shapiro, Cabrini Hospital and Monash University, Melbourne, Australia<br />

Background: In the CO.20 trial, the addition <strong>of</strong> BRIV, a tyrosine kinase<br />

inhibitor targeting vascular endothelial and fibroblast growth factor receptors<br />

(VEGFR2,3 and FGFR 1,2,3), to CET, increased objective response<br />

rate and progression free survival, but did not significantly increase overall<br />

survival. Previous clinical studies demonstrated modulation <strong>of</strong> circulating<br />

angiogenic factors (CAF) in CRC which occur on therapy or upon progression<br />

(Kopetz JCO 2010). Methods: CO.20 pts were randomized 1:1 to CET<br />

plus either BRIV (Arm A) or placebo (ARM B) in a double-blind design. Pts<br />

may have had 1 prior anti-VEGF based therapy, but no prior anti-EGFR<br />

therapy. Primary endpoint was overall survival (OS). Baseline pre-treatment<br />

plasma samples were analyzed using commercial Multi-Analyte ELISAs to<br />

measure CAF, immunologic, and growth factors, with an initial discovery<br />

and subsequent validation data set. Results: 750 pts were randomized (376<br />

in Arm A and 374 in Arm B). Median OS in the intent-to-treat population<br />

was 8.8 months in Arm A and 8.1 months in Arm B, hazard ratio<br />

(HR)�0.88; 95%CI�0.74-1.03; p�0.12. In an exploratory subgroup<br />

analysis, there is a statistically non-significant trend favoring the effect <strong>of</strong><br />

brivanib on OS among the 41% pts who received prior anti-VEGF therapy<br />

versus those who did not.Baseline plasma samples were collected from<br />

96% <strong>of</strong> pts, and analysis is ongoing. Results <strong>of</strong> the largest circulating<br />

biomarker analysis will be presented. CAF results will be compared with<br />

response and survival data to look for potential patient subgroups that may<br />

benefit more from the combination treatment. Conclusions: This large scale<br />

analysis will provide insights on the potential use <strong>of</strong> CAF or CAF pr<strong>of</strong>iles as<br />

predictive markers in CRC.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


3573 General Poster Session (Board #31B), Mon, 8:00 AM-12:00 PM<br />

The relationship between microRNA-126 and maintenance therapy with<br />

bevacizumab after bevacizumab plus chemotherapy as first-line treatment<br />

for patients with metastatic colorectal cancer: Results <strong>of</strong> the phase III<br />

Nordic ACT trial NCT00598156 translational study. Presenting Author:<br />

Torben Hansen, Danish Colorectal Cancer Group South, Vejle Hospital and<br />

University <strong>of</strong> Southern Denmark, Vejle, Denmark<br />

Background: Maintenance therapy during chemotherapy free intervals is a<br />

debatable topic in metastatic colorectal cancer (mCRC). In the Nordic ACT<br />

trial patients benefitting from induction chemotherapy plus bevacizumab<br />

were randomized to bevacizumab with or without erlotinib. MicroRNA-126<br />

(miRNA-126) is believed to influence angiogenesis in several ways, and we<br />

investigated the relationship between miRNA-126 and progression free<br />

survival (PFS) under maintenance therapy. Methods: The miRNA-126 was<br />

analysed by polymerase chain reaction on both genomic DNA (by the<br />

pri-miRNA-126 (A24G) single nucleotide polymorphism (SNP)) and paraffin<br />

embedded primary tumors, (which were classified as low or high<br />

miRNA-126 expressing tumours using the median value as cut-<strong>of</strong>f).<br />

Diagnostic biopsies were not processed due to limited tumour tissue. Of the<br />

159 randomized patients blood samples were available from 151 and<br />

primary tumours from 91. The PFS (from time <strong>of</strong> randomization) was<br />

compared using the Kaplan-Meier method and the log rank test. The Cox<br />

Regression analyses were used to test for independent contributions.<br />

Results: The A24G SNP was not related to PFS, but a significant association<br />

was seen between genotypes and miRNA-126 expression levels.Furthermore,<br />

the PFS was increased in patients with high tumor miRNA-126<br />

expression compared to patients with low expression, hazard ratio (HR)<br />

0.58 (95% confidence interval (CI) 0.38-0.90), p � 0.015. The median<br />

PFS was 6.2 months (95% CI 4.3-6.6 months) and 4.0 months (95% CI<br />

2.6-4.5 months), respectively. Results did not differ between treatment<br />

arms, but the Cox Regression analysis confirmed an independent prognostic<br />

value <strong>of</strong> the tumor miRNA-126 expression, HR 0.61 (95% CI 0.39-<br />

0.95), p � 0.030. Conclusions: The miRNA-126 may be a potential marker<br />

for bevacizumab containing maintenance therapy. A similar relationship<br />

with PFS has recently been demonstrated in patients with mCRC treated<br />

with chemotherapy only. The results call for validation.<br />

3575 General Poster Session (Board #31D), Mon, 8:00 AM-12:00 PM<br />

Molecular and clinicopathologic evidence <strong>of</strong> heterogeneity in KRASmutant<br />

colon cancers. Presenting Author: Vlad Calin Popovici, Swiss<br />

Institute <strong>of</strong> Bioinformatics, Lausanne, Switzerland<br />

Background: The activation <strong>of</strong> the MEK/ERK signaling cascade by KRAS or<br />

BRAF mutations has a high incidence in colorectal cancer (CRC). While<br />

these mutations are mutually exclusive, we have identified a highly<br />

conserved gene expression pattern, characteristic to BRAF mutants, that<br />

can also be observed in KRAS mutants (AACR 2011, JCO 2012), and<br />

which defines a distinct subpopulation. Its characterization is important as<br />

it may shed a light on the observed survival and treatment response<br />

variability <strong>of</strong> this group <strong>of</strong> CRCs. Methods: A set <strong>of</strong> 257 stage II and III<br />

KRAS mutant CRCs from the PETACC3 clinical trial and a subset <strong>of</strong> 150<br />

CRCs from TCGA project were used for this study. Gene expression data was<br />

available for all samples and, additionally, CNV and methylation data was<br />

available for some <strong>of</strong> the samples. Due to the limited number <strong>of</strong> MSI<br />

samples, the analyses were focused on MSS subpopulation. Results: The<br />

cluster analysis on the genes from the BRAF signature clearly indicated a<br />

major split <strong>of</strong> the KRAS mutants into two subgroups, that were further<br />

compared in terms <strong>of</strong> clinico-pathological and survival parameters. The<br />

BRAF-mutant-like (BML) subgroup was clearly enriched in MSI-H<br />

(p�0.001), right-sided (p�0.038) and mucinous histology (p�0.001)<br />

tumors. The enrichment in mucinous tumors remained significant also in<br />

stratified analysis by MSI status. In MSS, the BML subpopulation <strong>of</strong> KRAS<br />

mutants had the worst prognosis for overall survival (p�0.05, HR�1.6)<br />

and survival after relapse (p�0.004, HR�2.1). Analysis <strong>of</strong> differentially<br />

expressed genes between BML and the rest <strong>of</strong> KRAS mutants identified<br />

genes responsible for colon crypt differentiation, Wnt pathway activation<br />

and, more intriguingly, 50 other genes, all located on chromosome 20q,<br />

that had significantly different activation levels in BML tumors. Among<br />

these genes, there are a number <strong>of</strong> important tumor suppressors, like<br />

PLAGL2, TP53RK and POFUT1. No differences in type <strong>of</strong> KRAS mutation<br />

nor in PIK3CA mutations were found between BML and the rest <strong>of</strong> KRAS<br />

mutants. Conclusions: We identified a subgroup <strong>of</strong> KRAS mutants formed <strong>of</strong><br />

more aggressive, poorly differentiated tumors. All these results prove that<br />

KRAS mutant CRCs form a heterogeneous group tumors.<br />

Gastrointestinal (Colorectal) Cancer<br />

221s<br />

3574 General Poster Session (Board #31C), Mon, 8:00 AM-12:00 PM<br />

Cetuximab combined with infusional 5-fluorouracil/folinic acid (5-FU/FA)<br />

and oxaliplatin in metastatic colorectal cancer (mCRC): A pooled analysis<br />

<strong>of</strong> COIN and OPUS study data. Presenting Author: Julien Taïeb, Georges<br />

Pompidou European Hospital, Paris, France<br />

Background: Infusional 5-FU/FA � oxaliplatin is a widely used schedule in<br />

the first-line treatment <strong>of</strong> mCRC. In the randomized phase II OPUS study,<br />

the addition <strong>of</strong> cetuximab to one such regimen (FOLFOX4) significantly<br />

improved response and progression-free survival (PFS) in patients (pts)<br />

with KRAS wild-type (wt) mCRC. However, in the randomized phase III<br />

COIN study, a benefit for the addition <strong>of</strong> cetuximab to first-line fluoropyrimidine<br />

(administered as either infusional 5-FU or capecitabine) � oxaliplatin<br />

was not confirmed in pts with KRAS wt tumors. Methods: A pooled<br />

study-based analysis <strong>of</strong> treatment outcome in pts with KRAS wt tumors<br />

from the OPUS study and COIN subgroup who received infusional 5-FU/FA<br />

� oxaliplatin (as the OxMdG regimen) was carried out using a random<br />

effects model. Outcome in the pooled analysis was considered in the<br />

context <strong>of</strong> other randomized studies investigating first-line chemotherapy<br />

regimens �/- cetuximab in pts with mCRC. Results: The pooled KRAS wt<br />

population included 179 pts from the OPUS study and 244 from the<br />

OxMdG subgroup <strong>of</strong> the COIN study. A benefit for the addition <strong>of</strong> cetuximab<br />

to infusional 5-FU/FA was suggested for response (odds ratio 1.87, 95% CI<br />

1.07–3.28) and PFS (hazard ratio, HR 0.69, 95% CI 0.52–0.92) but<br />

overall survival (OS) did not show a statistically significant improvement<br />

(HR 0.90, 95% CI 0.73–1.11). These response and PFS data are similar to<br />

those <strong>of</strong> the KRAS wt population <strong>of</strong> the CRYSTAL study investigating<br />

infusional 5-FU/FA and irinotecan �/- cetuximab (response: odds ratio<br />

2.07, 95% CI 1.52–2.83; PFS: HR 0.70, 95% CI 0.56–0.87) whereas the<br />

improvement in OS was statistically significant in that study (HR 0.80,<br />

95% CI 0.67–0.95). Similar efficacy <strong>of</strong> FOLFOX � cetuximab and<br />

FOLFIRI � cetuximab in the first-line treatment <strong>of</strong> mCRC was also<br />

suggested by data from the randomized phase II CORE 1.2.001 and CELIM<br />

studies. Overall, the safety pr<strong>of</strong>ile <strong>of</strong> infusional 5-FU/FA and oxaliplatin �<br />

cetuximab was found to be acceptable and manageable. Conclusions: The<br />

pooled analysis supports the use <strong>of</strong> cetuximab combined with infusional<br />

5-FU/FA and oxaliplatin in the first-line treatment <strong>of</strong> KRAS wt mCRC.<br />

3576 General Poster Session (Board #31E), Mon, 8:00 AM-12:00 PM<br />

Prognostic impact <strong>of</strong> preoperative plasma tissue inhibitor <strong>of</strong> metalloproteinase<br />

1 (pTIMP-1) in colorectal cancer (CRC) patients (Pts): A pooled<br />

analysis <strong>of</strong> 1,024 stage II/III pts. Presenting Author: Nils Brünner, Section<br />

for Pathobiology, Department <strong>of</strong> Veterinary Disease Biology, Faculty <strong>of</strong> Life<br />

Sciences, University <strong>of</strong> Copenhagen, Frederiksberg C, Denmark<br />

Background: Identification <strong>of</strong> a marker with strong prognostic impact on<br />

clinical outcomes would aid in the management <strong>of</strong> pts with primary,<br />

curatively resected CRC. We conducted a pooled individual pt data analysis<br />

to confirm the prognostic value <strong>of</strong> pTIMP-1 for overall survival (OS) in stage<br />

II and III CRC. Methods: pTIMP-1 was determined in plasma using<br />

immunoassays. Samples and clinical data were included from 5 studies<br />

with a total <strong>of</strong> 1,042 stage II/III pts resected between 1991 – 2006<br />

(Holten-Andersen (HA), 2000 [n�378], HA, 2004 [n�226], Waas, 2005<br />

[52], Birgisson, 2010 [225], Nielsen, 2011[n�161]). To provide comparable<br />

measures across studies, pTIMP-1 values were rescaled using<br />

fractional ranks (FR, based on ranks and sample size). Pts were grouped by<br />

the median <strong>of</strong> the pTIMP-1 FR score. Stratified log-rank tests and<br />

multivariate Cox models assessed the association between OS and pT-<br />

IMP-1 FR. Results: Overall, 59% were male; median age 69 years; 56% had<br />

stage II disease and 50% had colon cancer. The median follow-up was 5.5<br />

years with 457 deaths. Continuous pTIMP-1 FR as a linear effect was<br />

significantly associated with OS (p � .0001, HR�1.38 for a 0.3 unit<br />

increase in FR score, 95% CI 1.25 to 1.52).Pts with pTIMP-1 FR above vs<br />

below the median had significantly shorter OS (HR, 1.72; 95% confidence<br />

interval[CI], 1.42 to 2.08, p �0.0001), which remained significant after<br />

adjusting for age, gender, stage, disease location, and serological CEA (HR,<br />

1.31; 95%CI, 1.07 to 1.60; p � 0.009). Results by stage are shown in the<br />

table. No evidence <strong>of</strong> an interaction between pTIMP-1 and stage or disease<br />

location (colon vs. rectal) was observed (all p � 0.6). Conclusions: pTIMP-1<br />

is a strong prognostic marker for predicting OS independent <strong>of</strong> age, gender,<br />

CEA, stage and site <strong>of</strong> disease in curatively resected stage II and III CRC.<br />

Stage II Stage III<br />

pTIMP-1 FR < median pTIMP-1 FR > median pTIMP-1 FR < median pTIMP-1 FR > median<br />

N (%) 281 (48%) 302 (52%) 240 (52%) 218 (48%)<br />

# <strong>of</strong> events 71 125 116 145<br />

5-yr OS rate<br />

73.8%<br />

62.6%<br />

53.9%<br />

34.4%<br />

(95% CI) (68.2%-79.4%) (56.8%-68.4%) (47.1%-60.6%) (27.6%-41.2%)<br />

HR (95% CI) ref 1.73 (1.29 – 2.31) ref 1.71 (1.34 – 2.19)<br />

P 0.0002 �.0001<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


222s Gastrointestinal (Colorectal) Cancer<br />

3577 General Poster Session (Board #31F), Mon, 8:00 AM-12:00 PM<br />

Is conversion to resection possible with hepatic arterial infusion (HAI) and<br />

systemic (SYS) even in previously treated patients (pts) with unresectable<br />

colorectal liver metastases (UnCLM)? Presenting Author: Nancy E. Kemeny,<br />

Memorial Sloan-Kettering Cancer Center, New York, NY<br />

Background: Previously, we showed thatHAI with FUDR � dexamethasone<br />

(Dex) plus SYS produced a 47 % resectability rate in a retrospective study<br />

<strong>of</strong> 49 pts with UnCLM. Methods: Prospectively evaluated UnCLM pts in a<br />

new protocol were combined with the above protocol (n�105 pts) and all<br />

were treated with HAI FUDR/Dex � Sys. Unresectability was defined as<br />

diffuse bilateral metastases, involvement <strong>of</strong> all hepatic/portal veins, and/or<br />

inability to preserve remaining liver with adequate perfusion. Factors<br />

associated with conversion were identified using a multivariate logistic<br />

regression model. Overall survival (OS) and progression free survival (PFS)<br />

were calculated from pump placement by the Kaplan-Meier method.<br />

Resectability was a time-dependent covariate in a Cox regression model.<br />

Results: 61 <strong>of</strong> the 105 pts had prior SYS (56 %with prior Oxali) and 45<br />

(74%) were progressing at the time <strong>of</strong> pump placement. In previously<br />

treated pts, 44% underwent resection, with a median OS <strong>of</strong> 45 mos. Of 44<br />

chemo-naïve pts, 57% underwent hepatectomy, with a median OS <strong>of</strong> 68<br />

mos. The following were significantly associated with resection conversion:<br />

lesion number [p�0.02], baseline CEA [p�0.04], females [p�0.03] and<br />

clinical risk score (CRS) [p�0.05]. In multivariate analysis, gender and<br />

CRS remained predictive <strong>of</strong> resectability. Surgery greatly reduced the<br />

hazard <strong>of</strong> death by 67% [HR: 0.33, 95%CI: 0.17-0.61, p�0.0004], after<br />

adjusting for several risk factors (Table). Median PFS was 12 mos for all<br />

pts. Conclusions: Even in previously treated pts,HAI � SYS is an approach<br />

to convert UnCLM to resection. Gender and CRS are associated with<br />

conversion to resectability.<br />

Multivariate Cox regression models.<br />

HR (95%CI) P value<br />

OS<br />

Surgery (yes vs no)* 0.33 (0.17-0.61) �0.001<br />

Gender (F vs M) 0.95 (0.51-1.77) 0.88<br />

CRS (>3vs3vs


3581 General Poster Session (Board #32B), Mon, 8:00 AM-12:00 PM<br />

Mutant (MT) KRAS codon 12 and 13 alleles in patients (pts) with<br />

metastatic colorectal cancer (mCRC): Assessment as prognostic and<br />

predictive biomarkers <strong>of</strong> response to panitumumab (pmab). Presenting<br />

Author: Marc Peeters, Antwerp University Hospital, Edegem, Belgium<br />

Background: Pmab is a fully human monoclonal antibody against the<br />

epidermal growth factor receptor (EGFR). Significant improvement in<br />

progression-free survival (PFS) was demonstrated in pts with wild-type<br />

(WT) KRAS mCRC treated with pmab�FOLFOX4 (1st-line; study<br />

20050203), pmab�FOLFIRI (2nd-line; study 20050181), and pmab<br />

monotherapy (study 20020408). In mCRC, mutations in KRAS codons 12<br />

and 13 are established biomarkers for lack <strong>of</strong> clinical benefit to anti-EGFR<br />

therapies. We retrospectively examined individual MT KRAS codon 12 and<br />

13 alleles as prognostic and predictive biomarkers <strong>of</strong> response in three<br />

phase 3 studies. Methods: Pts were randomized to receive FOLFOX4,<br />

FOLFIRI, or best supportive care �/- pmab 6.0 mg/kg Q2W in trials<br />

20050203, 20050181, and 20020408, respectively. The MT KRAS<br />

codon 12 and 13 alleles (G12A, G12C, G12D, G12R, G12S, G12V, G13D)<br />

were detected using the Therascreen K-RAS Mutation Kit (Qiagen). Results:<br />

MT KRAS codon 12 and 13 alleles were detected in 40% (440/1096),<br />

45% (486/1083), and 43% (184/427) <strong>of</strong> pts in trials 20050203,<br />

20050181, and 20020408, respectively. MT KRAS allele distribution was<br />

conserved across studies and balanced between treatment arms. Baseline<br />

demographic and clinical features were comparable between all MT KRAS<br />

allele subgroups. There was no consistent evidence that any individual MT<br />

KRAS allele, compared to the remaining MT KRAS alleles or the entire MT<br />

KRAS group, differentially impacted PFS or overall survival (OS) in control<br />

arm-treated or pmab-treated pts. Only in the pmab�FOLFOX4 arm <strong>of</strong> study<br />

20050203 were G13D (unfavorably) and G12V (favorably) significantly<br />

associated with OS. Response rates were similar between MT KRAS allele<br />

groups in the 1st- and 2nd-line mCRC treatment setting. Finally, in analyses<br />

<strong>of</strong> pts pooled from all 3 trials, only the G12A KRAS allele emerged as a<br />

significant negative predictive factor for OS. Conclusions: The lack <strong>of</strong><br />

consistent results across three lines <strong>of</strong> therapy indicates pts whose tumors<br />

harbor MT KRAS codon 12 or 13 alleles are unlikely to respond to pmab<br />

therapy. Currently, only pts with WT KRAS mCRC should be treated with<br />

EGFR antibodies.<br />

3583 General Poster Session (Board #32D), Mon, 8:00 AM-12:00 PM<br />

Is there any significant difference between tumor regression grade systems<br />

<strong>of</strong> rectal cancer? Presenting Author: Atthaphorn Trakarnsanga,<br />

Memorial Sloan-Kettering Cancer Center, New York, NY<br />

Background: Tumor regression grade (TRG) is a measure <strong>of</strong> histopathologic<br />

response <strong>of</strong> rectal cancer to preoperative chemoradiation (CRT) and<br />

correlates with outcomes. Several TRG systems have been reported<br />

including Mandard (5, 3 tier), Dowrak/Rodel (5, 3 tier), Memorial Sloan<br />

Kettering Cancer Center (MSKCC), and <strong>American</strong> Joint Committee <strong>of</strong><br />

Cancer (AJCC). The purpose <strong>of</strong> this study is to compare the different TRG<br />

systems and determine which one(s) best predict recurrence and survival.<br />

Methods: Review <strong>of</strong> prospective maintained database from 1998 to 2007<br />

identified 563 patients with locally advanced rectal cancer (T3/4 and/or<br />

N1) and treated with long-course CRT followed by total mesorectal<br />

excision. TRG was determined by measuring proportion <strong>of</strong> tumor mass<br />

replaced by fibrosis. Patients were then classified into the various TRG<br />

schemes which were compared by analyzing association with recurrence<br />

and survival using concordance index (CI) and reclassification index. CI is a<br />

measure that summarizes the predictive strength <strong>of</strong> a marker. Computing<br />

and contrasting CI across several markers is a way <strong>of</strong> selecting the best<br />

prognostic marker. Results: 75% <strong>of</strong> patients were noted to have clinical<br />

stage III disease by endorectal ultrasound or rectal MRI. Following<br />

resection with median follow-up <strong>of</strong> 39.3 months, 2% developed local<br />

recurrence and 17% developed distant metastasis. The median interval<br />

time between completion <strong>of</strong> CRT and surgery is 48 days. 20% demonstarted<br />

complete pathological response. CI <strong>of</strong> the 3 tier Mandard, 3 tier<br />

Dowrak/Rodel, MSKCC and AJCC are 0.665, 0.653, 0.683, and 0.694,<br />

respectively (higher number indicates better prediction). The AJCC was<br />

significantly more accurate in predicting recurrence than the 3- tier<br />

Mandard (p�0.002), and Dowrak/Rodel (p�0.006). AJCC had a higher CI<br />

than MSKCC although it did not reach significance (p�0.068). Comparing<br />

the 3 tier systems, MSKCC was most accurate and correctly reclassified 17<br />

% and 23 % <strong>of</strong> the patients Mandard and Dowrak/Rodel systems,<br />

respectively. Conclusions: TRG predicts recurrence and survival following<br />

combined modality therapy for rectal cancer. The TRG system that recently<br />

was proposed in the 7thAJCC staging is currently the most accurate<br />

predictor.<br />

Gastrointestinal (Colorectal) Cancer<br />

223s<br />

3582 General Poster Session (Board #32C), Mon, 8:00 AM-12:00 PM<br />

A phase II study <strong>of</strong> high-dose cetuximab plus irinotecan in colorectal<br />

cancer patients with KRAS-wild type tumors who progressed on prior<br />

standard dose cetuximab and irinotecan. Presenting Author: Ahmad Ali<br />

Fora, Roswell Park Cancer Institute, Buffalo, NY<br />

Background: Prior clinical data suggest a dose-related response to cetuximab<br />

(cmab) when combined with irinotecan in patients (pts) with KRAS<br />

WT tumors who do not develop grade � 2 rash with standard cmab dosing.<br />

However, the investigation <strong>of</strong> higher doses <strong>of</strong> cmab in the setting <strong>of</strong><br />

acquired or innate resistance to standard dose cmab has not been<br />

previously investigated. We conducted a phase II clinical trial <strong>of</strong> high-dose<br />

cmab plus irinotecan in KRAS WT pts who progressed on standard-dose<br />

cmab plus irinotecan. Methods: Pts who progressed within 4 weeks from<br />

receiving a minimum <strong>of</strong> 6 weeks <strong>of</strong> standard dose cmab plus irinotecan<br />

were included in this study. Cmab was administered at 500 mg/m2 /week<br />

and irinotecan was administered at the same dose/schedule on which each<br />

individual patient previously progressed. All pts received doxycyline 100<br />

mg PO bid starting day 1 <strong>of</strong> treatment. 12-week PFS rate was the primary<br />

endpoint. The regimen was considered interesting if 7/36 patients had<br />

stable disease or response at 12 weeks. The study was closed early after<br />

meeting its primary endpoint. Results: 20 pts were treated on study: median<br />

age 65.5 yrs (44-84), 14 pts were males. 8 pts had grade � 2<br />

hypomagnesaemia (3 grade 3 and 2 grade 4), 6 pts had grade � 2 skin rash<br />

(1 grade 3, 0 grade 4), and 4 pts had grade � 2 diarrhea (1 grade 3, 0 grade<br />

4). 1 pt had a confirmed PR and 12 pts had a SD (at 6 weeks). In 9 pts, SD<br />

was confirmed at 12 weeks (3 pts� 24 weeks). Median PFS and OS were<br />

2.8 and 6.6 months, respectively. The 1-year survival was 25%. Conclusions:<br />

High-dose cmab plus irinotecan is well tolerated with an acceptable rate <strong>of</strong><br />

diarrhea and skin toxicities but with an increased rate <strong>of</strong> grade 3-4<br />

hypomagnesemia. The prolonged disease stabilization and single response<br />

suggest that resistance to standard dose cmab plus irinotecan can be<br />

overcome by cmab dose escalation. The identification <strong>of</strong> predictive markers<br />

<strong>of</strong> response from dose escalation will be necessary to implement this<br />

strategy selectively in KRAS WT colorectal cancer patients.<br />

3584 General Poster Session (Board #32E), Mon, 8:00 AM-12:00 PM<br />

Association <strong>of</strong> genetic variants in obesity-related genes with tumor recurrence<br />

in stage II/III colon cancer. Presenting Author: Robert D. Ladner,<br />

University <strong>of</strong> Southern California Norris Comprehensive Cancer Center, Los<br />

Angeles, CA<br />

Background: High body mass index (BMI) is an established risk factor for<br />

colorectal cancer incidence and death. Recent studies found obesity before<br />

the diagnosis <strong>of</strong> colon cancer was associated with worse survival compared<br />

with normal weight. Single nucleotide polymorphisms (SNPs) <strong>of</strong> obesityrelated<br />

gene have been related with different cancer risk including<br />

colorectal cancer. Here we tested the hypothesis whether SNPs in obesityrelated<br />

genes (PPAR, LEP, NFKB, CD36, DRG1, NGAL, REGIA and<br />

DSCR1) may predict tumor recurrence in adjuvant colon cancer. Methods:<br />

Either blood or FFPE tissue specimens were obtained from 234 patients<br />

(107 females and 127 males; median age 59 years (range 22–78 years))<br />

with stage II (105 patients) or III (129 patients) colon cancer at the<br />

University <strong>of</strong> Southern California/Norris Comprehensive Cancer Center. The<br />

median follow-up was 4.4 years. SNPs in obesity-related genes were<br />

determined by PCR-RFLP or PCR-based direct sequence. The primary<br />

endpoint <strong>of</strong> the study was time to tumor recurrence (TTR). Results: in<br />

univariable analysis, PPAR rs1801282 and DSCR1 rs6517239 were<br />

independently associated with time to tumor recurrence (TTR). Patients<br />

with PPAR CC genotype had longer median TTR 9.4 months (95% C.I: 5.6,<br />

12.4�) compared to those with CG genotype, had median TTR 3.4 months<br />

(95% C.I: 1.7, 16.8�)(p�0.04, log-rank test). Patients with DSCR1 AA<br />

genotype had shorter median TTR 6.6 months (95% C.I: 4.0, 16.8�)<br />

compared to those with AG and GG genotypes, which had longer median<br />

TTR 9.4 months (95% C.I: 5.7, 10.7�)(p�0.027, log-rank test). The<br />

multivariate analysis adjusting for stage and type <strong>of</strong> adjuvant therapy<br />

showed a trend in the association between PPAR rs1801282 and DSCR1<br />

rs6517239 and time to recurrence (Wald test p�0.08 and 0.058,<br />

respectively). Conclusions: Our preliminary results demonstrated polymorphisms<br />

in obesity-related gene might be potential molecular markers to<br />

predict TTR in adjuvant colon cancer. Further large and biomarker<br />

embedded trial needed to confirm our findings.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


224s Gastrointestinal (Colorectal) Cancer<br />

3585 General Poster Session (Board #32F), Mon, 8:00 AM-12:00 PM<br />

FOLFOXIRI plus bevacizumab as first-line treatment <strong>of</strong> BRAF-mutant<br />

metastatic colorectal cancer patients. Presenting Author: Lisa Salvatore,<br />

U.O. Oncologia Medica 2, Azienda Ospedaliero-Universitaria Pisana, Istituto<br />

Toscano Tumori, Pisa, Italy<br />

Background: BRAF V600E mutation plays a negative prognostic role in<br />

metastatic colorectal cancer (mCRC) patients, leading to a median PFS <strong>of</strong><br />

4-6 months with first-line conventional treatments. Recently, results from a<br />

retrospective exploratory analysis suggested that an up-front intensive<br />

treatment with FOLFOXIRI plus bevacizumab could improve the outcome<br />

<strong>of</strong> BRAF mutant mCRC. Methods: Fifteen consecutive BRAF mutant mCRC<br />

patients treated with first-line FOLFOXIRI plus bevacizumab were included<br />

in a validation set. The primary endpoint was 6-months progression free<br />

rate (6m-PFR). Secondary endpoints were: overall survival, response rate<br />

and the pooled analysis <strong>of</strong> all main outcome parameters in both the<br />

exploratory and validation set. Results: In the validation set, 11 out 15<br />

patients included were progression-free at 6 months, for a 6m-PFR <strong>of</strong><br />

73.3%. Primary endpoint was met. At a median follow-up <strong>of</strong> 21.6 months,<br />

median progression-free survival was 9.2 months while median OS has not<br />

yet been reached. In the pooled data analysis <strong>of</strong> the validation set and the<br />

initial retrospective cohort 24 out 25 total patients were evaluable for<br />

response: partial or complete response was obtained in 17 (68%) and in 1<br />

(4%) patient, respectively, for an overall response rate <strong>of</strong> 72%. At a median<br />

follow-up <strong>of</strong> 34.1 months, the pooled set <strong>of</strong> patients showed a median PFS<br />

<strong>of</strong> 11.8 months and a median OS <strong>of</strong> 23.8 months. Conclusions: These data<br />

suggest that FOLFOXIRI plus bevacizumab could be a reasonable option for<br />

the first-line treatment <strong>of</strong> BRAF mutant metastatic colorectal cancer<br />

patients.<br />

3587 General Poster Session (Board #32H), Mon, 8:00 AM-12:00 PM<br />

Activity <strong>of</strong> the anti-IGF-1R antibody dalotuzumab (MK-0646) in KRASmutant<br />

colorectal cancer: Preclinical and clinical data. Presenting Author:<br />

Sriram Sathyanarayanan, Merck Research Laboratories, Boston, MA<br />

Background: KRAS mutated metastatic colorectal cancer (mCRC) is inherently<br />

resistant to EGFR targeted therapy and carries an inferior prognosis to<br />

wild-type disease with a median survival <strong>of</strong> approximately 16 months.<br />

Methods: The activity <strong>of</strong> dalotuzumab (Dz) has been investigated in KRAS<br />

mutant pre-clinical colon cancer models. In addition efficacy data relating<br />

to a sub-set <strong>of</strong> KRAS mutant mCRC patients enrolled in a randomized<br />

phase II/III study (initiated prior to the introduction <strong>of</strong> KRAS genotyping) <strong>of</strong><br />

Dz in combination with cetuximab (Cx) and irinotecan (Ir) are reported. All<br />

patients had received Cx and Ir with either placebo (n�20), weekly Dz<br />

(n�18), or 2 weekly Dz (n�31). Results: In pre-clinical models, Dz was<br />

found to be effective in inhibiting IGF-1 mediated cellular growth.<br />

Furthermore, in colorectal cancer xenograft models high IGF-1 was found<br />

to identify a sub-set <strong>of</strong> Dz responsive tumours. Combination studies<br />

demonstrated that Irinotecan exposure resulted in the activation <strong>of</strong> IGF-1R<br />

and PI3K signaling pathways, representing a possible cellular survival<br />

mechanism. In xenograft models the combination <strong>of</strong> Dz with irinotecan<br />

produced lasting tumor growth inhibition that persisted even after treatment<br />

withdrawal. Outcome data are reported for 69 patients with KRAS<br />

mutant mCRC enrolled in the randomized study. Efficacy data demonstrated<br />

differential activity between colon versus rectal tumours with<br />

evidence <strong>of</strong> activity in the rectal subgroup. Molecular analysis suggests that<br />

this observation may be mediated by differential IGF-1 expression.<br />

Conclusions: These data suggest that Dz in combination with irinotecan may<br />

have utility in the treatment <strong>of</strong> KRAS mutated colorectal cancer patients.<br />

Based on these data Dz is being further evaluated in a molecularly selected<br />

population <strong>of</strong> mCRC.<br />

Dz vs placebo arm<br />

Hazard ratio (95% CI)<br />

PFS OS<br />

Weekly Dz 2 weekly Dz Weekly Dz 2 weekly Dz<br />

Mean normalized IGF-1<br />

Cq values (rtPCR)<br />

All patients (n�69) 0.75 (0.35, 1.58) 1.14 (0.59, 2.20) 0.88 (0.42, 1.84) 0.96 (0.49, 1.90)<br />

Colon (n�37) 1.50 (0.60, 3.74) 1.63 (0.67, 3.94) 1.21 (0.47, 3.07) 1.31 (0.54, 3.19) 5.22<br />

Rectal (n�32) 0.17 (0.04, 0.79) 0.31 (0.08 1.22) 0.32 (0.09, 1.20) 0.37 (0.11, 1.18) 4.52<br />

p�0.043<br />

3586 General Poster Session (Board #32G), Mon, 8:00 AM-12:00 PM<br />

Neoadjuvant capecitabine, oxliplatin, and bevacizumab (CAPOX-B) in<br />

intermediate-risk rectal cancer (RC) patients defined by magnetic resonance<br />

(MR): GEMCAD 0801 trial. Presenting Author: Carlos Fernandez-<br />

Martos, Medical Oncology Department, Fundacion Instituto Valenciano De<br />

Oncologia, Valencia, Spain<br />

Background: Retrospective data suggest that RT might not be needed in all<br />

patients with stage II/III RC. Modern systemic therapy might have local<br />

efficacy similar to chemoradiation (CRT). Methods: A multicenter phase II<br />

trial was undertaken to evaluate safety and efficacy <strong>of</strong> neoadjuvant<br />

CAPOX-B in patients with T3 middle third rectal adenocarcinoma. Eligible<br />

patients (pts) had measurable disease at the baseline and candidate for R0<br />

total mesorectal escision (TME) with intermediate-risk defined by pelvic<br />

MR a) T3 with distal border <strong>of</strong> tumor � 5 cm from the anal verge and below<br />

the sacral promontory. b) tumor �2 mm from the mesorectal fascia. Pts<br />

received 4 cycles <strong>of</strong> Cap 2000 mg/m2 (d1-14), Ox 130 mg/m2 (d1) and B<br />

7.5 mg/kg (d1) every 3 weeks (last cycle without B). Pts undergo re-staging<br />

with MR. One radiologist reviewed all pre- and post-treatment MR scans<br />

independently. Pts without progression proceed to TME 4-6 weeks from the<br />

last cycle. If progression, pts were to be referred for pre-op cap/RT followed<br />

by TME. 1º Endpoint: Tumor Response (RECIST). Design: Simon 2-stage;<br />

28 pts 1st stage and 46 pts 2nd stage. We report data on the planned<br />

analysis <strong>of</strong> pts included for 1st stage. Results: 28 eligible pts (10F/18M)<br />

were enrolled from 7/09-5/11. Tumor response, compliance and toxicity<br />

details are shown in table below. Two pN2 pts received postop Cap/RT.<br />

Conclusions: Neoadjuvant CAPOX-B is active and safe. Early parameters <strong>of</strong><br />

efficacy are encouraging and seem similar to those observed with CRT.<br />

Efficacy<br />

MR response<br />

MR overall response<br />

MR complete response<br />

MR partial response<br />

MR stable disease<br />

p response<br />

pCR ( ypT0N0)<br />

pLN�<br />

TRG intermediate � total regression �<br />

Downstaging<br />

CRM free<br />

R0 resection<br />

Compliance<br />

4 cycles<br />

24*<br />

21 (87.5%) 95%; CI 68%-97%<br />

1 (4%)<br />

20 (83%)<br />

3 (13%)<br />

27v<br />

4 (15%)<br />

10 (37%)<br />

17 (63%)<br />

10 (37%)<br />

26 (96%)<br />

27 (100%)<br />

24 (86%)<br />

Toxicity G3� 14 (50%)<br />

Surgical complications G3� 3 (11%)<br />

Abbreviations: pCR, pathologic complete response; CRM, circumferential resection margin;<br />

TRG, tumor regression grade. � Dworak 2�3�4;*4MRnotdone. 1 died (diarrhea/renal<br />

failure), 1 perforation after 3th cycle, 2 withdrawal;v pts underwent surgery.<br />

3588 General Poster Session (Board #33A), Mon, 8:00 AM-12:00 PM<br />

Early tumor shrinkage in patients with metastatic colorectal cancer<br />

receiving first-line treatment with cetuximab combined with either CAPIRI<br />

or CAPOX: An analysis <strong>of</strong> the AIO KRK 0104 trial. Presenting Author:<br />

Dominik Paul Modest, Department <strong>of</strong> Hematology and Oncology, Klinikum<br />

Grosshadern and Comprehensive Cancer Center, LMU Munich, Munich,<br />

Germany<br />

Background: This study investigated the impact <strong>of</strong> early tumor-shrinkage<br />

(ETS) on progression-free (PFS) and overall survival (OS) in patients with<br />

metastatic colorectal cancer (mCRC) treated within the AIO KRK-0104trial.<br />

ETS was previously shown to be a predictor <strong>of</strong> treatment response to<br />

cetuximab. The present analysis, for the first time, evaluates the correlation<br />

<strong>of</strong> ETS with outcome parameters in patients undergoing capecitabinebased<br />

therapy. It also aims to correlate the predictive value <strong>of</strong> ETS and<br />

cetuximab-induced skin toxicity. Methods: The AIO KRK0104 trial was<br />

performed as a randomised study comparing capecitabine/oxaliplatin<br />

(CAPOX) plus cetuximab to capecitabine/irinotecan (CAPIRI) plus cetuximab<br />

in the first-line treatment <strong>of</strong> mCRC. 121 patients were available for<br />

evaluation <strong>of</strong> ETS at 6 weeks. ETS was defined as a relative change <strong>of</strong><br />

�20% in the sum <strong>of</strong> the longest diameters <strong>of</strong> target lesions compared to<br />

baseline. Results: Tumors <strong>of</strong> 63 patients (71% KRAS wild-type (wt), 100%<br />

skin reactions) developed ETS, 58 tumors did not develop ETS (53% KRAS<br />

wt, 86% skin reactions). ETS was associated with prolonged PFS (8.9 vs.<br />

4.7 months, p�0.001, hazard ratio: 0.37) and OS (31.6 vs. 15.8 months,<br />

p�0.005, hazard ratio: 0.48) in patients with KRAS wt tumors but not in<br />

patients with KRAS mutant mCRC. ETS occurred more frequently with<br />

CAPOX- vs CAPIRI-based therapy (p�0.05). There was a highly significant<br />

correlation between the occurrence <strong>of</strong> ETS and cetuximab-related skin<br />

toxicity <strong>of</strong> any grade (I-III) (p�0.002). ETS was documented more<br />

frequently in patients with liver metastasis (p�0.09), metastatic involvement<br />

<strong>of</strong> �2 organs (p�0.09), KRAS wild-type tumors (p�0.06) and no<br />

previous adjuvant chemotherapy (p�0.06). Conclusions: In patients with<br />

KRAS wild-type mCRC receiving capecitabine- and cetuximab-based firstline<br />

therapy ETS correlates with prolonged PFS and OS. ETS also correlates<br />

with cetuximab-induced skin toxicity. Both parameters may therefore serve<br />

as post-randomisation surrogate markers <strong>of</strong> favourable outcome for cetuximab-based<br />

treatment.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


3589 General Poster Session (Board #33B), Mon, 8:00 AM-12:00 PM<br />

Examining the effects <strong>of</strong> metformin on survival outcome in stage II/III<br />

colorectal cancer patients with diabetes mellitus. Presenting Author: Guek<br />

Eng Lee, Department <strong>of</strong> Medical Oncology, National Cancer Centre,<br />

Singapore<br />

Background: Insulin resistance and deregulation <strong>of</strong> the insulin-like growth<br />

factor (IGF) axis is implicated in colonic carcinogenesis. Metformin inhibits<br />

hepatic gluconeogenesis and increases insulin sensitivity. It induces AMPK<br />

activation, thus inhibiting mTOR and downstream survival and proliferation<br />

pathways. We evaluated the effects <strong>of</strong> metformin on survival outcomes <strong>of</strong><br />

patients with stage 2 and 3 colorectal cancer (CRC). Methods: Among 1455<br />

patients with stage II or III colorectal cancer, we identified 344 patients<br />

with both CRC and diabetes mellitus. 219 diabetic patients received<br />

metformin and 125 diabetic patients were not receiving metformin.<br />

Patient’s demographics, clinical and histopathologic characteristics, overall<br />

survival, time to recurrence and relapse-free survival were evaluated.<br />

Molecular analysis for AMPK, and mTOR pathway on archival tissue is<br />

ongoing. Results: After a median follow-up <strong>of</strong> 78 months (6.5 years), there<br />

was no difference in survival outcomes between diabetic and non-diabetic<br />

patients. Among diabetic patients, there were 99 relapses and 90 deaths.<br />

Metformin use was associated with improved overall survival (HR 0.23;<br />

95%CI: 0.15-0.35 p�0.01), time to recurrence (HR 0.63; 95%CI:<br />

0.41-0.96) and relapse-free survival (HR 0.47; 95%CI:0.33-0.67,p�0.01).<br />

In multivariate analysis, after adjustment for T stage, N stage and patients’<br />

age, metformin use remained associated with improved overall survival (HR<br />

0.18, p�0.01); time to recurrence (HR 0.55; p�0.04) and relapse-free<br />

survival (HR 0.44, p�0.01). Results <strong>of</strong> molecular correlative studies<br />

performed on archival tissue will be presented at the meeting. Conclusions:<br />

Among patients with diabetes and colorectal cancer, use <strong>of</strong> metformin is<br />

associated with improved survival outcomes in both univariate and multivariate<br />

analysis lending support to the hypothesis that it may have anti-cancer<br />

activity.<br />

3591 General Poster Session (Board #33D), Mon, 8:00 AM-12:00 PM<br />

First-line chemotherapy plus cetuximab in patients grouped according to<br />

prognostic risk factors: Analysis <strong>of</strong> the CRYSTAL and OPUS studies.<br />

Presenting Author: Gunnar Folprecht, University Hospital Carl Gustav<br />

Carus, Dresden, Germany<br />

Background: Analysis <strong>of</strong> randomized trials has shown that mCRC patients<br />

(pts) can be divided into prognostic risk groups according to baseline<br />

clinical parameters including ECOG performance status, white blood cell<br />

count (WBC), alkaline phosphatase (ALP) and number <strong>of</strong> metastatic (met)<br />

sites (Köhne C, et al. Ann Oncol 2002;13:308-17). The effect <strong>of</strong> adding<br />

cetuximab to first-line chemotherapy (CT) on overall survival (OS) in these<br />

groups <strong>of</strong> KRAS wild-type mCRC pts was investigated in the CRYSTAL and<br />

OPUS studies. Methods: Pt risk groups were: low-risk (LRG� ECOG 0/1, 1<br />

met site) intermediate-risk (IRG� ECOG �1, �1 met site, ALP �300 U/L<br />

or, ECOG�1, low WBC, 1 met site) and high-risk (HRG� ECOG�1, �1 met<br />

site, ALP�300 U/L or ECOG�1, high WBC, or ECOG�1, low WBC and �2<br />

met sites). Exploratory analyses comprised estimates <strong>of</strong> effects using Cox‘s<br />

proportional hazards model for OS on individual pt data and comparison <strong>of</strong><br />

treatment arms by log-rank test. Results: Data are shown in the table. In the<br />

pooled analyses, in both treatment arms OS was longest in LRG pts and<br />

shortest in HRG pts. Adding cetuximab to CT led to marked improvements<br />

in OS in the HRG (Hazard ratio [HR] 0.765, 95% CI 0.506–1.157,<br />

p�0.203) and IRG (HR 0.781, 95% CI 0.622–0.981, p�0.033), while<br />

improvements in LRG pts (HR 0.869, 95% CI 0.672–1.124, p�0.287)<br />

were observed only after prolonged follow up. Data were similar in the<br />

separate CRYSTAL and OPUS studies. Conclusions: The analysis confirms<br />

the concept <strong>of</strong> prognostic risk groups for OS according to baseline clinical<br />

parameters in pts with KRAS wt mCRC. Benefit from the addition <strong>of</strong><br />

cetuximab to first-line CT in terms <strong>of</strong> OS appears to be more pronounced in<br />

the IRG and HRG.<br />

CT CT � cetuximab<br />

Study/risk group N Median OS, mo (95% CI) N Median OS, mo (95% CI)<br />

Pooled<br />

LRG 164 25.7 (21.9–28.7) 173 27.0 (24.8–29.5)<br />

IRG 213 16.4 (14.9–20.0) 166 22.2 (19.5–25.7)<br />

HRG 59 12.6 (9.2–14.4) 46 17.7 (13.1–19.3)<br />

CRYSTAL<br />

LRG 127 26.1 (21.2–29.9) 132 28.2 (24.9–31.5)<br />

IRG 172 16.6 (14.8–20.4) 141 23.5 (20.0–29.6)<br />

HRG 43 12.8 (9.2–14.6) 31 16.7 (8.3–19.0)<br />

OPUS<br />

LRG 37 23.9 (19.5–n.e.) 41 26.0 (22.9–n.e.)<br />

IRG 41 16.2 (12.0–21.6) 25 18.4 (11.0–19.8)<br />

HRG 16 11.8 (4.8–18.5) 15 19.9 (13.1–30.4)<br />

Abbreviation: n.e., not estimable.<br />

Gastrointestinal (Colorectal) Cancer<br />

225s<br />

3590 General Poster Session (Board #33C), Mon, 8:00 AM-12:00 PM<br />

U.S. patients receiving resin 90Y microspheres for unresectable colorectal<br />

liver metastases: A multicenter study <strong>of</strong> 506 patients. Presenting Author:<br />

Andrew S. Kennedy, Cancer Centers <strong>of</strong> North Carolina, Cary, NC<br />

Background: Implantation <strong>of</strong> radioactive microspheres via the hepatic artery<br />

with Yttrium-90 ( 90Y) is termed “radioembolization, (RE)”. Resin microspheres<br />

were FDA cleared for use in colorectal liver metastases (mCRC) in<br />

2002. Rapid worldwide acceptance <strong>of</strong> this therapy has resulted in greater<br />

than 30,000 procedures conducted in the past 10 years. This investigatorinitiated<br />

study focuses on RE treatment outcomes in US-only patients since<br />

2002. Methods: A retrospective multi-institutional study was designed to<br />

analyze the outcome <strong>of</strong> consecutively treated mCRC patients undergoing<br />

RE by experienced treatment centers in the USA using resin 90Y microspheres.<br />

IRB approval was obtained by each center with independent data<br />

collection and analyses. Primary endpoints: CTC 3.0ae toxicity, RECIST<br />

response and survival; baseline treatment parameters, prior chemotherapy,<br />

and liver directed therapies. Results: 506 patients at 10 institutions (193<br />

M, 313 F) received 770 RE treatments; median age � 60.4 years (20.8 –<br />

91.9 years); median number <strong>of</strong> RE treatments per patient � 1.0 (1-5<br />

treatments). Active extrahepatic disease was present at first RE in 34.8%<br />

<strong>of</strong> patients. The majority (90%) <strong>of</strong> patients received prior chemotherapy,<br />

with 30.6% also having undergone prior hepatic surgery/ablative procedures.<br />

Median follow up after RE � 8.4 mo. (0.4 – 67.6 mo.) with median<br />

survival � 10.1 mo. (95% CI 9.1 – 12.0). For first RE treatment, median<br />

tumor volume was 146.0 mL (2.8 – 3228.0 mL). Median radiation activity<br />

delivered � 1.18 GBq (0.12 – 2.29 GBq), lung shunt median � 4.8 % (.02<br />

– 45%). Total grade 1-3 events were 32% GI, 44% fatigue and 1% liver<br />

failure. Only 2.4% <strong>of</strong> all treatments required an overnight stay postprocedure.<br />

Conclusions: The modern USA experience <strong>of</strong> 90Y therapy for<br />

unresectable, heavily pretreated mCRC liver metastases is encouraging<br />

with a median survival <strong>of</strong> 10.1 months after first RE procedure. Toxicity was<br />

mild and <strong>of</strong> short duration in most patients. RECIST response is being<br />

analyzed currently as are data <strong>of</strong> additional patients.<br />

3592 General Poster Session (Board #33E), Mon, 8:00 AM-12:00 PM<br />

A UGT1A1 *28 and *6 genotype-directed phase I study <strong>of</strong> irinotecan plus<br />

infusional leucovorin and 5-fluorouracil (sLV5FU2) in patients with previously<br />

untreated metastatic colorectal cancer (mCRC). Presenting Author: Yong Sang<br />

Hong, Department <strong>of</strong> Oncology, Asan Medical Center, University <strong>of</strong> Ulsan<br />

College <strong>of</strong> Medicine, Seoul, South Korea<br />

Background: We designed a phase I study to determine maximum tolerated dose<br />

(MTD) <strong>of</strong> irinotecan when combined with sLV5FU2 in mCRC patients (pts).<br />

Methods: Pts were genotyped for UGT1A1 *28 and *6, and stratified into 3<br />

groups according to the number <strong>of</strong> defective allele (DA), designated 0 (*1/*1), 1<br />

(*1/*28, *1/*6), and 2 (*28/*28, *6/*6, *6/*28). Within each group, the dose <strong>of</strong><br />

irinotecan was escalated (table) in combination with fixed dose <strong>of</strong> sLV5FU2.<br />

Plasma drug levels and dose-limiting toxicity (DLT) were evaluated at cycle 1.<br />

Results: A total <strong>of</strong> 43 pts were accrued: 19 for 0 DA, 20 for 1 DA and 4 for 2 DA<br />

group. The MTD was estimated as 300 mg/m2 /2-week for the 1 DA group with 2<br />

DLTs in the level 3, and the MTD was not reached for the 0 DA group with 1 DLT<br />

in the level 4 (table). The mean relative extents <strong>of</strong> glucuronidation, AUClast ratio<br />

<strong>of</strong> SN-38G to SN-38, were 9.36, 6.81, and 5.09 for the 0, 1, and 2 DA groups,<br />

respectively (P�0.017). Of the 43 pts, five pts showed AUClast, SN38 that<br />

exceeded 400 ng·h/mL (1.02 umol·h/L) and DLT was observed in 40% (2/5).<br />

The overall response rate was 67.4% (95% CI, 51.5-80.9) with 6 complete<br />

responses and 23 partial responses. Median progression-free and overall survival<br />

was 8.0 months (95% CI, 7.1-8.9) and 25.6 months (95% CI, 23.4-27.7),<br />

respectively. Grade 3 or 4 toxicity during all treatment cycles included<br />

neutropenia (79% [0 DA]; 90% [1 DA]; 75% [2 DA]), leucopenia (21%; 30%;<br />

0%), febrile neutropenia (0%; 10%; 0%) and diarrhea (0; 5%; 0) per patient.<br />

Conclusions: Dose-normalized exposure <strong>of</strong> SN38 was significantly higher in the 2<br />

DA UGT1A1 group. Higher doses <strong>of</strong> irinotecan based on UGT1A1 genotyping are<br />

feasible when combined with sLV5FU2 in mCRC pts. The recommended dose <strong>of</strong><br />

irinotecan was 330, 270, 150 mg/m2 /2-week for pts with 0, 1, 2 DA based on<br />

pharmacokinetic analysis.<br />

Level and dose<br />

Number <strong>of</strong> patients<br />

Group <strong>of</strong> irinotecan Treated DLT occurred<br />

DLTs<br />

0DA 1 240 * 4 0<br />

2 270 3 0<br />

3 300 6 0<br />

4 330 6 1 Gr 4 neutropenia<br />

1DA 1 240 * 3 0<br />

2 270 14 0<br />

3 300 3 2 Gr 4 neutropenia,<br />

Gr 3 febrile neutropenia<br />

2DA -1 150 * 4 0<br />

0 180 - -<br />

* Starting dose <strong>of</strong> irinotecan for each dose level (mg/m2 ).<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


226s Gastrointestinal (Colorectal) Cancer<br />

3593 General Poster Session (Board #33F), Mon, 8:00 AM-12:00 PM<br />

Phase II trial <strong>of</strong> combined chemotherapy with irinotecan, S-1, and<br />

bevacizumab (IRIS/Bev) in patients with metastatic colorectal cancer:<br />

Update analysis—Hokkaido Gastrointestinal Cancer Study Group (HGCSG)<br />

trial. Presenting Author: Satoshi Yuki, Department <strong>of</strong> Gastroenterology,<br />

Hokkaido University Hospital, Sapporo, Japan<br />

Background: The FIRIS study (Muro K et al. Lancet Oncol 2010;11:853–<br />

860) previously demonstrated the non-inferiority <strong>of</strong> Irinotecan plus S-1(IRIS)<br />

to FOLFIRI for metastatic colorectal cancer(mCRC), with progression-free<br />

survival (PFS) as the primary endpoint. We previously reported that IRIS<br />

plus bevacizumab(IRIS/bev) is very effective as first-line treatment (Komatsu<br />

Y et al. ESMO 2010). We now report the updated results <strong>of</strong> this study.<br />

Methods: Eligible patients had to have mCRC with a confirmed diagnosis <strong>of</strong><br />

adenocarcinoma, an age <strong>of</strong> �20 years, ECOG performance status (PS) <strong>of</strong><br />

0-1, and no history <strong>of</strong> prior chemotherapy. S-1 40-60 mg twice daily p.o.<br />

was given on days 1-14 and irinotecan 100 mg/m2 and bevacizumab 5<br />

mg/kg i.v. were given on days 1 and 15 <strong>of</strong> a 28-day cycle. The primary<br />

endpoint was safety. The secondary endpoints included overall response<br />

(OR), progression-free survival (PFS), and overall survival (OS). Results: The<br />

target number <strong>of</strong> 53 patients was enrolled as <strong>of</strong> March 2009. The results<br />

are reported for 52 patients with evaluable lesions. The clinical characteristics<br />

<strong>of</strong> the patients were as follows. The median age was 63.5 years (range,<br />

48 to 82). The male:female ratio was 3:2. The performance status on the<br />

Eastern Cooperative Oncology Group scale was 0. In January 2012, on<br />

safety analysis, the incidence <strong>of</strong> grade 3 or 4 neutropenia was 27%. The<br />

incidences <strong>of</strong> other grade 3 or 4 adverse reactions were as follows: diarrhea,<br />

17%; anorexia, 4%; stomatitis, 2%; hypertension, 21%; and gastrointestinal<br />

perforation, 0%. The overall response rate was 63.5%. Three patients<br />

had complete response. Thirty patients had partial response, 16 had stable<br />

disease, none had progressive disease, and 3 were not evaluable. Median<br />

progression-free survival was 17.0 months and median survival time was<br />

39.6 months. Conclusions: IRIS/Bev is a remarkably active and generally<br />

well-tolerated first-line treatment for patients with mCRC. Randomized<br />

control trial comparing this regimen with oxaliplatin containing regimen(X-<br />

ELOX or mFOLFOX6 plus bevacizumab) is being planned.<br />

3595 General Poster Session (Board #33H), Mon, 8:00 AM-12:00 PM<br />

Neoadjuvant radiotherapy (RT) combined with capecitabine (Cape) and<br />

sorafenib (Sor) in patients with locally advanced, K-ras-mutated rectal<br />

cancer (LARC): A phase I/II trial (SAKK 41/08). Presenting Author: Roger<br />

Von Moos, Kantonsspital Graubünden, Chur, Switzerland<br />

Background: 40% <strong>of</strong> patients (pts) with LARC present with K-ras mutation.<br />

Sor is an inhibitor <strong>of</strong> ras/raf and <strong>of</strong> VEGFR. Furthermore Sor has radiosensitizing<br />

effects. These facts build a strong rationale to use Sor in combination<br />

with preoperative RT and Cape in pts with K-ras mutant tumors. Methods:<br />

Pts with K-ras mutated mrT3-4 and / or mrN�, M0 disease were recruited<br />

in cohorts <strong>of</strong> 7 pts per dose level (DL). Dose limiting toxicity (DLT) was<br />

defined as any G3 or higher non hematological toxicity (tox) or haematological<br />

tox during � 7 days, which are possibly, probably or definitely related to<br />

trial treatment occurring during and up to 4 weeks after last administration.<br />

If � 2 out <strong>of</strong> 7 pts experienced a DLT, then the next cohort was treated at a<br />

higher dose level. If � 2 out <strong>of</strong> 7 pts suffered from a DLT, this DL would be<br />

considered too toxic and a dose level below would be tested in the next<br />

cohort. The highest dose level with � 2 out <strong>of</strong> 7 pts experienced a DLT<br />

would be recommended for the phase II part. In all dose levels RT was given<br />

in 25 fractions at 1.8Gy (45Gy). Results: In 8 centers in Switzerland a total<br />

<strong>of</strong> 21 pts in 3 cohorts have been treated in the phase 1 study. After<br />

observing severe skin toxicity and diarrhea (2 pt with skin toxicity G3, one<br />

patient (pt) with diarrhea G3 plus hand-foot syndrome (HFS) (G3)) an<br />

amendment was implemented to reduce the Sor dose from 400mg BID to<br />

once daily. In DL 1 after the amendment, 2 pts experienced a DLT (enteritis<br />

G3, dermatitis G3). Two pts in DL 2 suffered from a DLT (cystitis G3 plus<br />

HFS G3, cardiac ischemia G3). Further non dose-limiting G3 toxicities<br />

after the amendment were lymphopenia (G3) (1pt, DL 2) and hypocalcaemia<br />

(G3) (1 pt, DL 2). No Grade 4 toxicity was seen. DL 2 is the<br />

recommended dose for phase II. Conclusions: After reducing the Sor dose to<br />

400mg once daily, neoadjuvant treatment with Sor in combination with full<br />

dose Cape and RT was tolerable and the toxicities manageable.<br />

Dose level<br />

(each with 7 pts)<br />

Capecitabine (mg/m 2 )<br />

per day during 5 weeks<br />

Sorafenib (mg)<br />

per day during 5 weeks<br />

DL 1 before amendment 2 x 600 2 x 400<br />

DL 1 after amendment 2 x 600 1 x 400<br />

DL 2 after amendment 2 x 825 1 x 400<br />

3594 General Poster Session (Board #33G), Mon, 8:00 AM-12:00 PM<br />

Functional SNPs in vascular endothelial growth factor (VEGF-A) and overall<br />

survival (OS) in bevacizumab-treated patients with metastatic colorectal<br />

cancer (mCRC). Presenting Author: Janet E. Murphy, Massachusetts<br />

General Hospital Cancer Center, Boston, MA<br />

Background: Functional SNPs in VEGF-A are prognostic for OS in several<br />

malignancies, but not described definitively in mCRC. In bevacizumabtreated<br />

patients with advanced breast cancer, VEGF -2578AA and -1154A<br />

were predictive <strong>of</strong> OS—the latter SNP reported in a recent mCRC series as<br />

well. We examined the association between five functional VEGF-A SNPs<br />

and OS in a large cohort <strong>of</strong> bevacizumab-treated patients with mCRC.<br />

Methods: 403 patients with mCRC treated from 2004-2010 were included<br />

in a retrospective analysis. DNA extraction, genotyping, and SNP evaluation<br />

were performed according to standard protocols. Survival was calculated<br />

from the time <strong>of</strong> Stage IV diagnosis until death. Data were censored as<br />

<strong>of</strong> 12/31/2010. Results: There were 279 deaths in this group <strong>of</strong> 403<br />

patients (69%). Median age was 55.7 (24-86 y), and 54% <strong>of</strong> patients were<br />

male. Age, sex, race, tumor grade, chemotherapies, and curative surgery<br />

(metastatectomy to negative margins) were considered. Significant clinical<br />

predictors <strong>of</strong> OS in univariate Cox modeling were cetuximab treatment<br />

[HR�1.46; 95% CI 1.13-1.88; p�0.0014], irinotecan treatment<br />

[HR�2.10; 1.32-3.35; p�0.001], and curative surgery [HR�0.36; 0.25-<br />

0.75; p�0.001]. Significant negative interaction was found between both<br />

cetuximab and irinotecan and curative surgery, and in modeling that<br />

included this interaction, only surgery remained predictive. In multivariate<br />

analysis, no association was found between VEGF-A and OS (Table).<br />

Conclusions: There was no association between five functional VEGF-A<br />

SNPs and OS in this large bevacizumab-treated mCRC cohort.<br />

SNP Allelic pairs HR [95% CI] Allelic pairs HR [95% CI]<br />

VEGF-2578 C/A vs C/C 1.10 [0.82-1.46] A/A vs C/C 1.06 [0.77-1.46]<br />

VEGF-1154 A/G vs A/A 1.14 [0.89-1.47] G/G vs A/A 0.93 [0.63-1.37]<br />

VEGF-460 C/T vs T/T 1.02 [0.76-1.36] C/C vs T/T 0.95 [0.69-1.30]<br />

VEGF�405 C/G vs G/G 1.13 [0.87-1.47] C/C vs G/G 0.94 [0.67-1.32]<br />

VEGF�936 C/T vs C/C 1.14 [0.87-1.49] T/T vs C/C 0.90 [0.37-2.17]<br />

3596 General Poster Session (Board #34A), Mon, 8:00 AM-12:00 PM<br />

Treating patients with colorectal liver metastasis: A national decisionmaking<br />

analysis to understand choice <strong>of</strong> therapy. Presenting Author:<br />

Timothy M. Pawlik, Johns Hopkins Hospital, Baltimore, MD<br />

Background: Criteria for resectability <strong>of</strong> colon cancer liver metastases (CLM)<br />

are evolving, yet little is known about how physicians choose a therapeutic<br />

strategy for potentially resectable CLM. Methods: Physicians completed a<br />

national web-based survey that consisted <strong>of</strong> varied CLM case scenarios.<br />

Respondents choose among 3 treatment strategies: immediate liver resection(LR),<br />

preoperative chemotherapy followed by surgery(C¡LR), or palliative<br />

chemotherapy (PC). Data were analyzed using multinomial logistic<br />

regression, yielding relative risk ratios (RRR). Results: Of 219 respondents,<br />

79% practiced at academic centers and 63% were in practice �10 years.<br />

Median number <strong>of</strong> cases evaluated was 4/month. Surgical training varied:<br />

51% surgical oncology (SO), 44% hepatobiliary/transplant (HB/LT), 5% no<br />

fellowship. While each factor impacted choice <strong>of</strong> CLM therapy, the relative<br />

impact differed (p�0.01). Synchronous CLM presentation increased the<br />

choice <strong>of</strong> C¡LR (OR 4.27) and PC (OR 3.10) versus LR (p�0.001). For<br />

patients with more extensive intrahepatic disease (i.e., 5 tumors, both<br />

lobes) respondents strongly favored C¡LR (OR 5.12) and PC (OR 9.60)<br />

versus LR (p�0.001). The presence <strong>of</strong> hilar lymph-node disease was<br />

associated with a strong aversion to LR with surgeons more likely to choose<br />

C¡LR (OR 8.92) or PC (OR 49.9) (p�0.001). Interestingly, even the<br />

presence <strong>of</strong> a resectable solitary lung metastasis strongly deterred choice <strong>of</strong><br />

LR with respondents favoring C¡LR (OR 4.43) or PC (OR 6.97) (p�0.001).<br />

While other factors such as patient age, tumor size, and extent <strong>of</strong> resection<br />

impacted choice <strong>of</strong> therapy, the relative size <strong>of</strong> these effects was modest.<br />

After controlling for clinical factors, surgeons with more years in practice<br />

were more likely to choose LR (RRR 1.4). SO-trained surgeons were more<br />

likely than HB/LT-trained surgeons to choose C¡LR (RRR 2.5) or PC (RRR<br />

4.15)(p�0.001). Conclusions: This is the first study to define the relative<br />

impact <strong>of</strong> key clinical factors on choice <strong>of</strong> therapy for CLM. While clinical<br />

factors influence choice <strong>of</strong> therapy, surgical subspeciality and physician<br />

experience are also important determinants <strong>of</strong> care.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


3597 General Poster Session (Board #34B), Mon, 8:00 AM-12:00 PM<br />

Genetic variants <strong>of</strong> kinases suppressors <strong>of</strong> ras in KRAS-BRAF wild-type<br />

metastatic colorectal cancer patients treated with cetuximab and irinotecan.<br />

Presenting Author: Leonor Benhaim, University <strong>of</strong> Southern California<br />

Norris Comprehensive Cancer Center, Los Angeles, CA<br />

Background: Scaffold proteins kinase suppressor <strong>of</strong> ras (KSRs) have<br />

emerged as critical modulators <strong>of</strong> the EGFR pathway that binds to RAS<br />

promoting ERK activation. KSR1 and KSR2 have been shown to be over<br />

expressed in various solid tumours including colon cancer. In-vitro, the<br />

down-regulation <strong>of</strong> KSR results in reduction <strong>of</strong> cells proliferation and<br />

tumour growth independently to KRAS status. Moving from this biological<br />

rational we investigated the correlation between functional polymorphisms<br />

in the KSR genes and clinical outcomes in patients with KRAS and BRAF<br />

wild-type mCRC treated with cetuximab and irinotecan. Methods: Germline<br />

DNA was obtained from peripheral blood <strong>of</strong> patients with mCRC KRAS and<br />

BRAF wild- type resistant to irinotecan treated with a combination <strong>of</strong><br />

cetuximab and irinotecan. Seven functionally relevant SNP were selected<br />

on the basis <strong>of</strong> public literature resources and databases, 4 in the KSR1<br />

and 3 in the KSR2 genes and analyzed by PCR followed by direct<br />

sequencing. All candidate SNPs were evaluated for association with<br />

response rate, PFS and OS. Results: A total <strong>of</strong> 66 patients were included.After<br />

a median follow-up <strong>of</strong> 13.3 months, median PFS and OS were<br />

4.7 and 11.3 months respectively. Seventeen out <strong>of</strong> 66 patients (26%)<br />

achieved a RECIST response. In univariate analysis, KSR2 rs11068551<br />

any G(AG/GG) genotype was significantly associated with shorter PFS<br />

compared to that <strong>of</strong> A/A carriers (median 3.5 vs. 7.4 months, HR�1.74<br />

[95%CI: 1-3.04] p�0.049, log-rank test). This result remained significant<br />

in a multivariate model (HR�2.13 [95%CI: 1.15-3.95], p�0.017, logrank<br />

test). None <strong>of</strong> the other tested SNPs were significantly associated with<br />

outcomes. Conclusions: Beyond KRAS and BRAF, some proteins including<br />

KSR are involved in modulating the level <strong>of</strong> activation <strong>of</strong> the EGFR pathway.<br />

Our results show that the functionally relevant KSR2 rs11068551 could<br />

affect PFS in advanced mCRC patients treated with cetuximab and<br />

irinotecan. Preclinical studies to confirm and further explain these preliminary<br />

findings are ongoing.<br />

3599 General Poster Session (Board #34D), Mon, 8:00 AM-12:00 PM<br />

The effect <strong>of</strong> deprivation on uptake and outcomes in a population-based<br />

FOBt colorectal cancer screening program. Presenting Author: David<br />

Mansouri, Department <strong>of</strong> Surgery, University <strong>of</strong> Glasgow, Glasgow, United<br />

Kingdom<br />

Background: Population-based FOBt colorectal cancer screening has been<br />

shown to reduce cancer specific mortality and is used across the UK.<br />

Despite evidence that socioeconomic deprivation is associated with increased<br />

incidence <strong>of</strong> colorectal cancer, uptake <strong>of</strong> screening may be lower in<br />

those who are more deprived. The aim <strong>of</strong> this study was to assess the impact<br />

<strong>of</strong> deprivation on the screening process. Methods: A prospectively maintained<br />

database, encompassing the first screening round in a single<br />

geographical area, was analysed with deprivation categories calculated<br />

from the Scottish Index <strong>of</strong> Multiple Deprivation 2009. Results: Overall,<br />

395,698 individuals were invited to screening, 204,812(52%) participated<br />

and 6,094(3%) tested positive. 32% <strong>of</strong> screened individuals were in<br />

the most deprived quintile. Of the positive tests, 5,457(95%) agreed to be<br />

pre-assessed for colonoscopy. 839(16%) did not proceed to colonoscopy<br />

following pre-assessment. Of the 4,618 that attended for colonoscopy,<br />

cancer was detected in 7%. Colonoscopy results were not recorded in<br />

1,035(22%) cases. Lower uptake <strong>of</strong> screening was seen in males, those<br />

that were younger and those who were more deprived (p�0.001). Higher<br />

levels <strong>of</strong> deprivation were also associated with not proceeding to colonoscopy<br />

following pre-assessment (p�0.001). Higher positivity rates were<br />

seen in males, those that were older and more deprived (p�0.001). Despite<br />

higher positivity rates in the more deprived individuals (4% most deprived<br />

vs 2% least deprived, p�0.001), the positive predictive value <strong>of</strong> detecting<br />

cancer in those attending for colonoscopy was lower in those who were more<br />

deprived (6% most deprived vs 8% least deprived, p�0.040). Conclusions:<br />

Socioeconomic deprivation has a significant effect throughout the FOBt<br />

screening process. Individuals who are more deprived are less likely to<br />

participate in screening, less likely to complete the screening process and<br />

less likely to have cancer identified as a result <strong>of</strong> a positive test. This study<br />

adds further weight to existing evidence that individuals who are more<br />

deprived are less likely to engage in population-based FOBt colorectal<br />

cancer screening. Novel strategies to improve this are required.<br />

Gastrointestinal (Colorectal) Cancer<br />

227s<br />

3598 General Poster Session (Board #34C), Mon, 8:00 AM-12:00 PM<br />

Correlation <strong>of</strong> specific cytokine pr<strong>of</strong>iles with high blood neutrophil-tolymphocyte<br />

ratio and outcome in metastatic colorectal cancer. Presenting<br />

Author: Zhi-Yu Chen, Fudan University Shanghai Cancer Center, Shanghai,<br />

China<br />

Background: High blood neutrophil-to-lymphocyte ratio (NLR) has been<br />

previously identified as a poor prognostic marker in patients with metastatic<br />

colorectal cancer (mCRC). However, the underlying pathophysiology associated<br />

with this marker is not well defined. We validated this biomarker and<br />

investigated the relationship between circulating cytokines and high NLR.<br />

Methods: NLRs were calculated retrospectively from the ratio <strong>of</strong> peripheral<br />

blood absolute neutrophil and lymphocyte counts, and segregated based on<br />

previously determined cut<strong>of</strong>f <strong>of</strong> �5 and �5. Four cohorts with a total <strong>of</strong><br />

805 CRC pts were evaluated to confirm the prognostic clinical significance<br />

<strong>of</strong> NLR. Plasma cytokine levels were evaluated in exploratory (n�39) and<br />

validation cohorts (n�166) <strong>of</strong> previously untreated mCRC patients using<br />

multiplex-bead assays, and correlated with NLR. Results: High NLR is<br />

present in 20% <strong>of</strong> CRC patients and is associated with poor prognosis,<br />

independent <strong>of</strong> known prognostic factors (Table). Expression <strong>of</strong> 6 cytokines<br />

(IL-6, IL-8, IL-2Ra, HGF, M-CSF and VEGF) correlated with NLR�5 in both<br />

the exploratory and validation cohort. In the validation cohort, an additional<br />

14 cytokines correlated with NLR�5, including high EREG,AREG, and<br />

TGF-�, and low TRAIL. By hierarchial clustering, these 20 cytokines fall<br />

into 3 overlapping major clusters: angiogenic cytokines, inflammatory<br />

cytokines, and epidermal growth factor ligands. In the validation cohort, pts<br />

with a composite cytokine score below the median have longer survival than<br />

patients with high scores (40.0 mo vs 20.0 mo, HR � 2.24; 95%<br />

CI�1.47–3.41; P�0.001). Conclusions: High peripheral blood neutrophil/<br />

lymphocyte ratio (NLR) is associated with poor outcomes independent <strong>of</strong><br />

other known prognostic features. NLR correlates with discrete sets <strong>of</strong><br />

cytokines which may reflect important biological features in poor prognosis<br />

mCRC.<br />

Median OS (months) 3-yr survival rate<br />

Cohort (number <strong>of</strong> pts) HR 95% CI P value NLR5 NLR5<br />

Untreated mCRC (166) 2.7 1.8-4.3 �0.001 34.2 15.3<br />

Phase � mCRC (167) 2.2 1.5-3.1 �0.001 9.0 3.7<br />

Hepatectomy mCRC (208) 1.8 1.1-2.9 0.01 13.3 (DFS) 9.5 (DFS) 38% 8%<br />

Stage II/ III CRC (274) 2.4 1.1-5.1 0.02 90% 75%<br />

3600 General Poster Session (Board #34E), Mon, 8:00 AM-12:00 PM<br />

HER2 status in locally advanced rectal cancer and metachronous metastases:<br />

Opportunity for a new therapeutic approach? Presenting Author:<br />

Lena-Christin Conradi, Department <strong>of</strong> General and Visceral Surgery, University<br />

Medical Center, Georg-August-University, Göttingen, Germany<br />

Background: Even though the implementation <strong>of</strong> multimodal treatment<br />

strategies including neoadjuvant radiochemotherapy (RCT) has led to<br />

improved survival distant metastases are still limiting the prognosis <strong>of</strong><br />

rectal cancer patients. In this context, we investigated the HER-2 status in<br />

rectal cancer patients, UICC stages II and III. Our aim was to assess the<br />

HER-2 positivity rate in primary tumors and metachronous metastases.<br />

Methods: In this study 264 rectal cancer patients (192 male, 72 female;<br />

median age 64 years) from phase-II/-III-trials <strong>of</strong> the German Rectal Cancer<br />

Study Group (CAO/ARO/AIO-94 and 04) were included. HER-2 status was<br />

determined pretherapeutically in tumor biopsies as well as resection<br />

specimens and metachronous metastases (n�27) using immunohistochemistry<br />

(IHC0 to IHC3�) scoring and S-ISH-amplification detection. Tumors<br />

with IHC3� or S-ISH ratio ?2.0 were classified as HER-2 positive; results<br />

were correlated with clinicopathological parameters and long-term survival.<br />

Results: A positive HER-2 status was found in 12.4% <strong>of</strong> pre-treatment<br />

biopsies, in 29.3% <strong>of</strong> the resection specimens and 22.2% (n�6) <strong>of</strong><br />

metastases. With a median follow-up <strong>of</strong> 46.5 months patients with<br />

HER-2-positivity showed better disease free survival (p�0.06) and cancerspecific<br />

survival (CSS, p�0.05). The 5-years survival rate was 96.4%<br />

(HER-2-positive) versus 79.5% (HER-2-negative). In multivariate analyses<br />

HER-2 status was as an independent (p�0.0053) predictor for CSS along<br />

with (y)pN-status (p�0.0001) and R-status (p�0.023). Conclusions:<br />

HER-2 amplification is detectable in a significant proportion (about 30%)<br />

<strong>of</strong> primary tumors <strong>of</strong> patients with advanced rectal cancer. Furthermore<br />

HER-2 amplification was detectable in 22% <strong>of</strong> resected metachronous<br />

metastases during follow-up. Therefore HER-2 represents a promising<br />

target and should be further assessed within prospective clinical trials.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


228s Gastrointestinal (Colorectal) Cancer<br />

3601 General Poster Session (Board #34F), Mon, 8:00 AM-12:00 PM<br />

Cost-effectiveness analysis per life-year gained based on predictors <strong>of</strong><br />

response for first-line metastatic colorectal cancer therapy in Spain.<br />

Presenting Author: Cristobal Belda-Iniesta, Centro Integral Oncológico<br />

Clara Campal (CIOCC), Madrid, Spain<br />

Background: Economic crisis, increasing prevalence and cost <strong>of</strong> mCRC<br />

management endanger availability <strong>of</strong> biological therapies for public health<br />

systems due to budget limitations. We performed a cost-effectiveness<br />

analysis based on markers <strong>of</strong> response/resistance including biological<br />

therapies available in Spain for mCRC. Methods: We aimed to calculate<br />

incremental cost-effectiveness ratio (ICER) per life-year gained (LYG) and<br />

progression-free year gained based on predictive markers for mCRC.<br />

Efficacy data include randomized trials (RT) that guided on-label uses <strong>of</strong><br />

bevacizumab and cetuximab. Control arms from these trials were used as<br />

reference to calculate ICERs. Markers <strong>of</strong> clinical benefit (biological and<br />

radiological) were included in this model. Toxicity as predictor <strong>of</strong> efficacy<br />

was excluded for any therapy. Prices for drugs in Spain were assumed to<br />

represent the best-value for each drug including all possibilities to reduce<br />

pharmacy costs. For 1st line, median duration <strong>of</strong> therapy reported by RT<br />

was used to calculate the final budget. 70kg and 1.7 m were used as<br />

reference for patients dose calculations. If accessible, HR for PFS and OS<br />

were used instead <strong>of</strong> medians. Results: K-Ras status and early response<br />

measured by CT at 8 weeks were used as predictors <strong>of</strong> resistance and<br />

increased efficacy for cetuximab-based combinations. We have not identified<br />

any predictor marker for other drugs from RT. In this regard,<br />

FOLFIRI�cetuximab combination obtained an ICER below the widelyproposed<br />

Spanish threshold <strong>of</strong> 30,000 € per LYG if patients harbored wt<br />

K-Ras tumors and evidenced an objective response at 8 weeks. Other ICERs<br />

for different schedules were too distant from this limit, they will be<br />

presented at the meeting. Multivariate analysis confirmed the robustness <strong>of</strong><br />

results. Conclusions: 1st line FOLFIRI�cetuximab therapy for wt K-Ras<br />

patients that get an objective response measured by CT at 8 weeks is the<br />

only cost-effective therapy option for mCRC below usual health-economic<br />

thresholds for Spain. Our results are critical to design cost-effectiveness<br />

based clinical guidelines for mCRC that will contribute to financial<br />

sustainability <strong>of</strong> public health system in Spain.<br />

3603 General Poster Session (Board #34H), Mon, 8:00 AM-12:00 PM<br />

Improved early prediction <strong>of</strong> individual prognosis for patients with mCRC:<br />

Joint modeling <strong>of</strong> tumor shrinkage with volume data for PFS and OS.<br />

Presenting Author: Ulrich Robert Mansmann, Institute <strong>of</strong> Medical Informatics,<br />

Biometry and Epidemiology, Klinikum Grosshadern, University <strong>of</strong><br />

Munich, Munich, Germany<br />

Background: Piessevaux et al. (ESMO 2010) proposed a decrease in<br />

RECIST-based tumor size <strong>of</strong> 20% at week 8 following 1st-line therapy for<br />

mCRC as a predictor <strong>of</strong> clinical outcome (PFS and OS). We expanded upon<br />

this idea (ASCO GI 2012). A tumor volume algorithm was developed<br />

providing a better approximation to true tumor volume by using both the<br />

longest and the longest orthogonal diameters <strong>of</strong> a target lesion. In this study<br />

we compare the quality <strong>of</strong> early prediction-based individual patient<br />

prognosis based on tumor change according to either RECIST or the tumor<br />

volume algorithm. Methods: The prognostic method is combined with the<br />

volume algorithm and applied to the data from 2 studies (OPUS, n�337;<br />

CRYSTAL, n�1198) and 4 treatment regimens (FOLFOX4 �/- cetuximab<br />

and FOLFIRI �/- cetuximab), in patients with mCRC. The influence <strong>of</strong> the<br />

treatment regimen on early PFS and OS prognosis is studied by joint<br />

modeling. The quality <strong>of</strong> early prognosis depending on the tumor size<br />

assessment used is compared by the logarithmic scoring rule. Results:<br />

Individual volume shrinkage and baseline volume are considered prognostic<br />

factors. Individual predictions depend on the chemotherapy administered<br />

and whether cetuximab is added. Equivalent tumor baseline volumes<br />

and early volume changes, predict an up to 20% higher 1 year (y) PFS and<br />

2y OS rate for FOLFIRI than for FOLFOX. The addition <strong>of</strong> cetuximab to<br />

standard chemotherapy (CT) translates into a mean increase in shrinkage <strong>of</strong><br />

10% resulting in an improvement in 1y PFS rates <strong>of</strong> 23% and in 2y OS<br />

rates <strong>of</strong> 18%. The quality <strong>of</strong> individual prediction over the 4 regimens is<br />

combined in one logarithmic (log) score. The log score for the RECISTbased<br />

prediction is 2.45 which is significantly higher than that for the<br />

volume-based prediction <strong>of</strong> 1.97 (p�0.0001). Lower scores correspond to<br />

a better prediction. Conclusions: The tumor volume algorithm enables a<br />

more precise prediction <strong>of</strong> individual patient PFS and OS than RECISTbased<br />

tumor assessments. Based on early tumor changes for CT, �/cetuximab,<br />

it is possible to calculate quantitative information on a patient’s<br />

prognosis. The model has high potential to guide individual clinical<br />

decision making for mCRC patients.<br />

3602 General Poster Session (Board #34G), Mon, 8:00 AM-12:00 PM<br />

Aflibercept versus placebo in combination with FOLFIRI in previously<br />

treated metastatic colorectal cancer (mCRC): Mean overall survival (OS)<br />

estimation from a phase III trial (VELOUR). Presenting Author: Florence<br />

Joulain, san<strong>of</strong>i, Chilly Mazarin, France<br />

Background: VELOUR evaluated the efficacy and safety <strong>of</strong> the novel fusion<br />

protein aflibercept (VEGF Trap) in combination with FOLFIRI in mCRC<br />

patients previously treated with oxaliplatin. Median OS showed significant<br />

improvement with 12.06 months in placebo arm vs 13.50 months in<br />

aflibercept arm (p�0.0032; HR�0.82 [95.34% CI: 0.71 to 0.94]) [Van<br />

Cutsem, 2011]. Since survival data in oncology are usually right skewed,<br />

median survival is preferred for regulatory purposes. However, mean<br />

survival estimation can render a more meaningful estimate for long term<br />

benefit <strong>of</strong> interventions, and be effectively applied to clinical and economic<br />

decision support. Estimating mean OS also allows payers to derive an<br />

estimation <strong>of</strong> total costs and outcomes in the population. The purpose <strong>of</strong><br />

this study was to estimate the mean OS for VELOUR trial. Methods: During<br />

the trial follow-up period, mean OS is not observable and can be estimated<br />

by extrapolating the trial Kaplan-Meier curve using a survival function.<br />

Several standard parametric distributions were tested: exponential, weibull,<br />

lognormal, loglogistic and gompertz. Akaike’s Information Criteria (AIC),<br />

Bayesian Information Criteria (BIC) and graphical method were used to<br />

evaluate the goodness <strong>of</strong> fit <strong>of</strong> the distributions. Models were run by<br />

treatment arm separately or combining the two arms and using treatment as<br />

covariate to control for variation. Results: Using AIC/BIC and graphical<br />

method, loglogistic function best fit the VELOUR data both with and<br />

without treatment as covariate. Weibull distribution is used for sensitivity<br />

analysis. Conclusions: Using loglogistic function, mean OS benefit for<br />

aflibercept in combination with FOLFIRI is at least 2.9 months (versus 1.4<br />

months difference in median survival). The results have important implications<br />

for clinical and economic decision support. Study NCT00561470<br />

was funded by san<strong>of</strong>i, in partnership with Regeneron.<br />

Mean OS (months)<br />

Placebo* Aflibercept* Difference<br />

Loglogistic<br />

By treatment separately 18.9 25.5 6.6<br />

With treatment as covariate 20.3 23.2 2.9<br />

Weibull<br />

By treatment separately 14.9 17.9 3.0<br />

With treatment as covariate 15.0 17.7 2.7<br />

* � FOLFIRI<br />

3604 General Poster Session (Board #35A), Mon, 8:00 AM-12:00 PM<br />

Integrin genetic variants and stage-specific tumor recurrence in patients<br />

with stage II and III colon cancer. Presenting Author: Pierre Oliver Bohanes,<br />

University <strong>of</strong> Southern California Norris Comprehensive Cancer Center, Los<br />

Angeles, CA<br />

Background: Integrins are key elements in cancer biology regulating tumor<br />

growth, angiogenesis and lymphangiogenesis through interactions <strong>of</strong> the<br />

tumor cells with the microenvironment. Recent evidence showing that<br />

integrins are critical in cancer dormancy suggests that their differential<br />

expression or activity may be responsible for tumor recurrence. Moving<br />

from the hypothesis that integrins could have different effects in stage II<br />

and III colon cancer, we tested as a primary endpoint whether a comprehensive<br />

panel <strong>of</strong> germline single nucleotide polymorphisms (SNPs) in integrin<br />

genes could predict stage-specific time to tumor recurrence (TTR) in stage<br />

II and III colon cancer patients. Methods: A total <strong>of</strong> 234 patients, 105<br />

high-risk stage II and 129 stage III, treated with 5-fluorouracil-based<br />

chemotherapy at the University <strong>of</strong> Southern California were included in this<br />

study. The median follow-up time was 4.4 years. Whole blood samples were<br />

analyzed for 22 germline SNPs in integrin genes using PCR-RFLP or direct<br />

DNA-sequencing. Results: In the multivariate analysis,stage II colon cancer<br />

patients with at least one G allele for ITGB3 rs4642 had higher risk <strong>of</strong><br />

recurrence (HR�4.027, 95%CI 1.556-10.421, p�0.004). This association<br />

was also significant in the combined stage II-III cohort (HR�1.975,<br />

HR 95%CI 1.194-3.269, p�0.008). The predominant role <strong>of</strong> ITGB3<br />

rs4642 in stage II diseases was confirmed using recursive partitioning,<br />

showing that ITGB3 rs4642 was the most important factor in stage II<br />

diseases. In contrast, in stage III diseases, both ITGB1 rs2298141<br />

(HR�1.909, 95%CI 1.054-3.459, p�0.033) and ITGA4 rs7562325<br />

(HR�0.227, 95%CI 0.064-0.804, p�0.022) were associated with TTR.<br />

The latter showed a significant interaction between stages (p�0.048).<br />

Conclusions: This study identifies germline polymorphisms in integrin<br />

genes as independent stage-specific prognostic markers for stage II and III<br />

colon cancer. These data strengthen the role <strong>of</strong> tumor dormancy in early<br />

colon cancer and may help to select subgroups <strong>of</strong> patients who may benefit<br />

from integrin-targeted treatments.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


3605 General Poster Session (Board #35B), Mon, 8:00 AM-12:00 PM<br />

Phase II study <strong>of</strong> preoperative radiation with concurrent capecitabine,<br />

oxaliplatin, and bevacizumab followed by surgery and postoperative 5-FU,<br />

leucovorin, oxaliplatin (FOLFOX), and bevacizumab in patients with locally<br />

advanced rectal cancer: A trial <strong>of</strong> the Eastern Cooperative Oncology Group<br />

(E3204). Presenting Author: Steven J. Cohen, Fox Chase Cancer Center,<br />

Philadelphia, PA<br />

Background: Oxaliplatin and bevacizumab potentiate the antitumor activity<br />

<strong>of</strong> fluoroyprimidines and radiotherapy in preclinical and clinical studies <strong>of</strong><br />

colorectal cancer. This phase II trial evaluated the efficacy <strong>of</strong> the preoperative<br />

combination <strong>of</strong> capecitabine, oxaliplatin, and bevacizumab with<br />

radiotherapy in rectal cancer. Methods: Eligibility: resectable T3/T4 adenocarcinoma<br />

<strong>of</strong> the rectum � 12 cm. from anal verge. Preoperative treatment<br />

was capecitabine (825 mg/m² bid M-F), oxaliplatin (50 mg/m² weekly),<br />

bevacizumab (5 mg/kg D1,15,29), and pelvic external beam radiotherapy<br />

(50.4 Gy/28 fractions). Surgery was performed within 6-8 weeks. Within<br />

12 weeks <strong>of</strong> surgery, patients received 12 cycles <strong>of</strong> FOLFOX � bevacizumab<br />

(5 mg/kg) Q2 weeks. Pathologic complete response (path CR) was<br />

the primary endpoint. Secondary endpoints included resection rate and<br />

toxicity. Results: Fifty-seven patients (pts) enrolled from 7/06 to 5/10 and<br />

54 were eligible. <strong>Clinical</strong> staging: T3/4 (50/4), NX0/1/2 (2/16/31/5). Most<br />

common grade 3/4 non-hematologic toxicities during treatment were rectal<br />

pain (9), fatigue (8), and diarrhea (7). Two patients died <strong>of</strong> aspiration. Nine<br />

pts had early postsurgical complications (9 wound infections, 6 dehiscence,<br />

1 abscess) and 23 had late surgical complications (23 non-healing<br />

wounds, 12 dehiscence, 5 bowel obstruction/ileus, 2 abscess). Pathologic<br />

staging: T0/1/2/3 (11/3/15/19), N0/1/2 (32/10/7). Nine pts had path CRs<br />

(17%, 90% CI:[9%-27%]. Surgery was LAR (38) and APR (11). Twentytwo<br />

pts did not receive postoperative chemotherapy (39%), 16 due to<br />

adverse events. Five local recurrences have been reported. RFS and OS<br />

data are immature. Conclusions: The addition <strong>of</strong> oxaliplatin and bevacizumab<br />

to capecitabine and radiotherapy for locally advanced rectal cancer<br />

resulted in downstaging for the majority <strong>of</strong> pts but did not improve the<br />

pathologic complete response rate compared to historical controls. The<br />

combination resulted in significant early and late postsurgical complications.<br />

3607 General Poster Session (Board #35D), Mon, 8:00 AM-12:00 PM<br />

Six months versus twelve months <strong>of</strong> capecitabine as adjuvant chemotherapy<br />

for stage III (Dukes’ C) colon cancer: Initial safety report for the<br />

open-label randomized phase III study (JFMC37-0801). Presenting Author:<br />

Katsuyuki Kunieda, Gifu Prefectural General Medical Center, Gifu,<br />

Japan<br />

Background: The standard treatment duration <strong>of</strong> adjuvant chemotherapy<br />

(CT) in patients (pts) with stage III colon cancer is 6 months. On the other<br />

hand, no clinical trial showed the optimal treatment duration <strong>of</strong> oral<br />

chemotherapeutic agents in adjuvant setting for colon cancer. Sargent et al<br />

have reported that 83% <strong>of</strong> recurrences in stage II and III pts have occurred<br />

within the first 3 years after surgery and peak was observed around one year<br />

after surgery. Therefore, to clarify the benefit <strong>of</strong> 12 months administration<br />

<strong>of</strong> Capecitabine, we designed randomized phase III trial for a comparison <strong>of</strong><br />

6 months treatment and 12 months treatment <strong>of</strong> capecitabine as adjuvant<br />

CT for stage III colon cancer. Methods: JFMC37 is a multicenter, randomized<br />

Phase III trial. Patients with fully resected Stage III colon or recto<br />

sigmoid cancer were eligible. Capecitabine was administered orally as<br />

tablets, 2,500 mg/m²/day for 14 days followed by a 7-days rest. Treatment<br />

is continued to 8 cycles (6 months) in arm A (A) or 16 cycles (12 months) in<br />

arm B (B). Patients were randomized 1:1 to A or B. Data size was estimated<br />

by disease free survival as primary endpoint. The statistical design is based<br />

on superiority hypothesis; 5-yrs DFS is 60% in arm A, 67% in arm B<br />

;unilateral ��0.05, 1-��0.8;and planed accrual is 1200 pts. Results:<br />

Between September 2008 to December 2009, 1304 patients were<br />

enrolled and then randomized. Both arms were well balanced for mean age:<br />

(A) 64.1, (B) 63.8; ECOG PS (%0/1): (A) 95.0/5.0, (B) 97.1/2.9;<br />

involvement <strong>of</strong> lymph nodes (%N0/N1/N2): (A) 77.1/19.9/3.1, (B) 76.6/<br />

19.7/3.7. Treatment completion rate for A and B were 68.2% and 43.4%.<br />

Incidences <strong>of</strong> serious adverse events (SAEs) over 1% were neutropenia: (A)<br />

2.6%, (B) 3.8%, diarrhea: (A) 2.9%, (B) 2.1%, loss <strong>of</strong> appetite: (A) 1.3%,<br />

(B) 1.0%, fatigue: (A) 1.8%, (B) 1.2%, hand-hoot syndrome: (A) 16.4%,<br />

(B) 22.1%. Conclusions: There were no obvious differences in SAEs<br />

between arm A and arm B. Although twelve months <strong>of</strong> capecitabine showed<br />

a tendency to increase G3/4 hand-foot syndrome, we concluded that<br />

incidence <strong>of</strong> SAEs were acceptable and comparable to previously report.<br />

Gastrointestinal (Colorectal) Cancer<br />

229s<br />

3606 General Poster Session (Board #35C), Mon, 8:00 AM-12:00 PM<br />

Prognostic significance <strong>of</strong> intermediate mucinous carcinoma in patients<br />

with microsatellite stable stage II or III colon cancer. Presenting Author:<br />

Gun Min Kim, Department <strong>of</strong> Internal Medicine, Yonsei University College<br />

<strong>of</strong> Medicine, Seoul, South Korea<br />

Background: Mucinous adenocarcinoma (MC) is a histological subtype <strong>of</strong><br />

colorectal cancer with �50% <strong>of</strong> mucinous component <strong>of</strong> tumor volume.<br />

According to WHO classification, tumor less than 50% <strong>of</strong> the lesion<br />

composed <strong>of</strong> mucin was considered as non-mucinous carcinoma (NMC).Mucinous<br />

histology has been shown to be an independent adverse prognostic<br />

factor in some studies, but not in others. However, prognostic implication<br />

<strong>of</strong> intermediate mucinous carcinoma (IMC) which had focal mucinous<br />

component �50% <strong>of</strong> tumor volume has not been studied yet. The aim <strong>of</strong><br />

this study is to explore prognostic impact <strong>of</strong> IMC in patients with resected<br />

stage II or III colon cancer. Methods: This study involved 917 patients who<br />

underwent curative resection for stage II or III colon cancer in Severance<br />

Hospital, from 2003 to 2009. The proportion <strong>of</strong> mucinous component was<br />

reviewed and re-classified correctly by pathologists. MC was defined tumor<br />

having �50% mucinous component. Tumor with mucinous component but<br />

less than 50% regarded as IMC and less than 10% mucin as NMC. Results:<br />

Among 917 patients, 63 (6.9%) and 60 (6.5%) were MC and IMC,<br />

respectively. Median follow-up duration was 50.2 months (0.9~109.7) and<br />

Stage 3 was 48.1% (441/917). The similarities between MC and IMC were<br />

found as right colon dominant (MC: IMC: NMC � 60%: 67%: 34%,<br />

p�.001) and higher incidence <strong>of</strong> MSI-high compared to NMC (MC: IMC:<br />

NMC � 27%: 30%: 11%, p�.001). In stage III, the disease-free survival<br />

(DFS) rate at 5 years was 44% for MC, 56% for IMC, and 74% for NMC<br />

(p�.001). Subgroup analysis according to MSI status showed that DFS had<br />

significant inverse correlation with proportions <strong>of</strong> mucinous components in<br />

MSI-low or MSS group, but patients with MSI-high tumor showed higher<br />

five-year DFS rate (�85%) regardless <strong>of</strong> mucinous component. For stage II<br />

colon cancer patients, both mucinous histology and MSI had little clinical<br />

significance. Conclusions: In patients with curative resection for stage II or<br />

III colon cancer, IMC had similar characteristics to MC, which were right<br />

colon dominant and high incidence <strong>of</strong> MSI-high tumor. The prognostic<br />

impact <strong>of</strong> IMC was intermediate risk between MC and NMC in stage III<br />

patients with MSI-low or MSS tumor, but not in MSI-high group.<br />

3608 General Poster Session (Board #35E), Mon, 8:00 AM-12:00 PM<br />

Effect <strong>of</strong> postoperative complications on adjuvant chemotherapy use in<br />

stage III colon cancer. Presenting Author: Ryan P. Merkow, Northwestern<br />

University, Feinberg School <strong>of</strong> Medicine, Chicago, IL<br />

Background: The National Quality Forum has endorsed the use <strong>of</strong> adjuvant<br />

chemotherapy in stage III colon cancer, yet a substantial treatment gap<br />

exists in the United States. Our objective was to evaluate the contribution<br />

<strong>of</strong> postoperative complications on the use <strong>of</strong> adjuvant therapy after<br />

colectomy for cancer. Methods: Patients from the National Surgical Quality<br />

Improvement Program and the National Cancer Data Base who underwent<br />

colon resection for cancer were linked (2006-2008). The association <strong>of</strong><br />

complications on adjuvant chemotherapy use was assessed using multivariable<br />

regression models. Results: From 140 hospitals, 2368 patients<br />

underwent resection for stage III colon adenocarcinoma. Overall, 36.8%<br />

(871/2,368) patients were not treated with adjuvant therapy, <strong>of</strong> which<br />

47.8% (416/871) had documented severe comorbidities or advanced age<br />

(�80) as the reason for no adjuvant therapy receipt. Of the remaining 455<br />

patients, 21.3% (97/455) had �1 serious complication that could account<br />

for adjuvant therapy omission. The remaining 41.1% (358/871) patients<br />

did not have a documented reason for not recieving adjuvant therapy.<br />

Complications associated with adjuvant therapy omission were abscess/<br />

anastomotic leak (OR 1.91, 95% CI 1.02-3.59), renal failure (OR 7.16,<br />

95% CI 1.92-26.79), prolonged ventilation (OR 7.92, 95% CI 2.97-<br />

21.13), re-intubation (OR 5.69, 95% CI 2.13-15.18), and pneumonia (OR<br />

4.05, 95% CI 2.07-7.90). Abscess/anastomotic leak was associated with a<br />

28-day delay in time to adjuvant chemotherapy (73 vs. 45 days, p�0.05).<br />

Superficial surgical site infection did not decrease adjuvant therapy receipt<br />

but delayed the time to its use (57 vs. 44 days, p�0.05). The occurrence <strong>of</strong><br />

postoperative sepsis was associated with a 15-day delay to adjuvant<br />

chemotherapy (60 vs. 45 days, p�0.05). Conclusions: Serious postoperative<br />

complications explained nearly one quarter <strong>of</strong> the adjuvant chemotherapy<br />

treatment gap among stage III colon cancer patients. Postoperative<br />

complications affect treatment utilization and should be considered when<br />

calculating adherence with the Stage III adjuvant therapy for colon cancer<br />

measure. Judging provider performance using quality metrics is challenging<br />

without clinical data.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


230s Gastrointestinal (Colorectal) Cancer<br />

3609 General Poster Session (Board #35F), Mon, 8:00 AM-12:00 PM<br />

Reappraisal <strong>of</strong> the risks and benefits <strong>of</strong> major liver resection in patients<br />

with initially unresectable colorectal liver metastases. Presenting Author:<br />

Francois Cauchy, HBP Surgery, Hôpital Beaujon, Clichy, France<br />

Background: Improvements in both surgical technique and efficacy <strong>of</strong><br />

chemotherapy have increased the rate <strong>of</strong> resection for patients with initially<br />

unresectable colorectal liver metastases (IU-CRLM). We aimed to evaluate<br />

the short and long-term outcomes <strong>of</strong> major hepatectomy for such patients.<br />

Methods: From 2000 to 2011, 257 patients underwent major hepatectomy<br />

for CRLM. Seventy-eight (30%) <strong>of</strong> these patients were considered IU and<br />

required portal vein occlusion and/or �12 cycles or change in induction<br />

chemotherapy regimen to achieve resectability. Results: IU patients had<br />

respectively more lesions (5.6 vs.3.6, p�0.001), more frequently bilobar<br />

(70% vs.50% p�0.008) and synchronous (83.3% vs.70%, p�0.027)<br />

than initially resectable (IR) patients. Post-operative mortality (12.8%<br />

vs.1.7%, p�0.001) and major complications (46.2% vs.22.3%,<br />

p�0.0001) were higher in IU patients. An associated metabolic syndrome<br />

(HR 5.2, CI 1.2-21.9, p�0.025), high grade sinusoidal lesions (HR 2.4, CI<br />

1.1-5.8, p�0.044) and the need for vascular reconstruction (HR 6.3, CI<br />

1.2-34.4, p�0.032) were significant risk factors for major morbidity in IU<br />

patients. Significantly fewer IU patients received adjuvant chemotherapy in<br />

case <strong>of</strong> major postoperative complications compared to IR patients (47%<br />

vs. 83%, p�0.001). Overall 5-year survival was significantly lower in IU<br />

than in IR patients (26% vs.55%, p�0.032) and all IU patients had tumor<br />

recurrence within 3 years. The absence <strong>of</strong> adjuvant chemotherapy and<br />

tumor size �5 cm were the only factors associated with poor survival in<br />

multivariate analysis for IU patients. Conclusions: IU-CRLM patients<br />

requiring major liver resection displayed higher morbidity and mortality<br />

rates than IR ones, therefore compromising both short and long-term<br />

outcomes. Multimodal strategy should be reassessed in the presence <strong>of</strong><br />

metabolic syndrome, sinusoidal lesions or major vascular involvement.<br />

3611 General Poster Session (Board #35H), Mon, 8:00 AM-12:00 PM<br />

<strong>Clinical</strong> utility <strong>of</strong> the preoperative Glasgow prognostic score in patients<br />

undergoing potentially curative resection for colorectal cancer. Presenting<br />

Author: Donald C. Mcmillan, Department <strong>of</strong> Surgery, University <strong>of</strong> Glasgow,<br />

Glasgow, United Kingdom<br />

Background: There is now good evidence that, in addition to TNM stage, the<br />

pre-operative combination <strong>of</strong> the standardised measurements <strong>of</strong> C-reactive<br />

protein and albumin, the Glasgow Prognostic Score (mGPS) provides<br />

valuable prognostic information. The aim <strong>of</strong> the present study was to<br />

examine the clinical application <strong>of</strong> the pre-operative mGPS in a large<br />

mature cohort <strong>of</strong> patients undergoing potentially curative resection for<br />

colorectal cancer. Methods: From a prospectively maintained database,<br />

consecutive patients (n� 797) with histologically proven colorectal cancer<br />

who were considered to have undergone potentially curative resection<br />

between January 1997 and December 2010 in a single surgical unit at the<br />

Royal Infirmary, Glasgow were included in the study. Results: Patients with<br />

an elevated mGPS were more likely to be older (p�0.001), female<br />

(p�0.05), have colonic tumours (�0.001), present as an emergency<br />

(p�0.001), had higher TNM stage (p�0.05) and more likely to die <strong>of</strong> their<br />

disease (p�0.01). The median follow-up was 66 months and using 3 year<br />

cancer-specific mortality as an endpoint, the area under the receiver<br />

operator curve was 0.652 (95% CI, 0.591–0.714; p�0.001) for the<br />

mGPS and 0.668 (95% CI, 0.610–0.727; p�0.001) for TNM stage. The<br />

cancer-specific survival, at 3 years, varied between 86% and 64%<br />

according to the mGPS and between 88% and 72% according to TNM<br />

stage. The cancer-specific survival, at 3 years, in patients with a mGPS 0<br />

was 91% and 81% for TNM stage II and III respectively. The cancerspecific<br />

survival, at 3 years, in patients with a mGPS 1 was 89% and 66%<br />

for TNM stage II and III respectively. The cancer-specific survival, at 3<br />

years, in patients with a mGPS 2 was 77% and 43% for TNM stage II and III<br />

respectively. Conclusions: The results <strong>of</strong> the present study show the clinical<br />

utility <strong>of</strong> the pre-operative mGPS in predicting cancer specific survival <strong>of</strong><br />

potentially curative surgery for colorectal cancer.<br />

3610 General Poster Session (Board #35G), Mon, 8:00 AM-12:00 PM<br />

A population-based study <strong>of</strong> the effect <strong>of</strong> FDG PET/CT in the management<br />

<strong>of</strong> liver-limited colorectal adenocarcinoma (CRC) metastases. Presenting<br />

Author: Maria Yi Ho, Division <strong>of</strong> Medical Oncology, British Columbia Cancer<br />

Agency, Vancouver, BC, Canada<br />

Background: PET/CT scans are publically funded in British Columbia for<br />

staging in liver limited metastatic CRC. However, past studies have been<br />

equivocal about the utility <strong>of</strong> PET/CT as some report as high as a 20-30%<br />

change in management while others report �10% change in management.<br />

Our primary objective was to assess the effect <strong>of</strong> the addition <strong>of</strong> PET/CT to<br />

CT scanning for the management <strong>of</strong> liver limited colorectal cancer.<br />

Methods: Patients who underwent PET/CT scan for de novo liver limited<br />

metastatic disease from 2005-2011 in the province <strong>of</strong> British Columbia<br />

were identified using the PET/CT database. Patients recently completed or<br />

currently on chemotherapy were excluded. We determined the concordance<br />

rates between CT and PET/CT scans with respect to the extra-hepatic<br />

disease, the number <strong>of</strong> lesions in the liver and the location <strong>of</strong> liver lesions.<br />

Results: 349 patients were identified. The most common indications for<br />

PET/CT scans after an initial CT scan were: detection <strong>of</strong> extrahepatic<br />

disease (77%), confirmation <strong>of</strong> the malignant nature <strong>of</strong> the liver lesions<br />

(8%) and the extent <strong>of</strong> extrahepatic disease (15%). PET/CT and CT were<br />

discordant in 39% <strong>of</strong> cases for the extent <strong>of</strong> metastatic disease. PET/CT<br />

revealed extrahepatic disease in 27% <strong>of</strong> the cases for which CT only<br />

detected liver limited disease. In contrast, 13% <strong>of</strong> patients were downstaged<br />

when CT liver lesions were demonstrated not to be FDG avid.<br />

Concordance <strong>of</strong> PET/CT and CT scans on the number and location <strong>of</strong> liver<br />

lesions was 52% and 85%, respectively. PET/CT revealed additional<br />

number <strong>of</strong> liver lesions and multilobar disease in 26% and 12% <strong>of</strong> cases,<br />

respectively. Furthermore, the median time between PET/CT and CT were<br />

64.3 days and 64.1 days for concordant and discordant cases (p�0.88).<br />

Conclusions: PET/CT scans provided additional information compared to CT<br />

scans which could have implications for surgical management. Our study<br />

supports the utility and public funding <strong>of</strong> PET/CT in addition to CT in<br />

patients with potentially surgically curable metastatic CRC involving the<br />

liver.<br />

3612 General Poster Session (Board #36A), Mon, 8:00 AM-12:00 PM<br />

Does multiple mutational analysis <strong>of</strong> the EGFR pathway have a prognostic<br />

role in advanced colorectal cancer (CRC)? Presenting Author: Luisa Foltran,<br />

Azienda Ospedaliero-Universitaria, Udine, Italy<br />

Background: BRAF mutation is widely recognised as a strong negative<br />

prognostic factor in advanced CRC, while the prognostic value <strong>of</strong> KRAS<br />

mutations in codons 12-13 remains controversial. Exploring mutations in<br />

other downstream components <strong>of</strong> the EGFR pathway may have an impact<br />

on survival. Methods: A consecutive cohort <strong>of</strong> 201 metastatic CRC patients<br />

treated with systemic chemotherapy were analysed for KRAS (12-13-61-<br />

146), BRAF, PIK3CA and NRAS genotypes by pyrosequencing on PyroMark-<br />

TMQ96 ID instrument (Qiagen, Germany) with commercially available kits<br />

Anti-EGFR MoAb response (Diatech Pharmacogenetics, Italy). Accurate<br />

microdissection guaranteed more than 70% <strong>of</strong> cancer cells for each<br />

sample. For the purpose <strong>of</strong> the survival analysis 4 categories were created:<br />

(1) KRAS mutated (codons 12-13 only); (2) BRAF mutated; (3) any <strong>of</strong><br />

KRAS codons 61-146, PIK3CA or NRAS mutations; (4) all-wild type.<br />

Log-rank and Cox proportional tests were applied for statistical analysis.<br />

Results: KRAS mutations were present in 96 (47.8%) patients: 86 (42.8%)<br />

were in codons 12-13. BRAF mutations were found in 11 (5.5%) samples<br />

while PIK3CA and NRAS in 33 (16.4%) and 7 (3.5%), respectively. All<br />

mutations were mutually exclusive except for 24 (11.9%) patients with<br />

concomitant KRAS/PIK3CA mutations. Median survivals for different<br />

categories are shown. Patients harbouring BRAF mutation had the worst<br />

outcome (p�0.0006). Mutations <strong>of</strong> any codon <strong>of</strong> KRAS (12-13-61-146)<br />

also negatively impacted on survival (p�0.026), while the all wild-type<br />

(KRAS/BRAF/PIK3CA/NRAS) patients had the longest survival (p�0.002).<br />

Conclusions: This study suggests the usefulness for early molecular pr<strong>of</strong>iling<br />

for advanced CRC patients. Mutational analysis <strong>of</strong> all EGFR pathway<br />

components may identify different prognostic subgroups. This information<br />

may drive treatment selection in clinical practice and stratification in<br />

clinical trials.<br />

Mutational status<br />

Median survival<br />

(months)<br />

HR for death<br />

(95% CI)<br />

BRAF mutation 5.1 4.5 (2.1-9.7)<br />

KRAS mutation 12-13 only 15.8 1.56 (1.1-2.3)<br />

Any <strong>of</strong> KRAS 61-146 /NRAS/ PIK3CA mutation 14.7 1.8 (1.1-3.3)<br />

All W-T (reference category) 22.2 1<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


3613 General Poster Session (Board #36B), Mon, 8:00 AM-12:00 PM<br />

Neoadjuvant treatment <strong>of</strong> colorectal liver metastases (CRLM) with drug<br />

eluting beads trans-arterial chemoembolization (DEBIRI-TACE): A multiinstitute<br />

phase II study in resectable metastases. Presenting Author:<br />

Robert Peter Jones, University <strong>of</strong> Liverpool, Liverpool, United Kingdom<br />

Background: Perioperative chemotherapy confers 3-year progression free<br />

survival advantage following resection <strong>of</strong> CRLM and good pathologic<br />

response is associated with improved overall survival. However, systemic<br />

neoadjuvant chemotherapy can increase postoperative morbidity and<br />

mortality. TACE using preloaded Irinotecan eluting beads gives sustained<br />

delivery <strong>of</strong> drug directly to tumor, thereby maximising response and<br />

reducing systemic exposure. This study examined the feasibility and safety<br />

<strong>of</strong> neoadjuvant DEBIRI-TACE before CRLM resection. Methods: Patients<br />

with resectable CRLM received single DEBIRI-TACE (up to 200mg) 1<br />

month pre-hepatectomy. Primary end-point: tumor resectability, secondary<br />

end-points: safety, radiologic response (RECIST) and pathologic tumor<br />

response. Results: TACE attempted in 49 patients, successful in 40.<br />

Reasons for failed TACE included consent withdrawal (n�2), bilobar<br />

disease (n�2), tumour involving gallbladder wall (n�1), suspected hepatoma<br />

(n�1), arterial access difficulty (n�2), contrast medium allergy<br />

(n�1). Post-TACE complications: 1 acute pancreatitis (3%); 4 postembolization<br />

syndromes (10%). Imaging at 4 weeks post-TACE (30<br />

patients): complete response 0/40 (0%); partial response 1/40 (3%);<br />

stable disease 19/40 (48%); ‘progressive’ disease 10/40 (25%). 40<br />

patients proceeded to surgery, 38 underwent hepatectomy (2 peritoneal<br />

disease, resectability rate 95%). 30-day post-operative mortality 5%<br />

(n�2), neither death TACE related (1 intraoperative pneumomediastinum,<br />

1 aspiration pneumonia). 63 lesions (median 2 per patient) targeted with<br />

TACE. Histology: no residual disease 17%; 1-49% residual disease 59%;<br />

�50% residual disease 22%; no response 2%. Conclusions: Resection<br />

after neoadjuvant DEBIRI-TACE for CRLM is feasible and safe. Single<br />

treatment with DEBIRI-TACE resulted in tumor pathologic response similar<br />

to that seen after protracted systemic chemotherapy.<br />

3615 General Poster Session (Board #36D), Mon, 8:00 AM-12:00 PM<br />

Association <strong>of</strong> HER2 and IGF1 germline polymorphisms with outcome in<br />

metastatic colorectal cancer (mCRC) patients (pts) treated with secondline<br />

irinotecan (IR) with or without cetuximab (CB): The EPIC experience.<br />

Presenting Author: Dongyun Yang, University <strong>of</strong> Southern California Norris<br />

Comprehensive Cancer Center, Los Angeles, CA<br />

Background: EPIC, a multinational phase III clinical trial with IR � CB vs IR<br />

alone in mCRC pts in the second-line setting after failure <strong>of</strong> FOLFOX<br />

demonstrated a benefit for IR�CB in progression-free survival (PFS) and<br />

response rate (RR). We evaluated 6 functional germline polymorphisms<br />

involved in the IGF1 and HER2 for their potential role as molecular<br />

predictors <strong>of</strong> clinical outcome in pts treated in the EPIC study. Methods:<br />

DNA was extracted from all available formalin-fixed paraffin-embedded<br />

tumor samples from the EPIC trial. Genotyping was performed using<br />

PCR-RFLP assays and 5’ -end [g-33P] ATP’ labeled PCR-protocols.<br />

Univariate analysis (Fisher’s exact test for RR; log-rank test for PFS and OS)<br />

was performed to examine associations between polymorphisms and<br />

clinical outcome. Multivariate analysis (Logistic regression or Cox regression<br />

model) was conducted to control baseline patient characteristics and<br />

treatment. Results: 186 pts with available samples were treated either with<br />

IR/CB (arm A, 84 pts) or IR alone (arm B, 102 pts). Median age was 59 yrs<br />

(range 34-85yrs) for arm A and 61 yrs (range 25-90 yrs) for arm B. In arm<br />

A, 11/84 pts (13%) showed CR or PR, whereas 73/84 (87%) pts had SD or<br />

PD. For arm B, 6/102 pts (6%) showed CR or PR, whereas 96/102 pts<br />

(94%) had SD or PD. Median PFS for arm A was 3.0 months (95%CI 2.4-<br />

4.1 months) vs 2.7 months (95%CI 2.2-2.9 months) for arm B; median OS<br />

was 9.3 months (95%CI 7.1-21.1 months) for arm A vs 12.3 months<br />

(95%CI 10.4- 17.9 months) for arm B. KRAS mutation status was not<br />

significantly associated with outcome in our patient cohort. We found that<br />

HER2 rs 1136201 was significantly associated with response (RR: AA<br />

6.5%, AG 12.5%, and GG 27.3%, Fisher’s exact test p�0.045). IGF1 rs<br />

2946834 was significantly associated with PFS in both univariate and<br />

multivariate analyses (median PFS was 2.8 months in patients with CC or<br />

CT vs 1.8 months in patients with TT; log-rank p�0.009; Wald test<br />

p�0.008). Conclusions: Our study suggests the prognostic value <strong>of</strong> polymorphisms<br />

in the IGF1 and HER2-pathway in mCRC pts treated with IR� CB.<br />

Prospective validation <strong>of</strong> these findings in clinical trials is warranted.<br />

Gastrointestinal (Colorectal) Cancer<br />

231s<br />

3614 General Poster Session (Board #36C), Mon, 8:00 AM-12:00 PM<br />

Efficacy and safety <strong>of</strong> bevacizumab in metastatic colorectal cancer (mCRC):<br />

Pooled analysis from randomized controlled trials (RCTs). Presenting<br />

Author: Niall Christopher Tebbutt, Austin Hospital, Heidelberg, Australia<br />

Background: Bevacizumab (BV) with chemotherapy (CT) is a standard<br />

treatment for mCRC. This analysis pooled individual patient data from the<br />

clinical databases <strong>of</strong> seven RCTs (phase II or III) <strong>of</strong> BV in 1st- or 2nd-line<br />

treatment to further define clinical outcomes, including within subgroups.<br />

Methods: Patient data were pooled from 1st-line (AVF2107, NO16966,<br />

ARTIST, AVF2192, AVF0780, AGITG MAX) and 2nd-line (ECOG E3200)<br />

trials. All analyses were based on the intent-to-treat population. Overall and<br />

progression-free survival estimates (OS, PFS) were calculated by Kaplan-<br />

Meier methods. To assess differences in time to response variables by<br />

treatment arm (CT vs BV � CT), stratified random (overall) and fixed<br />

(subgroup comparisons) models were used to estimate pooled hazard ratios<br />

(HRs) and 95% confidence intervals (CIs), with each study included as a<br />

stratum. Results: Of the 3,763 pooled patients (CT [n�1773]; BV � CT<br />

[n�1990]), 58.8% were male, 39.6% were �65 years, and 45.7% had an<br />

ECOG performance status �1. OS and PFS were statistically significantly<br />

increased in BV-treated patients vs control patients. Safety data in this<br />

pooled analysis were consistent with the pr<strong>of</strong>ile <strong>of</strong> BV from the individual<br />

studies. Conclusions: The addition <strong>of</strong> bevacizumab to CT resulted in<br />

statistically significant improvements in survival outcomes for mCRC<br />

patients in the overall analysis, with PFS benefit extending across subgroups<br />

defined by CT intensity, CT regimen, and KRAS status.<br />

Overall: BV � CT vs CT<br />

HR (95% CI) P value<br />

OS 0.80 (0.71�0.90) .0003<br />

PFS 0.57 (0.46�0.71) �.0001<br />

Subgroup comparisons: BV � CT vs CT<br />

HR (95% CI) for OS HR (95% CI) for PFS<br />

Monotherapy (n�751) 0.86 (0.72–1.02) 0.56 (0.48–0.67)<br />

Doublets (n�3,012) 0.82 (0.76–0.89) 0.70 (0.64–0.76)<br />

Irinotecan regimen (n�1,027) 0.71 (0.61–0.83) 0.55 (0.47–0.64)<br />

Oxaliplatin regimen (n�1,985) 0.87 (0.79–0.96) 0.77 (0.70–0.85)<br />

KRAS wild-type patients (n�364) 0.70 (0.54–0.91) 0.57 (0.45–0.72)<br />

KRAS mutant patients (n�166) 0.85 (0.60–1.22) 0.54 (0.38–0.76)<br />

3616 General Poster Session (Board #36E), Mon, 8:00 AM-12:00 PM<br />

Incidence and prognostic impact <strong>of</strong> KRAS and BRAF mutations in patients<br />

undergoing liver surgery for colorectal metastases. Presenting Author:<br />

Georgios Karagkounis, The Johns Hopkins University School <strong>of</strong> Medicine,<br />

Baltimore, MD<br />

Background: Molecular biomarkers <strong>of</strong>fer the potential for refining prognostic<br />

determinants in patients undergoing cancer surgery. Among patients with<br />

colorectal cancer metastases, KRAS and BRAF are important biomarkers,<br />

but their role in patients undergoing surgical therapy for liver metastases is<br />

unknown. In this study, the prevalence and prognostic significance <strong>of</strong><br />

KRAS and BRAF mutations were determined in patients undergoing<br />

surgical therapy <strong>of</strong> colorectal liver metastases (CRLM). Methods: KRAS and<br />

BRAF analysis was performed on 202 patients undergoing curative intent<br />

surgical therapy <strong>of</strong> CRLM between 2003 and 2008. Tumor samples were<br />

analyzed for somatic mutations using sequencing analysis (KRAS: codon<br />

12/13, BRAF: V600E). The frequency <strong>of</strong> mutations was ascertained and<br />

their impact on outcome determined relative to other clinicopathologic<br />

factors. Results: KRAS gene mutations were detected in 58/202 patients<br />

(29%) undergoing surgery for CRLM, comparable to that reported in<br />

non-surgical patient series. In contrast, mutation in the BRAF gene was<br />

identified in very low frequency in this surgical cohort, found in only 4/202<br />

(2%) patients. KRAS mutations were associated with worse long-term<br />

survival (HR�2.13, CI�1.25-3.65), as well as risk <strong>of</strong> recurrence (HR�1.89,<br />

CI�1.12-3.19). ). In addition, KRAS mutation was also associated with<br />

risk <strong>of</strong> recurrence following surgical therapy (HR�1.89, CI�1.12-<br />

3.19).While other clinicopathologic features, including tumor number,<br />

CEA and primary stage were also associated with survival, KRAS status<br />

remained independently predictive <strong>of</strong> outcome. The low incidence <strong>of</strong> BRAF<br />

mutation limited the ability to determine its prognostic impact. Conclusions:<br />

While KRAS mutations were found in approximately one third <strong>of</strong> patients,<br />

BRAF mutations were found in only 2% <strong>of</strong> patients undergoing surgery for<br />

CRLM. KRAS status was an independent predictor <strong>of</strong> overall and diseasefree<br />

survival. Molecular biomarkers such as KRAS may help to refine our<br />

prognostic assessment <strong>of</strong> patients undergoing surgical therapy for CRLM.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


232s Gastrointestinal (Colorectal) Cancer<br />

3617 General Poster Session (Board #36F), Mon, 8:00 AM-12:00 PM<br />

Relationship between genetic markers and quality <strong>of</strong> life (QOL) in stage III<br />

colon cancer (CC) patients (pts) prior to adjuvant treatment (N0147).<br />

Presenting Author: Jeff A. Sloan, Mayo Clinic, Rochester, MN<br />

Background: In metastatic colorectal cancer (CRC), previous studies suggested<br />

potential relationships between certain drug metabolizing (DM)<br />

genes (DPYD, ERCC2, GSTP1 and TYMS) and QOL prior to receipt <strong>of</strong><br />

chemotherapy (Sloan, Plenary Session ASCO 2004). The current study is<br />

the first to examine relationships between similar DM genes and QOL in<br />

stage III CC pts. Methods: 1,583 CC pts on a randomized adjuvant stage III,<br />

phase III trial received FOLFOX, FOLFIRI, or either �/- Cetuximab.<br />

Genomic DNA and baseline QOL data via linear analogue self assessment<br />

(LASA) scales with 4 QOL variables were obtained. 53 DM SNPs were<br />

selected for known functional effect within potential regulatory sites.<br />

Two-sample t-tests assessed differences in QOL between pts with variants<br />

and wild-type status. Differences <strong>of</strong> at least 10 points on a 0-100 point<br />

QOL score were considered clinically significant. Results: 14 relationships<br />

between DM genes and QOL were detected at the nominal p � 0.05 level;<br />

six <strong>of</strong> these were related to DPYD, ERCC2, GSTP1 and TYMS. Relationships<br />

were also observed at the 0.05 level with polymorphisms <strong>of</strong> genes DPYS,<br />

ERCC1, MGMT, MTHFR, OPRT and UGT1A1 based on unadjusted and<br />

adjusted association tests. GSTP1 I105V and OPRT 5’UTR 28 A�G<br />

markers differed on fatigue scores (p�0.04, 0.005, respectively). Variants<br />

on MTHFR R594Q were associated with higher mental acuity scores<br />

(p�0.02). These relationships remained after adjusting for age, gender,<br />

race, T stage, PS, disease site, lymph nodes involved, and grade. The<br />

robustness <strong>of</strong> the observed associations varied with the multiple comparison<br />

adjustment method. Conclusions: Similar to the earlier study <strong>of</strong> stage IV<br />

CRC, we observed a possible relationship in pts with Stage III CC with<br />

regard to DM genes and QOL, although these relationships were more<br />

modest than observed in Stage IV pts. These DM genes may be in linkage<br />

disequilibrium with other genetic variants that more directly affect behavior<br />

and QOL. Further genome wide association studies are needed to clarify<br />

these relationships.<br />

3619 General Poster Session (Board #36H), Mon, 8:00 AM-12:00 PM<br />

Comparison <strong>of</strong> incidence and patterns <strong>of</strong> recurrence in colon cancer (Cca)<br />

treated by sentinel lymph node (SLN) mapping (M) versus conventional surgery.<br />

Presenting Author: Mohammed Saifullah Shaik, Hurley Medical Center/<br />

Michigan State University, Flint, MI<br />

Background: Recurrence <strong>of</strong> Cca patients (pts) depends on the initial stage at<br />

diagnosis. SLNM upstages more pts than conventional surgery (Conv. Surg.) for<br />

nodal metastasis, this might have an impact on the incidence and pattern <strong>of</strong><br />

recurrence. Hence comparative study was undertaken for recurrence in Cca pts<br />

undergoing SLNM Vs Conv.Surg. Methods: Group (Gp) A pts underwent SLNM<br />

with 1% Lymphazurin. The first 1-4 blue nodes were marked as SLNs followed by<br />

standard oncologic resection. Gp B pts underwent Conv. Surg. Data was<br />

collected for, TNM staging, Recurrence sites and follow up treatment. Minimum<br />

follow up <strong>of</strong> all pts was 12 month, with mean follow up period 44 months for both<br />

Gps. Results: There were 357 pts in GpA and 466 pts in GpB. For GpA pts, SLNM<br />

was successful in 99.6% <strong>of</strong> pts with average (Avg.) SLN was 2.8/pts. Avg. no. <strong>of</strong><br />

LNs in GpA was 15.4/pts vs. 12.8/pt in GpB (p�0.001). For GpA pts, 46% was<br />

LN �ve vs 34% in GpB . Excluding T0, Tis and M1 pts at diagnosis, recurrence<br />

was calculated for 313 pts in GpA vs. 314 pts in GpB. An overall recurrence was<br />

7.9% in GpA vs 22.2% (p�0.001) in GpB with distant mets being most common<br />

in both Gps. Recurrence <strong>of</strong> LN �ve and LN –ve pts also were significantly lower<br />

in GpA vs GpB (Table). The pattern <strong>of</strong> recurrence between the two Gps is shown<br />

in the Table. Conclusions: SLNM upstages significantly more pts in Cca than<br />

conv. surg. Even though pattern <strong>of</strong> recurrence was similar in both Gps, better<br />

staging may have resulted in better treatment, reducing the recurrence in SLNM<br />

Gp. Hence SLNM may be beneficial for Cca surgery.<br />

Node �ve<br />

Node -ve<br />

Gp. A Gp. B<br />

Gp. A Gp. B<br />

(SLN) (Conv.)<br />

(SLN) (Conv.)<br />

T-Stage<br />

1A. Incidence <strong>of</strong> recurrence<br />

(# 112) (# 112) p°-value T-Stage (# 201) (# 202) p°-value<br />

T-1 0 1 T-1 0 8<br />

T-2 1 2 T-2 1 4<br />

T-3 16 30 T-3 4 14<br />

T-4 0 6 T-4 3 5<br />

Total * 17 39 0.008 Total * 8 31 0.0005<br />

(15%) (34.8%)<br />

(4%) (15.3%)<br />

1B. Patterns <strong>of</strong> recurrence. Gp. A Gp. B<br />

Local 1 (4%) 5 (7.1%)<br />

Regional (Reg) 4 (16%) 12 (17%)<br />

Distant 20 (80%) 53 (76%)<br />

Total number <strong>of</strong> recurrences Gp A Gp B<br />

Local 1 (3%) 5 (6%)<br />

Reg. lymph node and s<strong>of</strong>t tissue 5 (15%) 16 (20%)<br />

Liver 13 (35%) 31 (37.3%)<br />

Lung 11 (30%) 10 (12%)<br />

Bone 1 (3%) 4 (4.8%)<br />

Peritoneum 0 (0%) 2 (2.4%)<br />

* Pts with Tis/T0/M1 disease were excluded (Gp A�41; Gp B� 61); ° Chi-square test.<br />

3618 General Poster Session (Board #36G), Mon, 8:00 AM-12:00 PM<br />

Pharmacogenetic pr<strong>of</strong>iling for oxaliplatin-based adjuvant chemotherapy<br />

(CT) benefit in stage II-III colon cancer (CC) patients: A GEMCAD study.<br />

Presenting Author: Jaime Feliu, Department <strong>of</strong> Medical Oncology, La Paz<br />

University Hospital, Madrid, Spain<br />

Background: Identifying molecular biomarkers for tumour recurrence is<br />

critical in successfully selecting early-stage CC patients who are more likely<br />

to benefit from adjuvant CT. We tested whether single nucleotide polymorphisms<br />

(SNP) within genes involved in biological pathways <strong>of</strong> interest for<br />

CC progression and treatment resistance would predict the risk <strong>of</strong> recurrence<br />

in stage II-III CC patients treated with oxaliplatin and fluoropyrimidines-based<br />

adjuvant CT. Methods: Genomic DNA was extracted from<br />

formalin-fixed paraffin-embedded samples from 202 surgically treated<br />

high-risk stage II (29.7%) and stage III (70.29%) CC patients receiving<br />

adjuvant CT (25.24% FOLFOX, 74.75% XELOX) from January 2004 to<br />

December 2008. Minimum follow-up was 36 months. Genotyping was<br />

performed for 62 SNPs in 31 genes using the MassARRAY (SEQUENOM)<br />

technology. Our results were validated in an independent cohort <strong>of</strong> 177<br />

stage II-III CC. Results: After a median follow-up <strong>of</strong> 51.4 months (7-96), 63<br />

patients (31.18%) had experienced a relapse and 31 (15.34%) had died.<br />

Three-year disease-free survival (DFS) and overall survival (OS) were<br />

72.2% (�/-0.032) and 89.1% (�/-0.022), respectively. Multivariate<br />

analysis showed that the risk <strong>of</strong> recurrence for patients with the E-selectine<br />

rs3917412 G�A G/G genotype was higher than for those with any A allele<br />

genotype [relative risk (RR): 2.02, 95% confidence interval (CI): 1.09-<br />

3.73, p�0.024]. In haplotype analysis, patients harboring the E-selectine<br />

rs3917412 G�A G/G and methylentetrahydr<strong>of</strong>olate reductase (MTHFR)<br />

rs1801133 C�T T/T haplotype had a higher risk <strong>of</strong> developing tumor<br />

recurrence compared to the E-selectine rs3917412 G�A any A and/or<br />

MTHFR rs1801133 C�T any C haplotype (RR: 3.39, 95% CI, 1.51-7.62,<br />

p�0.003). The ability to predict recurrence <strong>of</strong> this combined analysis was<br />

independently validated in the second cohort (RR: 3.75, 95% CI, 1.32-<br />

10.68, p�0.013). Conclusions: E-selectine and MTHFR SNPs were found<br />

to be independent prognostic markers for stage II-III CC patients, suggesting<br />

that this combined analysis provides additional prognostic information<br />

to that <strong>of</strong>fered by clinicopathologic variables.<br />

3620 General Poster Session (Board #37A), Mon, 8:00 AM-12:00 PM<br />

Effect <strong>of</strong> perioperative complications on recurrence and survival after<br />

resection <strong>of</strong> hepatic colorectal metastases: Analysis <strong>of</strong> data from a<br />

randomized controlled trial. Presenting Author: Camilo Correa-Gallego,<br />

Memorial Sloan-Kettering Cancer Center, New York, NY<br />

Background: Perioperative outcomes such as blood loss, transfusions and<br />

morbidity have been associated with cancer specific survival, but this is<br />

unsupported by prospective data. Methods: Patients were derived from a<br />

previously reported prospective randomized trial <strong>of</strong> acute normovolemic<br />

hemodilution (ANH) versus standard management during major hepatectomy.<br />

Patients with metastatic colorectal cancer (mCRC) were selected for<br />

analysis. Chemotherapy and survival data were obtained from the medical<br />

record. Disease extent was measured using a clinical risk scoring (CRS)<br />

system. Log-rank test and Cox proportional hazard model were used to<br />

evaluate recurrence free (RFS) and overall survival (OS). Results: This trial<br />

enrolled 130 patients, 90 <strong>of</strong> whom had mCRC. Median follow up was 54 m;<br />

median age was 53 yr; 56% were men. The CRS was �3 in 55% <strong>of</strong><br />

patients, and 57% had additional extrahepatic procedures. Morbidity and<br />

mortality were 33% and 3%. Chemotherapy was split before and after<br />

surgery in 70%; 10% received only preoperative and 19% only postoperative<br />

treatment, which was initiated after a median time <strong>of</strong> 6 wks (IQR �<br />

4-7). RFS and OS were 29 and 59 m. On multivariate analysis, (�)<br />

resection margin, high CRS and perioperative complications predicted<br />

shorter RFS and OS. Delayed initiation <strong>of</strong> postoperative chemotherapy (� 8<br />

wks) while most common in patients with complications (37 vs 12%; p:<br />

0.01), was not a significant predictor <strong>of</strong> shorter RFS or OS. Randomization<br />

(ANH vs standard) was also not significant in this regard. (See Table.)<br />

Conclusions: In this prospective randomized cohort, perioperative morbidity<br />

was a highly significant independent predictor <strong>of</strong> cancer specific outcome,<br />

associated with but not entirely explained by delayed initiation <strong>of</strong> chemotherapy.<br />

Variables affecting overall (OS) and recurrence free (RFS) survival.<br />

Median survival<br />

Multivariate<br />

months Univariate P P-HR<br />

CRS (high vs low)<br />

OS 32 vs 74 0.02 �0.01 – 3<br />

RFS 21 vs 46 0.03 �0.01 –3<br />

Margin (� vs -)<br />

OS 34 vs 71 0.05 0.03 – 0.5<br />

RFS 29 vs 32 0.2 0.04 – 0.5<br />

Complications (Y vs N)<br />

OS 27 vs 75 �0.01 �0.01 – 3.4<br />

RFS 18 vs 60 �0.01 �0.01 – 3.5<br />

Delayed chemotherapy (Y vs N)<br />

OS 30 vs 75 0.06 0.06 – 3<br />

RFS 29 vs 59 0.1 0.3<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


3621 General Poster Session (Board #37B), Mon, 8:00 AM-12:00 PM<br />

Colorectal cancer characteristics and outcomes in patients less than age<br />

50: A comparison <strong>of</strong> an urban university hospital with the NCI SEER<br />

database. Presenting Author: Edith P. Mitchell, Kimmel Cancer Center at<br />

Thomas Jefferson University, Philadelphia, PA<br />

Background: Cancer <strong>of</strong> the colon and rectum is the third most commonly<br />

occurring cancer, as well as the third leading cause <strong>of</strong> cancer deaths in<br />

<strong>American</strong> men and women. Colorectal cancer in younger patients is<br />

believed to have worse pathological features and prognosis than in older<br />

patients. The objective <strong>of</strong> this study was to assess pathological features and<br />

outcomes <strong>of</strong> CRC in patients less than age 50 using an institutional sample<br />

and comparing to the Surveillance, Epidemiology and End Results (SEER)<br />

database. Methods: Included in the study were a total <strong>of</strong> 4595 cases from<br />

the Tumor Registry at Thomas Jefferson University Hospital (TJUH) over a<br />

twenty year period from 1988 through 2007 and 290,338 cases from the<br />

Surveillance, Epidemiology and End Results (SEER) database from 1988<br />

through 2004. Patients less than age 50 were compared to those age 50<br />

and older. Results: Patients under age 50 with CRC presented with more<br />

advanced stage tumors in both data sets (�0.0001) , and had more poorly<br />

differentiated tumors than older patients (PTJUH�0.02754; PSEER�0.0001). Patients under 50 also had more mucinous/signet ring<br />

cell tumors with 12 percent to 8.1 percent in the TJUH data (p�0.002916)<br />

and 13.2 percent to 10.3 percent in the SEER data (p�0.0001), with<br />

younger males having the highest prevalence in both data sets. Younger<br />

patients had fewer proximal tumors than patients 50 and over, and a higher<br />

proportion <strong>of</strong> rectal tumors (p�0.001). Patients under age 50 were more<br />

likely to have positive nodes at all stages (PSEER �0.0001) relative to 50<br />

and over, as well as more likely to develop peritoneal metastases<br />

(PTJUH�0.3507),, but less likely to have lung metastases PTJUH�0.05249) than older pts. Despite their poor pathologic features, patients under age<br />

50 had better than or equal survival to those 50 and older. Conclusions:<br />

Colorectal cancer patients under age 50 presented with worse histological<br />

characteristics and metastasized much sooner, yet the younger patients<br />

had better than or equal survival to those ages 50 and older. Ongoing<br />

studies will assess differences in treatment and molecular features between<br />

younger and older colorectal cancer patients.<br />

3623 General Poster Session (Board #37D), Mon, 8:00 AM-12:00 PM<br />

Clinicopathologic and molecular associations <strong>of</strong> PIK3CA and PTEN mutation<br />

in colorectal cancer. Presenting Author: Fiona Lee Day, Ludwig<br />

Institute for Cancer Research, Parkville, Melbourne, Australia<br />

Background: The phosphatidylinositol 3-kinase (PI3K) signaling pathway<br />

has been implicated in the development <strong>of</strong> colorectal cancer (CRC),<br />

however a systematic analysis <strong>of</strong> pathway members and their association<br />

with clinical, pathological and other molecular features has been lacking.<br />

Methods: We determined the prevalence <strong>of</strong> PIK3CA (exons 9, 20) mutations<br />

in a large cohort (n�1092) <strong>of</strong> community-based patients (pts) with stage<br />

I-IV CRCs. A subset (n�744, 68%) were also tested for mutation in PTEN<br />

(exons 3,4,5,6,7,8), PTEN LOH and CIMP. All tumors were characterised<br />

for MSI, KRAS and BRAF mutations, and annotated for clinico-pathologic<br />

features and patient outcomes in a prospectively-recorded database.<br />

Detected novel PIK3CA exon-specific correlates were further explored in a<br />

combined analysis integrating the data <strong>of</strong> other published cohorts (total<br />

n�3677). Results: PIK3CA mutations occurred in 11.6% <strong>of</strong> primary<br />

tumors and were associated with older pt age (p�0.009), proximal site<br />

(p�0.001), lower lymphovascular invasion (p�0.018) and KRAS mutation<br />

(p�0.001), but not with gender, tumor stage or differentiation. Direct<br />

comparison <strong>of</strong> PIK3CA exon 9 and 20 mutations in the combined analysis<br />

(mixed effect logistic regression) finds both at increased frequency in<br />

KRAS-mutant tumors (p�0.098 for difference); however PIK3CA exon 20<br />

mutations uniquely correlated with BRAF mutation (p�0.002 for difference),<br />

MSI (p�0.001) and CIMP-high status (p�0.031). PTEN mutations<br />

were present in 5.8% <strong>of</strong> tumors and associated with proximal site<br />

(p�0.013), mucinous histology (p�0.013), MSI (p�0.001), CIMP-high<br />

(p�0.001), and BRAF mutation (p�0.001). There was a trend toward<br />

mutation <strong>of</strong> both PIK3CA and PTEN and strong selection for two hits to<br />

PTEN (mut/mut or mut/LOH, p�0.001). In our cohort neither PIK3CA or<br />

PTEN mutation were associated with pt survival outcomes. Conclusions: In<br />

CRC, common mutations in the PI3K signaling pathway are universally<br />

associated with proximal site, however display unique gene- and exonspecific<br />

associations with tumor histology, MSI, CIMP, KRAS and BRAF<br />

mutation. These need to be considered when exploring targeted therapeutic<br />

options for pts with CRC.<br />

Gastrointestinal (Colorectal) Cancer<br />

233s<br />

3622 General Poster Session (Board #37C), Mon, 8:00 AM-12:00 PM<br />

Novel colon cancer tumor suppressor gene, �-defensin 1, to predict<br />

recurrence in patients with stage II and III colon cancer. Presenting Author:<br />

Melissa Janae Labonte, University <strong>of</strong> Southern California Norris Comprehensive<br />

Cancer Center, Los Angeles, CA<br />

Background: Human �-defensin 1 (hBD-1) encoded by the DEFB1 gene is<br />

an antimicrobial peptide involved in the innate immune response and is<br />

expressed in epithelial cells, including the colon. hBD-1 has been shown to<br />

have tumor suppressor functions in urothelial cancer models. We tested<br />

whether 4 germline single nucleotide polymorphisms (SNPs) in DEFB1<br />

could predict time to tumor recurrence (TTR) in stage II and III colon<br />

cancer (CC) patients. We then sought to demonstrate if hBD-1 has tumor<br />

suppressor functions in CC models. Methods: A total <strong>of</strong> 234 patients, 105<br />

stage II and 129 stage III, treated with 5-FU-based chemotherapy at the<br />

University <strong>of</strong> Southern California were included. The median follow-up time<br />

was 4.4 yrs. SNPs were analyzed from whole blood samples using direct<br />

DNA-sequencing. To gain insight into hBD-1’s function, hBD-1 expression<br />

in CC cell lines was determined by qRT-PCR and Western blotting. hBD-1<br />

expression was induced ectopically and CC cells and viability, membrane<br />

permeability, cell-cycle and apoptosis analyzed. Results: Stage III patients<br />

with rs1800972 DEFB1 -44G containing genotypes had longer TTR than<br />

patients with C/C genotype (11.3 mo vs 2.7 mo; p�.008). In contrast in<br />

stage II, patients with rs1799946 DEFB1 -52A containing genotypes, had<br />

significantly longer TTR than patients with G/G genotype (16.8 mo vs 5.9<br />

mo; p�.017). In the multivariate analysis, both DEFB1 -44C/G and DEFB1<br />

-52G/A SNPs remained significant, respectively in stage III (HR�.419;<br />

95%CI .201-.87; p�.020) and in stage II (HR�.360; 95%CI .152-.853;<br />

p�.020). Haplotype <strong>of</strong> all four SNPs was significantly associated with<br />

stage-specific TTR (to be presented at the meeting). In CC cell line models,<br />

there was a loss <strong>of</strong> hBD-1 expression, and induction <strong>of</strong> hBD-1 expression<br />

resulted in a loss <strong>of</strong> cell viability, membrane permeability and the induction<br />

<strong>of</strong> apoptosis. Conclusions: The results demonstrate for the first time<br />

evidence <strong>of</strong> hBD-1’s potential influence on the microenvironment and its<br />

role as a tumor suppressor in CC. Further studies need to be conducted to<br />

better understand the role <strong>of</strong> hBD-1 in CC development and progression<br />

and evaluate the potential utility <strong>of</strong> hBD-1 as a therapeutic strategy.<br />

3624 General Poster Session (Board #37E), Mon, 8:00 AM-12:00 PM<br />

Methylations <strong>of</strong> NEUROG1, p16, and MLH1 and recurrence following<br />

adjuvant FOLFOX in colorectal cancer. Presenting Author: Hyun-Jung Lee,<br />

Department <strong>of</strong> Internal Medicine, Seoul National University Hospital,<br />

Seoul, South Korea<br />

Background: CpG island methylator phenotype (CIMP) is characterized by<br />

concurrent methylation <strong>of</strong> multiple CpG islands in tumor DNA, which can<br />

inactivate tumor suppressor genes or promote carcinogenesis. The prognostic<br />

impact <strong>of</strong> CIMP on treatment outcome <strong>of</strong> colon cancer patients receiving<br />

adjuvant 5-fluorouracil/leucovorin/oxaliplatin (FOLFOX) is unclear. We<br />

investigated CIMP markers in colorectal cancer patients treated with<br />

adjuvant FOLFOX. Methods: Sporadic colorectal cancer patients treated<br />

with curative resection followed by adjuvant FOLFOX were included. DNA<br />

was extracted from formalin-fixed paraffin-embedded surgical specimen. 8<br />

CpG island loci (CACNA1G, CRABP1, IGF2, MLH1, NEUROG1, P16,<br />

RUNX3 and SOCS1) were examined using MethyLight analysis. Diseasefree<br />

survival (DFS) was evaluated according to each methylation loci.<br />

Results: A total <strong>of</strong> 322 patients were included. Methylation at 1 or more loci<br />

was observed in 150 patients (46.6%) and 6 or more loci in 15 (4.7%).<br />

During a median follow-up duration <strong>of</strong> 39.7 months, 55 recurrences were<br />

observed. Three year DFS in the patient cohort was 84%. CRABP1 (23.9%)<br />

was the most frequently methylated loci, followed by p16 (22.7%) and<br />

NEUROG1 (20.8%). Patients having methylation at NEUROG1 (3 year<br />

DFS 78% in (�) vs. 86% in (-), p � 0.014) and p16 (3 year DFS 78% in<br />

(�) vs. 86% in (-), p � 0.12) had worse DFS, whereas methylation at MLH1<br />

had better DFS (3 year DFS 100% in (�) vs. 86% in (-), p � 0.19). In a<br />

combined analysis, patients with MLH1(-)/NEUROG1(�)/p16(�) had<br />

worst treatment outcome compared to MLH1(-)/NEUROG1(�) or p16(�),<br />

MLH1(-)/ NEUROG1(-) /p16(-), and MLH1(�) (3 year DFS 62%, 82%,<br />

87%, and 100%, respectively; p � 0.002). In multivariate analysis,<br />

NEUROG1(�)/p16(�) was associated with significantly higher recurrence<br />

compared with other patients (adjusted hazard ratio (HR) 2.15 (95%<br />

confidence interval (CI) 1.08 - 4.27, p � 0.029). Conclusions: Methylation<br />

status <strong>of</strong> NEUROG1, p16, and MLH1 is associated with recurrence<br />

following adjuvant FOLFOX in stage II/III colorectal cancer. Further<br />

validation and translational studies to improve treatment outcome in the<br />

subset <strong>of</strong> patients are warranted in the future.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


234s Gastrointestinal (Colorectal) Cancer<br />

3625 General Poster Session (Board #37F), Mon, 8:00 AM-12:00 PM<br />

Impact <strong>of</strong> “poorly differentiated clusters” in primary lesion as a novel<br />

prognostic indicator in colorectal liver metastasis. Presenting Author:<br />

Tsuyoshi Konishi, Gastroenterology Center, Cancer Institute Hospital,<br />

Tokyo, Japan<br />

Background: Preoperative predictors for survival are needed in colorectal<br />

liver metastasis in order to select poor-risk group that truly requires<br />

perioperative chemotherapy. This study aimed to elucidate survival predictors<br />

in patients undergoing curative hepatectomy for colorectal liver<br />

metastasis, particularly focusing on the impact <strong>of</strong> poorly differentiated<br />

clusters (PDC); a novel histologic grading system in primary lesion.<br />

Methods: A total <strong>of</strong> 424 consecutive patients undergoing curative resection<br />

<strong>of</strong> both primary colorectal cancers and metastatic liver lesions at two<br />

referral centers were enrolled in the study. Determinants <strong>of</strong> overall survival<br />

(OS) and recurrence free survival (RFS) after hepatectomy were investigated<br />

by univariate and multivariate analysis, using detailed clinicopathological<br />

parameters in primary and metastatic lesions. Cancer clusters <strong>of</strong> �5<br />

cancer cells and lacking a gland-like structure were counted under a x20<br />

objective lens in a field containing the highest number <strong>of</strong> clusters at<br />

invasive front <strong>of</strong> primary lesions, and tumors with �5,5to9,and�10<br />

clusters were classified as PDC grade (G)1, G2 and G3, respectively (n�65,<br />

132, and 227 tumors, respectively). Results: OS and RFS at 3 years were<br />

59% and 27%, respectively, with average follow up period <strong>of</strong> 43 months.<br />

PDC grade in primary lesion was strongly associated with both 3-year-OS<br />

(83%, 62%, and 51%, respectively, p�0.0001) and 3-year-RFS (55%,<br />

30%, and 17%, respectively, p�0.0001). Multivariate analysis revealed<br />

that PDC grade in primary lesion was the most potent prognostic factor after<br />

hepatectomy independent <strong>of</strong> T and N <strong>of</strong> primary lesion and size <strong>of</strong> liver<br />

metastasis. Conclusions: PDC grade in primary lesion is a novel potent<br />

prognostic indicator in colorectal liver metastasis, which is independent <strong>of</strong><br />

T and N. It is noteworthy that PDC grade can bias the survival in clinical<br />

studies targeting perioperative chemotherapy in colorectal liver metastasis.<br />

3627 General Poster Session (Board #37H), Mon, 8:00 AM-12:00 PM<br />

Effect <strong>of</strong> BSA dosing on 5-FU exposure among colorectal cancer patients<br />

depending on their gender and age. Presenting Author: Abebe Haregewoin,<br />

Myriad Genetics, Inc., Salt Lake City, UT<br />

Background: 5-fluorouracil (5-FU) remains the cornerstone <strong>of</strong> colorectal<br />

cancer (CRC) therapy and its dosing is based on body surface area (BSA).<br />

Although convenient, BSA dosing disregards factors that impact 5-FU PK<br />

such as genotype, age, gender, etc. Thus, patients receiving the same BSA<br />

based dose show a wide range <strong>of</strong> 5-FU plasma levels (measured as AUC in<br />

mg · h/L), resulting in ineffective or toxic doses affecting therapy outcome.<br />

An optimal therapeutic AUC target range <strong>of</strong> 20-30 mg · h/L has been<br />

proposed. Methods: Plasma samples from the initial treatment cycle <strong>of</strong> 927<br />

CRC patients who received 2,400 mg/m2 continuous infusion <strong>of</strong> 5-FU over<br />

44-48 hours, with or without bevacizumab, were tested for 5-FU exposure<br />

using an immune-based assay (OnDose). AUC results were analyzed to<br />

evaluate gender and age effects. Results: There are 391 (42.2%) female<br />

and 536 (57.8%) male patients with age ranging from 17-93 years<br />

(mean/SD: 58.5/11.8). See table. These analyses indicate that in comparison<br />

to the proposed optimal AUC target range <strong>of</strong> 20-30 mg · h/L, females<br />

receive supra-optimal doses compared to males (p�0.0083). Age also<br />

affects the 5-FU AUC (p�0.0002), with younger patients more likely to<br />

receive suboptimal doses than older patients. Conclusions: BSA dosing is<br />

an unreliable indicator <strong>of</strong> exposure and may lead to under dosing in males<br />

and younger patients, and overdosing in females and older patients,<br />

potentially resulting in corresponding diminished efficacy or increased<br />

toxicity. Data suggest that therapy could be optimizedby 5-FU dose<br />

adjustment in subsequent cycles based on PK monitoring so that AUC<br />

values will converge towards the target range irrespective <strong>of</strong> gender or age.<br />

A large clinical trial is in progress to validate these findings.<br />

Patient characteristic<br />

Below<br />

(8 - 19 mg · h/L)<br />

AUC target range<br />

Within<br />

(20 - 30 mg · h/L)<br />

Above<br />

(31 - 60 mg · h/L)<br />

Gender<br />

Female 169 (43.2%) 165 (42.2%) 57 (14.6%)<br />

Male 272 (50.7%) 210 (39.2%) 54 (10.1%)<br />

Age (years)<br />

< 45 68 (56.2%) 43 (35.5%) 10 (8.3%)<br />

46-50 60 (55.1%) 37 (33.9%) 12 (11%)<br />

51-55 79 (57.7%) 45 (32.8%) 13 (9.5%)<br />

56-60 66 (46.8%) 58 (41.1%) 17 (12.1%)<br />

61-65 74 (47.1%) 69 (44%) 14 (8.9%)<br />

66-70 41 (35.6%) 53 (46.1%) 21 (18.3%)<br />

> 70 53 (36.1%) 70 (47.6%) 24 (16.3%)<br />

3626 General Poster Session (Board #37G), Mon, 8:00 AM-12:00 PM<br />

Effect <strong>of</strong> the 12-gene colon cancer assay results on treatment recommendations<br />

in patients with stage II colon cancer. Presenting Author: Thomas H.<br />

Cartwright, Ocala Oncology, Ocala, FL<br />

Background: The Oncotype DX Colon Cancer Recurrence Score (RS) has<br />

been clinically validated as an independent predictor <strong>of</strong> individual recurrence<br />

risk in stage II colon cancer patients following surgery. As a result,<br />

physicians have been ordering the Oncotype DX assay for stage II colon<br />

cancer patients since January 2010, yet no data exist on the assay’s impact<br />

on adjuvant treatment planning in clinical practice. We performed a survey<br />

to characterize the impact <strong>of</strong> the assay on adjuvant treatment recommendations<br />

in stage II colon cancer. Methods: U.S. medical oncologists (n�346)<br />

who ordered Oncotype DX for �3 patients with stage II colon cancer were<br />

contacted and asked to complete a web-based survey regarding the single<br />

most recent stage II colon cancer patient for whom the assay was ordered.<br />

The survey was developed through cognitive interviews with four medical<br />

oncologists, and the protocol was institutional review board approved.<br />

Results: We analyzed survey results from 116 eligible physicians. Physicians<br />

were more <strong>of</strong>ten in community (86%) than academic or other<br />

practices, and had a median <strong>of</strong> 14.5 years (range, 2-40) <strong>of</strong> practice<br />

experience. The median patient age was 62 years (range, 32–85). Most<br />

patients (81%) had T3 disease. Patients had �8, 9-11 and �12 nodes<br />

examined 3%, 13% and 84% <strong>of</strong> the time and 38% had comorbidities. Of<br />

the 76 patients tested for MMR/MSI, 13 (17%) were MMR-D or MSI-H and<br />

46 (61%) were MMR-P or MSI-L; 17 (22%) unknown. Median RS was 20<br />

(range, 1-77). Before obtaining the RS, chemotherapy was planned in 52<br />

(45%) patients, observation in 40 (34%), and there was no recommendation<br />

in 24 (21%). For the 92 patients with pre-assay recommendations,<br />

recommended treatment changed after obtaining the RS for 27 patients<br />

(29%). Treatment intensity decreased for 18 (67%) and increased for 9<br />

(33%) <strong>of</strong> 27 changed recommendations. A significant trend <strong>of</strong> decreasing<br />

treatment intensity with lower RS values was observed (p�.0035).<br />

Conclusions: These findings indicate that for stage II colon cancer patients,<br />

treatment recommendations were changed by RS results in 29% <strong>of</strong><br />

patients. Use <strong>of</strong> the Oncotype DX assay may lead to reductions in treatment<br />

intensity, contributing to the assay’s cost effectiveness.<br />

3628 General Poster Session (Board #38A), Mon, 8:00 AM-12:00 PM<br />

Use <strong>of</strong> gene set analysis (GSA) for molecular classification <strong>of</strong> responders<br />

and nonresponders to FOLFOX therapy in colorectal cancer (CRC). Presenting<br />

Author: Stephen Yip, British Columbia Cancer Agency, Vancouver, BC,<br />

Canada<br />

Background: Folinic acid (FOL) fluorouracil (F) and oxaliplatin (OX) chemotherapy<br />

is a commonly used therapy for CRC. Lacking in current literature<br />

are clinically relevant classifiers for potential responders. GSA technique is<br />

statistical method that detects significance <strong>of</strong> sets <strong>of</strong> genes, instead <strong>of</strong><br />

examining a gene-by-gene basis. The objective <strong>of</strong> this study was to identify<br />

differential functionally annotated gene expression pr<strong>of</strong>iles associated with<br />

response to FOLFOX therapy in CRC tumors using gene-by-gene and GSA<br />

approaches. Methods: Genome wide expression pr<strong>of</strong>ile data were collected<br />

on pre-treatment tumor tissues from patients with unresectable CRC<br />

receiving FOLFOX therapy (n � 83, Affymetrix HG U133A, GSE28702,<br />

Tsuji et al. BJC 2012). Gene expression was compared between responders<br />

(n � 42) and Non-responders (n � 41). GSA was conducted on 3272<br />

curated gene sets from the Molecular Signatures Database (Subramanian,<br />

Tamayo et al. 2005, PNAS 102, 15545-15550) annotated by biological<br />

pathway, biochemical function and clinical behavior. Significant analysis<br />

<strong>of</strong> Microarray (SAM) and GSA (Tibshirani et al.) was done to identify gene<br />

sets associated with FOLFOX response. Results: Differential expressions <strong>of</strong><br />

23 genes were significantly associated with response, based on a single<br />

gene approach (p-value � 0.05). 13 <strong>of</strong> these were located on Chromosome<br />

17 (p � 0.001). Among these, the top 5 ranked genes included NPEPPS,<br />

MBTD1, CEP44, LTA4H and CPNE4 which are involved in metal ion<br />

binding and aminopeptidase activity. GSA revealed only 44 out <strong>of</strong> 3272<br />

gene sets were significantly associated with response, with a false discovery<br />

rate less than 25%. Increased expression <strong>of</strong> B-lymphocyte differentiation<br />

and Ras-signalling-related gene sets was associated with responders while<br />

mTOR signaling and hematopoietic stem cell-related genes set were<br />

associated with non-responders. Conclusions: Our data showed that differential<br />

biological pathways could be identified to predict response to<br />

FOLFOX therapy for CRC patients. Analysis may be useful to help define<br />

clinically relevant biologic subtypes among patients with metastatic colorectal<br />

cancer.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


3629 General Poster Session (Board #38B), Mon, 8:00 AM-12:00 PM<br />

Phase II trial evaluating a 12-week regimen <strong>of</strong> interdigitating FOLFOX<br />

chemotherapy plus pelvic chemoradiation for the simultaneous treatment<br />

<strong>of</strong> primary and metastatic rectal cancer. Presenting Author: Michael<br />

Michael, Peter MacCallum Cancer Centre, Melbourne, Australia<br />

Background: Current chemotherapy regimens used during chemoradiation<br />

(CRT) are adequate for radiosensitization but suboptimal for systemic<br />

control. The aim <strong>of</strong> this study was to assess tolerability, and local/systemic<br />

benefits <strong>of</strong> new regimen delivering intensive chemotherapy and radical<br />

radiotherapy in an interdigitating manner. Methods: Phase II prospective<br />

study for patients (pts) with untreated simultaneous symptomatic primary<br />

and metastatic rectal cancer. The treatment regimen: 12 weeks long.<br />

FOLFOX chemotherapy (oxaliplatin 100mg/m2, leucovorin 200mg/m2,<br />

5-FU 400mg/m2 bolus, all day 1, and 5FU continuous infusion [CI] 2.4<br />

g/m2/46 hours) was given in weeks 1, 6, and 11. Pelvic CRT: 25.2 Gy in 3<br />

weeks, 1.8 Gy/fr, with concurrent oxaliplatin 85mg/m2 day 1 and 5-FU CI<br />

200mg/m2/day given in weeks 3-5, and 8-10. Pts received, in 12 weeks, 3<br />

courses <strong>of</strong> FOLFOX and pelvic radiation 50.4 Gy with concurrent oxaliplatin/<br />

5-FU. All pt were staged with CT, MRI and FDG-PET before and posttreatment.<br />

Results: 26 pts treated. The mean age was 61 years, 69% male.<br />

Rectal primary MRI stage was T2 4%, T3 81% and T4 15%. Liver and lung<br />

metastases were present in 81%, and 35% <strong>of</strong> pts, respectively: 38% <strong>of</strong><br />

patients had more than one site <strong>of</strong> metastatic disease. 24 pts (92%)<br />

completed the 12-week treatment regimen. All pts received the planned<br />

radiation dose. 65% <strong>of</strong> pts received the planned number <strong>of</strong> oxaliplatin<br />

courses with 88% <strong>of</strong> pts receiving at least 75% <strong>of</strong> the protocol oxaliplatin<br />

dose. In this 12-week period, grade 3 toxicities were neutropenia 23%,<br />

diarrhoea 15%, and radiation perineal skin reaction 12%. Only grade 4<br />

toxicity was neutropenia: 15%. PET metabolic response (CR�PR) rate for<br />

rectal primary was 96%. Overall PET metabolic response rate for metastatic<br />

disease was 60% (CR rate 16%). Conclusions: It is thus feasible to<br />

deliver intensive chemotherapy and radical radiotherapy in an interdigitating<br />

manner to treat both primary and metastatic rectal cancer simultaneously.<br />

High completion and response rates are encouraging. This regimen is<br />

the subject <strong>of</strong> a current phase II neoadjuvant trial for resectable rectal<br />

cancer (TROG 09.01).<br />

3631 General Poster Session (Board #38D), Mon, 8:00 AM-12:00 PM<br />

A phase I dose-escalation study <strong>of</strong> TAS-102 in patients (pts) with refractory<br />

metastatic colorectal cancer (mCRC). Presenting Author: Manish Patel,<br />

Florida Cancer Specialists/Sarah Cannon Research Institute, Sarasota, FL<br />

Background: TAS-102 is an oral combination <strong>of</strong> a novel antimetabolite<br />

5-trifluorothymidine (FTD) and a thymidine phosphorylase inhibitor (TPI)<br />

that prevents degradation <strong>of</strong> FTD. In preclinical studies, TAS-102 is<br />

effective against human colorectal tumors with innate and acquired<br />

resistance to 5FU. A Japanese randomized phase 2 study demonstrated a<br />

significant survival improvement <strong>of</strong> TAS-102 over placebo in refractory<br />

mCRC (HR�0.56 p�0.0011, EMCC2011). The present study was conducted<br />

to determine the MTD <strong>of</strong> TAS-102 in Western pts with refractory<br />

mCRC. Methods: Pts with mCRC who had received at least 2 lines <strong>of</strong><br />

chemotherapy including a fluoropyrimidine, irinotecan and oxaliplatin were<br />

enrolled at an initial dose level <strong>of</strong> 30 mg/m2 BID days 1- 5 and days 8-12<br />

every 4wks using a standard 3�3 design. A second dose level <strong>of</strong> 35 mg/m2 ,<br />

which was the MTD for Japanese pts, was the maximal targeted dose in this<br />

study. Results: 12 pts received TAS-102 (30/35 mg/m2�3/9, M/F�7/5,<br />

Age 44-75, PS0/1�8/4). Pts received a median <strong>of</strong> 3 prior regimens for<br />

metastatic disease. No DLT was observed in the initial dose level (0/3) and<br />

one DLT (grade 3 febrile neutropenia) was observed at 35mg/m2 (1/9).<br />

Other drug-related � grade 3 toxicities observed in cycle 1 included<br />

neutropenia at 58% (7/12). To date, a median number <strong>of</strong> treatment cycles<br />

at the MTD has not been reached as 5 pts are still receiving therapy (range<br />

1-3� cycles). Conclusions: Western patients with refractory mCRC showed<br />

the same MTD as Japanese pts (35 mg/m2 BID, days 1- 5 and days 8-12<br />

every 4wks) and the safety pr<strong>of</strong>ile was consistent between the populations.<br />

The trial has entered an expansion phase to assess further safety and<br />

preliminary efficacy. A phase 3 trial is being planned.<br />

Gastrointestinal (Colorectal) Cancer<br />

235s<br />

3630 General Poster Session (Board #38C), Mon, 8:00 AM-12:00 PM<br />

Evaluation <strong>of</strong> the prognostic utility <strong>of</strong> miRNA and multiple genetic markers<br />

in stage IIA colon cancer. Presenting Author: Elizabeth Mambo, Asuragen<br />

Inc., Austin, TX<br />

Background: The variable prognoses <strong>of</strong> stage II colon cancer subjects have<br />

provoked a therapeutic dilemma <strong>of</strong> who should receive adjuvant therapy.<br />

Presently, prognosis is evaluated on <strong>of</strong>ten subjective histopathological<br />

features. Reliable, objective markers that can accurately predict recurrence<br />

risk are needed. Using retrospectively collected tumor samples with known<br />

clinical outcome, we evaluated the prognostic utility <strong>of</strong> miRNA together<br />

with clinicopathological features and molecular markers with suggestive<br />

prognostic value. Methods: We evaluated the expression <strong>of</strong> miRNA in 118<br />

stage IIA FFPE tumors (73 non-recurrent, NR; 45 recurrent, R) using the<br />

Affymetrix Gene Chip V1.0 microarray, and performed RT-qPCR to verify<br />

select miRNAs. We also sequenced KRAS codons 12/13, and BRAF codon<br />

600, and assessed MSI status using a fluorescent PCR-based assay. MMR<br />

protein expression was determined by IHC. An empirical approach was<br />

applied to screen miRNA using combinatorial criteria <strong>of</strong> microarray 2-way<br />

ANOVA analysis, Kaplan-Meier curve logrank, and single and multivariate<br />

Cox-regression. miRNA was the most significant predictor among all<br />

histopathological and molecular features (FDR�0.05). A miRNA classifier<br />

was built with nested two-layer cross-validation. Results: The top miRNA<br />

model was selected based on AUC estimates and Cox-regression p-value.<br />

This model included 30 miRNAs; 25 <strong>of</strong> these were common with the<br />

empirical approach, confirming the robustness <strong>of</strong> candidate selection. The<br />

miRNA classifier distinguished R from NR cases with an AUC <strong>of</strong> ~0.74.<br />

Among all histopathological and molecular features evaluated, grade was<br />

significantly associated with disease-free survival (logrank p�0.05). Both<br />

single variate and multivariate Cox analysis revealed that grade and PMS2<br />

status were strongly associated with recurrence (p�0.05). Lastly, lack <strong>of</strong><br />

expression <strong>of</strong> PMS2 and MLH1, BRAF V600E and MSI was highly<br />

correlated (� coefficient � 0.6), and associated with non-recurrent<br />

disease. Conclusions: These data provide strong support for miRNAs as<br />

independent prognostic biomarkers for stage IIA colon cancer.Validation<br />

work is ongoing to demonstrate the potential utility <strong>of</strong> these miRNAs in<br />

patient stratification.<br />

3632 General Poster Session (Board #38E), Mon, 8:00 AM-12:00 PM<br />

Surgical resection and peri-operative chemotherapy for colorectal cancer<br />

(CRC) liver metastases in routine clinical practice: A population-based<br />

outcomes study. Presenting Author: Christopher M. Booth, Queen’s University<br />

Cancer Research Institute, Kingston, ON, Canada<br />

Background: Resection <strong>of</strong> liver metastases combined with peri-operative<br />

chemotherapy is an important treatment option for patients with advanced<br />

CRC. Most <strong>of</strong> the literature describes outcomes achieved in highly selected<br />

patients treated at a few high volume institutions. Here we report the<br />

results <strong>of</strong> a population-based study <strong>of</strong> the management and outcome <strong>of</strong> all<br />

patients who underwent resection <strong>of</strong> CRC liver metastases in Ontario,<br />

Canada. Methods: All cases <strong>of</strong> CRC in Ontario who underwent surgical<br />

resection <strong>of</strong> liver metastases in 1994-2009 were identified using the<br />

population-based Ontario Cancer Registry (OCR). The OCR captures<br />

diagnostic and demographic information on ~98% <strong>of</strong> all incident cancer<br />

cases in Ontario. We linked electronic records <strong>of</strong> treatment to the OCR to<br />

identify surgery, neoadjuvant (NACT) and adjuvant chemotherapy (ACT).<br />

We describe time trends in treatment and survival using 3 study periods:<br />

1994-1999, 2000-2004, 2005-2009. Results: During 1994-2009, 2717<br />

patients underwent resection <strong>of</strong> CRC liver metastases in Ontario; mean age<br />

was 65 years and 61% were male. From 1994-2009 there was a 103%<br />

increase in the number <strong>of</strong> patients undergoing resection <strong>of</strong> liver metastases<br />

(117/year to 237/year) while incident cases <strong>of</strong> CRC during this time<br />

increased by 31% (5285/year to 6956/year). Use <strong>of</strong> NACT increased over<br />

the study period: 94-99, 11% (78/700); 00-04, 15% (124/830); 05-09,<br />

36%; (424/1187), (p�0.001). Use <strong>of</strong> ACT also increased: 94-99, 38%<br />

(263/700); 00-04, 40% (335/830); 05-09, 45% (532/1187), (p�0.006).<br />

In 2005-2009 there was substantial variation across geographic regions in<br />

use <strong>of</strong> NACT (range 19% to 46%, p�0.029) and ACT (range 31% to 56%,<br />

p�0.015). Five year overall survival during the 3 study periods was 36%<br />

(95%CI 32-39%), 40% (95%CI 36-43%), and 47% (95%CI 43-51%)<br />

(p�0.001). Conclusions: Resection <strong>of</strong> CRC liver metastases in routine<br />

practice in the general population <strong>of</strong> Ontario is associated with survival<br />

outcomes that are comparable to those reported in case series from leading<br />

comprehensive cancer centres. Survival improved over the study period<br />

despite a greater proportion <strong>of</strong> patients with CRC undergoing liver resection.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


236s Gastrointestinal (Colorectal) Cancer<br />

3633 General Poster Session (Board #38F), Mon, 8:00 AM-12:00 PM<br />

Triplet chemotherapy (TC) with FOLFIRINOX regimen in metastatic colorectal<br />

cancer (mCRC): Experience <strong>of</strong> the Val d’Aurelle Center. Presenting<br />

Author: Emmanuelle Samalin, Digestive Oncology CRLC Val d’Aurelle,<br />

Montpellier, France<br />

Background: TC is a treatment option for mCRC to improve the tumor<br />

response rate in selected patients (pts) and the conversion rate <strong>of</strong> initially<br />

non-resectable liver metastases. The aim <strong>of</strong> this study was to evaluate the<br />

impact and feasibility <strong>of</strong> FOLFIRINOX regimen in mCRC pts. Methods: All<br />

mCRC pts with non-resectable disease who have received FOLFIRINOX<br />

alone or combined with targeted therapies (bevacizumab or cetuximab)<br />

from October 2000 to December 2010 were selected for this analysis.<br />

<strong>Clinical</strong> data were collected in a mCRC specific data base and analyzed by<br />

the end <strong>of</strong> 2011. Results: Ninety two pts (52% <strong>of</strong> men), median age 59 yrs<br />

(range: 27-76) were treated with FOLFIRINOX (D1 oxaliplatin 85 mg/m² IV<br />

2H then irinotecan 180 mg/m² IV 90 min and elvorin 200 mg/m², then 5FU<br />

200 mg/m² and 2400 mg/m² by 46H infusion, D1�D15) alone (64%) or<br />

combined with cetuximab(30%) or bevacizumab (6%), as 1st-line in 82 pts<br />

(89%). Prophyllactic G-CSF was given in 58% <strong>of</strong> them. Primary tumor was<br />

located in colon (58%) or rectum (42%), and 64 (69%) <strong>of</strong> pts presented<br />

with synchronous metastases: liver 100%, lung 40%, peritoneum 17% and<br />

nodes 17%. Median number <strong>of</strong> cures was 8 (range: 1-12). There was 1<br />

toxic death. Grade 3-4 toxicities were: diarrhea 22%, neuropathy 21%,<br />

cutaneous 12%, neutropenia 28%, febrile neutropenia 0%, thrombopenia<br />

6%. Objective Response rate according to RECIST criteria was 72%<br />

[CI95% 61-81] including 10 pts with complete response (11%). The<br />

primary tumor was resected in 70 pts (76%) and 14% had KRAS mutated<br />

tumor. Among the pts with liver metastases, 63 (68%) pts were evaluated<br />

for secondary resectability by a multidisciplinary committee and 40 pts<br />

(43%) had resection achieved (70% R0). Median overall survival was 49<br />

months [CI95%28-62]. Conclusions: These results confirm the feasibility<br />

<strong>of</strong> FOLFIRINOX regimen with or without targeted therapies and its efficacy<br />

in terms <strong>of</strong> response rate and overall survival as 1rst-line treatment in<br />

selected mCRC pts.<br />

TPS3635^ General Poster Session (Board #38H), Mon, 8:00 AM-12:00 PM<br />

MAVERICC: A randomized phase II study <strong>of</strong> mFOLFOX6-bevacizumab (BV)<br />

versus FOLFIRI-BV with prospective biomarker stratification in previously<br />

untreated metastatic colorectal cancer (mCRC). Presenting Author: Sachdev<br />

P. Thomas, Illinois Cancer Care, Peoria, IL<br />

Background: The identification <strong>of</strong> prognostic and predictive biomarkers<br />

could significantly improve the risk-benefit ratio and cost-effectiveness <strong>of</strong><br />

1st-line mCRC regimens. This is the first prospective study <strong>of</strong> tumoral<br />

ERCC1 (chemo-resistance marker to platinum compounds) and plasma<br />

VEGF-A as potential biomarkers for oxaliplatin- and BV-containing regimens,<br />

respectively, in an effort to further define the optimal chemotherapy<br />

backbone with biologic therapies, including BV, for mCRC. Methods: In this<br />

randomized, open-label, global, phase II study, patients (N�360) with<br />

histologically or cytologically confirmed CRC and �1 measurable metastatic<br />

lesion are stratified at screening by tumoral ERCC1 mRNA expression<br />

(high vs low, cut<strong>of</strong>f <strong>of</strong> 1.7 [ERCC1/�-actin mRNA]). Eligibility criteria<br />

include completion <strong>of</strong> adjuvant therapy �12 months before screening and<br />

an ECOG performance status �1. Blood samples are collected to quantify<br />

plasma VEGF-A levels. Patients within each ERCC1 stratification group are<br />

randomized 1:1 to mFOLFOX6-BV or FOLFIRI-BV administered in 2-week<br />

cycles. BV will be given at a dose <strong>of</strong> 5 mg/kg IV q2w. Patients will remain on<br />

study treatment until disease progression (PD) or unacceptable toxicity. If<br />

oxaliplatin or irinotecan need to be discontinued, BV and 5-fluorouracil or<br />

capecitabine are to be continued until PD. The primary objectives are: 1) to<br />

assess ERCC1 and VEGF-A as biomarkers <strong>of</strong> progression-free survival (PFS)<br />

for oxaliplatin- and BV-containing regimens in 1st-line mCRC, and 2)<br />

within ERCC1 high patients, to test whether FOLFIRI-BV is associated with<br />

a prolonged 1st-line PFS compared to mFOLFOX6-BV. Secondary objectives<br />

include assessing the impact <strong>of</strong> these markers on overall survival,<br />

objective response, hepatic metastases resection, and safety. Exploratory<br />

endpoints include correlative analyses with additional tumor tissue, blood,<br />

and SNP markers. The first patient was enrolled in August 2011. An<br />

interim biomarker distribution assessment <strong>of</strong> the first 100 patients is<br />

planned, and the evaluation <strong>of</strong> the primary endpoints is estimated for early<br />

2015. <strong>Clinical</strong>trials.gov: NCT01374425.<br />

TPS3634 General Poster Session (Board #38G), Mon, 8:00 AM-12:00 PM<br />

A randomized, double-blind, phase (Ph) III study <strong>of</strong> the irinotecan-based<br />

chemotherapy FOLFIRI plus ramucirumab (RAM) or placebo (PL) in<br />

patients (pts) with metastatic colorectal carcinoma (mCRC) progressive<br />

during or following first-line therapy with bevacizumab (BEV), oxaliplatin<br />

(OXALI), and a fluoropyrimidine (FP) (RAISE) (NCT01183780). Presenting<br />

Author: Axel Grothey, Mayo Clinic College <strong>of</strong> Medicine, Rochester, MN<br />

Background: Vascular endothelial growth factor (VEGF) and the VEGF<br />

receptor-2 (VEGFR-2) regulate angiogenesis and are overexpressed in CRC.<br />

RAM is a fully human IgG1 monoclonal antibody that inhibits binding <strong>of</strong><br />

VEGF ligands to VEGFR-2 and inhibits VEGFR-2 activation and signaling.<br />

In a preclinical study, antiangiogenic and antitumor effects were observed<br />

when DC101, an antibody that targets murine VEGFR-2, was administered<br />

to mice bearing human colon cancer xenografts. Antitumor activity for RAM<br />

has been demonstrated in a Ph I study in pts with solid tumors over a wide<br />

range <strong>of</strong> RAM dose levels and in a Ph II study with RAM � mFOLFOX-6 as<br />

1st-line therapy in pts with mCRC. Methods: This ongoing, randomized,<br />

double-blind, placebo-controlled Ph III study includes mCRC pts with<br />

measurable or nonmeasurable disease and ECOG performance status <strong>of</strong><br />

0-1 who have experienced disease progression during or within 6 months<br />

following 1st-line therapy with BEV, OXALI, and any FP. Randomization is<br />

stratified by geographic region, KRAS status, and time to progression after<br />

initiation <strong>of</strong> 1st-line therapy. Pts are randomized 1:1 to either RAM 8 mg/kg<br />

or PL every 14 days. Pts in both arms receive FOLFIRI (irinotecan: 180<br />

mg/m2 , folinic acid: 400 mg/m2 , 5-fluorouracil: 400 mg/m2 bolus followed<br />

by 2400 mg/m2 continuous infusion over 46-48 hours). The primary<br />

endpoint is overall survival (OS). The sample-size estimate assumes 85%<br />

power to detect at least a 2.5-month median OS difference (HR � 0.8)<br />

between treatment arms with a 1-sided alpha <strong>of</strong> 0.025. Secondary<br />

endpoints include progression-free survival, tumor response, safety, pharmacokinetics,<br />

immunogenicity, and correlations between biomarkers and<br />

clinical outcome. CRC tissue and blood collection is mandatory for<br />

biomarker analyses. As <strong>of</strong> 05 January 2012, 238 <strong>of</strong> 1050 planned pts have<br />

been enrolled from 100 sites in North America, South America, Europe,<br />

Asia, and Australia.<br />

TPS3636 General Poster Session (Board #39A), Mon, 8:00 AM-12:00 PM<br />

TRICC-c: BIBF 1120 versus placebo in patients receiving oxaliplatin plus<br />

fluorouracil and leucovorin (mFOLFOX6) for advanced, chemorefractory<br />

metastatic colorectal cancer (mCRC)—A multicenter, randomized phase II<br />

trial in progress. Presenting Author: Andreas Berger, Department <strong>of</strong><br />

Internal Medicine I, Martin-Luther-University Halle-Wittenberg, Halle,<br />

Germany<br />

Background: Colorectal cancer (CRC) is the second leading cause <strong>of</strong><br />

cancer-related death in western countries. With advances in the treatment<br />

<strong>of</strong> metastatic CRC (mCRC) in the last decade using combination chemotherapy<br />

and bevacizumab, a monoclonal antibody against the vascular<br />

endothelial growth factor (VEGF), progression free survival (PFS) in first<br />

and second line setting was substantially improved. Tumour angiogenesis<br />

is also driven by other factors but VEGF including Platelet Derived Growth<br />

Factor (PDGF) and Fibroblast Growth Factor (FGF). BIBF1120 is a potent,<br />

orally available triple angiokinase inhibitor that blocks the receptor tyrosine<br />

kinase activity <strong>of</strong> human VEGFR1-3, FGFR1 and -3 and PDGFR� and -�.<br />

Thus, BIBF1120 could be an exciting addition to the treatment <strong>of</strong> patients<br />

with chemorefractory CRC. Methods: Randomized, double-blind, placebocontrolled,<br />

multicentre, phase II trial <strong>of</strong> BIBF1120 (orally, 200 mg, bid,<br />

d1-d14, Arm A) vs. placebo (Arm B) in patients receiving mFOLFOX6<br />

(oxaliplatin, 85 mg/m2 , fluorouracil, 400 mg/m2 bolus and 2400 mg/m2 over 46 h, leucovorin, 200 mg/m2 ) q14 d for patients (ECOG performance<br />

status 0-1) with chemorefractory mCRC who received one prior line <strong>of</strong><br />

chemotherapy. Scheduled are 90 patients per treatment arm. Primary<br />

endpoint: PFS, Secondary endpoints: objective response, overall survival,<br />

duration <strong>of</strong> overall response, safety. Additionally there will be an explorative<br />

analysis <strong>of</strong> predictive biomarkers for BIBF1120. 4 <strong>of</strong> planned 180 patients<br />

have been enrolled. We expect the last patient out in June 2013. (Trial<br />

identifier: NCT01362361.)<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


TPS3637 General Poster Session (Board #39B), Mon, 8:00 AM-12:00 PM<br />

The GAIN-C study (BP25438): Randomized phase II trial <strong>of</strong> RG7160<br />

(GA201) plus FOLFIRI, compared to cetuximab plus FOLFIRI or FOLFIRI<br />

alone in second-line KRAS wild type (WT) or mutant metastatic colorectal<br />

cancer (mCRC). Presenting Author: Andres Cervantes-Ruiperez, Department<br />

<strong>of</strong> Hematology and Medical Oncology, INCLIVA, University <strong>of</strong> Valencia,<br />

Valencia, Spain<br />

Background: GA201 is a novel, dual-acting, humanized, glycoengineered<br />

IgG1 anti-EGFR monoclonal antibody, with enhanced antibody-dependent<br />

cellular cytotoxicity (ADCC) activity in combination with signal inhibition.<br />

GA201 demonstrates significantly enhanced in vitro/vivo activity compared<br />

to cetuximab (cet) both as a single agent and in combination with<br />

irinotecan, in both KRAS mutant and BRAF mutant models and promising<br />

clinical activity in ph I and neo-adjuvant trials (Paz Ares et al, JCO 2011)<br />

including KRAS mutant mCRC. A randomized ph II program was launched:<br />

one study in NSCLC and GAIN-C in mCRC (NCT01326000), which is<br />

presented here. Methods: Main inclusion criteria are progression on 1L<br />

containing oxaliplatin, ECOG 0-1, and adequate hematological and liver<br />

function. Main exclusion criteria: prior anti-EGFR treatment. A total <strong>of</strong> 160<br />

patients in 2L mCRC (stratified for EGFR expression, disease progression<br />

before or after 6 months after starting 1L, prior treatment with bevacizumab<br />

Y vs N) will be randomized to receive either GA201 (day 1, 8 <strong>of</strong> cycle<br />

1 then q2W) or cet (qW) � FOLFIRI q2W (KRAS WT) or to receive GA201�<br />

FOLFIRI or FOLFIRI alone (KRAS mutant). Collection <strong>of</strong> archival tumor<br />

plus a mandatory fresh tumor biopsy at baseline were implemented<br />

because ph I data showed that EGFR expression is not concordant between<br />

the two specimen types and to optimize assessment <strong>of</strong> potential immune<br />

related biomarkers. The fresh tumor biopsy will be centrally analyzed for<br />

EGFR (immunohistochemistry) and KRAS status. Primary objective is<br />

progression free survival; secondary endpoints are to define objective<br />

response rates, the safety pr<strong>of</strong>ile, pharmacokinetics and pharmacodynamics.<br />

A comprehensive biomarker program (blood and tumor), mainly<br />

immune-phenotyping, immunohistochemistry in tumor samples (Ventana)<br />

and immune functional tests (including adaptive responses) were set up to<br />

investigate potential predictive biomarkers and the mode <strong>of</strong> action <strong>of</strong><br />

GA201. Study is ongoing worldwide in 9 countries with the safety run-in<br />

phase completed in Nov 2011. Recruitment is planned to be completed by<br />

end <strong>of</strong> April 2012.<br />

TPS3639 General Poster Session (Board #39D), Mon, 8:00 AM-12:00 PM<br />

Sequential first-line treatment for metastatic colorectal cancer with capecitabine,<br />

irinotecan, and bevacizumab: Capecitabine plus bevacizumab<br />

(Cape-Bev) versus capecitabine, irinotecan plus bevacizumab (CAPIRI-<br />

Bev): The AIO KRK 0110/ML22011 randomized, phase III trial. Presenting<br />

Author: Clemens Albrecht Giessen, Department <strong>of</strong> Hematology and Oncology,<br />

Klinikum Grosshadern and Comprehensive Cancer Center, LMU<br />

Munich, Munich, Germany<br />

Background: Several randomized trials have indicated that combination<br />

chemotherapy applied in metastatic colorectal cancer (mCRC) does not<br />

significantly improve overall survival when compared to the sequential use<br />

<strong>of</strong> cytotoxic agents (CAIRO, MRC Focus, FFCD 2000-05). The present<br />

study investigates the question in bevacizumab-based first-line treatment<br />

including escalation- and de-escalation strategies. Methods: The AIO KRK<br />

0110/ML22011 trial (<strong>Clinical</strong>Trials.gov NCT01249638) is a two-arm,<br />

multicenter, open-label randomized phase III trial comparing the efficacy<br />

and safety <strong>of</strong> Cape-Bev versus CAPIRI-Bev in the first-line treatment <strong>of</strong><br />

metastatic colorectal cancer. Patients with unresectable metastatic colorectal<br />

cancer, ECOG PS 0-1, will be assigned in a 1:1 ratio to receive either<br />

capecitabine 1250 mg/m2 bid for 14d (d1-14) plus bevacizumab 7.5<br />

mg/kg (d1) q3w (Arm A) or capecitabine 800 mg/m2 BID for 14d (d1-14),<br />

irinotecan 200 mg/m2 (d1) and bevacizumab 7.5 mg/kg (d1) q3w (Arm B).<br />

Patients included into this trial are required to consent to the analysis <strong>of</strong><br />

tumor tissue and blood for translational investigations. In Arm A, treatment<br />

escalation from Cape-Bev to CAPIRI-Bev is recommended in case <strong>of</strong><br />

progressive disease (PD). In Arm B, de-escalation from CAPIRI-Bev to<br />

Cape-Bev is possible after 6 months <strong>of</strong> treatment or in case <strong>of</strong> irinotecanassociated<br />

toxicity. Re-escalation to CAPIRI-Bev after PD is possible. The<br />

primary endpoint is time to failure <strong>of</strong> strategy (TFS). Secondary endpoints<br />

are ORR, OS, PFS, safety and quality <strong>of</strong> life. Conclusion: The AIO KRK<br />

0110 trial is designed for patients with disseminated, but asymptomatic<br />

mCRC who are not potential candidates for surgical resection <strong>of</strong> metastasis.<br />

Two bevacizumab-based strategies are compared: one starting as singleagent<br />

chemotherapy (Cape-Bev) allowing escalation to CAPIRI-Bev and<br />

another starting with CAPIRI-Bev and allowing de-escalation to Cape-Bev<br />

and subsequent re-escalation if necessary. By January 2012, 79 <strong>of</strong><br />

planned 516 patients have been enrolled.<br />

Gastrointestinal (Colorectal) Cancer<br />

237s<br />

TPS3638 General Poster Session (Board #39C), Mon, 8:00 AM-12:00 PM<br />

A pilot trial <strong>of</strong> a combination <strong>of</strong> therapeutic vaccines (GI-4000 and<br />

GI-6207) as adjunctive therapy with first-line therapy with bevacizumab<br />

plus either FOLFOX or FOLFIRI in stage IV patients with newly diagnosed<br />

Ras-mutant positive or negative metastatic colorectal cancer. Presenting<br />

Author: John Marshall, Lombardi Comprehensive Cancer Center, Georgetown<br />

University, Washington, DC<br />

Background: A promising approach for the treatment <strong>of</strong> cancer is the<br />

development <strong>of</strong> vaccines that target specific tumor antigens. In the<br />

metastatic CRC patient population, targeted and active immunotherapy<br />

may inhibit cancer progression and improve survival. This trial is designed<br />

to evaluate the efficacy, immunogenicity, and safety <strong>of</strong> GI-4000 plus<br />

standard therapy in patients with metastatic colorectal cancer. GI-4000 is<br />

a proprietary immunotherapy that uses whole, heat-killed recombinant<br />

Saccharomyces cerevisiae yeast (called Tarmogens � Targeted Molecular<br />

Immunogens). GI-4000 is designed to activate a cellular immune response<br />

to target cells with activating ras mutations. Tarmogens have been shown to<br />

elicit selective killing <strong>of</strong> target cells that express a number <strong>of</strong> cancer<br />

antigens, including mutated Ras, by activation <strong>of</strong> antigenspecific T cell<br />

mediated responses. Methods: The study population consists <strong>of</strong> subjects<br />

with metastatic colorectal cancer with an activating mutation in ras. Newly<br />

diagnosed subjects receive FOLFOX (or FOLFIRI) � bevacizumab (Bev) �<br />

GI- 4000; 3 weekly injections <strong>of</strong> GI-4000 are followed by 8 cycles <strong>of</strong> Bev �<br />

FOLFOX (or FOLFIRI); day 1 and 2 every 14 days. Doses <strong>of</strong> GI-4000 are<br />

administered on day 8 <strong>of</strong> each cycle. Upon completion <strong>of</strong> chemotherapy,<br />

GI-4000 continues along with Bev maintenance every 2 weeks for up to 5<br />

years or until subjects experience intolerance, disease recurrence, or death.<br />

If Bev is stopped, GI-4000 may continue on the same maintenance<br />

schedule alone. Subjects that have already completed standard chemotherapy<br />

(FOLFOX or FOLFIRI) may enter the study and receive Bev �<br />

GI-4000 every 2 weeks for up to 5 years. Enrollment is ongoing and will<br />

continue up to 52 subjects.<br />

TPS3640 General Poster Session (Board #39E), Mon, 8:00 AM-12:00 PM<br />

A randomized, phase II, multicenter, double-blind, placebo-controlled<br />

study evaluating onartuzumab (MetMAb) in combination with mFOLFOX6<br />

plus bevacizumab in patients with metastatic colorectal cancer. Presenting<br />

Author: Johanna C. Bendell, Sarah Cannon Research Institute, Nashville,<br />

TN<br />

Background: Dysregulation <strong>of</strong> the HGF/Met (Met) pathway has been linked<br />

with poor prognosis in colorectal cancer. Crosstalk between the Met and<br />

vascular endothelial growth factor (VEGF) pathways may be important<br />

during tumorigenesis. Aberrant activation <strong>of</strong> the HGF/Met pathway may<br />

promote angiogenesis via tumor cell secretion <strong>of</strong> angiogenic factors or<br />

directly activating endothelial cells. Onartuzumab (MetMAb) is a monovalent,<br />

monoclonal antibody that specifically binds to the Met receptor. The<br />

combination <strong>of</strong> onartuzumab and VEGF inhibition in preclinical models<br />

resulted in enhanced antitumor activity over either treatment alone.<br />

Preclinical efficacy data support the combination <strong>of</strong> onartuzumab with<br />

platinum agents. In phase I studies, onartuzumab has been generally well<br />

tolerated alone and in combination with bevacizumab. Adverse events most<br />

commonly associated with onartuzumab are peripheral edema and fatigue.<br />

Methods: This is a randomized, two-arm, phase II study in patients with<br />

previously untreated metastatic colorectal cancer. Patients (n�188) will<br />

be randomized (1:1) to either mFOLFOX6/bevacizumab/placebo or mFOL-<br />

FOX6/bevacizumab/onartuzumab. Oxaliplatin will be discontinued after 8<br />

cycles with remaining drugs continued until progression. The primary<br />

endpoint <strong>of</strong> this study is PFS in all patients. PFS by Met IHC diagnostic<br />

status (Met positive vs Met negative) will also be analyzed. Secondary<br />

endpoints include OS, ORR, safety, and biomarker analyses. Primary and<br />

secondary analyses will include all randomized patients and will be<br />

conducted according to assigned treatment arm. Kaplan–Meier methodology<br />

will be used to estimate median PFS for each treatment arm. An<br />

estimate <strong>of</strong> HR with 95% CI will be determined using a Cox regression<br />

model. Safety will be assessed in all patients receiving at least one dose <strong>of</strong><br />

any treatment. This study is open for accrual; further details can be found<br />

on <strong>Clinical</strong>Trials.gov (NCT01418222).<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


238s Gastrointestinal (Colorectal) Cancer<br />

TPS3641^ General Poster Session (Board #39F), Mon, 8:00 AM-12:00 PM<br />

A randomized, double-blind, placebo-controlled, multicenter, multinational,<br />

phase II trial <strong>of</strong> L-BLP25 in patients with colorectal carcinoma<br />

following R0/R1 hepatic metastasectomy. Presenting Author: Carl Christoph<br />

Schimanski, University <strong>of</strong> Mainz, Mainz, Germany<br />

Background: Approximately 15-20% <strong>of</strong> patients diagnosed with colorectal<br />

cancer (crc) develop metastatic disease. Surgical resection remains the<br />

only potentially curative treatment. 5-year survival following R0-resection<br />

<strong>of</strong> liver metastases lies ~28 -39%. Recurrence occurs in ~70% <strong>of</strong> pts.<br />

Adjuvant chemotherapy has not significantly improved clinical outcomes.<br />

The primary objective <strong>of</strong> the LICC trial (L-BLP25 in Colorectal Cancer) is to<br />

analyze whether L-BLP25, an active cancer immunotherapy, extends<br />

recurrence-free survival (RFS) time over placebo in colorectal cancer pts<br />

following R0/R1 resection <strong>of</strong> hepatic metastases. L-BLP25 targets MUC1<br />

glycoprotein, which is highly expressed in hepatic metastases from crc. In a<br />

phase IIB trial, L-BLP25 showed acceptable tolerability and a trend toward<br />

longer survival in pts with stage IIIB NSCLC. Methods: This is a multinational,<br />

phase II, multicenter, randomized, double-blind, placebo-controlled<br />

trial with a sample size <strong>of</strong> 159 pts from 20 centers in 3 countries. Pts must<br />

have stage IV cr adenocarcinoma limited to liver metastases. Following<br />

complete resection <strong>of</strong> the primary tumor and all syn-/metachronous<br />

metastases, eligible pts are randomized 2:1 to receive either L-BLP25 or<br />

placebo. Those allocated to L-BLP25 receive a single dose <strong>of</strong> 300 mg/m2 cyclophosphamide (CP) 3 days before first L-BLP25 dose, then primary<br />

treatment with sc L-BLP25 930 �g once weekly for 8 weeks, followed by<br />

maintenance doses at 6-week (years 1 and 2) and 12-week (year 3)<br />

intervals until recurrence. In the control arm, CP is replaced by saline<br />

solution and L-BLP25 by placebo. Primary endpoint: RFS time. Secondary<br />

endpoints: OS time, safety status, tolerability, RFS/OS in MUC-1 positive<br />

cancers. Exploratory immune response analyses are planned. First recruitment<br />

was <strong>of</strong> Q3 2011. To date, 8 <strong>of</strong> 20 centers are initiated and 4 pts<br />

recruited. Completion <strong>of</strong> recruitment is scheduled for Q3 2013. Primary<br />

endpoint will be assessed in Q3 2016: Follow-up will end Q3 2017. No<br />

interim analysis is planned. Design and implementation <strong>of</strong> this vaccination<br />

study in colorectal cancer is feasible. No major issues identified during<br />

setup <strong>of</strong> the study.<br />

TPS3643 General Poster Session (Board #39H), Mon, 8:00 AM-12:00 PM<br />

ATTACHE: A phase III, multicenter, randomized comparison <strong>of</strong> chemotherapy<br />

given prior to and post surgical resection versus chemotherapy<br />

given post surgical resection for hepatic metastases from colorectal cancer.<br />

Presenting Author: Jonathan Fawcett, Queensland Liver Transplant Service,<br />

Brisbane, Australia<br />

Background: No randomized studies have directly compared the role <strong>of</strong><br />

peri-operative to adjuvant chemotherapy for resectable liver metastases.<br />

Benefit from post operative compared to peri-operative treatment has been<br />

suggested in a recent retrospective study <strong>of</strong> 499 patients with resected<br />

colorectal liver metastases which showed improved survival with entirely<br />

post-operative chemotherapy. Given this data and that <strong>of</strong> the small<br />

randomised trials demonstrating improved surgical outcomes with adjuvant<br />

chemotherapy, the role <strong>of</strong> entirely post-operative chemotherapy as a means<br />

<strong>of</strong> improving outcomes while reducing the negative effects <strong>of</strong> pre-operative<br />

treatment needs to be examined. Methods: 200 patients randomized 1:1 to<br />

6 months <strong>of</strong> treatment post-operatively or 3 months <strong>of</strong> treatment preoperatively<br />

and 3 months post-operatively. Site investigators will nominate<br />

chemotherapy schedule (mFOLFOX6, XELOX or FOLFIRI when adjuvant<br />

oxaliplatin received previously) prior to randomisation. Primary endpoint:<br />

proportion <strong>of</strong> patients in each arm with surgical complications within 30<br />

days. Secondary endpoints: proportion <strong>of</strong> patients completing planned<br />

chemotherapy, post operative mortality rate (in each group), tolerability<br />

and safety <strong>of</strong> treatment, response rate by RECIST V1.1 and CEA, time to<br />

progression, time to treatment failure, overall survival, QoL (EORTC<br />

QLQ-C30 and QLQ-LMC21). A planned prospective meta-analysis with<br />

MRC (UK) and NSABP C-11 trials will have sufficient power to examine the<br />

effect <strong>of</strong> schedule (peri- or post-operative) on progression free survival<br />

(PFS). Eligibility: Patients with histologically proven colorectal cancer with<br />

radiologically confirmed, resectable liver metastases without evidence <strong>of</strong><br />

extra-hepatic disease are eligible. Patients with synchronous metastases<br />

who have undergone resection <strong>of</strong> the primary tumour are eligible but<br />

patients requiring combined resection <strong>of</strong> primary cancer and liver metastatic<br />

disease are excluded. Patients with involved hilar nodes or wound<br />

implant metastases will not be eligible. Trial Status: Study opened to<br />

accrual August 2011.<br />

TPS3642 General Poster Session (Board #39G), Mon, 8:00 AM-12:00 PM<br />

FOLFIRI plus 5 mg/kg <strong>of</strong> bevacizumab versus 10 mg/kg as second-line<br />

therapy in patients with metastatic colorectal cancer who have failed<br />

first-line bevacizumab plus oxaliplatin-based therapy: A randomized phase<br />

III study (EAGLE Study). Presenting Author: Masato Nakamura, Aizawa<br />

Hospital, Matsumoto, Japan<br />

Background: There has been much controversy about continuation <strong>of</strong><br />

bevacizumab (BV) with a second-line regimen after progression on a<br />

BV-containing first-line regimen. Recently, it was announced that a phase<br />

III study (AIO 0504 / ML 18147) met its primary endpoint <strong>of</strong> overall<br />

survival. It means BV-based regimen (5 mg/kg) extends survival when<br />

continued beyond initial treatment in patients with metastatic colorectal<br />

cancer(mCRC). The verified data from a randomized phase III study<br />

(E3200) indicates that BV at 10 mg/kg in the second-line setting followed<br />

by BV-naive treatment extends survival. The dose <strong>of</strong> BV 5 mg/kg could be<br />

lower than the recommended dose in the second-line CRC treatment. Thus<br />

we planned this second line phase III study (EAGLE study) to evaluate the<br />

appropriate dose <strong>of</strong> BV (5 or 10 mg/kg) with FOLFIRI in patients with mCRC<br />

who have failed BV plus oxaliplatin-based first line regimen. Methods:<br />

EAGLE is a multicenter randomized phase III study <strong>of</strong> FOLFIRI plus BV 5<br />

mg/kg versus 10 mg/kg in mCRC who have failed BV plus oxaliplatin-based<br />

first line regimen (UMIN000002557).The primary endpoint is PFS. The<br />

secondary endpoints are the toxicity, response rate, time to treatment<br />

failure, OS. Eligible patients were older than 20 years <strong>of</strong> age, had<br />

histologically proved CRC, ECOG performance status 0 or 1, adequate<br />

organ function, progression or difficult to continue after BV plus oxaliplatinbased<br />

first line regimen, more than four times administration <strong>of</strong> BV and<br />

oxaliplatin in the first-line. Patients were randomized 1:1 to FOLFIRI with<br />

BV 5mg/kg or 10 mg/kg.The planned sample size was 370 patients to<br />

detect 30% risk reduction (median PFS assumed to be 5.0 months in arm<br />

A, 7.0 months in arm B) with 90% power assuming a two-sided significance<br />

level <strong>of</strong> 0.05, an accrual time <strong>of</strong> 2.5 years and a follow-up time <strong>of</strong> 1<br />

year. Between September 2009 to January 2012, 387 patients were<br />

enrolled at 100 sites in Japan. This phase III study will answer the question<br />

<strong>of</strong> the appropriate dose <strong>of</strong> BV after progression on a BV-containing first-line<br />

regimen.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


LBA4000 Oral Abstract Session, Sat, 3:00 PM-6:00 PM<br />

A randomized, multicenter trial <strong>of</strong> epirubicin, oxaliplatin, and capecitabine<br />

(EOC) plus panitumumab in advanced esophagogastric cancer (REAL3).<br />

Presenting Author: Tom Samuel Waddell, Royal Marsden Hospital NHS<br />

Foundation Trust, London, United Kingdom<br />

The full, final text <strong>of</strong> this abstract will be available at<br />

abstract.asco.org at 12:01 AM (EDT) on Saturday, June 2,<br />

2012, and in the <strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong> online<br />

supplement to the June 20, 2012, issue <strong>of</strong> Journal <strong>of</strong><br />

<strong>Clinical</strong> Oncology. Onsite at the <strong>Meeting</strong>, this abstract will<br />

be printed in the Saturday edition <strong>of</strong> ASCO Daily News.<br />

4002 Oral Abstract Session, Sat, 3:00 PM-6:00 PM<br />

Randomized phase III study <strong>of</strong> irinotecan (CPT-11) versus weekly paclitaxel<br />

(wPTX) for advanced gastric cancer (AGC) refractory to combination<br />

chemotherapy (CT) <strong>of</strong> fluoropyrimidine plus platinum (FP): WJOG4007<br />

trial. Presenting Author: Shinya Ueda, Kinki University School <strong>of</strong> Medicine,<br />

Osakasayama, Japan<br />

Background: A combination CT <strong>of</strong> FP has been regarded as the standard<br />

first-line treatment for AGC. Although two randomized trials showed a<br />

survival benefit <strong>of</strong> second-line CT (CPT-11 or docetaxel) compared with<br />

best supportive care, no standard regimen has been established. In Japan,<br />

wPTX has been used more frequently than docetaxel as the second-line CT.<br />

The objective <strong>of</strong> this study was to compare CPT-11 with wPTX in patients<br />

(pts) with AGC refractory to FP. Methods: Patients with AGC refractory to the<br />

first-line FP regimen were randomized 1:1 to either CPT-11 (150 mg/m2 ,<br />

q2w) or wPTX (80 mg/m2 , days 1, 8, 15, q4w). The primary endpoint was<br />

overall survival (OS) and secondary endpoints were progression-free survival<br />

(PFS), overall response rate (ORR), adverse events and receiving rates<br />

<strong>of</strong> third-line CT. To demonstrate an increase in median OS from 5 months<br />

(wPTX) to 7.5 months (CPT-11) with 2-sided alpha 5% and 80% power,<br />

220 pts were required. Results: Between Aug 2007 and Aug 2010, 223 pts<br />

were enrolled; 112 pts were randomized to CPT-11 and 111 pts to wPTX.<br />

Baseline characteristics were well balanced between arms. Median OS was<br />

8.4 months for CPT-11 and 9.5 months for wPTX (HR 1.132; 95% CI,<br />

0.86-1.49; p�0.38). Median PFS was 2.3 months for CPT-11 and 3.6<br />

months for wPTX (HR 1.14; 95% CI, 0.88-1.49; p�0.33). The ORR was<br />

13.6% (12/88) for CPT-11 and 20.9% (19/91) for wPTX (p�0.20). The<br />

most common grade 3/4 adverse events were neutropenia (39.1% for<br />

CPT-11 vs. 28.7% for wPTX), anemia (30.0% vs. 21.3%), anorexia<br />

(17.3% vs. 7.4%) and fatigue (12.7% vs. 6.5%). Four (4%) CPT-11 and<br />

three (3%) wPTX recipients died within 30 days after the last administration.<br />

Subsequent CT was performed in 80 pts (71%) for CPT-11 and 97 pts<br />

(89%) for wPTX. Seventy-five pts (67%) in the CPT-11 group and 87 pts<br />

(80%) in the wPTX group received the crossover CT. Conclusions: The<br />

WJOG4007 trial, the first phase III study comparing second-line CT<br />

regimens for AGC, did not demonstrate the superiority <strong>of</strong> CPT-11 over<br />

wPTX. Thus, wPTX can be adopted as a control arm <strong>of</strong> future phase III trials<br />

<strong>of</strong> second-line CT for AGC.<br />

Gastrointestinal (Noncolorectal) Cancer<br />

239s<br />

LBA4001 Oral Abstract Session, Sat, 3:00 PM-6:00 PM<br />

Intergroup Trial <strong>of</strong> Adjuvant Chemotherapy in Adenocarcinoma <strong>of</strong> the<br />

Stomach (ITACA-S) trial: Comparison <strong>of</strong> a sequential treatment with<br />

irinotecan (CPT-11) plus 5-fluorouracil (5-FU)/folinic acid (LV) followed by<br />

docetaxel and cisplatin versus a 5-FU/LV regimen as postoperative treatment<br />

for radically resected gastric cancer. Presenting Author: Emilio<br />

Bajetta, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy<br />

The full, final text <strong>of</strong> this abstract will be available at<br />

abstract.asco.org at 12:01 AM (EDT) on Saturday, June 2,<br />

2012, and in the <strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong> online<br />

supplement to the June 20, 2012, issue <strong>of</strong> Journal <strong>of</strong><br />

<strong>Clinical</strong> Oncology. Onsite at the <strong>Meeting</strong>, this abstract will<br />

be printed in the Saturday edition <strong>of</strong> ASCO Daily News.<br />

LBA4003 Oral Abstract Session, Sat, 3:00 PM-6:00 PM<br />

Phase III randomized trial <strong>of</strong> definitive chemoradiotherapy (CRT) with<br />

FOLFOX or cisplatin and fluorouracil in esophageal cancer (EC): Final<br />

results <strong>of</strong> the PRODIGE 5/ACCORD 17 trial. Presenting Author: Thierry<br />

Conroy, Centre Alexis Vautrin, Vandoeuvre-lès-Nancy, France<br />

The full, final text <strong>of</strong> this abstract will be available at<br />

abstract.asco.org at 12:01 AM (EDT) on Saturday, June 2,<br />

2012, and in the <strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong> online<br />

supplement to the June 20, 2012, issue <strong>of</strong> Journal <strong>of</strong><br />

<strong>Clinical</strong> Oncology. Onsite at the <strong>Meeting</strong>, this abstract will<br />

be printed in the Saturday edition <strong>of</strong> ASCO Daily News.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


240s Gastrointestinal (Noncolorectal) Cancer<br />

4004 Oral Abstract Session, Sat, 3:00 PM-6:00 PM<br />

Optimum time to assess complete clinical response (CR) following chemoradiation<br />

(CRT) using mitomycin (MMC) or cisplatin (CisP), with or without<br />

maintenance CisP/5FU in squamous cell carcinoma <strong>of</strong> the anus: Results <strong>of</strong><br />

ACT II. Presenting Author: Robert Glynne-Jones, Mount Vernon Centre for<br />

Cancer Treatment, Middlesex, United Kingdom<br />

Background: In the ACT I trial, relapse-free survival was improved with CRT<br />

compared to RT alone, but the CR rate at 6 weeks was similar (39% CRT,<br />

30% RT alone; p�0.08). Most studies assessed CR between 6 and 12<br />

weeks following completion <strong>of</strong> CRT. We investigated the association<br />

between observation <strong>of</strong> CR at 3 different time-points and progression-free<br />

and overall survival (PFS, OS), to determine the optimal time to assess this<br />

early endpoint. Methods: ACT II recruited 940 pts between 2001 and<br />

2008. It compared, in a factorial design, CisP versus MMC when combined<br />

with 5-FU CRT, and two cycles <strong>of</strong> maintenance chemotherapy versus no<br />

maintenance. CR (complete absence <strong>of</strong> tumour and node negative) was<br />

assessed by rectal exam or imaging at 11, 18 and mandated CT at 26 weeks<br />

after the start <strong>of</strong> CRT. Median follow-up was 5 years. Pts were excluded<br />

from analysis if assessment was not done/ missing at one or more time<br />

points (n�245). Data from 695 pts were analysed. Results: Pt characteristics<br />

(all 940) median age 58 years; tumour site – canal (84%), margin<br />

(14%); stage T1-T2 (52%), T3-T4 (46%); N� (32%), N0 (62%). CisP did<br />

not improve OS (HR: 0.96, p�0.89) or PFS (HR: 0.85, p�0.43) compared<br />

to MMC. This is consistent with the lack <strong>of</strong> an absolute difference at 18 and<br />

26 weeks, but not at 11 weeks (where a difference was found). CR patients<br />

(compared to not-CR) had a significantly lower risk <strong>of</strong> progression/death.<br />

The association between these outcomes and response was strongest at 26<br />

weeks. 202/695 (29%) pts not in CR at 11 weeks were CR at 26 weeks.<br />

Conclusions: 29% <strong>of</strong> pts not in CR at 11 weeks achieved CR at 26 weeks.<br />

Early surgical salvage would not have been appropriate for these pts. We<br />

recommend assessment at 26 weeks in future trials.<br />

Absolute risk<br />

HR (95% CI)<br />

difference (95% CI)<br />

(CR vs not-CR)<br />

Pts with CR CR rate % MMC CisP PFS OS<br />

Week 11 429 65.6 57.9 7.7% (0.52, 14.9) 0.74 (0.56, 0.97) 0.68 (0.48, 0.97)<br />

p�0.04<br />

p�0.03<br />

p�0.03<br />

Week 18 527 75.4 76.2 0.8% (-7.2, �5.6) 0.54 (0.40, 0.73) 0.44 (0.31, 0.64)<br />

p�0.81<br />

p�0.001<br />

p�0.001<br />

Week 26 582 83.5 84.0 0.5% (-5.9, �5.1) 0.26 (0.19, 0.35) 0.23 (0.16, 0.33)<br />

p�0.88<br />

p�0.001<br />

p�0.001<br />

4006 Oral Abstract Session, Sat, 3:00 PM-6:00 PM<br />

Tivantinib (ARQ 197) versus placebo in patients (Pts) with hepatocellular<br />

carcinoma (HCC) who failed one systemic therapy: Results <strong>of</strong> a randomized<br />

controlled phase II trial (RCT). Presenting Author: Lorenza Rimassa,<br />

Department <strong>of</strong> Oncology, Humanitas Cancer Center, Rozzano, Italy<br />

Background: Tivantinib (T) is a selective, oral inhibitor <strong>of</strong> c-Met, the tyrosine<br />

kinase receptor for hepatocyte growth factor involved in tumor cell<br />

migration, invasion, proliferation and angiogenesis. T has shown promising<br />

results in HCC in phase 1 studies as monotherapy and in combination with<br />

sorafenib. Methods: This multi-center RCT, enrolled pts with unresectable<br />

HCC, 1 failed systemic therapy, ECOG PS �2. Child-Pugh B-C were<br />

excluded. Pts were randomized 2:1 to oral T {360 mg bid (A), 240 mg bid<br />

(B)} or placebo (P), stratifying by PS and vascular invasion (VI). Treatment<br />

continued until disease progression (PD) or unacceptable toxicity. RECIST<br />

1.1 response was evaluated by CT / MRI every 6 weeks. Crossover to<br />

open-label T was allowed after PD. Primary endpoint was time to tumor<br />

progression (TTP) in the intent-to-treat (ITT) population by central radiology<br />

review. Other endpoints included disease control rate (DCR), PFS, OS,<br />

efficacy in Met� (Met � 2� in �50% <strong>of</strong> tumor at immunohistochemistry)<br />

pts, safety. Results: Characteristics <strong>of</strong> the 107 enrolled HCC pts (A� 38,<br />

B� 33, P� 36) in T/P: 58/28 male; median age 70/68; PS 0 41/21; VI<br />

22/13; Met� 22/15; Met- 27/13. Dose A was reduced to B in all pts due to<br />

G�3 neutropenia (NEUT) rate. Major TTP, DCR and PFS benefits were<br />

obtained in Met� pts, with preliminary OS trend favoring T (HR�0.47) and<br />

no detrimental effect in Met- pts. DCR (95% CI) in T/P was 44 (31-56) /<br />

31(16-48)% for ITT and 50 (28-72) / 20 (4-48)% in Met� pts. Most<br />

common AEs in T were asthenia (26.8%), NEUT (25.4%), low appetite<br />

(25.4%); most common drug-related AEs were NEUT (25.4%), anemia<br />

(15.5%). Most frequent drug-related serious AE was neutropenic sepsis<br />

(4.2%). Efficacy was similar inA/Bwith less frequent NEUT in B (21.1% /<br />

6.1%). Final OS, PK, biomarker data will be presented. Conclusions:<br />

Compared to P, T significantly benefited second-line HCC pts, especially if<br />

Met�, with manageable safety pr<strong>of</strong>ile at 240 mg BID.<br />

Median:<br />

TTP (mos)<br />

T arm P arm HR CI* Log-rank p value<br />

ITT 1.6 1.4 0.64 0.43-0.94 0.04<br />

Met �<br />

PFS (mos)<br />

2.9 1.5 0.43 0.19-0.97 0.03<br />

ITT 1.7 1.5 0.67 0.44-1.04 0.06<br />

Met � 2.4 1.5 0.45 0.21-0.95 0.02<br />

*90%CI for primary TTP. 95%CI for other endpoints.<br />

4005 Oral Abstract Session, Sat, 3:00 PM-6:00 PM<br />

Evaluation <strong>of</strong> MET pathway biomarkers in a phase II study <strong>of</strong> rilotumumab<br />

(R, AMG 102) or placebo (P) in combination with epirubicin, cisplatin, and<br />

capecitabine (ECX) in patients (pts) with locally advanced or metastatic<br />

gastric (G) or esophagogastric junction (EGJ) cancer. Presenting Author:<br />

Kelly S. Oliner, Amgen Inc., Thousand Oaks, CA<br />

Background: R is an investigational, fully human, monoclonal antibody to<br />

HGF/SF, the ligand <strong>of</strong> the MET receptor. A double-blind, P-controlled,<br />

phase 2 study randomized 121 G/EGJ cancer pts 1:1:1 to 15 mg/kg R �<br />

ECX (Arm A, n � 40), 7.5 mg/kg R � ECX (Arm B, n � 42), or P � ECX (Arm<br />

C, n � 39). OS and PFS improved with the addition <strong>of</strong> R to ECX (Iveson et<br />

al, European Multidisciplinary Cancer Congress 2011; abstr 6.504). High<br />

tumor MET levels have been associated with poor prognosis in G cancer<br />

(Nakajima et al, Cancer 1999;85:1894-1902). We explored MET pathway<br />

biomarkers to identify pts who may benefit from R. Methods: MET protein<br />

levels and gene copy numbers were measured in archival tumor samples by<br />

immunohistochemistry (IHC) and fluorescence in situ hybridization, respectively.<br />

High and low MET subgroups were defined by several predefined<br />

strategies. Total HGF and soluble MET (sMET) protein in plasma were<br />

measured by ELISA and MSD assays, respectively. Treatment and biomarker<br />

effects on OS and PFS were analyzed by Cox proportional hazard<br />

models and Kaplan-Meier estimates. Results: Tumor samples were evaluable<br />

for MET protein from 62 pts in Arms A � B and 28 pts in Arm C. Pts<br />

with METHigh tumors (� 50% tumor cells positive) in Arms A � B had<br />

improved median OS than those in Arm C (11.1 mo [80% CI: 9.2-13.3] vs<br />

5.7 mo [80% CI: 4.5-10.4]; HR � 0.29, 95% CI: 0.11-0.76, p � 0.012).<br />

Conversely, pts with METLow tumors (� 50% positive) in Arms A � B had a<br />

trend toward unfavorable OS compared with those in Arm C (HR � 1.84,<br />

95% CI: 0.78-4.34). In the chemotherapy only arm (Arm C), pts with<br />

METHigh tumors had poorer OS (HR � 3.22, 95% CI: 1.08-9.63) than pts<br />

with METLow tumors. Similar trends were seen with PFS. Predefined<br />

dichotomization schemes for tumor MET gene copy number and baseline<br />

plasma levels <strong>of</strong> total HGF or sMET did not correlate with OS or PFS.<br />

Conclusions: High MET expression by IHC may predict clinical benefit to R<br />

� ECX in G/EGJ cancer pts. High MET expression may also be associated<br />

with poor prognosis for ECX-treated pts. Further investigation is needed to<br />

confirm these findings.<br />

4007 Oral Abstract Session, Sat, 3:00 PM-6:00 PM<br />

Activity <strong>of</strong> cabozantinib (XL184) in hepatocellular carcinoma: Results from<br />

a phase II randomized discontinuation trial (RDT). Presenting Author: Chris<br />

Verslype, Hepatology, University Hospitals Gasthuisberg, Leuven, Belgium<br />

Background: Cabozantinib (cabo) is an oral, potent inhibitor <strong>of</strong> MET and<br />

VEGFR2. MET over-expression has been observed in advanced hepatocellular<br />

carcinoma (HCC). Anti-VEGF pathway agents have shown clinical<br />

benefit in pts w/ HCC. Simultaneous targeting <strong>of</strong> the MET and VEGF<br />

signaling pathways with cabo may therefore be a promising treatment<br />

strategy. Methods: Eligible HCC patients (pts) were required to have<br />

measurable disease per RECIST, � 1 prior systemic regimen and Child-<br />

Pugh score <strong>of</strong> A. Pts received cabo at 100 mg qd over a 12 wk Lead-in<br />

stage. Tumor response (mRECIST) was assessed q6 wks. Treatment � wk<br />

12 was based on response: pts with PR continued open-label cabo, pts with<br />

SD were randomized to cabo vs placebo, and pts with PD discontinued.<br />

Primary endpoint in the randomized phase was progression free survival<br />

(PFS). The primary endpoint was overall response rate (RR) per mRECIST<br />

in the Lead-in stage. Results: Enrollment has been completed (n � 41); all<br />

pts are unblinded. Median age: 61 years (33 to 83). Males: 76%; Asian:<br />

37%. HCC etiology: Hep B 24%; Hep C 22%; alcohol abuse 20%; other<br />

38%. Extra-hepatic spread observed in 70%. Median number <strong>of</strong> prior<br />

systemic treatments was 1; prior sorafenib was 51%. Median baseline AFP<br />

was 368 ng/mL (3 – 259,298); 86% had elevated AFP at baseline. Median<br />

follow-up was 5.5 mos (0.8 -18.5). 29 pts (71%) completed the Lead-in<br />

stage. Median PFS from Study Day 1 was 4.2 mos. 2/36 pts evaluable for<br />

tumor assessment at 12 weeks achieved a confirmed PR (cPR) by original<br />

RECIST (RR 5%). One more pt randomized at Week 12 achieved a cPR at<br />

18 weeks. 28/36 pts (78%) with �1 post-baseline scan had tumor<br />

regression (with no apparent relationship to prior sorafenib therapy). The<br />

overall disease control rate (DCR � PR�SD) at Week 12: was 68% (Asian<br />

subgroup: 73%). AFP responses (defined as reduction from baseline <strong>of</strong><br />

�50% in pts with elevated AFP at baseline) in 26 pts with �1 postbaseline<br />

result: 10/26 (38%). Most common Gr 3/4 AEs: diarrhea (17%),<br />

palmar-plantar erythrodyesthesia (15%), and thrombocytopenia (10%).<br />

Conclusions: Cabo treatment exhibits activity in HCC pts regardless <strong>of</strong> prior<br />

sorafenib treatment. The safety pr<strong>of</strong>ile was comparable to that <strong>of</strong> other<br />

VEGFR TKIs.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


4008 Oral Abstract Session, Sat, 3:00 PM-6:00 PM<br />

A randomized controlled phase II study <strong>of</strong> the prophylactic effect <strong>of</strong><br />

urea-based cream on the hand-foot skin reaction associated with sorafenib<br />

in advanced hepatocellular carcinoma. Presenting Author: Zhenggang Ren,<br />

Zhongshan Hospital, Fudan University, Shanghai, China<br />

Background: Sorafenib (SOR) has become the standard treatment for<br />

advanced hepatocellular carcinoma (HCC). Hand-foot skin reaction (HFSR)<br />

is one <strong>of</strong> the most common adverse events associated with SOR and its<br />

occurrence can impact patient quality <strong>of</strong> life and lead to dose modification<br />

or interruption, both <strong>of</strong> which may negatively impact clinical outcomes.<br />

This randomized controlled trial is the first large prospective study to<br />

investigate the prophylactic effect <strong>of</strong> urea-based creams on HFSR associated<br />

with SOR. Methods: Patients with advanced HCC treated with SOR<br />

were randomly assigned 1:1 to receive prophylactic urea-based cream (Arm<br />

A) or best supportive care (BSC) following development <strong>of</strong> HFSR (Arm B).<br />

SOR was administered 800 mg daily. Urea-based cream was given twice<br />

daily for up to 12 weeks starting on Day 1. BSC was at the physician’s<br />

discretion and excluded urea-based creams. The primary endpoint was the<br />

incidence <strong>of</strong> all-grade HFSR in the first12 weeks. Results: Eight hundred<br />

sixty eight patients were enrolled; 439 patients in Arm A and 432 patients<br />

in Arm B. There was no difference <strong>of</strong> baseline characteristics between two<br />

arms. Over the 12 week period <strong>of</strong> study, the incidence <strong>of</strong> all-grade HFSR<br />

was significantly lower in Arm A compared to Arm B; n�246 (56.0%)<br />

patients in Arm A versus n�318 (73.6%) patients in Arm B, p�0.0001.<br />

The incidence <strong>of</strong> grade �2 HFSR tended to be lower in Arm A compared to<br />

Arm B, but did not reach statistical significance; n�96 (21.9%) patients<br />

Arm A versus n�126 (29.2%) patients in Arm B, p�0.1638. The median<br />

time to the first HFSR event was 2.5 fold longer in Arm A compared to Arm<br />

B; 84 days (95% CI 45-93 days) in Arm A and 34 days (95% CI 29-43<br />

days) in Arm B (p�0.001). Conclusions: This is the first large prospective,<br />

randomized control trial examining the prophylactic use <strong>of</strong> urea-based<br />

creams for treatment <strong>of</strong> HFSR associated with a multikinase inhibitor.<br />

Compared to BSC, prophylactic topical use <strong>of</strong> a urea-based cream appears<br />

to be effective in preventing and/or delaying the incidence <strong>of</strong> HFSR<br />

associated with SOR treatment in patients with advanced HCC.<br />

4010 Poster Discussion Session (Board #2), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Assessment <strong>of</strong> HER2 gene amplification in adenocarcinomas <strong>of</strong> the<br />

stomach or gastroesophageal junction in the INT-0116/SWOG9008 clinical<br />

trial. Presenting Author: Michael Alexander Gordon, Keck School <strong>of</strong><br />

Medicine <strong>of</strong> the University <strong>of</strong> Southern California, Norris Comprehensive<br />

Cancer Center, Los Angeles, CA<br />

Background: Trastuzumab has been approved for treatment <strong>of</strong> patients with<br />

HER2-positive metastatic gastric carcinoma; however, relatively little is<br />

known about the role <strong>of</strong> HER2 in the natural history <strong>of</strong> this disease.<br />

Methods: Patients enrolled in the INT-0116/SWOG9008 phase III gastric<br />

cancer clinical trial (n�559) were retrospectively evaluated for HER2 gene<br />

amplification by fluorescence in situ hybridization (FISH)(n�258), overexpression<br />

by immunohistochemistry (IHC)(n�148) and HER2 amplification<br />

by silver-enhanced in situ hybridization (n�77) based on availability <strong>of</strong><br />

tissue specimens. The purpose <strong>of</strong> the original clinical trial was to evaluate<br />

the benefit <strong>of</strong> post-operative 5-fluorouracil/leucovorin plus radiation therapy<br />

compared to surgery alone. Results: HER2 gene amplification rate by FISH<br />

was 10.9% in tumor tissue from 258 patients evaluated. HER2 status<br />

determined by FISH was 92% concordant with SISH. HER2 overexpression<br />

rate by IHC was 12.2% among 148 patients evaluated, with 90%<br />

agreement between FISH and IHC. There was a significant interaction<br />

between HER2 status by FISH and treatment with respect to both OS<br />

(p�0.034) and DFS (p�0.020). Among patients with HER2 non-amplified<br />

cancers, treated patients had a median OS <strong>of</strong> 44 months compared to 24<br />

months for patients in the surgery only arm (34 and 17 months respectively<br />

for DFS, p�0.003). Among 28 patients with HER2 amplified cancers, the<br />

medians for OS were 16 months in the treated arm, and 22 months in the<br />

surgery arm (p�0.55) (13 and 11 months respectively for DFS, p�0.87).<br />

We were unable to detect a statistically significant treatment benefit in this<br />

small subset <strong>of</strong> patients with HER2 amplification. HER2 amplification<br />

status was not an independent prognostic marker <strong>of</strong> OS among patients<br />

who received no postoperative chemotherapy or radiation therapy (p�0.76).<br />

Conclusions: Patients lacking HER2 amplification responded significantly<br />

to treatment as indicated by both OS and DFS.<br />

Gastrointestinal (Noncolorectal) Cancer<br />

241s<br />

4009 Poster Discussion Session (Board #1), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Adverse prognostic impact <strong>of</strong> heterogeneous HER2 gene amplification in<br />

patients with esophageal adenocarcinoma (EAC). Presenting Author: Harry<br />

H. Yoon, Mayo Clinic, Rochester, MN<br />

Background: HER2 expression in upper digestive cancer is reported to be<br />

heterogeneous, which substantially affects interpretation <strong>of</strong> HER2 positivity<br />

in the clinic. Yet the frequency and prognostic impact <strong>of</strong> HER2 genetic<br />

heterogeneity and polysomy 17 (poly17) are unknown in EAC. Methods:<br />

HER2 amplification (fluorescence in situ hybridization) and protein expression<br />

were examined in untreated surgical EAC specimens (N � 661) at<br />

Mayo Clinic. HER2 genetic heterogeneity was defined per ASCO/CAP as<br />

amplification (HER2/CEP17 ratio � 2) in 5-50% <strong>of</strong> cancer cells; poly17<br />

refers to � 3 copies <strong>of</strong> chromosome 17. Most tumors were T3-4 (68%) or<br />

lymph node (LN)-positive (73%). Cox models were used to assess diseasespecific<br />

(DSS) and overall survival (OS). Results: HER2 amplification was<br />

detected in 117 <strong>of</strong> 661 EACs (18%), <strong>of</strong> which 20 (17%) showed HER2<br />

heterogeneity. HER2 heterogeneous tumors had a significantly higher<br />

frequency <strong>of</strong> poly17 and high tumor grade. HER2 heterogeneity by<br />

amplification vs expression were correlated. Since heterogeneity was<br />

limited to HER2-amplified tumors, survival analysis was stratified by<br />

amplification status. In multivariable analysis, only HER2 heterogeneity<br />

and metastatic LN number were prognostic (Table). Conclusions: Among<br />

HER2 amplified EACs, 17% show HER2 heterogeneity, which is associated<br />

with increased poly17 and independently predicts 2-fold higher risk <strong>of</strong><br />

cancer-specific death. Among HER2-nonamplified cases, poly17 is independently<br />

associated with worse survival. These novel findings demonstrate<br />

aggressive subgroups in HER2-amplified and -nonamplified EACs that have<br />

important implications for HER2 analysis and evaluation <strong>of</strong> benefit from<br />

HER2 targeted therapy.<br />

DSS<br />

Variable<br />

a OS a<br />

HR p HR p<br />

HER2-amplified N � 117<br />

Heterogeneity, yes vs no 2.0b 0.025 2.0c 0.026<br />

Grade, 4 v 1-3 1.1 0.7 1.1 0.8<br />

T stage, 3-4 v 1-2<br />

No. LN<br />

1.3 0.2 1.3 0.3<br />

d 1.1 �.001 1.4 �.001<br />

Poly17, yes v no<br />

HER2-nonamplified N � 544<br />

0.7 0.1 0.7 0.1<br />

Heterogeneity None<br />

Grade, 4 v 1-3 1.3 0.01 1.3 0.007<br />

T stage, 3-4 v 1-2<br />

No. LN<br />

2.0 �.001 1.9 �.001<br />

d 1.1 �.001 1.1 �.001<br />

Poly17, yes v no 1.4 0.012 1.3 0.023<br />

a Adjusted for all covariates; b 95% confidence interval (CI) 1.1 – 3.8; c 95% CI 1.1 – 3.7; d Per metastatic LN.<br />

HR, hazard ratio.<br />

4011 Poster Discussion Session (Board #3), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Pr<strong>of</strong>iling <strong>of</strong> activated receptor tyrosine kinases in advanced gastric cancers<br />

identifies patients with poor prognosis. Presenting Author: Phillip Sangwook<br />

Kim, Prometheus Laboratories Inc., San Diego, CA<br />

Background: Metastatic gastric cancer (GCA) remains a therapeutic challenge<br />

due to its poor prognosis. Trastuzumab is the only known targeted<br />

therapy that has demonstrated a survival benefit in a small subset <strong>of</strong><br />

metastatic GCAs. Addition <strong>of</strong> molecular diagnostics for predicting prognosis<br />

and therapeutic outcomes can significantly enhance the clinical<br />

management <strong>of</strong> GCAs. Herein, we identify activated receptor tyrosine<br />

kinases (RTKs) in distinct GCA subsets that correlate with disease-free<br />

survival post-curative GCA surgery. Methods: Fresh frozen GCA tissues from<br />

434 stage I to IV GCA patients were pr<strong>of</strong>iled for HER1, HER2, HER3,<br />

p95HER2, c-MET and IGF1R RTKs using the multiplexed Collaborative<br />

Enzyme Enhanced Reactive (CEER) immunoassay. CEER is a highly<br />

sensitive and specific proximity assay that relies on the formation <strong>of</strong> a triple<br />

antibody complex surrounding the target protein. Results: p95HER2, a<br />

known trastuzumab resistance mechanism, was identified for the first time<br />

in 79% <strong>of</strong> IHC/FISH-based HER2(�) GCAs. Full-length HER2 expression<br />

was heterogeneous with ~20% <strong>of</strong> HER2(-) GCAs still expressing the HER2<br />

protein. 232/434 GCAs expressed at least one activated RTK analyzed in<br />

our study. Of these 232 patients, 121 patients that presented with stage II<br />

or III disease at the time <strong>of</strong> surgery; demonstrated a worse progression-free<br />

survival as compared to those where none <strong>of</strong> the analyzed RTKs were<br />

activated (32.6 months vs 76.5 months, p�0.0318). HER axis members<br />

were the most commonly activated RTKs with 64% <strong>of</strong> such GCAs.<br />

Approximately 71% <strong>of</strong> activated cMET tumors demonstrated a coactivation<br />

with a HER axis member with a preference for HER1 in ~61% <strong>of</strong><br />

cMET-activated GCAs. Patients with p-MET(�):p-HER1(�) GCAs had a<br />

worse prognosis as compared to those with p-MET(�):p-HER1(-) GCAs<br />

(46.2 months vs 82.8 months, p�0.0184). Conclusions: CEER-based RTK<br />

characterization revealed concomitantly activated signaling pathways in<br />

GCAs which could predict disease recurrence. Comprehensive molecular<br />

pr<strong>of</strong>iles <strong>of</strong> GCA tumors can allow for the implementation <strong>of</strong> these companion<br />

biomarkers in GCA therapeutic clinical trials.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


242s Gastrointestinal (Noncolorectal) Cancer<br />

4012 Poster Discussion Session (Board #4), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Impact <strong>of</strong> insulin-like growth factor 1 receptor (IGF1R) and amphiregulin<br />

(AREG) expressions on survival in patients with stage II/III gastric cancer<br />

enrolled in the ACTS-GC study. Presenting Author: Wataru Ichikawa,<br />

National Defense Medical College, Tokorozawa, Japan<br />

Background: Exploratory biomarker analysis was conducted to identify<br />

factors related to outcomes <strong>of</strong> patients enrolled in the ACTS-GC study, a<br />

randomized controlled trial comparing adjuvant S-1 administration with<br />

surgery alone in 1,059 patients with stage II/III gastric cancer. Methods:<br />

Formalin-fixed paraffin-embedded surgical specimens were retrospectively<br />

examined in 829 patients (78.3%), and 63 genes involved in pyrimidine<br />

metabolic pathway, growth factor signaling pathway, apoptosis, DNA<br />

repair, etc., were analyzed by quantitative real-time RT-PCR after TaqMan<br />

assay-based pre-amplification. Gene expression levels were normalized to<br />

the geometric mean expressions <strong>of</strong> GAPDH, ACTB, and RPLP0, used as<br />

reference genes. The expression <strong>of</strong> each gene was categorized into low and<br />

high values at those median. The impacts <strong>of</strong> gene expression on survival<br />

were analyzed using the 5-year survival data <strong>of</strong> the ACTS-GC. The<br />

Benjamini and Hochberg procedure was used to control the false discovery<br />

rate (FDR). Results: Among 63 screened genes, IGF1R and AREG most<br />

strongly correlated with overall survival (OS), with FDR <strong>of</strong> 0.0048 and<br />

0.018, respectively. OS was significantly worse in IGF1R high patients<br />

than in IGF1R low patients, but better in AREG high patients than in AREG<br />

low patients. The hazard ratio (HR) for death in the analysis <strong>of</strong> OS (S-1 vs.<br />

surgery alone) was lower in the high IGF1R group (HR, 0.55; 95%CI,<br />

0.40-0.76) than in the low IGF1R group (HR, 0.72; 95%CI, 0.49-1.06).<br />

Likewise, the HR for death in the analysis <strong>of</strong> OS (S-1 vs. surgery alone) was<br />

much smaller in the low AREG group (HR, 0.57; 95%CI, 0.41-0.79) than<br />

in the high AREG group (HR, 0.74; 95%CI, 0.51-1.08). Interactions were<br />

not statistically significant. Conclusions: IGFR1 gene expression was<br />

associated with poor outcomes after curative resection <strong>of</strong> stage II/III gastric<br />

cancer, whereas AREG gene expression was associated with good outcomes.<br />

No interaction for survival was evident between S-1 and these gene<br />

expressions.<br />

4014 Poster Discussion Session (Board #6), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Multivariate analysis including biomarkers in the phase III RADIANT-2<br />

study <strong>of</strong> octreotide LAR plus everolimus (E�O) or placebo (P�O) among<br />

patients with advanced neuroendocrine tumors (NET). Presenting Author:<br />

James C. Yao, University <strong>of</strong> Texas M. D. Anderson Cancer Center, Houston,<br />

TX<br />

Background: In this large phase III trial, median progression-free survival<br />

(PFS) improved by 5.1 mo with E�O compared to P�O in patients (pts)<br />

with NET associated with carcinoid syndrome. Baseline imbalances including<br />

WHO performance status (PS) and primary site favoring P�O confounded<br />

primary analysis. Chromogranin A (CgA) and 5-hydroxyindoleacetic<br />

acid (5-HIAA) are important biomarkers in NET. Analyses were performed<br />

to identify prognostic factors and adjust for baseline imbalances. Methods:<br />

Pts were randomized to E�O (n�216) or P�O (n�213). Potential<br />

prognostic factors including baseline CgA (�2�ULN vs �2�ULN), baseline<br />

5-HIAA (�median vs �median at baseline), age (�65 vs �65),<br />

gender, race, WHO PS (0 vs 1, 2), primary site (lung vs other), prior<br />

somatostatin analog use (yes vs no), duration from diagnosis (�6 mo, 6-24<br />

mo, 2-5 yr, �5 yr), and organs involved (liver, bone) were assessed in<br />

univariate analysis using the log rank test and stepwise regression using<br />

Cox proportional hazards model. Results: Median PFS (mo) was significantly<br />

longer for pts with nonelevated CgA (27 vs 11; p�.001) and nonelevated<br />

5-HIAA (17 vs 11; p�.001). Analyses also indicated age (14 vs 12;<br />

p�.01), WHO PS (17 vs 11; p�.004), liver involvement (14 vs not<br />

reached; p�.02), bone metastases (8 vs 15; p�.001), and lung as primary<br />

site (11 vs 14; p�.06) as potentially prognostic. Multivariate analysis<br />

indicated that significant prognostic factors for PFS included baseline CgA<br />

(HR, 0.47; CI, 0.34-0.65; p�.001), WHO PS (HR, 0.69; CI, 0.52-0.90;<br />

p�.006), bone involvement (HR, 1.52; CI, 1.06-2.18; p�.02), and lung<br />

as primary site (HR, 1.55; CI, 1.01-2.36; p�.04). Adjusted for covariates,<br />

a 38% reduction in risk <strong>of</strong> progression was observed for E�O (HR, 0.62;<br />

95% CI, 0.51-0.87; p�.003). Conclusions: In the phase III RADIANT-2<br />

trial, baseline CgA levels, WHO PS, lung as primary site, and bone<br />

involvement were important prognostic factors. Exploratory analysis adjusted<br />

for these prognostic factors indicated significant benefit <strong>of</strong> everolimus<br />

therapy.<br />

4013 Poster Discussion Session (Board #5), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Actionable targets in pancreatic cancer detected by immunohistochemistry<br />

(IHC), microarray (MA) fluorescent in situ hybridization (FISH), and<br />

mutational analysis. Presenting Author: Daniel D. Von H<strong>of</strong>f, Virginia G.<br />

Piper Cancer Center at Scottsdale Healthcare/TGen, Scottsdale, AZ<br />

Background: A great need exists for new therapeutic approaches for patients<br />

with pancreatic cancer. Methods: The study cohort included 1029 patients<br />

analyzedfor a.) up to 29 different immunohistochemical biomarkers – (e.g.,<br />

COX-2, MGMT, PGP, RRM1, TOPOI, TOPOII, SPARC etc.)b.); in up to 450<br />

patients’ specimens a whole genome expression analysis was performed<br />

using HumanHT-12 v4 beadChips ( Illumina Inc.,San Diego, CA) c.) in up<br />

to 695 patients FISH for c-Myc, EGFR, HER2 and TOPO2A gene copy<br />

amplifications; and d.) in up to 783 patients sequencing for KRAS, EGFR,<br />

PI3CA and BRAF, was performed. Results: IHC identified actionable targets<br />

included; 74% high COX-2; 57% negative ERCC1; 8% negative MGMT;<br />

22% negative MRP1; 47% negative PGP; 77% low RRM1, 44% high<br />

SPARC; 30% high TOPO2A; 61% high TOPOI and 73% negative TS. Other<br />

biologically important findings by IHC for possible new therapeutics<br />

included 27% negative PTEN; and 20% high PDGFR. Microarray results<br />

presented multiple overexpressed targets for consideration including 36%<br />

<strong>of</strong> specimens with overexpressed adenosine deaminase; 28% asparagine<br />

synthase; 17% BCL2; 20% survivin; 23% carboxylesterase; 67% DNMT1;<br />

40% thymidine phosphatase; 49% EPHA2 (and others in the src family <strong>of</strong><br />

kinases); 57% FOLR2; 41% HDAC1; 62% HiF1�; 23% IL2RA (CD25);<br />

46% NFkB1; 48% OGFR; 32% RARA; 26% VEGFR; and 43% vitamin D<br />

receptor. FISH yielded 2% amplified EGFR and 10% amplified Her2neu.<br />

Sequencing noted 73% mutated KRAS and 3% mutated PIK3CA.<br />

Conclusions: Examining actionable targets in patients’ pancreatic cancers<br />

(a)reiterates the commonality and importance <strong>of</strong> KRAS mutations in this<br />

disease (needs renewed targeting effort) (b)suggests that TOPO2 inhibitors<br />

(particularly if transport into tumor can be improved) should be examined<br />

in this disease (c)suggests other pathways to target including DNA repair,<br />

epigenetic, Src and inflammation (d) suggests protein turnover, amino acid<br />

targets and folate receptor2 as fresh areas to explore against the disease.<br />

Supported in part by a Stand Up To Cancer Dream Team Award.<br />

4015 Poster Discussion Session (Board #7), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Predictive and prognostic factors for treatment and survival in 305 patients<br />

with advanced gastrointestinal poorly differentiated neuroendocrine carcinoma:<br />

The NORDIC NEC study. Presenting Author: Halfdan Sorbye,<br />

Haukeland University Hospital, Bergen, Norway<br />

Background: Gastrointestinal poorly differentiated neuroendocrine carcinoma<br />

(GI-NEC) are aggressive tumors with Ki-67�20% and usually<br />

metastatic at diagnosis. Knowledge about GI-NEC is limited. We retrospectively<br />

reviewed clinical data to identify predictive and prognostic markers<br />

for advanced GI-NEC patients. Methods: Epidemiological, biochemical,<br />

histopathological, treatment and survival data were registered for advanced<br />

GI-NEC patients diagnosed during 2000-2009 at 12 Nordic university<br />

hospitals. Results: 305 patients were included. Palliative chemotherapy<br />

was given to 252 patients, median survival was 11 months. Response rate<br />

to 1st-line chemotherapy was 31%, 33% had stable disease. Ki-67�55%<br />

was by ROC analyses the best cut-<strong>of</strong>f value concerning correlation to<br />

response rate. Response rate to platinum-based chemotherapy was lower in<br />

patients with Ki-67�55% (14% vs.44%, p�0.001). Response rate for 84<br />

patients given 2nd-line chemotherapy was 18%, whereas 33% achieved<br />

SD. The most important negative prognostic factors for survival were poor<br />

performance status, primary colorectal tumors, and elevated baseline<br />

platelets or lactate dehydrogenase (LDH) levels. Patients with Ki-67�55%<br />

had longer median survival (15 months) than patients with Ki-67�55%<br />

(10 months) (p�0.001). Survival and response rates did not differ between<br />

the different platinum chemotherapy schedules (cisplatin-based vs. carboplatin-based)<br />

or morphology subtypes. 53 patients received best supportive<br />

care only with a median survival <strong>of</strong> 1 month. Conclusions: This is, to our<br />

knowledge, the largest study reporting patient and tumor characteristics,<br />

treatment and survival in advanced GI-NEC. Performance status, location<br />

<strong>of</strong> primary tumor and blood levels <strong>of</strong> platelets and LDH were the strongest<br />

prognostic factors for survival. Patients with Ki-67�55% had significantly<br />

longer survival than patients with higher Ki-67, but were less responsive to<br />

platinum-based chemotherapy. Our data indicate that to consider all<br />

GI-NEC as one single disease entity may not be correct.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


4017 Poster Discussion Session (Board #9), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Maintenance sunitinib (MS) or observation (O) in metastatic pancreatic<br />

adenocarcinoma (MPA): <strong>Clinical</strong> and translational results <strong>of</strong> a phase II<br />

randomized trial (NCT00967603). Presenting Author: Michele Reni, San<br />

Raffaele Scientific Institute, Milan, Italy<br />

Background: Combination chemotherapy (CT) prolonged progression-free<br />

survival at 6 months (PFS6) <strong>of</strong> patients (pts) with MPA from �20% with<br />

gemcitabine (GEM) to �50%. To prolong CT over 6 months has unproven<br />

benefit and is hampered by cumulative toxicity. This open-label, randomised,<br />

multi-centre phase II trial explored the role <strong>of</strong> MS in pts with MPA<br />

without progressive disease (PD) after CT, using an O group as calibration<br />

arm. The role <strong>of</strong> circulating endothelial cells (CEC) as biomarker <strong>of</strong> MS<br />

activity was explored. Methods: Adult pts with pathologic diagnosis <strong>of</strong> MPA,<br />

performance status (PS) �50%, without radiological PD or CA19.9 raise<br />

�20% after 6 months <strong>of</strong> CT were stratified based on PS and previous CT<br />

and randomized to O (arm A) or MS at 37.5 mg daily until PD or a maximum<br />

<strong>of</strong> 6 months (arm B). Primary endpoint was PFS6. Sample size (H0 10%;<br />

H1 30%; a .10; b .10) was 26 pts per arm and MS had to be considered <strong>of</strong><br />

interest with �5 pts PFS6. CEC were evaluated by flow cyt<strong>of</strong>luorometry on a<br />

baseline peripheral blood sample. Results: 56 pts (29 arm A; 27 arm B)<br />

were enrolled. One arm B pt had kidney cancer and was excluded. Pts<br />

characteristics were (A/B): median age 64/61 years; median PS 90/100;<br />

median CA19.9 45/32; previous CT: GEM 3/3; combination CT 26/22.<br />

Main grade 3-4 toxicity in arm B was 15% neutropenia; 12% thrombocytopenia<br />

and hand-foot syndrome, 8% diarrhea. Second line CT was given to<br />

76% <strong>of</strong> pts in both arms. One arm A (3%) and 6 arm B pts (23%; p�.01)<br />

were PFS6; 2y overall survival (OS) was 4% and 25% (p�.09). Blood<br />

sample for CEC analysis was available for 46 <strong>of</strong> 55 pts (84%). In arm A,<br />

median PFS was longer for pts with CEC number �30 (lower terzile group)<br />

when compared to �30 (2.9 versus 1.9 months; p.08). MS significantly<br />

increased PFS in CEC �30 group (3.4 months; p�.01). No PFS difference<br />

between arms was observed in �30 group (2.3 months). Conclusions: This<br />

is the first randomized trial on maintenance therapy in MPA. In arm B, the<br />

primary endpoint was fulfilled and 2y OS was remarkably high, suggesting<br />

that MS may have a role in pts with MPA. The number <strong>of</strong> CEC may be useful<br />

to predict benefit from MS.<br />

4019 Poster Discussion Session (Board #11), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Dual blockade <strong>of</strong> epidermal growth factor receptor (EGFR) and insulin-like<br />

growth factor receptor-1 (IGF-1R) signaling in metastatic pancreatic<br />

cancer: Phase I/randomized phase II trial <strong>of</strong> gemcitabine, erlotinib, and<br />

cixutumumab versus gemcitabine plus erlotinib (SWOG-0727). Presenting<br />

Author: Philip Agop Philip, Karmanos Cancer Institute, Wayne State<br />

University, Detroit, MI<br />

Background: Targeting a single pathway in pancreatic adenocarcinoma<br />

(PAC) is unlikely to impact its natural history. EGFR targeting with erlotinib<br />

added to gemcitabine (G) marginally improved the outcome <strong>of</strong> patients with<br />

advanced disease. We tested the hypothesis that if the EGFR and IGF-1R<br />

pathways are simultaneously blocked then progression free survival (PFS)<br />

would be significantly improved by abrogating reciprocal signaling that<br />

leads to resistance to either drug. Methods: This was a phase I/randomized<br />

phase II (RP2) study testing the anti-IGF-1R monoclonal antibody cixutumumab<br />

combined with erlotinib and G in patients with metastatic PAC. The<br />

control arm was erlotinib plus G. The primary endpoint <strong>of</strong> the RP2 portion<br />

<strong>of</strong> the study was PFS. Eligibility included patients with untreated metastatic<br />

disease, performance status 0/1, fasting blood glucose less than<br />

institutional upper limit <strong>of</strong> normal, and willingness to provide tissue and<br />

blood specimens for correlative studies. Results: In phase I portion (n�10)<br />

safety <strong>of</strong> cixutumumab 6 mg/kg IV/wk, erlotinib 100 mg/day orally and G<br />

1000 mg/m2 IV D 1, 8, and 15 <strong>of</strong> a 28-day cycle was established. In the<br />

RP2 portion (n�124) 114 eligible patients (median age 63) were included.<br />

On the cixutumumab arm, 59% <strong>of</strong> patients were female vs. 40% on<br />

control arm. Median PFS and overall survival were 3.7 and 6.7 months,<br />

respectively, in both arms <strong>of</strong> the study. Major grade 3 and 4 toxicities were<br />

(cixutumumab/control) elevation <strong>of</strong> transaminases (12%/6%), fatigue<br />

(16%/12%), gastrointestinal (35%/28%), neutropenia (21%/10%), and<br />

thrombocytopenia (16%/7%). Grade 3/4 hyperglycemia was seen in 16%<br />

<strong>of</strong> patients on cixutumumab. Grade 3 or 4 kin toxicity was similar in both<br />

arms <strong>of</strong> the study (� 5%). Five treatment-related deaths were reported: 2<br />

cardiac on each arm, 1 pulmonary on cixutumumab. Conclusions: IGF-1R<br />

inhibitor cixutumumab did not improve the progression free or overall<br />

survival <strong>of</strong> patients with metastatic PAC treated with erlotinib and G in a<br />

molecularly unselected population.<br />

Gastrointestinal (Noncolorectal) Cancer<br />

243s<br />

4018 Poster Discussion Session (Board #10), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

FOLFIRI.3 (CPT-11 plus folinic acid plus 5-FU) alternating with gemcitabine<br />

or gemcitabine (G) alone in patients (pts) with previously untreated<br />

metastatic pancreatic adenocarcinoma (MPA): Results <strong>of</strong> the randomized<br />

multicenter AGEO phase II trial FIRGEM. Presenting Author: Isabelle<br />

Trouilloud, Hôpital Européen Georges Pompidou, Paris, France<br />

Background: CPT-11 showed modest activity and tolerability in MPA. We<br />

developed a new strategy to improve efficacy and tolerability <strong>of</strong> CPT-11<br />

based regimen in MPA pts. Methods: Chemotherapy-naive pts with histologically<br />

proven MPA, bilirubin levels � 1.5 ULN and performance status (PS)<br />

0-1 were randomized in a phase II trial to receive either FOLFIRI.3 (CPT-11<br />

90 mg/m2 as a 60-min infusion on day (D) 1, leucovorin 400 mg/m2 as a<br />

2-hr infusion on day 1, followed by 5-FU 2000 mg/m2 as a 46-hr infusion<br />

and CPT-11 90 mg/m2 , repeated on D3, at the end <strong>of</strong> the 5-FU infusion,<br />

every 2 weeks) for 2 months alternarting with G (1000 mg/m² at a fixed<br />

dose rate <strong>of</strong> 10 mg/m²/min on D1, D8, D15, D29, D36 and D43) for 2<br />

months (arm A) or G alone (arm B). Using Fleming design the primary end<br />

point was rate <strong>of</strong> progression free survival (PFS) at 6 months from (H0)<br />

25% over (H1) 45% needing to include 49 pts/arm. Results: Between 2007<br />

and 2011, 98 pts were enrolled (males: 59, median age: 62 years, PS 0:<br />

32%). Median follow-up was 23 months. Grade 3-4 toxicities per pts (%) in<br />

arm A/B were diarrhea 13/0, nausea-vomiting 11/4, neutropenia 51/25<br />

and febrile neutropenia 4/0. No toxic death occurred. Response rate were<br />

40 and 11% as disease control rate (CR�PR�SD) were 73 and 52% in arm<br />

A and B, respectively. The primary endpoint <strong>of</strong> the trial was met with rate <strong>of</strong><br />

PFS at 6 months <strong>of</strong> 48% (95% CI: 33-63) in arm A while in arm B PFS was<br />

30% (95% CI: 17 - 44). One year PFS was 23% (95%CI: 11.5-36) and<br />

11% (95%CI: 4-21), respectively. Conclusions: FIRGEM strategy is feasible<br />

and efficient with a manageable toxicity pr<strong>of</strong>ile in good condition pts<br />

with MPA. A phase III trial comparing this strategy with the FOLFIRINOX<br />

triplet therapy focusing on quality <strong>of</strong> life should now be performed.<br />

4020 Poster Discussion Session (Board #12), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Long-term survival in patients with pancreatic cancer: Relevant factors in<br />

CONKO-001. Presenting Author: Marianne Sinn, Medical Department,<br />

Division <strong>of</strong> Hematology, Oncology and Tumor Immunology Charité -<br />

Universitätsmedizin Berlin, Berlin, Germany<br />

Background: Long-term survival (LTS) in patients with pancreatic cancer is<br />

still rare, even in resectable and potentially curative stages. Few prospective<br />

data are available to identify predictive factors. The CONKO-001 study<br />

establishing adjuvant gemcitabine (GEM) may provide data to answer this<br />

question. Methods: CONKO-001 patients (pts) with an overall survival � 5<br />

years were included in this analysis and compared to those with � 5 years.<br />

Central re-evaluation <strong>of</strong> the primary histology was done to confirm the<br />

diagnosis <strong>of</strong> pancreatic adenocarcinoma. Univariate analysis with the<br />

x²-test identified qualifying factors (p�0.10). Logistic regression with a<br />

stepwise selection process was used to investigate the influence <strong>of</strong> these<br />

covariates on LTS. Results: Of the 354 pts included in the intention-to-treat<br />

analysis <strong>of</strong> CONKO-001, 53 (15%) pts with an overall survival <strong>of</strong> more than<br />

5 years could be identified, for 39 (74%) tumor specimens could be<br />

obtained. In 38 (97% <strong>of</strong> pts with LTS) the diagnosis <strong>of</strong> adenocarcinoma<br />

was confirmed, 1 showed a high-grade neuroendocrine tumor. Relevant<br />

factors for all 53 pts with LTS compared to remaining 301 non-LTS pts in<br />

univariate analysis were active treatment (GEM) (68% in LTS pts vs 48% in<br />

non-LTS pts; p�0.006), tumor grading (G1 17% vs 3%, G2 64% vs 55%,<br />

G3 17% vs 40%; p�0.000), tumor-size (T2 15% vs 9%, T3 74% vs 84%;<br />

p�0.004) and lymph nodes (N0 47% vs 25% N1 53% vs 74%; p�0.003.<br />

Significance could not be demonstrated for resection margin (R0 vs R1),<br />

sex, age, Karn<strong>of</strong>sky performance status (�80% vs 80% vs �80%) and CA<br />

19-9 (40-100 U/ml vs �40 U/ml) at study entry. In the multivariate<br />

analysis tumor grading (gr) (odds ratio gr 3 vs gr 1�0.07; gr 3 vs gr 2�<br />

0.38; p�0.017) and active treatment (odds ratio GEM vs observation�0.38;<br />

p�0.004) were the only independent prognostic factors.<br />

Conclusions: Long-term survival can be achieved in adenocarcinoma <strong>of</strong> the<br />

pancreas. In pts with completely resected pancreatic cancer, tumor grading<br />

and active treatment with GEM were the only predictive factor for LTS.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


244s Gastrointestinal (Noncolorectal) Cancer<br />

4021 Poster Discussion Session (Board #13), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Phase I/II study <strong>of</strong> preoperative (pre-op) short course chemoradiation (CRT)<br />

with proton beam therapy (PBT) and capecitabine (cape) followed by early<br />

surgery for resectable pancreatic ductal adenocarcinoma (PDAC) <strong>of</strong> the<br />

head. Presenting Author: Theodore S. Hong, Massachusetts General<br />

Hospital, Boston, MA<br />

Background: Standard adjuvant 6 week CRT may delay and reduce<br />

tolerability <strong>of</strong> adjuvant gemcitabine-based chemotherapy. We explore the<br />

safety and efficacy <strong>of</strong> a one-week course <strong>of</strong> pre-op CRT with PBT and cape<br />

followed by early pancreaticoduodenectomy (PD). Methods: Patients with<br />

radiographically resectable, biopsy-proven PDAC <strong>of</strong> the head were enrolled<br />

from May 2007-December 2010 on this prospective, NCI-sponsored<br />

clinical trial. Eligibility included no CT involvement <strong>of</strong> SMA or celiac artery;<br />

No metastatic disease. Dose level 1 consisted <strong>of</strong> PBT delivered 3 Gy x 10.<br />

Pts in subsequent dose levels received 5 Gy x5inprogressively shortened<br />

schedules: level 2 (wk1MWF,wk2TTh), level 3 (wk1MTThF,wk2M),<br />

level 4 (wk 1 M-F). PBT was targeted at pancreatic mass with elective nodal<br />

coverage. Pts received Cape 825 mg/m2 BID wk 1 and 2 M-F. Surgery was<br />

performed 1-6 wks after completion <strong>of</strong> cape. Patients were recommended<br />

to receive 6 mo <strong>of</strong> gemcitabine after surgery. Genotyping <strong>of</strong> 15 genes<br />

(including KRAS, PIK3CA, BRAF, NRAS, TP53, IDH1) was performed.<br />

Results: 50 pts were enrolled on study. 48 patients are eligible for this<br />

analysis. 3 pts were treated at each <strong>of</strong> dose levels 1-3. 6 pts were at dose<br />

level 4, which was selected as MTD. No DLTs were observed. 35 patients<br />

were treated in the phase II portion at the MTD. Gr 3 toxicity was noted in 2<br />

pts (chest wall pain-1, colitis-1). 38 pts underwent PD. Reasons for no PD<br />

were: metastatic disease-9, unresectable tumor-1. Mean post-op length <strong>of</strong><br />

stay was 7 days (range 5-47). 6/38 (16%) resected pts had positive<br />

margins. 28/38 (74%) had positive nodes. Median follow up is 21 months<br />

among the 19 patients still alive. 4/48 (8%) local failures in ALL patients.<br />

mOS and mPFS are 18 and 10 months respectively for ALL patients and 27<br />

and 14 months for RESECTED patients. Of 28 patients with genotype data<br />

available, 22 (79%) had KRAS mutations. 10/22 KRAS mt and 4/6 KRAS<br />

wt pts are alive. Conclusions: Pre-op CRT with 1 wk <strong>of</strong> PBT and capecitabine<br />

followed by early surgery is well tolerated and associated with favorable<br />

local control. Complete genotype and DPC4 data will be presented at the<br />

meeting.<br />

4023 Poster Discussion Session (Board #15), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Correlation <strong>of</strong> hand-foot skin reaction (HFS) with treatment efficacy in<br />

pancreatic cancer (PC) patients (pts) treated with gemcitabine/capecitabine<br />

plus erlotinib: A subgroup analysis from the AIO-PK0104 randomized,<br />

cross-over phase III trial in advanced PC. Presenting Author: Michael<br />

Haas, Department <strong>of</strong> Hematology and Oncology, Klinikum Grosshadern and<br />

Comprehensive Cancer Center, LMU Munich, Munich, Germany<br />

Background: AIO-PK 0104 investigated the efficacy and safety <strong>of</strong> gemcitabine/erlotinib<br />

(G/E) followed by capecitabine (C) vs. C/E followed by G.<br />

The present subgroup analysis evaluated the correlation between Cassociated<br />

skin toxicity and outcome parameters in PC. Methods: Within<br />

this multicenter phase III trial, pts with confirmed advanced PC were<br />

randomly assigned to 1st-line treatment with either C (2,000 mg/m2 /d,<br />

d1-14 q d21) plus E (150 mg/d, arm A) or G (1,000 mg/m2 over 30 min<br />

weekly x 7, then d1, 8, 15 q d28) plus E (150 mg/d, arm B). A cross-over to<br />

either G (arm A) or C (arm B) was performed after treatment failure (e. g.<br />

disease progression or unacceptable toxicity). Time to treatment failure<br />

after 1st- and 2nd -line therapy (TTF2) was the primary study endpoint.<br />

Treatment-related skin toxicity was evaluated separately for each treatment<br />

arm/each regimen based on NCI-CTCv2. Results: Of 279 eligible pts, 47<br />

had locally advanced, 232 had metastatic disease and 141 pts received<br />

second-line chemotherapy. For the present subgroup analysis data on skin<br />

toxicity were available from 255 pts. For the 73 pts (29%) with a HFS (any<br />

grade documented at any time during the treatment strategy), TTF2 and OS<br />

both were significantly prolonged compared to pts without HFS (7.4 vs 4.0<br />

months, p�0.001 and 9.7 vs 5.5 months, p�0.002, respectively).<br />

Considering HFS during 1st-line treatment in 123 pts within the CE arm,<br />

these results could be confirmed for the 47 pts (38%) with a documented<br />

HFS <strong>of</strong> any grade (TTF2: 7.6 vs. 3.2 months, p�0.001; OS: 10.2 vs. 4.4<br />

months, p�0.001). In pts receiving 1st-line treatment with G/E (n�132)<br />

no difference in outcome was observed for pts with (n�13) or without<br />

(n�119) HFS <strong>of</strong> any grade (TTF2: 5.7 vs. 4.2 months, p�0.375; OS: 8.4<br />

vs. 6.6 months, p�0.505). Conclusions: The current subgroup analysis <strong>of</strong><br />

AIO-PK0104 supports the assumption <strong>of</strong> a correlation between HFS in PC<br />

pts treated with capecitabine or capecitabine/erlotinib and efficacy endpoints<br />

like TTF2 and OS. A capecitabin-associated HFS thus might be<br />

predictive for efficacy in patients with advanced PC.<br />

4022 Poster Discussion Session (Board #14), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

A phase IB/randomized phase II study <strong>of</strong> gemcitabine (G) plus placebo (P)<br />

or vismodegib (V), a hedgehog (Hh) pathway inhibitor, in patients (pts) with<br />

metastatic pancreatic cancer (PC): Interim analysis <strong>of</strong> a University <strong>of</strong><br />

Chicago phase II consortium study. Presenting Author: Daniel Virgil<br />

Thomas Catenacci, University <strong>of</strong> Chicago, Chicago, IL<br />

Background: Sonic Hh (SHh), the ligand for the Hh pathway, is overexpressed<br />

in �80% <strong>of</strong> PC. V, a small molecule antagonist <strong>of</strong> the Hh<br />

signaling pathway, has activity in preclinical PC models. Methods: We<br />

conducted a multi-center, placebo-controlled, phase IB/randomized phase<br />

II trial <strong>of</strong> GV or GP. Eligible pts, KPS 80-100, had previously untreated<br />

metastatic PC, or had completed adjuvant therapy � 6 months (mo) prior.<br />

Primary endpoint: progression-free survival (PFS). Correlatives: serial SHh<br />

serum levels; serial contrast perfusion CT imaging. To allow for early<br />

stopping due to lack <strong>of</strong> efficacy, a planned interim analysis was performed<br />

after approximately 50% <strong>of</strong> the expected number <strong>of</strong> PFS events occurred.<br />

The protocol stipulated that the trial would be terminated if the probability<br />

<strong>of</strong> rejecting the null hypothesis were the trial to continue (conditional<br />

power) was �10%. All pts received G 1000mg/m2 over 30 minutes, days<br />

(D) 1, 8, 15, Q28D. Pts, stratified by KPS (80 v 90/100), and disease<br />

status (newly-diagnosed/recurrent), were randomized to V (150 mg PO<br />

daily) or P. For pts on P, cross-over was allowed at progression. Results: 70<br />

evaluable pts (V/P 35/35) enrolled at 12 sites 2/10-6/11. Pt characteristics:<br />

median age 63/63 (range 49-79/48-82); KPS 80: 8/8; 90: 12/14;<br />

100: 15/13. Grade 3/4 toxicity (%pts): neutropenia 20/26; hyponatremia<br />

3/11; fatigue 9/6; hyperglycemia 14/6; elevated alkaline phosphatase<br />

9/11. Response (%): complete 0/3, partial 0/11, stable disease 49/31.<br />

Median PFS: 3.7/2.4 mo (95% CI: 2.4-4.6/1.9-3.7; adjusted HR 0.92<br />

[0.52-1.64]). Upon progression/unblinding, 23 GP pts crossed over to GV.<br />

Median overall survival (OS): 6.3/5.4 mo (95% CI:4.9-7.8/4.2-8.0, adjusted<br />

HR 0.97, [0.47-2.01]). 1-year survival (%): 24/24. Laboratory and<br />

radiological correlatives will be presented. Conclusions: GV has an acceptable<br />

toxicity pr<strong>of</strong>ile. This trial did not meet criteria for futility at this interim<br />

analysis. The study is expected to complete accrual <strong>of</strong> 112 pts in February<br />

2012. The final analysis will be reported after 90 events. Funded by NCI<br />

N01-CM-62201.<br />

4026 Poster Discussion Session (Board #18), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Ethnic gene pr<strong>of</strong>ile <strong>of</strong> genes involved in angiogenesis to predict regional<br />

bevacizumab efficacy difference in gastric cancer. Presenting Author:<br />

Takeru Wakatsuki, University <strong>of</strong> Southern California Norris Comprehensive<br />

Cancer Center, Los Angeles, CA<br />

Background: AVAGAST showed regional bevacizumab (Bev) efficacy difference<br />

(RBED), namely, Asian (Asi) patients (pts) with gastric cancer (GC)<br />

had no benefit whereas European and Pan-<strong>American</strong> patients had more<br />

benefit from Bev. Recently, germline gene polymorphisms in angiogenesis<br />

have been recognized as predictive marker for Bev efficacy. Allele frequency<br />

(AF) in gene polymorphisms may vary depending on ethnicity. We<br />

tested the hypothesis whether angiogenic pathway gene polymorphisms<br />

may have different AF among Asi, Caucasian (Cau), and Hispanic (Hisp) in<br />

GC and this disparity may explain RBED. Methods: Three-hundred pts<br />

[Japanese (Jap), Cau, and Hisp, 100 from each race] with histopathologically<br />

confirmed GC were collected from Japan, USA, Austria, and Italy<br />

between 1991 and 2011. These pts were divided into 2 groups as training<br />

set (n�50) and validation set (n�50) in each race. Seven functional gene<br />

polymorphisms previously reported as predictive marker were selected. All<br />

samples were analyzed using PCR-based direct DNA- sequencing. Fisher’s<br />

exact test was used to compare the distribution <strong>of</strong> AF among races. Results:<br />

Significant disparate distributions in favorable AF for Bev were shown<br />

among races in Table. Jap GC pts had significant lower AF <strong>of</strong> 5 predictive<br />

gene polymorphisms in training set, and among these 5, three predictive<br />

gene polymorphisms were also validated. Conclusions: Our preliminary<br />

results showed significant disparate AF distributions in predictive gene<br />

polymorphisms for Bev, and these disparities may explain RBED in<br />

AVAGAST. Further investigation is warranted to elucidate RBED.<br />

The favorable allele frequencies for bevacizumab.<br />

Training set Validation set<br />

Cau (%) Hisp (%) Jap (%) P value Cau (%) Hisp (%) Jap (%) P value<br />

VEGFR2 rs12505758 T/C 84.0 85.0 68.8 0.009 87.0 83.3 64.3 0.004<br />

VEGFR1 rs9582036 C/A 79.0 86.0 82.7 0.660 75.0 80.0 83.0 0.740<br />

VEGF-A rs699946 A/G 78.0 60.2 49.0 0.001 78.0 76.0 58.0 0.021<br />

VEGFR2 rs11133360 C/T 55.1 43.8 70.8 0.002 60.0 35.9 68.0 �0.001<br />

VEGF-A rs833061 C/T 52.0 35.0 29.6 0.020 50.0 40.0 33.0 0.200<br />

VEGF-A rs699947 A/C 51.0 34.0 27.6 0.013 51.0 36.0 29.0 0.026<br />

VEGF-A rs1570366 A/G 39.0 17.0 14.6 0.001 37.0 22.0 20.4 0.130<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


4027 Poster Discussion Session (Board #19), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Multicenter randomized phase II trial <strong>of</strong> cisplatin, irinotecan plus bevacizumab<br />

(PCA) versus docetaxel, cisplatin, irinotecan plus bevacizumab<br />

(TPCA) in patients (pts) with metastatic esophagogastric cancer (MEGCA).<br />

Presenting Author: Peter C. Enzinger, Dana-Farber Cancer Institute,<br />

Boston, MA<br />

Background: In MEGCA, PCA has a 65% response rate (RR) and 8.3 mos<br />

time to progression (Shah. JCO 2006); TPCA has a 66% RR and 8.9 mos<br />

progression-free survival (PFS) (Enzinger. ESMO 2008 updated). Methods:<br />

Chemo naive pts with MEGCA were stratified: ECOG (0/1 vs. 2) and disease<br />

site (Gastric or GEJ/esophageal) and randomized 1:1 to PCA or TPCA. PCA<br />

-- cisplatin 30mg/m2 and irinotecan 65mg/m2 on d1, 8, and bevacizumab<br />

10 mg/kg d1 docetaxel 30mg/m2, cisplatin 25mg/m2 and irinotecan<br />

50mg/m2 on d1, 8, and bevacizumab 10 mg/kg d1. Cycles were 3 weeks.<br />

Response assessment - q6wks (RECIST 1.1). Primary endpoint was PFS.<br />

Results: 85 patients: median age�61 (40-85); male/female� 70/15;<br />

ECOG: 0/1/2�(34/49/2); gastric/GE junction/esophageal�27/21/37; measurable/evaluable:<br />

78/7; sites <strong>of</strong> metastatic disease (# <strong>of</strong> pts): lymph nodes<br />

(34), liver (31), lung (11), bone (5), abdomen (4), peritoneum (4), other<br />

(11). There were two treatment-related deaths: esophageal hemorrhage,<br />

sudden death (both TPCA). Conclusions: Both regimens were efficacious<br />

and reasonably well tolerated. The addition <strong>of</strong> docetaxel to the attenuated<br />

PCA combination did not significantly improve efficacy.<br />

PCA (N�44) TPCA (N�41) P value<br />

Major response 56.8% (41.0, 71.7) 48.8% (32.9, 64.9) 0.518<br />

PFS median (N�85) 7.7mos (5.8, 10.1) 8.4mos (6.8, 9.8) 0.861<br />

OS 1-year (N�85) 52.6% (36.0, 66.8) 59.1% (42.2, 72.7) 0.866<br />

OS median (N�85)<br />

Grade III-IV toxicity %<br />

12.5mos (9.6, 15.5) 13.3mos (11.5, 15.4) 0.866<br />

Neutropenia 25.0 31.7 0.492<br />

Thrombocytopenia 4.5 4.9 1.000<br />

Diarrhea 22.7 31.7 0.465<br />

Fatigue 15.9 19.5 0.778<br />

Vomiting 9.1 12.2 0.733<br />

Infection 11.4 7.3 0.714<br />

Anorexia 2.3 9.8 0.192<br />

Thromboembolic 4.5 9.8 0.423<br />

HTN 4.5 4.9 1.000<br />

4029 Poster Discussion Session (Board #21), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

The pattern and timing <strong>of</strong> disease recurrence in squamous cancer <strong>of</strong> the<br />

anus: Mature results from the NCRI ACT II trial. Presenting Author: David<br />

Sebag-Montefiore, St James Institute <strong>of</strong> Oncology, Leeds, United Kingdom<br />

Background: Concurrent chemoradiation (CRT) is standard treatment for<br />

patients with squamous cell carcinoma <strong>of</strong> the anus. Although the majority<br />

<strong>of</strong> patients achieve long term disease control, locoregional failure is either<br />

detected early (failure to enter complete remission) or at a later date as<br />

locoregional recurrence. We explore the pattern and timing <strong>of</strong> disease<br />

recurrence within a phase III trial (ACT II), which mandated standardised<br />

radiation fields and doses (50.4Gy in 28 daily fractions). Methods: The UK<br />

ACT II trial recruited a total <strong>of</strong> 940 patients (2000 -2008) and compared<br />

cisplatin with mitomycin when combined with 5flurouracil CRT and two<br />

cycles <strong>of</strong> maintenance chemotherapy versus no maintenance using a<br />

factorial 2x2 design. Patient characterstics: T1/2 52%, T3/4 46%; N�<br />

32% N0 62%. The loco-regional response to treatment was assessed by<br />

clinical examination at 11, 18 and 26 weeks after CRT. <strong>Clinical</strong> examination<br />

was performed every two months for year 1; 3-monthly for year 2; then<br />

6-monthly for years 3-5. CT scans <strong>of</strong> chest/abdomen/pelvis were performed<br />

at 6,12 24 months. Sites <strong>of</strong> failure were recorded on a case record form<br />

(primary site [PS], inguinal [ING], pelvic nodes [PEL], metastatic [METS].<br />

Results: After a median follow up <strong>of</strong> 5 years, the crude pelvic failure (PF)<br />

rate is 18% (163/924). Of the 163 PF, 133 pts (82%) were pelvic only and<br />

30 (18%) had PF � metastasis. 93% <strong>of</strong> all pelvic recurrences were<br />

detected during the first three years (54% yr 1, 26% yr 2 and 13% yr 3).<br />

The pattern was similar for PF only and PF � mets (data not shown). Only<br />

46 (4%) <strong>of</strong> patients had [METS] as the first disease related event. 117/133<br />

(88%) patients with PF only are evaluable for analysis <strong>of</strong> the pattern <strong>of</strong><br />

failure., 53% occurred at the primary site (PS) only, 23% were PS � nodal<br />

(5% [ING] 13% [PEL] and 5% [ING�PEL], and 25% nodal only (6%<br />

[ING],14% [PEL] and 5% [ING�PEL]). Conclusions: The ACT II trial results<br />

with mature follow-up demonstrate a low rate <strong>of</strong> pelvic failure. Intensive<br />

follow up to detect potentially salvageable pelvic failure should be<br />

restricted to a maximum <strong>of</strong> three years in future trials as only 7% <strong>of</strong><br />

relapses occur beyond this time point.<br />

Gastrointestinal (Noncolorectal) Cancer<br />

245s<br />

4028 Poster Discussion Session (Board #20), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

A randomized phase II study <strong>of</strong> continuous versus intermittent S-1 plus<br />

oxaliplatin in first-line treatment <strong>of</strong> patients with metastatic gastric<br />

carcinoma. Presenting Author: Sook Ryun Park, Center for Gastric Cancer,<br />

National Cancer Center, Goyang, Gyeonggi, South Korea<br />

Background: We conducted a randomized phase II study to compare<br />

continuous vs. intermittent S-1 � oxaliplatin (SOX) after induction <strong>of</strong> 6<br />

cycles <strong>of</strong> SOX in patients (pts) with metastatic gastric cancer (MGC).<br />

Methods: Pts �18 yrs with chemo-naive MGC, normal organ function,<br />

ECOG PS 0-2, and measurable or evaluable lesion(s) were initially given an<br />

induction treatment <strong>of</strong> 6 cycles <strong>of</strong> SOX (S-1 40 mg/m2 bid on D1-14 �<br />

oxaliplatin 130 mg/m2 on D1 q 3 wks). Pts with CR/PR or SD were<br />

randomized to continue SOX (arm A) until progression/intolerable toxicity<br />

or discontinue until progression when SOX was re-administered (arm B).<br />

The primary endpoint was overall survival (OS), and secondary endpoints<br />

included progression-free survival (PFS), response rate, safety, quality <strong>of</strong><br />

life, and biomarker correlative studies. Results: From July 2007 to Dec<br />

2010, a total <strong>of</strong> 250 pts entered into the study. Median age was 53 yrs<br />

(range, 24-69); PS 0/1/2�13/219/18; M/F�162/88. Three pts did not<br />

receive treatment and 126 (50.4%) discontinued treatment before or at<br />

the completion <strong>of</strong> 6 cycles <strong>of</strong> SOX due to progression (45.6%), pt refusal<br />

(2.8%), or adverse events (2.0%). A total <strong>of</strong> 121 pts were randomized: 59<br />

(48.8%) to arm A and 62 (51.2%) to arm B. <strong>Clinical</strong> characteristics were<br />

well balanced between arms. SOX continuation resulted in a significant<br />

reduction in the risk <strong>of</strong> progression (median PFS, 10.5 months for arm A vs.<br />

7.2 months for arm B; HR�0.57, 95% CI 0.39-0.84, p�0.005). With a<br />

median follow-up <strong>of</strong> 34.0 months (range, 10.5-53.3 months), there was no<br />

significant difference in OS (median OS, 22.6 months for arm A vs. 22.7<br />

months for arm B; HR, 0.79, 95% CI 0.51-1.25, p�0.31). Arm A had<br />

higher rates <strong>of</strong> grade 3/4 fatigue (28.8% vs. 8.1%, p�0.004) and<br />

neuropathy (25.4% vs. 8.1%, p�0.014) but other grade 3/4 hematologic<br />

(neutropenia, 35.6% vs. 32.3%; thrombocytopenia, 23.7% vs. 21.0%;<br />

anemia, 15.3% vs. 6.5%) or non-hematologic toxicity rates were not<br />

significantly different. Conclusions: Continuous chemotherapy with SOX<br />

after induction therapy improved PFS but not OS. Supported by NCC Grant<br />

1010180 (S-1 and oxaliplatin was provided by JEIL Pharm. Co., Ltd. and<br />

san<strong>of</strong>i-aventis Korea Co., Ltd., respectively).<br />

4030 Poster Discussion Session (Board #22), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Phase II trials <strong>of</strong> cetuximab (CX) plus cisplatin (CDDP), 5-fluorouracil<br />

(5-FU) and radiation (RT) in immunocompetent (ECOG 3205) and HIVpositive<br />

(AMC045) patients with squamous cell carcinoma <strong>of</strong> the anal<br />

canal (SCAC): Safety and preliminary efficacy results. Presenting Author:<br />

Madhur Garg, Albert Einstein College <strong>of</strong> Medicine/Montefiore Medical<br />

Center, Bronx, NY<br />

Background: Epidermal growth factor receptor (EGFR) expression and HPV<br />

infection are common in SCAC. We therefore initiated 2 phase II studies to<br />

evaluate the safety and efficacy <strong>of</strong> the EGFR inhibitor CX given concurrently<br />

with CDDP/5-FU/RT in HIV-positive (AMC045) and immunocompetent<br />

(E3205) patients with SCAC. Methods: All patients received CX (400<br />

mg/m2 loading, then 250 mg/m2/wk IV x 6-8 wks) plus CDDP (75 mg/m2<br />

IV q28 days x 2) and 5-FU (1000 mg/m2/day IV infusion days 1-4 q 28<br />

days x 2) concurrently with RT (45-54 Gy) beginning with CX dose 2.<br />

Patients in E3205 also received 2 cycles <strong>of</strong> CDDP/5-FU alone prior to<br />

CX/CDDP/5-FU/RT; this was discontinued on recommendation <strong>of</strong> the NCI<br />

Anorectal Task Force after 28 patients. Both trials were powered to detect a<br />

reduction in 3-year local-regional failure (LRF) rate from 35% to 17.5%<br />

(alpha�0.10, beta�0.10), the primary end point. Other endpoints included<br />

progression free survival (PFS) and overall survival (OS). The results<br />

below include complete toxicity and preliminary efficacy data (including<br />

only the first 28 patients from E3205). Results: Expedited reporting was<br />

required for type I (any grade 5, grade 4 cardiac) or II (grade 4 RT skin,<br />

diarrhea) adverse events, with prespecified rates <strong>of</strong> �5% or �20%,<br />

respectively defined as unacceptable. Early stopping rules were not invoked<br />

for either trial. LRF rates data will be presented after more detailed case<br />

review is completed. Conclusions: CX plus CDDP/5-FU/RT is feasible in<br />

patients with SCAC, including patients with HIV infection. Preliminary<br />

safety and efficacy data appear encouraging, but accrual without neoadjuvant<br />

CDDP/5-FU continues in E3205, and additional followup <strong>of</strong> both<br />

study cohorts is required in order to determine whether pre-specified<br />

efficacy endpoints were met.<br />

AMC045 E3205<br />

No. 45 28<br />

Stage I/II/III 24%/42%/34% 11%/50%/39%<br />

Completed protocol therapy 37 (82%) 22 (79%)<br />

Type I/II adverse events 2 (4%)/0 1 (4%)/1 (4%)<br />

Colostomy rate 7% (1-18%) 14% (4-33%)<br />

2 year PFS rate (95% CI) 80% (61-90%) 92% (81-100%)<br />

2 year OS rate (95% CI) 89% (73-96%) 93% (83-100%)<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


246s Gastrointestinal (Noncolorectal) Cancer<br />

4031 Poster Discussion Session (Board #23), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Randomized phase II trial <strong>of</strong> gemcitabine plus S�1 combination therapy<br />

versus S�1 in advanced biliary tract cancer: Results <strong>of</strong> the Japan <strong>Clinical</strong><br />

Oncology Group study (JCOG0805). Presenting Author: Makoto Ueno,<br />

Kanagawa Cancer Center Hospital, Yokohama, Japan<br />

Background: Gemcitabine plus cisplatin combination (GC) therapy is the<br />

standard therapy for advanced biliary tract cancer (BTC). In previous trials,<br />

gemcitabine plus S-1 combination (GS) therapy and S-1 mono-therapy had<br />

shown considerable efficacy in patients with BTC. The aim <strong>of</strong> this trial is to<br />

evaluate the efficacy and safety <strong>of</strong> the two regimens and to determine which<br />

is more promising as a test arm regimen for a subsequent phase III trial.<br />

Methods: Chemotherapy-naive patients with recurrent or unresectable BTC<br />

(gallbladder [GB], intrahepatic biliary duct [IHBD], extrahepatic biliary<br />

duct [EHBD], ampulla <strong>of</strong> Vater [AV]), an ECOG PS <strong>of</strong> 0-1,andadequate<br />

organ function were randomly assigned to receive GS (gemcitabine: 1,000<br />

mg/m2 , iv, days 1 and 8; S-1: 60 mg/m2 , p.o., days 1 - 14, every 3 weeks) or<br />

S-1 (80 mg/m2 , p.o., days 1 - 28, every 6 weeks). We assumed that<br />

%1-year survival <strong>of</strong> one regimen is 30% and that <strong>of</strong> the other regimen is<br />

more than 40%. To ensure at least 85% probability <strong>of</strong> correct selection, 98<br />

eligible pts are required. The decision rule was that the regimen with higher<br />

%1-year survival will be considered as more promising regimen. Results:<br />

From February 2009 to April 2010, 101 pts (GB, n�38; IHBD, n�35;<br />

EHBD, n�20; AV, n�8) were randomized (GS, n�51; S-1, n�50). For the<br />

GS arm and S-1 arm, %1-year survivals were 52.9% and 40.0%, the<br />

median survival time were 12.5 and 9.0 months (hazard ratio 0.86 [95%CI<br />

0.54-1.36]; p�0.52), and the median progression-free survival time were<br />

7.1 and 4.2 months (0.44 [0.29-0.67]; p�0.0001). Grade 3/4 hematological<br />

toxicities were more frequent in the GS arm than in the S-1 arm,<br />

(percentage in GS/S-1 arms): neutropenia, 60.8/4.0; leukocytopenia,<br />

29.4/2.0; hemoglobin, 11.8/4.0; and thrombocytopenia, 11.8/4.0; respectively.<br />

Although two treatment-related deaths occurred in the GS arm<br />

(pneumonitis, acute myocardial infarction), other grade 3/4 non-hematological<br />

toxicities were infrequent and reversible in both arms. Conclusions: The<br />

GS arm was superior in %1-year survival to S-1. Here we consider GS to be<br />

more promising as the test arm for a subsequent phase III trial comparing<br />

with GC.<br />

4033 Poster Discussion Session (Board #25), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Observations <strong>of</strong> hepatocellular carcinoma (HCC) management patterns<br />

from the global HCC bridge study: First characterization <strong>of</strong> the full study<br />

population. Presenting Author: Joong-Won Park, National Cancer Center,<br />

Goyang, South Korea<br />

Background: HCC is a major health problem across the world. The global<br />

HCC BRIDGE study is the first global, large-scale, observational study to<br />

document the real-world experience <strong>of</strong> HCC patients from diagnosis to<br />

death. Methods: This longitudinal cohort study (started March 2009)<br />

includes HCC patients newly diagnosed between January 2005 and June<br />

2011 and treated at major medical centers, with data collected retrospectively<br />

and prospectively as recorded in patient charts. Full patient enrollment<br />

is expected at the end <strong>of</strong> January 2012. Results: At the time <strong>of</strong> the<br />

first interim analysis (July 2011), 12,442 treated HCC patients were<br />

enrolled at 42 sites in Asia (n�8909, 72% [China: n�6295, 71%; Japan:<br />

n�295, 3%]), Europe (n�2040, 16%) and North America 1493 (n�1493,<br />

12%). Mean age was 57 years; 82% were male. The predominant risk<br />

factor was HBV in Asia (76%) and HCV in Europe (48%), North America<br />

(45%) and Japan (69%). Most patients were diagnosed without surveillance<br />

(Asia, 82%; Europe, 73%; North America, 69%; Japan, 64%). In<br />

Asia, Europe and North America, the predominant BCLC stage at diagnosis<br />

was C (48%, 46%, 46%) followed by A (34%, 28%, 26%). First recorded<br />

treatments in Asia, Europe and North America were resection (31%, 15%,<br />

21%), transplantation (1%, 3%, 1%), TACE (49%, 30%, 36%), other<br />

locoregional therapy (12%, 35%, 23%) and systemic therapy (3%, 10%,<br />

8%). Treatments ever used (2005–2011) in Asia, Europe and North<br />

America were resection (33%, 17%, 24%), transplantation (2%, 6%,<br />

12%), TACE (57%, 35%, 48%), other locoregional therapy (20%, 42%,<br />

37%) and systemic therapy (9%, 20%, 21%). These results will be<br />

updated with data from the full study population (approx. 19,000 patients),<br />

and preliminary survival data will be presented. Conclusions: As the<br />

largest study <strong>of</strong> its type, in 19,000 patients worldwide, the HCC BRIDGE<br />

study provides valuable insights into global HCC disease characteristics<br />

and patient management. Based on prior analyses, differences in risk<br />

factors among regions confirm well-known trends, while other observed<br />

differences (e.g., treatment variations) may be related to country- and<br />

site-specific practices and patient characteristics.<br />

4032 Poster Discussion Session (Board #24), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Gemcitabine and oxaliplatin (GEMOX) alone or in combination with<br />

cetuximab as first-line treatment for advanced biliary cancer: Final analysis<br />

<strong>of</strong> a randomized phase II trial (BINGO). Presenting Author: David Malka,<br />

Institut Gustave Roussy, Villejuif, France<br />

Background: Gemcitabine-platinum chemotherapy (CTx) regimens are widely<br />

accepted as first-line standard <strong>of</strong> care for patients (pts) with advanced<br />

biliary cancers (ABC). EGFR overexpression has been observed in ABC,<br />

suggesting that the combination with anti-EGFR monoclonal antibodies<br />

may be appropriate. Methods: Patients with ABC, WHO performance status<br />

(PS) 0-1, and without prior palliative CTx were eligible for this international,<br />

open-label, two-stage, non-comparative, randomized phase II trial.<br />

Patients received GEMOX (gemcitabine, 1 g/m² [10 mg/m²/min] at day [D]1<br />

� oxaliplatin, 100 mg/m² at D2, arm A) or GEMOX � cetuximab (500<br />

mg/m² at D1 or 2, arm B), every 2 weeks. The primary endpoint was crude<br />

4-month progression-free survival (PFS) rate (H0, �40%; H1, �60%;<br />

planned sample size, 100 pts, increased to 150 pts by amendment to allow<br />

subgroup analyses). Secondary endpoints were objective response rate<br />

(ORR), disease control rate (DCR), PFS, overall survival (OS), and toxicity<br />

(NCI-CTC v3.0). Exploratory endpoints included early metabolic response<br />

as assessed by PET at 1 month, and tumor KRAS mutational analysis.<br />

Results: From Oct. 2007 to Dec. 2009, we enrolled 150 pts (median age,<br />

62 years; male, 57%; metastatic, 79%; cholangiocarcinoma, 84%; median<br />

follow-up, 30 months) (Table). Conclusions: GEMOX-cetuximab regimen<br />

was well tolerated and met its primary endpoint (4-month PFS<br />

�60%). However, median PFS and OS were similar in both arms.<br />

Exploratory analyses (e.g., KRAS tumor status) are underway to identify pt<br />

subgroups deriving benefit from the addition <strong>of</strong> cetuximab to CTx.<br />

Arm A Arm B<br />

Cycles (median) (n) 10 10<br />

Toxicity (%) 1<br />

Peripheral neuropathy 2<br />

46 49<br />

Neutropenia 16 22<br />

Fatigue 13 17<br />

Thrombocytopenia 19 11<br />

Gastrointestinal 15 13<br />

Allergy/hypersensitivity 1 8<br />

Rash 0 7<br />

Efficacy [95% CI]<br />

ORR (%) 3<br />

29 [18-40] 23 [13-33]<br />

DCR (%) 3<br />

77 [67-88] 87 [79-95]<br />

4-month PFS (%) 53 [41-64] 63 [52-74]<br />

PFS (median, months) 5.3 [3.7-6.6] 6.0 [5.0-7.4]<br />

OS (median, months) 12.4 [8.6-16.0] 11.0 [8.9-13.7]<br />

1 Grade 3-4, NCI-CTC v3.0.<br />

2 Grade 2-3, Modified Levi’s scale.<br />

3 In 131 evaluable pts.<br />

4034 General Poster Session (Board #40A), Mon, 8:00 AM-12:00 PM<br />

Final results <strong>of</strong> a phase I/II study in patients with pancreatic cancer,<br />

malignant melanoma, and colorectal carcinoma with trabedersen. Presenting<br />

Author: Helmut Oettle, Universitätsmedizin Berlin Charite, Campus<br />

Virchow-Klinikum, Berlin, Germany<br />

Background: TGF-�2 overexpression in solid tumors triggers key cancer<br />

pathomechanisms, i.e. suppression <strong>of</strong> antitumor immune responses system<br />

and metastasis. Trabedersen specifically inhibits TGF-�2 expression.<br />

In the clinical Phase I/II study we evaluate MTD, safety, pharmakokinetics<br />

(PK), and efficacy <strong>of</strong> i.v. trabedersen in patients with advanced tumors.<br />

Methods: A total <strong>of</strong> 61 patients with pancreatic cancer (PancCa, n�37),<br />

malignant melanoma (MM, n�19), or colorectal carcinoma (n�5) were<br />

treated with i.v. trabedersen as 2nd to 4th-line therapy with escalating doses<br />

in 2 treatment schedules. (1st schedule: 7d on, 7d <strong>of</strong>f; 2nd schedule: 4d on,<br />

10d <strong>of</strong>f; up to 10 cycles). Within the 1st schedule, the MTD was established<br />

at 160 mg/m2 /d. In the 2nd schedule dose-escalation was stopped before<br />

reaching MTD. In the Phase II-part <strong>of</strong> the study further PancCa and MM<br />

patients were treated with 140 mg/m2 /d. For assessment <strong>of</strong> PK parameters,<br />

plasma time pr<strong>of</strong>iles were analyzed for trabedersen and its n-1 to n-5<br />

metabolites by non-compartimental analysis. Results: Trabedersen was safe<br />

and well-tolerated. The only expected adverse reaction identified is<br />

non-serious and transient thrombocytopenia. Only 2 SAEs (gastrointestinal<br />

hemorrhage und pyrexia) were considered as possibly related to study<br />

medication. Further clinical development will focus on PancCa patients<br />

receiving 140 mg/m2 /d trabedersen as 2nd-line treatment. Survival analysis<br />

<strong>of</strong> these patients revealed a mOS <strong>of</strong> 13.4 months (n�9; 95% CI: 2.2,<br />

39.7). One PanCa patient had a complete response <strong>of</strong> liver metastases and<br />

is still alive after 75 months. Promising efficacy data were also seen in MM<br />

patients enrolled into the last cohort (140 mg/m2 /day) with a current mOS<br />

<strong>of</strong> 9.3 months (n�14; 95% CI: 6.5, 12.2). PK analyses showed for both<br />

treatment schedules that exposure to trabedersen was in the expected<br />

range for all doses and half-life <strong>of</strong> trabedersen (1.12 to 2.08 hrs) as well as<br />

clearance (2.22-4.37 L/h*m2 ) were independent <strong>of</strong> dose. Conclusions:<br />

Trabedersen showed excellent safety and encouraging survival results in<br />

the Phase I/II clinical study. A randomized, active-controlled study in 2nd line stage IV PanCa patients is in preparation.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


4035 General Poster Session (Board #40B), Mon, 8:00 AM-12:00 PM<br />

Updated results <strong>of</strong> the GEST study: Randomized phase III study <strong>of</strong><br />

gemcitabine plus S-1 (GS) versus S-1 versus gemcitabine (GEM) in<br />

unresectable advanced pancreatic cancer in Japan and Taiwan. Presenting<br />

Author: Akira Fukutomi, Shizuoka Cancer Center, Sunto-gun, Japan<br />

Background: The GEST study (Ioka et al. ASCO 2011, Abstract 4007)<br />

demonstrated the non-inferiority <strong>of</strong> S-1 to GEM with respect to the primary<br />

endpoint <strong>of</strong> overall survival (OS) in patients with pancreatic cancer (PC).<br />

We now report the updated results <strong>of</strong> this study. Methods: The GEST study<br />

was a randomized, 3-arm, phase III study. Chemotherapy-naive patients<br />

with unresectable advanced PC, an ECOG Performance status (PS) <strong>of</strong> 0-1,<br />

and adequate organ functions were randomly assigned to receive GEM<br />

(1000 mg/m2 , iv, d1, 8 and 15, q4w), S-1 (80/100/120 mg/day based on<br />

BSA, po, d1-28, q6w), or GS (GEM 1000 mg/m2 , iv, d1 and 8 plus S-1<br />

60/80/100 mg/day based on BSA po, d1-14, q3w). The primary endpoint<br />

was OS, used to assess the non-inferiority <strong>of</strong> S-1 and the superiority <strong>of</strong> GS<br />

to GEM. Patient information was updated in July 2011. Results: At the time<br />

<strong>of</strong> this follow-up analysis, median follow-up was 29.8 months with 795 OS<br />

events, compared with 18.4 months with 710 OS events out <strong>of</strong> 832<br />

patients at the previous analysis. Median OS was 8.8 months (95% CI:<br />

8.0–9.7) in the GEM group and 9.7 months (95% CI: 7.6-10.8) in the S-1<br />

group (HR�0.96, 97.5% CI: 0.79-1.17, p�0.001 for non-inferiority),<br />

which is consistent with prior results (HR�0.96, 97.5% CI: 0.78-1.18,<br />

p�0.001). In the GS group, median OS was 9.9 months (95% CI:<br />

9.0-11.2). The HR was 0.91 (97.5%CI: 0.75-1.11, p�0.28 for superiority<br />

versus the GEM group). On subgroup analysis, GS was associated with a<br />

non-statistically significant trend toward better OS compared with GEM<br />

among patients with locally advanced disease and those with a PS <strong>of</strong> 1.<br />

Median OS was 12.7 months (95% CI: 9.7–14.9) in the GEM group and<br />

15.9 months (95% CI: 13.0-19.7) in the GS group (HR�0.72, 95% CI:<br />

0.51-1.03) among patients with locally advanced disease, and 6.2 months<br />

(95% CI: 4.9–8.3) in the GEM group and 9.6 months (95% CI: 8.0-10.9)<br />

in the GS group (HR�0.62, 95% CI: 0.46-0.83) among patients with a PS<br />

<strong>of</strong> 1. Conclusions: The non-inferiority <strong>of</strong> S-1 to Gem in terms <strong>of</strong> the primary<br />

endpoint <strong>of</strong> OS was reconfirmed. Monotherapy with S-1 can be used as one<br />

<strong>of</strong> the standard treatments for advanced PC. As for GS therapy, there is<br />

room for further investigation.<br />

4037 General Poster Session (Board #40D), Mon, 8:00 AM-12:00 PM<br />

Incidence and characterization <strong>of</strong> venous thromboembolic events (VTE) in<br />

patients with pancreatic cancer: Effect <strong>of</strong> timing <strong>of</strong> VTE on survival.<br />

Presenting Author: Maithili A. Shethia, University <strong>of</strong> Texas M. D. Anderson<br />

Cancer Center, Houston, TX<br />

Background: Patients (pts) with pancreatic cancer are at high risk for VTE,<br />

and the occurrence <strong>of</strong> VTE can affect pts’ prognosis. The purpose <strong>of</strong> this<br />

study was to evaluate the incidence <strong>of</strong> VTE and the impact <strong>of</strong> timing <strong>of</strong> VTE<br />

(early vs. late) on survival. Methods: Medical record <strong>of</strong> 260 pts with<br />

pancreatic cancer, newly referred to UT MDACC during one year period<br />

from 1/1/2006 to 12/31/2006, were reviewed for the incidence <strong>of</strong> VTE<br />

during a 2-year follow-up period from the date <strong>of</strong> diagnosis. All VTE<br />

episodes were confirmed by radiologic studies. Survival analysis was<br />

conducted using Kaplan-Meier analysis and Cox proportional hazard<br />

models. Results: Of the 260 pts, 47 pts (18%) had 51 episodes <strong>of</strong> VTE<br />

during the 2-year follow-up. The median age <strong>of</strong> the pts with VTE was 61<br />

years (range: 28-86) and 53% were males. Of the 47 pts with VTE, 27<br />

(57%) had PE, 19 (40%) had DVT and 1 had concurrent PE/DVT. Three pts<br />

had recurrent VTE during the study period. Median follow-up time for OS<br />

was 192 days (range: 1-1652 days). Kaplan-Meier Survival analysis<br />

showed that those who developed VTE earlier (within 30 or 90 days) had<br />

shorter median overall survival (OS) compared with those who had VTE<br />

beyond these time points. The hazard ratios, 95% CI, and median OS at 1<br />

year are summarized in the table below. Conclusions: The incidence <strong>of</strong> VTE<br />

is high in pts with pancreatic cancer. The timing <strong>of</strong> VTE had a significant<br />

impact on OS; pts who had an early development <strong>of</strong> VTE had a shorter<br />

overall survival.<br />

Timing <strong>of</strong> VTE*<br />

1 year OS Median 1 year OS**<br />

Hazard ratio<br />

(95% CI) p value<br />

Early VTE<br />

vs. late VTE p value***<br />

Within 30 days 3.52 (1.71-7.23) 0.0006 116 vs. 295 0.0003<br />

Within 90 days 5.33 (1.85-15.35) 0.0019 152 vs. NR 0.0006<br />

* From the date <strong>of</strong> diagnosis <strong>of</strong> pancreatic cancer to date <strong>of</strong> diagnosis <strong>of</strong> VTE; **<br />

days; *** Log-rank; NR: not reached.<br />

Gastrointestinal (Noncolorectal) Cancer<br />

4036 General Poster Session (Board #40C), Mon, 8:00 AM-12:00 PM<br />

Efficacy and safety <strong>of</strong> bevacizumab combined with chemotherapy in the<br />

treatment <strong>of</strong> patients with metastatic well-differentiated duodenopancreatic<br />

endocrine tumors (BETTER study). Presenting Author: Michel<br />

Ducreux, Institut Gustave Roussy, Villejuif, France<br />

Background: Gastrointestinal neuroendocrine tumors (NET) overexpress<br />

vascular endothelial growth factor (VEGF), thus we hypothesized that<br />

Bevacizumab (Bv), a monoclonal antibody targeting VEGF in combination<br />

with chemotherapy may show an activity in duodeno-pancreatic NET.<br />

Methods: This multicenter, open-label, non-randomized, two- group phase<br />

II trial assessed in one group the efficacy and safety <strong>of</strong> Bv (7.5 mg/kg IV on<br />

day 1, every 3 weeks), combined with 5-FU/streptozotocin (stz): 5-FU 400<br />

mg/m²/d; Stz 500 mg/m²/d IV, d1-5/ every 42 days, during a minimum <strong>of</strong> 6<br />

months in patients (pts) with progressive, metastatic, well-differentiated<br />

duodeno-pancreatic NET (WHO 2000), not previously treated with chemotherapy,<br />

ECOG PS �2 and Ki 67 index � 15%. Primary endpoint was<br />

progression-free survival (PFS). Secondary endpoints were overall survival<br />

(OS), response rate, safety and quality <strong>of</strong> life. Study duration was 24<br />

months. Results: In the ITT population, 34 pts were enrolled, 22 (64.7%)<br />

were men, median age 55.3 years (37.0-77.6), 33 (97.1%) pts had<br />

ECOG-PS 0/1. Most metastases were in the liver 33 (97.1%) pts or lymph<br />

nodes 14 (41.2%) pts. Ki-67 proliferative index was 0-2% in 8 (25%) pts,<br />

3-4% in 5 (15.6%), 5-9% in 6 (18.8%), 10-14% in 12 (37.5%) and 15%<br />

in 1(3.1%) pt. Median treatment duration was 14.0 months; median<br />

number <strong>of</strong> cycles was 10. At 24 months, median PFS was 23.7 months<br />

[95%CI: 14.5; not reached] based on 18 events. PFS rate at 18 months<br />

was 62%. Tumor control rate was 100% (n� 34) including partial response<br />

in 19 (55.9%) pts and stable disease in 15 (44.1%) pts. Survival rate at 24<br />

months was 88%. Median OS was not reached, 5 patients died. CTC grade<br />

3/4 Adverse Events (AEs) occurred in 23 (67.6%) <strong>of</strong> pts, mainly digestive 5<br />

(14.7%) pts. G3/4 Bv targeted AEs were hypertension in 7 (20.6%) pts and<br />

thromboembolism in 4 (11.8%). Conclusions: Efficacy data in this phase II<br />

trial assessing a novel approach with Bv and 5-FU/stz in duodenopancreatic<br />

NET is encouraging and the toxicity pr<strong>of</strong>ile is acceptable.<br />

Results suggest that Bv may have a place in the treatment <strong>of</strong> pancreatic<br />

NET and warrant further investigation in a phase III trial.<br />

4038 General Poster Session (Board #40E), Mon, 8:00 AM-12:00 PM<br />

A pooled analysis <strong>of</strong> phase II trials <strong>of</strong> gemcitabine (GEM)-containing<br />

doublets plus bevacizumab (BEV): Should combination chemotherapy<br />

serve as the backbone in clinical trials <strong>of</strong> advanced pancreatic cancer<br />

(APC)? Presenting Author: Katherine Van Loon, University <strong>of</strong> California,<br />

San Francisco Comprehensive Cancer Center, San Francisco, CA<br />

Background: GEM has served as the chemotherapy platform for most phase<br />

III clinical trials in APC, inc. CALGB 80303 (GEM �/- BEV). However,<br />

GEM-based combination regimens may confer superior outcomes in select<br />

pts and represent a preferred backbone in clinical trial design testing<br />

targeted agents. Methods: Data was pooled from 5 phase II trials evaluating<br />

GEM-based cytotoxic doublets plus BEV in APC. 1 o endpoint was OS. 2o endpoints included ORR, CA19-9 response, and adverse events (AEs).<br />

Kaplan-Meier methods estimated time-to-event endpoints. The Cox proportional<br />

hazard model estimated univariate hazard ratios (HR) <strong>of</strong> death.<br />

Results: Of 261 pts, 90.7% were Caucasian, 95.4% had an ECOG PS 0-1,<br />

and 91.6% had metastatic disease. Median age � 60y. Pooled OS data (in<br />

mos), stratified for PS and stage, is shown in the table. ORR across all<br />

trials: CR 1.6%, PR 22.9%, SD 50.8%, PD 20.2%, NA 4.7%. HR for pts<br />

who achieved disease control (CR/PR/SD) was 0.35 vs. those with PD (95%<br />

CI 0.23-0.54, p�0.001). 76.5% <strong>of</strong> pts had elevated baseline CA19-9; <strong>of</strong><br />

these, 62% achieved �50% reduction (HR 0.50; 95% CI 0.34-0.73,<br />

p�0.001). BEV-related AEs �grade 3: HTN (10.6%), hemorrhage (9.5%),<br />

VTE (10.1%), cardiac events (3.4%), and bowel perforation (2.2%).<br />

Median OS in pts with grade 3-4 HTN was 13.4 mos vs. 9.8 mos in those<br />

without (HR 0.77; 95% CI 0.48-1.24, p�0.29). Conclusions: Recognizing<br />

the limitations <strong>of</strong> single-arm phase II trial design and cross-study comparisons,<br />

these results compare favorably to those from CALGB 80303. The<br />

standard paradigm <strong>of</strong> GEM �/- drug X in clinical trial development for APC<br />

needs to be reconsidered. Based on our data as well as the recent phase III<br />

FOLFIRINOX study, building on more intensive combination chemo regimens<br />

in future trials may represent a better strategy, especially for pts with<br />

good PS.<br />

gem/ox �<br />

bev<br />

gem/cap �<br />

bev<br />

gem/cis �<br />

bev<br />

gem/doc �<br />

bev<br />

gem/ox �<br />

bev<br />

Phase II<br />

total<br />

247s<br />

CALGB<br />

80303<br />

N 50<br />

Median OS 11.7<br />

PS<br />

0<br />

1<br />

2<br />

Stage<br />

Locally advanced<br />

Metastatic<br />

50<br />

9.8<br />

52<br />

8.4<br />

27<br />

10.4<br />

79<br />

8.4<br />

179†<br />

9.9<br />

11.1<br />

9.1<br />

3.3<br />

14.2<br />

9.5<br />

602<br />

5.8 - 5.9<br />

7.9<br />

4.8<br />

2.4<br />

9.9<br />

5.7<br />

† Reflects 4 studies with available raw data.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


248s Gastrointestinal (Noncolorectal) Cancer<br />

4039 General Poster Session (Board #40F), Mon, 8:00 AM-12:00 PM<br />

Baseline albumin (b-alb) as a potential predictive biomarker for the efficacy<br />

<strong>of</strong> bevacizumab (B) therapy (tx) in patients (pts) with advanced pancreas<br />

cancer (APCA): A comparative analysis. Presenting Author: Ludmila Katherine<br />

Martin, The Ohio State University Medical Center, Columbus, OH<br />

Background: Phase III studies <strong>of</strong> B in unselected pts with APCA have<br />

demonstrated no improvement in outcome. Recent data suggest certain<br />

subsets <strong>of</strong> APCA patients may benefit from B. Lower b- alb results in a<br />

15-20% increased rate <strong>of</strong> B clearance that may decrease exposure to B.<br />

The resulting clinical implications are not well understood. We evaluated<br />

the potential predictive and prognostic role <strong>of</strong> b-alb in pts with APCA<br />

receiving gemcitabine (G)-based tx with or without B. Methods: Relevant<br />

data were collected from 3 prospective phase II studies <strong>of</strong> G-based tx. Pts<br />

were grouped according to exposure to B (Gr 1) or no B (Gr 2) and by b-alb<br />

� 3.4 g/dL (� LLN) or � 3.4 g/dL (�LLN). Univariate and multivariate<br />

analyses <strong>of</strong> clinical outcome (OS, TTP) were conducted for each group and<br />

all pts. Results: 100 pts (46M, 54F) with median age 63 (range 28-82)<br />

were included. 94% had stage IV. Median b-alb was similar in both groups.<br />

<strong>Clinical</strong> outcomes by alb are outlined in the table. In Gr 1 but not Gr 2,<br />

b-alb � 3.4 g/dL was significantly associated with improved OS and TTP.<br />

For pts with b-alb �3.4 g/dL, maintenance <strong>of</strong> alb �3.4 g/dL throughout tx<br />

was significantly associated with improved survival in Gr 1 but not Gr 2.<br />

Multivariate analysis revealed significant association between alb � 3.4<br />

and OS regardless <strong>of</strong> B status (p�0.004) although this was strongly<br />

influenced by the survival differential in Gr 1. Conclusions: APCA pts with<br />

b-alb � 3.4 g/dL appear to derive significant benefit from B and this benefit<br />

is most pronounced in pts who maintain alb � 3.4 g/dL throughout B tx.<br />

This finding was not observed in pts treated without B. b-alb � 3.4 g/dL<br />

including maintenance <strong>of</strong> alb � 3.4 g/dL during B tx may predict for<br />

improved efficacy <strong>of</strong> B in APCA. These findings require further investigation<br />

in larger prospective trials.<br />

<strong>Clinical</strong> outcome according to alb.<br />

Group 1 (N�42) Group 2 (N�58)<br />

b-alb � 3.4 g/dL b-alb � 3.4 g/dL p b-alb � 3.4 g/dL b-alb � 3.4 g/dL p<br />

(N�28)<br />

(N�14)<br />

(N�43)<br />

(N�15)<br />

mOS (m) 10.7 3.1 0.00175 7.3 5.4 0.22<br />

mTTP (m) 7.7 2.7 0.009 3.9 2.8 0.76<br />

alb � 3.4 w/tx alb 2to � 3.4 w/tx p alb�3.4 w/tx alb 2to � 3.4 w/tx p<br />

(N�9)<br />

(N�19)<br />

(N�8)<br />

(N�29)<br />

mOS (m) 20.1 8.6 0.003 8.9 5.9 0.63<br />

4041 General Poster Session (Board #40H), Mon, 8:00 AM-12:00 PM<br />

Development <strong>of</strong> the SP120 rabbit monoclonal antibody for determining<br />

hENT1 status and predicting response to gemcitabine in pancreatic ductal<br />

adenocarcinoma. Presenting Author: Stefan Hubert Boeck, Department <strong>of</strong><br />

Hematology and Oncology, Klinikum Grosshadern and Comprehensive<br />

Cancer Center, LMU Munich, Munich, Germany<br />

Background: Human equilibrative nucleoside transporter 1 (hENT1) is the<br />

primary membrane channel through which gemcitabine (Gem) enters<br />

pancreatic tumor cells. Patients whose tumors have low expression <strong>of</strong><br />

hENT1 may derive little benefit from Gem therapy. Methods: We have<br />

developed and analytically validated a rabbit monoclonal antibody, SP120,<br />

immunohistochemistry (IHC) in vitro diagnostic (IVD) assay, used on the<br />

VENTANA BenchMark ULTRA automated slide staining platform, for<br />

assessing hENT1 expression in pancreatic adenocarcinoma. Results: SP120<br />

was shown to be sensitive and specific for membrane expression <strong>of</strong> hENT1<br />

with variable expression demonstrated across a panel <strong>of</strong> primary pancreatic<br />

adenocarcinoma samples. Using retrospectively collected primary tumor<br />

samples (n � 201) from a randomized controlled clinical adjuvant trial<br />

(RTOG 97-04), we developed and verified a hENT1 scoring algorithm and<br />

cut-<strong>of</strong>f for identifying patients least likely to benefit from Gem (hENT1low).<br />

We confirmed the predictive value <strong>of</strong> the hENT1 assay by showing<br />

there was no association with clinical outcome in the 5FU-treated control<br />

group. The distribution <strong>of</strong> hENT1 expression was similar between these<br />

samples and an independent set <strong>of</strong> 130 pancreatic adenocarcinoma<br />

specimens from the AIO-PK0104 study (77 were confirmed metastatic<br />

biopsies). Importantly, 100% concordance in hENT1 status (high vs. low)<br />

was demonstrated between 16 primary tumors and paired, contemporaneous<br />

metastatic lesions. Overall, using this cut-<strong>of</strong>f, approximately two thirds<br />

<strong>of</strong> pancreatic tumors displayed low-hENT1 expression across the different<br />

data sets (n � 363). Conclusions: The hENT1 IHC IVD has been developed<br />

and is being used in a pivotal trial <strong>of</strong> a novel lipid-conjugated Gem<br />

(CO-101, which enters tumor cells independently <strong>of</strong> hENT1), vs. Gem in<br />

hENT1-low pancreatic cancer patients (NCT01124786). This study is the<br />

first prospective test <strong>of</strong> the hypothesis that gemcitabine is not active in<br />

tumors with low hENT1 expression.<br />

4040 General Poster Session (Board #40G), Mon, 8:00 AM-12:00 PM<br />

Antitumor activity <strong>of</strong> nab-paclitaxel and gemcitabine in resectable pancreatic<br />

cancer. Presenting Author: Rafael Alvarez-Gallego, Centro Integral<br />

Oncológico Clara Campal, Madrid, Spain<br />

Background: Nab-paclitaxel in combination with gemcitabine has shown<br />

interesting clinical activity in patients with advanced pancreatic cancer<br />

(PDA) likely related to its ability to eliminate pancreas cancer stroma. In<br />

this study we explore clinical and pathological effects <strong>of</strong> the combination in<br />

patients with operable PDA. Methods: Patients with resectable o borderline<br />

resectable pancreatic cancer were treated with gemcitabine(1000mg/m2<br />

days 1, 8 and 15) and nab-paclitaxel(125mg/m2 days 1, 8 and 15) for two<br />

cycles prior to surgery. Response was assessed by FDG-PET, CA199 levels<br />

and elastography, an EUS-based non invasive assessment <strong>of</strong> tumor stroma.<br />

Results: 16 patients were included into the study. 2 patients (12.5%)<br />

showed a progression disease (both with hepatic metastases) and were not<br />

operated. Median value for PET SUVmax decreased from 7,1 pre-treatment<br />

to 4,6 post-treatment(p�0.004), including 7(50%) <strong>of</strong> patients with a<br />

partial metabolic response and the mean CA199 decreased from 2654 to<br />

52(p�0.02) with 43% patients having a more that 75 % decrement in<br />

tumor marker. The elastography ratio value diminished from 36 pretreatment<br />

to 18 post-treatment(p�0.003) and correlated with improvement<br />

in SUVmax(p�0.04) and CA199 response(p�0,07). Grade 3-4<br />

toxicities were neutropenia in 18% (none febrile neutropenia), thrombocytopenia<br />

in 12.5 and 6.2% transaminase elevation. So far 9 patients have<br />

been operated and in 8(89%) a complete resection (R0) was achieved. 1<br />

patient had a complete pathological response and 4 patients had near<br />

complete responses with only a few(� 5%) residual tumor. In-depth<br />

analysis <strong>of</strong> stromal composition after treatment showed, compared to a<br />

series <strong>of</strong> 10 cases untreated and treated with conventional chemoradiation,<br />

decreased my<strong>of</strong>ibroblast content, increase vessel density and distorted<br />

collagen fibers. Conclusions: Neoadjuvant treatment with gemcitabine plus<br />

nab-paclitaxel is feasible and results in significant clinical activity. Noninvasive<br />

elastography appears and attractive method to monitor tumor<br />

response. The rate <strong>of</strong> pathological responses and R0 resections in substantial<br />

for this setting. Biological studies <strong>of</strong> resected specimens show unique<br />

effects in tumor stroma.<br />

4042 General Poster Session (Board #41A), Mon, 8:00 AM-12:00 PM<br />

A phase III trial <strong>of</strong> ganitumab (GAN, AMG 479) with gemcitabine (G) as<br />

first-line treatment (tx) in patients (pts) with metastatic pancreatic cancer<br />

(MPC): An analysis <strong>of</strong> safety from the GAMMA trial (GEM and AMG 479 in<br />

Metastatic Adenocarcinoma <strong>of</strong> the Pancreas). Presenting Author: Charles<br />

S. Fuchs, Dana-Farber Cancer Institute, Boston, MA<br />

Background: GAN is an investigational, fully human, monoclonal antibody<br />

inhibitor <strong>of</strong> IGF1R. GAMMA is assessing the safety and efficacy <strong>of</strong> GAN plus<br />

G as first-line tx in MPC pts (<strong>Clinical</strong>Trials.gov ID: NCT01231347).<br />

Methods: This is an ongoing, global, phase III, double-blind study. Pts are<br />

randomized 2:2:1 to receive placebo, GAN 12 mg/kg, or GAN 20 mg/kg (IV;<br />

days 1 and 15 Q28D) with G 1000 mg/m2 (IV; days 1, 8, and 15 Q28D).<br />

The planned sample size is 825. Primary endpoint: overall survival. Key<br />

secondary endpoints: progression-free survival, 1-year survival rate, patientreported<br />

outcomes, and safety. This study includes multiple planned safety<br />

analyses conducted by an independent Data Monitoring Committee (DMC).<br />

The current predefined safety analyses occurred when 150pts received � 1<br />

cycle <strong>of</strong> tx. Results: As <strong>of</strong> Sep 16, 2011, 207 pts are included in this<br />

aggregate analysis: 50% male; median age, 63 yrs (range 36-83); ECOG<br />

PS 0/1, 50%/50%. Of the 207 pts, 204 pts received study tx, and 61 pts<br />

ended study tx. Most frequent adverse events (AE) are shown (table). Ten<br />

pts (5%) died during or within 30 days <strong>of</strong> the end <strong>of</strong> tx. Seven events were<br />

attributed to or associated with disease progression. One event <strong>of</strong> cardiac<br />

failure was reported to be possibly tx related. Pulmonary embolism was<br />

suspected but not confirmed. Conclusions: The GAMMA study continues<br />

per protocol. The only grade 3/4 AE occurring in more than 5% <strong>of</strong> patients<br />

to date is neutropenia.<br />

Any grade AE occurring in > 15% <strong>of</strong> pts or higher grade AE in > 3% <strong>of</strong> ptsa .<br />

Any grade-n(%)<br />

CTCAE v3.0<br />

Grade 3/4-n(%)<br />

CTCAE v3.0<br />

AE, MedDRA terms<br />

Thrombocytopenia b<br />

Neutropenia<br />

79 (39) 8 (4)<br />

c<br />

72 (35) 47 (23)<br />

Nausea 69 (34) 4 (2)<br />

Fatigue 56 (27) 9 (4)<br />

Decreased appetite 45 (22) 2 (1)<br />

Vomiting 38 (19) 4 (2)<br />

Pyrexia 36 (18) 0 (0)<br />

Diarrhea 35 (17) 1 (0)<br />

Leukopenia 34 (17) 7 (3)<br />

Alanine aminotransferase increased 28 (14) 9 (4)<br />

Hyperglycemia 27 (13) 9 (4)<br />

Aspartate aminotransferase increased 23 (11) 7 (3)<br />

Asthenia 21 (10) 6 (3)<br />

a Analysis includes all 3 tx arms and excludes 3 pts who did not receive any tx; b Includes<br />

platelets decreased; c Includes neutrophils decreased.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


4043 General Poster Session (Board #41B), Mon, 8:00 AM-12:00 PM<br />

Phase I/II study <strong>of</strong> 90Y-clivatuzumab tetraxetan ( 90Y-hPAM4) combined<br />

with gemcitabine (Gem) in advanced pancreatic cancer (APC): Final<br />

results. Presenting Author: Allyson J. Ocean, New York Presbyterian<br />

Hospital-Weill Cornell Medical College, New York, NY<br />

Background: A Phase I/II trial evaluated single and repeated cycles <strong>of</strong><br />

fractionated radioimmunotherapy (RAIT) with 90Y-labeled humanized mAb<br />

( 90Y-hPAM4) plus Gem as first-line therapy in Stage 3-4 APC. Methods:<br />

Cycles <strong>of</strong> Gem once-weekly x 4 with 90Y-hPAM4 on wks 2, 3 and 4 were<br />

repeated until progression, withdrawal or unacceptable toxicity. In <strong>Part</strong> I,<br />

90 2 Y doses were escalated with Gem fixed at 200 mg/m . In <strong>Part</strong> II, Gem was<br />

increased up to 1000 mg/m2 , with 90Y fixed at 12 mCi/m2 for cycle 1 and<br />

lowered for retreatment. Results: Of 100 pts entered, 10 withdrew early,<br />

while 90 (73 stage IV) received 1-4 cycles. In <strong>Part</strong> I, 38 pts received<br />

90 2 Y-hPAM4 weekly x3at6.5, 9, 12, or 15 mCi/m , with the same cycle<br />

repeated 1-3 times in 13 pts. By CT-RECIST criteria, 6 pts (16%) had PRs<br />

and 16 (42%) had stabilization as best response (58% disease control).<br />

After cycle 1, 52% (13/25) with PET-avid images had �25% SUV<br />

reduction, and 33% (9/27) with elevated CA19-9 levels decreased by<br />

�50%. The median OS was 7.7 mo., but 11.8 mo. for retreated pts [46%<br />

(6/13) survived �1 yr.], and with improved efficacy at higher 90Y doses.<br />

NCI-CTCv3 Grade 3-4 platelets or ANC developed in 20/38 (53%) after<br />

cycle 1 (all reversible to Grade 1) and in all retreated pts (irreversible in 4/9<br />

pts at 12 or 15 mCi/m2 ). In <strong>Part</strong> II, 52 pts received increased Gem without<br />

evidence <strong>of</strong> improved efficacy, while 13 pts were retreated with more<br />

acceptable toxicity at lower 90Y doses <strong>of</strong> 6.5 or 9 mCi/m2 . Treatment was<br />

well tolerated with no infusion reactions. Infections requiring IV antibiotics<br />

occurred at a low rate and responded to appropriate coverage (bacteremia/<br />

sepsis, 7%; febrile neutropenia, 4%; ascending cholangitis, 3%; pneumonia,<br />

2%; others 1%). One case <strong>of</strong> bleeding occurred, due to rectal tumor<br />

invasion. Anecdotal reports <strong>of</strong> good performance and decreased pain<br />

medication requirements require further validation. Conclusions: Fractionated<br />

RAIT with 90Y-hPAM4 combined with low-dose 200 mg/m2 GEM<br />

appears promising as a treatment regimen for APC. Hematologic toxicity<br />

was dose limiting. A 90Y-hPAM4 dose <strong>of</strong> 12 mCi/m2 for cycle 1 and 6.5<br />

mCi/m2 for cycle 2 have been selected as suitable for further clinical<br />

development in the first-line setting.<br />

4045 General Poster Session (Board #41D), Mon, 8:00 AM-12:00 PM<br />

A phase II multi-institutional study to evaluate gemcitabine and fractionated<br />

stereotactic body radiotherapy for unresectable, locally advanced<br />

pancreatic adenocarcinoma. Presenting Author: Joseph M. Herman, Sidney<br />

Kimmel Comprehensive Cancer Center at Johns Hopkins University,<br />

Baltimore, MD<br />

Background: Phase I/II studies with single-fraction (25 Gy) stereotactic<br />

body radiotherapy (SBRT) for pancreatic ductal adenocarcinoma (PDA)<br />

have shown local progression free survival (LPFS) rates <strong>of</strong> �90%, but are<br />

limited by late GI toxicity and minimal tumor response. We performed a<br />

phase II multi-center trial <strong>of</strong> gemcitabine (GEM) and fractionated SBRT to<br />

determine if a high rate <strong>of</strong> LPFS with reduced toxicity could be achieved.<br />

Methods: After multidisciplinary review, 32 pts with locally advanced PDA<br />

received GEM in sequence with SBRT (6.6 Gy in 5 consecutive daily<br />

fractions, 33 Gy total). LPFS, metastasis free survival (MFS), and overall<br />

survival (OS) were measured from date <strong>of</strong> tissue diagnosis. Objective tumor<br />

response (OTR) was assessed by RECIST/PERCIST. EORTC QLQ-C30/<br />

PAN26 questionnaires were used to measure QOL. Results: Median f-up<br />

was 12 mos (range, 2-23). Mean age was 69.9 yrs (SD, 9.8) and 62% were<br />

male. Pts received a mean <strong>of</strong> 2.2 (SD, 1.0) GEM doses prior to SBRT and<br />

8.3 (SD, 5.6) doses total. All pts completed SBRT. Median OS was 15.9<br />

months (95% CI, 12.7-18.8). Stratification by CA19-9 � or � 90 at<br />

diagnosis yielded a hazard ratio <strong>of</strong> 6.2 for � 90 (p�0.021). Median LPFS<br />

has not been reached and median MFS was 10.2 mos (95% CI, 2.9-17.5).<br />

LPFS rate at 1 year was 87%. OTR on CT was seen in 41%, while 41% had<br />

stable disease and 18% progressed. Tumor metabolic activity decreased in<br />

17/18 patients with pre/post-SBRT PET available. Mean peak SUV was 4.0<br />

pre-SBRT versus 2.4 post-SBRT (p�0.002). Median CA19-9 was reduced<br />

from 124.7 prior to SBRT to 43.9 afterwards. Acute toxicity included:<br />

grade 2 anorexia (37%), fatigue (28%), nausea (22%), abd pain (19%),<br />

weight loss (9%), diarrhea (3%); gr 3 nausea (9%); and gr 4 nausea (6%).<br />

Late gr �3 GI toxicity was seen in 9%. Mean QOL score 4 wks post-SBRT<br />

was similar to baseline (p�0.38). At 6 mos there was a trend towards<br />

improved QOL (p�0.07). Conclusions: Fractionated SBRT with GEM<br />

achieves high rates <strong>of</strong> LPFS and tumor response. Minimal grade �3 acute<br />

and late toxicity was observed. SBRT is more likely to benefit patients with<br />

Ca-19-9 �90. A combination <strong>of</strong> SBRT with more aggressive chemotherapy<br />

may further improve outcomes.<br />

Gastrointestinal (Noncolorectal) Cancer<br />

249s<br />

4044 General Poster Session (Board #41C), Mon, 8:00 AM-12:00 PM<br />

Evaluation <strong>of</strong> predictive factors for thromboembolic events in patients with<br />

advanced pancreatic cancer: Evaluation <strong>of</strong> the CONKO-004 (PROSPECT)<br />

study cohort. Presenting Author: Uwe Pelzer, Universitätsmedizin Berlin,<br />

Charité Centrum für Tumormedizin, Berlin, Germany<br />

Background: Patients (pts) with advanced pancreatic cancer (APC) <strong>of</strong>ten<br />

suffer from symptomatic thromboembolic events (sVTE). The CONKO-004<br />

trial showed that the low molecular weight heparin (LMWH) enoxaparin<br />

reduces sVTE (p�0.05, number needed to treat: 12) without increasing the<br />

rate <strong>of</strong> major bleeding when prophylactically applied. Our goal was to<br />

identify predictive factors for sVTE in pts with APC undergoing first-line<br />

chemotherapy. Methods: We analyzed the 152 (out <strong>of</strong> 312) pts randomized<br />

in the observation group.SVTE incidence was 9.9%. To identify foremost<br />

risk factors we used clinical parameters like performance status, stage,<br />

grading, primary or recurrent, gender, age, body mass index, erythropoietin<br />

stimulating agents (darbepoetin), as well as baseline laboraty parameters<br />

such as creatinine, hemoglobin, WBC, platelets, INR, ptt, CEA, carboanhydrat<br />

19-9, AST, ALT, AP and GGT. The multivariate logistic regression<br />

model with forward stepwise selection process was used for this estimation.<br />

With reference to a previously proposed scoring system (Khorana et al;<br />

Blood 2008) we evaluated the score as well. Results: No single parameter<br />

could be isolated demonstrating significant influence on the incidence <strong>of</strong><br />

sVTE in pts with APC. We verified the Kohrana scoring model with our<br />

152/312 pts. Applying the score to our pts we didn’t discriminate pts<br />

between the two high risks groups (Table). Conclusions: Predictive models<br />

may help to identify cancer pts at high risk for sVTE to consider preventive<br />

anticoagulation. Within a cancer entity at high risk for sVTE such as pts<br />

with APC we did not succeed in the identification <strong>of</strong> single predictive<br />

parameters.For pts with APC undergoing first line chemotherapy primary<br />

prevention with LMWH should therefore be considered for the whole group<br />

<strong>of</strong> pts for at least three months.<br />

0 1-2


250s Gastrointestinal (Noncolorectal) Cancer<br />

4047 General Poster Session (Board #41F), Mon, 8:00 AM-12:00 PM<br />

Multicenter phase II trial <strong>of</strong> temsirolimus (TEM) and bevacizumab (BEV) in<br />

pancreatic neuroendocrine tumor (PNET): Results <strong>of</strong> a planned interim<br />

efficacy analysis. Presenting Author: Timothy J. Hobday, Mayo Clinic,<br />

Rochester, MN<br />

Background: PNET has long had few effective therapies other than chemotherapy.<br />

Recent placebo-controlled phase III trials <strong>of</strong> the mTOR inhibitor<br />

everolimus and the VEGF/ PDGF receptor inhibitor sunitinib noted improved<br />

progression-free survival (PFS). However, objective response rates<br />

(RR) with these agents are still �10%. Preclinical studies suggest<br />

enhanced anti-tumor effects with combined mTOR and VEGF targeted<br />

therapy. Methods: We conducted a phase II trial <strong>of</strong> the mTOR inhibitor TEM<br />

(25 mg IV q week) and the VEGF-A monoclonal antibody BEV (10 mg/kg IV<br />

q 2 weeks) in patients (pts) with well or moderately differentiated PNET<br />

and progressive disease by RECIST within 7 months <strong>of</strong> study entry.<br />

Co-primary endpoints were RR and 6-month PFS. Planned enrollment is 50<br />

patients, with interim analysis for futility after the first 25 evaluable pts. Pts<br />

had no prior mTOR or VEGF targeted agents, ECOG PS 0-1, and adequate<br />

hematologic and organ function. Continued octreotide was allowed, but not<br />

required. Prior interferon, embolization, and � 2 chemotherapy regimens<br />

were allowed. Results: Confirmed PR was documented in 13 <strong>of</strong> the first 25<br />

(52%) evaluable patients. 21 <strong>of</strong> 25 (84%) patients were progression-free<br />

at 6 months. Both endpoints exceeded the protocol-defined criteria to<br />

continue enrollment. For 36 evaluable patients, the most common grade<br />

3-4 adverse events attributed to therapy were hypertension (14%), leukopenia<br />

(11%), lymphopenia (11%), hyperglycemia (11%), mucositis (8%),<br />

hypokalemia (8%), and fatigue (8%). Conclusions: The combination <strong>of</strong><br />

TEM/BEV has substantial activity in a multi-center phase II trial with RR <strong>of</strong><br />

52%, well in excess <strong>of</strong> single targeted agents in PNET. 6-month PFS was a<br />

notable 84% in a population <strong>of</strong> patients with RECIST criteria progression<br />

within 7 months <strong>of</strong> study entry. Accrual is ongoing.<br />

4049 General Poster Session (Board #41H), Mon, 8:00 AM-12:00 PM<br />

Addition <strong>of</strong> algenpantucel-L immunotherapy to standard <strong>of</strong> care (SOC)<br />

adjuvant therapy for pancreatic cancer. Presenting Author: Jeffrey M<br />

Hardacre, University Hospitals Case Medical Center, Cleveland, OH<br />

Background: Resected pancreatic cancer carries a one year survival rate <strong>of</strong><br />

~65%. Algenpantucel (HyperAcute-Pancreas, HAPa) exploits the hyperacute<br />

rejection mechanism <strong>of</strong> xenotransplantation by administering genetically<br />

modified, irradiated, allogeneic tumor cells expressing aGal moieties<br />

on their surface. We propose that addition <strong>of</strong> algenpantucel-L to SOC<br />

adjuvant therapy may improve survival for patients with resected pancreatic<br />

cancer. Methods: Open-label, dose-finding, Phase 2 study (16 sites)<br />

evaluating HAPa (100/300 M cells / dose) � SOC (RTOG-9704: Gemcitabine<br />

� 5-FU-XRT) for resected pancreatic cancer patients. Primary<br />

endpoint: 1-year disease-free survival (DFS). Secondary Endpoints: overall<br />

survival (OS), toxicity and immunologic analysis. Results: : 70 patients,<br />

median age 62 years, 47% female, 81% lymph node positive, median<br />

tumor size 3.2 cm, and 17% post-operative CA 19-9 � 180 received SOC<br />

� HAPa. 1-year DFS <strong>of</strong> 63% and OS <strong>of</strong> 86% compares favorably to<br />

historical controls (~45%% and 65%, respectively) for this high risk<br />

population. Subgroup analysis <strong>of</strong> patients receiving 300M cells /dose<br />

shows a higher 12-month DFS compared to 100M cells/dose, 81% vs. 52%<br />

(p � 0.02). Likewise, patients receiving 300M cells/dose tended to better<br />

1-year survival compared to a 100M cells/dose, 96% vs. 80% (p � 0.053).<br />

Forty-four (63%) developed strong (G1-G3) skin reactions at the injection<br />

sites including many long term survival patients with periodic skin flares at<br />

injection sites far beyond their last vaccination. In 13/45 (29%) tested<br />

patients, an increase <strong>of</strong> �30% in anti-mesothelin Ab titer was seen after<br />

immunization. Thirty-nine (56%) presented with eosinophilia (range: 5-<br />

45%), including one patient with persistent eosinophilia and stable lung<br />

metastasis for over 2 years. Conclusions: Addition <strong>of</strong> HAPa to standard<br />

adjuvant therapy for resected pancreatic cancer shows promising immunological<br />

activation and may improve survival. A multi-institutional, phase 3<br />

study is currently underway.<br />

4048 General Poster Session (Board #41G), Mon, 8:00 AM-12:00 PM<br />

A phase I study <strong>of</strong> nab-paclitaxel (A), gemcitabine (GEM), and capecitabine<br />

(X) in patients with metastatic pancreatic adenocarcinoma (MPA). Presenting<br />

Author: Thach-Giao Truong, University <strong>of</strong> California, San Francisco<br />

Comprehensive Cancer Center, San Francisco, CA<br />

Background: GEM-based doublets, such as GEM � capecitabine (X), may<br />

be beneficial in select pts with MPA. Given preclinical evidence <strong>of</strong> synergy<br />

when a taxane is added to GEM � X, as well as recent phase I/II study<br />

results showing substantial activity with GEM � nab-paclitaxel (A) (ORR <strong>of</strong><br />

48%, median OS � 12 mos at MTD), we conducted a phase I study<br />

evaluating the 3-drug combination <strong>of</strong> A, GEM, and X (AGX) given biweekly<br />

in pts with MPA. Methods: Pts with previously untreated MPA and ECOG PS<br />

0-1 were eligible. A (100-150 mg/m2 ) and GEM (750-1000 mg/m2 at 10<br />

mg/m2 /min) were both administered on day 4, and X (500-1000 mg/m2 bid) on days 1-7, <strong>of</strong> each 14-day cycle. A 3�3 dose escalation design was<br />

used, with expanded cohort at MTD. Primary objective was to establish<br />

MTD <strong>of</strong> this regimen; secondary objectives include safety and preliminary<br />

assessment <strong>of</strong> efficacy. DLT definitions: gr 3-4 ANC or neutropenic fever; gr<br />

4 plts; gr 2-4 hand-foot syndrome (HFS), neuropathy or diarrhea; or other gr<br />

3-4 non-heme toxicity. Results: Fifteen patients were enrolled across two<br />

dose levels (age range, 45-74 y). Final MTD was established at dose level 0,<br />

consisting <strong>of</strong> A 100 mg/m2, GEM 750 mg/m2, and X 750 mg/m2 bid. At<br />

dose level 1, two <strong>of</strong> 4 pts experienced DLTs (gr 3 LFT elevation, gr 3 ANC).<br />

For the entire study cohort, 10 pts (67%) experienced at least one gr 3-4<br />

AE, including LFTs (20%), N/V (13%), and fatigue (7%). Gr 3-4 heme<br />

toxicity was uncommon. Other notable AEs (any grade): fatigue (87%),<br />

rash/HFS (67%), N/V (53%), diarrhea (40%), neuropathy (33%). Median #<br />

cycles received � 4 (range, 2-16). 73% <strong>of</strong> pts d/c’ed study treatment due<br />

to disease progression. Best response for the entire cohort (n�14 evaluable<br />

pts): 2 PR, 8 SD, 4 PD (disease control rate � 71%). Of 12 pts with<br />

elevated CA19-9 at baseline, 5 (42%) had �50% biomarker decline.<br />

Conclusions: Recognizing the limits <strong>of</strong> cross-study comparisons and small<br />

sample size, these results do not match those reported at MTD in the phase<br />

I/II trial <strong>of</strong> GEM (1000 mg/m2) � A (125 mg/m2 ). The modest activity seen<br />

with this AGX regimen may have resulted from suboptimal dosing, and<br />

suggests that dose intensity may be an important factor when trying to<br />

incorporate nab-paclitaxel into multidrug regimens.<br />

4050 General Poster Session (Board #42A), Mon, 8:00 AM-12:00 PM<br />

A phase I/II study <strong>of</strong> the MEK inhibitor BAY 86-9766 (BAY) in combination<br />

with gemcitabine (GEM) in patients with nonresectable, locally advanced or<br />

metastatic pancreatic cancer (PC): Phase I dose-escalation results. Presenting<br />

Author: Jean-Luc Van Laethem, Hôpital Universitaire Erasme, Brussels,<br />

Belgium<br />

Background: Targeting the RAS–RAF–MEK–ERK pathway may be useful in<br />

the treatment <strong>of</strong> PC, as 75–90% <strong>of</strong> PCs have KRAS mutation. BAY is an<br />

orally bioavailable, potent, allosteric MEK 1/2 inhibitor that showed<br />

single-agent activity, and synergistic activity with GEM, in ectopic, orthotopic,<br />

syngenic and patient-derived xenograft PC models. Methods: The<br />

phase I part aimed to determine the maximum tolerated dose (MTD) <strong>of</strong> BAY<br />

in combination with GEM, and the pharmacokinetics <strong>of</strong> BAY and GEM.<br />

3�3 study design was used. After informed consent, eligible patients with<br />

advanced PC received GEM 1000 mg/m2 (30-min, once-weekly IV infusion<br />

for 7 out <strong>of</strong> 8 weeks in cycle 1 [C1], and 3 out <strong>of</strong> 4 weeks in subsequent<br />

cycles) and oral BAY 30 mg BID (dose level 1 [DL1]) or 50 mg BID (DL2).<br />

No escalation beyond DL2 was planned. MTD was the highest dose at which<br />

maximum 1 out <strong>of</strong> 6 evaluable patients displayed dose-limiting toxicities<br />

(DLT) during C1. Results: As <strong>of</strong> 6 Jan 2012, 17 patients were enrolled and<br />

treated (10 at DL1, 7 at DL2). DL1 cohort has completed; DL2 evaluation<br />

is ongoing. At DL1, DLTs occurred in 1/6 evaluable patients: a patient with<br />

extensive liver metastasis developed chemotherapy-associated steatohepatitis<br />

(CASH) and died <strong>of</strong> hepatic failure. Rare CASH cases have been<br />

reported to be associated with GEM alone, but co-involvement <strong>of</strong> BAY<br />

currently cannot be ruled out. To date, 3 patients have completed C1 at<br />

DL2 without DLTs. DL2 completion is expected in March 2012. BAY�GEM<br />

showed a manageable tolerability pr<strong>of</strong>ile. The most frequent treatmentrelated<br />

grade 3–4 adverse event (AE) at any DL was neutropenia (n�6).<br />

The most frequent clinically relevant BAY-related AE was acneiform rash<br />

(n�11), which was mostly grade 1–2 (one case grade 3) and manageable.<br />

BAY�GEM showed evidence <strong>of</strong> clinical efficacy: partial responses were<br />

seen in 4/10 patients at DL1 and 1/3 patients at DL2. No pharmacokinetic<br />

interaction between BAY and GEM was seen at DL1. Conclusions: In<br />

patients with advanced PC, BAY 30 mg BID in combination with GEM had a<br />

manageable safety pr<strong>of</strong>ile, with acneiform rash as the clinically most<br />

relevant toxicity attributable to BAY.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


4051 General Poster Session (Board #42B), Mon, 8:00 AM-12:00 PM<br />

Updated survival analysis <strong>of</strong> preoperative gemcitabine (gem) plus bevacizumab<br />

(bev)-based chemoradiation for resectable pancreatic adenocarcinoma.<br />

Presenting Author: Rachna T. Shr<strong>of</strong>f, University <strong>of</strong> Texas M. D.<br />

Anderson Cancer Center, Houston, TX<br />

Background: Preop therapy for resectable pancreatic cancer (PC) maximizes<br />

access to multimodality therapy and increases the R0 resection rate. Based<br />

on our phase I chemoradiation (chemoRT) trial in PC patients, we<br />

hypothesized that the addition <strong>of</strong> bev to gem-based chemoRT in pts with<br />

resectable PC may allow more pts to undergo pancreaticoduodenectomy<br />

(PD), improve the rate <strong>of</strong> R0 resections, and prolong overall survival.<br />

Methods: Pts with biopsy proven, stage I/II adenocarcinoma <strong>of</strong> the pancreatic<br />

head or uncinate process received 6 weekly infusions <strong>of</strong> gem (400<br />

mg/m2 ) and 3 infusions <strong>of</strong> bev (10 mg/kg, every 2 wks) with concomitant<br />

RT, 50.4 Gy at 1.8 Gy/fr. Pts were restaged 4-6 wks after the last dose <strong>of</strong><br />

radiation. Those without disease progression and with good PS underwent<br />

surgery. Pts with adequate recovery received bev (10 mg/kg) every 2 weeks<br />

for 3 mo starting ~ 6 wks after surgery. Results: This study enrolled 11 <strong>of</strong> a<br />

planned 31 pts but was terminated early due to unforeseen postop<br />

complications. Median age is 64 yrs; all pts had ECOG-PS 0-1. Median<br />

CA19-9 was 52. Median follow-up from surgery was 41 mths. All completed<br />

chemoRT; 10 underwent restaging and 1 pt died from cardiac arrest<br />

before restaging. 9 pts (90 %) went to surgery and underwent a successful<br />

R0 PD. Pathologic PR rate (�50 % tumor kill) was 56 %. As we have<br />

previously reported, preop grade 3-4 toxicities were infrequent and major<br />

postop complications occurred in 5 <strong>of</strong> the 9 pts (56%) who underwent PD.<br />

Median overall survival (OS) was 30.1 mths (range: 2.6 - 63.9) for the<br />

entire cohort. Of the 9 resected pts, median OS was 45.5 mths with 5 <strong>of</strong> 11<br />

pts (45%) achieving survival longer than best historical control (median<br />

58.2 mths). Conclusions: The addition <strong>of</strong> bev to our prior backbone <strong>of</strong><br />

gem-based preoperative chemoRT led to a higher resection rate (90%) than<br />

our previous protocols. Updated survival results <strong>of</strong> this study are promising.<br />

Taken together, these results may prompt further investigation <strong>of</strong> this<br />

regimen with modifications in the timing <strong>of</strong> bev delivery.<br />

4053 General Poster Session (Board #42D), Mon, 8:00 AM-12:00 PM<br />

International experts’ panel for the development <strong>of</strong> guidelines for the<br />

definition <strong>of</strong> time to event endpoints in clinical trials (DATECAN project):<br />

Results for pancreatic cancer. Presenting Author: Franck Bonnetain,<br />

Biostatistics and Epidemiology Unit, Centre Georges François Leclerc,<br />

Dijon, France and EA4184, College <strong>of</strong> Medicine, Dijon, France<br />

Background: Variability in the definition <strong>of</strong> survival endpoints in oncology<br />

trials was identified (Mathoulin et al.JCO 2008). Lack <strong>of</strong> a formal<br />

consensus could cause this, which limits inter-trial comparisons. The<br />

DATECAN project aimed at obtaining a formal consensus recommendation<br />

for defining survival endpoints for randomized clinical trials (RCTs) in the<br />

following cancer sites: pancreas, sarcoma/GISTs, breast, colorectal, gastric/<br />

œsophagus, head and neck, kidney-bladder. We report results for pancreatic<br />

cancer. Methods: Based on a literature review <strong>of</strong> RCTs (2006-2009),<br />

we identified survival endpoints and events currently used. A 2-round<br />

modified Delphi method using RAND scoring (range:1-9) was used to reach<br />

consensus. Academic research groups were contacted for participation in<br />

order to select clinicians and methodologists for Pilot and Scoring groups<br />

(�30 experts/localization). Results: The Pilot group identified 14 endpoints<br />

that needed definition through consensus, such as progression free survival<br />

(PFS), time-to-treatment failure, time to quality <strong>of</strong> life deterioration.<br />

Endpoint definitions were seeked by disease setting (detectable disease vs<br />

not). Amongst the 52 European experts contacted, 33 and 30 participated<br />

to the 1st and 2nd round respectively. The experts scored a total <strong>of</strong> 204<br />

events; a consensus was reached for 25 (12%) at the 1st round and 156<br />

(76%) at the 2nd round. As example PFS was defined as time interval<br />

between the date <strong>of</strong> randomization, and the day <strong>of</strong> first local, regional<br />

progression or occurrence <strong>of</strong> distant metastases (including liver or non liver<br />

metastases) or occurrence <strong>of</strong> 2nd pancreatic cancer or death (all causes),<br />

whichever occurs first. The consensus was finalized during a face-to-face<br />

meeting organized during the ESMO 2011 congress and general rules were<br />

proposed for final ratification. Conclusions: Based on this consensus, an<br />

European charter is being finalized and proposed for endorsement to all<br />

academic groups in order to harmonize results and to allow formal<br />

comparisons <strong>of</strong> pancreatic RCTs. The impact <strong>of</strong> these definitions on trial<br />

results will be also investigated in a further project.<br />

Gastrointestinal (Noncolorectal) Cancer<br />

251s<br />

4052 General Poster Session (Board #42C), Mon, 8:00 AM-12:00 PM<br />

A phase IB study <strong>of</strong> erlotinib in combination with gemcitabine and<br />

nab-paclitaxel in patients with previously untreated advanced pancreatic<br />

cancer: An Academic GI Cancer Consortium (AGICC) study. Presenting<br />

Author: Lawrence P. Leichman, Comprehensive Cancer Center at Desert<br />

Regional Medical Center, Palm Springs, CA<br />

Background: The combination <strong>of</strong> gemcitabine (gem) and nab-paclitaxel<br />

(nab) has demonstrated promising activity in advanced pancreatic cancer.<br />

Erlotinib (erl) adds modest benefit to gemcitabine. We initiated this phase<br />

IB study to evaluate the safety <strong>of</strong> the three drug combination and obtain<br />

preliminary evidence <strong>of</strong> efficacy. Methods: Patients (pts) with previously<br />

untreated locally advanced (la) or metastatic (met) pancreatic cancer with<br />

ECOG PS 0-1 were treated with gem and nab IV days 1,8,15 and once daily<br />

erl days 1-28 Q28 days. Standard 3�3 design was used with dose levels<br />

(DL) (gem,nab,erl): 1(1,000, 125, 100), -1 (1,000, 100, 100), -2 (1,000,<br />

75, 100), -3 (1,000,75,75). CT scans were obtained Q 2 cycles. DLT was<br />

defined as �grade (gr) 3 non-hematologic, febrile neutropenia, �gr 3<br />

thrombocytopenia, or missing �2 doses gem, nab or �5 doses erl within<br />

first cycle (C). Results: Nineteen pts were enrolled and completed a total <strong>of</strong><br />

62 cycles (range 0-11). Pt characteristics: M/F (9/10), White/Hispanic/AA<br />

(15/2/2), median age 63 (range 54-78), ECOG PS 1�11, met/la (12/7). In<br />

DL1, 1/3 pts had gr3 dehydration in C 2 and 1/3 had gr 4 neutropenia/<br />

sepsis in C 4. Although not formally DLT, 3 more pts were enrolled. Of the<br />

next 3 pts, 1 DLT <strong>of</strong> gr 3 diarrhea/gr 4 neutropenia in C 1 was noted and 1<br />

other pt DC’d therapy for neutropenia after 2 cycles. Of 3 pts in DL -1, 2<br />

DLTs (gr 3 optic neuropathy, too many missed doses) were observed. Of 3<br />

pts in DL -2, 2/3 had DLT (gr 3 esophagitis/fatigue and gr 3 transaminitis).<br />

Of 6 evaluable pts in DL -3, no DLTs were observed. Most common gr 3/4<br />

toxicities were neutropenia (9), dehydration, thrombocytopenia (3 each),<br />

and hypotension (2). Of 13 pts evaluable for response, 6 had partial<br />

response (46%), 5 stable disease (38%), and 2 progressive disease (15%)<br />

as best response. Median PFS and OS <strong>of</strong> entire cohort were 5.3 and 9.3<br />

months respectively. Conclusions: The combination <strong>of</strong> erlotinib with gemcitabine<br />

and nab-paclitaxel is not tolerable at standard single agent dosing <strong>of</strong><br />

all drugs. However, significant clinical activity was noted, even at DL -3.<br />

Further study <strong>of</strong> the combination will need to incorporate reduced dosing.<br />

4054 General Poster Session (Board #42E), Mon, 8:00 AM-12:00 PM<br />

Tumor IGF-1 expression as a predictive biomarker for IGF1R-directed<br />

therapy in advanced pancreatic cancer (APC). Presenting Author: Milind M.<br />

Javle, University <strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: IGF-1 up-regulates PC proliferation and invasiveness through<br />

activation <strong>of</strong> PI3K/Akt signaling pathway and down-regulates PTEN. We<br />

investigated IGF-1 expression in tissue and blood as potential predictive<br />

markers in phase II study <strong>of</strong> IGF1R-directed monoclonal antibody, MK-<br />

0646 in APC. Prior phase I established the MTD <strong>of</strong> MK0646 at 5 mg/kg<br />

with gemcitabine (G) and erlotinib (E) and 10 mg/kg with G alone. Methods:<br />

Patients (pts) with stage IV, previously untreated APC, ECOG PS 0-1,<br />

adequate hematologic and organ function were enrolled. Arm A: G 1,000<br />

mg/m2 over 100 min, weekly x 3, MK-0646 weekly x 4; Arm B: G 1000<br />

mg/m2 and MK-0646 � E 100 mg daily. Arm C (control) was G 1,000<br />

mg/m2 � E 100 mg. Cycles were repeated every 4 weeks. Pts were equally<br />

randomized in the 3 arms. Primary study objective was progression-free<br />

survival (PFS). Pre-treatment peripheral blood samples were measured for<br />

IGF-1 level by ELISA; archival core biopsies were analyzed for IGF-1 mRNA<br />

expression. RNA extraction from FFPE samples used Roche Transcriptor<br />

First Strand cDNA Synthesis Kit. TaqMan PreAmp technique was used to<br />

amplify target cDNA prior to TaqMan RT-PCR analysis. Cox proportional<br />

hazards model for PFS analyzed the interaction between tissue IGF-1<br />

expression and treatment. Results: 50 pts were enrolled (A�15,<br />

B�16,C�16 pts, 3 ineligible). Median PFS <strong>of</strong> arms A, B and C were 5.5<br />

months (95% CI: 3.9 – NA), 3.0 months (95% CI:1.8 – 5.6) and 2.0<br />

months (95% CI: 1.8 – NA), respectively (log-rank test; p � 0.17). Median<br />

OS <strong>of</strong> A was 11.3 months (95% CI: 8.9 – NA), B 8.9 months (95% CI: 5.3 –<br />

NA) and C 5.7 months (95% CI: 2.0 – NA) (log-rank test; p � 0.44). 35<br />

archival core biopsies were analyzed, 21 had adequate tissue for analysis.<br />

Using a Multivariable Cox proportional hazards model for PFS, where IGF-1<br />

was dichotomized at the median, there was a 76% reduction in the risk <strong>of</strong><br />

disease progression or death in arm A as compared with the control (arm C)<br />

at high IGF-1 level (p � 0.16). When IGF-1 was fitted as a continuous<br />

variable, this reduction was 96% (p � 0.08). There was no correlation<br />

between tissue and serum IGF-1. Conclusions: Tissue expression <strong>of</strong> IGF-1<br />

level may represent a promising predictive biomarker for IGF1R-directed<br />

therapy in APC.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


252s Gastrointestinal (Noncolorectal) Cancer<br />

4055 General Poster Session (Board #42F), Mon, 8:00 AM-12:00 PM<br />

A randomized phase III multi-institutional study <strong>of</strong> TNFerade biologic with<br />

5-FU and radiotherapy for locally advanced pancreatic cancer: Final results<br />

Presenting Author: Aaron Tyler Wild, Department <strong>of</strong> Radiation Oncology<br />

and Molecular Radiation Sciences, Johns Hopkins University School <strong>of</strong><br />

Medicine, Baltimore, MD<br />

Background: TNFerade biologic (TNF) is a novel means <strong>of</strong> selective delivery<br />

<strong>of</strong> TNF-� to tumor cells by gene transfer through intratumoral (IT) injection.<br />

TNF is a replication deficient adenovirus vector containing TNF-� cDNA<br />

ligated downstream from a radiation-inducible Egr-1 promoter, allowing<br />

spatiotemporal constraint <strong>of</strong> TNF-� production to the radiation field. Herein<br />

we report the final results <strong>of</strong> a multi-center, randomized, open-label,<br />

controlled phase III trial <strong>of</strong> TNF with chemoradiotherapy for locally<br />

advanced pancreatic ductal adenocarcinoma (PDA). Methods: Pts with<br />

locally advanced PDA were randomized 1:2 to standard <strong>of</strong> care (SOC;<br />

5-FU/RT followed by GEM) versus TNF � SOC. RT dose was 50.4 Gy in 28<br />

fractions. Concurrent 5-FU (200 mg/m2 /day IV) started on day 1 <strong>of</strong> RT each<br />

wk. TNF was injected IT ~4 hrs prior to RT weekly by percutaneous (PTA) or<br />

endoscopic (EUS) approach. 4 wks after RT, GEM (1000 mg/m2 IV) was<br />

given until progression or toxicity. Results: Of 277 pts, 187 were randomized<br />

to TNF and 90 to SOC. Demographic/baseline characteristics were<br />

similar between arms (all NS), as was GEM received (67 vs. 68%; 7.0 vs.<br />

7.6 total wks). Median f/up was 9.1 mos (range, 0.1-50.5). Median OS for<br />

TNF by ITT analysis was 10.1 mos (95% CI, 9.1-11.7) vs. 10.0 mos (95%<br />

CI, 7.6-11.2) for SOC (p�0.6). TNF delivery method did not affect OS (9.4<br />

mos for PTA vs. 11.5 for EUS; p�0.7). Baseline CA19-9 � 1000 was<br />

found to impart independent risk to TNF pts (HR�1.7; p�0.02). Subgroup<br />

analysis (SGA) <strong>of</strong> 86 pts with T1-T3 disease showed an OS benefit for TNF<br />

compared to SOC (10.9 vs. 9.0 mos, respectively; p�0.04). TNF resulted<br />

in more grade 1-2 fever/chills (p�0.001) as well as grade 3 (p�0.001) and<br />

4(p�0.05) toxicities (commonly lymphopenia, hypo/hyperkalemia, abd/<br />

chest pain) than SOC. Use <strong>of</strong> PTA vs. EUS did not affect grade 3/4 toxicity<br />

rates. Conclusions: TNF � SOC did not prolong OS for locally advanced<br />

PDA. SGA reveals a possible OS benefit for early stage (T1-T3) tumors and<br />

CA19-9 � 1000. PTA and EUS injection achieved similar rates <strong>of</strong> efficacy<br />

and toxicity. Grade 1-2 toxicity typical <strong>of</strong> systemic exposure to TNF-�<br />

(pyrexia/hypotension/chills) was common, but grade 3-4 was minimal.<br />

4057 General Poster Session (Board #42H), Mon, 8:00 AM-12:00 PM<br />

Activity <strong>of</strong> front-line FOLFIRINOX (FFX) in stage III/IV pancreatic adenocarcinoma<br />

(PC) at Memorial Sloan-Kettering Cancer Center (MSKCC). Presenting<br />

Author: Maeve A. Lowery, Memorial Sloan-Kettering Cancer Center,<br />

New York, NY<br />

Background: The PRODIGE/ACCORD trial recently established FFX as a<br />

treatment option for good performance status (PS) pts with stage IV PC<br />

(Conroy et al, NEJM 2011). We evaluated the activity and toxicity<br />

associated with FFX therapy in pts with advanced PC treated at MSKCC<br />

outside <strong>of</strong> a clinical trial. Methods: 80patients (pts) treated withFFX as<br />

1st-line therapyat MSKCC between 07/1/10 and 12/30/11, were identified<br />

from an institutional database (prior IRB approval). Records were reviewed<br />

for demographic, treatment, toxcity, and response data. Results: 61 and 19<br />

pts received FFX for stage IV and III PAC respectively. Demographics and<br />

outcomes are summarized in the table. Median starting dose <strong>of</strong> FFX was<br />

80% <strong>of</strong> that used in the PRODIGE/ACCORD trial. Median overall survival<br />

(OS) was 12.5 months (mo) (95% CI 9.5–15.5) in pts treated with 1st line<br />

FFX for stage IV PAC and 13.7 mo (95% CI 11.3–15.8) in pts with stage III<br />

PAC. 68% <strong>of</strong> pts with stage IV PAC who discontinued FFX for disease<br />

progression (PD) or toxicity received 2nd-line gemcitabine-based therapy.<br />

Conclusions: We observed activity and acceptable toxicity in carefully<br />

selected patients treated with FFX at 80% dose intensity and routine use <strong>of</strong><br />

growth factor support. Treatment with FFX resulted in median OS <strong>of</strong> � 1<br />

year in pts with stage IV PAC and is an active front-line regimen.<br />

1st line: Stage IV<br />

N�61<br />

1st line: Stage III<br />

N�19<br />

Age (yrs)<br />

Sex<br />

Male<br />

64 (range 46-80)<br />

-<br />

33(54%)<br />

58 (range 37-69)<br />

-<br />

11(58%)<br />

Female<br />

Primary tumor<br />

28(46%)<br />

-<br />

8(42%)<br />

-<br />

Head<br />

23(38%)<br />

14(74%)<br />

Body<br />

21(34%)<br />

2(10%)<br />

Tail<br />

Prophylactic growth factor<br />

Median number <strong>of</strong> cycles<br />

Dose reduction/discontinutation for toxicity<br />

17(28%)<br />

51(84%)<br />

7<br />

32(52%)<br />

3(16%)<br />

15(79%)<br />

6<br />

9(47%)<br />

GI<br />

11(18%)<br />

4(21%)<br />

Myelosupression<br />

4(7%)<br />

1(5%)<br />

Neuropathy<br />

9(15%)<br />

1(5%)<br />

Drug reaction<br />

4(7%)<br />

1(5%)<br />

Other<br />

Subsequent surgery/RT<br />

Best response*<br />

4(7%)<br />

N/A<br />

-<br />

2(10%)<br />

1(5%)/10(53%)<br />

-<br />

PR<br />

21(40%)<br />

4(21%)<br />

SD<br />

23(45%)<br />

13(68%)<br />

PD<br />

8(15%)<br />

2(11%)<br />

* Evaluable pts N�52 (1 pt not yet staged, 3 pts� 2 cycles FFX due to toxicity/5 pts due to<br />

non-treatment-related death).<br />

4056 General Poster Session (Board #42G), Mon, 8:00 AM-12:00 PM<br />

M402, a heparan sulfate mimetic and novel candidate for the treatment <strong>of</strong><br />

pancreatic cancer. Presenting Author: Birgit C. Schultes, Momenta Pharmaceuticals,<br />

Cambridge, MA<br />

Background: Recent advances in pancreatic cancer research implicate the<br />

involvement <strong>of</strong> several heparin-binding growth factors (such as HGF,<br />

HB-EGF, PDGF, hedgehogs, and TGFs) that control tumor-stroma interactions.<br />

We have rationally designed a heparan sulfate mimetic, M402, which<br />

has been previously shown to affect tumor progression and metastasis<br />

through disruption <strong>of</strong> multiple pathways. We hypothesized that M402<br />

could modulate tumor-stroma interactions and enhance the efficacy <strong>of</strong><br />

gemcitabine, and evaluated its efficacy in two preclinical models. Methods:<br />

A genetically engineered mouse model (GEMM; KrasLSLG12D p53LSLR172H )<br />

featuring spontaneous pancreatic tumor formation and metastasis assessed<br />

M402’s effect on tumorigenesis and metastasis. The orthotopic<br />

Capan-2 model in nude mice evaluated the effect <strong>of</strong> M402 on desmoplasia,<br />

a fibrotic response that hinders effective delivery <strong>of</strong> chemotherapeutics, via<br />

inhibition <strong>of</strong> sonic hedgehog (SHH) signaling in fibroblasts and stellate<br />

cells. In both models, M402 was studied as monotherapy and with<br />

gemcitabine. Results: In the GEMM, M402 significantly prolonged survival<br />

in combination with gemcitabine while each monotherapy showed modest<br />

efficacy. M402, alone and in combination, also reduced metastases and<br />

local invasion and inhibited epithelial-to-mesenchymal transition. In the<br />

Capan-2 model, gemcitabine was increasingly less effective as desmoplasia<br />

progressed over time. The addition <strong>of</strong> M402 to gemcitabine increased<br />

its efficacy with respect to primary tumor burden. Metastasis, invasion, and<br />

surrounding fibrotic lesions appeared particularly impacted by the combination<br />

treatment. M402 was also effective as monotherapy with dosedependency,<br />

which correlated with reduced SHH signaling. Conclusions:<br />

M402 can modulate tumor-stroma interactions involved in the metastatic<br />

and desmoplastic pathways in two pancreatic cancer models supporting<br />

the translation <strong>of</strong> these findings into a clinical study. A first-in-human study<br />

is planned that will evaluate the safety, pharmacokinetics, efficacy, and<br />

biomarker pr<strong>of</strong>iles <strong>of</strong> escalating M402 doses in combination with gemcitabine<br />

in patients with metastatic pancreatic cancer.<br />

4058 General Poster Session (Board #43A), Mon, 8:00 AM-12:00 PM<br />

Benefits <strong>of</strong> platinum-based chemotherapy (Pt-chemo) in pancreatic adenocarcinoma<br />

(PC) associated with BRCA mutations: A translational case<br />

series. Presenting Author: Olusola O. Faluyi, Princess Margaret Hospital,<br />

Toronto, ON, Canada<br />

Background: The prognosis <strong>of</strong> PC is poor with limited response to standard<br />

chemotherapy. Prior randomized studies <strong>of</strong> cisplatin and gemcitabine in PC<br />

demonstrated no additional benefit over gemcitabine alone. Preclinical<br />

data and case reports suggest that BRCA mutant PC may have increased<br />

sensitivity to Pt-chemo. Our case series characterizes the benefits <strong>of</strong><br />

Pt-chemo in germline BRCA mutant PC. Methods: Patients with PC and<br />

germline BRCA mutations were identified using the Ontario Pancreatic<br />

Cancer Study and Pharmacy databases. Review <strong>of</strong> clinical records provided<br />

demographic, treatment and survival data. Radiology review assessed<br />

responses to chemotherapy. RNA was extracted from tumor samples for<br />

gene expression studies using a panel <strong>of</strong> DNA repair genes and Nanostring<br />

technology. BRCA loss <strong>of</strong> heterozygosity (LOH) was also investigated.<br />

Results: We identified 14 PC patients with BRCA mutations (8 BRCA2, 6<br />

BRCA1). 11 <strong>of</strong> these had metastatic disease <strong>of</strong> which 5 received Pt-chemo.<br />

Of the 5 treated with Pt-chemo, there were 3 partial responses (PR) and 2<br />

complete responses (CR) using Recist criteria. For the remaining 6 not<br />

treated with Pt-chemo, there was 1 PR with gemcitabine. Additionally, two<br />

patients with locally advanced disease at diagnosis became resectable<br />

following Pt-chemo. Overall survival was superior for patients receiving<br />

Pt-chemo (33.0�25.6 vs 7.3�4.5 months; p�0.04). Gene expression<br />

and LOH results will be presented. Conclusions: Responses and survival<br />

associated with Pt-chemo observed in our case series <strong>of</strong> BRCA mutant PC<br />

adds to existing data supporting the use <strong>of</strong> Pt-chemo in this subgroup.<br />

Despite the low (2 to 5%) prevalence <strong>of</strong> BRCA mutations in PC, the benefits<br />

gained from personalizing treatment using Pt-chemo supports BRCA<br />

testing in selected patients.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


4059 General Poster Session (Board #43B), Mon, 8:00 AM-12:00 PM<br />

Inequal benefits from regionalization <strong>of</strong> cancer care: The pancreatic cancer<br />

surgery paradigm. Presenting Author: Theodore P. McDade, Surgical<br />

Outcomes Analysis & Research, University <strong>of</strong> Massachusetts Medical<br />

School, Worcester, MA<br />

Background: Regionalization has been proposed for high-level care, including<br />

multidisciplinary cancer treatment and complex procedures. Pancreatic<br />

resections can serve as a marker for both. Using Massachusetts<br />

Division <strong>of</strong> Health Care Finance and Policy (DHCFP) data, we investigated<br />

regionalization <strong>of</strong> surgery for pancreatic cancer (PCa), its potential effect<br />

on perioperative outcomes, and disparities in access to high-volume PCa<br />

surgery centers. Methods: Using MA DHCFP Hospital Inpatient Discharge<br />

Data, 2005-2009, 10,524 discharges for PCa were identified, <strong>of</strong> which<br />

746 were associated with pancreatic resection. Discharges with missing or<br />

out-<strong>of</strong>-state residence were excluded (n�704). Using geodetic methods<br />

and ZIP codes, center-to-center distances were calculated between patient<br />

(pt) and treating hospital. Median ZIP income was estimated from 2009<br />

census data. High volume hospitals (4 <strong>of</strong> 25 performing pancreatic<br />

resections in MA) were defined using Leapfrog Criteria (� 11 per year (87th percentile for MA). Chi-square and logistic regression analyses were<br />

performed using SAS s<strong>of</strong>tware. Results: Median age was 65. Pts were<br />

predominantly White (87.2%), with median ZIP income <strong>of</strong> $54,677. Pts<br />

travelled in-state up to 112 miles (median 15.4), with the majority<br />

resected at high volume hospitals (76%). Median length <strong>of</strong> stay (LOS) was<br />

8.0 days, with LOS�1 week associated with low volume hospitals<br />

(p�0.0002). Of 14 in-hospital deaths, 7 were at low volume hospitals<br />

(4.14% <strong>of</strong> 169 pts) compared to 7 at high volume hospitals (1.31% <strong>of</strong> 535<br />

pts) (p�0.0214). Predictors <strong>of</strong> shorter travel distance were: Black race (OR<br />

4.45 (95% CI 1.66-11.93)), operation at low volume hospital (OR 2.62<br />

(95% CI 1.81-3.77), and increased age (per year) (OR 1.02 (95% CI<br />

1.00-1.03), but not sex or median income. Conclusions: Using MA<br />

statewide discharge data, regionalization <strong>of</strong> pancreatic cancer surgery to<br />

high-volume, better-outcome centers is seen to be occurring. However, it is<br />

not uniform, and disparities exist between groups <strong>of</strong> cancer pts that do and<br />

do not travel for their care. In the current era <strong>of</strong> scrutiny on cost, quality,<br />

and access to cancer care, further study into predictors <strong>of</strong> pts receiving<br />

optimal care is warranted.<br />

4061 General Poster Session (Board #43D), Mon, 8:00 AM-12:00 PM<br />

Phase II trial <strong>of</strong> bevacizumab, irinotecan, cisplatin, and radiation as<br />

preoperative therapy in esophageal adenocarcinoma. Presenting Author:<br />

David Ilson, Memorial Sloan-Kettering Cancer Center, New York, NY<br />

Background: Preop chemo and radiotherapy (RT) with weekly irinotecan (I),<br />

cisplatin (C) and 5040 cGy is tolerable and active [Cancer, in press].<br />

Bevacizumab (B) � weekly I/C in advanced esophagogastric cancer<br />

increased response rate and PFS without an increase in toxicity [JCO 24:<br />

5201; 2006]. In a phase II trial in esophageal adenocarcinoma (EA) we<br />

combed B � I/C with RT as preop therapy. A toxicity analysis after 10<br />

patients (pts) undergoing surgery indicated no unexpected toxicity with B<br />

[JCO 27: Abstract 4573; 2009], and we completed a planned accrual <strong>of</strong> 33<br />

pts. Methods: Pts had resectable distal esophageal or Siewert’s I or II EA,<br />

T2-3 or N1 staged by EUS, PET, and CT scan. Induction chemo: I: 50-65<br />

mg/m2 � C: 30 mg/m2 weeks 1,2,4,5, B: 7.5 mg/kg weeks 1 and 4;<br />

Chemort: 180 cGy daily fractions to 5040 cGy, I/C weeks 7,8,10,11� B<br />

weeks 7,10. Esophagectomy was planned 6-8 weeks after RT. Postop: B:<br />

7.5 mg/kg every 3 weeks for 8 cycles. Results: 33 pts were enrolled. 26<br />

male: 7 female; distal esophagus 11 (33%), GEJ 22 (67%); 21 T3N1<br />

(64%); 10 T3N0 (30%); 2 T2N0-1 (6%). 25/33 pts went to surgery (76%),<br />

24 had R0 resection (73%). Pathologic CR 5 pts (15%). 8 pts did not going<br />

to surgery: 2 for adverse events (9%, 1 CVA due to patent foramen ovale, 1<br />

esophageal perforation due to endoscopic biopsy), 6 for progressive disease<br />

(18%). 21/24 pts (88%) received adjuvant B, 20 (95%) completed all<br />

cycles. Grade 3/4 toxicity in 30 evaluable pts during chemort: 24%<br />

neutropenia, 3% neutropenic fever, 23% esophagitis, 13% dehydration,<br />

13% thrombocytopenia, 7% pulmonary embolism, 3% nausea/vomiting.<br />

Surgical complications: 3 anastomotic leaks (12%), 4 respiratory failure<br />

(16%), 1 pulmonary embolism (4%), 2 post op deaths (8%). At a median<br />

follow up <strong>of</strong> 20 months, PFS was 14 months and OS was 30 months.<br />

Conclusions: The addition <strong>of</strong> B to preop chemoRT in EA was tolerable<br />

without increase in treatment toxicity or surgical complications. There was<br />

no suggestion <strong>of</strong> improved pathologic CR, PFS, or OS with the addition <strong>of</strong> B<br />

to I/C/RT. Evaluation <strong>of</strong> B in phase III trials <strong>of</strong> chemort does not appear<br />

warranted. Supported by a grant from Genentech.<br />

Gastrointestinal (Noncolorectal) Cancer<br />

253s<br />

4060 General Poster Session (Board #43C), Mon, 8:00 AM-12:00 PM<br />

Choice <strong>of</strong> chemotherapy in the treatment <strong>of</strong> metastatic squamous cell<br />

carcinoma <strong>of</strong> the anal canal. Presenting Author: Cathy Eng, University <strong>of</strong><br />

Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: Metastatic squamous cell carcinoma (SCCA) <strong>of</strong> the anal canal<br />

is an uncommon malignancy with no standard approach. The reported<br />

median overall survival (OS) is 9-12 months (M) following 5-FU � cisplatin<br />

(FC)-based therapy. The aim <strong>of</strong> this study is to evaluate first-line chemotherapy<br />

approaches in this patient (pt) population. Methods: A retrospective<br />

analysis was conducted <strong>of</strong> 428 pts with metastatic SCCA <strong>of</strong> the anal canal<br />

identified from the MDACC tumor registry between 1/1/2000 - 5/31/2011.<br />

Electronic medical records were reviewed for histology, date <strong>of</strong> diagnosis<br />

and/or recurrence, site <strong>of</strong> metastasis, type <strong>of</strong> therapy provided, response<br />

rate (RR), progression-free survival (PFS), OS, and lines <strong>of</strong> salvage therapy.<br />

All eligible pts were required to be treatment-naïve for metastatic disease<br />

and have radiographic imaging at MDACC. Waiver <strong>of</strong> informed consent was<br />

obtained. Results: 99 pts fulfilled all criteria; 10 were lost to follow-up; 12<br />

did not initiate chemotherapy. 77 pts were evaluable; M: F � 20:57;<br />

median age � 55 years (range: 37 - 82); HIV(�) � 5% (4/77); prior<br />

chemoXRT with curative intent: 70% (54/77), complete response (CR):<br />

87% (47/54), median time to development <strong>of</strong> metastatic disease �17M.<br />

29% (22/77) presented with metastatic disease. Sites <strong>of</strong> disease included<br />

distant lymph nodes (41%); liver (45 %); lung (25%); bone (15%); and<br />

brain (8%). The median follow up was 37M. 73% (56/77) <strong>of</strong> patients were<br />

treated with platinum-based therapy; 51% (n�39) received FC and 22%<br />

(n� 17) received carboplatin � paclitaxel (CP). The median PFS was 6M;<br />

FC trended better than CP for PFS (7M vs. 5M, p�0.067). The overall<br />

median OS � 29M. 40% (31/77) <strong>of</strong> pts received neoadjuvant first-line<br />

therapy followed by metastasectomy (68%), XRT (26%), or both (6%);<br />

resulting in a median OS � 35M. Conclusions: Metastatic SCCA <strong>of</strong> the anal<br />

canal is a malignancy in which 5-FU�cisplatin is a commonly used<br />

regimen. Our analysis suggests FC results in improved PFS over CP but is<br />

underpowered supporting further analysis. The short median PFS with<br />

front-line chemotherapy, and yet longer OS reflects the challenges in<br />

treating this patient population and the importance <strong>of</strong> multidisciplinary<br />

management in select cases.<br />

4062 General Poster Session (Board #43E), Mon, 8:00 AM-12:00 PM<br />

A phase II study <strong>of</strong> neoadjuvant therapy with cisplatin, docetaxel, panitumumab<br />

plus radiation therapy followed by surgery in patients with locally<br />

advanced adenocarcinoma <strong>of</strong> the distal esophagus (ACOSOG Z4051).<br />

Presenting Author: Carolyn E. Reed, Medical University <strong>of</strong> South Carolina,<br />

Charleston, SC<br />

Background: Multiple clinical trials have incorporated preoperative chemotherapy<br />

and radiation (RT) in an attempt to improve local tumor control,<br />

distant disease failure and overall survival rates for locally advanced but<br />

resectable adenocarcinoma <strong>of</strong> the distal esophagus and gastroesophageal<br />

junction (GEJ). This multicenter, cooperative group study combined active<br />

chemotherapy agents cisplatin (C), docetaxel (D) and the targeted EGFR<br />

agent panitumumab (P) in the induction phase followed by concurrent<br />

chemotherapy (CDP) and radiation. Pathologic complete response (pCR), a<br />

surrogate for improved survival, was the primary endpoint. Methods: From<br />

01/15/09 to 07/22/11, 70 patients (pts) with Siewert I or II adenocarcinomas<br />

and clinical stages T3N0M0, T2-3N1M0 or T2-3N0-1M1a (celiac LN<br />

� 2 cm) were accrued. Patients received cisplatin (40 mg/m2 ), docetaxel<br />

(40 mg/m2 ) and panitumumab (6 mg/kg) on weeks 1, 3, 5, 7, 9 with RT<br />

(5040 cGy, 180 cGy/day x 28d) beginning week 5. The decision rule had a<br />

90% power with a 0.10 significance level to detect a pCR rate <strong>of</strong> at least<br />

35%. Secondary objectives included near-pathologic complete response<br />

(near-pCR), toxicity, and overall and disease-free survival rates. Results:<br />

Five pts were ineligible. Of the remaining 65 pts (59 M, 6 F; median age<br />

61), 12 pts did not undergo surgery (5 progressed, 4 refused, 3 other). Of<br />

the 58 evaluable pts, the pCR rate was 32.8% (90% CI: 22.6% -42.9%)<br />

and near-pCR 22.4% (90% CI: 13.4%-31.4%). Total doses <strong>of</strong> C, D, and P<br />

were achieved in 76%, 80%, and 73%, respectively (n � 70). 66 pts<br />

(94%) received the total RT dose. Sixteen pts (23%) had a grade 4�<br />

non-heme adverse event possibly related to treatment. Venous thrombosis<br />

(5 pts) was most common. Conclusions: The CDP regimen in the neoadjuvant<br />

setting in patients with esophageal adenocarcinomas is active (pCR �<br />

near-pCR � 55.2%) and feasible. The toxicity though tolerable is substantial.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


254s Gastrointestinal (Noncolorectal) Cancer<br />

4063 General Poster Session (Board #43F), Mon, 8:00 AM-12:00 PM<br />

Bisphosphonates and esophageal cancer: A RADAR report. Presenting<br />

Author: Beatrice J. Edwards, Department <strong>of</strong> Medicine, Geriatrics Division,<br />

Northwestern University Feinberg School <strong>of</strong> Medicine, Chicago, IL<br />

Background: In 2009, the US Food and Drug Administration (FDA) reported<br />

on 23 patients who had developed distal esophageal cancer, with alendronate<br />

(ALN) within 2 years <strong>of</strong> initiation <strong>of</strong> therapy. Similarly, 31 cases <strong>of</strong><br />

esophageal cancer were reported from Europe and Japan. Esophagitis has<br />

been associated with oral BPs. Erosive esophagitis and persistent mucosal<br />

abnormalities have been noted with crystalline material (similar to ground<br />

ALN). Our objective was to assess the FDA Adverse event reporting system<br />

(FDA AERS) for a safety signal. Methods: The FDA Adverse Event Reporting<br />

System (AERS) database was searched using terms related to esophageal<br />

cancer combined with all drug names for bisphosphonates (search period:<br />

1996-2010). Disproportionality ratios were calculated: Proportional reporting<br />

ratio (PRR) and Empiric Bayes Geometric Mean (EBGM) determining<br />

that the esophageal cancer cases were more common with bisphosphonates<br />

than with other drugs in the database. Results: We identified 128<br />

cases <strong>of</strong> bisphosphonate-associated esophageal cancer; 114 (89%) female<br />

and 14 male (11%), mean age 72 � 12 yrs; the drugs included alendronate<br />

(n�96, 75%), risedronate sodium (n�14, 11%), ibandronic acid (n�10,<br />

7.8%), zoledronic acid (n�7, 5.4%) and pamidronate (n�1, 1%). Barrett’s<br />

esophagus was listed in 3 cases <strong>of</strong> esophageal carcinoma. A<br />

significant safety signal was found only for alendronate with a PRR� 6.4<br />

(95% C.I. 5.29, 7.730; p�0.001), EBGM � 6.3 (95% C.I. 5.1, 7.6<br />

p�0.001). Conclusions: Our analysis <strong>of</strong> FDA AERS identifies a larger<br />

number <strong>of</strong> cases <strong>of</strong> esophageal cancer than previously described, and a<br />

significant safety signal with alendronate use. Avoidance <strong>of</strong> oral bisphosphonates<br />

in patients with Barrett’s esophagus, and persistent mucosal abnormalities<br />

is recommended. Increased awareness and vigilance is needed for<br />

patients receiving oral bisphosphonate therapy.<br />

4065 General Poster Session (Board #43H), Mon, 8:00 AM-12:00 PM<br />

Trastuzumab (TRA) in combination with different first-line chemotherapies<br />

for treatment <strong>of</strong> HER2-positive metastatic gastric or gastroesophageal<br />

junction cancer (MGC): Findings from the German noninterventional<br />

observational study HerMES. Presenting Author: Susanna Hegewisch-<br />

Becker, Internistische Praxisgemeinschaft Eppendorf, Hamburg, Germany<br />

Background: The international phase III study ToGA has recently shown that<br />

TRA is effective in prolonging survival in HER2-positive MGC. However, few<br />

data are available for TRA as part <strong>of</strong> routine clinical practice. Methods: This<br />

non-interventional observational study was conducted to evaluate the<br />

efficacy, safety and feasibility <strong>of</strong> TRA in previously untreated pts with<br />

HER2-positive MGC. Results: Between Apr 2010 and Jan 2012, data from<br />

110 pts were collected. All pts were evaluable for safety. Baseline pt<br />

characteristics were as follows: median age 63 yrs (range 29–88); gender<br />

(male 70%; female 29%); ECOG PS (0: 25%; 1: 50%; 2: 15%; 3: 5%);<br />

distant mets (91%); liver mets (54%), lymph node mets (35%); peritoneal<br />

carcinomatosis (23%). The median duration <strong>of</strong> TRA treatment was 4.4<br />

months (0–17.1). According to the schedule <strong>of</strong> chemotherapy TRA was<br />

administered every 2–3 wks in a median dose <strong>of</strong> 4–6 mg/kg BW. Only 28%<br />

<strong>of</strong> pts received TRA according to the label in combination with cisplatin and<br />

5-FU or capecitabine. The remainder received: cisplatin, 5-FU and<br />

leucovorin (17%); 5-FU, leucovorin, oxaliplatin and docetaxel (8%); 5-FU,<br />

leucovorin and oxaliplatin (7%); capecitabine (6%); other combinations<br />

(25%); TRA monotherapy (7%). Although most pts didn’t receive cisplatinbased<br />

therapy, preliminary median progression-free survival was 6.8<br />

months, thus comparable to the ToGA data. Most common adverse events<br />

(AEs, all grades) were diarrhoea (7%), vomiting (5%) and nausea (5%).<br />

Most common grade 3/4 AEs were vomiting (3%), nausea (2%) and fatigue<br />

(2%). Health-related quality <strong>of</strong> life as assessed by EORTC QLQ-C30 and<br />

QLQ-STO22 remained stable during observation time. An updated analysis<br />

<strong>of</strong> approx. 200 pts will be presented at the meeting. Conclusions: TRA<br />

combined with diverse chemotherapies is safe and effective in the routine<br />

treatment <strong>of</strong> MGC. Cisplatin-free less toxic regimens are feasible and<br />

equally effective. The results are in line with those from the ToGA trial and<br />

suggest that treatment with TRA should be regarded as standard <strong>of</strong> care for<br />

pts with HER2-positive MGC.<br />

4064 General Poster Session (Board #43G), Mon, 8:00 AM-12:00 PM<br />

Combined analysis <strong>of</strong> randomized controlled trial (RCT) and patientpreference<br />

trial (PPT) evaluating second-line chemotherapy (SLC) in<br />

advanced gastric cancer (AGC). Presenting Author: Soonil Lee, Dankook<br />

University Hospital, Cheonan, South Korea<br />

Background: While prolonged overall survival (OS) from SLC compared to<br />

best supportive care (BSC) has recently been demonstrated for AGC (Park<br />

et al, ASCO 2011), the prognosis <strong>of</strong> pretreated AGC remains poor. We<br />

assessed whether patient preference, willing to participate onto RCT, or<br />

other parameters contributed to this OS improvement. Methods: In the<br />

phase III trial, pretreated AGC patients (n�372) were first <strong>of</strong>fered RCT. If a<br />

patient agreed to participate, randomly assigned 2:1 to SLC or BSC<br />

(n�202). If refused RCT, but agreed to receive treatment <strong>of</strong> their<br />

preference, they were <strong>of</strong>fered SLC or BSC (PPT; n�170). OS <strong>of</strong> RCT<br />

participants was compared to that <strong>of</strong> whole patient population according to<br />

prognostic subgroups. In addition, analyses <strong>of</strong> OS unadjusted for multiple<br />

comparisons were conducted across predefined subgroups. Results: Median<br />

OS was 6.3 months among 254 patients treated with SLC and 3.3 months<br />

among 118 patients treated with BSC (HR, 0.507; 95% CI, 0.405 to<br />

0.637; one-sided p�0.001). Compared to the RCT patients, younger age<br />

group, those with an ECOG performance status (PS) <strong>of</strong> 0, and with one prior<br />

chemotherapy were under-represented in the PPT patients. OS was<br />

comparable between RCT (5.0 months) and PPT (4.4 months) patients<br />

even after controlling for major prognostic factors (log-rank p�0.322). The<br />

OS benefit for SLC was preserved when analyzed according to baseline<br />

parameters. Parameters that were associated significantly with a prolonged<br />

OS included a PS <strong>of</strong> 0, the chemotherapy-free interval �3 months, and one<br />

prior chemotherapy. When the analysis was adjusted for these three<br />

parameters, patients who received SLC still had improved OS (HR, 0.554;<br />

95% CI, 0.441 to 0.696; p�0.001). Conclusions: The 54% participation<br />

rate obtained in the current study represents the best achievable rate for<br />

this kind <strong>of</strong> phase III RCT involving BSC only arm. Even if we consider<br />

differences in the baseline characteristics between RCT and PPT patients,<br />

it is concluded that the RCT results can be generalized for the patient<br />

population and provided additional evidence supporting the use <strong>of</strong> SLC in<br />

patients with pretreated AGC.<br />

4066 General Poster Session (Board #44A), Mon, 8:00 AM-12:00 PM<br />

Does in-house availability <strong>of</strong> multidisciplinary teams increase survival in<br />

upper GI cancer? Presenting Author: Christian Kersten, Center for Cancer<br />

Treatment, Sørlandet Hospital Trust, Kristiansand, Norway<br />

Background: Treatment <strong>of</strong> esophageal, gastric and pancreatic cancers (UGI)<br />

is recommended to be discussed by a multidisciplinary team (MDT),<br />

despite lack <strong>of</strong> evidence that this approach leads to increased survival. In<br />

2001, a cancer centre with irradiation and chemotherapy facilities was<br />

established in the Norwegian county <strong>of</strong> West Agder (WA), providing the<br />

potential for local in-house MDT meetings. Our primary objective was to<br />

evaluate the effect <strong>of</strong> the establishment <strong>of</strong> in-house MDT availability<br />

(iMDTa) on survival in a cohort <strong>of</strong> UGI patients in WA. We compared<br />

survival figures for WA with those <strong>of</strong> other Norwegian counties with<br />

complete, less complete and without iMDTa. Methods: We defined “iMDTa”<br />

as a single administrative institution with all departments on one campus,<br />

serving the population <strong>of</strong> one county. We compared cause-specific survival<br />

rates for 2000-04 and 2005-08 for UGI patients living in counties with<br />

(MDT-Yes), without (MDT-No) and with a mix (MDT-mix) <strong>of</strong> iMDTa, with the<br />

county <strong>of</strong> WA which had a change in iMDTa (MDT-Change) during the study<br />

period. Crude survival was modelled with Kaplan-Meier method and Cox<br />

regression analysis was used to adjust for age and region (sex and stage<br />

distributions were similar in all counties). Results: We analyzed 12530 UGI<br />

patients living in five Norwegian regions. Median age was 74 (17-98) yrs<br />

and median follow-up was 5 (0-138) months. The regions with the highest<br />

level <strong>of</strong> iMDTa achieved the largest increases in survival, compared to the<br />

counties with limited or no iMDTa. Median overall survival for all UGI<br />

patients in WA/MDT-Change increased from 129 to 300 days from 2000-8,<br />

p�0.001. Compared to the county with MDT-Mix, the county with<br />

MDT-Change reached a statistically significant reduction in the risk <strong>of</strong><br />

death (HR) for both esophageal (1.12- 0.60) and stomach cancers<br />

(0.87-0.63), but not for pancreatic cancers (1.04-1.01). Conclusions: In<br />

parallel with an increasing use <strong>of</strong> in-house MDT, we found a striking and<br />

more than two-fold increase in survival in the Norwegian county <strong>of</strong><br />

WA/MDT-Change. This survival gain is partly explained by increased use <strong>of</strong><br />

chemotherapy. During the same time period, no increase in survival was<br />

found in the MDT-No or MDT-Mix counties.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


4067 General Poster Session (Board #44B), Mon, 8:00 AM-12:00 PM<br />

Correlation between overall survival and other endpoints in clinical trials <strong>of</strong><br />

second-line chemotherapy for patients with advanced gastric cancer.<br />

Presenting Author: Kohei Shitara, Aichi Cancer Center Hospital, Nagoya,<br />

Japan<br />

Background: The correlation between progression-free survival (PFS) or<br />

time to progression (TTP) and overall survival (OS) has been evaluated in<br />

patients with advanced gastric cancer (AGC) who received first-line<br />

chemotherapy (Shitara, K et al. Invest New Drug 2011; Shitara K and<br />

Burzykowski T, et al, ASCO 2011). However, no corresponding analysis had<br />

been done in patients who underwent second-line chemotherapy for AGC.<br />

Methods: We evaluated the potential <strong>of</strong> PFS, TTP, response rate (RR), or<br />

disease control rate (DCR) to act as surrogates for OS in phase II and III<br />

trials <strong>of</strong> second-line chemotherapy for AGC by comprehensive literaturebased<br />

analysis. Correlations between each endpoint and OS were evaluated<br />

by Spearman rank correlation coefficient (�). Subgroup analyses by trial<br />

region or type <strong>of</strong> failure to previous chemotherapy were also conducted.<br />

Results: Fifty-six trials, including four randomized studies, were selected<br />

for analysis and covered a total <strong>of</strong> 61 treatment arms and 3,038 patients;<br />

34 studies were conducted in Asia, 20 studies in Non-Asian countries, and<br />

two studies in both regions. Median PFS were similar in Asian and<br />

Non-Asian trials (3.0 vs. 3.3 months). In contrast, median OS tended to be<br />

longer in Asian vs. Non-Asian trials (8.0 vs. 6.0 months; p�0.01). Median<br />

PFS/TTP and OS showed a moderate correlation with � <strong>of</strong> 0.51 (95% CI,<br />

0.31-0.73). Correlation tended to be higher in PFS (� � 0.62) than TTP (�<br />

� 0.29) and higher in non-Asian trials (� � 0.73) than Asian trials (� �<br />

0.32). Correlation between PFS/TTP and OS among the trials in which<br />

eligibility required failure to previous fluorouracil and cisplatin also showed<br />

low correlation (� � 0.48). The RR and DCR also did not show high<br />

correlation with OS (� � 0.30 for RR; 95% CI 0.04-0.56; � � 0.53 for<br />

DCR; 95% CI 0.31-0.75). The hazard ratio (HR) <strong>of</strong> PFS and OS in each<br />

arms <strong>of</strong> the four randomized studies showed a low correlation with � <strong>of</strong><br />

0.10. Conclusions: Our results indicate that PFS/TTP, RR, and DCR did not<br />

correlate sufficiently with OS to be used as surrogate endpoints in patients<br />

with AGC who underwent second-line chemotherapy. Further research is<br />

needed based on individual patient data from ongoing randomized trials.<br />

4069 General Poster Session (Board #44D), Mon, 8:00 AM-12:00 PM<br />

Customization <strong>of</strong> treatment for patients with esophageal adenocarcinoma<br />

(EAC) who achieve clinical complete response (cCR) after chemoradiation<br />

using initial standardized uptake value (iSUV) <strong>of</strong> 18f-fluorodeoxyglucose PET. Presenting Author: Akihiro Suzuki, University <strong>of</strong> Texas M. D. Anderson<br />

Cancer Center, Houston, TX<br />

Background: Patients with localized esophageal adenocarcinoma (EAC)<br />

receive preoperative chemoradiation followed by surgery (trimodality [TM]<br />

therapy) or definitive chemoradiotherapy (bimodality [BM] therapy) based<br />

on comorbidities and tumor geography. However, we cannot individualize<br />

recommendations beyond these parameters. We hypothesized that iSUV<br />

could customize therapy. Methods: Data source was our prospective<br />

database <strong>of</strong> fully staged EAC patients (2002-2010). All patients had a cCR<br />

(post-chemoradiation negative biopsy and post-chemoradiation physiologic<br />

uptake on PET). iSUV cut-point was derived by recursive partitioning.<br />

Results: For 323 cCR patients, the median follow-up was 40.8 months. 206<br />

(63.8%) patients had TM and 117 (36.2%) had BM therapy. Median OS <strong>of</strong><br />

TM patients and BM patients were 94.8 months (95 % CI; NA-NA) and<br />

36.5 months (95% CI; 30.5-42.4; p�0.001), respectively. Similar differences<br />

were observed in RFS (p�0.001). The median iSUV was 9.4 (range,<br />

0-58.0). Intriguingly, TM patients with iSUV <strong>of</strong> �6 had a better OS (94.8<br />

months; 95% CI; 39.1-150.5) and RFS (94.8 months; 95% CI; 18.2-<br />

171.4) compared to BM patients with iSUV <strong>of</strong> �6 OS (31.4 months; 95%<br />

CI; 21.0-41.9; p��0.001) and RFS (17.2 months; 95% CI; 14.5-19.8;<br />

p�0.001). However, the prognosis <strong>of</strong> TM and BM cCR patients with iSUV<br />

�6 was similar (OS, p�0.62 and RFS, p�0.46). The pathological CR<br />

(pathCR) rate in TM patients was similar irrespective <strong>of</strong> iSUV <strong>of</strong> �6<br />

(27.1%) vs. iSUV �6 (28.6%; p�0.85). Conclusions: Our unique data<br />

provide an insight into the fate <strong>of</strong> cCR EAC patients by iSUV. Patient with<br />

iSUV �6 dramatically benefit from surgery and TM therapy is encouraged.<br />

iSUV and pathCR do not correlate. iSUV can customize therapy <strong>of</strong> localized<br />

EAC patients.<br />

Gastrointestinal (Noncolorectal) Cancer<br />

255s<br />

4068 General Poster Session (Board #44C), Mon, 8:00 AM-12:00 PM<br />

Measuring transcripts <strong>of</strong> components <strong>of</strong> the BRCA1 DNA repair pathway,<br />

components <strong>of</strong> the hippo-YAP pathway, �-TrCP, HER2, AEG-1, EZH2 and<br />

other genes in advanced gastric cancer. Presenting Author: Jia Wei,<br />

Nanjing Drum Tower Hospital, Nanjing, China<br />

Background: The primary goal <strong>of</strong> this study was to classify gastric cancer in<br />

two different sub-groups. We speculated that HER2-positive gastric cancers<br />

could be regulated by �-TrCP, while tumors unaffected by �-TrCP<br />

activation could be more dependent on the Hippo-YAP signaling pathway,<br />

in which TAZ increases the expression <strong>of</strong> AXL. Gastric cancer can<br />

overexpress AEG-1 and EZH2. The relationship between EZH2 and the<br />

Hippo-YAP pathway was explored in this study. We also explored BRCA1<br />

complexes, including upstream components, such as 53BP1 and MMSET.<br />

Methods: Paraffin-embedded samples were collected from 132 advanced<br />

gastric cancer patients (p) treated with first-line FOLFOX, 58 <strong>of</strong> whom<br />

received second-line docetaxel-based treatment. Gene expression levels <strong>of</strong><br />

HER2, �-TrCP, TAZ, AXL, EZH2, AEG-1, BRCA1, and genes involved in<br />

DNA repair pathways (RAP80, PIAS1, PIAS4, MDC1, 53BP1, MMSET,<br />

and UBC9) were quantified by real-time PCR. BIM, IDO, and SPINK1 were<br />

also examined. Results: A close correlation was found for the majority <strong>of</strong> the<br />

genes, for example, BRCA1-MMSET (rho�0.28; P�0.002) and AXL-TAZ<br />

(rho�0.58; P�0.001). Median overall survival (OS) was 12.5 months (m).<br />

Among the 58 p receiving second-line docetaxel, in p with high levels <strong>of</strong><br />

BRCA1, OS was 24.9 m, while it was 19.1 m in p with intermediate levels<br />

and 9.5 m in p with low levels (P�0.009). OS was 28.6 in p with high<br />

levels <strong>of</strong> MMSET, 13.8 in p with intermediate levels and 12.3 in p with low<br />

levels (P�0.001). In the multivariate analysis, only BRCA1 was a significant<br />

marker for OS (Table). Conclusions: The study showed the predictive<br />

value <strong>of</strong> BRCA1, which could be useful for customizing chemotherapy with<br />

or without docetaxel. In addition, MMSET requires further study since it is<br />

involved in DNA repair upstream <strong>of</strong> BRCA1.<br />

HR 95% CI p<br />

�-TRCP<br />

1.22 1 Ref.<br />

ERBB2<br />

19.86 1 Ref.<br />

AEG-1<br />

1.12 1 Ref.<br />

BRCA1<br />

7.62 1 Ref.<br />

4070 General Poster Session (Board #44E), Mon, 8:00 AM-12:00 PM<br />

Investigatory study <strong>of</strong> biomarkers for gastric cancer based on a cDNA bank.<br />

Presenting Author: Takashi Oshima, Gastroenterological Center, Yokohama<br />

City University, Yokohama, Japan<br />

Background: We have constructed a cDNA bank using mRNA extracted from<br />

frozen specimens <strong>of</strong> gastric cancer tissue and adjacent normal gastric<br />

mucosa and studied biomarkers for the individualized therapy <strong>of</strong> gastric<br />

cancer and the identification <strong>of</strong> new treatment targets. We report currently<br />

available results. Methods: We studied 227 patients in whom at least 5<br />

years had elapsed since surgery for gastric cancer. The disease stage was IB<br />

in 29 patients, II in 66, III in 103, and IV in 29. Among the 139 patients<br />

who postoperatively received fluoropyrimidine anticancer agents, 82 with<br />

stage II or III disease were given adjuvant chemotherapy with S-1. A total<br />

116 genes were selected as candidate biomarkers on the basis <strong>of</strong> the<br />

results <strong>of</strong> comprehensive DNA microarray analysis, extraction from a serial<br />

analysis <strong>of</strong> gene expression (SAGE) database, and other studies. The<br />

relative expression levels <strong>of</strong> these 116 genes in gastric cancer tissue and<br />

adjacent normal mucosa were measured in each case by a quantitative<br />

polymerase chain reaction assay using the cDNA databank described<br />

above, and the relations between clinical histopathological factors and<br />

treatment outcomes were examined. Results: In patients who underwent<br />

gastrectomy, high expression levels <strong>of</strong> the secreted protein acidic and rich<br />

in cysteine (SPARC), sulfatase 1 (SULF1), and inhibin, beta A (INHBA)<br />

genes were significantly associated with poor outcomes. In patients with<br />

stage II or III disease who received adjuvant chemotherapy with S-1, high<br />

expression levels <strong>of</strong> the insulin-like growth factor receptor 1 (IGF-1R),<br />

KIAA1199, thymidylate synthase (TS), and regenerating IV (Reg IV) genes<br />

were significantly associated with poor survival. Conclusions: Investigatory<br />

studies using a cDNA bank <strong>of</strong> biomarkers for gastric cancer suggested that<br />

expression levels <strong>of</strong> the SPARC, SULFI, and INHBA genes are useful<br />

prognostic factors in patients who undergo gastrectomy for gastric cancer.<br />

Expression levels <strong>of</strong> the IGF-1R, KIAA1199, TS, Reg IV, and INHBA genes<br />

may be useful biomarkers in patients who receive adjuvant chemotherapy<br />

with S-1.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


256s Gastrointestinal (Noncolorectal) Cancer<br />

4071 General Poster Session (Board #44F), Mon, 8:00 AM-12:00 PM<br />

Efficacy and safety <strong>of</strong> bevacizumab combined with capecitabine in progressive,<br />

metastatic well-differentiated digestive endocrine tumors (BETTER<br />

study). Presenting Author: Emmanuel Mitry, Institut Curie, St. Cloud,<br />

France<br />

Background: Neuroendocrine digestive tumors are highly vascularized<br />

neoplasms known to express the vascular endothelial growth factor (VEGF).<br />

We assessed the activity <strong>of</strong> bevacizumab (Bv), a monoclonal antibody<br />

directed against VEGF combined with capecitabine (CAP). Methods: In this<br />

multicenter, open-label, non-randomized, two-group phase II trial, one<br />

group assessed Bv (7.5 mg/m² on d1/3 weeks) combined with CAP (1,000<br />

mg/m2 twice daily, orally d1-14, resumed on day 22). Eligible patients (pts)<br />

had progressive, metastatic well-differentiateddigestive tract endocrine<br />

tumors (WHO 2000 classification), ECOG-PS �2, Ki 67 index � 15%, no<br />

prior systemic chemotherapy and acceptable organ functions. The primary<br />

endpoint was progression-free survival (PFS). Secondary endpoints were<br />

overall survival, response rate, safety and quality <strong>of</strong> life. Pts were treated for<br />

a minimum <strong>of</strong> 6 months and study duration was 24 months. Results: From<br />

Jun 2007 to Oct 2009, 49 pts were enrolled, 26 (53.1%) were men;<br />

median age 60.3 years (41.2-82.3); ECOG-PS was 0/1 in 46 (93.9%) pts.<br />

Primary tumor site was: small intestine 40 (81.6%) pts, caecum 3 (6.1%),<br />

rectum 4 (8.2%) and stomach 2 (4.1%). Ki-67 proliferative index was<br />

0-2% in 17 (35.4%) pts, 3-4% in 14 (29.2%), 5-9% in 13 (27.1%),<br />

10-14% in 4 (8.3%) Metastatic sites were liver: 46 (93.9%) pts, lymph<br />

nodes: 24 (49.0%), peritoneum 23 (46.9%). Median treatment duration<br />

was 13.8 months; median number <strong>of</strong> cycles was 19. At 24 months, median<br />

PFS was 23.4 months [95%CI: 13.2; not reached] based on 26 events.<br />

PFS rate at 18 months was 55%. Tumor control rate was 87.8% (n�43)<br />

including partial response in 9 (18.4%) pts and stable disease in 34<br />

(69.4%) pts. Survival rate at 24 months was 85%, median overall survival<br />

was not reached, 8 patients died. CTC grade 3/4 Adverse Events (AEs)<br />

occurred in 41 (83.7%) pts, mainly digestive 14 (28.6%) pts. Main G3/4<br />

Bv targeted AE was hypertension in 15 (30.6%) pts. Conclusions: In this<br />

chemotherapy resistant tumor, the combination <strong>of</strong> capecitabine and<br />

bevacizumab, assessed in a phase II study, shows such encouraging results<br />

that this combination may become a reference in the medical treatment <strong>of</strong><br />

ileal NET.<br />

4073 General Poster Session (Board #44H), Mon, 8:00 AM-12:00 PM<br />

Cetuximab, paclitaxel, cisplatin and concurrent radiation in Chinese<br />

patients with locally advanced esophageal squamous cell carcinoma: An<br />

open-label, multicenter, phase II study. Presenting Author: Jinming Yu,<br />

Department <strong>of</strong> Radiation Oncology, Shandong Cancer Hospital and Institute,<br />

Jinan, China<br />

Background: In China more than 90% <strong>of</strong> esophageal malignancies are <strong>of</strong><br />

squamous cell carcinoma (SCC). We conducted this Chinese multicenter<br />

trial to determine the efficacy and safety <strong>of</strong> the addition <strong>of</strong> cetuximab with<br />

paclitaxel, cisplatin, and concurrent radiation for patients with esophageal<br />

SCC and to determine whether KRAS status predicts response. Methods:<br />

Patients with unresectable locally advanced cervical, upper or midesophageal<br />

SCC without distant metastasis were eligible for this open-label<br />

phase II trial. All patients received cetuximab (400 mg/m2 day 1 before<br />

chemoradiotherapy and 250 mg/m2 q1w � 7 weeks), paclitaxel (45 mg/m2 q1w � 7 weeks) and cisplatin (20 mg/m2 q1w � 7 weeks) with 59.4 Gy <strong>of</strong><br />

radiation. The primary end point was response rate. Second end points<br />

included toxicity, overall survival (OS), progression-free survival (PFS), and<br />

KRAS mutation status. Results: Fifty-five patients were enrolled and<br />

evaluable to safety. Non-hematological adverse events were generally grade<br />

1 or 2, and were most <strong>of</strong>ten rash (94.5%), mucositis (58.2%), fatigue<br />

(45.5%), nausea (41.8%) and hepatic dyfunction (40%). Hematologic<br />

adverse events included grade 3 neutropenia (32.7%) and grade 3 anemia<br />

(1.82%). Ten patients did not complete the protocol therapy (6 for<br />

chemotherapy dose delays, 1 for paciltaxel hypersensitivity, 1 by the<br />

treating physicians for unstated reasons, 1 for concurrent unrelated<br />

infection, and 1 for tracheo-esophageal fistula). The response rate was<br />

97.7%. The 1-year OS and median OS was 87.3% and 16.8 months, the<br />

1-year PFS and median PFS was 30.4% and 13.9 months, respectively. No<br />

mutations were detected at KRAS codons 12 or 13 in the 52 available<br />

specimens. Conclusions: Cetuximab can be safely administered with<br />

chemoradiation for Chinese patients with esophageal cancer and may<br />

improve the clinical response rate. KRAS mutations were too rare to be<br />

analyzed as a predictor <strong>of</strong> response.<br />

4072 General Poster Session (Board #44G), Mon, 8:00 AM-12:00 PM<br />

NeoFLOT: Multicenter phase II study <strong>of</strong> perioperative chemotherapy in<br />

resectable adenocarcinoma <strong>of</strong> the gastroesophageal junction or gastric<br />

adenocarcinoma. Presenting Author: Christoph Schulz, Department <strong>of</strong><br />

Hematology and Oncology, Klinikum Grosshadern and Comprehensive<br />

Cancer Center, LMU Munich, Munich, Germany<br />

Background: The rationale <strong>of</strong> the NeoFLOT-trial was the intensification <strong>of</strong><br />

neoadjuvant chemotherapy (NACT) by prolongation <strong>of</strong> preoperative treatment.<br />

This strategy is based on the notion that while perioperative<br />

treatment is notably beneficial in locally advanced gastroesophageal cancer<br />

(GEC), postoperative chemotherapy can only be applied in a fraction <strong>of</strong><br />

patients (pts). Methods: Pts with T3, T4 and/or N� adenocarcinoma (GEC)<br />

were eligible for this multicenter phase II trial. NACT consisted <strong>of</strong> 6 cycles<br />

<strong>of</strong> oxaliplatin 85 mg/m2 , leucovorin 200 mg/m2 , fluorouracil 2600 mg/m2 and docetaxel 50 mg/m2 (FLOT) applied q 2 wks. Staging was performed<br />

after 3 cycles to select pts with progressive disease (PD) for immediate<br />

surgery. Primary endpoint was the R0-resection rate. Secondary endpoints<br />

included the pathological complete response rate (pCR), histologic tumor<br />

regression grade (Becker 2003), safety, and progression-free survival<br />

(PFS). Results: From 10/2009 to 06/2011, 59 pts were enrolled <strong>of</strong> whom<br />

58 pts were assessable for safety. Median age was 61 years (range: 32-79),<br />

58.6% (34/58) had tumors <strong>of</strong> the gastroesophageal junction. 50 pts<br />

underwent surgery and were assessable for the primary endpoint. R0resection<br />

rate was 86.0% (43/50). pCR was achieved in 20.0% (10/50) <strong>of</strong><br />

pts. During NACT, 6.9% (4/58) <strong>of</strong> pts developed progressive disease. Dose<br />

reduction was performed in 43.1% (25/58) <strong>of</strong> pts resulting in a median<br />

dose intensity <strong>of</strong> 89.2%. Grade 3/4 neutropenia was observed in 29.3%<br />

(17/58) <strong>of</strong> pts, febrile neutropenia grade 3/4 in 1.7% (1/58). Common<br />

grade 3/4 non-hematologic adverse events were diarrhea (13.8% (8/58))<br />

and mucositis (6.9% (4/58)). Treatment related mortality was 3.4% (2/58)<br />

with 2 cases <strong>of</strong> sepsis. After a median follow-up <strong>of</strong> 9.1 months, median PFS<br />

and OS have not been reached. Conclusions: These data indicate that NACT<br />

with 6 cycles FLOT is well-tolerated and highly effective in resectable GEC.<br />

4074 General Poster Session (Board #45A), Mon, 8:00 AM-12:00 PM<br />

Oxaliplatin, irinotecan, bevacizumab followed by docetaxel, bevacizumab<br />

in inoperable gastric cancer: First efficacy results <strong>of</strong> a multicenter phase II<br />

trial (AGMT Gastric-3) <strong>of</strong> the Arbeitsgemeinschaft Medikamentöse Tumortherapie.<br />

Presenting Author: Ewald Woell, St. Vinzenz Krankenhaus Betriebs<br />

GmbH, Zams, Austria<br />

Background: In our previous phase II trials (AGMT-Gastric-1 and AGMT<br />

Gastric-2) efficacy <strong>of</strong> oxaliplatin and irinotecan as well as oxaliplatin,<br />

irinotecan and cetuximab was shown. Time to progression however was<br />

short suggesting acquired chemotherapy resistance. Therefore sequential<br />

chemotherapy combined with bevacizumab is investigated in the presented<br />

trial. Methods: Oxaliplatin 85 mg/m2 biweekly (q2w) and irinotecan 125<br />

mg/m2 q2w are administered for the first three months followed by<br />

docetaxel 50mg/m2 q2w for three months. Chemotherapy for 6 months is<br />

combined with bevacizumab 5 mg/kg q2w which is administered until<br />

progression. For this abstract 36 patients (pt.) with histologically proven<br />

unresectable and/or metastatic gastric adenocarcinoma have been evaluated<br />

in a first line setting. Median age: 62.5 years (range 26-80 years), PS<br />

0: 25 patients, PS 1: 10 patients, missing: 1 patient, single metastatic<br />

site: 24 patients, multiple metastases: 10 patients, missing: 2. Results:<br />

Frequently reported adverse events (more than 20% <strong>of</strong> pt.) were predominantly<br />

grade 1 or 2 and included diarrhea (25/36, 69%), polyneuropathy<br />

(17/36, 47%), nausea (17/36, 47%), fatigue (15/36, 42%), neutropenia<br />

(13/36, 36%), abdominal pain (11/36, 31%), hypokalemia (9/36, 25%).<br />

Grade 3 and 4 toxicities included neutropenia (6/36, 17%), diarrhea<br />

(3/36, 8%), hypokalemia (3/36, 8%), anemia in (2/36, 6%), leucopenia<br />

(2/36, 6%), thrombocytopenia (1/36, 3%), nausea in (1/36, 3%). Objective<br />

response rate after 3 cycles was available in 25 patients: CR 1/25<br />

(4%), PR 14/25 (56%), SD 8/25 (32%), PD 2/25 (8%). After 6 cycles<br />

there were 12 evaluable patients with CR 2/12 (16.7%), PR 5/12 (41.7%),<br />

SD 4/12 (33.3%) and PD 1/12 (8.3%). Conclusions: The combination <strong>of</strong><br />

oxaliplatin and irinotecan with bevacizumab followed by docetaxel with<br />

bevacizumab is feasible and very active in advanced gastric cancer.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


4075 General Poster Session (Board #45B), Mon, 8:00 AM-12:00 PM<br />

Post-recurrence survival in patients with previously resected esophageal<br />

adenocarcinoma (EAC). Presenting Author: Mark Lewis, Mayo Clinic<br />

College <strong>of</strong> Medicine, Rochester, MN<br />

Background: EAC recurrence after surgery with curative intent is believed to<br />

carry a uniformly dismal prognosis that may discourage further therapy. To<br />

date, the post-recurrence survival <strong>of</strong> patients has not been examined in<br />

EAC. Our aim was to examine site <strong>of</strong> recurrence in relation to outcome in<br />

EAC patients after surgery. Methods: Among EAC patients (N � 796)<br />

rendered margin-free at surgery performed at Mayo Clinic, most were T3-4<br />

and lymph node (LN)-positive; none received neoadjuvant therapy. The<br />

patient subset who had documented disease recurrence (N � 401) formed<br />

the current study population. Cox models were used to examine overall<br />

survival (OS) post-recurrence. Results: Among patients with recurrence,<br />

median time to recurrence (TTR) was 11 months. Site <strong>of</strong> recurrence<br />

included loco-regional (regional LNs, esophagogastric, anastomosis), chest,<br />

abdomen, or distant sites in 97 (27%), 144 (40%), 181 (50%), and 88<br />

(24%) patients, respectively. Most recurrences (66%) were limited to one<br />

site. Chest-involved recurrence was significantly associated with improved<br />

OS (hazard ratio [HR] 0.78, P � .047), even after adjusting for TTR,<br />

number <strong>of</strong> recurrence sites, tumor pathology, and palliative chemotherapy.<br />

This result was confirmed when multivariate analysis was restricted to<br />

patients who had only 1 recurrence site (Table) or who had biopsy-proven<br />

recurrence (P � .080). In separate models, abdomen-involved (HR � 1.3,<br />

P � .016) or bone-involved (HR � 1.6, P� .008) recurrences were<br />

independently associated with worse OS. Conclusions: Chest-involved<br />

recurrence <strong>of</strong> EAC independently predicts for improved survival, whereas<br />

abdominal and bony sites <strong>of</strong> recurrence predict for worse outcome. Primary<br />

tumor grade and node number were durable prognosticators after recurrence.<br />

These novel data provide useful prognostic information and have the<br />

potential to influence clinical decision-making.<br />

Predictors for post-recurrence survival in a multivariate model.<br />

HRa p<br />

Chest-only, yes v no 0.69b .021<br />

Grade, 4 v 1-3 1.36 .027<br />

Malignant LN, per additionalnode 1.06 .003<br />

Palliative chemo, yes v no 0.57 �.0001<br />

TTR, > v < 11 mos 0.99 .265<br />

a Adjusted for all variables. b 95% confidence interval 0.50 - 0.95.<br />

4077 General Poster Session (Board #45D), Mon, 8:00 AM-12:00 PM<br />

Phase II trial <strong>of</strong> tesetaxel, an oral taxane, as second-line therapy for patients<br />

with advanced gastroesophageal cancer. Presenting Author: Mary Frances<br />

Mulcahy, Northwestern University, Chicago, IL<br />

Background: Tesetaxel is a novel, orally administered taxane. It is not a<br />

substrate for P-glycoprotein, and preclinical data suggest tesetaxel has<br />

potent antitumor activity that exceeds standard taxanes in human tumor<br />

xenografts while causing substantially less neuropathy. Tesetaxel is not<br />

associated with hypersensitivity, thus eliminating the need for premedication<br />

and extended observation. A prior study showed an overall major<br />

response rate (ORR) <strong>of</strong> 20% with tesetaxel used as 2nd-line therapy in<br />

patients (pts) with advanced gastric cancer. We previously showed that<br />

“flat” (i.e., non weight-based) dosing with tesetaxel was associated with<br />

excessive variability; therefore, we amended the final trial design to<br />

evaluate whether we could extend the prior observations in a confirmatory<br />

Phase 2 trial using the MTD. Methods: Eligibility included: adenocarcinoma<br />

<strong>of</strong> stomach/GE junction; 1 prior fluoropyrimidine-platinum regimen; ECOG<br />

PS 0-1; and adequate organ function. Tesetaxel was administered orally<br />

once every 3 weeks starting at 27 mg/m 2 escalated to 35 mg mg/m2 pending tolerance. ORR was the primary endpoint; estimated overall<br />

survival (OS) was secondary. Results: To date, 24 pts -- from a final target <strong>of</strong><br />

27 – have been enrolled (10 men, 14 women; median age, 58 yrs [range,<br />

26-81]). Preliminary ORR in 15 evaluable cases to date are shown in the<br />

table below. At this time, only 4 pts have died and median survival is too<br />

early to estimate. Grade 3-4 adverse events have included neutropenia<br />

(27%), anemia, fatigue (13% each), and anorexia (7%). One pt (7%)<br />

experienced Grade 3 peripheral neuropathy. There have been no episodes<br />

<strong>of</strong> febrile neutropenia. Conclusions: Oral tesetaxel administered once every<br />

3 weeks is active against gastric cancer in the 2nd-line setting. The<br />

observed 20% ORR confirms earlier data and appears at least equivalent to<br />

docetaxel (ORR� 5%-19%), as reported in 4 prior studies in this<br />

population. Tesetaxel was well tolerated and was not been associated with<br />

hypersensitivity. We expect to present final ORR and estimated OS from<br />

this trial.<br />

Response N � 24<br />

Evaluable (n) 15<br />

<strong>Part</strong>ial 3 (20%)<br />

Stable disease 3<br />

Progression 9<br />

Too early 10<br />

Response duration, mos 1.5�,3�,4�<br />

Gastrointestinal (Noncolorectal) Cancer<br />

257s<br />

4076 General Poster Session (Board #45C), Mon, 8:00 AM-12:00 PM<br />

Prognostic impact <strong>of</strong> FHIT, APC, and HER2 status in resected gastric<br />

cancer: A clinical-biological risk stratification model. Presenting Author:<br />

Emilio Bria, Department <strong>of</strong> Pathology and Diagnostics, University <strong>of</strong><br />

Verona, Verona, Italy<br />

Background: The dismal prognosis <strong>of</strong> gastric cancer prompts for the<br />

identification <strong>of</strong> factors predictive <strong>of</strong> survival/response to treatment. The<br />

potential prognostic value <strong>of</strong> 11 molecular markers was investigated in a<br />

retrospective series <strong>of</strong> 208 resected gastric cancers. Methods: Molecular/<br />

clinicopathological data were correlated to cancer-specific/overall survival<br />

(CSS/OS) using a Cox model. Molecular data were: microsatellite instability,<br />

CDX2, APC, ß-catenin, E-Cadherin, FHIT, P53, p21, HER-2 (IHC/<br />

FISH), TOPO2A amplification (FISH) e PIK3CA mutation (exons 9/20).<br />

ROC analysis provided optimal cut-<strong>of</strong>fs and model validation. A logistic<br />

equation with regression analysis coefficients was constructed to estimate<br />

individual patients’ probability (IPP) <strong>of</strong> death. Internal cross-validation<br />

(100 simulations, 80% <strong>of</strong> the dataset) was accomplished. Multivariate<br />

model Ratios served to derive a prognostic continuous score to identify risk<br />

classes. Results: 208 patients were studied (median follow-up 16 months,<br />

range 1-155). Multivariate analysis selected 8 independent CSS predictors:<br />

sex (HR 1.53, p�0.04), stage (HR 5.40, p�0.0001), R (HR 2.69,<br />

p�0.0001), localization (HR 1.64, p�0.008), LN (HR 1.55, p�0.02),<br />

APC (HR 1.91, p�0.001), FHIT (HR 1.54, p�0.05) and HER-2 (HR 1.92,<br />

p�0.08). Independent OS predictors were: sex, age, stage, R, localization,<br />

LN, APC and HER-2. Cross-validation analysis confirmed APC, FHIT and<br />

HER-2 as the biological variables displaying significant correlation with<br />

CSS (replication: 98%, 51%, 31%). The multivariate model predicted a<br />

2-yr IPP <strong>of</strong> cancer-death/death with a prognostic accuracy <strong>of</strong> 0.87 (SE<br />

0.02)/0.91 (SE 0.02). A 2-class risk stratification model was developed<br />

with the ROC generated cut-<strong>of</strong>f prognostic score (AUC 0.87, SE 0.03;<br />

Sensitivity 85.3%, Specificity 78.9%); see table below. This 2-class model<br />

has a prognostic performance for CSS/OS <strong>of</strong> 0.82 (SE 0.03)/0.81 (SE<br />

0.02). Conclusions: FHIT, APC and HER-2 represent powerful prognostic<br />

factors for resected GC and may complement clinical parameters to<br />

accurately predict individual patient risk.<br />

Low risk (score < 6) High risk (score > 6) p value<br />

CSS 82.5% 16.4% �0.0001<br />

OS 79.7% 15.6% �0.0001<br />

4078 General Poster Session (Board #45E), Mon, 8:00 AM-12:00 PM<br />

Outcome <strong>of</strong> 58 trimodality-eligible esophagogastric cancer (EC) patients<br />

who achieved clinical complete response (cCR) after preoperative chemoradiation<br />

but then declined surgery. Presenting Author: Takashi Taketa,<br />

University <strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: For patients with EC who can withstand surgery, the preferred<br />

therapy is trimodality. However, after achieving a cCR (defined as postchemoradiation<br />

negative endoscopic biopsy for cancer and post-chemoradiation<br />

physiologic FDG uptake by PET), some patients are tempted to<br />

decline surgery. Literature is sparse on the outcome <strong>of</strong> such patients.<br />

Methods: Between 2002 and 2011, we identified 621 trimodality-eligible<br />

EC patients in our prospective database. All patients had to be trimodalityelgible<br />

and must have received preoperative chemoradiation and completed<br />

preoperative staging that included a repeat endoscopic biopsy and<br />

PET-CT prior to surgery among other routine tests. Results: Of 621<br />

trimodality-eligible patients identified, 58 patients declined surgery after<br />

completing chemoradiation. All patients had a cCR. The median age was<br />

69 (range, 47-85). Male (84.5%) and Caucasian (91.4%) were dominant.<br />

Baseline stage was II (44.8%) or III (51.7%) and histology was adenocarcinoma<br />

(67.2%) or squamous cell carcinoma (29.3%). 40 patients remain<br />

alive at a median follow up <strong>of</strong> 50.4 months (95% CI, 38.6-62.1). 5-year OS<br />

and relapse-free survival were 56.7�9.0% and 32.9�7.7%. Of 12<br />

patients with local recurrence during surveillance, 11 had salvage resection.<br />

Conclusions: Although, the outcome <strong>of</strong> EC patients with cCR who<br />

declined surgery appears reasonable, in the absence <strong>of</strong> a validated<br />

prediction/prognosis model, only trimodality therapy must be encouraged<br />

for trimodality-eligible patients. Supported by UT M. D. Anderson Cancer<br />

Center grants and generous donors.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


258s Gastrointestinal (Noncolorectal) Cancer<br />

4079 General Poster Session (Board #45F), Mon, 8:00 AM-12:00 PM<br />

Phase II study <strong>of</strong> oxaliplatin and sorafenib in advanced gastric cancer after<br />

failure <strong>of</strong> cisplatin-fluoropyrimidine-based (PF) treatment. Presenting Author:<br />

Rosa Gallego, Hospital Clinic, Barcelona, Spain<br />

Background: PF-based chemotherapy is the standard first line treatment in<br />

advanced GC. Although some drugs as taxanes or oxaliplatin (Ox) have<br />

shown efficacy in combination with P or F, no established second line<br />

exists. RAF/MEK/ERK pathway activation as well as VEGF expression have<br />

been related with more advanced GC. Sorafenib (S) is a potent Raf and<br />

VEGFR inhibitor and Ox has demonstrated non-cross resistance with P.<br />

Methods: A multicenter phase II to evaluate the efficacy <strong>of</strong> OX- S<br />

combination in gastric or GEJ adenocarcinoma (pts) with progressive<br />

disease after CF based first line chemotherapy, was planned. Other<br />

inclusion criteria: ECOG 0-2, measurable disease and adequate organ<br />

function. Treatment doses were Ox 130mg/m2 /3 weeks and S 800<br />

mg/bid/d until progression or toxicity. CT scan evaluation every 3 cycles.<br />

The study followed a A-Hern single-stage design (HR � 1.67, median<br />

expected PFS <strong>of</strong> 4.3m if treatment ineffective, sample size: 43). The study<br />

was closed with the inclusion <strong>of</strong> 40 pts, maintaining a power � 87%. Ten<br />

pts would have to be alive and free <strong>of</strong> progression at 12m to consider the<br />

study positive. Results: Forty pts were included and evaluable for PFS.<br />

Thirty-six pts were evaluable for response. Median age was 63 ( range 39 -<br />

76) years, ECOG 0/1/2 in 36/59/5 % <strong>of</strong> pts. PFS to 1st line was � 6m in<br />

59% <strong>of</strong> pts. One CR and, 47.2% <strong>of</strong> SD were observed. Gr ¾ toxicity (%)<br />

was: neutropenia (9.8), thrombocitopenia (7.3), neurotoxicity (4.9) and<br />

diarrhea (4.9). Median PFS was 3 (95 %CI: 2.3-4.1 )m and median OS was<br />

6.5 ( 95% CI: 5.2-9.6)m. PFS at 12 m was 0%. Median OS in pts with PFS<br />

to 1º line � 6m/ �6m was 9.7m and 5.6m respectively (p0.04).<br />

Conclusions: The combination <strong>of</strong> Oxaliplatin-Sorafenib in advanced GC<br />

patients previously treated with CF shows a safe pr<strong>of</strong>ile but these results do<br />

not support the implementation <strong>of</strong> a phase III trial. Our results show that<br />

PFS under a CP-based first line therapy determine subgroups <strong>of</strong> GC<br />

patients with different clinical behaviors.<br />

4081 General Poster Session (Board #45H), Mon, 8:00 AM-12:00 PM<br />

Safety <strong>of</strong> everolimus (EVE) in Asian patients (pts) with advanced gastric<br />

cancer (AGC) enrolled in the phase III GRANITE-1 study. Presenting<br />

Author: Kun-Huei Yeh, National Taiwan University Hospital, Taipei, Taiwan<br />

Background: Previous subanalyses <strong>of</strong> the RECORD-1 (Jpn J Clin Oncol<br />

2011;41:17-24) and RADIANT-3 (GICS 2011; abs. 289) studies <strong>of</strong> the<br />

oral mammalian target <strong>of</strong> rapamycin inhibitor EVE showed that incidences<br />

<strong>of</strong> stomatitis, rash, infections, and pneumonitis were greater in Asian pts;<br />

however, the number <strong>of</strong> Asian pts enrolled was small. GRANITE-1 provides<br />

an opportunity to evaluate EVE safety in a broader Asian population.<br />

Methods: In the randomized, double-blind, phase 3 GRANITE-1 study, pts<br />

aged �18 y with confirmed AGC and disease progression after 1 or 2 lines<br />

<strong>of</strong> systemic chemotherapy were randomized 2:1 to EVE 10 mg/d � best<br />

supportive care (BSC) or placebo � BSC. Randomization was stratified by<br />

region (Asia [China, Japan, Korea, Taiwan, Thailand, Hong Kong] vs rest <strong>of</strong><br />

world [ROW]) and previous lines <strong>of</strong> chemotherapy (1 vs 2). Study drug was<br />

continued until disease progression or unacceptable toxicity. Adverse<br />

events (AEs) were collected throughout the study and for 28 d after final<br />

dose <strong>of</strong> study drug and assessed according to the Common Terminology<br />

Criteria for Adverse Events, v 3.0. Primary endpoint was overall survival.<br />

Safety was a secondary endpoint. Results: Of the 656 pts in the study, 363<br />

(55%) were enrolled in Asia and received EVE (n�243) or placebo<br />

(n�120). EVE did not significantly reduce the risk <strong>of</strong> death in the overall<br />

(HR 0.90; 95% CI 0.75-1.08), Asian (HR 0.96; 95% CI 0.75-1.23), or<br />

ROW (HR 0.85; 95% CI 0.65-1.10) populations. The most common<br />

any-grade AEs among EVE-treated pts (Asia vs ROW) were decreased<br />

appetite (45% vs 51%), stomatitis (44% vs 35%), and fatigue (34% vs<br />

35%). Among EVE-treated pts, rates <strong>of</strong> any-grade rash, infections, and<br />

pneumonitis (Asia vs ROW) were 22% vs 18%, 24% vs 28%, and 4% vs<br />

2%, respectively; rates <strong>of</strong> grade 3/4 stomatitis, rash, infections, and<br />

pneumonitis (Asia vs ROW) were 4% vs 6%, 0% vs 0.5%, 6% vs 9%, and<br />

0.4% vs 1%, respectively. No grade 4 pneumonitis was observed. Only 1 pt<br />

(from Asia) discontinued due to pneumonitis (grade 3). Conclusions: The<br />

safety pr<strong>of</strong>ile <strong>of</strong> EVE in Asian pts with AGC was similar to that observed in<br />

ROW pts with AGC and with EVE in other cancers. No new safety signals<br />

were identified. Pneumonitis was relatively uncommon in Asian and ROW<br />

pts.<br />

4080 General Poster Session (Board #45G), Mon, 8:00 AM-12:00 PM<br />

Feasibility <strong>of</strong> perioperative chemotherapy with infusional 5-FU, leucovorin,<br />

and oxaliplatin with or without docetaxel, in elderly patients with locally<br />

advanced esophagogastric cancer. Presenting Author: Sylvie Lorenzen,<br />

National Center for Tumor Diseases, University <strong>of</strong> Heidelberg, Heidelberg,<br />

Germany<br />

Background: The aim <strong>of</strong> this study was to determine feasibility and efficacy<br />

<strong>of</strong> perioperative chemotherapy in elderly, potentially operable esophagogastric<br />

cancer patients. Methods: Patients aged 65 or older with locally<br />

advanced esophagogastric adenocarcinoma were randomized to perioperative<br />

chemotherapy consisting <strong>of</strong> four preoperative and four postoperative<br />

cycles <strong>of</strong> infusional 5-FU, leucovorin, and oxaliplatin (FLO) with or without<br />

docetaxel 50mg/m² (FLOT), every 2 weeks. Results: 44 patients with a<br />

median age <strong>of</strong> 70 years were randomized to either FLO (22) or FLOT (22)<br />

chemotherapy. In the FLO and FLOT group, 20 and 18 patients completed<br />

four cycles <strong>of</strong> preoperative chemotherapy and 17(77.3%) and 15(68.2%)<br />

patients proceeded to surgery, with 88.2% and 93.3% R0 resections,<br />

respectively. FLOT was associated with significantly more treatmentrelated<br />

NCI-CTC grade 3/4 adverse events, including neutropenia<br />

(p�.0001), leukopenia (p�.0001), stomatitis (p�0.02) and nausea<br />

(p�0.02) and a higher rate <strong>of</strong> patients experiencing a �10-points<br />

deterioration <strong>of</strong> EORTC QoL global health status scores (FLOT, 54%; FLO<br />

23.1%) at 8 weeks. No perioperative mortality was observed, however<br />

postoperative morbidity was nearly doubled with the FLOT regimen (60%<br />

versus 35.3% for FLO), mostly due to wound infections in the FLOT group<br />

(20%). 11 (64.7%) and 9 (60.0%) <strong>of</strong> patients in the FLO/FLOT group were<br />

able to undergo postoperative chemotherapy. Compared with the FLO<br />

group, the FLOT group had a better response rate (61.9% vs. 18.2%) and a<br />

trend towards an improved median disease free survival (21.1 vs. 12.0<br />

months; p�0.09). Conclusions: Age alone is not a contra-indication for the<br />

selection <strong>of</strong> patients for three-drug regimens in the multimodal treatment<br />

approach. FLOT seems to be more effective than FLO, but is associated<br />

with an increased toxicity and higher postoperative morbidity. Therefore,<br />

careful patient selection is warranted.<br />

4082 General Poster Session (Board #46A), Mon, 8:00 AM-12:00 PM<br />

A phase II study <strong>of</strong> tivantinib monotherapy in patients with previously<br />

treated advanced or recurrent gastric cancer. Presenting Author: Kei Muro,<br />

Aichi Cancer Center Hospital, Nagoya, Japan<br />

Background: Tivantinib is a selective, non-ATP competitive, small-molecule<br />

inhibitor <strong>of</strong> c-MET and is under development in several cancers such as<br />

NSCLC and HCC. Elevated expressions <strong>of</strong> c-MET and its ligand, hepatocyte<br />

growth factor, have been frequently found in gastric cancer, and are<br />

associated with a more aggressive phenotype. In this single-arm phase II<br />

study, we evaluated the efficacy <strong>of</strong> tivantinib monotherapy in Asian patients<br />

(pts) with previously treated metastatic gastric cancer (MGC). This is the<br />

first clinical trial evaluating the efficacy <strong>of</strong> a selective c-MET inhibitor for<br />

MGC. Methods: Eligibility criteria included MGC with at least 1 measurable<br />

lesion per RECIST; 1 or 2 prior chemotherapy regimens; ECOG PS 0 or 1;<br />

and normal CYP2C19 metabolism. Tivantinib was daily administered 360<br />

mg bid orally. The primary endpoint was disease control rate (DCR) defined<br />

as CR, PR or SD after 8 weeks administration. Pretreatment tumor tissue<br />

collection was required to evaluate the relationship between biomarkers<br />

and efficacy. Pharmacokinetic (PK) evaluation was also conducted. Results:<br />

Thirty patients received tivantinib and were eligible for analysis: median<br />

age 62.5 (41-70) years; ECOG PS 0/1 (8/22); 1/2 prior regimen (16/14);<br />

12 pts (40%) with prior gastrectomy. No objective response was observed,<br />

and DCR was 36.7% (11/30 pts). Median progression-free survival was 43<br />

(95% CI: 29.0-92.0) days. Grade 3 or 4 adverse events (AEs) occurred in<br />

13 pts (43.3%), in which neutropenia (n � 4) and anemia (n � 4) were<br />

recognized as drug-related. Only 2 pts discontinued due to AEs. There is no<br />

treatment-related death and novel safety concern. c-MET gene amplification<br />

(� 5 copies/cell) was observed in 4 pts (13.3%). No obvious<br />

relationship <strong>of</strong> treatment outcome with biomarkers including c-MET gene<br />

amplification, c-MET, p-c-MET and HGF expression in tumor and serum<br />

HGF was identified. Gastrectomy and/or ethnicity (Korean or Japanese) did<br />

not affect PK parameters. Conclusions: Tivantinib as a monotherapy showed<br />

only a modest efficacy in previously treated MGC, and further trial testing in<br />

combination would be warranted in MGC. It would be an important finding<br />

that gastrectomy do not affect PK pr<strong>of</strong>ile <strong>of</strong> tivantinib.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


4083 General Poster Session (Board #46B), Mon, 8:00 AM-12:00 PM<br />

Interim safety results from a phase II/III trial with 5-FU, oxaliplatin, and<br />

docetaxel (FLOT) versus epirubicin, cisplatin, and 5-FU (ECF) in patients<br />

with locally advanced, resectable gastric/oesophagogastric junction (OGJ)<br />

cancer: A study <strong>of</strong> the AIO. Presenting Author: Ulrike Koock, Krankenhaus<br />

Nordwest, UCT-University Cancer Center, Frankfurt, Germany<br />

Background: Perioperative ECF is a standard treatment for localized<br />

gastric/OGJ adenocarcinoma. However, 5-year survival rate remain below<br />

40%. The FLOT regime is an effective new combination with pathologic<br />

response rates in the 15% range. This phase II/III study compares both<br />

regimens in resectable stages. Methods: Pts are stratified by different<br />

baseline criteria and randomized to either 3 � 3 perioperative cycles <strong>of</strong><br />

ECF (Epirubicin 50 mg/m2 , d1 Cisplatin 60 mg/m², d1 5-FU 200 mg/m²,<br />

d1-d21, qd21) or 4 � 4 cycles <strong>of</strong> perioperative FLOT (Docetaxel 50mg/m2,<br />

d1 5-FU 2600 mg/m², d1 leucovorin 200 mg/m², d1 Oxaliplatin 85 mg/m²,<br />

d1, qd14). 5-FU can be replaced by Capecitabine in the ECF-arm (ECX).<br />

This is a preplanned toxicity analysis after 200 pts in order to decide<br />

whether to continue to a phase III trial. Results: 202 pts have been<br />

randomized so far. Median age is 62 yrs; 79% <strong>of</strong> pts are male. The<br />

Primaries were Gastric in 43.4%, OGJ in 53.2% and not evaluable/<br />

documented in 3.4% <strong>of</strong> pts. 190 pts were eligible for the safety analyses.<br />

Median no. <strong>of</strong> preoperative cycles was 3 and 4 with ECF/ECX and FLOT,<br />

respectively, and 68.7% vs. 66.0% <strong>of</strong> pts (ECF/ECX vs. FLOT) started<br />

postoperative chemotherapy (ct). Grade 3/4 side effects were observed in<br />

52.1% <strong>of</strong> ECF/ECX and 59.6% <strong>of</strong> FLOT pts with no significant differences<br />

between arms regarding individual grade 3/4 toxicities. Thromboembolic<br />

events occurred in 14.5% vs. 8.5% in pts with ECF/ECX vs. FLOT (p�.25).<br />

Serious adverse events occurred in 60.3% vs. 39.7% <strong>of</strong> pts with ECF/ECX<br />

vs. FLOT. Preoperative delay/interruptions <strong>of</strong> ct were observed in 74.0% vs.<br />

61.7% <strong>of</strong> pts with ECF/ECX vs. FLOT (p�.07). Dose modifications <strong>of</strong><br />

preoperative ct were performed in 28.1% vs. 22.3% <strong>of</strong> treatment cycles<br />

with ECF/ECX vs. FLOT, respectively. 121 pts underwent surgery. Severe<br />

surgical morbidity was similar in both arms (ECF/ECX, 15.8%; FLOT,<br />

14.1%). Surgical mortality was observed in 2 and 1 pts with ECF/ECX and<br />

FLOT. Toxic deaths were observed in 1 pt each. Conclusions: Perioperative<br />

FLOT is feasible and safe. This supports the continuation <strong>of</strong> the trial as a<br />

phase III.<br />

4085 General Poster Session (Board #46D), Mon, 8:00 AM-12:00 PM<br />

Relief <strong>of</strong> bowel-related symptoms with telotristat etiprate in octreotide<br />

refractory carcinoid syndrome: Preliminary results <strong>of</strong> a double-blind,<br />

placebo-controlled multicenter study. Presenting Author: Thomas M.<br />

O’Dorisio, University <strong>of</strong> Iowa Hospitals and Clinics, Iowa City, IA<br />

Background: Diarrhea associated with carcinoid syndrome has been attributed<br />

to tumor production <strong>of</strong> serotonin. Telotristat etiprate, (LX1032,<br />

LX1606), is an oral inhibitor <strong>of</strong> peripheral serotonin synthesis. This study<br />

explored the safety, tolerability, and efficacy <strong>of</strong> telotristat etiprate in<br />

carcinoid patients with octreotide-refractory diarrhea. Methods: Carcinoid<br />

patients with �4 bowel movements (BMs)/day on octreotide were randomized<br />

3:1 to receive telotristat etiprate or placebo as double-blind treatment.<br />

Patients enrolled in sequential, escalating dose cohorts <strong>of</strong> 150, 250, 350,<br />

or 500 mg tid, followed by a 500 mg tid expansion cohort. Patients were<br />

followed for toxicity, 24-hr urinary 5-HIAA (u5-HIAA), BM frequency, and<br />

self-reported bowel-related symptoms. Subjects were asked “In the past 7<br />

days, have you had adequate relief <strong>of</strong> your carcinoid syndrome bowel<br />

complaints such as diarrhea, urgent need to have a bowel movement,<br />

abdominal pain or discomfort?” Responses (yes or no) were analyzed as<br />

categorical variables. Results: 16 patients enrolled in the 4 escalating dose<br />

cohorts and 7 in the expansion cohort; 18 on telotristat etiprate and 5 on<br />

placebo. Median age: 62 yrs; mean 6.3 BMs/day (range, 4-10). AEs<br />

included primarily mild-moderate diarrhea, nausea, and abdominal discomfort.<br />

In treated subjects, adequate relief was reported as: Week 1 - 6/18<br />

(33.3%), Week 2 - 5/16 (31.3%), Week 3 - 5/15 (33.3%), and Week 4 -<br />

6/13 (46.0%). No placebo subjects reported improvement at any timepoint.<br />

Biochemical response (�50%reduction in u5-HIAA) and BM response<br />

(�30% reduction in daily BMs for 2 weeks) were associated with<br />

reporting <strong>of</strong> adequate relief. For evaluable telotristat etiprate-treated<br />

patients, 9/16 (56%) experienced a biochemical response and 5/18 (28%)<br />

experienced a clinical (BM) response; no placebo subjects achieved either<br />

biochemical or clinical response. Conclusions: Treatment with telotristat<br />

etiprate was associated with decreases in u5-HIAA and BM frequency, and<br />

with self-reported relief <strong>of</strong> bowel related symptoms. Treatment in an<br />

extension phase with open-label telotristat etiprate is ongoing.<br />

Gastrointestinal (Noncolorectal) Cancer<br />

259s<br />

4084 General Poster Session (Board #46C), Mon, 8:00 AM-12:00 PM<br />

Assessment <strong>of</strong> the 7th edition <strong>of</strong> the AJCC classification and a proposal <strong>of</strong> a<br />

new classification in patients with gastric cancer undergoing D2 gastrectomy.<br />

Presenting Author: Vincenzo Catalano, UOC Oncologia, A. O., Pesaro,<br />

Italy<br />

Background: Studies on Asian, US, and German patients have moved some<br />

criticisms on the validity <strong>of</strong> the 7th edition <strong>of</strong> the AJCC classification to<br />

discriminate outcome <strong>of</strong> gastric cancer stages. We investigated the effect<br />

<strong>of</strong> this AJCC classification in a high-quality surgical populations <strong>of</strong> patients<br />

receiving D2 lymphadenectomy. Methods: From the prospective database<br />

at San Salvatore Hospital, Pesaro, we identified 515 patientswith gastroesophageal<br />

junction (Siewert II and III) or stomach adenocarcinoma who<br />

underwent gastrectomy with curative intent from 1998 to 2010. Lymphadenectomy<br />

extended to the 3rd level 12p/b nodes (D2/D3) was performed in<br />

all patients. Overall survival (OS) probabilities, calculated from the date <strong>of</strong><br />

surgery to the date <strong>of</strong> death, from any cause, were estimated using the<br />

Kaplan-Meier method and compared using the log-rank test. Results: 58%<br />

<strong>of</strong> patients were male,median age was 73 years (range 36-96). Median<br />

number <strong>of</strong> examined lymph nodes was 32 (range, 1-89), and only 8.9% <strong>of</strong><br />

patients had less than 15 examined lymph nodes; 96 patients received<br />

adjuvant chemo- or chemoradiotherapy. As shown in the table, we proposed<br />

a revised staging system (Pesaro Staging System, PSS), which performs<br />

better than the 7th edition <strong>of</strong> AJCC classification in terms <strong>of</strong> survival<br />

differences between stages. Conclusions: This study confirms once again<br />

that the 7th edition <strong>of</strong> the AJCC classification does not discriminate<br />

adequately the outcome from stage to stage. In a European population <strong>of</strong><br />

patients undergoing gastrectomy plus at least D2 lymphadenectomy, the<br />

revised staging system, PSS, better defines patient prognosis.<br />

7th edition AJCC Staging System Pesaro Staging System<br />

Stage Groupings n 5-yr OS (%) p* Stage Groupings n 5-yr OS (%) p*<br />

IA T1N0 106 86.6 I T1N0-2, T2N0 165 85.7<br />

IB T2N0, T1N1 55 83.3 .9926 II T3-4aN0, T2N1-2 84 68.4 .0018<br />

IIA T3N0, T2N1, T1N2 61 67.6 .0447 IIIA T3-4aN1-2 105 53.8 .044<br />

IIB T4aN0, T3N1, T2N2, T1N3 65 64.8 .7468 IIIB TxN3, T4bNx 117 28.1 .0023<br />

IIIA T4aN1, T3N2, T2N3 58 56.3 .3845 IV TxNxM1 44 4.6 �.0001<br />

IIIB T4bN0-1, T4aN2, T3N3 87 35.6 .0376<br />

IIIC T4bN2-3, T4aN3 39 8.0 .0016<br />

IV TxNxM1 44 4.6 .0741<br />

* Compared with row above.<br />

4086 General Poster Session (Board #46E), Mon, 8:00 AM-12:00 PM<br />

Association <strong>of</strong> clinical complete response (cCR) after preoperative chemoradiation<br />

and pathological complete response (pathCR) in patients with<br />

gastroesophageal cancer (GEC) and indispensability <strong>of</strong> trimodality therapy<br />

(TMT). Presenting Author: Naga K Sucharita Cheedella, University <strong>of</strong> Texas<br />

M. D. Anderson Cancer Center, Houston, TX<br />

Background: TMT strategy has the highest level-1 evidence for treating<br />

localized GEC. High rates <strong>of</strong> cCR (defined as post-chemoradiation negative<br />

endoscopic biopsy and physiologic uptake on PET) are common and have<br />

questioned the benefit from surgery in patients with cCR after chemoradiation.<br />

We hypothesized that cCR would be associated with a high rate <strong>of</strong><br />

pathCR than � cCR. Methods: The data were analyzed retrospectively in<br />

563 patients who had esophagectomy for GEC in between 2002 and 2010<br />

at UTMDACC. Among them, 284 had TMT and post-chemoradiation<br />

endoscopic biopsies and PET (before surgery). Multiple statistical methods<br />

were used. Results: Of these 284 TMT patients, 218 (77%) patients<br />

achieved a cCR. However, only 67 (31%) <strong>of</strong> 218 had a pathCR. The<br />

sensitivity <strong>of</strong> cCR for pathCR was 97.1 % (67/69) but the specificity was<br />

low, 29.8 % (64/215). Intriguingly, 66 patients who had � cCR, only 2<br />

patients (3%) had a pathCR. The difference in the rate <strong>of</strong> pathCR between<br />

the cCR and � cCR groups was significant (P � 0.001). Conclusions: Our<br />

data show that cCR is frequent after chemoradiation but the pathCR rate is<br />

not high and it is associated with specificity that is too low for clinical<br />

implementation. Therefore, all TMT-eligible patients, irrespective <strong>of</strong> the<br />

achievement <strong>of</strong> cCR or � cCR must be encouraged to undergo surgery.<br />

Therapies that overcome chemoradiation resistance and could increase the<br />

pathCR rate are needed for esophageal preservation in select GEC patients.<br />

Supported by UTMDACC and generous donors.<br />

Path CR<br />

cCR<br />

� -<br />

Total<br />

� 67 151 218<br />

- 2 64 66<br />

Total 69 215 284<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


260s Gastrointestinal (Noncolorectal) Cancer<br />

4087 General Poster Session (Board #46F), Mon, 8:00 AM-12:00 PM<br />

Tolerability and efficacy <strong>of</strong> a biweekly docetaxel, fluorouracil, leucovorin,<br />

oxaliplatin regimen (TFOX) in previously advanced metastatic gastric and<br />

oesogastric junction (AGC) adenocarcinoma. Presenting Author: Simon<br />

Pernot, Hôpital Européen Georges Pompidou, Paris, France<br />

Background: The DCF regimen is superior to 5-FU-Cisplatin in term <strong>of</strong><br />

response rate (RR) and overall survival (OS) in advanced gastric cancer, but<br />

is also more toxic. Oxaliplatin is better tolerated and can effectively replace<br />

cisplatin in AGC patients (pts). We hypothesize that incorporating Docetaxel<br />

into a simplified FOLFOX regimen should be a tolerable and<br />

efficient option, and could facilitate the use <strong>of</strong> this drug in 1st line in AGC.<br />

Methods: TFOX regimen was given biweekly as follow: Docetaxel (50mg/<br />

m2), oxaliplatin (85mg/m2), leucoverin (400mg/m2) and 5FU continuous<br />

infusion 48h (2400mg/m2). Main inclusion criteria were: pts with histollogically<br />

proven metastatic or locally advanced gastric or oesogastric junction<br />

adenocarcinoma, previously untreated, PS�2. Using Fleming design the<br />

primary end point was response rate, needing a sample size <strong>of</strong> 40 pts.<br />

Results: Between 02/2008 and 07/2011, 41 pts with AGC were enrolled<br />

(oesogastric junction: 17 pts, diffuse type: 22 pts, metastatic: 37 pts);<br />

mean age was 53.5 years (31-73), 26 pts were male. PS 0/1/2: 11/24/6<br />

pts, 14 pts had a relapse after a R0 resection. The total number <strong>of</strong> cycles<br />

administered was 310 (mean 7.6 cycles/pts) and 90% <strong>of</strong> pts received at<br />

least 4 cycles. No toxic death occurred. Grade 3-4 toxicities were observed<br />

in 20 pts (48%). Grade 3-4 toxicities were neutropenia (29%) febrile<br />

neutropenia (7%), neurotoxicity (10%), asthenia (10%). ORR was 63%<br />

(n�26) [IC95% 48-78], with 2 complete responses. Disease control rate<br />

(CR�PR�SD) was 78% (n�32) [IC95% 65-90]. Median progression free<br />

survival and overall survival were 6.5 months [IC95% 5.02-10.85] and<br />

12.1 months [IC95% 7.25-15.28], respectively. Conclusions: TFOX is<br />

effective with a manageable toxicity pr<strong>of</strong>ile in first line treatment for AGC<br />

pts. Tolerability <strong>of</strong> this regimen compares favourably with DCF reported<br />

tolerability. TFOX may be an alternative option for intensive 1st line<br />

treatment and should now be evaluated in randomized trials.<br />

4089 General Poster Session (Board #46H), Mon, 8:00 AM-12:00 PM<br />

Assessment <strong>of</strong> HER2 status from an epidemiology study in tumor tissue<br />

samples <strong>of</strong> gastric and gastro-esophageal junction cancer: Spanish results<br />

<strong>of</strong> the HER-EAGLE study. Presenting Author: Lourdes Gomez, Hospital<br />

Virgen del Rocío, Sevilla, Spain<br />

Background: Overexpression <strong>of</strong> HER2 is an important biomarker in gastroesophageal<br />

junction (GEJ) and gastric cancer (GC) and a predictive factor<br />

for trastuzumab (T, Herceptin). The ToGA phase III trial showed the benefit<br />

in overall survival <strong>of</strong> adding trastuzumab to chemotherapy inHER2-positive<br />

advanced GEJ/GC patients, following validated scoring criteria by a central<br />

laboratory. This study examines the incidence <strong>of</strong> HER2 positivity (local<br />

laboratories) in GEJ/GC according to EMA Herceptin label. Methods:<br />

HER-EAGLE was an epidemiological, non–interventional, international<br />

study assessing HER2 status by IHC/ISH in tumor samples from any stage<br />

GEJ/GC patients. Neutral buffered 10% formalin embedded tissues (surgical<br />

excision specimens or minimum 6-8 biopsies) analyzed via validated<br />

Ventana and Dako methods and scoring criteria used in ToGA: HER2positive<br />

if IHC 3� or IHC 2� (FISH/SISH confirmed; HER2/CEP17 ratio �<br />

2.0); HER2-negative if IHC 0 or IHC 1�. Overall and subgroup estimates<br />

calculated with 95%CI. Data from the Spanish cohort are presented.<br />

Results: The Spanish cohort <strong>of</strong> HER-EAGLE included 1,954 participants<br />

(63.7% males) from 33 hospitals (Dec2010 to Aug2011), with 469<br />

biopsies (24.0%) and 1,479 excisions (75.7%). Samples were 13.7% GEJ<br />

and 86.3% GC, and the adenocarcinomas were 1,137 intestinal (58.2%),<br />

510 diffused (26.1%), 163 mixed (8.3%; Laureen classification), and 144<br />

not available (7.3%). Median time from sampling to HER2 analysis <strong>of</strong> 5.1<br />

years (range from days to 12 years). Total <strong>of</strong> 210 cases tested IHC 3�<br />

(10.7%), 215 IHC 2� (11.0%), 308 IHC 1� (15.8%), and 1,221 IHC 0<br />

(62.5%). Overall, HER2 positivity (IHC 3� or IHC2�/ISH�) was 14.1%<br />

(95%CI 12.61 – 15.75). HER2 positivity by histological type was higher in<br />

intestinal adenocarcinomas (19.5%) compared to diffused (4.5%) or in<br />

mixed (9.2%). Conclusions: HER-EAGLE confirmed the feasibility <strong>of</strong><br />

community based HER2 testing in GC as the first study with a high number<br />

<strong>of</strong> samples analyzed by local laboratories. The incidence <strong>of</strong> HER2-positivity<br />

(EMA label definition) was similar to the ToGA screening population, and it<br />

was again higher in adenocarcinomas <strong>of</strong> intestinal type (19.5%).<br />

4088 General Poster Session (Board #46G), Mon, 8:00 AM-12:00 PM<br />

Prognostic value <strong>of</strong> lemur tyrosine kinase-3 (LMTK3) polymorphism in<br />

Japanese (J) patients (PTS) with localized gastric adenocarcinoma (GAC).<br />

Presenting Author: Afsaneh Barzi, Norris Comprehensive Cancer Center,<br />

University <strong>of</strong> Southern California, Los Angeles, CA<br />

Background: LMTK3 is an estrogen receptor � (ER�) regulator. Recent<br />

studies show that [rs808419(r8) and rs9989661(r9)] and LMTK3 expression<br />

are prognostic in breast and colon cancers. Our group demonstrated<br />

that r9AA is associated with shorter time to recurrence in Caucasian(C) and<br />

Hispanic(H) females(F) with GAC. We investigated the significance <strong>of</strong><br />

LMTK3 polymorphism in J PTS with GAC. Methods: Blood or tissue samples<br />

<strong>of</strong> 169 J PTS who had surgery with/without adjuvant chemotherapy (ACT)<br />

were analyzed. Genomic DNA was extracted using the QIAmp kit; all<br />

samples were analyzed using PCR-based direct DNA-sequencing. The<br />

endpoints <strong>of</strong> the study were disease-free survival (DFS) and overall survival<br />

(OS). Kaplan-Meier curves and log-rank test were used for univariate<br />

analysis. Multivariate analysis was performed to test the interaction<br />

between polymorphism and gender adjusting for other variables. Results:<br />

60 F and 109 males were enrolled in this study, 17% stage(s) IB, 31% s II,<br />

36% s III, 17% s IV (AJCC-6). The median age was 67(31-88). 65% <strong>of</strong> PTS<br />

received S-1 based ACT. Median follow-up was 4 years(ys). Prognosis was<br />

worse in men with r9 AA than AG/GG, at 1 year 67% (95% CI 40-83%) with<br />

AA vs 99% (95% CI 91-99%) <strong>of</strong> AG/GG were alive (p� 0.039). Median<br />

survival was not reached in the AG/GG group; in the AA group median DFS<br />

and OS was 1yr (p� 0.03) and 2ys (p� 0.039) respectively. In the<br />

multivariate analysis adjusting for s, age, and ACT, males carrying AA had<br />

increased risk <strong>of</strong> disease recurrence (HR 3.84 95%CI 1.86-7.92, p�<br />

0.001) and dying (HR 3.47 95%CI 1.58-7.62 p�0.002) compared to<br />

those with AG/GG (HR�1, reference). Conclusions: r9 AA was associated<br />

with significantly worse DFS and OS in J male with GAC. These results<br />

confirm our previous findings that LMTK3 is an independent prognostic<br />

factor for localized GAC; interestingly the relationship between gender and<br />

prognostic significance is the opposite in J vs. C/H. The gender disparity<br />

can be due to the differences in the etiology (histological subtypes),<br />

management strategies, allele frequency, and degree <strong>of</strong> estrogen exposure<br />

in the two populations. Additional studies are warranted to identify the<br />

underlying biological mechanism.<br />

4090 General Poster Session (Board #47A), Mon, 8:00 AM-12:00 PM<br />

A prospective trial for defining a subset <strong>of</strong> patients with limited metastatic<br />

gastric cancer who may be candidates for bimodal treatment strategies:<br />

FLOT3. Presenting Author: Salah-Eddin Al-Batran, Krankenhaus Nordwest,<br />

UCT-University Cancer Center, Frankfurt, Germany<br />

Background: The utility <strong>of</strong> surgery for metastatic gastric cancer is debated.<br />

A prospective trial was performed to evaluate a prognostic model for<br />

selecting patients (pts) treated with systemic chemotherapy (ct) who may<br />

also be candidates for surgical intervention. Methods: Using a predefined<br />

algorithm pts with untreated gastric cancer were prospectively stratified<br />

into 3 groups: operable (OD), limited metastatic (LD), or extensive<br />

metastatic (ED) disease and treated with 5-FU, oxaliplatin, leucovorin and<br />

docetaxel (FLOT). LD was defined as: distant intra-abdominal lymph node<br />

metastases only or/and a maximum <strong>of</strong> 1 organ involved, normal serum<br />

alkaline phosphatase, � 5 liver lesions, no visible carcinomatosis (peritoneum<br />

or pleura), and ECOG � 1. All other metastatic pts were ED. Pts with<br />

OD received 4 preoperative ct cycles followed by surgery and 4 postoperative<br />

cycles. Pts with LD received 8 cycles with surgery allowed for complete<br />

macroscopic resection. Pts with ED received 8 cycles with surgery allowed<br />

for palliation only. The study had 80% power to detect a HR <strong>of</strong> 0.55 for<br />

overall survival in favor <strong>of</strong> the LD group (vs. ED group; 2-sided log-rank<br />

p�0.05). Results: 238 <strong>of</strong> 252 pts included were eligible (OD/LD/ED:<br />

51/60/127). LD pts had distant lymph nodes only (41%), liver (22%), lung<br />

(17%), localized peritoneal involvement (7%), or others (13%). A median<br />

<strong>of</strong> 8 ct cycles was applied to all groups. Median OS was 22.9 vs. 10.7<br />

months in pts with LD vs. ED, respectively (HR 0.37; 95% CI, 0.25 – 0.56;<br />

p �0.001). LD was the strongest predictor <strong>of</strong> OS in the multivariate<br />

analysis including all single determinants <strong>of</strong> LD status (p�.002). Surgical<br />

resection was conducted in 96%, 62%, and 12% in the OD, LD, and ED<br />

groups, <strong>of</strong> which R0 resection (primary) was achieved in 82%, 81% and<br />

33%, respectively. Within the LD arm, operated pts had better outcome<br />

than non-operated pts (median OS 31.3 vs. 15.9 months; p�.004) and pts<br />

with lymph node only involvement had best outcome. Conclusions: This<br />

clinical model identifies a subset <strong>of</strong> pts with (limited) metastatic gastric<br />

cancer who have a favorable outcome and who may be candidates for<br />

bi-modal treatment strategies.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


4091 General Poster Session (Board #47B), Mon, 8:00 AM-12:00 PM<br />

Neoadjuvant chemoradiotherapy with 5-fluorouracil-cisplatin combined<br />

with cetuximab in patients with resectable locally advanced esophageal<br />

carcinoma: A prospective phase I/II trial (FFCD-PRODIGE 3)—Preliminary<br />

phase II results. Presenting Author: Laetitia Dahan, APHM La Timone,<br />

Marseille, France<br />

Background: Chemoradiotherapy (CRT) for locally advanced esophageal<br />

cancer is based on 5FU combined with cisplatin. The anti-EGFR antibody<br />

cetuximab increases the efficacy <strong>of</strong> RT-based regimen for patients (pts)<br />

with cancer <strong>of</strong> the head and neck. This phase I/II trial was evaluating MTD,<br />

safety and pathologic complete response (pCR) rate <strong>of</strong> the combination <strong>of</strong><br />

cetuximab with platinum-based CRT in esophageal cancer. Methods:<br />

Inclusion criteria: squamous cell carcinoma or adenocarcinoma <strong>of</strong> the<br />

esophagus, stage II-III, WHO PS 0-1. A radiotherapy <strong>of</strong> 45 grays (1.8<br />

Gy/25F) was given over 5 weeks. During phase I, four patients experienced<br />

limited toxicity to dose level 0, and treatment was desescalated to dose<br />

level -1: weekly cetuximab (400 mg/m2 one week before start <strong>of</strong> radiotherapy<br />

and 250 mg/m2 during radiotherapy), and 5 FU (500 mg/m2 per<br />

day D1-D4) combined with cisplatin (40 mg/m2 D1) on week 1 and 5. Nine<br />

pCR over 27 resections were needed to show a pCR rate�20% (��5%,<br />

power � 90% ). Thirty three patients were included in the phase II. Results:<br />

From 07-2007 to 01-2011, 33 pts were enrolled, 20 squamous cell<br />

carcinoma (60.6%), 13 adenocarcinoma (39.4%), 25 (75.8%) stage III<br />

and 8 (24.2%) stage II. Among 32 pts who received CRT, the main grade<br />

3-4 toxicities were esophagitis (4 pts), leucopenia (1 pt), anaphylaxis<br />

reaction (1 pt), rash (1 pt). Resection was achieved in 27 pts (25 R0)/31<br />

who underwent surgery. Complete or near complete pathologic response<br />

(TRG grades 1 and 2 to Mandard) was achieved respectively in 5 (18.5%)<br />

and 6 (22.2%) pts. There were 4 deaths at 30 days post surgery. Severe<br />

postoperative complications were pulmonary complications (8), anastomotic<br />

stricture (4), gastric necrosis (1) and infection (7). The median<br />

overall survival was 15.7 months [11.01-.], and the median relapse free<br />

survival was 13.7 months [5.47-.]. Conclusions: Adding cetuximab to<br />

preoperative chemoradiotherapy including 5FU and cisplatin did not<br />

increase pCR. The recommended dose level <strong>of</strong> chemotherapy determined<br />

during phase I could explain those disappointing results. We won’t initiate a<br />

phase III trial.<br />

4093 General Poster Session (Board #47D), Mon, 8:00 AM-12:00 PM<br />

Randomized phase II study <strong>of</strong> preoperative concurrent chemoradiotherapy<br />

with or without induction chemotherapy with S-1 and oxaliplatin in patients<br />

with resectable esophageal cancer. Presenting Author: Dok Hyun Yoon,<br />

Department <strong>of</strong> Oncology, Asan Medical Center, University <strong>of</strong> Ulsan College<br />

<strong>of</strong> Medicine, Seoul, South Korea<br />

Background: The value <strong>of</strong> adding induction chemotherapy (ICT) to preoperative<br />

chemoradiotherapy followed by surgery has not been delineated well.<br />

Methods: Patients with stage II, III or IVA (by AJCC 6th ed.) esophageal<br />

cancer were randomly allocated to either 2 cycles <strong>of</strong> ICT (oxaliplatin 130<br />

mg/m2 on day 1 and S-1 at 40 mg/m2 bid on days 1-14, every 3 weeks),<br />

followed by concurrent chemoradiotherapy (CCRT) (46 Gy, 2 Gy/day with<br />

oxaliplatin 130 mg/m2 on day 1 and 21 and S-1 30 mg/m2 bid, 5 days/week<br />

during radiotherapy) and surgery (arm A, n�48), or the same chemoradiotherapy<br />

followed by surgery without ICT (arm B, n�49). Primary outcome<br />

was to compare pathologic complete response (pCR). Results: Thirty six and<br />

35 patients underwent surgery with or without ICT, respectively. pCR rate<br />

among those who underwent surgery was significantly lower in arm A<br />

(30.6% vs. 54.3%, p�0.043). However, no difference in progression-free<br />

survival (PFS) and overall survival (OS) was observed with a median<br />

follow-up <strong>of</strong> 19.5 mo (95% CI, 19.1-22.4). Two-year PFS rate was 63.8%<br />

in arm A and 55.2% in arm B (p�0.626) and 2-year OS rate was 70.1%<br />

and 62.6%, respectively (p�0.515). While 47 (arm A) and 48 (arm B)<br />

patients received at least 44 Gy <strong>of</strong> radiotherapy, relative dose intensity<br />

(RDI) for oxaliplatin during CCRT was significantly lower in arm A vs. arm B<br />

(92.7% � 19.6% vs. 99.7 � 1.8%, p�0.017). RDI for S1 did not<br />

significantly differ (94.1% � 17.3% vs. 98.5% � 5.9%, p�0.095). G3/4<br />

thrombocytopenia was significantly common in arm A (37.5% vs. 4.1%, p<br />

�0.001), which contributed to lower RDI <strong>of</strong> oxaliplatin. Three patients in<br />

arm A, compared to none in arm B, failed to survive for 90 days after<br />

surgery. Conclusions: Adding this ICT to preoperative chemoradiotherapy<br />

seems to cause lower pCR rate and higher toxicity during CCRT.<br />

Gastrointestinal (Noncolorectal) Cancer<br />

261s<br />

4092 General Poster Session (Board #47C), Mon, 8:00 AM-12:00 PM<br />

Positive association <strong>of</strong> gastric cancer surgery outcome with surgeon<br />

specialization in a Shanghai high-volume general hospital. Presenting<br />

Author: Zhenbin Shen, Department <strong>of</strong> General Surgery, Zhongshan Hospital,<br />

Fudan University, Shanghai, China<br />

Background: Numerous studies suggest positive relationship between<br />

hospital volume and cancer treatment outcomes, the surgeon’s experience<br />

and specialty training may also be important. This was examined in a high<br />

volume hospital in Shanghai among patients who underwent gastric cancer<br />

(GC) surgery. Methods: Data on consecutive patients (pts) undergoing R0 or<br />

R1 GC resection in Zhongshan hospital between January 2003 and June<br />

2010 were collected and analyzed. Follow-up on pts who were non-<br />

Shanghai residents were less complete therefore excluded. Post-operative<br />

mortality, pathologic results and survival outcome for pts treated by<br />

surgical training, i.e., sub-specialized vs., non-specialized, were obtained.<br />

Survival was calculated by the Kaplan-Meier method and Log-rank test was<br />

used to determine statistical significance. To determine whether subspecialty<br />

surgical training was an independent factor for overall survival<br />

(OS), univariate and multivariate analyses were performed using Cox<br />

proportional hazards regression. Results: Total 5,046 pts underwent R0 or<br />

R1 GC resection were identified.1594 pts had complete covariate data,<br />

survival information and were included in the study. Of them, the<br />

sub-specialized group included 217 cases treated by 3 surgeons, while the<br />

non-specialized group included 1377 cases treated by 52 surgeons. 5-year<br />

cumulative OS was higher in the sub-specialized group (62.9% vs. 54.6%,<br />

p�0.032). Multivariate analysis showed that tumor stage(p�0.001),<br />

location <strong>of</strong> tumor (p�0.003), vascular invasion (p�0.001) and surgeon<br />

(HR�1.54, p�0.001) were all associated with OS. The incidence <strong>of</strong><br />

positive margin was higher in non-specialized group (2.0% vs. 2.7%,<br />

p�0.001) and the probability <strong>of</strong> retrieved lymph nodes less than 15 was<br />

more in non-specialized group (25.9% vs. 7.3%, p�0.001). Postoperative<br />

mortality was also higher in non-specialized group than in specialized<br />

group(1.5% vs. 0.9%, p�0.001). Conclusions: In high volume general<br />

hospital, sub-specialty training is desirable in gastric cancer surgery, the<br />

quality <strong>of</strong> gastric cancer surgery can be further improved by sub-specialty<br />

training leading to better treatment outcome.<br />

4094 General Poster Session (Board #47E), Mon, 8:00 AM-12:00 PM<br />

Multicenter phase II study combining panitumumab with chemoradiation<br />

followed by surgery for patients with operable esophageal cancer (PACTstudy).<br />

Presenting Author: Sil Kordes, Academic Medical Center, Amsterdam,<br />

Netherlands<br />

Background: A consistent finding in many studies in patients with operable<br />

esophageal and gastro-esophageal junction cancer is that response to<br />

preoperative therapy, particularly the absence <strong>of</strong> residual disease in the<br />

surgical specimen, is an indicator <strong>of</strong> better disease-free and overall<br />

survival. One <strong>of</strong> the potential strategies to improve these local effects <strong>of</strong><br />

preoperative chemoradiation (CRT) is the concurrent inhibition <strong>of</strong> the<br />

epidermal growth factor receptor (EGFR). Therefore in our trial we evaluated<br />

the pathologic response <strong>of</strong> panitumumab in combination with neoadjuvant<br />

CRT as first line treatment <strong>of</strong> operable adenocarcinomas or squamous<br />

cell carcinomas <strong>of</strong> the esophagus. Methods: Patients with resectable<br />

cT1N1M0 or cT2-3N0-2M0 tumors received preoperative CRT consisting<br />

<strong>of</strong> 3 administrations <strong>of</strong> panitumumab (6mg/kg) in weeks 1-3-5, weekly<br />

administrations <strong>of</strong> carboplatin (AUC � 2) and paclitaxel (50 mg·m2 ) for 5<br />

weeks and concurrent radiotherapy (41.4 Gy in 23 fractions, 5 days per<br />

week) followed by surgery. The primary endpoint was the percentage <strong>of</strong><br />

pathologic complete responses (pCR). Results: From January 2010 till<br />

December 2011, 91 patients were enrolled. 3 patients were not resected<br />

due to the development <strong>of</strong> metastatic disease during CRT. Results <strong>of</strong> all<br />

patients will be presented at ASCO. The majority <strong>of</strong> the 74 patients<br />

operated thus far had T3 (85%) and node positive (77%) tumors. 21.6%<br />

reached pCR and 13.5% had less than 10% viable tumor cells in the<br />

resected specimen. R0-resection was achieved in 98.6%. There were 58<br />

patients with adenocarcinoma and 11 patients with squamous cell carcinoma.<br />

The pCR <strong>of</strong> these patients were 15.5% and 45.5%, respectively.<br />

Main grade 3 toxicities were esophagitis, fatigue, rash and anorexia. 1<br />

postoperative death occurred due to ischemia <strong>of</strong> the esophageal reconstruction.<br />

Conclusions: The addition <strong>of</strong> panitumumab to chemoradiotherapy with<br />

carboplatin and paclitaxel was feasible without increasing postoperative<br />

mortality, but did not improve the rate <strong>of</strong> pathologic complete responses<br />

compared to historical data.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


262s Gastrointestinal (Noncolorectal) Cancer<br />

4095^ General Poster Session (Board #47F), Mon, 8:00 AM-12:00 PM<br />

Two-year follow-up <strong>of</strong> a phase II study on catumaxomab as part <strong>of</strong> a<br />

multimodal approach in primarily resectable gastric cancer. Presenting<br />

Author: Carsten Bokemeyer, Department <strong>of</strong> Oncology/Hematology, University<br />

Hospital Hamburg, Hamburg, Germany<br />

Background: Perioperative chemotherapy (CT) has demonstrated as survival<br />

benefit in locally advanced gastric cancer (GC) in randomized trials.<br />

However, the overall cure rate is 30-40% and a significant number <strong>of</strong><br />

patients are not able to receive the postoperative part <strong>of</strong> their CT regimen.<br />

In Europe, the trifunctional antibody catumaxomab is approved for the<br />

treatment <strong>of</strong> malignant ascites based on a pivotal trial which also included<br />

GC patients. A new multimodal approach combining neoadjuvant CT,<br />

followed by gastrectomy and intraperitoneal (i.p.) immunotherapy with<br />

catumaxomab was assessed in a single-arm multicenter phase II study. We<br />

here report 2-year follow-up data. Methods: GC pts (T2/T3/T4, N�/–, M0)<br />

received 3 cycles <strong>of</strong> neoadjuvant fluoropyrimidin/platinum-based CT followed<br />

by ´en-bloc´ R0-gastrectomy. Catumaxomab was administered i.p. as<br />

intraoperative bolus (10 �g) followed by 4 consecutive 3-hour infusions <strong>of</strong><br />

10-150 �g. Primary safety endpoint was the rate <strong>of</strong> predefined postoperative<br />

complications observed during 30 days after surgery. Key efficacy<br />

endpoints included disease-free (DFS) and overall survival (OS). Results:<br />

The original study data presented at the WCGC in 2011 (Schuhmacher et<br />

al.,Ann Oncol (2011) 22(suppl. 5)) showed that the primary endpoint was<br />

met and the described application regimen is safe. At time <strong>of</strong> surgery,<br />

27.8% <strong>of</strong> patients were stage I, 27,8% <strong>of</strong> patients were stage II, 22,3% <strong>of</strong><br />

patients were stage III and 14,8% <strong>of</strong> patients were stage IV as assessed<br />

according to pTNM measures. At 24 months 39/54 (safety analysis set)<br />

patients were still alife,14/54 were dead, (one patient lost to follow-up),<br />

24/37 had no progression, only 13/37 patients relapsed (for 2 patients<br />

disease status was not recorded). At the 2 year cut <strong>of</strong>f DFS was 56.4%<br />

(95% CI: 41–69%), OS was 75% (95% CI: 60–85%). Conclusions:<br />

Catumaxomab as part <strong>of</strong> a multimodal therapy in primarily resectable GC is<br />

a feasible option. The 2-year follow up efficacy results show promising data<br />

for DFS and OS in a cohort <strong>of</strong> locally advanced gastric cancer pts.<br />

4097 General Poster Session (Board #47H), Mon, 8:00 AM-12:00 PM<br />

Sequential chemotherapy with dose-dense docetaxel, cisplatin, folinic acid<br />

and 5-fluorouracil (TCF-dd) followed by oxaliplatin, folinic acid, 5-fluorouracil,<br />

and irinotecan (COFFI) in locally advanced or metastatic gastric<br />

cancer (MGC). Presenting Author: Gianluca Tomasello, Istituti Ospitalieri,<br />

Cremona, Italy<br />

Background: MGC is a chemosensitive tumour. According to the Norton-<br />

Simon hypothesis, a reduction <strong>of</strong> tumor burden could best be achieved by<br />

shortening the interval between the cycles, and the resistance might be<br />

overcome by switching from initial chemotherapy to a new, non crossresistant<br />

one. We previously reported on the high efficacy <strong>of</strong> a new strategy<br />

using 2 sequential, non cross-resistant chemotherapy regimens in MGC<br />

(Cancer Chemother Pharmacol 11 Jan; 67 (1): 41-8). Aim <strong>of</strong> this study is to<br />

evaluate this therapeutic approach in a larger case series. Methods: 43<br />

patients (pts) treated at our centre from November 04 to April 11 were<br />

included. All pts were chemo-naïve and received 4 cycles <strong>of</strong> TCF-dd (see<br />

reference above for doses) every 2 weeks. Subsequently and irrespective <strong>of</strong><br />

their response, they received 4 cycles <strong>of</strong> COFFI (see reference above ) every<br />

2 weeks. In both regimens pegfilgrastim 6 mg sc on day 3 was included.<br />

Results: Median age: 63; 74% male. PS 0-1. After the first regimen<br />

(TCF-dd) 43 pts were evaluable for response.We registered 3 CR, 27 PR, 7<br />

SD, 5 PD and 1 not evaluable for an ORR at ITT <strong>of</strong> 70% (95% CI, 58-85).<br />

All pts proceeded to the second regimen (COFFI) and 40 pts were evaluable<br />

for response. The 3 CR observed after TCF-dd were maintained. Among the<br />

27 pts with PR after TCF-dd, 2 achieved CR, 14 improved the response, 6<br />

maintained PR, 3 progressed. Among the 7 pts with SD after TCF-dd, 1<br />

achieved CR, 3 achieved PR, 2 progressed and 1 is not valuable. Among the<br />

5 pts with PD after TCF-dd, 1 achieved CR, 4 achieved RP. After COFFI we<br />

observed 5 CR, 21 PR, 9 SD and 5 PD. The ORR in the 40 pts was 60%<br />

(95% CI, 50-80). Considering both regimen ORR was 93%. mTTP was<br />

12.1 months (95% CI, 8,1-16,2). mOS was 16.1 months (95% CI,<br />

12.7-21.6). At Dec ’11, 4 out <strong>of</strong> 8 pts who achieved CR are alive and<br />

disease free. Most frequent G3-4 toxicities were: astenia (32%), neutropenia<br />

(35%), anemia (14%), neurotoxicity (21%). Conclusions: The study<br />

confirms that a sequential dose-dense strategy using TCF-dd followed by<br />

COFFI is feasible and highly effective and deserves to be tested in a<br />

randomized study which is on going.<br />

4096 General Poster Session (Board #47G), Mon, 8:00 AM-12:00 PM<br />

A phase II study <strong>of</strong> capecitabine-oxaliplatin and cetuximab in patients<br />

(pts) with advanced/metastatic gastric cancer (G) or gastroesophageal<br />

junction (GEJ) adenocarcinoma. Presenting Author: Syma Iqbal, University<br />

<strong>of</strong> Southern California Norris Comprehensive Cancer Center, Los Angeles,<br />

CA<br />

Background: There have been limited advances in the treatment (tx) <strong>of</strong> pts<br />

with metastatic G or GEJ adenocarcinoma. The combination <strong>of</strong> fluroropyrimidines<br />

and platinums are standard regimens for this disease. The epidermal<br />

growth factor receptor pathway has been identified as a target and KRAS<br />

mutations are rare. Methods: The primary objective was to assess the<br />

proportion <strong>of</strong> pts with progression within 4 months. Secondary objectives<br />

include response (RR), progression free survival (PFS) overall survival (OS),<br />

toxicity and molecular correlates. A two-stage Simon phase II design was<br />

used to distinguish between a 30% vs. 50% 4 month PFS; with analysis<br />

planned at 27 and 53 pts. Eligible pts were chemonaive, metastatic/<br />

unresectable with measurable disease. Pts received capecitabine 850<br />

mg/m2 BID D1-14, oxaliplatin 130 mg/m2 IV D1, cetuximab 400 mg/m2<br />

(load) then 250 mg/m2 IV D1, 8, 15, q21 days. Blood and tissue were<br />

analyzed for genes in the DNA repair and EGFR pathways. Results: 61 pts<br />

were accrued, 2 GEJ and 59 G; 42 male, 19 female; median age 56 years<br />

(28-86); 54 pts were evaluable. The 4 month PFS rate 61% (95%CI<br />

47-73%), PFS 5.4 months (95% CI 3.5-6.7); RR 50% (95%CI: 36-64%),<br />

1 (1.9%) complete, 26 (48.1%) partial, 22 (40.7%) stable disease; 2<br />

(3.7%) pts became resectable; median OS 14.9 months (95% CI 8.8-<br />

22.2) with median follow-up <strong>of</strong> 21.8 months. 65% (39/60) <strong>of</strong> pts had<br />

grade 3 (36 pts) or 4 (3 pts) toxicity; most common, anorexia (13.3%),<br />

diarrhea (13.3%), fatigue (13.3%), nausea (15%), vomiting (15%); grade<br />

3 hand/foot (5.3%), acneiform rash (3.3%). SNP’s in LRIG4, (regulator <strong>of</strong><br />

EGF signaling) and PAI1 (tumor angiogenesis, cell proliferation, migration<br />

and adhesion) were significantly associated with RR (67% G/G vs 17% A/A<br />

,p�0.044) and PFS (5.8 months 5G/4G vs 3.4 months 5G/5G, p�0.017)<br />

respectively. Conclusions: XELOX and cetuximab has promising activity,<br />

comparable to existing regimens. Novel SNP’s were identified correlating<br />

with RR and PFS which may allow for optimization <strong>of</strong> pt selection.<br />

4098 General Poster Session (Board #48A), Mon, 8:00 AM-12:00 PM<br />

Lenalidomide for second-line treatment <strong>of</strong> advanced hepatocellular cancer<br />

(HCC): A Brown University Oncology Group phase II study. Presenting<br />

Author: Howard Safran, Brown University Oncology Group, Providence, RI<br />

Background: Lenalidomide inhibits fibroblast growth factor (FGF), vascular<br />

endothelial growth factor (VEGF) and multiple tumor growth pathways.<br />

There is no standard <strong>of</strong> care for patients who progress after sorafenib.<br />

Therefore, we performed a phase II study to determine the activity <strong>of</strong><br />

lenalidomide in second-line HCC therapy. Methods: Patients with advanced<br />

HCC who progressed on or were intolerant to sorafenib were eligible. Prior<br />

chemoembolization, RFA, or surgery were allowed. Eligibility criteria also<br />

included bilirubin �4 mg/dL, AST and ALT �5 times upper limit <strong>of</strong> normal,<br />

ECOG performance status 0-2, platelet count �60,000/mm3 , absolute<br />

neutrophil count �1000/mm3 , and creatinine �2mg/dL. Patients were<br />

treated with lenalidomide 25mg orally days 1-21 <strong>of</strong> a 28 day cycle until<br />

disease progression or unacceptable toxicities. The planned original<br />

sample size was 25 patients but when early activity was demonstrated the<br />

study was expanded to 40 patients. Results: The study has completed<br />

accrual <strong>of</strong> 40 patients. The median age was 60.5 years (17-88 years).<br />

Nineteen patients were Child-Pugh A, 16 patients were B, and 5 patients<br />

were C. Twenty four patients had extrahepatic disease. Preliminary data is<br />

available on the first 37 patients. One patient had grade 4 neutropenia.<br />

Grade 3 toxicities included ANC (n�2), fatigue (n�4), rash (n�2), arthritis<br />

(n�1), diarrhea (n�1), dehydration (n�2). One patient developed variceal<br />

bleeding which precipitated encephalopathy and death. Of the 32 patients<br />

with elevated baseline AFP, nine (28%) had a �50% reduction including<br />

one patient with a reduction in AFP from 56,900ng/ml to 5 ng/ml. Six <strong>of</strong><br />

the first 37 patients (16%) had a radiographic partial response. Two<br />

patients achieved a complete response and have not progressed at 36 and<br />

32 months. Conclusions: Lenalidomide can be administered to patients<br />

with advanced HCC and significant hepatic dysfunction. Promising, and in<br />

a small percentage <strong>of</strong> patients, dramatic and durable activity has been<br />

demonstrated. Investigations are underway to explore the mechanism <strong>of</strong><br />

action <strong>of</strong> lenalidomide in HCC.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


4099 General Poster Session (Board #48B), Mon, 8:00 AM-12:00 PM<br />

A phase II trial <strong>of</strong> temsirolimus (TEM) and bevacizumab (BEV) in patients<br />

with advanced hepatocellular carcinoma (HCC). Presenting Author: Jennifer<br />

J. Knox, Princess Margaret Hospital, Toronto, ON, Canada<br />

Background: There is strong rationale to combine an m-TOR inhibitor (TEM)<br />

with a VEGF inhibitor (BEV) as a potentially active and well tolerated<br />

treatment for HCC. Both agents have shown modest single agent activity in<br />

HCC and so evaluated here in a phase II trial. Methods: A modified 2-stage<br />

Simon design planned 25 or 50 patients (pts) to test the null hypothesis<br />

that true tumor response rate is at most 10% andtrue 6-mo progressionfree<br />

survival rate (PFS) (by RECIST) is at most 65%, or no better than single<br />

agent BEV (6 mo PR �2 pts or PFS 6 mo �18 out <strong>of</strong> 25.) Toxicity, TTP,<br />

PFS and survival were 2nd endpoints. Eligible pts had confirmed HCC with<br />

disease unresectable or amenable to other localised therapies, Child Pugh<br />

A liver status and no prior systemic therapy involving the VEGF or m-TOR<br />

class <strong>of</strong> agents. TEM was administered at starting dose 25 mg IV<br />

d1,8,15,22 with BEV at 10mg/kg IV d 1, 15, all q 28 days (1 cycle).<br />

Imaging was q 8 wks. Results: From 09/09 to 09/11, 27 eligible pts were<br />

enrolled with 25 evaluable for toxicity and efficacy. Med age 59 yrs, 85%<br />

male, PS 0/1: 35/65, 58% metastatic, �85% BCLC stage C. With med 6<br />

cycles (range 1-14) delivered, most pts (88%) experienced a grade 3�<br />

adverse event (a/e.) Common grade 3 a/es related to treatment included<br />

thrombocytopenia (40%), neutropenia (20%), leucopenia (12%), fatigue<br />

(8%), anemia, mucositis, dyspnea, diarrhea, bleeds, fistula, infections (4%<br />

each). There was one possible treatment related death. Per protocol dose<br />

reductions/discontinuation for TEM-related a/es were most common. There<br />

were 2 confirmed PRs and 16 pts progression-free by 6 mos. A third pt<br />

developed a late PR at cycle 13. Median TTP on study was 6 mos, median<br />

PFS was 7.4 mos and median survival was 8.3 mos, with 13 pts still alive.<br />

Accrual closed at end <strong>of</strong> stage 1 as neither the number <strong>of</strong> responses nor the<br />

PFS at 6 mos passed the futility stopping rule set for this combination.<br />

Conclusions: This multicenter study is the first HCC trial evaluating the<br />

BEV/TEM doublet. Despite manageable toxicity, the ORR and 6 mo PFS did<br />

not surpass assumptions based on single agent BEV in HCC. Further study<br />

<strong>of</strong> BEV/TEM combination in this advanced HCC population is not recommended.<br />

4101 General Poster Session (Board #48D), Mon, 8:00 AM-12:00 PM<br />

Serum folate, LINE-1 hypomethylation, and overall survival in a prospective<br />

cohort <strong>of</strong> hepatocellular carcinoma patients. Presenting Author: Abby<br />

B. Siegel, Columbia University, New York, NY<br />

Background: Folate plays a central role in DNA methylation reactions, but<br />

the relationship between folate and methylation status has not been<br />

studied in HCC patients. We hypothesized that low folate levels would<br />

correlate with global hypomethylation, and worsened survival in HCC<br />

patients. Methods: 72 newly-diagnosed HCC patients were enrolled in a<br />

prospective study, beginning 10/08, and followed until 9/11. We excluded<br />

those with previous malignancy, and uncompensated Child Pugh (CP) B or<br />

C disease. We used pyrosequencing to evaluate quartiles <strong>of</strong> LINE-1<br />

methylation in plasma as a surrogate for global tumor hypomethylation. We<br />

evaluated the relationship between folate deficiency (serum folate levels �<br />

6ng/mL), DNA hypomethylation (LINE-1 methylation � 25th percentile<br />

i.e., � 69.4%), and overall survival in our cohort. Results: Median age was<br />

60; 82% were men and 67% had hepatitis C. Mean serum folate levels<br />

were 12.1 ng/mL (SD � 5.3 ng/mL), while mean % LINE-1 methylation<br />

was 69.9 (SD � 4.6). In a univariate analysis, folate deficiency and LINE-1<br />

hypomethylation were significant predictors <strong>of</strong> poor overall survival (HR �<br />

3.77; 95% CI: 1.37, 10.41, and HR�3.50; 95% CI: 1.51, 8.13,<br />

respectively). These markers remained independent predictors <strong>of</strong> survival<br />

in a multivariate model including age, CP class, AJCC stage and AFP. HR<br />

for folate deficiency in this model was 3.52 (95% CI: 1.16, 10.70), and HR<br />

for LINE-1 hypomethylation was 2.74 (95% CI: 1.10, 6.85). We also found<br />

a significant association between folate deficiency and LINE-1 hypomethylation<br />

(p � 0.04). Conclusions: We provide novel data that folate deficiency<br />

is an independent predictor <strong>of</strong> worsened overall survival in patients with<br />

HCC in a multivariate model. Further, we show that global changes in DNA<br />

methylation assesed in plasma correlate with folate levels, supporting a<br />

possible mechanistic link between the two. Folate supplementation, and<br />

the use <strong>of</strong> other agents which modulate methylation, deserve further study<br />

in HCC.<br />

Gastrointestinal (Noncolorectal) Cancer<br />

263s<br />

4100^ General Poster Session (Board #48C), Mon, 8:00 AM-12:00 PM<br />

A phase I/II study <strong>of</strong> AZD6244 in combination with sorafenib in advanced<br />

hepatocellular carcinoma. Presenting Author: Su Pin Choo, National<br />

Cancer Center Singapore, Singapore<br />

Background: Preclinical studies have shown that pharmacological inhibition<br />

<strong>of</strong> the MEK/ERK pathway by AZD6244 enhanced the anti-tumor effect<br />

<strong>of</strong> sorafenib in both orthotopic and ectopic models <strong>of</strong> HCC. We conducted<br />

this study to determine tolerability, pharmacokinetics, and pharmacodynamics<br />

<strong>of</strong> AZD6244 when combined with sorafenib in advanced HCC. Methods:<br />

Patients with biopsy-proven unresectable BCLC B/C hepatocellular carcinoma<br />

were recruited. Only those with Childs-Pugh A or B (7) liver cirrhosis<br />

and without prior systemic therapy were included. Sorafenib at 400mg bd<br />

was given 1 week before initiation <strong>of</strong> AZD6244 which was escalated in<br />

subsequent cohorts from 75mg om based on 3�3 design. PK and PD<br />

studies and QOL assessments were performed. DCE-MRI imaging was<br />

performed to assess tumor vascularity in response to treatment. Results:<br />

Fourteen patients were recruited (including 2 replaced). 11 had evaluable<br />

disease. Characteristics: all male, all Chinese, 12 were BCLC C stage. Two<br />

DLTs were seen out <strong>of</strong> 6 patients at dose level 1 (AZD6244 at 50mg bd)<br />

which were grade 3 fatigue and grade 3 abdominal pain with elevated<br />

transaminases. When an additional dose level 1A was added (ADZ6244 at<br />

100mg om), 2 out <strong>of</strong> 3 patients had DLTs <strong>of</strong> grade 3 raised aspartate<br />

transaminase and grade 3 diarrhea. Thus, the MTD was determined to be<br />

AZD6244 at 75mg om when combined with sorafenib 400mg bd. Common<br />

toxicities were diarrhea (83%), rash (58%), fatigue (50%), hypertension<br />

(42%), anorexia/vomiting/thrombocytopenia (25%). Two patients had reversible<br />

LVEF dysfunction and there were no eye toxicities. PK <strong>of</strong> AZD6244<br />

showed oral clearance <strong>of</strong> 11.2 � 6.8 L/h and terminal half-life <strong>of</strong> 6.0 �2.0<br />

h.Objective responses were 3 PR, 6 SD and 2 PD.DCE-MRI measurements<br />

demonstrated significant reductions in permeability surface area product<br />

(PS, ml/100ml/min) and fractional intravascular blood volume (v1, ml/<br />

100ml) seven days after starting sorafenib. No additional antivascular<br />

activity was observed when AZD6244 was added to sorafenib. Conclusions:<br />

The recommended phase II dose for AZD6244 is 75mg om when combined<br />

with sorafenib 400mg bd for advanced HCC patients. This combination is<br />

feasible, shows activity and warrants further investigation.<br />

4102 General Poster Session (Board #48E), Mon, 8:00 AM-12:00 PM<br />

Phase I trial <strong>of</strong> temsirolimus (TEM) plus sorafenib (SOR) in advanced<br />

hepatocellular carcinoma (HCC) with pharmacokinetic (PK) and biomarker<br />

correlates. Presenting Author: Robin Katie Kelley, Helen Diller Family<br />

Comprehensive Cancer Center, University <strong>of</strong> California, San Francisco, San<br />

Francisco, CA<br />

Background: Mammalian target <strong>of</strong> rapamycin inhibitors added to SOR<br />

augment antitumor effect in HCC models. We developed a phase 1 trial to<br />

determine maximum tolerated dose (MTD) and recommended phase 2 dose<br />

(RP2D) <strong>of</strong> TEM plus SOR in HCC patients (pts). The study was approved<br />

and funded by the National Comprehensive Cancer Network (NCCN) from<br />

general research support from Pfizer, Inc., and conducted at 2 NCCN<br />

centers. Methods: Eligibility: �1 measurable site. No prior systemic therapy<br />

(Tx). ECOG �2, Child Pugh �7, bilirubin �2 mg/dL, platelets (PLT)<br />

�75,000/mcL. Design: 3�3 escalation. Dose-limiting toxicity (DLT) window<br />

28 days. MTD expansion cohort <strong>of</strong> 9 pts for PK and biomarkers.<br />

Endpoints: 1°: MTD, RP2D. 2°: Safety, toxicity, PK for TEM. Exploratory:<br />

Tumor necrosis, alpha fetoprotein (AFP)-L3, des-�-carboxyprothrombin,<br />

and circulating tumor cells (CTC) by slide-based assay. Results: 21 pts<br />

enrolled. Median age: 60 (47-77). Male/Female: 15/6. Etiology: HCV 9<br />

(43%), HBV 4 (19%), HBV�HCV 2 (10%), ETOH 2 (10%), unknown 4<br />

(19%). Toxicity: DL1: 1 DLT Grade (Gr) 3 PLT. All pts required reductions<br />

for adverse events (AE); de-escalated to DL-1 for intolerability. DL-1: 1 DLT<br />

Gr3 hand-foot syndrome (HFS). Most common related �Gr 3 AE: HypoPO4<br />

(52%); PLT (24%); transaminitis (19%); diarrhea, fatigue, HFS (10%<br />

each). Possibly related serious AE (SAE): Gr4 tumor rupture, Gr4 urosepsis,<br />

Gr3 dental infection with Gr2 ANC, Gr2 pneumonia (1 pt, 5% each). Best<br />

response: Confirmed partial response (PR) 2/21 (10%), stable disease (SD)<br />

11/21 (52%), progression 1/21 (5%), 7/21 (33%) not evaluable. Time on<br />

study: Range �1 to 19�months; median 3� months for pts who<br />

completed �1 cycle (16/21). 16/21 (76%) had baseline elevated AFP<br />

�20; 8/16 (50%) had �50% decline. CTC were detected in 5/5 <strong>of</strong> tested<br />

samples. Decreased tumor enhancement on Tx was seen. Conclusions:<br />

DL-1 is MTD and RP2D, lower than a prior trial in pts without HCC;<br />

tolerability may be impacted by cirrhosis. Encouraging durable radiographic<br />

and AFP responses occurred.<br />

Dose level (DL) TEM (mg IV/week) SOR (mg PO) Accrual to date<br />

-2 10 200 QD 0<br />

-1 (RP2D) 10 200 BID 9 � 5 expansion<br />

1 (starting) 15 200 BID 7<br />

2 15 400 BID 0<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


264s Gastrointestinal (Noncolorectal) Cancer<br />

4103 General Poster Session (Board #48F), Mon, 8:00 AM-12:00 PM<br />

A phase II trial <strong>of</strong> MEK inhibitor BAY 86-9766 in combination with<br />

sorafenib as first-line systemic treatment for patients with unresectable<br />

hepatocellular carcinoma (HCC). Presenting Author: Ho Yeong Lim, Division<br />

<strong>of</strong> Hematology-Oncology, Department <strong>of</strong> Medicine, Samsung Medical<br />

Center, Sungkyunkwan University School <strong>of</strong> Medicine, Seoul, South Korea<br />

Background: Sorafenib (S) is the only approved systemic treatment for<br />

unresectable HCC. Nevertheless, there remains an unmet medical need for<br />

more effective treatment options for this disease. BAY 86-9766 (B) is an<br />

oral, allosteric inhibitor <strong>of</strong> MEK, a key component <strong>of</strong> the MAP kinase<br />

pathway. This study evaluated the efficacy and safety <strong>of</strong> a combination<br />

therapy with B plus S in patients (pts) with HCC. Methods: This is a single<br />

arm, open-label, phase 2 study. Eligible were pts with unresectable HCC,<br />

Child-Pugh Class A, performance status (PS) 0-1, and no prior systemic<br />

anticancer therapy for HCC. Pts started Cycle 1 (21 days) with B 50 mg bid<br />

orally plus S 600 mg daily (200 mg AM, 400 mg PM) orally. If there was no<br />

hand-foot skin reaction, fatigue, or gastrointestinal toxicity � grade 2, S<br />

was escalated to 400 mg bid from Cycle 2 on. Treatment continued until<br />

progression or withdrawal criteria were met. Tumor assessment was<br />

performed every 6 weeks during treatment. Safety was evaluated every<br />

week for the first 6 weeks and every 3 weeks thereafter. Results: Seventy pts<br />

from Asia started study treatment. Pts were predominantly male (86%);<br />

median age was 56 years; 54% had PS <strong>of</strong> 0 and 46% PS <strong>of</strong> 1. The vast<br />

majority had liver cirrhosis (83%) and infection with HBV (76%) or HCV<br />

(17%). Sixty-five were evaluable for efficacy per protocol. Three pts (5%)<br />

had confirmed partial response and 25 pts (38%) had prolonged stable<br />

disease (�10 weeks), with a disease control rate <strong>of</strong> 43%. Median<br />

time-to-progression was 4.1 months. Survival data are not mature, yet. The<br />

most frequent drug-related adverse events (AEs) were rash (60%), diarrhea<br />

(59%), AST elevation (43%), vomiting (30%), nausea (29%), ALT elevation<br />

(26%), and anorexia (26%). There were 4 Grade 5 related AEs (hepatic<br />

failure, sepsis/hepatic encephalopathy, tumor lysis syndrome, and unknown<br />

cause, respectively). Dose modifications due to AEs were necessary<br />

in almost all pts. The median daily dose was 64.2 mg for B and 443.3 mg<br />

for S, respectively. Conclusions: B in combination with S showed antitumor<br />

activity in pts with HCC. However, frequent dose modifications due to AEs<br />

might have limited the treatment effect <strong>of</strong> this combination.<br />

4105 General Poster Session (Board #48H), Mon, 8:00 AM-12:00 PM<br />

Randomized phase II study <strong>of</strong> the x-linked inhibitor <strong>of</strong> apoptosis (XIAP)<br />

antisense AEG35156 in combination with sorafenib in patients with<br />

advanced hepatocellular carcinoma (HCC). Presenting Author: Ann Sing<br />

Lee, Tuen Mun Hospital, Tuen Mun, Hong Kong<br />

Background: XIAP inhibits caspases which are proteases responsible for<br />

apoptotic cell death. It is highly expressed in HCC. AEG35156 is a second<br />

generation antisense oligonucleotide targeting XIAP mRNA, thus lowers the<br />

apoptotic threshold <strong>of</strong> cancer cells. It also accumulates in the liver. This<br />

study is designed to assess the added benefit <strong>of</strong> combining AEG35156 with<br />

sorafenib. Methods: Patients with histologically or clinically diagnosed<br />

(AASLD criteria) HCC who had failed or were unsuitable for resection or<br />

ablative therapies were randomized (2:1) to receive either weekly injection<br />

<strong>of</strong> AEG35156 300mg in combination with sorefanib 400mg BID or<br />

sorefanib alone. The primary end point was progression-free survival (PFS).<br />

Other endpoints were overall survival (OS), response rates and safety.<br />

Results: 51 patients were recruited. 48 patents were evaluable. There were<br />

31 patients in the combination arm and 17 in the control arm. The median<br />

age was 60. 88% <strong>of</strong> patients were male. 81% <strong>of</strong> patients were hepatitis B<br />

carrier. 90% <strong>of</strong> patients belong to Child-Pugh class A. The median<br />

follow-up was 16.2 months. The PFS for the combination arm was 4.0<br />

months (95% CI: 1.2-4.1) and 2.6 months for control arm. The OS for the<br />

combination arm was 6.5 months (95% CI: 3.9-11.5) and 5.4 months for<br />

the control arm. There were 3 partial responders (Choi’s criteria) in the<br />

combination arm (10%, 95% CI: 3-27%) and none (0%) in the control<br />

arm. Patients who had the study treatment interrupted (PFS 4.0, 95% CI:<br />

2.4-5.4) or had dose modification (PFS 4.45, 95% CI: 1.0-6.5) according<br />

to protocol did significantly better than those who had no dose reduction<br />

(PFS 1.2, 95% CI: 1.2-4.0) and those in the control arm (PFS 2.6, 95% CI:<br />

1.2-5.4). This also applies to OS. Regarding toxicities, there were one<br />

AEG35156 related serious adverse event (SAE) <strong>of</strong> hypersensitivity and two<br />

sorafenib related gastrointestinal SAE. Conclusions: AEG35156 in combination<br />

with Sorafenib was well tolerated in patients with advanced HCC.<br />

Dose reduced AEG35156 in combination with sorafenib have shown more<br />

activity than sorafenib alone and warrants further investigation.<br />

4104 General Poster Session (Board #48G), Mon, 8:00 AM-12:00 PM<br />

A prospective cohort study using a Japanese nationwide survey to evaluate<br />

the therapeutic effects <strong>of</strong> surgery and percutaneous ablation for hepatocellular<br />

carcinoma. Presenting Author: Kiyoshi Hasegawa, Hepato-Biliary-<br />

Pancreatic Surgery Division, Department <strong>of</strong> Surgery, Graduate School <strong>of</strong><br />

Medicine, University <strong>of</strong> Tokyo, Tokyo, Japan<br />

Background: Which is the best treatment for less advanced hepatocellular<br />

carcinoma (HCC) with good liver function remains one <strong>of</strong> the most<br />

important and unsolved problems. To solve this problem, we conducted this<br />

study and evaluated the therapeutic impacts <strong>of</strong> surgical resection (SUR),<br />

percutaneous ethanol injection (PEI), and radi<strong>of</strong>requency ablation (RFA)<br />

on long-term outcomes in patients with HCC. Methods: A large-scale<br />

database constructed by a Japanese nationwide survey was used for this<br />

study. Between 2000 and 2005, 28,510 patients with HCC were treated<br />

by SUR, PEI, or RFA, among whom we identify 12,968 patients with no<br />

more than 3 tumors (�3cm) and liver damage <strong>of</strong> class A or B. The patients<br />

were divided into SUR group (n�5,361), RFA group (n�5,548), and PEI<br />

group (n�2,059). Rates <strong>of</strong> overall and recurrence-free survival were<br />

compared among them. Results: Median follow-up was 2.16 years. Overall<br />

survival rates at 3 and 5 years were respectively 85.3%/71.1% in the SUR<br />

group, 81.0%/61.1% in the RFA, and 78.9%/56.3% in the PEI. Recurrence-free<br />

survival rates at 3 and 5 years were 56.7%/36.2%, 42.8%/<br />

28.3%, and 35.7%/23.1%, respectively. On multivariate analysis, the<br />

hazard ratio for death was significantly lower in the SUR group than in the<br />

RFA (SUR vs. RFA:0.84, 95% confidence interval, 0.74-0.95; p�0.006)<br />

and the PEI (SUR vs. PEI:0.75, 0.64-0.86; p�0.0001). The hazard ratios<br />

for recurrence were also lower in the SUR group than in the RFA (SUR vs.<br />

RFA:0.74, 0.68-0.79; p�0.0001) and the PEI (SUR vs. PEI:0.59,<br />

0.54-0.65; p�0.0001). Conclusions: Surgical resection would provide<br />

longer overall and recurrence-free survival than either RFA or PEI in<br />

patients with HCC.<br />

4106 General Poster Session (Board #49A), Mon, 8:00 AM-12:00 PM<br />

Blood signatures for early liver cancer detection in patients with chronic<br />

hepatitis B. Presenting Author: Choong-Chin Liew, GeneNews Ltd, Richmond<br />

Hill, ON, Canada<br />

Background: Some 20-30% <strong>of</strong> patients with chronic hepatitis B (CHB)<br />

eventually develop hepatocellular carcinoma (HCC), a disease with poor<br />

prognosis and high mortality unless detected early. Ultrasound and/or<br />

alpha-fetoprotein (AFP) are widely used for HCC surveillance. However<br />

these technologies are operator-dependent, have low sensitivity and are<br />

considered inadequate for screening. This study describes a blood-based<br />

gene-expression signature prognostic for HCC in CHB patients. Methods:<br />

PaxGene was used to extract RNA from whole blood samples taken from<br />

more than 1,000 Malaysian patients (matched HCC, CHB, healthy controls).<br />

Complementary DNA was generated from isolated RNA via reverse<br />

transcription and analyzed using Affymetrix U133Plus 2.0 with MAS 5.0<br />

normalization. To minimize bias we developed a novel algorithm based on<br />

SentinelPair regression analysis to identify biomarker panels. A Monte<br />

Carlo search identified pair combinations that could differentiate between<br />

cancer and controls. Twelve genes were selected for qRT-PCR Cohort 1<br />

verification (n�139). Six genes were then short-listed for qRT-PCR Cohort<br />

2 verification using independent samples (n�150). Results: Multivariate<br />

quantitative microarray analysis <strong>of</strong> 147 samples (HCC�47; CHB�50;<br />

controls�50) identified 12 probe sets differentially expressed between<br />

HCC patients and controls and CHB (ROC AUC�0.90). These genes were<br />

evaluated using qRT-PCR on the same cohort (ROC AUC�0.86). Six genes<br />

were subsequently short listed for qRT-PCR Cohort 2 verification. The AUC<br />

<strong>of</strong> the 6-gene-combination <strong>of</strong> Cohort 2 (HCC�50; CHB�50; controls�50)<br />

was 0.80. Importantly, <strong>of</strong> the thirteen HCC with low AFP (�15ng/ml), 9<br />

were predicted as “positive.” Conclusions: Blood RNA biomarkers identified<br />

in this study may form the basis <strong>of</strong> a test that can be used in patients with<br />

CHB for detection <strong>of</strong> early HCC, potentially improving prognosis for this<br />

cancer. This blood-based test would represent a significant clinical<br />

alternative to ultrasound and/or AFP.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


4107 General Poster Session (Board #49B), Mon, 8:00 AM-12:00 PM<br />

Treatment <strong>of</strong> advanced or metastatic hepatocellular cancer (HCC): Final<br />

clinical results <strong>of</strong> a single-arm phase II study <strong>of</strong> bevacizumab and<br />

everolimus. Presenting Author: Gerhard Treiber, Department <strong>of</strong> Internal<br />

Medicine, Zollernalb Clinic, Balingen, Germany<br />

Background: mTOR inhibitors are emerging drugs for treatment (Tx) <strong>of</strong><br />

advanced hepatocellular cancer (HCC), their therapeutic effects may be<br />

potentiated by combination with anti-VEGF partners (Treiber G, Expert Rev<br />

Anticancer Ther 2009). Methods: Pts with histologically confirmed advanced<br />

HCC were included. Tx consisted <strong>of</strong> RAD001 in a daily start dose <strong>of</strong><br />

5 mg orally and bevacizumab 5mg/kg iv every 2 weeks, up to 24 weeks.<br />

Primary endpoint was TTP, secondary endpoints were OS, RR, changes in<br />

biomarkers, and safety and tolerability besides PK analysis. Pts with either<br />

a CPT Score � 9, a CLIP score � 3, or a Performance Status <strong>of</strong> ECOG � 2<br />

were not eligible. Radiological TTP was centrally evaluated by both,<br />

conventional(c)-, and modified(m)-RECIST criteria on a PP basis. Additionally,<br />

we included a combined endpoint (TTPcomb: time to either clinical or<br />

radiological progression leading to termination <strong>of</strong> treatment) as assessed by<br />

the local centers in all recruited patients. Results: 33 pts (ITT) were<br />

recruited (26 with Child A, 7 with Child B; median CLIP score was 2, mean<br />

age 67 y, and mean BMI 28). 17/33 pts. had previous Tx (including<br />

sorafenib � 3 months). Drop out other than PD was due to consent<br />

withdrawl, lost to FU, and AE (n�5), therefore 28 patients had received<br />

study medication �2 wks (PP). There were 65 CTC-AE grade 3/4 events<br />

occuring in 22 pts. Median Tx duration was 69 days on RAD001, and a<br />

median <strong>of</strong> 5 cycles <strong>of</strong> bevacizumab were applied. Dose adjustements for<br />

RAD001 were performed in 20 pts, mean cumulative individual RAD dose<br />

was 334 mg, there was no association between Cmin <strong>of</strong> RAD and TTP or<br />

OS. On ITT, median TTPcomb was 2.0 mo and median OS was 9.0 mo. On<br />

PP, median TTP approached 3.8 mo (conventional RECIST) and median<br />

OS 9.7 mo. Using (m)RECIST, 5 pts. achieved PR within the first 8 wks,<br />

and this was accompanied by a significant VEGF plasma decrease (median<br />

<strong>of</strong> -65%), on (c)RECIST best response seen was SD. Conclusions: Combination<br />

Tx <strong>of</strong> RAD001 and bevazicumab is tolerated acceptably. TTP and OS in<br />

our 1°/2°-line HCC patients on Tx is comparable to sorafenib monotherapy<br />

in the SHARP trial (1° line only).<br />

4109 General Poster Session (Board #49D), Mon, 8:00 AM-12:00 PM<br />

Biliary tract cancer: A large institutional experience. Presenting Author:<br />

Mairead Geraldine McNamara, Princess Margaret Hospital, Toronto, ON,<br />

Canada<br />

Background: Biliary tract cancers (BTCs) encompass both cholangiocarcinoma<br />

(CC), arising in intrahepatic, perihilar (klatskin), or distal biliary tree,<br />

ampulla <strong>of</strong> vater and gallbladder carcinoma (GBC). Prognosis is poor for<br />

majority <strong>of</strong> these patients (pts). Aim: To retrospectively review outcomes <strong>of</strong><br />

pts, as practices/treatments evolve, who presented to a multidisciplinary<br />

team at tertiary referral center Princess Margaret Hospital, Toronto, with a<br />

diagnosis <strong>of</strong> BTC. Methods: 1057 pts were followed from 01/87 - 09/11.<br />

Complete demographics, performance status (PS), disease site, histological<br />

diagnosis, percentage receiving surgery with curative intent, chemotherapy<br />

(CT), radiotherapy (RT), recurrence patterns and overall survival<br />

(OS) were analyzed. Results: The cohort includes 549 (52%) males, PS <strong>of</strong><br />

0-1 in 850 (80%), 2-3 in 87 (8%) pts. A histological diagnosis <strong>of</strong><br />

adenocarcinoma was confirmed in 885 (84%), 106 (10%) non diagnostic,<br />

66 (6%) other. Definitive surgery was performed in 41% and adjuvant CT or<br />

concurrent CT/RT given in 20% and 8% respectively. CT or CT/RT was<br />

given for unresectable/metastatic disease in first line palliative setting in<br />

395 (37%) and 18 pts (5%) respectively. Response to first line CT in GBC<br />

was 33% vs. 24% in CC (p�0.005) and med survival was 8.4 and 13.1 mo<br />

respectively (p�0.001). The OS for entire cohort <strong>of</strong> 1057 pts was 19.3 mo<br />

with survival for breakdown <strong>of</strong> tumor type, stage detailed in the table. At<br />

this time, 207 (20%) are still alive, 609 (58%) deceased, status unknown/<br />

pending in 241 (22%). Conclusions: This represents a large biliary cancer<br />

cohort with survival benchmarks obtained in modern era <strong>of</strong> multidisciplinary<br />

care. Different subsites clearly present at different stages and have<br />

different prognosis with treatments. Despite better responses for advanced<br />

disease on CT in GBC, survival was better in CC consistent with reported<br />

literature. Therapeutic advancement mandates finding additional drug<br />

options and appropriate adjuvant care.<br />

Location N (%)<br />

Stage I<br />

(%)<br />

Stage II<br />

(%)<br />

Stage III<br />

(%)<br />

Stage IV<br />

(%)<br />

Median<br />

survival<br />

(mo)<br />

Gallbladder 304 (29) 8 12 17 63 13.0<br />

Distal bile duct 248 (23) 32 32 6 30 22.2<br />

Ampulla <strong>of</strong> vater 186 (18) 51 23 13 13 46.8<br />

Klatskin 162 (15) 14 10 24 52 16.5<br />

Intrahepatic 157 (15) 13 18 10 59 20.4<br />

Overall 1,057 (100) 19.3<br />

Gastrointestinal (Noncolorectal) Cancer<br />

265s<br />

4108 General Poster Session (Board #49C), Mon, 8:00 AM-12:00 PM<br />

A phase I/II study <strong>of</strong> foretinib, an oral multikinase inhibitor targeting MET,<br />

RON, AXL, TIE-2, and VEGFR in advanced hepatocellular carcinoma<br />

(HCC). Presenting Author: Thomas Cheung Yau, Department <strong>of</strong> Surgery,<br />

The University <strong>of</strong> Hong Kong, Hong Kong, China<br />

Background: Hepatocyte growth factor (HGF)/MET signalling plays a pivotal<br />

role in tumor cell proliferation, migration and invasion in HCC and<br />

circulating levels <strong>of</strong> HGF correlate with poor prognosis. This phase I/II trial<br />

(MET111645) evaluated foretinib, an oral multikinase inhibitor targeting<br />

MET, RON, AXL, TIE-2 and VEGFR, as first-line therapy in Asian advanced-<br />

HCC patients. Methods: Asian patients with measurable, unresectable/<br />

metastatic HCC, no prior sorafenib or other multi-kinase inhibitors, ECOG<br />

PS 0-1, adequate organ function and Child-Pugh grade A were recruited.<br />

The phase I was a standard 3�3 dose escalation design with a phase II<br />

cohort expansion. The primary endpoint was safety and tolerability at the<br />

maximum tolerated dose (MTD) and the secondary endpoints included<br />

antitumor activity (objective response rate [ORR], disease stabilization rate<br />

[DSR; confirmed CR/PR or SD for at least 12 weeks], and time to<br />

progression [TTP] evaluated by central review according to modified<br />

RECIST), and overall survival (OS) at the MTD, plus pharmacokinetics<br />

(PK). Results: Thirteen patients were enrolled in phase I. Two dose-limiting<br />

toxicities (DLT) (renal failure, proteinuria) were observed at 45 mg once<br />

daily (QD) but no DLTs were observed at 30 mg QD. Thus, the MTD was<br />

determined to be 30 mg QD. A further 32 patients were enrolled at the<br />

MTD, for a study total <strong>of</strong> 39 patients treated at 30 mg QD. The most<br />

common AEs, independent <strong>of</strong> causality,were hypertension (36%), decreased<br />

appetite (23%), and pyrexia (21%). The most common SAEs were<br />

hepatic encephalopathy (10%) and ascites (8%). Two patients discontinued<br />

foretinib due to AEs. No dose reductions were reported. Thirty-eight<br />

patients were evaluable for efficacy. The ORR was 24% (95% CI 11-40),<br />

DSR 79% (95% CI; 63-90), and the median TTP was 4.2 months (95% CI<br />

2.7-7.5). Mature OS data will be presented. Mean steady-state exposures<br />

(AUC/Cmax) were comparable after administration <strong>of</strong> foretinib at 30 and 45<br />

mg. Conclusions: Foretinib has an acceptable safety, tolerability, and PK<br />

pr<strong>of</strong>ile in an Asian HCC population. It has demonstrated promising<br />

antitumor activity that warrants further testing in a randomized setting.<br />

4110 General Poster Session (Board #49E), Mon, 8:00 AM-12:00 PM<br />

Adjuvant chemotherapy following curative intent hepatectomy for intrahepatic<br />

cholangiocarcinoma: Results from a multi-institutional analysis <strong>of</strong><br />

575 patients. Presenting Author: Dario Ribero, Ospedale Mauriziano,<br />

Torino, Italy<br />

Background: Surgical resection alone is the standard <strong>of</strong> care for patients<br />

with resectable intrahepatic cholangiocarcinoma (IHC). This study evaluates<br />

the benefit <strong>of</strong> adjuvant chemotherapy ( AdjCTx) following curative intent<br />

hepatectomy for IHC. Methods: Clinicopathologic and long-term outcome<br />

data <strong>of</strong> 575 consecutive patients treated with curative intent hepatectomy<br />

for IHC (1995-2011) were extracted from a multi-institutional registry.<br />

After excluding operative mortality and M1 (n�46), Cox regression analysis<br />

was used to identify independent determinants <strong>of</strong> early recurrence (i.e.,<br />

within 3 years). Propensity scores, which are used in observational studies<br />

to reduce selection bias by equating groups on the basis <strong>of</strong> relevant<br />

covariates, were calculated and utilized to match patients who had or had<br />

not AdjCTx (one-to-one match). Cases whose propensity score deviated more<br />

than 0.10 were considered unmatched and excluded from the analysis.<br />

Primary end-point was recurrence-free survival (RFS) at 3-years. Results: At<br />

a median FU <strong>of</strong> 42 months, 247 patients had recurred. Predictors <strong>of</strong><br />

recurrence were LN metastases (HR 1.83 [1.36-2.44]), radical resection<br />

(HR 0.64 [0.45-0.9]), an elevated preoperative CA19.9 (HR 1.54 [1.15-<br />

2.07]), vascular invasion (HR 1.97 [1.49-2.61]), multiple tumors (HR<br />

2.21 [1.71-2.86]), and size (analysed as continuous variable) (HR 1.01<br />

[1.01-1.01]). After matching, no difference was observed between patients<br />

who had or had not AdjCTx (n�155 per group; 3-yrs RFS 28.3% vs. 38.0%,<br />

respectively; p�NS). When the analysis was restricted to patients who had<br />

gemcitabine, GEMOX or FOLFOX for 3 or more cycles (n�64 per group)<br />

again no difference emerged between patients who had or had not AdjCTx<br />

(3-yrs RFS 27.7% vs. 40.0% respectively, p�NS ). Conclusions: Our data<br />

suggest that AdjCTx following resection <strong>of</strong> IHC does not increase 3-years<br />

RFS.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


266s Gastrointestinal (Noncolorectal) Cancer<br />

4111 General Poster Session (Board #49F), Mon, 8:00 AM-12:00 PM<br />

A phase II trial <strong>of</strong> gemcitabine, irinotecan, and panitumumab in advanced<br />

cholangiocarcinoma, with correlative analysis <strong>of</strong> EGFR, KRAS, and BRAF:<br />

An interim report. Presenting Author: Davendra Sohal, Abramson Cancer<br />

Center at the University <strong>of</strong> Pennsylvania, Philadelphia, PA<br />

Background: Cholangiocarcinoma is an aggressive neoplasm. Current chemotherapy<br />

approaches achieve modest results. The epidermal growth<br />

factor receptor (EGFR) pathway appears to be associated with tumor stage,<br />

prognosis and response to therapy. This trial was designed to evaluate the<br />

tolerability and efficacy <strong>of</strong> the combination <strong>of</strong> panitumumab, a monoclonal<br />

anti-EGFR antibody, with gemcitabine and irinotecan, in patients with<br />

advanced cholangiocarcinoma. Molecular analysis <strong>of</strong> EGFR pathway genes<br />

was planned as well. Methods: Patients with advanced (unresectable or<br />

metastatic) cholangiocarcinoma, ECOG PS 0-2, and adequate liver, kidney<br />

and bone marrow function were treated with panitumumab (9 mg/kg) on<br />

day 1, and gemcitabine (1000 mg/m2 ) and irinotecan (100 mg/m2 ) on days<br />

1 and 8 <strong>of</strong> a 21-day cycle. Tissue specimens were collected at diagnosis for<br />

correlative molecular analyses. Primary objective is to evaluate the 5-month<br />

progression-free survival (PFS) rate. Secondary objectives include overall<br />

response rate (ORR), overall survival (OS) and toxicity <strong>of</strong> the combination.<br />

Mutational analysis <strong>of</strong> EGFR (del 19; 858), KRAS (codons 12, 13) and<br />

BRAF (V600E) was done on samples with adequate material for testing.<br />

Results: There have been 26 (<strong>of</strong> planned 42) patients recruited to the study.<br />

A median <strong>of</strong> 6 (0-30) cycles were administered. There were no treatment<br />

related deaths. The most common gr 3 or higher toxicities were neutropenia<br />

(10 pts, 38%), thrombocytopenia (10 pts, 38%), skin rash (10 pts, 38%)<br />

and diarrhea (3 pts, 12%). During the study, there were 3 CR, 6 PR, 10 SD<br />

(disease control rate <strong>of</strong> 90%), and 2 PD (by RECIST) in 21 evaluable pts.<br />

Two pts went on to have surgical resection. Median OS is 12.7 months. Of<br />

13 testable samples, no EGFR or BRAF mutations were identified; however,<br />

there were 7 KRAS mutations. Retrospective analysis showed no difference<br />

in OS by KRAS mutation status. Conclusions: Interim evaluation <strong>of</strong> this<br />

ongoing study showed encouraging tolerability and efficacy <strong>of</strong> this regimen.<br />

Several patients have KRAS mutations; there appears to be no association<br />

with response, however. The pre-specified efficacy criteria to continue<br />

enrollment were met.<br />

4113 General Poster Session (Board #49H), Mon, 8:00 AM-12:00 PM<br />

SWOG 0941: A phase II study <strong>of</strong> sorafenib and erlotinib in patients (pts)<br />

with advanced gallbladder cancer or cholangiocarcinoma. Presenting<br />

Author: Anthony B. El-Khoueiry, University <strong>of</strong> Southern California Norris<br />

Comprehensive Cancer Center, Los Angeles, CA<br />

Background: The treatment <strong>of</strong> pts with advanced biliary cancers represents<br />

a therapeutic challenge. The vascular endothelial growth factor and<br />

epidermal growth factor receptor pathways play an important role in biliary<br />

carcinogenesis, as evidenced by their up-regulation and prognostic impact.<br />

Methods: The primary objective was progression free survival (PFS) (improvement<br />

in PFS from 4 to 8 months); secondary endpoints included overall<br />

survival (OS), objective response rate, and toxicity. A two-stage design was<br />

used; if 13 or more eligible pts <strong>of</strong> the 25 initially accrued were alive without<br />

progression at 4 months, an additional 25 were to be accrued. Eligibility<br />

criteria included no prior treatment for advanced or metastatic disease,<br />

histologic diagnosis <strong>of</strong> gallbladder cancer or cholangiocarcinoma, presence<br />

<strong>of</strong> measurable disease, Zubrod performance status 0-1, AST/ALT � 5<br />

IULN, total bilirubin �1.5 IULN, adequate hematologic function. Pts were<br />

treated with sorafenib 400 mg PO BID and erlotinib 100 mg PO q daily<br />

continuously. Restaging scans were performed every 8 weeks. Results: 32<br />

eligible pts were accrued. 30 pts were evaluable for response. 2 patients (7<br />

%) had an unconfirmed partial response (95% CI:1%-23%), and 8 pts<br />

(27%) had stable disease. Median PFS was 2 months (95% CI: 2-3<br />

months). 22/32 patients progressed or died within 4 months <strong>of</strong> registration.<br />

Median OS was 6 months (95% CI: 3 -7 months). The study failed to meet<br />

its primary endpoint to proceed to the second stage <strong>of</strong> accrual. There were 3<br />

deaths on study, 1 <strong>of</strong> which was possibly related to treatment. 19 patients<br />

(59%) had grade 3 or 4 toxicities: AST/ALT (22%), bilirubin (16%),<br />

alkaline phosphatase (16%), hypertension (16%), diarrhea (9%), hepatic<br />

infection (6%). Conclusions: This multicenter study was the first to evaluate<br />

the combination <strong>of</strong> sorafenib and erlotinib in pts with advanced biliary<br />

cancers. Despite manageable toxicity, the study failed to meet its primary<br />

endpoint. Correlative studies on collected tissue and blood are ongoing;<br />

improved pt selection based on tumor biology and molecular markers is<br />

necessary for future evaluation <strong>of</strong> targeted therapies in this disease.<br />

4112 General Poster Session (Board #49G), Mon, 8:00 AM-12:00 PM<br />

<strong>Clinical</strong>, pharmacodynamic (PD), and pharmacokinetic (PK) evaluation <strong>of</strong><br />

cediranib in advanced hepatocellular carcinoma (HCC): A phase II study<br />

(CTEP 7147). Presenting Author: Andrew X. Zhu, Division <strong>of</strong> Hematology<br />

and Oncology, Massachusetts General Hospital, Boston, MA<br />

Background: Sorafenib remains the only approved systemic therapy in HCC.<br />

We performed a phase II study <strong>of</strong> cediranib (AZD2171)—a more potent and<br />

selective pan-VEGF receptor inhibitor—in advanced HCC patients (pts).<br />

Methods: Eligibility criteria included unresectable or metastatic measurable<br />

HCC, ECOG PS �2, CLIP score �3, and adequate organ function.<br />

Patients received cediranib at 30 mg po qd continuously (4-wk cycle). The<br />

primary endpoint was progression free survival (PFS). We also assessed<br />

overall survival (OS) and response rates, steady-state PK <strong>of</strong> cediranib, and<br />

blood circulating biomarkers. Results: Since 6/16/09, we have enrolled the<br />

targeted 17 pts required for the first stage <strong>of</strong> the planned study: ECOG<br />

0/1/2�5/11/1, CLIP 1/2/3�6/4/7, Child A/B�14/3, BCLC C�17. Nine<br />

pts had prior sorafenib. The best response was stable disease in five pts<br />

(29%). The median PFS was 5.3 months (95% CI: 3.5-9.7). The median<br />

OS was 11.7 months (95% CI: 7.5-13.6). Grade 3 toxicities included<br />

hypertension (29%), hyponatremia (12%), elevated SGOT (12%) and one<br />

pt each (6%) in SGPT, fatigue, hyperbilirubinemia, cardiac ischemia, and<br />

proteinuria. Grade 4 pulmonary embolism and brainstem hemorrhage<br />

occurred in 1 pt each. Steady-state PK parameters (mean�SD) were, Cmin, 22�21 ng/mL; Cmax, 55�33 ng/mL; AUC�, 887�503 ng*h/mL. Plasma<br />

levels <strong>of</strong> VEGF and PlGF increased and sVEGFR1, sVEGFR2 and Ang-2<br />

decreased significantly after cediranib treatment (p�0.05). PFS was<br />

inversely correlated with baseline levels <strong>of</strong> bFGF, sVEGFR2 and VEGF, and<br />

OS was inversely correlated with baseline levels <strong>of</strong> sVEGFR1, Ang-2,<br />

TNF-alpha and CD34�CD133� hematopoietic progenitor cells (p�0.05).<br />

Conclusions: Cediranib at 30 mg daily is associated with high frequency <strong>of</strong><br />

grade 3 hypertension and shows preliminary evidence <strong>of</strong> antitumor activity<br />

in advanced HCC pts. Exploratory studies confirmed potential PD and<br />

response biomarkers <strong>of</strong> anti-VEGF therapy. Cediranib exhibits similar PK in<br />

HCC pts as in those with other tumor types and normal/near normal hepatic<br />

function. This study was stopped by AstraZeneca after discontinuation <strong>of</strong><br />

cediranib development for unrelated factors.<br />

4114 General Poster Session (Board #50A), Mon, 8:00 AM-12:00 PM<br />

Multiparametric quantitative functional MRI for assessing early changes in<br />

volumetric functional tumor burden in hepatocellular carcinoma treated by<br />

intra-arterial therapies. Presenting Author: Vivek Gowdra Halappa, The<br />

Johns Hopkins Hospital, Baltimore, MD<br />

Background: Hepatocellular carcinoma (HCC) is the most common primary<br />

malignant disease <strong>of</strong> the liver. Intra-arterial therapy (IAT) has been shown<br />

to improve survival and is commonly used in unresectable HCC. Assessing<br />

response to IAT as early as possible using objective criteria is paramount for<br />

clinical care. The purpose <strong>of</strong> this study was to evaluate volumetric diffusion<br />

weighted (DWI) and contrast Enhanced MR imaging (CE-MRI) changes for<br />

assessing early treatment response to IAT in hepatocellular carcinoma.<br />

Methods: This study included 177 lesions <strong>of</strong> HCC in 107 patients (85 Male,<br />

mean age 64 years) with BCLC A (19) B (44) C (36) D (8) treated by IAT. All<br />

patients had pre-IAT and 3-4 weeks follow up MRI with DWI and CE-MR.<br />

Tumors were segmented using semi automated s<strong>of</strong>tware to provide volumetric<br />

functional analysis <strong>of</strong> DWI and CE- MRI as well as percent tumor<br />

burden. Statistical analyses include paired t-test, Kaplan-Meier curves, log<br />

rank test and multivariate analysis with cox model. Results: There was<br />

significant increase in volumetric ADC and significant decrease in volumetric<br />

arterial (AE) and venous (VE) enhancement after IAT. Patients with<br />

tumor burden <strong>of</strong> � 10% had improved survival compared to patients with<br />

high tumor burden <strong>of</strong> �10% (log rank p �.008 ) This survival benefit was<br />

larger among patients with tumor burden <strong>of</strong> � 10% who demonstrated an<br />

increase in volumetric ADC (�20%. Overall survival (OS) 33.5 mo) and<br />

decrease in volumetric VE (�50% OS 35.2 mo) compared to patients with<br />

high tumor burden �10% who no favorable response in ADC ( OS 14.1 mo)<br />

or VE (OS 16.5 mo) (Log rank p � .001) . In multivariate analysis factors<br />

associated with favorable prognosis were: increased ADC, (Hazard Ratio<br />

(HR) � 0.44, 95% CI �0.24 - 0.80) decreased VE, (HR � 0.43, 95% CI �<br />

0.22 - 0.84) and BCLC A and B (HR � 0.31, 95% CI �0.18 - 0.54).<br />

Conclusions: Volumetric functional analysis <strong>of</strong> DWI and CE-MR are reliable<br />

imaging biomarkers <strong>of</strong> treatment response and tumor necrosis were able to<br />

differentiate responders and non responders. Early assessment <strong>of</strong> treatment<br />

response by volumetric functional MRI predicts prognosis in patients<br />

undergoing IAT in HCC.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


4115 General Poster Session (Board #50B), Mon, 8:00 AM-12:00 PM<br />

Efficacy, safety, tolerability, and PK <strong>of</strong> the HDAC inhibitor resminostat in<br />

sorafenib-refractory hepatocellular carcinoma (HCC): Phase II SHELTER<br />

study. Presenting Author: Michael Bitzer, Medical University Clinic, Eberhard-Karls-University,<br />

Tuebingen, Germany<br />

Background: Resminostat (R), an oral HDAC inhibitor, was studied in the<br />

SHELTER trial evaluating safety, PK and efficacy in HCC patients (pts)<br />

refractory to sorafenib (S). R was explored as monotherapy and within a<br />

novel resensitization approach to overcome tolerance to S by the combination<br />

<strong>of</strong> both drugs. Methods: Pts with advanced HCC (BCLC B/C) were<br />

included in a multi-center, two-arm trial. Radiologic progression under S<br />

firstline therapy had to be confirmed by central review (RECIST) prior to<br />

study entry. A dose escalation <strong>of</strong> R (range 200 to 600 mg) combined with S<br />

(400 or 800 mg) was performed. Arm A investigated the drug combination<br />

(R�S), Arm B the monotherapy <strong>of</strong> R (600 mg). Primary objective was the<br />

progression-free survival rate (PFSR) after 12 weeks (w). Secondary<br />

objectives included safety, tolerability, tumor response, PFS, TTP, OS and<br />

the analyses <strong>of</strong> PK and biomarkers (BM), incl. AFP, VEGF, HDAC enzyme<br />

inhibition, histone acetylation and gene expressions in peripheral blood.<br />

Results: 50 pts were enrolled, dose escalation determined 600 mg R and<br />

400 mg S for Arm A. <strong>Clinical</strong> activity results <strong>of</strong> 15 evaluable pts from<br />

combination treatment revealing a PFSR <strong>of</strong> 66.6% after 12 w, not all<br />

patients in Arm B already reached the 12 w staging. Most frequent AE were<br />

CTC grade 1-2 GI complaints (nausea, vomiting) and skin disorders (rash,<br />

pruritus, HFSR). Grade 3-4 toxicity (SAE reports) consisted mainly <strong>of</strong><br />

non-hematological events mostly related to tumor disease. Plasma concentrations<br />

<strong>of</strong> both drugs correlated with administered doses and were in the<br />

expected range without obvious influence <strong>of</strong> preexisting liver disease. BM<br />

investigations revealed effective target modulation by R in both arms.<br />

Conclusions: The primary study objective was achieved for both treatment<br />

arms. R demonstrated a favorable PK, safety and tolerability pr<strong>of</strong>ile, even in<br />

combination with S and despite preexisting liver disease, includig cirrhosis.<br />

The observed clinical activity emphazises the resensitizing activity <strong>of</strong> R by<br />

an epigenetic mode-<strong>of</strong>-action to overcome tolerance to S and warrants<br />

further development, in particular for combination therapy in both, first and<br />

second line HCC treatment.<br />

4118 General Poster Session (Board #50E), Mon, 8:00 AM-12:00 PM<br />

Updated results from a phase III trial <strong>of</strong> sunitinib versus placebo in patients<br />

with progressive, unresectable, well-differentiated pancreatic neuroendocrine<br />

tumor (NET). Presenting Author: Aaron Vinik, EVMS Strelitz Diabetes<br />

Research Center and Neuroendocrine Unit, Norfolk, VA<br />

Background: In a double-blind phase III trial, sunitinib (Sutent; SU)<br />

improved progression-free survival (PFS) vs placebo (PBO; 11.4 vs 5.5<br />

mos; HR: 0.42, 95% CI: 0.26–0.66; P�0.0001) and was well tolerated in<br />

patients (pts) with unresectable, well-differentiated pancreatic NET that<br />

had progressed �12 mos before baseline. Initial overall survival (OS)<br />

revealed a benefit for SU vs PBO, although median OS had not been<br />

reached. We now report PFS assessed by blinded independent central<br />

review (BICR) and updated OS. Methods: Pts were randomized 1:1 to SU<br />

37.5 mg or PBO on a continuous daily dosing schedule, each with best<br />

supportive care. The primary endpoint was investigator-assessed PFS; OS<br />

was a secondary endpoint to be evaluated every 2 yrs for 5 yrs, or until 95%<br />

<strong>of</strong> pts had died. Additionally, BICR was performed retrospectively; baseline<br />

and on-study CT/MRI scans were evaluated by a 2-reader, 2-time-point<br />

lock, followed by a sequential locked-read, batch-mode paradigm by<br />

blinded, third-party radiologists. Results: 171 pts were randomized (SU,<br />

n�86; PBO, n�85) from Jun 2007 to Apr 2009. The trial ended when an<br />

independent data monitoring committee noted efficacy favoring SU and<br />

more serious AEs and deaths with PBO. The study was unblinded at<br />

closure; pts were <strong>of</strong>fered open-label SU and followed for survival. Median<br />

PFS by BICR was 12.6 mos (SU) vs 5.8 mos (PBO) (HR: 0.32, 95% CI:<br />

0.18–0.55; P�0.00001). At study closure, there were 9 and 21 deaths in<br />

the SU and PBO arms (HR: 0.41, 95% CI: 0.19–0.89; P�0.02). By Apr<br />

2011 (2 yrs additional follow-up) a total <strong>of</strong> 87 deaths occurred (51%),<br />

median OS was estimated at 33.0 mos in the SU arm, and 26.7 mos in the<br />

PBO arm (HR: 0.71, 95% CI: 0.47–1.09; P�0.11 [Table]). 69% <strong>of</strong> pts<br />

crossed over to SU on progression. Conclusions: BICR confirmed investigator<br />

assessment <strong>of</strong> PFS and demonstrated a 6.8-mo improvement in median<br />

PFS with SU. Updated OS favors SU, although this result is non-significant<br />

for reasons that may include crossover <strong>of</strong> treatment and limited statistical<br />

power.<br />

2-yr OS update, Apr 2011<br />

Deaths, n<br />

Censored, n<br />

Median OS (95% CI), mos<br />

SU<br />

n�86<br />

40<br />

46<br />

33.0 (25.6–NR)<br />

HR (95% CI) 0.71 (0.47–1.09)<br />

P 0.11<br />

Gastrointestinal (Noncolorectal) Cancer<br />

PBO<br />

n�85<br />

47<br />

38<br />

26.7 (16.4–35.3)<br />

267s<br />

4117 General Poster Session (Board #50D), Mon, 8:00 AM-12:00 PM<br />

Safety and efficacy <strong>of</strong> MET inhibitor tivantinib (ARQ 197) combined with<br />

sorafenib in patients (pts) with hepatocellular carcinoma (HCC) from a<br />

phase I study. Presenting Author: Robert E. Martell, Tufts Medical Center<br />

Cancer Center, Boston, MA<br />

Background: The multikinase inhibitor sorafenib is standard <strong>of</strong> care for pts<br />

with advanced HCC. Tivantinib, an oral, selective MET inhibitor, demonstrated<br />

synergistic antitumor activity when combined with sorafenib in<br />

several tumor models. The phase 1 dose-escalation study assessed the<br />

safety pr<strong>of</strong>ile <strong>of</strong> tivantinib plus sorafenib in pts with advanced solid tumors.<br />

Methods: Endpoints were safety, the recommended phase 2 dose (RP2D) <strong>of</strong><br />

tivantinib plus sorafenib, and antitumor activity. Dose escalation previously<br />

established the RP2D as tivantinib 360 mg twice daily (BID) plus sorafenib<br />

400 mg BID. Extension cohorts enrolled � 20 pts each with HCC or other<br />

tumors. Patients were treated until disease progression or unacceptable<br />

toxicity. After a safety review in HCC pts in other studies and a report <strong>of</strong><br />

febrile neutropenia in this study, the tivantinib dose for newly enrolled pts<br />

was reduced to 240 mg BID. Results: 20 pts with HCC (mean age, 62 yr;<br />

Child-Pugh [CP] A [n � 14], or CP B [n � 6]) were treated at the RP2D (n �<br />

10) or at the reduced tivantinib dose plus sorafenib (n � 10). 10 pts<br />

received � 1 previous systemic anticancer treatment (median, 0.5; range,<br />

0-3). The most common adverse events (� 25%) were rash (40%),<br />

palmar-plantar erythrodysesthesia syndrome (35%), fatigue and diarrhea<br />

(30% each), and nausea and anorexia (25% each). Neutropenia was<br />

reported in 2 pts. Best response was 1 complete response (CR), 1 partial<br />

response (PR), and 12 stable disease (SD). Overall response rate and<br />

disease control rate were 10% and 70%, respectively. Median progressionfree<br />

survival (mPFS) was 3.5 mo (95% CI, 2.8-11.1 mo). Among 8 pts<br />

previously treated with vascular endothelial growth factor (VEGF) inhibitors<br />

(6 sorafenib; 1 sunitinib; 1 sorafenib plus sunitinib), best response was 1<br />

CR, 1 PR, and 3 SD, and mPFS was 15.9 mo (95% CI, 1.6-15.9 mo). 2 pts<br />

are still on study. Conclusions: The combination <strong>of</strong> tivantinib (360 mg BID)<br />

plus sorafenib (240 mg BID) was well tolerated. Preliminary evidence <strong>of</strong><br />

antitumor activity indicates that combined inhibition <strong>of</strong> MET and angiogenic<br />

signals may have therapeutic potential in this setting, including pts<br />

pretreated with VEGF inhibitors.<br />

4119 General Poster Session (Board #50F), Mon, 8:00 AM-12:00 PM<br />

PAZONET: Results <strong>of</strong> a phase II trial <strong>of</strong> pazopanib as a sequencing<br />

treatment in progressive metastatic neuroendocrine tumors (NETs) patients<br />

(pts), on behalf <strong>of</strong> the Spanish task force for NETs (GETNE)—<br />

NCT01280201. Presenting Author: Enrique Grande Pulido, Hospital<br />

Ramon y Cajal, Madrid, Spain<br />

Background: Pazopanib is an oral tyrosine kinase inhibitor <strong>of</strong> the VEGFR,<br />

PDGFR and KIT that has demonstrated clinical activity in NETs. The aims<br />

<strong>of</strong> our study were to assess efficacy, safety and potential predictive<br />

biomarkers <strong>of</strong> pazopanib in pts with NETs who had previously failed to at<br />

least one antiangiogenic or mTOR inhibitor treatment. Methods: Pts<br />

presented locally-advanced or metastatic gastrointestinal or pancreatic<br />

NET disease documented as progressive. Pazopanib 800 mg was given<br />

daily until disease progression (DP) or unacceptable toxicity. Primary<br />

endpoint was <strong>Clinical</strong> Benefit Rate (CBR) defined as Complete Response<br />

plus <strong>Part</strong>ial Response (PR) plus Stable Disease (SD) at 6 months by<br />

RECIST-V-1.0. Optimal two-stage Simon design was utilized with H1 and<br />

H0 set at 70 % and 50 % respectively, Kaplan-Meier estimates waere used<br />

for the analysis <strong>of</strong> time-to-event variables: 95% CI. Results: 33 pts were<br />

evaluable for response per protocol. 54.5% males, mean age 60.7�10.3<br />

years. At 6 months, 2 pts had PR (6%), 26 SD (79%), and 5 DP (15%),<br />

thus the CBR was 85%. By subgroups, CBR was 100% in pts with no<br />

previous targeted therapy (7 pts), 89% in pts with previous mTOR<br />

inhibitors (9 pts), 83% in pts with previous antiangiogenics (12 pts), and<br />

60% in pts with previous antiangiogenics and mTOR inhibitors (5 pts). The<br />

sum <strong>of</strong> the longest diameter <strong>of</strong> target lesions had a decrease over 10% in<br />

22% <strong>of</strong> pts (29% without previous biological treatment, 22% mTOR<br />

without prior multitarget, 18% prior multitarget without mTor, and 20%<br />

with previous multitarget and mTOR. Median PFS was only reached in the<br />

subgroup <strong>of</strong> pts (26 pts) with previously treated with antiangiogenics and<br />

mTOR inhibitors (20.6 weeks, 95% CI, 10.4-30.8). Most frequent toxicities,<br />

any grade: asthenia (75%), diarrhoea (63%), nausea (42%). Translational<br />

studies on angiogenesis and inmunohistochemistry biomarkers and<br />

CTCs are ongoing. Conclusions: Pazopanib at 800 mg daily has promising<br />

activity in advanced NET regardless <strong>of</strong> previous treatment with other<br />

targeted therapies. This trial may introduce the concept <strong>of</strong> treatment<br />

sequencing with novel targeted agents in NETs.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


268s Gastrointestinal (Noncolorectal) Cancer<br />

4120 General Poster Session (Board #50G), Mon, 8:00 AM-12:00 PM<br />

Merkel cell polyomavirus in gastrointestinal neuroendocrine tumors. Presenting<br />

Author: Salvatore Lorenzo Renne, European Institute <strong>of</strong> Oncology,<br />

Milan, Italy<br />

Background: Cutaneous Merkel cell carcinoma (MCC) is a high grade<br />

neuroendocrine carcinoma potentially induced by Merkel Cell Polyomavirus<br />

(MCPyV), a clonally integrated host in the tumor cell genome.<br />

Integration <strong>of</strong> viral DNA and truncating mutations in the helicase domain <strong>of</strong><br />

the large T (LT) antigen, a protein implicated in the viral life cycle, have<br />

been detected in all MCPyV associated MCCs. These results suggest a role<br />

<strong>of</strong> the virus in tumor pathogenesis according to a two step model: a<br />

necessary but not sufficient integration <strong>of</strong> viral DNA followed by LT antigen<br />

premature truncations. Gastrointestinal neuroendocrine tumors (GI-NETs)<br />

share common phenotypical features with MCCs, therefore we evaluated<br />

whether MCPyVs may be present in GI-NET. Methods: Formalin–fixed and<br />

paraffin-embedded samples from 14 cases <strong>of</strong> liver metastases <strong>of</strong> G2<br />

GI-NETs, 9 small cell lung cancer (SCLCs) and 4 MCCs <strong>of</strong> the skin were<br />

screened for MCPyV-DNA positivity using two PCR primer sets mapping the<br />

LT antigen gene. Then to confirm the putative pathogenetic role <strong>of</strong> the<br />

virus, we seeked for premature truncation <strong>of</strong> LT antigen by sequencing the<br />

helicase portion using seven amplicons mapping 1356-2210 region <strong>of</strong> the<br />

MCPyV complete genome (RefSeq: NC_010277.1). Results: Viral DNA was<br />

detected in 7/14 GI-NETs, in 0/9 SCLCs and in 4/4 MCCs. We found a<br />

premature stop codon truncating the helicase domain in two GI-NETs. Both<br />

the patients had a rapid disease progression. We also found previously not<br />

reported alterations in MCCs: a single aminoacid deletion in 3 cases and a<br />

point mutation causing a single aminoacid substitution in another case.<br />

Conclusions: For the first time the potential tumorigenic signature <strong>of</strong> MCPyV<br />

has been detected in GI-NETs, suggesting a possible role in pathogenesis.<br />

The study is ongoing to confirm these observations that could support new<br />

diagnostic and therapeutical perspectives <strong>of</strong> a subset <strong>of</strong> GI NETs. Moreover<br />

we have shown that the clonal integration <strong>of</strong> MCPyV in MCCs produces<br />

different genetic alterations with unknown effects on cell biology.<br />

4122 General Poster Session (Board #51A), Mon, 8:00 AM-12:00 PM<br />

RAMSETE: A single-arm, multicenter, single-stage phase II trial <strong>of</strong> RAD001<br />

(everolimus) in advanced and metastatic silent neuro-endocrine tumours in<br />

Europe. Presenting Author: Marianne E. Pavel, Charite Universitatsmedizin,<br />

Berlin, Germany<br />

Background: The mammalian target <strong>of</strong> rapamycin (mTOR) signaling pathway<br />

is involved in the pathogenesis <strong>of</strong> neuroendocrine tumors (NET).<br />

Everolimus (RAD001), an oral mTOR inhibitor, has antitumor activity in<br />

patients (pts) with advanced NET. In this open-label, multicenter, phase II<br />

study (RAMSETE), the safety and efficacy <strong>of</strong> everolimus monotherapy was<br />

evaluated in pts with advanced nonsyndromic, nonpancreatic NET. Methods:<br />

Pts with advanced (unresectable or metastatic), progressive, nonsyndromic,<br />

nonpancreatic NET received everolimus (10 mg/day) as monotherapy.<br />

The primary endpoint was objective response rate (proportion <strong>of</strong><br />

pts with best overall complete response [CR] or partial response [PR] per<br />

RECIST v1.0) by central radiologic review. A secondary endpoint included<br />

progression-free survival (PFS). Results: By database s<strong>of</strong>t lock (December<br />

1, 2011), 73 pts from 16 European clinics received everolimus (median<br />

duration <strong>of</strong> treatment: 193 days). Fifty-five (75%) pts discontinued;<br />

reasons included disease progression (n�23), adverse events (AEs [n�23]),<br />

withdrawal <strong>of</strong> consent (n�4), death (n�3), and protocol deviation (n�2).<br />

In the per protocol population (N�60), 32 (53%) pts received prior<br />

antineoplastic therapy. The best response by central review was stable<br />

disease in 55%. By local review, 3 (5%) pts had a PR, with SD in 39 (65%)<br />

pts. Median PFS by central review was 185 days (95% CI: 158-255).<br />

Median PFS by local investigator review was 285 days (95% CI: 231-not<br />

estimable). 69 (95%) pts reported treatment-related AEs <strong>of</strong> any grade,<br />

including rash (n�28; 38%), diarrhea (n�20; 27%), mucosal inflammation<br />

(n�18; 25%), and decreased appetite (n�17; 23%). Treatmentrelated<br />

grades 3 and 4 AEs and serious AEs were reported by 27 (37%) and<br />

18 (25%) pts, respectively.Conclusions: In this open-label trial <strong>of</strong> everolimus<br />

in pts with advanced, nonsyndromic, extrapancreatic NET, a high rate<br />

<strong>of</strong> disease stabilization was achieved after prior tumor progression with<br />

favorable median PFS. This study further supports efficacy <strong>of</strong> everolimus in<br />

types <strong>of</strong> NET other than those studied in RADIANT-3 (pancreatic NET) and<br />

RADIANT-2 (NET associated with carcinoid syndrome).<br />

4121 General Poster Session (Board #50H), Mon, 8:00 AM-12:00 PM<br />

Treatment <strong>of</strong> advanced neuroendocrine tumors: Results <strong>of</strong> the UKINETS<br />

and NCRI randomized phase II NET01 trial. Presenting Author: Philippa<br />

Corrie, Oncology Centre, Cambridge University Hospitals NHS Foundation<br />

Trust (Addenbrooke’s Hospital), Cambridge , United Kingdom<br />

Background: Chemotherapy is widely used for advanced, unresectable<br />

neuroendocine tumours (NETs) but only four randomised trials are published.<br />

The first combined streptozocin (strep) and 5fluorouracil (5FU),<br />

reporting 63% response rate (RR), but subsequent retrospective studies<br />

had lower RRs (6-45%). Cisplatin has been combined with strep�5FU in a<br />

retrospective study with promising activity in NETs and capecitabine (cap)<br />

has been effectively substituted for 5FU in many tumours. The NET01 trial<br />

was designed to investigate whether cap�strep � cisplatin warrant further<br />

evaluation. Methods: Patients (pts) with histologically confirmed, unresectable,<br />

advanced and/or metastatic NETs <strong>of</strong> foregut, pancreatic or unknown<br />

primary site were randomised (ratio 1:1) to 6 cycles <strong>of</strong> 3-weekly cap 625<br />

mg/m2 po bd daily (D1-21), strep 1.0g/m2 IV (D1), with cisplatin 70mg/m2 IV(D1) (Cap-cist) or without (Cap-strep). The primary outcome measure was<br />

RR using RECIST criteria (v 1.0). 84 pts were required to test a RR <strong>of</strong><br />

�40% vs �60% with a significance level <strong>of</strong> 5% and 80% power in each<br />

arm. Central pathology review was performed; retrospective central radiology<br />

review was undertaken on 10% <strong>of</strong> randomly selected cases. Results: 86<br />

pts (58% male, median age 59 years) were randomised (44 Cap-strep, 42<br />

Cap-cist) with primary site – foregut 20%, pancreatic 48% and unknown<br />

33%. The grade was low 8 (11%), intermediate 47 (67%) and high 15<br />

(21%). Baseline characteristics were balanced between the 2 arms. 60%<br />

Cap-strep pts received �6 cycles compared with 33% Cap-cist pts. 17<br />

Cap-strep and 26 Cap-cist pts experienced grade �3 toxicities. Outcome<br />

results are summarised in the table. Ki67, mitotic index or primary site <strong>of</strong><br />

origin did not appear to predict for response. Conclusions: The novel<br />

Cap-strep � cisplatin regimens were associated with overall disease control<br />

and survival durations consistent with published studies. Cap-strep was<br />

better tolerated than Cap-cist. Further prospective randomised studies are<br />

required to determine optimal NET chemotherapy.<br />

Cap-cist Cap-strep<br />

Response<br />

14%<br />

8%<br />

PR<br />

64%<br />

74%<br />

SD<br />

22%<br />

18%<br />

PD<br />

PFS 33 (79)<br />

30 (68)<br />

No. progressions/deaths, n (%)<br />

9.7<br />

10.2<br />

Median (mo)<br />

OS 34 (40)<br />

No. deaths, n (%)<br />

34.7<br />

Median (mo)<br />

4123 General Poster Session (Board #51B), Mon, 8:00 AM-12:00 PM<br />

Circulating tumor cells as prognostic and predictive markers in neuroendocrine<br />

tumors. Presenting Author: Mohid Shakil Khan, University College<br />

London, London, United Kingdom<br />

Background: Neuroendocrine tumors (NETs) are a heterogeneous group <strong>of</strong><br />

tumors with variable survival. While Ki67 in tumour tissue is prognostic,<br />

there have been no prospectively validated circulating prognostic or<br />

predictive markers. Here we present results from a large prospective study<br />

<strong>of</strong> circulating tumor cells (CTCs) in patients with NETs. Methods: Patients<br />

about to commence therapy had a 7.5 ml blood sample taken at baseline<br />

with additional samples taken at 3-5 weeks after commencing treatment.<br />

CTCs were enumerated using the CellSearch system and counted independently<br />

by two observers. Radiological response was classified according to<br />

RECIST 1.1. PFS and OS were measured from start date <strong>of</strong> therapy, to date<br />

<strong>of</strong> progression and date <strong>of</strong> death, respectively. In order to define a<br />

prognostic cut <strong>of</strong>f value for CTCs, 90 patients were enrolled in a training set<br />

and 85 patients in a validation set. The optimal cut-<strong>of</strong>f level validated was<br />

CTC�1. Prognostic factors including Ki67 were evaluated using Cox<br />

regression. Changes in CTCs after treatment were divided into tertiles.<br />

Results: 138 patients with metastatic NET (31 pancreatic, 81 midgut, 12<br />

bronchopulmonary, 11 unknown primary, 3 hindgut) were recruited who<br />

were about to undergo treatment with somatostatin analogues(34), chemotherapy(28),<br />

PRRT(40), TAE(18), RFA(3), Sunitinib(4), interferon(4) and<br />

surgery(7). Median follow-up was 9.7 months (range 5-29). Patients with<br />

baseline CTC�1 had significantly worse PFS (HR 4.3 95%CI 1.8-10.3),<br />

and OS (HR 6.1 95%CI 1.8-20.3) than those without CTCs. In multivariate<br />

analysis CTC presence was associated with worse PFS (HR 3.7 95%CI<br />

1.5-8.9) and OS (HR 5.1 95%CI1.5-7.3). Changes in CTCs predicted<br />

response to therapy (Fisher’s exact p�0.001) and those with increase in<br />

CTCs by �33% post-therapy had significantly worse PFS (HR 23.1 95%CI<br />

5.37-99.0) and OS (HR18.9 95%CI 2.4-146) than a fall or �33%<br />

increase. Conclusions: In metastatic NETs, CTCs are a promising prognostic<br />

marker and may be an early marker <strong>of</strong> response to therapy. Further<br />

evaluation <strong>of</strong> CTCs in the context <strong>of</strong> therapeutic trials is required.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


4124 General Poster Session (Board #51C), Mon, 8:00 AM-12:00 PM<br />

Prevalence and prognostic significance <strong>of</strong> FGF receptor 2 (FGFR2) gene<br />

amplification in Caucasian and Korean gastric cancer cohorts. Presenting<br />

Author: Elaine Kilgour, AstraZeneca, Oncology Innovative Medicines,<br />

Macclesfield, United Kingdom<br />

Background: Gastric adenocarcinoma is the 4th most common cancer, and<br />

many patients present with metastatic or recurrent disease. The prognosis<br />

for these patients is poor, with median survival times <strong>of</strong> 11–12 months.<br />

Hence there is a need for identification <strong>of</strong> potential new drug targets. We<br />

have determined the prevalence <strong>of</strong> FGFR2 gene amplification (FGFR2amp)<br />

and its relationship to clinicopathological parameters and survival in<br />

Caucasian (n�408) and Korean (n�356) gastric cancers (GC) and determined<br />

the overlap with HER2 or cMET gene amplification. Methods:<br />

FGFR2, HER2 and cMET gene amplification was assessed by fluorescence<br />

in situ hybridisation in GC tissue microarrays from Caucasian and Korean<br />

surgically resected gastric carcinomas. Gene amplification was defined as a<br />

gene/centromeric probe ratio <strong>of</strong> �2.0 after measuring at least 50 tumour<br />

cells. Results: 7% (30/408) <strong>of</strong> Caucasian and 4% (15/356) <strong>of</strong> Korean GC<br />

showed FGFR2amp, and the incidence was not significantly different<br />

between these cohorts (p�0.092). FGFR2amp was significantly associated<br />

with lymph node status in both cohorts (p�0.0007, multivariate analysis),<br />

and in the Korean cohort with diffuse-type histology, but not with depth <strong>of</strong><br />

tumour invasion, age or gender. Patients with FGFR2amp showed significantly<br />

shorter overall survival in both the Caucasian (HR 2.37, 95% CI<br />

1.6–3.5; p�0.0001) and Korean (HR 2.33, 95% CI 1.28–4.25;<br />

p�0.0129) cohorts, by multivariate analysis. There was no overlap<br />

between FGFR2amp and HER2 or cMET amplification in the Korean<br />

cohort, but two <strong>of</strong> twenty-six FGFR2amp Caucasian gastric tumours also<br />

showed HER2 amplification. Conclusions: This is the first study to demonstrate<br />

FGFR2amp, at a prevalence <strong>of</strong> 4% and 7%, in two large GC cohorts <strong>of</strong><br />

Asian and Caucasian origin and that FGFR2amp is prognostic and significantly<br />

associated with lymph node metastasis. Furthermore, FGFR2amp is<br />

generally mutually exclusive with HER2 and cMET amplifications. Based<br />

on these observations, FGFR2 inhibitors warrant further investigation in the<br />

treatment <strong>of</strong> FGFR2 amplified GCs.<br />

4126 General Poster Session (Board #51E), Mon, 8:00 AM-12:00 PM<br />

Prevalence <strong>of</strong> functional tumors in neuroendocrine carcinoma: An analysis<br />

from the NCCN NET database. Presenting Author: Michael A. Choti, The<br />

Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University,<br />

Baltimore, MD<br />

Background: Neuroendocrine tumors (NETs) are increasing in incidence<br />

and prevalence. Identification and treatment <strong>of</strong> specific clinical NET<br />

syndromes are established, yet there is uncertainty regarding the prevalence<br />

<strong>of</strong> NET with hormone-related symptoms versus nonfunctional tumors.<br />

Methods: The National Comprehensive Cancer Network (NCCN)<br />

created a comprehensive database to characterize patients (pts) treated for<br />

NETs. This database was queried to identify pts presenting to 7 NCCN<br />

institutions with a confirmed NET diagnosis: including carcinoid (cNET),<br />

pancreatic NET (pNET), NET not otherwise specified (NOS), and pheochromocytoma<br />

(PCC) between 2004 and 2010. The primary aim <strong>of</strong> this analysis<br />

was to describe demographic and clinical characteristics <strong>of</strong> NET pts by<br />

functional (fxn) status at diagnosis (dx). Results: Among 1244 NET pts,<br />

26% (n�327) had an fxn tumor. Carcinoid syndrome (CS) occurred in 28%<br />

<strong>of</strong> cNET pts. The most common primary tumor sites among CS pts were<br />

small bowel (69%) and unknown (15%). Prevalence <strong>of</strong> hormonal syndrome<br />

(HS) among pNET pts was 22%, 24% among NOS pts and 37% among<br />

PCC pts. The majority <strong>of</strong> CS pts (74%), pNET HS pts (67%), and NOS HS<br />

pts (91%) had distant disease at dx, in contrast to 31% <strong>of</strong> PCC HS patients.<br />

Among CS pts with a known histologic grade, 91% were well differentiated<br />

(G1). Similarly, 86% <strong>of</strong> pNET HS and 67% <strong>of</strong> NOS HS pts with a known<br />

histologic grade had G1 NETs. The most common symptoms at dx among<br />

pts with CS included abdominal cramping (53%), change in bowel habits<br />

(48%), and flushing (40%). Among those tested, 85% <strong>of</strong> CS pts had a<br />

positive 5-HIAA test at dx. Among pNET HS pts, the most common<br />

symptoms present at dx were abdominal cramping (39%) and change in<br />

bowel habits (46%). The most common symptoms present at dx among<br />

NOS HS pts were abdominal cramping (35%), change in bowel habits<br />

(47%) and flushing (38%). Conclusions: Prevalence <strong>of</strong> CS in this NCCN<br />

database (28%) was slightly higher than the 10% previously reported in the<br />

literature. In contrast, the prevalence <strong>of</strong> HS among pNET pts (22%) was<br />

lower than previously reported. Approximately one quarter <strong>of</strong> cNET pts<br />

without metastatic disease had CS, warranting further analysis as CS most<br />

<strong>of</strong>ten occurs in the presence <strong>of</strong> liver metastasis.<br />

Gastrointestinal (Noncolorectal) Cancer<br />

269s<br />

4125 General Poster Session (Board #51D), Mon, 8:00 AM-12:00 PM<br />

A multi-institutional phase II open-label study <strong>of</strong> AMG 479 in advanced<br />

carcinoid and pancreatic neuroendocrine tumors. Presenting Author: Matthew<br />

Kulke, Dana-Farber Cancer Institute, Boston, MA<br />

Background: The IGF pathway is thought play an important role in<br />

neuroendocrine tumor progression. We therefore investigated AMG 479, a<br />

human monocolonal antibody against IGF1-R, in patients with metastatic<br />

progressive carcinoid and pancreatic neuroendocrine tumors (NETS).<br />

Methods: This open-label phase II study enrolled patients (�18 yrs) with<br />

metastatic low and intermediate-grade carcinoid and pancreatic NETs. Key<br />

inclusion criteria included evidence <strong>of</strong> progressive disease (by RECIST)<br />

within 12 months <strong>of</strong> enrollment, ECOG PS 0-2, and fasting blood sugar<br />

�160mg/dL. Prior treatments were allowed, and concurrent somatostatin<br />

analog therapy was permitted as long as patients remained on a stable<br />

dose. The primary endpoint was objective response rate. Secondary<br />

endpoints included overall survival (OS), progression-free survival (PFS),<br />

and safety. Results: 60 patients (30 carcinoid, 30 pancreatic NET) were<br />

treated with AMG 479 18mg/kg every 3 weeks and 54 patients were<br />

evaluable for response. There were no objective responders by RECIST.<br />

When best response to therapy was evaluated, 10/27 (37%) evaluable<br />

carcinoid patients and 8/26 (31%) evaluable pancreatic NET patients<br />

experienced 1-29% tumor shrinkage, while 17/27 (63%) <strong>of</strong> the carcinoid<br />

patients and 15/26 (58%) <strong>of</strong> the pancreatic NET patients appeared to<br />

experience continued tumor growth. Median PFS was 6.3 months (95% CI<br />

4.2-12.6) for the entire cohort; 10.5 months for carcinoid patients and 4.2<br />

months for pancreatic NET patients. The OS rate at 12 months was 70%<br />

(55%-81%) for the entire cohort. Median OS has not been reached.<br />

Treatment related grade 3/4 AEs were rare and consisted <strong>of</strong> hyperglycemia<br />

(4%), neutropenia (4%), thrombocytopenia (4%) and infusion reaction<br />

(1%). Conclusions: While well-tolerated, single-agent AMG 479 was not<br />

found to result in major tumor responses among patients with metastatic<br />

low-intermediate grade carcinoid or pancreatic NET. Subgroup analysis to<br />

identify characteristics <strong>of</strong> patients who may have benefited from therapy<br />

with AMG 479 is ongoing.<br />

4127 General Poster Session (Board #51F), Mon, 8:00 AM-12:00 PM<br />

Bevacizumab, pertuzumab, and sandostatin for patients (pts) with advanced<br />

neuroendocrine cancers (NET). Presenting Author: Irfan Firdaus,<br />

Sarah Cannon Research Institute/Oncology Hematology Care, Inc, Cincinnati,<br />

OH<br />

Background: Targeted agents are used to treat well-differentiated NET.<br />

Octreotide binds somatostatin receptors and improves time to progression<br />

(TTP) <strong>of</strong> carcinoid tumors, and VEGF pathway targeting improves TTP <strong>of</strong><br />

pancreatic NET. HER2 overexpression is reported in NET, and pertuzumab<br />

blocks HER2 receptor dimerization. This phase II trial combined bevacizumab,<br />

pertuzumab, and octreotide LAR for pts with advanced NET.<br />

Methods: Pts with advanced well-differentiated NET were enrolled. Pts had<br />

ECOG 0-1, and normal end-organ function, including left ventricular<br />

ejection fraction (LVEF) �50%. Pts received bevacizumab 15 mg/kg IV,<br />

pertuzumab 420 mg IV (following an 840 mg loading dose) day 1 <strong>of</strong> a 21<br />

day cycle, and octreotide LAR 30 mg IM every 28 days. Primary endpoint<br />

was objective response rate (ORR). Results: Between 6/10 and 4/11, 43 pts<br />

enrolled. 22 M/21 F, med age 62 (33-88), 32 (74%) carcinoid, 11 (26%)<br />

PNET. 26% had prior sandostatin, 72% prior surgery, 19% prior radiation,<br />

23% with � 2 previous systemic therapies. Treatment-related grade 3<br />

toxicities included: hypertension (28%), LVEF dysfunction (9%) (reversible<br />

with trial drug hold), and diarrhea (7%). No grade 4 toxicity was<br />

reported. Median treatment duration was 38 weeks (range: 0 – 76).<br />

Chromogranin A values were elevated in 27 pts; 59% had decreases during<br />

treatment. Seven pts (16%) achieved objective response [CR: 1 carcinoid<br />

(2%), PR: 6 (4 carcinoid, 2 PNET) (14%)]. ORR for carcinoid pts was<br />

15.6%. Median PFS for the entire group was 8.2 months (95% CI: 6.3 –<br />

NA). PFS for carcinoid pts was 8.5 months (95% CI: 6.3 – NA); PFS for<br />

PNET pts was 6.4 months (95% CI: 3.9 – NA). Median OS has not been<br />

reached at 14.3 months followup. Conclusions: The ORR <strong>of</strong> the combination<br />

<strong>of</strong> bevacizumab, pertuzumab, and octreotide LAR in NET pts was<br />

encouraging. <strong>Part</strong>icularly the response rate in carcinoid pts appears higher<br />

than historical data, warranting further investigation <strong>of</strong> this regimen in<br />

these pts.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


270s Gastrointestinal (Noncolorectal) Cancer<br />

4128 General Poster Session (Board #51G), Mon, 8:00 AM-12:00 PM<br />

Prognostic relevance <strong>of</strong> circulating PIGF levels in patients with neuroendocrine<br />

tumors. Presenting Author: Christian Fischer, Charité-Universitätsmedizin,<br />

Berlin, Germany<br />

Background: Placental growth factor (PlGF), a VEGF homolog implicated in<br />

tumor angiogenesis and adaptation to antiangiogenic therapy, is emerging<br />

as candidate target in malignancies. As antiangiogenic treatments are<br />

introduced in the management <strong>of</strong> neuroendocrine tumors (NETs), we<br />

addressed the expression and function <strong>of</strong> PlGF in NETs. Methods: Serum<br />

levels <strong>of</strong> PlGF were determined in two independent cohorts <strong>of</strong> NET patients<br />

collected retrospectively and prospectively (n�87 and n�84) using Roche<br />

Elecsys and correlated with clinical data. Expression <strong>of</strong> PlGF in tumors was<br />

evaluated by immunohistochemistry. PlGF effects on proliferation and<br />

migration were examined in vitro using NET cell lines. Results: Circulating<br />

and tumoral PlGF were found elevated in NET patients as compared to<br />

control sera and pancreatic tissues in a retrospective analysis restricted to<br />

patients with pancreatic NETs (pNETs). De novo expression <strong>of</strong> PlGF<br />

occurred primarily in the tumor stroma, suggesting paracrine stimulatory<br />

circuits. Indeed, PlGF enhanced proliferation and migration <strong>of</strong> NET cells,<br />

and elevated circulating PlGF levels correlated with advanced tumor<br />

grading, suggesting that PlGF supported a more aggressive phenotype. In<br />

line with this notion, circulating PlGF levels above median predicted<br />

reduced tumor related survival in pNET patients (p�0.012). Furthermore,<br />

pathologic PlGF levels were associated with poor prognosis within the<br />

subgroup <strong>of</strong> grade 2 tumors. Subsequent prospective PlGF determinations<br />

confirmed and extended our observation <strong>of</strong> elevated PlGF levels for both,<br />

pNETs (n�32, p�0.001) and midgut NETs (n�58, p�0.001). Since<br />

median follow up was too short to determine survival, time to progression<br />

(TTP) was analyzed instead. Most pNETs were progressive at the time <strong>of</strong><br />

sample collection, precluding an informative evaluation <strong>of</strong> TTP in this<br />

subgroup. However, PlGF levels above median were correlated with shorter<br />

TTP in midgut NETs (p�0.0091; TTP 27 months versus undefined, HR<br />

3.802). Conclusions: These data assign to PlGF a novel function in the<br />

pathobiology <strong>of</strong> NETs and propose PlGF as a prognostic parameter in<br />

patients with NETs.<br />

4130 General Poster Session (Board #52A), Mon, 8:00 AM-12:00 PM<br />

Patients with carcinoma <strong>of</strong> unknown primary and “colon cancer pr<strong>of</strong>ile”:<br />

Clinicopathologic features and survival data. Presenting Author: Gauri R.<br />

Varadhachary, University <strong>of</strong> Texas M. D. Anderson Cancer Center, Houston,<br />

TX<br />

Background: We have previously proposed a “colon cancer pr<strong>of</strong>ile” (CCP-<br />

CUP) favorable subset. CCP-CUP is ~ 8% <strong>of</strong> all CUP with features<br />

resembling lower gastroinestional cancers. Distinguishing this entity from<br />

other CUP pts is <strong>of</strong> significance given the progress in the treatment <strong>of</strong><br />

metastatic colorectal cancer (CRC). Additionally, emerging data suggests a<br />

high level <strong>of</strong> agreement between histology�IHC and tissue <strong>of</strong> origin<br />

pr<strong>of</strong>iling for CCP-CUP. Methods: The retrospective cohort includes 74 pts<br />

from MD Anderson (50) and Sarah Cannon (24) Cancer Centers from 2004<br />

-2010. Pts with CDX2� tumors and pathology suggesting a �GI pr<strong>of</strong>ile�<br />

were chosen. All pts met the definition <strong>of</strong> CUP and most had a negative<br />

colonoscopy. Results: 2 cohorts were created - Cohort 1 (34 pts), labeled<br />

“consistent with lower GI pr<strong>of</strong>ile” [CDX2�, CK20�, CK7-] and Cohort 2<br />

(40 pts), labeled “probable lower GI pr<strong>of</strong>ile” [CDX2�, irrespective <strong>of</strong><br />

CK7/CK20 status]. Most pts had a good PS; 58% women, median age 59<br />

yrs; 20 (27%) pts had ascites on presentation and the predominant sites <strong>of</strong><br />

metastases included liver (30%), carcinomatosis (50%), and nodes (51%).<br />

53 pts received first line CRC regimens (FOLFOX or FOLFIRI based), 15 pts<br />

received gemcitabine or taxane based and 3 ‘other’ regimens. OS was 37<br />

mo (C.I 22- 52). 6 <strong>of</strong> these were �outliers� (Stage 4 NED or indolent<br />

pathology). Excluding these, cohorts 1 and 2 had 32 and 36 pts - their OS<br />

were 37 and 21 mo respectively. There was no difference in OS <strong>of</strong> pts with<br />

or without ascites on presentation. Kras data was available for 17; 12 were<br />

Kras mutant. On multivariate analysis, the factors found to negatively<br />

influence survival were age, PS (<strong>of</strong> 2) and 3 or more sites <strong>of</strong> disease.<br />

Conclusions: Survival <strong>of</strong> IHC defined CCP-CUP pts (which may include<br />

colorectal, appendiceal and small bowel pr<strong>of</strong>ile) exceeds historical control<br />

and illustrates a new �favorable� subset. IHC is not without its limitations –<br />

nonetheless, pts with CDX2 � tumors and CUP should undergo evaluation<br />

for GI cancers and likely best served with an armamentarium <strong>of</strong> drugs used<br />

for CRC. Since carcinomatosis and liver mets are predominant sites, all<br />

patients with abdominal CUP should undergo optimal IHC (CDX-2, CK7,<br />

and CK20) testing to rule out a CCP-CUP.<br />

4129 General Poster Session (Board #51H), Mon, 8:00 AM-12:00 PM<br />

Well-differentiated grade 3 digestive neuroendocrine tumors: Myth or<br />

reality? The PRONET study group. Presenting Author: Jean-Yves Scoazec,<br />

Hôpital Edouard Herriot, Lyon, France<br />

Background: In contrast to the 2000 World Health Organization (WHO)<br />

classification <strong>of</strong> digestive neuroendocrine tumors (NET) in which morphologic<br />

differentiation was the first criterion, the 2010 WHO classification <strong>of</strong><br />

NET is based mostly on histologic grade. NET are now classified into three<br />

main categories: NET G1 (mitotic count �2/10 HPF and/or �2% Ki67<br />

index), NET G2 (2-20/10 HPF and/or 3-20%), and neuroendocrine<br />

carcinoma (NEC) <strong>of</strong> small or large cell type. While NET G1 and G2 are<br />

well-differentiated tumors, NEC are considered poorly differentiated G3<br />

tumors. We looked at the agreement between grade and differentiation to<br />

determine whether all NET can be readily classified according to the 2010<br />

WHO classification. Methods: We designed a 1-year prospective, epidemiologic<br />

study to assess the characteristics <strong>of</strong> newly diagnosed NET, including<br />

diagnostic pathology. From August 2010 to July 2011, all pathology<br />

laboratories in France were invited to register all incident cases <strong>of</strong><br />

gastroenteropancreatic (GEP) and thoracic NET, excluding small cell<br />

carcinoma. For GEP-NET, investigators were asked to indicate morphologic<br />

differentiation (according to WHO 2000) and elements <strong>of</strong> histologic grade<br />

(mitotic index, Ki67 index), according to ENETS. Results: Of 500 invited<br />

centers, 80 participated; 1417 incidental cases were included and 77<br />

excluded (duplicates or exclusion criteria), totaling 1340 cases; 778<br />

(58.1%) were GEP-NET; 660/778 (85%) were well differentiated, 72 (9%)<br />

poorly differentiated, and 46 (6%) adenocarcinoid, nonclassified, or not<br />

evaluable; 422 (54.2%) were G1, 220 (28%) G2, 104 (13.5%) G3, and<br />

32 (4.1%) had missing grades. Of those deemed G3, 72 (69%) were<br />

described as poorly differentiated, 21 (20%) as well differentiated (mean<br />

Ki67 index 35%, range 25%-60%), and 11 (10.5%) as adenocarcinoid.<br />

Conclusions: In this prospective, epidemiologic study, overall agreement<br />

between grade and differentiation was good. However, a significant<br />

proportion <strong>of</strong> G3 NET were classified as well differentiated and thus<br />

unclassifiable by 2010 WHO classification. This group <strong>of</strong> tumor deserves to<br />

be included in future classifications to help the clinician decide whether<br />

they should be treated as NET G1/G2 or NEC G3.<br />

4131 General Poster Session (Board #52B), Mon, 8:00 AM-12:00 PM<br />

Mutation pr<strong>of</strong>ling in patients with carcinoma <strong>of</strong> unknown primary using the<br />

Sequenom MassARRAY system. Presenting Author: Katherine Stemke<br />

Hale, University <strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: CUP management may be viewed as an epitome <strong>of</strong> personalised<br />

medicine as we apply our understanding <strong>of</strong> biological markers to this<br />

heterogeneous disease. There is a significant interest in evaluating actionable<br />

mutations in various signaling pathways, receptors and downstream<br />

effectors in CUP that may help us understand its biology and serve as the<br />

basis for unique targeted therapeutic approaches. Sequenom (SQM)<br />

Massarray platform enables rapid mutation pr<strong>of</strong>iling in solid tumors which<br />

we applied to CUP. Methods: 83 CUP samples were sent for DNA mass array<br />

analysis. Sequenom was run on 60 samples; 23 samples lacked sufficient<br />

quantity or quality <strong>of</strong> DNA. Data on clinicopathological features was<br />

collected. Results: Of the 60 patient samples run on SQM, 16 patients had<br />

changes including 11 mutations and 6 polymorphisms . The 6 polymorphisms<br />

were in MET . The mutations reported were :RAS (7; 6 KRAS, 1<br />

NRAS), IDH1 (2), PI3K (1) and MET (1). Atleast 50% <strong>of</strong> the tumors were<br />

poorly differentiated. Interestingly, the 2 patients with IDH1 mutations<br />

presented with osseous predominant metastatic disease. Of the 6 patients<br />

with KRAS mutations, 4 presented with a gastrointestinal pr<strong>of</strong>ile ( CDX2�<br />

or CK20�) on IHC; in all 4, the DNA sequencing analysis results matched<br />

the sequenom results. Conclusions: The (all-comers) overall mutation rate<br />

in a heterogenous CUP population is low (11/60, 18%). No �new� low<br />

frequency mutations were found using the current panel (Table). Rare and<br />

potentially targetable mutations may be identified with a larger panel.<br />

Focusing on CUP subsets using this approach may also provide a higher<br />

mutational yield.<br />

P13K/AKT pathway MEK pathway Receptors Downstream effectors<br />

AKT1, 2, 3 BRAF EGFR CDK4<br />

PIK3CA KRAS ALK CTNNB1<br />

PHLPP2 MEK1,2 KIT IDH1,2<br />

FRAP (mTOR) NRAS MET JAK2<br />

RICTOR PRKAG1/2 PDGFRA RET<br />

PDPK1 BRAF FLT3<br />

PIK3R1 TNK2(ACK1)<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


4132 General Poster Session (Board #52C), Mon, 8:00 AM-12:00 PM<br />

Final results <strong>of</strong> a phase Ib study <strong>of</strong> tivozanib and FOLFOX6 in patients (pts)<br />

with advanced gastrointestinal (GI) tumors. Presenting Author: Ferry<br />

Eskens, Erasmus University Medical Center, Daniel den Hoed Cancer<br />

Center, Rotterdam, Netherlands<br />

Background: Tivozanib is a potent, selective, oral small-molecule tyrosine<br />

kinase inhibitor <strong>of</strong> vascular endothelial growth factor receptors 1, 2 and 3<br />

with a long half-life. A Phase I study found tivozanib’s maximum tolerated<br />

dose (MTD) to be 1.5 mg/d and responses were observed in pts with<br />

colorectal cancer (CRC) and other tumors. Tivozanib has shown additive<br />

anti-tumor activity with 5-fluorouracil in a preclinical breast tumor model.<br />

FOLFOX6 is a standard chemotherapy for GI cancers. This open-label,<br />

Phase Ib study (NCT00660153) sought to determine the MTD, doselimiting<br />

toxicities (DLTs), pharmacokinetics (PK), and anti-tumor activity<br />

<strong>of</strong> escalating doses <strong>of</strong> tivozanib combined with a modified (m) FOLFOX6<br />

regimen (85 mg/kg2 oxaliplatin) in pts with advanced GI tumors. Methods:<br />

Tivozanib was administered once daily in 4-week cycles (3 weeks on, 1<br />

week <strong>of</strong>f) with mFOLFOX6 administered on Days 1 and 15 <strong>of</strong> each cycle.<br />

Pts also received a single dose <strong>of</strong> tivozanib on day -5 for PK analysis. Pts<br />

were allowed to continue tivozanib following discontinuation <strong>of</strong> mFOL-<br />

FOX6. Results: Twenty two pts (14:8 male:female; median age 58 years;<br />

91% Caucasian) received tivozanib 0.5 mg (n�9), 1.0 mg (n�3), or 1.5<br />

mg (n�10) and mFOLFOX6. Pts received a median <strong>of</strong> 5.2 (range 0.0 to<br />

25.1) months <strong>of</strong> treatment. DLTs were observed in 2 pts on tivozanib 0.5<br />

mg (reversible Grade [Gr] 3 diarrhea and Gr 3/4 transaminase elevations)<br />

and in 2 pts on tivozanib 1.5 mg (Gr 3 seizure and reversible Gr 3 vertigo).<br />

Other Gr 3/4 drug-related adverse events (AEs) included neutropenia,<br />

fatigue, and hypertension (n�2 each). Eight pts discontinued treatment<br />

due to AEs. The MTD for tivozanib with mFOLFOX6 was 1.5 mg. The<br />

disease control rate was 63% (�1% complete response, 27% partial<br />

response, 36% stable disease). Preliminary PK data showed no interaction<br />

between tivozanib and mFOLFOX6. Eight additional pts enrolled at the 1.5<br />

mg dose level are currently being evaluated. Final results will be presented.<br />

Conclusions: Tivozanib can be combined at its recommended dose <strong>of</strong> 1.5<br />

mg/day with mFOLFOX6 for pts with advanced GI tumors. The combination<br />

was tolerable and showed encouraging anti-tumor activity. A Phase II study<br />

<strong>of</strong> this combination for mCRC is ongoing.<br />

TPS4134 General Poster Session (Board #52E), Mon, 8:00 AM-12:00 PM<br />

Dasatinib combined with gemcitabine (Gem) in patients (pts) with locally<br />

advanced pancreatic adenocarcinoma (PaCa): Design <strong>of</strong> CA180-375, a<br />

placebo-controlled, randomized, double-blind phase II trial. Presenting<br />

Author: T.R. Jeffry Evans, Beatson West <strong>of</strong> Scotland Cancer Centre,<br />

Glasgow, United Kingdom<br />

Background: Dasatinib, a potent oral BCR-ABL and SRC family kinase<br />

(SFK) inhibitor, is approved for first- and second-line therapy <strong>of</strong> Philadelphia<br />

chromosome-positive chronic phase chronic myeloid leukemia (CML)<br />

in pts with newly diagnosed CML or CML resistant/intolerant to prior<br />

therapy. SRC expression and activity is upregulated in PaCa and correlates<br />

with reduced survival in resected high-grade PaCa (Morton, Gastroenterology<br />

2010) and resistance to Gem, a PaCa standard <strong>of</strong> care (Duxbury, J Am<br />

Coll Surg 2004). In preclinical PaCa studies, inhibition <strong>of</strong> SFKs with<br />

dasatinib reduces tumor cell proliferation, migration, and invasion; increases<br />

apoptosis; sensitizes cells to Gem; and inhibits development <strong>of</strong><br />

metastases in vivo either alone or in combination with Gem (Duxbury, Clin<br />

Cancer Res 2004; Duxbury, J Am Coll Surg 2004; Nagaraj, Mol Cancer<br />

Ther 2010; Morton, op cit). Phase I clinical studies <strong>of</strong> dasatinib and Gem<br />

therapy in PaCa have demonstrated feasibility and suggested efficacy <strong>of</strong> the<br />

combination (Uronis, ASCO 2009, abstract e15506). Methods: This<br />

double-blind phase II study tests whether addition <strong>of</strong> dasatinib to Gem is<br />

tolerable and improves efficacy in pts with histologically/cytologically<br />

confirmed unresectable locally advanced nonmetastatic PaCa. Eligible pts,<br />

aged �18 years with Eastern Cooperative Oncology Group performance<br />

status �1 and adequate organ function, are randomized 1:1 to Gem 1000<br />

mg/m2 IV once weekly (Weeks 1–3 <strong>of</strong> a 4-week cycle) plus either dasatinib<br />

100 mg once daily or matched placebo. Pts are treated until progression,<br />

unacceptable toxicity, withdrawal <strong>of</strong> consent, or study termination. The<br />

primary endpoint is OS, and secondary endpoints are progression-free<br />

survival and safety. Exploratory endpoints include freedom from distant<br />

metastases, measures <strong>of</strong> pain and fatigue, overall response rate, and<br />

carbohydrate antigen 19-9. Final study analysis will be conducted after<br />

135 deaths; all pts will be followed for survival. To date, 23/200 pts have<br />

enrolled; estimated primary completion date is March 2013. <strong>Clinical</strong>Trials.<br />

gov identifier: NCT01395017.<br />

Gastrointestinal (Noncolorectal) Cancer<br />

271s<br />

4133 General Poster Session (Board #52D), Mon, 8:00 AM-12:00 PM<br />

Methylguanine DNA methyl transferase (MGMT) expression to predict<br />

response to temozolomide (TMZ) in patients with digestive neuroendocrine<br />

tumors (NETs). Presenting Author: Pascal Hammel, Beaujon, Paris, France<br />

Background: TMZ is an orally given drug used in patients (pts) with NETs,<br />

with encouraging results in pancreatic NETs in a recent study (Strosberg et<br />

al, 2011). Higher treatment efficacy seems to be correlated to MGMT<br />

tumor deficiency (Kulke et al, 2010). Aim-To assess MGMT expression in<br />

digestive NETs and to correlate with the efficacy <strong>of</strong> TMZ-based therapies.<br />

Methods: All pts with well differentiated, progressive, non resectable NETs<br />

treated with TMZ-based chemotherapy were included. Treatment efficacy<br />

was defined according to RECIST. Pts with progression or only stable<br />

disease were considered as “non-responders”. Nuclear expression <strong>of</strong><br />

MGMT was assessed by immunohistochemistry on primary tumors or<br />

metastases and graded according the product <strong>of</strong> the intensity <strong>of</strong> staining (0<br />

to 3) and the rate <strong>of</strong> positive cells (%), leading to a score comprised<br />

between 0 and 300. A score � 80 was defined as “high” staining. Results:<br />

22 pts (age 59 years (36-81)) with pancreatic (14 pts), small bowel (5 pts)<br />

or other (3 pts) NETs, grade 1 (5 pts) or 2 (17 pts) (WHO 2010<br />

classification) were included. They received TMZ alone (19 pts) or<br />

combined with capecitabine (3 pts) as first line (3 pts) or 2� line (19<br />

pts).After a median <strong>of</strong> 6 cycles (3-16), objective response, stable disease<br />

and progression rates were seen in 32 % (7 pts, all with pancreatic NETs),<br />

41% (9 pts, 5 pancreatic) and 27% (6 pts, 4 small bowel), respectively.<br />

Median (range) MGMT score was 10 (0-300). A “High” MGMT score was<br />

seen in 36% <strong>of</strong> pts (small bowel 4, pancreas 3 pts) ; it was correlated with<br />

primary tumor location (more frequent in small bowel NETs, p�0.02) and<br />

predictive <strong>of</strong> the absence <strong>of</strong> response (p�0.02). A “Low “MGMT score<br />

tended to be associated with objective response (p�0.06) whereas none <strong>of</strong><br />

the pts with “high” score had tumor response. Response rate in pts with<br />

“low” MGMT score was 50%. Conclusions: MGMT deficiency is more<br />

frequent in pancreatic than small bowel NETs. Patients with pancreatic<br />

NET and low MGMT score are good candidates for TMZ, whereas those with<br />

high score should be treated with other drug in first intention. In patients<br />

with small bowel NET with most <strong>of</strong>ten high MGMT score, tumor stabilization<br />

using TMZ seems to be rare.<br />

TPS4135 General Poster Session (Board #52F), Mon, 8:00 AM-12:00 PM<br />

A phase III, multicenter, randomized, double-blind, placebo-controlled<br />

trial <strong>of</strong> ganitumab or placebo in combination with gemcitabine (G) as<br />

first-line therapy for metastatic adenocarcinoma <strong>of</strong> the pancreas: The<br />

GAMMA trial. Presenting Author: Charles S. Fuchs, Dana-Farber Cancer<br />

Institute, Boston, MA<br />

Background: Ganitumab (AMG 479) is an investigational, fully human,<br />

monoclonal antibody inhibitor <strong>of</strong> the IGF1R. In a randomized phase II study<br />

in patients with metastatic pancreatic cancer, addition <strong>of</strong> ganitumab 12<br />

mg/kg every 2 weeks (Q2W) to G prolonged progression-free survival (PFS)<br />

and overall survival (OS) (Kindler. Ann Onc. 2010;21:741P). Exposureefficacy<br />

analysis showed that patients with higher ganitumab exposure<br />

levels (AUCss at or above median) had longer PFS and OS (Lu. JCO.<br />

2011;29:4049). Methods: In this phase III study, patients are randomized<br />

2:2:1 to placebo � G, ganitumab 12 mg/kg � G, or ganitumab 20 mg/kg �<br />

G. Patients receive ganitumab or placebo IV days (D) 1 and 15 and G 1000<br />

mg/m2 IV D 1, 8, and 15 every 28 D. Patients receiving 20 mg/kg<br />

ganitumab are expected to achieve ganitumab levels above the median in<br />

phase 2. Key eligibility criteria: untreated metastatic adenocarcinoma <strong>of</strong><br />

the pancreas; ECOG score 0 or 1; � 18 years old; adequate organ function;<br />

and fasting (or non-fasting) glucose � 160 mg/dL. The primary endpoint is<br />

OS. A log-rank test stratified by ECOG, presence <strong>of</strong> liver metastases, and<br />

geographic region will compare OS independently for each ganitumab arm<br />

at an overall one-sided 2.5% significance level for declaring superiority <strong>of</strong><br />

ganitumab � G vs placebo � G. Key secondary endpoints include PFS,<br />

objective response, safety, and patient-reported outcomes. An additional<br />

objective is to define a subpopulation with improved OS based upon<br />

baseline levels <strong>of</strong> circulating biomarkers. Enrollment began in April 2011.<br />

As <strong>of</strong> January 23, 2012, 463 <strong>of</strong> 825 patients have been enrolled. The study<br />

is overseen by an independent data monitoring committee. Status: open.<br />

Supported by Amgen Inc. in collaboration with Takeda Global Research &<br />

Development Center, Inc.; <strong>Clinical</strong>Trials.gov: NCT01231347.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


272s Gastrointestinal (Noncolorectal) Cancer<br />

TPS4136 General Poster Session (Board #52G), Mon, 8:00 AM-12:00 PM<br />

Gemcitabine with nab-paclitaxel in neoadjuvant treatment <strong>of</strong> pancreatic<br />

adenocarcinoma: GAIN-1 study. Presenting Author: Neelam Vijay Desai,<br />

University <strong>of</strong> Florida, Gainesville, FL<br />

Background: Pancreatic adenocarcinoma remains a deadly disease with a<br />

dismal prognosis. Only 15% <strong>of</strong> patients present with resectable disease<br />

with 5-year survival only 20% despite adjuvant therapy. Neoadjuvant<br />

treatment may improve R0 resection rates, allows more patients curative<br />

surgery, is cost effective, and may improve overall survival by incorporating<br />

earlier systemic therapy. Methods: This is a prospective single arm<br />

open-label phase II trial using gemcitabine (G) and nab-paclitaxel (A) in the<br />

neoadjuvant treatment <strong>of</strong> resectable and borderline-resectable pancreatic<br />

cancer. Patients are stratified by risk group as defined by a predictive model<br />

published by Bao et al and the NCCN expert consensus statement (Table):<br />

low-risk resectable (Group 1) and high-risk resectable or borderlineresectable<br />

(Group 2). Group 1 receives 3 cycles <strong>of</strong> G�A then surgical<br />

resection. 1 cycle � G 1000mg/m2 �A 100mg/m2 IV on Days 1, 8, 15 Q28<br />

days. Group 2 receives 2 cycles <strong>of</strong> G�A followed by 2 weeks <strong>of</strong> hyp<strong>of</strong>ractionated<br />

IMRT (50.4Gy in 10 fx) with G 400mg/m2 IV weekly, then surgical<br />

resection. The primary endpoints are R0 resection rate and overall response<br />

rates. Secondary endpoints are PFS, OS, 30-day post-op mortality, toxicity,<br />

QOL and molecular analysis including DPC4 and SPARC levels in tumor<br />

and stroma. Enrollment has just begun with a total accrual goal <strong>of</strong> 60<br />

patients. This platform will allow targeted systemic therapy and tailored<br />

treatment stratification to optimize outcomes in curable pancreatic cancer.<br />

This study is registered with <strong>Clinical</strong>trials.gov (NCT01470417).<br />

Risk stratification Definition<br />

Low-risk -EUS Stage � IA OR<br />

-No vascular involvement on CT &<br />

a. EUS stage IB or IIA OR<br />

b. EUS stage � IIB but tumor � 2.6cm<br />

High-risk -EUS stage IB or IIA but vascular involvement on CT<br />

-No vascular involvement but EUS stage � IIB & tumor � 2.6 cm<br />

-(�) nodes on PET or FNA<br />

Borderline -Involvement <strong>of</strong> SMV/portal vein but w/o encasement <strong>of</strong> arteries, or<br />

short segment venous occlusion 2/2 tumor thrombus<br />

or encasement but w/ suitable vessels to allow<br />

resection & reconstruction<br />

-Gastroduodenal artery encasement up to hepatic artery<br />

w/ short segment involvement <strong>of</strong> hepatic artery,<br />

w/o extension to celiac axis<br />

-Tumor abutment <strong>of</strong> SMA �180 degrees <strong>of</strong> vessel wall<br />

TPS4138 General Poster Session (Board #53A), Mon, 8:00 AM-12:00 PM<br />

PaFLO: Pazopanib with 5-fluorouracil, leucovorin, and oxaliplatin (FLO) as<br />

first-line treatment in advanced gastric cancer: A randomized phase II<br />

study <strong>of</strong> the Arbeitsgemeinschaft internistische Onkologie (AIO). Presenting<br />

Author: Kirstin Breithaupt, Charité-Universitätsmedizin Berlin, Campus<br />

Virchow-Klinikum, Med Klinik m. S. Hämatologie u. Onkologie, Berlin,<br />

Germany<br />

Background: VEGF inhibition in gastric cancer shows promising improvement<br />

<strong>of</strong> remission rate and progression-free survival (Ohtsu et al., JCO<br />

2011). Pazopanib is an orally available tyrosine kinase inhibitor (TKI)<br />

selectively inhibiting VEGFR-1, -2, -3, c-kit and PDGFR. It is approved for<br />

treating renal cell cancer. A phase-I trial showed good tolerability <strong>of</strong><br />

pazopanib with full-dose FOLFOX in solid tumors (Brady et al., ASCO,<br />

2009). FLO is a widely used combination for advanced gastric cancer<br />

recommended in national guidelines. Methods: 75 Patients with HER-2negative<br />

locally advanced or metastatic adenocarcinoma <strong>of</strong> the stomach or<br />

the gastro-esophageal junction will be randomized in a 2:1 ratio to A: FLO<br />

(F 2600mg/m2 as 24h infusion, L 200mg/m2, O 85 mg/m2) d1 �<br />

pazopanib (800mg) d1-14 and B: FLO; repeated for 12 2-week cycles,<br />

followed by a maintenance therapy with pazopanib alone in A and an<br />

observation period in B until disease progression. Primary endpoint is<br />

progression-free survival rate (PFSR) at 6 months, secondary endpoints are<br />

PFSR at 9 and 12 months, median PFS, response rate, duration <strong>of</strong><br />

response, toxicity, tolerability and overall survival. Additionally, we evaluate<br />

the predictive and prognostic relevance <strong>of</strong> PIGF, VEGF, and the<br />

respective soluble receptors sVEGFR1 and sVEGFR2 as biomarkers for<br />

clinicopathological parameters, clinical response to treatment and tumor<br />

volume change. Based on a phase-III trial demonstrating a 6-month PFSR<br />

<strong>of</strong> 44% with FLO (Al-Batran et al., 2008), we estimate a 6-month PFSR <strong>of</strong><br />

55% in the experimental group. Given an alpha error <strong>of</strong> 0.1 and a beta error<br />

<strong>of</strong> 0.2 in a Simon 2-stage minimax design, in the first stage �12 <strong>of</strong> 30<br />

patients need to be progression free at 6 months to continue and after the<br />

second stage �25 <strong>of</strong> 50 patients should be progression free at 6 months to<br />

justify further evaluation. Randomization is performed to estimate selection<br />

bias according to pazopanib-specific exclusion criteria for comparison<br />

with historical data. Study protocol received ethics committee approval in<br />

November 2011 and is currently recruiting patients in 15 AIO centers.<br />

TPS4137 General Poster Session (Board #52H), Mon, 8:00 AM-12:00 PM<br />

PAN1: A randomized phase II study evaluating potential predictive biomarkers<br />

in the treatment <strong>of</strong> metastatic pancreatic cancer. Presenting Author: Yu<br />

Jo Chua, Medical Oncology Unit, The Canberra Hospital, Garran, Australia<br />

Background: Biomarker-based selection <strong>of</strong> cancer treatments has already<br />

entered routine clinical practice in some cancer types. Recent retrospective<br />

data suggests the human equilibrative nucleoside transporter 1 (hENT1) to<br />

be promising predictive marker for benefit from gemcitabine in pancreatic<br />

cancer, at least in resected disease. Methods: 80 patients will be randomised<br />

to receive either gemcitabine or FOLFOX chemotherapy (folinic<br />

acid (FOL) fluorouracil (F) and oxalipatin (OX)) with stratification based on<br />

hENT1 status. All patients will be required to have tumour samples tested<br />

for hENT1 prior to randomisation, with both hENT1 positive and negative<br />

patients eligible for inclusion. Patients will continue their assigned<br />

treatment until progression, or unacceptable toxicity. Primary endpoint:<br />

Progression free survival in each chemotherapy arm (FOLFOX/gemcitabine).<br />

Secondary endpoints: efficacy/activity <strong>of</strong> gemcitabine and FOLFOX<br />

as assessed by OS (overall survival), TTP (time to progression), RR<br />

(response rate according to RECIST v1.1), CA19.9 response; Percentage <strong>of</strong><br />

eligible patients able to be randomised within 14 days <strong>of</strong> screening; PFS in<br />

each biomarker-selected cohort (hENT1 positive/negative); Treatment<br />

related toxicity in each chemotherapy group (CTCAE v4.0) and in biomarker<br />

cohorts (hENT1 positive/negative); establishment <strong>of</strong> a tissue bank from<br />

patients treated with and without gemcitabine for further biomarker<br />

studies. Eligibility: Adult patients with radiologically and histologically<br />

confirmed metastatic pancreatic adenocarcinoma who have not previously<br />

received treatment for metastatic disease, and who have tumour tissue<br />

available for hENT1 testing are eligible for the study. Patients who do not<br />

have tumour tissue available will be required to undergo core biopsy to<br />

obtain tissue for hENT1 testing. Status: Opened to accrual July 2011.<br />

TPS4139 General Poster Session (Board #53B), Mon, 8:00 AM-12:00 PM<br />

A randomized, multicenter, double-blind, placebo (PBO)-controlled phase<br />

III study <strong>of</strong> paclitaxel (PTX) with or without ramucirumab (IMC-1121B;<br />

RAM) in patients (pts) with metastatic gastric adenocarcinoma, refractory<br />

to or progressive after first-line therapy with platinum (PLT) and fluoropyrimidine<br />

(FP). Presenting Author: Hansjochen Wilke, Kliniken Essen Mitte<br />

Center <strong>of</strong> Palliative Care, Essen, Germany<br />

Background: Vascular endothelial growth factor (VEGF) expression in gastric<br />

cancer (GC) is associated with more aggressive clinical disease. VEGF<br />

expression in resected GC is associated with tumor recurrence and shorter<br />

survival. Data from Phase 2 and 3 studies suggest that agents targeting the<br />

VEGF pathway improve the efficacy <strong>of</strong> some chemotherapy regimens in 1stand<br />

2nd-line treatment <strong>of</strong> patients with gastric or gastroesophageal<br />

carcinomas. RAM, a fully human monoclonal antibody, binds to the VEGF<br />

receptor-2 (VEGFR-2), potently blocks the binding <strong>of</strong> VEGF to VEGFR-2,<br />

inhibits VEGF-stimulated activation <strong>of</strong> VEGFR-2, and neutralizes VEGFinduced<br />

mitogenesis <strong>of</strong> human endothelial cells. Methods: Pts are randomized<br />

1:1 to receive PTX � RAM or PTX � PBO until disease progression or<br />

intolerable toxicity (28-day cycle; RAM/PBO 8 mg/kg Days 1, 15; PTX 80<br />

mg/m2 Days 1, 8, 15). Eligibility includes metastatic or locally advanced,<br />

unresectable gastric or gastroesophageal junction adenocarcinoma; prior<br />

first-line therapy with any PLT/FP doublet with or without anthracycline;<br />

progressive disease during or following first-line therapy; ECOG PS 0-1;<br />

bilirubin � 1.5 � upper limit <strong>of</strong> normal (ULN), transaminases � 3 � ULN<br />

for ALAT/ASAT if no liver metastases, � 5 � ULN if liver metastases;<br />

creatinine � 1.5 � ULN; absolute neutrophil count � 1.5 � 109 /L,<br />

hemoglobin � 9 g/dL; platelets � 100 � 109 /L. The primary endpoint is<br />

overall survival (OS). Secondary endpoints include progression-free survival,<br />

time to progression, best overall response, objective response rate,<br />

safety, patient-reported outcome measures, pharmacodynamics, immunogenicity,<br />

and pharmacokinetics. This study, powered at 90% to show an<br />

increase in OS (mdn: 7mPTX�PBO, 9.33 m PTX � RAM) at a 1-sided<br />

2.5% significance level, will randomize 663 pts. As <strong>of</strong> 18 January 2012,<br />

approximately 58% <strong>of</strong> planned pts were randomized. The IDMC reviewed<br />

this study 23 June and 01 December 2011 and recommended the study<br />

continue unmodified.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


TPS4140 General Poster Session (Board #53C), Mon, 8:00 AM-12:00 PM<br />

Lapatinib in combination with ECF/x in EGFR1 positive first-line metastatic<br />

gastric cancer (GC): A phase II randomized placebo controlled trial (EORTC<br />

40071). Presenting Author: Markus Hermann Moehler, University <strong>of</strong><br />

Mainz, Mainz, Germany<br />

Background: Survival <strong>of</strong> HER2� metastatic GC is prolonged by trastuzumab<br />

when administered with CF/X (VanCutsem, ASCO 2009). Lapatinib inhibits<br />

both EGFR1 and HER2, is active in HER2� GC lines, and has shown<br />

clinical activity in uncontrolled phase II GC trials. A phase III trial <strong>of</strong><br />

lapatinib with X � oxaliplatin in HER2� (FISH) GC is closed to recruitment.<br />

Additional unaddressed questions include the efficacy and safety <strong>of</strong><br />

lapatinib with ECF/X (epirubicin � cisplatin � 5-FU or capecitabine (X),<br />

which is a preferred chemotherapy (CT) regimen in GC), and its activity in<br />

patients (pts) with discordant FISH or IHC HER2 status or EGFR1�.<br />

Methods: This is a phase II, randomized, double- blind, placebo controlled,<br />

multicenter trial sponsored by the EORTC. About 480 pts with adenocarcinoma<br />

<strong>of</strong> the stomach or esophagogastric junction not amenable to curative<br />

surgery and without prior palliative CT are screened centrally for HER2/<br />

EGFR1 by FISH and IHC. Patients are enrolled into one <strong>of</strong> two strata: 1)<br />

HER2 FISH- and IHC 2/3�, or 2) HER2 IHC 0/� and EGFR1 FISH� or IHC<br />

2/3�. Pts HER2 FISH�/IHC 2/3� and pts without HER2/EGFR1 by<br />

FISH/IHC will be excluded. 168 pts are anticipated to be randomized to<br />

lapatinib 1,250 mg cont. until progression or placebo, administered 6<br />

cycles <strong>of</strong> ECF or ECX (72/96 in stratum 1/2, respectively).The primary<br />

endpoint is progression-free survival (PFS) in stratum 2 and 77 events are<br />

needed for 80% power to detect an increase in PFS from 4 to 6.5 months<br />

with lapatinib (HR�0.615, one-sided alpha 10%). Secondary endpoints<br />

include PFS, toxicity, response rate, overall survival, and correlation <strong>of</strong><br />

HER2/EGFR1 status with response. Currently, half <strong>of</strong> all screened patients<br />

(19/38) have been randomized. So far, 8/38 (21%) pts were HER2�<br />

according TOGA criteria. By FISH or IHC, 14/38 were EGFR1�, with 4/14<br />

pts double HER2/EGFR�. Enrolment continues in 5 centers with about<br />

4-10 patients per month. A safety cohort analysis will be performed in the<br />

first 15 pts receiving lapatinib. Conclusions: This is the first trial to analyze<br />

prospectively and separately the role <strong>of</strong> lapatinib combined with chemotherapy<br />

in EGFR1� GC pts stratified by FISH/ IHC.<br />

TPS4142 General Poster Session (Board #53E), Mon, 8:00 AM-12:00 PM<br />

Postoperation chemotherapy with S1 and docetaxel in curatively resected<br />

gastric cancer <strong>of</strong> stage III (POST trial). Presenting Author: Minkyu Jung,<br />

Gachon University Gil Hospital, Incheon, South Korea<br />

Background: Complete surgical resections remains the only chance for cure<br />

in patients with gastric cancer, but approximately from 40% to 80% <strong>of</strong><br />

patients still have recurrences and most patients ultimately die from their<br />

disease. The recent adjuvant trials in gastric cancer showed significantly<br />

improved survival in patients with adjuvant chemotherapy than those with<br />

surgery alone. However, further studies need for the effect <strong>of</strong> adjuvant<br />

chemotherapy following D2 dissected gastric cancer patients, especially in<br />

advanced gastric cancer. S-1 is an oral anticancer drug, a prodrug <strong>of</strong><br />

fluorouracil, very effective in gastric cancer. Docetaxel is the first drug that<br />

showed survival benefits when added to the two drugs in advanced gastric<br />

cancer patients. And docetaxel is also synergistic anti-cancer effect with<br />

S-1 in advanced gastric cancer. Base on this background, the aim <strong>of</strong> this<br />

study is to detect a significant increase in 3 –year disease free survival<br />

(DFS) <strong>of</strong> adjuvant chemotherapy with docetaxel and S-1(DS) relative to<br />

those with S-1 and cisplatin (SP) in patients with stage III gastric cancer<br />

Methods: This study is an open-label, phase 3, randomized controlled trial,<br />

multicenter in South Korea. Patients with stage III (AJCC 7th edition)<br />

gastric cancer who had had curative D2 gastrectomy is randomly assigned<br />

to receive adjuvant chemotherapy <strong>of</strong> eight 3-week cycles <strong>of</strong> intravenous<br />

docetaxel (35 mg/m2 on day 1 and 8 <strong>of</strong> each cycle) plus oral S-1 (35 mg/m2 twice daily on days 1 to 14 <strong>of</strong> each cycle) for 6 months (DS) or<br />

chemotherapy <strong>of</strong> eight 3-week cycles <strong>of</strong> oral S1 plus intravenous cisplatin<br />

(60 mg/m2 ). After satisfying the screening criteria, patients have been<br />

randomized to the SD or SP arm in a 1:1 ratio. The randomization is<br />

stratified by institution and stage <strong>of</strong> disease (IIIA vs. IIIB vs. IIIC). The each<br />

stratum has been randomized by using the method <strong>of</strong> randomly permuted<br />

block. The primary endpoint is 3 year DFS, will analyze by intention to treat.<br />

A total <strong>of</strong> 290 patients will be enrolled, 67 patients have been treated to<br />

day, with continuing accrual. The trial is registered at <strong>Clinical</strong>Trials.gov<br />

(NCT01283217).<br />

Gastrointestinal (Noncolorectal) Cancer<br />

273s<br />

TPS4141 General Poster Session (Board #53D), Mon, 8:00 AM-12:00 PM<br />

TOPGEAR: An international randomized phase III trial <strong>of</strong> preoperative<br />

chemoradiotherapy versus preoperative chemotherapy for resectable gastric<br />

cancer (AGITG/TROG/EORTC/NCIC CTG). Presenting Author: Trevor<br />

Leong, Peter MacCallum Cancer Centre, Melbourne, Australia<br />

Background: Optimal management <strong>of</strong> patients with resectable gastric<br />

cancer remains unknown. Since the INT0116 and MAGIC trials, there are 2<br />

standards <strong>of</strong> care for adjuvant therapy: postoperative chemoradiotherapy<br />

(CRT) and perioperative ECF chemotherapy. The important question arising<br />

from these studies is whether CRT is superior to chemotherapy alone as<br />

adjuvant therapy. This randomized phase II/III trial will compare CRT to<br />

chemotherapy alone in the preoperative setting. Methods: Patients with<br />

resectable adenocarcinoma <strong>of</strong> the stomach or gastroesophageal junction<br />

will be randomized to receive either preoperative chemotherapy alone<br />

(ECFx3 as per MAGIC regimen) or preoperative CRT (ECFx2 followed by<br />

45Gy <strong>of</strong> radiation with concurrent 5-FU). Following surgery, both groups<br />

will receive 3 further cycles <strong>of</strong> ECF. The trial is being conducted in two<br />

<strong>Part</strong>s; <strong>Part</strong> I (phase II component) will recruit 120 patients with the aim <strong>of</strong><br />

demonstrating sufficient efficacy and safety <strong>of</strong> preoperative CRT, as well as<br />

trial feasibility. <strong>Part</strong> II (phase III component) will recruit a further 632<br />

patients to provide a total <strong>of</strong> 752. The primary endpoint for <strong>Part</strong> I is<br />

pathological complete response rate, and for <strong>Part</strong> 2 it is overall survival.<br />

The trial includes formal quality <strong>of</strong> life and biological sub-studies. In<br />

addition, the trial incorporates a rigorous quality assurance program that<br />

includes real time central review <strong>of</strong> radiotherapy plans and central review <strong>of</strong><br />

surgical technique. Current status: This study is an international, intergroup<br />

trial led by the Australasian Gastro-Intestinal Trials Group (AGITG),<br />

in collaboration with the Trans-Tasman Radiation Oncology Group (TROG),<br />

European Organisation for Research and Treatment <strong>of</strong> Cancer (EORTC) and<br />

the NCIC <strong>Clinical</strong> Trials Group. To date, 36 patients have been recruited<br />

from 20 sites in Australia and New Zealand; European and Canadian sites<br />

will commence recruitment in 2012.<br />

TPS4143 General Poster Session (Board #53F), Mon, 8:00 AM-12:00 PM<br />

ST03: A randomized trial <strong>of</strong> perioperative epirubicin, cisplatin plus<br />

capecitabine (ECX) with or without bevacizumab (B) in patients (pts) with<br />

operable gastric, oesophagogastric junction (OGJ) or lower oesophageal<br />

adenocarcinoma. Presenting Author: Elizabeth C Smyth, The Royal Marsden<br />

Hospital, Sutton, United Kingdom<br />

Background: Perioperative ECX chemotherapy is a standard <strong>of</strong> care for<br />

localised gastric/OGJ/lower oesophageal adenocarcinoma (Cunningham<br />

NEJM 2006). B is a monoclonal antibody targeting VEGF-A, and in<br />

combination with chemotherapy results in improved response rates (RR)<br />

and progression free survival in advanced gastric cancer (Ohtsu JCO 2011).<br />

The aim <strong>of</strong> ST03 is to assess the safety and feasibility (stage I, first 200 pts)<br />

and efficacy (stage II) <strong>of</strong> the addition <strong>of</strong> B to perioperative ECX chemotherapy.<br />

Methods: ST03 is a multicentre, open-label, phase II/III randomised<br />

trial ongoing at 92 UK centres; sites in Germany will open in<br />

2012. Eligibility criteria are histologically proven, untreated, resectable,<br />

lower oesophageal, OGJ or gastric adenocarcinoma; age �18 years; WHO<br />

PS 0-1; and adequate cardiac ejection fraction (EF). Exclusion criteria are<br />

TIA/CVA or MI �1 year; uncontrolled hypertension; �Grade II NYHA heart<br />

failure; recent gastrointestinal inflammatory conditions or major surgery/<br />

trauma/open biopsy �28d <strong>of</strong> study entry. Pts receive 3 pre- and 3<br />

postoperative ECX (epirubicin 50 mg/m2 iv D1, cisplatin 60 mg/m2 iv D1<br />

and capecitabine 1250mg/m2 /D1-21) �/- B 7.5mg/kg D1 q3wk during<br />

chemotherapy, then 6 Bev q3wk (investigational arm). Surgery is prespecified<br />

and laparoscopic procedures allowed only after quality assurance<br />

review. All specimens undergo central pathology review; blood and tissue<br />

collection for translational correlates is ongoing. The primary outcome<br />

measures for Stage I (safety results including cardiac EF) have been<br />

reported (Okines ASCO 2011). The stage II primary outcome measure is<br />

overall survival. Secondary outcome measures are RR, resection rate,<br />

disease free survival, toxicity, and QoL. An MRI substudy is open, a PET<br />

substudy is planned. 558 <strong>of</strong> ~950 pts required have been recruited,<br />

accrual expected to complete in 2013. An embedded pilot study within<br />

ST03 randomising HER2 positive patients to ECX � lapatanib will open in<br />

2012. Trial sponsored and co-ordinated by the MRC <strong>Clinical</strong> Trials Unit and<br />

funded by Cancer Research UK (CRUK06/025, NCT00450203).<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


274s Gastrointestinal (Noncolorectal) Cancer<br />

TPS4144 General Poster Session (Board #53G), Mon, 8:00 AM-12:00 PM<br />

A phase II study <strong>of</strong> afatinib (BIBW 2992) in patients with advanced<br />

HER2-positive trastuzumab-refractory esophagogastric cancer. Presenting<br />

Author: Yelena Yuriy Janjigian, Memorial Sloan-Kettering Cancer Center,<br />

New York, NY<br />

Background: Trastuzumab, approved by the FDA, has been the standard <strong>of</strong><br />

care for patients (pts) with HER2-positive esophagogastric cancer. Acquired<br />

and de novo resistance to trastuzumab is an important clinical issue.<br />

Afatinib, an oral irreversible inhibitor <strong>of</strong> the ErbB-family <strong>of</strong> tyrosine kinase<br />

receptors, EGFR (ErbB1), HER2 (ErbB2), and HER4 (ErbB4), in combination<br />

with cetuximab, demonstrated a 40% partial response (PR) rate, with<br />

clinical benefit in �90% in lung cancer patients with acquired resistance<br />

to erlotinib. (Janjigian Y. ASCO 2011). MSKCC data in a HER2-positive<br />

NCI-N87 gastric cancer xenograft showed that while trastuzumab alone<br />

was minimally effective, single-agent afatinib resulted in near complete<br />

tumor regression by inducing apoptosis and downregulation <strong>of</strong> HER2,<br />

p-HER2, EGFR, p-EGFR with minimal additive benefit <strong>of</strong> trastuzumab. In<br />

light <strong>of</strong> these data and the efficacy <strong>of</strong> afatinib in patients with trastuzumabrefractory<br />

breast cancer, we designed a phase II study to determine if<br />

afatinib will benefit patients with trastuzumab-refractory HER2-positive<br />

esophagogastric cancer. We hypothesize that simultaneous inhibition <strong>of</strong><br />

ErbBB receptor family components with afatinib will overcome trastuzumab<br />

resistance. Molecular bases <strong>of</strong> trastuzumab resistance will be<br />

examined. Methods: Pts with metastatic HER2-positive (IHC 3� or FISH<br />

�2.0) esophagogastric cancer with disease progression on a trastuzumabcontaining<br />

regimen will receive afatinib 40 mg once daily. Primary<br />

endpoint RECIST 1.1 response (SD�CR�PR) at 4 months, with imaging<br />

every 8 wks. 13 pts will be enrolled in the 1st stage and if �1 responses are<br />

observed, additional 14 ps (total <strong>of</strong> 27) will be treated. An initial biopsy<br />

prior to the start <strong>of</strong> therapy, a second biopsy after 1 wk <strong>of</strong> afatinib, analysis<br />

<strong>of</strong> archival pre-trastuzumab tissue and blood sample for matched normal<br />

DNA control are mandated. Changes in signaling following afatinib therapy<br />

will provide insight into response heterogeneity. Degree <strong>of</strong> target inhibition<br />

will be correlated with responses. Archival baseline (pre-trastuzumab) and<br />

pre-afatinib tissue will be assessed for abnormalities in pathways implicated<br />

in trastuzumab resistance.<br />

TPS4146 General Poster Session (Board #54A), Mon, 8:00 AM-12:00 PM<br />

A multicenter, randomized, double-blind, phase III study <strong>of</strong> ramucirumab<br />

(IMC-1121B; RAM) and best supportive care (BSC) versus placebo (PBO)<br />

and BSC as second-line treatment in patients (pts) with hepatocellular<br />

carcinoma (HCC) following first-line therapy with sorafenib (SOR). Presenting<br />

Author: Andrew X. Zhu, Division <strong>of</strong> Hematology and Oncology, Massachusetts<br />

General Hospital, Boston, MA<br />

Background: Blockade <strong>of</strong> vascular endothelial growth factor (VEGF) signaling<br />

in HCC has been shown in preclinical models with monoclonal<br />

antibodies and small molecule multikinase inhibitors, and in clinical trials<br />

with SOR, to have anti-tumor activity and improve survival. RAM, a fully<br />

human monoclonal antibody, binds to the VEGF receptor-2 (VEGFR-2),<br />

potently blocks the binding <strong>of</strong> the VEGF ligand to VEGFR-2, inhibits<br />

VEGF-stimulated activation <strong>of</strong> VEGFR-2, and neutralizes VEGF-induced<br />

mitogenesis <strong>of</strong> human endothelial cells. In a single-arm, Phase 2 study <strong>of</strong><br />

RAM as 1st-line monotherapy in 42 pts with advanced HCC (Zhu, ILCA<br />

2010, abstr. O-033), preliminary results included median progression-free<br />

survival (PFS) <strong>of</strong> 4 months (m), time to progression (TTP) <strong>of</strong> 4.2 m, overall<br />

survival (OS) <strong>of</strong> 12 m and disease control rate <strong>of</strong> 70% (best overall<br />

response: 10% partial response, 60% stable disease). Methods: Pts with<br />

HCC with disease progression during or following 1st-line therapy with SOR<br />

(or who were intolerant to SOR) are randomized 1:1 to receive RAM (8<br />

mg/kg) or PBO once every 2 weeks, with BSC, until disease progression or<br />

intolerable toxicity. Eligibility includes prior SOR as 1st-line systemic<br />

treatment for HCC, Child-Pugh A, B7 or 8 cirrhosis, ECOG PS 0-1, bilirubin<br />

� 3 mg/dL, transaminases � 5 � upper limit <strong>of</strong> normal (ULN), creatinine<br />

� 1.2 � ULN, and platelets � 75�103 /�L. The primary endpoint is OS.<br />

Secondary endpoints include PFS, best objective response rate, TTP,<br />

patient-reported outcome measures <strong>of</strong> disease-specific symptoms and<br />

health-related quality <strong>of</strong> life, safety, and RAM pharmacokinetics, pharmacodynamics,<br />

and immunogenicity. With 544 patients, this study is designed<br />

to detect an increase in OS (median 6 m with PBO to 8 m with RAM;<br />

1-sided �� 2.5%, 85% power). As <strong>of</strong> 18 January 2012, approximately<br />

52% <strong>of</strong> planned pts have been randomized. The IDMC reviewed this study<br />

on 21 June and 3 November 2011 and recommended the study to continue<br />

unmodified.<br />

TPS4145 General Poster Session (Board #53H), Mon, 8:00 AM-12:00 PM<br />

DOCTOR: A randomized phase II trial <strong>of</strong> preoperative cisplatin, 5-fluorouracil,<br />

and docetaxel with or without radiotherapy based on poor early response<br />

to standard chemotherapy for resectable adenocarcinoma <strong>of</strong> the esophagus<br />

and/or OG junction. Presenting Author: Andrew Barbour, University <strong>of</strong><br />

Queensland, Brisbane, Australia<br />

Background: Surgery forms the mainstay <strong>of</strong> curative treatment for oesophageal<br />

and gastro-oesophageal junction adenocarcinoma (OAC). Preoperative<br />

chemotherapy (CTX) with or without concurrent radiotherapy (CRT) have<br />

resulted in modest improvements in outcome. Patients who demonstrate a<br />

histological response in the resected specimen following pre-operative<br />

therapy (CTX or CRT) have consistently better survival than non-responders.<br />

Recent data suggests that early metabolic response, assessed by FDG-PET<br />

scan performed 14�/-1 days after the start <strong>of</strong> CTX compared with a<br />

baseline PET scan, is predictive <strong>of</strong> a histological response and improved<br />

survival. Increasing the proportion <strong>of</strong> responders to pre-operative therapy<br />

remains one <strong>of</strong> the major challenges facing patients with localised OAC.<br />

Methods: 150 patients with resectable adenocarcinona <strong>of</strong> the oesophagus<br />

or GOJ will be registered and given 1 cycle <strong>of</strong> cisplatin and 5FU (CF).<br />

Patients will undergo a series <strong>of</strong> baseline tests including endoscopy,<br />

endoscopic ultrasound and 18FDG-PET. At Day 15 <strong>of</strong> the initial CF cycle<br />

the PET will be repeated. Patients with a PET response (� 35% reduction<br />

in SUVmax) to initial treatment will continue with CF as standard treatment<br />

prior to surgery. PET non-responders (� 35% reduction in SUVmax) will be<br />

randomized equally to receive either docetaxel, cisplatin and 5FU (DCF) or<br />

DCF plus radiatiotherapy. Following completion <strong>of</strong> the preoperative chemotherapy<br />

/chemoradiotherapy patients will have their cancer surgically<br />

removed. The primary end point is major histological response (�10%<br />

residual viable tumour) and secondary endpoints <strong>of</strong> progression-free and<br />

overall survival, toxicity, quality <strong>of</strong> life and feasibility <strong>of</strong> response-based<br />

therapy. Eligibility: T2 or greater tumours (by EUS or CT), or T1 tumours<br />

that are poorly differentiated, or T1 node positive, histologically proven<br />

invasive adenocarcinoma <strong>of</strong> the oesophagus or GOJ, technically resectable<br />

tumour which is sufficiently FDG avid on the initial PET staging scan.<br />

Status: Opened to accural 2009.<br />

TPS4147 General Poster Session (Board #54B), Mon, 8:00 AM-12:00 PM<br />

An open-label, randomized phase II study comparing first-line treatment<br />

with dovitinib (TKI258) versus sorafenib in patients with advanced hepatocellular<br />

carcinoma. Presenting Author: Ann-Lii Cheng, National Taiwan<br />

University Hospital, Taipei, Taiwan<br />

Background: Hepatocellular carcinoma (HCC) is a highly vascularized tumor<br />

that may rely on tumor angiogenesis for growth, invasion, and metastasis.<br />

Inhibition <strong>of</strong> the vascular endothelial growth factor receptor (VEGFR) and<br />

platelet-derived growth factor receptor (PDGFR) pathways temporarily<br />

prevents HCC tumor progression. However, because the fibroblast growth<br />

factor receptor (FGFR) pathway can serve as an escape pathway for these<br />

tumors, simultaneous inhibition <strong>of</strong> FGFR may be required to provide a<br />

sustainable antitumor response. Dovitinib (TKI258) has been shown to be a<br />

potent oral tyrosine kinase inhibitor that inhibits multiple angiogenic<br />

factors, including FGFR, VEGFR, and PDGFR, with IC50 values <strong>of</strong> � 20 nM.<br />

Preclinical studies in HCC xenograft models have demonstrated that the<br />

antitumor effects <strong>of</strong> dovitinib are superior to those <strong>of</strong> the kinase inhibitor<br />

sorafenib, which is approved for use in advanced HCC. These data support<br />

additional testing <strong>of</strong> dovitinib in advanced HCC patients. Methods: This<br />

study (NCT01232296) is an open-label, randomized phase II trial being<br />

conducted at multiple centers in the Asia-Pacific region. Adult patients are<br />

eligible if they have advanced HCC (Barcelona Clinic Liver Cancer stage C)<br />

with Child-Pugh class A cirrhotic status. Patients must also have an Eastern<br />

Cooperative Oncology Group (ECOG) performance status <strong>of</strong> 0 or 1 and<br />

cannot be eligible for (or have had disease progression after) surgical or<br />

locoregional therapies. Patients are randomized 1:1 to receive dovitinib<br />

(500 mg/day on a 5-days-on/2-days-<strong>of</strong>f dosing schedule) or sorafenib (400<br />

mg twice daily). Patients will be treated until disease progression, unacceptable<br />

toxicity, death, or discontinuation for any other reason. No treatment<br />

crossover will be permitted. The primary end point <strong>of</strong> this trial is overall<br />

survival. Secondary end points include time to tumor progression, disease<br />

control rate, time to definitive deterioration in ECOG status, safety, and<br />

pharmacokinetics. The pharmacodynamic effects <strong>of</strong> dovitinib and sorafenib<br />

on plasma/serum biomarkers will also be explored. As <strong>of</strong> 30 January 2012,<br />

93 <strong>of</strong> the planned 150 patients have been enrolled.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


TPS4148 General Poster Session (Board #54C), Mon, 8:00 AM-12:00 PM<br />

Sorafenib in combination with local microtherapy guided by gadolinium-<br />

EOB-DTPA enhanced MRI in patients with inoperable hepatocellular<br />

carcinoma (HCC) (SORAMIC). Presenting Author: Jens Ricke, Otto-von-<br />

Guericke-University Magdeburg, Magdeburg, Germany<br />

Background: HCC is a leading cause <strong>of</strong> cancer-related mortality in both men<br />

and women. It represents the fifth most common cancer worldwide with an<br />

increasing incidence. For the individual patient tumor stage at diagnosis<br />

(number and size <strong>of</strong> nodules, presence or absence <strong>of</strong> vascular invasion,<br />

presence or absence <strong>of</strong> extrahepatic spread), liver function and general<br />

health status are the principal prognostic factors. The clinical management<br />

<strong>of</strong> HCC requires a comprehensive, multidisciplinary approach. In early HCC<br />

curative treatment can be achieved by local ablation, resection or liver<br />

transplantation. In intermediate stages patients (pts) are <strong>of</strong>fered locoregional<br />

treatment with palliative intent (transarterial chemoembolisation<br />

(TACE), Yttrium-90-radioembolisation (SIRT)). In advanced disease systemic<br />

therapy with sorafenib (sor) has the potential to prolong survival <strong>of</strong><br />

pts and is standard <strong>of</strong> care in pts with preserved liver function. Studies on<br />

the combined use <strong>of</strong> locoregional and systemic therapy with their potential<br />

<strong>of</strong> a beneficial synergism are few and conducted in a small number <strong>of</strong> pts.<br />

Methods: This phase II-study is composed <strong>of</strong> three substudies with the<br />

following primary objectives: 1. In pts in whom local ablation is appropriate<br />

to determine if sor in combination with RFA prolongs the time-torecurrence<br />

in comparison with RFA plus placebo. Primary endpoint (PEP):<br />

time to recurrence, n � 290 pts 2. In pts in whom RFA is not appropriate<br />

(palliative treatment group) to determine if the combination <strong>of</strong> SIRT and sor<br />

improves the overall survival in comparison to sor alone. PEP: overall<br />

survival, n � 375 pts 3. To confirm in a 2-step procedure that Gd-EOB-<br />

DTPA enhanced MRI is non-inferior or superior compared with contrastenhanced<br />

multislice CT for stratification <strong>of</strong> pts to a palliative or a local<br />

ablation treatment strategy. PEP: correct stratification <strong>of</strong> pts to a palliative<br />

versus local ablation treatment strategy; n � 830 pts The trial has started<br />

in December 2010 as a multinational and multicentric study. 90 pts have<br />

been enrolled until January 31st 2012 with 51 pts randomized in the<br />

palliative arm and 14 pts treated in the curative arm.<br />

TPS4150 General Poster Session (Board #54E), Mon, 8:00 AM-12:00 PM<br />

TACE 2: A randomized placebo-controlled, double-blinded, phase III trial<br />

evaluating sorafenib in combination with transarterial chemoembolisation<br />

(TACE) in patients with unresectable hepatocellular carcinoma (HCC).<br />

Presenting Author: Tim Meyer, University College London, London, United<br />

Kingdom<br />

Background: TACE is regarded as the standard <strong>of</strong> care for patients with<br />

intermediate stage HCC (BCLC B), while sorafenib (S) is the current<br />

standard for advanced disease (BCLC C). The combination <strong>of</strong> S with TACE<br />

for intermediate disease is rational since sorafenib inhibits the action <strong>of</strong><br />

VEGF which may be induced by tumour hypoxia following TACE, and S also<br />

has direct anti-tumour effect. The combination has been shown to be safe<br />

in and active in phase I and II studies. Phase III trials are now required to<br />

determine whether TACE � S is superior to TACE alone in terms <strong>of</strong> survival.<br />

Methods: We are conducting a double blind multicentre trial to determine if<br />

the addition <strong>of</strong> S to TACE is superior to TACE alone. This is a UK NCRI trial<br />

sponsored by University College London (UCL) (EudraCT 2008-005073-<br />

36). Patients with intermediate stage HCC are randomised 1:1 to continuous<br />

S (400mg BD) or placebo (P). Key inclusion criteria include unresectable<br />

HCC confined to the liver, patent main portal vein, ECOG PS �1 and Child<br />

Pugh A liver score. After randomisation patients commence study drug and<br />

TACE is performed at 2-5 weeks using drug eluting beads loaded with<br />

150mg doxorubicin (Biocompatibles UK Ltd) according to a standard<br />

protocol. Further TACE is performed at the discretion <strong>of</strong> the investigator<br />

based on radiological response and patient tolerance. The primary outcome<br />

measure is progression free survival and central, external, real time<br />

radiology review is required to confirm progression. Secondary outcome<br />

measures are overall survival, time to progression, toxicity, disease control,<br />

quality <strong>of</strong> life and number <strong>of</strong> TACE procedures performed in 12 months.<br />

The target recruitment is 412 in order to detect a hazard ratio <strong>of</strong> 0.72 using<br />

a 2-sided level <strong>of</strong> significance <strong>of</strong> ��0.05 with 85% power. The trial was<br />

reviewed by the IDMC after the presentation <strong>of</strong> the SPACE trial (Lencioni et<br />

al J Clin Oncol 30, 2012). The IDMC concurred with the conclusion <strong>of</strong> the<br />

SPACE investigators that the combination is tolerated and results required<br />

confirmation in a phase III trial. Thus recruitment into the TACE2 trial will<br />

continue as planned.<br />

Gastrointestinal (Noncolorectal) Cancer<br />

275s<br />

TPS4149 General Poster Session (Board #54D), Mon, 8:00 AM-12:00 PM<br />

Randomized phase II trial <strong>of</strong> TACE plus everolimus as first-line therapy in<br />

localized unresectable hepatocellular carcinoma (HCC): The TRACER trial.<br />

Presenting Author: Ronnie Tung Ping Poon, Department <strong>of</strong> Surgery,<br />

University <strong>of</strong> Hong Kong, Hong Kong, China<br />

Background: Transcatheter Arterial Chemoembolization (TACE) has shown<br />

benefit in improving survival and is a widely used treatment for unresectable<br />

HCC. However, TACE induces hypoxia leading to up-regulation <strong>of</strong><br />

HIF-1� and VEGF signaling, and tumor progression. Everolimus is an oral<br />

inhibitor <strong>of</strong> mammalian target <strong>of</strong> rapamycin (mTOR) and its downstream<br />

signaling including HIF-1�. Everolimus has also been shown to increase<br />

chemosensitivity <strong>of</strong> HCC in preclinical study (Cancer Lett; 273(2):201-9).<br />

Combining everolimus with TACE may delay tumor progression. TRACER<br />

(Tace with Rad001 in Asian Centers for Evaluation and Research) aims to<br />

evaluate the efficacy and safety <strong>of</strong> everolimus plus TACE in patients with<br />

localised unresectable HCC. Methods: TRACER is a multinational, randomized,<br />

double-blind, placebo-controlled phase II trial. Patients aged �18<br />

years newly diagnosed with localized unresectable Child A HCC; ECOG PS<br />

<strong>of</strong> �2; �1 unidimensional lesion measurable according to RECIST; and<br />

adequate bone marrow, liver, and renal function. Exclusion criteria include<br />

main portal vein invasion and/or extrahepatic spread; prior local or systemic<br />

treatment for HCC, or contraindications to TACE. Eligible patients who have<br />

successfully undergone the first TACE are randomized 1:1 to oral everolimus<br />

7.5mg or matching placebo daily. TACE is repeated every 10 weeks or<br />

so. A short everolimus dose interruption <strong>of</strong> 48 hours before and after each<br />

TACE has been designed to allow recovery from complications <strong>of</strong> TACE. For<br />

standardization, doxorubicin-eluting beads are being used in every TACE.<br />

Patients shall exit the trial upon tumor progression, unacceptable toxicity,<br />

death, or trial discontinuation. All endpoints will be assessed on an ITT<br />

basis. The primary endpoint is time to progression base on Modified<br />

RECIST Criteria. Secondary endpoints include objective response rate,<br />

overall survival, and incidence <strong>of</strong> extrahepatic spread, safety and exploratory<br />

biomarkers. An independent data monitoring committee has been<br />

established to safeguard the trial subjects. Estimated total enrollment is<br />

80, <strong>of</strong> which 12 subjects have been enrolled. (<strong>Clinical</strong> Trial Registry<br />

Number: NCT01379521.)<br />

TPS4151 General Poster Session (Board #54F), Mon, 8:00 AM-12:00 PM<br />

A randomized phase III trial comparing sorafenib plus best supportive care<br />

(BSC) versus BSC alone in Child-Pugh B patients (pts) with advanced<br />

hepatocellular carcinoma (HCC): The BOOST study. Presenting Author:<br />

Bruno Daniele, G. Rummo Hospital, Benevento, Italy<br />

Background: The efficacy <strong>of</strong> sorafenib in pts with advanced HCC has been<br />

demonstrated in two randomized phase III trials (Llovet JM, NEJM<br />

2008;359:378; Cheng AL, Lancet Oncol 2009;10:25), both restricted to<br />

pts with well-preserved liver function (Child-Pugh A). Child-Pugh B (CPB)<br />

pts, that represent a relevant proportion <strong>of</strong> pts with advanced HCC in<br />

clinical practice, were not eligible. Despite this limitation, the marketing<br />

authorization <strong>of</strong> sorafenib by the main regulatory agencies was not<br />

restricted to Child A pts. CPB pts are different in terms <strong>of</strong> prognosis, and<br />

are potentially different in terms <strong>of</strong> balance between treatment efficacy and<br />

toxicity. Large observational studies [Marrero JA, ASCO 2011 (abstr<br />

4001)] are producing quite reassuring data about sorafenib tolerability in<br />

CPB pts, but the real efficacy <strong>of</strong> the drug in this setting remains<br />

substantially unknown, due to the lack <strong>of</strong> randomized trials. Methods:<br />

BOOST (B Child HCC patients – Optimization Of Sorafenib Treatment) is a<br />

randomized phase III trial comparing sorafenib � best supportive care<br />

(BSC) vs. BSC alone in CPB pts with advanced HCC. Pts are eligible if older<br />

than 18, with ECOG performance status 0-2. Pts assigned to experimental<br />

arm receive sorafenib 400 mg twice daily, with dose reductions and<br />

interruptions according to toxicity. Overall survival (OS) is the primary<br />

endpoint. In order to demonstrate a Hazard Ratio <strong>of</strong> death 0.70 in favor <strong>of</strong><br />

sorafenib (2-month improvement in median OS, from 4.5 to 6.5 months),<br />

with 80% power and � 0.05, 320 pts have to be randomized, 160 per arm.<br />

The BOOST trial (<strong>Clinical</strong>Trials.gov Identifier NCT01405573; Eudract<br />

number 2009-013870-42) is approved by the Ethical Committee <strong>of</strong> the<br />

National Cancer Institute, Napoli, Italy, as coordinating centre, and is<br />

currently under evaluation by several other Institutions. BOOST is a<br />

non-pr<strong>of</strong>it, academic trial. The trial has received a financial support by<br />

Italian Ministry <strong>of</strong> Health (FARM84SA2X), although the support is not<br />

enough to supply sorafenib to participating centers. BOOST is partially<br />

supported by AIRC (grant IG2009-9316). The study is open to all<br />

international Centers wishing to participate.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


276s Gastrointestinal (Noncolorectal) Cancer<br />

TPS4152 General Poster Session (Board #54G), Mon, 8:00 AM-12:00 PM<br />

Phase IIb randomized trial <strong>of</strong> JX-594, a targeted multimechanistic oncolytic<br />

vaccinia virus, plus best supportive care (BSC) versus BSC alone in<br />

patients with advanced hepatocellular carcinoma who have failed sorafenib<br />

treatment (TRAVERSE). Presenting Author: James M. Burke, Jennerex,<br />

Inc., San Francisco, CA<br />

Background: JX-594 is a first-in-class targeted oncolytic poxvirus designed<br />

to selectively replicate in and destroy cancer cells with epidermal growth<br />

factor receptor (EGFR)/ ras pathway activation. Direct oncolysis plus<br />

GM-CSF expression stimulates tumor vascular disruption and anti-tumor<br />

immunity (Nature Rev Cancer 2009). JX-594 was well-tolerated in Phase 1<br />

trials and was shown to replicate in metastatic tumors following intratumoral<br />

(IT) or intravenous (IV) administration (Lancet Oncol 2008 and<br />

Nature 2011). A randomized dose-finding Phase 2 trial has been completed<br />

with JX-594 in 30 patients with advanced HCC. Treatment with<br />

high-dose JX-594 was associated with prolonged survival vs low-dose<br />

JX-594 (median survival 14.1 mo vs 6.7 mo; Hazard Ratio 0.39, p�0.02)<br />

(AASLD <strong>Annual</strong> <strong>Meeting</strong>, 2011, LB1). Methods: TRAVERSE is a Phase 2b<br />

randomized, open-label, multi-center trial <strong>of</strong> JX-594 plus BSC versus BSC<br />

in patients with advanced HCC who have failed sorafenib treatment.<br />

Approximately 120 patients will be randomized 2:1 to experimental and<br />

control arm respectively. Randomization will be stratified by region (Asian<br />

vs non-Asian); sorafenib intolerant vs refractory; and presence vs absence<br />

<strong>of</strong> extra-hepatic disease. The primary objective is to determine overall<br />

survival in the 2 arms. Assuming a control median overall survival <strong>of</strong> 4.0<br />

months and a target hazard ratio <strong>of</strong> 0.57 (corresponding to an experimental<br />

arm median survival <strong>of</strong> 7.0 months), 73 events (deaths) will provide 70%<br />

power at 1-sided alpha � 0.05 to detect a difference in overall survival<br />

between the treatment groups using a stratified logrank test. Patients<br />

randomized to JX-594 will receive a dose <strong>of</strong> 109 plaque forming units (pfu)<br />

IV on Day 1 followed by five IT treatments between Day 8 and Week 18.<br />

Main inclusion criteria are advanced HCC having failed sorafenib (intolerance<br />

or radiographic progression during or � 3 months following last<br />

sorafenib), Child-Pugh A-B7 (no ascites), acceptable hematologic function.<br />

Enrollment has begun on this study with clinical trial registry number<br />

<strong>of</strong> NCT01387555.<br />

TPS4153 General Poster Session (Board #54H), Mon, 8:00 AM-12:00 PM<br />

The CLARINET study: Assessing the effect <strong>of</strong> lanreotide autogel on tumor<br />

progression-free survival in patients with nonfunctioning gastroenteropancreatic<br />

neuroendocrine tumors. Presenting Author: Patrick Delavault, Ipsen<br />

Innovation, Paris, France<br />

Background: Somatostatin analogs (SSAs) provide good symptom control in<br />

patients with neuroendrocine tumors (NETs). <strong>Clinical</strong> studies suggest SSAs<br />

also stabilize tumor growth. In a previous double-blind study (PROMID) in a<br />

mixed functioning and non-functioning gastroenteroNET population (n�85),<br />

octreotide LAR significantly increased time to tumor progression compared<br />

with placebo. The CLARINET study is being undertaken to explore the<br />

antiproliferative effects <strong>of</strong> SSAs in a more homogeneous population.<br />

Specifically, the CLARINET study is a double-blind, placebo-controlled<br />

trial assessing the effect <strong>of</strong> 120 mg <strong>of</strong> lanreotide Autogel (lanreotide Depot<br />

in the USA) on progression-free survival in patients with non-functioning<br />

gastroenteropancreatic NETs (GEP-NETs). Methods: CLARINET is a 96week,<br />

multinational study being conducted in collaboration with UKI NETS<br />

and ENETS in patients with well or moderately differentiated nonfunctioning<br />

GEP-NETs and a proliferation index (Ki67) <strong>of</strong> �10%. Patients<br />

are stratified by prior tumor progression status and presence/absence <strong>of</strong><br />

previous therapies, and treated with lanreotide Autogel 120 mg or placebo.<br />

The primary endpoint is time to either disease progression (using Response<br />

Evaluation Criteria In Solid Tumors) or death. Two baseline computed<br />

tomography scans (�12 weeks apart) are being performed, followed by<br />

additional scans at intervals up to 96 weeks. Secondary endpoints include<br />

proportion <strong>of</strong> patients alive and without tumor progression at 48 and 96<br />

weeks, time to progression, overall survival, safety, quality <strong>of</strong> life, plasma<br />

chromogranin A levels, tumor markers, and pharmacokinetic parameters.<br />

The primary study population for the analysis will be the intention-to-treat<br />

population. Recruitment is now complete (n�204) and, thus far, the drug<br />

safety monitoring committee has identified no safety concerns. The trial is<br />

registered with <strong>Clinical</strong>Trials.gov (NCT NCT00353496).<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


4500 Oral Abstract Session, Sat, 3:00 PM-6:00 PM<br />

Cardiac safety analysis for a phase III trial <strong>of</strong> sunitinib (SU) or sorafenib<br />

(SO) or placebo (PLC) in patients (pts) with resected renal cell carcinoma<br />

(RCC). Presenting Author: Naomi B. Haas, Abramson Cancer Center at the<br />

University <strong>of</strong> Pennsylvania, Philadelphia, PA<br />

Background: We performed a cardiac analysis <strong>of</strong> E2805, a well population<br />

<strong>of</strong> pts with resected high risk RCC. The objectives were to determine if pts<br />

treated with SU or SO had clinically significant decreases in left ventricular<br />

(LV) ejection fraction (EF) and to describe the frequency <strong>of</strong> clinically<br />

significant heart failure (HF). We also report the frequency <strong>of</strong> other events<br />

including (un)stable angina or myocardial infarction. Methods: EF was<br />

measured by multiple gated acquisition scan (MUGA) at baseline, 3, 6, and<br />

12 months (mo), and end <strong>of</strong> treatment, if symptoms developed, and 3 mo<br />

after the last abnormal assessment. The primary cardiac endpoint was<br />

defined as an EF decline � the institutional lower limit <strong>of</strong> normal (ILN) that<br />

was a � 16% decline from baseline, and that occurred � 6 mo into<br />

therapy. <strong>Clinical</strong>ly significant HF was � Grade 3 LV systolic or diastolic<br />

dysfunction (severe symptoms with any activity or from drop in EF<br />

responsive (Grade 3) or refractory (Grade 4) to therapy. Late LVEF events<br />

were a drop in LVEF <strong>of</strong> � 16% occurring after 6 mo <strong>of</strong> therapy. Event rates<br />

on each treatment arm were calculated, with 90% exact binomial confidence<br />

intervals (CI). Results: Post-baseline MUGAs are available for 1589<br />

<strong>of</strong> 1943 total pts accrued. 1293 pts had MUGA assessment � 6 mo. 21 pts<br />

had primary events (Table). 71 pts had worst LVEF declines <strong>of</strong> �16% from<br />

baseline, including 52 <strong>of</strong> which occurred � 6 mo from baseline, with the<br />

majority not meeting full primary event criteria. There were 11 reported<br />

grade 3 LV systolic events (5 SU, 4 SO and 2 PLC). 8 pts had cardiac<br />

ischemia possibly or probably from agent. Only one grade 4 event followed a<br />

primary LVEF event. 4 <strong>of</strong> 7 pts who began treatment with LVEF �ILN had<br />

primary LVEF events. Conclusions: In detailed followup <strong>of</strong> SU and SO use,<br />

cardiac function in pts starting with normal EF, was not impaired<br />

significantly over placebo. Ischemic events were uncommon and not clearly<br />

associated with treatment. The data demonstrate that SU or SO for<br />

adjuvant therapy will likely not cause significant cardiac toxicity.<br />

Arm Pts assessed 1° Cardiac endpt Rate 90% CI<br />

SU 397 9 2.3% 1.2-3.9%<br />

SO 394 7 1.8% 0.8-3.3%<br />

PLC 502 5 1.0% 0.4-2.1%<br />

CRA4502 Oral Abstract Session, Sat, 3:00 PM-6:00 PM<br />

Patient preference between pazopanib (Paz) and sunitinib (Sun): Results <strong>of</strong><br />

a randomized double-blind, placebo-controlled, cross-over study in patients<br />

with metastatic renal cell carcinoma (mRCC)—PISCES study, NCT<br />

01064310. Presenting Author: Bernard J. Escudier, Institut Gustave<br />

Roussy, Villejuif, France<br />

The full, final text <strong>of</strong> this abstract will be available at<br />

abstract.asco.org at 12:01 AM (EDT) on Saturday, June 2,<br />

2012, and in the <strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong> online<br />

supplement to the June 20, 2012, issue <strong>of</strong> Journal <strong>of</strong><br />

<strong>Clinical</strong> Oncology. Onsite at the <strong>Meeting</strong>, this abstract will<br />

be printed in the Saturday edition <strong>of</strong> ASCO Daily News.<br />

Genitourinary Cancer<br />

277s<br />

4501 Oral Abstract Session, Sat, 3:00 PM-6:00 PM<br />

Tivozanib versus sorafenib as initial targeted therapy for patients with<br />

advanced renal cell carcinoma: Results from a phase III randomized,<br />

open-label, multicenter trial. Presenting Author: Robert John Motzer,<br />

Memorial Sloan-Kettering Cancer Center, New York, NY<br />

Background: Tivozanib, a potent, selective, long half-life tyrosine kinase<br />

inhibitor targeting all three VEGF receptors, showed activity and tolerability<br />

in a Phase II trial (JCO 2011;29[18S]:4550). Methods: Patients (pts) with<br />

clear cell advanced renal cell carcinoma (RCC), prior nephrectomy,<br />

RECIST-defined measurable disease, and Eastern Cooperative Oncology<br />

Group (ECOG) performance status 0 or 1 were randomized 1:1 to tivozanib<br />

(T) 1.5 mg once daily for 3 weeks (wks) followed by 1 wk rest, or sorafenib<br />

(S) 400 mg twice daily continuously in a 4-wk cycle. Pts were treatment<br />

naïve or received no more than 1 prior systemic therapy for metastatic<br />

disease; pts receiving prior VEGF- or mTOR-targeted therapy were excluded.<br />

The primary endpoint was progression-free survival (PFS) per<br />

blinded, independent radiological review. 500 pts were to be enrolled to<br />

observe 310 events, yielding 90% power to detect medians <strong>of</strong> 9.7 and 6.7<br />

months (m) with 5% type I error (2-sided). Results: A total <strong>of</strong> 517 pts were<br />

randomized to T (n�260) or S (n�257). Demographics were well balanced<br />

between the 2 groups, except ECOG 0 (T: 45% vs S: 54%, p�0.035).<br />

Median PFS was 11.9 m for T vs 9.1 m for S (HR�0.797, 95% CI<br />

0.639–0.993; p�0.042). In the treatment-naïve stratum (70% <strong>of</strong> pts<br />

enrolled in each arm), the median PFS was 12.7 m for T vs 9.1 m for S (HR<br />

0.756, 95% CI 0.580–0.985; p�0.037). In all pts, objective response<br />

rate (ORR) for T was 33% vs 23% for S (p�0.014). The most common<br />

adverse event (AE; all grades/�grade 3) for T was hypertension (T:<br />

46%/26% vs S: 36%/18%) and for S was hand-foot syndrome (T: 13%/2%<br />

vs S: 54%/17%). Other important AEs included diarrhea (T: 22%/2% vs S:<br />

32%/6%), fatigue (T: 18%/5% vs S: 16%/4%), and neutropenia (T:<br />

10%/2% vs S: 9%/2%). Patient-reported outcome data are being analyzed.<br />

Overall survival data are not mature. Conclusions: Tivozanib demonstrated<br />

significant improvement in PFS and ORR compared with sorafenib as initial<br />

targeted treatment for advanced RCC. The safety pr<strong>of</strong>ile <strong>of</strong> tivozanib is<br />

favorable, and includes a low incidence <strong>of</strong> fatigue, diarrhea, myelosuppression,<br />

and hand-foot syndrome.<br />

4503 Oral Abstract Session, Sat, 3:00 PM-6:00 PM<br />

Axitinib for first-line metastatic renal cell carcinoma (mRCC): Overall<br />

efficacy and pharmacokinetic (PK) analyses from a randomized phase II<br />

study. Presenting Author: Brian I. Rini, Cleveland Clinic Taussig Cancer<br />

Institute, Cleveland, OH<br />

Background: Axitinib is a potent, selective, second-generation inhibitor <strong>of</strong><br />

vascular endothelial growth factor receptors with efficacy in mRCC. Due to<br />

PK and pharmacodynamic variability, some patients have sub-optimal drug<br />

exposures at the standard 5-mg twice daily (BID) dose. Prior analyses<br />

indicated higher drug exposure enhanced efficacy; thus, dose titration<br />

based on individual tolerability may optimize exposure and improve<br />

outcomes. A randomized phase II trial evaluated the efficacy and safety <strong>of</strong><br />

axitinib dose titration from 5 mg BID to a maximum <strong>of</strong> 10 mg BID in<br />

first-line mRCC. Methods: Patients with treatment-naïve mRCC received<br />

axitinib 5 mg BID for a 4-week lead-in period (cycle 1). Then, patients with<br />

2 consecutive weeks <strong>of</strong> blood pressure (BP) �150/90 mmHg, no axitinibrelated<br />

toxicities �grade 2, no dose reductions, and �2 antihypertensive<br />

medications were randomized in a double-blind fashion to axitinib 5 mg<br />

BID � dose titration with either axitinib (arm A) or placebo (arm B); those<br />

ineligible for randomization continued with same dose (arm C). Primary<br />

endpoint is objective response rate (ORR). Serial 6-hr PK sampling and<br />

24-hr ambulatory BP monitoring (ABPM) were performed on cycle 1 day 15<br />

in a subset <strong>of</strong> patients. Results: In all, 203 patients were accrued; 112<br />

randomized to arms A or B, and 91 in arm C. As <strong>of</strong> July 1, 2011, ORR (95%<br />

confidence interval [CI]) was 40.2% (31.0, 49.9) in A�B (blinded pooled<br />

analysis) and 56.0% (45.2, 66.4) in C. Median progression-free survival<br />

(mPFS) (95% CI) was 13.7 mo (9.2, not estimable [NE]) in A�B and 12.2<br />

mo (8.6, 16.7) in C. Patients with drug exposure above therapeutic<br />

threshold (AUC24 �300 ng·h/mL; n�27) on cycle 1 day 15 had longer<br />

mPFS and higher ORR than those with sub-therapeutic exposure (n�25)<br />

(mPFS [95% CI]: 13.9 mo [12.2, NE] vs 8.3 mo [5.5, NE]; ORR: 59% vs<br />

48%). Patients with mean increases <strong>of</strong> diastolic BP (�dBP) �15 mmHg<br />

(n�18) per ABPM had higher ORR than those with �dBP �15 mmHg<br />

(n�36) (61% vs 53%). Conclusions: Axitinib is effective in first-line<br />

treatment <strong>of</strong> mRCC, with high ORR and mPFS �1 yr. Preliminary results<br />

suggest therapeutic drug exposure and �dBP �15 mmHg on cycle 1 day<br />

15 may be associated with better outcomes.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


278s Genitourinary Cancer<br />

4504 Oral Abstract Session, Sat, 3:00 PM-6:00 PM<br />

Efficacy <strong>of</strong> cabozantinib (XL184) in patients (pts) with metastatic, refractory<br />

renal cell carcinoma (RCC). Presenting Author: Toni K. Choueiri, Lank<br />

Center for Genitourinary Oncology, Dana-Farber Cancer Institute/Brigham<br />

and Women’s Hospital, Harvard Medical School, Boston, MA<br />

Background: Cabozantinib (cabo) is an oral, potent inhibitor <strong>of</strong> MET and<br />

VEGFR2 that is currently undergoing evaluation in several oncology<br />

indications. Renal cell carcinoma (RCC) was chosen as an indication in this<br />

drug-drug interaction (DDI) study based on involvement <strong>of</strong> the MET and<br />

VEGFR signaling pathways in this disease. The primary objective <strong>of</strong> this<br />

study is to determine the effect <strong>of</strong> cabo on single dose PK <strong>of</strong> the CYP2C8<br />

substrate rosiglitazone (rosi). Anti-tumor activity was also evaluated.<br />

Methods: Eligible pts were required to have RCC with clear cell components<br />

with metastases, Karn<strong>of</strong>sky performance status <strong>of</strong> �70 and measurable<br />

disease by RECIST. Pts needed to have experienced PD following standard<br />

therapies. Cabo was given daily at a dose <strong>of</strong> 140 mg free base (equivalent to<br />

175 mg salt form) starting at Day 2. Rosi (4 mg) was given Day 1 and Day<br />

22 to complete PK assessment for DDI. Cabo was continued until PD. On<br />

Day 57 and every 8 weeks thereafter subjects underwent tumor assessments<br />

by mRECIST. Results: Enrollment is complete at 25 RCC pts; 17/25<br />

(64%) RCC pts had received � 2 prior agents; 13/25 (52%) with at least 1<br />

VEGF pathway inhibitor and 1 mTOR inhibitor. The majority <strong>of</strong> pts were in<br />

an intermediate (21/25) or poor (3/25) prognostic category (1/25 in<br />

favorable category) per Heng et al (JCO, 2009, v27, p5794). ORR by<br />

mRECIST: 7/25 (28%). Disease control rate (PR � SD): 72% at 16 weeks;<br />

19/21 (90%) pts with �1 post-baseline scan experienced tumor regression<br />

(range: 4 - 63% reduction in measurements). 10/25 (36%) pts remain on<br />

cabo. Median PFS is 14.7 months (95% CI: 7.3, upper limit not reached)<br />

with a median follow-up <strong>of</strong> 7.7 months. AEs � Grade 3 severity:<br />

hypophosphatemia (36%), hyponatremia (20%), and fatigue (16%). PK<br />

data suggest that clinically relevant doses <strong>of</strong> cabo do not alter the Cmax or<br />

AUC <strong>of</strong> rosi, consistent with no inhibition <strong>of</strong> CYP2C8. Conclusions: Cabo<br />

0-24h<br />

demonstrates encouraging anti-tumor activity in heavily pretreated RCC pts<br />

with a toxicity pr<strong>of</strong>ile similar to that <strong>of</strong> other VEGFR TKIs. PK data suggest<br />

no DDI between cabo and rosi (CYP2C8 substrate).<br />

4506 Oral Abstract Session, Sat, 3:00 PM-6:00 PM<br />

Randomized phase II trial <strong>of</strong> gemcitabine/cisplatin (GC) with or without<br />

cetuximab (CET) in patients (pts) with advanced urothelial carcinoma (UC).<br />

Presenting Author: Petros Grivas, University <strong>of</strong> Michigan, Ann Arbor, MI<br />

Background: In UCpts EGFR over-expression correlates with high grade,<br />

stage, progression, short survival. Preclinical data show enhanced antitumor<br />

effect with CET � chemotherapy (CT). In several solid tumors CET<br />

added to CT improved survival. CET is hypothesized to improve UC<br />

response to CT. Methods: Pts with ECOG PS 0-2, adequate organ function,<br />

unresectable, locally recurrent or metastatic UC were stratified by disease<br />

state and randomized (1:2) to C 70mg/m2 d1, G 1000mg/m2 d1, d8, d15<br />

(Arm A) or same CT � CET 500mg/m2 d1, d15 (Arm B) q28 days. Due to<br />

high thromboembolic events rate in Arm B, G dose was reduced to 800<br />

mg/m2 . 6 GC cycles were planned. Arm A pts progressing after 2 GC cycles<br />

had CET added. Arm B pts were to continue on CET monotherapy after 4-6<br />

CT cycles. Imaging was q8 weeks. Primary endpoint: RECIST response rate<br />

(RR); secondary: safety, PFS, survival. Since E-cadherin impacts UC<br />

response to CET in vitro, serum level (sE-cad) was assessed at baseline,<br />

after 2 cycles, end <strong>of</strong> CT and at progression. With 1-stage design, assuming<br />

RR 50% (Arm A) and at least 65% (Arm B), planned sample size was 27/54<br />

pts. RR improvement <strong>of</strong> 5% has 81% probability if true difference is 15%.<br />

Results: 88 pts were eligible and randomized, Arm A/B: median age 66/61<br />

years; bladder primary 71.4%/77.2%; metastatic disease 92.9%/89.5%,<br />

median CT cycles number 6/5. 28 Arm B pts continued CET (median 3<br />

cycles, range 1-29); 1 Arm A pt had CET added. Most common G3/4<br />

adverse events (AEs) (Arm A, B): neutropenia (34.5%, 33.9%), anemia<br />

(10.3%, 5.1%), thrombocytopenia (27.6%, 22%), thromboembolism<br />

(6.9%, 18.6%), hyponatremia (3.5%, 10.2%); Arm B only: rash 28.8%,<br />

fatigue 11.9%, hypomagnesemia 11.9% and 2 deaths. 84 pts were<br />

response evaluable (see table below). sE-cad did not correlate with<br />

outcome. sE-cad increased in Arm A, but decreased in Arm B over time.<br />

Conclusions: GC/CET was feasible with no outcome improvement but higher<br />

AEs. sE-cad did not correlate with outcome. Decreased sE-cad in Arm B<br />

suggests potential EGFR-mediated E-cad proteolysis.<br />

Endpoint Arm A (N�28) Arm B (N�56)<br />

RR% (95%CI) 57 (37-76) 62.5 (49-75)<br />

Median PFS (months) (95%CI) 8.5 (6-10) 7.6 (6-10)<br />

Median survival (months) (95%CI) 14 (13-unreached) 14 (12-22)<br />

4505 Oral Abstract Session, Sat, 3:00 PM-6:00 PM<br />

<strong>Clinical</strong> activity and safety <strong>of</strong> anti-PD-1 (BMS-936558, MDX-1106) in<br />

patients with previously treated metastatic renal cell carcinoma (mRCC).<br />

Presenting Author: David F. McDermott, Beth Israel Deaconess Medical<br />

Center, Boston, MA<br />

Background: BMS-936558 is a fully human monoclonal antibody that<br />

blocks the programmed death-1 co-inhibitory receptor (PD-1) expressed by<br />

activated T cells. In the initial portion <strong>of</strong> a phase I study, BMS-936558<br />

showed promising activity in patients (pts) with various solid tumors,<br />

including mRCC. We expanded accrual in order to better characterize<br />

antitumor and dose effects. Methods: RCC pts were treated with BMS-<br />

936558 administered IV Q2WK at 10 mg/kg initially, followed by additional<br />

pts at 1 mg/kg. Pts received up to 12 cycles (4 doses/cycle) <strong>of</strong><br />

treatment or until PD or CR. <strong>Clinical</strong> activity was assessed by RECIST 1.0.<br />

Results: Of 240 pts treated as <strong>of</strong> July 1 2011, 33 had mRCC and were<br />

treated at 1 (n�17) or 10 mg/kg (n�16). ECOG performance status was<br />

0/1 in 13/19 pts. The number <strong>of</strong> prior therapies was 1 (n�10), 2 (n�9), or<br />

�3 (n�14), and included prior immunotherapy (n�20) or antiangiogenic<br />

therapy (n�24); 31 pts had prior nephrectomy. Sites <strong>of</strong> metastatic disease<br />

included lymph node (n�26), liver (n�7), lung (n�28), and bone (n�10).<br />

Median duration <strong>of</strong> therapy was 19 wk (max 96 wk). The incidence <strong>of</strong> grade<br />

3-4 related AEs was 12% and included hypophosphatemia (6%), elevated<br />

ALT (3%), and cough (3%). <strong>Clinical</strong> activity was observed at both dose<br />

levels (Table). Some pts had a persistent reduction in overall tumor burden<br />

in the presence <strong>of</strong> new lesions and were not categorized as responders.<br />

Responses were noted in pts with visceral and bone metastases. Among the<br />

responders who were first treated �1 yr prior to data lock, 4 <strong>of</strong> 5 pts treated<br />

at 10 mg/kg had OR duration �1 yr, and 1 <strong>of</strong> 2 pts treated at 1 mg/kg was<br />

still on study with OR duration <strong>of</strong> 17.5 mo. Although the data for pts treated<br />

at 1mg/kg are not mature, the estimated progression-free survival (PFS)<br />

rate at 24 wk in pts receiving 10 mg/kg was 67% (Table). Conclusions:<br />

BMS-936558 is well tolerated and has durable clinical activity in pts with<br />

previously treated mRCC. Further development <strong>of</strong> BMS-936558 in pts with<br />

mRCC is ongoing.<br />

Dose<br />

(mg/kg) n a OR b uPR c RR d (%)<br />

PFS rate<br />

at 24 wk (%)<br />

1 16 3 2 31 TBD e<br />

10 16 5 - 31 67<br />

a Response evaluable pts.<br />

b CR or PR.<br />

c Unconfirmed PR.<br />

d Response rate [(OR � uPR) ÷ n].<br />

e To be determined (follow up ongoing).<br />

4507 Oral Abstract Session, Sat, 3:00 PM-6:00 PM<br />

Biomarker analysis and final results <strong>of</strong> INT70/09 phase II pro<strong>of</strong>-<strong>of</strong>-concept<br />

study <strong>of</strong> pazopanib (PZP) in refractory urothelial cancer (UC). Presenting<br />

Author: Andrea Necchi, Fondazione IRCCS Istituto Nazionale dei Tumori,<br />

Milan, Italy<br />

Background: Discouraging results have been achieved with standard and<br />

novel compounds in refractory UC. The final results with biomarker analysis<br />

<strong>of</strong> INT70/09 trial with pazopanib are presented. Methods: Pts failing � 1<br />

chemotherapy regimen for metastatic disease underwent PZP 800 mg once<br />

daily until PD or unacceptable toxicity. CT scan and PET scan were planned<br />

at baseline and q4weeks thereafter. Independent review <strong>of</strong> all CT scans was<br />

made and � 5 RECIST CR�PR were required to conclude for activity<br />

according to Simon’s 2-stage design. 50 mL <strong>of</strong> EDTA blood samples were<br />

collected at baseline and q4wks in all pts to analyze plasma VEGF,<br />

sVEGFR-1,-2 and -3, c-Kit, IL-6, 8 and 12 by multiplex ELISA plates.<br />

Results: 41 pts were enrolled from 02/2010 to 07/2011. 15 pts (37%) had<br />

UC <strong>of</strong> the upper urinary tract. 20/41 pts were treated in 3rd line or beyond.<br />

19 pts (46%) were CDDP-refractory and 22 (54%) had hepatic metastases.<br />

27 (66%) had ECOG PS 1-2. 7 pts (17%) had a confirmed PR, 24 had a SD<br />

(76% clinical benefit). 20 pts (49%) had a confirmed necrotic evolution <strong>of</strong><br />

metastases and/or a decreased SUV at PET consistent with PR. Median PFS<br />

and OS (95% CL) were 2.6 (1.7-3.7) and 4.7 mos (4.2-7.3), respectively<br />

but 7 pts (17%) had long-term cure for �10mos (4/7 beyond 2nd line). G3<br />

hypertension occurred in 2 pts, diarrhoea in 5, anemia and hand-foot<br />

syndrome in 3 pts each. Significant increase from T0 (baseline) to T1<br />

(�4wks) level was observed for VEGF (p�0.0001), IL6 (p�0.0129) and<br />

IL8 (p�0.0013) and decrease for VEGFR2 and c-Kit (p�0.0001 each).<br />

Rising IL8T1 levels significantly associated with RECIST progression at<br />

covariance analysis (p�0.0104). Elevated IL8T1 levels (IQ range,<br />

HR�2.11), liver mets (HR�2.33), PS (HR�3.73) and upper tract UC<br />

(HR�0.33) were significant variables for OS at multivariate Cox analysis.<br />

Conclusions: This is the 1st trial ever presented to meet the primary<br />

endpoint with a targeted drug in UC. A role for antiangiogenesis in UC has<br />

been set. Increasing levels <strong>of</strong> IL8 were associated with PD and worse<br />

outcome and may be included in a new prognostic model. Future investigation<br />

should aim at targeting IL8 to improve efficacy results by prolonging<br />

responses.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


4508 Oral Abstract Session, Sat, 3:00 PM-6:00 PM<br />

Bilateral testicular cancer within two prospective, population-based SWENO-<br />

TECA protocols in clinical stage I nonseminoma. Presenting Author:<br />

Torgrim Tandstad, St. Olav’s Hospital, Trondheim, Norway<br />

Background: Contralateral tumor (CLT) occurs in 3.5-5% in men diagnosed<br />

with testicular cancer. The precursor lesion, ITGCNU, transforms into<br />

invasive germ cell cancer in 50-100%. Although radiotherapy eradicates<br />

ITGCNU effectively, mandatory biopsy <strong>of</strong> the contralateral testis to detect<br />

ITGCNU is controversial. Whether, adjuvant chemotherapy (ACT) reduces<br />

the incidence <strong>of</strong> bilateral cancer is also uncertain. Methods: 988 patients<br />

with clinical stage 1 nonseminoma were included in two prospective,<br />

population-based SWENOTECA protocols. Thirteen patients were excluded<br />

due to previous contralateral biopsy, synchronous bilateral cancer or<br />

protocol violations. Treatment was either adjuvant chemotherapy (n�490),<br />

or surveillance (n�485). Contralateral testicular biopsy was recommended,<br />

but performed only in 283 patients. In case <strong>of</strong> ITGCNU radiotherapy<br />

to 16 Gy was recommended.The estimated cumulative incidence <strong>of</strong><br />

CLT was calculated using the Kaplan-Meier method. Results: With a median<br />

follow-up <strong>of</strong> 6.3 years, twenty-nine (3.9%) patients developed CLT including<br />

five patients with synchronous cancer. Biopsies showed ITGCNU in<br />

3.2%. The incidence <strong>of</strong> CLT was similar following ACT, 3.7 % (11/490),<br />

and surveillance, 3.1% (12/485), p�0.99. Biopsied patients had a risk <strong>of</strong><br />

developing CLT <strong>of</strong> 4.3% (9/283), and seven patients treated for CIS never<br />

developed CLT. Unbiopsied patients had a risk <strong>of</strong> 3.0 % (14/668). The<br />

proportion <strong>of</strong> bilateral cancers was similar in biopsy negative patients 3.6%<br />

(7/274) and unbiopsied patients 3.0 % (14/668). Young age at orchiectomy<br />

was a significant risk factor for metachronous cancer, HR 0.94 (CI:<br />

0.89-0.99), p�0.04. All patients with ITGCNU were <strong>of</strong>fered RT. One<br />

irradiated patient developed CLT cancer, and one developed CLT before RT<br />

was given. Conclusions: In this selected population ACT did not reduce the<br />

incidence <strong>of</strong> CLT. There was a high proportion <strong>of</strong> false negative biopsies,<br />

which might explain why biopsy negative patients had the same risk <strong>of</strong> CLT<br />

as patients not undergoing biopsy. Young patients had the highest risk <strong>of</strong><br />

developing contralateral cancer, the risk <strong>of</strong> CLT decreased by 6% yearly.<br />

4510 Oral Abstract Session, Tue, 9:45 AM-12:45 PM<br />

Effect <strong>of</strong> denosumab on prolonging bone-metastasis free survival (BMFS)<br />

in men with nonmetastatic castrate-resistant prostate cancer (CRPC)<br />

presenting with aggressive PSA kinetics. Presenting Author: Fred Saad,<br />

University <strong>of</strong> Montreal Hospital Center, Montreal, QC, Canada<br />

Background: Denosumab, an anti-RANK-ligand monoclonal antibody, has<br />

been shown to prolong BMFS by a median 4.2 months and with a 15% risk<br />

reduction vs. placebo in men with non-metastatic CRPC and baseline PSA<br />

value � 8.0 ng/mL and/or PSA doubling time (PSADT) �10.0 months. To<br />

determine the efficacy <strong>of</strong> denosumab in men at greatest risk for bone<br />

metastases, we evaluated BMFS in a subset <strong>of</strong> men with PSADT �6 months<br />

(previously reported in Smith MR, et al: J Clin Oncol. 23:2918-2925,<br />

2005). Methods: 1,432 men with non-metastatic CRPC (baseline medians:<br />

PSA: 12.3 ng/mL, PSADT: 5.1 months, ADT duration: 47.1 months) were<br />

randomized 1:1 to receive monthly subcutaneous denosumab 120 mg or<br />

placebo. The first patient enrolled February 2006; primary analysis cut-<strong>of</strong>f<br />

was July 2010, when �660 men had developed bone metastasis or died.<br />

The primary endpoint was BMFS (time to first bone metastasis or death<br />

from any cause). BMFS results are presented for men with baseline PSADT<br />

�6 months. Results: Median BMFS in the placebo group <strong>of</strong> men with<br />

PSADT �6 months was 6.5 months shorter than for the placebo group in<br />

the full population (18.7 months vs. 25.2 months), indicating that these<br />

men are at particularly high risk. In this group <strong>of</strong> men with PSADT �6<br />

months, denosumab prolonged BMFS by a median <strong>of</strong> 7.2 months and with<br />

a 23% reduction in risk compared with placebo (Table). Conclusions:<br />

Patients with shortened PSADT are at higher risk <strong>of</strong> developing bone<br />

metastasis and denosumab is markedly effective at prolonging BMFS in<br />

this subset <strong>of</strong> patients.<br />

BMFS<br />

median<br />

(months)<br />

Population<br />

Sample<br />

size<br />

All patients D: 716 D: 29.5<br />

P: 716 P: 25.2<br />

PSADT


280s Genitourinary Cancer<br />

LBA4512 Oral Abstract Session, Tue, 9:45 AM-12:45 PM<br />

Updated analysis <strong>of</strong> the phase III, double-blind, randomized, multinational<br />

study <strong>of</strong> radium-223 chloride in castration-resistant prostate cancer<br />

(CRPC) patients with bone metastases (ALSYMPCA). Presenting Author:<br />

Chris Parker, The Royal Marsden NHS Foundation Trust, Sutton, United<br />

Kingdom<br />

The full, final text <strong>of</strong> this abstract will be available at<br />

abstract.asco.org at 12:01 AM (EDT) on Monday, June 4,<br />

2012, and in the <strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong> online<br />

supplement to the June 20, 2012, issue <strong>of</strong> Journal <strong>of</strong><br />

<strong>Clinical</strong> Oncology. Onsite at the <strong>Meeting</strong>, this abstract will<br />

be printed in the Monday edition <strong>of</strong> ASCO Daily News.<br />

4514 Oral Abstract Session, Tue, 9:45 AM-12:45 PM<br />

A randomized phase II study <strong>of</strong> OGX-427 plus prednisone (P) versus P<br />

alone in patients (pts) with metastatic castration resistant prostate cancer<br />

(CRPC). Presenting Author: Kim N. Chi, British Columbia Cancer Agency,<br />

Vancouver, BC, Canada<br />

Background: Heat Shock Protein 27 (Hsp27) is a multi-functional chaperone<br />

protein that regulates cell signaling and survival pathways implicated<br />

in cancer progression. In prostate cancer models, Hsp27 complexes with<br />

androgen receptor (AR) and enhances transactivation <strong>of</strong> AR-regulated<br />

genes. OGX-427 is a 2nd generation antisense oligonucleotide that inhibits<br />

Hsp27 expression with in vitro and in vivo efficacy and was well tolerated<br />

with single agent activity in phase I studies. Methods: Chemotherapy-naïve<br />

pts with no/minimal symptoms were randomized to receive OGX-427 600<br />

mg IV x 3 loading doses then 1000 mg IV weekly with P5mgPOBIDorP<br />

only. Primary endpoint was the proportion <strong>of</strong> pts progression free (PPF) at<br />

12 weeks (PCWG2 criteria). A 2-stage MinMax design (H0 � 5%, HA �20%, ��0.1, ��0.1) with 32 pts/arm provides 70% power to detect the<br />

difference at 0.10 1-sided significance. Secondary endpoints include PSA<br />

decline, measurable disease response, and circulating tumour cell (CTC)<br />

enumeration. Results: 38 pts have been enrolled; 1st stage <strong>of</strong> accrual<br />

completed with 2nd stage accruing. In the 1st 32 pts randomized (17 to<br />

OGX-427�P, 15 to P), baseline median age was 71 years (53-89), ECOG<br />

PS 0 or 1 in 66% and 34% <strong>of</strong> pts, median PSA 66 (6-606), metastases in<br />

bone/lymph nodes/liver or lung was 75/56/9%, 31% had prior P treatment,<br />

and 93% had �5 CTC/7.5 ml. Predominantly grade 1/2 infusion reactions<br />

(chills, diarrhea, flushing, nausea, vomiting) occurred in 47% <strong>of</strong> pts<br />

receiving OGX-427�P. One pt on OGX-427�P developed hemolytic<br />

uremic syndrome. A PSA decline <strong>of</strong> �50% occurred in 41% <strong>of</strong> pts on<br />

OGX-427�P, and 20% <strong>of</strong> pts treated with P. A measurable disease partial<br />

response was seen in 3/8 (38%) evaluable pts on OGX-427�P and 0/9 pts<br />

on P. CTC conversion from �5 to�5/7.5 ml occurred in 50% <strong>of</strong> pts on<br />

OGX-427�P and 31% treated with P. Thus far, in 26 evaluable pts the PPF<br />

at 12 weeks was 71% (95% CI: 42-92) in OGX-427�P treated pts and<br />

33% (95% CI: 10-65) in pts on P. Conclusions: These data provide clinical<br />

evidence for the role <strong>of</strong> Hsp27 as a therapeutic target in prostate cancer<br />

and support continued evaluation <strong>of</strong> OGX-427 for pts with CRPC. Funded<br />

by a grant from the Terry Fox Research Institute.<br />

4513 Oral Abstract Session, Tue, 9:45 AM-12:45 PM<br />

Cabozantinib (XL184) in chemotherapy-pretreated metastatic castration<br />

resistant prostate cancer (mCRPC): Results from a phase II nonrandomized<br />

expansion cohort (NRE). Presenting Author: Matthew R. Smith, Massachusetts<br />

General Hospital Cancer Center, Boston, MA<br />

Background: Cabozantinib (cabo) inhibits MET and VEGFR2. High rates <strong>of</strong><br />

bone scan resolution, pain relief and overall disease control, independent<br />

<strong>of</strong> PSA changes, were previously reported in a phase II study in mCRPC<br />

patients (pts). This is a NRE cohort in docetaxel (D)-pretreated pts with a<br />

novel primary endpoint <strong>of</strong> bone scan response based on computer-aided<br />

quantitative assessment <strong>of</strong> bone scan lesion area (BSLA) and a doublereader,<br />

independent, blinded review (Nucl Med Commun, in press).<br />

Methods: D-pretreated (�225 mg/m2 ) CRPC pts with bone metastasis were<br />

required to have progressed in s<strong>of</strong>t-tissue or bone within 6 months <strong>of</strong> last<br />

dose <strong>of</strong> D. Pts received 100 mg cabo qd. Tumor response was assessed q6<br />

wks. Bone scan response (BSR) was defined by a �30% decline in BSLA.<br />

Pain intensity (worst pain over the past 24 hrs; BPI scale 0-10) and<br />

interference with sleep and daily activity were prospectively assessed using<br />

an IVR system. Analgesic use was collected by diary. Bone turnover markers<br />

and CTCs were assessed. Results: 93 D-pretreated pts were enrolled (89<br />

evaluable with �6 wks f/u). Median age was 67, 46% received cabazitaxel<br />

and/or abiraterone, 32% had visceral disease, 51% had fatigue, and 18%<br />

had anemia. 44% had worst pain �4 <strong>of</strong> which 95% were taking narcotics.<br />

Median CTC count was 49 and 80% had �5. Median f/u was 125 days<br />

(range, 23-305). Of 85 pts evaluable for BSR, 51 (60%) had a PR, 24<br />

(28%) SD, 5 (6%) PD and 5 (6%) d/c’d prior to f/u scan. 21/30 pts (70%)<br />

had reduction <strong>of</strong> measurable disease.16/33 pts (49%) with BPI �4 and<br />

�12 wks f/u had pain reduction durable for �6 wks; 46% had decreased<br />

narcotic use, including 27% who discontinued use. Sleep and daily activity<br />

were improved in pts with pain relief. Among pts with elevated serum<br />

levels, 74%, 67% and 47% had declines on treatment <strong>of</strong> �30% in CTx,<br />

NTx and bALP, respectively. In 59 pts with CTCs �5, 92% had a decrease<br />

<strong>of</strong> �30% and 39% converted to �5 CTCs at weeks 6 or 12. 12%<br />

discontinued cabo due to AEs. Most common Gr 3/4 AEs were fatigue<br />

(19%), nausea (10%) and anemia (10%). Conclusions: Cabo treatment<br />

resulted in high rates <strong>of</strong> bone scan response, durable pain relief, and<br />

reductions in bone turnover markers and CTCs in D-pre-treated CRPC pts<br />

with bone metastases.<br />

4515 Oral Abstract Session, Tue, 9:45 AM-12:45 PM<br />

Prostate-specific antigen decline (PSA) as a surrogate for overall survival<br />

(OS) in patients (pts) with metastatic castrate-resistant prostate cancer<br />

(mCRPC) who failed first-line chemotherapy. Presenting Author: Susan<br />

Halabi, Duke University Medical Center, Durham, NC<br />

Background: PSA kinetics, and more specifically a 30% decline in PSA<br />

following initiation <strong>of</strong> first-line chemotherapy with docetaxel, has been<br />

reported to be a surrogate endpoint for OS in mCRPC pts. The objective <strong>of</strong><br />

this analysis was to evaluate PSA kinetics as surrogate endpoints for overall<br />

survival (OS) in patients who were receiving second line chemotherapy<br />

following progression after docetaxel front line therapy. Methods: Data from<br />

a phase III trial <strong>of</strong> 755 mCRPC pts randomized to treatment with<br />

cabazitaxel in combination with prednisone (C�P) every 3 weeks or<br />

mitoxantrone in combination with prednisone (M�P) were used. All pts<br />

were previously treated with a docetaxel-containing regimen. PSA decline<br />

(�30% and �50% ) and PSA velocity within the first three months <strong>of</strong><br />

treatment were evaluated as potential surrogate endpoints for OS. The<br />

proportional hazards (PH) model was used to test for Prentice’s criteria and<br />

the proportion <strong>of</strong> treatment explained (PTE) was computed as a second test<br />

<strong>of</strong> surrogacy. PTE was defined as one minus the ratio <strong>of</strong> the treatment<br />

coefficient in the adjusted PH model (includes PSA decline or velocity) to<br />

the treatment coefficient in the unadjusted PH model. Results: Of 755<br />

men, 654 had sufficient PSA data to be included in the analysis. Treatment<br />

arm (C�P vs. M�P) was prognostic <strong>of</strong> OS with a hazard ratio (HR) <strong>of</strong> 0.65<br />

(95% CI�0.54-0.79, p�0.001). A 30% PSA decline within three months<br />

<strong>of</strong> treatment was associated with a HR <strong>of</strong> 0.46 (95% CI 0.37-0.57,<br />

p-value�0.001) for OS. After adjusting for treatment effect, the HR for<br />

30% PSA decline was 0.50 (95% CI� 0.40-0.62, p�0.001) but treatment<br />

arm remained statistically significant thus failing Prentice’s third<br />

criterion. The PTE for �30% decline in PSA within three months was 0.39<br />

(95% CI� 0.36-0.42) indicating a lack <strong>of</strong> surrogacy for OS. Similar results<br />

were observed for pts who experienced �50% decline in PSA and PSA<br />

velocity. Conclusions: Neither PSA decline (�30% and �50%) nor PSA<br />

velocity within the first three months <strong>of</strong> therapy are surrogate endpoints for<br />

OS in pts receiving second line chemotherapy.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


4516 Oral Abstract Session, Tue, 9:45 AM-12:45 PM<br />

A validated whole-blood RNA transcript-based prognostic model that<br />

predicts survival in men with castration-resistant prostate cancer. Presenting<br />

Author: William K. Oh, Division <strong>of</strong> Hematology and Medical Oncology,<br />

The Tisch Cancer Institute, Mount Sinai School <strong>of</strong> Medicine, New York, NY<br />

Background: Survival for patients with castration resistant prostate cancer<br />

(CRPC) is highly variable. We developed a whole blood RNA transcriptbased<br />

model as a prognostic biomarker in CRPC. Methods: Peripheral blood<br />

was collected from 62 men with CRPC in a training set and from 140<br />

patients with CRPC in a validation set on various treatment regimens. A<br />

panel <strong>of</strong> 168 inflammation and prostate cancer-related genes was evaluated<br />

using optimized quantitative polymerase chain reaction to assess<br />

biomarkers predictive <strong>of</strong> survival. A 2-class proportional hazard model was<br />

developed from time <strong>of</strong> CRPC diagnosis and time <strong>of</strong> blood draw. Results: A<br />

6-gene model (consisting <strong>of</strong> ABL2, SEMA4D, ITGAL, and C1QA, TIMP1,<br />

CDKN1A) separated CRPC patients into two classes: higher risk men who<br />

died within 2·2 years <strong>of</strong> developing CRPC and lower risk men who lived over<br />

2·2 years (log rank p�0·00083). The results were similar regardless <strong>of</strong> the<br />

survival time definition (CRPC diagnosis versus blood draw) and did not<br />

depend on whether they received chemotherapy in addition to hormone<br />

treatment. The model successfully validated in an independent cohort <strong>of</strong><br />

men with CRPC (p� 0.000001·7). Conclusions: Transcriptional pr<strong>of</strong>iling <strong>of</strong><br />

whole blood yields critical prognostic information in men with CRPC<br />

independent <strong>of</strong> treatment. The 6-gene model suggests possible dysregulation<br />

<strong>of</strong> the immune system, a finding that warrants further study. This<br />

model may play an important role in patient counseling, in patient<br />

stratification for clinical trials, and potentially as a predictive biomarker for<br />

immune-based therapeutic strategies.<br />

LBA4518 <strong>Clinical</strong> Science Symposium, Sat, 8:00 AM-9:30 AM<br />

Interim analysis (IA) results <strong>of</strong> COU-AA-302, a randomized, phase III study<br />

<strong>of</strong> abiraterone acetate in chemotherapy-naive patients (pts) with metastatic<br />

castration-resistant prostate cancer (mCRPC). Presenting Author: Charles<br />

J. Ryan, University <strong>of</strong> California, San Francisco, San Francisco, CA<br />

The full, final text <strong>of</strong> this abstract will be available at<br />

abstract.asco.org at 12:01 AM (EDT) on Saturday, June 2,<br />

2012, and in the <strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong> online<br />

supplement to the June 20, 2012, issue <strong>of</strong> Journal <strong>of</strong><br />

<strong>Clinical</strong> Oncology. Onsite at the <strong>Meeting</strong>, this abstract will<br />

be printed in the Saturday edition <strong>of</strong> ASCO Daily News.<br />

Genitourinary Cancer<br />

281s<br />

4517 Oral Abstract Session, Tue, 9:45 AM-12:45 PM<br />

18F-16�-fluoro-5�-dihydrotestosterone (FDHT) PET as a prognostic biomarker<br />

for survival in patients with metastatic castrate-resistant prostate<br />

cancer (mCRPC). Presenting Author: Karen A. Autio, Memorial Sloan-<br />

Kettering Cancer Center, New York, NY<br />

Background: At present there is no imaging biomarker for patients with<br />

mCRPC as the disease primarily metastasizes to bone, which is difficult to<br />

visualize and quantify using standard techniques. We have conducted<br />

clinical trials in which patients with progressive mCRPC are scanned using<br />

FDG-PET to examine glucose metabolism, and FDHT-PET. FDHT is a novel<br />

tracer and structural analog <strong>of</strong> dihydrotestosterone that binds to the<br />

androgen receptor (AR) to demonstrate AR overexpression, a key biologic<br />

feature <strong>of</strong> mCRPC. We previously reported an association between baseline<br />

FDG-PET and overall survival in mCRPC. We now present the prognostic<br />

utility <strong>of</strong> baseline FDHT-PET. Methods: We prospectively scanned mCRPC<br />

patients with FDG and FDHT-PET prior to a change in therapy as part <strong>of</strong><br />

imaging clinical trials. PET results were represented as the hottest lesion<br />

(SUVmax), or average <strong>of</strong> the five hottest lesions (SUVmaxavg). Five lesions<br />

per patient were recorded. If less than 5 lesions were captured, a value <strong>of</strong> 1<br />

was imputed for the remaining lesions. <strong>Clinical</strong> and lab parameters were<br />

collected. Univariate analysis was performed on PET, clinical and lab<br />

variables for association with overall survival (OS). Multivariate models <strong>of</strong><br />

prognostic factors were constructed. Results: 170 mCRPC patients were<br />

imaged with FDG and 116 also had FDHT-PET at baseline. Median survival<br />

was 21.2 months (95%CI: 19.1-24.0). On univariate analysis, FDHTmax,<br />

FDHTmaxavg, FDGmaxavg, PSA, hemoglobin, alkaline phosphatase, and<br />

LDH were associated with OS. FDHTmax and FDHTmaxavg were significantly<br />

correlated with one another. In a multivariate model, FDHTmaxavg<br />

and LDH were prognostic <strong>of</strong> survival. In a separate model assessing FDG,<br />

FDGmaxavg and LDH were also prognostic. Conclusions: FDHT not only<br />

allows for a visual assessment <strong>of</strong> the AR axis in prostate cancer, but appears<br />

to associate with overall survival in mCRPC. Increased AR activity as<br />

measured by FDHT SUVmaxavg held greater prognostic value than PSA or<br />

Gleason score. FDHT is also being evaluated as a clinical response indicator<br />

and pharmacodynamic measure with AR directed therapies.<br />

4519^ <strong>Clinical</strong> Science Symposium, Sat, 8:00 AM-9:30 AM<br />

Primary, secondary, and quality-<strong>of</strong>-life endpoint results from the phase III<br />

AFFIRM study <strong>of</strong> MDV3100, an androgen receptor signaling inhibitor.<br />

Presenting Author: Johann Sebastian De Bono, The Institute for Cancer<br />

Research, London, United Kingdom<br />

Background: MDV3100, a novel androgen receptor signaling inhibitor<br />

(ARSI), inhibits: 1) binding <strong>of</strong> androgens to AR, 2) AR nuclear translocation,<br />

and 3) association <strong>of</strong> AR with DNA. MDV3100 was active in a phase<br />

I-II trial enrolling pre- and post-docetaxel castration-resistant prostate<br />

cancer (CRPC) patients. The AFFIRM trial evaluated whether MDV3100<br />

could provide benefit to men with post-docetaxel CRPC. Methods: In this<br />

double-blind, multinational phase III study, patients who had received<br />

docetaxel-based chemotherapy were randomized 2:1 to MDV3100 160<br />

mg/day or placebo. Treatment with corticosteroids was allowed but not<br />

required. Patients were stratified by baseline ECOG and mean brief pain<br />

inventory score. The primary endpoint was overall survival (OS). Other<br />

efficacy endpoints included radiographic progression-free survival (rPFS),<br />

time to PSA progression (TTPP), s<strong>of</strong>t tissue objective response (PR�CR),<br />

PSA response, and quality <strong>of</strong> life (QoL) response (FACT-P). Results: 800<br />

patients were randomized to MDV3100 and 399 to placebo with respective<br />

median treatment durations <strong>of</strong> 8.3 and 3.0 months.Based on a planned<br />

interim analysis at 520 deaths, the Independent Data Monitoring Committee<br />

recommended the study be unblinded and placebo patients <strong>of</strong>fered<br />

MDV3100. Efficacy results are presented (Table). The most common<br />

MDV3100 events with an incidence higher than placebo were fatigue (34%<br />

vs 29%), diarrhea (21% vs 18%), and hot flush (20% vs 10%). Grade �3<br />

events <strong>of</strong> interest were cardiac disorders (0.9% MDV3100 vs 2% placebo),<br />

fatigue (6% MDV3100 vs 7% placebo), seizure (0.6% MDV3100 vs. 0%<br />

placebo), and LFT abnormalities (0.4% MDV3100 vs 0.8% placebo).<br />

Conclusions: MDV3100, a novel ARSI, is well-tolerated and significantly<br />

prolongs OS, slows disease progression, and improves QoL in men with<br />

post-docetaxel CRPC.<br />

MDV3100 Placebo HR (95% CI) P value<br />

OS, median 18.4 months 13.6 months 0.631 (0.529, 0.752) �0.0001<br />

rPFS, median 8.3 months 2.9 months 0.404 (0.350, 0.466) �0.0001<br />

TTPP, median 8.3 months 3.0 months 0.248 (0.204, 0.303) �0.0001<br />

PR�CR 25.1%� 3.8% 2.9%�1.0% - �0.0001<br />

PSA response 54.0% 1.5% - �0.0001<br />

QoL response 43.3% 17.8% - �0.0001<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


282s Genitourinary Cancer<br />

4520 <strong>Clinical</strong> Science Symposium, Sat, 8:00 AM-9:30 AM<br />

Neoadjuvant androgen pathway suppression prior to prostatectomy. Presenting<br />

Author: Elahe A. Mostaghel, Fred Hutchinson Cancer Research Center,<br />

Seattle, WA<br />

Background: Optimizing tissue androgen suppression may provide better<br />

local and systemic control <strong>of</strong> prostate cancer (PCa). Standard androgen<br />

deprivation therapy (ADT) has limited effect on tissue androgens which<br />

remain in a range which supports tumor survival. We determined whether<br />

targeting androgen metabolism using CYP17 and 5a-reductase (SRD5A)<br />

inhibitors would more effectively suppress tissue androgens and tumor<br />

volume. Methods: Open label, multicenter neoadjuvant study in men with<br />

localized PCa treated for 3 months prior to prostatectomy with zoladex and<br />

1) avodart 3.5 mg QD; 2) avodart and casodex 50 mg QD; or 3) casodex,<br />

avodart and ketoconazole 200 mg TID. Serum and tissue androgens were<br />

measured by LC/MS/MS. Data were compared to men treated with standard<br />

ADT (LHRH agonist plus Casodex), and untreated prostatectomy tissue.<br />

The primary outcome measure was suppression <strong>of</strong> tissue dihydrotestosterone<br />

(DHT). Results: 35 men with intermediate/high risk PCa were enrolled.<br />

Tissue DHT was suppressed 30 fold (� 95%) in all groups vs. LHRH<br />

agonist/Casodex (0.92 � 0.20 pg/mg vs. 0.03� 0.03 for all groups<br />

combined, p�0.0001). Tissue testosterone was 3-4 fold higher (consistent<br />

with SRD5A inhibition) in all treatment groups vs. LHRH agonist/Casodex<br />

(0.33 vs. 0.07 pg/mg, p � 0.05). Differences in DHT/T between groups 1<br />

through 3 were not statistically significant. There was no correlation<br />

between tissue and serum androgens, or tissue androgen and tumor volume<br />

(p� 0.05). In subset analysis, total serum DHEA declined significantly in<br />

group 3, with free DHEA unchanged, suggesting differential effect on free<br />

and total DHEA. Pathologic complete response (CR) was seen in 2 men,<br />

and an additional 8 men had �0.2 cc <strong>of</strong> tumor, with the largest number <strong>of</strong><br />

CR or near CR in the cohort treated with ADT, CYP17 and SRD5A inhibitor,<br />

4 <strong>of</strong> 12 men (33%). Due to small cohort size, differences were not<br />

statistically significant. Conclusions: Addition <strong>of</strong> high dose SRD5A inhibition<br />

(with and without CYP17 inhibition) achieves prostate DHT levels 30<br />

fold below standard ADT. In this relatively high risk population, CR or near<br />

CR was seen in 10 <strong>of</strong> 35 men receiving protocol therapy. Further<br />

suppressing the androgen receptor signaling axis may provide better local<br />

and systemic control <strong>of</strong> PCa.<br />

4522 Poster Discussion Session (Board #1), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Time from prior chemotherapy (TFPC) as a prognostic factor in advanced<br />

urothelial carcinoma (UC) receiving second-line systemic therapy. Presenting<br />

Author: Gregory R. Pond, McMaster University, Hamilton, ON, Canada<br />

Background: Prognostic factors for overall survival (OS) in patients receiving<br />

second-line chemotherapy for advanced platinum-pretreated UC include<br />

ECOG performance status (PS) �0, hemoglobin (Hb) �10g/dL and the<br />

presence <strong>of</strong> liver metastasis (LM) (Bellmunt J, J Clin Oncol 2010). We<br />

hypothesized that time from prior chemotherapy (TFPC) independently<br />

impacts OS. Methods: Of 11 available phase II trials evaluating second-line<br />

therapy for advanced UC (n�698), 6 trials with available baseline Hb, PS<br />

and LM were utilized (n�534). The trials evaluated vinflunine (2 trials),<br />

docetaxel plus vandetanib or placebo, paclitaxel-gemcitabine, nanoparticlealbumin-bound<br />

paclitaxel and paclitaxel-cetuximab. The Kaplan-Meier<br />

method was used to estimate OS from date <strong>of</strong> starting second-line therapy.<br />

Cox proportional hazards regression stratified for trial was used to evaluate<br />

the prognostic effect <strong>of</strong> factors on OS. TFPC was evaluated as a continuous<br />

variable, and based on cutpoints <strong>of</strong> 3, 6, 9 and 12 months (mo) from prior<br />

chemotherapy to first study treatment. The choice <strong>of</strong> optimal cutpoint for<br />

TFPC was determined by the maximum likelihood ratio �2 statistic. Results:<br />

Overall, 513 patients were evaluable. 64.1% received prior chemotherapy<br />

for metastatic disease. Median OS was 6.8 mo (95% CI: 6.1 to 7.4); range<br />

was 0 to 84.2 mo. Median OS was 5.2, 7.1, 8.8, 7.6 and 10.6 mo<br />

respectively for TFPC �3 (n�181), 3 to �6 (n�133), 6 to �9 (n�77), 9<br />

to �12 (n�45) and �12 (n�77) mo, respectively. Shorter TFPC was<br />

independently prognostic for decreased survival. The optimal cutpoint for<br />

TFPC was �3 mo, but no well-defined plateau was observed. PS�0<br />

(HR�1.72, p�0.001), LM (HR�1.41, p�0.002), Hb �10 g/dl (HR�1.59,<br />

p�0.001) and TFPC �3 mo (HR�1.67, p�0.001) were significantly<br />

prognostic in the multivariate model. Timing <strong>of</strong> prior chemotherapy<br />

(metastatic disease vs. perioperative) was not prognostic. Conclusions: A<br />

shorter duration <strong>of</strong> TFPC exhibited a significant negative prognostic impact<br />

on OS independent <strong>of</strong> known prognostic factors in patients receiving<br />

second-line therapy for advanced UC. If externally validated, TFPC should<br />

be a stratification factor in trials <strong>of</strong> second-line therapy for advanced UC.<br />

4521 <strong>Clinical</strong> Science Symposium, Sat, 8:00 AM-9:30 AM<br />

Effect <strong>of</strong> neoadjuvant abiraterone acetate (AA) plus leuprolide acetate<br />

(LHRHa) on PSA, pathological complete response (pCR), and near pCR in<br />

localized high-risk prostate cancer (LHRPC): Results <strong>of</strong> a randomized<br />

phase II study. Presenting Author: Mary-Ellen Taplin, Dana-Farber Cancer<br />

Institute, Boston, MA<br />

Background: LHRPC is infrequently cured with prostatectomy (RP). To date<br />

neoadjuvant androgen deprivation therapy (ADT) has not improved outcomes<br />

and residual intra-prostatic androgens remain. AA lowers serum<br />

testosterone (T) and DHT to � 1 ng/dL and has improved survival in<br />

advanced PC. Methods: We conducted a neoadjuvant, phase II trial <strong>of</strong><br />

AA/LHRHa in LHRPC. The primary aim was to evaluate intra-prostatic<br />

T/DHT with LHRHa vs LHRHa/AA. We report secondary endpoints <strong>of</strong> PSA,<br />

pCR and near pCR ([� 5mm residual tumor]) and safety. Eligibility: � 3<br />

positive biopsies and either Gleason � 7(4�3), T3, PSA � 20 ng/mL or<br />

PSA velocity � 2 ng/mL/year. For the first 12 wks men were randomized to<br />

LHRHa or LHRHa/AA/prednisone (P) 5mg qd. After 12 wks, a prostate<br />

biopsy was done to measure T/DHT. Men received 12 more wks <strong>of</strong><br />

LHRHa/AA/P followed by RP. Results: 58 men enrolled: 28 to initial LHRHa<br />

and 30 to initial AA/LHRHa. 2 withdrew prior to RP (1/group). Median age<br />

was 58 (50-75). pCR/near pCR was 14/56 (25%). Grade 3 AEs included<br />

elevated AST/ALT 5/58 (9%) and hypokalemia 3/58 (5%). No grade 4<br />

mineralocorticoid-related AEs were observed. Conclusions: Neoadjuvant<br />

ADT with AA was well tolerated in LHRPC. PSA (�0.2) declines were high<br />

and achieved earlier on AA/LHRHa compared to LHRHa. The pCR/near pCR<br />

rates were higher for 24 wks AA (34%) than 12 wks AA (15%). No new<br />

safety signals were seen with AA used with P 5 mg. These results support<br />

further evaluation <strong>of</strong> aggressive ADT as neoadjuvant/adjuvant therapy for<br />

LHRPC.<br />

Baseline<br />

12 wks AA/<br />

24 wks LHRHa<br />

n� 28<br />

24 wks AA/<br />

24 wks LHRHa<br />

n�30<br />

Gleason: 7/8/9/10 8/10/10/0 9/7/11/3<br />

PSA (median) 10.6 6.8<br />

PSA: < 10/10-20/> 20 12/9/7 20/6/4<br />

Elevated PSA velocity 6 3<br />

Stage T3 8 6<br />

Results n�27 n�29<br />

PSA: wk 4/8/12/16/20/24 4.34/1.35/1.06/0.20/0.09/0.06 0.65/0.17/0.10/0.09/0.06/0.05<br />

12 wk nadir PSA < 0.2 1/27 (4%) 26/29 (90%) p�0.0001<br />

24 wk nadir PSA < 0.2 23/27 (85%) 25/29 (86%) p�0.9131<br />

pCR 1/27 (4%) 3/29 (10%) p�0.3349<br />

Near pCR (tumor < 5mm) 3/27 (11%) 7/29 (24%) p�0.2034<br />

Total pCR/near pCR 4/27 (15%) 10/29 (34%) p�0.0894<br />

pT3 16/27 14/29<br />

Positive nodes 3/27 (11%) 7/29 (24%)<br />

Positive margins 5/27 (19%) 5/29 (17%)<br />

4523 Poster Discussion Session (Board #2), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Neoadjuvant chemotherapy with DD-MVAC and bevacizumab in high-risk<br />

urothelial cancer: Results from a phase II trial at the University <strong>of</strong> Texas<br />

M. D. Anderson Cancer Center. Presenting Author: Arlene O. Siefker-<br />

Radtke, University <strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: DD-MVAC has shown an improved 5-year survival in metastatic<br />

urothelial cancer; however, there is no prospective data on its use in the<br />

neoadjuvant setting. Previous work suggested that over-expression <strong>of</strong> VEGF<br />

was associated with a high risk <strong>of</strong> relapse in our neoadjuvant patients,<br />

leading to the hypothesis that combining a VEGFR inhibitor with chemotherapy<br />

may improve patient outcomes. Methods: Between 8/07 and<br />

12/10, 60 patients with urothelial carcinoma <strong>of</strong> the bladder or upper tract<br />

tumor were enrolled on a prospective phase II clinical trial. Eligibility<br />

requirements included at least one <strong>of</strong> the following: 3-D mass on EUA<br />

(cT3b disease), LVI, hydronephrosis, micropapillary features, tumor in a<br />

diverticula, and for upper tract tumors a high grade tumor or radiographically<br />

measurable sessile mass. The primary endpoint was pathologic<br />

down-staging to ��pT1N0M0. Results: Forty-four patients with bladder/<br />

urethral tumors and 16 patients with upper tract tumors were enrolled.<br />

Pathologic down-staging to �� pT1N0M0 occurred in 53% <strong>of</strong> patients<br />

overall (bladder/urethra 45%, upper tract 75%), and to �� pT0N0M0 in<br />

38% overall (bladder/urethra 39%, upper tract 38%). At a median<br />

follow-up <strong>of</strong> 26 months, the 2-year OS and DSS was 78% and 82%,<br />

respectively (bladder 2-yr OS and DSS 75%, 78%; upper tract 93%, 93%).<br />

The median OS and DSS have not yet been reached. The most common<br />

grade 3 or greater toxicity was neutropenia in 27% <strong>of</strong> patients, followed by<br />

fatigue in 10%. The following grade 3 toxicities were observed in � 10% <strong>of</strong><br />

patients: mucositis, DVT/PE, hypertension, nausea/vomiting, thrombocytopenia.<br />

One patient experienced cardiac ischemia. Conclusions: Neoadjuvant<br />

chemotherapy leads to pathologic down-staging in 45% <strong>of</strong> patients<br />

with bladder cancer. DD-MVAC appears an acceptable alternative to<br />

traditional M-VAC in the neoadjuvant setting. Although bevacizumab did<br />

not impact down-staging based upon historical expectations, determining<br />

the effect on recurrence requires longer follow-up.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


4524 Poster Discussion Session (Board #3), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Prognostic model for overall survival in patients with metastatic urothelial<br />

cancer treated with cisplatin-based chemotherapy. Presenting Author:<br />

Matt D. Galsky, Division <strong>of</strong> Hematology and Medical Oncology, The Tisch<br />

Cancer Institute, Mount Sinai School <strong>of</strong> Medicine, New York, NY<br />

Background: Patients with metastatic urothelial cancer (UC) treated with<br />

cisplatin-based chemotherapy have heterogeneous clinical outcomes. Pretreatment<br />

prognostic variables have previously been defined (Bajorin, JCO,<br />

1999) but have not been extensively validated or expanded upon. Methods:<br />

Pretreatment clinical parameters from 400 international patients with<br />

metastatic UC treated on phase II and III first-line clinical trials with<br />

cisplatin-based chemotherapy from 11/1997 to 10/2011 were evaluated<br />

(cisplatin � gemcitabine �/- paclitaxel or �/- antiangiogenic agent � 235;<br />

MVAC � 83; cisplatin � 5FU �/- paclitaxel � 79). Parameters were<br />

subjected to multivariable regression analysis to determine which patient<br />

characteristics had independent prognostic significance for survival. Results:<br />

Complete data were available on 361 patients, median survival was 13.9<br />

months (95% CI 12.4, 16.5); 247 have died. In multivariable analysis,<br />

ECOG performance status (�1), visceral metastases (lung, liver, bone), and<br />

white blood count (WBC, continuous variable) were significantly associated<br />

with poor survival (P � .05), whereas perioperative chemotherapy, hemoglobin,<br />

platelets, creatinine clearance, site <strong>of</strong> primary tumor, and number <strong>of</strong><br />

metastatic sites were not (Table). The cut-<strong>of</strong>f for WBC was established at<br />

10. Median survival times for patients with 0, 1, 2, or 3 risk factors were<br />

26.9, 16.8, 12.8, 9.7 months (log rank p value � 0.0001). When<br />

subjected to internal validation, the model achieved a bootstrap-corrected<br />

c-index <strong>of</strong> 0.60. External validation is ongoing utilizing data from a phase<br />

III trial (n�186) <strong>of</strong> MVAC versus docetaxel plus cisplatin. Conclusions: A<br />

model derived from pretreatment parameters from an international cohort<br />

<strong>of</strong> patients was constructed that confirmed, and expanded upon, known<br />

prognostic factors in metastatic UC. This model may be useful for patient<br />

counseling and clinical trial design. Better predictions require the identification<br />

<strong>of</strong> additional factors that affect survival.<br />

Prognostic model for overall survival.<br />

Parameter Hazard ratio 95% CI P<br />

WBC > 10 1.66 1.27, 2.18 0.0002<br />

Visceral metastases 1.49 1.15, 1.93 0.0024<br />

ECOG > 1 1.52 1.16, 2.00 0.0028<br />

4526 Poster Discussion Session (Board #5), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Neoadjuvant accelerated MVAC (AMVAC) in patients with muscle invasive<br />

bladder cancer: Results <strong>of</strong> a multicenter phase II study. Presenting Author:<br />

Elizabeth R. Plimack, Fox Chase Cancer Center, Philadelphia, PA<br />

Background: Standard methotrexate, vinblastine, doxorubicin and cisplatin<br />

(MVAC) demonstrates a survival benefit in the neoadjuvant setting for<br />

patients (pts) with muscle invasive bladder cancer (MIBC). Compared with<br />

standard MVAC, AMVAC yielded higher response rates with less toxicity in<br />

the metastatic setting. Methods: Pts with MIBC, cT2-T4a, and N0-N1 with<br />

CrCl ��50 and adequate hepatic and marrow function were eligible. Pts<br />

received 3 cycles <strong>of</strong> AMVAC (methotrexate 30 mg/m2 , vinblastine 3 mg/m2 ,<br />

doxorubicin 30 mg/m2 , cisplatin 70mg/m2 ) on day 1, with pegfilgrastim 6<br />

mg day 2 or 3, every 2 weeks. Pts with CrCl � 60 could receive cisplatin<br />

split over 2 days. Radical cystectomy (RC) with lymph node dissection was<br />

performed 4-8 weeks after the last dose <strong>of</strong> chemotherapy. Primary endpoint<br />

was pathologic complete response (pCR) rate. Results: Accrual is complete<br />

with 44 MIBC pts enrolled at 2 institutions (FCCC, TJU) over a 25 month<br />

period. Median age 64 (range 45-83). Three withdrew from study early and<br />

are not evaluable for response (2 physician discretion, 1 withdrawal <strong>of</strong><br />

consent). An additional 8 are currently receiving treatment on study with<br />

toxicity and response data pending. Of the 33 evaluable pts for whom final<br />

data is available, 30 received all 3 cycles <strong>of</strong> AMVAC at full dose. Three pts<br />

received � 3 cycles due to grade 3 fatigue (1), low platelets (1), and<br />

disease progression precluding RC (1). 32/33 pts underwent RC, all within<br />

8 weeks <strong>of</strong> last chemotherapy. Median time from start <strong>of</strong> chemotherapy to<br />

RC was 9.7 wks (range 4.6-13 wks). 13/33 pts (39.4%, 95% CI,<br />

22.7-56.1%) had a pCR. An additional 3 (9.1%) were downstaged to non<br />

muscle invasive disease. For the intent to treat cohort (n�36) 8 pts had<br />

grade 3-4 AMVAC related adverse events, the most common being anemia<br />

(3), fatigue (3) and neutropenia (2) and overall pCR rate was 36.1%. (95%<br />

CI, 20.4-51.8%). All pts will have completed study treatment by April<br />

2012. Final results will be presented. Conclusions: Neoadjuvant AMVAC is<br />

well tolerated and preliminary results show a pCR rate similar to that<br />

reported for standard 12-week MVAC, suggesting that AMVAC for three<br />

cycles (6 weeks) is a safe and efficient alternative.<br />

Genitourinary Cancer<br />

283s<br />

4525 Poster Discussion Session (Board #4), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Correlation <strong>of</strong> progression-free survival at 6 months (PFS6) with overall<br />

survival at 12 months (OS12) in an analysis <strong>of</strong> 10 trials <strong>of</strong> second-line<br />

therapy for advanced urothelial carcinoma (UC). Presenting Author: Benjamin<br />

Maughan, University <strong>of</strong> Utah, Salt Lake City, UT<br />

Background: Second-line therapy <strong>of</strong> advanced UC is a significant unmet<br />

need. While phase II trials have relied on response rate (RR) as a primary<br />

endpoint, response may not be suitable for assessing cytostatic agents and<br />

does not capture duration <strong>of</strong> benefit. We hypothesized that PFS6 correlates<br />

with OS12 and may be a robust intermediate endpoint for phase II trials.<br />

Methods: Ten trials with individual patient progression and survival data<br />

(n�646) evaluating chemotherapy and/or biologics following chemotherapy<br />

were combined. Progression was defined as tumor progression<br />

(RECIST 1.0 in 9 trials, WHO in 1 trial), or death from any cause.<br />

Unadjusted and adjusted binomial confidence intervals for PFS6, OS12<br />

and response were reported, with adjustment for variability between trials<br />

using random effects models. The relationship between PFS6 and OS12<br />

was assessed at the trial level using Pearson correlation and weighted linear<br />

regression with larger studies having more influence. The relationship<br />

between PFS6 and OS12 at the individual level was assessed using<br />

Pearson chi-square test with Yates continuity correction. Statistical analyses<br />

employed “R” statistical computing s<strong>of</strong>tware, version 2.8.0. Results:<br />

59% had visceral metastasis and performance status was 0, 1 and 2 in 50,<br />

39 and 8%, respectively. Median age was 65 years (31-88) and 77% were<br />

male. PFS6 was 22% (95% CI: 17-24%), and adjusted PFS6 was 23%<br />

(95% CI: 15-34%). The OS12 was 20% (95% CI: 17-24%), and adjusted<br />

OS12 was 21% (95% CI: 15-29%). The Pearson correlation between trial<br />

level PFS6 and OS12 was 0.66 (p � 0.037). The individual level<br />

agreement between PFS6 and OS12 was seen in 82% <strong>of</strong> patients (kappa �<br />

0.45). Among 560 patients evaluable for response and OS, the RR was<br />

22% (95% CI: 18-25%) and adjusted RR was 21% (95% CI: 13-32%).<br />

Trial level Pearson correlation between response and OS12 was 0.37 (p �<br />

0.30), and individual level agreement was seen in 78% (kappa�0.36).<br />

Conclusions: PFS6 is associated with OS12 at the trial and individual levels<br />

in patients receiving second-line therapy for advanced UC. The association<br />

<strong>of</strong> response with OS was suboptimal. Validation <strong>of</strong> PFS6 is warranted.<br />

4527 Poster Discussion Session (Board #6), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Defining the genetic basis <strong>of</strong> everolimus sensitivity in metastatic bladder<br />

cancer (MBC) by whole-genome sequencing (WGS). Presenting Author:<br />

Gopa Iyer, Memorial Sloan-Kettering Cancer Center, New York, NY<br />

Background: MBC is rarely cured with standard chemotherapy and novel<br />

agents are clearly needed. A phase II trial <strong>of</strong> everolimus in MBC resulted in<br />

1 near-complete response (CR), 1 partial response, and 3 minor responses.<br />

We used WGS and a Next Generation exon capture assay to identify the<br />

genetic aberrations underlying treatment response. Methods: In the near-CR<br />

case, tumor and peripheral blood DNA was subjected to WGS (Illumina)<br />

using 2x100 bp paired-end libraries for 40X haploid coverage. Raw<br />

sequence data was aligned to hg19 and somatic point mutation detection<br />

was performed. Tumor DNA from 13 additional patients on the trial was<br />

analyzed using an exon capture sequencing assay. Results: 184 exonic<br />

somatic mutations were identified in the everolimus complete responder by<br />

WGS, including a2bpframeshift truncation in TSC1 and a nonsense<br />

mutation in NF2. Sanger sequencing <strong>of</strong> an additional 96 high-grade<br />

bladder tumors found 5 (6.2%) tumors containing TSC1 alterations and no<br />

NF2 mutations. Of the 13 patients on the trial who underwent targeted<br />

exon sequencing, 3 (23%) possessed nonsense TSC1 mutations and 2 had<br />

minor treatment responses (17% and 24% tumor regression). 1 patient<br />

with a 7% tumor regression had a somatic missense TSC1 variant. 8 (89%)<br />

<strong>of</strong> 9 patients with tumor progression as best response were TSC1 wild-type.<br />

Patients with TSC1-mutant tumors remained on drug for longer duration<br />

(7.7 vs. 2 months, p�0.004, Wilcoxon rank sum test). Conclusions:<br />

Everolimus is an active agent in MBC in patients with TSC1 mutant tumors.<br />

The pattern <strong>of</strong> co-mutation in patients with TSC1 mutant tumors is complex<br />

and combination therapies will likely be required to achieve durable<br />

responses in most patients. Our results also highlight the potential utility <strong>of</strong><br />

WGS in identifying novel predictors <strong>of</strong> response to targeted inhibitors.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


284s Genitourinary Cancer<br />

4528 Poster Discussion Session (Board #7), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Association <strong>of</strong> long-term exposure to circulating platinum with adverse late<br />

effects in testicular cancer survivors. Presenting Author: Hink Boer, Department<br />

<strong>of</strong> Medical Oncology, University Medical Center Groningen, Groningen, Netherlands<br />

Background: Successful platinum(Pt)-based chemotherapy for testicular cancer<br />

(TC) comes at the price <strong>of</strong> late cardiovascular morbidity and neurotoxicity.<br />

Damage induced by circulating Pt could be an etiological mechanism. We<br />

investigated the relation between circulating Pt and late effects. Methods: In 96<br />

consecutive TC patients (med age 29 [17-53] at chemotherapy), 3 serum<br />

samples and a 24h-urine sample were collected on several time-points med 5 yrs<br />

(1-13) after treatment. Pt concentrations ([Pt]) were measured with a sensitive<br />

voltammetric method (Lancet 2000, 355:1075). Measured [Pt] combined with<br />

cisplatin dose, age, weight and height were analyzed simultaneously to construct<br />

a population pharmacokinetic (PK) model using NONMEM. Based on the PK<br />

parameters <strong>of</strong> each patient, individual [Pt] at 1, 3, 5, 10 yrs after start and AUC<br />

were calculated. Cardiovascular status, paresthesia and markers <strong>of</strong> vascular<br />

damage were assessed after a med follow-up (FU) <strong>of</strong> 9 yrs (3-15). Results: Decay<br />

<strong>of</strong> [Pt] was best described by a two compartment model. Mean terminal T1/2 was<br />

3.7 (�0.3) yrs. At all time-points [Pt] correlated with cisplatin dose and renal<br />

function during treatment. [Pt] at 3 yrs correlated with systolic blood pressure<br />

(BP) (r�.32, p�0.01) and creatinine clearance (CRCL) (r�-.25, p�0.02) at FU.<br />

Patients with increased BP had higher Pt AUCs than patients with normal BP<br />

(table). Pt AUCs were higher in patients with paresthesia (table). Conclusions:<br />

Known late effects <strong>of</strong> cisplatin-based chemotherapy such as hypertension and<br />

paresthesia are related to higher circulating [Pt]. Long-term exposure to Pt is<br />

involved in healthy tissue damage in TC survivors.<br />

[Pt] pg/g serum (mean�SD)<br />

AUC 1-5 yrs<br />

(pg/g •day)•103 n 3 yrs 5 yrs<br />

Cisplatin dose<br />

800mg<br />

43<br />

.048*<br />

53<br />

379 �118<br />

.028<br />

433 �140<br />

189 �55<br />

.013<br />

218 �66<br />

960 �250<br />

1109 �316<br />

CRCL (mL/min)<br />

>120<br />


4532 Poster Discussion Session (Board #11), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Serum tumor marker (STM) decline rates during high-dose chemotherapy<br />

(HDCT) to predict outcome for germ cell tumor (GCT) patients (pts).<br />

Presenting Author: Darren Richard Feldman, Memorial Sloan-Kettering<br />

Cancer Center, New York, NY<br />

Background: STM decline rates predict outcome for GCT pts during 1st-line conventional-dose chemotherapy. We investigated the importance <strong>of</strong> STM<br />

decline rates during salvage HDCT. Methods: HDCT included an initial low<br />

dose (LD) portion (1-2 cycles <strong>of</strong> paclitaxel plus ifosfamide [TI] � G-CSF) for<br />

stem cell collection followed by a high-dose (HD) portion (3 cycles <strong>of</strong><br />

carboplatin � etoposide or TI � carboplatin). Eligible pts had elevated STM<br />

(AFP �15 and/or HCG �2.2) at the start <strong>of</strong> LD and completed �1 HD<br />

cycle. Slope and t1/2 were calculated for HCG and AFP for cycles 1-2 <strong>of</strong> LD<br />

and HD starting with the peak value during days (d) 1-6 <strong>of</strong> LD and HD to<br />

avoid interference from lysis. T1/2 for AFP �7d and HCG �3.5d were<br />

categorized as satisfactory (SAT); normalization within 7d was also considered<br />

SAT, regardless <strong>of</strong> t1/2. Pts with elevated AFP and HCG had to have<br />

adequate decline in both to be considered SAT. Progression-free (PFS) and<br />

overall survival (OS) were compared for SAT vs. unsatisfactory (UNSAT) pts<br />

using the log-rank test. Results: Of 117 pts (115 male, 105 nonseminoma),<br />

the most common primary sites were testis (81) and mediastinum (26). 93,<br />

19, and 5 pts completed 3, 2, and 1 HD cycles, respectively. Overall,<br />

19/117 had SAT decline during LD (19/77 for HCG; 7/57 for AFP). For LD,<br />

there was no significant difference in PFS or OS for SAT vs. UNSAT decline.<br />

For HD, 17/96 had SAT decline (11/59 for HCG; 9/47 for AFP). SAT overall<br />

decline (HCG and AFP) during HD predicted superior PFS (p�0.0025) and<br />

OS (p�0.0046) with 2-year (yr) PFS 0.88 (0.59, 0.97) vs. 0.37 (0.26,<br />

0.47) and 3yr OS 0.82 (0.55, 0.94) vs. 0.37 (0.26, 0.48). SAT HCG<br />

decline alone also predicted superior PFS (p�0.0018) and OS (p�0.004)<br />

with 2yr PFS 0.91 (0.51, 0.99) vs. 0.32 (0.19, 0.46) and 3yr OS 0.91<br />

(0.51, 0.99) vs. 0.35 (0.22, 0.50) whereas only a non-significant trend for<br />

improved PFS (p�0.14) and OS (p�0.16) was seen for AFP alone.<br />

Conclusions: STM decline rates during the HD portion <strong>of</strong> salvage HDCT<br />

predicts favorable PFS and OS, mostly driven by HCG. However, UNSAT<br />

STM decline does not exclude long-term PFS/OS. STM decline rates during<br />

the initial LD portion <strong>of</strong> HDCT programs do not predict outcome; thus,<br />

UNSAT LD decline should not prohibit subsequent HDCT.<br />

4534 Poster Discussion Session (Board #13), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

A retrospective analysis <strong>of</strong> patients with intermediate-risk germ cell tumor<br />

(IRGCT) treated at Indiana University from 2000 to 2010. Presenting<br />

Author: Costantine Albany, Indiana University Melvin and Bren Simon<br />

Cancer Center, Indianapolis, IN<br />

Background: Based on the International Germ Cell Cancer Collaborative<br />

Group (IGCCCG), IRGCT represents 26% <strong>of</strong> non-seminomas and 10% <strong>of</strong><br />

seminomas with 2 year PFS <strong>of</strong> 74% (63-85%). Most patients are treated<br />

with 4 cycles <strong>of</strong> Bleomycin, Etoposide and Cisplatin (BEP). However, the<br />

optimal therapy for this heterogeneous group is not well defined. Our<br />

hypothesis was that many patients with IRGCT may not require BEPx4 as<br />

they are clinically closer to good risk rather than to poor risk disease.<br />

Methods: We conducted a retrospective analysis <strong>of</strong> all patients with<br />

testicular cancer seen at Indiana University (IU) between 2000- 2010. We<br />

identified 170 pts with IRGCT <strong>of</strong> whom 84 consecutive pts received their<br />

therapy at IU. 2 chemotherapy categories were identified: BEP (or equivalent)<br />

x4 or BEPx3 (�/-EPx1). Results: 45 pts received either BEPx4 (41),<br />

VIPx4 (2) or BEPx2 followed by HDCTx2 (2). 39 pts received BEPx3 (9) or<br />

BEPx3�EPx1 (30). Treatment decisions were based on clinical characteristics<br />

and markers amplitude. There were 78 (93%) non-seminoma, 6 (7%)<br />

seminoma. 27 (32%) stage II and 57 (68%) stage III. Mean AFP 3899<br />

(range 1000-9550; n�50), mean hCG 16354 (range 5000-49000;<br />

n�34). The overall 1 and 2 year Kaplan-Meier estimates were 93% and<br />

87% for PFS and 98% and 92% for OS. Results are depicted below.<br />

Conclusions: Our results for patients with IRGCT treated from 2000-2010<br />

are superior to the IGCCCG pts treated from 1975-1990. Routine administration<br />

<strong>of</strong> BEPx4 probably represents overtreatment in a substantial<br />

percent <strong>of</strong> IRGCT.<br />

BEPx3 (�/- EPx1) BEPx4 /other p value<br />

Number 39 45<br />

AFP mean (range) 3890 (1009- 8300) n�23 3907 (1000- 9550) n�28<br />

hCG mean (rRange) 16074 (5000-49000) n�14 16549 (5000- 36800) n�20<br />

Persistent GCT on post<br />

5 2<br />

chemo RPLND<br />

Salvage high dose<br />

4 6<br />

chemotherapy (HDCT)<br />

Death 2 2<br />

PFS 1 yr, 2 yrs 95%, 89% 91%, 85% 0.60<br />

OS 1 yr, 2 yrs 100%, 94% 96%, 90% 0.86<br />

Genitourinary Cancer<br />

285s<br />

4533 Poster Discussion Session (Board #12), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Phase II trial <strong>of</strong> bevacizumab (BEV)/high-dose chemotherapy (HDC) for<br />

refractory germ-cell tumors (GCT). Presenting Author: Yago Nieto, University<br />

<strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: HDC is a curative therapy for relapsed GCT. However, multiple<br />

prior relapses, cisplatin refractoriness (relapse/progression (PD) within 4<br />

weeks) or absolute refractoriness (no prior response), or very high tumor<br />

markers at relapse predict poor early event-free survival (EFS). The<br />

validated Beyer model (JCO 1996;14:2638) identifies groups with poor<br />

risk (5% EFS at 1-year post-HDC), intermediate risk (25% EFS) or good risk<br />

(�50% EFS). Given high VEGF expression in metastatic GCT and synergy<br />

between BEV and chemotherapy, we studied concurrent BEV/HDC in<br />

refractory GCT. Methods: Eligibility includes � 1st relapse/PD, poor/interm<br />

risk and no contraindications to HDC or BEV. Treatment consisted <strong>of</strong> 2<br />

cycles <strong>of</strong> HDC with stem-cell support following BEV (5 mg/kg) 1 week<br />

before each cycle. HDC-1 consists <strong>of</strong> a novel regimen <strong>of</strong> infusional<br />

gemcitabine with docetaxel/melphalan/carboplatin (BBMT 2005;11:297).<br />

HDC-2 includes ifosfamide/carboplatin/etoposide. Target accrual is 25 pts,<br />

to distinguish a 1-yr expected EFS <strong>of</strong> 15% (median time to progression<br />

[TTP], 9 months) from 50% 1-year EFS (median TTP, 2 years). Results: 21<br />

pts have been treated, median age 22 (range, 19-45) poor risk (N�15) or<br />

interm risk (N�6), cisplatin refractoriness (N�9) or absolute refr (N�12),<br />

median 3 prior relapses (1-4), median 3 prior regimens (2-6), PD at HDC:<br />

11 pts. Tumor sites: lungs (N�15), liver (8), bones (5), brain (5), retroperit<br />

(15), mediast (12). Histol at Dx: embryonal ca (4), chorio (3), mixed with<br />

teratoma (14). Prior surgery for metastases: 8 pts (6 abdomen, 1 liver, 1<br />

brain). Prior radiation: 6 pts. Toxicity <strong>of</strong> HDC-1: grade 3 mucositis in all<br />

pts, rash (8 G2, 2 G3) and transaminase elevation; 2 pts with marginal<br />

baseline renal function died from early sepsis; 1 pt died from late fungal<br />

pneumonia. After HDC-1, markers normalized in 14/17 evaluable pts. 15<br />

pts received HDC-2 at a median 49 (38-66) days after 1st stem cell<br />

infusion, which was well tolerated. 8 pts had residual lesions resected with<br />

findings <strong>of</strong> either teratoma (N�2) or no viable tumor (N�6). With median<br />

follow-up <strong>of</strong> 23 (3-43) months, 14 pts are alive in CR (67% EFS).<br />

Conclusions: BEV with HDCx2 shows encouraging preliminary results in<br />

heavily pretreated refractory GCT.<br />

4535 Poster Discussion Session (Board #14), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Large retroperitoneal lymph nodes (RPLN) as a novel risk factor for venous<br />

thromboembolism (VTE) in germ cell tumor (GCT) patients (pts) receiving<br />

first-line chemotherapy (chemo). Presenting Author: Ben Tran, Princess<br />

Margaret Hospital, Toronto, ON, Canada<br />

Background: VTE causes substantial morbidity and mortality in GCT pts.<br />

The Khorana model is a validated predictive model that stratifies VTE risk in<br />

cancer pts receiving chemo; Khorana score �3 (K3) identifies pts at high<br />

risk. Many GCT pts have bulky RPLN that can cause venous stasis in the<br />

lower limbs. This study examines the incidence <strong>of</strong> VTE in GCT pts and<br />

assesses large RPLN as a novel predictor <strong>of</strong> VTE risk. Methods: Retrospective<br />

data from the Princess Margaret Hospital GCT database was complemented<br />

by review <strong>of</strong> medical records. GCT pts receiving 1st line chemo<br />

between 2000-2010 were included. Pts diagnosed with VTE prior to chemo<br />

and pts receiving thromboprophylaxis (TP) were excluded. Pts diagnosed<br />

with VTE during or within 3 months <strong>of</strong> completing chemo were identified.<br />

Large RPLN were defined as having maximal diameter �5cm. Odds ratios<br />

for VTE risk with large RPLN and K3 were calculated. Discriminatory<br />

accuracy (DA) <strong>of</strong> each predictor was calculated using area under the<br />

receiver operating characteristic curves (AUROC). An external cohort from<br />

London Regional Cancer Program, with similarly collected data, was used<br />

to validate results. Results: In the test cohort 21 (10%) <strong>of</strong> 216 pts<br />

developed VTE. Both large RPLN (OR 6.2, p�0.001) and K3 (OR 11.8,<br />

p�0.001) were significantly associated with VTE. Positive predictive value<br />

(PPV) was lower for large RPLN compared to K3 (21% v 44%, p�0.014)<br />

but sensitivity was greater (71% v 40%, p�0.001), while DA showed no<br />

difference (AUROC 0.73 v 0.67, p�0.46). In the validation cohort 10 (9%)<br />

<strong>of</strong> 111 pts developed VTE. There was a non significant trend for associations<br />

between VTE and both large RPLN (OR 2.54, p�0.16) and K3 (OR<br />

4.5, p�0.13). When compared to K3, sensitivity was greater for large<br />

RPLN (60% v 20%, p�0.003), but there was no difference in PPV (14% v<br />

29%, p�0.25) or DA (AUROC 0.61 v 0.57, p�0.72). Conclusions: VTE<br />

occurs in 1 in 10 GCT pts receiving curative chemotherapy. Large RPLN is<br />

a novel and clinically applicable predictor <strong>of</strong> VTE risk, although prospective<br />

validation would be beneficial. Randomized controlled trials <strong>of</strong> TP in GCT<br />

pts should be considered in pts at high risk <strong>of</strong> VTE, identified by large RPLN<br />

or K3.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


286s Genitourinary Cancer<br />

4536 Poster Discussion Session (Board #15), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

An in-depth multicentered population-based analysis <strong>of</strong> outcomes <strong>of</strong><br />

patients with metastatic renal cell carcinoma (mRCC) that do not meet<br />

eligibility criteria for clinical trials. Presenting Author: Daniel Yick Chin<br />

Heng, Tom Baker Cancer Centre, University <strong>of</strong> Calgary, Calgary, AB,<br />

Canada<br />

Background: <strong>Clinical</strong> trials have strict eligibility criteria that exclude many<br />

patients to whom the trial results are later extrapolated to in clinical<br />

practice. Methods: mRCC patients treated with VEGF targeted therapy were<br />

retrospectively deemed ineligible for clinical trials (according to commonly<br />

used inclusion/exclusion criteria) if they had a Karn<strong>of</strong>sky Performance<br />

Status (KPS) �70%, brain metastases, non-clear cell histology, hemoglobin��9<br />

g/dL, creatinine �2x the upper limit <strong>of</strong> normal, platelet count <strong>of</strong><br />

�100x103 /uL, neutrophil count �1500/mm3 or corrected calcium��12<br />

mg/dL. Results: 894/2076 (43%) patients were deemed ineligible for<br />

clinical trials by the above criteria. Between ineligible versus eligible<br />

patients, the response rate, median progression free survival (PFS) and<br />

median overall survival <strong>of</strong> first-line targeted therapy were 21% vs 29%, 5.2<br />

vs 8.8 months and 14.5 vs 28.8 months (all p�0.0001), respectively.<br />

Second-line PFS (if applicable) was 3.2 months in the trial ineligible vs 4.4<br />

months in the trial eligible patients (p�0.0074). Patients who were<br />

excluded due to KPS�70, hemoglobin��9 g/dL, calcium ��12, brain<br />

metastases, and non-clear cell histology, had a hazard ratio (HR) for death<br />

<strong>of</strong> 2.8 (95%CI 2.4-3.4), 1.8 (95%CI 1.4-2.2), 1.8 (95%CI 1.2-2.7), 1.4<br />

(95%CI 1.1-1.8), and 1.4 (95%CI 1.1-1.7), respectively (all p�0.01).<br />

When adjusted by the Heng et al prognostic categories, the HR for death<br />

between trial ineligible vs trial eligible patients was 1.511 (95%CI�1.335-<br />

1.710, p�0.0001). Conclusions: The number <strong>of</strong> patients that are ineligible<br />

for clinical trials is high and their outcomes are inferior. Specific trials<br />

addressing the needs <strong>of</strong> protocol ineligible patients and assessing OS are<br />

required.<br />

<strong>Clinical</strong> trial<br />

ineligible N�894<br />

<strong>Clinical</strong> trial<br />

eligible N�1,182 P value<br />

Parameter<br />

Heng et al prognosis<br />

�0.0001<br />

Favorable<br />

9%<br />

25%<br />

Intermediate<br />

48%<br />

59%<br />

Poor<br />

43%<br />

16%<br />

Median KPS (%) (range) 80 (20-100) 90 (70-100) �0.0001<br />

Anemia (below LLN) 68.7% 51.4% �0.0001<br />

Hypercalcemia (above ULN) 14.5% 6.8% �0.0001<br />

Brain metastases present 19% 0% �0.0001<br />

Non-clear cell histology 26% 0% �0.0001<br />

4538 Poster Discussion Session (Board #17), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Characteristics <strong>of</strong> long-term and short-term survivors <strong>of</strong> metastatic renal cell<br />

carcinoma (mRCC) treated with targeted therapy: Results from the International<br />

mRCC Database Consortium. Presenting Author: Wanling Xie, Dana-Farber<br />

Cancer Institute, Boston, MA<br />

Background: Patients with mRCC have variable courses in terms <strong>of</strong> survival and<br />

response to targeted therapy. The patients at the two extremes <strong>of</strong> the survival<br />

spectrum need to be characterized. Methods: 2,161 patients with mRCC treated<br />

with targeted therapy were examined. 152 patients who survived 4 years or more<br />

after the initiation <strong>of</strong> targeted therapy (long-term) were compared with 218<br />

patients who survived 6 months or less (short-term) over the same time period<br />

(2004-2007). Results: Long-term survivors had fewer poor prognostic factors<br />

(PFs) such as Karn<strong>of</strong>sky performance status (KPS) �80%, diagnosis to treatment<br />

interval�1 yr, hypercalcemia, anemia, thrombocytosis and neutrophilia<br />

(all p�0.0001). Patients with favorable prognosis who responded to targeted<br />

therapy were more likely to be long term survivors. For those in the intermediate<br />

risk group, patients who were long-term survivors were more likely to have only 1<br />

poor prognostic factor (73% vs. 28%, p�0.0001) and KPS�80% (88% vs.<br />

69%, p�0.009) compared to those in the short term survivor group. On<br />

multivariable analysis adjusting for PFs, response to targeted therapy (PR or<br />

better) significantly predicted long term survivor status (odds ratio�6.3, 95%<br />

CI: 2.3,17.4, p�0.0004). Conclusions: Long term survivors had a higher<br />

response rate to targeted therapy, a longer treatment duration and more use <strong>of</strong><br />

second-line targeted therapy. Baseline prognostic criteria may be able to<br />

discriminate between long- and short- term survivors.<br />

Long-term Short-term<br />

survivor<br />

survivor<br />

Characteristic<br />

(N�152) (N�218) P value<br />

Median age (years) 61 61 0.26<br />

Heng et al prognostic category<br />

�0.0001<br />

Favorable<br />

42%<br />

2%<br />

Intermediate<br />

49%<br />

34%<br />

Poor<br />

3%<br />

60%<br />

Unknown<br />

5%<br />

4%<br />

KPS < 80% 7% 53% �0.0001<br />

Prior nephrectomy 95% 65% �0.0001<br />

>1 site <strong>of</strong> metastases 73% 83% 0.02<br />

Best response<br />

�0.0001<br />

Complete response<br />

4%<br />

0%<br />

<strong>Part</strong>ial response<br />

34%<br />

4%<br />

Stable disease<br />

53%<br />

19%<br />

Progressive disease<br />

3%<br />

43%<br />

Unknown<br />

6%<br />

34%<br />

Median duration <strong>of</strong> targeted<br />

therapy (months)<br />

23.6 2.0 -<br />

Median duration <strong>of</strong> second-line<br />

11.5<br />

0.8<br />

-<br />

targeted therapy (months)<br />

(N�90)<br />

(N�20)<br />

Median overall survival (months) 69.3 3.1 -<br />

LBA4537 Poster Discussion Session (Board #16), Sat, 8:00 AM-12:00 PM<br />

and 12:00 PM-1:00 PM<br />

Axitinib versus sorafenib as second-line therapy in Asian patients with<br />

metastatic renal cell carcinoma (mRCC): Results from a registrational<br />

study. Presenting Author: Shukui Qin, Nanjing Bayi Hospital, Nanjing,<br />

China<br />

The full, final text <strong>of</strong> this abstract will be available at<br />

abstract.asco.org at 12:01 AM (EDT) on Saturday, June 2,<br />

2012, and in the <strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong> online<br />

supplement to the June 20, 2012, issue <strong>of</strong> Journal <strong>of</strong><br />

<strong>Clinical</strong> Oncology. Onsite at the <strong>Meeting</strong>, this abstract will<br />

be printed in the Saturday edition <strong>of</strong> ASCO Daily News.<br />

4539 Poster Discussion Session (Board #18), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Phase III randomized sequential open-label study to evaluate efficacy and<br />

safety <strong>of</strong> sorafenib (SO) followed by sunitinib (SU) versus sunitinib followed<br />

by sorafenib in patients with advanced/metastatic renal cell carcinoma<br />

without prior systemic therapy (SWITCH Study): Safety interim analysis<br />

results. Presenting Author: Maurice Stephan Michel, University Hospital<br />

Mannheim, Mannheim, Germany<br />

Background: Several retrospective studies have investigated the sequential<br />

use <strong>of</strong> SO and SU. Some smaller trials support the use <strong>of</strong> SO followed by SU<br />

forming the rationale for this study. Methods: Pts with metastatic RCC<br />

unsuitable for cytokines without prior systemic therapy, ECOG PS 0/1,<br />

MSKCC score low or intermediate, and �1 measurable lesion (CT/MRI<br />

every 12 weeks) were randomized to SO-�SU or SU-�SO in standard<br />

dosage (primary endpoint total PFS from randomization to event during 2 nd<br />

line therapy). Treatment continues until progression or intolerability.<br />

Monitoring includes echocardiography and NT pro-BNP. Results: Baseline<br />

characteristics <strong>of</strong> 361 randomized pts (116 completed) are balanced<br />

between arms. Safety data <strong>of</strong> 333 pts are evaluable. AE occurring in �10%<br />

<strong>of</strong> pts are listed in tab. 1. Left-ventricular ejection fraction (LVEF) at<br />

screening, switch <strong>of</strong> treatment, and end <strong>of</strong> study are given in tab. 1. AE<br />

occurred in 93.4% and 92.8%; grade 3/4 AE in 59.9% and 50%; and SAE<br />

in 46.7% and 42.2% in the SO-�SU and SU-�SO arm, respectively.<br />

Updated results will be presented. Conclusions: AE frequencies are higher<br />

in 1 st than in 2nd line treatments. Typical AE pr<strong>of</strong>iles for SO and SU are<br />

observed. LVEF values are in a similar range.<br />

SO 1st SU 2nd SU 1st SO 2nd<br />

(n�167) % % (n�71) % % (n�166) % % (n�47) % %<br />

CTCAE All G 3/4 All G 3/4 All G 3/4 All G 3/4<br />

Hematotoxicity 6.6 1.2 19.7 5.6 16.9 9.0 0 0<br />

Hypertension 26.9 7.2 7.0 1.4 29.5 9.0 4.3 2.1<br />

Fatigue 28.1 5.4 22.5 2.8 31.3 5.4 17.0 0<br />

Weight loss 12.6 0.6 4.2 0 4.2 0.6 4.3 0<br />

Hair loss 28.1 0 1.4 0 3.0 0 4.3 0<br />

Rash 10.8 1.2 4.2 0 3.0 0 6.4 0<br />

Hand-Foot Skin Rct. 41.3 13.8 7.0 1.4 15.1 4.2 21.3 8.5<br />

Diarrhea 45.5 4.8 14.1 2.8 30.1 1.2 29.8 4.3<br />

Mucositis 4.8 0 4.2 0 13.8 2.4 2.1 0<br />

Nausea 18.6 1.2 15.5 1.4 22.9 0.6 4.3 0<br />

Vomiting 6.6 0 12.7 2.8 14.5 1.8 4.3 0<br />

SO->SU N Mean LVEF % �SD<br />

Screening 151 63.0 � 7.6<br />

Day <strong>of</strong> stopping 1st-line treatment 46 62.7 � 8.9<br />

End <strong>of</strong> study<br />

SU->SO<br />

23 60.3 � 10.5<br />

Screening 149 64.1 � 8.2<br />

Day <strong>of</strong> stopping 1st-line treatment 28 62.6 � 10.1<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


4540 Poster Discussion Session (Board #19), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

The use <strong>of</strong> tumor growth rate (TGR) in evaluating sorafenib and everolimus<br />

treatment in mRCC patients: An integrated analysis <strong>of</strong> the TARGET and<br />

RECORD phase III trials data. Presenting Author: Charles Ferte, Department<br />

<strong>of</strong> Medical Oncology, Institut Gustave Roussy, Villejuif, France<br />

Background: RECIST criteria may not be sufficient to evaluate targeted<br />

therapies in mRCC. TGR includes the time between each evaluation and is<br />

expected to overcome some <strong>of</strong> the RECIST pitfalls. How TGR is modified<br />

under targeted therapies and how it correlates with outcome in mRCC<br />

remains unknown. Methods: Medical records from all patients (pts) prospectively<br />

treated at Institut Gustave Roussy (IGR) in the TARGET (n�84) and<br />

in the RECORD (n� 52) phase III trials were extracted. Results were<br />

subsequently validated in the entire TARGET cohort (TARGET) (n�902).<br />

TGR was computed by dividing tumor shrinkage by the time between the<br />

two related evaluations (% RECIST x 100 / days). Typical treatment periods<br />

were assessed: BEFORE treatment (wash-out), UNDER treatment (first<br />

cycle), at PROGRESSION (pts still receiving the drug) and AFTER treatment<br />

interruption (wash-out). Results: As compared to placebo, TGR<br />

UNDER was significantly decreased following sorafenib (-23.6 vs. 20 (IGR)<br />

and -19 vs. 22 (TARGET)) and everolimus (-5.2 vs. 30 (IGR)). The great<br />

majority <strong>of</strong> pts (IGR) had a decrease in the TGR UNDER vs. BEFORE,<br />

regardless <strong>of</strong> the RECIST evaluation, both with sorafenib (28/29) or<br />

everolimus (36/37). TGR AFTER sorafenib or everolimus interruption was<br />

significantly higher than TGR at PROGRESSION in both settings (IGR)<br />

(14.6 vs. 31 and 17.9 vs. 32.1 respectively). No significant difference was<br />

observed between TGR AFTER vs. BEFORE either in sorafenib or in<br />

everolimus pts (IGR). High TGR UNDER is associated with poor progression<br />

free survival (HR� 2.6) and overall survival (HR� 2.3) in sorafenib-treated<br />

pts (multivariate analysis, trained in IGR and validated in TARGET cohorts)<br />

and with poor overall survival in everolimus-treated pts (IGR)(HR� 1.2).<br />

Conclusions: Adding TGR assessment in mRCC unravels interesting traits<br />

that make it potentially adaptable for clinical practice: (i) better evaluation<br />

<strong>of</strong> the tumor response, regardless <strong>of</strong> RECIST criteria, (ii) independent<br />

prognosis value, (iii) it suggests that maintaining sorafenib or everolimus<br />

after disease progression might benefit to patients and should be prospectively<br />

evaluated.<br />

4542 Poster Discussion Session (Board #21), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Sunitinib objective response (OR) in metastatic renal cell carcinoma<br />

(mRCC): Analysis <strong>of</strong> 1,059 patients treated on clinical trials. Presenting<br />

Author: Ana M. Molina, Memorial Sloan-Kettering Cancer Center, New<br />

York, NY<br />

Background: Sunitinib achieves robust OR and improved progression-free<br />

survival (PFS) in mRCC patients (N Engl J Med 2007;356:115). A<br />

retrospective analysis was performed to characterize the OR rate with<br />

sunitinib treatment (Tx) and OR-associated patient features and survival.<br />

Methods: Data from six phase II and III trials were pooled from 1,059<br />

patients who received sunitinib on the approved 50 mg/d 4-week-on/2-week<strong>of</strong>f<br />

schedule (n�689; 65%) or at 37.5 mg continuous once-daily dosing<br />

(n�370; 35%), in both the 1st- (n�783; 74%) and 2nd-line (n�276;<br />

26%) Tx settings. Median PFS and overall survival (OS) were estimated by<br />

the Brookmeyer and Crowley method, and compared between responders<br />

and nonresponders, and early and late responders (response � and �12<br />

weeks, respectively), by a log-rank test. Baseline characteristics were<br />

compared by Fisher-exact Test, T-Test, or Wilcoxon Rank-sum Test.<br />

Results: Of 1,059 patients, 398 (38%) had a confirmed investigatorassessed<br />

OR by RECIST (including 12 with a complete response), <strong>of</strong> whom<br />

105 (26%), 243 (61%), 314 (79%), and 342 (86%) had a response by 6,<br />

12, 18, and 24 weeks, respectively. Median (range) time to tumor response<br />

(TTR) in all patients was 10.6 (2.7–94.4) weeks, which was similar in the<br />

1st- and 2nd-line Tx settings. Responders had better baseline ECOG<br />

scores, more favorable risk factor classification based on published MSKCC<br />

data, a longer interval since initial diagnosis, more prior nephrectomy, and<br />

less presence <strong>of</strong> baseline bone metastases (all p�0.05). Early responders<br />

had more lung metastases (p�0.01). Median PFS (16.3 vs. 5.3 months)<br />

and OS (40.1 vs. 14.5 months) were significantly longer in responders vs.<br />

nonresponders (both p�0.001), with similar results regardless <strong>of</strong> 1st- or<br />

2nd-line Tx setting. Median OS did not differ between early and late<br />

responders (p�0.1438). Conclusions: OR was achieved in 38% <strong>of</strong> 1,059<br />

mRCC patients treated with sunitinib and was predicted by favorable<br />

pretreatment prognostic factors. Patients with OR had longer PFS and OS.<br />

Median TTR was 10.6 weeks, and characteristics and outcome <strong>of</strong> early and<br />

late responders were similar, except for higher frequency <strong>of</strong> lung metastases<br />

among early responders.<br />

Genitourinary Cancer<br />

4541 Poster Discussion Session (Board #20), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

PFS to predict long-term OS after first-line treatment for advanced renal<br />

cell carcinoma (aRCC): Correlation and power analysis <strong>of</strong> randomized trials<br />

(RCT). Presenting Author: Michele Milella, Regina Elena National Cancer<br />

Institute, Rome, Italy<br />

Background: Targeted agents (TA) have become standard 1st line aRCC<br />

treatment based on evidence <strong>of</strong> PFS advantage. Retrospective series<br />

indicatePFS as a reliable intermediate end-point in this setting; however,<br />

correlation, surrogacy testing, and validation are required. Methods: RCT<br />

evaluating the efficacy <strong>of</strong> TA as 1st line treatment for aRCC were eligible.<br />

PFS/OS Response/Disease Control rates (ORR, DCR) and HR were extracted<br />

from papers/updated presentations. Correlations between 6-, 9-,<br />

and 12-mo PFS and OS rates according to parametric (Pearson’s r) and<br />

non-parametric (Spearman’s Rho and Kendall’s Tau) coefficients (with<br />

95% CI) were analyzed to avoid lead-time biases. Regression analysis<br />

(parametric R2) and a power-analysis-model to determine patients’ sample<br />

necessary to detect 3%, 5% and 10% OS gain were developed. Results: Six<br />

RCT (4096 pts) were gathered. The best overall correlation between PFS<br />

and OS at concurrent timepoints was found at 9 mos. With regard to overall<br />

rates, 3- and 6-mo PFS significantly correlated with 9-mo OS, as shown in<br />

the table below. Pearson’s coefficients for the correlation between 3-mo<br />

PFS and 6- and 12-mo OS were 0.70 (p�0.01) and 0.67 (p�0.01); the<br />

correlation between 6-mo PFS and 12-mo OS was also significant (Pearson<br />

0.74, p�0.005; Spearman 0.83, p�0.005; Tau 0.71, p�0.001). The<br />

regression equation was: Y�0.391861 � 0.4914X [R2 0.44,<br />

p(slope)�0.01]; based on this model, the demonstration <strong>of</strong> a 3-mo PFS<br />

absolute difference <strong>of</strong> 6%, 10% and 21% (corresponding to a 9-mo OS<br />

benefit <strong>of</strong> 3%, 5% and 10%) would require 2043, 696 and 155 patients,<br />

respectively. A significant correlation was also found between DCR and OS.<br />

Conclusions: Early PFS is an acceptable intermediate end-point for OS in<br />

the context <strong>of</strong> 1st line TA for aRCC. Individual patient data analysis to verify<br />

Prentice criteria would be required for definitive confirmation.<br />

Correlation coefficient (p value)<br />

Sample<br />

Pearson Rho Tau<br />

Overall 0.66 (0.01) 0.78 (�0.01) 0.67 (0.03)<br />

0.75 (0.004) 0.91 (0.002) 0.79 (0.004)<br />

TA arm 0.69 (n.s.)<br />

0.94 (0.03) 0.85 (0.02)<br />

0.90 (0.01) 0.90 (0.04) 0.82 (0.03)<br />

Control arm 0.76 (n.s.)<br />

0.84 (n.s.)<br />

0.69 (n.s.)<br />

0.68 (n.s.)<br />

0.89 (0.04) 0.82 (0.03)<br />

4543 Poster Discussion Session (Board #22), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Association <strong>of</strong> inherited genetic variation with clinical outcome in patients<br />

with advanced renal cell carcinoma treated with mTOR inhibition. Presenting<br />

Author: Mark M. Pomerantz, Lank Center for Genitourinary Oncology,<br />

Dana-Farber Cancer Institute/Brigham and Women’s Hospital, Harvard<br />

Medical School, Boston, MA<br />

Background: Mammalian target <strong>of</strong> rapamycin (mTOR)-targeted therapy is<br />

standard in patients with metastatic renal cell carcinoma (mRCC). Predictive<br />

biomarkers for response are lacking. We sought to characterize<br />

outcome after mTOR inhibition based on germline variation in 2 critical<br />

genes in the mTOR pathway: phosphoinositide-3-kinase catalytic alpha<br />

(PIK3CA) and mTOR. Methods: 76 <strong>American</strong>s <strong>of</strong> European ancestry treated<br />

with temsirolimus or everolimus for mRCC were included. Germline DNA<br />

was genotyped for all common genetic variation (minor allele frequency<br />

[MAF] �0.05) across PIK3CA and mTOR, tagging with single nucleotide<br />

polymorphisms (SNPs) at r2�0.8. Associations between genotype and<br />

progression-free survival (PFS) and overall survival (OS) from the start <strong>of</strong><br />

mTOR-targeted therapy were measured using univariate Cox model. Results:<br />

Among 76 patients, 66% were poor or intermediate risk and 89% received<br />

prior systemic therapies. Median follow up was 23.7 months. Median PFS<br />

and OS were 4.3 and 12.7 months, respectively. Two intronic PI3KCA<br />

SNPs were significantly associated with PFS and OS, and a SNP in the 5’<br />

UTR <strong>of</strong> mTOR was associated with OS. Associations were maintained when<br />

adjusted for age, gender and MSKCC RCC risk categories using Cox PH<br />

model (Table). The PI3KCA SNPs are in modest linkage disequilibrium (r 2<br />

~0.36). Conclusions: We suggest that inherited variation at PIK3CA is<br />

associated with PFS and OS after mTOR inhibition. If results are validated,<br />

prospective studies could explore the role <strong>of</strong> genotyping in treatment<br />

selection for mRCC. Further work will be needed to identify causal alleles<br />

and define the mechanism underlying the associations.<br />

PFS and OS after mTOR-targeted therapy by genotype.<br />

Gene SNP MAF<br />

Median PFS<br />

Genotype (months)<br />

HR<br />

(95% CI)<br />

Median OS<br />

P value* (months)<br />

HR<br />

(95% CI) P value*<br />

PIK3CA rs2699905 0.25 TC, TT 6 1 13.1 1<br />

CC 4.4 2.1 0.006 12.2 1.86 0.03<br />

(1.23,3.57) (1.05,3.3)<br />

PIK3CA rs13082485 0.08 AA, AG 10.6 1 33.9 1<br />

GG 4.2 3.1<br />

(1.37,7)<br />

0.007 12.2 4.11<br />

(1.46,11.61) 0.008<br />

mTOR rs12732063 0.07 AA, AG 3.5 1 7.7 1<br />

GG 4.8 0.58<br />

(0.28,1.2)<br />

0.14 12.9 0.36<br />

(0.16,0.8)<br />

Abbreviations: HR, hazard ratio; CI, confidence interval. * Adjusted for age, gender, MSKCC risk groups.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.<br />

287s<br />

0.01


288s Genitourinary Cancer<br />

4544^ Poster Discussion Session (Board #23), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Phase II trial <strong>of</strong> RAD001 in renal cell carcinoma patients with non-clear<br />

cell histology. Presenting Author: Youngil Koh, Department <strong>of</strong> Internal<br />

Medicine, Seoul National University College <strong>of</strong> Medicine, Seoul, South<br />

Korea<br />

Background: In non-clear cell renal cell carcinoma (ncRCC), the efficacy <strong>of</strong><br />

VEGF tyrosine kinase inhibitor (TKI) is controversial. In the while, a mTOR<br />

inhibitor, temsirolimus showed a promising efficacy in ncRCC patients in<br />

ARCC trial. Hence, we investigated the role <strong>of</strong> everolimus in ncRCC with<br />

this phase II trial. Methods: ncRCC patients received everolimus 10 mg<br />

once daily until disease progression or unacceptable toxicity. We included<br />

patients who had received VEGF TKI previously, while excluded patients<br />

who received previous mTOR inhibitor. The primary end point was progression<br />

free survival (PFS). Results: A total <strong>of</strong> 49 patients were enrolled from 5<br />

centers. Their median age was 57 years (range 24-75 years) and male to<br />

female ratio was 37:12. Histology <strong>of</strong> the patients included papillary<br />

(n�29), chromophobe (n�8), collecting duct (n�2), sarcomatoid (n�4),<br />

and unclassifiable (n�6) RCC. Twenty-three patients had been treated with<br />

VEGF-TKI prior to the study enrollment. Among 49 patients, 46 patients<br />

underwent radiologic response assessment after everolimus treatment.<br />

<strong>Part</strong>ial response was observed in 5 patients (10.2%) and stable disease in<br />

25 patients (51.0%). Diseases <strong>of</strong> 16 patients (32.7%) progressed despite<br />

<strong>of</strong> everolimus administration. Histology <strong>of</strong> 5 patients who showed objective<br />

response to everolimus included chromophobe carcinoma (n�2), papillary<br />

carcinoma (n�2) and unclassifiable carcinoma (n�1). During the study<br />

period, 34 patients experienced PFS events and median PFS was 5.2<br />

months. Patients with chromophobe histology showed longer PFS than<br />

patients with the other histologies (median PFS 18.8 months vs. 3.5<br />

months, p�0.027). Estimated median PFS were not significantly different<br />

between patients VEGF-TKI treatment and patients without previous<br />

VEGF-TKI treatment (median PFS 7.1 vs. 3.7 months, p�0.110). Toxicity<br />

pr<strong>of</strong>iles were commensurable with previous reports. Conclusions: Everolimus<br />

shows considerable efficacy in ncRCC. Patients with chromophobe<br />

histology might earn benefit from everolimus treatment especially. Previous<br />

treatment with VEGF-TKI seems not to significantly influence outcome <strong>of</strong><br />

everolimus therapy in these patients. (<strong>Clinical</strong>Trials.gov number,<br />

NCT00830895)<br />

4546 Poster Discussion Session (Board #25), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Phase III AXIS trial <strong>of</strong> axitinib versus sorafenib in metastatic renal cell<br />

carcinoma: Updated results among cytokine-treated patients. Presenting<br />

Author: M. Dror Michaelson, Massachusetts General Hospital Cancer<br />

Center, Boston, MA<br />

Background: Axitinib, a potent and selective second-generation inhibitor <strong>of</strong><br />

vascular endothelial growth factor receptors, improved progression-free<br />

survival (PFS) compared to sorafenib in patients with metastatic renal cell<br />

carcinoma (mRCC) in the phase III AXIS trial. In patients previously treated<br />

with cytokines, median PFS (mPFS) was 12.1 mo. In a prior phase II study<br />

<strong>of</strong> axitinib for cytokine-refractory mRCC, mPFS was 13.7 mo and the 5-yr<br />

overall survival (OS) rate was 20.6%. We report updated PFS and OS for<br />

cytokine-treated patients from the AXIS trial. Methods: 723 patients with<br />

clear-cell mRCC and progressive disease after 1 systemic therapy were<br />

enrolled, <strong>of</strong> whom 251 received prior interleukin-2 (IL-2) or interferon-�<br />

(IFN-�). Patients were randomized 1:1 to axitinib (5 mg twice daily [BID]<br />

starting dose) or sorafenib (400 mg BID). Results: As <strong>of</strong> Jun 3, 2011, mPFS<br />

(95% confidence interval [CI]) for cytokine-treated patients was 12.0 mo<br />

(10.1, 13.9) with axitinib (n�126) vs 6.6 mo (6.4, 8.3) with sorafenib<br />

(n�125): hazard ratio [HR] (95% CI) 0.519 (0.375, 0.720); p�0.0001.<br />

For those treated with an IL-2-containing regimen, mPFS (95% CI) was<br />

15.7 mo (8.3, 19.4) with axitinib (n�37) vs 8.3 mo (4.7, 15.7) with<br />

sorafenib (n�38). For those treated with IFN-� alone, mPFS (95% CI) was<br />

12.0 mo (10.0, 13.8) with axitinib (n�89) vs 6.5 mo (6.4, 8.2) with<br />

sorafenib (n�87). As <strong>of</strong> Nov 1, 2011, median OS (95% CI) in the<br />

cytokine-treated subgroup was 29.4 mo (24.5, not estimable) with axitinib<br />

vs 27.8 mo (23.1, 34.5) with sorafenib: HR (95% CI) 0.813 (0.555,<br />

1.191); p�0.144. Adverse event (AE) pr<strong>of</strong>iles were similar in both arms.<br />

Few cytokine-treated patients (5.6%) discontinued axitinib due to toxicity.<br />

AEs reported in �25% <strong>of</strong> cytokine-treated patients receiving axitinib were<br />

diarrhea (49.2%), hypertension (47.6%), fatigue (35.7%), dysphonia<br />

(29.4%), and hand–foot syndrome (28.6%). Conclusions: Axitinib showed<br />

median PFS and OS <strong>of</strong> 1 and 2.5 yr, respectively, in cytokine-treated<br />

patients, confirming prior phase II study results, and was well tolerated.<br />

PFS was superior to that <strong>of</strong> sorafenib in second-line mRCC and compares<br />

favorably with historical results <strong>of</strong> other agents in second-line mRCC.<br />

4545 Poster Discussion Session (Board #24), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Safety and efficacy <strong>of</strong> MET inhibitor tivantinib (ARQ 197) combined with<br />

sorafenib in patients (pts) with renal cell carcinoma (RCC) from a phase I<br />

study. Presenting Author: Igor Puzanov, Vanderbilt University Medical<br />

Center, Nashville, TN<br />

Background: Inhibitors <strong>of</strong> vascular endothelial growth factor (VEGF) and<br />

VEGF receptor are standard therapy for RCC, and the MET signaling<br />

pathway is implicated in tumor angiogenesis. Tivantinib is an oral, selective<br />

MET inhibitor. In several tumor models, tivantinib plus sorafenib exhibited<br />

synergistic antitumor activity vs single-agent activity. This phase I doseescalation<br />

study assessed the safety <strong>of</strong> tivantinib plus sorafenib in pts with<br />

advanced solid tumors. Methods: Endpoints were safety, the recommended<br />

phase II dose (RP2D) <strong>of</strong> tivantinib plus sorafenib, and antitumor activity.<br />

Previously, dose escalation established the RP2D as tivantinib 360 mg<br />

twice daily (BID) plus sorafenib 400 mg BID. Extension cohorts enrolled �<br />

20 pts each with RCC or other tumors. Patients were treated until disease<br />

progression or unacceptable toxicity. Results: 20 pts (mean age, 60 yr)<br />

including 16 clear cell, 3 papillary, and 1 clear cell/chromophobe RCC pts<br />

received treatment at the RP2D (n � 19) or tivantinib 360 mg BID plus<br />

sorafenib 200 mg BID (n � 1). 4 pts are still on study. 16 pts (13 with clear<br />

cell RCC) received � 1 previous systemic therapy (median, 2; range, 0-4)<br />

including VEGF (14 pts) and/or mTOR (5 pts) inhibitors. The most common<br />

(� 25%) adverse events were rash (65%), diarrhea (45%), alopecia (40%),<br />

hypophosphatemia (35%), and fatigue, stomatitis, palmar-plantar erythrodysesthesia<br />

syndrome, and pruritus (25% each). Best response was partial<br />

response (PR) in 3 pts (all clear cell RCC pts) and stable disease (SD) in 15<br />

pts (11 clear cell, 3 papillary, and 1 clear cell/chromophobe RCC pts). 7 pts<br />

with SD had � 10% tumor size reduction. The overall response rate (ORR)<br />

and disease control rate (DCR; PR � SD) were 15% and 90%, respectively.<br />

Median progression-free survival (mPFS) was 12.7 mo (95% CI, 7.1-14.5<br />

mo). In 14 pts previously treated with a VEGF inhibitor, best response was 2<br />

PR and 10 SD, the ORR and DCR were 14% and 86%, respectively, and<br />

mPFS was 12.7 mo (95% CI, 5.3-NR mo). Conclusions: Oral combination<br />

therapy with tivantinib plus sorafenib was well tolerated and exhibited<br />

preliminary anticancer activity in pts with RCC, including pts pretreated<br />

with VEGF inhibitors.<br />

4547 Poster Discussion Session (Board #1), Mon, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

SWOG 0421: Prognostic and predictive value <strong>of</strong> bone metabolism biomarkers<br />

(BMB) in castration resistant prostate cancer (CRPC) patients (pts) with<br />

skeletal metastases treated with docetaxel (DOC) with or without atrasentan<br />

(ATR). Presenting Author: Primo Lara, University <strong>of</strong> California, Davis,<br />

Sacramento, CA<br />

Background: S0421, a phase III study <strong>of</strong> DOC �/- the endothelin antagonist<br />

ATR in CRPC pts with bone metastases, showed no overall survival (OS)<br />

benefit for DOC�ATR. While BMB may have a prognostic role in CRPC,<br />

their predictive role vis a vis bone-targeted therapy such as ATR is<br />

unknown. We prospectively assessed pre-treatment serum BMB from<br />

S0421 pts to validate their prognostic and predictive value. Methods: BMB<br />

for resorption (N-telopeptide, NTX and Pyridinoline, PYD) and formation<br />

(C-terminal collagen propeptide, CICP and bone alkaline phosphatase,<br />

BAP) were assayed [Quidel (PYD, CICP, BAP) and Wampole (NTX)]. Cox<br />

regression models for OS based on BMB adjusted for clinical variables were<br />

developed. An adjusted Cox model was fit with main effects and BMB x<br />

Treatment interaction to assess predictive value <strong>of</strong> ATR on OS. Results: Of<br />

1,038 pts, 855 (82%) submitted baseline serum: 778 (91%) were usable<br />

and analyzable. Pt characteristics: median age � 69 years; PS 0-1 � 91%,<br />

Bisphosphonate use � 61%; Gleason �7 � 56%; median PSA � 68; and<br />

bone mets only � 45%. BMB values (median; range): NTX (14 nM;<br />

9.3-23.9), BAP (64.7 u/L; 35-164), CICP (9.5 ng/mL; 6.5-17.4), and PYD<br />

(2.8 nmol/L; 2.2-3.9). Table below shows prognostic role <strong>of</strong> BMB. Pts with<br />

very high BMB (upper 25 %ile, n�47) not only have poor prognosis (HR �<br />

4.3, p�0.001) but have OS benefit from ATR (HR�0.34, median OS �<br />

13.6 vs. 6.7 months; interaction p�0.002**). Conclusions: S0421 validates<br />

the strong independent OS prognostic value <strong>of</strong> BMB in CRPC. Very<br />

high BMB levels appear to be significantly predictive <strong>of</strong> OS benefit with<br />

ATR. Further study <strong>of</strong> endothelin antagonists in CRPC should focus on this<br />

high-risk pt subset. (5R01-CA120469)<br />

Biomarker<br />

Hazard ratio<br />

(95% CI)<br />

Median survival,<br />

months ( median) p-value*<br />

BAP (u/l) 1.23 (1.14, 1.32) 22.8 vs. 15.4 �0.001<br />

CICP (ng/ml) 1.38 (1.26, 1.51) 24.5 vs. 14.6 �0.001<br />

NTx (nM) 1.40 (1.27, 1.54) 22.4 vs. 15.5 �0.001<br />

PYD (nmol/l) 1.52 (1.28, 1.81) 22.0 vs. 15.3 �0.001<br />

* Significance set at � 0.006 to control 2-sided error rate at 0.05. **Significance set at<br />

�0.01 to control overall 2-sided error rate at 0.05 (Bonferroni adjustment).<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


4548 Poster Discussion Session (Board #2), Mon, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

A phase I study <strong>of</strong> the androgen signaling inhibitor ARN-509 in patients<br />

with metastatic castration-resistant prostate cancer (mCRPC). Presenting<br />

Author: Dana E. Rathkopf, Sidney Kimmel Center for Prostate and Urologic<br />

Cancers, Memorial Sloan-Kettering Cancer Center, New York, NY<br />

Background: ARN-509 is a novel small molecule AR antagonist that impairs<br />

AR nuclear translocation and binding to DNA, inhibiting tumor growth and<br />

promoting apoptosis, with no partial agonist activity. (Clegg et al., 2012)<br />

We conducted a phase I trial to assess safety, pharmacokinetics (PK), and<br />

determine the recommended phase II dose (RP2D). Methods: Eligible<br />

patients with mCRPC received ARN-509 orally on a continuous daily dosing<br />

schedule. Seven doses (30, 60, 90, 120, 180, 240, and 300 mg) were<br />

tested using standard 3x3 dose escalation criteria. Once drug concentrations<br />

were achieved that met or exceeded optimal levels predicted<br />

preclinically, an additional 2 dose levels were tested to further confirm the<br />

safety margin <strong>of</strong> ARN-509 (390 and 480 mg). Anti-tumor activity was<br />

assessed by PSA, radiographic responses, and FDHT-PET imaging. Results:<br />

Thirty patients were enrolled. The most common grades 1-2 treatmentrelated<br />

adverse events were fatigue (38%), nausea (29%), and pain (24%).<br />

There was only 1 treatment-related grade 3 adverse event (abdominal pain)<br />

at 300 mg, possibly related to a higher pill burden. PK was shown to be<br />

linear and dose-dependent. At 12 weeks, 42% <strong>of</strong> patients have had � 50%<br />

PSA declines. Eleven (37%) patients have discontinued the study due to<br />

progression, with the longest patient still on study for more than 16<br />

months. FDHT-PET imaging demonstrated AR blockade at 4 weeks across<br />

multiple dose levels. Conclusions: In this phase I study, ARN-509 was<br />

shown to be safe and well tolerated with linear PK. Based on promising<br />

activity across all dose levels and pharmacodynamic evidence <strong>of</strong> AR<br />

antagonism, an optimal biologic dose <strong>of</strong> 240 mg daily was selected for<br />

phase II investigation. DOD/PCF PCCTC trial sponsored by Aragon Pharmaceuticals.<br />

4550 Poster Discussion Session (Board #4), Mon, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Tasquinimod and survival in men with metastatic castration-resistant<br />

prostate cancer: Results <strong>of</strong> long-term follow-up <strong>of</strong> a randomized phase II<br />

placebo-controlled trial. Presenting Author: Andrew J. Armstrong, Duke<br />

Cancer Institute, Durham, NC<br />

Background: Tasquinimod (T) is an oral quinoline-3-carboxamide derivative<br />

that binds S100A9 protein and has preclinical anti-angiogenic and<br />

anti-tumor activity. Between 12/07-6/09, 201 (134 T, 67 Placebo (P))<br />

men with metastatic CRPC were randomized and received treatment<br />

once-daily at an initial dose <strong>of</strong> 0.25 mg/day escalated to 1.0 mg/day over 4<br />

weeks. Placebo patients could cross over to T after 6 months or at disease<br />

progression. The primary endpoint <strong>of</strong> improved PCWG2 criteria-defined<br />

progression at 6 months was met (69 vs. 37% <strong>of</strong> patients (T/P) were<br />

progression free) with PFS <strong>of</strong> 7.6 vs. 3.3 months for pts on T vs. P1 with<br />

acceptable toxicity. This abstract provides the first analysis on symptomatic<br />

progression, overall survival (OS) as well as a multivariate analysis for PFS<br />

and OS. Methods: Survival data were collected between June 2011 and<br />

January 2012 with a median time to censoring <strong>of</strong> 32 months. Survival data<br />

was also evaluated in an exploratory multivariate model <strong>of</strong> known prognostic<br />

factors in CRPC. Results: An imbalance <strong>of</strong> several baseline prognostic<br />

criteria favored placebo (e.g. baseline PSA <strong>of</strong> 29 vs. 19 (T/P)) (JCO<br />

2011;20:4022). Time to symptomatic progression was longer in T treated<br />

patients (p�0.039, HR�0.42). Record <strong>of</strong> death (97 events) or survival<br />

�13 months was documented in 182 patients. Median time to death was<br />

34.2 vs. 30.2 months (T/P). Median time to death in the PCWG2<br />

bone-metastatic subgroup (N�92/44) was 34.2 vs. 25.6 months. A<br />

multivariate analysis <strong>of</strong> known prognostic factors including PSA, LDH, PSA<br />

kinetics, and hemoglobin demonstrated an adjusted HR for PFS <strong>of</strong> 0.54<br />

(95% CI 0.37,0.81) and OS <strong>of</strong> 0.72 (95% CI 0.46,1.12) in the total<br />

population and 0.63 (95% CI 0.37,1.07, n�136) in the bone-metastatic<br />

group. Conclusions: OS observed after tasquinimod treatment is longer than<br />

previously reported in this patient population. The current exploratory data<br />

indicates that the prolongation in PFS observed with tasquinimod treatment<br />

may lead to a survival advantage in men with metastatic CRPC. A<br />

phase III placebo-controlled study (NCT01234311) is ongoing in men with<br />

bone-metastatic CRPC powered to detect an OS improvement.<br />

Genitourinary Cancer<br />

289s<br />

4549 Poster Discussion Session (Board #3), Mon, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Safety and activity <strong>of</strong> the investigational agent orteronel (ortl) without<br />

prednisone in men with nonmetastatic castration-resistant prostate cancer<br />

(nmCRPC) and rising prostate-specific antigen (PSA): Updated results <strong>of</strong> a<br />

phase II study. Presenting Author: Daniel J. George, Duke University<br />

Medical Center, Durham, NC<br />

Background: Ortl is an investigational, oral, non-steroidal selective 17,20lyase<br />

inhibitor that suppresses androgen production. Ortl affects cortisol<br />

synthesis less than similar agents due to limited inhibition <strong>of</strong> 17�hydroxylase,<br />

and may permit steroid-free dosing. Ortl 300 mg BID was<br />

examined in patients (pts) with nmCRPC and rising PSA. Methods: Eligible<br />

pts had nmCRPC with PSA �2 ng/mL (PSA �8 ng/mL if doubling time �8<br />

mo), and surgical/medical castration, with testosterone (T) �50 ng/dL.<br />

Prior chemotherapy, ketoconazole, or concomitant corticosteroids were<br />

excluded. Starting dose <strong>of</strong> ortl was 300 mg BID and continued until PSA<br />

progression, metastases, or unacceptable toxicity. The primary endpoint<br />

was the percentage <strong>of</strong> pts with PSA �0.2 ng/mL after 3 mo. Secondary<br />

endpoints included safety, PSA kinetics, time to metastases, changes in<br />

endocrine markers and circulating tumor cells (CTCs). Results: 39 pts were<br />

enrolled with baseline demographics including median age 71 y, ECOG PS<br />

�1, median PSA 12.1 ng/mL (range 2.6-67.8), T 7.9 ng/dL (1.4–17.3),<br />

and ACTH 19 ng/L (n�33; 0-47); 3 pts had dose reduction due to adverse<br />

events (AEs). Gr �3 AEs occurred in 16 pts (drug-related in 14); Gr �3 AEs<br />

�5% were dyspnea (8%), hypertension (13%), fatigue, hypokalemia,<br />

pneumonitis (5% ea). 7 pts (18%) had serious AEs; most common was<br />

pneumonitis (2�Gr 3, 1�Gr 2). 8 pts discontinued due to AEs; 2 pts<br />

required corticosteroids. At 3 mo, 6 pts (16%) achieved PSA �0.2 ng/mL;<br />

PSA50 and PSA90 rates were 76% and 32%, respectively; median PSA<br />

declined by 83% (n�34); median T declined by 89% to 0.78 ng/dL<br />

(n�31), and median ACTH increased by 171% to 43 ng/L; median cortisol<br />

declined by 21%. At 6 mo, PSA50 and PSA90 rates were 45% and 21%,<br />

respectively. Median time to PSA progression was 14.8 mo. 17 pts (44%)<br />

were on treatment �6 mo. Of 37 pts with baseline CTC/7.5 mL assessed,<br />

only 1 had CTC �5 and converted to �5; 6 had 1–4 CTCs at baseline, none<br />

converted to �5 during treatment. Conclusions: Ortl without steroids<br />

produces marked and durable declines in T and PSA, has manageable<br />

toxicities, and is feasible in men with nmCRPC.<br />

4551 Poster Discussion Session (Board #5), Mon, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Radium-223 chloride (Ra-223) impact on skeletal-related events (SREs)<br />

and ECOG performance status (PS) in patients with castration-resistant<br />

prostate cancer (CRPC) with bone metastases: Interim results <strong>of</strong> a phase III<br />

trial (ALSYMPCA). Presenting Author: A. Oliver Sartor, Tulane Cancer<br />

Center, New Orleans, LA<br />

Background: Ra-223, a 1st-in-class alpha-pharmaceutical, targets bone<br />

metastases (mets) with high-energy alpha-particles <strong>of</strong> short range (�100<br />

�m). ALSYMPCA, a phase III, double-blind, randomized, multinational<br />

study, compared Ra-223 plus best standard <strong>of</strong> care (BSC) v placebo (pbo)<br />

plus BSC in patients (pts) with bone mets in CRPC. The primary endpoint<br />

was OS; secondary endpoints included SREs and ECOG PS. Methods:<br />

Eligible pts had progressive, symptomatic CRPC with � 2 bone mets on<br />

scintigraphy and no known visceral mets; were receiving BSC; and either<br />

previously received docetaxel, were docetaxel ineligible, or refused docetaxel.<br />

Pts were randomized 2:1 to 6 injections <strong>of</strong> Ra-223 (50 kBq/kg IV)<br />

q4 wks or matching pbo and stratified by prior docetaxel use, baseline ALP<br />

level, and current bisphosphonate use. Results: 921 pts were randomized<br />

from 6/2008-2/2011. In a planned interim analysis (n � 809), Ra-223<br />

significantly improved OS v pbo (median OS 14.0 v 11.2 mo, respectively;<br />

two-sided P � .00185; HR � .695; 95% CI, .552-.875).SREs were lower<br />

in the Ra-223 v pbo group, and time to 1st SRE was significantly delayed<br />

(median time to SRE 13.6 mo v 8.4 mo, respectively; P � .00046; HR �<br />

.610; 95% CI, .461-.807). The proportion <strong>of</strong> pts with ECOG PS deterioration<br />

(� 2 points) was less in Ra-223 v pbo group at Wk 12 and Wk 24 (4%,<br />

15/389 v 9%, 16/180 and 7%, 16/236 v 12%, 10/83, respectively). Time<br />

to ECOG PS deterioration (� 2 points) was significantly delayed by Ra-223<br />

v pbo (P � .003; HR � .62; 95% CI, .46-.85). Conclusions: Ra-233<br />

significantly delayed time to 1st SRE and SRE components, notably SCC.<br />

Fewer pts in the Ra-223 group had ECOG PS deterioration. Ra-223<br />

improves OS with excellent safety and may provide a new standard <strong>of</strong> care<br />

for CRPC pts with bone mets.<br />

Time to 1st event<br />

No. (%) <strong>of</strong> patients<br />

(Ra-223 vs Pbo)<br />

Ra-223 Pbo<br />

HR<br />

SRE component<br />

n � 541 n � 268 p value* (95%CI)<br />

External beam radiotherapy 122 (23) 72 (27) .0038 .65<br />

(.48-.87)<br />

Spinal cord compression 17 (3) 16 (6) .016 .44<br />

(.22-.88)<br />

Pathologic bone fracture 20 (4) 18 (7) .013 .45<br />

(.24-.86)<br />

Surgical intervention 9 (2) 5 (2) .69 .80<br />

(.27-2.4)<br />

*Not adjusted for multiplicity.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


290s Genitourinary Cancer<br />

4552 Poster Discussion Session (Board #6), Mon, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Development <strong>of</strong> a nomogram model using a very large sample <strong>of</strong> patients<br />

(pts) to predict the risk <strong>of</strong> 99mTc-bone scan (BS) positivity in pts receiving<br />

androgen deprivation therapy (ADT) for prostate cancer (PCa). Presenting<br />

Author: Ge<strong>of</strong>frey Gotto, Southern Alberta Institute <strong>of</strong> Urology, Calgary, AB,<br />

Canada<br />

Background: Decisions to start imaging pts with PCa can be difficult due to<br />

the heterogeneous nature <strong>of</strong> the disease. Nomograms can combine information<br />

on multiple disease factors to improve predictive accuracy and are not<br />

influenced by subjective confounders that may affect clinical judgment.<br />

However, currently available nomograms for PCa predict current or future<br />

risk <strong>of</strong> positive BS in ADT-naive pts only. We have developed a new<br />

nomogram to predict current BS positivity in ADT-treated pts with PCa<br />

using a very large pt sample. Methods: All BS records from 1650<br />

ADT-treated pts who received treatment at the Memorial Sloan-Kettering<br />

Cancer Center from 2000 to 2011 were analyzed. Pts were followed up<br />

from the start <strong>of</strong> ADT until the first positive BS or last hospital visit.<br />

Multivariate logistic regression analysis was used to model the variables<br />

that could be used for predicting likelihood <strong>of</strong> metastases and the variables<br />

were incorporated into the nomogram model. The current probability <strong>of</strong><br />

bone metastasis was generated by the nomogram using the pt variables at<br />

each BS and the predictive accuracy was quantified by calculating the<br />

concordance index (C-index). Results: In total, 3949 BS records were<br />

analyzed and 950 pts had a positive scan. There was an average <strong>of</strong> 1.1 prior<br />

negative scans before a positive result was recorded. Median prostatespecific<br />

antigen (PSA) was 2.5 ng/mL and mean PSA doubling time<br />

(PSADT) was 7.8 months. At the same time <strong>of</strong> the positive scans, 49.4% <strong>of</strong><br />

pts had a PSA �10 ng/mL. Based on clinical relevance and data<br />

availability, 13 variables were included in the nomogram model: number <strong>of</strong><br />

previous negative BSs, PSA, PSADT, prostatectomy, radiotherapy, brachytherapy,<br />

chemotherapy, immunotherapy, estrogen therapy, cryotherapy,<br />

clinical trial enrollment, and most recent primary and secondary Gleason<br />

grades. C-index for the nomogram was 0.76. Conclusions: This is the first<br />

nomogram to predict current BS positivity in pts with PCa following ADT.<br />

The nomogram was devised from a very large data set from a single center<br />

and provides high predictive accuracy.<br />

4554 Poster Discussion Session (Board #8), Mon, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Abiraterone in patients with metastatic castration-resistant prostate cancer<br />

progressing after docetaxel and MDV3100. Presenting Author: Ecaterina<br />

Ileana, Institut Gustave Roussy, Villejuif, France<br />

Background: Chemotherapy with docetaxel is the standard first-line treatment<br />

in patients with metastatic castration-resistant prostate cancer<br />

(mCRPC). In patients progressing after docetaxel, both abiraterone and<br />

MDV3100 have yielded improved survival for patients with mCRPC. The<br />

efficacy <strong>of</strong> abiraterone in patients pre-treated with MDV 3100 is unknown.<br />

Methods: We investigated abiraterone-prednisone in 24 patients with<br />

cancer progression after docetaxel followed by MDV3100. All patients<br />

received abiraterone 1000 mg/day plus prednisone 10mg/day. Prostatespecific<br />

antigen (PSA) response, symptom response, and time to progression<br />

were assessed. Results: Patient characteristics were as follows: median<br />

age: 74 years (53-84), median PSA: 108 ng/mL (2-2541), metastatic<br />

sites: bone: all 24 patients, liver/lung: 6 patients (25%), and lymph nodes :<br />

9 patients (38%). Five patients (21%) had a PSA decrease on abirateroneprednisone.<br />

Three patients (13%) achieved a PSA response, defined as a<br />

decrease <strong>of</strong> �50% in PSA, confirmed after� 4 weeks. The duration <strong>of</strong> PSA<br />

response was 2, 3 and 4.5 months. Six patients (29%) had a symptomatic<br />

response on the pain score and analgesic consumption was decreased.<br />

Treatment was well tolerated. Abiraterone-prednisone was discontinued in<br />

one patient due to edema and hypokaliemia. Conclusions: This study shows<br />

preliminary evidence that abiraterone-prednisone yields activity in patients<br />

with mCRPC pretreated with docetaxel and MDV3100.<br />

4553 Poster Discussion Session (Board #7), Mon, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Durable radiologic and clinical disease stability beyond PSA progression in<br />

patients with metastatic castration-resistant prostate cancer (mCRPC)<br />

treated with abiraterone acetate (AA). Presenting Author: Diletta Bianchini,<br />

The Royal Marsden Hospital and The Institute <strong>of</strong> Cancer Research, Sutton,<br />

United Kingdom<br />

Background: AA, a potent oral CYP17A1 inhibitor is approved for treatment<br />

<strong>of</strong> mCRPC with a survival advantage <strong>of</strong> 4.9 months. In clinical practice,<br />

response evaluation remains challenging for pts with mCRPC. CTC conversion<br />

from CTC � 5toCTC�5with treatment predicts for improved overall<br />

survival in mCRPC. We hypothesized that pts continue to have durable<br />

disease stability beyond PSA progression on AA. Methods: Prostate Specific<br />

Antigen (PSA) responses, radiological responses and CTC conversion rates<br />

were retrospectively analysed in pts treated on AA at our institution. CTCs,<br />

PSA and imaging were obtained at predefined time points during these<br />

studies. Radiological and PSA progression were defined by standard<br />

Prostate Cancer Working Group Criteria II. <strong>Clinical</strong> progression consisted <strong>of</strong><br />

worsening disease related pain, skeletal events or declining performance<br />

status.Pearson’s chi-squared test and the Kaplan-Meier method were used<br />

for this analysis. Results: 141 patients [ECOG Performance Status 0-2;<br />

Median Age: 69.7 (range 44.7-87.1); 85 post-docetaxel, 56 predocetaxel]<br />

received AA. The median duration <strong>of</strong> clinical and radiological<br />

stable disease (SD) was 16.8 months (n�55) and 5.6 months (n�75) in<br />

patients with a baseline CTCs count <strong>of</strong> � 5 cells/7.5mls and � 5 cells/7.5<br />

mls respectively. In the 105 patients with documented PSA progression on<br />

AA there was a median 5.7-month delay in detecting radiological and/or<br />

clinical progression (95% CI: 4.2, 8.4; range 0.3, 35.6 months). Radiological<br />

and clinical SD <strong>of</strong> � 1 year, � 2 years and � 3 years on AA was observed<br />

in 43/141 (30.5%), 21/141 (14.9%) and 12/141 (8.5%) respectively.<br />

Conclusions: Radiological and clinical disease stabilization beyond PSA<br />

progression is maintained in a high proportion <strong>of</strong> mCRPC patients treated<br />

with AA. Future studies should evaluate whether continued AA treatment<br />

beyond PSA and radiological progression can impact outcome.<br />

4555 Poster Discussion Session (Board #9), Mon, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Association <strong>of</strong> metabolic syndrome with poorer prostate cancer and overall<br />

survival in men receiving androgen deprivation therapy (ADT) for biochemical<br />

relapse. Presenting Author: Sarah Maria Rudman, Division <strong>of</strong> Cancer<br />

Studies, King’s College London, Guy’s Hospital, London, United Kingdom<br />

Background: The metabolic syndrome (MS) has been implicated in the<br />

development <strong>of</strong> prostate cancer (PC). In our previous study <strong>of</strong> a Veterans’<br />

Administration (VA) cohort, MS was associated with a shorter duration <strong>of</strong><br />

PC control with ADT. We report the impact <strong>of</strong> MS on overall survival (OS)<br />

and PC specific death for a cohort <strong>of</strong> patients with biochemical relapse.<br />

Methods: 273 pts (64 VA pts and 209 pts from Health Pr<strong>of</strong>essionals Follow<br />

up Study) treated with ADT for biochemical recurrence post radiation or<br />

prostatectomy for PC were included. The modified Adult Treatment Panel<br />

III criteria for MS was used to identify patients with MS status prior to the<br />

commencement <strong>of</strong> ADT. Cox models tested for association <strong>of</strong> MS status<br />

with OS or time to PC specific death. With 42% overall death rate, 31% MS<br />

prevalence, there was 90% power to detect HR� 1.81 (type I error rate<br />

�0.05). Results: 31% pts (84/273) had MS and 15% pts (40/273) died <strong>of</strong><br />

PC. Median follow-up was 9.5 years. The median OS with and without MS<br />

was 7.4 and 11.2 years respectively. Patients with hypertension, being<br />

African <strong>American</strong>, having diabetes and age were associated with increased<br />

risk <strong>of</strong> death from any causes, while hypertension and being African<br />

<strong>American</strong> were also associated with increased risk <strong>of</strong> PC specific death. A<br />

multivariate Cox regression model adjusted for age at diagnosis, race,<br />

definitive local therapy (RT vs. RP), PSA at diagnosis and Gleason score<br />

revealed MS was associated with a significantly increased risk <strong>of</strong> death from<br />

any cause and PC specific death (Table). Conclusions: In men receiving ADT<br />

for biochemically recurrent androgen dependent PC, the presence <strong>of</strong> MS at<br />

the commencement <strong>of</strong> ADT is associated with an increased risk <strong>of</strong> death<br />

from any cause as well as prostate cancer specifically.<br />

Overall (N�273)<br />

Comparison Median (years) Event Adjusted HR<br />

Endpoint (MS status) OS, (95%CI) (patient) (95% CI) P value*<br />

OS No (ref) 11.17 74 (189) 1<br />

(10-13.9)<br />

Yes 7.42 (5.33-12.1) 40 (84) 2.12<br />

(1.42, 3.16) �0.001<br />

PCa death No (ref) 25 (189) 1<br />

Yes 15 (84) 2.03 0.036<br />

(1.05, 3.94)<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


4556 Poster Discussion Session (Board #10), Mon, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Cytoreduction and androgen signaling modulation by abiraterone acetate<br />

(AA) plus leuprolide acetate (LHRHa) versus LHRHa in localized high-risk<br />

prostate cancer (PCa): Preliminary results <strong>of</strong> a randomized preoperative<br />

study. Presenting Author: Eleni Efstathiou, University <strong>of</strong> Athens, Athens,<br />

Greece; University <strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: Endocrine to “intracrine” androgen signaling transition, a<br />

milestone in the lethal progression <strong>of</strong> PCa, has not been characterized in<br />

localized high risk disease. Signaling heterogeneity under the selective<br />

pressure <strong>of</strong> castration may account for response differences. Methods: A<br />

single institution preoperative study <strong>of</strong> 12 weeks AA 1g/prednisone 5 mg �<br />

LHRHa compared to LHRHa (randomized 2:1) was conducted in patients<br />

(pts) with high risk PCa (clinical stage �T1c and biopsy Gleason score �8,<br />

or �T2b, Gleason � 7 and PSA � 10ng/ml). Primary aim: Assess<br />

difference in down staging (� ypT2) and safety. Secondary aims: Assess<br />

difference in androgen biosynthesis, androgen signaling (AR, AR variants,<br />

NKX 3.1, ERG, PSA) proliferation apoptosis and candidate treatment<br />

resistance pathways. Results: We report on 37 (50 enrolled) pts who had<br />

prostatectomy. AA�LHRHa was given to 25pts, LHRHa to 12. Median age<br />

is 61 ys (46-74). Preoperative PSA was �0.1ng/ml in 17/25 (68%)<br />

AA�LHRHa vs 0/12 LHRHa (p 0.0001). ypT2N0 occurred in 15/25 (60%)<br />

AA�LHRHa treated pts vs 4/12 (33%) LHRHa (p 0.17). Near complete<br />

cytoreduction (�6mm scattered cells) occurred in 6/25 (24%) AA�LHRHa<br />

vs 1/12 (8%) LHRHa. Lymph node infiltration in 7/25 (28%) AA�LHRHa<br />

vs 6/12 (50%) LHRHa. Margin positivity in 2/25 (8%) AA�LHRHa vs 4/12<br />

(33%) LHRHa (p 0.07). Grade 3 AA related AEs: elevated AST/ALT 4<br />

(discontinued AA), hypertension 3. One AA�LHRHa pt had postop pulmonary<br />

embolism. Androgen signaling and proliferation suppression is more<br />

pr<strong>of</strong>ound in AA�LHRHa treated remaining tumor cells (Table). Conclusions:<br />

Addition <strong>of</strong> AA to LHRHa results in greater cytoreduction, suppression <strong>of</strong><br />

PSA and androgen signaling compared to LHRHa in high risk PCa. Findings<br />

support the hypothesis that intracrine androgen signaling is a therapy target<br />

in patients with untreated PCa and form the foundation for a marker driven<br />

treatment strategy.<br />

Mean tumor expression (involvement)% by IHC (standard deviation).<br />

Marker AA�LHRHa LHRHa P value Wilcoxon<br />

AR 27 (26) 69 (22.3) 0.0001<br />

Nkx3.1 50 (29.8) 83 (14.3) 0.002<br />

CYP17 64 (22) 75 (14.4) 0.13<br />

Ki67 3.4 (5) 8.6 (5.7) 0.003<br />

4558 Poster Discussion Session (Board #12), Mon, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Exploratory analysis <strong>of</strong> survival benefit and prior docetaxel (D) treatment in<br />

COU-AA-301, a phase III study <strong>of</strong> abiraterone acetate (AA) plus prednisone<br />

(P) in metastatic castration-resistant prostate cancer (mCRPC). Presenting<br />

Author: Oscar B. Goodman, National Cancer Institute, Las Vegas, NV<br />

Background: AA, a selective androgen biosynthesis inhibitor, blocks the<br />

action <strong>of</strong> CYP17, thereby inhibiting adrenal and intratumoral androgen<br />

production. AA has demonstrated improved overall survival (OS) by 4.6<br />

months (mos) vs placebo (HR�0.74) in patients (pts) previously treated<br />

with D. Methods: COU-AA-301 is a randomized double blind study <strong>of</strong> AA (1<br />

g) � P (5 mg po BID) vs placebo � P administered to mCRPC pts post-D<br />

with a primary endpoint <strong>of</strong> OS. To further evaluate primary survival result<br />

robustness, we performed post hoc exploratory analyses to assess whether<br />

the timing <strong>of</strong> first and last dose <strong>of</strong> D and reason for D discontinuation<br />

impacted OS. Results: At randomization, treatment arms were balanced<br />

with respect to baseline characteristics, prior D use, and reasons for<br />

discontinuation. In both arms, almost half (45%) discontinued D due to<br />

progressive disease (PD); remainder discontinued D after completing all<br />

planned cycles (37%), due to toxicity (12%), or for other reasons (5%) per<br />

investigator. Median OS from first and last dose <strong>of</strong> D were longer with AA vs<br />

placebo (Table). Median OS was longer with AA vs placebo in pts who<br />

discontinued D for PD, or for all other reasons. Conclusions: These<br />

exploratory analyses suggest that the OS benefit <strong>of</strong> AA in mCRPC was<br />

maintained when calculated from first or last dose <strong>of</strong> prior D, and whether<br />

or not pts discontinued D for PD. Pts in AA arm <strong>of</strong> this study had a<br />

prolonged median OS <strong>of</strong> � 32 mos from time <strong>of</strong> initial D therapy. Congruity<br />

among these analyses and lack <strong>of</strong> dependence on D timing demonstrate<br />

robustness <strong>of</strong> the primary survival result.<br />

Median OS, mos (95% CI)<br />

Category a<br />

AA<br />

(N�797)<br />

Placebo<br />

(N�398)<br />

Time from D b<br />

From first dose D 32.6 (30.7, 35.0) 27.6 (25.9, 30.3)<br />

HR � 0.75 (0.65, 0.88); p�0.0002<br />

From last dose D 23.2 (22.4, 24.5) 19.4 (17.5, 20.8)<br />

HR � 0.74 (0.64, 0.86); p �0.0001<br />

Reason for D discontinuation c<br />

PD 14.2 (12.0, 15.8) 10.5 (9.3, 11.8)<br />

HR � 0.77 (0.62, 0.97); p�0.0222<br />

All other reasons 17.0 (15.6, 18.2) 12.6 (10.4, 14.9)<br />

HR � 0.73 (0.59, 0.89); p�0.0025<br />

a<br />

Investigator reported (limited source verification);<br />

b<br />

calculated from first or last dose <strong>of</strong> D;<br />

c<br />

calculated<br />

from randomization.<br />

Genitourinary Cancer<br />

4557 Poster Discussion Session (Board #11), Mon, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Time to testosterone (T) and PSA rises during first “<strong>of</strong>f treatment” interval<br />

(1OFF-2ON) <strong>of</strong> intermittent androgen deprivation (IAD) as prognostic for<br />

time to castration resistance (CRPC) and prostate cancer mortality (PCM) in<br />

men with biochemical relapse (BR). Presenting Author: Evan Y. Yu, Fred<br />

Hutchinson Cancer Research Center, Seattle, WA<br />

Background: A recent phase III trial <strong>of</strong> intermittent vs. continuous AD<br />

supports IAD as standard <strong>of</strong> care for men treated with AD for BR. To identify<br />

potential prognostic factors during 1OFF, times to T and PSA rises from our<br />

prospective trial <strong>of</strong> IAD in men with BR were analyzed in relation to times to<br />

CRPC and PCM. Methods: 72 men with BR after definitive local therapy<br />

were treated with IAD, each cycle consisting <strong>of</strong> 9 months <strong>of</strong> leuprolide and<br />

flutamide followed by a variable “<strong>of</strong>f treatment” interval. T and PSA were<br />

followed monthly; AD was resumed when PSA reached a pre-specified<br />

value. Cycles repeated until CRPC, defined as �2 PSA rises with T�50<br />

ng/dL. Markers <strong>of</strong> interest from 1OFF-2ON were time to first T�50, time<br />

from first T�50 to first PSA rise �0.1 ng/mL, time to first PSA rise �0.1,<br />

and PSA doubling time (PSAdt), calculated using the first 3 PSA measurements<br />

starting from first PSA �0.1. The associations <strong>of</strong> these markers with<br />

CRPC and PCM were evaluated using Cox proportional hazards models (or<br />

logistic regression if the Cox proportional hazards assumption was not met),<br />

controlling for age at study entry and Gleason score categorized to �7 or<br />

�7. Results: A 30-day increase in time to first T�50 was significantly<br />

associated with a 75% increase in the risk <strong>of</strong> PCM. A 30-day increase in<br />

time to PSA rise from 1OFF or after T�50 was significantly associated with<br />

a 23% or 72% reduction in the risk <strong>of</strong> CRPC, respectively. While neither <strong>of</strong><br />

the associations <strong>of</strong> PSAdt with CRPC or PCM were significant, they were <strong>of</strong><br />

moderate size: PSAdt displayed a moderate reduction in risk <strong>of</strong> CRPC by<br />

35% and PCM by 38%. Conclusions: During the first “<strong>of</strong>f treatment”<br />

interval <strong>of</strong> IAD, the time to first T�50 is prognostic for PCM. Time to first<br />

PSA rise after 1OFF and after first T�50 are both prognostic for CRPC.<br />

Time to CRPC Time to PCM<br />

Marker<br />

N HR P value 95% CI N HR P value 95% CI<br />

1OFF to T>50<br />

T>50 to PSA>0.1<br />

46<br />

45<br />

1.1<br />

0.28<br />

0.67<br />

0.05<br />

(0.71, 1.7)<br />

(0.08, 0.99)<br />

49<br />

48<br />

1.75<br />

0.28<br />

0.01<br />

0.19<br />

(1.14, 2.68)<br />

(0.03, 1.63) ��<br />

1OFF to PSA>0.1 57 0.77 0.01 (0.63, 0.93) 60 0.85 0.24 (0.65, 1.11)<br />

PSAdt 49 0.65 0.08 (0.4, 1.06) 52 0.62 0.29 (0.26, 1.5)<br />

�� Odds ratio.<br />

291s<br />

4559 Poster Discussion Session (Board #13), Mon, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

The effect <strong>of</strong> PSA frequency and duration on PSA doubling time (PSADT)<br />

calculations in men with biochemically recurrent prostate cancer (BRPC)<br />

after definitive local therapy. Presenting Author: Channing Judith Paller,<br />

The Johns Hopkins Hospital, Baltimore, MD<br />

Background: The prognostic nature <strong>of</strong> PSADT in men with BRPC makes it an<br />

attractive intermediate endpoint for assessing novel therapies in these<br />

patients. Although a number <strong>of</strong> investigational agents appear to favorably<br />

modulate PSADT, slowing in PSADT has also been observed in placebotreated<br />

men. We aimed to determine whether PSADT calculations could be<br />

influenced by the frequency and duration <strong>of</strong> PSA measurements. Methods:<br />

We performed a retrospective analysis <strong>of</strong> men with BRPC who chose to<br />

defer hormonal therapy. Eligible men were those with PSA values �0.2<br />

ng/mL and at least 6 values taken on average 3 mo apart, and whose local<br />

prostate cancer therapy was completed �1 year prior. To examine the<br />

influence <strong>of</strong> PSA frequency and duration on PSADT, we calculated median<br />

PSADT using different subsets <strong>of</strong> available PSA values (e.g. each vs every<br />

other; and first 3 vs. remaining PSA values). Results: After a median<br />

follow-up <strong>of</strong> 58 mo (range, 6-185 mo), 213 men with BRPC had �6 PSA<br />

values and 127 men had �9 PSA values for analysis. Men (77% white,<br />

23% black/other) had a median age <strong>of</strong> 61 y with Gleason score distribution<br />

as follows (�6: 30%; 7: 40%; �8: 18%; NOS: 12%). For men with �6<br />

data points: PSADT calculated using earlier values ranged from 13.2 to<br />

16.6 mo, compared to 16.6 to 17.8 mo, respectively, for the remaining<br />

values (within-patient change range: 0.6-1.2 mo). For men with �9 data<br />

points: PSADT calculated using earlier values ranged from 15.3 to 19.9 mo<br />

compared to 22.1 to 22.3 mo, respectively, for the remaining values<br />

(within-patient change range: 3.5 to 4.5 mo). When we examined the<br />

frequency <strong>of</strong> PSADT by using every other value, we found little difference<br />

(22.3 vs 22.1 mo). Conclusions: These data show that PSADT appears to<br />

increase even in the absence <strong>of</strong> therapy, and may be influenced by duration<br />

<strong>of</strong> PSA follow up. This finding explains PSADT slowing on placebo arms and<br />

calls into question the utility <strong>of</strong> PSADT as a surrogate endpoint. Placebocontrolled<br />

trials and the use <strong>of</strong> standard clinical endpoints are recommended<br />

to screen novel agents in men with BRPC to mitigate bias because<br />

<strong>of</strong> natural PSADT variability.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


292s Genitourinary Cancer<br />

4560 Poster Discussion Session (Board #14), Mon, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Development <strong>of</strong> a needle biopsy-based genomic test to improve discrimination<br />

<strong>of</strong> clinically aggressive from indolent prostate cancer. Presenting<br />

Author: Eric A. Klein, Glickman Urological and Kidney Institute, Cleveland<br />

Clinic, Cleveland, OH<br />

Background: Standardized, quantitative tools are needed to improve discrimination<br />

<strong>of</strong> clinically aggressive from indolent prostate cancer at diagnosis.<br />

We previously identified multiple genes and biological pathways in radical<br />

prostatectomy (RP) specimens associated with clinical recurrence and<br />

adverse pathology. We evaluated whether measurement <strong>of</strong> these genes in<br />

prostate needle biopsy (Bx) specimens predicts adverse pathology - high<br />

grade (GS � 4�3) and/or non-organ-confined disease (pT3) - in a separate<br />

study <strong>of</strong> AUA low-intermed risk pts treated w/ RP. Methods: All AUA<br />

low-intermed risk (cT1-T2a, GS 3�3/3�4) pts treated w/ RP �6 mos from<br />

Bx at Cleveland Clinic (1999-2007) w/ available Bx tissue and �3 pos<br />

cores were evaluated. Expression <strong>of</strong> 81 prognostic genes identified in<br />

previous RP studies and 5 reference genes was quantitated by RT-PCR in<br />

60 �m manually microdissected FPE prostate needle Bx tissue. Association<br />

<strong>of</strong> gene expression w/ adverse pathology was analyzed by logistic<br />

regression controlling false discovery rate (FDR) using Storey’s method.<br />

Results: RT-PCR was successful for 167 <strong>of</strong> 171 (98%) pts (92 AUA low, 75<br />

intermed). At RP, 58 pts (35%) had high-grade and/or non-organ confined<br />

disease. 58 <strong>of</strong> 81(72%) genes predicted high grade and/or non-organconfined<br />

disease (FDR�10%), including all stromal response and androgen<br />

genes and the majority <strong>of</strong> cellular organization genes (82%).<br />

Proportionately fewer proliferation (40%), stress response (29%), and<br />

basal epithelial (25%) genes were associated w/ adverse path. Androgen<br />

and cellular organization genes consistently predicted lower risk; stromal<br />

response and proliferation genes predicted higher risk; (majority <strong>of</strong> std odds<br />

ratios �1.5, comparable to the prognostic strength <strong>of</strong> ER in breast cancer).<br />

Conclusions: Genes and biological pathways associated w/ clinically aggressive<br />

prostate cancer in RP specimens can be reliably measured by RT-PCR<br />

from fixed prostate needle biopsies and predict high-grade, non-organconfined<br />

disease. These results support the potential value <strong>of</strong> a Bx-based<br />

assay to inform selection <strong>of</strong> immediate treatment or active surveillance for<br />

pts diagnosed with prostate cancer on needle Bx.<br />

4562 Poster Discussion Session (Board #16), Mon, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Predictive biomarkers in circulating tumor cells (CTC) from patients with<br />

castration-resistant prostate cancer (CRPC) through genomic analysis.<br />

Presenting Author: Daniel Costin Danila, Sidney Kimmel Center for<br />

Prostate and Urologic Cancers, Memorial Sloan-Kettering Cancer Center,<br />

New York, NY<br />

Background: Mutations in the ligand binding domain <strong>of</strong> androgen receptor<br />

(AR) in prostate cancer cells may alter their sensitivity to treatment with<br />

specific antiandrogens. To predict for tumor sensitivity to treatment with<br />

novel AR targeted therapies, we explored the frequency <strong>of</strong> mutation<br />

detection and copy number alteration in CTC isolated from patients with<br />

CRPC enrolled on trials with these targeted therapies. Methods: We used<br />

fluorescence-activated cell sorting (FACS) methodology to enrich EpCAM� ,<br />

CD45- , DAPI- cells. For mutation detection and genomic copy number<br />

alteration in CTC in low number <strong>of</strong> cancer cells found in clinical samples,<br />

we optimized next-gen deep sequencing by Illumina. Results: In patients<br />

with progressive CRPC , �10 or �50 EpCAM� events (EPE) were isolated<br />

by FACS in 88% or 58% <strong>of</strong> patients, in whom 32% and 10% had<br />

unfavorable (�5 cells/7.5 ml) CTC counts using CellSearch. EPE, expressing<br />

prostate-specific mRNAs, provide sufficient high quality DNA for<br />

genomic sequencing and copy number analysis. Adequate coverage was<br />

obtained from as few 50 EPE, with a recovery rate <strong>of</strong> 89% from FACS sorted<br />

samples. The detection threshold <strong>of</strong> a mutation was established at 1:4<br />

alleles. To further expand genomic pr<strong>of</strong>iling in CTC, we optimized Nimblegen<br />

exome mutation detection by deep sequencing on HiSeq PE101. Our<br />

initial analysis established the polymorphism frequency detection thresholds<br />

in heterogeneous cell populations, and confirmed the sequencing<br />

coverage. Somatic missense mutations in AR, APC and TP53 found in CTC<br />

but not in paired WBC were confirmed by Sanger sequencing. In parallel,<br />

copy number alterations in CTC are studied. Conclusions: Somatic mutations<br />

detected in CTC isolated from patients with CRPC can serve as<br />

predictive markers <strong>of</strong> tumor sensitivity to targeted therapies. We established<br />

standard operating procedures for specimen processing, and confirmed<br />

the sequencing coverage and polymorphism detection thresholds in<br />

heterogeneous cell population. Currently we are proceeding to clinical<br />

samples to study the associations between specific molecular alterations in<br />

CTC as predictive markers <strong>of</strong> sensitivity and clinical outcomes.<br />

4561 Poster Discussion Session (Board #15), Mon, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Gene expression biomarkers to predict overall survival <strong>of</strong> prostate cancer<br />

patients. Presenting Author: Chunde Li, Department <strong>of</strong> <strong>Clinical</strong> Oncology,<br />

Karolinska University Hospital, Stockholm, Sweden<br />

Background: Current clinical parameters are not accurate in the prediction<br />

<strong>of</strong> overall survival <strong>of</strong> prostate cancer patients. Methods: Driven by cancer<br />

stem cell hypothesis and by using a simple bioinformatics analysis, we<br />

identified 641 genes with either consistently high or low level <strong>of</strong> expression<br />

in human embryonic stem cells. We showed that these 641 genes had<br />

strong power to classify cancers into different biological subtypes and<br />

named them as embryonic stem cell gene predictors (ESCGPs). Nineteen<br />

selected ESCGPs and five control genes were analyzed by multiplex<br />

quantitative PCR in prostate fine needle aspiration (FNA) samples taken at<br />

diagnosis. The cohort included 189 patients diagnosed during 1986-<br />

2001. Of these patients, 97.9% had overall and cancer specific survival<br />

data and 77.9% were treated by medical or surgical castration as the<br />

primary treatment. The cohort was divided into a discovery and two<br />

validation subsets. The univariate and multivariate Cox proportional hazards<br />

and Kaplan-Meier plots were used in the survival analysis. A published<br />

dataset was used for an external validation. Results: Ten gene markers F3,<br />

WNT5B, VGLL3, CTGF, IGFBP3, c-MAF-a, c-MAF-b, AMACR, MUC1 and<br />

EZH2, showed a significant correlation to overall or cancer specific survival.<br />

Four <strong>of</strong> these genes, F3, IGFBP3, CTGF and AMACR, were independent <strong>of</strong><br />

all clinical parameters. An expression signature <strong>of</strong> F3, VGLL3 and IGFBP3<br />

characterized patients into three subtypes. The median overall survival was<br />

2.60 years in the high risk, 3.85 years in the intermediate risk and 7.98<br />

years in the low risk subtype, corresponding to a HR <strong>of</strong> 5.86 (95% CI<br />

2.91-11.78, p�0.001) for the high risk and 3.45 (95% CI 1.79-6.66,<br />

p�0.001) for the intermediate risk over the low risk subtype. Two gene<br />

markers, F3 and EZH2, were further verified by the external validation.<br />

Conclusions: The new ESCGP gene markers and the expression signatures<br />

can be used to predict the overall and cancer specific survival <strong>of</strong> prostate<br />

cancer patients.<br />

4563 Poster Discussion Session (Board #17), Mon, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Generation <strong>of</strong> a prognostic cancer stem-like gene expression signature in<br />

men undergoing radical prostatectomy for localized prostate cancer.<br />

Presenting Author: Adrian Stuart Fairey, University <strong>of</strong> Southern California,<br />

Los Angeles, CA<br />

Background: Novel biomarkers are needed to predict recurrence after<br />

radical prostatectomy for localized prostate cancer. Recent data suggest<br />

that cancer stem-like cells (CSC) are present in prostate tumors, and the<br />

CSC phenotype has been associated with an aggressive cancer phenotype.<br />

We investigated whether tumor mRNA levels <strong>of</strong> genes typically expressed<br />

by CSC are associated with disease recurrence. Methods: <strong>Clinical</strong>ly annotated<br />

prostatectomy specimens (FFPE) were used for this nested casecontrol<br />

study. Samples represented men who underwent radical<br />

prostatectomy and either experienced a recurrence (PSA or clinical;<br />

n�152) or no recurrence (controls; n�124) with minimum 5-year followup.<br />

Men were excluded if they received neoadjuvant hormone therapy.<br />

Cases and controls were frequency matched based on D’Amico risk group.<br />

Laser capture microdissection with mRNA extraction and quantitative<br />

RT-PCR were used to quantify the expression <strong>of</strong> 12 candidate CSCassociated<br />

genes (ALDH1A1, Axin2, Bmi1, CD133, CD44a, CTNNB1/�catenin,<br />

ITGA2/integrin �2, NANOG, Nkx3-1, Notch1, OCT 4, TACSTD2/<br />

Trop2). Univariable and multivariable analyses were conducted to measure<br />

associations with recurrence. Results: mRNA data were evaluable for 241 <strong>of</strong><br />

276 patients. Univariate Wilcoxon analysis showed that 4 genes (Axin2,<br />

p�0.001; CD44a, p�0.036; OCT 4, p�0.017; TACSTD2, p�0.008) were<br />

expressed at low levels in tumors <strong>of</strong> patients whose disease recurred.<br />

Recursive partitioning analysis identified 3 genes significantly predictive <strong>of</strong><br />

disease recurrence (Axin2, NANOG, CTNNB1), with cutpoints yielding<br />

odds ratios (OR) <strong>of</strong> 8.5 (Axin2low /NANOGhigh ;n�94, 95%CI 3.7-19.2) and<br />

5.1 (Axin2low /NANOGvery high /CTNNB1low ; n�26, 95%CI 1.7-14.8).<br />

Conclusions: We identified a novel CSC-associated 3 gene expression<br />

signature with the potential to predict recurrence after radical prostatectomy.<br />

Axin2 is known to interact with CTNNB1/�-catenin in the Wnt<br />

signaling pathway, which in turn has been shown to regulate expression <strong>of</strong><br />

NANOG, a key protein that mediates pluripotency. In vitro experiments and<br />

an independent cohort validation study are planned based on these novel<br />

findings.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


4564 Poster Discussion Session (Board #18), Mon, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Pretreatment (pre-tx) neutrophil to lymphocyte ratio (NLR) in metastatic<br />

castration-resistant prostate cancer (mCRPC) patients (pts) treated with<br />

ketoconazole (keto): Association with outcome and predictive model.<br />

Presenting Author: Avishay Sella, Assaf Har<strong>of</strong>eh Medical Center, Zerifin,<br />

Israel<br />

Background: The CYP17 inhibitor keto is active in mCRPC. The NLR, an<br />

index <strong>of</strong> systemic inflammation, is associated with prognosis in several<br />

types <strong>of</strong> cancer. We assessed the association between pre-tx NLR and<br />

outcome <strong>of</strong> mCRPC pts treated with keto. Methods: We performed an<br />

international multicenter retrospective study <strong>of</strong> pts with mCRPC, who were<br />

treated with keto. We analyzed the pre-tx NLR and previously described<br />

factors associated with keto tx outcome as prior response to hormonal tx,<br />

pre-tx PSADT, and extent <strong>of</strong> metastatic disease (limited vs extensive).<br />

Progression free survival (PFS) was determined by the Kaplan-Meier<br />

method. Multivariate analyses using Cox regression model were performed<br />

to determine their independent effect, and to form a predictive model. A<br />

survival tree analysis was used to find the best NLR cut-<strong>of</strong>f value. Results:<br />

From 1999-2011, 156 mCRPR pts (median age 69) were treated with<br />

keto. 78/156 (50%) had � 50% PSA decline. Overall median PFS was 8<br />

months (mos) (range 1-144). Excluded from the analysis were 23 pts<br />

without available data on pre-tx NLR, and those with recent (�1 mos)<br />

health event or tx (surgery, steroids, radiation) associated with a change <strong>of</strong><br />

blood counts. 133 pts were included in the analysis. 62 (47%) had an<br />

elevated pre-tx NLR �3. Risk factors associated with PFS (table) were<br />

pre-tx NLR �3, prior response to GnRH-a �24 mos and to antiandrogen<br />

(AA) �6 mos, and pre-tx PSADT �3 mos. The number <strong>of</strong> risk factors was<br />

used to categorize patients into three risk groups (table): favorable (0-1<br />

factors), intermediate (2 factors), and poor (3-4 factors). Conclusions: In<br />

mCRPC pts treated with keto, pre-tx NLR, prior response to hormonal tx,<br />

and pre-tx PSADT are associated with PFS, and may be used to categorize<br />

pts into risk groups.<br />

Factor PFS (mos) (HR, p value)<br />

NLR < 3vs>3 14 vs 3, (0.353, �0.0001)<br />

Response to prior GnRH-a<br />

12 vs 5, (0.513, 0.035)<br />

> 24 vs < 24 mos<br />

Response to prior AA<br />

18 vs 3, (0.445, 0.003)<br />

>6vs3vs


294s Genitourinary Cancer<br />

4569 Poster Discussion Session (Board #23), Mon, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Dual antiangiogenic therapy using lenalidomide and bevacizumab with<br />

docetaxel and prednisone in patients with metastatic castration-resistant<br />

prostate cancer (mCRPC). Presenting Author: Bamidele Adesunloye, Medical<br />

Oncology Branch, National Cancer Institute, National Institutes <strong>of</strong><br />

Health, Bethesda, MD<br />

Background: Previously, we had shown the potent anti�tumor activity <strong>of</strong><br />

dual anti-angiogenic therapy by combining bevacizumab (B) and thalidomide<br />

(T) with docetaxel (D) and prednisone (P) in mCRPC (Ning JCO<br />

2010). We hypothesized that combining lenalidomide (L), an analogue <strong>of</strong><br />

T, with B, D, and P would have a more favorable efficacy/toxicity pr<strong>of</strong>ile.<br />

Methods: All patients (pts) had chemotherapy�naïve mCRPC. Among the<br />

first 52 pts, 3 received L 15 mg daily, 3 had 20 mg daily, and the rest had<br />

25 mg daily for 14 days <strong>of</strong> every 21�day cycle (C). The protocol was<br />

recently amended to enroll 11 more pts at L 15 mg; 2 pts have now been<br />

enrolled in this expansion cohort. All pts received D 75 mg/m2 and B 15<br />

mg/kg on day 1 with P 10 mg and enoxaparin daily throughout each C.<br />

Pegfilgrastim was given on day 2. PSA each C with imaging after C2 and<br />

after every 3C. Dental exams with mandible CT scan at baseline, after C5,<br />

and every 6C. Results: 54 <strong>of</strong> 62 pts have been enrolled. Median age 65.5<br />

(51�82), Gleason score 8 (5�10), on�study PSA 85.2 ng/ml<br />

(0.15�3520), and pre�study PSA doubling time 1.49 months<br />

(0.52�6.73). Median number <strong>of</strong> Cs was 16 (3�38). PFS was 22 months<br />

and probability <strong>of</strong> survival at 12 months was 90%. Forty-six (85.2%) and<br />

42 (77.8%) pts had PSA declines <strong>of</strong> �50% and �75%, respectively. Of 30<br />

pts with measurable disease there were 1 CR and 25 PR (86.7% overall<br />

RR). 17/54 pts were <strong>of</strong>f study for radiographic disease progression and<br />

8/54 for other reasons. Grade �2 toxicities included neutropenia (34/54),<br />

anemia (23/54), thrombocytopenia (7/54), hypertension (12/54), perianal<br />

fistula (3/54), rectal fissure (1/54), myocardial infarction (1/54), and<br />

osteonecrosis <strong>of</strong> the jaw (ONJ) (12/54, 22.0%). At the time <strong>of</strong> diagnosis <strong>of</strong><br />

ONJ, 7/12pts were on bisphosphonates (BP), 2/12 had used BP previously,<br />

and 3/12 never used BP. The incidence <strong>of</strong> ONJ was comparable to 18.3%<br />

reported by Ning et al. A recent study <strong>of</strong> carboplatin plus weekly docetaxel<br />

reported an incidence <strong>of</strong> 29.3%. Conclusions: Dual anti-angiogenic therapy<br />

with, B and L, plus D and P was associated with high PSA (85.2%) and<br />

tumor (86.7%) responses in mCRPC, with manageable toxicities. The<br />

incidence <strong>of</strong> ONJ is comparable to other studies.<br />

4571 Poster Discussion Session (Board #25), Mon, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Preliminary quality-<strong>of</strong>-life outcomes for SWOG-9346: Intermittent androgen<br />

deprivation in patients with hormone-sensitive metastatic prostate<br />

cancer (HSM1PC)—Phase III. Presenting Author: Carol Moinpour, Fred<br />

Hutchinson Cancer Research Center, Seattle, WA<br />

Background: The relative quality <strong>of</strong> life (QOL) for patients with newly<br />

diagnosed, metastatic prostate cancer, treated with intermittent androgen<br />

deprivation (IAD) has been assumed and hypothesized, yet never compared<br />

in a well-powered randomized trial (RT) to continuous androgen deprivation<br />

(CAD). SWOG-9346 provided such a RT in which to test QOL differences<br />

between CAD and IAD in men with metastatic prostate cancer. Methods:<br />

Patients were randomized to CAD or IAD. Patients completed the SWOG<br />

QOL Questionnaire (SF-20/SF-36, Symptom Distress Scale, treatmentspecific<br />

symptoms, global QOL) at randomization and months (mo) 3, 9,<br />

and 15 post-randomization. Five QOL change scores at one time point (mo<br />

3) were designated as primary for the QOL endpoint and are reported in this<br />

abstract: impotence, libido, energy/vitality (E/V), physical function (PF),<br />

and emotional function (EF). Significance level was adjusted for 5<br />

comparisons (used p�0.01). Results: 615 patients in the CAD arm and<br />

633 in the IAD arm completed the QOL questionnaire at baseline. Change<br />

between baseline and 3 months differed for the two arms with CAD<br />

reporting statistically significantly more impotence and less libido than<br />

IAD. EF was also slightly better for the IAD arm. Conclusions: These results<br />

indicate better sexual function in men receiving IAD versus CAD through<br />

post-randomization month 3. Additional benefits for IAD may include<br />

better PF, E/V and EF. Ongoing analyses will address the role <strong>of</strong> missing<br />

data, additional follow-up assessments, and resumption <strong>of</strong> therapy in the<br />

IAD arm.<br />

Baseline treatment arm<br />

Impotence %<br />

(n)<br />

Libido %<br />

(n)<br />

EF^ score<br />

(n)<br />

PF^ score<br />

(n)<br />

E/V^ score<br />

(n)<br />

Descriptive results<br />

3-mo<br />

CAD IAD CAD IAD T-test p values*<br />

.85<br />

(546)<br />

.03<br />

(560)<br />

80.0<br />

(568)<br />

70.2<br />

(569)<br />

59.8<br />

(557)<br />

.82<br />

(585)<br />

.04<br />

(580)<br />

77.9<br />

(595)<br />

70.7<br />

(591)<br />

59.7<br />

(586)<br />

.87<br />

(487)<br />

.04<br />

(481)<br />

79.0<br />

(491)<br />

69.3<br />

489)<br />

58.9<br />

(478)<br />

.73<br />

(494)<br />

.11<br />

(486)<br />

79.6<br />

(500)<br />

71.3<br />

(500)<br />

60.0<br />

(489)<br />

*T-test <strong>of</strong> arm difference in baseline to 3-month change for patients with both assessments; ^0-100 scale.<br />

� 0.01<br />

� 0.01<br />

� 0.01<br />

0.08<br />

0.23<br />

4570 Poster Discussion Session (Board #24), Mon, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

A multinational phase III adjuvant study <strong>of</strong> immediate (I) versus deferred<br />

(D) chemotherapy (C)/hormone therapy (HT) after radical prostatectomy<br />

(RP): TAX-3501. Presenting Author: Mario A. Eisenberger, Sidney Kimmel<br />

Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD<br />

Background: TAX 3501 was designed to test I or D C/HT after RP. No<br />

adequately designed prospectively randomized surgical systemic adjuvant<br />

studies have ever been successfully completed. Methods: TAX-3501 was a<br />

randomized phase III study. Eligibility included post-operative predicted<br />

probability <strong>of</strong> 5-year freedom from progression <strong>of</strong> �60% (Kattan et al)<br />

after central pathology review, no prior C/HT, undetectable PSA, M0,<br />

normal hematology/chemistries, signed consent. Pts were randomized<br />

within 120 days after RP to I or D (at 1st progression) Rx with HT (leuprolide<br />

acetate 22.5mg S.C. q.3 months x6)�/- C (docetaxel 75mg/m2 q.3wks x<br />

6). F/U included a physical exam, PSA, CBC/chemical pr<strong>of</strong>ile, serum T,<br />

scans yearly or at progression (PSA� 0.4ng/ml, clinical or radiographic<br />

and death). Main objectives comparing PFS and 5-year progression-free<br />

rates after systemic treatment (2 � 2 factorial design, 4 arms). 1,696<br />

subjects would provide 90% power to detect the targeted treatment effect<br />

at a 5% 2-sided type I error level. Results: From 12/2005-9/2007399<br />

pts were registered, 228 randomized after central path review ICHT�55,<br />

IHT�55, DCHT�56, DHT�62, median age 62 (41-76), pre-op PSA<br />

(ng/ml) 9.38 (2.2 - 90.0), Gleason score- 8,T3a (19.6%),<br />

R�(65%),S.V.�(50%),N�(19.7%%), all had undetectable post op PSA.<br />

Predicted probability <strong>of</strong> no progression was 21% on 228pts. Study was<br />

terminated by sponsor (9/2007) due to poor accrual F/U continued from<br />

2007-2010. Small sample size precludes reliable analyzes 37/118 (31.3%)<br />

on the D arms received CHT/HT (all PSA progressions); ICHT 10/55, IHT<br />

14/55, DCHT 9/56, DHT 8/62 met progression endpoint after I/D Rx (1/31<br />

bone). AEs were characteristic and reversible (no grade 5). Leading causes<br />

<strong>of</strong> accrual impediment were inadequate site selection, no consensus<br />

regarding patient and treatment selection for this pt population, physician<br />

and patient bias re: treatment and evolving changes in the adjuvant<br />

treatment landscape during follow-up time (adjuvant RT). Conclusions: The<br />

role <strong>of</strong> adjuvant systemic treatment after RP remains undefined. <strong>Clinical</strong><br />

trials in this pt population are challenging. Supported by San<strong>of</strong>i<br />

NCT000283062.<br />

4572 General Poster Session (Board #1A), Sun, 8:00 AM-12:00 PM<br />

Intravesical sequential BCG and electromotive mitomycin versus BCG<br />

alone in high risk non-muscle invasive bladder cancer. Presenting Author:<br />

Savino Mauro Di Stasi, Department <strong>of</strong> Surgery/Urology, Tor Vergata<br />

University, Rome, Italy<br />

Background: In 2006, we reported that intravesical sequential bacillus<br />

Calmette-Guérin (BCG) and electromotive mitomycin in high risk nonmuscle<br />

invasive bladder cancer leads to higher disease-free interval, lower<br />

recurrence and progression, and to improved overall survival and diseasespecific<br />

survival compared with BCG alone. After an additional 6 years <strong>of</strong><br />

follow-up, we now report estimated 16-year results. Methods: From 1994<br />

through 2002, we randomly assigned 212 patients with high risk non-<br />

.muscle invasive bladder cancer to 81 mg BCG infused over 120 min once<br />

a week for 6 weeks (n�105) or to 81 mg BCG infused over 120 min once a<br />

week for 2 weeks, followed by 40 mg electromotive mitomycin (intravesical<br />

electric current 20 mA for 30 min) once a week as one cycle for three cycles<br />

(n�107). Complete responders underwent maintenance treatment: those<br />

assigned BCG alone had one infusion <strong>of</strong> 81 mg BCG once a month for 10<br />

months, and those assigned BCG and mitomycin had 40 mg electromotive<br />

mitomycin once a month for 2 months, followed by 81 mg BCG once a<br />

month as one cycle for three cycles. The primary endpoint was disease-free<br />

interval. Analyses were done by intention to treat. Results: Median follow-up<br />

was 121 months (IQR 70.5–163.5). Patients assigned sequential BCG and<br />

electromotive mitomycin had higher disease-free interval than did those<br />

assigned BCG alone (79 months [95% CI 27–139] vs 26 months<br />

[11–113]; difference between groups 53 months [39–67], log-rank<br />

p�0.0002). Patients assigned sequential BCG and electromotive mitomycin<br />

also had lower recurrence (45% [35–55] vs 62% [50–72], difference<br />

between groups 17% [6–28], log-rank p�0.0002); progression (12%<br />

[3–21] vs 28% [17.5–38.5], difference between groups 16% [5–27],<br />

log-rank p�0.003); overall mortality (44% [33–55] vs 59% [43–75],<br />

difference between groups 15% [2–28], log-rank p�0.01); and diseasespecific<br />

mortality (9% [2.5– 15.5] vs 23% [11–34], difference between<br />

groups 14% [4–24], log-rank p�0.0055). Conclusions: In patients with<br />

high risk non-muscle invasive bladder cancer intravesical BCG combined<br />

with electromotive mitomycin provided better results than BCG alone in<br />

terms <strong>of</strong> higher remission rates and longer remission times.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


4573 General Poster Session (Board #1B), Sun, 8:00 AM-12:00 PM<br />

Expression <strong>of</strong> MET and HGF in bladder cancer tumorigenesis and invasion.<br />

Presenting Author: Randy F. Sweis, University <strong>of</strong> Michigan, Ann Arbor, MI<br />

Background: The human MET gene encodes the hepatocyte growth factor<br />

(HGF) tyrosine kinase receptor. Limited studies in human bladder cancer<br />

have demonstrated that expression <strong>of</strong> MET protein is linked with disease<br />

progression and survival. We hypothesized that the expression <strong>of</strong> both MET<br />

and its ligand HGF are altered in bladder cancer. Methods: Expression <strong>of</strong><br />

MET and HGF in human bladder tissues was explored using Oncomine, an<br />

online compendium <strong>of</strong> cancer transcriptome pr<strong>of</strong>iles. The largest relevant<br />

dataset was identified and contained 157 samples (Sanchez-Carbayo, et al.<br />

2006). Normalized and log2 transformed mRNA expression values for<br />

Affymetrix U133 microarray probe sets corresponding to the genes <strong>of</strong><br />

interest were compiled and averaged for each gene. Mann-Whitney U<br />

testing was used to compare means. Nominal and standard least squares<br />

logistic regression was used to evaluate the predictive significance <strong>of</strong>, and<br />

relevance <strong>of</strong> clinicopathologic variables on, gene expression. Statistical<br />

analyses were carried out using JMP 9.0.2. Results: Expression <strong>of</strong> MET and<br />

HGF was compared across subsets <strong>of</strong> normal bladder tissue (n�48),<br />

superficial tumors (n�28), and invasive tumors (n � 81). Relative to<br />

normal tissue, MET expression was greater in superficial (p � 0.0008) and<br />

invasive tumors (p�0.0001). HGF expression was reduced in both invasive<br />

and superficial tumors vs. normal tissue (both p� 0.0001), but was higher<br />

in invasive vs. superficial tumors (p� 0.0007). In a logistic regression<br />

model <strong>of</strong> tumor samples, both MET and HGF correlated with tumor invasion<br />

(model p�0.0003). Interaction between MET and HGF was not significant<br />

(p�0.55). Neither MET nor HGF expression was predicted by regression<br />

using age, gender, grade, or nodal stage (p�0.16 and 0.27, respectively).<br />

Conclusions: Decreased HGF expression and MET over-expression are<br />

strongly associated with tumorigenesis and invasion in bladder cancer. The<br />

reduction in HGF expression is blunted in invasive vs. superficial tumors,<br />

suggesting a potential loss <strong>of</strong> negative feedback in more aggressive tumors.<br />

Expression <strong>of</strong> both genes could not be predicted by modeling clinicopathologic<br />

variables. MET signaling represents a potential therapeutic target in<br />

bladder cancer.<br />

4575 General Poster Session (Board #1D), Sun, 8:00 AM-12:00 PM<br />

Defining a nine-biomarker panel for predicting bladder cancer outcome in<br />

combination with smoking intensity: A report from the Los Angeles Cancer<br />

Surveillance Program. Presenting Author: Anirban Pradip Mitra, University<br />

<strong>of</strong> Southern California Keck School <strong>of</strong> Medicine and Norris Comprehensive<br />

Cancer Center, Los Angeles, CA<br />

Background: Urothelial carcinoma <strong>of</strong> the bladder (UCB) is a disease <strong>of</strong><br />

alterations in several cellular pathways. Routine molecular pr<strong>of</strong>iling studies<br />

do not account for smoking, a well established risk factor for UCB, and its<br />

influence on outcome. This study assessed the prognostic potential <strong>of</strong> a<br />

multi-pathway protein panel across all UCB stages in a population-based<br />

cohort after accounting for clinicopathologic factors and smoking history.<br />

Methods: 212 UCB patients from the LA CSP, part <strong>of</strong> the NCI/SEER cancer<br />

registry, were included. �Smoking intensity� analyzed biologic and molecular<br />

impact <strong>of</strong> smoking by combining smoking status, duration <strong>of</strong> smoking<br />

and number <strong>of</strong> cigarettes smoked daily into a composite covariate. Tumors<br />

were pr<strong>of</strong>iled for Bax, caspase-3, Apaf-1, Bcl-2, p53, p21, COX2, VEGF<br />

and E-cadherin alterations by IHC. Univariate analyses and multivariable<br />

modeling examined associations with outcome. Results: Median follow up<br />

was 13.2 years. Age, pathologic stage, adjuvant therapy (all p� 0.001) and<br />

surgical modality (p � 0.05) were associated with survival. Increasing<br />

smoking intensity was associated with worse outcome (P � 0.001). Apaf-1<br />

(p � 0.005), E-cadherin (p � 0.014) and p53 (p � 0.032) were<br />

univariately prognostic; E-cadherin remained prognostic after multivariate<br />

analysis (p � 0.04). Combined alterations in all 9 biomarkers were<br />

prognostic by univariate (p � 0.001) and multivariate (p � 0.006)<br />

analysis. A multivariate model that included all 9 biomarkers and smoking<br />

intensity was more accurate in predicting prognosis than models comprising<br />

<strong>of</strong> standard clinicopathologic covariates without (p � 0.001) or with (p<br />

� 0.018) smoking intensity. Conclusions: The study confirms detrimental<br />

effects <strong>of</strong> smoking on UCB prognosis. Apaf-1, E-cadherin and p53<br />

individually predicted UCB survival. Increasing number <strong>of</strong> biomarker<br />

alterations was significantly associated with worsening survival, although<br />

markers contained in the panel were not necessarily prognostic individually.<br />

Predictive value <strong>of</strong> the nine-biomarker panel with smoking intensity<br />

was significantly higher than that <strong>of</strong> routine clinicopathologic parameters<br />

alone.<br />

Genitourinary Cancer<br />

4574 General Poster Session (Board #1C), Sun, 8:00 AM-12:00 PM<br />

Results <strong>of</strong> a phase II study <strong>of</strong> pralatrexate in patients with advanced/<br />

metastatic relapsed transitional cell carcinoma <strong>of</strong> the urinary bladder.<br />

Presenting Author: Yohann Loriot, Institut Gustave Roussy, Villejuif, France<br />

Background: Antifolate agents have demonstrated activity in transitional<br />

cell carcinoma (TCC), a disease with very poor outcomes in advanced<br />

stages. Pralatrexate (FOLOTYN, Allos Therapeutics, Inc., Westminster,<br />

CO), a folate analogue targeting dihydr<strong>of</strong>olate reductase, is designed for<br />

enhanced uptake and accumulation in tumor cells. The objective <strong>of</strong> this<br />

study was to examine the activity and safety <strong>of</strong> pralatrexate in patients (pts)<br />

with advanced/metastatic TCC <strong>of</strong> the urinary bladder after failure <strong>of</strong> prior<br />

chemotherapy. Methods: Pts with histologically confirmed TCC (�50% TCC<br />

in tumor) received pralatrexate 190 mg/m² intravenously on days 1 and 15<br />

<strong>of</strong> a 28-day cycle supplemented with vitamin B12 and folic acid. Included<br />

pts had Eastern Cooperative Oncology Group performance status <strong>of</strong> 0–1,<br />

measurable disease, and prior treatment with �1 platinum- and/or methotrexate-based<br />

regimen in the recurrent/metastatic setting. Results: Thirty<br />

pts were enrolled and treated. All pts received prior platinum-based<br />

therapy, and 7 pts (23%) received methotrexate in a multidrug regimen.<br />

One pt had a confirmed partial response (PR); 4 additional pts had<br />

unconfirmed PRs. Twelve pts had stable disease. The median number <strong>of</strong><br />

cycles received was 2 (range, 1–24), and median time on treatment was 56<br />

days (range, 1–714). The median progression-free survival (PFS) and<br />

overall survival for all pts was 4.0 months (95% confidence interval [CI],<br />

2.1–4.5) and 9.3 months (95% CI, 5.6–13.2), respectively. Eight pts<br />

(27%) had PFS �6 months and 3 pts (10%) had PFS �12 months. Eight<br />

pts (27%) underwent dose reductions, all due to mucositis. Six pts (20%)<br />

discontinued the study due to treatment-related adverse events (AEs)—<br />

mainly mucositis. The most frequent treatment-related AEs were stomatitis<br />

(77%), asthenia (30%), vomiting (27%), and anemia, nausea, and<br />

neutropenia (23% each). Conclusions: Pralatrexate showed evidence <strong>of</strong><br />

activity and durable disease control when used as a single agent in pts with<br />

advanced bladder cancer, although the overall response rate was modest.<br />

Further study <strong>of</strong> pralatrexate in this setting should focus on improving drug<br />

delivery and evaluation <strong>of</strong> novel combination approaches.<br />

4576 General Poster Session (Board #1E), Sun, 8:00 AM-12:00 PM<br />

The impact <strong>of</strong> preoperative ASA score and serum albumin on early<br />

complication and survival rate <strong>of</strong> patients undergoing radical cystectomy<br />

for bladder cancer. Presenting Author: Hooman Djaladat, Institute <strong>of</strong><br />

Urology, University <strong>of</strong> Southern California, Los Angeles, CA<br />

Background: <strong>American</strong> <strong>Society</strong> <strong>of</strong> Anesthesiologist Score (ASA-S) is used to<br />

evaluate patient physical status before surgery. Serum albumin (Alb) is also<br />

a marker <strong>of</strong> nutritional status. We evaluated the impact <strong>of</strong> preoperative<br />

ASA-S and Alb on early complication rate and survival <strong>of</strong> patients who<br />

underwent radical cystectomy for bladder cancer. Methods: 1964 patients<br />

with primary bladder cancer underwent radical cystectomy between 1971<br />

and 2008 at USC. Preoperative serum Alb and ASA-S were available in<br />

1471 and 1140 patients respectively. Post cystectomy early complication<br />

was defined as any surgery related/unrelated event leading to lengthening<br />

hospital stay or re-admission within 90 days <strong>of</strong> surgery. Recurrence free<br />

survival (RFS) and overall survival (OS) for these cohorts were reviewed<br />

using a Kaplan-Meier and Cox proportional hazards models. Results: The<br />

demographic data <strong>of</strong> patients based on their serum Alb and ASA-S is<br />

presented in the Table. The median follow up was 12.4 years (0 - 36.6 yrs).<br />

Low serum Alb (�3.4 g/dL) and high ASA-S (3 or 4) were associated with<br />

higher early complication rate (43% vs. 33%, p� 0.03 and 40% vs. 28%,<br />

p� 0.0001 respectively). In multivariable analysis, low serum Alb level was<br />

an independent predictor <strong>of</strong> RFS (HR 1.35, 95% CI 1.00-1.81) and OS<br />

(HR 1.62, 95% CI 1.29-2.04). High ASA-S was an independent predictor<br />

<strong>of</strong> OS (HR 1.45, 95% CI 1.13-1.85), but not RFS. Conclusions: Preoperative<br />

low serum Alb and high ASA-S are independently predictive <strong>of</strong> post<br />

cystectomy decreased OS. Low serum Alb is also a risk factor for recurrence<br />

after cystectomy. These parameters potentially could be used in nomograms<br />

to predict post-cystectomy prognosis.<br />

Demographic data in patients who underwent cystectomy for bladder<br />

cancer based on preoperative serum Alb and ASA-S respectively.<br />

Pts No Male sex<br />

Alb< 3.4 137 92<br />

(67%)<br />

Alb> 3.4 1334 1062<br />

(79%)<br />

ASA�1, 2 400 320<br />

(80%)<br />

ASA�3, 4 740 574<br />

(78%)<br />

Median age<br />

(range) LVI<br />

74<br />

(51-90)<br />

67<br />

(30-93)<br />

60<br />

(35-90)<br />

69<br />

(33-92)<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.<br />

62<br />

(45%)<br />

373<br />

(28%)<br />

98<br />

(25%)<br />

226<br />

(30%)<br />

Multi<br />

focality<br />

46<br />

(33%)<br />

497<br />

(37%)<br />

150<br />

(38%)<br />

275<br />

(37%)<br />

High<br />

grade<br />

126<br />

(92%)<br />

1101<br />

(82%)<br />

317<br />

(79%)<br />

608<br />

(82%)<br />

295s<br />

Stage<br />

> pT3<br />

87<br />

(63%)<br />

461<br />

(35%)<br />

116<br />

(29%)<br />

258<br />

(35%)


296s Genitourinary Cancer<br />

4577 General Poster Session (Board #1F), Sun, 8:00 AM-12:00 PM<br />

FGFR3 protein expression and gene mutation in primary and metastatic<br />

urothelial carcinoma (UC) tumors. Presenting Author: Jonathan E. Rosenberg,<br />

Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute/<br />

Brigham and Women’s Hospital, Harvard Medical School, Boston, MA<br />

Background: FGFR3 protein expression may represent a valid therapeutic<br />

target in metastatic UC. The prevalence <strong>of</strong> both mutation and overexpression<br />

is unknown in metastatic UC. Methods: Tissue microarrays <strong>of</strong> formalin<br />

fixed paraffin-embedded urothelial carcinomas (UC) were stained for<br />

FGFR3 by immunohistochemistry (IHC) [primary (n�250); metastatic<br />

(n�31); <strong>of</strong> which (n�14) were paired]. FGFR3 immunostaining was scored<br />

as negative or positive based on previously reported scoring systems.<br />

FGFR3 mutation in primary tumors was assessed by iPlex and confirmed by<br />

hME sequencing (n�141) or Affymetrix OncoScan FFPE Express 2.0<br />

(primary: n�17; metastases n�31). Results: FGFR3 IHC positivity was<br />

present in 48% <strong>of</strong> metastases (95% CI�32-65%) and 26% <strong>of</strong> primary<br />

tumors, (95%�CI 21-32%), though strong staining was rare (�1%).<br />

Paired primary and metastatic tumors were both negative in 50% <strong>of</strong> cases,<br />

with 14% positive only in the metastasis, 14% positive only in the primary<br />

tumor, and 21% positive in both. If the primary tumor showed staining,<br />

71% <strong>of</strong> the metastases showed staining. FGFR3 IHC staining did not<br />

impact overall survival (p�0.8). FGFR3 mutations were observed in 9.6%<br />

<strong>of</strong> metastatic tumors (95% CI�3.3-25%), compared to 3.5% <strong>of</strong> primary<br />

tumors (95% CI�1.5%-8%). Co-occurrence <strong>of</strong> mutation and FGFR3 DNA<br />

copy number gain was observed in one specimen. Conclusions: FGFR3 IHC<br />

staining is present 26 % <strong>of</strong> primary tumors <strong>of</strong> patients who go on to develop<br />

metastatic disease, and nearly half <strong>of</strong> metastatic tumor sites. FGFR3<br />

mutation frequency in primary and metastatic tumor specimens is low.<br />

Further investigation <strong>of</strong> the frequency <strong>of</strong> FGFR3 protein expression in<br />

metastases is needed. The presence <strong>of</strong> FGFR3 protein by IHC staining in<br />

primary and metastatic specimens suggests that FGFR3 may represent a<br />

therapeutic target even in the absence <strong>of</strong> mutation. Further functional<br />

studies are needed.<br />

4579 General Poster Session (Board #2A), Sun, 8:00 AM-12:00 PM<br />

BRCA1, RAP80, and AEG-1 mRNA expression in resectable muscleinvasive<br />

bladder cancer (MIBC) patients (p) treated with neoadjuvant<br />

cisplatin-based chemotherapy. Presenting Author: Albert Font Pous, Institut<br />

Catala d’ Oncologia, Hospital Germans Trias i Pujol, Badalona, Spain<br />

Background: Cystectomy remains the standard treatment in MIBC and only<br />

a minority <strong>of</strong> p are treated with neoadjuvant chemotherapy, suggesting that<br />

predictive markers <strong>of</strong> chemotherapy outcome are needed. Low BRCA1<br />

mRNA expression is associated with an improvement in survival in bladder<br />

cancer p treated with cisplatin-based chemotherapy. However, BRCA1<br />

function can be modulated by other DNA repair genes. RAP80 is required<br />

for the accumulation <strong>of</strong> BRCA1 to sites <strong>of</strong> DNA breaks, and cells depleted<br />

<strong>of</strong> RAP80 exhibit hypersensitivity to irradiation. AEG-1 can induce BRCA1<br />

expression and cause chemoresistance. Methods: Paraffin-embedded pretreatment<br />

tumor samples were collected by transurethral resection from 65<br />

p with resectable MIBC stage T2-4N0M0 treated with neoadjuvant cisplatinbased<br />

chemotherapy. Gene expression levels <strong>of</strong> BRCA1, RAP80 and AEG-1<br />

were quantified by real-time quantitative PCR. Expression levels were<br />

divided into terciles and correlated with median survival (MS). Results: 33 p<br />

were treated with cisplatin, methotrexate and vinblastine (CMV) and 32 p<br />

with cisplatin and gemcitabine. Chemotherapy was followed by cystectomy<br />

in 60 p. Overall MS was not reached and 5-year survival was 51%. MS was<br />

45 months (m) and 5-year survival was 27% in 21 p with high BRCA1<br />

mRNA levels vs 168 m and 59% in 44 p with low and intermediate levels<br />

(p�0.05). MS was 50 m in 15 p with high AEG-1 levels, 45 m in 15 p with<br />

intermediate levels, and was not reached in 18 p with low levels, although<br />

these differences were not statistically significant (p�0.3). No differences<br />

in MS were observed according to RAP80 mRNA levels. Conclusions:<br />

BRCA1 can be a useful marker to predict the efficacy <strong>of</strong> neoadjuvant<br />

chemotherapy. Cisplatin-based chemotherapy should be recommended in<br />

p with low/intermediate BRCA1 expression. Further studies with larger<br />

numbers <strong>of</strong> p are warranted to elucidate the role <strong>of</strong> AEG-1 in this setting.<br />

4578 General Poster Session (Board #1G), Sun, 8:00 AM-12:00 PM<br />

A pre-cystectomy decision model to predict pathologic upstaging and<br />

oncologic outcomes in clinical stage T2 bladder cancer. Presenting Author:<br />

Siamak Daneshmand, University <strong>of</strong> Southern California Keck School <strong>of</strong><br />

Medicine and Norris Comprehensive Cancer Center, Los Angeles, CA<br />

Background: Neoadjuvant chemotherapy is the standard for advanced<br />

bladder cancer (BC), although its benefit in lower risk clinically organconfined<br />

muscle-invasive (cT2N0M0) BC is uncertain. This study aimed to<br />

define pre-cystectomy factors in cT2N0M0 BC that could predict pathological<br />

upstaging at cystectomy and oncological outcomes. Methods: 1,964<br />

institutional BC patients were reviewed. Neoadjuvant chemotherapy-naïve<br />

cT2N0M0 patients who underwent cystectomy were included. A multivariate<br />

classification and regression decision tree was used to assess hierarchical<br />

interactions <strong>of</strong> univariately significant variables, thereby identifying<br />

patients at greatest risk for upstaging and poor outcome. Results: 948<br />

cT2N0M0 patients were identified; 512 (54%) were upstaged at cystectomy.<br />

Pathological upstaging was associated with poor RFS and overall<br />

survival (OS) (both p � 0.001). Age, lymphovascular invasion (both P �<br />

0.01), multifocality (p � 0.004), deep muscle invasion, tumor growth<br />

pattern and hydronephrosis (all p � 0.001) were associated with upstaging.<br />

When these factors were included in a multivariate decision tree<br />

model, 71% <strong>of</strong> patients with hydronephrosis were upstaged and had the<br />

worst outcome (p � 0.001). In patients without hydronephrosis, tumor<br />

growth pattern was a second-tier discriminator; only 37% <strong>of</strong> patients with<br />

papillary tumors were upstaged. In patients with non-papillary tumors,<br />

deep muscle invasion was a third-tier discriminator; 70% <strong>of</strong> patients with<br />

invasion were upstaged. Age was a third-tier discriminator in patients with<br />

papillary � non-papillary features; 33% <strong>of</strong> patients �65 years were<br />

upstaged compared to 47% <strong>of</strong> patients �65 years. The decision tree was<br />

cross-validated and resulted in 3 risk groups – 5 year RFS/OS for low risk<br />

patients was 66%/48% versus 46%/27% for high risk patients (p �<br />

0.001). Conclusions: This study identified pre-cystectomy variables that<br />

predict upstaging and poor outcomes in cT2N0M0 BC. The decision tree<br />

approach highlighted the most crucial prognostic variables, and can aid in<br />

identifying low risk patients in a stepwise fashion who have lower probability<br />

<strong>of</strong> upstaging and better outcomes.<br />

4580 General Poster Session (Board #2B), Sun, 8:00 AM-12:00 PM<br />

Multidimensional investigation <strong>of</strong> HER2 in advanced urothelial carcinoma<br />

(UC). Presenting Author: Rachel S. Park, Lank Center for Genitourinary<br />

Oncology, Dana-Farber Cancer Institute, Boston, MA<br />

Background: Incidence <strong>of</strong> Her2 positivity and association with overall<br />

survival (OS) are controversial in advanced UC. Activating Her2 mutations<br />

have been identified in other cancers, but they have not been previously<br />

reported in UC. We determined Her2 status by immunohistochemistry<br />

(IHC), fluorescence in situ hybridization (FISH), and copy number gain<br />

(CNG) via array CGH <strong>of</strong> primary UC tumors from patients (pts) with<br />

metastatic disease. Targeted Her2 sequencing was performed at known<br />

mutation hotspots, and mutation effect was investigated in vitro. Methods:<br />

Tissue microarrays <strong>of</strong> formalin fixed paraffin-embedded tumor from 98 UC<br />

pts treated with platinum-based combination chemotherapy for metastatic<br />

disease were evaluated for Her2 protein and for Her2 gene amplification by<br />

using standard clinical protocols. Positive staining was defined as an IHC<br />

score <strong>of</strong> 3� or a FISH ratio <strong>of</strong> �2 using scoring criteria established for<br />

evaluation <strong>of</strong> breast cancer. Her2 CNG was evaluated by aCGH with cut<strong>of</strong>f<br />

log base 2 ratio � 0.9. Mutation status was validated by hME sequencing.<br />

OS was measured from start <strong>of</strong> treatment for metastatic disease. Association<br />

<strong>of</strong> OS and Her2 status was assessed by a Cox regression model.<br />

NIH-3T3 cells with Her2 V777L were assessed for growth, invasion, and<br />

Her2 kinase activation. Results: 22% <strong>of</strong> pts had 3� Her2 staining by IHC.<br />

21% <strong>of</strong> pts had FISH amplification. These were concordant in 78% <strong>of</strong> pts.<br />

CNG was identified in 16% and was concordant with FISH and IHC 85%<br />

and 88% <strong>of</strong> the time, respectively. Her2 status by any modality showed no<br />

significant association with OS in either univariate [HR�0.94, 95% CI:<br />

(0.52, 1.70), p�0.83] or multivariate [HR�1.12, 95% CI: (0.61, 2.06),<br />

p�0.72] analysis. Her2 mutations (V777L and L755S) were identified in 2<br />

pts (2%). In vitro analysis <strong>of</strong> V777L results in transformation <strong>of</strong> NIH-3T3<br />

cells, leading to increased growth, invasion on s<strong>of</strong>t agar, and Her2 kinase<br />

constitutive activation. Conclusions: Her2 overexpression or amplification<br />

in the primary tumor does not predict OS in pts with metastatic UC. Other<br />

research has suggested that V777L sensitizes cells to lapatinib, while<br />

L755S leads to lapatinib resistance. These rare oncogenic Her2 mutations<br />

occur and may be therapeutic targets in selected pts.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


4581^ General Poster Session (Board #2C), Sun, 8:00 AM-12:00 PM<br />

Phase II trial <strong>of</strong> neoadjuvant gemcitabine (G) and cisplatin (C) with<br />

sunitinib in patients (pts) with muscle-invasive bladder cancer (MIBC).<br />

Presenting Author: Arjun Vasant Balar, Memorial Sloan-Kettering Cancer<br />

Center, New York, NY<br />

Background: Response to neoadjuvant chemotherapy (NC) prior to radical<br />

cystectomy (RC) predicts improved overall survival (OS) in MIBC. Associations<br />

with enhanced survival include complete pathologic response (pT0;<br />

Grossman, NEJM 2003) and eradication <strong>of</strong> the muscle-invasive component<br />

(�pT2; Splinter, J Urol 1992). Sunitinib (S) is active in pretreated pts with<br />

advanced disease. We tested if S added to GC was safe, improved the rate<br />

<strong>of</strong> pT0, and improved the rate <strong>of</strong> �pT2. Methods: Cisplatin-eligible pts with<br />

cT2-4aN0 bladder cancer received G 1000 mg/m2 and C 35 mg/m2 on day<br />

(D) 1 and D8 with S 25 mg orally daily D1-14 <strong>of</strong> a 21D cycle for 4 cycles.<br />

RC plus pelvic lymph node dissection was required to assess response <strong>of</strong><br />

pT0 or �pT2. A Simon’s Minimax 2-stage design was used to test a null<br />

(H0) pT0 rate � 20% against alternative (H1) pT0 rate � 40% with Type I<br />

and II error rates <strong>of</strong> 0.05 and 0.10 respectively. Enrollment to the 2nd stage<br />

<strong>of</strong> 45 patients was planned if � 6 <strong>of</strong> the initial 24 evaluable pts achieved<br />

pT0. Primary endpoint was pT0N0 and secondary endpoints were: response<br />

defined as �pT2N0; safety; time to progression (TTP), and OS.<br />

Results: 18 pts (15M, 3F), median age 63 (54-76) were enrolled from 6/09<br />

and 10/11 after which financial support was withdrawn. 3 pts were<br />

inevaluable for response endpoints due to: 1.) withdrawal <strong>of</strong> consent, 2.)<br />

declining any surgery, 3.) partial cystectomy instead <strong>of</strong> RC. All 18 were<br />

evaluable for safety, TTP and OS. 1 <strong>of</strong> 15 pts had pT0N0 (6.6%; 95% CI<br />

0.34 – 29.8%) and 5 had �pT2N0 (33%; 95% CI 15-58%). 4 <strong>of</strong> 5 pts<br />

with status �pT2N0 were pTisN0. Median TTP was 10 months (95% CI<br />

3.5-NR). 3 pts were deceased at time <strong>of</strong> analysis; median OS not reached.<br />

Neutropenia due to the 3 drug combination required routine GCSF support<br />

on day 8 <strong>of</strong> each cycle. Grade 3/4 toxicities were anemia (11 pts),<br />

neutropenia (6), thromboembolic events (2), febrile neutropenia (2) and<br />

infection (2). Conclusions: Despite incomplete accrual, the pT0 rate was<br />

low suggesting that S does not add to GC. Residual non-invasive disease<br />

(�pT2) was common, including a large proportion <strong>of</strong> pts with pTisN0.<br />

Given these findings, response criteria for future NC studies should<br />

consider either �pT2 or � pTis as the primary endpoint.<br />

ABSTRACT<br />

WITHDRAWN<br />

Genitourinary Cancer<br />

297s<br />

4582 General Poster Session (Board #2D), Sun, 8:00 AM-12:00 PM<br />

PI3KCA mutations in advanced urothelial carcinoma: A potential therapeutic<br />

target? Presenting Author: Joaquim Bellmunt, University Hospital del<br />

Mar-IMIM, Barcelona, Spain<br />

Background: PI3KCA is frequently mutated in human cancer; however,<br />

information is scarce regarding its relevance in urothelial carcinoma (UC).<br />

We determined the prevalence <strong>of</strong> mutation and impact on clinical outcome<br />

<strong>of</strong> PI3KCA uniformly-treated patients with metastatic UC. Impact <strong>of</strong> PI3K<br />

and dual PI3K/mTOR inhibition was tested in vitro in UC cell lines with<br />

either H1047R or E545K mutation. Methods: 141 samples from invasive<br />

UC were scanned for mutations. Of those, complete clinical data was<br />

available from 85 cases treated with platinum-based combination chemotherapy<br />

for advanced or metastatic disease. DNA was extracted from FFPE<br />

material. Mutation status was determined by iPLEX sequencing and<br />

confirmed with hME sequencing. Overall survival (OS) was measured from<br />

beginning <strong>of</strong> treatment for metastatic disease to time <strong>of</strong> death or censored<br />

on the last known alive date. Cox proportional hazard model was used to<br />

assess the associations <strong>of</strong> PI3K mutational status and OS. Growth<br />

inhibitory effects <strong>of</strong> a specific PI3K inhibitor and a dual PI3K/mTOR<br />

inhibitor (both from Selleck) on UC cell lines with or without mutations<br />

were tested using MTT assays. Results: Mutations in the PI3KCA gene were<br />

observed in 14 (10%; 95% CI 6-16%) specimens. E545K was detected in<br />

all 14 specimens, though one specimen contained mutation at both E545K<br />

and H1047R. Among patients with clinical data, there was no statistically<br />

significant association between PI3KCA mutational status and OS (HR for<br />

having PI3KCA�0.49, 95% CI [0.15, 1.57], p-value 0.22). Preliminary in<br />

vitro experiments showed that cell growth was more potently inhibited with<br />

dual PI3K/mTOR inhibitors than with PI3K inhibitors. Conclusions: Mutations<br />

in the PI3KCA gene were detected in 10% <strong>of</strong> invasive UC and did not<br />

correlate with OS in patients with metastatic UC treated with platinumbased<br />

chemotherapy. PI3K inhibition in vitro impacts UC cell growth,<br />

though dual PI3K/mTOR inhibitors may have more significant effects than<br />

PI3K inhibition alone.<br />

4584 General Poster Session (Board #2F), Sun, 8:00 AM-12:00 PM<br />

PIK3CA gene alterations in bladder cancer: Analysis <strong>of</strong> correlative series <strong>of</strong><br />

transurethral resection (TUR)—Correlation with grade histology and tumor<br />

size. Presenting Author: Daniel E. Castellano, University Hospital 12 de<br />

Octubre, Madrid, Spain<br />

Background: PI3K pathway involvement in bladder cancer is well documented<br />

and activating mutations <strong>of</strong> the gene have been identified,<br />

particularly in tumors <strong>of</strong> low grade and stage. We have analyzed hot spot<br />

mutations and copy number in PIK3CA gene as well as mRNA expression<br />

levels in tumor tissue <strong>of</strong> pts with urothelial cell carcinoma (UCC). Methods:<br />

90 pts were analyzed. In 85 pts, fresh tissue from tumor and adjacent<br />

normal tissue was collected. All pts signed an informed consent and basic<br />

demoghrapics data were collected. Histology confirmation <strong>of</strong> UCC was<br />

required. PIK3CA alterations were analyzed by quantative-PCR. Results:<br />

Patient«s characteristics were: median age 73 years, Ta 45.5%, T1<br />

41.1%, T2 12.2%; high-grade histology 43.4%, low-grade 51,1%, PULMP<br />

4,4%. With a median follow-up <strong>of</strong> 10.0 months, 21% tumor recurrences<br />

were observed. 13% normal tissue and 51,8% tumor samples harbored<br />

alterations in PIK3CA gene, including alterations affecting the helical<br />

domain (mutations 65%, amplifications 60%, both 21%). According to T<br />

stage the distribution was: 10/36 Ta, 26/35 T1 y 6/10 T2. (p�0.05). When<br />

analyzing factors related to tumor recurrence, it was found that PIK3CA<br />

gene mutations in E542 or E545 had a lower recurrence risk (p-value<br />

²0.045). Differential expression analysis (genome-wide transcriptome analysis/microarray)<br />

showed that tumors bearing altered PIK3CA gene are more<br />

similar to non-tumoral samples comparing with tumors bearing wtPIK3CA<br />

gene. Moreover, overexpressed genes in mutant samples are mainly<br />

involved in G-protein and Wnt signaling, whereas underexpressed genes are<br />

involved in cell morphogenesis and cell polarity. Conclusions: We observed<br />

that PIK3CA gene alterations is an early carcinogenesis event in UCC pts,<br />

and more frequently found in larger tumor stages. These data support that<br />

PIK3CA status may constitute a good prognostic tool, as well as a<br />

therapeutic target in stratified groups for bladder cancer.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


298s Genitourinary Cancer<br />

4585 General Poster Session (Board #2G), Sun, 8:00 AM-12:00 PM<br />

External validation <strong>of</strong> somatic copy number alteration (SCNA) at chromosome<br />

1q23.3 in advanced urothelial carcinoma (UC). Presenting Author:<br />

Markus Riester, Department <strong>of</strong> Biostatistics and Computational Biology,<br />

Dana-Farber Cancer Institute, Boston, MA<br />

Background: Advanced UC is associated with multiple SNCAs. To identify<br />

SCNA predictors <strong>of</strong> poor survival in advanced UC, we evaluated SCNAs in<br />

two cohorts <strong>of</strong> high-risk urothelial carcinoma <strong>of</strong> the bladder. Methods: We<br />

obtained DNA copy number data from bladder cancer patients in two<br />

independent cohorts [Spanish cohort (n� 93, Agilent aCGH 180k array)<br />

and Brigham and Women’s/Dana-Farber (BW/DF) cohort (n � 48, Affymetrix<br />

OncoScan FFPE Express 2.0)]. For the BW/DF cohort, we acquired<br />

copy number information from 30 primary tumors and 33 metastases. For<br />

12 patients, both primary and matched metastases were available. The<br />

GISTIC method was used on GLAD segmented data to identify driver copy<br />

number lesions, and the NBC algorithm to identify recurrent breakpoints.<br />

Cox proportional hazards models were used to estimate the effect <strong>of</strong> the<br />

identified SCNAs on overall survival, defined as time from start <strong>of</strong><br />

chemotherapy for metastatic disease (Spanish) or from metastatic recurrence<br />

(BW/DF). Copy numbers were incorporated in the Cox models as<br />

continuous measures. Statistical significance <strong>of</strong> optimal cutpoints was<br />

estimated with permutation tests. Copy number pr<strong>of</strong>iles <strong>of</strong> primary tumors<br />

and metastases were compared with hierarchical clustering. Results:<br />

Amplification <strong>of</strong> 1q23.3 was independently associated with overall survival<br />

in both cohorts (Spanish cohort: HR 3.98; 95% 1.64-9.67; p � 0.03;<br />

C-statistic 0.54, 95% 0.45-0.65. BW/DF cohort: HR 6.28; 95% 1.84-<br />

21.4; p � 0.042; C-statistic 0.62, 95% 0.48-0.76). Amplifications at<br />

22q12.2 were enriched in patients who developed metastasis (p � 0.05).<br />

A breakpoint analysis identified possible truncations <strong>of</strong> ITPR1 (Spanish:<br />

15%; BW/DF: 25%) and PRIM2 (Spanish: 31% BW/DF: 12.5%) in large<br />

numbers <strong>of</strong> samples. For primary tumors with matched metastases, we<br />

observed that metastases clustered together with their primary tumors.<br />

Conclusions: External validation <strong>of</strong> the 1q23.3 amplification demonstrates<br />

the association <strong>of</strong> this SCNA with poor outcomes in advanced UC; the<br />

identification <strong>of</strong> the target <strong>of</strong> this copy number gain is ongoing. Gene<br />

truncations and their biological significance require further experimental<br />

investigation.<br />

4587 General Poster Session (Board #3B), Sun, 8:00 AM-12:00 PM<br />

Long-term progression-free survival (PFS) and overall survival (OS) to<br />

pemetrexed (P) as single agent in metastatic urothelial carcinoma (MUC): A<br />

Spanish Oncology Genitourinary Group (SOGUG) systematic review. Presenting<br />

Author: J. M. Cervera Grau, ITIC Benidorm, Valencia, Spain<br />

Background: The effect <strong>of</strong> pemetrexed in MUC is not well characterized.<br />

Vinflunine is the standar second-line in MUC with adjusted benefits and no<br />

more drugs have been aproved. SOGUG reports a systematic review <strong>of</strong> MUC<br />

patient series with high activity <strong>of</strong> P in monotherapy. Methods: Patients with<br />

locally advanced urothelial carcinoma and/or MUC whom received P 500<br />

mg/m(2) every 21 days with folic acid and vitamin B12 supplementation,<br />

were elected. These patients received P in second, third or fourth-line <strong>of</strong><br />

chemotherapy. Results: 44 patients have been reported (39males), median<br />

age 62 [41-82]. Of all 44 patients; 21, 22, and 1 patients received P as<br />

second-line, third and fourth-line respectively. A median <strong>of</strong> 4 courses were<br />

administered [1-12] and disease control (SD�PR�CR) was achieved in 19<br />

patients for an overall control rate <strong>of</strong> 43.2%. Four groups with significant<br />

differences in PFS (p�.033) were established (1.bone-metastasis, 2.visceral-metastasis,<br />

3. nodal-visceral-metastasis and 4.nodal-metastasis). OS<br />

was significant between 2nd and 4th group (p�.036) . Mean PFS and OS<br />

were 125days [17-606] and 219days [17-1168] respectively for all 44<br />

patients. Mean PFS and OS <strong>of</strong> 8 patients with bone metastasis were 75<br />

days and 134 days. Mean PFS and OS <strong>of</strong> 10 patients with visceral<br />

metastasis were 65 days and 144 days. Mean PFS and OS <strong>of</strong> 8 patiens with<br />

nodal�visceral metastasis were 154 and 228 days. Mean PFS and OS <strong>of</strong><br />

18 patients with nodal metastasis were 166 days and 299 days. Conclusions:<br />

Our longer and multicenter follow-up shows that single agent P in<br />

monotherapy as second, third and fourth line in MUC is associated with<br />

high activity and long-time survival specially in metastatic nodal MUC.<br />

These results suggest that P as monotherapy requires to be further studied,<br />

more patients are being collected.<br />

4586 General Poster Session (Board #3A), Sun, 8:00 AM-12:00 PM<br />

A phase II trial <strong>of</strong> neoadjuvant dasatinib (Neo-D) in muscle-invasive<br />

urothelial carcinoma <strong>of</strong> the bladder (miUCB): Hoosier Oncology Group<br />

GU07-122 trial. Presenting Author: Noah M. Hahn, Indiana University<br />

Melvin and Bren Simon Cancer Center, Indianapolis, IN<br />

Background: Neoadjuvant chemotherapy (NC) preceding radical cystectomy<br />

(RC) is an accepted standard for miUCB patients (pts). Dasatinib (D) is an<br />

oral tyrosine kinase inhibitor <strong>of</strong> Src mediated signaling, and has demonstrated<br />

promising preclinical anti-tumor activity, providing a rationale to<br />

evaluate Neo-D in human miUCB. Methods: A phase II trial was conducted<br />

to assess the safety and biologic activity <strong>of</strong> Neo-D in miUCB. Key eligibility<br />

criteria included: miUCB (T2-T4a, N0, M0), unsuitable or willing to forego<br />

platinum-based NC, adequate TURBT tissue available, ECOG PS 0-1,<br />

creatinine � 2 x ULN. Pts received D 100 mg po once daily for 28 �/- 7<br />

days followed by RC 8-24 hours after the last dose. The primary endpoint<br />

was feasibility defined as � 60% <strong>of</strong> pts completing therapy without<br />

treatment-related dose limiting toxicity (DLT) specified as grade 4 hematologic<br />

toxicity, grade 3 non-hematologic toxicity, or any grade 2 toxicity � 14<br />

days. Secondary endpoints included the assessment <strong>of</strong> biologic activity as<br />

assessed by pre- and post-treatment tumor tissue immunohistochemistry<br />

analysis <strong>of</strong> Src, pSrc, EPHA2, pEPHA2, FAK, pFAK, AKT, pAKT, CD31,<br />

Ki67, TUNEL. Results: The study completed accrual with enrollment <strong>of</strong><br />

25pts. 22 and 24 pts were respectively evaluable for feasibility and toxicity<br />

endpoints. Patient demographics included: median age – 62, M/F – 20/5,<br />

ECOG PS 0/1 – 19/6. Baseline tumor staging included: T2 – 17, T3 – 7.<br />

The study achieved its primary endpoint with 15 pts (68%) completing<br />

therapy without treatment related DLT’s. DLT’s included: fatigue (2 pts),<br />

DVT/PE, abdominal pain, supraventricular tachycardia, enteric fistula, and<br />

hematuria (1 pt each). Frequency <strong>of</strong> highest observed toxicity on study<br />

included: Grade 1 – 13%, Grade 2 – 38%, Grade 3 – 46%, Grade 4–4%.<br />

Among 22 patients, pathologic stage at RC was T1/Tis in 3 pts (14%), �T2<br />

in 19 pts (86%), and node positive in 6 pts (27%). Correlative analyses <strong>of</strong><br />

pre- and post-treatment tumor Src signaling are ongoing and will be<br />

updated at the meeting. Conclusions: Neoadjuvant dasatinib preceding RC<br />

is feasible in miUCB patients. Tumor tissue correlative studies may provide<br />

directions for further development.<br />

4588 General Poster Session (Board #3C), Sun, 8:00 AM-12:00 PM<br />

<strong>Clinical</strong> validation <strong>of</strong> the 7th AJCC-TNM nodal staging system in lymph<br />

node-positive patients undergoing radical cystectomy. Presenting Author:<br />

Eila C. Skinner, USC Institute <strong>of</strong> Urology, University <strong>of</strong> Southern California,<br />

Los Angeles, CA<br />

Background: The AJCC TNM staging system for bladder cancer has recently<br />

been updated. In the new 7th edition, staging <strong>of</strong> lymph node (LN)<br />

metastases is based on anatomical location instead <strong>of</strong> size as in the former<br />

6th edition. This study evaluated whether the prognostic value <strong>of</strong> nodal<br />

staging according to the 7th edition is superior to the 6th edition. Methods:<br />

Prospectively collected data <strong>of</strong> patients who underwent radical cystectomy<br />

(RC) at a high-volume center between 2002 and 2008 were reviewed.<br />

Patients who underwent RC for non-urothelial cancer, received neoadjuvant<br />

therapy or had non-LN metastatic disease at the time <strong>of</strong> RC were<br />

excluded. All patients underwent RC with extended lymphadenectomy up<br />

to the inferior mesenteric artery. LNs were submitted in predesignated<br />

anatomically defined packets. Detailed data on the number, size and<br />

location <strong>of</strong> LN metastases were recorded. Both the 6th and 7th edition AJCC<br />

TNM editions were used to stage LN positive (LN�) disease. Median<br />

follow-up time was 2.8 years. Kaplan-Meier analysis was used to estimate<br />

overall survival (OS) and recurrence-free survival (RFS). Results: A total <strong>of</strong><br />

637 patients were identified <strong>of</strong> whom 141 patients (22.1%) had LN�<br />

disease. In total, 83 patients (58.9%) received adjuvant chemotherapy.<br />

Administration <strong>of</strong> adjuvant chemotherapy did not differ significantly per<br />

N-stage according to both editions. Based on the 6th edition, 31 patients<br />

(22.0%) had N1 disease, 105 patients (74.5%) N2 disease and 5 patients<br />

(3.5%) N3 disease. Three-year RFS rates for N1, N2 and N3 disease were<br />

51%, 43% and 0% respectively (p � 0.87). Based on the 7th edition, 29<br />

patients (20.6%) had N1 disease, 60 patients (42.6%) N2 disease and 52<br />

patients (36.9%) N3 disease. Three-year RFS rates for N1, N2 and N3<br />

disease were 51%, 42% and 45%, respectively (p � 0.84). Conclusions:<br />

Neither the AJCC-TNM 7th edition (location-based) LN staging nor the 6th edition (size-based) staging performed well as a prognostic tool for the<br />

patients in this cohort. The new staging system moved a large percent <strong>of</strong><br />

patients into the N3 category, which did not have a worse prognosis than<br />

either the N1 or N2 categories. A better staging system for LN� bladder<br />

cancer patients needs to be developed.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


4589 General Poster Session (Board #3D), Sun, 8:00 AM-12:00 PM<br />

Mortality from other malignancies in bladder cancer survivors. Presenting<br />

Author: Hamed Ahmadi, USC Institute <strong>of</strong> Urology, University <strong>of</strong> Southern<br />

California, Los Angeles, CA<br />

Background: Other-cause mortality represents a considerable proportion <strong>of</strong><br />

overall long-term mortality in bladder cancer (BC) patients who underwent<br />

curative radical cystectomy (RC). Other malignancies besides BC are<br />

reportedly among the most common etiologies <strong>of</strong> other-cause mortality. We<br />

report on mortality from other malignancies in BC survivors who have<br />

undergone prior RC. Methods: Data on BC patients who died <strong>of</strong> other<br />

malignancies were derived from a cohort <strong>of</strong> 1964 patients with pathologic<br />

diagnosis <strong>of</strong> urothelial carcinoma <strong>of</strong> bladder who underwent RC with intent<br />

to cure due to BC at University <strong>of</strong> Southern California between 1971 and<br />

2008. For subsequently developed malignancies, the time interval between<br />

RC and occurrence <strong>of</strong> malignancy was calculated. Other malignancies<br />

mortality was evaluated with respect to gender, pathological stage <strong>of</strong><br />

BC, and type <strong>of</strong> malignancy. Results: Other malignancies were diagnosed in<br />

463 (23.5%) patients. They were the cause <strong>of</strong> death in 122 (6.21%)<br />

patients (109 male) who had a median age <strong>of</strong> 68.6 (range 43.4 – 88.7) at<br />

time <strong>of</strong> RC. Of all 122 patients with other malignancies, 22(18%) patients<br />

were diagnosed prior to RC, 5 (4%) concurrently with BC diagnosis, and 94<br />

(77%) subsequent to RC. At the time <strong>of</strong> cystectomy, 87/122 (71.3%)<br />

patients had lymph node-negative organ-confined disease, compared to<br />

56.3% in the whole cohort; and 18 (14.7%) patients had nodal disease,<br />

compared to 23.2% in the whole cohort. 116/122 (95%) <strong>of</strong> these patients<br />

had no evidence <strong>of</strong> BC recurrence at time <strong>of</strong> death. Secondary cancers<br />

occurred at a median <strong>of</strong> 7.7 years (Range 3 months – 29.9 years) after RC.<br />

Most common sites and types <strong>of</strong> malignancies that led to death were lung in<br />

43/122 (35.2%), predominantly non-small cell carcinoma in 17/<br />

43(39.5%); hematologic in 15/122 (12.3%), largely lymphoma in 6/15<br />

(40%); and colon in 11/122 (9%), primarily adenocarcinoma in 8/11<br />

(72.7%) patients. Conclusions: Lung, hematologic, and colon cancers are<br />

the most common causes <strong>of</strong> death due to other malignancies in BC<br />

survivors and more commonly found in male patients with lymph nodenegative<br />

organ-confined tumor. Continued screening for other cancers in<br />

the follow-up <strong>of</strong> BC survivors may be warranted.<br />

4591 General Poster Session (Board #3F), Sun, 8:00 AM-12:00 PM<br />

Novel oncogenic function <strong>of</strong> ATDC in bladder cancer. Presenting Author:<br />

Phillip Lee Palmbos, University <strong>of</strong> Michigan Health System, Ann Arbor, MI<br />

Background: Bladder cancer is a common and deadly disease, but the<br />

molecular events leading to its initiation and progression are incompletely<br />

understood. We recently identified Ataxia-Telangiectasia Group D Associated<br />

(ATDC) as a novel oncogene which drives tumor proliferation and<br />

invasion in pancreatic carcinoma (Cancer Cell, 2009). In this study, we<br />

describe the role <strong>of</strong> ATDC as an oncogene in bladder cancer. Methods: To<br />

further determine the oncogenic role <strong>of</strong> ATDC, we generated ATDC<br />

transgenic (tg) mice in which ATDC expression was driven by a CMV<br />

promoter and characterized the resulting tumors. Results: Interestingly, the<br />

dominant phenotype in these mice was the development <strong>of</strong> both noninvasive<br />

and invasive urothelial carcinomas (9% and 20% respectively,<br />

average age <strong>of</strong> onset 10-12 months <strong>of</strong> age). Histologically, these tumors<br />

were indistinguishable from human urothelial carcinomas. Gene expression<br />

pr<strong>of</strong>iling <strong>of</strong> invasive tumors derived from ATDC tg mice demonstrated a<br />

marked overlap with gene signatures <strong>of</strong> human invasive bladder cancers.<br />

ATDC was the 11th most highly up-regulated gene in bladder cancers<br />

represented in the Oncomine gene expression database. Analysis <strong>of</strong> a<br />

human bladder cancer tissue microarray (311 samples) by IHC showed<br />

elevated expression in 70% (173/252) <strong>of</strong> muscle-invasive carcinomas,<br />

22% (5/23) <strong>of</strong> papillary tumors and little or no expression in normal<br />

bladder urothelium. ATDC tg mouse bladder tumors demonstrated loss <strong>of</strong><br />

p53 signaling and down-regulation <strong>of</strong> PTEN expression, which correlated<br />

with ATDC induced methylation <strong>of</strong> the PTEN promoter by DNMT3A.<br />

Furthermore, ATDC knock-down in invasive cancer cell lines resulted in<br />

decreased proliferation, invasion and reactivation <strong>of</strong> p53-mediated signaling<br />

and PTEN expression. Conclusions: ATDC is a novel oncogene that is<br />

highly expressed in human bladder cancers and is sufficient to drive the<br />

development <strong>of</strong> invasive bladder tumors in tg mice. The mechanism by<br />

which ATDC drives bladder cancer formation involves alterations in p53<br />

and PTEN pathways known to be important in bladder tumorigenesis.<br />

Genitourinary Cancer<br />

299s<br />

4590 General Poster Session (Board #3E), Sun, 8:00 AM-12:00 PM<br />

Second-line treatment <strong>of</strong> advanced urothelial cancer with paclitaxel and<br />

RAD001 (everolimus) in a German phase II trial (AUO trial AB 35/09).<br />

Presenting Author: Guenter Niegisch, Department <strong>of</strong> Urology, Heinrich-<br />

Heine-University, Duesseldorf, Germany<br />

Background: Efficacy <strong>of</strong> second-line treatment after cisplatinum failure in<br />

patients (pts) with advanced urothelial cancer is limited. Based on<br />

encouraging preclinical data and results from several phase I trials in solid<br />

cancers, we evaluated the safety and efficacy <strong>of</strong> the combination <strong>of</strong><br />

paclitaxel and RAD001 (everolimus) in these pts. Methods: In this singlearm,<br />

multicenter phase II trial, pts failing prior cisplatinum based combination<br />

treatment for advanced urothelial cancer were treated with paclitaxel<br />

(175 mg/m² i.v., three-weekly) and the mTOR-inhibitor RAD001 (10 mg<br />

p.o., once daily). Pts were treated until tumor progression by RECIST<br />

criteria or until a maximum <strong>of</strong> 6 cycles was completed. A one-stage design<br />

was used to evaluate the overall response rate (ORR) as primary endpoint.<br />

Results: 27 pts (18 men, 9 women; median age 63 (35-76) years; ECOG �<br />

1: 11/27pts, hepatic metastases: 10 pts, Hb�10 g/dl: 8 pts; upper urinary<br />

tract: 9 pts, lower urinary tract 15 pts, both: 3 pts) were enrolled between<br />

07/09 and 12/11. As <strong>of</strong> 01/12, 19 pts are evaluable for the primary<br />

endpoint and the toxicity pr<strong>of</strong>ile (6 pts still on treatment, 1 patient has<br />

withdrawn consent). Main toxicities (all CTCAE grades) were anemia<br />

(10/19 pts), leucopenia (8/19 pts), and neuropathy (9/19 pts). Grade III/IV<br />

toxicities were anemia (4/19 pts) and leucopenia (4/19 pts). No treatment<br />

related deaths were observed. Median number <strong>of</strong> cycles was 4 (1-6).<br />

Complete remission (CR) and partial remission (PR) was observed in 0/19<br />

and 3/19 pts, respectively (ORR 16%). Stable disease (SD) was observed in<br />

10/19 pts. Median time-to-progression (TTP) was 2.7 months (95%<br />

confidence interval [CI] 1.6 – 4.4), median overall survival (OS) was 6.5 (CI<br />

4.5 – 9.2) months. Conclusions: Frequency and severity <strong>of</strong> toxicities were<br />

acceptable. Using the combination <strong>of</strong> RAD001 and paclitaxel as secondline<br />

treatment for advanced urothelial cancer after cisplatinum-failure,<br />

response rates and survival times comparable to vinflunine were observed.<br />

4592 General Poster Session (Board #3G), Sun, 8:00 AM-12:00 PM<br />

External validation <strong>of</strong> prognostic models for overall survival (OS) in patients<br />

(pts) with advanced cancer (UC) treated with cisplatin-based chemotherapy.<br />

Presenting Author: Andrea Borghese Apolo, Medical Oncology<br />

Branch, National Cancer Institute, National Institutes <strong>of</strong> Health, Bethesda,<br />

MD<br />

Background: The most commonly used model predicting OS for UC pts<br />

treated with cisplatin-based chemotherapy is based on 2-variables (visceral<br />

metastases and performance status), developed at MSKCC in 1999, and<br />

validated in a phase III study (DeSantis JCO 2011). A prognostic model <strong>of</strong><br />

OS for advanced UC pts based on 4 variables (visceral metastases,<br />

albumin, performance status, and hemoglobin) was developed using 308<br />

pts from MSKCC (ASCO 2007 abstr 5055). We report the discriminative<br />

ability <strong>of</strong> the 4- and 2- variable models for advanced UC pts using an<br />

independent dataset from CALGB 90102. Methods: The analysis was<br />

performed using an external multi-institutional dataset from CALGB<br />

90102. The primary measurement <strong>of</strong> predictive discrimination was Harrell’s<br />

c-index which was computed with 95% confidence interval (CI). To<br />

assess whether there was a statistically significant difference in discrimination<br />

between the two models, the U statistic was used to test whether the<br />

predictions <strong>of</strong> the 4-variable model in all possible pairs were more<br />

concordant with actual observations than the 2-variable model in the same<br />

pairs. Results: CALGB 90102 included 74 UC pts (58 males, 16 females),<br />

median age 64 years, treated with cisplatin, gemcitabine and gefitinib,<br />

enrolled from 7/02 to 4/05 with a median follow-up <strong>of</strong> 72.5 months.<br />

Visceral metastases were present in 64% (bone, 18%, liver, 31%, lung,<br />

43%), median KPS 90%. The MSKCC 2-variable risk group distribution<br />

was 30% �0, 65%� 1 and 5%�2. The median OS �12.7 months (95%<br />

CI�10.4-20.5) with 68 deaths observed. When applied to the CALGB<br />

cohort, the predictive accuracy for the 4- and 2-variable models were 0.63<br />

(95 CI� 0.56- 0.69) and 0.58 (95% CI� 0.52-0.65), respectively. There<br />

was a statistically significant difference in discrimination between the two<br />

models (p �0.019), with superiority <strong>of</strong> the 4-variable model compared to<br />

the 2-variable model. Conclusions: A 4-variable prognostic nomogram for<br />

survival in pts with advanced UC was superior to a 2-variable risk-group<br />

model. The 4-variable prognostic model may replace the widely used<br />

2-variable model and can be used in the design and conduct <strong>of</strong> future<br />

phase II and III trials in advanced UC.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


300s Genitourinary Cancer<br />

4593 General Poster Session (Board #3H), Sun, 8:00 AM-12:00 PM<br />

Activity <strong>of</strong> intravesical CG0070 in Rb-inactive superficial bladder cancer<br />

after BCG failure: Updated results <strong>of</strong> a phase I/II trial. Presenting Author:<br />

Terence W. Friedlander, University <strong>of</strong> California, San Francisco, San<br />

Francisco, CA<br />

Background: Loss <strong>of</strong> retinoblastoma (Rb) tumor suppressor activity occurs<br />

commonly in bladder cancer and leads to upregulation <strong>of</strong> the E2F-1<br />

transcription factor. CG0070 is a replication-competent oncolytic adenovirus<br />

genetically modified to express GMCSF under control <strong>of</strong> the human<br />

E2F-1 promoter. Updated results from a phase I study <strong>of</strong> CG0070 in<br />

patients with recurrent superficial bladder cancer (T1, Ta, Tcis) after BCG<br />

treatment are presented here. Methods: The V-0046 phase I/II study<br />

previously reported the safety and evidence <strong>of</strong> efficacy <strong>of</strong> single and<br />

multiple doses <strong>of</strong> intravesical CG0070. Efficacy was determined using<br />

quarterly cystoscopy, biopsy, and/or urine cytology. Tumor Rb status was<br />

assessed immunohistochemically in 18 tumors. Results: 35 patients were<br />

treated with either a single dose (n�13) <strong>of</strong> CG0070 ranging from 1x1012 to<br />

3x1013 viral particles (vp) or with weekly x6(n�9) or every 4 week x 3-6<br />

doses (n�13) ranging from 1x1012 to 1x1013 vp per dose. The most<br />

common adverse events regardless <strong>of</strong> schedule were flu-like illness,<br />

dysuria, hematuria, bladder spasm, and nocturia. No maximum tolerated<br />

dose was reached. Urine GMCSF and CG0070 levels were detectable in<br />

almost all patients suggesting in-vivo viral replication. The CR rate in the<br />

single dose cohort was 23% (3/13), and 64% (14/22) in the multi-dose<br />

cohorts. The CR rate for patients with either CIS or Ta tumors was 65%<br />

(15/23). Of all responders, 11 recurred with a remission duration ranging<br />

from 3.0 - 33.4 months and 6 patients, all in the multiple dose cohorts,<br />

remain in remission as <strong>of</strong> last follow-up with a remission duration ranging<br />

from 3.3 – 38.2 months. Phosphorylated (inactive) Rb was detected in 13<br />

<strong>of</strong> the 18 (72%) patients evaluated. Nine <strong>of</strong> these 13 patients (70%) had a<br />

complete response. All patients (5/5, 100%) with known phosphorylated<br />

Rb treated in the weekly cohort experienced a CR. Conclusions: CG0070<br />

was well tolerated with minimal toxicities. Complete responses were more<br />

frequently observed in the multiple-dose cohorts, including durable CRs in<br />

patients with inactive Rb and in patients with CIS. Further study in<br />

Rb-inactive superficial tumors is warranted.<br />

4595 General Poster Session (Board #4B), Sun, 8:00 AM-12:00 PM<br />

Late gastrointestinal (GI) complications in patients with stage I-II testicular<br />

seminoma treated with radiotherapy (RT). Presenting Author: Christopher<br />

Leigh Hallemeier, Mayo Clinic, Rochester, MN<br />

Background: For stage I-II testicular seminoma, RT is highly effective at<br />

eradicating disease in the abdominopelvic lymph nodes, but results in<br />

unnecessary exposure to normal tissues including the GI tract. The purpose<br />

<strong>of</strong> this study was to define the incidence and risk factors for late GI<br />

complications in this patient population. Methods: A retrospective review<br />

was performed <strong>of</strong> 251 patients with stage I-II testicular seminoma treated<br />

with curative intent RT at our institution from 1974-2009. All patients<br />

underwent orchiectomy and postoperative external beam RT to the involved<br />

and/or at-risk nodal basins. Potential late GI complications that were<br />

assessed included endoscopically-confirmed gastric or duodenal ulceration,<br />

small bowel obstruction (SBO), and biopsy-confirmed malignancy <strong>of</strong><br />

the GI tract. Risks were estimated using the Kaplan-Meier (KM) technique<br />

and univariate/multivariate analyses were performed using the Cox proportional<br />

hazards model. Results: Median age at diagnosis was 36 years (range<br />

18 – 80). <strong>Clinical</strong> stage was I (n�199) or II (n�52). Median abdominopelvic<br />

RT dose was 26 Gy (interquartile range 25 – 30). Median follow-up was<br />

15 years (range 0.1 – 38). KM estimates for any GI complication (ulcer,<br />

SBO, or GI malignancy) at 10, 20, and 30 years were 7, 10 and 24%,<br />

respectively. Four patients died as result <strong>of</strong> a GI complication. KM<br />

estimates for ulcer at 10, 20, and 30 years were 4, 7, and 9%, respectively.<br />

Age at RT (Hazard Ratio [HR] 1.05, 95% Confidence Interval [CI] 1.00 –<br />

1.10, p�0.03) and RT total dose (per Gy, HR 1.20, 95% CI 1.09 – 1.31,<br />

p�0.01) were associated with risk <strong>of</strong> ulcer. KM estimates for SBO at 10,<br />

20, and 30 years were 2%, 2%, and 3%, respectively. History <strong>of</strong><br />

inflammatory bowel disease was associated with risk <strong>of</strong> SBO (HR 43, 95%<br />

CI 7-325, p�0.01). KM estimates for GI malignancy at 10, 20, and 30<br />

years were 0.5, 3 and 16%, respectively. Age at RT was associated with risk<br />

<strong>of</strong> GI malignancy (HR 1.07, 95% CI 1.02-1.14, p�0.01). Conclusions: In<br />

patients with stage I-II testicular seminoma treated with RT, late GI<br />

complications were a relatively uncommon, but clinically significant source<br />

<strong>of</strong> late morbidity. Use <strong>of</strong> low dose, limited field, and/or proton RT may<br />

reduce these risks.<br />

4594 General Poster Session (Board #4A), Sun, 8:00 AM-12:00 PM<br />

Utilization and predictors <strong>of</strong> neoadjuvant chemotherapy for muscleinvasive<br />

bladder cancer in the Veterans Health Administration. Presenting<br />

Author: Gurdarshan Singh Sandhu, Washington University School <strong>of</strong><br />

Medicine, St. Louis, MO<br />

Background: The survival benefit with neoadjuvant chemotherapy for<br />

bladder cancer was established in 2003. However, adoption <strong>of</strong> this<br />

paradigm in clinical practice has been slow. We explored the use <strong>of</strong><br />

neoadjuvant chemotherapy and identified predictors <strong>of</strong> its use in a<br />

contemporary cohort in the Veterans Health Administration (VA). Methods:<br />

Using the national VA <strong>Clinical</strong> Cancer Registry, all patients diagnosed with<br />

clinical stage T2-4, N0 or Nx, M0 bladder cancer from 1997 to 2007 were<br />

stratified into surgically (radical cystectomy [RC], n�1,211) and nonsurgically<br />

managed groups (n�2,125). Receipt <strong>of</strong> neoadjuvant chemotherapy<br />

was defined as chemotherapy treatment up to 6 months before RC as well<br />

as initial treatment only with chemotherapy (without subsequent surgery or<br />

radiation) in the nonsurgical group. Temporal trends in neoadjuvant<br />

chemotherapy use were evaluated with a chi square test. Predictors <strong>of</strong><br />

neoadjuvant chemotherapy were examined using a multivariable logistic<br />

regression model incorporating demographic, socioeconomic, comorbid,<br />

pathologic and hospital factors. Results: 6.3% and 8.3% <strong>of</strong> patients<br />

received neoadjuvant chemotherapy in the surgical and non-surgical group,<br />

respectively. Analysis <strong>of</strong> temporal trends in chemotherapy use demonstrated<br />

an increase in neoadjuvant chemotherapy use over time (p�0.0001);<br />

from 3% (2003 and before) to 14% annually (2007). On multivariable<br />

analysis <strong>of</strong> both groups, older age and more recent diagnosis were<br />

predictive <strong>of</strong> neoadjuvant chemotherapy use (Table). Other covariates did<br />

not predict receipt <strong>of</strong> neoadjuvant chemotherapy. Conclusions: While<br />

overall use <strong>of</strong> neoadjuvant chemotherapy in the VA population with bladder<br />

cancer remains low, use there<strong>of</strong> is slowly increasing, with a more recent<br />

diagnosis most strongly predicting its use.<br />

Multivariable predictors <strong>of</strong> neoadjuvant chemotherapy in bladder cancer.<br />

OR 95% CI P value<br />

Age (per year older)<br />

Year <strong>of</strong> diagnosis (reference 2003 and before)<br />

1.02 1.00-1.04 �.0001<br />

2004 1.56 0.98-2.47 0.06<br />

2005 1.23 0.76-2.00 0.39<br />

2006 1.66 1.09-2.52 0.02<br />

2007 2.38 1.59-3.57 �0.0001<br />

4596 General Poster Session (Board #4C), Sun, 8:00 AM-12:00 PM<br />

Interim results <strong>of</strong> phase II trial <strong>of</strong> the cyclin-dependent kinase 4/6 inhibitor<br />

PD-0332991 in refractory retinoblastoma protein positive germ cell<br />

tumors. Presenting Author: David J. Vaughn, Abramson Cancer Center at<br />

the University <strong>of</strong> Pennsylvania, Philadelphia, PA<br />

Background: Deregulation <strong>of</strong> the retinoblastoma pathway in germ cell<br />

tumors (GCT) has been well documented. We previously reported that<br />

PD-0332991, a selective oral inhibitor <strong>of</strong> cyclin-dependent kinase 4/6, led<br />

to prolonged disease control in 3 patients (pts) with unresectable growing<br />

teratoma syndrome (NEJM, 2009). For these reasons, we initiated a phase<br />

II trial <strong>of</strong> PD-0332991 in pts with refractory retinoblastoma protein (Rb)<br />

positive (�) GCT. Methods: Pts with incurable refractory GCT that expressed<br />

Rb by immunohistochemistry were treated with PD-0332991 125 mg<br />

orally daily for 21 days followed by a 7 day break (cycle � 28 days). Tumor<br />

assessments were performed every 2 cycles. The primary endpoint was<br />

6-month progression-free survival (PFS) rate. Results: As <strong>of</strong> 1/12/2012,<br />

archived tumors from 36 pts with refractory GCT were stained for Rb and 35<br />

had � 1� staining. 18 <strong>of</strong> planned 24 pts have been enrolled and treated.<br />

Pt characteristics: 17 male, 1 female; median age, 31 years (range,<br />

17-56); median ECOG performance status, 1 (range, 0-1); median number<br />

<strong>of</strong> prior chemotherapy regimens, 2 (range, 1-6); median number prior<br />

surgeries, 3 (range, 1-6). Pt pathology: mature teratoma (MT), 7; teratoma<br />

with malignant transformation (TMT), 7; mixed GCT, 2; late relapse (LR),<br />

2. 16 pts are evaluable; 2 pts are too early to evaluate. 5 <strong>of</strong> 16 evaluable pts<br />

achieved 6-month PFS (3 MT, 1 TMT, 1 LR). Median PFS was 2 months<br />

(range, 0-17). No objective radiological responses were observed; 7<br />

patients had best response <strong>of</strong> stable disease by RECIST criteria (3 MT, 3<br />

TMT, 1 LR). Grade 3 toxicity included neutropenia (4 pts), thrombocytopenia<br />

(3 pts), anemia (1 pt), mucositis (1 pt). No grade 4 toxicity was seen.<br />

Analysis <strong>of</strong> archival tumor for predictive biomarkers including Rb and p16<br />

expression is being performed. Conclusions: PD-0332991 has resulted in<br />

6-month PFS in pts with refractory Rb � GCT, including pts with incurable<br />

teratomas.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


4597 General Poster Session (Board #4D), Sun, 8:00 AM-12:00 PM<br />

Bone metastases in patients with relapsed/refractory germ cell tumors<br />

(GCT) undergoing first salvage chemotherapy: A retrospective analysis <strong>of</strong> a<br />

large international database. Presenting Author: Karin Oechsle, University<br />

Medical Center Eppendorf, Hamburg, Germany<br />

Background: Bone metastases (mets) are rare events in GCT patients (pts)<br />

and data on characteristics, risk factors, treatment and outcome in these<br />

pts are largely missing. Methods: A subgroup <strong>of</strong> 104 ptswith bone metsfrom<br />

the IPFSG database was analyzed. All patients experienced unequivocal<br />

relapse/progression after � 3 cycles <strong>of</strong> cisplatin-based chemotherapy and<br />

received conventional-dose (CDCT) or high-dose salvage chemotherapy<br />

(HDCT). Results: 104 / 1594 pts presented with bone mets (6,5%) at first<br />

relapse. Histology was pure seminoma in 26%, and non-seminoma in 74%.<br />

At initial diagnosis, 54% <strong>of</strong> pts had presented with �poor risk� according to<br />

IGCCCG. Overall response rate (ORR; CR�PR) to 1st-line chemotherapy<br />

was 89%. Median PFS after primary treatment was 3 months (range 0 -<br />

140). Bone mets at first relapse were accompanied by abdominal, lung,<br />

mediastinal, liver and brain mets in 54%, 48%, 28%, 36%, and 14%,<br />

respectively. Tumor marker pr<strong>of</strong>iles were heterogeneous (elevation <strong>of</strong> AFP /<br />

ß-HCG: 51% / 37%). Salvage treatment was CDCT in 34%, and HDCT in<br />

66% <strong>of</strong> pts. ORR to salvage chemotherapy was 68% for the total cohort,<br />

and 43% vs. 81% for CDCT vs. HDCT (p�.01). For the total cohort, 2y-PFS<br />

and 5y-OS were 24% and 16%, respectively. 2y-PFS and 5y-OS were<br />

significantly higher in pts after HDCT compared to CDCT (2y-PFS 29% vs.<br />

14%, p�.01; 5y-OS 19% vs. 9%, p�.04). Conclusions: Pts relapsing with<br />

bone mets after platinum-based CTX were characterized by �poor risk�<br />

disease at initial diagnosis, but characteristics at first relapse were<br />

heterogeneous. Treatment response and outcome was improved in pts with<br />

HDCT compared to CDCT. Further prospective evaluation <strong>of</strong> characteristics<br />

and treatment approaches in GCT pts with bone mets is warranted.<br />

4599 General Poster Session (Board #4F), Sun, 8:00 AM-12:00 PM<br />

<strong>Clinical</strong> relevance <strong>of</strong> specialist pathologic testicular tumor review. Presenting<br />

Author: David N. Church, Oxford Cancer Centre, University <strong>of</strong> Oxford,<br />

Oxford, United Kingdom<br />

Background: Therapeutic options in stage IA/B testicular cancer include<br />

surveillance, retroperitoneal lymph node dissection, and adjuvant chemotherapy/radiotherapy;<br />

clinician and patient choice is guided by recurrence<br />

risk. Specialist pathologic review is important in informing this decision as<br />

it may alter diagnosis and staging. Methods: Analysis <strong>of</strong> a prospective<br />

supraregional database <strong>of</strong> 627 testicular tumors diagnosed in Oxford or<br />

referred for central review between 2004 and 2012. Tumor histology, stage<br />

(AJCC 7th ed.), and other factors predictive <strong>of</strong> relapse were compared<br />

between referring and final pathology reports. Results: Of 402 cases<br />

referred from 11 hospitals during the study period, 370 primary testicular<br />

tumors with an informative initial pathology report were analysed. There<br />

was a discrepancy between referring and final reports in 116 cases<br />

(31.4%), with significant variation between referring units in the frequency<br />

<strong>of</strong> discordance (20.0-89.9%, p�0.0001, x2 ). Changes to histological<br />

diagnosis in 34 cases (9.2%) were minor alterations in type and proportion<br />

<strong>of</strong> non-seminomatous germ cell tumor (NSGCT) elements <strong>of</strong> no clinical<br />

significance, with exception <strong>of</strong> one classical seminoma reclassified as<br />

spermatocytic seminoma. For seminomas (n�201) the commonest discrepancies<br />

were in identification <strong>of</strong> rete testis invasion (RTI) (15.9%); lymphovascular<br />

invasion (LVI) (9.5%); and spermatic cord invasion (SCI) (5.0%).<br />

RTI was more frequently under-reported (14.4%), and LVI over-reported<br />

(8.0%) on initial assessment. Disparity <strong>of</strong> tumor size in 2 cases (1.0%) was<br />

<strong>of</strong> no clinical significance. For NSGCT (n�150) the commonest discrepancies<br />

were in RTI (9.3%), LVI (7.3%) and SCI (6.7%), with LVI typically<br />

over-reported (4.0%) and RTI and SCI under-reported (6.7%, 5.3%<br />

respectively) on primary analysis. Central review resulted in change <strong>of</strong><br />

primary tumor stage in 45 cases (12.1%). 12 (6.0%) and 16 (8.0%)<br />

seminomas, and 12 (8.0%) and 5 (3.3%) NSGCTs were upstaged and<br />

downstaged respectively. Conclusions: Central pathology review results in<br />

frequent alteration <strong>of</strong> factors predictive <strong>of</strong> relapse in stage IA/B testicular<br />

cancer. <strong>Part</strong>icular attention should be directed to review <strong>of</strong> RTI, LVI and<br />

SCI to ensure accurate prognostication.<br />

Genitourinary Cancer<br />

301s<br />

4598 General Poster Session (Board #4E), Sun, 8:00 AM-12:00 PM<br />

Metastatic malignant transformation <strong>of</strong> teratoma to primitive neuroectodermal<br />

tumor (PNET): Results with PNET-based chemotherapy. Presenting<br />

Author: Amit Jain, Indiana University Melvin and Bren Simon Cancer<br />

Center, Indianapolis, IN<br />

Background: Metastatic germ cell cancers are highly chemosensitive and<br />

have 80% cure rate with cisplatin based chemotherapy. Post-chemotherapy<br />

teratoma can usually be surgically resected. However, teratoma,<br />

which is pleuripotent tissue, can undergo malignant transformation along<br />

mesodermal elements to PNET. Unlike teratoma, PNET can metastasize<br />

and render a patient unresectable and incurable. We report the results <strong>of</strong><br />

treatment <strong>of</strong> patients with PNET with cyclophosphamide � doxorubicin �<br />

vincristine (CAV) alternating with ifosfamide � etoposide (IE). Methods: We<br />

reviewed 81 patients with histologically confirmed PNET transformed from<br />

testicular teratoma at Indiana University from 1998 to 2011. We identified<br />

13 cases who were treated with chemotherapy comprising cyclophosphamide<br />

1000-1200 mg/m2 , doxorubicin 50-75 mg/m2 , and vincristine 2 mg<br />

alternating with ifosfamide 1.8 grams/m2 plus etoposide 100 mg/m2 for 5<br />

days. Treatment was given every 3 weeks with a maximum <strong>of</strong> 6 cycles or<br />

until progression or undue toxicity. Hematopoeitic growth factors were<br />

usually incorporated. The remaining 68 patients underwent surgical<br />

resection. Results: Ten patients had unresectable disease and 3 were<br />

treated in an adjuvant setting. Median age was 31 years (range 20-53). Ten<br />

<strong>of</strong> 13 patients had received prior platinum based chemotherapy. Eight <strong>of</strong><br />

10 evaluable patients achieved objective response. Five <strong>of</strong> those were<br />

rendered with no evidence <strong>of</strong> disease(NED) with further surgery. Although 4<br />

<strong>of</strong> the 5 patients subsequently relapsed, 1 patient remains alive with NED<br />

at 69 months. The 3 patients who received adjuvant treatment after<br />

retroperitoneal lymph node dissection (RPLND) are alive with NED at 1, 4<br />

and 8 years. Conclusions: CAV and IE alternating chemotherapy has high<br />

objective response rate for PNET transformed from teratoma and results in<br />

occasional long term disease free survival when combined with subsequent<br />

resection. We recommend adjuvant CAV alternating with IE chemotherapy<br />

for patients with PNET after RPLND due to the high probability <strong>of</strong> recurrent<br />

disease and their high chemosensitivity to this regimen.<br />

4600 General Poster Session (Board #4G), Sun, 8:00 AM-12:00 PM<br />

Quality <strong>of</strong> life and late toxicities in germ-cell cancer patients after<br />

high-dose chemotherapy. Presenting Author: Thomas Wuendisch, University<br />

Hospital Marburg, Marburg, Germany<br />

Background: The number <strong>of</strong> long term survivors after treatment for relapsed/<br />

refractory germ cell cancer (GCC) is increasing but little is known about<br />

quality <strong>of</strong> life (QOL) and late toxicities (LT). Methods: We assessed LT and<br />

QOL in GCC patients (pts), treated in a prospective, randomized, multicenter,<br />

phase III trial comparing single versus sequential high-dose<br />

chemotherapy (HDCT) (Lorch 2007). All 216 pts were asked to complete<br />

the European Organization <strong>of</strong> Research and Treatment <strong>of</strong> Cancer Quality <strong>of</strong><br />

Life Questionnaire (EORTC QLQ-C30) prior to HDCT, 6 weeks after and<br />

yearly thereafter. Results were analyzed according to standard methods<br />

(Fayers 2001). In addition pts were contacted at a median <strong>of</strong> 76 months<br />

(range 45-109) after HDCT to obtain information about current social and<br />

pr<strong>of</strong>essional activities as well as LT. Results: Among 216 pts, 92/216<br />

(42%) were alive and without evidence <strong>of</strong> disease one year after randomization.<br />

Median age <strong>of</strong> pts was 34 years (range 15-56). A median <strong>of</strong> 4<br />

conventional-dose cisplatin-based treatment cycles had been given prior to<br />

HDCT. Questionnaires and response to the survey were evaluable in 86/92<br />

(93%) pts overall and in 59/86 (69%) pts more than 3 years after HDCT.<br />

Values for global health status, role functioning and emotional functioning<br />

declined during the first year after HDCT, increased in the following years,<br />

but did not reach the reference values from large samples <strong>of</strong> the general<br />

population in Norway (Hjermstad 1998). Pts after sequential HDCT scored<br />

better in a number <strong>of</strong> items (e.g. emotional-, cognitive-, social-functioning,<br />

fatigue and dyspnoea) in comparison to single HDCT including high-dose<br />

cyclophosphamide. Persisting polyneuropathy was reported in 59%, ototoxicity<br />

or tinnitus in 56%, erectile dysfunction in 24%, hypertension in 23%,<br />

hypercholesterinemia in 17%, diabetes mellitus in 6%, nephrotoxicity in<br />

6%, myocardial infraction in 2% and second cancers in 1% <strong>of</strong> pts. Overall<br />

82 % <strong>of</strong> pts were employed, 30% exercised regularly. Conclusions: HDCT<br />

has a negative impact on QOL, including social and pr<strong>of</strong>essional life. Most<br />

common late toxicities were ototoxicity and polyneuropathy. Sequential<br />

HDCT without cyclophosphamide seems to result in a better outcome in<br />

respect to QOL.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


302s Genitourinary Cancer<br />

4601 General Poster Session (Board #4H), Sun, 8:00 AM-12:00 PM<br />

Residual tumor resections (RTR) in patients with germ cell tumors (GCT)<br />

after salvage chemotherapy. Presenting Author: Christian Winter, University<br />

<strong>of</strong> Duesseldorf, Duesseldorf, Germany<br />

Background: Residual tumor resections (RTR) after salvage chemotherapy<br />

are <strong>of</strong> utmost importance due to a much higher rate <strong>of</strong> vital carcinoma in<br />

the residual tumor. A complete RTR may lead to an improved long term<br />

survival <strong>of</strong> such patients (pts). Methods: A retrospective analysis was<br />

performed in 197 pts who underwent 209 RTRs in 2 referral centers<br />

(2003-2011) with identical surgical technique (2 surgeons). The RTR<br />

database was queried for pts who received salvage chemotherapy (conventional<br />

salvage chemotherapy (CCT) or high-dose salvage chemotherapy<br />

(HDCT)) and a subsequent RTR. Results: Sixty (29%) <strong>of</strong> all RTRs were<br />

performed after salvage chemotherapy. In 31 pts (52%) and 29 pts (48%)<br />

a CCT and HDCT was used as salvage regimen, respectively.Vital cancer<br />

was found in 48% and 45% <strong>of</strong> pts with CCT and HDCT. Respectively, vital<br />

cancer consisted <strong>of</strong> germ cell cancer and malignant transformed teratoma<br />

in 61% and 39% <strong>of</strong> the cases. In contrast, the percentage <strong>of</strong> vital cancer in<br />

pts after 1st-line treatment was 30% (p� 0.0365).This comparatively high<br />

rate is due to a 55% rate <strong>of</strong> intermediate/poor prognosis pts in the whole<br />

group <strong>of</strong> 209 RTRs. To date we have evaluated the oncological outcome in<br />

171 <strong>of</strong> 197 pts with a median follow-up <strong>of</strong> 23 months (1-227). In 142 pts<br />

no relapse was observed, whereas in 29 pts a GCT relapse occured (17%), 7<br />

(4.6%) <strong>of</strong> all evaluated pts had an “in-field”-relapse. 7 pts died due to<br />

tumor progression. The relapse rate in pts with RTR after salvage regime<br />

was significantly higher compared to RTR pts after primary chemotherapy<br />

(37% vs. 13%, p� 0.0008). The „in-field“-relapse rate in pts with RTR<br />

after salvage was only in 3.9%, 2 pts died due to tumor progression.<br />

Conclusions: Every second pts with RTR after salvage chemotherapy<br />

showed vital cancer in the residual tumor. The relapse rate in pts with RTR<br />

after salvage chemotherapy was significantly higher compared to RTR pts<br />

after first-line chemotherapy (p�0.0008) but the “infield” relapse rate was<br />

similar. A complete resection <strong>of</strong> all residual lesions after salvage chemotherapy<br />

is necessary to potentially improve the long-term disease-free<br />

survival. This is especially true for pts with malignant transformed teratoma<br />

and restricted options for repeated salvage chemotherapy.<br />

4603 General Poster Session (Board #5B), Sun, 8:00 AM-12:00 PM<br />

Phase I results <strong>of</strong> a phase I/II trial <strong>of</strong> BNC105P with everolimus in<br />

metastatic renal cell carcinoma (mRCC) patients previously treated with<br />

VEGFR tyrosine kinase inhibitors. Presenting Author: Thomas E. Hutson,<br />

US Oncology Research, LLC, McKesson Specialty Health, The Woodlands,<br />

TX, and Baylor Sammons Cancer Center-Texas Oncology PA, Dallas, TX<br />

Background: BNC105P is an investigational agent that destabilizes tubulin<br />

polymers leading to selective damage <strong>of</strong> tumor vasculature, causing<br />

disruption <strong>of</strong> blood flow to tumors, hypoxia and associated tumor necrosis.<br />

BNC105P also has a direct anti-proliferative action on cancer cells.<br />

Preclinical investigations have demonstrated that BNC105P is effective at<br />

selectively damaging the vasculature in both primary and metastatic<br />

lesions. Up regulation <strong>of</strong> the mTOR pathway has been identified as a<br />

survival response by the tumor to hypoxic insult. It follows that the<br />

combined use <strong>of</strong> BNC105P with an agent active against mTOR may<br />

improve clinical outcome in patients with progressive mRCC who are<br />

refractory to VEGFR-directed tyrosine kinase inhibitors (TKI). Methods: A<br />

phase I/II study in mRCC patients who have received 1-2 prior TKIs was<br />

undertaken. Using a classic 3�3 design, the phase I component <strong>of</strong> this<br />

study enrolled 12 subjects at 4 dose levels <strong>of</strong> BNC105P (4.2, 8.4, 12.6,<br />

16 mg/m2 ; IV infusion Days 1 and 8, 21-day repeating cycle). Everolimus<br />

was administered concurrently (10 mg p.o.). PK analysis was performed<br />

during Cycle 1. Results: The phase I component has been completed. The<br />

BNC105P / everolimus combination was well tolerated. No DLTs (drugrelated,<br />

during cycle 1) were observed in any <strong>of</strong> the phase I subjects.<br />

Toxicities on study deemed to be drug-related (either single agent or<br />

combination) included single grade 3 events <strong>of</strong> anemia and pericardial<br />

effusion. Grade 2 events (more than 1 occurrence) <strong>of</strong> fatigue, anemia and<br />

oral mucositis were also observed. Seven phase I subjects achieved at least<br />

disease stabilization with a minimum time on therapy <strong>of</strong> 18 weeks (6<br />

cycles). Across all subjects a median <strong>of</strong> 6 cycles (range: 1-15) was<br />

administered. PK analysis confirmed no drug-drug interaction. The randomized<br />

phase II component <strong>of</strong> the study continues and will compare<br />

everolimus given in combination with BNC105P to a sequential approach<br />

(everolimus followed by BNC105P). Conclusions: The MTD <strong>of</strong> BNC105P<br />

(16 mg/m2 ) can be combined with full dose everolimus and is being<br />

evaluated in the randomized phase II study.<br />

4602 General Poster Session (Board #5A), Sun, 8:00 AM-12:00 PM<br />

Can abdominal ultrasound replace CT scanning in long-term follow-up <strong>of</strong><br />

male patients with germ cell tumor? Presenting Author: Arthur Gerl,<br />

Oncology Practice, Ludwig Maximilians University Munich (LMU), Munich,<br />

Germany<br />

Background: To date, there is no international consensus on how best to<br />

follow patients (pts) with GCT after their initial management. In the<br />

absence <strong>of</strong> a generally accepted follow-up schedule the possible benefits <strong>of</strong><br />

regular CT scanning must be weighed against their cost, potential contrast<br />

media reactions and the long term risk <strong>of</strong> cumulative X-ray exposure. The<br />

present study focuses on the role <strong>of</strong> abdominal ultrasound in the follow-up<br />

<strong>of</strong> males with GCT and raises the question whether CT-scanning may be<br />

replaced by abdominal ultrasound. Methods: This retrospective singlecenter<br />

cohort study included 887 GCT pts followed between January 2001<br />

and November 2011. The follow-up schedule was predominately based on<br />

abdominal ultrasound performed by the same physician (A.G.). Patterns <strong>of</strong><br />

recurrence and the long-term outcome were analyzed. Results: 462 <strong>of</strong> 887<br />

pts (52.1%) had stage I, 258 (29.1%) stage II and 130 (14.7%) stage III<br />

disease (not evaluable in 37 pts). The median time between baseline and<br />

the most recent follow-up examination was 5.0 years. A total <strong>of</strong> 14.604<br />

abdominal ultrasound examinations (16.5/pt.), 1170 CT scans (1.32/pt.),<br />

and 956 chest X-rays (1.08/pt) were performed. A relapse occurred in 58<br />

pts (6.5%) with 11 <strong>of</strong> 58 pts experiencing multiple relapses. 34 <strong>of</strong> 58<br />

relapses (58.6%) were detected in pts with stage I GCT. The sites <strong>of</strong> relapse<br />

included the abdomen (n�42), other sites (n�10), and marker elevation<br />

only (n�10). 33 <strong>of</strong> 42 abdominal relapses (78.6%) were detected by<br />

abdominal ultrasound. The median size <strong>of</strong> abdominal lymph nodes was 25<br />

mm (range, 6-67mm). After a median follow-up <strong>of</strong> 5 years the GCT<br />

specific survival <strong>of</strong> the entire cohort was 98.4%. Regarding the subgroup <strong>of</strong><br />

pts with relapse the GCT specific survival was 94.7%. Conclusions:<br />

Ultrasound appears to be an appropriate method to detect abdominal<br />

recurrences in pts with GCT. However, training and cumulative experience<br />

is necessary to detect retroperitoneal recurrences at an early stage. The<br />

number <strong>of</strong> expensive and potentially harmful CT scans may be markedly<br />

decreased by abdominal ultrasound.<br />

4604 General Poster Session (Board #5C), Sun, 8:00 AM-12:00 PM<br />

Genetic determinants <strong>of</strong> long-term response to rapalog therapy in advanced<br />

renal cell carcinoma (RCC). Presenting Author: Martin Henner Voss,<br />

Memorial Sloan-Kettering Cancer Center, New York, NY<br />

Background: Temsirolimus (T) and everolimus (E) are rapalog inhibitors <strong>of</strong><br />

the mammalian target <strong>of</strong> rapamycin (mTOR) with efficacy in advanced RCC<br />

[NEJM;356(22):227-81; Lancet;372(9637):449-56]. With reported median<br />

progression-free survival (PFS) <strong>of</strong> 5.5 and 4.9 months (mo), respectively,<br />

benefit is typically modest, yet a subset <strong>of</strong> patients (pts) achieves<br />

long-term disease control evident by extended PFS. The oncogenomic<br />

background for this is unclear. Methods: We analyzed frozen specimens <strong>of</strong><br />

tumor and adjacent normal kidney from pts with advanced RCC and<br />

documented long-term response to T or E, defined as PFS <strong>of</strong> � 20mo.<br />

Samples from pts with PFS � 2mo served as comparators. Specimens were<br />

subjected to whole exome sequencing; a targeted next-generation sequencing<br />

platform was used for deep sequencing and investigation <strong>of</strong> copy<br />

number variations (CNV) in 230 genes <strong>of</strong> interest. Results: In 5 pts with<br />

long-term response to T [n�3] or E [n�2], histologic subtypes were clear<br />

cell [n�3] and unclassified [n�2] RCC. Median number <strong>of</strong> prior treatments<br />

was 2 (1-3). Two pts remain on therapy, 3 have stopped drug for disease<br />

progression; treatment duration was 20, 25�, 27, 28, and 28� mo. Three<br />

control pts progressed after 1, 2, and 2 mo on therapy. Mean target<br />

coverage was 86x for whole exome and 443x for targeted sequencing. Three<br />

<strong>of</strong> 5 long-term responders harbored acquired somatic mutations and/or<br />

CNV in genes encoding for members <strong>of</strong> the Phosphoinositide 3-Kinase<br />

(PI3K) / mTOR pathway. Effects on amino acid sequence and gene function<br />

suggest a hyperactive pathway in all 3. For the other 2 long-term responders<br />

genomic changes with possible indirect link to the pathway, but no<br />

apparent determinants <strong>of</strong> treatment response were seen. In 3 control<br />

patients with rapid disease progression no genomic alterations suggesting<br />

hyperactivation <strong>of</strong> the pathway were seen. Conclusions: In this retrospective<br />

discovery set <strong>of</strong> RCC pts with unusually long response to rapalog therapy,<br />

next generation sequencing using pre-treatment tissue specimens revealed<br />

plausible oncogenomic determinants <strong>of</strong> treatment benefit in 3 <strong>of</strong> 5 cases.<br />

These findings provide basis for further biomarker development and studies<br />

in a larger sample set are ongoing.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


4605 General Poster Session (Board #5D), Sun, 8:00 AM-12:00 PM<br />

Correlation <strong>of</strong> chromosome (Chr) 14 loss and 5q gain with outcomes <strong>of</strong><br />

pazopanib treatment in patients (pts) with metastatic clear cell renal cell<br />

carcinoma (mRCC). Presenting Author: Gary R. Hudes, Fox Chase Cancer<br />

Center, Philadelphia, PA<br />

Background: The identification <strong>of</strong> molecular prognostic and/or predictive<br />

determinants <strong>of</strong> outcome in pts with mRCC is an important challenge. We<br />

hypothesized that specific tumor DNA copy number alterations (CNAs) -<br />

loss <strong>of</strong> chr 14 or 14q (14/14q-), and gain <strong>of</strong> chr 5q (5q�) may predict<br />

likelihood <strong>of</strong> pazopanib treatment benefit in pts with mRCC. Methods: Pts<br />

DNA samples from the Phase II (VEG102616) pazopanib trial were<br />

analyzed by using Affymetrix OncoScan. Copy no. data were moving<br />

smoothed and normalized. The copy no. and allele difference were pr<strong>of</strong>iled<br />

according to their chromosome locations to interrogate CNA and LOH.<br />

Objective response (OR, RECIST) and progression free survival (PFS) were<br />

determined by investigators (IN) and an independent review (IR) committee.<br />

OR and PFS data were analyzed using exact and Kaplan-Meier tests.<br />

Results: Tumor DNA samples from 75 pts were adequate for CNA analysis.<br />

14/14q- was found in 35/75 pts (46.7%), and 5q� was found in 37/75<br />

(49.3%). 14/14q- was present in the tumors <strong>of</strong> 12/31 pts (39%) with OR<br />

by IR compared with 23/44 (52%) nonresponding pts (p�0.347). 14/14qdid<br />

not affect PFS. 5q� was present in tumors <strong>of</strong> 17/31 pts (55%) with OR<br />

and 20/44 (45%) nonresponding pts (p�0.486). PFS was longer in pts<br />

with 5q� tumors compared with those without 5q� (log rank p�0.026),<br />

with median PFS <strong>of</strong> 66 vs 35 wks, respectively. The odds <strong>of</strong> achieving OR<br />

decreased as the total number <strong>of</strong> chromosomal gains/losses increased<br />

(p�0.032, odds ratio 0.49), but this had no effect on PFS. In exploratory<br />

analysis, we examined the combined effect <strong>of</strong> both 14/14q- and 5q� on<br />

PFS (Table). Conclusions: Pts with 5q� tumors have significantly longer<br />

PFS, with no effect on OR rate. While 14/14q- alone had no effect on<br />

outcomes, the combination <strong>of</strong> 5q� and no 14/14q- was associated with<br />

significantly greater PFS. Pts with more genetically complex tumors were<br />

less likely to obtain OR with pazopanib.<br />

Median PFS wks (95% confidence limits)<br />

Copy no. change No. pts<br />

By IN By IR<br />

14/14q- , no 5q� 14 18 (8, 52) 28 (18, -)<br />

No 14/14q-, no 5q� 24 36 (20, 60) 44 (20, 84)<br />

14/14q-, 5q� 21 36 (24, -) 60 (28, -)<br />

No 14/14q-, 5q� 16 83 (36, 100) 84 (43, -)<br />

Total 75 p�0.005 p�0.071<br />

4607 General Poster Session (Board #5F), Sun, 8:00 AM-12:00 PM<br />

Evolving trends in non-clear cell renal cell cancer (RCC) epidemiology: A<br />

large registry analysis <strong>of</strong> 4,483 patients. Presenting Author: Ashok P. Pai,<br />

University <strong>of</strong> California, Davis, Sacramento, CA<br />

Background: Non-clear cell (ncc) RCC is uncommon but includes a<br />

heterogeneous group <strong>of</strong> histologic subtypes such as papillary, chromophobe<br />

(Chr), medullary (Med), and collecting duct (CD) cancers. We used a<br />

large cancer registry to define nccRCC clinical features and outcomes,<br />

identify prognostic variables, and to generate hypotheses for further study.<br />

Methods: Invasive RCC tumors in the California Cancer Registry from<br />

1998-2009 among adults � 18 years <strong>of</strong> age (n�38,251) were analyzed.<br />

Baseline clinical and tumor variables were collected. Primary outcome<br />

measures were 3-year cause-specific (CSS) and overall survival (OS). Uniand<br />

multivariate survival analysis were used to identify predictors <strong>of</strong> CSS<br />

and OS. Results: Of 38,251 RCC cases, 19,149 (50%) were <strong>of</strong> clear cell<br />

type; 14,619 (38.2%) were “unclassified.” Thus, 4,483 (11.7%) nccRCC<br />

cases were identified and included in this analysis. Of these, 3,304<br />

(73.7%) were diagnosed in 2004-09, suggesting a shift to more precise<br />

coding <strong>of</strong> histologic subtypes starting in the early 2000s. Histology<br />

distribution (n, %): papillary - 2,863 (63.9%); Chr - 1,507 (33.6%); and<br />

other including Med and CD - 113 (2.5%). Variables associated with<br />

significantly better OS and CSS (univariate analysis) were Chr histology,<br />

female sex, and higher socioeconomic status (SES). Significantly worse OS<br />

and CSS were seen in Med�CD, age � 65 yrs, no nephrectomy (Nx), and<br />

higher stage. Non-hispanic blacks had significantly worse OS and CSS,<br />

while the “targeted therapy era” (2004-2009) was associated with better<br />

OS. Multivariate analysis showed the following to be independently<br />

associated with outcomes (all p �0.001; Hazard Ratios [HR] shown for<br />

CSS and OS): Chr histology (0.48, 0.56), Med�CD histology (2.99, 2.42),<br />

no Nx (2.84, 3.18), regional stage (5.84, 1.98), distant stage (25.7,<br />

7.67); and non-hispanic blacks (1.5,2 p�0.006; 1.25, p�0.03). Age �<br />

65 yrs (HR 1.78, p�0.001) and high SES (HR 0.88, p�0.001) were<br />

associated with OS but not CSS. Conclusions: This is among the largest<br />

registry analyses <strong>of</strong> nccRCC ever performed, showing emerging trends in<br />

this uncommon RCC subset. <strong>Clinical</strong> variables associated with CSS and OS<br />

were identified that can inform the design <strong>of</strong> future clinical trials in<br />

nccRCC.<br />

Genitourinary Cancer<br />

303s<br />

4606 General Poster Session (Board #5E), Sun, 8:00 AM-12:00 PM<br />

Safety and efficacy <strong>of</strong> AMG 386 in combination with sunitinib in patients<br />

with metastatic renal cell carcinoma (mRCC) in an open-label multicenter<br />

phase II study. Presenting Author: Michael B. Atkins, Beth Israel Deaconess<br />

Medical Center, Boston, MA<br />

Background: AMG 386, an investigational peptide-Fc fusion protein,<br />

inhibits angiogenesis by disrupting the angiopoietin/Tie2 axis. We evaluated<br />

the safety and efficacy <strong>of</strong> AMG 386 plus sunitinib in patients (pts) with<br />

mRCC. Methods: Adults with mRCC who were naïve to angiogenesis<br />

inhibitors were sequentially enrolled to 2 cohorts: sunitinib 50 mg PO QD<br />

(4 wks on, 2 wks <strong>of</strong>f) plus AMG 386 at 10 mg/kg (A) or 15 mg/kg (B) IV QW.<br />

Primary endpoints: adverse events (AEs), dose interruptions/reductions due<br />

to AEs in the first 12 wks <strong>of</strong> treatment; secondary endpoints included:<br />

progression-free survival (PFS) and response rate (ORR). Results: 85 pts<br />

received �1 dose <strong>of</strong> study medication (A/B, n�43/42). In A/B, 88%/76%<br />

were male and 30%/36% were age �65; MSKCC risk scores were low<br />

(40%/36%) or intermediate (60%/62%). For A/B: median follow-up time<br />

was 19.6/12.0 mos; AMG 386 discontinuations due to AEs were 16%/<br />

29%; and grade �3 treatment-related AEs occurred in 72%/74% with<br />

virtually all attributed to sunitinib. Grade 3 AEs occurring with �5%<br />

frequency were hypertension, hand foot syndrome, asthenia, fatigue,<br />

elevated lipase, diarrhea, mucositis, vomiting, thrombocytopenia, and<br />

neutropenia, with no distinction between dose levels. The percentage <strong>of</strong> pts<br />

with sunitinib dose interruptions within the first 12 wks (A/B, 58%/57%)<br />

met the prespecified criteria. One pt in B had fatal acute pulmonary edema.<br />

No pt developed anti-AMG 386 antibodies. The Kaplan-Meier estimate<br />

(95% CI) <strong>of</strong> PFS was 13.9 (10.4, 19.2) mos in A; PFS in B is not yet mature<br />

with only 21% <strong>of</strong> pts having disease progression. ORR (95% CI) was 58%<br />

(42, 73) in A including 1 CR, and 59% (42, 74) in B. Conclusions: In pts<br />

with mRCC, AMG 386 at 10 and 15 mg/kg combined with sunitinib<br />

appeared to be tolerable. Reported sunitinib dose modifications for the<br />

observation period were within the prespecified range. Efficacy results<br />

suggest potential benefit for the addition <strong>of</strong> AMG 386 to sunitinib.<br />

4608 General Poster Session (Board #5G), Sun, 8:00 AM-12:00 PM<br />

Selecting renal cell carcinoma therapy: Ranking <strong>of</strong> patient perspective on<br />

toxicities. Presenting Author: Michael K.K. Wong, Norris Comprehensive<br />

Cancer Center, University <strong>of</strong> Southern California, Los Angeles, CA<br />

Background: Oral therapies (angiogenesis inhibitors and mammalian target<br />

<strong>of</strong> rapamycin (mTOR) inhibitors) for renal cell carcinoma (RCC) have<br />

demonstrated significant improvements in progression-free survival but<br />

also possess toxicities. The objective <strong>of</strong> this study was to evaluate whether<br />

different toxicities <strong>of</strong> oral RCC therapies had equal importance to patients.<br />

Methods: US adults from the Kidney Cancer Association with a self-reported<br />

diagnosis <strong>of</strong> RCC completed a web-enabled survey. Respondents were<br />

asked to select the 3 most and 3 least troublesome toxicities from a list <strong>of</strong><br />

20 common RCC therapy toxicities. For each respondent, a value <strong>of</strong> 1 was<br />

assigned to each <strong>of</strong> the 3 most troublesome toxicities, a value <strong>of</strong> -1 was<br />

assigned to each <strong>of</strong> the 3 least troublesome toxicities, and a value <strong>of</strong> 0 was<br />

assigned to the remaining toxicities. A straight count method was applied to<br />

estimate the mean relative importance <strong>of</strong> each toxicity. Respondents also<br />

answered 10 treatment-choice questions, each <strong>of</strong> which included a pair <strong>of</strong><br />

hypothetical RCC medication pr<strong>of</strong>iles described by survival, toxicities, and<br />

serious adverse events. Four toxicities including fatigue, mouth sores,<br />

hand-foot syndrome, and stomach problems were included in both exercises.<br />

Results: 264 <strong>of</strong> the 272 respondents completed the entire ranking<br />

exercise. Among the 20 toxicities, stomach problems was the most<br />

troublesome and was assigned an importance <strong>of</strong> 10. Changes in hair color<br />

was the least troublesome and was assigned an importance <strong>of</strong> 0. Patients<br />

ranked fatigue (8.2), mouth sores (7.7), hand-foot syndrome (6.6) by order<br />

<strong>of</strong> importance. When given choices among eliminating severe toxicities,<br />

fatigue was as important as stomach problems, both fatigue and stomach<br />

problems were more import than mouth sores, and mouth sores was more<br />

important than hand-foot syndrome; although not statistically significant.<br />

Conclusions: This statistical approach <strong>of</strong>fers insight into those toxicities<br />

important to patients on chronic RCC therapies.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


304s Genitourinary Cancer<br />

4609 General Poster Session (Board #5H), Sun, 8:00 AM-12:00 PM<br />

Post-axitinib systemic therapy in metastatic renal cell carcinoma (mRCC).<br />

Presenting Author: Housam Haddad, Cleveland Clinic Taussig Cancer<br />

Institute, Cleveland, OH<br />

Background: Axitinib is a potent and selective inhibitor <strong>of</strong> the VEGF receptor<br />

family with efficacy as a second-line therapy in mRCC. Evaluation <strong>of</strong> further<br />

systemic therapy following axitinib has not been previously reported.<br />

Methods: The medical records <strong>of</strong>mRCCpatients (pts) treated at The Cleveland<br />

Clinic or MD Anderson Cancer Center who received axitinib were<br />

retrospectively reviewed. Patient characteristics including best response<br />

and duration <strong>of</strong> treatment on axitinib and each subsequent line <strong>of</strong> therapy<br />

were recorded. Results: Eighty-one patients were initially identified; 29<br />

patients received at least one approved targeted agent following axitinib<br />

and are included in this analysis. The remainder <strong>of</strong> pts were excluded due<br />

to ongoing axitinib (n�16), lack <strong>of</strong> subsequent therapy (n�13), lost to<br />

follow up (n�11), received experimental agents post-axitinib (n�7) or were<br />

inevaluable (n�2). Three pts were excluded due to � 21 days on axitinib.<br />

Pt characteristics at start <strong>of</strong> axitinib included 76% male, median age 59<br />

(range, 44-78); 97% clear cell; 93% prior nephrectomy; 64% previouslytreated.<br />

Pts were favorable (39%) or intermediate (60%) risk based on<br />

Heng criteria. Overall response rate to axitinib was 52% and median<br />

duration <strong>of</strong> treatment <strong>of</strong> 11.2 months (range, 1.1-90). Thirty eight percent<br />

<strong>of</strong> patients had new brain, bone, and/or liver metastases at disease<br />

progression on axitinib. For the first subsequent therapy post axitinib<br />

(n�25), pts were treated with tyrosine kinase inhibitors (72%) or mTOR<br />

inhibitors (28%). Overall 8% <strong>of</strong> pts achieved partial responses (one pt each<br />

to sunitinib and pazopanib) and 52% had a best response <strong>of</strong> stable disease.<br />

Estimated median duration <strong>of</strong> therapy was 4.4 months (range, 0.2-27.5�).<br />

Fifteen pts received a second post-axitinib targeted therapy with mTOR<br />

inhibitors (60%) or TKIs (40%). One patient (7%) achieved a partial<br />

response to pazopanib and 8 patients (53%) had a best response <strong>of</strong> SD.<br />

Estimated median treatment duration was 4.8 months (range, 0.7-19.1�).<br />

Conclusions: There are objective responses and stable disease to postaxitinib<br />

targeted therapies, consistent with previous series and trials in<br />

refractory RCC patients.<br />

4611 General Poster Session (Board #6B), Sun, 8:00 AM-12:00 PM<br />

Outcome <strong>of</strong> rapid disease progression in the treatment break following<br />

cytoreductive nephrectomy (CN) after presurgical sunitinib in patients with<br />

primary metastatic renal cell carcinoma (RCC). Presenting Author: Axel<br />

Bex, The Netherlands Cancer Institute, Amsterdam, Netherlands<br />

Background: There is concern that interruption <strong>of</strong> tyrosine kinase inhibitors<br />

(TKI) triggers disease progression (PD) and metastasis. This is based on<br />

preclinical models and observation <strong>of</strong> rapid PD in the postsurgical treatment<br />

break in patients pretreated with TKI. Little is known about the<br />

frequency <strong>of</strong> PD during postsurgical TKI interruption and its outcome.<br />

These data may have implications for neoadjuvant or presurgical treatment<br />

concepts. Methods: Of 66 patients from two closed phase II trials<br />

investigating 2-3 months presurgical sunitinib for primary metastatic<br />

clear-cell RCC, 45 were evaluated in this retrospective analysis because<br />

they had absence <strong>of</strong> PD prior to planned surgery and underwent CN (35<br />

[78%] MSKCC intermediate and 10 [22%] poor risk). Patients had CT<br />

scans at the end <strong>of</strong> pretreatment and at 4 weeks post-nephrectomy before<br />

restarting sunitinib. In patients with postsurgical RECIST PD at 4 weeks<br />

when compared to the preoperative CT scan overall survival (OS) was<br />

measured from the time <strong>of</strong> nephrectomy and compared to patients without<br />

treatment break PD. Results: Median OS <strong>of</strong> 45 patients from the time <strong>of</strong><br />

surgery was 22 months (13.0-31.5 months). Overall 14 patients progressed<br />

during treatment break (31%), with new sites <strong>of</strong> disease in 5/14<br />

(35%). Reintroduction <strong>of</strong> sunitinib resulted in disease stabilisation or<br />

better in 13. Patients with poor risk were represented in the progressors<br />

(n�4[28%]) and non-progressors (n�6 [19%]) (p�0.7 fishers exact test).<br />

The hazard ratio (HR) for death associated with PD during treatment break<br />

was 1.90 (95% CI 0.89-4.08). OS for non-progressors was 25 months<br />

(15-NA) and 13 months (6-27) for progressors. Conclusions: A significant<br />

proportion <strong>of</strong> patients develop PD upon sunitinib withdrawal in a 4 week<br />

recovery period following CN. In view <strong>of</strong> the small sample size these data<br />

suggest a strong trend toward treatment break PD being associated with a<br />

poor outcome. It is not clear whether this PD is related to interruption <strong>of</strong><br />

TKI, surgery or a combination <strong>of</strong> factors. The EORTC SURTIME trial<br />

investigates treatment break PD after immediate CN and CN after sunitinib<br />

and may answer this question.<br />

4610 General Poster Session (Board #6A), Sun, 8:00 AM-12:00 PM<br />

Incidence and severity <strong>of</strong> cardiotoxicity in metastatic renal cell carcinoma<br />

(RCC) patients treated with targeted therapies. Presenting Author: Philip S.<br />

Hall, Stanford University, Internal Medicine Residency Program, Stanford,<br />

CA<br />

Background: Almost all patients with advanced RCC are treated with<br />

therapies targeting the hypoxia inducible factor axis. The potential for these<br />

agents to cause cardiovascular (CV) toxicity has been increasingly recognized<br />

but the overall incidence and extent have not been well described. We<br />

sought to identify and characterize the incidence and severity <strong>of</strong> CV toxicity<br />

among RCC patients treated with targeted therapies. Methods: Between<br />

2004 and 2011, all RCC patients treated with targeted therapies directed<br />

against the VEGF or mTOR axes were identified at our institution. The<br />

incidence <strong>of</strong> hypertension, left ventricular (LV) dysfunction, changes in<br />

serum markers <strong>of</strong> CV toxicity (e.g., troponin I, NT-proBNP) and heart failure<br />

were assessed and graded according to the Common Terminology Criteria<br />

for Adverse Events (CTCAE v.4.0). Results: Cardiovascular toxicity developed<br />

in 116 <strong>of</strong> 159 patients (73%). Excluding hypertension, 52 <strong>of</strong> 159<br />

(33%) developed cardiovascular toxicity. Cardiac toxicity ranged from<br />

asymptomatic drops in LV ejection fraction (LVEF) to rises in NT-proBNP to<br />

severe heart failure. Asymptomatic cardiotoxicity as defined by decrease in<br />

LVEF or increase in NT-proBNP occurred in 43 patients (27%). Symptomatic<br />

heart failure (grade 2-3) and grade 3-4 decrease in LVEF each occurred<br />

in 4%. Sunitinib was the most frequently used and most common <strong>of</strong>fending<br />

agent, with 66 <strong>of</strong> 101 sunitinib-treated patients (65%) developing a form<br />

<strong>of</strong> CV toxicity, or 32 <strong>of</strong> 101 (32%) excluding hypertension. However, it was<br />

notable that CV toxicity was observed in 68%, 66%, and 51% <strong>of</strong> patients<br />

treated with bevacizumab, sorafenib, and pazopanib as well. The mTOR<br />

inhibitors elicited significantly less CV toxicity, but sample sizes were<br />

small. Conclusions: Cardiovascular toxicity is an important adverse event<br />

related to targeted therapy administration. Close monitoring for the<br />

development <strong>of</strong> CV toxicity with the use <strong>of</strong> these agents should become<br />

standard <strong>of</strong> care as early detection <strong>of</strong> asymptomatic patients could preempt<br />

symptomatic toxicity and reduce treatment-related morbidity and mortality.<br />

4612 General Poster Session (Board #6C), Sun, 8:00 AM-12:00 PM<br />

Overall and progression-free survival with everolimus, temsirolimus, or<br />

sorafenib as second targeted therapies for metastatic renal cell carcinoma:<br />

A retrospective U.S. chart review. Presenting Author: Hongbo Yang,<br />

Analysis Group, Inc., Boston, MA<br />

Background: Tyrosine kinase inhibitors (TKIs) (sorafenib, sunitinib, pazopanib)<br />

and inhibitors <strong>of</strong> the mammalian target <strong>of</strong> rapamycin (mTORs)<br />

(everolimus, temsirolimus) are used for treating metastatic renal cell<br />

carcinoma (mRCC) following initial treatment with a TKI. This study’s<br />

objective was to establish the effectiveness <strong>of</strong> the most commonly used 2nd line treatments based on real-use data. Methods: mRCC patients who<br />

received a TKI as their 1st targeted therapy and everolimus, temsirolimus or<br />

sorafenib as their 2nd therapy were identified by oncologists (85% in<br />

community practice) throughout the US as part <strong>of</strong> an online chart review<br />

study. Baseline characteristics were assessed prior to 2nd targeted therapy,<br />

including type and duration <strong>of</strong> initial TKI, response, histological subtype,<br />

performance status, and sites <strong>of</strong> metastasis. Overall survival (OS) and<br />

progression free-survival (PFS) were assessed after initiating 2nd targeted<br />

therapy. OS and PFS were compared between treatment groups using Cox<br />

proportional hazards models adjusted for baseline characteristics. Results:<br />

Charts were reviewed for 233, 178, and 123 mRCC patients receiving<br />

everolimus, temsirolimus, and sorafenib after an initial TKI, respectively.<br />

Median age was 64 years and 70.4% were male. Prior to initiating a 2nd targeted therapy, 86.0% used sunitinib and 85.9% had disease progression.<br />

After adjusting for baseline characteristics, everolimus was associated<br />

with significantly prolonged OS compared to temsirolimus (hazard<br />

ratio [HR] 0.56; CI 0.40-0.78; p�0.001) and sorafenib (HR 0.65; CI<br />

0.42-0.99; p�0.047). Everolimus was associated with significantly longer<br />

PFS compared to temsirolimus (HR 0.73; CI 0.55-0.96; p�0.025), and<br />

non-statistically significant longer PFS compared to sorafenib (HR 0.75; CI<br />

0.53-1.07; p�0.110). Results were similar in the subgroup with sunitinib<br />

as 1st targeted therapy and the subgroup with progression on 1st TKI.<br />

Conclusions: Among mRCC patients with prior TKI treatment, everolimus<br />

was associated with significantly prolonged OS and PFS compared to<br />

temsirolimus and significantly prolonged OS compared to sorafenib.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


4613 General Poster Session (Board #6D), Sun, 8:00 AM-12:00 PM<br />

Xp11.2 translocations in adult renal cell carcinomas with clear cell and<br />

papillary features. Presenting Author: Susanne M. Chan, Department <strong>of</strong><br />

Anatomical Pathology, Sunnybrook Health Sciences Centre, University <strong>of</strong><br />

Toronto, Toronto, ON, Canada<br />

Background: Xp11.2 translocation renal cell carcinoma (RCC) is a rare<br />

tumor with unpredictable clinical course and prognosis in adults. Recognition<br />

<strong>of</strong> these tumors depends on the identification <strong>of</strong> a RCC with unique<br />

histology, particularly clear cell and papillary (CCP) features. Our objectives<br />

were to determine the incidence <strong>of</strong> Xp11.2 translocations in adult<br />

RCCs with clear cell and papillary features, and to characterize the<br />

clinicopathological features and prognosis <strong>of</strong> adult Xp11.2 RCCs. Methods:<br />

Slides for 1047 RCCs in adults (1999-2009) were retrieved from multiple<br />

institutions. Cases were reviewed histologically in order to detect any<br />

degree <strong>of</strong> clear cell and papillary change. Tissue microarrays were constructed<br />

from the clear cell and papillary RCCs as well as 40 non-papillary<br />

clear cell, 5 papillary, 3 chromophobe and 2 unclassified RCCs. Immunohistochemistry<br />

using TFE3, a marker highly sensitive (97.5%) and specific<br />

(99.6%) for Xp11.2 translocations was performed. Four pathologists<br />

independently reviewed the TFE3 results. <strong>Clinical</strong> and pathologic data were<br />

also retrieved. Results: Out <strong>of</strong> 1047 RCCs, 140 (13%) exhibited clear cell<br />

and papillary features. Four out <strong>of</strong> these 140 (3%) were positive for TFE3.<br />

All <strong>of</strong> the non-papillary clear cell, papillary, chromophobe and unclassified<br />

RCCs were negative for TFE3. Mean follow up for TFE3� RCCs was 55<br />

months. Conclusions: Xp11.2 translocation RCCs diagnosed by TFE3<br />

immunohistochemistry were identified in 3% <strong>of</strong> adult RCCs that had clear<br />

cell and papillary changes. These tumors appear to present with smaller<br />

tumour size, lower stage and better prognosis in comparison to non-Xp11.2<br />

CCPRCC and clear cell RCCs. In addition to Xp11.2 translocation carcinoma,<br />

coexistence <strong>of</strong> clear cell and papillary features may be present in<br />

other subsets <strong>of</strong> tumors that have yet to be characterized.<br />

Comparison <strong>of</strong> clinicopathological features and prognosis.<br />

TFE3� CCPRCC TFE3- CCPRCC Non-papillary clear<br />

(n�4)<br />

(n�136)<br />

cell RCC (n�40)<br />

Average age 54 62 60<br />

Average tumor size 4.1 6.0 5.2<br />

pT1 4 (100%) 80 (60%) 28 (70%)<br />

pT2 0 (0%) 17 (13%) 2 (5%)<br />

pT3 0 (0%) 35 (26%) 10 (25%)<br />

pT4 0 (0%) 1 (1%) 0 (0%)<br />

Local recurrence 0 (0%) 13 (10%) 9 (23%)<br />

Distant metastasis 0 (0%) 31 (23%) 5 (13%)<br />

Death due to RCC 0 (0%) 11 (8%) 4 (10%)<br />

4615 General Poster Session (Board #6F), Sun, 8:00 AM-12:00 PM<br />

Outcomes <strong>of</strong> patients (pts) with metastatic renal cell carcinoma (mRCC)<br />

treated with pazopanib after progression on other targeted therapies (TT): A<br />

single-institution experience. Presenting Author: Marc Ryan Matrana,<br />

University <strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: Pazopanib is an approved multi-tyrosine kinase inhibitor that<br />

prolongs progression-free survival (PFS) compared to placebo in treatmentnaive<br />

and cytokine-refractory mRCC. Outcomes and safety data on its use<br />

after TT are limited. Methods: We retrospectively reviewed pts with mRCC<br />

who received salvage pazopanib between 11/09-11/11. Kaplan-Meier<br />

method was used to estimate survival outcomes. PFS was calculated from<br />

start <strong>of</strong> pazopanib until progressive disease (PD) or death. Univariable and<br />

multivariable Cox proportional hazards models were fitted to evaluate<br />

associations <strong>of</strong> PFS with covariables. Results: 114 consecutive pts met<br />

inclusion criteria (median age 62.6 years, 66% males, 83% clear cell). All<br />

pts had PD after other TT (median # <strong>of</strong> prior TT 2, range 1-5; median time<br />

on prior TT 23.3 mos). 79% <strong>of</strong> pts had PD on sunitinib, 39% on sorafenib,<br />

19% on temsirolimus, 59% on everolimus, and 23% on bevacizumab.<br />

25% received prior chemotherapy and 16% received prior cytokines in<br />

addition to TT. 87% had prior nephrectomy. 11% had favorable-risk, 68%<br />

intermediate-risk, and 21% poor-risk per MSKCC criteria. 85 events (PD or<br />

death) occurred. Median OS was 17 mos (95% CI: 10.3-NA). Median PFS<br />

was 6.4 mos (95% CI: 4.5-9.5). By multivariable analysis, PFS was<br />

associated with male gender (HR�0.433, 95%CI: 0.269-0.696;<br />

p�0.0006), # <strong>of</strong> metastatic sites (HR�1.252; 95%CI: 1.04-1.503;<br />

p�0.016), hypertension exacerbation (HR�0.378; CI: 0.175-0.813;<br />

p�0.0128) and PS 2� vs.0-1 (HR�2.067; CI: 1.243-3.437; p�0.0052).<br />

58% discontinued pazopanib due to PD, 12% died <strong>of</strong> PD on treatment, and<br />

11% discontinued pazopanib due to adverse events (AEs), mostly GI<br />

complaints or fatigue. There were no treatment related deaths. Common<br />

AEs included: fatigue (44%), diarrhea (29%), nausea/vomiting (15%),<br />

anorexia (14%), hypertension exacerbation (11%), hypothyroidism (11%),<br />

hand-foot skin reaction (9%), and increase LFTs (4%). 86% <strong>of</strong> AEs were<br />

grade 1/2. Conclusions: In this retrospective study, pazopanib demonstrated<br />

efficacy in mRCC following PD with other TT. AEs were mild/<br />

moderate and manageable.<br />

Genitourinary Cancer<br />

305s<br />

4614 General Poster Session (Board #6E), Sun, 8:00 AM-12:00 PM<br />

A phase I study <strong>of</strong> pazopanib (P) combined with bevacizumab (B) in<br />

patients with metastatic renal cell carcinoma (mRCC) or other advanced<br />

refractory tumors. Presenting Author: Sylvie Negrier, Léon-Bérard Cancer<br />

Centre, Lyon, France<br />

Background: Since previous experiments <strong>of</strong> B with VEGFR tyrosine kinase<br />

inhibitors showed overlapping and limiting toxicities, a dose-finding study<br />

was designed to explore the safety and feasibility <strong>of</strong> the combination <strong>of</strong> a<br />

new VEGFR inhibitor P with B, in mRCC treatment-naive patients (pts) or in<br />

pts with other advanced refractory solid tumors. Methods: This bicentric<br />

trial is conducted with 3�3�3 escalation doses <strong>of</strong> P � B. The MTD is the<br />

highest dosage not expected to cause a dose limiting toxicity (DLT) in more<br />

than 2/3, 3/6 or finally 3/9 pts, during the first 8 weeks <strong>of</strong> treatment. The<br />

effect <strong>of</strong> B on steady-state pharmacokinetic (PK) <strong>of</strong> P is also investigated.<br />

Major inclusion criteria are: ECOG PS �1, ALT and AST ��2.5 x ULN,<br />

serum creatinine ��1.5 mg/dL or creatinine clearance ��50 mL/mn and<br />

absence <strong>of</strong> uncontrolled hypertension. Results: 15 pts were enrolled with<br />

mRCC (n�7), melanoma (n�2), adrenocortical carcinoma (n�1), mesothelioma<br />

(n�1), pancreatic cancer (n�1), oesophageal cancer (n�1), bladder<br />

cancer (n�1) and seminoma (n�1). Median age is 61 (43-78), 12 pts are<br />

male. No DLT were reported at DL1 (P 400 mg/d � B 7.5 mg/kg q2w) (n �<br />

9), or at the first step <strong>of</strong> DL2 (P 600 mg/d � B 7.5 mg/kg q2) (n�3), but the<br />

3 following nephrectomized pts experienced DLT: a grade 3 transaminitis,<br />

a grade 3 pulmonary embolism and a reversible microangiopathic hemolytic<br />

anemia. Inclusion <strong>of</strong> nephrectomized pts was completed, but enrolment<br />

<strong>of</strong> 3 additional non-nephrectomized pts at DL2 was approved by<br />

IDSMB in November 2011 and these treatments are ongoing. Preliminary<br />

results <strong>of</strong> PK analysis showed a significantly higher P AUC at steady-state in<br />

pts with DLT. Moreover, lower P apparent clearance was observed in<br />

nephrectomized pts. Conclusions: The MTD <strong>of</strong> the combination <strong>of</strong> P and B is<br />

400 mg/d and 7.5mg/kg respectively, in nephrectomized patients, but is<br />

not yet reached in other patients. Unexpected effect <strong>of</strong> nephrectomy status<br />

was observed on P pharmacokinetics.<br />

4617 General Poster Session (Board #6H), Sun, 8:00 AM-12:00 PM<br />

Circulating mir-21 and mir-378 are predictive <strong>of</strong> progression-free survival<br />

(PFS) in patients treated with everolimus in metastatic renal cell cancer<br />

(mRCC). Presenting Author: James Lin Chen, The University <strong>of</strong> Chicago,<br />

Chicago, IL<br />

Background: The oral mTOR inhibitor, everolimus, affects tumor growth by<br />

targeting cellular metabolic proliferation pathways and modestly delays<br />

RCC progression. We hypothesized that circulating microRNAs, which have<br />

been associated with renal cancer and inflammation, may serve as<br />

predictive biomarkers to help better define a population more sensitive to<br />

treatment. Methods: Plasma from mRCC pts refractory to VEGF inhibition<br />

were obtained prior to treatment with standard dose everolimus as part <strong>of</strong> a<br />

clinical trial examining FDG-PET as a potential predictive biomarker. As we<br />

were specifically interested in tumor response to drug, only pts who died,<br />

remained on trial, or had radiographic progression by RECIST criteria were<br />

pr<strong>of</strong>iled. Pts who were unable to tolerate drug were excluded. MicroRNAs<br />

were extracted and pr<strong>of</strong>iled without pre-amplification using Exiqon LNA<br />

PCR panels. Crossing point (Cp) values within 5 <strong>of</strong> the negative control were<br />

removed. MicroRNAs must have been present in �90% <strong>of</strong> samples and<br />

varied at least p � 0.10 from mean to be further analyzed. Cox-proportional<br />

hazards model and Kaplan-Meier analyses were performed. Results: 28<br />

patients had available plasma and met criteria for pr<strong>of</strong>iling. Pt characteristics<br />

included: 20 (71%) clear cell histology, median age 57.7 (43 – 76),<br />

median number <strong>of</strong> prior systemic therapies 2 (1 – 3). 103 microRNAs were<br />

expressed in at least 90% <strong>of</strong> all samples. Mir-21 and mir-378 were<br />

independently correlated with PFS (FDR: 0.02 and 0.06, respectively).<br />

Low circulating plasma mir-21 and mir-378 levels resulted in a median<br />

PFS prolongation <strong>of</strong> 370d vs. 101d (p�0.027) and 368d vs. 106d<br />

(p�0.001). Analysis <strong>of</strong> the clear cell cohort for mir-21 and mir-378 also<br />

demonstrated a significant median PFS difference <strong>of</strong> 350d vs. 173d<br />

(p�0.045) and 345d vs. 147d (p�0.004). Conclusions: Elevated levels <strong>of</strong><br />

circulating mir-21 and mir-378 have been associated with systemic<br />

inflammatory states, and in our study are correlated with decreased PFS in<br />

mRCC pts undergoing everolimus therapy. Further prospective studies will<br />

be required to validate these exploratory results for their potential role as<br />

prognostic or predictive biomarkers.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


306s Genitourinary Cancer<br />

4618 General Poster Session (Board #7A), Sun, 8:00 AM-12:00 PM<br />

Evaluation with DCE-US <strong>of</strong> antiangiogenic treatments in 539 patients<br />

allowing the selection <strong>of</strong> one surrogate marker correlated to overall survival.<br />

Presenting Author: Nathalie Lassau, Institut Gustave Roussy, Villejuif,<br />

France<br />

Background: A prospective study <strong>of</strong> dynamic contrast-enhanced ultrasound<br />

(DCE-US) with quantification for the evaluation <strong>of</strong> antiangiogenic treatments<br />

was launched (19 centers), supported by the French National<br />

Cancer Institute. The objectives were the diffusion <strong>of</strong> the standardized<br />

method, a cost evaluation and the identification <strong>of</strong> perfusion parameters<br />

predicting tumor response. Methods: All patients had DCE-US at baseline,<br />

D7, D14, D30, D60 and every two months. Each examination included a<br />

bolus injection <strong>of</strong> sonovue (Bracco) and 3 minutes <strong>of</strong> raw linear data with an<br />

Aplio (Toshiba). Raw data were analyzed with a mathematical model<br />

(patent PCT/IB2006/003742) to evaluate 7 parameters characterizing the<br />

tumor perfusion curve. Response to treatment was evaluated every 2<br />

months with RECIST criteria. In order to have sufficient follow-up data, the<br />

statistical analysis has to be performed more than 6 months after the<br />

inclusion <strong>of</strong> the last analyzed patient. Inclusions were closed in March<br />

2010. Results: A total <strong>of</strong> 539 patients have been included (mainly RCC<br />

(157) and HCC (107)); more than 2 000 DCE-US and 1700 CT-scan were<br />

performed. A follow-up more than 12 months showed that 3 parameters<br />

have a strong significant difference (p�0.0003) according to the response<br />

at 6 months. The decrease <strong>of</strong> more than 40% <strong>of</strong> AUC at one month is<br />

correlated to the TTP (p� 001) and OS (p� 0.04). Conclusions: Final<br />

results confirm the usefulness <strong>of</strong> this tool to monitor anti-angiogenic<br />

treatments. The criteria: the decrease <strong>of</strong> more than 40% <strong>of</strong> AUC at one<br />

month is predictive <strong>of</strong> response.<br />

4620 General Poster Session (Board #7C), Sun, 8:00 AM-12:00 PM<br />

Prospective exploratory analysis <strong>of</strong> the association between genetic polymorphisms<br />

and sunitinib toxicity and efficacy in metastatic renal-cell carcinoma.<br />

Presenting Author: Carlos Alberto Farfan, Hospital Universitario 12<br />

de Octubre, Madrid, Spain<br />

Background: Sunitinib (SU) is a multi-targeted receptor tyrosine kinase<br />

inhibitor treatment that is approved for metastatic renal cell carcinoma<br />

(mRCC). However, several patients either do not respond to treatment or<br />

they experience significant toxicity.Our study aims to find genetic markers<br />

<strong>of</strong> toxicity and efficacy using a commercially available DNA microarray<br />

genotyping system. Methods: 30 patients with newly diagnosed mRCC,<br />

from January 2010 to May 2011, were evaluated prospectively at Hospital<br />

12 de Octubre (Madrid, Spain). Pts received SU 50 mg/day, 4 wks on / 2<br />

wks <strong>of</strong>f treatment schedule. A total <strong>of</strong> 92 <strong>of</strong> single nucleotide polymorphisms<br />

(SNPs) in 34 genes in the pharmacokinetic and pharmacodynamic<br />

pathways <strong>of</strong> drugs were analyzed using Drug inCode pharmacogenetic<br />

service. SNPs in candidate genes, together with clinical characteristics<br />

were tested univariately for association with the number <strong>of</strong> days <strong>of</strong> SU<br />

treatment until the first reduction <strong>of</strong> dose, PFS and OS. Results: Complete<br />

analysis was possible in 25 pts. Pts with CYP1A2*1/*1.a low metabolizing<br />

genotype, had an increased risk <strong>of</strong> dose reductions due to toxicity compare<br />

to allele *1F (Median time to dose reduction : 2.33 months vs NR;<br />

p�0.006). Pts with CYP2C19*1/*1 , wild type genotype, had an increased<br />

risk <strong>of</strong> dose reductions due to toxicity vs. other genotypes (Median time to<br />

dose reduction: 2.8 vs 9.73 months; p�0.021). Catechol-O-methyltransferase<br />

(COMT) V158M polymorphisms, was associated with PFS and OS<br />

(Met/Met carriers median PFS and OS NR; Met/Val pts PFS� 15 months;<br />

OS�17.2 months and Val/Val pts PFS�3.3 months; OS� 4.4 months ;<br />

p�0.005 for PFS and p�0.003 for OS). Conclusions: This preliminary<br />

analysis suggests that CYP1A2 and CYP2C19 SNPs may be associated with<br />

toxicity in patients with RCC treated with SU. As CYP1A2 and CYP2C19<br />

activity could be affected by a variety <strong>of</strong> non-genetic factors, if these is<br />

confirmed, it could lead to the necessity <strong>of</strong> controlling toxic and dietary<br />

habits <strong>of</strong> pts treated with SU. SNPs associated with toxicity and survival in<br />

this preliminary analysis are being validated in an independent cohort <strong>of</strong><br />

RCC treated with SU (García-Donas J, et al. Lancet Oncol 2011).<br />

4619 General Poster Session (Board #7B), Sun, 8:00 AM-12:00 PM<br />

Use <strong>of</strong> bisphosphonates (Bis) combined with sunitinib (Su) to improve the<br />

response rate (RR), progression-free survival (PFS), and overall survival<br />

(OS) <strong>of</strong> patients (pts) with bone metastases (mets) from renal cell<br />

carcinoma (RCC). Presenting Author: Avivit Peer, Rambam Health Care<br />

Campus, Haifa, Israel<br />

Background: Bis are used to prevent skeletal events <strong>of</strong> bone mets, and may<br />

exhibit anti tumor effects. We aimed to evaluate whether Bis can bring a<br />

RR, PFS, and OS benefit to pts with bone mets from RCC that are treated<br />

with Su. Methods: We performed an international multicenter retrospective<br />

study <strong>of</strong> pts with bone mets from RCC who were treated with Su. Pts were<br />

divided into Bis users (group 1) and nonusers (group 2). The effect <strong>of</strong> Bis on<br />

RR, PFS and OS, was tested with adjustment for known prognostic factors<br />

using a chisquare test from contingency table and partial likelihood test<br />

from Cox regression model. Results: Between 2004-2011, 244 pts with<br />

metastatic RCC were treated with Su. 92 pts had bone mets, 41 group 1<br />

and 51 group 2. The groups were balanced regarding the following known<br />

prognostic factors: past nephrectomy, clear cell vs non clear cell histology,<br />

initial diagnosis to sunitinib treatment (tx) time, presence <strong>of</strong> � 2 mets<br />

sites, presence <strong>of</strong> lung/liver mets, ECOG performance status, anemia,<br />

calcium level � 10 mg/dL, elevated alkaline phosphatase (AP), pre-tx<br />

neutrophil to lymphocyte ratio (NLR) �3, sunitinib induced HTN, and the<br />

use <strong>of</strong> angiotensin system inhibitors. They were also balanced with regard<br />

to past cytokines/targeted tx, and mean sunitinib dose/cycle. Objective<br />

response was partial response/stable disease 85% (n�35) vs 71% (n�36),<br />

and progressive disease 15% (n�6) vs 29% (n�15) (OR 3.287, p�0.07)<br />

in group 1 vs 2 respectively. Median PFS was 15 vs 5 months (HR 0.433,<br />

p�0.035), and median OS not reached with a median folloup time <strong>of</strong> 43<br />

mos vs 12 months (HR 0.398, p�0.003), in favor <strong>of</strong> group 1. In<br />

multivariate analysis <strong>of</strong> the entire pt cohort (n�92), factors associated with<br />

PFS were Bis use (HR 0.433, p�0.035), pre-tx NLR �3 (HR 0.405,<br />

p�0.016), and elevated AP (HR�3.63, p�0.012). Factors associated<br />

with OS were Bis use (HR 0.32, p�0.003), elevated AP (HR 3.18,<br />

p�0.002), and Su induced HTN (HR 0.193, p� 0.001). Conclusions: Bis<br />

may improve the outcome <strong>of</strong> Su tx in RCC with bone mets. This should be<br />

investigated prospectively, and if validated applied in clinical practice and<br />

clinical trials.<br />

4621 General Poster Session (Board #7D), Sun, 8:00 AM-12:00 PM<br />

Prognostic role <strong>of</strong> body composition parameters in renal cell carcinoma<br />

(RCC). Presenting Author: Emilie Lanoy, Biostatistics and Epidemiology<br />

Unit, Institut Gustave Roussy, Villejuif, France<br />

Background: Previous studies have shown that body component i.e. muscle<br />

tissue (MT) and adipose tissue (AT) are linked to overall survival (OS) and<br />

progression free survival (PFS). The aim <strong>of</strong> our study is to analyze whether<br />

MT and AT have a prognostic role in metastatic RCC treated with targeted<br />

therapy. Methods: We investigated body mass index (BMI), MT and AT in<br />

RCC pts. Analysis <strong>of</strong> CT image was used to evaluate cross-sectional areas<br />

(cm2 ) <strong>of</strong> total AT (TAT), MT, and the grey level image (GLI) <strong>of</strong> MT, reflecting<br />

physical properties <strong>of</strong> the scanned tissue and used as a proxy to describe<br />

the quality <strong>of</strong> muscle. The 3rd lumbar vertebra (L3) was chosen as a<br />

landmark since L3 and whole-body measurements are linearly related.<br />

Images were analyzed using Slice-O-Matic s<strong>of</strong>tware V4.3 (Tomovision).<br />

Indexed on height MT (cm2 /m2 ), indexed on height TAT (cm2 /m2 ) and<br />

mean GLI <strong>of</strong> MT were computed and described stratified on sex. For each<br />

measurement, population was divided in two groups: patients with values �<br />

or �� median observed in patient <strong>of</strong> the same gender and OS and PFS were<br />

estimated using Kaplan-Meier method and compared with the log-rank<br />

test. Multivariable Cox proportional hazards model were adjusted for<br />

modified MSKCC risk group and treatment (active versus placebo). Results:<br />

There were 113 men aged <strong>of</strong> 60 (interquartile range�52-56) years with<br />

BMI <strong>of</strong> 26 (24-29) kg2 /m2 and 36 women aged <strong>of</strong> 58 (54-65) years with<br />

BMI <strong>of</strong> 23 (20-26) kg2 /m2 . After adjustment for MSKCC (OS and PFS) and<br />

active treatment (PFS), GLI <strong>of</strong> MT over the median was associated with<br />

longer OS (HR�1.85, 95%CI�[1.22;2.82], p�.004) and PFS (HR�1.81,<br />

95%CI�[1.22;2.65], p�.002) Conclusions: Quality <strong>of</strong> muscular tissue is<br />

independently associated with better outcome in RCC. Surprisingly, adipose<br />

tissue as well as BMI is not associated with survival in this study.<br />

OS in months PFS in months<br />

Label<br />

Median P25 P75 p Median P25 P75 p<br />

Indexed MT>� 14 cm 2 /m 2 (men)<br />

or 13 cm 2 /m 2 24 14 49 .1213 6 4 13 .3128<br />

(women)<br />

Indexed MT< 14 cm 2 /m 2 (men)<br />

or 13 cm 2 /m 2 19 10 39 5 3 9<br />

(women)<br />

Mean GLI >� 38 (men) or 36 (women) 29 18 62 .0011 8 4 15 .0007<br />

Mean GLI < 38 (men) or 36 (women) 14 7 30 4 3 8<br />

Indexed TAT >�326 cm 2 /m 2 (men)<br />

or 231 cm 2 /m 2 24 14 45 .1725 6 4 11 .6124<br />

(women)<br />

Indexed TAT


4622 General Poster Session (Board #7E), Sun, 8:00 AM-12:00 PM<br />

Adherence to oral anticancer drugs (OAD) in patients (pts) with metastatic<br />

renal cancer (mRCC): First results <strong>of</strong> the prospective observational multicenter<br />

IPSOC study (Investigating Patient Satisfaction with Oral Anticancer<br />

Treatment). Presenting Author: Pascal Wolter, Department <strong>of</strong><br />

General Medical Oncology, University Hospitals Leuven, Leuven, Belgium<br />

Background: Patient adherence to oral therapy is a critical issue in cancer<br />

treatment. The aim <strong>of</strong> this study is to investigate the prevalence and<br />

severity <strong>of</strong> non-adherence to OAD in mRCC and to identify factors<br />

predictive <strong>of</strong> non-adherence. Methods: Prospective observational multicenter<br />

trial performed at 11 Belgian academic and non-academic centers.<br />

All pts with mRCC starting OADs (sunitinib, pazopanib, everolimus or<br />

sorafenib) are eligible for the study. Pts are contacted by phone at baseline<br />

and at 1, 3, 6 and 12 months. At each contact, pts are asked to complete<br />

questionnaires investigating 1) medication adherence (MMAS), 2) patient<br />

satisfaction with treatment (CTSQ) and with treatment education (PS-<br />

CaTE), 3) the extent <strong>of</strong> information desire (EID), 4) quality <strong>of</strong> life (FACT-G<br />

and FKSI) and 5) the role <strong>of</strong> the pharmacist (SWiP). Adherence is measured<br />

using an electronic medication event monitoring system (MEMS, Aardex).<br />

Results: Between 02/2011 and 11/2011, 49 pts (m: 33, f: 16) with a<br />

median age <strong>of</strong> 63 years (range 25 - 87) have participated in the IPSOC<br />

study. Twenty-nine pts (64%) were treated with an OAD in first-line, 15 pts<br />

(33%) in second-line. With a median follow-up <strong>of</strong> 131 days (range 2 - 313)<br />

45 pts (92%) claimed to be fully adherent to their treatment (based on<br />

MMAS and CTSQ data). Four patients indicated to have missed at least one<br />

dose, <strong>of</strong> whom two indicated they occasionally forgot their medication and<br />

two others interrupted treatment because <strong>of</strong> side effects. Based on MEMS<br />

data, mean adherence, defined as the percentage <strong>of</strong> days with at least the<br />

prescribed number <strong>of</strong> dosage taken, was 98.91%. Conclusions: The IPSOC<br />

study, the first to examine adherence to OAD among mRCC pts, shows that<br />

mRCC pts are almost fully adherent to treatment recommendations. This<br />

seems to be in contrast to adherence data for other, long-lasting, anticancer<br />

treatments. Further investigations will focus on the question<br />

whether extensive counseling and participation in side-effect programs<br />

contribute to the high percentage <strong>of</strong> adherence in this study.<br />

4624 General Poster Session (Board #7G), Sun, 8:00 AM-12:00 PM<br />

Molecular characterization <strong>of</strong> SETD2, a histone methyltransferase, in clear<br />

cell renal cell carcinoma (ccRCC). Presenting Author: Thai Huu Ho,<br />

University <strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: Although the von Hippel-Lindau (VHL) tumor suppressor is<br />

mutated in 60% <strong>of</strong> ccRCC, deletion <strong>of</strong> VHL in mice is insufficient for<br />

tumorigenesis. Sequencing <strong>of</strong> ccRCC tumors identified mutations in<br />

SETD2, a histone H3 lysine 36 (H3K36) trimethyltransferase. We hypothesize<br />

that loss <strong>of</strong> SETD2 methyltransferase activity decreases H3K36<br />

trimethylation (H3K36Me3) in ccRCC, and contributes to the cancer<br />

phenotype. Methods: H3K36Me3 immunohistochemical (IHC) staining was<br />

quantitated in wild-type and mutant SETD2 nephrectomy tissue. To<br />

establish frequency <strong>of</strong> SETD2 loss <strong>of</strong> heterozygosity (LOH), genomic DNA<br />

was isolated from 51 VHL deficient ccRCC specimens and analyzed with<br />

Affymetrix single-nucleotide polymorphism (SNP) arrays. To evaluate<br />

alterations <strong>of</strong> histone modifications in advanced ccRCC, H3K36Me3 IHC<br />

was quantitated on tissue microarrays (TMAs) representing 28 paired<br />

ccRCC specimens with unaffected kidney parenchyma and 40 metastases.<br />

To identify kidney-specific H3K36Me3 binding sites, chromatin immunoprecipitation<br />

(ChIP) sequencing was performed on nephrectomy specimens.<br />

Results: Nephrectomy specimens with SETD2 truncating mutations<br />

have decreased H3K36Me3 (�50%) compared to uninvolved kidney<br />

tissue. Using SNP arrays, LOH at chromosome 3p (location <strong>of</strong> VHL and<br />

SETD2 genes) was detected in �90% <strong>of</strong> the tested tumors. IHC reveals a<br />

27.4% and 52.0% decrease in H3K36Me3 positive nuclei in primary<br />

ccRCC and metastases, respectively, when compared to matched adjacent<br />

unaffected kidney tissue (p �0.0001). Using ChIP sequencing, 421 and<br />

1,718 genomic regions were upregulated in the tumor and unaffected<br />

kidney tissue. Conclusions: SETD2 truncating mutations in patient-derived<br />

tissue have decreased H3K36Me3 indicating that SETD2 is a nonredundant<br />

H3K36 methyltransferase. LOH <strong>of</strong> VHL and SETD2 occurs in<br />

�90% <strong>of</strong> tested ccRCC tumors. Genomic regions enriched for H3K36Me3<br />

binding are being validated in additional patient-derived tissues. H3K36Me3<br />

is decreased in primary ccRCC and even more in metastases, suggesting<br />

that SETD2 methyltransferase activity is progressively misregulated in<br />

RCC. Loss <strong>of</strong> SETD2 activity may cooperate with VHL silencing to promote<br />

tumorigenesis.<br />

Genitourinary Cancer<br />

307s<br />

4623 General Poster Session (Board #7F), Sun, 8:00 AM-12:00 PM<br />

Trends in the use <strong>of</strong> cytoreductive nephrectomy for metastatic renal cell<br />

carcinoma in the VEGFR tyrosine kinase inhibitor era. Presenting Author:<br />

Che-Kai Tsao, Division <strong>of</strong> Hematology and Medical Oncology, The Tisch<br />

Cancer Institute, Mount Sinai School <strong>of</strong> Medicine, New York, NY<br />

Background: Two large randomized trials (SWOG, EORTC) published in<br />

2001 established the role <strong>of</strong> cytoreductive nephrectomy (CyNx) for the<br />

treatment <strong>of</strong> metastatic renal cell carcinoma (mRCC) in the cytokine era. A<br />

paradigm shift occurred in 2005 with the approval <strong>of</strong> VEGFR tyrosine<br />

kinase inhibitors (TKIs). We hypothesized that uncertainty regarding the<br />

role <strong>of</strong> CyNx in the VEGFR TKI era has resulted in a change in practice<br />

patterns. Methods: Using the Surveillance, Epidemiology and End Results<br />

(SEER) registry, we identified 2780 patients with histologically-confirmed<br />

mRCC between 2000 and 2008 who underwent CyNx or no surgery.<br />

Patients were separated into pre- or VEGFR TKI-eras (2000-2005 vs.<br />

2006-2008). Differences in baseline characteristics between these patient<br />

groups were assessed and controlled for in a logistic regression analysis to<br />

determine the likelihood <strong>of</strong> undergoing CyNx. Results: Overall, 1202 <strong>of</strong><br />

2780 patients (43%) underwent CyNx. CyNx increased from 41% in 2000<br />

to 49% in 2005, and decreased to 35% in 2008, with a 20% decreased<br />

likelihood <strong>of</strong> undergoing CyNx in the VEGFR TKI era compared to the<br />

pre-VEGFR TKI era. Logistic regression analysis showed that tumor size,<br />

age, race, marital status and pre- versus post-2005 periods were independent<br />

predictors <strong>of</strong> CyNx (Table). Patients who were non-Caucasian, single,<br />

with primary tumor �3cm, or older were less likely to undergo CyNx.<br />

Conclusions: Use <strong>of</strong> CyNx increased after supporting level I evidence was<br />

published in 2001, and decreased after regulatory approval <strong>of</strong> VEGFR TKIs<br />

in 2005. Racial and demographic differences exist in the utilization <strong>of</strong><br />

CyNx. The results <strong>of</strong> pending randomized trials evaluating the role <strong>of</strong> CyNx<br />

in the TKI-era are awaited to optimize use <strong>of</strong> this modality and address<br />

potential disparities.<br />

Logistic regression model for use <strong>of</strong> CyNx in 2000-2008.<br />

Variable (reference) Odds ratio Std error p value<br />

Post-2005 (Pre-2005) 0.802 0.092 0.0161<br />

Primary tumor size >3cm (


308s Genitourinary Cancer<br />

4626 General Poster Session (Board #8A), Sun, 8:00 AM-12:00 PM<br />

Impact <strong>of</strong> maximum standardized uptake value (SUVmax) evaluated by<br />

18-fluoro-2-deoxy-d-glucose positron emission tomography/computed tomography<br />

(FDG PET/CT) on survival for patients with advanced renal cell<br />

carcinoma. Presenting Author: Noboru Nakaigawa, Department <strong>of</strong> Urology,<br />

Yokohama City University Graduate School <strong>of</strong> Medicine, Yokohama, Japan<br />

Background: In this era <strong>of</strong> molecular targeting therapy when various<br />

systematic treatments can be selected, prognostic biomarkers are required<br />

for the purpose <strong>of</strong> risk-directed therapy selection. Numerous reports <strong>of</strong><br />

various malignancies have revealed that 18-Fluoro-2-deoxy-D-glucose<br />

( 18F-FDG) accumulation, as evaluated by positron emission tomography,<br />

can be used to predict the prognosis <strong>of</strong> patients. The purpose <strong>of</strong> this study<br />

was to evaluate the impact <strong>of</strong> the maximum standardized uptake value<br />

(SUVmax) from 18-fluoro-2-deoxy-D-glucose positron emission tomography/<br />

computed tomography (FDG PET/CT) on survival for patients with advanced<br />

renal cell carcinoma (RCC). Methods: A total <strong>of</strong> 67 patients with advanced<br />

or metastatic RCC were enrolled in this study. The FDG uptake <strong>of</strong> all RCC<br />

lesions diagnosed by conventional CT was evaluated by FDG PET/CT. The<br />

impact <strong>of</strong> SUVmax on patient survival was analyzed prospectively. Results:<br />

The mean duration <strong>of</strong> observation was 461 days (range, 7-1229 days). The<br />

SUVmax before treatment <strong>of</strong> 67 patients ranged between undetectable<br />

level and 16.6 (mean 7.6�3.6). The patients with RCC tumors showing<br />

high SUVmax before treatment demonstrated poor prognosis (p�0.001<br />

hazard ratio 1.289, 95% CI 1.161-1.430). The median survival time <strong>of</strong> 36<br />

patients with RCC showing SUVmax less or 7.0 was 1229�991 days, that<br />

<strong>of</strong> 21 patients with RCC showing SUVmax between 7.0 and 12.0 was<br />

446�202 days, and that <strong>of</strong> 10 patients RCC showing SUVmax higher than<br />

12.0 was 95�43 days (�7.0 vs. 7.0� �12.0 p�0.0052, 7.0� �12.0 vs.<br />

12.0�:p�0.0169, log-rank test). SUVmax demonstrated a tendency to<br />

predict the survival compared with the Memorial Sloan-Kettering Cancer<br />

Center classification (p �0.015 vs 0.315, multivariate Cox analyses).<br />

Conclusions: The survival <strong>of</strong> patients with advanced RCC can be predicted<br />

by evaluating their SUVmax using FDG PET/CT. FDG PET/CT has potency as<br />

an “imaging biomarker” to provide helpful information for the clinical<br />

decision-making.<br />

4628 General Poster Session (Board #8C), Sun, 8:00 AM-12:00 PM<br />

Sorafenib treatment <strong>of</strong> Asian patients with advanced renal cell carcinoma<br />

(RCC) in daily practice: Subset analysis <strong>of</strong> the large non-interventional<br />

PREDICT study. Presenting Author: Dingwei Ye, Cancer Hospital, Fudan<br />

University, Shanghai, China<br />

Background: Previous studies with sorafenib in Asian patients with advanced<br />

RCC were relatively small and used strict entry criteria. Here we<br />

investigated safety and efficacy in a large subset <strong>of</strong> Asian patients in the<br />

prospective, non-interventional PREDICT study <strong>of</strong> sorafenib in routine<br />

practice (NCT 00895674). Methods: Patients were eligible based on a<br />

diagnosis <strong>of</strong> advanced RCC and the decision by the investigator to prescribe<br />

sorafenib under compliance <strong>of</strong> the local product label. Tumor status,<br />

patients’ performance and physician assessment <strong>of</strong> efficacy and tolerability<br />

were collected up to 12 months. Results: Between Jan 2007 and June<br />

2010, 1092 patients were enrolled in China (n�1033), South Korea<br />

(n�55), the Philippines (n�3) and Indonesia (n�1). In the efficacy<br />

population (n�909), baseline characteristics were: 71% male; 89% �70<br />

years old; 89% clear cell histology; 78% prior nephrectomy; 56% prior<br />

systemic therapy; 16% high MSKCC risk; 35% ECOG PS �2; 5% brain<br />

metastases. Overall, 19% <strong>of</strong> patients had �1 concomitant disease at<br />

baseline; the most frequent concomitant diseases were hypertension<br />

(14%) and diabetes (6%). Initial sorafenib dose was 800 mg daily in 97%<br />

<strong>of</strong> patients, <strong>of</strong> whom 91% were also receiving 800 mg daily as last dose.<br />

Median duration <strong>of</strong> sorafenib treatment was 9.7 months (range 0.2–24.1),<br />

and in clinically relevant subgroups was as follows: treatment-naïve, 9.7<br />

months; high MSKCC risk, 9.3 months; brain metastases, 8.4 months; age<br />

�70 years, 7.6 months; ECOG PS 2, 9.7 months; ECOG PS 3, 6.1 months.<br />

At last follow up, 63% <strong>of</strong> physicians reported good/very good efficacy and<br />

59% good/very good tolerability. Sorafenib was well tolerated; �2% <strong>of</strong> the<br />

safety population (n�1022) reported serious drug-related adverse events<br />

(SDRAEs) and only 3% discontinued due to DRAEs. In all, 35% <strong>of</strong> patients<br />

reported a DRAE, with the most frequent being hand-foot skin reaction<br />

(21%), diarrhea (7%), rash (7%), alopecia (5%), hypertension (3%).<br />

Conclusions: In this large subset <strong>of</strong> Asian patients with advanced RCC<br />

treated in daily practice settings, sorafenib was well tolerated and provided<br />

benefit, including in clinically relevant patient subgroups.<br />

4627 General Poster Session (Board #8B), Sun, 8:00 AM-12:00 PM<br />

Natural history <strong>of</strong> malignant bone disease in renal cancer: Final results <strong>of</strong><br />

an Italian bone metastases survey. Presenting Author: Daniele Santini,<br />

Dipartimento di Medicina Oncologica, Università Campus Bio-Medico di<br />

Roma, Rome, Italy<br />

Background: bone metastases (mts) are an emerging clinical problem in<br />

renal cancer patients related to survival increase. We report the final data <strong>of</strong><br />

largest survey never published in literature. Methods: 398 renal cancer<br />

patients (pts) with evidence <strong>of</strong> bone mts, all died at the moment <strong>of</strong> study<br />

inclusion, have been included. Clinico-pathological data, data on survival<br />

and Skeletal Related Events (SRE) data and skeletal related therapies have<br />

been collected and statistically analyzed. Results: 286 males/112 females;<br />

median age: 63 (16-87); pts with bone mts at the moment <strong>of</strong> renal cancer<br />

diagnosis: 31.4%; pts with single bone mts: 31.1%. Type: lytic 77%,<br />

mixed: 14.6%, blastic: 7.6 %. Sites: spine (65.8%), pelvis (38.4%), long<br />

bones (31.6%), other (18.8%). Median time to bone mts: 8 months<br />

(0-288) (all patients); 24 months (1-288) (pts without bone mts at<br />

diagnosis). Pts with at least 1 SRE: 71.1%. Types <strong>of</strong> SREs: pathologic<br />

fracture (12.6%), radiotherapy (61.8%), spinal compression (7.6%), bone<br />

surgery (14.8%), hypercalcaemia (3.2%). Median number <strong>of</strong> SRE for<br />

patient: 1 (0-4). Median time to first SRE: 2 (0-72), to second SRE: 4<br />

(0-113), to third SRE: 11 (1-108). Median survival after bone mts<br />

diagnosis: 12 (1-178). Median survival after first SRE: 10 (0-144). Median<br />

survival in pts with at least one SRE: 14 (1-178); median survival in pts<br />

without SREs: 9 (0-62). In according with MKSCC criteria median time to<br />

skeletal disease was in patients with good prognosis was 24 (0-288),<br />

intermediate was 5 (0-180) and poor prognosis was 0 (0-77). A total <strong>of</strong> 168<br />

pts received zoledronic acid until performance status worsening or death.<br />

162 pts have been analysed as control group. The median time to first SRE<br />

in the zoledronic treated pts was 3 mths (0-101) compared with 1 mth (0 -<br />

25) in the control group (p� 0.05). 5 cases <strong>of</strong> ONJ have been diagnosed.<br />

Conclusions: The present survey is the largest descriptive study concerning<br />

the natural history <strong>of</strong> bone disease in renal cancer patients. The effects <strong>of</strong><br />

biological therpies on bone met will be presented during the meeting.<br />

4629 General Poster Session (Board #8D), Sun, 8:00 AM-12:00 PM<br />

Overall survival (OS) in metastatic renal cell carcinoma (mRCC) sequentially<br />

treated with different targeted therapies (TTs): Results from a large<br />

cohort <strong>of</strong> patients. Presenting Author: Giuseppe Procopio, Fondazione<br />

IRCCS Istituto Nazionale dei Tumori, Milan, Italy<br />

Background: Targeted Therapies (TTs) have definitely improved survival in<br />

patients with mRCC. However the optimal therapeutic strategy is not<br />

shared. This study was performed to assess the overall survival (OS) in a<br />

consecutive series <strong>of</strong> mRCC patients receiving TTs. Methods: Characteristics<br />

and outcomes <strong>of</strong> 336 patients affected by mRCC receiving TTs were<br />

collected from the database <strong>of</strong> Istituto Nazionale Tumori <strong>of</strong> Milan. The<br />

main characteristics <strong>of</strong> patients were: ECOG PS 0/1/2 186 (55%)/131<br />

(39%)/19 (6 %); clear-cell histology 291 (87%); previous nephrectomy<br />

293 (87%). According to Motzer criteria 32% <strong>of</strong> patients showed low risk,<br />

48% intermediate risk and 20% poor. Overall, 167 (50%) patients<br />

received one TTs, while 116 (34%), 42 (13%) and 11 (3.3%) received 2, 3<br />

and 4 TTs, respectively. 245 (73%) patients received sorafenib (So), 212<br />

(63%) sunitinib (Su), 33 (10%) a bevacizumab regimen and 73 (22%)<br />

other TTs, including everolimus, temsirolimus and axitinib. The Kaplan<br />

Meier curves were used to describe the survival.The uni- and multi-variate<br />

analyses for OS were carried out by means <strong>of</strong> Cox proportional hazard<br />

regression analysis. Results: Ata median follow-up <strong>of</strong> 43 months, 199<br />

patients (57 %) had died. The median OS was 24 months (95%CI:<br />

20.0-27.0) and the 5-year OS was 24.6 % (95 %CI: 18.7-30.8). In<br />

univariate analyses, there were no significant differences in the hazard<br />

ratios (HR) for So followed by Su compared to Su followed by So<br />

(HRSU-SO / SO-SU � 1.16; 95%CI: 0.57-2.33) or compared with other<br />

therapies (HROther sequential th. / SO-SU � 1.21; 95%CI: 0.78-1.88;<br />

p�0.674).In the multivariate analysis, in terms <strong>of</strong> OS any statistical<br />

difference was reported as regards the sequence used (Su/So vs So/Su;<br />

p�0.05) or bevacizumab regimen as compared to Su and/or So used<br />

sequentially (p�0.05). In the uni and multivariate analysis ECOG PS,<br />

nephrectomy, Fuhrman grade and number <strong>of</strong> sites <strong>of</strong> disease are independent<br />

predictive factors <strong>of</strong> outcome (p� 0.01). Conclusions: These data<br />

suggest that TTs improve OS in mRCC without any statistical difference<br />

when using different sequences <strong>of</strong> TTs. No cross-resistance between<br />

several sequences <strong>of</strong> TTs was documented.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


4630 General Poster Session (Board #8E), Sun, 8:00 AM-12:00 PM<br />

Hypoxia-inducible factor (HIF) 1� and 2� as predictive markers <strong>of</strong> outcome<br />

to VEGFR tyrosine kinase inhibitors (TKI) in renal cell carcinoma (RCC).<br />

Presenting Author: M.I. Saez, Hospital Universitario Virgen de la Victoria,<br />

Malaga, Spain<br />

Background: Relevant biomarkers in RCC are needed to identify appropriate<br />

candidates for selected targeted therapies. Mutations in the von Hippel-<br />

Lindau (VHL) gene result in the accumulation <strong>of</strong> HIF and increased<br />

expression <strong>of</strong> proangiogenic factors, including VEGF. Methods: Metastatic<br />

clear RCC patients with available baseline tumor samples who received<br />

first-line oral VEGFR-TKI were included in this analysis. VHL mutation/<br />

hypermethylation status and HIF1� and HIF2� immunohistochemical<br />

staining were analyzed from paraffin-embedded tumors. Additionally, a<br />

panel <strong>of</strong> candidate VEGF and VEGFR2 genetic SNPs was determined from<br />

peripheral blood samples. HIF was scored as negative or positive based on<br />

staining intensity (0-10% and � 10%, respectively). Results were evaluated<br />

for associations with clinical outcome. Results: 80 patients were<br />

included: 71 evaluable for HIF expression, 63 for VHL status and 52 for<br />

SNPs. 73% received treatment with sunitinib and median follow-up was<br />

21.5 months. Unlike VHL status, HIF1 and HIF2 positive expression<br />

showed a significant correlation with PFS and OS. HIF1� was also<br />

predictive for response rate (RR). On multivariate analysis adjusting for<br />

other prognostic factors, HIF1� and HIF2� remained the most significant<br />

independent predictive factors for survival (adjusted HR 0.09, 95% CI<br />

0.03-0.28, p � 0.0001 and HR 0.13, 95% CI 0.04-0.37, p � 0.0001;<br />

respectively). We did not find any statistically significant differences based<br />

on the VEGF and VEGFR2 SNPs analyzed. Conclusions: HIF1� and HIF2�<br />

levels represent the most important independent predictive factors <strong>of</strong><br />

outcome for VEGFR-TKI therapy in metastatic RCC. These findings may<br />

contribute to optimize treatment with targeted agents.<br />

HIF1� HIF2�<br />

HIF1� � HIF1� - HIF2� � HIF2� -<br />

N (%) 38 (53.5) 33 (46.5) 46 (64.8) 25 (35.2)<br />

RR (%) 67 27 55 37<br />

OR (p value) 0.18 (p�0.001) 0.50 (p�0.20)<br />

Median PFS (months) 22.8 13.6 21.4 9.6<br />

HR (p value) 0.36 (p�0.0001) 0.61 (p�0.04)<br />

Median survival (months) 43.7 16.6 42.5 20.3<br />

HR (p value) 0.31 (p�0.0001) 0.52 (p�0.04)<br />

4632 General Poster Session (Board #8G), Sun, 8:00 AM-12:00 PM<br />

Updated safety results from EXIST-2: Everolimus therapy for angiomyolipoma<br />

(AML) associated with tuberous sclerosis complex (TSC) or sporadic<br />

lymphangioleiomyomatosis (sLAM). Presenting Author: John Bissler, Cincinnati<br />

Children’s Hospital Medical Center, Cincinnati, OH<br />

Background: EXIST-2 (NCT00790400) is a randomized, double-blind,<br />

placebo-controlled, phase 3 trial assessing the efficacy and safety <strong>of</strong><br />

everolimus, an oral mTOR inhibitor, for treating AML in patients with TSC<br />

or sLAM. We have previously reported that everolimus resulted in a<br />

significantly higher AML response rate vs placebo (41.8% vs 0%; 95% CI:<br />

23.5–58.4; p�0.0001) with a consistent safety pr<strong>of</strong>ile (Bissler et al. JAm<br />

Soc Nephrol. 22, 2011, Abstract LB-PO3159). Here we present a 90-day<br />

safety update. Methods: 118 eligible patients were randomized 2:1 to<br />

receive everolimus 10 mg daily (n�79) or placebo (n�39). The primary<br />

efficacy endpoint was AML response rate (proportion <strong>of</strong> patients with best<br />

overall AML response status <strong>of</strong> “response”). Original cut-<strong>of</strong>f date for data<br />

analysis was 30 Jun 2011. An updated analysis <strong>of</strong> the safety data for the<br />

safety set (all patients receiving �1 dose <strong>of</strong> double-blind study drug with a<br />

valid post-baseline assessment) to 14 Oct 2011 are presented here.<br />

Results: As <strong>of</strong> 14 Oct 2011, median treatment duration was 48.1 and 45.0<br />

weeks for everolimus and placebo arms, respectively. Discontinuations in<br />

the double-blind period were the same in the everolimus arm as the initial<br />

analysis, but had increased by 4 patients in the placebo arm since initial<br />

analysis (3 due to disease progression, 1 withdrew consent). The majority <strong>of</strong><br />

adverse events (AEs) continued to be grade 1 or 2; the incidence <strong>of</strong> serious<br />

AEs was slightly higher than initially reported, particularly in the placebo<br />

arm (everolimus 20.3%, placebo 23.1%). AE incidence leading to discontinuation<br />

was the same as initially reported (everolimus 3.8%, placebo<br />

10.3%). In the updated data, 3 additional everolimus patients required<br />

dose interruption or reduction due to AEs; dose reduction/interruption<br />

remained more common in the everolimus arm (51.9% vs. 20.5%).<br />

Conclusions: Overall, the 90-day updated safety data analysis from the<br />

EXIST-2 trial has not revealed any additional safety concerns. No other<br />

patients receiving everolimus withdrew for any reason, whereas 3 more<br />

patients receiving placebo withdrew due to disease progression.<br />

Genitourinary Cancer<br />

309s<br />

4631 General Poster Session (Board #8F), Sun, 8:00 AM-12:00 PM<br />

Use <strong>of</strong> early tumor shrinkage as a response to VEGF inhibitors as a predictor<br />

<strong>of</strong> progression-free survival (PFS) and overall survival (OS) in patients with<br />

metastatic renal cell carcinoma (mRCC). Presenting Author: Viktor Gruenwald,<br />

Department <strong>of</strong> Hematology, Hemostasis, Oncology and Stem Cell<br />

Transplantation, Hannover Medical School, Hannover, Germany<br />

Background: Several novel targeted agents significantly prolong overall<br />

survival (OS) in metastatic renal cell cancer (mRCC) patients (pts.).<br />

Translational research, however, has not identified any prospectively<br />

validated prognostic or predictive biomarkers. Recently, progression free<br />

survival (PFS), overall response rate (ORR) and early tumor shrinkage were<br />

proposed as putative predictors for clinical outcome. In this study we aim to<br />

explore the potential role <strong>of</strong> treatment induced tumor remission as a<br />

prognostic or predictive parameter in mRCC. Methods: In our analyses we<br />

investigated the putative prognostic role <strong>of</strong> best response according to<br />

RECIST 1.1 criteria (complete remission (CR), partial remission (PR),<br />

stable disease (SD), progressive disease (PD)) in first line Tyrosine-kinase<br />

inhibitor (TKI) treatment in n�83 pts. Responders were defined by<br />

achieving CR, PR or SD at any time during the course <strong>of</strong> treatment.<br />

Furthermore, we tested whether tumor shrinkage <strong>of</strong> 10% or more within 12<br />

weeks <strong>of</strong> treatment qualifies as a potential cut-<strong>of</strong>f-parameter to predict PFS<br />

or OS. Uni- and multivariate analyses were performed using Log-rang test,<br />

Kaplan-Meier-, and Cox-regression analysis. Results: Univariate analyses<br />

revealed that first-line treatment non-responders had a significantly shorter<br />

OS than responders (p �0.0001). Tumor shrinkage <strong>of</strong> �10% or �10%<br />

also correlated with an OS <strong>of</strong> 14.5 vs. 29.1 mo. (p�0.001) and a PFS <strong>of</strong><br />

3.0 vs. 11.5 mo. (p �0.001). In multivariate analyses tumor shrinkage <strong>of</strong><br />

�10% was tested with other common variables such as ECOG, MSKCCscore,<br />

histology, and metastatic sites and proved to be a significant<br />

independent prognostic (HR 0.361; 95% CI 0.156-0.833) and predictive<br />

(HR 0.306; 95% CI 0.152-0.612) parameter. Conclusions: Our results<br />

outline that sensitivity to first line treatment <strong>of</strong> mRCC is an important<br />

prognostic factor in mRCC. Hereby tumor shrinkage <strong>of</strong> �10% within 12<br />

weeks <strong>of</strong> treatment reveals a novel cut-<strong>of</strong>f-parameter, which showed to be a<br />

promising prognostic and predictive marker in mRCC. Further investigations<br />

are needed to validate this parameter.<br />

4633 General Poster Session (Board #8H), Sun, 8:00 AM-12:00 PM<br />

Use <strong>of</strong> miRNA pr<strong>of</strong>iling in metastatic renal cell carcinoma to reveal a tumor<br />

suppressor effect for mir-215. Presenting Author: George M. Yousef,<br />

Department <strong>of</strong> Laboratory Medicine and Keenan Research Centre, St.<br />

Michael’s Hospital, Toronto, ON, Canada<br />

Background: Renal cell carcinoma (RCC) is the most common neoplasm <strong>of</strong><br />

the adult kidney. Metastatic RCC is difficult to treat. The five-year survival<br />

rate for metastatic RCC is �10%. Recently, microRNAs (miRNAs) have<br />

been shown to have a role in cancer metastasis and potential as prognostic<br />

biomarkers in cancer. Methods: We performed a miRNA microarray to<br />

identify a miRNA signature characteristic <strong>of</strong> metastatic compared to<br />

primary RCC. Results were validated by quantitative real time PCR. Target<br />

prediction analysis and gene expression pr<strong>of</strong>iling identified many <strong>of</strong> the<br />

dysregulated miRNAs could target genes involved in tumor metastasis. The<br />

effect <strong>of</strong> miR-215 on cellular migration and invasion was shown in a RCC<br />

cell line model. Results: We identified 65 miRNAs that were significantly<br />

altered in metastatic when compared to primary RCC. Nine (14%) miRNAs<br />

had increased expression while 56 (86%) miRNAs showed decreased<br />

expression. miR-10b, miR-196a, and miR-27b were the most downregulated<br />

while miR-638, miR-1915, and miR-149* were the most upregulated.<br />

A non-supervised 2D-cluster analysis showed that a sub-group <strong>of</strong> the<br />

primary tumors clustered under the metastatic arm with a group <strong>of</strong> miRNAs<br />

that follow the same pattern <strong>of</strong> expression suggesting they have an<br />

inherited aggressive signature. We validated our results by examining the<br />

expressions <strong>of</strong> miR-10b, miR-126, miR-196a, miR-204, and miR-215, in<br />

two independent cohorts <strong>of</strong> patients. We also showed that overexpression <strong>of</strong><br />

miR-215 decreased cellular migration and invasion in a RCC cell line<br />

model. In addition, through gene expression pr<strong>of</strong>iling, we identified direct<br />

and indirect targets <strong>of</strong> miR-215 that can contribute to tumor metastasis.<br />

Conclusions: Our analysis showed that miRNAs are altered in metastatic<br />

RCC and can contribute to kidney cancer metastasis through different<br />

biological processes. Dysregulated miRNAs represent potential prognostic<br />

biomarkers and may have therapeutic applications in kidney cancer.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


310s Genitourinary Cancer<br />

4634 General Poster Session (Board #9A), Sun, 8:00 AM-12:00 PM<br />

Evaluation <strong>of</strong> selective inhibitors <strong>of</strong> nuclear export (SINE) CRM1 inhibitors<br />

for the treatment <strong>of</strong> renal cell carcinoma (RCC). Presenting Author: Hiromi<br />

Inoue, University <strong>of</strong> California, Davis, Davis, CA<br />

Background: For the ~30% <strong>of</strong> patients who present with RCC at the<br />

metastatic stage, multi-kinase inhibitors have been used with moderate<br />

success: progression-free survival remains at only one to two years, and<br />

thus it is imperative to discover novel therapeutic approaches for metastatic<br />

disease. We asked whether (1) SINE inhibitors <strong>of</strong> chromosome region<br />

maintenance protein 1 (CRM1) attenuate key cell cycle regulatory and<br />

apoptotic molecules and whether these compounds exert salutary effects in<br />

a human RCC xenograft mouse model. Methods: Four RCC cell lines (ACHN,<br />

Caki-1, 786-O, and A498) with distinct genotypes, and primary normal<br />

human kidney (NHK) cell lines, were used in this study. The cells were<br />

treated with the chemically related SINE CRM1 inhibitors KPT-185 or 251<br />

and MTT assays were performed. In addition, cell cycle analyses, immun<strong>of</strong>luorescence<br />

for p53 and p21, and immunoblotting for CRM1, p53, p21,<br />

p27, and p-MDM2 were performed for all cell lines. RCC mice with Caki-1<br />

xenografts were treated with vehicle, the orally-available CRM1 inhibitor<br />

KPT-251, or sorafenib for 26 days. Tumor volume was measured over<br />

several days. Results: Both KPT185 and 251 specifically reduced CRM1<br />

protein levels in RCC cells. KPT-185 caused dose-dependent cytotoxicity<br />

in RCC cells, which was greater than sorafenib in RCC cell lines but less in<br />

NHK cells, suggesting a possible clinical advantage <strong>of</strong> KPT-185 over<br />

sorafenib. By FACS analysis, we showed that KPT-185 arrests the cell cycle<br />

in both G2/M and G1, and increased the sub-G0 cell population. KPT-185<br />

and 251 both increased p53 and p21 in RCC cells, and KPT-185 confined<br />

these proteins to the nucleus. In vivo, KPT-251 inhibited Caki-1 xenografts<br />

in mice compared to both vehicle and sorafenib without obvious systemic<br />

adverse effects. Conclusions: We introduce a completely novel therapeutic<br />

approach to the treatment <strong>of</strong> RCC based on inhibition <strong>of</strong> the nuclear export<br />

<strong>of</strong> key cell cycle regulatory proteins. Inhibition <strong>of</strong> CRM1 leads to forced<br />

nuclear retention, and thereby activation, <strong>of</strong> several key p53-pathway<br />

proteins, leading to cell cycle arrest and apoptosis in RCC cell lines in vitro<br />

and tumor growth inhibition in vivo.<br />

4636 General Poster Session (Board #9C), Sun, 8:00 AM-12:00 PM<br />

Efficacy and safety <strong>of</strong> cancer vaccine with 4-fold gene-modified allogeneic<br />

tumor cells: Results <strong>of</strong> the phase I/II ASET study in patients with advanced<br />

renal cell carcinoma. Presenting Author: Steffen Weikert, Department <strong>of</strong><br />

Urology, Charité-University Medicine Berlin, Berlin, Germany<br />

Background: MGN1601 was tested in the first-in-man phase I-II clinical<br />

study in patients with advanced renal cell carcinoma (RCC) who failed<br />

several previous therapy lines and had no further standard therapy<br />

available. MGN1601 consists <strong>of</strong> two active pharmaceutical ingredients in<br />

fixed combination: fourfold gene-modified allogeneic tumor cells expressing<br />

IL-7, GM-CSF, CD80 and CD154 through MIDGE vectors and a TLR-9<br />

agonist, the immunomodulator dSLIM. Methods: The ASET study is a<br />

multicentric, single-arm phase I-II clinical trial. <strong>Clinical</strong> response was<br />

evaluated using CT scans (RECIST 1.1 or immune related Response<br />

Criteria, irRC). Efficacy data were evaluated in terms <strong>of</strong> PFS and OS for the<br />

intended to treat and the treated per protocol (TPP) populations, clinical<br />

parameters and quality <strong>of</strong> life. Immune response was determined using<br />

DTH to MGN1601, LTT assay, frequency and activation <strong>of</strong> blood cells, and<br />

mRNA, chemokine and cytotoxic T cells analysis as well as tumor tissue<br />

evaluation. Results: Nine <strong>of</strong> 19 included patients completed the TPP, the<br />

others discontinued the study earlier due to PD. Median PFS in the TPP<br />

group was 12 wks (3 months) and OS (not reached yet) 46 wks (11<br />

months). Three patients achieved disease control (1 PR, 2 SD) after 12<br />

wks. Two patients are continuing treatment in the extension phase and are<br />

progression free since 37 and 46 wks, respectively. Re-evaluation <strong>of</strong> tumor<br />

response data using irRC revealed 1 additional patient with a delayed tumor<br />

response 4 wks after treatment stop. Herewith, 4 out <strong>of</strong> 9 TPP patients<br />

(45%) achieved disease control. Of 7 patients receiving targeted therapy<br />

upon stop <strong>of</strong> study treatment, 4 had substantial objective responses,<br />

providing evidence that the study drug is able to render their tumors more<br />

vulnerable to subsequent therapies. Immune analysis showed trends<br />

towards increases <strong>of</strong> T-, NKT-cell and pDC frequencies and other immune<br />

parameters in those patients with clinical responses, indicating anti-tumor<br />

immunity <strong>of</strong> study treatment. Conclusions: The therapeutic cancer vaccine<br />

MGN1601 shows promising efficacy in late stage mRCC patients. Results<br />

warrant further clinical studies with MGN1601.<br />

4635 General Poster Session (Board #9B), Sun, 8:00 AM-12:00 PM<br />

Genetic polymorphisms’ influence in outcome <strong>of</strong> metastatic renal cell<br />

cancer patients treated with VEGF-targeted agents. Presenting Author:<br />

Fabio Augusto Barros Schutz, Lank Center for Genitourinary Oncology,<br />

Dana-Farber Cancer Institute/Brigham and Women’s Hospital, Harvard<br />

Medical School, Boston, MA<br />

Background: Single nucleotide polymorphisms (SNP) in critical signaling<br />

pathways may impact the outcome <strong>of</strong> metastatic renal cell cancer (mRCC)<br />

patients (pts) treated with VEGF targeted agents. Methods: Germline DNA<br />

was extracted from 263 pts <strong>of</strong> European-<strong>American</strong> ancestry enrolled in a<br />

prospective protocol with full baseline and follow up clinical data. A total <strong>of</strong><br />

113 common SNPs within select genes involved in RCC pathogenesis,<br />

angiogenesis and drugs metabolism were genotyped and associations<br />

sought with clinical outcome. The primary endpoint was progression free<br />

survival (PFS), defined as time from VEGF targeted therapy to progression<br />

or death. Cox model tested for the association between SNPs with PFS in<br />

univariate and multivariate model that adjusted for age, gender, type <strong>of</strong><br />

VEGF inhibitor and MSKCC risk score. The false discovery rate (pFDR) was<br />

used to control for the number <strong>of</strong> tests performed. Results: The median<br />

follow-up time was 51.6 months (mo), 81% <strong>of</strong> pts had progression or death<br />

events. All patients were treated with an approved VEGF targeted agent<br />

(bevacizumab, sunitinib, sorafenib or pazopanib). Fifty three percent<br />

(140/263) <strong>of</strong> pts were treated with sunitinib. Two SNPs, one in HIF2� and<br />

one in VEGFR2, showed a significant association with the risk <strong>of</strong> progression.<br />

Compared to the dominant genotype (frequency 83%, median PFS<br />

10.1mo), the VEGFR2 rs2305948 variant genotype (frequency 17%)<br />

showed a superior PFS (median 19.2mo) corresponded to an adjusted<br />

hazard ratio (HR) <strong>of</strong> 0.56 (95%CI: 0.37-0.84; p�0.005). Similarly and<br />

compared to the dominant genotype (frequency 94%, median PFS 10.6mo),<br />

the HIF2a rs11687512 variant genotype (frequency 6%) had a superior<br />

PFS (median 26.5mo) with an adjusted HR <strong>of</strong> 0.33 (95%CI 0.16-0.68;<br />

p�0.002). The analysis adjusted for age, gender, type <strong>of</strong> VEGF targeted<br />

therapy (Sunitinib vs. others) and MSKCC risk score. False discovery rate<br />

was �20% with both SNPs. Conclusions: Inherited variants may influence<br />

the PFS <strong>of</strong> pts with mRCC treated with VEGF targeted agents. Validation <strong>of</strong><br />

these findings in an independent cohort is required. If validated, these<br />

results could help identifying subset <strong>of</strong> pts that are more likely to remain<br />

progression free on treatment.<br />

4637 General Poster Session (Board #9D), Sun, 8:00 AM-12:00 PM<br />

Longitudinal serum testosterone levels (T) in long-term testicular cancer<br />

survivors (TCSs) in relation to testicular cancer (TC) treatment, aging, and<br />

TC diagnosis itself. Presenting Author: Mette Sprauten, OUS, The Norwegian<br />

Radium Hospital, Oslo, Norway<br />

Background: Low T may increase the risk <strong>of</strong> cardiovascular disease,<br />

osteoporosis, and reduced quality <strong>of</strong> life. T might be reduced by TC, its<br />

treatment, ageing, and particularly their combination. Methods: T was<br />

retrieved from 311 TCSs after orchiectomy and prior to subsequent<br />

management with either surveillance/surgery only (S), radiotherapy (RT) or<br />

cisplatin based chemotherapy (CT). Human Chorionic Gonadotropin (hCG)<br />

was available for 211 TCSs. T was reassessed at surveys performed 9 (S9)<br />

and 18 years (S18) after treatment. T values were categorized into quartiles<br />

according to cut-<strong>of</strong>f values derived from 570 healthy controls (C) for each<br />

decadal age group. Statistical associations were assessed with Chi2 tests.<br />

Results: In TCSs about to receive RT or CT, T and hCG were higher when<br />

compared to those subsequently managed by S. TCSs were more likely to<br />

belong to the lowest T quartile, more so with increasing treatment intensity<br />

(table). The proportions <strong>of</strong> TCSs belonging to the corresponding T quartiles<br />

displayed no significant changes from S9 to S18. Conclusions: TCSs had<br />

lower T than C <strong>of</strong> similar age already after orchiectomy, possibly related to<br />

removal <strong>of</strong> the affected testicle and/or testicular dysgenesis syndrome.<br />

TCSs who were to receive RT or CT had slightly increased T when compared<br />

to S, probably due to hCG stimulation. T reduction by RT or CT has been<br />

described previously but rarely in longitudinal studies. Of note, longitudinal<br />

assessment <strong>of</strong> T without comparison to C might result in overestimation <strong>of</strong><br />

the treatment burden. Relatively stable age adjusted T levels from S9 to<br />

S18 for the TCSs independent <strong>of</strong> treatment are encouraging.<br />

C: n�570,<br />

(100%)<br />

S: n�69,<br />

(100%)<br />

RT: n�133,<br />

(100%)<br />

CT: n�109,<br />

(100%)<br />

Pretreatment<br />

75% 141 (24.7) 3 (4.3) 18 (13.5) 18 (16.5)<br />

OR(95%CI) Referent (R) 13.2 (4.0- 43.7) 3.5 (2.0- 6.2) 2.4 (1.3- 4.3)<br />

S9<br />

75% 141 (24.7) 9 (13.0) 12 (9.0) 12 (11.0)<br />

OR(95%CI) R 3.2 (1.4- 7.0) 5.2 (2.7- 10.0) 5.0 (2.6- 9.7)<br />

S18<br />

75% 141 (24.7) 6 (8.7) 9 (6.8) 10 (9.2)<br />

OR(95%CI) R 4.4 (1.8- 11.0) 6.9 (3.3- 4.4) 6.8 (3.4- 13.7)<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


4638 General Poster Session (Board #9E), Sun, 8:00 AM-12:00 PM<br />

Epidermal growth factor receptor (EGFR) expression and KRAS mutations<br />

in penile squamous cell carcinoma: Analysis <strong>of</strong> potential application <strong>of</strong><br />

anti-EGFR monoclonal antibodies. Presenting Author: Hongfeng Gou,<br />

Department <strong>of</strong> Abdominal Cancer, Cancer Center, West China Hospital,<br />

Sichuan University, Chengdu, China<br />

Background: Penile squamous cell carcinoma (SCC) is a rare cancer with<br />

poor prognosis and limited response to conventional chemotherapy. Anti-<br />

EGFR monoclonal antibodies (mAbs) have exhibited significant anti-tumor<br />

activities in several cancers such as colorectal carcinoma, head and neck<br />

cancer. KRAS mutations are linked to a poor response to mAbs and may<br />

thereby result in resistance to anti-EGFR agents. This study was aimed to<br />

examine EGFR expression and KRAS, BRAF mutations in penile SCC and to<br />

analyze the potential application <strong>of</strong> anti-EGFR monoclonal antibodies<br />

(mAbs) for this disease. Methods: Totally 150 penile SCC specimens from<br />

patients undergone surgical resection were included. EGFR expression was<br />

evaluated by immunohistochemistry. KRAS mutations at codons 12 and<br />

13, and the BRAF mutation at codon 600 were analyzed on DNA isolated<br />

from formalin fixed paraffin embedded tissues by direct genomic sequencing.<br />

Results: EGFR expression was positive in all specimens, and its<br />

overexpression rate was 92%. We failed to observe a significant correlation<br />

between EGFR expression and tumor grade or lymph node metastases. The<br />

detection <strong>of</strong> KRAS and BRAF mutations analysis were performed in 94 and<br />

83 tumor tissues, respectively. KRAS mutation was detected in only one<br />

sample and no patient was found to harbor BRAF V600E point mutation.<br />

Conclusions: We found the overexpression <strong>of</strong> EGFR and the absence <strong>of</strong><br />

KRAS and BRAF mutations in penile SCC. This suggests that anti-EGFR<br />

mAbs may be potentially considerable therapeutic agents in penile SCC.<br />

4640 General Poster Session (Board #9G), Sun, 8:00 AM-12:00 PM<br />

Dynamic sentinel lymph node biopsy in patients with invasive squamous<br />

cell carcinoma <strong>of</strong> the penis: A prospective study <strong>of</strong> the outcome <strong>of</strong> 500<br />

inguinal basins assessed in a single institution. Presenting Author: Wayne<br />

Lam, St. George’s Hospital, London, United Kingdom<br />

Background: Dynamic sentinel node biopsy (DSNB) in combination with<br />

ultrasound scan (USS) has been the technique <strong>of</strong> choice at our centre since<br />

2004 for the assessment <strong>of</strong> non palpable inguinal lymph nodes in patients<br />

with squamous cell carcinoma <strong>of</strong> the penis (SCCp). Sensitivity/falsenegative<br />

rates may vary depending on whether results are reported per<br />

patient or per node basin and with and without USS. The purpose <strong>of</strong> this<br />

study was to determine the long-term outcome <strong>of</strong> DSNB and ultrasoundguided<br />

fine needle aspiration cytology (FNAC) in our cohort <strong>of</strong> newly<br />

diagnosed patients and to analyse any variation in sensitivity <strong>of</strong> the<br />

procedure. Methods: A prospective cohort study over 6 years (2004 to<br />

2010). Inclusion criteria: New diagnosis SCCp, T1G2 or greater definitive<br />

histology, non-palpable nodes in inguinal basin. Exclusion: patient with<br />

persistent/untreated local disease. Sensitivity <strong>of</strong> the procedure was calculated,<br />

per node basin, per patient, DSNB alone, USS/DSNB combined.<br />

Minimum follow up 12 months. Results: 500 inguinal basins in 264<br />

patients underwent USS�/-FNAC and DSNB. 70 (14%) positive inguinal<br />

basins in 57(22%) patients were identified. 9 (2%) inguinal basins had no<br />

tracer uptake. 2 inguinal basins were confirmed false negative at 8 and 12<br />

months. 2 inguinal basins had positive USS�FNAC and negative DSNB.<br />

Overall sensitivity <strong>of</strong> the technique is reported in the table. Conclusions:<br />

DSNB in combination with USS has excellent performance characteristics<br />

to stage patients with clinically node-negative penile cancer with a 3%<br />

false negative rate. USS improves performance by 4% over DSNB alone.<br />

There is no difference in performance <strong>of</strong> the combined technique if it is<br />

reported per node basin or per patient.<br />

USS�/-FNAC � DSNB DSNB alone<br />

Technical (per inguinal basin) 97% 95%<br />

<strong>Clinical</strong> (per patient) 97% 93%<br />

Genitourinary Cancer<br />

311s<br />

4639 General Poster Session (Board #9F), Sun, 8:00 AM-12:00 PM<br />

Activity <strong>of</strong> insulin growth factor-receptor (IGF-1R) antibody cixutumumab<br />

combined with the mTOR inhibitor temsirolimus in patients with metastatic<br />

refractory adrenocortical carcinoma. Presenting Author: Aung Naing,<br />

Department <strong>of</strong> Investigational Cancer Therapeutics (Phase I Program),<br />

University <strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: Adrenocortical carcinoma (ACC) is a rare and aggressive<br />

endocrine malignancywithout an available effective systemic chemotherapy.<br />

IGF-R2 overexpression leading to the activation <strong>of</strong> the IGF1R/<br />

mTOR pathway is well described in ACC. Cixutumumab, a fully human IgG1<br />

monoclonal antibody directed at insulin growth factor-1 receptor (IGF-1R),<br />

was combined with temsirolimus on the basis <strong>of</strong> preclinical data. Methods:<br />

Patientsreceived cixutumumab, 3-6 mg/kg IV weekly, and temsirolimus,<br />

25 mg-37.5 mg IV weekly (4-week cycles), with restaging after 8 weeks.<br />

Results: Twenty-six patients were enrolled (13 [50%] men); median age, 47<br />

years; median number <strong>of</strong> prior therapies, 4. Five patients previously<br />

received an IGF-1R inhibitor and one, temsirolimus. The most frequent<br />

toxicities, at least possibly drug-related, were grade 1-2 thrombocytopenia<br />

(38%), mucositis (58%), hypercholesterolemia (31%), hypertriglyceridemia<br />

(35%), and hyperglycemia (31%). Eleven <strong>of</strong> 26 patients (42%)<br />

achieved stable disease (SD) � 6 months (duration range � 6to21<br />

months) with 3 <strong>of</strong> the 11 having received a prior IGF-1R inhibitor.<br />

Conclusions: Cixutumumab combined with temsirolimus was well tolerated<br />

and more than 40% <strong>of</strong> patients achieved prolonged SD. This study was<br />

supported by R21CA13763301A1 (AN), U01CA62461 (RK), and<br />

U01CA62487 (PL).<br />

4641 General Poster Session (Board #9H), Sun, 8:00 AM-12:00 PM<br />

Adjuvant radiotherapy after primary surgical resection in patients with<br />

adrenocortical carcinoma: Retrospective cohort analysis. Presenting Author:<br />

Mouhammed Amir Habra, University <strong>of</strong> Texas M. D. Anderson Cancer<br />

Center, Houston, TX<br />

Background: Adrenocortical carcinoma (ACC) is a rare malignancy with high<br />

recurrence and mortality rates. The role <strong>of</strong> adjuvant radiotherapy (RT) to<br />

improve outcome remains unclear. Considering the rarity <strong>of</strong> ACC, we<br />

conducted a historical cohort study to ascertain the effect <strong>of</strong> adjuvant RT<br />

on overall survival and recurrence rates. Methods: Patients were selected<br />

from the MD Anderson Cancer Center (MDACC) ACC registry (1998- 2011)<br />

who had primary tumor resection with no evidence <strong>of</strong> distant metastasis at<br />

the time <strong>of</strong> initial diagnosis and a minimum follow-up <strong>of</strong> 6-months. The<br />

adjuvant RT group included patients who received adjuvant RT within 3<br />

months <strong>of</strong> diagnosis. Control group included patients who did not receive<br />

RT and matched based on resection margin status and stage at diagnosis.<br />

Results: There were no significant differences between the adjuvant RT<br />

group (n�15) and comparison group (n �45) in gender distribution, age,<br />

tumor size, functional status, and use <strong>of</strong> adjuvant mitotane therapy. On<br />

multivariate Cox proportional hazard model for overall survival, the adjuvant<br />

RT group had hazard ratio <strong>of</strong> 1.981(95% confidence interval [CI] 0.894-<br />

4.391, p�0.0922) compared to the control group. The differences in<br />

median time to local recurrence, distant recurrence and <strong>of</strong> recurrence free<br />

survival were not significant between the two groups. In subgroup analysis<br />

including only the patients whose initial treatments were from outside <strong>of</strong><br />

MDACC, RT group (n�15) had hazard ratio <strong>of</strong> overall survival <strong>of</strong> 1.604<br />

(95% CI 0.712- 3.613, p�0.2543) compared to group without adjuvant<br />

RT (n� 32). Median times to local recurrence, distant recurrence, or <strong>of</strong><br />

recurrence free survival were also not significantly different between the<br />

two groups. Conclusions: To our knowledge, this is the largest single<br />

institution report about adjuvant RT use in ACC. In our study, RT did not<br />

appear to cause a significant difference in overall survival, recurrence rate,<br />

or time to recurrence. However, it is still possible that patients <strong>of</strong>fered<br />

adjuvant RT and control groups may have been inherently different. Hence,<br />

a prospective study is needed to clarify the role <strong>of</strong> adjuvant RT in patients<br />

with resectable ACC.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


312s Genitourinary Cancer<br />

4642 General Poster Session (Board #10A), Sun, 8:00 AM-12:00 PM<br />

Baseline covariates impacting overall survival (OS) in a phase III study <strong>of</strong><br />

men with bone metastases from castration-resistant prostate cancer.<br />

Presenting Author: Karim Fizazi, Institut Gustave Roussy, Villejuif, France<br />

Background: Prognostic models <strong>of</strong> OS in men with metastatic castrateresistant<br />

prostate cancer (M�CRPC), have been limited. Here we present<br />

an analysis <strong>of</strong> baseline covariates associated with OS from an international<br />

phase 3 study that demonstrated superiority <strong>of</strong> denosumab over zoledronic<br />

acid for prevention <strong>of</strong> skeletal-related events (SRE) in this population<br />

(Fizazi et al., Lancet 2011;377:813-822). Methods: Patients had confirmed<br />

bone metastases (BM) from CRPC (a rising PSA despite castrate<br />

testosterone levels) and no prior bone anti-resorptive therapy. Proportional<br />

hazards modeling with various selection strategies was used to assess the<br />

prognostic significance <strong>of</strong> baseline covariates in multivariate analyses.<br />

Study-specified factors (previous SRE [Y vs N], PSA level [�10 vs �10 ng<br />

/mL]) and additional variables (Cook et al., Clin Cancer Res 2006;12:3361-<br />

3367; Halabi et al., J Clin Oncol 2003;21:1232-1237; Halabi et al., J Clin<br />

Oncol 2008;26:2544-2549) were explored. As no difference in OS was<br />

observed between treatment arms, analyses were performed using the<br />

pooled overall patient population. Results: Analyses included all randomized<br />

subjects with available baseline covariate data (n�1745). At the<br />

primary analysis date (median study duration 12.2 months), OS was 51%.<br />

Various selection strategies produced consistent results. In multivariate<br />

analysis, bone-specific alkaline phosphatase (BAP) �146 �g/L (p�0.0001)<br />

and corrected urinary N-telopeptide (uNTx) �50 nmol/mmol (p�0.0008)<br />

were associated with shorter OS, as were prior SRE (p�0.0002), PSA �10<br />

ng /mL (p�0.0001), visceral metastases (p�0.0002), greater time from<br />

either diagnosis to first BM or first BM to randomization (p�0.0001 for<br />

both), ECOG performance status 2 vs. 0/1 (p�0.017), BPI-SF pain score<br />

�4 (p�0.0001), age (p�0.008), alkaline phosphatase �143 U/L<br />

(p�0.0001), and hemoglobin �128 g/L (p�0.0001). Conclusions: Besides<br />

known factors previously associated with OS in men with CRPC<br />

(Halabi et al., 2003), we show that bone-associated covariates (pain, prior<br />

SRE, BAP, and uNTx) are also important and independent prognostic<br />

factors for OS.<br />

4644 General Poster Session (Board #10C), Sun, 8:00 AM-12:00 PM<br />

Prognostic stratification <strong>of</strong> post-docetaxel metastatic castration resistant<br />

prostate cancer (mCRPC) from a phase III randomized trial. Presenting<br />

Author: Guru Sonpavde, Texas Oncology, Houston, TX, and Department <strong>of</strong><br />

Medicine, Section <strong>of</strong> Medical Oncology, Michael E. DeBakey Veterans<br />

Affairs Medical Center, Baylor College <strong>of</strong> Medicine, Houston, TX<br />

Background: A prognostic model for mCRPC post docetaxel is necessary to<br />

guide therapy. We retrospectively analyzed a phase III trial enrolling<br />

progressive mCRPC following docetaxel to construct a prognostic model.<br />

Additionally, we studied the impact <strong>of</strong> neutrophil-lymphocyte ratio (NLR), a<br />

potential marker for inflammatory and immune state. Methods: A phase III<br />

trial (SUN-1120) comparing prednisone combined with sunitinib (N�584)<br />

or placebo (N�289) for mCRPC following docetaxel-based chemotherapy<br />

was evaluated. The treatment arms were combined for analysis, since no<br />

statistical difference was observed in the primary endpoint <strong>of</strong> overall<br />

survival (OS). A logarithmic transformation was applied to non-normal<br />

factors. The Kaplan-Meier method was used for OS estimation. To identify<br />

an optimal prognostic model for survival, we used a Cox proportional<br />

hazards regression methods with forward stepwise selection, stratifying for<br />

ECOG PS, progression type (PSA or radiographic) and treatment group. A<br />

risk score was calculated and patients were categorized into risk groups to<br />

assess model performance. Results: Data from patients without missing<br />

data (n�806) were used to construct an optimal model. The factors used in<br />

the model that remained individually significant in multivariate analysis<br />

were: log-LDH (HR 2.77 [95% CI�2.23, 3.44], p�0.001), hemoglobin<br />

(0.81 [0.76, 0.87], p�0.001), log-NLR (1.63 [1.38, 1.92], p�0.001),<br />

�1 organ involved (1.53 [1.24, 1.88], p�0.001), log-alkaline phosphatase<br />

(1.14 [1.01, 1.30], p�0.041) and log-PSA (1.07 [1.00, 1.13],<br />

p�0.036). No clear cutpoints were identified; thus, these prognostic<br />

factors were used to group patients into 3 equally sized risk categories.<br />

Low, medium and high risk patients (n�268-270 per group) had median<br />

(95% CI) OS estimates <strong>of</strong> 23.7 (21.4-not reached), 13.5 (11.6-15.8) and<br />

7.3 (6.3-8.4) months, respectively. Conclusions: A prognostic risk model<br />

with readily available variables significantly discriminated between outcomes<br />

in post-docetaxel mCRPC and may provide valuable information in<br />

future studies. High NLR was associated with an independent poor<br />

prognostic impact, and warrants prospective validation.<br />

4643 General Poster Session (Board #10B), Sun, 8:00 AM-12:00 PM<br />

Phase I trial <strong>of</strong> NY-ESO-1/LAGE1 peptide vaccine for metastatic castration<br />

resistant prostate cancer (mCRPC). Presenting Author: Teresa Gray Hayes,<br />

Michael E. DeBakey VA Medical Center and Baylor College <strong>of</strong> Medicine,<br />

Houston, TX<br />

Background: The NY-ESO-1 and LAGE-1 antigens are amplified in malignancies<br />

including prostate cancer. Preclinical induction <strong>of</strong> immune responses<br />

and anti-tumor activity has been demonstrated upon administration <strong>of</strong><br />

these peptides. A phase I/II clinical trial evaluated the feasibility <strong>of</strong> a<br />

combined HLA class-I and class-II peptide vaccine in mCRPC. Methods:<br />

Men with progressive mCRPC, Performance Status �2, PSA �10 ng/ml<br />

and had appropriate HLA class I (A2) and class II haplotypes (DR4, DP4,<br />

DR13) were eligible. Three groups with 3 patients each received the<br />

vaccine subcutaneously every 2 weeks for 6 doses. Group 1 was treated<br />

with a peptide directed at a HLA class I haplotype (HLA A2), Group 2 was<br />

treated with a peptide reacting to a HLA class II haplotype (DR4, DP4, or<br />

DR13), and Group 3 received peptides directed at both Class I and II<br />

haplotypes. Group I and Group II received 1 dose <strong>of</strong> 1000 mcg and Group<br />

III received doses <strong>of</strong> 1000 mcg for each <strong>of</strong> 2 peptides. Androgendeprivation<br />

was continued. Results: Of 14 patients enrolled, all were<br />

evaluable for toxicities and 9 patients completed all 6 treatments per<br />

amended protocol and were evaluable for efficacy. The median baseline<br />

PSA was 85.19 ng/ml. Four patients were chemo naive and 5 were<br />

post-docetaxel. One patient had a grade 5 event (myocardial infarction)<br />

unlikely therapy-related. Potential therapy related toxicities were local<br />

grade 1 injection site erythema (n�5), fatigue (n�2), flu-like symptoms<br />

(n�1), myalgias (n�1), anorexia (n�1), nausea (n�1) and leukocytosis<br />

(n�1). The median PSA doubling time pre-therapy and during therapy were<br />

3.1 and 4.92 months, respectively. The PSA-DT was prolonged compared<br />

to baseline in 6 patients, including a negative PSA slope in 2 patients.<br />

Antigen-specific immune response determined by ELISPOT was observed,<br />

and its association with slowing <strong>of</strong> PSA-doubling time will be further<br />

examined. Conclusions: In men with mCRPC, HLA class-I and/or class-II<br />

restricted NY-ESO-1 and LAGE-1 peptides administered subcutaneously<br />

demonstrated excellent tolerability, slowing <strong>of</strong> PSA doubling time and<br />

antigen-specific immune responses. Further development <strong>of</strong> peptide vaccines<br />

alone or in combination with other regimens may be warranted.<br />

4645 General Poster Session (Board #10D), Sun, 8:00 AM-12:00 PM<br />

Inhibition <strong>of</strong> 3�-hydroxysteroid dehydrogenase by abiraterone: A rationale<br />

for increasing drug exposure in castration-resistant prostate cancer. Presenting<br />

Author: Nima Sharifi, University <strong>of</strong> Texas Southwestern Medical Center,<br />

Dallas, TX<br />

Background: Treatment with abiraterone acetate (abi) increases the survival<br />

<strong>of</strong> men with castration-resistant prostate cancer (CRPC). Resistance to abi<br />

invariably occurs, probably due in part to up-regulation <strong>of</strong> steroidogenic<br />

enzymes and/or other mechanisms that sustain the synthesis <strong>of</strong> dihydrotestosterone<br />

(DHT), which raises the possibility <strong>of</strong> reversing resistance by<br />

concomitant inhibition <strong>of</strong> other required steroidogenic enzymes. The 1,000<br />

mg daily abi dose was selected for the phase III trials despite the absence <strong>of</strong><br />

dose-limiting toxicities at higher doses. Based on the 3�-hydroxyl, �5- structure, we hypothesized that abi also inhibits 3�-hydroxysteroid dehydrogenase/isomerase<br />

(3�HSD), which is absolutely required for the intratumoral<br />

synthesis <strong>of</strong> DHT in CRPC, regardless <strong>of</strong> origins or routes <strong>of</strong> synthesis.<br />

Methods: We tested if abi inhibits recombinant 3�HSD2 activity in vitro or<br />

endogenous 3�HSD activity in LNCaP and LAPC4 cells, including conversion<br />

<strong>of</strong> [ 3H]-dehydroepiandrosterone (DHEA) to androstenedione (AD),<br />

androgen receptor (AR) nuclear translocation, expression <strong>of</strong> AR-responsive<br />

genes, and LAPC4 xenograft growth in orchiectomized mice supplemented<br />

with DHEA. Results: Abi has a mixed inhibition pattern <strong>of</strong> 3�HSD2 in vitro,<br />

blocks the conversion from DHEA to AD and DHT with an IC50 <strong>of</strong> � 1 �Min<br />

CRPC cell lines, inhibits AR nuclear translocation and expression <strong>of</strong><br />

TMPRSS2, and decreases CRPC xenograft growth in DHEA-supplemented<br />

mice. Conclusions: Abi blocks 3�HSD enzymatic activity, synthesis <strong>of</strong> AD<br />

and DHT, inhibits the AR-response, and suppresses growth <strong>of</strong> CRPC cells at<br />

concentrations that are clinically achievable. Variable abi inhibition <strong>of</strong><br />

3�HSD might account in part for the heterogeneous clinical response to<br />

abi. More importantly, 3�HSD inhibition with abi might be clinically<br />

harnessed to reverse resistance to CYP17A1 inhibition at the standard dose<br />

by dose-escalation, or simply by administration with food to increase drug<br />

exposure.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


4646 General Poster Session (Board #10E), Sun, 8:00 AM-12:00 PM<br />

Polymorphisms in fragile histidine triad gene (FHIT) associated with<br />

aggressive prostate cancer and cancer specific mortality. Presenting<br />

Author: Yan Ding, City <strong>of</strong> Hope, Duarte, CA<br />

Background: FHIT is a 1.5 Mb gene encoding a 16.8 kD triphosphatase<br />

known to promote apoptosis responding to DNA damage in epithelial cells.<br />

It resides in the most frequently observed fragile site in the human genome,<br />

FRA3B, and is frequently deleted in early development <strong>of</strong> multiple cancer<br />

types. More importantly, down regulation <strong>of</strong> FHIT protein has been<br />

associated with clinical features <strong>of</strong> tumors, such as the presence <strong>of</strong><br />

extraprostatic extension and Gleason Score � 8 in prostate cancer (CaP),<br />

advanced diseases in breast cancer, and shorter survival after platinumbased<br />

treatment for advanced non-small cell lung cancer. Previously, a<br />

genetic locus for CaP has been reported in FHIT. Here we evaluated<br />

association <strong>of</strong> aggressive CaP and death due to CaP with Single nucleotide<br />

polymorphisms (SNPs) in FHIT using cases and controls <strong>of</strong> European origin<br />

nested in the Prostate, Lung, Colorectal, and Ovarian (PLCO) Cancer<br />

Screening trial with 13 years <strong>of</strong> follow-up. Methods: We analyzed 659 SNPs<br />

on FHIT in 1,164 CaP patients (476 with nonaggressive CaP, Gleason � 7<br />

and stage � III; 688 with aggressive CaP, Gleason �� 7 and/or stage III/IV;<br />

63 with death due to CaP) and 1,167 controls using an incidence density<br />

sampling strategy. Stratified Cochran-Mantel-Haenszel test and logistic<br />

regression were performed to test association. Results: We detected<br />

significant association (p � 0.05) <strong>of</strong> incident CaP to 48 SNPs and<br />

aggressive CaP to 52 SNPs. After 50,000 permutations, two SNPs within a<br />

7 kb region, one for incident CaP and the other for aggressive CaP,<br />

remained marginally significant (corrected p � 0.053 and 0.093, respectively).<br />

The same risk allele for aggressive CaP was also associated with<br />

CaP-specific mortality (OR � 1.50, p � 0.027). The association was<br />

stronger after adjusting for Gleason, stage, and PSA at diagnosis, treatment<br />

options after diagnosis. Test on imputed SNPs within the 7 kb intronic<br />

region identified 7 additional SNPs in 2 linkage disequilibrium groups at<br />

higher risk to CaP-specific death (OR � 1.98, p � 0.00015 and OR �<br />

2.51, p � 0.0027). Conclusions: Risk SNPs in FHIT were implicated for<br />

aggressive disease and CaP specific death. Replication <strong>of</strong> these SNPs in<br />

other populations is warranted.<br />

4648 General Poster Session (Board #10G), Sun, 8:00 AM-12:00 PM<br />

Overall survival (OS) benefit with sipuleucel-T by baseline PSA: An<br />

exploratory analysis from the phase III IMPACT trial. Presenting Author:<br />

Gerald Chodak, Weiss Memorial Hospital, Chicago, IL<br />

Background: Sipuleucel-T is an autologous cellular immunotherapy approved<br />

for asymptomatic or minimally symptomatic metastatic castrateresistant<br />

prostate cancer. In the IMPACT trial, sipuleucel-T showed a<br />

22.5% reduction in risk <strong>of</strong> death vs the control group (hazard ratio<br />

[HR]�0.775 [95% CI 0.614, 0.979]; P�0.032). A pre-specified subgroup<br />

analysis for baseline prognostic variables showed homogeneous<br />

treatment effects consistently favoring sipuleucel-T. In patients (pts) with<br />

baseline PSA below vs above the median, there was a trend toward greater<br />

treatment effect (HR�0.685 vs. 0.865). In this exploratory analysis, we<br />

further sub-divide baseline PSA into quartiles to evaluate potential treatment<br />

effect patterns. Methods: The analysis included all randomized pts<br />

from IMPACT (n�512). Pts were categorized by baseline PSA quartile<br />

(Table), ECOG PS and by median for other baseline prognostic variables<br />

(i.e., LDH, PAP, ALP in bone-only disease, and Hgb). Median OS and HR<br />

were estimated using Kaplan-Meier and Cox models, respectively. Results:<br />

Increasing baseline PSA quartile was associated with markers <strong>of</strong> advanced<br />

disease. HRs suggest a consistent treatment effect in all subsets, although<br />

there is inadequate power to show significant results within each quartile.<br />

There was a trend toward an increased magnitude <strong>of</strong> treatment benefit in<br />

pts with a lower baseline PSA (Table). Results for other baseline prognostic<br />

variables also suggest a trend toward greater benefit in subjects with better<br />

prognostic features. However, results for baseline Hgb indicated an<br />

opposite trend. Conclusions: Although not adequately powered for significance,<br />

the results <strong>of</strong> this analysis support a consistent OS benefit with<br />

sipuleucel-T across PSA quartiles. The greater magnitude <strong>of</strong> benefit in pts<br />

with lower baseline PSA suggests that pts with less advanced disease may<br />

benefit more from treatment with sipuleucel-T.<br />

Baseline PSA (ng/mL)<br />

22.1–50.1 >50.1–134.1 >134.1<br />

n 128 128 128 128<br />

Median OS, months<br />

Sipuleucel-T 41.3 27.1 20.4 18.4<br />

Control 28.3 20.1 15.0 15.6<br />

Difference 13.0 7.1 5.4 2.8<br />

HR (95% CI) 0.51 (0.31, 0.85) 0.74 (0.47, 1.17) 0.81 (0.52, 1.24) 0.84 (0.55, 1.29)<br />

Genitourinary Cancer<br />

313s<br />

4647 General Poster Session (Board #10F), Sun, 8:00 AM-12:00 PM<br />

Guideline-discordant androgen deprivation therapy (ADT) use in localized<br />

prostate cancer (CaP) and cost implications: A population-based study.<br />

Presenting Author: Adam Rea Kuykendal, University <strong>of</strong> North Carolina at<br />

Chapel Hill, Chapel Hill, NC<br />

Background: ADT use in localized CaP has increased overall survival and is<br />

recommended by National Comprehensive Cancer Network (NCCN) guidelines<br />

in certain clinical situations. However, ADT may cause harm and is<br />

without benefit in other situations. Prior studies showed a decline in<br />

“inappropriate” ADT use coinciding with Medicare reimbursement changes<br />

in 2004-2005. This study examines recent trends in ADT use and<br />

quantifies the cost <strong>of</strong> guideline-discordant ADT. Methods: Patients in the<br />

Surveillance Epidemiology and End Results (SEER)-Medicare database<br />

diagnosed with non-metastatic CaP between 2004 and 2007, ages 66-80<br />

were included for analysis. PSA, Gleason score and clinical stage were used<br />

to define D’Amico risk categories. Logistic regression was used to examine<br />

factors associated with guideline-discordant ADT use. <strong>Annual</strong> direct cost<br />

was estimated using the current Medicare reimbursement amount for ADT.<br />

Results: Of 24,280 men included, 13% received guideline-discordant ADT.<br />

Discordant use declined from 15% in 2004 to 11% in 2007. In low-risk<br />

patients, 15% received discordant ADT, mostly due to simultaneous ADT<br />

with radiation. Discordant use was seen in 7% <strong>of</strong> intermediate and 16% <strong>of</strong><br />

high-risk patients, mostly from ADT monotherapy. African <strong>American</strong> (AA)<br />

(p�.001), older patients (p�.001) and those with more comorbidities<br />

(p�.001) were more likely to receive discordant ADT (Table). The estimated<br />

annual direct cost to Medicare from discordant ADT is $43,500,000.<br />

Conclusions: Approximately one in eight patients received ADT discordant<br />

with published guidelines, with AA and elderly patients disproportionately<br />

affected. Elimination <strong>of</strong> discordant use would result in substantial savings<br />

in healthcare costs.<br />

Covariate OR p value<br />

AA (vs. Caucasian) 1.35 �.001<br />

Age (vs. 65-69)<br />

70-74 1.58 �.001<br />

75-79 3.22 �.001<br />

Modified Charlson comorbidity (vs. 0)<br />

>0 1.34 �.001<br />

Diagnosis (vs. 2004)<br />

2005 .86 .008<br />

2006 .78 �.001<br />

2007 .71 �.001<br />

Risk group (vs. low risk)<br />

Intermediate .40 �.001<br />

High .88 .003<br />

Controls for SEER region, regional socioeconomic indicators, and marital status.<br />

4649 General Poster Session (Board #10H), Sun, 8:00 AM-12:00 PM<br />

Targeted next-generation sequencing (NGS) <strong>of</strong> advanced prostate cancer<br />

(PCA) using formalin-fixed tissue. Presenting Author: Himisha Beltran,<br />

Weill Cornell Medical College, New York, NY<br />

Background: The genomic landscape <strong>of</strong> advanced PCA is not well characterized,<br />

partly due to limited availability <strong>of</strong> frozen metastatic tissue. This has<br />

created a gap in our knowledge <strong>of</strong> treatment related and potentially<br />

targetable genomic alterations. The purpose <strong>of</strong> this study was to demonstrate<br />

feasibility <strong>of</strong> performing NGS to molecularly characterize advanced<br />

PCA using formalin-fixed paraffin embedded (FFPE) tissue. Methods: 25<br />

metastatic CRPC, 4 metastatic hormone naïve PCA, 3 primary localized<br />

PCA, and 18 benign matched prostates were evaluated (40mm FFPE tissue<br />

per case). High-density foci were captured and sequenced for 3230 exons<br />

<strong>of</strong> 182 cancer-related genes and 37 introns <strong>of</strong> 14 genes <strong>of</strong>ten rearranged in<br />

cancer to an average depth <strong>of</strong> �800x in a CLIA lab (Foundation Medicine).<br />

Recurrent mutations, copy number alterations, and fusions were validated<br />

using PCR and FISH. Results: In �90% <strong>of</strong> samples, there was sufficient<br />

DNA (�50 ng) for analysis. Recurrent high confidence cancer alterations in<br />

CRPC included: TMPRSS2-ERG fusion (44%), PTEN loss (44%), TP53<br />

mutation (40%), AR mutation (24%), AR gain (24%), RB mutation (28%),<br />

MYC gain (12%), and BRCA2 loss (12%). Overall 48% <strong>of</strong> CRPC harbored<br />

AR gene alterations. Additionally, there were mutations in CTNNB1 (12%),<br />

ATM (8%) and PIK3CA (4%). Copy number alterations not previously<br />

described in PCA included CCND1, CDK4/6 gains and CDKN2A/CDKN2B<br />

deletions. Hormone naïve metastatic and high risk localized PCA demonstrated<br />

similar frequency <strong>of</strong> TMPRSS2-ERG gene fusion, BRCA2 deletion,<br />

and TP53 mutations as CRPC, but AR alterations and MYC gain were not<br />

seen. Conclusions: This study demonstrates feasibility <strong>of</strong> in-depth, NGS<br />

based DNA analysis using FFPE tissue, even biopsy material. Frequent AR<br />

alterations in CRPC, mutations associated with disease progression, and<br />

potential drug targets were identified. Focused NGS has clinical potential<br />

to identify actionable genomic alterations in advanced PCA that can impact<br />

patient participation in trials as well as treatment and outcome. Treatment<br />

options include PARP inhibitors for patients with BRCA2 and ATM<br />

alterations (20% <strong>of</strong> cases) and PI3K/AKT inhibitors for PIK3CA mutations.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


314s Genitourinary Cancer<br />

4650 General Poster Session (Board #11A), Sun, 8:00 AM-12:00 PM<br />

Correlation <strong>of</strong> increased eosinophil count following sipuleucel-T treatment<br />

with outcome in patients (pts) with metastatic castrate-resistant prostate<br />

cancer (mCRPC). Presenting Author: Douglas G. McNeel, University <strong>of</strong><br />

Wisconsin-Madison, Madison, WI<br />

Background: Sipuleucel-T is the first autologous cellular immunotherapy<br />

approved for asymptomatic or minimally symptomatic mCRPC. In the<br />

IMPACT trial, pts treated with sipuleucel-T had transient increases in<br />

eosinophil counts compared with control. In this retrospective analysis, we<br />

assess potential correlations between eosinophilia, overall survival (OS),<br />

prostate cancer-specific survival (PCSS) and immune response following<br />

sipuleucel-T. Methods: Data from three phase III trials (D9901, D9902A,<br />

and IMPACT) were pooled. The analysis included CBCs performed at<br />

baseline and wks 2–34. Eosinophilia was defined as either �ULN (with<br />

normal baseline), or a max change from baseline within the top quartile, at<br />

any time between wks 2–16. Results: Increased eosinophil counts were<br />

seen in sipuleucel-T pts by wk 6, decreasing to near baseline by wk 14;<br />

eosinophil counts in control pts were stable. Of 377 sipuleucel-T pts<br />

eligible for analysis, 105 (27.9%) had eosinophilia. Baseline disease<br />

characteristics associated with eosinophilia were indicative <strong>of</strong> better<br />

prognosis (i.e., longer Halabi predicted survival [p�0.007], lower PSA<br />

[P�0.033], higher Hgb [p�0.001], and no prior docetaxel [p�0.012]). In<br />

univariate analyses, eosinophilia correlated with improved OS (HR�0.75;<br />

95%CI: 0.56–1.01; p�0.057) and PCSS (HR�0.71; 95%CI: 0.53–<br />

0.97; p�0.031); trends persisted after adjusting for Halabi. The magnitude<br />

<strong>of</strong> eosinophilia positively correlated with antigen-specific humoral<br />

responses (p �0.039 for wks 6, 14 and 26) and elevations in the cytokines<br />

at wk 6 (IL2 [p�0.011], IL5 [p�0.038] and TARC [p�0.001]). AEs<br />

occurring more frequently (p�0.05) in pts with eosinophilia were infusionrelated:<br />

pyrexia (33.3 v 21.3%) and nausea (19.0 v 10.7%). No cases <strong>of</strong><br />

hypereosinophilic syndrome were reported. Conclusions: Increases in eosinophils<br />

after sipuleucel-T correlated with improved OS and PCSS. Large<br />

increases in eosinophil counts were associated with humoral responses and<br />

Th2-type cytokine production. Further studies <strong>of</strong> eosinophilia as a biomarker<br />

for sipuleucel-T response, particularly in earlier disease settings,<br />

are <strong>of</strong> interest.<br />

4652 General Poster Session (Board #11C), Sun, 8:00 AM-12:00 PM<br />

Comparing the ability <strong>of</strong> clinicians and a nomogram model at predicting<br />

future 99mTc-bone scan (BS) positivity in patients (pts) with rising prostatespecific<br />

antigen (PSA) following radical prostatectomy (RP) for prostate<br />

cancer (PCa). Presenting Author: Michael W. Kattan, Department <strong>of</strong><br />

Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH<br />

Background: Nomograms that assist clinicians in predicting which PCa pts<br />

have a high risk <strong>of</strong> developing M1 disease have been developed, but<br />

clinicians still tend to use their own judgment <strong>of</strong> risk when managing<br />

individual pts. This is the first study to compare the ability <strong>of</strong> clinicians vs a<br />

nomogram at predicting future BS positivity in pts with PCa. Only androgen<br />

deprivation therapy- (ADT-) naive post-RP pts were considered, as there are<br />

currently no nomograms for predicting M1 disease in pts receiving ADT.<br />

The Slovin nomogram (Slovin et al. Clin Cancer Res 2005; 11: 8669-<br />

8673) was chosen based on considerations <strong>of</strong> the numbers <strong>of</strong> pts analyzed<br />

and the follow-up intervals used in its development. Methods: During an<br />

advisory board in June 2011, 24 PCa experts were given details <strong>of</strong> 25<br />

ADT-naive post-RP PCa pts with biochemical recurrence who were randomly<br />

selected from the Cleveland Clinic Database <strong>of</strong> 759 pts. The<br />

predicted 1-yr from today probability <strong>of</strong> freedom from metastasis was<br />

generated using the Slovin nomogram and estimated by the clinicians. The<br />

predictive accuracy was quantified using the concordance index (C-index).<br />

Results: All pts were Caucasian and median age was 62 yrs. Median PSA<br />

level at biochemical recurrence was 1.2 ng/mL and PSADT varied between<br />

1.3 and 11.7 months. The data used for each <strong>of</strong> the pts was acquired<br />

between 4 and 133 months after the RP procedure. Eight pts had a positive<br />

BS within 1 yr. Considerable variation between the clinicians’ predictions<br />

for each pt was observed. The nomogram gave a higher C-index compared to<br />

the clinicians (0.812 and 0.628, respectively; p�0.003). The clinicians<br />

typically overestimated the risk <strong>of</strong> developing M1 disease. The nomogram<br />

outperformed all <strong>of</strong> the clinicians (C-index range: 0.47 to 0.75). Urologists<br />

predictions were superior to those <strong>of</strong> oncologists (p�0.048). Conclusions:<br />

The Slovin nomogram was more accurate than expert clinicians in predicting<br />

1-yr BS positivity. We suggest nomograms should become an integral<br />

part <strong>of</strong> the clinical decision process in the PCa setting.<br />

4651 General Poster Session (Board #11B), Sun, 8:00 AM-12:00 PM<br />

Proton radiotherapy for prostate cancer in the Medicare population:<br />

Patterns <strong>of</strong> care and comparison <strong>of</strong> early toxicity with IMRT. Presenting<br />

Author: James B. Yu, Yale University School <strong>of</strong> Medicine, New Haven, CT<br />

Background: Proton radiotherapy (PRT) is a costly treatment used for<br />

prostate cancer despite little evidence supporting its use. We examined<br />

patterns <strong>of</strong> PRT use in the Medicare program and assessed the short-term<br />

toxicity <strong>of</strong> PRT vs. intensity modulated radiation therapy (IMRT). Methods:<br />

Using national Medicare claims from 2008-2009, we identified a sample<br />

<strong>of</strong> prostate cancer patients ages 66-94 who had received PRT or IMRT. We<br />

used multivariable logistic regression to identify patient and regional<br />

factors associated with receipt <strong>of</strong> PRT. We searched claims for procedure<br />

and diagnosis codes indicative <strong>of</strong> treatment-related complications and<br />

grouped the complications into genitourinary (GU), gastrointestinal (GI),<br />

and other complications. To compare the effect <strong>of</strong> PRT and IMRT on<br />

short-term toxicity, we used a Mahalanobis distance approach to match<br />

each PRT patient to two IMRT patients, achieving balanced distribution <strong>of</strong><br />

clinical and sociodemographic characteristics. We compared six-month<br />

and one-year outcomes between the two treatment groups using conditional<br />

logistic regression. Results: We identified 27,647 men; 421 (2%)<br />

received PRT and 27,226 (98%) received IMRT. Patients who received<br />

PRT were widely geographically distributed, with some patients traveling<br />

�500 miles for treatment. PRT patients were younger, healthier, and <strong>of</strong><br />

higher socioeconomic status. Although PRT was associated with a significant<br />

reduction in GU complications at six-months compared with IMRT<br />

(6.1% vs. 12.0%, OR 0.60 [95% CI 0.38-0.96], p�0.03), at one-year<br />

post-treatment there was no longer any difference in cumulative complication<br />

rates (18.9% vs. 21.9%, OR 0.96 [95% CI 0.61-1.53], p�0.88).<br />

There was no significant difference in GI or other complications at<br />

six-months or one-year post-treatment. Conclusions: Although PRT remains<br />

a scarcely used treatment, some prostate cancer patients traveled great<br />

distances for treatment. While PRT was associated with a reduction in<br />

six-month GU toxicity, there were no differences in toxicity at one-year.<br />

Further study on longer-term effects and other clinical and patient-reported<br />

outcomes is needed to inform the widespread application <strong>of</strong> PRT.<br />

4653 General Poster Session (Board #11D), Sun, 8:00 AM-12:00 PM<br />

Detecting prostate cancer in BRCA1 and BRCA2 mutation carriers: A role<br />

for EN2? Presenting Author: Emma Killick, Institute <strong>of</strong> Cancer Research,<br />

Sutton, United Kingdom<br />

Background: EN2 is part <strong>of</strong> the HOX gene family and plays a role in foetal<br />

development. More recently a potential oncogenic role for the protein has<br />

been postulated and its utility as a cancer biomarker has been explored in<br />

prostate cancer (PrCa) and breast cancer. Carriers <strong>of</strong> mutations in the<br />

BRCA1 and BRCA2 genes have an increased risk <strong>of</strong> PrCa (1.8-fold and<br />

5-fold respectively) and their tumours tend to be more aggressive and<br />

advanced than sporadic cases. Currently there is no national screening<br />

program for BRCA mutation carriers in the UK, and the IMPACT study was<br />

set up to evaluate PSA screening in this particular group. Here we analyse<br />

the efficacy <strong>of</strong> the urinary EN2 protein as a marker <strong>of</strong> early cancer detection<br />

within this higher risk group. Methods: First pass urine (without preceding<br />

digital rectal examination) was collected as part <strong>of</strong> the IMPACT screening<br />

study which enrolled individuals aged between 40 and 69 who were<br />

unaffected by PrCa at time <strong>of</strong> enrolment into the study (n� 418). All<br />

participants were from families harbouring a BRCA1 or BRCA2 mutation<br />

and were either BRCA mutation carriers themselves or controls with a<br />

negative predictive BRCA genetic test. They underwent annual PSA test<br />

with a PSA <strong>of</strong> � 3.0 ng/ml triggering a diagnostic biopsy. EN2 protein was<br />

measured in the urine using an ELISA; (positive ��42.5ng/ml). Results:<br />

Our initial results demonstrated urinary EN2 had a sensitivity <strong>of</strong> 66.67%<br />

and a specificity <strong>of</strong> 89.29% when discriminating which men had been<br />

diagnosed with PrCa; the ROC AUC was 0.816. The difference in EN2 level<br />

between those diagnosed with cancer and those who were not was<br />

significant (p ��0.001). There was trend towards higher EN2 levels in<br />

those with more aggressive tumours (median EN2 84.5ng/mL in Gleason<br />

�3�4 vs 111ng/mL in Gleason �4�3), however this was not statistically<br />

significant. In one PrCa case EN2 rise preceded PSA rise by 2 years.<br />

Further samples are in the process <strong>of</strong> being analysed, results from these will<br />

be included. Conclusions: Urinary EN2 protein measurement warrants<br />

further investigation as a PrCa biomarker in this higher risk group with<br />

genetic predisposition to PrCa.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


4654 General Poster Session (Board #11E), Sun, 8:00 AM-12:00 PM<br />

A phase II study <strong>of</strong> KX2-391, an oral inhibitor <strong>of</strong> Src kinase and tubulin<br />

polymerization, in men with bone-metastatic castration-resistant prostate<br />

cancer (CRPC): A PCCTC trial. Presenting Author: Emmanuel S. Antonarakis,<br />

Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins<br />

University, Baltimore, MD<br />

Background: KX2-391 is an oral small molecule inhibitor <strong>of</strong> Src kinase and<br />

tubulin polymerization. In phase 1 trials, PSA declines were seen in men<br />

with advanced prostate cancer. Methods: A single-arm phase 2 study <strong>of</strong><br />

KX2-391 in 31 men with chemo-naïve bone-metastatic CRPC was conducted<br />

at 5 PCCTC sites. Men received oral KX2-391 (40 mg BID) until<br />

disease progression or unacceptable toxicity. The primary endpoint was<br />

progression-free survival (PFS) at 24 wk (progression � clinical/<br />

radiographic progression or death, but not rising PSA; PCWG2 criteria); a<br />

50% success rate was predefined as clinically significant. Secondary<br />

endpoints were PSA progression-free survival (PPFS) at 24 wk (PSA<br />

progression � 25% PSA rise above nadir; PCWG2 criteria); median PFS;<br />

median PPFS; and max PSA decline. Exploratory outcomes included PK<br />

studies, CTC enumeration, and analysis <strong>of</strong> markers <strong>of</strong> bone resorption<br />

(urinary N-telopeptide [uNTx]; C-telopeptide [CTx]) and formation (bone<br />

alk phos [BAP]; osteocalcin). Results: The trial closed early after accrual <strong>of</strong><br />

31 men, due to a prespecified futility rule. In all, 26/31 men were<br />

evaluable for the primary endpoint; efficacy results are shown below. Also,<br />

2/11 men (18%) with unfavorable (�5) CTCs at baseline converted to<br />

favorable (�5) CTC counts on study, and 18/19 men (95%) with baseline<br />

favorable CTC counts maintained them. The proportion <strong>of</strong> men with<br />

declines in bone turnover markers was 32% (8/25) for uNTx, 21% (6/29)<br />

for CTx, 10% (3/29) for BAP, and 25% (7/28) for osteocalcin. In PK<br />

studies, median Cmax was 61 (range 16–129) ng/mL, and AUC was 156<br />

(35–348) ng*hr/mL. Common toxicities (�10%) included LFT elevations,<br />

leukopenia, thrombocytopenia, fatigue, nausea and constipation.<br />

Conclusions: KX2-391 dosed at 40 mg BID lacks antitumor activity in men<br />

with CRPC, but modulates bone turnover markers in some men. Because a<br />

Cmax <strong>of</strong> �142 ng/mL is required for tubulin polymerization inhibition,<br />

higher once-daily dosing will be used in future trials.<br />

Endpoint Value 95% CI<br />

Primary<br />

PFS at 24 wk (%) 7.7 2.3 – 24.3<br />

Secondary<br />

PPFS at 24 wk (%) 0 0 – 13.2<br />

Median PFS (wk) 18.6 12.1 – 24.0<br />

Median PPFS (wk) 5.0 4.1 – 11.4<br />

>30% PSA decline (%) 9.7 3.5 – 25.0<br />

4656 General Poster Session (Board #11G), Sun, 8:00 AM-12:00 PM<br />

Phase I results from a phase I/II study <strong>of</strong> orteronel, an oral, investigational,<br />

nonsteroidal 17,20-lyase inhibitor, with docetaxel and prednisone (DP) in<br />

metastatic castration-resistant prostate cancer (mCRPC). Presenting Author:<br />

Daniel Peter Petrylak, Columbia University Medical Center, New York,<br />

NY<br />

Background: The investigational agent orteronel (ortl, TAK-700) is a<br />

selective 17,20-lyase inhibitor that blocks androgen production. Since DP<br />

is standard chemotherapy in mCRPC, this phase 1/2 study examined ortl �<br />

DP in men with mCRPC. Methods: The primary phase I objective was to<br />

determine the maximum dose <strong>of</strong> ortl 200–400 mg BID that can be<br />

administered safely with DP in castrate men (testosterone [T] �50 ng/dL)<br />

with mCRPC, �1 prior chemotherapy and no ketoconazole/abiraterone<br />

�30 d prior. A 3�3 dose escalation was used with ortl 200 then 400 mg<br />

BID. Ortl dosed daily and D (75 mg/m2 q3w) � P (5 mg BID) was started on<br />

d8 <strong>of</strong> cycle 1 (28 d); cycles �2 � 21 d. Results: 14 men, median age 68 yrs<br />

(range 53–81), ECOG PS 0/1 (71%/29%), median PSA 59.4 ng/mL<br />

(5.2–1052), T 8.1 ng/dL (1.2–16.7), 9 with measurable disease, were<br />

treated. Median ortl exposure was 33.3 wks (6.3–59); 6 and 8 men<br />

received ortl 200 and 400 mg BID � DP, in cohorts 1 and 2, respectively.<br />

No dose-limiting toxicities (DLTs) occurred in cohort 1 (2 x 3 pts); in cohort<br />

2 (2 x 4 pts), 1 pt in group 1 had DLT (Gr3 related febrile neutropenia); no<br />

DLTs occurred in group 2. 3 pts discontinued due to AEs (2 D infusion<br />

reactions, 1 at each dose; 1 unrelated cardiorespiratory death at 200 mg).<br />

All men had at least 1 AE Gr �3 which were treatment-related in 11 men.<br />

The most common Gr �3 AEs were seen at both doses and included<br />

neutropenia 50% (n � 7), hyperglycemia 21% (n � 3), febrile neutropenia,<br />

infusion reaction, hyponatremia, decreased neutrophil count (2 each); at<br />

400 mg only: fatigue, hypophosphatemia, and decreased WBC count (2<br />

each). 7 men had serious AEs (SAEs), which were drug-related in 5. The<br />

most common SAE was febrile neutropenia in 2 men. At 3 mo, PSA50 and<br />

PSA90 rates were 86% and 36%, respectively; 12/14 men had a median<br />

PSA decline �70%; and median T declined to �0.2 ng/dL (0–10.7). The<br />

Cmax <strong>of</strong> D � ortl was similar to D alone. Conclusions: Ortl 200 mg and 400<br />

mg BID � DP appears safe and tolerable with androgen-lowering activity at<br />

the maximum planned dose in mCRPC. There is no evidence <strong>of</strong> AE<br />

potentiation when ortl is administered with DP. The phase II portion <strong>of</strong> the<br />

study will use ortl 400 mg BID � DP.<br />

Genitourinary Cancer<br />

315s<br />

4655 General Poster Session (Board #11F), Sun, 8:00 AM-12:00 PM<br />

Liver metastases (LM) to predict for short overall survival (OS) in metastatic<br />

castration-resistant prostate cancer (mCRPC) patients (pts). Presenting<br />

Author: William Kevin Kelly, Kimmel Cancer Center at Thomas Jefferson<br />

University, Philadelphia, PA<br />

Background: Patients withCRPC with LM represent a subset <strong>of</strong> patients with<br />

a poor prognosis. An exploratory analysis was performed to evaluate the<br />

difference in baseline characteristics and clinical outcomes in patients<br />

with and without LM from a randomized phase III trial (CALGB 90401) in<br />

men with mCRPC. Methods: Data from 1,050 men treated with docetaxel,<br />

prednisone with either bevacizumab or placebo were used. Pts were<br />

chemotherapy naïve, and had evidence <strong>of</strong> progressive mCRPC despite<br />

castrate testosterone levels and anti-androgen withdrawal, ECOG performance<br />

status � 2, and adequate bone marrow, hepatic and renal functions.<br />

The proportional hazards model was used to assess the prognostic significance<br />

<strong>of</strong> LM in predicting OS and progression free survival (PFS) adjusting<br />

for stratification factors. Results: Fifty-nine (5.6%) <strong>of</strong> the 1045 pts with a<br />

complete data set had documented LM. Patients with LM had higher<br />

baseline alkaline phosphatase (ALK, 167 vs 117 U/L, p �0.0205) and<br />

lactate dehydrogenase (LDH, 262 vs 205 U/L, p �0.0001) compared to<br />

patients without LM. There were strong associations between LM status<br />

and lung metastasis (p�0.0004) and other visceral disease (p��0.001)<br />

but not with bone disease. <strong>Clinical</strong> outcomes as a function <strong>of</strong> LM status are<br />

listed in the table. The median OS time in LM pts was 14.4 compared to<br />

22.6 months, with a hazard ratio (HR) 1.4. The HR for treatment effect<br />

(DP�B vs. DP) for LM was not statistically significant for either group.<br />

Conclusions: Compared to pts without LM, mCRPC with LM are characterized<br />

by higher LDH and ALK and have a poor OS despite having similar PFS<br />

and objectivebiochemical response to docetaxel based therapy.<br />

<strong>Clinical</strong> outcomes Liver mets at baseline<br />

No<br />

Yes<br />

HR*<br />

(n�986) (n�59) (95% CI) p value<br />

Median OS (months) 22.6<br />

14.4<br />

1.4 0.019*<br />

(95% CI )<br />

(21.5-24.1) (13.3-17.8) (1.1-1.8)<br />

Median PFS (months) 7.6<br />

5.7<br />

1.1 0.672*<br />

(95% CI)<br />

(7.2-8.1) (2.5-7.1) (0.7-1.7)<br />

>50% decline in PSA 64%<br />

55%<br />

0.216<br />

(95% CI)<br />

(60-67) (42-69)<br />

Objective response<br />

41%<br />

53%<br />

0.097<br />

(95% CI)<br />

(37-46) (39-67)<br />

* stratified log-rank p value<br />

4657 General Poster Session (Board #11H), Sun, 8:00 AM-12:00 PM<br />

A national survey <strong>of</strong> radiation oncologists and urologists on active surveillance<br />

for low-risk prostate cancer. Presenting Author: Simon P. Kim, Mayo<br />

Clinic, Rochester, MN<br />

Background: While active surveillance (AS) is well recognized as an<br />

acceptable treatment strategy for low-risk prostate cancer (PC), the extent<br />

to which radiation oncologists and urologists perceive AS as effective and<br />

routinely recommend it to patients is unknown. Therefore, we sought to<br />

assess the attitudes and treatment recommendations for low-risk PC from a<br />

national survey <strong>of</strong> PC specialists. Methods: A mail survey was sent to a<br />

population-based sample <strong>of</strong> 1,439 physicians in the U.S. from late 2011<br />

and early 2012. Physicians were queried about their attitudes regarding AS<br />

and treatment recommendations for patients diagnosed with low-risk PC<br />

(PSA�10 ng/dl; T1c; Gleason 6 in one <strong>of</strong> twelve cores). Pearson Chi-square<br />

and multivariate logistic regression were used to test for differences in<br />

attitudes and treatment recommendations by physician demographics,<br />

compensation structure, primary place <strong>of</strong> employment, and specialty.<br />

Results: Overall, 321 radiation oncologists and 322 urologists completed<br />

the survey for a 45% response rate. Most physicians reported that AS is<br />

effective for low-risk PC (71%) and stated that they were comfortable<br />

routinely recommending AS (67%). Urologists were more likely to agree<br />

that AS is effective (77% vs. 67%; p�0.005) and were comfortable<br />

recommending AS (74% vs. 61%; p�0.001) compared with radiation<br />

oncologists. Most physicians recommended radical prostatectomy (47%)<br />

or radiation therapy (32%), but fewer endorsed AS (21%) for low-risk<br />

disease. After adjusting for physician covariates, radiation oncologists were<br />

more likely to recommend radiation therapy (OR: 10.97; p�0.001), while<br />

urologists were more likely to recommend surgery (OR: 4.69; p�0.001)<br />

and AS (OR: 2.18; p�0.001) for low-risk PC. Conclusions: Although AS is<br />

widely viewed as effective by both radiation oncologists and urologists,<br />

most urologists continue to recommend surgery, while most radiation<br />

oncologists recommend radiation therapy. Our results may explain in part<br />

the relatively low contemporary use <strong>of</strong> AS in the U.S.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


316s Genitourinary Cancer<br />

4658 General Poster Session (Board #12A), Sun, 8:00 AM-12:00 PM<br />

Prostate-specific antigen (PSA) screening in the United States: Patterns <strong>of</strong><br />

use and PSA outcomes in screened versus unscreened men. Presenting<br />

Author: Ian M. Allen, UNC-CH School <strong>of</strong> Medicine, Chapel Hill, NC<br />

Background: PSA screening is a subject <strong>of</strong> substantial controversy. We<br />

examined patterns <strong>of</strong> PSA screening using a recent cohort from the<br />

population-based National Health and Nutrition Examination Survey<br />

(NHANES) study and compared PSA levels <strong>of</strong> previously screened vs. never<br />

screened men. Methods: NHANES is a cross-sectional study which collects<br />

health-related information (including cancer screening history, family<br />

history and socioeconomic variables) and blood samples from nationally<br />

representative samples <strong>of</strong> the US population. We included 2,078 previously<br />

screened men and 1,902 never screened men surveyed from 2003-8.<br />

Statistical analysis accounted for sampling weights. Results: 25% <strong>of</strong> men<br />

age 40-49 years had prior PSA screening; 56% age 50-59, and 72% age<br />

60-69. Screening rates were higher for men with family history (68% vs.<br />

50% no history, p�.001), health insurance (58% vs. 21% no insurance,<br />

p�.001), and Caucasians (59% vs. 44% non-Caucasian, p�.001). No<br />

significant differences were seen in the PSA values <strong>of</strong> screened vs. never<br />

screened men stratified by age (Table), or by race/ethnicity, insurance<br />

status, or family history. Conclusions: Rates <strong>of</strong> PSA screening in the US<br />

differ by age, family history, race/ethnicity and insurance status. However,<br />

no significant differences were seen in PSA values <strong>of</strong> screened vs.<br />

unscreened men, and very few patients under 60 years <strong>of</strong> age had PSA<br />

values �10. Despite questions about the appropriate role <strong>of</strong> PSA testing,<br />

population-based prostate cancer screening is routinely conducted among<br />

Caucasian and non-Caucasian men 50 years <strong>of</strong> age and older.<br />

PSA in screened versus unscreened individuals stratified by age.<br />

Screened (%) Unscreened (%) p value<br />

Age 49-49<br />

PSA 0-3.99 98.9 98.7 0.927<br />

PSA 4-9.99 0.7 0.9<br />

PSA >10 0.3 0.3<br />

Age 50-59<br />

PSA 0-3.99 92.9 94.6 0.066<br />

PSA 4-9.99 6.4 4.1<br />

PSA >10 0.6 1.2<br />

Age 60-69<br />

PSA 0-3.99 81.9 83.3 0.136<br />

PSA 4-9.99 15.7 12.2<br />

PSA >10 2.3 4.4<br />

4660 General Poster Session (Board #12C), Sun, 8:00 AM-12:00 PM<br />

A first-in-human phase I imaging study using hyperpolarized 1c-13 pyruvate<br />

(h-Py) in patients (pts) with localized prostate cancer (l-PCa).<br />

Presenting Author: Andrea Lynne Harzstark, University <strong>of</strong> California, San<br />

Francisco, San Francisco, CA<br />

Background: Preclinical studies demonstrated that the conversion <strong>of</strong> h-Py to<br />

hyperpolarized 13C lactate (h-lac) is detectable on MRI-spectroscopy and is<br />

a useful marker <strong>of</strong> differentiation in PCa. H-Py MRI provides more than<br />

10,000-fold enhancement in signal to noise ratio (SNR), allowing for rapid<br />

detection <strong>of</strong> metabolic alterations in vivo. Hyperpolarized compounds have<br />

not been previously studied in man. Methods: Pts with biopsy-proven<br />

untreated l-PCa were enrolled in a phase I study <strong>of</strong> h-Py MRI. Following a<br />

modified 3�3 design, 6 pts were enrolled at each dose level (0.14, 0.28<br />

and 0.43 mL/Kg): 3 to monitor kinetics <strong>of</strong> h-Py, and 3 to evaluate the<br />

spatial distribution <strong>of</strong> metabolism in PCa and normal prostate (nl-P). An<br />

expansion cohort <strong>of</strong> 15 pts explored the biological variability <strong>of</strong> metabolism.<br />

A dynamic nuclear polarization (DNP) system, the first human system<br />

anywhere, generated and delivered 230 mM sterile h-Py. IV injection <strong>of</strong><br />

h-Py was followed by imaging with a 3T MR scanner with custom transmit<br />

and receive coils. Monitoring included EKG, vital signs, and laboratory<br />

testing. Results: 31 pts were imaged. 23 pts had Gleason (G) 6, 6 pts G7,<br />

and 2 pts G8 PCa. Median age was 63 years (range 45-75); median PSA<br />

was 5.9 ng/mL (1.88-20.2). No dose limiting toxicities or �grade (gr) 2<br />

toxicity was observed. Toxicity included: gr 1dysgeusia (6 pts), gr 1<br />

hypokalemia, gr 1 hypocalcemia, gr 1 dizziness, and gr 2 diarrhea (1 pt<br />

each). Median time from dissolution <strong>of</strong> the agent to delivery into patients<br />

was 66 seconds (43-88). Signals from h-Py and h-Lac were seen in PCa<br />

and nl-P at all doses; 0.43 mL/Kg showed the best SNR and discrimination<br />

between PCa and nl-P and was therefore established as the phase II dose.<br />

There appeared to be an association between h-Lac levels and PCa grade.<br />

Conclusions: H-py metabolic imaging has minimal toxicity and provides the<br />

ability to discriminate Ca from nl-P based on increased levels <strong>of</strong> h-lac. The<br />

correlation with grade and changes with therapy require further study.<br />

4659 General Poster Session (Board #12B), Sun, 8:00 AM-12:00 PM<br />

Prevalence <strong>of</strong> nonmetastatic (M0) prostate cancer (PC) patients on continuous<br />

androgen deprivation therapy (ADT) in the United States. Presenting<br />

Author: Karynsa Cetin, Center for Observational Research, Amgen Inc,<br />

Thousand Oaks, CA<br />

Background: ADT is the cornerstone treatment <strong>of</strong> metastatic PC, but the<br />

nature and extent <strong>of</strong> its use in the M0 setting is less well-described. We<br />

sought to estimate the current prevalence <strong>of</strong> M0 PC patients actively<br />

receiving continuous ADT (�6 months) in the US. Methods: Two pointprevalent<br />

cohorts on 12/31/2008 with continuous insurance coverage in<br />

2008 were assembled: men aged 45-64 years (yrs) enrolled in commercial<br />

health plans (MarketScan) and men aged �67 yrs enrolled in fee-forservice<br />

(FFS) Medicare (Medicare 5% sample). Among those with evidence<br />

<strong>of</strong> PC and no evidence <strong>of</strong> metastases, we selected men who had continuous<br />

exposure to gonadotropin-releasing hormone agonists during at least the<br />

last 6 months <strong>of</strong> 2008 or received bilateral orchiectomy prior to 7/1/2008.<br />

The number <strong>of</strong> prevalent ADT users was extrapolated to the entire national<br />

commercially insured population aged 45-64 yrs and to the entire Medicare<br />

FFS population aged �65 yrs using person-level weights. Applying agespecific<br />

prevalence estimates to the US Census population on 12/31/<br />

2008, we estimated the number <strong>of</strong> prevalent ADT users in the total US<br />

male population aged �45 yrs. Results: An estimated 11,935 (95%<br />

confidence interval [CI]: 11,310-12,561) commercially insured men aged<br />

45-64 yrs and 115,468 (95% CI: 112,304-118,633) Medicare FFS men<br />

aged �65 yrs were M0 PC patients actively receiving continuous ADT for<br />

�6 months on 12/31/2008. Extrapolated to the total US male population<br />

aged �45 yrs, this estimate was 188,916 (95% CI: 184,104-193,727).<br />

Age-specific prevalence (N [95% CI]) on 12/31/2008 is presented in the<br />

table. Conclusions: We projected nearly 190,000 US men with M0 PC were<br />

actively receiving continuous ADT for �6 months at the end <strong>of</strong> 2008, and<br />

the vast majority (91%) <strong>of</strong> these men were aged �65 yrs. Additional work<br />

will address timing <strong>of</strong> initiation, duration, and other aspects <strong>of</strong> ADT use in<br />

this large population <strong>of</strong> M0 PC patients.<br />

Age, yrs Commercial and FFS Medicare males US males<br />

45-64 11,935 (11,310-12,561) 17,163 (16,264-18,062)<br />

65-74 28,087 (26,272-29,902) 43,851 (41,017-46,686)<br />

75-84 51,541 (49,549-53,532) 76,157 (73,214-79,100)<br />

>85 35,841 (34,195-37,487) 51,745 (49,368-54,121)<br />

4661 General Poster Session (Board #12D), Sun, 8:00 AM-12:00 PM<br />

Correlation <strong>of</strong> UHRF1 expression in primary prostate cancer patients with<br />

adverse prognosis. Presenting Author: Enrico Roggero, IOSI International<br />

Prostate Research Group, Bellinzona, Switzerland<br />

Background: Cancer <strong>of</strong> the prostate is a leading cause <strong>of</strong> cancer death in<br />

western countries. There is a great need to understand the clinical course <strong>of</strong><br />

the disease and to distinguish the indolent tumors from those with<br />

aggressive behavior. Epigenetic mechanisms play an essential role in<br />

cancer initiation and progression. Our groups have recently provided<br />

evidence that over-expression <strong>of</strong> UHFR1 (ubiquitin-like with PHD and ring<br />

finger domain) leads to prostate cancer progression by activating a robust<br />

epigenetic switch involving silencing <strong>of</strong> a network <strong>of</strong> tumor suppressor<br />

genes. The purpose <strong>of</strong> this study was to evaluate in a clinical setting the<br />

prognostic impact <strong>of</strong> UHFR1 expression. Methods: In a series <strong>of</strong> 225<br />

eligible patients (median age 63 years, range 44-75) with prostate cancer<br />

treated in a single institution with prostatectomy between 1990 and 1999<br />

we evaluated the tumor nuclear expression <strong>of</strong> UHRF1 by immunohistochemistry<br />

(IHC). <strong>Clinical</strong> and histological data <strong>of</strong> the series were also reviewed.<br />

The UHFR1 expression (evaluated in tissue microarrays by immunohistochemistry)<br />

and prognostic factors were analyzed using univariate analyses<br />

and multivariate logistic regression analysis to identify association with<br />

overall survival (OS). Results: The median FU for the series was 137 months<br />

(range 1-229), eighty-one patients died (median FU 85 months). In<br />

Ninety-seven patients (43%) the UHRF1 expression was positive. In<br />

univariate analyses Gleason Score (�7 vs 7-9), Stage Risk Group (TNM<br />

Stage �III vs Stage III and/or N�) and UHFR1 expression (negative vs<br />

positive) were significant prognostic factors for OS with p-value �0.0001.<br />

In multivariate analyses Gleason Score, Stage Risk Group and UHFR1<br />

expression were independent predictors for OS with respectively HRs <strong>of</strong><br />

2.77 (95% CI 1.72-4.46) p�0.0001, HRs <strong>of</strong> 1.96 (95% CI 1.14-3.37)<br />

p�0.014 and Hrs 1.35 (95% CI 1.02-1.78) p�0.030. Conclusions: Our<br />

results indicate that expression <strong>of</strong> UHFR1 protein, independently from<br />

historical prognostic factors, is linked with adverse prognosis for overall<br />

survival in a homogeneous primary prostate cancer treated group with<br />

long-term follow-up.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


4662 General Poster Session (Board #12E), Sun, 8:00 AM-12:00 PM<br />

Prostate-specific membrane antigen antibody drug conjugate (PSMA ADC):<br />

A phase I trial in metastatic castration-resistant prostate cancer (mCRPC)<br />

previously treated with a taxane. Presenting Author: Anthony E. Mega,<br />

Brown University Oncology Group, Providence, RI<br />

Background: The abundant expression <strong>of</strong> prostate specific membrane<br />

antigen (PSMA) type II transmembrane glycoprotein on prostate cancer<br />

cells provides a rationale for antibody therapy. PSMA ADC, a fully<br />

humanized antibody to PSMA linked to the potent antitubulin agent<br />

monomethyl auristatin E (MMAE), binds PSMA and is internalized within<br />

the prostate cancer cell where cleavage by lysosomal enzymes and releases<br />

free MMAE, causing cell cycle arrest and apoptosis. We report results from<br />

an ongoing phase 1 dose escalation study <strong>of</strong> PSMA ADC in subjects with<br />

taxane-refractory mCRPC. Methods: Eligibility requirements include progressive<br />

mCRPC following taxane-containing chemotherapy and ECOG status <strong>of</strong><br />

0 or 1. PSMA ADC was administered by IV infusion Q3W for up to 4 cycles.<br />

Adverse events, pharmacokinetics (PK), PSA, circulating tumor cells,<br />

clinical disease progression and immunogenic response to PSMA ADC were<br />

assessed. Serum PSMA ADC and total antibody were measured by ELISA,<br />

and free MMAE was measured by LC/MS/MS.The dosing cohorts range from<br />

0.4 mg/kg to 2.8 mg/kg, with dose escalation continuing. Results: 40<br />

subjects have been dosed in nine dose levels (0.4, 0.7, 1.1, 1.6, 1.8, 2.0,<br />

2.2, 2.5, 2.8 mg/kg). To date, PSMA ADC has been well tolerated with the<br />

most commonly seen adverse events being anorexia and nausea, and the<br />

most common laboratory abnormalities being reversible hematologic parameters<br />

and liver function tests. Antitumor activity has been manifested as<br />

reductions either in PSA or circulating tumor cells in the higher dose<br />

cohorts. Exposure to PSMA ADC increased with dose and was ~1,000-fold<br />

greater than MMAE exposure. Similar PK metrics were observed after the<br />

first and third doses. Dosing at the 2.8 mg/kg cohort is continuing and an<br />

MTD has not yet been reached. Conclusions: PSMA ADC is generally well<br />

tolerated in subjects with mCRPC, previously treated with taxane in doses<br />

up to 2.8 mg/kg. Antitumor activity at higher dose levels has been observed.<br />

The MTD has not yet been reached and enrollment is ongoing at higher dose<br />

levels.<br />

4664 General Poster Session (Board #12G), Sun, 8:00 AM-12:00 PM<br />

Response to ketoconazole (keto) or docetaxel (D) following clinical progression<br />

on abiraterone acetate (AA) in castrate-resistant prostate cancer<br />

(CRPC). Presenting Author: Rahul Raj Aggarwal, University <strong>of</strong> California,<br />

San Francisco, San Francisco, CA<br />

Background: Patients (pts) with CRPC treated with keto <strong>of</strong>ten experience a<br />

rise in serum adrenal androgen levels upon progression and may subsequently<br />

respond to more potent adrenal suppression with AA. In contrast,<br />

pts treated with AA do not have a rise in serum androgen levels upon<br />

progression, and thus may not respond to further androgen synthesis<br />

inhibition with keto but require other treatment modalities such as<br />

chemotherapy instead. The goal <strong>of</strong> this analysis is to report the outcomes <strong>of</strong><br />

pts with CRPC progressing on AA subsequently treated with either keto or<br />

D. Methods: Pts with chemotherapy-naïve CRPC were treated on previously<br />

reported phase 1 and 2 trials <strong>of</strong> AA, with doses ranging from 250 to 1000<br />

mg/day. Subsequent treatment following progression on AA was per<br />

individual investigator discretion, including treatment with either keto or D.<br />

Results: To date, following disease progression on AA, 6 and 14 pts have<br />

been subsequently treated with either keto or D respectively. Of the 14 pts<br />

treated with D, 11 (79%) had exposure to keto prior to AA. The keto- and<br />

D-treated cohorts were similar with respect to other baseline factors,<br />

including prior response to AA (4/6 (67%) pts with � 50% PSA decline on<br />

AA in keto cohort; 10/14 (71%) pts in D cohort); though pts treated with D<br />

were more likely to require opioid analgesics (0 pts in keto cohort; 7 (50%)<br />

in D). For pts treated with keto, none experienced a decline in PSA,<br />

improvement in bone scan, or an objective response; all had disease<br />

progression by 12 weeks. In contrast, for pts treated with D, 10 (71%) had<br />

a decline in PSA; 6 (43%) with a � 50% maximum PSA decline. Of 6 pts<br />

with measurable disease, 2 (33%) had an objective response. The median<br />

time to progression for the D cohort was 129 days (range 60 – 294).<br />

Conclusions: Although the cohort size is small, and post-AA therapy was not<br />

randomized, these data provide preliminary support for the hypothesis that<br />

CRPC progressing on AA is cross-resistant to further androgen synthesis<br />

inhibition with keto. Contrary to other reports, prior treatment with AA does<br />

not appear to decrease the likelihood <strong>of</strong> subsequent response to D, but this<br />

observation requires prospective validation.<br />

Genitourinary Cancer<br />

4663 General Poster Session (Board #12F), Sun, 8:00 AM-12:00 PM<br />

Results <strong>of</strong> telomerase activity measurements from live circulating tumor<br />

cells captured on a slot micr<strong>of</strong>ilter in a phase III SWOG-coordinated<br />

prostate cancer trial (S0421). Presenting Author: Amir Goldkorn, University<br />

<strong>of</strong> Southern California Norris Comprehensive Cancer Center, Los<br />

Angeles, CA<br />

Background: Analysis <strong>of</strong> circulating tumor cells (CTC) is a promising<br />

biomarker strategy in advanced prostate cancer, and telomerase activity<br />

(TA) is a recognized cancer marker. To test whether CTC TA is prognostic for<br />

survival (OS), we developed a novel Parylene-C slot micr<strong>of</strong>ilter capable <strong>of</strong><br />

capturing live CTC and used it to measure CTC TA as part <strong>of</strong> a Phase III<br />

SWOG-coordinated therapeutic trial in metastatic castration resistant<br />

prostate cancer (S0421). Methods: Blood samples were drawn into EDTA<br />

tubes and shipped overnight to a central processing site. After Ficoll<br />

centrifugation, low constant pressure was used to pass the mononuclear<br />

cell layer through two slot micr<strong>of</strong>ilters in series as published previously<br />

(filter1 captures CTC � background white blood cells; filter2 captures only<br />

background white blood cells). Filter-trapped cells were lysed in CHAPS<br />

buffer and assayed for TA using qPCR-based telomeric repeat amplification.<br />

In parallel, CTC were enumerated using CellSearch (J&J). Cox<br />

regression was used to evaluate the association between baseline (pretreatment)<br />

TA and OS overall, and within subgroups characterized by good<br />

prognosis (�5) vs. poor prognosis (��5) baseline CTC counts. CART<br />

regression was used to explore potential prognostic subgroups based on<br />

baseline PSA, CTC, and TA cutpoints. Results: Samples were obtained from<br />

263 patients. While no association was observed between TA and OS<br />

overall, in patients with baseline CTC ��5 (108 <strong>of</strong> 263 or 41% <strong>of</strong><br />

patients), TAfilter2 –TAfilter1 representing high CTC TA relative to background<br />

blood cells was associated with a hazard ratio (HR) <strong>of</strong> 1.14 (95% CI<br />

1.05-1.23, p�0.001) for OS after adjusting for risk factors and remained<br />

significant when also adjusting for CTC: HR 1.14 (95% CI 1.04-1.23;<br />

p�0.005). Exploratory CART regression assessing baseline PSA, CTC, and<br />

TA identified risk groups based only on CTC and TA values. Conclusions:<br />

Baseline TA from CTC live-captured on a new slot micr<strong>of</strong>ilter is the first CTC<br />

biomarker shown to be prognostic <strong>of</strong> OS in men with CTC counts ��5 ina<br />

prospective clinical trial. CTC TA may be useful for further identifying<br />

prognostic groups in this population.<br />

4665 General Poster Session (Board #12H), Sun, 8:00 AM-12:00 PM<br />

Phase I clinical trial <strong>of</strong> galeterone (TOK-001), a multifunctional antiandrogen<br />

and CYP17 inhibitor in castration resistant prostate cancer (CRPC).<br />

Presenting Author: Robert B. Montgomery, University <strong>of</strong> Washington,<br />

Seattle, WA<br />

Background: Galeterone is an oral steroid analog that suppresses prostate<br />

cancer growth by inhibiting CYP17, blocking androgen receptor and<br />

reducing androgen receptor levels. Methods: Open label, multicenter<br />

dose-finding study assessing the safety, pharmacokinetics and clinical<br />

effect <strong>of</strong> escalating doses <strong>of</strong> galeterone in chemotherapy naïve CRPC<br />

patients (pts). Pts were enrolled in cohorts from 650-2,600mg <strong>of</strong> galeterone<br />

daily for 12 wks. Study also explored effects <strong>of</strong> food supplement and<br />

scheduling. Pts remained on study until disease progression or dose<br />

limiting toxicity. Results: Pt characteristics included median age 69<br />

(47-92), median PSA 24 (6-200), and 46.9% with metastases. 36 <strong>of</strong> 49<br />

pts completed 12 wks <strong>of</strong> study with early discontinuation for toxicity (6),<br />

progression (5), or withdrawal <strong>of</strong> consent (2). Maximal tolerated dose was<br />

not reached. The frequency <strong>of</strong> AEs was 58% grade 1, 30% grade 2, 8%<br />

grade 3 and 1% grade 4. Transient LFT elevations were seen in 15 men (5<br />

with suspected or confirmed Gilberts). There was no trend for increasing<br />

toxicity with dose escalation and no PK difference in Gilberts pts. 9 SAEs<br />

were reported with one considered related to galeterone (rhabdomyolysis<br />

with acute renal failure in the context <strong>of</strong> high dose statin use). Overall<br />

11/49 pts (22%) demonstrated �50% PSA decline and an additional<br />

13/49 (26%) had 30-50% declines. <strong>Part</strong>ial response by RECIST was seen<br />

in 2 pts. Consistent with lyase inhibition, increased corticosteroids and<br />

suppressed androgens were seen with dose escalation. Analysis <strong>of</strong> limited<br />

plasma collected 4 - 30 hours after dosing showed high interpatient<br />

variability with no consistent relationship to dose or time after dosing.<br />

Conclusions: Galeterone was well tolerated in CRPC pts and demonstrated<br />

clinical activity. Galeterone is being reformulated with additional PK and<br />

phase II trials planned.<br />

PSA response to galeterone.<br />

Dose (mg/day) N<br />

Duration on<br />

treatment<br />

(months)<br />

>50% PSA<br />

decline<br />

% (n)<br />

317s<br />

>30% PSA<br />

decline<br />

% (n)<br />

650 6* 2-16 20% (1) 20% (1)<br />

975 with and w/out supplement 12* 1-21 18% (2) 55% (6)<br />

1,300 6 2-21 0% (0) 17% (1)<br />

1,950 single/split 13 1-12 23% (3) 54% (7)<br />

2,600 single/split<br />

*One pt nonevaluable.<br />

12 1-8 42% (5) 75% (9)<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


318s Genitourinary Cancer<br />

4666 General Poster Session (Board #13A), Sun, 8:00 AM-12:00 PM<br />

Regional variation in survival after metastatic prostate cancer diagnosis.<br />

Presenting Author: Darius Lakdawalla, University <strong>of</strong> Southern California,<br />

Los Angeles, CA<br />

Background: Survival after diagnosis <strong>of</strong> metastatic prostate cancer (mPC)<br />

averages 2-3 years. Substantial regional differences in survival have been<br />

documented for PC prior to 2000. We investigated the extent to which<br />

regional variation exists in survival for mPC patients during 2000-2007.<br />

Methods: Using the Surveillance, Epidemiology, and End Results (SEER)<br />

database linked to Medicare claims, we identified men (mean age, 77.6<br />

years) continuously enrolled in Medicare <strong>Part</strong>s A/B who were diagnosed<br />

with mPC between 2000 and 2007 and not diagnosed with another<br />

malignancy. The base-case model was limited to hospital service areas with<br />

�50 patients. A Cox proportional hazards model was used to estimate<br />

hazard ratios (HR) for overall survival (OS), adjusting for year <strong>of</strong> diagnosis,<br />

age, marital status, poverty and education covariates, Gleason score,<br />

comorbidities, and region covariates. Sensitivity <strong>of</strong> results was tested by<br />

limiting the analysis to patients surviving at least 6 months after diagnosis<br />

and by removing regional sample size limits. We report HRs for covariates,<br />

results <strong>of</strong> a Wald test <strong>of</strong> joint significance for region effects, and percentage<br />

difference from the mean for each region’s HR for death. Results: A total <strong>of</strong><br />

2696 patients with mPC met the inclusion criteria. OS was 37% at 3 years<br />

and mean HR for death was 4.8 (sd 2.1). HRs for death were positively<br />

correlated with age (HR�1.96, 95% CI: 1.6-2.3 for �80 years), PCspecific<br />

comorbidity index (HR�1.2, 95% CI: 1.1-1.3), and Gleason score<br />

(HR�6.6, 95% CI: 2.4-17.8 for poorly differentiated). Year <strong>of</strong> diagnosis,<br />

race, and socioeconomic status were not significantly associated with<br />

mortality. Wald test <strong>of</strong> joint significance for survival across regions <strong>of</strong><br />

P�.019 indicated significant differences in survival across regions and was<br />

robust in sensitivity analyses. Patients living in regions with the worst<br />

survival rates had HRs for death that were 20% higher than the mean<br />

(HR�5.8) and those living in regions with the best survival rates had HRs<br />

30% lower than the mean (HR�3.4). Conclusions: There are significant<br />

regional differences in survival for mPC patients in the 2000s. Further<br />

research is needed to determine if treatment differences play a role in this<br />

disparity.<br />

4668 General Poster Session (Board #13C), Sun, 8:00 AM-12:00 PM<br />

Incidence-based analysis <strong>of</strong> lethal prostate cancer in Swedish counties<br />

with high versus low incidence <strong>of</strong> prostate cancer in nationwide, populationbased<br />

registries. Presenting Author: Pär Stattin, Department <strong>of</strong> Surgery,<br />

Urology Service, Memorial Sloan-Kettering Cancer Center, New York, NY<br />

Background: Current data from randomized trials differ in the ratio <strong>of</strong><br />

benefit to harms from screening for prostate cancer (PC) using prostate<br />

specific antigen (PSA) testing and it is further unclear if decreases in PC<br />

mortality in the US can be attributed to introduction <strong>of</strong> screening or to<br />

changes in treatment. The aim <strong>of</strong> this study was to investigate if rapidly<br />

increased incidence <strong>of</strong> PC diagnosis due to early and rapidly increased rate<br />

<strong>of</strong> PSA testing, is associated with decreased incidence <strong>of</strong> lethal PC.<br />

Methods: In this incidence-based study using highly representative data<br />

from nation-wide population-based registries we analysed the incidence <strong>of</strong><br />

lethal prostate cancer in eight Swedish counties with the most rapid and<br />

largest increased incidence <strong>of</strong> PC diagnosis (high incidence counties) from<br />

1995 through 2002, and six counties with the smallest increased PC<br />

incidence (low incidence counties). We analysed the rate ratio (RR) in high<br />

vs. low incidence counties <strong>of</strong> cases with metastatic PC at diagnosis,<br />

(positive bone scan or PSA above 100 ng/ml), PC-specific mortality, and<br />

excess mortality (death among PC cases regardless <strong>of</strong> cause). <strong>Part</strong>icipants<br />

were men aged 50-74 years during 2000 to 2009 in the high versus low<br />

incidence counties contributing 4,528,134 versus 2,471,373 personyears<br />

at risk, respectively. There were 33,780 incident PC diagnoses and<br />

1,577 PC deaths in high incidence counties and 16,377 PC cases and 985<br />

PC deaths in low incidence counties. Results: In high vs. low incidence<br />

counties, RR <strong>of</strong> metastatic PC was 0.85 (95% confidence interval [CI] �<br />

0.79 to 0.92), RR <strong>of</strong> PC-specific mortality was 0.87 (95% CI � 0.81 to<br />

0.95) and RR <strong>of</strong> excess mortality in men with PC was 0.75 (95 %CI � 0.66<br />

to 0.86). Conclusions: Our study provides data on a population-level<br />

indicating a decreased incidence <strong>of</strong> lethal PC in areas with rapid and large<br />

increase in incident PC diagnosis due to increased rates <strong>of</strong> PSA testing,<br />

early diagnosis, and treatment with curative intent.<br />

4667 General Poster Session (Board #13B), Sun, 8:00 AM-12:00 PM<br />

The impact <strong>of</strong> prior therapy with ketoconazole (keto) on clinical outcomes<br />

after subsequent docetaxel treatment in metastatic castration-resistant<br />

prostate cancer (mCRPC) patients: Results from a randomized phase III<br />

trial (CALGB 90401). Presenting Author: Eric Jay Small, University <strong>of</strong><br />

California, San Francisco, San Francisco, CA<br />

Background: Small retrospective reviews have suggested the possibility <strong>of</strong> a<br />

decreased benefit <strong>of</strong> docetaxel in mCRPC patients (pts) previously treated<br />

with abiraterone acetate, a novel androgen synthesis inhibitor (ASI).<br />

CALGB 90401 was an intergroup (CALGB, ECOG) randomized phase 3 trial<br />

in which 1050 mCRPC pts were treated with docetaxel, which <strong>of</strong>fered the<br />

opportunity to evaluate the effect <strong>of</strong> prior treatment with keto, an earlier<br />

generation ASI, on clinical outcomes following docetaxel treatment in<br />

mCRPC pts. Methods: Data from 1,050 men randomized on CALGB 90401<br />

to treatment with docetaxel and prednisone with either bevacizumab or<br />

placebo were used. Pts were chemotherapy naïve, had evidence <strong>of</strong><br />

progressive mCRPC, an ECOG performance status � 2, and adequate bone<br />

marrow, hepatic and renal function. Randomization was stratified by age,<br />

prior history <strong>of</strong> a thromboembolic event, and 24-month predicted survival<br />

using the CALGB nomogram. The effect <strong>of</strong> prior keto use on overall survival<br />

(OS), progression-free survival (PFS), PSA decline, and objective response<br />

rate (RR) was estimated using proportional hazards and logistic regression<br />

models. Results: 277/1050 pts (26%) had received prior keto. Baseline<br />

characteristics including the 3 stratification factors, race, performance<br />

status, measurable disease, and baseline alkaline phosphatase and Hgb<br />

were balanced between prior keto treated pts and keto untreated pts. LDH<br />

and PSA were higher in the keto treated patients (211 vs. 201, p � 0.01;<br />

and 121 vs. 73, p � .0001; respectively.) There were no statistically<br />

significant differences in keto vs. non-keto treated patients with regards to<br />

OS (21.1 vs. 22.3 m, p � 0.643); PFS (8.1 vs. 8.6m p � 0.394 ); �50%<br />

decline in PSA (61% vs. 66% p�.112); or objective response (39% vs.<br />

43% p � 0.34). Conclusions: As measured by OS, PFS, PSA and RR, prior<br />

treatment with the ASI ketoconazole has no impact on the subsequent<br />

clinical impact <strong>of</strong> docetaxel in mCRPC pts.<br />

4669 General Poster Session (Board #13D), Sun, 8:00 AM-12:00 PM<br />

Association <strong>of</strong> decreased expression <strong>of</strong> protein phosphatase 2A subunit<br />

PR55� (PPP2R2C) with an increased risk <strong>of</strong> metastases and prostate<br />

cancer-specific mortality. Presenting Author: Elysia Sophia Spencer, Fred<br />

Hutchinson Cancer Research Center, Seattle, WA<br />

Background: The protein phosphatase 2A (PP2A) holoenzyme complex<br />

negatively controls cell growth, differentiation and apoptosis. Down regulation<br />

<strong>of</strong> PP2A activity is associated with oncogenic transformation <strong>of</strong><br />

papillomas and is a target for the small-T viral oncogene. The PR55�<br />

(2R2C) subunit regulates PP2A target specificity, inhibiting c-Src activity<br />

in some cell lines. c-Src activity enhances growth and invasion in prostate<br />

cancer. This study evaluates whether PPP2R2C protein levels in radical<br />

prostatectomy (RP) specimens were predictive for the development <strong>of</strong><br />

metastases or prostate-cancer specific mortality (PCSM). Methods: From<br />

1954-1997, 1004 consecutive patients underwent RP at Virginia Mason<br />

Medical Center in Seattle. Metastases were confirmed radiographically or<br />

by tissue diagnosis. Death certificates confirmed PCSM. Patients without<br />

stored tissue were excluded. 34 patients with metastases or PCSM were<br />

matched to patients without recurrence (NR) at a 1:2 ratio. Paraffinembedded<br />

tissue was stained with anti-PPP2R2C monoclonal antibody,<br />

clone 6D1 (Abnova, Taiwan). A pathologist evaluated the percentage and<br />

intensity <strong>of</strong> nuclei stained using a 4-point scale. Statistical analysis was<br />

performed on SAS. Results: Mean follow up was 10.26 years. PPP2R2C was<br />

detected in 13/18 (72.2%) PCSM, 18/22 (81.8%) metastases and 45/49<br />

(91.8%) NR patients. Mean expression was lower in cancerous vs benign<br />

glands, 1.74 vs 2.04 (p � 0.001). Expression intensity from PCSM vs NR<br />

was 1.45 vs 1.80 (p � 0.041). On multivariate analysis, expression was<br />

lower in metastases or PCSM versus NR 1.79 vs 2.35 (p � 0.002),<br />

independent <strong>of</strong> pre-op PSA and Gleason grade. Conclusions: PPP2R2C loss<br />

was highly correlated with metastasis and PCSM. Patients with the lowest<br />

levels <strong>of</strong> expression were at the highest risk for PCSM. As tissue was<br />

obtained at time <strong>of</strong> surgery, this analysis reflects a truly prognostic<br />

biomarker, independent <strong>of</strong> Gleason grade and PSA. We have shown that<br />

PPP2R2C scoring is a useful metric to identify patients at high risk <strong>of</strong><br />

developing advanced disease. With further validation may allow for more<br />

accurate individual patient treatment plans and counseling.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


4670 General Poster Session (Board #13E), Sun, 8:00 AM-12:00 PM<br />

African <strong>American</strong> race to predict for earlier failure <strong>of</strong> active surveillance:<br />

Results from the Duke Prostate Center. Presenting Author: Mitchell<br />

Bassett, Duke University Medical Center, Durham, NC<br />

Background: As concerns mount regarding overtreatment and overdiagnosis<br />

<strong>of</strong> prostate cancer (CaP), active surveillance (AS) is increasingly<br />

utilized in low risk patients. While African-<strong>American</strong> (AA) race is associated<br />

with adverse outcomes after prostatectomy, its effect on patients managed<br />

with AS is not known. Methods: A retrospective review identified 222<br />

patients managed with AS at the Duke Prostate Center from January 2005<br />

to September 2011. All men had CaP diagnosed on biopsy performed at our<br />

center, and elected AS over treatment. Failure was defined as progression<br />

to treatment. In men who failed AS, the reasons for failure, follow-up PSA<br />

and biopsy characteristics were analyzed. The primary outcome - time from<br />

diagnosis to failure <strong>of</strong> AS for a reason other than patient choice - was<br />

analyzed with univariable and multivariable Cox proportional hazards<br />

models. Results: In our AS cohort, 73% are Caucasian and 23% AA. Median<br />

follow-up is 25.4 months. Age, household income, BMI, PSA, clinical<br />

stage, family history, prostate volume, number <strong>of</strong> cores with cancer, and<br />

Gleason grade on initial biopsy did not differ by race. The number <strong>of</strong><br />

biopsies and PSA tests performed on AS did not differ by race. A higher<br />

proportion <strong>of</strong> AA men tended to fail from biopsy progression (72.7% vs.<br />

63.8%) while a lower proportion failed by choice (9.1% vs. 14.9%)<br />

compared to Caucasians (p � 0.114). AA men had a significantly shorter<br />

time to failure (HR 1.74, p � 0.045) compared to Caucasians. There was a<br />

trend toward increased Gleason grade 8 or higher cancer on follow-up<br />

biopsy in AA compared to Caucasian men (10% vs. 2.5%, p � 0.08). AA<br />

race remained a predictor (HR 1.76, p � 0.058) <strong>of</strong> failure on multivariable<br />

analysis, as did initial PSA (HR 1.90, p � 0.031) and number <strong>of</strong> cores with<br />

cancer on initial biopsy (HR 1.29, p � 0.013). Conclusions: AA race was<br />

associated with higher risk for failure <strong>of</strong> AS. There was a trend toward AA<br />

men failing due to biopsy progression and with higher grade cancer.<br />

Additional follow-up is necessary to determine how this affects the long<br />

term outcomes <strong>of</strong> these men.<br />

TPS4672 General Poster Session (Board #13G), Sun, 8:00 AM-12:00 PM<br />

Phase II trial <strong>of</strong> cabazitaxel in patients with advanced or metastatic<br />

transitional cell carcinoma <strong>of</strong> the urothelial tract who have progressed<br />

within less than 12 months after cisplatin-based chemotherapy: A Spanish<br />

Oncology Genitourinary Group (SOGUG) study. Presenting Author: Jose<br />

Angel Arranz Arija, Hospital General Universitario Gregorio Marañón,<br />

Madrid, Spain<br />

Background: Advanced transitional cell carcinoma <strong>of</strong> the urothelium (TCCU)<br />

on progression after previous cisplatin-based combination is generally an<br />

incurable disease. The appropriate management <strong>of</strong> these patients is still an<br />

unmet need. Many drugs have shown modest or no activity in previous<br />

phase 2 trials. Population heterogeneity in these studies emerges as one <strong>of</strong><br />

the key determinants that could explain the variable outcomes. Recently, in<br />

a phase III study in this setting, prognostic factors (PF) for overall survival<br />

were identified (Bellmunt J et al, JCO 2010). Taxanes are active drugs in<br />

2nd-line metastatic TCCU. Cabazitaxel (C) is a semi-synthetic taxane that<br />

is a poor substrate for the multidrug resistance system. C could be a valid<br />

alternative in this patient population. Methods: This is an open label phase<br />

II study <strong>of</strong> C in patients (pts) with advanced or metastatic TCCU who have<br />

progressed within 12 months after receiving a 1st-line platinum based<br />

chemotherapy. There are three treatment arms as patients will be assigned<br />

to one <strong>of</strong> three groups previously defined based on the presence <strong>of</strong> 0, 1 or<br />

2-3 PFs as defined by Bellmunt et al. The activity <strong>of</strong> C will be assessed<br />

separately in each group and overall. The primary endpoint is response rate<br />

(RR) evaluated according to RECIST 1.1, a maximum <strong>of</strong> 35 pts are needed<br />

in each subgroup (maximum number <strong>of</strong> pts required: 105). Secondary<br />

objectives are RR in the whole population, toxicity, progression-free<br />

survival and overall survival. In addition, an external validation <strong>of</strong> the<br />

prognostic model proposed by Bellmunt will be conducted, as well as a<br />

pharmacogenomic study in order to better define the toxicity pr<strong>of</strong>ile <strong>of</strong> the<br />

drug and the potential responders.<br />

Genitourinary Cancer<br />

319s<br />

4671 General Poster Session (Board #13F), Sun, 8:00 AM-12:00 PM<br />

Activity <strong>of</strong> oral VT-464, a selective CYP17-lyase inhibitor, in the LNCaP<br />

prostate cancer xenograft. Presenting Author: William Douglas Figg,<br />

Medical Oncology Branch, National Cancer Institute, National Institutes <strong>of</strong><br />

Health, Bethesda, MD<br />

Background: With the FDA approval <strong>of</strong> abiraterone acetate, inhibition <strong>of</strong><br />

CYP17 (17� hydroxylase/C17, 20-lyase) is now a validated approach to the<br />

treatment <strong>of</strong> castration-resistant prostate cancer. VT-464 is a novel,<br />

selective CYP17-lyase inhibitor with decreased activity against CYP17<br />

hydroxylase (less mineralcocorticoid and glucocorticoid effects). The study<br />

objectives were to observe the effects <strong>of</strong> VT-464 in a prostate cancer<br />

xenograft model and to compare its activity to abiraterone acetate and<br />

surgical castration. Methods: SCID mice were implanted subcutaneously<br />

with LNCaP cells. When tumors reached 100 mm3 , mice were randomized<br />

to receive vehicle (0.5% CMC in saline, 5 mL/kg), VT-464 at 15, 50, or 100<br />

mg/kg p.o. b.i.d. A second cohort <strong>of</strong> LNCaP tumor-bearing mice received<br />

vehicle, surgical castration, or VT-464, or abiraterone acetate at 100 mg/kg<br />

p.o. b.i.d. for 28 days. Terminal blood and tumor concentrations were<br />

analyzed on day 28, four hours after the last dose. Results: In the first<br />

LNCaP xenograft cohort, percent growth inhibition (� S.E.) <strong>of</strong> 9.6 (�15.6),<br />

38.5 (�12.4), and 73.9 (�13.2) was observed on day 21 <strong>of</strong> treatment for<br />

VT-464 doses <strong>of</strong> 15, 50, and 100 mg/kg, respectively. Growth reduction at<br />

100 mg/kg was statistically significant compared to vehicle control from<br />

day 7 to 28. VT-464 was well tolerated with insignificant weight loss at all<br />

doses. In the second cohort, VT-464-treated (100 mg/kg) mice had<br />

significantly reduced tumor volumes on day 28 compared to control and<br />

abiraterone acetate (p�0.05, p�0.01, respectively). Reduction in tumor<br />

volumes were similar between VT-464-treated (100 mg/kg) and castrate<br />

animals. Plasma and tumor analyses revealed much greater plasma and<br />

tumor exposure <strong>of</strong> VT-464 compared to abiraterone acetate. Conclusions:<br />

VT-464 exhibited dose-dependent growth inhibition with significantly<br />

reduced tumor volumes at the highest dose compared to abiraterone<br />

acetate. The activity in VT-464-treated animals was similar to that <strong>of</strong><br />

castrate animals. These preclinical results show promising activity <strong>of</strong><br />

VT-464 in the treatment <strong>of</strong> prostate cancer.<br />

TPS4673 General Poster Session (Board #13H), Sun, 8:00 AM-12:00 PM<br />

Design <strong>of</strong> a phase II randomized, open-label trial <strong>of</strong> DN24-02, an<br />

autologous cellular immunotherapy targeting HER2/neu, in patients with<br />

surgically resected urothelial cancer at high risk <strong>of</strong> recurrence. Presenting<br />

Author: Dean F. Bajorin, Memorial Sloan-Kettering Cancer Center, New<br />

York, NY<br />

Background: Despite surgical resection and the use <strong>of</strong> (neo)adjuvant<br />

chemotherapy, up to 50% <strong>of</strong> patients with invasive urothelial carcinoma<br />

will have disease recurrence and eventually die <strong>of</strong> metastatic disease. New<br />

approaches are needed for these patients. Approximately 50% <strong>of</strong> patients<br />

with high-risk pathologic features have evidence <strong>of</strong> HER2 expression in the<br />

primary tumor or involved lymph nodes. DN24-02 is an autologous cellular<br />

immunotherapy targeting HER2, which is based on Dendreon’s Antigen<br />

Delivery Cassette technology – the same platform used for sipuleucel-T (an<br />

FDA-approved treatment for asymptomatic or minimally symptomatic<br />

metastatic castrate-resistant prostate cancer). Each dose <strong>of</strong> DN24-02 is<br />

manufactured by culture <strong>of</strong> peripheral blood mononuclear cells obtained by<br />

leukapheresis with a recombinant antigen that comprises components <strong>of</strong><br />

the intra- and extracellular domains <strong>of</strong> the HER2 antigen linked to GM-CSF.<br />

A product similar to DN24-02 has shown evidence <strong>of</strong> safety and immune<br />

response in phase I studies. Objectives: The primary objective is to<br />

determine whether DN24-02, given as adjuvant therapy following surgical<br />

resection, can prolong survival compared to standard <strong>of</strong> care (SOC).<br />

Secondary objectives are to evaluate disease-free survival, safety, and<br />

immune responses to DN24-02. Methods: Eligible patients will have<br />

undergone surgical resection <strong>of</strong> a primary urothelial cancer, with either<br />

�pT2 or pN� staging, and HER2 expression �1� by IHC based on<br />

pathologic review. Approximately 180 patients will be randomized (1:1) to<br />

receive three IV infusions <strong>of</strong> DN24-02 as adjuvant therapy approximately<br />

every 2 weeks, or SOC. Stratification will include 1) neoadjuvant chemotherapy<br />

vs surgery alone; and 2) positive (pN�) vs negative lymph node<br />

involvement (pN0). Other adjuvant chemotherapy will not be allowed.<br />

Patients can be treated per SOC if there is disease recurrence. Patients will<br />

be monitored for evidence <strong>of</strong> recurrence by imaging, and followed for<br />

survival. Cellular and serological immune responses will be assessed at<br />

baseline and post-DN24-02 therapy.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


320s Genitourinary Cancer<br />

TPS4674 General Poster Session (Board #14A), Sun, 8:00 AM-12:00 PM<br />

Vinflunine maintenance therapy versus best supportive care (BSC) after<br />

platinum combination in advanced bladder cancer: A phase II, randomized,<br />

open-label study (MAJA study)—SOGUG 2011-02. Presenting Author:<br />

Sonia Maciá, Hospital General de Elda, Elda, Spain<br />

Background: Vinflunine is a novel microtubule inhibitor currently approved<br />

by EMA as treatment after platinum progression, in metastasic bladder<br />

cancer. It is distinguished from the other vinca-alkaloids because it binds<br />

relatively weakly to tubulin, suggesting an improved tolerance pr<strong>of</strong>ile as a<br />

result <strong>of</strong> less neuropathy. Based on the fact that no cumulative toxicity is<br />

expected and the results reported in second-line, we aim to test the role <strong>of</strong><br />

vinflunine in first line therapy, as maintenance treatment for patients who<br />

obtain clinical benefit after platinum. Methods: This is a multicenter,<br />

randomized, open label, pro<strong>of</strong>-<strong>of</strong>-concept study that will be performed in<br />

20 institutions members <strong>of</strong> the Spanish Oncology Genitourinary Group<br />

(SOGUG). Subjects will be randomized in a 1:1 ratio to receive vinflunine<br />

320 mg/m2 every 21 days plus BSC vs BSC alone until disease progression.<br />

Vinflunine dose will be 280mg/m2 for patients with PS�1, age � 75 years,<br />

prior pelvic radiotherapy or creatinine clearance Cr �60ml/min. Stratification<br />

factors are: 1) Scheduled dose at randomization (320 vs 280mg/m2)<br />

2) Liver metastasis (Y/N). Main inclusion criteria are: Subjects �18 and�<br />

80 years <strong>of</strong> age with histological diagnosis <strong>of</strong> transitional cell carcinoma <strong>of</strong><br />

the urothelial tract and measurable disease with radiological response or<br />

stabilization after 6 cycles <strong>of</strong> a platinum containing doublet for metastasic/<br />

advanced disease. Primary objective will be progression free survival (PFS),<br />

considering as clinically relevant 6 months in experimental arm. To<br />

guarantee an overall type-1 � error (one side) no greater than 0.05 and a<br />

type II (�) error 0.1 for the primary endpoint <strong>of</strong> PFS, a sample size <strong>of</strong> 86<br />

patients allocated in a 1:1 ratio is planned. Recruitment is scheduled to<br />

start by February 2012. A pharmacogenomic translational study will also<br />

be conducted.<br />

TPS4676 General Poster Session (Board #14C), Sun, 8:00 AM-12:00 PM<br />

Phase II trial <strong>of</strong> gemcitabine and cisplatin plus ipilimumab as first-line<br />

treatment for metastatic urothelial carcinoma. Presenting Author: Benjamin<br />

Adam Gartrell, Division <strong>of</strong> Hematology and Medical Oncology, The<br />

Tisch Cancer Institute, Mount Sinai School <strong>of</strong> Medicine, New York, NY<br />

Background: While metastatic urothelial carcinoma is a chemosensitive<br />

neoplasm, current therapeutic approaches are inadequate. Preclinical and<br />

clinical data suggests that bladder cancer is immunogenic. For example,<br />

CD8� tumor infiltrating lymphocytes correlate with survival in patients<br />

with muscle-invasive disease (Sharma, PNAS, 2007). However, immunotherapeutic<br />

approaches have been rarely investigated for advanced urothelial<br />

cancer. CTLA4 blockade with ipilimumab is a novel approach to<br />

immunotherapy that interrupts T-cell pathways responsible for immune<br />

down-regulation or tolerance. In a pro<strong>of</strong> <strong>of</strong> concept study, ipilimumab was<br />

administered to 12 patients with muscle-invasive disease preoperatively<br />

(Carthon, Clin Can Res, 2010). Treatment resulted in perivascular infiltration<br />

<strong>of</strong> cells positive for CD3, CD8, CD4, and granzyme and intriguing<br />

evidence <strong>of</strong> antitumor activity. In the current trial, we will explore a<br />

“phased” schedule <strong>of</strong> chemotherapy and ipilimumab with the goal <strong>of</strong><br />

“autovaccinating” patients to tumor antigen with chemotherapy prior to<br />

introduction <strong>of</strong> immune checkpoint blockade. Methods: We have initiated a<br />

phase II clinical trial <strong>of</strong> gemcitabine (G), cisplatin (C), plus ipilimumab in<br />

chemonaive patients with unresectable and/or metastatic urothelial cancer<br />

within the Hoosier Oncology Group network. During cycles 1 and 2, G<br />

(1000 mg/m2 D day 1 � 8) and C (70 mg/m2 D 1) will be administered<br />

every 21 days without ipilimumab. During cycles 3-6, GC plus ipilimumab<br />

(10 mg/kg day 1) will be administered every 21 days. Patients without<br />

evidence <strong>of</strong> disease progression after completion cycle 6 will continue<br />

single-agent ipilimumab “maintenance” every 3 months. The primary<br />

objective is to determine the 1-year overall survival. The trial will enroll 36<br />

patients and is powered to detect an improvement in 1-year survival from<br />

60% to 80%. Secondary objectives include progression-free survival,<br />

disease control rate, safety, and immunologic outcomes. Correlative<br />

studies will include serial measurements <strong>of</strong> the global composition <strong>of</strong><br />

immune cells in the blood by polychromatic flow cytometry, whole blood<br />

transcriptional pr<strong>of</strong>iling, and tumor-antigen specific CD8� T cells assays.<br />

TPS4675^ General Poster Session (Board #14B), Sun, 8:00 AM-12:00 PM<br />

Randomized phase II study <strong>of</strong> docetaxel with or without ramucirumab<br />

(IMC-1121B) or icrucumab (IMC-18F1) in patients with urothelial transitional<br />

cell carcinoma (TCC) following progression on first-line platinumbased<br />

therapy. Presenting Author: Daniel Peter Petrylak, Columbia<br />

University Medical Center, New York, NY<br />

Background: The vascular endothelial growth factor (VEGF) pathway may<br />

play an important role in the pathogenesis <strong>of</strong> bladder cancer. Two<br />

antibodies (ramucirumab or icrucumab) in combination with docetaxel are<br />

being tested in this clinical study. Ramucirumab is a fully human IgG1<br />

monoclonal antibody (MAb) that specifically binds with high affinity to<br />

VEGF receptor-2 (VEGFR-2), thereby blocking the interaction <strong>of</strong> VEGF<br />

ligands. Ramucirumab inhibits VEGF-mediated proliferation <strong>of</strong> human<br />

endothelial cells and migration <strong>of</strong> human leukemia cells. Icrucumab is a<br />

fully human IgG1 MAb that specifically binds with high affinity to VEGFR-1<br />

and blocks the binding <strong>of</strong> VEGF-A, VEGF-B, and placental growth factor<br />

(PlGF) to the receptor, thereby inhibiting subsequent signaling. Blockage<br />

<strong>of</strong> VEGFR-1 and VEGFR-2 by antibodies demonstrates antiangiogenic and<br />

antitumor activity and prevents dissemination and growth <strong>of</strong> lung metastases<br />

in several tumor xenograft models. Methods: This study includes<br />

patients (pts) with TCC with progressive disease after prior platinum-based<br />

therapy. Pts are randomized equally to 1 <strong>of</strong> 3 open-label treatments given<br />

on a 21-day cycle: docetaxel (75 mg/m2 ) on Day 1 (Arm A); docetaxel on<br />

Day 1 and ramucirumab (10 mg/kg) on Day 1 (Arm B); or docetaxel on Day<br />

1 and icrucumab (12 mg/kg) on Days 1 and 8 (Arm C). Randomization is<br />

stratified by the absence/presence <strong>of</strong> visceral metastases and receipt <strong>of</strong><br />

prior antiangiogenic therapy. The primary endpoint is progression-free<br />

survival (PFS). Secondary outcome measures include response rate and<br />

duration <strong>of</strong> response, overall survival, and pharmacodynamic markers,<br />

including circulating levels <strong>of</strong> PlGF, VEGF-A, VEGF-B, soluble VEGFR-1,<br />

and soluble VEGFR-2. Exploratory analyses <strong>of</strong> VEGF, VEGFR-1, and<br />

VEGFR-2 genetic polymorphisms will be performed. As <strong>of</strong> 12 January<br />

2012, approximately 20% <strong>of</strong> 138 planned pts were randomized in the US<br />

and Canada. The sample size will allow differentiation <strong>of</strong> an expected<br />

increase in median PFS from 3.0 months to 4.5 months with a one-sided<br />

alpha <strong>of</strong> 0.1 and a power <strong>of</strong> 71%. <strong>Clinical</strong>Trials.gov identifier:<br />

NCT01282463.<br />

TPS4677 General Poster Session (Board #14D), Sun, 8:00 AM-12:00 PM<br />

Phase II study <strong>of</strong> everolimus (E) in refractory germ cell tumors (GCTs).<br />

Presenting Author: Michal Mego, Department <strong>of</strong> Medical Oncology, School<br />

<strong>of</strong> Medicine, Comenius University, National Cancer Institute, Bratislava,<br />

Slovakia<br />

Background: GCTs represent a model for the cure <strong>of</strong> cancer. Nonetheless, a<br />

small proportion <strong>of</strong> patients (pts) develop disease recurrence. Because <strong>of</strong><br />

insufficient results in the treatment <strong>of</strong> relapsed GCTs, evaluation <strong>of</strong> new<br />

treatments is a priority. Loss <strong>of</strong> the tumor suppressor gene PTEN (phosphatase<br />

and tensin homolog) marks the transition from intratubular germ cell<br />

neoplasias (ITGCN) to invasive GCTs. In the testis, PTEN was abundantly<br />

expressed in germ cells whereas it was virtually absent from 56% <strong>of</strong><br />

seminomas as well as from 86% <strong>of</strong> embryonal carcinomas and virtually all<br />

teratomas. On the contrary, ITGCN intensely expressed PTEN, indicating<br />

that loss <strong>of</strong> PTEN expression is not in early event in GCTs development, but<br />

is associated with disease progression. PTEN inactivation is associated<br />

with dysregulation <strong>of</strong> PI3K/Akt pathway and increased mTOR signalling.<br />

We hypothesize that dysregulation <strong>of</strong> PI3K/Akt pathway due to PTEN<br />

inactivation in GCTs suggests that these pts could have greater benefit from<br />

mTOR inhibition. Methods: In December 2011, National Cancer Institute <strong>of</strong><br />

Slovakia launched an one arm, two-staged phase II study aimed to evaluate<br />

the efficacy and toxicity <strong>of</strong> E in pts with refractory GCTs. The primary<br />

objective is to determine the efficacy <strong>of</strong> E in patients with refractory GCTs.<br />

Secondary objectives includes favourable response rate, time to progression<br />

and safety. The pts with radiological and/or serological pro<strong>of</strong> <strong>of</strong><br />

relapsed GCTs, who were not amenable to be cured by chemotherapy or<br />

surgery and who failed at least two platinum-based regimens or one<br />

platinum regimen in case <strong>of</strong> platinum-refractory disease or primary mediastinal<br />

non-seminomatous GCTs were eligible. All other standard inclusion<br />

and exclusion criteria for study <strong>of</strong> salvage treatment in refractory GCTs pts<br />

were applied. E will be administered at a dose <strong>of</strong> 10mg daily until<br />

progression or unacceptable toxicity. Assuming a response rate <strong>of</strong> clinical<br />

interest <strong>of</strong> �40%, a minimal response rate <strong>of</strong> 20%, a probability <strong>of</strong> 5% for<br />

rejecting an active drug, and a probability <strong>of</strong> 20% to further evaluate an<br />

ineffective drug, 18 pts will be enrolled into the first cohort. If �4<br />

responses will be observed, the study will be terminated, otherwise, it will<br />

be continued with a second cohort <strong>of</strong> 20 pts.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


TPS4678 General Poster Session (Board #14E), Sun, 8:00 AM-12:00 PM<br />

Neoadjuvant therapy preceding cytoreductive nephrectomy to develop<br />

individualized first-line therapy with everolimus for advanced renal cell<br />

carcinoma (RCC). Presenting Author: Guru Sonpavde, Texas Oncology,<br />

Houston, TX, and Department <strong>of</strong> Medicine, Section <strong>of</strong> Medical Oncology,<br />

Michael E. DeBakey Veterans Affairs Medical Center, Baylor College <strong>of</strong><br />

Medicine, Houston, TX<br />

Background: Everolimus (E), an orally administered mTOR inhibitor, is the<br />

conventional agent for progressive disease following vascular endothelial<br />

growth factor (VEGF) inhibitors for advanced RCC. Given the excellent<br />

tolerability and convenient oral administration, a rationale may be made to<br />

develop E as first-line therapy for selected patients. The period between<br />

diagnosis and cytoreductive nephrectomy (CN) may be utilized for brief<br />

therapy to capture biologic activity in tumor tissue and facilitate individualized<br />

systemic therapy. The agent may be resumed following CN and<br />

continued until progression. Pharmacodynamic (PD) alterations from<br />

baseline to CN tumor tissue may predict progression-free survival (PFS).<br />

We hypothesized that the application <strong>of</strong> this paradigm to E may enable the<br />

employment <strong>of</strong> first-line E in patients predicted to optimally benefit.<br />

Methods: This is a 27-patient phase II trial being conducted at the Baylor<br />

College <strong>of</strong> Medicine and University <strong>of</strong> Texas Southwestern, and 5 patients<br />

have been enrolled currently. Patients with �3 poor risk factors or those<br />

who refuse or are ineligible for VEGF inhibitors and have untreated<br />

metastatic RCC are eligible. Adequate hematologic, renal and hepatic<br />

function is required. The primary endpoint is PFS and secondary endpoints<br />

are response, survival and toxicities. Exploratory endpoints are tumor<br />

tissue, peripheral blood mononuclear cell (PBMC) and plasma PD studies<br />

and pharmacokinetic (PK) studies, and their association with PFS. PD<br />

studies include DNA, mRNA, protein and miRNA studies to evaluate the<br />

mTORC1 among other pathways. Patients initially undergo 4 core biopsies<br />

<strong>of</strong> the primary renal mass for fresh frozen tumor. Patients then receive E 10<br />

mg orally once daily for 3-5 weeks. CN is performed within 24-48 hours<br />

after the last dose <strong>of</strong> E. At the time <strong>of</strong> CN and before separation from the<br />

vascular supply, 3 core biopsies are performed. E is resumed 2-4 weeks<br />

after CN and continued until progression or intolerable toxicities. <strong>Clinical</strong><br />

and laboratory assessments are performed every 4-5 weeks, and imaging is<br />

performed every 8 weeks. A progressing metastatic site will be biopsied (4<br />

core biopsies).<br />

TPS4680 General Poster Session (Board #14G), Sun, 8:00 AM-12:00 PM<br />

A phase II study <strong>of</strong> bevacizumab and erlotinib in subjects with advanced<br />

hereditary leiomyomatosis and renal cell cancer (HLRCC) or sporadic<br />

papillary renal cell cancer (RCC). Presenting Author: Eric A. Singer,<br />

Urologic Oncology Branch, Center for Cancer Research, National Cancer<br />

Institute, National Institutes <strong>of</strong> Health, Bethesda, MD<br />

Background: There are currently no treatment options <strong>of</strong> proven efficacy for<br />

patients with advanced papillary RCC. The evaluation <strong>of</strong> families with<br />

inherited forms <strong>of</strong> RCC has allowed for the identification <strong>of</strong> genetic<br />

alterations associated with papillary RCC. HLRCC is one such familial<br />

condition with a propensity for the development <strong>of</strong> a clinically aggressive<br />

variant <strong>of</strong> papillary RCC characterized by unique histopathologic features.<br />

Germline mutations in the gene for the Krebs cycle enzyme fumarate<br />

hydratase (FH) are the genetic hallmark <strong>of</strong> HLRCC. Mutational inactivation<br />

<strong>of</strong> FH leads to von Hippel-Lindau-independent upregulation <strong>of</strong> hypoxia<br />

inducible factors (HIF) and their downstream transcriptional targets such<br />

as vascular endothelial growth factor (VEGF) and transforming growth<br />

factor-alpha (TGF-�) / epidermal growth factor receptor (EGFR). Upregulation<br />

<strong>of</strong> the HIF pathway may also be important in sporadic papillary RCC,<br />

although the prevalence <strong>of</strong> FH inactivation in these patients is unknown.<br />

We propose to evaluate the activity <strong>of</strong> dual VEGF/EGFR blockade with<br />

bevacizumab/erlotinib in patients with HLRCC-associated RCC and sporadic<br />

papillary RCC. Methods: This is a single arm phase II study based on<br />

an open label Simon two-stage minmax design with a maximum accrual <strong>of</strong><br />

20 patients in each <strong>of</strong> two cohorts: Cohort 1- patients with HLRCC; Cohort<br />

2- patients with sporadic papillary RCC. The primary endpoint is RECIST<br />

overall response rate, assessed independently in each cohort. Patients will<br />

receive bevacizumab (10mg/Kg IV every 2 weeks) and erlotinib (150mg PO<br />

daily). Dose reductions and drug interruptions for toxicity are allowed.<br />

Patients will be evaluated for response every 8 weeks. Major eligibility<br />

criteria include: advanced RCC associated with HLRCC or sporadic papillary<br />

RCC; age �18 years; ECOG 0-2; measurable disease by RECIST; no<br />

brain metastases; no more than 2 prior regimens containing a VEGF<br />

inhibitor; and no prior therapy with bevacizumab. The prespecified activity<br />

goal for the first stage <strong>of</strong> accrual has been met for both cohorts.<br />

NCT01130519.<br />

Genitourinary Cancer<br />

321s<br />

TPS4679 General Poster Session (Board #14F), Sun, 8:00 AM-12:00 PM<br />

An observational study <strong>of</strong> metastatic renal cell carcinoma patients prior to<br />

initiation <strong>of</strong> initial systemic therapy. Presenting Author: Elizabeth R.<br />

Plimack, Fox Chase Cancer Center, Philadelphia, PA<br />

Background: The biology <strong>of</strong> metastatic RCC is extremely diverse, including a<br />

subpopulation <strong>of</strong> patients with indolent growth <strong>of</strong> metastases. Because <strong>of</strong><br />

the acute and chronic toxicity and assumed non-curative nature <strong>of</strong> current<br />

systemic targeted therapy, a select subset <strong>of</strong> patients may be better served<br />

with initial surveillance. Such an approach may allow for deferral <strong>of</strong><br />

systemic therapy to minimize overall treatment-related toxicity burden<br />

while maintaining treatment benefit. Methods: A prospective phase II trial is<br />

being conducted to characterize the clinical course <strong>of</strong> patients with mRCC<br />

who defer initial systemic treatment. Appropriate patients are selected by<br />

the treating physician based on an observed indolent growth pattern. As<br />

such, there is a 12 month window allowed between the first diagnosis <strong>of</strong><br />

metastatic disease and study entry. Patients must be treatment-naïve,<br />

asymptomatic, with histologically confirmed mRCC and clinically-evident,<br />

measurable disease to be eligible. Radiographic assessment is performed<br />

at baseline, every 3 months for year 1, every 4 months for year 2, then every<br />

6 months. The primary objective is to characterize the clinical outcome <strong>of</strong><br />

patients on observation in terms <strong>of</strong> time to RECIST defined disease<br />

progression and time to initiation <strong>of</strong> systemic treatment. Secondary<br />

endpoints include measurement <strong>of</strong> disease-related symptoms and depression/anxiety<br />

using standardized questionnaires (FKSI-DRS and HADS), as<br />

well as correlative endpoints analyzing the immune response over time for<br />

which blood is being collected for immune assays (TH1/TH2 phenotype,<br />

Tregs). Pts remain on study until initiation <strong>of</strong> systemic therapy due to<br />

radiographic disease progression, development <strong>of</strong> disease-related symptoms,<br />

withdraw <strong>of</strong> consent or clinical change that renders the patient<br />

unacceptable for further observation. Local therapy (e.g. surgery, radiation)<br />

during the observation period is permitted. Currently, 29 patients have<br />

been accrued at 5 collaborating sites. The target accrual <strong>of</strong> 50 pts provides<br />

adequate power for the primary descriptive endpoint as well as 80% power<br />

to detect changes from baseline for the correlate endpoints based on a<br />

two-sided Wilcoxon signed rank test.<br />

TPS4681 General Poster Session (Board #14H), Sun, 8:00 AM-12:00 PM<br />

EVERSUN: A phase II trial <strong>of</strong> everolimus alternating with sunitinib as<br />

first-line therapy for advanced renal cell carcinoma (RCC) (ANZUP trial<br />

0901). Presenting Author: Ian D. Davis, Austin Health, Heidelberg,<br />

Australia<br />

Background: Treatment <strong>of</strong> RCC has improved due to better understanding <strong>of</strong><br />

its biology. New targeted therapies have improved time to progression and<br />

overall survival but the optimal sequencing <strong>of</strong> agents is unknown. Currently<br />

drugs are given sequentially, usually starting with sunitinib and <strong>of</strong>ten<br />

followed by an mTOR inhibitor or another VEGFR-targeted therapy, but<br />

resistance to both drugs eventually occurs probably due to host adaptive<br />

responses. We hypothesize that resistance might be delayed by planned<br />

alternation <strong>of</strong> treatments. Methods: EVERSUN is a single-arm, two-stage,<br />

multicenter, phase II clinical trial aiming to determine the activity and<br />

safety <strong>of</strong> an alternating regimen <strong>of</strong> two therapies with different targets<br />

(sunitinib and everolimus) in patients with advanced RCC. Key eligibility<br />

criteria: RCC with a clear cell component; metastatic or locally advanced<br />

disease not suitable for resection; ECOG performance status 0-1; low or<br />

intermediate MSKCC prognostic score. The primary endpoint is the status<br />

<strong>of</strong> being alive and progression-free (RECIST 1.1) 6 months after registration.<br />

Target accrual <strong>of</strong> 55 subjects gives 95% power and 95% confidence<br />

to distinguish between 6-month progression free survival rates <strong>of</strong> 64% or<br />

lower vs 84% or higher using a Simon 2-stage minimax design. The criteria<br />

for further evaluation come from the pivotal trial <strong>of</strong> single agent sunitinib as<br />

first line therapy for RCC, in which the 6-month progression free survival<br />

rate was 74%. Trial treatment is administered in 12-week (wk) cycles<br />

consisting <strong>of</strong> 4 wks <strong>of</strong> sunitinib (50 mg daily) followed by 2 wks rest,<br />

followed by 5 wks <strong>of</strong> everolimus (10 mg daily) followed by 1 wk rest.<br />

Disease progression is interpreted as failure <strong>of</strong> the most recent drug taken.<br />

<strong>Part</strong>icipants who stop one drug because <strong>of</strong> toxicity or disease progression,<br />

on or before the 6 month assessment, will continue the other drug until<br />

subsequent progression or prohibitive toxicity on the second drug. EVER-<br />

SUN is an ANZUP Cancer Trials Group Ltd. trial coordinated by the NHMRC<br />

<strong>Clinical</strong> Trials Centre. Accrual commenced in September 2010 with 38/55<br />

participants recruited as <strong>of</strong> the 31-Jan-12 from 17 Australian sites<br />

(ACTRN12609000643279).<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


322s Genitourinary Cancer<br />

TPS4682 General Poster Session (Board #15A), Sun, 8:00 AM-12:00 PM<br />

Treatment <strong>of</strong> refractory metastatic renal cell carcinoma (RCC) with lenvatinib<br />

(E7080) and everolimus. Presenting Author: Ana M. Molina, Memorial<br />

Sloan-Kettering Cancer Center, New York, NY<br />

Background: Lenvatinib (E7080) is an oral tyrosine kinase inhibitor targeting<br />

vascular endothelial growth factor receptor (VEGFR) 1-3, fibroblast<br />

growth factor receptor 1-4, RET, KIT and platelet-derived growth factor<br />

receptor beta. Lenvatinib has demonstrated acceptable toxicity and antitumor<br />

activity in Phase I and II studies (Glen H et al. JCO 2008, 26;15S:<br />

3526;Yamada K et. al. Clin Cancer Res 2011, 17:2528-2537; Sherman S<br />

I et al. JCO 2011, 29; 15S: 5503). Improved understanding <strong>of</strong> the<br />

molecular basis <strong>of</strong> renal cell carcinoma (RCC) has led to the development <strong>of</strong><br />

targeted therapies (inhibitors <strong>of</strong> mTOR and <strong>of</strong> the VEGF pathway) that have<br />

shown clinical benefit in patients with metastatic RCC. However patients<br />

develop resistance and ultimately progress during treatment with available<br />

single agents. Combination therapy targeting different pathways have the<br />

potential to improve efficacy. Methods: This multicenter, randomized,<br />

open-label, phase Ib/II study is investigating everolimus (an oral mTOR<br />

inhibitor) in combination with lenvatinib in RCC patients with unresectable<br />

or metastatic disease (NCT01136733). The study is being conducted in<br />

two phases. The phase Ib dose-escalation component will determine the<br />

maximum tolerated dose (MTD) <strong>of</strong> the combination; starting doses are 12<br />

mg/day lenvatinib and 5 mg/day everolimus. A cohort expansion will follow<br />

to confirm the MTD and establish a recommended phase II dose (RP2<br />

dose). Approximately 150 patients will be enrolled in phase II and<br />

randomized 1:1:1 to receive: lenvatinib and everolimus at the RP2 dose<br />

(Arm A); lenvatinib 24 mg/day (Arm B); or everolimus 10 mg/day (Arm C).<br />

Patients with clear cell RCC, ECOG performance score �2 and progression<br />

during or after one prior VEGF-targeted treatment are eligible for phase II.<br />

The primary endpoint for Phase II is to compare progression-free survival <strong>of</strong><br />

the investigational arms (A and B) with single agent everolimus (Arm C).<br />

Three phase Ib dose escalation cohorts have completed enrollment, and<br />

enrollment <strong>of</strong> the expansion cohort commenced in November 2011.<br />

<strong>Part</strong>icipating phase II centers will be located in the US, UK, Spain, Poland<br />

and Czech Republic and are expected to enroll patients throughout 2012.<br />

TPS4684 General Poster Session (Board #15C), Sun, 8:00 AM-12:00 PM<br />

A phase II study <strong>of</strong> intermittent sunitinib in previously untreated patients<br />

(pts) with metastatic renal cell carcinoma (mRCC). Presenting Author:<br />

Laura S. Wood, Cleveland Clinic Taussig Cancer Institute, Cleveland, OH<br />

Background: Sunitinib is a standard <strong>of</strong> care initial treatment in mRCC. One<br />

challenge is balancing the acute and chronic toxicity <strong>of</strong> therapy with<br />

clinical benefit. Pre-clinical and retrospective clinical data support the<br />

concept that treatment breaks are feasible and not associated with a<br />

reduction in efficacy <strong>of</strong> sunitinib. It is hypothesized that an intermittent<br />

dosing regimen <strong>of</strong> sunitinib in mRCC pts is feasible, and may lead to<br />

prolonged duration <strong>of</strong> disease control with improved tolerance. Methods:<br />

Eligibility criteria include treatment-naïve clear cell mRCC, measurable<br />

disease, and adequate organ function. Pts will be treated for 4 cycles <strong>of</strong><br />

sunitinib (50 mg 4/2) in the absence <strong>of</strong> unacceptable toxicity or RECISTdefined<br />

progression. Pts with � 10% reduction in tumor burden per<br />

RECIST following 4 cycles will have sunitinib held, with re-staging CT scans<br />

approximately every 10 weeks thereafter to assess tumor burden. Sunitinib<br />

will be re-initiated in those patients with an increase in tumor burden <strong>of</strong><br />

10% or greater (per RECIST) compared to the scan obtained just prior to<br />

holding sunitinib. Pts who have RECIST tumor burden reduction <strong>of</strong> 10% or<br />

more compared to scans at the start <strong>of</strong> the most recent treatment period will<br />

again have sunitinib held. This intermittent sunitinib dosing will continue<br />

until RECIST-defined disease progression while on sunitinib. Patients not<br />

initially achieving at least a 10% reduction in tumor burden will continue<br />

sunitinib. The primary objective is the feasibility <strong>of</strong> intermittent sunitinib,<br />

defined as the proportion <strong>of</strong> patients eligible for and who undergo<br />

intermittent therapy. The alternative hypothesis is a feasibility rate <strong>of</strong> �<br />

80% vs. the null hypothesis <strong>of</strong> � 50%. Thirty pts provides 80% power<br />

based on a two-sided exact test with a .05 type I error. Secondary endpoints<br />

include PFS and toxicity. Twenty five <strong>of</strong> planned 30 patients have been<br />

enrolled.<br />

TPS4683 General Poster Session (Board #15B), Sun, 8:00 AM-12:00 PM<br />

Phase III trial <strong>of</strong> dovitinib (TKI258) versus sorafenib in patients with<br />

metastatic renal cell carcinoma after failure <strong>of</strong> anti-angiogenic (VEGFtargeted<br />

and mTOR inhibitor) therapies. Presenting Author: Robert John<br />

Motzer, Memorial Sloan-Kettering Cancer Center, New York, NY<br />

Background: Standard first- and second-line treatments in metastatic renal<br />

cell carcinoma (mRCC) target the vascular endothelial growth factor (VEGF)<br />

and mammalian target <strong>of</strong> rapamycin (mTOR) signaling pathways. However,<br />

signaling through other pathways, including the fibroblast growth factor<br />

receptor (FGFR) pathway, may account for tumor resistance to these<br />

standard therapies. Dovitinib (TKI258) is an oral FGF, VEGF, and plateletderived<br />

growth factor (PDGF) receptor tyrosine kinase inhibitor, with IC50 values <strong>of</strong> � 10 nM. In a phase II study <strong>of</strong> 59 RCC patients, many <strong>of</strong> whom<br />

had failed prior VEGF-targeted and mTOR inhibitor therapies, dovitinib<br />

(500 mg/day on a 5-days-on/2-days-<strong>of</strong>f schedule) was well tolerated and<br />

demonstrated promising anti-tumor effects, with progression-free survival<br />

(PFS) <strong>of</strong> 5.5 months (Angevin et al, ASCO 2011). Methods: Approximately<br />

550 patients from over 26 countries will be randomized 1:1 in this<br />

multicenter, open-label, randomized phase III trial (NCT01223027) to<br />

receive dovitinib (500 mg/day on a 5-days-on/2-days-<strong>of</strong>f schedule) or<br />

sorafenib (400 mg twice daily). Eligible mRCC patients must have failed 1<br />

VEGF-targeted therapy and 1 mTOR inhibitor (disease progression on or<br />

within 6 months <strong>of</strong> stopping the prior treatment). Patients will remain on<br />

study until disease progression, unacceptable toxicity, death, or discontinuation<br />

for any other reason. No treatment crossover is planned. The primary<br />

endpoint is PFS as determined by central radiology assessment according<br />

to RECIST v1.1, with evaluations performed every 8 weeks. Secondary<br />

endpoints include overall survival, overall response rate, safety, patientreported<br />

outcomes, and pharmacokinetics. The pharmacodynamic effects<br />

<strong>of</strong> dovitinib on plasma/serum biomarkers will also be explored. The data<br />

monitoring committee last reviewed the trial on 20 December 2011 and<br />

recommended that the trial continue as planned. This is the first third-line<br />

randomized clinical trial in mRCC to evaluate a multitargeted inhibitor <strong>of</strong><br />

FGFR.<br />

TPS4685 General Poster Session (Board #15D), Sun, 8:00 AM-12:00 PM<br />

Prospective assessment <strong>of</strong> circulating endothelial cells (CECs) as early<br />

markers <strong>of</strong> clear cell renal cell carcinoma (CCRCC) progression in first line<br />

setting: The Circles study (SOGUG 2011-01). Presenting Author: Daniel E.<br />

Castellano, University Hospital 12 de Octubre, Madrid, Spain<br />

Background: Since the identification <strong>of</strong> Von Hippel Lindau (VHL) gene<br />

mutations as a critical step in the development <strong>of</strong> most Clear Cell Renal Cell<br />

Carcinomas (CCRCC), neoangionesis inhibition has become a cornerstone<br />

in their treatment. Despite the proven efficacy <strong>of</strong> antiangiogenic drugs,<br />

most patients will not achieve partial response by RECIST criteria. Thus, to<br />

accurately determine tumor progression is a challenging question incompletely<br />

answered by traditional radiological assessment. In recent years<br />

CECs counts have been proposed as surrogate biomarkers <strong>of</strong> angiogenesis<br />

that could be used for monitoring tumor evolution while on targeted<br />

therapies. We aim to figure out if CECs elevations could anticipate<br />

radiological progression in CCRCC. Methods: An observational prospective<br />

study is being performed in 10 institutions members <strong>of</strong> the Spanish<br />

Oncology Genitourinary group (SOGUG). Patients older than 18 years with<br />

histologically proven CCRCC on first line treatment with any targeted drug<br />

who have not progressed after 3 months <strong>of</strong> therapy are considered elegible.<br />

Recruitment begun on August the 1st 2011 and 15 <strong>of</strong> the 75 scheduled<br />

patients have been included so far. CECs are periodically collected in<br />

peripheral blood every 6 weeks for 15 months or radiological tumor<br />

progression, whatever occurs first. Median CEC values will be calculated<br />

and stated by descriptive statistics (Cellsearch, VERIDEX). To explore the<br />

evolution <strong>of</strong> CECs counts along treatment a linear model will be constructed.<br />

An association among CECs counts changes and time to progression<br />

will be analyzed with Cox model.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


TPS4686 General Poster Session (Board #15E), Sun, 8:00 AM-12:00 PM<br />

A phase II biomarker assessment <strong>of</strong> tivozanib in oncology (BATON) trial in<br />

patients (pts) with advanced renal cell carcinoma (RCC). Presenting<br />

Author: Thomas E. Hutson, Texas Oncology–Baylor Charles A. Sammons<br />

Cancer Center, Dallas, TX<br />

Background: Tivozanib is an oral, potent, and selective tyrosine kinase<br />

inhibitor targeting all three vascular endothelial growth factor (VEGF)<br />

receptors; it showed efficacy and tolerability in a Phase II trial in pts with<br />

advanced RCC (Nosov et al. JCO 2011;29[18S]:4550). Preclinical studies<br />

have identified a 42-gene tivozanib resistance signature (Jie et al. EORTC-<br />

NCI-AACR 2010; abstract 608) that may be predictive <strong>of</strong> tivozanib<br />

sensitivity. This study (NCT01297244) is designed to evaluate these<br />

potential biomarkers for tivozanib activity in pts with advanced RCC.<br />

Methods: Pts with advanced RCC (stratified as clear cell or non-clear cell)<br />

who underwent nephrectomy and received �1 prior systemic treatment (no<br />

prior VEGF- or mammalian target <strong>of</strong> rapamycin–targeted therapy) entered<br />

this open-label, single-arm study conducted in the United States and<br />

Canada beginning January 2011. Pts receive tivozanib 1.5 mg/d orally (3<br />

weeks on, 1 week <strong>of</strong>f schedule). Primary objectives include correlation <strong>of</strong><br />

biomarkers in blood (e.g. VEGF, hepatocyte growth factor [HGF]) and tumor<br />

tissue (e.g. CD68, hypoxia-induced factor, VEGF, HGF, and gene expression<br />

pr<strong>of</strong>iles) with clinical activity and/or treatment-related toxicity, and<br />

6-month progression-free survival (PFS) rate. Secondary objectives include<br />

overall response rate (ORR), PFS, safety and tolerability, and pharmacokinetics.<br />

Contingency table methods will be used for biomarker correlation<br />

with ORR, and Cox proportional-hazards regression for correlation with<br />

PFS. Sample size (100 pts) is estimated based on clinical considerations<br />

and the precision with which 6-month PFS can be estimated. Biomarkers<br />

were defined at study outset; the requirement <strong>of</strong> prior nephrectomy ensured<br />

availability <strong>of</strong> primary tumors as controls for correlative analysis. As <strong>of</strong><br />

January 2012, the study completed enrollment <strong>of</strong> 100 pts, demonstrating<br />

a large number <strong>of</strong> pts can be enrolled to a biomarker study with well-defined<br />

candidate genes and critical inclusion criteria to facilitate compliance.<br />

Tivozanib biomarkers evaluated in this study, along with those evaluated in<br />

other solid tumors, may play an important role in optimizing patient<br />

selection.<br />

TPS4688 General Poster Session (Board #15G), Sun, 8:00 AM-12:00 PM<br />

Phase II study <strong>of</strong> dovitinib in first-line metastatic or nonresectable primary<br />

adrenocortical carcinoma (ACC): SOGUG study 2011-03. Presenting<br />

Author: Paula Jimenez, Hospital Universitario Central de Asturias, Oviedo,<br />

Spain<br />

Background: Dovitinib is a novel targeted therapy, that has proven to inhibit,<br />

among other tyrosin kinases, the fibroblast growth factor receptor (FGFR).<br />

Since this pathway has been proposed to play a major role in ACC, we aim to<br />

test the clinical efficacy <strong>of</strong> dovitinib in this tumor. Methods: An open label<br />

phase II trial has been designed in patients with advanced non-resectable<br />

ACC. The objective will be to obtain at least a 15% response rate according<br />

to RECIST criteria. Taking as a basis the two-stage Gehan model, 15<br />

patients would need to be included in the first stage to demonstrate a<br />

treatment efficacy <strong>of</strong> at least 15%. Sample size calculation was done based<br />

on the following parameters, probability <strong>of</strong> Type I error � � 0.05, power <strong>of</strong><br />

the test (1 - �) � 0.8. Main inclusion criteria are advanced non-resectable<br />

disease and no prior therapy (other than mitotane). Dovitinib scheduled<br />

dose matches currently employed standard in the drug development<br />

(500mg daily for 5 days then 2 days <strong>of</strong>f) for 6 months. If clinical benefit is<br />

obtained longer treatment will be allowed for particular patients. Since this<br />

is an extremely unfrequent disease 7 institutions, members <strong>of</strong> the SOGUG<br />

(Spanish Oncology Genitourinary Group), will participate. The active<br />

support <strong>of</strong> a big collaborative group will guarantee candidate patients to be<br />

refereed to such institutions. Starting January 26th 2012 recruitment is<br />

scheduled to last around 12 months. A translational research, including<br />

whole exome analysis, will be performed in order to improve our scarce<br />

knowledge <strong>of</strong> ACC.<br />

Genitourinary Cancer<br />

323s<br />

TPS4687 General Poster Session (Board #15F), Sun, 8:00 AM-12:00 PM<br />

Phase I/II study <strong>of</strong> high-dose interleukin 2, aldesleukin, in combination<br />

with the histone deacetylase inhibitor entinostat in patients with metastatic<br />

renal cell carcinoma. Presenting Author: Roberto Pili, Roswell Park Cancer<br />

Institute, Buffalo, NY<br />

Background: Immunosuppressive factors such as regulatory T cells (Tregs)<br />

limit the efficacy <strong>of</strong> immunotherapies. Histone deacetylase (HDAC) inhibitors<br />

have been shown to have anti-tumor activity in different malignancies<br />

and to induce immuno-modulatory effects. We have previously reported<br />

that a class I specific HDAC inhibitor, entinostat, has synergistic anti-tumor<br />

effect in combination with high dose interleukin-2 (IL-2) in a renal cell<br />

carcinoma model (Kato Y et al <strong>Clinical</strong> Cancer Res 2007). Our group has<br />

also recently showed that low dose entinostat induces STAT3 acetylation,<br />

down-regulates Foxp3 expression in Tregs, and blocks Tregs suppressive<br />

function without affecting T effector cells (Shen Li et al PLoSONE 2012).<br />

Methods: Based on these preclinical evidences we have initiated a Phase<br />

I/II clinical study with entinostat and high dose IL-2 in patients with<br />

metastatic renal cell carcinoma. The primary objective <strong>of</strong> the study is to<br />

evaluate the safety, tolerability and efficacy <strong>of</strong> this combination strategy.<br />

The main eligibility criteria are clear cell histology, no prior treatments and<br />

being fit to receive high dose IL-2. The Phase I portion consists <strong>of</strong> two dose<br />

levels <strong>of</strong> entinostat (3 and 5 mg) and a fixed standard dose <strong>of</strong> IL-2<br />

(600,000 units/Kg every 8 hrs.) according to a 3�3 design. The sample<br />

size <strong>of</strong> 36 patients in the Phase II portion is powered to detect an increase<br />

in objective response rate from 20% to 40% as compared to historical data<br />

with high dose IL-2 alone. Correlative studies include assessment <strong>of</strong> CD4�,<br />

CD8�, CD4�/Foxp3, NK cells in tumor and blood samples by immunohistochemistry<br />

and FACS analysis, and tumor metabolism by FDG PET. We are<br />

also exploring the relationship between entinostat exposure with pharmacodynamic<br />

endpoints. To date dose level one has been completed without<br />

DLT. Enrollment to dose level two has begun in the Fall 2011. The results<br />

from this study may confirm the role <strong>of</strong> entinostat in enhancing the effect <strong>of</strong><br />

IL-2 and may be translated into other combination strategies involving<br />

immunotherapies. The clinical trial and correlative studies are supported<br />

by the National Cancer Institute- R21CA137649.<br />

TPS4689 General Poster Session (Board #15H), Sun, 8:00 AM-12:00 PM<br />

CA184-043: A randomized, double-blind, phase III trial comparing ipilimumab<br />

versus placebo following a single dose <strong>of</strong> radiotherapy (RT) in<br />

patients (pts) with castration-resistant prostate cancer (CRPC) who have<br />

received prior treatment with docetaxel (D). Presenting Author: Charles G.<br />

Drake, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins<br />

University, Baltimore, MD<br />

Background: Ipilimumab (Ipi), a fully human monoclonal antibody which<br />

blocks CTLA-4, augments antitumor immune responses. Ipi has shown<br />

antitumor effects in prostate cancer model systems and clinical activity (via<br />

prostate-specific antigen [PSA] declines and RECIST response) in Phase<br />

1/2 investigations in CRPC, with a side effect pr<strong>of</strong>ile reflective <strong>of</strong> its<br />

mechanism <strong>of</strong> action. Preclinical data suggest that RT given prior to<br />

CTLA-4 blockade may increase antitumor activity. Methods: In this study,<br />

pts with CRPC who have progressed during or after D are randomized 1:1 to<br />

receive either a single dose <strong>of</strong> bone-directed RT followed by Ipi 10mg/kg, or<br />

RT followed by placebo. Within 2 days <strong>of</strong> RT administration (up to 5 lesions<br />

at 8 Gy on a single day) patients receive their initial dose <strong>of</strong> Ipi/placebo;<br />

Ipi/placebo is then given every 3 weeks for a total <strong>of</strong> 4 doses. Eligible pts<br />

may continue to receive blinded study drug every 12 weeks until they meet<br />

treatment stopping criteria, withdraw consent, are lost to follow-up, or<br />

study closure. The primary endpoint is overall survival (OS). Secondary<br />

endpoints include progression-free survival, pain response, and safety. The<br />

study is designed to detect a 3.8 month difference (HR�0.76) in median<br />

OS with 90% power and 0.05 2-sided type one error. The enrollment goal is<br />

800 randomized patients, with a single interim analysis for superiority <strong>of</strong><br />

OS planned at 435 events at approximately 33 months from first patient<br />

first visit (<strong>Clinical</strong>Trials.gov identifier: NCT00861614).<br />

Key inclusion criteria<br />

Metastatic CRPC<br />

At least 1 bone metastasis appropriate for RT<br />

Testosterone 2 prior cytotoxic chemo regimens for CRPC<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


324s Genitourinary Cancer<br />

TPS4690 General Poster Session (Board #16A), Sun, 8:00 AM-12:00 PM<br />

What is optimal timing <strong>of</strong> post prostatectomy radiotherapy? Is adjuvant<br />

radiotherapy equivalent to early salvage radiotherapy? The “RAVES” phase<br />

III randomized clinical trial. Presenting Author: Maria Pearse, Auckland<br />

City Hospital, Auckland, New Zealand<br />

Background: Three randomised trials have demonstrated a significant<br />

benefit <strong>of</strong> adjuvant post prostatectomy radiotherapy in patients with<br />

positive margins, extra capsular extension or seminal vesicle involvement,<br />

and it should be regarded as current standard <strong>of</strong> care. However, adopting<br />

this approach will expose nearly half <strong>of</strong> such patients to unnecessary<br />

radiotherapy and potential treatment morbidity. Salvage radiotherapy, if<br />

given early, is recognised to be effective. The RAVES trial is designed to<br />

compare these two approaches, and was developed in collaboration with<br />

the Trans Tasman Radiation Oncology Group (TROG), Australian and New<br />

Zealand Urogenital and Prostate Trials Group (ANZUP) and the Urological<br />

<strong>Society</strong> <strong>of</strong> Australia and New Zealand (USANZ). It aims to test the<br />

hypothesis that active surveillance with early salvage radiotherapy is<br />

non-inferior to adjuvant radiotherapy with respect to risk <strong>of</strong> biochemical<br />

failure (defined as PSA level � 0.40 ng/mL and rising). Methods: Patients<br />

must have at least one <strong>of</strong> the following risk factors: positive margins,<br />

extracapsular extension or seminal vesicle involvement. They must start<br />

radiotherapy within 4 months <strong>of</strong> radical prostatectomy (RP) and have an<br />

undetectable PSA (� 0.10 ng/ml) prior to randomisation. Patients receiving<br />

androgen deprivation or who have an artificial hip are excluded. Eligible<br />

patients are randomised to either: Arm 1: Adjuvant RT (64Gy in 32#)<br />

commenced within 4 months <strong>of</strong> RP, or Arm 2: Active surveillance with 3<br />

monthly PSA tests and commencement <strong>of</strong> early salvage RT (64Gy in 32#) if<br />

PSA rises � 0.20 ng/ml. Stratification is by seminal vesicle invasion,<br />

Gleason Score, pre-operative PSA, margin positivity (no/yes) and radiotherapy<br />

institution. A sample size <strong>of</strong> 470 patients is required to detect a<br />

10% non-inferiority margin in the 5-year biochemical failure-free rate<br />

between the adjuvant and active surveillance arms. As <strong>of</strong> 31 Jan 2012,<br />

186 patients have been recruited across 26 centres in Australia and New<br />

Zealand. A meta analysis with the MRC RADICALS and GETUG-17 trials<br />

will be prospectively designed to detect a survival difference between the<br />

two approaches.<br />

TPS4692^ General Poster Session (Board #16C), Sun, 8:00 AM-12:00 PM<br />

Comparison <strong>of</strong> two doses <strong>of</strong> cabazitaxel plus prednisone in patients (pts)<br />

with metastatic castration-resistant prostate cancer (mCRPC) previously<br />

treated with a docetaxel (D)-containing regimen. Presenting Author: Mario<br />

A. Eisenberger, Sidney Kimmel Comprehensive Cancer Center at Johns<br />

Hopkins University, James Buchanan Brady Urological Institute, Baltimore,<br />

MD<br />

Background: The phase III TROPIC study (NCT00417079) reported a<br />

significant improvement in overall survival (OS) for cabazitaxel (Cbz) �<br />

prednisone (P;CbzP) (25 mg/m2 IV Q3W/10 mg PO QD) vs mitoxantrone<br />

(M) � P (MP) (median OS 15.1 vs 12.7 mos; HR 0.70; P � 0.0001) in pts<br />

with mCRPC (also known as hormone-refractory prostate cancer) previously<br />

treated with a D-containing regimen. CbzP is approved by the FDA, EMA<br />

and other health authorities for the treatment <strong>of</strong> pts with mCRPC that has<br />

progressed after a D-containing regimen. Cbz toxicity is consistent with<br />

other taxanes; compared with M, more hematologic toxicities are reported<br />

(primarily Grade 3–4 neutropenia). Phase I/II studies identified 20 and 25<br />

mg/m2 as recommended doses; 25 mg/m2 was selected for the phase III<br />

TROPIC study. As pooled data show Grade 3–4 neutropenia incidence is<br />

lower with Cbz � 25 mg/m2 (61%) vs � 25 mg/m2 (74%), it is <strong>of</strong> interest to<br />

assess if reducing the Cbz approved dose in mCRPC lessens hematologic<br />

toxicity and is non-inferior in terms <strong>of</strong> efficacy. Methods: PROSELICA<br />

(NCT01308580) is a randomized, open-label, multinational, phase III<br />

study comparing 20 mg/m2 and 25 mg/m2 Cbz for efficacy and tolerability.<br />

Pts with a life expectancy � 6 mos, ECOG PS � 2, histologically/<br />

cytologically confirmed metastatic prostate adenocarcinoma resistant to<br />

hormone therapy and previously treated with a D-containing regimen are<br />

eligible. Pts are randomized 1:1 to receive Cbz 20 mg/m² or 25 mg/m² IV<br />

Q3W � P 10 mg PO QD, treated until disease progression, unacceptable<br />

toxicity or withdrawal <strong>of</strong> consent (max 10 cycles), and stratified according<br />

to ECOG PS, measurable disease (yes/no) and region. The primary endpoint<br />

is OS (non-inferiority design). Secondary endpoints include safety, progression-free<br />

survival (PCWG2 criteria), PSA and pain progression and response,<br />

tumor response in pts with measurable disease and health-related<br />

quality <strong>of</strong> life. Cbz PK and pharmacogenomics will be assessed in pt<br />

subgroups. Planned enrollment is 1,200 pts. Study start was in May 2011;<br />

as <strong>of</strong> Jan 2012, 270 pts had been enrolled. The first DMC meeting<br />

recommended continuing the study without change.<br />

TPS4691 General Poster Session (Board #16B), Sun, 8:00 AM-12:00 PM<br />

CA184-095: A randomized, double-blind, phase III trial to compare the<br />

efficacy <strong>of</strong> ipilimumab versus placebo in asymptomatic or minimally<br />

symptomatic patients (pts) with metastatic chemotherapy-naive castrationresistant<br />

prostate cancer (CRPC). Presenting Author: Tomasz M. Beer,<br />

Oregon Health & Science University Knight Cancer Institute, Portland, OR<br />

Background: Ipilimumab (Ipi), a fully human monoclonal antibody which<br />

blocks CTLA-4, augments antitumor immune responses. Ipi has demonstrated<br />

overall survival (OS) benefit in two Phase 3 trials for advanced<br />

melanoma, with side effects that were managed using product-specific<br />

treatment guidelines. In addition, in Phase 1/2 trials in metastatic CRPC,<br />

Ipi has shown clinical activity (as measured by prostate-specific antigen<br />

[PSA] declines and RECIST response) with no unexpected toxicities. While<br />

docetaxel is standard therapy for metastatic CRPC, its use may be delayed<br />

until pts develop symptoms. As such, this global (~150 sites in 25<br />

countries) Phase 3 study (<strong>Clinical</strong>Trials.gov identifier: NCT01057810) is<br />

evaluating Ipi vs placebo in chemotherapy-naïve pts with asymptomatic or<br />

minimally symptomatic CRPC without visceral metastases. Methods: The<br />

primary endpoint is OS; secondary endpoints include progression-free<br />

survival, time to pain progression, and time to non-hormonal systemic<br />

therapy. The study is designed to detect a 9.3 month difference (HR�0.7)<br />

in OS with 90% power and 0.05 two-sided significance. Pts are randomized<br />

at a 2:1 ratio to receive Ipi 10 mg/kg every 3 weeks for up to 4 doses or<br />

placebo, respectively, as induction therapy. Eligible pts will continue to<br />

receive maintenance therapy <strong>of</strong> blinded study drug every 12 weeks until<br />

treatment stopping criteria are met, withdrawal <strong>of</strong> consent, or study<br />

closure. The accrual goal is 600 pts randomized.<br />

Inclusion criteria<br />

Metastatic CRPC<br />

Asymptomatic or minimally symptomatic<br />

Progression during prior hormonal therapy<br />

Discontinuation <strong>of</strong> anti-androgens<br />

Testosterone < 50 ng/dl<br />

ECOG Performance Status 0-1<br />

Exclusion criteria<br />

Liver, lung, or brain metastases<br />

Prior immunotherapy or chemotherapy for metastatic CRPC<br />

Autoimmune disease<br />

HIV, Hep B, or Hep C infection<br />

Pelvic targeted radiotherapy within 3 months <strong>of</strong> study<br />

TPS4693 General Poster Session (Board #16D), Sun, 8:00 AM-12:00 PM<br />

A phase III, randomized, double-blind, multicenter trial comparing the<br />

investigational agent orteronel (TAK-700) plus prednisone (P) with placebo<br />

plus P in patients with metastatic castration-resistant prostate cancer<br />

(mCRPC) that has progressed during or following docetaxel-based therapy.<br />

Presenting Author: Robert Dreicer, Cleveland Clinic, Cleveland, OH<br />

Background: The investigational agent orteronel is a selective inhibitor <strong>of</strong><br />

17,20-lyase, a key enzyme in the testosterone synthesis pathway. In a<br />

phase 1/2 study in men with mCRPC, orteronel reduced prostate-specific<br />

antigen (PSA) levels, and inhibited testosterone and DHEA-S consistent<br />

with potent 17,20-lyase inhibition (Agus D, et al. J Clin Oncol 2012;30:s5<br />

abst 98). Docetaxel-based chemotherapy is an effective but noncurative<br />

therapy for mCRPC that has progressed on hormonal therapy; new therapeutic<br />

options are needed. Methods: This double-blind, multicenter study is<br />

assessing orteronel � P vs placebo � P in men with mCRPC<br />

(NCT01193257; C21005). Patients must have evidence <strong>of</strong> disease progression<br />

during or after receiving a total <strong>of</strong> �360 mg/m2 docetaxel within a<br />

6-mo period. Patients who are clearly intolerant to docetaxel or have<br />

progressive disease before receiving �360 mg/m2 are also eligible if they<br />

have received at least 225 mg/m2 <strong>of</strong> docetaxel within a 6-mo period and<br />

meet the other inclusion criteria. Other eligibility criteria include radiographically<br />

documented metastatic disease and baseline testosterone �50 ng/dL<br />

following surgical or medical castration. Prior adrenal-targeted therapies<br />

are not permitted. Men may have opioid-requiring bone pain. The planned<br />

sample size is 1083; men will be randomized 2:1 to receive orteronel 400<br />

mg twice daily (BID) plus P5mgBIDorplacebo plus P. The primary<br />

endpoint is overall survival; other endpoints are radiographic progressionfree<br />

survival, PSA decrease <strong>of</strong> �50% at 12 wks, pain response at 12 wks,<br />

safety, time to PSA progression, objective response by RECIST, circulating<br />

tumor cell and endocrine marker changes, and patient-reported outcomes.<br />

After disease progression, men may continue to receive study drug. Tumor<br />

specimens will be analyzed for biomarkers that may predict orteronel<br />

antitumor activity, including the TMPRSS2:ERG fusion gene. The same<br />

regimens are being evaluated in a concurrent phase 3 study in chemotherapy-naïve<br />

men with mCRPC (NCT01193244).<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


TPS4694 General Poster Session (Board #16E), Sun, 8:00 AM-12:00 PM<br />

A phase I/IIa study <strong>of</strong> the safety and efficacy <strong>of</strong> radium-223 chloride<br />

(Ra-223) with docetaxel (D) for castration-resistant prostate cancer (CRPC)<br />

patients with bone metastases. Presenting Author: Michael J. Morris,<br />

Memorial Sloan-Kettering Cancer Center, New York, NY<br />

Background: Ra-223is a first-in-class alpha-pharmaceutical that targets<br />

bone metastases (mets) with high energy alpha-particles <strong>of</strong> short range (�<br />

100 �m). In the ALSYMPCA trial, Ra-223 plus best standard <strong>of</strong> care (BSC)<br />

significantly improved OS vs placebo plus BSC in CRPC patients (pts) with<br />

bone mets (Parker et al. ECCO/ESMO 2011), and was shown to have an<br />

excellent safety pr<strong>of</strong>ile. Since D is an approved chemotherapy with<br />

demonstrated survival benefit for pts progressing after castrating hormone<br />

therapy, it is logical to explore combining Ra-223 plus D in pts with CRPC<br />

and bone mets. This study is being conducted in the Prostate Cancer<br />

<strong>Clinical</strong> Trials Consortium with an aim to establish the safe dose <strong>of</strong> Ra-223<br />

and D when used in combination in pts with bone mets with CRPC.<br />

Methods: This ongoing phase I/IIa trial (NCT01106352) is enrolling pts<br />

with bone mets from CRPC intended for treatment with D. D naïve pts are<br />

preferred, although up to 10 previous infusions <strong>of</strong> D are acceptable if D was<br />

well tolerated and the patient is in good condition and fulfills all eligibility<br />

criteria at study entry. Phase I dose-escalation study: minimum <strong>of</strong> 9 pts<br />

(max. 18 pts) will be included if no dose limiting toxicity (DLT) is observed.<br />

Dose escalation will follow a ‘3�3’ design, with no intra-patient dose<br />

escalation or overlapping <strong>of</strong> cohorts permitted. Ra-223 doses are 25 or 50<br />

kBq/kg, with number <strong>of</strong> Ra-223 injections (inj) (2, 4, 5, or 10) and interval<br />

between inj (3 or 6 wks) varying by escalation schema; D doses are 60 or 75<br />

mg/m2 q3wk. DLT will be assessed when 6 wks post-inj safety data are<br />

available after 1st Ra-223 �D inj. Phase IIa open-label expanded safety<br />

study: pts will be randomized 2:1 to the recommended dose <strong>of</strong> Ra-223 to<br />

be used with D or to treatment with D alone (approx. 42 pts will be<br />

included). Primary endpoint is to assess safety <strong>of</strong> combining Ra-223 with<br />

D. Exploratory efficacy endpoints for the open-label expanded safety<br />

portion <strong>of</strong> the study include change in disease-related biomarkers, time to<br />

1st radiological or clinical progression, CTC and pain assessment. Enrollment<br />

in dose-escalation portion <strong>of</strong> the study is complete; expanded safety<br />

cohort to begin enrolling in March 2012.<br />

TPS4696^ General Poster Session (Board #16G), Sun, 8:00 AM-12:00 PM<br />

First-line use <strong>of</strong> cabazitaxel in chemotherapy-naive patients with metastatic<br />

castration-resistant prostate cancer (mCRPC): A three-arm study in<br />

comparison with docetaxel. Presenting Author: Stephane Oudard, Georges<br />

Pompidou European Hospital, Paris, France<br />

Background: Docetaxel (D) in combination with prednisone (P) as first-line<br />

(1L) chemotherapy in patients (pts) with mCRPC is the current standard <strong>of</strong><br />

care. However, treatment is not curative and D-resistant disease typically<br />

develops. Cabazitaxel (Cbz) is a novel taxane active in D-sensitive and<br />

-resistant tumor models. <strong>Clinical</strong> activity <strong>of</strong> Cbz plus P (CbzP) was<br />

demonstrated in the Phase III TROPIC study in mCRPC pts previously<br />

treated with a D-containing regimen; CbzP showed a significant overall<br />

survival (OS) benefit vs mitoxantrone plus prednisone (median OS 15.1 vs<br />

12.7 months; HR 0.70; P � 0.0001). Therefore, it is <strong>of</strong> interest to<br />

determine if CbzP provides an OS advantage vs DP in 1L mCRPC pts.<br />

Methods: The phase III FIRSTANA study (NCT01308567) is a randomized,<br />

open-label, multinational trial in 1L mCRPC pts, designed to compare the<br />

efficacy <strong>of</strong> Cbz 25 mg/m² IV Q3W (Arm A) and Cbz 20 mg/m² IV Q3W (Arm<br />

B) vs D 75 mg/m2 IV Q3W (Arm C). P 10 mg PO QD is to be given<br />

concomitantly. Pts are stratified by ECOG PS (0–1 vs 2), measurable<br />

disease (yes/no) and region (depending on availability <strong>of</strong> Cbz as 2L). Pts<br />

with ECOG PS � 2, histologically/cytologically confirmed metastatic<br />

prostate adenocarcinoma, with no prior chemotherapy and with disease<br />

progression following medical or surgical castration are eligible. The<br />

primary endpoint is OS. Secondary endpoints include progression-free<br />

survival (PFS) (PCWG2 criteria), radiologic PFS, tumor response in measurable<br />

disease (RECIST 1.1), PSA response and PSA PFS, pain response and<br />

pain PFS, time to occurrence <strong>of</strong> any skeletal-related events, safety pr<strong>of</strong>ile<br />

and health-related quality <strong>of</strong> life. Cbz pharmacokinetics and pharmacogenomics<br />

will be assessed in pt subgroups. Pts will be treated until<br />

progression, unacceptable toxicity or pt request. Planned enrollment is<br />

1,170 pts; study size was calculated to achieve 90% power for OS. Study<br />

start was in May 2011; at January 2012, 219 pts were enrolled. The first<br />

DMC meeting recommended continuing the study without change.<br />

Genitourinary Cancer<br />

325s<br />

TPS4695 General Poster Session (Board #16F), Sun, 8:00 AM-12:00 PM<br />

A randomized, open-label, phase II study <strong>of</strong> MDV3100 alone or in<br />

combination with leuprolide and dutasteride as neoadjuvant therapy to<br />

prostatectomy in intermediate and high-risk prostate cancer. Presenting<br />

Author: Robert B. Montgomery, University <strong>of</strong> Washington School <strong>of</strong><br />

Medicine, Seattle, WA<br />

Background: MDV3100 is a potent androgen receptor (AR) signaling<br />

inhibitor (ARSI) that inhibits AR signaling via three mechanisms: inhibition<br />

<strong>of</strong> androgen binding to AR, inhibition <strong>of</strong> AR nuclear translocation, and<br />

inhibition <strong>of</strong> nuclear AR-DNA binding. In vivo, MDV3100 induces significant<br />

prostate cancer apoptosis, an effect not seen with anti-androgens. To<br />

date, the use <strong>of</strong> neoadjuvant androgen deprivation therapy has not led to an<br />

improvement in time to PSA progression (Soloway 2002; Aus 2002). While<br />

serum androgens may be suppressed using luteinizing hormone-releasing<br />

hormone agonists, intratumoral levels <strong>of</strong> androgens remain, driving continued<br />

AR signaling and prostate cancer survival. More effective inhibition <strong>of</strong><br />

AR signaling may improve local and systemic disease control. Methods:<br />

MDV3100-07 will assess the effect <strong>of</strong> 6 mos <strong>of</strong> neoadjuvant AR blockade<br />

with AR inhibition alone (MDV3100) or in combination with maximal<br />

suppression <strong>of</strong> androgens (MDV3100 �leuprolide [L] � dutasteride [D]).<br />

Eligible patients will have treatment-naive localized prostate cancer and be<br />

candidates for radical prostatectomy. Patients must have either PSA � 10<br />

ng/mL or Gleason score � 7(4� 3) with �3 cores containing tumor.<br />

Patients with evidence <strong>of</strong> metastatic/nodal disease are excluded. All<br />

patients receive MDV3100 (160 mg/d PO); those randomized to<br />

MDV3100�L�D therapy also receive L (22.5mg IM q3m) and D (0.5<br />

mg/day PO). Serum/tumor androgen levels will be serially assessed. Tissue<br />

from the diagnostic and prostatectomy specimens will be evaluated for<br />

androgen levels, AR signaling pr<strong>of</strong>iles, and selected markers <strong>of</strong> apoptosis<br />

and mitotic indices. The primary efficacy endpoint is pathological complete<br />

response (pCR) rate at time <strong>of</strong> radical prostatectomy. For each arm, the<br />

percent <strong>of</strong> patients who achieve a pCR will be compared to the percent pCR<br />

in patients treated with neoadjuvant leuprolide, estimated to be 5% in a<br />

mixed low-to-intermediate risk population. Target Accrual: 40 pts will be<br />

randomized 1:1 to MDV3100 or MDV3100�L�D therapy. Keywords:<br />

MDV3100, prostate cancer, androgen receptor, anti-androgen, Phase 2,<br />

neoadjuvant.<br />

TPS4697 General Poster Session (Board #16H), Sun, 8:00 AM-12:00 PM<br />

A phase II study <strong>of</strong> the androgen signaling inhibitor ARN-509 in patients<br />

with castration-resistant prostate cancer (CRPC). Presenting Author: Dana<br />

E. Rathkopf, Sidney Kimmel Center for Prostate and Urologic Cancers,<br />

Memorial Sloan-Kettering Cancer Center, New York, NY<br />

Background: ARN-509 is a novel small molecule androgen signaling<br />

inhibitor that impairs AR nuclear translocation and binding to DNA,<br />

inhibiting tumor growth and promoting apoptosis, with no partial agonist<br />

activity. Preclinical data suggests that the maximal therapeutic index <strong>of</strong><br />

ARN-509 can be achieved at low steady state plasma levels with minimal<br />

toxicity (Clegg et al, 2012). Enrollment in the Phase 1 dose escalation<br />

study <strong>of</strong> ARN-509 in patients with progressive CRPC with and without prior<br />

chemotherapy was completed in January 2012. The recommended Phase 2<br />

dose <strong>of</strong> 240 mg was determined based on safety, PSA kinetics, and<br />

pharmacokinetic and pharmacodynamic analysis (Rathkopf et al, GU<br />

ASCO, 2012). Methods: The primary objective <strong>of</strong> this Phase 2 study is to<br />

determine the PSA response at 12 weeks according to Prostate Cancer<br />

Working Group 2 (PCWG2) Criteria (Scher et al, 2008). Three expansion<br />

cohorts will enroll a total <strong>of</strong> 80-90 patients for treatment with 240 mg<br />

continuous oral ARN-509 daily. These cohorts include: 1) non-metastatic<br />

treatment-naïve CRPC (50 patients); 2) chemotherapy-naïve metastatic<br />

(m) CRPC (20 patients); and 3) chemotherapy-naïve, post abiraterone<br />

mCRPC (10-20 patients). The effect <strong>of</strong> food on the PK <strong>of</strong> ARN-509 and the<br />

effect <strong>of</strong> ARN-509 on ventricular repolarization will also be evaluated.<br />

Phase 2 enrollment is ongoing. DOD/PCF PCCTC trial sponsored by Aragon<br />

Pharmaceuticals. NCT01171898.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


326s Genitourinary Cancer<br />

TPS4698^ General Poster Session (Board #17A), Sun, 8:00 AM-12:00 PM<br />

TERRAIN: A randomized, double-blind, phase II study comparing MDV3100<br />

with bicalutamide (Bic) in men with metastatic castrate-resistant prostate<br />

cancer (CRPC). Presenting Author: Edwina S. Baskin-Bey, Astellas Pharma<br />

Europe, Ltd., Staines, United Kingdom<br />

Background: MDV3100 is a novel androgen receptor (AR) signaling inhibitor<br />

(ARSI) in clinical development for treatment <strong>of</strong> prostate cancer (PCa).<br />

Compared with Bic in nonclinical CRPC models, MDV3100 showed higher<br />

affinity AR binding, inhibited nuclear translocation and AR-DNA binding,<br />

showed no evidence <strong>of</strong> AR agonism, and caused tumor regression in<br />

Bic-resistant xenografts. MDV3100 has shown antitumor activity in men<br />

with advanced PCa. We present the study design <strong>of</strong> a trial comparing<br />

MDV3100 and Bic in men with progressive metastatic PCa. Methods:<br />

Multinational study (Table) with planned enrollment <strong>of</strong> 370 patients<br />

(randomized 1:1 to MDV3100 160 mg/d or Bic 50 mg/d). Inclusion criteria<br />

include metastatic (�2 bone lesions or s<strong>of</strong>t tissue disease at screening)<br />

progressive CRPC (�3 rising prostate-specific antigen [PSA] levels or new<br />

bone/s<strong>of</strong>t tissue disease), ongoing stable gonadotropin-releasing hormone<br />

(GnRH) analog therapy or surgical castration (serum testosterone �50<br />

ng/dL), Eastern Cooperative Oncology Group performance status 0–1, and<br />

life expectancy �1 year. Exclusion criteria include previous chemotherapy,<br />

current/prior antiandrogens (except if administered for �12 wk and<br />

discontinued no less than 6 mo before study). Patients will be stratified by<br />

time <strong>of</strong> bilateral orchiectomy or GnRH analog initiation (before/after<br />

diagnosis <strong>of</strong> metastases) and will receive treatment until occurrence <strong>of</strong><br />

radiographic progression/skeletal-related event, start <strong>of</strong> new antineoplastic<br />

therapy, or development <strong>of</strong> an adverse event requiring discontinuation.<br />

Results: The primary endpoint is progression-free survival; secondary<br />

endpoints are safety, PSA response, and time to PSA progression. Exploratory<br />

endpoints include circulating tumor cell conversion rate and quality <strong>of</strong><br />

life. Patients are currently enrolling. Conclusions: This ongoing, head-tohead,<br />

phase II trial is the first to prospectively assess whether MDV3100<br />

can provide improved antitumor effects vs Bic in men with metastatic<br />

CRPC.<br />

Country Sites, n<br />

Canada 4<br />

US-East 12<br />

US-Midwest 13<br />

US-South 11<br />

US-West 10<br />

France 9<br />

Germany 6<br />

Romania 3<br />

UK 5<br />

Belgium 5<br />

TPS4700 General Poster Session (Board #17C), Sun, 8:00 AM-12:00 PM<br />

Targeting castration resistant prostate cancer (CRPC) with autologous<br />

PSMA-directed CAR� T cells. Presenting Author: Susan F. Slovin, Sidney<br />

Kimmel Center for Prostate and Urologic Cancers, Memorial Sloan-<br />

Kettering Cancer Center, New York, NY<br />

Background: Based on our preclinical animal models, we initiated a phase I<br />

dose-escalating study to assess safety, dose requirement and targeting<br />

efficiency <strong>of</strong> genetically directed autologous human T cells targeted to<br />

Prostate Specific Membrane Antigen (PSMA). Our approach is based on the<br />

infusion <strong>of</strong> autologous PSMA-targeted T cells utilizing the P28z second<br />

generation chimeric antigen receptor (CAR) in patients (pts) with metastatic<br />

CRPC, following iv cyclophosphamide (Cy) (trial NCT01140373).<br />

For safety, the herpes simplex virus-1 thymidine kinase (hsvtk) gene is<br />

co-expressed with the P28z receptor, and renders T cells sensitive to<br />

ganciclovir for immediate T cell elimination if needed. The expression <strong>of</strong><br />

hsvtk enables PET imaging using radiolabeled FIAU to localize adoptively<br />

transferred T cells. The aims <strong>of</strong> the trial are to assess: (1) safety <strong>of</strong><br />

PSMA-targeted T cells; (2) biologic and anti-tumor effects; (3) T cell<br />

persistence at tumor site; and (4) immune response. Methods: Autologous T<br />

cells are activated from a leukapheresis product using anti-CD3/CD28<br />

Dynabeads. Release criteria include mean vector copy number by Q-PCR<br />

and vector identity by Southern blot, absence <strong>of</strong> Replication Competent<br />

Retrovirus and residual Dynabeads. Pts will be treated at 3 dose levels from<br />

107 to 108 CAR� T cells/kg. Four patients have been enrolled; 3 have been<br />

treated with 300mg/m2 <strong>of</strong> Cy one day before infusion <strong>of</strong> 107 CAR� T<br />

cells/kg. Pts underwent baseline and post treatment CT, bone and PET<br />

scans. Pts are followed weekly, then monthly with blood work including<br />

immune and vector sequence monitoring. Results: The first 3 pts within the<br />

first cohort were successfully treated without toxicity. Two had stable<br />

disease for greater than 6 months with the third patient having disease<br />

progression. There were no acute adverse events. Conclusion: We have<br />

established an ex vivo transduction, expansion and therapeutic protocol for<br />

the generation and testing <strong>of</strong> safe, clinical-grade, PSMA targeted T cells.<br />

Pts enrolled at the next dose level <strong>of</strong> 3x107CAR� T cells/kg will be<br />

assessed as described and by imaging the transduced T cell population<br />

using 18F-FIAU as a radiotracer. The data pertaining to the planned T cell<br />

imaging will be presented as well.<br />

TPS4699 General Poster Session (Board #17B), Sun, 8:00 AM-12:00 PM<br />

Prospect: A randomized, double-blind, phase III efficacy trial <strong>of</strong> PROST-<br />

VAC. Presenting Author: Olga Bandman, BN ImmunoTherapeutics, Mountain<br />

View, CA<br />

Background: PROSTVAC is a candidate cancer vaccine comprised <strong>of</strong> two<br />

recombinant poxviral vectors: vaccinia (V) and fowlpox (F), each with<br />

insertions <strong>of</strong> four human genes: PSA and three costimulatory molecules<br />

(TRICOM) - LFA-3, B7.1 and ICAM-1. This <strong>of</strong>f the shelf vaccine demonstrated<br />

a statistically significant overall survival (OS) benefit <strong>of</strong> 8.5 months<br />

while displaying a favorable side effect pr<strong>of</strong>ile in patients (pts) with<br />

asymptomatic to minimally symptomatic prostate cancer (mCRPC) in a<br />

randomized, placebo –controlled 122-pt Phase II trial. Data from this<br />

Phase II trial supported the design <strong>of</strong> a Phase III protocol that will rigorously<br />

test the hypothesis <strong>of</strong> OS benefit, as well as expand our understanding <strong>of</strong><br />

immune system response to cancer vaccines. Methods: 1200 pts will be<br />

randomized in a double-blind fashion to three arms: PROSTVAC,<br />

PROSTVAC�GM-CSF, or Placebo, at 1:1:1 ratio. A five-month treatment<br />

regimen will include a priming vaccination with PROSTVAC-V, and six<br />

booster vaccinations with PROSTVAC-F. Eligible pts will have asymptomatic<br />

or minimally symptomatic mCRPC, and progression despite androgen<br />

ablation and be chemotherapy-naïve. Pts with rapidly progressing disease<br />

will be excluded, as well as pts with risk factors for developing vacciniaassociated<br />

complications. The projected trial size is 400 pts per arm for at<br />

least 85% power. Primary endpoint is OS. The final analyses will be<br />

event-driven and will compare each active arm independently with placebo.<br />

Pts will be followed for 12 months after the projected number <strong>of</strong> events in<br />

each arm is realized. Secondary endpoint is proportion <strong>of</strong> event-free pts at 6<br />

months compared to placebo. A number <strong>of</strong> exploratory endpoints are<br />

planned, including immune response to immunizing antigen, non-vaccinecontained<br />

prostate antigens, tumor-associated antigens; changes in baseline<br />

biomarker levels and CTC levels, as well as characterization <strong>of</strong> T cell<br />

subpopulations. The thorough immune monitoring program would provide<br />

basis for future studies on the effects <strong>of</strong> cancer immunotherapy on immune<br />

system and facilitate search for potential biomarkers <strong>of</strong> such effects. The<br />

trial is currently open for enrollment. <strong>Clinical</strong>Trials.gov registry number:<br />

NCT00450463.<br />

TPS4701 General Poster Session (Board #17D), Sun, 8:00 AM-12:00 PM<br />

Randomized phase II clinical trial to assess MUC1 specific immune<br />

response to L-BLP25 vaccine in addition to standard therapy in newly<br />

diagnosed high-risk prostate cancer. Presenting Author: Nishith Singh,<br />

Laboratory <strong>of</strong> Tumor Immunology and Biology and Medical Oncology<br />

Branch, National Cancer Institute, Bethesda, MD<br />

Background: In high-risk prostate cancer, radiation therapy (RT) � androgen<br />

deprivation therapy (ADT) improve survival. Nonetheless, 10-year<br />

disease specific mortality is about 25%. L-BLP25 is a cancer vaccine<br />

containing the BLP25 lipopeptide that targets MUC1 tumor antigen. It may<br />

enhance immune targeting <strong>of</strong> cells that express MUC1 (e.g. prostate<br />

cancer). In murine models, RT synergizes with vaccine-induced anti-cancer<br />

immunity (augments T-cell mediated cancer cytolysis, up-regulates cellular<br />

Fas and co-stimulatory/adhesion molecules). ADT augments T-cell<br />

trafficking to prostate. Immune response to combining the three (L-BLP25<br />

� RT � ADT) is not known. The current trial intends to study this immune<br />

response to L-BLP25 � RT � ADT and compare it to RT�ADT alone. Using<br />

ELISPOT, endo-rectal MRI and serial prostate biopsies, this trial was<br />

designed to correlate systemic immune response with changes in tumor<br />

imaging and/or tumor microenvironment after treatment with L-BLP25.<br />

This trial may provide insight into immune response biomarkers that are<br />

most appropriate in this setting. Methods: A randomized (1:1), open-label,<br />

phase II trial <strong>of</strong> 42 pts is planned. Eligibility: Adult males with newly<br />

diagnosed high-risk prostate cancer (T3 or Gleason � 8 or seminal vesicle<br />

involvement or N1 or PSA�20) and HLA-A2/A3 positivity (to allow for<br />

ELISPOT analysis). The vaccine arm will receive RT � 2-year ADT �<br />

L-BLP25. Standard arm will receive RT � 2-year ADT. L-BLP25 vaccine<br />

schedule: biweekly X 5 starting with neo-adjuvant ADT, then 6 weekly X 4<br />

starting with RT. A single 300mg/m2 cyclophosphamide infusion (decreases<br />

suppressor T-cells) will be given 3 days before L-BLP25 to enhance<br />

immune response in the vaccine arm. The impact <strong>of</strong> L-BLP25 � RT�ADT<br />

on MUC-1-specific systemic immune response will be determined using<br />

interval peripheral blood ELISPOT assays. Endo-rectal coil MRI will be<br />

done before and after treatment to study prostate signal changes for<br />

correlative and predictive analysis. MRI-UltraSound guided lesion-targeted<br />

serial prostate biopsies will be obtained to assess immune response in<br />

tumor microenvironment. Two pts have been enrolled.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


LBA5000 Oral Abstract Session, Sat, 3:00 PM-6:00 PM<br />

Randomized phase III study <strong>of</strong> erlotinib versus observation in patients with<br />

no evidence <strong>of</strong> disease progression after first-line platin-based chemotherapy<br />

for ovarian carcinoma: A GCIG and EORTC-GCG study. Presenting<br />

Author: Ignace B. Vergote, University Hospital Leuven and KU Leuven,<br />

Leuven, Belgium<br />

The full, final text <strong>of</strong> this abstract will be available at<br />

abstract.asco.org at 12:01 AM (EDT) on Saturday, June 2,<br />

2012, and in the <strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong> online<br />

supplement to the June 20, 2012, issue <strong>of</strong> Journal <strong>of</strong><br />

<strong>Clinical</strong> Oncology. Onsite at the <strong>Meeting</strong>, this abstract will<br />

be printed in the Saturday edition <strong>of</strong> ASCO Daily News.<br />

LBA5002^ Oral Abstract Session, Sat, 3:00 PM-6:00 PM<br />

AURELIA: A randomized phase III trial evaluating bevacizumab (BEV) plus<br />

chemotherapy (CT) for platinum (PT)-resistant recurrent ovarian cancer<br />

(OC). Presenting Author: Eric Pujade-Lauraine, GINECO and Université<br />

Paris Descartes, Paris, France<br />

The full, final text <strong>of</strong> this abstract will be available at<br />

abstract.asco.org at 12:01 AM (EDT) on Saturday, June 2,<br />

2012, and in the <strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong> online<br />

supplement to the June 20, 2012, issue <strong>of</strong> Journal <strong>of</strong><br />

<strong>Clinical</strong> Oncology. Onsite at the <strong>Meeting</strong>, this abstract will<br />

be printed in the Saturday edition <strong>of</strong> ASCO Daily News.<br />

Gynecologic Cancer<br />

327s<br />

5001 Oral Abstract Session, Sat, 3:00 PM-6:00 PM<br />

Olaparib plus paclitaxel plus carboplatin (P/C) followed by olaparib maintenance<br />

treatment in patients (pts) with platinum-sensitive recurrent serous<br />

ovarian cancer (PSR SOC): A randomized, open-label phase II study.<br />

Presenting Author: Amit M. Oza, Princess Margaret Hospital, Toronto, ON,<br />

Canada<br />

Background: The oral PARP inhibitor olaparib has shown antitumor activity<br />

in pts with SOC. Our multicenter study compared the efficacy <strong>of</strong> (Arm A)<br />

olaparib capsules plus P/C for 6 cycles then maintenance olaparib<br />

monotherapy vs (Arm B) P/C alone for 6 cycles and no further therapy in pts<br />

with PSR SOC (NCT01081951). Methods: Pts received 6 x 21-day(d)<br />

cycles <strong>of</strong> olaparib (200 mg bid, d1–10/21) � P (175 mg/m2 iv, d1) � C<br />

(AUC4 iv, d1), then olaparib monotherapy as maintenance (400 mg bid,<br />

continuous) (Arm A), or 6 x 21d cycles <strong>of</strong> P (175 mg/m2 iv, d1) � C (AUC6<br />

iv, d1) then no further therapy (Arm B), until progression. Randomization<br />

(1:1) was stratified by number <strong>of</strong> platinum treatments and platinum-free<br />

interval. Primary endpoint: progression-free survival (PFS) by central review<br />

(RECIST 1.1). Secondary endpoints: overall survival (OS), objective response<br />

rate (ORR), safety. Archival tissue was collected where available for<br />

analysis <strong>of</strong> biomarker correlation. Results: Of 162 pts randomized (n�81<br />

per arm), 156 received treatment (Arm A, n�81; Arm B, n�75) and 121<br />

began the maintenance/no further therapy phase (Arm A, n�66; Arm B,<br />

n�55). Olaparib � P/C (AUC4) followed by maintenance olaparib showed a<br />

significant improvement in PFS vs P/C (AUC6) alone (HR � 0.51, 95% CI<br />

0.34, 0.77; P�0.0012; median � 12.2 vs 9.6 months). OS data are<br />

immature (total events: 14%). ORR was similar for Arm A and Arm B (64 vs<br />

58%). Most common AEs during the combination phase were alopecia (74<br />

vs 59%), nausea (69 vs 57%) and fatigue (64 vs 57%) for Arm A vs Arm B,<br />

respectively. Pts with grade �3 AEs (65 vs 57%), serious AEs (SAEs: 15 vs<br />

21%) and AEs leading to treatment discontinuation (19 vs 16%) were<br />

similar for Arm A vs Arm B. Most common AEs during maintenance/no<br />

further therapy were nausea (50 vs 6%) and vomiting (29 vs 7%). 29 vs<br />

16% <strong>of</strong> pts had grade �3 AEs, 9 vs 7% had SAEs and 8% vs N/A<br />

discontinued due to AEs in the olaparib vs no treatment arms, respectively.<br />

There were no fatal AEs. Conclusions: In pts with PSR SOC, olaparib plus<br />

P/C (AUC4) followed by olaparib 400 mg bid monotherapy maintenance<br />

treatment resulted in a significant improvement in PFS vs P/C (AUC6)<br />

alone.<br />

5003 Oral Abstract Session, Sat, 3:00 PM-6:00 PM<br />

Long-term follow-up <strong>of</strong> a randomized trial comparing conventional paclitaxel<br />

and carboplatin with dose-dense weekly paclitaxel and carboplatin in<br />

women with advanced epithelial ovarian, fallopian tube, or primary peritoneal<br />

cancer: JGOG 3016 trial. Presenting Author: Noriyuki Katsumata,<br />

Nippon Medical School Musashikosugi Hospital, Kanagawa, Japan<br />

Background: The primary analysis <strong>of</strong> the JGOG3016 trial (Lancet 2009,<br />

374:1331) showed that dose-dense weekly administration with paclitaxel<br />

and carboplatin (dd-TC) demonstrated statistically significant efficiency<br />

over tri-weekly administration withTC (c-TC) as first-line chemotherapy in<br />

patients with stage II-IV epithelial ovarian, fallopian tube or primary<br />

peritoneal cancer. We report the long-term follow-up results on progressionfree<br />

survival (PFS) and overall survival (OS). Methods: Patients with stage II<br />

to IV ovarian cancer were randomly assigned to receive c-TC (carboplatin<br />

AUC 6 and paclitaxel 180 mg/m2 on day 1) or dd-TC (carboplatin AUC 6 on<br />

day 1 and paclitaxel 80 mg/m2 on day 1, 8, 15). The treatments were<br />

repeated every 3 weeks for six cycles; in responding patients, additional<br />

three cycles were administered. Results: The analysis included eligible 631<br />

patients. At 6.4 years <strong>of</strong> median follow-up, there continues to be a highly<br />

statistically significant improvement in median PFS in favor <strong>of</strong> the dd-TC<br />

group compared with the c-TC group (28.1 vs. 17.5 months, hazard ratio<br />

[HR] 0.75, 95% CI, 0.62-0.91; P�0.0037). Median survival has not yet<br />

been reached in the dd-TC group, and OS at 5 years was higher in the dd-TC<br />

group than the c-TC group (58.6% vs. 51.0%, HR 0.79, 95% CI,<br />

0.63-0.99; P �0.0448). Conclusions: The dd-TC improves long-term PFS<br />

and OS in patients with advanced epithelial ovarian cancer.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


328s Gynecologic Cancer<br />

5004 Oral Abstract Session, Sat, 3:00 PM-6:00 PM<br />

Prospective assessment <strong>of</strong> cost, morbidity, and survival associated with<br />

lymphadenectomy in low-risk endometrial cancer. Presenting Author: Sean<br />

Christopher Dowdy, Mayo Clinic, Rochester, MN<br />

Background: Since 1999, patients with low-risk endometrial cancer (EC) as<br />

defined by the Mayo criteria have preferably not undergone lymphadenectomy<br />

(LND) at our institution. We assess survival, sites <strong>of</strong> recurrence,<br />

morbidity, and cost per up-staged case in this low-risk cohort. Methods:<br />

Consecutive patients with Mayo-defined low-risk EC managed without<br />

(non-LND) and with LND were compared. Cause-specific survival (CSS) was<br />

estimated using the Kaplan-Meier method and compared using the<br />

log-rank test. 30-day cost analyses were equated to 2010 Medicare dollars.<br />

Results: Among 1,393 consecutive surgically managed EC cases, 385<br />

(27.6%) met the Mayo low-risk criteria, accounting for 34.1% <strong>of</strong> type I EC.<br />

The 5-year CSS <strong>of</strong> the low-risk cases was 98.6 %. There were 80 LND cases<br />

(median # nodes, 29) and 305 non-LND cases. Complications within 30<br />

days occurred in 37.5% and 19.3% <strong>of</strong> LND and non-LND cases, respectively<br />

(P�0.001). Nodal metastasis was identified in a single LND case<br />

(1.3%). There were 11 recurrences, 6 <strong>of</strong> which were vaginal. Not a single<br />

recurrence was detected in the pelvic or paraaortic nodal areas in these<br />

385 patients, with a median follow-up <strong>of</strong> 5.4 years. The estimated<br />

prevalence (combining surgery and surveillance) <strong>of</strong> lymph node metastasis<br />

was 0.3%. The 5-year CSS in LND and non-LND cases was 97.3% and<br />

99.0%, respectively (P�0.32). Patients were more than seven times as<br />

likely to die <strong>of</strong> co-morbidities than from EC. The 30-day median cost <strong>of</strong> care<br />

was $15,678 for LND cases compared to $11,028 for non-LND cases<br />

(P�0.001). The estimated cost per up-staged low-risk case was $439,990<br />

if performed via endoscopy and $327,866 via laparotomy. If the 305<br />

non-LND cases had been subjected to LND, an estimated additional<br />

$1,418,189 would have been expended. Conclusions: For patients with<br />

low-risk EC as defined by the Mayo criteria, lymphadenectomy dramatically<br />

increases morbidity and 30-day cost <strong>of</strong> care without discernible short- or<br />

long-term benefits: CSS was 99% with a 0.3% rate <strong>of</strong> nodal metastasis. In<br />

these low-risk patients, hysterectomy with salpingo-oophorectomy alone is<br />

appropriate surgical management and should be standard <strong>of</strong> care.<br />

5006 Oral Abstract Session, Sat, 3:00 PM-6:00 PM<br />

A randomized, phase III trial <strong>of</strong> paclitaxel plus carboplatin (TC) versus<br />

paclitaxel plus cisplatin (TP) in stage IVb, persistent or recurrent cervical<br />

cancer: Japan <strong>Clinical</strong> Oncology Group study (JCOG0505). Presenting<br />

Author: Ryo Kitagawa, NTT Medical Center Tokyo, Tokyo, Japan<br />

Background: TC is a less toxic regimen in terms <strong>of</strong> milder nephropathy,<br />

neuropathy and no need <strong>of</strong> hospitalization. This multicenter phase III trial<br />

was designed to evaluate the clinical benefits <strong>of</strong> TC compared with TP<br />

which is current standard chemotherapy for stage IVB or recurrent cervical<br />

cancer. Methods: Patients (pts) with stage IVB or recurrent cervical cancer;<br />

not amenable to curative therapy; 0-1 prior platinum; no prior taxanes; were<br />

randomized with minimization method to either TP (T 135 mg/m2 24h d1<br />

� P 50 mg/m2 2h d2) or TC (T 175 mg/m2 3h d1 � C AUC5 1h d1), both<br />

for maximum 6 cycles every 21 days. Primary endpoint was overall survival<br />

(OS). Secondary endpoints were progression-free survival (PFS), toxicities,<br />

and the proportion <strong>of</strong> non-hospitalization periods (NHP) as a surrogate for<br />

QoL. The trial was powered at least 70% to confirm the non-inferiority <strong>of</strong> TC<br />

to TP (threshold hazard ratio [HR] 1.29) in terms <strong>of</strong> OS, and the planned<br />

sample size was 250 pts with one-sided alpha 5%. HR is estimated by a<br />

stratified Cox regression. Results: From 2/06 to 11/09, 253 pts were<br />

enrolled. 71% pts <strong>of</strong> TP arm and TC arm each received 6 cycles. Median<br />

follow-up is 17.4 mo. Results are as below. As an alpha level for an interim<br />

analysis was less than 0.0001, significance level for the final analysis is<br />

approximately 5% even after the multiplicity adjustment. Conclusions: This<br />

first randomized controlled trial comparing carboplatin doublet with cisplatin<br />

doublet showed significant non-inferiority <strong>of</strong> TC in terms <strong>of</strong> OS. More<br />

feasible and less toxic TC can be recommended as the new standard<br />

treatment for stage IVB or recurrent cervical cancer.<br />

TP TC<br />

OS (median) HR 0.99 (multiplicity adjusted 90%CI: 18.3 mo 17.5 mo<br />

0.79-1.25); noninferiority p�0.032<br />

PFS (median) HR 1.04 (95%CI: 0.80-1.35) 6.90 mo 6.21 mo<br />

Neutropenia G3-4 85.1% 76.4%<br />

Thrombocytopenia G3-4 3.3% 23.6%<br />

Febrile neutropenia G3-4 16.0% 7.3%<br />

Creatinine G2-4 9.8% 4.1%<br />

Neuropathy (motor) G3-4 0.8% 2.4%<br />

Neuropathy (sensory) G3-4 0% 4.9%<br />

Early treatment discontinuation (toxicity-related) 11.4% 10.0%<br />

NHP (p


5008 Oral Abstract Session, Sat, 3:00 PM-6:00 PM<br />

Survival after radiation therapy for early-stage endometrial carcinoma: The<br />

Oslo study revisited after up to 43 years <strong>of</strong> follow-up. Presenting Author:<br />

Kristina Lindemann, Akershus University Hospital, Lørenskog, Norway<br />

Background: There is an ongoing debate regarding the benefit <strong>of</strong> radiation in<br />

patients with early stage endometrial carcinoma. Data on long time risk<br />

conferred by radiation is scarce. This study is a long-term follow-up on<br />

survival and secondary cancers <strong>of</strong> a previously published randomized study<br />

(Aalders J. et al., Obstet Gynecol 1980; 56: 419-27). Methods: Between<br />

1968 and 1974, 568 patients with endometrial cancer FIGO stage I<br />

primarily treated with abdominal hysterectomy and bilateral salpingooophorectomy<br />

were included in the study. Patients were postoperatively<br />

randomized to receive either vaginal radium brachytherapy followed by<br />

external pelvic radiation 40 Gy (N�288) or brachytherapy alone (N�280).<br />

Survival data and data on incident secondary cancers were obtained by<br />

individual linkage to the Registry <strong>of</strong> Statistics Norway and Cancer Registry<br />

<strong>of</strong> Norway. By the end <strong>of</strong> follow-up at 1 November 2011, 45 (7.9%)<br />

patients were still alive. We used Cox proportional hazards model to<br />

estimate hazard ratios (HR) with 95% confidence intervals (95% CI). We<br />

also conducted analyses stratified by age groups. Results: After median 21<br />

(range 0-43.4) years <strong>of</strong> follow-up there was no significant difference in<br />

overall survival or relapse free survival between treatment arms with HR <strong>of</strong><br />

1.12 (95% CI: 0.95-1.33) and HR 0.88 (95% CI: 0.55-1.40), respectively.<br />

Patients treated with external radiation had significantly lower risk <strong>of</strong><br />

developing locoregional relapse (p�0.001). However, women younger than<br />

60 years had a significant poorer survival after external radiation (HR 1.36;<br />

95% Cl: 1.06-1.76). In this patient group the risk <strong>of</strong> secondary cancer was<br />

significantly increased (HR 1.9; 95% CI: 1.23-3.03). Conclusions: We<br />

observed no survival benefit <strong>of</strong> external pelvic radiation in early stage<br />

endometrial carcinoma. In women younger than 60 years, pelvic radiation<br />

decreased survival, probably due to increased risk <strong>of</strong> subsequent second<br />

neoplasms. Adjuvant external radiotherapy cannot be recommended to this<br />

patient group. Those who have received such treatment might eventually<br />

benefit from prolonged post treatment surveillance with respect to secondary<br />

cancer.<br />

5010 <strong>Clinical</strong> Science Symposium, Sat, 8:00 AM-9:30 AM<br />

Molecular determinants <strong>of</strong> outcome with mTOR inhibition in endometrial<br />

cancer (EC). Presenting Author: Helen Mackay, Princess Margaret Hospital,<br />

University Health Network, Toronto, ON, Canada<br />

Background: Deregulation <strong>of</strong> PI3K/AKT/mTOR signaling plays a significant<br />

role in EC biology. NCIC CTG has completed three phase II trials <strong>of</strong> 2 mTOR<br />

inhibitors (temsirolimus and ridaforolimus) in EC (IND 160A, IND 160B,<br />

IND192) demonstrating anti-tumor activity. PTEN expression, however,<br />

was not associated with response. In this study we are conducting<br />

additional molecular analyses on samples obtained from patients (pts)<br />

participating in these trials to identify mutations or other findings associated<br />

with mTOR response. Methods: Formalin fixed paraffin embedded<br />

(FFPE) tumor samples were collected from pts treated on the three trials.<br />

Tumor DNA was isolated and mutational pr<strong>of</strong>iling (MP) was performed<br />

using theOncoCarta Panel v1.0 which can detect 238 mutations in 19<br />

oncogenes including AKT1, 2, BRAF, CDK-4, EGFR, ERBB2, MET, H-, K-,<br />

N- RAS, PDGFRA, PIK3CA, RET. All mutations were verified by Sanger<br />

sequencing. For each gene found to be mutated in at least 1 patient, the<br />

relationship between presence/absence <strong>of</strong> mutations in that gene and<br />

objective anti-tumor response (RR) was assessed (RR vs. no response; early<br />

progression (PD) vs. no PD) using Fisher’s exact test. In addition, the<br />

presence <strong>of</strong> any mutation vs. no mutations was assessed in relationship to<br />

the same clinical outcomes. Results: MP was feasible in 73 <strong>of</strong> the 94<br />

eligible pts treated on the three trials. 44% (32 pts) had a mutation in at<br />

least one gene: 21 pts (29%) in PIK3CA, 10 (14%) KRAS, 4 (5%) MET, 3<br />

(4%) NRAS, 3 (4%) AKT and 1(1%) EGFR. 9 pts (12%) had � 1 mutation.<br />

No significant correlations were seen, in individual trials or within the<br />

pooled data set <strong>of</strong> 3 studies, between the presence/absence <strong>of</strong> any<br />

mutation and RR (p�1.0) or early PD (p�1.0). No significant association<br />

was seen between the presence <strong>of</strong> mutations within individual genes and<br />

RR or PD (including PIK3CA). Of interest, no response was observed in any<br />

<strong>of</strong> the 13 pts with a Ras mutation (non-significant). Additional analyses<br />

involving gene expression (including stathmin) and correlation <strong>of</strong> MP with<br />

tumor morphology are underway. Conclusions: Mutations are common in EC<br />

but we have not identified a statistical association between presence <strong>of</strong><br />

mutations in PIK3CA or any other gene and response to mTOR inhibition.<br />

Further analyses are ongoing.<br />

Gynecologic Cancer<br />

329s<br />

5009 <strong>Clinical</strong> Science Symposium, Sat, 8:00 AM-9:30 AM<br />

Impact <strong>of</strong> metformin use on risk <strong>of</strong> recurrence in high-grade endometrial<br />

cancers. Presenting Author: Emily Meichun Ko, University <strong>of</strong> North Carolina at<br />

Chapel Hill, Chapel Hill, NC<br />

Background: Obesity and diabetes (DM) have been linked to increased recurrence<br />

rates and worse survival in endometrial cancer. Metformin is widely used as the<br />

first line treatment for type II DM. Recent epidemiological evidence suggests<br />

that metformin may lower cancer risk and reduce cancer-related deaths among<br />

DM patients. However, no studies have assessed the effect <strong>of</strong> metformin use on<br />

endometrial cancer outcomes. Methods: A retrospective cohort analysis was<br />

conducted <strong>of</strong> all grade 3 endometrial cancer patients with DM at a single<br />

institution from 2005-2010. <strong>Clinical</strong>-pathologic data was abstracted, including<br />

metformin, insulin, sulfonamide and thiazolidinedione use at the time <strong>of</strong> cancer<br />

diagnosis. Statistical analysis were performed using summary statistics and<br />

multivariate cox analysis, with two tailed p-values �0.05 considered significant.<br />

Results: 55/329 patients with grade 3 cancer had DM. Mean age was 68.3 (sd<br />

8.4), and BMI 35.7 (sd 9.4). Stage distribution consisted <strong>of</strong> 58.2% stage I,<br />

7.3% stage II, 27.3% stage III, and 7.3% stage IV. Histology included 41.8%<br />

endometrioid, and 58.2% serous or clear cell. 56.4% <strong>of</strong> diabetics used<br />

metformin and 43.6% did not. Mean follow-up was 21.6 months (sd 15.0). After<br />

adjusting for BMI, myometrial invasion, stage, histology, adjuvant treatment,<br />

and non-metformin DM medications, metformin use was significantly associated<br />

with decreased cancer recurrence (HR 0.06, 95CI 0.01, 0.40; p�0.004).<br />

Conclusions: Metformin use was associated with a decreased risk <strong>of</strong> recurrence in<br />

high-grade endometrial cancer, suggesting that it may have a role as adjuvant<br />

and maintenance therapy for this obesity-driven disease.<br />

Metformin user (%) Metformin non-user (%)<br />

n�31 n�24<br />

Age 67.5 (9.1) 69.4 (7.4)<br />

BMI<br />

Stage<br />

35.4 (8.5) 36.2 (10.5)<br />

I 15 (48.4) 17 (70.8)<br />

II 3 (9.7) 1 (4.2)<br />

III 9 (29.0) 6 (25.0)<br />

IV<br />

Histology<br />

4 (12.9) 0 (0)<br />

Endometrioid 11 (35.5) 12 (50)<br />

Serous � Clear cell<br />

Other medications<br />

20 (64.5) 12 (50)<br />

Sulfonamide (Sulfo) 5 (16.1) 8 (0)<br />

Thiazolidinedione (Thz) 2 (6.5) 0 (0)<br />

Sulfo � Thz 2 (6.5) 2 (8.3)<br />

Insulin 0 (0) 4 (16.7)<br />

Insulin � Sulfo 1 (3.2) 0 (0)<br />

Insulin � Thz<br />

Adjuvant treatment<br />

0 (0) 2 (8.3)<br />

Chemotherapy 4 (12.9) 3 (12.5)<br />

Radiation 7 (22.6) 4 (16.7)<br />

Chemo � Radiation 11 (35.5) 7 (29.2)<br />

5011 <strong>Clinical</strong> Science Symposium, Sat, 8:00 AM-9:30 AM<br />

KRAS and EGFR mutations to distinguish adenocarcinomas and squamous<br />

cell carcinomas <strong>of</strong> the cervix. Presenting Author: Alexi Wright, Dana-Farber<br />

Cancer Institute, Boston, MA<br />

Background: Cervical adenocarcinomas (AC) have higher rates <strong>of</strong> recurrence<br />

and distant metastasis, compared with squamous cell carcinomas (SCC),<br />

and decreased survival in advanced disease. Yet, tailored treatments for<br />

cervical cancers have not emerged. The aim <strong>of</strong> this study was to compare<br />

the frequency and type <strong>of</strong> somatic mutations in cervical AC and SCC to<br />

identify novel therapeutic targets for both subtypes. Methods: Tumors from<br />

61 patients with cervical cancer (36 AC, 25 SCC) underwent genomic<br />

pr<strong>of</strong>iling by OncoMap, a multiplexed mass spectrometric genotyping<br />

technology that interrogates more than 400 known mutations in 33 cancer<br />

genes. Results: Overall, 31/61 (50.8%) tumors harbored candidate mutations,<br />

and 6/61 (9.8%) had �2 mutations. PIK3CA mutations were present<br />

in 21/64 (34.4%) <strong>of</strong> cervical cancers, with a trend towards higher rates in<br />

SCC compared with ACC (21/25 or 48.0% vs. 9/27 or 33.3%, p�0.10).<br />

KRAS mutations were present in 6/31 (16.7%) <strong>of</strong> AC, but none <strong>of</strong> the SCC<br />

(0%; 0/25). EGFR mutations were present in 9/25 (36.0%) <strong>of</strong> SCC,<br />

including G719S, but none <strong>of</strong> AC (0%; 0/31). Conclusions: The identification<br />

<strong>of</strong> distinct genomic alterations in SCC and AC <strong>of</strong> the cervix suggest<br />

different therapeutic rationales for each subtype. EGFR amplification in<br />

cervical SCC has been previously reported, but this is the first identification,<br />

to our knowledge, <strong>of</strong> activating EGFR mutations in cervical SCC.<br />

Together, this suggests that EGFR-inhibitors might be useful in selected<br />

patients with cervical SCC. Similarly, activating mutations in KRAS and<br />

PIK3CA in AC suggest that inhibition <strong>of</strong> the MAPK pathway (MEK<br />

inhibitors) and/or PI3K pathway may be beneficial in this subtype. Future<br />

studies should include more comprehensive genomic pr<strong>of</strong>iling strategies,<br />

such as targeted massively parallel sequencing, to detect multiple types <strong>of</strong><br />

genomic alterations.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


330s Gynecologic Cancer<br />

5012 Poster Discussion Session (Board #1), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

A phase II, open-label, multicenter study <strong>of</strong> IMC-1121B (ramucirumab;<br />

RAM) monotherapy in the treatment <strong>of</strong> persistent or recurrent epithelial<br />

ovarian (EOC), fallopian tube (FTC), or primary peritoneal (PPC) carcinoma<br />

(CP12-0711/NCT00721162). Presenting Author: Richard T. Penson,<br />

Massachusetts General Hospital, Boston, MA<br />

Background: VEGF receptor-mediated-signaling contributes to ovarian cancer<br />

pathogenesis. Elevated VEGF expression and serum levels are associated<br />

with poor clinical outcomes. We investigated RAM, a fully human<br />

VEGFR-2 antagonist antibody, in patients (pts) with persistent or recurrent<br />

EOC/FTC/PPC. Methods: Adult women with EOC/FTC/PPC who had completed<br />

�1 platinum (P)-based chemotherapeutic (ct) regimen and had a<br />

P-free interval (PFI) <strong>of</strong> �12 months (m), progression on, or persistent<br />

disease after P-based therapy were eligible. Any number <strong>of</strong> prior ct<br />

regimens was allowed. ECOG PS 0-1 and adequate organ function were<br />

required. Pts received 8 mg/kg RAM IV every 2 weeks. Primary endpoints<br />

were progression-free survival at 6m (PFS-6) and confirmed objective<br />

response rate (ORR) by RECIST 1.0. Results: 60 pts were treated; 1<br />

remains on study as <strong>of</strong> Dec 2011. Median age was 62 years (range 27-80).<br />

Median number <strong>of</strong> prior regimens was 3 (range 1– 14). 51 pts (85%)<br />

received � 2 prior regimens; 25 pts (42%) received �3 prior regimens. 45<br />

pts (75%) were P resistant or refractory, with 65% (39 pts) serous tumors.<br />

PFS-6: 34.2% (95% CI: 21.7% – 47%). Best overall response: 3 PR (5%),<br />

34 SD (57%), 20 PD (33%) and 3 not evaluable (5%). Median duration <strong>of</strong><br />

PR: 5.6m (3.7, 5.6, 17.5); median PFS: 3.5m (95% CI: 2.3 – 5.3).<br />

Median OS: 11.1m (95% CI: 8.3 – 17.0). No unexpected toxicities were<br />

observed. Grade (G) 3 adverse events (AEs) observed in �5% <strong>of</strong> pts were:<br />

headache (10%) and fatigue (8%). No G4 AEs were observed in �5% <strong>of</strong><br />

pts. 5 deaths occurred on RAM or within 30 days <strong>of</strong> discontinuation; 4 due<br />

to PD, and 1 due to intestinal perforation. 1 G4 bowel perforation and one<br />

G4 colo-vaginal fistula were noted. All 3 cases <strong>of</strong> perforation/fistula<br />

occurred in the setting <strong>of</strong> progressive, large-volume disease. Correlative<br />

biomarker studies are ongoing to identify patients most likely to benefit.<br />

Conclusions: Ramucirumab was reasonably tolerated and demonstrated<br />

single-agent activity in persistent or recurrent ovarian carcinoma, with<br />

approximately one-third <strong>of</strong> patients progression free at 6 months.<br />

5014 Poster Discussion Session (Board #3), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Phase I trial <strong>of</strong> metronomic oral topotecan in combination with pazopanib<br />

utilizing a daily dosing schedule to treat recurrent or persistent gynecologic<br />

tumors. Presenting Author: Todd D. Tillmanns, The West Clinic, Memphis,<br />

TN<br />

Background: Metronomic oral topotecan has antiangiogenic properties. Oral<br />

pazopanib is also a potent inhibitor <strong>of</strong> VEGF angiogenesis with clinical<br />

benefit. This study investigated the safety and efficacy <strong>of</strong> daily topotecan<br />

and pazopanib. Methods: This phase I, single-center, open-label, dose<br />

ranging study comprised 28-day cycles <strong>of</strong> daily oral topotecan/pazopanib at<br />

dose levels <strong>of</strong> 0.50/400, 0.25/800, 0.25/600, and 0.25/400 mg. A<br />

standard 3�3 dose escalation design was used. Dose limiting toxicity<br />

(DLT) was defined as � grade 3 adverse event (AE) occurring in cycle 1,<br />

noncompletion <strong>of</strong> cycle 1 at prescribed dose, or inability to start cycle 2 as<br />

scheduled due to toxicities. Imaging was conducted after every 2 cycles for<br />

response assessment. Confirmed responses were evaluated per RECIST<br />

1.0. Results: 25 extensively pretreated patients (pts) with gynecologic<br />

tumors (6 cervical, 7 endometrial, 8 ovarian, 4 other) were enrolled. Mean<br />

age was 61 years; 19 Caucasian, 6 African <strong>American</strong>. Median number <strong>of</strong><br />

cycles received was 4 (range 1-19). DLTs occurred � 2 pts for A (2/6), B<br />

(2/7), C (3/7). For level D, 1/5 had a DLT, but one dose level C pt with a DLT<br />

received level D dosing due to unavailability <strong>of</strong> pazopanib. 9/25 pts had any<br />

serious adverse event (SAE), and respiratory SAEs were most common<br />

(4/16 SAEs). Nausea, fatigue, dysgeusia, diarrhea, and vomiting were the<br />

most common conditions reported as non-serious AEs. The Table shows<br />

best overall response (28%) by dose level. Conclusions: DLTs in dose levels<br />

C and D were effectively managed with minor dose and schedule adjustments.<br />

4 pts with DLTs at these doses remained on treatment for 4, 5, and<br />

6 months, with one pt still on treatment at 9 months. Given the initial<br />

biologically favorable signal in this heavily pretreated group and low<br />

toxicity, we recommend a randomized phase II trial to define efficacy using<br />

the 0.25/600 regimen allowing for dose reduction if needed.<br />

Dose level CR PR SD PD NE<br />

A 0.50/400 0 0 2 (33.3%) 4 (66.7%) 0<br />

B 0.25/800 2 (28.6%) 2 (28.6%) 0 1 (14.3%) 2 (28.6%)<br />

C 0.25/600 1 (14.3%) 0 1 (14.3%) 4 (57.1%) 1 (14.3%)<br />

D 0.25/400 1 (20.0%) 1 (20.0%) 0 0 3 (60.0%)<br />

Overall 4 (16.0%) 3 (12.0%) 3 (12.0%) 9 (36.0%) 6 (24.0%)<br />

5013 Poster Discussion Session (Board #2), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Phase II trial <strong>of</strong> bevacizumab and pemetrexed for recurrent or persistent<br />

epithelial ovarian, fallopian tube, or primary peritoneal cancer. Presenting<br />

Author: Andrea R. Hagemann, Washington University School <strong>of</strong> Medicine<br />

and Siteman Cancer Center, St. Louis, MO<br />

Background: This phase II clinical trial evaluated the efficacy and safety <strong>of</strong><br />

the combination <strong>of</strong> bevacizumab, a VEGF inhibitor, and pemetrexed, a<br />

multi-targeted antifolate agent inhibiting thymidylate synthase, for recurrent<br />

or persistent epithelial ovarian, fallopian tube (FT) or primary peritoneal<br />

(PP) cancer. Methods: Patients with measurable recurrent/persistent<br />

epithelial ovarian, FT or PP cancer after at least one prior platinum- and<br />

taxane-containing regimen were eligible. Patients might have received � 2<br />

prior cytotoxic chemotherapy regimens but no prior bevacizumab. Pemetrexed<br />

500 mg/m2 IV and bevacizumab 15 mg/kg IV were given every 3<br />

weeks until progression, unacceptable adverse effects, or patient/physician<br />

choice. 32 patients were needed to have 90% power to detect the primary<br />

endpoint <strong>of</strong> 6-month PFS � 40% with a two-sided 0.05 significance.<br />

Secondary endpoints included toxicity and response by RECIST and<br />

CA-125 criteria. Results: Thirty-four patients received a median <strong>of</strong> 7<br />

treatment cycles (range, 2-26). Twenty-eight patients (82%, 95% CI:<br />

66-92) were platinum-sensitive. Median follow-up was 17.1 months<br />

(range, 2.7-31.2). The 6-month PFS was 58.2% (95%CI: 40-73) for all<br />

patients and 50% (19-81) for platinum-resistant patients. Objective<br />

response rates by RECIST criteria included (%; 95%CI): 0 CR, 14 PR<br />

(41%; 25-59), 18 SD (53%; 35-70) and 2 PD (6%; 1-20). Of 27 patients<br />

evaluable by CA-125, levels declined �50% in 17 (62%; 44-79), and<br />

�75% in 8 (30%; 16-49). 12-month OS was 88% (95%CI: 71-95) and<br />

median PFS was 7.8 months (95%CI: 4.7-10.7). Of the 34 patients, grade<br />

3-4 hematologic toxicities occurred in 53% (neutropenia 50%, leukopenia<br />

26%, thrombocytopenia 12%, anemia 9%). Other grade 3-4 toxicities<br />

included metabolic (29%), constitutional (18%), pain (18%) and gastrointestinal<br />

(15%). No bowel perforations occurred. Conclusions: Combined<br />

bevacizumab/pemetrexed is well tolerated and highly active for the treatment<br />

<strong>of</strong> recurrent ovarian cancer. The dose and schedule tested here<br />

warrant further investigation in phase III trials.<br />

5015 Poster Discussion Session (Board #4), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Randomized phase II study <strong>of</strong> docetaxel plus vandetanib (D�V) versus<br />

docetaxel followed by vandetanib (D-V) in patients with persistent or<br />

recurrent epithelial ovarian, fallopian tube, or primary peritoneal carcinoma<br />

(OC): SWOG S0904. Presenting Author: Robert L. Coleman, University <strong>of</strong><br />

Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: Antiangiogenesis therapy has led to antitumor effects both<br />

preclinically and clinically. Vandetanib (V) is an oral tyrosine kinase<br />

inhibitor <strong>of</strong> VEGFR-2/3, EGFR and RET. These targets are <strong>of</strong> interest in OC<br />

care, as targeting has shown anti-tumor efficacy, particularly in combination<br />

with taxanes. We explored the efficacy, safety, and toxicity <strong>of</strong> docetaxel<br />

(D) and V in women with recurrent OC. Methods: Women with resistant,<br />

refractory, or progressive/persistent OC were eligible for this randomized<br />

phase II study provided they had not received D or V for recurrent disease.<br />

Patients were allowed to receive other anti-VEGF targeted agents for<br />

primary therapy (stratification variable). Up to 3 additional cytotoxic<br />

regimens for recurrence were allowed. Patients were allocated 1:1 to D(75<br />

mg/m2 , I.V.)�V (100 mg daily, p.o.) or D(75 mg/m2 ). Patients receiving<br />

single agent D were allowed to crossover to V upon progression (D-V). The<br />

primary endpoint was PFS. Other objectives were: OS, objective response<br />

(ORR), and frequency/severity <strong>of</strong> adverse events. The study was designed<br />

with 84% power to detect a 1.55 PFS hazard ratio using a one-sided P <strong>of</strong><br />

0.1. Results: 131 patients were enrolled; 5 were excluded (1 ineligible, 4<br />

eligible but untreated). 9% had received prior anti-angiogenic therapy. 61<br />

patients on D�V were assessable for toxicity; 19 (31%) had treatmentrelated<br />

G4 events, primarily hematologic. Similarly, 17 (26%) <strong>of</strong> 65<br />

patients receiving D alone had G4 events, primarily hematologic. 34 (52%)<br />

patients crossed over to V; no G4 events were recorded among 32 evaluable<br />

patients. G3 diarrhea was observed in 14% <strong>of</strong> D�V patients; 5% D-V<br />

patients. G3 acneiform rash occurred in 2% and 0%, respectively. The<br />

median PFS estimates were 3.0 mos (D�V) vs 3.5 (D-V); HR (PFS): 0.98<br />

(80% CI:0.75-1.27). For OS, the median estimates were 14 mos (D�V) vs<br />

12 mos (D-V); HR (OS):0.84 (80% CI:0.56-1.28). ORR was 14% and<br />

17%, respectively. Crossover V response was 4% (1/27 measurable<br />

patients). Conclusions: D�V was well tolerated in this population however,<br />

did not prolong PFS with respect to D.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


5016 Poster Discussion Session (Board #5), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Phase II trial <strong>of</strong> irinotecan plus bevacizumab for heavily pretreated<br />

recurrent ovarian cancer. Presenting Author: Salvia Sanjay Jain, New York<br />

University School <strong>of</strong> Medicine, New York, NY<br />

Background: Irinotecan and bevacizumab have single agent activity in<br />

platinum sensitive (PSen) and resistant (PRes) ovarian cancer patients<br />

(pts). We sought to evaluate the efficacy and toxicity <strong>of</strong> irinotecan plus<br />

bevacizumab in these pts. The trial was designed to evaluate whether the<br />

progression free survival (PFS) at 6 months is at least 40% and with<br />

secondary objectives <strong>of</strong> estimating response rate (RR), duration <strong>of</strong> response<br />

(DoR), time to progression and toxicity. Methods: No limitation on number<br />

<strong>of</strong> prior regimens was placed, and prior use <strong>of</strong> study drugs was allowed.<br />

Irinotecan 250mg/m2 and bevacizumab 15mg/kg every 3 weeks were<br />

given. Due to treatment-related grade 3 toxicity (diarrhea and neutropenia)<br />

experienced by the first 5 pts on the study, the dose <strong>of</strong> irinotecan was<br />

amended to 175mg/m2. Results: 20 pts with recurrent ovarian cancer<br />

[PSen 5, PRes 15] <strong>of</strong> a planned 35 have been recruited thus far. Median<br />

age is 59 (45-78). Median number <strong>of</strong> prior regimens is 5 (3-12) with 9 pts<br />

demonstrating progressive disease (PD) on prior topotecan-containing<br />

regimens and 7 pts exhibiting PD on prior bevacizumab-containing regimens.<br />

4 pts discontinued treatment before 2 cycles (2 for protocol defined<br />

toxicity, 2 by patient/physician choice). <strong>Part</strong>ial response (PR) was observed<br />

in 2 PSen pts and 1 PRes pt, while stable disease (SD)was seen in 9 (2<br />

PSen, 7 PRes) out <strong>of</strong> the 15 pts assessable for response at this time. 3 pts<br />

demonstrated PD after 2 cycles <strong>of</strong> treatment. 12 <strong>of</strong> 13 pts with PR or SD by<br />

RECIST also had response by CA125 criteria. Median DoR thus far (SD plus<br />

PR) is 18 weeks (4-37). 6 pts have ongoing response (4-18 weeks). Of 19<br />

pts that received � 2 cycles, 3 had grade 3 diarrhea (2 before protocol<br />

amendment and 1 after). 2 pts had grade 3/4 neutropenia (1 before and 1<br />

after protocol amendment). Median PFS is 9.6 months (mts). Median<br />

overall survival is 15.5 mts. PFS rate at 6 mts is 61% with 95% confidence<br />

interval: (40%, 92%). Conclusions: Results <strong>of</strong> the trial to date suggest the<br />

hypothesis that the PFS at 6 mts is less than 40% can be rejected. Activity<br />

<strong>of</strong> this regimen is encouraging given the heavily pretreated nature <strong>of</strong> the<br />

pts. Dose-limiting diarrhea and neutropenia required protocol amendment.<br />

We continue to accrue study subjects at the amended dosing.<br />

5018 Poster Discussion Session (Board #7), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Lenalidomide (REV) in asymptomatic late recurrent ovarian cancer (ROC)<br />

patients with increasing CA 125: A GINECO phase II trial. Presenting<br />

Author: Frédéric Selle, Universite Pierre et Marie Curie, Oncology, GHU-Est<br />

Tenon, Paris, France<br />

Background: REV is a thalidomide analogue, with both immunomodulatory<br />

and anti-angiogenic properties that could confer antitumor effect in ROC.<br />

Methods: The aim <strong>of</strong> this study was to evaluate REV efficacy as single agent<br />

in patients (pts) with asymptomatic late ROC (�6mos) with increasing CA<br />

125, in 2nd or 3rd line. Primary endpoint was to estimate the rate <strong>of</strong><br />

non-progressive disease at 4 mos. Pts were treated with REV 20 mg daily in<br />

oral continuous regimen with systematically recommended anti-thrombotic<br />

prophylaxis (ATP). Imagery and CA 125 were performed every 8 weeks.<br />

Results: From 05/2009 to 09/2010, 45 pts were included with a median<br />

age <strong>of</strong> 63 years. Pt characteristics were: serous (78%), previous lines (one<br />

73%, two 27%), median platinum-free interval (PFI) (11.3 mos), PFI � 12<br />

mos (42%), measurable disease (73%), and ECOG performance status 0<br />

(84%). Efficacy: Rate <strong>of</strong> non progressive disease at 4 mos was 38%<br />

(95%CI, 23-53), 59 % (95%CI, 36-82) and 24 % (95%CI, 7-41) for the<br />

global population, pts relapsing over 12 mos and those relapsing between<br />

6-12 mos, respectively. Results were independent <strong>of</strong> the number <strong>of</strong><br />

previous lines. Median progression-free survival was 3.8 mos (95%CI,<br />

2.1-5.6) and 6.4 mos in the subset <strong>of</strong> pts with PFI � 12 mos. Response<br />

evaluation according to CA 125 (Rustin criteria) was: complete response<br />

(CR) 2.4%, partial response (PR) 17%, stable disease (SD) 71%. When<br />

using RECIST criteria alone, response evaluation was: 9.5% PR and 45%<br />

SD. Median duration <strong>of</strong> biological response was 6.6 mos. REV efficacy will<br />

be correlated to immunological parameters (lymphocyte phenotypes and<br />

cytokines). Safety: Grade 3-4 toxicity in more than 5% <strong>of</strong> pts was<br />

neutropenia (29%) and thrombo-embolic events (TEE) (11%). TEE occurred<br />

only in pts without ATP. Reasons for stopping treatment due to<br />

toxicity were TEE (3), allergy (2), arrhythmia (1), dyspnea (1) and<br />

neutropenia (1). Conclusions: REV demonstrated encouraging activity in<br />

ROC with good tolerability and manageable adverse events. A phase I <strong>of</strong><br />

REV combined with platinum-based chemotherapy is currently being<br />

conducted.<br />

Gynecologic Cancer<br />

331s<br />

5017^ Poster Discussion Session (Board #6), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Safety <strong>of</strong> front-line bevacizumab (BEV) combined with weekly paclitaxel<br />

(wPAC) and q3w carboplatin (C) for ovarian cancer (OC): Results from<br />

OCTAVIA. Presenting Author: Antonio Gonzalez-Martin, GEICO and Medical<br />

Oncology Service, Centro Oncológico M. D. Anderson International<br />

Spain, Madrid, Spain<br />

Background: In two randomized phase III trials in OC (GOG218 and ICON7),<br />

front-line BEV � q3w PAC � q3w C followed by BEV alone significantly<br />

improved progression-free survival (PFS) vs chemotherapy (CT) alone. In<br />

the Japanese NOVEL trial, wPAC � q3w C was more effective than q3w<br />

PAC � C, but toxicity limited CT delivery. The single-arm OCTAVIA study<br />

evaluated front-line BEV � wPAC � q3w C. Methods: Patients (pts)<br />

received 6–8 cycles <strong>of</strong> BEV (7.5 mg/kg, d1) � wPAC (80 mg/m2 d1, 8, 15)<br />

� C (AUC6, d1) iv q3w, with BEV q3w continued alone for a total <strong>of</strong> up to<br />

17 cycles (1 y) as front-line therapy for newly diagnosed OC (FIGO stage<br />

I–IIa [grade 3/clear cell] or stage IIb–IV [any grade]). The trial was designed<br />

to recruit a pt population similar to that enrolled in ICON7. The primary<br />

endpoint was PFS. Secondary endpoints included response rate, duration<br />

<strong>of</strong> response, overall survival, biological progression-free interval, and<br />

safety. Previously we reported safety findings from the concurrent CT<br />

phase. Here we present final safety results from the entire treatment period.<br />

Results: Between Jun 2009 and Jun 2010, 189 eligible pts were enrolled.<br />

Baseline characteristics: median age 55 y (range 24–79 y); ECOG 0 74%;<br />

FIGO stage I/II/III/IV 10%/10%/63%/17%; serous/clear cell/mixed 65%/<br />

6%/6%; 71% optimally debulked. Pts received a median <strong>of</strong> 6 CT cycles<br />

(range 1–8) and 17 BEV cycles (range 0–18). Of the 168 pts who received<br />

single-agent BEV, 135 completed 1y<strong>of</strong>therapy. In the entire treatment<br />

period, BEV was discontinued for adverse events (AEs) in 12% and disease<br />

progression (PD) in 10%. The most common grade �3 hematologic AEs<br />

were neutropenia (60%), anemia (8%), and thrombocytopenia (7%). The<br />

incidences <strong>of</strong> grade �3 AEs <strong>of</strong> special interest for BEV were: hypertension<br />

4.2% (grade 2/3/4: 9.0%/3.2%/1.1%); thromboembolic events 6.3%<br />

(grade 3/4: 3.7%/2.6%); bleeding 0.5% (grade 3), wound-healing complications<br />

0.5% (grade 3), and GI perforation 0.5% (grade 4). There was no<br />

grade �3 proteinuria or fistula/abscess. At the time <strong>of</strong> data cut-<strong>of</strong>f, 9 pts<br />

had died, all from PD. Conclusions: BEV combined with wPAC is feasible<br />

and well tolerated. BEV AEs were no more frequent with wPAC in OCTAVIA<br />

than with q3w PAC in ICON7.<br />

5019 Poster Discussion Session (Board #8), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

A phase II study <strong>of</strong> RO4929097 (RO), a gamma-secretase inhibitor, in<br />

advanced platinum (Pt)-resistant (R) ovarian cancer (OC): A study <strong>of</strong> the<br />

PMH, Chicago, and California phase II consortia. Presenting Author: Ivan<br />

Diaz-Padilla, Princess Margaret Hospital, Toronto, ON, Canada<br />

Background: Notch signalling pathway plays a critical role in regulating<br />

cellular differentiation, proliferation, and apoptosis in preclinical cancer<br />

models. The Notch receptor, its ligands, and down-stream effectors are<br />

over-expressed in OC. Inhibition <strong>of</strong> �-secretase-mediated Notch cleavage is<br />

a primary focus for the development <strong>of</strong> targeted therapeutics. Methods:<br />

Women (pts) with progressive, measurable (RECIST), Pt-R OC treated with<br />

� 2 chemotherapy regimens for recurrent disease were treated with oral RO<br />

at 20mg od, 3 days on/4 days <strong>of</strong>f every week, q3w. The primary objective<br />

was to determine the antitumor efficacy <strong>of</strong> RO in Pt-R OC by the<br />

progression-free survival (PFS) rate at the end <strong>of</strong> 4 cycles. Secondary<br />

objectives were to assess the safety <strong>of</strong> RO and to explore molecular<br />

correlates <strong>of</strong> outcome in archival tumor tissue. A Simon two-stage design<br />

was used. The study would open to second stage accrual if � 4 pts <strong>of</strong> the<br />

first 17 accrued remain progression-free at the end <strong>of</strong> 4 cycles. Results: 39<br />

pts have been enrolled after first-stage criteria were met. Median age was<br />

59 (range 26-81). Median number <strong>of</strong> cycles was 2 (range 1-18). 30 (83%)<br />

pts had high-grade serous OC. 34 pts were evaluable for response. 8 pts<br />

(20%) were progression-free after 4 cycles.12 pts (35%) had stable<br />

disease, with a median duration <strong>of</strong> 3.9 months (range 2.5-12.2). Median<br />

PFS was 1.3 months (1.2-2.3). The most common drug-related adverse<br />

events (AEs) <strong>of</strong> any grade (% pts) were: nausea (36), fatigue (28), anorexia<br />

(15), hypophosphatemia (15), anemia (13), and increased alanine aminotransferase<br />

[ALT] (13). There were 5 G3-4 AEs at least possibly related with<br />

RO: increased liver transaminases (2), diarrhea (1), headache (1), and<br />

hypophosphatemia (1). Intracellular Notch (NIC) and JAG1 protein expression<br />

on high-grade serous OC were correlated with response in 17 pts.<br />

Median PFS for pts with high NIC (n�6) was 3.3 months (1.0-not reached),<br />

compared to 1.3 months (1.1-2.6) for pts with low NIC (n�11), p�0.09.<br />

Conclusions: RO has limited clinical activity in unselected Pt-R OC as a<br />

single agent. Future studies need to assess potential for cohort enrichment<br />

using NIC expression.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


332s Gynecologic Cancer<br />

5020 Poster Discussion Session (Board #9), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Randomized trial <strong>of</strong> oral cyclophosphamide (C) with or without veliparib<br />

(V), an oral poly (ADP-ribose) polymerase (PARP) inhibitor, in patients with<br />

recurrent BRCA-positive ovarian, or primary peritoneal or high-grade serous<br />

ovarian carcinoma. Presenting Author: Shivaani Kummar, Developmental<br />

Therapeutics Clinic, National Cancer Institute, Bethesda, MD<br />

Background: V�C was well tolerated in a phase I trial and responses and<br />

prolonged disease stabilization were observed in BRCA � patients (pts).To<br />

assess the relative contribution <strong>of</strong> the PARP inhibitor to the efficacy<br />

observed for the combination, we conducted a randomized multicenter trial<br />

comparing the response rate (RR) <strong>of</strong> V and C to the RR <strong>of</strong> C alone in patients<br />

with deleterious BRCA mutations and recurrent ovarian, or primary peritoneal,<br />

fallopian tube or high-grade serous ovarian cancer. Methods: Pts were<br />

� 18 yrs, KPS � 70%, had adequate organ function, prior therapy with<br />

PARP inhibitors was allowed.Both drugs were administered orally qd; C 50<br />

mg, V 60 mg; 21 day cycles. Pts were randomized to receive either C alone<br />

or V�C. At disease progression, pts on C alone were allowed to cross over to<br />

the combination. Radiologic imaging was performed at baseline and q 3<br />

cycles for assessment <strong>of</strong> response. Dose reduction <strong>of</strong> V was allowed to 40<br />

mg for gr 3 non-hematologic or gr 4 hematologic toxicities. The study<br />

design had an 88% power to detect the difference between a 15% RR for C<br />

alone versus 35% for V�C, early closure if fewer responses were observed<br />

on the combination arm in the first 65 pts enrolled (half <strong>of</strong> the total<br />

projected accrual). Results: Total <strong>of</strong> 74 pts were enrolled (36 pts C, 38 pts<br />

V�C), median age 58 (37-79 yrs), # <strong>of</strong> prior therapies: median 4 (1-9), 2<br />

pts had prior PARP therapy. Treatment was well tolerated, Gr � 2 toxicities<br />

per arm for initial regimen (# <strong>of</strong> pts): C alone: lymphopenia (2), mucositis<br />

(1); V�C: lymphopenia (4), anemia (2), leucopenia (2), neutropenia (2). Of<br />

the 74 pts evaluable for response at the interim analysis, 3 PRs observed in<br />

36 pts on V�C and 5 PRs <strong>of</strong> 38 pts on C alone arm; thus accrual was<br />

stopped. PAR levels assessed by validated ELISA were inhibited (�80%) in<br />

PBMCs in 9/10 pts 4 hours post V, no inhibition with C alone. Conclusions:<br />

Addition <strong>of</strong> V, a PARP inhibitor, to C did not improve RR versus C alone.<br />

Exomic sequencing, gene expression studies, and Fanconi Anemia triple<br />

stain immun<strong>of</strong>luorescence (FATSI) assay for FancD2 nuclear foci formation<br />

using archival tissue are ongoing.<br />

5022 Poster Discussion Session (Board #11), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Use <strong>of</strong> chemotherapy (CT) in BRCA1/2-deficient ovarian cancer (BDOC)<br />

patients (pts) with poly-ADP-ribose polymerase inhibitor (PARPi) resistance:<br />

A multi-institutional study. Presenting Author: Joo Ern Ang, The<br />

Royal Marsden NHS Foundation Trust and The Institute <strong>of</strong> Cancer Research,<br />

Sutton, United Kingdom<br />

Background: Emerging clinical data point to the clinical utility <strong>of</strong> PARPi in<br />

BDOC. However, the impact <strong>of</strong> PARPi exposure on the prospects <strong>of</strong><br />

response to further CT remains unclear. In our previous single centre study,<br />

we provided the first clinical data relating to the use <strong>of</strong> post-PARPi CT (JCO<br />

2010: 28(15S), A5041). Our aim here was to re-examine this issue in a<br />

larger multi-institutional population. Methods: We included pts with advanced<br />

BDOC who received CT, having progressed on �200 mg bd<br />

olaparib. Pt, tumor and treatment characteristics and clinical outcomes<br />

were documented. Relationships between post-PARPi CT overall survival<br />

(OS) and other variables were explored using Cox regression. Results: We<br />

collected data on 75 pts (median age 51 y [range 31-77], BRCA1:BRCA2<br />

54:21, mean 3 previous lines <strong>of</strong> CT [95% CI 2.5-3.4], pre-PARPi platinum<br />

(Plt) resistance rate 49% and olaparib RECIST-response rate [RR] 39%<br />

[95% CI 28-50]). Following olaparib, most pts received Plt alone or in<br />

combination with taxane (Tx) or liposomal doxorubicin (PLD). Weekly Plt<br />

was used in 36% <strong>of</strong> all Plt-treated pts, mainly in combination with weekly<br />

Tx. Overall RECIST and CA125 (GCIG) RRs were 38% and 48%, respectively;<br />

these responses occurred independently <strong>of</strong> PARPi response or<br />

pre-PARPi Plt sensitivity (all p�.1). The median progression free survival<br />

and OS <strong>of</strong> RECIST responders were 7.9 m (95% CI 5.8-10.9) and 10.5 m<br />

(95% CI 1.4-19.6), respectively. In all pts, the median OS from the start <strong>of</strong><br />

post-PARPi CT was 7.9 m (95% CI 5.7-10.1) while that from diagnosis was<br />

64.9 m (95% CI 52.9-76.9). Factors associated with improved OS on<br />

post-PARPi CT in the MVA included best olaparib response <strong>of</strong> non-disease<br />

progression (p�.003, HR 0.28), optimal initial debulking (p�.01, HR<br />

0.36) and pre-PARPi Plt sensitivity (p�.05, HR 0.46). Conclusions: These<br />

data indicate potential for meaningful responses to CT in BDOC pts with<br />

PARPi resistance. Analysis to identify molecular predictors <strong>of</strong> response is<br />

ongoing.<br />

RR ([n responses]/[n evaluable])<br />

Regimen<br />

RECIST GCIG<br />

Plt single agent/combination 16/37 20/35<br />

Tx single agent 4/9 3/7<br />

PLD single agent 1/8 4/11<br />

Others 1/4 1/5<br />

Total 22/58 (38%) 28/58 (48%)<br />

5021 Poster Discussion Session (Board #10), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Phase I/II study <strong>of</strong> weekly paclitaxel with or without MLN8237 (alisertib),<br />

an investigational aurora A kinase inhibitor, in patients with recurrent<br />

epithelial ovarian, fallopian tube, or primary peritoneal cancer (OC), or<br />

breast cancer (BrC): Phase I results. Presenting Author: Gerald Steven<br />

Falchook, Department <strong>of</strong> Investigational Cancer Therapeutics, University <strong>of</strong><br />

Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: Preclinical data indicate that inhibiting aurora A kinase (AAK),<br />

a key mitotic regulator, in OC can enhance the activity <strong>of</strong> taxane-based<br />

chemotherapy. This is the first clinical study to evaluate the AAK inhibitor<br />

MLN8237 (A) in combination with paclitaxel (P). Methods: Eligible women<br />

(aged �18 y) had previously treated, metastatic or locally recurrent OC or<br />

BrC. OC pts had previously received a platinum and taxane, and had<br />

disease recurrence within 12 mo <strong>of</strong> discontinuing platinum. Pts received IV<br />

P (D1, 8, 15) with oral A (enteric coated tablet, 3 d on, 4 d <strong>of</strong>f, wkly for 3<br />

wks) in 28-d cycles. Doses were escalated in a 3�3 schema. Phase I<br />

endpoints were safety, response by RECIST and CA125, and pharmacokinetics<br />

(PK). Results: 28 pts were treated at doses <strong>of</strong> P80 mg/m2 � A10 mg<br />

BID (n�5), P80�A20 (n�6), P60�A20 (n�4), P60�A30 (n�6),<br />

P60�A40 (n�4) and P60�A50 (n�3). 25 pts had OC (20 ovarian, 4<br />

primary peritoneal, 1 mullerian) and 3 BrC. Median age was 59 y (range<br />

37–72). Pts received a median <strong>of</strong> 2.5 (1–16) and 2.5 cycles (1–15) <strong>of</strong> A<br />

and P, respectively. 11 pts are ongoing. 3 pts had DLTs: Gr 4 diarrhea<br />

(P80�A20), Gr 3 stomatitis (P80�A20) and Gr 4 neutropenia for �7 d<br />

with fever (P60�A30). MTD has not yet been reached. 27 pts had �1<br />

drug-related adverse event (AE); most common were neutropenia (n�18),<br />

anemia (n�13) and nausea (n�12). 21 pts had Gr �3 drug-related AEs;<br />

the most common was neutropenia (n�15). 5 pts had drug-related SAEs.<br />

No pts have died on study. Preliminary PK data (n�24) indicated fast<br />

absorption <strong>of</strong> A with a median Tmax <strong>of</strong> 3 hr. Geometric means <strong>of</strong> A AUC0–12hr on D3 were 3740 nM*hr (CV: 22%), 7180 nM*hr (31%), 11,900 nM*hr<br />

(49%) and 15,100 nM*hr (72%) at 10, 20, 30 and 40 mg BID,<br />

respectively. A exposures increased approximately dose proportionally.<br />

<strong>Part</strong>ial responses by RECIST (n�5) and/or CA125 (n�7) in 8 pts (29%; 7<br />

OC, 1 BrC); 3 pts had stable disease �6 mo. Conclusions: Emerging data<br />

from this first combination study suggest that P�A is generally well<br />

tolerated and has antitumor activity in pts with OC/BrC. Dose-escalation is<br />

ongoing to determine the recommended phase II dose.<br />

5023 Poster Discussion Session (Board #12), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

A novel autologous oxidized whole-tumor antigen vaccine therapy for<br />

recurrent ovarian cancer. Presenting Author: Lana Kandalaft, University <strong>of</strong><br />

Pennsylvania, Philadephia, PA<br />

Background: Despite surgical and chemotherapeutic advances, the death<br />

rate from ovarian cancer has not changed. We report here an enhanced<br />

dendritic cell vaccine platform developed for clinical testing. Furthermore<br />

we report the application <strong>of</strong> this novel platform comprising <strong>of</strong> dendritic cell<br />

(DC)-based autologous whole tumor antigen vaccination in a pilot study <strong>of</strong><br />

patients with recurrent ovarian cancer. Methods: To determine the optimal<br />

tumor lysate preparation for loading DCs, ovarian tumor lines were prepared<br />

by HOCl-oxidation, UVB-irradiation or freeze and thaw. Normal donor DCs<br />

were evaluated for tumor lysate uptake, cytokine and chemokine productions<br />

and phenotype. The optimal lysate preparation, was used in a phase I<br />

study where five patients with recurrent ovarian cancer with available tumor<br />

lysate from secondary debulking surgery underwent intranodal vaccination<br />

with OC-DC, an autologous DC preparation pulsed with HOCL oxidized<br />

autologous tumor cells. Feasibility, safety, and biological and clinical<br />

efficacy were evaluated. Results: Normal donor DCs pulsed with HOCloxidized<br />

tumor lines demonstrated the highest tumor lysate uptake,<br />

matured efficiently after LPS and IFN-gamma stimulation, and produced<br />

higher levels <strong>of</strong> proinflammatory cytokines and chemokines. In vitro, these<br />

lysates loaded DCs primed T cell responses against ovarian tumor associated<br />

antigens and effectively expanded against tumor specific T cells from<br />

donors and patients. Therapy was feasible and well tolerated in all subjects.<br />

Vaccination with OC-DC produced limited grade 1 toxicities and elicited<br />

tumor-specific T cell responses. Moreover specific HLA-A2-restricted<br />

responses were documented following vaccination and HER-2/neu specific<br />

T cells were expanded following 10 days <strong>of</strong> in vitro culture. Patients<br />

exhibiting immune response demonstrated clinical benefit including two<br />

patients who demonstrated remission inversion on vaccine maintenance.<br />

Conclusions: We developed a DC-HOCl oxidized whole tumor lysate vaccine<br />

which was safe and well-tolerated by patients. The vaccine was highly<br />

proinflammatory and elicited cellular and humoral anti-tumor responses<br />

establishing a platform for immune-combinatorial therapy.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


5025 Poster Discussion Session (Board #14), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Phase II trial <strong>of</strong> temsirolimus and bevacizumab for initial recurrence <strong>of</strong><br />

endometrial cancer. Presenting Author: Mark H. Einstein, Albert Einstein<br />

College <strong>of</strong> Medicine, Bronx, NY<br />

Background: We report the interim results <strong>of</strong> the endometrial arm <strong>of</strong> a<br />

multi-tumor protocol using temsirolimus and bevacizumab in endometrial<br />

cancer (EMCA) patients at the time <strong>of</strong> their initial recurrence. The primary<br />

aim <strong>of</strong> this trial is to assess treatment efficacy in terms <strong>of</strong> both confirmed<br />

tumor response and 6-month progression free survival (PFS). Methods:<br />

Women with a performance status <strong>of</strong> 0 or 1 who have had their first<br />

recurrence for EMCA were eligible. Subjects who had chemotherapy as part<br />

<strong>of</strong> their adjuvant treatment after front line surgical staging were also<br />

eligible. The regimen included Temsirolimus 25 mg IV weekly followed by<br />

bevacizumab 10mg/kg IV on days 1 and 15 <strong>of</strong> a 28 day cycle. A modified<br />

two-stage Simon design with fixed sample size was adopted with the null<br />

hypothesis being that the true tumor response rate is at most 25% and the<br />

true 6 month PFS rate is at most 50%. Results: We enrolled 26 evaluable<br />

subjects to the first stage <strong>of</strong> which one did not proceed with treatment. The<br />

median age at enrollment was 60 (range 40-80). 22 (85%) were white and<br />

19 (73%) were not Hispanic/Latino. 19 (73%) <strong>of</strong> subjects had prior<br />

raditation therapy, with 4 having a prior para-aortic boost. 5 (20%) subjects<br />

had a confirmed PR and 12 (48%) were progression-free at 6 months,<br />

which fell short <strong>of</strong> the futility stopping rule. An additional 5 (20%) subjects<br />

had a best response <strong>of</strong> confirmed SD, so 10 (40%) had overall clinical<br />

benefit from this regimen. AEs attributable to treatment were modest and<br />

included 16 grade 3 adverse events, <strong>of</strong> which the most common ones<br />

included hypertension, hyperglycemia, and neutropenia. There were 2<br />

grade 4 events that were possibly treatment related including a duodenal<br />

perforation and an anorectal infection. Conclusions: While there was<br />

clinical benefit <strong>of</strong> this regimen in women at the time <strong>of</strong> their first recurrence<br />

<strong>of</strong> EMCA, the combination <strong>of</strong> temsirolimus and bevacizumab did not<br />

achieve our prespecified efficacy assumptions. This differs from what has<br />

been reported with this combination as a second line therapy for recurrent<br />

EMCA, where prespecified response assumptions differ. Also, the regimen<br />

had comparable safety and toxicity to other cytotoxic chemotherapy<br />

regimens used in this setting.<br />

5028 Poster Discussion Session (Board #17), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Efficacy and safety <strong>of</strong> the APE (actinomycin D, cisplatin, etoposide)<br />

regimen for the management <strong>of</strong> high-risk gestational trophoblastic neoplasia.<br />

Presenting Author: Catherine Lhomme, Institut Gustave Roussy,<br />

Villejuif, France<br />

Background: Patients (pts) with high risk gestational trophoblastic neoplasia<br />

(GTN) or who fail low risk single agent chemotherapy (CT) require multi<br />

agent CT to be cured. The most common regimen is etoposide (E),<br />

methotrexate and actinomycin D (A) alternating weekly with cyclophophamide<br />

and vincristine (EMA/CO). Cisplatin (P) is a very active drug but its<br />

role is controversial and usually restricted to second line. We report results<br />

<strong>of</strong> a platinum based therapy: APE. Methods: We evaluated the efficacy and<br />

safety on 103 pts treated at Institut Gustave Roussy (IGR) (n�80) or other<br />

French centers (n�23) between 1983 and 2010 with APE for high risk<br />

GTN (defined by IGR criteria [Azab, Cancer, 1988] and/or FIGO score �6).<br />

Pts with brain metastasis were excluded. Results: Efficacy was evaluated on<br />

59 pts treated for high risk GTN in first line, and on 39 pts in �2nd line<br />

including 13 pts after multi agent CT. We excluded pts with placental site<br />

trophoblastic tumors (n�2), or with FIGO score �7 and without IGR criteria<br />

(n�3). Complete remission (CR) rate was 95%. Seven pts (7 %) relapsed<br />

and a second CR was obtained for all with surgery and/or CT. Only one<br />

patient died due to GTN, after successive CRs obtained with 3 regimens.<br />

Five year overall survival (median follow-up 6.6 years) was 98%. Toxicity<br />

was evaluated on 95 pts. No toxic death occurred. Given good efficacy and<br />

to avoid acute hematotoxicity and long-term G�1 neuro and ototoxicity<br />

APE regimen was modified as detailed in the Table (below). Long-term<br />

neuro (5 pts, G1), oto (2 pts, G1 and 2 pts, G2) and renal toxicities (1 pt,<br />

G1 ) were recorded. No long-term G2 toxicities were observed with APE3.<br />

One pt developed an AML 4 after 4cy APE and 6 cy EMA/CO. 37 pts <strong>of</strong> 40<br />

who wished to be pregnant succeeded and all <strong>of</strong> them had at least one live<br />

birth. Conclusions: With a 98% long-term overall survival rate, an excellent<br />

reproductive outcome, and no detectable long-term toxicity, APE-3 should<br />

be regarded as an alternative standard option to EMA/CO for high-risk GTN.<br />

A 0.3 mg/m2 /J E 100 mg/m2 /J Cisplatin Cycles N<br />

APE 1 J1–3,J14-15 J1–3,J14-15 100 mg/m²/J, J1 J1 � J28 38<br />

APE 2 J1–3 J1–3 100mg/m²/J, J1 J1 � J21 34<br />

APE 3 J1–3 J1–3 75mg/m²/J, J1 J1 � J21 23<br />

Gynecologic Cancer<br />

333s<br />

5026 Poster Discussion Session (Board #15), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Brief family history questionnaire for identification <strong>of</strong> Lynch syndrome in<br />

women with newly diagnosed endometrial cancer. Presenting Author: Sarah<br />

E. Ferguson, Princess Margaret Hospital, Toronto, ON, Canada<br />

Background: Endometrial cancer (EC) is <strong>of</strong>ten the sentinel cancer for<br />

women with Lynch syndrome (LS); however, it is underappreciated in this<br />

population. The Brief Family History Questionnaire (bFHQ) was developed<br />

to identify women with EC who have family histories suggestive <strong>of</strong> LS. The<br />

objective <strong>of</strong> our study was to evaluate the bFHQ compared to an extended<br />

family history (eFHQ) and medical record in identifying women with EC who<br />

may benefit from genetic cancer risk assessment. Methods: All women with<br />

newly diagnosed EC from July 2010 to June 2011 were asked to participate<br />

in a prospective screening protocol for LS which included completing two<br />

family history questionnaires; the bFHQ which is a 4-item self-report<br />

measure and the 37-item eFHQ administered by a research assistant.<br />

Family history was also extracted from the medical record. Using the bFHQ<br />

women were flagged as requiring additional investigation for LS based on<br />

predetermined criteria and the predictive ability <strong>of</strong> the flag was evaluated<br />

treating eFHQ as the gold standard. Comparisons were made between the<br />

bFHQ, eFHQ and medical record for families meeting Amsterdam II,<br />

<strong>Society</strong> Gynecologic Oncologist (SGO) 20-25% or the Ontario Ministry <strong>of</strong><br />

Health (MOH) testing criteria for LS, using generalized estimating equation<br />

logistic regression models. Results: 119 (N � 182, 65%) consented to the<br />

study and 106 (89%) completed the bFHQ. The median age was 61<br />

(26-91). The number <strong>of</strong> women who met testing criteria by the eFHQ was<br />

17 (16%) and 33 (31%) were flagged by the bFHQ. The sensitivity,<br />

specificity, PPV and NPV <strong>of</strong> the bFHQ was 88.2%, 79.8%, 45.5% and<br />

97.3%. There was no significant difference in the number <strong>of</strong> women who<br />

met Amsterdam II or SGO 20-25% testing criteria between the bFHQ,<br />

eFHQ and medical record (P � 0.05). The numbers <strong>of</strong> women meeting<br />

MOH criteria using the bFHQ (N�16, 15%) and the eFHQ were similar<br />

(N�17, 16%) (P � 0.7); however, more women met MOH criteria using the<br />

bFHQ and the eFHQ compared to the medical record (N�8, 7.6%) (P �<br />

0.011; P � 0.006). Conclusions: The patient-administered bFHQ is a<br />

highly effective tool in identifying women who meet MOH testing criteria for<br />

LS and is a good screening tool to identify women with EC for further<br />

genetic assessment.<br />

5029 Poster Discussion Session (Board #18), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

A snapshot <strong>of</strong> potentially personalized care: Molecular diagnostics in<br />

gynecologic cancer. Presenting Author: Elizabeth Sales, Massachusetts<br />

General Hospital, Boston, MA<br />

Background: Genetic abnormalities underlie the development <strong>of</strong> cancer. It<br />

has been proposed that tumors be recategorized by gene mutation such as<br />

BRAF in LG serous, TP53 in HG serous, and PIK3CA in clear cell and<br />

endometrioid tumors. These targets potentially represent an opportunity for<br />

personalizing cancer therapy. Methods: Gynecologic Oncology patients at<br />

the MGH Cancer Center can have their tumor genotyped for a panel <strong>of</strong><br />

mutations by SNaPshot, a validated, CLIA approved assay developed by<br />

MGH that uses DNA from FFPE tissue to interrogate 160 site-specific<br />

mutations across 15 genes (AKT1, APC, BRAF, CTNNB1, EGFR, ERBB2,<br />

IDH1, KIT, KRAS, MAP2K1, NOTCH1, NRAS, PIK3CA, PTEN, TP53). At<br />

present SNaPshot has no validated endpoints in GYN Cancers but may help<br />

identify a useful clinical trial. Results: Between 5/17/10 and 10/17/11,<br />

125 patients consented to SNaPshot genotyping. Patients had a median<br />

age <strong>of</strong> 59 (24-78) yrs. Tumors were ovarian 70(56%), uterine clear, UPSC,<br />

or MMMT 16(13%), uterine endometrioid 10(8%), fallopian tube 8(6%),<br />

PPC 7(6%), cervical 6(5%), uterine sarcomas (3), ACUP (2), vulvovaginal<br />

(2), metastatic (1). A mutation was identified in 41(33%), with 9 <strong>of</strong> these<br />

(23%) having 2 or 3 (n�2) mutations. In the 85 ovarian, FT, and PPC<br />

cancers 33% were �ve, but 50% were in TP53. The low mutation rate for<br />

TP53 is likely explained by copy number abnormalities (Amplification).<br />

50% <strong>of</strong> the 10 uterine tumors were �ve, with 3 <strong>of</strong> those 5 having multiple<br />

mutations in the PIK3CA pathway, while 69% <strong>of</strong> the non-endometrioid<br />

uterine tumors had mutations. Only 20% <strong>of</strong> the vulvo-vaginal and Cx<br />

tumors had mutations, both PIK3CA. 19% <strong>of</strong> the purely serous tumors<br />

(n�58) had TP53 mutations, and 37% <strong>of</strong> the purely clear/endometrioid<br />

tumors (n�19) had mutations in PIK3CA, PTEN or AKT. Certain rare<br />

tumors did not have identifiable mutations: granulosa cell tumors (2),<br />

ovarian small cell (2). 5 pts with a PIK3CA mutation were enrolled on a<br />

clinical trial (2 phase II, 3 phase I, 3 uterine, 1 ovary, 1 cervix).<br />

Conclusions: SNaPshot can identify potentially important therapeutic<br />

targets. However, the incidence <strong>of</strong> �drugable� targets in ovarian cancer is<br />

low, and �5% subjects eventually were treated on a relevant clinical trial.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


334s Gynecologic Cancer<br />

5030 Poster Discussion Session (Board #19), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

A comprehensive analysis <strong>of</strong> BRAF and KRAS mutation status in low-grade<br />

serous (LGS) and serous borderline (SB) ovarian cancer (OC). Presenting<br />

Author: Rachel Nicole Grisham, Memorial Sloan-Kettering Cancer Center,<br />

New York, NY<br />

Background: LGS OC develops in a step-wise pattern from serous cystadenoma<br />

to SB tumor to invasive LGSOC. LGSOC accounts for 6-22% <strong>of</strong> serous<br />

OC and is a chemoresistant disease. Prior studies have reported that<br />

LGS/SB ovarian tumors harbor BRAF mutations in 28-35% <strong>of</strong> cases,<br />

suggesting that LGS/SB OC may respond to mutant BRAF inhibitors.<br />

Methods: We genotyped 75 LGS and SB ovarian tumors for BRAF and KRAS<br />

hotspot mutations using a mass spectrometry based Sequenom assay. All<br />

samples were collected at MSKCC between 2000-2010. All specimens<br />

underwent two independent pathologic reviews for diagnostic confirmation<br />

and macrodissection to ensure 80% cellularity. The incidence and identity<br />

<strong>of</strong> BRAF and KRAS mutations were defined and results were correlated with<br />

stage, response to treatment, and outcome. Exon capture sequencing is<br />

underway to sequence all coding exons <strong>of</strong> 230 cancer associated genes to<br />

identify additional targetable alterations in LGS/SB OC. Results: Of 75<br />

samples examined, 56(75%) were SB and 19(25%) LGS histology. 57% <strong>of</strong><br />

samples showed KRAS mutation (KRAS�)(n�17, 23%) or BRAF mutation<br />

(BRAF�)(n�26, 35%). Mutation status was mutually exclusive. All BRAF<br />

� were V600E, KRAS� were G12D (n�9) or G12V (n�8). BRAF� was<br />

associated with early stage (I-II) at presentation and SB histology. 22<br />

(29%) patients were treated with chemotherapy, 2 (9%) with KRAS � and<br />

0 with BRAF�. 4(15%) BRAF� patients underwent resection <strong>of</strong> recurrent<br />

disease. All BRAF� patients are alive without evidence <strong>of</strong> disease at a<br />

median follow-up <strong>of</strong> 43.3 months (range 1.9- 129.3 months). Conclusions:<br />

V600E BRAF mutations are present in 35% <strong>of</strong> cases <strong>of</strong> SB/LGS ovarian<br />

cancer. Presence <strong>of</strong> BRAF � in SB/LGS OC is associated with good<br />

prognosis and surgical cure <strong>of</strong> disease. Patients with SB/LGS OC who<br />

require systemic therapy are unlikely to have BRAF�. Our group is<br />

performing a detailed search for additional targetable alterations in<br />

multiple signal transduction pathways through next generation sequencing<br />

technology.<br />

KRAS� BRAF� WT for KRAS/BRAF p value<br />

Stage<br />

I/II 10 (58.8%) 24 (92.3%) 11 (34.4%) �0.001<br />

III/IV 7 (41.2%) 2 (7.7%) 21 (65.6%)<br />

Histology<br />

SB 14 (82.4%) 25 (96.2%) 17 (53.1%) �0.001<br />

LGS 3 (17.6%) 1 (3.8%) 15 (46.9%)<br />

5032 Poster Discussion Session (Board #21), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Application <strong>of</strong> micro-RNA (miRNA) expression pr<strong>of</strong>iles for prognostication<br />

in low-risk endometrial carcinoma (LEC). Presenting Author: Johanne<br />

Ingrid Weberpals, The Ottawa Hospital, Ottawa, ON, Canada<br />

Background: Reliably predicting which LEC patients are most likely to recur<br />

is a challenge for the clinician with implications on adjuvant therapy.<br />

MiRNAs have been exploited for diagnosis and prognostication in a number<br />

<strong>of</strong> malignancies. We hypothesize that miRNA expression pr<strong>of</strong>iles differ in<br />

tumors from patients with recurrence compared to those without recurrence.<br />

Methods: The inclusion criteria for this study are informed consent,<br />

stage 1 disease, grade 1 or 2 tumors and endometrioid histology. RNA was<br />

extracted from formalin-fixed paraffin-embedded tissues and miRNA pr<strong>of</strong>iling<br />

was done using Agilent Human miRNA. Differentially expressed<br />

miRNAs were identified using GeneSpring GX s<strong>of</strong>tware and the two groups<br />

were compared using the student t-test. Results: The expression levels <strong>of</strong><br />

866 miRNAs were determined from LEC patients with recurrence (n�15)<br />

and without recurrence (n�16). The mean follow-up interval was 61.5<br />

months. The average age <strong>of</strong> cancer diagnosis for patients with and without<br />

recurrence was 60.2 (range 42-75) and 59.7 (range 44-86), respectively<br />

(p�0.91). Three <strong>of</strong> 15 patients with recurrence and 6 <strong>of</strong> 16 patients<br />

without recurrence received adjuvant brachytherapy following their primary<br />

surgery (p�0.43). 17 miRNAs were identified which can distinguish<br />

between the tumors with recurrence and those without recurrence (p�0.05).<br />

MiR-146a, miR-18a, miR-222, and miR-30a showed the highest fold<br />

change difference (�5 fold) in the tumors with recurrence compared to that<br />

did not recur. A decision tree prediction model for recurrent LEC was<br />

developed where a miRNA cut<strong>of</strong>f was used as a branch in the decision tree.<br />

This model identified those patients who were most likely to recur based on<br />

the expression <strong>of</strong> 4 dysregulated miRNAs (miR-222, miR-361-3p, miR-<br />

181c and miR-125b). Conclusions: These preliminary results show the<br />

miRNA expression pr<strong>of</strong>ile differs among LEC and can be used to distinguish<br />

an aggressive sub-group. Should future validation studies confirm this<br />

result, this information would be valuable in the design <strong>of</strong> a biomarker<br />

study to help decide which patients would benefit most from extended<br />

adjuvant treatment.<br />

5031 Poster Discussion Session (Board #20), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Topotecan plus carboplatin versus standard therapy with paclitaxel plus<br />

carboplatin (PC) or gemcitabin plus carboplatin (GC) or carboplatin plus<br />

pegylated doxorubicin (PLDC): A randomized phase III trial <strong>of</strong> the NOGGO-<br />

AGO-Germany-AGO Austria and GEICO-GCIG intergroup study (HECTOR).<br />

Presenting Author: Jalid Sehouli, Department <strong>of</strong> Gynecology, European<br />

Competence Center for Ovarian Cancer, Charité Campus Virchow Klinikum,<br />

Berlin, Germany<br />

Background: We present the efficacy data from a phase III study <strong>of</strong><br />

topotecan (T) plus carboplatin (C) versus standard therapy with paclitaxel<br />

plus carboplatin (PC) or gemcitabine plus carboplatin (GC) or carboplatin<br />

plus pegylated doxorubicin (PLDC). Methods: From 02/07 to 12/09, 590<br />

pts were screened and 550 pts were randomized to either T (0.75mg/m²/d1-<br />

3/q21d) � C (AUC 5/d1/q21d) or to standard therapy with CP or GC or<br />

PLDC based on patient preference. Progression free survival at 1 year was<br />

defined as primary endpoint. Results: Median number <strong>of</strong> cycles was 6<br />

(range 0-9) in both arms. Most patients preferred GC (78%) in the standard<br />

therapy arm.. Best Response (CR�PR) was 73.1% (95%CI) and 75.1%<br />

(95%CI) for the CA. Median follow-up was 18 (0-52) months for TC and 20<br />

(0-48) months for standard therapy. TC failed to show any advantage<br />

regarding 1-yr.-PFS or OAS. Conclusions: The combination <strong>of</strong> topotecan<br />

plus carboplatin failed to improve PFS or OAS in platinum sensitive<br />

relapsed ovarian cancer. In addition, carboplatin plus gemcitabine was well<br />

tolerated with lower rates <strong>of</strong> severe and long-lasting (neuropathy) toxicities<br />

compared to paclitaxel-carboplatin.<br />

1 year PFS rate (%): p�0.215<br />

PFS, median months (95%); p�0.472<br />

OAS, median moths (95%),<br />

p�0.340<br />

TC<br />

arm<br />

37.0%<br />

10 mo<br />

(9.2-10.8)<br />

25 mo<br />

(22.4-27.6)<br />

CP<br />

arm<br />

49.3%<br />

12 mo<br />

(9.1-14.9)<br />

29 mo<br />

(23.4-34.6)<br />

GC<br />

arm<br />

36.8%<br />

10 mo<br />

(9.1-10.9)<br />

33 mo<br />

(26.8-39.7)<br />

Neutropenia G3–4; p�0.015 27.8% 34.2% 40.7%<br />

Thrombocytopenia G3–4; p2; p2; p�0.073 10.0% 17.7% 8.5%<br />

Hand-foot syndrome G>2 0% 0% 0%<br />

Mucositis G>2p�0.380 3.0% 6.3% 3.7%<br />

Hypersensitivity, allergic reaction G>2;<br />

p�0.100<br />

15.2% 12.7% 8.5%<br />

Early treatment discontinuation<br />

(toxicity related, < 6 cycles); p�0.162<br />

15.9% 7.6% 13.2%<br />

5033 Poster Discussion Session (Board #22), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Randomized, phase III study <strong>of</strong> carboplatin plus paclitaxel for 8 cycles<br />

(CP8) versus carboplatin x 8 cycles plus paclitaxel x 4 cycles (C8P4) in<br />

advanced ovarian, fallopian, or primary peritoneal carcinoma. Presenting<br />

Author: Aristotelis Bamias, HECOG and University <strong>of</strong> Athens, Athens,<br />

Greece<br />

Background: The combination <strong>of</strong> carboplatin/paclitaxel represents the<br />

standard 1st-line chemotherapy in advanced OC, FC, and PPC. The optimal<br />

duration <strong>of</strong> paclitaxel treatment has not been defined, while its use is<br />

associated with cumulative neurotoxicity in about 50% <strong>of</strong> patients, which<br />

becomes long-term in 15-20% <strong>of</strong> cases. We, therefore, designed a<br />

randomized study to investigate the effect <strong>of</strong> administering 4 instead <strong>of</strong> 8<br />

cycles <strong>of</strong> paclitaxel in the combination carboplatin/paclitaxel on efficacy<br />

and tolerability <strong>of</strong> this treatment. Methods: Patients with FIGO stages IIC-IV<br />

OC, FC, PPC were included. Carboplatin AUC 6 and paclitaxel at 175<br />

mg/m2 were used. Both agents were administered for 8 cycles in the CP8<br />

arm, while paclitaxel was administered only for 4 cycles in the C8P4. The<br />

study was powered to detect a � 15% difference in survival rate to a<br />

baseline rate <strong>of</strong> 50 % at the 3-year time point. Results: 389 pts were<br />

randomized (2/2004-1/2008) and 380 were eligible for analysis (CP8:<br />

192, C8P4:188). The distribution (CP8 vs C8P4) <strong>of</strong> baseline characteristics<br />

were: stage III: 78% vs. 76%; IV: 12% vs. 15%, residual disease 0 cm:<br />

25% vs. 22%, ECOG PS 0: 68% vs. 64%, serous carcinomas: 79% vs.<br />

68%, tumor grade III: 56% vs. 63%. During a median follow up <strong>of</strong> 72.3<br />

months 231 patients (111 [58%] in CP8 arm and 120 [64%] in the C8P4<br />

arm) have died. Median PFS was significantly shorter in the C8P4 arm<br />

(21.41 vs. 16.46 months, HR [95% CI]: 1.36 [1.07-1.71], Wald’s<br />

p�0.011), while OS was similar between the two arms (53.41 vs. 46.59<br />

months, HR [95% CI]: 1.18 [0.91-1.53], Wald’s p�0.211). Lower grade 3<br />

or 4 neurotoxicity (1.9% vs. 10.8%, p� 0.001) but higher myelotoxicity<br />

(neutropenia 38.8% vs. 28.8%, p�0.031; thrombocytopenia 20% vs.<br />

8.3%, p�0.004) was observed in the C8P4 arm. Conclusions: Lowering the<br />

total number <strong>of</strong> cycles <strong>of</strong> paclitaxel in 1st-line chemotherapy <strong>of</strong> advanced<br />

OC, FC, PPC resulted in similar OS but shorter median PFS and is not<br />

recommended in this setting. The reduction <strong>of</strong> neurotoxicity by limiting the<br />

total paclitaxel cycles to 4 is achieved at the expense <strong>of</strong> higher myelotoxicity.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


5034 Poster Discussion Session (Board #23), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Use <strong>of</strong> HE4 and CA125 to predict surgical outcome and for prognostic<br />

value for progression-free survival (PFS) and overall survival (OS) in primary<br />

epithelial ovarian cancer (EOC) patients (pts). Presenting Author: Ioana<br />

Braicu, Charité University Department <strong>of</strong> Gynecology, Campus Virchow<br />

Clinic , Berlin, Germany<br />

Background: Optimal surgical cytoreduction and response to platinum (P)<br />

based chemotherapy (ChTh) remain the cornerstones <strong>of</strong> therapeutic management<br />

<strong>of</strong> primary EOC. Aim <strong>of</strong> this study was to analyze the predictive<br />

role <strong>of</strong> HE4 and CA125 as biomarkers (BM) for clinical outcome in primary<br />

EOC pts at diagnosis and during subsequent follow-up. Methods: In the<br />

European OVCAD project 275 pts with primary EOC were enrolled. Pts were<br />

eligible if radical cytoreductive surgery and P-based ChTh were applied.<br />

Plasma collected at first diagnosis and 6 months after 1st line ChTh in<br />

P-sensitive pts was analyzed for HE4 and CA125 levels using ELISA and<br />

Luminex technique, respectively. Results: Complete cytoreduction with no<br />

residual tumor disease (RTD) was obtained in 69.9% pts. HE4 and CA125<br />

expression in plasma at first diagnosis correlated with RTD, p � 0.002 and<br />

p�0.002, respectively. The sensitivity (SE) and specificity (SP) <strong>of</strong> the<br />

combinative use <strong>of</strong> both BM in predicting RTD was 64.8% and 73.5%,<br />

respectively. Pts having over-expression <strong>of</strong> both BM in plasma had a 6.1<br />

greater risk for RTD (p�0.001, OR: 6.107, 95% CI 2.41-15.46). Presistance<br />

occurred more frequently when both BM were over-expressed<br />

(p�0.028, OR� 3.1, 95%CI 1.13-8.46). Elevated BM levels during<br />

follow-up predicted recurrence (SE 90% and SP 71% for CA125 �55U/<br />

ml; SE 72.7% and SP 81.4% for HE4 �150pM) and when HE4 or CA125<br />

were positive, a SE <strong>of</strong> 86.4% and SP <strong>of</strong> 72.9% were achieved. Elevated<br />

CA125 and HE4 at 6 months following adjuvant therapy was associated<br />

with significantly poorer PFS (p�0.001, HR 9.6, 95%CI 3.93-23.44 with<br />

elevated HE4 or CA125, and HR�50.52, 95%CI 14.44-176.78, with<br />

elevated HE4 and CA125) and OS (p�0.001, HR�7.42 95%CI 1.43-<br />

38.42 with elevated HE4 or CA125 and HR�28.38 95%CI 6.50-123.97<br />

with elevated HE4 and CA125). Conclusions: The combinative use <strong>of</strong> HE4<br />

and CA125 appears to have a significant value in predicting optimal<br />

surgical outcome and development <strong>of</strong> P resistance disease in EOC pts.<br />

Elevated plasma levels 6 months after 1st line ChTh significantly correlate<br />

with OS and PFS in P-sensitive pts.<br />

5036 Poster Discussion Session (Board #25), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Use <strong>of</strong> an optimized primary ovarian cancer xenograft model to mimic<br />

patient tumor biology and heterogeneity. Presenting Author: Zachary C.<br />

Dobbin, University <strong>of</strong> Alabama at Birmingham, Birmingham, AL<br />

Background: Current xenograft and transgenic models <strong>of</strong> ovarian cancer are<br />

mainly homogeneous and poorly predict response to therapy. Use <strong>of</strong> patient<br />

tumors may represent a better model for tumor biology and <strong>of</strong>fer potential to<br />

test personalized medicine approaches, but poor take rates and questions<br />

<strong>of</strong> recapitulation <strong>of</strong> patient tumors have limited this approach. We have<br />

developed a protocol for improved feasibility <strong>of</strong> such a model and examined<br />

its similarity to the patient tumor. Methods: Under IRB and IACUC approval,<br />

23 metastatic ovarian cancer samples were collected at the time <strong>of</strong> tumor<br />

reductive surgery. Samples were implanted either subcutaneously (SQ),<br />

intraperitoneally (IP), in the mammary fat pad (MFP), or in the subrenal<br />

capsule (SRC) and monitored for tumor growth. Cohorts from 8 xenolines<br />

were treated with combined carboplatin and paclitaxel or vehicle, and<br />

response to therapy compared between xenografts and patients. Expression<br />

<strong>of</strong> tumor-initiating cell (TIC) markers ALDH1, CD133, and CD44 was<br />

assessed by immunohistochemistry in tumors from patients and treated<br />

and untreated xenografts. Results: At least one SQ implanted tumor<br />

developed in 91.3% <strong>of</strong> xenografts, significantly higher than in the MFP<br />

(63.6%), IP (23.5%), or SRC (8%). Xenografts were similar in expression<br />

<strong>of</strong> putative TIC’s compared to patient tumors. The patients and the<br />

xenografts also have similar responses to chemotherapy in that xenografts<br />

from patients with a partial response responded more slowly than those<br />

from patients achieving a complete response (45 vs 21 days, p�.004).<br />

Treated xenografts were more densely composed <strong>of</strong> TICs. ALDH1 increased<br />

to 36.1% from 16.2% (p�0.002) and CD133 increased to 33.8% from<br />

16.2% (p�0.026). Conclusions: Xenoline development can be achieved at<br />

a high rate when tumors collected from metastatic sites are implanted SQ.<br />

These xenografts are similar to patient tumors with regard to chemotherapy<br />

response and TIC expression.. This model may be a more accurate model<br />

for in vivo pre-clinical studies as compared to current models. Also, as<br />

treated xenografts become chemoresistant, this model is well positioned to<br />

evaluate targeted therapies aimed at the most aggressive populations in a<br />

heterogeneous tumor.<br />

Gynecologic Cancer<br />

335s<br />

5035 Poster Discussion Session (Board #24), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Functional pr<strong>of</strong>iling <strong>of</strong> clear cell ovarian cancer. Presenting Author: Rowan<br />

Miller, The Institute <strong>of</strong> Cancer Research, London, United Kingdom<br />

Background: Clear cell ovarian cancer represents up to 15% <strong>of</strong> epithelial<br />

ovarian cancers. In comparison to other subtypes, clear cell ovarian<br />

carcinomas have a poorer prognosis and are relatively resistant to standard<br />

platinum based chemotherapy. Recently, loss <strong>of</strong> function mutations in the<br />

tumour suppressor gene ARID1A were identified in up to 50% <strong>of</strong> ovarian<br />

clear cell carcinomas. We have adopted an integral functional and<br />

molecular pr<strong>of</strong>iling approach as a route to identify new genetic dependencies<br />

and therapeutic targets for this disease. Methods: Clear cell ovarian<br />

cancer cell lines were functionally pr<strong>of</strong>iled using high throughput screening<br />

with chemical and siRNA libraries. This has been integrated with molecular<br />

pr<strong>of</strong>iling data generated from exome and transcriptome sequencing to aid<br />

the discovery <strong>of</strong> novel targets. Results: Using functional screens we have<br />

now identified critical gene dependencies and potential therapeutics in a<br />

series <strong>of</strong> clear cell ovarian cancer models. The comparison <strong>of</strong> functional<br />

viability pr<strong>of</strong>iles for models characterized by ARID1A loss <strong>of</strong> function<br />

mutations is now enabling an analysis <strong>of</strong> synthetic lethal effects that could<br />

be used to target clear cell ovarian cancers carrying these mutations.<br />

Conclusions: The work undertaken so far provides the framework for the<br />

discovery <strong>of</strong> therapeutic targets for clear cell ovarian cancer using an<br />

integrated approach. Revalidation <strong>of</strong> these preliminary results is now<br />

underway to characterize new genetic dependencies for this disease.<br />

5037 General Poster Session (Board #17E), Sun, 8:00 AM-12:00 PM<br />

Effect <strong>of</strong> weekly administration <strong>of</strong> bevacizumab, gemcitabine, and oxaliplatin<br />

in patients with heavily pretreated ovarian cancer. Presenting Author:<br />

Yuji Ikeda, Ohki Memorial Cancer Center for Women, Tokorozawa, Japan<br />

Background: The combination therapy <strong>of</strong> gemcitabine with oxaliplatin<br />

(GEMOX) has high activity in patients with ovarian cancer. Bevacizumab<br />

(B), a vascular endothelial growth factor specific antibody, enhances<br />

chemotherapeutic efficacy through its anti-angiogenic function in various<br />

types <strong>of</strong> tumors. We evaluated the effect <strong>of</strong> weekly administration <strong>of</strong> B and<br />

GEMOX in heavily-pretreated patients with recurrent or refractory ovarian<br />

cancers (ROC). Methods: Nineteen patients with ROC received at least three<br />

or more cycles <strong>of</strong> weekly-B and GEMOX consisting <strong>of</strong> B (2mg/kg),<br />

gemcitabine (300mg/m2 ) and oxaliplatin (30mg/m2 ). The treatment was<br />

continued until the development <strong>of</strong> progressive disease. The response and<br />

adverse effects (AE) were evaluated using the response evaluation criteria<br />

in solid tumors (RECIST), CA125 Gynecologic Cancer Intergroup (GCIG)<br />

criteria, and common terminology criteria for adverse events (CTCAE)<br />

version 3.0. Results: Seventeen (89%) <strong>of</strong> the 19 patients were primarily<br />

stage 3 or 4. Fifteen patients (79%) had received more than three regimens<br />

<strong>of</strong> chemotherapy. All patients were pretreated with a platinum-containing<br />

regimen within 6 months and 16 <strong>of</strong> these patients (84%) were pretreated<br />

within 3 months. According to the RECIST evaluation, 2 patients (11%)<br />

had a complete response (CR), 6 patients (32%) had a partial remission<br />

(PR) and 5 patients (26%) had a stable disease (SD). The response rate<br />

(RR; CR�PR) and clinical benefit rate (CBR; CR�PR�SD) were 42% and<br />

68%, respectively. In 10 patients with serous adenocarcinoma, RR was<br />

60%. In 6 patients pretreated with weekly B and pegylated liposomal<br />

doxorubicin (PLD), RR was 50%. Median progression-free survival was 5<br />

months (range: 2-11 months). Hematological adverse effects (AE) with<br />

grade 3/4 were leukopenia (16%), neutropenia (10%), thrombocytopenia<br />

(5%), however, all AE were manageable. Conclusions: Weekly B and<br />

GEMOX administration had significant activity with mild AE in patients with<br />

ROC especially in serous adenocarcinoma. Notably, the activity was also<br />

observed in patients pretreated with weekly B and PLD. These results<br />

warrant further prospective study.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


336s Gynecologic Cancer<br />

5038 General Poster Session (Board #17F), Sun, 8:00 AM-12:00 PM<br />

Phase Ib study <strong>of</strong> AMG 386 in combination with paclitaxel (P) and carboplatin<br />

(C) in high-risk stage I and stages II-IV epithelial ovarian, primary peritoneal, or<br />

fallopian tube cancers. Presenting Author: Antonio Casado, Hospital Clínico San<br />

Carlos, Madrid, Spain<br />

Background: AMG 386 is a first-in-class investigational peptibody that inhibits<br />

angiopoietin-1/-2 binding to Tie2. We report on an ongoing, 2-part, open-label<br />

phase 1b study <strong>of</strong> AMG 386 � PC as first-line treatment <strong>of</strong> high-risk ovarian<br />

cancer. Methods: <strong>Part</strong> 1 enrolled women, GOG performance status 0/1, with<br />

newly diagnosed, FIGO high-risk stage I or stages II, IIIA-B (primary debulking<br />

surgery; PDS) or IIIC & IV (PDS or interval debulking surgery; IDS) ovarian<br />

cancer. Patients (pts) received AMG 386 at 15 mg/kg QW IV � P at 175 mg/m2 Q3W � C at AUC 6 Q3W for 6 cycles (or until disease progression or<br />

unacceptable toxicity); AMG 386 was withheld 4 weeks pre-/post-debulking. Pts<br />

completing 6 cycles continued AMG 386 QW as maintenance for up to 18<br />

months. <strong>Part</strong> 1: incidence <strong>of</strong> dose-limiting toxicities (DLTs) dictated expansion<br />

to 25 pts (part 2). Primary endpoint: DLT incidence; secondary: adverse events<br />

(AEs), PK and efficacy measures. Results: At the time <strong>of</strong> analysis, 16 pts had<br />

received �1 dose <strong>of</strong> AMG 386 � PC and completed �2 cycles <strong>of</strong> therapy (PDS,<br />

n�8; IDS, n�8). No DLTs occurred in part 1. 15 pts continue on-study; 6 pts<br />

have received a median <strong>of</strong> 6 doses <strong>of</strong> maintenance AMG 386. One pt<br />

discontinued due to progression. AEs are tabulated below. One serious AE<br />

(decreased appetite; maintenance phase) required hospitalization. AE incidence<br />

and type were similar for PDS or IDS. Combination therapy did not affect the PK<br />

<strong>of</strong> AMG 386 or PC. Non-neutralizing antibodies against AMG 386 were detected<br />

in 2 pts. Conclusions: AMG 386 at 15 mg/kg QW � PC is tolerable in pts with<br />

high-risk ovarian cancer. AMG 386 maintenance was tolerated and associated<br />

with few AEs. Updated toxicity and efficacy data will be presented.<br />

AEs<br />

Any; n (%)<br />

Of specific interest (grade 3<br />

Neutropenia<br />

Decreased appetite<br />

Localized edema<br />

Syncope<br />

Thrombocytopenia<br />

Anemia<br />

Ascites<br />

Hypertension<br />

Hypokalaemia<br />

Psoriasis<br />

All grade >4<br />

Neutropenia<br />

Thrombocytopenia<br />

Grade 5<br />

AMG 386 � PC<br />

n � 16<br />

16 (100)<br />

1(6)<br />

1(6)<br />

1(6)<br />

8 (50)<br />

5 (31)<br />

0<br />

0<br />

2 (13)<br />

2 (13)<br />

1(6)<br />

1(6)<br />

1(6)<br />

1(6)<br />

1(6)<br />

4 (25)<br />

3 (19)<br />

1(6)<br />

0<br />

AMG 386 maintenance<br />

n � 6<br />

4 (67)<br />

0<br />

0<br />

0<br />

2 (33)<br />

0<br />

1 (17)<br />

1 (17)<br />

0<br />

0<br />

0<br />

0<br />

0<br />

0<br />

0<br />

0<br />

0<br />

0<br />

0<br />

5040 General Poster Session (Board #17H), Sun, 8:00 AM-12:00 PM<br />

ESA use in women with cancer: Consequences <strong>of</strong> the 2008 FDA clinical<br />

alert. Presenting Author: Kimberly M. Dickinson, Warren Alpert Medical<br />

School <strong>of</strong> Brown University, Providence, RI<br />

Background: Erythropoietin stimulating agents (ESA) are used clinically as<br />

an alternative to blood transfusions in cancer patients suffering from<br />

symptoms <strong>of</strong> anemia. However, more recent randomized controlled trials <strong>of</strong><br />

ESA usage concluded that its use is associated with an increased risk <strong>of</strong><br />

tumor progression and death. As a result, in July 2008 the FDA issued a<br />

clinical alert restricting the use <strong>of</strong> ESA. A reduction in the prescribing <strong>of</strong><br />

ESA was immediately seen but changes in blood transfusion rates have not<br />

been examined. Methods: A retrospective chart review was conducted<br />

drawing from patients under treatment in the Program in Women’s<br />

Oncology at Women and Infant’s Hospital from one year before the clinical<br />

alert (August 2007-July 2008) to one year afterward (August 2008-July<br />

2009). The primary outcomes were blood transfusion and ESA administration<br />

rates compared across the two time periods. Results: The study<br />

population (n�776) included patients with a cancer diagnosis who<br />

received chemotherapy during one or both time periods. 165 (21.3%)<br />

patients received ESA treatment. The total number <strong>of</strong> ESA treatments<br />

administered in the study period <strong>of</strong> interest was 1,277, with the majority<br />

(60%) given prior to the FDA alert. The mean number <strong>of</strong> ESA treatments in<br />

the first time period was 6.39 per person as compared to 0.61 per person in<br />

the second time period. Of the study population, 186 (23.8%) patients<br />

received at least one blood transfusion. A total <strong>of</strong> 463 blood transfusions<br />

were administered during the entire study period but a significant difference<br />

was not observed in the proportion <strong>of</strong> those delivered prior to the FDA<br />

alert (52%) versus after the FDA alert (48%). The average number <strong>of</strong><br />

transfusions given in the first time period was 2.34 per person, as<br />

compared to 2.17 per person in the second period. Conclusions: Our results<br />

indicate that despite a steep decline in the use <strong>of</strong> ESA for chemotherapyinduced<br />

anemia, blood transfusion rates were not significantly different<br />

between the two periods. Interestingly, a slight downward trend was<br />

observed from before the FDA alert to after the alert. While more work is<br />

needed to understand the implications <strong>of</strong> these findings, it suggests that<br />

resource utilization did not increase despite the reduction in ESA use.<br />

5039 General Poster Session (Board #17G), Sun, 8:00 AM-12:00 PM<br />

The combination <strong>of</strong> intravenous bevacizumab and metronomic oral cyclophosphamide<br />

for recurrent platinum-resistant ovarian cancer. Presenting<br />

Author: Emma L. Barber, Northwestern University, Department <strong>of</strong> Gynecologic<br />

Oncology, Chicago, IL<br />

Background: This study was undertaken to determine the progression free<br />

survival and overall survival in heavily pre-treated patients with recurrent<br />

ovarian carcinoma treated with bevacizumab and metronomic oral cyclophosphamide.<br />

Methods: An IRB-approved retrospective review was performed<br />

for all patients with recurrent ovarian, fallopian tube or primary<br />

peritoneal carcinomas treated with intravenous bevacizumab 10mg/kg<br />

every 14 days and oral cyclophosphamide 50mg daily between January<br />

2006 and December 2010. Response to treatment was determined by<br />

change in disease status according to RECIST criteria and/or CA-125<br />

levels. Results: Sixty-six eligible patients were identified with a median age<br />

<strong>of</strong> 58 years. Fifty-five patients (83%) originally had optimal cytoreduction<br />

and all were platinum resistant. Median time from diagnosis to beginning<br />

bevacizumab and cyclophosphamide was 36 months. Median number <strong>of</strong><br />

prior chemotherapy treatments was 7.5 (range 3-16). Eight patients<br />

(12.1%) had side effects which required discontinuing bevacizumab and<br />

cyclophosphamide, most common were hypertension, proteinuria, and<br />

fatigue. There was one bowel perforation (1.5%). A complete response was<br />

noted in 7 patients (10.6%), partial response was seen in 21 patients<br />

(31.8%) with an overall response rate <strong>of</strong> 42.4%. Fifteen patients (22.7%)<br />

had stable disease and 23 (34.8%) had disease progression. Median<br />

progression free survival (PFS) for responders was 5 months (range 2-14)<br />

and 11 months (range 4-14) for those with a complete response. Median<br />

overall survival (OS) from start <strong>of</strong> bevacizumab and cyclophosphamide for<br />

responders was 20 months (range 2-56) and 9 months (range 1-51) for<br />

nonresponders. Conclusions: Bevacizumab and cyclophosphamide is an<br />

effective, well-tolerated chemotherapy regimen in heavily pre-treated<br />

patients with recurrent ovarian carcinoma which significantly improves PFS<br />

and OS in responders. Response rates were significantly better than the<br />

rates we have reported in this same group <strong>of</strong> patients receiving topotecan<br />

(22%) or liposomal doxorubicin (25%) and were superior to reported rates<br />

for single agent bevacizumab (18%) in patients with only 2-3 prior<br />

regimens.<br />

5041 General Poster Session (Board #18A), Sun, 8:00 AM-12:00 PM<br />

Carboplatin dosing in the treatment <strong>of</strong> epithelial ovarian cancer (EOC): A<br />

Gynecologic Oncology Group (GOG) study. Presenting Author: Roisin Eilish<br />

O’Cearbhaill, Memorial Sloan-Kettering Cancer Center, New York, NY<br />

Background: Optimal carboplatin dosing (CPRx) for patients (pts) with renal<br />

dysfunction or low creatinine (Cr) values in the setting <strong>of</strong> malnutrition and<br />

ascites is unknown. Multiple methods have been utilized to estimate Cr<br />

clearance (CrCl) but these perform differently in pts with abnormal Cr<br />

values. We sought to determine 1) the relationship between adverse events<br />

(AE) and baseline CrCl used for CPRx; 2) the effect on CPRx <strong>of</strong> using<br />

Cockcr<strong>of</strong>t-Gault (CG) �/- the NCI/CTEP recommended limits (CGL), Modification<br />

<strong>of</strong> diet in renal disease (MDRD) or Jelliffe Formula (J) renal function<br />

estimates. Methods: Retrospective data were drawn from pts treated on<br />

GOG 182, a phase III trial <strong>of</strong> carboplatin doublet vs triplet or sequential<br />

doublet combinations in stage III/IV EOC. For patient safety, the protocol<br />

was amended to assign the lower limit <strong>of</strong> Cr at 0.6mg/dl for CPRx. Area<br />

under the receiver operating characteristic curve (AUC) was used to<br />

describe associations between CrClJ and various AE. Sensitivity and<br />

positive predictive values (PPV) described the AE rate in pts with CrClJ �60ml/min. CPRx for each pt was calculated using J, CG, CGL and MDRD.<br />

Results: 3830 evaluable pts had a mean age 58.7yrs, mean BMI 26.8kg/m2 and mean baseline CrClJ 81.9ml/min (range 23.4-239). The AUC statistics<br />

(range 0.52-0.64) show that the log(CrClJ) was not a good predictor <strong>of</strong><br />

grade �3 AE (anemia, thrombocytopenia, febrile neutropenia, auditory,<br />

renal, metabolic, neurologic). A cut<strong>of</strong>f value <strong>of</strong> CrClJ �60 ml/min would<br />

have deemed 15% <strong>of</strong> pts treated on GOG182 ineligible. The range <strong>of</strong> PPV<br />

for the above AEs in pts with CrClJ �60 ml/min was 1.8-15%. Using CG,<br />

CGL, MDRD instead <strong>of</strong> J for CPRx would have resulted in �10% decrease<br />

in CPRx in 21%, 32% and 12% <strong>of</strong> pts, respectively. Using CG, CGL, MDRD<br />

instead <strong>of</strong> J for CPRx would have resulted in �10% increase in CPRx in<br />

45%, 9.6% and 5.2% <strong>of</strong> pts, respectively. Conclusions: Our data do not<br />

support excluding patients with CrClJ �60ml/min from clinical trials. The<br />

new GOG guidelines replacing J with CGL affect CPRx. The clinical<br />

significance <strong>of</strong> this change with regards to toxicity, particularly in pts with<br />

abnormally low Cr values, is yet to be determined.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


5042 General Poster Session (Board #18B), Sun, 8:00 AM-12:00 PM<br />

Detection <strong>of</strong> disseminated tumor cells in bone marrow as an independent<br />

prognostic factor in primary ovarian cancer patients. Presenting Author:<br />

Pauline Wimberger, AGO and Department <strong>of</strong> Gynecology and Obstetrics,<br />

University <strong>of</strong> Duisburg-Essen, Essen, Germany<br />

Background: Detection <strong>of</strong> disseminated tumor cells (DTC) in the bone<br />

marrow (BM)<strong>of</strong> breast cancer patients is associated with poor outcome.<br />

Recent studies demonstratedthat DTC may serve as a prognostic factor in<br />

ovarian cancer.The aim <strong>of</strong> our study was to evaluate the impact <strong>of</strong> BM<br />

status on survival in a large cohort <strong>of</strong> ovarian cancer patients. Methods: 365<br />

patients with primary ovarian cancer were included into this three-center<br />

prospective study. BM aspirates were collected preoperatively from iliac<br />

crest. Disseminatedtumor cells were identified by immunocytochemistry<br />

using the pancytokeratin antibodyA45B/B3 and by cytomorphology. Patient<br />

outcomes were evaluated using a multivariable Cox regression model.<br />

Results: Disseminated tumor cells were detected in 28% <strong>of</strong> all BM<br />

aspirates. The number <strong>of</strong> CK-positive cells ranged from 1 to 42 per 2x106 mononuclear cells. DTC status did not correlate with any <strong>of</strong> the established<br />

clinicopathogical factors. The overall survival was significantly shorter<br />

among DTC-positive patients compared to DTC-negative patients (51 mo,<br />

95% CI: 35 – 67 mo versus 32 mo, 95% CI: 22 – 42 mo; p � 0.003).<br />

However, disease-free survival was not related to DTC-positivity. In the<br />

multivariable analysis, BM status, FIGO stage, nodal status, resection<br />

status and age were independent predictors <strong>of</strong> reduced overall survival.<br />

Interestingly, a subset <strong>of</strong> DTCs may have stem cell properties since a subset<br />

<strong>of</strong> these cells (128 out <strong>of</strong> 228 cases) were SOX2 positive, which is an<br />

embryonic stem cell marker. Conclusions: Tumor cell dissemination into<br />

bone marrow is a common phenomenon in ovarian cancer. DTC detection<br />

has the potential to become an important biomarker for prognostication and<br />

may be included as a therapeutic target in future concepts.<br />

5044 General Poster Session (Board #18D), Sun, 8:00 AM-12:00 PM<br />

What do 676 primary and recurrent ovarian cancer (OC) patients expect<br />

from their doctors and therapy management? Results <strong>of</strong> a German survey <strong>of</strong><br />

the northeastern German <strong>Society</strong> <strong>of</strong> Gynecological Oncology (NOGGO).<br />

Presenting Author: Gülten Oskay-Özcelik, NOOGO, Berlin, Germany<br />

Background: The primary aim <strong>of</strong> this study was to investigate information<br />

needs and preferences among patients with ovarian cancer, focusing<br />

especially on doctor-patient relationships and therapy management.<br />

Methods: A 42-item questionnaire was developed and validated in a<br />

mono-centre phase I study and was then provided to primary and recurrent<br />

ovarian cancer patients via internet (online) or as a print-version. In the first<br />

part basic data (age, tumour status, therapy) were requested. In the second<br />

part, most <strong>of</strong> the questions try to evaluate the expectations and needs<br />

concerning their therapy management and doctor-patients communication.<br />

Results: From January to November 2009, a total <strong>of</strong> 676 (201 online; 475<br />

print version) patients with ovarian cancer from 44 German centres took<br />

part in the survey.The median age <strong>of</strong> the online group was 49 years (range<br />

19-84), for the print group 62 years (26-92). Nearly all patients (98.7%)<br />

had a primary surgery and a primary chemotherapy (89%). Asked for side<br />

effects during therapy, the most frequent answers were alopecia, paraesthesia/dysaesthesia<br />

and fatigue. Most <strong>of</strong> the patients were content with the<br />

completeness and understandability <strong>of</strong> the explanations about the therapies<br />

from their doctors . The three most important aspects, which were<br />

proposed by patients to improve therapy against ovarian cancer were:<br />

“Doctors should have more time for explanations”, “The therapy should not<br />

lead to any loss <strong>of</strong> hair”, and “The therapy should be more effective”.<br />

Conclusions: This study underlines the high need <strong>of</strong> ovarian cancer patients<br />

to discuss all details concerning treatment options and clinical management.<br />

As matter <strong>of</strong> fact, the physician involved in the treatment is the most<br />

important source <strong>of</strong> information.<br />

Gynecologic Cancer<br />

337s<br />

5043 General Poster Session (Board #18C), Sun, 8:00 AM-12:00 PM<br />

BRCA1 immunohistochemistry in high-grade serous ovarian cancers<br />

(HGS-OC) characterized for BRCA1 germ-line mutations. Presenting Author:<br />

David Michael Hyman, Memorial Sloan-Kettering Cancer Center, New<br />

York, NY<br />

Background: The optimal use <strong>of</strong> BRCA1 germline testing in patients with<br />

HGS-OC is unclear. Moreover, BRCA1 germline testing does not provide<br />

information on non-germline mechanisms <strong>of</strong> BRCA1 loss. We investigated<br />

the performance <strong>of</strong> BRCA1 immunohistochemistry (IHC) testing in HGS-OC<br />

with known BRCA1 mutation status. Methods: Eligible patients had<br />

HGS-OC, underwent surgery at a single institution between 1997 and<br />

2010, and were tested for germline BRCA1 mutations under IRB-approved<br />

protocols. FFPE tumor tissue was used in triplicate to construct a tissue<br />

microarray (TMA). Two pathologists, blinded to BRCA1 status, independently<br />

scored each sample as BRCA1 IHC present (normal) or absent<br />

(abnormal). Whole sections were stained for all samples with absent<br />

BRCA1 staining on TMA. A commercially available monoclonal antibody<br />

against BRCA1, clone MS110 from Calbiochem (OP92) was used with<br />

previously optimized conditions. Results: 123 samples (30 BRCA1 �; 93<br />

BRCA1 wild-type) were analyzed and 47 (38%) had abnormal BRCA1 IHC.<br />

Inter-observer agreement on BRCA IHC was high (kappa�0.87). BRCA IHC<br />

had a sensitivity <strong>of</strong> 83.3% (CI: 70.0-96.7%), specificity <strong>of</strong> 76.3% (CI:<br />

67.7-85.0%), PPV <strong>of</strong> 53.2% (CI: 38.9-67.5%), and NPV <strong>of</strong> 93.4% (CI:<br />

87.9-99.0%) for BRCA1 germline mutation. There was a trend towards<br />

improved overall survival in the BRCA IHC abnormal vs. normal patients<br />

(median survival 82 vs 61 wks, HR 0.70, p�0.12). Conclusions: BRCA1<br />

IHC is a reproducible and inexpensive test with a high NPV for absence <strong>of</strong> a<br />

BRCA1 germline mutation. The 22 (17.7%) cases with abnormal BRCA1<br />

IHC and wild-type BRCA1 germline status may have other mechanisms <strong>of</strong><br />

protein loss such as promoter hypermethylation or somatic mutation and<br />

are currently being tested for these changes. This rate <strong>of</strong> non-germline<br />

BRCA1 loss in HGS-OC is consistent with data generated by the Tumor<br />

Cancer Genome Atlas Network. BRCA1 IHC may be useful as a reliable<br />

biomarker for risk stratification in HGS-OC.<br />

Germline BRCA1 status<br />

Wild-type Mutated<br />

BRCA1 IHC<br />

Normal 71 (57.2%) 5 (4.0%)<br />

Abnormal 22 (17.7%) 25 (20.2%)<br />

5045 General Poster Session (Board #18E), Sun, 8:00 AM-12:00 PM<br />

Correlative study <strong>of</strong> low serum creatinine and hematological toxicities in<br />

Japanese patients with ovarian cancer treated by dose dense TC therapy.<br />

Presenting Author: Koji Matsumoto, Medical Oncology Division, Hyogo<br />

Cancer Center, Akashi, Japan<br />

Background: Dose dense TC (ddTC) is a novel standard therapy for patients<br />

(pts) with advanced ovarian cancer, although hematological toxicities<br />

(hemTX), especially anemia, may increase as observed in NOVEL trial<br />

(JGOG3016). Low serum creatinine (LCr), especially below 0.7 mg/dl, may<br />

lead to overestimation <strong>of</strong> GFR. GOG announced that pts with LCr should use<br />

a minimum value <strong>of</strong> 0.7mg/dl to estimate GFR. The correlation between<br />

LCr and hemTX treated by ddTC is unknown. Methods: GFR was determined<br />

using the Cockcr<strong>of</strong>t-Gault formula. Serum creatinine concentrations were<br />

measured using enzymatic assays. Minimum value <strong>of</strong> 0.7 mg/dl was not<br />

used during this period <strong>of</strong> time. The carboplatin clearance was then<br />

calculated by Calvert equation. HemTX were defined as, Grade 3 or 4 (by<br />

CTC-AE ver.4) neutropenia, anemia, and thrombocytopenia. Using electrical<br />

chart, frequency <strong>of</strong> hemTX and correlation between serum creatinine<br />

(less than 0.7 or not) were examined. Results: From Feb. 2010 to Dec.<br />

2011, 61 consecutive pts were treated with ddTC. LCr was observed in<br />

73% <strong>of</strong> pts. No treatment related death occurred. Among 61 pts, 50<br />

(82%), 31 (51%), and 12 (19.6 %) pts experienced Grade3/4 neutropenia,<br />

anemia and thrombocytopenia, respectively. HemTX in pts with LCr and the<br />

others were as in the Table. Conclusions: LCr is frequent in Japanese female<br />

pts. ddTC in practice setting seems safe, and hemTX <strong>of</strong> ddTC are similar<br />

with those observed in NOVEL trial. The rationale using a minimum value <strong>of</strong><br />

0.7 mg/dl should be further studied by larger population, such as pts in<br />

NOVEL trial.<br />

G3/4 neutropenia G3/4 anemia G3/4 thrombocytopenia<br />

Pts with LCr 36 (80%) 24 (53.3%) 9 (20%)<br />

Pts without LCr 14 (87.5%) 7 (44%) 3 (18.8%)<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


338s Gynecologic Cancer<br />

5046 General Poster Session (Board #18F), Sun, 8:00 AM-12:00 PM<br />

Neo-escape: Neoadjuvant extended sequential chemotherapy with adjuvant<br />

postoperative treatment for epithelial nonmucinous advanced inoperable<br />

peritoneal malignancy. Presenting Author: Christopher John Poole,<br />

University Hospital Coventry and Warwickshire NHS Trust, Coventry,<br />

United Kingdom<br />

Background: Neo-Escape was designed to exploit fully the modest non-cross<br />

resistance <strong>of</strong> carboplatin (CBDCA) and paclitaxel (ptx) in an extended<br />

sequential regimen, with dose-dense ptx, and address feasibility <strong>of</strong> combining<br />

gemcitabine (gem) with either CBDCA or ptx. Methods: A randomised<br />

phase II trial in patients (pts) with untreated (FIGO stage 3C/4) inoperable<br />

ovarian, fallopian, or primary peritoneal carcinoma to assess feasibility <strong>of</strong><br />

two regimens <strong>of</strong> sequential neoadjuvant-then-adjuvant chemotherapy (CT);<br />

(a) CBDCA AUC 2.5 and gem 1000mg/m2 repeated days 1 and8q3wksx<br />

6 cycles, then ptx 175mg/m2 q 2 wks x 6 cycles (CG-P) or (b) CBDCA AUC 6<br />

q 3 wks x 6 cycles, then ptx 175mg/m2 and gem 2000mg/m2 q2wksx6<br />

cycles (C-PG). All pts were considered for delayed 1o debulking surgery<br />

after neoadjuvant CT. The 1o feasibility outcome was % pts completing 12<br />

cycles <strong>of</strong> CT. Using Fleming’s single stage procedure 44 patients on each<br />

arm were needed to test null hypothesis <strong>of</strong> feasibility �60% with 5%<br />

1-sided significance level and 90% power. 2o outcomes included safety,<br />

PFS and ORR. Pts were stratified by serum albumin, stage and tumor<br />

differentiation. Results: 75 pts were recruited Sept 2007 - May 2011 (28<br />

CG-P; 47 C-PG), median age 62 yr (range 21-75). Recruitment to CG-P<br />

closed early due to futility. 52% had albumin �35g/L, 68% FIGO stage 3C<br />

and 80% poorly differentiated tumors. 64% on CG-P and 55% on C-PG had<br />

debulking surgery as planned and a further 4% on CG-P and 13% on C-GP<br />

after completion <strong>of</strong> all CT. For CG-P 35% achieved 0cm, 35% �1cm and<br />

30% � 1cm residuum; for C-GP 34% 0cm, 13% �1cm and 34% �1cm,<br />

19% TBC. 14/28 pts on CG-P completed all 12 cycles (feasibility 50%;<br />

95% CI 31-67%); 37/47 pts on C-PG (feasibility 79% (95% CI 64-88).<br />

Main reason for early discontinuation was toxicity on CG-P and disease<br />

progression on C-PG. Similar proportions <strong>of</strong> pts on each arm had dose<br />

reductions (68%) or delays (86% on CG-P; 89% on C-PG), mainly for<br />

toxicity. 82% <strong>of</strong> pts experienced grade 3/4 toxicity on CG-P; 72% on C-PG.<br />

Median PFS for CG-P is 14.3 mths (95% CI 11.6-15.7mths) and 13.0<br />

mths (95% CI 11.5-15.4mths) for C-PG. Conclusions: CG-P was not<br />

feasible at these doses using pre-specified criteria, but C-PG is feasible.<br />

5048 General Poster Session (Board #18H), Sun, 8:00 AM-12:00 PM<br />

NKTR-102 in patients with platinum-resistant ovarian cancer (PROC):<br />

Modeling CA125 response and its correlation with tumor response.<br />

Presenting Author: Yen Lin Chia, Nektar Therapeutics, San Francisco, CA<br />

Background: NKTR-102 is a unique topoisomerase 1 inhibitor that provides<br />

continuous exposure to SN38. In heavily pretreated pts with PROC (median<br />

<strong>of</strong> 3 prior therapies; 27 pts were platinum refractory), 145 mg/m2 NKTR-102 given q14d or q21d demonstrated significant anti-tumor<br />

activity (JCO 28:7s, 5013). We present a PK/PD model for CA125 kinetics<br />

that can be used to project GCIG response as an aide to selecting dose and<br />

schedule. Methods: Data from 55 pts with PROC and elevated CA125<br />

(median baseline � 515 U/mL) were fit with a PK/PD model developed to<br />

correlate CA125 dynamics with SN38 conc-time pr<strong>of</strong>iles predicted from<br />

individual pt dosing history: d[CA125]/dt � Kin*exp(beta*t)*(1-[SN38]/<br />

(IC50�[SN38]))-Kout*[CA125]. Results: CA125 pr<strong>of</strong>iles were well described<br />

by the model, with a population mean SN38 IC50 <strong>of</strong> 1.1 ng/mL.<br />

Typical min and max SN38 conc during treatment were 1.5 and 3 ng/mL<br />

for q14d and 0.9 and 2.4 ng/mL for q21d, indicating that both schedules<br />

resulted in SN38 exposure near the IC50. The half-life <strong>of</strong> CA125 decline<br />

was 6.4 days, similar to literature values. 46 <strong>of</strong> 55 pts had pre- and<br />

post-treatment tumor measurements for correlation <strong>of</strong> CA125 response and<br />

tumor size. Overall best response by GCIG and RECIST correlated in 57% <strong>of</strong><br />

pts. 33% <strong>of</strong> pts with GCIG response showed declines in tumor size, albeit<br />

insufficient to classify as RECIST response. 72% <strong>of</strong> pts with GCIG SD<br />

showed at least SD by RECIST. CA125 vs time was simulated for 1000 pts<br />

receiving 145 mg/m2 NKTR-102 q14d or q21d, to allow comparison <strong>of</strong><br />

CA125 and GCIG response between schedules (see table). The % pts per<br />

GCIG response was comparable between schedules, supporting use <strong>of</strong> the<br />

better-tolerated q21d regimen. Conclusions: The PK/PD model described<br />

CA125 pr<strong>of</strong>iles well, providing a tool to predict drug response from SN38<br />

PK data. Close correlation <strong>of</strong> CA125 with tumor size is consistent with<br />

historical use <strong>of</strong> CA125 as a surrogate marker. Predicted CA125 pr<strong>of</strong>iles for<br />

NKTR-102 q14d or q21d were similar, suggesting that the better-tolerated<br />

q21d schedule will produce results consistent with those from Ph 2.<br />

Model-predicted % pts by response.<br />

CR PR SD PD<br />

q14d 19 23 44 14<br />

q21d 16 22 46 16<br />

5047 General Poster Session (Board #18G), Sun, 8:00 AM-12:00 PM<br />

Changes <strong>of</strong> serum glycome in patients suffering from ovarian cancer.<br />

Presenting Author: Veronique Blanchard, Institute <strong>of</strong> Laboratory Medicine,<br />

<strong>Clinical</strong> Chemistry and Pathobiochemistry, Charité Berlin, Berlin, Germany<br />

Background: Protein glycosylation plays an important role in many biological<br />

processes. Most human serum proteins, with the exception <strong>of</strong> albumin,<br />

are glycosylated. Glycosylation is known to be altered with development <strong>of</strong><br />

diseases such as cancer. In the case <strong>of</strong> ovarian cancer, tumor markers<br />

among them CA-125 that are clinically used are known to have poor<br />

specificity. In addition, they fail to detect the disease at an early stage.<br />

Therefore, better biomarkers are needed. The aim <strong>of</strong> the present research<br />

work is to identify new potential glycan biomarkers by analyzing the serum<br />

N-glycome <strong>of</strong> patients suffering from ovarian cancer Methods: Serum was<br />

collected from 67 patients as well as from 33 healthy age-matching<br />

women. N-glycans were released from 10 ul serum by PNGase F digestion,<br />

permethylated and subsequently analyzed by means <strong>of</strong> MALDI-TOF mass<br />

spectrometry. The SPSS s<strong>of</strong>tware was used for the statistical analysis.<br />

Results: The N-glycome <strong>of</strong> patients was found to have more fucosylated<br />

structures, especially in tri- and tetraantennary sialylated glycans. The PCA<br />

analysis indicates that there are significant differences between the<br />

glycome <strong>of</strong> ovarian cancer patients in all stages <strong>of</strong> the disease and the<br />

glycome <strong>of</strong> healthy controls. We identified 14 potential structures that were<br />

divided in two categories, one <strong>of</strong> mon<strong>of</strong>ucosylated structures with high<br />

antennarity (sensitivity 94%, specificity 97%) and one containing highmannose<br />

structures and an asialylated structures (sensitivity 97%, specificity<br />

97%). Conclusions: Our study is the first trial to identify major<br />

differences between ovarian cancer sera and control sera, which could<br />

potentially be used in the future as biomarkers.<br />

5049 General Poster Session (Board #19A), Sun, 8:00 AM-12:00 PM<br />

Longitudinal health-related quality <strong>of</strong> life assessment: Implications for<br />

prognosis in ovarian cancer. Presenting Author: Donald Peter Braun,<br />

Cancer Treatment Centers <strong>of</strong> America, Zion, IL<br />

Background: Several studies in the oncology literature have demonstrated<br />

the prognostic value <strong>of</strong> baseline quality <strong>of</strong> life (QoL). However, there is little<br />

to no information on the prognostic effects <strong>of</strong> changes in QoL during<br />

treatment. We investigated whether changes in QoL could predict survival<br />

in ovarian cancer patients treated with an integrative model <strong>of</strong> care.<br />

Methods: We evaluated 137 ovarian cancer patients treated at our institution<br />

between Jan 2001 and Dec 2009 who were available for a minimum<br />

follow-up <strong>of</strong> 3 months. QoL was evaluated at baseline and after 3 months <strong>of</strong><br />

treatment using EORTC-QLQ-C30. The QLQ-C30 incorporates a global<br />

scale, 5 function scales and 8 symptom scales. Patient survival was defined<br />

as the time between date <strong>of</strong> first patient visit and date <strong>of</strong> death from any<br />

cause/date <strong>of</strong> last contact. Cox regression was performed to evaluate the<br />

prognostic significance <strong>of</strong> baseline and changes in QoL scores after<br />

adjusting for age, treatment history and stage at diagnosis. Results: Mean<br />

age at diagnosis was 51.1 years. 28 patients were newly diagnosed while<br />

109 were previously treated. Stage at diagnosis was I, 16; II, 15; III, 72; IV,<br />

28; and 6 indeterminate. Median overall survival was 33.5 months (95%<br />

CI: 11.5-55.6 months). Baseline QoL scale predictive <strong>of</strong> survival upon<br />

multivariate analysis was nausea/vomiting (p�0.04). Associations between<br />

changes in QoL and survival were observed for global function, appetite loss<br />

and constipation. Every 10-point increase (improvement) in global function<br />

from baseline to 3 months was associated with a 10% decreased risk <strong>of</strong><br />

death (HR�0.90; 95% CI�0.81 to 0.99, p�0.03). The corresponding<br />

HRs for 10-point increase (deterioration) in appetite loss and constipation<br />

scales were 1.20 (1.06 to 1.35; p�0.005) and 1.13 (1.02, 1.24;<br />

p�0.02) respectively. Conclusions: This exploratory study provides some<br />

preliminary evidence to indicate that ovarian cancer patients whose QoL<br />

improves within 3 months <strong>of</strong> treatment have a significantly increased<br />

survival time compared to those who fail to demonstrate improvement.<br />

These findings might be used in clinical practice to systematically address<br />

QoL-related problems <strong>of</strong> ovarian cancer patients throughout their treatment<br />

course.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


5050 General Poster Session (Board #19B), Sun, 8:00 AM-12:00 PM<br />

Technetium-99m sulfur colloid (TSC) as a phenotypic probe for predicting<br />

pharmacokinetics (PK) and palmar-plantar erythrodysesthesia (PPE) toxicity<br />

<strong>of</strong> pegylated liposomal doxorubicin (PLD) in patients (pts) with recurrent<br />

epithelial ovarian cancer (EOC). Presenting Author: Hugh Giovinazzo,<br />

University <strong>of</strong> North Carolina at Chapel Hill, Chapel Hill, NC<br />

Background: There is significant variability in the PK and PD (efficacy and<br />

toxicity) <strong>of</strong> PLD. Clearance (CL) <strong>of</strong> PLD has been proposed to occur<br />

primarily via uptake by cells <strong>of</strong> the mononuclear phagocyte system (MPS)<br />

mainly located in the liver, spleen, and blood. TSC is a radioactive colloid<br />

used clinically for diagnostic and functional imaging <strong>of</strong> the MPS. Our aim is<br />

to evaluate TSC as a phenotypic probe <strong>of</strong> MPS activity, which may predict<br />

PLD PK and PPE toxicity in pts (n�9) with EOC. Methods: Prior to<br />

administration <strong>of</strong> PLD on cycle 1 day 1 (C1D1), TSC was administered at<br />

10 mCi IVP � 1. Dynamic planar and SPECT/CT images <strong>of</strong> the liver, spleen<br />

and hands were acquired using a gamma camera. Blood samples were<br />

collected up to 60 min after TSC and analyzed for radioactivity using a<br />

gamma counter. On C1D1, PLD was administered at 40 mg/m2 alone or at<br />

30 mg/m2 in combination with carboplatin IV over 1 to 3 h. Serial PK<br />

samples were obtained from 0h to 672h post PLD dose. Plasma was<br />

processed to measure encapsulated and released doxorubicin (dox) using<br />

solid phase separation and HPLC. CL <strong>of</strong> PLD and TSC were calculated by<br />

non-compartmental analysis. The grade <strong>of</strong> PPE toxicity was determined by<br />

NCI CTCAE (v4.03) criteria. Results: Nine patients have undergone TSC<br />

imaging and eight patients completed TSC blood PK and PLD plasma PK<br />

studies. There was a positive linear relationship between TSC CL and<br />

encapsulated dox CL (R2 � 0.61, p�0.02). When patients were subdivided,<br />

there was a stronger relationship between TSC CL and encapsulated<br />

dox CL (R2 � 0.81, p�0.03) in pts receiving PLD monotherapy. A positive<br />

relationship using Spearman’s correlation (��0.84, p�0.006) was also<br />

found between maximum grade PPE toxicity developed and estimated AUC<br />

<strong>of</strong> encapsulated dox in hands [(TSC AUCBlood)/(TSC AUCHand)*Encapsulated Dox AUCPlasma]. Conclusions: These results suggest that TSC is a probe for<br />

MPS function and PLD PK and PD and may be used to individualize PLD<br />

therapy in pts with EOC. In addition, our findings suggest TSC may be able<br />

to predict the development <strong>of</strong> PPE in patients.<br />

5052 General Poster Session (Board #19D), Sun, 8:00 AM-12:00 PM<br />

Panitumumab and pegylated liposomal doxorubicin in platinum-resistant<br />

epithelial ovarian cancer with KRAS wild-type: The PaLiDo study, a phase II<br />

nonrandomized multicenter study. Presenting Author: Karina Dahl Steffensen,<br />

Department <strong>of</strong> <strong>Clinical</strong> Oncology and Radiotherapy, Vejle Hospital,<br />

Vejle, Denmark<br />

Background: Ovarian cancer (OC) patients with platinum-resistant recurrent<br />

disease have few therapeutic options and the response rates are only<br />

10-20% using non-cross-resistant chemotherapeutic agents. The increasing<br />

number <strong>of</strong> negative trials for OC treatment has prompted an evaluation<br />

<strong>of</strong> new biologic agents, which in combination with chemotherapy may<br />

result in improvement in survival. Panitumumab is a fully human monoclonal<br />

antibody specific to the epidermal growth factor receptor (EGFR). No<br />

previous studies have evaluated the effect <strong>of</strong> panitumumab in OC based on<br />

KRAS mutation status. The main purpose was to investigate the response<br />

rate in platinum-resistant, KRAS wild-type OC patients treated with<br />

pegylated liposomal doxorubicin (PLD) supplemented with panitumumab.<br />

Methods: Major eligibility criteria were confirmed stage I-IV primary<br />

epithelial ovarian/fallopian/peritoneal cancer patients with progression<br />

either during or within 6 months after end <strong>of</strong> first or second line<br />

platinum-based chemotherapy. Only patients with measurable disease by<br />

CA125 criteria and with KRAS wild type were eligible. Patients were treated<br />

with panitumumab 6 mg/kg day 1 and day 15 and with PLD 40 mg/m² day<br />

1, every 4 weeks. Tumor assessment was performed at baseline and at every<br />

third cycle according to CA-125 criteria. Results: A total <strong>of</strong> 46 patients were<br />

enrolled by 6 study sites in this multi-institutional phase II trial. Within the<br />

population evaluable for response (N�33), there was 8 CA125 responders<br />

for an overall response rate <strong>of</strong> 24.3 %. Progression-free and overall survival<br />

in the intention-to-treat population (N�43) was 2.7 months (2.5-3.2<br />

months, 95%CI) and 8.1 months (5.6-11.7 months, 95%CI), respectively.<br />

The most common treatment related grade 3 toxicities included skin<br />

toxicity (42%), fatigue (19%) and vomiting (12%). Conclusions: The<br />

combination <strong>of</strong> PLD and panitumumab demonstrates efficacy in platinum<br />

refractory/resistant patients although the dermatologic toxicity was considerable.<br />

Gynecologic Cancer<br />

339s<br />

5051 General Poster Session (Board #19C), Sun, 8:00 AM-12:00 PM<br />

Sexual function, sexual activity, and quality <strong>of</strong> life in women with ovarian<br />

and endometrial cancer. Presenting Author: Philipp Harter, Kliniken Essen<br />

Mitte, Essen, Germany<br />

Background: Gynecological cancer (GC) is generally assumed to have an<br />

impact on sexual function and activity. Although there are several studies<br />

addressing the issue, case control studies are currently limited. Methods:<br />

We performed a cross-sectional investigation <strong>of</strong> sexual function and activity<br />

utilizing the sexual activity questionnaire, the female sexual function<br />

index, and parts <strong>of</strong> the EORTC QLQ C30. Patients with gynecological<br />

cancer (GC) like ovarian and endometrial cancer were compared with a<br />

control group (C) <strong>of</strong> non-cancer patients. Inclusion <strong>of</strong> GC was only allowed if<br />

treatment was completed �12 months previously and patients were<br />

disease-free. Results: The questionnaires were sent out to 727 women (335<br />

x GC and 392 x C), 22.8% <strong>of</strong> which responded. Response rates in both<br />

groups were equivalent (79 pts with GC [23.6%] and 87 control subjects<br />

[22.2%]). Median age was 57 years (C) and 62 years (GC), respectively<br />

(p�0.237). 51.5% (C) and 59.5% (GC) were not sexually active, mainly<br />

owing to lack <strong>of</strong> a partner (37%) or lack <strong>of</strong> interest (21%) in controls and<br />

lack <strong>of</strong> interest (40%, p�0.05), self-reported physical problems (31.9%,<br />

p�0.05), and physical problems <strong>of</strong> the partner (21%, p�0.05). There<br />

were significant differences between both groups in the SAQ discomfort<br />

score (p�0.05). We did not observe significant differences in quality <strong>of</strong> life<br />

or other scores regarding sexuality. Conclusions: About half <strong>of</strong> the women in<br />

both groups were not sexually active. However, reasons for non-activity<br />

differ. Quality <strong>of</strong> sexuality tends to be impaired in GC patients, but this<br />

seems not to influence quality <strong>of</strong> life. A shift <strong>of</strong> priority caused by<br />

substantial anxiety regarding cancer specific survival might explain our<br />

findings.<br />

5053 General Poster Session (Board #19E), Sun, 8:00 AM-12:00 PM<br />

Effect <strong>of</strong> tumor-infiltrating T-lymphocytes on the aggressiveness <strong>of</strong> epithelial<br />

ovarian tumors. Presenting Author: Ines Vasconcelos, Department <strong>of</strong><br />

Gynaecology, European Competence Center for Ovarian Cancer, Campus<br />

Virchow Klinikum, Charité-Universitätsmedizin Berlin, Berlin, Germany<br />

Background: Borderline ovarian tumors (BOT) are a low-grade form <strong>of</strong><br />

ovarian malignancy with significantly less aggressive behavior than epithelial<br />

ovarian cancer (EOC). Since there is neither convincing scientific<br />

evidence nor a marker capable <strong>of</strong> predicting BOT’s behavior, we analyzed<br />

the role <strong>of</strong> immune tolerance in differential disease outcome. EOC and BOT<br />

were chosen due to similar disease etiology, despite different disease<br />

courses. Increased numbers <strong>of</strong> T-cell subpopulations infiltrate malignant<br />

tumors, so we used epigenetic tests to determine the prevalence <strong>of</strong><br />

regulatory T-cells (Treg) and overall-T-Lymphocytes (oTL) in EOC and BOT.<br />

Methods: The ovarian cancer samples were provided by the European<br />

multicentric OVCAD study. The BOT samples were obtained from Tumor<br />

Bank Ovarian Cancer at Charité Campus Virchow (Germany). Samples and<br />

clinical data were prospectively collected and documented using validated<br />

SOPs. DNA was bisulphite-converted and forwarded to methylationspecific<br />

RT-PCR for CD3 and FOXP3 loci. Results: We evaluated 90<br />

high-grade EOC, 12 BOT with invasive implants and 25 non-invasive BOT<br />

samples. Higher oTL-values correlate with mortality (p�0.008) and recurrence<br />

(p�0.001), while higher Treg levels correlate with recurrence<br />

(p�0.028). Patients with non-invasive BOT have lower oTL (p�0.019) and<br />

Treg (p�0.0005) levels than patients with invasive BOT and EOC, while<br />

BOT (both invasive and non-invasive) patients have lower Treg-to-oTL ratios<br />

than EOC patients (p�0.0005). Finally, oTL levels associate with overall<br />

survival in EOC (p�0.042). Conclusions: We observed a strict correlation<br />

between tumor-type, Treg and oTL levels, as well as Treg-to-oTL ratio. This<br />

is in agreement with our hypothesis that disease aggressiveness is associated<br />

with the amount tumor-infiltrating lymphocytes and may provide the<br />

explanation for differing disease courses in EOC and BOT.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


340s Gynecologic Cancer<br />

5054 General Poster Session (Board #19F), Sun, 8:00 AM-12:00 PM<br />

An updated safety analysis <strong>of</strong> OCEANS, a randomized, double-blind, phase<br />

III trial <strong>of</strong> gemcitabine (G) and carboplatin (C) with bevacizumab (BV) or<br />

placebo (PL) followed by BV or PL to disease progression (PD) in patients<br />

with platinum-sensitive (Plat-S) recurrent ovarian cancer. Presenting<br />

Author: Carol Aghajanian, Memorial Sloan-Kettering Cancer Center, New<br />

York, NY<br />

Background: OCEANS met its primary end point with a statistically<br />

significant and clinically meaningful hazard ratio for progression-free<br />

survival (PFS) <strong>of</strong> 0.484. Secondary end points included objective response<br />

rate, overall survival, and safety. Eleven additional months <strong>of</strong> safety data<br />

were collected after the data cut-<strong>of</strong>f date for the final PFS analysis event<br />

and updated safety analyses were performed. Methods: Eligible patients<br />

(pts) were randomized to arm A: GC (G [1000 mg/m 2 days 1 and 8] and C<br />

[AUC 4, day 1], q21d for 6–10 cycles) � concurrent PL (q21d), followed<br />

by PL until PD or unacceptable toxicity; or arm B: GC � concurrent BV (15<br />

mg/kg q21d), followed by BV until PD or unacceptable toxicity. All adverse<br />

events (AE) were recorded and graded per NCI-CTCAE v3.0. Results: The<br />

incidence <strong>of</strong> any grade AE was 100% in both arms and <strong>of</strong> SAEs was 25.3%<br />

(PL arm) and 35.6% (BV arm). The rates <strong>of</strong> proteinuria, hypertension<br />

(HTN), reversible posterior leukoencephalopathy syndrome (RPLS), thrombocytopenia<br />

and epistaxis were higher in the BV arm. More pts in the BV<br />

arm (20.6%) than in the PL arm (4.7%) experienced an AE that led to<br />

discontinuation <strong>of</strong> study drug, in the BV arm most commonly due to HTN<br />

(4%), proteinuria (2.8%), epistaxis (1.2%), thrombocytopenia (1.6%) and<br />

RPLS (0.8%). Median number <strong>of</strong> BV cycles in pts with G �3 proteinuria<br />

was 22.5 and the AE resolved in 91.7% <strong>of</strong> pts. Among the pts with G �3<br />

HTN, the median number <strong>of</strong> BV cycles was 16.5 and the AE resolved in<br />

72.7% <strong>of</strong> pts. Conclusions: The overall safety pr<strong>of</strong>ile was similar to that seen<br />

at the time <strong>of</strong> the final PFS analysis. Higher incidences <strong>of</strong> proteinuria and<br />

HTN were possibly related to longer BV treatment duration and resolved in<br />

the majority <strong>of</strong> pts.<br />

Adverse event, n (%)<br />

GC � PL<br />

(n�233)<br />

GC � BV<br />

(n�247)<br />

Bleeding (non-CNS), all G 64 (27.5) 158 (64)<br />

Bleeding (non-CNS), G >3 2 (0.9) 14 (5.7)<br />

Epistaxis, G >3 1 (0.4) 12 (4.9)<br />

HTN, all G 20 (8.6) 107 (43.3)<br />

HTN, G >3 1 (0.4) 44 (17.8)<br />

Fistula/abscess, all G 1 (0.4) 4 (1.6)<br />

GIP, all G 0 (0) 0 (0)<br />

Proteinuria, G >3 2 (0.9) 24 (9.7)<br />

RPLS, all G 0 (0) 2 (0.8)<br />

Thrombocytopenia, G >3 79 (34) 99 (40)<br />

5056 General Poster Session (Board #19H), Sun, 8:00 AM-12:00 PM<br />

The impact <strong>of</strong> interval cytoreduction and age in advanced-stage ovarian<br />

cancer: A GOG ancillary study. Presenting Author: Stuart M. Lichtman,<br />

Memorial Sloan-Kettering Cancer Center, New York, NY<br />

Background: To determine if an age group exists for which interval<br />

cytoreductive surgery (ICS) in patients with suboptimal residual disease at<br />

primary surgery influences progression free survival (PFS) and overall<br />

survival (OS) among women with advanced ovarian cancer treated on GOG<br />

182. Methods: GOG 182 was a prospective, randomized trial <strong>of</strong> first-line<br />

chemotherapy in patients with advanced ovarian cancer. Patients with both<br />

optimal and suboptimal residual disease were included, and those with<br />

suboptimal residual were considered for ICS, with intent registered and<br />

stratified prior to randomization. Patients were randomized to one <strong>of</strong> five<br />

chemotherapy arms, employing combinations <strong>of</strong> either two or three agents<br />

delivered intravenously, with a control arm <strong>of</strong> paclitaxel and carboplatin. A<br />

retrospective analysis was approved by the GOG Ancillary Study Committee<br />

to investigate the influence <strong>of</strong> age on treatment and outcomes. In that<br />

analysis, Cox regression was used to identify independent prognostic<br />

factors and estimate their covariate effects on the adjusted PFS and OS <strong>of</strong><br />

patients with suboptimal residual disease. Statistical significance was set<br />

at p�0.05. Results: Among the entire eligible study population, 28%<br />

(n�1,042) were registered with suboptimal residual disease (� 1 cm) and<br />

109 <strong>of</strong> these patients elected to undergo ICS. Hazard ratios (HR) were<br />

determined for patients undergoing ICS with reference to patients with<br />

suboptimal disease not undergoing ICS. Based on the most current<br />

follow-up data, the HR for progression or death was not statistically<br />

different between the groups, but the HR <strong>of</strong> death alone was significant at<br />

0.72 (95% CI: 0.57–0.92, p�0.008). There was no significant association<br />

<strong>of</strong> age with ICS in either the PFS or the OS model. Conclusions: In this<br />

trial, a patient’s age did not influence the effect <strong>of</strong> ICS on PFS or OS. There<br />

is no demonstrable benefit in PFS associated with ICS, but there was a<br />

statistically significant improvement in OS. To elucidate this finding,<br />

further study is warranted, likely in the form <strong>of</strong> a meta-analysis incorporating<br />

data from other prospective trials.<br />

5055 General Poster Session (Board #19G), Sun, 8:00 AM-12:00 PM<br />

The impact <strong>of</strong> obesity on time to recurrence in ovarian cancer: A<br />

retrospective study. Presenting Author: Karina E Hew, Mercy Medical<br />

Center, Baltimore, MD<br />

Background: There has been conflicting data regarding the relationship<br />

between obesity and progression free survival in patients with ovarian<br />

cancer. There has been some evidence to suggest that obesity results in<br />

altered tumor biology and a poorer prognosis in these patients. The aim <strong>of</strong><br />

this study was to examine whether obesity is a risk factor for time to<br />

recurrence in primary epithelial ovarian cancer. Methods: A multicenter<br />

retrospective chart review was performed at Mercy Medical Center and<br />

University <strong>of</strong> Michigan Medical Center. 591 patients were diagnosed with<br />

primary epithelial ovarian cancer between 2004-2009. However, 221<br />

patients were excluded from the analysis because <strong>of</strong> persistent or progressive<br />

disease, treatment with neoadjuvant chemotherapy, presence <strong>of</strong><br />

synchronous tumors or incomplete follow-up data. 370 patients were<br />

eligible for analysis. Data collected included: height and weight at the time<br />

<strong>of</strong> surgery, age, race, medical co-morbid illnesses, tumor stage, grade and<br />

histology. Treatment related data such as number <strong>of</strong> cycles <strong>of</strong> adjuvant<br />

chemotherapy; and optimal versus suboptimal tumor debulking was also<br />

collected. Body mass index (BMI) was defined according to WHO 2004<br />

criteria. Women with a BMI greater than 30 were categorized as obese. The<br />

diagnosis <strong>of</strong> recurrence was made by positive radiological or pathological<br />

diagnosis <strong>of</strong> cancer recurrence after patient had surgery, received adjuvant<br />

chemotherapy and had no clinical, radiological or serological evidence <strong>of</strong><br />

recurrence during this time. The time to recurrence was then quantified in<br />

terms <strong>of</strong> months from the initial surgery. Survival analyses were performed<br />

with the Kaplan-Meier method and compared using log-rank testing. Time<br />

to recurrence was analyzed using Mann-Whitney U and Wilcox W tests.<br />

Results: 130 (35%) obese patients were compared with 240 (65%) non<br />

obese patients. A recurrence was documented in 125 (47.9%) non obese<br />

patients and 49 (37.7%) obese patients. Time to recurrence between both<br />

BMI groups was found to be identical, at 15 months (p�1.0). The<br />

progression free survival was similar in both obese and non obese subjects<br />

(p�0.118). Conclusions: Obesity does not impact the time to recurrence in<br />

patients with primary ovarian cancer.<br />

5057 General Poster Session (Board #20A), Sun, 8:00 AM-12:00 PM<br />

The impact <strong>of</strong> interval from surgery to chemotherapy and age in advancedstage<br />

ovarian cancer: A GOG ancillary study. Presenting Author: Andrew<br />

Menzin, North Shore University Hospital, Manhasset, NY<br />

Background: To determine if an age group exists for which the interval from<br />

surgery to the initiation <strong>of</strong> chemotherapy influences progression free<br />

survival (PFS) and overall survival (OS) among women with advanced<br />

ovarian cancer treated on GOG 182. Methods: GOG 182 was a prospective,<br />

randomized trial <strong>of</strong> first-line chemotherapy in patients with advanced<br />

ovarian cancer, including those with optimal and suboptimal residual<br />

disease. Patients were randomized to one <strong>of</strong> five chemotherapy arms,<br />

employing combinations <strong>of</strong> either two or three agents delivered intravenously,<br />

with a control arm <strong>of</strong> paclitaxel and carboplatin. Chemotherapy was<br />

to be initiated within 12 weeks <strong>of</strong> primary surgery. A retrospective analysis<br />

was approved by the GOG Ancillary Study Committee to investigate the<br />

influence <strong>of</strong> age on treatment and outcomes. In that analysis, Cox<br />

regression was used to identify independent prognostic factors and estimate<br />

their covariate effects on the adjusted PFS and OS <strong>of</strong> the study<br />

population. Statistical significance was set at p�0.05. Results: The primary<br />

analysis <strong>of</strong> GOG 182 showed no differences in PFS or OS for any <strong>of</strong> the<br />

experimental arms when compared to the control regimen, and it was felt<br />

that the data from all arms could be aggregated for this analysis. Data for all<br />

regimens was pooled, and the time interval from surgery to chemotherapy<br />

was examined as a prognostic factor <strong>of</strong> survival. The interval had no<br />

statistically significant association with PFS (p�0.105). In the OS model,<br />

though, the functional form <strong>of</strong> the log interval was both significantly linear<br />

and nonlinear (p�0.001). After being flat until about 20 days (21.2 days;<br />

95% CI, 15.0–28.2 days), the plot <strong>of</strong> log interval against log hazard<br />

increases, suggesting a changepoint time after which the associated risk <strong>of</strong><br />

death increases. Conclusions: In this study, time interval from surgery to<br />

chemotherapy had no impact on PFS, but there was evidence <strong>of</strong> a potential<br />

time-dependent relationship with OS, which might be elucidated with a<br />

meta-analysis incorporating data from other prospective trials. Our observations<br />

did not appear to be influenced by patient age. Overall, these findings<br />

can reassure patients who are recuperating from extensive cancer surgery.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


5058 General Poster Session (Board #20B), Sun, 8:00 AM-12:00 PM<br />

Treatment reality in elderly patients with advanced ovarian cancer: A<br />

prospective analysis <strong>of</strong> the OVCAD consortium. Presenting Author: Linn<br />

Lena Woelber, University Medical Center Hamburg-Eppendorf, Hamburg,<br />

Germany<br />

Background: Approximately one third <strong>of</strong> women diagnosed with ovarian<br />

cancer are 70 years or older. Standard therapy <strong>of</strong> ovarian cancer including<br />

radical cytoreduction and combination chemotherapy has considerable<br />

morbidity and information regarding treatment reality in elderly patients<br />

with ovarian cancer is very limited. Methods: Patients with primary<br />

epithelial ovarian cancer FIGO-stages IIB-IV were prospectively included in<br />

5 European cancer centers. All patients underwent surgery with the intent<br />

<strong>of</strong> maximal cytoreduction and platinum-based chemotherapy. To analyze<br />

treatment strategies and outcome in the elderly, patients were subdivided<br />

in �70 years and �70 years <strong>of</strong> age and compared regarding clinicopathological<br />

variables and prognosis. Results: A total <strong>of</strong> 275 patients were<br />

included and followed for a median <strong>of</strong> 25 months. Median age <strong>of</strong> the total<br />

cohort was 58 (18-85) years with only 47 patients (17.1%) �70 years old.<br />

Age itself was not a prognostic factor for progression free survival (PFS) in<br />

multivariate analysis. 30-days mortality rate after primary surgery was<br />

3.6% in elderly patients compared to 0.6% in patients �70 years<br />

(p�0.153). Surgery was less radical in patients �70 (e.g. fewer lymph<br />

node dissections p�0.001) and the percentage <strong>of</strong> patients with residual<br />

disease after surgery was higher in elderly (44.7%) compared to younger<br />

patients (28.5%) despite similar FIGO stage distribution (p�0.029).<br />

Furthermore, elderly received more <strong>of</strong>ten mono-chemotherapy (p�0.001).<br />

Consequently, outcome was less favorable in patients �70 compared to<br />

patients �70 years (75% overall survival 16 vs. 28 months; p�0.002 and<br />

median PFS 14 vs. 20 months; p�0.182). Conclusions: In this prospective<br />

European multicenter study, ovarian cancer patients age 70 and older were<br />

treated significantly less radical and had unfavorable outcome compared to<br />

younger patients. Specific trials for elderly patients focusing on surgical as<br />

well as chemotherapeutic aspects are therefore highly desirable to gain<br />

more information in an aging society.<br />

5060 General Poster Session (Board #20D), Sun, 8:00 AM-12:00 PM<br />

The effect <strong>of</strong> the APPRISE mandate on use <strong>of</strong> erythropoiesis-stimulating<br />

agents and transfusion rates in ovarian cancer patients. Presenting Author:<br />

Jonathan David Boone, University <strong>of</strong> Alabama at Birmingham, Birmingham,<br />

AL<br />

Background: Erythropoiesis-stimulating agents (ESAs) have long been used<br />

to support chemotherapy-induced anemia in patients with epithelial<br />

ovarian cancer (EOC). Recent studies have demonstrated that ESAs may<br />

lead to increased tumor growth and shorter survival. The FDA mandated<br />

new guidelines (APPRISE, for Assisting Providers and Cancer Patients with<br />

Risk Information for the Safe use <strong>of</strong> ESAs) for consenting patients before<br />

ESA administration. We sought to quantify the change in ESA use and RBC<br />

transfusion rates after the APPRISE mandate was instituted. Methods: A<br />

retrospective chart review identified patients with EOC undergoing chemotherapy<br />

after initiating the APPRISE mandate. A similar subset <strong>of</strong> patients<br />

treated prior to the APPRISE guidelines served as a control cohort.<br />

Abstracted data included patient demographics, primary or second line<br />

treatment status, chemotherapy regimen, number <strong>of</strong> patients requiring<br />

ESA or RBCs, or both, and a cost savings analysis. Results: 84 patients with<br />

EOC were identified as having undergone 367 cycles <strong>of</strong> first or second line<br />

chemotherapy after the APPRISE guidelines were instituted. A matched set<br />

<strong>of</strong> 88 patients receiving 613 cycles <strong>of</strong> chemotherapy within a year prior to<br />

institution <strong>of</strong> the APPRISE guidelines was analyzed for comparison. The<br />

two groups were statistically similar. Most patients in each group were<br />

initially diagnosed with advanced stage disease, were receiving primary<br />

chemotherapy, and were receiving taxane/platinum-based chemotherapy.<br />

45 <strong>of</strong> 88 patients (51%) in the pre-APPRISE group received a total <strong>of</strong> 196<br />

ESA injections compared to 0 <strong>of</strong> 84 patients (0%) in the post-APPRISE<br />

group. In spite <strong>of</strong> discontinuing the use <strong>of</strong> ESAs and no change in<br />

transfusion thresholds, RBC transfusion in the post-APPRISE group was<br />

similar to that in the pre-APPRRISE group (8.3% vs. 14.8%, p�.28).<br />

Omission <strong>of</strong> ESAs in the post-APPRISE group resulted in a cost-savings <strong>of</strong><br />

an estimated $700,000 in billable charges. Conclusions: In our institution,<br />

the APPRISE guidelines resulted in complete cessation <strong>of</strong> ESA use in<br />

patients with EOC, resulting in considerable cost savings. Importantly, RBC<br />

transfusion rates did not increase after the guidelines were imposed.<br />

Gynecologic Cancer<br />

341s<br />

5059 General Poster Session (Board #20C), Sun, 8:00 AM-12:00 PM<br />

NGR-hTNF and doxorubicin in relapsed ovarian cancer (OC). Presenting<br />

Author: Domenica Lorusso, Department <strong>of</strong> Gynecologic Oncology, National<br />

Cancer Institute, Milan, Italy<br />

Background: NGR-hTNF (asparagine-glycine-arginine human tumor necrosis<br />

factor) is able to promote antitumor immune responses and to improve<br />

the intratumoral doxorubicin (D) uptake by selectively damaging tumor<br />

vessels. Methods: OC patients (pts) with progressive disease (PD) after � 1<br />

platinum/taxane regimen and with a platinum free interval lower than 6<br />

months (PFI �6) or ranging from 6 to 12 months (PFI 6-12) received<br />

NGR-hTNF (N) 0.8 �g/m2 and D 60 mg/m2 on day 1 every 3 weeks. Primary<br />

endpoint <strong>of</strong> this phase 2 trial was response rate by RECIST criteria with a<br />

target <strong>of</strong> � 6/37 responding pts. Secondary aims were progression free<br />

survival (PFS) and overall survival (OS). Results: 37 pts (median age 57<br />

years; PS 0/1 32/5; PFI � 6/6-12 25/12; prior regimens 1-5) were<br />

enrolled. Median baseline peripheral blood lymphocyte count (PBLC) was<br />

1.6/mL (interquartile range 1.2-2.1). In all, 177 cycles were given, with 18<br />

pts (49%) receiving � 6 cycles and 12 pts (32%) 8 cycles. Neither grade<br />

3/4 adverse events (AEs) related to N nor increase <strong>of</strong> D-related AEs were<br />

noted. Common grade 1/2 AEs included chills (65%). Eight pts (23%; 95%<br />

CI 12-39) had partial response (PR; 2 with PFI � 6 and 6 with PFI 6-12;<br />

median duration: 8.2 months). Fifteen pts had stable disease (SD, 43%;<br />

10 with PFI � 6 and 5 with PFI 6-12; median duration: 4.9 months) for an<br />

overall disease control (DC, PR�SD) rate <strong>of</strong> 66%. Mean changes from<br />

baseline in target tumor size after 2, 4, 6, and 8 cycles were 2%, -54%,<br />

-69%, and -77%, respectively. Median PFS was 5.0 months (95% CI<br />

3.1-6.9) and median OS was 17.0 months (10.4-23.6). In pts with PFI � 6<br />

or 6-12, median PFS were 3.8 and 7.8 months (p�.03) and median OS<br />

were 14.3 and 20.1 months (p�.14), respectively. Pts with DC had longer<br />

median OS than those with early PD (24.0 and 4.9 months, respectively,<br />

p�.02). Longer PFI (p�.03) and higher PBLC (p�.01) were associated<br />

with better PFS, while OS correlated only with PBLC (p�.001). In the<br />

subset with PFI � 6, pts with PBLC � or � 1.2/mL (1st quartile) had<br />

median PFS <strong>of</strong> 4.9 and 2.6 months (p�.02) and median OS <strong>of</strong> 15.8 and<br />

4.3 months (p�.0001), respectivel Conclusions: A randomized phase II<br />

trial is currently testing D � NGR-hTNF in pts with PFI � 6 (refractory/<br />

resistant). The role <strong>of</strong> PBLC as blood-based biomarker deserves further<br />

investigation.<br />

5061 General Poster Session (Board #20E), Sun, 8:00 AM-12:00 PM<br />

A phase Ib study <strong>of</strong> the combination <strong>of</strong> temsirolimus (T) and pegylated<br />

liposomal doxorubicin (PLD) in advanced or recurrent breast, endometrial,<br />

and ovarian cancer. Presenting Author: Marye Boers-Sonderen, Department<br />

<strong>of</strong> Medical Oncology, Radboud University Nijmegen Medical Centre,<br />

Nijmegen, Netherlands<br />

Background: PLD is active in metastatic breast, endometrial and ovarian<br />

cancer. Preclinical studies suggest that mTOR inhibitors (mTORi), such as<br />

T, have an additive therapeutic effect to chemotherapy and resistance to<br />

doxorubicin can be reversed by adding an mTORi. Therefore, the combination<br />

<strong>of</strong> T and PLD is highly promising. Methods: This phase I study assessed<br />

the maximum tolerated dose (MTD), safety and activity <strong>of</strong> the combination<br />

<strong>of</strong> T and PLD in advanced or recurrent breast, endometrial or ovarian<br />

cancer. Patients (pts) who were not previously treated with PLD or T, with<br />

adequate organ function were eligible. After a two week run in period with T<br />

iv once weekly, PLD iv once every four weeks was added. In case <strong>of</strong> clinical<br />

benefit, pts were treated for a maximum <strong>of</strong> 9 cycles combination therapy. T<br />

could be continued as monotherapy afterwards. The MTD was defined as<br />

the highest dose at which � 1 dose limiting toxicity (DLT) had been<br />

observed among 6 pts. FDG PET scans were performed at baseline and after<br />

2 and 6 wks to assess the effect on tumor metabolism. Pharmacokinetic<br />

(PK) sampling was performed during cycle 1. Results: 20 pts were enrolled.<br />

On the 4th dose level with 20 mg T and 40 mg/m2 PLD 2 DLTs occurred in<br />

6 pts: a grade 3 thrombocytopenic bleeding and a grade 3 skin toxicity.<br />

Therefore, the MTD was assessed at 15 mg T and 40 mg/m2 PLD. Adverse<br />

events (all grades/grade 3-4 in %) occurring most frequently were fatigue<br />

(84/5), nausea (84/16), mucositis (79/21), vomiting (74/16) and anorexia<br />

(74/0) Furthermore, rash and hand foot syndrome occurred both in 53% <strong>of</strong><br />

pts, with 11% and 21% grade 3 respectively. 3 pts had a confirmed PR and<br />

9 had SD (� 3 months). The mean progression free survival (PFS) was 4.9<br />

months with 2 pts still on treatment. Results <strong>of</strong> FDG PET and PK data are<br />

currently being analyzed and will be presented. Conclusions: The combination<br />

<strong>of</strong> T and PLD is safe and tolerable. The MTD was assessed at PLD 40<br />

mg/m2 once every 4 weeks and T 15 mg weekly. The activity <strong>of</strong> this<br />

combination in breast, endometrial and ovarian cancer pts is promising and<br />

warrants further studies.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


342s Gynecologic Cancer<br />

5062^ General Poster Session (Board #20F), Sun, 8:00 AM-12:00 PM<br />

Phase I safety study <strong>of</strong> farletuzumab, carboplatin, and pegylated liposomal<br />

doxorubicin (PLD) in patients with platinum-sensitive epithelial ovarian<br />

cancer (EOC). Presenting Author: Kenneth H. Kim, University <strong>of</strong> Alabama<br />

at Birmingham, Birmingham, AL<br />

Background: Farletuzumab (FAR) is a humanized monoclonal antibody that<br />

binds to folate receptor-�, a target which is largely absent in normal<br />

epithelium and over-expressed in EOC. A phase I and a phase II study<br />

indicate potential utility <strong>of</strong> FAR in combination with carboplatin/paclitaxel<br />

in recurrent platinum sensitive EOC. This study assessed safety and<br />

efficacy <strong>of</strong> FAR in combination with PLD/carboplatin, which has been<br />

demonstrated to have greater efficacy than carboplatin/paclitaxel in women<br />

with platinum-sensitive EOC. Methods: A multicenter, single-arm study<br />

enrolled 15 patients with platinum-sensitive EOC in first or second relapse<br />

defined by either CA-125 or RECIST. The primary endpoint is to assess<br />

safety <strong>of</strong> FAR plus PLD/carboplatin in this population. Patients were<br />

treated with weekly FAR 2.5 mg/kg plus carboplatin AUC 4-5 and PLD 30<br />

mg/m2 every 4 weeks for 6 cycles, followed by maintenance treatment with<br />

single agent FAR until disease progression. Initial dosing <strong>of</strong> weekly 2.5<br />

mg/kg was later amended to 7.5 mg/kg every 3 weeks. Results: 13 <strong>of</strong> 15<br />

patients completed 6 cycles <strong>of</strong> combination therapy; one received a total <strong>of</strong><br />

12 cycles <strong>of</strong> chemotherapy. Two patients progressed during combination<br />

therapy: one received 3 cycles, the other 5 cycles. The median number <strong>of</strong><br />

subsequent single agent maintenance cycles was 8 (range 0-13). No grade<br />

4 toxicities were observed. Of 20 grade 3 toxicities in 14 patients, 3 (1<br />

fatigue; 2 small bowel obstructions in the same patient) were deemed by<br />

the investigator to be possibly related to FAR. These events were seen<br />

during the combination phase and were deemed by the investigator to be<br />

also possibly related to chemotherapy. All patients demonstrated clinical<br />

benefit: 1 complete response, 10 partial response and 4 stable disease. 7<br />

patients have come <strong>of</strong>f study, all for disease progression; a median PFS <strong>of</strong><br />

11 months was noted. 8 patients remain on study. Conclusions: These<br />

preliminary data indicate that the safety pr<strong>of</strong>ile <strong>of</strong> FAR in combination with<br />

carboplatin and PLD is consistent with that historically recorded for<br />

carboplatin and PLD alone. Complete safety pr<strong>of</strong>ile will be assessed; PK<br />

data and final results will be presented.<br />

5064 General Poster Session (Board #20H), Sun, 8:00 AM-12:00 PM<br />

Application <strong>of</strong> an ointment with high radical protection factor as a<br />

prevention strategy against PPE. Presenting Author: Franziska Kluschke,<br />

Center <strong>of</strong> Applied Cutaneous Physiology (CCP), Department <strong>of</strong> Dermatology,<br />

Charité-Universitätsmedizin Berlin, Berlin, Germany<br />

Background: Pegylated liposomal doxorubicin has proved to be highly<br />

efficient in the treatment <strong>of</strong> various tumors. Depending on the application<br />

protocol, up to 80% <strong>of</strong> patients develop palmar-plantar erythrodysesthesia<br />

(PPE). So far, a prevention strategy is still unknown. Recently, it was shown<br />

that parts <strong>of</strong> the chemotherapeutics were excreted with the sweat onto the<br />

skin surface, spreading there homogeneously and penetrating into the<br />

stratum corneum. The formation <strong>of</strong> free radicals in the tissue results in<br />

PPE. The aim <strong>of</strong> the study was to investigate if a topically applied ointment<br />

containing antioxidants with high radical protection factor (RPF) can be a<br />

PPE prevention strategy. Methods: 20 patients with ovarian carcinoma, who<br />

had been treated with pegylated liposomal doxorubicin (40 mg/m2 ), were<br />

investigated. They applied the ointment at least twice daily, 2 days before<br />

and during 3 cycles <strong>of</strong> chemotherapy. Their skin condition was examined by<br />

a trained dermatologist. Results: From 20 patients enrolled in the study,<br />

only 12 (60%) met the conditions by applying the cream at least twice daily<br />

in the palmar and plantar regions. These patients did not develop PPE. One<br />

patient died in the 2nd cycle <strong>of</strong> therapy. 7 patients (35%) did not follow the<br />

ointment application protocol for various reasons; 6 <strong>of</strong> them developed PPE<br />

and resumed ointment application thereafter. As a result, PPE disappeared<br />

or was strongly reduced in these patients so that chemotherapy could be<br />

continued. Due to the small group <strong>of</strong> patients, the fact that PPE was not<br />

induced in patients who had applied the ointment regularly can be<br />

generalized only restrictedly. Far more interesting are the findings in those<br />

patients, who had stopped ointment application during chemotherapy and<br />

developed PPE, which disappeared after they resumed applying the<br />

ointment. The regression or distinct reduction <strong>of</strong> PPE after re-application <strong>of</strong><br />

the ointment clearly proves the efficacy <strong>of</strong> this strategy. Conclusions: The<br />

topical application <strong>of</strong> an ointment containing antioxidants with high radical<br />

protection factor (RPF) could be an efficient strategy against the development<br />

<strong>of</strong> PPE during chemotherapy.<br />

5063 General Poster Session (Board #20G), Sun, 8:00 AM-12:00 PM<br />

Factors associated with an increased risk <strong>of</strong> recurrence in women with<br />

ovarian granulosa cell tumors. Presenting Author: Anuj Suri, The University<br />

<strong>of</strong> North Carolina Hospitals, Division <strong>of</strong> Gynecologic Oncology, Chapel Hill,<br />

NC<br />

Background: There is emerging data in different malignancies, including<br />

gynecologic cancers, demonstrating factors independent <strong>of</strong> a patient’s<br />

tumor characteristics, which are associated with an increased risk <strong>of</strong> cancer<br />

recurrence. The goal <strong>of</strong> this study is to determine demographic and<br />

prognostic factors affecting recurrence <strong>of</strong> disease (RD) in women with<br />

ovarian granulosa cell tumors (GCT). Methods: A dual-institution retrospective<br />

analysis <strong>of</strong> patients diagnosed with GCT between 1995 and 2010.<br />

Demographics including age, race, BMI, stage, diabetes (DM), adjuvant<br />

treatment, and progression free survival (PFS) were extracted. Hazard<br />

ratios for recurrence were estimated by univariate and multivariate Cox<br />

regression models Results: One hundred nine women identified with a<br />

median age <strong>of</strong> 50 (range 12-87). Fifty-six (57.1%) were Caucasian, 32<br />

(32.7%) African <strong>American</strong>, and 10 (10.2%) were other. Median BMI was<br />

29 (range 12-57). Twenty-one patients had DM. The majority <strong>of</strong> women<br />

had stage I disease (89.0%), 7 (6.4%) had stage II/III disease, and 5 were<br />

unstaged. In univariate analysis, DM showed the strongest association with<br />

recurrence (HR 3.56, 95% CI: 1.57-8.11). Non-white race was also<br />

associated with higher risk <strong>of</strong> RD, though the association was not<br />

statistically significant (HR 1.74, 95% CI: 0.78-3.87). The association<br />

between DM and RD was also found in multivariate analysis controlling for<br />

all factors including non-white race and BMI (HR 2.99, 95% CI: 1.11-<br />

8.08). Only 19 (17%) patients received adjuvant chemotherapy consisting<br />

<strong>of</strong> one <strong>of</strong> two different adjuvant chemotherapy regimens bleomycin,<br />

etoposide, and cisplatin or paclitaxel and carboplatin, however there was no<br />

difference in outcome based on chemotherapy regimen (p�0.24).<br />

Conclusions: This is the largest study analyzing factors associated with risk<br />

<strong>of</strong> recurrence in women with ovarian GCT. Studies have demonstrated<br />

higher morbidity among diabetic patients with breast and colon cancer and<br />

this may be modifiable with metformin. Our results emphasize that<br />

diabetes is one <strong>of</strong> the strongest predictors <strong>of</strong> recurrent disease in patients<br />

with ovarian GCT. Further studies are necessary to evaluate the effect <strong>of</strong><br />

other factors such as adjuvant chemotherapy.<br />

5065 General Poster Session (Board #21A), Sun, 8:00 AM-12:00 PM<br />

A phase I, dose escalation trial to assess the safety and biological activity <strong>of</strong><br />

recombinant human interleukin-18 (SB-485232) in combination with<br />

pegylated liposomal doxorubicin in platinum-resistant recurrent ovarian<br />

cancer. Presenting Author: Fiona Simpkins, University <strong>of</strong> Miami Miller<br />

School <strong>of</strong> Medicine and Sylvester Comprehensive Cancer Center, Miami, FL<br />

Background: SB-485232, a recombinant human form <strong>of</strong> interleukin-18<br />

(IL-18), is being studied as a novel cytokine for tumor immunotherapy.<br />

IL-18 has been shown to increase the anti-tumor activity <strong>of</strong> doxorubicin in<br />

other cancer mouse models. Methods: This multi-center study enrolled 16<br />

patients (pts), and evaluated four cycles (28 days each) <strong>of</strong> standard<br />

pegylated liposomal doxorubicin (PLD) (40 mg/m2 day 1) in combination<br />

with increasing doses <strong>of</strong> IL-18 (1, 3, 10, 30, and 100 mg/kg) on days 2 and<br />

9 <strong>of</strong> each cycle witha1yrfollow-up to further evaluate safety and efficacy.<br />

Results: Of the 16 pts, 10 (63%) completed study treatment and 6 (37%)<br />

did not because <strong>of</strong> disease progression (5 pts) and PLD hypersensitivity (1<br />

pt). Thirteen (82%) were platinum resistant/refractory having received a<br />

median <strong>of</strong> � 3 prior lines <strong>of</strong> therapy. SB-485232 up to 100 mg/kg in<br />

combination with PLD had an acceptable safety pr<strong>of</strong>ile. All 16 pts<br />

experienced at least 1 AE including chills (81%), nausea (75%), anemia<br />

(63%), fatigue (56%), hyperglycemia, and pyrexia (50% each). Eight <strong>of</strong> 16<br />

pts had Grade 3 AEs including anemia (19%), and 6% each for abdominal<br />

pain, asthenia, dehydration, PLD hypersensitivity, edema, fatigue, hyperglycemia,<br />

hyperkalemia, jaundice, pain, nausea, vomiting, and pyelonephritis.<br />

There were no grade 4/5 AEs. Four subjects experienced 10 non-fatal<br />

SAEs with 3 related to study drug: anemia, PLD hypersensitivity, and<br />

cytokine release syndrome. Maximal SB-485232 biological activity, assessed<br />

by peripheral blood NK and T cell markers, was observed at<br />

10mg/kg-100mg/kg. This drug combination resulted in a 6% partial<br />

response rate (RR) and a 38% stable disease rate using RECIST 1.0.<br />

Median (95% CI) time <strong>of</strong> progression-free survival (PFS) was 4.50 (3.52,--)<br />

months. Conclusions: The SB-485232/PLD combination was tolerable with<br />

minimal toxicity. These preliminary efficacy data are comparable to the<br />

historical RR and PFS observed with PLD monotherapy in platinumresistant<br />

ovarian cancer. However, given the small sample size, this<br />

combination warrants further investigation.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


5066 General Poster Session (Board #21B), Sun, 8:00 AM-12:00 PM<br />

Effect <strong>of</strong> BRCA mutation on prognosis in patients with ovarian cancer: A<br />

systematic review and meta-analysis. Presenting Author: Antonis Valachis,<br />

Department <strong>of</strong> Oncology Sörmland Mälarsjukhuset, Eskilstuna, Sweden<br />

Background: To investigate whether the germ-line mutations <strong>of</strong> BRCA 1 and<br />

2 in patients with ovarian cancer are associated with disease outcome.<br />

Methods: A systematic search <strong>of</strong> the literature was performed using search<br />

terms related to BRCA, ovarian cancer and prognosis. Studies were<br />

considered eligible if they reported the outcome <strong>of</strong> ovarian cancer in<br />

patients with BRCA 1 and/or 2 mutations compared with patients with<br />

sporadic ovarian cancer or / and no BRCA mutation. Cohort and casecontrol<br />

designs were included. Pooled Hazard Ratios (HR) were estimated<br />

with fixed or random effects models, depending on between-studies<br />

heterogeneity. Results: Of 2,340 titles identified, 26 articles met the<br />

inclusion criteria. Of these, 6 studies were excluded from the analysis due<br />

to duplication <strong>of</strong> results (n�1) and lack <strong>of</strong> data to calculate HR (n�5).<br />

Patients with BRCA 1 or 2 mutations had better survival compared with<br />

control group both in cohort (HR: 0.58, 95% Confidence Interval (CI)<br />

0.42-0.79, p-value 0.0005) and in case-control (HR: 0.65, 95% CI<br />

0.45-0.93, p-value � 0.02) studies. When only patients with BRCA 1<br />

mutation were analyzed, the survival benefit remained significant (HR:<br />

0.67, 95% CI 0.53-0.85, p-value � 0.001). Furthermore, the differential<br />

survival benefit <strong>of</strong> the mutation groups was even more evident among<br />

patients with BRCA 2 mutation (HR: 0.43, 95% CI: 0.30-0.63, p-value �<br />

0.0001). A direct comparison <strong>of</strong> patients with BRCA 1 vs. BRCA 2<br />

mutations (4 studies) revealed a significantly better survival for BRCA 2<br />

mutation carriers (HR: 0.53, 95% CI: 0.33-0.85, p-value � 0.009).<br />

Conclusions: Our results confirm the hypothesis that BRCA status is a<br />

prognostic factor in patients with ovarian cancer. Based on these results,<br />

the use <strong>of</strong> BRCA status as a stratifying factor in therapeutic ovarian cancer<br />

trials seems to be fully justifiable. The stronger association between<br />

survival and BRCA 2 mutation, compared with BRCA 1, suggests a different<br />

nature <strong>of</strong> the dysfunction <strong>of</strong> these 2 genes.<br />

5068 General Poster Session (Board #21D), Sun, 8:00 AM-12:00 PM<br />

A phase II study <strong>of</strong> pazopanib in recurrent or persistent ovarian (EOC),<br />

peritoneal (PPC), or Fallopian tube cancer (FTC): A Spanish Ovarian Cancer<br />

Group (GEICO) study. Presenting Author: Ana Oaknin, Medical Oncology<br />

Department, Vall d´Hebron University Hospital, Barcelona, Spain<br />

Background: Pazopanib (P) is a potent and selective multi-targeted receptor<br />

tyrosine kinase inhibitor <strong>of</strong> VEGFR-1, VEGFR-2, VEGFR-3, PDGFR-a/�, and<br />

c-kit that inhibits angiogenesis. Signaling blockade <strong>of</strong> these pathways is<br />

associated with anti-tumor and anti- angiogenesis activity. Methods: Eligible<br />

patients (pts) had persistent or recurrent EOC/PPC /FTC up to 2 prior<br />

cytotoxic regimens. They had to have received at least a platinum–based<br />

line and fulfill platinum resistant criteria. Treatment consisted <strong>of</strong> P 800 mg<br />

orally QD until disease progression or prohibitive toxicity. The primary<br />

endpoint was <strong>Clinical</strong> Benefit Rate (CBR) defined as Complete Response<br />

(CR) plus <strong>Part</strong>ial Response (PR) plus Stable Disease (SD) � 8 weeks by<br />

RECIST v1.1.An optimal two-stage Simon design was utilized with H1and<br />

H0set at 60%and 40% respectively; Power �90% significance level <strong>of</strong> 5%<br />

(Stage 1: � 25pts; total�66). Correlative studies to identify angiogenic<br />

biomarkers to predict response to P were performed. Results: From 12/10 to<br />

7/11, 25 pts were enrolled, 21 pts had EOC, 2PPT, and 2FTC. Median age:<br />

64 years (range 43-81), ECOG 0/1/2:12/11/2 pts. Prior chemotherapy<br />

regimens 1/2:10/15 pts. Median weeks on treatment: 8 (range 4-25). Most<br />

frequent adverse events (AEs) were asthenia (56%), hypertension (36%)<br />

and diarrhea, nausea and anorexia (20% each).Grade 3 toxicities: Hypertension<br />

(6pts), ALT/AST elevations (3 pts), asthenia (2 pts), DVT (1pt), Fistula<br />

(1 pt), Anemia (1 pt). Six pts required dose reduction to 600 mg due to<br />

toxicity.Reasons for stopping study treatment: PD (18pts), AEs (3pts) and<br />

investigator decision (2pts). First stage analysis showed: PR:1/25, SD:<br />

9/25, CBR:10/25; 40% (95% CI 21.1%-61.3%).No correlation between<br />

GCIG CA-125 response and RECIST criteria was established, 8 versus 1<br />

response respectively. Median PFS was 1.83 months (95% CI 1.67-2).<br />

Conclusions: The CBR observed at the first stage did not reach the planned<br />

statistical hypothesis (CBR:12 pts). Therefore, the lack <strong>of</strong> activity <strong>of</strong> P in<br />

platinum resistant EOC/PPT/FT led to discontinuation <strong>of</strong> the study.<br />

Translational study results will be presented in an additional abstract.<br />

Gynecologic Cancer<br />

343s<br />

5067 General Poster Session (Board #21C), Sun, 8:00 AM-12:00 PM<br />

Management <strong>of</strong> antiangiogenics’ renovascular safety in ovarian cancer<br />

subgroup and intermediate results <strong>of</strong> the MARS study. Presenting Author:<br />

Vincent Launay-Vacher, Service ICAR, Pitie-Salpetriere Hospital, Paris,<br />

France<br />

Background: Anti-VEGF drugs (AVD) are widely used in cancer patients<br />

(pts). Hypertension (HTN) and proteinuria (Pu) are class-side-effects <strong>of</strong><br />

AVD, related to the inhibition <strong>of</strong> the VEGF pathway. The MARS study has<br />

been conducted to assess the renovascular tolerance <strong>of</strong> these drugs in the<br />

clinical setting. Methods: Hypertension (HTN) and proteinuria (Pu) are<br />

class-side-effects <strong>of</strong> anti-VEGF drugs (AVD), related to the inhibition <strong>of</strong> the<br />

VEGF pathway. The MARS study has been conducted to assess the<br />

renovascular tolerance <strong>of</strong> these drugs in the clinical setting. Results: Among<br />

77 OC pts been included, 38 completed the study to date (1-year follow-up<br />

(f/u)). Median age at inclusion (introduction <strong>of</strong> the AVD) was 62 years.<br />

Diabetes and HTN prevalences were 5.2% and 7.9%, respectively. Baseline<br />

renal assessment retrieved: Pu 13.2%, Hu 7.9%, mean aMDRD 80.9<br />

ml/min/1.73m 2<br />

and 3 pts with aMDRD�60. The incidence <strong>of</strong> de novo Pu<br />

during f/u was 36.4% (Table). All pts with Pu at inclusion improved, except<br />

one. Among pts with de novo Pu, 58.3% afterwards improved/normalized.<br />

No Grade 3/4 Pu has been reported (at inclusion or during f/u) and no Hu.<br />

17.1% developed HTN. In addition, a mean renal function decrease <strong>of</strong> -2.7<br />

ml/min/1.73m2 /year was observed and 4 pts had aMDRD�60 at the end <strong>of</strong><br />

f/u. 36.4% had grade 1 SCr increase (median increase <strong>of</strong> 15.9%) No<br />

thrombotic micro-angiopathy (TMA) has been reported. Conclusions: The<br />

results <strong>of</strong> the MARS subgroup <strong>of</strong> OC pts shows that 1) TMA remains rare, 2)<br />

Pu develops in 36.4% <strong>of</strong> the pts, however with no Grade 3/4, 3) less than<br />

20% developed HTN, and 4) renal function was not especially impaired.<br />

Furthermore, in case <strong>of</strong> a renovascular effect, investigators followed the<br />

recommendations from the French <strong>Society</strong> <strong>of</strong> Nephrology (Halimi JM et Al.<br />

Nephrol Ther 2008) and no treatment withdrawal for unmanageable<br />

renovascular toxicity occurred.<br />

Prevalence at inclusion<br />

Incidence during f/u<br />

Renovascular effects<br />

(%)<br />

(%)<br />

Pu*<br />

Grade 1<br />

13.2<br />

10.5<br />

36.4<br />

21.2<br />

Grade 2<br />

2.7<br />

15.2<br />

Grade 3-4<br />

0.0<br />

0.0<br />

HTN 7.9 17.1<br />

Hu<br />

Traces/�<br />

7.9<br />

5.3<br />

0.0<br />

���<br />

2.6<br />

SCr increase*<br />

- 36.4<br />

Grade 1<br />

36.4<br />

Grade 2-4<br />

*NCI-CTC v4.03.<br />

0.0<br />

5069 General Poster Session (Board #21E), Sun, 8:00 AM-12:00 PM<br />

A novel treatment for ovarian cancer (OC): Anti-Müllerian inhibiting<br />

substance type II receptor (MISRII) humanized monoclonal antibody (mAb)<br />

3C23K—Preclinical validation. Presenting Author: Christine Gaucher, LFB<br />

Biotechnologies, Loos, France<br />

Background: Expressed on most OC subtypes while displaying a restricted<br />

expression pr<strong>of</strong>ile in adult normal tissues, MISRII represents a potentialtarget<br />

for OC immunotherapy. We present here the preclinical assessment <strong>of</strong> a<br />

humanized anti-MISRII EMABling mAb, 3C23K. Methods: Either quantitative<br />

RT-PCR or immunohistochemistry (IHC) studies were performed to<br />

confirm MISRII expression pr<strong>of</strong>ile in Granulosa Cell Tumor (GCT) or<br />

Epithelial OC (EOC) patient samples and to evaluate tissue cross-reactivity.<br />

For in vitro and in vivo experiments, we have generated 4 patient-derived<br />

MISRII expressing EOC cell lines. Xenograft studies were conducted in<br />

swiss nude mice on established tumors (100 mm3 ). Mice received 2 to 3<br />

weekly i.p. injections (10 mg/kg/inj) for 4 to 6 wks and tumor volumes were<br />

compared with control groups. Comparison <strong>of</strong> i.p. vs i.v. injections were<br />

assessed as well as combination with carboplatin (once a week for 4 weeks,<br />

60 mg/kg/inj). In addition, 3C23K plasma level was monitored to determine<br />

half-life. Results: 1) Target validation: we confirmed by IHC the<br />

expression <strong>of</strong> MISRII in most OC tissue sections (4/4 GCT and 13/14 EOC),<br />

meanwhile, MISRII mRNA was only detected in 7/48 normal tissues. 2) In<br />

vitro assessment: tested in vitro 3C23K displayed both cytotoxic (ADCC)<br />

and anti-proliferative activities. 3) In vivo assessment: in the mouse<br />

xenograft models 3C23K exhibited a strong anti-tumoral activity as<br />

measured by tumor volume, with T/C ratios reaching values below 0.42<br />

shortly after the initiation <strong>of</strong> treatment. No differences in efficacy were<br />

noticed between i.p. and i.v. injections or between thrice vs twice a week<br />

administrations. In addition, similar half lives were observed for 3C23K<br />

injected either i.v. (96.9 h) or i.p (113.5 h). Finally, the combination <strong>of</strong><br />

3C23K with carboplatin (CP), a standard <strong>of</strong> care in OC, exhibited an even<br />

stronger anti-tumor activity with T/C values at D22 <strong>of</strong> 0.06 (3C23K�CP),<br />

0.18 (3C23K) and 0.69 (CP) vs vehicle. Conclusions: 3C23K represents a<br />

promising candidate for OC targeted therapy and a dose-escalation phase I<br />

study is planned in patients with OC.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


344s Gynecologic Cancer<br />

5070 General Poster Session (Board #21F), Sun, 8:00 AM-12:00 PM<br />

BRCA1 methylation status in high-grade serous ovarian cancer (HGSOC)<br />

patients (pts). Presenting Author: Ilary Ruscito, Department <strong>of</strong> Gynecology<br />

and Obstetrics, Sapienza University <strong>of</strong> Rome, Rome, Italy<br />

Background: Mutations in BRCA1/2 genes contribute to increase risk for<br />

breast and ovarian cancer (OC).Hypermethylation <strong>of</strong> CpG islands in gene<br />

promoter regions is considered a frequent mechanism associated with<br />

inactivation <strong>of</strong> tumor suppressor genes which contributes to oncogenic<br />

transformation. Studies identified methylation changes in OC pts. Most <strong>of</strong><br />

the studies analyzed a small cohort and combined different histological<br />

subtypes. HGSOC is a special entity <strong>of</strong> OC associated with poorer OS. Aim<br />

<strong>of</strong> this study was to analyze the clinical impact <strong>of</strong> BRCA1 promoter gene<br />

methylation status in HGSOC. Methods: The cohort comprised 127 HGSOC<br />

treated by cytoreduction followed by platinum-based chemotherapy at<br />

Department <strong>of</strong> Gynecology, Charité Medical University Berlin, Germany.<br />

Fresh frozen OC tissues provided by TOC were examined to access the<br />

tumoral tissue quality. Samples presenting at least 50% <strong>of</strong> tumour area<br />

were included within this study. DNA was extracted followed by sodium<br />

bisulfite conversion, and assessment <strong>of</strong> BRCA1 gene promoter methylation<br />

rate was determined by PCR. Statistical analisys has been carried out using<br />

SPSS s<strong>of</strong>tware. Results: In our study, 14.2% <strong>of</strong> the pts presented a<br />

hypermethylation <strong>of</strong> the BRCA1 promoter gene. The hypermethylation <strong>of</strong><br />

BRCA1 promoter gene was significantly more frequently encountered in the<br />

younger pts (�59 yrs: 77.8% in the Hypermethylation Group vs 47.7% in<br />

the Hypomethylation Group, p�0.022). No differences in methylation<br />

status between primary and metastatic tissue from OC pts could be<br />

observed. Also no correlation with FIGO stage was observed. Optimal tumor<br />

debulking could be reached in most <strong>of</strong> the pts (57.5%), without significant<br />

differences between both groups. Platinum response rates were similar<br />

between both groups. No differences in PFS and OS could be observed.<br />

Conclusions: The study shows that HGSOC pts presenting the BRCA1 gene<br />

promoter in the methylated status, developed the disease significantly<br />

earlier with respect to OC patients with unmethylated BRCA1 gene<br />

promoter. The clinical outcome was similar in HGSOC independently from<br />

BRCA1 methylation status.<br />

5072 General Poster Session (Board #21H), Sun, 8:00 AM-12:00 PM<br />

Exposure-response relationship <strong>of</strong> open-label (OL) AMG 386 monotherapy<br />

in patients (pts) with recurrent ovarian cancer. Presenting Author: Beth Y.<br />

Karlan, Cedars-Sinai Medical Center, Division <strong>of</strong> Gynecologic Oncology,<br />

Los Angeles, CA<br />

Background: AMG 386, an investigational peptibody, inhibits tumor angiogenesis<br />

by preventing angiopoietins 1/2 and Tie2 receptor interaction. A<br />

phase II, double-blind, controlled study (NCT00479817) evaluated toxicity<br />

and efficacy <strong>of</strong> AMG 386 � paclitaxel (P) in recurrent ovarian cancer<br />

pts. Increased AMG 386 exposure in that study was associated with longer<br />

progression-free survival (PFS; Lu et al., JCO 2010; 28 [Suppl]: 5042).<br />

Here, these analyses were applied to pts electing OL AMG 386 monotherapy<br />

after progression in the placebo group. Methods: Pts with recurrent<br />

epithelial ovarian, fallopian tube, or peritoneal cancer (�3 prior anticancer<br />

therapies, GOG �1) were randomized 1:1:1 to P IV QW (3 on/1 <strong>of</strong>f) � AMG<br />

386 (3 or 10 mg/kg) or placebo IV QW. In the placebo group, pts who<br />

progressed and stayed eligible could receive OL AMG 386 10 mg/kg IV QW<br />

monotherapy. In pts receiving AMG 386 monotherapy, post-hoc analyses<br />

evaluated PFS per RECIST, objective response rate (ORR), adverse events<br />

(AEs), and the relationships between high and low AMG 386 exposure<br />

(based on a median AUCSS <strong>of</strong> 10.6 mg*hr/mL) and efficacy (PFS per<br />

RECIST, ORR). Results: 161 pts were randomized. 18 pts received OL AMG<br />

386 monotherapy (range: 1-56 infusions). At the time <strong>of</strong> analysis, PFS was<br />

3.2 mo. ORR was 0%. 6 pts had stable disease (SD), 7 had progressive<br />

disease (PD). Common AEs were abdominal pain (n � 6), fatigue (n � 5),<br />

peripheral edema (n � 5), nausea (n � 5), urinary tract infection (n � 5),<br />

and vomiting (n � 5); grade � 3 AEs in more than 1 pt were pleural effusion<br />

(n � 2; all grade 3), small intestinal obstruction (n � 2; all grade 3), and<br />

ovarian cancer (n � 2; all grade 5). PFS was longer in the high- (n � 9) vs<br />

low-exposure (n � 9) group (7.2 vs 1.8 mo; HR � .266, p � .041).<br />

Precrossover PFS was similar between high- and low-exposure groups (5.5<br />

vs 4.5 mo; HR � 1.146, p � .812). For both groups, ORR was 0%. In the<br />

high-exposure group, 4 pts had SD, 3 had PD. In the low-exposure group, 2<br />

pts had SD, 4 had PD. Conclusions: In line with results from a phase II study<br />

<strong>of</strong> AMG 386 � P, pts with recurrent ovarian cancer who received AMG 386<br />

monotherapy and had high AMG 386 exposure had longer PFS than pts<br />

with low exposure. Toxicity was similar in both groups.<br />

5071 General Poster Session (Board #21G), Sun, 8:00 AM-12:00 PM<br />

Interval versus primary tumor debulking surgery in advanced ovarian<br />

cancer: Analysis <strong>of</strong> the European OVCAD data. Presenting Author: Christina<br />

Fotopoulou, Charité University Department <strong>of</strong> Gynecology, Campus Virchow<br />

Clinic, Berlin, Germany<br />

Background: The international scenery around optimal primary treatment <strong>of</strong><br />

advanced ovarian cancer (AOC) patients (pts) is currently being discussed,<br />

with large discrepancies and heterogeneity still existing between the<br />

national guidelines worldwide. Aim <strong>of</strong> the present study was to evaluate the<br />

differences in outcome <strong>of</strong> AOC-pts after primary (PDS) versus intervaldebulking-surgery<br />

(IDS) based on a prospectively assessed multicenter<br />

data set. Methods: Overall outcome was analyzed from the OVCAD database;<br />

a prospective, observational, multicenter project. AOC-pts who<br />

underwent surgery in five specialized gynecological cancer centers across<br />

three European countries between 02/2005 and 12/2008 were evaluated.<br />

Overall (OS) and progression free survival (PFS) were calculated by<br />

Kaplan-Meier-curves. Univariate and Cox-regression-analysis were applied.<br />

Results: Overall, 256 AOC pts (FIGO-stage III/IV) were evaluated. Fifty pts<br />

(19.5%) underwent IDS and 206 pts (80.5%) PDS. Despite the nonrandomized<br />

setting both groups were well balanced in terms <strong>of</strong> FIGO-stage,<br />

grading, histological subtype and presence <strong>of</strong> ascites. Different selection<br />

criteria were however present for each center. PDS pts presented significantly<br />

higher rates <strong>of</strong> intestinal resection (44.2% vs.24%; p�0.01) and<br />

lymphonodectomy compared to IDS ones (72.3%vs.48%; p�0.001), by<br />

equivalent complete tumor resection rates (67.5% vs.68%; p�0.82).<br />

Platinum response was significantly higher in PDS vs. IDS pts (80.6% vs.<br />

54%; p�0.001). 3-years OS was with 66.7% (95%CI: 60.2-73.2%)<br />

significantly better in PDS- versus 48.3% (95%CI: 34.2-62.5%) in IDS pts<br />

(p�0.001). Also 2-years PFS was with 31.9% (95%CI:24.8-39.1%)<br />

significantly higher in PDS- vs. 11.4% (95%CI: 0.9-22%) in IDS-pts<br />

(p�0.001). In multivariate analysis PDS, but not age, ascites, FIGO-stage,<br />

grading, histology or residual tumor were <strong>of</strong> prognostic significance for<br />

platinum response. In addition, multivariate analysis identified PDS and no<br />

residual tumor to positively and ascites to negatively affect OS and PFS.<br />

Conclusions: PDS appears to be associated with a more favorable outcome<br />

compared to IDS in highly specialized centers according to this nonrandomized<br />

data set.<br />

5073 General Poster Session (Board #22A), Sun, 8:00 AM-12:00 PM<br />

Targeting therapy based on preclinical analysis <strong>of</strong> clinical, molecular, and<br />

functional characteristics <strong>of</strong> individual high-grade serous ovarian cancers.<br />

Presenting Author: Monique Topp, Walter and Eliza Hall Institute <strong>of</strong><br />

Medical Research, Melbourne, Australia<br />

Background: Recent molecular exploration <strong>of</strong> high-grade epithelial ovarian<br />

cancer (OC) has revealed potential targets for novel therapy based on<br />

altered DNA repair function, deregulated pathways and recurrent amplifications<br />

(Cancer Genome Atlas Research Network. 2011. Nature 474).<br />

Improved pre-clinical models allowing analysis <strong>of</strong> specific molecular<br />

subsets <strong>of</strong> ovarian cancer are urgently required to test novel treatment<br />

strategies. Methods: We have generated a novel xenograft model <strong>of</strong> human<br />

high-grade serous OC (HG-SOC). Histologic, functional and molecular<br />

analysis <strong>of</strong> the novel xenograft cohort (at baseline and following xenotransplantation)<br />

allows stratification <strong>of</strong> individual HG-SOC for testing with<br />

appropriate targeted therapy. We perform functional analysis <strong>of</strong> in vitro<br />

Homologous Recombination (HR) DNA repair and drug response capabilities<br />

on fresh human HG-SOC immediately following surgical resection.<br />

Molecular classification (similar to Tothill [Clin Canc Res. 2008;14]);<br />

analysis <strong>of</strong> NHEJ pathway (Proc Natl Acad Sci. 2011;108) and other DNA<br />

repair genes (Proc Natl Acad Sci USA 2011;108) is performed. In vivo drug<br />

response is studied in murine xenografts. Results: Sixteen chemotherapynaive<br />

potentially HG-SOC samples and associated clinical data have been<br />

collected. Functional evidence <strong>of</strong> DNA repair (HR) capability and response<br />

to DNA damaging agents will be presented, including IHC for markers <strong>of</strong><br />

DNA damage (gH2AX), DNA repair (RAD51AP1) and apoptosis (capsase 3<br />

cleavage). Molecular classification, DNA repair gene and DNA repair<br />

pathway analyses are underway. Twelve HG-SOC have been transplanted<br />

and 6 <strong>of</strong> the first 8 have successfully xenografted, with serial transplantation<br />

and phenotyping <strong>of</strong> xenograft derivatives underway. In vivo drug<br />

response will be presented. Conclusions: This xenograft model will enable<br />

us to address hypotheses generated by recent molecular analyses <strong>of</strong> human<br />

HG-SOC (Cancer Genome Atlas Research Network. 2011. Nature 474; Clin<br />

Canc Res. 2008;14). <strong>Clinical</strong>, functional and molecular annotation will<br />

allow pre-clinical drug testing based on the plausible hypothesis approach.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


5074 General Poster Session (Board #22B), Sun, 8:00 AM-12:00 PM<br />

Proliferation pathway aberration frequencies in BRCA1- and BRCA2mutated<br />

ovarian cancers. Presenting Author: Deborah A. Zajchowski,<br />

Clearity Foundation, San Diego, CA<br />

Background: Large-scale genomic analyses <strong>of</strong> high-grade, advanced-stage<br />

serous ovarian cancers by The Cancer Genome Atlas (TCGA) project<br />

revealed aberrations in genes comprising key proliferation and survival<br />

pathways (RB-E2F, RAS, PI3K) in the majority <strong>of</strong> tumors. Patients with<br />

germline BRCA1/2-mutations have more favorable prognoses than non-<br />

BRCA carriers, and recent work suggests that BRCA2 carriers do better than<br />

BRCA1. We hypothesized that concurrent proliferation pathway aberrations<br />

and BRCA1/2 mutations in tumors might play a role in patient outcome.<br />

Methods: Mutation, copy number, and clinical data for 309 TCGA-pr<strong>of</strong>iled<br />

serous ovarian tumors were downloaded from the MSKCC cBIO web portal.<br />

Each tumor was scored as aberrant for a pathway if any gene (RB: RB1,<br />

CDKN2A, CCND1, CCND2, E2F3, CCNE1; PI3K: PIK3CA, PTEN, AKT1,<br />

AKT2; RAS: KRAS, BRAF, NF1) in that pathway was mutated, amplified, or<br />

deleted. Results: 205 <strong>of</strong> 309 tumors had an aberration in at least one <strong>of</strong><br />

these pathways. The frequency <strong>of</strong> pathway alteration differed significantly<br />

in BRCA1 (82%, 28/34), BRCA2 (52%, 17/33) and BRCA1/2 WT (66%,<br />

160/242) tumors (BRCA1 vs. BRCA2: Fisher’s p� 0.0096). BRCA1<br />

tumors more frequently contained alterations in multiple pathways than<br />

BRCA2 or WT tumors (41% vs. 24% or 25%, respectively). RB-E2F<br />

pathway alteration frequency was significantly different (BRCA1: 56%,<br />

BRCA2: 18%, WT: 43%, p�0.0043), but no significant differences in<br />

PI3K and RAS pathway aberration frequencies (BRCA1%: 41, 38; BRCA2%:<br />

36, 27; WT% 28, 27), respectively, were observed. In agreement with the<br />

previous report, BRCA2 patients had significantly better overall survival<br />

(OS) than either BRCA1 or WT patients (median OS months for BRCA1:<br />

35.9, BRCA2 45.4, BRCA1/2 WT 27.8; p�0.001). Presence <strong>of</strong> pathway<br />

alterations was not significantly associated with OS in BRCA1, BRCA2, or<br />

WT patients in this cohort. Conclusions: These results show a negative<br />

association between BRCA2 mutations and aberrations in key proliferation<br />

and survival pathways. Beyond BRCA1 and BRCA2 genetic mutations, the<br />

elevated frequency <strong>of</strong> pathway alteration in BRCA1 vs. BRCA2 tumors<br />

highlights differences that may be important for patient prognosis as well<br />

as therapy responses.<br />

5076 General Poster Session (Board #22D), Sun, 8:00 AM-12:00 PM<br />

Inflammatory and nutritional markers as predictors <strong>of</strong> longer hospital stay<br />

and suboptimal residual disease (RD) in ovarian cancer (OVCA). Presenting<br />

Author: Michelle Torres, Mayo Clinic, Rochester, MN<br />

Background: Advanced OVCA should be managed aggressively, and extensive<br />

surgery has been the most accepted initial treatment. Medically unfit<br />

patients or those with extensive disease, in which complete cytoreduction<br />

is unlikely, may not benefit from upfront radical surgery, and neoadjuvant<br />

chemotherapy might be an appropriate alternative. Thus, reliable preoperative<br />

indicators <strong>of</strong> surgical outcome are necessary for considering primary<br />

surgery vs. neoadjuvant chemotherapy. Our aim is to determine if Creactive<br />

protein (CRP), IL-6, albumin and Glasgow Prognostic Score (GPS –<br />

score based on CRP and albumin) correlate with overall survival (OS),<br />

length <strong>of</strong> hospital stay (LOS), surgical morbidity, and suboptimal cytoreduction.<br />

Methods: We randomly selected 50 stages III/IV OVCA who underwent<br />

surgery as a primary treatment between July 2002 and June 2009 at Mayo<br />

Clinic with serum albumin levels and frozen serum available. CRP and IL-6<br />

were measured in stored serum. Univariate and multivariate regression<br />

models were fit to evaluate associations with each <strong>of</strong> the outcomes. Results:<br />

Among the 50 patients, the mean age was 67.7 years. 34% had<br />

pretreatment albumin �3.5 g/ml, 22.4% had CRP level �10 mg/l, 26.5%<br />

had IL-6 �24 pg/ml and 45% had abnormal GPS score. At 1, 3 and 5 years<br />

following surgery, the OS was 75.6%, 49.8% and 36.9%, respectively. RD<br />

(0, �1, �1cm; p�0.001) was the only independent predictor <strong>of</strong> OS. Also,<br />

IL-6 (p�0.028) and stage (p�0.046) were independently associated with<br />

LOS, but no inflammatory or nutritional markers were significant associated<br />

with post surgical complications. Stage IV (p�0.019) and elevated CRP<br />

(p�0.044) were independent predictors <strong>of</strong> suboptimal surgery (RD �<br />

1cm). Conclusions: One-third <strong>of</strong> the patients in our series had low serum<br />

albumin at the time <strong>of</strong> the OVCA diagnosis, and at least one-fourth had<br />

elevated inflammatory markers. Advanced stage and elevated inflammatory<br />

markers (CRP and IL-6) were independent predictors <strong>of</strong> longer hospital stay<br />

and suboptimal debulking. These pilot data, if confirmed in a larger<br />

population, may help in the selection <strong>of</strong> candidates for neoadjuvant<br />

chemotherapy.<br />

Gynecologic Cancer<br />

345s<br />

5075 General Poster Session (Board #22C), Sun, 8:00 AM-12:00 PM<br />

Secondary surgery with intraoperative chemohyperthermia in recurrent<br />

ovarian adenocarcinoma. Presenting Author: Jean Marc Bereder, University<br />

Hospital Archet 2, Nice, France<br />

Background: Optimal treatment <strong>of</strong> peritoneal recurrences in ovarian cancer<br />

is debating with second line chemotherapies. We proposed association <strong>of</strong><br />

secondary surgery with heated intraperitoneal per operative chemotherapy<br />

(HIPEC). The aim <strong>of</strong> study is to determine prognostic factors in a single<br />

center cohort. Methods: Retrospective study <strong>of</strong> consecutive 169 patients<br />

with peritoneal recurrence from ovarian cancer were performed to evaluate<br />

HIPEC and to identify prognostic factors. Peritoneal Cancer Index (PCI)<br />

assess tumor load and completeness cytoreductive score (CCS) were used<br />

to give quality <strong>of</strong> resection CCS0 (no visible tumor), CCS1 (persistent<br />

diffuse lesions � 2.5mm), CCS2 (2.5mm �CC2� 25mm) and over CCS3<br />

status. HIPEC is performed with platinum based regimen. Endpoint was<br />

survival. Cox’s regression model was used for multivariate survival analysis<br />

and extending Cox model for modelling survival data. Results: We have<br />

operated on 197 procedures (HIPEC) in 169 patients from 2000 to 2011.<br />

Mean age was 58 years old range [28-75]. Median PCI was 10. After<br />

completion <strong>of</strong> resection, allocation <strong>of</strong> CCS was CCS0�120, CCS1�70,<br />

CCS2 & CCS3 �7. Procedure related mortality was 1% and morbidity 21%,<br />

mean length <strong>of</strong> hospital stay was 17 days range [7-51]. 3 and 5 years<br />

overall survival were respectively 64.7% and 37.4 %. Median survival was<br />

47.6 months and the median disease free survival was 20 months. PCI<br />

�10 (even if complete resection performed) and CCS2&3 were worse<br />

prognostic factors (HR respectively � 2.64 IC 95% [1.29-5.36] and �<br />

3.31 IC 95 % [1.55-7.08]). Modelling <strong>of</strong> these factors, is very strong to<br />

predict risk <strong>of</strong> death over the 2 first years after HIPEC. Conclusions: The<br />

chemo-hyperthermia is a standardized and reproducible feasible method.<br />

Less extensive disease and the quality <strong>of</strong> cytoreduction remain an independent<br />

factor <strong>of</strong> better outcome. To date HIPEC allows to reach the longest<br />

median time survival in peritoneal recurrent ovarian cancer. Modelling<br />

survival data is useful to know the risk <strong>of</strong> dying.<br />

5077 General Poster Session (Board #22E), Sun, 8:00 AM-12:00 PM<br />

Association <strong>of</strong> SNP genotype in ABCB1 with completion <strong>of</strong> intraperitoneal<br />

chemotherapy in ovarian and primary peritoneal cancer. Presenting Author:<br />

Sergio Enderica Gonzalez, Mayo Clinic, Rochester, MN<br />

Background: A standard <strong>of</strong> care (SOC) for the adjuvant treatment <strong>of</strong> ovarian<br />

and primary peritoneal cancer is a platinum-based intravenous (IV) chemotherapy<br />

doublet. Intraperitoneal (IP) chemotherapy is also a SOC, but the<br />

high incidence <strong>of</strong> grade 3/4 adverse events has limited its acceptance<br />

among clinicians. To identify genetic markers for completion <strong>of</strong> IP<br />

chemotherapy, we analyzed SNPs in four genes known to play a role in<br />

metabolism <strong>of</strong> platinum or taxane drugs. Methods: Patients diagnosed with<br />

primary or recurrent stage III or IV epithelial ovarian or primary peritoneal<br />

cancer who had primary or secondary debulking surgery at Mayo Clinic<br />

(Rochester, MN) between January 2007 and February 2009 followed by IP<br />

chemotherapy were included in this study. A cycle was defined as IV<br />

paclitaxel (135mg/m2 ) on day one, IP cisplatin (100mg/ m2 ) on day two,<br />

and IP paclitaxel (60mg/ m2 ) on day eight. With prior consent from the<br />

patient, peripheral blood was obtained before treatment for extraction <strong>of</strong><br />

germline DNA. Using a custom Illumina BeadXpress 96-plex panel, SNPs<br />

in GSTM1 (N�7), ABCB1 (N�57), CYP3A4 (N�7), and CYP2C8 (N�25)<br />

were genotyped. The association between SNPs in these subsets <strong>of</strong> genes<br />

and completion <strong>of</strong> IP chemotherapy was analyzed using linear regression.<br />

Results: Thirty-seven patients were included in this study and 16 (43.2%)<br />

completed IP chemotherapy. Twenty-two out <strong>of</strong> the fifty-seven ABCB1<br />

SNP’s were associated with the number <strong>of</strong> cycles (p�0.15). There were no<br />

significant associations for GSTM1, CYP3A4 and CYP2C8 SNP’s. The<br />

minor A allele at missense SNP rs2229109 in the ABCB1 gene was present<br />

in five (31.2%) who completed treatment and only one (4.8%) who did not<br />

(p�0.007). However, the later was due to catheter complication. There was<br />

no association between rs2229109 and overall or progression-free survival.<br />

Conclusions: Our findings suggest that SNP rs2229109 in ABCB1 gene is<br />

associated with completion <strong>of</strong> IP chemotherapy, and potentially, less<br />

toxicity from chemotherapy. Thus, these patients may be optimal candidates<br />

for IP chemotherapy. Further studies in a larger population are<br />

needed.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


346s Gynecologic Cancer<br />

5078 General Poster Session (Board #22F), Sun, 8:00 AM-12:00 PM<br />

BRCA mutations and outcome in epithelial ovarian cancer (EOC): Experience<br />

in ethnically diverse groups. Presenting Author: Tamar Safra, Department<br />

<strong>of</strong> Oncology, Tel Aviv Sourasky Medical Center, Sackler School <strong>of</strong><br />

Medicine, Tel-Aviv University, Tel Aviv, Israel<br />

Background: EOC patients with BRCA mutations have been reported to have<br />

better prognosis than non-hereditary (NH) matched cases, an advantage<br />

shown especially in the Ashkenazi-Jewish (AJ) population. We have<br />

analyzed our experience in our ethnically diverse patient cohort from NYC,<br />

Israel and Italy. Methods: A retrospective chart review <strong>of</strong> patients diagnosed<br />

with Stage IC-IV EOC between 1995-2008 at the NYU Cancer Institute, Tel<br />

Aviv Sourasky MC and Padova <strong>Clinical</strong> Cancer Centers. Out <strong>of</strong> �700<br />

patients, 183 were tested for BRCA mutations and evaluated. Results:<br />

Median age was 55.5 (range 31–83 years). Out <strong>of</strong> 183, 86 are carriers <strong>of</strong><br />

BRCA1/2 mutations and 97 tested negative, 67 and 19 are carriers <strong>of</strong><br />

BRCA1 and BRCA2 mutation respectively. Carrier frequency in EOC<br />

population is 46% (45/97) in AJ’s and 48% (41/86) in non-AJ’s. AJ<br />

patients had the following BRCA1 mutations: 185delAG (29), 5382insC<br />

(5), unknown (UK) (2) and BRCA2 mutations: 6174delT (8), UK (1).<br />

Non-AJ’s were divided by ethnicities into non-AJ, Caucasian, African-<br />

<strong>American</strong>, Hispanic, Middle Eastern or unknown. Non-AJ Jewish patients<br />

had BRCA1 mutations in 185delAG (7) and BRCA2 in 6174delT (1), UK<br />

(1). Non-Jewish Caucasians exhibited the widest variation <strong>of</strong> mutation<br />

types, with the following BRCA1 sites: 185delAG, K1702X (5223A�T),<br />

E1373X, 3829delT, 185delAT, IVS11�1G�A, 5385insC,<br />

5083del19�stop1670, 1720delAF�stop536, 1806C/T�Igu536Ter, del<br />

ex1a-2, cod1486ex14:4575delAstop1504, 5563G7A;Trp1815stop,<br />

5181delGTT(Val1688del) and UK and the following BRCA2 sites: 6174delT<br />

(2), 5301insA (1), 802delAT (1), cod2960ex22:9106C/t (1), cod68ex3:<br />

432delAstop79 (1), 7408A/T;Arg2394stop (1). One African-<strong>American</strong><br />

patient with BRCA1 at 1294del40, 1 Hispanic BRCA1 at 185delAG. OS<br />

was significantly prolonged for BRCA carriers at 93.6 months versus 63.2<br />

months [95% CI: 44.5-91.7] (p�0.0016) for NH. Conclusions: Our data<br />

reports a wide variety <strong>of</strong> BRCA mutations in an ethnically diverse EOC<br />

population and confirms that BRCA mutations carriers have a better<br />

prognosis with a longer median survival compared to NH population. A<br />

larger cohort might manifest prognostic differences between the different<br />

types <strong>of</strong> mutations.<br />

5080 General Poster Session (Board #22H), Sun, 8:00 AM-12:00 PM<br />

A matched pair analysis <strong>of</strong> intra- and postoperative catumaxomab in<br />

patients with ovarian cancer from a multicenter, single-arm phase II trial<br />

versus a consecutive single-center collective <strong>of</strong> ovarian cancer patients<br />

without immunotherapy. Presenting Author: Klaus Pietzner, Charite University<br />

Medicine <strong>of</strong> Berlin, Berlin, Germany<br />

Background: Advanced ovarian cancer is still connected to high mortality<br />

rates due to intraperitoneal tumor cells that survive radical cytoreductive<br />

surgery as well as adjuvant chemotherapy. These persistent tumor cells<br />

need to be targeted in order to improve survival. Catumaxomab has<br />

demonstrated the ability to kill EpCAM-positive intraperitoneal tumor cells<br />

<strong>of</strong> ovarian cancer patients in studies aiming to controll malignant ascites in<br />

the recurrent setting. This analysis was conducted to investigate the<br />

efficacy <strong>of</strong> intraperitoneal catumaxomab at the timepoint <strong>of</strong> primary<br />

cytoreductive surgery and postoperative period, prior to standard adjuvant<br />

chemotherapy. Methods: Ovarian cancer patients undergoing radical surgery<br />

received one intraoperative (10 �g) followed by four subsequent<br />

intraperitoneal (i.p.) dosages (10, 20, 50 and 150 �g) <strong>of</strong> catumaxomab on<br />

days 7, 10, 13, and 16, respectively. Because <strong>of</strong> the single arm design <strong>of</strong><br />

the study, the patients treated with catumaxomab were compared in a<br />

matched pair analysis to consecutive patients with primary ovarian cancer<br />

who received standard treatment without catumaxomab in a large center, in<br />

order to compare survival. The two main prognostic factors <strong>of</strong> stage and<br />

level <strong>of</strong> tumorreduction were chosen as matching criteria. Results: Of 58<br />

patients screened, 41 were treated with catumaxomab and available for<br />

survival evaluation. Median age was 57 years in the catumaxomab group<br />

and 59 years in the matched-pair control group. The most comon histology<br />

was the serous subtype with 70.7 % in the catumaxomab and 80.5 % <strong>of</strong> the<br />

patients in the control group. The median for overall survival was reached<br />

for the historical consecutive matched-pair control collective, but is not yet<br />

reached for the catumaxomab group. However, 3-year survival data were<br />

available for both groups and showed survival <strong>of</strong> 85.4% (35) in the<br />

catumaxomab group and 63.4% (26) in the matched-pair control group<br />

(p-value: 0.041; HR 2.073) Conclusions: There seems to be a trend to<br />

beneficial 3-year survival in the catumaxomab group, suggesting that a<br />

phase III trial is warranted.<br />

5079 General Poster Session (Board #22G), Sun, 8:00 AM-12:00 PM<br />

Serum IGFBP4 levels in combination with miliary disease: A candidate<br />

predictor <strong>of</strong> ovarian cancer progression-free survival. Presenting Author:<br />

Samantha Cohen, Mt Sinai School <strong>of</strong> Medicine, New York, NY<br />

Background: Insulin-like growth factor binding protein, IGFBP4, was shown<br />

to be highly expressed across all stages <strong>of</strong> epithelial ovarian cancer (EOC)<br />

and serum levels are elevated in EOC. Moreover, IGFBP4 levels are ~3x<br />

greater in women with malignant pelvic masses. We investigated whether<br />

ascites volume and the presence <strong>of</strong> miliary disease in combination with<br />

serum levels <strong>of</strong> IGFBP4 are independent predictors <strong>of</strong> survival. Methods: A<br />

prospective and retrospective analysis was performed. Patients were<br />

enrolled at the time <strong>of</strong> cytoreductive surgery. Ascites volume was either<br />

absent, �500 cc (low), or �� 500 cc (high), and the presence <strong>of</strong> miliary<br />

disease was recorded. The IGFBP4 cut<strong>of</strong>f was 1064.5 ug/ml based upon<br />

previous results. The Kaplan-Meier product limit method was used to<br />

estimate PFS probabilities. The Cox proportional hazards model was used<br />

to estimate hazard ratios (HR) and corresponding 95% CI. Results: 57<br />

cases were included in the analysis <strong>of</strong> ascites volume and miliary disease.<br />

Cytoreductive outcomes were complete gross resection (44.8%), optimal<br />

(��1cm residual disease; 44.8%), and suboptimal ( �1cm residual<br />

disease; 10.3%). Histologic subtypes: papillary serous (n�35; 61.4%),<br />

mucinous (n�15; 26.3%), endometrioid (n�4; 7.0%), and clear cell<br />

(n�3; 5.3%). Stage distribution was 21.1% I/II, and 78.9% III/IV. PFS<br />

was unaffected by ascites volume (p�0.341) or miliary disease. Among<br />

this cohort, 29 had IGFBP4 levels available for a separate analysis.<br />

Patients with high IGFBP4 and miliary disease were 5.5 times as likely to<br />

recur compared with patients with miliary disease and low IGFBP4<br />

(HR�5.55 [0.77, 39.82]), and the statistical significance was borderline<br />

(p�0.088). No statistically significant differences were detected between<br />

rates <strong>of</strong> recurrence among patients with high and low IGFBP4 values in<br />

combination with ascites volume. Conclusions: These exploratory studies<br />

suggest that patients with high IGFBP4 serum levels and miliary disease<br />

were � 5 times as likely to recur compared to women with miliary disease<br />

and low IGFBP4 levels. Future studies examining these variables using a<br />

larger population and examining the biologic basis <strong>of</strong> this relationship are<br />

planned.<br />

5081 General Poster Session (Board #23A), Sun, 8:00 AM-12:00 PM<br />

Progression-free survival (PFS) as a surrogate end point for overall survival<br />

(OS) in first-line treatment <strong>of</strong> advanced epithelial ovarian cancer (EOC).<br />

Presenting Author: Katrin Marie Sjoquist, NHMRC <strong>Clinical</strong> Trials Centre,<br />

University <strong>of</strong> Sydney, Sydney, Australia<br />

Background: The duration and cost <strong>of</strong> clinical trials <strong>of</strong> first-line chemotherapy<br />

in advanced EOC could be reduced if a surrogate endpoint were<br />

used in place <strong>of</strong> OS. The consensus <strong>of</strong> the Gynecological Cancer Intergroup<br />

was that PFS is the preferred primary end point for these trials due to<br />

potential confounding <strong>of</strong> post-progression therapy on OS. We performed a<br />

systematic review to assess the extent to which treatment effect on PFS is<br />

predictive <strong>of</strong> OS in this patient population. Methods: Randomised trials <strong>of</strong><br />

first-line chemotherapy comparing platinum containing regimens in advanced<br />

EOC were identified; trials with non-platinum backbone, maintenance<br />

strategies or biological containing therapies were excluded. Summary<br />

data (hazard ratios (HR), median PFS and OS and the ratios <strong>of</strong> medians<br />

between treatment arms) were extracted. Weighted least-square (WLS) R2 by trial sample size derived using linear regression was used to report<br />

correlation. Results: 15 eligible trials with 19 treatment comparisons were<br />

identified comprising a total <strong>of</strong> 16,598 patients. There was a good<br />

correlation between treatment effect on PFS and OS (correlation <strong>of</strong> HR: r,<br />

0.88, WLS R2 , 0.71; correlation <strong>of</strong> ratios <strong>of</strong> medians: r, 0.84, WLS R2 ,<br />

0.72). Good correlation between treatment effect on PFS and OS was also<br />

observed in various prognostic subgroups (Table). Conclusions: In clinical<br />

trials <strong>of</strong> first-line platinum based chemotherapy without biological agents<br />

in advanced EOC, treatment effect on PFS and OS is highly correlated. PFS<br />

could be considered as a primary end point when evaluating future first line<br />

strategies in advanced EOC.<br />

Prognostic factors across trials<br />

low vs. high split at median Correlation for HR (r) WLS R 2<br />

Stage IV (16%) 0.94 vs. 0.75 0.79 vs. 0.57<br />

Optimal debulking (62.6%) 0.86 vs. 0.95 0.69 vs. 0.83<br />

Age (59 years) 0.93 vs. 0.86 0.77 vs. 0.66<br />

ECOG performance status ECOG 2 and 3 (10.1%) 0.95 vs. 0.89 0.81 vs. 0.76<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


5082^ General Poster Session (Board #23B), Sun, 8:00 AM-12:00 PM<br />

Final results <strong>of</strong> a phase II trial <strong>of</strong> weekly nab-paclitaxel with GM-CSF as<br />

chemoimmunotherapy for platinum-resistant epithelial ovarian cancer.<br />

Presenting Author: Ron E. Swensen, Loma Linda University, Loma Linda,<br />

CA<br />

Background: Ovarian cancer patients with an anti-tumor immune response<br />

have a prolonged survival, suggesting that augmenting anti-tumor immunity<br />

may be therapeutic. We hypothesized that weekly nab paclitaxel (nabP)<br />

followed by GM-CSF may enhance anti tumor immunity and prolong time to<br />

progression (TTP). Methods: Eligible subjects had platinum resistant<br />

ovarian, primary peritoneal or fallopian tube cancer, and an elevated<br />

CA125. Conditional power estimate after 11 subjects showed 22 subjects<br />

had 80% power to show a response rate (RR) �21% if the true study RR is<br />

35%. Study end points were RR and TTP. Progression (DP) was doubling <strong>of</strong><br />

CA125 above the nadir. Complete response (CR) was a decline <strong>of</strong> CA125<br />

below institutional normal. <strong>Part</strong>ial response (PR) was a decline <strong>of</strong> �50%<br />

from baseline. Stable disease (SD) was all other scenarios. Subjects<br />

received nabP, 100mg/m2 days 1,8,15 followed by GM-CSF 250mcg days<br />

16-26 <strong>of</strong> a 28 day cycle until progression or 6 cycles were complete.<br />

Responding subjects received up to 6 more cycles <strong>of</strong> GM-CSF on days<br />

14-28. Results: 21 subjects received at least one dose <strong>of</strong> study medications.<br />

Median age was 61 (30-91) and had a median <strong>of</strong> 3 (1-13) prior<br />

regimens. Among those completing the study, the median TTP was 132<br />

days vs. 272 days on the prior platinum regimen. 9/21 (43%) had a PR and<br />

4/21 (19%) had a CR. Subjects with a response had a median TTP <strong>of</strong> 140<br />

days. Assay <strong>of</strong> serial T-lymphocyte counts against CEA, MUC1, CA125 and<br />

tt and influenza controls are planned. Conclusions: Weekly nabP with<br />

GM-CSF had manageable toxicity and induced a high response rate (62%<br />

by CA125) in patients with platinum resistant ovarian cancer, however this<br />

regimen did not prolong the TTP beyond the TTP observed in the prior<br />

platinum regimen.<br />

5084 General Poster Session (Board #23D), Sun, 8:00 AM-12:00 PM<br />

Prevalence <strong>of</strong> high-risk human papillomavirus in uterine cervical lesions<br />

among older Japanese women. Presenting Author: Kazuhiro Takehara,<br />

National Hospital Organization Kure Medical Center/Chugoku Cancer<br />

Center, Hiroshima, Japan<br />

Background: To estimate the prevalence and genotypes <strong>of</strong> high-risk human<br />

papillomavirus (HPV) among older Japanese women, using liquid-based<br />

cytology (LBC). Methods: ThinPrep LBC specimens were collected from<br />

11,039 Japanese women (age range, 14-98 years). After classifying<br />

cytodiagnosis, specimens were analyzed for HPV DNA using the multiplex<br />

polymerase chain reaction method. Cervical smear specimens from 1,302<br />

women showed positive results. To examine the prevalence <strong>of</strong> HPV in<br />

women defined as negative for intraepithelial lesion or malignancy (NILM),<br />

2,563 samples were randomly selected from the remaining 9,737 women.<br />

Comparisons were made between women �50 years <strong>of</strong> age (older age<br />

group) and women �50 years <strong>of</strong> age (younger age group). Written informed<br />

consent was obtained from all patients. In this study, the high-risk HPV<br />

genotypes encountered were 16, 18, 31, 33, 35, 45, 52, and 58. Results:<br />

In the older age group with abnormal smear findings, HPV genotypes were<br />

detected in 49.7% (148/298), including high-risk HPV genotypes in<br />

40.9% (122/298). In the younger age group with abnormal smear findings,<br />

HPV genotypes were detected in 71.7% (720/1004), including high-risk<br />

HPV genotypes in 58.1% (583/1,004). In NILM, HPV-positive rates were<br />

4.5% (39/873) in the older age group and 11.8% (199/1,690) in the<br />

younger age group. In high-grade squamous intraepithelial lesion (HSIL) or<br />

more severe cytological findings, HPV genotypes <strong>of</strong> each group (older age<br />

group/younger age group) were detected in 61.7%/83.1%, and high-risk<br />

HPV genotypes were detected in 56.4%/74.7% <strong>of</strong> women. In positive<br />

cervical smears, HPV 16 was the most frequently detected (28.5%) in the<br />

younger age group, while HPV 52 (31.3%) and 58 (27.2%) were detected<br />

more frequently than HPV 16 (18.4%) in the older age group. Conclusions:<br />

In Japan, although HPV infection as a cause <strong>of</strong> abnormal cervical cytology<br />

is more frequent among younger age groups than in older age groups,<br />

high-risk HPV infection was more highly associated with older individuals<br />

(older age group/younger age group: abnormal smear findings, 82.4%/<br />

81.0%; HSIL or more severe cytological findings, 91.3%/89.9%). In older<br />

age groups, HPV 52 and 58 were more frequent than HPV 16.<br />

Gynecologic Cancer<br />

347s<br />

5083 General Poster Session (Board #23C), Sun, 8:00 AM-12:00 PM<br />

Prospective multicenter study evaluating the survival <strong>of</strong> patients with<br />

locally advanced cervical cancer (LACC) and negative positron emission<br />

tomography with computerized tomography (PET-CT) in the para-aortic<br />

area undergoing laparoscopic para-aortic (PA) lymphadenectomy before<br />

chemoradiation therapy. Presenting Author: Sebastien Gouy, Institut<br />

Gustave Roussy, Villejuif, France<br />

Background: In three comprehensive cancer centers, patients with LACC<br />

were systematically staged using conventional and PET-CT imaging before<br />

chemoradiotherapy. If patients had no uptake in the PA area, laparoscopic<br />

extraperitoneal PA surgery was then performed to define radiation field<br />

limits more accurately. The aim <strong>of</strong> this study was to evaluate the<br />

therapeutic impact <strong>of</strong> this management. Methods: A prospective multicenter<br />

series <strong>of</strong> 237 patients treated from 2004 to 2011 for LACC with a<br />

negative PET-CT <strong>of</strong> the PA area and undergoing laparoscopic PA lymphadenectomy.<br />

Radiation fields were extended to PA area when PA nodes were<br />

involved. Chemoradiotherapy modalities were homogeneous between Institutions.<br />

Patients with a poor prognosis histologic subtype, peritoneal<br />

carcinomatosis or ovarian metastasis were excluded. Results: <strong>Clinical</strong><br />

stages were IB2 (n�79), IIA (n�10), IIB (n�120), III (n�23), IVA (n�5).<br />

The histologic types were squamous carcinoma (n�197), adenocarcinoma<br />

(n�34) and others (n�6). Twenty-nine (11%) patients had nodal involvement<br />

(false negative PET-CT results): 16 with PA nodal metastasis<br />

measuring � 5 mm and 13 � 5 mm. With a median follow-up <strong>of</strong> 18 (range,<br />

0-67) months, disease-free survival (DFS) at 2 years in patients without<br />

and with PA involvement was respectively 76% (68%-83%) and 61%<br />

(37%-80%)(p�.007). DFS at 2 years in patients without PA involvement<br />

or with PA metastasis measuring � or � 5 mm was respectively 76%<br />

(68%-83%), 89% (57%-98%) and 38% (14%-68%)(p�.0006).<br />

Conclusions: This is the largest series <strong>of</strong> patients reported undergoing such<br />

strategy. We obtained a similar survival rate for patients with PA nodal<br />

metastasis � 5 mm and patients without PA lymph node involvement<br />

suggesting that this strategy is highly efficient in such patients. Conversely,<br />

the survival <strong>of</strong> patients with PA nodal involvement � 5 mm remained poor,<br />

despite no extrapelvic disease at PET-CT imaging in this subgroup. Other<br />

treatment modalities should be evaluated for these patients.<br />

5085 General Poster Session (Board #23E), Sun, 8:00 AM-12:00 PM<br />

Nuclear Y-box binding protein-1 expression as a prognostic marker and<br />

correlation with epidermal growth factor receptor expression in cervical<br />

cancer. Presenting Author: Shin Nishio, Kurume University School <strong>of</strong><br />

Medicine, Kurume, Japan<br />

Background: Y-box binding protein-1 (YB-1) is a member <strong>of</strong> the cold shock<br />

protein family and functions in transcription and translation. Many reports<br />

indicate that YB-1 is highly expressed in tumor cells and is a marker <strong>of</strong><br />

tumor aggressiveness and clinical prognosis. Overexpression <strong>of</strong> epidermal<br />

growth factor receptor (EGFR) has been associated with poor outcomes in<br />

cervical cancer (CC). <strong>Clinical</strong> trials have shown that EGFR inhibitors are<br />

effective against CC (JCO 2011). Nuclear YB-1 expression correlates with<br />

EGFR expression in various types <strong>of</strong> cancer. Methods: Nuclear YB-1<br />

expression was immunohistochemically analyzed in tissue specimens<br />

obtained from 204 CC patients who underwent surgery. Associations <strong>of</strong><br />

nuclear YB-1 expression with clinicopathological factors such as survival<br />

and EGFR (HER1 and HER2) expression were investigated. Results:<br />

Nuclear YB-1 expression was observed in 41 (20%) <strong>of</strong> 204 cases and<br />

correlated with stage, tumor diameter, stromal invasion, and lymph-node<br />

metastasis. Nuclear YB-1 expression also correlated with both HER1<br />

expression (p�0.0114) and HER2 expression (p�0.0053). Kaplan-Meier<br />

survival analysis showed that nuclear YB-1 expression was significantly<br />

associated with poor outcomes in terms <strong>of</strong> progression-free survival<br />

(p�0.0033) and overall survival (p�0.0003). On multivariate analysis,<br />

stromal invasion, parametrial invasion, and nuclear YB-1 expression were<br />

independent predictors <strong>of</strong> survival. Conclusions: Nuclear YB-1 expression is<br />

a prognostic marker and correlates with EGFR expression in CC.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


348s Gynecologic Cancer<br />

5086 General Poster Session (Board #23F), Sun, 8:00 AM-12:00 PM<br />

Concurrent chemoradiotherapy with paclitaxel and cisplatin for adenocarcinoma<br />

<strong>of</strong> the cervix. Presenting Author: Yutaka Nagai, University <strong>of</strong> the<br />

Ryukyus, Okinawa, Japan<br />

Background: Adenocarcinoma <strong>of</strong> the cervix has a worse prognosis than its<br />

squamous counterpart, particularly when cancer cells spread beyond the<br />

uterine cervix. This is derived from the observation that adenocarcinoma is<br />

less sensitive to radiotherapy (RT) and chemotherapy It is unclear whether<br />

cisplatin-based concurrent chemoradiotherapy (CCRT) has the same effect<br />

on adenocarcinoma as on squamous cell carcinoma. Methods: We retrospectively<br />

analyzed 32 patients with stage IIB–IVA cervical adenocarcinoma<br />

who were treated with RT or CCRT. Fourteen patients were treated with RT<br />

from 1983–1996, 8 with CCRT using cisplatin alone (CCRT-P) from<br />

1997–2002, and 10 with CCRT using cisplatin�paclitaxel (CCRT-TP)<br />

after 2003. The patients were treated with external beam radiotherapy, and<br />

low-dose or high-dose rate intracavitary brachytherapy. For CCRT-P, the<br />

patients received 20 mg/m2 cisplatin for 5 days every 3 weeks, and for<br />

CCRT-TP, the patients received 50 mg/m2 cisplatin every 3 weeks and 50<br />

mg/ m2 paclitaxel weekly. Results: A complete response was achieved in<br />

7/14 patients in the RT, 4/8 patients in the CCRT-P, and 9/10 patients in<br />

the CCRT-TP group. Ten <strong>of</strong> the 14 patients in the RT, 7/8 patients in the<br />

CCRT-P, and 2/10 patients in the CCRT-TP group experienced locoregional<br />

recurrence. The 5-year overall survival rate in the RT, CCRT-P, and<br />

CCRT-TP groups was 7.1%, 25.0%, and 74.1%, respectively (p �<br />

0.0094), and their central disease-free survival rate was 21.4%, 12.5%,<br />

and 78.8%, respectively (p�0.0119). The acute toxicities associated with<br />

CCRT-TP are manageable. Regarding late toxicity <strong>of</strong> CCRT-TP, no grade 3/4<br />

adverse effect was observed. Conclusions: The present study demonstrated<br />

that CCRT-TP achieved much better local control for adenocarcinoma <strong>of</strong><br />

the cervix, leading to a decrease in locoregional recurrence. Prospective<br />

trials in larger series <strong>of</strong> patients are urgently needed.<br />

5088 General Poster Session (Board #23H), Sun, 8:00 AM-12:00 PM<br />

The impact <strong>of</strong> racial disparity on outcomes <strong>of</strong> patients with early-stage<br />

uterine endometrioid carcinoma in an equal-access environment. Presenting<br />

Author: Jared R. Robbins, Henry Ford Hospital, Detroit, MI<br />

Background: To determine if racial disparity exists between African <strong>American</strong><br />

(AA) and non-African <strong>American</strong> (NAA) patients with early stage uterine<br />

endometrioid carcinoma who had similar multidisciplinary management.<br />

Methods: Our prospectively-maintained database <strong>of</strong> 1,450 uterine cancer<br />

patients was reviewed for this IRB-approved study. We identified 766<br />

consecutive patients with endometrioid carcinoma 1988 FIGO stages I-II<br />

who underwent hysterectomy between 1987-2009. Patients with nonendometrioid<br />

carcinoma, mixed histologies and those who received preoperative<br />

treatments were excluded. For the purpose <strong>of</strong> data analysis, patients<br />

were divided into two groups; AA and NAA. Recurrence-free survival (RFS),<br />

disease specific (DSS) and overall survival (OS) was calculated from the<br />

date <strong>of</strong> hysterectomy using the Kaplan-Meier method. Cox regression<br />

modeling was used to explore the risks <strong>of</strong> various factors on recurrence.<br />

Results: Median follow-up was 5.1 years. 27% were AA and 73% were NAA.<br />

All patients underwent hysterectomy and oophorectomy. 80% had peritoneal<br />

cytology and 69% underwent lymphadenectomy. AA patients were<br />

more likely to have higher grade tumors, and more lymphovascular space<br />

involvement (LVSI). Although the two groups were balanced in regards to<br />

surgical staging and adjuvant treatment received, the five-year RFS and<br />

DSS were significantly lower in AA compared to NAA patients (91% vs<br />

84%, p�0.030; 95% vs 88%, p�0.011, respectively). Between the two<br />

groups, OS was not significantly different. On multivariate analysis and<br />

after adjusting for other prognostic factors, race (AA vs NAA) was not a<br />

significant predictor <strong>of</strong> outcome. Grade 3 tumors and the presence <strong>of</strong> LVSI<br />

were the only two independent predictors <strong>of</strong> RFS and DSS with p��0.001<br />

and p��0.001, respectively. Conclusions: In this large hospital-based<br />

study, AA race was associated with a higher incidence <strong>of</strong> adverse pathological<br />

features and worse recurrence-free and disease-specific survival.<br />

However, on multivariate analysis race was not an independent prognostic<br />

factor. Further studies are needed to elucidate possible underlying molecular<br />

mechanisms for these poorer outcomes.<br />

5087 General Poster Session (Board #23G), Sun, 8:00 AM-12:00 PM<br />

How is the current clinical management <strong>of</strong> endometrial cancer worldwide?<br />

An international survey by the North-Eastern German <strong>Society</strong> <strong>of</strong> Gynaecological<br />

Oncology (NOGGO). Presenting Author: Robert W. Kraetschell,<br />

Charité University Department <strong>of</strong> Gynecology, Campus Virchow Clinic,<br />

Berlin, Germany<br />

Background: Indication, technique and extent <strong>of</strong> lymph-node-dissection<br />

(LND) in endometrial cancer (EC) remains controversial and is strongly<br />

debated in cancer community. We conducted a national- and at a second<br />

step an international-survey evaluating the current status-quo <strong>of</strong> the<br />

surgical and medical management <strong>of</strong> EC. Methods: A validated 15-itemquestionnaire<br />

regarding surgical and adjuvant procedures <strong>of</strong> EC was sent to<br />

all major gynaecological cancer societies and study groups worldwide. The<br />

questionnaire could also be answered online. Results: In a phase-I-national<br />

trial, the questionnaire was validated on basis <strong>of</strong> 316 German institutions.<br />

On the phase-II-international survey a total <strong>of</strong> 302 questionnaires were<br />

answered from 24 countries, mainly from Japan (38.7%), Spain (8.3%),<br />

Austria (7%), United-Kingdom (6.3%), Italy (6%), USA (4.3%) and<br />

Canada (4%). The vast majority <strong>of</strong> the participating clinics were academic<br />

(62.8%), while 75.2% <strong>of</strong> them belonged to gynaecology. Only 0.7% <strong>of</strong> the<br />

clinics internationally reported never performing LND in EC. 62.3% <strong>of</strong> the<br />

clinics perform both a pelvic and paraaortic lymph node dissection. 59.1%<br />

<strong>of</strong> the participants performed a systematic lymph node dissection with the<br />

intention <strong>of</strong> both adequate staging and for therapeutic value. 15.05% <strong>of</strong><br />

the clinics perform LND up to the common-iliac-arteries, 9.03% up to the<br />

inferior-mesenteric-artery and 70.6% up to the renal-veins. The most<br />

common risk-factors to indicate LND were: high-grading (93%), nonendometrioid-histology<br />

(90.1%), lymphovascular-invasion (55.3%), bloodvessel<br />

invasion (45.4%) and tumor-diameter �2cm (38.4%). For advanced<br />

stage III&IV disease the vast majority (60% and 80%, respectively) <strong>of</strong> the<br />

physicians indicated systemic chemotherapy alone. Conclusions: This study<br />

presents the large variety in clinical management <strong>of</strong> EC worldwide,<br />

underlining so the high need <strong>of</strong> future prospective randomised trials which<br />

will establish standard and evidence based treatment guidelines for ECdisease.<br />

5089 General Poster Session (Board #24A), Sun, 8:00 AM-12:00 PM<br />

Association <strong>of</strong> smoking with pulmonary recurrences among women with<br />

intermediate- to high-risk early-stage endometrial adenocarcinoma. Presenting<br />

Author: Lori Elizabeth Weinberg, Case Comprehensive Cancer Center<br />

and Case Western Reserve University, Cleveland, OH<br />

Background: While a number <strong>of</strong> histologic risk factors have been identified<br />

in endometrial carcinoma, host factors are less well studied. Our aim was to<br />

examine the influence <strong>of</strong> smoking on the risk <strong>of</strong> pulmonary recurrences in<br />

women with early stage intermediate to high-risk endometrioid uterine<br />

cancer (EC). Methods: Women with surgically treated stage I or II EC from<br />

1994 to 2010 were included in this study if they had lymphovascular space<br />

invasion (LVSI), FIGO grade 2 or 3 histology (G2/3), or outer half<br />

myometrial invasion (OI). All demographic, medical and oncologic data<br />

were obtained from chart review. Patients were excluded if their follow-up<br />

period was less than six months or if they had a diagnosis <strong>of</strong> a second<br />

primary invasive cancer. We performed univariate and multivariate logistic<br />

regression analyses and Fisher’s Exact test for two-way analyses <strong>of</strong><br />

categorical variables to identify prognostic factors <strong>of</strong> pulmonary recurrences.<br />

Significance was determined by two-sided tests with ��0.05.<br />

Results: 349 patients were identified to meet histologic criteria, 3% were<br />

excluded. The remaining 337 patients had a median follow-up <strong>of</strong> 60<br />

months (range 6 to 194). The median age <strong>of</strong> this cohort was 68 years and<br />

median BMI was 30.7 kg/m2. 81 patients (24%) had a past or present<br />

history <strong>of</strong> smoking. There were 14 pulmonary recurrences (4.1%), 36<br />

(10.7%) distant recurrences and 58 (17.2%) total recurrences in the<br />

cohort. Smoking (present or past) was significantly associated with pulmonary<br />

recurrences, with an odds ratio <strong>of</strong> 1.09 (95% CI 1.04-1.15;<br />

P�0.0002). In a multivariate analysis adjusting for age, BMI, grade, LVSI,<br />

OI, cervical invasion, and adjuvant therapy, smoking was still a predictor <strong>of</strong><br />

pulmonary recurrences. Conclusions: Smoking is a significant and independent<br />

predictor <strong>of</strong> pulmonary recurrences among women with early stage EC.<br />

Refining our understanding <strong>of</strong> host factors that influence risk may further<br />

allow us to identify those who may benefit most from adjuvant therapies as<br />

well as intensive surveillance.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


5090 General Poster Session (Board #24B), Sun, 8:00 AM-12:00 PM<br />

External validation <strong>of</strong> a nomogram predicting overall survival (OS) <strong>of</strong> women<br />

with uterine leiomyosarcoma (ULMS). Presenting Author: Alexia Iasonos,<br />

Memorial Sloan-Kettering Cancer Center, New York, NY<br />

Background: A nomogram for predicting OS <strong>of</strong> women with ULMS was<br />

developed , given the poor prognostic performance <strong>of</strong> FIGO and AJCC<br />

staging systems. We aimed to determine the validity <strong>of</strong> the ULMS<br />

nomogram using independent, external cohorts. Methods: The ULMS<br />

nomogram incorporates 7 clinical characteristics: age at diagnosis, tumor<br />

size, grade, involvement <strong>of</strong> cervix, locoregional metastases (direct extrauterine<br />

spread, locoregional lymph node), distant metastases, and mitotic<br />

index (per 10 HPF) to predict OS following primary surgery. Two independent<br />

cohorts from sarcoma centers (1 US, 1 Europe) were included.<br />

Eligible women were treated at the institutions (1994-2010) and had<br />

undergone hysterectomy. Women with locally advanced or metastatic<br />

disease who underwent more extensive surgery were included if the primary<br />

tumor (uterus) was resected. Women who previously underwent resection <strong>of</strong><br />

the primary tumor or recurrences at other institutions were included if they<br />

received followup care at 1 <strong>of</strong> the centers. Women who presented with<br />

unresectable disease who never underwent surgery and those with insufficient<br />

information on any <strong>of</strong> the nomogram variables were excluded. Results:<br />

187 women with ULMS were identified who met the above criteria (median<br />

age 51 yrs, median tumor size 9 cm, median mitotic index 20). Tumors<br />

were generally high grade (88%), FIGO stage I-II (61%) without cervical<br />

involvement (93%) and without locoregional (77%) or distant metastases<br />

(83%). Median and 5-yr OS rates were 4.5 (95% CI 3.2-5.3) yrs and 46%,<br />

respectively; 65 women (35%) were alive at last follow up. The nomogram<br />

concordance index was 0.67(SE�0.02) which was as high as the concordance<br />

index from the initial cohort used for nomogram development. The<br />

concordance between actual OS and nomogram predictions suggests<br />

excellent calibration <strong>of</strong> the nomogram in the validation cohort. Predictions<br />

were within 1% <strong>of</strong> actual 5-yr OS rates, except for the patients with a 5-yr<br />

OS rate <strong>of</strong> greater than 0.68. Conclusions: The ULMS nomogram was<br />

externally validated and can be generalized to independent cohorts. The<br />

nomogram provides a more individualized and accurate estimation <strong>of</strong> OS <strong>of</strong><br />

women diagnosed with ULMS following primary surgery.<br />

5092 General Poster Session (Board #24D), Sun, 8:00 AM-12:00 PM<br />

Prognosic value <strong>of</strong> lower uterine segment involvement in high-grade<br />

endometrial cancers. Presenting Author: Paola A. Gehrig, Lineberger<br />

Comprehensive Cancer Center, University <strong>of</strong> North Carolina at Chapel Hill,<br />

Chapel Hill, NC<br />

Background: To determine the relationship <strong>of</strong> lower uterine segment (LUS)<br />

involvement and clinical-pathologic outcomes in high grade endometrial<br />

cancers (EC). Although LUS and prognosis has been previously reported in<br />

the literature, the results have been conflicting and limited to early stage<br />

cases without focus upon the highest risk histologies. Methods: A singleinstitution<br />

retrospective cohort analysis <strong>of</strong> all grade 3 EC from Jan 2005-<br />

Sept 2010 was performed. <strong>Clinical</strong>-pathologic data were abstracted. LUS<br />

status was determined based on permanent-section pathology. Statistical<br />

analyses were performed using univariate and bivariate analyses with<br />

t-tests, X2, and log-rank tests. Multivariate regressions were performed by<br />

cox modeling. Two sided p-values�0.05 were considered significant.<br />

Results: Of 329 cases, 52% were LUS�. Mean age was 66.1(SD 10.8) and<br />

BMI 31.7(SD 8.3). The majority were Caucasian (63.2%) and 30.1% were<br />

African-<strong>American</strong> (AA). Most women (80.2%) were overweight or obese,<br />

58.7% had hypertension, and 22.5% were diabetic. Most women had stage<br />

I disease (54.8%), but 8.6% had stage II disease, and 36.6% had stage<br />

III-IV disease. Histologic subtypes included 32.2% endometrioid, 47.4%<br />

serous/clear cell, and 17% carcinosarcoma. Thirty-nine percent had �<br />

50% myometrial invasion (MI) and 43.2% had LVSI. Most <strong>of</strong> the women<br />

(80.8%) underwent retroperitoneal node dissection (77.9% pelvic, 70.0%<br />

periaortic). Mean follow-up time was 24.5 months (range 0.13, 73.3). Age,<br />

HTN, and DM did not differ by LUS status. Statistically significant factors<br />

associated with LUS positivity included race AA (38.3 v 26.5%), obesity<br />

(57.5 v 46.7%), serous/clear cell (65.7 v 53.8%), LVSI (56.2 v 30.5%),<br />

deep MI (52.1 v 25.3%), and positive nodes (42.4 v 12.7%). LUS� was<br />

significantly associated with an increased rate <strong>of</strong> recurrence (HR 2.3, CI<br />

1.16-4.47, p �0.02) after adjusting for obesity, deep MI, LVSI, nodal<br />

status, stage, serous/clear cell histology, and adjuvant therapy. Conclusions:<br />

Lower uterine segment was independently associated with an increased<br />

rate <strong>of</strong> recurrence in high grade EC. This should be confirmed in<br />

prospective endometrial trials to see if this remains an independent<br />

predictor <strong>of</strong> recurrence.<br />

Gynecologic Cancer<br />

349s<br />

5091 General Poster Session (Board #24C), Sun, 8:00 AM-12:00 PM<br />

Pair box (PAX8) protein to predict disease recurrence and its association<br />

with outcome in women with endometrial cancer: A retrospective study <strong>of</strong><br />

229 patients. Presenting Author: Damanzoopinder Samrao, USC, Los<br />

Angeles, CA<br />

Background: Pax-8 is a member <strong>of</strong> the pair-box (PAX) family <strong>of</strong> transcription<br />

factor genes and it has been found to be overexpressed in numerous cancer<br />

cell lines including ovarian and endometrial. Possibly, inhibition <strong>of</strong> Pax8<br />

activity may even have an impact on cancer treatment. However the role <strong>of</strong><br />

Pax8 in human endometrial cancer has not yet been explored. Thus, the<br />

aim <strong>of</strong> this study is to determine its predictive value in disease outcome <strong>of</strong><br />

endometrial cancer. Methods: 229 patients with available clinical data and<br />

paraffin-embedded tissue were available for review and analysis. The<br />

clinical parameters used for modeling were age, histologic subtypes,<br />

myometrial depth <strong>of</strong> invasion, lympho-vascular invasion (LVI), FIGO grade,<br />

lymph nodes positive, recurrence, disease status, recurrence time and<br />

survival time. To test the association between Pax8 and the clinical<br />

parameters, Fisher’s exact test was performed. For survival analysis,<br />

Kaplan-Meier method was performed. Results: We found a strong association<br />

between PAX8� and high tumor grade (p�0.002), LVI � (p�0.018),<br />

and type II tumor subtype. Patients with tumor expressing Pax8 were more<br />

likely to present with shorter OS and DFS p� 0.00096 and p�0.018<br />

respectively. There was an association <strong>of</strong> PAX8 with OS (p�0.01486) with<br />

5-years OS probability <strong>of</strong> 80.04% for patients with Pax8- and 55.9% for<br />

patients with Pax8�. There was also an association <strong>of</strong> PAX8 and DFS<br />

probability (p�0.02028) with 5-years DFS probability was <strong>of</strong> 72.12% for<br />

patients with Pax8- versus 49.88% for patients with Pax8� expression.<br />

Conclusions: Pax8 is a reliable marker in endometrial cancer and its<br />

overexpression can predict poor outcome.<br />

5093 General Poster Session (Board #24E), Sun, 8:00 AM-12:00 PM<br />

Neoadjuvant chemotherapy plus radical surgery followed by chemotherapy<br />

in locally advanced cervical cancer. Presenting Author: Roberto Angioli,<br />

Department <strong>of</strong> Obstetrics and Gynecology, Campus Bio-Medico University<br />

<strong>of</strong> Rome, Rome, Italy<br />

Background: The aim <strong>of</strong> this study is to evaluate the efficacy, in terms <strong>of</strong><br />

overall survival and progression free survival, and safety <strong>of</strong> adjuvant<br />

chemotherapy after neoadjuvant chemotherapy followed by radical surgery<br />

both in patients with and without node metastases. Methods: Between June<br />

2000 to May 2007, all patients with diagnosis <strong>of</strong> locally advanced cervical<br />

cancer referred to the Division <strong>of</strong> Gynecologic Oncology <strong>of</strong> the University<br />

Campus Bio-Medico <strong>of</strong> Rome were elegible for this protocol.All enrolled<br />

patients received 3 cycles <strong>of</strong> platinum-based chemotherapy every 3 weeks<br />

according to the scheme cisplatin 100 mg/mq and paclitaxel 175 mg/mq.<br />

After neoadjuvant chemotherapy all patients with stable or progression to<br />

treatment were excluded from the protocol, all other were submitted<br />

classical radical hysterectomy and bilateral systematic pelvic lymph node<br />

dissection, and after to adjuvant treatment with 6 cycles <strong>of</strong> platinum based<br />

chemotherapy with cisplatin 100 mg/mq and paclitaxel 175 mg/mq.<br />

Results: 110 patients with local advanced cervical cancer received the<br />

treatment with neoadjuvant chemotherapy followed by radical surgery and<br />

adjuvant chemotherapy.Our study focused on clinical and operative data ,<br />

in terms <strong>of</strong> overall survival and disease free survival at 5 and 3 years. 5-year<br />

OS <strong>of</strong> our series was 78% at five years and 86% at 3-years, with<br />

encouraging results also in subgroup with and without node mestastases.<br />

Conclusions: The adjuvant chemotherapy regimen after neoadjuvant chemotherapy<br />

and radical surgery rappresents a valid treatment option for<br />

patients with locally advanced cervical cancer without lymph node involvement,<br />

both in terms <strong>of</strong> overall survival than in terms <strong>of</strong> disease-free interval,<br />

the results have also confirmed the validity <strong>of</strong> this approach in lymph node<br />

metastases, with a complication rate lower than the standard radiochemotherapy<br />

regime.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


350s Gynecologic Cancer<br />

5094 General Poster Session (Board #24F), Sun, 8:00 AM-12:00 PM<br />

The novel biomarker HE4 versus CA125 in detecting endometrial cancer: A<br />

case control prospective study. Presenting Author: Francesco Plotti,<br />

Campus Bio-Medico <strong>of</strong> Rome, Rome, Italy<br />

Background: In endometrial cancer, there are no markers routinely used in<br />

clinical practice. This study prospectively investigates the sensitivity and<br />

specificity <strong>of</strong> new marker HE4 in detection <strong>of</strong> endometrial cancer. Methods:<br />

Serum samples were prospectively obtained 24 hours before surgery from<br />

25 patients with endometrial cancer and from 25 patients with uterine<br />

benign pathology, operated from January 2011 to October 2011 at<br />

University Campus Bio-Medico <strong>of</strong> Rome. Preoperative CA125 levels were<br />

evaluated by a one-step “sandwich” radioimmunoassay. HE4 levels were<br />

determined using the HE4 enzymatic immune assay. The CA125 normal<br />

value is considered less than 35 U/mL. Two HE4 cut-<strong>of</strong>f are considered:<br />

less than 70 pmol/L and less than 150 pmol/L. The specificity analysis was<br />

performed using the parametric T-Test for comparing the HE4 series and<br />

the Mann-Whitney test for the CA125 series. The level <strong>of</strong> statistical<br />

significance is set at p � 0.05. Results: The sensitivity <strong>of</strong> CA125 in<br />

detecting endometrial cancer is 16% whereas the sensitivity <strong>of</strong> HE4 is 48%<br />

and 28 % for 70 pmol/L and 150 pmol/L cut-<strong>of</strong>f respectively. The<br />

specificity <strong>of</strong> HE4 is 100% (positive predictive value � 100%, negative<br />

predictive value � 65.79% and 58.14% considering the two HE4 cut-<strong>of</strong>f,<br />

respectively), whereas the CA125 specificity is 72 % (positive predictive<br />

value � 36.36%, negative predictive value � 46.15%) in detection <strong>of</strong><br />

endometrial cancer. Conclusions: HE4 has a good sensitivity and a<br />

specificity <strong>of</strong> 100% in detection <strong>of</strong> endometrial cancer and may be useful<br />

for detecting early stage endometrial cancer. In particular the HE4 at<br />

cut-<strong>of</strong>f <strong>of</strong> 70 pmol/L yields the best sensitivity and specificity.<br />

5096 General Poster Session (Board #24H), Sun, 8:00 AM-12:00 PM<br />

Phase II trial on cisplatin-doxorubicin hydrochloride-paclitaxel (TAP) combination<br />

as neoadjuvant chemotherapy (NACT) for locally advanced cervical<br />

adenocarcinoma (LACA). Presenting Author: Francesco Raspagliesi,<br />

National Cancer Institute <strong>of</strong> Milan, Milan, Italy<br />

Background: NACT followed by surgery is a different therapeutic approach<br />

to locally advanced cervical cancer that seems to <strong>of</strong>fer specific advantages<br />

over chemoradiation such as potential activity against distant micrometastases,<br />

a debulking effect improving subsequent surgical outcome, less<br />

toxicity, and an easier management <strong>of</strong> salvage therapy. This phase II trial<br />

was designed to evaluate the toxicity and activity <strong>of</strong> NACT with TAP in<br />

patients with LACA. Methods: Patients (pts) with FIGO stage IB2 (9 pts)- II<br />

(21 pts) uterine adenocarcinoma were treated with cisplatin 70 mg/mq,<br />

doxorubicin hydrochloride 45 mg/mq and paclitaxel 150 mg/mq day 1<br />

every 21 days for 3 cycles. Eight pts (26.6%) presented with positive<br />

lymphnodes ( �LY) at diagnosis (5 pelvic and 3 pelvic�paraortic). After the<br />

last cycle <strong>of</strong> chemotherapy patients underwent radical surgery with lymphnode<br />

dissection. <strong>Clinical</strong> responses to chemotherapy was evaluated<br />

according to Recist criteria. Pathological response was classified as no<br />

residual tumor (pCR), residual disease with �3 mm stromal invasion (pR1)<br />

or residual disease with �3 mm stromal invasion (pR2). Due to the slow<br />

accrual the trial was closed before the target population <strong>of</strong> 45 pts was<br />

reached. Results: Between 2003 to 2010, 30 women were enrolled. Two<br />

complete clinical responses (6.7%), 21 partial responses (70%) and 7<br />

stabilizations <strong>of</strong> disease (23.3%) were registered. All the patients underwent<br />

radical surgery and were assessable for pathologic responses. Five<br />

patients (16.6%) achieved a pCR , 8 (26.6 %) a pR1 response; 17 patients<br />

(56.7%) a pR2 response. At pathological assessment 10 pts (33.3%)<br />

presented with �LY (7 pelvic, 1 paraortic, 2 pelvic�paraortic). At a median<br />

follow up <strong>of</strong> 45 months progression free survival and overall survival were<br />

37 months (95% CI 1-98) and 45 months (95% CI 9-101�) respectively.<br />

Hematologic toxicity was the most relevant side effect with 13 pts (43 %)<br />

reporting grade 3-4 neutropenia. Conclusions: The TAP combination seems<br />

to be feasible with an acceptable toxicity pr<strong>of</strong>ile and a promising response<br />

rate for the treatment <strong>of</strong> LACA. The effect <strong>of</strong> NACT on lymphnodes status<br />

warrant further investigation.<br />

5095 General Poster Session (Board #24G), Sun, 8:00 AM-12:00 PM<br />

Association <strong>of</strong> adjuvant therapy in early-stage low-risk endometrial cancer<br />

with increased mortality: A statewide cancer registry analysis. Presenting<br />

Author: Michael R. Milam, University <strong>of</strong> Louisville, Louisville, KY<br />

Background: National Comprehensive Cancer Network (NCCN) guidelines<br />

state that patients with early stage low risk endometrial cancer (defined<br />

with 2009 criteria as stage IA endometrioid endometrial cancer) may be<br />

managed with observation with consideration <strong>of</strong> adjuvant therapy.The<br />

premise <strong>of</strong> this study is to review the patterns <strong>of</strong> care <strong>of</strong> those patients who<br />

received adjuvant therapy and its impact on survival. Methods: This is a<br />

retrospective cohort analysis <strong>of</strong> 1044 women from 2004-2008 in the<br />

Kentucky Cancer Registry (KCR) one <strong>of</strong> the affiliates utilized in the<br />

Surveillance, Epidemiology and End Results (SEER) Program database.<br />

Inclusion criteria for the patients in this analysis were those women with<br />

2009 Stage IA uterine cancer <strong>of</strong> endometrioid histology, moderate and well<br />

differentiated tumor grade, who received definitive primary surgery. Adjuvant<br />

therapy was defined as any postoperative radiotherapy and/or chemotherapy<br />

after definitive surgical treatment. Patients with adjuvant therapy<br />

after surgery (AT) were compared to those patients who underwent surgery<br />

only (SO). Chi-square tests were used to identify associations between type<br />

<strong>of</strong> treatment and clinical/demographic factors. K-M plots and Cox regression<br />

models were used to examine survival between the two treatment<br />

groups. Results: 5.3% (55/1044) <strong>of</strong> patients with early stage low risk<br />

endometrial cancer were treated with AT compared to 94.7% (989/1044)<br />

<strong>of</strong> SO patients.No statistical differences in mean age, race, tumor size,<br />

smoking status, insurance status, lymph node dissection and gynecologic<br />

oncology care were found among the AT or SO groups. Five year survival was<br />

significantly better in the SO cohort compared to the AT cohort (92% alive<br />

at 5 years for SO vs. 66% alive at 5 years; p�0.0001). Controlling for other<br />

confounders in the multivariate Cox regression analysis, SO patients had<br />

substantially less risk for death compared to the AT groups (HR: 0.21;<br />

95%CI 0.12-0.38; p�0.0001). Conclusions: In this statewide cancer<br />

registry analysis, adjuvant therapy after surgery in early stage low risk<br />

endometrial cancer patients is uncommon and is associated with an<br />

increased risk <strong>of</strong> mortality.<br />

5097 General Poster Session (Board #25A), Sun, 8:00 AM-12:00 PM<br />

Analysis <strong>of</strong> somatic mutation in miRNAs and miRNA regulation-related<br />

genes in gynecological cancers: New frameshift mutation <strong>of</strong> TNRC6A found<br />

in endometrial cancer. Presenting Author: Qi Mei, Department <strong>of</strong> Oncology,<br />

Tongji Hospital, Tongji Medical College, Huazhong University <strong>of</strong> Science<br />

and Technology, Wuhan, China<br />

Background: Micro RNAs (miRNAs) are small non-coding RNAs which plays<br />

an important role in tumorigenesis and mutation <strong>of</strong> a miRNA gene or<br />

miRNA regulation-related gene may cause butterfly effect on alteration <strong>of</strong><br />

global gene expression.In order to investigate this hypothesis,high resolution<br />

melting analysis (HRM) and denaturing high-performance liquid<br />

chromatography (DHPLC) were used to screen 20 miRNA genes and 6<br />

miRNA regulation-related genes for mutations in gynecologic cancers.<br />

Methods: In this study,182 DNA samples from breast cancer tumors,42<br />

from ovrian cancers and 28 from endometrial cancers were obtained.18<br />

human cancer cell lines were also included.We used HRM and/or DHPLC to<br />

screen mutations <strong>of</strong> these genes in human cancer cell lines and tumor<br />

tissues. All samples with curves that differed in shape compared to<br />

wild-type were directly sequenced for confirmation.Secondary structures<br />

for the wild-type and variant primary precursor <strong>of</strong> miRNA sequences<br />

obtained from sequencing were predicted using RNAfold s<strong>of</strong>tware.Western<br />

blot and immunohistochemistry were used to evaluate the related protein<br />

change in cancer cell line and tumor tissues. Results: Polymorphisms <strong>of</strong><br />

these genes were very frequently observed in gynecologic cancers.Two rare<br />

variants were found in the primary precursor sequences <strong>of</strong> mir-200c and<br />

mir-142 gene.Moreover,a novel frameshift mutation( c.3547_3548insA)<br />

in TNRC6A gene was found in two endometrial cacner cell lines (RL95-2<br />

and Hec-1A) and endometrial cancers (1/28).Western blot and immunohistochemistry<br />

showed loss <strong>of</strong> expression <strong>of</strong> TNRC6A protein in the endometrial<br />

cacner cell lines and endometrial cancers with this mutation.<br />

Conclusions: Two rare novel variants in 20 miRNA genes and a novel<br />

frameshift mutation in TNRC6A gene were identified in gynecologic<br />

cancers.Oue data indicate that mutations in TNRC6A gene may contribute<br />

to the cancer development and might be a potential new therapeutic target<br />

in endometrial cacner.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


5098 General Poster Session (Board #25B), Sun, 8:00 AM-12:00 PM<br />

Clinicopathologic characteristics <strong>of</strong> endometrial cancer in Lynch syndrome.<br />

Presenting Author: Fabrice Lecuru, European Hospital George<br />

Pompidou, Paris, France<br />

Background: Poor data exist on clinico-pathological description <strong>of</strong> endometrial<br />

cancer (EC) in Lynch syndrome (LS) compared with sporadic ones. To<br />

evaluate the clinico-pathological findings <strong>of</strong> Lynch-related EC to establish<br />

histological criteria to discriminate familial and sporadic EC and to decide<br />

the optimal management <strong>of</strong> patients. Methods: Retrospective study <strong>of</strong><br />

prospective cohort <strong>of</strong> patients with LS in our institution from 1999 to<br />

2011. We identified and described all cases <strong>of</strong> endometrial cancers.<br />

Management and follow-up were detailed. Results: Of a cohort <strong>of</strong> 126<br />

patients with LS, 10 women developed endometrial carcinoma. The<br />

median age at diagnosis was 51 years (41-58). Five patients had an<br />

identified mutation (2 hMLH1, 2 hMSH2 and 1 hMSH6). In 9 cases, EC<br />

was the first Lynch-related tumor to occur. No patient developed ovarian<br />

cancer. All, except 2 patients (1 serous carcinoma and 1 clear cell<br />

carcinoma), had endometrioid adenocarcinoma (80%). Tumor grade was<br />

grade 1 in 3 patients, grade 2 in 5 and grade 3 in 2 patients. Forty per cent<br />

<strong>of</strong> patients had lymphovascular space involvement (LVSI). The FIGO stages<br />

were as follows: stage IA (n�7), stage IB (n�2) and stage IIIC (n�1). Four<br />

in ten patients had tumor located in the lower uterine segment. With a<br />

median follow-up <strong>of</strong> 14 months (range9–40months), recurrence occurred<br />

in one patient with a stage IB grade 2 endometrioid adenocarcinoma with<br />

LVSI. Conclusions: EC in LS is characterized by early age at onset,<br />

localization in lower uterine segment, and high rate <strong>of</strong> LVSI. Other data on<br />

histology and survival do not differ from sporadic cancers. These results<br />

suggest that we can consider conservative treatment in patients with good<br />

prognosis tumors. Further studies are required.<br />

5100 General Poster Session (Board #26A), Sun, 8:00 AM-12:00 PM<br />

The effects <strong>of</strong> age on treatment and outcomes in women with stage IB-IIB<br />

cervical cancer. Presenting Author: Dario R. Roque, Women and Infants<br />

Hospital, Providence, RI<br />

Background: Advanced age may affect the treatment choice and subsequent<br />

outcome in elderly patients with cervical cancer. Given the potential<br />

for cure with either surgery or chemoradiation in early stage disease, we<br />

aimed to determine whether a patient’s age influenced the treatment<br />

received and the outcome. Methods: Our retrospective cohort identified a<br />

total <strong>of</strong> 303 patients diagnosed with Stage IB1 through IIB cervical<br />

carcinoma who were treated at our institution between 2000 and 2010.<br />

The eligible patients were divided into two groups based on age at the time<br />

<strong>of</strong> diagnosis: �65 and � 65 years. Adjusted odd ratios were calculated to<br />

determine variables associated with treatment received (chemoradiation or<br />

surgery). Single and multivariate Cox proportional hazards modeling were<br />

used to estimate hazard ratios for variables associated with disease specific<br />

survival. Results: Of the patients meeting inclusion criteria, 253 were �65<br />

years and 50 were � 65 years. The distribution <strong>of</strong> tumor histology, stage<br />

and grade was not different between the two groups. After adjusting for<br />

histology, stage and a validated comorbidity score, the odds ratio <strong>of</strong><br />

receiving chemoradiation vs. surgery for the cohort � 65 years was 1.69<br />

(OR 95% CI: 0.68-4.17). There was no significant difference in the type <strong>of</strong><br />

primary treatment received between the two groups (P � 0.16). Persistent<br />

disease was seen in 46 (18%) <strong>of</strong> the younger patients and in 19 (38%) <strong>of</strong><br />

the older patients (P � 0.02). In the elderly cohort the treatment received<br />

did not influence disease-specific or all-cause mortality. However, compared<br />

to women under 65, older women treated surgically had increased<br />

disease specific (HR 3.18, 95% CI: 0.98-10.3) and all-cause mortality<br />

(HR 6.53, 95% CI: 2.57-16.6). Conclusions: Age does not appear to be a<br />

factor influencing the treatment received by patients with Stage IB1-IIB<br />

cervical cancer. The type <strong>of</strong> treatment received does not seem to affect<br />

disease-specific mortality among older versus younger women. However,<br />

surgery was associated with a 6.5-fold increased risk <strong>of</strong> all cause mortality<br />

among older women when compared to women under 65 years.<br />

Gynecologic Cancer<br />

351s<br />

5099 General Poster Session (Board #25C), Sun, 8:00 AM-12:00 PM<br />

Prognostic factors and treatment-related outcomes in patients with uterine<br />

serous cancer (USC). Presenting Author: Maria deLeon, Yale University,<br />

New Haven, CT<br />

Background: USC an uncommon (10%) but aggressive form <strong>of</strong> uterine<br />

cancer, causes 50% <strong>of</strong> uterine cancer deaths. The purpose <strong>of</strong> this study is<br />

to report a large single-institution experience with USC investigating the<br />

effects <strong>of</strong> clinicopathological parameters and treatment on overall (OS) and<br />

disease-free survival (DFS). Methods: Records from our institution were<br />

reviewed from 1987-2009. All 334 USC patients (pts) identified were<br />

surgically staged. Postoperative treatments used were: (1) observation<br />

(OBS, n�33); (2) platinum-based chemotherapy (PCH, n�78); (3) whole<br />

pelvis radiation therapy (WPRT, n�16); (4) PCH and vaginal apex brachytherapy<br />

(PCHR, n�165); (5) vaginal apex brachytherapy (VAB, n�35). The<br />

cancer stages were 121 pts IA (36.2%), 36 IB (10.8%), 27 II (8.1%), 39<br />

IIIA (11.7%), 2 IIIB (0.6%), 32 IIIC1 (9.6%), 9 IIIC2 (2.7%), 28 IVA<br />

(8.4%) and 40 IVB (12%). Results: The 5-year OS for stage IA/IB, II,<br />

IIIA/IIIB, IIIC1/IIIC2, IVA/IVB were 82%, 70%, 64%, 36%, and 9%,<br />

respectively. The 5-year DFS for stage IA/IB, II, IIIA/IIIB, IIIC1/IIIC2,<br />

IVA/IVB were 82%, 68%, 56%, 24%, 6%, respectively. Advanced stage<br />

was associated with significantly shorter OS and DFS (p �0.01). The 5-year<br />

OS for stage IA/IB disease was 94% for pts who received PCHR, 90% for<br />

OBS, 75% for PCH, 65% for VAB and 0% for WPRT. The 5-year DFS for<br />

PCHR, OBS, PCH, VAB and WPRT for Stage IA/IB were 89%, 82%, 86%,<br />

72% and 0% respectively. For stage II-IVB pts, the 5-year OS was 51% for<br />

PCHR, 0% for OBS, 23% for PCH and 20% for WPRT. The 5-year DFS for<br />

stage II-IVB with PCHR, OBS, PCH and WPRT were 42%, 0%, 17% and<br />

13% respectively. Older age pts had worse survival, (p�0.01). Race and<br />

BMI did not impact survival. Incomplete surgical debulking (p�0.01),<br />

depth <strong>of</strong> myometrial invasion �50% (p�0.01) and lymph node metastasis<br />

(p�0.01) were all associated with worse prognosis. Conclusions: PCHR<br />

overall exhibited better OS and DFS regardless <strong>of</strong> disease stage. Age,<br />

incomplete surgical debulking, depth <strong>of</strong> myometrial invasion and lymph<br />

node involvement were independent prognostic factors in USC patients in<br />

this study.<br />

5101 General Poster Session (Board #26B), Sun, 8:00 AM-12:00 PM<br />

Phase II trial <strong>of</strong> topotecan, cisplatin, and bevacizumab for recurrent or<br />

persistent cervical cancer. Presenting Author: Israel Zighelboim, Washington<br />

University School <strong>of</strong> Medicine and Siteman Cancer Center, St. Louis,<br />

MO<br />

Background: The prognosis associated with recurrent or persistent cervical<br />

cancer is exceedingly poor. GOG-179 demonstrated a survival benefit with<br />

the combination <strong>of</strong> cisplatin and topotecan compared to single-agent<br />

cisplatin, with the former showing median PFS <strong>of</strong> 4.6 months, median OS<br />

<strong>of</strong> 9.4 months, and a 27% objective response rate. The role <strong>of</strong> angiogenesis<br />

in cervical carcinogenesis and progression has been well documented. We<br />

evaluated the activity and safety <strong>of</strong> the combination <strong>of</strong> topotecan, cisplatin<br />

and bevacizumab in patients with incurable carcinoma <strong>of</strong> the cervix.<br />

Methods: Patients with histologically proven measurable recurrent or<br />

persistent cervical carcinoma not amenable to curative intent treatment<br />

were eligible. No prior chemotherapy for recurrence was allowed. Cisplatin<br />

50 mg/m2 day 1, topotecan 0.75 mg/m2 days 1, 2 and 3 and bevacizumab<br />

15 mg/kg day 1 were prescribed in a 21-day cycle. Cytokine support was<br />

allowed at physician discretion. The primary endpoint was 6-month PFS.<br />

Additionally, objective clinical response and toxicity were evaluated.<br />

Accrual goal (N�27) was based on a 50% improvement goal in 6-month<br />

PFS in relation to GOG-179 (40% to 60%), with a one-sided 0.10<br />

significance and 80% power. Results: 27 eligible patients received a<br />

median <strong>of</strong> 3 treatment cycles (range, 1-19). All patients received radiotherapy<br />

as part <strong>of</strong> their first line treatment. Median follow-up was 8.5<br />

months (1.2-32.9). The 6-month PFS was 59% (95%CI: 38.0-74.7).<br />

Among 26 RECIST-evaluable patients, objective response rates were (%;<br />

95%CI): 1 CR (4%; 1-19.6), 7 PR (27%; 11.6-47.8), 11 SD (42%;<br />

23.4-63.1) and 7 PD (27%; 11.6-47.8). Median OS was 9.8 months<br />

(95%CI: 7.7-20.6) and median PFS was 7.1 months (95%CI: 2.0-12.1).<br />

Grade 3-4 hematologic toxicity occurred in 96% <strong>of</strong> patients (thrombocytopenia<br />

93% leukopenia 70%, anemia 70%, neutropenia 59%). Other grade<br />

3-4 toxicities were also common (metabolic 48%, pain 37%, genitourinary<br />

30%, constitutional 22% and gastrointestinal 19%). Conclusions: The<br />

addition <strong>of</strong> bevacizumab totopotecan and cisplatin results in a highly active<br />

but toxic regimen. Future efforts should focus on identification <strong>of</strong> predictive<br />

biomarkers and treatment modifications to minimize toxicity.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


352s Gynecologic Cancer<br />

5102 General Poster Session (Board #26C), Sun, 8:00 AM-12:00 PM<br />

Phase II trial <strong>of</strong> robotic stereotactic radiosurgery (SBRT) in patients with<br />

recurrent gynecologic malignancies. Presenting Author: Charles Kunos,<br />

Case Comprehensive Cancer Center and Case Western Reserve University,<br />

Cleveland, OH<br />

Background: Ablative radiation dose delivered by a robotic SBRT platform<br />

has shown progression-free survival benefit in two limited case series<br />

among patients with recurrent gynecological malignancies. The therapeutic<br />

impact <strong>of</strong> SBRT on disease progression (PD) was evaluated in the recurrent<br />

setting in this phase II trial. Methods: Fifty patients with recurrent and<br />

measurable gynecologic malignancy were treated with SBRT. The cohort<br />

included patients with recurrent ovarian (n �25), endometrial (n �14),<br />

cervical (n �9), or vulvar (n �2) cancer, 1 prior chemotherapy or radiation<br />

regimen, and GOG performance status 0, 1, 2. Patients underwent<br />

image-guided SBRT in 3 daily doses <strong>of</strong> 800 cGy � 2400 cGy. SBRT<br />

planning target volumes were determined by both the radiation and<br />

gynecologic oncologist using non-contrasted CT and 18F-FDG PET/CT<br />

overlays. The primary endpoints were 6-month clinical benefit rate (#<br />

complete response � # partial response � # stable disease without PD [by<br />

RECIST v1.0] / 50), and less than 30-day posttherapy toxicity. Results:<br />

Between July 2009 and September 2011, 50 patients were enrolled and<br />

have a median posttherapy follow-up <strong>of</strong> 9 months. At 3 months, 50%<br />

(n�25) had complete response, 46% (n�23) had partial response, and<br />

4% (n�2) had stable disease in SBRT-targeted lesions. Twenty-six patients<br />

(52%) have had non-SBRT target PD and 18 (36%) have died <strong>of</strong> PD. Of the<br />

50 patients, 33 had a PD-free interval <strong>of</strong> at least 6 months, for an overall<br />

clinical benefit rate <strong>of</strong> 66%. Less than 30-day posttherapy SBRT-related<br />

toxicities were grade 2 fatigue (n �9 [18%]), grade 2 nausea (n �3[6%],<br />

grade 3 nephropathy (n �2[4%]), and grade 4 hyperbilirubinemia (n<br />

�1[2%]). Conclusions: This is the first phase II clinical trial <strong>of</strong> SBRT<br />

showing a clinically relevant benefit <strong>of</strong> ablative radiation in the setting <strong>of</strong><br />

recurrent gynecological disease. Despite excellent control <strong>of</strong> targeted<br />

lesions with minimal toxicity, non-SBRT target PD rates are high, spurring<br />

interest for future SBRT-chemotherapy clinical trials.<br />

5104 General Poster Session (Board #27B), Sun, 8:00 AM-12:00 PM<br />

A phase II study <strong>of</strong> dose-dense neoadjuvant chemotherapy with weekly<br />

paclitaxel and carboplatin followed by chemoradiation in locally advanced<br />

cervical carcinoma. Presenting Author: Rajkumar Bikramjit Singh, All India<br />

Institute <strong>of</strong> Institute, New Delhi, India<br />

Background: We prospectively studied dose dense neoadjuvant chemotherapy<br />

(NACT) designed for an enhanced cell kill, better local control and<br />

eradication <strong>of</strong> micrometastases prior to standard concurrent chemoradiation<br />

(CRT) in locally advanced cervical cancer. Methods: Between June<br />

2010 and December 2011, 21 patients (median age - 51 years, range 35 -<br />

67) <strong>of</strong> cervical cancer with locally advanced disease received NACT using<br />

paclitaxel (60mg/m2 ) and carboplatin (AUC-2) weekly for 6 doses. Patients<br />

(pts) then received concurrent CRT (external and brachytherapy) with<br />

weekly cisplatin (40mg/m2 for 6 doses) at a mean interval <strong>of</strong> 15 days (range<br />

7–20 days). The primary end-point was response rate i.e. complete<br />

response (CR) and partial response (PR) 12 weeks post CRT. Results:<br />

Baseline stages were: stage 2A - 19%, 3B - 71.4%, 4A - 9.5%. Squamous<br />

cell carcinoma and adenocarcinoma constituted 95.2% and 4.7% pts<br />

respectively. Following NACT, 66.6% pts responded (CR -9.5% %; PR –<br />

57.1 %), 23.8% had stable disease (SD) and 4.7% had progressive disease<br />

(PD). A total <strong>of</strong> 18 pts completed NACT and CRT <strong>of</strong> which 17 were in CR<br />

and 1 in PR. One patient with stage 4A disease developed vesicovaginal<br />

fistula at end <strong>of</strong> NACT for which she underwent pelvic exenteration and was<br />

in pathological CR. After NACT, one patient developed choroid metastases<br />

and was taken <strong>of</strong>f study protocol while another patient was lost to follow up.<br />

At a mean follow up 5.8 months (range 1 - 14), 90% pts were in CR, 5% in<br />

PR and 5% had PD. During NACT, Grade 3/4 neutropenia, thrombocytopenia<br />

and anemia were seen in 33.3%, 4.7% & 9.5% <strong>of</strong> pts, respectively and<br />

grade 3/4 non-hematological toxicities in 9.5% pts. Following CRT, Grade<br />

3/4 neutropenia, thrombocytopenia and anemia were seen in 25%, 5% and<br />

10% <strong>of</strong> pts, respectively while 20% pts had grade 3/4 non-hematological<br />

toxicities. G-CSF was used in 30% pts during NACT and 25% pts during<br />

CRT, respectively. Conclusions: NACT with weekly paclitaxel and carboplatin<br />

followed by radical CRT is feasible and is associated with a high<br />

response rate in locally advanced cervical cancer. This approach needs to<br />

be studied in a phase III trial.<br />

5103 General Poster Session (Board #27A), Sun, 8:00 AM-12:00 PM<br />

Neoadjuvant chemotherapy <strong>of</strong> docetaxel and carboplatin in patients with<br />

stage Ib2 to IIb nonsquamous cervix cancer <strong>of</strong> the uterus. Presenting<br />

Author: Shoji Nagao, Saitama Medical University International Medical<br />

Center, Hidaka, Japan<br />

Background: Non-squamous carcinoma <strong>of</strong> the uterine cervix has a resistance<br />

to radiation therapy and chemotherapy, and this is associated with<br />

worse prognosis compared with squamous carcinoma. This is a phase II<br />

study to assess an efficacy and safety <strong>of</strong> neoadjuvant chemotherapy <strong>of</strong><br />

docetaxel and carboplatin (DC therapy) in patients with stage Ib2 to II<br />

non-squamous cervix cancer <strong>of</strong> the uterus. Methods: Patients with histologically<br />

confirmed, stage Ib2-II non-squamous uterine cervix cancer were<br />

received DC therapy prior to type III radical hysterectomy. IV doceaxel was<br />

administered at 60 mg/m2 followed by IV carboplatin administration based<br />

on AUC�6. The treatment was repeated every 21 days for a total <strong>of</strong> 1 to 3<br />

cycles. Surgery was performed as soon as sufficient response was obtained.<br />

Results: Fifty-three women with non-squamous cervical carcinoma (FIGO<br />

stage Ib2,15; IIa, 7; IIb, 31) received DC therapy. Median age and tumor<br />

size were 49 years old (range: 27-71) and 52mm (range: 10-92),<br />

respectively. Fourty <strong>of</strong> 47 patients (74%) received 2 cycles <strong>of</strong> chemotherapy.<br />

<strong>Clinical</strong> response occurred in 69.8% <strong>of</strong> patients (complete<br />

response, 5; partial response, 32; stable disease, 15; disease progression,<br />

1), and 96% (52/54) <strong>of</strong> patients completed the surgery. Incidences <strong>of</strong><br />

grade 3/4 hematological toxicities were 98% (53/54) for neutrocytopenia,<br />

4% (2/54) for thrombocytopenia, and 9% (5/54) for anemia. Febrile<br />

neutropenia was observed in three patients. Observed grade 3/4 nonhematological<br />

toxicities included were 5 for nausea, 2 for vomiting, 3 for<br />

constipation, 4 for allergic reaction. Median follow-up duration was 860<br />

days with a range <strong>of</strong> 147- 1635 days. The 2-years progression-free survival<br />

rate and 2-years overall survival rate were 63% and 79% in stage IB2, 71%<br />

and 86% in stage IIA2, and 67% and 86% in stage IIB, respectively.Twenty<br />

patients recurred and twelve patients died. Conclusions: Neoadjuvant<br />

chemotherapy using DC therapy was well tolerated and had good response.<br />

It seems to be benefical in patients with satge Ib2 to II nonsquamous<br />

cervical cancer.<br />

5105 General Poster Session (Board #27C), Sun, 8:00 AM-12:00 PM<br />

Prognostic significance <strong>of</strong> high-risk human papilloma virus (HPV), p16,<br />

and p53 status in women with vulvar squamous cell carcinoma (VSCC).<br />

Presenting Author: Ghassan Allo, University <strong>of</strong> Toronto, Toronto, ON,<br />

Canada<br />

Background: The incidence <strong>of</strong> VSCC is increasing. Studies suggest the<br />

presence <strong>of</strong> two histologically and molecularly distinct subsets <strong>of</strong> VSCC;<br />

one contingent on and another independent <strong>of</strong> HPV infection. However, it is<br />

uncertain if HPV status has prognostic significance. HPV oncoproteins can<br />

result in degradation <strong>of</strong> the tumor suppressor p53, cell cycle deregulation<br />

and abnormal expression <strong>of</strong> cyclin dependant kinase inhibitor p16. The aim<br />

<strong>of</strong> this study was to investigate HPV infection, p16 and p53 in relation to<br />

clinical parameters in women with VSCC. Methods: Sequential cases <strong>of</strong><br />

VSCC from patients (pts) treated at Princess Margaret Hospital (PMH) from<br />

2000 to 2008 were reviewed. HPV infection was evaluated by Roche Linear<br />

array. A tissue microarray was constructed. p16 and p53 immunohistochemistry<br />

was performed. <strong>Clinical</strong> data was abstracted from medical records and<br />

PMH Cancer Database. Survival analysis was performed using Kaplan-<br />

Meier curves and log rank test. Results: We identified124 pts with VSCC.<br />

HPV was detected in 43/123 (35%) pts (median age 71 � 16 yrs). HPV16<br />

was the most common serotype (38/43; 88.4%). p16 was expressed in<br />

30/115 (26%) pts and p53 in 59/117 (50.4%) pts. Median age <strong>of</strong> pts was<br />

not different in relation to HPV, p16 and p53 status. Expression <strong>of</strong> p16<br />

(p�0.0001) and loss <strong>of</strong> p53 (p�0.007) were associated with HPV<br />

infection. Pts with HPV positive tumors were less likely to recur (recurrence<br />

rate at 5 years (RR) 12.5% vs 50.3%, p�0.009). HPV positive VSCC were<br />

not associated with better 5 yr disease free survival (DFS), 58% vs 31%;<br />

p�0.15, or overall survival (OS), 61% vs 61% ; p�0.94. p16 positive<br />

tumors had a lower RR at 5 yrs, 23.8% vs 59%, p�0.006 and better 5yr<br />

DFS (61% vs 27% ; p�0.009) but not significant for OS (65% vs 59%;<br />

p�0.94). Among pts with HPV positive VSCC, OS and DFS were not<br />

different between p16 positive and negative VSCC. In the 46 pts treated<br />

with radiotherapy, HPV and p16 positive tumors were associated with a<br />

lower RR (p�0.004 and 0.005). p53 expression was not prognostic in any<br />

pt group. Conclusions: Women with HPV-positiveVSCC have a lower risk <strong>of</strong><br />

disease recurrence. p16-expressing VSCC are associated with reduced<br />

disease recurrence and improved DFS.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


5106 General Poster Session (Board #28A), Sun, 8:00 AM-12:00 PM<br />

ADXS11-001 immunotherapy targeting HPV-E7: Preliminary survival data<br />

from a phase II study in Indian women with recurrent/refractory cervical<br />

cancer. Presenting Author: <strong>Part</strong>ha Basu, Chittaranjan National Cancer<br />

Institute, Kolkata, India<br />

Background: ADXS11-001 immunotherapy is a live attenuated Listeria<br />

monocytogenes (Lm) bioengineered to secrete a HPV-16-E7 fusion protein<br />

targeting HPV transformed cells. The Lm vector serves as its own adjuvant<br />

and infects APC where it naturally cross presents, stimulating both MHC<br />

class 1 and 2 pathways resulting in specific T-cell immunity to tumors.<br />

Here we describe the preliminary survival data associated with ADXS11-<br />

001 administration in Lm-LLO-E7-015, a randomized phase II study being<br />

conducted in India in 110 patients with recurrent/refractory cervical cancer<br />

who have been treated previously with chemotherapy, radiotherapy or both.<br />

Methods: Patients are randomized to either 3 doses <strong>of</strong> ADXS11-001 at 1 x<br />

109 CFU or 4 doses <strong>of</strong> ADXS11-001 at 1 x 109CFU with cisplatin<br />

chemotherapy. Naprosyn and oral promethazine are given as premedications<br />

and a course <strong>of</strong> ampicillin is given 72h after infusion thereby clearing<br />

any residual vector. Patients receive CT scans at baseline and Days 84,<br />

184, 273, 365 and 545. The primary endpoint is 12 month survival.<br />

Results: As <strong>of</strong> January 26, 2012, 88 patients have received 200 doses <strong>of</strong><br />

ADXS11-001; with the percentage <strong>of</strong> patients alive at 6 months at 62%<br />

(34/55); at 9 months at 41% (15/37) and at 12 months at 40% (6/15).<br />

Tumor responses have been observed in both treatment arms with 3<br />

complete responses (elimination <strong>of</strong> tumor burden) and 4 partial responses<br />

(�30% reduction in tumor burden) by RECIST. One serious (Gr3) adverse<br />

event and 77 mild-moderate (Gr 1-2) adverse events possibly related to<br />

study treatment have been reported in 35% (31/88) <strong>of</strong> patients. The<br />

non-serious adverse events consisted predominately <strong>of</strong> transient, noncumulative<br />

flu-like symptoms associated with infusion that responded to<br />

symptomatic treatment, or resolved on their own within hours <strong>of</strong> treatment.<br />

Conclusions: This immunotherapy can be safely administered to patients<br />

with advanced cancer alone and in combination with chemotherapy.<br />

ADXS11-001 is well tolerated and presents a predictable and manageable<br />

safety pr<strong>of</strong>ile. Early signs <strong>of</strong> clinical benefit merit further investigation.<br />

Updated findings will be presented at the meeting.<br />

5108 General Poster Session (Board #28C), Sun, 8:00 AM-12:00 PM<br />

Preoperative geriatric assessment (GA) and surgical outcomes in older<br />

women with gynecological (gyn) cancer. Presenting Author: Daneng Li,<br />

Memorial Sloan-Kettering Cancer Center, New York, NY<br />

Background: GA can predict surgical outcomes in older patients (pts);<br />

however, pre-surgical evaluation for older pts with gyn malignancies has not<br />

been well-described. This study will determine the association between GA<br />

variables with post-operative morbidity and mortality. Methods: Women<br />

75yrs or older who had geriatric evaluation before any gyn surgery at<br />

Memorial Sloan Kettering Cancer Center (MSKCC) between 1/2010-6/<br />

2011 were identified. Pre-operative GA included: Mini-Cog Test (cognition),<br />

fall history, medication list, nutritional status (weight loss �10lbs,<br />

albumin), functional status (activities <strong>of</strong> daily living (ADL), instrumental<br />

I-ADL), and Charlson comorbidity index. Outcomes included: delirium,<br />

length <strong>of</strong> hospital stay (LOS), 30-day surgical adverse events (AE, grade<br />

1-5, via prospective-MSKCC surgical database), 30-day hospital readmission<br />

and 6-month mortality. Utilizing bivariate analyses, associations<br />

between GA measures and post-operative outcomes were evaluated.<br />

Results: 72 pts (median age 79yrs, range 75-92) with gyn cancer (54%<br />

uterine, 36% ovarian/peritoneal/tubal, 10% cervical/vaginal/vulvar) had<br />

gyn surgery. 34 pts (47%) had stage III/IV disease. 21pts (30%) had<br />

secondary cancer history. Pt’s baseline GA measures: ADL-dependent<br />

(13%), IADL-dependent (19%), weight loss (18%), fall history (18%), mini<br />

cog score (median 4, range 0-5), Charlson score (median 2, range 0-9).<br />

24pts (33%) had surgical AE; no significant association with age or GA.<br />

Median LOS was 2 days (range 0-20); 11pts (15%) required 30-day<br />

readmission. Delirium (p�0.01), nutrition (weight loss p�0.04, albumin<br />

p�0.04), anemia (p�0.003) and high comorbidity index (p�0.013) were<br />

associated with longer LOS. Six-month mortality was 8%; older age<br />

(p�0.02), poor functional status (lower ADL and IADL, p�0.001 and<br />

p�0.007), number <strong>of</strong> medications (p�0.05) and poor cognition (p�0.001)<br />

were associated with shorter survival. Conclusions: Surgical morbidity is<br />

common in older pts. Although AE’s were not associated with GA variables,<br />

GA can detect high-risk features for longer LOS and shorter survival.<br />

Further prospective studies with pre-operative GA and interventions are<br />

warranted.<br />

Gynecologic Cancer<br />

353s<br />

5107 General Poster Session (Board #28B), Sun, 8:00 AM-12:00 PM<br />

Cervical cancer treatment for operable lesions in a low-resource contemporary<br />

setting. Presenting Author: Caroline Dorothy Lynch, Indiana University,<br />

Indianapolis, IN<br />

Background: To compare HIV� and HIV- women with operable cervical<br />

cancer in a low resource contemporary setting. Methods: A retrospective<br />

study using well-matched controls from a Kenyan teaching and referral<br />

hospital. Results: 183 women were treated for cervical cancer between<br />

October 2007 and June 2011. The histologic subtype was squamous cell in<br />

all but one case. At presentation, 28 had operable lesions (Stage IA1–<br />

IIB1); 7 more received neoadjuvant chemotherapy prior to surgery. HIV<br />

seroprevalence was 54% (18/33) among initial operative cases and 57%<br />

among the neoadjuvant group (p�ns). Mean age was 42 (HIV�), and 43<br />

(HIV-), (range 25-64). HIV- vs. HIV� cervical cancer patients (mean CD4<br />

count 373, 50%�200) were detected by visual inspection with acetic acid<br />

(VIA) (18% (2/11)vs 68% (15/22) p�.099), symptoms (27%(3/11) vs<br />

14%(3/22) p�.43), or Pap smear (45% (5/11) vs .09% (2/22) p�.06),<br />

respectively.HIV� patients (two Stage IB1, two Stage IB2) did not require<br />

more downstaging than HIV- patients (two stage IIB, one stage IIIA) before<br />

surgery (18% (4/22) vs 27% (3/11) p�.63). Surgical treatments were not<br />

statistically different in either group and included radical hysterectomy(25),<br />

total abdominal hysterectomy(2), cesarean hysterectomy(1),<br />

and total vaginal hysterectomy(5). Postoperative complications included<br />

fever, dehiscence, DVT, ileus, fistula, and infectious complications (chest,<br />

urinary tract, wound). One HIV- patient suffered postoperative fever,<br />

vesicovaginal fistula, and wound dehiscence (overall complications<br />

.06%).Lymph node involvement was noted in 7 HIV� and 3 HIV- patients<br />

who underwent full staging procedures (p�.004). Conclusions: In patients<br />

with operable cervical cancer, HIV serostatus does not affect complication<br />

rate or influence need for downstaging prior to surgery compared to a<br />

well-matched control group. HIV� patients were not more likely to receive<br />

neoadjuvant chemotherapy but were more likely to have positive lymph<br />

nodes. VIA detected the majority <strong>of</strong> cervical cancers HIV� patients.<br />

5109 General Poster Session (Board #29A), Sun, 8:00 AM-12:00 PM<br />

Genome-wide association analysis in host characteristics <strong>of</strong> progression to<br />

high-grade cervical intraepithelial neoplasia for women with human papilloma<br />

virus infection and normal cytology. Presenting Author: Chyong-Huey<br />

Lai, Gynecologic Cancer Research Center, Chang Gung Memorial Hospital<br />

and Department <strong>of</strong> Obstetrics and Gynecology, Chang Gung University<br />

College <strong>of</strong> Medicine, Taoyuan, Taiwan<br />

Background: Although it is well accepted that persistent human papillomavirus<br />

(HPV) is necessary for carcinogenesis <strong>of</strong> cervical neoplasms, the<br />

molecular mechanism for progression is unclear. HPV testing is widely used<br />

for cervical cancer screening. The hazard ratio <strong>of</strong> developing cervical<br />

intraepithelial neoplasia grade 2 or more severe (CIN2�) in baseline<br />

HPV-positive/normal cytology women is 30-50 fold as compared with<br />

HPV-negative/normal cytoloy women. HPV positivity would cause substantial<br />

anxiety. It is important to identify a prognostic pr<strong>of</strong>ile for predicting<br />

progression. Methods: We collected blood samples from women aged � 30<br />

years in a population-based nested cohort study enrolling women with HPV<br />

infection (n � 871) or HPV-negative (n � 902) with normal cytology in<br />

2004-2009 for prospective follow-up. To identify the host genetic characteristics<br />

associated with cervical carcinogenesis, a genome-wide association<br />

study (analyzing 530,194 SNPs) was conducted in 23 cases who<br />

developed CIN2� and 62 viral type- and age-matched controls who were<br />

HPV-positive at baseline without developing CIN throughout the follow-up<br />

period. Results: One SNP with significant P values (rs16969682; P�4.58 x<br />

10-5 ,OR�5.9, 95% CI�2.52-13.96) located in SEC14-like 1 (SEC14L1)<br />

gene localized to chromosome 17 was identified with association between<br />

progression to CIN2� and controls without CIN throughout follow-up.<br />

SEC14L1 belongs to the widely-expressed SEC14-superfamily. This superfamily<br />

consists <strong>of</strong> � 500 members that are involved in biological functions<br />

including membrane trafficking and phospolipid metabolism. Furthermore,<br />

using T-cell based cDNA screening, SEC14L-1a is identified as a regulator<br />

<strong>of</strong> HIV-1 replication. Conclusions: The potential role <strong>of</strong> SEC14L1 in<br />

host-viral interaction will be further elucidated. Additional statistically<br />

significant SNPs including rs16969682 will be validated on cases with<br />

CIN2� (n � 250) and normal controls (normal cytology/HPV-negative at<br />

baseline without acquisition <strong>of</strong> HPV or abnormal cytology/histology during<br />

follow-up, n � 500).<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


354s Gynecologic Cancer<br />

5110 General Poster Session (Board #29B), Sun, 8:00 AM-12:00 PM<br />

Validation <strong>of</strong> the predictive value <strong>of</strong> modeled HCG residual production “P”<br />

in low-risk gestational trophoblastic neoplasia (GTN) patients treated in<br />

GOG-174 phase III trial. Presenting Author: Benoit You, Centre Hospitalier<br />

Lyon Sud; Hospices Civils de Lyon; EMR UCBL/HCL 3738, Faculté de<br />

Médecine Lyon-Sud, Université Claude Bernard Lyon1, Lyon, France<br />

Background: In low-risk GTN, chemotherapy is changed according to serum<br />

hCG levels. We previously showed mathematical modeling <strong>of</strong> hCG kinetics<br />

provides a parameter production “P”, a useful early predictor <strong>of</strong> methotrexate<br />

(MTX) resistance (You et al; Ann Oncol 2010; ASCO and ISSTD 2011).<br />

We applied this approach to patient cohort <strong>of</strong> GOG-174 trial, in which<br />

weekly MTX (Arm 1) was compared to dactinomycin (Arm 2). Methods:<br />

Database (210 patients, including 78 with resistance) was split into 2 sets.<br />

A 126 patient Model Set was initially used to adjust model parameters.<br />

Patient hCG kinetics from day7 to day50 were fit with NONMEM program<br />

to: “[hCG(time)] � hCG0i * exp(–k*time) � P”; where P is residual hCG<br />

tumor production, hCG0i is the initial hCG level, and k is the rate constant.<br />

Three putative P-based classifiers <strong>of</strong> resistance were assessed using ROC<br />

analyses. Then an 84 patient Test Set with blinded-resistance status was<br />

used to assess the validity <strong>of</strong> predictions. The primary endpoint was<br />

treatment resistance defined as relapse and/or lack <strong>of</strong> hCG normalization.<br />

Results: Due to initial surge in 14% patients, hCG kinetic modeling was<br />

started on day7. Individual hCG decline pr<strong>of</strong>iles <strong>of</strong> Model Set patients were<br />

modeled. There was no impact <strong>of</strong> treatment arm on variability <strong>of</strong> kinetic<br />

parameter estimates. The best P cut-<strong>of</strong>fs to discriminate resistant vs.<br />

sensitive patients were 7.7 in Arm 1 and 74.0 in Arm 2. They were<br />

combined to define 2 predictive groups with low vs. high risks <strong>of</strong> resistance<br />

(ROC AUC � 0.82; Se � 93.8%; Sp � 70.5%). The model was then<br />

applied to Test Set patient cohort. The predictive value <strong>of</strong> P-based<br />

predictive groups regarding resistance was reproducible (ROC AUC � 0.81;<br />

Se � 88.9% (95% CI: 70.8%-97.7%); Sp � 73.1% (95% CI: 60.0%-<br />

84.4%)). Both P and treatment arm were associated with resistance using<br />

multivariate logistic regression tests. Predictive value <strong>of</strong> P was less<br />

accurate in dactinomycin arm. Conclusions: The early predictive value <strong>of</strong><br />

the modeled kinetic parameter P regarding resistance appears promising in<br />

GOG-174 study, especially in MTX arm. This is the second positive<br />

evaluation <strong>of</strong> this procedure study. Prospective validation is warranted.<br />

TPS5112 General Poster Session (Board #30A), Sun, 8:00 AM-12:00 PM<br />

OPSALIN: A phase II placebo-controlled randomized study <strong>of</strong> ombrabulin<br />

in patients with platinum-sensitive recurrent ovarian cancer treated with<br />

carboplatin (Cb) and paclitaxel (P). Presenting Author: Eric Pujade-<br />

Lauraine, Université Paris Descartes, AP-HP, Hôpitaux Universitaires Paris<br />

Centre, Site Hôtel-Dieu, Paris, France<br />

Background: Most women with ovarian cancer have disease recurrence after<br />

responding to their first treatment with platinum-based chemotherapy and<br />

are considered to have platinum-sensitive disease if the relapse-free period<br />

is more than 6 months. Although CbP is standard first-line chemotherapy<br />

for ovarian cancer, patients with platinum-sensitive recurrent disease are<br />

<strong>of</strong>ten retreated with CbP. Ombrabulin (AVE8062) is a vascular disrupting<br />

agent and analogue <strong>of</strong> combretastatin A4 that damages established tumor<br />

vasculature causing tumor necrosis and has synergistic antitumor activity<br />

with platinum agents in tumor models in vivo (Cancer Sci. 2003;94:200).<br />

A phase I study showed that treatment with vivo ombrabulin plus CbP is<br />

feasible in patients with advanced solid tumors (NCI-AACR-EORTC 2010;<br />

Abs 386). We initiated OPSALIN, a phase II randomized trial, to evaluate<br />

whether the addition <strong>of</strong> ombrabulin to CbP improves outcomes in patients<br />

with platinum-sensitive recurrent ovarian cancer (NCT01332656;<br />

EFC10260). Methods: Eligibility criteria include age <strong>of</strong> at least 18 years,<br />

ECOG PS 0–2, measurable carcinoma <strong>of</strong> the ovarian epithelium, fallopian<br />

tube, or primary peritoneum that is platinum sensitive, and completion <strong>of</strong><br />

only one previous line <strong>of</strong> platinum-based chemotherapy. Exclusion criteria<br />

include uncontrolled brain metastases, peripheral neuropathy �grade 1,<br />

and a prior history <strong>of</strong> cardiovascular disease. Patients are being randomized<br />

(1:1) to receive either ombrabulin 35mg/m2 or placebo plus CbP every 3<br />

weeks. Assigned treatment will continue until disease progression or death,<br />

unacceptable toxicity, or consent withdrawal. The primary endpoint is<br />

progression-free survival stratified by the time <strong>of</strong> first disease recurrence<br />

(6–12 months or �12 months). Secondary endpoints include safety,<br />

response rate, overall survival, pharmacokinetics, and analysis <strong>of</strong> predictive/<br />

prognostic biomarkers. Planned randomization is a total <strong>of</strong> 150 patients at<br />

approximately 45 sites globally. Sixty-five patients have been randomized<br />

as <strong>of</strong> January 2012.<br />

5111 General Poster Session (Board #29C), Sun, 8:00 AM-12:00 PM<br />

Nomogram prediction for overall survival <strong>of</strong> patients diagnosed with<br />

cervical cancer. Presenting Author: Christoph Grimm, Department <strong>of</strong><br />

General Gynecology and Gynecologic Oncology, Comprehensive Cancer<br />

Center, Medical University <strong>of</strong> Vienna, Vienna, Austria<br />

Background: Cervical cancer is clinically staged based upon the International<br />

Federation <strong>of</strong> Gynecologists and Obstetricians (FIGO) system. FIGO<br />

stage is well established as prognostic parameter. It is well known that<br />

other additional parameters are useful to estimate overall survival (OS) in<br />

patients with cervical cancer. The aim <strong>of</strong> this multi-center was to create a<br />

nomogram to predict OS in patients diagnosed with cervical cancer.<br />

Methods: Cervical cancer databases <strong>of</strong> two large Austrian institutions were<br />

analysed. Characteristics known to predict OS were collected. For each<br />

patient association between each prognostic parameter and OS was<br />

assessed by multivariable modeling. The corresponding 3-year and 5-year<br />

OS probabilities were then determined using the nomogram. The constructed<br />

nomogram was then validated using the bootstrap correction<br />

technique. Results: Mean 5-year OS rates for patients with FIGO stage IA,<br />

IB, II, III, and IV were 99.0% (1.0), 88.6% (3.0), 65.8% (5.2), 58.7%<br />

(11.0), and 41.5% (14.7), respectively (p�0.001). Mean five-year OS<br />

time was 44.2 (30.9) months. Based on the multivariable model FIGO<br />

stage, tumor size, age, histologic subtype, lymph node ratio, and parametrial<br />

involvement were identified as nomogram parameters. The bootstrap<br />

sample corrections provided an estimated concordance probability (interquartile<br />

range) <strong>of</strong> 0.794 (0.779-0.805). Conclusions: Based on 6 easily<br />

available parameters a novel nomogram to predict 3-year and 5-year OS <strong>of</strong><br />

patients diagnosed with cervical cancer was constructed and internally<br />

validated. Application <strong>of</strong> this nomogram allows more accurate and individual<br />

prediction <strong>of</strong> patients’ prognosis.<br />

TPS5113 General Poster Session (Board #30B), Sun, 8:00 AM-12:00 PM<br />

A study <strong>of</strong> autologous dendritic cell therapy targeting mucin-1 for treatment<br />

<strong>of</strong> patients with epithelial ovarian cancer in first remission. Presenting<br />

Author: Jonathan S. Berek, Stanford Cancer Center, Stanford, CA<br />

Background: Cvac (Cvac) is an autologous dendritic cell immune stimulant<br />

targeting mucin-1 positive tumors. A phase IIa study demonstrated<br />

potential for treating mucin-1 positive epithelial ovarian carcinoma (EOC).<br />

A phase IIb study was designed to determine on a pilot basis if Cvac could<br />

demonstrate an effect on progression free and overall survival in patients<br />

with EOC in first or second remission. The current study, CANVAS (CANcer<br />

VAccine Study) is evaluating Cvac treatment <strong>of</strong> EOC patients in first<br />

remission after surgery and standard chemotherapy, with at least three<br />

cycles <strong>of</strong> platinum and taxane therapy. Endpoints include progression free<br />

survival, overall survival, safety evaluation and quality <strong>of</strong> life scores.<br />

Methods: Patients are eligible if they have stage III or IV mucin-1 positive<br />

EOC and have had optimal cytoreductive (�1cm residual disease) surgery.<br />

Apheresis to obtain immature dendritic cells occurs prior to chemotherapy.<br />

Cvac is manufactured during chemotherapy. Patients remain eligible for<br />

the study if they have a complete response to standard chemotherapy after<br />

surgery. Cvac or placebo is dosed every four weeks for three doses, and<br />

every twelve weeks for three additional doses. 1000 patients will be<br />

randomized to Cvac or placebo in 22 countries, with 800 expected to<br />

receive vaccine or placebo. CT or MRI imaging determines progression, and<br />

is centrally read prior to inclusion and at each timepoint. Recruitment<br />

commenced January 2012. Over 100 centers are participating in the study<br />

globally, with majority in Europe. Enrollment is expected to take 18-24<br />

months. An interim analysis is planned after 400 PFS events, and final<br />

analysis after 620 PFS events. Observation <strong>of</strong> 620 events would have<br />

approximately 90% power to detect a hazard ratio <strong>of</strong> 0.77 in PFS at a<br />

significance level <strong>of</strong> p�0.049 (approximately a 30% increase in median<br />

PFS in the Cvac group compared with the placebo group).<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


TPS5114 General Poster Session (Board #30C), Sun, 8:00 AM-12:00 PM<br />

ACRIN 6695 perfusion CT as prognostic imaging biomarker in ovarian<br />

cancer. Presenting Author: Chaan S. Ng, University <strong>of</strong> Texas M. D.<br />

Anderson Cancer Center, Houston, TX<br />

Background: Tumor size and the cell surface glycoprotein CA125 levels<br />

have been traditional biomarkers for ovarian carcinoma, but remain<br />

suboptimal for assessing patients receiving chemotherapy. Current morphological<br />

criteria do not adequately evaluate lesion necrosis from antiangiogenic<br />

therapy when no tumor volume change is measured. The<br />

evaluation <strong>of</strong> functional biomarkers rather than tumor volume may better<br />

distinguish responders from non-responders early in treatment with antiangiogenic<br />

therapy. Perfusion CT can evaluate changes in tumor vascularity<br />

including blood flow (BF), blood volume (BV), mean transit time (MTT) and<br />

capillary permeability surface product (PS) before and after antiangiogenic<br />

therapy with/out decrease in tumor volume. Objectives: The<br />

aims <strong>of</strong> the study are to evaluate the relationship between changes in tumor<br />

perfusion parameters and clinical outcomes <strong>of</strong> progression free survival,<br />

overall survival, and standard RECIST anatomic response criteria. A<br />

test-retest perfusion CT scan will also be performed to evaluate reproducibility<br />

<strong>of</strong> perfusion parameters in a subset <strong>of</strong> participants. Methods: In this<br />

collaborative trial, participants will be co-enrolled in the GOG-262 treatment<br />

trial. <strong>Part</strong>icipants will receive doublet chemotherapy <strong>of</strong> paclitaxel and<br />

carboplatin, followed by anti-angiogenic monoclonal therapy at cycle two.<br />

Perfusion CT will be performed at three time points: at baseline prior to<br />

therapy (T0), between days 18 and 21 <strong>of</strong> cycle one chemotherapy (T1), and<br />

at 8-10 days (T2) in anti-angiogenic monoclonal therapy. <strong>Part</strong>icipants with<br />

primary epithelial ovarian, peritoneal or fallopian tube cancer with optimally<br />

or suboptimally debulked FIGO Stage II, III or IV disease are eligible<br />

for the trial. Lesion eligibility will be evaluated by size and attenuation<br />

criteria. Accrual: ACRIN 6695 is activated at 12 GOG sites; 4/78<br />

participants have accrued Discussion: Perfusion CT has been readily<br />

incorporated into the pre-existing clinical CT protocols and during scheduled<br />

RECIST scans. These perfusion CT functional maps <strong>of</strong> BF, BV, MTT<br />

and PS have been generated using vendor provided s<strong>of</strong>tware without issue.<br />

Contact: Please contact Chaan Ng, MD cng@mdanderson.org for additional<br />

information.<br />

TPS5116 General Poster Session (Board #31B), Sun, 8:00 AM-12:00 PM<br />

A phase III trial <strong>of</strong> adjuvant chemotherapy following chemoradiation as<br />

primary treatment for locally advanced cervical cancer compared to<br />

chemoradiation alone: The OUTBACK TRIAL. Presenting Author: Linda R.<br />

Mileshkin, Peter MacCallum Cancer Centre, Melbourne, Australia<br />

Background: Cervical cancer is a global health problem and the most<br />

common cause <strong>of</strong> cancer-related death among women in developing<br />

nations. Despite the recently developed cervical cancer vaccine, many<br />

women will continue to die from cervical cancer for many decades unless<br />

existing treatments can be improved. Unscreened women <strong>of</strong>ten present<br />

with locally-advanced disease that has a 5 year overall survival (OS) rate <strong>of</strong><br />

60% or less following standard chemo-radiation. Although some evidence<br />

suggests that adjuvant chemotherapy following chemo-radiation may be <strong>of</strong><br />

value, its role remains controversial. Methods: OUTBACK is a randomized<br />

phase III Gynecologic Cancer InterGroup (GCIG) trial designed and led by<br />

the Australia New Zealand Gynaecological Oncology Group (ANZGOG) in<br />

collaboration with the NHMRC <strong>Clinical</strong> Trials Centre. <strong>Part</strong>icipating countries<br />

(groups) include Australia and New Zealand (ANZGOG), India, the<br />

USA and Canada (GOG, RTOG). OUTBACK is suitable for women with<br />

locally advanced cervical cancer (FIGO stage IB1 and node positive, IB2, II,<br />

IIIB or IVA). The primary objective is to determine if the addition <strong>of</strong><br />

adjuvant chemotherapy to standard cisplatin-based chemo-radiation improves<br />

OS. Women are randomized to either A) standard cisplatin-based<br />

chemo-radiation or B) standard cisplatin-based chemo-radiation followed<br />

by 4 cycles <strong>of</strong> carboplatin and paclitaxel chemotherapy. Secondary<br />

objectives are to compare progression-free survival, treatment-related<br />

toxicity, patterns <strong>of</strong> disease recurrence, quality <strong>of</strong> life and psycho-sexual<br />

health, and the association between radiation protocol compliance and<br />

outcomes. Blood and tumour samples are collected from consenting<br />

patients for future translational studies. 780 women will be enrolled to<br />

determine if the addition <strong>of</strong> adjuvant chemotherapy can improve the 5-year<br />

OS rate by � 10%. OUTBACK opened in Australia and New Zealand in<br />

2011. In early 2012 the trial opened in the USA and activation <strong>of</strong> GOG<br />

sites is ongoing. 15 patients have been randomized. It is expected that<br />

RTOG and India will open the trial later this year with recruitment<br />

increasing substantially once all sites are activated.<br />

Gynecologic Cancer<br />

355s<br />

TPS5115 General Poster Session (Board #31A), Sun, 8:00 AM-12:00 PM<br />

A phase II study <strong>of</strong> live-attenuated listeria monocytogenes immunotherapy<br />

(ADXS11-001) in the treatment <strong>of</strong> persistent or recurrent cancer <strong>of</strong> the<br />

cervix (GOG-0265). Presenting Author: Warner King Huh, University <strong>of</strong><br />

Alabama at Birmingham, Birmingham, AL<br />

Background: This is a GOG/NCI sponsored phase II study (NCT01266460,<br />

GOG 0265) <strong>of</strong> ADXS11-001 in patients with persistent or recurrent cancer<br />

<strong>of</strong> the cervix. ADXS11-001 is a live attenuated Listeria monocytogenes<br />

(Lm) immunotherapy bioengineered to secret a HPV E7 fusion protein<br />

targeting HPV-E7 transformed cells. A previous phase I dose escalation<br />

study determined the safety <strong>of</strong> ADXS11-001 in patients with late stage<br />

cervical cancer (Maciag PA. Vaccine. 2009 Jun 18;27(30):3975-83). The<br />

primary objectives <strong>of</strong> this study are: to evaluate the tolerability and safety <strong>of</strong><br />

ADXS11-001; and to assess the activity <strong>of</strong> ADXS11-001 in patients with<br />

persistent or recurrent cancer <strong>of</strong> the cervix. Secondary objectives are<br />

progression-free survival, overall survival and objective tumor response.<br />

Methods: Patient eligibility criteria: Females age � 18 years with persistent<br />

or recurrent squamous or non-squamous cell carcinoma, adenosquamous<br />

carcinoma, or adenocarcinoma <strong>of</strong> the cervix with documented disease<br />

progression (disease not amenable to curative therapy). Patients must have<br />

measurable disease as defined by RECIST 1.1; at least one “target lesion”<br />

as defined by RECIST 1.1; have had one prior systemic chemotherapeutic<br />

regimen for management <strong>of</strong> their disease; have adequate organ function<br />

and must be free <strong>of</strong> active infection and not on antibiotics. This protocol is a<br />

Simon 2-stage design with a planned sample size <strong>of</strong> up to 67 patients.<br />

Patients will receive ADXS11-001 at a dose <strong>of</strong> 1x109 CFU on Day 1 and<br />

repeat every 28 days for 3 total doses in the absence <strong>of</strong> disease progression<br />

or unacceptable toxicity, with each dose followed at 72 hours by a7day<br />

course <strong>of</strong> ampicillin, 500 mg QID. Tumor tissue and serum samples may be<br />

collected periodically for translational research. After completion <strong>of</strong> study<br />

treatment, patients are followed every 3 months for 2 years and then every<br />

6 months for 3 years. This phase II study just began enrolling and as <strong>of</strong><br />

January 27, 2012, 2 patients have been enrolled.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


356s Head and Neck Cancer<br />

5500 Oral Abstract Session, Sun, 8:00 AM-11:00 AM<br />

DeCIDE: A phase III randomized trial <strong>of</strong> docetaxel (D), cisplatin (P),<br />

5-fluorouracil (F) (TPF) induction chemotherapy (IC) in patients with<br />

N2/N3 locally advanced squamous cell carcinoma <strong>of</strong> the head and neck<br />

(SCCHN). Presenting Author: Ezra E.W. Cohen, The University <strong>of</strong> Chicago,<br />

Chicago, IL<br />

Background: IC is associated with lower distant failure (DF) rates in SCCHN<br />

but an improvement in overall survival (OS) has not been validated. The<br />

goal <strong>of</strong> this trial was to determine whether IC prior to chemoradiotherapy<br />

(CRT) improves survival compared to CRT alone. Methods: In this phase 3,<br />

open-label trial, subjects with pathologically confirmed SCCHN; N2/N3<br />

disease without metastases; no prior therapy; KPS ³ 70%; and intact organ<br />

function were randomized to CRT alone (CRT arm) [5 days <strong>of</strong> D (25 mg/m 2 ),<br />

F (600 mg/m 2 ), hydroxyurea (500 mg BID), and RT (150 cGy BID) followed<br />

by a 9 day break] or to 2 cycles <strong>of</strong> IC [D (75 mg/m2 ), P (75 mg/m2 ), F (750<br />

mg/m2 day 1-5)] followed by the same CRT (IC arm). Primary endpoint was<br />

OS. Secondary endpoints included DF free survival, failure pattern, and<br />

recurrence-free survival (RFS). 280 subjects provided 80% power to detect<br />

a hazard ratio HR�0.5 for OS (a�0.05). Results: 280 subjects were<br />

accrued from 2004-09 with minimum follow-up 24 months. Of 142<br />

patients randomized to IC, 91% received 2 cycles and 87% continued to<br />

CRT. Treatment adherence during CRT was high for docetaxel and<br />

hydroxyurea, but fewer than 75% <strong>of</strong> the patients received target dose <strong>of</strong><br />

5FU in both arms. RT was delivered without major deviations in 94% and<br />

95% <strong>of</strong> patients on IC and CRT arms, respectively. The most common grade<br />

3-4 toxicities during IC were febrile neutropenia (9%) and mucositis (8%),<br />

and during CRT (both arms combined) they were mucositis (45%),<br />

dermatitis (19%), and leukopenia (17%). Only grade 3-4 leukopenia and<br />

neutropenia rates were significantly higher in IC (p�0.002 and p�0.02,<br />

respectively). Table shows efficacy. Conclusions: High survival rates were<br />

observed in both arms. Further analysis and follow-up may provide insight<br />

into why the significant decrease in DF did not translate into improved OS.<br />

3-year outcomes.<br />

Endpoint<br />

IC arm<br />

(%)<br />

CRT arm<br />

(%) HR 95% CI p value<br />

OS 75 73 0.92 0.59-1.42 0.70<br />

DF-free survival 69 64 0.84 0.56-1.26 0.39<br />

RFS 67 59 0.76 0.52-1.13 0.18<br />

Cumulative incidence <strong>of</strong> DF 10 19 0.46 0.23-0.92 0.025<br />

Cumulative incidence <strong>of</strong><br />

locoregional failure<br />

9 12 0.79 0.37-1.68 0.55<br />

5502 Oral Abstract Session, Sun, 8:00 AM-11:00 AM<br />

A phase II, randomized trial (CONCERT-1) <strong>of</strong> chemoradiotherapy (CRT)<br />

with or without panitumumab (pmab) in patients (pts) with unresected,<br />

locally advanced squamous cell carcinoma <strong>of</strong> the head and neck<br />

(LASCCHN). Presenting Author: Jordi Giralt, Hospital Vall d’Hebron,<br />

Barcelona, Spain<br />

Background: Pmab is a fully human monoclonal antibody against the<br />

epidermal growth factor receptor. We evaluated the safety and efficacy in<br />

pts receiving CRT alone or CRT plus pmab (PCRT) as 1st-line treatment <strong>of</strong><br />

LASCCHN (sponsored and funded by Amgen Inc.). Methods: Pts with stage<br />

III, IVA, or IVB previously untreated LASCCHN <strong>of</strong> all sites excluding the<br />

nasopharynx were randomized 2:3 to open-label CRT or PCRT. CRT<br />

included cisplatin 100 mg/m2 for 3 cycles during standard fractionation<br />

radiotherapy (RT). PCRT included pmab 9.0 mg/kg � cisplatin 75 mg/m2 ,<br />

both administered with RT as in the CRT arm. The primary endpoint was<br />

local regional control (LRC) rate at 2 years; key secondary endpoints<br />

included PFS, OS, and safety. Preplanned HPV subset analysis, as<br />

determined by p16 immunohistochemistry, was performed on available<br />

samples. Results: Of 150 treated pts (87 pts PCRT, 63 pts CRT), 87% were<br />

men; median (range) age was 57 (39-77) years; ECOG PS 0: 68%. Of 99<br />

pts with tumor evaluable for HPV, 42% were HPV�. Overall, the 2-year LRC<br />

rate (95% CI) was 61% (50%-71%) for PCRT and 68% (54%-78%) for<br />

CRT. PFS events occurred in 40% <strong>of</strong> the PCRT and 35% <strong>of</strong> CRT arm (HR<br />

[95% CI] 1.15 [0.68-1.96]; p�0.61). Death occurred in 36% <strong>of</strong> the PCRT<br />

arm vs 24% <strong>of</strong> the CRT arm (HR [95% Cl] for OS 1.63 [0.88-3.02];<br />

p�0.12). Disease progression was the cause <strong>of</strong> death in 22% <strong>of</strong> PCRT pts<br />

and 10% <strong>of</strong> CRT pts. There were no differences in outcome by tumor HPV<br />

status. No difference in fatal adverse events (AEs) was seen between arms.<br />

Grade 3� AEs occurred in 85% vs 68% <strong>of</strong> pts (PCRT vs CRT). Differences<br />

in grade 3� toxicity between treatment arms (PCRT, CRT) were most<br />

pronounced for mucosal inflammation (55%, 24%), radiation skin injury<br />

(28%, 13%), dysphagia (40%, 27%), and rash (11%, 0%). RT delays <strong>of</strong><br />

�10 days occurred in 16% <strong>of</strong> the PCRT arm and 3% <strong>of</strong> the CRT arm.<br />

Median cisplatin cumulative dose received was 223.1 mg/m2 in the PCRT<br />

arm and 296.9 mg/m2 in the CRT arm, reflective <strong>of</strong> differences in planned<br />

dose. Conclusions: The addition <strong>of</strong> pmab to CRT did not show an increase in<br />

efficacy and was associated with increased toxicity. Further results <strong>of</strong> HPV<br />

biomarker analysis will be presented.<br />

5501 Oral Abstract Session, Sun, 8:00 AM-11:00 AM<br />

The PARADIGM trial: A phase III study comparing sequential therapy (ST)<br />

to concurrent chemoradiotherapy (CRT) in locally advanced head and neck<br />

cancer (LANHC). Presenting Author: Robert I. Haddad, Department <strong>of</strong><br />

Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School,<br />

Boston, MA<br />

Background: PARADIGM is a multicenter phase III study comparing TPF<br />

(docetaxel, cisplatin, and 5-fluorouracil)-based ST (Arm A) to upfront<br />

cisplatin CRT (Arm B) in patients (pts) with LAHNC. Pt accrual was<br />

terminated in 12/2008 due to slow enrollment with 145 <strong>of</strong> 300 planned<br />

analyzable patients accrued. Safety data was previously presented at the<br />

2010 ASCO annual meeting showing no unusual pattern <strong>of</strong> toxicities in<br />

either arm. Here we present the survival results. Methods: Pts were<br />

randomized to receive arm A-ST (as induction chemotherapy (ICT) with TPF<br />

x 3) followed by CRT with either weekly carboplatin and once daily<br />

radiotherapy, or weekly docetaxel and accelerated boost radiotherapy<br />

based on adequate response to ICT; or arm B - accelerated boost CRT with<br />

bolus cisplatin x 2. The primary endpoint was survival. With original accrual<br />

target <strong>of</strong> 300 analyzable patients, this study was powered at 80% to detect<br />

an improvement in 3-year survival from 55% (arm B) to 70% (arm A).<br />

Results: A total <strong>of</strong> 145 previously untreated pts were enrolled (Arm A: 70;<br />

Arm B: 75), <strong>of</strong> whom 127 were male and 127 were Caucasian. Median age<br />

was 55; patients had PS <strong>of</strong> 0 (97) or 1 (48). Sites <strong>of</strong> disease were<br />

oropharynx: 80, larynx: 24, hypopharynx: 15, and oral cavity: 26. Disease<br />

stages were II (1 pt), III (20 pts) and IV (124 pts). After a median follow-up<br />

<strong>of</strong> 49 months, 41 pts have died (20 in arm A and 21 in arm B). Three-year<br />

survival was 73% (arm A) and 78% (arm B) (HR1.09; 95% CI 0.59 to 2.03<br />

p�0.77). Three-year progression-free survival was 67% (arm A) and 73%<br />

(arm B) (HR 1.2; 95% CI 0.65 to 2.22; p�0.55). Patterns <strong>of</strong> failure will be<br />

presented at the meeting. Conclusions: Althoughthese results suggest no<br />

survival differences between CRT and ST for patients with LAHNC, the<br />

study was terminated before the planned accrual could be reached. HPV<br />

status for the oropharynx cases which represented the majority <strong>of</strong> patients<br />

was not available for stratification; and excellent survival was seen in both<br />

arms.<br />

5503 Oral Abstract Session, Sun, 8:00 AM-11:00 AM<br />

Randomized phase II trial <strong>of</strong> cisplatin and radiotherapy with or without<br />

erlotinib in patients with locally advanced squamous cell carcinoma <strong>of</strong> the<br />

head and neck (SCCHN). Presenting Author: Renato Martins, University <strong>of</strong><br />

Washington, Seattle, WA<br />

Background: The combination <strong>of</strong> cisplatin and radiotherapy is a standard<br />

treatment for patients with locally advanced SCCHN. Cetuximabradiotherapy<br />

is superior to radiotherapy alone in this population, validating<br />

EGFR as a target. Erlotinib, a small molecule inhibitor <strong>of</strong> EGFR, has activity<br />

in recurrent/metastatic disease. Adding EGFR inhibition to standard<br />

cisplatin-radiotherapy may improve efficacy. Methods: Patients with locally<br />

advanced SCCHN were randomized to receive cisplatin 100mg/m2 on days<br />

1,22 and 43 combined with 70Gy <strong>of</strong> radiotherapy (arm A) or the same<br />

chemoradiotherapy combined with erlotinib 150mg/day, starting one week<br />

prior to radiotherapy and continued to its completion (arm B). Randomizing<br />

204 patients had 80% power to compare a 60% complete response rate<br />

(CRR) to a 40% null rate. Available tumors were tested for p16, ERCC1 and<br />

EGFR FISH. Results: Between June 2006 and October 2011, 204 patients<br />

were randomized. Results presented are based on intention to treat. There<br />

were more females on arm A however no other differences in patient<br />

characteristics, including p16 positivity. Patients on arm B had more rash<br />

and gastrointestinal adverse events, but treatment arms did not differ for<br />

rates <strong>of</strong> other grade III/IV toxicities or serious adverse events (SAEs). Arm A<br />

had a CRR <strong>of</strong> 61% and arm B had a CRR <strong>of</strong> 68% (p�0.3). With a median<br />

follow-up <strong>of</strong> 23 months, there were only 29 events in the overall survival<br />

analysis and 46 in the progression-free survival (PFS) analysis. The 2 and 3<br />

year PFS were 71% and 63% for arm A and 78% and 73% for arm B<br />

(p�0.61). Conclusions: The addition <strong>of</strong> erlotinib did not increase the rate <strong>of</strong><br />

SAEs but failed to significantly increase CRR. As seen in other recent trials,<br />

our results in both arms are superior to historical comparisons. Updated<br />

PFS data will be presented. Supported by a grant from the investigatorinitiated<br />

trial program <strong>of</strong> Genentech/OSI.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


5504^ Oral Abstract Session, Sun, 8:00 AM-11:00 AM<br />

Safety and efficacy <strong>of</strong> panitumumab (pmab) in HPV-positive (�) and HPVnegative<br />

(-) recurrent/metastatic squamous cell carcinoma <strong>of</strong> the head and neck<br />

(R/M SCCHN): Analysis <strong>of</strong> the global phase III SPECTRUM trial. Presenting<br />

Author: Jan Stoehlmacher-Williams, University Hospital Carl Gustav Carus,<br />

Dresden, Germany<br />

Background: SPECTRUM evaluated the safety and efficacy <strong>of</strong> pmab, a fully human<br />

monoclonal antibody against the epidermal growth factor receptor, with platinumbased<br />

chemotherapy (CT) vs CT alone in patients (pts) with R/M SCCHN. This<br />

predefined analysis presents outcomes by tumor HPV status. Methods: All tumor<br />

samples were centrally reviewed. HPV status was determined using a validated<br />

immunohistochemistry assay to p16INK4A by an independent lab blinded to<br />

treatment assignments. Tumor samples were scored positive or negative according<br />

to prespecified criteria. Results: Of 657 enrolled pts (ITT), 443 (67%) had samples<br />

evaluable for HPV testing. Ninety-nine (22%) tumors were HPV� and 344 (78%)<br />

were HPV- . HPV� rates varied by site (37% oropharynx, 19% larynx, 15% oral<br />

cavity, and 13% hypopharynx) and geographic region (44% N America, 22% W<br />

Europe, 21% Asia Pacific, 19% S America, and 18% E Europe). Demographics<br />

were generally balanced except pts with HPV� vs HPV- tumors were more <strong>of</strong>ten<br />

non-smokers (31% vs 14%), had oropharyngeal tumors (47% vs 23%), and had<br />

poorly differentiated tumors (30% vs 13%). Efficacy results are shown (Table).<br />

Adverse events were generally balanced between HPV� and HPV- pts. Conclusions:<br />

Pts with HPV- R/M SCCHN administered pmab � CT had improved overall survival<br />

(OS) and progression-free survival (PFS), whereas no improvement in OS or PFS was<br />

observed in pts with HPV� tumors.<br />

ITT<br />

(n�657)<br />

HPV �<br />

(n�99)<br />

HPV -<br />

(n�344)<br />

OS<br />

Events - %<br />

HR<br />

74 73 77<br />

a (95% CI)<br />

P value<br />

0.87 (0.73-1.05) 1.00 (0.61-1.64) 0.76 (0.59-0.97)<br />

b<br />

0.14 0.98 0.02<br />

Median OS (pmab �<br />

CT vs CT) - mos<br />

11.1 vs 9.0 11.0 vs 12.6 11.7 vs 8.6<br />

QITP<br />

PFS<br />

0.253<br />

Events - %<br />

HR<br />

86 91 90<br />

a (95% CI)<br />

p-value<br />

0.78 (0.66-0.92) 1.21 (0.76-1.92) 0.67 (0.53-0.84)<br />

b<br />

0.004 0.43 0.001<br />

Median PFS (pmab �<br />

CT vs CT) - mos<br />

5.8 vs 4.6 5.6 vs 5.5 6.0 vs 5.1<br />

QITP<br />

ORR ITT<br />

(n�566)<br />

0.068<br />

c<br />

HPV �<br />

(n�88) c<br />

HPV -<br />

(n�297) c<br />

ORR (pmab �<br />

CT vs CT) - %<br />

36 vs 25 44 vs 32 35 vs 27<br />

P value odds ratio 0.007 0.27 0.13<br />

Abbreviations: QITP, quantitative interaction test p-value; ORR, objective response rate.<br />

a b c<br />

Stratified hazard ratio. Stratified log-rank test p-value. Pts with baseline measurable<br />

disease per modified RECIST 1.0.<br />

5506 Oral Abstract Session, Sun, 8:00 AM-11:00 AM<br />

PD-1:PD-L1(B7-H1) pathway in adaptive resistance: A novel mechanism<br />

for tumor immune escape in human papillomavirus-related head and neck<br />

cancers. Presenting Author: S<strong>of</strong>ia Lyford-Pike, Johns Hopkins Medical<br />

Institutions, Baltimore, MD<br />

Background: Human papillomavirus-associated head and neck squamous<br />

cell carcinomas (HPV-HNSCC) are associated with a strong host immune<br />

response. Despite ample tumor infiltrating lymphocytes (TILs), the cancer<br />

is able to persist and grow. Here, we provide evidence for an adaptive<br />

immune resistance mechanism mediated through the PD-1:PD-L1 pathway.<br />

Methods: We evaluated the peripheral blood and tumor infiltrating<br />

lymphocytes for PD-1 expression using flow cytometry in HPV-HNSCC<br />

patients. We evaluted PD-L1 expression within the tumors using immunohistochemistry.<br />

We performed functional assays evaluating IFN-gamma<br />

secretion <strong>of</strong> PD-1� and PD-1(-) CD8� T cells isolated from the tumor<br />

microenvironment <strong>of</strong> PD-L1� and PD-L1(-) HPV-related head and neck<br />

cancers. Results: We found the majority <strong>of</strong> CD8� TILs in HPV-HNSCC<br />

express the PD-1 co-inhibitory receptor. We report a striking association<br />

between expression <strong>of</strong> its ligand PD-L1 on tumor cells and tumor associated<br />

macrophages, and the presence <strong>of</strong> TILs. Interestingly, membranous<br />

PD-L1 staining on tumor cells and CD68� antigen presenting cells was<br />

geographically localized to the periphery <strong>of</strong> tumor nests and juxtaposed to<br />

fronts <strong>of</strong> infiltrating T cells. Functional assays demonstrated that PD-1dependent<br />

T cell inhibition required expression <strong>of</strong> PD-L1 in the tumor<br />

microenvironment. Finally, we report localized expression <strong>of</strong> PD-L1 in the<br />

deep crypts <strong>of</strong> normal tonsils, the site <strong>of</strong> HPV infection and origin <strong>of</strong><br />

HPV-HNSCC. Conclusions: Our findings support the role <strong>of</strong> the PD-1:PD-L1<br />

interaction in creating an immune-privileged site for initial viral infection<br />

and subsequently adaptive immune resistance once tumors are established<br />

and suggest a rationale for therapeutic blockade <strong>of</strong> this pathway in patients<br />

with HPV-HNSCC.<br />

Head and Neck Cancer<br />

357s<br />

5505 Oral Abstract Session, Sun, 8:00 AM-11:00 AM<br />

Cetuximab, docetaxel, and cisplatin (TPEx) as first-line treatment in<br />

patients with recurrent or metastatic (R/M) squamous cell carcinoma <strong>of</strong> the<br />

head and neck (SCCHN): Final results <strong>of</strong> phase II trial GORTEC 2008-03.<br />

Presenting Author: Joel Guigay, Department <strong>of</strong> Medical Oncology, Institut<br />

Gustave Roussy, Villejuif, France<br />

Background: Cetuximab in combination with platinum and 5FU (PFEx) has<br />

become a standard in first-line treatment <strong>of</strong> patients (pts) with R/M<br />

SCCHN. Cetuximab and taxane combinations have shown promising<br />

activity. This multicenter phase II study evaluates the efficacy and safety <strong>of</strong><br />

cetuximab, docetaxel and cisplatin combination (TPEx) as first-line treatment<br />

in pts with R/M SCCHN. Methods: Pts were required to have WHO PS<br />

0-1, no prior systemic therapy for R/M SCCHN, cumulative dose <strong>of</strong> cisplatin<br />

less than 300 mg/m² and no prior anti-EGFR therapy. Pts received<br />

docetaxel 75 mg/m² day 1, cisplatin 75 mg/m² day 1 and cetuximab (400<br />

mg/m² on day 1 <strong>of</strong> cycle 1 then 250 mg/m² weekly), repeated every 21 days<br />

x 4 cycles then followed by cetuximab 500 mg/m² every 2 weeks (wks) as<br />

maintenance therapy until disease progression or unacceptable toxicity.<br />

G-CSF support with lenograstim 150 �g/m²/day was delivered after each<br />

cycle <strong>of</strong> chemotherapy. Response was assessed every 6 wks, according to<br />

RECIST. The primary endpoint was objective response rate (ORR) at 12<br />

wks. Secondary endpoints were safety, best overall response, progressionfree<br />

survival (PFS), overall survival (OS) and biomarkers. Results: 54 pts<br />

have been enrolled: 52 males, median age 57.8years (28-69), 12 (22.2%)<br />

oropharynx, 55% metastatic.48 pts could be evaluated for ORR at 12 wks:<br />

partial response, stable disease and progression were respectively found in<br />

23 pts (PR 47.9%), 21 pts (SD 43.7%) and 4 pts (PD 8.3%). Best overall<br />

ORR was 54% (1 CR, 27 PR). The median PFS and OS were 7.1 and 15.3<br />

months, respectively. The 1-year OS was 58.6%. Median PFS after start <strong>of</strong><br />

maintenance therapy was 4.2 months. Toxicity was manageable with<br />

G-CSF support. Treatment related toxicities included G4 neutropenia<br />

(n�3). Grade 3 events were skin rash (n�5), hypersensitivity reaction<br />

(n�3), mucositis (n�1), fatigue (n�1) and febrile neutropenia (n�1).<br />

Alopecia was common. 1 toxic death was related to sepsis on feeding tube.<br />

Conclusions: Efficacy analysis demonstrates that TPEx regimen is effective<br />

and might be a relevant substitute for PFEx as first-line treatment in fit pts<br />

with R/M SCCHN.<br />

5507 Oral Abstract Session, Sun, 8:00 AM-11:00 AM<br />

Intra-arterial infusion <strong>of</strong> recombinant adenoviral human p53 gene combined<br />

chemotherapy for advanced oral cancer: A phase II double-blind<br />

randomized clinical trial. Presenting Author: Yi Li, Department <strong>of</strong> Head and<br />

Neck Oncology, West China Hospital <strong>of</strong> Stomatology, Sichuan University,<br />

Chengdu, China<br />

Background: The p53 tumor suppressor gene has been shown as having<br />

multiple anti-tumor functions and being capable <strong>of</strong> enhancing the efficacy<br />

<strong>of</strong> chemo-therapy. This study is to investigates clinical outcomes <strong>of</strong><br />

intra-arterial infusion <strong>of</strong>recombinant adenoviral human p53 gene (rAdp53)<br />

combined with chemotherapy in treatment <strong>of</strong> advanced oral cancer.<br />

Methods: Ninety-nine patients with stage III or IV oral carcinoma, satisfying<br />

with the inclusion criteria, were randomly allocated to Group I (G1: n�35;<br />

rAd-p53 plus chemotherapy), Group II (G2: n�33; rAd-p53), or Group III<br />

(G3: n�31; chemotherapy). Intra-artery infusion <strong>of</strong> rAd-p53 were given<br />

through superficial temporal artery reverse intubation at a dose 1-4 �1012 viral particles (VP) , once every 3 days for 6 times. Chemotherapy regimen<br />

consisted <strong>of</strong> oxaplatin 200 mg/m2 on day1, bleomycin 8 mg/m2 on day 2,<br />

6, 8 and Methotrexate 30 mg/m2 on day1, 8, for squamous cell carcinoma<br />

(SCC), and oxaplatin 200 mg/m2 on day1, 5-Fu 200 mg/m2 on day1,3 and<br />

cyclophosphamide 400 mg/m2 on day2, 8 for adenoid cystic carcinoma<br />

(ACC). Results: The complete response rate (CR) in G1 (48.5%) was<br />

significantly higher than in G2 (16.7%) and G3 (17.2%). The overall<br />

response rates (RR) were 88.6%, 54.5%, and 51.6% for G1, G2 and G3,<br />

respectively. The 3-year overall survival (OS) <strong>of</strong> G1 (82.9%) was significantly<br />

higher than the patients in G2 (60.6%) and G3 (58.1%). All those<br />

patients receiving rAd-p53 had a self-limited fever. Positive Bax immunostaining<br />

was detected in all the patients receiving rAd-p53. Conclusions:<br />

Intra-arterial infusion <strong>of</strong> combined rAd-p53 and chemotherapy may represent<br />

an alternative to the current standard treatment for the late stage oral<br />

carcinoma.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


358s Head and Neck Cancer<br />

5508 <strong>Clinical</strong> Science Symposium, Mon, 11:30 AM-1:00 PM<br />

An international, double-blind, randomized, placebo-controlled phase III<br />

trial (EXAM) <strong>of</strong> cabozantinib (XL184) in medullary thyroid carcinoma<br />

(MTC) patients (pts) with documented RECIST progression at baseline.<br />

Presenting Author: Patrick Sch<strong>of</strong>fski, Department <strong>of</strong> General Medical<br />

Oncology, University Hospitals Leuven, Leuven, Belgium<br />

Background: MTC arises from parafollicular cells <strong>of</strong> the thyroid gland,<br />

accounts for 5-8% <strong>of</strong> thyroid cancers and represents an unmet medical<br />

need. Cabozantinib (cabo) is an oral inhibitor <strong>of</strong> MET, VEGFR2, and RET.<br />

We conducted a phase III study <strong>of</strong> cabo vs placebo (P) in pts with<br />

progressive, unresectable, locally advanced or metastatic MTC. Methods:<br />

Eligible pts were required to have documented RECIST progression within<br />

14 months <strong>of</strong> screening. The primary efficacy measure was progression-free<br />

survival (PFS) as assessed by an independent review facility (IRF) using<br />

RECIST. Secondary efficacy measures included objective response rate<br />

(ORR) and overall survival (OS). The study has 90% power to detect a 75%<br />

increase in PFS and 80% power to detect a 50% increase in OS. Tumor<br />

assessments occurred every 12 weeks. Crossover between treatment arms<br />

was not allowed. Results: Between Sept 2008 and Feb 2011, 330 pts<br />

(median age 55 yrs; 67% male; 96% measureable disease; RET mutation<br />

status: pos 48%; neg 12%; unknown 39%; prior TKI exposure: yes 21%,<br />

no 78%, unknown 2%) were randomized 2:1 to cabo (140 mg free base<br />

[175 mg salt form] qd; n�219) or P (n�111). The planned primary PFS<br />

analysis included events through the date <strong>of</strong> the 138th event. As <strong>of</strong><br />

15June2011, 44.7% <strong>of</strong> pts on cabo and 13.5% on P were still receiving<br />

study treatment. Statistically significant PFS prolongation <strong>of</strong> 7.2 mo was<br />

observed; median PFS for cabo was 11.2 mo vs 4.0 mo for P (HR 0.28,<br />

95% CI 0.19-0.40, p�0.0001). PFS results favored the cabo group across<br />

subset analyses including RET status and prior TKI use. ORR was 28% for<br />

cabo vs 0% for P (p�0.0001). An interim analysis <strong>of</strong> OS (44% <strong>of</strong> the 217<br />

required events) did not show a difference between cabo and P. The most<br />

frequent grade �3 adverse events (cabo vs P) were diarrhea (15.9 vs<br />

1.8%), palmar-plantar erythrodysesthesia (12.6 vs 0%), fatigue (9.3 vs<br />

2.8%), hypocalcemia (9.3 vs 0%), and hypertension (7.9 vs 0%).<br />

Conclusions: This phase III study met its primary objective <strong>of</strong> demonstrating<br />

substantial PFS prolongation with cabo vs. P in a patient population with<br />

MTC and documented progressive disease in need <strong>of</strong> therapeutic intervention.<br />

5510 <strong>Clinical</strong> Science Symposium, Mon, 11:30 AM-1:00 PM<br />

A molecular diagnostic panel for thyroid cancer disease management.<br />

Presenting Author: Shih-min Cheng, Quest Diagnostics Nichols Institute,<br />

San Juan Capistrano, CA<br />

Background: In 2009, the <strong>American</strong> Thyroid Association revised its guidelines<br />

for patients with thyroid nodules and differentiated thyroid cancers,<br />

recommending BRAF, RAS, RET/PTC, and PAX8-PPAR� molecular testing<br />

in patients with indeterminate fine-needle aspirate (FNA) cytology. Alterations<br />

in any <strong>of</strong> these markers in thyroid nodules are strongly associated with<br />

malignancy. We studied the prevalence <strong>of</strong> these alterations in thyroid FNA<br />

specimens in order to establish a strategy to improve disease management.<br />

Methods: DNA and RNA were extracted from thyroid FNA specimens<br />

(N�149) and tested for BRAF mutations using allele-specific PCR (V600E<br />

and K601E); for RAS (H-, K-, N-) mutations using pyrosequencing (codons<br />

12, 13, and 61); and for RET/PTC1 and RET/PTC3 rearrangements and<br />

PAX8-PPAR� translocations using RT-PCR. Results: Overall, 90 (60.4%) <strong>of</strong><br />

the specimens had alterations in �1 <strong>of</strong> the 4 molecular markers (Table).<br />

BRAF V600E mutations (54.4%) were the most prevalent mutations in<br />

papillary thyroid cancer (PTC); no K601E mutations were found. RAS<br />

mutations were found in PTC, follicular adenoma (FA), and follicular<br />

thyroid cancer (FTC) specimens; half <strong>of</strong> these mutations involved N-RAS<br />

Q61. RET/PTC rearrangements were detected in 3.5% <strong>of</strong> PTC specimens<br />

and were evenly distributed between the 2 major subtypes, RET/PTC1 and<br />

RET/PTC3. PAX8/PPAR� translocations were detected in 18.8% <strong>of</strong> FTC<br />

but not in FA, probably due to small sample size. Out <strong>of</strong> 7 indeterminate<br />

thyroid nodules, 2 had BRAF mutations and 2 had RAS mutations. The<br />

presence <strong>of</strong> the 4 markers was generally mutually exclusive; only 1 PTC<br />

specimen had concurrent RAS and RET/PTC1 alterations. Conclusions: The<br />

prevalence <strong>of</strong> BRAF, RAS, RET/PTC, and PAX8/PPAR� alterations in<br />

thyroid cancer specimens highlights the potential for targeted therapeutic<br />

strategies. The mutually exclusive pattern <strong>of</strong> alterations also suggests a<br />

hierarchical screening strategy for samples with limited availability.<br />

Prevalence <strong>of</strong> marker alterations in thyroid FNA specimens (NT � not<br />

tested).<br />

Diagnosis #<br />

BRAF<br />

(%)<br />

RAS<br />

(%)<br />

RET/PTC<br />

(%)<br />

PAX8/PPAR�<br />

(%)<br />

Total<br />

(%)<br />

PTC 114 62 (54.4) 11 (9.6) 4 (3.5) NT 77 (67.5)<br />

FA 12 NT 2 (16.7) NT 0 2 (16.7)<br />

FTC 16 NT 5 (31.3) NT 3 (18.8) 8 (50%)<br />

Indeterminate 7 2 (28.6) 2 (28.6) 0 0 4 (57.1)<br />

5509^ <strong>Clinical</strong> Science Symposium, Mon, 11:30 AM-1:00 PM<br />

Reacquisition <strong>of</strong> RAI uptake in RAI-refractory metastatic thyroid cancers by<br />

pretreatment with the MEK inhibitor selumetinib. Presenting Author: Alan<br />

Loh Ho, Memorial Sloan-Kettering Cancer Center, New York, NY<br />

Background: Therapies for radioactive iodine (RAI)-refractory thyroid cancers<br />

<strong>of</strong> follicular origin (TC-FCO) are desperately needed. TC-FCO mouse<br />

models show that blocking mitogen-activated protein kinase (MAPK)<br />

signaling with a MAP kinase kinase 1/2 (MEK1/2) inhibitor increases<br />

sodium iodide symporter expression and iodine incorporation. This 20<br />

patient (pt) pilot study aimed to evaluate if the MEK1/2 inhibitor selumetinib<br />

(AZD6244, ARRY-142866) can reverse RAI-refractoriness in<br />

TC-FCO pts (NCT00970359). Methods: RAI-refractory TC-FCO disease was<br />

defined as: 1) non-RAI-avid lesion/s on a diagnostic or post-therapy RAI<br />

scan, 2) RAI-avid lesion/s that was stable or increased in size after RAI<br />

therapy, or 3) fluorodeoxyglucose (FDG)-avid lesion/s by positron emission<br />

tomography (PET) scan. rhTSH-stimulated lesional dosimetry with 124I PET<br />

was performed prior to and after 4 weeks <strong>of</strong> treatment with selumetinib (75<br />

mg orally bid). If the second 124I PET scan predicted a lesional RAI dose <strong>of</strong><br />

� 2000 cGy, therapeutic RAI was administered while on the drug. Primary<br />

endpoints were to evaluate changes in tumor iodine uptake and RECIST<br />

response after RAI therapy; changes in serum thyroglobulin (TG) after RAI<br />

was a secondary endpoint. Results: 24 pts were enrolled, 22 eligible, and<br />

20 evaluable. For the 20 evaluable pts, median age was 61 (range 44-77<br />

yrs), 11 were men. 19 pts had tumors analyzed for BRAF and N-,K- RAS<br />

mutations. 8 pts had BRAF mutant (MUT), 11 BRAF wild-type (WT)<br />

tumors; 1 pt to be analyzed. Selumetinib increased 124I uptake in 12 <strong>of</strong> the<br />

20 pts (4 <strong>of</strong> 8 BRAF MUT; 8 <strong>of</strong> the 12 other pts). The 8 <strong>of</strong> those 12 pts<br />

achieving sufficient iodine avidity to warrant RAI therapy included all 5 pts<br />

known to be NRAS MUT to date and 1 BRAF MUT pt. Of the 7 pts who have<br />

received RAI, 5 had partial responses (PRs); 2 stable disease (SD). Mean<br />

percent reduction in TG in this group (pre- vs 2 months post- RAI) was<br />

91%. No � grade 2 CTCAE toxicities attributable to selumetinib were<br />

observed. 1 pt was diagnosed with myelodysplastic syndrome � 51 weeks<br />

after RAI (unrelated to selumetinib). Conclusions: Selumetinib can enhance<br />

iodine uptake in a subset <strong>of</strong> RAI-refractory TC-FCO and may be<br />

particularly effective for RAS MUT tumors.<br />

5511 Poster Discussion Session (Board #1), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

A multicenter randomized controlled trial (RCT) <strong>of</strong> adjuvant chemotherapy (CT)<br />

in nasopharyngeal carcinoma (NPC) with residual plasma EBV DNA (EBV DNA)<br />

following primary radiotherapy (RT) or chemoradiotherapy (CRT). Presenting<br />

Author: Anthony T. C. Chan, Department <strong>of</strong> <strong>Clinical</strong> Oncology, Prince <strong>of</strong> Wales<br />

Hospital, The Chinese University <strong>of</strong> Hong Kong, Shatin, Hong Kong<br />

Background: The benefit <strong>of</strong> adjuvant CT in NPC is unclear. Administering CT after<br />

full-dose CRT presents challenges in treatment compliance and toxicity. Post-RT<br />

EBV DNA predicts poor survival and may be a biomarker <strong>of</strong> subclinical residual<br />

disease. We conducted a biomarker driven RCT using EBV DNA to select high<br />

risk NPC patients (pts) for adjuvant CT while sparing low risk pts from<br />

unnecessary toxicity. Methods: Biopsy proven NPC, AJCC stage IIB-IVB, detectable<br />

EBV DNA at 6-8 wks post-RT, no persistent locoregional disease or distant<br />

metastasis, ECOG 0 or 1, adequate organ function. Randomised with stratification<br />

for primary therapy (RT Vs CRT) and tumor stage (II/III Vs IV) to arm A<br />

(adjuvant cisplatin 40 mg/m2 and gemcitabine 1000mg/m2, both given on<br />

D1�8 q3w x 6 cycles) or arm B (clinical follow-up). EBV DNA and PET scan were<br />

performed before and 6 months after randomization. Primary endpoint was<br />

relapse free survival and secondary endpoints included toxicity <strong>of</strong> adjuvant CT.<br />

With a hazard ratio <strong>of</strong> 2, 100 pts were required with a power <strong>of</strong> 0.8 and an alpha<br />

at 0.05. This safety analysis was approved by DMSC. Results: From 9/2006 to<br />

12/2011, 514 pts consented for EBV DNA screening, 95 with detectable EBV<br />

DNA consented for adjuvant study. After work-up, 74 were eligible for randomization<br />

(37 to arm A; 37 to arm B). The two arms were well balanced in baseline<br />

characteristics. 80% received prior neoadjuvant and/or concurrent CT. Staging:<br />

IIB (36.5%), III (29.7%), IVA (18.9%), IVB (14.8%). Five pts refused adjuvant<br />

CT after randomization. Overall 65% and 57% completed 5 and 6 cycles<br />

respectively. Mean dose intensity (DI): 84% for cisplatin (22.5 mg/m2/wk, range<br />

0.0-26.7), 92% for gemcitabine (612.8 mg/m2/wk, range 333.3-777.8).<br />

Treatment related adverse events above CTC G2 were summarized in Table.<br />

Conclusions: Delivery <strong>of</strong> 6 cycles <strong>of</strong> adjuvant CT is feasible with acceptable<br />

toxicity after full dose RT or CRT.<br />

Events (No) G3 G4 G5<br />

Non-hematological<br />

Bleeding 1 1<br />

Electrolytes 2 1<br />

Fatigue 1<br />

Hearing 3<br />

Infection 1<br />

Mucositis 1<br />

syncope 2<br />

Weight loss 1<br />

Hematological<br />

Febrile neutropenia 1<br />

Hemoglobin 10<br />

Neutrophils 14 10<br />

Platelets 2<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


5512 Poster Discussion Session (Board #2), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Five years’ results <strong>of</strong> the German ARO 04-01 trial <strong>of</strong> concurrent 72 Gy<br />

hyperfractionated accelerated radiation therapy (HART) plus once weekly<br />

cisplatinum/5-FU versus mitomycin C/5-FU in stage IV head and neck<br />

cancer. Presenting Author: Volker Budach, Department <strong>of</strong> Radiooncology<br />

CCM/CVK, Charite University Medicine, Berlin, Germany<br />

Background: Are 6 cycles <strong>of</strong> once weekly DDP plus one cycle <strong>of</strong> 5-FU with<br />

concurrent HART superior to 2 cycles <strong>of</strong> MMC plus one cycle <strong>of</strong> 5-FU in<br />

terms <strong>of</strong> overall survival (OS) and metastases-free survival (MFS)? Methods:<br />

Eligibility: Stage IV SCC <strong>of</strong> oro(OP)- and hypopharynx(HP), KFS <strong>of</strong> �80%<br />

stratified for sites, N-status, grading, hemoglobin and center. The HART<br />

schedule was reported elsewhere (V.Budach, JCO 23;2005). HART was<br />

applied concurrently with DDP/5-FU at 30mg/m² days 1,8,15,22,29,36 or<br />

MMC/5-FU at 10mg/m2 day 1�36 and for both arms with 600mg/m² 5-FU<br />

days 1-5 as 120 hrs. c.i. TVD dose prescription was 72 Gy using<br />

3D-conformal or IMRT-TP. 364 patients were analysed using an ITT<br />

principle for OS, MFS, progression-free survival (PFS) and loco-regional<br />

control (LRC). Hazard ratio (HR) calculations were adjusted for competing<br />

risk factors. Results: Median follow-up was 48 mos. for both arms. Mean<br />

age was 55.4 years, 83/17% were male/female and 100% stage IV patients<br />

(UICC 2002). 58.5%/41.5% <strong>of</strong> all patients suffered from OP- or HP<br />

cancer, respectively. The OS and MFS at 4 years for the DDP- versus<br />

MMC-arm was 42.1% vs. 38.8% (n.s.) and 67.3% vs. 56.6% (p�.05),<br />

respectively. The LRC and PFS for the DDP versus MMC-arm was 58.6% vs.<br />

57.2% (n.s.) and 46.4% vs. 38.7% (n.s.). Seven items recorded for acute<br />

toxicity and 9 for late morbidity showed no significant differences between<br />

the treatment arms except for creatinine for the DDP-arm (p � 0.001)<br />

using nonparametric analyses <strong>of</strong> variances for repeated measurements. The<br />

overall compliance rates for RTX were 96%, DDP: 72%, 5-FU: 97%, MMC:<br />

86%, respectively. Conclusions: This phase III trial first establishes level<br />

IB-evidence for a once weekly DDP chemoradiation regimen. For MFS at 4<br />

yrs., DDP/5 FU-HART is superior to MMC/5 FU HART at equal levels <strong>of</strong><br />

acute and late radiation sequelae for both treatment arms. No significant<br />

differences were seen yet for OS, PFS or LRC. Chemoradiation with weekly<br />

DDP/5-FU or MMC/5-FU shows excellent compliance rates and can easily<br />

compete with other concurrent chemo- or bio-radiation schedules including<br />

induction TPF followed by radiation.<br />

5514 Poster Discussion Session (Board #4), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

A phase II study <strong>of</strong> temsirolimus/sorafenib in patients with radioactive<br />

iodine (RAI)-refractory thyroid carcinoma. Presenting Author: Eric Jeffrey<br />

Sherman, Memorial Sloan-Kettering Cancer Center, New York, NY<br />

Background: Sorafenib is an oral tyrosine kinase inhibitor <strong>of</strong> multiple<br />

targets, including RAF, VEGFR1, and VEGFR2. Published response rates<br />

with sorafenib in RAI refractory thyroid cancer have ranged from 11-23%.<br />

Temsirolimus is an intravenous inhibitor <strong>of</strong> mTOR; mutations in the<br />

PI3K/mTOR/AKT pathway occur late in thyroid cancer and may be<br />

associated with aggressive disease. Methods: The study was a single<br />

institution, phase II design. Primary objective was response rate. Eligible<br />

patients (pts) had progressive, RAI-refractory/fluorodeoxyglucose (18-F)avid,<br />

recurrent/metastatic, non-medullary, thyroid cancer; RECIST measurable<br />

disease; and adequate organ/marrow function. Sorafenib was given at<br />

200 mg orally twice a day and temsirolimus at 25 mg intravenously weekly.<br />

38 patients were enrolled; 37 were eligible and 36 patients started<br />

treatment between 12/18/09 – 10/20/11. Data cut<strong>of</strong>f date is 1/19/12.<br />

Fourteen pts are still actively on study. Results: Of the 37 eligible pts,<br />

demographics: female-46%; median age-64 years (30-81); histologypapillary<br />

(23)/follicular (1)/Hürthle (5)/poorly differentiated (6)/anaplastic<br />

(2); prior systemic treatment (21); prior sorafenib (6). Grade 4-5 adverse<br />

events at least possibly related to drug: grade 5 – sudden death, NOS (1 pt):<br />

grade 4 – colonic perforation (1 pt). Evaluation <strong>of</strong> best response: partial<br />

(PR) (8, including 3 with anaplastic/poorly differentiated); stable disease<br />

(SD) (21); progression (1); inevaluable (7). The partial response rate was<br />

38% in the cohort that previously did not receive any systemic treatment.<br />

There was no correlation <strong>of</strong> response to either BRAF (10) or RAS (5)<br />

mutational status. Median time on treatment is 203 days (range 14-638<br />

days) at the cut<strong>of</strong>f date. For the 21 pts with SD, the maximum percentage<br />

shrinkage by RECIST criteria ranged from -2% to -35% (one patient with<br />

unconfirmed PR) with a median time on treatment <strong>of</strong> 203 days (range<br />

39-503 days). Conclusions: The combination <strong>of</strong> sorafenib and temsirolimus<br />

shows promising results in a heavily pretreated population. This study was<br />

approved and funded by the National Comprehensive Cancer Network<br />

(NCCN) from general research support from Pfizer, Inc.<br />

Head and Neck Cancer<br />

359s<br />

5513 Poster Discussion Session (Board #3), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Cetuximab/radiotherapy (CET�RT) versus concomitant chemoradiotherapy<br />

(cCHT�RT) with or without induction docetaxel/cisplatin/5-fluorouracil<br />

(TPF) in locally advanced head and neck squamous cell carcinoma<br />

(LASCCHN): Preliminary results on toxicity <strong>of</strong> a randomized, 2x2 factorial,<br />

phase II-III study (NCT01086826). Presenting Author: Maria Grazia Ghi,<br />

Medical Oncology Department, Venezia, Italy<br />

Background: The standard treatment options for LASCCHN are cCHT�RT or<br />

CET�RT. Strategies to improve the efficacy with the integration <strong>of</strong><br />

induction chemotherapy are being investigated. Primary endpoints <strong>of</strong> this<br />

study were to compare: 1) the overall survival (OS) <strong>of</strong> induction vs. no<br />

induction arms; 2) the Grade(G)3-4 in-field toxicity <strong>of</strong> cCHT�RT vs.<br />

CET�RT. Methods: Patients (pts) with unresectable LASCCHN, stage<br />

III-IV, ECOG PS 0–1 were randomized to a 2x2 factorial design: Arm A1:<br />

cCHT�RT (2 cycles <strong>of</strong> ciplatin/5fluorouracil); Arm A2: CET�RTX; Arm B1:<br />

3 cycles <strong>of</strong> TPF followed by the same cCHT�RT; Arm B2: 3 cycles <strong>of</strong> TPF<br />

followed by CET�RT. A total <strong>of</strong> 204 deaths over 420 pts ( including the<br />

101 randomized in the phase II part <strong>of</strong> the study comparing cCHT�RT with<br />

or w/o induction TPF) were required to detect a HR <strong>of</strong> death <strong>of</strong> 0.675<br />

(A1�A2 vs. B1�B2; 2-sided a�0.05; b�0.20) and a 10% difference in<br />

G3-4 in-field mucosal toxicity (A1�B1 vs. A2�B2). Results: By February<br />

2012, 387 pts over 413 pts were evaluable for toxicity. 82% <strong>of</strong> pts were<br />

male; median age was 60y; PS: 0�77.8% and 1�22.2%. Disease stage<br />

was III (31%) or IV (69%). Sites <strong>of</strong> disease were oral cavity (21.7%),<br />

oropharynx (54.8%), hypopharynx (23.5%). At a median follow-up <strong>of</strong> 21<br />

months, 126 deaths occurred. Data on G3-4 in-field toxicity (primary<br />

endpoint) and compliance to cCHT�RT vs CET�RT are shown in the table.<br />

Conclusions: No advantage for CET�RT over cCHT�RT was observed<br />

regarding G3-4 in-field toxicities and feasibility. Pts are still being<br />

followed-up to assess OS.<br />

cCHT�RTX CET�RTX p<br />

In-field mucositis G3-4 29.4% 23% 0.15<br />

In-field skin reaction G3-4 11.1% 13.7% 0.43<br />

RT median dose, Gy (range) 66 (18-70.2) 70 (35-70.4) �0.01<br />

RT median duration, weeks (range) 7.2 (0.6-13.3) 8.4 (1-12.7) �0.01<br />

RT interruption > 3days 32.8% 38.7% 0.28<br />

Pts receiving 2 CHT cy or 7 weekly CET 91.3% 78.2% �0.01<br />

RT modification due to acute toxicity 18.5% 27.7% 0.08<br />

5515 Poster Discussion Session (Board #5), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Expression <strong>of</strong> p16, ERCC1, and EGFR amplification as predictors <strong>of</strong><br />

responsiveness <strong>of</strong> locally advanced squamous cell carcinomas <strong>of</strong> head and<br />

neck (SCCHN) to cisplatin, radiotherapy, and erlotinib: A phase II randomized<br />

trial. Presenting Author: Melissa Austin, University <strong>of</strong> Washington,<br />

Seattle, WA<br />

Background: This study evaluated the expression <strong>of</strong> p16 and ERCC1 and<br />

EGFR amplification as predictive and prognostic factors in a recently<br />

completed phase II randomized trial comparing cisplatin (100 mg/m2 on d<br />

1, 22, 43) with radiotherapy (70Gy) �/- erlotinib (150 mg/d during<br />

radiotherapy) in locally advanced SCCHN. p16 (HPV surrogate) is a known<br />

prognostic factor in oropharyngeal SCCHN, and high ERCC1 expression has<br />

been correlated with resistance to cisplatin. EGFR gene copy number may<br />

be prognostic in SCCHN and alter response to erlotinib. Methods: Pretreatment<br />

formalin-fixed, paraffin-embedded tumor tissue was available for<br />

84/204 patients. EGFR copy number was assessed using Vysis LSI DNA<br />

probes, and immunohistochemistry was performed on Leica Bond III<br />

machines using Lab Vision ERCC-1 (8F1) and CINtec p16 antibodies. All<br />

assays were performed in accordance with manufacturer instructions; all<br />

studies were reviewed by a single Pathologist (MA) who was blinded to<br />

patient outcome. Logistic regression and Cox regression models fit a<br />

treatment arm by marker interaction and tested selected linear contrasts.<br />

Results: Efficacy analysis showed no difference in complete response rate<br />

(CRR) and progression-free survival (PFS) between treatment arms. However,<br />

PFS in both arms was better than historical comparisons, with only 46<br />

events over a median follow-up <strong>of</strong> 23 months. Positivity in p16 was<br />

associated with greater CRR (OR 3.5, p�0.03) and longer PFS (HR�0.38;<br />

p� 0.07). The CRR effect was greater for the erlotinib arm (OR 8.1,<br />

p�0.01) than for Arm A (OR 1.5, p�0.56). The ERCC1 (�) rate was 46%<br />

(39/84).ERCC1 expression above the median was not associated with<br />

worse CRR (OR 0.69; p�0.46) or PFS (HR 0.99; p�0.98). Only 4 tumors<br />

showed EGFR amplification precluding further analysis. Conclusions: p16(�)<br />

tumors had a better outcome, similar to other recent trials, and erlotinib<br />

seemed to increase the CRR among p16(�) tumors. ERCC1 expression did<br />

not predict chemoradioresistance in this study. EGFR amplification was too<br />

rare in the tested population to assess predictive or prognostic value.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


360s Head and Neck Cancer<br />

5516^ Poster Discussion Session (Board #6), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Cilengitide with cetuximab, cisplatin, and 5-FU in recurrent and/or metastatic<br />

squamous cell cancer <strong>of</strong> the head and neck: The ADVANTAGE phase<br />

II trial. Presenting Author: Jan Baptist Vermorken, Department <strong>of</strong> Medical<br />

Oncology, Antwerp University Hospital, Edegem, Belgium<br />

Background: Prognosis for patients with recurrent and/or metastatic squamous<br />

cell cancer <strong>of</strong> the head and neck (R/M-SCCHN) is very poor. �v�5<br />

integrin is overexpressed in SCCHN and selective integrin blockade is being<br />

investigated as a treatment strategy. Methods: ADVANTAGE was a phase I/II<br />

study evaluating cilengitide with cetuximab and platinum-based chemotherapy.<br />

Eligible patients were �18 years with �1 measurable lesion,<br />

Karn<strong>of</strong>ski performance status �70%, and confirmed R/M-SCCHN. The<br />

phase II part reported herein was an open-label, randomized, controlled<br />

trial investigating progression-free survival (PFS) in patients treated with 2<br />

cilengitide 2000 mg regimens: once weekly (CIL1W) and twice weekly<br />

(CIL2W) administration combined with cisplatin, 5-FU, and cetuximab<br />

(PFE) as compared with PFE alone (control). Secondary objectives included<br />

overall survival (OS) and objective response (OR). Treatment-emergent<br />

adverse events (TEAEs) were monitored. Results: 184 eligible patients were<br />

enrolled. Overall, patient characteristics were similar across arms. Median<br />

duration <strong>of</strong> treatments was shorter in the CIL2W arm compared with the<br />

CIL1W arm (cilengitide: 16.1 vs 23.4 wk, cetuximab: 16.0 vs 22.6 wk,<br />

platinum: 16.0 vs 18.0 wk, and 5-FU: 14.6 vs 18.0 wk). Median PFS was<br />

6.4 months in the CIL1W group, 5.6 months in the CIL2W group and 5.7<br />

months in the control group, with CIL1W and CIL2W having HRs <strong>of</strong> 1.03<br />

(95% CI: 0.67–1.59) and 1.55 (95% CI: 0.99–2.43) vs control. The latter<br />

HR was possibly due to less compliance in the CIL2W arm. Median OS was<br />

12.4, 10.6, and 11.6 months in the CIL1W, CIL2W, and control groups,<br />

respectively. ORs were observed in 46.8%, 26.7%, and 35.5% <strong>of</strong> patients<br />

in the CIL1W, CIL2W, and control arms, respectively. Most common<br />

(�15%) grade 3/4 TEAEs were neutropenia, hypokalemia, leukopenia,<br />

stomatitis, and fatigue. No safety differences were noted between treatment<br />

arms. Overall, there were 7 drug-related TEAEs (2 in CIL1W, 2 in<br />

CIL2W, and 3 in control) leading to death. Conclusions: In this population,<br />

neither <strong>of</strong> the cilengitide-containing regimens was able to demonstrate a<br />

PFS benefit over PFE alone.<br />

5518 Poster Discussion Session (Board #8), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Lenvatinib treatment <strong>of</strong> advanced RAI-refractory differentiated thyroid<br />

cancer (DTC): Cytokine and angiogenic factor (CAF) pr<strong>of</strong>iling in combination<br />

with tumor genetic analysis to identify markers associated with<br />

response. Presenting Author: Douglas Wilmot Ball, The Johns Hopkins<br />

University School <strong>of</strong> Medicine, Baltimore, MD<br />

Background: Lenvatinib is an oral tyrosine kinase inhibitor targeting<br />

VEGFR1-3, FGFR1-4, RET, KIT and PDGFR�. In a phase II study <strong>of</strong><br />

lenvatinib, 58 patients (pts) with DTC were enrolled and a response rate <strong>of</strong><br />

50% was observed (Sherman, ASCO 2011). Methods: Pts received lenvatinib<br />

at a starting dose <strong>of</strong> 24 mg oral once daily in 28-day cycles. Serum<br />

was collected at baseline (BL), on day 8 and on day 36 and concentrations<br />

<strong>of</strong> 47 CAFs were measured using multiplex bead arrays and enzyme-linked<br />

immunosorbent assay (ELISA). 33 genes (443 mutations) were examined<br />

in archival tumor tissues (n�25). Association <strong>of</strong> baseline CAF, changes in<br />

CAF levels upon treatment and gene mutation status with treatment<br />

outcomes was investigated. Results: Combination <strong>of</strong> low baseline VEGF and<br />

ANG-2 (p�0.02 and HR�0.386) correlated with longer PFS. Both<br />

baseline and changes in CAF levels demonstrated an association with gene<br />

mutation status. High baseline levels <strong>of</strong> VEGF were observed in pts with<br />

wild type RAS and BRAF (p�0.035) whereas high baseline sTIE-2 levels<br />

associated with RAS mutation (p�0.023). Supporting pre-clinical mouse<br />

xenograft tumor model developed using ANG2-overexpressing human<br />

thyroid cancer cell line FTC-286 demonstrated reduced sensitivity to<br />

lenvatinib treatment. Increased levels <strong>of</strong> IL-10 and FGF-2 8-day posttreatment<br />

(8d post-tx) significantly associated with RAS (N- or K-) and<br />

BRAF mutation. Combination <strong>of</strong> gene mutation status with baseline CAF<br />

levels provided better prediction <strong>of</strong> longer PFS compared to gene mutation<br />

alone. Hierarchical clustering modeling using changes in CAF levels 8d<br />

post-tx with tumor shrinkage identified a set <strong>of</strong> CAFs that could predict two<br />

categories <strong>of</strong> lenvatinib response: longer PFS and greater tumor shrinkage<br />

or longer PFS in the absence <strong>of</strong> significant tumor shrinkage. Conclusions:<br />

CAF pr<strong>of</strong>iling identified potential predictive biomarkers <strong>of</strong> lenvatinib<br />

treatment outcomes in DTC. Combination <strong>of</strong> RAS and BRAF mutation with<br />

baseline VEGF and ANG-2 or treatment-associated changes in FGF-2 and<br />

IL-10 level correlated with lenvatinib treatment response.<br />

5517 Poster Discussion Session (Board #7), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Genomic pr<strong>of</strong>iling <strong>of</strong> a clinically annotated cohort <strong>of</strong> locoregionally advanced<br />

head and neck cancers (HNC) treated with definitive chemoradiotherapy.<br />

Presenting Author: Tanguy Y. Seiwert, The University <strong>of</strong> Chicago<br />

Medical Center, Chicago, IL<br />

Background: Recent advances in genomic technology and sequencing have<br />

allowed the near comprehensive characterization <strong>of</strong> genetic abnormalities<br />

in HNC, but their correlation with clinical outcome remains to be determined.<br />

We analysed a clinically annotated cohort <strong>of</strong> locoregionally advanced<br />

HNC for copy number changes and mutations and correlated with<br />

outcome in an exploratory fashion. Methods: 120 head and neck cancers<br />

(frozen tissue) and matching normal tissues/blood from patients with<br />

locoregionally advanced HNC treated with definitive chemoradiation �/induction<br />

at the University <strong>of</strong> Chicago were obtained from our tissue bank.<br />

DNA, RNA, protein were extracted. Barcoded, multiplexed sequencing<br />

libraries for 500 head and neck cancer associated genes (Agilent capture)<br />

were built and sequenced on Illumina HiSeq next generation sequencers.<br />

150 commonly copy number altered and HNC relevant genes were analysed<br />

for copy number alterations (CNA) using the Nanostring nCounter platform.<br />

Respective genetic aberrations in curated pathways were allocated to tumor<br />

subgroups. Exploratory analysis correlated data with induction response<br />

and progression free survival. Confirmatory HPV testing was performed<br />

using an E6, nested, multiplex PCR. Results: �80% <strong>of</strong> targeted sequences<br />

were successfully captured and sequenced. Commonly mutated genes<br />

included: TP53, CDKN2A, PIK3CA, NOTCH1. Of note PIK3CA mutations<br />

were enriched in HPV(�) tumors. Commonly copy number altered genes<br />

included amplifications <strong>of</strong> VEGF1, CCND1, MYC, EGFR, MDM4, CTTN, as<br />

well as PIK3CA, and deletions <strong>of</strong> CDKN2A, SUCLG2, FHIT, TGFBR2,<br />

PTPRD, IKBKG, TRAF6, and RASSF1. CCND1, and MYC were commonly<br />

co-amplified and correlated with poor progression free survival, representing<br />

a distinct HNC phenotype. Analysis for additional genes and influence<br />

on induction response will be presented. Conclusions: Targeted, medium<br />

throughput genomic pr<strong>of</strong>iling is feasible, and provides the majority <strong>of</strong> HNC<br />

specific genetic aberrations at a comparably low cost. Impact on outcome<br />

(CCND1/MYC amplification) and potential treatment targets (PIK3CA<br />

mutations in HPV(�) tumors) were identified.<br />

5519 Poster Discussion Session (Board #9), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Safety, molecular, and imaging responses to cetuximab administered in a<br />

window pre-operative study in squamous cell carcinoma <strong>of</strong> the head and<br />

neck (SCCHN). Presenting Author: Sandra Schmitz, Department <strong>of</strong> Head<br />

and Neck Surgery, St-Luc Hospital, Catholic University <strong>of</strong> Louvain, Brussels,<br />

Belgium<br />

Background: Only a minority <strong>of</strong> SCCHN pts benefits from anti-EGFR mAbs.<br />

Trials with pre- and post-therapy tumor biopsies are crucial to better<br />

characterize the molecular pathways involved in treatment response or<br />

resistance. Targeted agents (TA) are <strong>of</strong>ten investigated in unselected<br />

end-stage cancer pts, making difficult optimal translational research. One<br />

way to resolve this issue is to perform “window” studies where a TA is given<br />

during the incompressible period between the diagnosis and surgery.<br />

Methods: We conducted a phase I/II study: cetuximab (C) was given<br />

pre-operatively to treatment-naïve SCCHN pts selected for primary curative<br />

surgery. C (400mg/m2 first wk followed by 250mg/m2/wk) was given<br />

during 2 wks (day -15 until day -1, 3 infusions) before surgery (day 0).<br />

Tumour biopsies, FDG/PET, and CT were performed at diagnosis and<br />

surgery. In the phase I, we investigated the safety <strong>of</strong> preoperative C by<br />

progressively reducing the delay between the last dose <strong>of</strong> C and surgery<br />

(minimum delay : 24 hrs ; 3�3 phase 1 design). The aims <strong>of</strong> the phase II<br />

were (i) safety, (ii) C activity by FDG-PET (Po�0.10, P1�0.35, a�0.05 and<br />

b� 0.10). As controls, 5 additional pts were included without C treatment<br />

but with the same requirements regarding biopsies and imaging. Results:<br />

The phase 1 study (n�18) demonstrated that C infusion given 24 hrs<br />

before surgery was safe. Safety was confirmed in the phase II (n�20). 90 %<br />

had a FDG-PET partial response (PR) (EORTC guideline) in the C group and<br />

0% in the controls. 52% had a �SUVmax decrease <strong>of</strong> �50%. 2 pts<br />

evaluable by CT/MRI had a PR (RECIST). Then, we compared the pre-and<br />

post treatment biopsies by immunochemistry. For the whole group, C did<br />

not reduce Ki67 (p�0.1) and did not seem to induce apoptosis (caspase).<br />

However, for pts with �SUVmax decrease � 25% or � 50%, Ki67 was<br />

decreased (p�0.04 and 0.006). C induced downregulation <strong>of</strong> pEGFR<br />

(p�0.0004) and pERK (p�0.007) but not pAKT/AKT (Histoscore).<br />

Conclusions: Pre-operative study with C is safe. Our findings suggest that<br />

the main downstream molecular pathway blocked by C in SCCHN is the<br />

RAS/RAF/ERK. Further analyses are ongoing to better characterize molecular<br />

response and escape mechanisms.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


5520 Poster Discussion Session (Board #10), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Role <strong>of</strong> c-MET pathway in the outcome prediction <strong>of</strong> cetuximab-based<br />

therapy in patients with recurrent or metastatic squamous cell carcinoma <strong>of</strong><br />

the head and neck. Presenting Author: Victoria Casado, Oncology Department.<br />

Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid,<br />

Spain<br />

Background: Activation <strong>of</strong> the c-MET oncogene promotes tumor growth,<br />

invasion and metastasis in several tumor types. In addition, c-MET has<br />

been reported in consort with EGFR and/or be activated as a compensatory<br />

pathway in the presence <strong>of</strong> EGFR blockade resulting a mechanism <strong>of</strong><br />

acquired resistance to EGFR inhibitors in lung and colorectal carcinoma.<br />

We investigated the impact on cetuximab sensitivity <strong>of</strong> HGF, c-MET, c-MET<br />

activation and c-MET gene status in recurrent or metastatic squamous cell<br />

carcinoma <strong>of</strong> the head and neck (HNSCC) patients. Methods: A singleinstitution<br />

retrospective analysis including 50 HNSCC patients was carried<br />

out. For each case, formalin-fixed tumor specimens were assayed for HGF,<br />

total and phosphorylated Y1234/35 c-MET (p-c-MET) by immunohistochemistry<br />

and c-MET gene by FISH. Overexpression criteria were defined by ROC<br />

curves for each protein and amplification was defined by �2 copies in at<br />

least two <strong>of</strong> the three studied tumor areas. Results were analyzed for<br />

association with clinico-pathological features and survival outcomes for<br />

cetuximab-treated patients (median follow-up 75 months). Results: c-MET<br />

overexpression was detected in 26 (52%) <strong>of</strong> patients, c-MET amplification<br />

in 15 (30%) and p-c-MET in 12 (24%). Amplification was associated with<br />

c-MET overexpression (p�0.004). HGF overexpression was observed in 8<br />

(12%) associated with c-MET phosphorylation (p�0.001), suggesting a<br />

paracrine activation <strong>of</strong> receptor. No significant association with clinicopathological<br />

parameters was detected. Log-rank test showed significantly<br />

worse outcome in c-MET overexpressing patients for progression-free (PFS)<br />

(p�0.002) and overall survival (OS) (p�0.045); p-c-MET was correlated<br />

with worse PFS (p�0.014) and OS (p�0.001). In multivariate logistic<br />

regression analysis, p-c-MET was an independent prognostic factor for PFS<br />

(HR 6.5; 95% CI 1.5-8.9). Conclusions: HGF/c-MET pathway correlated<br />

with worse outcome in cetuximab-based regimen treated HNSCC patients,<br />

acting as a resistance mechanism for EGFR inhibitors and supporting a<br />

dual blocking HGF/c-MET and EGFR pathways for treatment <strong>of</strong> these<br />

patients.<br />

5522 Poster Discussion Session (Board #12), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Exploratory pharmacodynamics (PD) trial to investigate the mechanism <strong>of</strong><br />

action <strong>of</strong> RG7160 (GA201), a novel dual-acting, monoclonal antibody<br />

(mAb) designed to enhance antibody-dependent cellular cytotoxicity (ADCC),<br />

compared with cetuximab (C) in neoadjuvant head and neck squamous cell<br />

carcinoma (HNSCC). Presenting Author: Stéphane Temam, Institut Gustave<br />

Roussy, Villejuif, France<br />

Background: GA201 efficacy was superior to C in orthotopic xenograft<br />

models in terms <strong>of</strong> activity and PD. In a phase I study, objective responses<br />

and long lasting stable disease were observed in heavily pre-treated<br />

patients (pts). Methods: HNSCC pts (T2-4, any N, M0) received i.v.<br />

neo-adjuvant 700/1400 mg GA201 or C (as per SPC), on D1 and 8. Tumor<br />

biopsies were taken at baseline (BL) and pre-surgery. The primary objective<br />

was determination <strong>of</strong> changes in tumor immune cell infiltration after<br />

treatment. Peripheral CD3�, CD16�, CD56� cells, plasma cytokines,<br />

tumor EGFR and 18F-FDG-PET avidity were also studied. Results: To date,<br />

39 pts (15/10 vs 14 pts for 700/1400 GA201 vs C, respectively) were<br />

evaluable for the primary objective and 4 pts are on-going. Patient<br />

characteristics at BL were as follows: median age, 57/62 vs 62 years;<br />

gender, 11/10 vs 12 male; tumor size � 4 cm, 7/3 vs 3 pts. A greater<br />

proportion <strong>of</strong> pts receiving GA201 compared to C had metabolic tumor<br />

reduction (� 25% <strong>of</strong> SUVmax) (10/15 at 700 mg [1 pathological CR], 7/10<br />

at 1400 mg GA201 vs 7/14 C pts) and symptomatic relief (6/12 at 700 mg,<br />

6/7 at 1,400 mg GA201 vs 2/10 C pts). Table shows changes in PD<br />

markers. Conclusions: GA201 treatment, compared to C, exhibited more<br />

extensive tumor immune cell infiltration and a greater metabolic response.<br />

Consistent with the proposed mechanism <strong>of</strong> action <strong>of</strong> GA201, peripheral<br />

NK cells dropped and a unique cytokine pr<strong>of</strong>ile was observed. The data<br />

support the immunomodulatory superiority <strong>of</strong> GA201 - a mAb designed to<br />

enhance ADCC compared to C.<br />

Median change from BL (range)<br />

GA201<br />

C ‡<br />

700 mg* 1,400 mg †<br />

Tumor biopsy 1<br />

CD3� (cells/mm 2 ) 396 (–987, 2336) 205 134 (–45, 722)<br />

EGFR (intensity 3; %) –14 (–83, 25) – –5.5 (–65, 43)<br />

Peripheral blood<br />

CD16� and/or CD56�/CD3– % –97 (–100, –92) –98 (–100, –97) –1 (–71, 59)<br />

CD3� % –89 (–98, –73) –93 (–96, –78) –59 (–86, 178)<br />

IL-15 % 946 (0, 2113) 695 (649, 742) 0 (–53, 7)<br />

IL-1RA % 6,950 (706, 21,933) 1,201 (873, 1,529) –16 (–70, 19)<br />

*n� 14-15; † n � 1-2; ‡ n � 8-12 for paired samples.<br />

1<br />

Correlations with SUVmax are being investigated.<br />

Head and Neck Cancer<br />

361s<br />

5521 Poster Discussion Session (Board #11), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Differential expression <strong>of</strong> EGFR, HER2, P16, and high-risk (hr) HPV status<br />

in oropharyngeal (Or) and oral cavity (OC) squamous cell carcinoma (SCC).<br />

Presenting Author: Anne Sudaka, Centre Antoine Lacassagne, Nice, France<br />

Background: We aimed to better characterize the expression <strong>of</strong> epidermal<br />

growth factor receptors family in Or and OC SCC and correlate it to hr HPV<br />

status and anatomoclinical features. Methods: In formalin fixed paraffin<br />

embedded tumor tissues, chromogenic in situ hybridization (CISH) was<br />

performed to detect hr HPV and immunohistochemistry to evaluate EGFR<br />

(positive score: 4 to 6), HER2 (0�negative, 1�positive) and P16 (positive<br />

score:�70% labeled cells) expression. Baseline data were collected and<br />

analyze will be used. Results: Among 128 pts, 69 were tobacco users and<br />

58 male. Median age at diagnosis was 61 y [23-95]. EGFR, Her2, P16<br />

expression and hrHPV positivity were seen in 84 (65%), 12 (9%), 47<br />

(37%) and 47 (37%) respectively. P16 signal was linked with absence <strong>of</strong><br />

tobacco (31% vs. 72%, Pearson chi2 test: p�10-3 ), absence <strong>of</strong> alcohol<br />

(48% vs. 83%, p�10-3 ), Or site (53% vs. 76%, p�0.005), T1 (34% vs.<br />

19%, p�0.06), N0 (52% vs. 23%, p�0.003), absence <strong>of</strong> nodal capsular<br />

rupture (85% vs. 68%, p�0.02), WHO grade 3 (37% vs. 68%, p�0.001),<br />

hr HPV detection (17% vs. 70%, p�10-3 ). Multivariate analysis confirmed<br />

the link between P16 expression and hr HPV CISH positivity (OR�0.05, CI<br />

95% [0.01-0.21], p�10-3 ) , absence <strong>of</strong> tobacco (OR�9.2, CI 95%<br />

[2.1-40.7], p�0.03) and N0 (OR�0.2, CI 95% [0.04- 0.75], p�0.02).<br />

EGFR signal was linked with tobacco (31% vs 78%,p�10-3 ), alcohol (57%<br />

vs 81%, p�0.02), OC localization (77% vs 58%, p�0.03), well differentiated<br />

SCC (73% WHO grade 1-2 vs 56% grade 3, p�0.04), absence <strong>of</strong> hr<br />

HPV detection (74% vs 52%, p�0.01) and absence <strong>of</strong> P16 labeling (78%<br />

vs 44%, p�10-3 ). Multivariate analysis confirmed the link between EGFR<br />

positivity and tobacco (OR�0.32, CI 95% [0.1-0.9], p�0.03), alcohol<br />

(OR�9.5, CI 95% [1.2- 72.8], p�0.03) and OC localization (OR�0.3, CI<br />

95% [0.1-1], p�0.05). HER2 signal was linked with alcohol (5% vs 18%,<br />

p�0.04), history <strong>of</strong> tobacco associated neoplasia (10% vs 0%, p�0.02),<br />

Or site (14% vs 2%, p�0.03) and who grade 3 (5% vs 14%, p�0.06).<br />

Her2 labeling was not associated with tobacco, sex, hr HPV detection, P16<br />

positivity or EGFR one. Conclusions: Hr HPV associated SCC have a low<br />

expression <strong>of</strong> EGFR and are not associated with HER2 labeling.<br />

5523 Poster Discussion Session (Board #13), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Differences in biomarker expression in HNSCC according to p53 status.<br />

Presenting Author: Rebecca Anne Feldman-Moreno, Caris Life Sciences,<br />

Phoenix, AZ<br />

Background: Patients with p53 wildtype head and neck squamous cell<br />

carcinoma (HNSCC) tend to be HPV-positive, which associates with better<br />

prognosis. The purpose <strong>of</strong> this study was to explore biomarker expression<br />

pr<strong>of</strong>iles for insight into molecular differences in HNSCC patients based on<br />

p53 status. Methods: TP53 gene sequencing using the AmpliChip p53<br />

microarray (Roche Molecular Systems, Inc.) was attempted on 61 HNSCC<br />

patients previously tested with Caris Target Now tumor pr<strong>of</strong>iling service.<br />

DNA was extracted from a FFPE sample, amplified and processed on the<br />

AmpliChip p53 microarray to detect single base pair substitution and<br />

deletion mutations in exons 2-11andtheir flanking splice sites in the<br />

TP53 gene (GenBank X54156). EGFR FISH , HER2 IHC and 22 other<br />

predictive biomarkers, e.g. TS, TOPO2A, MGMT, etc., were assayed and<br />

retrospectively analyzed. All tests were performed in a CLIA-certified lab<br />

and interpreted by board-certified pathologists or cytogeneticists. Statistical<br />

analysis was performed using SPSS (PASW statistics17) for parametric<br />

and non-parametric tests <strong>of</strong> independence. Results: 52 cases provided<br />

sufficient quality DNA for p53 analysis and results revealed a mutation rate<br />

<strong>of</strong> 25% in HNSCC patients. Interestingly, only EGFR FISH and HER2 IHC<br />

(p�.002 and p�.004, respectively) were differentially expressed in<br />

wildtype vs. mutated p53. Matched-pair analysis in the p53 mutated<br />

subgroup (n�13) showed no significant trend regarding EGFR status<br />

(p�.763) but a slight trend towards HER-2 negativity (p�.020). In the p53<br />

wildtype subgroup (n�39), a strong association with EGFR FISH nonamplification<br />

(n�28, 71.8%, p�.001) as well as HER-2 negativity (n�38,<br />

97.4%, p�.001) was shown. Conclusions: To our knowledge, this is the first<br />

analysis <strong>of</strong> differential biomarker expression pr<strong>of</strong>iles in HNSCC based on<br />

p53 status. We hypothesize that the absence <strong>of</strong> EGFR amplification in the<br />

p53 wildtype cancers may be a contributing factor to the improved<br />

prognosis observed in HPV-positive HNSCC. Additionally, the strong<br />

association between p53 wildtype HNSCC patients and EGFR nonamplification<br />

suggests EGFR-targeted therapies like cetuximab would<br />

likely fail in p53 wildtype patients.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


362s Head and Neck Cancer<br />

5524 Poster Discussion Session (Board #14), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

New DNA methylation markers associated with oral cancer (OC) development<br />

(dvlpt). Presenting Author: Pierre Saintigny, University <strong>of</strong> Texas M. D.<br />

Anderson Cancer Center, Houston, TX<br />

Background: We have reported that DNA methyltransferase-3B (DNMT3B)<br />

gene expression is a strong predictor <strong>of</strong> OC dvlpt. Using high-throughput<br />

DNA methylation pr<strong>of</strong>iles <strong>of</strong> oral preneoplastic lesions (OPL), we identified<br />

86 candidate genes that were hypermethylated in patients (pts) developing<br />

OC and suggested that a CpG island methylation phenotype may occur early<br />

during OC dvlpt. Our aim was to validate some <strong>of</strong> the candidate genes and<br />

to study the value <strong>of</strong> LINE1 repetitive element methylation, a surrogate <strong>of</strong><br />

global DNA methylation, as biomarkers <strong>of</strong> risk to develop OC. Methods:<br />

Validation cohort included 40 pts with a median follow-up <strong>of</strong> 4.2 years,<br />

including 14 pts who developed OC. None <strong>of</strong> them were used in the<br />

discovery phase. Frozen OPL were collected prospectively and subject to<br />

DNA extraction. We performed a quantitative analysis <strong>of</strong> the degree <strong>of</strong><br />

methylation <strong>of</strong> LINE1, and AGTR1, FOXI2, PENK, and HOXA9 promoters<br />

CpG sites using pyrosequencing. Results: The degree <strong>of</strong> methylation <strong>of</strong><br />

AGTR1 (Mann-Whitney P�0.0001), FOXI2 (P�0.0002), PENK<br />

(P�0.0001), but not HOXA9 (P�0.0714) was significantly higher in pts<br />

who developed OC. On the contrary, LINE1 methylation was significantly<br />

lower in pts who developed OC (P�0.0001). The median percent <strong>of</strong><br />

methylation was used to dichotomize the pts into high versus low methylation<br />

levels. Oral cancer-free survival (OCFS) was significantly worse in pts<br />

with high levels <strong>of</strong> AGTR1 (Log-rank P�0.0229), FOXI2 (P�0.0170), and<br />

PENK (P�0.0142) promoter methylation. No significant difference was<br />

found with HOXA9. A Methylation Index (MI) was developed by averaging<br />

the percent <strong>of</strong> methylation <strong>of</strong> AGTR1, FOXI2, and PENK promoters; pts<br />

with a high MI had a worse OCFS (P�0.0031). On the other hand, pts with<br />

low levels <strong>of</strong> LINE1 methylation had a significantly worse OCFS (P�0.0118).<br />

Finally, in 26 pts, a positive correlation was observed between DNMT3B<br />

gene expression levels and the MI (rho�0.65, P�0.0003); surprisingly,<br />

there was a negative correlation with LINE1 methylation (rho�-0.74,<br />

P�0.0001). Conclusions: AGTR1, FOXI2 and PENK promoter methylation<br />

may be associated with increased OC risk in pts with OPL and correlate with<br />

DNMT3B expression. Global DNA hypomethylation is an early event in OC<br />

dvlpt.<br />

5526 Poster Discussion Session (Board #16), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Effects <strong>of</strong> low-level laser therapy in the prevention and treatment <strong>of</strong><br />

concurrent chemoradiotherapy induced oral mucositis: A triple-blind randomized<br />

controlled trial. Presenting Author: P.U. Prakash Saxena, Department<br />

<strong>of</strong> Radiotherapy & Oncology, Kasturba Medical College, Udupi, India<br />

Background: Oral mucositis (OM) is the most cumbersome acute side effect<br />

<strong>of</strong> concurrent chemoradiotherapy (CCRT) for head and neck cancer (HNC).<br />

OM associated pain affects oral function also may lead to CCRT break.<br />

Several modalities have been tried to prevent and treat this complication<br />

but none has been proved successful till date. We used prophylactic Low<br />

Level Laser Therapy (LLLT) for the prevention and treatment <strong>of</strong> CCRT<br />

induced OM. Methods: In this triple blind study, 221 HNC patients<br />

scheduled to undergo CCRT were randomized into laser (n�111) and<br />

placebo (n�110) group. Laser group received LLLT (Helium Neon laser,<br />

��632.8nm, P�24m, W�3.0J/cm2 ) while placebo received sham treatment<br />

daily prior to radiation for 45days. Oral Mucositis (OM) (RTOG/EORTC<br />

Scale), oral pain (Visual Analog Scale - VAS), dysphagia (Functional<br />

Impairment Scale - FIS), weight loss, duration <strong>of</strong> analgesic use were<br />

assessed. Data was analyzed using frequencies and percentage, generalized<br />

estimating equations (GEE) and Odds ratio. Results: There was<br />

significant reduction in incidence <strong>of</strong> severe OM (F�16.64, degree <strong>of</strong><br />

freedom df�8,876, p�0.0001) and its associated pain (F�25.06,<br />

df�8,876, p�0.0001) and dysphagia (F�20.17, df�8,876, p�0.0001)<br />

and loss <strong>of</strong> weight (F�87.56, df�8,876, p�0.0001) when compared<br />

between the Laser and placebo group. The duration <strong>of</strong> step III analgesia<br />

required was also significantly lower (p�0.001) in laser (3.2 �1.4days)<br />

than placebo (6.7 �2.6days) group. Conclusions: LLLT was able to<br />

decrease the incidence <strong>of</strong> CCRT induced severe OM its associated pain,<br />

dysphagia, weight loss and the duration <strong>of</strong> analgesic use.<br />

Statistical analysis <strong>of</strong> OM and FIS scores between laser and placebo group.<br />

Week<br />

Laser group<br />

OR (95% CI)<br />

Oral mucositis Functional Impairment Scale<br />

Placebo group<br />

OR (95% CI)<br />

Laser group<br />

OR (95% CI)<br />

Placebo group<br />

OR (95% CI)<br />

7 19.05 (9.78-37.12) 8.18 (3.03-22.05) 12 (6.77-21.2) 5.24 (3.15-8.72)<br />

6 12.72 (6.73-24.04) 5.17 (1.83-14.58) 9.5 (5.45-16.53) 4.7 (2.85-7.74)<br />

5 7.06 (3.88-12.83) 3.86 (1.37-10.84) 6.1 (3.62-10.28) 3.89 (2.35-6.45)<br />

4 3.65 (2.05-6.49) 3.24 (1.27-8.27) 2.7 (1.74-4.2) 2.48 (1.57-3.91)<br />

3 Ref Ref Ref Ref<br />

5525 Poster Discussion Session (Board #15), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Hearing evaluation <strong>of</strong> head and neck cancer patients (HNCP): Comparison<br />

<strong>of</strong> adverse event (AE) criteria. Presenting Author: A. Dimitrios Colevas,<br />

Stanford University School <strong>of</strong> Medicine, Stanford, CA<br />

Background: The incidence and severity <strong>of</strong> baseline hearing abnormality in<br />

HNCP is poorly characterized, as is the subsequent CDDP associated<br />

hearing loss. While formal behavioral audiograms can characterize CDDP<br />

associated hearing loss in great detail, most oncologists either rely on<br />

subjective evaluation <strong>of</strong> hearing loss or grade hearing impairment using the<br />

CTCAE. Two new grading systems for grading cisplatin ototoxicity have<br />

been developed for use in pediatrics, the Brock and Chang systems. This<br />

report is the first evaluation <strong>of</strong> these 3 hearing loss criteria in an adult<br />

cancer patient population. Methods: We conducted an electronic medical<br />

records (EMR) search for all HNCP between 2004 and 2010 treated at the<br />

Stanford Cancer Institute (SCI) and identified which patients had audiograms<br />

in the EMR, and who had received CDDP. Three investigators (RL,<br />

ADC, KC) independently graded these audiograms using the CTCAE v3 and<br />

v4, Brock and Chang criteria. Baseline distributions, changes following<br />

CDDP dosing, and inter-observer variability were calculated for hearing<br />

impairment grading using all four criteria. Results: We evaluated 460<br />

audiograms in 111 patients. Because 282/460 <strong>of</strong> the audiograms did not<br />

have threshold shifts measured at 1,2,3,4,6 and 8 kHz, CTCAE v4 could<br />

not be applied to them . We therefore limited all further comparisons to<br />

CTCv3, Brock and Chang criteria. The table below summarizes the analysis.<br />

Conclusions: The Brock and Chang ototoxity criteria scales deliver a<br />

distribution <strong>of</strong> hearing AE grades at baseline more compatible than the<br />

CTCAE v3 with respect to the expectation that most HNCP do not have<br />

severe baseline hearing loss. Both the Brock and Chang scales are more<br />

sensitive than the CTCv3 to CDDP associated hearing changes. The Brock<br />

scale, followed by the Chang scale, demonstrated highest inter-rater<br />

agreement. Additional data concerning performance characteristics and<br />

utility <strong>of</strong> these three grading systems will be presented. Replacement <strong>of</strong> the<br />

present hearing impairment criteria in the CTCAE with either the Brock or<br />

Chang scales should be considered.<br />

Ototoxicity scale CTCAE v3 Brock Chang<br />

Baseline median grade 2 0 0<br />

% inter-rater agreement 69 91 80<br />

% with grade changes following CDDP 33 44 47<br />

5527 Poster Discussion Session (Board #18), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

ARIX: A randomized trial <strong>of</strong> acupuncture versus oral care sessions in<br />

patients with chronic radiation-induced xerostomia following treatment for<br />

head and neck cancer. Presenting Author: Richard Simcock, Sussex Cancer<br />

Centre, Brighton, United Kingdom<br />

Background: Radiation induced xerostomia impairs quality <strong>of</strong> life <strong>of</strong> in head<br />

and neck cancer survivors. Prior small non-randomised studies suggest that<br />

acupuncture may alleviate symptoms. We report the first large randomised<br />

trial comparing 8 weeks <strong>of</strong> group acupuncture with oral care sessions as a<br />

control. Methods: 145 patients with radiation induced xerostomia�18<br />

months were recruited from 7 UK Cancer centres. Subjects received<br />

standardised group sessions <strong>of</strong> oral care (OC) education and 8 sessions <strong>of</strong><br />

weekly group acupuncture (A) in a randomised crossover design with a 4<br />

week washout (Group One OC/A and Group Two A/OC). Acupuncture was<br />

standardised to a technique previously piloted (3 auricular points and two<br />

distal points bilaterally). Patients completed the EORTC QLQ H&N questionnaire<br />

at baseline and weeks 5, 9, 13, 17 and 21 in addition to rating 4 key<br />

dry mouth symptoms. The primary outcome was patient reported improvement<br />

in dry mouth symptoms. The statistical analysis was longitudinal<br />

based on logistic regression models and adjustment for potential carry over.<br />

Stimulated and unstimulated saliva was measured. Results: An improvement<br />

for 5/7 symptoms was noted following acupuncture. In Group One<br />

24% showed improvement 8 weeks following A (19% after OC). In Group<br />

Two 26% reported improvement in severe dry mouth after A (14% after<br />

OC). The estimated OR <strong>of</strong> improved dry mouth 5 weeks after A compared to<br />

OC was 2.0 (p�0.031), after adjusting for effects <strong>of</strong> time, potential<br />

carry-over, centre and patient characteristics. The estimated odds ratios <strong>of</strong><br />

improvement in severity <strong>of</strong> symptoms for A compared to OC were (OR�1.67,<br />

p�0.048) for sticky saliva, (OR�2.08, p�0.011) for needing to sip to<br />

swallow food and (OR�1.71, p�0.013) for waking up at night needing to<br />

drink. Mean attendance rate for A was 89% and 80% for OC. There was no<br />

significant change in saliva production over time or by intervention. Mean<br />

global QoL score did not change significantly over time either within or<br />

between groups. No adverse events were seen. Conclusions: Eight sessions<br />

<strong>of</strong> weekly group acupuncture provides significantly better relief <strong>of</strong> radiation<br />

induced xerostomia than group oral care education.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


5528 Poster Discussion Session (Board #19), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

A phase II trial <strong>of</strong> cetuximab in high-risk premalignant lesions <strong>of</strong> the upper<br />

aerodigestive tract. Presenting Author: Joseph A Califano, Johns Hopkins<br />

University School <strong>of</strong> Medicine, Baltimore, MD<br />

Background: High risk head and neck premalignancy includes patients with<br />

prior HNSCC and loss <strong>of</strong> heterozygosity (LOH). These patients demonstrate<br />

30-60% rates <strong>of</strong> progression to malignancy despite conventional surgical<br />

excision. We performed a phase II trial using short term therapy with<br />

cetuximab, a humanized monoclonal antibody directed against EGFR, in<br />

the treatment <strong>of</strong> high risk premalignant lesions <strong>of</strong> the UADT. Methods:<br />

Inclusion criteria required; 1) presence <strong>of</strong> 3p, 9p21, or 17p LOH, and/or 2)<br />

surgically unresectable high grade premalignant lesions, and/or 3) high<br />

grade premalignancy after curative therapy for HNSCC. Patients received<br />

cetuximab 400 mg/m2 on week one followed by 250 mg/m2 on week 2-8 or<br />

observation, with the option for crossover to cetuximab for patients<br />

originally randomized to the observation arm. Histologic grade (1�benign,<br />

2�mild dysplasia, 3�moderate dysplasia, 4�severe dysplasia/carcinoma<br />

in situ, 5�invasive cancer) and change in grade <strong>of</strong> dysplasia was evaluated.<br />

Results: 19 patients were enrolled. Two patients discontinued therapy due<br />

to toxicity. We noted a decrease in grade <strong>of</strong> dysplasia in the cetuximab<br />

treated group (-1.0) vs. the observation group (-0.2), (Wilcoxon test p value<br />

� 0.18). In the observation group, 0/5 patients (0%) underwent complete<br />

resolution <strong>of</strong> dysplasia; while 4/12 (33.3%) treated patients had no<br />

remaining dysplasia after therapy (p � 0.26), three <strong>of</strong> these patients had<br />

complete resolution <strong>of</strong> dysplastic changes after cetuximab therapy alone.<br />

Of these three patients, two have had no recurrence <strong>of</strong> leukoplakia or<br />

dysplasia at 1.5 and 2 years. One patient treated with oral cavity dysplasia<br />

has had no recurrence <strong>of</strong> dysplasia or lesions within the oral cavity, but<br />

developed a hypopharyngeal cancer at 1.5 years after trial completion. Of 5<br />

patients randomized to observation, three underwent crossover to cetuximab<br />

therapy, and two <strong>of</strong> these three patients had complete resolution <strong>of</strong><br />

dysplasia. Conclusions: EGFR blockade with cetuximab alone can result in<br />

significant, durable, and complete clinical and histological resolution <strong>of</strong><br />

moderate to severe dysplasia in at least a subset <strong>of</strong> high risk patients.<br />

5530 Poster Discussion Session (Board #21), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

The influence <strong>of</strong> institutional head and neck cancer (HNC) clinical trial<br />

accrual on overall survival (OS): An analysis <strong>of</strong> RTOG 0129. Presenting<br />

Author: Evan John Wuthrick, Ohio State University Medical Center-James<br />

Cancer Hospital, Columbus, OH<br />

Background: NCCN recommends HNC patients (pts) be treated by providers<br />

with experience in HNC management. Whether treatment by experienced<br />

providers (measured by no. <strong>of</strong> pts treated) is associated with HNC survival<br />

outcome is unknown. Methods: Analysis included 471 pts in RTOG 0129<br />

with stage III-IVa HNC with known tumor HPV and smoking status<br />

randomized to cisplatin concurrent with standard or accelerated fractionation<br />

radiotherapy (RT). <strong>Part</strong>icipating centers were stratified into pt accrual<br />

tertiles (ATs) to 21 RTOG HNC trials from 1997-2002. As a surrogate for<br />

experience, we investigated the independent effect <strong>of</strong> AT on OS and<br />

progression-free survival (PFS) in multivariable cox proportional hazards<br />

models. Results: Median follow up was 4.8 yrs (range 1.1 - 6.5). In<br />

Kaplan-Meier analysis, OS and PFS for pts at centers in the two lower AT<br />

(low accruing centers [LAC]) were similar and compared to the upper AT<br />

(high accruing centers [HAC]). Median accrual in LAC vs HAC was 11 vs 82<br />

pts. Pts treated at LAC and HAC had similar age, HPV status, pack-years, N<br />

stage, comorbidities and study arm assignment (SAA), but pts at LAC had<br />

better performance status (PS)(Zubrod 0; 62% vs 52%; p�0.04), fewer T4<br />

tumors (27% vs. 35%; p�0.05) and were more likely uninsured (12% vs<br />

4%; p�0.009). Compared to HAC, LAC pts had significantly worse OS, (5<br />

yr 51.0% vs 69.1%; p � 0.002), and PFS (5 yr 42.7% vs 61.8%; p �<br />

0.001) and more locoregional failure (5 yr 36.4% vs 20.8%; p �0.001).<br />

Treatment at LAC was associated with ~90% increase in risk <strong>of</strong> death (OS<br />

HR 1.91, 95%CI 1.37-2.65) and progression (PFS HR 1.89, 95%CI<br />

1.39-2.56) when compared to pts treated at HAC, after adjustment for age,<br />

PS, pack-years, T and N stage, SAA and HPV status. Acute and chronic<br />

toxicities, RT dose, fractions, and treatment duration did not differ for pts<br />

at LAC vs HAC. RT at LAC was more likely than HAC to differ from protocol<br />

(18% [16.8% acceptable variation {AV}, 1.2% unacceptable deviation<br />

{UD}] vs 6% [4.7% AV, 1.3% UD]; p�0.001). Conclusions: Pts with HNC<br />

treated at LAC in RTOG trials were associated with significantly worse<br />

survival outcomes compared to pts treated at HAC.<br />

Head and Neck Cancer<br />

363s<br />

5529 Poster Discussion Session (Board #20), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Prognostic significance <strong>of</strong> the AJCC staging in patients with squamous cell<br />

carcinoma <strong>of</strong> the oropharynx. Presenting Author: Carlos Rodrigo Acevedo-<br />

Gadea, Yale University School <strong>of</strong> Medicine, New Haven, CT<br />

Background: There have been significant changes in the epidemiology <strong>of</strong><br />

head and neck squamous cell carcinomas (HNSCC), with an increase in the<br />

incidence <strong>of</strong> oropharyngeal (OP) cancer and opposite effect in other sites.<br />

Since the rise in OP cancer incidence is attributed to human papillomavirus<br />

(HPV), which is associated with a different biology and clinical behavior, we<br />

evaluated whether the current AJCC system retained its prognostic impact<br />

in this patient population. Methods: The Surveillance Epidemiology and<br />

End Results (SEER) registry was queried for patients with HNSCC diagnosed<br />

between 2004 and 2007. Overall survival (OS) was estimated by the<br />

Kaplan-Meier method and the Cox model was used to compare the survival<br />

curves for each AJCC stage. Patients were grouped into three anatomical<br />

locations: oral cavity (OC), larynx (L) and OP. Results: There were 26,520<br />

patients meeting eligibility criteria, including 8622 OP, 7332 OC, and<br />

10566 L. The AJCC staging retained its prognostic significance across all<br />

stages for patients with HNSCC <strong>of</strong> the OC and L. Patients with OP cancer,<br />

however, had similar 4-year survival for stages I through IVA, whereas stage<br />

IVB and IVC had a significantly decrease survival compared to IVA and IVB<br />

respectively (Table). Conclusions: The OS for stages III and IVA OP cancer is<br />

similar to those with stages I and II, in an effect that may be attributed to<br />

the increased frequency <strong>of</strong> HPV in this population, rendering the tumors<br />

more sensitive to chemotherapy and radiation. Therefore, the AJCC stage in<br />

OP cancer may be more useful in guiding the therapy than as a prognostic<br />

factor.<br />

4-year OS according to tumor site and stage.<br />

Oral cavity Larynx Oropharynx<br />

I 72.9% 71.6% 62.4%<br />

II 59.3% Vs I (HR 1.91, 64.0% Vs I (HR 1.51, 57.7% Vs I (HR 1.14,<br />

p � 0.001)<br />

p � 0.001)<br />

p � 0.22)<br />

III 44.5% Vs II (HR 1.51, 48.9% Vs II (HR 1.68, 67.2% Vs II (HR 0.81,<br />

p � 0.001)<br />

p � 0.001)<br />

p � 0.017)<br />

IVA 33.9% Vs III (HR 1.49, 39.3% Vs III (HR 1.29, 63.9% Vs III (HR 1.16,<br />

p � 0.001)<br />

p � 0.001)<br />

p � 0.014)<br />

IVB 27.2% Vs IVA (HR 1.46, 23.1% Vs IVA (HR 1.85, 42.6% Vs IVA (HR 1.99,<br />

p � 0.001)<br />

p � 0.001)<br />

p � 0.001)<br />

IVC 12.0% Vs IVB (HR 1.74, 16.6% Vs IVB (HR 1.28, 22.2% Vs IVB (HR 2.14,<br />

p � 0.001)<br />

p � 0.027)<br />

p � 0.001)<br />

5531 Poster Discussion Session (Board #22), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Outcomes <strong>of</strong> HIV patients treated with chemoradiotherapy (CRT) for<br />

squamous cell carcinoma <strong>of</strong> the head and neck (SCCHN). Presenting<br />

Author: Waleed F Mourad, Beth Israel Medical Center, New York, NY<br />

Background: SCCHN with HIV is a challenging clinical situation in the<br />

medical arena due to limited data. We report our outcomes and the impact<br />

<strong>of</strong> chemotherapy (CT) and HPV on this patient (pt) population Methods: This<br />

is a single institution retrospective study <strong>of</strong> 71 HIV pts with SCCHN treated<br />

from 1998-2011. The median age at RT and HIV diagnosis was 51 (32-72)<br />

and 34 (25-50) respectively. HIV duration was 11 yrs (6-20). Stage I-II, III<br />

and IV were 22, 27 and 51% respectively. Pts treated with RT alone were<br />

37% and CRT 63% {CDDP 91%}. Fifty pts were on HAART during<br />

treatment and 50% <strong>of</strong> pts with SCC <strong>of</strong> the oropharynx were HPV�. Median<br />

dose <strong>of</strong> 70, 63 and 54 Gy were delivered at 1.8-2 Gy/fraction to gross<br />

disease, high and low risk neck respectively. Twelve pts (17%) underwent<br />

neck dissection for N3 Results: With a median follow up <strong>of</strong> 50 months<br />

(12-140) the median weight loss was 20 lbs (6-40). Acute skin desquamation<br />

and mucositis grades � 2 and 3 were 76 and 24% respectively.<br />

Treatment breaks � 10 and 5 days were 7 and 21% respectively. One pt<br />

died from induction CT, 1 was hospitalized for grade 4 mucositis and 1<br />

developed osteoradionecrosis during CRT. Late dysphagia grades � 2, 3<br />

and 4 were 74, 15 and 11%. Late xerostomia grades � 2 and 3 were 77<br />

and 23% respectively. The median CD4 count and viral load before, during<br />

and after treatment were 370, 135, 100 and 0, 160, 260 respectively.<br />

Seven pts (10%) developed second primary malignancy. The 4 yr locoregional<br />

control (LRC) and overall survival (OS) were 69% and 55%<br />

respectively. Chi-square test shows significant relationship between LRC<br />

and RT duration, as well as between CT and lower CD4 counts and higher<br />

viral load (P�0.001). Positive trends were observed between (weight loss<br />

�10% and LRC) and (absence <strong>of</strong> second malignancy and OS) (P�0.271).<br />

There was no significant relationship between HPV �ve or CT on either LRC<br />

or OS. Conclusions: The addition <strong>of</strong> CT to RT yielded higher morbidity and<br />

mortality without clear benefit on LRC or OS. HIV per se seems to have a -ve<br />

impact on HPV �ve pts outcomes. Comparing observed rates <strong>of</strong> LRC and<br />

OS, our data show that RT�CT administered for HIV �ve pts with SCCHN<br />

appears to be less effective than HIV-ve pts. Larger size studies are<br />

warranted to optimize the management <strong>of</strong> this growing patient population.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


364s Head and Neck Cancer<br />

5532 Poster Discussion Session (Board #23), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Influence <strong>of</strong> extracapsular extension on lymph node staging for patients<br />

with squamous cell carcinoma <strong>of</strong> the head and neck. Presenting Author:<br />

Megan Ann Baumgart, Yale University School <strong>of</strong> Medicine, New Haven, CT<br />

Background: Although extracapsular extension (ECE) is a known poor<br />

prognostic factor in head and neck squamous cell carcinoma (HNSCC) and<br />

recommended to be collected by the current AJCC manual, it currently does<br />

not influence the final stage. We evaluated the prognostic impact <strong>of</strong> ECE in<br />

4 primary sites. Methods: The Surveillance Epidemiology and End Results<br />

(SEER) was queried for patients with squamous cell cancers <strong>of</strong> the oral<br />

cavity (OC), oropharynx (OP), hypopharynx (H) and larynx (L), known ECE<br />

status, M0, and diagnosed between 2004 and 2007. Survival curves were<br />

estimated by the Kaplan-Meier and compared by Cox proportional hazards<br />

models. Results: There were 23,384 patients meeting eligibility criteria,<br />

including 14,664 (62.7%) N0, 6483 (27.7%) N� without ECE (NECE),<br />

and 2237 (9.6%) with ECE. ECE was associated with decreased 3-year<br />

overall survival (OS), compared to NECE, for all lymph node stages<br />

including N1 (52% vs 61%, HR 1.32, p � 0.001), N2a (60% vs 76%, HR<br />

1.82, p�0.001), N2b (44% vs 62%, HR 1.62, p�0.001), N2c (34% vs<br />

47%, HR 1.43, p�0.001), N3 (31% vs 44%, HR 1.38, p�0.012). ECE<br />

was also an independent poor prognostic factor for cancer in the OC (HR<br />

1.43, p � 0.001), OP (HR 1.44, p � 0.001), H (HR 1.58, p � 0.01), and<br />

L (HR 1.28, p � 0.01). The comparison between ECE and NECE with one<br />

additional N degree showed no significant OS difference, except OP N1 and<br />

H N2a ECE, where survival was inferior to OP N2a and H N2b NECE<br />

respectively (Table). Conclusions: ECE is a significant predictor for poor<br />

outcomes in HNSCC, with survival equivalent to one additional degree <strong>of</strong> N<br />

involvement.<br />

N2a NECE vs<br />

N1 ECE<br />

N2b NECE vs<br />

N2a ECE<br />

N2c NECE vs<br />

N2b ECE<br />

N3 NECE vs<br />

N2c ECE<br />

Oral cavity<br />

(n�1,886)<br />

HR 1.19 (0.78-1.8;<br />

p�0.423)<br />

HR 0.77 (0.45-1.31;<br />

p�0.337)<br />

HR 0.96 (0.71-1.29;<br />

p�0.765)<br />

HR 0.98 (0.60- 1.62;<br />

p�0.950)<br />

Oropharynx<br />

(n�4,821)<br />

0.42 (0.30- 0.60;<br />

p��0.0001)<br />

0.85 (0.58-1.23;<br />

p�0.392)<br />

1.22 (0.95-1.56;<br />

p�0.124)<br />

0.72 (0.50-1.03;<br />

p�0.073)<br />

Hypopharynx<br />

(n�687)<br />

0.69 (0.38-1.23;<br />

p�0.208)<br />

0.57 (0.34-0.98;<br />

p�0.040)<br />

1.35 (0.83-2.20;<br />

p�0.222)<br />

0.96 (0.47-1.98;<br />

p�0.920)<br />

Larynx<br />

(n�1,346)<br />

1.30 (0.73-32;<br />

p�0.364)<br />

0.63 (0.35-1.12;<br />

p�0.113)<br />

0.81 (0.57-1.15;<br />

p�0.232)<br />

1.09 (0.66, 1.78;<br />

p�0.738)<br />

5534 General Poster Session (Board #21B), Sat, 1:15 PM-5:15 PM<br />

Phase II study <strong>of</strong> biweekly dose-intense docetaxel plus gemcitabine<br />

(GEM/DOC) in patients with recurrent locoregional or metastatic head and<br />

neck squamous cell carcinoma. Presenting Author: Ammar Sukari, Karmanos<br />

Cancer Institute, Detroit, MI<br />

Background: Patients with metastatic head and neck squamous cell<br />

carcinoma (HNSCC) have a poor prognosis, limited treatment options, and<br />

median survival <strong>of</strong> 6 to 9 months. Docetaxel and gemcitabine have both<br />

shown activity in HNSCC. The optimal combination, dosing, and scheduling<br />

<strong>of</strong> both drugs is, however, unknown. Thus, we investigated the efficacy<br />

and safety <strong>of</strong> biweekly dose intense GEM/DOC in patients with recurrent<br />

locoregional or metastatic HNSCC. Methods: An open-label, singleinstitution,<br />

single-arm, phase II study was conducted for patients who were<br />

previously treated with no more than two cytotoxic regimens. The patients<br />

received docetaxel (60 mg/m2IV) and gemcitabine (3000 mg/m2 IV) on day<br />

1. The treatments were repeated every 14 days (one cycle), until disease<br />

progression or unacceptable toxicity. The primary end point was response<br />

rate. RECIST-defined response was evaluated every 4 cycles and toxicities<br />

were evaluated at each cycle. Results: A total <strong>of</strong> 36 patients were enrolled<br />

(M:F 26:10; median age (range), 60 years (46-79); performance status<br />

0-1) , 29 <strong>of</strong> whom were response-evaluable. The patients received a<br />

median <strong>of</strong> 4 cycles (range 0-24). Of these 29 patients, none achieved<br />

complete response (CR) and 6 demonstrated a partial response (PR). Thus,<br />

the overall response rate was 21% (95% confidence interval [CI], 0.10 –<br />

0.38). Ten patients had stable disease (SD), resulting in tumor control (CR<br />

or PR or SD) in 16 <strong>of</strong> 29 patients (55%), whereas 13 patients (45%) had<br />

disease progression. The median response duration was 3.2 months (80%<br />

CI: 2.0 – 6.1 months). For all 36 patients, the median overall survival was<br />

4.2 months (95% CI: 2.4 – 7.0 months). Myelosuppression was the most<br />

common adverse event. Grade 3-4 neutropenia and anemia were observed<br />

in 10 (30%) and 13 (39%) patients, respectively. None <strong>of</strong> these patients,<br />

however, had febrile neutropenia or bleeding events, and there were no<br />

treatment-related deaths. Conclusions: The combination <strong>of</strong> biweekly dose<br />

intense GEM/DOC was tolerable and active regimen in patients with<br />

recurrent locoregional or metastatic HNSCC. Our findings warrant further<br />

investigation in a larger patient population.<br />

5533 General Poster Session (Board #21A), Sat, 1:15 PM-5:15 PM<br />

Longitudinal Oncology Registry <strong>of</strong> Head and Neck Carcinoma (LORHAN):<br />

Analysis <strong>of</strong> African <strong>American</strong> patients. Presenting Author: David N. Hayes,<br />

UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC<br />

Background: Previous retrospective analyses show poor outcomes for<br />

African <strong>American</strong> (AA) patients with head and neck carcinoma (HNC). Such<br />

racial disparities are not well understood, but patient (pt) demographics,<br />

comorbidities, tumor site, and human papillomavirus (HPV) status are<br />

suggested to play a role. Methods: The LORHAN registry was used to<br />

identify pts �18 yrs <strong>of</strong> age with de novo, non-metastatic HNC, and a record<br />

<strong>of</strong> survival time. This study evaluated pt and treatment characteristics by<br />

race, and conducted adjusted Cox regression analyses <strong>of</strong> overall survival<br />

(OS) and progression-free survival (PFS) to identify prognostic factors and<br />

interactions, and estimate hazard ratios (HRs) <strong>of</strong> AA vs. non-Hispanic white<br />

(NHW). Results: Baseline pt characteristics (see table below). The median<br />

OS/3-yr rate was 41.7 mo/51% in AA vs. 52.9 mo/70% in NHW,<br />

(age-adjusted HR: 1.73 [95% CI: 1.45, 2.08]). The median PFS/3-yr rate<br />

was 29.6 mo/43% in AA and 49.9 mo/62% in NHW, (age-adjusted HR:<br />

1.64 [95%CI: 1.40, 1.93]). Multivariate analysis <strong>of</strong> OS and PFS showed<br />

that AA race, stage III and IV, PS 2, and smoking were significantly<br />

associated with a worse HR, while OP demonstrated a favorable HR over<br />

other sites. Significant interaction tests led to subgroup analyses for OS<br />

showing an elevated risk in AA with OP (HR: 2.62 [95%CI: 1.95, 3.52])<br />

and similar risk elevations in male AA as well as in AA with PS 0-1 when<br />

compared to NHW in the corresponding subgroups. Conclusions: Controlling<br />

for other factors like PS and smoking, a worse prognosis was seen in AA<br />

males with OP localized SCCHN. Further investigation to improve the<br />

outcomes in this population is warranted.<br />

AA<br />

n�385<br />

NHW<br />

n�2,733 p value<br />

Age Yrs, mean (SD) 58.9 (10.1) 59.8 (10.8) 0.13<br />

Sex Male 295 (76.6%) 2,143 (78.4%) 0.43<br />

Smoking Yes 341 (88.6%) 2,133 (78.1%) �0.0001<br />

Alcohol Yes 331 (86.0%) 2,278 (83.4%) 0.19<br />

Site Hypopharynx 32 (8.3%) 102 (3.7%) �0.0001<br />

Larynx 134 (34.8%) 549 (20.1%)<br />

Oral cavity 43 (11.7%) 439 (16.1%)<br />

Oropharynx (OP) 143 (37.1%) 1,290 (47.2%)<br />

Stage I, II 155 (41.5%) 1,399 (52.1%) �0.0001<br />

III, IV 218 (59.3%) 1,284 (47.8%)<br />

PS 0, 1 308 (80.4%) 2,496 (91.7%) �0.0001<br />

HPV test in OP<br />

patients only<br />

Completed 21 (25.3%) 194 (24.1%)<br />

HPV result in OP<br />

patients only<br />

Pos 8 (38.1%) 159 (82.4%) �0.0001<br />

5535 General Poster Session (Board #21C), Sat, 1:15 PM-5:15 PM<br />

An open-labeled, multicentric clinical study <strong>of</strong> cetuximab combined with<br />

intensity-modulated radiotherapy (IMRT) plus concurrent chemotherapy in<br />

locoregionally advanced (LA) nasopharyngeal carcinoma (NPC): A 2-year<br />

follow-up report. Presenting Author: Chun-yan Chen, Cancer Center, Sun<br />

Yat-sen University, Guangzhou, China<br />

Background: To evaluate the safety and efficacy outcomes <strong>of</strong> concurrent<br />

cetuximab plus IMRT and cisplatin in Chinese patients with LA NPC.<br />

Methods: Patients with primary stage III-IVb (UICC/AJCC 2002 staging<br />

system) and non-keratinizing NPC were enrolled in this prospective,<br />

multicentric, phase II study. Cisplatin (80mg/m2 ,q3week) and cetuximab<br />

(400mg/m2 one w before radiation, and then 250mg/m2 /w) were given<br />

concurrently. The prescription dose <strong>of</strong> IMRT to GTVnx (primary tumor in<br />

nasopharynx) was 66 Gy - 75.9 Gy, GTVnd (positive cervical lymph nodes)<br />

was 60 Gy - 70Gy, The response rate was evaluated according to RECIST<br />

1.0 criteria, and adverse events (AEs) were graded according to NCI CTCAE<br />

V3.0 criteria. Results: From July 2008 to April 2009, 100 patients were<br />

enrolled (74 male), with median age <strong>of</strong> 43 years. The proportion <strong>of</strong> stage<br />

III, IVa and IVb patients were 71%, 22% and 7% respectively. 99% <strong>of</strong><br />

enrolled patients completed the planned treatment. AEs were within the<br />

expected range and manageable. No toxic death occurred during the<br />

treatment. Acneiform skin eruptions, mucositis, in-field dermatitis, xerostomia<br />

and neucopenia were the most common seen AEs, with 64% grade 2/3<br />

acneiform eruptions, 26% grade 2/3 in-field dermatitis, 90% � grade 2<br />

mucositis (2 cases <strong>of</strong> grade 4 mucositis with spontaneous bleeding), 40%<br />

� grade 2 xerostomia and 8% grade 2/3 neucopenia. With a median<br />

follow-up time <strong>of</strong> 23.5 months, the 2 year overall survival (OS), disease-free<br />

survival (DFS), local recurrence-free survival, regional (cervical lymph<br />

node) recurrence free survival and distant metastasis-free survival rates for<br />

the ITT population were 91%, 89%, 90%, 90% and 89%, respectively<br />

Multivariate analysis showed that N stage was the only prognostic factor for<br />

OS (p�0.0392, HR�2.946) and DFS (p�0.0062, HR�4.246) in these<br />

patients. Conclusions: Cetuximab combined with IMRT plus concurrent<br />

cisplatin in patients with LA NPC shows satisfactory 2-year locoregional<br />

control rate and 2-year overall survival. The combination seems to be well<br />

tolerated with a manageable side-effect pr<strong>of</strong>ile.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


5536 General Poster Session (Board #21D), Sat, 1:15 PM-5:15 PM<br />

Gingival cancer: 11 year follow-up at Karolinska University Hospital (2000-<br />

2010). Presenting Author: Rusana Simonoska, Department <strong>of</strong> ENT, Karolinska<br />

Institutet/Karolinska University Hospital, Stockholm, Sweden<br />

Background: In Sweden, approximately 500-600 cases <strong>of</strong> oral cancer are<br />

diagnosed every year. These include cancer <strong>of</strong> the gingiva, retromolar<br />

trigonum, bucca, hard palate, tongue and floor <strong>of</strong> the mouth. Each year,<br />

around 15 cases <strong>of</strong> gingival cancer (GC) are diagnosed in the Stockholm<br />

region. The goal <strong>of</strong> this study was to study the onset <strong>of</strong> symptoms,<br />

treatment, prognosis, and sequelae <strong>of</strong> GC in order to optimize the<br />

treatment. Methods: The study consists <strong>of</strong> a retrospective review <strong>of</strong> medical<br />

records <strong>of</strong> all diagnosed cases <strong>of</strong> GC in Stockholm region between<br />

2000-2010, identified through the ENT-clinic Karolinska Hospital patient<br />

database. Results: Our retrospective study comprised 156 patients diagnosed<br />

with GC. The average age was 72 years sharing equally between the<br />

sexes, 50% were smokers. 98% had a squamous cell carcinoma (scc).<br />

Presenting symptoms were <strong>of</strong>ten lump or ulceration in the gums, pain,<br />

bleeding or discomfort/misfit <strong>of</strong> dentures. Around 30% had premalignant<br />

lessons in the oral cavity before diagnosis. 3/4-th <strong>of</strong> all GC was localized in<br />

the lower jaw. 66% <strong>of</strong> the GC-patients presented as aT4 cancer. At<br />

presentation, 26% had a regional metastasis and <strong>of</strong> those 90% had their<br />

primary tumor in the lower jaw. Six cases had bilateral neck disease. 81%<br />

<strong>of</strong> the patients with regional metastasis had low to medium grade <strong>of</strong> scc<br />

differentiation <strong>of</strong> the primary tumor. 84% <strong>of</strong> all patients with regional<br />

metastasis had a T4 primary tumor. Neck dissection was performed in 38%<br />

(n�59). Of these 35 cases where staging neck, i.e. N0 at presentation and<br />

in 7 cases (20%) a positive neck disease was found. The risk for second<br />

primary was 15%. The overall 5-year survival was 24%. Conclusions:<br />

Advanced age and high number <strong>of</strong> T4 cancer at diagnosis partly explains<br />

the poor survival statistics. Almost 30% <strong>of</strong> the patients in our material have<br />

had premalignant lessons in the oral cavity before the cancer diagnosis and<br />

are at high risk for new tumors (second primary); therefore patients with GC<br />

should be followed up for at least 5 years, possibly longer in the presence <strong>of</strong><br />

premalignant lessons. GC <strong>of</strong> the lower jaw is more likely to metastasize than<br />

GC <strong>of</strong> the upper jaw. Due to 20% occult metastasis occurrence in the<br />

staging neck cases, we recommend staging neck dissection for patients<br />

with GC.<br />

5538 General Poster Session (Board #21F), Sat, 1:15 PM-5:15 PM<br />

A phase II study <strong>of</strong> sorafenib in combination with cisplatin and 5-fluorouracil<br />

in the treatment <strong>of</strong> recurrent or metastatic nasopharyngeal carcinoma.<br />

Presenting Author: Cong Xue, Sun Yat-sen University Cancer Center,<br />

Guangzhou, China<br />

Background: Nasopharyngeal carcinoma (NPC) is a disease with distinctive<br />

epidemiology and clinical behavior compared with Head and neck cancer<br />

(HNC) and more common in South-East Asia. Sorafenib monotherapy has<br />

been shown modest anticancer activity in HNC and NPC (C Elser et al; JCO<br />

2007). This study was to evaluate the efficacy and tolerability <strong>of</strong> sorafenib<br />

combined with cisplatin and 5-fluorouracil (5-FU) for patients with recurrent<br />

or metastatic NPC. Methods: A phase II, single arm clinical trial was<br />

conducted in three centers in China. Chemotherapy-naive histologically<br />

confirmed NPC patients with recurrent or metastatic disease were enrolled.<br />

Patients received oral sorafenib 400mg bid continuously until disease<br />

progression or unacceptable toxicity, cisplatin 80mg/m2 intravenously (iv)<br />

on day 1-5, and 5-FU 400mg/m2 iv infusion for 96 hours on day 1.<br />

Treatment was repeated every 21 days for a maximum <strong>of</strong> 6 cycles. Results:<br />

54 patients were enrolled. Most patients were with bone metastases<br />

(70.0%), followed by liver (56%) and lung metastases (56%). The median<br />

chemotherapy administrated was 4.0 cycles (range, 2-6cycles). The<br />

disease control rate (DCR) reached 90.7%, including one complete<br />

response (CR), 41 partial responses (PR) and 7 stable diseases (SD).<br />

Median PFS was 7.2 months (95% CI 6.8-8.4 months) and median OS was<br />

11.8 months (95% CI 10.6-18.7 months). Tumor cavitation after treatment<br />

was observed in 32.1% <strong>of</strong> patients with lung metastases. Decreased<br />

percentage <strong>of</strong> contrast update in responder patients (PR and CR) with liver<br />

metastases was also observed by Contrast-Enhanced Doppler ultrasound.<br />

The major toxicities include hand foot syndrome (HFS), myelo-suppression<br />

and gastrointestinal reaction.Dose reduction <strong>of</strong> sorafenib was required in<br />

22 patients (40.7%) and dose interruption in 14 patients (25.9%) because<br />

<strong>of</strong> toxicity. Conclusions: Sorafenib combined with cisplatin and 5-FU has an<br />

encouraging efficacy pr<strong>of</strong>ile with tolerable toxicity. Further randomized<br />

studies are needed to confirm the clinical benefit <strong>of</strong> sorafenib combined<br />

with chemotherapy in recurrent or metastatic NPC.<br />

Head and Neck Cancer<br />

365s<br />

5537 General Poster Session (Board #21E), Sat, 1:15 PM-5:15 PM<br />

Efficacy <strong>of</strong> concurrent cetuximab (C225) versus 5-fluorouracil/carboplatin<br />

(5FU/CBDCA) or cisplatin (CDDP) with intensity-modulated radiation<br />

therapy (IMRT) for locally advanced head and neck cancer (LAHNSCC).<br />

Presenting Author: Lauren Quinn Shapiro, Memorial Sloan-Kettering Cancer<br />

Center, New York, NY<br />

Background: We reported inferior outcomes for LAHNSCC patients (pts)<br />

that received concurrent C225 vs. high-dose CDDP with IMRT (IJROBP<br />

2011; 81:915). Prior to FDA approval <strong>of</strong> C225 for LAHNSCC, non-CDDP<br />

candidates at our institution were treated with 5FU/CBDCA (JNCI 1999;<br />

91:2081). The purpose <strong>of</strong> this study was to compare the outcomes <strong>of</strong><br />

concurrent C225 vs. 5FU/CBDCA vs. CDDP with IMRT for LAHNSCC.<br />

Methods: Retrospective review <strong>of</strong> records was performed for pts treated at<br />

MSKCC with concurrent C225 (n�49) vs. 5FU/CBDCA (n�52) vs. highdose<br />

CDDP (n�259) and IMRT from 11/02 to 04/08. Overall survival (OS),<br />

locoregional failure-free survival (LRFS), and late toxicity for all pts and<br />

oropharyngeal subset were obtained. Outcomes were analyzed using<br />

univariate/multivariate Cox proportional hazards model or competing-risks<br />

analysis. Multivariate OS analysis was confirmed by propensity score<br />

adjustment. Results: Pts were similar with regard to site, overall stage, and<br />

alcohol/tobacco history. Compared to pts treated with CDDP, those who<br />

received C225 or 5FU/CBDCA were older, with lower performance status,<br />

higher Charlson score, higher T stage, and worse renal function. With<br />

median follow-up <strong>of</strong> 53.1 months for survivors, the 4-year OS was 40.9%<br />

(95% CI 26.0-55.2%) for C225 vs. 70.2% (95% CI 55.5-80.9%) for<br />

5FU/CBDCA vs. 86.9% (95% CI 82.0-90.6%) for CDDP. Compared with<br />

CDDP, C225 (hazard ratio [HR] 4.01, p�0.0001) but not 5FU/CBDCA (HR<br />

1.54, p�0.15) was associated with worse OS on multivariate analysis.<br />

Propensity score analysis yielded a HR <strong>of</strong> 2.76 (p�0.01) for treatment with<br />

C225 vs. platinum-based chemotherapy. 4-year actuarial LRF for 5FU/<br />

CBDCA was similar to CDDP (9.7% vs. 6.3%, HR 1.51, p�0.42 by Gray’s<br />

method) and significantly lower than C225 (40.2%, HR 4.95, p�0.002).<br />

Late toxicity was as follows: C225 (6.1%) vs. CDDP (7.7%) vs. 5FU/CBDCA<br />

(21.2%). Conclusions: After adjusting for independent risk factors, there<br />

was no significant difference in OS or LRFS between 5FU/CBDCA and<br />

high-dose CDDP, but C225/RT resulted in significantly inferior OS and<br />

LRFS. Late toxicity was worst with 5FU/CBDCA.<br />

5539 General Poster Session (Board #21G), Sat, 1:15 PM-5:15 PM<br />

Patterns <strong>of</strong> care in elderly patients with squamous cell carcinoma <strong>of</strong> the<br />

head and neck: A SEER-Medicare analysis. Presenting Author: Noam<br />

Avraham Vanderwalde, Department <strong>of</strong> Radiation Oncology, University <strong>of</strong><br />

North Carolina at Chapel Hill, Chapel Hill, NC<br />

Background: Head and neck squamous cell carcinoma (HNSCC) is predominantly<br />

a disease <strong>of</strong> the elderly, however age and co-morbidity may affect<br />

therapy decisions. The purpose <strong>of</strong> this study was to assess the patterns <strong>of</strong><br />

care <strong>of</strong> elderly HNSCC patients and identify factors associated with<br />

non-receipt <strong>of</strong> surgery and chemoradiation (CRT). Methods: Using the<br />

Surveillance, Epidemiology, and End Results (SEER)-Medicare linked<br />

database (1992-2007) we identified a retrospective cohort <strong>of</strong> nonmetastatic<br />

HNSCC patients and categorized them into treatment cohorts.<br />

Comparisons were made between surgery vs. non-surgery, and CRT vs.<br />

non-CRT cohorts. Multivariate logistic regression models examined factors<br />

associated with non-receipt <strong>of</strong> treatment. Variables included tumor location,<br />

stage, year <strong>of</strong> diagnosis, age, gender, marital status, race, comorbidity,<br />

race-comorbidity interaction, SEER region, socioeconomic variables,<br />

and hospital affiliations. Results: The final cohort <strong>of</strong> 11,336 were 63%<br />

male and 83% white. 52% had no co-morbidities, 44% had oral cavity<br />

HNSCC, 52% were diagnosed between 2002 and 2007, 58% had surgery,<br />

and 21% received CRT. For the CRT vs. non-CRT model, increasing age<br />

(OR � 0.93; 95% CI 0.92 -0.94) and non-whites with co-morbidity (OR �<br />

0.71; 95% CI 0.55 – 0.92) were associated with decreased likelihood <strong>of</strong><br />

receiving CRT. More advanced staged disease, oropharyngeal tumor location,<br />

and diagnosis after 2002 were associated with increased likelihood <strong>of</strong><br />

receiving CRT. For the surgery versus non-surgery model, age was not<br />

associated with receipt <strong>of</strong> surgery (OR 0.99; 95% CI 0.99-1.00), nor was<br />

existence <strong>of</strong> comorbidity in white patients (OR 0.97; 95% CI 0.88-1.05).<br />

However, recent year <strong>of</strong> diagnosis, non-oral cavity tumor location, and<br />

advanced stage were all associated with reduced likelihood <strong>of</strong> receiving<br />

surgery. During the 15 year surveillance period, patients were less likely to<br />

receive surgery, and CRT was used more <strong>of</strong>ten. Conclusions: Age may<br />

influence the non-receipt <strong>of</strong> CRT in this elderly cohort but does not appear<br />

to be significantly associated with the non-receipt <strong>of</strong> surgery. There has<br />

been an increasing trend in the receipt <strong>of</strong> CRT with a decrease in surgery<br />

over the past 15 years.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


366s Head and Neck Cancer<br />

5540 General Poster Session (Board #21H), Sat, 1:15 PM-5:15 PM<br />

Prevalence, trends, and survival impact <strong>of</strong> human papillomavirus on<br />

oropharyngeal cancer in Indian population. Presenting Author: Ankur Bahl,<br />

Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute <strong>of</strong><br />

Medical Sciences, New Delhi, India<br />

Background: Among oropharyngeal squamous cell carcinoma (OSCC), the<br />

true prevalence <strong>of</strong> HPV remains variable and studies have estimated that up<br />

to 60% may be HPV positive. Patients with HPV positive tumor are<br />

usuallyyounger in age, less likely to have history <strong>of</strong> tobacco or alcohol<br />

consumption and associated with a better prognosis but this information for<br />

Indian patients is largely unknown. Methods: 105 newly diagnosed patients<br />

<strong>of</strong> OSCC were enrolled. HPV genotyping was done on the biopsy specimen<br />

by consensus polymerase chain reaction and reverse line-blot hybridization<br />

assay. HPV prevalence was studied according to gender, age, tobacco and<br />

alcohol use and high risk sexual behavior. Results: Overall HPV prevalence<br />

was 22.8%. HPV positive patients were younger by 8 years as compared to<br />

negative patients (P�0.003). No significant correlation between tobacco<br />

consumption, alcoholic habits, and HPV status was observed. The mean<br />

number <strong>of</strong> life time sexual partners in HPV positive patients was 1.66<br />

while, it was 1.33 in HPV negative patients (P�0.049). Incidence <strong>of</strong> high<br />

risk sexual behaviors was more in HPV positive patients (P�0.001). There<br />

were no significant associations between the two groups with respect to<br />

tumor size, nodal stage and the overall stage <strong>of</strong> the tumor. 16% <strong>of</strong> the base<br />

<strong>of</strong> tongue cancers and 40% <strong>of</strong> tonsillar carcinoma were positive (P � 0.02).<br />

Among positive samples, HPV 16 was the commonest (79%) followed by<br />

HPV 18 (12%). 96% <strong>of</strong> patients received treatment. At 18.8 months there<br />

was no significant difference in OS, EFS between HPV positive and negative<br />

OSCC (P � 0.97 and P� 0.51 respectively). Conclusions: The current study<br />

reconfirms that HPV positive OSCC patients are younger with high risk<br />

sexual behavior. Impact <strong>of</strong> smoking and alcohol consumption on HPV<br />

status was not found in this study. HPV positive rates were significantly<br />

higher for tonsillar cancer. Contrary to literature, we did not find any<br />

differences in OS or EFS between two groups. Small numbers <strong>of</strong> patients in<br />

the study group, short follow up period and significant tobacco smoking in<br />

HPV group may be the one <strong>of</strong> the reason for this.<br />

5542 General Poster Session (Board #22B), Sat, 1:15 PM-5:15 PM<br />

Phase II trial <strong>of</strong> chemoradiotherapy concurrent with S-1 plus cisplatin in<br />

patients with unresectable, locally advanced squamous cell carcinoma <strong>of</strong><br />

the head and neck (SCCHN): Results <strong>of</strong> the Japan <strong>Clinical</strong> Oncology Group<br />

study, JCOG 0706. Presenting Author: Makoto Tahara, National Cancer<br />

Center Hospital East, Kashiwa, Japan<br />

Background: To evaluate the efficacy and safety <strong>of</strong> chemoradiotherapy<br />

(CRT) concurrent with S-1 plus cisplatin in patients with unresectable<br />

locally advanced SCCHN. Methods: Eligibility criteria included histologically<br />

proven SCCHN with unresectable locally advanced lesions, PS 0-1,<br />

aged 20 to 75 years old, adequate organ function, and no prior treatment.<br />

Chemotherapy consisted <strong>of</strong> administration <strong>of</strong> S-1 twice daily on days 1-14<br />

at 60 mg/m2 /day, and cisplatin at 20 mg/m2 /day on days 8-11, repeated<br />

twice at a 5-week interval. Single daily radiation <strong>of</strong> 70 Gy in 35 fractions<br />

was given concurrently startingon day 1. For patients achieving an objective<br />

response after CRT, two additional cycles <strong>of</strong> chemotherapy wereadministered.<br />

The primary endpoint was clinical complete response rate (%CR),<br />

which was the proportion <strong>of</strong> complete response (CR) and good partial<br />

response (good PR). Good PR was defined as remaining tissue with tumor<br />

shrinkage, which was not regarded as residual tumor but rather as scar<br />

material.The planned sample size was 45 patients, which was calculated<br />

by SWOG’s two-stage attained design based on an expected %CR <strong>of</strong> 60%<br />

and a threshold <strong>of</strong> 45%, with a one-sided alpha <strong>of</strong> 0.1 and a power <strong>of</strong> 0.9.<br />

Results: From July 2008 to July 2010, 45 eligible subjects were accrued,<br />

including 43 males, with median age 63 years, ECOG PS 0/1 (36/9),<br />

oropharynx/ hypopharynx/larynx (26/15/4), T1/T2/T3/T4a/T4b (1/11/7/<br />

17/9) and N0/N2a/N2b/N2c/N3 (2/3/10/24/6). %CR was 64.4% (8 CR,<br />

21 good PR) on central review. After a median follow-up <strong>of</strong> 1.56 years,<br />

1-year local progression-free survival was 77.8%, with 1-year progressionfree<br />

survival <strong>of</strong> 70.9%, 1-year overall survival <strong>of</strong> 93.3% and 1-year time to<br />

treatment failure <strong>of</strong> 57.6%. Grade 3 or 4 toxicity included mucositis<br />

(46.7%), dysphagia (46.7%), anorexia (42.2%), radiation dermatitis<br />

(26.7%), neutropenia (26.7%) and febrile neutropenia (4.4%). No treatment-related<br />

deaths were observed. Conclusions: This combination showed<br />

promising efficacy with acceptable toxicities. Further investigation in a<br />

phase III trial is planned.<br />

5541 General Poster Session (Board #22A), Sat, 1:15 PM-5:15 PM<br />

Phase II trial <strong>of</strong> everolimus in patients with previously treated recurrent or<br />

metastatic squamous cell carcinoma <strong>of</strong> the head and neck (SCCHN).<br />

Presenting Author: Prakash Varadarajan, University <strong>of</strong> Texas Health Science<br />

Center at San Antonio, San Antonio, TX<br />

Background: The PI3K/Akt/mTOR pathway plays an important role in<br />

SCCHN pathogenesis and resistance to treatment. We conducted a phase II<br />

study <strong>of</strong> everolimus, an oral inhibitor <strong>of</strong> mTOR, in patients with refractory<br />

SCCHN. Methods: Patients with recurrent or metastatic SCCHN treated<br />

with at least 1 prior regimen, performance status (PS) 0-2, and adequate<br />

organ function, received everolimus 10 mg once daily orally. The primary<br />

endpoint was the disease control rate (DCR) defined as the proportion <strong>of</strong><br />

patients with a complete response, a partial response, or stable disease. A<br />

two-stage design was followed. Cytokines were measured in plasma and<br />

serum at baseline and post treatment. Results: 9 patients were enrolled.<br />

Median age 63 years (range 51 - 82), Male/Female, 6/3, performance<br />

status 0/1/2, 2/5/1. Primary site: oropharynx (2), oral cavity (4), larynx (1),<br />

others (2). Median number <strong>of</strong> palliative therapies in the recurrent/<br />

metastatic setting prior to study was 1 (range 1- 4). Median number <strong>of</strong><br />

cycles <strong>of</strong> everolimus delivered was 1 (range 1-5). No objective responses<br />

were seen. One patient had stable disease that lasted 6 months but all<br />

other patients had progression as best response. DCR was 11%. Median<br />

progression-free survival was 40 days. Four patients have died due to<br />

disease progression. Grade 3 adverse events were infrequent: pain (2<br />

patients), anemia (2 patients), and 1 patient each with pruritus, hypertransaminasemia,<br />

hyponatremia, lymphopenia, and fatigue. No Grade 4<br />

toxicities were reported. Biomarker analysis in 2 patients with available<br />

post treatment samples showed reduced plasma levels <strong>of</strong> VEGF-A, Gro-�,<br />

and IL-17, after everolimus treatment, but no reduction was observed in<br />

the plasma levels <strong>of</strong> IL-6 and IL-8, two cytokines <strong>of</strong>ten associated with<br />

SCCHN disease progression. Conclusions: Everolimus as monotherapy was<br />

well tolerated but did not have sufficient activity in this setting. This phase<br />

II study did not meet the predefined primary endpoint in the first stage <strong>of</strong><br />

accrual.<br />

5543 General Poster Session (Board #22C), Sat, 1:15 PM-5:15 PM<br />

Validation for a novel diagnostic standard in HPV-positive oropharyngeal<br />

squamous cell carcinoma. Presenting Author: Xiao-Jun Ma, Advanced Cell<br />

Diagnostics Inc, Hayward, CA<br />

Background: HPV testing is now widely advocated in the work up <strong>of</strong><br />

oropharyngeal squamous cell carcinoma (OPSCC). The �gold standard� for<br />

oncogenic HPV detection is the demonstration <strong>of</strong> transcriptionally active<br />

high-risk HPV in fresh tumour tissue. For clinical utility, testing strategies<br />

have necessarily focused on formalin-fixed paraffin-embedded (FFPE)<br />

tissue, but this has been at the expense <strong>of</strong> reduced sensitivity and<br />

specificity for oncogenic HPV. This study evaluates a novel RNA-based in<br />

situ hybridisation test against the analytical gold standard; detection <strong>of</strong><br />

high-risk HPV mRNA, derived from fresh frozen tissue samples, by<br />

quantitative reverse transcriptase polymerase chain reaction (qPCR).<br />

Methods: A tissue-microarray comprising FFPE cores from 79 OPSCC was<br />

tested using High Risk HPV RNAscope (Advanced Cell Diagnostics, USA)<br />

for HPV16, 18, 31, 33, 35, 52, and 58. Analytical accuracy and capacity<br />

for prognostic discrimination was determined by comparison with HPV RNA<br />

qPCR for HPV16, 18 and 33. Demographic and tumour parameters were<br />

compared by Chi-squared and Kruskal-Wallis tests. Test sensitivity and<br />

specificity were calculated against the standard. Kaplan–Meier survival<br />

estimates were constructed to assess prognostication. Results: High risk<br />

HPV RNAscope had sensitivity and specificity <strong>of</strong> 97% and 93% respectively<br />

(PPV 91%, NPV 98%) by comparison to the gold standard. Kaplan-<br />

Meier estimates <strong>of</strong> disease specific survival (DSS, p�0.002) and overall<br />

survival (OS, p�0.001) by RNAscope were similar to the gold standard<br />

(DSS, p�0.006, OS, p�0.002) and superior to p16 immunohistochemistry<br />

(IHC) or the combination <strong>of</strong> p16 IHC and DNA qPCR. Conclusions: We<br />

demonstrated that High Risk HPV RNAscope can be used to detect<br />

oncogenic HPV in FFPE OPSCC samples and has both excellent analytical<br />

and prognostic performance against the gold standard test for oncogenic<br />

HPV. These results raise the possibility that High Risk HPV RNAscope<br />

could be adopted as an ‘international standard’ test for OPSCC in clinical<br />

practice. As the oncology community approaches therapeutic de-escalation<br />

based on HPV status, such a reliable and efficacious test may have<br />

immediate application.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


5544 General Poster Session (Board #22D), Sat, 1:15 PM-5:15 PM<br />

A multi-institutional phase II trial <strong>of</strong> pazopanib monotherapy in advanced<br />

anaplastic thyroid cancer. Presenting Author: Keith Christopher Bible,<br />

Mayo Clinic, Rochester, MN<br />

Background: Pazopanib, an orally bioavailable multitargeted inhibitor <strong>of</strong><br />

kinases including VEGF-R, demonstrated impressive activity in metastatic<br />

differentiated thyroid cancer (49% durable RECIST PRs) and promising<br />

preclinical activity in anaplastic thyroid cancer (ATC) models, prompting its<br />

evaluation also as a candidate therapeutic in advanced ATC. Methods: A<br />

multicenter single arm phase II trial <strong>of</strong> 800 mg pazopanib daily was<br />

undertaken with the primary endpoint <strong>of</strong> RECIST response rate. The trial<br />

was designed such that there would be a 90% chance <strong>of</strong> detecting a<br />

response rate <strong>of</strong> �20% at the 0.10 significance level when the true tumor<br />

response rate is �5%. A pre-specified stopping rule designated that<br />

enrollment would cease unless 1 or more RECIST PRs�CRs were observed<br />

in the first 14 <strong>of</strong> 33 potential patients. Eligibility required informed<br />

consent, �18 years <strong>of</strong> age, performance status ECOG 0-2, systolic blood<br />

pressure (BP) �140 mm Hg and diastolic BP �90 mm Hg at entry, QTc<br />

interval �480 msecs, and measurable disease by RECIST criteria. Anatomical<br />

imaging and toxicity evaluations were required every 4 weeks. Results:<br />

Sixteen patients were enrolled. One patient withdrew prior to therapy,<br />

leaving 15 evaluable patients – 33.3% were male, with a median age <strong>of</strong> 66<br />

years (range 45-77); 11 <strong>of</strong> 15 patients had progressed through prior<br />

systemic therapy. Four patients required 1-2 dose reductions, with the<br />

most common severe toxicities (CTC-AE version 3.0 grades 3-5) hypertension<br />

(13%) and pharyngolaryngeal pain (13%). Reasons for treatment<br />

discontinuation included: disease progression (12 pts), death on study due<br />

to a vascular event possibly related to treatment (1 pt.), and intolerability<br />

(radiation recall tracheitis – 1 pt, and uncontrolled hypertension – 1 pt).<br />

Although transient disease regression was observed in several patients,<br />

there were no confirmed RECIST tumor responses, triggering study closure<br />

at time <strong>of</strong> interim analysis. Two patients are alive with disease 9.9 months<br />

and 2.9 years post-registration; the remaining 13 died <strong>of</strong> disease. The<br />

median time to progression was 62 days and the median survival time was<br />

111 days. Conclusions: Pazopanib has poor single agent activity in<br />

advanced anaplastic thyroid cancer.<br />

5546 General Poster Session (Board #22F), Sat, 1:15 PM-5:15 PM<br />

The role <strong>of</strong> family caregiver in the head and neck cancer: Psychological<br />

distress and quality <strong>of</strong> life evaluation. Presenting Author: Mario Airoldi, San<br />

Giovanni Antica Sede Hospital, Turin, Italy<br />

Background: The family caregiver (FCG) has become a hot topic. This figure<br />

among head and neck cancer patients is still largely un-investigated; aim <strong>of</strong><br />

our study was: 1) to describe in a more detailed way the role <strong>of</strong> FCG, 2) to<br />

evaluate quality <strong>of</strong> life (QoL) and psychological distress <strong>of</strong> FCGs and<br />

patients 3) to investigate relationships between FCG’s wellbeing and<br />

patient’s QoL and emotional pattern. Methods: Sixty couples <strong>of</strong> patients and<br />

their caregivers were enrolled in this observational cross-sectional study<br />

between April 2007 and May 2011 at 1st ENT Division, 2th Medical<br />

Oncology Division and 2th Radiotherapy Division <strong>of</strong> San Giovanni Battista<br />

Hospital <strong>of</strong> Turin. Inclusion criteria were: diagnosis <strong>of</strong> SCC, advanced stage<br />

(III-IV), completion <strong>of</strong> curative treatment and no evidence <strong>of</strong> disease at the<br />

enrolment. Psycho-oncological assessment was performed using: Distress<br />

Thermometer (DT), Stay-Trait Anxiety Inventory Manual in Y1 and Y2 form<br />

(STAI Y1-Y2), Beck Depression Inventory (BDI) and Montgomery-Asberg<br />

Depression Rating Scale (MDRS), EORTC-QLQ-C30 and Head and Neck-35<br />

module and Caregiver Quality <strong>of</strong> Life Index-Cancer (CQOLC). Results:<br />

Patients: state and trait anxiety are 46,7% (STAI Y1 mean value 40,2�10,2;<br />

cut-<strong>of</strong>f 40) and 36,7% (STAI Y2 mean value 36,7�8,2; cut <strong>of</strong>f 40)<br />

respectively; self reported and clinician rated depression are 31,6% (BDI<br />

mean value 8,2�5,3; cut-<strong>of</strong>f 9) and 48,3% (MDRS mean value 7,9�5,9;<br />

cut-<strong>of</strong>f 6) respectively.CGs: state and trait anxiety are 50% (STAI Y1 mean<br />

value 42,5�9,9; cut-<strong>of</strong>f 40) and 41,7% (STAI Y2 mean value 39,1�8,7;<br />

cut <strong>of</strong>f 40) respectively; self reported and clinician rated depression are<br />

28.3% (BDI mean value 7,3�4,7; cut-<strong>of</strong>f 9) and 41.7% (MDRS mean<br />

value 7,6�5,8; cut-<strong>of</strong>f 6) respectively.Data analysis underlined a positive<br />

association among emotional scales <strong>of</strong> patients and caregivers. Patients’<br />

psychological aspects are negatively associated with caregivers’ QoL and<br />

vice versa. Conclusions: Anxiety and depression are <strong>of</strong>ten present in FCGs<br />

and cured HNC patients. Long term patient’s QoL is the result <strong>of</strong> a frail<br />

balance between FCG and patiet emotional and psychological distress. A<br />

psychological support for FCG could improve patient well-being.<br />

Head and Neck Cancer<br />

367s<br />

5545^ General Poster Session (Board #22E), Sat, 1:15 PM-5:15 PM<br />

A phase I study <strong>of</strong> everolimus (E) plus weekly cisplatin (CDDP) plus<br />

intensity-modulated radiation therapy (IMRT) in head and neck (HN)<br />

cancer. Presenting Author: Matthew G. Fury, Memorial Sloan-Kettering<br />

Cancer Center, New York, NY<br />

Background: The PI3K/mTOR/eIF4E pathway is upregulated in many HN<br />

cancers, and expression <strong>of</strong> eIF4E in surgical margins has been associated<br />

with increased risk <strong>of</strong> recurrence (Cancer Res 2007; 67:2160. Clin Cancer<br />

Res 2004; 10:5820. JCO 1999; 17:2909). Daily E � weekly CDDP was<br />

well tolerated in a phase I study for patients with advanced solid tumors<br />

(Cancer Chemother Pharmacol 2011 Sep 13; Epub ahead <strong>of</strong> print]. E<br />

achieves radiosensitization in pre-clinical models. This study evaluated<br />

weekly CDDP � daily E given concurrently with IMRT in HN cancer.<br />

Methods: This was a single institution phase I study with a standard 3 � 3<br />

dose escalation plan. Patients (pts) received standard definitive IMRT (66<br />

Gy resected tumors, 70 Gy unresected tumors) concurrent with CDDP 30<br />

mg/m2 weekly X 6 and daily oral E according to the dose escalation plan.<br />

Four dose levels (DLs) <strong>of</strong> daily oral E were planned: 2.5 mg, 5 mg, 7.5 mg,<br />

and 10 mg. Toxicities were recorded according to NCI CTCAE v.3. Results:<br />

Thirteen (9M, 4F) pts enrolled, with median age 52y (range, 37 – 65y) and<br />

median KPS 90 (range, 80-100). Primary sites: oral cavity (4), salivary<br />

gland (4), oropharynx (2), nasopharynx (1), scalp (1), unknown primary (1).<br />

Stage: IVA (11), IVB (1), II (1). Prior treatment: surgery (10), chemotherapy<br />

(2). The most common � G.3 treatment related AEs were mucositis<br />

(functional 62%, clinical 31%), oral pain (31%), and lymphopenia (31%).<br />

There were no DLTs among 3 patients at DL1 or the first 3 patients at DL2.<br />

Among 4 pts at DL3, there were 2 DLTs: grade 3 mucositis with failure to<br />

thrive, and grade 3 mucositis with hypoxia. Among 3 additional patients<br />

enrolled at DL2, there was one 1 DLT: grade 3 mucositis with inability to<br />

tolerate E. There were no treatment-related deaths. With a median<br />

follow-up <strong>of</strong> 8.9 months, 3 patients have experienced recurrent disease,<br />

one <strong>of</strong> whom has died <strong>of</strong> disease. Median PFS has not been reached. Tissue<br />

correlates in process. Conclusions: When administered with weekly CDDP<br />

(30 mg/m2 ) and definitive IMRT for HN cancer, the phase II recommended<br />

dose <strong>of</strong> E is 5 mg/day.<br />

5547 General Poster Session (Board #22G), Sat, 1:15 PM-5:15 PM<br />

Antitumor activity <strong>of</strong> cabozantinib (XL184) in a cohort <strong>of</strong> patients (pts) with<br />

differentiated thyroid cancer (DTC). Presenting Author: Maria E. Cabanillas,<br />

University <strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: Cabozantinib (cabo) is an oral, potent inhibitor <strong>of</strong> MET,<br />

VEGFR2, and RET that is currently undergoing evaluation in several<br />

oncology indications. Differentiated thyroid cancer (DTC) pts were included<br />

in this study based on involvement <strong>of</strong> the MET, VEGFR, and RET signaling<br />

pathways in this disease. The primary objective <strong>of</strong> this study is to determine<br />

the effect <strong>of</strong> cabo on single dose PK <strong>of</strong> the CYP2C8 substrate rosiglitazone<br />

(rosi). Anti-tumor activity and safety were also evaluated. Methods: Metastatic<br />

DTC pts who were enrolled to this study were required to be<br />

RAI-refractory, have progressed on standard therapies and have measurable<br />

disease. Cabo was given daily at a dose <strong>of</strong> 140 mg free base<br />

(equivalent to 175mg salt form) starting at Day 2. Rosi (4mg) was given Day<br />

1 and Day 22 to complete PK assessment for drug-drug interaction (DDI).<br />

Cabo was continued until PD. On Day 57 and every 8 weeks thereafter<br />

subjects underwent tumor assessments by mRECIST. Results: 15 DTC pts<br />

were enrolled. Median number <strong>of</strong> prior regimens was 2 (11/15 pts had at<br />

least 1 prior VEGFR inhibitor). 8/15 pts (53%) had confirmed PRs<br />

including 1 pt with marked improvement <strong>of</strong> a bone infiltrating lesion; 6<br />

(40%) had best response <strong>of</strong> SD; all 14 pts with �1 post-baseline scan<br />

experienced tumor regression (range: -9 to -55%). Disease control rate (PR<br />

� SD): 80% at 16 weeks; 10/15 (67%) remain on cabo with a median<br />

follow-up <strong>of</strong> 7.3 months. Median PFS and OS have not been reached. Most<br />

common Gr 3/4 AEs were: diarrhea (20%), lipase increased (20%),<br />

hypertension (13%) and palmar-plantar erythrodyesthesia (13%); one<br />

related Gr 5 event reported: hemoptysis due to aorto-trachael fistula in a pt<br />

with history <strong>of</strong> prior mediastinal XRT and extensive neck surgeries. PK data<br />

suggest that clinically relevant doses <strong>of</strong> cabo do not alter the Cmax or<br />

AUC <strong>of</strong> rosi, consistent with no inhibition <strong>of</strong> CYP2C8. Conclusions: In<br />

0-24h<br />

pts with DTC, cabo treatment resulted in substantial anti-tumor activity.<br />

The safety pr<strong>of</strong>ile <strong>of</strong> cabo was comparable to that seen with other VEGFR<br />

TKIs. PK data suggest no DDI between cabo and rosi (CYP2C8 substrate).<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


368s Head and Neck Cancer<br />

5548 General Poster Session (Board #22H), Sat, 1:15 PM-5:15 PM<br />

Recurrent/metastatic salivary gland malignancies (RMSGM): Final report <strong>of</strong><br />

60 cases treated with cisplatin plus vinorelbine (DDP�VNB). Presenting<br />

Author: Fulvia Pedani, Department <strong>of</strong> Medical Oncology, San Giovanni<br />

Antica Sede Hospital, Turin, Italy<br />

Background: RMSGM are not amenable to the usual treatment with surgery<br />

and post-operative radiotherapy. The role <strong>of</strong> chemotherapy (CT) for RMSGM<br />

is palliative only. VNB showed moderate activity in our experience (Bull<br />

Cancer 85:892;1998) and in a randomized phase II trial we had demonstrated<br />

that the DDP�VNB combination had a better outcome than VNB<br />

alone (Cancer 91:541; 2001). In this abstract we report the final results <strong>of</strong><br />

this combination in 60 cases. Methods: From April 2001 to February 2009<br />

, 60 cases with RMSGM were enrolled. All patients received the following<br />

regimen: DDP 80 mg/sm d.1 � VNB 25 mg/sm d. 1,8 every 3 weeks. The<br />

study foresees a maximum <strong>of</strong> 6 cycles. Results: Patients characteristics<br />

were as follows: 35 males (58%) and 25 females (42%).; median age: 56<br />

yrs (range 20-68); median ECOG PS: 1 (0-2); histology: adenocarcinoma<br />

15 (25%), adenoid cystic ca. 34 (57%), others 11 (18%); site <strong>of</strong> disease:<br />

local 30 (50%) , mts �/- local 30 (50%). Forty-two pts received DDP�VNB<br />

as first line CT (70%) while 18 pts (30%) had the combination as<br />

second-line CT (30%). After a median <strong>of</strong> 5 cycles <strong>of</strong> first line DDP�VNB<br />

responses were: 3 CR (7%), 10 PR (24%), 14 NC (33%) and 15 PD (36%).<br />

After a median <strong>of</strong> 4 cycles <strong>of</strong> second line CT responses were: 0 CR; 1 PR<br />

(5%), 6 NC (33%) 11 PD (62%). Median survival: 10 months (3-29) for<br />

first line CT ; 4 months (1-12) for second line CT. G3-4 toxicity:<br />

neutropenia (20%), anemia (12%), nausea/vomiting (12%) , peripheral<br />

toxicity (3%). Conclusions: DDP�VNB is an effective first line CT in<br />

RMSGM ; second line CT has a low palliative activity. Toxicity seems<br />

acceptable. This regimen could be suitable for an integration with new<br />

biologic target agents.<br />

5550 General Poster Session (Board #23B), Sat, 1:15 PM-5:15 PM<br />

Preoperative recombinant adenoviral human p53 gene combined with<br />

intensity-modulated radiation therapy in treatment <strong>of</strong> stage IV papillary<br />

thyroid carcinoma: A randomized clinical study. Presenting Author: Jingqiang<br />

Zhu, West China Hospital in Sichuan University, Chengdu, China<br />

Background: Most <strong>of</strong> stage IV papillary thyroid carcinoma (PTC) is initially<br />

unresectable due to invading surrounding structures, and is also commonly<br />

resistant to standard radio- or chemo-therapy. P53 gene therapy could<br />

increase tumor radiosensitivity and inhibit tumor angiogenesis. This study<br />

is to evaluate the benefits <strong>of</strong> pre-operative recombinant adenoviral human<br />

p53 gene (rAd-p53) combined with intensity-modulated radiation therapy<br />

(IMRT) in treatment <strong>of</strong> stage IV PTC. Methods: Forty-six patients with<br />

historically-diagnosed stage IV PTC, satisfying the inclusion criteria, were<br />

randomly assigned into two groups: experimental group (EG) control group<br />

(CG). EG received intratumoral injection <strong>of</strong> 2-4�1012 rAd-p53 viral<br />

particles (VP) per 3 days for 10 times and IMRT once per day for 28 days at<br />

a total dose <strong>of</strong> 65 Gy. CG received the same IMRT therapy. Once month<br />

after the combined treatment, patients were assessed for resectability. A<br />

radical surgery was performed on the patients with a resectable tumor. All<br />

the patients received standard I131 therapy 1-1.5 months after surgery or<br />

after the last IMRT. Results: EG including 25 cases, 19 females and 6<br />

males, had a radical surgery rate <strong>of</strong> 76% (19/25), one-year survival rate <strong>of</strong><br />

96%, and no local recurrence and distant metastases for patients having a<br />

surgery. The tumors in six patients <strong>of</strong> EG were still unresectable. But all<br />

these six patients had a local partial response. The tissue sample analyses<br />

showed increased fibrous tissues, apoptotic cells, and increased Bax and<br />

p21 protein and mRNA levels. CG including 21 cases, 15 females and 6<br />

males, had a radical surgery rate <strong>of</strong> 47.6% (10/21), one-year survival rate<br />

<strong>of</strong> 81.0%, and distant metastatic rate <strong>of</strong> 20% (2/10) for patients having a<br />

surgery. The local partial response rate <strong>of</strong> unresectable patients in CG was<br />

72.7% (8/11). There were no changes in Bax and p21 protein and mRNA<br />

levels. The main side effects <strong>of</strong> rAd-p53 were self-limited fever, occurring<br />

in 24 <strong>of</strong> EG patients. Conclusions: Pre-perative rAd-p53 combinedwith<br />

IMRT in treatment <strong>of</strong> the stage IV PTC increases radical surgical rate and<br />

response rate, and improves long-term efficacy.<br />

5549 General Poster Session (Board #23A), Sat, 1:15 PM-5:15 PM<br />

Phase II study <strong>of</strong> temsirolimus and erlotinib in patients (pts) with<br />

recurrent/metastatic (R/M), platinum-refractory head and neck squamous<br />

cell carcinoma (HNSCC). Presenting Author: Julie E. Bauman, University <strong>of</strong><br />

New Mexico, Albuquerque, NM<br />

Background: The Epidermal Growth Factor Receptor (EGFR) is a validated<br />

target in HNSCC. In R/M disease, primary or acquired resistance to<br />

anti-EGFR therapy inevitably occurs. Downstream activation <strong>of</strong> the PI3K/<br />

Akt/mTOR pathway is an established resistance mechanism. We hypothesized<br />

that concurrent mTOR blockade may improve efficacy <strong>of</strong> anti-EGFR<br />

therapy and conducted this phase II study. Methods: We evaluated the<br />

combination <strong>of</strong> erlotinib 150 mg po daily and temsirolimus 15 mg IV<br />

weekly in pts with platinum-refractory R/M HNSCC and ECOG PS 0-2. The<br />

exact, single-stage phase II design had 80% power (5% significance level)<br />

to detect improvement in progression-free survival (PFS) from 2.3 to 4.4<br />

mos with 35 evaluable pts. Correlatives included determination <strong>of</strong> PIK3CA<br />

mutation (mut) status; p16, EGFR and PTEN expression; and immunomodulatory<br />

cytokine levels. Results: From Dec 2009 – Mar 2011, 12 pts<br />

enrolled. Six pts withdrew prior to first planned response assessment, due<br />

to toxicity (5) or death (1), prompting referral to the Data Safety and<br />

Monitoring Committee and early closure. Grade � 3 toxicities included<br />

fatigue (5), diarrhea (2), GI infection/peritonitis (2), dyspnea (2), HN<br />

edema (2), and neutropenia (1). Among 8 progression-evaluable pt,<br />

median PFS was 1.9 mos. Median OS was 4.1 mos. 4/12 tumors were<br />

p16(�); 4/10 were EGFR(�); 11/11 showed no PTEN expression. PIK3CA<br />

mut was present in 1/11; the mut(�) pt withdrew after 3 wk for<br />

diarrhea/peritonitis with minor response. There were no associations<br />

between tumor p16, EGFR or PTEN status and oncologic outcomes. Five <strong>of</strong><br />

6 toxicity-related withdrawals occurred in p16(-) pts. Immunomodulatory<br />

cytokine levels in pre- and post-temsirolimus plasma did not significantly<br />

differ or associate with toxicity. Two pts with � 4.4 mos PFS were p16(-)<br />

and PIK3CA mut(-). Conclusions: The combination <strong>of</strong> erlotinib and temsirolimus<br />

was poorly tolerated in this population, with toxicities predominant in<br />

p16(-) pts. Although the sample size was small, this R/M cohort demonstrated<br />

lack <strong>of</strong> PTEN expression and 9% PIK3CA mut, justifying further<br />

investigation <strong>of</strong> PI3K/Akt/mTOR pathway inhibitors in selected HNSCC pts.<br />

5551 General Poster Session (Board #23C), Sat, 1:15 PM-5:15 PM<br />

Association <strong>of</strong> smoking status with p16 and cyclin D1 (CCND1) expression<br />

with clinical characteristics and overall survival (OS) in oropharyngeal<br />

squamous cell carcinoma (OSC). Presenting Author: Mei-Kim Ang, National<br />

Cancer Centre, Singapore, Singapore<br />

Background: Smoking-related head and neck cancer (HNC) is genetically<br />

different, with higher mutation rates compared with non-smokers (NS).<br />

Human papillomavirus (HPV)-positive (�) OSC has a superior prognosis<br />

independent <strong>of</strong> treatment. Among HPV� patients (pt), current or prior<br />

smokers (CS) have poor OS compared with NS. Expression <strong>of</strong> p16, a known<br />

HPV surrogate, and CYD1, a cell cycle marker <strong>of</strong>ten dysregulated in HNC,<br />

was evaluated with respect to smoking status and OS. Methods: Expression<br />

<strong>of</strong> p16 and CYD1 was assessed by immunohistochemistry in 108 OSC pt<br />

treated between 1999-2009, using cut<strong>of</strong>fs <strong>of</strong> �70% (p16�) and �10%<br />

(CYD1�) stained tumor cells. Associations between expression, clinical<br />

characteristics and OS were evaluated by Kaplan-Meier method and<br />

compared by log rank test. Hazard ratio (HR) for death was estimated using<br />

Cox models. Results: 31 pt (28.7%) were p16� and 80 pt (75.5%) were<br />

CYD1 negative(-). p16� pt were younger (median age 57 v 66 yrs,<br />

p�0.002), more likely female (35.5% v 15.2%, p�0.035), NS (51.6% v<br />

13.9%, p�0.001) with lower combined age-comorbidity score (ageCS)<br />

(p�0.003). CYD1� pt were older (median 66 v 57 yrs, p�0.015), more<br />

likely CS (81.5% v 48.1%, p�0.002) with higher ageCS (p�0.018). At a<br />

median f/u <strong>of</strong> 65.7 months, median OS was 57.3 months. p16� pt had<br />

better OS than p16- pt (median OS not reached (NR) v 22.3 mths,<br />

p�0.001). CYD1� pt had poorer OS than CYD1- pt (median OS NR v 17.7<br />

mths, p�0.001). On multivariable analysis p16 and CYD1 status were<br />

independently associated with OS (HR 0.412, p�0.045 and HR 4.06,<br />

p�0.011 respectively), independent <strong>of</strong> smoking status (HR 5.01,<br />

p�0.008), ageCS (HR 1.32 per 1 point increase, p�0.001) and stage.<br />

Strikingly, among NS, 5-year OS in p16� compared with p16- pt was<br />

100% vs 67% (p�0.001). In contrast, among CS, p16 status had no<br />

association with OS (HR 0.97, p�0.943), while CYD1 status and ageCS<br />

were independent predictors <strong>of</strong> death (HR 4.70, p�0.025 and HR 1.28,<br />

p�0.001 respectively). Conclusions: In OSC, NS with high p16 expression<br />

have excellent prognosis. Among CS, pt with fewer comorbidities and low<br />

CYD1 expression have better OS. p16 status was not prognostic in the latter<br />

group <strong>of</strong> patients.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


5552 General Poster Session (Board #23D), Sat, 1:15 PM-5:15 PM<br />

Response <strong>of</strong> chemoradiation therapy after induction chemotherapy failure<br />

in locally advanced head and neck squamous cell carcinoma (LA-HNSCC).<br />

Presenting Author: Yoojoo Lim, Department <strong>of</strong> Internal Medicine, Seoul<br />

National University Hospital, Seoul, South Korea<br />

Background: Although induction chemotherapy (IC) for LA-HNSCC has<br />

shown high response rates, some patients do not respond to IC. Surgery has<br />

been the usual choice for the non-responding patients, but practically, not<br />

all <strong>of</strong> these patients are eligible for operation. The purpose <strong>of</strong> this study was<br />

to evaluate the efficacy <strong>of</strong> definitive radiation therapy (RT) for the HNSCC<br />

patients who failed IC. Methods: We have retrospectively analyzed the<br />

outcome <strong>of</strong> patients with LA-HNSCC who were initially treated with IC at<br />

Seoul National University Hospital between Jan. 2006 and Dec. 2010.<br />

Chemotherapeutic regimens used were 5-FU/cisplatin, docetaxel/5-FU/<br />

cisplatin, and docetaxel/cisplatin with or without cetuximab. After IC,<br />

patients were treated with RT, concurrent chemoradiation (CCRT), or<br />

surgery, either alone or with postoperative RT. Treatment modality was<br />

chosen on multi-disciplinary basis <strong>of</strong> our institution. Response was<br />

evaluated using RECIST criteria. Results: A total <strong>of</strong> 225 LA-HNSCC<br />

patients treated with IC were analyzed. Median age was 54 (range 24-77).<br />

Primary tumor locations were oral cavity (39.5%), nasal cavity/paranasal<br />

sinus (13.1%), oropharynx (13.1%), nasopharynx (13.1%), larynx (7.9%)<br />

and hypopharynx (13.1%). Among the 225 patients, 38 (16.9%) patients<br />

did not respond to IC [stable diseases (SD) � 23 (10.2%), progressive<br />

disease (PD) � 15 (6.7%)], and their median overall survival was 14.5<br />

months. Among the 38 non-responders, 14 (36.8%) patients were undertaken<br />

surgery and 22 (57.8%) unresectable or inoperable patients were<br />

treated with either definitive RT or CCRT. Responses to subsequent<br />

definitive RT or CCRT for 20 evaluable patients were as follows: complete<br />

response � 6 (30.0%), partial response � 7 (35.0%), SD � 3 (15.0%), PD<br />

� 4 (20.0%) Overall response rate to definitive RT or CCRT in nonresponder<br />

to IC was 65.0% (95%CI: 44.1%- 85.9%). Nine out <strong>of</strong> 15<br />

patients who showed PD to IC received definitive RT or CCRT and<br />

interestingly, 3 (33.3%) patients responded to definitive RT or CCRT.<br />

Conclusions: A significant portion <strong>of</strong> HNSCC patients who failed to IC can<br />

benefit from subsequent definitive RT or CCRT. Further prospective study is<br />

warranted.<br />

5554 General Poster Session (Board #23F), Sat, 1:15 PM-5:15 PM<br />

Expression and prognostic significance <strong>of</strong> directed therapy targets and<br />

mutational analysis <strong>of</strong> the EGFR pathway in malignant salivary gland<br />

tumors. Presenting Author: Jerome F. Cros, Georges Pompidou European<br />

Hospital, Paris, France<br />

Background: Malignant salivary gland tumors are rare and pleomorphic<br />

entities (24 sub types, WHO 2005). Curative treatment relies on surgery<br />

sometimes completed by radiotherapy. Management <strong>of</strong> non-surgical, locally<br />

advanced or metastatic tumors is difficult as conventional chemotherapies<br />

are ineffective. Directed therapies may provide important benefits for<br />

these patients. The aim <strong>of</strong> this work was to assess the expression and<br />

prognostic value <strong>of</strong> directed therapy targets on salivary gland tumors and<br />

search for mutations within the key players <strong>of</strong> the EGFR pathway. Methods:<br />

Immunochemistry on formalin fixed paraffin embedded (FFPE) samples<br />

from 124 patients for c-KIT, EGFR, c-ERBB2, MUC1, phospho-mTOR,<br />

androgen/estrogens/progesterone receptors and Ki67 was performed and<br />

related to progression free interval and survival. DNA was extracted from<br />

FFPE samples and conditional for treatment mutations <strong>of</strong> EGFR/KRAS/<br />

BRAF were assessed. An additional high throughput screening for mutations<br />

<strong>of</strong> key oncogenic genes was performed. Results: EGFR was the most<br />

expressed marker, found across almost all histotypes. Expression <strong>of</strong> the<br />

other markers was heterogeneous but some potential therapy leads were<br />

suggested by high levels <strong>of</strong> C-ERBB2 and androgen receptors in salivary<br />

duct carcinomas or C-KIT over expression in myoepithetial carcinomas.<br />

Tumor grade and a high proliferation index (Ki67�20%) were associated<br />

with progression free interval and survival. None <strong>of</strong> the other marker was.<br />

No mutation was found in EGFR/KRAS/BRAF. 7% (7/104) <strong>of</strong> the tumors<br />

harbored a mutation at the codon 61 <strong>of</strong> HRAS. In epithelial and epithelialmyoépithélial<br />

carcinoma, the mutation rate reached 33%. Mutations <strong>of</strong><br />

PI3K and p53 were the most frequently found in the broader screen.<br />

Conclusions: Several tumor subtypes expressed frequently some targets <strong>of</strong><br />

directed therapies suggesting potential therapy leads. The EGFR/MAPK<br />

pathway seems intact suggesting a low efficacy <strong>of</strong> small kinase inhibitors<br />

such as erlotinib or gefitinib. The mutation <strong>of</strong> H-RAS, known to be<br />

important but not sufficient to trigger urothelial carcinoma, could be a key<br />

oncogenic event in tumors with a myoepithelial component.<br />

Head and Neck Cancer<br />

369s<br />

5553 General Poster Session (Board #23E), Sat, 1:15 PM-5:15 PM<br />

The accuracy <strong>of</strong> 18F-fluorodeoxyglucose-positron emission tomography/<br />

computed tomography (FDG-PET/CT) in the staging <strong>of</strong> newly diagnosed<br />

nasopharyngeal carcinoma: A systematic review and meta-analysis. Presenting<br />

Author: Balamurugan A. Vellayappan, National University Cancer<br />

Institute, Singapore, Singapore, Singapore<br />

Background: The specific role <strong>of</strong> FDG-PET/CT in pretreatment staging <strong>of</strong><br />

nasopharyngeal carcinoma (NPC) remains to be validated. We performed a<br />

systematic review and meta-analysis to assess the diagnostic accuracy <strong>of</strong><br />

staging FDG-PET/CT for newly diagnosed NPC with reference to conventional<br />

staging modalities and/or clinical follow up. Methods: We searched<br />

MEDLINE, Cochrane central register <strong>of</strong> controlled trials, proceedings <strong>of</strong><br />

ASTRO and ASCO as well as Chinese databases (Chinese National<br />

Knowledge Infrastructure and CBMdisc) from the date <strong>of</strong> inception to<br />

September 2011 for relevant studies. Study quality was assessed using the<br />

Quality Assessment <strong>of</strong> Diagnostic Accuracy Studies (QUADAS) checklist.<br />

We determined the sensitivities and specificities across studies, pooled<br />

diagnostic odds ratios (DOR) and constructed summary receiver operating<br />

characteristic curves using hierarchical regression model. Results: We<br />

found 15 relevant studies (<strong>of</strong> which seven were in English) including 851<br />

patients. Of the 15 studies: five addressed primary tumor (T), nine<br />

addressed regional lymph nodes (N) and seven addressed distant metastasis<br />

(M). The combined sensitivity estimate for FDG-PET/CT in T classification<br />

was 0.77(95% confidence interval (CI) 0.59-0.95). The combined<br />

sensitivity estimate for N classification was 0.88 (95% CI 0.86-0.90),<br />

specificity 0.85(95% CI 0.83-0.88), DOR 82.4 (23.2 to 292.6) and<br />

Q-index was 0.90. For M classification, the combined sensitivity estimate<br />

was 0.82(95% CI 0.65-0.93), specificity 0.98 (95% CI 0.96 – 0.99), DOR<br />

120.9 (43.0 to 340.0) and Q-index was 0.89. Conclusions: FDG-PET/CT<br />

showed good accuracy in N and M but not T classification for newly<br />

diagnosed pre-treated NPC. FDG-PET/CT, together with Magnetic resonance<br />

imaging (MRI) <strong>of</strong> the nasopharynx, should be part <strong>of</strong> the routine<br />

staging investigations for NPC. Future research should evaluate the<br />

accuracy <strong>of</strong> FDG-PET/MRI fusion as a single staging modality for NPC.<br />

5555 General Poster Session (Board #23G), Sat, 1:15 PM-5:15 PM<br />

Association between salivary cytokine levels and chemoradiotherapyinduced<br />

toxicities in head and neck cancer patients. Presenting Author:<br />

Paolo Bossi, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy<br />

Background: Mucositis is a common complication <strong>of</strong> chemoradiotherapy<br />

(CTRT) for head and neck cancer (HNC), linked to a balance between proand<br />

anti-inflammation serum cytokines. No study has yet addressed the<br />

role <strong>of</strong> salivary cytokines in influencing toxicity severity. Methods: Twenty<br />

consecutive stage III (15%) and IV (85%) HNC patients (pts) were treated<br />

with radiotherapy (64-70 Gy) plus cisplatin (n�15), carboplatin (n�4) or<br />

cetuximab (n�1). Primary tumor site was oral cavity (15%), oropharynx<br />

(55%), nasopharynx (15%), larynx or hypopharynx (15%). Unstimulated<br />

saliva samples were collected according to standardized protocols before<br />

CTRT, during (3rd ,5th and 7th weeks) and two weeks after; concomitantly,<br />

mucositis grade (WHO classification), weight loss and need for feeding tube<br />

were evaluated. The salivary levels <strong>of</strong> 11 different cytokines (IFN�, IL1�,<br />

IL2, IL4, IL5, IL6, IL8, IL10, IL12p70, TNF� and TNF�) were analysed<br />

both in pts and in healthy donors (HD, n�10) by optimized bead-based<br />

multiplex immunoassay. The cytokine change during treatment was calculated<br />

as the difference between the mean <strong>of</strong> individual values and the<br />

baseline value. The Wilcoxon rank sum test was used for between-groups<br />

comparisons. Results: At baseline, no difference in cytokine levels was<br />

observed in pts as compared with HD, except IL8. A significant and<br />

progressive increase <strong>of</strong> IL1�, IL6, IL8, TNF� and IL10 levels was observed<br />

during treatment, with high levels persisting two weeks after treatment for<br />

all cytokines but IL1�. Significant association was shown between IL6<br />

increase and G3/4 mucositis (p�0.009) or feeding tube need (p�0.04).<br />

The same trend was observed for TNF�. Interestingly, IL8 increase<br />

appeared to be specifically linked to weight loss (�10%, p�0.003). In<br />

contrast, baseline cytokine salivary levels were not predictive <strong>of</strong> treatmentinduced<br />

toxicities. Conclusions: The increase <strong>of</strong> IL6, IL8 and TNF� salivary<br />

levels occurring in HNC patients with CTRT seems to be directly associated<br />

with mucositis severity, feeding tube prevalence and weight loss. Cytokines<br />

may represent a potential new target for preventive and/or therapeutic<br />

intervention.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


370s Head and Neck Cancer<br />

5556 General Poster Session (Board #23H), Sat, 1:15 PM-5:15 PM<br />

Organ preservation with daily concurrent chemoradiotherapy using a new<br />

superselective intra-arterial infusion for stage III, IV tongue cancer.<br />

Presenting Author: Iwai Tohnai, Department <strong>of</strong> Oral and Maxill<strong>of</strong>acial<br />

Surgery, Yokohama City University School <strong>of</strong> Medicine, Yokohama, Japan<br />

Background: Combined radiotherapy and intra-arterial chemotherapy, which<br />

delivers a very high concentration <strong>of</strong> anticancer agent to the tumor, is<br />

currently used to treat advanced head and neck cancer. However, chemotherapy<br />

and radiotherapy cannot be performed simultaneously on a daily<br />

basis under conventional methods because long-term catheterization is not<br />

possible. Therefore, we developed a new method using superselective<br />

intra-arterial infusions via the superficial temporal artery and the occipital<br />

artery, and used it to perform daily concurrent chemoradiotherapy for the<br />

treatment <strong>of</strong> advanced tongue cancer. Methods: Seventy-two patients with<br />

advanced tongue cancer underwent chemoradiotherapy using this superselctive<br />

intra-arterial infusion method. Catheters were inserted superselectively<br />

to the feeding arteries <strong>of</strong> the tumor via the superficial temporal artery<br />

and the occipital artery. Long-term catheterization is possible in this<br />

method. Daily concurrent combined therapy with radiotherapy (total dose:<br />

60 Gy), superselective intra-arterial chemotherapy using DOC (total dose:<br />

60 mg/m2 ) and CDDP (total dose: 150 mg/m2 ) was performed. The<br />

anticancer agent was injected in a bolus through the intra-arterial catheter<br />

simultaneously with radiotherapy. Results: No major complications were<br />

observed. <strong>Clinical</strong> efficacy was CR in 64 (88.9 %) patients and PR in 8<br />

(11.1 %). With a median follow-up period <strong>of</strong> 31 months (range 6 to 72<br />

months), the overall survive rate was 74.8 % and the local control rate was<br />

93.7%. Conclusions: This superselective intra-arterial infusion allows for<br />

long-term catheterization, unlike the conventional method, so that chemotherapy<br />

can be performed simultaneously with radiotherapy on a daily<br />

basis. This treatment method promises to be the strategy <strong>of</strong> choice for the<br />

treatment <strong>of</strong> advanced tongue cancer.<br />

5558 General Poster Session (Board #24B), Sat, 1:15 PM-5:15 PM<br />

Recombinant adenoviral human p53 gene combined with radio- and<br />

chemotherapy in treatment <strong>of</strong> advanced nasopharyngeal carcinoma: A<br />

randomized clinical study. Presenting Author: Guiping Lan, The People’s<br />

Hospital <strong>of</strong> Guangxi Zhuang Autonomous Region, Nanning, China<br />

Background: Studies showed p53 gene therapy could enhance the antitumor<br />

effect <strong>of</strong> both chemo- and radiotherapy. The anti-tumor function <strong>of</strong><br />

p53 gene is associated with up-regulated expression <strong>of</strong> p21 and Bax. Here,<br />

we investigate the beneficial role <strong>of</strong> recombinant adenoviral human p53<br />

gene (rAd-p53) combined with chemoradiotherapy in treatment <strong>of</strong> advanced<br />

nasopharyngeal carcinoma (NPC), and p21 and Bax protein<br />

expression in pre- and post-treatment tumor tissues. Methods: Sixty-three<br />

patients with historically-diagnosed advanced NPC, with ECOG performance<br />

status less than 2, life expectancy greater than 3 months and<br />

adequate organ function tests, were randomly assigned into two groups: 32<br />

in experimental group (EG) and 31 in control group (CG). EG received<br />

intratumoral injection <strong>of</strong> 1�1012 rAd-p53 viral particles (VP) per 3 days for<br />

6-8 times, carboplatin 300 mg/m2 on day1 and continuous infusion <strong>of</strong> a<br />

total dose <strong>of</strong> 2000 mg/m2 <strong>of</strong> 5-Fu on day1-5 for 3 cycles, and radiotherapy<br />

at a dose <strong>of</strong> 2Gy/fraction for a total dose <strong>of</strong> 70-76 Gy/35-38f. CG received<br />

the same chemo- and radiotherapy. Pre- and post-treatment tissue samples<br />

were analyzed for Bax and p21 protein levels. Results: The median<br />

follow-up time was 28 months. EG achieved a complete response rate (CR)<br />

<strong>of</strong> 62.5% and overall response rate (RR) <strong>of</strong> 90.6%, both rates being<br />

statistically significantly higher than that <strong>of</strong> CG (CR�35.5%, RR�74.2%).<br />

One-year <strong>of</strong> overall survival (OS) was 100.0% for EG and 90.3% for CG,<br />

and one year <strong>of</strong> progress free survival (PFS) was 96.7% and 77.4% for EG<br />

and CG, respectively. Two-year OS was 95.7% and 86.4% for EG and CG,<br />

respectively, and two-year <strong>of</strong> PFS was 79.6% and 73.5% for EG and CG,<br />

respectively. After treatment, both Bax and p21 protein levels in EG<br />

increased significantly. In CG, both proteins levels also increased after<br />

treatment but not significantly. The main side effects <strong>of</strong> rAd-p53 were<br />

self-limited fever, occurring in all <strong>of</strong> EG patients. Conclusions: RAd-p53 as<br />

a component <strong>of</strong> the comprehensive therapy for advanced NPC contributes<br />

significant beneficial effects. Increased levels <strong>of</strong> Bax and p21proteins after<br />

treatment represent two mechanisms <strong>of</strong> p53 anti-tumor activities.<br />

5557 General Poster Session (Board #24A), Sat, 1:15 PM-5:15 PM<br />

Gemcitabine versus cisplatin concurrent with radiation therapy in locally<br />

advanced head and neck squamous cell carcinoma. Presenting Author:<br />

Magda Mostafa, Kasr Al Aini Center <strong>of</strong> <strong>Clinical</strong> Oncology and Radiation<br />

Therapy (NEMROCK), Cairo, Egypt<br />

Background: In locally advanced head and neck squamous cell carcinoma<br />

(HNSCC) weekly cisplatin concurrent with radiation therapy is the standared<br />

treatment. However some patients cannot tolerate cisplatin. So we<br />

conduct a prospective randomized trial comparing cisplatin versus gemcitabine.<br />

Methods: This trial was done in Kasr El-Ainy Center <strong>of</strong> <strong>Clinical</strong><br />

Oncology and Radiation therapy (NEMROCK), during the period from<br />

March 2010 till June 2011. Sixty patients with locally advanced HNSCC<br />

were randomized to receive Cisplatin (30 mg/m2 ) weekly for 6 consecutive<br />

weeks (30 patients) or Gemcitabine (50 mg/m2 ) weekly for 6 consecutive<br />

weeks (30 patients) both concomitant with radiation therapy reaching a<br />

dose <strong>of</strong> 70 Gy over 7 weeks. Primary end points include response rate,<br />

progression free survival and toxicity. Toxicities were graded according to<br />

NCI-CTCAE v3.0. Results: Thewhole study group included 48 (80%) males<br />

and 12 (20%) females. Mean age was 47.9 (� 6.5) years (range 26-61).<br />

Both arms were comparable regarding their age, gender, performance<br />

status and stage. There were 9 (30%) CR, 7 (23.3%) PR, 2 (6.7%) SD and<br />

12 (40%) PD in cisplatin arm versus 12 (40%) CR, 4 (13.3%) PR, 1<br />

(3.3%) SD and 11 (36.7%) PD in gemcitabine arm. Median progression<br />

free survival (PFS) in cisplatin arm was 9 months versus 11months in<br />

gemcitabine arm with a hazard ratio <strong>of</strong> 0.08 (95% CI 0.005 – 1.47). We<br />

did not reach median overall survival. Radiotherapy induced skin toxicity<br />

(slight or patchy atrophy), nausea, vomiting, mucositis, salivary gland<br />

affection and weight loss were equally distributed in both arms. Dysphagia<br />

and fatigue were markedly higher in gemcitabine arm. While infection and<br />

neutropenia were slightly higher in cisplatin arm. Conclusions: Weekly<br />

gemcitabine 50mg/m2 concomitant with radiotherapy was found to be <strong>of</strong><br />

equal efficacy and toxicity comparable with weekly cisplatin in the<br />

treatment <strong>of</strong> locally advanced HNSCC.<br />

5559 General Poster Session (Board #24C), Sat, 1:15 PM-5:15 PM<br />

Outcomes <strong>of</strong> occult primary (OP) <strong>of</strong> the head and neck squamous cell<br />

carcinoma (HNSCC) treated with oropharynx (OPX)-targeted radiotherapy<br />

(RT) and concurrent chemotherapy (CCRT). Presenting Author: Kenneth<br />

Hu, Beth Israel Medical Center, New York, NY<br />

Background: OP <strong>of</strong> HNSCC is commonly treated with comprehensive<br />

mucosal RT including nasopharynx, OPX, hypopharynx and larynx and<br />

bilateral neck. We are reporting the long term outcomes <strong>of</strong> a conservative<br />

mucosal sparing technique consisting <strong>of</strong> targeting only the OPX mucosa<br />

and bilateral necks. Methods: This is a single-institution retrospective<br />

study. From January 1998-2010, a total <strong>of</strong> 68 patients with OP <strong>of</strong> HNSCC<br />

were treated with CCRT (90%) or RT alone (10%), 40% with IMRT. RT<br />

fields comprehended OPX mucosa only and bilateral necks. Gross disease<br />

in the neck received 70Gy, involved neck 63 Gy, OPX 60 Gy, uninvolved<br />

neck and ipsilateral retropharyngeal nodes 54 Gy. All fractions were given<br />

at the rate <strong>of</strong> 1.8-2 Gy/fraction. The median age was 58 (21-87), 80%<br />

Caucasian, and 75% males. Stage IVa (N2) was 75% <strong>of</strong> the population,<br />

while stages III (N1), and IVb (N3) were 9, and 16% respectively. 16<br />

patients (9N3�7 bulky N2) underwent neck dissection (ND). The median<br />

time interval from diagnosis to RT and RT duration were 60 (13-70) and 50<br />

days (49-63) respectively. The Median number <strong>of</strong> Chemotherapy cycles<br />

was 2 (1-3). Cisplatin was given to 70% <strong>of</strong> pts, while Carboplatin and<br />

Cetuximab were 10 and 20% respectively. Results: With a median follow-up<br />

<strong>of</strong> 5.3 years (1– 13.6), the loco-regional control, (LRC) for all stages is 95.6<br />

%. The median time to LRF is 18 months (12 - 63). Chronic CCRT toxicity<br />

was; grade (1) xerostomia (67%), dysphagia (35%), altered taste (28%),<br />

neck stiffness (15%), skin toxicity (12%), dysphonia (9%), and trismus<br />

(6%). One HIV patient developed grade 4 dysphagia. No patients experienced<br />

distant metastases. The emergence <strong>of</strong> primary was (1.5%) 1 patient<br />

developed subglottic SCC 2 years after CCRT, 2 patients failed in the neck<br />

(originally N3) all the 3 patients were salvaged successfully by surgery.<br />

Kaplan-Meier curve shows the 5-year cause-specific survival to be 100.<br />

Conclusions: Our data show that CCRT or definitive RT alone to the OPX and<br />

bilateral neck provides excellent oncologic and functional outcomes.<br />

Sparing the mucosal surfaces <strong>of</strong> the nasopharynx, hypopharynx, and larynx<br />

seems reasonable and likely reduces toxicity.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


5560 General Poster Session (Board #24D), Sat, 1:15 PM-5:15 PM<br />

Acetylated tubulin and risk <strong>of</strong> nodal metastases in squamous cell carcinoma<br />

<strong>of</strong> the head and neck (SCCHN). Presenting Author: Nabil F. Saba,<br />

Winship Cancer Institute, Emory University, Atlanta, GA<br />

Background: We previously established that AT expression predicts for<br />

response to chemotherapy in SCCHN (Saba et al, AACR 2010 meeting<br />

abstr 3744). We wanted to further examine the relation <strong>of</strong> AT expression<br />

and nodal metastases in SCCHN. Methods: We assessed AT expression in<br />

archival tissue specimens from primary SCCHN cases with (50 cases) and<br />

without (53 cases) lymph node metastases (NM) using standard immunohistochemistry<br />

(IHC). <strong>Clinical</strong> characteristics <strong>of</strong> patients were retrieved<br />

from the department <strong>of</strong> pathology under the guidelines <strong>of</strong> the institutional<br />

review board (IRB). IHC staining for AT was scored based on intensity and<br />

percentage <strong>of</strong> tumor cells stained: 0 � no staining, 1� �weak, 2� �<br />

moderate, 3� �moderate to high, 4� �high and a weighted index (WI)<br />

was calculated as percent stain x stain intensity. Wilcoxon two-sample test<br />

and Kruskal-Wallis test were used to estimate the relationships <strong>of</strong> the WI<br />

with nodal metastasis, node status, primary tumor location, grade, and<br />

stage. Log-rank test was used to examine the difference in OS and DFS<br />

among four groups based on quartiles <strong>of</strong> the WI. A Cox proportional hazard<br />

model was employed to estimate the adjusted effect <strong>of</strong> WI on OS and DFS.<br />

Results: A higher AT expression was associated with nodal metastasis<br />

(p�0.0155) and higher stage (p�0.0311). A lower AT expression was<br />

associated with oral cavity primary (p�0.0107). After adjusting for age,<br />

gender, race, and smoking status, a lower AT expression was significantly<br />

associated with better DFS in the subset <strong>of</strong> patients with NM by multivariate<br />

analysis (HR�1.008; 95% CI�1.002- 1.014; p�0.0123). Among<br />

patients with no NM there was a marginally significant difference in OS<br />

among four different groups based on quartiles <strong>of</strong> AT expression<br />

(p�0.0765). Conclusions: High AT expression is associated with nodal<br />

metastases in SCCHN and may be a marker <strong>of</strong> poor prognosis in patients<br />

with node positive disease. The value <strong>of</strong> AT as a prognostic marker in<br />

SCCHN needs to be better defined. This work is supported by a SPORE CDP<br />

Grant P50 CA128613-03 to NFS.<br />

5562 General Poster Session (Board #24F), Sat, 1:15 PM-5:15 PM<br />

Nomogram for prediction <strong>of</strong> prognosis in patients treated for oral cavity<br />

squamous cell carcinoma. Presenting Author: Pablo H. Montero, Head and<br />

Neck Service, Department <strong>of</strong> Surgery, Memorial Sloan-Kettering Cancer<br />

Center, New York, NY<br />

Background: TNM staging is an effective tool for prognostic prediction in<br />

patients with oral cavity cancer, but it disregards variables such as the<br />

anatomic subsite, medical comorbidity, lifestyle, etc. A nomogram has the<br />

ability to take into account many factors predictive <strong>of</strong> outcome for an<br />

individual patient, beyond the traditional TNM. We have developed a<br />

nomogram to accurately predict overall mortality (OM) and disease specific<br />

mortality (DSM) in individual patients treated for oral cancer. Methods:<br />

Demographic, host, and tumor characteristics <strong>of</strong> 1,617 oral cancer<br />

patients treated with surgery and adjuvant treatment between 1985 and<br />

2009 were available from a preexisting database. Recurrent disease was<br />

recorded in 509 patients, 328 died <strong>of</strong> cancer-related causes and 542 died<br />

<strong>of</strong> other causes. The median follow-up was 42 months (range 1-300<br />

months). Cox proportional hazards regression model was used for OM and<br />

competing risk regression was used for DSM. Death from other causes was<br />

treated as competing risk for DSM. Missing values in the predictors were<br />

multiply imputed before analysis. Variables analyzed as prognosis predictors<br />

included age, gender, race, alcohol/tobacco use, anatomic subsite,<br />

comorbidity, tumor size/thickness, bone/deep muscle invasion, nodal<br />

status/level, grade, vascular/perineural invasion, margin status and adjuvant<br />

therapy. Step-down method was used to select the statistically most<br />

powerful predictors for inclusion in the final nomogram for each outcome.<br />

Results: Age, severe comorbidity, subsite, tumor size, bone/deep muscle<br />

invasion, margin status, vascular and perineural invasion, nodal status and<br />

level, and adjuvant therapy were the variables with highest predictive value<br />

for OM. The most influential predictors <strong>of</strong> DSM were gender, tumor size,<br />

nodal status and location, subsite, margin status, grade and vascular<br />

invasion. Nomograms were generated for prediction <strong>of</strong> OM and DSM. The<br />

nomograms were internally validated with correction for over-fitting; biascorrected<br />

concordance index for OM was 75% and DSM 76%. Conclusions:<br />

We have developed nomograms that can accurately estimate OM and DSM<br />

based on tumor and host characteristics <strong>of</strong> an individual patient treated for<br />

oral cancer.<br />

Head and Neck Cancer<br />

371s<br />

5561 General Poster Session (Board #24E), Sat, 1:15 PM-5:15 PM<br />

Patient-reported symptoms following IMRT alone for oropharynx cancer: A<br />

survivorship study. Presenting Author: Gary Brandon Gunn, University <strong>of</strong><br />

Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: Patients (pts) with favorable oropharynx cancer (OPC) are<br />

<strong>of</strong>ten treated with single modality RT with excellent disease control. We<br />

present the pattern <strong>of</strong> pt-reported symptoms in OPC survivors treated with<br />

IMRT alone. This will represent a benchmark for later comparison to pts<br />

treated with other modalities (e.g., proton therapy, transoral surgery).<br />

Methods: Pts eligible for this cross-sectional questionnaire-based study had<br />

OPC treated with IMRT (no systemic therapy), in remission �18 mos,<br />

completed the MD Anderson Symptom Inventory – Head and Neck Module<br />

(MDASI-HN). <strong>Clinical</strong> data were tabulated and analyzed using nonparametric<br />

statistics. Results: 89 pts participated. OPC sub-site was tonsil<br />

in 58, base <strong>of</strong> tongue (BOT) in 30, and s<strong>of</strong>t palate in 1 pt. 65% were TX/1,<br />

28% T2, and 7% T3/4, while 53% were NX/1 with 47% N2. Mean RT dose<br />

was 66 Gy (SD�2.5). Median age was 54 years (SD�11). Mean follow-up<br />

at MDASI-HN was 74 months (SD�15). Mean MDASI-HN symptom rating<br />

(0 to 10 scale) was 1.3 (95% CI 0.97-1.6), with no significant difference<br />

by OPC sub-site. No single MDASI-HN items showed significant difference<br />

in the percentage <strong>of</strong> pts with moderate to severe (M/S) reports by OPC site,<br />

thou dry mouth approached significance (p�0.054), 53% in BOT vs. 32%<br />

for tonsil. Overall, 14% <strong>of</strong> pts were entirely symptom free (i.e. “0” across all<br />

22 symptom items), 33% reported only mild symptoms, 23% only<br />

moderate symptoms, and 31% reported any symptom as severe . The top 7<br />

symptoms were dry mouth, swallowing, sleep, taste, fatigue, distress, and<br />

teeth and these were reported at M/S levels by 39%, 24%, 19%, 17%,<br />

16%, 14%, and 10% <strong>of</strong> pts, respectively. For those with any M/S symptom,<br />

the median number <strong>of</strong> symptoms reported at M/S levels was 3. 1/89 pts had<br />

a feeding tube at the time <strong>of</strong> MDASI-HN completion. Conclusions: The late<br />

symptom burden for long-term OPC survivors following IMRT alone was low,<br />

with nearly half <strong>of</strong> pts with only minimal symptoms across all items.<br />

Considering other endpoints (e.g. disease control, acute toxicity, second<br />

malignancies, and functional measures), future treatment strategies should<br />

seek to match or exceed these results. Future RT symptom prevention/<br />

reduction strategies should focus on the top symptoms identified here.<br />

5563 General Poster Session (Board #24G), Sat, 1:15 PM-5:15 PM<br />

Head and neck cancer in Fanconi anemia and dyskeratosis congenita.<br />

Presenting Author: Blanche P. Alter, National Cancer Institute , Bethesda,<br />

MD<br />

Background: Fanconi Anemia (FA) and dyskeratosis congenita (DC) are<br />

inherited bone marrow failure syndromes (IBMFS) with extraordinarily high<br />

risks <strong>of</strong> cancer, particularly head and neck squamous cell carcinoma<br />

(HNSCC). The standardized incidence ratios (SIR) for solid tumors are 40<br />

in FA and 10 in DC, and for HNSCC 700 and 900 respectively. In the<br />

general population, cancers <strong>of</strong> the oropharynx may be associated with<br />

human papilloma virus (HPV) more than the oral cavity. The specific<br />

locations <strong>of</strong> HNSCC in FA and DC have not been reviewed, and thus the<br />

hypothetical role <strong>of</strong> HPV in these cancers is unknown. Methods: Literature<br />

cases were reviewed through PubMed searches using the terms “Fanconi<br />

anemia” and “dyskeratosis congenita”. All papers were reviewed, and<br />

details <strong>of</strong> the specific cancers recorded. FA literature included reports from<br />

1927 through 2011, and DC literature from 1910 through 2011. <strong>Part</strong>icipants<br />

in the National Cancer Institute (NCI) IBMFS retrospective/<br />

prospective protocol (NCI 02-C-0052, opened in 2002) were also analyzed<br />

for cancer. The HPV vaccine was not available during most <strong>of</strong> the time<br />

frame <strong>of</strong> this study. Results: There were 411 cases <strong>of</strong> cancer among 2190<br />

(19%) cases <strong>of</strong> FA described individually in the literature, and 21 cancer<br />

cases out <strong>of</strong> 116 (18%) FA patients in the NCI cohort. HNSCC were<br />

reported in 112 FA literature cases (5%), and 9 (8%) in the cohort. The<br />

oropharynx was the initial site for HNSCC in 31/112 literature cases (28%)<br />

and 3/9 NCI cases (33%). With regard to DC, 51/647 (8%) cases in the<br />

literature had cancer, as did 9 <strong>of</strong> 94 (10%) NCI participants. HNSCC were<br />

reported in 21/647 (3%) DC literature cases, and in 5/94 (5%) from the<br />

NCI cohort. Four <strong>of</strong> the 21 HNSCC (19%) in the literature were oropharyngeal,<br />

and 2/5 (40%) in the NCI cohort. The cumulative incidences <strong>of</strong> any<br />

HNSCC in the NCI FA and DC cohorts were 50% and 21% by 40 years <strong>of</strong><br />

age (censored at death from other causes). Conclusions: Patients with FA<br />

and DC have extremely high risks <strong>of</strong> HNSCC at substantially younger ages<br />

than the general population. Approximately 30% <strong>of</strong> these cancers occur in<br />

the oropharynx. The HPV vaccine could be important in prevention <strong>of</strong> these<br />

cancers. Future studies <strong>of</strong> the HPV status <strong>of</strong> oral cavity and oropharynx<br />

HNSCC tumors from patients with FA and DC will be informative.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


372s Head and Neck Cancer<br />

5564 General Poster Session (Board #24H), Sat, 1:15 PM-5:15 PM<br />

Should age affect our treatment decisions for head and neck cancer?<br />

Presenting Author: Estrella M Carballido, H. Lee M<strong>of</strong>fitt Cancer Center &<br />

Research Institute, Tampa, FL<br />

Background: Current opinion suggests elderly patients (pts) with head and<br />

neck cancer, those 65 or older, do not tolerate surgery, chemotherapy, or<br />

radiation as well as their younger counterparts. If this holds true, elderly pts<br />

may not be <strong>of</strong>fered standard treatments to prevent assumed complications.<br />

Methods: A retrospective cohort study at our comprehensive cancer center<br />

was conducted <strong>of</strong> newly diagnosed pts with head and neck squamous cell<br />

carcinoma to explore differences in treatment-related complications between<br />

older and younger groups. We included data from the first 199<br />

eligible pts (99 younger than 65 year old and 100 older than 65) evaluated<br />

between April 2009 and June 2010. Results: 79% <strong>of</strong> pts receiving<br />

treatment were male with a mean age <strong>of</strong> 54.9 and 71.6 years for the<br />

younger and older groups respectively. The older group had significantly<br />

more comorbidities (p � 0.001). The majority <strong>of</strong> older pts presented with<br />

oral cavity tumors (46%) while the oropharynx was the predominant site in<br />

the younger group (45%). 55% <strong>of</strong> younger and 49% <strong>of</strong> older pts presented<br />

with stage 4 disease across all sites. A total <strong>of</strong> 51 pts were p16 positive with<br />

no statistical differences between the groups. Surgery was the initial<br />

treatment for 57% <strong>of</strong> older pts (p � 0.008) while 46% <strong>of</strong> younger pts<br />

received concurrent chemotherapy and radiation as the primary treatment<br />

(p � 0.008). There was no statistically significant difference in surgical or<br />

radiation complications between the groups. Although most pts receiving<br />

chemotherapy experienced complications, older pts had slightly more<br />

(93% vs. 78%; p�0.031). The mean survival was 24.8 months with no<br />

statistical difference between groups. Significantly more pts in the older<br />

group, at last follow-up, were disease free (p � 0.012). Conclusions: The<br />

treatment <strong>of</strong> elderly pts with head and neck squamous cell carcinoma in our<br />

experience was congruent with that <strong>of</strong> younger pts. Elderly pts did not<br />

suffer more complications with surgery or radiation, however chemotherapy<br />

produced somewhat more complications in the elderly pts. Elderly pts did<br />

display less evidence <strong>of</strong> disease on follow-up. Age is always a consideration<br />

when treating individuals, but should not preclude the curative standard.<br />

5566 General Poster Session (Board #25B), Sat, 1:15 PM-5:15 PM<br />

ECOG 1308: A phase II trial <strong>of</strong> induction chemotherapy followed by<br />

cetuximab with low dose versus standard dose IMRT in patients with<br />

HPV-associated resectable squamous cell carcinoma <strong>of</strong> the oropharynx<br />

(OP). Presenting Author: Shanthi Marur, The Johns Hopkins University<br />

School <strong>of</strong> Medicine, Baltimore, MD<br />

Background: Human papillomavirus (HPV) infection is now recognized as a<br />

risk factor <strong>of</strong> oropharyngeal squamous cell carcinoma (OPSCC) and<br />

approximately 60% <strong>of</strong> OPSCC are HPV positive (�). ECOG 2399 using<br />

induction chemotherapy (IC) followed by chemoradiation demonstrated<br />

significantly improved 2-yr progression-free survival (PFS; 85% vs. 50%,<br />

p�0.05) and overall survival (OS; 94% vs. 58%p�0.004). These results<br />

have generated interest in development <strong>of</strong> less toxic regimens for HPV(�)<br />

patients with lower dose radiation. Methods: Design: This is a phase II<br />

clinical trial .The primary objective is the estimation <strong>of</strong> the 2-year PFS in<br />

the reduced dose RT arm. Secondary objectives include toxicity, OS,<br />

objective response, quality <strong>of</strong> life (QOL) and correlative studies <strong>of</strong> biomarkers.<br />

Eligibility Criteria: Patients with Stage III or IV resectable HPV(�)<br />

OPSCC. HPV(�) disease is defined as p16 IHC strongly (�) on at least 70%<br />

<strong>of</strong> cells, and/or HPV-16 ISH (�).Treatment : Patients received IC with a<br />

combination <strong>of</strong> paclitaxel 90mg/m2 on days 1,8, and 15, cisplatin<br />

75mg/m2 on day 1, and cetuximab loading dose <strong>of</strong> 400 mg/m2 on day 1,<br />

cycle 1 followed by cetuximab 250 mg/m2 weekly, thereafter. Each cycle<br />

was 21 days for a total <strong>of</strong> 3 cycles. After completion <strong>of</strong> IC, patients were<br />

assessed clinically with a complete head and neck exam and imaging<br />

studies. Those with clinical CR at primary site received radiation to 54Gy<br />

plus cetuximab. Patients with clinical PR or SD at primary site received<br />

standard dose RT with cetuximab. Correlative Studies: The expression <strong>of</strong><br />

biomarkers on tumor and blood samples and QOL parameters will be<br />

correlated with the clinical outcomes. Results: Current enrollment: Target<br />

accrual was 83 patients. The study accrued 90 patients from March 17,<br />

2010 to accrual on October 19, 2011. Baseline characteristics reveal;<br />

median age 57 years, 94% male, 95% white, 47% never smokers, 10%<br />

�10 pack year (pyr) smoking, 38% �10 pyr smoking, 84% not current<br />

smokers, 39% node (N) stage N2b and 29% N2c, 22% tumor (T) stage T1,<br />

52% T2, 16% T3, and 10% T4. Conclusions: Enrollment has just<br />

completed with no data analysis available.<br />

5565 General Poster Session (Board #25A), Sat, 1:15 PM-5:15 PM<br />

EGFR-KRAS genotyping and EGFR-CDX2 expression in sinonasal intestinal<br />

type adenocarcinomas: Toward a new targeted therapy? Presenting Author:<br />

Olivier Choussy, CHU Rouen-Service ORL, Rouen, France<br />

Background: Sinonasal carcinomas are very rare tumors which are sometimes<br />

not eligible for curative surgery. Alternative therapies (i.e. conventional<br />

chemotherapies/radiotherapy) are used but with poor results.<br />

Therefore, the need for a more efficient treatment is mandatory. Aims <strong>of</strong> our<br />

study: Comparing EGFR and KRAS genetic pr<strong>of</strong>iles, EGFR and CDX2<br />

phenotypes <strong>of</strong> sinonasal intestinal type adenocarcinomas (ITAC) with<br />

colorectal adenocarcinomas (CRC). Methods: 41 patients were treated in<br />

our institution between 1983 and 2007. All pathological specimens were<br />

reclassified according to the 2005 WHO classification. An immunohistochemical<br />

(IHC) study was carried out for EGFR and CDX2 expression. We<br />

were able to analyze 38 <strong>of</strong> the 41 specimens for KRAS and EGFR<br />

mutations. SNaPshot multiplex system was used to determine the presence<br />

<strong>of</strong> the most common mutations which are located in exon 18, 20 and 21 for<br />

EGFR and in exon 2 (codon 12 and13) for KRAS. Fragment analysis<br />

method was used for EGFR exon 19 deletions. Results: Thirty five <strong>of</strong> the 38<br />

patients were classified as ITAC (33 men and 2 women). The mean age was<br />

64.5 years. Exposure to wood work was found in 29 cases (85%). CDX2<br />

expression was present in 31 (89%) cases <strong>of</strong> ITAC and absent in all non<br />

intestinal adenocarcinomas (3 cases). EGFR was expressed in 29 ITACs<br />

(83%) with various degrees <strong>of</strong> IHC expression: 19 (56 %) 1�, 7 (21%) 2�,<br />

and 3 with 3� immunopositivity. No EGFR mutation was found in the<br />

whole population; 5 ITAC patients (14%) disclosed KRAS mutations.<br />

Conclusions: Histological, phenotype and genetic pr<strong>of</strong>iles <strong>of</strong> ITAC are very<br />

similar to those <strong>of</strong> colorectal adenocarcinoma. These results suggest that<br />

ITACs with wt KRAS could respond to anti MoAb anti-EGFR therapy in the<br />

same way as metastatic CRC. We propose that all sinonasal tumors should<br />

undergo: firstly, CDX2 IHC in order to confirm the ITAC histological subtype<br />

and then KRAS genotyping to select the wt population which could benefit<br />

from such anti EGFR targeted therapy.<br />

5567 General Poster Session (Board #25C), Sat, 1:15 PM-5:15 PM<br />

Young adults with squamous cell head and neck cancer: Ten years <strong>of</strong> a<br />

single institution’s experience (1999-2008). Presenting Author: Marcos<br />

Pantarotto Alves, Instituto Portugues Oncologia Francisco Gentil, Oporto,<br />

Porto, Portugal<br />

Background: Head and neck cancer <strong>of</strong> squamous cell type (SCHNC) is<br />

characterised by its aggressiveness and strong relationship with smoking<br />

and drinking habits. It is generally diagnosed in locally advanced stages,<br />

mainly in middle-aged men. People younger than 35yo are rarely affected<br />

by this disease, and little is known about its behaviour among young<br />

Portuguese adults. Our aim was to describe the clinical and epidemiological<br />

features <strong>of</strong> young adults with diagnosis <strong>of</strong> SCHNC in a single<br />

oncological centre in Portugal. Methods: Retrospective cohort including<br />

clinical registries’ databases for patients (pts) with diagnosis <strong>of</strong> SCHNC,<br />

with 3391 eligible pts from 1999 to 2008. Pts aged �18yo and �35yo at<br />

diagnosis with histopathological diagnosis in our Institution were included.<br />

Pts admitted to radiation therapy with no follow-up information were<br />

excluded. 39 pts fulfilled the criteria described, comprising the study<br />

population. Statistical analysis was carried out using SPSS v19.0 for Mac<br />

(IBM, 2010, EUA), with values <strong>of</strong> p�0,05 considered significant. Results:<br />

Male sex predominance (82%) was observed. The median age <strong>of</strong> presentation<br />

was 33 yo [22; 35], with 90% smokers with a median <strong>of</strong> 18,5<br />

package-years [1; 48]. Alcohol consumption was mild to moderate (n�9;<br />

23%) or heavy (n�15; 38%). The median <strong>of</strong> follow-up time was <strong>of</strong> 24<br />

months [1;151]. Locally advanced diseased was present at diagnosis in 24<br />

pts, where localised disease was commonest among women (57%) than in<br />

men (29%). Median overall survival (OS) was not reached on this<br />

population, although K-M graphs seems to show a significant difference in<br />

survival between sexes. Alcohol consumption was the most significant<br />

variable to influence the survival (p�0,01). Conclusions: SCHNC in young<br />

people is a rare condition, with distinct epidemiological and clinical<br />

features. Women seems to have greater survival rates, which may be<br />

explained by other risk factors already reported for this gender, namely HPV<br />

infection. The finding <strong>of</strong> an apparent relationship between alcohol consumption<br />

and OS, and the high prevalence <strong>of</strong> alcoholic intake habits on this<br />

parcel <strong>of</strong> the population, urge the need <strong>of</strong> development <strong>of</strong> public health<br />

programs addressed to this specific age group.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


5568 General Poster Session (Board #26A), Sat, 1:15 PM-5:15 PM<br />

Feasibility and short-term efficacy <strong>of</strong> four facio-cervical fields’ conformal<br />

radiotherapy for nasopharyngeal carcinoma. Presenting Author: Wang fang<br />

Zheng, Zhejiang Cancer Hospital, Hangzhou, China<br />

Background: Conventional bilateral facio-cervical fields increased the local<br />

control and overall rates for nasopharyngeal carcinoma, and generated<br />

severe late complications, while intensity-modulated radiation therapy<br />

(IMRT) may take up a lot <strong>of</strong> labor power and material resources.This study<br />

was to explore feasibility and short-term efficacy <strong>of</strong> four facio-cervical<br />

fields’ conformal radiotherapy (4F-CRT) for nasopharyngeal carcinoma.<br />

Methods: Between November 2007 and June 2009, 101 patients with<br />

histology-proven nonmetastatic NPC were rolled into this study and<br />

received 4F-CRT. 90 <strong>of</strong> all patients received four cycles chemotherapy.<br />

Results: The treatment was completed for all patients within 7 weeks. The<br />

complete response (CR) rate was 95.0% (96/101) and the partial response<br />

(PR) rate 5.0% (5/101). Overall response was 100% for nasopharyngeal<br />

lesions. The CR rate was 90.0% (91/101) and the PR rate 7.0% (7/101) in<br />

lymph node lesions. One year local control, regional control, overall survival<br />

(OS) rate, metastasis-free survival (MFS) and disease-free survival (DFS)<br />

rate are 100%, 100%, 100%, 97%, 97%, respectively. The major<br />

toxicities observed were grades 1-2 leukocytopenia. Conclusions: 4F-CRT<br />

can generate desirable dose distribution <strong>of</strong> tumor volume for nasopharyngeal<br />

carcinoma. The short-term efficacy <strong>of</strong> 4F-CRT is satisfactory and its<br />

acute toxicities are tolerable.<br />

5570 General Poster Session (Board #26C), Sat, 1:15 PM-5:15 PM<br />

Paranasal sinus squamous cell carcinomas (PNSSCC) incidence and<br />

survival trends. Presenting Author: Benjamin Ansa, Morehouse School <strong>of</strong><br />

Medicine , Atlanta , GA<br />

Background: PNSSCC are rare, accounting for 3 percent <strong>of</strong> all head and<br />

neck malignancies. They generally present with local invasion and have a<br />

high rate <strong>of</strong> local recurrence. There has been little information on the trends<br />

in incidence and survival <strong>of</strong> PNSSCC and no randomized trials to guide<br />

therapy. We analyzed the incidence rates and survival outcomes for<br />

patients diagnosed with PNSSCC in the SEER database. Methods: Data<br />

were obtained from the U.S. National Cancer Institute’s Surveillance,<br />

Epidemiology, and End Results (SEER) Program. Newly diagnosed PNS-<br />

SCC reported to SEER from 1973 through 2008 were categorized according<br />

to the patient’s sex, age, year <strong>of</strong> diagnosis, and stage. The incidence<br />

and survival were then examined and compared across different demographic<br />

and disease-related categories. Results: A total <strong>of</strong>2,323 patients<br />

were identified. The mean age was 62 years, range: 14-85 yrs; malesaccounted<br />

for 62.7%. Whites accounted for 75.8%, blacks for 12.2 % and<br />

12.0% were <strong>of</strong> asian or pacific islanders and other groups. Maxillary sinus<br />

primary accounted for 76.3%, ethmoid (10.2%), sphenoid (4.3%). The<br />

disease was localized in 9.5%, had regional spread in 72.0% and distant<br />

disease in 12.2%, at diagnosis. A statistically significant decrease (<strong>Annual</strong><br />

Percent Change <strong>of</strong> -1.6%, 95%CI� [-2.0, -1.1]) in the overall incidence<br />

was observed by Joinpoint analysis (p�0.01). The decrease in incidence<br />

was significant in patients with maxillary sinus (p�0.01) and was more<br />

pronounced in the white population compared to other groups. Both the<br />

observed and relative 5-year survival significantly increased over time and<br />

this was more notable in the white population compared to other groups.<br />

The observed 5-year survival rate increased from 24.9% (95% CI �<br />

[20.3-29.6]) during the 1975-1983 time-period to 40.8% (95% CI �<br />

[33.4-48.0]) during the most recent period <strong>of</strong> 2002-2008. Conclusions:<br />

There has been a decline in the overall incidence <strong>of</strong> PNSSCC more notably<br />

in the white population and for patients with maxillary sinus tumors. The<br />

5-year observed and relative survival has significantly increased. Our<br />

findings should incite more efforts to understand the factors behind the<br />

change in incidence and survival.<br />

Head and Neck Cancer<br />

373s<br />

5569 General Poster Session (Board #26B), Sat, 1:15 PM-5:15 PM<br />

Effects <strong>of</strong> vandetanib on body composition in patients with advanced<br />

medullary thyroid carcinomas: Results from a placebo-controlled study.<br />

Presenting Author: Marie-Helene Massicotte, Institut Gustave Roussy,<br />

Nuclear Medicine and Endocrine Oncology Service, Villejuif, France<br />

Background: Muscle (MT) and adipose tissue (AT) share common intra<br />

cellular pathways with tumor, thus it is not surprising to observe metabolic<br />

consequences with targeted therapies. The aim <strong>of</strong> the study was to assess<br />

the effects <strong>of</strong> vandetanib, a tyrosine kinase inhibitor (TKI) which demonstrated<br />

efficacy for the treatment <strong>of</strong> advanced medullary thyroid carcinoma<br />

(MTC), on MT and AT. Methods: 33 patients (25 men and 8 women, mean<br />

age <strong>of</strong> 54 years) with metastatic MTC received vandetanib 300 mg/d<br />

(n�23) or placebo (n�10) in the setting <strong>of</strong> the ZETA study. Cross-sectional<br />

areas (cm2 ) <strong>of</strong> visceral adipose tissue (VAT), subcutaneous adipose tissue<br />

(SAT) and MT were assessed by computed tomography imaging at 3rd<br />

lumbar vertebra and were indexed for height (cm2 /m2 ). Comparisons<br />

between treatment and placebo were made at 3 months, and long-term<br />

evolution was evaluated over 12 months. Results: At 3 months, compared to<br />

baseline, patients treated with vandetanib gained 1.5 kg, 1.3 cm2 /m2 <strong>of</strong><br />

MT, 5.1 cm2 /m2 <strong>of</strong> VAT and 4.5 cm2 /m2 <strong>of</strong> SAT. In contrast, patients under<br />

placebo lost 1.5 kg (p�0.02), 1.0 cm2 /m2 <strong>of</strong> MT (p�0.009), 5.5 cm2 /m2 <strong>of</strong> SAT (p�0.004) and gained significantly less VAT (0.6 cm2 /m2 )<br />

(p�0.02). At 12 months, compared to baseline, patients treated with<br />

vandetanib gained 2 kg (NS), lost 0.3 cm2 /m2 MT (NS), gained 3.4 cm2 /m2 <strong>of</strong> VAT (NS) and 8.7 cm2 /m2 <strong>of</strong> SAT (95% CI, 1.1 to 16.2). A significant<br />

decrease <strong>of</strong> calcitonin (defined as �50% compared to baseline) was<br />

associated with higher weight (p�0.01), VAT (p�0.01), and total adipose<br />

tissue (p�0.02). Conclusions: Beyond its proved efficacy in MTC treatment,<br />

and despite common intracellular pathways, vandetanib is the only studied<br />

TKI to preserve MT and to restore AT. Further research is needed to explore<br />

whether the relationship between changes <strong>of</strong> VAT and vandetanib treatment<br />

results from a direct metabolic action <strong>of</strong> vandetanib or is a<br />

consequence <strong>of</strong> biochemical tumor control.<br />

5571 General Poster Session (Board #27A), Sat, 1:15 PM-5:15 PM<br />

Impact <strong>of</strong> p16 status on the QOL effects <strong>of</strong> chemoradiation for locally<br />

advanced oropharynx cancer: Results <strong>of</strong> TROG 02.02. Presenting Author:<br />

Jolie Ringash, Princess Margaret Hospital and University <strong>of</strong> Toronto,<br />

Toronto, ON, Canada<br />

Background: We report the impact <strong>of</strong> p16 status on quality <strong>of</strong> life (QOL) for<br />

patients with stage III or IV (excluding T1-2N1 and M1) squamous cell<br />

carcinoma <strong>of</strong> the oropharynx (OPC) treated with concurrent chemoradiotherapy<br />

in a large international phase III trial (TROG 02.02/HeadSTART).<br />

Methods: The 861 patients accrued received definitive radiotherapy (RT)<br />

(70 Gy/7 weeks) concurrently with 3 cycles <strong>of</strong> either cisplatin (100mg/m2 )<br />

or cisplatin (75 mg/m2 ) plus tirapazamine (290 mg/m2 /day) by random<br />

assignment, as previously described. QOL was measured with the FACT-<br />

H&N at baseline, 2,6,12, 23 and 38 months. No significant difference in<br />

overall or subscale QOL score change from baseline was observed between<br />

arms at any subsequent time point; results for the oropharynx subgroup by<br />

p16 status are reported for both treatment arms combined. Results: Of 853<br />

eligible participants, 465 had OPC, for whom p16 status could be<br />

determined in 206. Of 179 who received adequate RT (� 60 Gy, no major<br />

deviations) and completed baseline QOL, 104 were p16� and 79 were<br />

p16-. p16� patients had better baseline ECOG PS, lower T-category,<br />

higher N-category, were younger and were less likely to be current smokers.<br />

Baseline mean FACT-H&N score was statistically and clinically significantly<br />

better in p16� patients (111 vs. 102, p�0.001). The drop in QOL<br />

from baseline to 2 months was more severe in p16� cases (-20.4 vs -9.1,<br />

p�0.001), resulting in an equalization <strong>of</strong> 2 month scores (p16�: 90.6,<br />

p16-: 93.6, p�0.16). At 6 and 12 months post-treatment, no difference in<br />

score changes from baseline by p16 status was seen (6 mo, p16�: -6.2,<br />

p16 -:-1.2, p�0.22; 12 mo, p16 �: -0.3, p16 -: �2.0, p�0.82).<br />

Conclusions: p16 associated oropharyngeal cancer has been shown to be a<br />

distinct entity with different demographic features. In our study, such<br />

patients exhibited better baseline QOL and a more severe drop immediately<br />

after treatment, but did not differ in long-term QOL response to the effects<br />

<strong>of</strong> aggressive concurrent chemoradiation. Given the favorable prognosis <strong>of</strong><br />

p16-associated oropharyngeal cancer, efforts to reduce the QOL burden <strong>of</strong><br />

treatment are warranted.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


374s Head and Neck Cancer<br />

5572 General Poster Session (Board #27B), Sat, 1:15 PM-5:15 PM<br />

Correlation between PI3K/PTEN/AKT/mTOR pathway genetic variations<br />

and clinical outcome in patients with squamous cell carcinoma <strong>of</strong> the head<br />

and neck receiving cetuximab-docetaxel treatment. Presenting Author:<br />

Alberto Fusi, Department <strong>of</strong> Hematology and Medical Oncology, Charité,<br />

CBF, Berlin, Germany<br />

Background: The PI3K/PTEN/AKT/mTOR pathway is one <strong>of</strong> the most<br />

targeted in human cancers and alterations in the axis are frequent events in<br />

squamous cell carcinoma <strong>of</strong> the head and neck (HNSCC). Specific genetic<br />

variations in this pathway have been associated with variation in recurrence,<br />

survival and response in lung cancer and esophageal cancer. The<br />

aim <strong>of</strong> this study was to determine whether single nucleotide nucleotide<br />

polymorphisms (SNPs) in AKT1, AKT2, FRAP1, PI3KCA and PTEN<br />

identified in other cancers were associated with treatment response and<br />

clinical outcome in patients with HNSCC. Methods: Genomic DNA was<br />

extracted from FFPE tissue specimens <strong>of</strong> 45 patients with recurrent or<br />

initially metastatic HNSCC who received a combined treatment with<br />

docetaxel and cetuximab in a phase II study. Eleven single SNPs in the<br />

genes AKT1, AKT2, FRAP1 (mTOR), PIK3CA and PTEN were genotyped by<br />

means <strong>of</strong> Real Time PCR system or by direct sequencing and analysed for<br />

association with response to therapy and survival. Results: None <strong>of</strong> the<br />

SNPs was related to treatment response. Two SNPs (AKT2:rs8100018 and<br />

PTEN:rs12569998) were associated with variation in progression risk. The<br />

AKT2:rs8100018 homozygous variant resulted in increased risk, with a HR<br />

<strong>of</strong> 4.83 (95% CI, 1.11-21.03) and the PTEN:rs12569998 homozygous<br />

variant in an increased risk, with a HR <strong>of</strong> 2.36 (95% CI, 1.24-4.50).<br />

AKT2:rs8100018 and PTEN:rs12569998 genetic variation had an additive<br />

effect on risk <strong>of</strong> tumor progression. The AKT2:rs8100018 homozygous<br />

variant was significantly associated with overall survival (OS) with HR <strong>of</strong><br />

3.57 (95% CI, 1.06-12.00) while presence <strong>of</strong> one <strong>of</strong> the two variant alleles<br />

<strong>of</strong> AKT1:rs3803304 with a lower risk <strong>of</strong> death (HR: 0.51; 95% CI,<br />

0.27-0.97). Conclusions: We found significant associations between common<br />

genetic variants in the PI3K/PTEN/AKT/mTOR pathway and outcome<br />

in patients with HNSCC. Despite the small sample size, patient cohort and<br />

treatment were homogeneous. As a consequence, evaluation <strong>of</strong> SNPs as a<br />

host factor may be considered in addition to tumor-specific mutations for<br />

personalized treatment development.<br />

5574 General Poster Session (Board #28A), Sat, 1:15 PM-5:15 PM<br />

Genetic alterations in HRAS gene in relation to outcome and response to<br />

cetuximab in head and neck squamous cell carcinoma. Presenting Author:<br />

Theodoros Rampias, Yale University School <strong>of</strong> Medicine, New Haven, CT<br />

Background: Aberrant signaling through RAS/MAPK pathway is implicated<br />

in resistance to EGFR-targeted agents in cancer. Genetic alterations in<br />

Hras gene such as mutations and specific polymorphisms are associated<br />

with aggressive phenotype in several smoking-related malignancies. We<br />

sought to determine the impact <strong>of</strong> Hras genetic alterations on response to<br />

cetuximab and prognosis in HNSCC. Methods: <strong>Clinical</strong> outcome according<br />

to Hras status was investigated in a retrospective cohort <strong>of</strong> 140 HNSCC<br />

specimens. Primary endpoints were overall survival (OS) and disease-free<br />

survival (DFS) and secondary endpoint was treatment response. For<br />

statistical analysis, T-test was used for continuous data and x2 –test for<br />

categorical data. Cetuximab-resistant cell lines harboring mutant Hras<br />

(BB49, T24) were infected with lentivirus expressing shRNA targeting the<br />

Hras or a scrambled- shRNA. MTT assay was used to determine the effect <strong>of</strong><br />

cetuximab on growth <strong>of</strong> lentivirus infected cells. Biochemical analysis<br />

involved immunoblotting for pERK1/2. Results: Mutationanalysis <strong>of</strong> tumor<br />

samples showed that 5.7% participants harbored Hras mutations and<br />

16.42% harbored Hras polymorphisms (rs12628, rs41258054) that are<br />

associated with tumorigenesis. Patients bearing tumors with mutated Hras<br />

had inferior mean OS ( 22.13vs 35.20, p�0.02) and a non-significant<br />

trend for inferior mean DFS. Patients with tumors containing Hras genetic<br />

alterations (mutation or polymorphism) had significantly inferior mean OS<br />

(p�0.02) compared to those harboring wt Hras and trended towards<br />

inferior DFS (p�0.07). Patients had received various treatments such as<br />

surgery plus/minus RT and various chemotherapy regimens. A subgroup<br />

analysis <strong>of</strong> 38 patients treated with cetuximab-based regimens showed that<br />

wt Hras was associated with higher likelihood <strong>of</strong> attaining CR or PR to<br />

treatment <strong>of</strong> borderline significance (p�0.06) due to small sample size.<br />

Silencing <strong>of</strong> Hras in Hras-mutant cell lines restored sensitivity to cetuximab<br />

and caused a direct downregulation <strong>of</strong> pERK1/2 levels. Conclusions: Hras<br />

genetic alterations are associated with aggressive clinical course and may<br />

affect response to cetuximab in HNSCC.<br />

5573 General Poster Session (Board #27C), Sat, 1:15 PM-5:15 PM<br />

Correlation <strong>of</strong> microbiomic pr<strong>of</strong>iles with disease status and MDR1 methylation<br />

in head and neck squamous cell carcinoma (HNSCC). Presenting<br />

Author: Pauline Funchain, Cleveland Clinic, Cleveland, OH<br />

Background: Recent studies <strong>of</strong> the aerodigestive tract have uncovered the<br />

functional importance <strong>of</strong> the microbiome, the total collection <strong>of</strong> microorganisms<br />

that reside within the human host. Furthermore, specific bacterial<br />

species have been shown to be etiologic agents or show potential as a<br />

screening biomarker in cancers including gastric, colon and pancreas.<br />

Here, we hypothesize that specific microbial populations may contribute to<br />

HNSCC pathogenesis via epigenetic modifications in inflammatory- and<br />

HNSCC-associated genes. Methods: Matched tumor and adjacent normal<br />

fresh frozen tissue specimens were collected from 55 prospectively<br />

enrolled HNSCC patients. Microbiomic pr<strong>of</strong>iles were obtained using Sanger<br />

sequencing <strong>of</strong> 16S rDNA PCR products. Methylation status <strong>of</strong> four genes<br />

previously linked to HNSCC or inflammation (MDR1, IL8, RARB, TGFBR2)<br />

was assessed in 49 samples by combined bisulfite restriction analysis and<br />

by Illumina HumanMethylation27 chip. Principle component analysis and<br />

multivariate analysis <strong>of</strong> covariance (MANCOVA) were used to identify<br />

bacterial subpopulations significantly associated with HNSCC and relevant<br />

clinical variables. Results: In this cohort, median age is 62, male:female<br />

ratio is 3:2, 16% are HPV�. Specific bacterial subpopulations associate<br />

with HNSCC over normal tissue and correlate with larger tumor size and<br />

higher nodal stage (p�0.05). Furthermore, microbial populations can<br />

separate tumors by tobacco (p�0.005) but not alcohol (p�0.7) status.<br />

Bacterial pr<strong>of</strong>iles are independent <strong>of</strong> HPV status. MDR1 promoter methylation<br />

is seen in tumor samples but not in normal oral mucosa (22/49 vs<br />

0/49), and associates with specific microbiomic subpopulations in the<br />

order Enterobacteriales and phylum Tenericutes (p�0.001). Additionally,<br />

MDR1 methylation correlates with regional nodal metastases. Conclusions:<br />

Specific bacterial subpopulations differ between normal and tumor tissue<br />

and show association to specific clinicopathologic features, suggesting a<br />

role for microbial pr<strong>of</strong>iling in HNSCC as a novel area <strong>of</strong> investigation for<br />

diagnosis, prognostication, and therapeutic targeting.<br />

5575 General Poster Session (Board #28B), Sat, 1:15 PM-5:15 PM<br />

Identification <strong>of</strong> head and neck cancers (SCCHN) that may respond to dual<br />

inhibition <strong>of</strong> EGFR and HER3 signaling. Presenting Author: David S.<br />

Shames, Genentech, South San Francisco, CA<br />

Background: Oncogenic signaling through the epidermal growth factor<br />

receptor family is one <strong>of</strong> the most frequent alterations found in human<br />

epithelial cancers. These receptor tyrosine kinases mediate their effects via<br />

high-level co-expression and homo- and heterodimerization events that<br />

drive tumor growth, metastasis, and survival. Extensive preclinical studies<br />

suggested that some cell lines depend on oncogenic autocrine signaling<br />

through HER3 (Wilson et al.). This phenotype was particularly prominent in<br />

cell lines derived from SCCHN and was strongly correlated with high HRG<br />

expression. Interestingly, two patients with SCCHN tumors that expressed<br />

high levels <strong>of</strong> HRG in our phase Ia trial (abstract #95245) <strong>of</strong> MEHD7954A,<br />

a dual-action human IgG1 antibody that blocks ligand binding to EGFR and<br />

HER3 (Schaefer et al.) had partial responses. To further explore the<br />

hypothesis that high-level HRG expression defines a sub-population <strong>of</strong><br />

SCCHN that may be sensitive to agents targeting HER3, and to identify<br />

other potential target indications for the development <strong>of</strong> MEHD7954A, we<br />

evaluated the expression <strong>of</strong> HRG in large cohorts <strong>of</strong> multiple solid tumor<br />

indications. Methods: HER3 and HRG expression was analyzed by qRT-PCR<br />

in 648 formalin-fixed paraffin embedded primary tumor samples from<br />

patients with NSCLC, SCCHN, melanoma, CRC and triple-negative breast<br />

cancer. Results: SCCHN-derived tumor samples had the highest levels <strong>of</strong><br />

HRG expression, exhibiting a bimodal distribution in SCCHN – a pattern<br />

that is clearly distinct when compared to other tumor types. These data<br />

suggest that high HRG levels and potentially HER3-dependent autocrine<br />

signaling occur more frequently in SCCHN than in other tumors. Further we<br />

investigated whether overexpression <strong>of</strong> HRG in SCCHN correlated with<br />

stage and disease outcome. Updated results from these extended studies<br />

will be presented. Conclusions: SCCHN tumors exhibit bimodal expression<br />

<strong>of</strong> HRG, suggesting that HRG expression levels may be useful in identifying<br />

a subset <strong>of</strong> patients most likely to benefit from inhibition <strong>of</strong> HER3 activity.<br />

Antitumor activity in such patients has been observed in a phase I study <strong>of</strong><br />

MEHD7954A (abstract #95245).<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


5576 General Poster Session (Board #28C), Sat, 1:15 PM-5:15 PM<br />

Predictors for response to cetuximab in a prospective clinical trial (E2303)<br />

<strong>of</strong> patients with head and neck squamous cell carcinoma (HNSCC).<br />

Presenting Author: Amanda Psyrri, University <strong>of</strong> Athens, Athens, Greece<br />

Background: Theidentification <strong>of</strong> resistance mechanisms to Epidermal<br />

Growth Factor Receptor (EGFR) inhibitors remains critical lack in the<br />

management <strong>of</strong> HNSCC. We sought to determine predictors for response to<br />

cetuximab in a phase II clinical trial. Methods: 63 patients (pts) with<br />

operable stage III/IV HNSCC participated in E2303, an Eastern Cooperative<br />

Oncology Group (ECOG) phase II trial <strong>of</strong> induction chemotherapy with<br />

weekly cetuximab, paclitaxel and carboplatin x 6 followed by chemoradiotherapy<br />

with the same regimen. A tissue microarray was constructed and<br />

EGFR, ERK1/2, Met, pAkt and STAT protein expression levels were<br />

assessed using AQUA. The objectives <strong>of</strong> analysis were to determine<br />

association <strong>of</strong> biomarkers with E2303 efficacy outcomes (best objective<br />

response (OR), overall survival (OS), progression-free survival (PFS), and<br />

event-free survival (EFS)). The logistic regression model was used to<br />

examine relationship between marker measurements (on a continuous<br />

scale) and OR. The univariate and multivariate Cox proportional hazards<br />

models were used to evaluate the relationship between markers and<br />

event-time distributions. Fisher’s exact test was used to evaluate differences<br />

in response rate between groups (high vs. low AQUA scores).<br />

Event-time distributions were estimated by the Kaplan-Meier method and<br />

compared by the log-rank test. Results: Cytoplasmic ERK1/2 levels weresignificantly<br />

associated with PFS and OS (p�0.03 and 0.01, respectively).<br />

Nuclear ERK1/2 levels were significantly associated with OS (p�0.02) and<br />

tended towards significance for PFS (p�0.09). The multivariate Cox<br />

regression analysis shows that cytoplasmic and nuclear ERK1/2 are<br />

significantly associated with OS and PFS after controlling for primary site<br />

and disease stage, respectively There was no significant association<br />

between cytoplasmic or nuclear ERK1/2 status and OR (p-values 0.98 and<br />

0.41, respectively).No association was found between expression <strong>of</strong> any <strong>of</strong><br />

other biomarkers and outcome measures. Our data analysis was based on<br />

35 pts with marker data available. Conclusions: Ras/MAPK/ERKpathway<br />

may be associated with resistance to cetuximab in HNSCC.<br />

5578 General Poster Session (Board #29B), Sat, 1:15 PM-5:15 PM<br />

A combined evaluation strategy <strong>of</strong> FDG PET/CT and contrast-enhanced diagnostic<br />

CT (ceCT) for lymph nodes in the neck. Presenting Author: Neetha Gandikota,<br />

Department <strong>of</strong> Nuclear Medicine, Mount Sinai Medical Center, New York, NY<br />

Background: Accurate assessment <strong>of</strong> cervical lymph nodes (LNs) for determination<br />

<strong>of</strong> metastatic involvement is <strong>of</strong> prime importance for management. Our<br />

objective was to determine the incremental value <strong>of</strong> combining metabolic and<br />

morphologic information in the differentiation between benign and malignant<br />

LNs. Methods: A total <strong>of</strong>101 pts with head and neck squamous cell (n�91) or<br />

thyroid cancer (n�6) or lymphoma (n�4) were included in the study. PET/CT<br />

and neck ceCT were acquired simultaneously or sequentially at staging (n�48)<br />

or restaging (n�53). In 131 LNs, variables evaluated includes SUVmax, size,<br />

shape (elliptical vs non-elliptical), presence <strong>of</strong> extracapsular extension (Ecextirregular<br />

margins), necrosis, and fatty hilum. Histopathology (n�96) and 12 mo<br />

follow-up (n�35) were used for confirmation <strong>of</strong> the findings. ROC analyses<br />

determined the SUVmax cut-<strong>of</strong>f. Results: Of131 LNs, malignancy wasconfirmed<br />

in 49 (37%).Results are shown in table. In the detection <strong>of</strong> malignancy, SUVmax<br />

<strong>of</strong> 4.5 yielded the best balance between the sensitivity and specificity,<br />

performing better than all CT variables alone. Combination analysis improved<br />

results only when SUVmax (4.5) was added to Ecext. However, best combination<br />

results were obtained at a SUV cut-<strong>of</strong>f <strong>of</strong> 3.7. Higher SUV cut-<strong>of</strong>f did not<br />

significantly improve overall performance <strong>of</strong> SUVmax alone (Table). Conclusions:<br />

In the differentiation <strong>of</strong> malignant from benign LNs, SUVmax (4.5) yields better<br />

results than CT variables alone. However combining a lower SUVmax (3.7) with<br />

Ecext produced the best results. Increasing SUVmax cut-<strong>of</strong>f only produced a gain<br />

in specificity at a significant cost <strong>of</strong> sensitivity.<br />

Univariate variables Sen Spec Odds ratio 95% CI P value<br />

SUVmax 82% 79% 16 5 – 54.6 � 0.01<br />

�/- ecext 41% 93% 10 3.4 – 28.8 � 0.01<br />

�/- necrosis 44% 88% 5.9 2.4 – 14.5 � 0.01<br />

Shape 54% 83% 5.8 2.5 - 13 � 0.01<br />

�/- fatty hilum 90% 15% 0.6 0.2 - 2 0.4<br />

Size (1.2 cm) 70% 58% - - 0.11<br />

Combined analyses SUV max<br />

and ecext<br />

SUV max<br />

and necrosis<br />

SUV max<br />

and elliptical<br />

shape<br />

Sen Spec Sen Spec Sen Spec<br />

SUVmax cut 3.7 95% 80% 100% 22% 100% 23%<br />

SUVmax cut 4.5 86% 80% 91% 44% 89% 23%<br />

SUVmax cut 5.3 76% 100% 82% 78% 73% 62%<br />

P value 0.005 0.007 0.03<br />

Head and Neck Cancer<br />

375s<br />

5577 General Poster Session (Board #29A), Sat, 1:15 PM-5:15 PM<br />

The impact <strong>of</strong> advanced age on outcomes in squamous cell carcinoma <strong>of</strong><br />

the head and neck. Presenting Author: Virginia Ann Moye, University <strong>of</strong><br />

North Carolina School <strong>of</strong> Medicine, Chapel Hill, NC<br />

Background: Limited data are available regarding outcomes in elderly head<br />

and neck cancer patients. This retrospective study was designed to<br />

characterize head and neck cancer in geriatric patients in the UNC Tumor<br />

Registry. Methods: Of 1,598 patients identified from the registry, 1,291<br />

patients were �70 years old (young group, YG) and 307 patients were � 70<br />

(elderly group, EG) at diagnosis. Both overall survival (OS), calculated from<br />

diagnosis to death or the last follow up, and relapse free survival (RFS) were<br />

censored at 60 months. Log-rank test was used to compare survival<br />

difference. Cox proportional hazard model was used to estimate hazard<br />

ratio, adjusting for potential confounding factors. All tests were performed<br />

in R 2.13.1. Results: EG patients were more likely to present at an early<br />

stage with 25% presenting at stage I and 37% presenting at stage IV<br />

compared to 17% and 51% respectively in YG. EG had a median OS <strong>of</strong> 35<br />

months (95% CI: 28-41 months) compared to approximately 60 months in<br />

YG (p � 0.0001 log rank test). The median RFS for EG and YG are 25 (95%<br />

CI: 20-32 months) and 44 (95% CI: 38-52 months) months respectively (p<br />

� 0.0001 log rank test). When controlling for possible confounders, EG<br />

patients were nearly twice as likely to die (HR 1.94, 95% CI 1.635-2.302,<br />

p�0.0001) and 70% more likely to relapse in 5 years compared to YG (HR<br />

1.704, 95% CI 1.449-2.004, p�0.05). The median life expectancy for<br />

elderly patients in our study was nearly 5 years for stage I/II and �2 years<br />

for stage III/IV. Conclusions: Poor OS and RFS in elderly patients compared<br />

to younger patients in this study indicate a need for further investigation<br />

considering comorbidities and aggressiveness <strong>of</strong> treatment. Significant<br />

differences in life expectancy in elderly patients with early versus late<br />

disease may help guide patients and clinicians in determining how<br />

aggressively to treat.<br />

5579 General Poster Session (Board #29C), Sat, 1:15 PM-5:15 PM<br />

A phase I/II study <strong>of</strong> concurrent nab-paclitaxel, carboplatin, and IMRT for<br />

locally advanced squamous cancer <strong>of</strong> the head and neck (LASCCHN).<br />

Presenting Author: Roy B. Tishler, Department <strong>of</strong> Radiation Oncology,<br />

Dana-Farber Cancer Institute/Brigham and Women’s Hospital, Boston, MA<br />

Background: We studied escalating doses <strong>of</strong> weekly nab-paclitaxel (Abraxane,<br />

AB) combined with weekly carboplatin (CP) and daily IMRT in patients<br />

with LASCCHN to determine the safety and optimal dosing <strong>of</strong> AB. The<br />

favorable biodistribution <strong>of</strong> AB, an albumin bound formulation <strong>of</strong> paclitaxel,<br />

may allow for intensified locoregional treatment without increasing<br />

normal tissue toxicity. Methods: An AB dose escalation was performed,<br />

initially using concurrent weekly CP (AUC � 1.5), Erbitux (ER) and daily<br />

IMRT to 70 Gy. After ER was dropped, dose escalation <strong>of</strong> AB from 20<br />

mg/m2 to 50 mg/m2 was performed in 10 mg/m2 increments using a 3 � 3<br />

study design. Results: A total <strong>of</strong> 28 patients have enrolled and all have<br />

completed treatment. Patients ranged in age from 42 to 72 years old<br />

(median � 59), with follow up from 3 to 48 months (median � 25). Most<br />

patient had oropharyngeal primaries (n�22) and stage III (n�8) or IV<br />

(n�20) disease; 18 were HPV�, 6 HPV- and 4 unknown. Initially 6<br />

patients received weekly AB (20 mg/m2 ), ER and CP, but ER was dropped<br />

due to greater than anticipated mucositis and dysphagia. Subsequently, 3<br />

patients each received CP with AB at 20, 30 and 40 mg/m2 weekly and a<br />

total <strong>of</strong> 13 received CP with AB at 50 mg/m2 . The median radiation dose is<br />

70 Gy and the median number <strong>of</strong> weekly chemotherapy cycles is 7. The<br />

primary toxicities were Grade 3 dysphagia, mucositis and dermatitis in 24<br />

<strong>of</strong> 28 patients. Only 3 (<strong>of</strong> 28) PEG tubes remain at 4, 6 and 14 months. No<br />

DLTs were observed at any dose level. There have been 5 recurrences, with<br />

4 being local-regional. Two patients have died <strong>of</strong> disease and 26 are alive.<br />

Conclusions: Concurrent AB, CP, and IMRT is a safe chemoradiotherapy<br />

combination in LASCCHN with early data demonstrating feasibility and<br />

efficacy.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


376s Head and Neck Cancer<br />

5580 General Poster Session (Board #30A), Sat, 1:15 PM-5:15 PM<br />

P16 and cyclin D1 (CYD1) as prognostic markers in hypopharyngeal (HSC)<br />

and oropharyngeal squamous cell carcinoma (OSC). Presenting Author:<br />

Siok Hoon Ang, Duke-NUS Graduate Medical School Singapore, Singapore,<br />

Singapore<br />

Background: In head and neck cancer (HNC), dysregulation <strong>of</strong> cell cycle<br />

proteins p16 and CYD1 are common. Variable associations <strong>of</strong> p16 over- and<br />

under- expression and CYD1 overexpression with overall survival (OS) have<br />

been described in different HNC sites. We evaluated the relationship <strong>of</strong><br />

p16 and CYD1 expression with clinical characteristics and OS in OSC and<br />

HSC. Methods: p16 and CYD1 expression was evaluated by immunohistochemistry<br />

in 77 HSC and 103 OSC patients (pts) and recorded as p16N<br />

(0% tumor cells stained), p16L (5-69%) and p16H (�70%), CYD1-<br />

(�10%) and CYD1� (�10%). OS between groups was evaluated by<br />

Kaplan-Meier method and compared by log rank test. Hazard ratio (HR) for<br />

death was estimated using multivariable Cox models. Results: Pts were<br />

predominantly Chinese (83.6% v 85.4%) with locally advanced HNC<br />

(91.4% v 92.2%). Compared to OSC, HSC pts were older (median age 67 v<br />

61 yrs), more likely male (89.3% v 74.0%), current or ex-smokers (83.3%<br />

v 63.6%) with higher comorbidity-age combined risk score (ageCCI), less<br />

likely p16H (6.5% v 30.1%)(all p�0.001) and had similar CYD�. p16H<br />

pts were younger (median age 58 (p16H) v 65 (p16L) v 66 (p16N) yrs,<br />

p�0.002), more likely non-smoker (51.4% v 23.4% v 13% p�0.001) with<br />

lowest ageCCI (p�0.001). <strong>Clinical</strong> characteristics did not differ by CYD1<br />

status. At median f/u <strong>of</strong> 50mths, median OS was 33 mths. Median OS was<br />

poor in HSC compared to OSC (23.9 v 72.1, p�0.001). Multivariate<br />

analysis showed associations <strong>of</strong> N2/N3 disease (HR 1.57, p�0.036),<br />

ageCCI (HR 1.22 per 1 pt increase, p�0.001), p16 (p16H: ref; p16L: HR<br />

2.34, p�0.045; p16N: HR 2.74, p�0.013) and CYD1� (HR 1.94,<br />

p�0.015) with death, independent <strong>of</strong> gender, smoking and site. Association<br />

<strong>of</strong> p16 with OS was seen mainly in OSC (median OS p16H: not reached<br />

(NR), p16L: 62, p16N: 22 mths, p�0.001) compared with median OS<br />

(HSC) (27 v 28 v 21, p�0.609). Similarly the association <strong>of</strong> CYD1 with OS<br />

was mainly in OSC (median OS CYD1-: NR v 23 mths, p�0.001 v HSC: 25<br />

v 25 mths, p�0.19). Conclusions: In OSC, p16 expression correlates with<br />

OS, with p16N associated with worst OS. CYD1 has an independent<br />

association with OS. Poorer OS in HSC may be due to adverse clinical<br />

characteristics. Assessment <strong>of</strong> p16 and CYD1 status in HSC did not predict<br />

for OS.<br />

5582 General Poster Session (Board #30C), Sat, 1:15 PM-5:15 PM<br />

HPV and survival in patients with oropharyngeal squamous cell cancer <strong>of</strong><br />

the head and neck (OPC) treated with induction chemotherapy followed by<br />

chemoradiotherapy (ST) versus chemoradiotherapy alone (CRT): The Dana-<br />

Farber experience. Presenting Author: Jochen H. Lorch, Department <strong>of</strong><br />

Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School,<br />

Boston, MA<br />

Background: HPV status is a major prognostic marker for survival in patients<br />

with OPC. We examined overall survival (OS) and progression free survival<br />

(PFS) in patients with OPC and known HPV status treated at Dana-Farber<br />

Cancer Institute with ST and CRT between 2002 and 2011. Methods: 280<br />

patients with OPC and known HPV status were identified retrospectively<br />

and clinical information was recorded. Results: 174 patients were treated<br />

with CRT (124 HPV positive, 50 HPV negative) and 106 patients were<br />

treated with ST (77 HPV positive, 29 HPV negative). For all 280 patients,<br />

OS and PFS were significantly better for patients who were HPV positive<br />

compared to those who were HPV negative. 3 year OS was 89.1% for HPV<br />

positive (95% CI, 83.8-94.7) and 70.5% for HPV negative patients<br />

(95%CI, 59.9-83%, HR 0.37, p�0.0007). Among HPV positive patients<br />

treated with CRT, 13/124 had died at 3 years (OS 88.5%, 95%CI<br />

81.7-95.9) while 13 deaths were recorded among 50 HPV negative<br />

patients (OS 72.2%, 95% CI 59.1-88.2, HR 0.38, p�0.011). PFS at 3<br />

years was also significantly better for HPV positive versus HPV negative<br />

patients(81.7% vs 58.8%, HR 0.42, p�0.006). In the group treated with<br />

ST, outcomes were similar despite a higher rate <strong>of</strong> stage IV versus stage III<br />

disease compared to the group treated with CRT (100% stage IV in ST<br />

versus 77% stage IV and 23% stage III disease in CRT group). Three year<br />

OS was 89.7% for HPV positive and 68.2% in the HPV negative group<br />

(8/77 HPV pos vs 10/29 HPV neg, HR 0.33, p�0.015). PFS was borderline<br />

better for HPV positive patients (81% vs 61.7%, HR 0.48, p� 0.058).<br />

Conclusions: We present the DFCI experience treating OPC with ST and CRT<br />

for patients with known HPV status over one decade.OS and PFS were<br />

superior for HPV positive versus HPV negative patients. Outcomes were<br />

virtually identical for patients treated with CRT versus ST despite a higher<br />

rate <strong>of</strong> stage IV disease in the ST group. Outcomes for the HPV negative<br />

patients in particular were superior compared to the published literature.<br />

5581 General Poster Session (Board #30B), Sat, 1:15 PM-5:15 PM<br />

The role <strong>of</strong> postoperative radiotherapy (PORT) in the management <strong>of</strong><br />

parotid gland malignancy (PGM). Presenting Author: Louis Harrison, Beth<br />

Israel Medical Center, New York, NY<br />

Background: To report the role <strong>of</strong> PORT in the management <strong>of</strong> PGM and to<br />

analyze the prognostic factors for optimal locoregional (LRC) and distant<br />

control (DC) Methods: This is a single institution retrospective study. From<br />

March 2001-2011, a total <strong>of</strong> 118 patients with PGM were treated with<br />

surgery and PORT (100%) <strong>of</strong> which 75% were treated with IMRT, 20 and<br />

10% received concurrent carboplatin and CDDP respectively. Indications<br />

for PORT were: recurrent or gross residual disease, �ve or close margins,<br />

nerve invasion and �ve lymph nodes (LN). PORT fields comprehended<br />

parotid bed and ipsilateral neck. Median PORT doses delivered were 70,<br />

63, 54 Gy (at 1.8-2 Gy/fraction) for gross, microscopic disease, and<br />

ipsilateral neck respectively. All PORT was delivered via 4-6 MV photons<br />

with daily 3-5 mm bolus. The median age was 52 (18-89). Females and<br />

Caucasian made up 56% and 64% <strong>of</strong> the population, respectively.<br />

Histologic findings were adenoid cystic carcinoma 20%, mucoepidermoid<br />

carcinoma 19%, adenocarcinoma 18%, pleomorphic carcinoma 16%,<br />

salivary duct carcinoma 9%, acinic cell carcinoma 8%, ex-pleomorphic<br />

carcinoma 7%, and sarcoma 3%. Pathological stages I, II, III, IV were 10,<br />

30, 20 and 40% respectively Results: With a median follow up <strong>of</strong> 56<br />

months (12-124), the LRC and DC for the whole cohort were 96% (5/118)<br />

and 93% (8/118) respectively. The median duration <strong>of</strong> PORT was 45 days<br />

(40-60). The median time to LR failure (LRF) and distant metastases were<br />

16 (12 - 30) and 28 months (24-48) respectively. Late RT toxicity was;<br />

grade I xerostomia 30%, altered taste 15%, trismus 6%, dysphagia and<br />

neck stiffness 2%. Kaplan-Meier curve shows the 5-year cause-specific<br />

survival to be 100%. Chi square and regression analysis tests show<br />

significant relationship (P�0.001) between salivary duct ca (70%) and<br />

distant metastases (DM), also males are at higher risk (17%) to develop<br />

LRF and DM, compared to females (6%). Poor prognostic features for DM<br />

and LRF were salivary duct ca, stage IV, PNI, and multiple LN involvement.<br />

Conclusions: Our data show that surgery followed by PORT to the tumor bed<br />

and ipsilateral neck provide excellent long standing oncologic and functional<br />

outcomes. Further studies are warranted to optimize the management<br />

<strong>of</strong> patients at higher risk <strong>of</strong> DM despite LRC and RT � chemotherapy<br />

administered.<br />

5583 General Poster Session (Board #31A), Sat, 1:15 PM-5:15 PM<br />

Phase II study <strong>of</strong> low-dose paclitaxel and cisplatin in combination with<br />

split-course concomitant twice-daily reirradiation (XRT) in recurrent squamous<br />

cell carcinoma <strong>of</strong> the head and neck (SCCHN): Long-term follow-up<br />

<strong>of</strong> Radiation Therapy Oncology Group (RTOG) protocol 9911. Presenting<br />

Author: Corey J. Langer, Abramson Cancer Center at the University <strong>of</strong><br />

Pennsylvania, Philadelphia, PA<br />

Background: Loco-regionally recurrent SCCHN or new second primary tumor<br />

(SPT) in a previous radiation field, if not curable by surgery, is virtually<br />

always fatal. Chemotherapy alone yields a median survival time (MST) <strong>of</strong> no<br />

more than 10 to 11 months, 1- and 2- year overall survival (OS) rates <strong>of</strong><br />

35% and 10-15% at best. Concurrent re-irradiation and chemotherapy is<br />

an alternative strategy. Methods: Eligibility for RTOG protocol 9911 stipulated<br />

measurable, recurrent SCCHN or SPT in a previous radiation field, PS<br />

0-1, and adequate end-organ indices. Patients (pts) received twice-daily<br />

XRT (1.5 Gy per fraction BID x5devery 2 weeks x4), plus cisplatin 15<br />

mg/m2 IV QD x 5 and paclitaxel 20 mg/m2 IV QD x 5 every 2 weeks x 4.<br />

G-CSF was administered days 6 - 13 <strong>of</strong> each 2-week cycle. Primary<br />

endpoints were OS and progression-free survival PFS at 1 and 2 yrs.<br />

Secondary endpoints included acute/late toxicities and patterns <strong>of</strong> failure.<br />

Results: 105 patients were enrolled from March 2000 through June 2003.<br />

100 patients were analyzable (76% male, med age 60 yrs). Oropharynx<br />

(41%) and oral cavity (27%) were the predominant primary sites. 23% had<br />

SPT. Median prior XRT dose was 65.7 Gy. 73% <strong>of</strong> pts completed all<br />

chemotherapy. Grade � 4 acute toxicity occurred in 28%, grade � 4 acute<br />

hematologic toxicity in 21%. 9 treatment-related deaths (9%) occurred:<br />

five in the acute setting, four late (including three carotid hemorrhages at<br />

116, 427 and 2772 days). 1-, 2-, and 5-year OS rates were 50%, 27%,<br />

and 15% respectively. PFS at 1, 2, and 5 yrs were 35%, 18%, and 7%.<br />

64.9% died from the incident cancer, 3.2% from SPT, and 21.2% from<br />

other, non-cancer causes. Estimated 10 yr survival is 5% with 6 pts still<br />

alive, including 2 free from relapse. Conclusions: Despite a high incidence<br />

<strong>of</strong> grade 5 toxicity, 1-, 2-, and 5- yr OS rates for split-course bid XRT and<br />

concurrent cisplatin/paclitaxel exceed results usually seen with chemotherapy<br />

alone. At 5-10 yrs, we have observed longterm survivors free <strong>of</strong><br />

recurrence.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


5584 General Poster Session (Board #31B), Sat, 1:15 PM-5:15 PM<br />

Sentinel lymph node biopsy for clinically N0 oral squamous cell carcinoma.<br />

Presenting Author: Hiroyuki Goda, Ehime University Graduate <strong>of</strong> Medicine,<br />

Department <strong>of</strong> Oral and Maxill<strong>of</strong>acial Surgery, Ehime, Toon, Japan<br />

Background: Regionallymph node metastasis is an important prognostic<br />

factor in oral squamous cell carcinoma (OSCC). Sentinel lymph node<br />

biopsy (SLNB) is a widely accepted procedure in various human malignancies.<br />

In clinically N0 (cN0) OSCC cases, SLNB has received considerable<br />

attention for its role in deciding whether to perform neck dissection. In this<br />

study, we assessed the efficiency <strong>of</strong> SLNB for cN0 OSCC case in a<br />

single-institution experience. Methods: 100 patients with cN0 OSCC<br />

underwent SLNB between 2001 and 2011, <strong>of</strong> which 95 were clinically T1<br />

and T2. The primary site was tongue, gingiva, oral floor, buccal mucosa,<br />

and lip in 50%, 36%, 8%, 5%, and 1%, respectively. The location <strong>of</strong><br />

sentinel lymph node (SLN) was determined by radioisotope (RI) method<br />

with preoperative lymphoscintigraphy and intraoperative use <strong>of</strong> a handheld<br />

gamma probe and/or dye method, and evaluated by histopathological<br />

examination and genetic analysis. Results: SLNB was performed with RI<br />

and dye method (79%), only dye method (14%), or only RI method (7%).<br />

SLN was successfully identified with RI method (100%) and dye method<br />

(71%). The average number <strong>of</strong> SLN was 2.5 with dye method and 1.9 with<br />

RI method. The rate <strong>of</strong> SLN identified side was 84% in ipsilateral, 10% in<br />

bilateral, and 6% in contralateral. Fifteen <strong>of</strong> 100 patients (15%) had<br />

metastasis-positive SLN, and 3 patients was up-grade to stage III and<br />

others to stage IVA. Eight patients with negative SLN developed latent neck<br />

lymph node metastasis. The sensitivity, specificity, accuracy, and negative<br />

predictive value was 65% (15/23), 100% (77/77), 92% (92/100) and<br />

91% (77/85). Disease specific survival rate for SLNB-negative patients<br />

were 98% (79/81), and for SLNB-positive patients were 73% (11/15),<br />

respectively. Conclusions: SLNB is a minimally invasive and highly reliable<br />

means <strong>of</strong> staging the cN0 neck for patients with OSCC. Patients with<br />

negative SLNB showed more excellent neck control rate and SLNB provides<br />

more accurate staging than elective neck dissection or wait and see.<br />

5586 General Poster Session (Board #31D), Sat, 1:15 PM-5:15 PM<br />

Importance <strong>of</strong> HPV involvement and FOXP3� T-cell status as prognostic<br />

factors in tonsilar squamous cell carcinoma. Presenting Author: Kwonoh<br />

Park, Department <strong>of</strong> Oncology, Asan Medical Center, University <strong>of</strong> Ulsan<br />

College <strong>of</strong> Medicine, Seoul, South Korea<br />

Background: Human papillomavirus (HPV) status is a strong and independent<br />

favorable prognostic factor for survival in tonsilar squamous cell<br />

cancer (TSCC). The reason for the improved survival in HPV associated<br />

TSCC is unclear. Recently, activation <strong>of</strong> immune surveillance mechanism<br />

against non-self Ag is postulated to one <strong>of</strong> reasonable causes as favorable<br />

prognosis <strong>of</strong> HPV associated with TSCC. Methods: We reviewed the medical<br />

records <strong>of</strong> histologically confirmed locally advanced TSCC patients, curatively<br />

treated in Asan Medical Center from January 2000 to December<br />

2008. The immunohistochemistry (IHC) assays for p16 and FOXP3 were<br />

done in TSCC pafaffin-embedded samples. Results: We identified 79<br />

patients who met the inclusion criteria. The median age was 54 years<br />

(range 32-76) and 16 (20%) patients were stage III and the others were all<br />

stage IV. With the median follow up <strong>of</strong> 62.9 months (95% CI, 59.2 - 66.7),<br />

sixty three (80%) were HPV-positive with p16 overexpression, and 38<br />

(48%) were Treg-positive with FOXP3. Treg involvement was significantly<br />

related to HPV positive status (P�0.011). The result was the same after<br />

adjustment <strong>of</strong> age, T&Nstage, smoking exposure and alcohol consumption.<br />

(Odd ratio � 6.54, 95% CI 1.58-27.1, P�0.01) Five-year overall<br />

survival (OS) rate in HPV-positive group was significantly higher than that <strong>of</strong><br />

HPV-negative group (78% and 63%, Hazard Ratio (HR)�0.347, 95% CI<br />

0.14-0.87, P�0.025), and 5-year OS <strong>of</strong> Treg-positive group was also<br />

higher than that <strong>of</strong> Treg-negative group (89% and 61%, HR�0.158, 95%<br />

CI 0.05-0.53, P�0.003). In multivariate analysis, the Treg status was an<br />

independent prognostic factor (HR�0.11, 95% CI 0.03-0.40, P�0.001),<br />

as well as HPV status. (HR�0.28, 95% CI 0.10 - 0.78, P�0.016).<br />

Conclusions: HPV positivity was associated with Treg positivity in TSCC and<br />

both were found to be favourable prognostic factors for survival.<br />

Head and Neck Cancer<br />

377s<br />

5585 General Poster Session (Board #31C), Sat, 1:15 PM-5:15 PM<br />

Neoadjuvant chemotherapy followed by definitive local treatment in locally<br />

advanced carcinoma maxillary sinus. Presenting Author: Vijay Patil, TMH,<br />

Mumbai, India<br />

Background: Locally advanced carcinoma <strong>of</strong> maxillary sinus has been<br />

historically reported to have poor prognosis. We evaluated the role <strong>of</strong><br />

neoadjuvant chemotherapy in improving the outcome in these patients.<br />

Methods: 41 patients with locally advanced borderline resectable (stage<br />

IVa) or unresectable maxillary carcinoma (stage IVb) were treated with<br />

induction chemotherapy between 2008 and 2011. The protocol included 2<br />

cycles <strong>of</strong> chemotherapy, response assessment and multidisciplinary clinic<br />

review for definitive local treatment. The demographic pr<strong>of</strong>ile, response to<br />

induction therapy, toxicity <strong>of</strong> chemotherapy, definitive treatment received,<br />

time to treatment failure (TTF) and overall survival (OS) were analysed.<br />

Univariate and multivariate analysis was performed to determine factors<br />

associated with response, TTF and OS. Results: The cohort <strong>of</strong> 41 patients<br />

had a median age <strong>of</strong> 48 years (22-71) with male preponderance (80.5%).<br />

The chemotherapy included two drugs (platinum and taxane) in 34 patients<br />

(82.9%) and three drugs (platinum, taxane and 5 FU) in 7(17.1%) The<br />

taxane utilized was docetaxel in 22 patients (53.7%) and paclitaxel in 19<br />

patients (46.3%). There was no complete response, stable disease in 18<br />

(43.9%), partial response in 16 (39%), and progression in 7 (17.1%)<br />

patients. All patients competed two cycles <strong>of</strong> chemotherapy, adequate dose<br />

intensity was maintained in 33 patients (78%) and there were no deaths.<br />

Post-induction, the treatment planned included surgery in 12 (29.3%),<br />

CT-RT in 24 (58.5%), radical RT in 1 (2.4%), palliative RT in 1 (2.4%) and<br />

palliative chemotherapy in 3 ( 7.3%) patients. Overall, the median TTF was<br />

10.2 months. With 16 deaths, the median OS was not reached. Only<br />

response post-induction was significantly associated with better TTF<br />

(p�0.02). Grade 3 tumor (p�0.04) and baseline serum albumin more than<br />

4 mg/dl (p�0.02) were associated with better OS. Grade 3-4 neutropenia,<br />

febrile neutropenia and loose motions were seen in 21.1%, 23.8% and<br />

15.8% respectively. Conclusions: In unresectable maxillary carcinoma,<br />

induction chemotherapy has clinically significant benefit with acceptable<br />

toxicity. Response to induction was the only significant factor for improved<br />

TTF.<br />

5587 General Poster Session (Board #31E), Sat, 1:15 PM-5:15 PM<br />

Pretreatment neurocognitive function (NCF) status in head and neck<br />

cancer (HNC) patients (pts) with comparison to control cohort. Presenting<br />

Author: Albiruni Ryan Abdul Razak, Princess Margaret Hospital, Toronto,<br />

ON, Canada<br />

Background: There is increasing evidence that NCF abnormalities may<br />

occur in cancer pts. Data on pre-treatment NCF in HNC pts are lacking.<br />

This study reports NCF in pts with newly diagnosed, curable HNC compared<br />

to controls. Methods: HNC pts underwent a 2-hour battery <strong>of</strong> NCF tests prior<br />

to radio �/-chemo(bio)therapy. Domains tested were intelligence (IQ),<br />

memory, language, attention, processing speed, executive function and<br />

manual dexterity. Test performances were transformed into Z-scores using<br />

normative data (score � -1 signified deficit). Pts also had self-reported<br />

assessments for NCF, quality <strong>of</strong> life (QOL), fatigue and affect. Data<br />

obtained were compared to non-cancer controls who underwent the same<br />

tests. Results: Eighty HNC and 30 control subjects were assessed. Objective<br />

NCF testing demonstrated that HNC and control cohorts were similar<br />

across all domains, except for IQ, with pts having higher scores (mean 0.55<br />

vs 0.12, p�0.03). However, individual analysis showed that 39% <strong>of</strong> HNC<br />

and 43% <strong>of</strong> control subjects had abnormal Z-scores in � 2 domains.<br />

Multivariable analysis <strong>of</strong> factors associated with � 2 abnormal NCF<br />

domains included: low education level, significant smoking history (� 10<br />

pack year), previous mild brain injury, gender, and group (pt vs control).<br />

Amongst pts, HPV -ve status and non-oropharyngeal tumors were also<br />

associated with decreased NCF. Pts reported statistically worse subjective<br />

baseline symptoms compared to controls: NCF (mean FACT-COG 33.7 vs<br />

18.2, p�0.002), QOL (FACT H&N 33.8 vs 14.9), fatigue (FACT-F 35.1 vs<br />

15.1), anxiety (HADS 7.0 vs 3.1) and depression (HADS 3.9 vs 1.2),<br />

p�0.01 for all five parameters. Conclusions: Objectively assessed NCF was<br />

similar between HNC pts and controls, but a proportion <strong>of</strong> participants in<br />

both cohorts have multi-domain abnormal Z-scores. Several patient demographics<br />

and disease characteristics were associated with abnormal NCFs.<br />

Subjectively, pts reported worse NCF, QOL, fatigue and affect. These data<br />

suggest that participant and disease characteristics may play a larger role<br />

in determining NCF than previously shown. Whether such characteristics<br />

impact subsequent NCF is under investigation in a longitudinal study.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


378s Head and Neck Cancer<br />

5588 General Poster Session (Board #31F), Sat, 1:15 PM-5:15 PM<br />

Second primary tumors in head and neck squamous cell carcinoma<br />

patients: A cross-sectional study. Presenting Author: Luciana Garcia<br />

Landeiro, <strong>Clinical</strong> Oncology, Faculdade de Medicina do ABC, Santo Andre,<br />

Brazil<br />

Background: Head and neck squamous cell carcinoma (HNSCC) patients<br />

(pts) are at elevated risk <strong>of</strong> developing a second primary tumor (SPT). Here<br />

we aimed to study the frequency <strong>of</strong> SPTs and those characteristics related<br />

to an increased probability <strong>of</strong> SPT occurrence. Methods: It is a crosssectional<br />

study in pts diagnosed with SCC primary located in oral cavity,<br />

oropharynx, hypopharynx or larynx, treated in two Brazilian institutions,<br />

with an index tumor diagnosed from Jan/2000 to Jan/2009. All pts were<br />

followed according the current recommendations, including clinical visits,<br />

imaging and endoscopic procedures. A SPT was defined following the<br />

Warren and Gates criteria (Am J Cancer 51:1358,1932), and they were<br />

classified as synchronous or metachronous if detected less or more than six<br />

months after the diagnosis <strong>of</strong> the index tumor, respectively. Results: 38 out<br />

<strong>of</strong> 318 HNSCC pts (12%) were diagnosed with 41 SPTs in their follow-up,<br />

being 12 synchronous (29%) and 29 metachronous (71%), all classified as<br />

SCC. Regarding the index tumor, it was primary located in oral cavity (114<br />

pts, 36%), oropharynx (87 pts, 27%), larynx (83 pts, 26%) and hypopharynx<br />

(34 pts, 11%), and staged as I-II (30%) or III-IV (67%). The SPTs were<br />

located more frequently in oral cavity (27%), lungs (27%), oropharynx<br />

(15%) and esophagus (12%) and staged as I-II (44%) or III-IV (42%).<br />

Among the 29 metachronous SPTs, 12 (41%) were diagnosed in the first<br />

two years <strong>of</strong> follow-up, 11 (38%) between two and five years, and 6 (21%)<br />

after five years. Considering all the studied HNSCC pts and index tumor<br />

characteristics, including primary site, persistence <strong>of</strong> tabacco and alcohol<br />

habits, and treatment, none was identified as related to an increased<br />

probability <strong>of</strong> developing a SPT. In the median follow-up <strong>of</strong> 40 mo., the<br />

median overall survival was not reached in all 318 HNSCC pts. No<br />

difference in overall survival was observed between those pts diagnosed or<br />

not with a SPT (HR 0.35, p�0.555). Conclusions: We found a 12%<br />

incidence <strong>of</strong> SPTs in these HNSCC pts. At diagnosis, SPTs were located<br />

more frequently in oral cavity and lungs, in advanced stages, and may occur<br />

even after five years after the index tumor. More efficient primary and<br />

secondary preventive strategies <strong>of</strong> SPTs must be developed.<br />

5590 General Poster Session (Board #31H), Sat, 1:15 PM-5:15 PM<br />

Gene expression as a predictor <strong>of</strong> radiotherapy or chemoradiotherapy<br />

response in head and neck squamous cell carcinoma patients: A molecular<br />

approach. Presenting Author: Antonio Lopez-Pousa, Hospital De Sant Pau,<br />

Barcelona, Spain<br />

Background: There are not validated markers <strong>of</strong> radioresistance in patients<br />

with a head and neck squamous cell carcinoma (HNSCC). The objective <strong>of</strong><br />

our study was to explore the relationship between response to radiotherapy<br />

and the expression at transcriptional level <strong>of</strong> a panel <strong>of</strong> genes related with<br />

the HNSCC carcinogenic process. Methods: The study group was 76<br />

consecutive HNSCC patients treated with radiotherapy (n�51) or chemoradiotherapy<br />

(n�25) with curative intention, and a minimum follow-up <strong>of</strong><br />

three years. Thirty-seven patients were T1-T2 tumors and 16 had an<br />

advanced tumor (T3-T4). Biopsy specimens were performed from the<br />

primary tumor before treatment and conveniently stored until processing.<br />

Expression <strong>of</strong> genes related with cell cycle (cyclin D1), apoptosis (Bcl-XL),<br />

angiogenesis (VEGF), inflammation (IL-8), and tissue invasiveness (MMP-2<br />

and MMP-9) were examined using a RT-PCR method. The continuous value<br />

<strong>of</strong> the mRNA expression level was evaluated with a classification and<br />

regression tree (CRT) method, considering the local control <strong>of</strong> the disease<br />

achieved with radiotherapy as the dependent variable. Results: During the<br />

follow-up 26 patients (33.8%) had a local failure. The CRT method found a<br />

significant relationship between VEGF and MMP-9 expression and local<br />

control <strong>of</strong> the disease. According the expression level <strong>of</strong> VEGF and MMP-9<br />

the patients were classified in three groups: the A group (n�19) defined by<br />

a low expression <strong>of</strong> VEGF, the B group (n�19) defined by a high expression<br />

<strong>of</strong> VEGF and a low expression <strong>of</strong> MMP-9 and the C group (n�38) defined by<br />

a high expression <strong>of</strong> both VEGF and MMP-9. The 5-year local recurrence<br />

free survival was: A group 94.7% (CI 95%: 84.7-100%), B group 83.3%<br />

(CI 95%: 66.1-100%), and C group 37.6% (CI 95%: 21.0-54.2%). There<br />

were significant differences in the local control <strong>of</strong> the disease according to<br />

the VEGF and MMP-9 expression values (test log rank, P�0.0001).<br />

Conclusions: Expression <strong>of</strong> VEGF and MMP-9 genes may be a radiosensitivity<br />

marker for patients with HNSCC treated with radical intention. We didn’t<br />

find prognostic significance for other genes explored.<br />

5589 General Poster Session (Board #31G), Sat, 1:15 PM-5:15 PM<br />

Pattern <strong>of</strong> symptoms in head and neck cancer (HNC) survivors treated with<br />

radiation therapy (RT): Association with systemic therapy. Presenting<br />

Author: David Ira Rosenthal, University <strong>of</strong> Texas M. D. Anderson Cancer<br />

Center, Houston, TX<br />

Background: We sought, in a large, long-term follow-up cohort, to determine<br />

benchmark cross-sectional analysis <strong>of</strong> HNC patient (pt) symptom pr<strong>of</strong>iles<br />

as a function <strong>of</strong> chemotherapy (CT)/RT strategy. Methods: Pts treated with<br />

RT with or without CT in remission � 18 mos. were surveyed with the MD<br />

Anderson Symptom Inventory–Head and Neck Module (MDASI-HN). <strong>Clinical</strong><br />

data were extracted. Data were tabulated, and group comparison<br />

performed using non-parametric analyses. Results: 250 pts participated;<br />

81% were male. Median age at RT was 54 years. 87% had oropharynx<br />

HNC. Most were T1/X (41%), 37% T2 and, 22% T3/4. For N-category,<br />

most were N2 (55%), �5% N3, and 40% NX-1. 18% had induction CT,<br />

26% concurrent, 5% both, and 50% had none. At a median follow-up time<br />

<strong>of</strong> 5.9 years (range 2-15), 11% <strong>of</strong> pts were entirely symptom free, 31%<br />

reported �mild symptom severity, 20% �moderate, and 38% reported �1<br />

symptom as severe. 16% <strong>of</strong> pts receiving RT alone and 11% sequential CT<br />

followed by RT were symptom free, vs. 1% <strong>of</strong> those receiving concurrent CT<br />

(chi-square p�0.01), and the symptom distribution pr<strong>of</strong>ile was distinct<br />

(p�0.03). The proportion <strong>of</strong> pts who received concurrent CT reporting any<br />

severe symptom item was 44%, vs. 36% <strong>of</strong> those not receiving concurrent<br />

CT (p�n.s.). No difference was seen in the moderate to severe (M/S)<br />

symptom report by treatment group. For all MDASI-HN items, most pts<br />

rated “0” or “not present”, except dry mouth and difficulty swallowing<br />

items, where mild symptoms were most likely (36% and 33%). The most<br />

common symptoms rated M/S were dry mouth, swallowing, choking,<br />

fatigue, and mouth and throat mucus reported by 42%, 23%, 18%, 16%,<br />

and 16%. MVA demonstrated T-stage and primary site, but not CT cohort<br />

correlated with M/S symptom report. Conclusions: Cumulatively, most pts<br />

had no more that mild symptom severity, but a substantial group <strong>of</strong> pts<br />

experience M/S levels. The symptom severity pr<strong>of</strong>ile was highest with<br />

concurrent CT, though this effect appears mediated by disease specific<br />

factors. The addition <strong>of</strong> sequential CT to RT did not to appear to alter M/S<br />

symptom report substantially; however, concurrent pts were almost never<br />

symptom free, in contrast to induction and no CT cohorts.<br />

5591 General Poster Session (Board #32A), Sat, 1:15 PM-5:15 PM<br />

A phase II trial <strong>of</strong> the multitargeted kinase inhibitor lenvatinib (E7080) in<br />

advanced medullary thyroid cancer (MTC). Presenting Author: Martin<br />

Schlumberger, Institut Gustave Roussy, Villejuif, France<br />

Background: Lenvatinib is an oral tyrosine kinase inhibitor targeting<br />

VEGFR1-3, FGFR1-4, RET, KIT and PDGFR�. In phase I studies <strong>of</strong><br />

lenvatinib partial responses (PR) were observed in thyroid as well as<br />

melanoma, endometrial, and renal cancers. Methods: Patients (pts) with<br />

unresectable MTC and disease progression demonstrated by RECIST<br />

during the prior 12 months were enrolled. Pts may have received prior<br />

VEGFR targeted therapy. Pts were treated with a starting dose <strong>of</strong> lenvatinib<br />

24 mg once daily in 28 day cycles until disease progression or development<br />

<strong>of</strong> unmanageable toxicities. Primary end point was Response Rate (RR) by<br />

RECIST. Tumor genetic analysis and circulating cytokine and angiogenic<br />

factors (CAF) analysis were performed. Results: 59 pts were enrolled (med<br />

age: 52; Male: 63%;) and are evaluable for response. 54% <strong>of</strong> pts required<br />

dose reduction for management <strong>of</strong> toxicity, and 22% were withdrawn from<br />

therapy due to toxicity. The most common treatment-related adverse events<br />

were proteinuria 58% (Gr3: 2%), diarrhea 56% (Gr3: 5%), hypertension<br />

48% (Gr3: 7%), fatigue 44% (Gr3: 5%), decreased appetite 41% (Gr3:<br />

5%), nausea 34% (Gr3: 0), and weight decreased 32% (Gr3: 3%). No Gr4<br />

events were reported for these event categories. Confirmed PRs were<br />

observed in 21pts (RR: 36%, 95% CI: 24-49) based on independent<br />

imaging review (IIR) and 29 pts (RR: 49%, 95% CI: 36-62) based on<br />

investigator assessment. For pts who received prior VEGFR-directed treatment<br />

(n�26) RR�35% (IIR); with no prior VEGFR-directed treatment<br />

(n�33) RR�36 % (IIR). Median PFS by IRR is 9.0 mo (95% CI: 7.0-)<br />

(based on minimum 8 mo. f/u, 46% events observed). There was no clear<br />

difference in treatment response between RET-mutant (RET-mu) and<br />

RET-wild type (RET-wt) patients. Low baseline levels <strong>of</strong> ANG2, sTie-2, HGF<br />

and IL-8 were associated with greater tumor shrinkage and prolonged PFS<br />

whereas high baseline levels <strong>of</strong> VEGF and sVEGFR3 were associated with<br />

greater tumor shrinkage. Conclusions: Lenvatinib administered orally at a<br />

dose <strong>of</strong> 24 mg once daily to patients with MTC is associated with<br />

manageable toxicity and a RR <strong>of</strong> 36%, identifying lenvatinib as a promising<br />

new potential therapeutic agent for treating patients affected with this<br />

disease.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


5592 General Poster Session (Board #32B), Sat, 1:15 PM-5:15 PM<br />

Final results <strong>of</strong> a phase II study <strong>of</strong> sorafenib in combination with<br />

carboplatin and paclitaxel in patients with metastatic or recurrent squamous<br />

cell cancer <strong>of</strong> the head and neck (SCCHN). Presenting Author:<br />

George R. Blumenschein, University <strong>of</strong> Texas M. D. Anderson Cancer<br />

Center, Houston, TX<br />

Background: Cytotoxic chemotherapy (CT) is the current standard treatment<br />

for metastatic/recurrent SCCHN. The prognosis for these patients (PTS) is<br />

poor with a median progression free survival (PFS) <strong>of</strong> 4 months with CT.<br />

Survival in PTS with SCCHN may correlate inversely with the number <strong>of</strong><br />

angiogenic growth factors secreted. Sorafenib is a potent inhibitor <strong>of</strong> c-Raf,<br />

b-Raf, VEGFR-1/2/3 and PDGFR-�. To investigate the hypothesis that a<br />

multi-targeted TKI added to chemotherapy would improve outcomes in<br />

patients with metastatic/recurrent SCCHN, we performed a phase II trial <strong>of</strong><br />

paclitaxel and carboplatin and sorafenib (PCS). Methods: PTS were<br />

required to have ECOG PS 0-1, measurable disease, controlled blood<br />

pressure, and may have received one regimen <strong>of</strong> induction, concomitant or<br />

adjuvant CT, but not CT for recurrent/metastatic disease. Sites <strong>of</strong> primary<br />

disease excluded nasopharynx and paranasal sinus. Treatment consisted P<br />

200mg/m2 and C AUC 6 intravenously on day 1 followed by S 400 mg orally<br />

bid (days 2-19) in every 3-week cycles. Primary endpoint was PFS,<br />

targeting median PFS <strong>of</strong> 6 months (M). Secondary endpoints include<br />

overall survival (OS), response rate (RR), exploratory biomarkers, and<br />

toxicity. Results: Forty-eight PTS with SCCHN were enrolled with 44 PTS<br />

evaluable for PFS and response using RECIST. Median age: 56 years<br />

(22-79 years), 89% male, median ECOG PS 1. Median follow-up was 24.1<br />

M. RR was 55% (n�24), disease control rate was 84% (n�37), median<br />

PFS was 8.51 M (95% CI: 5.98 ~ 13 months), and median OS was 22.6 M<br />

(95% CI: 13.1 - NA months). Grade ³3 treatment related toxicities included<br />

hand-foot syndrome (n�10), neutropenia (n�5), pain (n�6), elevated<br />

lipase (n�4), elevated amylase (n�3), anemia (n�3), fatigue (n�2),<br />

hypertension (n�2), neuropathy (n�2), febrile neutropenia (n�2), and<br />

thrombocytopenia (n�2). Conclusions: The combination <strong>of</strong> PCS was well<br />

tolerated and had encouraging activity in recurrent/metastatic SCCHN.<br />

Blood-based and tissue biomarkers are being analyzed. Final outcomes,<br />

toxicity, and correlative biomarker data will be reported.<br />

5594 General Poster Session (Board #32D), Sat, 1:15 PM-5:15 PM<br />

Long-term survival after distant metastasis in oropharyngeal carcinoma.<br />

Presenting Author: Sean M. McBride, Harvard Radiation Oncology Program,<br />

Boston, MA<br />

Background: The possibility <strong>of</strong> cure after distant metastases (DM) in<br />

oropharyngeal carcinoma (OPC) is uncertain. We conducted a retrospective<br />

cohort study to determine outcome and factors predictive <strong>of</strong> survival after<br />

metastasis in patients with OPC. Methods: From 2003-2010, 24 patients<br />

definitively treated (96% with concurrent chemo-radiation) at the Massachusetts<br />

General Hospital for OPC subsequently developed DM. At initial<br />

diagnosis, HPV status was available in 13 patients, 12 <strong>of</strong> whom were<br />

positive; six patients had T4 and 12 N2c-3 disease. Imaging studies<br />

pertinent to the diagnosis <strong>of</strong> DM were re-reviewed; 83% <strong>of</strong> patients had DM<br />

pathologically confirmed. Overall survival (OS) was defined from 1st<br />

radiographic evidence <strong>of</strong> DM to either death or last follow-up. Cox<br />

regression was used to evaluate factors predictive <strong>of</strong> OS after DM. Results:<br />

Median time to DM after initial treatment was 8.25 months (range,<br />

1.6-24). Twenty (83%) patients had isolated distant failure; <strong>of</strong> these, 6<br />

had limited (1 lesion or 2 adjacent lesions) single-organ disease. Eighteen<br />

(75%) had treatment after DM (83% had chemotherapy, 61% radiation,<br />

and 33% surgery). Median survival after distant metastasis was 19.2<br />

months. Median follow-up for survivors was 23.5 months (range, 7.9-<br />

60.3). In multivariate analysis, limited single-organ disease (HR�0.14,<br />

p�0.01, 95% CI 0.031, 0.67) was predictive <strong>of</strong> improved survival after<br />

DM; T4 disease at initial diagnosis predicted for decreased OS after DM<br />

(HR�3.9, p�0.01, 95% CI 1.31, 11.89). For patients with limited<br />

single-organ disease, the 2-year OS was 82% compared to 32% with<br />

extensive metastases (p � 0.006). Of the 6 patients with limited,<br />

single-organ metastatic disease, two remained alive at 59.4 and 60.5<br />

months without evidence <strong>of</strong> disease; both had HPV-positive tumors. One<br />

patient had a solitary hepatic failure and received neoadjuvant chemotherapy<br />

followed by partial hepatectomy; the other had a solitary lung<br />

metastasis and received wedge resection followed by radiation. Conclusions:<br />

Limited, single-organ metastatic disease predicts improved survival after<br />

DM in OPC. Cure is possible in this group. Patients with limited,<br />

single-organ distant disease should be considered for aggressive, local<br />

salvage treatment.<br />

Head and Neck Cancer<br />

379s<br />

5593 General Poster Session (Board #32C), Sat, 1:15 PM-5:15 PM<br />

TPF induction chemotherapy and pathologic response for patients with<br />

locally advanced and resectable oral squamous cell carcinoma. Presenting<br />

Author: Lai-ping Zhong, Department <strong>of</strong> Oral and Maxill<strong>of</strong>acial Surgery,<br />

Ninth People’s Hospital, School <strong>of</strong> Medicine, Shanghai Jiao Tong University,<br />

Shanghai, China<br />

Background: The role <strong>of</strong> induction chemotherapy in locally advanced and<br />

resectable oral squamous cell carcinoma has not been well issued.<br />

Methods: A prospective, open label, parallel, and interventional randomized<br />

control trail has been performed to evaluate the induction chemotherapy <strong>of</strong><br />

TPF protocol in resectable oral squamous cell carcinoma (OSCC) patients<br />

at clinical stage III and IVA. The patients received two cycles <strong>of</strong> TPF<br />

induction chemotherapy (75 mg/m2 docetaxel d1, 75mg/m2 cisplatin d1,<br />

and 750mg/m2 5-fluorouracil d1-5) followed by radical surgery and<br />

post-operative radiotherapy with a dose from 54 to 66 Gy (the experimental<br />

group) or surgery and post-operative radiotherapy (the control group).<br />

Post-surgical pathologic examination was performed to determine a positive<br />

response or negative response. A positive response was defined as<br />

absence <strong>of</strong> any tumor cells (pathologic complete response) or presence <strong>of</strong><br />

scattered foci <strong>of</strong> a few tumor cells (minimal residual disease with �10%<br />

viable tumor cells). The primary endpoint is the survival rate; the secondary<br />

endpoint is the local control and safety. This study has been approved by<br />

institutional ethics committee at Ninth People’s Hospital, School <strong>of</strong><br />

Medicine, Shanghai Jiao Tong University. Survival analysis was conducted<br />

with the Kaplan-Meier method. Results: 256 patients were enrolled in this<br />

trail and 224 patients (111 in experiment group and 113 in control group)<br />

finished the whole treatment protocol. After a median follow-up <strong>of</strong> 21<br />

months (ranging 6-43 m). The pathologic positive response rate was 29.7%<br />

(33/111), and negative response rate was 70.3% (78/111). The patients<br />

with positive response had a better disease free survival (38.5�2.1m,<br />

95%CI 34.4-42.6m, P�0.003) compared with those with negative response<br />

(24.6�2.1m, 95%CI 20.6-28.7m) and control group (31.0�1.6m,<br />

95%CI 27.9-34.1m). The toxicity <strong>of</strong> induction chemotherapy could be<br />

tolerated. Conclusions: Pathologic positive response to TPF induction<br />

chemotherapy could benefit the patients with locally advanced and<br />

resectable OSCC. However, further long-term follow-up is needed to<br />

confirm the benefit on survival and local control.<br />

5595 General Poster Session (Board #32E), Sat, 1:15 PM-5:15 PM<br />

Progressive increasing in the risk <strong>of</strong> second and subsequent malignant<br />

tumors in patients with a head and neck cancer: A validation study with<br />

SEER data. Presenting Author: Xavier Leon, Hospital Sant Pau, Barcelona,<br />

Spain<br />

Background: Patients with a head and neck squamous cell carcinoma<br />

(HNSCC) index tumor have a increased risk (2-4% per year) for appearance<br />

<strong>of</strong> a second malignant neoplasms (SMN) and higher risk for successive<br />

malignant tumors during the follow-up. The objective <strong>of</strong> our study was to<br />

validate this concept with data <strong>of</strong> patients included in the Surveillance<br />

Epidemiology and End Results (SEER) program (1973-2008). Methods:<br />

We performed a population-based cohort study <strong>of</strong> 149.328 patients with a<br />

primary oral cavity, oropharynx, hypopharynx and larynx squamous cell<br />

carcinoma index tumor, included in the SEER program, to analize the<br />

actuarial survival-free <strong>of</strong> second and successive tumors. A total <strong>of</strong> 11.948<br />

(8%) patients had one or more malignant tumors before the diagnosis <strong>of</strong><br />

HNSCC. Results: During the follow-up period, a total <strong>of</strong> 31.507 new SMN<br />

appeared. There was a progressive and significant increase in the risk <strong>of</strong><br />

SMN. The annual hazard ratio for 2nd to 7th successive SMN was 2.3%,<br />

2.7%, 3.9%, 5.4%, 8.9% and 19.1% annual risk respectively. Additionally<br />

11 patients had 8th SMN. We observed a progressive increase in the<br />

risk <strong>of</strong> appearence <strong>of</strong> new malignant tumors located in and outside the<br />

aerodigestive tract. The increase in the risk <strong>of</strong> SMN was higher for tumors<br />

located in the aerodigestive tract than tumors located outside aerodigestive<br />

tract. It was a tendency towards the increase in the proportion <strong>of</strong> HNSCC<br />

with the appearance <strong>of</strong> new tumors, with a decrease in the proportion <strong>of</strong> the<br />

malignant tumors located in the lung and in locations outside the<br />

aerodigestive tract. There were significant differences inthe risk <strong>of</strong> SMN<br />

between second and third, third and fourth, fourth and fifth, and sist and<br />

seventh tumors (p�0.0001). Conclusions: In patients with HNSCC there is<br />

a progressive increase in the risk <strong>of</strong> appearance <strong>of</strong> successive tumors.<br />

Previous tobacco and alcohol consumption, persistence in the exposure to<br />

carcinogens and individual susceptibility (genetic) could play a role in the<br />

increased risk <strong>of</strong> SMN. We have validated this concept with data from the<br />

SEER program.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


380s Head and Neck Cancer<br />

5596^ General Poster Session (Board #32F), Sat, 1:15 PM-5:15 PM<br />

Contralateral parotid volume as an objective marker for adaptive radiotherapy<br />

in head and neck cancer. Presenting Author: Patrick James<br />

Gagnon, Southcoast Centers for Cancer Care, Fairhaven, MA<br />

Background: Deformable image registration simplifies the process <strong>of</strong> adaptive<br />

radiotherapy and allows for sophisticated dosimetric analyses <strong>of</strong><br />

anatomic changes. Lacking is an objective measurement that could reliably<br />

predict for undesirable dosimetric changes during a course <strong>of</strong> radiotherapy.<br />

We analyzed daily changes in contralateral parotid volume and correlated<br />

these changes to mean dose delivered to the parotid gland over a course <strong>of</strong><br />

radiotherapy in four patients. Methods: Daily cone-beam CT (CBCT) scans<br />

<strong>of</strong> four patients were imported into the MIM Maestro s<strong>of</strong>tware. The<br />

simulation CT scan was registered to 35 daily CBCT’s per patient using a<br />

sequence <strong>of</strong> rigid and deformable registrations. Parotid contours were<br />

resampled and a deformed reconstruction <strong>of</strong> the planning CT was exported<br />

to the Eclipse treatment planning s<strong>of</strong>tware. The CT reconstructions were<br />

extended superiorly and inferiorly and the original fluence map was<br />

re-computed on the reconstructions with the isocenter at daily treatment<br />

position. Mean dose percent change and mean volume percent change<br />

were calculated for all CBCT’s and averaged across all patients. Average<br />

mean dose percent change was plotted against average mean volume<br />

change and evaluated with simple regression analysis. Results: A mean <strong>of</strong><br />

31 CBCT’s per patient were evaluated. Contralateral parotid volume<br />

percent changes for all patients were -31%, -40%, -40%, and -22% with a<br />

mean percent change <strong>of</strong> -33%. Mean dose percent changes were 25%,<br />

35%, 19%, and 25% with a mean percent change <strong>of</strong> 26% and a mean<br />

absolute dose increase to the parotids <strong>of</strong> 6.8 Gy. Simple regression analysis<br />

identified a coefficient <strong>of</strong> determination (R2 ) <strong>of</strong> 0.23 where the independent<br />

variable is volume percent change and the dependent variable is mean<br />

dose percent change. Conclusions: Daily or weekly volume assessment <strong>of</strong><br />

critical volumes is feasible utilizing deformable image registration and<br />

CBCT. Our data suggests there may be a correlation between parotid<br />

volume changes and increases in mean parotid dose though no causality is<br />

inferred. This relationship could be useful as a simple objective volumetric<br />

threshold measurement to prompt adaptive re-planning and warrants<br />

further investigation.<br />

TPS5598 General Poster Session (Board #32H), Sat, 1:15 PM-5:15 PM<br />

LUX-H&N 1: A phase III, randomized trial <strong>of</strong> afatinib versus methotrexate<br />

(MTX) in patients (pts) with recurrent/metastatic (R/M) head and neck<br />

squamous cell carcinoma (HNSCC) who progressed after platinum-based<br />

therapy. Presenting Author: Jean-Pascal H. Machiels, Clinique Universitaires<br />

St-Luc, Brussels, Belgium<br />

Background: EGFR (ErbB1) is expressed in 90% <strong>of</strong> HNSCC and associated<br />

with poor prognosis. Despite clinical benefit (CB) <strong>of</strong> EGFR-targeted<br />

treatments such as cetuximab treatment resistance will occur resulting in a<br />

need for novel targeted treatments to improve prognosis. Afatinib is an<br />

ErbB Family Blocker that irreversibly blocks signaling from all relevant<br />

ErbB Family dimers and may overcome limitations <strong>of</strong> current EGFRtargeted<br />

treatments in HNSCC. In a randomized, pro<strong>of</strong>-<strong>of</strong>-concept, Phase II<br />

trial, afatinib demonstrated comparable anti-tumor activity to cetuximab in<br />

pts with R/M HNSCC progressing after platinum-based therapy (Seiwert TY,<br />

et al; THNO, November 2011; Abs 40). Methods: LUX-H&N 1 is a phase III,<br />

randomized, open-label trial (NCT01345682) evaluating efficacy and<br />

safety <strong>of</strong> afatinib vs. MTX in pts with R/M HNSCC who have progressed after<br />

platinum-based therapy. Key eligibility criteria: confirmed R/M HNSCC not<br />

amenable to salvage surgery/radiotherapy; documented PD after cisplatin<br />

and/or carboplatin for R/M disease; any other than 1 previous platinumbased<br />

regimen for R/M disease; ECOG status 0 or 1; no PD �3 months <strong>of</strong><br />

completion <strong>of</strong> curatively intended treatment for locoregionally advanced or<br />

metastatic HNSCC; and no prior EGFR-targeted small molecules treatment.<br />

Eligible pts are stratified into four strata: ECOG performance score (0<br />

vs. 1) and prior use <strong>of</strong> EGFR-targeted antibody therapy (Yes/No) given in the<br />

R/M setting. Pts are randomized 2:1 to: afatinib (40mg/d orally) or MTX<br />

(40mg/m2 IV weekly). Dose changes are permitted according to absence/<br />

presence <strong>of</strong> drug-related AEs (afatinib: escalation to 50mg/d and/or<br />

reduction to 40, 30 then 20mg/d; MTX: escalation to 50mg/m2 and/or<br />

reduction to 40, 30 then 20mg/m2 ). Pts receive continuous treatment until<br />

PD or AEs requiring withdrawal. Randomized treatment may continue<br />

beyond PD in case <strong>of</strong> CB as judged by the investigator. The primary<br />

endpoint is PFS and secondary endpoints include OS, OR, health-related<br />

QOL and safety. Target enrolment is 474 pts and completion <strong>of</strong> pt<br />

recruitment and data analyses are awaited.<br />

5597 General Poster Session (Board #32G), Sat, 1:15 PM-5:15 PM<br />

<strong>Clinical</strong> outcomes and prognostic factors <strong>of</strong> 582 nasopharyngeal carcinoma<br />

patients treated with intensity-modulated radiotherapy. Presenting Author:<br />

Jin Yi Lang, Sichuan Cancer Hospital and Institute, Chengdu, China<br />

Background: To evaluate the 5-yrs clinical outcomes and prognostic factors<br />

<strong>of</strong> 582 nasopharyngeal carcinoma (NPC) patients treated with intensity<br />

modulated radiotherapy (IMRT). Methods: 582 NPC patients primarily<br />

treated with IMRT in Sichuan Cancer Hospital from Jan.2001 to Dec.2004<br />

were analyzed retrospectively, including 460 males and 122 females. The<br />

prescription dose was delivered as follows: gross target volume (GTVnx)<br />

67-76Gy in 30-33 fractions, positive neck lymph nodes (GTVln-R/L)<br />

60-70Gy in 30-33 fractions, The lower neck and the supraclavicular fossae<br />

were irradiated with the conventional 2D technique using an anterior field.<br />

The Kaplan-Meier method was used to calculate the local-regional control<br />

rate (LRC) and overall survival rate (OS). Acute and late toxicities were<br />

graded according to the Radiation Therapy Oncology Group (RTOG)<br />

radiation morbidity scoring criteria, meanwhile analyze the prognostic<br />

factors. Results: The median follow-up interval was 66.4 months. The<br />

5-year local control, regional control, distant metastasis-free survival,<br />

disease free survival, disease specific survival and overall survival rate was<br />

89.8%, 95.2%, 74.1%, 69.6%, 83.2% and 77.1%. 133 patients died<br />

during the follow-up. The 5-year DMFS <strong>of</strong> IMRT and IMRT combined with<br />

chemotherapy was 62.1% and 70.9%, the OS <strong>of</strong> them was 75.9% and<br />

79.1%. The incidence <strong>of</strong> grade 3 acute and late toxicity was 38.3% and<br />

4.2% respectively.Univariate analysis revealed that gender, T/N stage,<br />

clinical stage, radiotherapy interruption, anemia and weight loss was the<br />

significant prognostic factor for the OS. Multivariate analysis showed that N<br />

stage, radiotherapy interruption, chemotherapy, the volume <strong>of</strong> GTVnx, age,<br />

anemia and weight loss was the independent prognostic factors for the OS.<br />

Conclusions: The 5-year local control and overall survival rate <strong>of</strong> NPC<br />

treating with IMRT was 89.8% and 77.1%. The clinical stage, N stage ,<br />

volume <strong>of</strong> GTVnx and chemotherapy was the main prognostic factor for the<br />

OS. The acute and late toxicities were mainly grade I and II. Distant<br />

metastasis was the main pattern <strong>of</strong> failure.<br />

TPS5599 General Poster Session (Board #33A), Sat, 1:15 PM-5:15 PM<br />

LUX head and neck 2: A randomized, double-blind, placebo-controlled,<br />

phase III study <strong>of</strong> afatinib as adjuvant therapy after chemoradiation in<br />

primarily unresected, clinically high-risk, head and neck cancer patients.<br />

Presenting Author: Barbara Burtness, Fox Chase Cancer Center, Philadelphia,<br />

PA<br />

Background: Locally advanced squamous cell cancer <strong>of</strong> the head and neck<br />

(SCCHN) is treated with curative intent, but recurrence and death are<br />

common. SCCHN frequently over-expresses EGFR (ErbB1). Co-expression<br />

<strong>of</strong> other HER family members such as HER2 (ErbB2) may contribute to<br />

resistance to EGFR inhibition, which is the only validated targeted therapy<br />

in SCCHN. Methods: The trial investigates if adjuvant afatanib, an irreversible<br />

ErbB family blocker, which has shown preclinical activity against all<br />

ErbB dimers including EGFR and HER2, reduces the risk <strong>of</strong> recurrence in<br />

high-risk patients who have no evidence <strong>of</strong> disease following platinumbased<br />

chemoradiation with or without neck dissection. Patients are eligible<br />

who have received definitive chemoradiation to a minimum <strong>of</strong> 66 Gy, with<br />

concurrent cisplatin (�200 mg/m2 ) or carboplatin (�AUC 9), for SCC <strong>of</strong><br />

the oral cavity, oropharynx, or hypopharynx or larynx. Patients with base <strong>of</strong><br />

tongue or tonsil cancer and �10 pack years <strong>of</strong> tobacco use, as well as those<br />

with nasopharynx, sinus or salivary gland cancer, are excluded. Adequate<br />

bone marrow, liver and kidney function is required. Prior therapy with<br />

investigational agents or EGFR inhibitors is not permitted. Randomization<br />

must take place within 16 weeks <strong>of</strong> the completion <strong>of</strong> chemoradiation with<br />

or without subsequent neck dissection. Patients are randomized 2:1 to<br />

afatinib 40 mg po qd or placebo, and treatment continues for 18 months in<br />

the absence <strong>of</strong> disease recurrence, second primary tumors, or intolerance<br />

to the study medication. Dose escalation to 50 mg qd is undertaken in<br />

patients with no side effects, and stepwise dose reduction to 30 or 20 mg<br />

po qd for diarrhea, skin toxicity or other adverse events is permitted. The<br />

primary endpoint is disease-free survival (DFS). The study is planned to<br />

accrue approximately 669 patients worldwide, with a 90% power to detect<br />

a hazard ratio <strong>of</strong> 0.72. Secondary endpoints are DFS at 2 years, overall<br />

survival, health-related quality <strong>of</strong> life, and safety.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


TPS5600 General Poster Session (Board #33B), Sat, 1:15 PM-5:15 PM<br />

A phase III study <strong>of</strong> standard fractionation radiotherapy with concurrent<br />

high-dose cisplatin versus accelerated fractionation radiotherapy (RT) with<br />

panitumumab in patients with locally advanced stage III and IV squamous<br />

cell carcinoma <strong>of</strong> the head and neck (SCCHN) (NCIC <strong>Clinical</strong> Trials Group<br />

HN.6). Presenting Author: John N. Waldron, Princess Margaret Hospital,<br />

Toronto, ON, Canada<br />

Background: Standard treatment for locally advanced SCCHN, chemoradiotherapy<br />

(CRT), leads to significant acute and long-term morbidity. The<br />

demonstration that EGFR antibody (Ab) therapy improves outcome when<br />

added to standard RT (Bonner NEJM 2006) and that altered fractionation<br />

RT improves outcome compared to standard RT that is <strong>of</strong> similar magnitude<br />

as CRT (Bourhis Lancet 2006), led to the hypothesis that the combination<br />

<strong>of</strong> the two treatment strategies will improve efficacy compared to standard<br />

CRT with good tolerability. Methods: HN.6 is a Canadian phase III<br />

randomized study comparing standard RT 70Gy/35 over 7 weeks �<br />

cisplatin 100mg/m2 d 1, 22, 43 to accelerated RT 70Gy/35 over 6 weeks �<br />

panitumumab (anti EGFR Ab)) 9mg/kg 1 week prior to RT, d15, 36. Key<br />

eligibility criteria are: SCC <strong>of</strong> oral cavity, oropharynx, larynx or hypopharynx;<br />

TanyN�M0 or T3-4N0M0; adequate organ function and PS. Primary<br />

endpoint is progression free survival (PFS). Secondary endpoints include<br />

OS, local PFS, regional PFS, distant metastases, swallowing related QOL,<br />

functional swallowing outcomes, and economic evaluation (healthcare<br />

utilization, health utilities, indirect costs). Tissue and blood collection will<br />

allow biomarker evaluation including HPV status. Real time quality review<br />

<strong>of</strong> RT plans with plan data and radiology archiving will allow future analysis<br />

<strong>of</strong> RT parameters relative to toxicity and patterns <strong>of</strong> failure. <strong>Clinical</strong><br />

epidemiological data will be prospectively collected and correlated with<br />

biomarkers and outcome. Planned sample size is 320 patients over 3.2<br />

years with 3 more years <strong>of</strong> follow-up and target hazard ratio � 0.7 (absolute<br />

difference in control arm 2 year PFS <strong>of</strong> 12%, power 80%, 2-tail type 1 error<br />

0.05). If superiority is not demonstrated, noninferiority will be tested. One<br />

interim analysis is planned. Conduct to Date: Study activation: Dec 2008<br />

and completed accrual in Nov 2011. In Oct 2011, the DSMC recommended<br />

trial continuation. Supported by CCSRI grant 021039 and Amgen<br />

Inc. <strong>Clinical</strong>Trials.gov: NCT00820248.<br />

TPS5602 General Poster Session (Board #33D), Sat, 1:15 PM-5:15 PM<br />

A pilot study <strong>of</strong> raltegravir and cisplatin in head and neck squamous cell<br />

carcinoma (HNSCC). Presenting Author: Julie E. Bauman, University <strong>of</strong><br />

New Mexico, Albuquerque, NM<br />

Background: Metnase is a recently characterized DNA repair component<br />

present in anthropoid primates. Metnase, the fusion <strong>of</strong> a SET histone<br />

methylase domain and a Transposase nuclease (Tn) domain, enhances<br />

DNA double-stranded break (DSB) repair. The Tn domain trims free DNA<br />

ends for optimal end-joining, and is required to re-start stalled replication<br />

forks. The SET domain di-methylates H3K36 at the DSB, recruiting<br />

non-homologous end-joining repair components. Cisplatin (CDDP), the<br />

central chemotherapy in HNSCC, covalently binds DNA leading to intraand<br />

interstrand crosslinks (ICL). In addition to blocking strand transcription<br />

and segregation, the ICL stalls DNA replication fork progression<br />

leading to collapse and DSB. The role <strong>of</strong> Metnase in DNA repair, including<br />

overcoming the stalled replication fork, makes it a potential therapeutic<br />

target. We hypothesize that Metnase inhibition may be a useful adjunct to<br />

CDDP. 3D modeling <strong>of</strong> the Metnase Tn domain identified significant<br />

homology with human immunodeficiency virus 1 (HIV-1) integrase. A<br />

virtual screen <strong>of</strong> the Chem Div library identified the HIV-1 integrase<br />

inhibitor, raltegravir, as a lead compound for inhibiting the Metnase Tn<br />

domain. We designed a pilot study in HNSCC to evaluate potentiation <strong>of</strong><br />

CDDP DNA damage by raltegravir. Methods: The primary objective <strong>of</strong> this<br />

window-<strong>of</strong>-opportunity study is to analyze biomarkers <strong>of</strong> DNA damage, fork<br />

arrest, and apoptosis in serial tumor biopsies from patients (pts) with<br />

HNSCC undergoing CDDP, with and without raltegravir. Eligible pts: 1)<br />

have HNSCC with tumor site amenable to repeat, awake biopsy; 2) are<br />

appropriate candidates for CDDP. Pts are treated with 2 doses <strong>of</strong> CDDP 30<br />

mg/m2 . One CDDP dose is administered with raltegravir 400 mg bid for 5<br />

days, starting the day before CDDP. Pts undergo 3 research biopsies, at<br />

baseline and 48-72 hours after each CDDP. Serial biopsies will be<br />

evaluated for expression changes in �H2AX, Annexin V, pChk1, pChk2, and<br />

p53BP1 by immunohistochemistry. Numerical increase in biomarker<br />

expression in tumors exposed to CDDP-raltegravir vs. CDDP will justify<br />

development <strong>of</strong> a phase I/II study. Four <strong>of</strong> 12 pts have enrolled and<br />

completed all biopsies. Supported by a grant from the <strong>American</strong> Cancer<br />

<strong>Society</strong>.<br />

Head and Neck Cancer<br />

381s<br />

TPS5601 General Poster Session (Board #33C), Sat, 1:15 PM-5:15 PM<br />

Phase II trial <strong>of</strong> neoadjuvant chemotherapy for HPV-associated squamous<br />

cell carcinoma <strong>of</strong> the oropharynx followed by reduced-dose radiotherapy/<br />

chemoradiotherapy for responders or standard dose chemoradiotherapy for<br />

nonresponders. Presenting Author: Bhoomi Mehrotra, H<strong>of</strong>stra North Shore-<br />

LIJ School <strong>of</strong> Medicine, New Hyde Park, NY<br />

Background: Recent studies have established an improved outcome for HPV<br />

associated oropharyngeal squamous cell cancers with 3 year OS rates<br />

approaching 90%. Given that current aggressive treatment modalities for<br />

HNC include IMRT and chemotherapy (CT), they adversely affect the long<br />

term QOL <strong>of</strong> these patients who are considered highly curable. The primary<br />

objective <strong>of</strong> our study is to confirm the activity (CR � PR at 3 months post<br />

completion RT/CRT) <strong>of</strong> induction TPF followed by dose reduced IMRT �/concurrent<br />

CT, in nonsmokers with HPV associated, locally advanced SCC<br />

<strong>of</strong> H&N. Secondary objectives include: to define objective tumor RR to<br />

induction chemotherapy and to subsequent RT based treatment, per<br />

RECIST version 1.1 criteria, assess PFS and OS at 2 years,assess<br />

locoregional disease and distant disease control at 2 years, and monitoring<br />

<strong>of</strong> QOL instruments for HNC. Methods: Institutional IRB approval has been<br />

obtained for this study. Eligibility criteria include p16 expressing or ISH �<br />

SCC <strong>of</strong> the oropharynx, T1-3, N1-2, age � 17 years, adequate marrow and<br />

organ function and adequate PS. Exclusion criteria include T4 or N3<br />

disease, significant smoking history � 10 pack year lifetime exposure.<br />

Patients would be treated with standard TPF induction for 3 cycles followed<br />

by risk assessment. Pts. achieving locoregional CR or CR/PR at primary site<br />

would receive dose reduced RT (66 Gy) �/- CT, while those with SD/PD<br />

would receive standard CRT with full dose XRT. Post therapy assessments<br />

include serial evaluation <strong>of</strong> functional quality-<strong>of</strong>-life, including M. D.<br />

Anderson Dysphagia Inventory (MDADI) and Oropharyngeal swallowing<br />

efficiency (OPSE) measures <strong>of</strong> swallowing function, treatment related<br />

symptoms, dietary status assessments and formal sialometric measurement<br />

<strong>of</strong> parotid function. Interim analysis would include RR assessment at<br />

3 months post treatment in the first 12 patients who received dose reduced<br />

RT and compared with historical controls. Early stopping rules are built into<br />

the protocol. This study is currently open and patient accrual is ongoing.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


382s Health Services Research<br />

6000 Oral Abstract Session, Sat, 3:00 PM-6:00 PM<br />

Racial disparities in breast cancer survival. Presenting Author: Jeffrey H.<br />

Silber, The Children’s Hospital <strong>of</strong> Philadelphia and The University <strong>of</strong><br />

Pennsylvania Perelman School <strong>of</strong> Medicine, Philadelphia, PA<br />

Background: Reducing racial disparities in breast cancer survival has been a<br />

federal priority since the early 1990’s. We present a new method to assess<br />

disparities using sequential multivariate matching. We ask if racial disparities<br />

have increased or decreased over time and if so, what were potential<br />

reasons for such changes. Methods: We studied all women over 65 years <strong>of</strong><br />

age in the Medicare fee for service system diagnosed with breast cancer<br />

between 1991 and 2005 who were treated in one <strong>of</strong> 12 SEER sites (the<br />

sites in SEER since 1991). There were 5,251 black patients (74% early<br />

stage (I-III), 9% late stage (IV) and 17% missing stage) and 72,695 white<br />

patients (81% early stage, 5% late stage and 14% missing stage). All black<br />

cases represented the focal group for all matches. Using multivariate<br />

matching and the propensity score, white controls were matched to blacks<br />

in steps: (1) White controls matched to black cases on age and year <strong>of</strong><br />

diagnosis; (2) Age, year <strong>of</strong> diagnosis, and stage; (3): Age, year, stage,<br />

estrogen receptor status, grade, and 30 comorbidities. We then compare<br />

5-year survival in the Pre and Post-Taxane periods (1991-1998, 1999-<br />

2005). Results: When whites were matched to blacks on age and diagnosis<br />

year, 5-year Kaplan-Meier survival was 69.2% vs. 56.7%, P � 0.0001.<br />

Matching additionally on stage, differences � 64.1% vs. 56.7%, P �<br />

0.0001; Matching further on tumor characteristics and 30 comorbidities,<br />

the disparity reduced to 61.6% vs. 56.7%, P � 0.0001. Comparing trends<br />

over time, white-black differences in survival matched for age and year were<br />

67.6% vs. 55.2% (P � 0.0001) in the pre-Taxane era (difference �<br />

12.4%) and 71.2% vs. 58.7% (P � 0.0001) in the post Taxane era<br />

(difference � 12.5%); age and year matched paired racial differences were<br />

not different across eras (P � 0.389). Conclusions: While there may have<br />

been some improvements in overall survival, racial disparities in breast<br />

cancer survival have not improved, despite important policy initiatives and<br />

treatment advances. Adjusting for presentation at diagnosis does reduce<br />

differences in survival, but even these differences remain large and<br />

significant, suggesting that differences in both presentation and treatment<br />

given presentation are contributing to this disparity.<br />

6002 Oral Abstract Session, Sat, 3:00 PM-6:00 PM<br />

An international study <strong>of</strong> multitrial data investigating quality <strong>of</strong> life and<br />

symptoms as prognostic factors for survival in different cancer sites.<br />

Presenting Author: Chantal Quinten, European Organisation for Research<br />

and Treatment <strong>of</strong> Cancer Headquarters, Brussels, Belgium<br />

Background: The prognostic value for survival <strong>of</strong> HRQOL data derived from<br />

self-report questionnaires, has been well documented in cancer research.<br />

The objective <strong>of</strong> this study was to examine the prognostic value <strong>of</strong> HRQOL<br />

parameters for different cancer sites using one standardized and validated<br />

patient self-assessment tool. Methods: A total <strong>of</strong> 11 different cancer sites,<br />

pooled from 30 European Organisation for Research and Treatment <strong>of</strong><br />

Cancer (EORTC) Randomized Controlled Trials (RCTs), were selected for<br />

this study. For each cancer site, univariate and multivariate Cox proportional<br />

hazard modeling was used to assess the prognostic value (p�0.05) <strong>of</strong><br />

15 HRQOL parameters, assessed with the EORTC QLQ-C30 at baseline<br />

before randomization, for overall survival. Models were adjusted for the<br />

parameters age, gender, distant metastasis, World Health Organization<br />

performance status and stratified by clinical study. Results: A total <strong>of</strong> 7,417<br />

patients completed the EORTC QLQ-C30 before randomization. For brain<br />

cancer cognitive functioning (CF) (hazard ratio (HR) �0.95; p�.0001) was<br />

prognostic. For breast cancer nausea and vomiting (NV) (HR�1.17;<br />

p�0.0011) was a prognostic indicator. For colorectal cancer physical<br />

functioning (PF) (HR�0.93; p�.0001), NV (HR�1.07; p�.0001), and<br />

appetite loss (AP) (HR�1.07; p�.0001) predicted survival. For esophageal<br />

cancer PF (HR�0.88; p�0.0072) and for head and neck cancer NV<br />

(HR�1.14; p�0.0097) were prognostic. For lung cancer PF (HR�0.94;<br />

p�0.0006) and pain (HR�1.08; p�0.0001), for melanoma dyspnea<br />

(HR�1.06; p�.0001), for ovarian cancer NV (HR�1.2; p�.0001), for<br />

pancreatic cancer global QOL (HR�0.83; p�0.0073), for prostate cancer<br />

role functioning (RF) (HR�0.96; p�0.006) and AP (HR�1.07; p�.0001),<br />

and for testis cancer RF (HR�0.81; p�0.0144) were predictors <strong>of</strong><br />

survival. Conclusions: Our findings show that different HRQOL parameters<br />

provide prognostic information for survival for patients with different tumor<br />

sites and that no single HRQOL scale can predict survival in all cancer<br />

patients. Thus, each cancer site needs careful examination and no single<br />

QOL paramenter can predict survival in all cancer diseases.<br />

6001 Oral Abstract Session, Sat, 3:00 PM-6:00 PM<br />

Competing causes <strong>of</strong> mortality in long-term survivors <strong>of</strong> head and neck<br />

cancer. Presenting Author: Shrujal S. Baxi, Memorial Sloan-Kettering<br />

Cancer Center, New York, NY<br />

Background: Most head and neck cancers (HNC) recur within the first 3 yrs<br />

<strong>of</strong> diagnosis. Patients who live beyond this time point continue to have an<br />

excess risk <strong>of</strong> mortality. We investigated the causes <strong>of</strong> mortality in 3-yr<br />

survivors <strong>of</strong> HNC. Methods: We completed a retrospective cohort study<br />

using the Surveillance Epidemiology and End Results (SEER) database. We<br />

identified patients with a diagnosis <strong>of</strong> non-metastatic HNC between 1992<br />

and 2005 who survived 3 yrs from diagnosis. The primary outcome was<br />

death from all-causes, HNC, non-HNC malignancy, cardiovascular disease<br />

(CVD), and pulmonary disease (PD). We estimated the age-adjusted<br />

standardized mortality ratio (SMR; observed-to-expected (O/E)) and the<br />

absolute excess risk (AER; O-E), for all-cause and cause-specific mortality<br />

compared to US population data. Results: 31,772 long-term survivors were<br />

identified with disease arising in the larynx (40%), oral cavity (29%),<br />

oropharynx (26%), nasopharynx (3%) and hypopharynx (1%). In this<br />

cohort, 8191 (26%) were female, 3021 (10%) were black and 15,321<br />

(48%) were younger than 60 yrs. In this cohort, 16,697 (53%) had<br />

locally-advanced disease and 8353 (26%), 8721(27%) and 13,919<br />

(44%) received radiation alone, surgery alone or both. With a median<br />

follow-up <strong>of</strong> 6.4 years (3-17 years), there were 10,757 deaths due to: HNC<br />

(24%), non-HNC malignancy (31%), CVD (21%), PD (6%), and other<br />

causes (19%). Compared to the US population, the all-cause SMR was 5.4<br />

(95% CI 5.3-5.6) with an AER <strong>of</strong> 336 deaths per 10,000 person-years (py).<br />

The greatest excess risk was attributable to non-HNC malignancies (956<br />

per 10,000 py), CVD (533 per 10,000 py) and pulmonary disease (136 per<br />

10,000 py). Conclusions: HNC survivors experience excess mortality<br />

compared to the general population. Competing causes <strong>of</strong> mortality<br />

challenge long-term survival. Additional analytic studies with detailed data<br />

on tobacco history, treatment characteristics and co-morbidities are<br />

required to further evaluate associations with specific causes <strong>of</strong> death and<br />

individualize risk in this heterogenous population <strong>of</strong> survivors.<br />

6003 Oral Abstract Session, Sat, 3:00 PM-6:00 PM<br />

Regional variation in Medicare spending and survival among older adults<br />

with advanced cancer. Presenting Author: Gabriel Brooks, Dana-Farber<br />

Cancer Institute, Boston, MA<br />

Background: There is substantial regional variation in medical spending in<br />

the United States. Whether this variation extends to spending for cancer<br />

care and is associated with cancer survival outcomes is unknown. Methods:<br />

Patients aged 65 and older with incident advanced cancer were identified<br />

from the Surveillance, Epidemiology and End Results (SEER)-Medicare<br />

linked database. Advanced cancers included lung (stage IIIB/IV), pancreas<br />

(stage III/IV) colorectal, breast and prostate (stage IV). Medicare spending<br />

from 2 months pre-diagnosis until death or 12 months post-diagnosis was<br />

totaled in 2010 USD for Medicare-enrolled advanced cancer patients from<br />

80 hospital referral regions (HRR’s). HRRs were then ranked by mean<br />

composite 14-month spending and grouped into quintiles. The hazard ratio<br />

for death was assessed in each disease cohort for the comparison between<br />

the quintiles with lowest (Q1, referent) and highest (Q5) composite<br />

spending. Results: Mean composite 14-month spending ranged from<br />

$143,870 to $303,902 in HRR’s with the lowest and highest spending<br />

(Mason City, IA and Los Angeles, CA). As shown in the table, increased<br />

spending was associated with decreased risk <strong>of</strong> death for lung, colorectal<br />

and pancreas cancer but not for prostate or breast cancer. Conclusions:<br />

There is substantial regional variation in Medicare spending for patients<br />

with advanced cancer. This variation is associated with modest differences<br />

in risk <strong>of</strong> death between the lowest and highest quintiles <strong>of</strong> spending for<br />

lung, colorectal and pancreas cancer.<br />

Cohort Patients<br />

Mean spending<br />

Cohort Quintile 1 Quintile 5 Q1/Q5<br />

ratio<br />

HR for death,<br />

Q5 vs Q1 p<br />

Lung 36,312 $236,553 $192,477 $292,213 1.52 0.91 (0.87-0.94) �.0001<br />

Colorectal 9,912 $315,087 $261,545 $370,685 1.42 0.90 (0.83-0.97) 0.008<br />

Pancreas 6,993 $220,424 $169,396 $278,345 1.64 0.84 (0.78-0.91) �.0001<br />

Prostate 5,596 $153,376 $117,055 $195,460 1.67 1.09 (0.91-1.30) 0.363<br />

Breast 3,344 $222,661 $173,479 $270,266 1.56 1.01 (0.87-1.19) 0.859<br />

Composite 62,157 $229,058 $183,615 $281,740 1.53 NA NA<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


6004 Oral Abstract Session, Sat, 3:00 PM-6:00 PM<br />

Effect <strong>of</strong> early palliative care on health care costs in patients with<br />

metastatic NSCLC. Presenting Author: Joseph A. Greer, Massachusetts<br />

General Hospital, Boston, MA<br />

Background: Introducing palliative care soon after diagnosis for patients<br />

with metastatic non-small cell lung cancer (NSCLC) leads to improvements<br />

in quality <strong>of</strong> life, mood, end-<strong>of</strong>-life care, and possibly survival. We sought to<br />

investigate whether early palliative care is also associated with health care<br />

cost savings. Methods: This secondary analysis is based on a randomized<br />

controlled trial <strong>of</strong> 151 patients with newly-diagnosed, metastatic NSCLC<br />

presenting to an outpatient clinic at a tertiary cancer center between<br />

6/2006 and 7/2009. <strong>Part</strong>icipants received either early palliative care<br />

integrated with standard oncology care or standard oncology care alone. We<br />

queried participants’ electronic health records as well as our institution’s<br />

billing database to collect data on frequency and costs <strong>of</strong> outpatient clinic<br />

visits, inpatient hospitalizations, chemotherapy administration, and hospice<br />

services. The primary outcome was the difference in average resource<br />

use costs during the final month <strong>of</strong> life between groups. Results: By<br />

18-month follow up, 133 (88.1%) participants had died, and 125 (82.8%)<br />

had available data for this analysis. <strong>Part</strong>icipants in the early palliative care<br />

group had a mean cost savings <strong>of</strong> $2,282 (median�$2,432) per patient in<br />

total health care expenditures during the final month <strong>of</strong> life compared to<br />

the standard care group. The difference was primarily accounted for by<br />

lower costs for inpatient visits (mean saving per patient�$3,110) and<br />

chemotherapy administration (mean saving per patient�$640). Although<br />

expenditures for outpatient clinic visits were similar between groups, the<br />

costs for hospice services were greater for the early palliative care group<br />

because <strong>of</strong> the longer lengths <strong>of</strong> stay in hospice care (mean cost per<br />

patient�$1,125). Conclusions: Early palliative care for individuals diagnosed<br />

with metastatic NSCLC not only improves multiple patient outcomes<br />

but also may be associated with lower hospital resource use costs, primarily<br />

through decreased inpatient visits and chemotherapy administration at the<br />

end <strong>of</strong> life.<br />

6006 Oral Abstract Session, Sat, 3:00 PM-6:00 PM<br />

A comparison <strong>of</strong> primary care providers’ and oncologists’ preferences for<br />

different models <strong>of</strong> cancer survivorship care. Presenting Author: Winson Y.<br />

Cheung, British Columbia Cancer Agency, Vancouver, BC, Canada<br />

Background: There is increasing interest in developing more efficient and<br />

effective strategies for coordinating and delivering cancer and non-cancer<br />

related follow-up care to survivors. The objectives <strong>of</strong> this nationwide survey<br />

were to describe and compare US physician preferences for different<br />

cancer survivorship care models. Methods: The Survey <strong>of</strong> Physician Attitudes<br />

Regarding the Care <strong>of</strong> Cancer Survivors (SPARCCS) was mailed to<br />

PCPs and oncologists in order to evaluate their views regarding physician<br />

responsibilities, knowledge levels about survivorship, and cancer follow-up<br />

testing. Using weighted univariate and multivariate models, we analyzed<br />

PCPs’ and oncologists’ preferences for different cancer survivorship care<br />

models (PCP/shared vs. oncologist vs. non-physician provider) and examined<br />

how physician attitudes towards and self-efficacy with their own skills<br />

during breast and colorectal cancer follow-up affected these preferences.<br />

Results: Of 3,434 physicians surveyed, 2,202 (64%) responded <strong>of</strong> whom<br />

2,026 (59%) provided eligible outcomes for this study: 938 (46%) PCPs<br />

and 1,088 (54%) oncologists. In unadjusted analyses, most PCPs (51%)<br />

supported a PCP/shared care system whereas the majority <strong>of</strong> specialists<br />

(59%) strongly endorsed an oncologist-based model (p�0.001). A number<br />

<strong>of</strong> PCPs and oncologists (23% for both) preferred to involve non-physician<br />

providers. A significant proportion <strong>of</strong> cancer specialists (87%) did not feel<br />

that PCPs can take on the primary role for cancer follow-up. Many PCPs<br />

believed that they have the skills to perform breast and colorectal cancer<br />

follow-up (57%), detect recurrent cancers (74%), and <strong>of</strong>fer psychosocial<br />

support (50%), but only a minority (32%) were willing to assume exclusive<br />

responsibility. In adjusted analyses, PCPs already involved with cancer<br />

surveillance (43%) were more likely to prefer a PCP/shared care system<br />

than an oncologist-based survivorship care model (OR 2.08, 95%CI<br />

1.34-3.23, p�0.001). Conclusions: PCPs and oncologists have different<br />

preferences for models <strong>of</strong> cancer survivorship care. Prior involvement with<br />

cancer follow-up was one <strong>of</strong> the strongest predictors <strong>of</strong> PCPs’ willingness to<br />

assume this responsibility.<br />

Health Services Research<br />

383s<br />

6005 Oral Abstract Session, Sat, 3:00 PM-6:00 PM<br />

Cancer patient acceptance, understanding, and willingness to pay for<br />

pharmacogenetic testing (PGT). Presenting Author: Henrique Hon, Ontario<br />

Cancer Institute, Princess Margaret Hospital, Toronto, ON, Canada<br />

Background: PGT <strong>of</strong>fers the potential to improve cancer therapy through the<br />

use <strong>of</strong> specialized tests that can predict the level <strong>of</strong> efficacy and/or toxicity<br />

<strong>of</strong> specific treatments in an individual. However, there is currently little<br />

knowledge concerning cancer patient attitudes towards such testing in the<br />

clinical setting. Methods: We interviewed a broad cross-section <strong>of</strong> 278<br />

cancer patients (20% lung, 19% breast, 20% colorectal, 40% other) using<br />

hypothetical time, efficacy, toxicity and willingness-to-pay trade-<strong>of</strong>f PGT<br />

scenarios. Results: 153 potentially curable patients and 125 incurable<br />

patients received a separate series <strong>of</strong> trade-<strong>of</strong>f scenarios. For curative<br />

patients, 70% accepted chemo that had a 5% absolute improvement in<br />

cure rate and �5% <strong>of</strong> severe toxicity. Of these, 99% wanted PGT where the<br />

test identifies a subset <strong>of</strong> patients benefiting from chemo; the same<br />

individuals were willing to pay a median $2,000 (range: $0-25,000) for<br />

PGT and would accept a median wait time for PGT results <strong>of</strong> 21 days<br />

(0-90). Patient preferences were insensitive to variation <strong>of</strong> fractions <strong>of</strong><br />

individuals carrying the genetics associated with lack <strong>of</strong> benefit. In the<br />

incurable scenario, 90% <strong>of</strong> patients accepted palliative chemo with an<br />

80% response rate and a severe side effect rate <strong>of</strong> 5%. Of these, 98%<br />

wanted PGT, where there test identifies individuals at highest risk <strong>of</strong> severe<br />

toxicity; the same individuals were willing to pay a median $1,000<br />

($0-15,000) for PGT, and would accept PGT turnaround times <strong>of</strong> 14 days<br />

(1-90). Patient preferences were insensitive to variation <strong>of</strong> fractions <strong>of</strong><br />

individuals carrying the genetics associated with severe toxicity. The<br />

majority <strong>of</strong> patients (76% adjuvant; 87% metastatic) wanted to be involved<br />

in decision making regarding PGT; however, one in five patients (20%<br />

adjuvant; 22% metastatic) admitted that they lacked a basic understanding<br />

<strong>of</strong> what PGT means and its clinical implications. Conclusions: Among<br />

cancer patients willing to undergo chemo, almost all wanted PGT and were<br />

willing to pay for it, waiting several weeks for results. While patients had a<br />

strong desire to be involved in decision making for PGT, a considerable<br />

proportion lacked the necessary knowledge to make informed choices.<br />

6007 Oral Abstract Session, Sat, 3:00 PM-6:00 PM<br />

Who gets a referral to oncologists and subsequent treatments for stages III<br />

and IV non-small cell lung cancer (NSCLC)? Presenting Author: Bernardo<br />

H.L. Goulart, Fred Hutchinson Cancer Research Center, Seattle, WA<br />

Background: We identified the physicians initially involved in the management<br />

<strong>of</strong> stages III and IV NSCLC, and explored associations <strong>of</strong> patient and<br />

their initial physician factors with referrals to oncologists and subsequent<br />

receipt <strong>of</strong> guideline-based therapies (GBTs) endorsed by the National<br />

Comprehensive Cancer Network. Methods: Using a retrospective cohort<br />

design, we identified patients with a new diagnosis <strong>of</strong> stages III and IV<br />

NSCLC from 01/01/2000 to 12/31/2005 included in the Surveillance,<br />

Epidemiology, and End Results-Medicare database. After collecting patient<br />

sociodemographic, tumor, and treatment data, we linked Unique<br />

Physician Identifier Numbers (UPINs) from Medicare claims to the <strong>American</strong><br />

Medical Association Masterfile database to identify the initial physicians<br />

and subsequent referrals to cancer specialists, defined as surgeons,<br />

radiation oncologists and oncologists. We used logistic regression to<br />

explore associations between: 1) patient and initial physician independent<br />

variables with referrals to oncologists; 2) referrals to different combinations<br />

<strong>of</strong> cancer specialists with receipt <strong>of</strong> stage-specific GBTs, adjusted for<br />

confounders. The follow-up period was 12 months or up to 12/31/2006.<br />

Results: For 28,977 patients, mean age was 75 years, 53% were male,<br />

83% were white, 51% had stage IV, 37% initially saw an internal medicine<br />

doctor, 84% saw at least an oncologist, 31% saw all 3 types <strong>of</strong> cancer<br />

specialists, and 44% received GBTs. Younger age, white race, stage IV,<br />

higher income, lower co-morbidity index, initial physicians other than<br />

family practice doctors, and referral to pulmonologists were associated with<br />

higher likelihood <strong>of</strong> referral to oncologists (P�0.01 for all factors).<br />

Compared to those who saw only an oncologist, those who saw only a<br />

surgeon and/or a radiation specialist were less likely to receive GBTs<br />

(OR�0.3; 95%CI�0.3-0.4). Among patients who had no referrals or who<br />

saw specialties other than oncology, 14% received GBTs. Conclusions:<br />

Seeing an oncologist is a critical step in the standard treatment <strong>of</strong><br />

advanced NSCLC. Yet, race, income, and the type <strong>of</strong> initial physician may<br />

constitute barriers <strong>of</strong> access to oncologists, which can result in substandard<br />

care.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


384s Health Services Research<br />

6008 Oral Abstract Session, Sat, 3:00 PM-6:00 PM<br />

Oncologists’ and primary care providers’ awareness <strong>of</strong> late effects <strong>of</strong> cancer<br />

treatment: Implications for survivorship care. Presenting Author: Larissa<br />

Nekhlyudov, Harvard Medical School and Harvard Vanguard Medical<br />

Associates, Boston, MA<br />

Background: There is a growing population <strong>of</strong> cancer survivors, many <strong>of</strong><br />

whom may experience late or long-term effects (LEs) <strong>of</strong> treatment. The goal<br />

<strong>of</strong> our study was to describe and compare primary care providers’ (PCP) and<br />

oncologists’ awareness <strong>of</strong> LEs. Methods: The Survey <strong>of</strong> Physician Attitudes<br />

Regarding the Care <strong>of</strong> Cancer Survivors was completed by a nationally<br />

representative sample <strong>of</strong> 1,072 PCPs and 1,130 oncologists (cooperation<br />

rate 65%). Respondents were asked to report for each <strong>of</strong> four standard<br />

chemotherapy drugs used to treat breast or colorectal cancer the five LEs<br />

they had observed and/or had seen reported in the literature. We described<br />

and compared the physicians’ responses and, using separate multinomial<br />

logistic regression models, determined predictors <strong>of</strong> their ability to identify<br />

the main LEs for all agents, adjusting for physician demographics and<br />

practice characteristics. Results: Almost all (95.3%) oncologists identified<br />

cardiac dysfunction as a LE <strong>of</strong> doxorubicin (Adriamycin), and peripheral<br />

neuropathy as LEs <strong>of</strong> both taxol (97.3%) and oxaliplatin (96.6%); while<br />

these LEs were identified by only 55.1%, 21.8% and 21.8% <strong>of</strong> PCPs,<br />

respectively. Most oncologists identified premature menopause (71.4%)<br />

and secondary malignancies (62.0%) as LEs <strong>of</strong> cytoxan, compared with<br />

only 14.8% and 17.2% <strong>of</strong> PCPs. Main LEs for all four chemotherapy drugs<br />

were identified by 65% (n�741) <strong>of</strong> oncologists and only 6% (n�60) <strong>of</strong><br />

PCPs. In adjusted models, oncologists were more likely to identify the main<br />

LEs for all chemotherapy drugs if they spent 51-90% (OR 1.87, 95% CI<br />

1.21-2.88) or �90% (OR 1.82, 95% CI 1.08-3.08) <strong>of</strong> their time on<br />

patient care, and less likely if they were not board certified (OR 0.58, 95%<br />

CI 0.37-0.89). African <strong>American</strong> PCPs were less likely to identify the main<br />

LEs for all chemotherapy drugs (OR 0.19, 95% CI 0.08-0.45). Conclusions:<br />

Oncologists <strong>of</strong>ten identified the main LEs <strong>of</strong> cancer drugs while PCPs did<br />

not. While not surprising, these findings emphasize that in the transition <strong>of</strong><br />

patients from oncology to primary care settings, PCPs should be informed<br />

about the LEs <strong>of</strong> cancer treatment so that they may be better prepared to<br />

recognize and address these among survivors in their post-treatment care.<br />

6010 <strong>Clinical</strong> Science Symposium, Sun, 8:00 AM-9:30 AM<br />

Do insurance status and neighborhood characteristics explain why minorities<br />

underutilize NCI cancer centers? Presenting Author: Lyen C. Huang,<br />

Department <strong>of</strong> Surgery, Stanford University, Stanford, CA<br />

Background: National Cancer-Institute (NCI) designated cancer centers<br />

provide some <strong>of</strong> the highest quality cancer care in the US, in part due to the<br />

availability <strong>of</strong> cutting edge technologies and access to cancer clinical trials.<br />

Racial/ethnic minorities suffer from persistent disparities in cancer outcomes,<br />

and these groups are typically under-represented in clinical trials.<br />

This may be due in part to under-utilization <strong>of</strong> NCI centers by these groups.<br />

Methods: A unique dataset linking the California Cancer Registry with<br />

California patient discharge abstracts was used to identify patients undergoing<br />

resection for a primary diagnosis <strong>of</strong> colorectal cancer (CRC) (1996-<br />

2006). Travel distance to treatment hospital was determined using GIS<br />

s<strong>of</strong>tware. Chi-square analysis correlated patient demographics, clinical<br />

characteristics, insurance status, and neighborhood socioeconomics with<br />

NCI center use. Multivariable regression models were constructed to<br />

predict the likelihood <strong>of</strong> using an NCI center. Results: 95,994 CRC patients<br />

were identified. Median travel distance for care was �5 miles. Only 12,659<br />

(13%) lived within a 5 mile radius <strong>of</strong> an NCI center; and <strong>of</strong> those, fewer<br />

than 10% used the center for CRC care (n�1130). Black (OR 0.83 95%CI<br />

0.72-0.95) and Hispanic (OR 0.72 95%CI 0.65-0.81) patients were less<br />

likely than white patients to use NCI centers. Neighborhood socioeconomics,<br />

but not insurance status, were significantly correlated with NCI<br />

under-utilization. Asian populations were more likely to use NCI centers<br />

than white patients (OR 1.40 95%CI 1.28-1.54). Conclusions: Black and<br />

Hispanic patients are less likely to use nearby NCI hospitals for CRC care.<br />

Outreach efforts in communities with low socioeconomic status and<br />

educational attainment may increase use <strong>of</strong> NCI centers, improve CRC<br />

outcomes, and increase minority enrollment in clinical trials.<br />

CRA6009 <strong>Clinical</strong> Science Symposium, Sun, 8:00 AM-9:30 AM<br />

Patterns <strong>of</strong> decision making about cancer clinical trial participation among<br />

the online cancer treatment community: A collaboration between SWOG<br />

and NexCura. Presenting Author: Joseph M. Unger, SWOG Statistical<br />

Center, Seattle, WA<br />

The full, final text <strong>of</strong> this abstract will be available at<br />

abstract.asco.org at 12:01 AM (EDT) on Sunday, June 3,<br />

2012, and in the <strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong> online<br />

supplement to the June 20, 2012, issue <strong>of</strong> Journal <strong>of</strong><br />

<strong>Clinical</strong> Oncology. Onsite at the <strong>Meeting</strong>, this abstract will<br />

be printed in the Sunday edition <strong>of</strong> ASCO Daily News<br />

6011 <strong>Clinical</strong> Science Symposium, Sun, 8:00 AM-9:30 AM<br />

Insurance status and cancer disparities in adolescents and young adults<br />

age 15 to 39: National Cancer Data Base, 1998-2003. Presenting Author:<br />

Anthony Robbins, <strong>American</strong> Cancer <strong>Society</strong>, Atlanta, GA<br />

Background: Since the mid-1970s there has been little progress in<br />

improving cancer survival for adolescents and young adults (AYAs, defined<br />

by the National Cancer Institute as individuals 15 to 39 years <strong>of</strong> age), in<br />

contrast to the substantial improvements for children and older adults. The<br />

association between insurance status and cancer disparities (stage at<br />

diagnosis, survival, and treatment) in AYA patients has not been examined<br />

in a national sample. Methods: The National Cancer Data Base, a national<br />

hospital-based cancer registry, was used to examine insurance status and<br />

cancer disparities in 177,359 AYA cancer patients. Cases were diagnosed<br />

during 1998�2003 and followed for vital status through 2008. We<br />

examined the association between insurance status and stage at diagnosis,<br />

stage-specific survival, and receipt <strong>of</strong> the most frequently used stagespecific<br />

treatments for common AYA sites (thyroid, female breast, non-<br />

Hodgkin lymphoma, and acute myeloid leukemia). Results: Insurance<br />

status was strongly related to cancer disparities in AYA patients. For<br />

example, compared to patients with private insurance, uninsured patients<br />

were 1.71 times more likely to be diagnosed at stage IV (95% confidence<br />

interval [CI], 1.63�1.79), had higher risk <strong>of</strong> death within every stage<br />

[stage I, hazard ratio (HR) (95% CI), 2.43 (2.13�2.76); stage II, 1.87<br />

(1.70�2.06); stage III, 1.55 (1.43�1.68); stage IV, 1.39 (1.31�1.48);<br />

and unstaged (leukemias, CNS tumors, etc.), 1.67 (1.58�1.76), and for<br />

the four sites listed above, were less likely to receive the most frequently<br />

used stage-specific treatments. Insurance status remained a strong independent<br />

predictor for each <strong>of</strong> these outcomes in multivariate models adjusting<br />

for patient, hospital, and tumor factors. Conclusions: In a large national<br />

sample, insurance status was a strong independent predictor <strong>of</strong> cancer<br />

disparities in AYA patients. The Affordable Care Act <strong>of</strong> 2010 should<br />

facilitate the acquisition <strong>of</strong> adequate health insurance by AYA, who are the<br />

most under- and uninsured age group in the US population, and should<br />

contribute to remediation <strong>of</strong> such disparities.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


6012 <strong>Clinical</strong> Science Symposium, Sun, 8:00 AM-9:30 AM<br />

Racial differences in the impact <strong>of</strong> financial hardship on the intensity <strong>of</strong><br />

end-<strong>of</strong>-life cancer care. Presenting Author: Reginald D. Tucker-Seeley,<br />

Dana-Farber Cancer Institute, Boston, MA<br />

Background: Research suggests that Black cancer patients have higher<br />

end-<strong>of</strong>-life (EOL) medical costs than White patients; and that Black,<br />

compared with White, families are more likely to use all or most <strong>of</strong> their<br />

savings to pay for EOL care. Although Black cancer patients receive more<br />

intense EOL care than Whites, research has yet to determine the effect <strong>of</strong><br />

financial hardship on receipt <strong>of</strong> intensive EOL care, and whether the effect<br />

varies by the patient’s race. Methods: Coping with Cancer (CwC) is a<br />

longitudinal, multi-site cohort study <strong>of</strong> advanced cancer patients and their<br />

informal caregivers recruited from September 2002-February 2008. CwC<br />

was designed to investigate Black/White differences in EOL care. The<br />

purpose <strong>of</strong> this analysis was to determine the association between baseline<br />

financial hardship and receipt <strong>of</strong> intensive EOL care in the last week <strong>of</strong> life<br />

(CwC deceased cohort N�342), and to identify racial differences in this<br />

association. Financial hardship was defined as whether the household had<br />

to use all or most <strong>of</strong> their savings due to the family member’s illness<br />

(response �yes/no). Intensive EOL care was defined as the receipt <strong>of</strong><br />

ventilation or resuscitation in the last week <strong>of</strong> life assessed by medical<br />

record review and patient’s caregiver. Results: Patients reporting financial<br />

hardship had higher odds <strong>of</strong> receiving intensive EOL care (OR � 2.83, CI:<br />

1.33, 6.05). After adjusting for socio-demographic characteristics the<br />

significant association remained (OR � 2.55, CI: 1.13, 5.81). Racestratified<br />

fully adjusted models revealed no statistically significant association<br />

between financial hardship and intensive EOL care for Whites;<br />

however, for Blacks, those reporting financial hardship had over five times<br />

higher odds <strong>of</strong> receiving intensive EOL care (OR � 5.21, CI: 1.51, 17.99)<br />

compared to those not reporting financial hardship. Conclusions: Financial<br />

hardship was associated with greater likelihood <strong>of</strong> receiving intensive EOL<br />

care. The intensity <strong>of</strong> EOL care received by White patients was insensitive<br />

to financial hardship; in contrast Black patients reporting depletion <strong>of</strong> life<br />

savings for cancer care were much more likely to die receiving intensive<br />

EOL care than their non-financially strained counterparts.<br />

6014 Poster Discussion Session (Board #2), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Adjuvant chemotherapy (AC) initiation and early discontinuation in elderly<br />

patients (EPs) with stage III colon cancer (CC). Presenting Author: Jenny J.<br />

Ko, British Columbia Cancer Agency, Vancouver, BC, Canada<br />

Background: Research suggests that EPs with cancer are commonly<br />

undertreated, but the precise reasons for this observation are unclear. Our<br />

aims were to 1) evaluate the impact <strong>of</strong> advanced age on AC use (none vs<br />

capecitabine vs FOLFOX) for stage III CC, 2) determine the specific reasons<br />

for selecting and discontinuing a particular regimen, and 3) examine if the<br />

effect <strong>of</strong> AC on outcomes is modified by age. Methods: Patients diagnosed<br />

with stage III CC from 2006 to 2008 and referred to any 1 <strong>of</strong> 5 cancer<br />

centers in British Columbia, Canada were identified. Descriptive statistics<br />

were used to summarize treatment patterns in young patients (YPs) aged<br />

�70 vs EPs aged �/�70 years. Logistic regression was used to evaluate<br />

the association between AC and cancer-specific survival (CSS) in YPs and<br />

EPs. Results: We identified 810 patients: 51% men, 52% YPs and 48%<br />

EPs, and 74% received AC in the entire cohort. When compared to YPs,<br />

EPs had worse ECOG and more comorbidities (both p�0.01). EPs were less<br />

likely than YPs to receive AC (57 vs 91%, p�0.01). Frequent reasons for no<br />

treatment included age, comorbidities and perceived minimal benefit from<br />

AC. Among those treated with AC, EPs were less likely to receive FOLFOX<br />

(32 vs 74%, p�0.01) in favor <strong>of</strong> capecitabine due to patient preference,<br />

age and comorbidities. Once started on AC, EPs had similar rates <strong>of</strong> early<br />

treatment discontinuation as YPs (70 vs 62%, p�0.08). Reasons for early<br />

discontinuation were comparable between EPs and YPs (Table). Receipt <strong>of</strong><br />

either FOLFOX or capecitabine was correlated with improved CSS, compared<br />

to surgery alone. Age did not modify CSS, irrespective <strong>of</strong> AC choice<br />

(interaction p for capecitabine and age�0.26; interaction p for FOLFOX<br />

and age�0.40). Conclusions: EPs with stage III CC frequently received<br />

either no adjuvant treatment or capecitabine monotherapy due to advanced<br />

age and comorbidities. The treatment effect <strong>of</strong> AC on CSS is similar across<br />

age groups, with comparable side effects and rates <strong>of</strong> discontinuation<br />

between EPs and YPs. AC should not be withheld from CC patients based<br />

on advanced age alone.<br />

Reasons for early discontinuation<br />

Side effects p Progressive disease p Patient choice p<br />

YPs 59% 0.99 21% 0.39 6% 0.27<br />

EPs 58% 16% 10%<br />

Health Services Research<br />

385s<br />

6013 Poster Discussion Session (Board #1), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Toxicity <strong>of</strong> chemotherapy dosing using actual body weight in obese versus<br />

normal-weight patients: A systematic review and meta-analysis. Presenting<br />

Author: Kathryn Cunningham Hourdequin, Dartmouth Hitchcock Medical<br />

Center, Lebanon, NH<br />

Background: Because weight-based chemotherapy calculations can be very<br />

large in obese patients, oncologists <strong>of</strong>ten empirically reduce doses due to<br />

fear <strong>of</strong> excess toxicity. The resulting underdosing may negatively impact<br />

survival. We performed a systematic review and meta-analysis to determine<br />

whether, among adults receiving chemotherapy dosed by actual body<br />

weight (ABW), obese patients experience differing toxicity or survival<br />

compared to normal-weight patients. Methods: We searched MEDLINE,<br />

Cochrane Library, Web <strong>of</strong> Science, and <strong>Clinical</strong>Trials.gov through October<br />

2011 and reviewed reference lists. We included studies that compared<br />

outcomes <strong>of</strong> obese versus normal-weight adults receiving chemotherapy<br />

dosed according to ABW (�/- 5% variability). Studies followed subjects for<br />

at least one cycle <strong>of</strong> chemotherapy and reported at least one pre-specified<br />

outcome. Two authors independently abstracted data from eligible studies.<br />

We used random effects models to pool odds ratios (OR) for hematologic<br />

and non-hematologic toxicities. We summarized survival qualitatively.<br />

Results: Of 3,921 studies, five met inclusion criteria, for a total <strong>of</strong> 6,877<br />

subjects. Based on three studies, Grade 3/4 hematologic toxicity occurred<br />

less <strong>of</strong>ten in obese patients than normal-weight patients (OR 0.68, 95% CI<br />

0.51-0.89, I2�29%). A fourth study comparing leukocyte nadirs had<br />

variable results depending on the regimen, dosing, and patient comorbidities.<br />

Based on two studies, Grade 3/4 non-hematologic toxicity<br />

occurred less <strong>of</strong>ten in obese patients than normal-weight patients (OR<br />

0.74, 95% CI 0.63-0.87, I2�0%). A third study found rates <strong>of</strong> infection<br />

did not significantly differ. Three <strong>of</strong> four studies reported reduced overall<br />

survival in obese patients (statistical significance not reported). Conclusions:<br />

Contrary to common belief, obese patients receiving chemotherapy based<br />

on ABW appear to have lower rates <strong>of</strong> both hematologic and nonhematologic<br />

toxicities compared to normal-weight patients. These results<br />

do not support the practice <strong>of</strong> empiric dose reduction in obese patients.<br />

Further research should explore etiologies for the reduced survival in this<br />

group.<br />

6015 Poster Discussion Session (Board #3), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Comorbidity, chemotherapy toxicity, and outcomes among older women<br />

receiving adjuvant chemotherapy for breast cancer (BC). Presenting Author:<br />

Heidi D. Klepin, Wake Forest School <strong>of</strong> Medicine, Winston-Salem, NC<br />

Background: Comorbidity increases with age. We evaluated how comorbidity<br />

was associated with toxicity and clinical outcomes among older women<br />

with BC who received adjuvant chemotherapy. Methods: Cancer and<br />

Leukemia Group B (CALGB 49907) enrolled 633 women �65 years with<br />

early stage BC and randomized them to standard adjuvant chemotherapy<br />

(AC or CMF) or capecitabine (N Eng J Med 360:2055, 2009). 329 women<br />

on the Quality <strong>of</strong> Life companion study completed a pre-treatment health<br />

survey (Older <strong>American</strong> Resources and Services Questionnaire, physical<br />

health subscale). The survey evaluates 14 conditions and the degree to<br />

which each interferes with daily activities (rated from 1 “not at all” to 3 “a<br />

great deal”). Comorbidity was analyzed as follows: 1) total number 2)<br />

comorbidity burden score (summed conditions multiplied by interference<br />

rating); and 3) individual diseases. Primary outcomes were: 1) grade 3-5<br />

toxicity (incident and cumulative); 2) time to relapse (TTR), and 3) overall<br />

survival (OS). Contingency table methods were used to evaluate the<br />

association between comorbidity and toxicity. Cox proportional hazards<br />

models were used to evaluate TTR and survival outcomes. Results: Among<br />

329 women [median age 71 years (range 65-89), 86% white, 98% ECOG<br />

performance score 0-1, 75% stage 1-2] the median number <strong>of</strong> comorbidities<br />

was 2 (0-10), median comorbidity burden score was 3 (0-25), and<br />

most common conditions were arthritis (58%) and hypertension (54%).<br />

Few patients had diabetes (17%), heart disease (16%), and pulmonary<br />

disease (9%). No comorbidity measure was associated with toxicity or TTR.<br />

With a median follow-up <strong>of</strong> 5.4 years, increasing comorbidity was associated<br />

with shorter OS. For each additional comorbid condition the hazard <strong>of</strong><br />

death increased by 18% (HR 1.18 [95% CI; 1.06-1.33]) after adjusting for<br />

age, tumor size, treatment, node status and receptor status. Comorbidity<br />

burden score was similarly associated with OS (HR 1.08 [95% CI;<br />

1.03-1.14]), while no specific condition was independently associated.<br />

Conclusions: Among older women with good functional status, comorbidity<br />

was not associated with toxicity or BC relapse. However, comorbidity<br />

burden was associated with shorter OS.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


386s Health Services Research<br />

6016 Poster Discussion Session (Board #4), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Refused versus recommended adjuvant chemotherapy in the United States: A<br />

reason for concern. Presenting Author: Elizabeth Butzer Habermann, Department<br />

<strong>of</strong> Surgery, University <strong>of</strong> Minnesota, Minneapolis, MN<br />

Background: We designed this study to identify factors driving treatment<br />

recommendations and refusal in surgically treated patients with gastrointestinal<br />

(GI) cancers eligible for adjuvant chemotherapy. Methods: Using the 2001-2008<br />

California Cancer Registry (CCR), we constructed a cohort <strong>of</strong> 22,469 patients<br />

surgically treated for three GI cancers and eligible for adjuvant chemotherapy<br />

(stage III colon cancer in patients � 80 years, Ib-IVM0 gastric cancer, or<br />

nonmetastatic resected pancreatic adenocarcinoma). Information on chemotherapy<br />

receipt, recommendation, and refusal was utilized to construct our<br />

outcome measures. Multivariate logistic regression models identified predictors<br />

<strong>of</strong> recommendation and refusal <strong>of</strong> adjuvant chemotherapy across three GI cancer<br />

sites, adjusting for potential confounders. Results: Of those eligible for adjuvant<br />

therapy, only 61.4% were recommended treatment and 54.3% received it.<br />

Refusal rate was 3.5%. Persons who were older, black, publicly insured or<br />

uninsured, or with gastric cancers were less likely to have adjuvant chemotherapy<br />

recommended. Yet, older, Medicaid-insured, and those treated for<br />

gastric cancer were more likely to refuse adjuvant therapies. Women were as<br />

likely as men to be recommended adjuvant therapies but more likely to refuse<br />

them (Table). Conclusions: Many patients eligible for adjuvant chemotherapy<br />

after GI cancer surgery have not been so advised. Striking variations exist<br />

between recipients <strong>of</strong> treatment recommendations versus those who refuse<br />

them. These results demonstrate a wide implementation gap between the<br />

recommendations arising from cancer clinical trials versus actual clinical<br />

practice.<br />

Age<br />

75-79 vs. 18-64<br />

Race<br />

Black vs. white<br />

Gender<br />

Female vs. male<br />

Insurance status<br />

Medicaid vs. private<br />

Medicare vs. private<br />

Disease site<br />

Colon vs. pancreas<br />

Gastric vs. pancreas<br />

Adjuvant chemotherapy<br />

recommended vs. other<br />

odds ratio (95% CI)<br />

0.38<br />

(0.34-0.41)<br />

0.80<br />

(0.72-0.89)<br />

1.06<br />

(1.00-1.12)<br />

0.85<br />

(0.76-0.96)<br />

0.86<br />

(0.80-0.92)<br />

1.56<br />

(1.43-1.70)<br />

0.68<br />

(0.62-0.75)<br />

Adjuvant chemotherapy<br />

refused vs. received<br />

odds ratio (95% CI)<br />

5.93<br />

(4.46-7.88)<br />

1.18<br />

(0.82-1.70)<br />

1.36<br />

(1.12-1.66)<br />

1.65<br />

(1.13-2.42)<br />

0.88<br />

(0.70-1.10)<br />

1.17<br />

(0.84-1.62)<br />

1.82<br />

(1.28-2.59)<br />

6018 Poster Discussion Session (Board #6), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Use <strong>of</strong> palliative chemotherapy and targeted agents in elderly patients with<br />

metastatic colorectal cancer (mCRC). Presenting Author: Matthew Chan,<br />

British Columbia Cancer Agency, Vancouver, BC, Canada<br />

Background: Elderly patients are increasingly diagnosed with advanced<br />

cancers, but they are consistently underrepresented in clinical trials, which<br />

may lead to undertreatment. Our aims were to 1) evaluate the impact <strong>of</strong><br />

advanced age on patterns <strong>of</strong> first-line chemotherapy and bevacizumab use<br />

in mCRC, 2) examine the reasons for treatment choices and 3) compare<br />

adverse events and treatment discontinuations in elderly vs young patients.<br />

Methods: A random sample <strong>of</strong> mCRC patients diagnosed from 2006 to<br />

2007 and referred to any 1 <strong>of</strong> 5 regional cancer centers in British<br />

Columbia, Canada was reviewed. Summary statistics were used to describe<br />

treatment patterns between the elderly (�/�70 years) and young (�70<br />

years). Cox regression was used to determine the effect <strong>of</strong> systemic therapy<br />

on overall survival, controlling for age and confounders. Results: We<br />

identified 800 patients: 43% elderly and 57% young; 56% men; and 26 /<br />

36/38%ECOG0/1/2�, respectively. Fewer elderly patients were given<br />

chemotherapy (52% vs 79%, p�0.001). Among those treated, most<br />

common first-line palliative regimens for elderly vs young included:<br />

capecitabine (50 vs 15%), FOLFIRI (26 vs 38%), and FOLFOX (15 vs<br />

37%) (all p�0.001). Those aged �/�70 were also less likely to receive<br />

bevacizumab in their regimens (22 vs 50%, p�0.001). The most frequent<br />

reasons for no systemic therapy were similar between age groups: patient<br />

choice (31 vs 28%), poor ECOG (16 vs 17%), and significant co-morbidity<br />

(11 vs 13%). Risk <strong>of</strong> chemotherapy (p�0.30) and bevacizumab (p�0.39)<br />

adverse events were comparable between elderly and young as were rates <strong>of</strong><br />

early chemotherapy (p�0.07) and bevacizumab (p�0.79) discontinuation.<br />

Receipt <strong>of</strong> systemic therapy �/- bevacizumab was associated with<br />

improved survival from mCRC (HR for death 0.50, 95% CI 0.31-0.62,<br />

p�0.001), regardless <strong>of</strong> advanced age (p interaction for age and treatment<br />

� 0.33). Conclusions: Elderly patients with mCRC are more likely to receive<br />

no chemotherapy, capecitabine monotherapy, or a regimen without bevacizumab.<br />

However, in carefully selected elderly patients, adverse events,<br />

treatment discontinuations, and overall survival benefit from treatment<br />

appear similar to those observed for younger patients.<br />

6017 Poster Discussion Session (Board #5), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Endocrine therapy use among Medicaid-insured breast cancer survivors<br />

with hormone receptor-positive tumors. Presenting Author: Racquel Elizabeth<br />

Kohler, University <strong>of</strong> North Carolina at Chapel Hill, Chapel Hill, NC<br />

Background: Estrogen receptor positive (ER�) and progesterone receptor<br />

positive (PR�) cancers account for the majority <strong>of</strong> breast cancer diagnoses<br />

and deaths. Among women with ER� or PR� breast cancers, endocrine<br />

therapy (ET) is the cornerstone <strong>of</strong> adjuvant therapy and reduces 5-year risk<br />

<strong>of</strong> recurrence by as much as 40%. Observational studies in Medicare and<br />

privately-insured populations suggest that ET is underutilized. We sought<br />

to characterize ET use in a low-income Medicaid-insured population in<br />

North Carolina. Methods: We used Medicaid claims data matched to NC<br />

Central Cancer Registry records for women ages 18-64 diagnosed with in<br />

situ, stage I or II breast cancer from 2003-2007. We excluded dual<br />

eligibles and included only cases enrolled in Medicaid for at least 12 <strong>of</strong> the<br />

15 months following the index diagnosis. We defined our outcome as<br />

receipt <strong>of</strong> any ET medication (tamoxifen, letrozole, exemestane, or anastroxole)<br />

in prescription claims during the 15-month period post-diagnosis,<br />

among women with ER� or PR� disease. In multivariate logistic regressions,<br />

independent variables included age, race, tumor characteristics,<br />

receipt <strong>of</strong> other breast cancer treatments, co-morbidity, rural/urban residence,<br />

reason for Medicaid eligibility, involvement in the Breast and<br />

Cervical Cancer Control Program (BCCCP), patient-centered medical home<br />

enrollment, and diagnosis year. Results: Of the 269 women who met<br />

inclusion/exclusion criteria and were ER� or PR�, only 45% filled a<br />

prescription for ET during the study period. In multivariate analyses, being<br />

involved in the CDC-affiliated BCCCP was significantly associated with<br />

higher likelihood <strong>of</strong> receipt <strong>of</strong> guideline-recommended endocrine therapy<br />

(Marginal Effect: 0.299, p�0.01), but other independent variables were<br />

not significantly correlated with receipt <strong>of</strong> ET. Conclusions: Results suggest<br />

that ET is substantially underutilized in this low-income, vulnerable<br />

population and that intervention efforts to improve ET use may be<br />

important. Qualitative research is needed to understand the more nuanced,<br />

behavioral reasons for ET underuse, which may be related to symptom<br />

burden, cost, and patient-provider communication.<br />

6019 Poster Discussion Session (Board #7), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

An assessment <strong>of</strong> the collective efforts <strong>of</strong> clinical trials to provide<br />

evidence-based practice guidelines in cancer care. Presenting Author:<br />

Shane Lloyd, Yale University School <strong>of</strong> Medicine, New Haven, CT<br />

Background: It is unclear how many <strong>of</strong> the recommendations in the National<br />

Comprehensive Cancer Network (NCCN) guidelines are based Category 1<br />

(C1: “high level evidence”), or whether current cancer research efforts are<br />

focusing on the knowledge gap between guidelines and supporting evidence.<br />

We defined the knowledge gap as the percent <strong>of</strong> NCCN recommendations<br />

based on lower levels <strong>of</strong> evidence, and assessed the relation<br />

between the knowledge gap and cancer research efforts in terms <strong>of</strong> clinical<br />

trial enrollment. Methods: We examined the active, phase 3 randomized<br />

controlled trials (RCTs) listed on clinicaltrials.gov for the 17 most prevalent<br />

solid tumors. We used the NCCN guidelines to tally the percentage <strong>of</strong> C1<br />

recommendations. We used the Pearson coefficient and linear regression to<br />

examine whether enrollment in active phase 3 RCTs is associated with 1)<br />

the knowledge gap and 2) measures <strong>of</strong> burden to society including<br />

prevalence, incidence, person years life lost (PYLL) and disability adjusted<br />

life years (DALY). Results: We identified 834 treatment recommendations<br />

for the 17 included cancer types. Overall, 15% <strong>of</strong> the NCCN recommendations<br />

were rated as C1, varying from 40% for hepatobiliary to 0% for<br />

endometrial cancer. There was no association between RCT enrollment and<br />

the proportion <strong>of</strong> C1 recommendations in univariate or multivariate<br />

analyses. RCT enrollment positively correlated with measures <strong>of</strong> burden to<br />

society including prevalence (p-value � 0.001), incidence (p-value 0.001),<br />

and DALY (p-value 0.038). Breast and prostate cancers have a large<br />

amount <strong>of</strong> RCT enrollment per PYLL and a relatively small knowledge gap.<br />

Melanoma, ovarian, and endometrial cancers have a moderate amount <strong>of</strong><br />

RCT enrollment per PYLL and a relatively large knowledge gap. Lung,<br />

esophagus, bladder and CNS cancers have a low amount <strong>of</strong> RCT enrollment<br />

per PYLL and a moderate to large knowledge gap. Conclusions: RCT<br />

enrollment is not correlated to the knowledge gap. Current planned<br />

enrollment in RCTs is partially predicted by burden to society, with the<br />

strongest correlation to incidence and prevalence. Most recommendations<br />

in the NCCN guidelines are not based on C1 evidence.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


6020 Poster Discussion Session (Board #8), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

A critical evaluation <strong>of</strong> oncology clinical practice guidelines. Presenting<br />

Author: Bradley Norman Reames, University <strong>of</strong> Michigan, Ann Arbor, MI<br />

Background: <strong>Clinical</strong> Practice Guidelines (CPGs) play an essential role in<br />

cancer care today, but there are significant concerns regarding their<br />

content and reliability. In 2011, the Institute <strong>of</strong> Medicine (IOM) report<br />

“<strong>Clinical</strong> Practice Guidelines We Can Trust” created standards for developing<br />

trustworthy CPGs. Using these standards as a benchmark, we sought to<br />

evaluate recent oncology guidelines. Methods: CPGs and consensus statements<br />

addressing the screening, evaluation or management <strong>of</strong> the four<br />

leading causes <strong>of</strong> cancer-related mortality in the US (non-small cell lung,<br />

breast, prostate and colorectal cancers) published between January 2005<br />

and December 2010 were identified using MEDLINE. A standardized<br />

scoring system based on the eight standards set forth by the IOM was<br />

devised, and the methodology, content and disclosure policies <strong>of</strong> CPGs<br />

were critically evaluated by four independent reviewers. All CPGs were<br />

given two scores; points were awarded out <strong>of</strong> a possible 8 major criteria and<br />

20 sub-criteria. Results: We identified 168 CPGs for inclusion in the study;<br />

45% were from US groups. None <strong>of</strong> the CPGs fully met all the IOM<br />

standards. On average, CPGs only met 2.8 <strong>of</strong> 8 standards set forth by the<br />

IOM (mean 2.8 points out <strong>of</strong> 8, SD 1.7; 8.3 out <strong>of</strong> 20, SD 4.3). Less than<br />

half <strong>of</strong> CPGs were based on a systematic review. Only half <strong>of</strong> CPG panels<br />

addressed conflicts <strong>of</strong> interest. Overall, the CPGs were most consistent with<br />

IOM standards for transparency regarding the development process,<br />

articulation <strong>of</strong> recommendations, and use <strong>of</strong> external review. Most did not<br />

comply with standards for inclusion <strong>of</strong> patient and public involvement in<br />

the development or review process, nor did they specify their process for<br />

updating. CPGs from the US had higher overall scores than CPGs from<br />

international groups. CPGs addressing non-small cell lung cancer had<br />

higher overall scores (mean 3.9) than those for other cancers. Conclusions:<br />

The vast majority <strong>of</strong> oncology CPGs fails to meet the IOM standards for<br />

trustworthy guidelines. Notably, most CPGs are not based on systematic<br />

reviews, lack full disclosure, and do not include all relevant stakeholders in<br />

the guideline process. This highlights the need for improved CPG development<br />

processes.<br />

6022 Poster Discussion Session (Board #10), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Trade-<strong>of</strong>fs associated with axillary lymph node dissection: Implications <strong>of</strong><br />

the eligibility versus enrollment in ACOSOG Z0011. Presenting Author:<br />

Monica Shalini Krishnan, Harvard Radiation Oncology Program, Boston,<br />

MA<br />

Background: Results from ACOSOG Z0011 suggest axillary lymph node<br />

dissection (ALND) may not be necessary for patients following positive<br />

sentinel lymph node biopsy (SLNB). Concerns have been raised regarding<br />

generalizability <strong>of</strong> this trial, given the low-risk patient population. It is<br />

uncertain whether a subgroup who would have been eligible for ACOSOG<br />

Z0011 but were not adequately represented in the study may still benefit<br />

from ALND. Methods: We constructed a decision analysis using a Monte<br />

Carlo model to simulate axillary recurrence risk, lymphedema, and quality<br />

<strong>of</strong> life <strong>of</strong> women aged 45, 55 and 75 y/o with Stage II cancers following<br />

breast conserving surgery (BCS) with positive SLNB who were then treated<br />

with ALND and whole-breast radiation (BRT) or BRT alone. Women were<br />

divided into two risk groups based on the Memorial Sloan-Kettering Cancer<br />

Center non-sentinel lymph node (NSLN) nomogram: those with risk <strong>of</strong><br />

residual nodal involvement <strong>of</strong> 30-60% (high risk); and those with risk less<br />

than 30% (low risk, similar to the Z0011 patients). Probabilities and<br />

utilities for health states were derived from prior studies. Results: BRT alone<br />

resulted in improved quality-adjusted life expectancy (QALE) in the<br />

low-risk group, while ALND with BRT resulted in improved QALE in the<br />

high-risk group. Overall survival (OS) was similar at 5 years with both<br />

treatment strategies in both groups but was superior with ALND at 20 years<br />

in the high risk group (Table). Differences in outcomes decreased with<br />

increasing age. In the low-risk group, sensitivity analysis showed BRT alone<br />

is preferred unless the axillary recurrence risk with BRT is greater than<br />

1.6% or the lymphedema risk with ALND is less than 10%. In the high-risk<br />

group, ALND with BRT is the preferred strategy unless the axillary<br />

recurrence risk with BRT is less than 2.3%. Conclusions: Patients who<br />

would have been eligible for ACOSOG Z0011 but are at higher risk <strong>of</strong> having<br />

residual nodal disease following BCS and positive SLNB may benefit from<br />

ALND plus BRT rather than BRT alone.<br />

Overall survival and QALE in 55 y/o women.<br />

5 yr OS 20 yr OS QALE (yrs)<br />

BRT ALND BRT ALND BRT ALND<br />

Low risk (NSLN 0-30%) 88% 88% 47% 47% 15.53 15.46<br />

High risk (NSLN 31-60%) 86% 86% 38% 42% 13.55 14.36<br />

Health Services Research<br />

6021 Poster Discussion Session (Board #9), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Cancer patients’ trade-<strong>of</strong>fs for efficacy, toxicity, and cost. Presenting<br />

Author: Yu-Ning Wong, Fox Chase Cancer Center, Philadelphia, PA<br />

Background: When making treatment decisions, cancer patients (pts) must<br />

make trade-<strong>of</strong>fs between efficacy, toxicity (tox) and cost. However, little is<br />

known about how individual characteristics influence these decisions,<br />

particularly as many face high out <strong>of</strong> pocket costs. Methods: We presented<br />

cancer pts with hypothetical scenarios that asked them to choose between<br />

2 treatments <strong>of</strong> varying levels <strong>of</strong> efficacy, tox and cost. Each scenario<br />

included 9 choice pairs. Pts were given 2 <strong>of</strong> 3 scenarios described in the<br />

Table. Tox was also varied. Demographics, cost concerns and numeracy<br />

were assessed. Within each scenario, we used latent class methods to<br />

distinguish pt groups with discrete preferences. We then used regressions<br />

with group membership probabilities as covariates to identify associations.<br />

Results: We enrolled 400 pts. Median age was 61 years (range 27-90). 63%<br />

were female. 41% were college educated. 51% had an annual income<br />

�$60K. 25% were enrolled at a community hospital. 98% were insured.<br />

Within each <strong>of</strong> the 3 scenarios, we identified 3 pt classes with preferences<br />

for survival or aversion to high cost or toxicity. Across each <strong>of</strong> the scenarios,<br />

�6% <strong>of</strong> pts in the group averse to high cost chose the costlier treatment.<br />

�92% <strong>of</strong> pts in the group that favored survival chose the highest efficacy<br />

treatment. �65% <strong>of</strong> pts in the group with aversion to tox chose the lower<br />

tox treatment. Within each <strong>of</strong> the scenarios, pts in the group with<br />

preference for survival were more likely to have an income <strong>of</strong> �$60K<br />

(p�.05) and greater numeracy skills (p�.05). In scenarios 2 and 3, pts<br />

with concerns about treatment costs were more likely to be in the class that<br />

was averse to high cost (p�.05 for both). Conclusions: Even in hypothetical<br />

scenarios presented to insured pts, socioeconomic status was predictive <strong>of</strong><br />

treatment choice. Higher income pts may be more likely to focus on survival<br />

when making decisions while those with greater cost concerns may be more<br />

likely to avoid costly treatment, regardless <strong>of</strong> survival or tox. This raises the<br />

possibility that health plans with greater cost-sharing may have the<br />

unintended consequence <strong>of</strong> increasing disparities in care.<br />

Efficacy Range <strong>of</strong> costs<br />

1 DFS 86% vs 73% $500-$10,000 over 6 m<br />

2 DFS 73% vs 67% $250- $6,000 over 6 m<br />

3 2 yr survival 23% vs 15% $60 - $800 every 3 wk<br />

6023 Poster Discussion Session (Board #11), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

The involvement <strong>of</strong> partners in breast cancer treatment decision making.<br />

Presenting Author: Sarah T. Hawley, University <strong>of</strong> Michigan, Ann Arbor, MI<br />

Background: Incorporating partners into treatment decision making is an<br />

important element <strong>of</strong> patient-centered care, yet little is known about the<br />

role <strong>of</strong> partners in the decision process. Methods: We surveyed 503 partners<br />

<strong>of</strong> a population-based sample <strong>of</strong> breast cancer survivor 4 years after<br />

diagnosis (RR� 76%, N�382).The outcome was partners’ reports <strong>of</strong><br />

decision regret. Independent variables included decision making process<br />

measures (partners’ reports <strong>of</strong> sufficient treatment information receipt and<br />

sufficient involvement in decision making), race/ethnicity, age, education<br />

and income. Multivariable logistic regression was used to assess associations<br />

between decision regret and race/ethnicity, controlling for other<br />

variables. Results: 49% <strong>of</strong> partners were white, 14% African <strong>American</strong>,<br />

15% more-acculturated Latino, and 18% less-acculturated Latino. One<br />

quarter (26%) <strong>of</strong> partners reported that they received insufficient information<br />

and one third (35%) desired more involvement in decision-making.<br />

Compared to whites, less-acculturated Latino partners more <strong>of</strong>ten reported<br />

that they received insufficient information (41% vs. 18%, p�0.05) and<br />

desired more involvement in decision-making (49% vs. 14%, p�0.001).<br />

Overall 30% <strong>of</strong> partners reported high decision regret. Multivariate analyses<br />

showed factors associated with high decision regret were lessacculturated<br />

Latinos, insufficient information receipt and desire for more<br />

involvement (Table). Conclusions: Most partners <strong>of</strong> breast cancer survivors<br />

reported low decision regret and positively appraised their involvement in<br />

the decision process. Less acculturated Latinos reported more dissatisfaction<br />

with the decision process. Findings suggest the need for culturally<br />

appropriate treatment decision support interventions that include partners.<br />

Factors associated with high decision regret<br />

Race/ethnicity<br />

White<br />

African <strong>American</strong><br />

Latino–more acculturated<br />

Latino–less acculturated<br />

Sufficient information<br />

Yes<br />

No<br />

Sufficient involvement<br />

Yes<br />

No<br />

Controlled for age, education, and income.<br />

387s<br />

OR (95%CI)<br />

N�382<br />

1.00<br />

1.34 (0.58-3.12)<br />

1.82 (0.78-4.27)<br />

2.43 (1.00-5.89)<br />

1.00<br />

2.33 (1.25-4.31)<br />

1.00<br />

1.91 (1.04-3.48)<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


388s Health Services Research<br />

6024 Poster Discussion Session (Board #12), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Improving the quality <strong>of</strong> breast cancer surgical treatment decisions.<br />

Presenting Author: Steven J. Katz, University <strong>of</strong> Michigan Medical School,<br />

Ann Arbor, MI<br />

Background: Ensuring breast cancer treatment decisions are high quality<br />

(i.e., informed and preference-concordant) is a key component <strong>of</strong> patient<br />

centered care. Methods: A web-based decision tool, including an interactive<br />

preference clarification exercise, was developed over a one-year period with<br />

input from health communication experts, clinicians, and women with<br />

breast cancer. Newly diagnosed early stage breast cancer patients from two<br />

cancer centers were recruited and randomized to view the tool before or<br />

after completing a survey. Mean scores for key outcome measures,<br />

including surgical treatment knowledge (4 true/false questions), decision<br />

satisfaction (12 questions each with a 5-point Likert scale from strongly<br />

agree to strongly disagree), and preference-concordant decisions, were<br />

compared between the groups using t-tests. Concordance between preferences<br />

and surgical choices was evaluated using the chi-square test.<br />

Results: 110 subjects were recruited and 105 completed the study. Their<br />

mean age was 57 years, 60% had a college degree or more, and 81% were<br />

white. Those viewing the website first had higher scores on several decision<br />

outcomes than those taking the survey first (Table). Knowledge scores were<br />

also higher among those viewing the website before the survey (3.0 vs.<br />

2.61, p�.23). The risk <strong>of</strong> recurrence was the most important treatment<br />

attribute, followed by retaining the natural breast, in both groups. Concordance<br />

between treatment choice and computer generated treatment was<br />

65% for website first and 61% for survey first groups. Conclusions: A tool<br />

focused on improving knowledge and preference-concordant decisions<br />

produced positive results on breast cancer surgical treatment decision<br />

making. Further work should assess the impact <strong>of</strong> the tool in larger and<br />

more diverse populations.<br />

Website first Survey first<br />

Response 1-5<br />

Decision outcomes<br />

(strongly agree – strongly disagree)<br />

I’m unsure what decision to make 4.12 3.35*<br />

The surgical treatment decision is hard 3.76 2.80<br />

for me<br />

I’m aware <strong>of</strong> the choices I have to treat 1.61 1.83*<br />

my breast cancer<br />

I am satisfied with my surgical treatment 1.44 1.65<br />

decision<br />

I feel the surgical treatment decision<br />

matches my values<br />

*P�0.05, ^P�0.10.<br />

1.32 1.77^<br />

6026 Poster Discussion Session (Board #14), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Is care coordination associated with improved care quality for comorbid<br />

conditions in cancer survivors? Presenting Author: Claire Frances Snyder,<br />

The Johns Hopkins University School <strong>of</strong> Medicine and Sidney Kimmel<br />

Comprehensive Cancer Center, Baltimore, MD<br />

Background: Many cancer survivors have comorbid conditions, adding<br />

complexity to their already complicated care and requiring greater care<br />

coordination. We assessed the role <strong>of</strong> care coordination in comorbid<br />

condition care for cancer survivors. Methods: Using SEER-Medicare, we<br />

examined 7 published indicators <strong>of</strong> quality comorbid condition care in<br />

survivors <strong>of</strong> loco-regional breast, prostate, or colorectal cancer who were<br />

diagnosed in 2004, in fee-for-service Medicare, and survived �3 years.<br />

Comorbid condition care was evaluated during the transition from initial<br />

cancer treatment to survivorship (i.e. days 366-1095 post-diagnosis).<br />

Coordination risk was categorized as Likely, Possible, or Unlikely using an<br />

index developed and tested as part <strong>of</strong> the ACG case-mix adjustment and<br />

predictive modeling tool. The index factors in the number <strong>of</strong> unique<br />

providers, number <strong>of</strong> specialties, the percentage majority source <strong>of</strong> care,<br />

and generalist visits. We tested the hypothesis that lower coordination risk<br />

would be associated with better comorbid condition care using logistic<br />

regression, adjusting for socio-demographics, cancer type, and comorbidity.<br />

Results: The sample included 8661 survivors (53% prostate, 22%<br />

breast, 26% colorectal; mean age 75; 65% male, 85% white). Our<br />

hypothesis was not supported. Compared to patients with Unlikely coordination<br />

issues, patients with Likely coordination issues were more likely to<br />

receive appropriate care on 4 indicators and less likely on 1. Possible<br />

coordination issues were associated with better care on 1 indicator and<br />

worse care on 1 indicator. To explore this finding further, we conducted<br />

post-hoc analyses examining the role <strong>of</strong> each component <strong>of</strong> the coordination<br />

risk index. Having more unique providers was generally associated with<br />

better comorbid condition care, in contrast to the calculation <strong>of</strong> the index<br />

which considers more unique providers a greater risk for coordination<br />

issues. Conclusions: These findings suggest that traditional metrics <strong>of</strong> care<br />

coordination may not be valid for survivors <strong>of</strong> cancer. Understanding the<br />

role <strong>of</strong> care coordination in cancer survivorship care requires development<br />

and application <strong>of</strong> alternative coordination measures.<br />

6025 Poster Discussion Session (Board #13), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Does unblinding <strong>of</strong> treatment assignment impact participant perceptions in<br />

clinical trials? Presenting Author: Ann H. <strong>Part</strong>ridge, Dana-Farber Cancer<br />

Institute, Boston, MA<br />

Background: Blinding patients to treatment regimen is an important<br />

component <strong>of</strong> high quality randomized clinical trials although concern<br />

exists about how receipt <strong>of</strong> a placebo will impact participants’ views. In<br />

ECOG5103, patients were randomized to receive adjuvant chemotherapy<br />

for breast cancer with either placebo (arm A) or bevacizumab (arms B and<br />

C) and treatment assignment was unblinded by 24 weeks. We sought to<br />

determine if unblinding was associated with differential changes in<br />

participants’ views. Methods: The Decision-Making/Quality <strong>of</strong> Life component<br />

(DM-QOL) included all patients enrolling on E5103 between 1/5-<br />

6/8/10. Women were surveyed pretreatment and after unblinding. Wilcoxon<br />

rank sum testing was used to examine differences. Results: 572 patients on<br />

DM-QOL started protocol therapy; 118 on arm A, 454 on arm B/C; the two<br />

groups (arm A vs. B/C) were well balanced. 516 patients participated in<br />

pretreatment, 514 in unblinding survey. Pretreatment, 32% reported<br />

�moderate chance <strong>of</strong> recurrence in 5 years, 27% �moderate chance <strong>of</strong><br />

serious problem from treatment, 99% feeling at least somewhat informed,<br />

98% at least somewhat confident in study participation, most “extremely”<br />

confident. Overall, median response scores did not change over time, with<br />

no statistically significant differences between the groups in changes in<br />

perceptions <strong>of</strong>: recurrence risk (p�0.45); chances <strong>of</strong> a serious problem<br />

(p�0.12), feeling informed (p�0.99). However, they differed in confidence<br />

about study participation (p�0.04): after unblinding, a higher % <strong>of</strong><br />

placebo-treated patients had increased confidence (38% vs. 25%), and a<br />

higher % <strong>of</strong> bevacizumab-treated patients had decreased confidence (29%<br />

vs. 22%), although 44% across all arms had no change. Conclusions: In a<br />

placebo-controlled, double-blinded RCT, unblinding did not significantly<br />

affect most participants’ views, regardless <strong>of</strong> receipt <strong>of</strong> placebo or experimental<br />

drug. However, confidence in study participation may have been<br />

affected by knowledge <strong>of</strong> receipt <strong>of</strong> bevacizumab; publicity surrounding<br />

this experimental therapy may have affected results.<br />

6027 Poster Discussion Session (Board #15), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Patterns <strong>of</strong> diagnostic imaging and cumulative radiation exposure among<br />

long-term young adult cancer survivors. Presenting Author: Corinne Daly,<br />

<strong>Clinical</strong> Decision Making & Healthcare, Toronto, ON, Canada<br />

Background: Young adults surviving a diagnosis <strong>of</strong> malignancy have a<br />

considerable life expectancy, however, little is known about radiation<br />

exposure from diagnostic imaging in these patients. This study aims to<br />

describe patterns <strong>of</strong> imaging and radiation exposure in young adult cancer<br />

survivors (YAS) and cancer-free controls in Ontario, Canada. Methods: We<br />

conducted a population-based retrospective study. Young adults aged<br />

20-44 diagnosed with an invasive malignancy between 1992 and 1999<br />

who lived at least 5 years without recurrent disease were identified in the<br />

Ontario Cancer Registry. YAS were matched 1:5 to randomly selected<br />

cancer-free controls on calendar year <strong>of</strong> birth, sex, and geographic location.<br />

Radiological procedures were identified through Ontario Health Insurance<br />

Plan administrative data. The rate at which individuals underwent diagnostic<br />

procedures after surviving 5-years was compared between survivors and<br />

controls using Poisson regression. Cumulative radiation exposure due to<br />

computed tomography (CT) and standard x-ray were calculated. Results:<br />

20,911 YAS and 104,524 controls had a median <strong>of</strong> 13.5 years observation<br />

time after cancer diagnosis/referent date. The rate <strong>of</strong> CT scanning after 5<br />

year survival was higher in YAS (rate ratio� 3.59, 95% CI: 3.46 – 3.73)<br />

and varied according to malignancy type (Table). Over the course <strong>of</strong><br />

diagnosis, treatment and surveillance to 10 years, the average YAS was<br />

exposed to 33.8 mSv <strong>of</strong> radiation, 4.4 times the radiation exposure an<br />

individual in the general population received. 47% <strong>of</strong> YAS cumulative dose<br />

was attributed to exposure 5 years or more after diagnosis. Conclusions: YAS<br />

undergo imaging and are exposed to diagnostic irradiation at a significantly<br />

higher rate than controls even after 5 years <strong>of</strong> recurrence free survival.<br />

Alternative imaging techniques not associated with exposure to radiation<br />

should be considered for these patients.<br />

CT after 5 years<br />

n<br />

post-diagnosis<br />

YAS Rate ratio 95% CI<br />

All malignancy types 20,911 3.59 3.46 - 3.73<br />

Breast 4,581 3.21 2.96 - 3.48<br />

Gynecological 2,782 2.38 2.12 - 2.67<br />

Hodgkin’s lymphoma 1,071 6.69 5.82 - 7.69<br />

Melanoma 2,088 2.23 1.94 - 2.57<br />

Non-Hodgkin’s lymphoma 1,277 8.56 7.60 - 9.63<br />

Thyroid 2,388 1.74 1.55 - 1.96<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


6028 Poster Discussion Session (Board #16), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

How do elderly cancer survivors fare? A comparison <strong>of</strong> characteristics,<br />

health status, health behaviors, and spending between Medicare beneficiaries<br />

with and without a cancer history. Presenting Author: Xuehua Ke,<br />

University <strong>of</strong> Maryland School <strong>of</strong> Pharmacy, Batlimore, MD<br />

Background: Prior research on cancer (Ca) survivors has been limited to<br />

small cohorts or claims data with limited measures. In this study we used<br />

population-based survey data to characterize multiple dimensions <strong>of</strong> the<br />

survivor experience including socioceonomic status, health status and<br />

behaviors, and healthcare spending, comparing survivors to beneficiaries<br />

w/o a Ca history. Methods: Data were pooled from the 1997-2007 Medicare<br />

Current Beneficiary Survey (MCBS) including linked claims (3 yrs prior to<br />

MCBS enrollment to concurrent). Ca survivors were identified from ICD-<br />

9-CM codes on claims or self report <strong>of</strong> a prior Ca dx, survived 2� yrs from<br />

observed Ca dx, and had no active treatment, hospice enrollment or death<br />

in the selected survivorship yr. Beneficiary characteristics and healthcare<br />

use were based on self report augmented by claims-based measures.<br />

Bivariate and multivariate analyses were used to compare survivors’<br />

characteristics and spending to controls. Results: The study included<br />

3,921 survivors and 7,056 subjects w/o Ca. Among survivors, breast<br />

(21%), prostate (21%), and colorectal (16%) were most common Ca sites.<br />

Compared to controls without (w/o) Ca, survivors had higher income and<br />

assets, were more likely to have supplemental medical and prescription<br />

drug coverage (74% vs. 70%), had more comorbid conditions as measured<br />

by Charlson Comorbidity Index score ��2 (18% vs. 15%), and a smoking<br />

history (60% vs. 53%); survivors were less likely to avoid visiting doctors<br />

(22% vs. 30%), and more satisfied with the quality <strong>of</strong> medical care<br />

received (96% vs. 92%). Survivors had higher annual healthcare spending<br />

(mean $12,095 vs. $8,928) and outpatient drug spending ($2,205 vs.<br />

$1,898). All differences in healthcare use and spending remained significant<br />

after adjusting for socioeconomic status, health conditions, and<br />

behaviors. Conclusions: Elderly Ca survivorshad more supplemental insurance<br />

coverage, stronger preferences for medical care, used more preventive<br />

services, and had higher spending compared to beneficiaries w/o Ca. These<br />

patterns may reflect the effects <strong>of</strong> survivorship on need for healthcare and<br />

care seeking behaviors.<br />

6030 Poster Discussion Session (Board #18), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Longitudinal changes in health care utilization by adult survivors <strong>of</strong><br />

childhood cancer in the Childhood Cancer Survivor Study (CCSS). Presenting<br />

Author: Jacqueline N. Casillas, UCLA, Los Angeles, CA<br />

Background: The incidence <strong>of</strong> late effects increases as childhood cancer<br />

survivors age. Survivors require lifelong care focused on the risks arising<br />

from prior cancer therapy (survivor-focused care). Methods: We assessed<br />

longitudinal changes in health care utilization in adult survivors <strong>of</strong><br />

childhood cancer participating in the CCSS. Utilization at baseline and<br />

most recent follow-up was classified into one <strong>of</strong> three mutually exclusive<br />

hierarchical categories: no health care, general medical care, or survivorfocused<br />

care. Relative risk (RR) and 95% confidence intervals (CI) were<br />

calculated for predictors <strong>of</strong> reduction in care over time from survivorfocused<br />

to general or no care. Multivariable models, adjusted for key<br />

treatment exposures, were created to assess the risk factors for reductions<br />

in level <strong>of</strong> care over time. Results: Among 8591 eligible survivors, mean age<br />

at last follow-up was 35.1 years (SD�7.8) with a mean <strong>of</strong> 11.6 years<br />

(SD�2.2) since baseline. Of 3993 (46%) survivors who reported survivorfocused<br />

care at baseline, 2383 (59.7%) reported a lower level <strong>of</strong> care at<br />

follow-up. Among 4598 (54%) not receiving survivor-focused care at<br />

baseline, 915 (20%) reported survivor-focused care at follow-up. Baseline<br />

predictors <strong>of</strong> a decreased level <strong>of</strong> care were no health insurance (RR�1.5,<br />

95% CI 1.2-1.9), male sex (RR�1.4, 95% CI 1.2-1.6), being 10-19 years<br />

from diagnosis compared with 20� years (RR�1.4, 95% CI 1.1-1.7). In<br />

contrast, factors associated with a maintenance in survivor-focused care<br />

were Canadian residency compared to U.S. residency with insurance<br />

(RR�0.7, 95% CI 0.6-0.9), unemployment (RR�0.8, 95% CI 0.7-0.9),<br />

physical limitations (RR�0.7, 95% CI 0.6-0.9), cancer-related pain<br />

(RR�0.7, 95% CI 0.5-0.8), poor emotional health (RR�0.7, 95% CI<br />

0.5-0.9), having mild-moderate (RR�0.5, 95% CI 0.4-0.6) or severedisabling<br />

chronic health condition (RR�0.6, 95% CI 0.5-0.7). Conclusions:<br />

Less than a third <strong>of</strong> adult survivors <strong>of</strong> childhood cancer report survivorfocused<br />

care. Rates decrease over time. Targeted interventions to maximize<br />

survivor-focused care in at-risk survivors should be tested so preventive and<br />

risk-reducing opportunities are not lost.<br />

Health Services Research<br />

6029 Poster Discussion Session (Board #17), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Patient-centered medical homes may improve breast cancer surveillance<br />

among survivors. Presenting Author: Stephanie B. Wheeler, University <strong>of</strong><br />

North Carolina at Chapel Hill, Chapel Hill, NC<br />

Background: Community Care <strong>of</strong> North Carolina (CCNC) initiated a medical<br />

home (MH) program in the early 1990s focused on improving care in<br />

Medicaid-insured populations. CCNC has been successful in improving<br />

asthma, diabetes, and cardiovascular disease outcomes, but has not been<br />

examined in the context <strong>of</strong> cancer care. We sought to determine whether<br />

CCNC enrollment was associated with improved cancer surveillance among<br />

breast cancer survivors. Methods: Using state cancer registry records linked<br />

to Medicaid claims, we identified women ages 18-64 diagnosed with stage<br />

0, I, or II breast cancer from 2003-2007. We included only cases insured<br />

by Medicaid for at least 12 <strong>of</strong> 15 months following the index cancer<br />

diagnosis. Reflecting ASCO guidelines for breast cancer surveillance for<br />

survivors (2006), we defined outcomes as time to first surveillance<br />

mammogram post-diagnosis and overall receipt <strong>of</strong> mammogram by 15months<br />

post-diagnosis. Our primary independent variable was enrollment<br />

in CCNC, categorized as never enrolled, enrolled up to 6 months, and<br />

enrolled 7 months or more. We used multivariate Cox proportional hazards<br />

stratified by receipt <strong>of</strong> radiation therapy (RT) and logistic regressions.<br />

Results: 840 women were included in our sample. Approximately half were<br />

enrolled in CCNC for at least some time during the study period, 38% for<br />

more than 7 months post-diagnosis. Among women who received RT, being<br />

in a MH for at least 7 months corresponded to earlier follow-up mammogram<br />

(Hazard Ratio: 1.34; p�0.028), controlling for all other factors.<br />

Enrollment in a MH for at least 7 months post-diagnosis also was<br />

associated with overall receipt <strong>of</strong> mammogram by 15 months (p�0.01).<br />

Interaction terms indicated that women enrolled in MHs and living in a<br />

rural area had a statistically significant higher likelihood <strong>of</strong> receiving<br />

mammography. Conclusions: Results suggest that MH enrollment is associated<br />

with improved cancer surveillance among breast cancer survivors<br />

insured by Medicaid. Given the growing population <strong>of</strong> cancer survivors and<br />

increased emphasis on MHs in the Affordable Care Act, more research is<br />

needed to explore how patient-centered medical homes can be enhanced to<br />

improve the transition from cancer patient to cancer survivor.<br />

6031 Poster Discussion Session (Board #19), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

How radiation oncologist accessibility influences treatment choice and<br />

quality in early-stage breast cancer: A SEER database analysis. Presenting<br />

Author: Thomas M. Churilla, The Commonwealth Medical College, Scranton,<br />

PA<br />

Background: Mastectomy and breast conserving therapy (BCT, partial<br />

mastectomy and adjuvant radiotherapy) are equivalent in survival for<br />

treatment <strong>of</strong> early stage breast cancer. This study evaluated the impact <strong>of</strong><br />

radiation oncologist accessibility on choice <strong>of</strong> mastectomy versus BCT, and<br />

the receipt <strong>of</strong> radiotherapy after BCT. Methods: In the NCI SEER database,<br />

breast cancer cases from 2004-2008 were selected with the following<br />

criteria: T2N1M0 or less, lobular or ductal histology, and treatment with<br />

simple mastectomy or partial mastectomy (�/-) adjuvant radiation. The<br />

HRSA Area Resource File was combined to define average radiation<br />

oncologist density (ROD, number <strong>of</strong> radiation oncologists/100K people) by<br />

county over the same time period. Tumor characteristics, demographic<br />

information, and ROD were evaluated with respect to mastectomy rates and<br />

receipt <strong>of</strong> radiation therapy after BCT in univariate and multivariate<br />

analyses. Results: In the 118,961 cases analyzed, mastectomy was<br />

performed 33.3% <strong>of</strong> the time relative to BCT. After adjustment for<br />

demographic and tumor variables, the odds <strong>of</strong> having mastectomy versus<br />

BCT were inversely associated with ROD (OR [95% CI] � 0.94 [0.93-<br />

0.96]; p�0.001). Adjuvant radiation therapy was not administered in<br />

23.4% <strong>of</strong> BCT cases. Likewise, the odds <strong>of</strong> having BCT without adjuvant<br />

radiation were inversely associated with ROD (0.96 [0.95-0.98]; p�0.001,<br />

table). Conclusions: There was a significant, inverse and linear relationship<br />

between ROD and mastectomy rates independent <strong>of</strong> demographic and<br />

tumor variables. An inverse trend was also observed for the omission <strong>of</strong><br />

radiotherapy after BCT. Access to radiation oncologists was a factor in<br />

surgical choice and receiving appropriate BCT in early stage breast cancer.<br />

Radiation<br />

oncologist<br />

density a<br />

(percentile)<br />

Mastectomy<br />

rate b (%)<br />

Odds <strong>of</strong><br />

omission <strong>of</strong><br />

radiotherapy<br />

after BCT c [95% CI] p value<br />

0-10 th d 45.4 1.30 1.22-1.39 �0.001<br />

10-25 th 40.6 0.92 0.85-0.99 0.020<br />

25-75 th 37.9 1.00 Referent --<br />

75-90 th 40.0 0.85 0.79-0.92 �0.001<br />

90-100 th 36.4 0.89 0.83-0.95 �0.001<br />

a Number <strong>of</strong> radiation oncologists/100K.<br />

b Mastectomy/[Mastectomy � BCT w/ radiation].<br />

c Multivariate.<br />

d 0-10 th represents 0 radiation oncologists/100K.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.<br />

389s


390s Health Services Research<br />

6032 Poster Discussion Session (Board #20), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Life expectancy (LE) and the receipt <strong>of</strong> conservative versus active treatment<br />

in men with prostate cancer (CaP): A population-based study. Presenting<br />

Author: Trevor Joseph Royce, UNC-CH School <strong>of</strong> Medicine, Chapel Hill, NC<br />

Background: Prostate-specific antigen (PSA) screening increases the diagnosis<br />

<strong>of</strong> low-risk and potentially clinically insignificant CaP, which raises<br />

concern for possible overtreatment. The National Comprehensive Cancer<br />

Network (NCCN) guidelines recommend surveillance (conservative management)<br />

for patients with less than 10 years LE diagnosed with low-risk<br />

cancer. In contrast, NCCN recommends active treatment for high-risk CaP,<br />

the most aggressive form <strong>of</strong> this disease, irrespective <strong>of</strong> LE. We examine<br />

patterns <strong>of</strong> care in CaP patients in the Surveillance, Epidemiology and End<br />

Results (SEER) registry by LE. Methods: 152,578 men with non-metastatic<br />

CaP diagnosed from 2004-8 were included. Gleason, PSA, and clinical<br />

stage were used for risk-categorization per D’Amico criteria. The sample<br />

was dichotomized into men 76 years and younger (who have an average LE<br />

<strong>of</strong> 10 years or more based on the US Social Security Administration<br />

actuarial period life tables) vs. 77 years and older (less than 10 years<br />

average LE). Logistic regression models examined factors associated with<br />

each treatment modality. Results: 56% <strong>of</strong> patients age 77 and older with<br />

low-risk CaP received conservative management, and 44% active treatment<br />

(Table). However, conservative management was just as common in<br />

older patients with high-risk cancer (62%); 21% <strong>of</strong> younger patients with<br />

high-risk CaP also received conservative management. Multivariable analysis<br />

showed decreased use <strong>of</strong> conservative management over time in older<br />

patients (OR 0.78 for 2008 vs. 2004, 95%CI 0.71-.86, p�.001). African<br />

<strong>American</strong> race, being unmarried, and older age were also significantly<br />

associated with conservative management. Conclusions: There may be<br />

overtreatment <strong>of</strong> low-risk CaP patients age 77 and older, which is worsening<br />

in recent years. Correspondingly, there appears to be undertreatment <strong>of</strong><br />

elderly patients with high-risk CaP, the most aggressive form <strong>of</strong> this<br />

disease.<br />

Percent <strong>of</strong> patients receiving each treatment stratified by CaP risk and age.<br />

Low risk Intermediate risk High risk<br />

�77 �77 �77<br />

Conservative 22 56 13 47 21 62<br />

Radical prostatectomy 35 1 50 2 39 2<br />

Beam radiation 22 28 28 42 35 33<br />

Brachytherapy 21 15 8 9 6 3<br />

6034 Poster Discussion Session (Board #22), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Do patients with advanced-stage non-small cell lung cancer (AS NSCLC)<br />

live longer if managed within a clinical trial setting? Presenting Author:<br />

Taher Abu Hejleh, Division <strong>of</strong> Hematology, Oncology and Marrow Transplantation,<br />

Department <strong>of</strong> Internal Medicine, University <strong>of</strong> Iowa Hospitals and<br />

Clinics, Iowa City, IA<br />

Background: Treatment outcomes <strong>of</strong> AS NSCLC (stages IIIB and IV) are<br />

poor. There is an argument that participation in a clinical trial (CT) may<br />

confer survival benefit, probably, through enhancing quality <strong>of</strong> care. In this<br />

study, we explore the survival outcomes and perceived care quality for AS<br />

NSCLC patients (pts) treated within vs outside a CT. Methods: Data were<br />

obtained from surveys <strong>of</strong> newly diagnosed AS NSCLC pts studied by the<br />

Cancer Care Outcomes Research and Surveillance Consortium (CanCORS),<br />

a large cohort <strong>of</strong> pts across the United States. Pts who did not complete the<br />

baseline survey were excluded as this was associated with worse performance<br />

status (PS). Baseline characteristics according to CT participation<br />

were determined. Association between CT enrollment and survival was<br />

explored utilizing univariate and multivariate survival analysis after adjusting<br />

for age, comorbidities and self-reported PS. Results: Of 815 AS NSCLC<br />

pts, 56 (7%) were enrolled on a CT. Chemotherapy trials comprised 67% <strong>of</strong><br />

all trials. Of the 815 pts, 697 (86%) died. Median survival for pts within vs<br />

outside a CT was 62 vs 64 months. Neither age, comorbidities nor recalled<br />

PS differed significantly between pts within vs outside a CT (P�0.2085,<br />

0.5818 and 0.1678 respectively). On the multivariate survival model, CT<br />

enrollment did not correlate with longer survival (P�0.8811) and only<br />

presence <strong>of</strong> comorbidities was associated with worse survival (P�0.0021).<br />

Comparing pts according to CT enrollment, there was no significant<br />

difference in symptom management, receiving hospice care (P�0.606),<br />

death location (P�0.2018), or following pts’ wishes (P�0.8321). However,<br />

perception <strong>of</strong> the overall cancer care quality was greater among CT<br />

enrollees (P�0.0171). Conclusions: Management <strong>of</strong> AS NSCLC pts within<br />

a CT setting conveyed a perception <strong>of</strong> superior care that did not translate<br />

into survival benefit after adjusting for differences in age, comorbidities,<br />

and self-reported PS. These findings suggest that providing cancer care<br />

within a CT should not imply a survival benefit when counseling AS NSCLC<br />

pts about entering CTs.<br />

6033 Poster Discussion Session (Board #21), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Timeliness <strong>of</strong> care and stage at diagnosis <strong>of</strong> non-small cell lung cancer<br />

(NSCLC) with the implementation <strong>of</strong> a cancer care coordination program<br />

(CCCP) at a VA medical center. Presenting Author: Susan Alsamarai,<br />

Yale-New Haven Hospital, New Haven, CT<br />

Background: Timeliness <strong>of</strong> care improves patient satisfaction and may<br />

improve outcomes. A CCCP was established in Nov 2007 to improve<br />

timeliness <strong>of</strong> care <strong>of</strong> NSCLC patients at the Veterans Affairs Connecticut<br />

(VACT) Healthcare System. Methods: We performed a retrospective cohort<br />

analysis <strong>of</strong> patients diagnosed with NSCLC at VACT between 2005-2010.<br />

We compared timeliness <strong>of</strong> care and stage at diagnosis before and after the<br />

implementation <strong>of</strong> the CCCP. Results: Data from 352 patients was<br />

analyzed: 163 with initial abnormal imaging between 1/1/2005 and<br />

10/31/2007, and 189 with imaging between 11/1/2007 and 12/31/2010.<br />

Variables associated with a longer interval between the initial abnormal<br />

image and the initiation <strong>of</strong> therapy were: (1) earlier stage (mean <strong>of</strong> 130<br />

days for stages I/II vs. 87 days for stages III/IV, p�0.001),(2) lack <strong>of</strong><br />

cancer-related symptoms (145 vs 60 days, p�0.001), (3) presence <strong>of</strong><br />

medical co-morbidities (111 vs 76 days, p�0.01), and (4) depression<br />

(127 vs 98 days, p�0.029). Substance abuse increased the interval from<br />

initial abnormal image to tissue diagnosis by 29 days (p�0.032) but did<br />

not affect the interval from image to treatment. The mean interval between<br />

diagnosis and initiation <strong>of</strong> treatment was 19 days longer in blacks vs.<br />

non-blacks (55 vs 36 days, p�0.0118) although the overall time from<br />

abnormal image to diagnosis and to treatment was not statistically<br />

different. In a multivariate model adjusting for stage, histology, reason for<br />

initial imaging, and presence <strong>of</strong> a primary care provider, implementation <strong>of</strong><br />

a CCCP resulted in a mean reduction <strong>of</strong> 25 days in the time between the<br />

first abnormal image and initiation <strong>of</strong> cancer treatment (126 to 101 days,<br />

p�0.0154). The percent <strong>of</strong> patients diagnosed at stages I and II increased<br />

from 32% to 48% (p�0.0065) after the implementation <strong>of</strong> a CCCP.<br />

Conclusions: A centralized, multidisciplinary, hospital-based CCCP can<br />

improve timeliness <strong>of</strong> NSCLC care, and may also help ensure that<br />

incidental, early stage lung cancers are treated.<br />

6035 Poster Discussion Session (Board #23), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Results <strong>of</strong> a cluster randomized trial to evaluate a nursing lead supportive<br />

care intervention in newly diagnosed breast and colorectal cancer patients.<br />

Presenting Author: Jonathan Sussman, Juravinski Cancer Centre, Hamilton,<br />

ON, Canada<br />

Background: Patient transitions during the early phases <strong>of</strong> cancer care from<br />

initial diagnosis through oncology consultation are <strong>of</strong>ten poorly coordinated<br />

resulting in unmet need, poor continuity, and resultant distress. It has been<br />

proposed that better coordination <strong>of</strong> care during this period would improve<br />

the care experience from the patient’s perspective. We designed a<br />

randomized trial to test a community based nursing lead coordination <strong>of</strong><br />

care intervention in newly diagnosed breast and colorectal cancer patients.<br />

Methods: Cluster randomized control trial in 193 newly diagnosed breast<br />

and colorectal cancer patients enrolled through surgical practices within 7<br />

days <strong>of</strong> cancer surgery in Toronto, Canada. Surgical practices were<br />

randomized between a standardized nursing intervention and a control<br />

group involving usual care practices. The intervention consisted <strong>of</strong> a<br />

standardized in person supportive care assessment with ongoing supportive<br />

care by telephone or in person that included linkage to community services<br />

using protocol specified guidelines according to identified needs. The<br />

primary outcomes measured at 8 weeks were validated patient reported<br />

outcomes (PROs) <strong>of</strong> 1) unmet need (SCNS) and 2) continuity <strong>of</strong> care<br />

(CCCQI). Secondary outcomes included 1) quality <strong>of</strong> life (EORTC QLQ-<br />

C30), 2) health resource utilization, and 3) level <strong>of</strong> uncertainty with care<br />

trajectory (MUIS) at 8 weeks. Results: 121 breast and 72 colorectal<br />

patients were randomized through 28 surgical practices. The intervention<br />

group had a median <strong>of</strong> 6 nursing contacts over the study period. There were<br />

no differences between groups on PROs <strong>of</strong> unmet need, continuity <strong>of</strong> care,<br />

quality <strong>of</strong> life, or uncertainty. Health service utilization did not differ<br />

between groups. Conclusions: A specialized oncology nursing intervention<br />

early in the care trajectory did not result in improved supportive care<br />

outcomes for patients.<br />

Intervention, n � 85<br />

Mean (SD)<br />

Control, n � 101<br />

Mean (SD)<br />

Difference<br />

[95% CI]<br />

SCNS Summary Score 2.0 (0.7) 1.9 (0.7) 0.1 [-0.3, 0.7]<br />

CCCQI Summary Score 4.0 (0.6) 4.0 (0.5) 0.0 [-0.2, 0.2]<br />

EORTC Global Health 66 (20) 67 (22) 1 [-5.6, 6.9]<br />

MUIS Summary Score 46 (15) 47 (14) 1 [-5.2, 2.8]<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


6036 Poster Discussion Session (Board #24), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Which women with breast cancer do, and do not, undergo receptor status<br />

testing? A population-based study. Presenting Author: Linda Sharp, National<br />

Cancer Registry Ireland, Cork, Ireland<br />

Background: Receptor status is a determinant <strong>of</strong> breast cancer (BC)<br />

treatment and prognosis. <strong>Clinical</strong> practice guidelines advise that all women<br />

diagnosed with BC may undergo estrogen (ER), progesterone (ER) and<br />

human epidermal growth factor 2 (HER2) receptor tests. We conducted a<br />

population-based study to investigate what proportions <strong>of</strong> women undergo<br />

testing and predictors <strong>of</strong> test receipt. Methods: Incident BCs (ICD10 C50)<br />

diagnosed 2006-2008 were identified from the National Cancer Registry<br />

Ireland. Receptor tests were identified from registration records augmented<br />

by review <strong>of</strong> selected medical records. For each <strong>of</strong> the three receptors<br />

separately, logistic regression was used to identify which socio-demographic<br />

(age, marital status, smoking status, area <strong>of</strong> residence, and<br />

deprivation category) and clinical factors (grade, tumour size (T), nodal<br />

status (N), distant metastasis (M)) were associated with undergoing<br />

testing. Adjusted odds ratios (OR) and 95% confidence intervals were<br />

computed. Results: 7619 BCs were included. 7% were not tested for any <strong>of</strong><br />

the receptors. 73% were tested for all three. Considering each receptor<br />

separately, 90% had an ER test, 86% a HER2 test and 80% a PR test.<br />

Women with T4 tumours were less likely to have ER and HER2 tests, and<br />

more likely to have a PR test, than women with T1 tumours. After<br />

adjustment for clinical variables, age was not a significant predictor for ER<br />

and HER2 testing, while older women (�70) were more likely than younger<br />

women to have a PR test. For all three tests, non-married women were<br />

significantly less likely to be tested (ER: OR�0.65, 95%CI 0.52-0.8; PR:<br />

OR�0.81, 95%CI 0.7-,0.93; HER2: OR�0.72, 95%CI 0.62-0.85) and<br />

current smokers more likely (ER: OR�1.55, 95%CI 1.14-2.1: PR:<br />

OR�1.38, 95%CI 1.15-1.66; HER2: OR�1.25, 95%CI 1.01-1.55). Area<br />

<strong>of</strong> residence at diagnosis was a significant predictor <strong>of</strong> having each test,<br />

although the geographical patterns varied. Conclusions: More than onequarter<br />

<strong>of</strong> women with BC do not have one or more <strong>of</strong> the receptor tests.<br />

After adjusting for clinical variables, socio-demographic factors were<br />

significant predictors <strong>of</strong> test receipt. In the interests <strong>of</strong> equity, the reasons<br />

underlying these associations should be further investigated.<br />

6038 General Poster Session (Board #1A), Mon, 1:15 PM-5:15 PM<br />

Somatic mutation spectrum <strong>of</strong> non-small cell lung cancers (NSCLCs) from<br />

African <strong>American</strong>s (AAs). Presenting Author: Philip Edward Lammers,<br />

Vanderbilt-Ingram Cancer Center, Nashville, TN<br />

Background: In the AA population, previous studies have presented conflicting<br />

data on the frequency <strong>of</strong> EGFR mutations (Reinersman JTO 2011;<br />

Leidner JCO 2009), while frequencies <strong>of</strong> other gene mutations and<br />

translocations, including anaplastic lymphoma kinase (ALK), have not<br />

been described. Methods: 161 archival FFPE tumor specimens from self<br />

reported AA patients with any stage NSCLC from 1997-2010 were<br />

collected from 3 sites in Tennessee (132 samples) and one site in Michigan<br />

(29 samples). Samples were evaluated for known recurrent driver mutations<br />

in EGFR, KRAS, BRAF, NRAS, AKT1, PI3KCA, PTEN, HER-2, MEK1<br />

by standard SNaPshot/sizing assays, and translocations in ALK by FISH.<br />

<strong>Clinical</strong> data was collected on 119 patients. Chi-square was used to<br />

compare the frequency <strong>of</strong> mutations in subgroups and Kaplan-Meier and<br />

log rank were used to calculate and compare PFS between groups. Results:<br />

5.0% <strong>of</strong> tumors had EGFR mutations, 14.9% had KRAS mutations, 0.6%<br />

had a BRAF, AKT1, PI3KCA, or HER2 mutation, and 0% had NRAS, PTEN,<br />

or MEK1 mutations. Of 35 ‘pan-negative’ non-squamous specimens, 0 had<br />

ALK translocations. PFS was the same in those with and without KRAS<br />

mutation (p�0.74) and showed a trend towards improvement in those with<br />

EGFR mutation (p�0.08). The frequency <strong>of</strong> EGFR mutations was higher in<br />

samples from Detroit versus those from Tennessee (17% vs 2.3%,<br />

p�0.01), as was the frequency <strong>of</strong> adenocarcinoma (62% vs 44%,<br />

p�0.05). The frequency <strong>of</strong> EGFR mutations in never smokers was higher in<br />

the samples from Detroit versus Tennessee (83% vs 7.1%, p�0.01).<br />

Conclusions: In the largest tumor mutational pr<strong>of</strong>iling study <strong>of</strong> NSCLC from<br />

AAs to date, EGFR mutations occurred less frequently than would be<br />

expected from a North <strong>American</strong> population. We noted a regional difference,<br />

with fewer EGFR mutations in Tennessee than in Michigan, a finding<br />

that may have been the result <strong>of</strong> more adenocarcinoma samples from<br />

Michigan. The rates <strong>of</strong> other mutations and translocations including ALK<br />

were low. While lung cancer tumors should continue to undergo routine<br />

molecular testing to prioritize therapy, future comprehensive genotyping<br />

efforts should focus on identifying novel driver mutations in this population.<br />

Funding: 5RC1CA162260 R01CA060691 R01CA87895.<br />

Health Services Research<br />

391s<br />

6037 Poster Discussion Session (Board #25), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Racial differences in delay <strong>of</strong> treatment for localized prostate cancer (CaP):<br />

A population-based study. Presenting Author: William Allen Stokes, University<br />

<strong>of</strong> North Carolina at Chapel Hill, Chapel Hill, NC<br />

Background: African-<strong>American</strong>s (AA) are diagnosed with more advanced<br />

CaP than Caucasians (CA) and are more likely to die from CaP. Treatment<br />

delay is a potentially modifiable obstacle to care and clinically may be more<br />

important in AA patients because <strong>of</strong> more aggressive cancer at diagnosis.<br />

We examined time from diagnosis to curative treatment (surgery or<br />

radiation) in AA and CA patients in the Surveillance, Epidemiologic and<br />

End Results (SEER)-Medicare linked database. Methods: 21,454 CA and<br />

2,506 AA patients who were diagnosed with non-metastatic CaP from<br />

2004-08 and received treatment within 12 months <strong>of</strong> diagnosis were<br />

included. Linear regression was used to examine factors associated with<br />

number <strong>of</strong> days from diagnosis to treatment initiation, and logistic<br />

regression to assess odds <strong>of</strong> treatment within 6 months <strong>of</strong> diagnosis.<br />

Results: AA patients were more likely to have high-risk CaP than CA patients<br />

(39 vs. 35%), and less likely to have low-risk CaP (27 vs. 31%) (p�.001).<br />

Time to treatment was significantly prolonged for AA patients in all risk<br />

groups <strong>of</strong> CaP, and the difference was most prominent for high-risk patients<br />

(median 105 days for AA vs. 96 days for CA, p�.002). Racial differences in<br />

time to treatment persisted in multivariable analysis (Table). Sensitivity<br />

analyses examining the proportion <strong>of</strong> AA and CA patients initiating<br />

treatment within 6 months <strong>of</strong> diagnosis revealed similar results. Conclusions:<br />

AA patients, especially those with high-risk CaP, experience longer treatment<br />

delays than CA patients. This is the first large-scale study to examine<br />

treatment delays in AA and CA patients with CaP. The differences found<br />

may contribute to our understanding <strong>of</strong> racial disparities in CaP treatment<br />

outcomes.<br />

Covariate Coefficient (days) P value<br />

Race (vs. CA)<br />

AA 6.7 �.001<br />

Risk group (vs. low-risk)<br />

Intermediate 1.2 .25<br />

High 4.2 �.001<br />

Age (vs. 65-69)<br />

70-74 -1.4 .15<br />

>75 -2.2 .05<br />

Modified Charlson comorbidity (vs. 0)<br />

>0 -0.1 .90<br />

Controls for SEER region, regional socioeconomic indicators, marital status, and<br />

treatment modality.<br />

6039 General Poster Session (Board #1B), Mon, 1:15 PM-5:15 PM<br />

Do patient-reported symptoms predict for emergency department visits? A<br />

population-based analysis. Presenting Author: Lisa Catherine Barbera,<br />

Odette Cancer Centre, Toronto, ON, Canada<br />

Background: Since 2007 in Ontario, Canada, the Edmonton Symptom<br />

Assessment System (ESAS) has been routinely used to assess symptoms in<br />

cancer patients in both ambulatory and home-care settings. The purpose <strong>of</strong><br />

this study was to determine the relationship between individual patient<br />

symptoms, and their severity, with the likelihood <strong>of</strong> an emergency department<br />

(ED) visit. Methods: The cohort includes all cancer patients in Ontario<br />

who completed an ESAS assessment between January 2007 and March<br />

2009. We linked multiple provincial health databases to describe the<br />

cohort and determine if an ED visit occurred within 7 days <strong>of</strong> the patient’s<br />

first ESAS. Multivariate logistic regression was used to determine the<br />

association between symptom scores (absent: score 0; mild: 1-3; moderate:<br />

4-7; severe: 8-10) and the likelihood <strong>of</strong> an ED visit. Results: The cohort<br />

included 45,118 unique patients whose first assessment contributes to the<br />

study. 3.8% (n�1732) had an ED visit. The patients with ED visits were<br />

more likely to be men, to have lung or gastro-intestinal cancer, to have had<br />

recent radio or chemotherapy, and to have a shorter survival. The proportion<br />

<strong>of</strong> patients with ED visits increased from 2% to 10-12% as individual<br />

symptom scores increased from 0 to 10. Anxiety and depression were not<br />

associated with ED visits in the model, regardless <strong>of</strong> severity. Pain, nausea,<br />

drowsiness, appetite and shortness <strong>of</strong> breath with moderate or severe<br />

scores were associated with ED visits. Tiredness and wellbeing were the<br />

only symptoms to show a significant association for mild, moderate and<br />

severe scores. A well being score <strong>of</strong> 7-10 (reference score�0) had the<br />

highest odds ratio <strong>of</strong> 1.8 (95% CI 1.4-2.3). Conclusions: Worsening<br />

symptoms clearly contribute to ED visits. While specific symptoms like pain<br />

are obvious targets for management in the outpatient setting, constitutional<br />

symptoms like wellbeing or fatigue are associated with even higher odds.<br />

Though difficult to manage, such symptoms also warrant detailed assessment<br />

in order to optimize patient outcomes.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


392s Health Services Research<br />

6040 General Poster Session (Board #1C), Mon, 1:15 PM-5:15 PM<br />

Utilization <strong>of</strong> accelerated partial breast irradiation for ductal carcinoma in<br />

situ, 2002-2007: Report from the National Cancer Data Base. Presenting<br />

Author: Tomasz Czechura, North Shore University HealthSystem, Evanston,<br />

IL<br />

Background: The use <strong>of</strong> accelerated partial breast irradiation using brachytherapy<br />

(APBI-b) for patients with invasive cancer is increasing but data for<br />

ductal carcinoma in situ (DCIS) cases are limited. The <strong>American</strong> <strong>Society</strong> <strong>of</strong><br />

Radiation Oncology (ASTRO) guidelines suggest that APBI-b should be<br />

used only on a �cautionary� or �on trial� basis for women with DCIS. The<br />

purpose <strong>of</strong> this study was to examine utilization trends and correlates <strong>of</strong><br />

APBI-b use for patients with DCIS. Methods: A total <strong>of</strong> 70,043 postlumpectomy<br />

patients from the National Cancer Database diagnosed with DCIS<br />

between 2002 and 2007 were studied. Chi-square tests and logistic<br />

regression models were used to determine trends and factors related to<br />

APBI-b use. Results: The use <strong>of</strong> APBI-b increased from 0.7% in 2002 to<br />

10.0% in 2007 (p�0.001). Independent predictors APBI-b use were age,<br />

race, insurance status, comorbidity index, facility type, and facility location.<br />

Older patients were more likely to use APBI-b; relative to patients<br />

30-39 years old, the OR for patients 80-89 years old was 5.9 (95% CI:<br />

3.7-9.6). APBI-b use was higher in whites (4.9%), compared to blacks<br />

(4.4%), Hispanics (2.8%), and Asian pacific islanders (1.5%; p�0.001).<br />

Compared to noninsured, Medicare (OR�2.1, 95% CI: 1.3-3.3) and<br />

managed care patients (OR�2.0, 95% CI: 1.3-3.2) were more likely to<br />

undergo APBI-b. 2.3% <strong>of</strong> community cancer programs, 5.7% <strong>of</strong> comprehensive<br />

community programs and 4.1% <strong>of</strong> academic/research programs<br />

utilized APBI-b for treating DCIS (p�0.001). The use <strong>of</strong> APBI-b varied<br />

significantly by facility location; the West and South regions <strong>of</strong> the country<br />

were more likely to use APBI-b (OR�14, 95% CI: 10.1-19.6) than the<br />

Northeast region. 91% <strong>of</strong> the APBI-b patients fit the ASTRO �cautionary�<br />

guideline and 9% <strong>of</strong> patients fit the ASTRO �on trial� guidelines. In 2002,<br />

4% <strong>of</strong> ABPI-b patients fit the �on trial� guidelines which increased to 8.6%<br />

in 2007 (p�0.001). Conclusions: The use <strong>of</strong> APBI-b for DCIS increased<br />

from 2002-2007. APBI-b use varies by socioeconomic and facility factors<br />

with age being the most significant factor. Future studies are needed to<br />

determine the indications for APBI-b in patients with DCIS.<br />

6042 General Poster Session (Board #1E), Mon, 1:15 PM-5:15 PM<br />

Prospective evaluation <strong>of</strong> perceived barriers to medication adherence by<br />

patients on oral antineoplastics. Presenting Author: Benyam Muluneh,<br />

University <strong>of</strong> North Carolina Hospitals and Clinics Department <strong>of</strong> Pharmacy,<br />

Chapel Hill, NC<br />

Background: Appropriate use <strong>of</strong> oral antineoplastics (OA), especially those<br />

with a food-effect, is a challenge for patients and clinicians. Patient<br />

adherence is essential to optimize outcomes, minimize toxicity, minimize<br />

bias in clinical trials, and reduce health care costs. As the use <strong>of</strong> OA grows,<br />

due to the narrow therapeutic margin, it is critical to understand the<br />

barriers to patient adherence. The purpose <strong>of</strong> this study was to analyze<br />

cancer patients’ use <strong>of</strong> OAs and identify opportunities to improve patient<br />

adherence. Methods: We developed and tested a 30-question survey to<br />

address frequency and reasons for reducing/skipping doses; sources <strong>of</strong><br />

information for OA use; perceived importance <strong>of</strong> food-drug effects, and<br />

ease <strong>of</strong> understanding directions on vial label. Surveys, consisting <strong>of</strong> Likert<br />

scale and multiple choice questions, were distributed to adult cancer<br />

patients on OAs at the UNC Cancer Hospital clinics. Results: Seventy-seven<br />

patients taking OAs with CML, RCC, breast cancer, and GI tumors<br />

completed the survey with a response rate <strong>of</strong> 97%. This was a welleducated<br />

population with 71% having completed some college; 54%<br />

female and 58% older than 50 years. Forty-three percent <strong>of</strong> patients taking<br />

drugs with a significant food-drug effect (sorafenib, pazopanib, lapatinib,<br />

imatinib, nilotinib, and capecitabine) did not think about the last time they<br />

ate before taking their OA and 23% did not know that their OA had a<br />

food-drug effect. In addition, 21% <strong>of</strong> patients indicated they intentionally<br />

skipped/cut back on their OAs and 38% <strong>of</strong> those did not inform their<br />

physician. Although 97% reported no difficulty reading instructions on<br />

drug vial, nearly 20% had some difficulty understanding the directions.<br />

Conclusions: There are three main barriers associated with appropriate use<br />

<strong>of</strong> OAs: confusion or misunderstanding about the timing <strong>of</strong> drug with food;<br />

reducing/stopping drug without informing MD; and difficulty understanding<br />

directions on the drug vial label. A multipronged integrated approach<br />

involving the pharmacist, physician and nurse is needed to optimize<br />

communication <strong>of</strong> directions for optimal OA use.<br />

6041 General Poster Session (Board #1D), Mon, 1:15 PM-5:15 PM<br />

Conflict <strong>of</strong> interest disclosure in <strong>of</strong>f-label oncology clinical trials. Presenting<br />

Author: Blair Billings Irwin, Duke University Medical Center, Durham,<br />

NC<br />

Background: Off-label prescribing <strong>of</strong> anti-cancer targeted therapies is<br />

increasingly prevalent. Post-regulatory evidence and reporting requirements<br />

for reimbursement for <strong>of</strong>f-label oncologic indications are less<br />

stringent than the threshold for United States Food and Drug Administration<br />

registration. What are the prevalence, reliability, and predictors <strong>of</strong><br />

conflict <strong>of</strong> interest (COI) and funding disclosure statements in published<br />

reports <strong>of</strong> studies <strong>of</strong> cancer targeted therapies conducted in the postregulatory<br />

setting? Methods: As a part <strong>of</strong> a federally-funded systematic<br />

review, manuscripts were included in the analysis if they were used to<br />

support one <strong>of</strong> 19 indications for cancer targeted therapies that were<br />

<strong>of</strong>f-label but reimbursable according to authoritative compendia in 2006 or<br />

before. Studies were categorized according to trial design, trial results,<br />

average impact factor <strong>of</strong> journals, and the presence <strong>of</strong> COI and funding<br />

disclosure statements. Results: Sixty-nine studies were included in the<br />

systematic review. Prevalence <strong>of</strong> COI and funding disclosures was low, at<br />

33% and 58%, respectively; time trends showed some improvement<br />

between 2002 to 2007, but only 60% <strong>of</strong> studies had disclosures by 2007.<br />

Predictors <strong>of</strong> COI disclosure were publication in high impact factor journals<br />

(p�0.001), large study sample size (p�0.001), enrollment exclusively in<br />

the US (p�0.04), and study <strong>of</strong> the targeted therapy in combination with<br />

other agents as opposed to the study drug alone (p�0.03). Studies with<br />

multiple sites were more likely to include a funding disclosure statement<br />

(p�0.01). Conclusions: There is need for more consistent disclosure <strong>of</strong><br />

potential sources <strong>of</strong> bias in COI and funding statements in studies <strong>of</strong><br />

<strong>of</strong>f-label indications for anti-cancer targeted therapies.<br />

6043 General Poster Session (Board #1F), Mon, 1:15 PM-5:15 PM<br />

Bias in reporting <strong>of</strong> endpoints <strong>of</strong> efficacy and toxicity in randomized clinical<br />

trials (RCTs) for women with breast cancer (BC). Presenting Author:<br />

Francisco Emilio Vera Badillo, Princess Margaret Hospital, Toronto, ON,<br />

Canada<br />

Background: Phase III RCTs are designed to assess clinically important<br />

differences in endpoints that reflect benefit to patients. Accurate and<br />

unbiased reporting is essential to guide rational therapy. Here we evaluate<br />

the quality <strong>of</strong> reporting <strong>of</strong> the primary endpoint (PE) and <strong>of</strong> toxicity in RCTs<br />

for BC. Methods: PUBMED was searched from 1995-2011 to identify RCTs<br />

for BC. Scales for assessing bias in reporting <strong>of</strong> the PE and <strong>of</strong> toxicity were<br />

developed. For the PE, scales assessed whether the concluding statement<br />

<strong>of</strong> the abstract (CSAbs) was (a) based on the PE, (b) described appropriately<br />

a statistically positive or negative result for the PE, or (c) was based on<br />

secondary endpoints. Bias and completeness <strong>of</strong> reporting <strong>of</strong> toxicity was<br />

assessed using a hierarchy scale <strong>of</strong> whether reporting occurred in the<br />

CSAbs, elsewhere in the abstract, in the discussion <strong>of</strong> the article or only in<br />

the results. Association <strong>of</strong> bias with Journal Impact Factor (JIF); changes in<br />

the PE compared to protocol information in clinicaltrials.gov and funding<br />

source was also evaluated. Results: 164 trials were evaluated; 33% showed<br />

bias in reporting <strong>of</strong> the PE. The PE was more likely to be reported in the<br />

CSAbs if statistically significant [OR 5.2, 95% CI 1.9-14.3, p�0.001]. A<br />

statistically negative PE was not reported in the CSAbs in 27% <strong>of</strong> trials. Of<br />

the 30 trials where protocol information was available in clinicaltrials.gov,<br />

there were non-significant associations for the PE to show a positive result<br />

if had been changed, and for greater bias in reporting the PE if it was<br />

unchanged. 67% <strong>of</strong> studies showed bias in reporting <strong>of</strong> toxicity. Only 14%<br />

mentioned toxicity in CSAbs. When the PE was positive, bias in reporting <strong>of</strong><br />

toxicity was more common [OR 2.0, 95% CI 1.0-3.9, p�0.04]. There was<br />

no apparent association between bias in reporting <strong>of</strong> either the PE or<br />

toxicity and JIF or funding source, but a non-significant association<br />

between change in the PE and industry funding. Conclusions: Bias in<br />

reporting <strong>of</strong> the PE is common especially for studies with a negative PE.<br />

Reporting <strong>of</strong> toxicity is poor especially for studies with a positive PE.<br />

Changing <strong>of</strong> the PE appears to be a strategy to increase the likelihood <strong>of</strong><br />

observing a statistically significant result.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


6044 General Poster Session (Board #1G), Mon, 1:15 PM-5:15 PM<br />

Unintended consequences <strong>of</strong> health information technology: Evidence<br />

from Veterans Affairs Colorectal Cancer Oncology Watch Intervention.<br />

Presenting Author: John Bian, South Carolina College <strong>of</strong> Pharmacy,<br />

Columbia, SC<br />

Background: Although health information technology (HIT) has been hypothesized<br />

to help achieve greater adherence to recommended clinical guidelines<br />

and increase use <strong>of</strong> preventive care, well-conducted empirical studies<br />

evaluating this hypothesis are lacking. This study evaluated the ongoing<br />

Colorectal Cancer (CRC) Oncology Watch intervention, a clinical reminder<br />

system, implemented in 8 Veterans Affairs (VA) hospitals in 2008 for<br />

improving CRC (1) screening adherence among average- or higher-risk<br />

veterans, and (2) surveillance. Methods: VA administrative data were used<br />

to construct 4 cross-sectional groups <strong>of</strong> average-risk age 50-64 veterans,<br />

one for each <strong>of</strong> 2006, 2007, 2009, and 2010. We applied a linear<br />

probability model with hospital fixed-effects for estimation, using a natural<br />

experiment in which the 8 hospitals served as the intervention and other<br />

121 hospitals as a control and with 2006-2007 designated as the<br />

pre-intervention period and 2009-2010 as the post-intervention period.<br />

Results: The sample included 4,352,082 veteran-years in 4 years. The<br />

proportions adherent to screening were 37.6%, 31.6%, 34.4%, and<br />

33.2% in the intervention in 2006, 2007, 2009, and 2010, respectively,<br />

and the corresponding proportions in the control were 31.0%, 30.3%,<br />

32.3%, and 30.9%. Regression analysis showed that among those eligible<br />

for screening, the intervention was associated with a 2.2-percentage-point<br />

decrease in likelihood <strong>of</strong> adherence (P-value�0.0001). Additional analyses<br />

showed that the intervention was associated with a 5.6-percentagepoint<br />

decrease in likelihood <strong>of</strong> screening by colonoscopy among the<br />

adherent, but with increased total colonoscopies (any indication) <strong>of</strong> 3.6 per<br />

100 veterans age 50-64. Conclusions: The intervention may have slightly<br />

reduced CRC screening rates for the studied population. This consequence<br />

may have been caused by an unintentional shift <strong>of</strong> limited VA colonoscopy<br />

capacity from average-risk screening to higher-risk screening and to CRC<br />

surveillance, or by physicians’ fatigue due to the large number <strong>of</strong> clinical<br />

reminders implemented in the VA.<br />

6046 General Poster Session (Board #2B), Mon, 1:15 PM-5:15 PM<br />

Association <strong>of</strong> increases in quality <strong>of</strong> care with the NCI Community Cancer<br />

Center Program (NCCCP) pilot. Presenting Author: Michael T. Halpern, RTI<br />

International, Washington, DC<br />

Background: The NCCCP pilot is an initiative designed to enhance research<br />

and improve cancer care at community hospitals. As part <strong>of</strong> a multi-method<br />

evaluation <strong>of</strong> this pilot, we assessed changes in quality <strong>of</strong> care among the<br />

16 pilot NCCCP hospitals over time (before vs. after program initiation) and<br />

in comparison to a group <strong>of</strong> 25 similar hospitals that did not participate in<br />

the NCCCP. Methods: We compared changes in 5 NQF-approved quality <strong>of</strong><br />

care measures (3 for breast cancer, 2 for colon cancer) from 2006/07<br />

(before NCCCP initiation) vs. 2008/09/10 (post-initiation) for NCCCP and<br />

comparison group hospitals. Data were collected from all study hospitals<br />

using the Commission on Cancer’s Rapid Quality Reporting System, which<br />

allowed near real-time tracking <strong>of</strong> quality <strong>of</strong> care process measures. Results:<br />

Analyses included 18,608 breast cancer and 7,031 colon cancer patients.<br />

Patient-level concordance rates for all 5 quality <strong>of</strong> care measures increased<br />

significantly among both NCCCP and comparison group hospitals. The<br />

change (from baseline to post-NCCCP) in quality <strong>of</strong> care among NCCCP<br />

hospitals was significantly greater than the change among comparison<br />

group hospitals for two measures: radiation therapy following breast<br />

conserving surgery (RT-BCS) and hormonal therapy for women with<br />

hormone receptor positive breast cancer (HT). For the RT-BCS measure,<br />

NCCCP patients from underserved populations also experienced significantly<br />

greater changes in concordance than did corresponding populations<br />

from comparison group hospitals. In multivariate regression analyses<br />

controlling for patient characteristics, the change for the HT measure<br />

among NCCCP hospitals was significantly greater than that among comparison<br />

group hospitals. Conclusions: While both NCCCP and comparison group<br />

hospitals showed improved quality <strong>of</strong> care, participation in the NCCCP was<br />

associated with significantly greater improvements for a subset <strong>of</strong> measures.<br />

Including a separate comparison hospital group was critical for<br />

assessing changes associated with NCCCP participation while controlling<br />

for broader U.S. trends in improved quality <strong>of</strong> care. Future work will<br />

examine how hospital networks may have facilitated improvements in<br />

quality <strong>of</strong> care.<br />

Health Services Research<br />

393s<br />

6045 General Poster Session (Board #2A), Mon, 1:15 PM-5:15 PM<br />

Osteoporosis screening among prostate cancer survivors treated with<br />

androgen deprivation therapy. Presenting Author: Alicia Katherine Morgans,<br />

Massachusetts General Hospital Cancer Center, Boston, MA<br />

Background: Androgen deprivation therapy (ADT), the standard systemic<br />

treatment for prostate cancer, has adverse effects including bone loss and<br />

fractures. Current national guidelines suggest that men receiving ADT<br />

undergo dual energy x-ray absorptiometry (DXA) before and during ADT to<br />

better characterize fracture risk. We assessed receipt <strong>of</strong> DXA testing in a<br />

population-based cohort <strong>of</strong> men treated continuously with ADT for at least<br />

1 year and identified factors associated with testing. Methods: Using<br />

Surveillance, Epidemiology, and End Results-Medicare data, we identified<br />

men aged �65 with local or regional prostate cancer diagnosed during<br />

2001-2007 and followed through 2009 who received at least 1 year <strong>of</strong><br />

continuous ADT. We identified receipt <strong>of</strong> DXA testing in the 18-month<br />

period beginning 6 months before the first dose <strong>of</strong> ADT. We used logistic<br />

regression to identify factors associated with DXA testing, including patient<br />

and tumor characteristics and the physicians with whom they had outpatient<br />

visits. Results: Among 28,960 men treated with ADT for �1 year,<br />

6.5% had at least one DXA scan from 6 months before the first dose <strong>of</strong> ADT<br />

through 1 year after. DXA testing increased over time, with men initiating<br />

ADT in 2007-2009 more likely to be tested than those treated in<br />

2001-2002 (10.0% vs. 3.4%, OR 2.29, 95% CI 1.83-2.85). Men aged<br />

�85 were less likely than men aged 66-69 to undergo testing (OR 0.76,<br />

95% CI 0.65-0.89). Black men were less likely than white men to undergo<br />

testing (OR 0.92, 95% CI 0.61-0.86), as were men living in areas with<br />

lower educational attainment (P�.001). Compared with men seeing a<br />

urologist but no medical oncologist or primary care provider (PCP), men<br />

seeing a medical oncologist and a urologist (OR 2.11, 95% CI 1.39-3.21)<br />

and those seeing a medical oncologist, urologist and PCP (OR 2.59, 95%<br />

CI 2.01-3.34) had higher odds <strong>of</strong> testing. Conclusions: Few men receiving<br />

ADT for prostate cancer undergo DXA testing, with particularly low rates <strong>of</strong><br />

testing among older men, black men, and those living in areas with low<br />

educational attainment. Visits with a medical oncologist were associated<br />

with increased odds <strong>of</strong> testing. Interventions are needed to increase bone<br />

density testing among men receiving long-term ADT.<br />

6047 General Poster Session (Board #2C), Mon, 1:15 PM-5:15 PM<br />

Geographic and network analysis <strong>of</strong> oncology trials: Portfolio assessment <strong>of</strong><br />

<strong>Clinical</strong>Trials.gov. Presenting Author: Steven Kunyuan Cheng, Oregon<br />

Health & Science University, Portland, OR<br />

Background: Increasing complexity and specificity <strong>of</strong> cancer trials (CT)<br />

necessitates collaboration across the CT network to optimize research<br />

efforts. The relationship among CT sites and networks provide unique<br />

insights into improving coordination and accrual. Methods: 5971 interventional<br />

CTs registered between 2007 and 2010 were aggregated by trial and<br />

location. CTs in the Aggregate Analysis dataset from <strong>Clinical</strong>Trials.gov<br />

(AACT) were identified using MeSH terms. The distribution <strong>of</strong> mid-phase<br />

(MP)(phase I/II,II) and late-phase (LP)(PII/III,PIII) CTs for the ten most<br />

represented cancer types by number <strong>of</strong> sites was assessed using network<br />

graph theory and geographic analysis comparing distribution <strong>of</strong> trials across<br />

metropolitan statistical areas. Results: 66,566 CT sites were identified<br />

across the sample, 59.6% were in the United States (MP: 50.2%; LP:<br />

42.9%). Geographical availability <strong>of</strong> CTs and local cancer incidence rates<br />

were highly correlated (0.797, p� 0.001) but varied depending upon<br />

disease type. Network density (the degree <strong>of</strong> dyadic interconnections<br />

between sites studying similar cancer types) showed overall that MP trials<br />

were less dense with sparser interconnections among sites than LP trials.<br />

Network density <strong>of</strong> LP trials was higher for cancer types that had poorer<br />

correlation between geographic distribution <strong>of</strong> incidence and enrolling sites<br />

(-0.777, p�0.008). MP trials did not show a similar trend. Conclusions:<br />

The relationship between the distribution <strong>of</strong> CT and site location can be<br />

envisaged through geographic and network analysis <strong>of</strong> CT registry data. LP<br />

high-density networks should strive to diversify trial locations to better meet<br />

regional incidence rates.<br />

Incidence and trials MP (mid-phase) LP (late-phase)<br />

Pearson Corr.<br />

(all p


394s Health Services Research<br />

6048 General Poster Session (Board #2D), Mon, 1:15 PM-5:15 PM<br />

Electronic prompt to improve outpatient code status documentation for<br />

advanced lung cancer. Presenting Author: Jennifer S. Temel, Massachusetts<br />

General Hospital Cancer Center, Boston, MA<br />

Background: Rates <strong>of</strong> documentation <strong>of</strong> end-<strong>of</strong>-life care preferences in the<br />

medical record remain low, even among patients with incurable malignancies.<br />

The goal <strong>of</strong> this study was to assess the impact <strong>of</strong> electronic prompts<br />

to encourage oncology clinicians to document code status in the outpatient<br />

electronic health record (EHR) <strong>of</strong> patients with advanced lung cancers.<br />

Methods: We conducted two clinician focus groups (n�15) at an affiliated<br />

academic medical center to determine the appropriate content and timing<br />

<strong>of</strong> the electronic reminders. Based on the focus groups, we developed<br />

email reminders that were timed to the start <strong>of</strong> each new chemotherapy<br />

regimen. Between 6/09 and 1/11, 102 eligible patients with advanced<br />

lung cancer were approached, and 100 (98%) agreed to participate in the<br />

prospective study. Email reminders were sent to oncology clinicians at the<br />

patient’s next outpatient visit and with each new chemotherapy regimen.<br />

Using a pre-post design, we compared study participants to a retrospective<br />

cohort <strong>of</strong> 100 consecutive historical controls who began chemotherapy for<br />

advanced lung cancer at least one year prior to the start <strong>of</strong> this study. The<br />

primary outcome measure was the documentation <strong>of</strong> code status in the<br />

EHR. Results: Study participants were similar to historical controls, with no<br />

significant differences in age, gender, performance status, histology or<br />

initial cancer therapy received. At one year follow-up, 33/98 (34%) <strong>of</strong><br />

participants had a code status documented in the outpatient EHR compared<br />

with 12/83 (15%) <strong>of</strong> historical controls, p�0.003. Mean time to<br />

code status documentation was significantly shorter in study participants<br />

(8.6 months [95% CI 7.6-9.5]) compared with controls (10.5 months<br />

[95% CI 9.8-11.3]), p�0.004. Conclusions: Email prompts triggered by<br />

changes in chemotherapy improved the rate and timing <strong>of</strong> code status<br />

documentation in the EHR.<br />

6050 General Poster Session (Board #2F), Mon, 1:15 PM-5:15 PM<br />

How do social factors explain outcomes in non-small cell lung cancer<br />

among Hispanics/Latinos in California? Presenting Author: Manali I. Patel,<br />

Stanford University Medical Center, Palo Alto, CA<br />

Background: Hispanics in the United States have a lower age-adjusted<br />

incidence and mortality rate from non-small cell lung cancer compared<br />

with non-Hispanic whites. Previous studies have demonstrated the influence<br />

<strong>of</strong> nativity on survival among Hispanic patients but no studies have<br />

evaluated the interplay <strong>of</strong> nativity, clinical factors, social factors, and<br />

neighborhood factors on survival among Hispanic patients with non-small<br />

cell lung cancer. Methods: All Hispanic patients with non-small cell lung<br />

cancer between the years <strong>of</strong> 1988-2008 were identified in the California<br />

Cancer Registry (CCR). Kaplan Meier curves depict survival by nativity<br />

status among Hispanics with non-small cell lung cancer. Cox proportional<br />

hazard models estimate the hazard <strong>of</strong> mortality by race with adjustment for<br />

individual covariates (age, gender, marital status), clinical factors (histologic<br />

grade, surgery, radiation, and chemotherapy), and social and neighborhood<br />

factors (neighborhood and ethnic enclave status). Results: A total <strong>of</strong><br />

4,062 Hispanic patients with non small cell lung cancer were included.<br />

Overall, there was a 7% decreased risk <strong>of</strong> disease-specific mortality for<br />

foreign-born patients as compared with US-born patients (HR 0.93,<br />

p�0.08, 95% CI 0.87-1.00) although not-statistically significant. Adjustment<br />

for individual patient factors and clinical factors conferred a statistically<br />

significant 16% decreased risk <strong>of</strong> disease-specific mortality compared<br />

with US-born patients (HR 0.84, p�0.0001, 95% CI 0.78-0.91). Adjustment<br />

for socioeconomic status and neighborhood socioeconomic and<br />

ethnic enclave status did not explain the differences in survival (HR 0.84, p<br />

�0.001, 95% CI 0.78-0.91). Conclusions: Overall, foreign-born Hispanics<br />

with non-small cell lung cancer have a decreased risk <strong>of</strong> disease-specific<br />

mortality compared with US-born Hispanics with non-small cell lung<br />

cancer but social factors do not explain this survival advantage. Further<br />

investigation is needed to understand the drivers <strong>of</strong> the survival advantage<br />

outcomes in foreign-born populations.<br />

6049 General Poster Session (Board #2E), Mon, 1:15 PM-5:15 PM<br />

Positron emission tomography/computed tomography (PET/CT) for the<br />

diagnosis <strong>of</strong> recurrent cancer (PETREC): A multicenter, prospective cohort<br />

study. Presenting Author: John J. You, McMaster University, Hamilton, ON,<br />

Canada<br />

Background: The clinical utility <strong>of</strong> PET/CT in patients with suspected cancer<br />

recurrence remains unclear. The aim <strong>of</strong> this multi-center, prospective,<br />

comparative effectiveness study is to assess the impact <strong>of</strong> PET/CT on<br />

clinical management <strong>of</strong> patients with suspected cancer recurrence. Methods:<br />

Patients were eligible if cancer recurrence (non-small cell lung, breast,<br />

head and neck, ovarian, esophageal, Hodgkin’s or non-Hodgkin’s lymphoma)<br />

was clinically suspected, and if conventional imaging (e.g. X-ray,<br />

ultrasound, CT, or MRI) was non-diagnostic. As a pre-requisite to PET/CT<br />

booking, clinicians were asked at enrolment to indicate their planned<br />

management if PET/CT were not available. Patients then underwent<br />

18FDG-PET/CT. Clinicians were then asked to indicate their management<br />

plan based on PET/CT findings. Patients were followed up once at 3<br />

months. The primary outcome was change in planned management after<br />

PET/CT and was assessed independently and in duplicate by external<br />

outcome adjudicators using all available source documents. Results: 101<br />

patients (mean age 64 y, 45% male, median 1.3 y since last treatment)<br />

were enrolled from 4 centers in Ontario, Canada between April 2009 and<br />

June 2011. Distribution <strong>of</strong> tumor types was: non-small cell lung (55%),<br />

breast (19%), ovarian (10%), esophageal (6%), lymphoma (6%), head and<br />

neck (4%). 8 patients did not complete the study (non-adherence to<br />

protocol, 2; death, 5; disease progression prior to PET/CT, 1), <strong>of</strong> whom 2<br />

did not receive PET/CT. PET/CT changed planned management in 52<br />

(53%) patients (Table). At 3 months, planned management was carried out<br />

in 46/52 (88%) patients. Conclusions: In patients with suspected cancer<br />

recurrence, PET/CT changes planned management from non-treatment to<br />

treatment for approximately 1 in every 3 patients (“number needed to<br />

scan” � 3) and contributes importantly to clinical management.<br />

Impact on planned management<br />

PET/CT for suspected<br />

cancer recurrence (N�99)<br />

No change 47 (47)<br />

Major change*<br />

Minor change<br />

38 (38)<br />

<strong>Clinical</strong>/imaging follow-up to biopsy 9 (9)<br />

Imaging to clinical follow-up 2 (2)<br />

Biopsy to clinical/imaging follow-up<br />

Data are n (%). * No treatment to treatment.<br />

3 (3)<br />

6051 General Poster Session (Board #2G), Mon, 1:15 PM-5:15 PM<br />

Improving the impact <strong>of</strong> clinical research: A systematic analysis <strong>of</strong> kidney<br />

cancer trials. Presenting Author: Bradford Richard Hirsch, Duke Cancer<br />

Institute, Durham, NC<br />

Background: <strong>Clinical</strong> trials are essential to advancing cancer care, but the<br />

means to evaluate and improve the portfolio have been lacking. This study<br />

analyzes the renal cell carcinoma (RCC) trial portfolio using the database for<br />

the Aggregate Analysis <strong>of</strong> <strong>Clinical</strong>Trials.gov. As RCC is an area <strong>of</strong> promise, what<br />

insights can an evaluation <strong>of</strong> the portfolio provide? Methods: 40,970 clinical<br />

studies registered with <strong>Clinical</strong>Trials.gov between October 2007 and September<br />

2010 were aggregated by specialty using MeSH terms and submitted<br />

conditions. 8,942 oncology trials were identified and categorized by cancer<br />

type. 108 trials opening in October 2007 or later were identified as evaluating<br />

treatments for RCC. Descriptive statistics were used to characterize trial<br />

design, study agent(s), accrual and sponsorship. Results: 52 trials (48%)<br />

assessed agents already recommended as 1st or 2nd-line treatments in the<br />

National Comprehensive Cancer Network (NCCN) RCC Guidelines at the time<br />

<strong>of</strong> study initiation and 19 (18%) studied other FDA-approved treatments. 37<br />

trials (34%) included a novel (non-FDA approved) agent or vaccine. Total<br />

anticipated or actual accrual was 12,753, with 50% accrued/accruing to trials<br />

<strong>of</strong> only NCCN agents, 34% to novel agents and 16% to other FDA-approved<br />

agents. Industry was identified as a sponsor or collaborator in 48% <strong>of</strong> trials<br />

assessing only NCCN agents, 73% <strong>of</strong> novel agent trials and 53% <strong>of</strong> other<br />

FDA-approved agent trials. As shown in the table, a minority <strong>of</strong> trials were<br />

randomized, blinded or late-phase (Phase III or IV), regardless <strong>of</strong> approval<br />

status <strong>of</strong> study agent. Conclusions: The majority <strong>of</strong> new studies and accrual in<br />

RCC assess questions <strong>of</strong> treatment sequence and setting for established<br />

therapies, many <strong>of</strong> which lack rigorous design. Across the portfolio, studies are<br />

predominantly industry sponsored. Optimizing clinical research includes<br />

increasing studies <strong>of</strong> novel therapeutics and improving the comparative<br />

effectiveness research portfolio by increasing utilization <strong>of</strong> pragmatic designs,<br />

registries and late-phase programs.<br />

Trials Randomized Blinded Phase III or IV<br />

Agent<br />

n % n % n % n %<br />

NCCN-recommended 52 48% 17 33% 3 6% 7 13%<br />

Other FDA-approved 19 18% 3 16% 0 0% 1 5%<br />

Novel 37 34% 9 24% 3 8% 4 11%<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


6052 General Poster Session (Board #3A), Mon, 1:15 PM-5:15 PM<br />

Influence <strong>of</strong> comorbidity on guideline concordant care for breast cancer:<br />

Findings from the Center for Disease Control and Prevention National<br />

Program <strong>of</strong> Cancer Registry (NPCR) patterns <strong>of</strong> care study. Presenting<br />

Author: Gretchen Genevieve Kimmick, Duke University Medical Center,<br />

Durham, NC<br />

Background: Comorbidity burden predicts cancer treatment and may<br />

influence outcome. We explore the relationship <strong>of</strong> specific comorbid<br />

illnesses with receipt <strong>of</strong> guideline concordant care for early stage breast<br />

cancer. Methods: The NPCR’s Patterns <strong>of</strong> Care study reabstracted the<br />

medical records <strong>of</strong> breast cancer cases diagnosed in 2004 from 7 cancer<br />

registries. We included women with nonmetastatic in situ and invasive<br />

breast cancer, known hormone receptor status, node status, and tumor<br />

size. Guideline-concordant management, including surgery, radiation,<br />

chemotherapy and endocrine components, was based on NCCN guidelines<br />

using tumor size, nodal and hormone receptor status. Comorbidity was<br />

measured according to the Adult Comorbidity Evaluation Index (ACE).<br />

Multivariate logistic regression models were used to determine factors<br />

associated with guideline-concordant care, and included overall ACE<br />

scores and 26 separate ACE comorbidity categories, as well as age, race,<br />

hormone receptor status, and HER2 status. Results: The study sample<br />

included 6904 women (mean age 58.7 and range 20-99 years, 76% white,<br />

45% with ACE comorbidity score <strong>of</strong> 0, 70% ER and/or PR�, 13%<br />

HER2�). Overall, 64% received guideline-concordant care. Receipt <strong>of</strong><br />

guideline-concordant care varied by overall comorbidity burden (71% for<br />

none; 65% for minor; 63% for moderate; 50% for severe; p�0.05). The<br />

presence <strong>of</strong> hypertension (OR 1.26, 95% CI 1.08-1.48) predicted receipt<br />

<strong>of</strong> guideline concordant care, whereas, peripheral artery disease (OR 0.44,<br />

95% CI 0.21-0.93), diabetes (OR 0.78, 95% CI 0.63-0.97) and dementia<br />

(OR 0.31, 95% CI 0.13-0.74) predicted lack <strong>of</strong> guideline concordant care.<br />

Older age, black race, and hormone receptor positivity were associated with<br />

less, and HER2 positivity with receipt <strong>of</strong> more guideline-concordant care.<br />

Conclusions: Overall those with more comorbidity burden received less<br />

guideline-concordant care. However, the effects vary by specific conditions.<br />

The odds <strong>of</strong> receiving guideline-concordant care was greater in those<br />

with hypertension and less in those with peripheral arterial disease,<br />

diabetes, and dementia.<br />

6054 General Poster Session (Board #3C), Mon, 1:15 PM-5:15 PM<br />

A brief educational intervention to increase communication about complementary<br />

and alternative medicine (CAM) in community oncology settings.<br />

Presenting Author: Lorenzo Cohen, University <strong>of</strong> Texas M. D. Anderson<br />

Cancer Center, Houston, TX<br />

Background: The widespread use <strong>of</strong> CAM in academic oncology settings has<br />

been well documented. However, there is a lack <strong>of</strong> communication between<br />

patients and health care pr<strong>of</strong>essionals on CAM that may have negative<br />

health consequences. No comprehensive study <strong>of</strong> CAM use in community<br />

oncology settings exists. We examined the benefits <strong>of</strong> a brief educational<br />

intervention on nurse discussions <strong>of</strong> CAM with patients. Methods: A<br />

multi-site, randomized trial <strong>of</strong> an educational intervention (brief video,<br />

resource list) designed to encourage oncology nurses to discuss CAM use<br />

with their patients was conducted within the MD Anderson CCOP network.<br />

Nurses (N�175) and patients (baseline N�699 and different set <strong>of</strong><br />

patients after intervention N�650) completed questions about CAM use,<br />

communication, and knowledge. Results: Nurses were 97% female, 96%<br />

non-Hispanic white. Two months after the intervention, nurses in the<br />

intervention group reported that they were more likely to ask about CAM use<br />

than those in the control group (OR�4.2; p�.005) and asked more <strong>of</strong> their<br />

last 5 patients about CAM use (p�.003). No significant intervention effect<br />

was found for the proportion <strong>of</strong> patients in the clinic who indicated they<br />

were asked about CAM use after the intervention (OR�1.6, p�.10).<br />

Approximately 40% <strong>of</strong> patients reported using CAM following their cancer<br />

diagnosis yet the majority <strong>of</strong> nurses estimated less than 25% <strong>of</strong> their<br />

patients used CAM. Conclusions: CAM use in community-based oncology<br />

patients is high and there is an underestimation <strong>of</strong> use by the oncology<br />

nurses. This very brief intervention significantly improved how <strong>of</strong>ten nurses<br />

reported asking patients about their CAM use. However, the patients <strong>of</strong> the<br />

nurses did not reflect this change in communication. Additional types <strong>of</strong><br />

interventions are needed to increase communication between patients and<br />

nurses.<br />

Health Services Research<br />

395s<br />

6053 General Poster Session (Board #3B), Mon, 1:15 PM-5:15 PM<br />

Examining patient choices for metastatic breast cancer drugs. Presenting<br />

Author: Mary Lou Smith, Research Advocacy Network, Plano, TX<br />

Background: Patients with metastatic breast cancer face difficult drug<br />

decisions. Our previous research (ASCO Proc 2011, abstr 6044) focused<br />

on general benefit and toxicity showed that conjoint analysis (CA) allows<br />

patients to express preferences; our current research quantifies patient<br />

preference for specific drug pr<strong>of</strong>iles (capecitabine and paclitaxel). Methods:<br />

Research Advocacy Network and CBWhite conducted research using CA for<br />

DOD Center <strong>of</strong> Excellence for Individualization <strong>of</strong> Therapy in Breast Cancer.<br />

An online survey was sent by four breast cancer organizations (N�641).<br />

Questions elicited views on trade-<strong>of</strong>fs between benefit and type/severity/<br />

duration <strong>of</strong> toxicity. CA questions present pairs <strong>of</strong> hypothetical treatments<br />

and ask respondents their preferred alternative; a follow-up question asks<br />

whether the person would take the treatment if it were the only option<br />

available. Analysis <strong>of</strong> response patterns allows study <strong>of</strong> treatment preferences<br />

for combinations <strong>of</strong> benefit and described toxicity. Results: See table.<br />

Preferences show much greater attention to benefit than to toxicity. When<br />

CA is used to examine impact <strong>of</strong> biomarkers, focus on benefit continues.<br />

Paclitaxel pr<strong>of</strong>ile (IV) set with moderate PN lasting 1 year post treatment:<br />

with 33% benefit LH, 6% <strong>of</strong> respondents change treatment decision if<br />

biomarker predicts 27% vs 60% toxicity likelihood; with 27% toxicity LH,<br />

22% <strong>of</strong> respondents change treatment decision if biomarker predicts 20%<br />

vs 50% benefit likelihood. Conclusions: For patients with metastatic<br />

disease, CA shows much greater attention to benefit than toxicity, and high<br />

likelihood to take treatment with at least 30% chance <strong>of</strong> benefit for any<br />

toxicity tested here. These results suggest biomarkers (for the pr<strong>of</strong>iled<br />

drugs) predicting benefit are more likely to be used to affect patient<br />

treatment decisions than biomarkers for toxicity.<br />

Prediction <strong>of</strong> % selecting treatment with benefit and toxicity likelihood (LH)<br />

under two scenarios (N�641).<br />

10%<br />

Toxicity LH<br />

20% 40% 60%<br />

IV drug pr<strong>of</strong>ile—moderate peripheral neuropathy (PN) for 1 year<br />

Benefit LH<br />

20% N/A 77.4 72.3 65.3<br />

30% N/A 89.8 86.4 81.6<br />

50% N/A 97.1 96.1 94.3<br />

Oral drug pr<strong>of</strong>ile—moderate Hand-Foot Syndrome during treatment<br />

10% 67.6 65.2 60.8 N/A<br />

30% 94.7 93.0 91.4 N/A<br />

50% 99.0 98.3 97.4 N/A<br />

6055 General Poster Session (Board #3D), Mon, 1:15 PM-5:15 PM<br />

Self-reported conflicts <strong>of</strong> interest (sfCOI) <strong>of</strong> authors and the interpretation<br />

<strong>of</strong> randomized phase III trials (RCT) and related editorials (REd) in cancer<br />

research. Presenting Author: Giovanni Mendonca Bariani, Instituto do<br />

Cancer do Estado de Sao Paulo, Sao Paulo, Brazil<br />

Background: Growing participation from industry in cancer research has<br />

resulted in increased reporting <strong>of</strong> COI. We aimed to test any association<br />

between author’s conclusion and sfCOI in cancer studies. Methods: All RCT<br />

and REd published in 6 major cancer journals in a 3.5 year period were<br />

selected. Two investigators blinded to COI disclosure independently<br />

analyzed each RCT and REd, classifying authors’ conclusions as highly<br />

positive, positive, neutral, negative, and highly negative with respect to<br />

author’s opinion on the experimental therapy. The agreement rate between<br />

investigators for conclusion classification was 90% (consensus was achieved<br />

for the remaining 10%). We also collected data on study results, COI and<br />

sponsorship. COI was defined as any self-reported financial tie between<br />

author and industry except for research funds. Predictors <strong>of</strong> positive/highly<br />

positive conclusions <strong>of</strong> RCT and <strong>of</strong> REd were tested separately in logistic<br />

regression multivariable models. Results: From Jan 2008 to Oct 2011,<br />

1,485 articles were retrieved: 150 RCT and 140 REd were eligible. Among<br />

the RCT, 82 (55%) were positive, and 78 (52%) were entirely or partially<br />

funded by industry. Any sfCOI was present in 103 (69%) RCT and in 71<br />

(47%) REd. Conclusions <strong>of</strong> REd and RCT were: 7.3% and 11.3% highly<br />

positive, 42.7% and 57.3% positive, 8.0% and 2.0% neutral, 29.3% and<br />

18.7% negative, and 12.7% and 10.7% highly negative, respectively.<br />

Multivariable analysis showed that RCT positive result was the only<br />

significant predictor for positive conclusion by RCT authors (OR�109,<br />

95% CI: 21-567; p�0.001). The only factor associated with positive<br />

conclusions <strong>of</strong> REd authors was a positive conclusion by RCT author<br />

(OR�42, 95% CI: 7-244; p�0.001). While 64 (43%) RCT reported<br />

negative results, 103 (68.7%) RCT authors interpreted studies positively.<br />

Logistic regression for discordance between RCT result and RCT conclusion<br />

did not find any association with COI. Conclusions: Theinterpretation <strong>of</strong> RCT<br />

results by authors was not influenced by sfCOI or trial sponsorship. Authors<br />

<strong>of</strong> REd were not influenced by study results or by their sfCOI when<br />

discussing cancer RCT.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


396s Health Services Research<br />

6057 General Poster Session (Board #3F), Mon, 1:15 PM-5:15 PM<br />

The cost-effectiveness <strong>of</strong> improving cancer screening compliance. Presenting<br />

Author: Michael S. Broder, <strong>Part</strong>nership for Health Analytic Research,<br />

LLC, Beverly Hills, CA<br />

Background: Expensive treatments and a growing number <strong>of</strong> cancer patients<br />

have resulted in increased spending on cancer care. Within a framework we<br />

developed for measuring the value <strong>of</strong> quality improvement (QI), we describe<br />

the cost-effectiveness <strong>of</strong> improving compliance with cancer screening<br />

measures compared to other quality measures. Methods: We used our<br />

framework to examine 18 Healthcare Effectiveness Data and Information<br />

Set (HEDIS) 2010 quality measures, synthesize related cost-effectiveness<br />

(CE) data and describe measure-specific QI-adjusted incremental costeffectiveness<br />

ratios (ICERs). For each measure we: 1) quantified current<br />

compliance; 2) reviewed literature for ICERs; 3) estimated per-person<br />

steady state cost and quality-adjusted life years (QALYs); 4) estimated<br />

affected population size; 5) estimated the cost <strong>of</strong> QI; and 6) calculated<br />

QI-adjusted ICERs at full compliance, defined as 95%. We assumed<br />

per-person QI costs did not change with compliance and varied this in<br />

sensitivity analyses. We compared QI-adjusted ICERs for 3 cancer screening<br />

measures to the remaining measures. Results: Published ICERs for the<br />

cancer screening measures were $43,180/QALY (breast), $5,102/QALY<br />

(cervix) and $15,173/QALY (colon) and for other measures from $195/<br />

QALY (drug treatment) to $35,616/QALY (flu shots). Incorporating QI costs<br />

for cancer screening measures gave QI-adjusted ICERs <strong>of</strong> $64,549/QALY<br />

(breast), $15,463/QALY (cervix) and $22,991/QALY (colon), respectively.<br />

Incorporating QI costs for all 18 measures resulted in QI-adjusted ICERs<br />

from $195/QALY (drug treatment) to $9,075,868/QALY(antidepressant<br />

management), with a median <strong>of</strong> $15,463/QALY. Reaching 95% compliance<br />

with the 3 cancer measures would cost $5.1 billion and add 160,000<br />

QALYs ($32,640/QALY) and with all 18 measures would cost $13.4 billion<br />

and add 5.8 million QALYs ($2,313/QALY). Conclusions: Improving compliance<br />

with cancer screening may be cost-effective at a threshold <strong>of</strong><br />

$50k/QALY, although improving care on all HEDIS measures may be even<br />

more cost-effective. Accurate assessment <strong>of</strong> the cost <strong>of</strong> increasing cancer<br />

screening requires integration <strong>of</strong> both the cost-effectiveness <strong>of</strong> the screening<br />

tests and the cost <strong>of</strong> the QI programs needed to change practice.<br />

6059 General Poster Session (Board #3H), Mon, 1:15 PM-5:15 PM<br />

Health care costs associated with venous thromboembolism in select<br />

high-risk ambulatory solid tumor patients in the United States. Presenting<br />

Author: Alok A. Khorana, University <strong>of</strong> Rochester, Rochester, NY<br />

Background: Venous thromboembolism (VTE) is an increasingly common<br />

complication <strong>of</strong> cancer and its treatment, including chemotherapy. VTE<br />

has significant clinical consequences, including mortality. However, contemporary<br />

data on the healthcare costs <strong>of</strong> cancer-associated VTE are<br />

limited. We examined the real-world economic burden <strong>of</strong> VTE in ambulatory<br />

patients initiating chemotherapy for select common high-risk solid tumors.<br />

Methods: Healthcare claims data (2004-2009) from the IMS/PharMetrics<br />

Patient-Centric database were collected for propensity score-matched<br />

adult cancer (lung, colorectal, pancreas, stomach, bladder and ovary)<br />

patients with VTE (n�912) and without VTE (n�2,736) after initiating<br />

chemotherapy. Healthcare resource utilization (inpatient, outpatient medical,<br />

and outpatient prescription drug claims) and costs were compared<br />

between the two cohorts during 12 months’ follow-up after the index VTE<br />

event. Incremental costs <strong>of</strong> VTE were adjusted for demographic and clinical<br />

covariates. Results: Cancer patients with VTE had ~3-times as many<br />

all-cause hospitalizations (mean 1.38 vs. 0.55 per patient) and days in<br />

hospital (10.2 vs. 3.4 per patient), and more outpatient claims (331 vs.<br />

206 per patient) than matched cancer patients without VTE (all P�0.0001).<br />

Cancer patients with VTE incurred significantly higher overall (all-cause)<br />

inpatient costs (mean $21,299 vs. $7,459 per patient), outpatient costs<br />

(mean $53,660 vs. $34,232 per patient) and total healthcare costs (mean<br />

$74,959 vs. $41,691 per patient) than cancer patients without VTE (all<br />

P�0.0001). Total VTE-related healthcare costs were (mean) $9,247 per<br />

VTE patient over 12 months. Adjusted incremental all-cause healthcare<br />

costs <strong>of</strong> VTE were (mean) $30,538 per patient across the selected tumors,<br />

ranging from $11,946 per patient for gastric cancer to $38,983 per<br />

patient for pancreatic cancer. Conclusions: VTE results in significant<br />

inpatient and outpatient resource utilization, and increased all-cause (in<br />

addition to VTE-related) healthcare costs. Measures to prevent outpatient<br />

cancer-associated VTE may reduce healthcare utilization and costs in<br />

high-risk cancer patients.<br />

6058 General Poster Session (Board #3G), Mon, 1:15 PM-5:15 PM<br />

Treatment and treatment outcomes <strong>of</strong> advanced NSCLC patients in routine<br />

clinical care: Results <strong>of</strong> the retrospective LUCEOR study. Presenting<br />

Author: Jason Scott Agulnik, Jewish General Hospital, Lady Davis Institute<br />

for Medical Research, McGill University, Montreal, QC, Canada<br />

Background: Although NSCLC is the most common type <strong>of</strong> lung cancer,<br />

published data on both treatment patterns and outcomes in routine clinical<br />

care are scarce. Collecting this data systematically across countries will<br />

help to understand, compare and improve patient care. Methods: 1,490<br />

patient charts were reviewed retrospectively from 40 sites in Australia,<br />

Belgium, Canada, France, Germany, Italy, Sweden, Turkey, the Netherlands<br />

and the UK, 1436 <strong>of</strong> those evaluable. Patients deceased before April<br />

2010 were eligible if �18 years, diagnosed with advanced stage IIIb/IV<br />

NSCLC and had received active 1st line anti-cancer treatment. Demographic<br />

data, disease and, treatment history and resource use were<br />

collected from start <strong>of</strong> first line treatment until death. Results: Mean age at<br />

NSCLC diagnosis was 64 years [SD�10.6], 67% <strong>of</strong> patients had stage IV<br />

disease at diagnosis and 66% were male. Confirmed tumor histology was<br />

adenocarcinoma in 50% <strong>of</strong> patients, squamous and large cell carcinoma in<br />

23% and 14%, respectively. Tumour histology was not available in 13% <strong>of</strong><br />

patients. Mean number <strong>of</strong> treatment lines was 1.7 [range 1-8]. Biomarker<br />

analysis was performed in 19% <strong>of</strong> patients (<strong>of</strong> those 26% and 13% tested<br />

for EGFR and KRAS mutation, respectively) within less than one month <strong>of</strong><br />

diagnosis. 1st line therapy included mainly platinum doublets (86%,<br />

mostly carboplatine/gemcitabine [23%]), 2nd line included erlotinib (35%)<br />

and pemetrexed (18%), and 3rd line included erlotinib (38%) and<br />

docetaxel (17%). Most treated patients had ECOG performance status <strong>of</strong> 1.<br />

Stable disease (based on RECIST) was highest during 1st line therapy with<br />

30%. Median PFS from start <strong>of</strong> 1st line treatment was 3.8 months<br />

[SD�7.1], with a median OS <strong>of</strong> 7.1 months [SD�11.6]. Conclusions: The<br />

majority <strong>of</strong> patients were diagnosed with an adenocarcinoma and a number<br />

<strong>of</strong> patients were tested for EGFR or KRAS mutation. Overall treatment<br />

outcomes observed in routine care seem lower than those observed in<br />

clinical trials. Also current therapeutic options for NSCLC seem to provide<br />

limited clinical benefit, underpinning the need for new treatment alternatives<br />

to prolong PFS and OS.<br />

6060 General Poster Session (Board #4A), Mon, 1:15 PM-5:15 PM<br />

Use and spending on antineoplastic therapy (AT) in the Medicare population<br />

and the role <strong>of</strong> supplemental coverage. Presenting Author: Amy J.<br />

David<strong>of</strong>f, Marlene and Stewart Greenebaum Cancer Center, Baltimore, MD<br />

Background: Oral antineoplastic agents (OAA) represent a growing sector <strong>of</strong><br />

cancer therapy. Prior studies <strong>of</strong> older adult cancer patients (pts) examined<br />

use <strong>of</strong> infused/ injected chemotherapy (IICT) covered by Medicare <strong>Part</strong> B.<br />

We examined trends in IICT and OAA use, and the effect <strong>of</strong> supplemental<br />

coverage on AT use and spending. Methods: We used Medicare Current<br />

Beneficiary Survey data (1997-2007). Newly diagnosed cancer pts were<br />

selected using ICD-9CM codes. IICT use was identified from <strong>Part</strong> B claims;<br />

OAAs were identified from self reported prescription medication events.<br />

Supplemental coverage and pt characteristics were assessed from administrative<br />

records and self-report. Total spending summed payments from all<br />

sources during the cancer diagnosis and subsequent year. Logistic regression<br />

examined factors associated with use <strong>of</strong> any and type <strong>of</strong> AT;<br />

Generalized Linear Models estimated factors associated with spending.<br />

Results: Of 1,836 newly diagnosed cancer pts 559 (31%) received AT; 395<br />

(21%) used IICT and 253 (15%) used OAAs. Spending per user was<br />

$7,544(AT), $9,892 (IICT), and $1,535 (OAA). Supplemental coverage<br />

was associated with increased odds <strong>of</strong> AT use compared to no supplemental<br />

coverage (see Table). Cancer site and age were key predictors <strong>of</strong> spending<br />

among users. OAA spending increased during 2006-7 relative to 2004-5.<br />

Conclusions: AT use in Medicare beneficiaries is sensitive to the presence<br />

but not type <strong>of</strong> supplemental coverage. OAAs were used by a relatively large<br />

proportion <strong>of</strong> cancer pts receiving AT, although spending was less than for<br />

IICT during this largely pre-<strong>Part</strong> D period. With the growing number <strong>of</strong><br />

relatively new OAAs, and more in the pipeline, monitoring the role <strong>of</strong><br />

supplemental coverage, and particularly the role <strong>of</strong> <strong>Part</strong> D on access and<br />

spending is a critical area for ongoing research.<br />

Adjusted effects <strong>of</strong> supplemental coverage on AT use.<br />

AT IICT OAA<br />

(OR)<br />

Supplemental coverage (ref�none)<br />

Employment related w/ Rx 1.92** 1.88** 2.25**<br />

Other private w/ Rx 1.93** 1.79* 2.20**<br />

Public w/ Rx 1.85** 1.86** 2.00*<br />

Medical only 1.92** 1.85** 2.33**<br />

OR signif @ **p�.05; * p�.10. Models controlled for site, demographics, health<br />

status, income, region, year, and months observed.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


6061 General Poster Session (Board #4B), Mon, 1:15 PM-5:15 PM<br />

Disparities in preoperative imaging for breast cancer: Not so black and<br />

white. Presenting Author: Richard J. Bleicher, Fox Chase Cancer Center,<br />

Philadelphia, PA<br />

Background: Controversy exists about racial/ethnic differences in the care <strong>of</strong><br />

breast cancer patients. Imaging plays a critical role in evaluation, but little<br />

national data exists on breast imaging by race. This study was performed to<br />

determine if imaging disparities exist in Medicare patients, when adjusting<br />

for socioeconomic (SE) factors. Methods: Surveillance Epidemiology and<br />

End Results (SEER) data linked to Medicare claims were reviewed for<br />

patients diagnosed with invasive nonmetastatic breast cancer between<br />

2000 and 2005, undergoing surgery as their 1st treatment modality.<br />

Diagnostic imaging was reviewed during the interval between the 1st physician visit and surgery (“preoperative interval”[POI]) and imaging use<br />

was defined as �1 imaging-specific claim. SE factors included rural/urban<br />

setting, and education, poverty, and median income per census tract.<br />

Results: Among 39,790 patients, there were 34,774 White (87%), 2,341<br />

Black (5.9%), 1,459 Hispanic (3.7%), and 1,216 Asian/Pacific Islander<br />

(3.1%) patients. Univariate racial differences were noted for all imaging<br />

types (p’s �0.0001 to 0.007). During the POI, when adjusting for<br />

demographics, tumor characteristics, and SE factors in multivariable<br />

analysis, use <strong>of</strong> mammogram, ultrasound (US), breast MRI, CT, and bone<br />

scan were not different between Blacks and Whites. Education level was a<br />

predictor for US (p�0.007), breast MRI (p�0.0001), CT (p�0.006) and<br />

bone scan use (p�0.002), but not mammography (p�0.90). There were<br />

varying correlations between Hispanics or Asians vs Whites for all imaging<br />

types. Stage, histology, U.S. region, and comorbidity index were most<br />

consistently predictive <strong>of</strong> imaging use <strong>of</strong> all types. When evaluating 6 mos<br />

prior to the 1st physician visit along with the POI, 96.4% <strong>of</strong> all patients had<br />

�1 mammogram, with 94.1% <strong>of</strong> Blacks having mammograms vs 96.5% <strong>of</strong><br />

Whites (OR 0.7, 95% CI 0.6-0.9; p�0.001). Conclusions: There is little<br />

racial difference in imaging performed during the POI for Medicare patients<br />

having breast cancer, and efforts to determine causes <strong>of</strong> survival disparities<br />

in such patients should be focused elsewhere. Whether these findings<br />

apply to the privately insured or the uninsured requires additional exploration.<br />

6063 General Poster Session (Board #4D), Mon, 1:15 PM-5:15 PM<br />

Rural versus urban differences among patients (pts) with hormone-receptor<br />

positive (HR�) breast cancer (BC) and a 21-gene assay recurrence score (RS).<br />

Presenting Author: Molly Andreason, University <strong>of</strong> Wisconsin, Madison, WI<br />

Background: Differences in BC outcomes have been noted between rural and urban<br />

women. The influence <strong>of</strong> tumor biology vs treatment factors as the driver <strong>of</strong> these<br />

differences remains unclear. The Oncotype Dx 21-gene assay RS predicts distant<br />

recurrence risk for HR�BC;aRS�18 indicates possible chemotherapy benefit. We<br />

assessed rural vs urban differences in RS and therapies received (endocrine therapy;<br />

chemotherapy for RS�18). Methods: Retrospective chart review was conducted on<br />

BC pts diagnosed 2005–2010 with a 21-gene assay RS. Stage, grade, medications<br />

and receptor status were abstracted; RS information was obtained from Genomic<br />

Health. Rural-Urban Commuting Area (RUCA) codes defined rural vs urban status.<br />

Comparisons between rural vs urban pts were made using two-sample tests,<br />

chi-square or Fisher’s exact test for discrete data such as RS (� 18 vs � 18), stage<br />

and ER status. T- or Wilcoxon rank sum test were used for continuous data (RS<br />

0-100 and age.) All tests were at a two-sided significance level <strong>of</strong> 0.05. Results: Of<br />

495pts with RS, 488 had RUCA codes (91% white, 61% postmenopausal). For<br />

rural vs urban pts, mean RS was 18 vs 19, p�0.15. The table shows univariate<br />

analysis for other predictors. For treatment, 321/354 (97%) rural vs 118/134<br />

(94%) urban pts started endocrine therapy (p�0.2). For RS�18, 17/55 (31%) rural<br />

vs 83/165 (50%) urban pts received chemotherapy (p�0.02, Chi-square test).<br />

Conclusions: Rural pts did not have significantly different RS compared with urban<br />

pts, but received significantly less chemotherapy for RS�18. This suggests that for<br />

HR� BC, rural vs urban discrepancies may be explained in part by treatment factors.<br />

We suggest prospective comparison or confirmation in another cohort.<br />

BC in rural vs urban women.<br />

Rural<br />

N�134 (%)<br />

Urban<br />

N�354 (%) p value<br />

RS #<br />

Age<br />

18 (8.8) 19 (9.0) 0.15*<br />

#<br />

BMI<br />

58 (10.3) 56 (10.1) 0.08*<br />

#<br />

30.0 (7.3) 28.8 (7.5) 0.12*<br />

Hormone use at diagnosis<br />

Yes 17 (13) 60 (17) 0.28 ^<br />

No 113 (84) 279 (79)<br />

Unknown 4 (3) 15 (4)<br />

Stage<br />

I 92 (69) 247 (70) 1 ^<br />

II 36 (27) 95 (27)<br />

Unknown 6 (5) 12 (4)<br />

Grade<br />

1 36 (27) 116 (33) 0.13 ^<br />

2 71 (53) 191 (54)<br />

3 24 (18) 41 (12)<br />

Unknown 3 (2) 6 (2)<br />

# Mean (SD); *t-test; ^ Chi-square test (excludes unknown).<br />

Health Services Research<br />

397s<br />

6062 General Poster Session (Board #4C), Mon, 1:15 PM-5:15 PM<br />

Patient-related predictors <strong>of</strong> poor-quality pain care in advanced lung<br />

cancer patients. Presenting Author: Inga Tolin Lennes, Massachusetts<br />

General Hospital Cancer Center, Boston, MA<br />

Background: Unrelieved pain remains a major problem for all patients,<br />

including those with cancer, despite national standards for pain management.<br />

Screening and addressing pain is an integral part <strong>of</strong> oncology visits<br />

and an ASCO QOPI indicator <strong>of</strong> quality oncology care. The goal <strong>of</strong> this study<br />

was to assess associations between patient-related factors, particularly<br />

patient distress, and meeting ASCO quality metrics for appropriate management<br />

<strong>of</strong> pain in patients with advanced lung cancers. Identification <strong>of</strong><br />

patient related factors could lead to targeted quality improvement efforts.<br />

Methods: From 8/07 to 9/10, we recruited consecutive new patients in a<br />

multidisciplinary thoracic oncology clinic to participate in a research<br />

database for which patients completed self-report instruments for distress,<br />

depression (PHQ-9) and anxiety (GAD-7), at their first oncology visit. We<br />

then performed a QOPI chart audit for patients with advanced lung cancer<br />

who received care at our institution. A composite measure <strong>of</strong> appropriate<br />

pain management was calculated. Components <strong>of</strong> the composite measure<br />

included 1) documentation <strong>of</strong> pain assessment, and 2) for patients with<br />

moderate-severe pain (�4 on 10 point scale) plan <strong>of</strong> care documentation.<br />

Results: 253 patients completed baseline assessments and had follow up.<br />

Pain was assessed in 252 (99%) patients. Over a third (88/253) <strong>of</strong> newly<br />

diagnosed advanced lung cancer patients had at least moderate pain on<br />

their first visit to the medical oncologist. Almost half <strong>of</strong> patients with<br />

moderate-severe pain were depressed (40/88). Of the patients with<br />

moderate-severe pain, 54/88 (63%) had a plan <strong>of</strong> care related to pain<br />

documented in the oncologist’s note. In total, 219 (87%) patients received<br />

appropriate pain assessment and care. In a multivariate model including<br />

age, sex, histology, ECOG PS, provider, depression, and anxiety, only<br />

depression independently predicted inadequate pain care. Depressed<br />

patients were 3 times more likely to receive poor quality pain care (OR<br />

2.75, 95% CI 1.04-7.25, p�0.04). Conclusions: At initial oncology visit,<br />

pain is present in over a third <strong>of</strong> patients with advanced lung cancer.<br />

Depression is highly co-morbid with pain and appears to be a risk factor for<br />

inadequate pain care.<br />

6064 General Poster Session (Board #4E), Mon, 1:15 PM-5:15 PM<br />

Impact <strong>of</strong> county-level surgical specialist density on breast (BrCa) and lung<br />

cancer (LuCa) mortality. Presenting Author: Andrei Karpov, British Columbia<br />

Cancer Agency, Vancouver, BC, Canada<br />

Background: Variations in distribution <strong>of</strong> the surgical workforce may result<br />

in differential access to cancer screening and treatment. Our aim was to<br />

explore the relationship between county-level surgical specialist density<br />

and BrCa and LuCa mortality. Methods: Using data from Area Resource File,<br />

US Census and National Cancer Institute, regression models that controlled<br />

for cancer incidence, county demographics and socioeconomic<br />

factors were constructed to examine the association among a) general<br />

surgeon (GS) and radiation oncologist (RO) density with BrCa mortality and<br />

b) thoracic surgeon (ThS) and RO density with LuCa mortality. Plastic (PS)<br />

and transplant surgeons (TrS) were used as surgical controls as they were<br />

not expected to correlate significantly with BrCa or LuCa mortality. Results:<br />

A total <strong>of</strong> 1,557 and 2,044 US counties were analyzed for BrCa and LuCa,<br />

respectively: mean incidences were 119 and 75 and death rates were 25<br />

and 59 per 100,000 people, respectively, for BrCa and LuCa. Mean<br />

specialist densities were 7.72 (GS), 0.80 (RO), and 0.97 (PS) [for BrCa<br />

counties] and 0.55 (ThS), 0.55 (RO), and 0.01 (TrS) [for LuCa counties]<br />

per 100,000. When compared to counties with no surgical specialist, those<br />

with at least one GS and RO for BrCa and at least one ThS and RO for LuCa<br />

were associated with decreased mortality (Table). Increasing the density <strong>of</strong><br />

GS and RO beyond 9 and 1 per 100,000 did not result in significant<br />

reductions in BrCa mortality. Likewise, increasing the density <strong>of</strong> ThS and<br />

RO to above 1 each per 100,000 failed to yield further improvements in<br />

LuCa mortality. Counties with more elderly residents also correlated with<br />

worse BrCa and LuCa outcomes. Conclusions: The presence <strong>of</strong> specific<br />

surgical specialists is associated with lower BrCa and LuCa mortality. There<br />

appears to be a threshold at which point further increase in their density do<br />

not contribute to continued improvements in outcomes. Distributing the<br />

surgical workforce across all counties will <strong>of</strong>fer population-based improvements<br />

in BrCa and LuCa mortality.<br />

BrCa LuCa<br />

Specialist Effect on mortality* p Effect on mortality* p<br />

GS -0.21 0.001 n/a n/a<br />

RO -0.10 0.003 -0.03 0.032<br />

ThS n/a n/a -0.04 0.009<br />

PS -0.04 0.29 n/a n/a<br />

TrS n/a n/a -0.01 0.84<br />

*Beta estimates<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


398s Health Services Research<br />

6065 General Poster Session (Board #4F), Mon, 1:15 PM-5:15 PM<br />

Prevalence and characteristics <strong>of</strong> patients with stage IV solid tumors who<br />

receive no anticancer therapy. Presenting Author: Alexander C. Small,<br />

Division <strong>of</strong> Hematology and Medical Oncology, The Tisch Cancer Institute,<br />

Mount Sinai School <strong>of</strong> Medicine, New York, NY<br />

Background: Clinicians caring for patients with cancer are well aware that a<br />

subset <strong>of</strong> patients who present with metastatic solid tumors never receive<br />

anticancer therapy for reasons including poor functional status, comorbidities,<br />

and patient preference. The prevalence and characteristics <strong>of</strong> this population<br />

have not previously been described. Methods: The National Cancer Database<br />

was queried for patients diagnosed with metastatic (stage IV) solid tumors<br />

including breast, cervix, colon, kidney, small-cell and non-small cell lung<br />

[NSCLC and SCLC], prostate, rectum and uterus. Patients who received<br />

neither radiation therapy nor systemic therapy were identified. Other factors<br />

such as age, race, income, insurance status, and diagnosis year were assessed.<br />

In an exploratory analysis, log-binomial regression was used to estimate<br />

prevalence ratios (PR) for the proportion <strong>of</strong> untreated stage IV to treated stage<br />

IV cancer cases according to these factors. Results: From 2000-2008,<br />

773,233 patients with stage IV cancer were identified <strong>of</strong> whom 159,284<br />

(21%) received no anticancer therapy (Table). Patients with NSCLC accounted<br />

for 55% <strong>of</strong> untreated patients. Across all cancer types, older age (PR range<br />

1.37-1.49, all p�0.001), black race (PR range 1.05-1.32, all p�0.001),<br />

lack <strong>of</strong> medical insurance (PR range 1.47-2.46, all p�0.001), and lower<br />

income (except uterus) (PR range 0.91-0.98 for every $10,000 income, all<br />

p�0.001) were associated with increased prevalence <strong>of</strong> not receiving treatment.<br />

Conclusions: Approximately 20% <strong>of</strong> patients who present with stage IV<br />

solid tumors never receive anticancer therapy. These findings have potential<br />

implications with regards to healthcare policy and access to care.<br />

Prevalence <strong>of</strong> patients with metastatic cancer who receive no anticancer<br />

therapy (2000-2008).<br />

Cancer Type<br />

Untreated<br />

stage IV<br />

Total<br />

stage IV % untreated<br />

Cancer-specific<br />

% <strong>of</strong> total<br />

untreated cases<br />

Kidney 12,079 47,417 25.5% 7.6%<br />

NSCLC 87,400 353,748 24.7% 54.9%<br />

Uterus 297 1,217 24.4% 0.2%<br />

SCLC 21,085 99,206 21.3% 13.2%<br />

Rectum 4,305 26,140 16.5% 2.7%<br />

Colon 18,816 119,748 15.7% 11.8%<br />

Cervix 1,407 9,535 14.8% 0.9%<br />

Breast 7,313 57,148 12.8% 4.6%<br />

Prostate 6,582 59,074 11.1% 4.1%<br />

Total 159,284 773,233 20.6% 100.0%<br />

6067 General Poster Session (Board #4H), Mon, 1:15 PM-5:15 PM<br />

The use <strong>of</strong> the word “cure” in oncology. Presenting Author: Kenneth David<br />

Miller, Sinai Hospital <strong>of</strong> Baltimore, Baltimore, MD<br />

Background: Use <strong>of</strong> the word “cure” in cancer care reflects a balance <strong>of</strong><br />

physician and patient optimism, realism, medico-legal concerns, and even<br />

superstition. The purpose <strong>of</strong> this study was to survey a group <strong>of</strong> clinicians<br />

regarding the frequency and determinants <strong>of</strong> using the word “cure” in their<br />

practice. Methods: In 2011, 180 oncology clinicians at the Dana-Farber<br />

Cancer Institute were invited to complete a survey regarding the word<br />

“cure” in cancer care. <strong>Part</strong>icipants completed a 19 question survey<br />

regarding how commonly their patients are cured, how <strong>of</strong>ten they use the<br />

word cure, in what circumstances they would tell a patient that they are<br />

cured, the timing <strong>of</strong> telling a patient that they are cured, and hesitancy in<br />

using the word cure. Three patient case scenarios were presented to elicit<br />

participants’ views regarding whether patients were cured and whether they<br />

need continued follow-up. Results: The 117 participants who provided<br />

answers to the cure questions (65% <strong>of</strong> the original 180 invitees), were<br />

evenly divided between males and females,73% were medical doctors, and<br />

56% had 10 or more years <strong>of</strong> experience since their training. Eighty-one<br />

percent <strong>of</strong> respondents were hesitant to tell a patient that they are cured<br />

and 63% would never tell a patient that they are cured. Only 7% feel that<br />

greater than 75% <strong>of</strong> their patients are, or will be, cured. This varied<br />

significantly by subspecialty (p�0.001). The participating clinicians reported<br />

that only 34% (sd: 30%) <strong>of</strong> patients ask if they are cured. In<br />

considering 20-year survivors <strong>of</strong> seminoma, large cell lymphoma, and<br />

estrogen positive breast cancer, 81%, 73%, and 47% <strong>of</strong> clinicians,<br />

respectively, believed that the patients were cured and 33%, 38%, and<br />

52% recommended annual oncology follow-up <strong>of</strong> the patients. Twentythree<br />

percent <strong>of</strong> clinicians believed that patients should never be discharged<br />

from the cancer center. Conclusions: Oncologists report that<br />

patients are hesitant to ask whether they are cured, and the clinicians are<br />

hesitant to tell, although this varied by cancer subspecialty. <strong>Annual</strong><br />

oncology follow-up was frequently endorsed, even after 20 years in<br />

remission.<br />

6066 General Poster Session (Board #4G), Mon, 1:15 PM-5:15 PM<br />

Utility <strong>of</strong> positron emission tomography (PET) scans on the management <strong>of</strong><br />

cancers <strong>of</strong> unknown primary. Presenting Author: Hao Chen, British Columbia<br />

Cancer Agency, Vancouver, BC, Canada<br />

Background: PET scans can be potentially useful in the diagnostic and<br />

staging workup <strong>of</strong> certain cancers. Although frequently ordered, its precise<br />

role in the investigation and management <strong>of</strong> cancers <strong>of</strong> unknown primary<br />

(CUP) remains poorly defined. Our main study aims were to 1) compare the<br />

utility <strong>of</strong> PET vs. CT scans in determining the primary site, lymph node<br />

status, and metastases for patients with CUP, and 2) describe the overall<br />

survival <strong>of</strong> patients for whom the primary site was determined by PET vs.<br />

those who were not. Methods: Patients diagnosed with CUP in British<br />

Columbia, Canada from 2006 to 2009, evaluated at 1 <strong>of</strong> 5 regional cancer<br />

centers in the province, and underwent a PET scan were reviewed. We<br />

measured concordance rates between PET and CT scans and constructed<br />

regression models to characterize the effect <strong>of</strong> PET scans on treatment and<br />

survival. Results: A total <strong>of</strong> 175 patients were included: median age was 60<br />

years and 76% were men. PET scans were most commonly performed for<br />

the following indications: locating primary (45%); staging (42%); and<br />

others (13%). Among those in whom CT did not detect the primary site,<br />

PET revealed the location <strong>of</strong> the primary in 9% <strong>of</strong> cases. CTs and PETs were<br />

concordant in demonstrating nodal status and metastases in 91% and 95%<br />

<strong>of</strong> patients, respectively. PET scans were able to show additional nodal<br />

involvement and uncover metastatic disease in 9% and 5%, respectively,<br />

when compared to CT scans. Identification <strong>of</strong> the site <strong>of</strong> primary cancer by<br />

PET did not substantially modify subsequent therapy (p�0.37) and it also<br />

failed to significantly improve the median overall survival (10.1 vs. 7.3<br />

months, p�0.57) when compared to those in whom the location <strong>of</strong> the<br />

primary was unconfirmed. Conclusions: In this retrospective cohort <strong>of</strong> CUP<br />

patients, CT and PET scans appear to provide a similar level <strong>of</strong> diagnostic<br />

and staging information. For a small proportion <strong>of</strong> CUP patients, PETs were<br />

superior in clarifying the primary site, nodal status, and metastases, but<br />

these did not alter therapy or increase overall survival. Based on these<br />

findings and considering the cost implications <strong>of</strong> intensive imaging studies,<br />

the diagnostic value <strong>of</strong> PET scans over CT scans appears limited to a small<br />

subset <strong>of</strong> patients with CUP.<br />

6068 General Poster Session (Board #5A), Mon, 1:15 PM-5:15 PM<br />

Sown the seeds: An analysis <strong>of</strong> published trials in clinical oncology.<br />

Presenting Author: Natalie M. Spradlin, Vanderbilt University Medical<br />

Center, Nashville, TN<br />

Background: Competition in the pharmaceutical marketplace has resulted<br />

in “seeding trials” which appear to serve little scientific purpose other than<br />

to gain a financial hold in the marketplace. Hallmarks <strong>of</strong> “seeding” clinical<br />

trials [Kessler, et al N Eng J Med 1994] include trial design that does not<br />

support stated research goals, industry sponsorship without novel findings<br />

or poor scientific rationale, and drugs introduced into an already crowded<br />

therapeutic class, i.e. “me too” drugs. We evaluated clinical trials published<br />

in high impact oncology journals to ascertain the prevalence <strong>of</strong> trials<br />

bearing the characteristics <strong>of</strong> a seeding trial. Methods: We conducted a<br />

PubMed search using “clinical trial”, “oncology”, published 2/2005 –<br />

2/2010, in Ann Oncol, J Clin Oncol, Lancet, Lancet Oncol, ortheN Engl J<br />

Med. All phase I/II, II and III studies were included. Phase I, hematology,<br />

radiation oncology, and pediatric studies were excluded. Data collected<br />

included disease site, phase, funding source, journal, and whether the trial<br />

fit criteria for “seeding” as previously defined. Results: 1781 articles have<br />

been evaluated to date. 528 met criteria for analysis. 130 were considered<br />

seed studies. Seed studies per journal were Ann Oncol 26 <strong>of</strong> 128, J Clin<br />

Oncol 88 <strong>of</strong> 340, Lancet 8<strong>of</strong>22,Lancet Oncol 5 <strong>of</strong> 16, and N Engl J Med 3<br />

<strong>of</strong> 22. Studies published per disease site and percent seed studies were<br />

melanoma 19 and 31.6%, breast 106 and 30.2%, GI 126 and 20.6%,<br />

lung 101 and 29.7%, GU 65 and 24.6%, gynecology 40 and 30%, head<br />

and neck 24 and 12.5%, CNS 24 and 12.5%, sarcoma 11 and 18.2%, and<br />

advanced cancer 12 with zero seeds. Overall phase totals were phase I/II<br />

16, phase II 272, and phase III 240. Seed studies per phase were phase II<br />

28.7%, phase III 21.7% and zero for phase I/II. 210 studies were solely<br />

industry sponsored, and <strong>of</strong> these, 37.1% were seed studies. Funding was<br />

not listed for 73 studies. Conclusions: The integrity <strong>of</strong> clinical trials is<br />

challenged in the race to claim major market share. Our analysis <strong>of</strong> clinical<br />

trials published in high impact oncology journals found seeding trials<br />

account for 24.6% <strong>of</strong> publications. 37.1% <strong>of</strong> industry sponsored trials are<br />

seed studies. Seed studies are more common among the major disease<br />

sites or sites for which targeted therapies exist.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


6069 General Poster Session (Board #5B), Mon, 1:15 PM-5:15 PM<br />

Effectiveness <strong>of</strong> a screening intervention to identify clinical-trial-eligible<br />

patients. Presenting Author: Leo Chen, University <strong>of</strong> British Columbia,<br />

Vancouver, BC, Canada<br />

Background: Advancements in cancer therapy require clinical trials, but<br />

only 3% <strong>of</strong> all cancer patients (CP) participate in trials causing many<br />

studies to be delayed or fail to complete. Literature indicates that accrual to<br />

clinical trials is primarily driven by MD related factors. The objective <strong>of</strong> this<br />

study was to evaluate whether the screening and identification <strong>of</strong> potentially<br />

eligible patients (PEP) for specific clinical trials would lead to an<br />

increased rate <strong>of</strong> accrual (RA). Methods: During a 4 month period, the<br />

charts <strong>of</strong> CP attending the outpatient clinics <strong>of</strong> 12 MDs were reviewed to<br />

determine eligibility for 21 phase II-IV trials for CP with BR, GI, GU, GY, or<br />

LG cancer. Trials were included if they had been open for � 4 months and<br />

would remain open � 8 months from the start <strong>of</strong> the intervention. A<br />

screening coordinator with minimal clinical background reviewed the<br />

electronic record <strong>of</strong> new and followup CP to determine eligibility according<br />

to protocol specified criteria. PEP were identified for medical oncologist by<br />

attaching notices to CP charts. <strong>Part</strong>icipating MDs were surveyed regarding<br />

the helpfulness and accuracy <strong>of</strong> the forms. A negative-binomial regression<br />

model was used to compare RA and find 95% CI for relative rates. Results:<br />

Between May 1 to August 31, 2011 a total <strong>of</strong> 2,098 charts were screened<br />

for eligibility for 21 trials, and 120 PEP were identified. Of these, 15 were<br />

randomized to the referred study, 4 to a different study, and 4 CP were<br />

<strong>of</strong>fered but declined the referred study. Four month RA for included trials<br />

were 61 before, 73 during and 51 after the intervention. Relative rates<br />

adjusted for MD bookings were 0.85 (95% CI: 0.67, 1.06, p � 0.15)<br />

before and 0.70 (95% CI: 0.54, 0.90, p � 0.005) after, relative to during<br />

the intervention. 33 completed questionnaires were received: 22 (67%)<br />

were helpful and 23 (70%) were accurate. Screening required a 1.0 Full<br />

Time Equivalent position during the period <strong>of</strong> the intervention. Conclusions:<br />

Manual screening <strong>of</strong> patient records to determine clinical trial eligibility is<br />

labor intensive and increases enrolment to specific clinical trials. Notifications<br />

were deemed mostly helpful and accurate by oncologists. Screening<br />

interventions should be considered to improve clinical trial accrual.<br />

6071 General Poster Session (Board #5D), Mon, 1:15 PM-5:15 PM<br />

Burnout prevalence in medical and radiation oncologists at British Columbia<br />

Cancer Agency (BCCA). Presenting Author: Catherine A. Fitzgerald, BC<br />

Cancer Agency, Vancouver Island Centre, Victoria, BC, Canada<br />

Background: Burnout, reported to affect 30-60% <strong>of</strong> oncology workers, is a<br />

syndrome <strong>of</strong> psychological distress typically manifesting in three dimensions:<br />

Emotional Exhaustion (EE), Depersonalization (DP) and Low Personal<br />

Accomplishment (PA). Causal factors include workload, dealing with<br />

terminally ill patients and difficulties maintaining a balance between<br />

pr<strong>of</strong>essional and personal life. As workload rises due to increased complexity<br />

<strong>of</strong> therapy and increasing prevalence <strong>of</strong> cancer patients, burnout may<br />

increase, especially in times <strong>of</strong> financial constraint. We sought to determine<br />

the prevalence <strong>of</strong> burnout in medical and radiation oncologists<br />

working at BCCA, which provides all radiation and the majority <strong>of</strong> medical<br />

oncology services to BC’s 4.5 million people. Methods: In March 2011,<br />

BCCA oncologists were invited to participate in a confidential online survey<br />

consisting <strong>of</strong> basic demographics and the 22 item MasLach Burnout<br />

Inventory (MBI) instrument, the latter a validated tool measuring distress in<br />

the three main dimensions <strong>of</strong> burnout. Normative data for physicians were<br />

used to interpret the results. Results: Response rate was 59%, female:male<br />

40:60% with similar response rates for medical and radiation oncology (60<br />

v 59%). Of the 73 who indicated their age range, 34 (47%) were between<br />

35 and 44 years old. Respondents indicated that they had considered<br />

reducing their Full Time Equivalent (FTE) (67%) or leaving BC (46%). In<br />

those with at least 2 scores at a severe level, these rates were 76% and<br />

71% respectively. Conclusions: Over 60% <strong>of</strong> responding BCCA oncologists<br />

report burnout in at least one domain <strong>of</strong> the MBI tool. Many have<br />

considered leaving the province or reducing their hours. These data are<br />

consistent with Grunfeld’s survey <strong>of</strong> Ontario oncologists (CMAJ 2000),<br />

although the rate <strong>of</strong> burnout is higher in this survey. Further research into<br />

ways to lessen burnout in oncology is urgently needed.<br />

Percentage <strong>of</strong> responding BCCA oncologists (n�78) with “severe” score on<br />

burnout (MBI) survey.<br />

Burnout factor<br />

Emotional<br />

exhaustion Depersonalization<br />

% scores at<br />

“severe”<br />

level<br />

Low personal<br />

achievement<br />

At least<br />

1 factor<br />

More<br />

than<br />

1 factor<br />

All 3<br />

factors<br />

51% 44% 28% 64% 44% 14%<br />

Health Services Research<br />

399s<br />

6070 General Poster Session (Board #5C), Mon, 1:15 PM-5:15 PM<br />

Disparities in the treatment and outcomes <strong>of</strong> lung cancer among HIVinfected<br />

people in Texas. Presenting Author: Gita Suneja, Department <strong>of</strong><br />

Radiation Oncology, University <strong>of</strong> Pennsylvania School <strong>of</strong> Medicine, Philadelphia,<br />

PA<br />

Background: HIV-infected (HIV�) people are at elevated risk for lung cancer<br />

and have higher mortality following lung cancer diagnosis than uninfected<br />

(HIV-) individuals. The disparity in survival is partly due to advanced stage<br />

at diagnosis, but it is unclear whether HIV� people with lung cancer are<br />

less likely to receive cancer treatment, which could worsen survival.<br />

Methods: We included adults � 18 years <strong>of</strong> age with lung cancer reported to<br />

the Texas cancer registry (N�156,930). HIV status was determined by<br />

linkage with the enhanced Texas HIV/AIDS Reporting System. We compared<br />

HIV� and HIV- lung cancer cases with respect to demographic and<br />

clinical characteristics. For non-small cell lung cancer (NSCLC) cases, we<br />

identified predictors <strong>of</strong> cancer treatment (surgery, radiation, and chemotherapy)<br />

using logistic regression. We used Cox regression to evaluate the<br />

effects <strong>of</strong> HIV and treatment on lung cancer-specific mortality. Results:<br />

Compared with HIV- lung cancer cases (N�156,593), HIV� lung cancer<br />

cases (N�337) were more likely to be young, non-Hispanic black, male,<br />

and to have distant stage disease (53.7% vs. 44.4%). HIV� cases were<br />

less likely to receive cancer treatment than HIV- cases (60.3% vs. 77.5%;<br />

odds ratio 0.39, 95%CI 0.30-0.52 after adjustment for diagnosis year,<br />

age, sex, race, stage, and histologic subtype). In Cox models adjusted for<br />

these variables, both HIV infection (hazard ratio [HR] 1.34, 95%CI<br />

1.15-1.56) and lack <strong>of</strong> cancer treatment (HR 1.69, 95%CI 1.66-1.72)<br />

were associated with higher lung cancer-specific mortality. After adjustment<br />

for cancer treatment, the association between HIV and lung cancer<br />

mortality was attenuated (HR 1.25, 95%CI 1.06-1.47). The association<br />

between HIV and lung cancer-specific mortality was stronger among<br />

untreated lung cancer cases (HR 1.32, 95%CI 1.01-1.72) than treated<br />

cases (adjusted HR 1.16, 95%CI 0.94-1.43; p-interaction�0.34).<br />

Conclusions: In this population-based study, HIV� people with NSCLC were<br />

less likely to be treated for lung cancer than their HIV- counterparts. This<br />

lack <strong>of</strong> treatment may be partly responsible for higher cancer-related<br />

mortality in HIV� cases. Further investigation is needed to understand<br />

disparities in cancer treatment for HIV� people.<br />

6072 General Poster Session (Board #5E), Mon, 1:15 PM-5:15 PM<br />

<strong>Clinical</strong> and cost impact <strong>of</strong> diagnostic slide review. Presenting Author:<br />

Justin Cuff, Aegis Review, San Francisco, CA<br />

Background: Diagnostic error is postulated to represent a significant source<br />

<strong>of</strong> preventable mortality, morbidity, and cost. A primary pathologic diagnosis<br />

<strong>of</strong> cancer has a relatively high error rate necessitating cost effective<br />

solutions to mitigate diagnostic related harm. Methods: A review <strong>of</strong> 773<br />

consecutive inbound transfers to Stanford, with review <strong>of</strong> prior pathology<br />

material, was performed to measure diagnostic discrepancy. Information<br />

was collected about the originating practice size and type, subspecialty<br />

area, additional testing, and a description <strong>of</strong> the disagreement. For a subset<br />

<strong>of</strong> cases, itemized payment data was collected to explore the financial<br />

impact <strong>of</strong> slide review. Results: We found that 9% <strong>of</strong> cases have diagnostic<br />

discrepancies when comparing the incoming working diagnosis with the<br />

result <strong>of</strong> the pathology slide review. Major diagnostic discrepancies, likely<br />

to alter treatment, were identified in 3% <strong>of</strong> cases. Discrepancy rates<br />

differed significantly between subspecialty areas (p�0.01) although few<br />

areas were spared major error. Disagreements about classification were<br />

common as were over/under calling malignancy, grading errors, and<br />

incorrect ordering or interpretation <strong>of</strong> ancillary testing. The subset <strong>of</strong> cases<br />

(n�107) with billing information demonstrated ~42,800 unique charge<br />

events associated with these episodes <strong>of</strong> care. The mean charge for patients<br />

was $292,813 (median $130,467). Typical <strong>of</strong> charge data the dollar<br />

amounts exhibited strong right-skew. Average reimbursement was $76,017<br />

(median $23,999) and the median charge:reimbursement ratio was 3.3.<br />

When analyzing cases by service line, only 1.4% <strong>of</strong> charges related to the<br />

diagnostic review <strong>of</strong> the prior pathology material, demonstrating a remarkably<br />

small impact on the overall cost. While additional testing was<br />

performed on 10% <strong>of</strong> cases, there was no evidence that cases with a<br />

discrepancy were associated with increased overall costs. Conclusions:<br />

Diagnostic slide review on prior material is relatively inexpensive in terms <strong>of</strong><br />

the cost <strong>of</strong> overall care and permits the detection <strong>of</strong> significant errors that<br />

may affect treatment planning. Pathology slide review represents a clinically<br />

impactful and low cost way to prevent errors in diagnosis from<br />

becoming errors in management.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


400s Health Services Research<br />

6073 General Poster Session (Board #5F), Mon, 1:15 PM-5:15 PM<br />

Colorectal cancer survivors’ needs and preferences for survivorship information.<br />

Presenting Author: Talya Salz, Memorial Sloan-Kettering Cancer<br />

Center, New York, NY<br />

Background: We assessed colorectal cancer (CRC) survivors’ needs and<br />

preferences for information to guide development <strong>of</strong> a survivorship care<br />

plan. Methods: We conducted a survey <strong>of</strong> survivors treated for stage I-III<br />

CRC at two hospitals in New York City. <strong>Part</strong>icipants completed treatment<br />

6-24 months before the interview and had not received a survivorship care<br />

plan. We evaluated whether survivors knew their basic treatment history,<br />

whether they understood ongoing risks, and their preferences for receiving<br />

survivorship information. Results: 175 CRC survivors completed the survey<br />

between June 2010 and November 2011. Survivors generally remembered<br />

basic information about their diagnosis and treatment: 88% accurately<br />

reported the location <strong>of</strong> their cancer; 95-100% accurately reported<br />

whether they had surgery, chemotherapy, or radiation; and 90-95%<br />

correctly reported the completion date (within 6 months) for each treatment.<br />

Survivors knew less about the risks <strong>of</strong> local and distant recurrences<br />

(69% and 77% correct, respectively) and <strong>of</strong> getting another CRC compared<br />

to unaffected individuals (40% correct). More than three quarters <strong>of</strong><br />

participants received information about their cancer, their treatment<br />

history, ongoing oncology visits, and testing to be done by the oncologist<br />

(77-86% across categories). Across these categories <strong>of</strong> information,<br />

93-99% <strong>of</strong> those who received information found it useful. Most survivors<br />

did not receive information about symptoms to report to their doctors,<br />

returning to work, financial issues, or legal issues (59-95% across<br />

categories); but those who received this information found it useful<br />

(67%-100% across categories). Conclusions: Even without receiving survivorship<br />

care plans, CRC survivors generally understood their cancer<br />

diagnosis and treatment. However, many lacked knowledge <strong>of</strong> ongoing<br />

risks, prevention, and nonmedical survivorship issues. Most survivors found<br />

the survivorship information they received useful. The greatest benefit <strong>of</strong><br />

survivorship care plans to survivors may not be summarizing past care as<br />

much as helping survivors understand their risks and plan for the future.<br />

6075 General Poster Session (Board #5H), Mon, 1:15 PM-5:15 PM<br />

Out-<strong>of</strong>-pocket (OOP) health care expenditure burden for Medicare beneficiaries<br />

with cancer. Presenting Author: Ilene H. Zuckerman, University <strong>of</strong><br />

Maryland School <strong>of</strong> Pharmacy, Baltimore, MD<br />

Background: Concern is increasing about the OOP burden faced by cancer<br />

patients (pts). Medicare beneficiaries have multiple comorbidities, have<br />

limited financial resources, and may face substantial cost sharing under<br />

traditional Medicare if they do not have generous supplemental coverage.<br />

We examined OOP spending and burden relative to income for Medicare<br />

beneficiaries with cancer, compared to a non-cancer comparison group.<br />

Methods: We used Medicare Current Beneficiary Survey data (1997-2007).<br />

Newly diagnosed cancer pts were selected using ICD-9CM codes on claims<br />

after a 12 mos washout period. OOP spending was assessed using self<br />

report for the index(diagnosis) and subsequent year. Pt characteristics were<br />

self reported. Generalized Linear Models estimated effects <strong>of</strong> pt characteristics<br />

on OOP spending; logistic regression identified pt characteristics<br />

associated with high burden, defined as OOP spending �20% <strong>of</strong> income.<br />

Results: The cohort included 1,869 beneficiaries with, and 10,057<br />

beneficiaries without cancer. Relative to the non-cancer cohort, cancer pts<br />

were older, had greater comorbidities, and were more likely to lack<br />

supplemental coverage (22 vs 16%) (all at p�0.01).OOP spending was<br />

$4,727 or 11.4% <strong>of</strong> total spending for cancer pts. The unadjusted<br />

difference between cancer and non-cancer pts in OOP spending was<br />

$1,518 (p�.001); with adjustment for patient characteristics, cancer<br />

patients faced an incremental $956 (p�.01) in OOP spending. Median-<br />

[mean] OOP/income was 10%[24%] for beneficiaries with, compared to<br />

6%[14%] without cancer (p�.001). Over ¼ (28%) <strong>of</strong> beneficiaries with<br />

cancer spent 20% <strong>of</strong> their income OOP, compared to 16% <strong>of</strong> beneficiaries<br />

without cancer (p�.001). Supplemental insurance and higher income were<br />

protective against high OOP burden, whereas assets, comorbidity, and<br />

receipt <strong>of</strong> cancer-directed radiation and antineoplastic therapy were<br />

associated with higher OOP burden. Conclusions: Medicare beneficiaries<br />

with cancer face higher OOP burden than their counterparts without<br />

cancer; some <strong>of</strong> the higher burden was explained by higher comorbidity<br />

burden and lack <strong>of</strong> supplemental insurance. Financial pressures may<br />

discourage some elderly patients from pursuing treatment.<br />

6074 General Poster Session (Board #5G), Mon, 1:15 PM-5:15 PM<br />

Speed <strong>of</strong> accrual into published phase III oncology trials: A comparison<br />

across geographic locations. Presenting Author: Nancy R. Ruther, Gundersen<br />

Lutheran Health System, La Crosse, WI<br />

Background: There is a perception that patient accrual may be slower in the<br />

US than it is in Europe (Wang-Gillam A et al, J Clin Oncol 2010;28:3803-<br />

3807). However, a systematic study has not been performed. We seek to<br />

determine the speed at which patients are accrued into published phase III<br />

oncology trials across geographic locations and to identify factors that may<br />

influence this process. Methods: We searched MEDLINE and identified all<br />

original phase III oncology therapeutic trials published in 2006-2010 (N �<br />

567). Trials with no reported activation and completion dates were<br />

excluded (n � 20). Accrual speed per trial was expressed as patients per<br />

month and was calculated by dividing the number <strong>of</strong> patients enrolled by<br />

number <strong>of</strong> months a trial was open. Results: The 547 trials included in our<br />

study enrolled 281,340 patients. Most trials were for adults only (95.6%),<br />

late stage cancers (77.5%), and involved multinational participation<br />

(44.1%). Funding sources were cooperative groups (45.5%), industry<br />

(33.8%) and academic centers (20.5%). The top 5 cancers studied were<br />

hematologic (19.2%), gastrointestinal (16.5%), lung (16.3%), breast<br />

(15.4%), and gynecologic (8.4%). Trial results were negative (57.4%),<br />

positive (38.2%), or equivalent (4.4%). The mean (�SD) accrual speeds<br />

varied according to trial location (multinational [25.0�25.4], US<br />

[13.3�16.5], other countries [11.4�15.7], Europe [8.7�11.8]; [P�<br />

.001]), funding source (industry [28.3�24.7], cooperative groups<br />

[13.3�19.0], academic [6.8�6.5]; [P� .001]), and cancer type (breast<br />

[24.0�29.4], gastrointestinal [20.2�24.2], lung [19.3�22.7], gynecologic<br />

[15.3�19.2], hematologic [11.2�10.7]; [P� .001]). After adjusting<br />

for funding source, accrual speeds were significantly different across trial<br />

locations only in 2 cancers: gastrointestinal (multinational [25.2�3.7], US<br />

[24.1�8.2], Europe [11.5�3.7], other [7.5�8.5]; P� .046) and gynecologic<br />

(multinational [28.9�5.4], other [10.5�7.8], US [6.6�5.3], Europe<br />

[4.2�5.9]; P� .004) studies. Conclusions: Among published phase III<br />

oncology trials, multinational studies accrued patients faster in gastrointestinal<br />

and gynecologic cancers and at a similar speed in other cancers.<br />

6076 General Poster Session (Board #6A), Mon, 1:15 PM-5:15 PM<br />

Racial disparities in depressive symptom prevalence and selective serotonin<br />

reuptake inhibitor (SSRI) utilization in cancer patients: An analysis<br />

from ECOG E2Z02: Symptom Outcomes and Practice Patterns (SOAPP).<br />

Presenting Author: Fengmin Zhao, Dana-Farber Cancer Institute, Boston,<br />

MA<br />

Background: Rates <strong>of</strong> diagnosis and treatment <strong>of</strong> depression in primary care<br />

vary widely by race. It is unclear whether such disparity exists in oncology.<br />

Methods: 3106 patients with cancer <strong>of</strong> the breast, prostate, colon/rectum,<br />

or lung were enrolled from multiple sites in a longitudinal study <strong>of</strong><br />

symptoms. Patient-reported depressed mood (PRD) on a 0-10 numerical<br />

rating scale and clinician-reported psychological distress (CRD) were used<br />

to identify depressive patients at baseline. Patients also rated depressed<br />

mood 4-5 weeks after baseline. A 2-point change was considered clinically<br />

significant for change in PRD. Logistic regression models were used to<br />

examine the effect <strong>of</strong> race on prevalence <strong>of</strong> depression and antidepressant<br />

use. Ordinal logistic regression models examined the effect <strong>of</strong> race on<br />

changes in depression. Results: Of the 3106 patients, 2648 were white,<br />

364 were black, and 94 were other race. At baseline, 20% had moderate/<br />

severe PRD; CRD was 28%. Black patients had higher rates <strong>of</strong> depression<br />

than whites, but the difference was not significant after adjusting for other<br />

covariates (PRD: 24% vs. 20%, adjusted OR�1.1, P�0.6; CRD: 30% vs.<br />

27%, adjusted OR�1.06, P�0.8). Improvement in depressed mood varied<br />

by baseline PRD score (57% with severe depression, 51% with moderate,<br />

and 14% with mild). In patients with moderate/severe depression at<br />

baseline, blacks were less likely to have depression improvement than<br />

whites (adjusted OR�0.41, P�0.008 for moderate depression, adjusted<br />

OR�0.35, P�0.01 for severe depression). Among patients with depression<br />

defined by CRD, 29% were taking an SSRI (blacks: 14%, whites: 31%),<br />

Blacks were less likely to take an SSRI than whites (adjusted OR�0.38,<br />

P�0.01). Conclusions: Less than one third <strong>of</strong> patients with depressive<br />

symptoms are taking SSRI antidepressants. Black race is not only associated<br />

with less improvement in depressive symptoms over time but also<br />

lower probability <strong>of</strong> SSRI utilization.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


6077 General Poster Session (Board #6B), Mon, 1:15 PM-5:15 PM<br />

Cost-effectiveness <strong>of</strong> biomarker-directed bevacizumab for first-line therapy<br />

<strong>of</strong> persons with metastatic colorectal cancer. Presenting Author: Kristin<br />

Berry, Fred Hutchinson Cancer Research Center, Seattle, WA<br />

Background: When added to first-line chemotherapy for metastatic colorectal<br />

cancer, bevacizumab provides a moderate survival increase, but is<br />

associated with significant adverse events and high cost. As only a minority<br />

<strong>of</strong> patients respond to antiangiogenic therapy, a diagnostic that identifies<br />

potential responders ex ante could potentially change the benefit to risk<br />

pr<strong>of</strong>ile <strong>of</strong> bevacizumab and likely improve cost-effectiveness. Since efforts<br />

to identify a predictive test have so far been unsuccessful, a novel strategy<br />

may be to measure tumor response after bevacizumab treatment initiation<br />

using an in vivo biomarker test to guide decisions on whether patients<br />

should continue treatment. The objective is to estimate the costeffectiveness<br />

<strong>of</strong> biomarker-directed use <strong>of</strong> bevacizumab in first-line treatment<br />

<strong>of</strong> metastatic colorectal cancer compared to standard care. Methods:<br />

The analysis takes the perspective <strong>of</strong> the US healthcare system. A decision<br />

model was built to estimate the incremental effect <strong>of</strong> adding bevacizumab<br />

to IFL, FOLFIRI, 5FU/LV and FOLOFOX/XELOX regimens. Survival data,<br />

estimated through a meta-analysis <strong>of</strong> four clinical trials, combined with<br />

utilities from the literature were used to calculate quality-adjusted life<br />

years (QALYs). Costs were based on Medicare reimbursement and expert<br />

opinion. Assumed performance characteristics <strong>of</strong> the biomarker technology<br />

(sensitivity, specificity – provided by our industry partner) were 75% and<br />

95% 10 days after commencement <strong>of</strong> bevacizumab. Results: The biomarkerdirected<br />

bevacizumab strategy dominated usual care, providing both an<br />

average per patient gain <strong>of</strong> 0.015 QALYs and a cost-savings <strong>of</strong> $21,038.<br />

The cost-effectiveness <strong>of</strong> the biomarker technology was most influenced by<br />

the cost <strong>of</strong> the test and colorectal cancer utilities. Variations in biomarker<br />

specificity and sensitivity changed the magnitude <strong>of</strong> cost-savings and<br />

benefit gain but did not change the overall result <strong>of</strong> dominance. Conclusions:<br />

Modeling predicts that a validated marker which could identify responders<br />

to bevacizumab will save money and improve quality-adjusted life years.<br />

The results support additional investment in identifying such a diagnostic.<br />

6079 General Poster Session (Board #6D), Mon, 1:15 PM-5:15 PM<br />

Surveillance after potentially curative breast cancer treatment: The impact<br />

<strong>of</strong> HMOs. Presenting Author: Robert J. Avino, Saint Louis University, St.<br />

Louis , MO<br />

Background: Over 230,000 new cases <strong>of</strong> breast cancer are diagnosed in the<br />

US annually. Most patients receive curative-intent treatment. Posttreatment<br />

surveillance is commonly done. We have previously documented<br />

dramatic variation in surveillance intensity among ASCO experts. The only<br />

surveillance modalities endorsed by ASCO for asymptomatic patients are<br />

<strong>of</strong>fice visit and mammogram. It is commonly believed that health maintenance<br />

organizations (HMOs) restrict test utilization. We sought to determine<br />

the effect <strong>of</strong> HMO penetration rate on the known variation in<br />

surveillance strategies. Methods: The 3245 ASCO members who indicated<br />

that breast cancer was a major clinical focus <strong>of</strong> their practice were surveyed<br />

regarding their surveillance practices. Members were asked to consider 4<br />

idealized clinical vignettes and indicate their surveillance plan for each. A<br />

menu <strong>of</strong> 12 testing modalities was <strong>of</strong>fered. Practice patterns were stratified<br />

by HMO penetration rate (0-14%, 14-22%, 22-33%, 33-61%) in each<br />

physician’s practice location. Repeated-measures ANOVA was employed<br />

for analysis. Results: Of the ASCO members surveyed, 1012 responded;<br />

915 were evaluable. The modalities most frequently recommended were<br />

<strong>of</strong>fice visit, CBC, liver function tests (LFTs), and diagnostic mammogram.<br />

There was significant variation (p�0.05) in the recommended frequency <strong>of</strong><br />

utilization <strong>of</strong> diagnostic mammogram, but in no other modalities. For<br />

example, in year 1, diagnostic mammogram was recommended 1.9 � 1.8<br />

(mean �SD) times for the 0-14% penetration rate cohort and 1.5 � 1.1<br />

times for the 14-22% cohort. Conclusions: We found little evidence that<br />

HMOs restrict test utilization. The HMO penetration rate in the clinician’s<br />

practice location cannot account for the overall variation we have documented<br />

previously.<br />

Health Services Research<br />

401s<br />

6078 General Poster Session (Board #6C), Mon, 1:15 PM-5:15 PM<br />

Deviations from guideline-based therapy for febrile neutropenia in cancer<br />

patients. Presenting Author: Jason D. Wright, Columbia University College<br />

<strong>of</strong> Physicians and Surgeons, New York, NY<br />

Background: Febrile neutropenia (FN) is a common cause <strong>of</strong> morbidity in<br />

cancer patients. We examined guideline and non-guideline based care for<br />

cancer patients hospitalized with FN and examined how treatment influenced<br />

outcomes. Methods: The Perspective database was used to examine<br />

the treatment <strong>of</strong> solid tumor patients with FN from 2000-2010. To capture<br />

initial decision-making, we examined treatment within 48 hrs <strong>of</strong> admission.<br />

Based on evidence-based guidelines we determined the appropriate<br />

use <strong>of</strong> guideline-based antibiotics as well as use <strong>of</strong> treatments not routinely<br />

recommended, vancomycin and granulocyte-colony stimulating factors<br />

(GCSF). Hierarchical mixed effects models were developed to examine the<br />

influence <strong>of</strong> patient, physician, and hospital characteristics on the quality<br />

<strong>of</strong> treatment. Patients were stratified into low and high-risk groups and the<br />

effect <strong>of</strong> initial treatment on outcome (nursing home discharge and death)<br />

examined. Results: Among 25,231 patients with FN, within 48 hours <strong>of</strong><br />

admission blood cultures were obtained in 90%, guideline-based antibiotic<br />

administered to 79%, vancomycin to 37%, and GCSF to 63%. Patients<br />

treated at high-volume hospitals, by high-volume physicians and patients<br />

managed by hospitalists were more likely to receive guideline-based<br />

antibiotics (p�0.05). Vancomycin use increased with time from 17% in<br />

2000 to 55% in 2010 while GCSF use only decreased from 73% to 55%.<br />

Patients treated by internists and hospitalists were more likely to receive<br />

vancomycin; high-volume physicians were less likely to treat with both<br />

vancomycin and GCSF (p�0.05). Among patients who received GCSF,<br />

15% received only one dose and 22% received 2 doses <strong>of</strong> filgrastim.<br />

Among low-risk patients prompt initiation <strong>of</strong> guideline-based antibiotics<br />

decreased the risk <strong>of</strong> discharge to a nursing facility (OR�0.78; 95% CI,<br />

0.66-0.91) and death (OR�0.65; 95% CI, 0.50-0.85). Conclusions: There<br />

is substantial variability in the initial treatment for FN in cancer patients.<br />

While use <strong>of</strong> appropriate empiric antibiotics is high, use <strong>of</strong> non guidelinebased<br />

treatments such as vancomycin and GCSF are common. Physician<br />

and hospital factors are the most important predictors <strong>of</strong> both guideline and<br />

non guideline-based treatment.<br />

6080 General Poster Session (Board #6E), Mon, 1:15 PM-5:15 PM<br />

The relationship between symptom burden and perceived comorbidity in<br />

outpatients with common solid tumors. Presenting Author: Christine<br />

Ritchie, Unviersity <strong>of</strong> California, San Francisco, San Francisco, CA<br />

Background: Cancer patients have high symptom burden, but it is unclear<br />

whether comorbid conditions are associated with patient-reported symptom<br />

burden (the number and severity <strong>of</strong> symptoms). Methods: The Eastern<br />

Cooperative Oncology Group (ECOG) enrolled 3106 patients with invasive<br />

cancer <strong>of</strong> the breast, colon/rectum, prostate and lung, regardless <strong>of</strong> phase<br />

<strong>of</strong> care or stage <strong>of</strong> disease. At baseline, patients completed the M.D.<br />

Anderson Symptom Inventory (MDASI) and were also asked how bothered<br />

they were by difficulties related to health problems other than cancer.<br />

Symptom burden (SB) was defined as the number <strong>of</strong> symptoms scored 7 or<br />

higher on the 13-item MDASI physical scale. Variance-weighted least<br />

squares regression was used to identify factors associated with increasing<br />

levels <strong>of</strong> being bothered by comorbid conditions. Results: About 65% <strong>of</strong><br />

patients were at least a little bothered by symptoms associated with<br />

comorbidities. There was a strong association between increased bother by<br />

comorbidities and increased SB (p�0.0001). Among those not bothered<br />

by difficulties related to comorbidities, �1 symptom was rated 7 or worse<br />

by 29% <strong>of</strong> patients, compared to 51% in patients bothered by difficulties<br />

related to comorbidities (Fisher’s exact p�0.0001). Except for hair loss,<br />

the severity <strong>of</strong> all symptoms was higher in those patients reporting bother<br />

due to other perceived comorbidities. Factors associated with increased<br />

bother include SB (p�0.0001), taking medications for diabetes<br />

(p�0.0001), impaired ECOG performance status (p�0.03), older age<br />

(p�0.004), and being perceived by the clinician as having higher degree <strong>of</strong><br />

difficulty for providing care (p�0.0001). Conclusions: Symptom burden<br />

varies significantly according to the degree to which cancer patients<br />

perceive bother that they attribute to comorbid health problems. Perception<br />

<strong>of</strong> comorbidity is worth exploring as a stratification factor in symptom<br />

research and as an indicator for complexity in patient care.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


402s Health Services Research<br />

6081 General Poster Session (Board #6F), Mon, 1:15 PM-5:15 PM<br />

Physician age and surveillance intensity following curative-intent treatment<br />

for breast cancer patients. Presenting Author: Steven F. Abboud,<br />

Saint Louis University , St. Louis, MO<br />

Background: Breast carcinoma is a large health care concern for patients,<br />

physicians, and society. 2.5 million women have been treated for breast<br />

cancer and are candidates for surveillance in the US. We have documented<br />

dramatic variation in post-treatment surveillance strategies utilized by<br />

ASCO experts caring for such patients. Since it is <strong>of</strong>ten asserted that<br />

younger physicians order more tests than older physicians, we sought to<br />

measure the effect <strong>of</strong> clinician age on post-treatment surveillance intensity<br />

for breast cancer patients by analyzing a recent survey <strong>of</strong> ASCO members.<br />

Methods: We surveyed the 3245 ASCO members who indicated that breast<br />

cancer treatment was a major focus <strong>of</strong> their practice. 4 succinct clinical<br />

vignettes describing generally healthy women with breast cancer <strong>of</strong> varying<br />

prognoses and a menu <strong>of</strong> 12 surveillance modalities were <strong>of</strong>fered. The<br />

menu was chosen after a literature search indicated that no other<br />

surveillance tests were commonly used. We analyzed data from one <strong>of</strong> the 4<br />

idealized vignettes only (the patient with TNM IIA carcinoma) and stratified<br />

responses by clinician age. Practice patterns were compared by years after<br />

completion <strong>of</strong> training (0-10, 11-20, 21-30, 30-40, �40 years), a<br />

surrogate measure <strong>of</strong> physician age. Statistical analysis employed ANOVA.<br />

Results: There were 1012 responses; 915 were evaluable. Statistically<br />

significant differences were observed across age strata for CBC, liver<br />

function tests (LFTs), and serum CEA level only. For example, ASCO<br />

clinicians in practice for 0-10 years after completion <strong>of</strong> training recommended<br />

CBCs 1.3 � 1.4 (mean � SD) times in year 1. Those � 40 years<br />

after completion <strong>of</strong> training recommended CBCs 2.4 � 1.3 times in year 1<br />

(p�0.001). Conclusions: Younger physicians recommend statistically significantly<br />

fewer CBCs, LFTs, and serum CEA levels during post-treatment<br />

surveillance than older physicians. However, the magnitude <strong>of</strong> the difference<br />

is clinically small for all 3 modalities and does not explain the known<br />

overall variation in surveillance practice among clinically active experts.<br />

6083 General Poster Session (Board #6H), Mon, 1:15 PM-5:15 PM<br />

Chemotherapy in the oldest old: The feasibility <strong>of</strong> cytotoxic therapy in the<br />

80� population. Presenting Author: Shelly Sud, Division <strong>of</strong> Medical<br />

Oncology, The Ottawa Hospital Cancer Centre, Department <strong>of</strong> Medicine,<br />

University <strong>of</strong> Ottawa, Ottawa, ON, Canada<br />

Background: The incidence <strong>of</strong> most common malignancies increases with<br />

age. As life expectancy improves globally, more elderly cancer patients will<br />

be candidates for systemic therapy. There is little data investigating<br />

chemotherapy (CT) in those over 80 – the “oldest old”. Our hypothesis is<br />

that CT in the 80� population may be associated with significant toxicities<br />

and is therefore not feasible for many patients. Methods: A retrospective<br />

chart review was undertaken to report outcomes <strong>of</strong> patients �80 years old<br />

who initiated CT for solid tumors at the Ottawa Hospital Cancer Center<br />

between Nov. 2005 and Jan. 2010. Baseline data on patient demographics,<br />

cancer type and CT were collected. Primary endpoints included: rates<br />

<strong>of</strong> CT dose reduction, omission, delay and discontinuation due to toxicity,<br />

hospitalization and blood transfusion rates. Results: CT was initiated on<br />

212 occasions (32% lung, 31% GU, 24% GI, 13% other cancer). Median<br />

age was 83 (range 80-92) and 60% <strong>of</strong> patients were male. Where data were<br />

available, 60% had a good performance score (ECOG 0-1) and 63% were<br />

current or ex-smokers. 82% had Charlson risk index scores <strong>of</strong> �5, 37% had<br />

�6 baseline medications, 18% lived alone independently. At baseline,<br />

11% were anemic, 12% had leukocytosis, and 45% had impaired renal<br />

function (eGFR�60). Most patients had stage 4 disease (76%), were<br />

treated with palliative intent (75%) and were receiving first line CT (77%).<br />

Initial dose was adjusted in 34% <strong>of</strong> cases. Therapy was discontinued due to<br />

toxicity in 30% <strong>of</strong> cases, and 53% <strong>of</strong> patients required dose reduction,<br />

omission or delay. In 38% <strong>of</strong> cases, patients were hospitalized during their<br />

course <strong>of</strong> therapy or within 30 days there<strong>of</strong>. Blood transfusions were<br />

required in 24%. Factors associated with risk <strong>of</strong> hospitalization included<br />

baseline number <strong>of</strong> medications �6 (OR 1.96, 95% CI 1.1-3.5) and<br />

baseline anemia (OR 2.55, 95% CI 1.07-6.05). Initial dose reduction at<br />

cycle 1 did not significantly affect rates <strong>of</strong> hospitalization, transfusion or CT<br />

discontinuation. Conclusions: CT in the 80� population is associated with a<br />

significant risk <strong>of</strong> hospitalization, transfusion and discontinuation due to<br />

toxicity, even when doses are adjusted from the outset. We plan to<br />

prospectively validate these findings.<br />

6082 General Poster Session (Board #6G), Mon, 1:15 PM-5:15 PM<br />

Comparison <strong>of</strong> drug approval between health Canada (HC) and the U.S.<br />

Food and Drug Administration (FDA). Presenting Author: Doreen Ezeife,<br />

University <strong>of</strong> Calgary, Calgary, AB, Canada<br />

Background: Differences in drug approval processes between countries can<br />

impact patient access to new therapies. In Canada, patients can freely<br />

access a new treatment after regulatory approval by Health Canada (HC)<br />

followed by funding approval from the provincial government. The aims <strong>of</strong><br />

this study were to delineate the Canadian drug approval timeline and to<br />

compare the time to drug approval between HC and the US FDA. Methods:<br />

Cancer drugs approved by the FDA from 1989 to 2011 were reviewed. For<br />

each drug, the following endpoints were determined: publication date <strong>of</strong><br />

phase I and pivotal phase III trial, date <strong>of</strong> FDA and HC approval, HC<br />

submission date, and funding approval in Alberta (AB). Time intervals<br />

between the aforementioned endpoints were calculated. Results: Of 55<br />

FDA-approved drugs, 51 drugs are approved by HC with 40 <strong>of</strong> these drugs<br />

funded in AB. HC approval occurs an average <strong>of</strong> 14.4 months post FDA<br />

approval (95% CI -36.9 to 66.1, sign rank test p�0.0001). However, there<br />

was no significant difference between the mean time from Phase I to FDA<br />

approval (48.5 months; 95% CI 21.2 to 75.8) and Phase I to HC approval<br />

(61.5 months; 95% CI 32.4 to 90.5). Most drugs (74%) were approved by<br />

the FDA prior to publication <strong>of</strong> the phase III trial. There was a trend towards<br />

faster drug approval from Phase III to FDA approval compared to HC<br />

(-14.97 versus 0.1 months, p � 0.05). HC submission occurs before FDA<br />

drug approval 77% <strong>of</strong> the time (mean 3.0 months prior; 95%CI: -59.1 to<br />

43.4, p � 0.0206). HC approval occurs on average 17 months post HC<br />

submission. AB funding approval occurs on average 22 months after HC<br />

approval. The time interval from Phase I to AB funding approval was<br />

significantly shorter for targeted compared to cytotoxic agents (mean time<br />

58 vs. 120 months; p � 0.039). Conclusions: HC drug approval lags behind<br />

FDA approval by about 14 months. Time from Phase III to drug approval<br />

tends to be shorter for the FDA compared to HC. This is the first<br />

documentation, to our knowledge, <strong>of</strong> the time required to bring a drug from<br />

phase I trial to provincial funding approval.<br />

6084 General Poster Session (Board #7A), Mon, 1:15 PM-5:15 PM<br />

Does drug cost drive drug adherence? The designer drug phenomenon.<br />

Presenting Author: Jalal Ebrahim, St. Michael’s Hospital, University <strong>of</strong><br />

Toronto, Toronto, ON, Canada<br />

Background: Cancer patients (pts) are on many medications, both for<br />

malignancy and supportive therapies. The cost <strong>of</strong> oral medications are<br />

funded by either the pt, public, or private mechanisms. Low adherence<br />

rates are observed in oral treatments, and non-adherence is the primary<br />

cause <strong>of</strong> treatment failures. To our knowledge, cost-related adherence to<br />

oral therapy in the context <strong>of</strong> malignancy has not been studied extensively<br />

in the existing literature. We assessed the relationships between oral<br />

medication costs and adherence rates. Methods: This cohort study enrolled<br />

453 pts at 3 outpatient heme/onc clinics in the Greater Toronto Area. A<br />

7-item survey was designed to assess pt demographics, self-reported<br />

adherence to oral medication, type <strong>of</strong> drug coverage (private payer, public<br />

payer, self payer) and patients’ perceived cost <strong>of</strong> oral drugs. Oral medications<br />

were recorded and actual monthly costs were calculated. Descriptive<br />

statistics were used to describe frequencies. Spearman Rank Order<br />

Correlations and Chi-Square Analyses were used to examine relationships<br />

between variables. Results: Of 453 pts, 50% had a private drug plan, 24%<br />

paid out <strong>of</strong> pocket, 44% had government funding and 4% reported their<br />

physician had arranged funding. 51% <strong>of</strong> pts had oral drug costs <strong>of</strong> �<br />

$100/month. Self reported adherence to prescribed oral medications was<br />

80%. As the cost <strong>of</strong> prescribed medications increased, so did self reported<br />

adherence (r�0.144, p�0.002). There was also a significant relationship<br />

between drug coverage and oral drug costs (c²�23.78, df�12, p�0.02).<br />

Pts paying out <strong>of</strong> pocket were significantly less likely than all other pts to<br />

have oral drug costs <strong>of</strong> �$500/month (11% vs 19%) Conclusions: As oral<br />

drug costs increase, so does likelihood <strong>of</strong> adhering to prescribed regimen.<br />

This implies that further pt education about the efficacy and importance <strong>of</strong><br />

medications may help with adherence, regardless <strong>of</strong> cost. It is possible that<br />

pts are provided with more education regarding newer and more expensive<br />

agents than they are regarding older and cheaper agents, regardless <strong>of</strong><br />

efficacy. Disparities observed in medication costs between those with<br />

private drug plans vs. those without suggests financial restrictions may<br />

affect prescription patterns in this group.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


6085 General Poster Session (Board #7B), Mon, 1:15 PM-5:15 PM<br />

Physical activity (PA) and physical function (PF) in adult cancer survivors<br />

(CS). Presenting Author: Peter Crawford Birks, British Columbia Cancer<br />

Agency, Vancouver, BC, Canada<br />

Background: The longevity <strong>of</strong> CS is increasing, underscoring the importance<br />

<strong>of</strong> lifestyle modifications and preventive care, consisting <strong>of</strong> regular PA and<br />

maintenance <strong>of</strong> PF. Our aims were to 1) compare PA and PF among<br />

short-term CS, long-term CS and non-cancer controls (NCC) and 2) identify<br />

other clinical factors associated with decreased PA and PF. Methods: We<br />

identified CS and NCC from the US National Health and Nutrition<br />

Examination Survey (NHANES). CS were categorized into �/�5 (shortterm)<br />

and �5 years (long-term) based on date <strong>of</strong> diagnosis. Multivariate<br />

weighted regression models were constructed to examine for differences in<br />

28 and 26 measures <strong>of</strong> PA and PF, respectively, while controlling for age,<br />

gender, ethnicity, education, income, and other confounders. We explored<br />

specific and general PA domains such as aerobic activity and decline from<br />

past activity level as well as PF areas including ADLs, IADLs and mobility.<br />

Results: In total, 26,185 subjects were identified: 968 short-term CS,<br />

1,367 long-term CS, and 23,832 NCC. For the entire cohort, the mean age<br />

was 50 years, 52% were men, and 50% were white. Comparing across the<br />

3 patient groups, CS were generally older than NCC (60 vs 45 years,<br />

p�0.01). In terms <strong>of</strong> PA, short-term CS reported less general activity<br />

compared to 1 year ago (OR 0.58, 95%, CI 0.46-0.73, p�0.01) than<br />

long-term CS and NCC. In comparison to NCC, both groups <strong>of</strong> CS also<br />

described less current activity than their baseline from 10 years ago (OR<br />

0.63, 95% CI 0.48-0.82, P�0.001 and OR 0.67, 95% CI 0.55-0.83,<br />

P��0.01). There were significant differences among patient groups in<br />

several areas <strong>of</strong> PF (Table). Other factors that were associated with lower PA<br />

and PF included advanced age, low education, and low income. Conclusions:<br />

PA between CS and NCC were similar, although current activity as<br />

compared to that <strong>of</strong> the preceding year or past decade appears lower in CS,<br />

which may reflect their older age. Impairments in several measures <strong>of</strong> PF<br />

were observed among CS, which could impact their quality and quantity <strong>of</strong><br />

life.<br />

Limited work<br />

OR (95%CI) p<br />

Limited<br />

mobility OR<br />

(95%CI) p<br />

Limited<br />

leisure<br />

activities OR<br />

(95%CI) p<br />

NCC 1.00 0.01 1.00 0.02 1.00 0.05<br />

CS 5y 1.61 (1.3-1.9) 1.44 (1.1-1.9) 1.02 (0.6-1.7)<br />

6087 General Poster Session (Board #7D), Mon, 1:15 PM-5:15 PM<br />

Involvement <strong>of</strong> low- and middle-income countries (LMCs) in oncology<br />

randomized controlled trials (RCTs). Presenting Author: Janice C. Wong,<br />

Princess Margaret Hospital, Toronto, ON, Canada<br />

Background: Lower costs and fewer regulatory barriers make clinical trials in<br />

LMCs attractive, but little is known about characteristics <strong>of</strong> such trials and<br />

roles <strong>of</strong> investigators from LMCs. We describe trends in participation <strong>of</strong><br />

investigators from LMCs in oncology RCTs over a decade. Methods: We used<br />

Medline to identify all RCTs published in English between January 1 1998<br />

and December 31 2008 evaluating treatment in five solid tumours (lung,<br />

breast, colorectal, stomach, liver) with high mortality in LMCs as per<br />

GLOBOCAN 2002. Data on author affiliations and roles (first, middle or<br />

corresponding author), trial characteristics, funding and study interventions<br />

were abstracted using an electronic form. Countries were stratified<br />

into low-, middle- and high-income using World Bank data. Interventions<br />

were categorized as requiring basic, limited, enhanced or maximal resources<br />

as per the Breast Health Global Initiative classification. Logistic<br />

regression identified factors associated with investigator participation from<br />

LMCs. Results: 454 trials were identified: lung (n�177), breast (n�165),<br />

colorectal (n�82), stomach (n�29), liver (n�7). The annual number <strong>of</strong><br />

trials published increased from 10 trials in 1998 to 86 in 2008. The<br />

proportion <strong>of</strong> trials with at least one LMC author also increased over time<br />

from 20% in 1998 to 29% in 2008 (p�0.01), but almost all LMC authors<br />

were from middle-income countries. In 17% <strong>of</strong> trials involving LMC<br />

authors, they were first or corresponding authors. 67% <strong>of</strong> trials involving<br />

LMC investigators evaluated interventions requiring enhanced or maximal<br />

resources for implementation. Factors associated with LMC participation<br />

included industry funding (OR�3.57, p�0.0001), use <strong>of</strong> placebo arm<br />

(OR�2.59, p�0.02) and metastatic setting (OR�3.87, p�0.0003).<br />

Conclusions: An increasing number <strong>of</strong> oncology RCTs involve LMC investigators<br />

primarily in non-leadership roles. These trials are commonly industryfunded<br />

and <strong>of</strong>ten test interventions that require at least enhanced resources<br />

for implementation. To minimize concerns <strong>of</strong> exploitation and facilitate<br />

future collaboration it is crucial that interventions are locally feasible and<br />

investigators receive appropriate authorship.<br />

Health Services Research<br />

403s<br />

6086 General Poster Session (Board #7C), Mon, 1:15 PM-5:15 PM<br />

The NCI Community Cancer Centers Program (NCCCP): A model for<br />

reducing cancer health care disparities. Presenting Author: Donna M.<br />

O’Brien, NCI NCCCP Program, Bethesda, MD<br />

Background: In 2007, NCI launched the NCCCP, a public-private partnership<br />

with 16 community hospital cancer centers in 14 states, to explore<br />

methods to improve patient access to advanced cancer care in the<br />

community. With 40% <strong>of</strong> its NCI funding directed to reduce disparities<br />

across the cancer continuum, the NCCCP aims to: 1) Enhance access to<br />

care; 2) Improve quality <strong>of</strong> care; and 3) Increase clinical trials accrual. This<br />

approach supports priorities in the 2009 ASCO Policy Statement: Disparities<br />

in Cancer Care. Methods: A disparities workplan was developed to<br />

support the three aims. NCI and the sites worked as a learning collaborative<br />

to develop strategies and metrics for: race and ethnicity data tracking; near<br />

real-time reporting <strong>of</strong> adherence to Commission on Cancer (CoC) treatment<br />

quality measures; community outreach and patient navigation to increase<br />

cancer screening; and improved clinical trial underserved accrual. The<br />

tools and resources supporting these efforts will be discussed.<br />

(http://ncccp.cancer.gov/About/Progress.htm). Results: Evaluation <strong>of</strong> the 3<br />

year pilot shows improvement for underserved populations: Concordance<br />

with CoC treatment quality measures for radiation therapy for breast<br />

conserving surgery among Medicaid patients improved from 59.5 percent<br />

to 84.8 percent (p�.05). Increased community screening events (from<br />

992 to 1,585) and community partnerships focused on underserved<br />

populations (from 78 to 195). Increased accrual to NCI trials (minority<br />

accrual from 82 to 151 and elderly from 200 to 641). Conclusions: To be<br />

effective in reducing healthcare disparities, a multi-level approach is<br />

needed. This includes having: organizations which demonstrate a strong<br />

community-oriented mission; commitment by hospital management; engagement<br />

<strong>of</strong> private practice physicians; targeted training <strong>of</strong> staff; use <strong>of</strong><br />

standardized data collection and metrics; involvement <strong>of</strong> strategic partners<br />

with aligned goals at the national and local level; support by relevant NCI<br />

experts; and sharing best practices across a learning collaborative. The<br />

NCCCP disparities model was used in a variety <strong>of</strong> community settings<br />

targeting different underserved populations and has demonstrated effect in<br />

care in the respective communities.<br />

6088 General Poster Session (Board #7E), Mon, 1:15 PM-5:15 PM<br />

Beyond the performance rate: Understanding differences in treatment<br />

status using the Rapid Quality Reporting System (RQRS). Presenting<br />

Author: Erica J. McNamara, Commission on Cancer, <strong>American</strong> College <strong>of</strong><br />

Surgeons, Chicago, IL<br />

Background: RQRS is a cancer registry based tool for rapid case ascertainment<br />

and real-time care tracking using 5 National Quality Forum approved<br />

quality measures for breast (BC) and colon cancer (CC) and 1 <strong>American</strong><br />

<strong>Society</strong> <strong>of</strong> <strong>Clinical</strong> Oncology, National Comprehensive Cancer Network and<br />

Commission on Cancer (CoC) endorsed rectal cancer (RC) measure. RQRS<br />

was introduced in 2009 for testing at selected CoC accredited programs.<br />

The purpose <strong>of</strong> this study was to evaluate the proportion <strong>of</strong> cases with<br />

treatment administered for 4 measures where care is concordant with<br />

treatment either “administered” or “considered but not administered”<br />

(CNA). Methods: Case information on 19,631 breast, colon and rectal<br />

cancer patients diagnosed from 2009 – 2010 submitted by 64 RQRS<br />

participating sites were reviewed for 4 measures: (1) hormone therapy<br />

within 1 year <strong>of</strong> diagnosis (dx) for AJCC T1c, N0M0, or stage II or II;<br />

hormone receptor positive BC (HT), (2) multi-agent chemotherapy within 4<br />

months <strong>of</strong> dx; age � 70; hormone receptor negative BC (MAC), (3) adjuvant<br />

chemotherapy within 4 months <strong>of</strong> dx; age �80; AJCC Stage III CC (ACT),<br />

and (4) radiation therapy within 6 months <strong>of</strong> dx; age �80; AJCC T4N0M0<br />

or stage III patients receiving surgical resection for RC (AdjRT). Treatment<br />

status was defined as concordant (administered or CNA) or non-concordant<br />

(NC – not part <strong>of</strong> planned first course therapy or received after required time<br />

frame). Patient demographics and Charlson-Deyo (CD) co-morbidity scores<br />

were compared by concordance status using Chi-square tests. Results: The<br />

aggregate concordance rate was 83.3%. Among these 889 (5.4%) were<br />

CNA. Co-morbidity score was � 0 for 15% <strong>of</strong> cases. Patients age � 70, with<br />

Medicare insurance, CD score � 0 were significantly more likely to be CNA<br />

compared to completed or NC. Patient choice was the most common reason<br />

for CNA (64%). Conclusions: Among patients treated at RQRS test sites for<br />

which RQRS measures applied and care was concordant, only 5% were<br />

CNA. Patients over age 70 or with CD � 0 were more likely to have<br />

treatment CNA. Inclusion <strong>of</strong> CNA as concordant care has a minor impact on<br />

concordance rates and provides cancer programs with important information<br />

for targeting care review and quality improvement programs.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


404s Health Services Research<br />

6089 General Poster Session (Board #7F), Mon, 1:15 PM-5:15 PM<br />

Use and duration <strong>of</strong> chemotherapy (CT) in patients (pts) with metastatic breast<br />

cancer (MBC) according to tumor subtype (TS) and line <strong>of</strong> therapy (tx).<br />

Presenting Author: Davinia Shien Seah, Dana-Farber Cancer Institute, Boston,<br />

MA<br />

Background: Benefits <strong>of</strong> CT vary widely in MBC pts. We aimed to describe the impact<br />

<strong>of</strong> breast cancer TS and line <strong>of</strong> tx on the duration <strong>of</strong> CT. Methods: In a retrospective<br />

analysis, we identified 205 pts treated with CT for MBC at the Dana-Farber Cancer<br />

Institute between 2005-8. TS were classified as hormone receptor (HR)� (ER�/PR�/<br />

HER2-), triple negative (TN) (ER-/PR-/HER2-) or HER2� (HER2�, any HR). CT<br />

duration was defined as �time from start <strong>of</strong> one CT line to the start <strong>of</strong> the next CT.�<br />

Chi-square, Kruskal-Wallis, and Kaplan-Meier methods were used. Results: Median<br />

follow-up was 54 months (m). Most pts had received adjuvant tx, though 23% had<br />

MBC at diagnosis. Almost all HER2� pts had concurrent anti-HER2 tx. CT duration<br />

was longest in 1st line with consecutive stepwise decrease. The proportion <strong>of</strong> pts<br />

who received CT beyond the 3rd line varied by TS: 52% <strong>of</strong> HER2� pts received � 4<br />

CT lines, compared with 37% <strong>of</strong> HR� and 29% <strong>of</strong> TN pts. Duration in later CT lines<br />

also differed by TS. Median CT duration in HER2� pts was � 4 m in all 6 lines, but<br />

HR� and TN pts had a median CT duration <strong>of</strong> �4m from the 4th line onwards.<br />

Quartile estimates suggested a substantial fraction <strong>of</strong> HER2� and HR� pts<br />

receiving �6m <strong>of</strong> CT in 5th and 6th line; by contrast, CT duration for TN pts were<br />

consistently brief in later CT lines. Conclusions: TS determines the likely duration<br />

and number <strong>of</strong> lines <strong>of</strong> CT for MBC. Among HR� and HER2� pts who receive CT<br />

beyond 3rd line, there are substantial rates <strong>of</strong> prolonged tx suggesting clinical<br />

benefit. For TN pts, the opportunity for additional lines and the CT duration declined<br />

more dramatically. The true role <strong>of</strong> advanced (� 3rd) CT lines for all MBC in<br />

improving quality <strong>of</strong> life, survival, and the predictors <strong>of</strong> such benefit, warrant further<br />

study.<br />

CT characteristics.<br />

HR� TN HER2�<br />

No. <strong>of</strong> pts, N (%) 102 49 45 22 58 28<br />

No. <strong>of</strong> CT lines<br />

median, min, xax<br />

Pts receiving nth CT line<br />

N, %<br />

3 1 9 3 1 8 4 1 9<br />

1 102 100 45 100 58 100<br />

2 79 77 36 80 44 76<br />

3 56 55 26 58 40 69<br />

4 38 37 13 29 30 52<br />

5 24 24 8 18 24 40<br />

6<br />

CT duration<br />

Median, 1st, 3rd quartile<br />

9 9 6 13 19 33<br />

1 6.5 2.9 12.0 5.8 2.8 10.7 9.0 4.3 13.1<br />

2 4.4 2.6 7.9 3.3 2.0 6.3 5.1 2.8 11.3<br />

3 3.6 1.9 6.0 2.8 2.1 6.8 6.3 2.9 8.3<br />

4 4.0 2.1 7.3 3.4 1.8 8.1 4.7 2.7 8.9<br />

5 3.5 1.9 6.5 2.6 1.9 3.2 4.0 2.3 7.0<br />

6 3.3 2.1 7.4 2.0 1.8 2.3 4.2 1.9 6.1<br />

6091 General Poster Session (Board #7H), Mon, 1:15 PM-5:15 PM<br />

Recruitment <strong>of</strong> cancer patients to National Institute for Health research<br />

cancer research network portfolio studies between 2001 and 2011.<br />

Presenting Author: Matthew Cooper, National Institute for Health Research<br />

Cancer Research Network, Leeds, United Kingdom<br />

Background: The National Institute for Health Research Cancer Research<br />

Network (NCRN) was established in 2001 in England, United Kingdom to<br />

improve cancer patient outcomes by improving the coordination, integration<br />

and speed <strong>of</strong> cancer research. Methods: Baseline recruitment <strong>of</strong> cancer<br />

patients in England to clinical studies was around 4% <strong>of</strong> incident<br />

population. Research Networks were established initially in England<br />

(NCRN) and then across the UK, co-terminus with clinical cancer service<br />

networks, and a per capita based funding model used to provide a research<br />

infrastructure to support recruitment to a nationally defined research<br />

portfolio. Results: Within 3 years, as the networks were established,<br />

recruitment <strong>of</strong> patients to studies doubled from 10,000 to 20,000 cancer<br />

patients per year. Recruitment has continued to increase year on year,<br />

supported initially by underspend that had accrued from earlier years in the<br />

life <strong>of</strong> NCRN, and more recently from additional resources invested via the<br />

NIHR comprehensive networks. Data for 2010/11 show that over 45,000<br />

cancer patients are now recruited into portfolio studies in England each<br />

year, with over 50,000 across the whole <strong>of</strong> the UK. Conclusions: Dedicated,<br />

targeted, clinician-led National Health Service investment into supporting<br />

national portfolio studies, has delivered an unprecedented five-fold increase<br />

in recruitment <strong>of</strong> cancer patients into clinical trials across the<br />

United Kingdom. This required coordinated research infrastructure, close<br />

cooperation with research funders, particularly Cancer Research UK and<br />

the National Cancer Research Institute, and the enthusiasm and hard work<br />

<strong>of</strong> many clinicians, patients and others working to deliver clinical cancer<br />

care in the National Health Service in the United Kingdom.<br />

6090 General Poster Session (Board #7G), Mon, 1:15 PM-5:15 PM<br />

Health-related quality <strong>of</strong> life assessment in EORTC cancer clinical trials.<br />

Presenting Author: Efstathios Zikos, European Organisation for Research<br />

and Treatment <strong>of</strong> Cancer Headquarters, Brussels, Belgium<br />

Background: Over the last three decades health-related quality <strong>of</strong> life<br />

(HRQOL) has become an important part <strong>of</strong> the randomised controlled trials<br />

(RCTs) conducted by the European Organisation for Research and Treatment<br />

<strong>of</strong> Cancer (EORTC). This review aims to undertake a descriptive<br />

database evaluation <strong>of</strong> all the HRQOL studies conducted in EORTC since<br />

1980. Methods: The EORTC protocol database (n�785) was reviewed,<br />

restricting the search to between 1980 and 2011 (n�735). We investigated<br />

the number <strong>of</strong> HRQOL studies conducted in EORTC trials, the RCTs’<br />

status and the use <strong>of</strong> HRQOL tools since 1980. Results: 157 protocols with<br />

HRQOL assessment were identified involving 70,903 patients. 73 studies<br />

ended as defined in the protocol; 27 studies closed early due to poor<br />

accrual; 17 are at the final analysis <strong>of</strong> the primary end point stage; 14<br />

studies are still open to recruitment; 11 are closed to patient entry; and 15<br />

new RCTs are pending activation with HRQOL. The majority <strong>of</strong> phase III<br />

(n�135) and phase II/III (n�9) RCTs have HRQOL as secondary endpoint.<br />

EORTC also conducted a number <strong>of</strong> large scale field studies (n�11), where<br />

HRQOL was the primary endpoint. During the early period <strong>of</strong> 1980 to 1989<br />

HRQOL was assessed in 12 EORTC RCTs by using a small number <strong>of</strong><br />

HRQOL items, but from 1990 to 2000, HRQOL was assessed in 97 RCTs<br />

using more comprehensive HRQOL tools. Between 2001 and 2011 the<br />

number <strong>of</strong> RCTs with HRQOL was 48. The EORTC clinical groups with the<br />

most RCTs containing HRQOL were Radiation Oncology (n�22), Genito-<br />

Urinary (n�20), Gynaecological (n�16), Breast Cancer (n�16), Lung<br />

(n�13), Gastrointestinal, (n�13) and Brain (n�10). The EORTC HRQOL<br />

tools were used in 90% <strong>of</strong> the trials, with other validated tools being used<br />

when required. Conclusions: Our review <strong>of</strong> EORTC RCTs has shown how<br />

patient perspective has been constantly considered <strong>of</strong> major importance in<br />

oncology during the last three decades. The inclusion <strong>of</strong> patient perspective<br />

in drug development shows that a more comprehensive HRQOL assessment<br />

has taken place over time as better instruments have become available. As<br />

the positive value <strong>of</strong> patient perspective grows to clinicians, regulatory<br />

bodies and industry, we expect that EORTC will continue its support by<br />

including HRQOL endpoints where appropriate.<br />

6092 General Poster Session (Board #8A), Mon, 1:15 PM-5:15 PM<br />

Characterizing fatigue associated with sunitinib (SU) in patients (pts) with<br />

metastatic renal cell carcinoma (mRCC). Presenting Author: David Cella,<br />

Department <strong>of</strong> Medical Social Sciences, Northwestern University Feinberg<br />

School <strong>of</strong> Medicine, Chicago, IL<br />

Background: Fatigue is common in cancer pts and associated with use <strong>of</strong><br />

tyrosine kinase inhibitors (TKIs) such as SU. Limited data exist on the time<br />

pattern <strong>of</strong> fatigue with TKI therapy. Methods: Data from treatment-naïve<br />

mRCC pts in SU arms <strong>of</strong> two clinical trials were analyzed retrospectively.<br />

Study 1; 375 pts were randomized to SU 50 mg/d on a 4 weeks-on-2-weeks<strong>of</strong>f<br />

schedule (Schedule 4/2), for up to 30 cycles. Study 2; pts were<br />

randomized to SU 50 mg/d Schedule 4/2 (Group 1; n�146) or 37.5 mg/d<br />

continuous daily dosing (CDD; Group 2; n�146). In both trials, fatigue was<br />

measured with the question to pts: “I feel fatigued” over the past week<br />

(5-point rating scale, not at all-very much), and with the provider-rated<br />

Common Terminology Criteria for Adverse Events (CTCAE). In addition to<br />

descriptive pr<strong>of</strong>iles, Study 1 used two modeling approaches; repeated<br />

measures model (M1), with time as a categorical predictor; and random<br />

intercept-slope model (M2), with time as a continuous predictor. Study 2<br />

calculated mean absolute values <strong>of</strong> within-cycle rate <strong>of</strong> change (from one<br />

assessment to the next) through the first 6 treatment cycles. Results: In<br />

Study 1, representing fatigue across cycles, M1 showed that the initial<br />

increase in patient-reported fatigue was worst during Cycle 1; mean values<br />

at all subsequent cycles were numerically better. For CTCAE fatigue, M1<br />

showed that all but one <strong>of</strong> the pair-wise comparisons <strong>of</strong> the cycle means<br />

were not significantly different. M2 showed that the overall trend for<br />

patient-reported fatigue and CTCAE fatigue was not statistically different<br />

from zero. In Study 2, the mean absolute rate <strong>of</strong> change for fatigue during 6<br />

treatment cycles was greater for Group 1 (4/2) compared to Group 2 (CDD):<br />

0.042 vs. 0.032, respectively; P�0.003, t-test. Conclusions: In Study 1,<br />

pts reported notable fatigue in Cycle 1, which improved or stabilized,<br />

thereafter. In Study 2, Schedule 4/2 was associated with more within-cycle<br />

fluctuation in fatigue. These findings illustrate how SU-associated fatigue<br />

occurs early in therapy and continues with more within-cycle fluctuation<br />

associated with 4/2 dosing. This may help patient-clinician communications<br />

and interventions that support maintaining effective therapy.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


6093 General Poster Session (Board #8B), Mon, 1:15 PM-5:15 PM<br />

Unanticipated hospital admissions in patients undergoing radiotherapy<br />

with or without concurrent chemotherapy: Incidence and predictive factors.<br />

Presenting Author: Nabeel H. Arastu, University <strong>of</strong> North Carolina at Chapel<br />

Hill and Brody School <strong>of</strong> Medicine at East Carolina University, Greenville,<br />

NC<br />

Background: Unanticipated admissions are burdensome for patients and<br />

the healthcare system. An improved understanding <strong>of</strong> their frequency and<br />

predictive factors can inform initiatives to prevent such admissions and<br />

mitigate their associated human and financial costs. Methods: Electronic<br />

medical records <strong>of</strong> 500 patients undergoing external beam radiotherapy<br />

(RT) at our center in 2010 were reviewed. Unanticipated admission within<br />

90 days <strong>of</strong> initiating RT, and associated clinical factors, were recorded.<br />

Chi-squared and uni- and multivariate logistic regression was used to<br />

examine factors associated with admission. Results: Unanticipated admissions<br />

occurred in 20% (101/500) <strong>of</strong> patients, mean length <strong>of</strong> stay was 4<br />

days (range 1-16), and the mean interval between the start <strong>of</strong> RT and<br />

admission was 32 days (0-86 days). The most common indications for<br />

admissions were pain (19% <strong>of</strong> admissions), respiratory distress (15%), and<br />

neurologic symptoms (13%). On univariable analysis, 33% <strong>of</strong> patients<br />

treated for palliative intent were admitted (vs. 16% <strong>of</strong> curative intent<br />

patients, p�0.001), as were 26% <strong>of</strong> patients receiving concurrent chemotherapy<br />

(vs. 17% receiving RT alone, p�0.02). Multivariable analysis<br />

showed treatment intent, chemotherapy, and marital status to be associated<br />

with unplanned admissions (Table). A highly variable rate <strong>of</strong> unanticipated<br />

admission per diagnosis was observed (e.g. 4% for breast, 19% for<br />

GI/GU/GYN/ENT, and 37% for metastatic sites). Conclusions: Rates <strong>of</strong><br />

unanticipated admissions are �20% in patients undergoing RT. Approximately<br />

1/3 <strong>of</strong> patients receiving palliative RT, and more than 1/4 receiving<br />

concurrent chemoradiation, experienced an unplanned admission. Prophylactic<br />

measures should be studied in these high-risk patients to reduce<br />

admission rates, as unplanned admission may be an important quality <strong>of</strong><br />

care indicator in oncology.<br />

Odds <strong>of</strong> an unanticipated admission: Multivariate analysis.<br />

Covariate OR P value<br />

Concurrent chemotherapy (vs. RT alone) 3.4 �0.001<br />

Palliative intent (vs. curative) 2.7 �0.001<br />

Not married (vs. married) 2.3 �0.001<br />

6096 General Poster Session (Board #8E), Mon, 1:15 PM-5:15 PM<br />

Cost-effectiveness <strong>of</strong> adjuvant radiotherapy for older women with early<br />

hormone-receptor positive breast cancer. Presenting Author: Katherine<br />

Elizabeth Reeder-Hayes, University <strong>of</strong> North Carolina Hospital, Chapel Hill,<br />

NC<br />

Background: Radiation therapy (XRT) following breast conserving surgery<br />

decreases local recurrence at the expense <strong>of</strong> additional morbidity and<br />

treatment costs. However, its utility in elderly women at low risk <strong>of</strong><br />

recurrence has been questioned. This study assessed the cost-effectiveness<br />

<strong>of</strong> adding XRT to hormonal therapy (HT) in women over 70 with stage I,<br />

hormone-receptor positive (HR�) breast cancer after breast conserving<br />

surgery. Methods: A decision tree model was used to assess the costs and<br />

benefits <strong>of</strong> XRT � HT versus HT alone in 10,000 women age 70� with<br />

stage I HR� breast cancer. Using a societal perspective, we considered<br />

medical costs and quality <strong>of</strong> life effects <strong>of</strong> initial treatment, recurrences,<br />

and metastatic disease as well as long-term XRT-associated complications<br />

including breast fibrosis, chronic pneumonitis and cardiac disease. Probabilities<br />

<strong>of</strong> recurrence and death were modeled on recent clinical trial<br />

results, while toxicity probabilities were taken from literature review. The<br />

primary health outcome was incremental quality-adjusted life years (QALYs)<br />

gained. One-way and probabilistic sensitivity analyses (PSA) were performed<br />

to assess the sensitivity <strong>of</strong> model results and conclusions to various<br />

parameter estimates. Results: In the base-case scenario, the incremental<br />

cost-effectiveness ratio (ICER) for the addition <strong>of</strong> XRT was $923,017/<br />

QALY. The ICER was highly sensitive to variations in utility weights,<br />

particularly those reflecting patient preferences for initial treatment with or<br />

without XRT and those reflecting the decrement in quality <strong>of</strong> life resulting<br />

from breast fibrosis. In PSA, XRT was associated with lower qualityadjusted<br />

life expectancy at higher cost in 58% <strong>of</strong> simulations. Conclusions:<br />

In women over 70 with stage I HR� breast cancer, the addition <strong>of</strong> XRT to<br />

HT is not cost-effective at a willingness-to-pay threshold <strong>of</strong> $100,000/<br />

QALY, and is associated with little or no improvement in quality-adjusted<br />

life expectancy. Providers should be aware that the cost-effectiveness <strong>of</strong><br />

XRT in this population is strongly influenced by patient preferences<br />

surrounding recurrence and toxicity risks, and should weigh these factors<br />

when making shared decisions with patients.<br />

Health Services Research<br />

405s<br />

6095 General Poster Session (Board #8D), Mon, 1:15 PM-5:15 PM<br />

Association between financial relationships with commercial interests and<br />

research merit at the ASCO <strong>Annual</strong> <strong>Meeting</strong> (AM). Presenting Author:<br />

Beverly Moy, Massachusetts General Hospital, Boston, MA<br />

Background: Financial relationships with commercial interests (COI) are<br />

common in cancer research. There are few data examining the correlation<br />

between COI and research merit. <strong>Meeting</strong> placement prominence (MP) and<br />

peer review score (PRS) are indicators <strong>of</strong> research merit. We examined the<br />

association between ASCO AM abstracts whose authors disclose COI and<br />

both MP and PRS. Methods: We reviewed abstracts presented at the ASCO<br />

AM in 2006 and 2008-2011. We evaluated associations between COI<br />

disclosed by any author and PRS and MP (order <strong>of</strong> prominence: plenary<br />

session (PS), clinical science symposium (CSS), oral presentation (OP),<br />

poster discussion session (PDS), vs. general poster session (GPS)). Chisquare<br />

tests, T-tests, and logistic regressions <strong>of</strong> COI were used to assess<br />

associations with MP, PRS, and year. Results: Of 12,446 total abstracts<br />

accepted for presentation, 78% <strong>of</strong> PS, 59% <strong>of</strong> CSS, 54% <strong>of</strong> OP, 52% <strong>of</strong><br />

PDS, and 39% <strong>of</strong> GPS report at least one COI. Abstracts selected for PS,<br />

CSS, OP, and PDS had more COI compared to those selected for GPS (p �<br />

0.05). Stock ownership COI were more frequently disclosed in PS (30%),<br />

CSS (30%), OP (22%), and PDS (22%) compared to GPS (16%) (p �<br />

0.05). Employment COI were more frequently reported among abstracts<br />

presented at PS (39%), CSS (37%), OP (27%), and PDS (27%) compared<br />

to GPS (21%) (p � 0.05). Consultant COI were more likely to have higher<br />

MP than GPS placement (OR for PS�5.5; CSS�2.4; OP�2.2; PDS�1.9).<br />

Similarly, honoraria COI were more likely to have higher MP than GPS<br />

placement (OR for PS�3.9; CSS�1.8; OP�2.1; PDS�1.7). Better PRS<br />

was associated with COI (OR 0.17; p � 0.05). The relationship <strong>of</strong> better<br />

PRS with any COI strengthened over time from 2006, 2008-2011 (PRS<br />

times year interaction OR�0.65, p�0.001). Conclusions: ASCO abstracts<br />

whose authors report COI have higher merit as measured by MP and PRS.<br />

This suggests a dependence on industry relationships for access to<br />

important data that will lead to prominent cancer research. These relationships<br />

will require further investigation and ongoing management. To our<br />

knowledge, this is the first study examining the scientific merit <strong>of</strong> research<br />

with relation to COI.<br />

6097 General Poster Session (Board #8F), Mon, 1:15 PM-5:15 PM<br />

The impact <strong>of</strong> race/ethnicity concordance between patients and their<br />

navigators in time to diagnostic resolution <strong>of</strong> breast and cervical cancer<br />

screening abnormalities. Presenting Author: Marjory Charlot, Boston University<br />

Medical Center, Boston, MA<br />

Background: Patient navigators have been shown to reduce cancer disparities<br />

among racial/ethnic minorities by improving timely diagnosis and<br />

treatment <strong>of</strong> cancer. For a group <strong>of</strong> navigators who received cultural<br />

competency training, we sought to determine if racial/ ethnic concordance<br />

<strong>of</strong> the navigator and patient improved time to diagnostic resolution <strong>of</strong><br />

cancer screening abnormalities. Methods: Demographic data on 1466<br />

patients and their 23 navigators from the Boston Patient Navigation<br />

Research Program were used to assess concordance by race and ethnicity.<br />

All participants with either breast (n�751) or cervical (n�715) screening<br />

abnormalities were followed up to one year. Kaplan-Meier survival curves<br />

and proportional hazards regression models examined the effect <strong>of</strong> race/<br />

ethnicity concordance on time to definitive diagnosis, adjusting for age,<br />

race, language, economic status, insurance status, and severity <strong>of</strong> screening<br />

abnormality. Analyses were performed separately for the breast and<br />

cervical groups. Results: Of the 1466 patients, 32% were White, 27%<br />

Black, 31% Hispanic, and 10% Asian. Navigators were 61% White, 17%<br />

Black, 13% Hispanic and 9% Asian. Fifty eight percent <strong>of</strong> patientnavigator<br />

pairs were concordant by race/ethnicity. Overall rate <strong>of</strong> diagnostic<br />

resolution <strong>of</strong> cancer screening abnormalities within 365 days was high at<br />

90%. In both the breast and cervical cancer screening groups, racial/ethnic<br />

concordance was not associated with time to diagnostic resolution, with an<br />

aHR 1.03 (95% CI: 0.85, 1.25) for breast patients and HR 1.02 (95% CI:<br />

0.85, 1.21) for cervical patients. Conclusions: Patient-navigator racial/<br />

ethnic concordance is not associated with time to diagnostic resolution <strong>of</strong><br />

cancer screening abnormalities, in part because overall rates <strong>of</strong> diagnostic<br />

resolution were high. Our data suggest that with cultural competency<br />

training, navigators can be equally effective with patients from different<br />

ethnic and cultural backgrounds.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


406s Health Services Research<br />

6098 General Poster Session (Board #8G), Mon, 1:15 PM-5:15 PM<br />

Relationship between Oncotype DX testing and the use <strong>of</strong> chemotherapy in<br />

high-risk patients (pts). Presenting Author: Winston Wong, CareFirst<br />

BlueCross BlueShield, Baltimore, MD<br />

Background: CareFirst BlueCross BlueShield (CFBCBS) insurance network<br />

partnered with Cardinal Health Specialty Solutions (CHSS) to develop a<br />

cancer care pathway for network physicians in 2008. The program included<br />

a recommendation for molecular diagnostic testing with the Oncotype DX<br />

assay for pts with early-stage estrogen receptor-positive breast cancer.<br />

Based on NCCN guidelines, the pathway suggested adjuvant chemotherapy<br />

for all pts with Oncotype DX Recurrence Scores (RS) in the high-risk<br />

category. We aimed to determine the RS risk distribution among pts who<br />

received Oncotype DX testing and assess the patterns <strong>of</strong> care that followed.<br />

Methods: Using data from CFBCBS, CHSS proprietary claims s<strong>of</strong>tware, and<br />

Genomic Health, we retrospectively identified a cohort <strong>of</strong> women with<br />

breast cancer who were treated on the CFBCBS clinical care pathways<br />

program from 8/2008 to 6/2011 and received Oncotype DX testing. We<br />

determined the number <strong>of</strong> pts with a RS value in the low- (RS �18),<br />

intermediate- (RS 18-30), and high-risk (RS �31) groups along with the<br />

number <strong>of</strong> pts who subsequently received chemotherapy in each category.<br />

Results: Of 1174 women who received Oncotype DX testing, 53% <strong>of</strong> pts<br />

were in the low-, 35% in intermediate-, and 12% in the high-risk groups.<br />

Five percent <strong>of</strong> low-, 41% <strong>of</strong> intermediate-, and 74% percent <strong>of</strong> pts in the<br />

high-risk category were treated with chemotherapy. Twenty-six percent <strong>of</strong><br />

pts in the high-risk group did not receive chemotherapy. Conclusions: The<br />

proportionate use <strong>of</strong> chemotherapy in the low and intermediate risk groups<br />

was as expected based on adjuvant chemotherapy guidelines; however, the<br />

underuse <strong>of</strong> chemotherapy in 26% <strong>of</strong> high-risk pts was an unexpected<br />

finding. Further study is needed to determine: (1) why physicians avoided<br />

chemotherapy in 26% <strong>of</strong> high-risk pts; (2) the overall number <strong>of</strong> appropriate<br />

pts who underwent Oncotype DX testing; and, (3) the tumor characteristics<br />

that may have driven the underutilization <strong>of</strong> chemotherapy in the<br />

high-risk population.<br />

6100 General Poster Session (Board #9A), Mon, 1:15 PM-5:15 PM<br />

Quality <strong>of</strong> life <strong>of</strong> elderly patients undergoing concomitant chemoradiation<br />

therapy for head and neck cancers, including assessment <strong>of</strong> geriatric<br />

parameters. Presenting Author: Martine Extermann, H. Lee M<strong>of</strong>fitt Cancer<br />

Canter & Research Institute, Tampa, FL<br />

Background: Data on the quality <strong>of</strong> life <strong>of</strong> older patients undergoing<br />

concomitant chemoradiation therapy (CCRT) for head and neck (H&N)<br />

cancer are very scarce and no study has focused specifically on them.<br />

Furthermore, no study has assessed the contribution <strong>of</strong> geriatric symptoms<br />

to their quality <strong>of</strong> life. Methods: We prospectively assessed patients aged 65<br />

and older undergoing curative intent CCRT for H&N cancers, either alone or<br />

adjuvantly after surgery. We used the Quality <strong>of</strong> life-Radiation Therapy<br />

Instrument (QOL-RTI), with its H&N module (Trotti et al., 1998). In<br />

addition we created a 12-items senior adult questionnaire (SAQ). Patients<br />

were assessed at baseline, at 4 weeks, at the end <strong>of</strong> treatment (EOT), and 2<br />

months after EOT (recovery). Results: Fifty patients were enrolled. Median<br />

age was 69 years (range 65-87). Eighty-two percent <strong>of</strong> patients had locally<br />

advanced stage IV disease. Twenty-eight percent had prior surgery. All<br />

patients were treated with IMRT, 92% at 70 Gy. The most frequent<br />

chemotherapy regimen was cisplatin q3wks (58%), followed by weekly<br />

carboplatin (24%). Patients had on average 4 comorbidities (CIRS-G),<br />

54% <strong>of</strong> them a grade 3 or 4 disease. Forty-four percent were independent<br />

in IADL, and 98% were ECOG PS 0 or 1. The baseline scores were QOL-RTI:<br />

7.72 (SD 1.36), H&N module 7.7 (SD 2.16), SAQ 8.21 (SD 1.54). At EOT,<br />

the scores were 6.22 (1.26), 4.59 (1.82), 7.38 (1.38) respectively, and at<br />

recovery 7.17 (1.25), 6.06 (1.66), 7.96 (1.16). The scores paralleled<br />

functional evolution, as 24% <strong>of</strong> patients had an ECOG PS 2 and 76% were<br />

IADL dependent at EOT; 16% ECOG 2-3 and 55% IADL dependent at<br />

recovery. Cronbach alphas for the 3 QOL measures were 0.88, 0.89, and<br />

0.81, suggesting adequate internal consistency reliability. The SAQ was<br />

low-to-moderately correlated with the other two QOL measures (r�0.22 to<br />

0.59) at different points <strong>of</strong> assessment. Conclusions: Older H&N cancer<br />

patients experience significant impact <strong>of</strong> CCRT on their function, and on<br />

their quality <strong>of</strong> life on all three measures. Most recover after two months,<br />

although some may take longer. A geriatric module adds significant<br />

information to the general QOL-RTI and H&N questionnaires.<br />

6099 General Poster Session (Board #8H), Mon, 1:15 PM-5:15 PM<br />

Utilization <strong>of</strong> bone densitometry and administration <strong>of</strong> bisphosphonates to<br />

prevent osteoporosis in patients with nonmetastatic prostate cancer receiving<br />

anti-androgen therapy. Presenting Author: Muhammad Atif Khan,<br />

University <strong>of</strong> Arkansas for Medical Sciences Hospital, Little Rock, AR<br />

Background: Prostate cancer subjects with PSA relapse who are treated with<br />

androgen deprivation therapy (ADT) are recommended to have baseline and<br />

serial bone densitometry (BD) and receive intravenous bisphsphonates<br />

(BP). Utilization <strong>of</strong> BD and BP therapy was evaluated in a retrospective<br />

SEER Medicare database analysis. Methods: A cohort study <strong>of</strong> men aged<br />

aged � 65 years with a nonmetastatic incident diagnosis <strong>of</strong> prostate cancer<br />

between 2004 and 2008 was conducted. Data were obtained from the<br />

Surveillance, Epidemiology and End Results (SEER) linked Medicare<br />

claims. Medicare claims were used to select prostate cancer cases who had<br />

ever received ADT and intravenous BP as part <strong>of</strong> their treatment. ADT was<br />

defined as an orchiectomy, goserelin, leuprolide, leuprolide implant, or<br />

triptorelin. BD and treatment with pamidronate or zoledronic acid were<br />

identified using Medicare HCPCS codes. One-sided exact binomial test <strong>of</strong><br />

proportion was used to determine if the physician compliance rate is<br />

consistent with 80%; meaning equal to or higher than 80%. Results:<br />

157,974 newly diagnosed prostate cancer cases were identified. Of those,<br />

100,865 were age 65 and above and had no bone metastases. Subjects<br />

who did not have ADT claims were excluded. 30,846 patients were eligible<br />

for analysis. Cases were further stratified by use <strong>of</strong> BD and BP therapy.<br />

Results revealed 86.8% (N�26,774) on ADT did not receive either a BD or<br />

intravenous BP therapy. Approximately 2.9% (N�885) <strong>of</strong> the cases on ADT<br />

received BP treatment without ever receiving a BD. 9.3% (N�2,863) <strong>of</strong> the<br />

cases on ADT received a BD without receiving intravenous BP, while only<br />

1.05% (N�324) <strong>of</strong> the cases on ADT received both a BD and BP. A<br />

compliance rate <strong>of</strong> 1.05%, those patients receiving both bone densitometries<br />

to screen for bone loss and preventive therapy, was well below the<br />

expected rate <strong>of</strong> 80%. Conclusions: Contrary to the recommendations, BD<br />

assessment and BP use is under utilized in men receiving ADT for non<br />

metastatic prostate cancer. Education <strong>of</strong> practicing physicians regarding<br />

better screening and preventative measures for osteoporosis in this population<br />

is warranted.<br />

6101 General Poster Session (Board #9B), Mon, 1:15 PM-5:15 PM<br />

Long-term central venous catheter use among cancer patients in administrative<br />

claims data. Presenting Author: Allison Nicole Lipitz Snyderman,<br />

Memorial Sloan-Kettering Cancer Center, New York, NY<br />

Background: This study’s objective was to examine administrative claims<br />

data’s capacity to serve as a surveillance tool for long-term catheter use and<br />

related bloodstream infections among cancer patients. Population-based<br />

estimates and efforts to track catheter use and infections for this group are<br />

limited. As such, we sought to explore catheter use documentation using a<br />

cohort <strong>of</strong> colorectal cancer patients. Methods: We performed a retrospective<br />

analysis using the population-based SEER-Medicare dataset for patients<br />

66 years or older diagnosed with colorectal cancer in 2005-2007 (n �<br />

54,870). Insertions and removals <strong>of</strong> long-term central venous catheters<br />

(i.e., tunneled, ports/pumps, peripherally inserted central catheter [PICCs])<br />

were identified by billing codes within 2 years <strong>of</strong> diagnosis. Factors<br />

associated with catheter use were identified in multivariable logistic<br />

regression analysis. Results: Findings were consistent with clinical expectations.<br />

A total <strong>of</strong> 11,775 patients (21%) had at least one documented<br />

long-term catheter insertion within 2 years <strong>of</strong> diagnosis, 19% within 6<br />

months. Of those with catheters, approximately 25% had more than one<br />

insertion. Sixty-eight percent had at least one port/pump, 8% at least one<br />

tunneled catheter, and 33% at least one PICC. Seventy-four percent <strong>of</strong><br />

catheterized patients had IV chemotherapy. Of patients stages I, II, III, and<br />

IV, 10%, 18%, 37%, and 39% had catheters, respectively. Adjusted for<br />

other factors, patients with catheters were significantly more likely to be<br />

younger, female, black (vs. white), and have comorbidities (vs. none<br />

documented in year prior to diagnosis), compared to patients without<br />

catheters. By the end <strong>of</strong> the study, 34% <strong>of</strong> patients with ports/pumps had<br />

documented removal and 42% died (for patients with tunneled catheters,<br />

28% and 52%, respectively). Conclusions: Findings support the use <strong>of</strong><br />

claims data to capture long-term catheter use in cancer patients, providing<br />

a foundation for exploration <strong>of</strong> its capacity to capture infections. Given<br />

extensive documented catheter use (21%), infections may be a significant<br />

problem. Claims data may <strong>of</strong>fer a low-burden method for surveillance and<br />

study, aiding the development <strong>of</strong> targeted initiatives.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


6102 General Poster Session (Board #9C), Mon, 1:15 PM-5:15 PM<br />

Assessment <strong>of</strong> cancer registries (CR) in low- and middle-income countries<br />

(LMCs). Presenting Author: Leah L. Zullig, Department <strong>of</strong> Health Policy and<br />

Management, University <strong>of</strong> North Carolina at Chapel Hill, Chapel Hill, NC<br />

Background: CRs are crucial for cancer control, yet few global standards<br />

exist. This study identifies characteristics <strong>of</strong> quality CRs in LMCs and<br />

globally. Methods: A Medline search was conducted in PubMed to identify<br />

peer-reviewed articles describing: 1) CR development and functionality 2)<br />

types <strong>of</strong> CRs and 3) implementation, development and utility <strong>of</strong> CRs in<br />

LMCs. We examined articles describing CRs and assessed benchmarking<br />

data that could be used to define quality characteristics. Full-text,<br />

English-language articles were reviewed. References cited were searched<br />

for additional articles. We categorized characteristics into 4 domains:<br />

comparability, completeness, validity and timeliness. Results: The literature<br />

search yielded 16 key characteristics within 4 domains that may define<br />

high quality CRs. In the “comparability” domain, the key characteristic was<br />

use <strong>of</strong> standard definitions. CRs in the US, Europe and China generally<br />

adhere to the International Classification <strong>of</strong> Diseases for Oncology and<br />

histological verification <strong>of</strong> disease; in 2007 only 40% <strong>of</strong> African registries<br />

did so. In the “completeness” domain the key characteristic was population<br />

coverage. African CRs monitor 8% <strong>of</strong> the population, the Costa Rican<br />

registry covers 90%, US and Madras cancer registries reach 96%. In the<br />

“validity” domain the key criterion was pathologic diagnosis confirmation.<br />

Most LMC CRs are not pathology-based. CRs in wealthier settings like Hong<br />

Kong report histologic confirmation in �85% <strong>of</strong> cases. In the “timeliness”<br />

domain standards for timely data reporting are largely undocumented.<br />

Conclusions: No consensus exists on characteristics <strong>of</strong> quality CRs in a<br />

global context. The current study provides an initial set <strong>of</strong> metrics. A Delphi<br />

panel <strong>of</strong> international experts is planned to further address this. Based on<br />

this literature review, CRs in LMCs have limited reporting, validation and<br />

regional population coverage.<br />

6104 General Poster Session (Board #9E), Mon, 1:15 PM-5:15 PM<br />

Differential receipt <strong>of</strong> sentinel lymph node biopsy within practice-based<br />

research networks. Presenting Author: Anne-Marie Meyer, University <strong>of</strong><br />

North Carolina at Chapel Hill, Chapel Hill, NC<br />

Background: Sentinel lymph node biopsy (SLNB) for breast cancer was<br />

introduced into clinical practice in the late 1990s as an alternative to<br />

axillary lymph node dissection (ALND). Provider-based research networks<br />

(PBRNs) are believed to promote diffusion <strong>of</strong> innovations like SLNB into<br />

clinical practice; however, evidence <strong>of</strong> this association is limited. This<br />

study examines the diffusion <strong>of</strong> SLNB for early-stage breast cancer through<br />

the Community <strong>Clinical</strong> Oncology Program (CCOP), a community-based<br />

PBRN. Methods: We identified women undergoing breast conserving<br />

surgery with axillary staging for stage I or II breast cancer between January<br />

2000 and December 2003 using Surveillance Epidemiology and End<br />

Results-Medicare data (n�6,226). The primary outcome was receipt <strong>of</strong><br />

SLNB vs. ALND, and exposure was care received from CCOP physicians or<br />

institutions between diagnosis and surgery. Exposure was quantified as<br />

both a binary measure <strong>of</strong> ever seeing a CCOP, and as a proportion <strong>of</strong> all their<br />

claims associated with a CCOP. Covariates included race, age, marital<br />

status, education, Medicaid eligibility, comorbidity, tumor grade, stage,<br />

estrogen receptor status, year <strong>of</strong> diagnosis, SEER region, and other<br />

institutional characteristics such as NCI center designation, cooperative<br />

group, and medical school affiliation. Multivariable generalized linear<br />

modeling with generalized estimating equations was used to measure<br />

association between CCOP exposure and receipt <strong>of</strong> SLNB. Results: Women<br />

who received a higher proportion <strong>of</strong> their care from a CCOP-affiliated<br />

physician or hospital were more likely to receive SLNB. A 10% increase in<br />

the proportion <strong>of</strong> CCOP-affiliated claims was associated with a greater odds<br />

<strong>of</strong> receiving SLNB (OR 1.14; 95% CI 1.08, 1.20), after controlling for<br />

covariates. Similarly, sensitivity analysis <strong>of</strong> the binary indicator <strong>of</strong> CCOP<br />

exposure also showed greater odds <strong>of</strong> receiving SLNB (OR 1.32; 95%CI<br />

1.01, 1.74). Conclusions: The quality <strong>of</strong> cancer care delivered in community<br />

settings can be influenced by provider-based research networks. Our<br />

findings contribute to the growing body <strong>of</strong> evidence that community-based<br />

PBRNs can facilitate adoption <strong>of</strong> cancer innovations outside <strong>of</strong> academic<br />

medical centers.<br />

Health Services Research<br />

407s<br />

6103 General Poster Session (Board #9D), Mon, 1:15 PM-5:15 PM<br />

Development <strong>of</strong> a disease-specific measure <strong>of</strong> quality <strong>of</strong> life in myelodysplastic<br />

syndromes (MDS): The “QUALMS-1”. Presenting Author: Gregory Alan Abel,<br />

Dana-Farber Cancer Institute, Boston, MA<br />

Background: Studies assessing the quality <strong>of</strong> life (QoL) experienced by patients<br />

with MDS have almost universally relied upon generic measures; however,<br />

disease-specific QoL tools can allow for more sensitive assessments <strong>of</strong> the<br />

impact <strong>of</strong> changes in disease status. Methods: Using a clinical impact method <strong>of</strong><br />

instrument development, individual and combined focus groups were conducted<br />

with 32 members <strong>of</strong> our institution’s MDS community (patients, their caregivers,<br />

and health care providers) to identify MDS-relevant QoL domains and associated<br />

question topics. <strong>Part</strong>icipants’ rankings <strong>of</strong> the importance <strong>of</strong> the domains and<br />

question topics were compared, collapsing patients/caregivers into one group<br />

and physicians/other providers into another. A draft scale was constructed taking<br />

a greater number <strong>of</strong> questions from the more highly-ranked domains. Results:<br />

“Fatigue” was ranked as the most important domain (see table). None <strong>of</strong> the 12<br />

domains were ranked significantly differently by patients/caregivers versus<br />

providers. The two groups ranked 5 <strong>of</strong> 60 question topics differently: “Too tired<br />

for routine tasks” (providers higher; p� .05); “limited availability <strong>of</strong> support<br />

beyond the family” (providers higher; p� .02); “organizing life around transfusion/MD<br />

appointments” (providers higher, p� .03); “bruising” (patients/<br />

caregivers higher, p� .05) and “anger over diagnosis” (providers higher, p�<br />

.03). Conclusions: A high level <strong>of</strong> agreement in the rankings <strong>of</strong> domains and<br />

question topics between MDS patients/caregivers and providers suggests that<br />

the QoL experience <strong>of</strong> MDS patients is consistently compromised. The resulting<br />

38-item draft QUALMS-1 tool is now being piloted (cognitive debriefing and<br />

behavioral coding) in a new cohort <strong>of</strong> MDS patients, with the ultimate goal <strong>of</strong><br />

validation in a multi-institutional setting.<br />

Domain<br />

Mean ranking<br />

(n�32; 1� most important,<br />

12� least important)<br />

Fatigue 1.4<br />

Emotional health 3.3<br />

Uncertainty/sense <strong>of</strong> control 4.6<br />

Disease information 5.5<br />

Disruption <strong>of</strong> life/logistics <strong>of</strong> care 5.7<br />

Family relationships 6.6<br />

Symptoms other than fatigue 6.6<br />

Financial concerns 7.0<br />

Awareness <strong>of</strong> positives/hope 7.1<br />

Social/role functioning 7.9<br />

Sexual health 11.0<br />

Appearance/self-image 11.0<br />

6105 General Poster Session (Board #9F), Mon, 1:15 PM-5:15 PM<br />

Comparative effectiveness <strong>of</strong> erlotinib versus no treatment for advanced<br />

non-small cell lung cancer patients in the Veterans Administration.<br />

Presenting Author: Benjamin Kim, UCSF School <strong>of</strong> Medicine, San Francisco,<br />

CA<br />

Background: Erlotinib (E) is FDA-approved for second-line treatment <strong>of</strong><br />

unselected advanced non-small cell lung cancer (aNSCLC) patients.<br />

<strong>Clinical</strong> trials have shown increased response rates with E in aNSCLC<br />

patients having East Asian ethnicity, female gender, never-smoking history,<br />

or adenocarcinoma histology. The objective <strong>of</strong> this study was to compare<br />

the real-world effectiveness <strong>of</strong> E versus no treatment (NT) for first- and<br />

second-line treatment <strong>of</strong> aNSCLC patients in the Veterans Administration<br />

(VA). Methods: All incident aNSCLC cases diagnosed in the VA during 2007<br />

were identified through the VA Central Cancer Registry, a subset <strong>of</strong> which<br />

underwent medical record abstraction through the VA External Peer Review<br />

Program. Abstracted records were then merged with national VA Decision<br />

Support System pharmacy and Corporate Data Warehouse encounters data<br />

through 2010. Treatment lines were determined through adaptation <strong>of</strong> a<br />

published algorithm. The impact <strong>of</strong> E versus NT on overall survival for each<br />

treatment line was then evaluated using 1:N greedy propensity score<br />

matching (PSM) and 2-stage residual inclusion (2SRI) estimation. Results:<br />

Among 1965 aNSCLC patients assessed for first-line treatment, 78 (4%)<br />

received E and 782 (40%) received NT; among 1124 assessed for<br />

second-line treatment, 128 received E (7%) and 638 (33%) received NT.<br />

1:3 PSM <strong>of</strong> E cases to NT controls showed no difference in overall survival<br />

by Kaplan-Meier estimation in the first- (median 2.6 vs. 2.9 mos.,<br />

p-value�0.86) and second-line (3.4 vs. 3.2 mos., p�0.85) settings.<br />

Chemotherapy use by Veterans Integrated Service Network was selected as<br />

the instrumental variable for 2SRI. Expected mean survival <strong>of</strong> E versus NT<br />

was similarly not statistically prolonged in the first- (E:NT survival time ratio<br />

2.55, p�0.35) and second-line (2.49, p�0.32) settings. Conclusions: For<br />

a population with few aNSCLC patients having 3 out <strong>of</strong> the 4 clinicopathologic<br />

characteristics associated with improved response, E did not appear<br />

to confer a survival advantage over NT. Linking mutation results to<br />

observational datasets may improve comparative effectiveness research <strong>of</strong><br />

personalized medicine in oncology.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


408s Health Services Research<br />

6106 General Poster Session (Board #9G), Mon, 1:15 PM-5:15 PM<br />

Advanced gastrointestinal cancer patients’ perceptions <strong>of</strong> prognosis and<br />

goals <strong>of</strong> treatment. Presenting Author: Areej Raed El-Jawahri, Massachusetts<br />

General Hospital, Boston, MA<br />

Background: Patients with advanced cancer require accurate perception <strong>of</strong><br />

their illness in order to make informed decisions regarding their care.<br />

However, little is known about the accuracy <strong>of</strong> these patients’ illness and<br />

prognostic understanding. The objectives <strong>of</strong> this study are to 1) examine<br />

prognostic understanding in patients with advanced gastrointestinal (GI)<br />

cancers, 2) assess patient preferences for prognostic information, and 3)<br />

explore associations <strong>of</strong> perceptions with quality <strong>of</strong> life (QOL) and mood.<br />

Methods: Cross-sectional study <strong>of</strong> 50 patients within 6-12 weeks post<br />

diagnosis <strong>of</strong> advanced GI cancers (gastric, esophageal, pancreatic, and<br />

hepatobiliary). We assessed patients’ perceptions <strong>of</strong> prognosis with a<br />

15-item questionnaire, the Perception <strong>of</strong> Treatment and Prognosis Questionnaire.<br />

QOL and mood were assessed using the Functional Assessment<br />

<strong>of</strong> Cancer Therapy-General (FACT-G) and hospital anxiety and depression<br />

scales (HADS), respectively. Results: We enrolled 50/62 (80%) consecutive<br />

eligible patients within an 11-month period. 50% (25/50) <strong>of</strong> participants<br />

responded that the primary goal <strong>of</strong> their cancer treatment was to “cure their<br />

cancer.” Similarly, 54% (27/50) reported that they were “somewhat” to<br />

“extremely likely” to be cured from their cancer. Only 22% (10/49)<br />

reported having a discussion about their end-<strong>of</strong>-life care preferences with<br />

their oncologist. 76% (38/50) reported wanting to know as many details as<br />

possible about their cancer diagnosis and treatment and 64% (32/50)<br />

rated this information as “extremely important.” Patients who perceived<br />

knowing about their prognosis as “extremely helpful” reported a better QOL<br />

(p � 0.009), lower symptoms <strong>of</strong> anxiety (p � 0.02) and depression (p �<br />

0.02). Conclusions: Although the majority <strong>of</strong> patients report that they desire<br />

detailed information about their prognosis, half incorrectly perceived their<br />

cancer as curable and the majority did not discuss their end-<strong>of</strong>-life care<br />

preferences with their oncologist. Patients who found learning about their<br />

prognosis to be extremely helpful reported better QOL and mood. Studies <strong>of</strong><br />

interventions to increase advanced cancer patients’ knowledge <strong>of</strong> their<br />

prognosis and to encourage end-<strong>of</strong>-life discussions are warranted.<br />

6108 General Poster Session (Board #10A), Mon, 1:15 PM-5:15 PM<br />

Assessing the clinical significance <strong>of</strong> real-time quality <strong>of</strong> life data in cancer<br />

patients treated with radiation therapy. Presenting Author: Michele Y.<br />

Halyard, Mayo Clinic, Scottsdale, AZ<br />

Background: This pilot study evaluated whether providing clinicians with<br />

patient(pt) QOL results and symptom management pathways linked to QOL<br />

domains at the time <strong>of</strong> clinical appointment would result in improvement in<br />

QOL and treatment (tx) satisfaction. The objective was to obtain preliminary<br />

effect size estimates and logistical evidence for design <strong>of</strong> a larger,<br />

definitive trial. Methods: Oncology pts receiving 5-7 weeks <strong>of</strong> radiotherapy<br />

(RT) electronically completed QOL assessments (LASA) at baseline and<br />

biweekly prior to seeing clinicians. Was It Worth It (WIWI) and Interpersonal<br />

Patient-Provider Relationship (IPPRS) were measured at tx end. Pt endpoints<br />

(pro-rated primary endpoint LASA area under the curve (AUC), LASA<br />

changes from baseline, and WIWI responses) and clinician endpoints<br />

(IPPRS) were compared between the control group (Phase 1: no QOL<br />

feedback) and the intervention group (Phase 2: QOL feedback) via<br />

Wilcoxon, Chi-square and Fisher Exact tests. There was 80% power to<br />

detect a 10 point difference in average AUC. Results: 148 pts enrolled (79<br />

Phase 1, 69 Phase 2) from 11/28/2008 to 09/20/2011 (sites GI (27%),<br />

Lung (22%) and Head and Neck (52%)). 68% received RT and chemo.<br />

There were consistently moderate effect sizes observed but no statistically<br />

significant differences in any AUC nor end <strong>of</strong> tx change from baseline<br />

scores. 20% fewer pts in phase 2 reported clinical deficits in overall QOL<br />

(pain). In pts receiving 7 weeks <strong>of</strong> RT, end <strong>of</strong> tx average overall QOL, mental<br />

well-being (WB), physical WB and pain severity were significantly better in<br />

Phase 2 pts. WIWI results showed 76% found participation worthwhile,<br />

95% would participate again, and 92% would recommend the study to<br />

others. No differences between groups were found in communication<br />

between clinicians and pts (IPPRS). Conclusions: Preliminary estimates<br />

indicate potentially clinically significant improvements <strong>of</strong> moderate effect<br />

size in mental and physical WB and pain severity when clinicians received<br />

QOL real time with symptom management pathways. Further study is<br />

warranted in larger trial setting.<br />

6107 General Poster Session (Board #9H), Mon, 1:15 PM-5:15 PM<br />

Association between baseline physical function and comorbidity status<br />

with patient-reported quality <strong>of</strong> life after prostate cancer treatments:<br />

Combined analysis <strong>of</strong> two prospective cohort studies. Presenting Author:<br />

Ronald C. Chen, University <strong>of</strong> North Carolina at Chapel Hill, Chapel Hill, NC<br />

Background: Treatment-related bowel, urinary, and sexual dysfunction in<br />

prostate cancer patients varies by treatment type, baseline function and<br />

other patient factors. To better predict patient outcomes after treatment,<br />

we examined the impact <strong>of</strong> comorbidity on these quality <strong>of</strong> life (QOL)<br />

outcomes in a secondary data analysis <strong>of</strong> two pooled, prospective cohort<br />

studies. Methods: A total <strong>of</strong> 697 patients from 3 academic hospitals who<br />

received radical prostatectomy, external beam radiation, or brachytherapy<br />

were included. Using a validated instrument, patients reported bowel,<br />

urinary, and sexual symptoms pretreatment, and at 3, 12, 24, and 36<br />

months after treatment. Baseline physical function was measured by the<br />

physical component summary score (PCS) <strong>of</strong> the SF-12 using patient<br />

report. Comorbidity as measured by the Index <strong>of</strong> Co-Existent Disease<br />

(ICED) was obtained from medical record review. Repeated QOL measurements<br />

were analyzed using a mixed modeling method, by random coefficient<br />

modeling. Separate models were built for each outcome using bowel,<br />

urinary, and sexual scale scores at each time point.Covariates in all models<br />

included baseline age, education, ICED, and PCS. Results: Approximately<br />

70% <strong>of</strong> patients had one or more comorbid conditions at baseline. After<br />

adjusting for age and education in mixed-models, we found baseline<br />

comorbidity was independently associated with more sexual dysfunction<br />

(p�.001) and urinary incontinence (p�.03). Worse baseline physical<br />

functioning was independently associated with more bowel problems<br />

(p�.001) and sexual dysfunction (p�.001). There were no treatment by<br />

comorbidity or physical functioning interactions. Conclusions: Comorbidity<br />

and worse physical functioning at baseline are significantly associated with<br />

poorer bowel, urinary, and sexual function after treatment for prostate<br />

cancer, but the associations do not appear to differ by treatment. Patients<br />

with comorbidity recovered more slowly. This information may help patients<br />

and their physicians anticipate outcomes after surgical and radiation<br />

treatments.<br />

6109 General Poster Session (Board #10B), Mon, 1:15 PM-5:15 PM<br />

Changes in adjuvant breast cancer chemotherapy regimen selection over<br />

time in the community. Presenting Author: Debra A. Patt, Texas Oncology,<br />

US Oncology, Austin, TX<br />

Background: Adjuvant breast cancer (BC) chemotherapy (CT) treatment has<br />

evolved over time due to improved understanding <strong>of</strong> risk conveyed by<br />

pathology and patient (pt) characteristics, as well as emergence <strong>of</strong> new and<br />

mature data for survival and toxicity. We aimed to evaluate how CT regimen<br />

selection has changed in recent years in various subgroups. Methods: Using<br />

iKnowMed EHR data from The US Oncology Network, we retrospectively<br />

identified female pts diagnosed with stage I-III BC between 1/2007 and<br />

12/2010 at practices with EHR data available at time <strong>of</strong> diagnosis. Pts with<br />

�5 <strong>of</strong>fice visits, those with a second primary or previous cancer diagnoses<br />

were excluded. Age, ER, HER2, nodal status, stage and year <strong>of</strong> diagnosis<br />

were captured. CT utilization was determined by the number <strong>of</strong> pts who<br />

received CT within 6 months <strong>of</strong> their diagnosis. <strong>Clinical</strong> trial pts were<br />

included. Regimens were categorized by the initial CT title and drugs<br />

assigned. Pts with metastatic regimens were excluded. CT regimens were<br />

analyzed by subgroups and trended over time. Results: During the time<br />

period, 26,095 stage I-III BC pts were identified. A CT regimen within 6<br />

months <strong>of</strong> diagnosis was documented in 56% <strong>of</strong> pts. CT utilization was<br />

83% in HER2� pts, 85% in ER-/HER2- pts, and 45% in ER�/HER2- pts.<br />

CT utilization decreased overall with increasing pt age (71%, 64%, 51%,<br />

33%, and 13% for pts in their 4th ,5th ,6th ,7th and �8th decade <strong>of</strong> life). In<br />

HER2� pts, use <strong>of</strong> non-anthracycline containing regimens increased from<br />

26 to 62%, and anthracyline-taxane combination regimens decreased from<br />

33 to 15%. In HER2-/ER� pts, the most used non-anthracycline regimen<br />

was docetaxel � cyclophosphamide (TC) at 41%. Anthracycline-taxane<br />

combinations were used more <strong>of</strong>ten in the HER2-/ER- group (32%).<br />

Conclusions: In the 4 year study period, this data suggests that ER and<br />

HER2 status may drive chemotherapy choice more than nodal status.<br />

Anthracycline containing regimens are being used less <strong>of</strong>ten possibly due<br />

to similar efficacy but less cardiac toxicity, particularly when combined<br />

with trastuzumab. These results suggest a change away from anthracyclines<br />

in specific subgroups with the controversy over the benefits <strong>of</strong> that<br />

change unsettled, and cost implications yet unquantitated.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


6110 General Poster Session (Board #10C), Mon, 1:15 PM-5:15 PM<br />

Frequency <strong>of</strong> cognitive impairment (CI) in elderly patients (pts) suffering<br />

from malignancies and impact on therapeutic decision. Presenting Author:<br />

Thibault De La Motte Rouge, Medical Oncology Department, AP-HP,<br />

Salpetriere Hospital, University Paris VI, Paris, France<br />

Background: Therapeutic decision remains complex when a cancer is<br />

diagnosed in elderly pts. Our aim was to evaluate the frequency <strong>of</strong> CI in this<br />

population and its impact on therapeutic decision. Methods: An oncogeriatric<br />

evaluation including Comprehensive Geriatric Assessment (CGA) is<br />

systematically performed for all elderly cancer pts referred in our unit. We<br />

reviewed data <strong>of</strong> all pts assessed by geriatric oncologist at our institution<br />

from January 2009 to June 2011. Results: 378 pts were identified, among<br />

them a CI was noted in 87. Median age was 84 years (range 71 –94), 70 %<br />

� 80 years. Most <strong>of</strong> the pts (78/87) were referred at the time <strong>of</strong> diagnosis.<br />

Metastatic disease was diagnosed in 32 pts (52%) and Diffuse Large B Cell<br />

Lymphoma stage III or IV in 21 pts (81 %). In 41 pts, CI was already<br />

diagnosed: Alzheimer disease (AD) (n�38) and Vascular Dementia (n�3).<br />

CGA help to identify CI in 46 additional pts: AD (n�36); Vascular Dementia<br />

(n�2) and Mild Cognitive Impairment (n�8). 45/87 pts (52 %) were<br />

dependant for at least one activity <strong>of</strong> daily living (ADL). As a result <strong>of</strong> CGA<br />

and benefit/risk oncologic assessment, best supportive care was recommended<br />

in 12 pts. Among them, only 4 pts presented with advanced<br />

metastatic disease (main reason for palliative care). Pts in whom “best<br />

supportive care” decision (n�12) was recommended were more dependants<br />

than those who received specific anticancer therapy (n�75):<br />

dependence for at least 2 ADL: 10/12 pts (83%) versus 16/75 (21%); and<br />

presented more AD already diagnosed (11/12 versus 30/75). In the<br />

remaining 75 pts, specific cancer therapy was proposed, including chemotherapy<br />

(n�67), surgery (n�5), radiotherapy (n�3) and hormonotherapy<br />

(n�9). Treatment was initiated as recommended in all but 4 pts (best<br />

supportive care decision taken following discussion with pts and relatives).<br />

During the follow-up, only 11/75 pts needed to be placed in nursing home<br />

because <strong>of</strong> loss <strong>of</strong> autonomy. A survival � 1 year was observed in 27/75<br />

(36%) pts. An update <strong>of</strong> cognitive performance will be presented.<br />

Conclusions: Our data support that even if CI is frequent in elderly pts with<br />

malignancies, specific anticancer therapy remains feasible and should be<br />

considered in most elderly pts with CI.<br />

6112 General Poster Session (Board #10E), Mon, 1:15 PM-5:15 PM<br />

An integrated analysis <strong>of</strong> comprehensive geriatric assessment (CGA) in elderly<br />

patients with non-small cell lung cancer (NSCLC) (JCOG1115-A). Presenting<br />

Author: Hiroshi Katayama, JCOG Operations Office, National Cancer Center,<br />

Tokyo, Japan<br />

Background: The number <strong>of</strong> elderly NSCLC patients is increasing all over the<br />

world. Actual age and ECOG PS is not always efficient to select �fit-elderly� for<br />

chemotherapy. We integrated the data from two phase III trial (JCOG0207,<br />

JCOG0803/WJOG4307L) and evaluated 4 CGA items as well as age and PS to<br />

determine whether they would predict survival, response and toxicities. Methods:<br />

Eligibility criteria <strong>of</strong> both trials included histologically or cytologically proven<br />

NSCLC; stage III/IV; age 70 or more; ECOG PS <strong>of</strong> 0-1; unfit for bolus cisplatin; no<br />

prior treatment; adequate organ function. Patients were randomized to receive<br />

either weekly docetaxel (D) or D plus cisplatin (DP) in J0207, and either<br />

3-weekly D or DP in J0803/W4307L. This study included all eligible pts for<br />

whom all <strong>of</strong> 4 CGA items (Activity <strong>of</strong> Daily Living [ADL], Instrumental Activity <strong>of</strong><br />

Daily Living [IADL], Mini-Mental State Examination [MMSE], Geriatric Depression<br />

Scale [GDS-15]) were assessed before treatment. Primary endpoint was<br />

overall survival (OS). Survival curves are estimated by Kaplan-Meier method and<br />

multivariate analysis for OS adjusting baseline factors were performed by using<br />

stratified Cox regression model. Results: This analysis included 331 pts (J0207/<br />

J0803-W4307L: 105/226) in total. The result <strong>of</strong> multivariate analysis for OS<br />

was shown in the table. After adjusting baseline factors, MMSE as well as sex and<br />

PS was significantly associated with OS. None <strong>of</strong> the CGA items were associated<br />

with response and toxicities. Conclusions: It is not actual age but MMSE and PS<br />

that is useful for predicting prognosis <strong>of</strong> elderly pts. MMSE should be taken into<br />

consideration to determine the indication <strong>of</strong> chemotherapy for elderly pts.<br />

mOS Multivariate analysis<br />

n<br />

(months) HR 95%CI p<br />

Sex<br />

Male 243 12.8 1<br />

Female<br />

Stage<br />

88 24.1 0.44 0.30-0.66 �.001<br />

III 102 15.4 1<br />

IV<br />

PS<br />

229 14.5 1.25 0.89-1.75 0.194<br />

0 119 21.2 1<br />

1<br />

Age<br />

212 11.7 1.66 1.18-2.32 0.0033<br />

70-74 96 17.1 1<br />

75-79 181 15.0 0.96 0.67-1.37 0.810<br />

80-<br />

ADL<br />

54 11.3 1.45 0.91-2.29 0.119<br />

20 300 15.0 1<br />

-19<br />

IADL<br />

31 11.3 1.15 0.68-1.95 0.591<br />

5 267 15.0 1<br />

-4<br />

MMSE<br />

64 14.1 1.01 0.69-1.49 0.956<br />

30 133 21.2 1<br />

-29<br />

GDS-15<br />

198 13.5 1.46 1.05-2.03 0.025<br />

-4 207 14.8 1<br />

5- 124 15.0 0.84 0.61-1.15 0.268<br />

Health Services Research<br />

409s<br />

6111 General Poster Session (Board #10D), Mon, 1:15 PM-5:15 PM<br />

Cancer survivorship outcomes in immigrants. Presenting Author: Phyllis<br />

Noemi Butow, University <strong>of</strong> Sydney, Sydney, Australia<br />

Background: Immigration is increasing world-wide. Cancer survivorship is<br />

now recognised as a period <strong>of</strong> difficult adjustment for all patients, and<br />

possibly more so for immigrants. We explored disparities in quality <strong>of</strong> life<br />

outcomes for immigrant (IM) versus Anglo-Australian (AA) cancer survivors.<br />

Methods: In a cross-sectional design, cancer survivors were recruited<br />

through the New South Wales, Queensland and Victorian Cancer Registries<br />

in Australia. IM participants, their parents and grandparents were born in a<br />

country where Chinese, Greek, or Arabic is spoken and spoke one <strong>of</strong> those<br />

languages. AAs were born in Australia and spoke English. All were<br />

diagnosed with cancer 1-3 years previously. Questionnaires (completed in<br />

preferred language) included the Hospital Anxiety and Depression Scale<br />

(anxiety/ depression), FACT-G (quality <strong>of</strong> life) and Supportive Care Needs<br />

Survey (unmet needs). Outcomes were compared between AA and IM<br />

groups in adjusted regression models that included age, gender, socioeconomic<br />

status, education, marital status, religion, time since diagnosis<br />

and cancer type (prostate, colorectal, breast and other). Results: There were<br />

599 participants (response rate 41%). Consent was unrelated to demographic<br />

and disease variables. AA and IM groups were similar except that<br />

immigrants had higher proportions in the low and highly educated groups (p<br />

� 0.0001), and higher socioeconomic status (p � 0.0003). In adjusted<br />

analyses (see table), IMs had clinically significant higher depression<br />

(possible range 0-21), greater unmet information and physical needs, and<br />

lower quality <strong>of</strong> life than AAs. The possible range for the latter three is<br />

0-100. Conclusions: Immigrants experience poorer outcomes in cancer<br />

survivorship, even after adjusting for socio-economic, demographic and<br />

disease differences. Interventions are required to improve their adjustment<br />

after cancer. Results highlight areas <strong>of</strong> unmet need that might be better<br />

addressed by the health system (particularly with regard to provision <strong>of</strong><br />

information and support.<br />

Outcome<br />

(possible range) Immigrant Anglo-Australian P<br />

Depression<br />

4.6 2.7 �.0001<br />

(0-21)<br />

Unmet information needs 19.2 10.7 �.0001<br />

(0-100)<br />

Unmet physical needs<br />

14.8 10.2 .0006<br />

(0-100)<br />

Quality <strong>of</strong> life<br />

76.6 82.7 �.0001<br />

(0-100)<br />

6113 General Poster Session (Board #10F), Mon, 1:15 PM-5:15 PM<br />

Implications <strong>of</strong> smoking for quality <strong>of</strong> life and illness perceptions <strong>of</strong> lung<br />

cancer patients. Presenting Author: Sarah Danson, Academic Unit <strong>of</strong><br />

<strong>Clinical</strong> Oncology, Weston Park Hospital, Sheffield, United Kingdom<br />

Background: Adverse publicity about smoking may lead to feelings <strong>of</strong> guilt<br />

among lung cancer patients or pessimism about their future, potentially<br />

compromising health-related quality <strong>of</strong> life (QoL) and adherence with<br />

medical advice. QoL is an important outcome in clinical trials, particularly<br />

where survival rates are low. We aim to compare QoL and illness perceptions<br />

<strong>of</strong> lung cancer patients depending on smoking history. Methods:<br />

<strong>Clinical</strong> data, measures <strong>of</strong> QoL (EORTC-QLQ-C30�LC13) and illness<br />

perceptions (Brief IPQ – includes items to assess perceived severity,<br />

causality, timeline, understanding, emotional impact, and control over<br />

illness) were completed by 190 newly diagnosed lung cancer patients<br />

(Non-small cell: 75%; Mean age: 68 years, range: 48-85 years; 58%<br />

males). Of these, 24% were current smokers, 69% former smokers, and<br />

7% never smoked. Results: Although overall QoL was similar between the<br />

three smoker groups, there was a significant effect <strong>of</strong> smoking status on<br />

QoL subscale emotional function (EF)(F(2,168)�4.08,p�.019). Those<br />

who never smoked had significantly higher EF than current smokers<br />

(p�.03). There was also a significant effect <strong>of</strong> smoking history on cough<br />

(F(2,166);�5.40 ,p�.005) with smokers reporting significantly greater<br />

levels than former smokers (p�.004). Smokers were more likely than<br />

former smokers (p�.015) to attribute their lung cancer to smoking<br />

(F(2,161)�16.49,p�.000). Furthermore, there was a significant effect <strong>of</strong><br />

smoking on the perceived timeline <strong>of</strong> illness (F(2,144)�3.33,p�.039),<br />

with smokers being more pessimistic about this than former smokers<br />

(p�.043). Conclusions: These findings have implications for planning the<br />

care <strong>of</strong> lung cancer patients. In addition to the different treatment needs <strong>of</strong><br />

smokers and former smokers, smokers may need greater support to cope<br />

with the emotional effects <strong>of</strong> their illness. Our findings suggest that<br />

smokers blame their illness on their own behaviour, and have a more<br />

pessimistic view <strong>of</strong> likely survival time, so they may need more encouragement<br />

to accept life-prolonging treatments. These results may also inform<br />

tailored smoking-cessation advice.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


410s Health Services Research<br />

6114 General Poster Session (Board #10G), Mon, 1:15 PM-5:15 PM<br />

Impact <strong>of</strong> oncology drug shortages on patient treatment. Presenting Author:<br />

Daniel Jacob Becker, St. Luke’s-Roosevelt and Beth Israel Medical Center,<br />

Continuum Cancer Centers <strong>of</strong> New York, New York, NY<br />

Background: Cancer drug shortages increased considerably over the past 5<br />

years, but quantitative analyses <strong>of</strong> the scope and effects are limited. We<br />

assessed the effects <strong>of</strong> drug shortages on outpatient medication use in a<br />

single New York City university hospital. Methods: We examined pharmacy<br />

records for drug shortages, defined by the ASHP as supply issues that affect<br />

“how the pharmacy prepares or dispenses a drug product or influences<br />

patient care when prescribers must use an alternative agent.” We examined<br />

outpatient records for all cancer patients treated with infusional antineoplastic<br />

and/or supportive medications from 4/2010-9/2010 and from 4/2011-<br />

9/2011, and performed subgroup analysis on Aug/Sept 2011, the peak <strong>of</strong><br />

medication availability problems at our hospital. We compared proportions<br />

<strong>of</strong> patients treated with specific medications in 2010 and 2011 with<br />

Pearson’s chi-square test. Results: Twelve medications were in shortage<br />

during the study period in 2010 and 22 in 2011. Between 4/1/10 and<br />

9/30/10, 335 cancer patients were treated, and 379 patients were treated<br />

between 4/1/11 and 9/30/11. Median patient age was 60 years. Cancer<br />

site <strong>of</strong> origin was comparable between years: breast (40%), GI (15%),<br />

lymphoma (10%), GU (8%), and lung (7%). Drugs considered in shortage<br />

were used for 170 (50.8%) patients in 2010 and 241 (63.6%) patients in<br />

2011 (p�0.0005). Of 235 patients treated in Aug/Sept 2011, there were<br />

23 (9.8%) documented therapy changes due to shortages, compared with<br />

0 changes among 211 patients treated in Aug/Sept 2010 (p�0.0001).<br />

Among patients treated in Aug/Sept 2010 24 (11.4%) patients received<br />

paclitaxel and 19 (9.0%) received docetaxel. Among patients treated in<br />

Aug/Sept 2011, 11 (4.7%) received paclitaxel and 38 received docetaxel<br />

(16.2%), a 69% decrease for paclitaxel and 80% increase for docetaxel<br />

from 1 year prior (p�0.009, and p�0.024, respectively). The estimated<br />

cost <strong>of</strong> a single treatment with paclitaxel for 1 patient with BSA 1.75 was<br />

$47.59 versus $858.39 for docetaxel, a 1704% increase. Conclusions:<br />

Oncology drug shortages affected the majority <strong>of</strong> patients in our center and<br />

increased at an alarming rate. Drug shortages have substantial economic<br />

costs and mandate treatment changes that may affect efficacy and toxicity.<br />

6116 General Poster Session (Board #11A), Mon, 1:15 PM-5:15 PM<br />

Pregnancy outcomes in women with cancer. Presenting Author: Andrea<br />

Milbourne, University <strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: Parenthood after cancer is a critical concern for many cancer<br />

patients (pts). Pregnancy (prg) during cancer is an emotional time for about<br />

1/1000 pregnant women. No randomized controlled studies exist examining<br />

the impact <strong>of</strong> cancer treatment (tx) on the developing fetus nor on the<br />

woman with cancer. Methods: From 2002-2011, women presenting for<br />

cancer tx during prg were approached for this IRB-approved prospective<br />

database study. All pts provided written consent. Results: To date 143 pts<br />

are evaluable. The median age at diagnosis was 32.1 years and median<br />

gestational age (GA) at enrollment was 18.2 weeks. 95/143 (66.4%) are<br />

White, 19 (13.3%) are African <strong>American</strong>, 17 (11.9%) are Hispanic and 12<br />

(8.4%) are Asian/Other. Primary cancers included breast (n�59, 41.3%),<br />

hematologic (n�29, 20.3%), melanoma (n�13, 9%), GYN (n�11, 8%),<br />

GI (n�8, 5.6%), head/neck (n�7, 6%) and other (n�16, 11%) (brain�4,<br />

GU�1, thyroid�3, head/neck�7, thoracic�1, sarcoma�6, unknown primary�1).<br />

111/143 (77.6%) <strong>of</strong> prgs resulted in live births. Median birth<br />

weight was 6.5 lbs. Median follow-up time for pts was 32.3 months. To<br />

date, 3/19 pts who terminated prgs have died (1.6%). Most terminations<br />

occurred in the 1st trimester. To date, 79 pts (55.2%) are NED and 23 pts<br />

have died; <strong>of</strong> these 19 (1.7%) had live births. No major malformations were<br />

observed in the 74/143 (52%) <strong>of</strong> pts who received chemotherapy (CTx)<br />

during pregnancy. 57% received FAC/FEC; other regimens included ABVD<br />

(n�5), cytarabine (n�5), CHOP/R-CHOP, and platinum-based regimens.<br />

Median GA at the start <strong>of</strong> CTx was 19.7 wks. Median number <strong>of</strong> CTx cycles<br />

during prg was 4. Other pts underwent surgery (n�32), no tx (n�14),<br />

deferred tx until after delivery (n�17), radiation (2), transplant (3), other<br />

(1). Conclusions: Cancer diagnosis during prg is compatible with successful<br />

tx and prg outcome. Cancer tx during the 2nd and 3rd trimester can be<br />

safely given and in our pts did not result in adverse prg outcomes. Tx during<br />

the 1st trimester is usually not recommended. Thus cancer pts in their 1st<br />

trimester need to be extensively counseled about their disease as well as<br />

about the risks to the prg. In our pts continuation or termination <strong>of</strong> prg were<br />

not associated with an increased risk <strong>of</strong> death.<br />

6115 General Poster Session (Board #10H), Mon, 1:15 PM-5:15 PM<br />

<strong>Part</strong>icipation <strong>of</strong> teenagers and young adults (TYA) in cancer clinical trials<br />

(CCT): What can we learn from six years <strong>of</strong> accrual data in England?<br />

Presenting Author: Lorna Anne Fern, University College Hospitals London,<br />

London, United Kingdom<br />

Background: We reported underrepresentation <strong>of</strong> TYA in CTT in England,<br />

2005-06. Since 2005 national healthcare policies and research initiatives<br />

aimed at increasing participation <strong>of</strong> TYA in CCT have been implemented.<br />

We aimed to determine if this has improved accrual rates (AR). Methods: We<br />

analyzed accrual by age during 2005-2010 to UK Cancer Research<br />

Network interventional trials recruiting newly diagnosed patients (pts) with<br />

leukaemia, lymphoma, bone/s<strong>of</strong>t tissue sarcoma, central nervous system<br />

and germ cell tumours. AR were expressed as the ratio <strong>of</strong> pts entered onto<br />

trial compared to population incidence cases for 2005-08; for 2009-10,<br />

mean incidence <strong>of</strong> 2005-08 was used. Results: 2005-10 showed an AR<br />

increase <strong>of</strong> 10.3% for pts 10-14 yrs, 17.9% for pts 15-19 yrs but only<br />

4.6% for pts 20-24 yrs (Table). In 2010 AR was 54.4% for pts 10-14 yrs,<br />

43.3% for 15-19 yr olds and 20.6% for pts 20-24. <strong>Annual</strong> increases <strong>of</strong> AR<br />

were observed for pts 15-19 yrs, but in no other age groups, 0-59 yrs. We<br />

looked at AR for children and younger teenagers , 5-14 yrs, vs older TYA,<br />

15-24 yrs. Overall, AR for 5-14 yr olds was greater, 53.7% vs 23.0% for pts<br />

15-24 yrs; however, during 2005-10 AR increased by 9.7% for pts aged<br />

15-24 compared to 7.8% for pts aged 5-14. Conclusions: AR for TYA has<br />

improved in between 2005-10. Most benefit is evident for older teenagers;<br />

AR for young adults remain disappointing. Changes relate to increased trial<br />

availability and access, centralisation <strong>of</strong> care for TYA, amendments to age<br />

eligibility criteria to reflect tumour biology and increased collaboration<br />

between adult and paediatric clinical research groups. Strategies to<br />

improve AR for young adults require further development.<br />

Number recruited (R), incidence (I), and AR for pts 0-24 yrs.<br />

0-4 5-9 10-14 15-19 20-24<br />

R I AR R I AR R I AR R I AR R I AR<br />

2005 203 350 58.0 129 249 51.8 107 243 44.0 99 388 25.5 84 524 16.0<br />

2006 207 352 58.8 132 245 53.9 120 276 43.5 110 361 30.5 67 519 12.9<br />

2007 225 356 63.2 115 212 54.2 111 237 46.8 110 358 30.7 58 565 10.3<br />

2008 243 317 76.7 149 204 73.0 131 242 54.1 121 341 35.5 76 558 13.6<br />

2009 200 345 58.0 129 230 56.1 115 252 45.6 144 364 39.6 104 543 19.2<br />

2010 232 345 67.2 132 230 57.4 137 252 54.4 158 364 43.4 112 543 20.6<br />

%D 9.2 5.6 10.3 17.9 4.6<br />

6117 General Poster Session (Board #11B), Mon, 1:15 PM-5:15 PM<br />

Financial burdens (FB), quality <strong>of</strong> life, and psychological distress among<br />

advanced cancer patients (ACP) in phase I trials and their spousal<br />

caregivers (SC). Presenting Author: Fay J. Hlubocky, University <strong>of</strong> Chicago<br />

Pritzker School <strong>of</strong> Medicine, Chicago, IL<br />

Background: FBs have been identified as a significant predictor <strong>of</strong> stress for<br />

cancer patients and their caregivers/partners. FBs may indirectly affect the<br />

quality <strong>of</strong> life and psychological distress <strong>of</strong> clinical trial subjects, particularly<br />

those with advanced disease on Phase I trials. Methods: A convenience<br />

sample <strong>of</strong> ACP enrolling in phase I trials was assessed at baseline (T1) and<br />

one month (T2) using several measures including: depression (CES-D),<br />

state-trait anxiety (STAI-S/T), quality <strong>of</strong> life/qol (FACIT-Pal), and global<br />

health (SF-36). Data on FB were obtained via the questions <strong>of</strong> Ell (2008),<br />

querying subjects on: employment status, unexpected medical costs,<br />

concerns regarding wages (e.g. termination, use <strong>of</strong> sick time), and financial<br />

stress. Results: 104 subjects (52 Phase I ACPs and 52 SC) were separately<br />

interviewed at T1 and T2. For the total population: median age 61<br />

(28-78y); 50% male; 100% married; 87% Ca; 67%� HS educ; 55% GI<br />

dx; 53% income �$65,000 yr, with45% ACPs employed full/part-time;<br />

55% retired; 66% SC employed full/part-time with 34% retired. At T1,<br />

82% <strong>of</strong> ACP reported medical cost concerns and 79% reported financial<br />

stress. For SC at T1, 69% reported medical costs concerns; 83% employed<br />

SC reported wage concerns; and 81% reported financial stress. For both<br />

ACP and SC, rates remained consistent with the exception <strong>of</strong> increased<br />

self-report re medical cost concerns at 85% and 72% respectively at T2. At<br />

T2, ACP who reported unexpected medical costs had higher STAI-S (31 �<br />

10v29�12, p�0.02) and CES-D scores (12 � 11v10� 9, p�0.04). SC<br />

with self-reported financial stress had higher STAI-S anxiety (39 � 17v35<br />

� 13, p�0.03) at T2. In regression analyses, ACP with medical costs<br />

concerns had poorer FACIT-Pal QOL over time. Also, SC with medical cost<br />

concerns at T2 was negatively associated with SF-36 scores. ACP and SC<br />

qualitative responses re FBs revealed salient themes: insurance coverage,<br />

unexpected costs associated with research study participation, Medicare,<br />

bankruptcy, and worry re financial stability after ACP death. Conclusions:<br />

FBs are negatively associated with quality <strong>of</strong> life among clinical trial<br />

subjects and SC in phase I trials.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


6118 General Poster Session (Board #11C), Mon, 1:15 PM-5:15 PM<br />

Influence <strong>of</strong> hospital characteristics on immediate breast reconstruction<br />

following mastectomy. Presenting Author: Catherine A. Richards, Mailman<br />

School <strong>of</strong> Public Health, Columbia University, New York, NY<br />

Background: Immediate breast reconstruction (IBR) following mastectomy<br />

is underutilized in the U.S. Racial, economic and geographic factors are<br />

associated with lower rates <strong>of</strong> IBR. Prior research has explored the<br />

association <strong>of</strong> individual and surgeon-level factors with the use <strong>of</strong> IBR, with<br />

little attention paid to hospital characteristics. Methods: We analyzed data<br />

from the 2008 Nationwide Inpatient Sample (NIS), a 20% random sample<br />

<strong>of</strong> academic, public and private U.S. hospitals. We used ICD-9 codes to<br />

identify women diagnosed with invasive breast cancer or DCIS who<br />

underwent mastectomy, and IBR (natural or expander/implant). If a<br />

hospital performed at least one IBR during 2008, they were classified as<br />

performing reconstruction. Relative risk regression was used to assess the<br />

hospital factors associated with a hospital performing IBR. Results: Of the<br />

3,518 hospitals that performed mastectomy in 2008 only 50.4% performed<br />

at least one IBR. For hospitals that did not perform IBR, the average<br />

number <strong>of</strong> mastectomies was 5, compared to 35 at hospitals that did<br />

perform IBR (p�0.01). Among hospitals that did perform IBR, the mean<br />

proportion <strong>of</strong> mastectomy patients that had IBR was 34% (SD�20). In a<br />

multivariable adjusted model, urban/teaching (RR�3.47) and urban/nonteaching<br />

(RR�2.86) hospitals were significantly more likely to perform IBR<br />

compared to rural hospitals. Hospitals with a high proportion <strong>of</strong> privately<br />

insured patients (RR�1.10) were significantly more likely to perform IBR<br />

compared to hospitals with a low proportion <strong>of</strong> privately insured patients. In<br />

contrast, hospitals with a high proportion <strong>of</strong> publically insured patients<br />

(RR�0.24) and hospitals with a high proportion <strong>of</strong> female patients � 70<br />

years old (RR�0.75) were significantly less likely to perform IBR. Hospital<br />

region, hospital ownership status and the proportion <strong>of</strong> nonwhite patients<br />

were not significantly associated with IBR. Conclusions: Almost half <strong>of</strong> all<br />

U.S. hospitals where mastectomies are performed do not have any patients<br />

who have undergone IBR. The likelihood a hospital will perform IBR varies<br />

significantly by hospital characteristics.<br />

6120 General Poster Session (Board #11E), Mon, 1:15 PM-5:15 PM<br />

Do patient tracking, follow-up, and referral practices contribute to breast<br />

cancer disparities in a large urban area? Presenting Author: Christine B.<br />

Weldon, Center for Business Models in Healthcare, Chicago, IL<br />

Background: Chicago Black women are 62% more likely to die from breast<br />

(BC) cancer than White women. Previous data from 39 Chicago hospitals<br />

suggested significant quality deficits in breast cancer screening and<br />

treatment (Chicago Breast Cancer Quality Consortium, 2010). Patient<br />

tracking, follow up and referral practices may influence quality <strong>of</strong> care for<br />

minority women (Mojica et al, Cancer Control, 2007). Our goal is to<br />

evaluate tracking, follow up and referral practices during screening,<br />

diagnosis and treatment <strong>of</strong> BC at Chicago hospitals servicing Black women.<br />

Methods: Using the framework approach <strong>of</strong> qualitative research, we<br />

conducted interviews with providers <strong>of</strong> BC screening and care from 20<br />

Chicago institutions with Black patients averaging 50% <strong>of</strong> patient base (15<br />

community, 3 academic and 2 public hospitals). Informants included<br />

surgeons, medical oncologists, radiologists, mammography technicians,<br />

internists, nurses, administrators, and patient navigators. Interviews were<br />

transcribed, and thematic and statistical analyses were performed (simple<br />

frequencies and Fisher’s exact test). Results: Six <strong>of</strong> the 20 sites (30%)<br />

follow up with patients who did not show for a scheduled mammography<br />

visit. Five <strong>of</strong> these sites (83%, 5/6) have a low “no-show” rate (below 20%),<br />

compared to 4 sites (29%, 4/14) with low “no-show” rates among the 14<br />

sites without follow-up (p�0.05). Seven <strong>of</strong> the 20 sites (25%) direct<br />

diagnosed patients to their next step in care by providing referrals and<br />

guidance, while other 13 sites rely on a primary care physician or leave the<br />

patient without a clear care plan. BC patients at 6 <strong>of</strong> the 7 sites directing<br />

care (83%, 5/6) are referred to a mid- or high-volume surgeon (3� BC<br />

surgeries / month), compared to patients from only 1 <strong>of</strong> the 13 sites not<br />

directing care (p�0.001). Nine <strong>of</strong> the 20 sites track diagnosed BC patients<br />

through their care. Five <strong>of</strong> them (56%, 5/9) also track survivors, compared<br />

to none (0%, 0/11) <strong>of</strong> the 11 sites who do not track patients (p�0.008).<br />

Conclusions: Poor tracking, follow up and referral practices for breast<br />

cancer screening and treatment are associated with suboptimal care and<br />

may contribute to outcome disparities for Black women in Chicago.<br />

Health Services Research<br />

411s<br />

6119 General Poster Session (Board #11D), Mon, 1:15 PM-5:15 PM<br />

Why do cancer patients die in the emergency department (ED)? Analysis <strong>of</strong><br />

283 deaths in North Carolina EDs in 2008. Presenting Author: Ashley<br />

Nicole Leak, UNC Gillings School <strong>of</strong> Global Public Health, Department <strong>of</strong><br />

Health Policy and Management, Chapel Hill, NC<br />

Background: Emergency departments (ED) in the US are utilized by cancer<br />

patients for symptom management, treatment side effects, oncologic<br />

emergencies, and/or end <strong>of</strong> life care. EDs have become a place where acute<br />

care needs are addressed and there are discussions about end <strong>of</strong> life care.<br />

The purpose <strong>of</strong> this study was to describe characteristics <strong>of</strong> cancer patients<br />

who died in the ED, highlighting lung cancer patients and their ED visit<br />

characteristics. Methods: A secondary data analysis <strong>of</strong> ED visit data fromthe<br />

North Carolina Disease Event Tracking and Epidemiologic Collection Tool<br />

(NC DETECT), a population database <strong>of</strong> 110 <strong>of</strong> the 112 EDs in NC in 2008.<br />

This was a descriptive, retrospective analysis providing descriptive statistics<br />

<strong>of</strong> patient demographics including: sex, age at death, insurance,<br />

cancer type, and visit categories (hour, day, month). Free text chief<br />

complaints, as recorded by the health care provider, were cleaned and<br />

categorized. Results: There were37,760 ED visits by 27,644 patients<br />

associated with cancer; 283 (1%) <strong>of</strong> these visits resulted in death in the<br />

ED. The most common chief complaints <strong>of</strong> those who died were respiratory<br />

distress (17.3%), neurological changes (13.4%), and pain (5.7%). Of all<br />

cancer deaths, 63% were male with a mean age <strong>of</strong> 66 (SD 14.2). Over a<br />

third (N� 104, 36.7%) <strong>of</strong> cancer ED visits resulting in death had a code for<br />

lung cancer listed. Medicare was the insurance provided for almost half<br />

(47.3%) <strong>of</strong> these patients. Majority <strong>of</strong> deaths occurred on a patient’s first<br />

ED visit (70.9%). Although a third (34.7%) <strong>of</strong> deaths occurred during<br />

weekends, over a third (37.5%) occurred during weekday clinic hours.<br />

Conclusions: Even though deaths in the ED were infrequent, this study<br />

provides insight into reasons patients visit the ED. Some patients were<br />

enrolled in Hospice and/or had a DNR documented. This study can inform<br />

future research associated with precipitating factors leading up to the ED<br />

visit (e.g. worsening shortness <strong>of</strong> breath, location <strong>of</strong> care prior to ED visit).<br />

This study illustrates the importance <strong>of</strong> research on discussion <strong>of</strong> end <strong>of</strong> life<br />

care needs (advanced directives, code status, use <strong>of</strong> Hospice services) with<br />

cancer patients and their family before they reach the final stage <strong>of</strong> their<br />

disease.<br />

6121 General Poster Session (Board #11F), Mon, 1:15 PM-5:15 PM<br />

Competing event risk stratification may improve the design and efficiency<br />

<strong>of</strong> clinical trials: Secondary analysis <strong>of</strong> SWOG 8794. Presenting Author:<br />

Brent Shane Rose, Harvard University, Boston, MA<br />

Background: Efficiency <strong>of</strong> clinical trials may be improved by stratifying<br />

according to competing event risk. We aimed to test whether effect and<br />

sample size estimates would be altered when adjusting for competing event<br />

risk, using data from the SWOG 8794 trial <strong>of</strong> adjuvant radiation therapy<br />

(RT) for high-risk post-operative prostate cancer. Methods: The primary<br />

outcome was metastasis-free survival (MFS), defined as time to first<br />

occurrence <strong>of</strong> metastasis or death from any cause (i.e., competing mortality<br />

(CM)). We developed separate risk scores for time to metastasis and CM<br />

using backward stepwise competing risks regression. Risk factors for<br />

metastasis were PSA, Gleason score, and seminal vesicle invasion, and for<br />

CM were age, performance status, and Charlson comorbidity index.<br />

Treatment effects were estimated using Cox models adjusted for risk<br />

scores. We identified an enriched subgroup <strong>of</strong> 75 patients at high risk <strong>of</strong><br />

metastasis and low risk <strong>of</strong> CM, based on risk score cut<strong>of</strong>fs. Sample size<br />

estimates assumed type I and II error <strong>of</strong> 0.10 and 0.20, and accrual and<br />

follow-up times <strong>of</strong> 6 and 6 years. Results: The mean CM risk score was<br />

significantly lower in the RT vs. control arm (p�0.001). The effect <strong>of</strong> RT on<br />

MFS (HR 0.70; 95% CI, 0.53-0.92; p�0.010) was attenuated when<br />

controlling for metastasis and CM risk (HR 0.76; 95% CI, 0.58-1.00;<br />

p�0.049). The hazard for CM was reduced by RT (HR 0.82; 95% CI,<br />

0.59-1.14; p�0.24), but this effect was attenuated when controlling for<br />

CM risk (HR 0.94; 95% CI, 0.67-1.31; p�0.71). In contrast, there was no<br />

difference in the adjusted and unadjusted HRs for metastasis (0.50; 95%<br />

CI, 0.31-0.81; p�0.005 and 0.49; 95% CI, 0.30-0.81; p�0.005).<br />

Compared to the whole cohort, the enriched subgroup had the same<br />

baseline 10-year incidence <strong>of</strong> MFS (40%; 95% CI 22-57%), but a higher<br />

incidence <strong>of</strong> metastasis (30% (95% CI, 15-47%) vs. 20% (95% CI,<br />

15-26%)). A randomized trial in an enriched population could have<br />

achieved 80% power with 44% less patients (313 vs. 709 patients,<br />

respectively), due to the differing event composition. Conclusions: Stratification<br />

based on competing event risk may improve the design and<br />

efficiency <strong>of</strong> clinical trials. These findings should be externally validated.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


412s Health Services Research<br />

6122 General Poster Session (Board #11G), Mon, 1:15 PM-5:15 PM<br />

Variation in health-related quality <strong>of</strong> life (HRQOL) by ECOG performance<br />

status (PS) and fatigue among patients with chronic lymphocytic leukemia<br />

(CLL). Presenting Author: Christopher Flowers, Emory University, Atlanta,<br />

GA<br />

Background: Clinicians and investigators commonly use ECOG PS and<br />

clinician-reported patient (pt) fatigue as surrogates for HRQOL, a multifaceted<br />

construct that comprehensively looks at the pt perspective on<br />

disease and well-being. Because limited data exist on the relationships<br />

between PS, fatigue, and HRQOL for CLL pts, we examined the associations<br />

between these measures, and 3 validated HRQOL instruments: the<br />

Functional Assessment <strong>of</strong> Cancer Therapy-Leukemia (FACT-Leu), EQ-5D,<br />

and Brief Fatigue Inventory (BFI). Methods: Data were collected in<br />

CONNECT CLL, a prospective US observational registry initiated in 2010.<br />

Patient demographics and clinical characteristics were provided by clinicians.<br />

Patient HRQOL was self-reported at enrollment using the FACT-Leu,<br />

EQ-5D, and BFI. Scores were analyzed by ECOG PS (0, 1, 2-4) and<br />

clinician-reported fatigue (yes, no). Differences in HRQOL scores were<br />

assessed by ANOVA. Results: HRQOL data were reported by 899 pts from<br />

148 community, 10 academic, and 3 government centers. ECOG PS was<br />

available on 711 pts. Overall HRQOL, measured by mean FACT-Leu,<br />

FACT-G and EQ-5D Visual Analogue Scale (VAS), worsened with ECOG PS<br />

severity and was worse in pts with fatigue (all p�0.0001). All FACT-Leu<br />

domains except social/family were worse in pts with fatigue and those with<br />

higher ECOG PS. Mean EQ-5D pain/discomfort, mobility, self care and<br />

usual activities domain scores worsened in severity as ECOG worsened and<br />

for pts with fatigue (all p�0.011). BFI data indicated that global fatigue,<br />

fatigue severity and fatigue-related interference worsened by ECOG severity<br />

and were associated with clinician-reported fatigue (all p�0.0001).<br />

Conclusions: Initial CONNECT CLL results confirm that HRQOL worsens<br />

with worsening ECOG PS and was worse among pts with fatigue, especially<br />

in physical/functioning domains, pain/discomfort, and mobility. These<br />

results indicate that baseline ECOG PS and physician-rated fatigue are<br />

rapid assessments that predict robust measures <strong>of</strong> HRQOL. Future analyses<br />

are planned to examine how HRQOL, ECOG PS and fatigue change over<br />

time with changes in treatment and CLL disease status.<br />

6124 General Poster Session (Board #12B), Mon, 1:15 PM-5:15 PM<br />

Prevalence and predictors <strong>of</strong> adherence to antiemesis prophylaxis in lung<br />

cancer: A population-based study. Presenting Author: Daniel Richard<br />

Gomez, University <strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: Nausea/vomiting is a significant toxicity in the treatment <strong>of</strong><br />

lung cancer, but barriers exist to the delivery <strong>of</strong> prophylaxis. We studied<br />

compliance and predictors <strong>of</strong> prophylactic antiemetics with chemotherapy<br />

in this setting. Methods: We used a Texas state registry <strong>of</strong> clinical data<br />

linked with Medicare claims from 2001-2007. Our study population was<br />

incident lung cancers treated with platinum agents within 12 months <strong>of</strong><br />

diagnosis. To define guideline-adherent care, we assessed compliance to<br />

the <strong>American</strong> <strong>Society</strong> <strong>of</strong> <strong>Clinical</strong> Oncology recommended prophylactic<br />

agents dexamethasone and a 5-HT3 antagonist. Adherence was scored as a<br />

binary variable and defined as administration within 24 hours <strong>of</strong> the first<br />

day <strong>of</strong> the first cycle <strong>of</strong> chemotherapy. We utilized a logistic regression<br />

model to evaluate the role <strong>of</strong> the following factors in predicting adherence:<br />

concurrent radiation therapy (RT), race (black vs. white), histology (small<br />

cell vs. non-small cell), rural location, Charlson Comorbidity Index (CCI),<br />

household income (by quartiles [Q]), education, and treatment year<br />

(binary). Results: Of 31,762 patients in the database, 5155 patients met<br />

the above criteria. The adherence rate to dexamethasone and 5-HT3<br />

antagonists increased over time, from 51.5% in 2001 to 71.6% in 2007.<br />

Patients treated in the years 2005-2007 were 1.739 times more likely to<br />

be adherent to prophylaxis than were those from 2001-2004. Variables<br />

that predicted adherence (adjusted odds ratio [OR], 95% confidence<br />

interval [CI]) were: age (OR�1.013, CI [1.001, 1.026]), treatment year<br />

(OR�1.756, CI [1.548, 1.991]), race (black vs. white OR�0.684, CI<br />

[0.548,0.853]), median income (higher vs. lower OR�1.83 [Q2 vs. Q1];<br />

OR�1.296 [Q3 vs. Q1]; OR�1.496 [Q4 vs. Q1]), CCI (1� vs. 0,<br />

OR�0.613, CI [0.530,0.709]), and concurrent RT [yes vs. no OR�1.358,<br />

CI [1.197,1.541]). Conclusions: Compliance with guidelines for prophylactic<br />

antiemetics is suboptimal, but increasing over time. Several characteristics<br />

predict for improved adherence, including white race, median income,<br />

advanced age, and the receipt <strong>of</strong> concurrent RT. These findings highlight<br />

substantial economic disparities in supportive care <strong>of</strong> lung cancer in the<br />

state <strong>of</strong> Texas.<br />

6123 General Poster Session (Board #12A), Mon, 1:15 PM-5:15 PM<br />

Loss to follow-up <strong>of</strong> cancer patients in the poorest district <strong>of</strong> the United<br />

States. Presenting Author: Antranik Mangardich, Lincoln Medical and<br />

Mental Health Center, Bronx, NY<br />

Background: Loss to follow-up (LFU) <strong>of</strong> cancer patients is a serious<br />

dilemma, and has only been narrowly studied. Lincoln Medical and Mental<br />

Health Center (LMMHC) serves South Bronx (SB), the poorest district in the<br />

nation. The purpose <strong>of</strong> this study was to assess rates <strong>of</strong> LFU and correlate it<br />

with age, sex, ethnicity, race, cancer types, and stage at diagnosis.<br />

Methods: We collected data from 1,552 patients diagnosed with invasive<br />

cancer in LMMHC between 2006-2010. The data collected were age, sex,<br />

ethnicity, race, type <strong>of</strong> cancer, stage, LFU, treatment, and vital status.<br />

Results: From the 1,552 patients, roughly 25 % were LFU, with 50%<br />

receiving some initial form <strong>of</strong> treatment. The remaining percentages are<br />

shown below (Table). A higher rate <strong>of</strong> LFU was with patients younger than<br />

65 (OR: 1.38, 95% CI: 1.08-1.76). There was no correlation between sex<br />

and LFU. Non-Hispanics were more likely to be LFU compared to Hispanics<br />

(OR: 1.39, 95% CI: 1.07 – 1.8). Blacks were more likely to be LFU<br />

compared to non-Blacks (OR: 1.43, 95% CI: 1.12–1.82). There was no<br />

significance between LFU and stage at diagnosis. Looking at cancer<br />

specific data, colon cancer (C) and head and neck cancers (HN) had the<br />

highest percentage <strong>of</strong> LFU (30% each). There was higher LFU rate for C<br />

compared to breast cancer (B) (OR: 1.7, 95% CI: 1.03-2.8), prostate<br />

cancer (P) (OR: 1.88, 95% CI: 1.18-3.02), and lung cancer (L) (OR: 1.64<br />

95% CI: 0.94- 2.8). HN patients were more likely LFU compared to B (OR:<br />

2.4, 95% CI: 1.13-5.2), P (OR: 2.69, 95 % CI: 1.28-5.68), and L (OR:<br />

2.3, 95% CI: 1.05-5.19) patients. There was no significant difference<br />

between C and HN patients in respect to LFU. Conclusions: In the SB, LFU<br />

rates are related to age, ethnicity, race, and type <strong>of</strong> cancer. Younger<br />

patients, blacks, non-Hispanics, and those with C and HN cancers were<br />

most likely to be LFU, the latter likely due to the lack <strong>of</strong> a HN surgeon at<br />

LMMHC. We hope that with focus on race, ethnicity, and cancer-specific<br />

disparities in LFU rates, we will improve the retention rate <strong>of</strong> our cancer<br />

patients in the future.<br />

Continued to<br />

Sent to nursing<br />

Transferred<br />

LFU<br />

follow up<br />

home/hospice<br />

to other facility<br />

371 (24%) 863 (55%) 167 (11%) 151 (10%)<br />

No Rx Rx No Rx Rx No Rx Rx No Rx Rx<br />

190 (51%)<br />

Rx�treatment<br />

180 (49%) 190 (22%) 672 (78%) 105 (63%) 67 (37%) 93 (62%) 58 (38%)<br />

6125 General Poster Session (Board #12C), Mon, 1:15 PM-5:15 PM<br />

In-hospital mortality <strong>of</strong> patients admitted through the emergency department<br />

<strong>of</strong> a comprehensive cancer center. Presenting Author: Ahmed F.<br />

Elsayem, University <strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: Cancer is a common presenting condition for emergency<br />

departments (EDs); however, there is limited information on outcomes <strong>of</strong><br />

ED cancer patients subsequently admitted to the hospital. The purpose <strong>of</strong><br />

this study is to describe outcomes <strong>of</strong> patients with hematologic malignancies<br />

versus those with solid tumors admitted through the ED <strong>of</strong> a<br />

comprehensive cancer center. Methods: We queried the ED database <strong>of</strong> The<br />

University <strong>of</strong> Texas MD Anderson Cancer Center for calendar year 2010 and<br />

linked it to our institutional data warehouse, including tumor registry data.<br />

We classified all leukemia and related disorders, lymphoma, multiple<br />

myeloma, and bone marrow transplant patients as hematologic malignancies,<br />

and remaining cancers as solid tumors. Descriptive statistics, including<br />

chi-square, and t-tests were used in two-sided comparisons. All<br />

statistical analyses were performed using SPSS version 15. Results:<br />

20,732 total ED visits were made by 9,320 unique cancer patients. Of<br />

these, 5,364 (58%) were admitted to the hospital at least once (range 1-13<br />

admits). ED admissions constituted 39% <strong>of</strong> total unique patients admitted<br />

(N�13,753). The main admission indications for solid tumor patients were<br />

infectious complications (particularly pneumonia), intractable pain, or<br />

dehydration. For hematologic malignancies, the main indication was<br />

neutropenic fever. 211/1656 (13%) <strong>of</strong> liquid tumor patients were admitted<br />

to the Intensive Care Unit (ICU) compared to 484/3708 (13%) <strong>of</strong> solid<br />

tumor patients (P�NS). Of all patients admitted through the ED, 587/<br />

5364 (10.9%) died during hospitalization. The hematologic hospital<br />

mortality rate was 225/1653 (13.6%) versus 362/3708 (9.8%) for solid<br />

tumors (P�0.001). Only 242/8389 (3%) <strong>of</strong> patients admitted directly<br />

from outpatient clinics died during the hospitalization (p�0.001).<br />

Conclusions: Patients admitted through the ED, particularly those with<br />

hematologic malignancies, have a high hospital mortality rate. ED-based<br />

palliative care interventions may be justified to improve quality <strong>of</strong> life and<br />

prevent unnecessary costly interventions and ICU admission. Further<br />

research should define predictors <strong>of</strong> poor outcomes in cancer patients<br />

admitted through the ED.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


6126 General Poster Session (Board #12D), Mon, 1:15 PM-5:15 PM<br />

Caring for Alaska native prostate cancer survivors in primary care: A survey<br />

<strong>of</strong> Alaska Tribal Health System providers. Presenting Author: Jon C. Tilburt,<br />

Mayo Clinic, Rochester, MN<br />

Background: Little is known about the actual constraints <strong>of</strong> doing so in the<br />

distinctive geographic context <strong>of</strong> Alaska. We performed a survey <strong>of</strong> primary<br />

care providers (PCPs) within the Alaska Tribal Health System (ATHS) to<br />

learn more about their attitudes and practices surrounding prostate cancer<br />

(PC) survivorship care. Methods: We surveyed primary care physicians,<br />

nurse practitioners, and physician assistants practicing in the ATHS. We<br />

administered surveys to assess attitudes about PSA monitoring, responsibility<br />

for long-term surveillance, supporting emotional health and medical<br />

needs, their degree <strong>of</strong> confidence in managing patients on androgen<br />

deprivation therapy (ADT), as well as their comfort level in managing<br />

common side effects <strong>of</strong> treatment. Results: Of the 221 providers surveyed,<br />

114 responded (52%). Most PCPs indicated a preference for annual PSA<br />

monitoring (69%), but several (27%) indicated monitoring every 6 months,<br />

and a few (4%) indicated monitoring every 24 months. Most (60%) thought<br />

PCPs should manage cancer surveillance, but many (40%) thought a<br />

specialist (urologist or medical oncologist) should perform that function.<br />

When asked about supporting patients’ emotional needs, PCPs indicated<br />

that support groups (63%) followed by survivorship clinics (16%) or on-site<br />

specialist visits (14%) were the most appropriate venues to address these<br />

concerns. Most respondents thought that medical needs <strong>of</strong> PC survivors<br />

could be addressed locally with appropriate specialty input (71%) or<br />

potentially through dedicated survivorship clinics (14%). Only 46% indicated<br />

being �moderately� or �very� confident managing ADT. Most PCPs<br />

indicated being moderately or very comfortable monitoring for recurrence<br />

(59%), managing erectile dysfunction (66%), addressing urinary incontinence<br />

(63%), and addressing emotional needs (61%). Conclusions: PCPs<br />

practicing in the ATHS express comfort with monitoring for PC recurrence,<br />

but express some concerns about the full range <strong>of</strong> issues in PC survivorship.<br />

Constructing cancer survivor care models in geographically dispersed and<br />

resource-limited contexts for Alaska Natives is an ongoing clinical and<br />

policy challenge.<br />

6128 General Poster Session (Board #12F), Mon, 1:15 PM-5:15 PM<br />

Financial relationships with commercial interests (COI) among abstracts at<br />

the ASCO <strong>Annual</strong> <strong>Meeting</strong>. Presenting Author: Angela R. Bradbury, <strong>Clinical</strong><br />

Genetics, Fox Chase Cancer Center, Philadelphia, PA<br />

Background: ASCO has a formal policy that requires disclosure <strong>of</strong> COI with<br />

entities having a commercial interest in research. There are limited data<br />

describing the frequency and type <strong>of</strong> COI reported in cancer research and<br />

how these have changed over time. Methods: We reviewed author-reported<br />

COI for abstracts submitted for the ASCO <strong>Annual</strong> <strong>Meeting</strong> in 2006 and<br />

2008–2011. COI are classified as employment, stock, consultant, honoraria,<br />

research, expert witness, and other. Logistic regression models were<br />

used to evaluate the association between COI and acceptance at the annual<br />

meeting and change over time. Results: 37% <strong>of</strong> all abstracts reported at<br />

least one author with a COI. Any COI increased significantly from 33% in<br />

2006 to 39% in 2011 (p�0.001). Any COI and all categories <strong>of</strong> COI were<br />

significantly more prevalent among abstracts accepted for presentation<br />

compared to those accepted for publication only (Table). Among abstracts<br />

accepted for presentation, any COI increased from 39% in 2006 to 48% in<br />

2011 (p�0.001) Increases by year were statistically significant for overall<br />

(p�0.001), honoraria (p�0.045), and research (p�0.002) COI.<br />

Conclusions: Many authors submitting abstracts accepted for presentation<br />

at the ASCO <strong>Annual</strong> <strong>Meeting</strong> disclose COI. Reported COI have increased<br />

over time and are significantly more frequent among abstracts accepted for<br />

presentation than for publication only. These data suggest either that<br />

cancer research increasingly involves relationships with industry or that<br />

COI reporting has become more common. Policies to manage and foster<br />

these relationships will continue to be required and revised.<br />

COI among abstracts submitted for the ASCO <strong>Annual</strong> <strong>Meeting</strong> (n�21,108).<br />

COI<br />

Accepted for<br />

presentation<br />

(n�12,448)<br />

Publication<br />

only<br />

(n�8,660)<br />

OR (CI)<br />

in 2006<br />

Multiplicative<br />

increase in OR (CI)<br />

for each year<br />

post-2006<br />

Any COI 45% 25% 2.0 (1.8-2.3) * 1.06 (1.03-1.10)*<br />

Employment 24% 13% 2.1 (1.8-2.5)* 1.01 (0.97-1.06)<br />

Stock 19% 9% 2.2 (1.8-2.6)* 1.03 (0.98-1.08)<br />

Consultant 27% 12% 2.5 (2.1-2.9)* 1.09 (0.99-1.09)<br />

Honoraria 23% 10% 2.4 (2.0-2.8)* 1.05 (1.001-1.10)***<br />

Research 29% 13% 2.2 (1.9-2.5)* 1.07 (1.03-1.12)**<br />

Expert witness 1.7% 0.5% 3.1 (1.6-5.8)* 1.0 (0.9-1.3)<br />

*p�0.001, **p� 0.02, ***p�0.002.<br />

Health Services Research<br />

413s<br />

6127 General Poster Session (Board #12E), Mon, 1:15 PM-5:15 PM<br />

Geographic and socioeconomic determinants <strong>of</strong> participation by elderly<br />

patients in surgical oncology trials. Presenting Author: John H. Stewart,<br />

Wake Forest School <strong>of</strong> Medicine, Winston-Salem, NC<br />

Background: A recent report by our group demonstrated that �0.5% <strong>of</strong><br />

elderly patients with breast, prostate and lung cancer participate in surgical<br />

oncology trials. However, little work to date has evaluated the roles <strong>of</strong><br />

regional healthcare infrastructure and socioeconomic factors on clinical<br />

trial participation in this cohort. Methods: The NCI Cooperative Group<br />

Surgical Oncology Trial (CGSOT) database was queried for patients treated<br />

for breast, prostate and lung cancer cancer between 2000 and 2008<br />

(n�13,541). Geographical Information Systems data were used to evaluate<br />

proximity to healthcare facilities while regional socioeconomic characteristics<br />

were obtained from the 2000 US Census and Area Resource file.<br />

Counts were used to create a proportion <strong>of</strong> respondents with the corresponding<br />

95% confidence interval calculated using the central limit theorem.<br />

Independent t-tests were used to assess differences in outcome measures<br />

between the two age groups. Results: We found that 4136 participants in<br />

the NCI CGSOT database were 65 years <strong>of</strong> age or older. Interestingly,<br />

92.7% <strong>of</strong> this cohort was white and 85.7% was female. Socioeconomic<br />

determinants <strong>of</strong> participation are shown in the table. Conclusions: The work<br />

presented herein suggests that elderly minority patients are less likely to<br />

participate in surgical oncology trials than their white counterparts.<br />

Furthermore, elderly participants tend to reside in less affluent areas.<br />

Future work will utilize interventions that improve the recruitment <strong>of</strong> elderly<br />

patients to these trials.<br />

Geographic<br />


414s Health Services Research<br />

6130 General Poster Session (Board #12H), Mon, 1:15 PM-5:15 PM<br />

Multidisciplinary assessment and reporting <strong>of</strong> fitness to drive in brain tumor<br />

patients: A gray matter. Presenting Author: Alexander V. Louie, London<br />

Regional Cancer Program, London, ON, Canada<br />

Background: In some jurisdictions, there is a legal requirement for physicians<br />

to report medically unfit drivers. Objectives <strong>of</strong> this study are to<br />

determine physician knowledge and attitudes on reporting legislation and<br />

driving assessment, and review our institution’s experience in evaluating<br />

fitness to drive in brain tumour patients. Methods: Physicians caring for<br />

brain tumour patients in South-western Ontario were identified by public<br />

databases and surveyed by mail. Survey questions elicited demographics,<br />

opinions, and factors influencing the decision to report. Patients receiving<br />

brain radiotherapy at our institution between January and June 2009 were<br />

identified and details <strong>of</strong> driving assessment were extracted. Fisher’s exact<br />

test and a logistic regression model were used to determine differences in<br />

responses between specialists and family physicians and factors influencing<br />

reporting. Results: Surveys (n�467) were distributed with 198 (43%)<br />

responses. Most (76%) felt that reporting guidelines were unclear. Neurologists<br />

(43%) and Family Physicians (22%) were felt to be the most<br />

responsible to report unfit drivers. Compared to specialists, Family Physicians<br />

were less likely: to be comfortable with reporting (p�0.02), to<br />

consider reporting (p�0.001), or discuss the implications <strong>of</strong> driving<br />

(p�0.001). Perceived barriers in assessing fitness to drive included: lack<br />

<strong>of</strong> tools to assess (57%) and the impact on the patient-physician relationship<br />

(34%). 158 patients were retrospectively reviewed. Forty-eight patients<br />

(30%) were reported to the provincial licensing authority and 64<br />

(41%) were advised not to drive. 53 patients experienced seizures, <strong>of</strong><br />

which 36 (68%) had a documented discussion on driving. Only 30 (56%)<br />

<strong>of</strong> these patients were reported to the licensing authority despite legal<br />

requirements. Age, primary disease, previous neurosurgery and seizures<br />

were predictive <strong>of</strong> reporting (p�0.05). On logistic regression modeling,<br />

seizures (OR 12.4) and primary CNS disease (OR 15.5) remained predictive<br />

<strong>of</strong> reporting. Conclusions: Despite guidelines and laws, the assessment<br />

<strong>of</strong> fitness to drive in patients with brain tumours is not routinely conducted<br />

or documented in a multidisciplinary setting.<br />

6132 General Poster Session (Board #13B), Mon, 1:15 PM-5:15 PM<br />

Predictors <strong>of</strong> retention in a cervical cancer survivorship study. Presenting<br />

Author: Lari B. Wenzel, University <strong>of</strong> California, Irvine, Irvine, CA<br />

Background: Women with cervical cancer experience numerous quality <strong>of</strong><br />

life (QOL) disruptions. A completed pilot study indicated that QOL and key<br />

biomarkers could be improved with psychosocial telephone counseling<br />

(PTC). A confirmatory trial has recently completed accrual.The purpose <strong>of</strong><br />

this study is to identify predictors <strong>of</strong> retention in this large biobehavioral<br />

trial, and examine retention differences between the pilot and confirmatory<br />

trial. Methods: Women with stage I – III cervical cancer diagnosed 9–30<br />

months earlier were randomized to either PTC or usual care. PROs and<br />

biological specimens were collected prior to randomization (N�204), and<br />

3 (T2) and 9 months post baseline. Sociodemographic, clinical and QOL<br />

variables were evaluated from baseline to T2 to identify predictors <strong>of</strong> study<br />

retention. Multiple measures, including the PROMIS Depression and<br />

Anxiety scales were entered into a multivariate stepwise logistic regression<br />

model to predict study retention. Results: In the multivariate model, being<br />

randomized to counseling (OR�4.4, 95% CI: 1.5-12.6), having clinically<br />

significant depression, defined as �1 SD above the 50th percentile<br />

(OR�4.2, 95% CI:1.7-10.5) and being single (OR�3.4, 95% CI: 1.3-9.2)<br />

were the most significant predictors <strong>of</strong> drop-out in the study overall. Among<br />

those in the counseling arm, depression is the strongest predictor <strong>of</strong><br />

dropout, with a five times higher dropout rate (OR�5.2, 95% CI:1.9-15.6)<br />

than those with low to moderate levels <strong>of</strong> depression, or no depression.<br />

However, retention rates improved substantially overall from the pilot study<br />

(72%) to the confirmatory study (84%), in both study arms, and retention<br />

increased among Latinas from 60% in the pilot study to 83% in the current<br />

study, and from 52% to 81% among those with less than a high school<br />

education. Conclusions: Results from this analysis <strong>of</strong> predictors <strong>of</strong> retention<br />

indicate that enhanced attention to sociocultural disparities can improve<br />

study retention. Attending to baseline clinical depression may be a further<br />

consideration.<br />

6131 General Poster Session (Board #13A), Mon, 1:15 PM-5:15 PM<br />

Sociodemographic and patient disparities in use <strong>of</strong> first-line chemotherapy<br />

(CT) in older adult patients (pts) with advanced non-small cell lung cancer<br />

(AdvNSCLC): Analysis <strong>of</strong> Surveillance, Epidemiology, and End Results<br />

(SEER)-Medicare data. Presenting Author: Josephine Feliciano, University<br />

<strong>of</strong> Maryland Greenebaum Cancer Center, Baltimore, MD<br />

Background: <strong>Clinical</strong> trials have demonstrated that CT improves survival<br />

and quality <strong>of</strong> life in pts with AdvNSCLC. Our prior studies suggest that only<br />

26% <strong>of</strong> older pts diagnosed with AdvNSCLC received CT through 2002. In<br />

this study, we examined more recent patterns, and the effect <strong>of</strong> sociodemographics,<br />

race, and age on receipt <strong>of</strong> CT in older pts with AdvNSCLC.<br />

Methods: Medicare pts diagnosed with AdvNSCLC from 2001-2005 were<br />

identified in SEER registries. Medicare enrollment data provided demographic<br />

information (age, race, sex, marital status), augmented with census<br />

tract level data on median household income. Comorbid conditions at<br />

baseline were identified by ICD9-CM diagnoses on claims. Receipt <strong>of</strong> 1st<br />

line CT was identified based on agent-specific procedure codes in claims.<br />

Logistic regression examined predictors <strong>of</strong> 1st line CT. Results: Of 31,341<br />

pts with Adv NSCLC, 34% received 1st line CT. As reflected in the table,<br />

poor predicted PS (PredPS), multiple medical comorbidities, and black<br />

race were associated with lower likelihood <strong>of</strong> CT receipt. Being female,<br />

married, having higher income, and being diagnosed at a later year were<br />

associated with a higher likelihood <strong>of</strong> receiving CT. Conclusions: Even when<br />

controlling for factors such as PS, our results suggest the presence <strong>of</strong> age,<br />

race, and socio-economic disparities in the receipt <strong>of</strong> 1st line CT for older<br />

patients with AdvNSCLC.<br />

Multivariate association <strong>of</strong> pt characteristics with receipt <strong>of</strong> 1st line CT.<br />

Covariate Odds ratio 95% CI<br />

Dx age (yrs)<br />

--<br />

--<br />

70-74<br />

0.81<br />

0.76, 0.88<br />

75-79<br />

0.57<br />

0.53, 0.61<br />

80-84<br />

0.33<br />

0.30, 0.35<br />

>85<br />

0.13<br />

0.11, 0.15<br />

Race<br />

--<br />

--<br />

Non-Hispanic Black<br />

0.83<br />

0.76, 0.92<br />

Prior year Medicaid or MSP 0.86 0.79, 0.94<br />

Female 1.09 1.03, 1.15<br />

Married 1.54 1.46, 1.63<br />

Dx Yr<br />

--<br />

--<br />

2002<br />

1.12<br />

1.04, 1.22<br />

2003<br />

1.25<br />

1.15, 1.35<br />

2004<br />

1.27<br />

1.17, 1.38<br />

2005<br />

1.32<br />

1.22, 1.43<br />

6133 General Poster Session (Board #13C), Mon, 1:15 PM-5:15 PM<br />

Patient satisfaction with participation in phase II/III NCCTG clinical trials:<br />

Was it worth it? (N0392). Presenting Author: Cynthia Chauhan, North<br />

Central Cancer Treatment Group, Wichita, KS<br />

Background: Patient (pt) trial experience may give insight to quality <strong>of</strong> life<br />

(QOL) factors affecting accrual, retention and outcome. While pts are vital<br />

to clinical trial success, we know little about their post-trial opinions. This<br />

trial examined pt opinion <strong>of</strong> their trial and treatment decision making<br />

(TDM). Methods: Pts enrolled on designated North Central Cancer Treatment<br />

Group (NCCTG) phase II or III treatment trials completed the TDM<br />

Control Preferences Scale at baseline and the Was It Worth It (WIWI)<br />

satisfaction assessment at the ends <strong>of</strong> cycle 1 and active protocol<br />

treatment. The primary endpoint was the proportion <strong>of</strong> pts reporting<br />

worthwhile participation. Fisher Exact tests compared pt opinion across<br />

subgroups. Results: As <strong>of</strong> 01/15/2012, 264 pts were enrolled on 25<br />

protocols and treated at 79 sites. 86% <strong>of</strong> pts were in phase II studies and<br />

89% had stage IV disease. At the end <strong>of</strong> cycle 1, pts felt the trial was<br />

worthwhile (74%), would do it again (85%), and would recommend it to<br />

others (82%). 85% <strong>of</strong> pts reported undiminished QOL and only 7% rated<br />

the trial worse than expected. End <strong>of</strong> treatment responses were similar.<br />

Satisfaction rates varied by tumor site (p�0.04). 11% <strong>of</strong> pts having tumor<br />

response rated the trial as not worthwhile, and 66% <strong>of</strong> pts with progressive<br />

disease rated it worthwhile (see table). Pts with concordant preferred and<br />

actual TDM roles rated participation higher than pts with discordant TDM<br />

roles (see table). Conclusions: Most pts endorsed their clinical trial<br />

experience. Contrary to popular beliefs, treatment outcome did not have an<br />

overwhelming impact on pt satisfaction, and was just one <strong>of</strong> many factors<br />

such as TDM role discordance. Assessing pt satisfaction will inform future<br />

study design that can potentially improve pt accrual and retention.<br />

Answered yes to. ..<br />

Response by end <strong>of</strong> Tx WIWI scores CPS by 1-month WIWI scores<br />

CR/PR PD SD P value<br />

Concordant<br />

(N�230)<br />

Discordant<br />

(N�28) P value<br />

Worth it? 89% 66% 82% 0.01 75% 64% 0.13<br />

Do again? 92% 93% 81% 0.07 87% 68% 0.01<br />

QOL maintained? 83% 71% 84% 0.01 85% 75% 0.38<br />

Study expectations<br />

met or better?<br />

92% 81% 85% �0.01 94% 86% 0.11<br />

Recommend? 91% 90% 80% 0.15 83% 68% 0.04<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


6134 General Poster Session (Board #13D), Mon, 1:15 PM-5:15 PM<br />

Relationship <strong>of</strong> MRI tests and referral <strong>of</strong> malignant adnexal masses to<br />

gynecologic oncologists for surgery. Presenting Author: Rachel Kupets,<br />

Sunnybrook Health Sciences Centre, University <strong>of</strong> Toronto, Toronto, ON,<br />

Canada<br />

Background: To evaluate the patterns <strong>of</strong> radiologic imaging by family<br />

physicians and gynecologists in the work up <strong>of</strong> women found to have an<br />

adnexal mass on pelvic ultrasound. To evaluate whether advanced imaging<br />

tests are associated with improved referral <strong>of</strong> high risk adnexal masses to<br />

gynecologic oncologists. Methods: Centralized provincial databases <strong>of</strong><br />

healthcare utilization were used to identify women aged 45 and older who<br />

received a pelvic ultrasound between 2006-2008. Subsequent imaging<br />

tests ordered by physician specialty were identified. Of those women who<br />

proceeded to laparotomy, logistic regression was performed to determine<br />

which imaging tests were associated with referral <strong>of</strong> high risk adnexal<br />

tumors to a gynecologic oncologist. Results: 193, 261 women had a pelvic<br />

ultrasound; 19, 949 (10.3%) had a laparotomy. 2223 and 627 women<br />

were categorized with benign and malignant adnexal masses respectively.<br />

Up to 12% <strong>of</strong> women had a pelvic MRI and 58% <strong>of</strong> women had a CT scan<br />

after a pelvic ultrasound.Family physicians referred 58% and gynecologists<br />

referred 47% <strong>of</strong> high risk ovarian masses to a gynecologic oncologist<br />

respectively after imaging.Gynecologic Oncologists operated on only 55%<br />

<strong>of</strong> women with malignant adnexal masses. On multivariate analysis factors<br />

significant for surgery by a gynecologic oncologist include a preoperative CT<br />

Scan OR 3.58 (p�.001) and CT Scan and MRI OR 7.78 (p�.001).<br />

Preoperative MRI alone had an OR <strong>of</strong> 1.86 (p�0.09) and was not<br />

significant. Mean time to surgery significantly increased when further<br />

imaging tests were performed after a pelvic ultrasound (100 days), CT (131<br />

days), MRI (170 days), CT and MRI (179 days),P 0.002. Conclusions: The<br />

addition <strong>of</strong> a pelvic MRI to a pelvic ultrasound does not improve the referral<br />

<strong>of</strong> high risk adnexal masses to a gynecologic oncologist. A consensus on<br />

appropriate imaging and triage is needed when an adnexal mass is<br />

identified on ultrasound.<br />

6136 General Poster Session (Board #13F), Mon, 1:15 PM-5:15 PM<br />

Relationship between whole-body tumor burden and quality <strong>of</strong> life in<br />

patients with neur<strong>of</strong>ibromatosis. Presenting Author: Scott Randall Plotkin,<br />

Massachusetts General Hospital Cancer Center, Boston, MA<br />

Background: NF1, NF2, and schwannomatosis are a group <strong>of</strong> related<br />

genetic disorders in which affected individuals share the predisposition to<br />

develop multiple nerve sheath tumors. While previous studies have investigated<br />

the relationship between cutaneous tumor burden and quality <strong>of</strong> life,<br />

the relation between internal tumors and quality <strong>of</strong> life is unknown.<br />

Methods: As part <strong>of</strong> an IRB-approved research study, we performed<br />

whole-body MRI and administered the short form (SF)-36 to 245 adult<br />

subjects with NF. The number and location <strong>of</strong> internal nerve sheath tumors<br />

in each patient was identified by a board-certified radiologist and tumor<br />

volume was calculated using semi-automated volumetric analysis. One<br />

sample t-tests were used to compare subjects’ SF-36 scores to general<br />

population means. Independent linear regression analyses controlling for<br />

age and gender effects were used to relate whole-body tumor count,<br />

volume, and distribution (via Gini coefficient) to each domain <strong>of</strong> the SF-36.<br />

Results: 245 patients (142 with NF1, 53 with NF2, 50 with schwannomatosis)<br />

completed the study. On the SF-36, subjects with NF1 showed<br />

reduced quality <strong>of</strong> life in the physical role, emotional role, and mental<br />

health domains compared to the normal population (p�0.05). Subjects<br />

with NF2 showed reductions in the physical functioning, physical role,<br />

general health, and social functioning domains while subjects with schwannomatosis<br />

showed reductions in the physical role and bodily pain domains<br />

(p�0.05). In linear regression analysis, increased tumor number, increased<br />

tumor volume, and decreased Gini coefficient were correlated with<br />

decreased physical functioning in patients with NF2 (p�0.01). There was<br />

also a trend for increasing tumor volume to be correlated with decreased<br />

physical role and increased bodily pain in patients with NF1 and with<br />

increased bodily pain in patients with schwannomatosis (p�0.10).<br />

Conclusions: In our multi-institutional cohort, patients with all forms <strong>of</strong><br />

neur<strong>of</strong>ibromatosis show selected deficits in quality <strong>of</strong> life. Internal tumor<br />

burden does not correlate with these deficits, with the exception <strong>of</strong> physical<br />

function in NF2 patients.<br />

Health Services Research<br />

415s<br />

6135 General Poster Session (Board #13E), Mon, 1:15 PM-5:15 PM<br />

Patient-reported toxicity after treatment for cancers <strong>of</strong> the head and neck.<br />

Presenting Author: Christine E. Hill-Kayser, Hospital <strong>of</strong> the University <strong>of</strong><br />

Pennsylvania, Philadelphia, PA<br />

Background: Patients with cancers <strong>of</strong> the head and neck (HNC) may be at<br />

risk for significant late effects after treatment. Quality <strong>of</strong> life may be altered<br />

significantly as a result. This Internet based study evaluates patient<br />

perceptions <strong>of</strong> toxicity after treatment for HNC. Methods: Patient-reported<br />

data were gathered via a convenience sample frame from HNC survivors<br />

voluntarily utilizing a publically available, free, Internet-based tool for<br />

creation <strong>of</strong> survivorship care plans. Available at www.livestrongcareplan.com<br />

and through the OncoLinkwebsite, the tool allows survivors to enter data<br />

regarding diagnosis, demographics, and treatments, and provides customized<br />

guidelines for future care. During use <strong>of</strong> the tool, HNC survivors are<br />

queried regarding late effects associated with specific treatments. All data<br />

have been maintained with IRB approval. Results: Of 3866 survivors using<br />

this tool between 4/2010 and 10/2011, 140 (4%) had undergone primary<br />

treatment for HNC. Median diagnosis age was 52y and median current age<br />

56y. When queried regarding treatment received, 88% reported having<br />

undergone radiation, 73% surgery, and 67% chemotherapy. Many patientreported<br />

late effects were associated with local tumor control, and included<br />

difficulty swallowing/speaking (83%), decreased saliva production (88%),<br />

dental problems (49%), thyroid problems (33%), decreased neck mobility<br />

(60%), eye and vision deficits (9%), and tinnitus (40%). In addition, 53%<br />

noted concerns regarding cognitive function. Users <strong>of</strong> the tool reported that<br />

they would share it with their healthcare team in 55% <strong>of</strong> cases. The most<br />

common reasons for not sharing it were “I did not think they would care,”<br />

and “ I did not want to upset or anger them.” Conclusions: Survivors using<br />

this tool anonymously and voluntarily report significant toxicity after<br />

treatment for HNC. Not all survivors appear comfortable discussing these<br />

issues with healthcare providers, despite major potential impact on quality<br />

<strong>of</strong> life. The data reported here may be <strong>of</strong> significant impact in future study<br />

<strong>of</strong> outcomes after HNC, as well as patient counseling and survivor care.<br />

6137 General Poster Session (Board #13G), Mon, 1:15 PM-5:15 PM<br />

Predictive factors on the receipt and completion <strong>of</strong> adjuvant chemotherapy<br />

in a mixed sociodemographic cohort <strong>of</strong> patients. Presenting Author: Blase<br />

N. Polite, The University <strong>of</strong> Chicago , Chicago, IL<br />

Background: African <strong>American</strong>s are more likely to die from breast, colon and<br />

lung cancer compared to whites. They are also less likely to receive and<br />

complete recommended therapy. The reasons for this are unknown.<br />

Methods: Breast, colon, and lung cancer patients eligible for adjuvant<br />

chemotherapy (stage IB-III breast, stage IIB-III colon, and Stage IB, II<br />

lung) and who received surgery at 2 urban medical centers, were approached<br />

between 2008 and 2010 and completed a self-administered<br />

survey focusing on sociodemographic factors, including social support,<br />

anxiety, depression, co-morbidities, trust and God Locus <strong>of</strong> Control (e.g.,<br />

Whatever happens to my cancer is God’s will). These variables were related<br />

to receipt and completion <strong>of</strong> chemotherapy. 191 patients were approached,<br />

161 consented and 30 refused (16%); out <strong>of</strong> the 161, 23 were<br />

deemed ineligible because <strong>of</strong> previous cancers, non-curative stage or<br />

receipt <strong>of</strong> chemotherapy in the neoadjuvant setting. Results: Final sample<br />

consisted <strong>of</strong> 138 pts. Median age: 54; sex:81% F; Race/Ethnicity:47% AA,<br />

38% white, 9% Hispanic, 6% Asian/PI; Cancer Type: 73% Breast, 21%<br />

Colon, 6% Lung. Compared to whites, AA were more likely to have<br />

incomes�20K (35% vs. 10%, p�.0002), have any comorbidities (80% vs.<br />

57%, p�0.006), and have high levels <strong>of</strong> God Locus <strong>of</strong> Control (56% vs<br />

27%, p�0.003). There were no differences by race in chemotherapy<br />

receipt (89% vs. 88%) or relative dose intensity (RDI) <strong>of</strong> chemo��85%<br />

(58% vs. 67%). No factor in our model predicted chemotherapy receipt or<br />

RDI at a significant level. Pts with high God Locus <strong>of</strong> Control were<br />

somewhat more likely to start chemotherapy (OR 1.7, p�0.25) but were<br />

less likely to complete therapy (OR 0.55, p�0.19). The magnitude <strong>of</strong> these<br />

relationships held after controlling for age, race, comorbidities, cancer<br />

type, functional status, and social support. Conclusions: In a mixed<br />

sociodemographic cohort <strong>of</strong> patients, receipt and completion <strong>of</strong> adjuvant<br />

chemotherapy was similar by measured sociodemographic variables. Only<br />

high God Locus <strong>of</strong> Control was suggestive <strong>of</strong> being less likely to complete<br />

chemotherapy. Further attention and research on the role <strong>of</strong> religious<br />

beliefs in the cancer decision making process is warranted.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


416s Leukemia, Myelodysplasia, and Transplantation<br />

6500^ Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Effect <strong>of</strong> anti-CD19 BiTE blinatumomab on complete remission rate and<br />

overall survival in adult patients with relapsed/refractory B-precursor ALL.<br />

Presenting Author: Max Topp, Department <strong>of</strong> Internal Medicine II, Division<br />

<strong>of</strong> Hematology and Medical Oncology, Wuerzburg, Germany<br />

Background: Blinatumomab is a bispecific T-cell engaging (BiTE) antibody<br />

that directs cytotoxic T-cells to CD19 expressing target cells. Methods: In<br />

adult patients with relapsed/refractory B-precursor ALL, a phase II dose<br />

ranging trial is being conducted to evaluate efficacy and safety <strong>of</strong><br />

blinatumomab. The primary endpoint is the rate <strong>of</strong> hematological complete<br />

remission (CR) or CR with partial hematological recovery (CRh*) within 2<br />

cycles <strong>of</strong> blinatumomab treatment. Blinatumomab is administered by<br />

continuous intravenous infusion for 28-days followed by a 14-day treatmentfree<br />

interval. Responding patients can receive 3 additional cycles <strong>of</strong><br />

treatment or proceed to bone marrow transplantation. Three dose levels<br />

were explored as shown in the table. Results: In total 36 patients have been<br />

enrolled, 25 are currently evaluable. Seventeen out <strong>of</strong> 25 treated patients<br />

(68%) reached a hematological CR/CRh* and a minimal residual disease<br />

(MRD) response (MRD level �10 -4 ) within the first 2 cycles. Five out <strong>of</strong> 17<br />

responders (29%) showed a CRh* due to partial recovery <strong>of</strong> platelets. For<br />

the first 18 patients, response duration is 7.1 months and the median<br />

follow-up time for overall survival (OS) is 9.7 months (median not reached).<br />

Six cases <strong>of</strong> relapses have been recorded <strong>of</strong> which 3 were CD19� , and 3<br />

CD19- . As final dose 5 �g/m²/day in week 1 and 15 �g/m²/day for the<br />

remaining treatment (cohort 2a and 3) was selected. In these cohorts<br />

(n�12), the most common treatment emergent adverse events (TEAEs, all<br />

grade 1-2) were pyrexia (67%), headache (33%) and tremor (33%). TEAEs<br />

<strong>of</strong> grade �3 (7 in 5 patients, no grade 4), irrespective <strong>of</strong> relationship, were<br />

infections, confusion, epilepsy, hypertension and thrombocytopenia.<br />

Conclusions: The final dose was well-tolerated and produced an exceptionally<br />

high complete remission rate. A global phase 2 study to confirm these<br />

data is underway.<br />

Summary <strong>of</strong> dose cohorts and outcomes (Oct 2011).<br />

Cohort<br />

Patients<br />

treated<br />

Week 1, cycle 1<br />

(�g/m 2 /day)<br />

Week 2,<br />

cycle 1<br />

(�g/m 2 /day)<br />

Weeks 3-4,<br />

cycle 1 and<br />

subsequent cycles<br />

(�g/m2 /day) CR/CRh*<br />

1 7 15 15 15 5<br />

2a (final dose) 5 5 15 15 4<br />

2b 6 5 15 30 3<br />

3 (final dose) 7 5 15 15 5<br />

Total 25 17<br />

6502 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

A phase II trial <strong>of</strong> azacitidine (NSC-102816) and gemtuzumab ozogamicin<br />

(NSC-720568) as induction and post-remission therapy in patients <strong>of</strong> age<br />

60 and older with previously untreated non-M3 acute myeloid leukemia:<br />

SWOG S0703 protocol—Report on the good-risk patients. Presenting<br />

Author: Sucha Nand, Loyola University Medical Center, Maywood, IL<br />

Background: In pts with AML, response rates to standard chemotherapy<br />

decrease and early death rates increase with age. Whereas pts � 56 yrs <strong>of</strong><br />

age have a complete remission (CR) rate <strong>of</strong> 65% and 30 day mortality risk<br />

<strong>of</strong> �3%, the respective figures are 33% and 31% in pts � 75 yrs. This trial<br />

was designed to improve safety and efficacy by using agents with low<br />

toxicity, different mechanisms <strong>of</strong> action and possible synergy. The treatment<br />

could be given in the outpatient setting. Methods: Newly diagnosed<br />

pts with non-M3 AML, 60 yrs or older, were treated as follows: Induction:<br />

Hydroxyurea 1500 mg b.i.d till WBC �10,000/mcL, followed by azacitidine<br />

75 mg/m2/day s/cu or iv days 1-7, gemtuzumab ozogamicin (GO)<br />

3mg/m2 on D8. Induction treatment was repeated for residual disease on<br />

D14. Pts achieving CR received a consolidation treatment identical to the<br />

induction treatment. This was followed by 4 cycles <strong>of</strong> azacitidine 75/m2/<br />

day, D1-7, given q 4 weeks. Subsequent therapy was optional. Promoter<br />

and global methylation studies were performed at defined time points.<br />

Results: Pts were stratified into good risk (age 60-69 or Zubrod 0-1) and<br />

poor risk (age �70 and Zubrod 2 or 3) groups. The results presented here<br />

are from the good risk cohort. Eighty-three pts were entered <strong>of</strong> whom 79 are<br />

evaluable. Median age was 71 yrs (60-88) and 49 were males. Thirty-five<br />

achieved CR/CRi (44%: 95% CI 33%-56%). Median overall survival was<br />

11 months and median relapse-free survival with patients achieving a<br />

CR/CRi was 8 months. Six patients (8%), 3 with disease progression, died<br />

within 30 days <strong>of</strong> registration. The main non-hematologic toxicity was<br />

neutropenic fever (Grade 3 or higher), seen in 27 patients. Conclusions: The<br />

combination <strong>of</strong> hydroxyurea, azacitidine and GO is associated with a low<br />

induction mortality, can be given in the outpatient setting and produces<br />

results which compare favorably with standard chemotherapy regimens in<br />

elderly patients with AML. Our results are sufficiently encouraging to<br />

warrant further studies <strong>of</strong> this approach. <strong>Clinical</strong> Trials.govIdentifier:<br />

NCT00658814.<br />

6501 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Inotuzumab ozogamycin (I0), a CD22 monoclonal antibody conjugated to<br />

calecheamicin, given weekly, for refractory-relapse acute lymphocytic<br />

leukemia (R-R ALL). Presenting Author: Elias Jabbour, University <strong>of</strong> Texas<br />

M. D. Anderson Cancer Center, Houston, TX<br />

Background: Patients (pts) with R-R ALL have a poor prognosis. CD22 is<br />

highly expressed in ALL. We have previously reported on 49 pts with R-R<br />

ALL treated with IO at the dose <strong>of</strong> 1.8 mg/m2 every 3-4 weeks; the overall<br />

response rate was 57%. From preclinical studies, lower dose more frequent<br />

exposure schedules to IO may <strong>of</strong>fer better anti-ALL activity. Methods: Pts<br />

with R-R ALL received IO 0.8 mg/m2 on Day 1, 0.5 mg/m2 on Day 8, and<br />

0.5 mg/m2 on Day 15. The total dose per course remained the same 1.8<br />

mg/m2. Courses were given every 3-4 weeks. Early stopping rules were<br />

implemented for an overall response rate � 40%. Results: 20 pts were<br />

treated so far. Median age was 57 yrs (range 22–84). Cytogenetics were:<br />

Ph� in 7 (35%), t(4:11) in 2 (10%) , diploid in 3 (15%), and other in 8<br />

(40%). CD22 was expressed in � 50% blasts in all pts and in � 90% in 14<br />

(70%). Median number <strong>of</strong> courses so far was 2 (1–4). 9 (45%) pts received<br />

IO as salvage 1 (S1), 5 (25%) as salvage 2 (S2), 6 (30%) as salvage 3 (S3).<br />

one pt had allo SCT before IO. Median follow-up is 22 weeks (range, 7-43).<br />

Overall, 2/20 pts (10%) achieved CR, 6 had CRp (30%), 2 had marrow CR<br />

(10%), 8 were resistant, 2 died within 4 wks. Overall, CR�CRp�mCR (OR)<br />

was 50%: S1 6/9 (67%); S2 2/5 (40%); S3 2/6 (33%). Of the 10<br />

responders, 7 pts (70%) became MRD negative. There were no clear<br />

correlations between OR and pts/ALL characteristics or with CD22 expression<br />

vs. � 90%. In 16 pts with evaluable pharmacokinetics studies, the<br />

median end <strong>of</strong> the infusion levels <strong>of</strong> IO were 117 ng/ml and 77 ng/ml in<br />

responders and non-responders, respectively. Median survival was 7�<br />

months. Median OR duration was 5� months. Median survival in 10<br />

responders has not been reached. Liver function abnormalities (LFA) were<br />

noted as IO related in 7 (35%) pts, severe and reversible in 2 (10%). 4 pts<br />

were able to proceed to allo SCT (1 in CR, 3 in CRp) after a median <strong>of</strong> 4<br />

weeks from the last infusion <strong>of</strong> IO; No cases <strong>of</strong> veno-occlusive disease were<br />

observed. Conclusions: IO given weekly is active in R-R ALL with an OR <strong>of</strong><br />

50%. LFAs occur and are reversible. This study <strong>of</strong> IO weekly schedule is<br />

ongoing. IO and chemotherapy combinations are being pursued.<br />

6503 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

PACE: A pivotal phase II trial <strong>of</strong> ponatinib in patients with CML and<br />

Ph�ALL resistant or intolerant to dasatinib or nilotinib, or with the T315I<br />

mutation. Presenting Author: Jorge E. Cortes, University <strong>of</strong> Texas M. D.<br />

Anderson Cancer Center, Houston, TX<br />

Background: Ponatinib is a potent, oral, pan-BCR-ABL inhibitor active<br />

against the native enzyme and all tested resistant mutants, including the<br />

uniformly resistant T315I mutation. Methods: The PACE (Ponatinib Ph�ALL<br />

and CML Evaluation) trial started Sept 2010. Pts with refractory CML (CP,<br />

AP or BP) or Ph�ALL resistant or intolerant (R/I) to dasatinib or nilotinib or<br />

with T315I received 45 mg ponatinib once daily. The trial is ongoing;<br />

enrollment completed Sept 2011. Data as <strong>of</strong> 17 Jan 2012 are reported.<br />

Results: 449 pts were enrolled, 5 <strong>of</strong> whom were ineligible (post-imatinib,<br />

non-T315I) but treated. Median age was 59 (18-94) yrs; 53% male.<br />

Diagnoses were: 271 CP-CML (R/I�207; T315I�64); 79 AP-CML (R/<br />

I�60; T315I�19); 94 BP/ALL (R/I�48; T315I�46). Median time from<br />

diagnosis to ponatinib was 6 yrs. Prior TKIs included imatinib (96%),<br />

dasatinib (85%), nilotinib (66%), bosutinib (7%); 94% failed �2 prior<br />

TKIs, 59% failed �3 prior TKIs. 83% had a history <strong>of</strong> resistance to<br />

dasatinib or nilotinib; 12% were purely intolerant. In CP, best response to<br />

most recent dasatinib or nilotinib was MCyR 25%. Frequent mutations<br />

confirmed at entry: 29% T315I, 8% F317L, 4% E255K, 4 % F359V, 3%<br />

G250E. Median follow-up was 6.6 months. Response rates are presented<br />

in the table. Overall, 64% remained on therapy (77% CP). Most frequent<br />

reasons for discontinuation were progression (12%) and AE (10%). Most<br />

common drug-related AEs were thrombocytopenia (33%), rash (33%), dry<br />

skin (26%). Conclusions: Ponatinib has substantial activity in heavily<br />

pretreated pts and those with refractory T315I. Response rates continue to<br />

improve with longer follow-up. Multivariate analyses <strong>of</strong> predictors <strong>of</strong><br />

outcome will be presented.<br />

n Response to ponatinib / N evaluable (%)<br />

Overall R/I T315I<br />

CP-CML<br />

MCyR* 126/258 (49) 88/197 (45) 38/61 (62)<br />

CCyR 105/258 (41) 70/197 (36) 35/61 (57)<br />

MMR 68/265 (26) 40/205 (20) 28/60 (47)<br />

AP-CML<br />

MHR* 38/57 (67) 31/43 (72) 7/14 (50)<br />

MCyR 27/72 (38) 18/55 (33) 9/17 (53)<br />

CCyR 12/72 (17) 8/55 (15) 4/17 (24)<br />

BP-CML/Ph�ALL<br />

MHR* 33/89 (37) 17/46 (37) 16/43 (37)<br />

MCyR 30/82 (37) 14/41 (34) 16/41 (39)<br />

CCyR 23/82 (28) 11/41 (27) 12/41 (29)<br />

*Primary endpoints: MCyR in CP; MHR in AP, BP/Ph�ALL (baseline MHR excluded).<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


6504 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Dasatinib versus imatinib (IM) in newly diagnosed chronic myeloid leukemia<br />

in chronic phase (CML-CP): DASISION 3-year follow-up. Presenting<br />

Author: Andreas Hochhaus, Universitätsklinikum Jena, Jena, Germany<br />

Background: In the phase 3 DASISION trial <strong>of</strong> dasatinib v IM in patients<br />

(pts) with newly diagnosed CML-CP, dasatinib had higher 12-month rates<br />

<strong>of</strong> complete cytogenetic response (CCyR) and major molecular response<br />

(MMR) (Kantarjian, NEJM 2010;362:2260). By 12 months confirmed<br />

CCyR (cCCyR) rates for dasatinib v IM were 77% v 66%, P�0.001,<br />

meeting the primary endpoint. Methods: Pts were randomized to receive<br />

dasatinib 100 mg once daily (QD; n�259) or IM 400 mg QD (n�260).<br />

Results: Minimum 24-month follow-up (median 26.6 months) is reported<br />

here. 24-month molecular response rates were higher for dasatinib v IM:<br />

MMR (BCR-ABL �0.1%) 64% v 46%, P�0.0001; MR4 (BCR-ABL<br />

�0.01%) 29% v 19%, P�0.0053; MR4.5 (BCR-ABL �0.0032%) 17% v<br />

8%, P�0.0032. MMR rates were higher for dasatinib in all Hasford risk<br />

groups (high 73% v 56%; intermediate 61% v 50%; low 73% v 56%). Of<br />

pts who achieved MMR at 12 months, on dasatinib v IM, 97% v 92% had<br />

maintained their MMR at 24 months, respectively. Pts receiving dasatinib v<br />

IM had faster responses; median time to CCyR and MMR was 3.2 v 6.0 and<br />

15 v 36 months, respectively. In an intent-to-treat analysis, fewer pts<br />

receiving dasatinib (n�9; 3.5%) transformed to accelerated/blast phase v<br />

IM (n�15; 5.8%) on study or during follow-up after discontinuation.<br />

24-month overall and progression-free survival were similar for dasatinib v<br />

IM: 95.4% v 95.2% and 93.7% v 92.1% (follow-up is ongoing). Few<br />

additional adverse events (AEs) were reported between 12 and 24 months<br />

in both arms, with grade 3/4 nonhematologic AE rates �1%. In each arm,<br />

10 pts had a BCR-ABL mutation detected at time <strong>of</strong> discontinuation. For<br />

dasatinib v IM, 23% v 25% discontinued treatment for drug-related AEs<br />

(7% v 5%), progression (5% v 7%), failure (3% v 4%), unrelated AEs (2% v<br />

�1%), death (2% v �1%), and other (4% v 8%). Few pts discontinued<br />

between 12 and 24 months for dasatinib (n�19; 7%) and IM (n�16; 6%).<br />

Conclusions: Updated data with minimum 36-month follow-up will be<br />

presented, including mutation analyses in pts who discontinued, progressed,<br />

or had suboptimal response. Pts receiving dasatinib had a lower<br />

transformation rate and higher molecular responses v pts receiving IM,<br />

supporting the use <strong>of</strong> dasatinib in newly diagnosed CML-CP.<br />

6506 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Six-year (yr) follow-up <strong>of</strong> patients (pts) with imatinib-resistant or -intolerant<br />

chronic-phase chronic myeloid leukemia (CML-CP) receiving dasatinib.<br />

Presenting Author: Neil P. Shah, University <strong>of</strong> California, San Francisco,<br />

San Francisco, CA<br />

Background: Dasatinib, a potent BCR-ABL inhibitor, is approved for use as<br />

1st- and 2nd-line therapy for CML pts with newly diagnosed disease or<br />

resistance/intolerance to imatinib. This ongoing study in pts with imatinibresistant/-intolerant<br />

CML provides the longest follow-up <strong>of</strong> a secondgeneration<br />

BCR-ABL inhibitor. Methods: Study design has been described<br />

(Shah, J Clin Oncol 2008). Pts with imatinib-resistant/-intolerant CML<br />

(N�670) were randomized to dasatinib 100 mg once daily (QD), 50 mg<br />

twice daily (BID), 140 mg QD, or 70 mg BID. Results: Five-yr data are<br />

reported here; 6-yr data will be presented. After a minimum <strong>of</strong> 5 yrs, 151<br />

pts (74%) remain on QD dosing, 85 <strong>of</strong> whom (56%) are on �100 mg QD<br />

dosing. Overall, 205 pts (31%) remain on study therapy with 55 pts (53%)<br />

originally randomized to BID dosing having switched to QD dosing. For pts<br />

randomized to the 100 mg QD arm (n�167), progression-free survival<br />

(PFS) at 5 yrs is 57%, overall survival (OS) is 78% with an overall 5% rate<br />

<strong>of</strong> transformation to advanced disease. In exploratory analyses, 42% and<br />

60% <strong>of</strong> pts had BCR-ABL levels �10% (International Scale) at 1 and 3<br />

months (mos), respectively. In a landmark analysis, BCR-ABL �10% at 1<br />

or 3 mos was associated with higher 5-yr PFS. For dasatinib 100 mg QD,<br />

nonhematologic adverse events (AEs; all grades) generally first occurred in<br />

�24 mos. Cumulative rates <strong>of</strong> AEs in the 100 mg QD arm were headache<br />

(33%), diarrhea (28%), fatigue (26%), and pleural effusion (24%). For<br />

dasatinib 100 mg QD, cytopenias (grades 3/4) generally first occurred in<br />

�12 mos. The most common hematologic AEs (grades 3/4) in the 100 mg<br />

QD arm were neutropenia (36%) and thrombocytopenia (24%). AEs were<br />

managed by dose/schedule modifications. Conclusions: Five-yr follow-up <strong>of</strong><br />

pts who switched to dasatinib 100 mg QD after imatinib-resistance/<br />

intolerance shows high rates <strong>of</strong> PFS, and OS with an overall low rate <strong>of</strong><br />

transformation. Exploratory analyses suggest that achievement <strong>of</strong> a fast and<br />

deep response (�10% BCR-ABL) at 1 or 3 mos after initiation <strong>of</strong> dasatinib<br />

100 mg QD may be associated with a higher PFS. Dasatinib 100 mg QD<br />

was generally well tolerated over 5 yrs. Six-yr data will be presented.<br />

Leukemia, Myelodysplasia, and Transplantation<br />

6505^ Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Switch to nilotinib versus continued imatinib in patients (pts) with chronic<br />

myeloid leukemia in chronic phase (CML-CP) with detectable BCR-ABL<br />

after 2 or more years on imatinib: ENESTcmr 12-month (mo) follow-up.<br />

Presenting Author: Jeffrey Howard Lipton, Princess Margaret Hospital,<br />

Toronto, ON, Canada<br />

Background: Nilotinib induced significantly faster and deeper molecular<br />

responses vs imatinib in the ENESTnd trial. Achieving these deeper<br />

molecular responses may increase patient eligibility for future TKI discontinuation<br />

studies. Methods: CML-CP pts (N � 207) who achieved a<br />

complete cytogenetic response but were still BCR-ABL positive by RQ-PCR<br />

after � 24 mo on imatinib were randomized 1:1 to receive nilotinib 400 mg<br />

BID (n � 104) or to continue their imatinib dose (400 or 600 mg QD [n �<br />

103]). The primary endpoint was confirmed CMR (undetectable BCR-ABL<br />

by RQ-PCR with a sample sensitivity <strong>of</strong> � 4.5 logs in 2 consecutive<br />

samples). Other endpoints included molecular responses (MMR � 0.1% IS ,<br />

MR4 � 0.01% IS , and MR4.5 � 0.0032% IS ) and BCR-ABL ratio over time.<br />

Results: Rate <strong>of</strong> confirmed CMR was higher in the nilotinib arm vs imatinib<br />

by 12 mo (12.5% vs 5.8%) (Table). Rate <strong>of</strong> CMR (undetectable BCR-ABL<br />

in at least 1 sample) by 12 mo was significantly higher on nilotinib vs<br />

imatinib (23.1% vs 10.7%; P � .02). Rates <strong>of</strong> MMR, MR4 ,MR4.5 , and<br />

CMR were also superior in pts switched to nilotinib, and these pts had<br />

significantly shorter times to achieve these responses. Imatinib-treated pts<br />

had minimal evidence <strong>of</strong> improvement in molecular response vs a median<br />

0.5-log reduction in BCR-ABL by 12 mo for the nilotinib cohort. With<br />

12-mo follow-up, 84% <strong>of</strong> pts remained on nilotinib and 96% on imatinib.<br />

The nilotinib safety pr<strong>of</strong>ile was consistent with prior studies. Both drugs<br />

were well tolerated. Conclusions: Twice as many pts achieved deeper<br />

molecular responses after switching to nilotinib vs staying on imatinib.<br />

Nilotinib<br />

400 mg BID<br />

(n � 104)<br />

Molecular response by 12 mo (ITT population), %<br />

Confirmed CMR 12.5<br />

P � .108*<br />

CMR 23.1<br />

P � .02*<br />

Molecular response by 12 mo (in pts without the response at baseline), %<br />

MMR n � 24<br />

75.0<br />

MR<br />

P � .006**<br />

4 n � 74<br />

48.6<br />

MR<br />

P � .006**<br />

4.5 n � 94<br />

33.0<br />

P � .008**<br />

CMR<br />

*Stratified Cochran-Mantel-Haenszel test. **Stratified log-rank test.<br />

n � 101<br />

20.8<br />

P � .03**<br />

417s<br />

Imatinib<br />

400 or 600 mg QD<br />

(n � 103)<br />

5.8<br />

10.7<br />

n � 28<br />

35.7<br />

n � 78<br />

25.6<br />

n � 91<br />

16.5<br />

n � 100<br />

10.0<br />

6507 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

The Bruton’s tyrosine kinase (BTK) inhibitor PCI-32765 (P) in treatmentnaive<br />

(TN) chronic lymphocytic leukemia (CLL) patients (pts): Interim<br />

results <strong>of</strong> a phase Ib/II study. Presenting Author: John C. Byrd, The Ohio<br />

State University Comprehensive Cancer Center and Arthur G. James Cancer<br />

Hospital and Solove Research Institute, Columbus, OH<br />

Background: Fludarabine based therapy, while effective, carries significant<br />

risk <strong>of</strong> morbidity and mortality in the elderly pts. As such, older CLL pts<br />

represent a high priority for new therapeutic approaches. PCI-32765 (P) an<br />

oral, selective, irreversible inhibitor <strong>of</strong> BTK inhibits CLL cell proliferation,<br />

migration and adhesion. A multi-cohort Phase Ib/II trial evaluated 2 doses<br />

<strong>of</strong> single-agent P in both TN and relapsed/ refractory (R/R) CLL/SLL pts.<br />

Mature follow-up <strong>of</strong> the TN pts is reported. Methods: Pts �65 yrs old with<br />

active CLL requiring Tx by IWCLL guidelines were treated with oral P at<br />

doses <strong>of</strong> 420 mg or 840 mg administered daily for 28-day cycles until<br />

disease progression (PD). Response was evaluated according to 2008<br />

IWCLL criteria. Results: 31 pts were enrolled- 26 pts (420mg) and 5 pts<br />

(840 mg). The 840 mg cohort was terminated after comparable activity and<br />

safety between doses was shown in R/R pts. Median age 71 yrs (range<br />

65-84) with 74% <strong>of</strong> pts �70 yrs. 19/31 (61%) had baseline cytopenias<br />

(Hgb � 11g/dl or plts �100,000). Unmutated IgVH was present in 43% <strong>of</strong><br />

pts. The majority <strong>of</strong> AEs have been Gr��2 in severity, most commonly<br />

diarrhea, nausea, and fatigue. Gr �3 non-heme AEs potentially related to P<br />

in 19% <strong>of</strong> pts. 10% <strong>of</strong> pts experienced Gr �3 infections or cytopenias. With<br />

a median follow-up <strong>of</strong> 10.7 mos on 420 mg cohort 73% (19/26) achieved a<br />

response by IWCLL criteria with 65% partial responses (PR) and 8%<br />

complete remissions with no morphologic evidence <strong>of</strong> CLL on marrow. An<br />

additional 12% (3/26) <strong>of</strong> pts achieved nodal responses (NR) with lymphocytosis.<br />

Median follow-up in the 840 mg cohort is 4.6 mo, at 2 cycle<br />

assessment 2/5 achieved a PR and 1 pt with a NR. ORR was independent <strong>of</strong><br />

high risk factors. 84% <strong>of</strong> pts remain on study, reasons for discontinuation<br />

� AE (3), investigator decision (1) and PD (1). There have been no deaths.<br />

Estimated 12 mo median PFS for the 420 mg cohort is 93%. Conclusions:<br />

PCI-32765 is highly active and well tolerated in elderly TN CLL pts. The<br />

high ORR, including marrow clearance and very low PD rate with the single<br />

agent suggests that P warrants further study as a first-line treatment<br />

approach in elderly pts.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


418s Leukemia, Myelodysplasia, and Transplantation<br />

6508 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

A phase Ib/II study evaluating activity and tolerability <strong>of</strong> BTK inhibitor<br />

PCI-32765 and <strong>of</strong>atumumab in patients with chronic lymphocytic leukemia/<br />

small lymphocytic lymphoma (CLL/SLL) and related diseases. Presenting<br />

Author: Samantha Mary Jaglowski, The Ohio State University, Columbus,<br />

OH<br />

Background: Bruton’s tyrosine kinase (BTK) is a non-receptor kinase that is<br />

critical for B-cell receptor (BCR) signaling in normal and malignant B<br />

lymphocytes. PCI-32765 (P), an oral, potent and irreversible BTK inhibitor,<br />

antagonizes BCR signaling in CLL cells and abrogates protective<br />

features <strong>of</strong> the microenvironment. P is highly active as a single agent in<br />

CLL/SLL patients (pts), and this phase Ib/II study builds upon single-agent<br />

experience by combining P with <strong>of</strong>atumumab (O), an anti-CD20 monoclonal<br />

antibody. We present initial safety and efficacy data from cohort 1.<br />

Methods: Pts with relapsed/refractory (R/R) CLL/SLL following �2 prior<br />

therapies (Tx), including a purine-nucleoside analog (PA), are treated with<br />

420 mg P daily, in 28-day cycles, until disease progression. O is added at a<br />

dose <strong>of</strong> 300 mg on day (D) 1 <strong>of</strong> cycle 2, followed by 2000 mg on D8, 15,<br />

and 22 <strong>of</strong> cycle 2, D1, 8, 15, and 22 <strong>of</strong> cycle 3, and on D1 <strong>of</strong> cycles 5-8.<br />

Results: As <strong>of</strong> November 2011, 27 patients with either CLL/SLL/PLL<br />

(n�24) or Richter’s transformation (RT, n�3) have been enrolled and have<br />

received at least 6 cycles <strong>of</strong> treatment. The median age is 66 (range<br />

51-85), 9 were Rai stage III/IV. Median number <strong>of</strong> prior Tx is 3 (range<br />

2-10), 15 pts had bulky disease (� 5 cm); 11 pts were PA refractory.<br />

Poor-risk molecular features were common (del(17p) 10 pts, del(11q) 9<br />

pts). No grade (G) 3 or 4 infusion reactions, neutropenia, or thrombocytopenia<br />

have been observed. The majority <strong>of</strong> adverse events (AE) were G1/2.<br />

G3/4 AE included anemia (11%), pneumonia (11%), UTI (7%), hyponatremia<br />

(7%). 24/24 CLL/SLL/PLL pts have achieved PR (100% ORR) within 6<br />

cycles; 2/3 RT pts had PR. With median follow-up <strong>of</strong> 6.5 mo (range<br />

5.3-10.2 mo), 23 CLL/SLL/PLL pts and 1 RT pt remain on study; 1<br />

CLL/SLL pt went to transplant in PR; 2 RT pts progressed. Conclusions:<br />

PCI-32765 combined with <strong>of</strong>atumumab is well tolerated and highly active<br />

(100% ORR) in pts with heavily pre-treated R/R CLL/SLL. Rapid onset <strong>of</strong><br />

response, low relapse rate, and favorable safety pr<strong>of</strong>ile make this combination<br />

worthy <strong>of</strong> further study. Cohorts evaluating other Tx sequences are<br />

currently underway.<br />

6510 Poster Discussion Session (Board #2), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

The prognostic significance <strong>of</strong> early molecular and cytogenetic response for<br />

long-term progression-free and overall survival in imatinib-treated chronic<br />

myeloid leukemia (CML). Presenting Author: Rudiger Hehlmann, Medizinische<br />

Fakultät Mannheim der Universität Heidelberg, Mannheim, Germany<br />

Background: In the face <strong>of</strong> competing first line treatment options for CML<br />

early prediction <strong>of</strong> prognosis on imatinib is desirable to assure favorable<br />

survival or otherwise consider the use <strong>of</strong> an alternative therapy. We sought<br />

to evaluate the prognostic impact <strong>of</strong> early response landmarks. Methods: A<br />

total <strong>of</strong> 1,303 newly diagnosed imatinib-treated patients (pts) from the<br />

randomized CML Study IV were investigated to correlate molecular and<br />

cytogenetic response at 3 and 6 months with progression-free and overall<br />

survival (PFS, OS). Median follow-up was 4.7 years (range 0-9). The<br />

BCR-ABL expression was determined by quantitative RT-PCR and standardized<br />

according to the international scale (BCR-ABLIS ). The proportion <strong>of</strong><br />

Philadelphia-chromosome positive metaphases (Ph�) was determined by<br />

conventional metaphase analysis. To confirm the prognostic significance <strong>of</strong><br />

early molecular response, an independent validation sample <strong>of</strong> 174 pts<br />

treated with imatinib within the IRIS trial was analyzed. Results: The<br />

persistence <strong>of</strong> �10% BCR-ABLIS at 3 months separated a high-risk group<br />

(28% <strong>of</strong> pts; 5-year OS: 87%) from a group with 1-10% BCR-ABLIS (41%<br />

<strong>of</strong> pts; 5-year OS: 94%; p�0.012), and from a group with �1% BCR-ABLIS (31% <strong>of</strong> pts; 5-year OS: 97%; p�0.004). By cytogenetics high-risk<br />

patients could be identified by the persistence <strong>of</strong> �35% Ph� (27% <strong>of</strong> pts;<br />

5-year OS: 87%) as compared to �35% Ph� (73% <strong>of</strong> pts; 5-year OS: 95%;<br />

p�0.036). At 6 months the �1% BCR-ABLIS group (37% <strong>of</strong> pts; 5-year<br />

OS: 89%) showed inferior survival compared to �1% (63% <strong>of</strong> pts; 5-year<br />

OS: 97%; p�0.001); survival <strong>of</strong> the �0% Ph� group (34% <strong>of</strong> pts; 5-year<br />

OS: 91%) was inferior to 0% Ph� (66% <strong>of</strong> pts; 5-year OS: 97%;<br />

p�0.015). Regarding the IRIS pts 3 month BCR-ABLIS �10% (25% <strong>of</strong><br />

pts; 8-year OS: 81%) was associated with inferior survival compared to<br />

�10% (75% <strong>of</strong> pts; 8-year OS: 93%; p�0.011). Conclusions: Failure to<br />

achieve the response landmarks <strong>of</strong> 10% BCR-ABLIS or 35% Ph� at 3<br />

months <strong>of</strong> imatinib treatment and 1% BCR-ABLIS or 0% Ph� at 6 months<br />

identifies high-risk patients which might benefit from an early change <strong>of</strong><br />

therapy.<br />

6509^ Poster Discussion Session (Board #1), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Nilotinib versus imatinib in patients (pts) with newly diagnosed chronic myeloid<br />

leukemia in chronic phase (CML-CP): ENESTnd 3-year (yr) follow-up (f/u).<br />

Presenting Author: Hagop Kantarjian, University <strong>of</strong> Texas M. D. Anderson Cancer<br />

Center, Houston, TX<br />

Background: In ENESTnd, nilotinib significantly reduced progression to AP/BC<br />

and demonstrated superior rates <strong>of</strong> MMR, MR4 , and MR4.5 vs imatinib with f/u <strong>of</strong><br />

2 yrs. Methods: 846 pts with Ph� CML-CP were randomized to nilotinib 300 mg<br />

BID (n � 282), nilotinib 400 mg BID (n � 281), or imatinib 400 mg QD (n �<br />

283). Here, we report 3-yr f/u data. Results: Both nilotinib doses continued to<br />

demonstrate significantly higher rates <strong>of</strong> MMR, MR4 , and MR4.5 vs imatinib. In a<br />

landmark analysis, pts with BCR-ABL transcript levels � 10% at 3 months (mo)<br />

had a higher probability <strong>of</strong> achieving MMR by 1 and 2 yrs vs pts with transcript<br />

levels � 10%. No new progressions occurred on treatment since the 2-yr<br />

analysis; rates <strong>of</strong> progression to AP/BC including events on treatment (n � 2, 3,<br />

12) and those occurring both on treatment and after discontinuation (n � 9, 6,<br />

19) were significantly lower for nilotinib 300 mg BID and nilotinib 400 mg BID<br />

vs imatinib, respectively. At 3 yrs, OS considering only CML-related deaths was<br />

significantly higher for nilotinib vs imatinib. The safety pr<strong>of</strong>iles <strong>of</strong> nilotinib and<br />

imatinib were similar to those at 2 yrs. Conclusions: 3-yr f/u confirms the<br />

superiority <strong>of</strong> nilotinib vs imatinib and an acceptable tolerability pr<strong>of</strong>ile for the<br />

treatment <strong>of</strong> pts with newly diagnosed Ph� CML-CP.<br />

Nilotinib<br />

300 mg BID<br />

(n � 282)<br />

Response by 3 yrs, %<br />

MMR 73<br />

P � .0001<br />

MR4 50<br />

P � .0001<br />

MR4.5 32<br />

Nilotinib<br />

400 mg BID<br />

(n � 281)<br />

Imatinib<br />

400 mg QD<br />

(n � 283)<br />

70<br />

53<br />

P � .0001<br />

44<br />

26<br />

P � .0001<br />

28<br />

15<br />

3-mo landmark analysis: BCR-ABL transcript levels*, %<br />

< 1% (n � 120, 123, 41)<br />

> 1to< 10% (n � 89, 95, 133)<br />

> 10% (n � 24, 28, 88)<br />

Freedom from progression to AP/BC, %<br />

Estimated 3-yr rate on core treatment<br />

P � .0001<br />

76/89<br />

40/67<br />

4/29<br />

99.3<br />

P � .0003<br />

MMR by 1 yr/2 yrs, %<br />

72/91<br />

38/54<br />

14/29<br />

98.7<br />

71/78<br />

31/52<br />

2/20<br />

95.2<br />

Including events after discontinuation<br />

P � .0059<br />

96.7<br />

P � .0185<br />

98.1<br />

93.5<br />

OS, %<br />

Estimated 3-yr OS rate<br />

P � .0496<br />

95.1<br />

P � .0076<br />

97.0<br />

94.0<br />

Only CML-related deaths<br />

P � .4413<br />

98.1<br />

P � .0639<br />

98.5<br />

95.2<br />

Patients with BCR-ABL mutation, n<br />

Any mutation<br />

T315I<br />

P � .0356<br />

11<br />

3<br />

P � .0159<br />

11<br />

2<br />

21<br />

3<br />

* Patients with unevaluable/missing PCR assessments at 3 mo, atypical transcripts, or MMR by 3 mo were<br />

excluded.<br />

6511 Poster Discussion Session (Board #3), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Efficacy and safety <strong>of</strong> bosutinib (BOS) for Philadelphia chromosome–<br />

positive (Ph�) leukemia in older versus younger patients (pts). Presenting<br />

Author: Tim H. Brummendorf, Universitätsklinikum Aachen, Universitätsklinikum<br />

Hamburg-Eppendorf, Aachen and Hamburg, Germany<br />

Background: BOS is an oral dual Src/Abl kinase inhibitor with potent activity<br />

in Ph� leukemia. Methods: Efficacy and safety <strong>of</strong> BOS 500 mg/d was<br />

evaluated in older (�65 y; n � 119) and younger (�65 y; n � 451) pts in 3<br />

cohorts: chronic phase chronic myeloid leukemia (CP CML) after imatinib<br />

(IM; CP2L cohort; n � 287); CP CML after IM � dasatinib (DAS) and/or<br />

nilotinib (NIL; CP3L cohort; n � 119); and accelerated/blast phase<br />

(AP/BP) CML or acute lymphoblastic leukemia after IM � DAS and/or NIL<br />

(ADV cohort; n � 164). Results: Baseline events (�65yvs�65 y) included<br />

respiratory disorders (35% vs 13%), cardiac disorders (29% vs 9%), and<br />

diabetes (4% vs 4%). Median baseline medications were 3 (�65 y) and 5<br />

(�65 y). Median BOS duration was 11 mo and median follow-up was 31<br />

mo for all pts. 80% <strong>of</strong> ³65 y and 67% <strong>of</strong> �65 y pts discontinued BOS,<br />

including 32% and 18% due to an adverse event (AE; most commonly<br />

thrombocytopenia [6% vs 3%]). Rates <strong>of</strong> response were similar or lower in<br />

older versus younger pts (Table). On-treatment transformation to AP/BP<br />

CML was similar between groups. Incidences <strong>of</strong> nonhematologic treatmentemergent<br />

AEs were generally similar between older and younger pts,<br />

notably (all grades/grade �3 for �65yvs�65 y): diarrhea (85%/9% vs<br />

81%/8%), infection (56%/15% vs 49%/10%), and edema (8%/0% vs<br />

4%/�1%). Common grade �3 lab abnormalities (�65yvs�65 y) were<br />

thrombocytopenia (35% vs 35%), neutropenia (21% vs 25%), and anemia<br />

(19% vs 19%). Conclusions: BOS demonstrated similar efficacy and<br />

acceptable safety in both older and younger pts across Ph� leukemia<br />

cohorts.<br />

CP2L CP3L ADV<br />

>65 y 65 y 65 y


6512 Poster Discussion Session (Board #4), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

BELA trial update: Bosutinib (BOS) versus imatinib (IM) in patients (pts)<br />

with newly diagnosed chronic phase chronic myeloid leukemia (CP CML)<br />

after 30 months <strong>of</strong> follow-up. Presenting Author: Carlo Gambacorti-<br />

Passerini, University <strong>of</strong> Milan-Bicocca, Monza, Italy<br />

Background: The BELA study compared the efficacy and safety <strong>of</strong> BOS (dual<br />

Src/Abl kinase inhibitor) with IM in newly diagnosed CP CML. Methods: 502<br />

pts with newly diagnosed CP CML were randomized to BOS 500 mg/d (n �<br />

250) or IM 400 mg/d (n � 252) and stratified by Sokal risk group and<br />

geographic region. Efficacy analyses included all randomized pts (ITT);<br />

safety analyses included all treated pts (BOS, n � 248; IM, n � 251). Data<br />

described below are for �24 mo <strong>of</strong> follow-up; updated data for �30 mo <strong>of</strong><br />

follow-up will be presented. Results: Median treatment duration was 27.5<br />

mo in both cohorts; 63% <strong>of</strong> BOS pts and 71% <strong>of</strong> IM pts were still receiving<br />

treatment. The primary reason for BOS discontinuation was a treatmentemergent<br />

adverse event (TEAE; 24% vs 7% with IM); the primary reason for<br />

IM discontinuation was disease progression (13% vs 4% with BOS).<br />

Cumulative complete cytogenetic response (CCyR) rates by 24 mo were<br />

79% for BOS and 80% for IM. Cumulative major molecular response<br />

(MMR) rates by 24 mo were 59% for BOS and 49% for IM (P � 0.019),<br />

including 16% and 12% <strong>of</strong> pts with complete molecular response (4.0-log<br />

sensitivity). On-treatment transformation to accelerated/blast phase occurred<br />

in 4 (2%) BOS pts and 13 (5%) IM pts. Deaths were reported for 7<br />

BOS pts (6 due to CML progression) and 13 IM pts (10 due to CML<br />

progression); 24-mo Kaplan-Meier overall survival estimates were 97%<br />

(BOS) and 95% (IM). BOS was associated with higher incidences <strong>of</strong><br />

gastrointestinal events than IM (diarrhea [70% vs 25%], vomiting [32% vs<br />

16%]; primarily transient), but lower incidences <strong>of</strong> edema (13% vs 40%)<br />

and musculoskeletal events (cramps [4% vs 22%], bone pain [4% vs<br />

10%]). Grade �3 TEAEs in �2% <strong>of</strong> BOS or IM pts were diarrhea (12% vs<br />

1%), vomiting (3% vs 0%), and rash (2% vs 1%). Grade �3 lab<br />

abnormalities (�15% <strong>of</strong> pts) with BOS and IM were neutropenia (10% vs<br />

24%), thrombocytopenia (14% vs 15%), elevated alanine aminotransferase<br />

(23% vs 4%), and hypophosphatemia (6% vs 20%). Conclusions: BOS<br />

was effective for newly diagnosed CP CML and had a distinct toxicity<br />

pr<strong>of</strong>ile. With continued follow-up both on-treatment transformation to<br />

accelerated/blast phase and overall survival continue to favor BOS versus<br />

IM.<br />

6514^ Poster Discussion Session (Board #6), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Reductions in JAK2V617F allele burden with ruxolitinib treatment in<br />

COMFORT-II, a phase III study comparing the safety and efficacy <strong>of</strong><br />

ruxolitinib to best available therapy (BAT). Presenting Author: Alessandro<br />

M. Vannucchi, University <strong>of</strong> Florence, Florence, Italy<br />

Background: Ruxolitinib is a potent and selective JAK1/2 inhibitor approved<br />

for the treatment <strong>of</strong> myel<strong>of</strong>ibrosis (MF) based on results <strong>of</strong> the phase 3<br />

COMFORT studies. Ruxolitinib demonstrated rapid and durable reductions<br />

in splenomegaly and improved MF-related symptoms and quality <strong>of</strong> life <strong>of</strong><br />

patients (pts) with MF. Since one measure <strong>of</strong> efficacy is molecular<br />

response, this analysis correlates changes in mutant allele burden (%V617F)<br />

with spleen size reduction in COMFORT-II. Methods: COMFORT-II is a<br />

randomized, open-label, phase 3 study comparing ruxolitinib 15 or 20 mg<br />

twice daily (BID) with BAT. The primary endpoint was a � 35% reduction in<br />

spleen volume from baseline (BL) at week 48. Change in %V617F was<br />

measured by allele specific qPCR. Pts were stratified by reduction in<br />

%V617F (� 10%, 10-20%, � 20%) and results were correlated with<br />

achievement <strong>of</strong> the primary endpoint. Results: More pts in the ruxolitinib<br />

arm had � 10% V617F reductions compared with BAT (41% vs 5%; P �<br />

.01; Table). The majority <strong>of</strong> reductions � 20% were gradual and progressive<br />

over the course <strong>of</strong> the study; 2 pts had rapid reductions from 48% to<br />

1% and 45% to 9% over 48 weeks. In the ruxolitinib arm, significantly<br />

more pts with a � 20% V617F reduction achieved the primary endpoint<br />

compared with pts with a � 10% reduction (79% vs 30%; P � .004); in<br />

each group, gender did not affect spleen response. For pts with � 10%<br />

reductions (15 mg BID, n � 16; 20 mg BID, n � 24), the average total daily<br />

dose (TDD) was ruxolitinib 29.6 mg; pts with � 20% reductions (15 mg<br />

BID, n � 3; 20 mg BID, n � 11) had a TDD <strong>of</strong> 35.3 mg. Conclusions: Pts<br />

who received ruxolitinib had larger reductions in JAK2V617F allele burden<br />

compared with BAT. %V617F reductions were gradual over the course <strong>of</strong><br />

the 48-week study; longer follow-up is needed to determine the extent <strong>of</strong><br />

allele burden reduction.<br />

Ruxolitinib<br />

(N � 145) a<br />

BAT<br />

(N � 69) a<br />

n(%)<br />

JAK2V617F � at BL 110 (76) 49 (71) b<br />

Median (range) %V617F<br />

JAK2V617F<br />

84.5 (5-96) 81 (1-95)<br />

� with week-48 data<br />

%V617F reduction<br />

68 (62) 22 (45)<br />

Median<br />

< 10%<br />

-7.0% 0%<br />

c<br />

40 (59) 21 (95)<br />

10-20% 14 (21) 1 (5)<br />

> 20% 14 (21) 0<br />

a b c<br />

Pts with %V617F data. Unknown JAK-mutation status, n � 4. Includes pts with increased<br />

%V617F (range, 0-7%).<br />

Leukemia, Myelodysplasia, and Transplantation<br />

419s<br />

6513 Poster Discussion Session (Board #5), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Subcutaneous omacetaxine mepesuccinate in patients with chronic phase<br />

(CP) or accelerated phase (AP) chronic myeloid leukemia (CML) resistant/<br />

intolerant to two or three approved tyrosine-kinase inhibitors (TKIs).<br />

Presenting Author: Franck E. Nicolini, Centre Hospitalier Lyon Sud, Pierre<br />

Bénite, France<br />

Background: Omacetaxine mepesuccinate (“omacetaxine”) is a first-inclass,<br />

reversible, transient inhibitor <strong>of</strong> protein elongation that facilitates<br />

tumor cell death without depending on BCR-ABL signaling. <strong>Clinical</strong> activity<br />

was shown in two phase 2, open-label, multicenter studies in treatmentresistant<br />

CML. Methods: A subset from the phase 2 studies included<br />

patients in CP or AP who were resistant/intolerant to �2 approved TKIs.<br />

Omacetaxine 1.25 mg/m2 was given SC twice daily: �14 consecutive<br />

days/28-day cycle for induction, �7 days/cycle as maintenance. Secondary<br />

results were calculated for responses by number <strong>of</strong> prior TKIs. Results: Of<br />

122 patients (median age, 60 years), 62 had received 2 TKIs (100%<br />

imatinib; 76% dasatinib; 24% nilotinib) and 60 had received all 3<br />

approved TKIs. Prior non-TKI therapies (eg, 52% hydroxyurea, 34%<br />

interferon) were common. Of 45 CP patients in the 2 TKI group, 12 (27%)<br />

had major cytogenetic response (MCyR; median duration <strong>of</strong> 17.7 mo), and<br />

<strong>of</strong> 36 in the 3 TKI group, 4 (11%) had MCyR (median duration not<br />

reached). Of 17 AP patients in the 2 TKI group, 6 (35%) had major<br />

hematologic response (MaHR; median duration, 13.4 mo), and <strong>of</strong> 24 in the<br />

3 TKI group, 5 (21%) had MaHR (median duration, 6.4 mo). Median<br />

months <strong>of</strong> survival in the 2 and 3 TKI groups were 30.1 and not reached for<br />

CP and 12.0 and 24.6 for AP. Treatment-related grade 3/4 adverse events<br />

(AEs) occurred in 52 (84%) patients in the 2 TKI group and 42 (70%) in<br />

the 3 TKI group (most common: thrombocytopenia [71%, 48%]). Fifteen<br />

(24%) and 16 (27%) had an AE leading to discontinuation, primarily<br />

disease progression. There were 20 deaths (primarily disease progression),<br />

11 in the 2 TKI group and 9 in the 3 TKI group; 4 were considered related to<br />

treatment (sepsis in 2, pancytopenia, febrile neutropenia). Conclusions: In<br />

patients with heavily pretreated CML, response to omacetaxine therapy<br />

occurred whether they had received 2 or 3 prior TKIs. The drug was well<br />

tolerated. Tolerability was similar across the TKI groups in both CP and AP<br />

patients. Support: Teva Pharmaceutical Industries Ltd.<br />

6515^ Poster Discussion Session (Board #7), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Combination <strong>of</strong> the Bruton’s tyrosine kinase (BTK) inhibitor PCI-32765<br />

with bendamustine (B)/rituximab (R) (BR) in patients (pts) with relapsed/<br />

refractory (R/R) chronic lymphocytic leukemia (CLL): Interim results <strong>of</strong> a<br />

phase Ib/II study. Presenting Author: Susan Mary O’Brien, University <strong>of</strong><br />

Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: BTK is an essential mediator <strong>of</strong> B cell receptor signaling and a<br />

critical kinase for lymphoma cell survival. PCI-32765 (P), an oral,<br />

selective, irreversible inhibitor <strong>of</strong> BTK, inhibits proliferation, migration and<br />

adhesion in CLL cells, and is highly active as a single agent for the<br />

treatment <strong>of</strong> R/R CLL pts. (O’Brien ASH 2011). BR produces an overall<br />

response rate (ORR) <strong>of</strong> 59% in R/R CLL (Fischer JCO 2011). We report<br />

interim data on P combined with BR. Methods: R/R CLL pts received P 420<br />

mg orally daily for 28-day (D) cycles (C) until disease progression (PD). B<br />

was administered 70 mg/m2 on D1 and D2 combined with R 375 mg/m2 on<br />

D0 for C1 and 500 mg/m2 on D1 for subsequent courses for a maximum <strong>of</strong><br />

6 cycles. Response was evaluated according to IWCLL criteria. Results: 30<br />

pts were enrolled. Median age <strong>of</strong> pts was 62 yrs (range 41-82). 46% <strong>of</strong> pts<br />

were Rai stage III/IV and the median # <strong>of</strong> prior therapies was 2 (range 1-4).<br />

37% and 13% were considered refractory (treatment free interval �12 mo)<br />

to a purine analog containing regimen or BR, respectively. Bulky disease<br />

was present in 52%. Adverse events (AE) have been consistent with that<br />

expected with BR. Gr 3/4 neutropenia and thrombocytopenia have been<br />

noted in 47% and 10% <strong>of</strong> pts, respectively. Grade �3 non-hematologic<br />

AEs potentially related to P included rash (3 pts) and fatigue and tumor<br />

lysis reported in 2 pts each. There were no Gr 3/4 infusion reactions. There<br />

have been no discontinuations (D/C) due to AE and no deaths on study. At a<br />

median follow-up <strong>of</strong> 4.9 mos (range 2.7-8.3 mo) 16 pts have completed BR<br />

and 14 pts are still receiving BR. The ORR is 90% (27/30 pts) (CR 10%,<br />

PR 80%). 2 additional pts achieved a nodal response with residual<br />

lymphocytosis. Responses appear independent <strong>of</strong> high-risk clinical or<br />

genomic features. 90% <strong>of</strong> pts remain on study; reasons for D/C include PD<br />

(n�2) and 1 pt pursuing SCT. Conclusions: PCI-32765, in combination<br />

with BR, is highly active. The high ORR, low rate <strong>of</strong> PD, and good<br />

tolerability compares very favorably with historical controls, warranting<br />

additional investigation <strong>of</strong> this combination.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


420s Leukemia, Myelodysplasia, and Transplantation<br />

6516^ Poster Discussion Session (Board #8), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Phase II trial <strong>of</strong> the combination <strong>of</strong> <strong>of</strong>atumumab and lenalidomide in<br />

patients with relapsed chronic lymphocytic leukemia (CLL). Presenting<br />

Author: Lorenzo Falchi, University <strong>of</strong> Texas M. D. Anderson Cancer Center,<br />

Houston, TX<br />

Background: Based on the activity <strong>of</strong> <strong>of</strong>atumumab and lenalidomide as<br />

monotherapy, we evaluated efficacy and tolerability <strong>of</strong> the combination in<br />

pts with relapsed CLL. Methods: Thirty-six pts entered this phase II study.<br />

All pts had an indication for therapy, adequate liver/renal function and<br />

received prior fludarabine. Ofatumumab IV was given weekly for 4 weeks<br />

(300mg w 1; 1,000 mg w 2-4), monthly during months 2-6, and every<br />

other month during months 7-24. Lenalidomide 10 mg PO/day started on<br />

day 9 and continued for 24 months. Responses were assessed (2008 IWG<br />

criteria) at month 3, 6 and every 6 months. Results: Thirty-four pts are<br />

evaluable. Median age was 64 yrs (34-82), 59 % <strong>of</strong> the pts had Rai stage<br />

III-IV disease, median �-2M level was 4.1 mg/dL (1.7-16), 62 % <strong>of</strong> the pts<br />

had unmutated IgHV, 26% del(17p) and 12% del(11q). Median number <strong>of</strong><br />

prior treatments was 2 (1-8), all pts had been treated with FCR and 29%<br />

were fludarabine-refractory. The overall response (OR) rate is 65% (22 pts);<br />

7 pts (21%) achieved a complete response (CR) including 4 MRD-negative<br />

CR, and 15 pts (44%) achieved a partial response (PR). Twelve pts are still<br />

on therapy. Median duration <strong>of</strong> response has not been reached with a<br />

median follow up <strong>of</strong> 13 months. Seven pts discontinued therapy despite<br />

ongoing response: HSCT (3 pts), toxicity (2 pts) and physician choice (2<br />

pts); 3 pts discontinued therapy because <strong>of</strong> loss <strong>of</strong> response (at 12, 16 and<br />

16 months). Eighty-two % <strong>of</strong> the pts are alive. No deaths occurred on<br />

therapy, 6 deaths occurred after therapy discontinuation: CLL (4), HSCT<br />

(1) and CLL/lung cancer (1). Lenalidomide daily dose was 10 mg (9 pts),<br />

7.5 mg (4 pts), 5 mg (13 pts), �5 mg (8 pts). G3-4 toxicities included<br />

neutropenia (16 pts, 47%), thrombocytopenia (3 pts, 9%) and anemia (2<br />

pts, 6%). One pt (3%) experienced G4 pulmonary embolism while on ESAs.<br />

One pt (3%) had G3 infusion reaction to <strong>of</strong>atumumab. Fourteen G3<br />

infections occurred: pneumonia (4), fever/bacteremia (5), parotitis (1),<br />

cellulitis (2), HZV (1), and CNS toxoplasmosis (1). G1-2 tumor flare<br />

reaction occurred in 8 pts (24%). Conclusions: The combination <strong>of</strong><br />

<strong>of</strong>atumumab and lenalidomide was well tolerated. It induced durable<br />

responses in 65% <strong>of</strong> FCR pre-treated pts with relapsed CLL.<br />

6518^ Poster Discussion Session (Board #10), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

A phase I/II study <strong>of</strong> the selective phosphatidylinositol 3-kinase-delta<br />

(PI3K�) inhibitor, GS-1101 (CAL-101), with <strong>of</strong>atumumab in patients with<br />

previously treated chronic lymphocytic leukemia (CLL). Presenting Author:<br />

Richard R. Furman, Weill Cornell Medical College, New York, NY<br />

Background: PI3K� is expressed in cells <strong>of</strong> hematopoietic origin where it<br />

regulates the survival and proliferation <strong>of</strong> malignant B-cells. GS-1101 is an<br />

orally bioavailable, small-molecule inhibitor that selectively targets PI3K�<br />

and is highly active in patients with hematologic malignancies. Methods:<br />

This Phase 1/2 study evaluated repeated 28-day cycles <strong>of</strong> GS-1101 in<br />

combination with <strong>of</strong>atumumab. GS-1101 (150mg BID) was co-administered<br />

with a total <strong>of</strong> 12 infusions <strong>of</strong> <strong>of</strong>atumumab over 24 weeks (300mg<br />

initial dose either on Day 1 or Day 2 (relative to the first dose <strong>of</strong> GS-1101),<br />

followed 1 week later by 1,000mg weekly for 7 doses, followed 4 weeks<br />

later by 1,000mg every 4 weeks for 4 doses). Thereafter, each subjects<br />

received single-agent GS-1101 as long as the subject was benefitting.<br />

Results: Accrual is complete with 21 subjects enrolled and 11 evaluable.<br />

Six subjects started <strong>of</strong>atumumab treatment on Day 1 and 5 on Day 2.<br />

Median [range] age was 63 [54-76] years. The majority (9/11; 82%) <strong>of</strong><br />

patients had bulky adenopathy. The median [range] number <strong>of</strong> prior<br />

therapies was 3 [1-6], including prior exposure to alkylating agents (10/11;<br />

90%), rituximab (9/11; 82%), purine analogs (8/11; 72%), alemtuzumab<br />

(3/11; 28%) and/or <strong>of</strong>atumumab (2/11;18%). At the data cut<strong>of</strong>f, the<br />

median [range] treatment duration was 5 [0-7] cycles. Almost all subjects<br />

(9/11;82%) experienced marked and rapid reductions in lymphadenopathy<br />

within the first 2 cycles. The lymphocyte mobilization that is expected with<br />

PI3K� inhibition was significantly reduced in magnitude and duration and<br />

persisted past Cycle 1 in only 1 patient. Early follow up data support a<br />

favorable safety pr<strong>of</strong>ile and confirm a lack <strong>of</strong> clinically significant myelosuppression.<br />

Elevated baseline levels <strong>of</strong> CCL3, CCL4, CXCL13, and TNFa were<br />

significantly reduced after 28 days <strong>of</strong> treatment. Conclusions: GS-1101/<br />

<strong>of</strong>atumumab <strong>of</strong>fers a well-tolerated noncytotoxic combination regimen with<br />

substantial activity in previously treated patient with bulky adenopathy.<br />

Data on the complete cohort <strong>of</strong> 21 subjects will be presented.<br />

6517 Poster Discussion Session (Board #9), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Pretreatment prognostic factors associated with outcomes for first-line<br />

FCR-based treatments in previously untreated CLL. Presenting Author:<br />

William G. Wierda, University <strong>of</strong> Texas M. D. Anderson Cancer Center,<br />

Houston, TX<br />

Background: First-line chemoimmunotherapy with fludarabine, cyclophosphamide,<br />

and rituximab (FCR) demonstrated improved outcomes, including<br />

survival, for fit patients (pts) with CLL. Modifications <strong>of</strong> this regimen,<br />

including intensified rituximab (FCR3), addition <strong>of</strong> mitoxantrone (FCMR) or<br />

addition <strong>of</strong> alemtuzumab fir high-risk CLL (CFAR), were evaluated but did<br />

not improve outcomes in historic comparisons. Methods: We correlated<br />

outcomes, including complete remission (CR), time-to-treatment failure<br />

(TTF) and overall survival (OS), with new and traditional pretreatment<br />

prognostic factors to identify high-risk pts. Results: All pts (N�473) had an<br />

NCI-WG indication for treatment and received a first-line FCR-based<br />

regimen on trial; the intended treatment was 6 courses. Patient characteristics<br />

correlated with outcomes are presented in the table. Factors not<br />

associated with outcomes included absolute lymphocyte count; platelet<br />

count; performance status; spleen size; liver size; and number <strong>of</strong> involved<br />

lymph node sites. Conclusions: We identified the following as high-risk<br />

pretreatment features for patients going on first-line FCR-based therapy:<br />

advanced age, presence <strong>of</strong> 17p del, high B2M (�4mg/l), and unmutated<br />

IGHV gene. Pts with these features should be pursued with new treatment<br />

modalities and novel agents in order to improve outcomes.<br />

Pretreatment<br />

characteristic Higher CR Longer TTF Longer OS<br />

Younger age UVA UVA UVA; MVA<br />

Female - UVA; MVA -<br />

Earlier Rai stage UVA; MVA - UVA<br />

Lower ß-2 microglobulin (


6520 Poster Discussion Session (Board #12), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

A randomized phase II study <strong>of</strong> sapacitabine in MDS refractory to<br />

hypomethylating agents. Presenting Author: Guillermo Garcia-Manero,<br />

University <strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: Sapacitabine is an orally administered nucleoside analogue<br />

which causes single-strand DNA breaks and induces G2 cell cycle arrest. A<br />

multi-center, randomized phase 2 study was conducted to evaluate 3 dose<br />

regimens in older patients with MDS refractory to hypomethylating agents.<br />

The primary endpoint is 1-year survival. Secondary endpoints include the<br />

rate <strong>of</strong> CR, CRp, PR, major hematological improvement (HI) or stable<br />

disease and their corresponding durations. The study uses a selection<br />

design to identify a dose regimen which produces a better 1-year survival<br />

rate in the event that all three dose regimens are active. The planned<br />

sample size is 60 patients (20 patients in each arm). Methods: Eligible<br />

patients must be � 60 years with intermediate-2 or higher risk MDS<br />

previously treated with hypomethylating agents and 6 - � 20% blasts in<br />

bone marrow, ECOG 0-2, adequate renal and hepatic functions. Patients<br />

were randomized 1:1:1 to receive sapacitabine every 4 weeks at 200 mg<br />

b.i.d. x 7 days (Arm G), 300 mg q.d. x 7 days (Arm H), or 100 mg q.d. x 5<br />

days per week x 2 weeks (Arm I). The number <strong>of</strong> cycles is not limited.<br />

Results: As <strong>of</strong> January 2012, 61 patients were enrolled and 56 had � 30<br />

days <strong>of</strong> follow-up. Mean follow-up was 173 days. Median age was 71. Two<br />

or 3 cytopenias were present in 42 patients and 14 have received both<br />

azacitidine and decitabine. Baseline bone marrow had 6-10% blasts in 31<br />

patients and 11-19% blasts in 25 patients. Median number <strong>of</strong> cycles was 2<br />

and 18 patients received � 4 cycles. To date, 8 patients have responded (2<br />

CRs, 2 CRp, and 4 major HIs): 15% (Arm G), 16% (Arm H) and 12% (Arm<br />

I). Time to response is 1-3 cycles. Additionally, 6 patients achieved stable<br />

disease lasting longer than 16 weeks. More than 50% <strong>of</strong> patients have lived<br />

16 weeks or longer. Three deaths occurred within 30 days <strong>of</strong> randomization<br />

and 1 death was considered to be related to sapacitabine. Common adverse<br />

events (all grades, regardless <strong>of</strong> causality) included fatigue, nausea,<br />

diarrhea, constipation, edema, dyspnea, pyrexia, pneumonia, febrile neutropenia,<br />

anemia, neutropenia and thrombocytopenia, most <strong>of</strong> which were<br />

mild to moderate. Conclusions: Sapacitabine appears to be safe and active<br />

across all 3 dose regimens. Updated data will be presented at the meeting.<br />

6522 Poster Discussion Session (Board #14), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Efficacy and tolerability <strong>of</strong> lenalidomide (LEN) in patients (pts) 75 and older<br />

versus those younger than 75 with RBC transfusion-dependent low/int-1-risk<br />

MDS and del 5q. Presenting Author: Pierre Fenaux, Hopital Avicenne, Bobigny,<br />

France<br />

Background: LEN is the approved treatment for pts with RBC transfusiondependent<br />

Low/Int-1-risk MDS and del 5q. In 2 multicenter trials (MDS-003/-<br />

004) LEN lead to RBC transfusion independence (TI) for � 26 wks in 35–58% <strong>of</strong><br />

pts and cytogenetic response (CyR) in 25–73%. Most common grade (G) 3–4<br />

adverse events (AE) were neutropenia and thrombocytopenia, a safety concern in<br />

elderly pts. We evaluated efficacy and tolerability <strong>of</strong> LEN in pts � 75yvs�75 y<br />

in MDS-003/-004 trials. Methods: Pts received LEN 5 mg � 28 d, 10 mg � 21 d,<br />

or 10 mg � 28 d (all 28 d cycles). Dose reductions were required for G4<br />

neutropenia (both trials) and platelet counts � 30x109 /L (MDS-003) or � 25 x<br />

109 /L (MDS-004). Results: 32% <strong>of</strong> the 286 pts were � 75 y. Baseline (BL)<br />

characteristics, LEN treatment, and optional G-CSF use are shown in Table. In<br />

pts � 75yvs�75 y: RBC-TI � 26 wks was 39 vs 47% (P � NS); CyR was 64 vs<br />

54% (P � NS); 2-y AML progression rates were 10 vs 19% (P � NS); 2-y overall<br />

survival rates were 62 vs 73% (P � .001); G3–4 AE: neutropenia (63 vs 76%; P<br />

� .024), thrombocytopenia (56 vs 49%; P � NS), infection (36 vs 20%; P �<br />

.003); median time to recovery to ANC � 1x109 /L was 0.7 vs 0.7 mo (P � NS)<br />

and to platelet counts � 100x109 /L was 3.3 vs 1.7 mo (P � NS). 59% pts � 75<br />

y and 49% pts � 75 y discontinued LEN due to AE (33 vs 26%; P � NS), lack <strong>of</strong><br />

effect (32 vs 49%; P � NS), or death (15 vs 6%; P � NS). Dose reduction and<br />

discontinuation rates are shown in Table. Conclusions: Pts � 75 y and � 75 y<br />

had comparable response and AML progression rates. In pts � 75 y, LEN<br />

appears to be well tolerated but the higher infection rate justifies close follow-up.<br />

< 75 y > 75 y P value<br />

BL characteristics<br />

Female, %<br />

Median time from diagnosis, y<br />

Median RBC transfusion burden, units/8 wks<br />

Neutropenia (< 1x10<br />

65<br />

3<br />

6<br />

79<br />

3<br />

6<br />

.019<br />

NS<br />

NS<br />

9 /L), %<br />

Thrombocytopenia (< 100x10<br />

11 8 NS<br />

9 /L), %<br />

del 5q, %<br />

Isolated<br />

Plus > 1 abnormality<br />

IPSS risk, %<br />

Low<br />

Int-1<br />

ECOG, %<br />

0<br />

1/2<br />

LEN treatment<br />

Median duration, mo<br />

Median total dose per cycle, mg<br />

Median % <strong>of</strong> assigned dose cycle 1–4<br />

Reason for dose reduction, %<br />

Neutropenia<br />

Thrombocytopenia<br />

Reason for discontinuation, %<br />

Neutropenia<br />

Thrombocytopenia<br />

Optional G-CSF use<br />

Pts treated, %<br />

14<br />

71<br />

24<br />

27<br />

45<br />

25<br />

26<br />

14<br />

126<br />

68<br />

33<br />

17<br />

2<br />

4<br />

37<br />

13<br />

67<br />

31<br />

40<br />

37<br />

12<br />

42<br />

9<br />

108<br />

56<br />

25<br />

30<br />

3<br />

3<br />

30<br />

NS<br />

NS<br />

NS<br />

.003<br />

.009<br />

NS<br />

NS<br />

NS<br />

.019<br />

NS<br />

NS<br />

NS<br />

Leukemia, Myelodysplasia, and Transplantation<br />

421s<br />

6521 Poster Discussion Session (Board #13), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Use <strong>of</strong> single polymorphism arrays (SNP-A) to modify prognostic scoring<br />

systems for myelodysplastic syndromes. Presenting Author: Manojkumar<br />

Bupathi, Cleveland Clinic Foundation/MetroHealth Medical Center-<br />

Department <strong>of</strong> Translational Hematology and Oncology Research, Cleveland,<br />

OH<br />

Background: MDS are hematologic neoplasms characterized by dysplasia<br />

and cytopenias. Two commonly used risk stratifications, IPSS (Greenberg<br />

et al Blood 1997) and IPSS-R are based on clinical factors (marrow blasts,<br />

number <strong>of</strong> cytopenias and genetic abnormalities determined by conventional<br />

karyotyping), with the IPSS based on 4 cytogenetic risk groups and<br />

the IPSS-R based on 5 cytogenetic risk groups defined by Schanz et al JCO<br />

2012. A drawback to metaphase cytogenetics (MC) is its inability to detect<br />

small cryptic chromosomal defects and uniparental disomy (UPD). SNP-A<br />

can identify these small cytogenetic lesions/UPD and has been shown to be<br />

<strong>of</strong> prognostic value in MDS. The goal <strong>of</strong> this investigation was to determine<br />

if SNP-A detected defects could be used to refine the IPSS and IPSS-R.<br />

Methods: SNP-A karyotyping was performed as previously described (Tiu et<br />

al, Blood, 2011). We reviewed data from 327 patients idenfitying SNP-A<br />

results, MC, and the IPSS/IPSS-R-related clinical factors were identified.<br />

Overall survival (OS) measured from the date <strong>of</strong> sampling for SNP-A<br />

karyotyping was the primary endpoint. Data were analyzed using Cox<br />

proportional hazards models. Results: SNP-A lesions not detected by MC<br />

were grouped as 1) none or only gains/losses; 2) UPD only or gains�losses<br />

but no UPD; and 3) UPD�other defects. The frequencies/median os in<br />

months (mo) <strong>of</strong> the groups were: 66%/20.0, 23%/12.7 and 11%/5.8,<br />

respectively, p�.0001 for OS. Combining the SNP-A groups with MC resulted<br />

in revised definitions <strong>of</strong> cytogenetic risk that had better discrimination than<br />

the underlying models; and when combined with the relevant clinical<br />

factors resulted in refined IPSS (IPSSSNP-A ) and IPSS-R (IPSS-RSNP-A ) risk<br />

stratifications (Table). Conclusions: Detection <strong>of</strong> chromosomal gains, losses,<br />

and UPD by SNP-A has prognostic value in MDS and can be used to refine<br />

current risk stratifications.<br />

Median os in mo (% <strong>of</strong> pts) for each model.<br />

Risk groups<br />

Model 1 2 3 4 5<br />

IPSS 39.1 (21%) 20.0 (37%) 8.9 (21%) 4.6 (21%) ---<br />

IPSSSNP-A 47.0 (15%) 27.1 (30%) 15.3 (26%) 7.2 (18%) 2.5 (12%)<br />

IPSS-R 39.1 (9%) 30.4 (32%) 17.6 (12%) 16.7 (14%) 4.6 (28%)<br />

IPSS-R SNP-A 41.8 (23%) 27.7 (14%) 15.1 (34%) 6.6 (17%) 2.6 (12%)<br />

6523 Poster Discussion Session (Board #15), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Results <strong>of</strong> a phase II trial <strong>of</strong> azacitidine (AZA) with or without epoetin beta<br />

(EPO) in lower-risk MDS. Presenting Author: Claude Gardin, Hematology,<br />

Hopital Avicenne APHP, University <strong>of</strong> Paris 13, Bobigny, France<br />

Background: Although anemia <strong>of</strong> IPSS low and int-1 (LR) MDS initially<br />

responds to erythroid stimulating agents (ESA) in 40-50% <strong>of</strong> patients (pts),<br />

response is generally transient. Anemia recurrence with RBC cell transfusion<br />

dependency (RBC-TD) is an indicator <strong>of</strong> poor prognosis, even in<br />

absence <strong>of</strong> progression to higher risk MDS. AZA leads to RBC transfusion<br />

independence (RBC-TI) in 30–40% <strong>of</strong> LR-MDS (Lyons, JCO, 2009), but it<br />

has not been prospectively tested in LR-MDS patients resistant to ESA. It<br />

also remains unknown if the addition <strong>of</strong> ESA to AZA would be useful in such<br />

pts. Methods: In this randomized phase-II trial (GFMAzaEpo-2008-1 trial,<br />

NCT01015352), the Groupe Francophone des Myélodysplasies (GFM)<br />

compared AZA 75mg/m2/d for 5 days every 28 days for 6 cycles (AZA arm)<br />

to the same treatment plus EPO 60000U/week (AZA�EPO arm). Inclusion<br />

criteria were LR-MDS resistant to at least 12 weeks <strong>of</strong> ESA, with RBC-TD �<br />

4 RBC units in the previous 8 weeks. Responders in both arms were eligible<br />

for maintenance up to 12 monthly cycles, unless progression or loss <strong>of</strong><br />

erythroid response occurred. The primary endpoint was RBC-TI (HI-E major<br />

using IWG 2000 criteria) after 6 courses. Results: Between 2009 and<br />

2010, the 98 planned pts were included: M/F 65/28; median age 71y<br />

(41-84); 5 pts were excluded (one consent withdrawal, 2 early unrelated<br />

events and 2 with exclusion criteria). In the remaining 93 pts, IPSS risk was<br />

low in 69 and int-1 in 23 pts, with no imbalance between arms. Five pts<br />

received � 4 cycles and 16 received only 4 or 5 cycles, mainly due to<br />

toxicity or progression. RBC-TI after 6 courses was observed in 16.7%<br />

(8/48) in the AZA arm and 18 % (8/45) in the AZA�EPO arm (P�1), and in<br />

19.5% (8/41) and 26 % (8/31) respectively, in the 72 pts who received �<br />

6 cycles (P�.57). Overall best response rate (at least HI-E minor using IWG<br />

2000 criteria) was 35 and 33% in the AZA and AZA�EPO arms,<br />

respectively (P�0.99). Of note, only 9 pts in the AZA�EPO arm required at<br />

least one hospitalization for a SAE, compared to 22 pts in the AZA arm<br />

(P�0.015). Conclusions: Erythroid response to AZA, in LR MDS with<br />

demonstrated ESA resistance, appears lower than expected, with no<br />

improved outcome when combined with an ESA. The latter may however<br />

improve tolerance <strong>of</strong> AZA in LR MDS.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


422s Leukemia, Myelodysplasia, and Transplantation<br />

6524^ Poster Discussion Session (Board #16), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Evaluation <strong>of</strong> the first-in-class antimitochondrial metabolism agent CPI-<br />

613 in hematologic malignancies. Presenting Author: Timothy S. Pardee,<br />

Wake Forest University Health Sciences, Winston-Salem, NC<br />

Background: Altered metabolism is a hallmark <strong>of</strong> cancer, including hematologic<br />

malignancies. Most tumor cells use glycolysis even under aerobic<br />

conditions (the Warburg effect). This altered metabolism is a possible<br />

therapeutic target. The novel lipoate derivative CPI-613 is a first-in-class<br />

agent that targets a key mitochondrial enzyme, pyruvate dehydrogenase<br />

complex (PDH). Methods: CPI-613 was tested against leukemia cell lines in<br />

vitro and in vivo. It is also the subject <strong>of</strong> a phase I clinical trial for patients<br />

with hematologic malignancies. Results: CPI-613 was active against HL60,<br />

Jurkat, and K562 cells, with an average IC50 value <strong>of</strong> 14 �M (range 12.2 –<br />

16.4). CPI-613 was synergistic with doxorubicin, with Combinatorial Index<br />

(CI) values <strong>of</strong> 0.478 to 0.765. CPI-613 sensitivity increased with knockdown<br />

<strong>of</strong> p53 or MN1 expression, despite their increased resistance to<br />

standard therapy. CPI-613 was synergistic with nilotinib against BCR-ABLexpressing<br />

cells and with sorafenib against Flt3 ITD-expressing cells. CI<br />

values for nilotinib � CPI was 0.059 (�/-0.002) and for sorafenib � CPI<br />

was 0.581 (�/-0.052). In vivo, CPI-613 synergized with doxorubicin;<br />

median survival improved from 12 days with doxorubicin to 16 days with<br />

CPI-613 plus doxorubicin (p�0.0001). In the phase I clinical trial, 7 <strong>of</strong> 13<br />

evaluable patients had a response <strong>of</strong> stable disease or better, for an overall<br />

response rate <strong>of</strong> 54% (see table). CPI-613 was well tolerated, with no<br />

evidence <strong>of</strong> marrow suppression and no dose limiting toxicity identified.<br />

Conclusions: The novel agent CPI-613 has activity against a variety <strong>of</strong><br />

hematological malignancies, including acute leukemias. The therapeutic<br />

index appears high, with only minor toxicities observed.<br />

Patient # Diagnosis Dose CPI-613 mg/m 2<br />

Best response # <strong>of</strong> cycles<br />

1 NHL 420 NE 0<br />

2 AML 420 PD 1<br />

3 Myeloma 840 PD 1<br />

4 AML 840, 1386, 2100 CR 15<br />

5 Hodgkin’s 840 PD 1<br />

6 AML 1386 PD 0<br />

7 Myeloma 1386 SD 3<br />

8 Myeloma 1386 SD 4<br />

9 AML 1386 PD 1<br />

10 MDS 2100 SD 7<br />

11 MDS 2100 SD 6<br />

12 AML 2100 MLFS 2<br />

13 AML 2940 NE 0<br />

14 Burkitt’s 2940 SD 4<br />

15 AML 2940 PD 1<br />

SD� stable disease; CR�complete remission; MLFS�morphologically leukemia-free state;<br />

PD�progressive disease; NE�not evaluable.<br />

6526 Poster Discussion Session (Board #18), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

A phase II study <strong>of</strong> bortezomib added to standard daunorubicin and<br />

cytarabine during induction therapy and to intermediate-dose cytarabine<br />

(Int-DAC) for consolidation in patients with previously untreated acute<br />

myeloid leukemia (AML) age 60-75 years: CALGB study 10502. Presenting<br />

Author: Eyal C. Attar, Massachusetts General Hospital, Boston, MA<br />

Background: We conducted a clinical trial to determine the complete<br />

remission (CR) rate in older AML patients treated with bortezomib (Bz) plus<br />

daunorubicin and cytarabine induction therapy and the maximal tolerated<br />

dose (MTD) <strong>of</strong> Bz when added to Int-DAC consolidation therapy. Methods:<br />

Ninety-five adults (60-75 yrs old; median, 67) with previously untreated<br />

AML (including therapy-related and prior MDS) received Bz 1.3 mg/m2 IV<br />

on days 1, 4, 8 and 11 with daunorubicin 60 mg/m2 on days 1-3 and<br />

cytarabine 100 mg/m2 by continuous IV infusion on days 1-7. Patients who<br />

achieved a CR received 2 courses <strong>of</strong> consolidation chemotherapy with<br />

cytarabine 2 gm/m2 over 3 hours on days 1-5 (Int-DAC) with Bz administered<br />

on days 1, 4, 8 and 11. Three cohorts with escalating dose levels <strong>of</strong><br />

Bz were tested (0.7, 1.0, and 1.3 mg/m2 ). Dose limiting toxicities were<br />

assessed during the first cycle <strong>of</strong> consolidation. Predictors <strong>of</strong> response,<br />

including CD74 expression, were assessed. Results: The CR frequency was<br />

65% (62/95). An additional 4% (4/95) achieved CR with incomplete<br />

platelet recovery. The CR rate according to ELN Genetic Groups was 90%<br />

(9/10) for favorable, 71% (12/17) for Int-I, 52% (17/33) for Int-II, and<br />

46% (5/11) for adverse. Six patients had grade 3 sensory neuropathy.<br />

There were 6 treatment-related deaths during cycle 1 <strong>of</strong> induction and 2<br />

during cycle 2. Bz plus Int-DAC proved tolerable at the highest dose tested.<br />

The median disease-free (DFS) and overall survivals (OS) for patients on<br />

this study were 8.2 and 12.1 months, respectively. Lower CD74 expression<br />

was associated with CR/CRp (p�.04) but not with DFS or OS. Conclusions:<br />

The addition <strong>of</strong> Bz 1.3 mg/m2 IV on days 1, 4, 8, and 11 to standard “3 �<br />

7” daunorubicin and cytarabine induction chemotherapy for AML is well<br />

tolerated and results in a remission rate at least as high as would be<br />

expected with ”3�7” alone in older patients. The maximum tested dose <strong>of</strong><br />

Bz given in combination with Int-DAC for remission consolidation was 1.3<br />

mg/m 2<br />

and proved to be tolerable. Further testing <strong>of</strong> this regimen in a phase<br />

III study is planned.<br />

6525 Poster Discussion Session (Board #17), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Initial salvage therapy in first relapse AML: A phase IIb study <strong>of</strong> CPX-351<br />

versus investigator’s choice—A subset analysis by prognostic group.<br />

Presenting Author: Stuart Goldberg, John Theurer Cancer Center, Hackensack,<br />

NJ<br />

Background: CPX-351 encapsulates cytarabine and daunorubicin within<br />

liposomes at a 5:1 molar ratio, shown in vitro to maximize synergy.<br />

Liposomal encapsulation enables preferential uptake <strong>of</strong> drug within leukemic<br />

blasts followed by intracellular release enhancing efficacy in AML. A<br />

randomized Phase 2b study in 1st relapse AML completed 1-year follow up<br />

with improvement in event free (EFS) and overall survival (OS). This report<br />

describes patient outcomes with a focus on the unfavorable subset per the<br />

European Prognostic Index (EPI) (Breems DA, et al. J Clin Oncol, 2005 Mar<br />

20;23:1969-1978). Methods: Patients (pts) �65yo in 1st relapse AML<br />

after an initial CR �1 month, ECOG PS� 0-2, serum creatinine� 2.0<br />

mg/dL, total bilirubin � 2.0 mg/dL, ALT/AST � 3 x ULN, and LVEF �50%<br />

were eligible. Pts with APL, daunorubicin exposure �368 mg/m2 (or other<br />

anthracycline equivalent), active CNS leukemia, uncontrolled infections<br />

were excluded. Pts were randomized 2:1 to receive up to 2 inductions and 2<br />

consolidation courses <strong>of</strong> CPX-351 (100 u/m2 ; D 1, 3, 5) or investigators<br />

choice <strong>of</strong> intensive salvage treatment. Post remission allo-transplantation<br />

was permitted. 126 pts were stratified by EPI into favorable, intermediate,<br />

and unfavorable groups (with projected 1-year OS <strong>of</strong> 70%, 49%, and 16%).<br />

The primary efficacy endpoint was % survival at 1-year. Results: CPX-351<br />

increased the rate <strong>of</strong> morphologic leukemia-free state (77% vs. 60%), the<br />

rate <strong>of</strong> CR � CRi (51% vs. 41%), and the median EFS (4.0 vs. 1.4 mo) and<br />

OS (8.5 vs. 6.3 mo) in the overall patient population. Significant survival<br />

improvement occurred amongst those with unfavorable EPI scores<br />

(HR�0.55, p-value�0.02) with improved 1-year survival (29% vs. 10%).<br />

CPX-351 treatment resulted in more prolonged cytopenias with increased<br />

febrile neutropenia and grade 3-4 infections, but not in the D 60 mortality<br />

(overall: 15% vs. 16%; unfavorable: 16% vs. 24%). Conclusions: CPX-351<br />

is highly active in AML and induces remission even in patients with prior<br />

cytarabine and anthracycline exposure. Efficacy was evident in every<br />

prognostic group, particularly in the unfavorable EPI group where a<br />

statistically significant improvement in 1-year survival was observed.<br />

6527 Poster Discussion Session (Board #19), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Stage I findings <strong>of</strong> a two-stage phase II study to assess the efficacy, safety,<br />

and tolerability <strong>of</strong> barasertib (AZD1152) compared with low-dose cytosine<br />

arabinoside (LDAC) in elderly patients (pts) with acute myeloid leukemia<br />

(AML). Presenting Author: Giovanni Martinelli, Department <strong>of</strong> Hematology/<br />

Oncology Seràgnoli, Bologna, Italy<br />

Background: Barasertib is the pro-drug to barasertib-hQPA, a selective<br />

Aurora B kinase inhibitor with preliminary anti-AML activity in a Phase I/II<br />

study (Löwenberg et al. Blood 2011;118:6030). Methods: AML pts aged<br />

�60 y considered unsuitable for intensive chemotherapy were randomized<br />

2:1 to open-label barasertib 1200 mg (7-day iv infusion) or LDAC 20 mg<br />

(sc twice daily for 10 days) in 28-day cycles (NCT00952588). The primary<br />

endpoint was improved objective complete response rate (OCRR: CR � CRi<br />

[Cheson criteria, but requiring CRi confirmation �21 days after first<br />

appearance, with partial recovery <strong>of</strong> platelets and neutrophils]). Secondary<br />

endpoints included duration <strong>of</strong> response (DoR), overall survival (OS) and<br />

safety. Results: 74 pts (barasertib, 48; LDAC, 26) received treatment and<br />

all completed �1 cycle. A significant improvement in OCRR was observed<br />

with barasertib (35.4% [17/48] vs 11.5% [3/26]; difference, 23.9% [95%<br />

CI, 2.7-39.9]; P�0.05); barasertib responses were seen in all cytogenetic<br />

risk groups and appeared to be durable (median DoR [range], 82 [28-321]<br />

days; vs LDAC 30-85 days). Although not formally sized to compare OS<br />

data, a trend favoring barasertib was observed (HR�0.88, 95% CI,<br />

0.49-1.58; P�0.663; median OS, 8.2 vs 4.5 mo). Stomatitis and febrile<br />

neutropenia were the most common adverse events (AEs) in the barasertib<br />

arm with higher incidences vs LDAC (71% vs 15%; 67% vs 19%,<br />

respectively). Grade �3 AEs with a greater incidence in the barasertib arm<br />

were febrile neutropenia (50% vs 19%), stomatitis (29% vs 0%) and<br />

pneumonia (25% vs 8%); grade �3 infection rates were also higher with<br />

barasertib (40% vs 23%). For both arms, there were similar discontinuation<br />

rates due to AEs (barasertib 8.3% vs LDAC 7.7%), deaths due to AEs<br />

(12.5% vs 11.5%) and 30-day mortality (12.5% vs 15.4%). Cumulative<br />

toxicities were not observed with barasertib treatment. Conclusions: In this<br />

population with poor prognosis using standard chemotherapy, barasertib<br />

showed a significant improvement in OCRR vs LDAC, and a safety pr<strong>of</strong>ile<br />

that was manageable and consistent with previous studies.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


6528 Poster Discussion Session (Board #20), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Effect <strong>of</strong> fludarabine, cytarabine, filgrastim, and gemtuzumab ozogamicin<br />

(FLAG-GO) on relapse-free survival in patients with newly diagnosed<br />

core-binding factor acute myelogenous leukemia. Presenting Author: Gautam<br />

Borthakur, University <strong>of</strong> Texas M. D. Anderson Cancer Center,<br />

Houston, TX<br />

Background: Prior modulation with fludarabine increases cytarabinetriphosphate<br />

(ara-CTP) accumulation and granulocyte-colony-stimulating<br />

factor (G-CSF) increases the fludarabine-triphosphate (F-ara-ATP) levels in<br />

leukemic blasts. Our front-line regimen <strong>of</strong> fludarabine, cytarabine and<br />

filgrastim (FLAG) based on this rationale showed improved event-free<br />

survival compared to anthracycline and cytarabine based regimens in<br />

patients (pts) with core-binding factor acute myelogenous leukemia (CBF-<br />

AML). Medical Research Council AML 15 trial reported survival benefit<br />

from addition <strong>of</strong> gemtuzumab ozogamicin (GO) to chemotherapy regimens<br />

in patients with favorable-risk cytogenetics AML. Methods: In a clinical trial<br />

combining GO (3 mg/m2 IV) with FLAG (FLAG-GO) in newly diagnosed<br />

CBF-AML, pts received GO on day 1 <strong>of</strong> induction and <strong>of</strong> post-remission<br />

cycles 2 or 3 and 5 or 6 in addition to FLAG. FLAG regimen was comprised<br />

<strong>of</strong> fludarabine 30 mg/m2 and cytarabine 2 gm/m2 IV daily (both for 5 days in<br />

induction and 3-4 days in post-remission cycles) with filgrastim started on<br />

day-1 and continued till neutrophil recovery. Pts who received one<br />

non-FLAG-GO induction could enroll irrespective <strong>of</strong> their remission status.<br />

Results: Fifty pts [30 with t(8;21) and 20 with Inv16] have been enrolled [5<br />

<strong>of</strong> 50 (10%) were in remission at enrollment from prior induction]. Median<br />

age is 48 years (range, 19-76), median WBC count 12.3 x106 /L (range,<br />

1.3-97.2); 12 patients (24%) were �60 years <strong>of</strong> age and frequency <strong>of</strong> kit<br />

mutation is 8%. Complete remission with or without platelet recovery<br />

(CR/CRp) was achieved in 43/45 (96%) pts with 2 deaths in induction.<br />

With 6 planned consolidations, the median number <strong>of</strong> consolidation<br />

treatments received is 5 (range, 0-6). Two-thirds <strong>of</strong> pts needed dose<br />

reduction during post-remission cycles. Seven (14%) pts [t(8;21)� 5, Inv<br />

16�2] relapsed and with a median follow-up <strong>of</strong> 34 months (range, 15-54<br />

months) the relapse-free survival rate is 83%. Conclusions: FLAG-GO is a<br />

highly effective regimen and has resulted in high rate <strong>of</strong> relapse-free<br />

survival in pts with newly diagnosed CBF AML.<br />

6530 Poster Discussion Session (Board #22), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

<strong>Clinical</strong> trial and compassionate use experience with glucarpidase for<br />

methotrexate toxicity. Presenting Author: Brigitte C. Widemann, Pediatric<br />

Oncology Branch, National Cancer Institute, National Institutes <strong>of</strong> Health,<br />

Bethesda, MD<br />

Background: High dose methotrexate (MTX) is used to treat acute lymphoblastic<br />

leukemia (ALL), osteogenic sarcoma, non-Hodgkin lymphoma<br />

(NHL), and other cancers. MTX-associated renal impairment with delayed<br />

MTX elimination develops after 2 to 10% <strong>of</strong> treatment cycles, exposing<br />

patients to elevated MTX concentrations with potential for enhanced MTX<br />

toxicity and prolonged hospitalization. Glucarpidase, a recombinant form <strong>of</strong><br />

carboxypeptidase G2, rapidly hydrolyzes MTX into inactive metabolites and<br />

provides an alternate way <strong>of</strong> clearance in patients with delayed MTX<br />

elimination. Methods: From November 1993 to June 2009, 492 patients<br />

experiencing renal toxicity and delayed elimination <strong>of</strong> MTX were treated<br />

with glucarpidase (50 U/kg intravenously) in compassionate use trials<br />

conducted in the US and EU. Their outcomes are presented here. Results:<br />

The median age <strong>of</strong> the 492 patients was 18 yrs (range: 5 wks to 85 yrs).<br />

Sixty-three percent were male. Forty-one percent had NHL, 30% osteogenic<br />

sarcoma, 23% ALL, and 7% other malignancies. The median<br />

pre-glucarpidase MTX concentration was 17 �mol/L. Seventy-six percent <strong>of</strong><br />

patients received 1 dose <strong>of</strong> glucarpidase, 22% received 2 doses, and 2%<br />

received 3 doses. The first dose <strong>of</strong> glucarpidase was given at a median <strong>of</strong> 3<br />

days after MTX administration. One-hundred and fifty-six patients had MTX<br />

concentrations determined by HPLC. At the first measurement (median 15<br />

minutes post-glucarpidase) MTX was reduced by a median <strong>of</strong> 99% relative<br />

to the pre-glucarpidase baseline. At the last measurement (median 40 hrs<br />

post-glucarpidase) median MTX reduction remained at 99% compared with<br />

baseline. In 410 patients with pre-glucarpidase renal impairment measured<br />

as CTCAE Grade 2 or higher, 64% recovered to Grade 0 or 1 after a<br />

median <strong>of</strong> 12.5 days post-glucarpidase. Glucarpidase was well-tolerated<br />

overall; adverse events included paresthesia (2.0%), flushing (1.8%) and<br />

headache (1.0%). Eight percent <strong>of</strong> patients died within 30 days <strong>of</strong><br />

glucarpidase administration <strong>of</strong> causes unrelated to glucarpidase, as judged<br />

by the treating physician. Conclusions: Glucarpidase is well-tolerated and<br />

reduces MTX concentrations by 99% within 15 min <strong>of</strong> administration in<br />

patients with impaired renal clearance <strong>of</strong> MTX.<br />

Leukemia, Myelodysplasia, and Transplantation<br />

423s<br />

6529 Poster Discussion Session (Board #21), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Allogeneic stem cell transplantation for patients over age 65 with acute<br />

myelogenous leukemia and myelodysplastic syndrome. Presenting Author:<br />

Henry Jacob Conter, University <strong>of</strong> Texas M. D. Anderson Cancer Center,<br />

Houston, TX<br />

Background: ASCT represents a potentially curative approach for AML and<br />

MDS, diseases that primarily affects patients in 7th and 8th decade <strong>of</strong> life.<br />

Here, we report outcomes <strong>of</strong> patients older than 64 treated at the MD<br />

Anderson Cancer Center from 1996 until December 2011. Methods: 182<br />

patients older than 64 received an ASCT for AML (n�143) or MDS (n�39).<br />

Outcomes <strong>of</strong> interest were transplant-related mortality (TRM), incidence <strong>of</strong><br />

acute and chronic graft-versus-host disease (GVHD), incidence <strong>of</strong> relapse,<br />

and overall survival (OS) - which were estimated from the date <strong>of</strong><br />

transplant. Results: The median age <strong>of</strong> patients was 67 (range 65-79). Most<br />

patients were transplanted with active disease (table). Median follow-up for<br />

alive patients was 12.6 months (n�63; range 0-118). The cumulative<br />

incidence <strong>of</strong> 100-day, 1 year, and 3 year TRM was 14%, 18%, and 21%,<br />

respectively. 26% <strong>of</strong> patients developed grade II-IV acute GVHD and 35%<br />

suffered from chronic GVHD. The actuarial incidence <strong>of</strong> relapse was 46% at<br />

1 year and 53% at 3 and 5 years. Actuarial OS was estimated to be 45% at<br />

1 year, 28% at 3 years, and 21% at 5 years. 3 year OS for patients<br />

transplanted in CR and with active disease was 40% versus 23% (p�0.02).<br />

OS <strong>of</strong> patients age 65-69 or �69 was 30% vs. 20% (p�0.06) at 3 years for<br />

all patients; compared to 38% versus 27% (p�0.23) for patients in those<br />

age groups transplanted in CR. Conclusions: Although a significant minority<br />

<strong>of</strong> patients older than 64 years may achieve long-term disease control, new<br />

approaches are needed to reduce TRM and relapse in this cohort <strong>of</strong><br />

patients.<br />

Category Parameter Number (%)<br />

Donor type Matched related donor 87 (48)<br />

Matched unrelated donor 73 (40)<br />

Mismatched unrelated donor 17 (9)<br />

Preparative regimens Fludarabine/melphalan 85 (47)<br />

Fludarabine/busulfan 61 (34)<br />

Fludarabine/idarubicin 13 (7)<br />

Immunosuppression Tacrolimus/mini-methotrexate 147 (81)<br />

Tacrolimus/mycophenolate 8 (4)<br />

Post-transplant cyclophosphamide 16 (9)<br />

Disease status at ASCT Primary induction failure 21 (12)<br />

Complete remission 1 38 (21)<br />

Complete remission 2 or 3 15 (8)<br />

Refractory/untreated disease 120 (66)<br />

6531 Poster Discussion Session (Board #23), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

BCR-ABL kinase domain mutations and resistance in Ph� acute lymphoblastic<br />

leukemia from the imatinib to the second-generation TKI era.<br />

Presenting Author: Simona Soverini, Department <strong>of</strong> Hematology/Oncology<br />

Seràgnoli, Bologna, Italy<br />

Background: Advent <strong>of</strong> 2nd-generation TKIs has brought additional treatment<br />

options for Philadelphia-positive (Ph�) acute lymphoblastic leukemia<br />

(ALL) patients (pts). To analyze the changes they have determined in<br />

mutation frequency and type, we have reviewed the database recording the<br />

results <strong>of</strong> BCR-ABL mutation analyses done in our laboratory from 2004<br />

through 2011. Methods: 781 tests on 258 pts were performed by direct<br />

sequencing. Results: 143 pts were analyzed because <strong>of</strong> imatinib resistance;<br />

101 (71%) had one or more mutations (a single mutation in 91 pts; two<br />

mutations in 10 pts). Three mutation types were by far the most frequent:<br />

T315I (38 pts, 37%), E255K (19 pts, 18%) and Y253H (19 pts, 18%). Of<br />

84 pts who had developed resistance to 2nd- or 3rd-line therapy with<br />

dasatinib, nilotinib or bosutinib after imatinib failure, 65 (77%) were<br />

positive for Bcr-Abl mutations; 30 (46%) carried multiple mutations (up to<br />

four) and in 19 <strong>of</strong> them (63%) this was consequence <strong>of</strong> multiple lines <strong>of</strong><br />

therapy. The most frequent newly acquired mutation in this setting was the<br />

T315I, detected in 35/57 (61%) cases acquiring mutations on dasatinib.<br />

Mutation analysis was also performed in 15 resistant pts enrolled in a study<br />

<strong>of</strong> dasatinib as 1st-line treatment <strong>of</strong> Ph� ALL; 12 pts were positive, 11 <strong>of</strong><br />

them had a T315I. Taking advantage <strong>of</strong> a next-generation sequencer<br />

(Roche 454), allowing a high sensitive and quantitative mutation scanning<br />

<strong>of</strong> Bcr-Abl, serially collected samples from 24 selected cases who developed<br />

mutations and resistance to one or more TKIs were retrospectively<br />

analyzed to study the kinetics <strong>of</strong> expansion <strong>of</strong> mutant clones. Results will<br />

be presented. Conclusions: Although 2nd generation TKIs are more potent<br />

and have much fewer insensitive mutations, long-term disease control<br />

remains a problem and the T315I becomes an even tougher enemy. The<br />

high genetic instability fosters mutational events anytime during TKI<br />

treatment and some mutation types (T315I, Y253H) have been observed to<br />

emerge and take over very quickly (from �0.01% to 90% in one-two<br />

months). Supported by PRIN, AIL, AIRC, Fondazione CARISBO.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


424s Leukemia, Myelodysplasia, and Transplantation<br />

6532 Poster Discussion Session (Board #24), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Outcomes <strong>of</strong> patients older than 60 years with acute lymphoblastic<br />

leukemia: Survey from the Surveillance, Epidemiology, and End Results<br />

(SEER) program. Presenting Author: Jae Hong Park, Leukemia Service,<br />

Memorial Sloan-Kettering Cancer Center, New York, NY<br />

Background: The cure rate <strong>of</strong> pediatric acute lymphoblastic leukemia (ALL)<br />

has increased over the last 4 decades to above 80%, compared to a much<br />

smaller improvement in adults aged � 60 years. However, outcome<br />

information on older ALL patients (age � 60 years) is limited. Only a few<br />

clinical trials include the older patients, apply the same regimens developed<br />

for adults <strong>of</strong> all ages, and report a very poor outcome with no<br />

improvement over time. We therefore conducted a population-based survey<br />

<strong>of</strong> older ALL patients focusing on early death (ED) rates and changes in<br />

outcome over the last 30 years. Methods: Data from 9 population-based<br />

cancer registries that participate in the National Cancer Institute’s SEERprogram<br />

were used to identify patients aged 60 or older with a diagnosis <strong>of</strong><br />

ALL. Survival rates at 1, 6, 12 and 24 months were estimated using<br />

actuarial methods for 4 calendar periods: 1980-1985, 1986-1992,<br />

1993-1999, and 2000-2006. ED was defined as death occurring within<br />

one month <strong>of</strong> ALL diagnosis. Results: A total <strong>of</strong> 1066 ALL patients were<br />

identified. The ED rate significantly improved over the four study time<br />

periods from 20.2% in 1980-1985 to 13.2% in 2000-2006 (p�0.03).<br />

The overall survival (OS) at 6 months improved from 32.8% in 1980-1985<br />

to 45.3% in 2000-2006, but at 24 months, only a modest difference in OS<br />

was noted across the time period (13.1% in 1980-85 vs. 17.5% in<br />

2000-06). The median survival increased from 3 months to 6 months from<br />

the period 1980-1999 to 2000-2006. Conclusions: Although the longterm<br />

OS for patients aged 60 and over remains poor, there has been a slight<br />

improvement in early mortality and median OS from 1980s to the early 21st century. While the progress in reducing ED and increasing survival at 6<br />

months is encouraging, and may be reflecting better supportive care<br />

measures, the limited improvement indicates poor tolerance and lack <strong>of</strong><br />

efficacy <strong>of</strong> the toxic, long and complex chemotherapy regimens designed<br />

for younger adults. Therefore, future studies should be designed specifically<br />

for older ALL patients, focusing on novel, effective, but less toxic<br />

therapies, to further improve the short-term OS seen in the past decades<br />

and possibly a better overall outcome.<br />

6534 General Poster Session (Board #14A), Mon, 1:15 PM-5:15 PM<br />

Impact <strong>of</strong> treatment prior to allogeneic stem cell transplantation for<br />

myelodysplastic syndromes: A study on behalf <strong>of</strong> the SFGM-TC and the<br />

GFM groups. Presenting Author: Gandhi Laurent Damaj, Centre Hospitalier<br />

Universitaire Amiens Sud, Amiens, France<br />

Background: Until the early 2000s, treatment prior to SCT varied according<br />

to cytogenetics and % BM blasts and was mainly induction-type chemotherapy<br />

(ICT). The role <strong>of</strong> Azacitidine (AZA) prior to transplant remains<br />

uncertain. Methods: 163 consecutive pts (M/F�104/67; age � 18)<br />

underwent alloSCT between 2005 and 2009 from sibling (n�96) or HLAallele-MUD<br />

(10/10) (n�67). The SC source was blood (n�142) or marrow<br />

(n�21) and conditionings were myeloablative (n�33); nonmyeloablative<br />

(n�130). Pts who did not received neither prior ICT nor AZA were excluded<br />

since the main objective <strong>of</strong> this study was to investigate the impact <strong>of</strong> prior<br />

therapy on pts’ outcome. Results: At diagnosis, FAB/WHO was: 24 RA/RARS/<br />

RCMD, 52 RAEB-1, 74 RAEB-2, 13 RAEB-t/AML; Cytogenetic: fav<br />

(n�93), int (n�32), unfav (n�37). 98 pts received prior ICT (ICT-group)<br />

and 65 had received AZA, either alone (AZA-group; n�48) or AZA preceded<br />

or followed by intensive CT (AZA-chemo-group; n�17). In AZA groups, the<br />

drug was started 38 to 941 days from transplant and discontinued 6 to 438<br />

days before transplant with a median number <strong>of</strong> 4 cycles (range 1-26).<br />

Overall, 119 pts were considered responders at SCT (CR, PR, mCR),<br />

including 33 (69%) treated with AZA-alone, 9 (53%) with AZA-chemo and<br />

77(78%) with ICT.While there were no differences between AZA-group and<br />

ICT-group in terms <strong>of</strong> OS, EFS, relapse and NRM, having a poor risk<br />

cytogenetic and receiving AZA-chemo prior to SCT were, in multivariate<br />

analyses, the most important factors that adversely influenced OS with<br />

[HR�3.03; 95% CI 1.84-4.96] (p�.0001) and [HR�2.72; 95% CI,<br />

1.38-5.35] (p�.004), respectively. Pts <strong>of</strong> AZA-chemo group had the same<br />

rate <strong>of</strong> relapse but significantly higher NRM rate (p�.059) than those who<br />

received either AZA or ICT alone. Conclusions: With the goal <strong>of</strong> downstaging<br />

underlying disease before allo-SCT, AZA treatment appears to be a<br />

valid therapeutic approach, but mainly in pts receiving AZA alone with an<br />

outcome at least equivalent to pts receiving ICT prior to SCT. Allo-SCT in<br />

pts who required both AZA and chemotherapy had less satisfactory<br />

outcomes, possibly reflecting additional toxicity and/or more resistant<br />

disease.<br />

6533 Poster Discussion Session (Board #25), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Impact <strong>of</strong> donor source on allogeneic stem cell transplantation for Philadelphia<br />

chromosome-negative acute lymphoblastic leukemia. Presenting Author:<br />

Satoshi Nishiwaki, Department <strong>of</strong> Hematology and Oncology, Nagoya<br />

University Graduate School <strong>of</strong> Medicine, Nagoya, Japan<br />

Background: Although allogeneic stem cell transplantation (allo-SCT) could<br />

improve the outcome <strong>of</strong> adult Philadelphia chromosome-negative acute<br />

lymphoblastic leukemia [Ph(-) ALL], the impact <strong>of</strong> the donor source,<br />

particularly the position <strong>of</strong> cord blood (CB) transplantation, is still uncertain.<br />

Methods: We retrospectively analyzed 1726 adult Ph(-) ALL patients<br />

transplanted at the first time between 1998 and 2009 with myeloablative<br />

preparative regimens who were registered in the Japan <strong>Society</strong> for Hematopoietic<br />

Cell Transplantation database. Two hundred and thirty-three<br />

received CB transplantation [first complete remission (CR1): 95, subsequent<br />

CR: 53, non-CR: 85], 809 received allo-SCT from unrelated donor<br />

(URD) (CR1: 434, subsequent CR: 158, non-CR: 217), and 684 received<br />

allo-SCT from related donor (RD) (CR1: 388, subsequent CR: 89, non-CR:<br />

207). Results: Overall survival (OS) in patients after CB transplantation in<br />

CR1 was comparable with that after allo-SCT from URD or RD [57% in CB,<br />

64% in URD, and 65% in RD at 4 years, respectively, P�0.11]. Donor<br />

source was not a significant risk factor for OS in multivariate analysis.<br />

Although URD was a favorable factor for relapse and an unfavorable factor<br />

for non-relapse mortality (NRM), CB was not a significant factor for them<br />

[Relapse: 22% in CB, 17% in URD, and 24% in RD at 3 years, respectively<br />

(P�0.02); NRM: 27% in CB, 23% in URD, and 13% in RD at 3 years,<br />

respectively (P�0.0001)]. Among CB recipients in CR1, age at allo-SCT<br />

(45 years or older) was solely a significant adverse prognostic factor in<br />

multivariate analysis. Among patients younger than 45 years who received<br />

allo-SCT in CR1, OS after CB transplantation was significantly better than<br />

that after allo-SCT from mismatched URD (4-year OS: 68% vs. 49%,<br />

P�0.04). Similarly, OS was not different by donor source in subsequent CR<br />

or non-CR [Subsequent CR: 48% in CB, 39% in URD and 48% in RD,<br />

P�0.33; non-CR: 18% in CB, 21% in URD, and 15% in RD, P�0.20 at 4<br />

years, respectively]. Conclusions: Allo-SCT using CB led to similar outcomes<br />

as either RD or URD in any disease status. CB transplantation is a<br />

good alternative for adult Ph(-) ALL patients without a suitable RD or URD.<br />

6535^ General Poster Session (Board #14B), Mon, 1:15 PM-5:15 PM<br />

Immune-reconstitution after combined haploidentical and umbilical cord<br />

blood transplantation. Presenting Author: Koen van Besien, Weill Cornell<br />

Medical College, New York, NY<br />

Background: Umbilical cord blood (UCB) stem cells are frequently employed<br />

for allogeneic stem cell transplantation. However, delayed myeloid<br />

and lymphoid immune-reconstitution with UCB transplantation leads to<br />

increased risk <strong>of</strong> infections. We recently reported a pilot study combining<br />

UCB with a HLA haploidentical donor (Liu et al. Blood. 2011). Here, we<br />

report the immune reconstitution after the haploidentical-cord blood<br />

transplantation. Methods: Pts received reduced-intensity conditioning with<br />

fludarabine/melphalan/rATG and were assessed for lymphocyte subsets,<br />

serological response to pneumococcal vaccination, Cylex Immuknow assay<br />

which measures amount <strong>of</strong> ATP secreted by CD4 lymphocytes on stimulation<br />

by PHA, and T-cell spectratyping. Results: 45 pts, enrolled between<br />

01/2007 and 04/2011, are included in this analysis. The median absolute<br />

lymphocyte subset counts at serial time-points are shown in the Table. The<br />

median absolute values <strong>of</strong> CD3, CD4 and CD8 counts remained below the<br />

normal range throughout the 1st year post-transplant. The NK-cells and<br />

B-cells reached normal range by day 30 and day 100, respectively.<br />

Serological response to pneumococcal vaccination was seen in 7/17<br />

(defined as seroconversion or �3 fold rise in titers <strong>of</strong> serotype 14, 19F and<br />

23F) evaluable pts at a median 74 days after vaccination. Immuknow assay<br />

showed that 6/6 pts tested within first 30 days <strong>of</strong> transplant had low values<br />

whereas 5/8 tested �1yr had normal/high values. Of the 9 pts who were<br />

assessed for T-cell receptor repertoire by T-cell spectratyping, 7 had a<br />

polyclonal, complex T cell repertoire similar to healthy controls. CMV<br />

viremia requiring treatment occurred in 14 patients. EBV reactivation<br />

occurred in 18 pts and 5 developed PTLD 68 to 314 days after transplant.<br />

Conclusions: Haploidentical cord blood transplantation leads to rapid<br />

myeloid recovery and recovery <strong>of</strong> B-cell and NK cell numbers. T-cell<br />

recovery is more delayed, but at 1-year most pts have a polyclonal T cell<br />

repertoire and robust T-cell responses.<br />

Absolute count, median<br />

Day 30<br />

N�28<br />

Day 100<br />

N�31<br />

Day 180<br />

N�23<br />

Day 365<br />

N�15<br />

Total lymphocyte 256 747 959 1,354*<br />

CD3 3.5 25 114 447<br />

CD4 2 6 70 194<br />

CD8 2.5 8 30 146<br />

CD19 1 297* 401 439<br />

CD56 (CD3-)<br />

* Within normal range.<br />

220* 228 267 150<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


6536 General Poster Session (Board #14C), Mon, 1:15 PM-5:15 PM<br />

Impact <strong>of</strong> ATG on new HLA groups for unrelated donor allogeneic stem cell<br />

transplantation. Presenting Author: Soo Jung Lee, Department <strong>of</strong> Hematology/Oncology<br />

and Stem Cell Transplantation Unit, Kyungpook National<br />

University Hospital, Kyungpook National University School <strong>of</strong> Medicine,<br />

Daegu, South Korea<br />

Background: The outcomes <strong>of</strong> unrelated donor transplantation have improved<br />

with refinements in HLA testing. Recently, grouping <strong>of</strong> HLA<br />

matching for unrelated donor was suggested, defining by well-matched,<br />

partially-matched, and mismatched. In the current study, the role <strong>of</strong> ATG<br />

for each group was evaluated. Methods: A total <strong>of</strong> 92 patients diagnosed as<br />

hematologic diseases and received allogeneic stem cell transplantation<br />

(SCT) from unrelated donor were retrospectively analyzed. Results: Nineteen<br />

patients were classified as well-matched, 42 as partially-matched,<br />

and 31 as mismatched. Among them, 57 patients received anti-thymocyte<br />

globulin (ATG) as graft-versus-host disease (GVHD) prophylaxis. The overall<br />

survival (OS) rate was higher for well-matched group (83%) compared to<br />

partially-matched (54%) and mismatched (34%, p�0.076). For partiallymatched<br />

group, the OS was significantly improved with ATG (83.3% vs.<br />

38.6%, p�0.018). But, the OS was not different between groups with or<br />

without ATG for well-matched (87.5% vs. 66.7%, p�0.487) and mismatched<br />

(32.4% vs. 41.7%, p�0.215). ATG decreased the cumulative<br />

incidence <strong>of</strong> grade 3-4 acute GVHD (10% vs. 40.2%, p�0.068) and severe<br />

chronic GVHD (21.2% vs. 52.2%, p�0.037). The use <strong>of</strong> ATG (HR�0.248,<br />

p�0.029) was related with favorable OS for partially-matched group.<br />

However, the favorable effect <strong>of</strong> ATG was not observed in well-matched and<br />

mismatched groups. Conclusions: ATG effectively improved survival rate for<br />

partially-matched group in unrelated donor transplantation. However, the<br />

positive effect <strong>of</strong> ATG was not observed in well-matched and mismatched<br />

group.<br />

6538 General Poster Session (Board #14E), Mon, 1:15 PM-5:15 PM<br />

The outcomes <strong>of</strong> allogeneic stem cell transplantation in AML patients with<br />

monosomal karyotypes. Presenting Author: Sang Kyun Sohn, Department<br />

<strong>of</strong> Hematology/Oncology, Kyungpook National University Hospital, Kyungpook<br />

National University School <strong>of</strong> Medicine, Daegu, South Korea<br />

Background: The prognosis <strong>of</strong> acute myeloid leukemia (AML) with monosomal<br />

karyotype (MK) was reported extremely poor. We investigated the role<br />

<strong>of</strong> allogeneic stem cell transplantation (allo-SCT) for those with MK.<br />

Methods: A total <strong>of</strong> 114 patients who received allo-SCT for treatment <strong>of</strong><br />

AML were retrospectively analyzed. All patients were treated with standard<br />

induction chemotherapy with anthracycline and cytarabine. Cytogenetic<br />

abnormalties were grouped according to recently published MRC criteria<br />

and MK was defined as at least two autosomal monosomies or one<br />

monosomy plus one or more structural abnormality. Results: Thirteen<br />

patients had favorable cytogenetic risk, 78 intermediate, 11 adverse<br />

without MK, and 12 adverse with MK at the time <strong>of</strong> diagnosis. Among 12<br />

patients with MK, 5 (41.7%) achieved CR after induction therapy, 1<br />

(8.3%) relapsed and 6 (50.0%) refractory at the time <strong>of</strong> allo-SCT. The<br />

2-year overall survival (OS) was significantly lower for patients with MK<br />

(17.5%) compared to favorable (76.9%), intermediate (61.0%), and<br />

adverse without MK (36.4%, p�0.017). In the multivariate analysis, those<br />

with MK was related with extremely poor outcomes (HR 6.02, p�0.008),<br />

which was independent risk factor for OS. Survival benefit was observed in<br />

MK group with chronic GVHD compared to those without chronic GVHD.<br />

The median survival was 272 days with chronic GVHD (95% CI 204-339<br />

days) compared to 159 days (95% CI 76-172 days) without chronic GVHD<br />

(p�0.010). Conclusions: The prognosis remained poor in patients with MK<br />

despite <strong>of</strong> allo-SCT. Innovative approaches to induce GLV effects are<br />

needed to improve SCT outcomes in patients with MK.<br />

Leukemia, Myelodysplasia, and Transplantation<br />

425s<br />

6537 General Poster Session (Board #14D), Mon, 1:15 PM-5:15 PM<br />

Low day 100 transplant-related mortality (TRM) and relapse rate following<br />

cl<strong>of</strong>arabine (CLO) in combination with cytarabine, total body irradiation<br />

(TBI), and allogeneic stem cell transplantation (AlloSCT) in children,<br />

adolescents, and young adults (CAYA) with poor-risk acute leukemia.<br />

Presenting Author: Kavita Radhakrishnan, New York Medical College,<br />

Valhalla, NY<br />

Background: CAYA with ALL or AML in third complete remission (CR3),<br />

refractory relapse (RR) or induction failure (IF) have an extremely poor<br />

prognosis, �20% EFS (Gaynon, BJH, 2005;Wells, JCO, 2003). CLO, an<br />

inhibitor <strong>of</strong> DNA polymerase and ribonucleotide reductase, has significant<br />

activity in CAYA with relapsed ALL/AML (Jeha, JCO 2006,2009) and<br />

synergy with cytarabine (Faderl, Blood, 2005). We sought to determine<br />

safety, day-100 TRM, and overall survival (OS) associated with CLO,<br />

cytarabine and TBI followed by AlloSCT in CAYA with poor-risk ALL/AML.<br />

Methods: This is a multi-center phase I/II trial <strong>of</strong> a novel conditioning<br />

regimen <strong>of</strong> CLO (dose escalation: 40mg/m2 [n�3], 46 mg/m2 [n�3], 52<br />

mg/m2 [n�19]) x5d, sequential (4 hrs later) cytarabine 1000 mg/m2 x6d<br />

and TBI (1200cGy) followed by AlloSCT from matched related or unrelated<br />

donors in CAYA with ALL/AML in CR3, RR or IF. Patients with unrelated<br />

donors received R-ATG. GVHD prophylaxis consisted <strong>of</strong> tacrolimus and<br />

MMF (Bhatia/Cairo, BBMT, 2009). Kaplan-Meier method was used to<br />

determine the probabilities <strong>of</strong> engraftment, GVHD, TRM and OS. Results:<br />

25 pts, median age: 11.3 yrs (1.5-20.7); M:F: 19:6, ALL/AML: 22:3 (10<br />

CR3, 3 RR, 12 IF), 10 related donors, 15 unrelated donors (9 BM/PBSCs,<br />

6 UCB). Median TNC and CD34 dose was 4.47x108 /kg and 4.84x106 /kg<br />

for BM/PBSCs and 4.0x107 /kg and 2.8x105 /kg for UCB, respectively.<br />

Probabilities <strong>of</strong> neutrophil, platelet engraftment and grade II-IV aGVHD<br />

were 100%, 92.9% and 47.5%, respectively. CLO dose was tolerable at<br />

52mg/m2 /d x5d without dose limiting toxicity. Probability <strong>of</strong> Day 100 TRM<br />

was only 4.3%. Probability <strong>of</strong> 1-yr PFS and OS were 51% (CI95: 28-71%),<br />

and 43% (CI95: 22-63%) respectively. Conclusions: Preliminary results<br />

suggest this novel regimen followed by AlloSCT is safe and well tolerated in<br />

CAYA with poor-risk ALL/ AML with CLO dose 52 mg/m2 . Results are<br />

encouraging with respect to low risk <strong>of</strong> day 100 TRM and leukemic relapse<br />

associated with this conditioning regimen in this poor-risk population.<br />

6539 General Poster Session (Board #14F), Mon, 1:15 PM-5:15 PM<br />

Effect <strong>of</strong> IL-22 on intestinal stem cells, GVHD-related tissue damage, and<br />

GVL. Presenting Author: Alan M. Hanash, Memorial Sloan-Kettering Cancer<br />

Center, New York, NY<br />

Background: Intestinal graft vs. host disease (GVHD) is a major complication<br />

<strong>of</strong> allogeneic bone marrow transplantation (allo-BMT). Strategies to<br />

limit GVHD by selective promotion <strong>of</strong> epithelial regeneration in the absence<br />

<strong>of</strong> immunosuppression are largely unknown. Methods: We investigated the<br />

role <strong>of</strong> IL-22 in allo-BMT by utilizing wild type and IL-22 knockout (KO)<br />

mice as donors or recipients in allo-BMT. Results: We found that administration<br />

<strong>of</strong> an anti-IL-22 neutralizing antibody to allo-BMT recipients during<br />

the first month post-BMT led to increased GVHD mortality, indicating that<br />

IL-22 functioned to reduce GVHD severity post-BMT. In contrast, use <strong>of</strong><br />

IL-22 KO donor T cells did not impair their ability to eliminate A20<br />

lymphoma cells upon tumor challenge, thus indicating that IL-22 was not<br />

essential for donor T cells to mediate graft vs. lymphoma (GVL) responses.<br />

BMT with WT donors and recipients indicated that IL-22 levels in small and<br />

large intestine were increased after BMT and after radiation injury (RI)<br />

without BMT. IL-22 upregulation after RI was dependent on the presence<br />

<strong>of</strong> IL-23p40. Although intestinal IL-22 levels were increased after T<br />

cell-depleted (TCD) BMT, intestinal IL-22 was reduced by GVHD, as IL-22<br />

production was mediated by host-derived innate lymphoid cells (ILC) that<br />

were eliminated by GVHD. Furthermore, host-derived IL-22 was critical for<br />

reduction <strong>of</strong> GVHD morbidity, mortality, and intestinal pathology. GVHD in<br />

IL-22 KO mice led to increased apoptosis in epithelial crypts where the<br />

intestinal epithelial stem/progenitor cell niche is located. Immunohistochemistry<br />

and immun<strong>of</strong>luorescence demonstrated IL-22 receptor expression<br />

on intestinal stem cells (ISC) and progenitors. Allo-BMT in Lgr5-LacZ<br />

reporter mice indicated that ISC were targeted by GVHD, and GVHD in<br />

IL-22 KO mice led to dramatic ISC depletion. Conclusions: IL-22 is critical<br />

for protection <strong>of</strong> host epithelium during GVHD and critical for protection <strong>of</strong><br />

ISC, and it does not contribute to donor T cell GVL responses. These<br />

findings may have broad relevance for protection <strong>of</strong> ISC and intestinal<br />

epithelium in clinical GVHD and other inflammatory intestinal diseases,<br />

and may be useful for clinical separation <strong>of</strong> pathologic GVH and therapeutic<br />

GVL responses.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


426s Leukemia, Myelodysplasia, and Transplantation<br />

6540 General Poster Session (Board #14G), Mon, 1:15 PM-5:15 PM<br />

Outcomes <strong>of</strong> second allogeneic hematopoietic stem cell transplantation<br />

(SCT) for patients with acute lymphoblastic leukemia (ALL). Presenting<br />

Author: L. M. Poon, University <strong>of</strong> Texas M. D. Anderson Cancer Center,<br />

Houston, TX<br />

Background: Disease relapse is the major cause <strong>of</strong> failure after allogeneic<br />

hematopoietic stem cell transplantation (SCT) for acute lymphoblastic<br />

leukemia (ALL). Treatment options for these patients (pts) are limited and<br />

only a second SCT provides a realistic chance for long term disease<br />

remission. Methods: We retrospectively analyzed the outcomes <strong>of</strong> 31 pts<br />

with ALL who relapsed following their first allogeneic SCT, and went on to<br />

receive a second allogeneic SCT. Univariate analysis was used to assess for<br />

risk factors which influenced treatment-related mortality (TRM), and<br />

progression free survival (PFS) following second SCT. Results: 31 pts were<br />

evaluable for response with 2 early deaths within 30 days <strong>of</strong> SCT. The<br />

median age <strong>of</strong> the pts was 26 years (range 7- 49), and the median duration<br />

between their first SCT (SCT1) to relapse was 9.5 months (range 2.2-32.6<br />

months). 39% <strong>of</strong> pts were transplanted in active disease (n�12). The<br />

complete remission (CR) rate post SCT was 89%; 83% <strong>of</strong> pts transplanted<br />

with active disease attained CR. With a median follow-up <strong>of</strong> 3 years among<br />

survivors, PFS, and OS rates at 1 year were estimated at 23%. The TRM<br />

rate was 41% at 12 months. We did not identify any factors that impacted<br />

TRM through univariate analysis. We found a significant relationship<br />

between the time to progression following SCT 1 and PFS following SCT 2<br />

(p�0.02, HR�0.93/month). Conclusions: In summary, a second transplant<br />

remains an effective method for achieving response in a highly refractory<br />

patient population. Pts with longer PFS following SCT1 have a better PFS<br />

following SCT2. While long-term survival is limited, a significant proportion<br />

<strong>of</strong> pts remain disease-free for up to one year following SCT2, providing a<br />

window <strong>of</strong> time to administer preventive interventions. Notably, our 4<br />

long-term survivors, received novel therapies in the form <strong>of</strong> a single<br />

umbilical cord blood unit in addition to the PBSC to augment the immune<br />

response (n�2), a change in the stem cell source for the SCT from cord to<br />

mismatched adult unrelated (n�1), and post SCT maintenance therapy<br />

with 5-azacytidine (n�1), underscoring the need for a fundamental change<br />

with the methods for the second transplant to improve outcome.<br />

6542 General Poster Session (Board #15A), Mon, 1:15 PM-5:15 PM<br />

Impact <strong>of</strong> antithymocyte globulin (ATG)-containing conditioning regimens<br />

on patients undergoing allogeneic stem cell transplantation (allo-CST) for<br />

poor risk cytogenetic IPSS myelodysplastic syndrome (PRC-MDS): A report<br />

from the French <strong>Society</strong> <strong>of</strong> Stem Cell Transplantation and Cell Therapy<br />

(SFGM-TC). Presenting Author: Ibrahim Yakoub-Agha, University Hospital,<br />

Lille, France<br />

Background: Due to a risk <strong>of</strong> relapse <strong>of</strong> underlying disease in patients with<br />

PRC-MDS, the use <strong>of</strong> ATG, incorporated within the conditioning regimen<br />

prior to allo-SCT, is still controversial. Methods: Inclusion criteria included<br />

patients aged over 18 (n�101) who received allo-SCT transplanted<br />

between 1999 and 2009 from either a sibling (n�68) or HLA-allele-MUD<br />

(10/10) (n�33) for PRC-MDS. HLA matching was double-checked by the<br />

national Bone Marrow Donor Registry. Results: According to the FAB/WHO<br />

classification at diagnosis, 22 pts had RA/RARS/RCMD, 40 RAEB1, 30<br />

REAB2 and 9 RAEB-t/AML. 34 pts had progressed to a more advanced<br />

disease before allo-SCT. At diagnosis, 89 patients had an IPSS int-2 or<br />

higher. At transplant, 36 pts were responders (CR, PR, CRm) and 62 with<br />

progressive disease (relapsed/refractory, untreated or stable disease without<br />

hematological improvement). Median age at transplantation was 54<br />

years (range, 22-69). Pts received myeloablative conditioning (n�46) and<br />

nonmyeloablative (n�55). In this series, 48 patients received ATG as part<br />

<strong>of</strong> conditioning (’ATG’ group), whereas 53 did not (’no-ATG’ group). As <strong>of</strong><br />

April 1st 2011, 44 patients died <strong>of</strong> relapse and 22 <strong>of</strong> TRM. 3-year relapse,<br />

overall and event-free survival rates were not significantly different between<br />

the two groups. In contrast, the cumulative incidence <strong>of</strong> grade 2-4 acute<br />

GVHD was 48% in the no-ATG group and 30% ATG group (P �.005).<br />

Although the cumulative incidence <strong>of</strong> chronic GVHD was similar in the<br />

no-ATG and ATG groups, a trend for a lower TRM was observed in the ATG<br />

group (p�.06). In multivariate analysis, the absence <strong>of</strong> use <strong>of</strong> ATG was<br />

associated with an increased risk <strong>of</strong> acute grade 2-4 [HR � 1.92, p�.044].<br />

Conclusions: The addition <strong>of</strong> ATG to the conditioning regimen resulted in a<br />

decreased incidence <strong>of</strong> acute GVHD without increasing relapse rates and<br />

compromising survival <strong>of</strong> patients undergoing allo-SCT for poor risk<br />

cytogenetic MDS.<br />

6541 General Poster Session (Board #14H), Mon, 1:15 PM-5:15 PM<br />

Randomized phase III trial <strong>of</strong> haploidentical HCT with or without an add<br />

back strategy <strong>of</strong> HSV-TK donor lymphocytes in patients with high-risk acute<br />

leukemia. Presenting Author: Claudio Bordignon, Università Vita-Salute,<br />

Istituto Scientifico San Raffaele; MolMed, Milan, Italy<br />

Background: Suicide gene therapy applied to allogeneic stem cell transplantation<br />

has been tested in several trials. When exposed to ganciclovir, donor<br />

lymphocytes expressing herpes simplex thymidine kinase suicide gene (TK<br />

cells) are selectively eliminated. In a phase I-II trial (Ciceri, Bonini, Lancet<br />

Oncology 2009,489-500), TK cells were infused in patients after haploidentical<br />

transplantation. Of 50 patients enrolled, 28 received transduced-TK<br />

cells and 22 achieved rapid immune reconstitution, with a non-relapse<br />

mortality <strong>of</strong> 14%. GvHD after DLI-TK infusion was reported in 30% <strong>of</strong><br />

patients and in all cases was fully controlled by ganciclovir treatment.<br />

Methods: High risk leukemia patients in first or subsequent complete<br />

remission are currently randomized in a 2-arm (3:1 ratio) phase III trial with<br />

or without the add-back strategy <strong>of</strong> HSV-TK donor lymphocytes in patients<br />

underwent T-depleted haploidentical stem cell transplant. Primary end<br />

point is disease-free survival (DFS), while secondary end points included<br />

overall survival, non-relapse mortality, immune reconstitution, acute and<br />

chronic GvHD incidence, and relapse incidence. For the primary analysis <strong>of</strong><br />

DFS 170 patients (127 patients in the experimental group and 43 patients<br />

in the control) are required to detect an hazard ratio <strong>of</strong> 0.55 with at least<br />

80% power for 2-sided 0.05 level test. Patients are eligible for the study if<br />

they are older than 18 years-old, absence <strong>of</strong> timely and suitable HLA<br />

matched family or unrelated donor, and have a ECOG performance status �<br />

2. Patients are stratified by state <strong>of</strong> disease, ECOG PS and country before<br />

randomization. Results: Transduced lymphocytes are given to patients<br />

between day �21 and day �49 after haploidentical HCT in the absence <strong>of</strong><br />

spontaneous immune reconstitution and/or development <strong>of</strong> GvHD. In<br />

absence <strong>of</strong> immune reconstitution and GvHD after the first TK-cells<br />

add-back further 3 infusions could be administered 30 days after the last<br />

infusion. TK lymphocytes dose is 1 x10^7 CD3/Kg. Conclusions: The study<br />

(TK008) is currently open to accrual in EU, US, Israel (clinicaltrials.gov:<br />

00914628).<br />

6543 General Poster Session (Board #15B), Mon, 1:15 PM-5:15 PM<br />

Minimum tolerable interval <strong>of</strong> 90yttrium ibritumomab-tiuxetan to autologous<br />

stem cell transplantation after high-dose chemotherapy with carmustin,<br />

etoposide, cytarabine, and melphalan for relapsed or refractory<br />

aggressive B-cell non-Hodgkin lymphoma. Presenting Author: Justin Hasenkamp,<br />

University Medicine Goettingen, Goettingen, Germany<br />

Background: High-dose therapy and autologous stem cell transplantation<br />

(ASCT) in patients (pts) with aggressive B-NHL failing from immunochemotherapy<br />

including rituximab show poor outcome with 3y PFS <strong>of</strong> 39%<br />

(Gisselbrecht et al. JCO 2010). Combining BEAM with 90yttrium ibritumomab<br />

tiuxetan is a promising option to enhance the efficacy <strong>of</strong> the<br />

high-dose regimen. Methods: Pts without disease progression during<br />

salvage therapy <strong>of</strong> relapsed or refractory CD20� aggressive B-NHL were<br />

included in this prospective, multicenter, phase I/II trial. Primary endpoint<br />

was the maximum tolerated dose <strong>of</strong> 90Yttrium ibritumomab tiuxetan given<br />

as close as possible to ASCT defined as �2 pts with dose limiting toxicity in<br />

a 6�6 pts cohort. First, we reduced the time interval <strong>of</strong> 90yttrium ibritumomab tiuxetan (0.4 mCi/kg body weight) to ASCT from 14 to 12 and<br />

then 10 days. Subsequently it was planned to increase the 90yttrium ibritumomab tiuxetan dose. Results: From 2006 to 2009 26 pts, median<br />

age 58y (34-66) were enrolled. Histology included 14 DLBCL, 6 FL III°, 5<br />

transformed FL and 1 aggressive B-NHL without further subtyping. 11/26<br />

pts had relapse/progression within 1y after diagnosis. Secondary IPI was<br />

�1 in 14 pts. Response to salvage therapy was CR in 6/26 pts and PR in<br />

12/26 pts. 20/26 pts achieved CR after ASCT. Engraftment showed no<br />

significant differences in pts <strong>of</strong> different cohorts with median recovery <strong>of</strong><br />

leukocytes (�1/nl) and platelets (�25/nl) at d �10 and �13. Two early<br />

deaths (d �7, �18) occurred due to infections. 90Yttrium ibritumomab<br />

tiuxetan (0.4 mCi/kg body weight) at d -10 <strong>of</strong> ASCT was determined as<br />

minimum tolerated interval <strong>of</strong> radioimmunotherapy to ASCT after BEAM.<br />

Median follow-up <strong>of</strong> the survivors is 3.3 y. 3y PFS and OS is 67% (95% CI,<br />

49%-85%) and 75% (95% CI, 58%-92%), respectively. Main cause <strong>of</strong><br />

death was relapse/progression with 27% (95% CI, 6%-38%) at 3y.<br />

Conclusions: Z-BEAM followed by ASCT was safe and feasible and results in<br />

a high response rate with durable remissions in rituximab pretreated<br />

patients with aggressive B-NHL. Extended studies at the maximum<br />

tolerated dose and confirmation <strong>of</strong> superiority compared to conventional<br />

ASCT are warranted.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


6544 General Poster Session (Board #15C), Mon, 1:15 PM-5:15 PM<br />

Allogenic stem cell transplant (ASCT) as initial salvage for patients (pts)<br />

with acute myeloid leukemia (AML) refractory to high-dose cytarabinebased<br />

induction chemotherapy. Presenting Author: Naval Guastad Daver,<br />

University <strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: Outcomes <strong>of</strong> pts with AML who are refractory to High-dose<br />

Cytarabine (HiDAC) based induction are dismal. ASCT as initial salvage<br />

may be effective and potentially superior to repeat induction with combination<br />

chemotherapies in such pts. Methods: 1597 AML pts were treated with<br />

HiDAC-based induction at MD Anderson Cancer Center between 1995 and<br />

2009. 285 primary refractory pts were identified. 28 (10%) <strong>of</strong> these<br />

underwent ASCT as initial salvage and were reviewed. Results: Median age<br />

was 56 years (36 to 77) and 50% were males. Median ECOG PS was 1<br />

(0-2). Antecedent hematological disorders were present in 14 pts (50%).<br />

Median white cell count, hemoglobin, platelet count and bone marrow (BM)<br />

blast percentage at diagnosis were 3.5 x 109 /l (0.6 to 73.6), 7.8 g/l (6.5 to<br />

11.4), 44 x 109 /l (9 to 615), and 43% (6-82), respectively. 9 pts (32%)<br />

had complex cytogenetics. FLT3 mutations were identified in 2 (15%) <strong>of</strong><br />

13 evaluated pts. All pts were refractory to HiDAC-based induction. HiDAC<br />

was combined with an anthracycline in 16 pts (57%) and nonanthracycline<br />

in 12 pts (43%). Median time from induction to ASCT was<br />

76 days (28 to 184). Median BM blast and peripheral blast at ASCT were<br />

28% (3 to 82) and 4% (0 to 41). 21 pts (75%) had matched related donors<br />

(18 sibling and 3 haploidentical) and 7 (25%) had matched unrelated<br />

donors. Conditioning regimens were melphalan-based, busulfan-based,<br />

fludarabine-based or others in 7 (25%), 10 (36%), 8 (29%) and 3 pts<br />

(10%); respectively. Complete remission (CR) was achieved in 23 <strong>of</strong> 28 pts<br />

undergoing ASCT (CR � 82%) with median time to CR <strong>of</strong> 31 days (26 to<br />

134). 12 pts relapsed with median time to relapse <strong>of</strong> 5 months (2 to 19). 8<br />

pts remain alive with median follow up <strong>of</strong> 80 months (28 to 118). Median<br />

overall survival (OS) for the entire group is 20 months (5 to 118). In<br />

historical series <strong>of</strong> pts with AML refractory to HiDAC-based induction,<br />

salvage chemotherapies induced CR rates <strong>of</strong> 22% with median OS <strong>of</strong> 4<br />

months. Conclusions: Initial salvage with allogenic SCT is feasible and<br />

yields superior outcomes to salvage chemotherapy in primary HiDAC<br />

refractory AML pts. Randomized studies are warranted to further explore<br />

this treatment option.<br />

6546 General Poster Session (Board #15E), Mon, 1:15 PM-5:15 PM<br />

Neurotoxicity after high-dose melphalan. Presenting Author: Jose Eugenio<br />

Najera, M. D. Anderson Cancer Center Orlando, Orlando, FL<br />

Background: High dose melphalan (HDM) at 200 mg/m2 is the standard<br />

preparative regimen for patients with multiple myeloma (MM) and lightchain<br />

amyloidosis (AL) undergoing autologous hematopoietic stem cell<br />

transplantation (auto-HCT). Neurotoxicity has been seen with HDM. In this<br />

report we describe the incidence, clinical manifestations and outcome <strong>of</strong><br />

HDM- associated neurotoxicity. Methods: We performed a chart review <strong>of</strong> all<br />

patients who received HDM and auto-HCT for MM or AL between January<br />

2007 to December 2009 at the University <strong>of</strong> Texas MD Anderson Cancer<br />

Center (MDACC). HDM- associated encephalopathy was defined as altered<br />

mental status, seizure or unexplained loss <strong>of</strong> consciousness within 30 days<br />

<strong>of</strong> auto-HCT. Patients with documented hemorrhagic or embolic stroke, or<br />

metabolic abnormalities were excluded. Results: 451 patients were included.<br />

Median age at auto-HCT was 59 years (range: 35-80). Thirty<br />

patients (6.6%) had AL and 61 patients (13.5%) had a pre-transplant<br />

serum creatinine <strong>of</strong> � 1.5 mg/dl. Nine patients (2.0%) developed HDMassociated<br />

encephalopathy with a median <strong>of</strong> 13 days (range 4-22) from<br />

auto-HCT. Among patients with encephalopathy, 8 (89%) developed<br />

changes in mental status ranging from drowsiness and confusion to loss <strong>of</strong><br />

consciousness, while one patient had tonic-clonic seizures (11%). Of the<br />

affected patients there were 6 (66%) females, 8 patients (89%) � 59 years<br />

<strong>of</strong> age and only 2 patients (22%) had a creatinine clearance <strong>of</strong> � 60<br />

ml/min. One patient was dialysis-dependent. A CT scan or MRI was<br />

obtained in all 9 patients. Only one patient had imaging abnormalities<br />

reported as posterior reversible encephalopathy syndrome (PRES). Electroencephalogram<br />

(EEG) was performed on 6 patients. Epileptiform activity<br />

was seen in one patient with clinical seizures. Mild generalized slowing was<br />

noted in 2 other patients with mental status changes. Cerebrospinal fluid<br />

was obtained in 2 patients and did not show any abnormalities. Complete<br />

resolution <strong>of</strong> neurologic symptoms was seen in all patients prior to hospital<br />

discharge, and there were no deaths. Conclusions: HDM-induced encephalopathy<br />

was seen in only 2% patients, and it is associated with complete<br />

neurologic recovery without any increase in transplant-related mortality.<br />

Leukemia, Myelodysplasia, and Transplantation<br />

427s<br />

6545 General Poster Session (Board #15D), Mon, 1:15 PM-5:15 PM<br />

A phase II trial 90y-ibritumomab tiuxetan in combination with reduced<br />

intensity regimen <strong>of</strong> fludarabine (flu) and melphalan (mel) followed by<br />

allo-HCT in patients with refractory B-cell lymphoma. Presenting Author:<br />

Auayporn Nademanee, City <strong>of</strong> Hope, Duarte, CA<br />

Background: RIC allo-HCT has reduced transplant-related mortality (TRM)<br />

in patients with relapsed NHL. However, relapses do occur despite<br />

potential graft vs. lymphoma (GVL) effect. We hypothesized that adding<br />

Zevalin to Flu/Mel may improve disease control and reduce relapse post<br />

allo-HCT. Methods: Patients received In- Zevalin on day -21 followed by<br />

90 2 Y- Zevalin 0.4mCi/kg on day -14, flu 25 mg/m daily on days -9 to -5 and<br />

mel 140 mg/m2 on day -4. Rituximab (R) level was measured on Day -22<br />

and -15 and if the level was � 10 �g/ml, R was not given prior to In-Zevalin<br />

or 90Y- Zevalin to enhance biodistribution. Tacrolimus and sirolimus was<br />

used for GVHD prophylaxis. Between 10/2007 and 11/2011, 31 were<br />

treated. Median age 55 (range 27-67), median regimen �3 (range 2-8).<br />

Median time from diagnosis to HCT was 20 mo (range 5-105). Histology:<br />

DLBCL (including transformed lymphoma)�14 (45%), MCL�7 (23%),<br />

FL�5 (16%) and SLL/CLL�5 (16%). Disease status at HCT: 1CR�7,<br />

Relapse�9, �2CR�5, primary refractory �10. Fifteen had chemoresistant<br />

and 19 had FDG PET� at HCT. Donors: sib�13, URD�18. Results: All<br />

patients engrafted with the median time to ANC �500 and platelet �<br />

20,000 was 14 (range 10-28) and 13.5 days (range 11-28), respectively.<br />

There were 10 deaths from disease progression (2) infection (5) GVHD (1)<br />

and multi-organ failure (2). TRM at day �100 and at 1 yr was 6.5% and<br />

17%, Five with DLBCL relapsed between 3-7 mos. Grade II-IV acute GVHD<br />

was 65%, Grade III-IV was 16%, chronic GVHD was 65%. Fifteen became<br />

PET- at day �100 while 4 remained PET� and relapsed. Twenty-one are<br />

alive at a median followup <strong>of</strong> 24 mos (range 2-46). The 2 yrs OS and PFS<br />

was 65% (95% CI, 51-75%) and 57% (95% CI, 46-67%), respectively.<br />

Univariate analysis identified disease status predict for OS and PFS while<br />

histology predict for PFS. Conclusions: This study demonstrates the<br />

feasibility <strong>of</strong> adding Zevalin to flu/mel in the allo-HCT setting for B-cell NHL<br />

and suggest that this approach could be used to provide early disease<br />

control before GVL effect takes place. Innovative approaches should be<br />

explored in DLBCL.<br />

6547 General Poster Session (Board #15F), Mon, 1:15 PM-5:15 PM<br />

Effect <strong>of</strong> radioimmunotherapy-based conditioning for autologous stem cell<br />

transplantation on poor-risk molecular pr<strong>of</strong>iling in diffuse large B-cell<br />

lymphoma. Presenting Author: Tanya Siddiqi, City <strong>of</strong> Hope, Duarte, CA<br />

Background: Gene expression pr<strong>of</strong>iling has improved risk stratification <strong>of</strong><br />

diffuse large B-cell lymphoma (DLBCL) in the first line setting as shown by<br />

DNA microarray analysis and immunohistochemistry (IHC). 5-year overall<br />

survival (OS) for germinal center B-cell-like (GCB) DLBCL is significantly<br />

better than that for non-GCB DLBCL in newly diagnosed patients. However,<br />

in the relapsed/refractory setting, autologous stem cell transplant (ASCT)<br />

appeared to overcome the poor prognosis <strong>of</strong> cell <strong>of</strong> origin as defined by IHC<br />

using CD10, Bcl6 and MUM1 and there was no difference in survival<br />

between GCB and non-GCB groups. Ibritumomab tiuxetan (Z)-BEAM<br />

conditioning regimen for ASCT in DLBCL has produced promising response<br />

rates and progression free survival (PFS). The role <strong>of</strong> poor risk molecular<br />

markers in predicting outcome with the novel conditioning regimen<br />

Z-BEAM is unknown. Methods: We evaluated cell <strong>of</strong> origin (GCB, non-GCB)<br />

using IHC in 36 patients undergoing ASCT with Z-BEAM conditioning from<br />

2002 to 2010. Median age <strong>of</strong> the patients was 54.5 years. There were 12<br />

females and 24 males. 6 patients were in 1st CR/PR ( 17%), 12 had<br />

induction failure disease (33%), 9 were in 1st relapse (25%), 1 in 2nd relapse (3%) and 8 were in �2nd CR (22%). 27 patients had chemosensitive<br />

disease (75%) and all had prior rituximab exposure. Median number <strong>of</strong><br />

prior treatments was 2. IPI at transplant was mainly low risk (78%). 22<br />

patients had GCB (61%) and 14 (39%) had non-GCB DLBCL. Results:<br />

Median follow up is 25.6 months. 12 patients relapsed (33%) and 9 died<br />

(25%). The major cause <strong>of</strong> death is relapse/disease progression. At 2 years,<br />

median OS is 79% and median PFS is 64%. No significant difference in 2<br />

year OS and PFS is noted between the GCB and non-GCB groups (p�0.07<br />

and 0.06 respectively). Conclusions: Z-BEAM conditioning for ASCT may<br />

overcome the poor prognostic effect <strong>of</strong> non-GCB DLBCL in relapsed/<br />

refractory disease. Further evaluation <strong>of</strong> cell <strong>of</strong> origin using new markers<br />

like GCET1, LMO2 and FOXP1 could confirm these results as there seems<br />

to be better concordance <strong>of</strong> these markers with DNA microarray analysis.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


428s Leukemia, Myelodysplasia, and Transplantation<br />

6548 General Poster Session (Board #15G), Mon, 1:15 PM-5:15 PM<br />

Dose-intensive cyclophosphamide, etoposide, cisplatin reinduction for<br />

relapsed/refractory aggressive non-Hodgkin lymphoma (r/r-aNHL). Presenting<br />

Author: Jan-Willem Henning, University <strong>of</strong> Calgary and Tom Baker<br />

Cancer Center, Calgary, AB, Canada<br />

Background: Approximately 2/3 r/r-aNHL patients (pts) respond to salvage<br />

R-ICE or R-DHAP, 1/2 proceed to autologous stem cell transplantation<br />

(ASCT), and 1/3 achieve 3 year (yr) progression-free survival (PFS);<br />

however, PFS is only 20% if prior Rituximab, time to progression (TTP)�1yr,<br />

or age-adjusted International Prognostic Index (aaIPI)�2-3 [JCO 2010;28:<br />

4184-90]. Since 1995, we re-induced poor prognosis r/r-aNHL with<br />

dose-intensive Cyclophosphamide 5.25g/m2 , Etoposide 1.05g/m2 and<br />

Cisplatin 105mg/m2 (DICEP), G-CSF days (d) 14-19, and apheresis<br />

d19,20, or 21. Rituximab was added d0,7 after 2006. Methods: We<br />

retrospectively analyzed 113 consecutive transplant eligible r/r-aNHL pts<br />

[diffuse large B-cell�95, transformed�9, peripheral T-cell�6, other�3]<br />

who received one cycle <strong>of</strong> DICEP (n�93) or R-DICEP (n�20) from<br />

1995-2009. Patient characteristics included: median age�49yr (22-69);<br />

primary refractory�68; TTP�1yr�85; elevated LDH�60; ECOG 2-4�42;<br />

aaIPI 2-3�59; bulk�10cm�26. Results: Of 113 pts, 77% responded to<br />

DICEP and 90% (102) proceeded to ASCT. The median CD34� cells<br />

collected was 19x106 /kg (0.3-142). Early treatment-related mortality<br />

(TRM) occurred in 3 pts (2.7%), and 4 others developed late second<br />

cancers (MDS/AML�2). With 94 months median follow-up (26-194), 5<br />

and 10yr OS rates for all 113pts are 48% and 41%, and PFS rates are 42%<br />

and 37%, respectively. 5 year PFS rates for ASCT vs no-ASCT are 46% vs<br />

9%, for relapse aaIPI�0-1 vs aaIPI�2-3 are 53.3% vs 32.1% (p�0.01),<br />

and for TTP�1yr vs �1yr are 63.9% vs 35.2% (p�0.009). Other<br />

predictors <strong>of</strong> inferior PFS in univariate analysis were elevated LDH, ECOG<br />

2-4, no response to DICEP; however, PFS for 27 pts who failed prior<br />

Rituximab-chemotherapy (56%) was similar to other 86 pts (38%) (logrank<br />

p�0.09). Predictors <strong>of</strong> PFS and OS in multivariate analysis include:<br />

TTP�1yr, elevated LDH, bulk, no response to DICEP. Conclusions: (R)DI-<br />

CEP is an effective re-induction regimen for r/r-aNHL, leading to excellent<br />

stem cell mobilization and a high chance <strong>of</strong> proceeding to ASCT. Long-term<br />

PFS and OS rates compare favourably to reports <strong>of</strong> other re-induction<br />

regimens, and a prospective multicentre trial is warranted.<br />

6550 General Poster Session (Board #16A), Mon, 1:15 PM-5:15 PM<br />

The comparison <strong>of</strong> up-front autologous peripheral stem cell transplantation<br />

in first remission and long-lasting intensive chemotherapy protocols in<br />

highly aggressive non-Hodgkin’s lymphomas: Results <strong>of</strong> a retrospective<br />

analysis. Presenting Author: Mustafa Ozturk, Gulhane Faculty <strong>of</strong> Medicine,<br />

Departments <strong>of</strong> Medical Oncology and Bone Marrow Transplantation Unit,<br />

Ankara, Turkey<br />

Background: Optimal therapy in adult patients with Burkitt’s and lymphoblastic<br />

lymphoma are still unknown. Despite long–lasting and intensive<br />

chemotherapy protocols, post-transplant relapse is common and the<br />

chemotherapy period is time consuming. Methods: In this retrospective<br />

study, the results <strong>of</strong> induction and consolidation chemotherapies followed-by<br />

high dose therapy (HDT) and autologous peripheral stem cell<br />

transplantation (APSCT) (n�21), which was given in a time period <strong>of</strong> 3<br />

months were compared to long–lasting and intensive chemotherapy protocols<br />

(LICP) (n�39) in patients with Burkitt’s and Lymphoblastic lymphoma,<br />

without bone marrow or central nervous system involvement at<br />

presentation. Cyclophosphamide, vincristine, prednisolone, doxorubicin<br />

(Adriamycin) with or without L–asparaginase were given as induction<br />

chemotherapy, followed-by a consolidation chemotherapy with DHAP and<br />

up-front HDT. Results: More than 60% <strong>of</strong> patients in both groups were in<br />

the intermediate and high risk group according to International Prognostic<br />

Index. There was no significantly difference according to age, gender, bulky<br />

disease, stage, Burkitt’s and Lymphoblastic lymphoma ratio in both groups<br />

(p�0,05). Median follow up was 32,3 months (min-max: 0-229 months).<br />

When HDT and LICP groups were compared: CR achieved in 76,3% and<br />

82,1% <strong>of</strong> patients (p�0,05); Median PFS was 10,4 months (min-max:<br />

6,1-20,3) and 10,1 months (min-max: 6,3- 69,0) (p�0,05); One year<br />

overall survival rate was 64,7% and 74,4% and five year overall survival<br />

rate was 50,1% and 56,7% (p�0,05) respectively. Conclusions: The<br />

current study suggests that induction and consolidation chemotherapies<br />

followed by HDT and APSCT have similar CR, PFS and OS rates when<br />

compared to more time consuming LICP therapies. This provides a short<br />

period <strong>of</strong> treatment with a high rate <strong>of</strong> survival in adult patients with highly<br />

aggressive lymphomas in first remission.<br />

6549 General Poster Session (Board #15H), Mon, 1:15 PM-5:15 PM<br />

Effect <strong>of</strong> lenalidomide induction therapy on peripheral blood stem cell<br />

collection in patients undergoing autologous stem cell transplant for<br />

multiple myeloma. Presenting Author: Divaya Bhutani, Karmanos Cancer<br />

Institute, Detroit, MI<br />

Background: Autologous stem cell transplant (ASCT) remains part <strong>of</strong><br />

standard therapy for Multiple Myeloma (MM). Lenalidomide (LEN) is a<br />

newer, effective therapy for MM. It has been suggested that prior LEN<br />

therapy is associated with an increased risk <strong>of</strong> stem cell collection failure,<br />

particularly when only G-CSF is used for mobilization. Methods: We<br />

conducted a retrospective chart review <strong>of</strong> 310 consecutive MM pts who<br />

underwent pheresis to collect stem cells for first ASCT between July 1,<br />

2007 and June 30, 2011 at the Karmanos Cancer Institute. We compared<br />

differences in quantity <strong>of</strong> CD34 cells collected, days needed to collect the<br />

target number <strong>of</strong> cells (� 2.5 x 10*6 CD34� cells/kg), days to platelet and<br />

neutrophil engraftment. We also evaluated the association between CD34�<br />

cells collected and the number <strong>of</strong> cycles <strong>of</strong> LEN therapy. Results: Of 310<br />

patients, 90% were mobilized with only G-CSF initially. Patients were<br />

analyzed as two groups: LEN exposed (LEN(�); n � 128) and LEN<br />

naive(LEN(-); n � 182). Median age in both groups was 58 years. No<br />

differences in race, sex and MM stage distribution were observed between<br />

the two groups. The median number <strong>of</strong> stem cells collected in the LEN(�)<br />

group was significantly less than the LEN(-) group (6.46 vs. 7.56 x 10*6<br />

CD34 cells/kg; p� 0.0004). In addition, the median number <strong>of</strong> pheresis<br />

sessions required for adequate stem cell collection were significantly more<br />

in the LEN(�)group as compared to LEN(-) group (2 vs.1 sessions;<br />

p�0.002). In the LEN(�) group, there was a negative correlation between<br />

CD34� cells collected and the prior number <strong>of</strong> cycles <strong>of</strong> LEN (p�0.0001).<br />

There was no statistically significant excess in the number <strong>of</strong> stem cell<br />

collection failures with G-CSF in the LEN(�) group (7% vs. 4% p�0.31).<br />

All pts who failed collection after G-CSF were successfully collected with<br />

Cytoxan or Plerixafor priming. LEN exposure had no effect on post-ASCT<br />

neutrophil or platelet recovery. Conclusions: Although Lenalidomide exposure<br />

is associated with a slightly lower CD34� stem cell yield and on<br />

average an extra session <strong>of</strong> pheresis when G-CSF is used for mobilization,<br />

collection failure is uncommon and post-ASCT engraftment is normal.<br />

6551 General Poster Session (Board #16B), Mon, 1:15 PM-5:15 PM<br />

Validation <strong>of</strong> the M. D. Anderson Symptom Inventory multiple myeloma<br />

module. Presenting Author: Nina Shah, University <strong>of</strong> Texas M. D. Anderson<br />

Cancer Center, Houston, TX<br />

Background: Validated scales exist for quality <strong>of</strong> life assessment in patients<br />

with multiple myeloma (MM); however, no existing tool comprehensively<br />

assesses disease- and treatment-related symptoms in these patients. The<br />

aim <strong>of</strong> this study was to validate a module <strong>of</strong> the M. D. Anderson Symptom<br />

Inventory developed for patients with MM (MDASI-MM). The MDASI-MM<br />

includes 13 core symptoms, 6 interference items, and 7 MM-specific<br />

items: constipation, muscle weakness, diarrhea, sore mouth/throat, rash,<br />

difficulty concentrating, and bone aches. Methods: The MDASI-MM was<br />

administered to 2 cohorts <strong>of</strong> patients with MM. The first cohort (N�86)<br />

consisted <strong>of</strong> cross-sectional symptom data from patients at the beginning<br />

<strong>of</strong> induction chemotherapy or autologous hematopoietic stem cell transplant<br />

(HSCT). This cohort was used to demonstrate i) known-group validity,<br />

by detecting differences in groups by performance status; ii) concurrent<br />

validity, by correlating selected items and subscales from the MDASI-MM<br />

with those from the Short Form Health Survey (SF-12) and the EORTC-<br />

QLQC30; and iii) reliability, by computing Cronbach alpha values. The<br />

second cohort (N�53) consisted <strong>of</strong> symptom data collected at 2 points:<br />

prior to transplant and 7 days post-HSCT. This cohort was used to<br />

demonstrate the sensitivity <strong>of</strong> the MDASI-MM to detect acute worsening <strong>of</strong><br />

patients’ symptoms post-HSCT. Results: The MDASI-MM detected significant<br />

differences in symptoms and interference levels according to patients’<br />

performance status (P � .001). MDASI-MM scores correlated with those<br />

from SF-12 subscales for physical, emotional, cognitive, and social<br />

functioning (all P �.001) and with comparable items from the EORTC-<br />

QLQC30 (such as pain, fatigue, difficulty concentrating, constipation, and<br />

diarrhea, all P � .001). Cronbach alphas were 0.85 for symptom severity<br />

and 0.87 for interference. The MDASI-MM’s overall mean scores worsened<br />

between pre- and post-HSCT (1.32 vs. 2.55 (range � 0-10), P � .001).<br />

Conclusions: The MDASI-MM is a valid, reliable, and concise tool that can<br />

be used to quantitatively assess symptom severity and interference in<br />

clinical trials and care <strong>of</strong> MM patients.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


6552 General Poster Session (Board #16C), Mon, 1:15 PM-5:15 PM<br />

Secondary neutropenia (SN) after autologous hematopoietic stem cell<br />

transplantation (AHSCT) in patients (pts) with lymphoma. Presenting<br />

Author: Sunita Nathan, Rush University Medical Center, Chicago, IL<br />

Background: Plerixafor for mobilization <strong>of</strong> autologous stem cells (ASC) has<br />

increased the yield <strong>of</strong> transplanted ASC and is evolving as an important<br />

option for mobilization. A prior study reported a 10% incidence <strong>of</strong> SN after<br />

AHSCT (BMT 2009 Aug; 44(3): 175-83). Incidence and outcome <strong>of</strong> SN in<br />

the setting <strong>of</strong> Plerixafor/G-CSF (P/G), is unknown. We report the incidence<br />

and possible etiology for the development <strong>of</strong> SN in pts undergoing AHSCT<br />

for lymphoma at our institution. Methods: Data from 80 consecutive AHSCT<br />

pts over a 2 year period were reviewed. All pts were mobilized using P/G.<br />

Demographics, AHSCT indication, prior therapies (Rx), conditioning regimen<br />

(CR), engraftment and post-transplant complications were identified<br />

and collected. SN was defined as ANC �1000 after initial engraftment.<br />

Results: 80 pts underwent an AHSCT for lymphoma from 2009-11. 70 pts<br />

had evaluable data. 37 (52.86%) pts (average age 55.4 yrs) were male and<br />

33 (47.14%) pts (average age 48.3 yrs) female. 25 (35.7%) pts had �2<br />

comorbidities. Indications included relapsed/ refractory B-NHL, T-NHL<br />

and HL. 47 (67%) pts had Stage IV ds, 48.9% with BM involvement. 17<br />

(24.3%) pts had �2 Rx, 18 (25.7%) with loco-regional XRT and 3 (4.3%)<br />

with RIT. CR included BEAM, BEC and Benda-EAM �/- Rituxan. # <strong>of</strong><br />

CD34� cells given ranged; 1.77-19.99 x 10^6/kg. Neutrophil engraftment<br />

occurred at a median <strong>of</strong> 11 days. 26 (37.14%) pts developed SN<br />

possibly from PCP prophylactic antibiotics and infections. Prior BM<br />

involvement, Rx or XRT had no role. 5 (45.4%) pts with Benda-EAM CR had<br />

SN. Associated morbidity/mortality were not noted. Conclusions: We conclude<br />

that secondary neutropenia is common after autologous stem cell<br />

transplant using the Plerixafor/GCSF combination for mobilization and is<br />

higher than reported in the literature (37% vs 10%). Patients who received<br />

this combination for mobilization should be followed up closely in the<br />

post-transplant period for secondary neutropenia. About half the patients<br />

who received the Benda-EAM conditioning regimen developed secondary<br />

neutropenia warranting its use with caution outside <strong>of</strong> a clinical trial.<br />

6554 General Poster Session (Board #16E), Mon, 1:15 PM-5:15 PM<br />

An anthropometric study in children with chronic myeloid leukemia on<br />

imatinib. Presenting Author: Vamshi Krishna Reddy Goteke, Nizam’s<br />

Institute <strong>of</strong> Medical Sciences, Hyderabad, India<br />

Background: The long-term adverse effects, especially in children with<br />

chronic myeloid leukemia (CML) on imatinib are unknown. There is very<br />

little literature addressing the adverse effects on growth in children on<br />

imatinib. We analysed the effect <strong>of</strong> imatinib on anthropometry in children<br />

with CML. Methods: The records <strong>of</strong> children � 18 years with CML diagnosed<br />

at our institute between 2003 and 2011 were retrospectively analysed.<br />

Children who received imatinib for at least one year and on regular follow up<br />

were eligible for growth assessment. The data was analysed using WHO<br />

AnthroPlus v1.0.4 and SPSS.v19 s<strong>of</strong>tware and the Height for age and BMI<br />

for age/sex “z” scores were computed. Results: A total <strong>of</strong> 61 children were<br />

started on imatinib. But, only 37 children were eligible for assessment <strong>of</strong><br />

growth. Of them 3 were further excluded as the WHO AnthroPlus s<strong>of</strong>tware<br />

supported data only till 19 completed years. Median age was 12 years<br />

(range: 5 – 17). 17 children were in the prepubertal age (5-11years) at<br />

commencement <strong>of</strong> imatinib. The mean duration <strong>of</strong> imatinib therapy was<br />

41.24 months (range: 12–91). The overall z- scores for height for age<br />

(HAZ) on follow up were significantly (p �0.029) lower, compared to<br />

baseline. However when analysed according to age groups, the HAZ was<br />

found significantly (p�0.005) lower among 5-11 year age group compared<br />

to age group <strong>of</strong> �12 years. No significant differences were observed in BMI<br />

for age/sex z scores by age groups, over the period. Conclusions: Imatinib<br />

appears to cause growth retardation in prepubertal children. Further follow<br />

up may shed more light on the degree <strong>of</strong> stunting and the deficit in the final<br />

adult height.<br />

Leukemia, Myelodysplasia, and Transplantation<br />

429s<br />

6553 General Poster Session (Board #16D), Mon, 1:15 PM-5:15 PM<br />

Cost-effectiveness analysis <strong>of</strong> bendamustine plus rituximab versus CHOP-R<br />

in treatment-naive patients with mantle cell (MCL) and indolent lymphomas<br />

(IL). Presenting Author: Wayne Su, United BioSource Corporation,<br />

Bethesda, MD<br />

Background: To assess the cost-effectiveness <strong>of</strong> bendamustine plus rituximab<br />

(B-R) vs cyclophosphamide, doxorubicin, vincristine, prednisone,<br />

and rituximab (CHOP-R) for treatment-naive patients with MCL or IL from a<br />

US healthcare payer perspective. Methods: A discrete event simulation was<br />

developed for a mixed population <strong>of</strong> patients with MCL or IL. Two arms were<br />

simulated, each containing 1000 identical MCL or IL patients treated with<br />

B-R or CHOP-R. Input data for baseline characteristics, overall response,<br />

and risks for treatment-related adverse events (AEs) and infections were<br />

obtained from the NHL 1-2003 trial (n�549, primarily stage IV MCL and<br />

IL); gaps were filled by consultation with experts. Direct medical costs and<br />

utilities were estimated based on US databases and published literature.<br />

Costs and benefits were discounted at 3% per annum. Base case model<br />

predictions were performed by selecting regression models that had a best<br />

fit to progression free survival (PFS). Robustness <strong>of</strong> these models was<br />

evaluated by testing other models with reasonably good fit. Results: In the<br />

base case, model predicts longer PFS for B-R than for CHOP-R in MCL<br />

(average <strong>of</strong> 49.8 vs 28.6 months) and IL (67.9 vs 51.0). Quality-adjusted<br />

life-years (QALYs) per patient were higher for B-R than CHOP-R for MCL<br />

(3.51 vs 2.68) and IL (4.42 vs 3.58). For MCL, total per-patient costs were<br />

$115,191 for B-R and $100,261 for CHOP-R; for IL, respective costs were<br />

$134,814 and $110,065, resulting in incremental cost-effectiveness<br />

ratios (ICERs) for B-R vs CHOP-R <strong>of</strong> $18,161 per QALY for MCL and<br />

$29,549 for IL (ICER�$50,000 to be cost-effective). Higher complete<br />

and partial response rates for B-R than for CHOP-R impacted subsequent<br />

treatment costs, which were lower for B-R by $21,632 for MCL and<br />

$24,961 for IL, as well as AE costs, lower by $10,113 and $10,570,<br />

respectively. The model results were robust with respect to the use <strong>of</strong><br />

alternative regression models for PFS estimation. Conclusions: In patients<br />

with MCL or IL, the model demonstrates that B-R is a cost-effective<br />

alternative to CHOP-R. Alternative regression models confirmed robustness<br />

<strong>of</strong> results. Support: Teva Pharmaceutical Industries Ltd.<br />

6555 General Poster Session (Board #16F), Mon, 1:15 PM-5:15 PM<br />

Prevalence <strong>of</strong> autoimmune manifestations in patients with large granular<br />

lymphocytic leukemia: Exploratory analysis <strong>of</strong> a series <strong>of</strong> consecutive<br />

patients. Presenting Author: Bruno Bockorny, Department <strong>of</strong> Medicine,<br />

University <strong>of</strong> Connecticut, Farmington, CT<br />

Background: Review <strong>of</strong> literature suggests certain autoimmune abnormalities<br />

to accompany LGL leukemia. The present study was set to identify the<br />

prevalence <strong>of</strong> autoimmune phenomena in LGL leukemia patients and<br />

compare it with same in general population. Methods: We conducted both<br />

retrospective and prospective analyses in a series <strong>of</strong> consecutive patients<br />

(n�11) with LGL leukemia that had been followed in outpatient setting.<br />

Median age was 71 years ( � 14 years). The presence <strong>of</strong> associated<br />

autoimmune disorders in our cohort was compared to the published<br />

prevalence <strong>of</strong> these conditions in general population. Statistical analysis:<br />

The obtained values were tested for statistical significance using Fisher’s<br />

exact test for small number <strong>of</strong> observations (95% confidence); a p value �<br />

0.05 was considered significant. Results: A total <strong>of</strong> 45% patients in our<br />

study were diagnosed with autoimmune conditions. Identified autoimmune<br />

disorders included rheumatoid arthritis [RA] (n�3), Hashimoto thyroiditis<br />

(n�3), Felty syndrome (n�1), aplastic anemia (n�1), pernicious anemia<br />

(n�1), and leukocytoclastic vasculitis (n�1). Statistical analysis showed<br />

the following significance: Hashimoto thyroiditis (p�0.044), RA (p�0.003),<br />

Felty syndrome (p�0.001), aplastic anemia (p�0.001), pernicious anemia<br />

(p�0.001), and leukocytoclastic vasculitis (p�0.001). Statistical<br />

difference was also noted for autoimmune serologic abnormalities, with<br />

ANA positivity present in 63.6% <strong>of</strong> the cases (p� 0.001) and RF present in<br />

50% <strong>of</strong> the patients (p�0.005). Conclusions: Nearly half <strong>of</strong> LGL leukemia<br />

patients in our cohort had autoimmune conditions, thus significantly<br />

exceeding the overall prevalence in general population. Statistically significant<br />

differences were recorded for RA, Hashimoto thyroiditis, Felty syndrome,<br />

aplastic anemia, pernicious anemia, leukocytoclastic vasculitis,<br />

ANA and RF positive serologies. The prevalence <strong>of</strong> Hashimoto thyroiditis<br />

and ANA positivity in our patient cohort was significantly higher than the<br />

one in existing literature. Given the relatively small size <strong>of</strong> the analyzed<br />

cohort, larger studies are expected to confirm our findings.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


430s Leukemia, Myelodysplasia, and Transplantation<br />

6556 General Poster Session (Board #16G), Mon, 1:15 PM-5:15 PM<br />

Maintenance therapy with arsenic after induction in an alternative and<br />

long-term case for the prevention <strong>of</strong> recurrence <strong>of</strong> acute promyelocytic<br />

leukemia during the period between November 2005 and December 2011.<br />

Presenting Author: Mozaffar Aznab, Kermanshah University <strong>of</strong> Medicine,<br />

Kermanshah, Iran<br />

Background: Arsenic trioxide has been used in the first line treatment <strong>of</strong><br />

acute promyelocytic leukemia and also for recurrence after ATRA and<br />

chemotherapy. In this study, it we used it in induction <strong>of</strong> remission and<br />

maintenance therapy Methods: Between November 2005 to December<br />

2011, 42 patients admitted in our department with APL diagnosis. There<br />

were 27 male and 15 women with a median age <strong>of</strong> 28 years. Arsenic<br />

trioxide started at 0.15 mg/kg intravenous infusion till patient’s bone<br />

marrows achieve to complete remission. Then, after 28 days rest, we did<br />

consolidation with Arsenic trioxide with the same dosage for 28 more days.<br />

We continued treatment with 14 days courses <strong>of</strong> Arsenic trioxide every 3-4<br />

months for 2 years, as maintenance therapy Results: Four patients died<br />

during the first 20 days <strong>of</strong> treatment. Thirty-eight patients achieved to<br />

remission. Two patients refused to continue treatment after achieving to<br />

remission and excluded from this study. Thirty-six patients finished whole<br />

treatment. After a median follow-up <strong>of</strong> 54 months, 4 patients died due to<br />

disease relapse and one patient relapsed and initiated treatment with<br />

Arsenic and ATRA. Five patients faced leukocytosis over 100,000/ml. In<br />

these cases we were obligated to discontinue Arsenic for 3-4 days and did<br />

chemotherapy by Danourobicin for 2 days. Totally 8 patients died during<br />

remission induction and long-term follow up. One year, 3 and 5 years RFS<br />

were 97%, 87.1% and 79.4 respectively and we didn’t observe any relapse<br />

for patients who remained in remission after 5 years. Finally, 31 patients<br />

are alive and free <strong>of</strong> disease. The overall survival was 79.5% for our cohort.<br />

Conclusions: Arsenic trioxide is an effective treatment as the first line<br />

therapy for new cases <strong>of</strong> APL and long term therapy with will reduce the risk<br />

<strong>of</strong> diseases recurrence without any major toxicity in long time.<br />

6558 General Poster Session (Board #17A), Mon, 1:15 PM-5:15 PM<br />

Response rates in patients with relapsed/refractory acute myeloid leukemia<br />

with FLT3-ITD mutation using 5-azacitadine plus sorafenib. Presenting<br />

Author: Mona Lisa Alattar, University <strong>of</strong> Texas at Houston Health Science<br />

Center, Houston, TX<br />

Background: The outcome <strong>of</strong> patients with relapsed acute myeloid leukemia<br />

(AML) with FLT3-ITD mutation is poor. Sorafenib is an inhibitor <strong>of</strong> FLT-3<br />

kinase activity in nanomolar concentrations. We conducted this phase II<br />

study to evaluate the efficacy and tolerability <strong>of</strong> the combination <strong>of</strong><br />

Sorafenib and 5-Aza for the treatment <strong>of</strong> patients with refractory or<br />

relapsed AML with the particular emphasis on those with FLT3-ITD<br />

mutation. Methods: Patients were eligible if they had relapsed or refractory<br />

AML and were 18 years <strong>of</strong> age or older. They received 5-Aza 75 mg/m2 daily<br />

x 7 together with Sorafenib 200 mg BID X 28 days; cycles were repeated in<br />

approximately 1-month intervals. Responses were assessed after first cycle<br />

and then at the discretion <strong>of</strong> the treating physician at the time <strong>of</strong> the best<br />

peripheral blood response. Results: 32 patients with AML with a median age<br />

<strong>of</strong> 61 years (range, 24-87) were enrolled. They included 13 (41%) with<br />

diploid cytogenetics, 9 (28%) with complex cytogenetics, and 10 (31%)<br />

with miscellaneous chromosomal abnormalities. FLT-3-ITD was detected<br />

prior to the initiation <strong>of</strong> treatment in 30/32 patients. Patients had received<br />

a median <strong>of</strong> 2 prior treatments (range, 1-6). 12 (38%) patients had<br />

received �3 prior regimens and 11 had failed therapy with a FLT3 kinase<br />

inhibitor (6 with AC220, 1 with PKC412, and 5 with sorafenib, either as<br />

monotherapy or with plerixafor); 2 had failed 2 prior FLT3 inhibitors. 10<br />

patients were inevaluable as they never received therapy or discontinued it<br />

before response assessment at one month. The overall CR/CRi rate among<br />

the 22 evaluable patients is 50%, including 9 (41%) with CRi and 2 (9%)<br />

with CR; with a median <strong>of</strong> 3 (range, 1-9) treatment cycles. The median time<br />

to achieving CR/CRi was 3 months (range, 1-4) and the median duration <strong>of</strong><br />

CR/CRi was 3 months (range, �1–5.5). Three patients proceeded to<br />

allogeneic stem cell transplant. The most common study drug-related<br />

adverse events were rash and fatigue with no deaths attributable to study<br />

medications. Conclusions: Combination <strong>of</strong> 5-Aza and Sorafenib is promising<br />

for the treatment <strong>of</strong> patients with relapsed and refractory AML with<br />

FLT-3-ITD mutation and may allow them to proceed to stem cell transplant.<br />

6557 General Poster Session (Board #16H), Mon, 1:15 PM-5:15 PM<br />

Effect <strong>of</strong> ROR1-targeting small molecules on chronic lymphocytic leukemia<br />

(CLL) cells. Presenting Author: Hakan Mellstedt, Karolinska University<br />

Hospital Solna, Stockholm, Sweden<br />

Background: There is a great interest to develop targeting drugs for CLL,<br />

both MAbs and small molecules, to improve the outcome for the disease.<br />

ROR1, a receptor tyrosine kinase, is overexpressed on CLL cells but not on<br />

normal cells. ROR1 is constitutively phosphorylated and siRNA transfection<br />

induces leukemic cell death. The aim <strong>of</strong> the study is to produce small<br />

molecules inhibiting the cytoplasmic tyrosine kinase activity <strong>of</strong> ROR1,<br />

which could induce specific killing <strong>of</strong> leukemic cells. Methods: A highthroughput<br />

screening assay in 384-format has been developed measuring<br />

phosphorylation <strong>of</strong> a substrate peptide by human recombinant intracellular<br />

ROR-1 kinase domain. A collection <strong>of</strong> 80.000 small molecules has been<br />

screened generating two chemical series. Novel leads have been synthesized<br />

and structure-activity-relationship has been developed using the<br />

assay. Results: Three compounds were selected (KAN0173631,<br />

KAN0438063, KAN0438175T). Freshly isolated leukemic CLL cells were<br />

used as targets in addition to PBMC from healthy donors to test cytotoxicity.<br />

The three compounds induced specific apoptosis <strong>of</strong> CLL cells (Annexin V/PI<br />

and MTT) both at 24 and 48h. Efficacy index (EI), i.e. killing <strong>of</strong> leukemic<br />

cells in relation to normal PBMC was favourable. The most promising drug<br />

KAN0438063 had an EI <strong>of</strong> 40 i.e. killed 40 times more CLL cells than<br />

PBMC at an IC50 <strong>of</strong> 10�M. The compounds induced PARP cleavage and<br />

cleavage <strong>of</strong> caspases 8 and 9 as well as down regulation <strong>of</strong> Mcl-1 and Bcl-xl<br />

(Western Blot). ROR1 was dephosphorylated (Western Blot). The selective<br />

apoptotic effect was compared to other small molecules targeting non-<br />

ROR1 structures in CLL (PCI-32765, CAL-101, R406, R788, STK-<br />

156485, STK-156133) and our compounds were significantly more<br />

effective (EI) (p�0.001). Conclusions: We have developed effective and<br />

selective series <strong>of</strong> compounds targeting ROR1 with promising ADMET<br />

properties. ROR1 targeting small molecules might also be effective for<br />

other tumor cells expressing ROR1 as has been noted for e.g. pancreatic<br />

carcinoma cells. Our model molecules will be further optimized and tested<br />

in animal tumor models. These molecules represent the first small<br />

molecules targeting ROR1 – a “survival factor” in CLL.<br />

6559 General Poster Session (Board #17B), Mon, 1:15 PM-5:15 PM<br />

Post hoc analysis <strong>of</strong> relationship between baseline white blood cell count<br />

and survival outcome in a randomized phase III trial <strong>of</strong> decitabine in older<br />

patients with newly diagnosed acute myeloid leukemia. Presenting Author:<br />

Chris Arthur, Royal North Shore Hospital, St. Leonards, Australia<br />

Background: In a large phase III trial (N�485), patients (pts) �65y with<br />

newly diagnosed acute myeloid leukemia (AML) received 1-h IV infusion <strong>of</strong><br />

decitabine (DAC) 20 mg/m2 for 5 consecutive days every 4 wk or treatment<br />

choice (TC) with supportive care or cytarabine 20 mg/m2 subcutaneous<br />

injection for 10 consecutive days every 4 wk (NCT00260832; Kantarjian et<br />

al. JCO, in press). Enrolled pts had white blood cell (WBC) count �40<br />

x109 /L; baseline WBC counts were relatively low (median [range] DAC: 3.1<br />

x109 /L [0.3-127.0]; TC: 3.7 x109 /L [0.5-80.9]). This post hoc analysis<br />

assessed baseline WBC count and survival outcome. Methods: Overall<br />

survival (OS) and progression-free survival (PFS) were summarized by<br />

baseline WBC subgroups (�1, 1-5, �5 x109 /L). Results: Mature survival<br />

data (2010) were based on intent-to-treat (ITT) population (446 deaths:<br />

TC, n�227; DAC, n�219). OS was 5.0 mo for TC vs 7.7 mo for DAC<br />

(nominal P�.037). For each WBC subgroup, differences in OS for TC vs<br />

DAC were not significant (NS), but hazard ratios (HR) favored DAC for all<br />

subgroups (Table). There was a significant difference in PFS in favor <strong>of</strong> DAC<br />

for patients with baseline WBC 1–5 x109 /L (P�.005; HR�0.67) and �5<br />

x109 /L (P�.027; HR�0.71). Conclusions: These data are consistent with<br />

overall results, with a trend toward improved outcome with DAC regardless<br />

<strong>of</strong> baseline WBC count in older pts with newly diagnosed AML. Further<br />

analyses are warranted.<br />

Outcome WBC x10 9 /L<br />

TC<br />

Median, a mo<br />

Event/n<br />

OS All pts 5.0<br />

227/243<br />

�1 4.9<br />

20/22<br />

1-5 5.9<br />

106/115<br />

�5 4.3<br />

94/99<br />

PFS All pts 2.1<br />

221/243<br />

�1 2.2<br />

20/22<br />

1-5 2.9<br />

110/115<br />

�5 2.1<br />

98/99<br />

DAC<br />

Median, a mo<br />

Event/n<br />

7.7<br />

219/242<br />

8.6<br />

22/25<br />

9.5<br />

104/119<br />

6.3<br />

88/93<br />

3.7<br />

221/242<br />

5.5<br />

24/25<br />

3.8<br />

111/119<br />

3.4<br />

89/93<br />

Nominal P value b<br />

HR (95% CI) c<br />

P�.037<br />

HR�0.82 (0.68, 0.99)<br />

P�NS<br />

HR�0.85 (0.42, 1.70)<br />

P�NS<br />

HR�0.78 (0.58, 1.03)<br />

P�NS<br />

HR�0.82 (0.60, 1.10)<br />

P�.003<br />

HR�0.75 (0.62, 0.91)<br />

P�NS<br />

HR�1.05 (0.56, 1.97)<br />

P�.005<br />

HR�0.67 (0.51, 0.89)<br />

P�.027<br />

HR� 0.71 (0.53, 0.97)<br />

a Kaplan-Meier product limit estimates. b 2-sided log-rank test. c Stratified Cox regression<br />

model (age and cytogenetic risk as strata; treatment group as covariate). HR �1 means<br />

advantage for DAC.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


6560 General Poster Session (Board #17C), Mon, 1:15 PM-5:15 PM<br />

Increased risk <strong>of</strong> MDS/AML post radiation treatment for breast cancer:<br />

Analysis <strong>of</strong> SEER data 2001-2008. Presenting Author: Henry G. Kaplan,<br />

Swedish Cancer Institute, Seattle, WA<br />

Background: Ionizing radiation is a known cause <strong>of</strong> myeloid leukemia, but it<br />

is not known whether therapeutic doses for breast cancer (BC) pose risk. We<br />

hypothesized that BC radiation treatment is associated with increased risk<br />

<strong>of</strong> myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML))<br />

as seen in a previously conducted institutional cohort study. Methods: We<br />

used 2001-2008 Surveillance, Epidemiology and End Results (SEER)<br />

database records to identify a cohort <strong>of</strong> first primary BC patients treated<br />

with radiation who were unlikely to be treated with chemotherapy (stage 0<br />

and estrogen receptor positive (ER�) stage I). We identified subsequent<br />

MDS/AML diagnoses in the BC cohort, using SEER to query appropriate<br />

ICD-O-3 codes. We compared observed MDS/AML rates in the BC cohort to<br />

expected rates, estimated for first primary MDS/AML in the entire population,<br />

and calculated observed/expected rate ratios with 95% confidence<br />

intervals (CI). Results: We estimated an increased overall risk <strong>of</strong> MDS/AML<br />

in the breast cancer cases treated with radiation compared to the general<br />

population (RR�4.23, 95% CI 3.41, 5.18). The relative risk was higher for<br />

stage I ER� breast cancer cases �65 years <strong>of</strong> age (RR�7.75, 95% CI<br />

5.18, 11.05) than for stage 0 cases �65 years <strong>of</strong> age (RR�3.65, 95% CI<br />

1.57, 7.05). Among women 65 years and older, there was a more modest<br />

increased risk, only seen for stage 1 ER� breast cancer cases (RR�1.52)<br />

(table). Conclusions: Our results suggest that radiation treatment for breast<br />

cancer is associated with increased risk <strong>of</strong> MDS/AML. The effect appears to<br />

be age-dependent.<br />

Expected and observed MDS/AML incidence rates in breast cancer cases<br />

with radiation treatment (per 100,000 person years).<br />

Expected<br />

rate<br />

Person<br />

years<br />

Observed rate<br />

(n�cases)<br />

Rate ratio<br />

(95% CI)<br />

Stage 0, age


432s Leukemia, Myelodysplasia, and Transplantation<br />

6564 General Poster Session (Board #17G), Mon, 1:15 PM-5:15 PM<br />

Prognostic implication <strong>of</strong> monosomal karyotype in patients with acute<br />

myeloid leukemia who received allogeneic hematopoietic cell transplantation.<br />

Presenting Author: Yunsuk Choi, Department <strong>of</strong> Hematology, Asan<br />

Medical Center, University <strong>of</strong> Ulsan College <strong>of</strong> Medicine, Seoul, South<br />

Korea<br />

Background: Monosomal karyotype (MK), defined as at least two autosomal<br />

monosomies or one single autosomal monosomy with one or more structural<br />

cytogenetic abnormalities, has been associated with worse outcomes in<br />

acute myeloid leukemia (AML). We evaluated the prognostic impact <strong>of</strong> MK<br />

on outcomes after allogeneic hematopoietic cell transplantation (allo-HCT)<br />

in AML. Methods: We retrospectively analyzed 169 adult patients with AML,<br />

who received allo-HCT in the first complete remission (CR) between 2000<br />

and 2009. All patients had cytogenetic results at the diagnosis <strong>of</strong> AML.<br />

Patients were classified as having good, intermediate, or poor risk cytogenetics<br />

according to the NCCN guideline, and MK status was also determined.<br />

Results: MK was observed in 12 patients (7.1%); <strong>of</strong> whom, 11 patients also<br />

had complex karyotype (CK). Compared to patients without MK, those with<br />

MK had significantly lower overall survival (OS) (62.2% vs. 0%) and event<br />

free survival (EFS) (58.9% vs. 0%), and higher relapse probability (RP)<br />

(22.9% vs. 66.7%) at 5 years (all, p�0.001). 22 patients with CK also<br />

showed inferior outcomes, but CK with MK was associated with more<br />

inferior outcomes than CK without MK. Multivariate analysis showed that<br />

MK had a significantly adverse impact on OS (hazard ratio [HR], 5.192;<br />

p�0.001), EFS (HR, 4.531; p�0.001), and RP (HR, 6.558; p�0.001)<br />

after allo-HCT (Table). Conclusions: MK is a major prognostic factor<br />

predicting extremely worse outcomes in AML patients who underwent<br />

allo-HCT in the first CR.<br />

Multivariate analysis.<br />

OS EFS RP<br />

HR (95% CI) p HR (95% CI) p HR (95% CI) p<br />

WBC count, >60<br />

(x103 /�L)<br />

1.990 (1.107-3.576) 0.021 1.885 (1.074-3.307) 0.027 -<br />

Time from diagnosis to 2.621 (1.216-5.650)<br />

HCT, >250, days<br />

Cytogenetics<br />

0.014 2.378 (1.115-5.071) 0.025 2.749 (1.140-6.629) 0.024<br />

Good 0.755 (0.231-2.463) 0.634 (0.196-2.054) 0.447 0.766 (0.180-3.248) 0.717<br />

Intermediate 1 1 1<br />

Poor, CK- 1.286 (0.576-2.872) 0.539 1.098 (0.496-2.435) 0.817 3.199 (1.338-7.648) 0.009<br />

Poor, CK�, MK- 1.686 (0.703-4.043) 0.242 2.171 (1.014-4.648) 0.046 3.525 (1.506-8.252) 0.004<br />

Poor, CK�,MK� 5.192 (2.552-10.561) �0.001 4.531 (2.255-9.108) �0.001 6.558 (2.739-15.703) �0.001<br />

6566 General Poster Session (Board #18A), Mon, 1:15 PM-5:15 PM<br />

Bendamustine retreatment <strong>of</strong> CLL in the outpatient setting. Presenting<br />

Author: Georg Guenther, Oncological Medical Practice, Potsdam, Germany<br />

Background: Several studies have proven that bendamustine is a highly<br />

active drug in the therapy <strong>of</strong> CLL (Knauf et al. J Clin Oncol. 2009;27:4378-<br />

84; Fischer et al. J Clin Oncol. 2011;29:3559-3556). Nevertheless, only<br />

few data exist about the real-life use and efficacy <strong>of</strong> bendamustinetherapies<br />

in the non-trial setting. The project group <strong>of</strong> internal oncology<br />

(p.i.o.) therefore implemented a registry for patients in the routine use <strong>of</strong><br />

bendamustine in the therapy <strong>of</strong> CLL (Sauer et al. Onkologie 2010;33(suppl<br />

6):12). Since 2008 a total <strong>of</strong> 616 patients have been registered, out <strong>of</strong><br />

which 473 have been thoroughly documented. To evaluate whether a<br />

retreatment with bendamustine in relapsed CLL is still active and well<br />

tolerable we present comparative data on therapies in patients with (group<br />

A) or without (group B) bendamustine pretreatment. Methods: 228 documented<br />

patients in relapsed situation were retrospectively appointed to one<br />

<strong>of</strong> two groups (see table below). Results: In Group A the ORR was 80 % and<br />

the median PFS lasted 15.6 month. In the patients without prior bendamustine<br />

treatment the ORR was 84% and the median PFS lasted 21.2 month.<br />

In both groups the median OS has not been reached yet. The grade 3/4<br />

toxicities were mostly hematologic and comparable in both groups. Similar<br />

grade 3/4 toxicities were observed for neutropenia (about 25% <strong>of</strong> the<br />

patients), thrombocytopenia (16%) and infections (5%). Conclusions:<br />

Bendamustine is broadly used in the routine treatment <strong>of</strong> CLL. Bendamustine-therapies<br />

show impressionable high activity and tolerability, even in an<br />

advanced-line context with prior exposition to bendamustine. The treatment<br />

results are comparable to results <strong>of</strong> clinical trials and underline the<br />

quality and feasibility <strong>of</strong> bendamustine in the outpatient treatment.<br />

Nevertheless, it is to note that this is a registry and that both groups<br />

therefore are not composed <strong>of</strong> randomized patient-populations (cf. ECOG,<br />

patients in lines and ratio <strong>of</strong> rituximab combinations).<br />

Group A Group B<br />

n 83 145<br />

Gender m/w 53/30 88 /57<br />

Median age (range) 51-84 (74) 45-93 (72)<br />

Ratio B/B�rituximab in % 57/43 51/49<br />

ECOG 0/1/2 in % 11/64/25 21/64/15<br />

Binet A/B/C in % 4/54/42 4/45/51<br />

Line 2./3./4./5./6.� in % 30/30/22/13/5 56/26/10/6/2<br />

Med. B-dose-intensity over 4 weeks 151,9 mg/m² 151,3 mg/m²<br />

6565 General Poster Session (Board #17H), Mon, 1:15 PM-5:15 PM<br />

Predicting outcome based on minimal residual disease (MRD) using<br />

multiparameter flow cytometry (FC) in acute myeloid leukemia (AML)<br />

patients (Pts). Presenting Author: Sagar D. Sardesai, Roswell Park Cancer<br />

Institute, Buffalo, NY<br />

Background: Most AML pts will experience relapse caused by persistent<br />

leukemic cells during complete remission (CR). The aim <strong>of</strong> our study was to<br />

predict outcome in AML pts in CR by assessing their MRD using standard<br />

4-colour FC panels available at our institute. Methods: We queried our AML<br />

database between 1/2004 to 10/2010 for newly diagnosed untreated AML<br />

pts �18 years <strong>of</strong> age who achieved CR after one induction regimen.<br />

Treatment included 7�3 or similar intensive approaches. The gating<br />

strategy used a series <strong>of</strong> Boolean regions that best defined the diagnostic<br />

abnormal population based on its expression patterns using three 4-colour<br />

FC panels (F7-CD38,CD10,CD19,CD34; F9-C11b,CD33,CD13,CD34; F38-<br />

CD15,CD56,CD7,CD34). The same regions were applied to post-induction<br />

samples, and the number <strong>of</strong> residual events was determined. Results: 140<br />

AML patients with a median age <strong>of</strong> 64 (range 24-93) years, including 74<br />

females (52.8%) were analyzed; 67 (47.8%) patients relapsed. Eightyeight<br />

(62.8%) pts were CD34-positive (CD34�) at diagnosis. Normal<br />

karyotype was detected in 27 (30.7%): FLT-3 ITD was detected in 4/23<br />

(17.4%) and NPM-1 mutation was detected in 1/15 (0.07%) samples.<br />

Median follow-up for CD34� pts was 17.9 months. Forty-two (30%) pts<br />

underwent stem cell transplantation (SCT) in first CR: 31 (22.1 %) were<br />

allogeneic and 11 (7.9%) autologous. In multivariate analysis, a detectable<br />

MRD in any <strong>of</strong> the 3 panels at or beyond week 16 among CD34� AML pts in<br />

CR was significantly associated with inferior relapse free survival (F7:<br />

P�0.035 , F9: P�0.027, F38: P�0.042). In addition, MRD � week 16<br />

using panel F9 was also significantly associated with inferior overall<br />

survival (P�0.0224). In pair-wise comparison, those who were MRDnegative<br />

� week 16 did not benefit from SCT compared to the non-SCT<br />

group. Similar analyses among CD34– AML pts did not achieve statistical<br />

significance. Conclusions: MRD� by FC in CD34� AML is an independent<br />

prognostic factor and can identify pts who may benefit from SCT/more<br />

intensive therapy to maintain remission. However, the value <strong>of</strong> studying<br />

MRD in CD34– AML requires further consideration. More effort is needed<br />

to identify stem cells in CD34– AML.<br />

6567 General Poster Session (Board #18B), Mon, 1:15 PM-5:15 PM<br />

Effect <strong>of</strong> epitopes derived from the mutated region <strong>of</strong> cytoplasmatic<br />

nucleophosmine 1 (NPM1) on CD4� and CD8� T-cell responses in<br />

patients with acute myeloid leukemia. Presenting Author: Jochen Greiner,<br />

Department <strong>of</strong> Internal Medicine III, University <strong>of</strong> Ulm, Ulm, Germany<br />

Background: Mutations <strong>of</strong> the nucleophosmin gene (NPM1mut) are one <strong>of</strong><br />

the most frequent molecular alterations in AML and constitute an important<br />

prognostic marker. The impact <strong>of</strong> NPM1mut on leukemogenesis and<br />

progression remains to be elucidated. Immune responses against NPM1mut<br />

might contribute to the favourable prognosis <strong>of</strong> AML patients with<br />

NPM1mut. Therefore, we examined T cell responses against NPM1mut.<br />

Methods: NPM1 wildtype as well as NPM1mut were screened for HLA-<br />

A*0201 binding T cell epitopes with the help <strong>of</strong> different algorithm<br />

programs. Ten peptides with most favourable characteristics were tested<br />

with ELISpot analysis for interferon-� and granzyme B in 33 healthy<br />

volunteers and 30 AML patients. Tetramer assays against most interesting<br />

epitopes were performed and chromium release assays were used to show<br />

the cytotoxicity <strong>of</strong> peptide-specific CD8� T cells. Moreover, HLA-DRbinding<br />

epitopes were used to test the role <strong>of</strong> CD4� T cells in NPM1<br />

immunogenicity. Results: Two epitopes (#1 and #3) derived from NPM1mut<br />

induced CD8� T cell responses in a high frequency. In healthy volunteers,<br />

immune responses were detected in 39%/18% against #1 and #3, and in<br />

33%/44% <strong>of</strong> NPM1mut AML patients against #1 and #3. NPM1-peptide<br />

primed effector T cells showed specific lysis <strong>of</strong> pulsed T2 cells as well as<br />

leukemic blasts in chromium release assays. In tetramer assays a significant<br />

CD8� T cell population could be detected. To obtain a robust and<br />

continuous T cell reaction, the help <strong>of</strong> CD4� T cells is indispensable.<br />

Therefore, we investigated the increase <strong>of</strong> CD8� T cell responses by the<br />

activation <strong>of</strong> CD4� T cells stimulated with longer peptides called overlapping<br />

peptides (OL). Potent HLA-DR epitopes were predicted and several<br />

favourable peptides (OL 1 to 8) were synthesized. OL8 showed favourable<br />

results to activate both CD8� and CD4� T cells. Conclusions: Taken<br />

together, NPM1mut represents a candidate for immunotherapeutic approaches<br />

and we hypothesize that it is also potentially involved in<br />

immunogenic rejection <strong>of</strong> NPM1mut leukemic blasts. Therefore, NPM1mut<br />

is a promising target structure for specific immunotherapies in AML<br />

patients.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


6568 General Poster Session (Board #18C), Mon, 1:15 PM-5:15 PM<br />

Incidence <strong>of</strong> second and secondary malignancies in patients with CLL: A<br />

single institution experience. Presenting Author: Samir Dalia, H. Lee<br />

M<strong>of</strong>fitt Cancer Center & Research Institute, Tampa, FL<br />

Background: Patients with chronic lymphocytic leukemia (CLL) have a<br />

higher incidence <strong>of</strong> second malignancies than the general population with<br />

one study showing the risk at 2.2 times the general popualtion. The<br />

increased incidence is thought to be due to immunosupression which<br />

results in decreased cell surveillance and proliferation <strong>of</strong> malignant cells.<br />

Our study aims to present the rate <strong>of</strong> second malignancies by cancer type in<br />

patients with CLL at our institution. Methods: The M<strong>of</strong>fitt Cancer Center<br />

Total Cancer Care (TCC) database was used to identify patients who had a<br />

diagnosis <strong>of</strong> CLL between January 1993-December 2009. Individual<br />

charts were reviewed to confirm the diagnosis <strong>of</strong> CLL, collect demographic<br />

data, and to assess for the presence <strong>of</strong> a second malignancy under an IRB<br />

approved protocol. A second malignancy was defined as another malignancy<br />

or transformation <strong>of</strong> CLL reported in the medical record. Second<br />

malignancy data was placed in three categories; skin cancers, solid tumor<br />

malignancy, and hematologic malignancy. Results: 546 CLL patients were<br />

included in the study. Median age was 62.5 years. 84 (43%) were Stage 0<br />

and 62 (32%) were Stage 1 RAI at diagnosis indicating earlier disease. 266<br />

(49%) patients had a second or secondary malignancy. A total <strong>of</strong> 304<br />

cancers were identified. 14% <strong>of</strong> patients had more than one malignancy.<br />

Melanoma was identified in 44 (16.5%) patients and non-melanoma skin<br />

cancer was identified in 54 (20%). Lung cancer was identified as the most<br />

frequent solid tumor malignancy with 36 (13.5%) cases, followed by<br />

prostate (35), breast (21), colorectal (15), and bladder (14). 10 patients<br />

had a Richter’s transformation <strong>of</strong> their CLL. 26 patients developed either<br />

myelodysplastic syndrome or acute myelogenous leukemia. Conclusions:<br />

Second malignancies are frequent in CLL patients. Immunosupression,<br />

increased UV light exposure, longer life expectancy in low risk CLL, and<br />

tertiary cancer center referral bias are likely reasons for these increased<br />

rates. Further research is needed to identify the precise mechanism which<br />

cause patients with CLL to have higher rates <strong>of</strong> second malignancies and to<br />

identify if there is an increased risk <strong>of</strong> a specific type <strong>of</strong> malignancy in<br />

patients with CLL.<br />

6570 General Poster Session (Board #18E), Mon, 1:15 PM-5:15 PM<br />

Phase II study <strong>of</strong> AZD2171 for the treatment <strong>of</strong> patients with myelodysplastic<br />

syndromes. Presenting Author: Shannon Eileen O’Mahar, University <strong>of</strong><br />

Wisconsin, Madison, WI<br />

Background: Inhibition <strong>of</strong> vascular endothelial growth factor receptors<br />

(VEGFR) can block growth and trigger apoptosis in neoplastic cells.<br />

AZD2171 (cediranib) is a highly potent, orally bioavailable, VEGFR-1/2<br />

inhibitor. We conducted a phase II study <strong>of</strong> the efficacy <strong>of</strong> AZD2171 for the<br />

treatment <strong>of</strong> MDS. Methods: Adults with MDS (IPSS Int-2 or High) were<br />

eligible if they exhibited adequate organ function and ECOG 0-2. The<br />

primary endpoint was proportion <strong>of</strong> responses according to the IWG criteria<br />

assessed at one and every 3 months. Prior investigation <strong>of</strong> cediranib at 45<br />

mg daily in patients with acute leukemia demonstrated toxicity concerns<br />

and therefore, the starting dose <strong>of</strong> this study was lowered to 30 mg daily.<br />

Results: A total <strong>of</strong> 16 pts with MDS (median age 73 years) were enrolled at a<br />

30 mg starting dose, and all were evaluable. Median baseline marrow blasts<br />

were 12.0 % (range 2-18); 3 pts (18.8 %) had low, 6 (37.5 %)<br />

intermediate, and 7 (43.8 %) had high risk cytogenetics. Prior therapy<br />

included azacitidine (n�7), decitabine (n�2), cytarabine (n�2), erythropoietin-stimulating<br />

agents (ESAs) (n�2), lenalidomide (n�1), or none<br />

(n�6). Patients were treated for a median <strong>of</strong> two 28-day cycles (range 1 to<br />

11). There were no confirmed responses. Patients with baseline blasts �<br />

5% showed no significant reduction in the blast count at 4 and 12 weeks.<br />

Median OS was 4.7 mo (95% CI: 2.6 – 11.6). Median TTP was 3.8 mo<br />

(95% CI: 1.7 – 10.8). Grade 4 hematological adverse events at least<br />

possibly related to cediranib were neutropenia (n�2) and thrombocytopenia<br />

(n�4). Grade 3 hematological adverse events at least possibly related to<br />

study treatment included: neutropenia (n�3), thrombocytopenia (n�2),<br />

and anemia (n�2). Grade 3 non-hematological adverse events included<br />

fatigue (n�4), dyspnea (n�3), dehydration (n�2), diarrhea (n�2), nausea<br />

(n�2), asthenia (n�1), and hypertension (n�1). Hypertension and proteinuria<br />

was uncommon with the 30 mg/day dose. Conclusions: With no<br />

confirmed response from 16 patients, cediranib was determined to be<br />

ineffective at a dose <strong>of</strong> 30 mg daily in our patient population. Supported by<br />

NCI N01-CM62205, NCI P30-CA014520 and the UW Carbone Comprehensive<br />

Cancer Center.<br />

Leukemia, Myelodysplasia, and Transplantation<br />

433s<br />

6569 General Poster Session (Board #18D), Mon, 1:15 PM-5:15 PM<br />

Induction chemotherapy with high-dose cytarabine and mitoxantrone in<br />

elderly acute myeloid leukemia (AML) patients. Presenting Author: Muthalagu<br />

Ramanathan, UMass Memorial Medical Center and UMass Medical<br />

School, Worcester, MA<br />

Background: Induction chemotherapy is <strong>of</strong>ten not used in elderly patients<br />

with AML due to poor prognosis and inferior outcomes. We present here our<br />

experience in treating 20 patients age�70yrs with an induction regimen<br />

consisting <strong>of</strong> high dose cytarabine and mitoxantrone (HiDAC/MITO).<br />

Methods: We analyzed all patients age � 70 years who underwent induction<br />

with HiDAC/MITO at our institution from January 2009 to December 2011.<br />

20 patients received HiDAC at 3 g/m2 daily on days 1-5 and mitoxantrone<br />

80mg/m2* once on day 2(*one patient received 60mg/m2). The end points<br />

analyzed were complete remission (CR) and induction mortality at day 30,<br />

overall survival and progression free survival. Results: Median age was 75<br />

years (70-83). 11 (55%) patients were male and 9 (45%) patients were<br />

female. Median duration <strong>of</strong> follow up <strong>of</strong> 10 (50%) patients who are still<br />

alive at the time <strong>of</strong> this analysis is 439 days (38-1095). High risk<br />

cytogenetics was noted in 11 (55%) patients including two patients with<br />

FLT3 mutated cytogenetics. Intermediate risk cytogenetics was noted in 9<br />

(45%) patients including 2 patients with NPM1 mutated FLT 3 WT AML.<br />

12 (60%) patients had secondary AML. The age unadjusted charlson<br />

comorbidity index (CCI) was 3 (2-9). 15 (75%) patients attained CR and 3<br />

(15%) patients attained a CR with incomplete platelet recovery (CRp). CR<br />

�CRp rate was 90%. Two (10%) patients were refractory to induction and<br />

5 (25%) patients relapsed at a later time point after induction. Median<br />

duration <strong>of</strong> hospital stay was 28 days (22-60). Median time to ANC �1K<br />

was 23.5 days (18-29), excluding 2 patients who had refractory disease.<br />

Median time to platelet recovery �100K was 25 days (20-44), excluding 2<br />

patients with refractory disease and 3 patients with CRp. Regimen related<br />

toxicity included cardiac toxicity in 4 patients and bacteremia in 11<br />

patients. Five (25%) patients were consolidated with stem cell transplant.<br />

Median overall survival (OS) was 151 days (38-1095) and progression free<br />

survival (PFS) was 131 days (38-1049). Conclusions: HiDAC/MITO induction<br />

is well tolerated by elderly patients with AML and demonstrates an<br />

overall response (CR�CRp) rate <strong>of</strong> 90% coupled with no induction deaths.<br />

6571 General Poster Session (Board #18F), Mon, 1:15 PM-5:15 PM<br />

Overall and cancer-specific survival <strong>of</strong> breast, colon, and lung cancer in<br />

patients with and without chronic lymphocytic leukemia: A SEER populationbased<br />

study <strong>of</strong> over 1 million patients. Presenting Author: Benjamin M.<br />

Solomon, Mayo Clinic, Rochester, MN<br />

Background: Chronic lymphocytic leukemia (CLL) is associated with an<br />

increased risk <strong>of</strong> developing second cancers. However, it is unknown<br />

whether CLL alters the natural history <strong>of</strong> these cancers once they occur.<br />

Methods: All patients with breast (n�583,838), colon (n�412,932),<br />

prostate (n�632,922), lung (n�489,290), kidney (n�95,902), pancreas<br />

(n�82,121) and ovarian (n�61,958) cancer reported to the Surveillance,<br />

Epidemiology, and End Results (SEER) Program from 1990 to 2007 were<br />

identified. Overall survival (OS; death due to any cause) and cancer-specific<br />

survival (death due to cancer site <strong>of</strong> interest) were examined, comparing<br />

patients with or without pre-existing CLL. Age- and sex-adjusted hazard<br />

ratios (HRs) were calculated. Results: OS for patients with pre-existing CLL<br />

was inferior for patients with breast (HR�1.61; p�0.001), colon<br />

(HR�1.65; p�0.001), kidney (HR�1.41; p�0.001), prostate (HR�1.75;<br />

p�0.001), and lung (HR�1.22; p�0.001) cancer after adjusting for age<br />

and sex. After excluding CLL-related deaths, OS remained shorter among<br />

patients with breast (p�0.001), colon (p�0.001), kidney (p�0.03),<br />

prostate (p�0.001), and lung (p�0.001) cancer. Cancer-specific survival<br />

was inferior for patients with breast (HR�1.29; p�0.03), colon (HR�1.75;<br />

p�0.001), and lung cancer (HR�1.17; p�0.001) who had pre-existing<br />

CLL after adjusting for age and sex. Conclusions: Several common cancers,<br />

including breast, colon, and lung, have inferior overall and cancer-specific<br />

survival when there is coexistent CLL.<br />

HRs for OS by cancer type.<br />

OS, including CLL deaths OS, excluding CLL deaths<br />

Cancer HR 95% C.I. p value HR 95% C.I. p value<br />

Breast 1.61 1.44-1.80 �0.001 1.38 1.21-1.57 �0.001<br />

Colon 1.65 1.52-1.78 �0.001 1.59 1.46-1.74 �0.001<br />

Kidney 1.41 1.19-1.68 �0.001 1.25 1.02-1.52 0.03<br />

Lung 1.22 1.15-1.29 �0.001 1.20 1.12-1.28 �0.001<br />

Ovary 1.13 0.86-1.49 0.38 0.98 0.73-1.33 0.91<br />

Pancreas 1.00 0.84-1.19 1.00 0.98 0.82-1.17 0.83<br />

Prostate 1.75 1.62-1.90 �0.001 1.35 1.22-1.50 �0.001<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


434s Leukemia, Myelodysplasia, and Transplantation<br />

6572 General Poster Session (Board #18G), Mon, 1:15 PM-5:15 PM<br />

Treatment outcomes in patients older than age 60 with acute myeloid<br />

leukemia (AML) in the nonclinical trial setting. Presenting Author: Steven<br />

Duffy, SUNY Upstate Medical University, Syracuse, NY<br />

Background: Optimal treatment <strong>of</strong> older adults with AML remains challenging.<br />

While AML tends to be a disease <strong>of</strong> older adults, this population<br />

experiences greater treatment-related toxicity and worse overall survival<br />

than younger patients. Only fit older adults enter clinical trials and thus the<br />

results may not apply to the entire population. Methods: We conducted a<br />

retrospective analysis <strong>of</strong> patients � 60 years with AML (APL excluded)<br />

diagnosed prior to 2008 with IRB approval. The association <strong>of</strong> clinical<br />

factors (age, sex, comorbidities, prior chemotherapy), leukemia (prior<br />

myelodysplastic syndrome or myeloproliferative neoplasm, cytogenetics,<br />

WBC count), and therapy (induction chemotherapy, palliative chemotherapy,<br />

and best supportive care) as they relate to overall survival was<br />

evaluated using bivariate and multivariate regression analyses. Results: Of<br />

87 patients (median age 73), 45% were male, 58% had high risk<br />

cytogenetics, 38% had prior MDS/MPD, 92% were chemotherapy naive,<br />

and 21% were in the high/very high Charlson risk class. The majority (67%)<br />

received standard dose induction chemotherapy (IC), 9% received (palliative<br />

intent) low-dose chemotherapy (LDC), and 24% received best supportive<br />

care (BSC). The median overall survival (OS) <strong>of</strong> the entire cohort was 2.5<br />

months. On bivariate analysis high WBC (�50,000 at presentation) was<br />

negatively associated (1.0 vs 2.7 months p �0.01) with survival. OS for IC,<br />

LDC, and BSC were 3.1, 2.8, and 0.7 months, respectively (p�0.001). On<br />

multivariable analysis, IC conferred longer survival when compared to LDC<br />

and BSC combined (OR 0.33 CI 0.2-0.6, p�0.001). High WBC was<br />

associated with a decreased survival time (OR 2.96 CI 1.6-5.5, p�0.02).<br />

Mortality during induction or consolidation chemotherapy was 38%. At<br />

5-year follow-up, only 4 patients were alive. Conclusions: In a non-clinical<br />

trial setting, OS <strong>of</strong> older adults with AML remains dismal. While IC <strong>of</strong>fers a<br />

chance <strong>of</strong> longer survival, mortality with IC is unacceptably high. Further<br />

studies are required to identify and validate tools for risk stratification in<br />

older adults with AML as well as to utilize therapies with an improved<br />

toxicity pr<strong>of</strong>ile.<br />

6574 General Poster Session (Board #19A), Mon, 1:15 PM-5:15 PM<br />

Evaluation <strong>of</strong> combination therapies with MOR00208, an Fc-enhanced<br />

humanized CD19 antibody, in models <strong>of</strong> lymphoma. Presenting Author:<br />

Mark Winderlich, MorphoSys AG, Martinsried/Planegg, Germany<br />

Background: MOR00208 (formerly XmAb5574) is a Fc-engineered humanized<br />

anti-CD19 antibody with superior cytotoxicity in a number <strong>of</strong> in vitro<br />

and in vivo models <strong>of</strong> lymphoma and leukemia, currently being evaluated in<br />

patients with relapsed/refractory CLL. Bendamustine (BEN), an alkylating<br />

agent, fludarabine (FLU), a purine analogue, and the CD20 antibodies<br />

rituximab (RTX) and <strong>of</strong>atumumab (OFA) were evaluated for their ability to<br />

enhance the cytotoxicity <strong>of</strong> MOR00208. Methods: Utilizing CD19/CD20�<br />

MEC-1 CLL cells, cytotoxicity was assessed in vitro by flow cytometry.<br />

MOR00208 was tested alone and in combination with BEN, FLU, RTX and<br />

OFA using peripheral blood mononuclear cells as effector cells. Median<br />

survival (MS) was evaluated in a murine model <strong>of</strong> disseminated lymphoma,<br />

using i.v.-administered RAMOS cells (CD19�) and treatment with<br />

MOR00208 alone and in combination with FLU. Combinatorial effects<br />

were classified according to Chou-Talalay (Pharmacol Rev. 2006 Sep;58(3):<br />

621-81). Results: MOR00208-mediated in vitro cytotoxicity was synergistically<br />

enhanced by BEN, FLU and the CD20 antibodies irrespective <strong>of</strong> their<br />

different modes <strong>of</strong> action (Combination Index � 1). In vivo, FLU alone at<br />

doses up to the maximum tolerated dose (MTD) did not prolong MS,<br />

whereas MOR00208 at 3 mg/kg increased MS by 26% compared to the<br />

isotype control. Co-administration <strong>of</strong> MOR00208 with 125 mg/kg FLU<br />

significantly enhanced MS by 65%. Despite its lack <strong>of</strong> single agent activity,<br />

FLU thus significantly enhanced the cytotoxicity <strong>of</strong> MOR00208. Conclusions:<br />

The cytotoxic in vitro activity <strong>of</strong> MOR00208 on CD19� B cells was<br />

synergistically enhanced by all studied drugs irrespective <strong>of</strong> their effector<br />

mechanisms. In vivo, the activity <strong>of</strong> MOR00208 in an aggressive lymphoma<br />

model was potentiated in combination with FLU. These results provide a<br />

mechanistic rationale for MOR00208 drug combinations which warrant<br />

further evaluation in clinical trials.<br />

6573 General Poster Session (Board #18H), Mon, 1:15 PM-5:15 PM<br />

A simple but effective model to decrease early deaths in acute promyelocytic<br />

leukemia (APL). Presenting Author: Anand P. Jillella, Department <strong>of</strong><br />

Medicine, Division <strong>of</strong> Hematology/Oncology, Georgia Health Sciences<br />

University, Augusta, GA<br />

Background: Recent reports suggest that approximately 30% <strong>of</strong> patients<br />

with APL die during induction. This has been confirmed in large populationbased<br />

studies in Sweden and the US. A recent analysis <strong>of</strong> SEER data from<br />

13 population-based cancer registries with 1400 APL patients in the US<br />

showed that 17% <strong>of</strong> all patients and 24% <strong>of</strong> patients greater than 55 years<br />

<strong>of</strong> age die within one month <strong>of</strong> diagnosis. The most common causes <strong>of</strong><br />

death are bleeding, infection, differentiation syndrome and multi-organ<br />

failure. Patients who survive induction have an excellent cure rate with few<br />

late relapses. Hence, decreasing early deaths is a high priority both at<br />

experienced as well as smaller centers with limited leukemia treatment<br />

experience in this highly curable disease. Methods: At Georgia Health<br />

Sciences University, between 7/2005 and 6/2009, 19 patients were<br />

diagnosed with APL. Seven patients (5 high-risk and 2 low-risk) died during<br />

induction resulting in an unusually high mortality rate <strong>of</strong> 37%. All patients<br />

who survived induction are still in remission at present. The high early<br />

death rate prompted us to develop a simple, 2 page treatment algorithm<br />

that focuses on quick diagnosis, prompt initiation <strong>of</strong> therapy, and proactive<br />

and aggressive management <strong>of</strong> all the major causes <strong>of</strong> death during<br />

induction. We also made our treatment protocol available to smaller<br />

treatment centers and helped the treating oncologists manage the patient<br />

during the first few days after diagnosis. Results: From 11/2010 to<br />

12/2011, we treated 4 patients at GHSU and helped manage 4 patients at<br />

2 outreach sites. The age range was 30 to 60; two patients were high-risk, 5<br />

intermediate- and one low-risk. There were no deaths during induction and<br />

all eight patients proceeded to consolidation treatment. Conclusions: While<br />

we recognize that this is a small cohort, our own experience and a similar<br />

approach pioneered by investigators in Brazil clearly shows this to be an<br />

effective intervention to decrease early deaths in APL. We believe our<br />

experience warrants large scale implementation <strong>of</strong> our protocol in an<br />

attempt to reduce early APL mortality.<br />

6575 General Poster Session (Board #19B), Mon, 1:15 PM-5:15 PM<br />

Cancer testicular antigens as a prognostic markers <strong>of</strong> chronic lymphocytic<br />

leukemia. Presenting Author: Julio C. Chavez, H. Lee M<strong>of</strong>fitt Cancer Center<br />

& Research Institute, Tampa, FL<br />

Background: CLL is the most common leukemia in adults and has a variable<br />

course and prognosis. <strong>Clinical</strong> staging systems and the use <strong>of</strong> biomarkers<br />

such as cytogenetics and the IGVH mutational status remain the gold<br />

standard for stratification. Cancer testis antigens (CTAs) expression analyses<br />

have shown prognostic impact in other hematological malignancies<br />

such as acute leukemias and multiple myeloma. With the analysis <strong>of</strong> CTAs<br />

we aim to discover additional molecular markers that can help us predict<br />

the clinical course and outcome in CLL patients. Methods: We analyzed the<br />

expression by RT-PCR <strong>of</strong> 39 known CTAs including members <strong>of</strong> the MAGE,<br />

GAGE, MAD-CT and SSX CTAs families in a group <strong>of</strong> 59 CLL patients seen<br />

at our institution. The M<strong>of</strong>fitt Cancer Center Total Cancer Care (TCC)<br />

database was used to correlated these findings with relevant clinical and<br />

molecular markers such as Rai stage, demographic data, initial WBC count,<br />

IGVH mutational status, CD38 expression, cytogenetics by FISH analysis<br />

and treatment outcomes. Overall survival (OS) and progression free survival<br />

(PFS) were calculated using Kaplan Meier curves with a SPSS statistical<br />

s<strong>of</strong>tware Results: Deletion 13q was the most common gene abnormality in<br />

35/59 (59.3%). 29/59 (49.1%) required at least one line <strong>of</strong> treatment.<br />

MAGE family CTAs were present in 53/59 (83.9%), MAD-CT in 24/59<br />

(40.7%), SSX in 14/59 (23.7%) and GAGE in 12/59(20.3%). 35/59<br />

(59.3%) CLL patients expressed 2 or more MAGE family CTAs. No<br />

differences were noted in the Rai staging, initial white blood cell count, B<br />

symptoms, extranodal disease, presence <strong>of</strong> CD38, or unmutated IVGH. The<br />

OS was not significantly different among all CTAs families with a tendency<br />

towards a better OS those expressing the SSX family (p�0.63). Additionally,<br />

CLL patients with single expression <strong>of</strong> MAD-CT1 (p�0.05) and<br />

MAGE-B2 (p�0.038) antigens correlated with improved survival.<br />

Conclusions: Patients with SSX family have a trend towards improved<br />

survival. When analyzed by individual antigen, MAD-CT1 and MAGE-B2<br />

expression was associated with improved survival. Further studies with a<br />

larger number <strong>of</strong> patients are needed to assess the real value <strong>of</strong> the<br />

expression <strong>of</strong> CTAs in CLL.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


6576 General Poster Session (Board #19C), Mon, 1:15 PM-5:15 PM<br />

AML patients with IDH1 or IDH2 mutations treated with hypomethylating<br />

agents: A case series. Presenting Author: Christopher Brent Benton,<br />

University <strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: Isocitrate dehydrogenase 1 (IDH1) and IDH2 mutations play a<br />

significant role in acute myeloid leukemia (AML), yet optimal treatment for<br />

AML patients with IDH mutations remains unclear. IDH mutations have<br />

been associated with a globally hypermethylated genomic state, as the<br />

result <strong>of</strong> increased levels <strong>of</strong> 2-HG, a regulator <strong>of</strong> histone methylation. We<br />

sought to retrospectively determine responses <strong>of</strong> patients with IDHmutated<br />

AML to treatment with hypomethylating agents. Methods: We<br />

reviewed clinical data for AML patients treated at The University <strong>of</strong> Texas<br />

M. D. Anderson Cancer Center from June 2001 to December 2011 whose<br />

stored leukemia samples from bone marrow aspirateshad been tested<br />

retrospectively for IDH1 and IDH2 mutations. We searched three databases<br />

that contained records for 407 AML patients. We selected patients who had<br />

been treated with the hypomethylating agents 5-azacitidine or decitabine,<br />

alone or in conjunction with other therapeutics. We retrospectively reviewed<br />

the patients’ medical records to obtain demographic, laboratory,<br />

treatment, and clinical data. We evaluated response measured by remission<br />

status, reduction in bone marrow blasts and peripheral blood blasts,<br />

and time to relapse. Results: We identified 104 patients (25.6%) who had<br />

either an IDH1 or IDH2 mutation, and <strong>of</strong> these, 8 patients had also been<br />

treated with hypomethylating agents. A ninth AML patient, who is IDH2mutant,<br />

is currently undergoing treatment with decitabine alone. Of these 9<br />

patients, three had a durable complete remission without recovery <strong>of</strong> blood<br />

counts (CRp), three had a partial response (PR), with or without blood<br />

count recovery, and three had no response. All 9 patients did have an initial<br />

decrease in the percentage <strong>of</strong> bone marrow blasts, peripheral blood blasts,<br />

or both. Conclusions: This series <strong>of</strong> patients demonstrates the possibility<br />

that AML patients with IDH1 or IDH2 mutations benefit from the use <strong>of</strong><br />

hypomethylating agents, and suggests it is worthwhile to prospectively<br />

investigate treatment <strong>of</strong> patients with IDH-mutated AML using hypomethylating<br />

agents.<br />

6578 General Poster Session (Board #19E), Mon, 1:15 PM-5:15 PM<br />

Analysis <strong>of</strong> outcomes <strong>of</strong> patients (pts) with blastic plasmacytoid dendritic<br />

cell neoplasm (BPDCN). Presenting Author: Naveen Pemmaraju, University<br />

<strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: BPDCN, formerly known as CD4 � CD56 � hematodermic<br />

tumor or blastic NK cell lymphoma, is a rare and aggressive hematologic<br />

malignancy. Little is known about outcomes <strong>of</strong> pts with BPDCN. Methods:<br />

We conducted a retrospective review <strong>of</strong> pts meeting following criteria:<br />

pathological diagnosis <strong>of</strong> BPDCN confirmed by an experienced hematopathologist<br />

and age � 18. Results: 13 pts, diagnosed October 1998-July<br />

2011, were identified. Median (med) age: 62 years (range 20-86 years), 12<br />

(92%) were male. Bone marrow (BM) involved in 9 (69%), skin 8 (62%),<br />

lymph nodes 5 (38%), brain 1 and uterine/ovarian 1 pt. Immunophenotype<br />

by flow cytometry: CD4� (11/11 pts), CD56� (10/11 pts), TCL-1� (4/4<br />

pts). Karyotype (8 pts): del(12) (1pt), complex (2), and diploid (5). Med<br />

CBC: WBC 5.8 x 109 /L (1.7-76.5), Hb 12.3 g/dL (8.3-17.0), platelet 107 x<br />

109 /L (44-255). Med BM blasts: 22% (0-77). 10 pts received first-line<br />

therapy with Hyper-CVAD alternating with high-dose methotrexate(MTX)<br />

and cytarabine (HCVAD) (in 1 pt following one cycle <strong>of</strong> CHOP), CHOP (2),<br />

oral MTX (1). Med follow-up time: 9 months (mo) (2-14 mo). Med number<br />

chemotherapy regimens: 1 (1-5). Complete remission (CR) in 10 pts. Med<br />

CR1: 19 mo (4-39 mo). Med overall survival (OS) 29 mo (1-44 mo). 9 pts<br />

have died, unknown cause (3), multi-organ failure (6). 10 pts received<br />

HCVAD as part <strong>of</strong> first line therapy: med OS: 29 mo (1-44 mo), med CR1:<br />

21 mo (4-39 mo), 9/10 (90%) pts achieved CR1; 1 pt did not achieve CR1<br />

(died, pneumonia, day 15). 9 pts (69%) had BM involvement (7 with other<br />

organs involved besides BM). 4 (31%) pts had no BM involvement; all 4 <strong>of</strong><br />

these pts had skin involvement only. Med OS, med CR1 <strong>of</strong> pts with BM<br />

involvement: 23 mo (1-44mo), 22 mo (4-39mo), respectively. Med OS,<br />

med CR1 <strong>of</strong> skin only pts: 29 mo (2-31 mo), 7 mo (6-19 mo), respectively,<br />

p�not significant. 4 pts received stem cell transplant (SCT) (2 allogeneic<br />

,2 autologous). Med OS for pts receiving SCT (n�4) was 31 mo(8-31 mo)<br />

versus med OS for non-SCT group (n�9) <strong>of</strong> 29 mo (1-44), p�not<br />

significant. Conclusions: Despite intensive multi-agent chemotherapy and<br />

SCT, response rates are low and survival is short. Better understanding <strong>of</strong><br />

the biologic basis <strong>of</strong> this disease and novel treatment approaches for<br />

treatment <strong>of</strong> BPCDN are crucial.<br />

Leukemia, Myelodysplasia, and Transplantation<br />

435s<br />

6577 General Poster Session (Board #19D), Mon, 1:15 PM-5:15 PM<br />

Phase I dose escalation study <strong>of</strong> TG02 in patients with advanced hematologic<br />

malignancies. Presenting Author: Gail J. Roboz, Weill Medical College<br />

<strong>of</strong> Cornell University, New York, NY<br />

Background: TG02 is a novel multikinase inhibitor with a unique spectrum<br />

<strong>of</strong> activity, targeting both the cell cycle regulatory cyclin-dependent kinases<br />

(CDKs) 1 and 2 and the transcriptional regulators CDKs 7 and 9. TG02 also<br />

inhibits the emerging oncogenic MAPK ERK5 and the DNA damage<br />

response mediator CDK5. TG02 kills primary blasts from a variety <strong>of</strong><br />

hematologic cancers and is curative in the MV4-11 model <strong>of</strong> FLT3-mutant<br />

AML. Methods: This is a first-in-man,single arm, open label, phase I dose<br />

escalation trial. The primary endpoints are dose-limiting toxicity (DLT),<br />

maximally tolerated dose (MTD ) and recommended phase 2 dose (RP2D).<br />

Patients (pts) � 18 years with advanced hematological malignancies or<br />

newly diagnosed AML pts � 65 years unfit for intensive therapy were<br />

enrolled onto daily (A) and intermittent (B, 5 days on 2 days <strong>of</strong>f X 2 weeks)<br />

schedules. Pts had acceptable organ function and ECOG PS 1-2. Definition<br />

<strong>of</strong> DLT was G3-4 AST or ALT �7 days, G4 AST or ALT, G4 hyperbilirubinemia,<br />

any other NCI CTC G3-4 events not due to underlying disease.<br />

Dose levels on arm A were 10mg to 70mg and 15mg-150 mg on arm B.<br />

Results: Forty-five pts have received at least one dose <strong>of</strong> study drug. Median<br />

age was 66 years (range, 37-87) and 80% were ECOG 0-1. Disease types<br />

enrolled included: AML (80%), high-risk MDS (22%), and CML-BC (3%).<br />

The median number <strong>of</strong> previous regimens was 3 (range, 1-12). The MTD on<br />

arm A was defined at 50 mg daily based on 2 DLTs at the 70mg dose level<br />

(G4 hyperbilirubinemia, G4 fatigue). Enrollment to arm B has competed<br />

dose levels 15 (N�3), 30 (N�3), 50 (N�3), 70 (N�3), 100mg (N�3),<br />

and enrollment at 150mg is ongoing without DLT to date. Common drug<br />

related adverse events were nausea (42%), vomiting (23%), fatigue (18%),<br />

decreased appetite (15%), constipation and diarrhea (13% each). Preliminary<br />

PK demonstrated dose proportional increases in exposure and a T1/2 ,<br />

supporting once daily dosing. Conclusions: The MTD for TG02 has been<br />

determined for the daily schedule at 50mg. Enrollment continues on the<br />

intermittent schedule. Schedules <strong>of</strong> every other day and week on/week <strong>of</strong>f<br />

dosing will also be evaluated.<br />

6579 General Poster Session (Board #19F), Mon, 1:15 PM-5:15 PM<br />

Outcome <strong>of</strong> acute myeloid leukemia in patients with a history <strong>of</strong> autoimmune<br />

disease. Presenting Author: Courtney Denton Dinardo, Hospital <strong>of</strong><br />

the University <strong>of</strong> Pennsylvania, Philadelphia, PA<br />

Background: Increased risks <strong>of</strong> solid and lymphoid malignancies are<br />

described in patients with autoimmune (AI) disease. The association<br />

between AI disease, AI treatment (particularly anti-TNF therapy), and AML<br />

is less established. Moreover, the pathologic characteristics <strong>of</strong> AML in this<br />

population have not been evaluated. Here we describe our AML experience<br />

in patients with prior AI disease. Methods: Patients with AML from 1992 to<br />

2011 were identified from our database, and AI disorder was verified<br />

through chart review. Patients were included if they had an AI disorder that<br />

required systemic treatment, and diagnosed � 1 year prior to AML.<br />

Patients were excluded if details including the year <strong>of</strong> AI diagnosis or<br />

treatment(s) received were not documented. A total <strong>of</strong> 22 patients were<br />

included for analysis. Results: Median age at AML diagnosis was 59 years<br />

(range 32 – 78), 4 (18%) were men. 10 (48%) had received an anti-TNF<br />

agent, for a median duration <strong>of</strong> 30 months (range 2 – 120). Median latency<br />

from AI diagnosis to AML was 9.4 yrs. All FAB subtypes were represented<br />

except M0 and M6, 9 (41%) had M4 or M5 disease. 10/21 patients (45%)<br />

had normal cytogenetics, and 5 had favorable cytogenetics (2 with acute<br />

promyelocytic leukemia (APML)). Standard induction was given to 19<br />

patients, and APML-directed therapy for those with APML. One patient<br />

received an autologous transplant in CR1, and 5 an allogeneic transplant (1<br />

in CR1, 4 in CR2). 9 patients are alive in CR1. 13 relapsed, and 4 (31%)<br />

are alive in CR2 or CR3 (3 following allogeneic transplant in later CR).<br />

Median OS was 68.1 months. In univariate analysis, factors associated<br />

with OS were cytogenetics, FAB subtype, and gender. Anti-TNF therapy<br />

may be associated with poorer prognosis; this was not statistically significant.<br />

Median OS was 14.1 months for poor, 68.1 months for intermediate,<br />

and not reached for favorable risk cytogenetics. Conclusions: Median OS<br />

was higher (� 5 years) than expected. Younger age and an increased<br />

incidence <strong>of</strong> favorable risk AML may account for this finding: whether this<br />

is a meaningful biologic association or stochastic event can only be verified<br />

with larger studies. Our observations do not support a label <strong>of</strong> “secondary<br />

leukemia” or therapy-related neoplasm in this population.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


436s Leukemia, Myelodysplasia, and Transplantation<br />

6580 General Poster Session (Board #19G), Mon, 1:15 PM-5:15 PM<br />

L-arginine depletion by PEG-arginase I, a new potential therapy for acute<br />

lymphoblastic leukemia. Presenting Author: Ofelia Crombet Ramos, Louisiana<br />

State University Health Sciences Center, New Orleans, LA<br />

Background: Advances in therapies have resulted in an overall complete<br />

remission rate <strong>of</strong> approximately 85% for childhood acute lymphoblastic<br />

leukemia (ALL). In contrast, the overall remission rate <strong>of</strong> adults with<br />

leukemia continues to be poor, only about 40% in cases <strong>of</strong> T cell-ALL<br />

(T-ALL). Therefore, it is imperative to generate new therapies that alone or<br />

in combination with other treatments could potentially increase the<br />

percentages <strong>of</strong> complete responders or be used to treat the refractory ALL<br />

population. Our published results show that a pegylated form <strong>of</strong> human<br />

arginase I (peg-Arg I) prevented T-ALL cell proliferation in vitro and in vivo<br />

through the induction <strong>of</strong> tumor cell apoptosis. Interestingly, the antileukemic<br />

effects induced by peg-Arg I did not affect the anti-tumor activity<br />

<strong>of</strong> normal T cells, suggesting an anti-tumor specific effect. Our hypothesis<br />

states that peg-Arg I has an anti-tumoral effect on B-ALL and T-ALL cells in<br />

vitro and that the sensitivity <strong>of</strong> ALL cells to peg-Arg I depends on their<br />

expression <strong>of</strong> argininosuccinate synthase (ASS) and their ability to produce<br />

L-arginine de novo from citrulline. Methods: Malignant T cell proliferation<br />

was tested using nonradioactive cell proliferation yellow tretrazolium salt<br />

kit. Apoptosis studies were based on the expression <strong>of</strong> annexin V. Western<br />

blot assays were conducted to determine enzymatic expression in different<br />

cell lines. Results: The results <strong>of</strong> our in vitro experiments showed that<br />

peg-Arg I had a pro-apoptotic and anti-proliferattive effect on B-ALL cells<br />

similar to the one previously seen on T-ALL cells. These effects can be<br />

overcome in cell lines able that express ASS and therefore to produce<br />

L-arginine de novo. Conclusions: Our results suggest the role <strong>of</strong> ASS in the<br />

ALL-apoptosis induced by peg-Arg-I. Our next steps include: _Understand<br />

why ASS-expressing ALL cells do not undergo apoptosis when cultured with<br />

peg-Arg-I_Determine the role <strong>of</strong> ASS in the anti-leukemic effect induced by<br />

peg-Arg-I in vivo. Completion <strong>of</strong> this research is expected to lead to a better<br />

understanding <strong>of</strong> how peg-Arg-I kills ALL cells and could provide the<br />

foundation for a novel therapy for ALL patients.<br />

6582 General Poster Session (Board #20A), Mon, 1:15 PM-5:15 PM<br />

Phase II study <strong>of</strong> the oral MEK inhibitor selumetinib (AZD6244) in<br />

advanced acute myeloid leukemia (AML). Presenting Author: Nitin Jain,<br />

The University <strong>of</strong> Chicago Medical Center, Chicago, IL<br />

Background: RAS-RAF-MEK-ERK pathway is activated in �75% <strong>of</strong> AML<br />

patients (pts). Selumetinib (AZD6244) is an orally bioavailable small<br />

molecule inhibitor <strong>of</strong> the MEK kinase. We report here the results <strong>of</strong> a phase<br />

II multicenter trial using selumetinib in advanced AML pts. Methods: Pts<br />

�18 years (yrs) with relapsed/refractory AML or �60 yrs old with untreated<br />

AML who were not candidates for standard chemotherapy were eligible. Pts<br />

received selumetinib 100mg twice daily. Pts were screened for KRAS,<br />

NRAS and FLT3 mutations. Analysis for p-ERK (downstream <strong>of</strong> MEK) was<br />

performed by flow-cytometry. Results: 47 pts (27 men) were enrolled.<br />

Median age was 69 yrs (range, 26-83). 85% were �60 yrs. Disease stage<br />

included previously untreated AML (13 pts, all were � 60 yrs; 11 had prior<br />

therapy for antecedent hematologic disease), primary refractory AML (14<br />

pts), 1st relapse (8 pts) and beyond first relapse (12 pts). Overall, 51% pts<br />

had secondary AML. Median number <strong>of</strong> prior therapies was 2 (range, 0-6).<br />

51% had poor-risk cytogenetics. 10 pts had a FLT3 ITD, 3 pts had an<br />

NRAS mutation and one pt had a KRAS mutation. Selumetinib was well<br />

tolerated. Median number <strong>of</strong> cycles administered was 1 (range, 1-21).<br />

49% received �2 cycles. Grade �3 adverse events possibly related to the<br />

drug included fatigue, dyspnea, and nausea occurring in 9%, 9% and 6%<br />

respectively. 1 pt has partial response, 3 pts had minor response (�50%<br />

decrease in blood and/or marrow blasts lasting �4 weeks), 2 pts had<br />

unconfirmed minor response (uMR: �50% decline in marrow blasts<br />

without a follow up confirmatory biopsy), 4 pts had stable disease (median:<br />

16.5 weeks, range: 9-85). No pt with FLT3 ITD or NRAS mutation<br />

responded. The sole pt with KRAS mutation had uMR with hematologic<br />

improvement in platelets. Analysis <strong>of</strong> p-ERK showed baseline activation in<br />

15/21 (71%) pts, but no difference in baseline levels in the responders vs<br />

non-responders (p�0.27). Conclusions: Administration <strong>of</strong> selumetinib in<br />

advanced AML is associated with modest antileukemic activity, which is<br />

independent <strong>of</strong> baseline p-ERK levels. Given its favorable toxicity pr<strong>of</strong>ile,<br />

combination with drugs that target relevant signaling pathways in AML<br />

should be considered. (Sponsored by NCI grant NO1-CM-62201)<br />

6581 General Poster Session (Board #19H), Mon, 1:15 PM-5:15 PM<br />

Quantitative detection <strong>of</strong> ZAP-70 expression in chronic lymphocytic leukemia.<br />

Presenting Author: Peixuan Zhu, Creatv MicroTech, Inc., Potomac,<br />

MD<br />

Background: Intracellular ZAP-70 protein was recently recognized as<br />

prognostic marker in chronic lymphocytic leukemia (CLL). The specific<br />

objective <strong>of</strong> this study was to develop a simple and sensitive assay, for the<br />

quantitative detection <strong>of</strong> ZAP-70 protein in leukemic cells. Methods:<br />

Components <strong>of</strong> the assay system include sample preparation, immunomagnetic<br />

fluorescence assay, Signalyte-II spectr<strong>of</strong>luorometer. The leukemic<br />

cells were isolated from CLL patient blood samples and lysed to release the<br />

intracellular ZAP-70 protein. ZAP-70 protein was captured by magnetic<br />

beads coated with anti-ZAP70 capture antibody, and recognized by a<br />

fluorescent detector antibody, forming an immuno-sandwich complex. This<br />

complex was dissociated for measurement <strong>of</strong> the fluorescence signal,<br />

which was proportional to ZAP-70 concentration, using the spectr<strong>of</strong>luorometer.<br />

Results: The assay conditions were extensively optimized by selecting<br />

an optimal pair <strong>of</strong> capture/detector antibodies, conjugation <strong>of</strong> fluorescence<br />

dye, cell lysis condition and excitation/emission wavelengths. The protocol<br />

was further validated with two positive controls, Jurkat cell lysate and<br />

recombinant ZAP70 protein (rZAP70). The limit <strong>of</strong> detection was determined<br />

to be lower than 125 Jurkat cells and 39 pg <strong>of</strong> rZAP70 protein. The<br />

signal response was linear over a wide range <strong>of</strong> concentration, from 625 to<br />

40,000 Jurkat cells per test (R2�0.9987) and from 0 to 40,000 pg<br />

rZAP70 protein per test (R2�0.9928). The results from 20 CLL patients<br />

correlated strongly with flow cytometry analysis. The concordance between<br />

the two methods for positive and negative results was 100% (7/7) and 92%<br />

(12/13), respectively, while the overall concordance between the two<br />

methods was 95%. Conclusions: The assay is a simple, reliable, and<br />

reproducible method for quantitative detection <strong>of</strong> ZAP-70 in patient<br />

leukemic cells, without the need for cell fixation or permeabilization. The<br />

entire assay could be completed in 5.5 hours. The ZAP-70 signal was linear<br />

over a wide dynamic range, which we believe enables quantitative assessment<br />

<strong>of</strong> small changes in ZAP-70 expression over the course <strong>of</strong> the disease<br />

and in response to therapeutic intervention.<br />

6583 General Poster Session (Board #20B), Mon, 1:15 PM-5:15 PM<br />

Real-world response monitoring and tolerability <strong>of</strong> imatinib-treated chronic<br />

myeloid leukemia patients captured in a retrospective research registry.<br />

Presenting Author: David D. Stenehjem, Huntsman Cancer Institute, Salt<br />

Lake City, UT<br />

Background: Monitoring tolerability and response to imatinib (IM) is an<br />

important aspect <strong>of</strong> chronic myeloid leukemia (CML) management. The<br />

objective <strong>of</strong> this study is to assess real-world tolerability and response<br />

monitoring in IM treated CML patients (pts). Methods: A comprehensive<br />

retrospective outcomes research registry <strong>of</strong> CML pts was created from the<br />

University <strong>of</strong> Utah electronic health record system. Study inclusion was<br />

limited to pts diagnosed with CML in chronic phase in 2001 to 2010 and<br />

treated with IM as a first-line therapy. Utilization and outcome <strong>of</strong> cytogenetic<br />

and molecular testing within 18 months <strong>of</strong> IM initiation, rates <strong>of</strong><br />

adverse drug events (ADEs), and therapy modifications were evaluated by<br />

chart review. Results: A total <strong>of</strong> 92 pts were treated with IM as first-line<br />

therapy. Within the first 18 months <strong>of</strong> treatment, cytogenetic testing was<br />

recorded in 45 pts (49%) and <strong>of</strong> these 33 pts (73%) achieved a complete<br />

cytogenetic response (CCyR) in a median <strong>of</strong> 241 days (range: 110-542);<br />

molecular testing was completed in 48 pts (52%) and <strong>of</strong> these 24 pts<br />

(50%) achieved at least a major molecular response (MMR) in a median <strong>of</strong><br />

254 days (range: 99-546). Imatinib associated ADEs <strong>of</strong> any grade (n � 60)<br />

occurred in 42 (46%) pts resulting in dose reductions in 15 pts (36%) in a<br />

median <strong>of</strong> 77 days and discontinuation <strong>of</strong> IM occurred in 9 (21%) pts in a<br />

median <strong>of</strong> 130 days. The IM dose was increased to �400 mg in 21 (23%)<br />

pts in a median <strong>of</strong> 457 days (range: 21-2112). Of pts diagnosed between<br />

2006 to 2010 (n � 34; 37%), 8 (25%) pts transitioned to dasatinib or<br />

nilotinib in a median <strong>of</strong> 397 days (range: 147 to 1057). Reasons for<br />

therapy change included physician documented suboptimal response or<br />

treatment failure (n � 5) and ADEs to IM (n � 3). Conclusions: Utilization <strong>of</strong><br />

cytogenetic and molecular testing within 18 months <strong>of</strong> IM initiation was<br />

lower than the National Comprehensive Cancer Network or European<br />

LeukemiaNet CML guidelines would suggest. Further research is warranted<br />

to understand limited response monitoring and outcomes in non-monitored<br />

pts. The ADE rate was similar to clinical trial data. The impact <strong>of</strong> ADEs on<br />

subsequent treatment and outcomes in CML pts deserves further study.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


6584 General Poster Session (Board #20C), Mon, 1:15 PM-5:15 PM<br />

Ofatumumab retreatment and maintenance in patients with fludarabinerefractory<br />

CLL. Presenting Author: Anders Österborg, Karolinska University<br />

Hospital, Stolkholm, Sweden<br />

Background: In Study 406, the anti-CD20 monoclonal antibody <strong>of</strong>atumumab<br />

(<strong>of</strong>a), given as monotherapy over 6 months, showed 47% overall response rate<br />

(ORR) in patients (pts) with chronic lymphocytic leukemia (CLL) refractory to<br />

fludarabine and alemtuzumab (FA-ref), or to fludarabine with bulky (�5cm)<br />

lymphadenopathy (BF-ref). The effects <strong>of</strong> <strong>of</strong>a retreatment (retx) and maintenance<br />

(mt) are unknown. Methods: Pts who responded to <strong>of</strong>a and then progressed<br />

or had stable disease (SD) in Study 406, were <strong>of</strong>fered retx in Study 416<br />

(NCT00802737; GSK/Genmab) with <strong>of</strong>a 1 x 300 mg � 7 x 2000 mg weekly<br />

followed by mt with <strong>of</strong>a 2000 mg monthly for up to 2 years (if SD or better).<br />

Primary endpoint was ORR (1996 NCI-WG). Safety and time to event outcomes<br />

were also assessed. Results: Of 29 pts enrolled, 7 had SD and 22 had partial<br />

response (PR) and progressed in Study 406. Pts were defined per Study 406: 17<br />

FA-ref, 11 BF-ref, 1 “other”. Pretreatment characteristics were similar between<br />

groups. Pts received a median <strong>of</strong> 12 doses (range 3-32); 86% had 8 doses, 3%<br />

received all 32 doses. 72% <strong>of</strong> pts had infusion-related adverse events (AEs),<br />

41% at 1st infusion, mostly grade 1-2. The most common grade �3 AE<br />

occurring up to 60 days after last tx was infection (38%); the most common was<br />

pneumonia (17%). 3 pts (10%) had fatal infections, all bronchopneumonia.<br />

<strong>Clinical</strong> efficacy is shown in Table. Comparative analysis to Study 406 is<br />

ongoing. Conclusions: The response to <strong>of</strong>atumumab (<strong>of</strong>a) as induction retreatment<br />

and progression-free survival in this limited number <strong>of</strong> pts was similar to<br />

1st treatment (Study 406). Ofa maintenance had some clinical benefit for pts<br />

with advanced CLL. Ofa was well-tolerated with no unexpected toxicities.<br />

Efficacy <strong>of</strong> <strong>of</strong>a retx.<br />

FA-ref (n�17) BF-ref (n�11) Total (n�29)<br />

Response %<br />

ORR (95% CI )<br />

At 8 wks (end <strong>of</strong> weekly retx): 59 (33, 82) 18 (2, 52) 45 (26, 64)<br />

CR 6 0 3<br />

Nodular PR 6 0 3<br />

PR 47 18 38<br />

SD 29 55 38<br />

PD 6 0 3<br />

Not evaluable (NE) 6 27 14<br />

Through 24 wks (4 months mt) 24 (7, 50) 18 (2, 52) 21 (8,40)<br />

Through 52 wks (11 months mt) 24 (7, 50) 18 (2, 52) 24 (10,44)<br />

CR 12 0 7<br />

PR 12 18 17<br />

SD 47 64 52<br />

PD 18 10<br />

NE 12 18 14<br />

Time to event outcomes Median (95% CI), months<br />

Duration <strong>of</strong> response 11 (7, 11) 24 (NA) 24 (7, 24)<br />

PFS 8 (3, 13) 7 (2,12) 8 (5, 12)<br />

Overall survival 20 (11,28) 11 (5,NA) 18 (11, NA)<br />

6586 General Poster Session (Board #20E), Mon, 1:15 PM-5:15 PM<br />

Albumin as a prognostic factor for overall survival in newly diagnosed<br />

patients with acute myeloid leukemia (AML). Presenting Author: Awais M.<br />

Khan, University <strong>of</strong> Alabama at Birmingham Comprehensive Cancer Center,<br />

Birmingham, AL<br />

Background: Hypoalbuminemia (HA) is an adverse prognostic factor in<br />

multiple neoplastic diseases. Severe hypoalbuminemia (�3.0 g/dl) at day<br />

�90 post allogeneic hematopoietic cell transplant (AHCT) was reported as<br />

an independent predictive variable for non-relapse mortality and overall<br />

survival (Kharfan-Dabaja, et al Biol Blood Marrow Transplant 2009; 15).<br />

We examined the prognostic value <strong>of</strong> serum albumin level prior to induction<br />

chemotherapy in patients with newly diagnosed AML. Methods: Data were<br />

collected retrospectively in newly diagnosed AML patients receiving induction<br />

chemotherapy (3� 7 regimen). Primary objective was to examine the<br />

relationship between serum albumin at baseline and probability <strong>of</strong> achieving<br />

complete remission (CR) or incomplete remission (CRi) and overall<br />

survival (OS). The Kaplan–Meier method used to estimate median overall<br />

survival; chi-square test used for comparison <strong>of</strong> categorical variables and<br />

t-test for continuous variables. Log rank test used to compare Kaplan–<br />

Meier survival estimates between two groups. Results: Between November<br />

2004 to July 2007, 135 patients who received 3�7 induction chemotherapy<br />

were included. Patient baseline characteristics were similar between<br />

patients with serum albumin � 3.5 g/dl (HA) and those with serum<br />

albumin � 3.5 g/dl (no HA) with respect to age, sex, FAB subtype, history <strong>of</strong><br />

antecedent MDS, karyotype, and chemotherapy . In patients with HA, mean<br />

age was 60 years compared to 56.5 years in non HA group. The median OS<br />

for patients with HA was 221 days (95%CI 149.5-292.5) compared to 421<br />

days (95%CI 236.7-605) with normal serum albumin (p�0.005). (Figure-1)<br />

The CR/CRi rate was 64%% for HA and 77.6% for those with normal<br />

albumin (p�0.09). In a multivariable Cox regression analysis including age<br />

� 60 years, history <strong>of</strong> MDS, karyotype, and serum albumin level at<br />

baseline; only age, karyotype and serum albumin were independent<br />

predictors <strong>of</strong> OS [Hazard ratio 0.47 (95%CI 0.31-0.71) (p�0.005) for<br />

normal serum albumin group]. Conclusions: In newly diagnosed AML, we<br />

demonstrate that hypoalbuminemia � 3.5 g/dl is an independent covariate<br />

for overall survival with conventional chemotherapy management. The<br />

prognostic value <strong>of</strong> low serum albumin should be validated in a prospective<br />

study.<br />

Leukemia, Myelodysplasia, and Transplantation<br />

437s<br />

6585 General Poster Session (Board #20D), Mon, 1:15 PM-5:15 PM<br />

Phase I study <strong>of</strong> TH-302, a hypoxia-activated cytotoxic prodrug, in subjects<br />

with advanced leukemias. Presenting Author: Marina Konopleva, University<br />

<strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: TH-302 is a 2-nitroimidazole prodrug <strong>of</strong> the DNA alkylator,<br />

bromo-isophosphoramide mustard designed to be selectively activated in<br />

hypoxic conditions. Preclinical data in mice with ALL have demonstrated<br />

marked expansion <strong>of</strong> hypoxia in areas <strong>of</strong> marrow leukemia infiltrates (Benito<br />

et al., PlosOne, in press). TH-302 also exhibited specific hypoxiadependent<br />

cytotoxicity when tested against primary ALL and AML samples<br />

in vitro. Based on these findings, a phase 1 study <strong>of</strong> TH-302 was designed<br />

for advanced leukemias. Methods: Eligibility: ECOG � 3, relapsed/<br />

refractory leukemias for which no standard therapy options were available,<br />

and acceptable hepatorenal function. A standard 3�3 dose escalation<br />

design was used with 40% dose increments. TH-302 was administered IV<br />

over 30-60 minutes daily on Days 1-5 <strong>of</strong> a 21-day cycle. The objectives<br />

were to determine the MTD and PK pr<strong>of</strong>ile <strong>of</strong> TH-302 with this schedule<br />

and to assess preliminary clinical activity <strong>of</strong> TH-302. Results: 34 subjects<br />

with previously treated AML (n�26), ALL (n�6) or CML in blast phase<br />

(n�1) received TH-302 at doses <strong>of</strong> 120 (n�4), 170 (n�4), 240 (n�3),<br />

330 (n�3), 460 (n�16) or 550 (n�4) mg/m². No skin or mucosal toxicity<br />

was noted in participants treated with TH-302 doses �240 mg/m2 .At330<br />

mg/m2 , grade 2 dermatological toxicities included skin ulcer (n�1) and<br />

hand/foot syndrome (n�1). Grade 2 mucositis (n�3) and grade 2 skin<br />

toxicity (e.g. skin rash, skin ulcers; n�3) were reported at 460 mg/m2 ; none<br />

were dose-limiting. Two <strong>of</strong> 3 evaluable subjects treated at the 550 mg/m2 cohort experienced DLTs <strong>of</strong> grade 3 esophagitis. Eight subjects had stable<br />

disease or better after 1 cycle. One ALL subject (at 120 mg/m2 ) cleared<br />

marrow blasts with persistent neutropenia. One AML subject (at 550<br />

mg/m2 ) achieved CRp after 1 cycle with resolution <strong>of</strong> leukemia cutis.<br />

Conclusions: TH-302 administered daily for 5 consecutive days every 3<br />

weeks is well-tolerated with increased incidence <strong>of</strong> skin and mucosal<br />

toxicity at higher dose levels. <strong>Clinical</strong> activity has been noted with a few<br />

objective responses, but majority <strong>of</strong> cytoreductions in the AML subset were<br />

transient. <strong>Clinical</strong> trials combining TH-302 with various chemotherapeutics<br />

with established efficacy in AML and ALL are planned.<br />

6587 General Poster Session (Board #20F), Mon, 1:15 PM-5:15 PM<br />

<strong>Clinical</strong> impact <strong>of</strong> dose intensity <strong>of</strong> initial tyrosine kinase inhibitor (TKI)<br />

therapy in accelerated-phase CML (CML-AP). Presenting Author: Maro<br />

Ohanian, University <strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: For patients (pts) presenting with CML-AP, TKIs are recommended.<br />

Although generally well tolerated, dose reductions and/or interruptions<br />

are <strong>of</strong>ten required. The impact <strong>of</strong> TKI dose intensity (DI) on CML-AP<br />

outcome has not been described. Aim: To determine impact <strong>of</strong> TKI dose<br />

interruptions and/or reductions on outcome in de novo CML-AP. Methods:<br />

Overall, 58 CML-AP pts (median age 46 yrs) were treated on consecutive or<br />

parallel clinical trials with TKIs as initial therapy: imatinib (n�36),<br />

dasatinib (n�5), or nilotinib (n�17). CML-AP features included: blasts<br />

�15% (n�8), basophils �20%, (n�22), platelets �100x109 /L (n�3),<br />

cytogenetic clonal evolution (n�22), or �1 feature (n�3). We examined<br />

the impact <strong>of</strong> dose reductions, treatment interruptions, and DI (ratio <strong>of</strong> the<br />

dose received vs. the intended dose for the entire treatment duration) on<br />

event-free (EFS), transformation-free (TFS), and overall survival (OS).<br />

Results: Among 58 pts, 37 required treatment interruptions and/or dose<br />

reductions (29 required dose reductions). 12 pts required dose reductions<br />

and/or interruptions due to hematologic toxicity and 23 for nonhematologic<br />

toxicities. Pts were divided into 3 DI tiers: 100% (n� 21),<br />

90-99%, (n�10), and �90% (n�27). Outcomes were analyzed based on<br />

DI tiers and presence or absence <strong>of</strong> dose reductions as shown in the table.<br />

Pts with dose reductions and/or interruptions for hematologic toxicities vs.<br />

non-hematologic toxicities had a 24-mo EFS rate <strong>of</strong> 87% vs. 100%<br />

(p�0.05). 24-mo EFS by dose reductions and/or interruptions within 6 mo<br />

or later were 100% and 91%, respectively (p �0.7). Conclusions: DI has no<br />

significant impact on de novo CML-AP outcome. However, dose interruptions<br />

and/or reductions for hematologic toxicities are associated with worse<br />

24-mo EFS, suggesting myelosuppression may in some patients be more a<br />

feature <strong>of</strong> the disease than toxicity from therapy.<br />

Outcome % Dose reduced Not reduced P value 100% 90-99%


438s Leukemia, Myelodysplasia, and Transplantation<br />

6588 General Poster Session (Board #20G), Mon, 1:15 PM-5:15 PM<br />

Impact <strong>of</strong> ABL kinase domain mutations on the outcome <strong>of</strong> patients with<br />

chronic myeloid leukemia (CML). Presenting Author: Kendra Lynn Sweet,<br />

H. Lee M<strong>of</strong>fitt Cancer Center & Research Institute, Tampa, FL<br />

Background: Patients with CML who develop resistance to imatinib commonly<br />

have mutations in the BCR-ABL kinase domain (KDM). Studies<br />

looking at outcomes in patients with P-loop versus non-P-loop mutations<br />

within the ABL-Kinase Domain have produced conflicting results. Methods:<br />

The Total Cancer Care (TCC) database was used to identify patients with<br />

CML treated at M<strong>of</strong>fitt Cancer Center (MCC). Descriptive data were<br />

reported, chi square test was used for categorical variables, and Kaplan<br />

Meier curves were used for OS and PFS. Log rank test was used to compare<br />

survival times between groups. Results: Between 1992 and 2011, 540<br />

CML patients were treated at MCC. Of those, 51% were male and 71% were<br />

under the age <strong>of</strong> 60. Sixty percent (n�322) were diagnosed after 2001. Of<br />

the 540 patients, 6.5% (n�35) were found to have mutations <strong>of</strong> which 26<br />

were detected in patients diagnosed after 2001. Of the 35 patients, 74%<br />

(n�26) had single mutations and 26% (n�9) had compound mutations.<br />

P-loop mutations were seen in 17% (n�6) and 43% (n�15) had T315I<br />

mutations. Patients with KDM progressed to accelerated or blast phase in<br />

46% (n�16) <strong>of</strong> cases compared to 27% (n�136) without mutations<br />

(p�0.03). Median OS was 126 months, 109 months, and not reached in<br />

patients with P-loop, T315I, and non-P-loop mutations respectively<br />

(p�0.17). The corresponding median PFS was 85 months, 89 months, and<br />

not reached (p�0.20). In patients with one mutation median OS was not<br />

reached compared to 105 months in patients with compound mutations<br />

(p�0.27). After 2001, patients with KDM had a median PFS <strong>of</strong> 75 months<br />

and OS <strong>of</strong> 126 months while neither was reached in the non-mutation<br />

cohort (p�0.007, p�0.26 respectively). Median PFS in patients with<br />

single mutations was 85 months versus 10 months in those with compound<br />

mutations (p�0.037). Patients with KDM had additional Ph� clones on<br />

cytogenetics in 49% <strong>of</strong> cases compared with 19% <strong>of</strong> cases in the<br />

non-mutation group (P � 0.005). Conclusions: T315I and P-loop KDM<br />

predict PFS and OS in CML patients, and convey a trend for worse<br />

prognosis. The presence <strong>of</strong> additional Ph� clones in patients with<br />

BCR-ABL KDM indicates a higher level <strong>of</strong> genetic instability and clonal<br />

evolution, which may be the contributing factor to poor outcomes.<br />

6590 General Poster Session (Board #21A), Mon, 1:15 PM-5:15 PM<br />

The large granular lymphocytic leukemia registry: A detailed analysis <strong>of</strong> 79<br />

patients with LGL leukemia and rheumatoid arthritis. Presenting Author:<br />

Kirsten Marie Boughan, Drexel University College <strong>of</strong> Medicine, Philadelphia,<br />

PA<br />

Background: The purpose <strong>of</strong> this study is to analyze patients enrolled in the<br />

LGL leukemia registry to distinguish the similarities between LGL leukemia<br />

and rheumatoid arthritis in order to access overlapping immune mechanisms<br />

that may be responsible for neutrophil mediated destruction.<br />

Methods: A retrospective chart review was performed on 79 patients<br />

enrolled in the LGL registry at Penn State Cancer Institute. All patients<br />

enrolled in the study had a diagnosis <strong>of</strong> both rheumatoid arthritis and<br />

potentially LGL leukemia. Data was collected for age, sex, RF factor<br />

positivity, family history, autoimmune disease, T-cell receptor gene rearrangement,<br />

and bone marrow invasion. Results: Of 79 patients the mean<br />

age <strong>of</strong> onset for LGL leukemia was 60 years old with no discrepancy noted<br />

between sexes, 37 M, 42 F. 49 patients were positive for rheumatoid<br />

factor. 27 patients had rheumatoid arthritis in a first degree relative with no<br />

discrimination between maternal or paternal inheritance. 22 patients were<br />

positive for any other autoimmune process. 60 patients were positive for<br />

T-cell receptor gene rearrangement. Of the remaining 19 patients that were<br />

negative for T-cell receptor rearrangement, 12 had evidence <strong>of</strong> bone<br />

marrow invasion (CD3/CD8� infiltrate in �20% bone marrow) and two<br />

showed bone marrow invasion <strong>of</strong> NK cell LGL (CD3/CD8-, CD57�) (Table).<br />

Conclusions: Patients with T cell LGL leukemia and rheumatoid arthritis<br />

appear to be clinically similar with regard to age, duration <strong>of</strong> disease, and<br />

other autoimmune disorders as patients with rheumatoid arthritis alone.<br />

Our patient population showed those with TLGL and RA also tends to have a<br />

positive family history <strong>of</strong> RA in up to 20% as opposed to 5-10% in RA<br />

patients. Given that RA and TLGL have a significantly higher expression <strong>of</strong><br />

the HLA-DR4 haplotype than healthy patients, it is conceivable that with<br />

shared genetic alterations, and gene environment interactions that may<br />

promote posttranslational modification, there may be a loss <strong>of</strong> tolerance<br />

resulting in T cell activation, and eventual transformation into a T cell<br />

clone.<br />

LGL/RA No. (%) Bone marrow invasion No. (%)<br />

N 79 74<br />

TCR � 60 (75%) 72 (97%)<br />

TCR- 19 (24%) 12 (16%)<br />

6589 General Poster Session (Board #20H), Mon, 1:15 PM-5:15 PM<br />

A phase I/II trial <strong>of</strong> combination <strong>of</strong> PKC412 and 5-azacytidine (AZA) for the<br />

treatment <strong>of</strong> patients with refractory or relapsed (R/R) acute myeloid leukemia<br />

(AML) and myelodysplastic syndrome (MDS). Presenting Author: Aziz Nazha,<br />

University <strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: Midostaurin (PKC412) is a FLT3 kinase inhibitor with activity in<br />

acute myeloid leukemia (AML). Hypomethylating agents play an important role<br />

in the treatment <strong>of</strong> AML and MDS. We investigated the safety (phase I) and<br />

clinical activity (phase II) <strong>of</strong> combination <strong>of</strong> 5-azacytidine (AZA) and PKC412 in<br />

pts with R/R AML and MDS. Method: Pts �18 years with MDS, CMML or AML,<br />

who failed prior therapies with performance status �2, adequate liver (bilirubin<br />

� 2x ULN, ALT � 2.5x ULN) and renal (creatinine � 2x ULN) functions were<br />

eligible. Pts received AZA 75 mg/m2 subcutaneously or intravenously for 7 days<br />

<strong>of</strong> each cycle (Days 1-7) and PKC412 at 25 mg (cohort 1) and 50 mg (cohort 2;<br />

target dose) orally twice daily for 14 days (Days 8-21). Pts were to receive up to<br />

12 cycles if benefit from treatment. Supportive care was standard. Results: 14<br />

pts were included in phase I. 1 pt was inevaluable for DLT (withdrawal before<br />

completing cycle#1). 6 pts treated in cohort 1 and 7 in cohort 2. 1 pt in cohort 2<br />

received PKC412 dose as per cohort 1 dose by pt error. Pt characteristics and<br />

responses are summarized in the Table. The overall response rate (ORR) was<br />

3/13(21%) (2 with CRi, 1 pt dropped BM blasts form 27% to 7% after 2 cycles<br />

and went to transplant. 1/4 with FLT3-ITD achieved CRi, 2 <strong>of</strong> the non-responders<br />

received prior FLT3 inhibitors (1 had developed FLT3 D835). All toxicities were<br />

grade 1 (nausea 1 pt, vomiting 1 pt, dry skin 1 pt, and rash 1 pt) with no<br />

difference between the 2 groups. No DLT was identified. Conclusions:<br />

PKC412�AZA is safe and well tolerated at the intended doses with good ORR in<br />

high risk pts with R/R AML. Phase II study is enrolling pts with FLT3-ITD.<br />

Updated result will be presented.<br />

Patients characteristics and responses.<br />

Parameter<br />

Number 14<br />

Age, years (range) 61 (38-86)<br />

Median N <strong>of</strong> prior therapies, N (range) 1 (1-7)<br />

Dose level, N<br />

Cohort 1 6<br />

Cohort 2 7<br />

Inevaluable (cohort 2) 1<br />

Median N <strong>of</strong> cycles (range) 2 (1-5)<br />

AML history, N (%)<br />

de novo 6 (43)<br />

MDS related 7 (50)<br />

Therapy-related AML 1(7)<br />

Cytogenetic risk group, N (%)<br />

Intermediate 5 (31)<br />

Unfavorable 9 (69)<br />

FLT3-ITD status, N (%)<br />

Positive 4 (29)<br />

Negative 9 (64)<br />

ND 1(7)<br />

Response rate, N (%)<br />

CRi 2 (14)<br />

PR 1(7)<br />

SD 3 (21)<br />

6591 General Poster Session (Board #21B), Mon, 1:15 PM-5:15 PM<br />

Pharmacokinetics (PK) and pharmacodynamics (PD) <strong>of</strong> rituximab administered<br />

by faster infusion in patients with previously untreated diffuse large<br />

B-cell (DLBCL) or follicular lymphoma (FL). Presenting Author: Michael<br />

Brewster, Roche Products Limited, Welwyn Garden City, United Kingdom<br />

Background: Faster infusion <strong>of</strong> rituximab (R) can improve patient (pt)<br />

convenience and reduce strain on oncology unit resources. To assess the<br />

safety, PK and PD <strong>of</strong> a 90-min infusion rate for R in NHL pts, a prospective,<br />

open-label, phase III, multicenter, single-arm trial (RATE) was conducted.<br />

Safety results have been previously presented (ASH 2011). Methods: Pts<br />

(�18 yrs old) with untreated DLBCL or FL, without clinically significant<br />

cardiovascular disease, received R-chemotherapy. R (375 mg/m2 ) was<br />

given at the standard infusion rate (4–6 h) in Cycle 1. Pts who did not<br />

experience Grade 3/4 infusion-related reactions (IRRs) and who had<br />

circulating lymphocyte counts � 5000/�l before Cycle 2, received subsequent<br />

R (375 mg/m2 ) infusions at a 90-min rate: 20% over 30 min then<br />

80% over the next 60 min. Pts (n�14) who completed all cycles at the<br />

90-min infusion rate gave further serum samples up to 16 wks after the last<br />

cycle for determination <strong>of</strong> PK parameters, including R terminal half-life<br />

(t½), maximum serum concentration (Cmax), systemic clearance (CL) and<br />

volume <strong>of</strong> distribution (V). Results: For pts (n�365) that received the<br />

90-min infusion rate at Cycle 2, serum R concentrations increased over the<br />

course <strong>of</strong> the study. Estimated median values were: CL, 107 ml/d; V at<br />

steady state, 3830 ml; t½, 24.6 d. At Cycle 2, 50.5% <strong>of</strong> pts had<br />

undetectable CD19� levels; this percentage increased during the study<br />

(68.3% Cycle 6, 87.5% Cycle 8). Conclusions: Peak R levels were higher<br />

than seen in published data <strong>of</strong> q3w R regimens but comparable with levels<br />

seen for q1w R regimens. The estimated median values for CL, V, t½ and B<br />

cell (CD19�) depletion with the 90-min infusion rate were similar to<br />

previously reported estimates for infusions at the standard rate. These<br />

results, in combination with the safety pr<strong>of</strong>ile previously reported, show<br />

that a faster infusion <strong>of</strong> R can be an acceptable option for selected NHL pts<br />

who tolerated the standard infusion rate at Cycle 1 without a severe IRR.<br />

Serum R concentration (�g/ml)<br />

Cycle 2, Day 1 Cycle 6, Day 1 Cycle 8, Day 1<br />

Trough Peak Trough Peak Trough Peak<br />

N 329 335 241 238 34 36<br />

Mean (SD) 30 (26) 228 (64) 80 (35) 275 (72) 92 (51) 299 (91)<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


6592 General Poster Session (Board #21C), Mon, 1:15 PM-5:15 PM<br />

Phase II study <strong>of</strong> omacetaxine (OM) and low-dose AraC (LDAC) in older<br />

patients with acute myelogenous leukemia (AML) and high-risk myelodysplastic<br />

syndrome (MDS). Presenting Author: Tapan M. Kadia, University <strong>of</strong><br />

Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: Treatment <strong>of</strong> AML in patients (pts) � 70 yrs <strong>of</strong> age with<br />

intensive chemotherapy is associated with high rates <strong>of</strong> early mortality and<br />

little benefit. Newer, lower-intensity approaches with novel mechanisms<br />

are needed. OM is a semisynthetic plant alkaloid which has demonstrated<br />

activity in AML as a single agent and with chemotherapy. Methods: We<br />

studied a low intensity program combining subcutaneous (SQ) OM with SQ<br />

LDAC in pts �/� 60 yrs, with AML or MDS, a performance status (PS) <strong>of</strong><br />

�/�2, and adequate organ function. Initially, 6 pts were enrolled at the<br />

following doses: OM 1.25 mg/m2 SQ Q12 hrs x 3 days with AraC 20 mg SQ<br />

Q12 hrs x 7 days on a 4 week cycle. If safety is confirmed, the phase II<br />

portion would commence at the safe dose levels. Up to 12 courses can be<br />

given. The primary endpoint was to determine the complete remission (CR)<br />

rate. Secondary endpoints were: CR duration, DFS, OS, safety, and early<br />

mortality. Results: 17 pts were enrolled on study so far. The median age was<br />

74 yrs (range, 64-81); the median PS was 1 (0-1). The karyotypes in these<br />

pts were: diploid in 6 (35%), complex with chromosome (chr) 5 and/or 7<br />

abnormality (abnl) in 4 (24%), complex without chr 5 and/or 7 abnl in 2<br />

(12%), 11q abnl in 1 (6%), poor metaphases in 1 (6%), and other in 3<br />

(18%). Four pts with prior MDS were treated with a median <strong>of</strong> 2 prior<br />

therapies (1-3). Median bone marrow blast % at the start <strong>of</strong> therapy was 40<br />

(15-87). The median WBC, hemoglobin, and platelets were 2.1 (0.4–<br />

24.8), 8.9 (7.7–10.7), and 45 (14–104), respectively. These pts have<br />

received a median <strong>of</strong> 1 (1-3) cycle <strong>of</strong> therapy. Of the 11 pts evaluable for<br />

response, there were 2(18%) CR, 1(9%) CRp, 1(9%) PR for an ORR <strong>of</strong><br />

4/11 (36%). Five pts had no response and were taken <strong>of</strong>f study. Two pts<br />

died on study: 1 on day 6 and 1 on day 27. Both pts were 74 yrs with a<br />

complex karyotype. One died <strong>of</strong> pneumonia and multi-organ failure and the<br />

2nd died from cardiac arrest. Other than the deaths, serious adverse events<br />

included grade 3 transaminitis in 1 and grade 3 heart failure in 1.<br />

Conclusions: OM and LDAC appears to be tolerable in older pts with AML.<br />

The combination appears to have activity. Stopping boundaries for futility<br />

and safety have not been breached. Enrollment is ongoing.<br />

6594 General Poster Session (Board #21E), Mon, 1:15 PM-5:15 PM<br />

Monitoring and switching patterns in chronic myelogenous leukemia (CML)<br />

pts treated with imatinib (IM): A chart review analysis. Presenting Author:<br />

Lei L. Chen, Novartis Pharmaceuticals, East Hanover, NJ<br />

Background: The objective <strong>of</strong> this study is to evaluate compliance to<br />

National Comprehensive Cancer Network (NCCN) and European LeukemiaNet<br />

(ELN) guidelines regarding monitoring frequency and switching<br />

practices in CML patients (pts) treated with IM in community practices.<br />

Methods: Physicians treating CML pts were selected from 28 community<br />

practices throughout the USA. Each physician reviewed and extracted up to<br />

15 pt charts <strong>of</strong> CML pts in chronic phase not previously enrolled in clinical<br />

trials. Pts were required to have initiated 1st-line IM between 1/2006 -<br />

10/2010. Data on 1st-line IM treatment response monitoring frequency<br />

and outcomes, and corresponding therapy switching patterns were extracted.<br />

Results: Information was collected on treatment monitoring and<br />

response from a total <strong>of</strong> 297 pt charts. Median IM duration was 320 days.<br />

Percentages <strong>of</strong> pts monitored according to both guidelines are reported. In<br />

addition, the proportion <strong>of</strong> pts who missed milestones and the proportion <strong>of</strong><br />

pts who did not switch to a 2nd-line tyrosine kinase inhibitor (TKI) when<br />

they missed milestones are reported. Conclusions: There is considerable<br />

undermonitoring <strong>of</strong> CML pts receiving IM, which is likely to result in a<br />

significant portion <strong>of</strong> pts with suboptimal response left undetected. The<br />

switching rate to 2nd-line TKI among pts with suboptimal response to IM is<br />

much lower than recommended by either NCCN or ELN guidelines.<br />

Milestone<br />

Mos Response % Monitored<br />

% Missed the<br />

milestone (among<br />

monitored pts)<br />

% Switched to<br />

2nd-line TKI<br />

(among pts who<br />

missed milestone)<br />

0-3 CHR †‡ 4-6 PCyR<br />

(N�297) 98.7 10.2 16.7<br />

†‡ 7-12 CCyR<br />

(N�285) 60.4 11.6 5.0<br />

†‡ 13-18 CCyR<br />

(N�284) 61.3 29.9 17.3<br />

† Pts previously achieved CCyR (N�145) 39.3 1.8 0.0<br />

Pts did not achieve CCyR* (N�119) 38.7 32.6 33.3<br />

19-24 Pts previously achieved CCyR (N�172) 36.6 3.2 0.0<br />

Pts did not achieve CCyR* (N�73) 13.7 30.0 33.3<br />

25-30 Pts previously achieved CCyR (N�156) 23.7 2.7 100.0<br />

Pts did not achieve CCyR* (N�83) 7.2 50.0 66.7<br />

31-36 Pts previously achieved CCyR (N�158) 22.8 2.8 100.0<br />

0-12 MMR<br />

Pts did not achieve CCyR* (N�77) 5.2 25.0 0.0<br />

‡ (N�297) 71.7 28.6 9.8<br />

13-18 (N�264) 72.7 13.0 12.0<br />

19-36 (N�245) 71.8 9.7 23.5<br />

† NCCN; ‡ ELN; * Did not achieve CCyR/not previously monitored.<br />

Leukemia, Myelodysplasia, and Transplantation<br />

439s<br />

6593 General Poster Session (Board #21D), Mon, 1:15 PM-5:15 PM<br />

Prognostic and predictive significance <strong>of</strong> smudge cell percentage on<br />

routine blood smear in chronic lymphocytic leukemia patients: A single<br />

center study from northern India. Presenting Author: Ajay Gogia, AIIMS,<br />

New Delhi, India<br />

Background: Smudge cells are ruptured lymphocytes seen on routine blood<br />

smears <strong>of</strong> chronic lymphocytic leukemia (CLL) patients. We evaluated<br />

prognostic and predictive significance <strong>of</strong> smudge cells percentage on a<br />

blood smear in CLL patients. Methods: We calculated smudge cell percentages<br />

(ratio <strong>of</strong> smudged to intact cells plus smudged lymphocyte) on<br />

archived blood smears <strong>of</strong> 185 untreated CLL patients registered at I.R.C.H,<br />

AIIMS, New Delhi over a period <strong>of</strong> 11 years. Results: There were 135 males<br />

and 50 females. The median age was 60 years (28-89). Median absolute<br />

lymphocyte count was 42 X109 /L. <strong>Clinical</strong> Rai stage distribution was: stage<br />

0 - 10%, stage I - 15%, stage II - 40%, stage III -15 % and stage IV - 20%.<br />

The median smudge cells percentage was 27% (4% to 76%). There was no<br />

correlation <strong>of</strong> proportion <strong>of</strong> smudge cells with age, sex, lymphocyte count,<br />

organomegaly, ZAP 70 � or CD 38 � CLL patients, but there was<br />

significant correlation with stage <strong>of</strong> disease. Median smudge cell percentage<br />

in stage 0 & I -33% (12-76), stage II- 31% (12-61) and stage<br />

III&IV-21% (4-51) [ p��0.001]. One third <strong>of</strong> early stage (0, I &II) patients<br />

required treatment during follow up, at the end <strong>of</strong> 5 years <strong>of</strong> follow up 55%<br />

required treatment with smudge cell � 30%, against 24% patients<br />

requiring treatment with smudge cells � 30% [p�0.01]. The percentage <strong>of</strong><br />

smudge cells as a continuous variable correlated with OS [HR 0.96, p�<br />

0.001]. The 5-year survival rate was 51% for patients with 30% or less<br />

smudge cells compared with 81% for patients with more than 30% <strong>of</strong><br />

smudge cells. Thirty percent <strong>of</strong> patients died during follow up. Median OS<br />

was 5 years with median follow up period <strong>of</strong> 3.9 years. Smudge cells<br />

percentage (�30% vs. �30%) had significant association with OS [HR<br />

0.47, 95%CI (0.32-0.71), p�0.001)]. Conclusions: Simple and inexpensive<br />

detection <strong>of</strong> smudge cells on blood smears on routine diagnostic test<br />

useful in predicting progression free and OS in CLL patients and may be<br />

beneficial in countries with limited recourses.<br />

6595 General Poster Session (Board #21F), Mon, 1:15 PM-5:15 PM<br />

Treatment outcome <strong>of</strong> acute myeloid leukemia (AML) in HIV� patients.<br />

Presenting Author: Irene Ang Dy, Leukemia Program, Continuum Cancer<br />

Centers <strong>of</strong> New York, New York, NY<br />

Background: AML is diagnosed more frequently in HIV� patients than<br />

previously. The results with standard AML treatment in such patients have<br />

not been evaluated. We evaluated whether the results justify standard<br />

aggressive treatment <strong>of</strong> AML in HIV� patients. Methods: We identified 5<br />

HIV� patients at our institution who were subsequently diagnosed with<br />

AML and 68 previously reported HIV� patients with AML through PubMed<br />

from 1986-2011. The median age at the time <strong>of</strong> AML diagnosis was 40<br />

years (range 7-70 years). Of the 26 patients with known karyotype, 7 had<br />

favorable, 7 intermediate and 12 had unfavorable cytogenetics. The<br />

majority <strong>of</strong> treated patients received standard intensive induction therapy<br />

and complete responders (CR) received consolidation therapy. HIV was<br />

pre-, post-, and concurrently diagnosed in 47, 2 and 19 AML patients<br />

respectively. The Kaplan-Meier method examined whether CD4 count, AML<br />

treatment and CR attainment affected overall survival. Cox proportional<br />

hazard modeling, adjusted for age and CD4 count determined whether AML<br />

treatment and CR attainment were associated with death from AML.<br />

Results: The final analysis included pre- and concurrently HIV diagnosed<br />

AML patients (n�66). HIV infection occurred at a median <strong>of</strong> 5 years (range<br />

0.25-28 years) prior to the diagnosis <strong>of</strong> AML in 47 patients. The most<br />

common FAB types were M4 (22.6%) and M2 (22.6%). CR was achieved in<br />

71.7% (n�33/46) <strong>of</strong> treated patients and 51.5% (n�17/33) <strong>of</strong> them<br />

relapsed with a median CR duration <strong>of</strong> 9.2 months. Median survival <strong>of</strong><br />

patients with CD4 count � 200 and � 200 was 7 vs. 13.4 months<br />

(p�0.03); median survival <strong>of</strong> untreated and treated patients was 1.0<br />

vs.13.2 months (p � 0.001) and median survival <strong>of</strong> treated patients who<br />

did and did not achieve CR was 2 vs. 21 months respectively (p � 0.001).<br />

All treated patients and those who achieved CR were less likely to die from<br />

AML than untreated patients and those who failed to respond (H.R�0.05;<br />

95% CI, 0.01-0.17 and H.R. � 0.11; 95% C.I, 0.04-0.35, respectively).<br />

Conclusions: Standard AML treatment and CR were associated with longer<br />

survival in HIV� patients regardless <strong>of</strong> CD4 count. More than half <strong>of</strong><br />

patients studied achieved CR. HIV� AML patients should be <strong>of</strong>fered<br />

standard AML therapy.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


440s Leukemia, Myelodysplasia, and Transplantation<br />

6596 General Poster Session (Board #21G), Mon, 1:15 PM-5:15 PM<br />

Omacetaxine mepesuccinate in chronic-phase chronic myeloid leukemia<br />

(CML) in patients resistant, intolerant, or both to two or more tyrosinekinase<br />

inhibitors (TKIs). Presenting Author: Luke Paul Akard, Indiana<br />

Blood and Marrow Transplantation, Indianapolis, IN<br />

Background: Omacetaxine mepesuccinate (“omacetaxine”) is a first-inclass,<br />

reversible, transient inhibitor <strong>of</strong> protein elongation that facilitates<br />

tumor cell death without depending on BCR-ABL signaling. <strong>Clinical</strong> activity<br />

was shown in two phase 2, open-label, multicenter studies <strong>of</strong> patients with<br />

treatment-resistant CML who had failed at least prior imatinib, many <strong>of</strong><br />

whom were also resistant to or intolerant <strong>of</strong> dasatinib and/or nilotinib.<br />

Methods: A subset <strong>of</strong> data from the phase 2 studies included patients in<br />

chronic phase who were resistant/intolerant to �2 approved TKIs. Omacetaxine<br />

1.25 mg/m2 was given subcutaneously twice daily: �14 consecutive<br />

days/28-day cycle for induction, �7 days/cycle as maintenance.<br />

Patients had never achieved or lost response to �2 TKIs (R group), were<br />

intolerant <strong>of</strong> �2 TKIs (I), or resistant to 1 and intolerant <strong>of</strong> another (R/I)<br />

were evaluated. Results: Of 81 patients (median age, 58 years), 69 were R,<br />

7 I, and 5 R/I. Major cytogenetic response occurred in 13 (19%) in the R<br />

group (median duration not reached), 2 (29%) I (median duration 7.4<br />

months), and 1 (20%) R/I (duration 17.7 months). For all patients, cycles<br />

<strong>of</strong> exposure and study duration were 7 cycles and 9.1 months (R); 4 cycles,<br />

7.3 months (I); and 2 cycles, 2.3 months (R/I). Median overall survival in<br />

months were 33.9 (R), not reached (I), and 25.0 (R/I). Of 66 (81%)<br />

patients with treatment-related grade 3/4 adverse events (AEs), the most<br />

common were thrombocytopenia in 44 R, 6 I, and 4 R/I patients and<br />

neutropenia in 32 R, 4 I, and 1 R/I. Fifteen patients had an AE leading to<br />

discontinuation (10 R, 2 I, 3 R/I), primarily disease progression. There were<br />

9 deaths (the most common were disease progression, sepsis), 8 I, 1 R/I; 2<br />

were considered related to treatment (both sepsis). Conclusions: This<br />

subset analysis <strong>of</strong> patients with chronic-phase CML and prior therapy with<br />

�2 TKIs shows that omacetaxine provides efficacy and tolerability across<br />

TKI-R, I, and R/I groups. Interpretation <strong>of</strong> the I and the R/I group data was<br />

limited by small sample sizes. Support: Teva Pharmaceutical Industries<br />

Ltd.<br />

6598 General Poster Session (Board #22A), Mon, 1:15 PM-5:15 PM<br />

Comparison <strong>of</strong> fludarabine (F), cyclophosphamide (C), and rituximab (R)<br />

versus FCR plus bevacizumab (B) in relapse chronic lymphocytic leukemia<br />

(CLL). Presenting Author: Hun Ju Lee, University <strong>of</strong> Texas M. D. Anderson<br />

Cancer Center, Houston, TX<br />

Background: There is significant data to show that vascular endothelial<br />

growth factor (VEGF) is important in the development and progression <strong>of</strong><br />

CLL. Bevacizumab (B) is an anti-VEGF monoclonal antibody associated<br />

with improvement in progression free survival (PFS) when combined with<br />

chemotherapy in pts with solid malignancies. FCR is the most active<br />

chemoimmunotherapy in treatment <strong>of</strong> CLL. We report data on a phase II<br />

trial combining FCR-B and comparing the results to those seen in a trial<br />

with FCR for pts with relapsed CLL (Badoux et al. Blood, March 2011).<br />

Methods: FCR consisted <strong>of</strong> F: 25 mg/m2 and C: 250 mg/m2 on D2-4; R: 375<br />

mg/m2 on D1 (for the first course) and 500 mg/m2 for courses 2-6, every 4<br />

weeks for 6 courses. FCR-B consisted as above for FCR and (B) 10 mg/kg<br />

was given on D3 <strong>of</strong> each cycle. Indications for treatment and response<br />

assessment were according to 1996 NCI-WG guidelines for both groups.<br />

Results: Baseline characteristics, complete remission (CR), overall response<br />

rate (ORR), PFS and OS <strong>of</strong> relapsed CLL pts are shown (Table).<br />

Conclusions: In relapsed pts with CLL, FCR-B showed a trend toward<br />

improved PFS compared to FCR.<br />

Characteristics FCR-B (%)[range] FCR (%)[range] P-value<br />

N 62 284<br />

Median age, yrs 64 [30-84] 60 [31-84]<br />

Median # <strong>of</strong> prior treatments 2 [1-5] 2 [1-10]<br />

Alkylating agent and (F) refractory 5 (8) 33 (11)<br />

Prior exposure to FCR 52 (84) 78 (27)<br />

Rai stage<br />

0-II 26 (42) 154 (54)<br />

III 9 (14) 34 (12)<br />

IV 27 (44) 96 (34)<br />

Karyotype/FISH<br />

Diploid/13q- 22 (36) 97 (34)<br />

11q- 25 (40) 16(6)<br />

� 12 3 (5) 13 (5)<br />

Complex 1 (2) 22 (8)<br />

Abn 17p 11 (18) 20 (7)<br />

Others 0 14 (5)<br />

Response<br />

CR 13 (21) 85 (30)<br />

ORR 44 (71) 210 (74)<br />

Overall Median (mo) [95% CI]<br />

PFS 34 [Not reached (NR)] 23 [18-27] Not significant (NS)<br />

OS NR 47 [40-53] NS<br />

Prior chemoimmunotherapy (FCR)<br />

PFS 34 [16-52] 20 [17-22] NS<br />

OS 38 [NR] 43 [36-49] NS<br />

6597 General Poster Session (Board #21H), Mon, 1:15 PM-5:15 PM<br />

FLT3 mutations in myelodysplastic syndromes (MDS) and chronic myelomonocytic<br />

leukemia (CMML). Presenting Author: Pavan Kumar Bhamidipati,<br />

Synergy Medical Education Alliance/Michigan State University,<br />

Saginaw, MI<br />

Background: FLT3 mutations occur in one third <strong>of</strong> acute myeloid leukemia<br />

(AML) patients (pts) and predict poor outcome. The incidence and impact<br />

<strong>of</strong> FLT3 in MDS/ CMML is unknown. Methods: We conducted a retrospective<br />

review at MDACC to identify FAB MDS/ CMML pts with FLT3 mutations at<br />

diagnosis. Results: From 1996 to 2010; 2052 MDS/ CMML pts had<br />

mutation analysis. 45 (2.2%) had FLT3 mutations (internal tandem<br />

duplication-ITD or D835) at diagnosis. 29 pts had MDS and 16 had CMML.<br />

Median (Med) age was 64 years (21 to 83) and 69% were males. FAB<br />

groups: 3 pts with refractory anemia (RA), 11 pts with refractory anemiaexcess<br />

blasts (RAEB), 18 pts with refractory anemia-excess blasts in<br />

transformation (RAEB-T) and 13 pts with CMML. IPSS: 3 in Low (7%), 16<br />

in Int-1 (36%), 11 in Int-2 (24%), and 15 in High (33%). Med white count,<br />

hemoglobin, platelet count and marrow blast percent at diagnosis were 5.2<br />

x109 /l (1.2 to 211), 10.0 g/l (6.8 to 14.9), 78 x 109 /l (8 to 429), and 14%<br />

(1 to 28), respectively. FLT3 ITD and FLT3 D835 mutations were present<br />

in 32 (71%) and 13 pts (29%), respectively. Karyotype was diploid in 30<br />

(66%); -5/-7 in 5 (11%), 11q in 1 (2%), and others in 9 pts (19%). All 5<br />

pts with -5/ -7 had the ITD mutation. Concurrent mutations were identified<br />

in RAS, NPM1 and C-Kit in 6 (13%), 3 (7%) and 1 (2%) pt, respectively.<br />

Med overall survival (OS) for FLT3 pts was 15 months compared to 17<br />

months for non FLT3 pts (P�0.9). 18 pts had RAEB-T: 13 (72%) received<br />

AraC-based therapy and 3 (17%) received hypomethylating therapy (HMT)<br />

with complete remission (CR) in 14 pts (78%). 14 pts had RA/ RAEB: 5<br />

(36%) received AraC-based therapy and 7 (50%) received HMT with CR in<br />

5 (37%) and hematological improvement (HI) in 4 (28%). 13 pts had<br />

CMML: 4 (31%) received AraC-based therapy and 6 (46%) received HMT<br />

with CR in 3 (23%) and HI in 3 (23%). Repeat FLT3 was available on 16<br />

pts achieving any response and was absent/ decreased in 14 (88%), stable<br />

in 1 (6%) and increased in 1 (6%). Notably, the 14 pts with absent/<br />

decreased FLT3 had med OS <strong>of</strong> 27 months versus 12 months for remaining<br />

group (P�0.004). Conclusions: FLT3 occurs in MDS/ CMML at a lower<br />

frequency than AML and does not predict poor outcome. Pts who achieve<br />

absent/ decreased FLT3 seem to have significantly improved overall<br />

survival.<br />

6599 General Poster Session (Board #22B), Mon, 1:15 PM-5:15 PM<br />

Expression levels <strong>of</strong> nucleoside transporter hENT1 and dCK enzyme, as<br />

prognostic factors in patients with AML treated with cytarabine. Presenting<br />

Author: Carmen Corrales-Alfaro, Instituto Nacional de Cancerologia, Mexico<br />

City, Mexico<br />

Background: Cytarabine has been set as the main drug included in different<br />

schemas for the treatment <strong>of</strong> AML patients. This drug requires inflow to the<br />

cell by the nucleoside transporter hENT1 and its activation rate is limited<br />

by dCK enzyme. Therefore decreased expression levels <strong>of</strong> these genes may<br />

influence response rate to this drug. Methods: AML patients � 15 years,<br />

without previous treatment were included. After informed consent signature,<br />

peripheral blood was obtained and DNA was extracted by standard<br />

methodology. Amplification <strong>of</strong> hENT1 and dCK genes was done by RT-PCR.<br />

<strong>Clinical</strong> parameters were registered. All patients received cytarabine �<br />

doxorrubicine as induction regimen (7 � 3 schema). Descriptive statistics<br />

was used. Uni- and multivariate analysis was done to determine factors<br />

influencing on response and overall survival. This protocol was approved by<br />

local IRB. Results: From January till December 2011, 22 patients have<br />

been included. Median age was 36 years (15-72 y), 52.3 % were male.<br />

According with FAB classification, M2 subtype was the most frequent<br />

(28.5 %). All patients had an adequate performance status (ECOG 1 & 2 �<br />

34 & 66 %, respectively). Cytogenetic risk was considered as intermediate<br />

risk in 38 %, followed by unfavorable in 28 %. Complete response was<br />

achieved in 58 %. Higher hENT1 expression levels was the only factor<br />

influencing on response and survival by Spearmen correlation analysis.<br />

Conclusions: These are preliminary results but are highly suggestive that<br />

pharmacogenetic analysis regarding cytarabine inflow may be decisive in<br />

AML patients. Our results require to be confirmed with a larger sample, and<br />

a longer follow-up.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


6600 General Poster Session (Board #22C), Mon, 1:15 PM-5:15 PM<br />

Real-world study <strong>of</strong> imatinib treatment patterns and outcomes among<br />

veteran patients with chronic myeloid leukemia. Presenting Author: Lizheng<br />

Shi, Tulane University, New Orleans, LA<br />

Background: Imatinib (IM) is the most commonly used drug in the treatment<br />

<strong>of</strong> chronic myeloid leukemia-chronic phase (CML-CP) patients (pts). This<br />

study investigated the treatment patterns and outcomes for veteran<br />

CML-CP pts initiated on IM. Methods: Pts (age �18 yrs) with �1 CML<br />

diagnosis codes documented (ICD-9 CM: 205.1x) between 1/1/2000 and<br />

12/31/2010 were identified from the VISN 16 data warehouse. Pts were<br />

required to have initiated IM as the first-line therapy in CML-CP. Accelerated<br />

and blastic phases (A/BP) were identified based on WHO classification<br />

using CBC information. IM dose adjustments were assessed as dose<br />

increase from �400 mg to �600mg or from 400-600mg to �800 mg<br />

daily. Rates <strong>of</strong> IM discontinuation (no use <strong>of</strong> IM for �60 days) and<br />

switching to other drug therapy (dasatinib/nilotinib (DS/NL), hydroxyurea,<br />

and interferon-alpha) were estimated. Time to discontinuation, progression<br />

to A/BP, and survival were assessed using Kaplan-Meier analysis (K-M).<br />

Overall survival was measured from IM initiation while survival among pts<br />

with disease progression was measured from the date <strong>of</strong> progression.<br />

Results: Among the 137 pts selected, average age was 64.8 yrs and<br />

follow-up time was 4.0 yrs. 16.8% <strong>of</strong> pts had dose increase from �400mg<br />

to �600mg and 21.7% <strong>of</strong> these pts switched to other therapy after dose<br />

increase. 13.1% <strong>of</strong> pts had dose increase from 400-600mg to �800 mg<br />

with 22.2% <strong>of</strong> these pts switching to other therapy later. During the study,<br />

83.8% <strong>of</strong> the 74 pts who discontinued IM did not use other drug therapies;<br />

and 16.2% (12 pts) switched, among which 9 pts took DS/NL. K-M showed<br />

that 25.6% and 42.4% pts discontinued IM treatment by year 1 and 2 and<br />

8.1% and 16.0% pts experienced disease progression by year 1 and 2,<br />

respectively. Among the 28 patients who had disease progression, 32.1%<br />

continued IM use after progression and only 7.1% switched to other<br />

therapies (50% switching to DS/NL). The mortality rates were 3.0% and<br />

9.5% by year 1 and 2 after IM initiation, and 21.7% and 42.7% by year 1<br />

and 2 after disease progression, respectively. Conclusions: The majority <strong>of</strong><br />

IM-treated patients, including patients with disease progression, discontinued<br />

IM use without switching to other effective therapies.<br />

6602 General Poster Session (Board #22E), Mon, 1:15 PM-5:15 PM<br />

A phase II study <strong>of</strong> cl<strong>of</strong>arabine and daunorubicin in patients age 60 or older<br />

with newly diagnosed adult acute myeloid leukemia. Presenting Author:<br />

Carlos Enrique Vigil, Roswell Park Cancer Institute, Buffalo, NY<br />

Background: The outcome <strong>of</strong> acute myeloid leukemia (AML) patients (pts)<br />

�60 years (yrs) old remains poor, with a median overall survival <strong>of</strong> a few<br />

months. Neither the addition <strong>of</strong> other drugs during induction <strong>of</strong> the<br />

traditional 7�3 regimen nor an increase <strong>of</strong> cytarabine or daunorubicin<br />

doses has improved their overall survival. We have therefore designed a<br />

combination <strong>of</strong> cl<strong>of</strong>arabine and daunorubicin. Aims: Determine the safety<br />

and efficacy <strong>of</strong> cl<strong>of</strong>arabine and daunorubicin in pts � 60 yrs <strong>of</strong> age.<br />

Methods: Induction-cl<strong>of</strong>arabine 20 mg/m2 /d in a 1-hour IV for 5 days<br />

followed 3 hrs later by daunorubicin 50 mg/m2 IV over 5 minutes on days 1,<br />

3, and 5. Pts who did not achieve complete remission (CR) or CR without<br />

platelet recovery (CRp) were eligible for a second round <strong>of</strong> induction as<br />

above. Consolidation - 2 cycles <strong>of</strong> cl<strong>of</strong>arabine 20mg/m2 /d for 3 days and<br />

daunorubicin 50 mg/m2 /day on days 1 and 3. Results: Twenty-one pts were<br />

enrolled. The median age was 69 (range 60-85) yrs. Seven pts (34%) had<br />

secondary AML and 52% had complex karyotype. Twenty-one pts completed<br />

one induction cycle, one pt received a second induction due to<br />

residual disease and 6 pts underwent consolidation therapy. The principal<br />

toxicity was grade �3 infections and prolonged thrombocytopenia and<br />

neutropenia, which occurred in 18% and 21% <strong>of</strong> pts, respectively. Three<br />

deaths were documented from treatment complications (sepsis and fungal<br />

infections). Eight (38.1%) pts achieved either a CR or CRp. Responses<br />

were observed in 5/11 (45.5%) pts with high risk features (complex<br />

karyotype and/or presence <strong>of</strong> FLT3). The median disease free-survival was<br />

1.9 (range, 0.9-8) months and the median overall survival was 6.5 (range,<br />

4.2-14.2) months. The 1-yr overall survival was 35%. Conclusions: These<br />

preliminary results suggest that cl<strong>of</strong>arabine in combination with daunorubicin<br />

is safe, well-tolerated with minimal toxicity, and active for the upfront<br />

treatment <strong>of</strong> �60 yrs old AML pts, including those with adverse features.<br />

Future trials will explore additional modifications to this regimen in order to<br />

optimize therapy for this group <strong>of</strong> pts with high risk features and frequent<br />

treatment failure.<br />

Leukemia, Myelodysplasia, and Transplantation<br />

441s<br />

6601 General Poster Session (Board #22D), Mon, 1:15 PM-5:15 PM<br />

A comparison <strong>of</strong> CR versus CRi response following CPX-351 treatment <strong>of</strong><br />

newly diagnosed AML in elderly patients (pts). Presenting Author: Jeffrey E.<br />

Lancet, H. Lee M<strong>of</strong>fitt Cancer Center & Research Institute, Tampa, FL<br />

Background: A Phase 2b study randomized untreated elderly AML pts to<br />

CPX-351 or 7�3. CPX-351 improved leukemia clearance (88% v 71%,<br />

�5% marrow blasts), CR�CRi rate (67% v 51%), and was particularly<br />

effective in pts with adverse karyotype and antecedent hematologic<br />

disorders (sAML). Survival following CRi is usually inferior compared to CR.<br />

CPX-351 markedly prolongs plasma drug levels and maintains the 5:1<br />

molar ratio for optimal leukemic cell killing potentially delaying hematologic<br />

recovery among CRi patients. Consequently, we compared the<br />

characteristics and outcomes <strong>of</strong> pts achieving CR v CRi. Methods: Untreated<br />

AML pts, aged 60-75, PS� 0-2, SCr � 2.0 mg/dL, total bilirubin<br />

�2.0 mg/dL, ALT/AST �3 x ULN, and LVEF �50% were eligible. Pts were<br />

randomized 2:1 to receive up to 2 inductions and 2 consolidations with<br />

CPX-351 (100 u/m2 ; D 1, 3, 5; 90 min infusion) or 7�3 (cytarabine�100<br />

mg/m 2<br />

and daunorubicin�60 mg/m2 ). Consolidation with stem cell transplantation<br />

(SCT) was permitted. The 1o endpoint was CR�CRi rate. The<br />

7�3 control arm had only a single CRi among 21 responders. The CPX-351<br />

arm had 15 CRi (27%) and 41 CR (73%) allowing a CR v CRi comparison to<br />

be made. Results: CR and CRi pts were balanced by age, race, and PS. The<br />

CRi group had more males (87% v 51%), more baseline WBC�20K (27% v<br />

15%), and more adverse karyotype (40% v 27%) and sAML (47% v 29%).<br />

A smaller proportion <strong>of</strong> CRi pts received post-remission chemotherapy<br />

(47% v 73%) but had similar rates <strong>of</strong> SCT (13% v 20%). Most CRi pts had<br />

delayed platelet recovery (80%). By 1-year more CRi pts had relapsed<br />

(54% v 39%) and more had died (54% v 34%). Contributing causes<br />

included: relapsed AML (7 CRi v 10 CR pts), complications post SCT (1 CRi<br />

v 1 CR pt), chemotherapy complications (0 CRi v2CRpts) and unknown<br />

causes (0 CRiv1CRpt). The survival curves were not significantly different<br />

(p�0.39). Conclusions: More CRi patients had adverse karyotype and sAML<br />

and most (53%) received no post remission chemotherapy. Survival was<br />

not significantly different compared to CR patients but was markedly better<br />

than that <strong>of</strong> non-responders. These data suggest that CRi following<br />

CPX-351 provides clinically meaningful benefit, a finding that needs to be<br />

confirmed in a larger randomized study.<br />

6603 General Poster Session (Board #22F), Mon, 1:15 PM-5:15 PM<br />

Factors associated with outcome <strong>of</strong> secondary MDS and AML: Review <strong>of</strong><br />

2,182 patients at MDACC. Presenting Author: Francesco Paolo Tambaro,<br />

University <strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: Secondary acute myeloid leukemia (sAML) and myelodysplastic<br />

syndromes (sMDS) have been associated with prior treatment for other<br />

malignancies, particularly chemotherapy, and associated with high-risk<br />

features and poor clinical outcomes. Methods: We identified 2,182 patients<br />

(pts) (57% male) diagnosed with AML (n�1,073; all FAB/WHO represented)<br />

or MDS (n�1,109; 984 MDS, 125 MDS/MPD, all WHO represented)<br />

and who had 1 (n�1,907), 2 (n�239), or 3 (n�36) prior<br />

malignancies (PM). Pt characteristics and outcomes were analyzed. Results:<br />

The median age was 67 yrs. Prior hematological neoplasms (25%), breast<br />

(16%), prostate (16%), skin (9%) were most common. The prior malignancy<br />

reflected prior treatment, 1,252 pts underwent surgery, 939<br />

received radiation, and 1,211 received chemotherapy alone or in combination;<br />

124 were untreated. Types <strong>of</strong> prior malignancy and prior treatment<br />

associated with AML and MDS will be presented. Median time to AML/MDS<br />

was 64 months (mo) and 46 mo (p�0.000) for pts with 1 or �1 PM,<br />

respectively, and correlated with type <strong>of</strong> PM (p�0.000) and previous<br />

monotherapy (surgery vs RT vs chemo) (p�0.020). For both AML and MDS,<br />

the incidence <strong>of</strong> poor-risk cytogenetics was nearly twice for pts who<br />

received prior chemotherapy compared to those who underwent surgery or<br />

radiation alone. Factors associated with survival in secondary AML and<br />

MDS are reported in the table. Conclusions: In conclusion, outcomes for pts<br />

with secondary AML and MDS are poor, especially for pts treated with prior<br />

chemotherapy. Type <strong>of</strong> prior malignancy and treatment are correlated with<br />

outcomes. Furthermore, greater number <strong>of</strong> prior malignancies correlates<br />

with inferior outcomes.<br />

AML MDS<br />

Variable<br />

Survival (mo) p Survival (mo) p<br />

#PM 1vs�1 8.6 vs 5 0.004 13.5 vs 10.3 .012<br />

Type prior malignancy Kidney vs lung 12 vs 4 0.00 19.2 vs 9.8 .000<br />

Prior treatment Surgery vs RT vs Chemo 11.7 vs 5 vs 3.5 0.000 19 vs 18 vs 12 .003<br />

Prior treatment Chemo vs No Chemo 10.1 vs 6.7 0.002 18.3 vs 11 .000<br />

Karyotype Bad vs Int vs Good 4.7 vs 12 vs 58.6 0.000 8.9 vs 21 vs 26<br />

Prior hematologic<br />

malignancy<br />

No vs Yes 8.6 vs 6.8 0.08 13.6 vs 11.1 .03<br />

FAB M3 vs M1 61 vs 4 0.000 NA<br />

IPSS High vs Int-1 vs Int-2 vs Low NA NA 7.6 vs 18 vs 9.7 vs 34.7 .000<br />

# cytopenias 0 vs 1 vs 2 vs 3 NA NA 15 vs 16.7 vs 9.6 vs 6.2 .000<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


442s Leukemia, Myelodysplasia, and Transplantation<br />

6604 General Poster Session (Board #22G), Mon, 1:15 PM-5:15 PM<br />

Pooled safety analysis <strong>of</strong> omacetaxine mepesuccinate in patients with<br />

chronic myeloid leukemia (CML) resistant to tyrosine-kinase inhibitors<br />

(TKIs). Presenting Author: Meir Wetzler, Roswell Park Cancer Institute,<br />

Buffalo, NY<br />

Background: Subcutaneous omacetaxine mepesuccinate (“omacetaxine”)<br />

is a reversible, transient inhibitor <strong>of</strong> protein elongation that does not<br />

depend on BCR-ABL signaling. It showed significant clinical activity in two<br />

phase 2, open-label, multicenter studies <strong>of</strong> patients with CML who were<br />

resistant/intolerant to prior TKI therapy. However, omacetaxine tolerability<br />

data across the 3 phases <strong>of</strong> CML are limited. Methods: Safety data were<br />

pooled from all patients in the 2 studies plus a small pilot study.<br />

Omacetaxine 1.25 mg/m2 was given subcutaneously twice daily: �14<br />

successive days per 28-day cycle for induction, �7 days/cycle as maintenance.<br />

Days <strong>of</strong> dosing could be adjusted and recombinant growth factors<br />

given, as clinically indicated. Results: Of 207 pooled patients (median age,<br />

57 years), 108 were in chronic (CP), 55 in accelerated (AP), and 44 in blast<br />

phase (BP). Median treatment was 4 cycles (range, 1–41). On study, 80%<br />

<strong>of</strong> patients had �1 hematologic adverse event (AE), 60% had a serious AE<br />

(SAE), 30% had a hematologic SAE, 34% had an SAE that resulted in<br />

hospitalization, and 15% discontinued treatment due to an SAE (most<br />

commonly disease progression). There were 49 (24%) deaths (15 CP, 11<br />

AP, and 23 BP). Of the 8 that were treatment related (5 CP, 2 AP, 1 BP),<br />

the most common cause was sepsis (3 patients). The most common AEs<br />

were thrombocytopenia (60%), anemia (51%), diarrhea (40%), neutropenia<br />

(37%), and nausea (30%). Injection site reactions (eg, injection site<br />

pain, erythema) occurred in 32%, with the majority being low grade.<br />

Infections (eg, pneumonia, bronchitis) occurred in 51%. Treatment-related<br />

grade 3/4 hematologic AEs included thrombocytopenia (52%), neutropenia<br />

(31%), anemia (30%), leukopenia (12%), and febrile neutropenia<br />

(11%). The most common treatment-related grade 3/4 nonhematologic AE<br />

was fatigue (4.3%). Overall, there were 34 (16%) grade 3/4 infections (12<br />

CP, 11 AP, 11 BP). Conclusions: In the largest safety analysis to date,<br />

subcutaneous omacetaxine 1.25 mg/m2 showed an acceptable safety<br />

pr<strong>of</strong>ile in all phases <strong>of</strong> CML. AEs were primarily hematologic, and grade 3/4<br />

nonhematologic AEs were not common. Support: Teva Pharmaceutical<br />

Industries Ltd.<br />

6606 General Poster Session (Board #23A), Mon, 1:15 PM-5:15 PM<br />

Prospective evaluation <strong>of</strong> serial 2-hydroxyglutarate in acute myeloid leukemia<br />

(AML) to determine response to therapy and predict relapse. Presenting<br />

Author: Amir Tahmasb Fathi, Massachusetts General Hospital/Harvard<br />

Medical School, Boston, MA<br />

Background: Mutations in isocitrate dehydrogenase (IDH1 and IDH2) occur<br />

in 10-20% <strong>of</strong> AML patients and result in production <strong>of</strong> 2-hydroxyglutarate<br />

(2-HG). We hypothesize that serial 2-HG quantification may be used to<br />

monitor response and predict relapse in patients with AML. Methods: We<br />

are conducting a prospective study at MGH and DFCI to (1) assess changes<br />

in serum and urine 2-HG levels during treatment in patients with newly<br />

diagnosed AML, (2) compare 2-HG levels in serum, urine, and bone marrow<br />

and, (3) serially compare 2-HG levels with IDH1/2mutant allele burden. In<br />

those with elevated serum 2-HG (�1000ng/ml), 2-HG is monitored serially<br />

or at relapse. Results: To date, 20 patients have been enrolled, 5 (25%) <strong>of</strong><br />

whom had elevated baseline serum 2-HG. All 5 were found to have IDH1/2<br />

mutations. The serum 2-HG for these patients was significantly higher than<br />

for those who were IDH-wt (median 1933 vs 87ng/mL; p�0.001). Urine<br />

2-HG was also higher in IDH-mutant patients (median 30500 vs 4230ng/<br />

mL; p�0.021), as was bone marrow 2-HG (median 9870 vs 309ng/mL;<br />

p�0.005). Serum 2-HG levels strongly correlated with that in urine (R2 0.987). Serum 2-HG decreased in all IDH mutant patients on therapy, but<br />

more rapidly in those receiving induction chemotherapy (Table), all <strong>of</strong><br />

whom achieved remission, than those receiving hypomethylatingagents.<br />

Conclusions: Patients with IDH-mutant AML have markedly elevated serum<br />

and urine 2-HG. 2-HG levels decreased with therapy and there is a strong<br />

correlation between serum and urine 2-HG. Data on the sensitivity <strong>of</strong> serial<br />

2-HG monitoring as well as comparison with mutant IDH1/2allele burden<br />

will be presented. This data suggests that serial 2-HG quantification may be<br />

a valuable non-invasive biomarker in this genetically defined subset <strong>of</strong><br />

AML.<br />

Serial analysis <strong>of</strong> serum 2HG (ng/mL) during therapy in IDH-mutant AML.<br />

Patient 1 (5-azacitidine)<br />

(IDH2 R140Q)<br />

Patient 2 (induction)<br />

(IDH1 R132H)<br />

Patient 3 (induction)<br />

(IDH1 R132H)<br />

Patient 4 (induction)<br />

(IDH2 R140Q)<br />

Patient 5 (5-azacitidine)<br />

(IDH2 R140Q)<br />

Day 0 Day 7 Day 14 Day 30 Day 60 Day 110<br />

1,933 2,125 2,006 1,599 2,077 60<br />

2,070 1,019 295 363 195<br />

66,207 1,477 1,043 407 154<br />

1,054 249 60 84 106<br />

1,296 786 935 503<br />

6605 General Poster Session (Board #22H), Mon, 1:15 PM-5:15 PM<br />

Analysis <strong>of</strong> imatinib (IM)-related, low-grade (LG), non-hematologic (heme)<br />

adverse events (AEs) in patients (pts) with chronic myeloid leukemia (CML)<br />

switched to nilotinib (NIL): ENRICH study update. Presenting Author:<br />

Michael J. Mauro, Knight Cancer Institute at Oregon Health & Science<br />

University, Portland, OR<br />

Background: This study update assesses change in chronic LG non-heme<br />

AEs in adult Ph� CML-CP pts switched from IM to NIL. Methods: Pts were<br />

eligible if treated with IM 400 mg/d for �3 mo, and had IM-related grade<br />

(G) 1/2 non-heme AEs persisting �2 mo or recurring �3 times and<br />

persisting despite best supportive care. Pts received NIL 300 mg BID.<br />

Primary endpoint is change in IM-related LG non-heme AEs at 3 mo.<br />

Disease response was monitored by RQ-PCR and pt-reported outcomes<br />

measured by 2 quality-<strong>of</strong>-life (QoL) questions and MD Anderson Symptom<br />

Inventory (MDASI)-CML. Results: 47 pts were enrolled as <strong>of</strong> data cut-<strong>of</strong>f<br />

(11/14/2011). There were 168 baseline IM-related non-heme AEs: 121<br />

G1, 47 G2. 37 pts completed 3 mo NIL, by 3 mo 103 <strong>of</strong> 154 IM-related<br />

AEs (67%) improved (92 resolved, 11 improved from G2 to G1), 47<br />

unchanged, 4 increased in severity. 13 pts were dose reduced for<br />

NIL-related AEs; 8 pts re-escalated after AEs recovered to G1 or resolved.<br />

34 G3 AEs occurred in 15 pts; investigators reported 19 AEs as suspected<br />

NIL-related (increased bilirubin, lipase, or blood glucose; hypokalemia;<br />

hypophosphatemia; pruritus; bronchitis; dehydration; rash; arthralgia;<br />

pleural effusion). 8 pts discontinued NIL: 6 for AEs, 2 withdrew consent.<br />

No G4 AEs were reported. 32 pts had MMR (3-log reduction <strong>of</strong> Bcr-Abl;<br />

�0.1% IS) at entry; 16 additional pts achieved MMR on NIL. At entry, 18<br />

pts had 4-log reduction and 10 pts 4.5-log reduction in Bcr-Abl. 16 and 13<br />

additional pts achieved 4- and 4.5-log reduction, respectively, on NIL. At 3<br />

mo (n�34) 62% and 53% pts reported QoL improvement from baseline in<br />

the previous 24 h and 7 d, respectively. Reduction in MDASI-CML<br />

severity/interference scores indicates symptom improvement. Mean reductions<br />

in MDASI-CML from baseline at 3 mo: severity, 1.21 (n�34);<br />

interference, 1.55 (n�33). Conclusions: 3 mo after switching to NIL,<br />

~70% baseline chronic LG non-heme IM-related AEs improved and �53%<br />

<strong>of</strong> pts reported improvement in QoL. Molecular responses were maintained<br />

or improved in all patients. Switch from IM to NIL in majority <strong>of</strong> pts reduces<br />

IM-related toxicities and preserves/induces molecular response in CML-CP.<br />

6607 General Poster Session (Board #23B), Mon, 1:15 PM-5:15 PM<br />

Interim results <strong>of</strong> a phase I/II randomized study <strong>of</strong> cl<strong>of</strong>arabine, idarubicin,<br />

and cytarabine (CIA) versus fludarabine, idarubicin, and cytarabine (FIA)<br />

for newly diagnosed or relapsed patients (pts) with acute myeloid leukemia<br />

(AML). Presenting Author: Michael Mathisen, University <strong>of</strong> Texas M. D.<br />

Anderson Cancer Center, Houston, TX<br />

Background: Outcomes <strong>of</strong> pts with AML remain suboptimal. The addition <strong>of</strong><br />

cladribine to 3�7 has been shown to improve complete remission (CR)<br />

rates and 3-year survival. The aim <strong>of</strong> this study was to assess the efficacy<br />

and safety <strong>of</strong> idarubicin � cytarabine (IA, idarubicin 10 mg/m2 on days<br />

1-3, cytarabine 1 g/m2 on days 1-5) combined with two other nucleoside<br />

analogs, cl<strong>of</strong>arabine (C) or fludarabine (F), in pts with newly diagnosed and<br />

relapsed/refractory (RR) AML. Methods: Pts with newly diagnosed or RR<br />

non-M3 AML with normal organ function were eligible. Pts with RR disease<br />

were treated in the phase I portion defining the MTD <strong>of</strong> C. The starting dose<br />

<strong>of</strong> C was 15 mg/m2 with doses escalating to 25 mg/m2 on days 1-5 in<br />

subsequent cohorts. During phase II, patients were randomized in a<br />

Bayesian design to C at the MTD with IA or fludarabine (F) at 30 mg/m2 on<br />

days 1-5 with IA. Up to 6 consolidation cycles were planned according to an<br />

attenuated schedule using the same drugs. Dose adjustments were made<br />

for elderly pts or pts with poor PS. Results: 9 pts were enrolled in the phase I<br />

portion. The overall response rate (ORR) in this group was 44%. DLT were<br />

observed at C 20 mg/m2 and included hand/foot syndrome (HFS), elevated<br />

bilirubin, and prolonged myelosuppression. The MTD was 15 mg/m2 on<br />

days 1-5. 50 evaluable pts were enrolled (16 newly diagnosed, 34 RR) in<br />

the phase II. In the frontline cohort, median age was similar in both groups<br />

(C 56, F 55), as were the cytogenetic pr<strong>of</strong>iles. The CR rate was 100% in<br />

both groups (9 CIA, 7 FIA). Detailed efficacy information can be found in<br />

the Table. More than half <strong>of</strong> the RR cohort were receiving therapy as salvage<br />

2 or higher, and most had short first CR durations. Notable toxicities<br />

included elevated liver function tests for both groups, and HFS in the C<br />

group. There were 4 deaths on study, though most pts were receiving<br />

therapy as second salvage and were over the age <strong>of</strong> 60. Conclusions: CIA<br />

and FIA are effective regimens for newly diagnosed or RR AML with<br />

manageable toxicity pr<strong>of</strong>iles. The ORR were 100% and 32% for newly<br />

diagnosed and RR AML pts, respectively, with low early mortality rates. The<br />

study is ongoing.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


6608 General Poster Session (Board #23C), Mon, 1:15 PM-5:15 PM<br />

Impact <strong>of</strong> immunoglobulin heavy chain variable region mutational status on<br />

the outcome <strong>of</strong> patients with chronic lymphocytic leukemia harboring<br />

isolated 13q deletion. Presenting Author: Gelenis C. Domingo, H. Lee<br />

M<strong>of</strong>fitt Cancer Canter & Research Institute, Tampa, FL<br />

Background: Several prognostic factors can predict the course <strong>of</strong> chronic<br />

lymphocytic leukemia (CLL). Among them, the IGVH mutational status and<br />

the presence <strong>of</strong> cytogenetic abnormalities are the strongest predictors <strong>of</strong><br />

outcome. Mutated IGVH and deletion 13q independently confer a survival<br />

advantage. CLL patients with mutated IGVH in combination with deletion<br />

13q have a better prognosis when compared to their unmutated IGVH<br />

counterparts. However, there is limited data on the outcome <strong>of</strong> patients<br />

harboring favorable deletion 13q and the unfavorable unmutated IGVH.<br />

This study aimed at identifying patients with these two indicators in order<br />

to obtain important prognostic information. Methods: We used the M<strong>of</strong>fitt<br />

Cancer Center Total Cancer Care (TCC) database to find patients with a<br />

diagnosis <strong>of</strong> CLL between January 1993 and December 2009. Individual<br />

charts were reviewed for demographic data and CLL cytogenetics, including<br />

IGVH mutation status and presence <strong>of</strong> deletion 13q. We analyzed the<br />

impact <strong>of</strong> having deletion 13q in combination with an unmutated IGVH on<br />

the overall survival (OS) for this subset <strong>of</strong> CLL patients using Kaplan Meier<br />

curves with SPSS statistical s<strong>of</strong>tware. Results: 546 patients were identified<br />

during the aforementioned time period with a diagnosis <strong>of</strong> CLL. Median age<br />

was 62.5 years. 144 (26.4%) <strong>of</strong> these patients had IGVH and cytogenetic<br />

analysis available. 53 patients had 13q deletion as their sole genetic<br />

abnormality. Patients with unmutated IGVH and positive for deletion 13q<br />

were 19/53 (35.8%). Patients with mutated IGVH and positive for deletion<br />

13q were 34/53 (64.2%). Patients with mutated IGVH and positive for<br />

deletion 13q had an OS <strong>of</strong> 17 years. While patients with unmutated IGVH<br />

and positive deletion 13q had a lower median OS <strong>of</strong> 12 years (91.2% vs<br />

78.9%, p�0.05). Hazard ratio for patients with IGVH mutated and positive<br />

deletion 13q was 0.4, p�0.05. Conclusions: Mutated IGVH appears to be<br />

associated with improved OS in patients with isolated 13q deletion when<br />

compared to patients with unmutated IGVH and isolated 13q deletion.<br />

Further research is needed to assess these mutations in relation to other<br />

cytogenetic abnormalities in CLL.<br />

6610 General Poster Session (Board #23E), Mon, 1:15 PM-5:15 PM<br />

Assessment <strong>of</strong> age as its own risk factor in AML. Presenting Author: Thomas<br />

Buchner, University <strong>of</strong> Muenster, Muenster, Germany<br />

Background: Patients’ age is an important issue in treatment decisions for<br />

AML, while its role in this disease remains poorly explained. Methods: In the<br />

AMLCG 1999 trial 1223 patients (pts) were 16-59y and 1470 pts were<br />

60-85y <strong>of</strong> age. Their treatment was randomized between TAD-HAM vs<br />

HAM-HAM induction (TAD, standard dose thioguanine, cytarabine, daunorubicin<br />

60mg/m² x 3; HAM, high-dose cytarabine 3g/m² x 6, mitoxantrone<br />

10mg/m² x 3), TAD consolidation and monthly maintenance vs autologous<br />

SCT, any chemotherapy � vs - G-CSF priming. All randomization was done<br />

upfront. Pts <strong>of</strong> �60y received routine double induction and full dose HAM<br />

while pts <strong>of</strong> 60�y preferentially received only one course induction and<br />

HAM at 1g instead <strong>of</strong> 3g cytarabine /m² x6.Results: With little differences<br />

according randomizations, pts �60y and 60�y achieved a complete<br />

remission rate (CR) <strong>of</strong> 70.2% and 53.5% (p�.001), overall survival (OS) at<br />

5y <strong>of</strong> 41.3% and 12.9% (p�.001) and a relapse rate (RR) <strong>of</strong> 49.0 and<br />

72.0% (p�.001). We also focussed on pts around 60y <strong>of</strong> age and<br />

compared the 172 pts <strong>of</strong> 57-59y with the 261 pts <strong>of</strong> 60-62y excluding pts<br />

undergoing allogeneic stem cell transplantation. According to their similar<br />

age the two groups showed similar baseline characteristics. In contrast and<br />

due to the cut-<strong>of</strong>f point for age adaption at 60y they differed considerably in<br />

treatment. Expressed by the cumulative dosage <strong>of</strong> cytarabine, the difference<br />

between the two groups was by factor 2.9. This difference, however,<br />

did not translate into a different outcome being 62% vs 60% CR, 28% vs<br />

21% 5y OS (p�0.25), and 73% vs 73% RR at 5y. A multivariable analysis<br />

in all pts between 16 and 85y <strong>of</strong> age identified cytogenetik/ molecular risk<br />

and age as a continuous variable, to be risk factors predicting CR, OS, as<br />

well as RR. In pts <strong>of</strong> 16-60y those below and above the median age <strong>of</strong> 47y<br />

differed in their CR rate by 75% vs 66% (p�.001), their OS by 49% vs<br />

35% (p�.001) and in their RR by 45% vs 53% (p�.007). In pts <strong>of</strong> 60-85y<br />

those below and above the median age <strong>of</strong> 67y differed in their CR rate by<br />

57% vs 51% (p�.023), and their OS by 16% vs 11% (p�.001), while their<br />

RR was similarly 71%. Conclusions: The outcome in pts with AML is<br />

substantially determined by patients’ age as its own risk factor, and not by<br />

treatment intensity.<br />

Leukemia, Myelodysplasia, and Transplantation<br />

443s<br />

6609 General Poster Session (Board #23D), Mon, 1:15 PM-5:15 PM<br />

A new paradigm in CLL: Minimizing toxicity by using the minimum effective<br />

dose (MED) <strong>of</strong> lenalidomide for older patients with CLL. Presenting Author:<br />

Nicole Lamanna, Memorial Sloan-Kettering Cancer Center, New York, NY<br />

Background: While CIT has proven active for younger patients with CLL;<br />

studies from Germany and MDACC with fludarabine or fludarabine- based<br />

regimens did not benefit the patient over the age <strong>of</strong> 65 or 70. As the median<br />

age <strong>of</strong> diagnosis is 72, the development <strong>of</strong> safe, active therapies for older<br />

patients is an unmet need in CLL. Lenalidomide is an active drug in CLL.<br />

The current clinical trial was designed to assess whether low-dose continuous<br />

lenalidomide therapy can provide long-term disease control with<br />

minimum toxicity in patients older than 65. Methods: Patients initiate<br />

lenalidomide at 2.5mg daily (28-day cycle) and only dose escalate for<br />

progressive disease (POD). Results: 18 patients have been enrolled: 12 men<br />

and 6 women, med age 70 (range 65-84) with Rai-intermediate (7 pts) or<br />

high-risk (11 pts; 61%) disease; med WBC 50.5 (6.7-326.3), HGB 10.3<br />

(8.9-13.9), PLT 121 (44-215), �-2 microglobulin 4.9 (3.0-10.2). 16<br />

(89%) patients had chromosomal abnormalities: 3 with 13q, 6 with tris 12,<br />

3 with 11q, and 4 with 17p. 13 (72%) patients have unmutated IGHV. 5<br />

(28%) patients were previously untreated; 13 (72%) had prior therapy<br />

(range 1-5; med 2). Median dose <strong>of</strong> lenalidomide is 2.5mg (range<br />

2.5mg-10mg); median no. cycles is 7 (range 1-28). 11 patients (61%) are<br />

still on therapy (7 previously treated): 6 at 2.5mg; 4 at 5mg, and 1 at<br />

10mg. 7 have discontinued therapy: 1 for ITP; 3 for POD including one with<br />

Richter’s, 2 for desquamating rash; and 1 withdrew consent. Only 2<br />

patients had tumor lysis and 10 had tumor flare (8 given steroids briefly). In<br />

these older patients on long-term continuous treatment, therapy has been<br />

generally well tolerated. There have been no deaths on study. Pneumonia<br />

developed in 4 patients (3 <strong>of</strong> these had prior therapy). 1 patient with history<br />

<strong>of</strong> atrial fibrillation developed DVT/PE. Main hematologic toxicity is<br />

neutropenia seen in 11 patients but transient. Grade 3/4 thrombocytopenia<br />

and anemia was seen in 5 and 0 patients, respectively. Conclusions:<br />

Low-dose continuous lenalidomide therapy iappears to be well-tolerated<br />

and may <strong>of</strong>fer long term disease control in some patients with CLL.<br />

Additional accrual and longer follow-up will be required to further assess<br />

the utility <strong>of</strong> this strategy.<br />

6611 General Poster Session (Board #23F), Mon, 1:15 PM-5:15 PM<br />

Disease response and survival outcomes in acute myeloid leukemia (AML)<br />

patients unfit for chemotherapy treated with 10-day decitabine as initial<br />

therapy. Presenting Author: Theodore Stewart Gourdin, University <strong>of</strong><br />

Maryland Marlene and Stewart Greenebaum Cancer Center, Baltimore, MD<br />

Background: AML is primarily a disease <strong>of</strong> older adults, with a median age <strong>of</strong><br />

67 years at diagnosis. Many patients are considered unfit for chemotherapy<br />

due to poor performance status and/or comorbidities, prompting investigation<br />

<strong>of</strong> less intensive approaches. A clinical trial in which 10-day courses <strong>of</strong><br />

decitabine 20mg/m2 were given every 28 days as initial therapy demonstrated<br />

a promising complete remission (CR) rate and disease-free survival<br />

(DFS). We used this regimen to treat AML patients unfit for chemotherapy<br />

outside <strong>of</strong> a clinical trial. Methods: Charts <strong>of</strong> 32 newly diagnosed AML<br />

patients treated with 10-day courses <strong>of</strong> decitabine 20mg/m2 as initial<br />

therapy at the University <strong>of</strong> Maryland Greenebaum Cancer Center between<br />

September 2010 and November 2011 were retrospectively reviewed. If<br />

patients attained bone marrow blasts �5%, 5-day courses were administered<br />

subsequently and were continued until disease progression. Results:<br />

Median age at diagnosis was 74 years (range, 52-86). 18 patients were<br />

male and 14 female. 23 were Caucasian, 7 African-<strong>American</strong> and 1<br />

Hispanic. 10 patients (31%) had performance status � 2. All had<br />

significant cardiac, pulmonary and/or renal disease. Karyotype risk group<br />

was unfavorable in 17 patients, intermediate in 14 and unknown in one.<br />

There were 9 CRs and 1 partial remission, for an overall response rate <strong>of</strong><br />

31%, following a median <strong>of</strong> 2 (range, 1 to 4) courses. 21 patients did not<br />

respond to treatment following 1 to 6 courses, and one died within 29 days<br />

<strong>of</strong> treatment initiation. 21 patients (65%) were hospitalized at least once<br />

for fever or infection during treatment. Of the 9 patients who achieved CR,<br />

5 had normal, 3 complex and 1 unknown karyotypes. Median DFS was 390<br />

days (range, 87�-468�) and median OS 180 days (range, 15-623�).<br />

Six-month OS was 37.5% and 1-year OS 16.5%. Conclusions: AML<br />

patients unfit for chemotherapy treated with 10-day decitabine 20mg/m2<br />

outside <strong>of</strong> a clinical trial had a higher response rate than reported for 5-day<br />

decitabine, but a lower response rate than in the reported clinical trial <strong>of</strong><br />

the 10-day regimen. Additional novel treatment approaches are needed for<br />

these patients.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


444s Leukemia, Myelodysplasia, and Transplantation<br />

6612 General Poster Session (Board #23G), Mon, 1:15 PM-5:15 PM<br />

High frequency <strong>of</strong> BCR-ABL oncogene in pediatric acute lymphoblastic<br />

leukemia (ALL) patients as revealed by RT-PCR and interphase FISH:<br />

Association with disease biology and treatment outcome. Presenting<br />

Author: Zafar Iqbal, Molecular Genetic Pathology Unit, Department <strong>of</strong><br />

Pathology, College <strong>of</strong> Medicine and KKUH, King Saud University, Riyadh,<br />

Saudi Arabia; and Haematology, Oncology and Pharmacogenetic Engineering<br />

Sciences (HOPES) Group, HSRL, Zoology Department, University <strong>of</strong> the<br />

Punjab, Lahore, Pakistan<br />

Background: ALL is a complex genetic disease involving many fusion<br />

oncogenes (FGs) 1 frequency <strong>of</strong> which can vary in different ethnic groups2,3 thus having implication in differential diagnosis, prognosis and treatment.<br />

Methods: We studied FGs in 101 pediatric ALL patients using RT-PCR1 at<br />

Day 0, Day 15 and Day 29 and Interphase FISH, and their association with<br />

clinical features and treatment outcome. Results: Five most common FGs<br />

i.e. BCRABL (t 22; 9), TCF3-PBX1 (t 1; 19), ETV6-RUNX1 (t 12; 21),<br />

MLL-AF4 (t 4; 11) and SIL-TAL1 (del 1p32) were found in 88.1%<br />

(89/101) patients. Frequency <strong>of</strong> BCR-ABL was 44.5% (45/101). Patients<br />

with BCR-ABL had significantly lower survival (43.733 weeks �4.241) as<br />

compared to others except MLL-AF4 and significantly higher TLC count.<br />

Overall survival was lower (52.2�3.75) than the patients with ETV6-RUN X<br />

1 (65.2� 9.9) which may be due to overall high frequency <strong>of</strong> poor<br />

prognostic FGs (71%) as compared to ETV6-RUNX 1 (17.1%) (p�0.01).<br />

Conclusions: This is the first study from Pakistan correlating molecular<br />

markers, disease biology and treatment response. It is the highest reported<br />

frequency <strong>of</strong> BCR-ABL in pediatric ALL and was associated with disease<br />

biology and survival. Some authors have reported BCR-ABL frequency<br />

higher than in West2,4,5 while others reported 45% frequency <strong>of</strong> ETV6-<br />

RUNX16 . These and our data reflect strong interplay <strong>of</strong> genetic and<br />

environmental factors in biology <strong>of</strong> pediatric ALL and its correlation with<br />

disease biology and treatment2,3 . Our data indicates immediate need for<br />

large clinical trials <strong>of</strong> imatinib, dasatinib and nilotinib in paediatric ALL<br />

treatment in our ethnic group. This study will lead to unravel the<br />

mechanisms <strong>of</strong> BCR-ABL Leukemogenesis and to find population-specific<br />

biomarkers and drug targets. References: 1) van Dongen JJ, et al.,<br />

Leukemia. 1999 Dec; 13(12):1901-28. 2) Iqbal Z, et al. J Pediatr<br />

Hematol Oncol. 2007 Aug; 29(8):585. 3) Ariffin H, et al. J Pediatr Hematol<br />

Oncol. 2007 Jan; 29(1):27-31. 4) Ramos C, et al. 2011 Sep; 139(9):1135-<br />

42. 5) Artigas A CG, et al. Rev Med Chil. 2006 Nov; 134(11):1367-76. 6)<br />

Karrman K, et al. Br J Haematol. 2006 Nov; 135(3):352-4.<br />

6614 General Poster Session (Board #24A), Mon, 1:15 PM-5:15 PM<br />

CDKN2A-deletion to predict relapse in adult B-cell acute lymphoblastic<br />

leukemia (B-ALL). Presenting Author: Preet Paul Singh, Mayo Clinic,<br />

Rochester, MN<br />

Background: CDKN2A locus on chromosome 9p21 is implicated in altered<br />

molecular pathways leading to leukemogenesis as a tumor suppressor by<br />

inhibiting the proliferative kinases CDK4/CDK6 and blocking the cell-cycle<br />

division during G1/S phase. Deletion <strong>of</strong> CDKN2A locus is frequently seen in<br />

ALL, although prognostic significance remains unclear. Methods: Retrospective<br />

chart review at Mayo Clinic between 8/2005-11/2011 was performed<br />

on adult B-ALL patients (pts) who underwent fluorescent in situ hybridization<br />

(FISH) studies at diagnosis. Survival estimate was calculated using<br />

Kaplan-Meier statistics. Event-free survival (EFS) was defined as time<br />

between diagnosis and induction failure, relapse, or death, while overall<br />

survival (OS) as time between diagnosis and death Results: Out <strong>of</strong> 27 pts<br />

who had FISH studies for CDKN2A, 15 (56%) pts had CDKN2A deletions<br />

(8 homozygous/7 hemizygous), while 12 (44%) pts had diploid cytogenetics<br />

(none had abnormalities <strong>of</strong> MLL, bcr/abl fusion or CDKN2A deletion on<br />

FISH). There was no significant difference in median age, gender, WBC,<br />

hemoglobin, platelet count, LDH or complete remission (CR) rate (p�0.924)<br />

between the two groups. CDKN2A-deleted pts had higher circulating blasts<br />

(34% vs 16%, p�0.017). Of 13 CDKN2A-deleted pts, chromosome 9p<br />

was intact in 9 (69%) pts using routine cytogenetics. In the CDKN2Adeleted<br />

group, allogeneic stem cell transplant (ASCT) was performed in 7<br />

(47%) pts at first CR and 1 at second CR. Four <strong>of</strong> 15 (27%) CDKN2Adeleted<br />

pts also had bcr/abl fusion. Five-year EFS was poorer in CDKN2Adeleted<br />

pts compared to normal FISH group (22% vs 60%, p�0.042),<br />

while 5-year OS was 72% vs 60% (p�0.308), respectively. Excluding the 4<br />

bcr/abl fusion pts, 5-year EFS was still statistically significant (p�0.008).<br />

In the CDKN2A deleted group, both 5-yr EFS (80% vs 0%, p�0.006) and<br />

OS (100% vs 42%, p�0.016) were significantly superior in pts who<br />

received ASCT. Conclusions: CDKN2A deletions predict earlier relapse in<br />

pts with B-ALL. Majority <strong>of</strong> 9p abnormalities were only found by FISH<br />

testing and missed by conventional cytogenetics. In CDKN2A deleted pts,<br />

ASCT gave overall survival advantage. Further investigation through larger<br />

cohorts <strong>of</strong> pts is needed to validate these findings.<br />

6613 General Poster Session (Board #23H), Mon, 1:15 PM-5:15 PM<br />

Safety and efficacy <strong>of</strong> dose escalated liposomal amphotericin B in adult<br />

leukemia patients with refractory invasive fungal infections: A single<br />

institution report. Presenting Author: Sherry Mathew, Memorial Sloan-<br />

Kettering Cancer Center, New York, NY<br />

Background: Leukemia patients are at risk for invasive fungal infections(IFI).<br />

Empiric therapy <strong>of</strong> suspected IFI is begun due to fever despite<br />

appropriate antibiotics during neutropenia or infiltrates on chest CT.<br />

Liposomal amphotericin B (L-AmB) is used for empiric anti-fungal therapy<br />

at 3-5 mg/kg. Data is lacking on the efficacy <strong>of</strong> L-AmB dose escalation in<br />

patients with CT progression and clinical decline. Methods: Patients who<br />

received 10 mg/kg <strong>of</strong> L-AmB from 2002-11. They were required to be �18<br />

years old and escalated from 5 to 10 mg/kg L-Amb for at least 7 doses.<br />

Patient information was collected from pharmacy and medical records.<br />

Successful treatment was defined as resolution <strong>of</strong> fever at least 1 week after<br />

high dose L-AmB and/or improvement or stability on chest CT. Renal and<br />

hepatic toxicity was assessed. <strong>Clinical</strong> remission was defined as recovery <strong>of</strong><br />

a normal neutrophil count. IRB approval was obtained for this study.<br />

Results: 58 patients were evaluated. At the time <strong>of</strong> presumed or probable<br />

IFI, 19 had completed induction therapy, 5 had received supportive<br />

therapy, and 34 had received 2 or more cycles <strong>of</strong> therapy. High dose L-AmB<br />

was begun for CT findings and fever, CT findings only, and fever only in 14,<br />

26, and 17 patients, respectively. Patients received a median <strong>of</strong> 15 doses<br />

<strong>of</strong> high dose L-AmB, and all received concomitant therapy with an<br />

echinocandin except two: 1 received nothing and 1 received voriconazole.<br />

Treatment success was seen in 28 (48%) patients. Twenty-nine (50%)<br />

patients were alive at 12 weeks and 10 (17%) patients proceded to BMT.<br />

Only 11 (39%) <strong>of</strong> the responding patients achieved a clinical remission. All<br />

patients received IV hydration and electrolyte repletion. Six patients had a<br />

grade 2 increase in serum creatinine and 10 patients developed grade 2 or<br />

3 hepatic toxicity. There were no grade 4 adverse events. Conclusions:<br />

Although it has been reported that L-AmB at 10mg/kg was inferior to 3<br />

mg/kg in untreated patients, our study supports a safe dose escalation<br />

strategy to 10mg/kg in treating progressive presumed or probable IFI and<br />

should be considered a clinical alternative for these high risk patients.<br />

6615 General Poster Session (Board #24B), Mon, 1:15 PM-5:15 PM<br />

Time to ATRA in suspected newly diagnosed acute promyelocytic leukemia<br />

and association with early death rate at a non-cancer center institution: Are<br />

we meeting the target? Presenting Author: Gulam Abbas Manji, Albany<br />

Medical Center, Albany, NY<br />

Background: ATRA administration in suspected APL patients (sAPL) is<br />

thought to impact early death rate (EDR) (Tallman and Manji, Blood Cells<br />

Mol Dis. 2011). Delay in ATRA therapy at specialized centers was<br />

associated with EDR (Altman et al Blood 2011). EDR within this study was<br />

significantly lower compared to the SEER database (12% vs 17.3%) and<br />

hence may not reflect overall delay in ATRA therapy. We determined time to<br />

ATRA therapy in sAPL, and fraction <strong>of</strong> sAPL that did not have disease.<br />

Methods: Retrospective analysis <strong>of</strong> patients that received ATRA for newly<br />

diagnosed s APL between 01/01/98-12/31/11 at Albany Medical Center.<br />

Time to hematologist evaluation, ATRA ordered and administered, and<br />

mortality data was collected from cancer registry, medical record, and<br />

chemo-pharmacy database. Results: A total <strong>of</strong> 39 patients with newly<br />

diagnosed sAPL were administered ATRA (46% male, mean age 50y). APL<br />

diagnosis: 29/39 (75%) true APL (APL); 9/10 ATRA-treated non-APL<br />

(A-nAPL); and one patient for whom cytogenetic data unavailable. EDR<br />

amongst APL was 5/29 (17%) compared to 2/9 (22%) within A-nAPL. Time<br />

variables were compared between APL patients that died early (�30d) to<br />

those that survived 30 days, and included: time to hematologist response<br />

(0.9d vs. 0.4d); time to ATRA ordered (2.9d vs. 2.0d); time to ATRA<br />

administered (3.4d v. 2.2d); and time elapsed between hematologist<br />

response to ATRA administered (2.5d v. 1.9d), respectively. Cryoprecipitate<br />

was administered to 1/5 (20%) patients who expired within 30d<br />

compared to 10/23 (43%) who survived. Overall mortality for APL was 9/29<br />

(31%) compared to 4/9 (44%) for A-nAPL group. Compared to recent<br />

reports, time to ATRA administration in our institution was later (2.0d vs.<br />

2.4d). Conclusions: Our data indicate that APL patients who survived 30d<br />

received ATRA early. EDR at our institution is comparable to that reported<br />

by SEER database and may be attributed to delay in ATRA administration.<br />

Higher fraction <strong>of</strong> patients that survived 30d received cryoprecipitate.<br />

Hence aggressive blood product support may contribute to improved<br />

survival. Timing at which these products were administered is currently<br />

being evaluated.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


6616 General Poster Session (Board #24C), Mon, 1:15 PM-5:15 PM<br />

Epigenetically induced expression <strong>of</strong> CD30 in T-PLL: A rationale for the use <strong>of</strong><br />

brentuximab vedotin. Presenting Author: Zainul Hasanali, Penn State Hershey<br />

Medical Center, Hershey , PA<br />

Background: T-cell prolymphocytic leukemia (T-PLL) is characterized by high<br />

lymphocyte counts, skin involvement and enlarged organs. Treatment with<br />

alemtuzumab (alem) induces complete remission (CR) in �50% <strong>of</strong> patients,<br />

duration is short (median: 6 mos). Therapy with the histone deacetylase<br />

inhibitors (HDACi) vorinostat (SAHA) or romidepsin (romi) and cladribine<br />

(2-CdA), may augment mAb activity. Methods: Five T-PLL patients were studied<br />

(Table). Diagnosis was made by pathology, flow cytometry and TCR studies. Alem<br />

(30 mg sq d1,3,5) and 2-CdA (5mg/m2 d1-5), followed by alem, 2-CdA and<br />

SAHA (400mg qdX5days) or romi (14mg/m2d 1,8,15) was given. One patient<br />

with skin lesions expressed CD30 and received brentuximab vedotin (SGN-35).<br />

Results: All patients achieved CR (mean: 10; median: 8 mos). Relapsed patients<br />

responded with repeat dosing. Genes assayed indicate involvement <strong>of</strong> the MAPK<br />

pathway (p53, DUSP2, TRIB1, C/EBP). mRNA expression <strong>of</strong> the membrane receptor<br />

CD30 (TNFRSF8) increased following therapy in 4/5 patients (mean: 14; Range:<br />

1.3-69 fold). CD30 protein was confirmed by IHC and Western blotting. One patient<br />

with relapsed T-PLL involving skin had a dramatic clearing <strong>of</strong> lesions and blood counts<br />

after one dose <strong>of</strong> SGN35. Conclusions: Therapy with HDACi and 2-CdA overcomes<br />

resistance and augments clinical activity <strong>of</strong> alem and SGN-35, as evidenced by<br />

derepression <strong>of</strong> CD30 and response to treatment. We present a rationale for<br />

epigenetic resensitization to mAbs and their drug conjugates (ADC) in T-PLL. A<br />

prospective multicenter clinical trial is warranted.<br />

Patient characteristics.<br />

Age, years<br />

Range 64-77<br />

Median 71<br />

Sex<br />

Male 3<br />

Female 2<br />

Karn<strong>of</strong>sky score 90% (5)<br />

WBC at diagnosis<br />

Range 120-300<br />

Median 211<br />

Peripheral blood flow<br />

CD4� 5<br />

CD8� 2<br />

CD2� 5<br />

CD5� 5<br />

CD7� 5<br />

Treatment<br />

Alemtuzumab 5<br />

Cladribine 5<br />

Vorinostat 3<br />

Romidepsin 1<br />

Brentuximab vedotin 1<br />

TCR-rearrangement 5<br />

Remission 5/5 (100%)<br />

Remission duration (months)<br />

Pt 1 16, 8�<br />

Pt 2 8, 5, 4<br />

Pt 3 6*<br />

Pt 4 13, 1*<br />

Pt 5 6, 3�<br />

Current status<br />

Alive 2<br />

Hematologic remission 2<br />

CD30 mRNA expression fold change after initial treatment<br />

Pt 1 �69.0<br />

Pt 2 �1.3<br />

Pt 3 -3.5<br />

Pt 4 �11.8<br />

Pt 5 �2.8, �4.2†<br />

Average �11.8<br />

Std. dev. -/� 27.3<br />

* Patient deceased with no evidence <strong>of</strong> disease.<br />

† Fold change after treatment <strong>of</strong> relapse.<br />

6618 General Poster Session (Board #24E), Mon, 1:15 PM-5:15 PM<br />

The prognostic role <strong>of</strong> serum albumin in patients receiving induction<br />

chemotherapy for acute myeloid leukemia (AML). Presenting Author:<br />

Hossein Sadrzadeh, Massachusetts General Hospital/Harvard Medical<br />

School, Boston, MA<br />

Background: Serum albumin has been investigated as a prognostic tool in<br />

the care <strong>of</strong> patients with hematologic malignancies, including multiple<br />

myeloma and myelodysplastic syndromes. However, its prognostic utility in<br />

patients with AML is unknown. We hypothesized that a lower serum<br />

albumin is associated with worse outcomes following induction chemotherapy<br />

for AML. Methods: We conducted a retrospective medical record<br />

review <strong>of</strong> 166 adult, non-promyelocytic AML patients who had received<br />

induction chemotherapy at Massachusetts General Hospital from 1992 to<br />

2007. Patient characteristics were summarized as numbers and percentages<br />

for categorical variables. The Kaplan Meier method was used to<br />

estimate median disease-free survival (DFS) and overall survival (OS). We<br />

dichotomized our patients by serum albumin �3 and �3, and determined<br />

the association <strong>of</strong> albumin with 60-day survival and complete remission<br />

(CR) rate using Fisher’s exact test. Association <strong>of</strong> albumin with DFS and OS<br />

was summarized using Cox regression in both univariate and multivariable<br />

analyses. Results: Of 166 patients, 125 (75%) achieved CR and 143<br />

(86%) were alive at 60 days following diagnosis. After risk-adjusting for age<br />

and LDH, we found that a serum albumin level �3 mg/dL was associated<br />

with decreased 60-day survival (OR 0.30, p�0.015) and CR rate (OR 0.41,<br />

p�0.02) compared to patients with serum albumin �3. There was no<br />

association between serum albumin and DFS (p�0.88) or OS (p�0.31). As<br />

expected, younger age was associated with better induction outcomes.<br />

Conclusions: Serum albumin was negatively associated with short-term<br />

outcomes in patients receiving induction chemotherapy. A serum albumin<br />

level less than 3, clinically relevant to oncologic patients, was associated<br />

with a significantly decreased CR rate and lower 60-day survival after<br />

induction chemotherapy. This data suggests that serum albumin, a<br />

surrogate commonly used for nutritional status and suppressed in inflammatory<br />

comorbid states, has prognostic utility for AML patients undergoing<br />

induction chemotherapy.<br />

Leukemia, Myelodysplasia, and Transplantation<br />

445s<br />

6617 General Poster Session (Board #24D), Mon, 1:15 PM-5:15 PM<br />

A seasonal pattern <strong>of</strong> presentation in younger patients with de novo acute<br />

myeloid leukemia (AML). Presenting Author: Benjamin Jacob Drapkin,<br />

Massachusetts General Hospital/Harvard Medical School, Boston, MA<br />

Background: Seasonal variation in AML has been studied in a wide variety <strong>of</strong><br />

populations and locations, with primarily negative results. These investigations<br />

may have been undermined by the heterogeneity <strong>of</strong> the disease, as de<br />

novo and secondary AML can vary in disease presentation and trajectory,<br />

likely reflecting distinct pathogenesis.We investigated seasonal variation in<br />

the incidence <strong>of</strong> AML diagnosed at Massachusetts General Hospital (MGH)<br />

from 1992 through 2011, focusing on de novo disease among younger<br />

patients. Methods: We assembled a database <strong>of</strong> 511 biopsy-proven cases <strong>of</strong><br />

adult-onset AML (age � 18 years), from the MGH electronic medical<br />

record, using a systematic search algorithm with IRB approval. We<br />

subdivided this database into three cohorts: (1) de novo AML diagnosed<br />

prior to age 50 (2) de novo AML diagnosed after age 50, and (3) secondary<br />

AML, preceded by chemotherapy, myelodysplasia or myeloproliferative<br />

disease. Diagnosis dates were grouped into quarters (Jan-Mar, Apr-Jun,<br />

Jul-Sep, Oct-Dec). Divergence <strong>of</strong> quarterly incidence from a null hypothesis<br />

<strong>of</strong> uniformity was evaluated by chi square analysis. Results: Among patients<br />

under 50-years-old with de novo AML, we found a 44% increase in<br />

incidence between October and December (Table, p�0.04). There was no<br />

significant variation throughout the rest <strong>of</strong> the calendar year. Furthermore,<br />

the incidence <strong>of</strong> both de novo AML diagnosed after age 50 and secondary<br />

AML conformed to a uniform quarterly distribution. As expected, the<br />

majority <strong>of</strong> AML patients under age 50 demonstrated intermediate-risk<br />

cytogenetics, most frequently normal karyotype. Conclusions: We found a<br />

significantly increased incidence <strong>of</strong> de novo AML in younger patients<br />

between the months <strong>of</strong> October and December, diagnosed at MGH from<br />

1992 through 2011. This may reflect a local environmental or infectious<br />

exposure that is not relevant to the pathogenesis <strong>of</strong> AML in older patients or<br />

those in whom AML develops due to prior therapy or previous hematological<br />

disorder. Investigation <strong>of</strong> this exposure is ongoing.<br />

Quarter All cases Under 50 y/o Over 50 y/o Secondary<br />

Jan-Mar 23% 17% 26% 23%<br />

Apr-Jun 25% 23% 25% 27%<br />

Jul-Sep 25% 24% 24% 26%<br />

Oct-Dec 27% 36% 26% 24%<br />

Total patients 511 108 182 221<br />

p value 0.51 0.04 0.97 0.77<br />

6619^ General Poster Session (Board #24F), Mon, 1:15 PM-5:15 PM<br />

Initial characterization <strong>of</strong> the toxicity and efficacy <strong>of</strong> elacytarabine (CP-<br />

4055), a novel antileukemic agent, using a multidimensional exposureresponse<br />

relationship model. Presenting Author: Murray P. Ducharme,<br />

Learn and Confirm Inc., Quebec, QC, Canada<br />

Background: Elacytarabine (CP-4055), an elaidic acid ester <strong>of</strong> ara-C, is a<br />

novel antineoplastic agent developed to treat hematologic malignancies<br />

(HM). It is metabolized to active ara-CTP and inactive ara-U. In 3 studies,<br />

diverse elacytarabine monotherapy doses and regimens were given to<br />

patients with advanced HM and solid tumours. This analysis aimed to 1)<br />

describe the pharmacokinetics (PK) <strong>of</strong> elacytarabine in these patients and<br />

2) investigate the relationship between PK and toxicity and efficacy in HM.<br />

Methods: Population PK analyses were run with ADAPT 5 (ITS) and<br />

included 146 patients given a 2h, 4h or 120h continuous infusion (CIV) <strong>of</strong><br />

elacytarabine. Elacytarabine, ara-C and ara-U plasma concentrations (Cp)<br />

were simultaneously modeled and explained. Standard model discrimination<br />

criteria were used to select the best model. With the model, different<br />

dosing regimens were simulated to find average exposure and % <strong>of</strong> patients<br />

with efficacious or toxic exposure. A multi-dimensional exposure-response<br />

relationship (MDERR) was developed with pre-clinical and clinical data.<br />

Results: The best model was a 2-compartment (CPT) model with linear<br />

elimination and formation rates for the metabolism <strong>of</strong> elacytarabine to<br />

ara-C. Mean PK parameters were Vc � 4.12 L/m2 ,CL�5.27 L/h/m2 and<br />

T1/2 � 7.57 h. Elacytarabine PK included a peripheral CPT with a<br />

non-linear distribution process to reflect saturable binding to red blood<br />

cells, otherwise its PK was linear. Ara-C and ara-U PK were described by 2and<br />

1-CPT linear models, respectively. The MDERR model related efficacy<br />

and toxicity thresholds to elacytarabine dosing regimens, and indicated<br />

that a 120 h CIV dosing regimen is preferable, allowing elacytarabine to<br />

exceed efficacious Cp for longer than the other investigated regimens. A<br />

minimum dose <strong>of</strong> 1000 mg/m2 /day is needed for most patients to receive<br />

efficacious exposure. Conclusions: Elacytarabine is a promising antileukemic<br />

agent for patients with advanced HM. Its PK, toxicity and efficacy<br />

were characterized in patients given diverse dosing regimens. An MDERR<br />

model, which will be further refined or confirmed in upcoming clinical<br />

trials, was proposed.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


446s Leukemia, Myelodysplasia, and Transplantation<br />

6620 General Poster Session (Board #24G), Mon, 1:15 PM-5:15 PM<br />

Azacitidine as induction therapy <strong>of</strong> elderly patients with acute myelogenous<br />

leukemia. Presenting Author: Cyrus Khan, West Penn Allegheny Health<br />

System, Pittsburgh, PA<br />

Background: Effective alternative treatment <strong>of</strong> elderly patients (aged � 60<br />

years) with acute myelogenous leukemia (AML) remains challenging.<br />

Elderly patients with AML respond poorly to standard induction regimens<br />

and have high treatment related mortality. In a prior retrospective analysis<br />

<strong>of</strong> elderly patients with AML performed at our institution, azacitidine (AZA)<br />

showed an overall response rate (ORR) <strong>of</strong> 60% with limited toxicity.<br />

Methods: This is a prospective, phase II open-label study using AZA in<br />

patients �60 years with AML. Inclusion criteria: Newly-diagnosed non-M3<br />

AML and ECOG � 2. Patients with circulating blast count �30,000/mcl<br />

were treated with hydroxyurea until � 30,000/mcl. AZA was administered<br />

at 100 mg/m2 subcutaneously for 5 consecutive days every 28 days until<br />

disease progression or significant toxicity. Results: In all, 15 patients were<br />

enrolled last follow-up being 8/2/11. The mean age <strong>of</strong> patients is 74 years<br />

(range: 64–82). Mean ECOG score was 1. Mean baseline bone marrow<br />

blast count was 52% (range: 25–92%). ORR was 46% (n�7) with<br />

complete response (CR) in 3 (20%) patients and partial response (PR) in 2<br />

(13%) patients according to NCI response criteria, and hematologic<br />

improvement (HI) in 2 (13%) patients according to IWG response criteria.<br />

The mean number <strong>of</strong> days on treatment was 198 (range: 13–724). The<br />

mean time to best response among responders was 95 days (range:<br />

44-279). The mean number <strong>of</strong> days hospitalized for diagnosis plus<br />

treatment or disease-related complication was 19 (range: 5–56), with the<br />

majority <strong>of</strong> therapy administered in an outpatient setting. Mean overall<br />

survival (OS) from diagnosis for all patients was 355 days (range: 13–908)<br />

and mean OS for responders was 532 days (range: 120–908). Nonhematological<br />

toxicity was limited to mild injection site skin reaction and<br />

fatigue in 73% (11/15). No treatment-related deaths were observed. The<br />

dose and schedule <strong>of</strong> AZA remained constant in a majority <strong>of</strong> the patients.<br />

Conclusions: This study suggests that a 5-day schedule <strong>of</strong> SC AZA at 100<br />

mg/m2 to elderly patients with newly-diagnosed AML is a feasible,<br />

well-tolerated and effective alternative to standard induction chemotherapy.<br />

6622 General Poster Session (Board #25A), Mon, 1:15 PM-5:15 PM<br />

Use <strong>of</strong> granulocytes concentrates from a single high yield apheresis to<br />

support more than one patient. Presenting Author: Fleur M. Aung, University<br />

<strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: Allogeneic granulocyte transfusion therapy is sometimes the<br />

only therapeutic option in immunocompromised neutropenic leukemic/<br />

HSCT patients with life threatening bacterial and fungal infections.<br />

Collection <strong>of</strong> larger cell doses <strong>of</strong> granulocyte concentrates (GC) by automated<br />

apheresis following G-CSF/steroids administration has renewed<br />

interest and its use has grown steadily. Methods: Donors were recruited from<br />

family/friends <strong>of</strong> the patients and informed consent was obtained. GCs were<br />

collected via donor stimulation with a single subcutaneous dose <strong>of</strong> G-CSF<br />

(600 mcg). First time donors received an oral dose <strong>of</strong> dexamethasone (8<br />

mg). GCs were harvested by Hetastarch-assisted leukapheresis 12 hours<br />

later via peripheral venous access with the continuous flow cell separator<br />

Spectra (COBE Caridian BCT, Lakewood, CO). Results: GCs were collected<br />

from 93 donors (67M: 26F; age median 40 (19-73 years) and 1½xblood<br />

volume processed median 7048 (3212-8085 mL). The pre/post wbc were<br />

median 8.1 (4.1-22.8)/ 42.7 (22.4-77.5). The volume collected was<br />

based on the post G-CSF wbc count (� 35 K/UL - 800 mL; �35 K/UL<br />

between 600-700 mL). The original yield (10 x10e) was median 9.7<br />

(5.1-17.2 units), volume median 718 (538-860 mL) and WBC median<br />

132.7 (84.2 -241.6 K/UL per bag). The Split GCs were median 4.48<br />

(2.70-7.72 units), with neutrophils median 87% (54-97% per bag). GCs<br />

were transfused to 51 patients (31M: 20F; age median 58 [19-83 years])<br />

who received median 3.5 (1-18 split units), containing wbcs median<br />

10734.69 [5778-42 -162698.09 (x10^3/uL)/unit) and achieved a WBC<br />

increment median 0.2 (0.0-8.3K/UL). Conclusions: The GCs were split due<br />

to the primary care team’s reluctance to transfuse the entire volume <strong>of</strong> the<br />

product due to volume restrictions or for some other medical reason. The<br />

same rationale to split platelets from high yield single apheresis was used<br />

to split our GC products. We found that GCs from each donor was utilized<br />

maximally and also brought partial relief to other patients who lacked<br />

donors. We would like to make the case that when adequate numbers <strong>of</strong><br />

GCs are collected it may be feasible to split the unit. This may <strong>of</strong>fer a<br />

glimmer <strong>of</strong> hope to other patients in need <strong>of</strong> GCs who lack a donor network<br />

support.<br />

6621 General Poster Session (Board #24H), Mon, 1:15 PM-5:15 PM<br />

Phase I dose escalation study <strong>of</strong> bortezomib in combination with lenalidomide<br />

in patients with myelodysplastic syndromes (MDS) and acute myeloid<br />

leukemia (AML). Presenting Author: Philip C. Amrein, Massachusetts<br />

General Hospital/Harvard Medical School, Boston, MA<br />

Background: Both bortezomib (Bz) and lenalidomide have clinical activity in<br />

patients with MDS and AML. We conducted a phase I dose escalation study<br />

to determine the maximal tolerated dose (MTD) <strong>of</strong> Bz in combination with<br />

lenalidomide. Methods: Patients with MDS (IPSS score � 0.5 or therapyrelated)<br />

received Bz by IV bolus on Days 1, 4, 8, and 11 and lenalidomide<br />

10 mg/day PO on Days 1-21 in 28 day cycles for up to 9 cycles. Three doses<br />

<strong>of</strong> Bz were tested (0.7, 1.0, or 1.3 mg/m2 ). Cohorts consisted <strong>of</strong> 3-6<br />

patients; the dose <strong>of</strong> Bz was escalated if there were � 2 dose limiting<br />

toxicities (DLTs). Growth factor support and transfusions were permitted.<br />

Dose limiting toxicities (DLTs) were assessed during the first cycle and were<br />

defined as severe neutropenia (absolute neutrophil count � 250/ul),<br />

thrombocytopenia (platelet count � 10,000/ul), grade � 2 neurotoxicity,<br />

or other grade � 3 non-hematologic toxicity. Following determination <strong>of</strong> the<br />

MTD, enrollment opened to patients with relapsed and refractory AML and<br />

those with untreated high risk disease for whom induction therapy was not<br />

indicated. Responses were assessed by IWG 2006 criteria for MDS and<br />

IWG 2003 criteria for AML. Results: 23 patients (14 men) were enrolled;<br />

one patient was inevaluable due to disease progression prior to starting<br />

protocol therapy. The median age was 73 years (range 54-87). There was 1<br />

DLT observed, neutropenia, in 6 patients treated with 1.0 mg/m2 Bz and no<br />

DLTs at 0.7 or 1.3 mg/m2 . The median number <strong>of</strong> cycles was 2 (range 2-9).<br />

Grade � 3 toxicities possibly attributable to the treatment at any dose level<br />

were: anemia (2), thrombocytopenia (10), leukopenia (3), infection (1),<br />

rash (2), dyspnea (1), dizziness (1), hypotension (1), pneumonia (2) and<br />

neuropathy (1). Among the 14 patients with MDS, 1 patient with RARS<br />

experienced a CR and 2 with RAEB-2 experienced marrow CR (mCR).<br />

Among the 8 patients with AML, there was 1 CR. Conclusions: The maximal<br />

tested dose <strong>of</strong> Bz (1.3 mg/m2 ) in combination with lenalidomide is safe.<br />

Responses were seen in MDS and high risk AML. Future testing <strong>of</strong> this<br />

regimen is planned.<br />

6623 General Poster Session (Board #25B), Mon, 1:15 PM-5:15 PM<br />

Novel risk factors for osteonecrosis <strong>of</strong> the jaw in multiple myeloma: A<br />

RADAR report. Presenting Author: Jaimee S. Holbrook, Department <strong>of</strong><br />

Dermatology, Northwestern University Feinberg School <strong>of</strong> Medicine, Chicago,<br />

IL<br />

Background: Osteonecrosis <strong>of</strong> the jaw (ONJ) was identified in 2001 and is<br />

commonly seen in multiple myeloma (MM). The objective <strong>of</strong> this study is to<br />

evaluate novel risk factors in MM cases from a retrospective database. We<br />

hypothesized that ONJ may be related to stem cell transplant, chronic<br />

kidney disease and active smoking. Methods: We conducted a retrospective<br />

case-control study <strong>of</strong> 231 MM cases (from January 1, 1998 to September<br />

30, 2010) identified from the electronic data warehouse (EDW) at<br />

Northwestern University (NU). The EDW is cross-institutional and integrates<br />

clinical data across NU. It comprises more than 2.3TB <strong>of</strong> data on<br />

roughly 2 million patients. The search terms used were: bisphosphonates,<br />

pamidronate, zoledronic acid, multiple myeloma, plasma cell disorders,<br />

osteonecrosis <strong>of</strong> the jaw, jaw abscess, dental abscess, among other terms<br />

described in literature. Data was abstracted onto a standardized form by 2<br />

trained abstractors and validated by a clinician reviewer (BJE). Known and<br />

hypothesized new risk factors were abstracted, including duration <strong>of</strong><br />

myeloma, treatment used, duration <strong>of</strong> bisphosphonate use, renal function<br />

indices, chemotherapy (vincristine, doxorubicin (A), dexamethasone (D) ,<br />

thalidomide (T), cisplatin (P), cyclophosphamide (C), etoposide (E), novel<br />

agents [bevacizumab, sorafenib, angiostatin]) GCSF, smoking, and MM<br />

clinical stage. Analyses included T test, Wilcoxon, and log rank analysis.<br />

Results: ONJ occurring after MM diagnosis was identified in 33 cases out <strong>of</strong><br />

a total <strong>of</strong> 233 cases <strong>of</strong> MM. ZOL, VAD, DT-PACE, and diabetes were more<br />

common in ONJ cases. Log rank analysis identified 2 risk factors for ONJ,<br />

the use <strong>of</strong> DT-PACE (p� 0.003), and complete and partial remission<br />

(p�0.007). Stem cell transplant and chronic kidney disease were not<br />

associated with ONJ. Conclusions: We identified novel risk factors for ONJ<br />

in MM, mainly partial or complete remission and use <strong>of</strong> DT-PACE. These<br />

results should prompt clinicians to heightened awareness and increased<br />

surveillance for the symptoms <strong>of</strong> ONJ for patients treated with DT-PACE.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


6624 General Poster Session (Board #25C), Mon, 1:15 PM-5:15 PM<br />

Adverse events (AEs) and the return <strong>of</strong> myel<strong>of</strong>ibrosis (MF)-related symptoms<br />

after interruption or discontinuation <strong>of</strong> ruxolitinib (RUX) therapy.<br />

Presenting Author: Srdan Verstovsek, University <strong>of</strong> Texas M. D. Anderson<br />

Cancer Center, Houston, TX<br />

Background: RUX is a JAK1/JAK2 inhibitor that demonstrated significant<br />

clinical benefit in patients (pts) with MF in COMFORT-I (NCT00952289),<br />

a phase 3, randomized (1:1), double-blind, placebo (PBO)-controlled study<br />

(N�309). Methods: Pts assessed MF-related symptoms daily using the<br />

modified Myel<strong>of</strong>ibrosis Symptom Assessment Form v2.0; Total Symptom<br />

Score (TSS) was calculated from individual scores for abdominal discomfort,<br />

pain under left ribs, early satiety, night sweats, itching, and bone/<br />

muscle pain. Therapy was interrupted if platelet or absolute neutrophil<br />

count fell below 50,000/�L or 500/�L, respectively. AEs were evaluated<br />

during treatment interruption or discontinuation. The protocol suggested<br />

an optional tapering strategy and possible addition <strong>of</strong> steroids if RUX<br />

therapy was discontinued for reasons other than thrombocytopenia. Results:<br />

In RUX-treated patients with treatment interruption, TSS gradually returned<br />

to baseline levels over approximately 1 week. The most common AE<br />

leading to treatment interruption or discontinuation in each group was<br />

thrombocytopenia (n�11, RUX) and abdominal pain (n�4, PBO). Of<br />

these, only 1 RUX pt discontinued for thrombocytopenia. A summary <strong>of</strong><br />

new onset/worsening AE data after treatment interruption or discontinuation<br />

is presented (Table). Of 58 pts who discontinued study treatment, 23<br />

(n�10, RUX; n�13, PBO) experienced a total <strong>of</strong> 43 SAEs (19, RUX; 24,<br />

PBO) that showed no specific pattern or difference between treatment<br />

groups. Four <strong>of</strong> 21 RUX-treated pts had dose tapering following study drug<br />

discontinuation. Conclusions: Apart from the expected return <strong>of</strong> MF-related<br />

symptoms, there was no pattern <strong>of</strong> AEs to suggest that RUX interruption or<br />

discontinuation is associated with a specific withdrawal syndrome.<br />

RUX (N�155) PBO (N�151)<br />

Treatment interruption, n 49 54<br />

Mean duration, days 16 9<br />

Grade >3 AEs, n 8 7<br />

SAEs, n 3<br />

3<br />

Gastrointestinal hemorrhage; Anemia; pulmonary edema;<br />

fatigue and neutropenic hepatic encephalopathy<br />

fever; urosepsis<br />

and acute gout<br />

Treatment discontinuation, n 21 37<br />

Grade >3 AEs, n 12 17<br />

SAEs, n 10 13<br />

6626^ General Poster Session (Board #26B), Mon, 1:15 PM-5:15 PM<br />

Health-related quality <strong>of</strong> life (HRQoL) and symptom burden in patients<br />

(Pts) with myel<strong>of</strong>ibrosis (MF) in the COMFORT-II study. Presenting Author:<br />

Jean-Jacques Kiladjian, Hôpital Saint-Louis et Université Paris Diderot,<br />

Paris, France<br />

Background: Ruxolitinib has demonstrated rapid and durable reductions in<br />

splenomegaly and improved disease-related symptoms and QoL in 2 phase<br />

3 studies (COMFORT-I and -II) in pts with primary MF (PMF), postpolycythemia<br />

vera-MF (PPV-MF), or post-essential thrombocythemia-MF<br />

(PET-MF). The prevalence <strong>of</strong> individual symptoms among these pts has not<br />

been defined. We evaluated the baseline HRQoL and symptoms among pts<br />

enrolled in COMFORT-II. Methods: COMFORT-II is a randomized, openlabel,<br />

multicenter, phase 3 study comparing ruxolitinib with best available<br />

therapy. HRQoL and symptoms were assessed at baseline using the<br />

European Organisation for the Research and Treatment <strong>of</strong> Cancer QoL<br />

Questionnaire–Core 30 (EORTC QLQ-C30) and Functional Assessment <strong>of</strong><br />

Cancer Therapy–Lymphoma (FACT-Lym); this analysis summarizes these<br />

scores for all pts, regardless <strong>of</strong> assigned treatment. Results: In COMFORT-II<br />

(N � 219), 52% <strong>of</strong> pts were aged � 65 years and 57% were male. By IPSS<br />

criteria (Cervantes et al. 2009), 40% had intermediate-2 and 60% had<br />

high-risk MF. Mean (95% CI) EORTC global health status/QoL (53.7<br />

[50.6-56.7]; median, 50.0) and FACT-General total scores (73.0 [70.8-<br />

75.2]) were comparable to those for pts <strong>of</strong> similar age with other cancers<br />

(Oliva et al. 2011: median global health status score <strong>of</strong> 50 for acute<br />

myeloid leukemia [AML]). The most frequent symptoms (reported as “quite<br />

a bit” or “very much”) were fatigue (54%), dyspnea (30%), insomnia<br />

(30%), pain (29%), night sweats (23%), and itching (21%), and there were<br />

differences in baseline symptoms across MF subtypes (Table). Conclusions:<br />

This analysis shows that pts with MF experience severe disease-related<br />

symptoms and have diminished HRQoL similar to pts with AML, but<br />

because pts with MF have a longer life expectancy (an average <strong>of</strong> 2.3 to 4<br />

years for high and intermediate-2 risk pts, respectively), they may suffer<br />

with a reduced QoL for many years.<br />

% “quite a bit” or “very much”<br />

PMF<br />

(N � 116)<br />

PPV-MF<br />

(N � 68)<br />

PET-MF<br />

(N � 35)<br />

Fatigue 59.2 46.1 53.2<br />

Dyspnea 28.7 30.2 31.2<br />

Insomnia 32.7 25.4 28.2<br />

Pain 25.9 28.6 41.9<br />

Night sweats 21.3 27.3 18.2<br />

Itching 16.8 28.8 21.3<br />

Weight loss 12.1 22.7 6.3<br />

Leukemia, Myelodysplasia, and Transplantation<br />

6625^ General Poster Session (Board #26A), Mon, 1:15 PM-5:15 PM<br />

Association <strong>of</strong> cytokine levels and reductions in spleen size in COMFORT-<br />

II, a phase III study comparing ruxolitinib to best available therapy (BAT).<br />

Presenting Author: Claire N. Harrison, Guy’s and St. Thomas’ NHS<br />

Foundation Trust, London, United Kingdom<br />

Background: Ruxolitinib is a potent and selective oral JAK1/2 inhibitor that<br />

has been approved for the treatment <strong>of</strong> myel<strong>of</strong>ibrosis (MF). Ruxolitinib has<br />

demonstrated rapid and durable reductions in splenomegaly and improved<br />

disease-related symptoms and quality <strong>of</strong> life (QoL) in 2 phase 3 studies<br />

(COMFORT-I and –II) in patients (pts) with primary MF (PMF), postpolycythemia<br />

vera-MF (PPV-MF), or post-essential thrombocythemia-MF<br />

(PET-MF). This analysis evaluated associations between cytokine levels<br />

and spleen size reductions in the COMFORT-II study. Methods: COM-<br />

FORT-II is a randomized (2:1), open-label, phase 3 study comparing the<br />

safety and efficacy <strong>of</strong> ruxolitinib with BAT. Spleen volume was measured by<br />

MRI every 12 weeks and spleen length by palpation at each study visit.<br />

Plasma samples were analyzed using Rules Based Medicines Human MAP<br />

v1.6; 89 cytokines were measured at BL and wks 4, 24, and 48. Simple<br />

linear regression was used to evaluate the correlation between BL or change<br />

from BL in cytokine levels with % change <strong>of</strong> spleen size. Results: In the<br />

ruxolitinib arm, association was found between changes in TNF-� and<br />

leptin levels and % spleen volume reduction at wk 24 (TNF-�: N� 93;<br />

correlation coefficient [R] � 0.43; false discovery rate adjusted P value [P]<br />

� .01; leptin: N � 96; R � -0.28; P � .09) and wk 48 (TNF-�:N� 86; R<br />

� 0.43; P � .01; leptin: N � 87; R � -0.34; P � .02) that was not<br />

observed with BAT and was independent <strong>of</strong> JAK2V617F status. For<br />

ruxolitinib-treated JAK2V617F� pts, higher leptin levels at BL were<br />

associated with greater % spleen length reductions at wk 48 (N � 45; R �<br />

-0.44; P � .11). A clear trend was observed for increased leptin levels that<br />

preceded weight gain on ruxolitinib treatment. For ruxolitinib-treated<br />

JAK2V617F� pts, decreased IL-8 at wk 4 was associated with % spleen<br />

volume reductions at wk 24 (N � 68; R � 0.28; P � .24) and wk 48 (N �<br />

60; R � 0.38; P � .15). Conclusions: This analysis has shown statistically<br />

significant associations between changes in cytokine levels and spleen size<br />

reductions. Further analysis is in progress to determine associations<br />

between cytokine levels and QoL or symptoms and to confirm these<br />

observations in an independent data set.<br />

6627 General Poster Session (Board #26C), Mon, 1:15 PM-5:15 PM<br />

Post hoc analysis <strong>of</strong> association between treatment response and various<br />

indicators <strong>of</strong> efficacy and safety in a randomized phase III trial <strong>of</strong><br />

decitabine in older patients with acute myeloid leukemia. Presenting<br />

Author: Mark D. Minden, Princess Margaret Hospital, Toronto, ON, Canada<br />

Background: In a recent, large phase III trial (NCT00260832; Kantarjian,<br />

JCO; in press), 485 patients �65y with newly diagnosed acute myeloid<br />

leukemia (AML) received, every 4 wks, treatment choice (TC) <strong>of</strong> either<br />

supportive care or cytarabine (20 mg/m 2 subcutaneous injection, 10<br />

consecutive days) or decitabine (DAC) 20 mg/m2 (1-h intravenous [IV]<br />

infusion, 5 consecutive days). This post hoc analysis investigated relationships<br />

between response to treatment and indicators <strong>of</strong> efficacy and safety.<br />

Methods: Response was defined as morphologic complete remission (CR),<br />

or CR with incomplete blood count recovery (CRi) or partial response (PR).<br />

Transfusions (red blood cell [RBC] or platelets [PLT]), IV antibiotic use, and<br />

dose modifications were tabulated for responders and nonresponders to<br />

DAC or TC during the treatment period. Results: Fewer responders than<br />

nonresponders had dose modifications (30.4% vs 64.5%, respectively;<br />

P�.0001). Antibiotic use and transfusions were similar in both groups.<br />

Overall survival for responders was 16.1–18.5 mo vs 4.2–4.9 mo for<br />

nonresponders. Conclusions: These data suggest that response to DAC or TC<br />

treatment predicts clinically relevant benefits, with fewer dose modifications<br />

in older patients with newly diagnosed AML. The number <strong>of</strong><br />

transfusions and antibiotic use was impacted by the longer survival time <strong>of</strong><br />

responders vs nonresponders. Data on the impact <strong>of</strong> early response are<br />

being analyzed.<br />

Parameter*<br />

Nonresponders<br />

(n�383)<br />

447s<br />

CR�CRi�PR responders<br />

(n�102) p value †<br />

Overall survival, median, mo<br />

IV antibiotic use<br />

4.2–4.9 16.1–18.5 –<br />

Number <strong>of</strong> patients 383 102<br />

Yes, n (%)<br />

PLT transfusions<br />

77 (20.1) 14 (13.7) .143<br />

Number <strong>of</strong> patients 279 75<br />

Mean (SD) 11.64 (14.7) 13.67 (21.7) .447<br />

Median (range)<br />

RBC transfusions<br />

7 (1–110) 7 (1–136)<br />

Number <strong>of</strong> patients 342 97<br />

Mean (SD) 10.68 (12.2) 14.38 (14.5) .023<br />

Median (range)<br />

Dose modifications<br />

7 (1–102) 11 (1–89)<br />

Number <strong>of</strong> patients 383 102<br />

Yes, n (%) 247 (64.5) 31 (30.4) �.0001<br />

*All patients who had both response and parameter data. † Based on 2 sample t test to compare<br />

means; 2 proportion Z test to compare proportions.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


448s Leukemia, Myelodysplasia, and Transplantation<br />

6628 General Poster Session (Board #27A), Mon, 1:15 PM-5:15 PM<br />

Regional variation in treatment costs and survival for elderly patients with<br />

myelodysplastic syndromes. Presenting Author: Xiaomei Ma, Yale University<br />

School <strong>of</strong> Medicine, New Haven, CT<br />

Background: Myelodysplastic syndromes (MDS) occur primarily in the<br />

elderly (�65 years). The expected 5-year cost for an elderly MDS patient<br />

tops $63,200 in 2009 US$. This study assessed regional variation in the<br />

cost <strong>of</strong> care and survival for elderly MDS patients. Methods: Using the<br />

Surveillance Epidemiology and End Results–Medicare data, we identified<br />

primary MDS patients aged 66-99 years diagnosed from 2001-2007, had<br />

continuous fee-for-service coverage for <strong>Part</strong>s A and B, and had no history <strong>of</strong><br />

other cancer. We assigned patients to Dartmouth Atlas <strong>of</strong> Health Care<br />

Hospital Referral Regions (HRRs) based on their residence at time <strong>of</strong><br />

diagnosis. We also selected controls from a 5% sample <strong>of</strong> Medicare<br />

beneficiaries without cancer and matched controls 1:1 to patients by HRR,<br />

age, sex, pre-diagnosis cost and comorbidity. All Medicare claims through<br />

2009 were tallied, and MDS-related costs were defined as the difference<br />

between the payments for a patient and a matched control. Results: With<br />

6244 patients in 73 HRRs, the average 3-year MDS-related cost varied<br />

across HRRs, ranging from $12,900 to $83,600 (2009 US$). Patients in<br />

high-spending regions had more comorbidities and higher Medicare costs<br />

before diagnosis and were more likely to be racial minorities and live in<br />

lower-income areas. Three-year survival ranged from 13.0% to 62.1%.<br />

However, there was no significant correlation between 3-year costs and<br />

3-year survival (��-0.06, p�0.61). The hazard ratios (HR) for higher<br />

spending regions compared to the lowest-expenditure region were near 1,<br />

after controlling for covariates including MDS subtype (2nd quartile:<br />

HR�1.03, 95% CI: 0.94-1.12; 2nd quartile: HR�1.04, 95% CI: 0.95-<br />

1.14; 4th quartile: HR�0.98, 95% CI: 0.90-1.08). Conclusions: We<br />

observed considerable regional variation in Medicare expenditure on elderly<br />

MDS patients during the first 3 years post-diagnosis. However, patients in<br />

higher cost regions had similar 3-year survival to patients in lower cost<br />

regions. Given the substantial economic burden <strong>of</strong> MDS and Medicare’s<br />

current fiscal challenges, it is important to further assess the factors<br />

associated with higher regional costs and to improve the care <strong>of</strong> MDS<br />

patients in a cost-efficient way.<br />

6630 General Poster Session (Board #27C), Mon, 1:15 PM-5:15 PM<br />

Preliminary safety and efficacy <strong>of</strong> ruxolitinib in patients (pts) with primary<br />

and secondary myel<strong>of</strong>ibrosis (MF) with platelet counts (PC) <strong>of</strong> 50–<br />

100x109 /L. Presenting Author: Moshe Talpaz, Comprehensive Cancer<br />

Center, University <strong>of</strong> Michigan, Ann Arbor, MI<br />

Background: Ruxolitinib (RUX) has demonstrated clinical benefit as therapy<br />

for MF. This study explores the safety and efficacy <strong>of</strong> RUX in pts with MF<br />

and low PC, where clinical data are limited. Methods: In this phase II study,<br />

pts with intermediate-1 to high-risk MF, and PC <strong>of</strong> 50–100x109 /L started<br />

RUX at 5 mg BID. Doses could be increased in 5 mg QD increments every 4<br />

weeks (wks) beginning at wk 4. Doses were decreased or held for PC<br />

�35x109 /L and �25x109 /L respectively. Assessments: Total Symptom<br />

Score (TSS, using the modified MF Symptom Assessment Form v2.0, and<br />

comprised <strong>of</strong> scores from 0�absent to 10�worst imaginable for night<br />

sweats, itching, bone/muscle pain, early satiety, abdominal discomfort and<br />

pain under left ribs); Patient Global Impression <strong>of</strong> Change (PGIC, a 7-point<br />

scale ranging from “very much improved” to “very much worse”); spleen<br />

size by palpation and by MRI (data not yet available); and safety. Results: At<br />

the time <strong>of</strong> analysis, 23 pts had completed �4 wks on study. At baseline:<br />

mean PC�72x109 /L; high (4%), intermediate-2 (52%) and intermediate-1<br />

(44%) risk group; mean spleen length�16.6 cm; mean TSS�25.5. At the<br />

time <strong>of</strong> analysis, 4%, 40%, 26%, 26% and 4% <strong>of</strong> pts were receiving RUX 5<br />

mg QD, 5 mg BID, 5 mg AM/10 mg PM, 10 mg BID, and 10 mg AM/15 mg<br />

PM, respectively. At wk 4 (all pts on 5 mg BID), mean TSS improved 14%;<br />

13% had �50% improvement. At wk 8 (52% on 5 mg AM/10 mg PM),<br />

mean TSS improved 23%; 30% had �50% improvement. Mean spleen<br />

length reduction <strong>of</strong> 22% (wk 4) and 27% (wk 8) was observed, and PGIC<br />

scores <strong>of</strong> “much improved” or “very much improved” were reported in 35%<br />

(wk 4) and 39% (wk 8) <strong>of</strong> pts. There were no Grade 4 PC, no dose holds for<br />

AEs and no discontinuations; 1 pRBC transfusion-dependent pt had Grade<br />

4 anemia. Three pts experienced a total <strong>of</strong> 4 SAEs (fever; hypnagogic<br />

dreams; and spleen pain/pneumonitis) which resolved while on treatment.<br />

Conclusions: Effects on spleen size, PGIC, and TSS, even over the first<br />

weeks at low doses, are superior to those observed in the placebo group in<br />

the COMFORT-I study. These preliminary findings suggest a dosing strategy<br />

starting with 5 mg BID RUX with subsequent dose optimization may be<br />

efficacious and well tolerated in MF pts who have low platelets.<br />

6629 General Poster Session (Board #27B), Mon, 1:15 PM-5:15 PM<br />

History <strong>of</strong> autoimmunity to predict clinical response to DNA methyltransferase<br />

inhibitors (DNMTI) in myelodysplastic syndromes (MDS), and<br />

MDS-derived acute myeloid leukemias (AML). Presenting Author: Ashkan<br />

Emadi, The Johns Hopkins University, Baltimore, MD<br />

Background: MDS is comprised <strong>of</strong> a heterogeneous group <strong>of</strong> clonal myeloid<br />

disorders. Chronic immune stimulation has been reported as a trigger for<br />

the development <strong>of</strong> a subset <strong>of</strong> MDS, with increased autoreactive cytotoxic<br />

T cells present in the bone marrow. Autoimmunity has been associated with<br />

MDS and other marrow failures. DNMTIs, 5-azacytidine and decitabine, are<br />

approved for the treatment <strong>of</strong> MDS. In addition to epigenetic impacts,<br />

these agents may have immunomodulatory effects, including augmentation<br />

<strong>of</strong> MAGE-related anti-tumor response. These reports and clinical observations<br />

led us to hypothesize that MDS patients with a history <strong>of</strong> autoimmunity<br />

may be more responsive to DNMTIs. Methods: To identify patients with<br />

MDS, a retrospective database review (2007-2011) was performed at<br />

Johns Hopkins Hospital (JHH) and Massachusetts General Hospital (MGH).<br />

The MGH data also included those with AML whose disease had progressed<br />

from MDS. Past medical history <strong>of</strong> autoimmune disorders, diagnosis, blood<br />

counts, flow cytometry and cytogenetics were reviewed. Patients with<br />

aplastic anemia, paroxysmal nocturnal hemoglobinuria or non-malignant<br />

etiologies <strong>of</strong> cytopenia were excluded. Patients with MDS were further<br />

studied if they were treated with DNMTI. Results: Of 137 patients with MDS<br />

or MDS/AML, 23 had a documented history <strong>of</strong> autoimmunity in the medical<br />

record. Of these, 15 (65.2%) experienced a response to therapy as defined<br />

by the International Working Group. Of 114 patients without a documented<br />

history <strong>of</strong> autoimmunity, 34 (29.8%) achieved a response during therapy, a<br />

significantly lower percentage as compared to those with autoimmune<br />

conditions (p-value 0.002, t-test). The majority <strong>of</strong> responding patients with<br />

a history <strong>of</strong> autoimmunity displayed a normal karyotype (9 <strong>of</strong> 15 patients).<br />

Conclusions: A history or co-presence <strong>of</strong> an autoimmune disorder may<br />

predict a high likelihood <strong>of</strong> achieving a clinical response to DNMTIs.<br />

Correlative studies in this unique population <strong>of</strong> patients with MDS and<br />

MDS/AML and prospective clinical studies are needed to improve our<br />

understanding <strong>of</strong> the possible mechanism <strong>of</strong> action <strong>of</strong> DNMTIs in these<br />

patients.<br />

6631 General Poster Session (Board #28A), Mon, 1:15 PM-5:15 PM<br />

Correlation <strong>of</strong> TCR diversity with immune reconstitution after cord blood<br />

transplant. Presenting Author: Ryan Emerson, Adaptive Biotechnologies,<br />

Seattle, WA<br />

Background: Umbilical cord blood is an important source <strong>of</strong> hematopoietic<br />

stem cells for allogeneic transplants used to treat hematologic disorders<br />

and malignancies. Stem cells from cord blood has several advantages over<br />

other sources (peripheral blood or marrow) including reduced incidence <strong>of</strong><br />

graft-versus-host disease (GvHD) which allows less strict donor-patient<br />

HLA-type matching requirements. However, hematopoietic recovery (both<br />

myeloid and platelet engraftment) and immune reconstitution is significantly<br />

delayed in CBT patients resulting in patients remaining at high risk<br />

<strong>of</strong> infection for an extended period <strong>of</strong> time. Though the high risk <strong>of</strong><br />

infections to patients is well-documented and recognized, physicians lack<br />

an objective means to monitor each patient’s progress toward functional<br />

reconstitution <strong>of</strong> the adaptive immune system. Our project aims to test if<br />

our measure <strong>of</strong> TCR diversity is a better proxy <strong>of</strong> immune reconstitution in<br />

CBT patients. Methods: To do this, we compare our proposed measure <strong>of</strong><br />

immune reconstitution, TCRB CDR3 sequence diversity and richness over<br />

time, against the observed clinical course <strong>of</strong> severe infection in patients<br />

who have received CBT. Previously, peripheral blood mononuclear cells<br />

(PBMC) were collected and cyropreserved from 35 CBT recipients at six<br />

time points, pre-transplant, and 28, 56, 100, 180, and 365 days<br />

post-transplant. Concordantly, clinical outcomes for these patients were<br />

collected for up to four years post-transplant. We use a t-test to test if size<br />

<strong>of</strong> TCR repertoire differs between CBT patients with non-relapse related<br />

mortality and all other CBT patients. Results: Our TCR sequencing method<br />

was successful at measuring immune reconstitution, and TCR diversity is<br />

predictive <strong>of</strong> subsequent mortality from severe infection by 56 days<br />

post-transplant. Conclusions: TCR repertoire sequencing is a viable method<br />

to monitor immune reconstitution.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


6632 General Poster Session (Board #28B), Mon, 1:15 PM-5:15 PM<br />

Post hoc analysis <strong>of</strong> relationship between baseline white blood cell count<br />

and renal and hepatic function and response in a randomized phase III trial<br />

<strong>of</strong> decitabine in patients age 65 or older with acute myeloid leukemia.<br />

Presenting Author: Jacques Delaunay, University <strong>of</strong> Nantes, Nantes,<br />

France<br />

Background: A phase III trial (NCT00260832) in patients (N�485) �65y<br />

with newly diagnosed acute myeloid leukemia (AML) was conducted<br />

(Kantarjian, JCO; in press). Every 4 wk, patients received decitabine (DAC)<br />

20 mg/m 2 (1-h intravenous infusion, 5 successive days) or treatment<br />

choice (TC) with either supportive care or cytarabine (20 mg/m2 subcutaneous<br />

injection daily, 10 successive days). This post hoc analysis examined<br />

whether baseline (BL) renal and hepatic function and white blood cell<br />

(WBC) counts were associated with response to DAC or TC. Methods: For<br />

patients with available data, BL WBC count and markers <strong>of</strong> renal function<br />

(blood urea nitrogen [BUN], creatinine) and liver function (ALT, AST,<br />

albumin) were tabulated for patients with/without a response to DAC or TC.<br />

Response was defined as morphologic complete remission (CR), CR with<br />

incomplete blood count recovery (CRi), or partial remission (PR). Results:<br />

Nonresponders had a higher mean BL creatinine vs responders (86.78 vs<br />

80.23 mmol/L, respectively; P�.005); with no differences in BL BUN<br />

levels. There were no other between-group differences. Conclusions: This<br />

analysis suggests that there is no relationship between BL WBC or hepatic<br />

function and response to treatment with DAC or TC. Although there was no<br />

difference in BL BUN, higher mean creatinine levels in nonresponders may<br />

suggest a prognostic relationship but further studies are needed to clarify.<br />

Parameter<br />

Nonresponder<br />

to DAC or TC<br />

Responder to DAC or TC<br />

(CR�CRi�PR) P value<br />

WBC (10 9 /L)<br />

n 372 101<br />

Mean (SD) 9.25 (13.98) 7.02 (12.25) .118<br />

Median (range) 3.72 (0.33-126.60) 2.70 (0.43-95.57)<br />

ALT (U/L)<br />

n 372 100<br />

Mean (SD) 25.32 (35.53) 20.79 (14.53) .054<br />

Median (range) 18 (5-614) 17 (6-103)<br />

AST (U/L)<br />

n 371 101<br />

Mean (SD) 23.82 (15.93) 21.74 (9.84) .106<br />

Median (range) 20 (5-203) 18 (9-65)<br />

Albumin (g/L)<br />

n 377 102<br />

Mean (SD) 34.18 (5.47) 35.07 (5.34) .139<br />

Median (range) 35 (19-48) 36 (16-44)<br />

BUN (mmol/L)<br />

n 377 102<br />

Mean (SD) 6.97 (2.65) 6.48 (2.27) .064<br />

Median (range) 6.40 (1.3-23.7) 6.15 (2.9-19.5)<br />

Creatinine (mmol/L)<br />

n 377 102<br />

Mean (SD) 86.78 (27.08) 80.23 (18.88) .005<br />

Median (range) 81 (38-254) 78 (44-169)<br />

TPS6634 General Poster Session (Board #29A), Mon, 1:15 PM-5:15 PM<br />

Dasatinib plus SMO antagonist versus dasatinib alone for treating patients<br />

(pts) with newly diagnosed Philadelphia chromosome-positive (Ph�) chronic<br />

myeloid leukemia in chronic phase (CML-CP): Design <strong>of</strong> CA180-363, a<br />

phase II, open-label randomized trial. Presenting Author: Geralyn C.<br />

Trudel, Bristol-Myers Squibb, Montreal, QC, Canada<br />

Background: Dasatinib 100 mg once daily (QD) is approved as first-line<br />

therapy for pts with newly diagnosed Ph� CML-CP and those who are<br />

resistant or intolerant to prior therapy, including imatinib, based on<br />

demonstrated efficacy and tolerability. BMS-833923 (SMO antagonist),<br />

selectively blocks hedgehog (Hh) pathway signaling by binding to and<br />

antagonizing Smoothened (SMO), preventing downstream signaling and<br />

activation <strong>of</strong> target genes. In preclinical studies, SMO loss or inhibition<br />

reduces hematopoietic stem cell renewal, induces CML stem cell depletion,<br />

and inhibits imatinib-resistant CML cell growth (Dierks, Cancer Cell<br />

2008; Zhao, Nature 2009). In pts with CML, Hh signaling genes (Sonic<br />

hedgehog, SMO, and Gli1) are significantly upregulated compared with<br />

control pts (Long, J Exp Clin Cancer Res 2011), suggesting that drug<br />

resistance and disease recurrence may be overcome by targeting the Hh<br />

signaling pathway. Methods: CA180-363 is a phase 2 trial to assess<br />

dasatinib activity with or without SMO antagonist in pts with newly<br />

diagnosed Ph� CML-CP. Pts receive dasatinib 100 mg QD for 12 months<br />

and are then randomized 1:1 to continuing dasatinib 100 mg QD with or<br />

without SMO antagonist up to 24 months, followed by dasatinib 100 mg<br />

QD alone until end <strong>of</strong> study (60 months). Randomization is stratified by<br />

Sokal score at diagnosis and major molecular response (MMR) at 12<br />

months (yes/no). Eligible pts are aged �18 years, have an Eastern<br />

Cooperative Group (ECOG) performance status <strong>of</strong> 0-2, and previously<br />

untreated Ph� CML-CP diagnosed within 6 months <strong>of</strong> enrollment. Primary<br />

endpoint (evaluated in pts who do not achieve MMR prior to randomization)<br />

is comparison <strong>of</strong> MMR rates for dasatinib alone vs dasatinib plus SMO<br />

antagonist. Other endpoints (evaluated in all pts) are complete molecular<br />

response, progression-free survival, event-free survival, transformation-free<br />

survival, and safety <strong>of</strong> the combination regimen. Recruitment started in<br />

September 2011 (14 pts enrolled to date); estimated primary completion<br />

date is November 2014. <strong>Clinical</strong>Trials.gov ID: NCT01357655.<br />

Leukemia, Myelodysplasia, and Transplantation<br />

449s<br />

6633 General Poster Session (Board #28C), Mon, 1:15 PM-5:15 PM<br />

Phase II study <strong>of</strong> short CHOP-rituximab combination with early consolidation<br />

with ibritumomab-tiuxetan-Y90 (IT-Y90 ) in non-pretreated patients age<br />

65 to 80 with CD20� diffuse large B-cell lymphoma (DLBCL). Presenting<br />

Author: Frederic Peyrade, Anticancer Center, Centre Antoine-Lacassagne,<br />

Nice, France<br />

Background: IT-Y90was approved in follicular lymphoma, and some data<br />

indicates that it could be used in DLBCL. It has been reported that early<br />

TEP-TDM negativity is associated with a longer survival in DLBCL, which<br />

suggest that these patients could benefit <strong>of</strong> a less intensive protocol.<br />

Methods: We conducted an international, open-label, phase II nonrandomised<br />

study, in patients aged between 65 and 80 with age-adjusted<br />

(aa) IPI score <strong>of</strong> 0 and 1 and CD20� DLBCL. The primary objective was to<br />

evaluate the efficacy <strong>of</strong> 3 cycles <strong>of</strong> RCHOP14 followed by, in case <strong>of</strong><br />

complete response (CR), an injection <strong>of</strong> IT-Y90 . Results: Thirty patients<br />

(M/F� 1) were included. Median age was 72.6 years (range 65 - 80).<br />

Patients had a stage III/IV, elevated LDH and IPI�1 in 38%, 16.6 and<br />

57% respectively. 25 patients received the full treatment. Five obtained an<br />

uncomplete FDG-PET response and were excluded after 2 RCHOP cycles.<br />

23 patients received the full IT-Y90 dosage (0.4mCi/Kg) and two received<br />

the attenuated dosage (0.3mCi/Kg). Mean treatment time was 54 days<br />

(median 52; min 48-max 69). The CR rate after RCHOP treatment was<br />

85% [95% CI: 65-94]. No treatment-related deaths occurred. Grade III-IV<br />

neutropenia and thrombocytopenia was observed in 45% and 4%, respectively.<br />

Five patients experienced at least one febrile neutropenia. After<br />

IT-Y90 , mean duration time for platelets �50G/l was 2.08 weeks (median<br />

2; min 0-max 5). Two patients required platelet transfusion. With a median<br />

follow-up <strong>of</strong> 29.5 months [95% CI: 23-39], the estimated 3-year PFS was<br />

90% [95% CI: 80-100] and the 3-year OS was 100% [95% CI: 100-100].<br />

Among patients treated with 90Y-IT, only 3 relapsed were recorded (at 6, 16<br />

and 24 months). Conclusions: this study suggested the feasibility, and<br />

efficacy <strong>of</strong> a short regimen including one injection <strong>of</strong> IT-Y90for selected<br />

DLBCL in complete response after 3 cycles <strong>of</strong> RCHOP14. This results need<br />

to be confirmed by further studies.<br />

TPS6635 General Poster Session (Board #29B), Mon, 1:15 PM-5:15 PM<br />

Targeting microenvironment-mediated resistance in leukemias: Phase I<br />

trial <strong>of</strong> mobilization and elimination <strong>of</strong> FLT3-ITD� acute myelogenous<br />

leukemia (AML) stem/progenitor cells by plerixafor/g-CSF/sorafenib. Presenting<br />

Author: Michael Andreeff, University <strong>of</strong> Texas M. D. Anderson<br />

Cancer Center, Houston, TX<br />

Background: FLT3-ITD AML are associated with poor prognosis. We<br />

identified Sorafenib (S) as potent inhibitor <strong>of</strong> FLT3-ITD (Zhang W, JNCI,<br />

2008; Borthakur G., Haematologica, 2010). FLT3-ITD is associated with<br />

overexpression <strong>of</strong> chemokine receptor CXCR4 and we found increased in<br />

vivo activity <strong>of</strong> S combined with CXCR4 inhibitor Plerixafor (P) and G-CSF<br />

(G) (Zeng Z et.al. Blood 2009). Here we report first data testing this<br />

concept in patients with R/R FLT3-ITD AML. Methods: G (10 ug/kg) and<br />

P(240 ug/kg) were given s.c. QOD on days 1 – 13, S (400-600mg), S on d 1<br />

- 28(one cycle). G/P was held when blasts � 5x104/ uL. CD34, 38, 123,<br />

CXCR4 (1D9, 12G5), VLA4, CD44 and phospho-proteins were measured by<br />

flow cytometry. Results: 10 patients have been treated so far : 2 achieved<br />

CRp, 4 PR and 4 failed (NR), for an overall response rate <strong>of</strong> 6/10; 3/6<br />

responders and 4/4 NR were previously treated with FLT3 inhibitors. 4/10<br />

pts. developed hyperleukocytosis (and missed 1 to 5 doses <strong>of</strong> G/P), 6 skin<br />

rash and 3 hypertension. Analysis <strong>of</strong> cells mobilized in 22 cycles revealed a<br />

29-fold increase in WBC, 41-fold in absolute blasts, 77-fold in granulocytes.<br />

Increase in circulating stem/progenitor cells was as follows: CD34�:<br />

231-fold, CD34�/38- : 90-, CD34�/38-/123�(LSC) : 148-, CXCR4�:<br />

139-, VLA-4� : 68- and CD44�: 82-fold. Increase in LSC was correlated<br />

with baseline blasts and VLA4, not with CXCR4. FISH confirmed mobilization<br />

<strong>of</strong> leukemic cells. Increased levels <strong>of</strong> pERK and pAKT were observed in<br />

mobilized cells. Conclusion: The combination <strong>of</strong> G-CSF�Plerixafor appears<br />

superior in increasing circulating leukemic blasts and stem/progenitor cells<br />

in FLT3-ITD AML, as compared to Plerixafor alone in R/R AML(blast<br />

increase 2.1-fold; Uy et al. Blood, in press). Treatment resulted in 2/10<br />

CRp and 4/10 PRs. Mobilized stem/progenitor cells displayed increased<br />

MAPK/AKT signaling and increased CXCR4 expression. This is the first<br />

clinical report <strong>of</strong> G-CSF/Plerixafor for the “mobilization” <strong>of</strong> AML cells,<br />

aimed at removing them from their protective bone marrow microenvironment.<br />

The initial results are providing pro<strong>of</strong>-<strong>of</strong>–principle <strong>of</strong> this concept.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


450s Leukemia, Myelodysplasia, and Transplantation<br />

TPS6636 General Poster Session (Board #29C), Mon, 1:15 PM-5:15 PM<br />

A single-arm, open-label, multicenter study <strong>of</strong> complete molecular response<br />

(CMR) in patients with newly diagnosed Philadelphia chromosome–<br />

positive (Ph�) chronic myeloid leukemia in chronic phase (CML-CP)<br />

treated with nilotinib. Presenting Author: Michael R. Savona, Sarah<br />

Cannon Center for Blood Cancers, Tennessee Oncology, PLLC, Nashville,<br />

TN<br />

Background: With more-potent tyrosine kinase inhibitors, increasingly<br />

sensitive methods are required to monitor treatment response and quantify<br />

minimal residual disease (MRD). Molecular monitoring is recommended by<br />

National Comprehensive Cancer Network and European LeukemiaNet<br />

guidelines; major molecular response (MMR) correlates with optimal<br />

long-term outcomes. CMR, while implying absence <strong>of</strong> BCR-ABL transcripts,<br />

is defined by levels undetectable by current technology (generally<br />

�4.5 logs reduced below a standardized baseline using quantitative<br />

real-time polymerase chain reaction [RT-PCR]). Newer technologies that<br />

can detect larger reductions will be important in analyzing correlations<br />

between decreased disease burden and long-term outcomes and may assist<br />

in selecting patients for “stopping” therapy. Methods: This US study plans<br />

to enroll 100 adults with newly diagnosed Ph� CML-CP within 6 months <strong>of</strong><br />

diagnosis to receive nilotinib 300 mg twice daily (BID) for up to 2 years.<br />

BCR-ABL transcripts are quantified using RT-PCR at a central laboratory.<br />

An exploratory molecular assay (digital detection [DiD]) with an additional<br />

�1-log sensitivity and mutation detection and quantification will be<br />

evaluated. Primary endpoint: the rate <strong>of</strong> confirmed CMR at 24 months<br />

(CMR confirmed by second PCR sample within 3 months; �4.5-log<br />

reduction <strong>of</strong> BCR-ABL from standardized baseline on the international<br />

scale [IS], equivalent to BCR-ABL �0.0032% IS). Dose escalation to 400<br />

mg BID is allowed per protocol. Secondary endpoints: rate <strong>of</strong>, time to, and<br />

duration <strong>of</strong> complete cytogenetic response (CCyR), MMR, and CMR; rate <strong>of</strong><br />

and time to loss <strong>of</strong> CCyR, MMR, and CMR and to progression to accelerated<br />

phase/blast crisis; rate <strong>of</strong> CMR in dose-escalated patients; and event-free,<br />

progression-free, and overall survival. Exploratory endpoints: BCR-ABL<br />

trends and correlation <strong>of</strong> mutations with response. The DiD assay data will<br />

provide information on BCR-ABL trends below the current CMR threshold<br />

and assist in evaluating lower levels <strong>of</strong> MRD.<br />

TPS6638 General Poster Session (Board #30B), Mon, 1:15 PM-5:15 PM<br />

Prospective, observational registry <strong>of</strong> branded imatinib (IM) and nilotinib<br />

(NIL) exposure in pregnant women: Voluntary post-authorization safety<br />

study. Presenting Author: Matthews Juma, Novartis Pharmaceuticals, East<br />

Hanover, NJ<br />

Background: Family planning decisions for cancer patients <strong>of</strong> childbearing<br />

age are impacted by their diagnosis. IM and NIL are category D drugs<br />

(demonstrated risk to the fetus based on mechanism <strong>of</strong> action and findings<br />

in animals; however, the benefits <strong>of</strong> therapy may outweigh the potential risk<br />

to the fetus); women should be advised not to become pregnant when<br />

taking these drugs. Limited data exist concerning safety <strong>of</strong> these drugs<br />

during pregnancy and consequences <strong>of</strong> interrupting treatment. The registry<br />

does not recommend patients receiving IM or NIL become pregnant, but<br />

serves only to document exposures that occur, and collect information on<br />

pregnancy outcome, maternal course <strong>of</strong> disease, and infant follow-up.<br />

Methods: The registry intends to enroll 150 women (�18 years) treated with<br />

branded IM and NIL within 6 mo before or during pregnancy (generic IM<br />

and NIL reports are excluded). Schedule <strong>of</strong> visits and treatment is<br />

according to local standard <strong>of</strong> care. Women are divided into 2 exposure<br />

cohorts: 1) pregnancy/fetal: received tyrosine kinase inhibitors (TKIs)<br />

within 14 d <strong>of</strong> conception or at any time during pregnancy; 2) interrupted<br />

TKI: received TKIs within 6 mo before conception but interrupted TKIs in<br />

preparation for/due to pregnancy. To identify signals <strong>of</strong> teratogenicity, the<br />

registry uses a general population baseline rate <strong>of</strong> birth defects, and the<br />

prevalence <strong>of</strong> defects in cohorts 1 and 2 may be compared. Cases are<br />

quantitatively analyzed for the emergence <strong>of</strong> unique defects or patterns <strong>of</strong><br />

defects. Primary objective: monitor TKI-exposed pregnancies to estimate<br />

the prevalence <strong>of</strong> birth defects (calculated by dividing number <strong>of</strong> birth<br />

defects by total number <strong>of</strong> exposed live births from cohort 1). Secondary<br />

objectives are to: 1) determine the impact <strong>of</strong> treatment interruption on<br />

maternal disease status; 2) assess and estimate the prevalence <strong>of</strong> serious<br />

adverse pregnancy outcomes; and 3) assess and estimate the prevalence <strong>of</strong><br />

developmental delays and functional deficits among infants during the first<br />

year <strong>of</strong> life. Maternal disease status is analyzed, comparing disease status<br />

at registration, pregnancy outcome, and 1 y after birth, stratified by length<br />

<strong>of</strong> TKI interruption.<br />

TPS6637 General Poster Session (Board #30A), Mon, 1:15 PM-5:15 PM<br />

VALOR, an adaptive design, pivotal phase III trial <strong>of</strong> vosaroxin or placebo in<br />

combination with cytarabine in first relapsed or refractory acute myeloid<br />

leukemia. Presenting Author: Farhad Ravandi, University <strong>of</strong> Texas M. D.<br />

Anderson Cancer Center, Houston, TX<br />

Background: The promising phase 2 activity/tolerability pr<strong>of</strong>ile <strong>of</strong> vosaroxin<br />

with cytarabine in first relapsed or refractory AML (N�69) with median<br />

overall survival (OS) 7.1 mo, combined CR 28% (CR 25%), median LFS 25<br />

mo and 30-day all-cause mortality 3%, supported phase 3 trial. VALOR<br />

(NCT01191801), a phase 3, randomized, controlled, double-blind trial,<br />

evaluates vosaroxin and cytarabine versus placebo and cytarabine in<br />

patients with first relapsed or refractory AML and incorporates an adaptive<br />

design. Primary objective is OS; secondary/tertiary objectives include CR<br />

rates, safety, EFS, LFS, and transplantation rate. Key eligibility criteria:<br />

persistent AML or first relapsed AML after 1 or 2 induction cycles that<br />

include at least 1 regimen <strong>of</strong> cytarabine with an anthracycline/anthracenedione;<br />

adequate cardiac, hepatic, renal function; and adults with no upper<br />

age restriction. Methods: VALOR is recruiting patients at over 100 sites in<br />

14 countries in North America, Europe, and Australia/NZ. Enrollment<br />

opened Dec 2010; 183 patients enrolled through Dec 2011. Adaptive<br />

design: Base case assumed 40% improvement in median OS (hazard ratio,<br />

HR�0.71), 90% power, 2-sided alpha <strong>of</strong> 0.05. At the interim analysis<br />

(50% events), DSMB may recommend a 50% sample size increase from<br />

450 to 675 evaluable patients if results indicate a larger sample size is<br />

required to reduce the risk <strong>of</strong> failing to confirm a clinically meaningful OS<br />

benefit (Mehta, Pocock, Stat Med 2010). In consideration <strong>of</strong> FDA and EMA<br />

guidance on Data Monitoring Committees and adaptive design with respect<br />

to trial integrity, operational bias, firewalls, and data confidentiality and<br />

archival, VALOR uses the Access Control Execution System (ACES ) to<br />

store, share, and archive confidential DSMB reports in a secure environment<br />

with an audit trail, and to facilitate communication between the<br />

DSMB and trial sponsor. The DSMB periodically reviews a combination <strong>of</strong><br />

blinded and unblinded analyses while the study team remains blinded as<br />

specified in the DSMB charter. The DSMB recommended VALOR continue<br />

as planned after reviewing safety data in Dec 2011.<br />

TPS6639 General Poster Session (Board #30C), Mon, 1:15 PM-5:15 PM<br />

JAKARTA: A phase III, multicenter, randomized, double-blind, placebocontrolled,<br />

three-arm study <strong>of</strong> SAR302503 in patients with intermediate-2<br />

or high-risk primary myel<strong>of</strong>ibrosis (MF), post-polycythemia vera (PV) MF, or<br />

post-essential thrombocythemia (ET) MF with splenomegaly. Presenting<br />

Author: Animesh Dev Pardanani, Mayo Clinic, Rochester, MN<br />

Background: SAR302503 is an orally administered selective JAK-2 inhibitor<br />

being evaluated for the treatment <strong>of</strong> MF. In a phase 1 study,<br />

SAR302503 reduced spleen size and disease-related symptoms and<br />

provided clinical benefit, including a reduction in the JAK2V617F allele<br />

burden, with an acceptable safety pr<strong>of</strong>ile in patients with primary MF,<br />

post-PV MF, or post-ET MF (J Clin Oncol 2011;29:789). An ongoing<br />

open-label extension <strong>of</strong> this trial confirmed the durability <strong>of</strong> those results<br />

and found that doses between 400–500 mg/day <strong>of</strong> SAR302503 represented<br />

the optimal balance between safety and efficacy (Pardanani, ASH<br />

2011;Abs 3838). These results led us to initiate a Phase 3 trial in<br />

December 2011 to evaluate SAR302503 at doses <strong>of</strong> 400 mg/day and 500<br />

mg/day, compared with placebo, for the treatment <strong>of</strong> patients with MF<br />

(NCT01437787; EFC12153). Methods: Eligibility criteria include age <strong>of</strong> at<br />

least 18 years, platelets �50,000/�l, ECOG PS 0–2, and a diagnosis <strong>of</strong><br />

high- or intermediate-risk 2 primary MF, post-PV MF, or post-ET MF<br />

according to IPSS criteria (Blood 2009;113:2895). Patients are being<br />

randomized (1:1:1) to receive 400 mg or 500 mg <strong>of</strong> SAR302503 or<br />

placebo orally once a day for at least 6 consecutive 28-day cycles. Assigned<br />

treatment will continue as long as patients are deriving benefit or until<br />

progression or unacceptable toxicity. Cross-over is allowed after 6 cycles or<br />

in case <strong>of</strong> progression before completion <strong>of</strong> 6 cycles according to predefined<br />

criteria. The primary endpoint is spleen response (�35% reduction<br />

in spleen volume by MRI/CT at the end <strong>of</strong> cycle 6). Key secondary<br />

objectives are symptom response rate and durability <strong>of</strong> spleen response.<br />

Additional secondary endpoints include assessment <strong>of</strong> allele burden and<br />

bone marrow fibrosis reduction. It is planned to randomize 75 patients per<br />

treatment group at approximately 130 sites worldwide.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


TPS6640 General Poster Session (Board #31A), Mon, 1:15 PM-5:15 PM<br />

An open-label, multicenter, expanded access study assessing the safety<br />

and efficacy <strong>of</strong> oral ruxolitinib administered to patients with primary<br />

myel<strong>of</strong>ibrosis (PMF), post-polycythemia myel<strong>of</strong>ibrosis (PPV MF) or postessential<br />

thrombocythemia myel<strong>of</strong>ibrosis (PET-MF). Presenting Author:<br />

Philipp D. le Coutre, University <strong>of</strong> Medicine Berlin, Charité Campus<br />

Virchow, Berlin, Germany<br />

Background: Myel<strong>of</strong>ibrosis (MF) can be primary in origin or can progress<br />

from polycythemia vera (PPV-MF) or essential thrombocythemia (PET-MF).<br />

MF is characterized by cytopenias, reactive bone marrow fibrosis, splenomegaly,<br />

and constitutional symptoms including weight loss, fever, and<br />

night sweats. MF is associated with dysregulation <strong>of</strong> the Janus kinase<br />

pathway, irrespective <strong>of</strong> JAK2V617F mutation status. Recently, ruxolitinib<br />

(INCB018424) was approved for the treatment <strong>of</strong> MF on the basis <strong>of</strong> results<br />

from 2 phase 3 trials (COMFORT-I and -II). These trials demonstrated<br />

marked reductions in splenomegaly and symptom burden, and improvements<br />

in health-related quality <strong>of</strong> life (QoL) measures. Methods: JAK<br />

Inhibitor rUxolitinib in Myel<strong>of</strong>ibrosis Patients (JUMP) is a global, phase 3b<br />

expanded access trial (NCT01493414) designed to assess the safety and<br />

efficacy <strong>of</strong> ruxolitinib in adult pts with PMF, PPV-MF or PET-MF who are<br />

treatment naïve, and are intolerant <strong>of</strong>, or had progressed on any prior<br />

therapy. Inclusion criteria: peripheral blast count <strong>of</strong> �10%, adequate liver<br />

and renal function and intermediate-2 or high risk MF according to IPSS<br />

criteria or intermediate-1 risk MF with palpable spleen length � 5 cm. The<br />

primary endpoint is to assess the safety <strong>of</strong> ruxolitinib. Additional endpoints<br />

include the proportion <strong>of</strong> pts with a � 50% reduction in palpable spleen<br />

length, % change in white blood cell and platelet counts from baseline<br />

(BL), and change in packed red blood cell transfusion requirements.<br />

Changes from BL in QoL scores will be assessed using validated ECOG PS,<br />

FACT-Lymphoma, and FACIT instruments. Pts with a BL platelet count �<br />

200,000/�L will receive oral ruxolitinib 20 mg BID; pts with a BL platelet<br />

count <strong>of</strong> 100,000/�L to 200,000/�L will receive 15 mg BID; dose<br />

modifications are permitted for safety and efficacy. Global enrollment is<br />

open and currently includes 277 pts. This is planned to be the largest study<br />

ever conducted in pts with MF.<br />

Country #<strong>of</strong>Pts<br />

Austria 24<br />

Belgium 15<br />

Brazil 24<br />

Germany 173<br />

Spain 28<br />

Argentina 3<br />

Canada 3<br />

Netherlands 3<br />

Italy 4<br />

Total 277<br />

TPS6642^ General Poster Session (Board #31C), Mon, 1:15 PM-5:15 PM<br />

A phase Ib, open-label, dose-finding study <strong>of</strong> ruxolitinib in patients (pts)<br />

with primary myel<strong>of</strong>ibrosis (PMF), post-polycythemia vera-myel<strong>of</strong>ibrosis<br />

(PPV-MF), or post-essential thrombocythemia-myel<strong>of</strong>ibrosis (PET-MF) and<br />

baseline platelets (PLTs) 50 to


452s Lung Cancer—Non-small Cell Local-Regional/Small Cell/Other Thoracic Cancers<br />

7000 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Is consolidation chemotherapy after concurrent chemoradiotherapy beneficial<br />

for locally advanced non-small cell lung cancer? A pooled analysis <strong>of</strong><br />

the literature. Presenting Author: Satomi Yamamoto, National Hospital<br />

Organization Kinki-Chuo Chest Medical Center, Sakai, Japan<br />

Background: There is a growing interest on the efficacy <strong>of</strong> maintenance<br />

chemotherapy to metastatic non-small cell lung cancer (NSCLC) and<br />

adjuvant chemotherapy to early stage NSCLC. However, the role <strong>of</strong><br />

consolidation chemotherapy (CCT) after concurrent chemo-radiotherapy in<br />

locally advanced NSCLC is undetermined. Methods: We systematically<br />

searched PubMed for phase II/III trials examining survival <strong>of</strong> locallyadvanced<br />

NSCLC treated with concurrent chemo-radiotherapy between<br />

January 1, 1995 and October 31, 2011. Median overall survival (mOS) and<br />

corresponding 95% confidence interval (CI) were collected from each study<br />

and pooled. We extracted log-transformated hazards and its standard errors<br />

under the assumption that survival follows an exponential distribution, and<br />

computed a pooled mOS and its 95% CI using random-effect model.<br />

Collected trial arms were divided into two groups by the presence <strong>of</strong> CCT:<br />

Arm with CCT (CCT�) and without CCT (CCT-). Results: Forty-five studies<br />

were identified including 9 phase III studies and 36 phase II studies with<br />

51 arms (CCT�: 29, CCT-: 22). <strong>Clinical</strong> data were comparable in clinical<br />

stage, performance status, cancer histology, gender, and median age<br />

between the two groups. I2 values for assessing heterogeneity were 15.3,<br />

9.1 and 24.2% in overall, CCT� and CCT- studies, respectively. There was<br />

no statistical difference in pooled mOS between CCT� (18.5 month,<br />

95%CI: 16.7-20.5) vs CCT- (18.1 month, 95%CI: 16.5-20.2). In regard to<br />

the � grade 3 toxicities in pneumonitis, esophagitis, and neutropenia,<br />

there was no difference between the two groups throughout the whole<br />

treatment courses. In random effect models, predicted hazard ratio <strong>of</strong><br />

CCT� to CCT- was 0.98 (95%CI: 0.8-1.13, p�0.7574). These models<br />

estimated that addition <strong>of</strong> CCT could not yield more than 3 months <strong>of</strong><br />

survival prolongation for patients with locally advanced NCSLC. Conclusions:<br />

The pooled analysis on publication basis failed to provide evidence that<br />

consolidation chemotherapy yields significant survival benefit for locally<br />

advanced NSCLC.<br />

7002 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Phase II study <strong>of</strong> pemetrexed (P) plus carboplatin (Cb) or cisplatin (C) with<br />

concurrent radiation therapy followed by pemetrexed consolidation in<br />

patients (pts) with favorable-prognosis inoperable stage IIIA/B non-small<br />

cell lung cancer (NSCLC). Presenting Author: Hak Choy, University <strong>of</strong> Texas<br />

Southwestern Medical Center, Dallas, TX<br />

Background: There is no consensus chemotherapy regimen with concurrent<br />

radiation therapy (CRT) for inoperable stage IIIA/B NSCLC. P synergizes<br />

with ionizing radiation, as well as with Cb and C in preclinical models.<br />

These doublets have shown efficacy and favorable toxicity pr<strong>of</strong>iles in phase<br />

II/III trials. Methods: In this open-label randomized phase II trial, 98pts<br />

with inoperable stage IIIA/B NSCLC (all histologies) were randomized (1:1)<br />

to P 500 mg/m2 plus Cb AUC 5 (PCb) or P 500 mg/m2 plus C 75 mg/m2 (PC) intravenously (IV) every 21 days for 3 cycles. All pts received CRT<br />

64–68 Gy (2 Gy/day, 5 days/week, Days 1–45). Consolidation P 500<br />

mg/m2 IV every 21 days for 3 cycles began 3 weeks after completion <strong>of</strong><br />

CRT. The primary endpoint was 2-year overall survival (OS); secondary<br />

endpoints included median OS, time to progression (TTP), overall response<br />

rate (ORR), and toxicity. Results: Since Jun 2007, 98 pts were enrolled<br />

(PCb: 46; PC: 52).Pts were followed until Oct 2011. Mean dose compliance<br />

was PCb: 95.7% P, 97.1% Cb; PC: 89.7% P, 89.1% C. Mean dose<br />

compliance for CRT was PCb: 95.7%; PC: 88.1%. CRT dose interruptions<br />

occurred in PCb: 32.6% and PC: 40.4%. Two-year OS was PCb: 45.2%<br />

(95% confidence interval [CI], 29.3-59.8); PC: 57.6% (95% CI, 41.6-<br />

70.7); p�0.270. Median OS (months) was PCb: 18.7 (95% CI, 12.9-not<br />

assessable [N/A]); PC: 27.0 (95% CI, 23.2-N/A). Median TTP (months)<br />

was PCb: 8.8 (95% CI, 6.0-10.7); PC: 13.1 (95% CI, 8.3-N/A); p�0.057.<br />

The ORR rates were PCb: 52.2% (complete response [CR], 6.5%; partial<br />

response [PR], 45.7%); PC: 46.2% (CR, 3.8%; PR, 42.3%). Grade 4<br />

treatment-related toxicities (% PCb/% PC) were: anemia, 0/1.9; neutropenia,<br />

6.5/3.8; thrombocytopenia, 4.3/1.9; and esophagitis, 0/1.9. No<br />

drug-related deaths were reported. Conclusions: While conclusions are<br />

limited by the size <strong>of</strong> the trial, this study suggests OS and TTP advantages<br />

for the C-containing arm. Both combinations with CRT appear well<br />

tolerated.<br />

7001 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

GILT study: Oral vinorelbine (NVBo) and cisplatin (P) with concomitant radiotherapy<br />

(RT) followed by either consolidation (C) with NVBo plus P plus best<br />

supportive care (BSC) or BSC alone in stage (st) III non-small cell lung cancer<br />

(NSCLC): Final results <strong>of</strong> a phase (ph) III study. Presenting Author: Rudolf M.<br />

Huber, Medizinische Klinik Innenstadt, München, Germany<br />

Background: Concurrent chemo-radiotherapy (CT-RT) is considered as a standard<br />

in st III NSCLC. Published trials with C after CT-RT show encouraging but<br />

discordant results. This ph III was set up to assess C in st III NSCLC. Methods:<br />

Patients (pts) received NVBo 50 mg/m² D1, D8, D15 � P 20 mg/m² D1-D4 q4w /<br />

2 cycles (cy) � RT (66 Gy / 33 Fr). C for OR�SD pts: NVBo 60-80 mg/m² D1D8<br />

� P 80 mg/m² D1q3w/2cy�BSC (Arm A) or BSC (Arm B). PFS was the<br />

primary endpoint. Results: From 07/05 to 05/09, 279 pts received CT/RT and<br />

201 pts (72%) were randomised to receive CT�BSC or BSC as C. Toxicity (tox)<br />

G3-4 (% pt) CT-RT/ C (Arm A/B): anaemia 3.2/3.5/1.1; thrombopenia 2.5/1.2/<br />

0.6; neutropenia (N) 11.2/11.7/5.7; febrile N 1.4/1.0/0; nausea (G3) 5.0/4.7/<br />

2.9; vomiting (G3) 3.9/3.5/2.0; anorexia 3.6/1.2/3.0; dysphagia 1.8/2.3/1.0;<br />

fatigue 3.3/ 2.3/1.0; pneumonia/ pneumonitis 2.6/0/2.0; CT-RT pain 2.2;<br />

CT-RT oesophagitis 8.6; 3 toxic deaths. Conclusions: In this ph III, NVBo�P�RT<br />

reports a high level <strong>of</strong> efficacy (OR 60.7%; DCR 86.0%) and low tox. The DCR is<br />

significantly improved in pts who received C with NVBo� P in eval pts<br />

(p�0.0084). Lung tox was not enhanced by using NVBo�P as C. However no<br />

survival advantage for C was achieved.<br />

Randomisation (N�201)<br />

CT-RT<br />

N�242 eval /279<br />

CT�BSC<br />

N� 76 eval /96<br />

BSC<br />

N�89 eval/ 105<br />

p<br />

Male (%) 71.0 71.9 71.4<br />

Median age<br />

(y, range)<br />

60.3 [33.9-75.7] 60.3 [34.1-75.9] 59.5 [40.4-75.1]<br />

SCC/ADC* (%) 53.0 / 36.2 54.2 / 36.5 53.3 / 36.2<br />

St IIIA / IIIB (%) 17.6 / 82.4 20.8/ 79.2 19.0 / 81.0<br />

ORR ITT (%) 95% CI 55.6 [49.5-61.5] 29.2 [20.4-39.4] 24.8 [16.8-34.2] p�0.48<br />

SCC / ADC* (%)<br />

60.5 / 54.5 34.6 / 25.7 23.6 / 28.9<br />

ORR eval (%) 95% CI 60.7 [54.3-66.9] 36.8 [26.0-48.6] 29.2 [20.0-39.8] p�0.30<br />

SCC / ADC* (%)<br />

66.9 / 57.6 42.9 / 33.3 28.3 / 31.4<br />

DCR ITT (%) 78.5 66.7 56.2 p�0.12<br />

DCR eval (%) 86.0 84.2 66.3 P�0.0084<br />

Median time<br />

Registration-Rando (m)<br />

3.0 2.9<br />

Median PFS ** (m) 6.4 [5-8.7] 5.5 [3.8-7.4] p�0.63<br />

Median OS ** (m) 20.8 [13.5-25.3] 18.5 [13.6-24.7] p�0.87<br />

4 year survival ** (%) 25.3 21.4<br />

* SCC: Squamous; ADC: Adenocarcinoma. ** Calculated from the time <strong>of</strong> randomisation, after CT-RT, on ITT.<br />

7003 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

A phase III study comparing amrubicin and cisplatin (AP) with irinotecan<br />

and cisplatin (IP) for the treatment <strong>of</strong> extended-stage small cell lung cancer<br />

(ED-SCLC): JCOG0509. Presenting Author: Yoshikazu Kotani, Department<br />

<strong>of</strong> Respiratory Medicine, Kobe University Hospital, Kobe, Japan<br />

Background: IP is the standard treatment for ED-SCLC, however <strong>of</strong>ten cause<br />

severe diarrhea. AP have shown promising activity in SCLC with fewer<br />

diarrhea. We conducted a phase III trial comparing AP with IP. Methods:<br />

Eligibility criteria included patients (pts) with chemotherapy-naïve, ED-<br />

SCLC, aged 20 to 70, and ECOG PS 0-1. Pts were randomized to receive<br />

either IP or AP, balancing for site, sex, and PS. IP comprised administration<br />

<strong>of</strong> I (60 mg/m2 ) iv on days 1, 8, and 15, and P (60 mg/m2 ) iv on day1,every<br />

4 weeks. AP comprised administration <strong>of</strong> A (40 mg/m2 ) iv on day 1-3, and P<br />

(60 mg/m2 ) iv on day1, every 3 weeks. The planned sample size was 141<br />

pts in each arm with a one-sided alpha <strong>of</strong> 5% and power <strong>of</strong> 70% and a<br />

non-inferiority margin <strong>of</strong> hazard ratio (HR) as 1.31. The primary endpoint<br />

was overall survival (OS). The secondary endpoints were response rate (RR),<br />

progression-free survival (PFS), adverse events (AEs), and quality <strong>of</strong> life<br />

(QOL). We evaluated pts’ QOL twice: at the baseline and after completion <strong>of</strong><br />

the second course. Results: 284 pts were randomized to IP (n�142) and AP<br />

(n�142). Median age was 63, 84% were male, and 56% had PS 0. When<br />

191pts enrolled, more febrile neutropenia (FN) was observed in AP than<br />

anticipated, and the initial dose <strong>of</strong> A was decreased from 40 mg/m2 to 35<br />

mg/m2 . At the second interim analysis conducted after the completion <strong>of</strong><br />

patient accrual, the median OS <strong>of</strong> AP (15.0 m) was much worse than that <strong>of</strong><br />

IP (18.3 m) and the HR (1.41; 96.3% CI, 1.03-1.93) exceeds even the<br />

non-inferiority margin, so the Data and Safety Monitoring Committee<br />

recommended early publication <strong>of</strong> the results. Median PFS was 5.7 (IP) vs.<br />

5.2 months (AP) (HR 1.43, 95% CI, 1.13-1.82). RR was 69.5% (IP) vs.<br />

77.9% (AP) (p�0.14). AEs in IP and AP arm were Grade 4 neutropenia<br />

(22.5% vs. 78.6%), G3-4 FN (10.7% vs. 32.1%), and G3-4 diarrhea<br />

(7.1% vs.1.4%). Proportion <strong>of</strong> improvement in physical status <strong>of</strong> QOL was<br />

37.1%(IP) vs. 31.7%(AP), (odds ratio 0.72; 95%CI, 0.43-1.22; P�0.227).<br />

Conclusions: AP showed more bone marrow suppression than expected<br />

although it caused less diarrhea. The non-inferiority <strong>of</strong> AP to IP was not<br />

demonstrated and IP remains the standard treatment for ED-SCLC.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


Lung Cancer—Non-small Cell Local-Regional/Small Cell/Other Thoracic Cancers<br />

7004 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Phase III trial <strong>of</strong> concurrent thoracic radiotherapy (TRT) with either the first<br />

cycle or the third cycle <strong>of</strong> cisplatin and etoposide chemotherapy to<br />

determine the optimal timing <strong>of</strong> TRT for limited-disease small cell lung<br />

cancer. Presenting Author: Keunchil Park, Department <strong>of</strong> Medicine,<br />

Samsung Medical Center, Sungkyunkwan University School <strong>of</strong> Medicine,<br />

Seoul, South Korea<br />

Background: Concurrent thoracic radiotherapy (TRT) with chemotherapy<br />

has been regarded as optimal treatment for limited-disease small cell lung<br />

cancer (SCLC). However, the issue on how early TRT should be commenced<br />

is not yet defined. Methods: A total <strong>of</strong> 219 patients with limited-disease<br />

SCLC, who were enrolled from July 2003 to June 2010, received four<br />

cycles <strong>of</strong> cisplatin plus etoposide (cisplatin 70 mg/m2 on day 1 and<br />

etoposide 100 mg/m2 on days 1 to 3 every 3 weeks). We randomly assigned<br />

these patients to receive concurrent TRT, beginning with the first cycle<br />

(initial arm) or the third cycle (delayed arm) <strong>of</strong> chemotherapy. In both arms,<br />

patients received 2.1 Gy once-daily in 25 fractions over a period <strong>of</strong> five<br />

weeks, with a total dose <strong>of</strong> 52.5 Gy. Patients with partial or complete<br />

response were recommended to receive prophylactic cranial irradiation<br />

(PCI). Results: Approximately 82% <strong>of</strong> patients completed planned four<br />

cycles <strong>of</strong> chemotherapy with 52.5 Gy TRT (81.1% and 82.4% in the initial<br />

and delayed arm, respectively). After a median follow-up <strong>of</strong> 4.9 years<br />

(range, 1.2 – 8.1 years), the median overall survivals were 24.1 and 26.8<br />

months (P�0.60) in the initial and delayed arm, respectively. Progressionfree<br />

survival and complete response rates were 12.2 vs. 12.1 months<br />

(P�0.94) and 36.0% vs. 38.0% (P�0.77) in the initial and delayed arms,<br />

respectively. PCI was given to 49.5% and 55.6% <strong>of</strong> patients in the initial<br />

and delayed arms, respectively (P � 0.37). Febrile neutropenia was<br />

significantly more frequent with the initial arm, occurring in 21.6% <strong>of</strong><br />

patients, as compared with 10.2% in the delayed arm (P � 0.02). All grade<br />

esophagitis occurred in 45.0% and 37.0% <strong>of</strong> the initial and delayed arms,<br />

respectively (p � 0.23). Conclusions: TRT (52.5 Gy, once daily) beginning<br />

with the third cycle <strong>of</strong> chemotherapy showed comparable survival outcomes<br />

and complete response rates with TRT beginning with the first cycle <strong>of</strong><br />

chemotherapy, with a lower frequency <strong>of</strong> febrile neutropenia.<br />

7006 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Comprehensive genomic characterization <strong>of</strong> squamous cell carcinoma <strong>of</strong><br />

the lung. Presenting Author: Ramaswamy Govindan, Washington University<br />

School <strong>of</strong> Medicine, St. Louis, MO<br />

Background: A third <strong>of</strong> patients with non-small cell lung cancer are<br />

diagnosed with squamous cell carcinoma (SCC) histology. This report<br />

describes findings from the comprehensive genomic analyses <strong>of</strong> 178 SCC<br />

samples. Methods: The Cancer Genome Atlas (TCGA) is conducting DNA,<br />

RNA, and miRNA sequencing along with DNA copy number pr<strong>of</strong>iling,<br />

quantification <strong>of</strong> mRNA expression and promoter methylation on surgically<br />

resected samples from previously untreated patients with stage I-III SCC <strong>of</strong><br />

the lung. Results: The demographics <strong>of</strong> 178 patients enrolled in the study:<br />

median age 68 years (range: 40-85); female 47 (26%) and history <strong>of</strong><br />

tobacco smoking 171 (96%). Over 30 sites <strong>of</strong> significant somatic copy<br />

number alteration (SCNA) were identified. Exome sequencing <strong>of</strong> 178 lung<br />

SCC and matched normal samples revealed 13 significantly mutated genes<br />

with a False Discovery Rate (FDR) <strong>of</strong> �0.01 and high expression levels,<br />

including TP53, CDKN2A, PTEN, KEAP1, and NFE2L2. Apart from the<br />

near universal loss <strong>of</strong> TP53 and CDKN2A, alterations in the NFE2L2/<br />

KEAP1 and PI3K/AKT pathways were found in 35% and 43% <strong>of</strong> tumors<br />

analyzed. mRNA expression pr<strong>of</strong>iling revealed four distinct expression<br />

subtypes, each one enriched with distinct mutations and SCNAs - classical<br />

(37%): NFE2L2 and KEAP1 mutations, FGFR kinase alterations, increased<br />

global methylation and the highest rate <strong>of</strong> tobacco use; basal (24%):<br />

alterations in FGFR kinases; secretory (24%): PDGFRA alterations; primitive<br />

(15%): RB1 mutations. Rearrangements involving several known<br />

tumor suppressors were detected by whole genome shotgun sequencing <strong>of</strong><br />

20 tumor/normal pairs and confirmed by RNA sequencing including PTEN,<br />

RB1, NOTCH1, NF1 and CDKN2A. CDKN2A loss by one <strong>of</strong> several<br />

mechanisms (deletion, mutation, rearrangement with loss <strong>of</strong> function and<br />

methylation) was observed in 72% <strong>of</strong> specimens. Potential therapeutic<br />

targets for clinical trials with currently available drugs were identified in<br />

127 patients (75%). Conclusions: SCC <strong>of</strong> the lung is a distinct molecular<br />

subtype <strong>of</strong> lung cancer potentially amenable to distinct molecularly<br />

targeted therapies.<br />

7005 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

SWOG 0802: A randomized phase II trial <strong>of</strong> weekly topotecan with and without<br />

AVE0005 (aflibercept) in patients with platinum-treated extensive-stage small<br />

cell lung cancer (E-SCLC). Presenting Author: Jeffrey Warren Allen, University <strong>of</strong><br />

Tennessee Health Science Center, Memphis, TN<br />

Background: Topotecan (T) (oral and IV) is the only FDA-approved second-line<br />

chemotherapy for patients with SCLC. Weekly T is associated with less toxicity<br />

than the daily x 5 regimen. AVE0005 (aflibercept) (A) is a novel recombinant<br />

human fusion protein which binds to circulating VEGF, thereby inhibiting its<br />

interaction with cell surface receptors. We sought to evaluate the efficacy and<br />

toxicity <strong>of</strong> weekly IV T (4 mg/m2) with or without A (6 mg/kg Q21D) in patients<br />

with relapsed SCLC following one line <strong>of</strong> platinum-based chemotherapy for E or<br />

limited-stage (L) SCLC. Methods: Patients were randomized 1:1 to T or A�T.<br />

Eligible patients had adequate organ function, ECOG PS 0-1, and no recent<br />

bleeding or cardiac events. Patients with brain metastases stable for � 3 months<br />

prior to study entry were allowed. Primary endpoint was 3-month PFS. Patients<br />

were stratified as platinum-sensitive (PS) or platinum-refractory (PR). PR<br />

patients had progressed within 90 days <strong>of</strong> last chemotherapy for E and 6 months<br />

for L. This report is limited to the PR stratum. Results: 98 patients were<br />

registered. 1 was ineligible and 1 withdrew consent, leaving 96 evaluable for the<br />

primary endpoint (91 for toxicity (see Table for characteristics)). 3-month PFS<br />

was 26% for A�T versus 9% for T (P�0.01). Overall survival was similar in each<br />

arm (4.6 mos (A�T) versus 3.9 mos (T) (P�0.25)). There was 1 partial response<br />

with A�T. Disease control rate (DCR) was 28% with A�T and 12% with T.<br />

Toxicity was mainly hematologic with 14% and 19% <strong>of</strong> patients experiencing a<br />

Grade 4 event with T and A�T, respectively. There was one treatment-related<br />

death with T (renal failure). Conclusions: This study met its primary endpoint <strong>of</strong><br />

improved 3-month PFS with A�T (p�0.01), warranting further study in the PR<br />

setting. <strong>Clinical</strong> benefit with A�T was achieved primarily through improved DCR.<br />

Toxicity was manageable and less than is seen with standard dose T. Accrual to<br />

the PS cohort is ongoing.<br />

Patient characteristics.<br />

T(n�46) A�T(n�50)<br />

Age (median) 63 60<br />

Gender<br />

Male (%)<br />

Female (%)<br />

Race<br />

Caucasian (%)<br />

Other (%)<br />

PS<br />

0(%)<br />

1(%)<br />

Stage<br />

Extensive (%)<br />

Limited (%)<br />

67<br />

33<br />

83<br />

17<br />

39<br />

61<br />

70<br />

30<br />

7007 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Prognostic and predictive effects <strong>of</strong> KRAS mutation subtype in completely<br />

resected non-small cell lung cancer (NSCLC): A LACE-bio study. Presenting<br />

Author: Frances A. Shepherd, Princess Margaret Hospital, University <strong>of</strong><br />

Toronto, Toronto, ON, Canada<br />

Background: We reported previously that KRAS is weakly prognostic and not<br />

significantly predictive <strong>of</strong> benefit from adjuvant chemotherapy (ACT) in<br />

NSCLC (Tsao et al. Proc ESMO 2012, Abst 4217). In colorectal cancer<br />

(CRC), KRAS mutation predicts resistance to EGFR monoclonal antibodies,<br />

but recent reports suggest that this may not be true for all mutation<br />

subtypes (De Roock et al. JAMA 2010). Smoking-related KRAS mutations<br />

in NSCLC differ from those <strong>of</strong> CRC. To explore the influence <strong>of</strong> KRAS<br />

mutation subtype, we undertook an analysis <strong>of</strong> KRAS subtype in LACE-Bio.<br />

Methods: KRAS mutation was determined in blinded fashion in 3 laboratories<br />

by direct sequencing. Exploratory analyses were performed to identify<br />

relationships between mutation status and subtype, overall (OS) and<br />

disease-free survival (DFS) using a Cox model stratified by trial and<br />

adjusted for covariates. Results: KRAS subtype was available in 1,532<br />

patients (756 observation [OBS], 776 ACT). There were 300 mutations<br />

(275 codon 12, 24 codon 13, 1 codon 14). In the OBS arm, there was no<br />

difference in prognosis for OS for codon 12 (Hazard Ratio [HR] mutation v<br />

wild-type [WT] KRAS 1.04, CI .77-1.4) or codon 13 (HR 1.01, CI<br />

0.47-2.17) mutations. A trend for benefit from ACT was observed in WT<br />

(HR ACT v OBS 0.89 CI 0.76-1.04, p�0.15) but not in patients with codon<br />

12 mutations (HR 0.95, CI .67-1.35, p�0.77). In patients with codon 13<br />

mutations, chemotherapy was deleterious (HR 5.78, CI 2.06-16.2,<br />

p�0.001), test for equality among 3 HRs p�0.002. Results were similar<br />

for DFS. Among codon 12 mutations, there was no prognostic effect based<br />

on specific amino acid substitution. Patients with G12A or G12R mutations<br />

appeared to derive greater benefit from ACT (HR 0.66 p�0.48) compared<br />

to those with G12C or G12V (HR 0.94 p�0.77) or G12D or G12S (HR 1.39<br />

p�0.48) or WT, but the differences were not significant (comparison <strong>of</strong> 4<br />

HRs p�0.76). Conclusions: In this study, patients with KRAS codon 13<br />

mutations had significantly poorer outcomes with ACT. These results<br />

require further confirmation and should be interpreted with caution in view<br />

<strong>of</strong> the small number <strong>of</strong> patients with codon 13 mutations. Supported by<br />

unrestricted grants from San<strong>of</strong>i Aventis and LNCC.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.<br />

453s<br />

48<br />

52<br />

94<br />

6<br />

22<br />

78<br />

83<br />

17


454s Lung Cancer—Non-small Cell Local-Regional/Small Cell/Other Thoracic Cancers<br />

7008 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Accuracy <strong>of</strong> FDG-PET to diagnose lung cancer in the ACOSOG Z4031 trial.<br />

Presenting Author: Eric L Grogan, Vanderbilt University Medical Center,<br />

Nashville, TN<br />

Background: Fluoro-deoxyglucose positron emission tomography (FDG-PET)<br />

is recommended for diagnosis and staging <strong>of</strong> known or suspected NSCLC.<br />

Meta-analyses examining the accuracy <strong>of</strong> FDG-PET to diagnose lung cancer<br />

demonstrated high sensitivity 94% and specificity 83% but were performed<br />

in select centers. The purpose <strong>of</strong> this study is to evaluate the<br />

accuracy <strong>of</strong> FDG-PET to diagnose NSCLC and examine enrolling site<br />

differences in the national prospective ACOSOG Z4031 trial. Methods:<br />

1,074 patients with clinical stage I (cT1-2N0M0) known or suspected<br />

NSCLC were enrolled between 2004 and 2006 in the Z4031 study and<br />

underwent surgical resection. The final diagnosis was determined by<br />

pathological examination. FDG-PET results were abstracted from radiology<br />

interpretations included in the case report forms. FDG-PET avidity was<br />

categorized based on either radiologist description or reported maximum<br />

standard uptake value (SUV). The four categories were: not avid and not<br />

cancerous (SUV�0), low avidity and likely not cancerous (SUV�0 and<br />

�2.5), avid and probably cancerous (SUV�2.5 and �5) and highly avid<br />

and likely cancerous (SUV�5). Sensitivity analysis <strong>of</strong> FDG-PET diagnostic<br />

accuracy was performed for varying levels <strong>of</strong> avidity and by preoperative<br />

lesion size. Differences in accuracy by enrolling site were examined.<br />

Results: There were 51 enrolling sites in 39 cities with 969 eligible<br />

participants. Preoperative FDG-PET results were available for 682 participants.<br />

Lung cancer prevalence was 83%. FDG-PET sensitivity was 82%<br />

(95% CI: 79-85), and specificity was 31% (95% CI: 23-40). Positive and<br />

negative predictive values were 85% and 26%, respectively and accuracy<br />

improved with lesion size. There were 80 false positive scans and 69% were<br />

granulomas. False negative scans occurred in 101 patients (11were<br />

�10mm) with adenocarcinoma, squamous, bronchoalveolar cell and neuroendocrine<br />

tumors responsible for 64%, 12%, 10% and 8% <strong>of</strong> false<br />

negative results, respectively. Specificity did not differ between the 8 sites<br />

with �25 patients (p�0.74). Conclusions: In a national surgical population<br />

with clinical stage I NSCLC, FDG-PET to diagnose lung cancer performed<br />

poorly compared to published studies. Reasons for poor test performance<br />

should be explored.<br />

7010 Poster Discussion Session (Board #2), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

The SELECT study: A multicenter phase II trial <strong>of</strong> adjuvant erlotinib in<br />

resected epidermal growth factor receptor (EGFR) mutation-positive nonsmall<br />

cell lung cancer (NSCLC). Presenting Author: Joel W. Neal, Stanford<br />

University, Palo Alto, CA<br />

Background: Cancers with activating EGFR mutations are exquisitely<br />

sensitive to EGFR tyrosine kinase inhibitors (TKIs) and retrospective data<br />

suggests adjuvant TKIs may improve outcomes in EGFR mutants. This<br />

prospective trial investigates the safety and efficacy <strong>of</strong> adjuvant erlotinib in<br />

EGFR mutation-positive NSCLC. Methods: Patients (pts) with surgically<br />

resected stage IA-IIIA NSCLC harboring activating EGFR mutations were<br />

treated with 150 mg/day <strong>of</strong> erlotinib for 2 years (y) after completion <strong>of</strong> any<br />

standard adjuvant chemotherapy and/or radiotherapy. The trial was designed<br />

to enroll 36 patients, and powered to demonstrate a primary<br />

endpoint <strong>of</strong> 2 y disease free survival (DFS) exceeding 85%, which would<br />

suggest improvement over the historically expected 70% 2 y DFS in early<br />

stage EGFR-mutant NSCLC (J Thorac Oncol 6:569). Results: Thirty-six pts<br />

were enrolled at five sites between 1/08 and 11/09; 53% stage I; 19%<br />

stage II; 28% stage IIIA. Toxicities were typical <strong>of</strong> erlotinib; no grade 4 or 5<br />

events or pneumonitis occurred. 8 pts (22%) required one dose reduction<br />

to 100 mg/day and 5 (14%) two reductions to 50 mg/day for grade 3 or<br />

persistent grade 2 toxicities. 11 pts discontinued before 2 full years (�1<br />

month (mo) [4], 1-12 mo [2] and 12-23 mo [5]) for toxicities [6], patient<br />

preference [3], prostate cancer [1] and recurrence [1]. After a median<br />

follow-up <strong>of</strong> 2.5 y, the 2 y DFS from enrollment is 94% (95% CI 80%,<br />

99%). 10 patients have recurred, 1 during erlotinib treatment and the<br />

others after stopping erlotinib (interval before recurrence 2 mo [1], 6-12<br />

mo [4], �12 mo [4]). Genotyping on repeat biopsies from seven <strong>of</strong> the<br />

recurrent cases is underway, as is assessment <strong>of</strong> response to subsequent<br />

erlotinib therapy. Two pts have died <strong>of</strong> recurrence: one at 1.5 y who stopped<br />

erlotinib after 1 mo for toxicity, and one at 2 y who progressed while on<br />

erlotinib. Conclusions: This is the first prospective study to report the<br />

efficacy <strong>of</strong> adjuvant erlotinib in NSCLC pts with EGFR mutations. This<br />

approach is feasible and yields excellent 2y DFS compared to historical<br />

genotype-matched controls. This trial was subsequently expanded to 100<br />

pts to permit subgroup analysis by stage.<br />

7009 Poster Discussion Session (Board #1), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Stages I-II non-small cell lung cancer treated using either lobectomy by<br />

video-assisted thoracoscopic surgery (VATS) or stereotactic ablative radiotherapy<br />

(SABR): Outcomes <strong>of</strong> a propensity score-matched analysis. Presenting<br />

Author: Suresh Senan, Department <strong>of</strong> Radiation Oncology, VU University<br />

Medical Center, Amsterdam, Netherlands<br />

Background: VATS procedures are increasingly used in early-stage NSCLC.<br />

As high local control rates are also seen with stereotactic ablative<br />

radiotherapy (SABR), we performed a propensity score-matched analysistocompare<br />

loco-regional control (LRC) after both treatments. Methods:<br />

Patients with stage I-II NSCLC treated at 6 hospitals (1 university and 5<br />

regional hospitals) with VATS lobectomy were eligible. Details <strong>of</strong> SABR<br />

patients were obtained from a single-institutional database. All VATSlobectomies<br />

were performed in accordance with ESTS guidelines. Patients<br />

were matched using propensity scores based on cTNM, age, gender,<br />

Charlson comorbidity score, lung function and performance score. Matching<br />

was performed blinded to all outcomes. Excluded were: synchronous<br />

lung tumors, COPD GOLD class 4 or history <strong>of</strong> prior lung cancer. A total <strong>of</strong><br />

86 VATS- and 527 SABR patients were eligible for matching (1:1 ratio,<br />

caliper distance <strong>of</strong> 0.025 without replacement). Loco-regional failure was<br />

defined as recurrence in/adjacent to the radiation planning target volume or<br />

surgical margins, the ipsilateral hilum or mediastinum. Recurrences were<br />

either biopsy-confirmed or PET-positive and reviewed by a tumor board.<br />

Patients upstaged during VATS and those developing recurrence were<br />

treated in accordance with national guidelines. Results: The matched<br />

cohort consisted <strong>of</strong> 128 patients with cT1-3N0 NSCLC following SABR<br />

(n�64) or VATS-lobectomy (n�64). Median follow-up was 30 and 16<br />

months, respectively. The groups were well matched on baseline variables.<br />

SABR patients had better LRC rates at 1- and 3-years (96.8% and 93.3%<br />

vs. 86.9% and 82.6%, respectively, p� .03). Three-year progression-free<br />

survival (PFS) did not significantly differ after SABR (79.3% versus<br />

63.2%, p � .09). Distant recurrence rates and overall survival (OS) did not<br />

significantly differ. Conclusions: Although loco-regional control was superior<br />

after SABR compared to VATS-lobectomy, PFS and OS did not differ at<br />

this time-point. Our findings support the current randomized controlled<br />

trial evaluating both treatments (ACOSOG Z4099).<br />

7011 Poster Discussion Session (Board #3), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Pilot SCAT trial: Spanish customized adjuvant chemotherapy (CT) based on<br />

BRCA1 mRNA expression levels (l) in resected stage II-IIIA non-small cell<br />

lung cancer (NSCLC) patients (p). Presenting Author: Jose Miguel Sanchez,<br />

Hospital M. D. Anderson Internacional, Madrid, Spain<br />

Background: Surgical resection is the standard treatment in early stages <strong>of</strong><br />

NSCLC. Nonetheless, long-term survival rate even after surgical resection<br />

is disappointing. Several randomized trials and meta-analyses confirmed<br />

the survival benefit <strong>of</strong> adjuvant cisplatin-based CT for stage II and IIIA.<br />

BRCA1 is a component <strong>of</strong> multiple DNA repair pathways, functions as a<br />

molecular determinant <strong>of</strong> response to cytotoxic chemotherapeutics agents,<br />

and is an independent prognostic variable in resected p. Since BRCA1<br />

mRNA expression has been linked to differential sensitivity to cisplatin and<br />

antimicrotubule drugs, BRCA1 expression may provide additional information<br />

for customizing adjuvant CT. Methods: Completely resected p with<br />

pathological lymph node involvement (N1, N2) received 4 cycles <strong>of</strong><br />

customized adjuvant CT according BRCA1 l. Low l: gemcitabine-cisplatin,<br />

intermediate l: docetaxel-cisplatin, high l: docetaxel. Overall survival (OS)<br />

was the primary endpoint. Multivariate analysis for time to relapse (TTR)<br />

and survival was performed. Results: Eighty-three p were elegible. Most <strong>of</strong><br />

the patients were male (83%). Type <strong>of</strong> surgery: 23% pneumonectomy, and<br />

77% lobectomy or bilobectomy. Fifty-three per cent had low BRCA1 l,<br />

33,7% intermediate l, and 13,3% expressed high l. Most <strong>of</strong> the tumors<br />

with adenocarcinoma histology had low l (71%). With a median follow-up <strong>of</strong><br />

41.6 months (m), median TTP for the whole group was 22.9 m (13.4-<br />

32.5): 20.3 m for low l, 56.5 m for intermediate l, and 51.9 m for high l<br />

(P�0.31). Median OS for the whole group was 63.6 m (33.3-93.9): 55 m<br />

for low l, and not reached for intermediate and high l (P�0.58). Forty-three<br />

p (51.8%) are still alive. Conclusions: Selection <strong>of</strong> customized CT based on<br />

BRCA1 expression l is feasible and could increase time to relapse and<br />

survival in resected N1-N2 p. No statistical differences for survival: 55<br />

months for gemcitabine-cisplatin, and not reached for docetaxel-cisplatin<br />

and for docetaxel as single agent. Randomized phase III SCAT trial with the<br />

addition <strong>of</strong> a non-pharmacogenomic control arm is ongoing (372 patients<br />

included).<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


Lung Cancer—Non-small Cell Local-Regional/Small Cell/Other Thoracic Cancers<br />

7012 Poster Discussion Session (Board #4), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Feasibility trial <strong>of</strong> adjuvant chemotherapy with docetaxel (DOC) plus<br />

cisplatin (CDDP) followed by maintenance chemotherapy <strong>of</strong> S-1 in completely<br />

resected non-small cell lung cancer (NSCLC): Thoracic Oncology<br />

Research Group (TORG) 0809. Presenting Author: Seiji Niho, Division <strong>of</strong><br />

Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Chiba,<br />

Japan<br />

Background: Maintenance chemotherapy could prolong overall and/or<br />

progression-free survival in advanced NSCLC. S-1 is an oral anticancer<br />

agent comprised <strong>of</strong> tegafur, 5-chloro-2, 4-dihydroxypyridine, and potassium<br />

oxonate. The TORG 0809 study was conducted to evaluate the<br />

feasibility and efficacy <strong>of</strong> maintenance chemotherapy <strong>of</strong> S-1 following<br />

DOC�CDDP in patients (pts) with curatively resected stage II and IIIA<br />

NSCLC. Methods: Pts received 3 cycles <strong>of</strong> DOC (60mg/m2 d1) plus CDDP<br />

(80mg/m2 d1), q3-4w, and subsequently S-1 at 40mg/m2 twice a day for<br />

14 consecutive days, q3w, for more than 6 months (max 1 year). The<br />

primary endpoint was determination <strong>of</strong> feasibility, which was defined as the<br />

proportion <strong>of</strong> pts who had completed maintenance for 8 cycles <strong>of</strong> S-1 or<br />

more. If the lower confidence interval (CI) <strong>of</strong> this proportion was 50% or<br />

more, the feasibility <strong>of</strong> the treatment was considered confirmed. The<br />

sample size was set to be 125. Results: Between Jun 2009 and Nov 2010,<br />

131 pts were enrolled, <strong>of</strong> whom 129 pts were eligible and assessable. The<br />

median age was 63 (23-74 years); PS 0: 107, 1: 22; p-stage IIA: 19, IIB:<br />

30, IIIA: 80; adenocarcinoma: 99, non-adenocarcinoma: 30. Of 129 pts,<br />

109 pts (84.5%) completed 3 cycles <strong>of</strong> DOC�CDDP. 106 pts initiated the<br />

maintenance S-1 and 66 pts (51.2%, 95% CI: 42.5-59.8) completed 8<br />

cycles or more S-1 treatment; this percentage was less than our previously<br />

defined criterion for treatment feasibility, since 32 pts terminated maintenance<br />

S-1 at 3 cycles or less. Grade 3/4 toxicities during the maintenance<br />

S-1 included anemia (7.3%), neutropenia (3.7%), anorexia (3.7%),<br />

dyspnea (1.8%), infection with neutropenia <strong>of</strong> grade 0 to 2 (1.8%). Febrile<br />

neutropenia was not observed. One pt developed interstitial pneumonia and<br />

sepsis, resulting in treatment-related death. Recurrence-free survival data<br />

will be presented. Conclusions: The toxicity level was acceptable, although<br />

the results did not meet our criterion for feasibility. Modification <strong>of</strong> the<br />

treatment schedule <strong>of</strong> maintenance S-1, such as a 2-week rest period<br />

rather than 1-week, might improve treatment compliance.<br />

7014 Poster Discussion Session (Board #6), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Quantitating pathologic response to neoadjuvant chemotherapy in KRASmutated<br />

versus nonmutated lung cancers. Presenting Author: Jamie E.<br />

Chaft, Memorial Sloan-Kettering Cancer Center, New York, NY<br />

Background: Trials <strong>of</strong> neoadjuvant chemotherapy in non-small cell lung<br />

cancers (NSCLC) have demonstrated that pathologic response correlates<br />

with downstaging and survival. There is controversy as to whether KRAS<br />

mutated patients (pts) have the same benefit with adjuvant chemotherapy<br />

as non-mutated pts however the impact <strong>of</strong> KRAS mutations on response to<br />

neoadjuvant therapy has not been reported. Methods: Pts with resectable<br />

stage I-III non-squamous NSCLCs were treated with 4 cycles <strong>of</strong> cisplatin,<br />

docetaxel, and bevacizumab, followed by surgical resection. Tumors were<br />

tested for KRAS mutations. Percent treatment effect (necrosis, inflammation,<br />

fibrosis) was quantified histologically. Pts were followed until disease<br />

recurrence and/or death. Binary variables were compared using the Fisher’s<br />

Exact Test. Survival was assessed by pathological response based on the<br />

literature (�90 vs �90%) using the Kaplan-Meier method, stratified by<br />

stage (IB-IIB vs. III) and by KRAS mutation. Results: We accrued 51 pts.<br />

34/51 (67%) had clinical stage IIIA disease. 48 had molecular testing:<br />

14/48 (29%) had KRAS mutations and 4/48 (8%) had EGFR mutations.<br />

The median follow-up was 2 years (yr) (range, 3-63 months). 41 pts had<br />

resection and analysis for pathological response. The 2 yr disease free<br />

survival (DFS) and overall survival (OS) were superior in the 11/41 (27%)<br />

with �90% versus those with �90% treatment effect: DFS 91% vs. 56%,<br />

p�0.029 and OS 100% vs. 60%, p�0.013. These outcomes remained<br />

significant when adjusted for stage. For pts with KRAS mutations, 0 <strong>of</strong> 10<br />

tumors analyzed had �90% treatment effect whereas 11 <strong>of</strong> 31 (35%) pts<br />

with wild-type KRAS had �90%, p�0.039. There was an inferior median<br />

OS for KRAS mutated pts (22 months) compared to pts without a known<br />

KRAS mutation, (Not Reached, p�0.03). Conclusions: Following neoadjuvant<br />

chemotherapy, pts with tumors having �90% necrosis had improved<br />

survival. There were no patients with KRAS mutation that achieved �90%<br />

treatment effect in response to cisplatin, docetaxel, and bevacizumab.<br />

Adaptive adjuvant trial strategies to improve upon outcomes <strong>of</strong> those with<br />

�90% necrosis at resection and new approaches specifically targeting<br />

KRAS-mutated NSCLCs are needed.<br />

7013 Poster Discussion Session (Board #5), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

MAGE-A3 cancer immunotherapeutic in resected stage IB-III NSCLC<br />

patients with or without sequential or concurrent chemotherapy. Presenting<br />

Author: Jean-Louis Pujol, CHU - Maladies Respiratoires, Montpellier,<br />

France<br />

Background: Previous trials with the MAGE-A3 recombinant (rec) protein<br />

formulated with an immunostimulant have shown induction <strong>of</strong> specific<br />

immune responses and signals <strong>of</strong> clinical activity in different cancer<br />

diseases. In this phase I/II study (NCT 00455572), we evaluated the safety<br />

pr<strong>of</strong>ile <strong>of</strong> the MAGE-A3 cancer immunotherapeutic (CI), formulated with<br />

the rec MAGE-A3 protein and the AS15 immunostimulant, and the<br />

induction <strong>of</strong> specific immune response with or without adjuvant chemotherapy<br />

(CT). Methods: MAGE-A3 CI was administered i.m. 8q3w in<br />

resected MAGE-A3� stage IB-III NSCLC patients (pts). Three cohorts (C)<br />

were evaluated: Immunization with concurrent cisplatin plus vinorelbine<br />

(C1), sequentially after the same CT (C2) or with no CT (C3). The<br />

anti-MAGE-A3 humoral and cellular immune responses were evaluated by<br />

ELISA and intracellular cytokine staining (T cells producing both IFNg and<br />

TNF) respectively. Adverse Events (AEs) were graded according to CTCAE v.<br />

3.0. Results: A total <strong>of</strong> 38/55 treated pts received the 8 doses schedule<br />

(15/19 in C1, 14/18 in C2, 9/18 in C3). Almost all pts reported at least one<br />

AE, mostly general constitutional disorders and administration site reactions.<br />

Six patients reported related grade 3 AEs. No related grade 4-5 AE or<br />

related SAE were reported. Immunogenicity results in the total treated<br />

population are reported in the table below. Conclusions: In this trial,<br />

patients in cohorts 2 and 3 correspond to the two populations enrolled in<br />

the Phase III MAGRIT trial evaluating the same MAGE-A3 CI. The phase I/II<br />

results suggest that this CI is well tolerated and induces in all treated pts<br />

specific antibodies against MAGE-A3 after 4 doses in presence or not <strong>of</strong><br />

concurrent or sequential adjuvant CT. About 25% <strong>of</strong> the treated pts are<br />

considered as CD4 responders in presence or not <strong>of</strong> concurrent or<br />

sequential adjuvant CT.<br />

C1<br />

n/N<br />

C2<br />

n/N<br />

455s<br />

C3<br />

n/N<br />

Cohort<br />

Seropositivity*<br />

-<br />

-<br />

-<br />

After 4 doses<br />

15/15<br />

15/15<br />

12/12<br />

After 8 doses<br />

15/15<br />

13/13<br />

9/9<br />

CD4 response** 4/11 4/15 2/8<br />

CD8 response** 1/11 1/15 0/8<br />

n: number <strong>of</strong> seropositive/responding patients; N: Total number <strong>of</strong> pts with<br />

available samples; *: Anti-MAGE-A3-Antibodies; **: Responders are pts with a<br />

response ratio baseline/any timepoint ��4 folds.<br />

7017 Poster Discussion Session (Board #9), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Updated results <strong>of</strong> a phase III trial comparing standard thoracic radiotherapy<br />

(RT) with or without concurrent daily low-dose carboplatin in<br />

elderly patients (pts) with locally advanced non-small cell lung cancer<br />

(NSCLC): JCOG0301. Presenting Author: Hiroaki Okamoto, Yokohama<br />

Municipal Citizen’s Hospital, Yokohama, Japan<br />

Background: We previously reported the superiority <strong>of</strong> combined chemoradiotherapy<br />

(CRT) over RT alone in elderly pts with locally advanced<br />

NSCLC (Atagi et al. ECCO2011). One and a half years follow-up data from<br />

last accrual are presented. Methods: Pts older than 70 years with unresectable<br />

stage III NSCLC were randomized to either RT alone (RT arm), a total<br />

dose <strong>of</strong> 60 Gy, or CRT arm including the same RT plus concurrent<br />

chemotherapy with carboplatin 30 mg/m2 /day, 5 days/week � 20 days. The<br />

primary endpoint was overall survival (OS). The planned sample size was<br />

100 pts in each arm with one-sided alpha <strong>of</strong> 5% and 80% power to detect a<br />

difference in median survival time (MST) from 10 months in RT arm to 15<br />

months in CRT arm. Results: Between Sep 2003 and May 2010, 200 pts<br />

were randomized. Baseline characteristics were similar in the RT (n�100)<br />

vs CRT (n�100) arms: median age, 77 vs 77 years; stage IIIB (n), 46 vs<br />

49; PS 0/1/2 (n), 41/55/4 vs 41/56/3. The second planned interim analysis<br />

was performed 10 months after the completion <strong>of</strong> accrual. In accordance<br />

with the pre-specified stopping rule, the JCOG Data and Safety Monitoring<br />

Committee recommended early publication <strong>of</strong> this trial because <strong>of</strong> the<br />

difference in OS favoring the CRT arm. In the updated analysis, OS was<br />

better in the CRT arm than the RT arm (HR � .64, 95% CI � .46-.89,<br />

one-sided p � .0033 by stratified log-rank test). In each arm (RT/CRT),<br />

MST was 16.5 mo/22.4 mo with 3-year OS <strong>of</strong> 14.3%/34.6%, response rate<br />

<strong>of</strong> 44.9%/54.6% (p�.201) and median progression-free survival <strong>of</strong> 6.9<br />

mo/8.9 mo (p�.003). Gr 3/4 toxicities were (RT/CRT): neutropenia<br />

0%/57.3%, infection 4.1%/12.5%, dysphagia 0%/1.0%, late RT toxicities<br />

7.4%/7.5%. The pattern <strong>of</strong> relapse site and post-protocol treatment were<br />

almost similar between the arms. Even after an adjustment by the Cox<br />

regression analysis with six variables [stage, PS, sex, age, histology,<br />

smoking status], CRT arm showed better survival (HR�.71, p�.038).<br />

Conclusions: The CRT using daily carboplatin is considered to be the<br />

standard treatment for elderly pts with locally advanced NSCLC.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


456s Lung Cancer—Non-small Cell Local-Regional/Small Cell/Other Thoracic Cancers<br />

7018 Poster Discussion Session (Board #10), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

SWOG S0533: A pilot trial <strong>of</strong> cisplatin (C)/etoposide (E)/radiotherapy (RT)<br />

followed by consolidation docetaxel (D) and bevacizumab (B) (NSC-<br />

704865) in three cohorts <strong>of</strong> patients (pts) with inoperable locally advanced<br />

stage III non-small cell lung cancer (NSCLC). Presenting Author: Antoinette<br />

J. Wozniak, Karmanos Cancer Institute, Wayne State University,<br />

Detroit, MI<br />

Background: Bevacizumab combined with chemotherapy has improved<br />

survival in the treatment <strong>of</strong> advanced NSCLC. This pilot trial was conducted<br />

to determine if bevacizumab could be incorporated into the standard<br />

chemotherapy/RT for locally advanced NSCLC. Methods: Pts with unresectable<br />

stage III NSCLC, PS 0-1, and adequate organ function were eligible.<br />

Pts were accrued in two strata, low and high risk (squamous histology,<br />

hemoptysis, tumor with cavitation or near a major vessel). Pts were treated<br />

with C 50mg/m2 (d 1 and 8), E 50mg/m2 (d 1-5) for 2 cycles concurrent<br />

with RT (64.8 Gy at 1.8 Gy/fx for 36 fxs) followed by consolidation D<br />

75mg/m 2<br />

and B 15 mg/kg for 3 cycles (Cohort 1). If safety was established<br />

then accrual would continue to Cohort 2 (B, d 15, 36, 57) and then Cohort<br />

3 (B d 1, 22, 43). Results: 29 pts (17 Low, 12 High) were registered, all to<br />

Cohort 1. 26 pts (stage IIIB 19 pts, squamous 4 pt, adenosquamous 1 pt)<br />

were evaluable. 25 completed chemo/RT. Grade 3/4 toxicities during<br />

chemo/RT included: neutropenia 10 pts, thrombocytopenia 2, anemia 2,<br />

febrile neutropenia 3, esophagitis 2, pneumonitis 1. 21 were assessed for<br />

safety with D/B consolidation. The major adverse events during D/B<br />

consolidation were pneumonitis (Gr 3-2pt)and2episodes <strong>of</strong> fatal<br />

hemoptysis in the high risk group resulting in closure <strong>of</strong> this stratum. The<br />

low risk stratum subsequently closed because <strong>of</strong> poor accrual. Median<br />

overall survival was 23 mos for low risk pts and 17 mos for the high risk<br />

stratum. Conclusions: In this trial, bevacizumab could not be successfully<br />

integrated into chemo-radiation for stage III NSCLC, particularly in pts<br />

considered at high risk for hemoptysis. The trial suffered from several<br />

temporary closures as mandated by CTEP when new bevacizumab-related<br />

toxicities were reported, contributing to slow accrual. In lower risk pts the<br />

data are insufficient to determine safety or efficacy. Supported in part by<br />

the following PHS Cooperative Agreement grant numbers awarded by the<br />

National Cancer Institute, DHHS: CA32102 and CA38926.<br />

7020 Poster Discussion Session (Board #12), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Favorable outcomes with chemoradiation and surgery for locally advanced<br />

non-small cell lung cancer: The BC Cancer Agency Vancouver experience.<br />

Presenting Author: Wen Wen Shan, University <strong>of</strong> British Columbia, Vancouver,<br />

BC, Canada<br />

Background: The role <strong>of</strong> surgery following concurrent platinum-based<br />

chemotherapy and radiation for locally advanced non-small cell lung<br />

cancer (NSCLC) remains controversial, with high surgical mortality rates<br />

reported in a large randomized clinical trial. In this retrospective study, we<br />

evaluated the safety and efficacy <strong>of</strong> concurrent chemoradiation with or<br />

without surgery over an 11 period at the BC Cancer Agency. Methods:<br />

Patients were identified by the Vancouver Centre Pharmacy database.<br />

Charts were reviewed and data extracted included patient characteristics,<br />

weight loss, performance status, and method <strong>of</strong> mediastinal staging.<br />

Outcome measures were overall survival, pathological response rate, and<br />

treatment-associated morbidity and mortality. Results: Between January<br />

1999-2010, 177 patients were identified with locally advanced NSCLC<br />

(stage IIIA/B) treated with platinum and etoposide and �40Gy radiation<br />

therapy, with or without surgical resection. The majority <strong>of</strong> treatment plans<br />

were reached by a multidisciplinary conference consensus. 74% (n�131)<br />

<strong>of</strong> patients received chemoradiation alone (bimodality therapy) and 36%<br />

(n�46) received chemoradiation followed by surgical resection (trimodality<br />

therapy). Among the trimodality therapy group, 16 patients underwent<br />

pneumonectomy and 30 lobectomy. Conclusions: In this series, bimodality<br />

therapy for patients with locally advanced NSCLC had similar treatment<br />

associated mortality and survival outcomes as reported in the literature.<br />

Trimodality therapy was associated with low treatment mortality rates and<br />

favourable survival. These two groups cannot be directly compared in this<br />

retrospective study. However, these results support a multidisciplinary<br />

approach to identify and carefully select patients with locally advanced<br />

NSCLC to undergo additional surgical resection following concurrent<br />

chemoradiation.<br />

Treatment-associated complications and survival.<br />

Bimodality Trimodality<br />

Complications<br />

Hospitalization 24% 11%<br />

Death on treatment 5% 2%<br />

Survival<br />

Median OS 19.6 mo 68 mo<br />

5-year OS 17.5% 53.2%<br />

7019 Poster Discussion Session (Board #11), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

A randomized, multicenter phase II study investigating additional weekly<br />

cetuximab to concurrent chemoradiotherapy in locally advanced non-small cell<br />

lung carcinoma: Reporting on the efficacy. Presenting Author: Michel M. van den<br />

Heuvel, Medical Oncology Department, The Netherlands Cancer Institute/Antoni<br />

van Leeuwenhoek Ziekenhuis, Amsterdam, Netherlands<br />

Background: Modest benefits from concurrent chemoradiotherapy (CRT) in<br />

patients with locally advanced NSCLC warrant more effective treatment regimen.<br />

Cetuximab, a monoclonal antibody against the epidermal growth factor receptor<br />

has shown activity in NSCLC. Feasibility data and toxicity have been published<br />

previously. We report treatment outcome <strong>of</strong> a multicenter phase II study <strong>of</strong> the<br />

combination <strong>of</strong> high dose accelerated RT and daily dose cisplatin with or without<br />

weekly cetuximab. Methods: Patients with locally advanced NSCLC received<br />

accelerated RT (66 Gy in 24 fractions) and concurrent daily cisplatin (6 mg/m2 )<br />

with (Arm A) or without (Arm B) additional weekly cetuximab (400 mg/m2 loading dose one week prior to the RT start followed by weekly 250 mg/m2 ). The<br />

Objective Local Response Control (OLRC) was determined at 6 and 24 weeks<br />

after treatment using response evaluation criteria in solid tumours criteria.<br />

Results: Between Feb 2009 and May 2011, 102 patients were included. Median<br />

follow-up was 13 months. Patients and tumor characteristics are shows in the<br />

Table. Stage distribution was: II (8%), IIIa (51%), and IIIb (40%). The CRT was<br />

well tolerated. The OLRC at 24 weeks was 79% in Arm A and 80% in Arm B. The<br />

one-year progression free survival and overall survival were 58% (45%-76%) and<br />

76% (64%-91%) for Arm A and 49% (35%-68%) and 72% (58%-89%) for Arm<br />

B respectively. Conclusions: The addition <strong>of</strong> cetuximab to low dose cisplation<br />

CRT does not improve OLRC in an unselected patient cohort but data on<br />

longterm disease control and survival are to be awaited.<br />

Characteristics <strong>of</strong> patients treated with concurrent chemoradiotherapy.<br />

CRT CRT � cetuximab<br />

Age (years, range) 63 (37-78) 62 (29-80)<br />

Female / Male 29% / 71% 33% / 67 %<br />

Ethnic origin (Asian/non-Asian) 2%/98% 6%/94%<br />

Staging:<br />

4%<br />

12%<br />

-IIa/b<br />

54%<br />

47%<br />

-IIIa<br />

-IIIb<br />

41%<br />

41%<br />

Histology:<br />

32%<br />

33%<br />

-Adeno<br />

28%<br />

24%<br />

-Large cell<br />

38%<br />

41%<br />

-Squamous<br />

-Other<br />

2%<br />

2%<br />

Smoking history<br />

3%/49%/49%<br />

3%/37%/61%<br />

(Never/former/current)<br />

42%/56%/2%<br />

39%/61%/0%<br />

PS 0/1/2<br />

FEV1 (%)<br />

87% (39%-151%)<br />

77% (40%-121%)<br />

GTV (cm3 )<br />

PTV (cm 3 )<br />

107 (25-907)<br />

547 (66-1766)<br />

120 (5-460)<br />

451 (49-1855)<br />

SUVmax 13.7 (6.3-35.9) 10.9 (6.2-31.8)<br />

7021 Poster Discussion Session (Board #13), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Comparison between diameters <strong>of</strong> the whole nodule and solid area and the<br />

proportion <strong>of</strong> GGO in the tumor shadow on HRCT in predicting less-invasive<br />

lung cancer and recurrence after complete resection in patients with<br />

clinical N0 NSCLC. Presenting Author: Haruhisa Matsuguma, Division <strong>of</strong><br />

Thoracic Surgery, Tochigi Cancer Center, Utsunomiya, Japan<br />

Background: Prediction <strong>of</strong> less-invasive lung cancer is important in selecting<br />

candidates for limited surgical resection. A greater proportion <strong>of</strong><br />

ground-glass opacity (%GGO) is well-known to be strongly associated with<br />

less-invasive lung adenocarcinoma. Recently, the diameter <strong>of</strong> the solid area<br />

(SolidØ) has been reported to also be a simple and better marker for the<br />

same purpose compared to the diameter <strong>of</strong> the whole nodule (NoduleØ).<br />

The aim <strong>of</strong> this study was to confirm that SolidØ can completely replace<br />

%GGO in predicting less-invasive lung cancer. Methods: From 1987 to<br />

2009, 433 patients with clinical T1a-2bN0 NSCLC underwent complete<br />

resection, and their preoperative HRCT images were preserved in DICOM<br />

format. NoduleØ and SolidØ were precisely measured using s<strong>of</strong>tware.<br />

%GGO was calculated using the method we previously published (Eur J<br />

Cardiothorac Surg 25:1102-6, 2004). Less-invasive lung cancer was<br />

defined as having no vascular, lymphatic, nor pleural invasion. We<br />

compared the three parameters with regard to predicting less-invasive lung<br />

cancer and recurrence. Results: Among the 433 patients, 220 were male,<br />

367 had an adenocarcinoma histology, and 58 experienced recurrence.<br />

Table shows percentages and numbers <strong>of</strong> non-less-invasive lung cancer<br />

cases. Among each category <strong>of</strong> SolidØ, greater %GGO is associated with<br />

less-invasive lung cancer. ROC analysis also showed that the area under the<br />

curve <strong>of</strong> %GGO was the highest (0.859, 95% CI 0.824 – 0.893), followed<br />

by SolidØ (0.806, 0.767 – 0.846), and NoduleØ (0.671, 0.620 – 0.721).<br />

Regardless <strong>of</strong> SolidØ, no patient with a greater %GGO (� 50%) experienced<br />

recurrence. Conclusions: Although SolidØ is a better prognostic<br />

indicator <strong>of</strong> non-invasiveness compared to NoduleØ, %GGO remains<br />

important.<br />

SolidØ %GGO 0-9% 10-49% 50-79% 80-100%<br />

31mm~ 22/22 (100) 30/33 (91) 1/3 (33) 0/0 (0)<br />

25~30mm 15/21 (71) 28/31 (90) 3/5 (60) 0/0 (0)<br />

21~25mm 36/30 (87) 38/64 (59) 0/4 (0) 0/0 (0)<br />

16~20mm 40/49 (82) 48/76 (63) 3/12 (25) 0/1 (0)<br />

11~15mm 14/22 (64) 24/59 (41) 2/27 (7) 0/3 (0)<br />

6~10mm 3/5 (60) 3/7 (43) 0/18 (0) 0/19 (0)<br />

0~5mm 0/1 (0) 0/2 (0) 0/2 (0) 0/23 (0)<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


Lung Cancer—Non-small Cell Local-Regional/Small Cell/Other Thoracic Cancers<br />

7022 Poster Discussion Session (Board #14), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Vorinostat (V) intratumoral levels and biomarker response in a window <strong>of</strong><br />

opportunity trial in patients with resectable aerodigestive tract cancer.<br />

Presenting Author: Konstantin H. Dragnev, Dartmouth-Hitchcock Medical<br />

Center, Lebanon, NH<br />

Background: Aerodigestive tract cancer (ADT) is the leading cause <strong>of</strong><br />

cancer-related death in the US. New effective treatments are needed.<br />

Among a panel <strong>of</strong> drugs causing growth inhibition <strong>of</strong> lung cancer cells and<br />

repression <strong>of</strong> cyclin D1, the histone deacetylase inhibitor V was found most<br />

potent. To translate studies into the clinic, a window <strong>of</strong> opportunity trial <strong>of</strong> V<br />

was conducted. Methods: Pts with resectable ADT received 400 mg V once<br />

daily orally for 7 days prior to surgical resection. Serum samples were<br />

obtained before the last V dose and at the time <strong>of</strong> biopsy. Tumor tissue was<br />

immediately snap-frozen in liquid nitrogen, stored at -70°C, homogenized<br />

in three parts PBS (1:3g/v). V was quantitated with a LC-MS/MS assay.<br />

Post- versus pre-treatment tumor biopsies were scored for necrosis, acute<br />

and chronic inflammation, and immunohistochemical changes in Ki-67,<br />

cyclin E, cyclin D1, EGFR, phospho-EGFR, p21, p27 and caspase. Results:<br />

15 pts enrolled, 9 pts took V for a median <strong>of</strong> 7 days (3-9), underwent<br />

resection and had adequate pretreatment samples. Median age 66, 4<br />

women, 4 former, 5 current smokers, 1 squamous cell lung carcinoma, 8<br />

adenocarcinoma (1 esophageal). PK analyses confirmed plasma (8pts) and<br />

tumor (7pts) concentrations above the detection limits. There was considerable<br />

interindividual variation in plasma V concentrations (7.3-192.4<br />

ng/ml; CV 95%), at 178-500 min from last V dose, and in intratumoral V<br />

levels (15.0-80.3 ng/g; CV 59%). All pts had wild-type EGFR, 4 pts had<br />

KRAS codon 12 mutations, 78% showed reduced Ki-67 expression, 50%<br />

had decreased cyclin E, 78% exhibited necrosis, chronic or acute inflammation<br />

in the post-treatment biopsies. Most cases with KRAS mutations<br />

had biomarker responses. Changes in multiple biomarkers were observed<br />

across the range <strong>of</strong> intratumoral V levels. Conclusions: This is the first report<br />

<strong>of</strong> V concentrations in human tumors. Preoperative treatment with V<br />

achieved intratumoral concentrations comparable to serum with evidence<br />

<strong>of</strong> necrosis, decreased Ki-67 and cyclin E, and other biomarker responses<br />

in resected ADT. The results demonstrate the value <strong>of</strong> window <strong>of</strong> opportunity<br />

trials in the investigation <strong>of</strong> novel cancer therapeutics.<br />

7024 Poster Discussion Session (Board #16), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Predictors <strong>of</strong> survival after resections <strong>of</strong> synchronous lung cancers located<br />

in mutiple lobes: A multi-institutional pooled analysis. Presenting Author:<br />

Tawee Tanvetyanon, H. Lee M<strong>of</strong>fitt Cancer Center & Research Institute,<br />

Tampa, FL<br />

Background: Surgical resection is a treatment option for patients with<br />

multiple primary lung cancers. However, for synchronous disease, it is<br />

<strong>of</strong>ten difficult to differentiate this condition from metastatic disease,<br />

especially when cancers are distributed in multiple lobes or in both lungs.<br />

Some people believe that surgery should be avoided when patients have<br />

bilateral cancers or when all cancers have the same histology. To date,<br />

however, available evidences are limited by small sample size. Methods:<br />

Studies (published 2008-2011) <strong>of</strong> curative resection for patients with<br />

synchronous (� 2-year interval) multiple lung cancers located in � 2 lobes,<br />

but without radiographic evidence <strong>of</strong> distant metastasis, were identified<br />

from literature. Corresponding authors were contacted and individual<br />

patient data were obtained. Patients with multiple cancers, but localized<br />

only to one lobe, were not included. Databases were pooled and multivariable<br />

Cox Proportional Hazard models were fit to adjust for confounders.<br />

Results: There were 467 patients included from 6 studies. Median overall<br />

survival was 52.0 months (95% CI: 45.6-63.7). Postoperative (30-day)<br />

mortality rate was 1.9%. In a multivariable model, study site or having<br />

pneumonectomy did not independently impact on survival. However, age,<br />

gender, nodal stage, tumor location, and histological similarity were<br />

independent predictors <strong>of</strong> survival. Advanced age increased mortality<br />

(p�0.012). Male sex increased mortality compared with female: HR 1.64<br />

(95% CI: 1.23-2.19, p�0.0008). N2 or N1 increased mortality over N0:<br />

HR 1.85 (95% CI 1.29-2.66, p�0.0008) and 1.97 (1.43-2.73,<br />

p�0.0001), respectively. Unilateral location increased mortality over<br />

bilateral location: HR 1.62 (95% CI 1.23-2.13, p�0.0002). Different<br />

histology increased mortality over similar histology: HR 1.45 (95% CI<br />

1.11-1.90, p�0.0069). Conclusions: In this largest multi-institutional<br />

database <strong>of</strong> resected multiple lung cancers <strong>of</strong> multiple lobes to date, we<br />

found no evidence <strong>of</strong> inferior survival among patients having bilateral<br />

cancers or having all cancers with the same histology. In fact, the survival<br />

among such patients appears superior to their counterparts.<br />

7023 Poster Discussion Session (Board #15), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Use <strong>of</strong> a proliferation-based mRNA signature to predict outcome in<br />

early-stage non-small cell lung adenocarcinoma. Presenting Author: Carmen<br />

Behrens, University <strong>of</strong> Texas M. D. Anderson Cancer Center, Houston,<br />

TX<br />

Background: Adjuvant treatment <strong>of</strong> patients with early-stage lung adenocarcinoma<br />

is based on post-surgical pathological staging and patient performance<br />

status. Disparate outcomes within each staging group suggest that<br />

additional prognostic markers could improve our understanding <strong>of</strong> riskbenefit<br />

and potentially lead to better treatment decisions. A proliferationbased,<br />

mRNA expression pr<strong>of</strong>ile was applied to public microarray data <strong>of</strong><br />

surgically treated lung adenocarcinomas and a cohort <strong>of</strong> FFPE samples to<br />

test its potential prognostic utility. Methods: Public expression data<br />

(Director’s Consortium, DC) were derived from Affymetrix HG-U133A<br />

arrays. <strong>Clinical</strong> FFPE samples were assayed by quantitative PCR. A cell<br />

cycle progression (CCP) score was calculated from the expression average<br />

<strong>of</strong> 31 cell cycle genes normalized by 15 housekeeper genes. The prognostic<br />

value <strong>of</strong> the CCP score to predict stage I and II patient outcomes was<br />

evaluated by Cox proportional hazards analysis with disease-related death<br />

as the primary outcome measure. Results: In 256 DC cases, the CCP score<br />

was a significant predictor <strong>of</strong> death in univariate (p�0.0001) and multivariate<br />

analysis (p�0.001, HR 1.57, 95%CI 1.20-2.05) using age, stage,<br />

gender, smoking status and treatment as covariates. Similarly, in a second<br />

data set (GSE31210, n�204) the CCP score was highly associated with<br />

death (univariate, p�0.001; multivariate analysis, p�0.003, HR 1.81,<br />

95% CI 1.24-2.66). Using quantitative PCR, the signature was applied to<br />

381 FFPE samples with a median follow-up <strong>of</strong> 5 years collected at the MD<br />

Anderson Cancer Center and the European Institute for Oncology. In the<br />

presence <strong>of</strong> clinical covariates (as above and tumor size and pleural<br />

invasion), the CCP score remained the most significant predictor <strong>of</strong> death in<br />

univariate (p�0.0003) and multivariate analysis (p�0.007, HR 1.50,<br />

95% CI 1.11-2.02). Conclusions: A 46 gene mRNA signature is a<br />

significant predictor <strong>of</strong> disease-related death in early-stage lung adenocarcinoma,<br />

providing independent prognostic value in the presence <strong>of</strong> clinical<br />

variables. This molecular predictor <strong>of</strong> cancer survival will be studied in<br />

additional cohorts for its ability to impact clinical treatment decisions.<br />

7025 Poster Discussion Session (Board #17), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Prognostic impact <strong>of</strong> the number <strong>of</strong> examined lymph nodes (LNs) in<br />

resected node negative (pNo) non-small cell lung cancer (NSCLC). Presenting<br />

Author: Obiageli Uchenna Ogbata, University <strong>of</strong> Tennessee Health<br />

Science Center, Memphis, TN<br />

Background: In the US, 29% <strong>of</strong> patients undergo curative-intent surgery for<br />

NSCLC. Absence <strong>of</strong> LN metastasis and the extent <strong>of</strong> LN examination<br />

influence survival. 44% <strong>of</strong> patients with pathological node negative (pN0)<br />

disease die within 5 years. There is no consensus on the optimal number <strong>of</strong><br />

LNs to be examined to determine pN0 stage. We hypothesized that patients<br />

with few examined nodes may have missed nodal metastasis and increasing<br />

the number <strong>of</strong> LNs examined would improve survival. Methods: Retrospective<br />

SEER database analysis <strong>of</strong> NSCLC resections from 1998 to 2002, with<br />

survival updated to 2008. Patients with first primary, pN0 NSCLC, with one<br />

or more LNs examined, met our study criteria. Cox regression and<br />

competing risk models were used for survival analysis. A p value �0.05 was<br />

considered statistically significant. Results: 8,137 patients met inclusion<br />

criteria and were evaluated. A median <strong>of</strong> 6 LNs were examined in this<br />

cohort. Patients who underwent pneumonectomy and lobectomy had more<br />

LNs examined than those who had sub-lobar resections. Higher number <strong>of</strong><br />

LNs examined was associated with better overall survival (OS) and lung<br />

cancer specific survival (LCSS), at a plateau <strong>of</strong> 8 LNs for both outcomes<br />

(Table). Having mediastinal LNs examined also resulted in increased OS<br />

and LCSS (p�0.05). Conclusions: The pathologic assessment <strong>of</strong> LNs in<br />

surgical specimens is <strong>of</strong>ten suboptimal. Examining more LNs may have<br />

increased the likelihood <strong>of</strong> correct staging. Lower numbers <strong>of</strong> LNs examined<br />

probably result in understaging. Examining 8 or more total LNs and<br />

mediastinal LNs examined improved both OS and LCSS.<br />

#<strong>of</strong>LN<br />

examined<br />

Hazard<br />

ratio<br />

for OS<br />

P value<br />

for OS<br />

95% CI<br />

for OS<br />

Hazard<br />

ratio<br />

for LCSS<br />

P value<br />

for LCSS<br />

457s<br />

95% CI<br />

for LCSS<br />

2 0.97 0.652 0.85 – 1.11 0.88 0.138 0.74 – 1.04<br />

3 0.93 0.262 0.81 – 1.06 0.87 0.101 0.74 – 1.02<br />

4 0.94 0.342 0.82 – 1.07 0.82 0.026 0.69 – 0.98<br />

5 0.83 0.008 0.72 – 0.95 0.78 0.008 0.66 – 0.94<br />

6 0.82 0.008 0.71 – 0.95 0.77 0.004 0.64 – 0.92<br />

7 0.86 0.052 0.74 – 1.00 0.76 0.005 0.62 – 0.92<br />

8 0.78 0.003 0.66 – 0.92 0.71 0.002 0.58 – 0.88<br />

9 0.85 0.052 0.73 – 1.00 0.79 0.026 0.64 – 0.97<br />

10 0.81 0.022 0.68 – 0.86 0.75 0.016 0.60 – 0.95<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


458s Lung Cancer—Non-small Cell Local-Regional/Small Cell/Other Thoracic Cancers<br />

7026 Poster Discussion Session (Board #18), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

The effect <strong>of</strong> comorbidity on stage-specific survival in resected non-small<br />

cell lung cancer patients. Presenting Author: MARK Krasnik, Rigshospitalet,<br />

Copenhagen, Denmark<br />

Background: TNM classification has been the traditional cornerstone <strong>of</strong><br />

decision-making in treatment choices <strong>of</strong> lung cancer. In addition to TNM<br />

stage and lung function, there is no other objective information to support<br />

this decision. Severe comorbidity affects overall survival, and may be an<br />

independent prognostic factor. In this study we quantified the effect <strong>of</strong><br />

comorbidity on stage-specific survival in resected non-small cell lung<br />

cancer (NSCLC) patients in a large population-based setting. Methods:<br />

Among 3,152 NSCLC patients from the Danish Lung Cancer Registry who<br />

underwent surgical resection between 1 January 2005 and 31 December<br />

2010, mortality hazard ratios were calculated during three consecutive<br />

time periods following surgery (0-1 month, 1 month - 1 year and �1 year)<br />

according to Charlson comorbidity score (CCS 0, 1-2, 3�), ECOG performance<br />

status, lung function, age, sex, pathological T and N stage according<br />

to the revised 7th edition TNM lung cancer staging system, using Cox<br />

proportional hazard modelling. The Kaplan-Meier method was used to<br />

describe stage-specific survival according to the Charlson comorbidity<br />

score. Results: Increasing severity <strong>of</strong> comorbidity was independently<br />

associated with significantly higher death rates throughout the three<br />

periods <strong>of</strong> follow-up [HR1.18 for CCS 1-2 and 2.06 for CCS 3� in 0-1<br />

month, 1.13 for CCS 1-2 and 1.57 for CCS 3� during 1 month - 1 year and<br />

1.14 for CCS 1-2 and 1.84 for CCS 3� after 1 year]. Stage-specific<br />

five-year survival in patients with severe comorbidity (CCS 3�) was<br />

significantly lower than in patients without comorbid disease [e.g., 38%<br />

(95% CI 23-53%) for pT1 and CCS 3� vs. 69% (62-75%) for pT1 and CCS<br />

0]. Conclusions: This study shows that severe comorbidity affects survival <strong>of</strong><br />

NSCLC patients undergoing surgical resection by as much as a single stage<br />

increment. As the survival <strong>of</strong> NSCLC patients after different treatment<br />

regiments is mainly based on the TNM classification, further research is<br />

necessary to fully identify which patients are most likely to benefit from<br />

surgery, but also to provide a basis for developing a better prediction<br />

instrument for the survival after combination treatment involving radiotherapy<br />

and chemotherapy.<br />

7028 Poster Discussion Session (Board #20), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Randomized phase III study comparing etoposide and cisplatin (EP) with<br />

irinotecan and cisplatin (IP) following EP plus concurrent accelerated<br />

hyperfractionated thoracic radiotherapy (EP/AHTRT) for the treatment <strong>of</strong><br />

limited-stage small-cell lung cancer (LD-SCLC): JCOG0202. Presenting<br />

Author: Kaoru Kubota, National Cancer Center Hospital East, Kashiwa,<br />

Japan<br />

Background: Four cycles <strong>of</strong> EP plus AHTRT is the standard treatment for<br />

LD-SCLC. IP demonstrated statistically significant overall survival (OS)<br />

improvement compared to EP for extensive-stage SCLC (JCOG9511; Noda,<br />

et al.NEJM, 2002). EP plus AHTRT followed by 3 cycles <strong>of</strong> IP is feasible<br />

with acceptable toxicities for LD-SCLC (Kubota, et al. CCR,2005). Methods:<br />

Eligibility criteria included patients with previously untreated LD-SCLC<br />

with measurable lesion, ECOG PS <strong>of</strong> 0-1, age: 20��, ��70 years old.<br />

Eligible patients received one cycle <strong>of</strong> EP (etoposide 100 mg/m2 on days<br />

1-3 and cisplatin 80mg/m2 on day 1) plus AHTRT (1.5 Gy b.i.d. total 45<br />

Gy/3 weeks). Patients who achieved CR, good PR, PR or SD with induction<br />

EP/AHTRT were randomized to receive either 3 cycles <strong>of</strong> consolidation EP<br />

or IP (irinotecan 60 mg/m 2 and cisplatin 60 mg/m2 on days 1, 8, 15).<br />

Patients with CR or good PR after consolidation chemotherapy received<br />

prophylactic cranial irradiation. The primary endpoint is OS after the<br />

randomization. The planned sample size for randomization is 250 with a<br />

one-sided alpha <strong>of</strong> 2.5% and at least 70% power to detect a difference<br />

between gruops, 30% in EP versus 42.5% in IP group in 3-year survival.<br />

Results: From Sep 2002 to Sep 2006, 281 patients from 36 institutions<br />

were registered. After the induction EP/AHTRT, 258 patients were randomized<br />

to consolidation EP (n�129) or IP (n�129). Patient demographics<br />

were well balanced between the two groups. At the final analysis, the<br />

superiority <strong>of</strong> IP in OS was not demonstrated (hazard ratio <strong>of</strong> IP to EP,<br />

1.085 [95%CI: 0.80-1.46]; one sided p�0.70, stratifield log-rank test).<br />

Grade 3/4 neutropenia (95%/78%), anemia (35%/39%), thrombocytopenia<br />

(21%/5%), neutropenic fever (17%/14%), diarrhea (2%/10%) were<br />

observed in EP and IP groups, respectively. Conclusions: EP plus AHTRT<br />

followed by 3 cycles <strong>of</strong> IP failed to demonstrate survival advantage over 4<br />

cycles <strong>of</strong> EP plus AHTRT which still is the standard treatment for LD-SCLC.<br />

EP IP<br />

Median OS 3.2 years 2.8 years<br />

3-year OS 53% 47%<br />

5-year OS 36% 34%<br />

Median PFS 1.1 years 1.0 year<br />

7027 Poster Discussion Session (Board #19), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Randomized phase II trial comparing two schedules <strong>of</strong> thoracic radiotherapy<br />

(TRT) in limited disease small-cell lung cancer (LD SCLC).<br />

Presenting Author: Bjørn Henning Grønberg, Department <strong>of</strong> Cancer Research<br />

and Molecular Medicine, Norwegian University <strong>of</strong> Science and<br />

Technology and The Cancer Clinic, St. Olavs Hospital-Trondheim University<br />

Hospital, Trondheim, Norway<br />

Background: Concurrent chemotherapy and TRT is standard therapy for<br />

SCLC if all lesions can be included in a radiotherapy field (LD). Several<br />

schedules <strong>of</strong> TRT are used. One study showed that two fractions a day<br />

improved local control and overall survival (OS), but this schedule has not<br />

been compared to a commonly used 3 wks schedule. Methods: Eligible pts<br />

had LD SCLC and PS 0-2. Pleural fluid was accepted if negative cytology.<br />

Pts received 4 cycles <strong>of</strong> PE (cisplatin 75 mg/m2 IV day 1 and etoposide 100<br />

mg/m2 IV day 1-3 q 3 wks) and were randomly assigned to 3 wks <strong>of</strong> 3D<br />

conformal TRT [A] 42 Gy (2.8 Gy x 1/day) or [B] 45 Gy (1.5 Gy x 2/day). TRT<br />

started 3-4 wks after the first PE. All responders received prophylactic<br />

cranial irradiation (PCI) 2 Gy x 15 � 6 wks after last PE. Pts reported health<br />

related quality <strong>of</strong> life (HRQoL) on EORTC QLQ C30 � LC13. Primary<br />

endpoint: 1-year local failure. Secondary: OS, toxicity and HRQoL (dysphagia<br />

and dyspnea; a difference � 10 points was considered significant). 75<br />

pts were required in each arm to show a 30% improvement <strong>of</strong> local disease<br />

control with ��.05 and ��.8. Results: 159 eligible pts were enrolled at 18<br />

sites in Norway May 05 – Jan 11 (A: 85, B: 74). Median age 60 (40-85);<br />

52% men, 84 % PS 0-1, 11% pleural fluid. Mean no. <strong>of</strong> PE-cycles was 3.8,<br />

97% completed TRT, 82 % PCI (no difference between arms). Response<br />

rates were similar (A: 92%, B: 94%; p�.8), but more pts on Arm B had CR<br />

(A: 13%, B: 35%; p�.01). There was no difference in local failure as first<br />

site <strong>of</strong> progression at 1 year (A: 17%, B: 12%; p�.4) or 1-year PFS (A:<br />

44%, B: 50% ; p�.4). There was similar grade 3-4 es<strong>of</strong>agitis (A: 33%, B:<br />

37 %; p�.7) and pneumonitis (A: 6 %, B: 7 %; p�.9). 2 pts (1 on each<br />

arm) died from pneumonitis. Pts in Arm B reported more dysphagia (A: 64<br />

points, B: 73 points), but not more dyspnea (A: 29 points, B: 28 points).<br />

1-year OS was similar (A: 77%, B: 76%; p�.9). Currently, 2-year survival<br />

among those followed � 2 years (n�130) favors Arm B (A: 40%, B: 55%;<br />

p�.09) and so far (all pts followed � 1-year; 103 events) median OS favors<br />

Arm B (A: 18.7 mos, B: 26.6 mos; p�.34). Conclusions: Twice daily TRT<br />

resulted in more CRs, slightly more dysphagia, similar 1-year local control<br />

and 1-year PFS. There are indications <strong>of</strong> improved 2-year and median OS in<br />

this arm.<br />

7029 Poster Discussion Session (Board #21), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Randomized phase II trial <strong>of</strong> pemetrexed/cisplatin with or without CBP501<br />

in patients with advanced malignant pleural mesothelioma (MPM). Presenting<br />

Author: Lee M. Krug, Memorial Sloan-Kettering Cancer Center, New<br />

York, NY<br />

Background: CBP501, a synthetic duodecapeptide, enhances the cytotoxicity<br />

<strong>of</strong> cisplatin in cell lines and xenografts, including NCI-H226 human<br />

mesothelioma. CBP501 increases cisplatin influx into tumor cells through<br />

an interaction with calmodulin, and inhibits cell cycle progression by<br />

abrogating DNA repair at the G2 checkpoint. Phase I clinical trials<br />

combining CBP501 with cisplatin alone or with pemetrexed showed<br />

acceptable safety pr<strong>of</strong>iles and suggested activity. The most common<br />

toxicity <strong>of</strong> CBP501 is infusion-related urticaria, which is easily managed<br />

with diphenhydramine. Methods: Chemotherapy-naive patients with unresectable<br />

MPM were stratified by histology (epithelioid vs other) and<br />

performance status (PS 0-1 vs 2) and randomized 2:1 to Arm A:<br />

pemetrexed/cisplatin plus CBP501 25 mg/m2 IV (42 pts planned), or Arm<br />

B: pemetrexed/cisplatin alone at standard doses (21 pts planned). Patients<br />

continued on treatment until progression or intolerance; no maintenance<br />

therapy was delivered. The primary endpoint is progression free survival<br />

(PFS) in Arm A; if � 23 <strong>of</strong> the 42 patients remain free <strong>of</strong> progression more<br />

than 4 months, the combination will be deemed worthy <strong>of</strong> further study. In<br />

addition to standard CT imaging to assess response and PFS, PET scans,<br />

pulmonary function tests, and mesothelin levels were performed. Results:<br />

Enrollment was completed in October 2011. 65 pts from 14 institutions<br />

were randomized, and 63 were treated. Patient characteristics in the two<br />

arms were similar and as follows: median age 65, 82% male, 71%<br />

epithelioid histology, 8% PS2. Grade 3/4 treatment-related toxicities were<br />

uncommon, no different than expected from standard chemotherapy, and<br />

comparable in the two arms. 70% <strong>of</strong> patients treated with CBP501 had<br />

infusion reactions, all grade 1-2. The best overall response rate in evaluable<br />

patients (modified RECIST, investigator assessed) was 12/37 (32%) (95%<br />

CI 18-50) in Arm A, and 3/22 (14%) (95% CI 3-35) in Arm B. The median<br />

number <strong>of</strong> chemotherapy cycles was 5.5 in Arm A, 5 in Arm B. Conclusions:<br />

Data on the primary endpoint <strong>of</strong> PFS in Arm A are maturing and will be<br />

presented at the meeting.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


Lung Cancer—Non-small Cell Local-Regional/Small Cell/Other Thoracic Cancers<br />

7030 Poster Discussion Session (Board #22), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Amatuximab, a chimeric monoclonal antibody to mesothelin, in combination<br />

with pemetrexed and cisplatin in patients with unresectable pleural<br />

mesothelioma: Results <strong>of</strong> a multicenter phase II clinical trial. Presenting<br />

Author: Raffit Hassan, National Cancer Institute, Bethesda, MD<br />

Background: Amatuximab (MORAb-009) is a chimeric monoclonal antibody<br />

to mesothelin, a cell surface glycoprotein highly expressed in many cancers<br />

including malignant mesothelioma (MM). Based on safety <strong>of</strong> amatuximab<br />

in phase I clinical trial and pre-clinical studies showing synergy in<br />

combination with chemotherapy, a single arm phase II study <strong>of</strong> amatuximab<br />

plus pemetrexed (P) and cisplatin (C) was initiated in pts. with MM.<br />

Methods: Eligibility criteria included pts. with unresectable epithelial or<br />

biphasic pleural MM, no prior chemotherapy and KPS � 70%. Pts.<br />

received amatuximab 5 mg/kg on days 1 and 8 with P 500 mg/m2 andC75<br />

mg/m2 (PC) given on day 1, <strong>of</strong> each 21 day cycle for 6 cycles. Pts. with<br />

objective response or stable disease received amatuximab monotherapy<br />

until disease progression. Primary endpoint was progression-free survival<br />

(PFS) at 6 months. Secondary endpoints were overall survival (OS),<br />

objective response rate and safety <strong>of</strong> amatuximab with PC. Results: From<br />

2/2009 to 10/2010, 89 pts. were enrolled at 26 sites. Pt. characteristics:<br />

median age 67 yrs. (range 46-80), 78% male, 70% with KPS �90%, 89%<br />

epithelial MM, 11% biphasic MM and 88% had stage III/IV disease.<br />

Median number <strong>of</strong> PC plus amatuximab cycles was 5 (range 1-6) and 56<br />

(63%) pts. received single agent amatuximab. In addition to the expected<br />

toxicity from PC, hypersensitivity reactions (12.4%; Grade 3/4�4.5%)<br />

from amatuximab were noted. By independent radiological review 30<br />

(39%) pts. had partial response and 39 (51%) had stable disease. PFS at 6<br />

months was 52% (95% CI: 39.5-63.5) with a median PFS <strong>of</strong> 6.1 months<br />

(95% CI: 5.4-6.5). As <strong>of</strong> 1/10/12 the median OS was 14.5 months (95%<br />

CI: 12.4-18.5) with 31 pts. still alive and 7 pts. receiving maintenance<br />

amatuximab. Conclusions: Amatuximab in combination with PC was generally<br />

well-tolerated in this study with 90% <strong>of</strong> pts. having an objective tumor<br />

response or stable disease by independent radiological review. Although<br />

PFS is not significantly different from historical results <strong>of</strong> PC alone, the<br />

median OS is 14.5 months with 35% <strong>of</strong> pts. still alive. Updated OS and<br />

biomarker data will be presented at the meeting.<br />

7032 Poster Discussion Session (Board #24), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Next-generation sequencing <strong>of</strong> thymic malignancies. Presenting Author:<br />

Milan Radovich, Indiana University School <strong>of</strong> Medicine, Indianapolis, IN<br />

Background: Thymic malignancies are rare with ~500 cases in the US per<br />

year. Apart from standard chemotherapy, treatment options are limited.<br />

Further, the challenge <strong>of</strong> histologic subtyping <strong>of</strong> these tumors along with an<br />

inadequate understanding <strong>of</strong> the transcriptional biology is a hindrance to<br />

the development <strong>of</strong> targeted therapies. Methods: Thymic malignancies and<br />

normal tissues were obtained from the Indiana University Simon Cancer<br />

Center. The WHO subtypes <strong>of</strong> our samples include: (4) type A, (2) A/B, (1)<br />

B2, (5) B3, (1) C, and (3) normal tissues. RNA was sequenced on a Life<br />

Technologies SOLiD sequencer. For gene expression, reads were mapped to<br />

the genome using BioScope and outputs imported into <strong>Part</strong>ek GS. In<br />

<strong>Part</strong>ek, statistical comparison <strong>of</strong> gene expression as well as PCA &<br />

clustering analyses were performed. Results: Unsupervised hierarchical<br />

clustering <strong>of</strong> gene expression values revealed 100% concordance between<br />

gene expression clusters and WHO subtype. A subsequent unsupervised<br />

clustering <strong>of</strong> 705 pre-miRNAs also showed substantial concordance<br />

between clusters and subtype. By analyzing the dendrograms, A&A/B<br />

tumors were significantly different from B type tumors, as well as C and<br />

normal. A substantial differentiator was a large cluster <strong>of</strong> overexpression in<br />

A&A/B tumors that was nearly absent in the others on chr19q13.42<br />

corresponding to the miR-515 cluster (43 miRNAs). When comparing A &<br />

A/B tumors vs. B type tumors, 1334 genes are differentially expressed (258<br />

downregulated) (FDR�5%). 95/258 genes are predicted to be downregulated<br />

by the miR-515 cluster including several transcription factors and<br />

tumor suppressors. When looking at mutations, we detected no recurrent<br />

gene fusions, though we are detecting several point mutations and small<br />

insertion deletions. These are being followed up by exome sequencing.<br />

Conclusions: Analyses reveal that RNA-seq can be used to accurately<br />

subtype Thymic Malignancies and the development <strong>of</strong> an expression based<br />

diagnostic is feasible. Further, these data support that the main differentiator<br />

<strong>of</strong> thymomas may lie in the expression <strong>of</strong> a single miRNA cluster. In<br />

addition, several mutations in key pathways are implicated. Ongoing<br />

analyses include: alternative splicing, noncoding RNAs, viral analysis and<br />

exome sequencing.<br />

459s<br />

7031 Poster Discussion Session (Board #23), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Comparison <strong>of</strong> prognostic impact between metabolic and radiologic response<br />

after induction chemotherapy for resectable malignant pleural<br />

mesothelioma: A multicenter study. Presenting Author: Yasuhiro Tsutani,<br />

Hiroshima University, Hiroshima, Japan<br />

Background: Induction chemotherapy followed by extrapleural pneumonectomy<br />

(EPP) for resectable malignant pleural mesothelioma (MPM) is<br />

controversial. To select optimal candidates for EPP, we evaluates the<br />

usefulness <strong>of</strong> metabolic response on fluorodeoxyglucose-positron emission<br />

tomography (FDG-PET) compared with radiologic response on highresolution<br />

computed tomography (HRCT) after induction chemotherapy to<br />

predict prognosis for patients with resectable MPM who underwent EPP in<br />

the setting <strong>of</strong> multicenter study. Methods: We performed HRCT and<br />

FDG-PET before and after induction platinum-based chemotherapy on 50<br />

patients with clinical T1-3 N0-2 M0 MPM who underwent EPP �<br />

postoperative hemithoracic radiation. A decrease <strong>of</strong> more than 30% in<br />

tumor maximum standardized uptake value (SUVmax) was defined as a<br />

metabolic responder. Radiologic response using modified RECIST or<br />

metabolic response and surgical results were analyzed. Results: In all<br />

cohort patients, median overall survival (OS) from diagnosis was 20.5<br />

month (95% confidence interval [CI], 15.0-26.0 month). Fourteen patients<br />

were classified as metabolic responders (28%) and 36 as nonresponders<br />

(72%). Metabolic responders significantly correlated to OS with<br />

median OS for metabolic responders <strong>of</strong> not reached (3-year OS <strong>of</strong> 60.0%)<br />

versus 18.7 month (95% CI, 13.3-24.2 month, 3-year OS <strong>of</strong> 26.5%) for<br />

non-responders (P � .025). No correlation was observed between OS and<br />

radiologic response with median OS for radiologic responders <strong>of</strong> 25.7<br />

month (n � 20, 95% CI, 14.5-37.0 month, 3-year OS <strong>of</strong> 35.8%) versus<br />

17.7 month (n � 30, 95% CI, 12.8-22.6 month, 3-year OS <strong>of</strong> 35.1%) for<br />

non-responder (p � .216). Based on the multivariate Cox analyses,<br />

decreased SUVmax (%) (p � .010) was a significantly independent factor<br />

for OS as well as epithelioid subtype (p � .044). Conclusions: Metabolic<br />

response on FDG-PET has higher predictive value <strong>of</strong> prognosis than<br />

radiologic response on HRCT after induction chemotherapy for patients<br />

with resectable MPM. Patients with metabolic responder and/or epithelioid<br />

subtype can be good candidates for EPP after induction chemotherapy.<br />

7033 Poster Discussion Session (Board #25), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Phase II study <strong>of</strong> cixutumumab (IMC-A12) in thymic malignancies.<br />

Presenting Author: Arun Rajan, National Cancer Institute, Bethesda, MD<br />

Background: The IGF-1 receptor (IGF-IR) is expressed in thymoma (T) and<br />

thymic carcinoma (TC). Prolonged SD has been seen with anti-IGF-IR<br />

therapy in thymic tumors in phase I trials. Cixutumumab (C) is a fully<br />

human IgG1 monoclonal antibody (ab) that targets IGF-IR. Methods:<br />

Patients (pts) with recurrent T or TC, PS 0-2, measurable disease, and<br />

normal organ function received C at 20 mg/kg IV q3wks until progression or<br />

intolerable toxicity. Primary endpoint was response rate. Correlative studies<br />

included immune cell subset (central memory, effector memory, naïve and<br />

regulatory T cells), circulating endothelial progenitor (CEP) cell, serum<br />

IGF-1 and IGF-IR in PBMCs, anticytokine ab, and tumor mutational<br />

analysis. Results: Forty-five pts enrolled from two centers (20 female,<br />

T�33, med age 52 yrs, range, 26-86). Median number <strong>of</strong> prior systemic<br />

treatments: 3 (range, 1-11). Median number <strong>of</strong> cycles <strong>of</strong> C administered: 6<br />

(range, 1-41). In 30 evaluable T pts there were 4 (13%) PR, 23 (77%) SD,<br />

and 3 (10%) PD. Corresponding numbers in 12 TC pts were 0, 5 (42%) and<br />

7 (58%). Median PFS for T and TC was 40 and 9 wks respectively.<br />

C-related grade 3/4 AEs were hyperglycemia (8%), hyperuricemia, weight<br />

loss, lipase elevation, chest wall pain (5% each) and AST elevation (3%).<br />

Two pts died while on study: one due to respiratory insufficiency related to T<br />

after 5 cycles and one due to worsening myasthenia and an acute coronary<br />

event after 13 cycles. In 8 <strong>of</strong> 33 T pts autoimmune (AI) symptoms<br />

developed (4 new-onset) due to immune thrombocytopenic purpura,<br />

myositis, myocarditis, colitis, PRCA and food allergy. In the first 13 pts a<br />

trend towards increased numbers <strong>of</strong> central and effector memory T cells<br />

and a decrease in naïve T cells and CEPs was seen. High-titer anticytokine<br />

abs, including anti-IFNa, anti-IL-12, and anti-IL-17A, were found in 13/27<br />

pts tested. No EGFR, KRAS or BRAF mutation, HER2 amplification or ALK<br />

translocations were detected in 11 tumors analyzed. Conclusions: C<br />

monotherapy is well tolerated and active in T. Accrual will continue until 37<br />

T pts are enrolled. Activity in TC is unworthy <strong>of</strong> further investigation. CEP<br />

and immune cell subset analysis suggests therapy may impact angiogenesis<br />

and immune response. Development <strong>of</strong> AI symptoms during treatment<br />

needs further evaluation.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


460s Lung Cancer—Non-small Cell Local-Regional/Small Cell/Other Thoracic Cancers<br />

7034 General Poster Session (Board #34A), Sat, 1:15 PM-5:15 PM<br />

Epidermal growth factor receptor mutation status and adjuvant chemotherapy<br />

in resected advanced non-small cell lung cancer. Presenting<br />

Author: Si-Yu Wang, Cancer Center, Sun Yat-sen University, Guangzhou,<br />

China<br />

Background: Mutations in the epidermal growth factor receptor (EGFR) are<br />

associated with response to chemotherapy in patients with advance<br />

NSCLC. The purpose <strong>of</strong> this study was to assess the association <strong>of</strong><br />

mutations in the EGFR tyrosine kinase domain and the efficacy <strong>of</strong> adjuvant<br />

chemotherapy in patients with fully resected IIIA-N2 NSCLC tumors.<br />

Methods: Tumor samples (n �150) from patients in our prior trial with<br />

IIIA-N2 NSCLC who either had or had not received paclitaxel/vinorelbine<br />

plus carboplatin chemotherapy following removal <strong>of</strong> the tumor were<br />

analyzed for EGFR mutations in exons 19 and 21. The association <strong>of</strong> the<br />

presence <strong>of</strong> EGFR mutations and survival following treatment was assessed.<br />

Results: Mutations were identified in 33 (22%) patients (n�13 in<br />

the no chemotherapy [observation] arm and n�20 in the chemotherapy<br />

arm). Fourteen patients (9.3%) had deletion mutations in exon 19, and 19<br />

patients (12.7%) had a substitution mutation in exon 21. Compared with<br />

patients wild-type for EGFR, patients with EGFR mutations had numerical<br />

but not statistically significant improved disease-free survival (35 months<br />

[95% CI, 14.6-55.4] versus 23 months [95% CI, 17.3-28.7], respectively,<br />

P�0.339) and overall survival (36 months [95% CI, 27.9 to 44.1] versus<br />

26 months [95% CI, 20.1 to 31.9], respectively p�0.271) regardless <strong>of</strong><br />

treatment. Patients with wild-type EGFR had greater overall survival with<br />

chemotherapy compared to no adjuvant therapy (HR�1.920; 95%CI,<br />

1.245-2.963; p�0.003). In contrast, EGFR mutant patients in the<br />

observation group compared to the chemotherapy group had longer median<br />

disease-free survival (35 months [95% CI, 20.9 to 49.1] versus 27 months<br />

[95% CI, 5.3 to 48.7], respectively, p�0.671) and overall survival (33<br />

months [95% CI, 24.2 to 41.8] versus 40 months [95% CI, 31.8 to 48.2]<br />

respectively, p �0.360). Conclusions: In this study, the status <strong>of</strong> mutations<br />

in exons 19 and 21 <strong>of</strong> EGFR was associated with different clinical<br />

outcomes in patients with resected IIIA-N2 NSCLC tumors either treated<br />

with or without adjuvant chemotherapy. These findings suggest that a<br />

patient’s treatment should be customized to their EGFR mutational status.<br />

7036 General Poster Session (Board #34C), Sat, 1:15 PM-5:15 PM<br />

Intrapleural combination bevacizumab with cisplatin therapy for non-small<br />

cell lung cancer caused by non-small cell lung cancer. Presenting Author:<br />

Nan Du, Department <strong>of</strong> Oncology, First Affiliated Hospital, Chinese PLA<br />

General Hospital, Beijing, China<br />

Background: Malignant pleural effusion (MPE) is a common complicate <strong>of</strong><br />

advanced non-small cell lung cancer (NSCLC). Bevacizumab, a humanized<br />

monoclonal antibody to VEGF, has been shown to be efficient in suppressing<br />

the pleural fluid accumulation; however, it has not been reported that<br />

whether bevacizumab can be used intrapleurally for the treatment <strong>of</strong> MPE.<br />

The present study is to evaluate the efficacy and safety <strong>of</strong> combined<br />

intrapleural therapy with bevacizumab and cisplatin, an antineoplastic<br />

agent, in controlling MPE. Methods: A total <strong>of</strong> 65 NSCLC patients with MPE<br />

were randomly assigned to one <strong>of</strong> two groups: intrapleural bevacizumab<br />

(300mg) with cisplatin (60mg) therapy (n�35) and intrapleural cisplatin<br />

(60mg) therapy (n�30). Results: The curative efficacy following combination<br />

or cisplatin mono therapy were 85.71% and 56.67%, respectively,<br />

being combination therapy group (p�0.05) is significantly higher. In<br />

addition, the combination therapy showed a higher efficacy in the patients<br />

with VEGF positive (p�0.01). Among the 25 cases that showed initial<br />

resistance to chemotherapy, 22 responded well to the combination<br />

therapy. Furthermore, combined bevacizumab with cisplatin therapy significantly<br />

reduced the VEGF expression, as shown by quantitative RT-PCR. All<br />

procedures were well tolerated by the patients, with wwwno severe side<br />

effect detected. Conclusions: Intrapleural combined bevacizumab with<br />

cisplatin therapy was effective and safe in managing MPE caused by<br />

NSCLC; the expression level <strong>of</strong> VEGF in MPE can be a prognostic marker for<br />

bevacizumab therapy.<br />

7035 General Poster Session (Board #34B), Sat, 1:15 PM-5:15 PM<br />

Adjuvant carboplatin, docetaxel, bevacizumab, and erlotinib versus chemotherapy<br />

alone in patients with resected non-small cell lung cancer: A<br />

randomized phase II study <strong>of</strong> the Sarah Cannon Research Institute (SCRI).<br />

Presenting Author: David Michael Waterhouse, Oncology Hematology<br />

Care/SCRI, Cincinnati, OH<br />

Background: Adjuvant chemotherapy improves overall survival (OS) in<br />

patients (pts) with resected non small cell lung cancer (NSCLC). Bevacizumab<br />

and erlotinib improve survival in pts with advanced unresectable<br />

NSCLC. This randomized phase II multicenter pilot study examined the<br />

safety and efficacy <strong>of</strong> chemotherapy and bevacizumab followed by bevacizumab<br />

and erlotinib vs. chemotherapy alone in pts with resected NSCLC.<br />

Methods: Eligible pts had completely resected (R0) stage IB, II, or IIIA<br />

NSCLC (TNM 6th Ed.), any NSCLC histology, and an ECOG PS 0-1. Pts were<br />

excluded for preoperatively confirmed N2 disease; N2 disease found at<br />

surgery was allowed. Pts were randomized 1:1 to carboplatin AUC�5,<br />

docetaxel 60 mg/m2 , and bevacizumab 15 mg/kg IV d1 q 21 days x 4,<br />

followed by maintenance bevacizumab 15 mg/kg d1 and erlotinib 150 mg<br />

PO daily x 8 cycles (Arm A) or carboplatin AUC�6 and docetaxel 75 mg/m2 IV d1 q 21 days x 4 (Arm B). The primary endpoint was 1-year disease-free<br />

survival (DFS); safety, 2-year DFS, and OS were secondary endpoints.<br />

Results: 106 pts were enrolled from 2/2008 to trial closure 1/2012 (A�54;<br />

B�52). Baseline features were balanced: age 63 yrs (42–87); 54% male;<br />

IB (46%), IIA (8%), IIB (27%), IIIA (18%); adenocarcinoma (61%),<br />

squamous (31%). Arm A received a median <strong>of</strong> 7 cycles (1-12); Arm B 4<br />

cycles (1-4). 1- and 2-year DFS by stage are shown in the table. The 1-year<br />

DFS for all stages were 78% (A) and 88% (B) (p�.66). The 3-year OS for all<br />

stages were 81% (A) and 63% (B). Neutropenia was the most common<br />

grade 3/4 hematologic toxicity (A 18%; B 29%). Severe non-hematologic<br />

toxicity was rare: fatigue (A 6%), diarrhea (B 6%). Bronchopleural fistulae<br />

occurred in 2 pts (1 per arm), and grade 3 gastrointestinal hemorrhage was<br />

seen in 1 pt (A). Conclusions: This pilot study demonstrated that bevacizumab<br />

and erlotinib could be safely added to platinum-doublet chemotherapy<br />

in the adjuvant setting in all histologies. Larger randomized studies<br />

will best define the roles <strong>of</strong> these agents in pts with resectable NSCLC.<br />

IB II IIIA<br />

A B A B A B<br />

N�24 N�25 N�19 N�19 N�11 N�8<br />

1-year DFS 93% 91% 83% 100% 100% 83%<br />

2-year DFS 84% 91% 71% 83% 75% 83%<br />

7039 General Poster Session (Board #34F), Sat, 1:15 PM-5:15 PM<br />

A prospective phase II study <strong>of</strong> induction erlotinib therapy in stage IIIA-N2<br />

non-small cell lung cancer. Presenting Author: Wenzhao Zhong, Guangdong<br />

Lung Cancer Institute, Guangdong General Hospital and Guangdong<br />

Academy <strong>of</strong> Medical Sciences, Guangzhou, China<br />

Background: Stage IIIA NSCLC represents a heterogeneous group <strong>of</strong><br />

patients with ipsilateral mediastinal (N2) lymph nodes involvement. The<br />

epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs)<br />

provide dramatic responses in patients with non-small cell lung cancer<br />

(NSCLC) carrying EGFR mutations. The purpose <strong>of</strong> this study is to evaluate<br />

the role <strong>of</strong> induction EGFR-TKI therapy in IIIA-N2 NSCLC. Methods:<br />

Patients with resectable NSCLC <strong>of</strong> stage IIIA-N2 were assigned to either<br />

induction erlotinib arm or induction gemcitabine/carboplatin(GC) arm<br />

based on the EGFR mutations analysis (NCT00600587) . Study Design:<br />

Phase II, factorial Assignment, Safety/Efficacy; Primary outcome measures:<br />

Response to Induction Therapy; Estimated enrollment: 5 cases with<br />

partial response in the erlotinib arm. Results: From January 2008 till June<br />

2011, 24 patients with IIIA-N2 NSCLC diagnosed by mediastinoscopy or<br />

PET/CT have been enrolled. Twelve cases with EGFR mutation were<br />

assigned to the erlotinib arm (30-42 days), while 12 cases harboring<br />

wild-type EGFR received the GC regimen (3 cycles). The primary end point<br />

<strong>of</strong> the response rates were 58% (7/12) for the erlotinib arm and 33% (4/12)<br />

for the GC arm (P�0.49). The pathological N2 complete response rates<br />

were 17% for the erlotinib arm and 25% for the GC arm (P�0.64). In the<br />

erlotinib arm, the most common failure model after initial therapy was<br />

distant metastases (9/10), especially brain metastases (3/9). The median<br />

progression free survival time was 7 months for the erlotinib arm and 9<br />

months for the GC arm (P�0.27). The median overall survival was 27<br />

months for the erlotinib arm and 23 months for the GC arm (P�0.52). In<br />

addition, four cases in the erlotinib arm got partial response to the 2nd<br />

EGFR-TKI therapy after progression to erlotinib induction therapy and the<br />

following thoracotomy. Conclusions: Induction erlotinib therapy in IIIA-N2<br />

NSCLC with EGFR activating mutation is a promising strategy. Distance<br />

metastases were major failure model. A multicenter, randomized, phase II<br />

study evaluating efficacy and safety <strong>of</strong> erlotinib vs GC as neoadjuvant<br />

therapy for stage IIIA-N2 NSCLC patients with EGFR mutations<br />

(NCT01407822) is currently recruiting participants.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


Lung Cancer—Non-small Cell Local-Regional/Small Cell/Other Thoracic Cancers<br />

7040 General Poster Session (Board #34G), Sat, 1:15 PM-5:15 PM<br />

Matched-pair comparison <strong>of</strong> outcome <strong>of</strong> patients with clinical stage I<br />

non-small cell lung cancer treated with resection or stereotactic radiosurgery.<br />

Presenting Author: Julia Shelkey, Penn State Hershey Medical<br />

Center, Hershey, PA<br />

Background: Stereotactic body radiotherapy (SBRT) is an alternative to<br />

surgery alone for certain patients with clinical stage I non-small cell lung<br />

cancer (NSCLC), but comparing their effectiveness is difficult because <strong>of</strong><br />

differences in patient selection and staging. Methods: Two databases were<br />

combined which contained 132 patients treated by lobectomy (LR) and 48<br />

by sublobar resection (SLR) and 137 patients managed with SBRT after<br />

negative staging between 1999-2009. We compared rates <strong>of</strong> overall<br />

survival (OS), disease-free survival (DFS), and locoregional control (LRC)<br />

between patients treated with surgery and SBRT to each other and in<br />

relation to possible prognostic factors. We then performed a matched-pair<br />

analysis comparing surgery and SBRT results. Median follow-up for the<br />

entire study population was 25.8 months. Results: On univariate analysis,<br />

OS was significantly correlated with histology, the Charlson Comorbidity<br />

Index, tumor size, aspirin use, and use <strong>of</strong> SBRT; DFS was correlated only<br />

with histology; and no variable was significantly correlated with LRC.<br />

Multivariate analysis found improved OS in patients with adenocarcinoma<br />

and those undergoing surgical resection. The NSCLC “not otherwise<br />

specified” histology was associated with poorer DFS. Overall survival was<br />

significantly poorer for SBRT patients in the matched-pair analysis than for<br />

patients treated with surgery, but DFS and LRC were not significantly<br />

different between these groups. Conclusions: Our retrospective study has<br />

demonstrated similar LRC and DFS in patients treated with SBRT or<br />

surgery, but worse OS in the former group, when patients were matched for<br />

prognostic factors. Our investigation suggests that randomized trials are<br />

needed to eliminate selection bias in treatment assignment in order to<br />

accurately compare outcomes between these approaches.<br />

7042 General Poster Session (Board #35A), Sat, 1:15 PM-5:15 PM<br />

A phase II study <strong>of</strong> cisplatin (P), S-1 (S), and concurrent thoracic<br />

radiotherapy (TRT) for locally advanced non-small cell lung cancer (LA-<br />

NSCLC): Okayama Lung Cancer Study Group trial 0501. Presenting<br />

Author: Daizo Kishino, National Hospital Organization Yamaguchi-Ube<br />

Medical Center, Ube-shi, Yamaguchi, Japan<br />

Background: We previously reported an efficacy and safety <strong>of</strong> fractionated<br />

schedule <strong>of</strong>P and docetaxel (D) (days 1, 8, 29, and 36, each) and<br />

concurrent TRT (DP-TRT) for LA-NSCLC (JCO 2010). Although the median<br />

survival time (MST: 26.3 months) was excellent, grade 3 or greater<br />

pneumonitis (10%) and esophagitis (14%) were observed and treatmentrelated<br />

death was 3%.Thus, further improvement in the safety as well as<br />

efficacy is strongly warranted. S, an oral fluoropyrimidine, is a new active<br />

agent possessing a radio-sensitizing effect. Additionally, combining S and<br />

P <strong>of</strong>fered an active and safe regimen for metastatic NSCLC. The objective<br />

<strong>of</strong> this study was to assess the efficacy and safety <strong>of</strong> S � P with concurrent<br />

TRT for LA-NSCLC. Methods: Patients with stage IIIA/IIIB, aged �75 years<br />

and PS 0-1, and without any prior chemotherapy were eligible for this<br />

study. Patients were treated with P (40 mg/m² on day 1, 8, 29 and 36) and<br />

S (40 mg/m²/dose b.i.d. on days 1-14 and 29-42) and TRT (60 Gy/30 fr<br />

over 6 weeks starting on day 1). Primary endpoint was response rate (RR),<br />

and required sample size was 48 patients. Results: Between 2006 and<br />

2009, 48 patients were enrolled (37 men; median age, 66 years; PS 0/1,<br />

36/14; IIIA/IIIB, 23/25; sq/non-sq, 22/26). <strong>Part</strong>ial response was observed<br />

in 37 patients (77%; 95% confidence interval: 63-88%). The response<br />

rate was higher in older patients (�65 yrs) than younger (�65 yrs) (89% vs.<br />

64%, p�0.041). At a median follow-up <strong>of</strong> 40 months, median progressionfree<br />

survival and MST were 9.3 months and 31.3 months, respectively. No<br />

difference in efficacy (response and survivals) was observed stratified by<br />

histology (sq vs. non-sq). Toxicities were generally mild, including G3/4<br />

neutropenia (44%), G3/4 thrombocytopenia (13%), G3 febrile neutropenia<br />

(8%) and G3 pneumonitis (4%). No one developed Gr3/4 esophagitis. No<br />

toxic deaths occurred. Conclusions: This chemoradiotherapy regimen yielded<br />

a favorable overall survival data. Also, it was well-tolerated in patients with<br />

LA-NSCLC as compared with concurrent DP-TRT therapy especially in term<br />

<strong>of</strong> TRT-related toxicities. A phase III trial <strong>of</strong> this regimen vs. DP-TRT is now<br />

planned.<br />

461s<br />

7041 General Poster Session (Board #34H), Sat, 1:15 PM-5:15 PM<br />

Amplification <strong>of</strong> fibroblast growth factor receptor type 1 gene (FGFR1) in<br />

samples from 101 NSCLC patients (pts) with squamous cell carcinoma<br />

(SCC) histology. Presenting Author: Alex Martinez Marti, Medical Oncology<br />

Department, Vall d’Hebron University Hospital, Barcelona, Spain<br />

Background: The FGFR1 gene is located in the chromosomal region 8p12<br />

and encodes a transmembrane receptor tyrosine kinase. Amplification <strong>of</strong><br />

FGFR1 has been reported in lung cancer, predominantly in SCC (in up to ~<br />

20%) and has been considered a potential target for treatment with<br />

anti-FGFR1 agents. Different methods and cut<strong>of</strong>f levels to determine<br />

FGFR1 amplification have been reported but as yet no consensus has been<br />

reached on standard method. The aim <strong>of</strong> this study is to assess FGFR1<br />

amplification determined by FISH analysis in a set <strong>of</strong> samples from<br />

101SCC pts and to explore their clinical features. Methods: Tumor samples<br />

from 101SCC pts diagnosed at our institution from August 2007 to August<br />

2011 were screened for FGFR1 amplification by FISH using the ZytoLight<br />

SPEC FGFR1/CEN8 probe. FGFR1 was considered FISH positive with a<br />

ratio � 2.2 and FISH FGFR1 was polysomic with 3 or more signals in<br />

�30% <strong>of</strong> tumor cells, any other result was considered as negative. For<br />

exploratory analyses FGFR1 amplification was considered positive if a<br />

median <strong>of</strong> 6 or more gene copies were identified and FISH were polysomic<br />

with more than 2 gene copies but less than 6. Results: Pts characteristics:<br />

median age 76 yrs (range 45-80), 91% male, 33% current smokers, 67%<br />

former smokers, stage: 8% IA/ 15% IB/ 11% IIA/ 11% IIB/ 24% IIIA, 12%<br />

IIIB/ 19% IV. With a median follow up <strong>of</strong> 48 months, the median overall<br />

survival was 18 months. FISH FGFR1 was positive (ratio � 2.2) in 7 (6.9%)<br />

pts: 6 were male, 4 former smokers. FISH FGFR1 was considered negative<br />

but polysomic (3 or more signals in �30% <strong>of</strong> tumor cells) in 43/94 (45%)<br />

pts. If we use for FISH positivity a cut<strong>of</strong>f <strong>of</strong> 6 or more copies only 3 patients<br />

were considered as positive for FGFR1 amplification (2 were also FISH<br />

positive by ratio�2.2). All those 5 patients considered FISH negative by<br />

gen copy number (but positive by ratio) were polysomic. Conclusions: In our<br />

experience FGFR1 amplification detected by FISH isrelatively uncommon<br />

in SCC, although a relevant proportion <strong>of</strong> FGFR1 FISH negative tumors had<br />

polysomy. Standarization <strong>of</strong> methods to determine FGFR1 amplification<br />

and the potential clinical significance <strong>of</strong> the presence <strong>of</strong> FGFR1 polysomy<br />

are needed.<br />

7043 General Poster Session (Board #35B), Sat, 1:15 PM-5:15 PM<br />

Predictors for locoregional recurrence for clinical stage III-N2 non-small<br />

cell lung cancer with nodal downstaging after induction chemotherapy and<br />

surgery. Presenting Author: Feiran Lou, Memorial Sloan-Kettering Cancer<br />

Center, New York, NY<br />

Background: Pathologic downstaging following induction chemotherapy in<br />

patients with stage III-N2 NSCLC is a well-known positive prognostic<br />

indicator. However, the predictive factors for locoregional recurrence (LRR)<br />

in these patients are largely unknown. Methods: Between 1998-2008, 153<br />

patients with clinically or pathologically-staged III-N2 NSCLC from two<br />

cancer centers in the United States were treated with induction chemotherapy<br />

and surgery. All patients had pathologic N0-1 disease, and no one<br />

received postoperative radiotherapy. LRR were defined as disease recurrence<br />

at the surgical site, lymph nodes (levels 1-14 including supraclavicular)<br />

or both. Overall survival (OS) was calculated using the Kaplan and<br />

Meier method and comparisons were made using the log-rank test.<br />

Univariate and multivariate Cox proportional hazards model were used to<br />

assess the association <strong>of</strong> LRR and risk factors. Results: The median follow<br />

up time for survivors was 39.3 months. Baseline pretreatment N2 nodal<br />

involvement was staged by either pathologic confirmation (18.2%) or<br />

PET/CT (81.8%). Overall, the 5 year LRR rate was 30.8% (n�38), with<br />

LRR being the first site <strong>of</strong> failure in 51% (22 <strong>of</strong> 43). The 5 year OS for<br />

patients with LRR compared to those without was 21% versus 60.1%,<br />

respectively (p�0.001). Using multivariate analysis, significant predictor<br />

for LRR was pN1 versus pN0 disease at time <strong>of</strong> surgery (p�0.001, HR<br />

3.43, 95% CI 1.80-6.56) and trended for squamous histology (p�0.072,<br />

HR 1.93, 95% CI 0.94-3.98). The 5-year LRR rate for N1 versus N0<br />

disease was 62% versus 20%, respectively. Neither single versus multistation<br />

N2 disease (p�0.291) nor initial staging by mediastinoscopy versus<br />

PET/CT (p�0.306) were predictors for LRR. We found that N1 status was<br />

also predictive for higher distant recurrence rate (p�0.021, HR 1.91, 95%<br />

CI 1.10-3.30) but only trended for poorer OS (p�0.123, HR 1.48, 95% CI<br />

0.90-2.44). Conclusions: LRR remains high in resected stage III-N2<br />

NSCLC patients after induction chemotherapy and nodal downstaging,<br />

particularly in patients with persistent N1 disease. Postoperative radiotherapy<br />

may be needed for these high-risk patients.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


462s Lung Cancer—Non-small Cell Local-Regional/Small Cell/Other Thoracic Cancers<br />

7044 General Poster Session (Board #35C), Sat, 1:15 PM-5:15 PM<br />

Higher incidence <strong>of</strong> EGFR exon 19 deletion in younger (age 40 or younger)<br />

patients with adenocarcinoma <strong>of</strong> the lung. Presenting Author: Eri Sugiyama,<br />

Division <strong>of</strong> Thoracic Oncology, National Cancer Center Hospital East,<br />

Kashiwa, Chiba, Japan<br />

Background: The proportion <strong>of</strong> younger patients (� 40 years) with lung<br />

cancer is reported to be 2-5%. The most frequent histologic type <strong>of</strong> them is<br />

adenocarcinoma, however little is known about the pathological and<br />

molecular characteristics <strong>of</strong> younger patients with lung adenocarcinoma.<br />

Methods: Between July 1992 and April 2011, a total <strong>of</strong> 7443 patients were<br />

diagnosed as lung cancer in National Cancer Center Hospital East and 165<br />

patients <strong>of</strong> whom (2.2%) who were 40 years or younger were identified.<br />

Among them, 44 patients with adenocarcinoma who underwent surgical<br />

resection were selected for this study. In addition, 185 elderly patients with<br />

� 40 years who underwent surgical resection matching gender and<br />

smoking status were selected as a control group. Histological predominant<br />

growth pattern and any coexisting variant pattern, the status <strong>of</strong> EGFR<br />

mutations were compared between these two groups. Results: The median<br />

age in � 40 years patients was 37 years (range, 21 to 40 years) and that in<br />

elderly patients was 68 years (range, 42 to 83 years). Between these two<br />

groups, there were no significant differences in the distribution <strong>of</strong> histological<br />

predominant growth patterns (lepidic; 31.8% vs. 26.5%, papillary;<br />

34.1% vs. 37.3%, acinar; 9.1% vs. 16.2%, and solid; 25.0% vs. 20.0%,<br />

p�0.78) and the incidence <strong>of</strong> EGFR mutations (40.9% vs. 45.9%;<br />

p�0.55). However, signet-ring cell component were significantly found in<br />

the younger patients than elderly (11.4% vs. 0%; p�0.01). The incidence<br />

<strong>of</strong> EGFR exon 19 deletion was significantly higher in younger patients than<br />

elderly, in contrast, that <strong>of</strong> EGFR exon 21 L858R was significantly higher in<br />

elderly patients (exon 19 del; 31.7% vs. 18.9%, L858R; 4.6% vs. 25.4%,<br />

p�0.0091). Three <strong>of</strong> 17 adenocarcinomas (17.7%) with EGFR-wild type<br />

in younger patients showed positive for ALK translocation. Conclusions:<br />

Younger patients with lung adenocarcinoma showed significantly higher<br />

proportion <strong>of</strong> EGFR exon 19 deletion genotype and containing histologically<br />

signet-ring cell component comparing with elderly patients. EGFR<br />

exon 19 deletion genotype may be related to pathogenesis <strong>of</strong> lung<br />

adenocarcinoma in younger patients.<br />

7046 General Poster Session (Board #35E), Sat, 1:15 PM-5:15 PM<br />

Molecular pr<strong>of</strong>iling <strong>of</strong> patients with non-small cell lung cancer (NSCLC)<br />

using bronchoscopic ultra-micro sampling. Presenting Author: Yuichi<br />

Sakairi, Department <strong>of</strong> General Thoracic Surgery, Graduate School <strong>of</strong><br />

Medicine, Chiba University, Chiba, Japan<br />

Background: Genetic information is essential for molecular targeted therapy<br />

for personalized medicine, although tissue sampling for genetic analysis<br />

remains challenging. In this study, we investigated the utility <strong>of</strong> bronchoscopic<br />

ultra-micro samples compared with conventional histological materials<br />

for multiple gene analyses for NSCLC. Methods: Patients with NSCLC<br />

proven by on-site cytological examinations during bronchoscopic survey<br />

were eligible. Following conventional needle aspiration biopsy by flexible<br />

bronch<strong>of</strong>iberscope (from primary lesion) or convex probe endobronchial<br />

ultrasound (from lymph nodes), the used needle was rinsed with 20 ml <strong>of</strong><br />

saline and this ultra-micro sample (MS) was used for both cytological<br />

diagnosis and genetic analysis. DNA and RNA were extracted from each<br />

sample. Gene mutations [EGFR (epidermal growth factor receptor), K-ras,<br />

BRAF] and anaplastic lymphoma kinase (ALK) fusion genes were examined<br />

by high resolution melting analysis and direct sequencing. The results <strong>of</strong><br />

cytological diagnoses and gene pr<strong>of</strong>iles using MS were compared to<br />

conventional histological diagnoses. Results: A total <strong>of</strong> 79 lesions (30<br />

primary and 49 metastatic nodes) from 76 patients were eligible. Adenocarcinoma<br />

(N � 46), squamous cell carcinoma (N � 25), and NSCLC (N � 8)<br />

were pathologically confirmed in histological lesion cores; however, only 29<br />

malignancies (37%) were detected with MS. DNA and RNA could be<br />

extracted from all histological samples and MS. In 35 cases, genetic<br />

disorders were identified, including EGFR mutations (N � 12), K-ras<br />

mutations (N � 5), BRAF mutation (N � 1), ALK fusion genes (N � 3), and<br />

silent mutations (N � 13); these results were identical for both histological<br />

samples and MS. Conclusions: Bronchoscopic ultra-micro samples (biopsy<br />

needle rinse fluids) are useful for multiple genetic examinations.<br />

7045 General Poster Session (Board #35D), Sat, 1:15 PM-5:15 PM<br />

A phase II trial <strong>of</strong> induction gefitinib monotherapy followed by cisplatindocetaxel<br />

and concurrent thoracic irradiation in patients with EGFRmutant<br />

locally advanced non-small-cell lung cancer (LA-NSCLC):<br />

LOGIK0902/OLCSG0905 intergroup trial. Presenting Author: Akiko<br />

Hisamoto, Department <strong>of</strong> Respiratory Medicine, Okayama University Hospital,<br />

Okayama, Japan<br />

Background: We previously reported efficacy and safety <strong>of</strong> cisplatindocetaxel<br />

and concurrent thoracic radiotherapy (TRT) for LA-NSCLC<br />

(Segawa Y and Kiura K. JCO 2010). However, its cure rate remains<br />

unsatisfied, and further improvement in the treatment outcome is strongly<br />

warranted. Recently, systemic chemotherapy has been individualized by<br />

intensive anti-cancer researches through the discovery <strong>of</strong> certain molecular<br />

targets in the metastatic NSCLC. Especially, gefitinib, EGFR tyrosine<br />

kinase inhibitor, is quite active and now one <strong>of</strong> the standard chemotherapy<br />

for untreated metastatic EGFR-mutant NSCLC (Maemondo M and Inoue A.<br />

NEJM 2010). Even in the locally advanced setting, approximately 30% <strong>of</strong><br />

Japanese NSCLC patients possess EGFR-mutant tumors. Given all <strong>of</strong> these<br />

backgrounds, investigation on the role <strong>of</strong> adding gefitinib monotherapy to<br />

the standard concurrent chemoradiotherapy might cause a new paradigm<br />

shift in this setting. Methods: Patients have to meet the following eligibility<br />

criteria: LA-NSCLC; active tumor EGFR mutations (exons 19 and 21) but<br />

without 790M; measurable lesions; performance status (PS) <strong>of</strong> 0 or 1; age<br />

� 75 years; and V20 � 35%. The primary endpoint is set as 2-year<br />

progression-free survival (PFS) rate; assuming that 75% in eligible patients<br />

would indicate potential usefulness, whereas 60% would be the lower limit<br />

<strong>of</strong> interest (� � 0.05, ß � 0.20), the estimated accrual number is 46<br />

patients. The secondary endpoint includes toxicity, response rate and<br />

overall survival. Patients will receive the induction treatment, specified as<br />

gefitinib monotherapy (250 mg/body) for 8 weeks. Patients who have not<br />

progressed during the induction therapy will receive cisplatin and docetaxel<br />

(40 mg/m2 ; days 1, 8, 29, 36, each) and concurrent three-dimensional<br />

conformal TRT (2-Gy, single, daily fractions for 5 consecutive days each<br />

week to provide a total dose <strong>of</strong> 60 Gy). Enrollment began in 2010, and will<br />

complete by 2015. UMIN registration number <strong>of</strong> 000005086.<br />

7047 General Poster Session (Board #35F), Sat, 1:15 PM-5:15 PM<br />

Relationship between tumor size and survival in non-small cell lung cancer<br />

(NSCLC): An analysis <strong>of</strong> the Surveillance, Epidemiology, and End Results<br />

(SEER) registry. Presenting Author: Jianjun Zhang, University <strong>of</strong> Texas<br />

M. D. Anderson Cancer Center, Houston, TX<br />

Background: Tumor size is a known prognostic factor for early stage (I and II)<br />

NSCLC, but its significance in node positive or locally advanced NSCLC has<br />

not been determined. We sought to evaluate its prognostic value in early<br />

and locally advanced NSCLC and create a nomogram incorporating tumor<br />

size and other prognostic variables to predict survival. Methods: The SEER<br />

registry was queried for patients (pts) with NSCLC, aged 20-103 and<br />

diagnosed between 1998 and 2003. Pts with extra-pulmonary metastases<br />

or distant lymph node metastases were excluded. Tumor size was analyzed<br />

as a continuous variable. Other demographic variables included age,<br />

gender, race, histology, primary tumor extension, node status and primary<br />

treatment modality (surgery vs radiation). Log-rank test was performed to<br />

evaluate the relationship between these variables and overall survival (OS).<br />

Cox proportional hazard model was used to evaluate whether tumor size was<br />

an independent prognostic factor. Results: 52,287 eligible pts were divided<br />

into 16 subgroups based on primary tumor extension and node status. For<br />

example, in group 1, tumor was confined to one lung with no nodes<br />

involved; pts in group 12 had tumor invading the mediastinum (T4 by<br />

extent) and positive ipsilateral mediastinal nodes. Tumor size had a<br />

significant effect on OS in almost all groups after adjustment for age, sex,<br />

race, histology, node status, primary tumor extension and primary treatment<br />

modality. Our model incorporating tumor size had significantly better<br />

predictive accuracy (larger C index) than our alternative model not<br />

including this information (p�0.0001). We then developed a nomogram<br />

incorporating tumor size, age, gender, race, histology, nodule stage, and<br />

tumor extension with the intent to predict OS. In subsequent bootstrap<br />

verification, the predicted 2-year OS from the nomogram was almost<br />

identical to the actual observed 2-year OS with a very slim biases <strong>of</strong><br />

estimates. Conclusions: Tumor size is an independent prognostic factor,<br />

including pts with node positive or locally advanced NSCLC. We successfully<br />

created a nomogram incorporating tumor size and other clinical<br />

variables to predict survival.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


Lung Cancer—Non-small Cell Local-Regional/Small Cell/Other Thoracic Cancers<br />

7048 General Poster Session (Board #35G), Sat, 1:15 PM-5:15 PM<br />

Breath analysis for early detection <strong>of</strong> lung cancer. Presenting Author:<br />

Geetha D. Vallabhaneni, Winship Cancer Institute, Emory University,<br />

Atlanta, GA<br />

Background: Lung cancer (LC) is the leading cause <strong>of</strong> cancer death<br />

worldwide. Early detection is critical to reduce LC related mortality. Breath<br />

analysis can be used as a non invasive diagnostic tool in conjunction with<br />

CT screening. We assessed the diagnostic value <strong>of</strong> breath volatile organic<br />

compounds (BVOCs) in the alveolar breath <strong>of</strong> female non-small cell lung<br />

cancer (NSCLC) patients. Methods: This is a prospective case-control study<br />

that enrolled newly diagnosed NSCLC and cancer-free female controls. The<br />

study aims: 1) To compare the BVOC signature in female NSCLC patients to<br />

a control group without LC 2) To compare the BVOC signature between<br />

NSCLC and breast cancer (BC) patients. The study was restricted to women<br />

because the pilot data was generated from female subjects with BC.<br />

Consenting eligible patients provided a total <strong>of</strong> five deep breath samples<br />

into a proprietary breath sampler developed at the Georgia Tech Research<br />

Institute (GTRI) for collecting alveolar breath. Breath samples were<br />

collected five minutes apart from each subject. The collected samples were<br />

analyzed for BVOCs using thermal desorption/gas chromatographic/mass<br />

spectrometric (TD/GC/MS) technique. Statistical differences in BVOCs in<br />

breath samples from cases and controls were assessed using this technique.<br />

Support vector machine classification method was employed to<br />

compare the BVOCs pattern between LC and BC patients. Results: We<br />

enrolled 25 NSCLC and 25 cancer-free subjects. TD/GC/MS analysis<br />

identified a total <strong>of</strong> 422 BVOCs among the controls and NSCLC subjects.<br />

75 unique BVOCs that were significantly different between cases and<br />

controls were employed for statistical modeling. This 75 BVOCs signature<br />

has a sensitivity <strong>of</strong> 0.75, a specificity <strong>of</strong> 0.75 and a classification accuracy<br />

<strong>of</strong> 75% for NSCLC and controls. Statistical analysis on the full BVOCs data<br />

set from NSCLC and previously collected BVOCs data from BC patients<br />

achieved a classification accuracy <strong>of</strong> 88%. Conclusions: Unique BVOCs<br />

pattern can identify patients with established NSCLC. Future studies in<br />

at-risk patients will assess the utility <strong>of</strong> this approach as a less invasive and<br />

minimal risk screening tool for early detection <strong>of</strong> NSCLC. Supported by the<br />

Kennedy Seed Grant awarded by the Winship Cancer Institute <strong>of</strong> Emory<br />

University.<br />

7050 General Poster Session (Board #36A), Sat, 1:15 PM-5:15 PM<br />

Case-control study <strong>of</strong> prophylactic cranial irradiation in nonmetastatic<br />

non-small cell lung cancer. Presenting Author: Simon Cheng, Department<br />

<strong>of</strong> Radiation Oncology, Columbia University Medical Center, New York, NY<br />

Background: Prophylactic cranial irradiation (PCI) reduces the incidence <strong>of</strong><br />

brain metastases in NSCLC patients after primary therapy, but its impact<br />

on survival is uncertain. We report on the largest study <strong>of</strong> survival in<br />

patients treated with and without PCI for non-small cell lung cancer<br />

(NSCLC). Methods: We reviewed 17 Surveillance, Epidemiology and End<br />

Results (SEER) registries for a retrospective study on patients who had PCI<br />

as part <strong>of</strong> their primary treatment for NSCLC from 1988 - 97. Cases were<br />

limited to those with non-metastatic (Stage I-III) NSCLC. To balance the<br />

cohorts, we matched each PCI patient with four non-PCI patients on stage,<br />

histology, race and sex. Associations between treatment type, clinical<br />

factors, and demographics were assessed using the Chi-squared test.<br />

Survival time was calculated as the number <strong>of</strong> months from diagnosis to the<br />

date <strong>of</strong> death. Survival was censored as <strong>of</strong> the last month when patients<br />

were known to be alive. Overall (OS) and cancer cause-specific survival<br />

(CSS) were investigated using the Kaplan-Meier, competing risks, Cox<br />

proportional hazards, and log-rank tests. Results: We found 472 PCI<br />

matched to 1,888 non-PCI patients. Characteristics were balanced across<br />

groups: race (p � 1.00), sex (p � 0.95), histology (p � 1.00), stage (p �<br />

1.00), and surgery (p � 0.81). PCI group was younger, median age 64 vs<br />

68 (p � 0.01). PCI vs no PCI median OS was 8 vs 10 months (p � 0.01).<br />

OS was 14% vs 28% at 2 years and 5% vs 12% at 5 years, PCI vs no PCI<br />

respectively (p � 0.01). Stage III OS was also different; 10% vs 21% at 2<br />

years, PCI vs no PCI respectively (p � 0.01). Median CSS was the same at 9<br />

months in both groups. Median follow-up was 14 years. Conclusions: PCI<br />

was not associated with improved OS or CSS in these NSCLC patients, and<br />

PCI may have a detrimental effect on OS. In limited-stage small cell lung<br />

cancer, a retrospective SEER analysis during the 1988 – 97 period showed<br />

a survival benefit in treated patients with PCI, which has been confirmed<br />

with prospective studies. To date, 4 prospective trials examining PCI for<br />

NSCLC have shown a reduced incidence <strong>of</strong> brain metastases, but the effect<br />

on survival is unclear. Further investigation is needed to determine whether<br />

PCI for NSCLC increases the risk <strong>of</strong> other causes <strong>of</strong> death.<br />

463s<br />

7049 General Poster Session (Board #35H), Sat, 1:15 PM-5:15 PM<br />

Outcomes <strong>of</strong> combined modality treatment in elderly patients with stage III<br />

non-small cell lung cancer. Presenting Author: Ludimila Cavalcante, Mount<br />

Sinai Medical Center, Miami Beach, FL<br />

Background: The use <strong>of</strong> combined modality treatment (CMT) has been<br />

demonstrated to improve survival in patients with inoperable, locally<br />

advanced NSCLC. Elderly patients have been traditionally excluded from<br />

prospective clinical trials investigating CMT and thus the optimal treatment<br />

strategy for these patients remains unclear. We retrospectively evaluated<br />

the outcomes <strong>of</strong> elderly patients who received concurrent and sequential<br />

chemoradiation. Methods: All lung cancer patients age 70 or older who<br />

received chemotherapy and radiation treatment at Mt. Sinai Medical Center<br />

for stage III NSCLC between 1997 and 2010 were analyzed from tumor<br />

registry data. Among 282 lung cancer elderly patients diagnosed with stage<br />

III disease, 64 patients (22.7 %) received CMT as part <strong>of</strong> their treatment.<br />

Patients who underwent surgery and palliative radiation were excluded.<br />

Statistical analysis was performed by log-rank, Kaplan-Meier methods, and<br />

t-test. Results: Median age at diagnosis for the CMT group was 75 years<br />

(70-87), male/female: 34/30, TNM: T4N0-1: 12, T3N1: 1, TXN2: 41,<br />

TXN3: 10. Concurrent chemoradiation was delivered in 43 cases and<br />

sequential in 21 cases. RT doses ranged from 50 to 63 Gy. The most<br />

common chemotherapy regimens used were carboplatin and taxol (44 %)<br />

and carboplatin and etoposide (15%). The most common treatment-related<br />

toxicities were esophagitis (42%), anemia (39%), and pneumonia (24%).<br />

Median survival (MST) was 19 months and 11 months in the concurrent<br />

and sequential chemoradiation groups (p�0.67). Observed two, three and<br />

five-year overall survival (OS) in the CMT group was 49.1%, 27.5%, and<br />

12.5%, respectively. Conclusions: Although CMT is not commonly used in<br />

elderly patients with lung cancer, the treatment is feasible and beneficial<br />

for selected patients in the community setting. Interestingly, the MST <strong>of</strong><br />

elderly patients who received CMT in this cohort was comparable to that <strong>of</strong><br />

younger historical controls with CMT. There was also a trend for increased<br />

survival in the concurrent versus sequential chemoradiation group.<br />

7051 General Poster Session (Board #36B), Sat, 1:15 PM-5:15 PM<br />

Correlation <strong>of</strong> pretreatment surgical staging and PET SUVmax with outcomes<br />

in NSCLC. Presenting Author: Giancarlo Moscol, Albert Einstein<br />

Medical Center, Philadelphia, PA<br />

Background: The AJCC staging does not recognize the number <strong>of</strong> involved<br />

mediastinal lymph nodes (LN), nodal stations or PET findings as prognostic<br />

factors in non-small cell lung cancer (NSCLC). Most clinical studies report<br />

poorer survival with greater number <strong>of</strong> involved LN and maximal tumor<br />

standardized uptake value (SUVmax) � 5. We hypothesized that clinical<br />

outcomes can be predicted by (1) number <strong>of</strong> involved LN, (2) number <strong>of</strong><br />

involved nodal stations, (3) SUVmax <strong>of</strong> primary tumor and (4) SUVmax <strong>of</strong><br />

mediastinum. Methods: Records <strong>of</strong> patients diagnosed with NSCLC stages<br />

I-IIIA from 1/1/99 to 12/31/10 were reviewed. Data collected included age,<br />

type <strong>of</strong> surgery, AJCC stage, number <strong>of</strong> LN/stations sampled and SUVmax<br />

<strong>of</strong> primary tumor/mediastinum. Primary outcomes were tumor recurrence<br />

and death. Descriptive statistics were used to summarize demographics.<br />

Prognostic factors were analyzed by a multivariate analysis using Cox’s<br />

proportional hazard model. Survival was estimated by the Kaplan-Meier<br />

method. Results: There were 369 patients with complete clinical data; 207<br />

underwent surgical staging with mediastinoscopy (62) or VATS/open<br />

thoracotomy (145). The median number <strong>of</strong> LN sampled was 9 (range 1-35)<br />

and the median number <strong>of</strong> stations biopsied was 3 (range 1-9). PET was<br />

performed in 89 patients; 59 scans showed tumor SUVmax � 5. A total <strong>of</strong><br />

65/207 patients received chemotherapy. With median follow-up time <strong>of</strong> 29<br />

months (range 1-116), 73 recurrences and 90 deaths were observed. Cox<br />

analysis showed greater recurrence risk for patients with 4 or more involved<br />

LN (HR� 4.66, CI�1.19-18.18, p�0.027) and higher mortality for<br />

patients with tumor SUVmax � 5 (HR�14.83, CI�2.32-94.9, p�0.004).<br />

Outcomes did not correlate with the number <strong>of</strong> involved nodal stations<br />

(p�0.76 for recurrence, p�0.51 for mortality) or with SUVmax <strong>of</strong> mediastinum<br />

(p�0.17 for recurrence, p�0.93 for mortality). Conclusions: The<br />

number <strong>of</strong> involved LN was an independent prognostic factor for recurrence<br />

but not for mortality. A tumor SUVmax �5 was independently associated<br />

with greater mortality. The number <strong>of</strong> involved stations and the SUVmax <strong>of</strong><br />

mediastinum did not correlate with worse outcomes. Our results are limited<br />

due to the small number <strong>of</strong> cases with PET/CT scans and short follow-up.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


464s Lung Cancer—Non-small Cell Local-Regional/Small Cell/Other Thoracic Cancers<br />

7052 General Poster Session (Board #36C), Sat, 1:15 PM-5:15 PM<br />

5-day dosing schedule <strong>of</strong> temozolomide in relapsed sensitive or refractory<br />

small cell lung cancer (SCLC) and methyl-guanine-DNA methyltransferase<br />

(MGMT) analysis in a phase II trial. Presenting Author: Alexander Edward<br />

Dela Cruz Drilon, Memorial Sloan-Kettering Cancer Center, New York, NY<br />

Background: MGMT promoter methylation and loss <strong>of</strong> MGMT activity are<br />

associated with improved sensitivity to alkylating agents. We recently<br />

reported that the oral alkylating agent temozolomide is active in 2nd- and<br />

3rd-line treatment <strong>of</strong> relapsed SCLC at 75mg/m2 /day for 21 out <strong>of</strong> 28 days<br />

(Pietanza et al, Clin Can Res 2012). Here we evaluate the 5-day dosing<br />

schedule <strong>of</strong> temozolomide in a second cohort <strong>of</strong> patients and analyze<br />

MGMT activity in both cohorts <strong>of</strong> patients on the same study. Methods:<br />

Patients with disease progression after 1 or 2 prior chemotherapy regimens<br />

received temozolomide at 200mg/m2 /day for 5 out <strong>of</strong> 28 days (n�25).<br />

Those with sensitive (S-SCLC, n�16) and refractory (R-SCLC, n�9)<br />

disease were enrolled to assess safety. Available tumor tissue from patients<br />

treated according to either schedule was assessed for MGMT promoter<br />

methylation status by PCR and MGMT expression by immunohistochemistry<br />

(IHC). Results: An overall response rate <strong>of</strong> 12% was noted (3 partial<br />

responses: 1 S-SCLC, 2 R-SCLC). 4 patients had stable disease for at least<br />

3 cycles. Median progression-free survival and overall survival for all<br />

patients were 1.3 months and 7.9 months, respectively. Grade �3<br />

thrombocytopenia and neutropenia was observed in 20% with a shorter<br />

mean duration <strong>of</strong> myelosuppression compared to the 21-day schedule.<br />

Results from MGMT evaluation <strong>of</strong> tumor samples from both dosing<br />

schedules were combined. Promoter methylation <strong>of</strong> MGMT was not<br />

significantly associated with response (p�0.23). However, patients that<br />

lacked MGMT expression by IHC had a higher response rate compared to<br />

those with MGMT-positive tumors (43% vs. 13%, p � 0.052; see table).<br />

Conclusions: The 5-day dosing schedule <strong>of</strong> temozolomide is both active and<br />

safe in patients with relapsed SCLC. A strong trend toward increased<br />

response was demonstrated in patients with MGMT-negative tumors<br />

compared to patients with MGMT-positive tumors by IHC. A trial combining<br />

temozolomide with the PARP inhibitor, ABT888, is planned.<br />

MGMT expression by IHC (n�38) PR<br />

Response<br />

SD � POD p<br />

Negative (n � 14) 43% (n�6) 57% (n�8) 0.052<br />

Positive (n � 24) 13% (n�3) 88% (n�21)<br />

7054 General Poster Session (Board #36E), Sat, 1:15 PM-5:15 PM<br />

Are the presenting characteristics and prognosis <strong>of</strong> primary salivary<br />

gland-type lung cancers (SG) similar to those <strong>of</strong> other non-small cell lung<br />

cancers (NSCLC)? Presenting Author: John M. Varlotto, Penn State Hershey<br />

Cancer Institute, Hershey, PA<br />

Background: SG, both adenoid cystic (ACC) and mucoepidermoid (ME)<br />

sub-types, are rare lung cancers. We choose to investigate the incidence <strong>of</strong><br />

these rare sub-types and assess their difference in presentation and<br />

prognostic characteristics in comparison to adenocarcinomas (Ad) and<br />

squamous cell carcinomas (SCC) during the same time period. Methods:<br />

The SEER-17 database was used to collect data during the years 1988-<br />

2008. Differences between populations were determined by the chi-square<br />

test. Survival curves were generated as Kaplan-Meier techniques. Cox<br />

proportional hazards test was used to compare survival differences. Results:<br />

During the 20-year study period, ACC (n �100) and ME (n� 178)<br />

accounted for 0.03% and 0.06% <strong>of</strong> NSCLCs. Mean follow-up was 34.5<br />

months for all patients. In comparison to ACC, patients with ME were<br />

significantly more likely to be younger (52 yr vs. 60yr), Asian(11.7% vs.<br />

7%), have Stage I disease (62.9% vs 24.0%), and less likely to be in the<br />

mainstem bronchi (17.2% vs. 6.3%). In comparison to patients with<br />

patients presenting with either SCC or Ad, both ME and ACC were<br />

significantly less likely to present with Stage IV disease (26.6% SCC,<br />

41.29% Ad, 16.73% SG), have nodal involvement (35.1% SCC, 27.4%<br />

Ad, 23.37% SG), and be older (70 SCC, 68 Ad, 58 years SG). Stratified by<br />

stage and treatment, there was no survival (OS) or disease-specific survival<br />

difference (DSS) between ACC and ME. The OS <strong>of</strong> the combined group <strong>of</strong><br />

ME and ACC was significantly better stage per stage than either Ad (Hazard<br />

ratio (HR) range � 0.26- 041), and SCC (HR range � 0.17-0.56). Lung<br />

Cancer-Specific survival at 2,3,5 years for surgically-resected Stage I ACC<br />

and ME were 83.5%, 80.4%, and 80.4%; and 82.6%, 78.0% and 78%,<br />

respectively. Conclusions: Patients with ACC and ME have rare sub-types <strong>of</strong><br />

lung cancer that present differently and have better survival than patients<br />

presenting with either <strong>of</strong> the more common histologic sub-types (SCC and<br />

Ad) <strong>of</strong> NSCLC. We encourage prospective, multi-institutional studies <strong>of</strong><br />

these rare sub-types so that care can be optimized. Optimal care may differ<br />

for SG because <strong>of</strong> their stage per stage better prognosis than other NSCLCs.<br />

7053 General Poster Session (Board #36D), Sat, 1:15 PM-5:15 PM<br />

High-throughput targeted resequencing <strong>of</strong> lung adenocarcinoma. Presenting<br />

Author: Byung Chul Kim, Personal Genomics Institute, Genome<br />

Research Foundation, AICT, Suwon, South Korea<br />

Background: The mutational pr<strong>of</strong>iles are crucial for cancer diagnosis and<br />

classification, which lead to tailoring the best therapeutic strategy to each<br />

patient. Here, we present target sequencing <strong>of</strong> 30 hot spot genes in lung<br />

adenocarcinoma to identify new hot spot mutations in never-smoker,<br />

female lung adenocarcinoma patients. Methods: We targeted 30 genes that<br />

are known to be mutated frequently in lung adenocarcinoma. The exome<br />

capture was done using selector technology and sequencing was done using<br />

Illumina GA IIx Genome Analyzer. Exomic short reads from Illumina<br />

Pipeline 1.4 were aligned to human genome (NCBI build 36) with BWA<br />

0.5.0. Variations and small indels were called by Samtools 0.1.7 and<br />

filtered by custom R scripts. Potential effects <strong>of</strong> these identified alterations<br />

were assessed with sequence analysis. Results: On average, 1.5 billion<br />

bases were uniquely mapped, and mean depth and coverage <strong>of</strong> exon regions<br />

were 38X and 96%, respectively. Filtering <strong>of</strong> data against public SNP<br />

database (dbSNP130), resulted in ~2,000 novel genetic alterations per<br />

sample, including single nucleotide variations, and small insertion and<br />

deletions in protein-coding regions. Among them, there were novel variants<br />

in LTK and EPHA5 genes. Conclusions: Our results demonstrated the<br />

feasibility <strong>of</strong> high-throughput mutation pr<strong>of</strong>iling with lung adenocarcinoma<br />

samples. We identified novel variants in hot target genes, which may<br />

provide an insight into the tumorigenesis signaling <strong>of</strong> lung adenocarcinoma.<br />

7055 General Poster Session (Board #36F), Sat, 1:15 PM-5:15 PM<br />

Would screening benefit 75- to 84-year-old patients with non-small cell<br />

lung cancer (NSCLC)? Presenting Author: Jessica Lake, Penn State College<br />

<strong>of</strong> Medicine, Hershey, PA<br />

Background: The Lung Cancer Screening Trial has shown an overall survival<br />

(OS) benefit and reduced lung cancer mortality in the 55-74 age group<br />

(gp). We chose to evaluate whether NSCLC patients aged 75-84 are<br />

increasing in the USA and whether they would benefit from aggressive<br />

therapy. Methods: SEER-17 was used to calculate NSCLC rates during the<br />

years 2000-2008. SEER-9 was used to estimate the proportional change in<br />

both 55-74 and 75-84 gp from 1973-2008. OS was analyzed in a modern<br />

population from SEER-17 (2004-2008) to assess the effects <strong>of</strong> increasingly<br />

aggressive therapy (observation(Ob), radiation (RT) or lobectomy<br />

(LB)) for a proposed screening population with T1N0 tumors. Chi-square<br />

test and Cox Regression (CR) were used to evaluate OS. Paired T-tests<br />

assessed changes in rates and proportions over time. Results: The 55-74 gp<br />

rose from 64.4% in 1973 to 67.25% in 1984, but fell to 58.8% by 2008,<br />

while the 75-84 gp rose from 12.1% in 1973 to 24% in 2008 (p�0.01),<br />

similar in both sexes. The rates/100,000 have been increasing in the<br />

75-84 gp (p�0.02), mainly in females (p�0.003) while the rates in the<br />

55-74 gp did not vary, but fell for men (p�0.03). In the Ob gp (n�1344),<br />

NSCLC was the most common cause <strong>of</strong> death (COD) in the 55-74 (29.8%)<br />

and 75-84 gp (40.6%), more than all other CODs combined (median<br />

survival (MS) � 11mn). CR revealed that OS was associated with the 55-74<br />

gp (0.59) and females (0.62) (p � 0.001). In the RT gp (n�1870), MS was<br />

14mn and lung cancer was the most common COD at 27.7% (55-74) and<br />

28.8% (75-84), again more than all other CODs combined. CR found that<br />

females (0.68) and black race (0.72) had better OS (p�0.017), but age<br />

was not. MS was 24 mn in the LB group (n�9384). COD from NSCLC and<br />

all other CODs was 8.2% and 6.1% (55-74) and 10.9% and 11.4%<br />

(75-84). CR showed that 55-74 (0.36), females (0.58), and Asians (0.73)<br />

had lower death rates (all p�0.015). Mean OS between the 55-74 (26.0)<br />

and 75-84 (24.2) gp showed a small yet significant difference. Conclusions:<br />

Rates and proportions <strong>of</strong> NSCLC have been steadily increasing in the 75-84<br />

gp. These data show that COD by lung cancer decreased significantly with<br />

increasingly aggressive treatment and treatment reduced the effects <strong>of</strong> age<br />

gp on survival. We feel that screening may be <strong>of</strong> benefit to the 75-84 gp.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


Lung Cancer—Non-small Cell Local-Regional/Small Cell/Other Thoracic Cancers<br />

7056 General Poster Session (Board #36G), Sat, 1:15 PM-5:15 PM<br />

Prognostic value <strong>of</strong> MAGE-A3 and PRAME gene expression in non-smallcell<br />

lung cancer (NSCLC). Presenting Author: Albert Linder, Lungenklinik<br />

Hemer, Hemer Nordrhein-Westfalen, Germany<br />

Background: We investigated the expression <strong>of</strong> some cancer–testis genes<br />

and their association with disease prognosis. Here we report our results for<br />

the expression, in resected NSCLC samples, <strong>of</strong> two tumor-specific antigens<br />

(Ags): MAGE-A3 and PRAME, both under evaluation in clinical trials.<br />

Methods: We conducted a single-center, uncontrolled, retrospective study<br />

<strong>of</strong> tissue from resected stage I–III NSCLC patients (pts): 650 were in stage<br />

I, 215 in stage II and 395 in stage III. 1260 FFPE tumor tissue samples<br />

from the tumor tissue bank <strong>of</strong> the Department <strong>of</strong> Pathology, Lungenklinik<br />

Hemer (Germany), were tested for MAGE-A3 and PRAME expression by<br />

specific qRT-PCR assays. The prognostic value <strong>of</strong> these Ags was determined<br />

by estimating the median overall survival (OS), disease-free interval<br />

(DFI) and disease-free survival (DFS). Results: Expression rates were 36%<br />

for MAGE-A3 and 66% for PRAME. The co-expression rate was 30%.<br />

Almost all tumors expressing MAGE-A3 also expressed PRAME. We<br />

observed no difference overall in Ag expression according to stage, tumor<br />

size and pts’ age. Squamous tumors expressed MAGE-A3 and PRAME more<br />

frequently than did adenocarcinomas (43 vs. 27 % and 80 vs. 44%<br />

respectively), and PRAME expression rates were higher in males than in<br />

females (70 vs. 53%). In the overall population, which included stages<br />

IA–IIIB, no prognostic value was detected for the expression <strong>of</strong> either Ag. In<br />

subset analyses, we found in 83 stage IIIB pts a trend to worse OS linked to<br />

MAGE-A3 expression (HR�1.977, p�0.0312). In 395 stage IB pts,<br />

PRAME expression was associated with worse OS (HR�1.483, p�0.0344)<br />

and DFS (HR�1.492, p�0.0167). Conclusions: The MAGE-A3 expression<br />

rate found is consistent with the results observed in the phase III MAGRIT<br />

trial (J-H Kim et al., 2011). No prognostic value for either PRAME or<br />

MAGE-A3 was observed in the overall population, in contrast to previous<br />

reports with smaller sample sizes (Gure et al., 2005, Boli et al., 2002) and<br />

in other tumors (Vourch’jourdain et al., 2009, Epping et al., 2008). We<br />

observed a trend toward poor prognostic value <strong>of</strong> MAGE-A3 in stage IIIB pts<br />

and <strong>of</strong> PRAME in stage IB pts.<br />

7058 General Poster Session (Board #37A), Sat, 1:15 PM-5:15 PM<br />

Prognostic role <strong>of</strong> FOXP3/CD4 ratio in resectable NSCLC. Presenting<br />

Author: Marta Usó, Fundación para la Investigación del Hospital General<br />

Universitario de Valencia, Valencia, Spain<br />

Background: Tregs play a critical role in immune tolerance to tumor cells<br />

and have been related to prognosis. The aim <strong>of</strong> this study was to determine<br />

the expression <strong>of</strong> Tregs makers genes by RT-PCR and to correlate them with<br />

clinico-pathological and prognostic variables in NSCLC. Methods: RNA was<br />

isolated from frozen lung specimens (tumor and normal lung) from<br />

resectable NSCLC patients (n�175). RT-PCR was performed to analyze the<br />

expression <strong>of</strong>: CD4, CD8A, CTLA-4, FoxP3, IL-10, CD25, CD127 and<br />

TGF�-1. Relative expression was normalized by an endogenous gene<br />

(GUSB) using the Pfaffl formulae. Statistical analyses were considered<br />

significant at p�0.05. Results: Tumor samples had significantly lower<br />

expression <strong>of</strong> CD127 (x0.45) and a tendency toward higher expression <strong>of</strong><br />

CD25 (x1.81), FoxP3 (x1.57) and CTLA-4 (x1.53) compared to normal<br />

lung tissues, reflecting a Treg phenotype infiltrating the tumor. Survival<br />

analyses revealed that patients with higher FoxP3 expression had reduced<br />

OS (median 29.8 vs 67 months, p� 0.026). We also found that those<br />

patients with high levels FoxP3/CD4 ratio had worse TTP and OS (median<br />

22.1 months vs NR, p� 0.021 and 29.8 vs NR months, p�0.003,<br />

respectively). A multivariable Cox regression model for TTP and OS was<br />

built using variables that were found significant in the univariate analysis<br />

(nodal involvement, histology, tumor size, PS, and FoxP3/CD4 ratio). This<br />

analysis revealed that FoxP3/CD4 ratio was an independent prognostic<br />

marker for both TTP and OS (see table). Conclusions: FoxP3 is a transcription<br />

factor necessary and sufficient for induction <strong>of</strong> the immunosuppressive<br />

functions in Tregs. In concordance, our results indicate that higher levels <strong>of</strong><br />

FoxP3/CD4 ratio in tumor samples is a poor prognostic markers for TTP and<br />

OS. Furthermore, the multivariable COX regression analysis showed that<br />

FoxP3/CD4 ratio is an independent prognostic marker. Therefore, this ratio<br />

could be used as a new biomarker <strong>of</strong> prognosis in resectable NSCLC.<br />

Supported by grants PS09-01149 and RD06/0020/1024 from ISCIII.<br />

TTP OS<br />

HR 95% IC P HR 95% IC P<br />

Histology 1.14 1.01-1.29 .027* - - -<br />

Nodal involvement 1.38 1.06-1.81 .016* 1.34 1.00-1.80 .045*<br />

Tumor size 2.505 1.449-4.330 .001* 2.030 1.173-3.512 .011*<br />

FoxP3/CD4 ratio 2.11 1.24-3.63 .006* 2.42 1.38-4.24 .002*<br />

465s<br />

7057 General Poster Session (Board #36H), Sat, 1:15 PM-5:15 PM<br />

Prognostic value <strong>of</strong> pathologic complete response to preoperative chemotherapy<br />

in early-stage non small cell lung cancer: Combined analysis <strong>of</strong> two<br />

IFCT randomized trials. Presenting Author: Guillaume Mouillet, University<br />

Hospital, Besançon, France<br />

Background: Our study aimed to evaluate whether pathologic complete<br />

response (pCR) in early-stage non-small cell lung cancer (NSCLC) following<br />

neoadjuvant chemotherapy resulted in improved outcome and to determine<br />

predictive factors for pCR. Methods: Eligible patients with stage IB or II<br />

NSCLC were included in two consecutive Intergroupe Francophone de<br />

Cancérologie Thoracique (IFCT) phase III trials evaluating platinum-based<br />

neoadjuvant chemotherapy, with pCR defined by the absence <strong>of</strong> viable<br />

cancer cells in the resected surgical specimen. Results: Among the 492<br />

patients analyzed, 41 (8.3%) achieved pCR. In the pCR group, 5-year<br />

overall survival (OS) was 80.0% compared to 55.8% in the non-pCR group<br />

(p�0.0007). In multivariate analyses, pCR was a favorable prognostic<br />

factor <strong>of</strong> OS (RR�0.34; 95% CI�0.18-0.64) in addition to squamous cell<br />

carcinoma (SCC), weight loss �5%, and stage IB disease. Five-year<br />

disease-free survival (DFS) was 80.1% in the pCR group compared to<br />

44.8% in the non-pCR group (p�0.0001). Two patients (4.9%) in the pCR<br />

group experienced disease recurrence compared to 193 patients (42.8%)<br />

in the non-pCR group. SCC subtype was the only independent predictor <strong>of</strong><br />

pCR (OR�4.30; 95% CI�1.90-9.72). Conclusions: Our results showed<br />

that pCR after preoperative chemotherapy was a favorable prognostic factor<br />

in Stage IB-II NSCLC. Our study is the largest published series evaluating<br />

pCRs following preoperative chemotherapy. The only factor predictive <strong>of</strong><br />

pCR was SCC. Identifying molecular predictive markers for pCR may help in<br />

distinguishing patients likely to benefit from neoadjuvant chemotherapy<br />

and in choosing the most adequate preoperative chemotherapy regimen.<br />

7059 General Poster Session (Board #37B), Sat, 1:15 PM-5:15 PM<br />

A systematic analysis <strong>of</strong> high-dose radiation in the treatment <strong>of</strong> surgically<br />

unresectable, locally advanced non-small cell lung cancer. Presenting<br />

Author: Madhusmita Behera, Department <strong>of</strong> Hematology and Medical<br />

Oncology, Emory University School <strong>of</strong> Medicine, Atlanta, GA<br />

Background: The 5-year survival rate for locally advanced non-small cell<br />

lung cancer (NSCLC) is � 25% with concomitant chemoradiotherapy.<br />

High-dose radiation appears to confer additional benefit based on surrogate<br />

endpoints from relatively small clinical trials. However, survival advantage<br />

<strong>of</strong> higher-dose radiation delivered by conventional fractionation <strong>of</strong> 1.8-2 Gy<br />

is unclear. We conducted a systematic review <strong>of</strong> pooled outcome data from<br />

published results <strong>of</strong> studies that evaluated the role <strong>of</strong> high-dose radiation in<br />

the management <strong>of</strong> locally advanced NSCLC. Methods: A detailed search <strong>of</strong><br />

published clinical trials was conducted using computerized databases<br />

(MEDLINE, EMBASE, Cochrane library) and meeting proceedings. Single<br />

arm studies using high dose (�66Gy) or randomized trials <strong>of</strong> high versus<br />

standard radiation dose (60-63Gy) for locally advanced NSCLC were<br />

selected. A systematic analysis <strong>of</strong> extracted data was performed using<br />

Comprehensive Meta Analysis (Version 2.2.048) s<strong>of</strong>tware under the random<br />

effect model. <strong>Clinical</strong> outcome in patients treated with high versus<br />

standard radiation dose was compared using point estimates for weighted<br />

values <strong>of</strong> median overall survival (OS), progression free survival (PFS), and<br />

response rate (RR). Treatment-related toxicities data between the two arms<br />

was compared using T-test and chi-square test. Results: Analytic data was<br />

retrieved from the results <strong>of</strong> 44 eligible studies reported between 1994 and<br />

2012. The studies enrolled 3699 patients, 62% males, 2095 and 1604<br />

treated with standard and high dose radiation respectively. The weighted<br />

median age was 63 and 61 years (p�0.66) for standard and high dose<br />

patients respectively. The weighted median OS was 19.1 vs. 16.4 months<br />

(p� 0.97); weighted RR <strong>of</strong> 76% vs. 70% (p�0.79) and survival rates after<br />

up to 3 years <strong>of</strong> follow up <strong>of</strong> 39% vs. 46% (p�0.45) for standard and high<br />

dose arms respectively. There was no significant difference in the rate <strong>of</strong><br />

grade �3 toxicities between the two treatment groups. Conclusions:<br />

High-dose radiation therapy does not result in improved survival over<br />

standard radiotherapy dose in patients with locally advanced NSCLC.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


466s Lung Cancer—Non-small Cell Local-Regional/Small Cell/Other Thoracic Cancers<br />

7060 General Poster Session (Board #37C), Sat, 1:15 PM-5:15 PM<br />

miRNA pr<strong>of</strong>iling in resectable NSCLC by multiplex next-generation sequencing.<br />

Presenting Author: Sandra Gallach, Fundación para la Investigación<br />

del Hospital General Universitario de Valencia, Valencia, Spain<br />

Background: Early-stage NSCLC has a relapse rate around 40% within 5<br />

years. With the advent <strong>of</strong> microRNA (miRNA), which seems to regulate<br />

many genes critical for tumorigenesis, there is a growing interest in the<br />

characterization <strong>of</strong> miRNAome in NSCLC specimens and to correlate them<br />

with prognosis. Next generation sequencing is a useful tool to study the<br />

miRNA content <strong>of</strong> solid tumors. Here, we applied high-throughput SOLiD<br />

transcriptome sequencing to study miRNAs expression in a cohort <strong>of</strong><br />

early-stage NSCLC patients (tumor vs normal lung). Methods: RNA was<br />

isolated from frozen lung specimens (tumor and normal lung) from<br />

resectable NSCLC patients (n�35). Samples with a RIN � 7 were analyzed<br />

and enriched in the miRNA fraction. miRNAs were sequencing using a<br />

bar-code multiplex SOLiD protocol. Data normalization was carried out by<br />

rescaling all data according to their counts. Readings were mapped against<br />

mature and no-mature miRNAs using miRBase. Statistical analysis was<br />

performed with CLCbio s<strong>of</strong>tware and considered significant when padj�0.005.<br />

Results: Using the SOLiD high throughput sequencing, we<br />

performed a systemic miRNA expression pr<strong>of</strong>iling analysis <strong>of</strong> paired<br />

samples (tumor vs normal lung). A total <strong>of</strong> 1268 miRNAs (mature and<br />

no-mature) have been detected in at least one sample. The differential<br />

expression between normal and tumor samples shown that 6 miRNAs<br />

(miR-193b, miR-182, miR-96, miR-148a, miR-299, miR-590) were<br />

upregulated and 7 (miR-145, miR-133a, miR-218, miR-125a, miR-30a,<br />

miR-126 and miR-139) were down-regulated significantly in tumor samples<br />

compared with normal lung tissues. We are performing studies using<br />

qRT-PCR in an independent cohort to further validate these findings.<br />

Conclusions: The deep sequencing technology used for differential miRNA<br />

expression is useful and novel. The use <strong>of</strong> barcoding allows multiplexing<br />

and lowers cost per sample. Several miRNAs were differentially expressed<br />

between tumor and normal tissue, but this point needs to be further<br />

validated in an independent cohort. Supported by grants TRA09-0132<br />

(MICINN) and RD06/0020/1024 (ISCIII).<br />

7062 General Poster Session (Board #37E), Sat, 1:15 PM-5:15 PM<br />

Comparative effectiveness <strong>of</strong> five treatment strategies for early-stage<br />

non-small cell lung cancer in the elderly. Presenting Author: Shervin<br />

Shirvani, University <strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: Early-stage lung cancer incidence among older adults is<br />

expected to increase due to demographic trends and CT-based screening,<br />

yet optimal treatment in the elderly remains controversial. Using the<br />

SEER-Medicare cohort spanning 2001-2007, we determined survival<br />

outcomes associated with five strategies used in contemporary practice.<br />

Methods: Treatment strategy and covariates were determined in 10,923<br />

patients age�66 with stage IA-IB non-small cell lung cancer. Cox regression,<br />

adjusted for patient and tumor factors, compared overall and<br />

disease-specific survival for lobectomy, sublobar resection, conventional<br />

radiation, stereotactic body radiotherapy (SBRT) and observation. In a<br />

second analysis, propensity-score matching was used for pairwise comparison<br />

<strong>of</strong> SBRT with other strategies. Results: Median age was 75 years and<br />

29% had substantial comorbidity. Treatment distribution was lobectomy<br />

(59%), sublobar resection (11.7%), conventional radiation (14.8%),<br />

observation (12.6%), and SBRT (1.1%). In unmatched Cox regression with<br />

median follow up <strong>of</strong> 3.2 years, SBRT was associated with the lowest risk <strong>of</strong><br />

death within six months <strong>of</strong> diagnosis (HR 0.47; 95%CI 0.37-0.61; referent<br />

is lobectomy). After six months, sublobar resection and SBRT were<br />

associated with similar overall survival (P�0.51) and were both modestly<br />

worse than lobectomy. With propensity-score matching, survival after SBRT<br />

was similar to lobectomy (HR 0.90; 95%CI 0.64-1.27). Conventional<br />

radiation and observation were associated with poor outcomes in all<br />

analyses. Disease-specific survival outcomes followed similar trends.<br />

Conclusions: This population-based experience provides strong support <strong>of</strong><br />

SBRT as an alternative to surgery or conventional radiation for select<br />

patient populations. Evaluation <strong>of</strong> this new therapy in randomized trials is<br />

urgently needed.<br />

7061 General Poster Session (Board #37D), Sat, 1:15 PM-5:15 PM<br />

SOX2 and FGFR1 gene copy number in surgically resected non-small cell<br />

lung cancer (NSCLC). Presenting Author: Luca Toschi, Humanitas Cancer<br />

Center, Rozzano, Italy<br />

Background: SOX2 is a member <strong>of</strong> the SRY-related HMG-box family <strong>of</strong><br />

transcription factors and has been shown to be frequently amplified and<br />

overexpressed in squamous cell lung cancer, with conflicting results<br />

regarding its prognostic relevance. Similarly, FGFR1, a transmembrane<br />

tyrosine kinase receptor belonging to the fibroblast growth factor receptor<br />

family, has been recently reported to be amplified in squamous cell lung<br />

carcinomas, suggesting a potential role for FGFR1 as a therapeutic target in<br />

NSCLC. Aim <strong>of</strong> the present study is to evaluate SOX2 and FGFR1 gene copy<br />

number in surgically resected NSCLCs, to investigate their prognostic<br />

relevance and their association with clinico-pathological characteristics.<br />

Methods: SOX2 and FGFR1 gene copy number was assessed by fluorescence<br />

in situ hybridization (FISH) in tissue microarray cores from 447<br />

surgically resected NSCLCs. Each patient was given a score ranging from 1<br />

to 6 according to increasing mean copy number per cell <strong>of</strong> each gene, with<br />

6 indicating true gene amplification. Results: SOX2 and FGFR1 FISH was<br />

successfully performed in 445 patients (pts), which were grouped as �<br />

(score 5-6) and - (score 1-4). Using this scoring system 105 (23.6%) pts<br />

tested SOX2�, while 74 (16.6%) pts resulted FGFR1�. True gene<br />

amplification for SOX2 and FGFR1 was observed in 19 (4.3%) and 37<br />

(8.3%) cases, respectively. SOX2� and FGFR1� status was significantly<br />

associated with squamous histology (p�.001). Additionally, SOX2� pts<br />

had a significantly higher chance <strong>of</strong> being former/current smokers, male<br />

and FGFR1�. FGFR1 gene status had no prognostic impact in the whole<br />

population and in the squamous cell carcinoma subgroup. Conversely,<br />

SOX2� pts had significantly longer overall survival compared with SOX2pts<br />

(HR 0.68, p�.020). When restricting survival analysis to squamous cell<br />

histology, stage I-II SOX2� pts had a significant survival advantage<br />

compared with SOX2- group (HR 0.38, p�.006), while no difference was<br />

observed in stage III-IV pts. Conclusions: Increased SOX2 and FGFR1 gene<br />

copy number is a common event in lung cancer pts with squamous cell<br />

histology. SOX2 gene gain is a favorable prognostic factor in surgically<br />

resected pts, particularly in early stage squamous cell cancers.<br />

7063 General Poster Session (Board #37F), Sat, 1:15 PM-5:15 PM<br />

FGFR1 amplification in squamous cell lung cancers. Presenting Author:<br />

Michele L. Cote, Karmanos Cancer Institute, Wayne State University,<br />

Detroit, MI<br />

Background: Squamous cell carcinoma <strong>of</strong> the lung (SqCCL) is the second<br />

most common type <strong>of</strong> lung cancer. There are currently no established<br />

targeted therapies that take advantage <strong>of</strong> SqCCL-specific genetic abnormalities.<br />

Amplification <strong>of</strong> the fibroblast growth factor receptor type 1 gene<br />

(FGFR1) has been identified as a driver event in breast cancers and in<br />

SqCCL. The demographic characteristics and prognosis <strong>of</strong> patients with<br />

these tumors remains to be defined. Methods: DNA from 123 surgically<br />

resected, fresh frozen, and clinically annotated SqCCLs was extracted<br />

using a resin-based or phenol-based methodology. DNA quantity and<br />

quality was assessed using the Quantifiler kit (ABI). Specimen histology<br />

was validated, and the proportion <strong>of</strong> tumor cells was �60%. Copy number<br />

variation (CNV) analysis was performed using quantitative PCR with<br />

primers specific for exons 12 and 16 <strong>of</strong> FGFR1 and data analysis using<br />

CopyCaller (ABI). Samples with a predicted CNV <strong>of</strong> at 2 or greater in at least<br />

one exon were scored as amplified. Chi-squared tests were used to examine<br />

clinical and demographic differences between patients with and without<br />

amplification <strong>of</strong> FGFR1. Survival differences were examined by Kaplan<br />

Meier and Cox regression models. Results: Men comprised the majority <strong>of</strong><br />

the population (n�91, 74%) and most cases were pathological stage I<br />

(n�75, 61%) or stage II (n�32, 26%). Thirty (24.4%) tumors showed<br />

amplification <strong>of</strong> FGFR1. There was no association between amplification<br />

and sex (p�0.18) or stage (p�0.37). The majority <strong>of</strong> the patients were<br />

white (n�114, 92.7%), but the proportion <strong>of</strong> tumors that were amplified<br />

was higher among non-whites (5/9, 55.5%) than whites (25/114, 21.9%,<br />

p�0.02). Overall, median survival was 32.2 months. In univariate survival<br />

analysis, amplification was associated with an increased hazard <strong>of</strong> death<br />

(HR�1.55, Wilcoxon p�0.03). Tumor stage at diagnosis was also associated<br />

with increased risk <strong>of</strong> death (HR�1.67, p�0.0009). Only stage<br />

remained a significant predictor <strong>of</strong> death in the multivariate survival models<br />

(HR�1.65, p�0.004). Conclusions: Somatic amplification <strong>of</strong> FGFR1 is a<br />

relatively common event in SqCCL and may be associated with race and<br />

overall survival.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


Lung Cancer—Non-small Cell Local-Regional/Small Cell/Other Thoracic Cancers<br />

7064 General Poster Session (Board #37G), Sat, 1:15 PM-5:15 PM<br />

Survival following segmentectomy and lobectomy for stage I non-small cell<br />

lung cancer. Presenting Author: Cardinale B. Smith, Division <strong>of</strong> Hematology<br />

and Medical Oncology, The Tisch Cancer Institute, Mount Sinai School<br />

<strong>of</strong> Medicine, New York, NY<br />

Background: Although lobectomy is considered the standard surgical<br />

treatment forstage IA non–small cell lung cancer (NSCLC), limited resections<br />

are frequently performed for patients with poor lung function or high<br />

operative risk. Recent studies suggest that segmentectomy may be the<br />

superior limited resection procedure. The objective <strong>of</strong> this study was to<br />

compare survival among patients with stage IA (�3cm) NSCLC undergoing<br />

lobectomy vs. segmentectomy. Methods: Using theSurveillance, Epidemiology<br />

and End Results registry we identified 15,180 cases <strong>of</strong> stage IA NSCLC<br />

that underwent lobectomy or segmentectomy. We used logistic regression<br />

to determine propensity scores for patients undergoing segmentectomy<br />

based on the patient’s preoperative characteristics. Overall and lung<br />

cancer-specific survival <strong>of</strong> patients treated with lobectomy versus segmentectomy<br />

was compared after adjusting, stratifying, or matching patients<br />

based on their propensity score. We also performed secondary analyses in<br />

subgroups <strong>of</strong> age (�70 vs. �70 years) and tumor size �2 cm. Results:<br />

Overall, 1,200 (8%) patients underwent segmentectomy. Among the entire<br />

cohort, analyses adjusting for propensity scores did not demonstrate a<br />

difference in outcomes among patients treated with lobectomy versus<br />

segmentectomy, (adjusted hazard ratio [HR] for overall survival 1.11, 95%<br />

confidence interval [CI]: 0.94 – 1.30 and lung cancer-specific survival<br />

1.11, 95% CI: 0.98 – 1.25). Similarly, secondary analyses showed no<br />

difference in overall (HR: 1.12, 95% CI: 0.90 – 1.40) and lung cancerspecific<br />

survival (HR: 1.04, 95% CI: 0.88 – 1.24) among patients with<br />

tumors �2 cm (T1a tumors). For patients �70 years <strong>of</strong> age, no difference<br />

in overall survival was observed (HR: 0.97, 95% CI: 0.73 – 1.28); however,<br />

a lung cancer-specific survival advantage <strong>of</strong> lobectomy was observed (HR:<br />

1.19, 95% CI: 1.00 – 1.40). Finally, among those �70 years overall<br />

survival (HR: 1.03, 95% CI: 0.87 – 1.22) and lung cancer-specific survival<br />

(HR: 1.18, 95% CI: 0. 97 – 1.44) showed equivalence <strong>of</strong> the two surgical<br />

groups. Conclusions: Segmentectomy and lobectomy may lead to equivalent<br />

survival rates among patients with stage IA NSCLC. These study<br />

findings should be confirmed in prospective studies.<br />

7066 General Poster Session (Board #38A), Sat, 1:15 PM-5:15 PM<br />

Prospective study <strong>of</strong> tumor suppressor gene (TSG) methylation as a<br />

prognostic biomarker in surgically resected non-small cell lung cancer<br />

(NSCLC). Presenting Author: Christopher G. Azzoli, Memorial Sloan-<br />

Kettering Cancer Center, New York, NY<br />

Background: In the New England Journal <strong>of</strong> Medicine, Brock et al. (2008)<br />

published a nested case-control study which tested the association<br />

between early recurrence <strong>of</strong> NSCLC after surgical resection, and TSG<br />

promoter methylation in tumor and lymph nodes detected by methylationspecific<br />

PCR (MSP). They reported that promoter methylation <strong>of</strong> the TSGs<br />

p16, CDH13, RASSF1A, and APC was significantly associated with early<br />

recurrence in 51 patients with stage I NSCLC compared to matched<br />

patients without early recurrence. We attempted to confirm these findings.<br />

Methods: In a prospective study, fresh frozen tumor tissue was acquired<br />

after surgical resection in 107 patients with stage I-IIIA NSCLC between<br />

2003-2008. The promoter methylation status <strong>of</strong> the same 4 genes<br />

examined by Brock, et al (p16, CDH13, RASSF1A, APC), as well as 6<br />

additional TSGs (MGMT, WIF-1, METH-2, GSTP1, SOCS3, DAPK) were<br />

assessed in the tumor tissue using quantitative MSP (MethyLight assay),<br />

with any amount <strong>of</strong> methylation scored as positive. Methylation status was<br />

correlated with clinical features, pathologic stage, disease-free survival<br />

(DFS), and overall survival (OS). Results: No significant associations were<br />

observed between promoter methylation <strong>of</strong> individual TSGs and DFS. No<br />

significant associations were observed between the number <strong>of</strong> methylated<br />

TSGs and DFS, or OS. Increased RASSF1A methylation was observed in<br />

poorly-differentiated and undifferentiated tumors compared to tumors that<br />

were well- or moderately-differentiated (p�0.031). Increased WIF-1 methylation<br />

and GSTP1 methylation were associated with increasing T (p�0.01)<br />

and N stage (p�0.028), respectively. Squamous cell carcinomas (SQCCs)<br />

were characterized by increased p16 methylation (p�0.0314) and decreased<br />

APC methylation (p�0.0146) compared to tumors <strong>of</strong> non-SQCC<br />

histologies. Conclusions: In this prospective study, we did not confirm that<br />

the promoter methylation <strong>of</strong> p16, CDH13, RASSF1A, and APC, or 6 other<br />

TSGs, was prognostic for early recurrence in surgically resected NSCLC.<br />

467s<br />

7065 General Poster Session (Board #37H), Sat, 1:15 PM-5:15 PM<br />

Phase II study <strong>of</strong> concurrent cetuximab (C225) plus definitive thoracic<br />

radiotherapy (XRT) followed by adjuvant docetaxel in poor prognosis<br />

patients with locally advanced non-small cell lung cancer (LA-NSCLC).<br />

Presenting Author: Thomas J. Dilling, H. Lee M<strong>of</strong>fitt Cancer Center &<br />

Research Institute, Tampa, FL<br />

Background: Recursive partitioning analysis has shown that ECOG PS � 2,<br />

male gender, or age � 70 are prognostic <strong>of</strong> poor outcome in LA-NSCLC pts.<br />

Concurrent chemoradiotherapy improves survival, but toxicity is concerning<br />

in this frail pt cohort. We therefore opened this trial <strong>of</strong> concurrent<br />

definitive-dose XRT (70 Gy in 35 fractions) and C225 (250 mg/m2 /dose),<br />

followed by adjuvant C225 and docetaxel (60 mg/m2 q3weeks x 3 cycles).<br />

Methods: Eligible patients had pathologically-proven LA-NSCLC (stage IIA –<br />

“dry” IIIB), not surgically resectable. They had ECOG PS 2 OR weight loss<br />

�5% in 3 months OR age �70. The primary objective was progression-free<br />

survival (PFS). Secondary objectives included overall survival (OS) and<br />

overall response rate (ORR). Results: From 5/2008 to 11/2010, 32 pts were<br />

evaluated for our single-institution, IRB-approved prospective clinical trial.<br />

3 pts were screen-failures and 2 more withdrew consent prior to treatment,<br />

leaving 27 evaluable patients. 1 was removed due to poor therapy<br />

compliance and 2 were taken <strong>of</strong>f trial due to grade 3 toxicity from the C225<br />

loading dose, but were followed under intent-to-treat analysis. Median<br />

follow-up and OS was 10.5 months. Median PFS was 7.8 months. ORR �<br />

59.3%. 8 early/sudden deaths were reported, so the trial closed early:<br />

radiation pneumonitis (RP) was the apparent cause in 1 and probably/<br />

possibly in 4 others, with insufficient information in 2 additional pts; 1 pt<br />

died <strong>of</strong> failure to thrive. Of the remaining pts, 3 developed grade 3� RP and<br />

2 had grade 2 RP. Conclusions: Pts enrolled on this trial had improved OS<br />

compared with poor-PS historical controls (10.5 vs 6.3 months) and<br />

comparable OS compared with good-PS historical controls (10.5 vs 11.9<br />

months) treated with RT (Werner-Wasik M et al. Int J Radiat Oncol Biol<br />

Phys. 2000;48:1475-82). However, pulmonary toxicity is a concern.<br />

Analysis <strong>of</strong> RP events is ongoing. Updated results will be presented at the<br />

meeting.<br />

7067 General Poster Session (Board #38B), Sat, 1:15 PM-5:15 PM<br />

Preoperative survivin, ERCC1, and PTEN expression in stage III non-small<br />

cell lung cancer (NSCLC) patients (pts) treated with neoadjuvant and<br />

definitive chemoradiation and association with overall survival (OS).<br />

Presenting Author: Marta Batus, Rush University Medical Center, Chicago,<br />

IL<br />

Background: Thoracic radiation and concurrent chemotherapy consisting <strong>of</strong><br />

platinum based doublets has produced modest improvement in long term<br />

survival for patient with locally advanced (LA) NSCLC. There is relatively<br />

little information regarding molecular pr<strong>of</strong>iles and outcome in LA-NSCLC<br />

patients (pts) treated with chemoradiation. The objective <strong>of</strong> this retrospective<br />

study is to evaluate potential relationships between expression <strong>of</strong> DNA<br />

repair enzyme ERCC1 and enzymes involved in cell survival – survivin and<br />

PTEN. Methods: Stage III NSCLC pts who were treated with chest radiation<br />

(40-60Gy) and concurrently with platinum doublet and who had sufficient<br />

pretreatment tissue were included in this study. Immunohistochemistry<br />

was used to detect nuclear and cytoplasmic expression (frequency 0-4 and<br />

intensity 0-4) <strong>of</strong> survivin, and PTEN, and for nuclear expression <strong>of</strong> ERCC1.<br />

Product <strong>of</strong> intensity and frequency was calculated for all markers and<br />

correlated with overall survival (OS). Results: 97 pts had adequate tumor<br />

samples for analysis. 53 women, median age 67. 48 pts with ERCC1 prod<br />

��6 had longer OS than 41 pts with ERCC1 prod �6 (19.6 vs 1.0 months,<br />

p�0.034). 16 pts with ERCC1 prod �6, PETN prod ��6 and survivin prod<br />

�4 had significantly lower OS than 68 pts with ERCC1��6, PETN �6 and<br />

survivin ��4 (17.2 vs 40.2 months, p�0.001). Conclusions: The association<br />

<strong>of</strong> inferior survival in LA-NSCLC pts whose tumors express high<br />

survivin, low PTEN, and high ERCC1, suggests that combining inhibitors <strong>of</strong><br />

survivin and or <strong>of</strong> PI3KCA with chemoradiation and developing strategies to<br />

inhibit DNA repair might improve outcomes in this group <strong>of</strong> pts.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


468s Lung Cancer—Non-small Cell Local-Regional/Small Cell/Other Thoracic Cancers<br />

7068 General Poster Session (Board #38C), Sat, 1:15 PM-5:15 PM<br />

Phase II trial: Concurrent chemotherapy and radiotherapy with nitroglycerin<br />

in locally advanced non-small cell lung cancer patients. Presenting<br />

Author: Dolores De la Mata, Instituto Nacional de Cancerología, Mexico<br />

City, Mexico<br />

Background: The treatment <strong>of</strong> choice for locally advanced non small cell<br />

lung cancer (NSCLC) is concurrent chemoradiation (CRT). However, efforts<br />

to improve treatment results include targeted therapy and the use <strong>of</strong><br />

radiosensitizers. Nitroglycerin (NTG), a nitric oxide (NO) donor agent,<br />

reduces expression <strong>of</strong> Hypoxia-Induced Factor, which is associated to both<br />

chemo and radio resistance. Methods: This is phase II trial in patients with<br />

locally advanced NSCLC treated with chemotherapy (CT) based on cisplatin<br />

and vinorelbin with NTG concurrent with radiation therapy. A 25 mg NTG<br />

patch was administered to the patients during the first 5 days <strong>of</strong> each<br />

induction treatment cycle and during chemo-radiotherapy. Blood samples<br />

<strong>of</strong> VEGF were taken before any treatment and after two cycles <strong>of</strong> CT. The<br />

protocol is registered with <strong>Clinical</strong>Trials.gov, number NCT00886405.<br />

Results: 35 patients were enrolled in this trial. Median Follow up was 16.6<br />

months (SD �13.6). Mean age was <strong>of</strong> 59.9 years (�10.8), 68.6% <strong>of</strong> the<br />

patients were smokers. ECOG status was 0 in 22.9%, 1 in 65.7% and 2 in<br />

11.5%, respectively. Histopathology was adenocarcinoma in 68.6%,<br />

epidermoid in 17.1% and undifferentiated in 14.3%. Stage distribution<br />

was: IIIa, 57.6% and IIIb, 42.5%. All patients completed CRT treatment<br />

and four underwent surgical treatment. Toxicity pr<strong>of</strong>ile related to NTG was<br />

grade 1 and 2 headache in 17.1%. Grade 3 and 4 toxicities were<br />

esophagitis (17.1%), neutropenia (62.9%), and nausea (5.7%). Sixty-four<br />

per cent <strong>of</strong> patients achieved partial response after CT and 75.8% after<br />

CRT. PFS was 11.8 months (95%, IC 7.8-15.6) and OS was 42.9 months<br />

(95%IC 31.3-52.1). After two cycles <strong>of</strong> CT, plasma VEGF levels were<br />

significantly lower (Median 132�79 vs. 53�78 pg/ml, p�0.001). No<br />

differences on PFS and OS were found between patients with a reduction �<br />

93 pg/ml (median <strong>of</strong> differences between VEGFR before and after chemotherapy).<br />

Conclusions: The addition <strong>of</strong> NTG to induction CT, and concurrent<br />

CRT on locally advanced NSCLC patients seems to increase the response<br />

rate, PFS and OS with an acceptable toxicity pr<strong>of</strong>ile. A prospective trial is<br />

warranted to confirm these findings.<br />

7070 General Poster Session (Board #38E), Sat, 1:15 PM-5:15 PM<br />

Multicenter phase II study <strong>of</strong> concurrent high-dose (72Gy) threedimensional<br />

conformal radiotherapy (3D-CRT) without elective nodal<br />

irradiation with chemotherapy using cisplatin and vinorelbine for unresectable<br />

stage III non-small cell lung cancer (NSCLC). Presenting Author:<br />

Hidehito Horinouchi, Division <strong>of</strong> Internal Medicine and Thoracic Oncology,<br />

National Cancer Center Hospital, Tokyo, Japan<br />

Background: The optimal dose <strong>of</strong> radiotherapy remains unclear in concurrent<br />

chemoradiotherapy for unresectable stage III NSCLC. We previously<br />

concluded that the recommended dose for further trial was 72Gy in a phase<br />

I study (Sekine et al., Int J Radiat Oncol Biol Phys. 953-959, 2012).<br />

Methods: Eligible patients (unresectable stage III NSCLC, age between 20<br />

and 74, PS 0-1, V20 � 30%) received cisplatin (80 mg/m2 day 1) and<br />

vinorelbine (20 mg/m2 days 1 and 8) repeated every 4 weeks for 3-4 cycles.<br />

The 3D-CRT started at the first day <strong>of</strong> chemotherapy at a total dose <strong>of</strong> 72 Gy<br />

in 36 fractions. The primary endpoint was a 2-year survival rate and the<br />

planned sample size was 60 to reject the rate <strong>of</strong> 45% under the expectation<br />

<strong>of</strong> 65% with a power <strong>of</strong> 90% and an alpha error <strong>of</strong> 5%. Results: Thirty-one<br />

patients from 4 institutions were enrolled between May 2009 and March<br />

2010. This trial was terminated early due to slow accrual and grade 5<br />

pulmonary toxicities in 2 patients. There were 25 men and 6 women with a<br />

median (range) age <strong>of</strong> 59 (32-72) years. Of these, 23 had adenocarcinoma<br />

and 21 had stage IIIA disease. The median (range) V20 value was 20<br />

(9-30). The full planned dose <strong>of</strong> radiotherapy could be administered in 30<br />

(97%) patients, and 26 (84%) <strong>of</strong> the patients received 3-4 cycles <strong>of</strong><br />

chemotherapy. During the chemoradiotherapy, grade 3-4 febrile neutropenia,<br />

infection and esophagitis were noted in 5, 4 and 1 <strong>of</strong> the 31 patients,<br />

respectively. After completion <strong>of</strong> the planned chemoradiotherapy, 5 patients<br />

had grade 3 or higher radiation pneumonitis, and 2 (6%) <strong>of</strong> these<br />

patients died at 6.6 and 7.3 months after the treatment started. The overall<br />

response rate was 97% (95% confidence interval: 83.3-99.9). Twenty-four<br />

patients are alive and thirteen patients experienced recurrence (2 had<br />

loco-regional recurrences, 7 had distant recurrence and 4 had mixed<br />

recurrence pattern) at a median follow up <strong>of</strong> 16.4 months. Conclusions:<br />

Concurrent high-dose (72Gy) 3D-CRT with chemotherapy using cisplatin<br />

and vinorelbine may have a too excessive incidence <strong>of</strong> pulmonary toxicities<br />

to warrant any further evaluation.<br />

7069 General Poster Session (Board #38D), Sat, 1:15 PM-5:15 PM<br />

Mediastinal lymph node examination and survival in resected early-stage<br />

non-small cell lung cancer in the Surveillance, Epidemiology, and End<br />

Results (SEER) database. Presenting Author: Raymond U. Osarogiagbon,<br />

Boston Baskin Cancer Foundation, Germantown, TN<br />

Background: Pathologic nodal stage is a key prognostic factor in resectable<br />

non-small cell lung cancer (NSCLC). Mediastinal lymph node (MLN)<br />

metastasis connotes a poor prognosis. Yet, some NSCLC resections in the<br />

US do not include MLN examination. Methods: We analyzed SEER data<br />

from 1998 to 2002 to quantify the long-term survival impact <strong>of</strong> failure to<br />

examine MLN in resected NSCLC. We used Kaplan-Meier methods to<br />

compare the unadjusted survival difference between patients with, and<br />

without, MLN examination. We used Cox proportional hazards and competing<br />

risk models to serially adjust for the impact <strong>of</strong> risk factors on survival<br />

differences. Results: Sixty-two percent <strong>of</strong> patients with pathologic N0 or N1<br />

NSCLC had no MLN examined. Men, African-<strong>American</strong>s, patients with<br />

more advanced stage, and those who had less than pneumonectomy were<br />

less likely to have MLN examination. Five-year all-cause mortality (46.9% v<br />

51.7%, p�.001), and lung cancer-specific mortality (31.5% v 36%,<br />

p�.001), rates were higher in those without MLN examination. After<br />

adjustment for potential confounders, MLN examination was associated<br />

with a 6% reduction in all-cause mortality (HR, 0.94; CI, 0.89-0.99;<br />

p�.014), and 10% reduction in lung cancer-specific mortality (HR, 0.90;<br />

95% CI, 0.84-0.96; p�.002) rates. The excess risk in 1 year’s cohort <strong>of</strong><br />

U.S. lung resections was 2,700 lives over 5 years. Conclusions: Failure to<br />

examine MLN was a common practice in �MLN-negative� NSCLC resections,<br />

which significantly impaired long-term survival. Efforts to understand<br />

the etiology <strong>of</strong> this quality gap, and measures to eliminate it, are warranted.<br />

7071 General Poster Session (Board #38F), Sat, 1:15 PM-5:15 PM<br />

Anatomic segmentectomy versus lobectomy for clinical stage I non-small<br />

cell lung cancer: A propensity-matched analysis. Presenting Author:<br />

Matthew J. Schuchert, Department <strong>of</strong> Cardiothoracic Surgery, University <strong>of</strong><br />

Pittsburgh Medical Center, Pittsburgh, PA<br />

Background: Although anatomic segmentectomy is considered a “compromised”<br />

procedure by many surgeons, new information from several retrospective,<br />

single-institution series has countered negative premises regarding<br />

tumor recurrence and patient survival. The primary objective <strong>of</strong> this study<br />

was to utilize propensity score matching to compare outcomes following<br />

these anatomic resection approaches for stage I NSCLC. Methods: Patients<br />

undergoing lobectomy (n�392) vs. segmentectomy (n�793) for clinical<br />

stage I NSCLC were matched 1:1 using a propensity score that accounted<br />

for the potential confounding effects <strong>of</strong> pre-operative patient variables.<br />

Matching based on propensity scores produced 312 patients in each group.<br />

Primary outcome variables included recurrence-free and overall survival.<br />

Factors affecting survival were assessed by proportional hazards (Cox)<br />

regression and Kaplan-Meier survival function estimates. Results: Perioperative<br />

mortality was 1.2% in the segmentectomy group and 2.5% in the<br />

lobectomy group (p�0.38). Ninety-day mortality was 2.6% and 4.8%<br />

(p�0.20), respectively. At a mean follow-up <strong>of</strong> 5.4 years, no differences<br />

were noted in locoregional (5.5% vs. 5.1%, p�1.00), distant (14.8% vs.<br />

11.6%, p�0.29) or overall recurrence rates (20.2% vs. 16.7%, p�0.30)<br />

when comparing segmentectomy with lobectomy. Furthermore, no significant<br />

differences were noted in time to recurrence (p�0.415) or overall<br />

survival (p�0.258) when comparing the matched groups. Five year<br />

freedom from recurrence (95% CI) was: Segment 0.70 [95% CI: (0.63,<br />

0.78) vs. Lobe 0.71 [95% CI: 0.64, 0.78]. Overall survival (95% CI) was:<br />

Segment 0.54 [95% CI: (0.47, 0.51) vs. Lobe 0.60 [95% CI: 0.54, 0.67].<br />

Segmentectomy was not found to be an independent predictor <strong>of</strong> recurrence<br />

(HR: 1.11, 95% CI: 0.87, 1.40) or overall survival (HR � 1.17, 95%<br />

CI: 0.89.1.52). Conclusions: In this large propensity-matched comparison,<br />

anatomic segmentectomy is associated with similar time to recurrence and<br />

overall survival rates when compared to lobectomy for clinical stage I<br />

NSCLC. These results will need further validation by prospective, randomized<br />

trials (CALGB 140503).<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


Lung Cancer—Non-small Cell Local-Regional/Small Cell/Other Thoracic Cancers<br />

7072^ General Poster Session (Board #38G), Sat, 1:15 PM-5:15 PM<br />

Size distribution and metastasis in discarded intrapulmonary lymph nodes<br />

(LN) after lung cancer resection. Presenting Author: Laura Elizabeth Miller,<br />

University <strong>of</strong> Tennessee Health Science Center, Memphis, TN<br />

Background: Lymph node (LN) status is the most important prognostic<br />

determinant after resection <strong>of</strong> lung cancer. 18% <strong>of</strong> a SEER cohort and 12%<br />

<strong>of</strong> a Memphis cohort had no LNs examined (pNx). Patients with pNx have<br />

inferior survival to T-category matched pN0 patients with at least 1 LN<br />

examined. The optimal number <strong>of</strong> LN needed to safely declare a patient<br />

pN0 may be �10. Less than 20% <strong>of</strong> resections in SEER achieve this. We<br />

hypothesized that a significant number <strong>of</strong> intrapulmonary LNs are left<br />

unexamined and some may harbor metastatic disease. We report the size<br />

characteristics <strong>of</strong> materials examined in a re-dissection protocol to test this<br />

hypothesis. Methods: Prospective study <strong>of</strong> lung resection specimens redissected<br />

after signout <strong>of</strong> the final pathology report. Remnant lung material<br />

was dissected with thin cuts and all LN-like material was retrieved for<br />

microscopic examination, irrespective <strong>of</strong> size or location. The size <strong>of</strong><br />

non-LN tissue, LN without metastasis and LN with metastases were<br />

compared using the Wilcoxon-Mann-Whitney test. Results: 112 specimens<br />

were examined and 1,094 LN-like materials were retrieved. 749 (69%)<br />

proved to be LN tissue. 71 (10%) LNs retrieved had metastasis. Non-LN<br />

tissue was significantly smaller than LN tissue (p�0.0001). LNs with<br />

metastasis were significantly larger than those without metastasis (p<br />

�0.0001). 60% <strong>of</strong> materials �2cm were LNs with metastasis. 7% <strong>of</strong><br />

materials �1cm were LN with metastasis. 52% <strong>of</strong> LNs with metastasis,<br />

and 55% <strong>of</strong> LNs without metastasis measured from 0.5 to 1.5cm (Table).<br />

Majority <strong>of</strong> LNs �2cm had metastatic disease, but 40% did not; a notable<br />

proportion <strong>of</strong> LNs with metastasis were small. Nearly equal percentages <strong>of</strong><br />

LNs with and without metastasis were found in the range <strong>of</strong> 0.5-1.5cm.<br />

Conclusions: Statistical differences in size between lymph nodes with and<br />

without metastasis is clinically meaningless due to broad overlap. LN size<br />

alone is not an adequate predictor <strong>of</strong> LN metastasis.<br />

Size (cm) Size range (cm), n (%)<br />

Mean Median 0-0.5 >0.5-1 >1-1.5 1.5-2 >2<br />

Non-LN, n�345 0.48 0.4 252 (73) 66 (19) 17 (5) 10 (3) 0 (0)<br />

LN without 0.69 0.6 281 (42) 302 (45) 72 (11) 15 (2) 8 (1)<br />

metastasis<br />

n�678<br />

LN with 1.32 1.2 8 (11) 20 (28) 17 (24) 14 (20) 12 (17)<br />

metastasis<br />

n�71<br />

7074 General Poster Session (Board #39A), Sat, 1:15 PM-5:15 PM<br />

Factors influencing recurrence following anatomic lung resection for<br />

non-small cell lung cancer. Presenting Author: Rodney Jerome Landreneau,<br />

University <strong>of</strong> Pittsburgh Shadyside Medical Center, Pittsburgh, PA<br />

Background: Anatomic lung resection provides the patient with the best<br />

chance for cure in the setting <strong>of</strong> early-stage non-small cell lung cancer<br />

(NSCLC). Despite complete (R0) resection, up to 20-30% <strong>of</strong> patients will<br />

develop recurrent disease. In the current study, we analyze the impact <strong>of</strong><br />

surgical and pathologic variables upon recurrence patterns following<br />

anatomic lung resection for clinical stage I NSCLC. Methods: A total <strong>of</strong><br />

1,192 patients (394 segmentectomies, 805 lobectomies) with clinical<br />

stage I NSCLC were evaluated. The primary outcome variable was recurrence.<br />

Multivariate analysis was performed based upon clinical (age,<br />

gender, comorbidities), surgical (operation, approach, surgical margin) and<br />

pathological (pleural invasion, tumor size and histology) variables. Predictors<br />

<strong>of</strong> recurrence were identified by proportional hazards regression.<br />

Differences in recurrence patterns between groups are illustrated by log<br />

rank tests applied to Kaplan-Maier estimates. Results: A total <strong>of</strong> 243<br />

recurrences (20.3%) were recorded at a mean follow-up <strong>of</strong> 35.6 months<br />

(71 locoregional, 172 distant). There was no significant difference in<br />

recurrence patterns when comparing segmentectomy and lobectomy.<br />

Multivariate analysis demonstrated that a margin:tumor ratio � 1, angiolymphatic<br />

invasion and the presence <strong>of</strong> only mild-moderate tumor inflammation<br />

were predictors <strong>of</strong> recurrence risk. Conclusions: Recurrence following<br />

anatomic lung resection is influenced predominantly by pathological<br />

variables (tumor size, angiolymphatic invasion, tumor inflammation).<br />

Optimization <strong>of</strong> surgical margin in relation to tumor size may improve<br />

outcomes. Extent <strong>of</strong> resection (segmentectomy vs. lobectomy) does not<br />

appear to have an impact on recurrence-free survival when adequate<br />

margins are obtained. These data have implications regarding the potential<br />

use <strong>of</strong> adjuvant therapy in selected Stage I patients at high risk for<br />

recurrence.<br />

Segmentectomy<br />

(n�394)<br />

Lobectomy<br />

(n�805) P value<br />

Recurrence 81(20.4%) 162 (20.1%) 0.88<br />

Locoregional 23 (5.9%) 48 (6.0%) 1.00<br />

Distant 58 (11.9%) 114 (14.1%) 0.79<br />

5-yr freedom from recurrence 69% 70% 0.53<br />

469s<br />

7073 General Poster Session (Board #38H), Sat, 1:15 PM-5:15 PM<br />

N0321: A phase II study <strong>of</strong> bortezomib, paclitaxel, carboplatin (CBCDA),<br />

and radiotherapy (RT) for locally advanced non-small cell lung cancer<br />

(NSCLC). Presenting Author: Steven E. Schild, Mayo Clinic, Scottsdale, AZ<br />

Background: In preclinical studies, combinations <strong>of</strong> bortezomib and RT<br />

result in synergistic tumor killing (Cancer Chemother Pharmacol. 2007;59:<br />

207-15). Our group reported the results from the phase I portion <strong>of</strong> this<br />

study previously (J Clin Oncol. 200;28 (suppl; abstr 7085)). Based on<br />

these data, we undertook a phase II study <strong>of</strong> bortezomib in combination<br />

with paclitaxel/CBCDA and RT. Methods: Based on results from our<br />

previously reported phase I trial, systemic therapy included bortezomib<br />

(1.2 mg/m² IV days 1,4,8,11), paclitaxel (175 mg/m² IV day 2), and<br />

carboplatin (CBCDA; AUC�6 day 2) given every 3 weeks for 2 cycles.<br />

Thoracic radiotherapy (RT) included 60Gy/30 daily fractions starting day 1.<br />

The primary endpoint was the 12-month survival rate. If 41 or more <strong>of</strong> these<br />

60 patients (68%) were alive at least 12 months after study entry, the trial<br />

would be deemed as positive and further study would be warranted. Results:<br />

27 pts were enrolled to the phase II portion: M/F�17/10; stage IIIA/<br />

IIIB�13/14; ECOG PS 0/1�9/18; and median age: 58 (range 43-79). 18<br />

pts (67%) completed therapy per protocol. At a planned interim analysis,<br />

23 <strong>of</strong> 26 evaluable patients (88.5%, 95% CI: 70-98%) survived for at least<br />

6 months, which exceeded the boundary <strong>of</strong> 21 or more needed to continue<br />

to full accrual. This trial could have continued per protocol, but was closed<br />

early due to slow accrual. With a median follow-up <strong>of</strong> 15.0 months in the<br />

17 patients still alive, the 12-month survival rate was 71% (95% CI: 49 –<br />

85). The median OS was 19 months (95% CI: 11 – no upper). Grade 3 or<br />

higher adverse events (regardless <strong>of</strong> attribution) were reported in 22 (82%)<br />

patients. Grade 4/5 adverse events were reported in 15 (56%) patients.<br />

There was one grade 5 event (pneumonitis). The most common (5 or more<br />

patients) grade 3/4 adverse events were fatigue (22%), leukopenia (63%),<br />

neutropenia (67%), and thrombocytopenia (44%). Conclusions: The addition<br />

<strong>of</strong> bortezomib resulted in high levels <strong>of</strong> severe hematologic toxicity, but<br />

also demonstrated evidence <strong>of</strong> activity with a 12-month survival rate <strong>of</strong><br />

71% and a median OS <strong>of</strong> 19 months. Further testing <strong>of</strong> this regimen in a<br />

larger study appears warranted.<br />

7075 General Poster Session (Board #39B), Sat, 1:15 PM-5:15 PM<br />

The Lung Cancer Symptom Scale (LCSS) as a prognostic indicator <strong>of</strong> overall<br />

survival in malignant pleural mesothelioma (MPM) patients: Post hoc<br />

analysis <strong>of</strong> a phase III study. Presenting Author: Ping Wang, Eli Lilly and<br />

Company, Indianapolis, IN<br />

Background: To determine the patients likely to benefit from therapy,<br />

clinicians seek factors associated with outcomes. This analysis investigates<br />

prognostic effect <strong>of</strong> baseline patient-reported symptoms on overall survival<br />

(OS) in the presence <strong>of</strong> demographic/clinical factors among MPM patients.<br />

Methods: Intent-to-treat patients (N�448) were included in the analysis.<br />

LCSS consists <strong>of</strong> 6 symptom items and 3 general items, each ranging from<br />

0 (no symptoms) to 100 (worst). Average symptom burden index (ASBI)<br />

was the mean <strong>of</strong> 6 symptom items. Patients were classified into subgroups<br />

based on ASBI: low symptom burden (LSB; ASBI�25) and high symptom<br />

burden (HSB; ASBI �25). Univariate Cox models were used to determine<br />

the prognostic effect <strong>of</strong> 7 demographic/clinical factors, 9 LCSS items, and<br />

ASBI on OS. Multivariate Cox model was used to determine the prognostic<br />

effect <strong>of</strong> ASBI in the presence <strong>of</strong> 7 demographic/clinical factors. Kaplan-<br />

Meier curves <strong>of</strong> OS were generated for patients with LSB and HSB and<br />

compared using both univariate and multivariate Cox models. Results: In<br />

the univariate analysis, significant prognostic effects on OS were observed<br />

for baseline Karn<strong>of</strong>sky performance status (KPS; 90-100 vs. 70-80, hazard<br />

ratio [HR]�0.45, p�0.0001) and baseline stage (I/II/III vs. IV, HR�0.63,<br />

p�0.0001). In addition, significant prognostic effects were observed for 5<br />

LCSS symptom items (anorexia, cough, dyspnea, fatigue, and pain), 3<br />

LCSS general items (interference with activity level, symptom distress, and<br />

quality <strong>of</strong> life; HRs�1.08-1.20 for every 10 points worsening, all p�0.02),<br />

as well as ASBI (HR�1.32, p�0.0001). In the multivariate Cox model,<br />

ASBI remained a significant prognostic factor <strong>of</strong> OS (HR�1.22, p�0.0001),<br />

along with baseline KPS and stage. Patients with LSB (N�265) had<br />

significantly better OS than those with HSB (N�181) (12.9 vs. 6.9<br />

months, HR�0.46, unadjusted p�0.0001; HR�0.59, p�0.0001 adjusted<br />

for the 7 demographic/clinical variables). Conclusions: The results<br />

suggest that baseline patient-reported symptoms as measured by LCSS<br />

provide distinct prognostic information in the presence <strong>of</strong> demographic and<br />

clinical measures in MPM.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


470s Lung Cancer—Non-small Cell Local-Regional/Small Cell/Other Thoracic Cancers<br />

7076 General Poster Session (Board #39C), Sat, 1:15 PM-5:15 PM<br />

NGR-hTNF as second-line treatment in malignant pleural mesothelioma<br />

(MPM). Presenting Author: Gilda Rossoni, Department <strong>of</strong> Oncology, Istituto<br />

Scientifico San Raffaele, Milan, Italy<br />

Background: NGR-hTNF exploits the asparagine-glycine-arginine (NGR)<br />

peptide for selectively targeting tumor necrosis factor (TNF) to cancer<br />

endothelial cells. Tumor hypervascularity is an independent predictor <strong>of</strong><br />

poor overall survival (OS) in MPM patients (pts). Methods: We report<br />

long-term results <strong>of</strong> a phase II trial that assessed NGR-hTNF in MPM pts<br />

with performance status (PS) � 2 and radiologic progressive disease (PD)<br />

after a pemetrexed-based regimen. NGR-hTNF was given at 0.8 �g/m2 every 3 weeks (q3w) in 43 pts and weekly (q1w) in 14 pts. Primary endpoint<br />

was progression free survival (PFS), with restaging done q6w by MPMmodified<br />

RECIST criteria, while secondary aims included disease control<br />

(DC) rate and OS. We also tested the impact on outcome <strong>of</strong> neutrophil to<br />

lymphocyte ratio (NLR) at baseline (median 3; interquartile range 2-5).<br />

Median follow-up was 32.5 months (95% CI 27.5-37.5). Results: Baseline<br />

characteristics were (n�57): median age 57 years; M/F 35/22; PS 0/1-2<br />

31/26; EORTC score good/poor 45/12. Median treatment free interval on<br />

prior therapy was 4.3 months. Only one drug-related grade � 3 adverse<br />

events (AEs) was noted, common grade 1/2 AEs being transient chills<br />

(75%). No higher toxicity was reported using the q1w schedule. Median<br />

PFS was 2.8 months (95% CI 2.2-3.3). DC rate was 46% (95% CI 32-59;<br />

26/57 pts; 1 partial response, 25 stable diseases) and was maintained for a<br />

median time <strong>of</strong> 4.7 months (95% CI 4.0-5.4). OS rates at 1 and 2 years<br />

were 47% and 16%, respectively. Median OS was longer in pts with DC<br />

than in those with early PD (16.2 and 8.3 months, respectively; p�.02).<br />

According to schedule, 6-month PFS rates were 11% and 36% and 2-year<br />

OS rates were 12% and 29% for q3w and q1w, respectively. In pts with DC,<br />

median PFS were 4.4 and 9.1 months and median OS were 13.3 and 24.8<br />

months for q3w and q1w, respectively. By Cox analyses, a PS <strong>of</strong> 0 (p�.01)<br />

and a low baseline NLR (p�.004) were the only variables associated with<br />

improved OS. Median OS in pts stratified by NLR � 2,3to4,and�5were 15.7, 10.5, and 4.2 months, respectively (p�.0002 for trend). Conclusions:<br />

NGR-hTNF showed promising PFS and OS in this study. A double-blind<br />

phase III trial is currently testing best investigator choice � NGR-hTNF<br />

q1w in pemetrexed-pretreated MPM pts (NCT01098266).<br />

7078 General Poster Session (Board #39E), Sat, 1:15 PM-5:15 PM<br />

Elevated PDGFRB gene copy number gain as prognostic in resected<br />

malignant pleural mesothelioma. Presenting Author: Anne S. Tsao, University<br />

<strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: PDGF/PDGFR pathway has been implicated in malignant<br />

pleural mesothelioma (MPM) carcinogenesis. We sought to evaluate the<br />

incidence <strong>of</strong> PDGFRB gene copy number gain (CNG) and PDGFR pathway<br />

protein expression by immunohistochemistry (IHC) in the tumor cell<br />

cytoplasm, membrane, nucleus, and stroma, and correlate it to patient<br />

clinical outcome. Methods: 88 archived tumor blocks from resected MPM<br />

with full clinical information were used to perform the analyses. IHC<br />

biomarkers for PDGFR�,� and p-PDGFR�, and fluorescence in situ<br />

hybridization were performed for analysis <strong>of</strong> PDGFRB gene CNG. Spearman’s<br />

rank correlation, Wilcoxon rank-sum test or Kruskal-Wallis test, BLiP<br />

plots, and Kaplan-Meier method were used to assess the biomarkers and<br />

their correlation to clinical outcome. Results: There were several correlations<br />

identified between the IHC biomarkers; however, none associated<br />

with patient demographics or histology subtype, with the exception <strong>of</strong> high<br />

cytoplasmic PDGFR� occurring in patients with no prior known asbestos<br />

exposure (p�0.029). In the CNG analysis, PDGFRB gene CNG in � 10% <strong>of</strong><br />

tumor cells had lower cytoplasmic p-PDGFR� (p�0.029), while PDGFRB<br />

gene CNG in � 40% <strong>of</strong> tumor cells had a higher cytoplasmic PDGFR�<br />

(p�0.04). PDGFRB gene CNG status did not associate with patient<br />

demographics or tumor characteristics. Patients with PDGFRB CNG �<br />

40% <strong>of</strong> tumor cells had an improved relapse-free survival (RFS) [HR 0.25<br />

(95% CI 0.09, 0.72), p�0.0096]. In the patients with PDGFRB CNG �<br />

40% <strong>of</strong> cells, the addition <strong>of</strong> chemotherapy appeared to also improve RFS<br />

(p�0.017). In the multi-covariate analyses for RFS, there was no association<br />

with any IHC biomarker. In the overall survival (OS) analysis, having<br />

PDGFRB gene CNG � 40% <strong>of</strong> tumor cells correlated with an improved OS<br />

[HR 0.32 (95% CI 0.11, 0.89), p�0.029] and the addition <strong>of</strong> perioperative<br />

chemotherapy led to a trend towards improved OS (p�0.089).<br />

Conclusions: PDGFRB CNG � 40% <strong>of</strong> tumor cells is a potential prognostic<br />

biomarker in surgically resected MPM tumors. Adding chemotherapy to<br />

patients with PDGFRB CNG � 40% <strong>of</strong> cells improved RFS and led to a<br />

trend towards improved OS. Future validation <strong>of</strong> this biomarker in prospective<br />

trials is needed.<br />

7077 General Poster Session (Board #39D), Sat, 1:15 PM-5:15 PM<br />

Phase II trial <strong>of</strong> anti-transforming growth factor-beta (TGFß) monoclonal<br />

antibody GC1008 in relapsed malignant pleural mesothelioma (MPM).<br />

Presenting Author: James Stevenson, University <strong>of</strong> Pennsylvania, Philadelphia,<br />

PA<br />

Background: TGF� is a pleiotropic cytokine overexpressed by MPM. Based<br />

on preclinical data documenting a key role for TGF� in promoting growth<br />

and progression <strong>of</strong> MPM, we are conducting a phase II trial <strong>of</strong> GC1008 in<br />

patients (pts) with progressive MPM. Methods: Pts with progressive MPM by<br />

modified RECIST criteria and PS 0-1 with 1-2 prior systemic therapies (at<br />

least 1 pemetrexed-based) are eligible. Treatment plan: GC1008 3mg/kg<br />

IV over 90 minutes every 21 days. Responses are assessed by modified<br />

RECIST every 6 weeks. The primary endpoint is progression-free survival<br />

(PFS) rate at 3 months; secondary objectives include safety with GC1008,<br />

response rate by modified RECIST, time to progression (TTP), and overall<br />

survival (OS). Results: The modified Gehan stage 1 stopping criterion <strong>of</strong><br />

1/13 pts with 3 month PFS has been exceeded. To date, 13 pts (10 PS 0; 3<br />

PS 1) with MPM (median age 69; 2F, 11M; 11 epithelial, 1 sarcomatoid, 1<br />

biphasic) enrolled. Treatment-related toxicities include G1/2 fatigue (3<br />

pts), nausea (1 pt) and xerosis (1 pt). Other adverse events possibly related<br />

to GC1008 were rapid disease progression in 1 pt after 2 cycles, and G2<br />

skin keratoacanthoma in 1 pt after 5 cycles. Three pts met the primary<br />

objective <strong>of</strong> 3 month PFS at 4.1, 4.2 and 9 months each. Stable disease<br />

(SD) was seen in 3 pts (23%). Median TTP is 1.4 months (95% CI 1.2-�);<br />

median OS is 13 months (95% CI 6-�). Increased serum mesothelin levels<br />

have closely tracked disease progression. Serum from 6/13 pts showed new<br />

antibodies against MPM tumor lysates as measured by immunoblotting.<br />

Two <strong>of</strong> 3 pts with SD had anti-tumor antibody responses. Mean baseline<br />

plasma level <strong>of</strong> TGF� was 2447 pg/ml but did not correlate with baseline<br />

plasma TGF� or TTP. Conclusions: GC1008 was well tolerated in pretreated<br />

MPM patients. SD occurred in 3 pts, all with prior disease progression.<br />

Evidence for humoral anti-tumor immunity was seen in nearly half <strong>of</strong><br />

enrollees and in 2 <strong>of</strong> 3 pts with SD. OS compares favorably to prior<br />

single-agent studies in pretreated MPM.<br />

7079^ General Poster Session (Board #39F), Sat, 1:15 PM-5:15 PM<br />

Phase II trial <strong>of</strong> BNC105P as second-line chemotherapy for advanced<br />

malignant pleural mesothelioma (MPM): Australasian Lung Cancer Trials<br />

Group and NHMRC <strong>Clinical</strong> Trials Centre Collaboration. Presenting Author:<br />

Anna K. Nowak, School <strong>of</strong> Medicine and Pharmacology, University <strong>of</strong><br />

Western Australia, Perth, Australia<br />

Background: BNC105P is a tubulin polymerization inhibitor that acts as a<br />

Vascular Disrupting Agent (VDA), has direct cytotoxic effects, and had<br />

preclinical and phase I activity in MPM. This aim <strong>of</strong> this study was to<br />

determine activity and safety <strong>of</strong> BNC105P as second-line therapy after<br />

pemetrexed and a platin in MPM. Methods: Eligible patients had progressive<br />

MPM, prior pemetrexed and platinum, measurable disease by modified<br />

RECIST, ECOG 0-1, and adequate organ and cardiovascular function.<br />

Important exclusions included recent thromboembolic, cardiovascular or<br />

cerebrovascular disease, or therapeutic anticoagulation. Pts received<br />

BNC105P (16 mg/m2 IV) Day 1 � 8 q21d until progression or toxicity. The<br />

primary endpoint was centrally reviewed objective tumour response rate<br />

(RR); the Simon 2-stage design assumed a RR <strong>of</strong> interest <strong>of</strong> 20% and a RR<br />

<strong>of</strong> no interest <strong>of</strong> 5%, with � � � � 0.05. Continuation past first stage<br />

accrual required �1 objective response in 24 patients. Results: 30 subjects<br />

were accrued over 10 months (90% male; median age 65 (range 41-83);<br />

77% ECOG PS 1; histology epithelioid (67%), biphasic (10%), sarcomatoid<br />

(7%), other/unspecified (17%)). All pts received at least one dose <strong>of</strong><br />

study drug; pts received a median <strong>of</strong> 2 cycles and median dose intensity<br />

was 100%. No significant haematologic, biochemical, peripheral neurotoxic<br />

or cardiac adverse events (AEs) including hypertension were observed.<br />

Grade 3 or 4 AEs occurred in 10 pts (33%). There were 2 deaths on study: 1<br />

due to stroke, the other due to pneumonia and respiratory failure. We<br />

observed 1 partial response (3%) and 13 pts with SD as their best response<br />

(43%). Median progression free survival was 1.5 mo (95% CI 1.4-2.4);<br />

median overall survival was 8.7 mo (95% CI 3.8-NR). Lung function and<br />

QOL data was collected. Biomarker analyses correlating to pharmacological<br />

changes induced by BNC105P are ongoing and will be presented.<br />

Conclusions: BNC105P was safe and tolerable but its single agent response<br />

rate failed to meet the pre-specified primary endpoint <strong>of</strong> interest.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


Lung Cancer—Non-small Cell Local-Regional/Small Cell/Other Thoracic Cancers<br />

7080 General Poster Session (Board #39G), Sat, 1:15 PM-5:15 PM<br />

Circulating tumor cell (CTC) as a significant clinical marker in epithelioidtype<br />

malignant pleural mesothelioma (MPM). Presenting Author: Kazue<br />

Yoneda, Hyogo College <strong>of</strong> Medicine, Nishinomiya, Japan<br />

Background: Circulating tumor cell (CTC) is a surrogate <strong>of</strong> distant metastasis,<br />

and our preliminary study suggested that CTC detected by an<br />

EpCAM-based immuno-magnetic separation system (“CellSearch”) was a<br />

useful clinical marker in the diagnosis <strong>of</strong> malignant pleural mesothelioma<br />

(MPM) (Tanaka F. et al ASCO 2008). Methods: Patients who presented at<br />

our institute to receive pleural biopsy with suspicion <strong>of</strong> MPM were<br />

prospectively enrolled. CTCs in 7.5mL <strong>of</strong> peripheral blood were quantitatively<br />

evaluated with the CellSearch system. Results: Among 136 eligible<br />

patients, 104 were finally diagnosed with MPM, and 32 were with<br />

non-malignant diseases (NM). CTC was positive (CTC�1) in 32.7%<br />

(37/104) <strong>of</strong> MPM pts, and in 9.4% (3/32) <strong>of</strong> NM pts (p�0.011).<br />

CTC-count was significantly higher in MPM (range, 0-9) than in NM (range,<br />

0-1; p�0.007). According to a ROC curve analysis, the CTC-test provided a<br />

significant diagnostic performance in discrimination between MPM and<br />

NM (AUC� 0.623; P�0.036). Among MPM pts, CTC-positivity and<br />

CTC-count were significantly increased with tumor progression (p�0.026<br />

and p�0.008, respectively). For all MPM pts, there was no significant<br />

difference in overall survival between CTC-positive and negative pts.<br />

However, in a planned subset analysis, CTC was a significant factor to<br />

predict poor prognosis (median survival time, 8.0 months for CTC-positive<br />

pts, and 20.3 months for negative pts; p�0.012) in pts with epithelioidtype<br />

MPM in which CTC was exclusively positive; a multivariate analysis<br />

confirmed that CTC, along with PS, was an independent prognostic factor<br />

(HR�2.38; P�0.006). Conclusions: CTC was a significant diagnostic<br />

marker in discrimination between MPM and NM. CTC-positivity was a<br />

significant and independent prognostic factor to predict a poor prognosis <strong>of</strong><br />

epithelioid MPM.<br />

Epithelioid Non-epithelioid<br />

No. <strong>of</strong> pts 79 25<br />

CTC-positive pts 32 (40.5%) 2 (8.0%)<br />

OS (median) 8.0m (CTC�1) vs. 20.3m 3.3m (CTC�1) vs. 5.7m<br />

(CTC�0)<br />

(CTC�0)<br />

(p�0.012)<br />

(p�0.482)<br />

7082 General Poster Session (Board #40A), Sat, 1:15 PM-5:15 PM<br />

Malignant pleural mesothelioma (MPM) in elderly patients: A multicenter<br />

survey. Presenting Author: Federica Grosso, Oncology SS Antonio e Biagio<br />

General Hospital, Alessandria, Italy<br />

Background: The incidence <strong>of</strong> malignant pleural mesothelioma (MPM) in<br />

elderly patients (pts) is increasing in Western countries. Elderly pts with<br />

MPM are under-represented in clinical trials, and there are no specific<br />

guidelines for their management. The aim <strong>of</strong> this study was to perform a<br />

retrospective survey on this patient population in four Oncology Departments<br />

with high MPM accrual and expertise. Methods: The clinical records<br />

<strong>of</strong> elderly pts (�70 years old) with MPM referred to the participating<br />

centers from January 2005 to November 2011 were reviewed. For each<br />

patient, age and gender, histology, Eastern Cooperative Oncology Group<br />

Performance Status (ECOG-PS), Charlson Comorbidity Index (CCI) and<br />

treatment modalities were collected. The study endpoint was overall<br />

survival (OS). Results: Out <strong>of</strong> a total <strong>of</strong> 610 cases, 210 elderly pts were<br />

identified (34% <strong>of</strong> the whole MPM population observed in the study<br />

period). Pts characteristics were: median age 75 yrs (range 70-92), M/F<br />

132/78, epithelial/non-epithelial histology 140/70, ECOG-PS 0/1/2/<br />

unknown 130/67/9/4. CCI was 0 in 128 pts (61%), �4 in 59 pts (28%).<br />

Treatment was multimodality therapy including surgery in 16, chemotherapy<br />

in 153 (73%) and best supportive care only in 41 pts (19%).<br />

Chemotherapy was mainly pemetrexed-based. Median OS was 11.3 months.<br />

Non-epithelial histology, age �75 yrs and the presence <strong>of</strong> comorbidities<br />

(CCI �1, p�.003; CCI �4, p�0.0001) were significantly correlated to a<br />

shorter OS. Conclusions: Pemetrexed-based chemotherapy is feasible in<br />

selected elderly pts with MPM. Comorbidity is a significant prognostic<br />

factor, and should be carefully considered in patient selection. Prospective<br />

dedicated trials in elderly pts with MPM selected according to comorbidity<br />

scales are warranted.<br />

471s<br />

7081 General Poster Session (Board #39H), Sat, 1:15 PM-5:15 PM<br />

Phase II study <strong>of</strong> bortezomib with cisplatin as first-line treatment <strong>of</strong><br />

malignant pleural mesothelioma (MPM): EORTC 08052. Presenting Author:<br />

Mary O’Brien, The Royal Marsden Hospital, Sutton, United Kingdom<br />

Background: Cisplatin is one <strong>of</strong> the most active drugs available in MPM<br />

while bortezomib has shown some activity in single agent phase II studies<br />

against MPM. This was a prospective phase II study <strong>of</strong> cisplatin and<br />

bortezomib (CB) in the first line treatment <strong>of</strong> MPM. Methods: Patients with<br />

histological proven MPM, with performance status (PS) 0/1, were eligible.<br />

The doses were cisplatin 75mg/m2 /3 wks and bortezomib 1.3mg/m2 day 1,<br />

4, 8, 11 every 3 wks. The primary end-point was progression free survival<br />

rate at 18 wks (PFSR�18). The 2-stage Simon design (a�0.1; b � 0.05,<br />

P0�0.50 and P1�0.675) was used. In the first step <strong>of</strong> the study 37 eligible<br />

patients were planned. If more than 19 patients were alive and free <strong>of</strong><br />

progression at 18 wks the total sample size was increased to 76 eligible<br />

patients. Results: Between 2007 and 2010 82 patients were entered. The<br />

median follow-up time is 32.3 months The median age was 55 years<br />

(range: 22-77yrs), male/female: 55/27 , PS 0/1: 9/73, Stage T1: 10%; T2:<br />

42%, T3: 25%; T4: 23% and N0: 57%; N1: 4%; N2: 33%; N3: 6%. The<br />

median number <strong>of</strong> cycles received was 4 and 38% received 6 cycles.<br />

Cisplatin/ bortezomib dose intensity was 98/ 80%. Toxicity (grade 3/4):<br />

neutropenia 10%, thrombocytopenia 11%, anaemia 1%. Grade 3-4<br />

hyponatraemia/ hypokalaemia occurred in 46/ and 17%. Grade 2 tinnitus,<br />

grade 3 fatigue occurred in 16%, and 12%, <strong>of</strong> patients. Motor/sensory/<br />

other neurotoxicity was grade 1: 6/28/7%, grade 2: 2/26/2% and grade 3:<br />

1/7/2% respectively. There were 2 toxic deaths at 32 and 74 days due to<br />

acute pneumonitis and cardiac arrest. The PFRS-18 (including symptomatic<br />

progression) was 53% (80% confidence intervals, CI, 42-64%). The<br />

overall survival was 13.5 months (95% CI 10.5-15) with 56% (95% CI<br />

44-66%) alive at 1 year. The PFS was 5.1 months (95% CI 3.3-6.5).<br />

Conclusions: On the basis <strong>of</strong> the PFRS-18, the null hypothesis could not be<br />

rejected, although CB gave predictable toxicity and was as active as other<br />

reported regimens in MPM.<br />

7083 General Poster Session (Board #40B), Sat, 1:15 PM-5:15 PM<br />

SWOG 0722: A phase II study <strong>of</strong> mTOR inhibitor everolimus (RAD001) in<br />

malignant pleural mesothelioma (MPM). Presenting Author: Linda L.<br />

Garland, Arizona Cancer Center, Tucson, AZ<br />

Background: MPM preclinical studies demonstrate activation <strong>of</strong> AKT<br />

pathway proteins, including mTOR, and provide the rationale to test<br />

everolimus, an mTOR inhibitor, in MPM. Methods: Unresectable MPM<br />

patients (pts) after 1-2 systemic regimens (including a platinum-based<br />

regimen), with PS 0-1, measurable disease and adequate organ function<br />

were treated with daily oral everolimus 10 mg. Study endpoints were<br />

progression-free survival (PFS) at 4 months, response rate,overall survival<br />

(OS), and frequency/severity <strong>of</strong> toxicities. Results: 61 pts were registered<br />

between 12/2008 and 9/2011; there were 57 evaluable pts for the primary<br />

endpoint <strong>of</strong> 4-month PFS. Median age was 65.9 yrs; M/F 43/14; prior<br />

regimens (1/2: 58%/42%); PS 0/1 (21%/79%); epithelial subtype 61%;<br />

biphasic 7%; NOS 28%; pending 4%. 5 pts remain on treatment.<br />

Preliminary data for 35 <strong>of</strong> 57 pts are as follows: RR 0% by standard RECIST<br />

and DCR <strong>of</strong> 14/35 (40%); data by mod RECIST are not yet available.<br />

Estimated 4-month PFS was 34% with an estimated median PFS <strong>of</strong> 3<br />

months. The null hypothesis <strong>of</strong> 4-month PFS � 30% could not be rejected<br />

(p�0.28). 41 pts have died; median OS is estimated at 5 months. Frequent<br />

grade 1-3 toxicities were fatigue (55.4%), hypertriglyceridemia (41.1%),<br />

anemia (39.3%), nausea (33.9%), oral mucositis (32.1%), anorexia<br />

(32.1%), diarrhea (26.8%), and hyperglycemia (26.8%). One pt each had<br />

grade 4 and grade 5 dyspnea. Conclusions: The trial did not achieve the<br />

primary endpoint <strong>of</strong> an improvement <strong>of</strong> PFS at 4 months from 30% to 50%.<br />

Inhibition <strong>of</strong> mTOR using single agent everolimus is not active in unselected<br />

MPM patients. Other strategies for targeting the activated AKT<br />

pathway in MPM remain <strong>of</strong> interest.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


472s Lung Cancer—Non-small Cell Local-Regional/Small Cell/Other Thoracic Cancers<br />

7084 General Poster Session (Board #40C), Sat, 1:15 PM-5:15 PM<br />

Outcomes <strong>of</strong> sarcomatoid malignant pleural mesothelioma, a distinct<br />

clinical entity. Presenting Author: Elisabetta Gattoni, Oncology ASL21,<br />

Casale Monferrato, Italy<br />

Background: Sarcomatoid malignant pleural mesotheliomas (SMPM), accounting<br />

for 10% <strong>of</strong> MPM, are resistant to chemotherapy (CT) with median<br />

survival in the range <strong>of</strong> 6 months. Here we report on clinical outcome <strong>of</strong> a<br />

large series <strong>of</strong> SMPM patients treated at our Oncological Department,<br />

located in a particularly asbestos-polluted area in Piedmont (Italy).<br />

Methods: Patients consecutively diagnosed, treated and included in our<br />

MesoDB between 1993 and 2010 were considered for this study. <strong>Clinical</strong><br />

and pathological data, treatments, response to CT and outcomes were<br />

analyzed for the present study. Diagnosis was always confirmed by the same<br />

expert pathologist. Results: Data <strong>of</strong> 672 patients were considered for the<br />

analysis. Among these, 53 (8%) were diagnosed with SMPM (17F/36M).<br />

Median age was 67,6 years. Asbestos exposure was occupational certain in<br />

12, possible in 15, domestic in 3 and environmental in 23. The predominant<br />

symptom at diagnosis was thoracic pain in 34 patients (64%),<br />

followed by dyspnea in 16 (30%), fatigue and weight loss in 2 and palpable<br />

mass in 1. The predominant radiological finding at diagnosis was diffuse<br />

pleural thickening in 23 patients (43%), pleural effusion in 14 (26%),<br />

pleural effusion and pleural thickening in 10 (19%), localized mass in 4<br />

(8%), unknown in 2 (4%). Forty-eight patients received first line CT with<br />

the following regimens: carboplatin/cisplatin and pemetrexed in29, pemetrexed<br />

in 8, cisplatin and raltitrexed in 3, cisplatin and gemcitabine in 3,<br />

gemcitabine in 2, doxorubicin based combination in 3 patients. The<br />

median number <strong>of</strong> cycles was 4. Overall response rate to first line CT was as<br />

follows: 11 SD (21%), 31 PD (58%) and 6 (11%) patients were not<br />

evaluable for response. Median progression free survival was 3.3 (95%CI<br />

2.6-4.1) months. Eleven patients received a second line CT and no<br />

responses or disease stabilization was observed. Median OS was 7.6<br />

months. Conclusions: This large series <strong>of</strong> SMPM suggests that standard CT<br />

has only negligible impact on the prognosis, underlying a highly unmet<br />

clinical need. SMPM patients should be treated within phase I-II studies<br />

with investigational agents. Research should focus to a deeper knowledge<br />

<strong>of</strong> tumor biology and to the identification <strong>of</strong> druggable targets.<br />

7086 General Poster Session (Board #40E), Sat, 1:15 PM-5:15 PM<br />

Phase II study <strong>of</strong> amrubicin (AMR) combined with carboplatin (CBDCA) for<br />

refractory relapsed small cell lung cancer (SCLC): North Japan Lung Cancer<br />

Group 0802. Presenting Author: Ryota Saito, Tohoku University Hospital,<br />

Sendai, Japan<br />

Background: AMR, a new anthracycline agent, has achieved some promising<br />

results for advanced SCLC both in the first-line and the second-line<br />

setting. However the efficacy <strong>of</strong> AMR alone against refractory relapsed<br />

SCLC was relatively low in previous studies. This study was conducted to<br />

evaluate the safety and efficacy <strong>of</strong> the combination <strong>of</strong> AMR plus CBDCA in<br />

patients with refractory relapsed SCLC. Methods: Patients with advanced<br />

SCLC who relapsed within 90 days after the completion <strong>of</strong> first-line<br />

chemotherapy received AMR (30 mg/m2 , day1-3) and CBDCA (AUC 4.0,<br />

day 1) every 3 weeks. The primary endpoint <strong>of</strong> this study was overall<br />

response rate (ORR), and secondary endpoints were progression-free<br />

survival (PFS), overall survival and toxicity pr<strong>of</strong>ile. Assuming that ORR <strong>of</strong><br />

45% in eligible patients would indicate potential usefulness while ORR <strong>of</strong><br />

20% would be the lower limit <strong>of</strong> interest, with alpha � 0.10 and beta �<br />

0.10, at least 24 patients were required. Results: From September 2008 to<br />

May 2011, 30 patients were enrolled from 10 institutions. One patient was<br />

excluded because <strong>of</strong> ineligible histology. Patient characteristics were:<br />

Male/Female 26/3; median age 67 (range 50-81); Performance status<br />

0/1/2 9/16/4. The median number <strong>of</strong> treatment cycles were 4 (range 1-7).<br />

The objective responses evaluated by RECIST were: CR 0, PR 10, SD 14,<br />

PD 5. The ORR was 34% and the disease control rate was 83%. Median<br />

PFS was 3.5 months and median survival time was 7.3 months. Grade 3-4<br />

neutropenia was observed in 23 patients (79%) and grade 3-4 thrombocytopenia<br />

was observed in 7 patients (24%). One patient (3%) suffered from<br />

grade 3-4 febrile neutropenia. Other grade 3 non-hematological toxicities<br />

such as infection, interstitial lung disease, hyponatremia, hypoglycemia,<br />

were observed in 7 patients (24%). No treatment related death was<br />

observed. Conclusions: This is the first prospective study <strong>of</strong> AMR combined<br />

with CBDCA for refractory relapsed SCLC, which was effective and well<br />

tolerated. Further investigation <strong>of</strong> this treatment is warranted.<br />

7085 General Poster Session (Board #40D), Sat, 1:15 PM-5:15 PM<br />

NGR-hTNF and doxorubicin in relapsed small-cell lung cancer (SCLC).<br />

Presenting Author: Raffaele Cavina, Humanitas Cancer Center, Rozzano,<br />

Italy<br />

Background: By selectively damaging tumor vasculature, NGR-hTNF (asparagine-glycine-arginine<br />

human tumor necrosis factor) is able to improve<br />

intratumoral uptake <strong>of</strong> doxorubicin (D). A phase I trial selected NGR-hTNF<br />

0.8 �g/m2 /day(d)1 and D 75 mg/m2 /d1 every 3 weeks (q3w) for phase II.<br />

Methods: SCLC pts failing one or more platinum-based regimen received<br />

NGR-hTNF until progressive disease (PD) and D up to 550 mg/m2 . This<br />

phase II trial (n�27 pts) had progression-free survival (PFS) as primary end<br />

point, with tumor response assessed q6w by RECIST, while secondary end<br />

points included adverse events (AEs), disease control rate (DCR), and<br />

overall survival (OS). Results: Among 28 pts enrolled (median age 63 years;<br />

M/F 19/9; PS 0/1-2 13/15; prior lines 1/2-3 20/8), 16 pts had platinumresistant<br />

(R) disease (PD�3 months) and 12 pts platinum-sensitive (S)<br />

disease (PD�3 months). At baseline, median neutrophil to lymphocyte<br />

ratio (NLR) was 4 (range 1-20). A total <strong>of</strong> 114 cycles were given (range<br />

1-10), with 13 pts (46%) having � 4 cycles and 9 pts (32%) � 6 cycles.<br />

No grade 3/4 AEs related to NGR-hTNF were noted, while grade 1/2 AEs<br />

were transient chills (61%). NGR-hTNF did not apparently increase<br />

D-related AEs. Median PFS was 3.2 months (95% CI 2.6-3.8) and 1-year<br />

OS rate was 34%. Overall, DCR was 55% (95% CI 35-74), comprising 6<br />

partial responses (22%; median duration: 6.3 months) and 9 stable<br />

diseases (33%; median duration: 4.1 months). Mean changes from<br />

baseline in target tumor size after 2, 4, and 6 cycles were -9%, -29%, and<br />

-32%, respectively for R disease and -11%, -20%, and -43%, respectively<br />

for S disease. In pts pretreated with � 2 lines, DCR was 75%, median PFS<br />

was 5.1 months, and 1-year OS rate was 44%. In pts with R or S disease,<br />

DCR were 50% and 58% (p�.72), median PFS were 2.7 and 4.1 months<br />

(p�.07), and 1-year OS rates were 27% and 42% (p�.65), respectively. At<br />

multivariate analyses, PFS was not related to baseline characteristics,<br />

while an improved OS was associated only with a low NLR. The 1-year OS<br />

rate was 48% in pts with NLR � 4 and 10% in pts with NLR � 4(p�.007),<br />

while in pts with NLR � 4, 1-year OS rate was 46% in R disease and 50%<br />

in S disease. Conclusions: This combination is well tolerated and active in<br />

platinum resistant and sensitive SCLC and further development is <strong>of</strong><br />

interest.<br />

7087 General Poster Session (Board #40F), Sat, 1:15 PM-5:15 PM<br />

<strong>Clinical</strong> trial design in small cell lung cancer: Surrogate endpoints and<br />

statistical evolution. Presenting Author: Myles Nickolich, Case Comprehensive<br />

Cancer Center, Cleveland , OH<br />

Background: Small cell lung cancer (SCLC) is a devastating disease for<br />

which little recent therapeutic advance has been achieved. SCLC trial<br />

design and reporting may have an impact on the interpretation <strong>of</strong> studies<br />

conducted. Furthermore the use <strong>of</strong> surrogate endpoints in SCLC has not<br />

been explored. Methods: Through examining all phase II and III SCLC trials<br />

published in the Journal <strong>of</strong> <strong>Clinical</strong> Oncology (8,471 patients from 66 trials<br />

between 1983-2010), we examined how SCLC trial reporting and design<br />

has evolved, determining if the type I error, power, and sample size<br />

calculations were provided. We assessed primary endpoints for all trials and<br />

sought to discover if response rate (RR) or progression-free survival (PFS)<br />

correlated with overall survival (OS). We analyzed response and survival<br />

outcomes for associations using Pearson correlation coefficient and differences<br />

between data groups by ANOVA followed by Tukey’s multiple<br />

comparison procedure. Results: There were increased reporting trends <strong>of</strong><br />

statistical design in power (16.7% in 1986-1996 to 77.8% <strong>of</strong> trials in<br />

2006-2010 [p�0.001]) and type I error (22.2% in 1986-1996 to 72.2%<br />

in 2006-2010 [p�0.005]). 72.2% <strong>of</strong> trials published in 1986-1996<br />

failed to report a primary endpoint whereas only 5.56% <strong>of</strong> trials in<br />

2006-2010 failed to do so (p�0.004). Of phase II trials, primary endpoint<br />

was identified as RR in 65%, OS in 25%, and PFS in 10% (p�0.0001).<br />

There is a strong correlation between RR and both PFS (p�0.013) and OS<br />

(p�0.012) in extensive-stage disease (ED) but not limited-stage disease<br />

(LD) (p�0.978 for PFS, p�0.193 for OS). This correlation is for partial<br />

response rate (pRR) but not complete response rate. RR (p�0.029)<br />

exhibits a negative trend over time with a dramatic and significant decrease<br />

in RR across all studies starting in 2005. A strong positive correlation exists<br />

between PFS and OS for LD (p�0.036) and ED (p�0.058). We found no<br />

change in median overall survival (p�0.383). Conclusions: Over time there<br />

has been improvement in reporting the essential statistical data for proper<br />

interpretation <strong>of</strong> SCLC trials. No change in survival was seen over the past<br />

28 years. RR should be evaluated as a surrogate endpoint for OS in ED but<br />

not LD. pRR correlates with OS and PFS in both LD and ED.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


Lung Cancer—Non-small Cell Local-Regional/Small Cell/Other Thoracic Cancers<br />

7088 General Poster Session (Board #40G), Sat, 1:15 PM-5:15 PM<br />

Efficacy <strong>of</strong> rechallenge chemotherapy in patients with sensitive relapsed<br />

small cell lung cancer. Presenting Author: Kazushige Wakuda, Division <strong>of</strong><br />

Thoracic Oncology, Shizuoka Cancer Center, Shizuoka, Japan<br />

Background: Treatment efficacy <strong>of</strong> rechallenge chemotherapy recommended<br />

for patients with sensitive-relapse small cell lung cancer (SCLC)<br />

has not been fully clarified. Methods: We defined sensitive relapse as<br />

treatment-free interval (TFI) � 90 days. Sixty-five sensitive-relapse SCLC<br />

patients who received second-line chemotherapy at the Shizuoka Cancer<br />

Center between September 2002 and May 2011 were separated into those<br />

treated with rechallenge chemotherapy (rechallenge group) and those<br />

treated with other regimens (other group) for comparison and analysis <strong>of</strong><br />

treatment efficacy. Results: No significant differences in age, gender, ECOG<br />

performance status at relapse, disease extent at diagnosis, or response to<br />

first-line treatment were found between the two groups, but TFI was<br />

significantly longer in the rechallenge group, which included 19 sensitiverelapse<br />

patients. The other group included 46 sensitive-relapse patients,<br />

21 <strong>of</strong> whom received amrubicin. There was no significant difference in<br />

overall survival (OS) between the two groups (median survival time (MST):<br />

rechallenge group 14.4 months, other group 13.1 months, p � 0.51). In<br />

the patients treated with amrubicin, MST was 12.6 months. Comparing the<br />

rechallenge group with the patients treated with amrubicin, there was also<br />

no significant difference in OS (p � 0.38). Both the rechallenge and other<br />

group included 11 patients with ex-sensitive relapse (TFI � 180 days).<br />

There was no significant difference in OS between the two groups (MST<br />

15.7 vs. 26.9 months, p � 0.46). Conclusions: Rechallenge chemotherapy<br />

did not prove superior to other chemotherapies, suggesting that monotherapy,<br />

such as amrubicin, might be reasonable as second-line chemotherapy<br />

for sensitive-relapse SCLC patients.<br />

7090 General Poster Session (Board #41A), Sat, 1:15 PM-5:15 PM<br />

Phase II trial <strong>of</strong> huKS-IL2 with cyclophosphamide (CTX) in patients with<br />

extensive disease small-cell lung cancer (ED-SCLC). Presenting Author:<br />

Oleg Gladkov, Chelyabinsk Regional <strong>Clinical</strong> Oncology Dispensary, Chelyabinsk,<br />

Russia<br />

Background: huKS-IL2 immunocytokine is a humanized antibody specific<br />

for EpCAM, fused at its Fc end to two molecules <strong>of</strong> IL2. Results <strong>of</strong> a phase<br />

1b study <strong>of</strong> huKS-IL2 plus low-dose CTX, and preclinical data, provided a<br />

rationale to evaluate this combination in SCLC, which is <strong>of</strong>ten EpCAMpositive.<br />

Methods: Patients (pts) with ED-SCLC responding (PR/CR) to 4<br />

cycles <strong>of</strong> first-line Pt-based chemotherapy were randomized 1,5:1 to<br />

receive huKS-IL2/CTX or best supportive care (BSC). Pts in the huKS-IL2/<br />

CTX arm received six 21-day cycles <strong>of</strong> CTX 300 mg/m2 on day (d) 1, and 1.5<br />

mg/m2 /d huKS-IL2 on d2–4, followed by 21-d cycles <strong>of</strong> CTX on d1 and<br />

huKS-IL2 on d2 until progression. Pts were stratified for prophylactic<br />

cranial irradiation (PCI) and response to chemotherapy. Primary endpoint<br />

was PFS rate at 6 months (mo). Secondary endpoints included OS rates at<br />

12 and 18 mo from start <strong>of</strong> Pt-based chemotherapy, median PFS and OS,<br />

safety, and immunogenicity. Results: 108 pts (64 huKS-IL2/CTX arm, 44<br />

BSC arm) were randomized and treated. Baseline characteristics were<br />

balanced between arms; however, higher % <strong>of</strong> males (75 vs 61.4%) and <strong>of</strong><br />

ECOG PS 0 (45.3 vs 38.6%) were observed in the active arm. Median age<br />

was 61.7 (32; 81) and 59.2 (45; 74) years in the active and BSC arm,<br />

respectively. Most pts were in PR at randomization (2 pts–1ineach arm –<br />

had CR). PCI was used in 15 (23.4%) and 11 (25%) pts in the active and<br />

BSC arm, respectively. No significant differences in PFS or OS were<br />

observed in the active vs BSC arm: PFS rate was 6.4 vs 12.2%; median PFS<br />

was 1.5 vs 1.4 mo; OS rates at 12 and 18 mo were 52 vs 61% and 24 vs<br />

29%, respectively; median OS was 12.3 vs 14.1 mo. One PR (45%<br />

reduction vs randomization baseline) was observed in the active arm. In a<br />

subset <strong>of</strong> pts who received PCI, median PFS was 1.7 vs 1.5 mo, median OS<br />

21.5 vs 14.3 mo in the active vs BSC arm, respectively. AEs observed more<br />

frequently in the active arm were flu-like symptoms, rash, hypotension,<br />

lymphopenia, LFT and creatinine elevations; all Grade 3/4 AEs related to<br />

huKS-IL2 were reversible/manageable. Conclusions: HuKS-IL2/CTX was<br />

well tolerated, but showed no benefit in pts with ED-SCLC in PR/CR after<br />

chemotherapy. A trend for improved PFS and OS was observed in pts who<br />

received prior PCI.<br />

473s<br />

7089 General Poster Session (Board #40H), Sat, 1:15 PM-5:15 PM<br />

Genome-wide association study <strong>of</strong> survival in small cell lung cancer<br />

patients treated with irinotecan and cisplatin chemotherapy. Presenting<br />

Author: Ji-Youn Han, Center for Lung Cancer, National Cancer Center,<br />

Goyang, South Korea<br />

Background: The prognosis <strong>of</strong> patients with extensive-disease small-cell<br />

lung cancer (ED-SCLC) remains poor. Despite a high initial response rate to<br />

chemotherapy, most patients die from rapid recurrence. We conducted a<br />

genome-wide analysis study (GWAS) to examine whether germline genetic<br />

variations are prognostic factors in ED-SCLC patients treated with irinotecan<br />

and cisplatin (IP) chemotherapy. Methods: We prospectively collected<br />

blood samples from 139 patients who participated in two phase II studies<br />

<strong>of</strong> IP chemotherapy as first-line therapy and conducted a GWAS using<br />

overall survival (OS) as the endpoint. Germline DNA was genotyped using<br />

the Affymetrix 5.0 or 6.0 array sets. Associations between OS and single<br />

nucleotide polymorphisms (SNPs) were investigated using multivariate Cox<br />

regression analysis. Adjustments for age, performance status (PS) and<br />

gender were made. We selected most promising SNPs with p�1�10�6 in<br />

the GWAS allelic association analyses. Results: A total <strong>of</strong> 426,019 SNPs<br />

were genotyped and 343,530 SNPs passed quality control (HWE, p�10-7 ,<br />

minor allele frequency�1%). We found 7 SNPs showing a significant<br />

association with OS. Patients harboring rs16950650 CT, rs7186128 AG<br />

or GG, rs17574269 AG, rs8020368 CC, rs4655567 CC, rs2166219 TT<br />

and rs2018683 TT genotypes showed shorter OS compared to patients<br />

with control alleles. Hazards ratios <strong>of</strong> death for each risk variants were 30.2<br />

(95% CI, 8.3-109.0, p�2.1X10-7 ), 2.5 (95% CI, 1.7-3.5, p�3.8X10-7 ),<br />

7.6 (95% CI, 3.4-16.6, p�4.5X10-7 ), 3.4 (95% CI, 2.1-5.5, p�3.5X10-7 ),<br />

4.9 (95% CI, 2.7-9.0, p�3.1X10-7 ), 5.8 (95% CI, 2.9-11.7, p�8.4X10-7 )<br />

and 7.8 (95% CI, 3.4-17.7, p�8.0X10-7 ), respectively. Among these<br />

SNPs, rs4655567, rs8020368, rs2018683 and rs17574269 were significantly<br />

associated with a refractory relapse. In multivariate logistic analysis<br />

including age, gender and PS, rs8020368 CC genotype showed higher risk<br />

<strong>of</strong> refractory relapse than patients with TT or TC genotype (HR�14.9 [95%<br />

CI, 1.9-114.8], p�0.010). Conclusions: This exploratory GWAS identified<br />

several candidate SNPs that might predictive for the outcome <strong>of</strong> patients<br />

with ED-SCLC receiving IP chemotherapy.<br />

7091 General Poster Session (Board #41B), Sat, 1:15 PM-5:15 PM<br />

Randomized phase II study <strong>of</strong> recombinant human endostatin in combination<br />

with chemotherapy in previously untreated extensive-stage small-cell<br />

lung cancer (NCT00912392). Presenting Author: Shun Lu, Shanghai Lung<br />

Cancer Center, Shanghai Chest Hospital, Shanghai, China<br />

Background: Endostar (recombinant human endostatin) is a novel antiangiogenesis<br />

drug developed in China for non-small cell lung cancer (NSCLC).<br />

Because <strong>of</strong> promising efficacy signals, we performed a randomized phase II<br />

trial to assess the efficacy and safety <strong>of</strong> adding endostar to first-line<br />

standard chemotherapy for treatment <strong>of</strong> chemonaive extensive-stage smallcell<br />

lung cancer (SCLC). Methods: Extensive-stage SCLC patients with a<br />

performance status 0-2 were randomly assigned to endostar group (endostar<br />

7.5mg/m2 D1-D14 with carboplatin AUC�5 plus etoposide 60mg/m2 D1-D5 <strong>of</strong> a 21-day cycle for six cycles) or the control group (the same dose<br />

<strong>of</strong> carboplatin plus etoposide). Patients in endostar treatment group with<br />

CR, PR and SD were treated with single-agent endostar until progression or<br />

unacceptable toxicity. The primary end point is progression-free survival<br />

(PFS). The secondary end points are overall survival (OS) and response rate<br />

(RR). Results: 140 patients were enrolled from June 2009 to June 2011,<br />

and 137 patients were included in full analysis set. 68 patients were<br />

randomly assigned to the endostar treatment and 69 patients to the control<br />

group. Median age was 57 years and 80.9% for male in the endostar group<br />

while median age was 58 years and 85.5% for male in the control group.<br />

Median PFS was similar for endostar and control group (6.2 v 5.9 months,<br />

P�0.163, HR 0.762; 95%CI 0.519-1.119). Median overall survival (OS)<br />

was also similar for both groups (12.4 v 12.3 months, P�0.475, HR<br />

0.835; 95%CI 0.508-1.371). Overall RR were 76.5% for endostar group<br />

and 68.1% for the control group (p�0.275). 20 patients in the endostar<br />

group completed six cycles <strong>of</strong> therapy and subsequently treated with<br />

single-agent endostar as maintenance therapy and the median PFS and OS<br />

were 6.8 and 12.4 months respectively. The rate <strong>of</strong> � 3 grade adverse<br />

events occurred in both groups was similar and no new or unexpected safety<br />

signals for endostar were observed. Conclusions: The addition <strong>of</strong> endostar to<br />

carboplatin plus etoposide for treatment <strong>of</strong> extensive stage SCLC didn’t<br />

improve the PFS significantly, with an acceptable toxicity pr<strong>of</strong>ile. And no<br />

improvement in OS was observed.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


474s Lung Cancer—Non-small Cell Local-Regional/Small Cell/Other Thoracic Cancers<br />

7092 General Poster Session (Board #41C), Sat, 1:15 PM-5:15 PM<br />

Circulating tumor cells (CTCs) in patients (pts) with small cell lung cancer<br />

(SCLC) as a marker <strong>of</strong> disease burden and therapeutic response, and<br />

predictor <strong>of</strong> disease relapse. Presenting Author: Anjana Ranganathan,<br />

University <strong>of</strong> Pennsylvania, Philadelphia, PA<br />

Background: Currently there are no validated biomarkers for assessment or<br />

prediction <strong>of</strong> disease burden or activity in SCLC. Enumeration <strong>of</strong> CTCs by<br />

CellSearch is FDA approved, highly reproducible and validated in other<br />

malignancies. Application as a prognostic and predictive marker in SCLC is<br />

limited due to a lack <strong>of</strong> studies documenting serial monitoring in pts on<br />

therapy. Methods: We are conducting a prospective study serially enumerating<br />

CTCs in pts with newly diagnosed SCLC. CTC number (per 7.5 ml<br />

peripheral blood) and percentage <strong>of</strong> CTCs demonstrating DNA damage and<br />

apoptosis based on �H2AX and M30 staining respectively, are being<br />

assessed prior to initiation <strong>of</strong> chemotherapy, during each cycle and at<br />

relapse. We are correlating CTC number with disease stage, number <strong>of</strong><br />

metastatic sites, response to therapy, and time to progression (TTP).<br />

Results: 21 SCLC pts are evaluable. 9 pts with limited disease (LD) had<br />

median baseline CTC value <strong>of</strong> 1 (0-8); only 2 <strong>of</strong> 9 pts had �5 CTCs. 12 pts<br />

with extensive disease (ED) had median baseline CTC value <strong>of</strong> 80.5<br />

(0-37780); 8 <strong>of</strong> 12 pts had �5 detectable CTCs (p value 0.02). Amongst<br />

the 12 pts with ED, median baseline CTC count was higher in pts with �3<br />

(n�3) compared to 1-2 sites <strong>of</strong> metastatic disease (n�9) (2668 vs 71; p<br />

value 0.52). Median percentage <strong>of</strong> CTCs positive for �H2AX and M30 was<br />

also higher for pts with �3 compared to 1-2 metastatic sites (83, 164.5 vs.<br />

2, 7.5) (p-value 0.40, 0.69). Serial CTC data are available on 3 pts with<br />

ED; all had responsive disease and reduction in CTC number to 0-1 after 2<br />

cycles <strong>of</strong> chemotherapy. As this protocol is ongoing, correlation <strong>of</strong> CTCs<br />

with updated response status and TTP will be presented at the ASCO<br />

meeting. Conclusions: CTCs can be isolated and serially enumerated in pts<br />

with SCLC. Baseline CTCs correlate directly with disease stage. In pts with<br />

� 3 metastatic sites, baseline CTCs tend to be greater and show higher<br />

levels <strong>of</strong> DNA damage and apoptosis compared to those with 1-2 metastatic<br />

sites. Reduction in CTCs is associated with radiographic response to<br />

therapy. Correlation between absolute number at baseline and TTP is not<br />

yet known.<br />

7094 General Poster Session (Board #41E), Sat, 1:15 PM-5:15 PM<br />

A phase II study <strong>of</strong> second-line bendamustine in relapsed or refractory<br />

small cell lung cancer (SCLC). Presenting Author: Christine Marie Lovly,<br />

Vanderbilt Ingram Cancer Center, Nashville, TN<br />

Background: Bendamustine is an alkylating agent with a nitrogen mustard<br />

group and purine-like benzimidazole group. Bendamustine in combination<br />

with carboplatin has shown efficacy as first line therapy in extensive stage<br />

SCLC with RR 73%, TTP 5.2 months and OS 8.3 months [Köster, et al, JCO<br />

2007]. This study aims to investigate the efficacy and safety <strong>of</strong> single agent<br />

bendamustine as 2nd or 3rd line therapy in patients with extensive-stage<br />

disease, small-cell lung cancer (ED-SCLC). Methods: This is an open-label,<br />

single-arm, multicenter phase II trial. Eligible patients had previously<br />

treated ED-SCLC, up to 2 prior regimens, ECOG performance status 0-2,<br />

evaluable/measurable disease, and adequate marrow, renal and hepatic<br />

function. Patients with stable treated brain metastases were allowed.<br />

Patients were treated with bendamustine (120mg/m2 IV days 1 and 2 every<br />

3 weeks) for up to 6 cycles. Evaluation occurred every 2 cycles. Primary<br />

endpoint was TTP; secondary endpoints include RR, PFS, OS, and toxicity.<br />

Results: 48 patients were enrolled; 56% were male and 96% were<br />

Caucasian. 33 patients were evaluable for response. There was 1 CR, 9 PR,<br />

13 SD (48% disease control rate) and 10 PD. Median TTP was 3.37<br />

months (95% CI 2.30 to 4.47 months). At the time <strong>of</strong> analysis, 13 patients<br />

were alive and with a median overall survival <strong>of</strong> 4.77 months (95% CI 3.67<br />

to 6.07 months). 5 patients (10.4%) required dose reductions due to AEs,<br />

2 due to fatigue, 1 due to neutropenia, 1 due to pancytopenia and 1 due to<br />

pneumonia. Grade 3/4 AEs included fatigue (18.8%), dyspnea (14.5%),<br />

infection without neutropenia (12.5%), anemia (8.3%), neutropenia (8.3%),<br />

and diarrhea (8.3%). Conclusions: These data indicate that single agent<br />

bendamustine appears to be well tolerated and effective in the second or<br />

third line setting for patients with SCLC.<br />

7093 General Poster Session (Board #41D), Sat, 1:15 PM-5:15 PM<br />

NCIC IND.190: A phase I trial <strong>of</strong> MK-0646 in combination with cisplatin<br />

and etoposide in extensive-stage small cell lung cancer (ES SCLC).<br />

Presenting Author: Peter Michael Ellis, Juravinski Cancer Centre, Hamilton,<br />

ON, Canada<br />

Background: Increased serum levels <strong>of</strong> insulin like growth factor (IGF), plus<br />

overexpression <strong>of</strong> the IGF-receptor (IGFR) are implicated in SCLC cell<br />

growth and proliferation, making suppression <strong>of</strong> the IGFR a potential<br />

therapeutic target. MK-0646 is a monoclonal antibody directed against the<br />

IGFR. The aim <strong>of</strong> this study was to determine the recommended phase II<br />

dose (RP2D) <strong>of</strong> cisplatin, etoposide plus MK-0646. Methods: We conducted<br />

a phase I study <strong>of</strong> two dose levels <strong>of</strong> MK-0646 (DL1 5mg/kg, DL2<br />

10mg/kg IV weekly) in combination with cisplatin (25mg/m2 ) and etoposide<br />

(100mg/m2 ) IV D1-3, q21d, for patients with chemotherapy-naive ES<br />

SCLC, PS 0-2. Patients with treated stable brain metastases were eligible.<br />

Patients completing 4-6 cycles <strong>of</strong> combination therapy could continue<br />

single agent MK-0646 until disease progression. Primary outcome was<br />

determination <strong>of</strong> the RP2D. Secondary outcomes included ORR (RECIST<br />

1.1), and toxicity (CTCAEv3). Results: A total <strong>of</strong> 12 patients were treated<br />

(DL1 – 3, DL2 – 9). The median age was 63 years (48-70), with 6 males<br />

and 6 females. Most subjects were good ECOG PS (PS1–8,PS2–4)and<br />

had 4 or more sites <strong>of</strong> disease (n�8). No DLTs were observed in DL1 or<br />

DL2. In an expanded DL2 cohort, 1 patient died from neutropenic sepsis<br />

during cycle 1. The median number <strong>of</strong> treatment cycles <strong>of</strong> chemotherapy<br />

was 4 (DL1) or 5 (DL2) and MK-0646 was 6 (DL1&2). Dose delays were<br />

observed for chemotherapy (DL1 - 2, DL2 – 6) and MK-0646 (DL1 – 3, DL2<br />

– 7). The confirmed ORR was 72.7% (PR 8, SD 2, PD 1, non-evaluable 1).<br />

Grade �3 toxicities (any cycle) occurring in more than 1 patient included:<br />

neutropenia (92%); thrombocytopenia (25%); leukopenia (50%); anemia<br />

(17%); fatigue (33%); joint pain (17%); thrombosis (25%). Grade 2 or 3<br />

hyperglycemia was observed in 1 <strong>of</strong> 3 (DL1) and 5 <strong>of</strong> 9 (DL2). Eight SAEs<br />

were observed in 3 patients (thrombosis, febrile neutropenia, infection,<br />

syncope, fatigue 2, dyspnea, back pain). Conclusions: MK-0646 can be<br />

combined at full dose with standard doses <strong>of</strong> cisplatin and etoposide<br />

(25mg/m2 and 100mg/m2 D1-3) with a RP2D <strong>of</strong> MK-0646 10mg/kg/week.<br />

The observed toxicities are consistent with that expected from cisplatin and<br />

etoposide except for hyperglycemia, which appears dose dependent.<br />

7095 General Poster Session (Board #41F), Sat, 1:15 PM-5:15 PM<br />

ABEL trial: A phase II randomized trial adding bemiparin (B) to chemoradiotherapy<br />

(CT-RT) in limited-stage small cell lung cancer (SCLC)—Final<br />

results. Presenting Author: Bartomeu Massuti, Alicante University Hospital,<br />

Alicante, Spain<br />

Background: Low molecular weight heparins (LMWH) could improve therapeutic<br />

outcomes in patients with advanced cancer. Heparin binding growth<br />

associated molecule is a member <strong>of</strong> growth factors involved in proliferation,<br />

angiogenesis and metastases in SCLC. LMWH associated with CT increase<br />

survival in cancer patients (Cochrane DB Systematic Reviews 2011).<br />

LMWH do not increase hemorrhagic events when used in neoplastic<br />

patients. Methods: Open randomized phase II multicentric trial to assess<br />

efficacy and safety <strong>of</strong> adding bemiparin to standard CT-RT in patients with<br />

SCLC with limited disease. CT: Cis/carboplatin � etoposide x 4-6 cycles.<br />

Concurrent early thoracic RT (45-50 Gy) and prophylactic cranial irradiation<br />

in case <strong>of</strong> response. Patients (p) were randomly allocated to addition <strong>of</strong><br />

B at daily subcutaneous dose <strong>of</strong> 3.500 UI during 26 weeks. Primary<br />

end-point: progression free survival (PFS). Secondary end-points: response<br />

(RECIST), safety pr<strong>of</strong>ile (CTC), venours thromboembolic (VTE) and hemorrhagic<br />

events incidence and overall survival (OS). Sample size 130 p.<br />

Preplanned interim analysis after inclusion 30 p. Results: From October-05<br />

to January/10 39 p were included. Study was closed after interim analysis<br />

due to poor recruitment. 38 evaluable p. ITT analysis: 20 CR-RT � Bvs18<br />

CT-RT. No significant disbalance in patient characteristics and prognostic<br />

factors. Median PFS 58.6 weeks (CT-RT�B) vs 38.8 w (CT-RT) (HR 2.576;<br />

Log-rank p�0,018). RR: 65% (CT-RT�B) vs 55% (CT-RT) (NS); Median<br />

OS 161.8 w (CT-RT�B) vs 49.3 w (CT-RT) (HR 2.96; Log-rank p�0,012).<br />

2-year survival rate: 68.6% (CT-RT�B) vs 29.4% (CT-RT) (p�0,0042).<br />

Safety: G3-4 AE: 50% vs 67%; bleeding events: 10% vs 27% (NS);<br />

thrombocytopenia 15% vs 50% (p�0.024); VTE: 0 vs 22% (p�0.041).<br />

Conclusions: Addition <strong>of</strong> B (3.500 u/daily s.c. during 26 w) to CT-RT in<br />

SCLC with LD significantly increases PFS and OS. B does not increase<br />

hemorragic events and significantly reduces VTE. In spite <strong>of</strong> early closure <strong>of</strong><br />

the trial due to poor accrual, these results warrants further research in this<br />

approach trying to improve current outcomes <strong>of</strong> SCLC. <strong>Clinical</strong>Trials.gov Id:<br />

NCT00324558.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


Lung Cancer—Non-small Cell Local-Regional/Small Cell/Other Thoracic Cancers<br />

7096 General Poster Session (Board #41G), Sat, 1:15 PM-5:15 PM<br />

Investigation <strong>of</strong> PARP1 as a therapeutic target in small cell lung cancer.<br />

Presenting Author: Lauren Averett Byers, University <strong>of</strong> Texas M. D.<br />

Anderson Cancer Center, Houston, TX<br />

Background: Small cell lung cancer (SCLC) is an aggressive malignancy that<br />

differs from non-small cell lung cancer (NSCLC) in its metastatic potential<br />

and response to treatment. To date, no molecularly-targeted agent has<br />

prolonged survival <strong>of</strong> SCLC patients. Using a proteomic approach, we<br />

previously identified high expression <strong>of</strong> the DNA repair protein poly<br />

(ADP-ribose) polymerase 1 (PARP1) in SCLC cell lines and tumors. Here we<br />

test in vitro sensitivity <strong>of</strong> SCLC to PARP inhibition or knockdown. Methods:<br />

Cell lines were treated with PARP inhibitor olaparib or AG014699 for 14d<br />

�/- chemotherapy. Relative cell viability was assessed by cell count. siRNA<br />

against PARP1 was compared with scrambled siRNA and mock transfected<br />

cells. To assess DNA repair, RAD51 foci were counted after 1�M or5�M<br />

olaparib and after 8Gy irradiation (RT). Results: SCLC cell lines were highly<br />

sensitive to PARP inhibition by olaparib (IC50s �0.5 �M for H69; �2�M<br />

in H524, H82, and H526) and AG014699 (IC50s �0.5 �M for H82, H69,<br />

and H524; 2.2 �M for H526 and H841). In contrast, A549 (NSCLC) was<br />

resistant to both drugs (IC50s �8�M). Because BRCA1/2 and PTEN<br />

mutations are associated with greater sensitivity to PARP inhibitors, we<br />

compared SCLC sensitivity with that <strong>of</strong> BRCA1-mutated (HCC1395) and<br />

PTEN-mutated (MDA-MB-468) breast lines. As expected, HCC1395 and<br />

MDA-MB-468 were sensitive to both PARP inhibitors. Remarkably, however,<br />

SCLC cell lines were as sensitive or more so. Combination <strong>of</strong> olaparib<br />

with topotecan or irinotecan (commonly used in SCLC) decreased tumor<br />

cell viability more than either agent alone (p �0.03). Consistent with the<br />

drug studies, knockdown <strong>of</strong> PARP1 by siRNA decreased growth <strong>of</strong> SCLC<br />

compared with that <strong>of</strong> controls. RAD51 foci increased in SCLC after<br />

olaparib treatment (�4-fold) and RT (�18-fold). Conclusions: SCLC lines<br />

were as sensitive to PARP inhibition as BRCA1- or PTEN-mutated breast<br />

cancer lines. Moreover, PARP inhibition enhanced the effect <strong>of</strong> chemotherapy<br />

on SCLC lines. Increased formation <strong>of</strong> RAD51 foci in SCLC cells<br />

after olaparib or RT suggests a deficiency in homologous recombination<br />

that may account for the sensitivity to PARP inhibitors. These results<br />

support the investigation <strong>of</strong> PARP inhibition as a novel therapeutic<br />

approach in SCLC lung cancer.<br />

7098 General Poster Session (Board #42A), Sat, 1:15 PM-5:15 PM<br />

Impact <strong>of</strong> PET staging in limited-stage SCLC. Presenting Author: Eric<br />

Xanthopoulos, Department <strong>of</strong> Radiation Oncology, University <strong>of</strong> Pennsylvania<br />

School <strong>of</strong> Medicine, Philadelphia, PA<br />

Background: Although PET/CT has been established for the initial staging <strong>of</strong><br />

NSCLC, it is still unproven for SCLC. This study examines clinical outcome<br />

in patients with limited-stage SCLC staged with and without PET/CT.<br />

Methods: An institutional database was reviewed to identify all patients that<br />

presented with limited-stage SCLC and treated definitively with concurrent<br />

chemoradiation. Overall survival and associations were assessed by the<br />

Kaplan-Meier approach, log-rank tests and Cox modeling. Results: From<br />

01/04 – 08/10, 54 consecutive limited-stage SCLC patients were treated<br />

with concurrent chemoradiation at the University <strong>of</strong> Pennsylvania. 40<br />

patients underwent PET staging; 14 underwent CT staging only; all had MR<br />

<strong>of</strong> the brain. Patient characteristics were well distributed, including age (p<br />

� 0.35), race (p � 0.21), sex (p � 0.93), dose (45 Gy, p � 0.89), and<br />

fractions per day (p � 0.89). PET-staged patients had median survival <strong>of</strong><br />

32 vs 15 months in patients without PET (p � 0.03). Survival rate was<br />

57% vs 29% at 24 months. Median time to distant failure <strong>of</strong> 29 vs 11<br />

months (p � 0.05). Median time to local failure was 41 vs 12 months (p �<br />

0.03). Median followup was 38 months in the 19 surviving PET-staged<br />

patients and 40 in the 2 surviving non-PET patients (p � 0.59). Lack <strong>of</strong><br />

PET-staging (OR � 0.92, p � 0.04) and race (OR � 1.02, p � 0.03)<br />

associated with increased distant failure on multivariate Cox analysis. Only<br />

PET-staging (OR � 0.93, p � 0.04) associated with increased overall<br />

survival on univariable Cox modeling. Conclusions: Median and overall<br />

survival <strong>of</strong> PET-staged SCLC patients compared favorably with those who<br />

were not. These findings are presumably due primarily to identification <strong>of</strong><br />

CT-occult distant disease by PET. This study underscores the value <strong>of</strong> PET<br />

in staging patients with SCLC.<br />

475s<br />

7097 General Poster Session (Board #41H), Sat, 1:15 PM-5:15 PM<br />

Expression pattern and biologic relevance <strong>of</strong> Bcl-2 and Mcl-1 in pulmonary<br />

neuroendocrine tumors. Presenting Author: Rajasree Pia Chowdry, Emory<br />

University, Atlanta, GA<br />

Background: Bcl-2 protein overexpression is frequently described in small<br />

cell lung cancer (SCLC) but the biological relevance remains unclear. To<br />

better elucidate the role <strong>of</strong> Bcl2 dysfunction in SCLC, we assessed the total<br />

and the biologically relevant phosphorylated forms <strong>of</strong> the anti-apoptotic<br />

Bcl2 family members (Bcl2, pBcl-2, Mcl-1, pMcl-1) in pulmonary neuroendocrine<br />

(NE) tumors. Methods: We analyzed archival samples <strong>of</strong> pulmonary<br />

carcinoid, SCLC and large-cell NE carcinoma (LCNEC) by immunohistochemistry<br />

(IHC) using specific antibodies. Protein expression was assessed<br />

by light microscopy using standard scoring <strong>of</strong> intensity (0, 1, 2 and 3) and<br />

percent cell staining and correlated with age, gender, smoking status,<br />

tumor type, stage and survival. Significant differences in expression pattern<br />

and correlation with clinical variables were assessed by Wilcoxon twosample<br />

test, t-test, ANOVA, Spearman and Pearson correlation coefficients<br />

and Cox regression model. Results: We retrieved 77 cases: carcinoid (16)<br />

SCLC (41) and LCNEC (20); median age (61 yrs), gender (62% females).<br />

Bcl2 and pBcl2 expression was higher in SCLC and LCNEC compared to<br />

benign carcinoid tumor. pBcl-2 showed stronger correlation with adverse<br />

features including smoking, malignant histology, higher stage at diagnosis<br />

and worse survival. No significant association <strong>of</strong> Mcl1 or pMcl-1 expression<br />

with clinical or pathologic parameters. Conclusions: pBcl-2 expression<br />

pattern and correlation with adverse prognostic features indicate a key role<br />

in SCLC biology and relevance as a therapeutic target.<br />

Biomarker Carcinoid SCLC LCNEC P value<br />

Cytoplasmic Bcl-2 7 97 124 0.0005<br />

Cytoplasmic pBcl-2 42 132 83 0.0007<br />

Cytoplasmic Mcl-1 230 232 253 0.5327<br />

Cytoplasmic pMcl-1<br />

Correlation <strong>of</strong> biomarkers and OS<br />

131 173 165 02919<br />

HR (95% CI)<br />

Cytoplasmic Bcl-2 1.01 (1.00-1.02) 0.0167<br />

Cytoplasmic pBcl-2 1.57 (1.09-2.26) 0.0165<br />

Cytoplasmic Mcl-1 1.12 (0.73-1.70) 0.6131<br />

Cytoplasmic pMcl-1<br />

Correlation <strong>of</strong> biomarkers and PFS<br />

0.96 (0.67-1.40) 0.8462<br />

HR (95% CI)<br />

Cytoplasmic Bcl-2 1.01 (1.00-1.02) 0.0072<br />

Cytoplasmic pBcl-2 1.39 (1.02-1.89) 0.0383<br />

Cytoplasmic Mcl-1 1.19 (0.82-1.73) 0.3531<br />

Cytoplasmic p-Mcl1 1.00 (1.00-1.01) 0.0402<br />

7099 General Poster Session (Board #42B), Sat, 1:15 PM-5:15 PM<br />

A phase II trial <strong>of</strong> pazopanib in relapsed/refractory small cell lung cancer<br />

(SCLC). Presenting Author: Leena Gandhi, Dana-Farber Cancer Institute,<br />

Boston, MA<br />

Background: Despite response to initial therapy, SCLC has high rates <strong>of</strong><br />

relapse or distant metastasis and limited 2nd-line therapeutic options.<br />

Angiogenesis is an essential part <strong>of</strong> cell invasion, dissemination, and<br />

outgrowth <strong>of</strong> distant metastases and therefore is a potential therapeutic<br />

target in SCLC. Pazopanib (Votrient, GSK) is a potent, competitive inhibitor<br />

<strong>of</strong> the tyrosine kinase activity <strong>of</strong> VEGFR-1, VEGFR-2, VEGFR-3, PDGF, and<br />

c-kit. We initiated a phase II single-arm trial <strong>of</strong> pazopanib in relapsed or<br />

refractory SCLC to determine impact on disease progression. Methods:<br />

Patients were eligible if they had progressive disease following up to two<br />

lines <strong>of</strong> prior therapy. Patients were treated at the FDA-approved dose <strong>of</strong><br />

800 mg pazopanib once daily. The primary endpoint was progression-free<br />

rate (PFR) at 8 weeks. Secondary endpoints included median progressionfree<br />

survival, overall survival, and safety. The trial followed a Simon 2-stage<br />

design to limit accrual if no therapeutic benefit was observed. Results: To<br />

date, 27 <strong>of</strong> 30 planned subjects have been enrolled since October 2010.<br />

Two did not complete cycle 1 and were considered inevaluable for<br />

response. Major toxicities (mostly grade 1/2) were those previously described<br />

including nausea, fatigue, hypertension, electrolyte abnormalities,<br />

and AST/ALT elevations (grade 3 in 4 subjects). Three subjects were<br />

removed from study due to toxicity: 1 with grade 3 nausea, 1 with grade 3<br />

drop in the cardiac ejection fraction, and 1 with multiple grade 2 toxicities<br />

including diarrhea, fatigue, and nausea. A fourth was removed due to grade<br />

1 hemoptysis despite clinical response. The PFR at 8 weeks <strong>of</strong> 21 subjects<br />

evaluable for response to date was 52%; 4 <strong>of</strong> these 11 subjects had<br />

chemo-refractory disease. There were no confirmed responses, but tumor<br />

regressions ranged from 2-20%. Median progression-free survival is 14.1<br />

weeks. Conclusions: In patients with SCLC, single-agent pazopanib demonstrated<br />

a notable rate <strong>of</strong> stable disease (including among chemo-refractory<br />

patients) and a median PFS that exceeds that <strong>of</strong> historical PFS rates <strong>of</strong> � 2<br />

months on ineffective 2nd-line therapies. These data suggest that the<br />

potential for pazopanib in SCLC treatment should be further investigated.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


476s Lung Cancer—Non-small Cell Local-Regional/Small Cell/Other Thoracic Cancers<br />

7100 General Poster Session (Board #42C), Sat, 1:15 PM-5:15 PM<br />

Amrubicin and carboplatin with pegfilgrastim in patients with extensivestage<br />

small-cell lung cancer (ES-SCLC): A phase II study <strong>of</strong> the Sarah<br />

Cannon Research Institute (SCRI). Presenting Author: Dianna Shipley,<br />

Tennessee Oncology, PLLC/Sarah Cannon Research Institute, Nashville,<br />

TN<br />

Background: Amrubicin is a novel anthracycline associated with high<br />

objective response rates (ORR) in patients (pts) with relapsed SCLC.<br />

Amrubicin improved the ORR and progression-free survival (PFS) in<br />

relapsed SCLC vs. topotecan, but not overall survival (OS) in a phase III<br />

study. Amrubicin with cisplatin/carboplatin for elderly Japanese pts was<br />

safe and active. We conducted a multicenter phase II study evaluating<br />

amrubicin and carboplatin in newly diagnosed ES-SCLC. Methods: Eligible<br />

pts had untreated ES-SCLC, measurable/evaluable disease (RECIST v. 1.1)<br />

and an ECOG PS �2. Pts received 4 cycles <strong>of</strong> amrubicin 30 mg/m2 on days<br />

1-3 and carboplatin AUC�5 both IV day 1 every 21 days with restaging<br />

every 6 weeks. Pegfilgrastim 6 mg sq was administered on day 4 <strong>of</strong> each<br />

cycle. The primary endpoint was 1-year OS. Secondary endpoints included<br />

ORR, PFS, OS, and toxicity. Results: 78 pts were enrolled from 3/2010 to<br />

7/2011. Baseline characteristics included: median age 65 yrs (range<br />

45-84); 56% female. 64% completed 4 cycles <strong>of</strong> treatment. Eleven (14%)<br />

pts showed complete responses and 47 (60%) pts partial responses, for an<br />

ORR <strong>of</strong> 74% (95% confidence interval 65%-82%). Twelve (15%) pts had<br />

stable disease. Median PFS and OS were 5.3 and 9.5 months, respectively.<br />

The 1-year OS was 36%. Grade 3/4 myelosuppression was the most<br />

common toxicity (thrombocytopenia 44%, neutropenia 34%, febrile neutropenia<br />

12%, anemia 26%), but was manageable. Severe non-hematologic<br />

toxicities (�5%) included hypokalemia 17%, fatigue 13%, dehydration<br />

10%, hyponatremia 10%, pneumonia 9%, and nausea/vomiting 8%. 1 pt<br />

died from sepsis and another from aspiration pneumonia. Conclusions:<br />

First-line ES-SCLC treatment with amrubicin and carboplatin induced<br />

several complete responses and is considered highly active. Myelosuppression<br />

was managed effectively with growth factor support. These results are<br />

comparable to historical data with platinum-doublet chemotherapy. A<br />

larger randomized study would be required to best assess this regimen’s<br />

impact on survival.<br />

7102 General Poster Session (Board #42E), Sat, 1:15 PM-5:15 PM<br />

Incidence <strong>of</strong> venous thromboembolism in lung cancer and its effect on<br />

survival. Presenting Author: Mohammad Mozayen, McLaren Regional<br />

Medical Center, Michigan State University, Flint, MI<br />

Background: Lung cancer is a major risk factor <strong>of</strong> venous thromboembolism<br />

(VTE). The incidence <strong>of</strong> VTE in different histological patterns <strong>of</strong> lungs<br />

cancer and its impact at the disease outcome is not well studied. We aim to<br />

evaluate the incidence rate <strong>of</strong> VTE in lung cancer and its effect on survival.<br />

Methods: Patients (pts) with lung cancer diagnosis from cancer database <strong>of</strong><br />

community hospital were reviewed. Pts’ medical records were checked for<br />

VTE over 3 years after diagnosis. Pts’ demographics, pathology, TNM stage<br />

and overall survival were studied. Development <strong>of</strong> VTE was primary<br />

outcome and 3 year overall survival was the secondary outcome. Results: A<br />

total 2,164 pts with lung cancer between 1995 and 2008 were included.<br />

Median age <strong>of</strong> the study population was 70 years. Males were 53%, African<br />

<strong>American</strong>s were 7%. 200 pts (9%) were diagnosed with VTE. Out <strong>of</strong> 1783<br />

pts with non-small cell lung cancer (NSCLC), 176 pts (9.9%) had VTE<br />

whereas out <strong>of</strong> 381 pts with small cell lung cancer (SCLC) 24 pts (6.3%)<br />

had VTE (p�0.015). Among NSCLC pts, 13.5% <strong>of</strong> pts with adenocarcinoma<br />

had VTE, whereas 6.6% <strong>of</strong> pts with squamous cell carcinoma had<br />

VTE (p�0.05). The incidence <strong>of</strong> VTE in stages I, II, III, IV <strong>of</strong> all pts were<br />

(8.4%, 7.3%, 13%, 7.5%) (p�0.007) respectively. In NSCLC, three years<br />

overall survival <strong>of</strong> pts with and without VTE was 22% and 24% respectively<br />

(p�0.34). In SCLC, three years overall survival <strong>of</strong> pts with and without VTE<br />

was 21% and 11% respectively (p�0.09). Conclusions: There is a higher<br />

incidence <strong>of</strong> VTE in non-small cell lung cancer than in small cell lung<br />

cancer. Among the NSCLC, VTE’s incidence was higher in adenocarcinoma<br />

than squamous carcinoma. VTE maybe higher at advanced stages (stage<br />

III) in both NSCLC and SCLC combined. There was no significant difference<br />

in the 3 years overall survival <strong>of</strong> lung cancer patients with and without VTE.<br />

7101 General Poster Session (Board #42D), Sat, 1:15 PM-5:15 PM<br />

Comparison <strong>of</strong> treatment patterns, health care utilization, and direct costs<br />

in elderly patients with distant-stage small cell lung cancer (SCLC) versus<br />

non-small cell lung cancer (NSCLC) using the SEER-Medicare database.<br />

Presenting Author: Sudeep Karve, RTI Health Solutions, Research Triangle<br />

Park, NC<br />

Background: Limited data exist on real-world treatment patterns, healthcare<br />

utilization, and associated costs <strong>of</strong> advanced SCLC among elderly patients<br />

in the US, and there are no recent comparisons between patients with<br />

advanced SCLC and advanced NSCLC. Methods: We retrospectively analyzed<br />

administrative claims data for elderly patients (�65 years) from the<br />

linked Surveillance, Epidemiology and End Results (SEER)-Medicare<br />

database for 2000-2008. Patients with a new diagnosis <strong>of</strong> distant stage<br />

lung cancer receiving cancer-directed therapy (ie, surgery, radiation,<br />

biologics, and/or chemotherapy) were grouped by tumor type (SCLC<br />

[n�5,855] vs NSCLC [n�24,090]). Survival was compared using Kaplan-<br />

Meier Log-rank; categorical measures with Chi-square statistics; and<br />

continuous measures with t-tests. Results: Compared to SCLC patients, a<br />

significantly greater proportion <strong>of</strong> patients with NSCLC received radiation<br />

therapy (75.6% vs 65.4%; p�0.001) and surgery (13.6% vs 7.8%;<br />

p�0.001). Chemotherapy was received by 85.5% <strong>of</strong> SCLC patients and<br />

60.3% <strong>of</strong> NSCLC patients (p�0.001). Significantly higher proportions <strong>of</strong><br />

SCLC patients also received red blood cell (20.7% vs 10.9; p�0.001) and<br />

platelet transfusions (5.6% vs 1.8%; p�0.001) as well as growth factor<br />

support (58.9% vs 39.5%; p�0.001). Survival did not differ significantly<br />

between groups (p�0.424), with the mean (10.4 months vs 11.1 months)<br />

and median (7.4 months vs 5.9 months) survival for SCLC and NSCLC<br />

noted accordingly. Total lifetime lung cancer-related costs ($44,167 vs<br />

$37,932; p�0.001) and all-cause costs ($70,548 vs $67,175; p�0.001)<br />

per patient for SCLC exceeded those for NSCLC. The primary drivers <strong>of</strong> cost<br />

included resource utilization across 3 care settings: hospitalizations, <strong>of</strong>fice<br />

visits, and hospital outpatient visits. Conclusions: Overall total lifetime and<br />

disease-related costs per advanced SCLC and NSCLC patient were high,<br />

and costs for SCLC exceeded those for NSCLC. Survival estimates coupled<br />

with per patient costs for both cancers underscores the unmet medical<br />

need for patients with distant stage SCLC and NSCLC.<br />

7103 General Poster Session (Board #42F), Sat, 1:15 PM-5:15 PM<br />

A phase (Ph) I/II study <strong>of</strong> belinostat (Bel) in combination with cisplatin,<br />

doxorubicin, and cyclophosphamide (PAC) in the first-line treatment <strong>of</strong><br />

advanced or recurrent thymic malignancies. Presenting Author: Anish<br />

Thomas, National Cancer Institute, Bethesda, MD<br />

Background: Belinostat is a hydroxamic acid histone deacetylase (HDAC)<br />

pan-inhibitor with single agent activity in thymic malignancies. PAC has<br />

activity against thymic cancers. Synergy between Bel and several chemotherapeutic<br />

agents, including P, A, and C, has been demonstrated in<br />

preclinical models. Methods: Patients with histologically confirmed, treatment<br />

naive advanced thymic malignancies, PS�2, measurable disease,<br />

and adequate renal, hepatic and hematopoietic functions were eligible.<br />

Ph1 evaluated safety and tolerability <strong>of</strong> the combination using increasing<br />

dose levels (DL) <strong>of</strong> Bel (1000-2000 mg/m² over 48 h CIVI) and PAC<br />

(50/50/500 mg/m² IV/cycle) (3�3 dose escalation schema), administered<br />

every 21 days for no more than 6 cycles followed by optional maintenance<br />

Bel every 4 weeks. Primary end point <strong>of</strong> Ph2 is overall response rate (ORR).<br />

Results: From March 2010 to January 2012, 13 patients were enrolled [7<br />

thymoma (T), 6 thymic carcinoma (TC); 8 in Ph1 and 5 in Ph2; median age:<br />

49 years (range, 23-76)]. In Ph1, 6 patients were treated at DL1 (Bel 1000<br />

mg/m2� PAC) and 2 patients at DL2 (Bel 2000 mg/m2� PAC). Dose<br />

Limiting Toxicities were Grade 3 nausea and diarrhea, and Grade 4<br />

neutropenia and thrombocytopenia. Recommended phase II dose (RP2D)<br />

was set at DL1. Most common grade 3/4 treatment-related adverse events<br />

(AE) were lymphocytopenia (100%), leucopenia (85%), neutropenia (77%)<br />

thrombocytopenia (54%), anemia (38%), hypophosphatemia (38%), hypomagnesemia,<br />

hypokalemia, elevated AST, prolonged QTc and infusioncatheter<br />

related thromboembolic complications (23% each). Outcomes<br />

included one complete response (CR; T at DL1), 6 partial responses (PR; 4<br />

T, 2 TC; 4 in Ph1, 2 in Ph2) and 6 stable disease (SD; 2 T, 4 TC; 3 each in<br />

Ph1 and Ph2). Four patients previously deemed unresectable underwent<br />

surgical resection. Conclusions: Belinostat in combination with PAC has<br />

activity in thymic malignancies with a predicable AE pr<strong>of</strong>ile. ORR was 54%<br />

including 33% PR in the TC subgroup. RP2D <strong>of</strong> the combination has been<br />

defined. Accrual to Ph2 part and molecular pr<strong>of</strong>iling <strong>of</strong> patient tumors is<br />

ongoing.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


Lung Cancer—Non-small Cell Local-Regional/Small Cell/Other Thoracic Cancers<br />

7104 General Poster Session (Board #42G), Sat, 1:15 PM-5:15 PM<br />

A prospective, phase II trial <strong>of</strong> induction chemotherapy with docetaxel/<br />

cisplatin for Masaoka stage III/IV thymic epithelial tumors. Presenting<br />

Author: Silvia Park, Division <strong>of</strong> Hematology-Oncology, Department <strong>of</strong><br />

Medicine, Samsung Medical Center, Sungkyunkwan University School <strong>of</strong><br />

Medicine, Seoul, South Korea<br />

Background: Thymic epithelial tumors (TETs), thymoma and thymic carcinoma,<br />

are the most common tumor <strong>of</strong> the anterior mediastinum. Initial<br />

complete resection is the most powerful prognostic indicator <strong>of</strong> survival.<br />

However, it is obviously related to stage. Here, we report the result <strong>of</strong> a<br />

prospective phase II study <strong>of</strong> neoadjuvant docetaxel/cisplatin in patients<br />

with locally advanced TETs. Methods: In this open-label, phase II, nonrandomized<br />

study, patients with histologically proven, Masaoka stage III/IV<br />

TETs at presentation were enrolled. Patients received docetaxel 75mg/m2 I.V for 1 hr, followed by I.V cisplatin 75mg/m2 over 1.5hr on day 1 <strong>of</strong> every<br />

3 week. After 3 cycles <strong>of</strong> chemotherapy, subsequent surgical resection was<br />

performed, if resectable. Results: From March 2007 to July 2011, a total <strong>of</strong><br />

27 TETs patients were entered into the trial. The median age was 54<br />

(range, 15-68), and Masaoka stage at presentation was III (n�8, 29.6%),<br />

IVA (n�17, 63.0%), and IVB (n�2, 7.4%). Histologic type by the WHO<br />

includes type B1 (n�2, 7.4%), B2 (n�3, 11.1%), B3 (n�5, 18.5%), and<br />

thymic carcinoma designated as type C (n�16, 59.3%). After completion<br />

<strong>of</strong> neoadjuvant chemotherapy, 17 (63.0%) achieved PR and 10 (37.0%)<br />

had SD. Nineteen patients (70.4%) underwent surgical resection, and 8<br />

patients did not (surgeons’ decision, n�5; patients’ refusal, n�2; radiation<br />

therapy, n�1). Of the 19 patients undergoing surgical resection, 17<br />

(89.5%) had complete resections and 2 (10.5%) did not. Major side<br />

effects <strong>of</strong> chemotherapy include grade 3 anorexia (n�1), nausea (n�2),<br />

diarrhea (n�3), alopecia (n�1). After completion <strong>of</strong> surgical resection,<br />

adjuvant therapy was performed as follows: radiotherapy (RT, n�8),<br />

chemotherapy (CTx, n�6), radiotherapy with chemotherapy (RT � CTx,<br />

n�2) and observation (n�3). Overall, 4yr OS and PFS was 79.4% and<br />

40.6%. Patients with complete resection showed 93.8% <strong>of</strong> 4yr OS and<br />

50.2% <strong>of</strong> 4yr PFS whereas patients without complete resection showed<br />

47.6% <strong>of</strong> 4yr OS and 26.7% <strong>of</strong> 4yr PFS, respectively (OS, p�0.06; PFS,<br />

p�0.27). Conclusions: Neoadjuvant docetaxel/cisplatin was well tolerated<br />

and feasible, and improved the surgical respectability in patients with<br />

advanced TETs.<br />

7106 General Poster Session (Board #43A), Sat, 1:15 PM-5:15 PM<br />

A 19-gene prognostic GEP signature (DecisionDx-Thymoma) to determine<br />

metastatic risk associated with thymomas. Presenting Author: Yesim<br />

Gokmen-Polar, Indiana University School <strong>of</strong> Medicine, Indianapolis, IN<br />

Background: The treatment <strong>of</strong> thymomas is predominantly based on the<br />

stage <strong>of</strong> disease. Histologic classification is <strong>of</strong> limited value as all types <strong>of</strong><br />

thymomas can give rise to metastases. In order to better predict the<br />

metastatic behavior <strong>of</strong> these tumors, we performed genome-wide gene<br />

expression analysis and identified a set <strong>of</strong> genes associated with presence<br />

or absence <strong>of</strong> metastases. In the current study, we sought to further develop<br />

and validate the gene signature using quantitative RT-PCR analysis.<br />

Methods: Thymomas with archived blocks and long-term follow-up data<br />

were reviewed. This training set study consisted <strong>of</strong> 50 cases, including 34<br />

cases on which the discovery microarray analysis had been performed. RNA<br />

was extracted from 5 x 10-micron thick sections in a CAP-accredited CLIA<br />

certified laboratory and analyzed by RT-PCR using custom TLDA cards on<br />

an ABI7900HT instrument. Expression data and biostatistical analysis<br />

were performed using GeNorm and JMP Genomics (SAS). Predictive<br />

modeling using <strong>Part</strong>ition Tree Analysis (PTA) and Logistic Regression<br />

Analysis (LRA) was performed. Metastasis-free survival (MFS) was assessed<br />

using Kaplan-Meier analysis. Results: A 19-gene expression pr<strong>of</strong>ile<br />

(GEP) signature was developed using a cohort <strong>of</strong> 50 thymomas for<br />

predicting metastasis. PTA yielded ROC <strong>of</strong> 0.97 (met. accuracy � 96%,<br />

non-met. accuracy � 81%), while LRA yielded ROC <strong>of</strong> 0.895 (met.<br />

accuracy � 87%, non-met. accuracy � 85%). PTA classification showed<br />

5-year MFS rates <strong>of</strong> 100% and 31% for predicted low risk (Class 1) and<br />

high risk (Class 2) <strong>of</strong> metastasis (median MFS � NR and 4.1 yrs, resp.,<br />

P�0.0001 Log-Rank), respectively. LRA showed 5-year MFS rates <strong>of</strong><br />

100% and 17% for predicted Class 1 and high risk Class 2 <strong>of</strong> metastasis<br />

(median MFS � NR and 2.9 yrs, resp., P�0.0001 Log-Rank), respectively.<br />

Analysis <strong>of</strong> additional cohorts is ongoing. Conclusions: We have successfully<br />

completed development <strong>of</strong> a 19-gene signature (DecisionDx-Thymoma)<br />

that appears to predict metastatic behavior <strong>of</strong> thymomas more accurately<br />

than traditional staging. If validated in larger cohort, this signature will<br />

provide insight for the future management <strong>of</strong> patients with this rare<br />

malignancy.<br />

477s<br />

7105 General Poster Session (Board #42H), Sat, 1:15 PM-5:15 PM<br />

Neoadjuvant treatment <strong>of</strong> primary inoperable or local recurrent thymoma<br />

with octreotide LAR to improve tumor resectability. Presenting Author:<br />

Berthold Schalke, Department <strong>of</strong> Neurology, University <strong>of</strong> Regensburg,<br />

Regensburg, Germany<br />

Background: The therapeutic outcome for unresectable, locally advanced,<br />

malignant thymoma is poor. Most important factor for long-term survival in<br />

thymoma patients is complete resection (R0) <strong>of</strong> the tumor. The study was<br />

performed to evaluate the efficacy <strong>of</strong> octreotide LAR plus prednisone in<br />

patients with primary inoperable or local recurrent thymoma to reduce<br />

tumor size. Methods: This was an open label, single-arm study in patients<br />

with inoperable or local recurrent thymoma. Patients were considered<br />

unlikely to achieve R0 resection at enrollment. Octreotide LAR was<br />

administered once every 2 weeks in combination with prednisone. Two<br />

stages were planned according to Fleming’s one sample multiple testing<br />

procedure for phase II clinical trials. The objective <strong>of</strong> the study was to show<br />

that octreotide LAR is effective in this patient population with respect to<br />

tumor shrinkage. Response was defined as decrease in tumor volume <strong>of</strong> at<br />

least 20% at month 3 as compared to baseline. Results: 17 thymoma<br />

patients at Masaoka stage III were recruited. Octreotide LAR showed a<br />

response in 15 <strong>of</strong> 17 patients (88.24%) at week 12. Two patients had<br />

discontinued the study before week 12 due to unsatisfactory therapeutic<br />

effect or adverse events. At Week 12, 5 patients (29.41%) operable for<br />

radical resection. 10 patients (58.82%) were not operable for radical<br />

resection. 16 <strong>of</strong> 17 patients (94.12%) experienced adverse events (AEs).<br />

The most frequent AEs were gastrointestinal disorders (70.59%), infections<br />

and infestations (64.71%), and blood/lymphatic system disorders<br />

(41.18%). Conclusions: Octreotide LAR was shown to be effective in<br />

patients with inoperable thymoma with respect to tumor shrinkage.Octreotide<br />

LAR was generally well tolerated. The reported AEs are in<br />

accordance with the known safety pr<strong>of</strong>ile.<br />

7107 General Poster Session (Board #43B), Sat, 1:15 PM-5:15 PM<br />

Factors affecting survival <strong>of</strong> patients with Masaoka stage IV thymic<br />

epithelial tumors (TET). Presenting Author: Yaman Suleiman, Indiana<br />

University School <strong>of</strong> Medicine, Indianapolis, IN<br />

Background: Thymoma and thymic carcinoma (TC) are rare tumors, but<br />

represent the most common neoplasms <strong>of</strong> the anterior mediastinum. The<br />

vast majority <strong>of</strong> TET present in early stages with little data existing on<br />

factors influencing survival in patients with advanced or stage IV disease.<br />

Methods: A retrospective analysis was performed on patients with confirmed<br />

TET (histology and with tissue blocks) seen at IUSCC diagnosed between<br />

1976 and 2011. Patient demographics including Masaoka stage, histology,<br />

and sites <strong>of</strong> metastasis were linked with progression free survival (PFS)<br />

and overall survival (OS). Results: Our analysis included 102 patients with<br />

stage IV TET: 50 presented de novo and 52 developed stage IV disease<br />

following primary treatment. When stratified by tumor histology (thymoma<br />

or TC), patients with TC had considerably poorer PFS (p�1.87x10-7 ) and<br />

OS (p�7.72x10-8 ). The median PFS for TC was 13 months (range 4 to 39)<br />

and the MST was 36 months (range 4 to 115). PFS at 5-years was 21% and<br />

0% but the five-year OS was 84% and 29% for thymoma and TC,<br />

respectively. Ten year OS was 55% for patients with thymoma and 0% for<br />

those with TC. Pleural (�3 vs. �3) metastases were significantly associated<br />

with a better PFS (p�0.036) and OS (p�0.0003). The PFS and OS <strong>of</strong><br />

patients with lung nodules trended with those with pleural metastasis.<br />

Patients with pleural metastasis and lung nodules sites did considerably<br />

better than those with visceral disease (PFS, p�0.004, OS, p�2.09x10-5 ).<br />

Conclusions: Patients with TC have significantly poorer PFS and OS when<br />

compared to thymoma confirming that TC is a distinct clinical entity from<br />

thymoma. Patients with thymoma may have prolonged survival, despite<br />

having residual disease. Although current staging places patients with<br />

pleural metastasis (Masaoka stage IVA) and those with lung nodules (stage<br />

IVB) as separate categories, our data would suggest that those with lung<br />

nodules have similar survival with those who have pleural metastasis.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


478s Lung Cancer—Non-small Cell Local-Regional/Small Cell/Other Thoracic Cancers<br />

7108 General Poster Session (Board #43C), Sat, 1:15 PM-5:15 PM<br />

Factors influencing outcome in thymic epithelial tumors (TET). Presenting<br />

Author: Ryan Frederick Porter, Indiana University School <strong>of</strong> Medicine,<br />

Indianapolis, IN<br />

Background: TET represent a heterogeneous collection <strong>of</strong> rare tumors,<br />

thymoma and thymic carcinoma (TC). Prognosis is <strong>of</strong>ten based on WHO<br />

histology and Masaoka Stage (MS) with small series <strong>of</strong>ten lacking long-term<br />

follow-up or advance stage disease. We examined predictors <strong>of</strong> relapse,<br />

disease-free survival (DFS) and overall survival (OS) based on WHO<br />

classification and MS. Methods: A retrospective analysis was performed on<br />

patients(pts) with TET seen at IUSCC from 1976 to 2011 with available<br />

tissue blocks. MS, WHO histology, demographics, autoimmune coexistence<br />

and non-TET neoplasms were correlated with DFS and OS (via<br />

Kaplan-Meier analysis). Results: Of 251 pts identified, MS at diagnosis was<br />

I(n�74), II(n�43), III(n�71), IVa(n�38), IVb(n�12) and indeterminate<br />

(n�13). The histology were thymoma (n�195) and TC (n�56). Median age<br />

was 51(10-88) and (M:F �121:130). Autoimmune disease was present<br />

(n�82) (myasthenia gravis n�52) and non-TET neoplasms (n�36).<br />

Therapy after diagnosis included surgery alone (n�115), radiation (n�29),<br />

chemo (n�71), chemo-radiotherapy (n�29), indeterminate(n�7). Median<br />

follow up was 69 months (1 to 399.5). DFS for stages I,II,III at 5 years was<br />

83%, 71% and 39%; DFS was associated with MS (p�.001) and WHO<br />

classification (p�.028). The 5yr and 10yr OS for stages I, II, III, IVa, IVb<br />

were 90%, 94%, 80%, 55%, 14% and 65%, 52%, 53%, 28%, 0%. The<br />

5yr OS for WHO classification were: A/AB-89%, B1/B2/B3-98% and<br />

C-30% and 10yr survivals were 89%, 62%, 0%. OS was associated with<br />

MS (p�.01) , WHO classification (p�.001) and autoimmune disease<br />

(p�.03). Late relapses (�5yr) occurred in 13 (23%) pts, including 3 with<br />

Stage I and 2 with WHO A classification. Conclusions: MS, WHO classification<br />

and autoimmune disorders were statistically significant with OS and<br />

with DFS for MS and WHO. TC prognosis is significantly worse than<br />

thymoma. The improved OS with autoimmune disease requires further<br />

investigation but raises the possibility <strong>of</strong> benefit derived from earlier<br />

diagnosis and/or heightened immune surveillance. These data support the<br />

notion that all TET, regardless <strong>of</strong> histologic subtype or clinical stage, are<br />

malignant with a capacity for metastasis. As relapses beyond 5 years are<br />

common, lifelong follow-up for TET is recommended.<br />

TPS7110 General Poster Session (Board #43E), Sat, 1:15 PM-5:15 PM<br />

Vinorelbine plus cisplatin versus gefitinib in resected non-small cell lung<br />

cancer haboring activating EGFR mutation (WJOG6410L). Presenting<br />

Author: Hirohito Tada, Department <strong>of</strong> General Thoracic Surgery, Osaka City<br />

General Hospital, Osaka, Japan<br />

Background: Vinorelbine plus cisplatin after completely resected stage II-III<br />

non-small cell lung cancer (NSCLC) is a standard therapy. Stage IV<br />

non-small cell lung cancer (NSCLC) harboring mutations in the epidermal<br />

growth factor receptor (EGFR) is quite sensitive to tyrosine kinase inhibitors<br />

(TKIs). Three randomized trials demonstrated that gefitinib is superior to<br />

platinum based chemotherapy in progression free survival. Retrospective<br />

analysis <strong>of</strong> adjuvant TKIs therapy for patient with resected lung cancer<br />

harboring EGFR mutation showed favorable trend toward improvement in<br />

disease free survival (DFS) and overall survival (OS). (J Thorac Oncol.<br />

2011;6: 569–575) BR19 comparing adjuvant gefitinib vs. placebo for<br />

completely resected NSCLC without any selection <strong>of</strong> biomarker was closed<br />

early and did not show any benefit <strong>of</strong> adjuvant gefitinib, even in the EGFR<br />

mutation positive cohort. The randomized trial in adjuvant therapy with<br />

erlotinib vs. placebo for patients with overexpression <strong>of</strong> EGFR protein has<br />

complete enrolment already. We conduct the first randomized phase III<br />

trial comparing adjuvant gefitinib with chemotherapy in patients with<br />

completely resected stage II-III NSCLC harboring EGFR mutations. Methods:<br />

Patients who have undergone complete resection and have EGFR mutation,<br />

deletions in exon 19, or L858R point mutation at exon 21 and without<br />

T790M mutation are randomly assigned gefitinib 250mg a day for 2 years<br />

or vinorelbine 25mg/m2 days 1 and 8, plus cisplatin 80mg/m2 day 1, every<br />

3 weeks for 4 courses. The primary endpoint is DFS and secondary<br />

endpoints are OS and safety. On the basis <strong>of</strong> previous studies, we assume<br />

the hazard ratio <strong>of</strong> DFS is 0.65. To demonstrate this improvement in DFS,<br />

230 patients in total would be needed during 3-year accrual period. This<br />

trial has begun from September 12 among 22 institutes in Japan. Until now<br />

(January 31. 2012), 13 cases have been enrolled. Five cases were enrolled<br />

in latest 1 month.<br />

TPS7109 General Poster Session (Board #43D), Sat, 1:15 PM-5:15 PM<br />

International tailored chemotherapy adjuvant trial: Itaca trial. Presenting<br />

Author: Silvia Novello, University <strong>of</strong> Turin, Orbassano, Italy<br />

Background: This is an ongoing phase III multicenter randomized trial<br />

comparing adjuvant pharmacogenomic-driven chemotherapy, based on<br />

thymidilate synthase (TS) and excision-repair cross-complementing -1<br />

(ERCC1) gene expression versus standard adjuvant chemotherapy in<br />

completely resected stage II-IIIA non-small cell lung cancer (EudraCT #:<br />

2008-001764-36). Methods: In all registered patients, before randomization,<br />

expression <strong>of</strong> ERCC1 and TS is assessed by qRT-PCR on paraffinembedded<br />

tumor specimens in a central laboratory. Randomization is<br />

stratified by stage and smoking status. Trial was emended on Feb, 2011<br />

with the 7th staging system. Primary end point is overall survival; secondary<br />

end points include recurrence-free survival, therapeutic compliance, toxicity<br />

pr<strong>of</strong>ile and comparative evaluation <strong>of</strong> ERCC1 and TS mRNA versus<br />

protein. It is assumed that the 5-year survival in the control arm is 45% and<br />

the hazard reduction associated to the experimental treatment is 30%.<br />

With a power <strong>of</strong> 90% to detect the estimated effect with log-rank test, a<br />

significant level <strong>of</strong> 5% (2 tails), 336 events have to be observed; the<br />

expected total number <strong>of</strong> patients is 700. The final statistical analysis will<br />

group together all control arms and all tailored chemotherapies groups.<br />

Efficacy analysis will be done on an intent-to-treat basis. Cox proportional<br />

hazard model will be used for estimating hazard ratios after adjusting for<br />

relevant variables. Within 45 days post-surgery, patients in each genetic<br />

pr<strong>of</strong>ile are randomized to receive either a standard chemotherapy selected<br />

by the investigator (cisplatin/vinorelbine, cisplatin/docetaxel or cisplatin/<br />

gemcitabine) or an experimental treatment (tailored arms) selected as<br />

follows: 1) high ERCC1 and high TS 4 cycles <strong>of</strong> single agent paclitaxel 2)<br />

high ERCC1 and low TS 4 cycles <strong>of</strong> single agent pemetrexed 3) low ERCC1<br />

and high TS 4 cycles <strong>of</strong> cisplatin/gemcitabine 4) low ERCC1 and low TS) 4<br />

cycles <strong>of</strong> cisplatin/pemetrexed. All chemotherapy regimens are administered<br />

for a total <strong>of</strong> 4 cycles on a 3-weekly basis. Currently, 312 patients<br />

have been randomized from 26 institutions mainly located in Italy and<br />

Germany (average enrolment: 13 patients/month).<br />

TPS7111 General Poster Session (Board #43F), Sat, 1:15 PM-5:15 PM<br />

Postoperative follow-up <strong>of</strong> lung cancer: Randomized trial comparing two<br />

follow-up programs in completely resected non-small cell lung cancer<br />

(IFCT-0302). Presenting Author: Virginie Westeel, Besançon University<br />

Hospital, Besançon, France<br />

Background: There are no robust data published on the follow-up after<br />

surgery for non-small cell lung cancer (NSCLC). Current international<br />

guidelines are informed by expert opinion. Most <strong>of</strong> them recommend<br />

regular follow-up with clinic visit and thoracic imaging, either chest X-ray <strong>of</strong><br />

Chest CT-scan. The IFCT-0302 trial addresses the question whether a<br />

surveillance program with chest CT-scan and fiberoptic bronchoscopy can<br />

improve survival compared to a follow-up only based on physical examination<br />

and chest x-ray. There is no such trial ongoing over the world. Methods:<br />

The IFCT-0302 trial is a multicenter open-label controlled randomized<br />

phase III trial. The objective <strong>of</strong> the trial is to compare two follow-up<br />

programs after surgery for stage I-IIIa NSCLC. The primary endpoint is<br />

overall survival. Patients are randomly assigned to arm 1, minimal<br />

follow-up, including physical examination and chest x-ray; or arm 2, a<br />

follow-up consisting <strong>of</strong> physical examination and chest x-ray plus chest CT<br />

scan and fiberoptic bronchoscopy (optional for adenocarcinomas). In both<br />

arms, follow-up procedures are performed every 6 months during the first<br />

two postoperative years, and every year between the third and the fifth<br />

years. The main eligibility criteria include: completely resected stage I-IIIA<br />

(6th UICC TNM classification) or T4 (in case <strong>of</strong> nodules in the same lobe as<br />

the tumor) N0 M0 NSCLC, surgery within the previous 8 weeks. Patients<br />

who have received and/or who will receive pre/post-operative chemotherapy<br />

and/or radiotherapy are eligible. Statistical considerations: 1,744 patients<br />

is required. Accrual status: 1,568 patients from 119 French centers had<br />

been included. The end <strong>of</strong> accrual can be expected for September 2012.<br />

Ancillary study: Blood samples are collected in 1000 patients for genomic<br />

high density SNP micro-array analysis. This collection will contribute to the<br />

French genome wide association study (gwas) <strong>of</strong> lung cancer gene susceptibility,<br />

and the genetic factors predictive <strong>of</strong> survival and lung cancer<br />

recurrence will be analyzed.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


Lung Cancer—Non-small Cell Local-Regional/Small Cell/Other Thoracic Cancers<br />

TPS7112 General Poster Session (Board #43G), Sat, 1:15 PM-5:15 PM<br />

IFCT-GFPC-0701 MAPS trial, a multicenter randomized phase III trial <strong>of</strong><br />

pemetrexed-cisplatin with or without bevacizumab in patients with malignant<br />

pleural mesothelioma (MPM). Presenting Author: Gerard Zalcman,<br />

Caen University Hospital, Caen, France<br />

Background: MPM median OS does not exceed 13 months with pem/CDDP<br />

doublet. U.S. Intergroup phase II trial <strong>of</strong> gemcitabine/CDDP, with or<br />

without bevacizumab, gave an appealing 15.6 months median OS in the<br />

bevacizumab arm. French Intergroup aimed to test pem/CDDP with<br />

bevacizumab (PCB), in a randomized phase III trial. Methods: Eligible<br />

patients had unresectable histologically proved MPM, no prior chemo, PS<br />

0-2, no thrombosis, nor bleeding. Primary endpoint: The primary outcome<br />

will be survival. The secondary endpoint will be Progression-Free Survival.<br />

Patients received pem 500 mg/m2 , CDDP 75 mg/m2 (PC),at D1, and<br />

vitamin B12 �B9 substitution, with (arm B) or without bevacizumab (arm<br />

A), 15 mg/kg Q21D, for 6 cycles. Arm B nonprogressive patients received<br />

bevacizumab maintenance therapy until progression or toxicity. 445<br />

patients to be recruited during a period 48 months, with at least 24 months<br />

<strong>of</strong> follow-up, and 385 events (deaths), will be needed to assure a power <strong>of</strong><br />

80% and detect at least a 4.3 months <strong>of</strong> median survival increase. This<br />

hypothesis leads to a Hazard Ratio (HR) <strong>of</strong> 1.33 and a 3-years survival <strong>of</strong><br />

14.7% in control arm and 23.6 % in experimental arm, with an absolute<br />

difference <strong>of</strong> 8.9% in survival rates. Accrual status: The first patient was<br />

included in February 2008. On January 31, 2012, 257 patients from 85<br />

French centers had been enrolled. The end <strong>of</strong> accrual can be expected for<br />

September 2013. Ancillary studies: For molecular biomarker analyses,<br />

thoracoscopic tissue specimens (TS, ERCC1, MSH2, TUBB3, NF2, p16,<br />

RASSF1A methylation,15 microRNAs ) and blood samples (micro-RNAS,<br />

VEGF, osteopontin, SRMP) at diagnosis are centrally collected. Finally, a<br />

prospective study comparing PET-CT to standard CT with central blinded<br />

analysis, is currently on-going for evaluation <strong>of</strong> response, and accuracy <strong>of</strong><br />

modified RECIST criteria for mesothelioma.<br />

479s<br />

TPS7113 General Poster Session (Board #43H), Sat, 1:15 PM-5:15 PM<br />

CA184-156: A randomized, multicenter, double-blind, phase III trial<br />

comparing the efficacy <strong>of</strong> ipilimumab (Ipi) plus etoposide/platinum (EP)<br />

versus EP in subjects with newly diagnosed extensive-stage disease small<br />

cell lung cancer (ED-SCLC). Presenting Author: David R. Spigel, Sarah<br />

Cannon Research Institute, Nashville, TN<br />

Background: EP (4-6 cycles) is standard <strong>of</strong> care 1st-line therapy for<br />

metastatic SCLC, and no multinational studies have reported any improvement<br />

beyond that reported for EP. Evidence <strong>of</strong> an ongoing immune<br />

response to SCLC tumors suggests that immunotherapy that enhances this<br />

immune response to SCLC may enhance the clinical benefit <strong>of</strong> EP. Ipi, a<br />

fully human monoclonal antibody which blocks CTLA-4, augments antitumor<br />

immune responses. Because some responses to Ipi may differ from<br />

those observed with cytotoxic therapies, immune-related response criteria<br />

(irRC) were derived from WHO criteria to better capture response patterns<br />

observed with Ipi. A randomized Phase 2 study <strong>of</strong> Ipi with paclitaxel/<br />

carboplatin (PC) in pts with ED-SCLC showed significant improvement in<br />

progression-free survival (PFS), as measured by irRC, over PC alone in pts<br />

receiving Ipi and PC in a phased regimen (Ipi started after 2 cycles <strong>of</strong> PC).<br />

Furthermore, addition <strong>of</strong> Ipi did not exacerbate PC toxicity, and immunerelated<br />

adverse events were managed using protocol-specific guidelines.<br />

This multicenter phase III study in pts with ED-SCLC (<strong>Clinical</strong>Trials.gov<br />

identifier NCT01450761) will determine whether adding Ipi to EP increases<br />

OS vs EP alone. Methods: EP consists <strong>of</strong> 4 cycles <strong>of</strong> etoposide (100<br />

mg/m2 , IV on Days 1-3 every 3 weeks [Q3W]) and cisplatin (75 mg/m2 , IV)<br />

or carboplatin (AUC�5, IV) once Q3W. Pts will be randomized to receive 4<br />

doses <strong>of</strong> Ipi (10 mg/kg, IV) in Arm A or placebo in Arm B, Q3W during<br />

induction, starting after 2 cycles <strong>of</strong> EP (phased schedule). Eligible pts will<br />

then receive blinded study drug (Ipi in Arm A; placebo in Arm B) Q12W<br />

until disease progression or unacceptable toxicity. The primary objective is<br />

to compare OS. Secondary objectives are to compare OS in those who<br />

receive blinded study drug, compare PFS between study arms, and to<br />

estimate best overall response rate and duration <strong>of</strong> response. First-line<br />

ED-SCLC pts with ECOG performance �1 will be included. Pts with<br />

symptomatic CNS metastases or a history <strong>of</strong> autoimmune disease will be<br />

excluded. The study will randomize 1100 pts at a 1:1 ratio.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


480s Lung Cancer—Non-small Cell Metastatic<br />

LBA7500 Oral Abstract Session, Mon, 3:00 PM-6:00 PM<br />

LUX-Lung 3: A randomized, open-label, phase III study <strong>of</strong> afatinib versus<br />

pemetrexed and cisplatin as first-line treatment for patients with advanced<br />

adenocarcinoma <strong>of</strong> the lung harboring EGFR-activating mutations. Presenting<br />

Author: James Chih-Hsin Yang, National Taiwan University Hospital,<br />

Taipei, Taiwan<br />

The full, final text <strong>of</strong> this abstract will be available at<br />

abstract.asco.org at 12:01 AM (EDT) on Monday, June 4,<br />

2012, and in the <strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong> online<br />

supplement to the June 20, 2012, issue <strong>of</strong> Journal <strong>of</strong><br />

<strong>Clinical</strong> Oncology. Onsite at the <strong>Meeting</strong>, this abstract will<br />

be printed in the Monday edition <strong>of</strong> ASCO Daily News.<br />

7502 Oral Abstract Session, Mon, 3:00 PM-6:00 PM<br />

SELECT: Randomized phase III study <strong>of</strong> docetaxel (D) or pemetrexed (P)<br />

with or without cetuximab (C) in recurrent or progressive non-small cell<br />

lung cancer (NSCLC) after platinum-based therapy. Presenting Author:<br />

Edward S. Kim, University <strong>of</strong> Texas M. D. Anderson Cancer Center,<br />

Houston, TX<br />

Background: SELECT investigated whether the addition <strong>of</strong> C to standard<br />

chemotherapy improved progression-free survival (PFS) in patients (pts)<br />

with recurrent or progressive NSCLC after failure <strong>of</strong> platinum-based<br />

therapy. Methods: SELECT was a multicenter, open label, randomized<br />

phase III trial. Per investigator choice, pts received either P (500 mg/m2 )or<br />

D (75 mg/m2 ) on day 1 and then were randomized within each group to<br />

chemotherapy plus C (400/250 mg/m2 ) (initial/weekly) or chemotherapy<br />

alone. Therapy was given for up to six 3-week cycles; pts randomized to C<br />

continued weekly monotherapy until disease progression or unacceptable<br />

toxicity. The primary objective was PFS for PC vs. P as determined by an<br />

Independent Review Committee (IRC). Secondary endpoints included<br />

overall survival (OS), objective response rate (ORR) and duration <strong>of</strong><br />

response (DOR) by IRC, and safety. Preplanned subgroup analyses for<br />

epidermal growth factor receptor (EGFR) staining intensity by immunohistochemistry<br />

and histology were performed. Results for PC vs. P only are<br />

presented. Results: Between Jan 2005 and Feb 2010, 938 total pts were<br />

randomized. Baseline demographics were comparable between PC (n�301)<br />

andP(n�304): median age 64 years; male 60%; Caucasian 88%; KPS<br />

80-100/60-70 84%/16%; squamous/non-squamous 24%/76%. Median<br />

PFS (months) PC: 2.89 and P: 2.76 (hazard ratio [HR] �1.03 [95%<br />

confidence interval (CI)�0.87-1.21]; p�0.76). Median OS (months) PC:<br />

6.93 and P: 7.79 (HR�1.01 [95% CI�0.86-1.20]; p�0.86). ORR PC:<br />

6.6% and P: 4.3% (odds ratio �1.59 [95% CI�0.78-3.26]; p�0.20).<br />

Median DOR (months) PC: 4.17 and P: 6.93 (HR�1.58 [95% CI�0.74-<br />

3.36]; p�0.24). There were no statistical differences in efficacy based on<br />

histology or EGFR staining intensity. More drug-related AEs/SAEs were<br />

observed in the PC arm, with differences mainly attributable to skin<br />

toxicities, GI (diarrhea/stomatitis), and hypomagnesemia. Conclusions: The<br />

addition <strong>of</strong> C to P did not improve efficacy in this pt population. Further<br />

biomarker analyses are planned. The safety pr<strong>of</strong>iles for C and P were<br />

consistent with existing data and no new safety signals were observed.<br />

LBA7501 Oral Abstract Session, Mon, 3:00 PM-6:00 PM<br />

TAILOR: Phase III trial comparing erlotinib with docetaxel in the secondline<br />

treatment <strong>of</strong> NSCLC patients with wild-type (wt) EGFR. Presenting<br />

Author: Marina Chiara Garassino, U.O. Oncologia Medica, A.O. Fatebenefratelli<br />

e Oftalmico, Milan, Italy<br />

The full, final text <strong>of</strong> this abstract will be available at<br />

abstract.asco.org at 12:01 AM (EDT) on Monday, June 4,<br />

2012, and in the <strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong> online<br />

supplement to the June 20, 2012, issue <strong>of</strong> Journal <strong>of</strong><br />

<strong>Clinical</strong> Oncology. Onsite at the <strong>Meeting</strong>, this abstract will<br />

be printed in the Monday edition <strong>of</strong> ASCO Daily News.<br />

7503 Oral Abstract Session, Mon, 3:00 PM-6:00 PM<br />

Phase II double-blind, randomized study <strong>of</strong> selumetinib (SEL) plus<br />

docetaxel (DOC) versus DOC plus placebo as second-line treatment for<br />

advanced KRAS mutant non-small cell lung cancer (NSCLC). Presenting<br />

Author: Pasi A. Janne, Dana-Farber Cancer Institute, Boston, MA<br />

Background: KRAS mutations are the most common (~20%) oncogenic<br />

alteration in NSCLC. There are no effective targeted therapies for this<br />

subset <strong>of</strong> NSCLC. Selumetinib (AZD6244, ARRY-142866) inhibits MEK1/2<br />

signaling downstream <strong>of</strong> KRAS. We prospectively evaluated SEL � DOC vs<br />

DOC � placebo in advanced KRAS mutant NSCLC based on preclinical<br />

observations (NCT00890825). Methods: Patients (pts) with stage IIIB-IV,<br />

KRAS mutant NSCLC, who had received prior chemotherapy, received iv<br />

DOC 75 mg/m2 , and po SEL 75 mg or placebo BD. The primary endpoint<br />

was overall survival (OS); secondary endpoints included: progression-free<br />

survival (PFS), objective response rate (RR), duration <strong>of</strong> response, change<br />

in tumor size, proportion <strong>of</strong> patients alive and progression-free at 6 mo, and<br />

safety and tolerability. Results: Between April 2009 and June 2010, 422<br />

pts were screened across 67 centers in 12 countries; 113 had KRAS<br />

mutant NSCLC and 87 were randomized (DOC, 43; SEL/DOC, 44).<br />

Baseline characteristics were balanced (DOC vs SEL/DOC): WHO PS 0,<br />

49%/48%; Female, 54%/52%; KRAS codon 12, 90%/93%. Median<br />

number <strong>of</strong> cycles: DOC, 4; SEL/DOC, 5. Most frequent grade 3/4 hematologic<br />

toxicity (DOC vs SEL/DOC): neutropenia (54.8%/67.4%), febrile<br />

neutropenia (0%/15.9%); most frequent grade 3/4 non-hematologic toxicity:<br />

dyspnea (11.9%/2.3%) asthenia (0%/9.1%), respiratory failure (4.8%/<br />

6.8%), acneiform dermatitis (0%/6.8%). Discontinuation due to AEs was<br />

similar: 18.2% SEL/DOC vs 11.9% DOC. OS was longer for SEL/DOC vs<br />

DOC (9.4 mo vs 5.2 mo; 56 events, median follow-up 219 days) but did not<br />

reach statistical significance; hazards were non proportional (HR 0.80;<br />

80% CI 0.56, 1.14; 1-sided p�0.2069). All secondary endpoints,<br />

including RR (DOC 0%, SEL/DOC 37%; p�0.0001) and PFS (DOC 2.1 mo,<br />

SEL/DOC 5.3 mo; 71 events; HR � 0.58; 80% CI 0.42, 0.79; 1-sided<br />

p�0.0138), were significantly improved for SEL/DOC vs DOC. Conclusions:<br />

This is the first prospective study to demonstrate a clinical benefit <strong>of</strong> a<br />

targeted therapy (SEL � DOC) for patients with KRAS mutant cancer <strong>of</strong> any<br />

type. Our findings could have implications for the treatment <strong>of</strong> NSCLC and<br />

other KRAS mutant cancers.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


7504 Oral Abstract Session, Mon, 3:00 PM-6:00 PM<br />

Analysis <strong>of</strong> resistance mechanisms to ALK kinase inhibitors in ALK�<br />

NSCLC patients. Presenting Author: Robert Charles Doebele, University <strong>of</strong><br />

Colorado Anschutz Medical Campus, Aurora, CO<br />

Background: Patients with anaplastic lymphoma kinase (ALK) gene fusions<br />

derive significant clinical benefit from crizotinib, an ALK inhibitor; moreover,<br />

next generation ALK kinase inhibitors are in development. Unfortunately,<br />

drug resistance develops after initial benefit (acquired) or occurs in<br />

patients who never derive a benefit (intrinsic). This study aimed to define<br />

molecular mechanisms <strong>of</strong> resistance to ALK kinase inhibitors in ALK�<br />

non-small cell lung cancer (NSCLC) patients. Methods: 30 ALK� crizotinibtreated<br />

NSCLC patients experienced radiologic disease progression, <strong>of</strong><br />

whom 7 progressed only in the CNS. Of the 23 patients with extra-CNS<br />

progression, a biopsy was attempted on 19. One <strong>of</strong> the patients without<br />

tissue post-crizotinib then proceeded to a second generation ALK inhibitor<br />

and tissue was obtained post progression on this drug. We performed<br />

molecular analysis and initiated cell lines from tumor tissue for these 19<br />

patients. Results: 15 patients had material evaluable for molecular analysis.<br />

Six patients (40%) developed secondary mutations in the kinase<br />

domain <strong>of</strong> ALK. Two novel mutations were identified in samples from ALK�<br />

NSCLC patients, F1174C and D1203N. Two patients each demonstrated<br />

G1269A or L1196M mutations. Two patients each, one with a resistance<br />

mutation, exhibited new onset ALK copy number gain (CNG). Four patients<br />

demonstrated the presence <strong>of</strong> another oncogenic driver (1 with EGFR<br />

mutation; 3 with KRAS mutation) with or without a persistent ALK gene<br />

rearrangement. One patient lacked an ALK gene fusion on progression<br />

biopsy, but had no identifiable alternate oncogene alteration. 3 patients<br />

retained ALK positivity with no identifiable resistance mechanism. Data on<br />

additional patients and an in vitro model <strong>of</strong> copy number gain will be<br />

presented. Conclusions: ALK kinase inhibitor resistance in ALK� NSCLC<br />

occurs through a diverse array <strong>of</strong> kinase domain mutations, ALK gene<br />

fusion CNG, and emergence <strong>of</strong> second (same cell) or separate (different<br />

cell) oncogenic drivers. In order to overcome resistance it will be important<br />

to differentiate patients that preserve ALK dominance (secondary mutations<br />

and CNG) versus those that have diminished ALK dominance<br />

(separate or second oncogenic drivers).<br />

7506 Oral Abstract Session, Mon, 3:00 PM-6:00 PM<br />

A randomized phase III trial <strong>of</strong> single-agent pemetrexed (P) versus<br />

carboplatin and pemetrexed (CP) in patients with advanced non-small cell<br />

lung cancer (NSCLC) and performance status (PS) <strong>of</strong> 2. Presenting Author:<br />

Rogerio Lilenbaum, Cleveland Clinic Florida, Weston, FL<br />

Background: No standard <strong>of</strong> care exists for patients with advanced NSCLC<br />

and PS 2 and clinical practice ranges from supportive care to combination<br />

chemotherapy. Methods: In a Brazilian multicenter phase III randomized<br />

trial, advanced NSCLC patients, with any histology at first, amended to<br />

non-squamous only, PS 2, no prior chemotherapy, and adequate organ<br />

function, were randomized to P alone (500 mg/m2) or CP (AUC 5 � same<br />

P) administered every 3 weeks for 4 cycles. Stratification factors included<br />

stage (IIIB vs. IV); age (�70 vs. �70); and weight loss (�5 kgvs.�5kg).<br />

The primary endpoint was overall survival and the study was powered to<br />

demonstrate an improvement in median survival from 2.9 to 4.3 months<br />

based on a prior CALGB trial. Results: A total <strong>of</strong> 217 patients were enrolled<br />

from 8 centers in Brazil and 1 in the US from April 2008 to July 2011.<br />

Twelve patients were ineligible and excluded. The 2 arms (P�102;<br />

CP�103) were balanced for patient characteristics. 14 patients had<br />

squamous and another 12 had unknown histology. The response rates were<br />

P � 10% and CP � 24% (p�0.019). In the ITT population, the median<br />

PFS was P � 3.0 mo and CP � 5.9 mo (HR�0.46, 95% CI 0.34; 0.63,<br />

p�0.001) and median OS was P � 5.6 mo vs. CP � 9.1 mo (HR�0.57,<br />

95% CI 0.41; 0.79, p�0.001). 1-year survival rates were 22% and 39%<br />

respectively. Similar results were seen when squamous patients were<br />

excluded from the analysis. Grade ¾ anemia (5.5%; 12%) and neutropenia<br />

(2.8%; 5.6%) were more frequent in CP. There were 4 treatment-related<br />

deaths in the CP arm. 30% <strong>of</strong> patients in each arm received 2nd line therapy<br />

Conclusions: Combination chemotherapy with CP significantly improves<br />

survival, with acceptable safety, in eligible patients with advanced NSCLC<br />

and PS 2, and represents a new standard.<br />

Lung Cancer—Non-small Cell Metastatic<br />

481s<br />

7505 Oral Abstract Session, Mon, 3:00 PM-6:00 PM<br />

Multiplex testing for driver mutations in squamous cell carcinomas <strong>of</strong> the<br />

lung. Presenting Author: Paul K. Paik, Memorial Sloan-Kettering Cancer<br />

Center, New York, NY<br />

Background: While the majority <strong>of</strong> lung adenocarcinomas (ADCL) harbor an<br />

identifiable driver mutation, targeted therapies for squamous cell lung<br />

carcinomas (SQCLC) have lagged in development due to a paucity <strong>of</strong><br />

druggable oncogenic events. Three targets, which together occur in up to<br />

50% <strong>of</strong> SQCLC, have been recently identified (FGFR1 amplification, DDR2<br />

mutations, PIK3CA mutations/PTEN loss). Comprehensive molecular analysis<br />

<strong>of</strong> SQCLC tumors by The Cancer Genome Atlas is ongoing, with new<br />

therapeutic targets on the horizon. Methods: We have instituted prospective,<br />

multiplex testing <strong>of</strong> SQCLC tumors (Squamous Cell Lung Cancer<br />

Mutation Analysis Program, “SQ-MAP”). Tests include FISH for FGFR1<br />

amplification (defined as FGFR1:CEP8 � 2in�10% <strong>of</strong> cells), IHC for loss<br />

<strong>of</strong> PTEN expression, and Sequenom MassARRAY for PIK3CA mutations<br />

(and others, below). We are also incorporating targeted exon sequencing<br />

(Agilent SureSelect/Ion Torrent) <strong>of</strong> a panel <strong>of</strong> over 80 lung cancer<br />

oncogenes and tumor suppressors in preparation for future studies. All<br />

tests were performed on formalin-fixed paraffin-embedded samples and<br />

with Institutional Review Board/Biospecimen Utilization Committee approval.<br />

Results: 40 SQCLC patient specimens have been processed through<br />

SQ-MAP over 3 months. 8 samples were excluded (insufficient tissue in 4,<br />

reclassification to ADCL in 4). Data are available for 28 patients. PTEN IHC<br />

was performed on 15 samples to date. Molecular results are summarized in<br />

the table. Events were non-overlapping. Results for the �80 gene sequencing<br />

component will be described. Based on SQ-MAP, accrual to 2 approved<br />

clinical trials (FGFR1 inhibition; PI3K inhibition) has begun, with a third<br />

planned (DDR2 mutations). Conclusions: Actionable defects were detected<br />

in 60% (95% CI: 37-75%) <strong>of</strong> SQCLC specimens. Mutation data for �80<br />

other oncogenes and tumor suppressors will be available shortly (including<br />

DDR2). SQ-MAP serves as a platform supporting both personalized care<br />

and research.<br />

Test Frequency 95% CI<br />

FGFR1 amplification 25% (7/28) 11-45%<br />

PTEN loss, complete 20% (3/15) 5-49%<br />

PIK3CA mutation 11% (3/28) 2-28%<br />

KRAS mutation 4% (1/28) 0-18%<br />

EGFR mutation 0%<br />

BRAF mutation 0%<br />

HER2 mutation 0%<br />

AKT1 mutation 0%<br />

NRAS mutation 0%<br />

MEK1 mutation 0%<br />

Any 60% 37%-75%<br />

LBA7507 Oral Abstract Session, Mon, 3:00 PM-6:00 PM<br />

PARAMOUNT: Final overall survival (OS) results <strong>of</strong> the phase III study <strong>of</strong><br />

maintenance pemetrexed (pem) plus best supportive care (BSC) versus<br />

placebo (plb) plus BSC immediately following induction treatment with<br />

pem plus cisplatin (cis) for advanced nonsquamous (NS) non-small cell<br />

lung cancer (NSCLC). Presenting Author: Luis Paz-Ares, University Hospital<br />

- Virgen del Rocio, Seville, Spain<br />

The full, final text <strong>of</strong> this abstract will be available at<br />

abstract.asco.org at 12:01 AM (EDT) on Monday, June 4,<br />

2012, and in the <strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong> online<br />

supplement to the June 20, 2012, issue <strong>of</strong> Journal <strong>of</strong><br />

<strong>Clinical</strong> Oncology. Onsite at the <strong>Meeting</strong>, this abstract will<br />

be printed in the Monday edition <strong>of</strong> ASCO Daily News.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


482s Lung Cancer—Non-small Cell Metastatic<br />

7508 <strong>Clinical</strong> Science Symposium, Sat, 8:00 AM-9:30 AM<br />

<strong>Clinical</strong> activity <strong>of</strong> crizotinib in advanced non-small cell lung cancer<br />

(NSCLC) harboring ROS1 gene rearrangement. Presenting Author: Alice<br />

Tsang Shaw, Massachusetts General Hospital Cancer Center, Boston, MA<br />

Background: Chromosomal rearrangements <strong>of</strong> the ROS1 receptor tyrosine<br />

kinase gene define a new molecular subset <strong>of</strong> NSCLC. In cell lines, ROS1<br />

rearrangements lead to expression <strong>of</strong> oncogenic ROS1 fusion kinases and<br />

sensitivity to ROS kinase inhibition. We examined the efficacy and safety <strong>of</strong><br />

crizotinib, a small molecule tyrosine kinase inhibitor <strong>of</strong> MET, ALK and ROS,<br />

in patients with advanced, ROS1-rearranged NSCLC. Methods: Patients<br />

with advanced NSCLC harboring ROS1 rearrangement, as determined<br />

using a break-apart FISH assay, were recruited into an expansion cohort <strong>of</strong><br />

a phase 1 study <strong>of</strong> crizotinib. Patients were treated with crizotinib at the<br />

standard oral dose <strong>of</strong> 250 mg BID. The objective response rate (ORR) was<br />

determined based on RECIST 1.0. The disease control rate (DCR; stable<br />

disease [SD] � partial response [PR] � complete response [CR]) was<br />

evaluated at 8 weeks. Results: Thirteen patients within the ROS expansion<br />

cohort received crizotinib and all were evaluable for response. The median<br />

age was 47 yrs (range 31–72), and all but one <strong>of</strong> the patients were<br />

never-smokers. All patients had adenocarcinoma histology. 12/13 patients<br />

were tested for ALK rearrangement and all were negative. The median<br />

number <strong>of</strong> prior treatments was 1 (range 0–3). To date, the ORR is 54%<br />

(7/13), with 6 PRs and 1 CR, with 6 responses achieved by the first<br />

restaging scan at 7–8 wks. There was 1 additional unconfirmed PR at the<br />

time <strong>of</strong> data cut-<strong>of</strong>f. The DCR at 8 wks was 85% (11/13). Median duration<br />

<strong>of</strong> treatment was 20 wks (range 4�–59�). All responses are ongoing, and<br />

12 patients continue on study. One patient had disease progression at first<br />

restaging and was discontinued from the study. The pharmacokinetics and<br />

safety pr<strong>of</strong>ile <strong>of</strong> crizotinib in this group <strong>of</strong> patients were similar to that<br />

observed in patients with ALK-positive NSCLC. Conclusions: Crizotinib<br />

demonstrates marked antitumor activity in patients with advanced NSCLC<br />

harboring ROS1 rearrangements. Like ALK, ROS defines a distinct subpopulation<br />

<strong>of</strong> NSCLC patients for whom crizotinib therapy may be highly<br />

effective. This study represents the first clinical validation <strong>of</strong> ROS as a<br />

therapeutic target in cancer.<br />

7510 <strong>Clinical</strong> Science Symposium, Sat, 8:00 AM-9:30 AM<br />

Discovery <strong>of</strong> recurrent KIF5B-RET fusions and other targetable alterations<br />

from clinical NSCLC specimens. Presenting Author: Marzia Capelletti,<br />

Dana-Farber Cancer Institute, Boston, MA<br />

Background: Many NSCLCs have driving oncogenic alterations including in<br />

EGFR, KRAS, ERBB2, BRAF, ALK and ROS1. <strong>Clinical</strong>ly effective drugs are<br />

approved for EGFR and ALK and clinical trials are underway for other<br />

genomic targets. Thus having a means <strong>of</strong> identifying genomic alterations in<br />

routine formalin fixed paraffin embedded (FFPE) clinical specimens is<br />

critical. Methods: We sequenced 24 FFPE NSCLC specimens with a next<br />

generation sequencing (NGS) assay that captures and sequences 2574<br />

coding exons <strong>of</strong> 145 cancer relevant genes plus 37 introns from 14 genes<br />

<strong>of</strong>ten rearranged in cancer. Tumors from 643 additional patients were<br />

genotyped for KIF5B-RET. Results: We identified 50 alterations in 21 genes<br />

with at least one in 83% (20/24) <strong>of</strong> tumors (range 1-7). In 72% (36/50) <strong>of</strong><br />

NSCLCs, at least one alteration was associated with a current clinical<br />

treatment or targeted therapy trial, including mutations in KRAS, BRAF,<br />

EGFR, MDM2, CDKN2A, CCNE1, CDK4, NF1 and PIK3CA. We also found<br />

an 11,294,741 bp pericentric inversion on chromosome 10 generating a<br />

novel gene fusion joining exons 1-15 <strong>of</strong> KIF5B to exons 12-20 <strong>of</strong> RET<br />

(K15:R12) in a Caucasian never smoker. This fusion gene contains the<br />

kinesin motor and coiled-coil domains <strong>of</strong> KIF5B and the entire RET tyrosine<br />

kinase domain. In 643 additional tumors we identified 11 fusion positive<br />

patients who were all wild type for known oncogenes (frequency <strong>of</strong> 6.3%<br />

(10/159)). Four unique KIF5B-RET variants were found: 8 K15:R12, 3<br />

K16:R12, 1 K22:R12 and 1 K15:R11. We introduced K15:R12 into<br />

Ba/F3 cells and observed IL-3 independent growth consistent with oncogenic<br />

transformation. KIF5B-RET Ba/F3 cells were sensitive to sunitinib,<br />

sorafenib and vandetinib, multi-targeted kinase inhibitors that inhibit RET,<br />

but not gefitinib, an EGFR kinase inhibitor. Sunitinib, but not gefitinib,<br />

inhibited RET phosphorylation in these cells. Conclusions: We identified<br />

both known and novel genomic alterations from NSCLC FFPE specimens<br />

using a single test. Our findings suggest that RET inhibitors should be<br />

tested in prospective clinical trials in NSCLC patients bearing KIF5B-RET<br />

rearrangements and that NGS is a feasible approach to stratifying patients<br />

for treatment based on their genomic pr<strong>of</strong>iles.<br />

7509 <strong>Clinical</strong> Science Symposium, Sat, 8:00 AM-9:30 AM<br />

<strong>Clinical</strong> activity and safety <strong>of</strong> anti-PD1 (BMS-936558, MDX-1106) in<br />

patients with advanced non-small-cell lung cancer (NSCLC). Presenting<br />

Author: Julie R. Brahmer, The Sidney Kimmel Comprehensive Cancer<br />

Center at Johns Hopkins University, Baltimore, MD<br />

Background: BMS-936558 is a fully human mAb that blocks the programmed<br />

death-1 (PD-1) co-inhibitory receptor expressed by activated T<br />

cells. We report here that BMS-936558 mediates antitumor activity in<br />

heavily pretreated patients (pts) with advanced NSCLC, a tumor historically<br />

not considered to be responsive to immunotherapy. Methods: BMS-936558<br />

was administered IV Q2WK to pts with various solid tumors, including<br />

NSCLC, at doses <strong>of</strong> 0.1 to 10 mg/kg during dose-escalation and/or cohort<br />

expansion. Pts with advanced NSCLC previously treated with at least 1 prior<br />

chemotherapy regimen were eligible. Pts received up to 12 cycles (4<br />

doses/cycle) <strong>of</strong> treatment or until PD or CR. <strong>Clinical</strong> activity was assessed<br />

by RECIST 1.0. Results: Of 240 pts treated as <strong>of</strong> July 1, 2011, 75 NSCLC<br />

pts were treated at 1 (n�17), 3 (n� 19), or 10 mg/kg (n�39). ECOG<br />

performance status was 0/1/2 in 24/49/2 pts. Tumor histology was<br />

squamous (n�17), non-squamous (n�52), or unknown (n�6). The number<br />

<strong>of</strong> prior therapies was 1 (n�9),2(n�20), �3 (n�45), or unknown<br />

(n�1). Ninety-five percent <strong>of</strong> pts had received platinum-based chemotherapy<br />

and 40% had a prior tyrosine kinase inhibitor. Sites <strong>of</strong> metastatic<br />

disease included lymph node (n�50), liver (n�12), lung (n�62), and bone<br />

(n�13). Median duration <strong>of</strong> therapy was 10 wk (max 100 wk). Common<br />

related AEs were fatigue (17%), nausea (11%), pyrexia (7%), pruritus<br />

(7%), dizziness (7%), and anemia (7%). The incidence <strong>of</strong> grade 3-4 related<br />

AEs was 8%. There was 1 drug-related death due to pulmonary toxicity.<br />

<strong>Clinical</strong> activity was observed at all dose levels (Table) and in multiple<br />

histologies. Several pts had prolonged SD. Some had a persistent decrease<br />

in overall tumor burden in the presence <strong>of</strong> new lesions and were not<br />

categorized as responders. At the time <strong>of</strong> data lock, <strong>of</strong> 10 OR pts, 3 had<br />

responses lasting �6 mo and 5 were on study with response duration <strong>of</strong><br />

1.8-5.5 mo. Conclusions: BMS-936558 is well tolerated and has encouraging<br />

clinical activity in pts with previously treated advanced NSCLC. Further<br />

development <strong>of</strong> BMS-936558 in pts with advanced NSCLC is warranted.<br />

Dose (mg/kg) na ORb uPRc RRd (%)<br />

1 18 1 - 6<br />

3 18 3 2 28<br />

10<br />

aResponse evaluable pts.<br />

bCR or PR.<br />

cUnconfirmed PR.<br />

dResponse rate [(OR � uPR) ÷ n].<br />

37 6 1 19<br />

7511 Poster Discussion Session (Board #1), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

A randomized double-blind trial <strong>of</strong> carboplatin plus paclitaxel (CP) with<br />

daily oral cediranib (CED), an inhibitor <strong>of</strong> vascular endothelial growth factor<br />

receptors, or placebo (PLA) in patients (pts) with previously untreated<br />

advanced non-small cell lung cancer (NSCLC): NCIC <strong>Clinical</strong> Trials Group<br />

study BR29. Presenting Author: Scott Andrew Laurie, Ottawa Hospital<br />

Cancer Centre, Ottawa, ON, Canada<br />

Background: In NCIC CTG study BR24, CED 30 mg/d � CP increased<br />

objective response rate (RR) and progression-free survival (PFS), but there<br />

were concerns regarding toxicity in some pts. BR29 tested a lower dose <strong>of</strong><br />

CED 20 mg/d limiting accrual to pts without significant weight loss/<br />

hypoalbuminemia. Methods: Consenting, eligible adult pts with advanced<br />

incurable NSCLC <strong>of</strong> any histology were randomized to receive CED 20 mg/d<br />

or PLA with up to 6 cycles <strong>of</strong> C (AUC � 6) P (200 mg/m2 ); non-progressing<br />

pts continued CED/PLA after CP until progression, unacceptable toxicity or<br />

pt request. The primary endpoint was overall survival (OS). An interim<br />

analysis (IA) for PFS was planned after 170 events in the first 260 pts; the<br />

study would continue if the hazard ratio (HR) for PFS was � 0.7. Accrual<br />

continued until the required number <strong>of</strong> events was reached then held<br />

pending IA. Results: The trial was halted when the IA (n�260) revealed a<br />

HR for PFS <strong>of</strong> 0.89 (95% CI 0.66-1.20). A final analysis including all 306<br />

randomized pts (median age 62, male 55%, PS 0 26%, PS 1 74%,<br />

adenocarcinoma 64%, squamous 13%, other histology 23%. RR was<br />

significantly higher with CED (52% vs 34 %, p � 0.001). For CED/PLA,<br />

respectively, median OS and PFS were 12.2/12.1 [HR: 0.95 (0.69-1.30,<br />

p�0.74)] and 5.5/5.5 months [HR: 0.91 (0.71-1.18, p�0.5)]. Grade �3<br />

hypertension (15% vs 3%, p�0.0002), anorexia (7% vs 1%, p�0.02) and<br />

diarrhea (16% vs 1%, p�0.0001) were all significantly increased with<br />

CED; there were 2 deaths possibly-related to CED [1 each hemorrhage,<br />

leukoencephalopathy (prior radiation)]. Conclusions: Adding a lower dose <strong>of</strong><br />

CED to CP increased RR and toxicity, but not PFS or OS.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


7512 Poster Discussion Session (Board #2), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Randomized phase II study <strong>of</strong> carboplatin and paclitaxel with either<br />

linifanib or placebo for advanced nonsquamous NSCLC. Presenting Author:<br />

Suresh S. Ramalingam, Winship Cancer Institute, Emory University,<br />

Atlanta, GA<br />

Background: Linifanib is a potent and selective inhibitor <strong>of</strong> VEGF and PDGF<br />

receptors with modest single-agent activity in NSCLC. We evaluated the<br />

combination <strong>of</strong> linifanib with carboplatin (C) and paclitaxel (P) for first-line<br />

therapy <strong>of</strong> advanced non-squamous NSCLC. Methods: Patients (pts) with<br />

stage IIIB/IV, non-squamous NSCLC, stratified by ECOG PS and gender,<br />

were randomized to receive up to six 3-wk cycles <strong>of</strong> C (AUC 6 mg/ml/min)<br />

and P (200 mg/m2 ) with daily placebo (Arm A), linifanib 7.5 mg (Arm B), or<br />

linifanib 12.5 mg (Arm C). The primary endpoint was progression-free<br />

survival (PFS); secondary endpoints included overall survival (OS), 12 m<br />

survival rate, and objective response rate (ORR). Safety was assessed by<br />

NCI-CTCAE v3.0. Results: 138 pts were randomized at 37 sites in 6<br />

countries. Baseline characteristics were: median age, 61 y; men, 57%;<br />

smoker, 84%. Efficacy results are shown in the table. Thrombocytopenia<br />

was the only Grade 3/4 AE significantly higher on linifanib (Arm B: 16.7%;<br />

Arm C: 29.8%) vs. placebo (2.1%). Other adverse events (AEs) related to<br />

the dose <strong>of</strong> linifanib were diarrhea, thrombocytopenia, hypertension,<br />

weight loss, palmar-plantar erythrodysaesthesia syndrome, and hypothyroidism.<br />

Analysis <strong>of</strong> samples for predictive biomarkers including serum VEGF<br />

and placental growth factor are underway. Conclusions: The addition <strong>of</strong><br />

linifanib to chemotherapy was tolerable at the doses tested and resulted in<br />

a significant improvement in PFS, with a modest survival improvement for<br />

Arm C in first-line therapy <strong>of</strong> advanced non-squamous NSCLC.<br />

Arm A<br />

N�47<br />

Arm B<br />

N�44<br />

Arm C<br />

N�47<br />

Median PFS, m [95% CI] 5.4 [4.2, 5.7] 8.3 [4.2, 10.8] HR 0.51, P�0.022* 7.3 [4.6, 10.9] HR 0.64, P�0.118*<br />

Median OS, m [95% CI] 11.3 [8.8, 17.0] 11.4 [6.6, 14.8]<br />

13.0 [8.3, 18.9]<br />

HR�1.08, P�0.779*<br />

HR�0.89, P�0.650*<br />

12 m survival rate, % 45 44 54<br />

ORR, % 25.5 43.2 31.9<br />

Received 2L therapy, n 26 13 18<br />

*Stratified log-rank test; p value compared with CP � placebo.<br />

7514 Poster Discussion Session (Board #4), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Activity <strong>of</strong> cabozantinib (XL184) in metastatic NSCLC: Results from a<br />

phase II randomized discontinuation trial (RDT). Presenting Author: Beth<br />

A. Hellerstedt, US Oncology Research, LLC, McKesson Specialty Health,<br />

The Woodlands, TX, and Texas Oncology, Central Austin Cancer Center,<br />

Austin, TX<br />

Background: Dysregulation <strong>of</strong> MET and VEGFR2 signaling has been observed<br />

in NSCLC and MET upregulation has been implicated in resistance<br />

to EGFR inhibitors. Cabozantinib (cabo) is an oral, potent inhibitor <strong>of</strong> MET<br />

and VEGFR2. A RDT evaluated activity and safety in 9 tumor types. Here we<br />

report on the metastatic NSCLC cohort which included patients who<br />

received prior EGFR and VEGF pathway targeted therapy. Methods: All<br />

eligible patients (pts) were required to have measurable disease at<br />

baseline. Pts received cabo at 100 mg qd over a 12 wk Lead-in stage.<br />

Tumor response (mRECIST) was assessed q6 wks. Treatment � wk 12 was<br />

based on response: pts with PR continued open-label cabo, pts with SD<br />

were randomized to cabo vs placebo, and pts with PD discontinued. Results:<br />

Enrollment to this cohort is complete (n � 60); all pts are unblinded.<br />

Baseline characteristics: median age 67 years; adenocarcinoma 72% and<br />

squamous cell 28%; 6 pts with known EGFR mutation (<strong>of</strong> 28 tested), all<br />

having received prior erlotinib; bone metastases 20%; median prior lines <strong>of</strong><br />

therapy 3 (range 0 - 6); prior exposure to anti-EGFR therapy 50%, prior<br />

exposure to anti-VEGF pathway therapy 32%. Median follow-up was 2<br />

months (range 0.4 - 22.3 months). At Wk 12, ORR per RECIST was 10%<br />

and overall disease control rate (PR�SD) was 40%. Objective tumor<br />

regression was observed in 30/47 pts (64%) with post-baseline tumor<br />

assessments, some <strong>of</strong> whom had known driver mutations in KRAS (3 pts) or<br />

EGFR (4 pts). 24 pts (40%) completed Lead-in stage with 15 randomized<br />

to continue cabo (N � 8) or to placebo (N � 7). No differences with respect<br />

to PFS were observed between treatment arms in the randomized phase <strong>of</strong><br />

the study. Median PFS from study day 1 for all pts was 4.2 months. Most<br />

common Grade 3/4 AEs were fatigue (13%), Palmar-plantar erythrodyesthesia<br />

(8%), diarrhea (7%) and asthenia (7%); one related Grade 5 AE <strong>of</strong><br />

hemorrhage was reported during Lead-in stage. Conclusions: Cabo treatment<br />

demonstrates activity in heavily pretreated metastatic NSCLC pts<br />

with 4.2 months median PFS, 10% RECIST response, and 64% rate <strong>of</strong><br />

objective tumor regression. The safety pr<strong>of</strong>ile <strong>of</strong> cabo was comparable to<br />

that seen with other VEGFR TKIs. Future studies are warranted in NSCLC.<br />

Lung Cancer—Non-small Cell Metastatic<br />

483s<br />

7513 Poster Discussion Session (Board #3), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

CALGB 30704: A randomized phase II study to assess the efficacy <strong>of</strong><br />

pemetrexed or sunitinib or pemetrexed plus sunitinib in the second-line<br />

treatment <strong>of</strong> advanced non-small cell lung cancer (NSCLC). Presenting<br />

Author: Rebecca Suk Heist, Massachusetts General Hospital Cancer<br />

Center, Boston, MA<br />

Background: Second line chemotherapy improves survival modestly and<br />

new strategies are needed. This trial was designed to evaluate the paradigm<br />

<strong>of</strong> an anti-VEGF strategy with or without standard chemotherapy in<br />

previously treated NSCLC. Methods: Patients with Stage IIIB/IV NSCLC, PS<br />

0-1 progressive after first-line chemotherapy were eligible for randomization<br />

to P (pemetrexed 500 mg/m2 d1), S (sunitinib 37.5 mg d1-21), or<br />

P�S (pemetrexed 500 mg/m2 d1 � sunitinib 37.5 mg d1-21). Pts were<br />

stratified by PS (0/1), stage (IIIB/IV), and gender (M/F). Primary objective<br />

was 18-week PFS rate; secondary objectives were response, OS, and<br />

toxicity. Target accrual was 225. The study was terminated early due to<br />

decreasing accrual rates. Results: Between 4/08 and 9/11, 130 pts<br />

registered; 128 pts treated (P�41, S�46, P�S�41). Median follow-up<br />

99 weeks. Baseline characteristics in the three arms (P/S/P�S) were well<br />

balanced: Male (54%/54%/54%); PS 0 (32%/35%/34%); Stage IV (88%/<br />

83%/93%). Histology was predominantly adenocarcinoma (66%/57%/<br />

66%); squamous was allowed (10%/15%/15%). Toxicity was higher in the<br />

S-containing arms: Grade 3/4/5 hematologic toxicity: P 5/0/0 (12%), S<br />

8/1/0 (21%), P�S 5/9/0 (36%); Grade 3/4/5 non-hematologic toxicity<br />

(excluding disease related deaths): P 6/2/0 (20%), S 21/3/1 (58%), P�S<br />

21/3/1 (63%). The 18-week PFS rate in the three arms was: P 51%<br />

(38-69), S 36% (24-54), P�S 47% (34-65). There is an overall statistically<br />

significant difference in OS between the three arms (2-sided<br />

p�0.0179) with HR 0.65 (95%CI: 0.38-1.13) for P/S; HR 0.47 (95%CI:<br />

0.27- 0.82) for P/P�S. Median OS was 10.5 mo (8.3-22.5) for P, 7.0 mo<br />

(6.0-13.0) for S, 6.7 (4.1-10.4) mo for P�S. Median PFS was 4.4 mo<br />

(1.7-8.8) for P, 3.3 mo (2.7-4.3) for S, 3.7 mo (2.5-4.3) for P�S(p�0.3).<br />

Fewer patients in the P�S arm received any subsequent therapy (29%)<br />

than either P (54%) or S (57%). Conclusions: Pemetrexed had a superior<br />

toxicity pr<strong>of</strong>ile to either sunitinib or the combination <strong>of</strong> pemetrexed and<br />

sunitinib. OS was significantly better with P alone compared to the two<br />

S-containing arms, with P�S performing worst for OS.<br />

7515 Poster Discussion Session (Board #5), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Randomized phase III trial <strong>of</strong> S-1 plus cisplatin versus docetaxel plus<br />

cisplatin for advanced non-small-cell lung cancer (TCOG0701). Presenting<br />

Author: Nobuyuki Katakami, Institute <strong>of</strong> Biomedical Research and Innovation,<br />

Kobe, Japan<br />

Background: Although molecularly targeted therapy improves outcome <strong>of</strong><br />

selected patients with advanced non-small-cell lung cancer (NSCLC), most<br />

<strong>of</strong> the patients ultimately become candidates <strong>of</strong> cytotoxic chemotherapy,<br />

which is the cornerstone <strong>of</strong> patient management. S-1 plus cisplatin (SP)<br />

has shown activity and good tolerability in phase II settings. Docetaxel plus<br />

cisplatin (DP) is the only third-generation regimen that demonstrated<br />

statistically significant improvement <strong>of</strong> overall survival and quality <strong>of</strong> life by<br />

head to head comparison with a second-generation regimen, vindesine plus<br />

cisplatin, in patients with advanced NSCLC. Methods: Patients with<br />

previously untreated stage IIIB or IV NSCLC, an ECOG PS <strong>of</strong> 0-1 and<br />

adequate organ functions were randomized to receive either oral S-1 80<br />

mg/m2 /day (40 mg/m2 b.i.d.) on days 1 to 21 plus cisplatin 60 mg/m2 on<br />

day 8 every 5 weeks or docetaxel 60mg/m2 on day 1 plus cisplatin 80<br />

mg/m2 on day 1 every 3 weeks, both up to 6 cycles. The primary endpoint is<br />

overall survival (OS). Non-inferiority study design was employed as upper<br />

confidence interval (CI) limit for HR�1.322. Secondary endpoints include<br />

progression-free survival (PFS), response, safety, and quality <strong>of</strong> life (QOL).<br />

Results: From April 2007 to December 2008, 608 patients from 66 sites in<br />

Japan were randomized to SP (n�303) or DP (n�305). Patient demographics<br />

were well balanced between the two groups. Two interim analyses were<br />

preplanned. At the final analysis, total <strong>of</strong> 480 death events were observed.<br />

The primary endpoint was met. OS for SP was non inferior to DP (median<br />

survival, 16.1 v 17.1 months, respectively; HR�1.013; 96.4% CI,<br />

0.837-1.227). PFS was 4.9 months in the SP arm and 5.2 months in the<br />

DP arm. Statistically significantly lower rate <strong>of</strong> febrile neutropenia (7.4% v<br />

1.0%), grade 3/4 neutropenia (73.4% v 22.9%), grade 3/4 infection<br />

(14.5% v 5.3%), grade 1/2 alopecia (59.3% v 12.3%) were observed in the<br />

SP arm than in the DP arm. QOL data investigated by EORTC QLQ-C30 and<br />

LC-13 favored for the SP arm. Conclusions: S-1 plus cisplatin is a standard<br />

first-line chemotherapy regimen for advanced NSCLC.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


484s Lung Cancer—Non-small Cell Metastatic<br />

7516 Poster Discussion Session (Board #6), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Three-arm randomized phase II study <strong>of</strong> carboplatin (C) and paclitaxel (P)<br />

in combination with cetuximab (CET), IMC-A12, or both for advanced<br />

non-small cell lung cancer (NSCLC) patients who will not receive bevacizumab-based<br />

therapy: An Eastern Cooperative Oncology Group (ECOG)<br />

study (E4508). Presenting Author: Nasser H. Hanna, Indiana University,<br />

Indianapolis, IN<br />

Background: Pre-clinical evidence supports the clinical investigation <strong>of</strong><br />

inhibitors to the insulin-like growth factor receptor (IGFR) and the epidermal<br />

growth factor receptor (EGFR) alone and in combination with one<br />

another in patients (pts) with NSCLC. Methods: Pts w/ chemotherapy-naïve,<br />

advanced NSCLC, not eligible for bevacizumab-based therapy, ECOG PS<br />

0/1 were eligible. Pts with uncontrolled or untreated brain mets and/or<br />

severe hyperglycemia were ineligible. Pts were randomized to receive: C<br />

AUC6iv� P 200 mg/m2 iv on day 1 every 3 wks combined with either CET<br />

iv weekly (arm A), IMC-A12 iv every 2 wks (arm B), or both (arm C). Patients<br />

with non-progressive disease (PD) after 12 weeks <strong>of</strong> therapy were permitted<br />

to continue on maintenance antibody therapy until PD. The primary<br />

objective was PFS. Secondary objectives included OS and toxicity. The<br />

design required 180 eligible patients and had an 88% power to detect a<br />

60% increase in median PFS for either comparison (arm A vs C or arm B vs<br />

C) (1-sided 0.10 level test). Results: Characteristics for arms A (n�39)/B<br />

(n�42)/C (n�48): males were 51%/57%/54%; median age was 60, 62,<br />

60; PS 0 49%/52%/60%; stage IV 87%/83%/94%; adenocarcinoma<br />

44%/26%/42%. The study was closed prematurely due to safety concerns<br />

after 129 eligible pts were treated. 13 patients died during treatment<br />

(A�6; B�2; C�5), including 9 w/in ~1 mo <strong>of</strong> starting therapy. G3-5<br />

toxicities, all attributions, A/B/C: anemia 16%/7%/13%; neutropenia<br />

27%/29%/42%; febrile neutropenia 7%/5%/4%; thrombocytopenia 7%/<br />

10%/13%; pneumonia 9%/2%/6%; hyperglycemia 2%/10%/15%; rash<br />

5%/2%/8%; hyponatremia 2%/5%/17%; thromboembolic events 2%/10%/<br />

0%; fatigue 16%;22%/13%. The estimated median PFS and OS for arms<br />

A/B/C were 3.4, 4.3, and 4.1 mos and 11.7, 8.6, and 8.4 mos,<br />

respectively. Conclusions: Based upon apparent lack <strong>of</strong> efficacy and<br />

excessive premature deaths, this study does not support the continued<br />

investigation <strong>of</strong> C � T � IMC-A12 alone or in combination with CET in pts<br />

with advanced NSCLC not eligible for bevacizumab.<br />

7518 Poster Discussion Session (Board #8), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Randomized phase II trial <strong>of</strong> erlotinib (E) plus high-dose celecoxib (HD-C)<br />

or placebo (P) in advanced non-small cell lung cancer. Presenting Author:<br />

Karen L. Reckamp, City <strong>of</strong> Hope, Duarte, CA<br />

Background: Cyclooxygenase-2 (COX-2)-dependent signalling represents a<br />

potential mechanism <strong>of</strong> resistance to epidermal growth factor receptor<br />

(EGFR) tyrosine kinase inhibitor (TKI) therapy in NSCLC. This is mediated<br />

in part through an EGFR-independent activation <strong>of</strong> MAPK/Erk by the COX-2<br />

metabolite PGE2. In addition, PGE2 promotes downregulation <strong>of</strong> Ecadherin<br />

and epithelial to mesenchymal transition. We hypothesize that<br />

EGFR and COX-2 inhibition with E and HD-C will augment EGFR TKI<br />

efficacy by increasing tumor E-cadherin expression and blocking PGE2mediated<br />

resistance to EGFR inhibition. Methods: Pts with stage IIIB/IV<br />

NSCLC who progressed following at least one line <strong>of</strong> therapy or refused<br />

standard chemotherapy were randomized to E (150mg/day)/HD-C<br />

(600mg/2x day) vs E/P in a 28-day cycle. Pts were stratified by smoking<br />

status and ECOG PS. The primary endpoint was PFS with 80% power to<br />

detect a 50% improvement; assessments were performed every 2 cycles.<br />

Secondary endpoints included response rate, OS and evaluation <strong>of</strong> molecular<br />

markers in tissue and serum to assess targeting COX-2-related pathways<br />

and evaluate EGFR TKI-resistance. All pts were required to have pretreatment<br />

tissue available. Results: 107 pts were enrolled with comparable<br />

baseline characteristics in both arms. Disease control rate (DCR) was<br />

similar in both arms, and a trend toward improved PFS was seen in the<br />

E/HD-C arm with HR 0.81 (see Table). Analysis <strong>of</strong> those with EGFR wild<br />

type revealed a significantly increased PFS while those with EGFR mutation<br />

had similar PFS in both groups. Safety analysis showed similar toxicity in<br />

both arms. Additional biomarker correlations will be presented. Conclusions:<br />

The combination <strong>of</strong> E/C in metastatic NSCLC with HD-C is well tolerated<br />

and demonstrates significantly improved efficacy in EGFR wt population.<br />

This warrants further study into the COX-2-dependent mechanisms <strong>of</strong><br />

primary resistance to EGFR TKI therapy. Supported by NIH 1P50 CA90388,<br />

K12 CA01727 and medical research funds from the Dept <strong>of</strong> Veterans’<br />

Affairs.<br />

E/HD-C (n�54) E/P (n�53) p value<br />

PFS (mo) 5.4 2.9 0.31<br />

PFS (EGFR mut, mo) 10.8 9.2 0.73<br />

PFS (EGFR wt, mo) 3.6 1.8 0.045<br />

PFS (EGFR na, mo) 2.7 2.7 0.96<br />

DCR (%) 59 55 0.70<br />

7517 Poster Discussion Session (Board #7), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

SWOG S0635 and S0636: Phase II trials in advanced-stage NSCLC <strong>of</strong><br />

erlotinib (OSI-774) and bevacizumab in bronchioloalveolar carcinoma<br />

(BAC) and adenocarcinoma with BAC features (adenoBAC), and in neversmokers<br />

with primary NSCLC adenocarcinoma (adenoCa). Presenting<br />

Author: Howard Jack West, Swedish Cancer Institute, Seattle, WA<br />

Background: Despite recent changes to lung adenoCa pathologic classification,<br />

adv stage BAC remains a definable and clinically applicable entity.<br />

Patients (pts) with BAC, as well as never-smoking (NS) pts with adv lung<br />

adenoCa, have emerged as relevant clinical subpopulations with a high<br />

probability <strong>of</strong> clinical benefit from epidermal growth factor receptor (EGFR)<br />

tyrosine kinase inhibitors (TKIs), likely related to the high proportion <strong>of</strong><br />

activating mutations in the EGFR gene in such pts. Based on these results<br />

and the potential for increased clinical activity conferred by addition <strong>of</strong> B to<br />

E, SWOG initiated a pair <strong>of</strong> phase II trials <strong>of</strong> this combination in pts with<br />

adv BAC (S0635) or NS pts with adv lung adenoCa (S0636). Methods: NS<br />

pts with BAC or adenoBAC were preferentially enrolled on S0636. A total <strong>of</strong><br />

78 and 85 eligible pts were enrolled and treated on the S0635 and S0636<br />

trials, respectively. Patients received E 150 mg PO daily and B 15 mg/kg IV<br />

q21 days until progression or prohibitive toxicity. Tumor tissue submission<br />

for pathologic review and assessment <strong>of</strong> molecular markers was mandated.<br />

Results: Pt demographics <strong>of</strong> the two trials are as shown in the table below.<br />

RECIST response rate among 61 BAC pts on S0635 with measurable<br />

disease was 18%, and among 53 NS pts on S0636, it was 47%. Median<br />

progression-free survival is 5 and 8 months (mo) on S0635 and S0636,<br />

respectively; median overall survival (OS) is 17 and 26 mo on S0635 and<br />

S0636, respectively. Toxicity consisted primarily <strong>of</strong> rash, diarrhea, and<br />

hypertension; no treatment-related deaths were reported. Molecular marker<br />

studies will be presented separately. Conclusions: In populations selected<br />

by clinical parameters, E withB is associated with modest toxicity and<br />

encouraging clinical efficacy that exceeded the prespecified OS threshold<br />

<strong>of</strong> 16 mo in studies <strong>of</strong> both adv BAC and NS pts, exceeding two years in NS<br />

pts.<br />

Trial Median age (range) Treatment naïve Performance status 0/1 Race<br />

S0635 (BAC) 69.2 (39.1-92.8) 12% 96% 92% White<br />

8% Black<br />

S0636 (NS) 61.3 (31.2-84.8) 13% 97% 66% White<br />

25% Asian<br />

5% Black<br />

4% Other<br />

7519^ Poster Discussion Session (Board #9), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

A randomized placebo-controlled phase III study <strong>of</strong> intercalated erlotinib<br />

with gemcitabine/platinum in first-line advanced non-small cell lung<br />

cancer (NSCLC): FASTACT-II. Presenting Author: Tony Mok, The Chinese<br />

University <strong>of</strong> Hong Kong, Hong Kong, China<br />

Background: FASTACT, a randomized, phase 2 study in advanced NSCLC,<br />

found that intercalated erlotinib with 1st-line platinum-based chemotherapy<br />

(CT) significantly prolongs progression-free survival (PFS) (HR<br />

0.47, p�0.0002) versus CT alone (Mok et al. JCO 2009). FASTACT-II is a<br />

confirmatory, randomized, phase 3, placebo-controlled, double-blind study<br />

in a large patient population Methods: Patients with untreated stage IIIB/IV<br />

NSCLC and ECOG PS 0/1 were randomized (1:1) to receive up to 6 cycles <strong>of</strong><br />

gemcitabine (1,250 mg/m2 on d1 and 8) plus platinum (carboplatin<br />

5�AUC or cisplatin 75 mg/m2 on d1) q4w, with either intercalated<br />

erlotinib (150 mg/day on d15–28) or placebo. Non-progressing patients<br />

received maintenance erlotinib or placebo until progression, unacceptable<br />

toxicity or death. Stratification was by disease stage, histology, smoking<br />

status and CT regimen. Primary endpoint was PFS; secondary endpoints<br />

included overall response rate (ORR), overall survival (OS), safety, QoL and<br />

biomarker analyses. Results: From Apr 2009 to Sep 2010, 451 patients<br />

were randomized to receive erlotinib (n�226) or placebo (n�225).<br />

Baseline demographics were well balanced across the arms; overall, 49%<br />

were never smokers, 40% were female and 76% had adenocarcinoma. PFS<br />

was significantly prolonged with erlotinib vs placebo: median 7.6 vs 6.0<br />

months, respectively; HR 0.57 (95% CI 0.46–0.70); p�0.0001. ORR was<br />

significantly improved with erlotinib vs placebo: 42.9% vs 17.8%;<br />

p�0.0001. A non-significant trend towards longer OS was seen with<br />

erlotinib vs placebo (median 18.3 vs 14.9 months; HR 0.78 [95% CI<br />

0.60–1.02]; p�0.069); 73% <strong>of</strong> placebo patients received a 2nd-line<br />

EGFR TKI. Except for skin rash with erlotinib, there was no clinically<br />

significant difference in toxicity between arms. A total <strong>of</strong> 283 patients<br />

(62.7%) provided samples for biomarker analyses, including EGFR mutation<br />

status, results <strong>of</strong> which will be presented. Conclusions: Intercalation <strong>of</strong><br />

erlotinib with CT significantly prolonged PFS in 1st-line advanced NSCLC.<br />

Biomarker analysis will determine the significance <strong>of</strong> this regimen in<br />

patients with or without activating EGFR mutations.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


7520 Poster Discussion Session (Board #10), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Overall survival (OS) results from OPTIMAL (CTONG0802), a phase III trial<br />

<strong>of</strong> erlotinib (E) versus carboplatin plus gemcitabine (GC) as first-line<br />

treatment for Chinese patients with EGFR mutation-positive advanced<br />

non-small cell lung cancer (NSCLC). Presenting Author: Caicun Zhou,<br />

Shanghai Pulmonary Hospital, Tongji University School <strong>of</strong> Medicine,<br />

Shanghai, China<br />

Background: The OPTIMAL study demonstrated significant superiority for E<br />

versus GC in terms <strong>of</strong> progression-free survival (PFS), objective response<br />

rate, tolerability and quality <strong>of</strong> life (QoL) in first-line advanced NSCLC<br />

patients with EGFR activating mutations (Act Mut�). Here we report OS<br />

data from OPTIMAL (<strong>Clinical</strong>Trials.gov NCT00874419). Methods: Chemotherapy-naive<br />

Chinese patients with advanced NSCLC and EGFR Act<br />

Mut�, ECOG performance status (PS) 0–2 and measurable disease were<br />

randomized to E (150 mg/day), or GC, and stratified by histology, smoking<br />

status and mutation type. OS at final data cut-<strong>of</strong>f (15 Nov 2011) was<br />

evaluated for the entire intent-to-treat (ITT) population. Subgroup analysis<br />

<strong>of</strong> OS by gender, histology, smoking status, PS, presence <strong>of</strong> skin rash and<br />

type <strong>of</strong> mutation was performed. Details <strong>of</strong> second- or later-line therapy<br />

were also documented for each patient. Results: A total <strong>of</strong> 165 patients<br />

were randomized to treatment and 154 patients received at least one dose<br />

<strong>of</strong> study drug (ITT population; E, n�82; GC, n�72). A total <strong>of</strong> 7 patients<br />

are still responding to erlotinib in the E arm. Post-study therapy included<br />

chemotherapy (doublet, n�38, or mono, n�8), or experimental drugs in<br />

clinical trials (n�10) in the E arm, and EGFR tyrosine kinase inhibitor (TKI)<br />

therapy (n�49) or chemotherapy (n�7) in the GC arm. Post-study<br />

treatment was not received by 26 and 16 patients in the E and GC arms,<br />

respectively. A total <strong>of</strong> 84 deaths were reported (E, n�47; GC, n�37). OS<br />

did not differ significantly between the two treatment arms (HR�1.065,<br />

p�0.6849), and no significant difference in OS was observed in the<br />

different subgroups. Conclusions: The lack <strong>of</strong> a statistically significant<br />

difference in OS in the OPTIMAL study was possibly due to a high level <strong>of</strong><br />

cross-over to EGFR TKI therapy in the GC arm. However, the significant<br />

benefits reported with E in terms <strong>of</strong> PFS, QoL and tolerability in this study<br />

suggest that E should be considered as one <strong>of</strong> the standard first-line<br />

treatments for patients with advanced EGFR Act Mut� NSCLC.<br />

7522 Poster Discussion Session (Board #12), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

EGFR compound mutants and survival on erlotinib in non-small cell lung<br />

cancer (NSCLC) patients (p) in the EURTAC study. Presenting Author:<br />

Rafael Rosell, Catalan Institute <strong>of</strong> Oncology, Barcelona, Spain; Pangaea<br />

Biotech, USP Institut Universitari Dexeus, Barcelona, Spain<br />

Background: When EGFR compound mutants (exon 19 deletions or L858R<br />

plusT790M) are present at the time <strong>of</strong> clinical resistance to erlotinib,<br />

patients attain longer overall survival (OS) (Oxnard et al. CCR 2011). The<br />

H1975 cell line, harboring L858R plus T790M prior to treatment, is<br />

sensitive to gefitinib (Sordella et al. Science 2004; Faber et al. Cancer<br />

Discovery 2011), though the compound mutant may become dominant<br />

after multiple passages in vitro, leading to resistance (Pao et al. PLoS Med<br />

2005). Methods: The EURTAC trial (clinicaltrials.gov NCT00446225)<br />

randomized 174 p with EGFR exon 19 deletions or L858R mutations to<br />

receive erlotinib or chemotherapy. Progression-free survival (PFS) was 9.7<br />

months (m) vs 5.2 m, respectively (P�0.0001). No differences in OS were<br />

observed.123 p with remaining available pre-treatment tumor tissue were<br />

re-analyzed for the concomitant presence <strong>of</strong> the T790M mutation with an<br />

allelic discrimination Taqman assay in the presence <strong>of</strong> a PNA clamp<br />

designed to inhibit the amplification <strong>of</strong> the exon 20 wild-type (wt) allele.<br />

This assay is capable <strong>of</strong> detecting the T790M allele at a ratio <strong>of</strong> 1:5000 wt<br />

alleles. Results: The T790M mutation was detected in 21/64 (32.8%) p in<br />

the erlotinib arm and 26/59 (44.1%) in the chemotherapy arm. PFS was<br />

12.1 m for p with mutant T790M in the erlotinib arm, 8.8 m for p with wt<br />

T790M in the erlotinib arm, 6.3 m for p with mutant T790M in the<br />

chemotherapy arm, and 4.5 m for p with wt T790M in the chemotherapy<br />

arm (P�0.0001). OS was not reached for p with mutant T790M in the<br />

erlotinib arm, 16.1 m for p with wt T790M in the erlotinib arm, 22.6 m for<br />

p with mutant T790M in the chemotherapy arm, and 18.4 m for p with wt<br />

T790M in the chemotherapy arm (P�0.04). Conclusions: Our unexpected<br />

finding that p with EGFR compound mutants attain the maximum benefit<br />

from erlotinib suggests a need for more sensitive assays to detect EGFR<br />

compound mutants and for studies <strong>of</strong> inhibitors targeting the EGFR T790M<br />

mutation. Whole genome sequencing may provide greater understanding <strong>of</strong><br />

the genetic factors involved in the differences in OS.<br />

Lung Cancer—Non-small Cell Metastatic<br />

485s<br />

7521 Poster Discussion Session (Board #11), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Updated overall survival results <strong>of</strong> WJTOG 3405, a randomized phase III<br />

trial comparing gefitinib (G) with cisplatin plus docetaxel (CD) as the<br />

first-line treatment for patients with non-small cell lung cancer harboring<br />

mutations <strong>of</strong> the epidermal growth factor receptor (EGFR). Presenting<br />

Author: Tetsuya Mitsudomi, Department <strong>of</strong> Thoracic Surgery, Aichi Cancer<br />

Center Hospital, Nagoya, Japan<br />

Background: WJTOG3405 met its primary endpoint <strong>of</strong> progression free<br />

survival (PFS) (9.2 months (mo.) for G vs. 6.3 mo. for CD, hazard ratio (HR)<br />

0.489, 95% confidence interval (CI): 0.336-0.710). (Mitsudomi et al.,<br />

Lancet Oncol., 2010). However, the impact on overall survival (OS) was not<br />

clear then because <strong>of</strong> relatively short follow-up period. Methods: Overall<br />

survival (OS) was re-evaluated using updated data (data cut<strong>of</strong>f, 31 July,<br />

2011, median follow-up, 34 months) for 172 patients. Results: Eighty-two<br />

events had occurred (48%). Median survival time (MST) for G arm was 36<br />

mo. (95% CI: 26.3 -) which was not significantly different from 39 mo.<br />

(95% CI: 31.2 -) for CD arm (HR 1.185, 95% CI 0.767-1.829).<br />

Multivariate analysis using Cox proportional hazards model revealed that<br />

none <strong>of</strong> covariates (treatment arm, smoking status, sex, age, postoperative<br />

recurrence or IIIB/IV, and mutation type) significantly affected OS. In the G<br />

arm, MST <strong>of</strong> patients with exon 19 deletion (36 mo.) was comparable to<br />

that <strong>of</strong> patients with L858R (35 mo.). In the CD arm, 78 patients (91%)<br />

received EGFR-TKI as the 2nd or later line treatment, whereas in the G arm,<br />

52 patients (61%) received platinum doublet. Accordingly, 130 patients<br />

received both platinum doublet and EGFR-tyrosine kinase inhibitor (TKI)<br />

and 34 patients received EGFR-TKI without platinum doublet in their<br />

whole courses <strong>of</strong> therapy. MST for the former and the latter group were 36<br />

months (95% CI: 31.2-45.7) and 45 months (95% CI: 25.6-), without<br />

significant difference. Conclusions: This update OS analysis revealed that G<br />

for advanced NSCLC with EGFR mutation <strong>of</strong>fers distinct survival benefit <strong>of</strong><br />

3 years. There was no difference in OS whether the first-line treatment was<br />

G or CD, in accordance with the precedent studies. The reason why PFS<br />

difference was not translated into OS difference is probably due to high<br />

cross over rate to EGFR-TKI. However, it was noteworthy that 40% <strong>of</strong><br />

patients in the G arm could be managed without platinum doublet and yet<br />

had similar outcome.<br />

7523 Poster Discussion Session (Board #13), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Sensitivity to EGFR inhibitors based on location <strong>of</strong> EGFR exon 20 insertion<br />

mutations within the tyrosine kinase domain <strong>of</strong> EGFR. Presenting Author:<br />

Daniel Botelho Costa, Beth Israel Deaconess Medical Center/Harvard<br />

Medical School, Boston, MA<br />

Background: Epidermal growth factor receptor (EGFR) mutations (M) define<br />

an important subgroup <strong>of</strong> non-small-cell lung cancer (NSCLC). Most<br />

patients whose tumors harbor exon 19 deletions or L858R EGFR M have<br />

responses to reversible ATP-mimetic EGFR tyrosine kinase inhibitors<br />

(TKIs), gefitinib and erlotinib. Exon 20 insertion M comprise ~5% <strong>of</strong> EGFR<br />

M, occur at the N-lobe <strong>of</strong> EGFR after its C-helix (AA M766), and nearly all<br />

NSCLCs with EGFR exon 20 insertion M display lack <strong>of</strong> responses to EGFR<br />

TKIs (Yasuda H. Lancet Oncol 2011). Methods: We have 1) compiled<br />

genotype-clinical outcomes <strong>of</strong> EGFR exon 20 insertion M NSCLCs to EGFR<br />

TKIs, 2) generated a comprehensive panel <strong>of</strong> exon 20 EGFR M constructs<br />

using site-directed mutagenesis and introduced them into Ba/F3 cells for in<br />

vitro analysis, and 3) compared NSCLC cell lines with EGFR M to a novel<br />

malignant pleural effusion-derived cell line. Results: The disease control<br />

rate <strong>of</strong> gefitinib or erlotinib was significantly higher in EGFR exon 20<br />

insertion M located within the C-helix (3/3,100%) when compared to M<br />

following the C-helix (1/14, 7%; p�0.00059). The NSCLC with EGFR-<br />

A763_Y764insFQEA (located within the C-helix <strong>of</strong> EGFR) achieved a<br />

partial response to erlotinib that lasted 18 months. Most other exon 20<br />

insertion M-positive NSCLCs did not respond (p�0.07). Eight representative<br />

exon 20 insertion M were studied (including EGFR-<br />

A763_Y764insFQEA, Y764_S765insHH, A767_V769dupASV,<br />

D770_N771insNPG, H773_V774insH). All, but A763_Y764insFQEA,<br />

were resistant to micromolar concentrations (C) <strong>of</strong> EGFR TKIs. Ba/F3 cells<br />

with EGFR-A763_Y764insFQEA underwent apoptosis upon exposure to<br />

nanomolar C <strong>of</strong> erlotinib. A patient-derived cell line with EGFR-<br />

A763_Y764insFQEA had phosphorylated EGFR, ERK and AKT inhibited by<br />

nanomolar C <strong>of</strong> erlotinib. Conclusions: Not all EGFR exon 20 insertion<br />

mutations are resistant to EGFR TKIs, and in specific EGFR-<br />

A763_Y764insFQEA is an EGFR TKI-sensitive M. This finding has clinical<br />

implications for the care <strong>of</strong> the 10,000 cases <strong>of</strong> EGFR exon 20 insertion M<br />

NSCLC diagnosed yearly and points towards the need to define the<br />

molecular mechanisms that underlie differential responses to EGFR TKIs.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


486s Lung Cancer—Non-small Cell Metastatic<br />

7524 Poster Discussion Session (Board #14), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Chemotherapy with erlotinib or chemotherapy alone in advanced NSCLC<br />

with acquired resistance to EGFR tyrosine kinase inhibitors (TKI). Presenting<br />

Author: Sarah B. Goldberg, Massachusetts General Hospital Cancer<br />

Center, Boston, MA<br />

Background: EGFR-mutant NSCLC has an oncogene-addicted phenotype<br />

that confers sensitivity to EGFR TKIs. Prior data suggests EGFR addiction<br />

persists after development <strong>of</strong> TKI resistance, leading many clinicians to<br />

continue TKI along with chemotherapy (chemo); however, this strategy has<br />

not been formally evaluated. Methods: An institutional database was<br />

reviewed to identify patients (pts) with advanced NSCLC and acquired<br />

resistance to EGFR TKIs by Jackman criteria. Pts were included if they<br />

subsequently received chemo. Objective response rate (RR) to chemo/<br />

erlotinib or chemo alone was assessed by blinded radiographic review and<br />

compared by Fisher’s exact test and multivariable logistic regression.<br />

Progression-free survival (PFS) and overall survival (OS) from TKI failure<br />

(defined as chemo initiation date) were compared by log-rank test and<br />

multivariable Cox analysis. Results: 78 pts were eligible (34 chemo/<br />

erlotinib, 44 chemo alone). 70 pts (90%) had a documented EGFR<br />

mutation and were on TKI for a median <strong>of</strong> 15 months (range 4-51); in the 8<br />

pts with unknown genotype the median duration on TKI was 11 months<br />

(range 5-16). Baseline characteristics were well balanced except more pts<br />

received erlotinib as initial TKI in the chemo/erlotinib group. RR was<br />

evaluable in 57 pts and was higher with chemo/erlotinib compared to<br />

chemo alone (41% vs 18%; OR 0.31, 95% CI 0.09, 1.04; p�0.08). RR<br />

adjusted for chemo regimen and time to TKI failure yielded OR 0.20 (95%<br />

CI 0.05, 0.78; p�0.02), favoring chemo/erlotinib. Median PFS was 4.4<br />

months in the chemo/erlotinib group and 4.2 months in the chemo alone<br />

group (crude HR�0.84, 95% CI 0.52, 1.38; p�0.50; adjusted HR 0.79,<br />

95% CI 0.48, 1.29; p�0.34). There was no significant difference in OS in<br />

the crude or adjusted analyses. Conclusions: This is the first study, to our<br />

knowledge, to show that continuation <strong>of</strong> an EGFR TKI with chemo<br />

compared to chemo alone significantly increases the RR in pts with<br />

advanced NSCLC and acquired TKI resistance, though we did not observe<br />

an impact on PFS or OS. Continued concurrent TKI may be a valuable<br />

strategy, particularly for pts with symptomatic progression, when a higher<br />

response rate may be beneficial. Further study is warranted.<br />

7526 Poster Discussion Session (Board #16), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Continuation <strong>of</strong> EGFR/ALK inhibition after local therapy <strong>of</strong> oligoprogressive<br />

disease in EGFR mutant (Mt) and ALK� non-small cell lung cancer (NSCLC).<br />

Presenting Author: Andrew James Weickhardt, University <strong>of</strong> Colorado Cancer<br />

Center, Denver, CO<br />

Background: This study aimed to investigate the benefits <strong>of</strong> local treatment <strong>of</strong><br />

limited disease progression and continuation <strong>of</strong> crizotinib (C) or EGFR-TKI in<br />

patients with ALK� or EGFR-Mt metastatic NSCLC, respectively. Methods:<br />

Patients with advanced ALK� NSCLC treated with C (n�38) and EGFR-Mt<br />

NSCLC treated with EGFR TKIs (erlotinib or gefitinib) (n�27) were identified.<br />

Patients were evaluated for initial response, site <strong>of</strong> first progression (CNS or extra<br />

CNS (eCNS)) and median progression free survival (PFS1). A subset <strong>of</strong> patients<br />

with limited sites <strong>of</strong> progression (oligoprogressive disease) suitable for local<br />

therapy received either radiation or surgery to these sites and continued on the<br />

same TKI. The subsequent pattern <strong>of</strong> progression and median progression free<br />

survival from the time <strong>of</strong> first progression (PFS2) were recorded. Results: In 38<br />

ALK� patients, PFS1�9.0 months on C. In 27 EGFR-Mt patients, PFS1�13.8<br />

months on EGFR-TKI. CNS was the first site <strong>of</strong> progression in 13/28 (46%) <strong>of</strong><br />

ALK� patients and 5/23 (22%) EGFR-Mt patients. 25 <strong>of</strong> 51 patients who<br />

progressed (49%) were deemed suitable for local therapy (15 ALK�, 10<br />

EGFR-Mt; 24 with radiotherapy (Body: 3 XRT, 9 SBRT; CNS: 7 WBRT, 5 SRS), 1<br />

with surgery (adrenalectomy)) and continuation <strong>of</strong> the same targeted therapy.<br />

19/25 patients progressed post local therapy, median PFS2 � 6.2 months. In<br />

patients who progressed at PFS1 only in the CNS, there was a trend to longer<br />

PFS2 than patients who progressed at PFS1 in eCNS (7.1 months vs 4.0<br />

months, p�0.26). 60% who progressed only in the CNS at PFS1, progressed<br />

eCNS at PFS2 (n�10). Conclusions: Progression <strong>of</strong> oncogene addicted NSCLC<br />

on relevant targeted therapies is <strong>of</strong>ten suitable for local therapy (oligoprogressive<br />

disease). Continuation <strong>of</strong> the targeted therapy following local therapy is<br />

associated with �6 months <strong>of</strong> additional disease control.<br />

Sites <strong>of</strong> first progression and median PFS1 and PFS2 in patients continuing TKI<br />

after local therapy <strong>of</strong> oligoprogressive disease.<br />

Site <strong>of</strong> progression Number <strong>of</strong> patients<br />

PFS1<br />

(months)<br />

PFS2<br />

(months)<br />

CNS 1st site <strong>of</strong> prog. 10 10.9 7.1<br />

eCNS 1st site <strong>of</strong> prog. 15 9.6 4.0<br />

All patients 25 10.0 6.2<br />

* Includes 3 patients who prog. eCNS and CNS at PFS1.<br />

7525 Poster Discussion Session (Board #15), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Response to EGFR tyrosine kinase inhibitor (TKI) retreatment after a<br />

drug-free interval in EGFR-mutant advanced non-small cell lung cancer<br />

(NSCLC) with acquired resistance. Presenting Author: Stephanie Heon,<br />

Dana-Farber Cancer Institute, Boston, MA<br />

Background: Patients (pts) with advanced NSCLC and sensitizing EGFR<br />

mutations who initially respond to gefitinib or erlotinib eventually develop<br />

acquired resistance to the TKIs. Anecdotal and retrospective reports<br />

suggest that EGFR-TKI resistant cancers can respond again to gefitinib or<br />

erlotinib after an interval <strong>of</strong>f the TKI. This retrospective study was<br />

undertaken to investigate the impact <strong>of</strong> EGFR-TKI retreatment after a<br />

drug-free interval in EGFR mutant NSCLC with acquired resistance to<br />

gefitinib or erlotinib. Methods: Pts with stage IV or relapsed NSCLC with<br />

sensitizing EGFR mutations and acquired resistance to EGFR-TKI seen at<br />

the DFCI/MGH between 8/00 and 8/11 who were retreated with single<br />

agent gefitinib or erlotinib after an EGFR-TKI-free interval were identified<br />

from a prospective trial. The objective tumor response (CR, PR, SD, PD)<br />

was determined using RECIST 1.1. Results: 19 pts were eligible and had<br />

adequate scans for radiographic assessments after the reinstitution <strong>of</strong> an<br />

EGFR-TKI. The response rate and median PFS to the initial course <strong>of</strong><br />

gefitinib (n�4) or erlotinib (n�15) were 16/19 (84%) and 9.8 months<br />

(95% CI, 7.8-11.3) respectively. All pts were retreated with erlotinib after<br />

1 to 4 intervening systemic regimens. The median interval from EGFR-TKI<br />

discontinuation to erlotinib retreatment was 11 months (range, 2-46). 4 <strong>of</strong><br />

the 19 pts (21%) had PD as the best response to erlotinib retreatment, 14<br />

(74%) had SD for at least 1 month, and 1 (5%) had a PR. The median PFS<br />

was 4.4 months (95% CI, 3.0-6.7). 3 pts remained on erlotinib without<br />

progression for 6 months. 3 pts had their tumors rebiopsied before (n�2) or<br />

during (n�1) erlotinib retreatment; 1 <strong>of</strong> the 3 had EGFR T790M in<br />

association with the initial sensitizing EGFR mutation, and another had a<br />

secondary PIK3CA mutation. Conclusions: Our findings suggest that erlotinib<br />

retreatment is an option for EGFR mutated NSCLC with acquired<br />

resistance to EGFR-TKI after a drug-free interval and progression on<br />

intervening therapy. Additional advanced NSCLC pts without a documented<br />

EGFR mutation who fulfill the clinical definition <strong>of</strong> acquired<br />

resistance are undergoing review to expand our cohort.<br />

7527 Poster Discussion Session (Board #17), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Local therapy as a treatment strategy in EGFR-mutant advanced lung<br />

cancers that have developed acquired resistance to EGFR tyrosine kinase<br />

inhibitors. Presenting Author: Helena Alexandra Yu, Memorial Sloan-<br />

Kettering Cancer Center, New York, NY<br />

Background: The utility <strong>of</strong> EGFR directed therapy for the treatment <strong>of</strong> EGFR<br />

mutant lung cancer is limited by the development <strong>of</strong> acquired resistance<br />

(AR) to EGFR tyrosine kinase inhibitor (TKI) therapy, which occurs after a<br />

median <strong>of</strong> 16 months (mos). There are no approved targeted therapies after<br />

disease progression on EGFR TKI therapy. Local therapy for oligometastatic<br />

disease is used with regularity in other solid tumors, and can lead to long<br />

term survival in selected individuals. EGFR mutant lung cancers with AR to<br />

TKI therapy can follow an indolent course that is amenable to local therapy<br />

to treat progression <strong>of</strong> disease when used in conjunction with continued<br />

EGFR inhibition. Outcomes following local therapy in this setting have not<br />

been assessed. Methods: Patients (pts) with AR to EGFR TKI’s who received<br />

local therapy excluding treatment <strong>of</strong> CNS metastases or local therapy prior<br />

to AR were identified in an IRB-approved prospective registry <strong>of</strong> 184 pts<br />

with AR enrolled from August 2004- November 2011. We collected<br />

treatments as well as progression free survival (PFS) and overall survival<br />

(OS) after local therapy. Results: 18 pts received local therapy. Treatments<br />

included surgical resection and radiation to lung, lymph nodes, adrenal<br />

gland or bone. Median age was 57, 56% were women, and 61% were never<br />

smokers. 14 had EGFR Exon 19 deletions, and 4 L858R. The median time<br />

to development <strong>of</strong> AR on TKI was 19 mo (range 5-33). Known mechanisms<br />

<strong>of</strong> AR included T790M (11 pts), MET amplification (1 pt) and small cell<br />

transformation (1 pt). The median PFS after local therapy was 10 mo (95%<br />

CI: 4-NA), median time from local therapy until change in systemic therapy<br />

was 22 mo (95%CI: 6 - 30), and median OS from local therapy was 41 mo<br />

(95% CI: 26-NA). Local therapy was tolerated well, with 85% <strong>of</strong> pts<br />

restarting TKI therapy within one month <strong>of</strong> completing local therapy.<br />

Conclusions: Local therapy is well tolerated and in combination with<br />

continued EGFR TKI therapy prolongs time until change in systemic<br />

therapy is required and may lead to outcomes that are superior to currently<br />

available treatment options in selected individuals.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


7528 Poster Discussion Session (Board #18), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Rebiopsy <strong>of</strong> non-small cell lung cancer patients with acquired resistance to<br />

EGFR-TKI: Comparison between T790M mutation-positive and -negative<br />

populations. Presenting Author: Akito Hata, Division <strong>of</strong> Integrated Oncology,<br />

Institute <strong>of</strong> Biomedical Research and Innovation, Kobe, Japan<br />

Background: The secondary epidermal growth factor receptor (EGFR)<br />

mutation T790M accounts for approximately half <strong>of</strong> acquired resistances to<br />

EGFR-tyrosine kinase inhibitors (TKI). A recent report has demonstrated<br />

the presence <strong>of</strong> T790M predicts a favorable prognosis and indolent<br />

progression, compared to the absence <strong>of</strong> T790M after TKI failure. However,<br />

rebiopsy to confirm T790M status can be challenging due to limited tissue<br />

availability and procedural feasibility, and little is known regarding the<br />

differences among patients with or without T790M. Methods: We investigated<br />

73 patients harboring EGFR sensitive mutations who had undergone<br />

rebiopsy to confirm the emergence <strong>of</strong> T790M after TKI failure. The peptide<br />

nucleic acid-locked nucleic acid PCR clamp method was used in EGFR<br />

mutational analyses. Patient characteristics (age, gender, smoking history,<br />

performance status, EGFR mutation site, initial TKI, response to initial TKI,<br />

line <strong>of</strong> initial TKI, progression-free survival with initial TKI, and biopsy site)<br />

and postprogression survivals (PPS) after initial TKI failure, were retrospectively<br />

compared in patients with and without T790M. Results: We identified<br />

T790M in 2 (10%) <strong>of</strong> 21 central nervous system (CNS) (19 cerebrospinal<br />

fluid and 2 brain tissue) specimens, and in 20 (38%) <strong>of</strong> 52 other lesions<br />

(25 lung tissue, 24 pleural effusion, and 3 lymph node) (p � 0.0225).<br />

Other characteristics had no statistical association with the detection <strong>of</strong><br />

T790M. Median PPS in patients with T790M was 34.0 months, and in<br />

those without T790M, 14.5 months (p � 0.0038). Although none <strong>of</strong> our<br />

patients received TKIs continuously after initial failure, 56 (77%) patients<br />

were re-administered TKIs. Regardless <strong>of</strong> T790M status, PPS in patients<br />

with TKI re-administration (23.4 months) was significantly longer than<br />

without re-administration (10.4 months) (p � 0.0085). Conclusions: The<br />

emergence <strong>of</strong> T790M in CNS is rare compared with other lesions. Patients<br />

with T790M after TKI failure have significantly better prognosis than those<br />

without T790M. The effectiveness <strong>of</strong> TKI re-administration or continuous<br />

administration beyond progression is suggested after initial TKI failure.<br />

7530 Poster Discussion Session (Board #20), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

First-line dacomitinib (PF-00299804), an irreversible pan-HER tyrosine<br />

kinase inhibitor, for patients with EGFR-mutant lung cancers. Presenting<br />

Author: Mark G. Kris, Memorial Sloan-Kettering Cancer Center, New York,<br />

NY<br />

Background: Dacomitinib irreversibly inhibits EGFR, HER2 and HER4, and<br />

showed superior activity vs. reversible EGFR tyrosine kinase inhibitors in<br />

EGFR-mutant lung cancer models, including resistant forms. This openlabel<br />

Phase II study evaluates dacomitinib as 1st-line treatment (tx) for<br />

patients (pts) with lung cancers. Pts with sensitizing EGFR deletions/<br />

mutations in exons 19 or 21 are reported here. Methods: Pts had stage<br />

IIIB/IV adenocarcinoma, no prior systemic tx, had smoked �10 pack years<br />

(none within 15 years <strong>of</strong> enrollment) or had known EGFR mutation. Pts<br />

received dacomitinib orally once daily continuously at 45 mg, or 30 mg<br />

with the option to escalate to 45 mg; evaluation was every 28 days.<br />

Endpoints included progression-free survival rate at 4 months (PFS at 4M,<br />

primary); PFS, and partial response (PR) rate. Results: 92 pts enrolled; 47<br />

had EGFR mutation in exons 19 (n�25) or 21 (n�22), 33 were female and<br />

27 Asian. 34/46 evaluable pts with EGFR exon 19 or 21 mutations had a<br />

PR (PR rate � 74%; 95% CI: 59–86; exon 19 � 72%; exon 21 � 76%).<br />

PR rates and preliminary PFS were not significantly different for exons 19<br />

and 21. Preliminary PFS at 4M was 96% (95% CI: 84–99), preliminary<br />

PFS rate at 1 year was 77% (95% CI: 61–87) and preliminary median PFS<br />

was 17 months (95% CI: 13–24). Median tx duration was 13.1 months. For<br />

pts with EGFR wild-type lung cancers, PR and PFS at 4M rates were 7%<br />

(n�14; 95% CI: 0–34) and 33% (n�14; 95% CI: 11–58), respectively,<br />

and for pts with EGFR unknown lung cancers, 46% (n�22; 95% CI:<br />

24–68) and 68% (n�24; 95% CI: 45– 83), respectively. 7 pts had lung<br />

cancers with non-sensitizing EGFR mutations; 2 had a PR and 3 SD. For all<br />

92 pts, common side effects included dermatitis acneiform (grade 3/4 �<br />

17%/0) and diarrhea (14%/0). 3/46 pts with EGFR exon 19 or 21<br />

mutations discontinued tx due to drug-related toxicity. Conclusions: 74% <strong>of</strong><br />

pts in this cohort with EGFR exon 19 or 21 mutant lung cancers<br />

experienced PRs with 1st-line dacomitinib; preliminary PFS rate was 77%<br />

at 1 year; preliminary median PFS was 17 months; further research is<br />

planned in this pt population. As dacomitinib is well tolerated, with<br />

preclinical activity against HER2, a cohort <strong>of</strong> pts with HER2 mutant lung<br />

cancers is recruiting.<br />

Lung Cancer—Non-small Cell Metastatic<br />

487s<br />

7529 Poster Discussion Session (Board #19), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Concordance <strong>of</strong> driver mutations in primary and matched metastasis from<br />

patients with non-small cell lung cancer (NSCLC) using next-generation<br />

sequencing (NGS). Presenting Author: Stéphane Vignot, Institut Gustave<br />

Roussy, INSERM U981, Villejuif, France<br />

Background: Progress in treating NSCLC continues to be limited by high<br />

recurrence rates after definitive treatment <strong>of</strong> localized disease and frequent<br />

presentation with metastatic disease. In these pts, diagnostic samples are<br />

<strong>of</strong>ten limited in size and may be inadequate for genotyping necessary in<br />

this disease. In applying genomic pr<strong>of</strong>iling to aid treatment planning for<br />

patients with metastatic NSCLC, it is critical to determine whether archived<br />

primary tumor adequately reflects the genomic spectrum <strong>of</strong> the patient’s<br />

metastatic disease or whether biopsy <strong>of</strong> a metastatic site is required. We<br />

undertook this study using NGS technology to characterize paired primarymetastatic<br />

biopsy genomic pr<strong>of</strong>iles. Methods: Primary and matched metastatic<br />

tumor pairs plus adjacent normal tissue from 15 pts with NSCLC<br />

(adenocarcinoma 9/squamous 4/large cell 2) and heavy smoking history<br />

were analyzed using a targeted NGS assay in a CLIA laboratory (Foundation<br />

Medicine). Genomic libraries were captured for 3230 exons in 182<br />

cancer-related genes plus 37 introns from 14 genes <strong>of</strong>ten rearranged in<br />

cancer and sequenced to average median depth <strong>of</strong> 778X with 99% <strong>of</strong> bases<br />

covered �100X. Results: Among the 30 tumors, 373 somatic alterations<br />

were identified; 96 were known NSCLC drivers including: TP53 (14 pts),<br />

KRAS (4 pts), SOX2 (4 pts), STK11 (2 pts), CDKN2A (2 pts) and<br />

SMARCA4 (2 pts) in full agreement with their respective histological type.<br />

In all cases (15/15), primary and metastatic tumors from the same patient<br />

demonstrated 90% driver mutation concordance (74/82, 95 % CI, 82-<br />

95%). For other mutations, the majority <strong>of</strong> which are likely passengers, the<br />

69% (201/291) concordance rate was significantly lower (p� .001).The<br />

total number <strong>of</strong> alterations between primaries (190) and metastases (183)<br />

was remarkably similar. Conclusions: These results suggest that genomic<br />

pr<strong>of</strong>iles <strong>of</strong> primary tumor can identify the key somatic alterations present in<br />

matched NSCLC metastases and therefore is a suitable specimen for<br />

clinical decision making in the majority <strong>of</strong> cases. Our data does not support<br />

the routine need for a new biopsy upon recurrence since variability in<br />

mutational status is low.<br />

7531 Poster Discussion Session (Board #21), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

A randomized discontinuation phase II trial <strong>of</strong> ridaforolimus in non-small<br />

cell lung cancer (NSCLC) patients with KRAS mutations. Presenting<br />

Author: Gregory J. Riely, Memorial Sloan-Kettering Cancer Center, New<br />

York, NY<br />

Background: Mutations in KRAS are present in ~25% <strong>of</strong> patients with<br />

advanced NSCLC. Preclinical data support the role <strong>of</strong> mammalian target <strong>of</strong><br />

rapamycin (mTOR) in KRAS mediated oncogenesis. Ridaforolimus is an<br />

inhibitor <strong>of</strong> mTOR which has been shown to have efficacy in advanced<br />

endometrial cancer and s<strong>of</strong>t tissue sarcoma. Everolimus, another mTOR<br />

inhibitor was previously evaluated in unselected patients with advanced<br />

NSCLC and found to have a response rate �5%. We hypothesized that by<br />

enrichment for patients with NSCLC and KRAS mutations, treatment with<br />

ridaforolimus would be associated with prolonged stable disease relative to<br />

available standard treatments for NSCLC. Methods: Patients with stage<br />

IIIB/IV non-small cell lung cancer with KRAS mutation who had received<br />

prior chemotherapy for NSCLC began treatment with oral ridaforolimus 40<br />

mg once daily on a 5 day/week schedule. After 8 weeks, patients with<br />

�30% tumor shrinkage remained on ridaforolimus and patients with<br />

�20% tumor growth discontinued treatment. Patients with stable disease<br />

were randomized 1:1 to placebo or ridaforolimus. The primary endpoint <strong>of</strong><br />

the study was progression-free survival (PFS) after randomization. Results:<br />

79 patients were enrolled (40 women, median age 58 [range 28-85]). The<br />

overall response rate (CR�PR) at 8 weeks was 1/79 (1%, 95% CI 0-7%).<br />

28 patients with stable disease at 8 weeks were randomized to ridaforolimus<br />

or placebo. Median PFS based on investigator assessment from<br />

randomization was significantly longer with ridaforolimus (4 months) than<br />

placebo (2 months, p�0.013, HR 0.36). Median OS from randomization<br />

was 18 months in the ridaforolimus treated arm and 5 months in the<br />

placebo treated group, (HR 0.46, p�0.09). The most common grade �3<br />

adverse events were fatigue (10%), mucositis/stomatitis (10%), pneumonia<br />

(10%), dyspnea (9%), diarrhea (6%), and hyperglycemia (6%).<br />

Conclusions: In patients with KRAS mutant NSCLC who had stable disease<br />

after 8 weeks <strong>of</strong> ridaforolimus, ridaforolimus was associated with prolonged<br />

progression-free survival. Further evaluation <strong>of</strong> ridaforolimus in this patient<br />

population is warranted.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


488s Lung Cancer—Non-small Cell Metastatic<br />

7532 Poster Discussion Session (Board #22), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Characteristics <strong>of</strong> NSCLCs harboring NRAS mutations. Presenting Author:<br />

Kadoaki Ohashi, Vanderbilt University, Nashville, TN<br />

Background: We sought to determine the frequency and clinical characteristics<br />

<strong>of</strong> patients with non-small cell lung cancers (NSCLCs) harboring NRAS<br />

mutations. We used preclinical models to identify targeted therapies likely<br />

to be <strong>of</strong> benefit against NRAS mutant lung cancer cells. Methods: We<br />

reviewed data in the Catalogue <strong>of</strong> Somatic Mutations in Cancer (COSMIC)<br />

and clinical history from patients with NSCLC whose tumors underwent<br />

systematic screening for driver mutations including NRAS. Patient characteristics<br />

examined included age, gender, race, smoking history, disease<br />

stage, treatment history, and overall survival (OS). 6 NSCLC cell lines with<br />

NRAS mutations were screened for sensitivity against multiple targeted<br />

agents. Gene expression was pr<strong>of</strong>iled using Affymetrix U133A arrays in 5<br />

NRAS mutant NSCLC cell lines, 8 with EGFR mutations and 17 with KRAS<br />

mutations. Results: Among 4524 patients with NSCLC tested, NRAS<br />

mutations were present in 29 (0.64%). The types <strong>of</strong> substitutions found<br />

were Q61H/K/L/R and G12A/C/D/R/S, with NRAS Q61L the most common<br />

(n�14; 48%). One tumor had a concurrent KRAS mutation. 83% had<br />

adenocarcinoma histology, with no significant differences in gender. While<br />

90% <strong>of</strong> patients were former or current smokers, smoking-related G:C�T:A<br />

transversions were significantly less frequent in NRAS than in KRASmutant<br />

NSCLC (KRAS: 66%, NRAS: 13%, p�0.05). Systemic chemotherapy<br />

showed limited efficacy in 7 patients with metastatic disease<br />

(median OS 7 mos). 5 <strong>of</strong> 6 NRAS mutant lung cancer cell lines were<br />

sensitive to the MEK inhibitors, AZD6244 and GSK1120212, while other<br />

targeted agents (against EGFR, ALK, MET, IGF-1R, PIK3CA, BRAF) were<br />

minimally effective. Gene expression pr<strong>of</strong>iles <strong>of</strong> NRAS mutant cell lines<br />

were distinct from those with KRAS or EGFR mutations. Conclusions: NRAS<br />

mutations define a distinct subset <strong>of</strong> NSCLCs (~1%) with potential<br />

sensitivity to MEK inhibitors. While NRAS gene mutations are more<br />

common in current/former smokers, the types <strong>of</strong> mutations are not those<br />

classically associated with smoking.<br />

7534 Poster Discussion Session (Board #24), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Native and rearranged ALK copy number and rearranged ALK cell count in<br />

NSCLC: Implications for ALK inhibitor therapy. Presenting Author: D. Ross<br />

Camidge, University <strong>of</strong> Colorado Cancer Center, Aurora, CO<br />

Background: ALK rearranged NSCLC responds well to ALK inhibitors.<br />

<strong>Clinical</strong>ly, �15% cells showing rearrangements by break-apart FISH<br />

classifies tumors as ALK(�). Native ALK copy number gain has also been<br />

reported. We explored the significance <strong>of</strong> native and rearranged ALK copy<br />

number on crizotinib outcomes and whether �15% reflected a clear<br />

biological distinction in the frequency <strong>of</strong> ALK(�) cells. Methods: Copy<br />

number and genomic status <strong>of</strong> ALK assessed by FISH. A total <strong>of</strong> 1426<br />

NSCLC clinical specimens, 174 ALK(�) and 1252 ALK(-) by standard<br />

criteria, and 26 NSCLC cell lines (2 ALK(�) and 24 ALK(-)) were<br />

investigated. Results: Native ALK gene mean copy number was significantly<br />

higher in ALK(-) than in ALK(�) cases (2.8, SD 0.93, range 1.2-11.4 vs.<br />

1.8, SD 0.79; range 0.6 to 5.2; p�0.01). Frequency <strong>of</strong> native ALK copy<br />

number gain (�3 copies/cell in �40% cells) was 19% in ALK(�) and 62%<br />

in ALK(-) tumors (p�0.001). In ALK(-) tumors, abundant focal amplification<br />

<strong>of</strong> native ALK was rare (0.8%); scanty amplification (�10% tumor<br />

cells) occurred in 1.1%, and duplication <strong>of</strong> the entire native ALK or <strong>of</strong> the<br />

ALK 3’ end occurred in 3.5%. Among ALK(-) cell lines, mean native ALK<br />

copy number ranged 2.1-6.9 and was not correlated with in vitro crizotinib<br />

sensitivity (IC50s 0.34-2.8 uM). In (�) patients, neither native or rearranged<br />

ALK copy number, nor percentage cell count correlated with<br />

maximal tumor shrinkage or PFS with crizotinib. <strong>Clinical</strong> specimens with<br />

0-9%, 10-15%, 16-30%, 31-50% and �50% <strong>of</strong> ALK� cells were found in<br />

79.3%, 8.5%, 1.4%, 2.7% and 8.1% <strong>of</strong> cases, respectively. Conclusions:<br />

Lower native ALK copy number in ALK(�) NSCLC suggests ALK fusion<br />

occurs early, preceding chromosomal instability. Elevated native ALK copy<br />

number rarely reflects focal amplification and native ALK copy number<br />

increases alone are not associated with sensitivity to ALK inhibition in vitro.<br />

Neither native or rearranged copy number, nor positive cell count within<br />

ALK(�) tumors influences clinical benefit from ALK inhibition. As 8.5% <strong>of</strong><br />

ALK(-) cases fall within 5% <strong>of</strong> the established �15% cell positive<br />

threshold, further investigation <strong>of</strong> ALK status by other diagnostic techniques<br />

in this subset may be warranted.<br />

7533 Poster Discussion Session (Board #23), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Results <strong>of</strong> a global phase II study with crizotinib in advanced ALK-positive<br />

non-small cell lung cancer (NSCLC). Presenting Author: Dong-Wan Kim,<br />

Seoul National University Hospital, Seoul, South Korea<br />

Background: Approximately 3–5% <strong>of</strong> NSCLC harbors ALK gene rearrangements.<br />

Crizotinib is a first-in-class, oral, small-molecule competitive ALK<br />

inhibitor with anti-MET activity. Methods: PROFILE 1005 is an ongoing<br />

global, multicenter, open-label, single-arm, phase II study evaluating the<br />

safety and efficacy <strong>of</strong> crizotinib (250 mg BID in 3-week cycles) in patients<br />

with advanced ALK-positive NSCLC who progressed after �1 chemotherapy<br />

for recurrent/advanced/metastatic disease. Tumor response was<br />

evaluated by RECIST 1.1 every 6 weeks. Patient-reported symptoms and<br />

global quality <strong>of</strong> life (QOL) were assessed using the EORTC QLQ-C30 and<br />

LC-13 at baseline, day 1 each cycle and at end <strong>of</strong> treatment. Results: As <strong>of</strong><br />

June 2011, 439 patients were evaluable for safety and 255 patients for<br />

tumor response. Median age was 53 years. The majority <strong>of</strong> patients were<br />

female (53%), never smokers (65%), and had adenocarcinoma (92%),<br />

ECOG PS 0–1 (83%) and �2 prior chemotherapy regimens (85%). Among<br />

patients evaluable for efficacy, median treatment duration was 25 weeks<br />

(77% <strong>of</strong> patients still ongoing). ORR was 53% (95% CI: 47–60), disease<br />

control rate at 12 weeks was 85% (95% CI: 80–89), median duration <strong>of</strong><br />

response was 43 weeks (96% CI 36–50) and median PFS was 8.5 months<br />

(95% CI: 6.2–9.9). The most frequent treatment-related AEs were visual<br />

effects (50%), nausea (46%), vomiting (39%), and diarrhea (35%), mostly<br />

grade 1–2. 29 patients (6.6%) had treatment-related SAEs, including<br />

dyspnea and pneumonitis (4 patients each; 0.9%), and febrile neutropenia<br />

and renal cyst (2 patients each; 0.5%). A statistically significant (p�0.05)<br />

and clinically meaningful (� 10 points) improvement from baseline was<br />

observed for patient-reported overall pain, pain in chest, cough, dyspnea,<br />

insomnia, fatigue and global QOL. Conclusions: Crizotinib demonstrated a<br />

high response rate and PFS, favorable tolerability pr<strong>of</strong>ile and improvement<br />

in patient-reported symptoms. These results provide strong evidence for<br />

crizotinib as a standard <strong>of</strong> care for advanced ALK-positive NSCLC.<br />

7535 General Poster Session (Board #44A), Sat, 1:15 PM-5:15 PM<br />

Association <strong>of</strong> epithelial to mesenchymal transition with efficacy <strong>of</strong><br />

EGFR-TKIs in non-small cell lung cancer patients with EGFR wild type.<br />

Presenting Author: Shengxiang Ren, Shanghai Pulmonary Hospital, Tongji<br />

University School <strong>of</strong> Medicine, Shanghai, China<br />

Background: The epithelial to mesenchymal transition (EMT) is a fundamental<br />

biological process during which epithelial cells change to a mesenchymal<br />

phenotype, it has a pr<strong>of</strong>ound impact on cancer progression and<br />

treatment. The purpose <strong>of</strong> this study was to investigate the role <strong>of</strong> EMT to<br />

predict the efficacy <strong>of</strong> epidermal growth factor receptor tyrosine kinase<br />

inhibitors (EGFR-TKIs) in advanced non-small cell lung cancer (NSCLC)<br />

patients. Methods: We evaluated the correlation between EMT and sensitivity<br />

to EGFR-TKIs in advanced NSCLC patients. Immunohistochemistry was<br />

used to examine the expression <strong>of</strong> EMT markers, E-cadherin, fibronectin,<br />

N-cadherin and vimentin and ARMs was used to detect the EGFR mutation<br />

in tumor specimens obtained before the treatment. The EMT phenotype<br />

status was determined according to the expression <strong>of</strong> E-cadherin, fibronectin,<br />

N-cadherin and vimentin. Results: 101 patients enrolled into this study<br />

and 97 had enough tissue to perform the EMT examination. The median<br />

age was 59 years old, male/female:56/45, never smoker/smoker: 71/30,<br />

adenocarcinoma/non-adeno: 73/28, PS 0-1/2-3: 83/18, Stage IIIB/IV:<br />

2/99, 1st /2nd-4th line: 13/88. The response rate and progression free<br />

survival(PFS) in the whole population were 45.5% and 7.6 months<br />

respectively. EMT test revealed that 46(47.4%) samples were epithelial<br />

phenotype, while 51(52.6%) were mesenchymal phenotype. 38 <strong>of</strong> 79<br />

patients harbour activating EGFR mutation. Among the patients with EGFR<br />

wild type or unknown status, patients with an epithelial phenotype had a<br />

higher response rate(40% Vs 17.6%, P�0.056) and significantly longer<br />

PFS(7.6 m Vs 3.8 m, P�0.045) than these with a mesenchymal phenotype.<br />

However, the response rate(85.7% Vs 64.7%, P�0.249) and<br />

PFS(15.2 m Vs 12.2 m, P�0.420) were similar in the patients with<br />

activated EGFR mutation. Conclusions: Patients with an epithelial phenotype<br />

got more benefit from the treatment <strong>of</strong> EGFR-TKIs in the advanced<br />

EGFR wild type NSCLC patients, which indicated that the EMT might be a<br />

potential marker to guide the individual therapy <strong>of</strong> EGFR-TKIs in this<br />

subpopulation.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


7536 General Poster Session (Board #44B), Sat, 1:15 PM-5:15 PM<br />

A randomized, double-blind, placebo-controlled phase II study <strong>of</strong> tigatuzumab<br />

(CS-1008) in combination with carboplatin/paclitaxel in patients<br />

with chemotherapy-naive metastatic/unresectable non-small cell lung cancer<br />

(NSCLC). Presenting Author: Joachim Von Pawel, Asklepios Klinikum<br />

Gauting, Munich, Germany<br />

Background: Tigatuzumab (T), a humanized monoclonal antibody that acts<br />

as a DR5 agonist induces apoptosis in human cancer cell lines and has<br />

shown activity in a phase 1 trial; phase 2 trials are ongoing in several tumor<br />

types. In NSCLC cells, single-agent T shows anti-cancer activity. Methods:<br />

Patients (pts) with histologically/cytologically confirmed (TNM ed6) NSCLC<br />

stage IIIB/IV, measurable disease by RECIST (v1.0), and ECOG-PS 0-1<br />

were enrolled at 15 European sites. Pts received T or placebo (PL) IV �<br />

carboplatin/paclitaxel (C/P) every 3 weeks (wks) (1 cycle) for up to 6 cycles.<br />

In the case <strong>of</strong> complete response (CR)/partial response (PR) or stable<br />

disease, T or PL was given as maintenance therapy. The dosage regimen<br />

was T 10 mg/kg or PL at wk 1/cycle 1 and 8 mg/kg or PL every 3 wks<br />

thereafter; P: 175 mg/m2 every 3 wks; C: AUC 6 every 3 wks. Primary<br />

endpoint: progression-free survival (PFS); secondary endpoints: overall<br />

survival (OS) and objective response rate (ORR) (CR � PR), safety. Efficacy<br />

was assessed in both the full population and a prospectively-defined FCGR<br />

genotype subset. Results: 97/100 pts (9.3% stage IIIB wet; 90.7% stage<br />

IV; 70.1% non-squamous; 29.9% squamous) were eligible for efficacy<br />

analyses (49 to T; 48 to PL). Median PFS (95% CI) was 5.4 months (3.3,<br />

6.6) for T vs 4.3 months (4.1, 5.8) for PL; HR � 0.96; 95% CI: 0.6, 1.6.<br />

Median OS (95% CI) was 8.4 months (6.9, 16.3) for T vs 9.0 months (7.6,<br />

14.5) for PL (HR � 0.95; 95% CI: 0.6, 1.6). 12 pts (24.5%) in the T arm<br />

and 11 pts (22.9%) in the PL arm had PR; no pt had CR. In a subset <strong>of</strong> pts<br />

(n � 25) with FCGR2A-H131R or FCGR3A-V158F genotypes, there was a<br />

non-significant trend towards increased PFS with T vs PL (HR � 0.47; 95%<br />

CI: 0.16, 1.35) but no difference in OS. T was well tolerated; the only<br />

increased grade 3/4 toxicity was grade 3 neutropenia (16% with T vs 4%<br />

with PL). The number <strong>of</strong> treatment cycles was similar in both treatment<br />

arms. Conclusions: T does not improve the efficacy <strong>of</strong> C/P as treatment for<br />

systemic therapy-naïve, unselected advanced NSCLC pts. T was well<br />

tolerated and is being investigated in other settings.<br />

7539 General Poster Session (Board #44E), Sat, 1:15 PM-5:15 PM<br />

A novel targeted oral insulin-like growth factor-1 receptor (IGF-1R) inhibitor<br />

and its implications for patients with non-small cell lung cancer<br />

(NSCLC): A phase I clinical trial. Presenting Author: Simon Ekman,<br />

Department <strong>of</strong> Oncology, Uppsala University Hospital, Uppsala, Sweden<br />

Background: The small-molecule IGF-1R inhibitor picropodophyllin (PPP)<br />

is the active compound in the oral suspension AXL1717. PPP has shown<br />

extensive preclinical antitumor effects against a wide range <strong>of</strong> cancers<br />

supporting its use as a single agent treatment. Methods: The clinical phase<br />

Ia/b study on advanced progressive cancer patients with various solid<br />

tumors and without remaining treatment options aimed at establishing the<br />

recommended phase II dose (RPTD) and the maximum tolerated dose<br />

(MTD) <strong>of</strong> AXL1717. Phase Ia consisted <strong>of</strong> single day dosing and phase Ib<br />

multiday dosing (to an accumulated total <strong>of</strong> 28 days <strong>of</strong> treatment; 2*28<br />

days following amendment) with 3 weeks intermission between treatments.<br />

Non-progressing patients could continue treatment in the extension study<br />

without time limitation (treated 119 weeks). PK samples were obtained at<br />

10 different time-points after the morning dose when appropriate. Results:<br />

Phase Ia included 16 patients treated with 78 BID doses ranging from<br />

5-2900 mg AXL1717 BID without any dose-limiting toxicity. Phase Ib<br />

included 39 patients treated with doses between 290-930 mg BID in<br />

periods between 7-28 days, totally for 147 weeks. Phase Ib showed that<br />

AXL1717 was well tolerated and neutropenia was the only detected<br />

dose-related, reversible, dose-limiting toxicity (DLT). RPTD dose was set at<br />

390 mg BID to minimize neutropenia. Although the study was not designed<br />

for efficacy assessment, some patients, mainly with NSCLC, showed signs<br />

<strong>of</strong> clinical benefit, including 3 partial responses and 12 patients with stable<br />

disease. The 15 patients with NSCLC and treatment duration <strong>of</strong> more than<br />

2 weeks with single agent AXL1717 in 3rd or 4th line showed a median<br />

time to progression <strong>of</strong> 31 weeks and a survival time <strong>of</strong> 60 weeks with 4<br />

patients being alive at cut-<strong>of</strong>f. Down-regulation <strong>of</strong> IGF-1R on granulocytes<br />

and increases <strong>of</strong> serum IGF-1 were seen. The systemic exposure <strong>of</strong><br />

AXL1717 was dose-dependent and sufficient for antitumor effects.<br />

Conclusions: AXL1717 has a good safety pr<strong>of</strong>ile and demonstrated promising<br />

clinical benefits in this severely ill and heavily pretreated patient cohort,<br />

especially in patients with NSCLC.<br />

Lung Cancer—Non-small Cell Metastatic<br />

489s<br />

7537 General Poster Session (Board #44C), Sat, 1:15 PM-5:15 PM<br />

Intratumoral heterogeneity <strong>of</strong> EGFR mutation and clinical significance in<br />

Chinese patients with advanced non-small cell lung cancer. Presenting<br />

Author: Hua Bai, Peking University School <strong>of</strong> Oncology, Beijing Cancer<br />

Hospital and Institute, Beijing, China<br />

Background: Whether exist intratumoral heterogeneity <strong>of</strong> epidermal growth<br />

factor receptor (EGFR) gene mutation remains controversial. This study<br />

aims to evaluate intratumoral heterogeneity <strong>of</strong> EGFR and its clinical<br />

significance in patients with non-small cell lung cancer (NSCLC). Methods:<br />

Samples were collected from 85 patients with stage IIIa-IV who had<br />

palliative surgery resection. All <strong>of</strong> the bulk tissues had been routinely<br />

detected EGFR mutation, in which contained 30 wild-type and 55<br />

mutant-type EGFR. 28–34 tumor foci for each sample were microdissected.<br />

EGFR mutation was detected by ARMS. EGFR copy number was<br />

detected by FISH. 23 patients received EGFR-tyrosine kinase inhibitor<br />

(TKIs) therapy. Results: 1740 tumor foci from 55 EGFR mutant-type<br />

(Group-M) and 959 foci from 30 wild-type cases(Group-W) were fractionated.<br />

Mean mutation contents were 73.9%(1285/1740) in Group-M and 0<br />

in Group-W. Accordingly, 28.2%(24/85)consisted <strong>of</strong> cells with both<br />

wild-type and mutated-type EGFR, with the proportion <strong>of</strong> EGFR mutant<br />

cells ranging from 1% to 90%, whereas 71.8%(61/85) samples contained<br />

pure mutant-type (n�31) or wild-type EGFR cells(n�30). 31 patients<br />

(36.5%) presented EGFR FISH positive. The patients with mutation<br />

heterogeneity, regardless <strong>of</strong> high or low mutation contents, presented lower<br />

EGFR copy number than those with pure mutation cases (16.7% vs<br />

71.0%,p�0.05). Among 23 patients who received EGFR-TKIs therapy, the<br />

mean mutation content was higher in partial response (71.1%) and stable<br />

disease group (43.4%) than in progressive disease group (5.0%) (p�0.005).<br />

Conclusions: This study showed that the heterogeneity <strong>of</strong> EGFR mutation<br />

was found in introtumoral region with different degree. The response to<br />

EGFR-TKIs was correlated with EGFR mutant content.<br />

7540 General Poster Session (Board #44F), Sat, 1:15 PM-5:15 PM<br />

Differential progression-free survival (PFS) to erlotinib according to EGFR<br />

exon 19 deletion type in non-small cell lung cancer (NSCLC) patients (p) in<br />

the EURTAC study. Presenting Author: Enric Carcereny Costa, Catalan<br />

Institute <strong>of</strong> Oncology, Hospital Germans Trias i Pujol, Barcelona, Spain<br />

Background: Different exon 19 deletion types have shown different in vitro<br />

sensitivity to erlotinib, with the lower IC50 for deletion E746_A750<br />

(ELREA) (Yuza et al. Cancer Biol Ther 2007). This information prompted us<br />

to examine outcome according to type <strong>of</strong> exon 19 deletion in the EURTAC<br />

study. Methods: The EURTAC trial (clinicaltrials.gov NCT00446225)<br />

randomized 174 p with EGFR exon 19 deletions or L858R mutations to<br />

receive erlotinib or chemotherapy. Progression-free survival (PFS) was 9.7<br />

months (m) vs 5.2 m, respectively (P�0.0001). Exon 19 deletions were<br />

divided into two groups: ELREA vs non-ELREA deletions. Results: Exon 19<br />

deletions were present in 57 p in the erlotinib arm and in 58 p in the<br />

chemotherapy arm. ELREA deletions were found in 41 p (71.9%) in the<br />

erlotinib arm and in 38 p (65.5%) in the chemotherapy arm. Non-ELREA<br />

deletions were found in 16 p in the erlotinib arm and in 20 p in the<br />

chemotherapy arm. There were no differences in p characteristics between<br />

treatment arms according to type <strong>of</strong> deletion. PFS for p with ELREA<br />

deletions was 9.4 m in the erlotinib arm and 4.6 in the chemotherapy arm<br />

(HR, 0.36; P�0.0004). PFS for p with non-ELREA deletions was not<br />

reached in p in the erlotinib arm and was 5.3 m for p in the chemotherapy<br />

arm (HR, 0.17; P�0.001). The multivariate analysis identified erlotinib<br />

arm (P�0.001) and non-ELREA deletions (P�0.001) as independent<br />

markers <strong>of</strong> longer PFS. Overall survival (OS) for p with ELREA deletions was<br />

17 m in the erlotinib arm and 18.4 in the chemotherapy arm (P�0.575).<br />

OS for p with non-ELREA deletions was not reached in the erlotinib arm and<br />

19.5 m in the chemotherapy arm (P�0.216). Response rate (RR) for p with<br />

ELREA deletions was 53.6% in the erlotinib arm vs 15.7% in the<br />

chemotherapy arm (P�0.004). RR for p with non-ELREA deletions was<br />

68.7% in the erlotinib arm vs 10% in the chemotherapy arm (P�0.001).<br />

Conclusions: To date, no biological reason has been identified that can<br />

explain the greater sensitivity to erlotinib in p with non-ELREA exon 19<br />

deletions. Our findings indicate the need to define the type <strong>of</strong> deletion prior<br />

to treatment since this information can be helpful in predicting the<br />

duration <strong>of</strong> response.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


490s Lung Cancer—Non-small Cell Metastatic<br />

7541 General Poster Session (Board #44G), Sat, 1:15 PM-5:15 PM<br />

A phase II multicenter study <strong>of</strong> aflibercept (AFL) in combination with<br />

cisplatin (C) and pemetrexed (P) in patients with previously untreated<br />

advanced/metastatic nonsquamous non-small cell lung cancer (NSCLC).<br />

Presenting Author: Hongbin Chen, Roswell Park Cancer Institute, Buffalo,<br />

NY<br />

Background: AFL is a recombinant human fusion protein that acts as a<br />

decoy receptor and prevents the interaction <strong>of</strong> vascular endothelial growth<br />

factor (VEGF)-A, VEGF-B, and placental growth factor with their receptors.<br />

This study evaluated the efficacy and safety <strong>of</strong> AFL in combination with<br />

first-line chemotherapy <strong>of</strong> C and P in NSCLC. Methods: This phase II, single<br />

arm, open label, multicenter trial in patients with previously untreated,<br />

locally advanced, or metastatic NSCLC excluded patients with squamous<br />

histology, cavitating lesions, ECOG � 1, uncontrolled hypertension, or<br />

brain/CNS metastases. All patients received IV AFL 6 mg/kg, P 500 mg/m2 ,<br />

and C 75mg/m2 , every 3 weeks for up to 6 cycles. For those who completed<br />

the combined chemotherapy, Q3W administration <strong>of</strong> AFL was to continue<br />

until disease progression, intolerable toxicity, or any other cause for<br />

withdrawal. The primary endpoints were objective response rate (ORR) and<br />

progression free survival (PFS). Planned sample size was 72 patients.<br />

Results: The study was closed prematurely due to 3 confirmed cases <strong>of</strong><br />

reversible posterior leukoencephalopathy syndrome (RPLS). A total <strong>of</strong> 42<br />

patients were enrolled. Median age was 61.5 years; 54.8% were male,<br />

85.7% white and 50% ECOG 0. A median <strong>of</strong> 4 cycles <strong>of</strong> AFL was<br />

administered. The most common treatment-emergent adverse events<br />

(TEAEs) <strong>of</strong> any grade were nausea (69%) and fatigue (67%), with<br />

hypertension (36%) as the most common grade 3/4 TEAE. The 3 patients<br />

with RPLS were Caucasian women. Two had a history <strong>of</strong> hypertension and<br />

both experienced elevated BP and reduced CrCl. Of the 38 patients<br />

evaluable for response, ORR was 26.3% (95% CI, 12.3-40.3%) and<br />

median PFS was 5 months (95% CI, 4.3-7.1). Conclusions: The rate <strong>of</strong><br />

RPLS observed in this study with AFL � C � P was higher than anticipated.<br />

A meta-analysis <strong>of</strong> safety from three large placebo-controlled studies<br />

reported no RPLS among 1333 patient treated with AFL � chemotherapy,<br />

and none was reported in prior combination studies <strong>of</strong> AFL � PorAFL� C<br />

� docetaxel. Though the study was stopped early, ORR and PFS were in<br />

accordance with most historical first-line NSCLC studies.<br />

7543 General Poster Session (Board #45A), Sat, 1:15 PM-5:15 PM<br />

Phase II study <strong>of</strong> the HSP90 inhibitor AUY922 in patients with previously<br />

treated, advanced non-small cell lung cancer (NSCLC). Presenting Author:<br />

Edward B. Garon, David Geffen School <strong>of</strong> Medicine, University <strong>of</strong> California<br />

Los Angeles, Los Angeles, CA<br />

Background: AUY922 is a highly potent, non-geldanamycin, HSP90 inhibitor.<br />

A Phase I study in advanced solid tumors provided a recommended<br />

Phase II dose <strong>of</strong> 70 mg/m2 . HSP90 acts as a chaperone <strong>of</strong> client proteins,<br />

including those relevant in NSCLC pathogenesis. This Phase II study<br />

assessed AUY922 in pts with previously treated advanced NSCLC stratified<br />

by molecular status. Methods: Pts with advanced NSCLC who progressed<br />

following �2 prior lines <strong>of</strong> chemotherapy, received AUY922 70 mg/m2 as a<br />

1-hr infusion once-weekly. Pts were assigned to 4 strata: EGFR activating<br />

mutations (EGFR mut), KRAS mut, ALK rearranged (ALK�), or EGFR/KRAS/<br />

ALK wild type (wt). Phase II Bayesian hypothesis testing design allowed<br />

sharing <strong>of</strong> information between strata based on assessing similarity in<br />

observed response data. Primary endpoint was confirmed (RECIST) objective<br />

response rate (ORR) or stable disease (SD) at 18 wks. Secondary<br />

endpoints included OS, PFS, and safety/tolerability. Results: A total <strong>of</strong> 112<br />

pts (median age 60 years; 45% male; 86% adenocarcinoma; 28 [25%] pts<br />

KRAS mut; 35 [31%] EGFR mut; 14 [12%] ALK�; 31 [28%] EGFR/KRAS/<br />

ALK wt) were treated at the December 16th 2011 cut<strong>of</strong>f. Most pts were<br />

heavily pretreated: 61% had received �3 prior systemic regimens; 64%<br />

had WHO PS 1 or 2. Mean duration <strong>of</strong> exposure was 9.1 wks. The most<br />

frequent adverse events (AEs, any grade [Gr]) were diarrhea (73%), visual<br />

AEs (71%), and nausea (43%). Most AEs were Gr 1/2; Gr 3/4 AE’s were rare<br />

(all �10%). The study is ongoing, and 29 pts were still receiving treatment<br />

at the cut<strong>of</strong>f date. Preliminary clinical activity <strong>of</strong> AUY922 (RECIST;<br />

investigator assessed) was seen with partial responses in 13/101 (13%)<br />

pts: 2/8 (25%) ALK� pts, 6/33 (18%) EGFR mut pts, 4/30 (13%)<br />

EGFR/KRAS/ALK wt pts, 0/26 (0%) KRAS mut pts, and 1/4 (25%) pts <strong>of</strong><br />

unknown status. In ALK� pts, responses were seen in crizotinib (CRZ)naive<br />

pts, and SD was seen with tumor shrinkage in CRZ-resistant pts. PFS<br />

data will be presented. Conclusions: AUY922 had an acceptable safety<br />

pr<strong>of</strong>ile and demonstrated preliminary activity in heavily pretreated pts with<br />

advanced NSCLC. This trial demonstrates clinical activity <strong>of</strong> AUY922 in<br />

EGFR mut and EGFR/KRAS/ALK wt NSCLC pts in addition to ALK� pts.<br />

7542 General Poster Session (Board #44H), Sat, 1:15 PM-5:15 PM<br />

Skin toxicity associated with outcome to erlotinib in non-small cell lung<br />

cancer (NSCLC) patients (p) with EGFR mutations in the EURTAC study.<br />

Presenting Author: Cristina Buges, Catalan Institute <strong>of</strong> Oncology, Hospital<br />

Germans Trias i Pujol, Barcelona, Spain<br />

Background: Rash is reported in 75% <strong>of</strong> p treated with erlotinib and has<br />

been associated with improved overall and progression-free survival (PFS)<br />

in unselected p although not specifically in p with EGFR mutations.<br />

Methods: The EURTAC trial (clinicaltrials.gov NCT00446225) randomized<br />

174 p with EGFR exon 19 deletions or L858R mutations to receive<br />

erlotinib or chemotherapy. Overall PFS was 9.7 months (m) vs 5.2 m,<br />

respectively (P�0.0001). Rash was defined according to NCI CTC v3.0 and<br />

dichotomized by grades 0-1 vs 2�. Grade 2 is macular or popular eruption<br />

or erythema with pruritus or other associated symptoms, localized desquamation<br />

or other lesions covering �50% <strong>of</strong> body surface area. We examined<br />

outcome in the erlotinib arm according to rash grade. Results: 16pinthe<br />

erlotinib arm had no rash (grade 0), 30 had grade 1, and 40 had grade 2�.<br />

80% <strong>of</strong> cases <strong>of</strong> rash grade 2� occurred in p with exon 19 deletions, while<br />

only 20% were in p with L858R mutations (P�0.039). There were no<br />

differences in rash grade according to sex, age or smoking status. PFS was<br />

11.2 m in p with rash grade 2� and 8.4 m in p with grade 0-1 (HR, 0.52;<br />

P�0.018). There was no correlation between rash grade 0-1 vs 2� and<br />

response or overall survival (OS). Interestingly, when p were divided into<br />

those with no rash (16 p) and those with grade 2� (40 p), PFS was 11.2 m<br />

for p with grade 2� vs 2.7 m for p with no rash (P�0.031), and OS was<br />

24.9 m for p with grade 2� vs 16.1 m for p with no rash (P�0.033).<br />

Conclusions: Although the biological reasons for the association <strong>of</strong> skin rash<br />

with better outcome to erlotinib have not been elucidated, this sub-analysis<br />

<strong>of</strong> the EURTAC trial has enabled us to confirm for the first time that skin<br />

rash – especially grade 2� - can predict better outcome to erlotinib in p<br />

with EGFR mutations. In addition, the correlation between rash and type <strong>of</strong><br />

EGFR mutation warrants further investigation.<br />

7544 General Poster Session (Board #45B), Sat, 1:15 PM-5:15 PM<br />

GAIN-(L): Efficacy and biomarker findings <strong>of</strong> RG7160 (GA201), a novel,<br />

dual-acting monoclonal antibody (mAb) designed to enhance antibodydependent<br />

cellular cytotoxicity (ADCC), in combination with first-line<br />

cisplatin and pemetrexed in metastatic nonsquamous NSCLC. Presenting<br />

Author: James F. Spicer, King’s College London, Guy’s Hospital Campus,<br />

London, United Kingdom<br />

Background: GA201, a humanized, engineered IgG1 anti-Epidermal Growth<br />

Factor Receptor (EGFR) mAb designed to enhance ADCC, has shown<br />

promising clinical activity in phase I and in the neoadjuvant treatment <strong>of</strong><br />

head and neck cancer. This phase Ib study (NCT01185847) aimed to<br />

determine the safety, pharmacokinetics (PK), activity and recommended<br />

phase II dose (RP2D) <strong>of</strong> GA201 in combination with chemotherapy in<br />

non-squamous NSCLC. Methods: Successive cohorts received GA201<br />

1000 mg or 1400 mg (IV d1, d8 then 2-weekly (q2W)) in combination with<br />

chemotherapy at standard doses. Data cut <strong>of</strong>f was 7 months after enrolling<br />

the last patient. Results: 14 patients (4 female) with performance status<br />

0-1 were enrolled. No maximum tolerated dose was reached. Most common<br />

adverse events (AEs – all grades) included rash (100%), hypomagnesaemia<br />

(71%), infusion-related reactions (64%), mucosal inflammation (57%)<br />

and anemia (50%). AEs <strong>of</strong> � grade 3 included rash (71%), skin fissure<br />

(21%), dry skin (14%), paronychia (14%), and asthenia (14%). Median<br />

timeto improvement <strong>of</strong> rash grade 3 was 11 days. AEs led to dose reduction<br />

for 4 patients and discontinuation for 1 patient. There were 6 confirmed<br />

partial responses (43%, 5 in 1400 mg cohort) and 7 patients (50%) with<br />

stable disease ��9 weeks. Duration <strong>of</strong> response ranged between 5 and 42<br />

weeks (3 patients still ongoing). Preliminary biomarker analysis shows a<br />

correlation between tumor-infiltrating CD16� immune cells and target<br />

lesion shrinkage at first tumor assessment [R(spear)�-0.65 (p�0.02)]; no<br />

apparent correlation between EGFR H-score and response was found. PK<br />

data supports 1400 mg as the RP2D (d1, d8 then q2W). Conclusions: The<br />

RP2D <strong>of</strong> GA201 in combination with chemotherapy was established to be<br />

1400 mg. The incidence <strong>of</strong> EGFR associated rash was high and guidelines<br />

to reduce its severity were implemented with a noted improvement in<br />

tolerability. Promising antitumor activity was observed. Biomarker data<br />

support the mode <strong>of</strong> action <strong>of</strong> GA201 via ADCC. A randomized phase II trial<br />

<strong>of</strong> this combination is ongoing and fully recruited.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


7545 General Poster Session (Board #45C), Sat, 1:15 PM-5:15 PM<br />

FGFR1 amplification and EGFR mutation in Chinese squamous cell lung<br />

cancer. Presenting Author: Zhigang Wei, Department <strong>of</strong> Thoracic Medical<br />

Oncology, Department <strong>of</strong> Thoracic Surgery, Peking University School <strong>of</strong><br />

Oncology, Beijing Cancer Hospital and Institute, Beijing, China<br />

Background: Squamous cell lung cancer (SCC) lacks for effective targeted<br />

therapies. FGFR1 amplification has emerged as a potential biomarker. This<br />

study aimed to explore clinicopathologic characteristics <strong>of</strong> FGFR1 amplification<br />

in Chinese SCC patients and further explore the correlation between<br />

FGFR1 amplification and EGFR mutations. Methods: One hundred seventyseven<br />

SCC patients were included in this retrospective study. Gene copy<br />

number <strong>of</strong> FGFR1 and EGFR mutations were detected by fluorescence in<br />

situ hybridization (FISH) and denaturing high-performance liquid chromatography<br />

(DHPLC), respectively. Results: FGFR1 amplifications were detected<br />

in 24.9% (44/177) Chinese SCC patients. FGFR1 amplification in<br />

SCC was more common in male (28.0%, 40/143) and smokers (28.7%,<br />

39/136) than female (11.8%, 4/34, p�0.049) and nonsmokers (12.2%,<br />

5/41, p�0.032). FGFR1 amplification and EGFR mutations were mutually<br />

exclusive (p�0.006), fourty-one <strong>of</strong> 139(29.4%) patients with wild-type<br />

EGFR had FGFR1 amplification, while 3 <strong>of</strong> 38 (7.9%) patients with EGFR<br />

mutation had FGFR1 amplification. Conclusions: FGFR1 amplification was<br />

common in Chinese squamous cell lung cancer, and mutually exclusive<br />

with EGFR mutations.<br />

7547 General Poster Session (Board #45E), Sat, 1:15 PM-5:15 PM<br />

Delay <strong>of</strong> chemotherapy through use <strong>of</strong> post-progression erlotinib in patients<br />

with EGFR-mutant lung cancer. Presenting Author: Ge<strong>of</strong>frey R. Oxnard,<br />

Dana-Farber Cancer Institute, Boston, MA<br />

Background: 1st-line tyrosine kinase inhibitor (TKI) therapy for patients<br />

(pts) with EGFR-mutant lung cancer prolongs progression free survival<br />

(PFS) and improves quality <strong>of</strong> life. When pts develop acquired resistance to<br />

TKI, chemotherapy is the only approved systemic treatment. Anecdotal<br />

evidence suggests that change <strong>of</strong> therapy can be delayed in some pts<br />

through continued TKI beyond progression (PD), but the effectiveness <strong>of</strong><br />

this approach has not been quantified. Methods: Through an IRB-approved<br />

mechanism, pts with advanced lung cancer and EGFR sensitizing mutations<br />

treated on 3 prospective trials <strong>of</strong> 1st-line erlotinib were identified.<br />

Only pts with RECIST PD while on erlotinib were studied. Time from PD<br />

until use <strong>of</strong> an alternate systemic therapy (or death) and characteristics at<br />

PD (development <strong>of</strong> new extra-thoracic metastases or cancer-related<br />

symptoms) were assessed. Results: 42 eligible pts were identified with the<br />

following characteristics: median age 70, 86% female, 93% non-Asian,<br />

50% never-smokers, 83% adenocarcinoma, 100% EGFR-mutant (55%<br />

exon 19, 36% exon 21, 9% exon 18). Median PFS on erlotinib was 13<br />

months. After PD, alternate systemic therapy was delayed �3 months in 19<br />

pts (45%; 95%CI: 31%-60%), through a combination <strong>of</strong> post-PD erlotinib<br />

(16 pts), radiation (6 pts), and/or surgery (3 pts), or on observation alone (2<br />

pts). These 19 pts commonly had exon 19 deletions and were more likely to<br />

have no cancer-related symptoms at PD (Table), and had a median post-PD<br />

survival <strong>of</strong> 29 months. Alternate systemic therapy was delayed �12<br />

months in 8 pts (19%). Conclusions: In a subset <strong>of</strong> pts with EGFR-mutant<br />

lung cancer and acquired resistance to TKI, chemotherapy can be delayed<br />

with the aid <strong>of</strong> post-PD TKI and local treatment modalities. This indolent<br />

PD is likely due to ongoing tumor EGFR dependence. In pts who are<br />

tolerating TKI and have asymptomatic progression, we recommend this<br />

palliative treatment strategy as an option for delaying chemotherapy and<br />

maximizing quality <strong>of</strong> life.<br />

Delay <strong>of</strong> alternate systemic therapy<br />

>3m<br />

(n�19)<br />


492s Lung Cancer—Non-small Cell Metastatic<br />

7549 General Poster Session (Board #45G), Sat, 1:15 PM-5:15 PM<br />

Phase III study (MONET1) <strong>of</strong> motesanib plus carboplatin/paclitaxel (C/P) in<br />

patients with advanced nonsquamous non-small cell lung cancer (NSCLC):<br />

Asian subgroup analysis. Presenting Author: Yukito Ichinose, National<br />

Kyushu Cancer Center, Fukuoka, Japan<br />

Background: MONET1 evaluated overall survival (OS) in patients (pts) with<br />

nonsquamous NSCLC receiving motesanib (an oral VEGFR 1, 2 and 3,<br />

PDGFR and Kit inhibitor) plus C/P compared with pts receiving placebo<br />

plus C/P. Analysis <strong>of</strong> the total population (N�1090) showed that motesanib<br />

� C/P did not significantly improve OS vs C/P alone (primary<br />

endpoint). Here we present results <strong>of</strong> a subgroup analysis <strong>of</strong> Asian pts.<br />

Methods: Asian pts (Japan, S. Korea, Philippines, Hong Kong, Taiwan,<br />

Singapore) with stage IIIB/IV or recurrent nonsquamous NSCLC and no<br />

prior systemic therapy for advanced NSCLC were analysed. Pts were<br />

randomized to up to six 3-wk cycles <strong>of</strong> C (AUC 6 mg/mL·min) and P (200<br />

mg/m 2 ) with either motesanib 125 mg QD (Arm A) or placebo QD (Arm B)<br />

orally continuously. Results: 227 Asian pts (incl. 106 Japanese pts) with<br />

nonsquamous NSCLC were randomized (Arm A/B, n�110/117); 198 had<br />

adenocarcinoma (n�97/101). Median age was 60 y (range 30–78); 80%<br />

had stage IV disease. At the time <strong>of</strong> analysis, 139 pts had died (118 pts<br />

with adenocarcinoma). Pts received a median <strong>of</strong> 164 days <strong>of</strong> motesanib vs<br />

125 days <strong>of</strong> placebo (vs 106 and 126 days in non-Asian pts). Median<br />

follow-up was 63 wks. Efficacy results are shown in the table. Motesanib/<br />

placebo-related AEs were seen in 94/74% <strong>of</strong> pts respectively; Gr �3<br />

related AEs in 48/22%. Most common emergent AEs were (Arm A/B)<br />

alopecia (78/76%), diarrhea (63/33%), and nausea (55/43%); gallbladder<br />

disorders (Gr 1–2) were seen in 9/2% <strong>of</strong> pts. Gr �3 AEs more frequent in<br />

Arm A vs B included neutropenia (36/22%) and hypertension (13/3%).<br />

Emergent Gr 5 events were seen in (Arm A/B) 5/4% vs 16/11% in<br />

non-Asian pts. Conclusions: In contrast to non-Asian pts, in the subgroup <strong>of</strong><br />

Asian pts with advanced nonsquamous NSCLC, motesanib plus C/P<br />

treatment was associated with increased OS, PFS, and objective response<br />

rates (ORR) compared with C/P alone, with no excess <strong>of</strong> treatment-related<br />

mortality.<br />

Asian subgroup All non-Asian pts<br />

Motesanib �C/P (n�110) Placebo � C/P (n�117) Motesanib �C/P(n�431) Placebo � C/P (n�432)<br />

Median OS (mos)<br />

Median PFS (mos)<br />

20.9*<br />

7.0<br />

14.5 10.9 10.7<br />

† ORR (%) 62<br />

5.3 5.5 5.4<br />

† 27 34* 25<br />

CR �1 0 �1 �1<br />

PR 61 27 33 25<br />

*p�0.05; †p�0.001 vs placebo � C/P<br />

7551 General Poster Session (Board #46A), Sat, 1:15 PM-5:15 PM<br />

A randomized phase II study <strong>of</strong> axitinib in combination with pemetrexed/<br />

cisplatin (pem/cis) as first-line therapy for nonsquamous non-small cell<br />

lung cancer (NSCLC). Presenting Author: Chandra Prakash Belani, Penn<br />

State Hershey Cancer Institute, Hershey, PA<br />

Background: Axitinib is a potent and selective second-generation inhibitor<br />

<strong>of</strong> VEGF receptors 1, 2, and 3 that has promising single-agent activity in<br />

advanced NSCLC. Efficacy and safety <strong>of</strong> axitinib (in 2 dosing schedules)<br />

combined with pem/ciswere evaluatedfor non-squamous NSCLC. Methods:<br />

Patients with confirmed stage IIIB, IV, or recurrent non-squamous NSCLC<br />

and ECOG performance status (PS) 0 or 1 were stratified by gender and PS,<br />

and randomized 1:1:1 to receive six 21-day cycles <strong>of</strong> axitinib continuously<br />

plus pem/cis (arm I); axitinib on Days 2 through 19 followed by a 3-day<br />

interruption plus pem/cis (arm II); or pem/cis alone (arm III). Axitinib was<br />

administered at a starting dose <strong>of</strong> 5 mg BID. Pem/cis (500/75 mg/m2 ) was<br />

infused on Day 1 <strong>of</strong> each cycle. Primary endpoint was progression-free<br />

survival (PFS). Results: Baseline characteristics <strong>of</strong> patients in arm I<br />

(n�55), arm II (n�58), or arm III (n�57) ranged between 59–62 yr<br />

median age; 62–65% male; 71–85% White; 73–85% current/exsmokers;<br />

and 43–47% PS 0. There were no significant differences in PFS<br />

or overall survival (OS) between axitinib-containing arms I and II compared<br />

with pem/cis alone, but objective response rates (ORR) were higher (Table).<br />

Most common all causality grade 3 adverse events (AEs) in arm I, II, and III,<br />

respectively, were hypertension (20%, 17%, 0%); neutropenia (18%,<br />

10%, 9%); nausea (16%, 5%, 7%); vomiting (13%, 5%, 4%); fatigue<br />

(11%,16%,16%); and anemia (7%, 14%, 9%). Grade 4 AEs observed in<br />

�1 patient were asthenia and pulmonary embolism (2 patients each in arm<br />

II). Conclusions: Axitinib combined with pem/cis was generally well tolerated,<br />

but efficacy was not significantly better than pem/cis alone in<br />

non-squamous NSCLC.<br />

Median PFS (95% CI), mo<br />

HR (95% CI) vs Arm III<br />

P<br />

Median OS (95% CI), mo<br />

HR (95% CI) vs Arm III<br />

P<br />

ORR (95% CI), %<br />

Treatment Difference, %<br />

(95% CI) vs Arm III<br />

P<br />

Arm I Arm II Arm III<br />

8.0 (6.5, 10)<br />

0.892 (0.560, 1.423)<br />

0.355<br />

16.6 (12.6, 22.4)<br />

1.079 (0.663, 1.756)<br />

0.632<br />

45.5 (32.0, 59.4)<br />

19.2 (1.7, 36.6)<br />

0.013<br />

7.9 (6.2, 9.5)<br />

1.019 (0.640, 1.623)<br />

0.545<br />

14.7 (11.5, 18.1)<br />

1.391 (0.871, 2.222)<br />

0.891<br />

39.7 (27.0, 53.4)<br />

13.4 (-3.7, 30.3)<br />

0.069<br />

7.1 (5.8, 9.2)<br />

–<br />

15.9 (11.1, –)<br />

–<br />

26.3 (15.5, 39.7)<br />

–<br />

7550 General Poster Session (Board #45H), Sat, 1:15 PM-5:15 PM<br />

Final results <strong>of</strong> a randomized, double-blind, phase II study <strong>of</strong> gemcitabine<br />

plus vandetanib or plus placebo in the treatment <strong>of</strong> advanced (stage<br />

IIIB/IV) non-small cell lung cancer (NSCLC) elderly patients (ZELIG study<br />

NCT00753714). Presenting Author: Cesare Gridelli, SG Moscati Hospital,<br />

Avellino, Italy<br />

Background: Vandetanib (V) is a once-daily oral inhibitor <strong>of</strong> VEGFR, EGFR<br />

and RET signaling. Single-agent gemcitabine (G) is a standard <strong>of</strong> care<br />

option for unselected patients (pts) unfit for doublet platinum based<br />

chemotherapy. This study assessed the progression-free survival (PFS)<br />

benefit <strong>of</strong> G�V compared to G plus placebo (P) in pts with advanced NSCLC<br />

aged � 70 years. Methods: Eligible pts (stage IIIB/IV NSCLC; WHO PS 0-2;<br />

all histologies; chemonaïve, aged �70) were randomized 1:1 to receive G<br />

1200 mg/m2 i.v. day 1 and 8 <strong>of</strong> each 21-day cycle, up to 6 cycles plus V<br />

100 mg/day or plus P until progression/toxicity. The primary objective was<br />

PFS (80% power to detect a hazard ratio [HR] � 0.667). Secondary<br />

endpoints included overall survival (OS), objective response rate (ORR),<br />

disease control rate (DCR), and safety. Results: Between Oct 2008-May<br />

2010, 124 pts (median age 75 yrs (70-84); 72.6% male; 57.2% WHO PS<br />

0-1; 74.2% past/never-smoker; 58.1% adenocarcinoma; 89.5% stage IV)<br />

were randomized to G�V (n�61) or G�P (n�63). Baseline characteristics<br />

were similar in both arms. At data cut-<strong>of</strong>f (Apr11), 87.9% pts<br />

progressed and 73.4% pts had died. PFS was significantly prolonged for<br />

G�V (HR�0.729; 95% CI 0.484-1.096; p�0.0417), median PFS<br />

G�V�6.0 months, G�P�5.5 months. No differences were seen in ORR<br />

(14.8% and 12.7%; p � 0.74), DCR (72.1% and 66.7%; p �0.51), OS<br />

(HR�1.024 [95% CI 0.667-1.571] p�0.8960), proportion <strong>of</strong> pts alive at<br />

1-year G�V�31.1% and G�P�30.2% (p�0.90). Adverse events (AEs)<br />

observed for V 100 mg were generally consistent with previous NSCLC<br />

studies <strong>of</strong> V 100 mg. Common AEs (any grade) occurring with a greater<br />

frequency in the G�V arm included skin toxicity (34.4% vs 15.9%) and<br />

hypertension (9.8% vs 3.2%). Diarrhea and neutropenia were similar in<br />

both arms (14.8% and 14.3%; 19.7% and 19.0%). Conclusions: Despite a<br />

marginally statistically significant improvement in PFS the study did not<br />

met the primary and secondary end points. The combination G�V was well<br />

tolerated in this clinical setting.<br />

7552 General Poster Session (Board #46B), Sat, 1:15 PM-5:15 PM<br />

Molecular marker analysis <strong>of</strong> SWOG S0636, a phase II trial <strong>of</strong> erlotinib and<br />

bevacizumab in never-smokers with advanced NSCLC. Presenting Author:<br />

Philip C. Mack, University <strong>of</strong> California, Davis, Sacramento, CA<br />

Background: S0636 investigated the combination <strong>of</strong> erlotinib and bevacizumab<br />

in never-smoking NSCLC patients with confirmed adenocarcinoma<br />

histology (H. West ASCO 2011). Patient eligibility was not restricted by<br />

molecular selection. Median PFS and OS were encouraging at 8 and 26<br />

months. An analysis <strong>of</strong> molecular markers was undertaken, focusing<br />

initially on the EGFR pathway. Methods: EGFR analysis included gene copy<br />

number, mutation and protein expression. Copy number was conducted by<br />

FISH using the Colorado scoring system. An immunohistochemistry H score<br />

was developed for EGFR protein expression analysis, ranging from 0 to<br />

400. Specimens were evaluable from 42 <strong>of</strong> the 85 eligible patients.<br />

Results: FISH positivity was identified in 17/35 pts (49%), 11 with high<br />

polysomy and 6 with true gene amplification. EGFR activating mutations<br />

were seen in 10/33 pts (30%). IHC H-score �200 was observed in 17/40<br />

pts (43%). All EGFR markers were significantly correlated with one<br />

another. In the EGFR WT subgroup, FISH-positive patients outperformed<br />

FISH-negative pts (mPFS 20 vs, 6 months, p�0.06). Conclusions: Careful<br />

analysis <strong>of</strong> EGFR markers (mutation, FISH and IHC) identified S0636<br />

patients with favorable PFS and encouraging trends for OS. EGFR FISH and<br />

IHC provided additional predictive information beyond that <strong>of</strong> EGFR<br />

mutation status. Supported in part by DHHS: CA32102 and CA38926, and<br />

in part by Genentech.<br />

Group N mPFS (mos) p value mOS (mos) 2-Year OS p value<br />

FISH positive 17 20 NR 0.82<br />

FISH negative 18 6 0.02 17 0.39 0.08<br />

EGFR mutant 10 36 NR 0.53<br />

EGFR wild type 23 8 0.09 26 0.57 0.69<br />

FISH � OR mutant 20 20 NR 0.72<br />

FISH – AND WT 13 6 0.03 15 0.40 0.08<br />

IHC: 0-100 11 6 ref. 18 0.25 ref.<br />

101-200 12 8 0.26 NR 0.61 0.27<br />

201-400 17 19 0.03 NR 0.74 0.06<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


7553 General Poster Session (Board #46C), Sat, 1:15 PM-5:15 PM<br />

Radiographic assessment and therapeutic decisions at RECIST progression<br />

in EGFR-mutant NSCLC treated with EGFR tyrosine kinase inhibitors.<br />

Presenting Author: Mizuki Nishino, Dana-Farber Cancer Institute, Boston,<br />

MA<br />

Background: EGFR mutated advanced NSCLC treated with EGFR TKIs<br />

typically progresses after initial response due to acquired resistance. TKI<br />

therapy is <strong>of</strong>ten continued beyond RECIST progression (PD). We investigated<br />

the frequency <strong>of</strong> this practice and patterns <strong>of</strong> RECIST PD via imaging<br />

findings, as well as the association between patient characteristics and<br />

discontinuation <strong>of</strong> TKI among patients (pts) who progressed while on TKI.<br />

Methods: Among a cohort <strong>of</strong> 101 advanced NSCLC pts with sensitizing<br />

EGFR mutations treated with first-line erlotinib or gefitinib at DFCI, 70 pts<br />

treated between 2002 and 2010 had at least two CT scans for retrospective<br />

radiographic assessments using RECIST1.1; 56 pts had experienced PD by<br />

the data closure date <strong>of</strong> June 2011. Results: Among 56 pts experiencing<br />

PD, 46 (82%) were female, median age was 63 (range 35-79), 28 (50%)<br />

were never-smokers, 32 (57%) had distant mets, 32(57%) had exon 19<br />

deletion, and 50 (89%) received erlotinib. 49 pts (88%) continued TKI<br />

therapy for at least 2 mos beyond retrospectively assessed PD. 31/32<br />

(97%) pts who progressed by increase <strong>of</strong> target lesions continued TKI.<br />

13/16 (81%) pts who progressed by new lesion remained on TKI. Two pts<br />

with PD in non-target lesions discontinued therapy at PD. 5/6 (83%) pts<br />

with both increase <strong>of</strong> target lesions and new lesion at PD continued TKI. In<br />

49 continuing pts, the median time from RECIST PD to termination <strong>of</strong> TKI<br />

was 10.1 mos (range: 2.2-64.2 mos). 15/49 (31%) pts who continued TKI<br />

received additional chemo compared to 0/7 pts who discontinued (Fisher’s<br />

p�0.17). Pts who discontinued therapy (n�7) were significantly younger<br />

(median 48 yrs) than those who continued TKI at PD (median 64 yrs,<br />

Wilcoxon p�0.003). Median OS beyond RECIST PD among those who<br />

continued TKI was 31.8 mos (95% CI 15.9- not reached) and though<br />

underpowered, this did not appear to be impacted by TTP when adjusted in<br />

a Cox model (p�0.84). Conclusions: 88% <strong>of</strong> EFGR-mutant NSCLC pts who<br />

progressed on first-line TKI continued therapy beyond RECIST PD, which is<br />

not the single determining factor for terminating TKI in EGFR-mutant<br />

NSCLC pts. Additional progression criteria specific to this population are<br />

needed to better guide therapeutic decision making.<br />

7555 General Poster Session (Board #46E), Sat, 1:15 PM-5:15 PM<br />

N0821: A phase II first-line study <strong>of</strong> a combination <strong>of</strong> pemetrexed (P),<br />

carboplatin (C), and bevacizumab (B) in elderly patients with good<br />

performance status (PS < 2). Presenting Author: Grace K. Dy, Roswell Park<br />

Cancer Institute, Buffalo, NY<br />

Background: In a retrospective exploratory analysis <strong>of</strong> E4599, patients (pts)<br />

� 70 yo had a higher frequency <strong>of</strong> and more severe toxicities without<br />

apparent survival benefit from the addition <strong>of</strong> B to C�paclitaxel. We<br />

hypothesized that in this pt population, B will have better safety and<br />

efficacy pr<strong>of</strong>ile when used in combination with C�P. Methods: Pts �/� 70<br />

yo with previously untreated stage IIIB/IV (TNM 6th ed) nonsquamous<br />

NSCLC, ECOG PS 0-1, measurable disease and adequate organ function<br />

were eligible. C at AUC 6, P at 500 mg/m2 and B at 15 mg/kg were<br />

administered on day 1 <strong>of</strong> each 21-day cycle for up to 6 cycles followed by<br />

maintenance P�B in patients with CR, PR or SD. The primary endpoint was<br />

6-month progression-free survival (PFS) rate. The treatment would be<br />

considered promising based on a single arm one-stage binomial design if<br />

34 or more successes out <strong>of</strong> 55 patients were observed. This design had an<br />

exact significance level <strong>of</strong> 0.05 at 93% power to detect a true success rate<br />

<strong>of</strong> at least 70%. Polymorphisms in VEGFA, FPGS, GGH, SLC19A1 and<br />

TYMS in germline DNA were correlated with treatment outcome. Results:<br />

58 eligible pts were enrolled; 29 males/29 females. Median age was 75.<br />

Median treatment cycles received was 6. Grade 3 or higher adverse events<br />

(AE) were reported in 49 (85%) pts. There were no treatment-related<br />

deaths. The most common grade 3/4 AEs(regardless <strong>of</strong> attribution) were<br />

hypertension (10%), fatigue (28%), dehydration (9%), neutropenia (43%)<br />

and thrombocytopenia (21%). There were 3 (5%) grade 3/4 hemorrhagic<br />

events. 8 (14%) had grade 4 neutropenia and 3 (5%) had grade 4<br />

thrombocytopenia. Grade 3/4 ischemic/thromboembolic events occurred in<br />

6 pts (10%). Thirty-four out <strong>of</strong> the first 54 (63%, 95% CI: 48.7-75.7%)<br />

evaluable pts met the primary endpoint (4 pts were lost to follow-up prior to<br />

6 months). The confirmed ORR was 37.9% (95% CI: 25.5-51.6%).<br />

Median time to treatment failure was 4.8 months (95% CI: 3.9-6.4).<br />

Median PFS was 7.1 months (95% CI: 5.9-11.7), median OS was 13.7<br />

months (95% CI: 9.4-15.7). Results <strong>of</strong> SNP analysis will be presented.<br />

Conclusions: C�P�B followed by maintenance P�B is an active and<br />

tolerable first-line regimen for elderly patients with good PS.<br />

Lung Cancer—Non-small Cell Metastatic<br />

493s<br />

7554 General Poster Session (Board #46D), Sat, 1:15 PM-5:15 PM<br />

Phase II study <strong>of</strong> pemetrexed (Pem) and cisplatin (Cis) plus cetuximab<br />

(Cet) followed by Pem and Cet maintenance therapy in patients (Pts) with<br />

advanced nonsquamous non-small cell lung cancer (NSCLC). Presenting<br />

Author: Gerald Schmid-Bindert, University Medical Center, Mannheim,<br />

Germany<br />

Background: A prospective, nonrandomized, multicenter study was conducted<br />

to assess the effect <strong>of</strong> adding cet to pem and cis in pts with<br />

advanced nonsquamous NSCLC. Methods: 113 Caucasian performance<br />

status 0-1 pts received 1st line pem (500 mg/m2 ) and cis (75 mg/m2 )on<br />

day 1 (21d cycle) for 4-6 cycles and cet (400 mg/m2 loading dose followed<br />

by 250 mg/m2 ) weekly. Non-progressive pts received pem 500 mg/m2 on<br />

day 1 (21d cycle) plus cet (250mg/m2 weekly) until progression. Pts<br />

received vitamin B12/folic acid and dexamethasone. Primary endpoint was<br />

objective response rate (ORR) (RECIST 1.0). Secondary endpoints were<br />

progression free survival (PFS), 1 year survival rate, translational research<br />

(TR) and safety. Results: Pts’ characteristics: median age 59.7 years, 64%<br />

male, 50% PS 0, 92% stage IV, and 78% adenocarcinoma. All pts<br />

completed � 1 cycle <strong>of</strong> induction therapy and 45% and 43% completed �<br />

1 cycle <strong>of</strong> maintenance with pem and cet, respectively. ORR (n�109) was<br />

38.5% (80% CI 32.2-45.1), all partial responses. Disease control rate<br />

(response/stable disease) was 59.6% (80% CI: 53.1-65.9). Median PFS<br />

was 5.82 months (80% CI: 4.40-6.70). One year survival rate was 0.45<br />

(80% CI: 0.39-0.51). Significant associations were seen between high<br />

EGFR by IHC and increased PFS (cytoplasm: HR�0.46, p�0.035;<br />

membrane: HR�0.41, p�0.008), and between high nuclear TTF-1 and<br />

increased ORR (OR�7.73, p�0.021) / PFS (HR�0.21, p�0.001) / OS<br />

(HR�0.25, p�0.035). Of 113 pts evaluated for safety, 73 (64.6%) pts<br />

had drug related CTC Grade 3/4 adverse events (AE): most frequent were<br />

neutropenia (14.2%), rash (15%), and vomiting (8.8%). Drug related<br />

serious AEs were reported in 27.4% pts: most frequent were anemia<br />

(5.3%), neutropenia (5.3%), vomiting (3.5%), and rash, renal failure,<br />

diarrhea and fatigue (1.8% each). There were 2 potential on-study drug<br />

related deaths (sudden death and large intestinal perforation). Conclusions:<br />

Pem, cis and cet appeared efficacious and tolerable. These results support<br />

further evaluation in a randomized trial. The TR outcomes are hypothesis<br />

generating given the study’s size and nonrandomized nature.<br />

7556 General Poster Session (Board #46F), Sat, 1:15 PM-5:15 PM<br />

Targeting EGFR and ERBB3 in lung cancer patients: <strong>Clinical</strong> outcomes in a<br />

phase I trial <strong>of</strong> MM-121 in combination with erlotinib. Presenting Author:<br />

Lecia V. Sequist, Massachusetts General Hospital Cancer Center, Boston,<br />

MA<br />

Background: Erlotinib is effective in NSCLCs with wild-type EGFR and<br />

shows enhanced benefit in EGFR mutation-positive cancers. However,<br />

resistance invariably develops, <strong>of</strong>ten involving persistent ErbB3 signaling<br />

and activation <strong>of</strong> the PI3K/AKT survival pathway. We present the full results<br />

<strong>of</strong> the Phase 1 study evaluating the safety and tolerability <strong>of</strong> erlotinib plus<br />

MM-121 a fully human IgG2 monoclonal antibody (mAb) to ErbB3.<br />

Methods: Eligible patients had advanced stage NSCLC, and ECOG 0-2.<br />

Seven cohorts were enrolled, evaluating varying levels <strong>of</strong> the MM-121 and<br />

erlotinib, as well as alternate MM-121 infusion schedules. Tumor response<br />

was assessed every 8 weeks. Dose levels were determined by safety and<br />

pharmacokinetic (PK) data, and immunogenicity, efficacy endpoints and<br />

exploratory biomarker evaluations were performed. Results: From February<br />

2010 – July 2011 32pts entered the study (median age 63y; 45% male;<br />

18% ECOG 0, 82% ECOG 1. 56% had adenocarcinoma and 30% pts<br />

received 3 or more lines <strong>of</strong> prior therapies (range 0-7) with 91% having had<br />

prior platinum. 65% had wild-type EGFR status and were never treated or<br />

never responded to EGFR-TKIs (EGFRwt) and 28% had acquired resistance<br />

to erlotinib treatment (EGFRresist). The most common toxicities (any<br />

grade) observed were diarrhea (82%), rash (64%), and fatigue (64%).<br />

DLTs observed in different cohorts were diarrhea, mucositis, rash and<br />

failure to thrive. <strong>Clinical</strong> activity was observed including 1 PR (an EGFR TKI<br />

naïve EGFR mutant) and 14 SD. Average duration <strong>of</strong> disease stabilization<br />

was 21.6 wks (range 7.1 -89.3 wks). Median PFS is 8.1 weeks and the 16<br />

week PFS rate was 41% in the overall population. For EGFRwt and<br />

EGFRresist pts the median PFS were 7.7 weeks and 15.1 weeks, and the<br />

16 week PFS rates were 32% and 44%, respectively. 7/20 EGFRwt pts and<br />

5/9 EGFRresist pts achieved SD. Conclusions: This study suggests that the<br />

combination <strong>of</strong> erlotinib and MM-121 has an acceptable safety pr<strong>of</strong>ile in<br />

heavily pre-treated NSCLC patients with and without EGFR mutations.<br />

Early signals <strong>of</strong> patient benefit were seen and a large randomized phase II<br />

study is ongoing.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


494s Lung Cancer—Non-small Cell Metastatic<br />

7557 General Poster Session (Board #46G), Sat, 1:15 PM-5:15 PM<br />

Interim analysis <strong>of</strong> afatinib monotherapy in patients with metastatic<br />

NSCLC progressing after chemotherapy and erlotinib/gefitinib (E/G) in a<br />

trial <strong>of</strong> afatinib plus paclitaxel versus investigator’s choice chemotherapy<br />

following progression on afatinib monotherapy. Presenting Author: Martin<br />

H. Schuler, Department <strong>of</strong> Medical Oncology, West German Cancer Center,<br />

University Duisburg-Essen, Essen, Germany<br />

Background: The benefit <strong>of</strong> sustained ErbB family blockade in NSCLC<br />

patients with acquired resistance (AR) to EGFR TKIs is unknown. We<br />

investigated afatinib, an irreversible blocker <strong>of</strong> EGFR (ErbB1), HER2<br />

(ErbB2) and ErbB4 receptor tyrosine kinases, in patients with metastatic<br />

NSCLC, who had failed chemotherapy and E/G. Methods: This was a Phase<br />

III, randomized, open-label, multi-center trial. Patients with pathologically<br />

confirmed Stage IIIB/IV metastatic NSCLC after �1 line <strong>of</strong> chemotherapy<br />

who failed E/G received oral afatinib 50 mg until disease progression (<strong>Part</strong><br />

A). After progression, patients with clinical benefit (�12 wks) were eligible<br />

to continue afatinib 40 mg plus paclitaxel or receive investigator’s choice<br />

chemotherapy (<strong>Part</strong> B). Primary endpoint for <strong>Part</strong> A was PFS (RECIST 1.1;<br />

CT scan every 6 wks). Available tumor samples were collected for central<br />

EGFR mutation testing; local mutation data were also collected. An interim<br />

analysis <strong>of</strong> <strong>Part</strong> A, assessing afatinib monotherapy, is reported. Results:<br />

<strong>Part</strong> A enrolled April 2010 through to May 2011; 1154 patients received<br />

afatinib monotherapy. The majority had adenocarcinoma (85%), 57% were<br />

female, 43% were Asian, 54% were never smokers. Best response to prior<br />

E/G was CR (2%), PR (31%), SD (42%) and PD (20%). Median PFS for<br />

afatinib was 3.3 mths; 88 patients (8%) achieved an objective tumor<br />

response, 648 (56%) had SD. For EGFR mutation positive patients (n�49,<br />

centrally confirmed), PFS was 4.2 vs. 2.6 mths for EGFR mutation negative<br />

patients (n�35). When applying clinical enrichment criteria for AR, PFS<br />

was 4.2 mths for those with enrichment (n�597) vs. 2.8 mths for those<br />

without (n� 557; logrank test p�0.0001). The most common grade 3/4<br />

adverse events were diarrhea (17%) and rash/acne (11%). In <strong>Part</strong> A, 99<br />

patients remain on treatment. Conclusions: Afatinib monotherapy provided<br />

a clinically meaningful benefit in this large, treatment-refractory NSCLC<br />

trial, similar to LUX-Lung 1. Those clinically enriched for AR to EGFR TKIs<br />

achieved prolonged disease control upon continued ErbB blockade.<br />

7559 General Poster Session (Board #47A), Sat, 1:15 PM-5:15 PM<br />

Final overall survival and updated biomarker analysis results from the<br />

randomized phase III ICOGEN trial. Presenting Author: Yan Sun, Cancer<br />

Hospital, Chinese Academy <strong>of</strong> Medical Sciences, Beijing, China<br />

Background: A total <strong>of</strong> 399 pretreated patients with advanced NSCLC were<br />

randomly assigned to receive gefitinib or icotinib in the phase III ICOGEN<br />

trial, the first head-to-head phase III trial <strong>of</strong> EGFR-TKIs. The results <strong>of</strong> the<br />

primary endpoint, PFS, have been reported previously. This report represents<br />

the final OS and biomarker analysis results. Methods: EGFR mutation<br />

was evaluated by using Scorpion ARMS (QIAGEN, n�152). Overall survival<br />

was analyzed by Cox proportional-hazards model analysis at 82% maturity.<br />

Results: Median OS was 13.3 months for icotinib and 13.9 months for<br />

gefitinib (hazard ratio [HR] � 0.90; 95% CI, 0.79 to 1.02; P � .109). The<br />

EGFR mutation rate was 43% in the icotinib group and 59% in the gefitinib<br />

group. Compared to wild type patients, patients with EGFR mutation had<br />

longer PFS (median, 6.2m vs. 2.3m; P�.00001) as well as OS (median,<br />

20.5m vs. 7.7m; P�.00001). There were no significant differences in PFS<br />

or OS between the two treatment groups in EGFR mutation-positive<br />

subgroup (median PFS, 7.8m vs. 5.3m for icotinib and gefitinib, respectively,<br />

P �.3162; median OS, 20.9m vs. 20.2m for icotinib and gefitinib,<br />

respectively, P �.7611.) or in EGFR mutation-negative subgroup (median<br />

PFS, 2.3m vs. 2.2m for icotinib and gefitinib, respectively, P �.1531;<br />

median OS, 7.8m vs. 6.9m for icotinib and gefitinib, respectively, P<br />

�.7885.). Conclusions: There is no statistically significant difference<br />

between icotinib and gefitinib in PFS or OS when given to NSCLC patients.<br />

This suggests that icotinib can provide similar OS benefits to gefitinib in<br />

advanced NSCLC patients. Moreover, EGFR mutation status is the strongest<br />

predictor in identifying which patients are most likely to benefit from<br />

icotinib.<br />

7558 General Poster Session (Board #46H), Sat, 1:15 PM-5:15 PM<br />

Afatinib monotherapy in patients with metastatic squamous cell carcinoma<br />

<strong>of</strong> the lung progressing after erlotinib/gefitinib (E/G) and chemotherapy:<br />

Interim subset analysis from a phase III trial. Presenting Author: Joo-Hang<br />

Kim, Yonsei Cancer Center, Yonsei University College <strong>of</strong> Medicine, Seoul,<br />

South Korea<br />

Background: Patients with squamous NSCLC have limited treatment options.<br />

For those deriving benefit from EGFR TKIs, it is unclear whether<br />

sustained ErbB family blockade <strong>of</strong>fers benefit upon progression. We<br />

evaluated afatinib, an irreversible blocker <strong>of</strong> EGFR (ErbB1), HER2 (ErbB2)<br />

and ErbB4 receptor tyrosine kinases, in patients with metastatic NSCLC<br />

who had failed chemotherapy and E/G. Here we describe a pre-specified<br />

analysis <strong>of</strong> those with squamous histology in <strong>Part</strong> A. Methods: This<br />

randomized Phase III, open-label, multi-center trial enrolled patients with<br />

pathologically confirmed metastatic NSCLC after failing �1 line <strong>of</strong> cytotoxic<br />

chemotherapy and E/G. In <strong>Part</strong> A, patients received oral afatinib 50<br />

mg until disease progression. Those with clinical benefit (�12 wks) who<br />

progressed were eligible to receive afatinib plus paclitaxel or investigator’s<br />

choice chemotherapy (<strong>Part</strong> B). Primary endpoint was PFS (RECIST 1.1).<br />

Following an amendment, an interim analysis <strong>of</strong> <strong>Part</strong> A was performed to<br />

assess afatinib monotherapy. Results: Patient enrolment into <strong>Part</strong> A was<br />

from April 2010 to May 2011. Of 1154 afatinib-treated patients, 91 (8%)<br />

had squamous histology; 18/91 and 40/91 had CR/PR and SD on prior E/G,<br />

respectively (by investigator). Median age was 63 yrs, 71% were male, 76%<br />

were current/ex-smokers. Median PFS on afatinib was 3.7 mths in the<br />

squamous histology subset. Of 91 patients, 42 had PFS �3 mths; 13 had<br />

PFS <strong>of</strong> �6 mths. In evaluable patients (n�77), 1 CR and 3 PRs were<br />

confirmed; 51 and 22 patients had best overall response <strong>of</strong> SD and PD,<br />

respectively. Of the 31 patients with PD on prior E/G with no intervening<br />

chemotherapy, 10 achieved confirmed disease control (2 PR; 8 SD) on<br />

afatinib. Most commonly reported grade 3/4 adverse events (AEs) in <strong>Part</strong> A<br />

were diarrhea (13%) and rash/acne (12%). The safety pr<strong>of</strong>ile in the<br />

squamous histology subset was similar to that observed for the whole trial.<br />

Conclusions: Afatinib monotherapy demonstrated encouraging activity in<br />

treatment-refractory NSCLC patients with squamous histology that merits<br />

further evaluation.<br />

7560 General Poster Session (Board #47B), Sat, 1:15 PM-5:15 PM<br />

A randomized phase III study <strong>of</strong> a cisplatin (CDDP) and docetaxel (DOC)<br />

with or without irinotecan (CPT) in pts with advanced NSCLC: Okayama<br />

Lung Cancer Study Group OLCSG 0403 trial. Presenting Author: Toshiaki<br />

Okada, Chugoku Central Hospital, Fukuyama, Japan<br />

Background: CDDP-based doublet chemotherapy provides a significant but<br />

modest survival prolongation in untreated advanced NSCLC with MST <strong>of</strong><br />

around 12 months. Based on the favorable efficacy pr<strong>of</strong>ile in our previous<br />

phase II trial <strong>of</strong> triplet chemotherapy with CDDP, DOC, and CPT (response<br />

rate: 57.1%, MST: 17 months) (JTO 2007), we conducted a phase III study<br />

to compare this regimen with CDDP and DOC doublet therapy in the<br />

first-line setting. Methods: Pts were randomly allocated to the triplet<br />

therapy (A arm; CDDP 60 mg/m2 , day 1; DOC 60 mg/m2 , day 1; and CPT 60<br />

mg/m2 , day 2; q3 wks, determined based on our phase I study result) or a<br />

standard doublet chemotherapy (B arm; CDDP 80 mg/m2 , and DOC 60<br />

mg/m2 , day 1; q3 wks). The primary endpoint was OS whilst secondary<br />

endpoints included response rates, PFS, and toxicity. Results: Between<br />

2004 and 2009, 120 pts were enrolled (A/B: 60 pts each). Objective<br />

response was comparable between the arms (18 pts [30%] each, p �<br />

0.571). As for toxicity, grade 3-4 neutropenia, principal toxicity, was<br />

observed in 93% vs. 58% (p � 0.001). Although grade 3 febrile<br />

neutropenia was observed in 53% and 18% <strong>of</strong> the pts (p � 0.001), none <strong>of</strong><br />

whom further developed toxic deaths. At a median follow-up time <strong>of</strong> 64.2<br />

months, median PFS time and 1-year PFS rate were 5.3 vs. 4.3 months and<br />

13.3% vs. 11.7%, respectively (stratified logrank test; p � 0.872). There<br />

was also no difference in OS (MST, 16.8 vs. 12.2 months; 1-year OS rate,<br />

68.0% vs. 53.0%; stratified logrank test; p � 0.846). Subgroup analysis<br />

for OS showed that never smokers (HR � 0.366; 95% CI � 0.072-1.866)<br />

tended to benefit from the triplet chemotherapy compared with eversmokers<br />

(HR � 1.380; 95% CI � 0.771-2.473) despite no statistical<br />

significance (p for interaction � 0.150). Similar trend in interaction<br />

between treatment effect and smoking status was also observed in PFS.<br />

Conclusions: This triplet chemotherapy failed to produce a significant<br />

antitumor activity as compared with the standard doublet therapy.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


7561 General Poster Session (Board #47C), Sat, 1:15 PM-5:15 PM<br />

A phase II study <strong>of</strong> erlotinib monotherpay in patients with previously treated<br />

advanced non-small cell lung cancer (NSCLC) without EGFR gene mutation<br />

who have never/light smoking history: NEJ006/TCOG0903. Presenting<br />

Author: Yoshiki Ishii, Dokkyo Medical University School <strong>of</strong> Medicine,<br />

Shimotoga, Japan<br />

Background: Epidermal growth factor receptor (EGFR) tyrosine kinase<br />

inhibitors (TKIs) have demonstrated good response in NSCLC harboring<br />

EGFR gene mutation. However, survival benefit from erlotinib was observed<br />

also in NSCLC patients with wild type (wt) EGFR gene in subsets <strong>of</strong> several<br />

phase III trials. Additionally, smoking status could be a predictive factor <strong>of</strong><br />

erlotinib efficacy, This study aimed to evaluate the efficacy <strong>of</strong> erlotinib in<br />

Japanese NSCLC patients with wt-EGFR and find a biomarker that predicts<br />

the efficacy <strong>of</strong> erlotinib other than EGFR gene mutation. Methods: The<br />

primary endpoint was an objective response rate. Secondary endpoints<br />

included disease control rate, overall survival, and safety. Advanced NSCLC<br />

patients without EGFR gene mutation who had received one to three prior<br />

chemotherapy regimens and who had never smoked or light smoked<br />

(Brinkman Index: less than200) were eligible in this study. EGFR gene<br />

status was evaluated by the PNA-LNA PCR clamp method. Erlotinib was<br />

administered daily (150mg/day) until disease progression or unacceptable<br />

toxicities. Results: Forty seven patients were enrolled between March 2010<br />

and November 2011. One patient was excluded for evaluation because<br />

having mutation <strong>of</strong> EGFR. Efficacy and safety were evaluated among 46<br />

patients. Best responses were PR 7 (15.2%), SD 12 (26.1%), PD 26<br />

(46.4%), NE 1 (2.2%). Response rate and disease control rate were 15.2%<br />

(95%CI: 4.9-25.5%) and 41.3 % (95%CI 27.1-55.5%) respectively.<br />

Grade 3 or 4 adverse events were anorexia (4), skin rash (2), neutropenia<br />

(1), leukopenia (1), anemia (2), elevation <strong>of</strong> AST/ALT (1), rectal ulcer (1),<br />

and cerebral infarction (1).Two patients suffered grade 3 interstitial lung<br />

disease. Conclusions: This is the first report to evaluate Erlotinib efficacy in<br />

selected NSCLC who do not possess EGFR gene mutation. Erlotinib showed<br />

significant anti-tumor activity in pretreated never or light smoked Japanese<br />

NSCLC patients without EGFR gene mutation. The results <strong>of</strong> biomakr<br />

analysis will be presented at the meeting.<br />

7564 General Poster Session (Board #47F), Sat, 1:15 PM-5:15 PM<br />

Elderly and younger patients with non-small cell lung cancer (NSCLC)<br />

derive similar benefit from second-line chemotherapy: A retrospective<br />

subset analysis <strong>of</strong> second-line chemotherapy trials conducted from the<br />

Hellenic Cooperative Research Group (HORG). Presenting Author: S<strong>of</strong>ia<br />

Agelaki, Department <strong>of</strong> Medical Oncology, University General Hospital <strong>of</strong><br />

Heraklion, Heraklion, Greece<br />

Background: Elderly patients (pts) achieve a similar survival benefit, with<br />

acceptable toxicity, from first-line chemotherapy for the treatment <strong>of</strong><br />

advanced NSCLC compared with their younger counterparts. There have<br />

been no second-line trials specifically designed for elderly pts and few data<br />

exist on the efficacy and tolerability <strong>of</strong> second-line therapy in this<br />

population. Moreover, little if any information exists on the frequency <strong>of</strong><br />

administration <strong>of</strong> second-line chemotherapy in these pts. Methods: The files<br />

<strong>of</strong> 2004 pts with advanced NSCLC enrolled into first-line chemotherapy<br />

trials performed by HORG from 1995 to 2007 were reviewed. A total <strong>of</strong> 600<br />

pts who received second-line chemotherapy within the context <strong>of</strong> clinical<br />

trials were identified. Patients’ data were analysed for efficacy and toxicity<br />

according to age. Results: Second-line chemotherapy was administered in<br />

24% and 34% <strong>of</strong> pts �65 and �65 years old after failure <strong>of</strong> prior therapy<br />

(p�0.0001). A total <strong>of</strong> 219 (24.8%) <strong>of</strong> 600 pts who received second-line<br />

treatment were �65 years old (median age 70 yrs, range 65-82). Response<br />

rates to second-line therapy were 11.9% for older pts compared to 12.3%<br />

for younger pts (p�ns). TTP was 2.8 and 3.1 months for older and younger<br />

pts, respectively (p�ns). Elderly pts receiving second-line chemotherapy<br />

had a median survival <strong>of</strong> 7.7 months compared with 8.2 months for younger<br />

pts (p�ns). Similar rates <strong>of</strong> haematological and non-haematological<br />

toxicities were encountered between the two groups. Conclusions: The<br />

participation <strong>of</strong> elderly pts to second-line chemotherapy trials was lower<br />

compared to younger patients. There was no significant difference in<br />

outcome or toxicity between elderly and younger pts. For elderly pts with<br />

advanced NSCLC and good performance status, second-line chemotherapy<br />

is appropriate. However, specific second-line trials in older pts are required<br />

since those included in the current analysis were probably highly selected<br />

to fit the inclusion criteria.<br />

Lung Cancer—Non-small Cell Metastatic<br />

495s<br />

7563 General Poster Session (Board #47E), Sat, 1:15 PM-5:15 PM<br />

First-line gefitinib for elderly patients with advanced non-small cell lung<br />

cancer (NSCLC) harboring EGFR mutations: A combined analysis <strong>of</strong> NEJ<br />

studies. Presenting Author: Sodai Narumi, Department <strong>of</strong> Respiratory<br />

Medicine, Tohoku University Graduate school <strong>of</strong> Medicine, Sendai, Japan<br />

Background: The first-line treatment with gefitinib has been established as a<br />

standard <strong>of</strong> care for advanced NSCLC patients with EGFR mutation by<br />

previous studies including NEJ002. Since the percentage <strong>of</strong> elderly<br />

population in those studies was lower than that <strong>of</strong> real situation, further<br />

validation <strong>of</strong> its usefulness for elderly patients with EGFR-mutated NSCLC<br />

is warranted. Methods: The efficacy and safety data <strong>of</strong> fit elderly patients<br />

(70 years or older with PS 0-2) with EGFR-mutated NSCLC who received<br />

the first-line gefitinib in NEJ001 (phase 2), NEJ002 (phase 3), and<br />

NEJ003 (phase 2) were combined. Same population treated with the<br />

first-line carboplatin plus paclitaxel in NEJ002 (n�34) were also examined<br />

their efficacy and safety for reference. Regarding the toxicity and quality <strong>of</strong><br />

life (QOL), the comparison between elderly patients and younger patients<br />

(� 70 years old) both treated with first-line gefitinib in NEJ002 was also<br />

performed. Results: Data from 71 patients (7 from NEJ001, 33 from<br />

NEJ002, 31 from NEJ003) treated with first-line gefitinib were combined.<br />

Patients’ characteristics were as follows; mean age: 76.8 years (range<br />

70-89), female: 73%, never smoker: 75%, PS 0-1: 92%. Overall response<br />

rate (73.2%) and median progression-free survival (14.3 months) in this<br />

group were significantly better than those in the reference group (26.5%<br />

and 5.7 months, respectively). Despite fewer patients received second-line<br />

chemotherapy in the first-line gefitinib group, its median overall survival<br />

(30.8 months) was not inferior to that <strong>of</strong> reference group (26.4 months).<br />

Regarding toxicities such as liver dysfunction, rash, diarrhea, and pneumonitis,<br />

and QOL examined by self questionnaires, there were no difference<br />

between the elderly patients and younger patients in NEJ002. Conclusions:<br />

First-line gefitinib still achieved high efficacy with acceptable toxicity in fit<br />

elderly patients with advanced NSCLC harboring EGFR mutation. Considering<br />

that elderly patients tend to receive fewer treatment lines compared<br />

with younger patients, first-line gefitinib would be strongly recommended<br />

for this population to avoid the risk <strong>of</strong> missing its administration.<br />

7565 General Poster Session (Board #47G), Sat, 1:15 PM-5:15 PM<br />

Randomized phase II trial <strong>of</strong> carboplatin combined with weekly paclitaxel<br />

(CP) and docetaxel alone (D) in elderly patients (pts) with advanced<br />

non-small cell lung cancer (NSCLC): NJLCG 0801. Presenting Author:<br />

Shunichi Sugawara, Sendai Kousei Hospital, Sendai, Japan<br />

Background: Standard first-line chemotherapy for elderly NSCLC pts has<br />

been considered as a monotherapy with vinorelbine or gemcitabine globally.<br />

However, we have demonstrated the high efficacy <strong>of</strong> CP for elderly pts<br />

in our previous trial (Ann Oncol 2010). Meanwhile, D has been considered<br />

as an alternative option for this population in Japan according to the result<br />

<strong>of</strong> WJTOG9904 (JCO 2006). Thus we compared the two regimens to select<br />

the proper candidate for future phase III trial. Methods: Eligible pts were<br />

aged 70 years or older with newly diagnosed stage IIIB/IV NSCLC; ECOG<br />

performance status 0-1; adequate organ function; written informed consent.<br />

Pts were randomized to receive carboplatin (AUC 6) on day 1 and<br />

paclitaxel (70mg/m2 on day 1, 8, and 15) every 4 weeks or D (60mg/m2 on<br />

day 1) every 3 weeks. The primary endpoint was overall response rate<br />

(ORR), and secondary endpoints were progression-free survival (PFS),<br />

overall survival, and toxicity pr<strong>of</strong>ile. Assuming that ORR <strong>of</strong> 40% would be<br />

potential usefulness while ORR <strong>of</strong> 20% would be the lower limit <strong>of</strong> interest,<br />

40 pts in each arm were required if expect 10% loss to follow up. Results:<br />

Between July 2006 and September 2010, 84 pts were enrolled and 41 pts<br />

in CP arm and 42 pts in D arm were eligible (median age, 76 years; 75%<br />

male; 72% stage IV). Median treatment cycle was 4 in each arm (CP, range<br />

1-6; D, range 1-8). ORRs were 51% (95%CI: 36-66%) and 26% (95%CI:<br />

12-39%) in the CP and D arm, respectively. With a median follow-up <strong>of</strong><br />

18.4 months, median PFS were 6.5 and 3.9 months in the CP and D arm,<br />

respectively (Logrank, P�0.0027). Grade 3 or severer toxicities were as<br />

follows: neutropenia (CP, 56% and D, 79%), anemia (CP, 15% and D,<br />

7%), thrombocytopenia (CP, 10% and D, 0%), infection (CP, 20% and D,<br />

25%). One treatment-related death due to neutropenia, pneumonia, and<br />

lethal arrhythmia occurred in D arm but none in CP arm. Conclusions: The<br />

platinum doublet CP achieved higher activity with an acceptable toxicity<br />

pr<strong>of</strong>ile for elderly pts with advanced NSCLC compared to monotherapy with<br />

D. The superiority <strong>of</strong> CP to the monotherapy in this trial is consistent with<br />

results <strong>of</strong> recent IFCT-0501 trial (Lancet 2011).<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


496s Lung Cancer—Non-small Cell Metastatic<br />

7566 General Poster Session (Board #47H), Sat, 1:15 PM-5:15 PM<br />

Rare and complex mutations <strong>of</strong> epidermal growth factor receptor (EGFR)<br />

and efficacy <strong>of</strong> tyrosine kinase inhibitor (TKI) in patients with non-small<br />

cell lung cancer (NSCLC). Presenting Author: Bhumsuk Keam, Department<br />

<strong>of</strong> Internal Medicine, Seoul National University Hospital, Seoul, South<br />

Korea<br />

Background: Other than in-frame deletional mutation (MT) in exon 19 and<br />

Leu858Arg point MT in EGFR gene, there are many rare and complex MT.<br />

The purpose <strong>of</strong> this study was to investigate was to clarify the clinical<br />

significance <strong>of</strong> rare and complex MT, and the efficacy <strong>of</strong> EGFR TKI in these<br />

patients. Methods: Rare MTs were defined any MT other than deletional MT<br />

in exon 19, Leu858Arg in exon 21, and Thr790Met in exon 20. Complex<br />

MT was defined two different EGFR MTs co-occurring within the single<br />

tumor sample. We have analyzed consecutive database <strong>of</strong> NSCLC patients<br />

who were treated with either gefitinib or erlotinib at Seoul National<br />

University Hospital between Jan. 2002 and Dec. 2011. For analysis <strong>of</strong><br />

EGFR MT, coding sequences from exon 18 to 21 were amplified by nested<br />

PCR and subjected to direct sequencing methods. Results: Among the<br />

1,159 TKI treated patients, 301 patients had EGFR MT (177 patients<br />

(58.8%) with del-19, 104 patients (34.6%) with Leu858Arg, and 20<br />

patients (6.6%) with rare MT). Twenty-three (7.6%) patients have complex<br />

MT, and 55 (18.3%) patients have Gln787Gln polymorphism particularly<br />

associated with Leu858Arg MT. Identified rare MT were follows; exon 18:<br />

Leu692Phe, Glu707Lys, Glu709Gly, Lys714Asn, Gly7Ala, Thr725Thr,<br />

exon 19: Ala755Asp Lys737Thr, exon 20: Val786Met, exon21: Ala871Gly,<br />

Gly873Gln, Leu833Val, Val834Leu, Arg836Cys, Pro848Leu, Ala859Thr,<br />

Leu861Gln. When categorizing 3 groups (group A: classic MT alone, group<br />

B: complex MT with classic � rare MT, group C: rare MT alone or complex<br />

MT with rare MTs), response rate (RR) to TKI was different between each<br />

groups (RR� 78.7% in group A vs. 69.2% in group B vs. 38.5% in group C,<br />

respectively, P � 0.001). Progression-free survival (PFS) was also poorer in<br />

rare MT (median PFS: 12.1 mo vs. 7.6 mo vs. 2.1 mo, respectively, P�<br />

0.020,). Gln787Gln polymorphism <strong>of</strong> exon 21 was not associated with RR<br />

or PFS (P� 0.101, P� 146, respectively) Conclusions: In this large case<br />

series, NSCLC patients who harboring rare MT other than del-19 and<br />

Leu858Arg, does not seems response to EGFR TKI. However, complex MT<br />

with the classical MT showed similar treatment efficacy toward EGFR TKI<br />

with that <strong>of</strong> classical MT alone.<br />

7569 General Poster Session (Board #48C), Sat, 1:15 PM-5:15 PM<br />

Phase I/II study <strong>of</strong> amrubicin and nedaplatin in patients with untreated,<br />

advanced non-small cell lung cancer. Presenting Author: Hiroaki Senju,<br />

National Nagasaki Medical Center, Ohmura, Japan<br />

Background: We conducted a phase I/II study <strong>of</strong> combination chemotherapy<br />

with nedaplatin (CDGP) and amrubicin (Amr) for patients with untreated,<br />

advanced non small-cell lung cancer (NSCLC). Methods: Eligible patients<br />

were having adequate organ function and PS <strong>of</strong> 0-1. CDGP was given on day<br />

1 and amrubicin on days 1, 2 and 3. The treatment was repeated every 3<br />

weeks. We fixed the dose <strong>of</strong> CDGP as 100 mg/m2, and escalated the dose <strong>of</strong><br />

amrubicin from a starting dose <strong>of</strong> 25 mg/m2 by 5mg/m2 per each levels<br />

until the maximum tolerated dose (MTD). The MTD was defined as the dose<br />

level at which at least two <strong>of</strong> three or two <strong>of</strong> six patients experienced a<br />

dose-limiting toxicity (DLT). Results: Between June 2009 and May 2011,<br />

36 patients were enrolled. In the phase I study, two DLTs occurred in six<br />

patients at level 2; dose level 1 was therefore recommended (25 mg/m2<br />

Amr, 100mg/m2 CDGP). DLTs included cerebral infarction and grade 4<br />

thrombocytopenia. In the phase II study, including phase I study, a total <strong>of</strong><br />

36 patients were enrolled and 132 cycles <strong>of</strong> chemotherapy were conducted.<br />

Grade 3 or 4 neutropenia, grade 3 anemia and grade 3 or 4<br />

thrombocytopenia occurred in 75%, 16.6% and 19.4% in all cycles,<br />

respectively. Febrile neutropenia occurred in 4cycles (3%) but all <strong>of</strong> them<br />

were controllable. Eighteen patients achieved a partial response and the<br />

overall response rate was 51.4%. Conclusions: Combination <strong>of</strong> CDGP and<br />

Amr was highly effective and well tolerable in patients with untreated,<br />

advanced NSCLC.<br />

7568 General Poster Session (Board #48B), Sat, 1:15 PM-5:15 PM<br />

A phase II study <strong>of</strong> erlotinib followed by chemotherapy on progression in an<br />

unselected population <strong>of</strong> advanced non-small cell lung cancer (NSCLC)<br />

patients. Presenting Author: Leora Horn, Vanderbilt-Ingram Cancer Center,<br />

Nashville, TN<br />

Background: Erlotinib is now accepted as first line therapy for advanced<br />

NSCLC patients with EGFR mutated tumors. However, some patients with<br />

EGFR wild-type tumors may benefit as well, and there are no accepted<br />

biomarkers to define this subset <strong>of</strong> patients or to define which patients<br />

benefit from chemotherapy. We conducted a single arm phase II trial <strong>of</strong><br />

erlotinib followed by chemotherapy on progression (carboplatin � paclitaxel<br />

�/- bevacizumab) in treatment naïve patients with stage IV NSCLC.<br />

Methods: In this multicenter prospective phase II trial, patients with stage<br />

IV NSCLC were eligible for enrollment. Blood, frozen tissue and FFPE<br />

biopsies for molecular analysis were mandatory at the time <strong>of</strong> study entry<br />

and optional at the time <strong>of</strong> progression on erlotinib. Additional eligibility<br />

included ECOG performance status (PS) 0-2, measurable disease, and<br />

adequate marrow, renal and hepatic function. Patients with stable treated<br />

brain metastases were allowed. Patients were treated with erlotinib 150 mg<br />

daily until disease progression at which time they underwent a biopsy and<br />

were switched to carboplatin/paclitaxel �/- bevacizumab. Disease status<br />

was assessed after 4 weeks <strong>of</strong> erlotinib to detect early progression. Results:<br />

From 10/07 to 06/11, 127 patients were enrolled: 59% male, 90%<br />

Caucasian, 6% African <strong>American</strong>, 4% Asian, 30% never smokers. 7<br />

patients failed screening. Among evaluable patients on erlotinib, 13% had<br />

PR, 48% SD, and 27% PD. Median PFS on erlotinib was 4.7 months (95%<br />

CI 3.5 to 8.3 months). In patients who went on to receive chemotherapy on<br />

protocol, 24% had PR, 27% SD, and 20% PD. In patients who went on to<br />

receive chemotherapy on protocol median progression-free survival (PFS)<br />

on chemotherapy was 8.1 months (95% CI 5.5 to 10.9 months). Among<br />

the 55 patients who did not receive chemotherapy, 23 had a decline in PS,<br />

14 patients refused, 2 patients enrolled on another protocol, and 6 patients<br />

had other therapy. Conclusions: This is the first study to report on PFS in<br />

unselected NSCLC patients following first line therapy with erlotinib.<br />

Translational studies on this large prospectively collected cohort <strong>of</strong> tissue<br />

samples prior to initiation <strong>of</strong> erlotinib are ongoing.<br />

7570 General Poster Session (Board #48D), Sat, 1:15 PM-5:15 PM<br />

Older patient participation in SWOG lung cancer trials: Comparative<br />

analysis from 1993 to 2008. Presenting Author: Paul Joseph Hesketh,<br />

Lahey Clinic, Tufts University School <strong>of</strong> Medicine, Burlington, MA<br />

Background: In 1999, SWOG published a study demonstrating the underrepresentation<br />

<strong>of</strong> patients 65 years and older on clinical trials (Hutchins<br />

NEJM). A second report published in 2006 demonstrated increasing<br />

proportions <strong>of</strong> patients � 65 years (31% 1997-2000; 38% 2001-2003)<br />

being enrolled into SWOG trials (Unger JCO). Older patient enrollment was<br />

still disproportionately low compared to the US cancer population. The<br />

current analysis focuses on patients with lung cancer from 1993-2008.<br />

Methods: The proportions <strong>of</strong> enrollment onto SWOG lung cancer treatment<br />

trials by age (65-69, 70-79, �80 years) and gender were computed for<br />

4-year time intervals between 1993 and 2008; corresponding rates in the<br />

US were derived from US Census and National Cancer Institute SEER data.<br />

Proportions in the SWOG trials were compared to the SEER proportions<br />

using a 1-sample binomial test <strong>of</strong> proportions. Time trends within SWOG<br />

were evaluated using linear regression. Results: The proportion <strong>of</strong> patients<br />

65-69 was significantly higher than the US population between 1993 and<br />

2004, but was not significantly different after 2004. Proportions <strong>of</strong><br />

patients on SWOG trials 70-79 years old and � 80 were significantly<br />

smaller than the US population. Females were underrepresented from<br />

1993-2004. Between 2005 and 2008 female enrollment was not significantly<br />

different from the US population. Conclusions: Currently, the<br />

proportion <strong>of</strong> patients 65-69 years <strong>of</strong> age and <strong>of</strong> female gender enrolled in<br />

SWOG trials is representative <strong>of</strong> the general lung cancer population.<br />

Although some progress has been made in increasing trial participation <strong>of</strong><br />

patients � 70, enrollment remains disproportionately low. The disparity is<br />

most evident in patients � 80. Continued efforts are needed to increase<br />

older patient participation in lung cancer trials. Supported in part by PHS<br />

Cooperative Agreement grants awarded by the National Cancer Institute:<br />

DHHS, CA32102 and CA38926.<br />

1993-1996 1997-2000 2001-2004 2005-2008<br />

SEER SWOG SEER SWOG SEER SWOG SEER SWOG<br />

Age 65-69 16% 20%� 16% 19%� 16% 19%� 16% 17%<br />

Age 70-79 34% 18%� 35% 18%� 36% 24%� 37% 23%�<br />

Age > 80 16% 1%� 17% 1%� 18% 3%� 20% 4%�<br />

Female gender** 42% 34%� 45% 39%� 47% 39%� 49% 46%<br />

� p � -.01; ** time trend p�0.05.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


7571 General Poster Session (Board #48E), Sat, 1:15 PM-5:15 PM<br />

Phase II study <strong>of</strong> amrubicin (AMR) for patients (pts) with non-small cell<br />

lung cancer (NSCLC) as third-line or fourth-line chemotherapy: Hokkaido<br />

Lung Cancer <strong>Clinical</strong> Study Group trial (HOT) 0901. Presenting Author:<br />

Toshiyuki Harada, Hokkaido Social Insurance Hospital, Sapporo, Japan<br />

Background: Although an increasing number <strong>of</strong> NSCLC pts receive thirdline<br />

chemotherapy with the established benefit <strong>of</strong> second-line chemotherapy,<br />

the role <strong>of</strong> cytotoxic agent in this setting has not yet been well<br />

defined prospectively. AMR, third-generation synthetic anthracycline agent,<br />

has found favorable clinical activity and acceptable toxicity for NSCLC as<br />

well as small cell lung cancer. This prospective trial was conducted to<br />

evaluate the efficacy and safety <strong>of</strong> AMR for NSCLC pts as third-line or<br />

fourth-line chemotherapy. Methods: Eligible pts had a performance status 0<br />

to 2, failure <strong>of</strong> second-line or third-line chemotherapy, and adequate organ<br />

function. Pts received AMR 35 mg/m2 intravenously on days 1-3 every 3<br />

weeks. The primary endpoint was disease control rate (DCR: CR � PR �<br />

SD). Secondary endpoints were overall survival (OS), progression-free<br />

survival (PFS), response rate (CR � PR), and toxicity pr<strong>of</strong>ile. The estimated<br />

accrual was 37 pts to confirm a DCR <strong>of</strong> 50% as desirable target level and a<br />

DCR <strong>of</strong> 30 % as uninteresting with alpha � 0.05 and beta � 0.20. Results:<br />

From August 2009 to May 2011, 41 pts were enrolled from 10 institutions.<br />

Patient characteristics were: male/female 29/12; median age 66 (range<br />

43–74); performance status 0/1/2 16/24/1; adenocarcinoma/squamous<br />

cell carcinoma/large cell carcinoma/not other specified 30/8/2/1; EGFR<br />

mutation positive/negative/unknown 7/26/8; treatment lines 3rd/4th 26/<br />

15. The median number <strong>of</strong> treatment cycles was 2 (range 1-9). The<br />

objective responses were CR 0, PR 4, SD 22, PD 14, and NE 1, giving a<br />

DCR <strong>of</strong> 61.0% (95% CI, 46.0-75.9%). Overall response rate was 9.8%<br />

(95% CI, 0.6-18.8%). Median PFS was 2.6 months, whereas median<br />

survival time was not reached. Grade 3/4 hematological toxicities were<br />

neutropenia (68%), anemia (12%), thrombocytopenia (12%), and febrile<br />

neutropenia (17%). Grade 3/4 non-hematological toxicities were anorexia<br />

(12%), nausea (10%), and pneumonitis (2%). No treatment-related death<br />

was observed. Conclusions: AMR shows significant clinical activity with<br />

acceptable toxicities as third-line or fourth-line chemotherapyfor advanced<br />

NSCLC.<br />

7573 General Poster Session (Board #48G), Sat, 1:15 PM-5:15 PM<br />

Cutaneous toxicity secondary to erlotinib therapy in patients with non-small<br />

cell lung cancer in the NCIC CTG BR.21 study: Time course and correlation<br />

with survival. Presenting Author: Roman Perez-Soler, Montefiore Medical<br />

Center/Albert Einstein College <strong>of</strong> Medicine, Bronx, NY<br />

Background: Cutaneous rash <strong>of</strong>ten develops in erlotinib-treated patients<br />

(pts) and is thought to be associated with improved response and survival.<br />

This analysis focuses on incidence, severity, and time course <strong>of</strong> skin rash<br />

secondary to erlotinib, as well as the correlation between rash development<br />

and overall survival (OS) in pts in the NCIC CTG BR.21 (NCT00036647)<br />

trial. Methods: Pts with stage IIIB or IV non–small cell lung cancer (NSCLC)<br />

who had failed first- or second-line chemotherapy were randomized 2:1 to<br />

erlotinib or placebo. Cutaneous toxicity was graded per NCI CTC v2.0 and<br />

managed at the discretion <strong>of</strong> the treating investigator. This retrospective<br />

analysis used a landmark approach, dividing pts into rash and no-rash<br />

groups, by the appearance <strong>of</strong> rash by week 10. Results: A total <strong>of</strong> 366 <strong>of</strong><br />

488 erlotinib-treated pts (75%) and 40 <strong>of</strong> 243 placebo-treated pts (16%)<br />

developed rash at any time point. Of the former, 75% <strong>of</strong> rash episodes<br />

occurred by week 2. The incidence <strong>of</strong> grades 1 and 2 rash peaked at week<br />

3; grade 3 rash peaked at week 5. The erlotinib dose was reduced in 48 pts<br />

(10%) due to rash. In most erlotinib-treated pts with rash, severity<br />

decreased or plateaued over time (Table). Some 311 erlotinib-treated pts<br />

(rash group) developed rash by week 10. Median OS in the erlotinib-treated<br />

rash (n�311) and no-rash (n�65) groups were 37.4 and 11.1 weeks,<br />

respectively (hazard ratio�0.51 [95% confidence interval, 0.38–0.68];<br />

P�.0001). Baseline factors significantly associated with prolonged OS in<br />

the rash population included race (Asian vs white), number <strong>of</strong> affected<br />

organs (�3 vs�3), and smoking status (never vs ever). Conclusions: For<br />

most pts, rash secondary to erlotinib presented within the first 2 weeks,<br />

peaked during weeks 3–5, decreased thereafter, and did not necessitate<br />

dose reduction, thus supporting SATURN results (Perez-Soler, ASCO<br />

2011, abst. 7610). Development <strong>of</strong> rash by week 10 <strong>of</strong> erlotinib therapy<br />

was associated with significantly improved OS.<br />

Initial grade n*<br />

Lesser<br />

Grade at last assessment, no. (%)<br />

Same Greater<br />

1 207 – 175 (85) 32 (15)<br />

2 125 51 (41) 71 (57) 3 (2)<br />

3 19 12 (63) 6 (32) 1 (5)<br />

4 2 0 (0) 2 (100) 0 (0)<br />

* Of 353 pts with rash grade levels evaluated over time.<br />

Lung Cancer—Non-small Cell Metastatic<br />

497s<br />

7572 General Poster Session (Board #48F), Sat, 1:15 PM-5:15 PM<br />

Treatment with EGFR tyrosine kinase inhibitors beyond progression in<br />

long-term responders to erlotinib in advanced non-small cell lung cancer: A<br />

case-control study <strong>of</strong> overall survival. Presenting Author: Martin Faehling,<br />

Klinik für Kardiologie und Pneumologie, Klinikum Esslingen, Esslingen,<br />

Germany<br />

Background: EGFR-tyrosine kinase inhibitor (TKI) such as erlotinib lead to<br />

prolonged disease stabilization in some patients with advanced NSCLC. It<br />

is so far not clear how to treat patients who progress after prolonged<br />

response to erlotinib. TKI therapy beyond progression with added chemotherapy,<br />

radiotherapy or best supportive care (BSC) may improve survival<br />

compared to chemotherapy, radiotherapy or BSC alone. Methods: We<br />

retrospectively analyzed all NSCLC patients treated with erlotinib at our<br />

institutions since 2004 who progressed after at least stable disease on<br />

erlotinib for at least six months (n�41). Twenty-seven patients were<br />

treated with TKI beyond progression (TKI patients), <strong>of</strong> whom 24 received<br />

erlotinib and 3 afatinib. Fourteen patients did not receive further TKI<br />

treatment after progression (controls). Overall survival (OS) from progression<br />

on TKI and OS from diagnosis <strong>of</strong> lung cancer was analyzed for the<br />

whole population and case-control subpopulations <strong>of</strong> pairs matched for<br />

gender, smoking status, and histology. Results: Treatment with TKI and<br />

chemotherapy was well tolerated with no increase in grade 3 and 4<br />

toxicities. TKI-patients had a significantly longer OS from progression on<br />

TKI (case control: median 21.0 vs. 3.0 months, HR 0.175) and longer OS<br />

from diagnosis <strong>of</strong> lung cancer (case control: median 28.5 vs. 15.3 months,<br />

HR 0.335). Conclusions: In long-term erlotinib responders, treatment with<br />

TKI beyond progression in addition to chemotherapy or radiotherapy is<br />

feasible and well tolerated with limited toxicity. TKI-treatment beyond<br />

progression improved OS compared to treatment with TKI-free chemotherapy<br />

or radiotherapy.<br />

7574 General Poster Session (Board #48H), Sat, 1:15 PM-5:15 PM<br />

Efficacy <strong>of</strong> pemetrexed as second-line therapy in advanced NSCLC after<br />

either treatment-free interval or maintenance therapy with gemcitabine or<br />

erlotinib in IFCT-GFPC 05-02 phase III study. Presenting Author: Olivier<br />

Bylicki, Hospices Civils de Lyon, Lyon, France<br />

Background: Continuous exposure <strong>of</strong> tumor cells to maintenance therapy in<br />

advanced NSCLC might lead to resistance to subsequent treatments.<br />

IFCT–GFPC 0502 study showed a progression-free survival (PFS) benefit<br />

with gemcitabine (G) or erlotinib (E) maintenance compared to observation<br />

(O) after cisplatin-G induction chemotherapy. The trial included a predefined<br />

second-line therapy with pemetrexed (P), allowing post-hoc assessment<br />

<strong>of</strong> its efficacy according to previous maintenance treatment or<br />

treatment-free interval. Methods: Stage IIIB/IV NSCLC patients (pts) with a<br />

PS <strong>of</strong> 0-1 were randomized after 4 cycles <strong>of</strong> cisplatin-G chemotherapy to O<br />

or to receive maintenance therapy with G or E until disease progression. P<br />

was given as second-line treatment on disease progression in all arms. PFS<br />

and OS were assessed from the beginning <strong>of</strong> P therapy according to<br />

randomization arm. Tumor response to P and tolerance were also analyzed.<br />

Results: Of the 464 pts randomized to either O (155), G (154) or E (155),<br />

360 pts (78 %) received P as second-line therapy, i.e. 130 (84%), 114<br />

(74%) and 116 (75%) in O, G and E arm, respectively. Baseline<br />

characteristics remained well balanced between arms (overall median age<br />

<strong>of</strong> 58 years, 28% female, 91% stage IV, 41% PS 0, 65/19/16%,<br />

adenocarcinoma/squamous/other, 10% non-smokers and 56% responders<br />

to induction CT). Median number <strong>of</strong> delivered P cycles was 3 (1-40) in all<br />

arms. Response rate was 19%, 7% and 15% for non-squamous pts in O, G<br />

and E, respectively. Median PFS did not significantly differ between G and<br />

O (4.2 vs 3.9 months, HR [95% CI] 0.81 [0.62-1.06]) or E and O (4.2 vs<br />

3.9 months, HR 0.83 [0.64-1.09]). OS data showed a non-significant<br />

improvement with G vs O (HR 0.81 [0.61-1.07]) or E vs O (HR 0.80<br />

[0.61-1.05]), with a median <strong>of</strong> 7.5, 8.3 and 9.1 months for O, G and E,<br />

respectively. Results were similar when analysis was restricted to nonsquamous<br />

pts. Grade 3-4 treatment-related AEs were similar in O (33.1%),<br />

G (31.6%) and E (25%). Conclusions: Maintenance therapy with continuation<br />

<strong>of</strong> G or switch to E does not impair the efficacy <strong>of</strong> second-line P by<br />

comparison with administration after a treatment-free interval.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


498s Lung Cancer—Non-small Cell Metastatic<br />

7575 General Poster Session (Board #49A), Sat, 1:15 PM-5:15 PM<br />

Prospective randomized phase II trial <strong>of</strong> oral vinorelbine (NVBo) and<br />

cisplatin (P) or pemetrexed (Pem) and P in first-line metastatic or locally<br />

advanced non-small cell lung cancer (M or LA NSCLC) patients (pts) with<br />

nonsquamous (non SCC) histologic type: NAVoTRIAL01—Preliminary<br />

results. Presenting Author: Jaafar Bennouna, Institut de Cancerologie de<br />

l’Ouest/Site René Gauducheau, Nantes, France<br />

Background: NVBo�P is considered as a standard treatment (trt) in M or LA<br />

NSCLC. The recent approval <strong>of</strong> Pem�P as front line chemotherapy (CT) for<br />

non-SCC demonstrates that today, histology could become a “new guidance”<br />

to treat patients (pts). Phase III trial (Scagliotti. JCO 2008) showed<br />

efficacy <strong>of</strong> Pem�P in patients with non-SCC pts. Moreover, the higher<br />

chemosensitivity <strong>of</strong> non-SCC is recognised and reported with other chemotherapies<br />

(Ardizzoni.JNCI 2007). In GLOB 3 study, NVBo�P also showed<br />

better survival in adenocarcinoma (11.7m) than in SCC (8.9m) (Tan.Ann <strong>of</strong><br />

Oncol 2009). This trial was set up to assess the efficacy <strong>of</strong> NVBo�P<br />

compared to Pem�P for pts with non-SCC histological type. The primary<br />

end-point was disease control rate (DCR) including overall response rate<br />

(ORR) and stable disease (SD). Methods: Pts were randomised to receive<br />

NVBo 80 mg/m² D1D8 (60 at Cycle 1) � P 80 mg/m² D1 q3w or Pem 500<br />

mg/m² � P 75 mg/m² D1 q3w. After 4 cycles <strong>of</strong> trt, for pts showing a<br />

disease control, single agent as continuation maintenance (CM) was<br />

proposed until progression or toxicity. Given that Pem�P is now considered<br />

as a reference trt in non-SCC, pts were randomised on a 2/1 basis and<br />

stratified according to stage (IIIB - IV - relapse), non-SCC confirmed by<br />

histology or cytology, gender, smoking status and centre. Results: From<br />

10/09 to 02/11, 153 pts were randomised to receive NVBo�P (102 pts) or<br />

Pem�P (51 pts). The efficacy between the 2 arms is similar: The ORR/DCR<br />

(Recist 1.1) after 4 cycles <strong>of</strong> trt are 26%/75% in the NVBo arm (100 eval<br />

pts in ITT population) and 24%/78% in the Pem arm (50 eval pts in ITT<br />

population). At the end <strong>of</strong> December 2011, 7 patients are still treated in<br />

CM: 5 pts in the NVBo arm and 2 pts in the Pem arm. Conclusions: The2CT<br />

regimen, NVBo�P orPem�P have a similar efficacy in terms <strong>of</strong> ORR/DCR<br />

for pts with advanced non-SCC NSCLC. These results appear to be in line<br />

with the published data. As the analysis is currently in progress, final<br />

results including safety data, PFS and OS will be presented during the<br />

meeting.<br />

7577 General Poster Session (Board #49C), Sat, 1:15 PM-5:15 PM<br />

The relevance <strong>of</strong> stable disease (SD) as a surrogate end-point in advanced<br />

non-small cell lung cancer (NSCLC) patients treated with erlotinib (E) as<br />

the second/third line. Presenting Author: Francesco Grossi, Lung Cancer<br />

Unit, National Institute for Cancer Research, Genova, Italy<br />

Background: SD has <strong>of</strong>ten been viewed as a result with an uncertain clinical<br />

value.With the advent <strong>of</strong> E in advanced NSCLC, increasing numbers <strong>of</strong><br />

patients (pts) achieve SD as a best response. A common observation in<br />

clinical practice is that a high proportion <strong>of</strong> patients receiving E have<br />

durable SD.The aim <strong>of</strong> this analysis was to compare the progression-free<br />

survival (PFS) and overall survival (OS) in pts with advanced NSCLC who<br />

achieved confirmed SD or complete/partial response (CR�PR) after second/<br />

third line treatment with E. Methods: Data from 684 Italian pts from the<br />

TRUST trial (Tiseo M et al. Lung Cancer 2008) were analyzed. E was<br />

administered orally at 150 mg per day and was continued until disease<br />

progression, development <strong>of</strong> unacceptable toxicity or patient’s refusal. We<br />

define confirmed SD (cSD) if the patients’ PFS was �5 months (response to<br />

E was evaluated per protocol every two months) and reconfirmed SD (rSD) if<br />

the patients’ PFS was �8 months. Results: Pts’ characteristics included the<br />

following: median age, 67 years (31-89); females/males� 219/465 pts<br />

(32%/68%); never/former smokers� 191/493 pts (28%/72%); squamous/<br />

adeno/BAC/large cell/NOS� 163/361/26/21/113 pts (24%/53%/4%/3%/<br />

16%); ECOG PS 0-1/2-3� 557/127 pts (81%/19%). In 83 pts (12%), E<br />

was administered as the first-line therapy in pts who were unable to receive<br />

chemotherapy; 305 pts (45%) had received 1 prior line <strong>of</strong> chemotherapy,<br />

and 296 pts (43%) had received 2 prior lines <strong>of</strong> chemotherapy. A total <strong>of</strong><br />

428 pts were evaluable for a response: 44 pts (10%) exhibited CR�PR,<br />

226 pts (53%) attained SD, which consisted <strong>of</strong> 139 pts (32%) with cSD<br />

and 83 pts (19%) with rSD. The median PFS estimates (with 95% CI) were<br />

13.1 months (9.8-16.4) for CR�PR pts, 9.9 months (8.2-11.4) for cSD<br />

pts and 14 months (12-16) for rSD. The median OS estimates (with 95%<br />

CI) were 24.0 months (17-31) for CR�PR pts, 17.9 months (14.4-21.5)<br />

for cSD pts and 24.9 months (18.1-31.7) for rSD. Conclusions: Our<br />

findings are the first to demonstrate the relevance <strong>of</strong> achieving stable<br />

disease with E treatment. Pts obtaining cSD or rSD had durable PFS and<br />

OS comparable, particularly for rSD, with PFS and OS <strong>of</strong> those having<br />

CR�PR.<br />

7576 General Poster Session (Board #49B), Sat, 1:15 PM-5:15 PM<br />

Difference in incidence and pattern <strong>of</strong> salvage treatment after failure to<br />

first-line epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-<br />

TKI) monotherapy and standard cytotoxic chemotherapy in pts with<br />

advanced non-small cell lung cancer (NSCLC) harboring EGFR mutations:<br />

Okayama Lung Cancer Study Group experience. Presenting Author: Yuka<br />

Kato, Okayama University Hospital, Okayama, Japan<br />

Background: EGFR-TKI (E) therapy yielded a better PFS than standard<br />

cytotoxic chemotherapy (C) therapy and a comparable OS in untreated pts<br />

with EGFR-mt tumors, suggesting each <strong>of</strong> the treatments is now crucial for<br />

such subpopulation. But, it has not been fully evaluated yet which <strong>of</strong> each<br />

should be initiated first, and to what degree both <strong>of</strong> two are actually<br />

administered in early line setting in the treatment course. We here<br />

investigated a potential difference in incidence and pattern <strong>of</strong> delivery <strong>of</strong><br />

subsequent crossover therapy after failure to each <strong>of</strong> the treatments in pts<br />

with EGFR-mt tumors. Methods: Consecutive 79 pts with advanced EGFR-mt<br />

NSCLC were retrospectively assessed who underwent E therapy (n � 39) or<br />

standard C therapy (n � 40) in the 1st-line setting between 2007 and<br />

2011. Results: In E group 16 (41%) <strong>of</strong> 39 pts were still on 1st-line E<br />

therapy. Nine (39%) <strong>of</strong> the remaining 23 could not receive standard C<br />

therapy after failure to E therapy due to symptomatic CNS metastasis(mets)<br />

in 6, skeletal events in 2, and patient refusal in 1, whilst in C group only one<br />

(3%) <strong>of</strong> 40 failed to receive subsequent E therapy because <strong>of</strong> relapse<br />

(carcinomatous lymphangitis) (�2-test; p � .001). Also, at the time <strong>of</strong><br />

relapse to the 1-st line therapy, PS deteriorated more frequently in E group<br />

(8/23 vs. 7/40; p � .042) and, relapse with symptomatic CNS mets<br />

seemed frequently observed in E group (6/23 vs. 4/40; p � .093).<br />

Multivariate analysis revealed type <strong>of</strong> regimens undergone in the 1st-line<br />

setting (E vs. C) correlated with failure <strong>of</strong> administration <strong>of</strong> subsequent<br />

crossover therapy (odd ratio: 3.224, 95%CI: .1.032 – 5,417, p � .004).<br />

Conclusions: It seems that pts with EGFR-mt tumors who were treated with<br />

1st-line C had better opportunity to receive post-progression E therapy than<br />

those treated with 1st-line E had chance to receive subsequent C therapy,<br />

possibly due to difference in PS deterioration rate and relapse pattern.<br />

7578 General Poster Session (Board #49D), Sat, 1:15 PM-5:15 PM<br />

KIF5B-RET: Discovery <strong>of</strong> a novel fusion oncogene in lung adenocarcinomas<br />

by a systematic screen for tyrosine kinase fusions and identification <strong>of</strong><br />

patients for a RET targeted therapy trial. Presenting Author: Yoshiyuki<br />

Suehara, Memorial Sloan-Kettering Cancer Center, New York, NY<br />

Background: The mutually exclusive pattern <strong>of</strong> major targetable driver<br />

oncogenes in lung adenocarcinomas (ADC) suggests that other similar<br />

driver oncogenes may exist. We therefore performed a systematic screen for<br />

tyrosine kinase (TK) fusions in cases without known driver oncogenes by<br />

measuring aberrantly high RNA expression <strong>of</strong> kinase domain (KD) exons<br />

relative to more 5’ exons. Methods: We studied 74 patients whose lung ADC<br />

lacked mutations in KRAS, EGFR, BRAF, HER2, and ALK fusions. A<br />

NanoString-based assay was designed to query the transcripts <strong>of</strong> 90 TKs at<br />

two points: 5’ to the KD and within or 3’ to the KD. Tumor RNAs were<br />

hybridized to the NanoString probes and analyzed for outlier 3’ to 5’<br />

expression ratios. The assay was validated on samples with known ALK and<br />

ROS fusions. Presumed novel fusion events were followed up by rapid<br />

amplification <strong>of</strong> cDNA ends (RACE) and confirmatory RT-PCR. Results: The<br />

NanoString assay identified aberrant 5’ to 3’ ratios in ROS and RET in 2<br />

cases, respectively, out <strong>of</strong> 74. RACE analysis isolated a novel GOPC-ROS<br />

fusion in the former and a novel KIF5B-RET fusion in the latter, both<br />

confirmed by RT-PCR. Further screening by RT-PCR for KIF5B-RET<br />

identified one more positive sample in the study set that had not been<br />

detected by NanoString. At the RNA level, both fusions joined exon 15 <strong>of</strong><br />

KIF5B to exon 12 <strong>of</strong> RET, thus retaining a portion <strong>of</strong> the dimerization<br />

domain <strong>of</strong> KIF5B and the entire KD <strong>of</strong> RET, analogous to RET fusions in<br />

papillary thyroid carcinoma (TC). One KIF5B-RET patient was a 60 y.o.<br />

female never smoker, the other, a 73 y.o. male former smoker. Conclusions:<br />

The novel KIF5B-RET fusion described here and also recently reported by<br />

Ju YS et al. (Genome Res, Dec 22, 2011) defines a new subset <strong>of</strong> lung ADC<br />

with a potentially targetable driver oncogene. Based on these genetic data<br />

and the preclinical activity <strong>of</strong> the RET inhibitor XL184 (Exelixis) in<br />

papillary TC and its known activity in medullary TC with RET mutations, we<br />

have initiated prospective testing for KIF5B-RET as part <strong>of</strong> our lung ADC<br />

screening panel in anticipation <strong>of</strong> a planned phase 2 trial with XL184 in<br />

patients with KIF5B-RET or related variant RET fusions.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


7579 General Poster Session (Board #49E), Sat, 1:15 PM-5:15 PM<br />

Family history <strong>of</strong> lung cancer in never smokers with non-small cell lung<br />

cancer (NSCLC) and its association with tumors harboring epidermal<br />

growth factor receptor (EGFR) mutations. Presenting Author: Elizabeth<br />

Mary Gaughan, Beth Israel Deaconess Medical Center/Harvard Medical<br />

School, Boston, MA<br />

Background: Inherited susceptibility to lung cancer is an understudied<br />

subject, however it has been described among never smokers (�100<br />

cigarettes/lifetime). Never smokers with NSCLC comprise an important<br />

subgroup <strong>of</strong> patients enriched for tumors harboring oncogene aberrations in<br />

the EGFR and ALK genes. We aimed to better characterize the incidence <strong>of</strong><br />

family history <strong>of</strong> lung cancer in the setting <strong>of</strong> routine tumor genotyping<br />

among never smokers with NSCLC. Methods: Clinicopathologic data plus<br />

tumor genotype (EGFR, KRAS, ALK) from 230 consecutive never smokers<br />

seen at Beth Israel Deaconess Medical Center and Dana-Farber Cancer<br />

Institute was compiled. We retrospectively analyzed the incidence <strong>of</strong> a<br />

family history <strong>of</strong> any cancer and lung cancer in these patients. Results: In<br />

our cohort, the average age was 56 years, 67% <strong>of</strong> the patients were women,<br />

75% were white, 41% had advanced NSCLC and 87% had adenocarcinoma<br />

histology. In these tumors, 98/230 (43%) had an EGFR mutation,<br />

16/155 (10%) had KRAS mutations and 27/127 (17%) had an ALK<br />

translocation. Family history <strong>of</strong> any cancer was common (57%) and specific<br />

family history <strong>of</strong> lung cancer was present in 42/230 cases (18%). Out <strong>of</strong><br />

thecases with a family history <strong>of</strong> any cancer, 22/53 (41.5%) EGFRmutated,<br />

1/6 (17%) KRAS-mutated and 3/20 (15%) ALK-translocated<br />

cohorts had a family history <strong>of</strong> lung cancer. The rate <strong>of</strong> family history <strong>of</strong> lung<br />

cancer to family history <strong>of</strong> cancer was significantly higher in the EGFRmutated<br />

cohort when compared to the ALK translocated plus KRASmutated<br />

cohorts (p�0.023). Conclusions: Family history <strong>of</strong> lung cancer is<br />

common in never smokers with NSCLC, and there seems to be a particular<br />

link in families in which the proband has an EGFR-mutated tumor. Further<br />

study <strong>of</strong> families with EGFR-mutated NSCLC may yield insights into the<br />

pathogenesis <strong>of</strong> this tumor type.<br />

7581 General Poster Session (Board #49G), Sat, 1:15 PM-5:15 PM<br />

NGR-hTNF plus chemotherapy as first-line therapy <strong>of</strong> non-small cell lung<br />

cancer (NSCLC). Presenting Author: Vanesa Gregorc, Department <strong>of</strong><br />

Oncology, Istituto Scientifico San Raffaele, Milan, Italy<br />

Background: NGR-hTNF (asparagine-glycine-arginine human tumor necrosis<br />

factor) is a selective vascular targeting agent able to improve intratumoral<br />

chemotherapy uptake. Methods: Chemo-naive, stage IIIb-IV NSCLC<br />

patients (pts), stratified by histology (nonsquamous or squamous) and PS<br />

(0 or 1), were randomized to cisplatin 80 mg/m2 /day(d)1 plus either<br />

pemetrexed 500 mg/m2 /d1 (nonsquamous) or gemcitabine 1,250 mg/m2 /<br />

d1�8 (squamous) every 3 weeks (q3w) up to 6 cycles with (arm A) or<br />

without (arm B) NGR-hTNF 0.8 �g/m2 /d1 q3w until progression. Progression<br />

free survival (PFS) was primary end point <strong>of</strong> this phase 2 trial<br />

(��20%, ��20%, HR�0.62, and n�102). Secondary end points included<br />

adverse events (AEs), response rate (RR) and overall survival (OS).<br />

Results: 59 pts were assigned to arm A and 57 to arm B. Baseline<br />

characteristics in arm A/B were: median age 62/63; male 34/37; PS 1<br />

21/21; squamous 15/15; nonsmokers 14/12; EGFR mutations 5/5. For the<br />

nonsquamous group, 281 cycles (mean 6.5; range 1-18) were given in arm<br />

A and 190 (mean 4.7; range 1-6) in arm B, while for the squamous group,<br />

88 (mean 6.3; range 1-19) in arm A and 48 (mean 3.7; range 1-6) in arm<br />

B. Most common grade 3/4 AEs were neutropenia 14% vs 17% and fatigue<br />

7% vs 11% in arm A and B, respectively. No grade 3/4 AEs related to<br />

NGR-hTNF or bleeding in pts with squamous histology were noted. Median<br />

follow-up was 12.4 months. In the whole study population, median (m)PFS<br />

was 5.8 vs 5.7 months (HR�0.95), RR was 23% vs 19%, and 1-year OS<br />

was 62% vs 62% in arm A and B, respectively. In the nonsquamous subset,<br />

a trend toward longer mPFS in arm A compared to arm B was noted in pts<br />

with a PS <strong>of</strong> 1 (6.7 vs 4.6 months, respectively), nonsmoking history (6.2 vs<br />

3.4), younger age (6.5 vs 3.4), and EGFR mutations (7.2 vs 3.3). In the<br />

squamous subset, mPFS was 5.1 vs 3.9 months (HR�0.71) and mOS was<br />

14.2 vs 10.8 months (HR�0.73) in arm A and B, respectively. In these pts,<br />

ORR was 36% in arm A and 23% in arm B, while median changes from<br />

baseline in target tumor size after 2, 4, and 6 cycles were -29%, -45%, and<br />

-41%, respectively in arm A, and -18%, -22%, and -14%, respectively in<br />

arm B. Conclusions: Regardless <strong>of</strong> histology, NGR-hTNF can be safely given<br />

with standard chemotherapy, showing tolerability and hint <strong>of</strong> activity in<br />

squamous NSCLC.<br />

Lung Cancer—Non-small Cell Metastatic<br />

499s<br />

7580 General Poster Session (Board #49F), Sat, 1:15 PM-5:15 PM<br />

Comparison <strong>of</strong> the KRAS/EGFR mutation pr<strong>of</strong>ile and survival <strong>of</strong> “collegiate<br />

smokers” and never smokers with advanced lung cancers. Presenting<br />

Author: Anna M. Varghese, Memorial Sloan-Kettering Cancer Center, New<br />

York, NY<br />

Background: In practice, we encounter patients (pts) with lung cancers who<br />

state they smoked only while in college. The impact <strong>of</strong> this degree <strong>of</strong><br />

tobacco use on cancer biology is unknown. We have shown that EGFR<br />

mutations are less common in pts who smoked � 15 pack years (PY). We<br />

hypothesize that among pts with lung cancer the KRAS / EGFR mutation<br />

pr<strong>of</strong>ile and overall survival (OS) <strong>of</strong> “collegiate smokers” (former smokers<br />

who smoked between 101 lifetime cigarettes and 5 PY) will be distinct<br />

from never smokers and former smokers with � 15 PY. Methods: We<br />

collected age, sex, stage, and survival for pts evaluated from 2004 - 2009<br />

with stage IIIB/IV lung cancer with known KRAS and EGFR status. ALK<br />

testing was not available routinely during this time. Smoking history was<br />

obtained using a patient completed survey. The Fisher exact test was used<br />

to compare mutation pr<strong>of</strong>iles. The log rank test was used to compare OS.<br />

Results: Smoking history and clinical data were available for 852 pts with<br />

Stage IIIB/IV lung cancer with known KRAS and EGFR status: 307 never<br />

smokers, 178 current smokers, and 367 former smokers. Of the former<br />

smokers, 55 were “collegiate smokers”, 61 had smoked 5-15 PY, and 251<br />

had smoked � 15 PY. The KRAS / EGFR mutation pr<strong>of</strong>iles by smoking<br />

history are shown below (Table). The KRAS / EGFR mutation pr<strong>of</strong>ile <strong>of</strong><br />

“collegiate smokers” is distinct from those <strong>of</strong> pts who were never smokers<br />

(p � .001) and former smokers with � 15 PY (p � .001) but similar to that<br />

<strong>of</strong> pts who were former smokers with 5-15 PY (p � 0.9). Median OS after<br />

diagnosis <strong>of</strong> stage IIIB/IV lung cancer for “collegiate smokers” was 25<br />

months (mos), compared to 32 mos for never smokers (p � 0.4) and 21<br />

mos for former smokers with � 15 PY (p � 0.63). Conclusions: “Collegiate<br />

smokers” are a distinct group <strong>of</strong> pts with a higher incidence <strong>of</strong> KRAS<br />

mutations and lower incidence <strong>of</strong> EGFR mutations compared to never<br />

smokers. These data suggest that even a small amount <strong>of</strong> cigarette smoking<br />

influences the biology <strong>of</strong> lung cancer. These findings reinforce the<br />

importance <strong>of</strong> doing mutation testing routinely for all pts with lung cancer<br />

to guide clinical care.<br />

Total EGFR KRAS<br />

N N % N %<br />

Never smokers 307 149 49 13 4<br />

�Collegiate smokers� 55 15 27 8 15<br />

Former smokers with > 15 pack years 251 27 11 100 40<br />

7582 General Poster Session (Board #49H), Sat, 1:15 PM-5:15 PM<br />

Thymidylate synthase (TS) gene expression in patients with ALK positive<br />

(�) non-small cell lung cancer (NSCLC): Implications for therapy. Presenting<br />

Author: David R. Gandara, University <strong>of</strong> California Davis Cancer Center,<br />

Sacramento, CA<br />

Background: ALK� NSCLC represents a molecular target-defined patient<br />

population highly responsive to the ALK inhibitor crizotinib. Previous<br />

reports have also suggested increased sensitivity <strong>of</strong> ALK� NSCLC to the<br />

chemotherapeutic agent pemetrexed. Thymidylate synthase (TS) is a<br />

candidate predictive biomarker for pemetrexed activity. Here we report<br />

analysis <strong>of</strong> the Response Genetics Inc. (RGI) database for this association<br />

and implications for therapy. Methods: ALK fusion was identified by a novel<br />

RT-PCR assay (Danenberg et al: ASCO 2010). For TS, RNA from microdissected<br />

formalin-fixed paraffin-embedded tumors was analyzed as previously<br />

described, reported as the ratio <strong>of</strong> gene expression to �-actin. For<br />

reference, a TS level �2.33 is the cutpoint for sensitivity. Results: TS levels<br />

were available from 63 ALK� patients and 1,698 ALK- control lung<br />

adenocarcinoma patients. All ALK� patients had adenocarcinomas without<br />

EGFR or KRAS mutations. Median age: 59.0 (range 33-88), gender<br />

(male/female) 32/31 (51%/49%). Median TS RNA level in ALK� patients<br />

was 2.02, range (0.55-19.44), and in ALK- patients was 3.32 (0.36-<br />

53.51), p�0.0001 (Mann-Whitney test). The majority <strong>of</strong> ALK� patients<br />

(N�43, 68%) had a TS level �2.33 cutpoint, compared to only 32% <strong>of</strong><br />

ALK- patients (N�551, p�0.0001). Conclusions: This analysis demonstrates<br />

relatively low TS gene expression in ALK� patient tumors as<br />

determined by RT-PCR. These data provide a mechanism <strong>of</strong> action<br />

supportive <strong>of</strong> pemetrexed sensitivity for ALK� NSCLC.<br />

TS expression N Median 95% CI Range Mean 95% CI p value<br />

ALK� patients 63 2.02 1.60-2.11 0.55-19.44 2.53 1.89-3.16 �0.0001<br />

ALK- patients 1698 3.32 3.15-3.45 0.36-53.51 7.87 1.68-14.05<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


500s Lung Cancer—Non-small Cell Metastatic<br />

7583 General Poster Session (Board #50A), Sat, 1:15 PM-5:15 PM<br />

Correlation between thymidylate synthetase (TS) expression and progressionfree<br />

survival (PFS) in patients receiving pemetrexed (pem) for advanced<br />

NSCLC: A prospective study. Presenting Author: Marianne Nicolson,<br />

Aberdeen Royal Infirmary, Aberdeen, United Kingdom<br />

Background: Pem efficacy in non-squamous (NS) NSCLC is hypothesised to<br />

be associated with TS expression. Our primary objective was to assess the<br />

correlation between TS and PFS, prospectively. Methods: Context: A Phase<br />

II, single-arm, exploratory, multicenter, UK study with appropriate approvals<br />

and consents. Key eligibility criteria: ECOG PS 0-1, NS-NSCLC<br />

histology, stage IIIB/IV. Patients (n�70) received pem/cisplatin induction<br />

x 4. Non-progressive patients continued on maintenance pem until tumor<br />

progression or discontinuation. All patients were followed until death or<br />

study closure. TS by IHC (H-scores) and qPCR (delta CQ values normalized)<br />

were assessed on diagnostic FFPE samples. Kaplan-Meier estimates for<br />

median PFS (mPFS) and Cox regression to determine the statistical<br />

correlation between PFS and TS IHC nuclear score, (continuous variable)<br />

were performed. Maximal Chi-square analysis identified the optimal association<br />

cutpoints between PFS and low vs. high TS scores. Results: Patient<br />

demographics were unremarkable, 32/70 (45.7%) were female. 55 (78.6%)<br />

had adenocarcinoma; 15 were not otherwise specified. Maintenance pem<br />

was started in 43/70 (61.4%) patients. Evaluable patients had at least one<br />

dose <strong>of</strong> study treatment and a valid TS score (n�60). For the evaluable<br />

population, the mPFS from start <strong>of</strong> induction was 5.5 months (95% CI<br />

3.9-6.9). A statistically significant correlation between PFS and TS IHC<br />

nuclear scores was observed [p�0.0001, HR� 1.01 (95% CI 1.01,<br />

1.02)], suggesting higher TS values are associated with shorter PFS. When<br />

the population was dichotomized at the identified optimal H-score cutpoint<br />

<strong>of</strong> 70, the mPFS in the low expression group (n�40) was 7.1 months<br />

(5.7-8.3) compared to 2.6 months (1.3-4.1) in the high expression group<br />

(n�20) [adjusted p�0.0015, HR�0.28 (95% CI 0.16-0.52)]. Similar<br />

trends with cytoplasmic TS IHC (p�0.09) and TS qPCR (p�0.05) levels<br />

were observed. Statistical correlations were seen among the TS levels. Data<br />

is preliminary. Conclusions: Study suggests statistically significant association<br />

between low TS IHC nuclear expression and improved PFS in this<br />

Phase 2 single-arm trial.<br />

7585 General Poster Session (Board #50C), Sat, 1:15 PM-5:15 PM<br />

Expression <strong>of</strong> folate pathway regulators and pemetrexed (pem) activity in<br />

patients (pts) with advanced non-small cell lung cancer (NSCLC). Presenting<br />

Author: Letizia Gianoncelli, Humanitas Cancer Center, Rozzano, Italy<br />

Background: Thymidylate synthase (TS) expression has been reported to<br />

predict pem activity in pts with advanced NSCLC. Besides TS, pem inhibits<br />

multiple enzymes <strong>of</strong> the folate pathway including dihydr<strong>of</strong>olate reductase<br />

(DHFR), glycinamide ribonucleotide formyltransferase (GART) and aminoimidazole<br />

carboxamide ribonucleotide formyltransferase (AICAR). Aim <strong>of</strong><br />

present study was to investigate whether these or other biomarkers<br />

influence pem activity in NSCLC pts. Methods: Advanced pretreated NSCLC<br />

pts who received at least two cycles <strong>of</strong> single agent pem were considered<br />

eligible if they had available tumor tissue to assess at least one <strong>of</strong> the<br />

proposed biomarkers. TS, DHFR, GART and AICAR protein expression was<br />

assessed by immunohistochemistry (IHC) in FFPE tumor samples. Pts were<br />

grouped as IHC� and IHC– according to staining intensity. Additionally,<br />

presence <strong>of</strong> EGFR and KRAS mutations was investigated. Results: Ninetysix<br />

pts were included in the study. The majority <strong>of</strong> subjects was male<br />

(72%), smokers (93%), with adenocarcinoma (72%). Response rate (RR),<br />

disease control rate (DCR), progression-free survival (PFS) and overall<br />

survival (OS) were 12%, 47%, 2.3 and 9.5 months, respectively. Most pts<br />

resulted TS-(72%), AICAR-(82%), DHFR� (68%), GART- (61%). EGFR<br />

and KRAS mutations were identified in 15% and 25% <strong>of</strong> assessable cases.<br />

None <strong>of</strong> the biomarkers was significantly associated with pts characteristics<br />

(gender, histology, smoking status), nor with RR or DCR. GART- pts<br />

experienced longer PFS when compared with the GART� group (HR 0.56,<br />

p�.052), while no difference was observed according to TS, DHFR and<br />

AICAR status. A significantly longer OS was observed for TS- (HR 0.52,<br />

p�.012), GART- (HR 0.50, p�.037) and AICAR- (HR 0.40, p�.028) pts.<br />

No significant difference in the distribution <strong>of</strong> EGFR mutant pts receiving<br />

EGFR TKIs after pem failure was observed between IHC� and IHC- groups.<br />

Conclusions: Low TS, GART and AICAR protein expression predicts significantly<br />

prolonged survival in single agent pem-treated pts with advanced<br />

NSCLC. The lack <strong>of</strong> PFS difference according to TS and AICAR status<br />

suggests prognostic rather than predictive relevance for these biomarkers.<br />

7584 General Poster Session (Board #50B), Sat, 1:15 PM-5:15 PM<br />

A randomized, double-blinded, phase II study <strong>of</strong> paclitaxel/carboplatin<br />

(PC) plus CT-322 versus PC plus bevacizumab (Bev) as first-line treatment<br />

for advanced nonsquamous non-small cell lung cancer (NSCLC). Presenting<br />

Author: Eugene Henry Paschold, Piedmont Hematology-Oncology<br />

Associates, Winston-Salem, NC<br />

Background: CT- 322 is a pegylated protein engineered from the tenth type<br />

III human fibronectin domain thatspecifically blocks VEGFR-2 signaling by<br />

all known ligands. This international randomized study assessed the<br />

efficacy and safety <strong>of</strong> PC � CT-322 compared to PC � Bev as first-line<br />

treatment for advanced non-squamous NSCLC. Methods: In addition to<br />

paclitaxel 200 mg/m2and carboplatin AUC 6 given on day 1 <strong>of</strong> a 21 day<br />

cycle(maximum 6 cycles), subjects, stratified by ECOG performance<br />

status, disease stage, and site, were randomized 1:1 to receive either<br />

CT-322 2 mg/kg on days 1, 8, and 15 (arm A) or Bev 15 mg/kg on day 1 and<br />

placebo on days 8 and 15 (arm B). CT -322 and Bev/placebo were<br />

continued as maintenance until disease progression, unacceptable toxicity,<br />

or withdrawal <strong>of</strong> consent. The primary endpoint was progression-free<br />

survival (PFS); secondary endpoints included overall survival (OS), response<br />

rate, and safety. Results: 255 subjects were randomized to arm A<br />

(n�127) or arm B (n�128). Baseline characteristics were wellbalanced.<br />

After a median follow- up <strong>of</strong> 13.8 months, 9 subjects on arm A and 24 on<br />

arm B remained on study treatment. The median treatment duration was 19<br />

weeks in arm A and 26.3 weeks on arm B. The median PFS in arm A was<br />

5.6 months compared to 6.8 months in arm B (HR 1.51, 1 -sided<br />

p�0.997). In all prespecified subgroups, PFS was favored in arm B. The<br />

response rate was 25.2% in arm A compared to 32.8% in arm B. Median<br />

OS was 12.5 months in arm A compared to 15.2 months in arm B. The<br />

most common grade � 3 adverse events (Arm A vs B) were neutropenia<br />

(47.2% vs 49.2%), thrombocytopenia (11.8% vs 6.3%), and fatigue<br />

(10.2% in both arms). Grade � 3 hypertension was more frequently<br />

reported in arm A (8.7% vs 3.1%). Grade � 3 venous thrombosis (5.5% vs<br />

6.3%), proteinuria (1.6% vs 2.3%), and bleeding events (0.8% vs 1.6%)<br />

were similar. Conclusions: PC � CT-322 failed to improve PFS, OS, or<br />

response rate compared to PC � Bev. However, thesafety pr<strong>of</strong>ile in the PC<br />

� CT-322 arm was consistent with the anti-angiogenic mechanism <strong>of</strong><br />

action, suggesting that CT-322 has biological activity as an inhibitor <strong>of</strong><br />

VEGFR-2.<br />

7586 General Poster Session (Board #50D), Sat, 1:15 PM-5:15 PM<br />

Single nucleotide polymorphisms (SNPs) <strong>of</strong> the platinum pharmacogenetic<br />

and VEGF pathways: Association with survival <strong>of</strong> platinum-treated stage IV<br />

non-small cell lung cancer (NSCLC) patients. Presenting Author: Sinead<br />

Cuffe, Princess Margaret Hospital, University <strong>of</strong> Toronto, Toronto, ON,<br />

Canada<br />

Background: Two potentially important host pathways in lung cancer<br />

systemic therapy are: (i) the pharmacogenetic pathway <strong>of</strong> platinum agents<br />

(DNA repair, metabolism, and multidrug resistance genes); and (ii) the<br />

vascular endothelial growth factor (VEGF) pathway. We investigated the<br />

relationship between SNPs in these two pathways and clinical outcome in<br />

platinum-treated NSCLC patients. Methods: 188 platinum-treated Stage IV<br />

NSCLC patients underwent SNP genotyping for the platinum-related (48<br />

SNPs in 7 genes) and VEGF (64 SNPs in 3 genes) pathways. SNPs were<br />

selected from the literature and through tagging. Association <strong>of</strong> SNPs and<br />

overall (OS) and progression free survival (PFS) were assessed using<br />

multivariate Cox proportional hazards models. Results: 72% were Caucasian;<br />

73%, adenocarcinoma; 92%, ECOG PS 0-1; median age, 60 years;<br />

54% received � one line <strong>of</strong> systemic therapy; 10% received anti-VEGF<br />

therapy/placebo; Median OS, 1.3 yrs; median follow up, 2.2 yrs. The top<br />

significant SNPs in the platinum-related pathway were in ABCC2<br />

(rs8187710 and rs2756109, r2�0.68). The G variants <strong>of</strong> the top SNP,<br />

ABCC2 rs8187710 (4554G�A), were associated with worse OS (adjusted<br />

hazard ratio [aHR], 2.62; 95%CI: 1.5-4.5; p�0.0005) and PFS (aHR,<br />

1.97; 95%CI: 1.2-3.4; p�0.01). Functionally, 4554G�A impairs ATPase<br />

activity and is associated with higher cellular accumulation <strong>of</strong> ABCC2<br />

substrates [PMID: 22027652]; furthermore, ABCC2 expression is associated<br />

with cisplatin resistance and clinical outcome in other cancers [PMID:<br />

17145840]. Within the VEGF pathway, the top significant SNPs were in<br />

the same haplotype block <strong>of</strong> VEGFR1/FLT1 (rs1324057, rs7324547,<br />

r2�1.0): for rs1324057, the aHR for OS was 1.59 (95%CI 1.2-2.1);<br />

p�0.001; and the aHR for PFS, 1.48 (95%CI 1.1-1.9); p�0.004.<br />

VEGFR1/FLT1 rs7324547 has been associated with esophageal cancer<br />

risk [PMID: 21751195], but has not been assessed in lung cancer.<br />

Conclusions: SNPS <strong>of</strong> the VEGFR1 and ABCC2 genes are strongly associated<br />

with OS and PFS in this cohort <strong>of</strong> platinum-treated advanced NSCLC<br />

patients. Future studies should assess whether these are predictive or<br />

prognostic markers.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


7587 General Poster Session (Board #50E), Sat, 1:15 PM-5:15 PM<br />

Sorafenib and continued erlotinib or sorafenib alone in patients with<br />

advanced non-small cell lung cancer progressing on erlotinib: A randomized<br />

phase II study <strong>of</strong> the Sarah Cannon Research Institute (SCRI).<br />

Presenting Author: Victor Gian, Tennessee Oncology, PLLC/Sarah Cannon<br />

Research Institute, Nashville, TN<br />

Background: Erlotinib is an oral epidermal growth factor receptor kinase<br />

inhibitor used in the treatment <strong>of</strong> advanced non-small-cell lung cancer<br />

(NSCLC). Resistance develops in patients (pts) who initially respond to<br />

erlotinib leading to progressive disease (PD). Sorafenib is an oral inhibitor<br />

<strong>of</strong> vascular endothelial and platelet-derived growth factor receptors and Raf<br />

kinases which play important roles in PD. This randomized phase II study<br />

evaluated the role <strong>of</strong> sorafenib and continued erlotinib or sorafenib alone in<br />

pts with progressive NSCLC following initial benefit with erlotinib. Methods:<br />

Eligible pts had IIIB/IV NSCLC, an ECOG PS 0-2, and had received �2<br />

lines <strong>of</strong> therapy with the last being single-agent erlotinib. Pts must have PD<br />

following clinical benefit (complete/partial response/stable disease) from<br />

erlotinib for �8 weeks. Pts were randomized 1:1 to continue erlotinib at the<br />

dose administered at the time <strong>of</strong> PD with the addition <strong>of</strong> sorafenib 400 mg<br />

orally twice daily (Arm A) or to sorafenib alone (Arm B). Cycles were 28 days<br />

with restaging every 2 cycles. The primary endpoint was progression-free<br />

survival (PFS). Results: 52 pts were enrolled from 2/2008 to 3/2011 (A 24;<br />

B 28). Baseline characteristics were balanced between arms and included:<br />

median age 65 years (41-87); 65% female; 69% adenocarcinoma/large<br />

cell; and 96% PS �2. 41% <strong>of</strong> pts were either never smokers or smoked<br />

�100 cigarettes/lifetime. Pts received a median <strong>of</strong> 8 weeks <strong>of</strong> treatment<br />

per arm (0.6–67 weeks). The median PFS was 3.1 (95% CI 1.7-3.7) and<br />

2.3 (1.7-3.6) months for A and B, respectively (p�.84). There were no<br />

grade 3/4 hematologic toxicities in either arm except grade 3 anemia in 1 pt<br />

(A). Severe nonhematologic toxicities in �5% included: fatigue 17%(A)/<br />

7%(B); diarrhea 17%/0; dehydration 13%/7%; hand-foot skin reaction<br />

8%/8%, and anorexia 4%/7%. Conclusions: Sorafenib has modest activity<br />

when used in combination with erlotinib or as a single agent in pts with PD<br />

following benefit with erlotinib alone. Additional study to identify potential<br />

subsets <strong>of</strong> refractory pts who might derive the greatest benefit from<br />

sorafenib are warranted.<br />

7589 General Poster Session (Board #50G), Sat, 1:15 PM-5:15 PM<br />

ALK and MET genes in advanced lung adenocarcinomas: The Lung Cancer<br />

Mutation Consortium experience. Presenting Author: Marileila Varella-<br />

Garcia, Department <strong>of</strong> Medicine/Medical Oncology, University <strong>of</strong> Colorado<br />

Cancer Center, Aurora, CO<br />

Background: The Lung Cancer Mutation Consortium (LCMC) consisting <strong>of</strong><br />

14 US Cancer Centers was established to evaluate a panel <strong>of</strong> molecular<br />

mutations in advanced lung adenocarcinoma. ALK gene fusions and MET<br />

gene amplification were assessed by FISH in CLIA certified laboratories.<br />

Methods: Molecular tests were performed in stage IIIB or IV lung adenocarcinoma.<br />

To date, FISH assays have been completed in 901 patients for ALK<br />

(ALK break-apart, Abbott Molecular) and in 654 patients for MET (in<br />

house/Abbott Molecular reagents). ALK� specimens were defined by split<br />

3’ALK/5’ALK signals (gap �2 signal diameters) or single 3’ALK signals in<br />

�15% <strong>of</strong> tumor cells. MET gene amplification (MET�) was defined by ratio<br />

mean MET/mean CEP7 �2. Results: The ALK� patient subset (N�75,<br />

8.3%) compared to the ALK- had significantly lower age at diagnosis (52<br />

vs. 60, p�0.001) and less frequent heavy smoking history (61% neversmokers<br />

among ALK� vs. 31% among ALK-, p�0.001; pack-year for<br />

current/former smokers 17 vs. 40, p�0.003). Liver metastases were<br />

significantly more frequent among ALK� than ALK- (23% vs.10%,<br />

p�0.004); no difference was detected in bone, brain and adrenal gland<br />

metastases. MET� (N�29, 4.4%) was significantly associated with female<br />

sex (72% female among MET� vs. 39% among MET-, p�0.001) and<br />

marginally more frequent in patients with adrenal metastasis; no difference<br />

was detected for age at diagnosis and smoking history. Follow-up on 73<br />

ALK� patients indicated that 56% received crizotinib as targeted therapy.<br />

Response was unknown in 8% and unreportable in 22% patients enrolled<br />

in ongoing randomized trials. Among patients with evaluable response,<br />

complete response, partial response, stable disease, and progressive<br />

disease were found respectively in 3%, 66%, 28%, and 3%. Conclusions:<br />

The LCMC successfully tested ALK and MET FISH in a large number <strong>of</strong> lung<br />

adenocarcinomas. It was demonstrated that directing positive patients to<br />

specific interventions is feasible, and that grouping <strong>of</strong> testing and trials<br />

within consortia may maximise relevant trial accrual in rare molecular<br />

subtypes. Supported by NCI-GO award. Submitted on behalf <strong>of</strong> the LCMC.<br />

Lung Cancer—Non-small Cell Metastatic<br />

501s<br />

7588 General Poster Session (Board #50F), Sat, 1:15 PM-5:15 PM<br />

<strong>Clinical</strong> characteristics <strong>of</strong> KRAS mutations in NSCLC and their impact on<br />

outcomes to first-line chemotherapy. Presenting Author: Mohit Butaney,<br />

Dana-Farber Cancer Institute, Boston, MA<br />

Background: KRAS is one <strong>of</strong> the most commonly mutated oncogenes in<br />

non-small cell lung cancer (NSCLC). While the impact <strong>of</strong> EGFR mutations<br />

and EML4-alk translocations has been well-described, there is limited<br />

information about the impact <strong>of</strong> these somatic mutations on response to<br />

chemotherapy. Methods: We retrospectively reviewed the demographics<br />

and clinical outcomes <strong>of</strong> patients with KRAS mutations and compared<br />

these to patients who were KRAS wild-type (WT). Eligible pts received<br />

1st-line IV chemo for stage IV NSCLC at DFCI and had known information<br />

about both KRAS and EGFR status. Since the biology and impact <strong>of</strong> EGFR<br />

mutations on response to chemo is well-described, we excluded such pts<br />

from the analysis. The primary endpoint was progression-free survival (PFS)<br />

with first-line chemo; secondary endpoints included radiographic response<br />

rate (RR) and overall survival (OS). Results: Between 2/05 and 8/11, there<br />

were 63 eligible KRAS pts and 97 eligible WT pts. The groups were similar<br />

in age (median 65yrs in both groups), % female (K 62, WT 54) race (K 89%<br />

white/6% black, 5% other; WT 86% white,/6% black/8% other), histology<br />

(K 90%adeno/8% NSCLC NOS; WT 86% adeno/9%NSCLC NOS), and % <strong>of</strong><br />

pts receiving 1/2/3 agents in 1st line (K 11/56/33; WT 18/53/30). KRAS<br />

pts were less likely to be never or light smokers (4% vs 33% for WT).<br />

Nonsmokers were more likely to harbor KRAS transition rather than<br />

transversion mutations (3 transition, 1 transversion), while the converse<br />

held for smokers (51 transversions, 8 transitions). Median PFS was similar<br />

for KRAS vs WT (K .65 vs WT 4.8 months, p�0.81). RR (29% for both<br />

groups), disease control rates (K 73% vs WT 78%), and median OS (K 13.5<br />

vs WT 12.1 months, p�.525) were also similar. Outcomes <strong>of</strong> KRAS pts to<br />

2nd line chemotherapy (PFS 2.2, OS 8.6) are similar to those seen for WT<br />

patients in this setting. There was no significant difference in outcomes<br />

based on gender, smoking status, drug received (pemetrexed-based vs<br />

taxane based), or specific KRAS genotype. Conclusions: Pts with KRAS<br />

mutations experience similar outcomes to standard chemotherapy as those<br />

who are wild-type for EGFR and KRAS. Going forward, these data can serve<br />

as a reference for control arms <strong>of</strong> KRAS-specific randomized trials.<br />

7590 General Poster Session (Board #50H), Sat, 1:15 PM-5:15 PM<br />

Weekly nab-paclitaxel in combination with carboplatin as first-line therapy<br />

in elderly patients (pts) with advanced non-small cell lung cancer (NSCLC).<br />

Presenting Author: Mark A. Socinski, University <strong>of</strong> Pittsburgh Medical<br />

Center, Pittsburgh, PA<br />

Background: The median age <strong>of</strong> advanced NSCLC pts in the US is 71 years;<br />

yetelderly pts (�70 years) are generally undertreated, with only �30%<br />

receiving systemic therapy. Hence, better, more tolerable therapeutic<br />

options are needed for this cohort. In a phase III trial nab-paclitaxel (nab-P,<br />

130 nm albumin-bound paclitaxel particles) � carboplatin (C) vs solventbased<br />

paclitaxel (sb-P) � C, significantly improved ORR, the primary<br />

endpoint (25% vs 33%, P � 0.005), with a 1-month improvement in OS (P<br />

� NS) and improved safety. This analysis evaluated efficacy and safety in<br />

pts �70 and �70 years old. Methods: Pts with untreated stage IIIB/IV<br />

NSCLC and with an ECOG score <strong>of</strong> 0/1 were randomized 1:1 (stratified by<br />

age [�70 vs �70 years], region, stage, gender, and histology) to C AUC 6<br />

day 1 and either nab-P 100 mg/m2 on days 1, 8, 15 or sb-P 200 mg/m2 day<br />

1 q 21 days. ORR and progression-free survival (PFS) were determined by<br />

blinded centralized review. Results: Of the phase III study population, 15%<br />

were elderly (156/1052; 74 pts in the nab-P/C and 82 in the sb-P/C arms).<br />

Most elderly pts were male (72%), Caucasian (71%), and had stage IV<br />

disease (64%). In pts both �70 and �70 years old, ORR was higher in the<br />

nab-P/C vs sb-P/C arm (�70: 34% vs 24%, P � 0.196; �70: 32% vs<br />

25%, P � 0.013). In pts �70 years old, PFS trended in favor <strong>of</strong> nab-P/C<br />

(median 8.0 vs 6.8 months, HR: 0.687, P � 0.134); OS was significantly<br />

improved (median 19.9 vs 10.4 months, HR: 0.583, P � 0.009). In<br />

contrast, PFS (6.0 vs 5.8 median months, HR: 0.903, P � 0.256) and OS<br />

(11.4 vs 11.3 median months, HR: 0.999, P � 0.988) were similar for<br />

both treatment arms in pts �70 years old. Adverse events were similar in<br />

pts �70 years old vs the entire study population, with less grade 3/4<br />

neutropenia (P � 0.05) and neuropathy (P � 0.05) and increased<br />

thrombocytopenia (P � NS) and anemia (P � 0.05) in the nab-P/C vs<br />

sb-P/C arms. In pts �70 years old, patient-reported FACT-taxane subscales<br />

revealed significantly less neuropathy, pain, and hearing loss in the nab-P/C<br />

vs sb-P/C arms (P � 0.001). Conclusions: In elderly pts with advanced<br />

NSCLC, nab-P/C as first-line therapy was well tolerated and led to improved<br />

ORR and PFS, with significantly longer OS vs sb-PC.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


502s Lung Cancer—Non-small Cell Metastatic<br />

7591 General Poster Session (Board #51A), Sat, 1:15 PM-5:15 PM<br />

EML4-ALK-gene rearrangement comparative analysis in circulating tumor<br />

cells and in tumor tissue <strong>of</strong> patients with lung adenocarcinoma. Presenting<br />

Author: Marius Ilie, Laboratory <strong>of</strong> <strong>Clinical</strong> and Experimental Pathology,<br />

Hospital Pasteur, Academic Hospital, Nice, France<br />

Background: The implementation <strong>of</strong> new theranostic biomarkers in Oncology<br />

is leading to impressive therapeutic improvements. In patients with<br />

lung cancer, the possibility to use Circulating Tumor Cells (CTCs) as a<br />

non-invasive theranostic approach is a clinically appealing challenge.<br />

Adenocarcinomas with EML4-ALK rearrangement are a new molecular<br />

subgroup <strong>of</strong> lung tumors with very good response to Crizotinib, an ALK<br />

inhibitor. We have thus aimed at developing an informative assay characterizing<br />

the ALK-gene status in CTCs isolated from patients with lung cancer.<br />

Methods: CTCs were isolated preoperatively using Isolation by Size <strong>of</strong><br />

Epithelial Tumor cells method (ISET) from 65 patients with lung adenocarcinoma<br />

and blindly screened for ALK-gene status. ALK break-apart<br />

fluorescence in situ hybridization (FISH) (LSI ALK dual colour probes set)<br />

and immunochemistry using an anti-ALK antibody (5A4 clone) were blindly<br />

performed on CTCs and corresponding tumor tissues and results were<br />

compared. Results: Two patients consistently showed ALK-gene rearrangement<br />

and strong ALK protein expression in CTCs and corresponding tumor<br />

samples. Negative results (both ALK FISH and ALK immunochemistry)<br />

were found in CTCs and corresponding tumor samples from the other 63<br />

patients. Conclusion: We have developed an approach allowing to characterize<br />

ALK-gene status in CTCs from patients with lung cancer and shown<br />

consistent results in CTC and tumor tissues. These preliminary results<br />

encourage larger studies and open new avenues for non-invasive, real-time,<br />

theranostic monitoring <strong>of</strong> cancer patients.<br />

7593 General Poster Session (Board #51C), Sat, 1:15 PM-5:15 PM<br />

Frequency <strong>of</strong> anaplastic lymphoma kinase (ALK) positive tumors among<br />

African <strong>American</strong> non-small cell lung cancer (NSCLC) patients. Presenting<br />

Author: Shirish M. Gadgeel, Karmanos Cancer Institute, Wayne State<br />

University, Detroit, MI<br />

Background: ALK gene rearrangement is a recently identified molecular<br />

alteration in the tumors <strong>of</strong> a small proportion <strong>of</strong> NSCLC patients. Previous<br />

reports have shown that the rate <strong>of</strong> EGFR gene mutations may vary in<br />

NSCLC patients <strong>of</strong> different ethnic background. It is unclear if similar<br />

differences exist with regards to ALK positivity. We analyzed ALK expression<br />

in tumors <strong>of</strong> AA NSCLC patients and also assessed the epidemiologic<br />

features and outcomes <strong>of</strong> these patients, as well as the occurrence <strong>of</strong><br />

known oncogene mutations. Methods: We identified 260 tumor tissues <strong>of</strong><br />

AA NSCLC patients, enrolled on several epidemiologic studies conducted<br />

at our institution. Immunohistochemistry, using the anti-Alk D5F3 antibody<br />

(Cell Signaling Technology) was used to visualize ALK expression.<br />

Fifty-three <strong>of</strong> these tumors also underwent mutation analysis using the<br />

OncoCarta Panel V1 (Sequenom) to evaluate 238 mutations in 19<br />

oncogenes. Results: Of the 260 tumors analyzed, 6 (2.3%, 95% CI-<br />

0.5-4.1%) were ALK positive. All ALK positive patients had adenocarcinoma<br />

histology (6/157) while no patients with other NSCLC histologies<br />

were ALK positive, p�0.04. There was no difference in the rate <strong>of</strong> positivity<br />

based on sex, 1% (1/99) in males versus 3.1% (5/161) in females,<br />

p�0.27. Consistent with prior studies, patients with ALK positive tumors<br />

were slightly younger (median age 56 years vs 61 years, p � 0.27) and the<br />

positive rate was higher in never smokers 11% (2/18) than in ever smokers<br />

1.6%, p�0.06. There was no correlation <strong>of</strong> ALK positivity with the stage <strong>of</strong><br />

the disease at diagnosis. The survival <strong>of</strong> ALK positive and negative patients,<br />

adjusted for age, sex and stage, was similar (HR�0.77; 95% CI 0.19-<br />

3.13). Fifty-three tumors have been assessed for oncogene mutations in<br />

addition to ALK expression. Of these, 2 (3.8%) were ALK positive and<br />

neither <strong>of</strong> these tumors carried other tested mutations such as EGFR, Kras,<br />

PIK3 or AKT. Conclusions: The rate <strong>of</strong> ALK positive tumors among<br />

African-<strong>American</strong> NSCLC patients is similar to the general population and<br />

the demographic features <strong>of</strong> ALK positive AA patients is also similar to the<br />

general ALK positive NSCLC patient population.<br />

7592 General Poster Session (Board #51B), Sat, 1:15 PM-5:15 PM<br />

Safety and efficacy analysis by histology <strong>of</strong> weekly nab-paclitaxel in<br />

combination with carboplatin as first-line therapy in patients (pts) with<br />

advanced non-small cell lung cancer (NSCLC). Presenting Author: Markus<br />

Frederic Renschler, Celgene Corporation, Summit, NJ<br />

Background: Treatment <strong>of</strong> advanced NSCLC differs by histology, with fewer<br />

options and poorer outcomes in pts with squamous histology. In a phase III<br />

trial <strong>of</strong> nab-paclitaxel (nab-P, 130 nm albumin-bound paclitaxel particles)<br />

� carboplatin (C) vs solvent-based paclitaxel (sb-P) � C, the primary<br />

endpoint <strong>of</strong> ORR was significantly improved from 25% to 33%, p � 0.005,<br />

with a 1-month improvement in OS (p � NS) and improved safety. This<br />

analysis evaluated efficacy and safety by histology. Methods: Pts with<br />

untreated stage IIIB/IV NSCLC were randomized 1:1 (stratified by age,<br />

histology, region, stage, and gender) to C AUC 6 day 1 and either nab-P 100<br />

mg/m2 on days 1, 8, 15 or sb-P 200 mg/m2 day1q21days. ORR and PFS<br />

were determined by blinded centralized review. Results: In squamous pts,<br />

nab-P/C produced a significantly higher ORR (41% vs 24%, p � 0.001),<br />

similar PFS (5.6 vs 5.7 mo, HR: 0.865) and �1-month improvement in OS<br />

(10.7 vs 9.5 mo, HR: 0.890) vs sb-P/C (Table). nab-P/C was as effective as<br />

sb-P/C in nonsquamous pts for ORR (26% vs 25%, p � 0.808), PFS (6.9<br />

vs 6.5 mo, HR: 0.933), and OS (13.1 vs 13.0 mo, HR: 0.950). In both<br />

squamous and nonsquamous pts, nab-P/C vs sb-P/C produced lower rates<br />

<strong>of</strong> grade 3/4 neuropathy (3% vs 11% and 3% vs 12%, respectively, p �<br />

0.001 both), neutropenia (43% vs 51%, p � NS, and 50% vs 63%, p �<br />

0.008), and higher but manageable rates <strong>of</strong> anemia (27% vs 4% and 28%<br />

vs 9%, p � 0.001 both) and thrombocytopenia (21% vs 7% and 16% vs<br />

11%, p � 0.001 both). Conclusions: In pts with advanced NSCLC, nab-P/C<br />

demonstrated a favorable risk-benefit pr<strong>of</strong>ile as a first-line therapy regardless<br />

<strong>of</strong> histology. Significantly improved ORR and a positive trend in OS<br />

were observed in pts with squamous histology.<br />

Squamous<br />

ORR, %<br />

95% CI<br />

Median PFS, mo<br />

95% CI<br />

Median OS, mo<br />

95% CI<br />

Nonsquamous<br />

ORR, %<br />

95% CI<br />

Median PFS, mo<br />

95% CI<br />

Median OS, mo<br />

95% CI<br />

nab-P/C sb-P/C RR/HR P<br />

n � 229<br />

41<br />

35, 47<br />

5.6<br />

5.5, 6.7<br />

10.7<br />

9.4, 12.5<br />

n � 292<br />

26<br />

21, 31<br />

6.9<br />

6.0, 8.1<br />

13.1<br />

11.5, 16.1<br />

n � 221<br />

24<br />

19, 30<br />

5.7<br />

5.4, 6.7<br />

9.5<br />

8.6, 11.6<br />

n � 310<br />

25<br />

20, 30<br />

6.5<br />

5.6, 7.0<br />

13.0<br />

11.1, 14.3<br />

1.680<br />

1.271, 2.221<br />

0.865<br />

0.680, 1.101<br />

0.890<br />

0.719, 1.101<br />

1.304<br />

0.788, 1.358<br />

0.933<br />

0.750, 1.159<br />

0.950<br />

0.779, 1.158<br />

�0.001<br />

0.245<br />

0.284<br />

0.808<br />

0.532<br />

0.611<br />

7594 General Poster Session (Board #51D), Sat, 1:15 PM-5:15 PM<br />

Update on the large-scale screening <strong>of</strong> ALK fusion oncogene transcripts in<br />

archival NSCLC tumor specimens using multiplexed RT-PCR assays.<br />

Presenting Author: Tianhong Li, University <strong>of</strong> California, Davis, Sacramento,<br />

CA<br />

Background: The ALK inhibitor crizotinib <strong>of</strong>fers a new standard <strong>of</strong> care for<br />

advanced NSCLC patients with EML4-ALK fusion oncogenes. We previously<br />

reported a 4.0% frequency <strong>of</strong> EML4-ALK fusion oncogene transcripts<br />

detected in 1889 NSCLC specimens in the RGI database (Li et al., ASCO<br />

2011). Methods: Patented single and multiplexed RT-PCR assays suitable<br />

for rapid and accurate detection <strong>of</strong> all variants <strong>of</strong> ALK fusion oncogene<br />

transcripts were used as previously described, including all 9 known<br />

EML4-ALK fusion gene transcripts and ALK RNA levels (Danenberg, ASCO<br />

2010). The sensitivity and specificity on archival formalin-fixed, paraffinembedded<br />

tumor specimens are 99% and 100%, respectively. We here<br />

update the detection <strong>of</strong> EML4-ALK fusion transcripts in the RGI database.<br />

Results: Between 12/2009 and 09/2011, 4750 NSCLC specimens in the<br />

RGI database were tested for the presence <strong>of</strong> ALK fusion transcripts. We<br />

found 152 (3.2%) NSCLC cases with EML4-ALK fusion positivity, including<br />

87 (57.2%) V1, 15 (9.9%) V2, 47 (30.9%) V3, and 3 (2.0%) V5a<br />

variants. Median age (range): 61.1 (33-96). Female: 74 (49%). All<br />

EML4-ALK-positive tumors were adenocarcinomas. No EGFR or K-Ras<br />

mutation was detected in ALK fusion-positive samples. Expression <strong>of</strong><br />

chemotherapy-related biomarkers was available from 63 (female: 31,<br />

49%) EML4-ALK-positive cases in the database: 43 (68%) had low TS<br />

level <strong>of</strong> �2.33; 40 (63.5%) had low ERCC1 level <strong>of</strong> �1.7, and 25 (40%)<br />

had low RRM1 level <strong>of</strong> �0.97. Conclusions: This RT-PCR assay provides a<br />

tool for rapid, large-scale screening <strong>of</strong> NSCLC FFPE tissues for EML4-ALK<br />

fusion gene transcripts. The relative value <strong>of</strong> this RT-PCR assay as a<br />

companion diagnostic test for drugs targeting ALK merits evaluation in<br />

comparison with the FDA approved ALK FISH test.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


7595 General Poster Session (Board #51E), Sat, 1:15 PM-5:15 PM<br />

Survival outcome segregated by KRAS mutation status in newly diagnosed<br />

stage IV non-small cell lung cancer (NSCLC) patients (pts) treated with<br />

first-line chemotherapy. Presenting Author: Anna K. Brady, University <strong>of</strong><br />

Pennsylvania, Philadelphia, PA<br />

Background: KRAS mutations (MT) form a distinct subset <strong>of</strong> NSCLC,<br />

generally considered to have poor prognosis. Although KRAS MT is a<br />

well-establised prognostic and predictive marker in colorectal cancer, its<br />

role in NSCLC remains ambiguous. Pts with KRAS MT NSCLC do not<br />

respond well to epidermal growth factor receptor (EGFR) directed tyrosine<br />

kinase inhibitor therapy, but little is known about the ability <strong>of</strong> KRAS MT to<br />

predict outcome after first-line chemotherapy in newly diagnosed advanced<br />

or recurrent NSCLC. Methods: We analyzed outcomes <strong>of</strong> consecutive pts<br />

with newly identified Stage IV non- squamous NSCLC treated at University<br />

<strong>of</strong> Pennsylvania (Penn) between 05/2008 and 7/2010 and then compared<br />

survival based on KRAS status [MT vs wild type (WT)] using chi square,<br />

Kaplan-Meier methods, and Cox regression models. Results: Of 106<br />

consecutive new pts with Stage IV non squamous NSCLC treated at Penn,<br />

49 (46%) underwent molecular analysis for KRAS MT. Fifteen (34%) were<br />

KRAS MT. Of 34 KRAS WT pts, 6 were positive for EGFR MT. The median<br />

age <strong>of</strong> all 49 pts was 61 years; 83% were Caucasian, 45% male and 60%<br />

had a �10 pack year smoking history. Median duration <strong>of</strong> follow up is 16.4<br />

mos. Majority <strong>of</strong> pts (92%) had adenocarcinoma histology. Most pts (88%)<br />

had ECOG PS 0-1 at presentation. Forty three pts received first line<br />

platinum-based combination chemotherapy (platinum and pemetrexed in<br />

31 pts). KRAS MT was associated with smoking (p�0.04), but not with<br />

gender or age. Overall survival (OS) <strong>of</strong> pts with KRAS MT was similar to<br />

KRAS WT pts [median OS 15.6 vs. 19.0 mos; HR 1.24 (95% CI<br />

0.57-2.67)]. Univariate analyses demonstrated superior OS among women<br />

compared to men (HR 0.40, 95% CI 0.20-0.85) in the entire pt cohort.<br />

Conclusions: In our population <strong>of</strong> stage IV non squamous NSCLC, pts with<br />

KRAS MT had similar OS to those with KRAS WT tumors. OS was slightly<br />

higher in KRAS WT pts, but this may have been confounded by the<br />

inclusion <strong>of</strong> 6 EGFR MT pts in this cohort. The potential prognostic or<br />

predictive role <strong>of</strong> KRAS MT in NSCLC pts undergoing chemotherapy<br />

requires further prospective study.<br />

7597 General Poster Session (Board #51G), Sat, 1:15 PM-5:15 PM<br />

Insulin-like growth factor 1 (IGF-1R) protein expression (PE) and gene copy<br />

number (GCN) for discrimination <strong>of</strong> response and outcome to figitumumab<br />

in NSCLC. Presenting Author: Murry W. Wynes, Division <strong>of</strong> Medical<br />

Oncology, University <strong>of</strong> Colorado Denver, Aurora, CO<br />

Background: Early clinical data suggested figitumumab (F), an anti-IGF-1R<br />

antibody, had clinical activity in several tumor types. However, two phase<br />

III studies, A4021016 carboplatin/paclitaxel (CP) �/- F in 1st line<br />

treatment or A4021018 erlotinib (E) �/- F in 2nd line therapy for patients<br />

with advanced NSCLC, were closed prematurely due to futility. Neither<br />

study used biomarker driven selection criteria. The aim <strong>of</strong> this study was to<br />

retrospectively examine the phase III specimens in order to determine if<br />

IGF-1R PE or GCN could discriminate response and/or outcome to F.<br />

Methods: IGF-1R PE was determined by immunohistochemistry (IHC) and<br />

the tumor specimens were scored using the H-score method (0-300) with<br />

separate enumeration <strong>of</strong> membranous and cytoplasmic components and<br />

then combination <strong>of</strong> these scores. GCN was evaluated by bright field silver<br />

in situ hybridization (SISH) with recording the average copy number for 50<br />

tumor cells. Results: IHC was evaluable in 25 CP�F, 20 CP, 27 E�F and<br />

26 E specimens whereas SISH was evaluable in 24, 17, 21 and 22<br />

specimens, respectively. Median PE for membrane, cytoplasm and both in<br />

A4021016 were 130, 110 and 120 and in A4021018 they were 135, 140<br />

and 140. Median GCN was 1.88 and 1.98 for A4021016 and A4021018,<br />

respectively. There were no significant differences in PE or GCN between<br />

treatment arms within each study. Neither PE nor GCN were able to<br />

discriminate between response/non-response or disease control/progression<br />

in either study. Likewise, neither study showed a significant difference<br />

in OS or PFS when using the median cut-points for either PE or GCN.<br />

Conclusions: Neither IGF1R PE or GCN were able to discriminate response<br />

or outcome in the limited quantity <strong>of</strong> specimens available from two studies<br />

that examined the IGF-1R targeted therapy figitumumab.<br />

Lung Cancer—Non-small Cell Metastatic<br />

7596 General Poster Session (Board #51F), Sat, 1:15 PM-5:15 PM<br />

Visual effects in anaplastic lymphoma kinase (ALK)-positive advanced<br />

non-small cell lung cancer (NSCLC) patients treated with crizotinib.<br />

Presenting Author: Ravi Salgia, The University <strong>of</strong> Chicago, Chicago, IL<br />

Background: Crizotinib is an ALK inhibitor indicated in the US for advanced<br />

ALK-positive NSCLC. Visual effects reported by patients treated with<br />

crizotinib were characterized using the Visual Symptom Assessment<br />

Questionnaire (VSAQ). Methods: Patients with previously treated, advanced<br />

ALK-positive NSCLC were administered 250 mg BID crizotinib in an<br />

ongoing phase II study (PROFILE1005, NCT00932451; Pfizer). Patients<br />

completed the VSAQ at day 1 <strong>of</strong> each 21 day cycle and at end <strong>of</strong> treatment.<br />

The VSAQ has a recall period <strong>of</strong> 3 weeks and consists <strong>of</strong> 7 questions<br />

assessing presence, frequency, timing, duration and degree <strong>of</strong> bother <strong>of</strong><br />

visual effects and their impact on Activities <strong>of</strong> Daily Living (ADL). The<br />

visual effects assessed included appearance <strong>of</strong> overlapping shadows/after<br />

images, flashing lights and streamers/strings/floaters, difficulty adapting to<br />

lights and seeing at night. Patients rated degree <strong>of</strong> bother on a 5-point scale<br />

ranging from ‘not at all’ to ‘extremely’. Impact on ADL was measured using<br />

a 10-point scale (0: no effect; 10: completely prevented ADL). Frequency<br />

analyses were performed. Results: As <strong>of</strong> June 1 2011, visual effects as<br />

identified by VSAQ were reported by 63% (114/182) <strong>of</strong> patients at cycle 2<br />

(C2), 57% at C3 (85/149), 52% at C4 (64/123) and 41% at C5 (46/112).<br />

The most commonly experienced visual events were appearance <strong>of</strong> flashing<br />

lights (C2:81%; C3:82%; C4:84%; C5: 76%), streamers/strings/floaters<br />

(C2: 83%; C3:78%; C4: 81%; C5:87%) and overlapping shadows/after<br />

images (C2:70%; C3:77%; C4:87%; C5:84%). Most patients reported<br />

each event to last �1 minute (C2:61%; C3:71%; C4:77%; C5: 70%).<br />

Majority <strong>of</strong> patients reported event frequency at each cycle <strong>of</strong> � 7 days/wk<br />

(50–78%). Patients reported that the visual effects occurred mostly in the<br />

morning (52–62%) and/or evening (62–73%). Majority <strong>of</strong> patients reported<br />

that visual effects were not at all or a little bothersome (C2:62%;<br />

C3:61%; C4:66%; C5:65%). Majority <strong>of</strong> patients indicated no or minimal<br />

impact on ADL (C2:80%; C3:80%; C4:83%; C5:87%). Conclusions: Visual<br />

effects identified by VSAQ in patients treated with crizotinib were frequent,<br />

but were reported to be transient with no or minimal impact on ADL.<br />

7598 General Poster Session (Board #51H), Sat, 1:15 PM-5:15 PM<br />

<strong>Clinical</strong> presentation <strong>of</strong> hepatotoxicity-associated crizotinib in ALKpositive<br />

(ALK�) advanced non-small cell lung cancer (NSCLC). Presenting<br />

Author: Patrick Schnell, Pfizer Inc., New York, NY<br />

Background: Severe hepatotoxicity has been observed with most receptor<br />

tyrosine kinase inhibitors. Crizotinib, a first-in-class oral inhibitor <strong>of</strong> ALK,<br />

was recently FDA approved for the treatment (tx) <strong>of</strong> advanced (ALK�)<br />

NSCLC. We provide a review <strong>of</strong> hepatotoxicity in the crizotinib clinical<br />

development program. Methods: Relevant tx-emergent adverse event (AE)<br />

reports from 2 pooled single-arm trials in patients (pts) with ALK� NSCLC<br />

who received crizotinib 250 mg BID (n�588) up to Jun 2011 were<br />

reviewed. Five reports <strong>of</strong> severe hepatotoxicity received through Dec 13,<br />

2011 were also evaluated. Results: Incidence <strong>of</strong> elevation in ALT and AST<br />

lab values by CTCAE grade are shown in the table. Additionally, 16<br />

all-grade (7 grade 3 or 4) cases <strong>of</strong> pertinent hepatic events have been<br />

reported. Transaminase elevations generally occurred in the first 2 months<br />

<strong>of</strong> tx and were usually reversible. Pts usually resumed tx at a lower dose<br />

without recurrence; however, 4 (�1%) required permanent discontinuation<br />

from tx. Five (�1%) cases <strong>of</strong> severe, potentially drug-related hepatotoxicity<br />

(2 fatal) were reported through Dec 13, 2011, with an estimated exposure<br />

<strong>of</strong> 1400 pts. Pts presented with fatigue, nausea, anorexia, weakness, and<br />

abdominal pain. Time from starting crizotinib to onset <strong>of</strong> hepatotoxicity was<br />

�6 weeks in 4 cases, while significant transaminase elevation was<br />

observed after 6 months in the fifth case. Significant hepatic metastases<br />

and abnormal enzymes at baseline were present in 1 (fatal) case, while<br />

baseline transaminases were normal in 3 cases, or just above the upper<br />

limit <strong>of</strong> normal in 1 case. Conclusions: Crizotinib is uncommonly associated<br />

with severe hepatotoxicity and was reported in �1% <strong>of</strong> pts. Nausea,<br />

malaise, decreased appetite, vomiting, and abdominal pain may indicate<br />

hepatic toxicity. ALT and total bilirubin should be monitored monthly and<br />

as clinically indicated. Discontinuation <strong>of</strong> crizotinib is recommended when<br />

severe drug-induced hepatic toxicity is suspected or diagnosed.<br />

Causality/grade<br />

N � 588<br />

Increased ALT<br />

n (%)<br />

503s<br />

Increased AST<br />

n (%)<br />

All causality<br />

All grade 86 (14.6) 63 (10.7)<br />

Grade 3/4 30 (5.1) 14 (2.4)<br />

Tx related<br />

All grade 73 (12.4) 52 (8.8)<br />

Grade 3/4 24 (4.1) 10 (1.7)<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


504s Lung Cancer—Non-small Cell Metastatic<br />

7599 General Poster Session (Board #52A), Sat, 1:15 PM-5:15 PM<br />

A large retrospective analysis <strong>of</strong> the activity <strong>of</strong> pemetrexed (PEM) in<br />

patients (pts) with ALK-positive (ALK�) non-small cell lung cancer<br />

(NSCLC) prior to crizotinib (CRIZ). Presenting Author: Giorgio V. Scagliotti,<br />

University <strong>of</strong> Turin, Orbassano, Italy<br />

Background: Retrospective small cohort studies suggest ALK� NSCLC may<br />

be particularly sensitive to PEM. Methods: PROFILE 1005 (NCT00932451;<br />

Pfizer) is a large phase 2 multi-center, single-arm study <strong>of</strong> CRIZ in<br />

previously treated ALK� NSCLC. Eligibility criteria included pts with<br />

progressive disease from the PEM arm <strong>of</strong> companion trial PROFILE 1007.<br />

We retrospectively assessed objective response rate (ORR) and time to<br />

progression (TTP; 1st dose to objective progression) in pts who received<br />

PEM prior to CRIZ. ORR with CRIZ post-PEM was assessed. Results: Of the<br />

439 ALK� pts enrolled as <strong>of</strong> June 2011, 369 (84.1%) received prior PEM<br />

(single agent or combination; any line advanced/metastatic) with a cumulative<br />

ORR <strong>of</strong> 18.7%. In pts who received PEM combinations 1st-line (1L;<br />

n�120) or 2nd-line (2L; n�60), ORR was 24.2% and 16.7%; and median<br />

TTP was 6.9 months (mo; 95% CI: 6.0–7.4) and 6.9 mo (95% CI:<br />

4.8–8.8), respectively. In pts who received 2L single-agent PEM (n�80),<br />

ORR was 12.5% and median TTP was 5.3 mo (95% CI: 3.0–6.6). In pts<br />

treated with PEM 3rd-line (3L) or later (n�138), ORR was 16.7%. By line<br />

<strong>of</strong> PEM treatment, ORR was lower and TTP shorter than that reported in<br />

small ALK� cohorts evaluating multiple lines <strong>of</strong> treatment. In 2L, previously<br />

reported ORR and median PFS with single-agent PEM in adenocarcinoma<br />

NSCLC were 12.8% (n�158) and 3.5 mo (n�283), and comparable<br />

to our findings. Pts who received 2L single-agent PEM (n�80) subsequently<br />

achieved higher ORR with CRIZ (54%; 95% CI: 42–65). Similar<br />

analyses in the 1L and 3L groups are ongoing. Conclusions: This retrospective<br />

study in predominantly never-smokers with ALK� NSCLC reports lower<br />

ORR and shorter TTP with PEM than that reported in smaller retrospective<br />

ALK� NSCLC cohorts. This finding may be in part due to the inclusion <strong>of</strong><br />

crossover pts from PROFILE 1007. This analysis and previous reports<br />

observed a tendency to a higher response rate and better PFS or TTP with<br />

PEM than in unselected populations in 2L, which may not be specifically<br />

related to ALK status but to a higher sensitivity to cytotoxic agents in<br />

never-smokers. Results from an ongoing phase 3 trial (PROFILE 1007) will<br />

provide additional information.<br />

7601 General Poster Session (Board #52C), Sat, 1:15 PM-5:15 PM<br />

<strong>Clinical</strong> benefit from pemetrexed before and after crizotinib exposure in<br />

patients with ALK positive non-small cell lung cancer (ALK� NSCLC).<br />

Presenting Author: Eamon Berge, University <strong>of</strong> Colorado Health Sciences<br />

Center, Aurora, CO<br />

Background: Crizotinib (Criz) produces high response rates and prolonged<br />

progression free survival (PFS) in ALK� NSCLC. Retrospective analyses<br />

suggest enhanced sensitivity to pemetrexed (Pem) in Criz-naïve ALK�<br />

NSCLC. Different biological mechanisms <strong>of</strong> Criz resistance have recently<br />

been discovered. <strong>Clinical</strong> cross-resistance between Criz and Pem has not<br />

been previously investigated. Methods: Patients with stage IV ALK� NSCLC<br />

treated in sequence with Pem then Criz (Pem-Criz), or Criz then Pem<br />

(Criz-Pem) were identified. Excluding CNS only progression events when no<br />

new systemic treated was started, PFS was compared using Cox Proportional<br />

Hazards. Correlations between molecular features <strong>of</strong> ALK positivity<br />

and Pem PFS were analyzed using Spearman correlations. Treatment<br />

sequence effect was explored using Fisher’s exact test. Results: We<br />

identified 19 Pem-Criz and 9 Criz-Pem ALK� patients. Pem was given as<br />

monotherapy in 37% vs. 66%, and as median 2nd vs. 3rd line cytotoxic in<br />

the Pem-Criz and Criz-Pem groups, respectively. For the Pem-Criz group,median<br />

PFS was 8.9 months with Pem (range: 1-21.5 months), and 14.7<br />

months with Criz (range:2.9� -27.5 months). For the Criz-Pem group,<br />

median PFS was 8.4 months with Criz (range: 2-29 months), and 4.4<br />

months with Pem (range: 0.5-10.5� months). Patients were more likely to<br />

progress on Pem given after Criz than before, but confidence intervals were<br />

wide (HR 1.51, 95% CI 0.626-3.66). Neither native or rearranged signal<br />

copy number, nor percent cells ALK� in the tumors correlated with PFS on<br />

Pem. Conclusions: Both Criz and Pem appear to be active drugs in<br />

ALK�NSCLC given in either order. Numerically, benefit from Pem was less<br />

when given post-Criz compared to pre-Criz, however this difference was not<br />

statistically significant. Several potential confounders exist in our small<br />

dataset (notably differences in line <strong>of</strong> therapy and use in combo vs.<br />

monotherapy). To fully address whether there may be a significant<br />

difference in overlap <strong>of</strong> induced specific drug resistance mechanisms, the<br />

clinical effect <strong>of</strong> Criz/Pem sequencing should be explored in a larger series<br />

adjusting for confounding effects.<br />

7600 General Poster Session (Board #52B), Sat, 1:15 PM-5:15 PM<br />

<strong>Clinical</strong> characteristics <strong>of</strong> ALK� NSCLC patients (pts) treated with crizotinib<br />

beyond disease progression (PD): Potential implications for management.<br />

Presenting Author: Gregory Alan Otterson, Department <strong>of</strong> Internal<br />

Medicine, The Ohio State University Comprehensive Cancer Center, Columbus,<br />

OH<br />

Background: Crizotinib is a first-in-class oral ALK inhibitor for the treatment<br />

(tx) <strong>of</strong> advanced ALK-positive (ALK�) NSCLC. Dramatic and prolonged<br />

responses to crizotinib are common, but pts do experience PD. Methods:<br />

<strong>Clinical</strong> characteristics <strong>of</strong> ALK� NSCLC pts enrolled onto two multicenter,<br />

single arm trials <strong>of</strong> crizotinib (A8081001, PROFILE 1005) with investigatordefined<br />

PD who were allowed to continue crizotinib if, in the investigator’s<br />

opinion, there was reasonable evidence <strong>of</strong> ongoing clinical benefit were<br />

assessed. A period <strong>of</strong> �2 wks was chosen as a reasonable minimum<br />

duration <strong>of</strong> post-PD crizotinib tx. Results: As <strong>of</strong> 1 June 2011, 146 pts<br />

(A8081001, n�61, PROFILE 1005, n�85) had PD. PD was observed in<br />

new lesions only (1 organ site, n�62; �1 organ site, n�18), target � new<br />

lesions (� 1 organ site, n�55), clinical PD (n�9), and no site assessment<br />

(n�2). Most common new lesions in single organ sites were brain (brain<br />

MRI not mandatory; n�25), liver (n�20), bone (n�4), and kidney (n�1).<br />

Of the 146 pts, 78 (53%) received crizotinib post-PD for at least 2 wks,<br />

91% <strong>of</strong> whom had ECOG PS 0 or 1 at PD. In these 78 pts, PD was observed<br />

in new lesions only (1 organ site, n�39; �1 organ site, n�4), target � new<br />

lesions (� 1 organ site, n�29), and clinical PD (n�6); the most common<br />

sites for single organ PD were brain (n�20), liver (n�9), and other (n�10).<br />

Best response before PD (% CR/PR, SD, PD) was 62/27/12 in the 78 pts<br />

who received �2 weeks’ tx post-PD and 31/37/32 in the 68 pts who<br />

received �2 weeks’ tx post-PD or discontinued. Median duration <strong>of</strong><br />

crizotinib tx post-PD (n�78) was 10 weeks (range 2–84). 20 pts with PD in<br />

brain only (concurrent local tx or radiation permitted) continue to receive<br />

crizotinib (range 3–82 weeks post-PD). Conclusions: Following initial<br />

crizotinib tx, PD most commonly occurred at a single organ site in ALK�<br />

NSCLC pts. The majority <strong>of</strong> pts receiving crizotinib post-PD had good PS,<br />

and tended to have single-site PD (most <strong>of</strong>ten the brain or liver), and a prior<br />

response on crizotinib. Given these observations, pts may be able to<br />

continue with crizotinib for a period <strong>of</strong> time following clinical or documented<br />

progression.<br />

7602 General Poster Session (Board #52D), Sat, 1:15 PM-5:15 PM<br />

A first-in-human phase I/II study <strong>of</strong> ALK inhibitor CH5424802 in patients<br />

with ALK-positive NSCLC. Presenting Author: Katsuyuki Kiura, Department<br />

<strong>of</strong> Respiratory Medicine, Okayama University Hospital, Okayama, Japan<br />

Background: Anaplastic lymphoma kinase (ALK) is a tyrosine kinase<br />

constitutively activated in a subset <strong>of</strong> non-small cell lung cancer (NSCLC)<br />

following chromosomal gene translocation. CH5424802 was identified as<br />

a potent, selective, and oral ALK inhibitor with a unique chemical scaffold,<br />

showing preferential antitumor activity against NSCLC cells expressing<br />

EML4-ALK fusion in vitro and in vivo (Cancer Cell, 2011). Our results<br />

support the potential <strong>of</strong> CH5424802 as a new therapeutic opportunity for<br />

patients (pts) with ALK-positive NSCLC. Methods: Pts with ALK-positive<br />

NSCLC received CH5424802 twice daily orally until progressive disease or<br />

toxicity was observed. In the phase I portion, dose was escalated using an<br />

accelerated titration scheme. The primary objectives were to investigate the<br />

safety, tolerability and pharmacokinetic (PK) parameters under fasting<br />

conditions, and to determine the recommended dose (RD) for use in the<br />

phase II portion. The primary objectives <strong>of</strong> the phase II portion were to<br />

investigate the efficacy and safety at the RD. The phase I portion was<br />

amended to include an investigation under non-fasting conditions in<br />

addition to fasting conditions. Results: 15 pts (M/F: 9/6) were treated with<br />

CH5424802 under fasting conditions in 6 cohorts (20 mg bid to 300 mg<br />

bid) and 9 pts (M/F: 2/7) were treated under non-fasting conditions in 2<br />

cohorts (240 mg bid and 300 mg bid). Median age was 42.5 years. The<br />

highest dose level defined in the protocol (300 mg bid) did not reach the<br />

MTD. Thus, a DLT was not determined. Toxicities were mild to moderate.<br />

The most frequent toxicity was grade 1 myalgia. Grade 3 toxicity (cases)<br />

occurred as follows; hypophosphatemia (2), neutropenia (2), blood CPK<br />

increased (1) and hypermagnesemia (1). PK data from the fasting cohorts<br />

showed dose-dependent increases <strong>of</strong> Cmax and AUC. All pts at all dose<br />

levels achieved tumor regression. At dose levels 240 mg bid or more under<br />

fasting conditions, all 7 pts with measurable lesions achieved a partial<br />

response. So far, 11 pts have been on study treatment � 6 months.<br />

Conclusions: CH5424802 was well tolerated with promising efficacy in pts<br />

with ALK-positive NSCLC. The phase II portion is ongoing.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


7603 General Poster Session (Board #52E), Sat, 1:15 PM-5:15 PM<br />

MIMEB: A phase II trial to evaluate FDG-PET/FLT-PET and DCE-MRI for<br />

early prediction <strong>of</strong> efficacy in patients with advanced non-small cell lung<br />

cancer treated with erlotinib and bevacizumab. Presenting Author: Matthias<br />

Scheffler, Lung Cancer Group Cologne, Department I <strong>of</strong> Internal<br />

Medicine and Center for Integrated Oncology, University Hospital Cologne,<br />

Cologne, Germany<br />

Background: Since the introduction <strong>of</strong> targeted therapy into the treatment <strong>of</strong><br />

NSCLC, molecular imaging tools gain in importance for assessment <strong>of</strong><br />

pharmacodynamics, pharmacokinetics and prediction <strong>of</strong> therapeutic outcome.<br />

Tumor metabolism (by FDG-PET), proliferation (by FLT-PET), and<br />

vascularization (by DCE-MRI) can be noninvasively assessed to quantify the<br />

effects <strong>of</strong> targeted therapy on tumor biology. We set up a trial to evaluate<br />

the effects <strong>of</strong> combined erlotinib (E) and bevacizumab (B) treatment in<br />

patients with advanced non-squamous cell NSCLC and to identify prospectively<br />

patients who benefit from this combination. Methods: Patients with<br />

non-squamous NSCLC stage IV without prior systemic therapy received at<br />

least six weeks <strong>of</strong> combined E and B. FLT and FDG-PET scans as well as<br />

DCE-MRI-scans were performed at baseline, after 1 week <strong>of</strong> therapy and<br />

after 6 weeks <strong>of</strong> therapy. Standard uptake values <strong>of</strong> the PET scans and<br />

vascularization parameters <strong>of</strong> the DCE-MRI scans were analyzed and<br />

coregistrated. Tumor specimens were analysed within a network screening<br />

panel. The primary objective <strong>of</strong> this trial was to evaluate the accuracy <strong>of</strong> the<br />

imaging tools for early prediction <strong>of</strong> nonprogression and PFS and its<br />

association with molecular markers. Results: Of the 40 patients, all<br />

received FDG-PET analyses, whereas FLT-PET was performed in 38<br />

patients and DCE-MRI in 36 patients. Median OS was 13.4 months,<br />

median PFS was 5.9 months. Until January 2012, tissue specimens from<br />

25 patients have been genetically analysed. First results show that even<br />

patients with KRAS, PI3K and BRAF mutations pr<strong>of</strong>iting from the combination<br />

either by response or prolonged PFS could be identified by early<br />

imaging assessment. Conclusions: The efficacy <strong>of</strong> E �B in unselected<br />

patients with NSCLC was comparable with platinum-based chemotherapy.<br />

In order to identify imaging-based predictive biomarkers to identify the<br />

subpopulation <strong>of</strong> patients with pronounced benefit <strong>of</strong> this combination<br />

FDG-, FLT- PET and DCE–MRI were successfully implemented in the<br />

treatment plan. Results <strong>of</strong> the imaging analysis and the correlation with<br />

tissue-based biomarkers will be presented.<br />

7605 General Poster Session (Board #52G), Sat, 1:15 PM-5:15 PM<br />

Impact <strong>of</strong> disease progression (DP) date determination method on post<br />

progression survival (PPS) and DP metrics in advanced lung cancer.<br />

Presenting Author: Sumithra J. Mandrekar, Mayo Clinic and NCCTG,<br />

Rochester, MN<br />

Background: Overall survival (OS) can be partitioned into progression-free<br />

survival (PFS) and PPS. PPS helps to understand trial results, especially<br />

when PFS and OS data on trial treatment effects are discordant. At ASCO<br />

2011, we reported that the magnitude <strong>of</strong> difference in the PFS estimates<br />

using different DP date methods was large enough to alter trial conclusions.<br />

Here, we investigate the impact <strong>of</strong> the DP date method on 1) PPS<br />

estimates, and 2) predictive utility <strong>of</strong> DP metrics on subsequent OS (SOS).<br />

Methods: Individual patient (pt) data from 14 trials were pooled. DP date<br />

was determined using: reported progression date (RPD) (method 1, M1),<br />

one day after last progression-free (PF) scan (M2), and midpoint between<br />

last PF scan and RPD (M3). PPS was estimated using the method <strong>of</strong><br />

Kaplan-Meier for the 3 DP date methods. A flexible landmark analysis at 2,<br />

4, and 6 months (mos) using Cox proportional hazards model was used to<br />

assess the impact <strong>of</strong> DP status (progression versus no-progression) on SOS<br />

(using M1, M2, or M3). Results: Among NSCLC (SCLC), 87% (91%) <strong>of</strong> pts<br />

reported DP. As expected, the PPS estimates were the lowest for RPD,<br />

highest for M2, and in-between for M3 (a direct consequence <strong>of</strong> the DP<br />

date method); with no difference by arm for the randomized trials.<br />

Regardless <strong>of</strong> the DP date method, patients who were progression-free had<br />

improved SOS (NSCLC: Hazard ratio, HR��0.33; p � 0.0001; SCLC:<br />

HR��0.48; p � 0.002) at each landmark time point, with comparable<br />

concordance index (SCLC: 0.57-0.65; NSCLC: 0.63-0.67), i.e., ability to<br />

discriminate patients with different SOS outcomes. Conclusions: While the<br />

DP date methods do not impact the predictive utility <strong>of</strong> the DP metrics, they<br />

significantly impact PPS estimates. The translation <strong>of</strong> a significant treatment<br />

effect on PFS to an effect on OS is influenced by PPS (longer PPS<br />

dilutes effect on OS). Standards for declaring DP date are thus critical to<br />

trial design and for trial go/no-go decisions.<br />

Progression date determination<br />

method<br />

NSCLC (10 trials; n�610)<br />

(median OS � 9.6)<br />

Median PFS, and PPS estimates (mos)<br />

SCLC (4 trials; n�114)<br />

(median OS � 8.7)<br />

PFS PPS PFS PPS<br />

Method 1 4.3 3.8 2.8 4.7<br />

Method 2 2.5 5.9 1.2 7.1<br />

Method 3 3.4 4.7 2.0 5.6<br />

Lung Cancer—Non-small Cell Metastatic<br />

505s<br />

7604 General Poster Session (Board #52F), Sat, 1:15 PM-5:15 PM<br />

Meta-analysis <strong>of</strong> progression-free survival as a predictor <strong>of</strong> overall survival<br />

in locally advanced or metastatic non-small cell lung cancer trials.<br />

Presenting Author: Evadne Jane Turner, Boehringer Ingelheim Pty Ltd,<br />

Sydney, Australia<br />

Background: Access to new oncology treatments for non-small-cell lung<br />

cancer (NSCLC) could be expedited if progression free survival (PFS) was<br />

accepted by payers as a valid endpoint predictive for overall survival (OS).<br />

We investigated the relationship between PFS and OS in advanced NSCLC,<br />

which has previously been limited by available data. Methods: A systematic<br />

review <strong>of</strong> the published literature was conducted (1988 to August 2011;<br />

Medline/Embase/Cochrane). Identified studies were assessed against the<br />

following criteria for inclusion in the analysis: randomised design; NSCLC;<br />

advanced disease (Stage IIIb or IV); pharmacological treatment in 2 or more<br />

groups; reported outcomes included median OS and median PFS. For each<br />

pair <strong>of</strong> treatment groups compared, the median OS difference and the<br />

median PFS difference were calculated. The data were analysed with<br />

locally weighted least squares regression (LOWESS) to validate the linear<br />

relationship. Unweighted and weighted linear regression was used to test<br />

the significance <strong>of</strong> the relationship between OS difference and PFS<br />

difference. Results: A total <strong>of</strong> 124 clinical trials, with 40,568 patients,<br />

were included in the analysis. The studies ranged from middle aged cohorts<br />

to the elderly. Median performance status (ECOG/WHO) was 1. 70% <strong>of</strong><br />

studies were first line and therapy type was predominantly chemotherapy<br />

(57%). 19 studies (15%) reported use <strong>of</strong> crossover or rescue therapy.<br />

Weighted linear regression demonstrated a highly significant linear relationship<br />

between OS difference and PFS difference (Table). The results<br />

suggest that a 1 month difference in PFS is associated with approximately<br />

1.3 months difference in OS. Conclusions: Analysis <strong>of</strong> NSCLC trials showed<br />

the treatment effect on PFS was predictive <strong>of</strong> the treatment effect on OS.<br />

With increased use <strong>of</strong> multiple lines <strong>of</strong> treatment and crossover/rescue<br />

therapy in new NSCLC trials OS differences are frequently confounded,<br />

making PFS an important outcome for health care decision making.<br />

Statistic<br />

Parameter Estimate SE 95% CI t-statistic p value<br />

Intercept -0.260 0.209 (-0.674, 0.154) -1.242 0.214<br />

Slope 1.317 0.160 (1.000, 1.634) 8.226 �0.001<br />

R-squared 36.6%<br />

7606 General Poster Session (Board #52H), Sat, 1:15 PM-5:15 PM<br />

Metastatic NSCLC: Treatment patterns, outcomes, and costs <strong>of</strong> newer<br />

agents. Presenting Author: Adrian G. Sacher, Division <strong>of</strong> Medical Oncology<br />

and Hematology, Princess Margaret Hospital, University <strong>of</strong> Toronto, Toronto,<br />

ON, Canada<br />

Background: New therapies for metastatic NSCLC have improved survival in<br />

clinical trials. The increased cost <strong>of</strong> these agents has led to variable drug<br />

funding and treatment across jurisdictions. Here we review patterns,<br />

real-world outcomes and costs <strong>of</strong> metastatic NSCLC treatment in the<br />

province <strong>of</strong> Ontario. Methods: All patients diagnosed with metastatic<br />

NSCLC from 2005-2009 were identified from the Ontario Cancer Registry<br />

with demographic, histologic and mortality data. Treatment records from<br />

the Ontario New Drug Funding Program and centralized treatment database<br />

were linked. Statistical analysis involved Wilcoxon rank sum, Kruskal-<br />

Wallis, Cochran-Armitage trend and likelihood ratio tests where appropriate.<br />

Results: 8113 metastatic NSCLC patients were identified. The median<br />

age was 68; 39% had adenocarcinoma, 14% squamous carcinoma and<br />

43% histology not otherwise specified. Most were treated in regional cancer<br />

centers (92%). The majority (76%) did not receive systemic therapy; 23%<br />

received first-line chemotherapy, most commonly platinum doublets. More<br />

patients received systemic therapy over time (19% in 2005 v 26% in<br />

2009, p �0.0001). Older patients (p �0.0001) and those with squamous<br />

histology (p �0.0001) were less likely to receive systemic therapy. Center<br />

<strong>of</strong> diagnosis did not affect the likelihood <strong>of</strong> treatment (p�0.46). The<br />

median time from diagnosis to death was significantly greater among<br />

patients selected to receive chemotherapy (9.3 v 3.2 months, p �0.0001),<br />

and with cisplatin/gemcitabine compared to other doublets (10.7 v 8.2<br />

months, p�0.004). 31% <strong>of</strong> treated patients received second-line chemotherapy,<br />

predominantly with docetaxel (52%) or pemetrexed (41%).<br />

Pemetrexed use increased significantly over time (8% in 2005 v 71% in<br />

2009, p�0.0001), as did the mean drug cost <strong>of</strong> second-line therapy<br />

($5939/patient in 2005 v $10,057 in 2009). A longer median survival was<br />

also seen with pemetrexed in adenocarcinoma (17.9 v 15.2 months for<br />

docetaxel, p �0.02). Conclusions: Most metastatic NSCLC patients do not<br />

undergo systemic treatment. First- and second-line treatment outcomes in<br />

this population-based study were similar to clinical trials, confirming better<br />

outcomes with new agents at greater cost.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


506s Lung Cancer—Non-small Cell Metastatic<br />

7607 General Poster Session (Board #53A), Sat, 1:15 PM-5:15 PM<br />

Does change in health-related quality <strong>of</strong> life (HRQoL) score predict<br />

survival? Analysis <strong>of</strong> a lung cancer RCT. Presenting Author: Divine Ewane<br />

Ediebah, European Organisation for Research and Treatment <strong>of</strong> Cancer<br />

Headquarters, Brussels, Belgium<br />

Background: Over 60 cancer clinical trials have shown that baseline<br />

health-related quality <strong>of</strong> life (HRQoL) scores are prognostic for patient<br />

survival. Few studies have investigated the added value <strong>of</strong> change in<br />

HRQoL scores. Our aim was to investigate if change in HRQoL scores from<br />

baseline over time is also associated with survival. Methods: We analyzed<br />

data from an EORTC 3-arm randomized clinical trial (RCT) in advanced<br />

non-small-cell lung cancer (NSCLC) patients, comparing<br />

gemcitabine�cisplatin, versus paclitaxel�gemcitabine, versus standard<br />

arm paclitaxel�cisplatin. HRQoL was measured in 394 patients using the<br />

EORTC QLQ-C30 at baseline and after each chemotherapy cycle. The<br />

prognostic significance <strong>of</strong> sex, age and WHO performance status (0-1 vs. 2)<br />

and the 15 QLQ-C30 subscales were assessed with Cox proportional hazard<br />

models stratified for treatment (level <strong>of</strong> significance 0.05). Changes in<br />

HRQoL scores from baseline to each chemotherapy cycle assessment were<br />

categorized as “improved”, “stable” and “worsened” using a threshold <strong>of</strong><br />

10 points difference. Due to expected attrition, the analysis was limited to<br />

changes from baseline up to cycle 3. Results: There were 248 patients in<br />

cycle 1, 212 in cycle 2 and 196 in cycle 3. We performed analyses<br />

separately using data at cycle 1, cycle 2, and cycle 3. In all analyses,<br />

HRQoL in various subscales and socio-demographic and clinical variables<br />

(physical functioning (hazard ratio [HR] 0.91, 95% CI 0.85-0.98;<br />

p�0.0103), pain (1.11, 1.05-1.17; p� 0.0004), age (0.98, 0.97-1.00,<br />

p�0.0413) and WHO performance status (1.77, 1.09-2.89; p�0.0218)<br />

at cycle 1; pain (1.11, 1.03-1.20; p�0.0016), age (0.98, 0.96-1.00;<br />

p�0.0217) and sex (0.63, 0.42-0.95; p�0.0081) at cycle 2; and role<br />

functioning (0.93, 0.88-1.00; p�0.0128) and age (0.98, 0.96-1.00;<br />

p�0.0081) at cycle 3) predicted survival; however, change in HRQoL was<br />

only an independent predictor for improvement at cycle 1. Conclusions: Our<br />

findings suggest that change from baseline over time in HRQoL, as<br />

measured on subscales <strong>of</strong> the EORTC QLQ-C30, contains added prognostic<br />

value for survival independent <strong>of</strong> baseline HRQoL scores. Further work is<br />

needed to assess the robustness and sensitivity <strong>of</strong> these findings.<br />

TPS7609 General Poster Session (Board #53C), Sat, 1:15 PM-5:15 PM<br />

Pilot trial <strong>of</strong> molecular pr<strong>of</strong>iling and targeted therapies in advanced thoracic<br />

malignancies: Non-small cell lung cancer, small cell lung cancer and<br />

thymic malignancies (CUSTOM). Presenting Author: Ariel Lopez-Chavez,<br />

Oregon Health & Science University, Portland, OR<br />

Background: For decades, clinical trial design strategies have relied upon<br />

the broad classification <strong>of</strong> tumors into tumor site and histopathology.<br />

Several lines <strong>of</strong> evidence have shown that a molecular approach to patient<br />

selection may be better in its capacity to improve patient outcomes.<br />

CUSTOM is an innovative clinical trial that allows for the parallel evaluation<br />

<strong>of</strong> multiple targeted therapies in molecularly selected patients with<br />

multiple cancer histological subtypes and the prospective evaluation <strong>of</strong><br />

multiple molecular biomarkers. Methods: All patients with advanced lung<br />

cancer and thymic malignancies and a good performance status are eligible<br />

independently <strong>of</strong> previous lines <strong>of</strong> treatment. The trial begins with tumor<br />

biopsy and molecular pr<strong>of</strong>iling using a multi-platform approach to identify<br />

oncogenic alterations in more than 34 genes for treatment allocation (12<br />

genes) and exploratory purposes. Depending upon the specific biomarker<br />

identified, patients are then triaged into 5 experimental arms. Erlotinib for<br />

sensitizing EGFR mutations; AZD6244 for KRAS, NRAS, HRAS and BRAF<br />

mutations; MK2206 for PIK3CA, AKT and PTEN mutations and PIK3CA<br />

amplification; Lapatinib for ERBB2 mutations or amplification; Sunitnib<br />

for KIT mutations and PDGFRA mutations and amplification. Patients not<br />

eligible for the experimental treatment arms are enrolled into a standard<br />

treatment arm. Upon disease progression patients are eligible for repeat<br />

biopsy and molecular pr<strong>of</strong>iling in order to identify molecular changes and<br />

mechanisms <strong>of</strong> resistance. All patients are followed until death. The<br />

primary endpoint is response rate and secondary endpoints include<br />

progression-free survival, duration <strong>of</strong> response and overall survival. With<br />

three disease types and five experimental drugs, there are 15 possible<br />

treatment arms which will be under active consideration. For each <strong>of</strong> these,<br />

the study will be conducted as an optimal two-stage phase II trial in order to<br />

rule out an unacceptably low 10% clinical response rate in favor <strong>of</strong> a<br />

modestly high response rate <strong>of</strong> 40%. As <strong>of</strong> January 11, 2012, two hundred<br />

and sixteen patients have been enrolled.<br />

7608 General Poster Session (Board #53B), Sat, 1:15 PM-5:15 PM<br />

Three-year survival in stage IV non-small cell lung cancer (NSCLC): The<br />

importance <strong>of</strong> metastatic distribution. Presenting Author: Daniel D. Karp,<br />

University <strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: Although long term survival (LTS) is an important goal <strong>of</strong> lung<br />

cancer treatment, those with metastatic disease at presentation have a<br />

median <strong>of</strong> only 8-9 months in most series with a small “tail” on the survival<br />

curve. We analyzed a large cohort <strong>of</strong> Stage IV pts by performance status<br />

(PS), smoking history, number and sites <strong>of</strong> disease to assess factors<br />

associated with 36 month survival. Methods: From 2004–2008, 526 newly<br />

diagnosed untreated pts with Stage IV NSCLC received initial treatment at<br />

our institution. Sites <strong>of</strong> disease were analyzed according to lung, brain,<br />

bone, liver, adrenal, skin, malignant effusion, lymphangitic, bulky pleural<br />

disease, and “other”. No patient had definitive surgery. Results: Overall,<br />

58/526 pts (10.8%) survived 36 months or more. 453 pts died within 36<br />

months. 15 pts alive with follow-up time less than 36 months were removed<br />

from further analysis. Of those, 38/58 (65.5%) had only 1 metastatic site<br />

(p�0.001). 14 (24.1%) had 2 sites, only 6 (10.3%) had 3 or more sites.<br />

Those 19 pts with lung to lung spread were the most common (32.8%) with<br />

LTS – reflecting the M1 status in the new International Staging System.<br />

Only 1 LTS pt had liver mets at diagnosis – a solitary lesion treated with<br />

radi<strong>of</strong>requency ablation. 20 pts had PS�0 (0.019) and 29 had PS�1<br />

(NS). PS�2 pts made up 6. 2 pts had PS�3 initially. 1 unknown. Only<br />

6/58 (10.3%) were current smokers, 23 (39.7%) were never-smokers<br />

(p�0.001). Adenocarcinoma made up 65.5%. Conclusions: Progress in<br />

lung cancer survival has been slow. Very few LTS pts have more than 1 site<br />

<strong>of</strong> metastatic disease at presentation, virtually none with liver disease<br />

(p�0.02). Number <strong>of</strong> metastatic sites and presence <strong>of</strong> liver metastases<br />

appear to be important stratification variables for lung cancer clinical trials.<br />

TPS7610 General Poster Session (Board #53D), Sat, 1:15 PM-5:15 PM<br />

TIME: A phase IIb/III randomized, double-blind, placebo-controlled study<br />

comparing first-line therapy with or without TG4010 immunotherapy<br />

product in patients with stage IV non-small cell lung cancer (NSCLC).<br />

Presenting Author: Elisabeth A. Quoix, Strasbourg University Hospital,<br />

Strasbourg, France<br />

Background: TG4010 is an immunotherapy product based on a poxvirus<br />

(MVA) coding for the MUC1 tumor-associated antigen and interleukin-2. A<br />

previous study, TG4010.09, which evaluated the combination <strong>of</strong> first-line<br />

chemotherapy with and without TG4010 in advanced NSCLC, achieved its<br />

primary endpoint based on 6-month progression-free survival (PFS) and<br />

showed that the pre-treatment level <strong>of</strong> activated Natural Killer (aNK) cells<br />

may be a potential predictive biomarker for TG4010 efficacy (E. Quoix et<br />

al., Lancet Oncol. 2011;12:1125-33). Methods: TIME is a double-blind<br />

phase IIb/III study comparing the combination <strong>of</strong> first-line therapy with<br />

TG4010 or placebo in stage IV NSCLC patients, Performance Status (PS) 0<br />

or 1 with a MUC1 expressing tumor by immunohistochemistry. The Phase<br />

IIb part <strong>of</strong> the study aims at prospectively validating aNK level as a<br />

predictive biomarker with PFS as a primary endpoint, by comparing the two<br />

treatment arms in two subgroups defined according to the level <strong>of</strong> aNK cells<br />

at baseline (normal or high). Bayesian criteria, derived from the TG4010.09<br />

study results, will be used to confirm that, with a large probability, the true<br />

hazard ratio is �1 in patients with normal level <strong>of</strong> aNK cells and �1 in<br />

patients with high level <strong>of</strong> aNK cells. The Phase III part <strong>of</strong> the study will<br />

then compare, by using a frequentist approach, the two treatment arms<br />

with overall survival as a primary endpoint in the patient population<br />

confirmed to be <strong>of</strong> interest in the Phase IIb part. The phase III part is<br />

powered to detect a 27% reduction in the hazard rate <strong>of</strong> death. Phase IIb<br />

and III parts <strong>of</strong> the study will enroll respectively 206 and 800 patients. A<br />

dynamic minimization procedure will be applied at randomization for<br />

histology, prescription <strong>of</strong> bevacizumab, type <strong>of</strong> chemotherapy, PS and<br />

center. If qualifying for, patients will receive maintenance therapy after<br />

chemotherapy according to labeling. The study TIME is open to recruitment<br />

and referenced in <strong>Clinical</strong>Trials.gov with the identifier NCT01383148.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


TPS7611 General Poster Session (Board #53E), Sat, 1:15 PM-5:15 PM<br />

CA184-104: Randomized, multicenter, double-blind, phase III trial comparing<br />

the efficacy <strong>of</strong> ipilimumab (Ipi) with paclitaxel/carboplatin (PC)<br />

versus placebo with PC in patients (pts) with stage IV/recurrent non-small<br />

cell lung cancer (NSCLC) <strong>of</strong> squamous histology. Presenting Author: Martin<br />

Reck, Hospital Großhansdorf, Großhansdorf, Germany<br />

Background: Years <strong>of</strong> research in advanced NSCLC have not improved<br />

outcomes for the squamous subtype beyond those <strong>of</strong> standard platinum<br />

doublets. Evidence <strong>of</strong> responses to immune therapies in NSCLC <strong>of</strong><br />

squamous cell histology supports investigation in this subtype. Ipi, a fully<br />

human monoclonal antibody which blocks CTLA-4, augments antitumor<br />

immune responses. Ipi improved overall survival (OS) in advanced melanoma,<br />

with side effects managed using product-specific treatment guidelines;<br />

immune-related response criteria (irRC) were derived from WHO<br />

criteria to better capture response patterns observed with Ipi. A randomized<br />

Phase 2 study <strong>of</strong> Ipi/PC in Stage IV NSCLC pts showed significant<br />

improvement in progression-free survival (PFS), as measured by mWHO or<br />

irRC, with a trend toward improved OS, over chemotherapy alone in pts<br />

receiving phased Ipi/PC (Ipi started after 2 cycles <strong>of</strong> PC). Phased Ipi/PC<br />

appeared to show efficacy in tumors <strong>of</strong> squamous histology. Addition <strong>of</strong> Ipi<br />

did not exacerbate PC toxicity, and immune-related adverse events were<br />

managed using protocol-specific guidelines. A Phase 3 trial (<strong>Clinical</strong>Trials-<br />

.gov identifier NCT01285609) is examining whether phased Ipi/PC will<br />

prolong OS in chemotherapy-naïve pts with squamous NSCLC. Methods:<br />

Stage IV/recurrent squamous NSCLC with ECOG 0-1 will be included; pts<br />

with CNS metastases or history <strong>of</strong> autoimmune disease will be excluded.<br />

Pts are randomized to receive 2 cycles <strong>of</strong> PC (175 mg/m2 and AUC�6,<br />

respectively; IV), followed by 4 cycles <strong>of</strong> study drug (Ipi in Arm A, placebo in<br />

Arm B; IV) with 4 additional cycles <strong>of</strong> PC (total 6 cycles). Pts without<br />

progressive disease (PD) after induction receive maintenance therapy with<br />

blinded study drug Q12W until PD per mWHO. The study will randomize<br />

920 pts 1:1 between arms. The primary endpoint <strong>of</strong> this study is OS;<br />

secondary endpoints include OS among pts who receive blinded therapy,<br />

PFS and best overall response rate.<br />

TPS7613^ General Poster Session (Board #53G), Sat, 1:15 PM-5:15 PM<br />

The GALAXY Trial (NCT01348126): A randomized IIB/III study <strong>of</strong><br />

ganetespib (STA-9090) in combination with docetaxel versus docetaxel<br />

alone in subjects with stage IIIB or IV NSCLC. Presenting Author: Glenwood<br />

D. Goss, The Ottawa Hospital Cancer Centre, Ottawa, ON, Canada<br />

Background: Hsp90 is a molecular chaperone required for proper folding<br />

and activation <strong>of</strong> many cancer-promoting proteins and is recognized as a<br />

key facilitator <strong>of</strong> cancer cell growth and survival. In pre-clinical models,<br />

Hsp90 inhibition causes degradation <strong>of</strong> multiple client proteins and leads<br />

to cancer cell death. Ganetespib is a resorcinolic Hsp90 inhibitor that has<br />

shown potent anti-tumor activity in patients with lung, breast, and other<br />

cancers that had progressed on standard treatment agents. Moreover,<br />

combination <strong>of</strong> ganetespib with docetaxel results in synergistic antiproliferative<br />

effects in several human non-small cell lung carcinoma (NSCLC)<br />

tumor xenografts. Ganetespib is well tolerated and is devoid <strong>of</strong> severe liver<br />

or common ocular toxicities that have been observed with some other<br />

Hsp90 inhibitors. Diarrhea is the most common adverse event and is<br />

manageable with appropriate supportive care. In a recent report, ganetespib<br />

administered at 200 mg/m2 weekly showed activity in pretreated patients<br />

with advanced NSCLC patients with ELM4-ALK translocation and KRAS<br />

mutations. Methods: Stage 1 (240 subjects): randomized, international<br />

open-label Phase 2B study in subjects that progressed on or after one prior<br />

systemic therapy for stage IIIB or IV NSCLC: patients are prospectively<br />

stratified for ECOG performance status, histology, total LDH, interval since<br />

diagnosis, and smoking status. Co-primary endpoints are PFS in the ITT<br />

population, and PFS in patients with KRAS mutations. Main secondary<br />

endpoints include ORR, disease control rate, OS and clinical activity in<br />

different molecular subtypes, including BRAF, HER2, EGFR, EML4-ALK.<br />

Patients on the control arm are treated with docetaxel 75 mg/m2 on day 1 <strong>of</strong><br />

a three-week cycle. In the combination arm, ganetespib 150 mg/m2 is<br />

given on day 1 (with docetaxel) and day 15 <strong>of</strong> a three-week cycle. At the<br />

time <strong>of</strong> submission 90 subjects had been enrolled in Stage 1.<br />

Lung Cancer—Non-small Cell Metastatic<br />

507s<br />

TPS7612 General Poster Session (Board #53F), Sat, 1:15 PM-5:15 PM<br />

AvaALL: Open-label randomized phase IIIb trial evaluating the efficacy and<br />

safety <strong>of</strong> standard <strong>of</strong> care with or without continuous bevacizumab (BV)<br />

treatment beyond disease progression in patients (pts) with advanced<br />

nonsquamous non-small cell lung cancer (NSCLC) after first-line (1L)<br />

treatment with BV plus platinum-doublet chemotherapy (CT). Presenting<br />

Author: Cesare Gridelli, SG Moscati Hospital, Avellino, Italy<br />

Background: Pts with NSCLC have a poor prognosis upon progression<br />

following 1L CT. Although the disease may have progressed on 1L CT, it is<br />

likely still dependent on VEGF-mediated pathways. Thus, persistent VEGF<br />

suppression along with second- and third-line cytotoxic regimens may yield<br />

clinical benefit. Analyses have associated this strategy with improved<br />

survival in NSCLC (Nadler et al, Oncologist 2011) and mCRC (Grothey et al<br />

JCO 2008). Methods: AvaALL (MO22097; NCT01351415), a multicenter<br />

(144 centers, 20 countries) open-label randomized (1:1) 2-arm phase IIIb<br />

study, recruits pts with advanced non-squamous NSCLC that has progressed<br />

after 1L treatment with BV plus a platinum-doublet and at least 2<br />

cycles <strong>of</strong> BV maintenance monotherapy. Pts in the experimental arm<br />

receive BV at the same dose from induction (7.5 or 15 mg/kg IV on D1 <strong>of</strong><br />

every 21-day cycle) continuously along subsequent lines, plus investigator’s<br />

choice <strong>of</strong> second-line agent (limited to pemetrexed, docetaxel, or<br />

erlotinib) and subsequent lines <strong>of</strong> treatment. Pts in the control arm receive<br />

investigator’s choice <strong>of</strong> agent alone in second and subsequent lines <strong>of</strong><br />

treatment, but no further BV treatment. Stratifications include nature <strong>of</strong><br />

second-line standard treatment, number <strong>of</strong> cycles <strong>of</strong> BV maintenance (�6<br />

vs. �6) and smoking status. The primary endpoint is overall survival (OS).<br />

Secondary endpoints include 6-, 12-, and 18-month OS rates, progressionfree<br />

survival, time to progression at second and third progressive disease<br />

(PD), response rate, disease control rate, duration <strong>of</strong> response at second<br />

and third PD, and safety <strong>of</strong> BV across multiple lines <strong>of</strong> treatment.<br />

Exploratory objectives include quality <strong>of</strong> life assessment through multiple<br />

treatment lines, and biomarker analysis. Assuming a median OS beyond PD<br />

<strong>of</strong> 7.9 months in the control group and 10.1 months in the treatment arm<br />

(HR 0.78), 528 events are required to achieve 80% power for the log-rank<br />

test at a 2-sided significance level <strong>of</strong> 5%. Enrollment began in June 2011.<br />

TPS7614 General Poster Session (Board #53H), Sat, 1:15 PM-5:15 PM<br />

ASPIRATION: Phase II study <strong>of</strong> continued erlotinib beyond RECIST<br />

progression in Asian patients (pts) with epidermal growth factor receptor<br />

(EGFR) mutation-positive non-small cell lung cancer (NSCLC). Presenting<br />

Author: Keunchil Park, Samsung Medical Center, Seoul, South Korea<br />

Background: First-line erlotinib (an EGFR tyrosine-kinase inhibitor) significantly<br />

increased progression-free survival (PFS) vs chemotherapy in phase<br />

III trials <strong>of</strong> pts with EGFR mutation-positive NSCLC. Discontinuation <strong>of</strong><br />

erlotinib on RECIST disease progression (PD) may lead to rapid disease<br />

flare-up; continued erlotinib beyond RECIST PD may extend clinical benefit<br />

by slowing progression <strong>of</strong> this life-threatening disease. We describe<br />

ASPIRATION, a large, Asian, multicenter, single-arm, open-label, phase II<br />

trial (NCT01310036), which will increase understanding <strong>of</strong> first-line<br />

erlotinib and erlotinib continuation beyond RECIST PD in pts with<br />

EGFR-mutated NSCLC. Methods: Pts (n�204) �18 yrs with stage IV/<br />

recurrent NSCLC, �1 measurable lesion (�10mm), ECOG performance<br />

status (PS) 0-2 and positive EGFR mutation status established by local<br />

pathology laboratory (that underwent voluntarily QA/QC) are eligible. All pts<br />

receive erlotinib 150mg/day. Tumor response is evaluated using RECIST<br />

(v1.1). The primary endpoint is PFS. At investigator’s discretion, pts may<br />

continue on erlotinib beyond RECIST PD, e.g. if they have slow PD (�6<br />

months <strong>of</strong> partial response/stable disease), asymptomatic minimal PD, or<br />

new brain metastasis controlled locally. Pts should not continue erlotinib if<br />

they have extracranial PD with symptoms; rapid PD and/or worsening <strong>of</strong> PS;<br />

or life-threatening complications. Pts continuing erlotinib who present with<br />

second RECIST PD will discontinue. Secondary endpoints include objective<br />

response rate, disease control rate, overall survival, and safety. For the<br />

exploratory biomarker study, pre-treatment tumor tissue blocks are collected;<br />

remaining tissue (after EGFR mutation testing for eligibility) will be<br />

analyzed centrally to study the association <strong>of</strong> biomarkers and clinical<br />

outcomes. Pre-treatment and post-treatment plasma and serum samples<br />

will be obtained at various time points for biomarker assays, including<br />

EGFR mutations and other candidate NSCLC biomarkers. Recruitment<br />

began in Apr 2011; the estimated final data collection for the primary<br />

endpoint is Dec 2014.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


508s Lung Cancer—Non-small Cell Metastatic<br />

TPS7615 General Poster Session (Board #54A), Sat, 1:15 PM-5:15 PM<br />

ARCHER: Dacomitinib (D; PF-00299804) versus erlotinib (E) for advanced<br />

(adv) non-small cell lung cancer (NSCLC)—A randomized double-blind<br />

phase III study. Presenting Author: Michael Boyer, Sydney Cancer Centre,<br />

Camperdown, Australia<br />

Background: D is a highly selective irreversible small molecule inhibitor <strong>of</strong><br />

all catalytically active members <strong>of</strong> the HER (human epidermal growth<br />

factor receptor) family <strong>of</strong> tyrosine kinases. In a randomized phase II trial in<br />

patients (pts) who had received 1–2 prior systemic therapy regimens for adv<br />

NSCLC, D demonstrated significantly longer progression-free survival (PFS)<br />

vs. E in the overall population (12.4 vs. 8.3 weeks; HR�0.66, P�0.012),<br />

with benefit consistent across several clinical and molecular subgroups.<br />

Median PFS in the KRAS wild-type (WT) subgroup was 16.1 vs. 8.3 weeks<br />

for D and E, respectively (HR�0.55, P�0.006). Methods: Based on phase<br />

II data, a randomized, double-blinded phase III clinical trial (ARCHER;<br />

NCT01360554) was designed to compare the efficacy <strong>of</strong> D with E in two<br />

co-primary populations <strong>of</strong> pts with adv NSCLC: (a) all enrolled pts with adv<br />

NSCLC, and (b) pts with KRAS WT NSCLC. Pts with locally adv/metastatic<br />

pathologically confirmed NSCLC, radiologically measurable disease, 1 or 2<br />

prior chemotherapy regimens, ECOG PS 0–2, and tissue available for<br />

molecular analysis will be randomized to receive D 45 mg or E 150 mg<br />

orally once daily. As <strong>of</strong> Jan 31, 2012, 117 <strong>of</strong> a planned 800 pts have been<br />

enrolled. The primary endpoint is PFS. Secondary endpoints include overall<br />

survival, objective response rate, duration <strong>of</strong> response, safety and tolerability,<br />

and pt-reported outcomes <strong>of</strong> health-related quality <strong>of</strong> life and disease-/<br />

treatment-related symptoms. Study design provides 90% and 80% power<br />

to detect �33% and �45% improvement in PFS in all pts receiving D vs.<br />

E, and in pts with KRAS W T NSCLC, respectively, and HR �0.75 and<br />

�0.69 using a 1-sided stratified log-rank test at a significance level <strong>of</strong><br />

0.015 and 0.01, respectively. The final primary analysis stratified log-rank<br />

test will include ECOG PS, KRAS mutation status and EGFR mutation<br />

status as stratification factors. The sample sizes above will also allow the<br />

assessment <strong>of</strong> OS in the co-primary populations with adequate power.<br />

Post-hoc analyses will be performed to explore EGFR, HER family, and<br />

KRAS mutation status, as well as other tumor-derived biomarkers collected<br />

from all pts in this trial.<br />

TPS7617 General Poster Session (Board #54C), Sat, 1:15 PM-5:15 PM<br />

Prophylactic cranial irradiation after reaching complete response, partial<br />

response, or stable disease in non-small cell lung cancer patients treated<br />

with epidermal growth factor receptor tyrosine kinase inhibitors (ProACT).<br />

Presenting Author: Amanda Tufman, Ludwig Maximilians University Munich,<br />

Munich, Germany<br />

Background: Patients with non-small cell lung cancer (NSCLC) who respond<br />

to treatment with epidermal growth factor receptor tyrosine kinase inhibitors<br />

(EGFR TKIs) seem to be at increased risk <strong>of</strong> central nervous system<br />

relapse, and may benefit from prophylactic cranial irradiation (PCI) more<br />

than other NSCLC patients. Methods: This study investigates the safety and<br />

efficacy <strong>of</strong> combining PCI with EGFR TKIs in stage IV NSCLC. Patients with<br />

stage IV NSCLC, no evidence <strong>of</strong> brain metastases, and an indication for first<br />

or later line therapy with an EGFR TKI will be enrolled. Those with complete<br />

response (CR), partial response (PR), or stable disease (SD) following 6<br />

weeks <strong>of</strong> therapy and no evidence <strong>of</strong> brain metastases on MRI will be<br />

treated with PCI. Neurocognitive function, depression indices, quality <strong>of</strong><br />

life, symptoms, and ability to function independantly will be assessed at<br />

baseline, before PCI, and at 6 week and then 3 month intervals following<br />

PCI. MRI will be repeated 6 weeks following PCI and at 3 month intervals,<br />

and serum markers <strong>of</strong> blood brain barrier permeability (neuron-specific<br />

enolase (NSE), protein S100 beta) will be assessed at all visits. Safety data<br />

will be formally reviewed after the first 10 patients. The primary endpoint<br />

for efficacy is overall survival. Secondary endpoints include progression<br />

free survival, site <strong>of</strong> progression, quality <strong>of</strong> life and neurological disability.<br />

Planed subgroup analyses based on mutation status, line <strong>of</strong> treatment with<br />

TKIs, comorbidity, precise histology and molecular biology will be carried<br />

out.<br />

TPS7616 General Poster Session (Board #54B), Sat, 1:15 PM-5:15 PM<br />

The MetLUNG study: A randomized, double-blind, phase III study <strong>of</strong><br />

onartuzumab (MetMAb) plus erlotinib versus placebo plus erlotinib in<br />

patients with advanced, MET-positive non-small cell lung cancer (NSCLC).<br />

Presenting Author: David R. Spigel, Sarah Cannon Research Institute/<br />

Tennessee Oncology PLLC, Nashville, TN<br />

Background: Increased Met signaling is associated with poor prognosis in<br />

NSCLC and can result in acquired resistance to epidermal growth factor<br />

receptor (EGFR)-targeted drugs in EGFR-mutant lung tumors. Onartuzumab<br />

(MetMAb) is a humanized monovalent monoclonal antibody that<br />

binds to Met with high specificity, preventing HGF ligand binding and<br />

blocking downstream signaling. Onartuzumab has shown anticancer activity<br />

in preclinical studies (Merchant et al. AACR 2008:Abstr. 1336) and in a<br />

phase I study <strong>of</strong> patients with advanced solid tumors (Moss et al. Ann Oncol<br />

2010;21(Suppl. 8):Abstr. 504P). In a phase II study, patients with<br />

Met-positive tumors (�50% <strong>of</strong> tumor cells stain 2� or 3� intensity by IHC)<br />

(Met Dx�) who received onartuzumab � erlotinib had nearly tw<strong>of</strong>old<br />

reduction in the risk <strong>of</strong> disease progression and threefold reduction in the<br />

risk <strong>of</strong> death compared with erlotinib alone (Spigel et al. J Clin Oncol<br />

2011;29(Suppl.):Abstr. 7505). An increased incidence <strong>of</strong> peripheral<br />

edema was observed in patients treated with onartuzumab. Methods: This is<br />

a randomized, phase III, multicenter, double-blind, placebo-controlled<br />

study. Adults with Met Dx� tumors who failed at least 1 but no more than 2<br />

prior lines <strong>of</strong> platinum-based chemotherapy for advanced NSCLC are<br />

eligible. Around 480 patients will receive (1:1) erlotinib (150 mg PO daily)<br />

� placebo or onartuzumab (15 mg/kg IV Q3W) until disease progression,<br />

unacceptable toxicity, patient/physician decision to discontinue, or death.<br />

Patients are stratified for Met expression (2� vs 3�), prior lines <strong>of</strong> therapy<br />

(1 vs 2), histology (squamous vs nonsquamous) and EGFR-activating<br />

mutation status (yes vs no). The primary endpoint is OS. Secondary<br />

endpoints include PFS, response rates, safety, patient-reported outcomes,<br />

and PK. An IDMC will monitor safety every 6 months after the 1st patient is<br />

enrolled and evaluate 1 interim analysis when ~67% <strong>of</strong> the total OS events<br />

have occurred. This study is open for accrual; further details can be found<br />

on <strong>Clinical</strong>Trials.gov (NCT01456325).<br />

TPS7618 General Poster Session (Board #54D), Sat, 1:15 PM-5:15 PM<br />

Weekly nab-paclitaxel in combination with carboplatin as first-line therapy<br />

in patients (pts) with advanced non-small cell lung cancer (NSCLC):<br />

Analysis <strong>of</strong> patient-reported neuropathy and taxane-associated symptoms.<br />

Presenting Author: Vera Hirsh, McGill University – Royal Victoria Hospital,<br />

Montreal, QC, Canada<br />

Background: Neuropathy and its associated symptoms are dose-limiting<br />

toxicities <strong>of</strong> taxane-based regimens. Measuring disease symptoms has<br />

utility for maintaining a balance between the benefits and costs <strong>of</strong><br />

treatment. In a phase III trial nab-paclitaxel (nab-P, 130 nm albuminbound<br />

paclitaxel particles) � carboplatin (C) vs solvent-based paclitaxel<br />

(sb-P) � C significantly improved the primary endpoint <strong>of</strong> overall response<br />

rate (ORR) (25% vs 33%, P � 0.005), with a 1-month improvement in<br />

median overall survival (OS; P � NS), and an improved tolerability pr<strong>of</strong>ile in<br />

pts with advanced NSCLC. Here we report on patient-assessed neuropathy<br />

and taxane-associated symptoms, important factors in a palliative patient<br />

setting. Methods: Pts with untreated stage IIIB/IV NSCLC were randomized<br />

1:1 to C AUC 6 day 1 and either nab-P 100 mg/m2 on days 1, 8, 15 (n �<br />

521) or sb-P 200 mg/m2 day1(n� 531) q 21 days. Treatment was<br />

continued until disease progression. Safety was assessed per the Common<br />

Terminology Criteria for Adverse Events (CTCAE) v3.0. The mean change<br />

from baseline to day 1 <strong>of</strong> each cycle for the neuropathy, pain, and hearing<br />

subscales <strong>of</strong> Functional Assessment <strong>of</strong> Cancer Therapy (FACT)-Taxane v<br />

4.0 were assessed. Results: 1031 (98%) pts completed FACT-Taxane at<br />

baseline, and 987 (94%) during follow-up or at completion <strong>of</strong> treatment.<br />

Baseline scores for neuropathy and pain were well balanced. Significant<br />

treatment effects favoring nab-P/C were noted for patient-reported neuropathy<br />

(P � 0.001), neuropathic pain hands/feet (P � 0.001), and hearing<br />

loss (P � 0.002). Physician-assessed rates <strong>of</strong> neuropathy were significantly<br />

lower in the nab-P/C arm vs the sb-P/C arm: 47% vs 63%, P � 0.001, all<br />

grades; 3% vs 12%, P � 0.001, grade 3/4, respectively. More pts treated<br />

with nab-P/C vs sb-P/C had no neuropathy (53% vs 47%, P � 0.001).<br />

Conclusions: nab-P/C was associated with statistically and clinically<br />

significant reductions in patient-reported neuropathy, neuropathic pain in<br />

the hands and feet, and hearing loss compared with sb-P/C. Patientreported<br />

outcomes for neuropathy were consistent with physician assessment.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


TPS7619 General Poster Session (Board #54E), Sat, 1:15 PM-5:15 PM<br />

A multicenter randomized phase III trial <strong>of</strong> customized chemotherapy<br />

versus standard <strong>of</strong> care for first-line treatment <strong>of</strong> elderly patients with<br />

advanced non-small cell lung cancer (EPIC). Presenting Author: George R.<br />

Simon, Hollings Cancer Center, Medical University <strong>of</strong> South Carolina,<br />

Charleston, SC<br />

Background: Personalizing therapy based on an individual patient’s molecular<br />

pr<strong>of</strong>ile is a potentially promising approach to optimize efficacy with the<br />

available agents. Optimizing efficacy in the elderly is particularly relevant<br />

owing to their increased propensity to suffer therapy-induced toxicity. In<br />

this proposal we will attempt to demonstrate optimization <strong>of</strong> therapy in<br />

good performance EGFR mutation negative elderly patients by taking in to<br />

consideration the histology <strong>of</strong> the tumor, the ERCC1 (marker <strong>of</strong> platinum<br />

resistance), RRM1 (for gemcitabine resistance) and TS (for pemetrexed<br />

resistance) status. Methods: Untreated advanced stage NSCLC patients age<br />

�70 years with measurable disease will be enrolled. Patients will be<br />

randomized in a 2:1 randomization to experimental arm (A) or standard arm<br />

(B). In arm A, treatment with single or dual-agent chemotherapy will be<br />

selected based on histology, ERCC1 (E), RRM1(R) and TS (T) expression at<br />

the RNA level. The cut <strong>of</strong>f for high (�, therefore resistant) or low (-,<br />

therefore sensitive) expression have been previously defined. Patients with<br />

squamousNSCLC who are E-R� will be treated with single agent carboplatin,<br />

E�R- with gemcitabine, E-R- with carboplatin and gemcitabine and<br />

E�R� with docetaxel or vinorelbine. In non-squamous NSCLC patients<br />

E-T� will be treated with carboplatin, E�T- withpemetrexed, E-T- with<br />

carboplatin and pemetrexed, E�T�R- with gemcitabine and E�T�R�<br />

with docetaxel or vinorelbine. In arm B treatment with single or dual-agent<br />

chemotherapy will be at the discretion <strong>of</strong> the care provider, i.e., standard <strong>of</strong><br />

care. The primary endpoint <strong>of</strong> the trial is overall survival (OS). The<br />

secondary endpoint is progression-free survival (PFS). The tertiary endpoint<br />

is disease response. Treatment will continue to maximum <strong>of</strong> 6 cycles if<br />

tolerated or until disease progression. A sample size <strong>of</strong> 453 patients will<br />

give us 90% power to detect a three-month improvement in median survival<br />

(8 to 11 months; corresponding to a HR <strong>of</strong> 0.73) with the log rank test at a<br />

significance level <strong>of</strong> 5%(1-sided) allowing for a 10% failure rate in gene<br />

analyses.<br />

Lung Cancer—Non-small Cell Metastatic<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.<br />

509s


510s Lymphoma and Plasma Cell Disorders<br />

8000 Oral Abstract Session, Sat, 3:00 PM-6:00 PM<br />

CALGB 50401: A randomized trial <strong>of</strong> lenalidomide alone versus lenalidomide<br />

plus rituximab in patients with recurrent follicular lymphoma.<br />

Presenting Author: John Leonard, Weill Cornell Medical College, New York,<br />

NY<br />

Background: Lenalidomide (L) and rituximab (R) are active as single agents<br />

in follicular (FL) and other B-cell lymphomas, although combination<br />

strategies have not been previously assessed in a randomized fashion.<br />

Methods: CALGB 50401 is a randomized phase II study, initially designed<br />

to evaluate 3 regimens: R alone (375 mg/m2 weekly x 4), L alone (15 mg<br />

cycle 1, then escalated to 20 mg cycles 2-12, administered days 1-21 q 28<br />

days x 12 cycles) or the combination <strong>of</strong> L� R (other 2 arms combined). The<br />

R alone arm was discontinued due to slow accrual with 3 enrolled subjects.<br />

Eligibility included recurrent FL, prior therapy with rituximab alone or in<br />

combination, and TTP <strong>of</strong> � 6 months from last rituximab dose. Prophylactic<br />

ASA or LMW heparin was recommended for patients at high risk for<br />

thrombosis. Results: Of 94 pts registered to L or LR, 89 (45 L and 44 LR)<br />

received at least one dose and had adequate data for analysis. Baseline<br />

characteristics include median age 63 (range 34-85) and 60% with<br />

intermediate- or high-risk FLIPI. Grade 3-4 adverse events (AE) were most<br />

commonly neutropenia (16% L,19% LR), fatigue (9% L, 14% LR) and<br />

thrombosis (16% - 7 pts L, 4% - 2 pts LR, p�0.158), and overall were seen<br />

in 49% (L) and 52% (LR) with 9% grade 4 in each arm. The full regimen<br />

was completed in 33% (L) and 59% (LR) <strong>of</strong> patients, with the difference<br />

due to more progressions or non-responders in the L group. In both arms<br />

about 19% <strong>of</strong> subjects discontinued therapy early due to AEs and dose<br />

intensity was over 80%. Objective response rates are L - 49% (13% CR)<br />

and LR - 75% (32% CR). With a median follow-up <strong>of</strong> 1.5 years (range 0.1-<br />

3.6 years), median EFS is 1.2 years (L) and 2.0 years (LR), p�0.0063,<br />

log-rank test. Conclusions: Lenalidomide � rituximab is more active than<br />

lenalidomide alone in patients with recurrent FL with similar toxicity. A<br />

trend toward lower thrombosis risk with LR may relate to greater anti-tumor<br />

efficacy. The LR regimen warrants further study in FL including as a<br />

backbone for addition <strong>of</strong> novel agents in relapsed and frontline settings.<br />

8002 Oral Abstract Session, Sat, 3:00 PM-6:00 PM<br />

ABVD (8 cycles) versus BEACOPP (4 escalated cycles �> 4 baseline) in<br />

stage III-IV high-risk Hodgkin lymphoma (HL): First results <strong>of</strong> EORTC<br />

20012 Intergroup randomized phase III clinical trial. Presenting Author:<br />

Patrice P. Carde, Institute Gustave Roussy, Villejuif, France<br />

Background: Escalated BEACOPP and derivatives achieved superior time to<br />

treatment failure (FFTF) over COPP/ABVD, resulting in higher overall<br />

survival (OS) for advanced HL. However, later clinical trials have failed to<br />

confirm OS superiority over ABVD. Methods: Eligibility criteria: clinical<br />

stage III/IV HL, International prognostic score (IPS) � 3, age�60. We<br />

compared ABVD (8 cycles) vs. BEACOPP (escalated 4 cycles � baseline 4),<br />

without irradiation. Randomization was stratified for institution and IPS.<br />

Primary endpoint was EFS, defined as treatment discontinuation, no<br />

complete response (CR) after 8 cycles, progression, relapse or death.<br />

Additional endpoints were CR, progression free survival (PFS), OS, quality<br />

<strong>of</strong> life and secondary malignancies. Outcomes were reviewed by study<br />

coordinators to ensure consistency across pts. Results: From 2002-2010,<br />

549 pts were randomized (ABVD 275, BEACOPP 274): stage IV 74%, PS<br />

0, 1, 2: 34, 48 and 17%, B-symptoms 81%, median age 35.2y, males<br />

75%. IPS was 4 or higher for 59% <strong>of</strong> pts. Histology reviewed no HL in 4<br />

cases. CR was 83% in both arms. With a median follow-up <strong>of</strong> 3.8 yrs, EFS<br />

at 4 yrs was 63.7% vs. 69.3% (HR � 0.86, 95%CI�0.64 to 1.15,<br />

p�0.312). PFS at 4 yrs was 72.8% vs. 83.4% (HR � 0.58, 95%CI�0.39<br />

to 0.85, p�0.005). OS at 4 yrs was 86.7 vs. 90.3 (HR � 0.71,<br />

95%CI�0.42 to 1.21, p�0.208). Toxic deaths occurred in 6 and 5 pts,<br />

with early discontinuation (prior to cycle 5) in 12 & 26 pts, respectively.<br />

There were 5 crossovers to BEACOPP and 10 to ABVD. Second malignancies<br />

occurred in 8 ABVD and 10 BEACOPP pts (myelodysplasia/leukemia 2<br />

and 4, lung 2 and 1, NHL 3 and 2, other 1 and 3); cumulative incidence<br />

curves did not differ significantly. Conclusions: The primary endpoint (EFS)<br />

was similar between treatment arms. However, more progressions/relapses<br />

were observed with ABVD, while early discontinuations were more frequent<br />

with BEACOPP. Nevertheless, even in this high-risk group, OS was not<br />

improved with BEACOPP. Additional considerations (treatment burden and<br />

cost, fertility issues, long term relapses and immediate and late morbidity)<br />

may guide physician/patient decisions toward ABVD or BEACOPP.<br />

8001 Oral Abstract Session, Sat, 3:00 PM-6:00 PM<br />

A phase III randomized intergroup trial (S0016) comparing CHOP plus<br />

rituximab with CHOP plus iodine-131-tositumomab for front-line treatment<br />

<strong>of</strong> follicular lymphoma: Results <strong>of</strong> subset analyses and a comparison<br />

<strong>of</strong> prognostic models. Presenting Author: Oliver W. Press, Fred Hutchinson<br />

Cancer Research Center, Seattle, WA<br />

Background: Advanced follicular lymphomas (FL) are considered incurable<br />

with chemotherapy and there is no consensus on the best treatment.<br />

Outcomes are variable, but can be partially predicted by defined prognostic<br />

factors. SWOG and CALGB compared the safety and efficacy <strong>of</strong> 2<br />

immunochemotherapy regimens in a Phase III trial enrolling 554 patients<br />

between 3/1/2001 and 9/15/2008. Methods: Patients were eligible if they<br />

had bulky stage II, III or IV FL and had not received prior therapy. Patients<br />

randomized to CHOP-R received 6 cycles <strong>of</strong> CHOP every 21 days � 6 doses<br />

<strong>of</strong> rituximab. Patients randomized to CHOP-RIT received 6 cycles <strong>of</strong> CHOP,<br />

followed by consolidative radioimmunotherapy with tositumomab/iodine<br />

I-131 tositumomab. A Cox proportional hazards multi-variable regression<br />

analysis assessed the prognostic impact <strong>of</strong> age, stage, LDH, LN size and<br />

number, performance status, hemoglobin, �2 microglobulin, BM involvement,<br />

and B symptoms. The prognostic value <strong>of</strong> 3 multi-variable models<br />

were compared. Results: Outcomes were outstanding with either CHOP-R or<br />

CHOP-RIT (2 yr PFS: 76% vs 80% [ p �0.11]; 2 yr OS: 97% vs 93% [p<br />

�0.08], respectively). Subset analyses so far have not identified any<br />

subgroups clearly benefitting to a greater degree from CHOP-R or CHOP-<br />

RIT in terms <strong>of</strong> both PFS and OS. Cox multivariable regression analysis<br />

identified serum-�2M, LDH level, and FLIPI index as the strongest<br />

prognostic factors associated with worse PFS and OS. Conclusions: Both<br />

regimens produced outstanding PFS and OS, and no statistically significant<br />

differences between them were observed. FLIPI, FLIPI2, and LDH � �2M<br />

models were all strong predictors <strong>of</strong> patient outcomes. A combination <strong>of</strong><br />

LDH � �2M was as good as the FLIPI index, and was simpler to apply.<br />

(Supported in part by NCI grants CA32102 and CA38926 from the NCI and<br />

GlaxoSmithKline.)<br />

PFS OS<br />

Parameter<br />

HR (95% Cl) p HR (95% Cl) p<br />

LDH 1.59 (1.17-2.17) .003 2.46 (1.49-4.07) .0004<br />

Serum-�2M 1.70 (1.27-2.28) .0004 2.22 (1.31-3.76) .003<br />

�2M and LDH 2.25 (1.51-3.31) �.0001 3.96 (2.02-7.78) �.0001<br />

FLIPI 2.28 (1.54-3.35) �.0001 3.65 (1.82-7.18) .0002<br />

8003 Oral Abstract Session, Sat, 3:00 PM-6:00 PM<br />

Plasma viral DNA as a marker <strong>of</strong> tumor response in EBV(�) Hodgkin<br />

lymphoma in a phase III study (E2496). Presenting Author: Jennifer Ann<br />

Kanakry, Johns Hopkins School <strong>of</strong> Medicine, Baltimore, MD<br />

Background: Epstein-Barr virus (EBV) is associated with Hodgkin lymphoma<br />

(HL) and can be detected by in situ hybridization (ISH) <strong>of</strong> viral nucleic acid<br />

(EBER) in tumor cells. Studies have suggested a correlation in HL between<br />

plasma EBV DNA and EBER ISH. We previously studied the DNase<br />

sensitivity <strong>of</strong> plasma EBV DNA and found plasma EBV <strong>of</strong> patients with<br />

EBV(�) HL was not protected from DNase digestion, consistent with<br />

tumor-derived DNA, while plasma EBV <strong>of</strong> patients with HIV without EBV(�)<br />

tumors was protected from DNase digestion, consistent with virion DNA.<br />

We sought to determine whether plasma EBV could serve as a surrogate for<br />

EBER ISH and whether reappearance <strong>of</strong> plasma EBV predicts treatment<br />

failure. Methods: Specimens from a Cancer Cooperative Intergroup Trial<br />

(E2496/Stanford V versus ABVD for HL) were used to compare pretreatment<br />

plasma EBV DNA copy number, assessed by real-time quantitative<br />

PCR, with EBV status by EBER ISH. An ROC analysis was performed using<br />

patients with both pretreatment plasma EBV and EBER results (n�121),<br />

identifying a cut<strong>of</strong>f <strong>of</strong> 60 viral copies/100 �L plasma (95% concordance,<br />

92% sensitivity, 96% specificity for EBV status by EBER). Using this<br />

cut<strong>of</strong>f, pretreatment plasma specimens (n�274) were designated EBV(�)<br />

(n�54) or EBV(-) (n�220), as were serial follow-up specimens. Cox<br />

proportional hazard models were constructed to evaluate plasma EBV as a<br />

prognostic factor for failure-free survival (FFS). FFS was estimated by the<br />

Kaplan-Meier method. Results: Pretreatment EBV(�) plasma was associated<br />

with treatment failure with a hazard ratio <strong>of</strong> 2.1 (95% CI 1.2-3.6,<br />

p�0.01) after adjusting for International Prognostic Score, treatment arm,<br />

and histology. Of the EBV(�) patients with follow-up specimens (n�45),<br />

patients with EBV(�) plasma beyond 1 month <strong>of</strong> therapy (n�9) had inferior<br />

FFS compared to those who cleared their plasma <strong>of</strong> EBV (n�36), (3-year<br />

FFS 44% versus 69%, respectively; log rank p�0.03). Conclusions: HL<br />

patients with EBV(�) plasma at baseline have inferior FFS compared to<br />

others. Among patients with EBV(�) plasma at baseline, those in whom<br />

plasma EBV persists or reappears after initiation <strong>of</strong> therapy have inferior<br />

FFS. Such patients may benefit from experimental or intensified therapies.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


8004 Oral Abstract Session, Sat, 3:00 PM-6:00 PM<br />

High-dose chemotherapy followed by allogeneic stem cell transplantation<br />

in high-risk relapsed and refractory aggressive non-Hodgkin lymphoma:<br />

Results <strong>of</strong> a prospective study <strong>of</strong> the German high-grade non-Hodgkin<br />

lymphoma study group. Presenting Author: Bertram Glass, Asklepios Klinik<br />

St. Georg, Hamburg, Germany<br />

Background: The role <strong>of</strong> allogeneic stem cell transplantation (alloSCT) in<br />

high-risk aggressive NHL is poorly defined; the role <strong>of</strong> graft-versuslymphoma<br />

effect is unclear. Reduced intensity conditioning has shown<br />

limited efficacy in patients with refractory disease. Methods: Patients with<br />

primary refractory disease, early relapse (�12 months) or relapse after<br />

autologous SCT <strong>of</strong> aggressive B-cell (n�61) or T-cell (n�23) NHL were<br />

enrolled from June 2004 to March 2009. Myeloablative conditioning<br />

(fludarabine 125mg/m2 , busulfan 12mg/kg, cyclophosphamide 120 mg/<br />

kg) was followed by alloSCT from related (n�24) or unrelated (n�60)<br />

donors. 57/84 patients received a 10 HLA-loci compatible graft. Results:<br />

Overall survival at 3 years was 42% (95% CI, 31% to 52%), progressionfree<br />

survival (PFS) was 40% (95% CI 29% to 50%). Non-relapse mortality<br />

(NRM) was 12% at 100 days and 35% at one year. Graft-versus-host<br />

disease (GVHD) and infection were the predominant causes <strong>of</strong> NRM.<br />

Relapse rate was 30% with latest relapse at day �327. Patients with an<br />

HLA fully compatible donor (plus ATG with unrelated donors) (n�40) had<br />

the best outcome (NRM at 1y 10.4 vs 57.2%, PFS at 3y 64.7% vs 31.8%,<br />

p � 0.0001). GVHD � grade I correlated with improved PFS (HR 0.45,<br />

p�0.0088). Patients with refractory disease or early relapse (n�60)<br />

experienced PFS at 3y <strong>of</strong> 33%. Conclusions: Lymphoma-debulking (highdose)<br />

chemotherapy followed by alloSCT shows excellent results in heavily<br />

pretreated patients with early relapse or primary refractory aggressive<br />

lymphoma. There was evidence <strong>of</strong> graft-versus-lymphoma activity in this<br />

setting. For patients with a fully matched (10/10) related or unrelated<br />

donor the results compare favorably to autoSCT or alloSCT following<br />

reduced-intensity conditioning.<br />

8006 Oral Abstract Session, Sat, 3:00 PM-6:00 PM<br />

R-CVP versus R-CHOP versus R-FM as first-line therapy for advanced-stage<br />

follicular lymphoma: Final results <strong>of</strong> FOLL05 trial from the Fondazione<br />

Italiana Linfomi (FIL). Presenting Author: Massimo Federico, Department<br />

<strong>of</strong> Oncology and Haematology, University <strong>of</strong> Modena and Reggio Emilia,<br />

Modena, Italy<br />

Background: The optimal chemotherapy regimen for patients with advanced,<br />

active follicular lymphoma (FL) has not been established yet. We<br />

conducted a randomized trial comparing R-CVP with R-CHOP and R-FM.<br />

Methods: Previously untreated patients with advanced FL were randomly<br />

assigned to receive 8 doses <strong>of</strong> rituximab associated to 8 cycles <strong>of</strong> CVP, or 6<br />

cycles <strong>of</strong> CHOP or FM (fludarabine 25 mg/m2 day 1-3, mitoxantrone 10<br />

mg/m2 day 1). No maintenance therapy was allowed. The principal study<br />

end point was Time to Treatment Failure (TTF). Events in TTF were failure<br />

<strong>of</strong> induction therapy, progressive or relapse disease and death from any<br />

causes. In order to show a hazard ratio between each experimental arms<br />

and standard arm <strong>of</strong> 0.53 we planned to accrue 534 patients (178 per arm)<br />

with 4 years <strong>of</strong> accrual and 1 year <strong>of</strong> follow-up. Statistical tests were<br />

two-sided with an alpha error <strong>of</strong> 0.05, adjusted by Bonferroni for multiple<br />

arm comparison, and power <strong>of</strong> 90%. Results: Between March 2006 and<br />

September 2010, 534 patients were enrolled; 30 were subsequently<br />

excluded due to violation <strong>of</strong> inclusion criteria. Median patient age was 56<br />

years (range 30-75), 63% <strong>of</strong> patients had stage IV disease, 37% had 3-5<br />

FLIPI and 27% 3-5 FLIPI2 scores. At the end <strong>of</strong> induction treatment the<br />

overall response rate (CR� PR) for the whole group was 91% (p�0.247).<br />

After a median follow-up <strong>of</strong> 34 months 208 events for TTF were recorded;<br />

3-year TTF was 46%, 64% and 61% for patients treated with R-CVP,<br />

R-CHOP and R-FM respectively (R-CHOP vs R-CVP p�0.007; R-FM vs<br />

R-CVP p�0.021; R-FM vs R-CHOP p�0.969). The 3-year overall survival<br />

rate (OS) was 98%, 95% and 93% for R-CVP, R-CHOP and R-FM group,<br />

respectively (P�NS). Patients treated with R-FM had a higher rate <strong>of</strong> g<br />

III-IV neutropenia (64% vs 28% R-CVP, p�0.001; and vs 50% R-CHOP,<br />

p�0.015). During follow-up second malignancies were registered as late<br />

events in 23 patients (2%, 3% and 8% in R-CVP, R-CHOP and R-FM,<br />

respectively). Conclusions: This trial showed that R-CVP was associated<br />

with an inferior 3-year TTF and PFS compared with R-FM and R-CHOP. OS<br />

was similar among study arms but R-FM showed a higher rate <strong>of</strong> secondary<br />

tumors.<br />

Lymphoma and Plasma Cell Disorders<br />

511s<br />

8005 Oral Abstract Session, Sat, 3:00 PM-6:00 PM<br />

A pooled analysis <strong>of</strong> 1,144 patients with HIV-associated lymphoma:<br />

Assessment <strong>of</strong> lymphoma-, HIV-, and treatment-specific factors on clinical<br />

outcomes. Presenting Author: Stefan K. Barta, Albert Einstein College <strong>of</strong><br />

Medicine, Bronx, NY<br />

Background: Non-Hodgkin Lymphoma (NHL) remains the most common<br />

malignancy in patients with HIV. Outcomes have significantly improved<br />

over the last decade, but there is no accepted consensus regarding the<br />

optimal initial therapeutic approach. Our objective was to assess the<br />

effects <strong>of</strong> clinical factors on response and survival. Methods: We performed<br />

a systematic review to search for prospective clinical phase II or III trials<br />

that assessed therapeutic interventions for HIV-associated NHL and<br />

assembled individual patient data from 16 trials published between 2000<br />

and 2011 including 1144 patients (median N�62/trial, range 17-195).<br />

Treatment factors included type <strong>of</strong> chemotherapy (CHOP, N�642;<br />

EPOCH, N�178; CDE, N�191; intensive regimens, N�155) and rituximab<br />

use (N�542). Endpoints included complete response (CR), progression-free<br />

survival (PFS), and overall survival (OS). We used logistic<br />

regression and Cox proportional hazard models adjusted for age, sex,<br />

age-adjusted International Prognostic Index (IPI), baseline CD4 count,<br />

baseline HIV viral load, use <strong>of</strong> concurrent antiretroviral therapy, and<br />

histology. Odds ratios (OR) � 1 for CR denote improved CR, and hazard<br />

ratios (HR) � 1 indicate reduced risk <strong>of</strong> progression or death. Results:<br />

Among the lymphoma- and HIV-specific covariates evaluated, only a higher<br />

IPI score was associated with inferior CR rate, PFS and OS (p�0.001).<br />

Rituximab was associated with a higher CR rate (OR 1.75; p�0.017),<br />

better PFS (HR 0.39, p�0.001) and OS (HR 0.39, p�0.001); patients<br />

with a higher baseline CD4 count benefited more from rituximab (HR for OS<br />

0.57 if baseline CD4 count �100/ul vs. �100/ul; p�0.001). For all<br />

histologies, initial therapy with the EPOCH regimen resulted in a better CR<br />

rate (OR 1.78, p�0.039), PFS (HR 0.61, p�0.032) and OS (HR 0.47,<br />

p�0.001) when compared to CHOP. Conclusions: In this pooled analysis<br />

including 1144 patients with HIV-associated NHL, the addition <strong>of</strong> rituximab<br />

to chemotherapy was associated with significantly improved CR rate,<br />

PFS, and OS, specifically for patients with a baseline CD4 count �100/uL.<br />

Treatment with infusional EPOCH was also more effective than CHOP.<br />

8007 Oral Abstract Session, Sat, 3:00 PM-6:00 PM<br />

A subgroup analysis <strong>of</strong> small lymphocytic and marginal zone lymphomas in<br />

the Eastern Cooperative Oncology Group protocol E4402 (RESORT): A<br />

randomized phase III study comparing two different rituximab dosing<br />

strategies for low tumor burden indolent non-Hodgkin lymphoma. Presenting<br />

Author: Michael E. Williams, University <strong>of</strong> Virginia Medical Center,<br />

Charlottesville, VA<br />

Background: Management <strong>of</strong> low tumor burden (LTB) indolent lymphoma in<br />

the rituximab (R) era is uncertain. We hypothesized that R could delay the<br />

need for chemotherapy and that maintenance R (MR) would be superior to<br />

R retreatment (RR) at progression. E4402 is a randomized phase III study<br />

comparing MR and RR for previously untreated, LTB (by GELF criteria)<br />

small lymphocytic lymphoma (SLL), marginal zone lymphoma (MZL) and<br />

FL. Results for the FL subset was previously presented (Kahl, et al. Blood<br />

2011; 118(21): LBA 6); we now report outcomes for the non-FL patients<br />

(pt). Methods: Pt received R 375 mg/m2 weekly x 4, with responders<br />

randomized to MR (1 doseRq3mo)orRR(Rqwkx4atprogression), each<br />

continued until treatment failure. The primary endpoint, time to treatment<br />

failure (TTTF), was defined as progression within 6 mo <strong>of</strong> last R, no<br />

response to RR, initiation <strong>of</strong> alternative therapy, or inability to complete<br />

protocol therapy. Pt were evaluated q 3 mo, with CT scans q 6 mo.<br />

Secondary endpoints: time to first cytotoxic therapy (TTCT), quality <strong>of</strong> life<br />

(QOL) and safety. Results: From 11/03 to 9/08,137 non-FL pt were<br />

enrolled. Complete or partial response was achieved in 57 (41%), who were<br />

randomized to MR (n�32) or RR (n�25). 136 pt were stage III-IV (1 IE),<br />

and all had PS 0-1; for MR vs RR, median age 66 vs 64, and M:F 47:53%<br />

vs. 28:72%, respectively. The mean no. <strong>of</strong> R doses/pt (incl. 4 induction<br />

doses) was 17.9 (range 5- 30) for MR and 5.8 (range 4-12) for RR. With a<br />

median follow-up <strong>of</strong> 4.3 yr, TTTF was 3.74 yr for MR vs. 1.07 yr for RR<br />

(p�.0002; HR 4.95). At 3 yr, 100% <strong>of</strong> MR vs. 70% <strong>of</strong> RR pt (p�.0002)<br />

remained free <strong>of</strong> cytotoxic therapy. Grade 3-4 toxicities occurred in 2 MR<br />

pt, 1 neutropenia and 1 encephalopathy. Conclusions: A planned subgroup<br />

analysis <strong>of</strong> non-FL pt showed significant benefit in TTTF and TTTC for MR<br />

but with 2 grade 4 toxicities. This differs from the FL pt in this trial, for<br />

whom response to induction was higher (70 vs. 41%; p�.0001) and where<br />

no TTTF benefit was observed with MR. LTB non-follicular indolent<br />

lymphoma pt who achieve a CR or PR to induction R benefit from MR<br />

therapy.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


512s Lymphoma and Plasma Cell Disorders<br />

8009 Oral Abstract Session, Sun, 8:00 AM-11:00 AM<br />

Phase I/II study <strong>of</strong> carfilzomib plus melphalan-prednisone (CMP) in elderly<br />

patients with de novo multiple myeloma. Presenting Author: Brigitte Kolb,<br />

University Hospital, Reims, France<br />

Background: Melphalan-prednisone � thalidomide (MPT) or bortezomib<br />

(MPV) are approved in frontline MM patients (pts) �65 years. Both<br />

regimens demonstrated significant benefit over MP alone in terms <strong>of</strong> PFS<br />

and OS but this benefit could be hampered by the risk <strong>of</strong> peripheral<br />

neuropathy (PN). Carfilzomib (Cfz) is a novel proteasome inhibitor that has<br />

demonstrated promising activity and favorable toxicity pr<strong>of</strong>ile, with low<br />

rates <strong>of</strong> PN. This phase I/II study was designed to determine the maximum<br />

tolerated dose (MTD) <strong>of</strong> CMP, to assess safety and evaluate efficacy <strong>of</strong> this<br />

combination in newly diagnosed MM �65. Methods: In Phase I, Cfz was the<br />

only escalating agent starting at 20 mg/m2 (level 1) with maximal planned<br />

dose 36 mg/m2 (level 3), given IV on days 1, 2, 8, 9, 22, 23, 29, 30 for<br />

nine 42-day cycles. Oral melphalan 9 mg/m² and prednisone 60mg/m²<br />

were given on days 1 to 4, for all dose levels. Based on toxicity assessment,<br />

the study was amended to add dose level 4 (Cfz 45 mg/m2). MTD<br />

determination was based on occurrence <strong>of</strong> Dose limiting toxicities (DLTs)<br />

during the first cycle only. DLTs were defined as any grade 4 hematologic<br />

toxicity or preventing administration <strong>of</strong> 2 or more <strong>of</strong> the 8 Cfz doses <strong>of</strong> the<br />

first treatment cycle except grade 4 thrombocytopenia without bleeding or<br />

grade 4 neutropenia lasting � 7 days; or grade � 3 febrile neutropenia; or<br />

any other grade � 3 nonhematologic toxicity. Results: As <strong>of</strong> January 20th<br />

2012, 24 pts have been enrolled in the phase I: 6 pts at level 1 (Cfz 20), 6<br />

at level 2 (Cfz 27), 6 at level 3 (Cfz 36), and 6 at level 4 (Cfz 45). There<br />

were 2 DLTs at level 4 (fever and hypotension not related to sepsis) and the<br />

MTD was considered to be 36 mg/m². Then, 16 additional pts were<br />

included in the phase II at level 3. Overall, 40 pts have been enrolled into<br />

the phase I/II study and 26 pts are evaluable for response. The ORR was<br />

92% including 42% at least VGPR. These results compare favorably to<br />

those achieved with MPV, MPT, MPR or lenalidomide-dex (ORR 71, 76, 80<br />

and 85%, respectively) in the same population. Conclusions: Frontline<br />

carfilzomib (36 mg/m2) � MP is a tolerable and very effective combination<br />

in elderly MM pts. Treatment is ongoing, with updated toxicity and efficacy<br />

data to be presented at the meeting.<br />

8011 Oral Abstract Session, Sun, 8:00 AM-11:00 AM<br />

Stringent complete response (sCR) in patients (pts) with newly diagnosed<br />

multiple myeloma (NDMM) treated with carfilzomib (CFZ), lenalidomide<br />

(LEN), and dexamethasone (DEX). Presenting Author: Andrzej J. Jakubowiak,<br />

University <strong>of</strong> Chicago, Chicago, IL<br />

Background: Combination treatment (tx) with CFZ, LEN, and DEX (CRd) is<br />

well tolerated and highly active in NDMM. In a phase 1/2 study, CRd<br />

provided rapid reduction <strong>of</strong> disease by 68% after cycle (C) 1 and 94%<br />

�partial response (PR) at a median <strong>of</strong> 8C, including 65% �very good PR<br />

and 53% �near CR (nCR), which improved to 79% �nCR after C12 (ASH<br />

2011, Abstr 631). Here, we examine the clinical significance <strong>of</strong> the<br />

response rates with longer follow-up. Methods: Pts with NDMM were treated<br />

in 28-day (d) C with CFZ 20–36 mg/m2 IV (d1, 2, 8, 9, 15, 16), LEN 25 mg<br />

PO (d1–21) and DEX 40/20 mg PO weekly (C1–4/5–8). After C4,<br />

autologous stem cell transplant (ASCT) candidates achieving �PR could<br />

collect stem cells but then continued CRd with the option to proceed to<br />

ASCT. After C8, pts received CRd maintenance, using the last tolerated<br />

doses, with LEN/DEX at the same schedule but a modified CFZ schedule<br />

(d1, 2, 15, 16). Response was assessed by IMWG criteria plus nCR.<br />

Results: As <strong>of</strong> Nov 30, 2011, median follow-up was 14 mo (range 4–25)<br />

with 33/53 (62%) pts achieving �nCR and 42% sCR. After a median <strong>of</strong><br />

13C (range 1–25), 36 pts completed C8 and continued CRd maintenance,<br />

64% achieving sCR. 20/22 pts in CR evaluated for minimal residual<br />

disease (MRD) by multiparameter flow cytometry had no MRD. Progressionfree<br />

survival (PFS) rate was 97% at 12 and 92% at 24 mo. All pts who<br />

achieved sCR have maintained response for a median <strong>of</strong> 9 mo (range<br />

1–20). Extended CRd tx was well tolerated. During CRd maintenance, the<br />

most common toxicities (all grades) were lymphopenia (30%), leukopenia<br />

(26%), and fatigue (25%), and peripheral neuropathy remained limited<br />

(11%, all G1/2). There were no tx discontinuations due to toxicity during<br />

maintenance and limited dose modifications (CFZ 19%, LEN 28%, DEX<br />

31%). Conclusions: CRd is highly active in NDMM, providing rapid and<br />

deep responses. Extended tx was well tolerated and resulted in improved<br />

depth <strong>of</strong> response with a high sCR rate and a significant proportion <strong>of</strong> pts<br />

without evidence <strong>of</strong> MRD. Responses were durable with very promising<br />

PFS. All pts who achieved sCR remained on CRd with sustained sCR. These<br />

results compare favorably to other frontline regimens.<br />

8010 Oral Abstract Session, Sun, 8:00 AM-11:00 AM<br />

A phase I/II trial <strong>of</strong> cyclophosphamide, carfilzomib, thalidomide, and<br />

dexamethasone (CYCLONE) in patients with newly diagnosed multiple<br />

myeloma. Presenting Author: Joseph Mikhael, Mayo Clinic, Scottsdale, AZ<br />

Background: Carfilzomib is a proteasome inhibitor that irreversibly binds its<br />

target with favorable toxicity pr<strong>of</strong>ile that has shown significant activity in<br />

relapsed multiple myeloma (MM). Here we used carfilzomib as the center <strong>of</strong><br />

a 4 drug induction regimen designed for MM patients pre stem cell<br />

transplant (SCT). Methods: We conducted a phase I safety run in 6 patients<br />

with no DLT observed before expanding to phase II. The phase II regimen is<br />

shown below. Treatment was for 4 cycles with expected SCT post induction.<br />

For the phase II portion <strong>of</strong> this trial, the primary endpoint is the proportion<br />

<strong>of</strong> patients who have � very good partial response to treatment. All patients<br />

received herpes zoster prophylaxis and ASA daily. Results: Twenty seven<br />

patients were enrolled. Median age was 65 (range 27-74). ORR for<br />

evaluable patients (n�17) at phase II dosing is 100%: CR 35%, VGPR<br />

48%, PR 18% after 4 cycles <strong>of</strong> CYCLONE. Grade 3 toxicity was reported in<br />

52% <strong>of</strong> patients and 14% experienced a grade 4 toxicity. Grade 3/4<br />

toxicities occurring in �5% <strong>of</strong> patients included fatigue, neutropenia,<br />

lymphopenia, thromboembolism, myopathy. Toxicities <strong>of</strong> any grade seen in<br />

�20% <strong>of</strong> patients included fatigue, constipation, lethargy, thrombocytopenia,<br />

somnolence, creatinine increased, malaise. Four patients (20%)<br />

developed grade 1 sensory neuropathy; no higher grade or painful neuropathy<br />

was evident. All patients are alive. All patients advancing to SCT<br />

successfully collected stem cells. One patient with t(4;14) disease who was<br />

not transplanted has progressed. 94% remain progression free. Conclusions:<br />

The 4 drug CYCLONE regimen has remarkable efficacy (83% �VGPR) and<br />

manageable toxicity in newly diagnosed patients with multiple myeloma.<br />

Especially notable was the low incidence <strong>of</strong> neuropathy and depth <strong>of</strong><br />

response (CR 35%) after only 4 cycles. Given the relative lack <strong>of</strong> toxicity an<br />

extension <strong>of</strong> this regimen at higher doses <strong>of</strong> carfilzomib (20/45mg/m2 ) has<br />

been initiated.<br />

Agent Dose level Route Day Cycle length<br />

Carfilzomib 20 mg/m2 cycle 1<br />

27mg/m2 IV 1, 2, 8, 9, 15, 16 28 days<br />

� cycle 2<br />

Thalidomide 100 mg Orally 1-28 28 days<br />

Cyclophosphamide 300 mg/m2 Orally 1,8,15 28 days<br />

Dexamethasone 40 mg Orally 1,8,15,22 28 days<br />

8012 Oral Abstract Session, Sun, 8:00 AM-11:00 AM<br />

PANORAMA 2: A phase II study <strong>of</strong> panobinostat in combination with<br />

bortezomib and dexamethasone in patients with relapsed and bortezomibrefractory<br />

multiple myeloma. Presenting Author: Melissa Alsina, H. Lee<br />

M<strong>of</strong>fitt Cancer Center & Research Institute, Tampa, FL<br />

Background: Patients (pts) with multiple myeloma (MM) refractory to<br />

bortezomib (BTZ) and an immunomodulatory drug have limited treatment<br />

options and a poor prognosis. In a phase I study <strong>of</strong> pts with relapsed or<br />

relapsed/refractory MM treated with panobinostat (PAN) � BTZ, clinical<br />

responses were observed overall and in pts with BTZ-refractory disease. We<br />

report results in PANORAMA 2, a trial in relapsed and BTZ-refractory pts.<br />

Methods: PANORAMA 2 is a single-arm, phase II study <strong>of</strong> PAN � BTZ �<br />

dexamethasone (Dex) in pts with relapsed and BTZ-refractory MM. Treatment<br />

phase 1 (TP1) consists <strong>of</strong> eight 3-week cycles <strong>of</strong> oral PAN �<br />

intravenous BTZ � oral Dex. Pts demonstrating clinical benefit enter<br />

treatment phase 2 (TP2) which consists <strong>of</strong> four 6-week cycles <strong>of</strong> PAN �<br />

BTZ � Dex. The primary endpoint is overall response (� partial response<br />

[PR]) in TP1. Results: Fifty-five pts with BTZ-refractory MM were enrolled<br />

with 10 pts ongoing and 28 in follow-up. The median age was 61 years<br />

(range 41-88 years). Pts were heavily pretreated: the median number <strong>of</strong><br />

prior regimens was 4 (range 2-11), and most pts (64%) received prior<br />

autologous stem cell transplant. Twenty-seven (49%) and 36 (65%) pts<br />

had BTZ and Dex in their most recent prior line <strong>of</strong> therapy, respectively.<br />

Eighteen pts achieved � PR for an overall response rate <strong>of</strong> 33% (1 near<br />

complete response and 17 PR), and 13 pts achieved MR for a clinical<br />

benefit rate <strong>of</strong> 56%. Three pts achieved a VGPR. Eighteen pts completed<br />

TP1 and entered TP2, and 2 have completed � 12 cycles. Common<br />

adverse events (AEs) <strong>of</strong> any grade included thrombocytopenia (66%),<br />

fatigue (64%), diarrhea (62%), nausea (58%), dyspnea (40%), anemia<br />

(38%), decreased appetite (36%), and dizziness (36%). Common grade<br />

3/4 AEs included thrombocytopenia (58%), fatigue (17%), anemia (15%),<br />

pneumonia (15%), neutropenia (13%), and diarrhea (13%). Only 1 pt<br />

(2%) experienced grade 3/4 peripheral neuropathy. Conclusions: PAN<br />

synergizes with BTZ in recapturing responses in heavily pretreated,<br />

BTZ-refractory MM pts. The combination <strong>of</strong> PAN and BTZ is a promising<br />

treatment for pts with BTZ-refractory MM that is generally well tolerated,<br />

with several pts receiving therapy long term.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


8013 Oral Abstract Session, Sun, 8:00 AM-11:00 AM<br />

Phase I trial <strong>of</strong> obatoclax mesylate in combination with bortezomib for<br />

treatment <strong>of</strong> relapsed multiple myeloma. Presenting Author: A. Keith<br />

Stewart, Mayo Clinic, Scottsdale, AZ<br />

Background: Obatoclax mesylate (GX15-070MS) is a BH3 mimetic that<br />

inhibits Bcl-2 protein family members including MCL-1, a dominant target<br />

in myeloma (MM). Obatoclax (OBX) inhibited viability <strong>of</strong> 14 MM cell lines<br />

(mean IC50 215 nM) and primary MM samples while exhibiting pre clinical<br />

synergy with bortezomib (BTZ). Sensitivity correlated with basal levels <strong>of</strong><br />

Mcl-1 and Bcl-XL, but not Bcl2, Bim, Bax or Bak expression. Methods: We<br />

report a phase I trial <strong>of</strong> OBX in combination with BTZ. Eligibility required<br />

measureable disease, � 1 prior MM therapy, �10 cycles <strong>of</strong> prior BTZ and<br />

did not progress on prior BTZ therapy, creatinine �2 ULN. Starting dose<br />

level 1 was OBX 14 mg/m2 24-hour continuous iv. infusion days 1, 8, 15 <strong>of</strong><br />

a 21-day cycle. BTZ given at 1.3mg/m2 iv. days 1, 4, 8 and 11. After<br />

protocol amendment OBX level 1 dosing was 30 mg/m2 , level 2 was 40<br />

mg/m2 IV both by continuous 3 hour infusion days 1, 8 and 15 on a 21 day<br />

schedule. Pre med. with famotidine was required. Results: Eleven patients<br />

were accrued, median age 62 (range: 46-77), median time from diagnosis<br />

was 4.7 years. Median <strong>of</strong> 2.5 cycles (range: 1-10). Median follow-up for<br />

patients still alive is 11.6 months (range: 0.9-35.5). At dose level 1, there<br />

were 2 DLTs. After amendment 8 patients were accrued (3 hour infusion): 4<br />

at amended dose level 1 and 4 at dose level 2. All patients are now <strong>of</strong>f<br />

treatment. 10 patients are evaluable for response: 2 patients at original<br />

dose level 1 (2 PR), 3 patients at dose level 1 (2 PR, 1 MR), no patients at<br />

dose level 2 responded: overall PR <strong>of</strong> 40%, clinical benefit response in<br />

50% (95% CI: 19-81%). 6 patients had disease progression and 2 patients<br />

died. 4 DLTs were seen: at original dose level 1 grade 4 thrombocytopenia<br />

and delay <strong>of</strong> therapy � 15 days. At dose level 2, 1 patient had grade 3<br />

somnolence, a 2nd patient grade 3 euphoria and grade 4 thrombocytopenia.<br />

No DLTs were seen at amended dose level 1. Common adverse events<br />

<strong>of</strong> any grade included GI, hematologic and neurologic e.g. euphoria,<br />

decreased level <strong>of</strong> consciousness, psychosis, speech. Conclusions: In<br />

summary MTD is OBX 30mg/m2 by 3 hour iv infusion once weekly, BTZ 1.3<br />

mg/m2 days 1,4,8, and 11. Major toxicities were central neurologic and<br />

hematologic. This P2C consortium study was supported by NCI N01-<br />

CM62205.<br />

8015 Oral Abstract Session, Sun, 8:00 AM-11:00 AM<br />

Efficacy and side-effect pr<strong>of</strong>ile <strong>of</strong> long-term bisphosphonate therapy in<br />

patients (pts) with multiple myeloma (MM): MRC myeloma IX study results.<br />

Presenting Author: Gareth J Morgan, Institute <strong>of</strong> Cancer Research, Royal<br />

Marsden Hospital, London, United Kingdom<br />

Background: Bisphosphonates (BPs) are recommended in pts with osteolytic<br />

lesions from MM. However, data on the long-term efficacy and safety <strong>of</strong><br />

BPs beyond 2yissomewhat limited. The MRC Myeloma IX study has<br />

already revealed significant overall survival (OS) and progression-free<br />

survival (PFS) benefits for zoledronic acid (ZOL) over clodronate (CLO) in<br />

MM pts (N � 1960) initiating chemotherapy (Morgan GJ, et al. Lancet.<br />

2010). We now report the efficacy and safety <strong>of</strong> BP therapy with long-term<br />

follow-up. Methods: Newly diagnosed MM pts were randomized to ZOL (4<br />

mg IV q 21-28 days) or CLO (1600 mg/day PO) plus antimyeloma therapy.<br />

BPs continued at least until disease progression. PFS and OS were<br />

estimated using Kaplan-Meier methodology. Hazard ratios (HRs) were<br />

calculated using stratified Cox models. Adverse events (AEs) were monitored<br />

continuously and analyzed using cumulative incidence functions.<br />

Results: At a median follow-up <strong>of</strong> 5.8 y in 1960 evaluable pts, ZOL<br />

improved PFS (HR � 0.88; P � .01) and OS (HR � 0.88; P � .03) vs CLO.<br />

Both BPs were generally well tolerated, and acute renal failure events were<br />

similar between groups (ZOL 5.2% vs CLO 5.8% at 2 y, with incidence<br />

plateaued thereafter). Overall incidence <strong>of</strong> confirmed osteonecrosis <strong>of</strong> the<br />

jaw (ONJ) has remained low (ZOL 3.7% vs CLO 0.5%; P � .0001). ONJ<br />

incidence was lower among pts receiving thalidomide-containing regimens<br />

(1.4%) vs no thalidomide (2.76%; P � .041). Events were generally<br />

low-grade, most occurred between 8 and 30 mo (median time to ONJ �<br />

23.7 mo), and cumulative incidence plateaued at ~36 mo. Ten pts had<br />

data on ONJ recovery: complete recovery in 4 pts, improvement in 2 pts, no<br />

change in 3 pts in the ZOL group; and no change in 1 CLO pt. Dental<br />

surgery or trauma preceded ONJ in 6 ZOL pts. Conclusions: ZOL provided<br />

sustained PFS and OS improvements vs CLO during long-term therapy in<br />

the MRC Myeloma IX study. Overall incidence <strong>of</strong> AEs was similar between<br />

groups, with no notable changes during long-term therapy. ONJ incidence<br />

remained low during long-term (� 2.5 y) therapy and was reduced in pts<br />

receiving thalidomide (possibly because <strong>of</strong> anticytokine effects <strong>of</strong> this<br />

agent).<br />

Lymphoma and Plasma Cell Disorders<br />

8014 Oral Abstract Session, Sun, 8:00 AM-11:00 AM<br />

Bendamustine, bortezomib, and dexamethasone (BVD) in elderly patients<br />

with relapsed/refractory multiple myeloma (RRMM): The Intergroupe Francophone<br />

du Myélome (IFM) 2009-01 protocol. Presenting Author: Philippe<br />

Rodon, Hematology, General Hospital, Blois, France<br />

Background: Bortezomib (V) plus dexamethasone (D) is a treatment <strong>of</strong><br />

choice <strong>of</strong> RRMM. In small series, the addition <strong>of</strong> an alkylator was<br />

beneficial. Bendamustine (B) showed a high activity in advanced MM. The<br />

IFM 2009-01 trial evaluates the combination <strong>of</strong> B, V and D in elderly pts<br />

with MM progressive on or after 1st line therapy. Methods: We conducted a<br />

phase 2 trial combining B 70 mg/m2 D1-8, V 1.3 mg/m2 D1-8-15-22 and D<br />

20 mg D1-8-15-22 every 28 days. 4 cycles were administered. In<br />

responders (PR or better), 2 additional cycles were provided followed by a<br />

maintenance phase with 6 cycles given every 2 months. Inclusion criteria<br />

were progression on or after 1 prior line <strong>of</strong> therapy, measurable disease, PS<br />

ECOG �3, ANC � 1.5x109 /l, platelets � 100x109 /l, creatinine � 250<br />

mcmol/l, AST and ALT � 3xULN. Pts with prior exposure to bortezomib<br />

were excluded. Response was evaluated according to IMWG criteria.<br />

Primary end point was response at end <strong>of</strong> cycle 4, secondary objectives<br />

overall response rate (ORR), progression-free survival (PFS), overall survival<br />

(OS) and toxicity. Results: The present analysis was restricted to the first 4<br />

cycles. From 03/2010 to 07/2011, 73 pts were included, median age 75.8<br />

years (range 66-86). Median time from diagnosis to inclusion was 29<br />

months. All pts received only 1 prior therapy: MP in 12, MP-thalidomide in<br />

44, lenalidomide-dexamethasone (LD) in 14, other in 3. 42 pts (57.5%)<br />

were responders at end <strong>of</strong> cycle 4 [CR: 8 (10.9%), VGPR: 9 (12.3%), PR:<br />

25 (34.2%), SD: 10 (13.6%), progression: 11 (15%), early discontinuation:<br />

10 (13.6%)]. 6pts/10 were in PR and 1pt/10 in VGPR at time <strong>of</strong><br />

discontinuation. ORR was 67.1% (49/73 pts). 11 pts died (MM: 6, sepsis:<br />

4, renal failure: 1). Adverse events grade 3-4 were neutropenia: 16 pts,<br />

thrombocytopenia: 7 pts, sepsis: 12 pts, gastro-intestinal: 8 pts, anaphylaxis:<br />

1 pt. 2 pts had DVT. Peripheral neuropathy grade�1 occurred in 9<br />

pts, all grade 2. Treatment was stopped in 20 pts (lack <strong>of</strong> efficacy: 11,<br />

toxicity: 9). Conclusions: These results compare favorably with those<br />

achieved with VD or LD. The triplet BVD combination is very effective and<br />

tolerable in elderly pts with MM in 1st progression.<br />

8016 Oral Abstract Session, Sun, 8:00 AM-11:00 AM<br />

Pomalidomide (POM) with or without low-dose dexamethasone (LoDEX) in<br />

patients (pts) with relapsed/refractory multiple myeloma (RRMM): Outcomes<br />

in pts refractory to lenalidomide (LEN) and/or bortezomib (BORT).<br />

Presenting Author: Ravi Vij, Washington University School <strong>of</strong> Medicine, St.<br />

Louis, MO<br />

Background: Response and survival outcomesare poor in pts with RRMM<br />

who are refractory to BORT and immunomodulatory drugs. POM�LoDEX<br />

has demonstrated activity in pts with advanced multiple myeloma who have<br />

received multiple lines <strong>of</strong> therapy. This analysis evaluates outcomes in pts<br />

with disease refractory to LEN, BORT, or both, as well in pts with disease<br />

refractory to both LEN and BORT who had received prior transplant.<br />

Methods: In the MM-002 phase II trial, pts received POM (4 mg/day on days<br />

1–21 <strong>of</strong> each 28-day cycle) alone (n�108) or in combination with LoDEX<br />

(40 mg/week) (n�113). Refractory disease was defined as documented<br />

progression during treatment or within 60 days <strong>of</strong> the last dose <strong>of</strong><br />

treatment. Response rates were assessed using European Group for Bone<br />

Marrow Transplantation (EBMT) criteria by independent adjudication<br />

committee. Results: Patients who received POM�LoDEX were refractory to<br />

LEN (77%), BORT (73%), or both (61%). Forty-two percent were refractory<br />

to both LEN and BORT and had received prior transplant. Overall, 20% <strong>of</strong><br />

pts achieved � PR, median PFS was 3.5 months, and 1-year survival rate<br />

was 59%. Response rates and duration were comparable in pts with disease<br />

refractory to LEN, BORT, or both, and in pts who had received prior<br />

transplant (response rate 25-34%, median duration <strong>of</strong> response 5.7-7<br />

months). Survival outcomes were similar across the groups (median PFS<br />

3.8-4.6 months; 1-year survival rate 60-67%). Conclusions: POM, given at<br />

4 mg/day on days 1–21 <strong>of</strong> each 28-day cycle in combination with LoDEX,<br />

40 mg/week, is effective in pts with RRMM refractory to LEN, BORT, or<br />

both, and including those with prior transplant. These results suggest a lack<br />

<strong>of</strong> cross-resistance between POM and LEN, and confirm activity not only in<br />

BORT-refractory pts but also those for whom transplant has failed.<br />

Refractory to:<br />

LEN<br />

n�87 BORT<br />

n�82<br />

LEN and BORT<br />

n�69<br />

LEN and BORT<br />

with prior transplant<br />

n�47<br />

POM�LoDEX<br />

> PR, % 25 29 28 34<br />

> MR, % 41 46 46 53<br />

Median duration <strong>of</strong> response, months* 7 5.8 6.2 5.7<br />

Median PFS, months 3.8 3.8 3.8 4.6<br />

1-year survival rate, %<br />

* In patients achieving �PR.<br />

65 60 61 67<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.<br />

513s


514s Lymphoma and Plasma Cell Disorders<br />

8017 Oral Abstract Session, Sun, 8:00 AM-11:00 AM<br />

Phase I study <strong>of</strong> twice-weekly dosing <strong>of</strong> the investigational oral proteasome<br />

inhibitor MLN9708 in patients (pts) with relapsed and/or refractory<br />

multiple myeloma (MM). Presenting Author: Sagar Lonial, Winship Cancer<br />

Institute, Emory University, Atlanta, GA<br />

Background: MLN9708 is the first oral proteasome inhibitor to enter clinical<br />

investigation in MM. This study (NCT00932698) assessed safety, MTD,<br />

and response rate with twice-weekly oral MLN9708 in pts with relapsed<br />

and/or refractory MM, and characterized plasma pharmacokinetics (PK)<br />

and blood pharmacodynamics. Methods: Pts aged �18 yrs with measurable<br />

MM received MLN9708 on d 1, 4, 8, and 11 <strong>of</strong> 21-d cycles. In the<br />

dose-escalation phase, pts required �2 prior therapies (including bortezomib,<br />

thalidomide/lenalidomide, and corticosteroids). At the MTD (2.0<br />

mg/m2 ), pts were enrolled to relapsed and refractory [RR], bortezomibrelapsed<br />

[VR], proteasome-inhibitor [PI] naïve, and carfilzomib [CZ] expansion<br />

cohorts. Results: 57 pts (53% M) were enrolled, 37 to the expansion<br />

cohorts (16 RR, 14 VR, 6 PI naïve, 1 CZ). Median age was 65 yrs (range<br />

50-86). Median number <strong>of</strong> prior lines <strong>of</strong> therapy was 4 (range 1-28); 88%,<br />

84%, 61%, and 5% had prior bortezomib, lenalidomide, thalidomide, and<br />

carfilzomib, respectively. Pts have received a median <strong>of</strong> 3 cycles (range<br />

1-24) to date (data cut-<strong>of</strong>f Dec 1, 2011); 7 (12%) have received �13<br />

cycles. Drug-related AEs were seen in 89% <strong>of</strong> pts, including fatigue (46%),<br />

thrombocytopenia (40%), and nausea (30%); 63% had drug-related grade<br />

�3 AEs, including thrombocytopenia (33%), neutropenia (14%), fatigue<br />

(9%), and rash (7%). Only 6 (11%) pts had drug-related peripheral<br />

neuropathy (PN; no grade �3). 7 pts discontinued due to AEs. 2 pts died on<br />

study, due to PD and an unrelated cardiac disorder. Of 46 responseevaluable<br />

pts, 6 have achieved �PR, with 1 sCR (PI naïve cohort) and 5<br />

PRs (2 in dose-escalation, 1 in RR, 2 in VR cohorts), and 1 VR pt has<br />

achieved MR, with duration <strong>of</strong> disease control <strong>of</strong> up to 18.6 mo. PK<br />

analyses showed MLN2238 (biologically active hydrolysis product) has<br />

linear plasma PK (0.8-2.23 mg/m2 ), Tmax <strong>of</strong> 0.5-1.25 hr, and terminal<br />

half-life <strong>of</strong> 4-6 d. A dose-dependent increase in whole blood 20S<br />

proteasome inhibition was observed. Conclusions: Current data suggest<br />

MLN9708 has clinical activity in heavily pretreated MM pts, with durable<br />

responses/disease control, and is generally well tolerated with infrequent<br />

low-grade PN.<br />

8019 <strong>Clinical</strong> Science Symposium, Mon, 11:30 AM-1:00 PM<br />

Daratumumab, a CD38 mab, for the treatment <strong>of</strong> relapsed/refractory<br />

multiple myeloma patients: Preliminary efficacy data from a multicenter<br />

phase I/II study. Presenting Author: Torben Plesner, Vejle Hospital, Vejle,<br />

Denmark<br />

Background: Daratumumab (HuMax-CD38) is a human CD38 monoclonal<br />

antibody with broad-spectrum killing activity; it effectively kills CD38expressing<br />

tumor cells via antibody-dependent cell-mediated cytotoxicity<br />

(ADCC), complement-dependent cytotoxicity (CDC) and apoptosis. From an<br />

ongoing first-in-human (FIH) dose-escalation study (<strong>Clinical</strong>Trials.gov<br />

CT00574288), it has been shown that daratumumab has an acceptable<br />

safety pr<strong>of</strong>ile (Gimsing: ASH 2011 abstract 1873). The objectives <strong>of</strong> this<br />

FIH study are to establish the safety pr<strong>of</strong>ile and MTD. In addition efficacy is<br />

evaluated. Methods: Pts �18 years and previously diagnosed with MM<br />

requiring systemic therapy and considered relapsed or refractory (RR) to at<br />

least two different prior lines <strong>of</strong> therapy and not eligible for salvage ASCT<br />

were enrolled. The design <strong>of</strong> this study encompasses an accelerated<br />

dose-escalation based on a classical 3�3 design. Daratumumab is administrated<br />

overa9wkperiod encompassing 2 pre-doses and 7 full-doses. The<br />

doses range from 0.005 mg/kg to 24 mg/kg. The decision to dose escalate<br />

is based on recommendation by an external Independent Data Monitoring<br />

Committee. Evaluation <strong>of</strong> efficacy data was according to Rajkumar (Blood<br />

2011;117:4691-5). The results presented in this abstract are based on<br />

preliminary data analyzed before database lock. Results: Data from 23 pts<br />

including the 4 mg/kg group are collected. Preliminary efficacy evaluation<br />

is based on best paraprotein response as reflected by change in serum<br />

and/or urine M-component. For groups � 1 mg/kg, 3/17 pts achieved a<br />

reduction in serum M-component (12%, 14%, 19%), in the 2 mg/kg group,<br />

1/3 pts had a reduction in urine M-component (55%), and in the 4 mg/kg<br />

group, 3/3 pts had a reduction in the serum M-component <strong>of</strong> 49%, 55, and<br />

64%, respectively. In the 4 mg/kg group a marked reduction in the<br />

percentage <strong>of</strong> plasma cells in the bone marrow was seen in all pts (80%,<br />

89%, and 97%). The toxicity was manageable. Conclusions: Daratumumab<br />

treatment resulted in reductions in M-component and bone marrow plasma<br />

cells in pts with RR MM. Toxicity has been manageable. Additional data<br />

will be presented at the meeting.<br />

8018^ <strong>Clinical</strong> Science Symposium, Mon, 11:30 AM-1:00 PM<br />

Phase II, randomized, double blind, placebo-controlled study comparing<br />

siltuximab plus bortezomib versus bortezomib alone in pts with relapsed/<br />

refractory multiple myeloma. Presenting Author: Robert Z. Orlowski,<br />

University <strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: Preclinical studies <strong>of</strong> siltuximab (S), a chimeric anti-IL-6<br />

mAb, in combination with bortezomib (B) indicate an additive to synergistic<br />

effect in multiple myeloma (MM) cell lines. This randomized study<br />

evaluated the safety and efficacy <strong>of</strong> S�B compared with placebo (plc)�B<br />

in pts with relapsed/refractory MM after 1�3 prior tx lines, measurable<br />

disease but no prior B exposure. Methods: 286 pts were randomized 1:1 to<br />

S�B: B�plc. S 6 mg/kg or plc was given IV q2w. B 1.3 mg/m2 was given IV<br />

on d 1, 4, 8, 11, 22, 25, 29, 32 for a max <strong>of</strong> 4 <strong>of</strong> 42-d cycles and then<br />

reduced to q1w for 35-d cycles. B was stopped for pts with PD/<br />

intolerability, and high dose oral dexamethasone (dex) 40 mg could then be<br />

started qd on d 1�4, 9�12, 17�20 for a max <strong>of</strong> 4 <strong>of</strong> 28-d cycles and on d<br />

1�4 <strong>of</strong> subsequent cycles until PD, while S/plc continued. Primary<br />

endpoint was PFS by EBMT criteria censored at the start <strong>of</strong> dex/subsequent<br />

tx. Secondary endpoints included overall response rate (ORR), OS, and<br />

safety before dex. Results: 142 and 144 pts received S�B and B�plc,<br />

respectively. Baseline demographics and disease characteristics were well<br />

balanced across S�B and B�plc, except for age (median 64 vs. 61 yrs) and<br />

myeloma type (IgG 65 vs. 71%, IgA 27 vs. 20%). Median tx duration was<br />

5.1 mo in both grps. Median PFS was 8.1 mo in S�B and 7.6 mo in B�plc<br />

(HR 0.869, p � 0.345). ORR (CR�PR) was 55% in pts on S�B and 47%<br />

on B�plc (p � 0.213); CR rates were 11 and 7% (p � 0.342),<br />

respectively. With 24.5 mo median follow up, median OS was 30.8 mo for<br />

S�B and 36.9 mo for B�plc (HR 1.353 for S�B, p � 0.103). Fewer pts on<br />

S�B than B�plc moved to dex (23 vs. 31%) and had subsequent SCT (5<br />

vs. 11%). Gr �3 AEs occurred in 91% on S�B and 74% on B�plc.<br />

Common gr �3 AEs in S�B were neutropenia (49%), thrombocytopenia<br />

(48%), leukopenia (14%). SAEs occurred in 29% on S�B and 24% on<br />

B�plc. Death occurred within 30 d <strong>of</strong> last study agent administration<br />

pre-dex in 8% on S�B and 5% on B�plc. Conclusions: The combination <strong>of</strong><br />

S�B had higher response rates but did not prolong survival compared with<br />

B�plc. A negative survival trend heavily influenced by differences in dex<br />

and SCT rescue was noted. S�B appears to be generally well tolerated but<br />

had a higher incidence <strong>of</strong> hematologic AEs.<br />

8020 <strong>Clinical</strong> Science Symposium, Mon, 11:30 AM-1:00 PM<br />

A randomized phase II study <strong>of</strong> elotuzumab with lenalidomide and low-dose<br />

dexamethasone in patients with relapsed/refractory multiple myeloma.<br />

Presenting Author: Philippe Moreau, Hematology Department, University<br />

Hospital, Nantes, France<br />

Background: Elotuzumab (Elo) is a humanized monoclonal IgG1 antibody<br />

targeting CS1, a cell surface glycoprotein. CS1 is highly expressed on<br />

�95% <strong>of</strong> multiple myeloma (MM) cells, with lower expression on natural<br />

killer cells and little to no expression on normal tissues. A phase 1 trial <strong>of</strong><br />

Elo plus lenalidomide and low-dose dexamethasone (Elo/Len/Dex) demonstrated<br />

an 82% objective response rate (ORR) in patients (pts) with<br />

relapsed/refractory (RR) MM (Lonial et al. J Clin Oncol, in press). Methods:<br />

In this phase 2 study, previously treated pts with MM were randomized to<br />

Elo 10 or 20 mg/kg IV (days 1, 8, 15, and 22 every 28-days in first 2 cycles<br />

and days 1 and 15 <strong>of</strong> subsequent cycles), Len 25 mg PO (days 1-21) and<br />

Dex 40 mg PO weekly. Prophylaxis for infusion-related reactions (IRs) was<br />

administered prior to each Elo infusion. Treatment continued until disease<br />

progression or unacceptable toxicity. The primary objective was to assess<br />

efficacy (ORR �partial response [PR]) according to IMWG criteria. Results:<br />

Among73 pts (median age 63 years; range, 39-82), 55% had received �2<br />

prior therapies, 60% had received prior bortezomib, and 62% prior<br />

thalidomide. ORR was 82% for all pts including 48% � very good PR<br />

(VGPR). The ORRs were 92% in the 10 mg/kg group (n�36) and 73% in<br />

the 20 mg/kg group (n�37). Median time to best response was 2.2 months<br />

(range, 0.7-17.5). After a median follow-up <strong>of</strong> 14.1 months, median<br />

progression-free survival (PFS) has not been reached, with PFS rates <strong>of</strong><br />

75% (10 mg/kg) and 65% (20 mg/kg). Elo/Len/Dex also showed encouraging<br />

activity in pts with high-risk cytogenetics (ORR 80%) and/or Stage 2-3<br />

MM (ORR 81%). The most common grade 3/4 toxicities were neutropenia<br />

(16%), lymphopenia (16%), and thrombocytopenia (16%). Investigatordesignated<br />

IRs were reported in 12% <strong>of</strong> pts (all grades); 1 pt (1.4%) had<br />

grade 3 IR (rash). Conclusions: Elo/Len/Dex was generally well tolerated and<br />

resulted in a high ORR, and PFS not reached after 14.1 months <strong>of</strong> median<br />

follow-up in pts with RR MM. Updated results will be presented at the<br />

meeting. Two phase 3 trials <strong>of</strong> 10 mg/kg Elo/Len/Dex are ongoing in newly<br />

diagnosed MM (ELOQUENT1; CA204-006; NCT01335399) and RR MM<br />

(ELOQUENT2; CA204-004; NCT01239797).<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


8021 Poster Discussion Session (Board #1), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

R-CHOP14 compared to R-CHOP21 in elderly patients with diffuse large<br />

B-cell lymphoma (DLBCL): Final analysis <strong>of</strong> the LNH03-6B GELA study.<br />

Presenting Author: Richard Delarue, Hôpital Necker, Paris, France<br />

Background: In 2000, the GELA established a new standard for elderly<br />

patients with DLBCL, demonstrating survival advantage <strong>of</strong> R-CHOP21 over<br />

CHOP21. Based on RICOVER-60 results, the DSHNHL proposed R-<br />

CHOP14 as a standard. Comparison between both regimens is lacking. We<br />

report the results <strong>of</strong> the final analysis <strong>of</strong> the randomized phase III trial<br />

LNH03-6B, with a median follow-up <strong>of</strong> 56 months. Methods: Pts between<br />

60 and 80 years old with DLBCL and aaIPI�1 were eligible. They were<br />

randomized between R-CHOP14 and R-CHOP21 for 8 cycles. G-CSF<br />

prophylaxis was given according to physician decision. Primary objective<br />

was to evaluate the efficacy <strong>of</strong> R-CHOP14 compared to R-CHOP21 as<br />

measured by the EFS. Results: 602 pts were randomized, 600 were<br />

evaluable, 304 with R-CHOP14 and 296 with R-CHOP21. Median age was<br />

70 years. Pts characteristics were similar between the two arms. Percentage<br />

<strong>of</strong> pts with baseline IPI3-5 was 72% in R-CHOP14 arm and 78% in<br />

R-CHOP21 arm. Median interval between 2 cycles was 14 d in R-CHOP14<br />

arm and 21 d in R-CHOP21 arm. In R-CHOP14 arms, 89% <strong>of</strong> cycles were<br />

administered with G-CSF. Median dose-intensity for R-CHOP14 arm was<br />

88% for cyclophosphamide and doxorubicin. There was no difference in<br />

median dose-intensity according to G-CSF administration at first cycle.<br />

Response rate (CR�CRu) was 71% in R-CHOP14 arm and 74% in<br />

R-CHOP21 arm (p�0.42). The 3-y EFS was 56% in R-CHOP14 arm and<br />

60% in R-CHOP21 (HR 1.04; CI95% 0.82-1.31; p�0.79). Moreover,<br />

there is no difference between both arms regarding 3-y PFS (60% vs. 62%;<br />

HR 0.99; CI95% 0.78-1.26; p�0.90), DFS (72% vs. 67%; HR 0.80;<br />

CI95% 0.58-1.10; p�0.80) and OS (69% vs. 72%; HR 0.96; CI95%<br />

0.73-1.26; p�0.75). Finally, percentage <strong>of</strong> patients with at least one<br />

serious adverse event (R-CHOP14: 51%; R-CHOP21: 47%) and rate <strong>of</strong><br />

toxic death (4.6% and 4.7% respectively) were similar. Conclusions:<br />

Results <strong>of</strong> the final analysis <strong>of</strong> LNH03-6B demonstrate similar efficacy and<br />

safety pr<strong>of</strong>ile between R-CHOP14 and R-CHOP21.<br />

8023 Poster Discussion Session (Board #3), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

A multicenter phase II study <strong>of</strong> bendamustine with rituximab in patients<br />

with relapsed/refractory diffuse large B-cell lymphoma (DLBCL). Presenting<br />

Author: Michinori Ogura, Department <strong>of</strong> Hematology and Oncology,<br />

Nagoya Daini Red Cross Hospital, Aichi, Japan<br />

Background: Effective salvage therapies are needed in patients (pts) with<br />

relapsed/refractory DLBCL after R-CHOP. Therapy with bendamustine plus<br />

rituximab (B-R) was well tolerated and effective in the preceding phase I<br />

study in relapsed/refractory aggressive B-cell non-Hodgkin lymphoma,<br />

including DLBCL. This phase II study assessed the efficacy and safety <strong>of</strong><br />

B-R in pts with relapsed/refractory DLBCL. Methods: Pts with histologically<br />

confirmed DLBCL (excluding transformed disease) and 1-3 prior therapies<br />

received rituximab 375 mg/m2 IV on day 1 and bendamustine 120 mg/m2 IV on days 2 and 3 <strong>of</strong> each 21-day cycle, for up to 6 cycles. Recovery <strong>of</strong><br />

neutrophil count to �1,000/mm3 and platelet count to �75,000/mm3 were required prior to the start <strong>of</strong> each cycle; treatment delays �2 weeks<br />

resulted in discontinuation. The primary endpoint was overall response rate<br />

(ORR); secondary endpoints included complete response (CR) rate, progression-free<br />

survival (PFS), and safety. Results: A total <strong>of</strong> 63 pts were enrolled;<br />

data from 59 pts were available. Median age was 67 (range, 36-75) years<br />

with 37 pts over 65 years. The majority <strong>of</strong> pts (64.4%) had 1 prior therapy;<br />

57 pts (96.6%) were previously treated with rituximab-containing combination<br />

chemotherapy and 8 (13.6%) had prior auto-PBSCT. Pts received a<br />

median <strong>of</strong> 4 (range, 1-6) treatment cycles. Sixteen (27.1%) pts completed<br />

6 treatment cycles; most common reasons for early discontinuation were<br />

disease progression (n�15) and failure to meet criteria to start the next<br />

cycle (n�13). Among 59 pts evaluable for response, ORR was 62.7% with<br />

a 37.3% CR rate. The median PFS was 200 days (95% CI, 109-410). Most<br />

common grade 3/4 adverse events (AEs) included CD4 lymphocytes<br />

decreased (66.1%), neutropenia (54.2%), and thrombocytopenia (10.2%).<br />

Four (6.8%) pts discontinued due to serious AEs (cytomegalovirus infection,<br />

infection, pneumonia, and pneumonia/respiratory failure). Conclusions:<br />

B-R demonstrated promising activity in pts with relapsed/refractory DL-<br />

BCL. Toxicity was primarily hematologic and generally manageable. These<br />

results suggest that B-R is a promising salvage regimen for pts with<br />

relapsed/refractory DLBCL after R-CHOP.<br />

Lymphoma and Plasma Cell Disorders<br />

515s<br />

8022 Poster Discussion Session (Board #2), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Role <strong>of</strong> radiotherapy for elderly DLBCL patients in the rituximab (R) era:<br />

Final results <strong>of</strong> the RICOVER-60-no-rx study <strong>of</strong> the DSHNHL. Presenting<br />

Author: Gerhard Held, Saarland University Hospital, Homburg, Germany<br />

Background: 117/306 (38%) <strong>of</strong> the total <strong>of</strong> 1,222 elderly (61-80 y) DLBCL<br />

pts. treated in RICOVER-60 with 6xR-CHOP-14�2R were assigned to<br />

receive additional radiotherapy (Rx) to bulky disease (Pfreundschuh et al.,<br />

Lancet Oncol. 2008). To study the relevance <strong>of</strong> Rx to bulky disease (Bx),<br />

166 pts. were prospectively treated without Rx in the R-CHOP-14-noRx<br />

amendment <strong>of</strong> RICOVER-60. Methods: The outcome <strong>of</strong> 166 R-CHOP-14noRx<br />

patients was compared with the 306 patients who had received<br />

6xR-CHOP-14�2R plus radiotherapy to Bx (�7.5 cm) in RICOVER-60.<br />

Methods: The outcome <strong>of</strong> 166 R-CHOP-14-noRx patients was compared<br />

with the 306 patients who had received 6xR-CHOP-14�2R plus radiotherapy<br />

to Bx (�7.5 cm) in RICOVER-60. Results: 164/166 R-CHOP-noRx<br />

patients are evaluable (median observation: 39 mos). Patients in R-CHOPnoRx<br />

were older (71 vs. 69 y.; median; p�0.018), more frequently in<br />

advanced stages (60% vs. 50%; p�0.037), and with extranodal involvement<br />

(63% vs. 53%; p�0.024), while Bx was more frequent in R-CHOP-<br />

14-Rx (38% vs. 29%; p�0.038). Overall response to therapy, EFS and OS<br />

were similar in the two studies adjusting for the prognostic imbalances<br />

between the cohorts. Patients with Bx who received received additional<br />

radiotherapy to Bx in R-CHOP-14-Rx had a better 3-year EFS (80% vs.<br />

54%; p�0.001), a better PFS (88% vs. 62%; p�0.001), and a better OS<br />

(90% vs. 65%; p�0.001) compared to R-CHOP-14-noRx. This was due<br />

the worse outcome <strong>of</strong> pts. with Bx in R-CHOP-14-noRx not achieving CR or<br />

CRu after 6xR-CHOP, since there was no difference in 3-year EFS in<br />

patients with Bx in CR or CRu after 6xR-CHOP-14 with and without<br />

additional radiotherapy (3-year EFS and PFS: 84% vs. 75%; p�0.430);<br />

OS 87% vs. 79%; p�0.839). Conclusions: In the R era, radiotherapy to<br />

bulky disease does not improve the outcome <strong>of</strong> elderly pts. in CR/CRu after<br />

completion <strong>of</strong> R-CHOP-14 immunochemotherapy, but appears to be<br />

beneficial for pts. with Bx not achieving CR/Cru. By restricting Bx<br />

radiotherapy to patients not achieving a CR/CRu, 43% <strong>of</strong> the patients with<br />

Bx could be spared radiotherapy. Supported by Deutsche Krebshilfe.<br />

8024 Poster Discussion Session (Board #4), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Evidence for rituximab (R) underdosing in subpopulations <strong>of</strong> elderly<br />

patients with DLBC: Results <strong>of</strong> the RICOVER-60 study <strong>of</strong> the DSHNHL.<br />

Presenting Author: Michael Pfreundschuh, Saarland University Hospital,<br />

Homburg, Germany<br />

Background: Gender and weight independently influence R clearance and R<br />

serum elimination half life (Mueller et al., Blood 2012). To investigate<br />

whether the differences in R pharmacokinetics translate into different<br />

outcomes, we analyzed elderly patients (pts) with different R pharmacokinetics<br />

treated in the RICOVER-60 study. Methods: 1222 elderly pts.<br />

(61-80 y) were randomized to receive 6 or 8 cycles <strong>of</strong> CHOP-14 with or<br />

without R given on days 1, 15, 29, 43, 57, 71, 85, and 99. For this study,<br />

subgroup analyses were performed for pts with faster R clearance: elderly<br />

male (vs. female) pts and elderly weighty (upper quartile: �77kg) vs. slim<br />

(lower quartile: �60 kg) female pts. Results: Elderly females had a slower R<br />

clearance (8.21 ml/h vs. 12.68 ml/h; p�0.003) and a prolonged R half life<br />

compared to men (t1/2�30.7 vs. t1/2�24.7 d; p�0.003). Female pts had<br />

a higher 3-year PFS (68% vs. 61%; p�0.062) and OS (74% vs. 68%<br />

p�0.086). The differences in outcome were due to a greater outcome<br />

improvement by the addition <strong>of</strong> R in females: the difference in 3-year PFS<br />

between female and male pts was 5.1% (p�0.448) in pts. receiving<br />

CHOP-14 only and 7.7% (p�0.053) when R was added. In a multivariate<br />

analysis the relative risk for progression in male compared to female<br />

patients was not significantly elevated after CHOP-14 (1.1; p�0.348), but<br />

was significantly higher after R-CHOP-14 (1.6; p�0.004). With respect to<br />

weight, addition <strong>of</strong> R resulted in a significantly improved 3-year PFS (74%<br />

vs. 49%; p�0.006) in female pts with a body weight within the lower<br />

quartile (�60 kg) who have an R serum half life <strong>of</strong> �38.1 days, while there<br />

was no improvement by the addition <strong>of</strong> R (72% vs. 71%; p�0.816) in<br />

female pts. with a body weight within the upper quartile (�77kg), who have<br />

a serum half life <strong>of</strong> �29.3 days. Conclusions: The reduced benefit <strong>of</strong> adding<br />

R to CHOP in elderly DLBCL pts. with a shorter rituximab serum half life<br />

(and hence lower serum levels) suggests that the respective subpopulations<br />

(males and weighty females) are underdosed when R is dosed based on<br />

body surface area at 375 mg/m2 . Ongoing studies <strong>of</strong> the DSHNHL<br />

investigate whether higher R doses for pts with a shorter R serum half life<br />

can improve the outcome <strong>of</strong> the respective pts.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


516s Lymphoma and Plasma Cell Disorders<br />

8025 Poster Discussion Session (Board #5), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Outcome <strong>of</strong> elderly DLBCL patients with 6xCHOP-14 and 8 rituximab (R)<br />

applications given over an extended period (SMARTE-R-CHOP-14 trial <strong>of</strong><br />

the DSHNHL). Presenting Author: Niels Murawski, Saarland University<br />

Hospital, Homburg, Germany<br />

Background: 6xCHOP-14 with 8xR given over an extended period improved<br />

3-year EFS (67% vs. 54%; p�0.030) and OS (80% vs. 67%, p�0.034) <strong>of</strong><br />

poor-prognosis patients (IPI�3-5) in the SMARTE-R trial compared to the<br />

RICOVER-60 trial where patients received 8xR every 2 weeks. Because we<br />

had recently shown (Mueller et al., Blood 2012) that elderly male patients<br />

have a faster R clearance (12.68 ml/h vs. 8.21 ml/h; p�0.003) and shorter<br />

serum elimination half life (t1/2�24.7 vs. t1/2�30.7 days; p�0.003)<br />

than females, we analyzed whether these differences translated into<br />

different outcomes by comparing the results achieved by elderly female and<br />

male patients in the RICOVER-60 and SMARTE-R trials. Methods: In<br />

SMARTE-R, 189 evaluable elderly (61-80 y) pts. with DLBCL received 6<br />

cycles <strong>of</strong> 2-weekly CHOP-14 combined with 8xR on days -4, -1, 10, 29,<br />

57, 99, 155, and 239. The primary endpoint was event-free survival (EFS).<br />

306 pts treated within the RICOVER-60 trial with 6xCHOP-14 � 8 R given<br />

on days 1, 15, 29, 43, 57, 71, 85 and 99 served as controls. Results: The<br />

3-year EFS <strong>of</strong> 51 poor-prognosis male patients in SMARTE-R was 67%<br />

compared to 47% <strong>of</strong> 66 poor-prognosis male patients treated in RI-<br />

COVER-60 (p�0.037); the respective figures were 71% vs. 53% (p�0.051)<br />

for PFS and 80% vs. 60% (p�0.027) for OS. In contrast, female poor-risk<br />

patients had only a small benefit from the extended rituximab exposure in<br />

SMARTE-R (n�48) compared to RICOVER-60 (n�57): 67% vs. 61%<br />

(p�0.354) for EFS; 71% vs. 67% (p�0.489) for PFS; and 80% vs. 76%<br />

(p�0.528) for OS. Conclusions: Elderly male patients with poor-prognosis<br />

DLBCL who have a faster R clearance and shorter R serum elimination half<br />

life than female patients, benefit significantly from the longer R exposure in<br />

SMARTE-R with a gain <strong>of</strong> 20% in 3-year OS, while the outcome <strong>of</strong> female<br />

patients was only slightly improved. Even though R maintenance has failed<br />

to demonstrate any benefit in the primary treatment <strong>of</strong> DLBCL to date,<br />

these results underline the importance <strong>of</strong> a minimum exposure time <strong>of</strong> R in<br />

order to exploit its full therapeutic potential in DLBCL. Supported by<br />

Deutsche Krebshilfe.<br />

8027 Poster Discussion Session (Board #7), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Retreatment with brentuximab vedotin in CD30-positive hematologic<br />

malignancies: A phase II study. Presenting Author: Nancy Bartlett, Washington<br />

University, Siteman Cancer Center, St. Louis, MO<br />

Background: Brentuximab vedotin comprises an anti-CD30 antibody conjugated<br />

by a protease-cleavable linker to a microtubule-disrupting agent,<br />

MMAE. In pivotal phase 2 studies in patients (pts) with relapsed/refractory<br />

Hodgkin lymphoma (HL) or systemic anaplastic large cell lymphoma<br />

(sALCL), objective response rates were 75% and 86% and median<br />

durations <strong>of</strong> response were 6.7 and 12.6 mo, respectively. A phase 2 study<br />

was initiated to investigate if pts who have previously responded to<br />

brentuximab vedotin could achieve another remission with retreatment<br />

(<strong>Clinical</strong>Trials.gov #NCT00947856). Methods: Pts had a CD30-positive<br />

hematologic malignancy, achieved an objective response (per Cheson<br />

2007) with prior brentuximab vedotin treatment, and experienced relapse<br />

after discontinuing treatment. Brentuximab vedotin was administered IV<br />

1.8 mg/kg every 21 days; antitumor activity was assessed by the investigator.<br />

Results: 14 HL pts and 8 sALCL (5 ALK-negative) pts were enrolled<br />

(median age 34 yr, range 16–72). Pts had received a median <strong>of</strong> 4 prior<br />

chemotherapy regimens (range 2–12). Median time since the previous<br />

brentuximab vedotin treatment was 6.9 mo (range 1–44). Median number<br />

<strong>of</strong> retreatment cycles was 7 (range 1� to 32�). Adverse events (AEs) in<br />

�25% <strong>of</strong> pts were nausea (41%), fatigue (36%), peripheral sensory<br />

neuropathy (36%), and diarrhea (27%). The most common Grade 3/4 AEs<br />

were anemia, fatigue, and hyperglycemia (3 pts each). Of the 11 pts who<br />

had pre-existing peripheral neuropathy, 3 (27%) had worsening with<br />

retreatment. Best clinical responses in pts with HL were 3 CR, 5 PR, 3 SD,<br />

3 PD. Among pts with sALCL, 5 achieved a CR, 1 had PD, and 2 were not<br />

yet evaluated. Of the 8 pts with CR in retreatment, previous best responses<br />

to brentuximab vedotin treatment were 4 PR and 4 CR. Median duration <strong>of</strong><br />

retreatment response was 10.8 mo (range 0� to 10.8), and in pts who<br />

achieved CR, the median duration <strong>of</strong> response was not reached (range 0�<br />

to 10.5 mo); 11 pts remain on retreatment. Conclusions: Retreatment with<br />

brentuximab vedotin was generally well tolerated. Objective responses were<br />

observed (13 <strong>of</strong> 20; 65%) in this heavily pretreated population. Enrollment<br />

to the phase 2 retreatment study is ongoing.<br />

8026 Poster Discussion Session (Board #6), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Multicenter, phase II study <strong>of</strong> bendamustine in refractory or relapsed T-cell<br />

lymphoma: The BENTLY trial. Presenting Author: Remy Gressin, University<br />

Hospital Grenoble, Grenoble, France<br />

Background: T-cell lymphomas have a poor prognosis with few options <strong>of</strong><br />

effective treatment. This study determined the efficacy and safety <strong>of</strong><br />

bendamustine as a single agent in the treatment <strong>of</strong> refractory or relapsed<br />

T-cell lymphomas. Methods: Patients with histologically confirmed peripheral<br />

T-cell lymphoma (PTCL) or cutaneous T-cell lymphoma (CTCL), who<br />

had previously received at least one line <strong>of</strong> chemotherapy were selected.<br />

Bendamustine was administered IV at the dosage <strong>of</strong> 120 mg/m2 on days 1<br />

and 2 every 3 weeks, for 6 cycles. Treatment response was assessed using<br />

the IWC for non-Hodgkin’s lymphoma. The primary end point was overall<br />

response rate (ORR). Secondary end points were duration <strong>of</strong> response<br />

(DoR), progression-free survival (PFS), and overall survival (OS),<br />

NCT00959686. Results: Twenty two female and 38 male were included.<br />

The median age was 66 years with more 1/4 <strong>of</strong> them � 75. Histology was<br />

predominantly angio-immunoblastic lymphadenopathy (n�32) and PTCLnos<br />

(n�23). The median previous line <strong>of</strong> chemotherapy was 1 (1-3). Nearly<br />

one half (45%) <strong>of</strong> the patients was refractory to the last previous<br />

chemotherapy and the median duration <strong>of</strong> the best previous response was<br />

6.6 (1.5-67) months. The disease was disseminated in the majority <strong>of</strong> case<br />

(87%) and the international prognostic index (IPI) was high (3–5) in 68%<br />

<strong>of</strong> the patients. Twenty patients (33%) received less than 3 cycles <strong>of</strong><br />

bendamustine. The major reason for early discontinuation was disease<br />

progression. In the Intent-To-Treat (ITT) population, the best ORR was<br />

50%, including complete response (CR) in 28% and partial response (PR)<br />

in 22 %. Bendamustine showed a consistency in the efficacy as a function<br />

<strong>of</strong> major disease characteristics. The median values for DoR, PFS and OS<br />

were 3.5, 4 and 6 months respectively. The most frequent grade 3/4 AEs<br />

were neutropenia (30%), thrombocytopenia (24%) and infections (20%).<br />

Conclusions: Bendamustine is active in high risk refractory and relapsed<br />

T-cell lymphoma with manageable toxicity.<br />

8028 Poster Discussion Session (Board #8), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Everolimus (EVE) for relapsed/refractory classical Hodgkin lymphoma<br />

(cHL): Open-label, single-arm, phase II study. Presenting Author: Patrick<br />

B. Johnston, Mayo Clinic, Rochester, MN<br />

Background: Effective treatment for relapsed/refractory cHL is lacking. The<br />

oral mammalian target <strong>of</strong> rapamycin inhibitor EVE showed promising<br />

efficacy and acceptable toxicity in cHL. We conducted a study to confirm<br />

EVE safety and efficacy in relapsed/refractory cHL. Methods: In this<br />

multicenter, open-label, 2-step, phase 2 study, adults with cHL that<br />

progressed after high-dose chemotherapy with autologous hematopoietic<br />

stem cell transplant (AHSCT) or a gemcitabine-, vinorelbine-, or vinblastinecontaining<br />

regimen received EVE 10 mg/d until disease progression or<br />

unacceptable toxicity. Response was assessed every 12 wk via integrated<br />

positron emission tomography/computed tomography (CT) with contrast or<br />

CT with contrast. Primary endpoint was overall response rate (ORR) per<br />

modified response criteria for malignant lymphoma. Adverse events (AEs)<br />

were assessed during, and for �4 wk after, EVE treatment. Results: 55<br />

patients were enrolled. Of the 38 currently evaluable patients, 37% were<br />

men, median age was 32.5 y, 53% were pretreated with AHSCT, and 95%<br />

were pretreated with gemcitabine, vinorelbine, or vinblastine; 71% had<br />

disease progression during previous therapies or discontinued previous<br />

treatment due to progression. 23 patients discontinued treatment, most<br />

commonly due to disease progression (n � 11). ORR was 37% (Table).<br />

Median progression-free survival (PFS) was 7.2 mo. The most common<br />

hematologic AEs were thrombocytopenia (39%) and anemia (24%); the<br />

most common nonhematologic AEs were fatigue (47%), cough (29%),<br />

dyspnea (26%), headache (21%), and rash (21%). Grade 3/4 AEs, most<br />

commonly thrombocytopenia (18%), were observed in 45% <strong>of</strong> patients.<br />

Conclusions: EVE showed a favorable ORR and median PFS in the first 38<br />

evaluable patients with highly pretreated, relapsed/refractory cHL. The AE<br />

pr<strong>of</strong>ile was consistent with that previously observed for EVE. These<br />

preliminary results confirm those <strong>of</strong> an earlier study and support further<br />

evaluation <strong>of</strong> EVE in cHL.<br />

Best overall response, n (%)<br />

EVE 10 mg/d<br />

N�38<br />

ORR 14 (36.8)<br />

Complete response* 1 (2.6)<br />

<strong>Part</strong>ial response 13 (34.2)<br />

Stable disease 10 (26.3)<br />

Progressive disease 5 (13.2)<br />

Unknown<br />

*Defined as resolution <strong>of</strong> all adenopathy.<br />

9 (23.7)<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


8029 Poster Discussion Session (Board #9), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

A multicenter phase II study <strong>of</strong> vorinostat in patients (pts) with relapsed or<br />

refractory indolent B-cell non-Hodgkin lymphoma (B-NHL) or mantle cell<br />

lymphoma (MCL). Presenting Author: Kiyoshi Ando, Department <strong>of</strong> Hematology<br />

and Oncology, Tokai University, Kanagawa, Japan<br />

Background: Most pts with indolent B-NHL relapse even after rituximabcontaining<br />

chemotherapy. Vorinostat is an orally active histone deacetylase<br />

(HDAC) inhibitor, and is under investigation as monotherapy or in combination<br />

regimens in various hematologic malignancies. It has been reported<br />

that the histone acetyltransferase genes (EP300 and CREBBP) are frequently<br />

mutated in NHL, suggesting the potential usefulness <strong>of</strong> HDAC<br />

inhibitors in its treatment. Methods: We conducted a phase 2 study to<br />

investigate the efficacy, safety and tolerability <strong>of</strong> vorinostat in pts with<br />

indolent B-NHL or MCL with the overall response rate (ORR) against<br />

follicular lymphoma (FL) as the primary endpoint. The primary hypothesis<br />

was to demonstrate the efficacy in FL pts as measured by the ORR.<br />

Assuming the true ORR with vorinostat <strong>of</strong> 50% and without vorinostat <strong>of</strong><br />

less than 25%, 39 pts will provide 90% power to demonstrate an<br />

ORR�25%. Vorinostat was administered at the dose <strong>of</strong> 200 mg BID for 14<br />

consecutive days in a 21-day cycle. The ORR and PFS were determined by<br />

an independent review panel in the Full Analysis Set (FAS) population.<br />

Results: A total <strong>of</strong> 56 pts with a median age <strong>of</strong> 60 years (range: 33 - 75)<br />

were enrolled, and 50 <strong>of</strong> them (39 with FL, 11 with other subtypes <strong>of</strong><br />

indolent B-NHL or MCL) were included in the FAS population. The number<br />

<strong>of</strong> prior therapeutic regimens was 2 (range: 1 - 4) and 41 pts have<br />

previously received rituximab-based regimens.. The percentages <strong>of</strong> pts with<br />

FLIPI at low risk, intermediate risk and high risk were 41%, 39%, and<br />

21%, respectively. The ORR in 39 pts with FL was 49% (95% CI: 32, 65),<br />

and the pre-specified statistical success criterion was met. The median<br />

PFS in 39 pts with FL was 17.5 months (range: 2, 22�), especially the<br />

median PFS in 19 responders with FL has not yet been reached. The major<br />

toxicities were hematologic and gastrointestinal ones, which were reversible<br />

and manageable. Grade 3/4 infection was observed in 2%. Conclusions:<br />

This phase 2 study demonstrated that oral vorinostat is an agent with<br />

sustained antitumor activity in pts with relapsed or refractory indolent<br />

B-NHL, with acceptable safety pr<strong>of</strong>iles, warranting further investigations.<br />

8031 Poster Discussion Session (Board #11), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

CNS-directed chemotherapy including high-dose chemotherapy with autologous<br />

stem cell transplantation (HD-ASCT) for CNS relapse <strong>of</strong> aggressive<br />

lymphomas: Final analysis <strong>of</strong> a phase II study. Presenting Author: Agnieszka<br />

Korfel, Department <strong>of</strong> Hematology and Oncology, Charite Campus<br />

Benjamin Franklin, Berlin, Germany<br />

Background: The outcome <strong>of</strong> patients with CNS relapse <strong>of</strong> aggressive<br />

lymphoma (secondary CNS lymphoma, SCNSL) is poor with no standard<br />

therapy established thus far. Here we present the final analysis <strong>of</strong> a<br />

prospective multicenter phase II study using an intensive induction<br />

regimen followed by high-dose chemotherapy and autologous stem-cell<br />

transplantation. Methods: Adult immunocompetent patients �65 years<br />

with SCNSL were eligible. Induction chemotherapy consisted <strong>of</strong> two cycles<br />

MTX/IFO (methotrexate 4g/m2 iv. d1, ifosfamide 2g/m2 iv. d3-5 and i.th.<br />

liposomal cytarabine 50mg d6) and one cycle AraC/TT (cytarabine 3g/m2 d1-2, thiotepa 40mg/m2 iv. d2 and i.th. liposomal cytarabine 50mg d3).<br />

Then, patients without progression received high-dose chemotherapy with<br />

carmustine 400mg/m2 iv. d -5, thiotepa 2x5mg/kg iv. d -4 to -3 and<br />

etoposide 150mg/m2 iv. d -5 to -3 followed by ASCT d0. Results: Thirty<br />

eligible patients (median age 58 years) were enrolled. Three patients had<br />

T-cell and 27 aggressive B-cell lymphoma. Pre-treatment was CHOP-like in<br />

29 patients, including rituximab in 26. CNS relapse occurred after a<br />

median <strong>of</strong> 8.5 (3-80) months and was intracerebral in 23 and meningeal<br />

in13 patients (combined in 7); 6 had concomitant systemic lymphoma.<br />

After induction therapy CNS response was found in 22 (73%) patients<br />

(8xCR, 14xPR), 3 patients had SD, 4 patients PD and 1 patient no<br />

response evaluation. HD-ASCT was performed in 24 patients; resulting in<br />

15 CR (63%), 2 PR (8%) and 7 PD (29%). Myelotoxicity was the most<br />

frequent WHO grade 3-4 adverse event with infections in 8/30 pts on<br />

MTX/IFO (27%), 5/23 (22%) on AraC/TT and 11/20(55%) on HD-ASCT.<br />

One patient died due to septic diverticulitis and one developed persisting<br />

fecal incontinence. The median follow up was 21 months. The median PFS<br />

was 12.1 months (95% CI 6.4-17.7) in all patients and 30.4 (95%CI<br />

2.5-58.3) months after HD-ASCT, the median overall survival was 27.4<br />

months and not reached, respectively. Conclusions: This first prospective<br />

study on SCNSL demonstrates that lasting remissions can be achieved with<br />

CNS-directed HD-ASCT in a large proportion <strong>of</strong> patients and probably cure<br />

in some.<br />

Lymphoma and Plasma Cell Disorders<br />

517s<br />

8030 Poster Discussion Session (Board #10), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Interim analysis <strong>of</strong> the largest prospective trial to date in adult CD20positive<br />

post-transplant lymphoproliferative disorder (PTLD): Introducing<br />

risk-stratified sequential treatment (RSST). Presenting Author: Ralf Ulrich<br />

Trappe, Department <strong>of</strong> Internal Medicine II: Hematology and Oncology,<br />

University Medical Center Schleswig-Holstein, Campus Kiel, Kiel, Germany<br />

Background: The prospective, multicenter international phase II PTLD-1<br />

trial <strong>of</strong> sequential treatment (ST, 4 cycles <strong>of</strong> weekly rituximab followed by 4<br />

cycles <strong>of</strong> CHOP-21 � G-CSF) in adult CD20-positive PTLD demonstrated<br />

excellent efficacy (90% overall response rate, ORR) and safety (11%<br />

treatment-related mortality, TRM). As the response to rituximab predicted<br />

overall survival (OS), the trial was amended in 2007 introducing riskstratified<br />

sequential treatment (RSST) according to the response to<br />

rituximab (NCT00590447). Methods: Following rituximab on days 1, 8, 15<br />

and 22, RSST consisted <strong>of</strong> 4 3-weekly courses <strong>of</strong> rituximab monotherapy<br />

for patients (pts) in complete remission (CR, low risk) while all others (high<br />

risk) received 4 cycles <strong>of</strong> R-CHOP-21 � G-CSF. Key exclusion criteria were<br />

CNS involvement, HIV infection, severe organ dysfunction not related to<br />

PTLD, and ECOG � 2. Primary endpoint was ORR. This is an analysis <strong>of</strong> the<br />

first 91 patients treated with RSST. Results: 79/91 pts had monomorphic,<br />

12 polymorphic PTLD. 41/91 pts were kidney, 27 liver, 12 heart, 7 lung or<br />

heart�lung, 3 heart�kidney and 1 kidney�pancreas transplant recipients.<br />

Median age at diagnosis was 60 years (range 20-82). 73/91 pts had late<br />

PTLD and 39/85 PTLDs were EBV-associated. 1 pt died before initiation <strong>of</strong><br />

treatment; 5 pts discontinued treatment after 4 cycles rituximab. TRM <strong>of</strong><br />

RSST was 7/90 (8%) including 5 deaths with unknown remission status.<br />

ORR was thus 74/80 (93%, 95%CI: 84-97%; CR: 62/80 [78%]). 24/90<br />

pts (27%) achieved CR with 4 cycles <strong>of</strong> rituximab. After a median follow up<br />

<strong>of</strong> �3 years, relapse rate in low risk pts was not increased by rituximab<br />

consolidation in RSST compared to CHOP consolidation in ST (3/23 vs.<br />

5/14, p�0.104]). In patients in PD after rituximab, R-CHOP was more<br />

effective than CHOP in achieving CR (15/23 vs. 3/11, p�0.038). OS at 3<br />

years was higher with RSST (70%, 95% CI: 60-82%) compared to ST<br />

(61%, 95%CI 49-72%) but this difference was not significant. Conclusions:<br />

With RSST 27% <strong>of</strong> pts were classified as low risk and achieved durable<br />

tumor control without chemotherapy while R-CHOP seems more efficient<br />

than CHOP in in high risk patients.<br />

8032 Poster Discussion Session (Board #12), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Phase Ib trial <strong>of</strong> AVL-292, a covalent inhibitor <strong>of</strong> Bruton’s tyrosine kinase<br />

(Btk), in chronic lymphocytic leukemia (CLL) and B-non-Hodgkin lymphoma<br />

(B-NHL). Presenting Author: Jennifer R. Brown, Dana-Farber<br />

Cancer Institute, Boston, MA<br />

Background: Btk supports activation, survival, and proliferation <strong>of</strong> malignant<br />

B cells in CLL and B-NHL. AVL-292 is an oral, potent (IC50 � 0.5nM),<br />

selective small molecule covalent inhibitor <strong>of</strong> Btk. Methods: Patients (pts)<br />

with previously treated CLL or B-NHL were administered AVL-292 in<br />

escalating cohorts <strong>of</strong> 125, 250, or 400 mg po QD using a 3�3 design in<br />

continuous 28 day cycles until progressive disease (PD) or toxicity. Key<br />

objectives were safety, DLT, MTD, PK, and Btk occupancy. Plasma<br />

AVL-292 levels were assessed by LC-MS-MS. Btk occupancy by AVL-292<br />

was assessed by covalent probe assay in peripheral blood mononuclear<br />

cells. Results: 12 pts have enrolled (3 each at 125 and 250 mg and 6 at<br />

400 mg: 5M/7F; median age 68 years, range 45-79; median 2.5 prior<br />

therapies, range 1-10) including 8 CLL (2 with 17p-, 2 with 11q22-, 2 with<br />

both 17p-/11q22-; 2 mutated IGHV, 5 unmutated IGHV, 1 missing) and 4<br />

B-NHL (1 diffuse large B cell lymphoma (DLBCL); 1 follicular (FL); 2<br />

marginal zone (MZL)). Median time on treatment is 65 days, range 28-158.<br />

Ten <strong>of</strong> 12 pts continue on treatment: 1 pt (DLBCL) discontinued for PD<br />

after 1 cycle and 1 pt (FL) for DLT (Gr 4 plts; 400 mg QD). No other DLTs or<br />

grade 4 adverse events (AEs) have occurred and MTD has not been reached.<br />

Most frequent AEs reported include transient diarrhea, rash/skin infection,<br />

URI, nausea, and fatigue. Gr 3 AEs include 1 atrial fibrillation (not drug<br />

related) and 1 ANC low (probably drug related). To date, 8 <strong>of</strong> 8 CLL patients<br />

have stable disease (SD) with median 28% decrease from baseline lymph<br />

node measurement (range 3-40% decrease). Six <strong>of</strong> 8 pts with CLL<br />

experienced increased ALC in cycle 1: median increase 89.5% (range<br />

50-212%). Two <strong>of</strong> 4 NHL pts have SD (both MZL), 1 PD (DLBCL), and 1<br />

not evaluable due to DLT (FL). Full Btk occupancy was achieved with dose<br />

levels � 250 mg. Multiple dose PK exposure (AUClast) was linear with no<br />

accumulation from Day 1 to 15. Conclusions: AVL-292 was well tolerated<br />

from 125-400 mg po QD and early efficacy analysis in CLL and B-NHL<br />

shows 10/11 efficacy evaluable pts with SD. Full Btk occupancy was<br />

achieved with � 250 mg QD and PK was predictable with no accumulation.<br />

Dose escalation is ongoing and MTD cohort expansion is planned.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


518s Lymphoma and Plasma Cell Disorders<br />

8033 Poster Discussion Session (Board #13), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Oral weekly MLN9708, an investigational proteasome inhibitor, in combination<br />

with lenalidomide and dexamethasone in patients (pts) with previously<br />

untreated multiple myeloma (MM): A phase I/II study. Presenting<br />

Author: Paul Gerard Guy Richardson, Dana-Farber Cancer Institute, Boston,<br />

MA<br />

Background: MLN9708 is an oral, reversible 20S proteasome inhibitor. The<br />

feasibility <strong>of</strong> combining a proteasome inhibitor with an immunomodulatory<br />

drug and a steroid in previously untreated MM has been demonstrated with<br />

the RVD regimen. This is the first study <strong>of</strong> MLN9708 in combination with<br />

lenalidomide and dexamethasone (NCT01217957). Here we report the<br />

phase (Ph) 1 MTD and preliminary Ph 2 results. Methods: Pts with<br />

previously untreated MM, aged �18 yrs with measurable disease received<br />

oral MLN9708 (phase 1: 1.68–3.95 mg/m2 ) days 1, 8, and 15, lenalidomide<br />

25 mg days 1–21, and dexamethasone 40 mg days 1, 8, 15, and 22,<br />

for up to twelve 28-day cycles. Primary objectives were determination <strong>of</strong><br />

safety, MTD, and recommended phase 2 dose (RP2D) (Ph 1), and<br />

CR�VGPR rate (Ph 2). Results: At data cut-<strong>of</strong>f (Dec 1, 2011), 29 pts had<br />

been enrolled (15 Ph 1, 14 Ph 2). Median age was 64 yrs (range 40–82);<br />

69% ISS stage II/III. In Ph 1, the MLN9708 MTD was determined as 2.97<br />

mg/m2 and the RP2D as 2.23 mg/m2 ; for Ph 2, the RP2D was converted to<br />

a 4.0 mg fixed dose based on population PK results. Ph 1 pts have received<br />

a median <strong>of</strong> 6 treatment cycles (range 1–11), 8 received �6 cycles; 6<br />

stopped to receive ASCT, 7 are ongoing. Ph 2 pts received a median <strong>of</strong> 1<br />

(range 1–2), all are ongoing. Grade �3 hematologic toxicity was reversible<br />

and included anemia (n�2) and thrombocytopenia (n�1). Grade �3<br />

nonhematologic toxicity included erythematous rash, syncope, and vomiting<br />

(2 pts each). All-grade drug-related peripheral neuropathy was seen in 6<br />

pts (21%), including grade 2 with pain in 2 (both Ph 1 at doses above the<br />

MTD). Two pts discontinued due to AE; there were 5 pts who had serious<br />

drug-related AE (all Ph 1). Of 19 response-evaluable pts (Ph 1 � Ph 2), all<br />

achieved �PR, including 5 CR (1 sCR), 4 VGPR, and 10 PR; all remain in<br />

response with duration <strong>of</strong> confirmed response <strong>of</strong> up to 9.5 months. Of 4<br />

response-evaluable Ph 2 pts, 1 has achieved VGPR and 3 PR to date.<br />

Conclusions: Oral MLN9708 plus lenalidomide and dexamethasone appears<br />

well tolerated with manageable toxicity. These data show antitumor<br />

activity at the RP2D in pts with previously untreated MM, with �PR in all<br />

pts to date.<br />

8035 Poster Discussion Session (Board #15), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Response rates to single-agent carfilzomib in patients refractory or intolerant<br />

to both bortezomib and immunomodulators in trial PX-171-003-A1.<br />

Presenting Author: David Samuel DiCapua Siegel, John Theurer Cancer<br />

Center, Hackensack, NJ<br />

Background: Patients (pts) with relapsed and refractory multiple myeloma<br />

(RR MM) who are refractory or intolerant to both bortezomib (BTZ) and<br />

immunomodulators (IMiDs; thalidomide [Thal] or lenalidomide [Len]) (ie,<br />

“double-refractory/intolerant”), have few therapeutic options and a poor<br />

prognosis. Carfilzomib (CFZ), a next generation proteasome inhibitor (PI),<br />

has shown durable single-agent activity in clinical studies including<br />

003-A1, an open-label, single-arm phase 2b trial in RR MM. The present<br />

analysis describes clinical activity in pts from 003-A1 with double-refractory/<br />

intolerant disease and in other groups <strong>of</strong> clinical interest including pts with<br />

disease refractory to all 5 approved classes <strong>of</strong> anti-MM tx (alkylators,<br />

anthracyclines, corticosteroids, IMiDs, and PIs) in clinical use (“refractory<br />

to all approved tx”). Methods: Pts from 003-A1 with double-refractory/<br />

intolerant disease were analyzed, as were pts with disease refractory to all<br />

approved tx. CFZ was given on days 1, 2, 8, 9, 15, 16 <strong>of</strong> 28-day cycles (C),<br />

(20 mg/m2 in C1; 27 mg/m2 in C2–12). Primary endpoint was overall<br />

response rate (ORR). Secondary endpoints included duration <strong>of</strong> response<br />

(DOR), overall survival, and safety. Results: The study ORR was 22.9% with<br />

median DOR <strong>of</strong> 7.8 mo (N�266). 228 pts (86%) with double-refractory/<br />

intolerant disease had ORRs <strong>of</strong> 20.6% and a median DOR <strong>of</strong> 7.4 mo.<br />

Conclusions: Single-agent CFZ demonstrated clinically meaningful, durable<br />

responses in pts with double-refractory/intolerant MM or disease refractory<br />

to all 5 approved classes <strong>of</strong> tx. The ORRs across groups <strong>of</strong> clinical interest<br />

were generally consistent with results for the entire study population. These<br />

results are notable for a next-generation PI and demonstrate the activity <strong>of</strong><br />

single-agent CFZ in pts with advanced stage MM.<br />

Refractory status n ORR (>PR), % DOR, mo<br />

Refractory/intolerant to BTZ and (>1 IMiD) 228 20.6 7.4<br />

To BTZ in any prior regimen 194 16.5 7.8<br />

To Thal in any prior regimen 118 17.8 5.6<br />

To Len in any prior regimen 221 22.2 7.8<br />

Refractory to all approved tx 44 20.5 7.8<br />

8034 Poster Discussion Session (Board #14), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Weekly dosing <strong>of</strong> the investigational oral proteasome inhibitor MLN9708 in<br />

patients (pts) with relapsed/refractory multiple myeloma (MM): A phase I<br />

study. Presenting Author: Shaji Kumar, Mayo Clinic, Rochester, MN<br />

Background: Phase 1 studies are evaluating IV and oral dosing <strong>of</strong> the<br />

reversible proteasome inhibitor MLN9708 in multiple tumor types. We<br />

report the safety, MTD, pharmacokinetics (PK), pharmacodynamics, and<br />

preliminary responses with weekly oral MLN9708 in pts with relapsed/<br />

refractory MM (NCT00963820). Methods: Pts aged �18 yrs received<br />

MLN9708 on d 1, 8, and 15 <strong>of</strong> 28-d cycles. In the dose-escalation phase,<br />

pts required �2 prior therapies (including bortezomib, thalidomide/<br />

lenalidomide, and corticosteroids). At the MTD, pts were to be enrolled to<br />

relapsed and refractory (RR), bortezomib-relapsed (VR), proteasome inhibitor<br />

(PI) naïve, and carfilzomib (CZ) expansion cohorts. Results: 36 pts have<br />

been enrolled to date (data cut-<strong>of</strong>f: Dec 1, 2011), 32 in the dose-escalation<br />

phase (0.24–3.95 mg/m2 ) and 8 to expansion cohorts (2 RR, 5 VR, 1 PI<br />

naïve; RR and VR cohorts each include 2 pts from MTD dose-escalation<br />

cohort). Median age was 64.5 yrs (range 40–79), 53% were male, and<br />

median number <strong>of</strong> prior lines <strong>of</strong> therapy was 3.5 (range 1-13), including<br />

92%, 92%, 56%, and 8% who had prior bortezomib, lenalidomide,<br />

thalidomide, and carfilzomib, respectively. Pts have received a median <strong>of</strong> 2<br />

cycles (range 1–11); 5 pts remain on treatment. Among 24 DLT-evaluable<br />

pts, 3 DLTs were seen: 2 at 3.95 mg/m2 (1 grade 3 rash, 1 grade 3 GI AEs)<br />

and 1 at 2.97 mg/m2 (grade 3 GI AEs). The MTD was determined as 2.97<br />

mg/m2 . Overall, 69% <strong>of</strong> pts had drug-related AEs, and 28% had related<br />

grade �3 AEs, including thrombocytopenia (17%), diarrhea (11%),<br />

nausea, neutropenia, and fatigue (each 8%). Only 3 (8%) pts had<br />

drug-related peripheral neuropathy (PN; no grade �3). 2 pts discontinued<br />

due to AEs. In 18 response-evaluable pts, 1 had a VGPR at 3.95 mg/m2 ,1<br />

had a PR at 2.97 mg/m2 , and 8 have achieved SD durable for up to 9.5<br />

mos. PK analyses showed linear plasma PK (0.8–3.95 mg/m2 ), Tmax <strong>of</strong><br />

0.5-2 hr, and terminal half-life <strong>of</strong> 7 d for MLN2238 (biologically active<br />

hydrolysis product). There was a trend for a dose-dependent increase in<br />

whole blood 20S proteasome inhibition. Conclusions: Current data suggest<br />

weekly oral MLN9708 is generally well tolerated with infrequent PN, and<br />

shows early signs <strong>of</strong> antitumor activity.<br />

8036 Poster Discussion Session (Board #16), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Clarithromycin, pomalidomide, and dexamethasone (ClaPD) in relapsed or<br />

refractory multiple myeloma. Presenting Author: Adriana C. Rossi, Weill<br />

Cornell Medical College, New York Presbyterian Hospital, New York, NY<br />

Background: Clarithromycin has been shown to enhance anti-myeloma<br />

activity <strong>of</strong> lenalidomide�dexamethasone in the upfront treatment <strong>of</strong><br />

multiple myeloma (MM). Pomalidomide is an immunomodulatory agent<br />

effective in relapsed/refractory MM (RRMM). We hypothesized that clarithromycin<br />

may similarly enhance pomalidomide � dexamethasone in RRMM.<br />

We now report updated results from a phase 2 trial <strong>of</strong> ClaPD in RRMM.<br />

Methods: 73 patients with RRMM were enrolled in a single-institution<br />

phase 2 study <strong>of</strong> ClaPD. All subjects had � 3 prior lines <strong>of</strong> therapy, one <strong>of</strong><br />

which must have included lenalidomide. ClaPD is clarithromycin 500mg<br />

twice daily; dexamethasone 40mg weekly; and pomalidomide 4mg for days<br />

1-21 <strong>of</strong> a 28-day cycle. All patients had VTE prophylaxis with aspirin.<br />

Monthly disease response evaluation included immunoelectrophoresis and<br />

free light chain analysis; bone marrow biopsy with skeletal imaging was<br />

used to confirm MM responses. Treatment continued as tolerated until<br />

disease progression. Results: The 66 patients who completed � 1 cycle <strong>of</strong><br />

ClaPD are reported. Median number <strong>of</strong> cycles was 6 (range 1-17).<br />

Responses were progressive disease: 10%, stable disease: 21%, minimal<br />

response: 12%, partial response: 33%, very good partial response: 18%,<br />

stringent complete remission: 5%, for an overall response rate (ORR) <strong>of</strong><br />

56% and �VGPR rate <strong>of</strong> 23%. Median time to PR was 1.25 cycles (range<br />

1-8). Median PFS was 5 months. Response and PFS were not different in<br />

patients refractory to lenalidomide (85%), bortezomib (82%), or doublerefractory<br />

patients (76%). After a median follow up <strong>of</strong> 12 months,28 pts<br />

(42%) remain on study without progression and 56pts (85%) are alive. Two<br />

pts withdrew due to toxicity (1 Grade 3 fatigue, 1 Grade 4 muscular<br />

weakness). One patient withdrew consent. Conclusions: ClaPD is highly<br />

effective for heavily pre-treated RRMM, particularly in lenalidomiderefractory<br />

disesase and compares favorably to previously published Phase 2<br />

data <strong>of</strong> Pom/Dex (ORR 56% vs 40% - Lacy et. al JCO 2009) without excess<br />

toxicity. Response to ClaPD is rapid, well tolerated, and sustained over 7<br />

months in most subjects. These data support the clinical efficacy <strong>of</strong><br />

pomalidomide based regimens in RRMM.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


8037 Poster Discussion Session (Board #17), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Connect MM: The multiple myeloma (MM) disease registry—Incidence <strong>of</strong><br />

second primary malignancies (SPM). Presenting Author: Robert M. Rifkin,<br />

US Oncology Research, Denver, CO<br />

Background: Advances in the treatment <strong>of</strong> MM have greatly improved<br />

clinical outcomes for patients (pts). SPM occurrence has been observed in<br />

early and late stage MM as well as with increasing age. US SEER Cancer<br />

Registry reports a background incidence rate <strong>of</strong> SPM 2.1/100 person-yrs<br />

(PY) among persons � 65 yrs <strong>of</strong> age. However, incidence <strong>of</strong> SPM in MM pts<br />

and the relationship to therapy still warrants further exploration. Methods:<br />

Connect MM is a US-based observational registry designed to characterize<br />

pts with newly diagnosed MM from 266 US sites. Initiated in Sep 2009,<br />

patient data were collected at baseline and each subsequent quarter with a<br />

standardized form. On Dec 14, 2011, Connect MM reached full enrollment<br />

at 1,500 pts. Results: As <strong>of</strong> Jan 13, 2012, preliminary retrospective SPM<br />

data is available for 1015 pts. Median age was 67 yrs, 56.8% male, and<br />

median follow-up was 10.6 mo (0.03-24.9 mo). 12 pts (7 male) with<br />

median age <strong>of</strong> 68.5 yrs developed SPM. Median time to SPM was 8.5 mo<br />

(0.8-17.7 mo) after treatment initiation. 11 invasive SPM were observed -<br />

4 hematological (heme): 1 AML, 1 MDS, 1 CMML and 1 DLBCL, and 7<br />

solid: 2 melanoma skin, 1 bronchus, 1 breast, 1 prostate, 1 tonsil, and 1<br />

gastric carcinoid. Also, 1 pt had non-melanoma skin cancer (NMSC), and 1<br />

pt with invasive skin cancer also had NMSC. Of the 4 pts developing heme<br />

SPM, 3 had bortezomib (BORT), 1 had thalidomide (THAL), 1 had<br />

lenalidomide (LEN), 2 had melphalan (MEL), and all 4 pts had steroids. Of<br />

the 7 pts who developed solid tumor SPM, 5 had BORT, 5 had LEN, 2 had<br />

doxorubicin, 1 had MEL, 5 had steroids, 1 had irradiation, and 1 had not<br />

received MM treatment. 2 pts with prior history <strong>of</strong> invasive malignancy<br />

developed solid tumor SPM (1 pt also developed NMSC).1 pt who received<br />

irradiation developed NMSC. Early overall incidence <strong>of</strong> invasive SPM was<br />

1.21/100 PY (95% CI 0.60, 2.16) for all pts and 1.20/100 PY (95% CI<br />

0.44,2.62) for pts � 65 yrs <strong>of</strong> age. Conclusions: This preliminary analysis<br />

shows that SPM occurred at an expected rate in this disease specific<br />

registry <strong>of</strong> patients with NDMM and appeared to occur irrespective <strong>of</strong> MM<br />

treatment administered. Incidence rates <strong>of</strong> SPM may increase over time as<br />

patients receive transplantation and alkylators. Prospective observation will<br />

continue.<br />

8039 Poster Discussion Session (Board #19), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Second cancer risk 40 years after cure for Hodgkin lymphoma. Presenting<br />

Author: Michael Schaapveld, The Netherlands Cancer Institute, Antoni van<br />

Leeuwenhoek Hospital, Amsterdam, Netherlands<br />

Background: During the last decades Hodgkin Lymphoma (HL) treatment<br />

changed towards less toxic chemotherapy schemes and smaller radiation<br />

fields. The impact <strong>of</strong> these changes on second cancer (SC) risk is still<br />

unknown. Methods: We calculated standardized incidence ratios (SIR),<br />

comparing SC risk after HL treatment with expected risk, based on cancer<br />

incidence in the general population, and compared SC risk between<br />

treatment modalities, accounting for competing events, in a large Dutch<br />

cohort comprising 3,390 5-years HL survivors, aged 15-51 years at HL<br />

treatment and diagnosed between 1965-2000. Results: The median<br />

follow-up was 18.2 years; 23% <strong>of</strong> the patients was followed �25 years.<br />

During follow-up 734 SCs and 92 third cancers (TC) occurred. The SIR for<br />

any SC was 4.5 (95% confidence interval (95%CI) 4.1-4.9). SC risk was<br />

still elevated after 35 years <strong>of</strong> follow-up (SIR 3.9; 95%CI 2.5-5.8) and<br />

cumulative incidence (CI) reached 47.1% (95%CI 43.6-50.5) at 40 years<br />

follow-up. For TCs the SIR was 5.5 (95%CI 4.4-6.9); the 20-year CI was<br />

22.3% (95%CI 17.8-27.2). Risks <strong>of</strong> NHL and leukemia strongly decreased<br />

in more recent treatment periods (P-trend �0.001). The CI <strong>of</strong> solid tumors<br />

(ST) between 5-19 years after HL treatment did not differ for patients<br />

treated between 1965-1979, 1980-1989 or 1990-2000 (P�0.21; 19year<br />

CI 9.1%, 11.6% and 11.4%, respectively). Radiotherapy (RT) above<br />

the diaphragm increased risk <strong>of</strong> STs above the diaphragm (hazard ratio<br />

(HR) 2.4, P�0.001), while subdiaphragmatic RT was associated with a<br />

1.7-fold increased HR <strong>of</strong> a subdiaphragmatic ST (P�0.001). An incomplete<br />

mantle field was associated with significantly lower breast cancer<br />

(BC) risk (hazard ratio (HR) 0.4, 95%CI 0.2-0.8). A cumulative procarbazine<br />

dose �4.2 g/m2 yielded a 1.3-fold increased HR (95%CI 1.0-1.7) for<br />

non-breast STs and a 2-fold (95%CI 1.2-3.1) increased HR for gastrointestinal<br />

STs, but was associated with a strongly decreased BC risk (HR 0.3,<br />

95%CI 0.2-0.6). Conclusions: SC risk after HL has decreased with<br />

treatment changes over the last decades, due to strongly decreasing risk <strong>of</strong><br />

leukemia and NHL. Smaller radiation fields and procarbazine doses �4.2<br />

g/m2 are associated with lower breast cancer risk, while high procarbazine<br />

doses increase risk <strong>of</strong> gastrointestinal STs.<br />

Lymphoma and Plasma Cell Disorders<br />

519s<br />

8038 Poster Discussion Session (Board #18), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Risk factors for development <strong>of</strong> second primary malignancies (SPM) after<br />

autologous stem cell transplant (ASCT) for multiple myeloma. Presenting<br />

Author: Amrita Y. Krishnan, City <strong>of</strong> Hope, Duarte, CA<br />

Background: Studies from the CALGB and IFM have suggested an increased<br />

incidence <strong>of</strong> SPM post ASCT in patients on lenalidomide maintenance.<br />

Patients with MM as well as patients post ASCT are inherently at higher risk<br />

<strong>of</strong> SPM. Therefore, assessment <strong>of</strong> risk factors associated with SPM would<br />

be useful in therapeutic decisions re preASCT therapy and post ASCTmaintenance.<br />

Methods: We conducted a retrospective cohort study <strong>of</strong> 841<br />

consecutive MM patients who underwent at least one ASCT at City <strong>of</strong> Hope<br />

from 1989 to 2009. Sixty cases with 70 SPMs were identified. A nested<br />

case-control study was also conducted to understand the role <strong>of</strong> therapeutic<br />

exposures associated with SPMs. Controls were MM patients post ASCT<br />

matched by year <strong>of</strong> HCT (�5 years). Results: The median length <strong>of</strong> follow up<br />

was 3.3 yrs. (range 0.3-19.9). Median age at ASCT was 56 yrs (range<br />

18-77). 62% had received a single autologous HCT, 27% tandem<br />

autologous HCT, 11% had received multiple HCTs (72 had a second<br />

allogeneic HCT)). The overall cumulative incidence <strong>of</strong> any SPM was 7.4%<br />

at 5 years and 15.9% at 10 years; the cumulative incidence <strong>of</strong> SPMs for<br />

patients �55 years approached 21.9% at 10 years. The cumulative<br />

incidence <strong>of</strong> MDS/AML was 1.8% and <strong>of</strong> solid tumors was 13.0%. Factors<br />

examined included age, race, sex, number and individual therapeutic<br />

exposures ( pre-ASCT, conditioning, and post-ASCT), disease status at<br />

ASCT. Multivariate Cox regression analysis revealed non-Hispanic whites<br />

(RR�2.4, 95% CI, 1.2-4.6, p�0.01) and older age (�55) at diagnosis <strong>of</strong><br />

MM (RR�2.3, 95% CI, 1.3-4.1, p�0.004) to be associated with an<br />

increased risk <strong>of</strong> developing SPMs. Only cumulative thalidomide exposure<br />

(both pre-ASCT and post-ASCT) demonstrated a trend toward a positive<br />

association (OR�3.5, 95% CI, 0.6-19.4, p�0.15). Six patients (3 cases<br />

and 3 controls) were exposed to lenalidomide prior to development <strong>of</strong> SPM<br />

(OR�1.0, 95% CI, 0.14-7.10). Conclusions: This single institution analysis<br />

identified non-hispanic whites and older age to be associated with<br />

increased risk <strong>of</strong> developing SPM in pts post ASCT for MM. The trend<br />

towards increased risk with thalidomide exposure may be suggestive <strong>of</strong> a<br />

class effect from IMIDs that is not restricted to lenalidomide alone.<br />

8040 Poster Discussion Session (Board #20), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Impact <strong>of</strong> t (11;14) on the outcome <strong>of</strong> autologous hematopoietic cell<br />

transplantation (Auto-HCT) in multiple myeloma. Presenting Author: Koji<br />

Sasaki, Beth Israel Medical Center, New York, NY<br />

Background: Approximately 15-20% <strong>of</strong> patients with multiple myeloma<br />

(MM) present with t(11;14)(q13;q32) involving IgH and CCND1-XT genes.<br />

In this study, we report the impact <strong>of</strong> the t(11;14) on the outcome <strong>of</strong><br />

patients with MM. Methods: We performed a retrospective chart review on<br />

patients with MM who underwent high-dose chemotherapy followed by<br />

auto-HCT at the M.D. Anderson Cancer Center between 2/2000 and<br />

8/2010, and had conventional cytogenetic (CC) or fluorescent in situ<br />

hybridization (FISH) results available before transplant. The primary<br />

objective was to compare the progression free survival (PFS) and overall<br />

survival (OS) <strong>of</strong> patients with t(11;14) to patients without chromosomal<br />

abnormalities. Results: CC or FISH studies were available for 1239<br />

patients: 863 normal, 28 with t(11;14), 348 with other abnormalities.<br />

Concurrent high-risk abnormalities on CC or FISH were seen in 15/28<br />

patients with t(11;14): del(13q) in 11 , del(17p) in 3, and t(14;16)(q32;<br />

q23) in 1. Induction treatment in patients with t(11;14) was: bortezomib �<br />

dexamethasone �/- thalidomide/lenalidomide : 15 (53%), thalidomide or<br />

lenalidomide � dexamethasone: 11 (39%), others 2 (8%); they received<br />

auto-HCT after a median <strong>of</strong> one line (1-7) <strong>of</strong> therapy. Median follow up in<br />

surviving patients was 39 months. There was no significant difference in<br />

median time from diagnosis to auto-HCT from diagnosis (6.9 vs. 7.7<br />

months, p�1.0), disease status at auto HCT (�PR1: 82 vs. 76%, �PR1: 7<br />

vs. 11%, relapsed 10 vs. 13%), complete remission (CR: 21% vs. 32%;<br />

p�0.30), very good partial remission (VGPR: 29% vs. 21%; p�0.23) or<br />

overall response (75% vs. 85%; p�0.18) between patients with t(11;14)<br />

and normal karyotype. Median PFS in patients with t(11;14) and normal<br />

karyotype was 15.7 months and 35.9 months, respectively (p�0.017).<br />

Median OS in patients with t(11;14) and normal karyotype was 51.4<br />

months and 88.4 months, respectively (p�0.03). There was no difference<br />

in PFS (p�0.25) or OS (p�0.71) in patients with t(11;14), with or without<br />

other high-risk chromosomal abnormalities. Conclusions: In this large<br />

single center study with a long follow up, we demonstrated that t(11;14) in<br />

MM is associated with a shorter PFS and OS in the context <strong>of</strong> auto-HCT.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


520s Lymphoma and Plasma Cell Disorders<br />

8041 Poster Discussion Session (Board #21), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Metronomic therapy for heavily pretreated relapsed/refractory multiple<br />

myeloma (RRMM). Presenting Author: Xen<strong>of</strong>on Papanikolaou, Myeloma<br />

Institute for Research and Therapy, Little Rock, AR<br />

Background: RRMM represents a true challenge in MM therapy. Based on<br />

the effectiveness <strong>of</strong> metronomic therapy in solid tumors, we developed a<br />

treatment <strong>of</strong> metronomically scheduled therapy (MT) for RRMM (Hollmig,<br />

ASH 2004). We are now updating our experience with a median follow up <strong>of</strong><br />

25.6 months. Methods: We identified 187 patients treated with MT from<br />

03/2004 to 11/2011. Results: The median age was 61 years (range<br />

36-83); the median number <strong>of</strong> prior therapies was 14 (range 1-51). 79% <strong>of</strong><br />

patients had prior HDT, 99% had a prior exposure to bortezomib, 98% to an<br />

IMiD, and 95% to their combination . The median number <strong>of</strong> completed<br />

MT cycles was 1 (range 1-5). 63% <strong>of</strong> patients had a response <strong>of</strong> MR or<br />

better (6% CR ,7% VGPR, 36% PR, 16% MR). The median overall (OS)<br />

and progression free (PFS) survivals were 11.3 and 3.7 months respectively.<br />

91 <strong>of</strong> 187 patients had gene expression pr<strong>of</strong>iling (GEP) prior to<br />

initiation <strong>of</strong> MT, <strong>of</strong> which 53% were high risk according to the 70-gene<br />

(GEP70) risk model. OS was affected by elevated CRP (� 8mg/L,<br />

HR�1.71, p�0.009) and cytogenetic abnormalities within 6 months <strong>of</strong><br />

initiation <strong>of</strong> MT (HR�2.45, p�0.001). For the 91 patients with GEP data<br />

available, OS was correlated with elevated CRP � 8mg/L (HR�2.11,<br />

p�0.009) and GEP70 high-risk (HR�2.65, p�0.001), which also showed<br />

a trend towards shorter PFS. Hematological toxicity grading was difficult as<br />

69% <strong>of</strong> patients presented with grade ��3 thrombocytopenia within 90<br />

days prior to starting MT. Grade 4 leucopenia , anemia thrombocytopenia<br />

due to MT occurred in 74%, 17%, 89% <strong>of</strong> patients respectively. Incidence<br />

<strong>of</strong> grade 4 neutropenic fever was 1%. Grade 4 or worse incidence <strong>of</strong> all<br />

non-hematological toxicities was below 9%. Most patients were treated in<br />

the outpatient setting (95%) and secondary admissions due to regimen<br />

toxicity occurred in 20%. Conclusions: MT is an effective salvage treatment<br />

in RRMM, with a high ORR and a favorable toxicity pr<strong>of</strong>ile.<br />

MT regimen.<br />

Agent Days Dosage<br />

Bortezomib 1,4,7,10,13,16 1 mg/m2 Thalidomide 1-16 200 mg<br />

Dexamethasone 1,4,7,10,13,16 20-40 mg<br />

Doxorubicin<br />

1-16 3 mg/m2 IV continuous infusion<br />

hydrochloride<br />

Cisplatin 1-16 1.5-3 mg/m 2 IV continuous infusion<br />

Rapamycin 1-16 3mg on day 1 , 1mg days 2-16<br />

8043 Poster Discussion Session (Board #23), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Long-term results <strong>of</strong> the phase II trial <strong>of</strong> the oral mTOR inhibitor everolimus<br />

(RAD001) in relapsed or refractory Waldenstrom macroglobulinemia.<br />

Presenting Author: Irene M. Ghobrial, Dana-Farber Cancer Institute,<br />

Boston, MA<br />

Background: The mammalian target <strong>of</strong> rapamycin (mTOR) signal pathway<br />

controls cell proliferation and survival. The trial’s goal was to determine the<br />

anti-tumor activity and safety <strong>of</strong> single-agent everolimus (TORC1 inhibitor)<br />

in patients with relapsed/refractory Waldenstrom macroglobulinemia (WM).<br />

Methods: Eligible patients had measurable disease (IgM monoclonal<br />

protein �1000 mg/dL with �10% marrow involvement or nodal masses �2<br />

cm), a platelet count �75,000 x 106 /L, a neutrophil count �1,000 x<br />

106 /L. Patients received everolimus 10 mg PO daily. Tumor response was<br />

assessed after cycles 2 and 6 and then every 3 cycles until progression.<br />

Results: 60 pts were treated. The median age was 64 years (range, 43-85).<br />

The median number <strong>of</strong> prior therapies was 3 (range, 1-11). All but two<br />

patients (97%) had received prior rituximab based therapy and 64% <strong>of</strong><br />

patients had received prior alkylator based therapies. The overall response<br />

rate (complete response CR� partial response PR� minimal response MR)<br />

was 73% (95% CI: 60-84%), with a PR <strong>of</strong> 50% and 23% MR. The median<br />

time to progression (TTP), progression-free survival (PFS), and overall<br />

survival (OS) for the entire study population is 28 months (mos), (95% CI:<br />

18-not reached (NR)), 22 mos (95% CI: 12 0-NR), and 55 mos (95% CI:<br />

55-NR), respectively. The estimated PFS at 12 and 24 months is 62%<br />

(95%CI: 51-75%), and 48% (95%CI: 37-63%), respectively. The 30<br />

patients who achieved a PR responded after a median <strong>of</strong> 2 months (range,<br />

1-26) <strong>of</strong> treatment. The median duration <strong>of</strong> response (DR) for these<br />

patients has not yet been reached and 19 <strong>of</strong> these patients remain in<br />

response after a median follow up <strong>of</strong> 31 months (range, 3-54). All but two<br />

patients had a decrease in their serum IgM. Grade 3 or higher related<br />

toxicities were observed in 67% <strong>of</strong> patients. The most common were<br />

hematological toxicities with cytopenias. Pulmonary toxicity occurred in<br />

5% <strong>of</strong> patients. Dose reductions due to toxicity occurred in 63% <strong>of</strong><br />

patients. Conclusions: Everolimus has high single-agent activity with an<br />

overall response rate <strong>of</strong> 73% and manageable toxicity in patients with<br />

relapsed WM, and <strong>of</strong>fers a potential new therapeutic strategy for this patient<br />

population.<br />

8042 Poster Discussion Session (Board #22), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Detection <strong>of</strong> the MYD88 L265P mutation in Waldenström’s macroglobulinemia<br />

using a highly sensitive allele-specific PCR assay. Presenting Author:<br />

Lian Xu, Dana-Farber Cancer Institute, Boston, MA<br />

Background: Waldenström’s macroglobulinemia (WM) is a B-cell malignancy<br />

characterized by bone marrow (BM) infiltration with lymphoplasmacytic<br />

cells and production <strong>of</strong> an IgM paraprotein. By whole genome<br />

sequencing, we recently identified a somatic mutation (L265P) in the<br />

MYD88 gene in 27/30 (90%) WM patients (Treon et al, ASH 2011). To<br />

expand this finding for possible diagnostic testing, we developed an<br />

allele-specific PCR assay for MYD88 L265P and evaluated this assay in a<br />

large cohort <strong>of</strong> WM patients. Methods: An allele-specific PCR assay was<br />

developed with a threshold <strong>of</strong> detection <strong>of</strong> 0.125% for MYD88 L265P.<br />

DNA from bone marrow aspirates from 99 patients with the clinicopathological<br />

diagnosis <strong>of</strong> WM was used for assessment <strong>of</strong> MYD88 L265P expression<br />

by both allele-specific PCR and Sanger sequencing. Findings were correlated<br />

with clinical parameters using ANOVA. Results: We observed that<br />

85/99 (86%) WM patients were positive for MYD88 L265P using the<br />

allele-specific PCR assay. Of the 85 allele-specific PCR positive patients,<br />

81 demonstrated a detectable mutation peak by Sanger sequencing. All 14<br />

allele-specific PCR negative patients remained negative by Sanger sequencing.<br />

By the allele-specific PCR assay, MYD88 L265P positive patients<br />

showed greater bone marrow involvement, higher serum IgM and lower<br />

serum IgA and IgG levels versus MYD88 L265P negative patients<br />

(p�0.008). Conclusions: MYD88 L265P is highly expressed in BM samples<br />

<strong>of</strong> WM patients using an allele-specific PCR assay, and is associated with<br />

greater bone marrow disease burden and serum IgM levels. Use <strong>of</strong><br />

allele-specific PCR provides a simple and sensitive diagnostic tool for<br />

detection <strong>of</strong> the MYD88 L265P mutation.<br />

8044 General Poster Session (Board #32A), Mon, 1:15 PM-5:15 PM<br />

Survival with splenectomy in splenic marginal zone lymphoma: Analysis <strong>of</strong><br />

the Surveillance, Epidemiology and End Results (SEER) database. Presenting<br />

Author: Adam J. Olszewski, Alpert Medical School <strong>of</strong> Brown University,<br />

Providence, RI<br />

Background: The role <strong>of</strong> splenectomy as the primary therapy for splenic<br />

marginal zone lymphoma has been questioned. We studied relative survival<br />

in SMZL and the impact <strong>of</strong> splenectomy on lymphoma-specific survival<br />

(LSS). Methods: SMZL cases (diagnosed in 1993-2008) were derived from<br />

the SEER database. Age, sex and race-matched actuarial survival data were<br />

summarized. Factors predictive <strong>of</strong> splenectomy were studied using logistic<br />

regression with a subsequent propensity score (PS)-weighted survival<br />

analysis. Results: 1071 patients were identified with a median age <strong>of</strong> 69<br />

years (range, 25-96) and a median follow up <strong>of</strong> 33 months. 53% were<br />

women and 92% were white. 70% <strong>of</strong> the lymphomas were stage III/IV with<br />

B symptoms recorded in 22.3%. 54% <strong>of</strong> patients underwent splenectomy.<br />

The significant factors predictive <strong>of</strong> surgery included younger age (p�10- 14), stage I/II (p�10-15 ), B-symptoms (p�0.003), no prior malignancy<br />

(p�0.002), with significantly lower rates in black patients (OR 0.41,<br />

95%CI 0.21-0.80, p�0.008), on the Pacific Coast (OR 0.62, 95%CI<br />

0.47-0.81, p�0.0004) and with decreasing rates over time (p�0.0002).<br />

The actuarial 5-year relative survival was 82.8% (95%CI 77.9-86.7%)<br />

with no difference by sex (p�0.79), race or stage. 54% <strong>of</strong> deaths were<br />

related to SMZL with the estimated LSS <strong>of</strong> 80.8% (95%CI 78-84%).<br />

Splenectomy was not associated with improved LSS in propensity scorestratified<br />

log-rank test (p�0.60) or in the PS-weighted Cox model<br />

(HR�0.93, 95%CI 0.62-1.38, p�0.70). Advancing age (HR 1.05, 95%CI<br />

1.03-1.07, p�10-8 ) and presence <strong>of</strong> B-symptoms (HR�1.98, 95%CI<br />

1.30-3.01, p�0.002) were significantly predictive <strong>of</strong> death from SMZL,<br />

with no evidence <strong>of</strong> improvement in LSS over the years (HR�0.97,<br />

95%CI�0.91-1.03, p�0.34). There was also no detectable impact <strong>of</strong><br />

splenectomy on survival in patients who ultimately died <strong>of</strong> SMZL (p�0.83).<br />

Conclusions: In this cohort, the largest studied to date, splenectomy did not<br />

demonstrate a survival benefit in SMZL. Patients continue to have<br />

decreased survival despite advances in other indolent B-cell lymphomas.<br />

The role <strong>of</strong> splenectomy versus chemoimmunotherapy remains to be<br />

determined.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


8045 General Poster Session (Board #32B), Mon, 1:15 PM-5:15 PM<br />

Survival outcomes in marginal zone lymphomas in the rituximab era:<br />

Analysis <strong>of</strong> the Surveillance, Epidemiology, and End Results (SEER)<br />

database. Presenting Author: Jorge J. Castillo, The Miriam Hospital,<br />

Providence, RI<br />

Background: Despite prognostic models developed for marginal zone<br />

lymphoma (MZL), the impact <strong>of</strong> different characteristics and treatments on<br />

survival in the population is largely unknown. We studied survival <strong>of</strong> MZL<br />

patients included in the SEER database. Methods: Records <strong>of</strong> MZL adult<br />

cases diagnosed between 1989-2008 were studied using descriptive<br />

methods and analysis <strong>of</strong> overall (OS) and lymphoma-specific (LSS) survival<br />

based on Kaplan-Meier function, stratified log-rank tests and Cox proportional<br />

hazard models. Results: 13,957 patients with MZL were identified<br />

and classified as splenic (SMZL; n�1,111, 8%), nodal (NMZL; n�4,101,<br />

29%) or extranodal MALT-type MZL (MALT; n�8,745, 63%). The median<br />

age was 68 years, 74% <strong>of</strong> patients were white and 55% were women.<br />

Median follow-up was 40 months. MALT was more common in non-<br />

Caucasians (p�10-27 ). B-symptoms were more common in SMZL (p�10- 5). Both LSS and OS were significantly better for MALT (p�10-60 ) with no<br />

difference between SMZL and NZML (p�0.30). 10-year LSS estimates<br />

were 67% for SMZL, 67% for NMZL, 84% for MALT. There was evidence<br />

for improved LSS since 2000 in MALT (HR 0.69, 95% CI 0.59-0.82,<br />

p�0.0003) and NMZL (HR 0.64, 95% CI 0.54-0.77, p�10-5 ), but not for<br />

SMZL (HR 0.85, 95% CI 0.56-1.28, p�0.43). Similar results were found<br />

for OS. There were differences in survival in MALT subtypes depending on<br />

primary site (p�10-12 ; Table). Conclusions: In the rituximab era, survival<br />

has improved for MALT and NMZL, but not for SMZL, possibly due to<br />

disparate treatment paradigms. The prognosis <strong>of</strong> MALT is different depending<br />

on primary site <strong>of</strong> involvement.<br />

Site n 5-year OS (%) 95% CI<br />

Skin 664 87.1 83.5-90.0<br />

Thyroid 179 85.8 78.0-90.9<br />

Ocular 1,108 82.4 79.4-85.0<br />

Salivary 719 82.4 78.7-85.5<br />

Connective tissue 190 81.7 73.9-87.4<br />

Breast 277 78.9 72.2-84.2<br />

Gastric 3,207 72.1 70.3-73.9<br />

Lung 737 72.1 67.9-75.9<br />

Bowel 791 71.1 67.2-74.7<br />

Other 392 72.7 67.0-77.7<br />

8047 General Poster Session (Board #32D), Mon, 1:15 PM-5:15 PM<br />

The utility <strong>of</strong> serum lactate dehydrogenase (LDH) in the follow-up <strong>of</strong><br />

patients with diffuse large B-cell lymphoma (DLBCL). Presenting Author:<br />

Navneeth Rao Bongu, Nebraska Medical Center, Omaha, NE<br />

Background: The prognostic significance <strong>of</strong> high serum LDH is well<br />

established for both indolent and aggressive lymphomas at diagnosis. The<br />

performance characteristics <strong>of</strong> LDH in predicting relapse after treatment in<br />

patients (pts) with DLBCL has not been well studied. The determination <strong>of</strong><br />

utility <strong>of</strong> LDH monitoring in pts with DLBCL would have widespread<br />

practice implications. Methods: We searched the Nebraska Lymphoma<br />

Study Group database to identify pts with DLBCL who were treated with a<br />

rituximab-based regimen, from 2000 to 2010, and sustained a complete<br />

response (CR). For the pts who relapsed (RP), after sustaining a CR, we<br />

collected the LDH level at relapse and the LDH level 3 months prior<br />

(considered baseline). For pts who never relapsed (NR), we collected the<br />

last 2 LDH levels at follow-up; levels had to be at least 3 months apart. The<br />

relative increase <strong>of</strong> LDH was compared, to baseline, among RP vs. NR.<br />

Results: We identified 129 pts, 15 pts were excluded from the analysis as<br />

their LDH results were not available, 27 relapsed and 87 didn’t. Median<br />

age <strong>of</strong> pts was 57 years. Only 9/27 RP (33%) had increase in LDH above<br />

upper limit <strong>of</strong> normal (ULN) at relapse. The mean increase in LDH at<br />

relapse was 1.2 fold above the ULN for RP vs. 0.83 for NR (p�0.59). The<br />

mean increase in LDH, from baseline, was 1.1 fold in NR vs. 1.3 in RP<br />

(p�0.3). The likelihood ratio (LR) <strong>of</strong> relapse was 4.65 for pts who had 1.5<br />

fold increase in LDH above baseline (1.5xLDH) vs. those who didn’t<br />

(p�0.03). The sensitivity, specificity, positive and negative predictive<br />

values <strong>of</strong> 1.5xLDH for detecting relapse, compared to clinical and imaging<br />

findings were 0.18, 0.95, 0.55, and 0.79 respectively. Also, 1.5xLDH at<br />

relapse was significantly associated with the presence <strong>of</strong> fever (LR�5.74;<br />

p�0.03) but not with other symptoms including drenching night sweats,<br />

anemia, or new/progressive lymphadenopathy. Conclusions: A 1.5 fold<br />

increase in LDH, over a period <strong>of</strong> 3 months, is associated with increased<br />

likelihood <strong>of</strong> relapse from DLBCL. Modest elevations in LDH (�1.5 fold <strong>of</strong><br />

baseline) doesn’t seem to be associated with relapse. LDH, when elevated<br />

at least 1.5 fold <strong>of</strong> baseline, is a specific (i.e. 56%), but not a sensitive (i.e.<br />

19%) marker, for relapse <strong>of</strong> DLBCL.<br />

Lymphoma and Plasma Cell Disorders<br />

521s<br />

8046 General Poster Session (Board #32C), Mon, 1:15 PM-5:15 PM<br />

Early determination <strong>of</strong> treatment sensitivity in HIV-related Hodgkin lymphoma<br />

by FDG-PET/CT after two cycles <strong>of</strong> ABVD chemotherapy: A<br />

European Cooperative Study Group on AIDS and Tumors (GECAT) study.<br />

Presenting Author: Nicolas Mounier, CHU l’Archet, Nice, France<br />

Background: The high prognostic value <strong>of</strong> FDG-PET/CT performed after 2<br />

cycles <strong>of</strong> chemotherapy for HIV negative Hodgkin lymphoma (HL) is well<br />

known. However, experience with PET in HIV-related HL needs to be further<br />

studied as nodal FDG uptake can be observed in various opportunistic<br />

infections and AIDS-related conditions. Methods: A total <strong>of</strong> 45 consecutive<br />

HL patients (pts) were enrolled in 10 centers from the GECAT. There were<br />

42 males and 3 females. Median age was 46 yo, range [26;64]. Median<br />

CD4 count was 391/mm3 , range [33;1191]. Viral load was negative in 38<br />

pts (84%) and uncontrolled in 5 pts (11%). Forthy three pts (96%)<br />

received concomitant HAART. HL was staged III-IV in 25 pts. International<br />

Prognostic Index scored 3-5 in 24 pts. All PET studies were performed after<br />

2 ABVD cycles. They were scored, blinded to treatment outcome, according<br />

to the 5-point Deauville visual scale. It was considered as negative when<br />

scored 1-3 (i.e tumor FDG uptake less or equal than liver uptake) and<br />

positive when scored 4-5 (i.e. more than liver uptake or new lesions).<br />

Chemotherapy was not modified : 4-8 cycles <strong>of</strong> ABVD, as initially planned.<br />

Results: Overall, 35 pts (78%) achieved a CR after the end <strong>of</strong> treatment.<br />

Three pts received Involved Field Radiation Therapy. At a median follow-up<br />

<strong>of</strong> 18 months, 3 pts relapsed and 2 <strong>of</strong> them died from HL. The 2 yr OS and<br />

PFS were estimated at 94 and 90%, respectively. PET after 2 cycles <strong>of</strong><br />

ABVD was negative in 40 pts (89%) and positive in 5 pts (11%). Patients<br />

with negative PET had a significantly better outcome than those with<br />

positive PET in term <strong>of</strong> 2 yr PFS (94% vs 60%, P�0.005), and 2 yr OS<br />

(100% vs 60%, P�0.0002). The negative predictive value was estimated<br />

at 97% and specificity at 93%. All patients who were PET-negative after<br />

the 2nd cycle stayed PET-negative after the 4th cycle and entered a<br />

durable CR. Conclusions: This largest study in HIV-positive HL showed that<br />

interim PET could play a central role in driving risk-tailored treatment. In<br />

further studies, de-escalation strategies should be tested for patients<br />

responding after 2 cycles <strong>of</strong> ABVD and those not reponding should be<br />

managed with intensive salvage strategies.<br />

8048 General Poster Session (Board #32E), Mon, 1:15 PM-5:15 PM<br />

Association <strong>of</strong> low expression <strong>of</strong> VEGF121 soluble is<strong>of</strong>orm with prognostic<br />

impact and survival in patients with activated B-cell-like (ABC-like) diffuse<br />

large B-cell lymphoma (DLBCL) from the CORAL trial. Presenting Author:<br />

Catherine Thieblemont, Saint-Louis Hospital, Paris, France<br />

Background: Gene expression pr<strong>of</strong>iling (GEP) has demonstrated that survival<br />

<strong>of</strong> DLBCL is influenced by the origin <strong>of</strong> the tumor cells (COO),<br />

germinal center-like (GC-like) vs ABC-like (Alizadeh et al. 2000), but also<br />

by the tumor microenvironment, particularly the angiogenesis (Lenz et al.,<br />

2008). To further understand the prognostic impact <strong>of</strong> these tumoral<br />

characteristics, we analysed the prognostic impact <strong>of</strong> COO and the<br />

expression <strong>of</strong> biomarkers involved in angiogenesis, in a selected population<br />

<strong>of</strong> 36 patients with relapsed/refractory DLBCL included in CORAL (Gisselbrecht,<br />

ASCO 2011). Methods: Expression <strong>of</strong> 5 biomarkers including VEGF<br />

(is<strong>of</strong>orms 121, 165, and 189), and their receptors (VEGFR-1 and R-2) was<br />

assessed by quantitative qRT-PCR after total RNA extraction and cDNA<br />

synthesis <strong>of</strong> tumor samples. COO was determined by GEP, and progression<br />

free- (PFS) and overall- (OS) survivals were analysed. Results: In the study<br />

population, VEGF121 expression below the median was associated to a<br />

better outcome, with 4 year-PFS at 63% vs 33% (p�.053), and a<br />

4-year-OS at 79% vs 37% (p � 0.032), respectively. VEGF-165 -189,<br />

VEGF-R1, -R2 did not have any significant impact. Eighteen patients were<br />

predicted as ABC-like DLBCL and 18 as GCB-like DLBCL. In patients with<br />

ABC-like DLBCL, low VEGF121 level was associated to a significantly<br />

better survival than in those with high VEGF121 level: 4-year OS at 100%<br />

vs 36% (p�.011). The type <strong>of</strong> induction treatment, R-ICE or R-DHAP, did<br />

not influence the outcome. The differences in outcome according to VEGF<br />

is<strong>of</strong>orms were not significant among CGB patients. Conclusions: Our data<br />

suggest that angiogenesis plays a central role in ABC-like DLBCL. VEGF121<br />

seemed to be a key player in this context and should be proposed as a target<br />

in the treatment <strong>of</strong> patients with ABC-like DLBCL.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


522s Lymphoma and Plasma Cell Disorders<br />

8049 General Poster Session (Board #32F), Mon, 1:15 PM-5:15 PM<br />

Transformation <strong>of</strong> follicular lymphoma in the era <strong>of</strong> immunochemotherapy:<br />

A population-based study from British Columbia. Presenting Author:<br />

Abdulwahab J. Al-Tourah, British Columbia Cancer Agency-Fraser Valley<br />

Centre and University <strong>of</strong> British Columbia, Surrey, BC, Canada<br />

Background: Several published series have established that the risk <strong>of</strong><br />

transformation <strong>of</strong> follicular lymphoma (FL) to aggressive lymphoma is<br />

approximately 3% /year (15%-20% at 5 years). The addition <strong>of</strong> rituximab<br />

(R) to chemotherapy (immuno-chemotherapy) has significantly improved<br />

the outcome <strong>of</strong> patients with FL. The impact <strong>of</strong> immuno-chemotherapy on<br />

the risk <strong>of</strong> transformation remains unknown. We assessed whether the<br />

introduction <strong>of</strong> immuno-chemotherapy has altered this risk. Methods: We<br />

examined the Lymphoid Cancer Database <strong>of</strong> the British Columbia Cancer<br />

Agency for FL patients treated with immuno-chemotherapy. Inclusion<br />

criteria: FL grades 1-3A by WHO criteria; only patients requiring treatment<br />

at diagnosis were included. Exclusion criteria: FL grade 3B or composite<br />

histology (FL and DLBCL) at diagnosis; pts who received anthracyclinebased<br />

chemotherapy; and HIV positivity. The diagnosis <strong>of</strong> transformation<br />

was confirmed by biopsy when possible (n�19; 79%) but patients who<br />

were considered to have transformed based on pre-defined clinical assessment<br />

(n�5; 21%) were also included in the analysis. Results: We identified<br />

261 pts with FL grade 1-3A requiring treatment at diagnosis, who received<br />

immuno-chemotherapy; median f/u 47 months (0.2-116), median age, 61<br />

y (34-86). Treatment: 243 (93%), R-CVP <strong>of</strong> which 145 (59%) also<br />

received maintenance R; 9 (4%), R-Fludarabine combination. 24 pts<br />

developed transformed aggressive lymphoma. The risk <strong>of</strong> transformation for<br />

the entire group was approximately 2% per year or 10% at 5 years.<br />

However, pts treated with maintenance R (n�151) had a lower risk <strong>of</strong><br />

transformation compared to pts who only received R-chemo at induction<br />

(n� 110), 8% vs 20% at 5 years respectively, (P� 0.003). The posttransformation<br />

outcome remains poor with a median survival <strong>of</strong> 6 months.<br />

Conclusions: We and other groups have demonstrated that the risk <strong>of</strong><br />

transformation from FL to aggressive lymphoma is approximately 15% to<br />

20% by 5 years. Our study suggests that the introduction <strong>of</strong> immunochemotherapy<br />

has reduced this risk to less than 10%. This effect is<br />

particularly apparent when patients receive maintenance R. The outcome<br />

for patients who develop transformation remains poor.<br />

8051 General Poster Session (Board #32H), Mon, 1:15 PM-5:15 PM<br />

Efficacy <strong>of</strong> alemtuzumab (ALZ) in combination with dose-adjusted EPOCH<br />

(DA-EPOCH) in untreated nodal peripheral T-cell lymphoma (PTCL).<br />

Presenting Author: Cliona Grant, National Cancer Institute, Bethesda, MD<br />

Background: The survival <strong>of</strong> patients (pts) with PTCL (excluding ALCL) is<br />

disappointing following anthracycline-based therapy and novel approaches<br />

are needed. In diffuse large B-cell lymphoma, the addition <strong>of</strong> anti-CD20<br />

therapy to CHOP has significantly improved outcome and we investigated if<br />

in PTCL, adding ALZ – which targets CD52, expressed on most PTCLs – was<br />

feasible and improved outcome. Methods: This was a single-center phase<br />

I/II trial <strong>of</strong> ALZ, in combination with DA-EPOCH in patients with treatmentnaïve<br />

CD52� PTCL. The MTD <strong>of</strong> 30 mg ALZ was combined with DA-EPOCH<br />

for phase II evaluation resulting in an intention-to-treat population (ITTP)<br />

<strong>of</strong> 30 pts. Results: Patient (n�30) characteristics: median (range) age 50<br />

(17-77); M:F 1:1; Stage III/IV 27 (90%); IPI �2 22 (73%). Histologies:<br />

ATLL 11 (37%), PTCL NOS 6 (20%), AITL 5 (16%), Hepatosplenic 3<br />

(10%), Peripheral T NK cell 2 (7%), Other 3 (10%). At 67 months median<br />

follow-up, 11 patients were alive, 10 disease-free. Median overall (OS) and<br />

event-free survivals (EFS) were 15.4 and 6.7 months respectively. Outcome<br />

was substantially better for pts with ‘nodal’ (AITL, PTCL-NOS, Other)<br />

compared to ‘non-nodal’ (ATLL and ‘extranodal’ histologies) PTCL subtypes.<br />

3-year OS for the two groups was 58.3% and 27.8% respectively<br />

(p�0.082); 3-yrs EFS was 50% (median 27.0 months) and 22.2%<br />

(median: 5 months) respectively (p�0.041). Half <strong>of</strong> the ‘nodal’ PTCL pts<br />

had sustained long-term complete remissions demonstrated by a plateau in<br />

the EFS curve at 27 months. There were 3 treatment related deaths: 2 from<br />

neutropenic sepsis; 1 from toxoplasma. Other toxicities included CMV and<br />

BK virus reactivation in 53% and 30% respectively. Febrile neutropenia<br />

was seen in 20% and grade 4 thrombocytopenia in 12 % <strong>of</strong> cycles.<br />

Conclusions: Although it has significant infectious toxicity, ALZ 30mg and<br />

DA-EPOCHin untreated PTCL is feasible and associated with a long-term<br />

favorable outcome in nodal but not extranodal or leukemic PTCL. A phase<br />

III study <strong>of</strong> ALZ 30mg with anthracycline-based therapy is ongoing and we<br />

await the results with interest.<br />

8050 General Poster Session (Board #32G), Mon, 1:15 PM-5:15 PM<br />

Long-term follow-up results <strong>of</strong> a phase I/II study <strong>of</strong> concurrent chemoradiotherapy<br />

for localized nasal NK/T-cell lymphoma (NKTCL): JCOG0211.<br />

Presenting Author: Motoko Yamaguchi, Mie University, Tsu, Japan<br />

Background: Concurrent chemoradiotherapy has been regarded as one <strong>of</strong><br />

the standard management for localized nasal NKTCL. However, its longterm<br />

efficacy and toxicity is not known. Methods: The JCOG0211 trial is a<br />

phase I/II study <strong>of</strong> concurrent chemoradiotherapy consisting <strong>of</strong> radiotherapy<br />

(RT) <strong>of</strong> 50 Gy and 3 cycles <strong>of</strong> DeVIC (carboplatin, etoposide,<br />

ifosfamide, dexamethasone) for newly diagnosed, localized nasal NKTCL<br />

(JCO 2009). Patients (Pts) with newly diagnosed, localized diseases (IE &<br />

contiguous IIE with cervical node involvement) who were 20-69 yrs <strong>of</strong> age<br />

with PS 0-2 were eligible. 3-D conformal RT planning with a wide margin<br />

(� 2 cm to the gross tumor, the entire nasal cavity and the nasopharynx)<br />

and a 2-step cone down were required. 33 pts were enrolled in the study,<br />

27 <strong>of</strong> whom were treated with RT and a 2/3-dose <strong>of</strong> DeVIC, which was<br />

selected as the recommended phase II dose in the preceding phase I<br />

portion <strong>of</strong> the trial. All pts completed RT without any protocol violations.<br />

Long-term follow-up results on overall survival (OS), progression-free<br />

survival (PFS) and toxicity were evaluated. Results: The median follow-up<br />

was 69 months (range, 62-96). The pt (N�33) characteristics were as<br />

follows: median age 54 yrs (range, 21-68); stage IIE 33%; B symptom (�)<br />

36%; elevated serum LDH 21%. %5-yr OS and PFS were 73% (95%CI,<br />

54-85%) and 67% (95%CI, 48-80%), respectively. 11 pts (33%) experienced<br />

disease recurrence. Two achieved a 2nd CR by salvage chemotherapies<br />

followed by allogeneic stem cell transplantation, and the remaining 9<br />

pts died <strong>of</strong> disease. There was no observed death and disease progression<br />

after 34 and 31 months, respectively. One pt experienced Grade 3 irregular<br />

menstruation for 3 years. No other Grade 3 or 4 late non-RT-associated<br />

adverse events (AEs) were observed. One pt received plastic surgery due to<br />

Grade 4 RT dermatitis. No other Grade 3 or greater RT-associated late AEs<br />

were encountered. Conclusions: Both survival benefit and disease control<br />

from concurrent chemoradiotherapy with RT and DeVIC are maintained<br />

during a 5-yr follow-up, indicating the excellent efficacy <strong>of</strong> this approach as<br />

a first-line therapy for localized nasal NKTCL. Long-term toxicity is<br />

acceptable.<br />

8052 General Poster Session (Board #33A), Mon, 1:15 PM-5:15 PM<br />

Radioimmunotherapy efficiency and safety in consolidation and relapse<br />

treatment <strong>of</strong> aggressive B-cell non-Hodgkin lymphoma: An updated analysis<br />

<strong>of</strong> 230 patients <strong>of</strong> the international RIT-network. Presenting Author:<br />

Karin Hohloch, Hematology and Oncology, Georg August University Göttingen,<br />

Goettingen, Germany<br />

Background: In aggressive lymphoma, few reliable clinical data about the<br />

use <strong>of</strong> radioimmunotherapy exist. Pts. with DLBCL registered in the<br />

international RIT-Network (RIT-NT) were analyzed with regard to Indication,<br />

line <strong>of</strong> therapy and outcome. Methods: The RIT-NT recruited 1111 pts.<br />

between Dec. 2006 and 2010. It is a web-based registry that collects<br />

observational data from RIT-treated patients with malignant lymphoma.<br />

232 pts. with DLBCL registered in the database were evaluated in the<br />

analysis. Results: 232 pts with DLBCL are registered, 17 pts were excluded.<br />

Histologic subtypes: 190 DLBCL, 15 primary mediastinal, 9 large cell<br />

anaplastic, 1 intravascular. Median age was 62 years (range 17-88), 27%<br />

<strong>of</strong> pts �70 years old. Stage I 16pts, II 54 pts, III 60 pts, IV 68 pts; 6<br />

extranodal involvement, for 11 pts. stage is not documented. 187 pts had<br />

1-3 previous chemotherapies (Ctx), 21 pts 4-6 previous Ctx, 1 pts had 7<br />

previous Ctx, for 6 pts. previous Ctx is not documented. 15 pts. had<br />

previous RIT and 24 pts a stem cell transplantation prior to RIT. 6 pts had<br />

bone marrow infiltration prior to RIT, 3 with more than 25%. 87 pts had RIT<br />

as first line (8 pts. conditioning, 68 pts consolidation, 1 primary therapy,<br />

10 other), and 84 pts received RIT in relapse (2d to 8 th. line therapy) (2<br />

pts. conditioning, 31 pts consolidation, 26 recurrence, 19 therapy refractory,<br />

6 other). Grade IV° hematotoxicity occured for neutrophils and<br />

platelets, grade III° for hemoglobin after RIT. Median time to recovery <strong>of</strong><br />

blood count was 81 days (range 0-600 days). ORR was 63,3 %; CR 54,4%;<br />

PR 8,8%, SD 0,9%, PD 23,7%, N.D. 12%. CR rate first line was 76,3 %,<br />

for relapse 44,3%. Mean overall survival (OS) in first line was 26,5 months<br />

14,3 months for pts. treated in relapse or refractory disease. Conclusions:<br />

Most pts. with DLBCL in the RIT-N received RIT as consolidation after first<br />

line therapy with excellent CR rates and OS compared to published data<br />

from risk groups . In relapsed DLBCL RIT is a safe and feasible treatment<br />

leading to satisfactory response rates with low toxicity. A prospective<br />

randomized phase III trial in front line DLBCL is currently planned (ZEST).<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


8053 General Poster Session (Board #33B), Mon, 1:15 PM-5:15 PM<br />

LR-CD: Lenalidomide combination therapy for untreated low-grade B-cell<br />

NHL. Presenting Author: Craig B. Reeder, Mayo Clinic, Scottsdale, AZ<br />

Background: Lenalidomide (LEN) is an immunomodulatory agent that has<br />

shown significant single-agent anti-lymphoma activity in patients with<br />

relapsed disease and in combination may enhance the efficacy <strong>of</strong> rituximab<br />

by various mechanisms. There is limited data on the role <strong>of</strong> LEN in<br />

combination regimens for treatment-naïve patients. This phase II single<br />

arm trial was designed to evaluate tumor response, toxicity (AE) and<br />

survival with a combination <strong>of</strong> LEN, rituximab, cyclophosphamide (CTX)<br />

and dexamethasone (DEX) (LR-CD) in symptomatic untreated patients with<br />

low grade B-cell non-Hodgkin lymphoma. Methods: Eligibility required age<br />

�18, ECOG PS �2, confirmed diagnosis <strong>of</strong> low-grade B-cell lymphoma<br />

(FL1-2, SLL, MZL or LPL/WM), measurable nodes �2cm or IgM �400mg/<br />

dL(LPL/WM), ANC �1400/mm3 , platelets �100,000/mm3 , creatinine<br />

�2mg/dL, signed informed consent and in need <strong>of</strong> therapy. Treatment<br />

consisted <strong>of</strong> IV rituximab 375mg/m2 D1, oral LEN 20mg D1-21, CTX<br />

250mg/m2 D1, 8, 15, DEX 40mg D1, 8, 15, 22 and ASA 325 mg daily in a<br />

28 day cycle. Treatment continued 2 cycles beyond best response (max<br />

12). Toxicity was assessed by NCI CTCAE v3.0. Results: 28 patients have<br />

enrolled at Mayo Clinic with 25 evaluable for toxicity and 21 for response:<br />

Median age 65(43-83), 72% male, FL 24%, MZL 28%, LPL/WM 38%, and<br />

SLL 4%. Median number <strong>of</strong> cycles given was 5. Overall response in 21<br />

patients with response data is 95% (95% CI 76-100%) with 19% CR (95%<br />

CI 5-42%), and 76% PR (confirmed and unconfirmed, 95% CI 53-92%).<br />

The ORR in 8 patients with LPL/WM with response data was 88%, all PR<br />

(95% CI 47-100%). At a median f/u <strong>of</strong> 14.7 months 96% are alive without<br />

progression. 1 death (unrelated) occurred 7.7 months after completing 12<br />

cycles <strong>of</strong> treatment. The most common grade �3 AEs were neutropenia<br />

(37.5%), leukopenia (16.7%), anemia (12.5%) and fatigue (12.5%).<br />

Conclusions: The combination LR-CD with the novel agent LEN is feasible,<br />

well tolerated and produces high response rates in symptomatic patients<br />

with low grade B-cell NHL. Toxicities are manageable and similar to<br />

reported data <strong>of</strong> single agent LEN.<br />

8055 General Poster Session (Board #33D), Mon, 1:15 PM-5:15 PM<br />

Association <strong>of</strong> prior rituximab exposure among patients undergoing autologous<br />

stem cell transplantation for relapsed/refractory diffuse large B-cell<br />

lymphoma with inferior outcomes in males compared with females.<br />

Presenting Author: Aleksandr Lazaryan, Taussig Cancer Institute, Cleveland<br />

Clinic, Cleveland, OH<br />

Background: The combination <strong>of</strong> rituximab with CHOP remains the standard<br />

upfront therapy <strong>of</strong> patients (pts) with advanced diffuse large B-cell<br />

lymphoma (DLBCL). Gender disparity in survival <strong>of</strong> pts with DLBCL appears<br />

to be significantly modified by the use <strong>of</strong> rituximab in both upfront (Ngo,<br />

2008; Pfreundschuh, 2009; Riihijarvi, 2010) and post-transplant maintenance<br />

(CORAL trial) settings. The presence <strong>of</strong> interaction between gender<br />

and prior rituximab exposure remains unknown in pts with relapsed/<br />

refractory DLBCL undergoing autologous stem cell transplantation (ASCT).<br />

Methods: We conducted a retrospective cohort study <strong>of</strong> 320 consecutive pts<br />

who underwent ASCT for relapsed/refractory DLBCL at our center from<br />

01/1998 to 12/2010. Relapse-free survival (RFS) and overall survival (OS)<br />

were estimated by Kaplan-Meier method and by Cox proportional hazards<br />

regression analysis in SAS 9.1 (Cary, NC). Results: Among all pts (median<br />

age�54 yrs (IQR, 45-61); 92% Caucasians) 210 (66%) were males and<br />

110 (34%) were females. 85 pts (27%) received more than two prior<br />

chemotherapy regimens. 174 pts (55%) had pre-ASCT exposure to<br />

rituximab as a part <strong>of</strong> induction or salvage regimens. Preparative regimen<br />

for ASCT uniformly consisted <strong>of</strong> Bu/Cy/VP-16. In the stratified univariate<br />

analysis <strong>of</strong> pts treated with rituximab prior to the ASCT, male pts had<br />

inferior RFS (p�0.02; hazard ratio [HR]�1.86, 95% CI, 1.09-3.18) and<br />

OS (p�0.03; HR�1.94, 95% CI, 1.05-3.58). In pts who did not receive<br />

rituximab prior to the ASCT, no difference in RFS or OS according to gender<br />

was observed (all p�0.4). Gender disparity among rituximab recipients was<br />

even more evident for both RFS (p�0.004; HR�2.22, 95% CI, 1.28-<br />

3.83) and OS (p�0.005; HR�2.42, 95% CI, 1.3-4.5) in the multivariable<br />

analyses controlling for age at transplant, number <strong>of</strong> prior chemotherapies,<br />

and pre-ASCT disease response. Conclusions: Our data support an emerging<br />

concern that male patients with DLBCL have inferior outcomes when<br />

treated with rituximab. The present study has extended this observation to<br />

pts with relapsed/refractory DLBCL treated with ASCT.<br />

Lymphoma and Plasma Cell Disorders<br />

523s<br />

8054 General Poster Session (Board #33C), Mon, 1:15 PM-5:15 PM<br />

Final results <strong>of</strong> phase I/II trial <strong>of</strong> vorinostat in combination with cyclophosphamide,<br />

etoposide, prednisone, and rituximab (R-CVEP) for elderly<br />

patients with relapsed/refractory diffuse large B-cell lymphoma (DLBCL).<br />

Presenting Author: David J. Straus, Memorial Sloan-Kettering Cancer<br />

Center, New York, NY<br />

Background: Standard treatment <strong>of</strong> relapsed/refractory DLBCL in elderly<br />

patients who are not candidates for autologous stem cell transplantation<br />

(auSCT) has not been established. Cyclophosphamide (C), etoposide (E),<br />

prednisone (P) and procarbazine (CEPP) has been used by many clinicians<br />

based on limited data (Blood 76: 1293-98, 1990). Vorinostat (V) is a<br />

histone deacetylase inhibitor that is approved for relapsed cutaneous T-cell<br />

lymphoma and has activity in B-cell lymphomas. This trial defined the<br />

maximum tolerated dose (MTD) <strong>of</strong> V added to standard therapy and<br />

determined the response rate <strong>of</strong> this combination. Methods: Patients �age<br />

60 with relapsed/refractory DLBCL not candidates for auSCT were enrolled<br />

on R-CVEP (R 375mg/m2 IV, d1; C 600mg/m2 d1 and 8, E 70mg/m2 IV d1,<br />

140mg/m2 d2 and 3; V PO and Pred 60mg/m2 PO d1-10) every 28 days for<br />

6 cycles. In the phase I component V was administered at doses <strong>of</strong><br />

300mg/d or 400mg/d for 10 days. The phase I was a 3 � 3 design and the<br />

phase II a two stage design requiring 8/20 complete responses (CR) for<br />

expansion. Assessment <strong>of</strong> response utilized end-<strong>of</strong>-treatment positron<br />

emission tomography (PET) (JCO 25: 579-86, 2007). Quality <strong>of</strong> life (QOL)<br />

was measured with the FACT-Lym v.4. Results: 27 pts. were enrolled. 1<br />

died before treatment. For 26 pts: median age 76 yrs. (69-88), 14 females<br />

and 12 males, baseline PS (ECOG) 1 (0-2). Median follow-up for survivors:<br />

9.2 mo. Phase I: 6 pts. at 300mg/d (no dose-limiting toxicity-DLT), 6 pts.<br />

at 400mg/d (2 grade 3 neutropenia � DLT). MTD 300mg/d x 10d. For 20<br />

pts. at V 300mg/m2 (6 phase I � 14 phase II): 2 <strong>of</strong>f study for toxicity, 1<br />

withdrew consent, 6 CR (30%), 5 partial response (PR) (25%), 6<br />

progressed (30%). Phenotypic overall responses (OR): germinal center<br />

(GC) 4/8 (2 CR), non-GC 6/10 (3 CR), transformed CLL 1/2 (1 CR). Median<br />

progression-free survival: 10 mo. QOL results will be presented. Conclusions:<br />

OR rate for V added to conventional chemotherapy and R was 55% (CR<br />

30%, PR 25%) in relapsed/refractory DLBCL in elderly pts. not candidates<br />

for auSCT. This could provide a baseline for comparison with future clinical<br />

trials in this understudied population.<br />

8056 General Poster Session (Board #33E), Mon, 1:15 PM-5:15 PM<br />

Effect <strong>of</strong> short-duration chemoimmunotherapy plus radioimmunotherapy<br />

on response rates in relapsed follicular lymphoma: A U.K. NCRI Lymphoma<br />

Group Study, CR UK/07/038. Presenting Author: Timothy M Illidge,<br />

Manchester Cancer Research Centre, Manchester, United Kingdom<br />

Background: Radioimmunotherapy (RIT) and rituximab (R) maintenance<br />

have been investigated after chemotherapy (CT) or R-CT in first line<br />

treatment <strong>of</strong> follicular lymphoma (FL), but less is known about the efficacy<br />

<strong>of</strong> RIT after R-CT in relapsed FL. This phase II study was conducted to<br />

examine efficacy and safety <strong>of</strong> abbreviated R-CT followed by 90Y Ibritumomab<br />

tiuxetan in patients with recurrent FL. Methods: Patients (pts) had<br />

FL, at 1st or 2nd relapse after response to R-CT or CT alone. All had<br />

WHO/ECOG performance status �2, and adequate bone marrow (BM),<br />

kidney, liver and cardiac function. BM involvement by lymphoma had to be<br />

�25% prior to infusion <strong>of</strong> 90Y Ibritumomab tiuxetan, but could be higher at<br />

study entry. Pts received 3 cycles <strong>of</strong> either R-CHOP or R-CVP, followed by<br />

90Y Ibritumomab tiuxetan (15MBq/Kg, maximum 1200 MBq). No maintenance<br />

R was given. Results: 52 pts were enrolled from 6/2008 to 7/2010.<br />

Median age was 62 years (range 31-87). 46% had high FLIPI score at<br />

entry, 31% intermediate. 80% were at 1st recurrence. Median duration <strong>of</strong><br />

best prior remission was 27.7 months. 65% pts had previously received R<br />

and 25% doxorubicin. 71% pts had R-CHOP and 29% R-CVP, prior to RIT.<br />

Overall response rate (ORR) after 3 cycles R-CT was 94% (CR/CRu 10%)<br />

and after RIT ORR 96% (CR/CRu 28%). Grade 3/4 thrombocytopenia<br />

occurred in 58% pts (median duration 16 days): 3/52 pts after R-CT and<br />

27/52 after RIT. Grade 3/4 neutropenia occurred in 62% pts: 15/52 pts<br />

after R-CT and 27/52 after RIT. 5 pts had a total 8 grade 3 infections, only<br />

1 attributable to RIT. 6 pts required platelets and 4 red cell transfusions. 1<br />

pt developed myelodysplasia 10 months after 90Y Ibritumomab tiuxetan,<br />

but no other second malignancy was recorded. PFS was 31.4 months (95%<br />

CI 15.8 - not reached). There was no difference in PFS according to FLIPI<br />

score at entry or reinduction CT (R-CHOP vs R-CVP). Pts with CR/CRu<br />

following RIT showed a trend to improved PFS compared to those with PR<br />

(12 month PFS 90% versus 63%, p�0.06). Conclusions: Abbreviated R-CT<br />

and consolidation with 90Y Ibritumomab tiuxetan is an option for pts with<br />

recurrent FL, with responses <strong>of</strong> comparable duration to those seen after a<br />

full course R-CHOP.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


524s Lymphoma and Plasma Cell Disorders<br />

8057 General Poster Session (Board #33F), Mon, 1:15 PM-5:15 PM<br />

Phase I study cohort evaluating an optimized administration schedule <strong>of</strong><br />

SAR3419, an anti-CD19 DM4-loaded antibody drug conjugate (ADC), in<br />

patients (pts) with CD19 positive relapsed/refractory b-cell non-Hodgkin’s<br />

lymphoma (NCT00796731). Presenting Author: Bertrand Coiffier, Hospices<br />

Civils de Lyon Sud, Pierre-Bénite, France<br />

Background: The recommended dose (RD) <strong>of</strong> SAR3419 administered<br />

intravenously every 3 weeks (q3w) for 6 cycles is 160 mg/m2 and is 55<br />

mg/m2 when administered weekly (q1w) for 8-12 doses. Reversible corneal<br />

deposits were dose limiting (DLT) in the q3w schedule. The q1w schedule<br />

was well tolerated and active. However, based on the occurrence <strong>of</strong><br />

late/cumulative adverse events (AE) at the RD supported by pharmacokinetic<br />

(PK) analyses showing ADC plasma accumulation after 4 weekly<br />

doses, an optimized schedule consisting <strong>of</strong> 4 weekly doses followed by 4<br />

bi-weekly doses at the RD was tested. Methods: The q1w study was<br />

extended to treat 25 pts with the optimized schedule for 8 to 12 doses.<br />

Results: Twenty-one pts were evaluable. Main histologies were diffuse large<br />

B-cell (DLBCL) (9; 43%) and follicular (6; 29%). Median number <strong>of</strong> prior<br />

regimens was 2 [1-8], 6 pts received prior autologous transplantation and<br />

95% <strong>of</strong> pts were Ann Arbor stage III-IV at study entry. Median number <strong>of</strong><br />

doses received was 8 as planned with a median relative dose intensity <strong>of</strong><br />

1.0 [0.8-1.0]. Most frequent AEs were asthenia (1 pt with grade 3) and<br />

gastrointestinal disorders in 7 pts each. No AE fulfilled the defined DLT<br />

criteria. Reversible grade 1 blurred vision/corneal event occurred in 1 pt.<br />

Grade 3-4 haematological toxicities were minor consisting <strong>of</strong> non complicated<br />

neutropenia in 4 pts, thrombocytopenia in 2 pts and anemia in 1 pt<br />

with no transfusion support. Six (29%) pts, among them 3 with DLBCL,<br />

achieved an objective response including 3 CRu (1 in a pt refractory to last<br />

regimen). In addition, 9 (43%) pts had stable disease. Response duration<br />

was [8-35�] weeks, 5 pts still responding at the cut-<strong>of</strong>f date. Conclusions:<br />

The optimized administration schedule shows an improved safety pr<strong>of</strong>ile<br />

compared to prior tested schedules. The clinical efficacy is preserved<br />

essentially in aggressive lymphoma. The optimized schedule is being<br />

assessed in 2 phase II studies evaluating SAR3419 either as a single agent<br />

or in combination with rituximab in pts with DLBCL histology.<br />

8059 General Poster Session (Board #33H), Mon, 1:15 PM-5:15 PM<br />

High affinity CD3 RECRUIT TandAb for T cell-mediated lysis <strong>of</strong> CD19� tumor B cells. Presenting Author: Eugene Zhukovsky, Affimed Therapeutics<br />

AG, Heidelberg, Germany<br />

Background: CD19 is expressed from early B cell development to the<br />

differentiation into plasma cells and is an attractive target for B cell<br />

malignancies either lacking CD20 expression or refractory to anti-CD20<br />

antibody therapies. T cells are potent tumor killing effector cells that are<br />

not recruited by native antibodies. The CD3 RECRUIT-TandAb AFM11, a<br />

human bispecific tetravalent antibody with two binding sites for both CD3<br />

and CD19, is a novel therapeutic for the treatment <strong>of</strong> NHL that harnesses<br />

the cytotoxic nature <strong>of</strong> T cells. Methods: A bispecific anti-CD19/anti-CD3<br />

tetravalent TandAb with humanized and affinity matured variable domains<br />

was constructed. The TandAb’s binding, T-cell mediated cytotoxic activity,<br />

and cytokine release were characterized in a panel <strong>of</strong> in vitro assays. In vivo<br />

efficacy was evaluated in a murine NOD/scid xenograft model reconstituted<br />

with human PBMC. Results: AFM11 mediates highly potent target tumor<br />

cell lysis in cytotoxicity assays: EC 50 values are low to sub-picomolar range<br />

in a panel <strong>of</strong> CD19 � cell lines and primary B-CLL tumor cells. The cytotoxic<br />

activity <strong>of</strong> tetravalent AFM11 is superior to that <strong>of</strong> alternative bivalent<br />

antibody formats possessing only a single binding site for both CD19 and<br />

CD3. High affinity binding <strong>of</strong> AFM11 to CD19, and more so to CD3 (low to<br />

sub-nanomolar Kd), is essential for efficacious T cell recruitment. The high<br />

affinity bivalent binding <strong>of</strong> AFM11 to CD3 does not trigger T cell activation<br />

in the absence <strong>of</strong> CD19 � target cells in functional in vitro assays. AFM11<br />

activates T cells only in the presence <strong>of</strong> its targets and mediates lysis while<br />

sparing antigen-negative bystanders. AFM11 induces down-modulation <strong>of</strong><br />

the CD3/TCR complex in the absence <strong>of</strong> target cells and at high concentrations.<br />

Also, AFM11-treated T cells can be re-engaged for target cell lysis.<br />

These features <strong>of</strong> AFM11-induced T cell activation may contribute additional<br />

safety with no compromise <strong>of</strong> efficacy. Finally, AFM11 demonstrates<br />

a robust dose-dependent inhibition <strong>of</strong> subcutaneous Raji tumors in mice.<br />

Conclusions: AFM11 is a novel highly efficacious drug candidate for the<br />

treatment <strong>of</strong> B cell malignancies with an advantageous safety pr<strong>of</strong>ile.<br />

8058 General Poster Session (Board #33G), Mon, 1:15 PM-5:15 PM<br />

Hodgkin’s disease and HIV infection (HD-HIV): Prognostic factors in 596<br />

patients (pts) within the group <strong>of</strong> European Cooperation on AIDS and<br />

Tumors (GECAT). Presenting Author: Michele Spina, National Cancer<br />

Institute, Aviano, Italy<br />

Background: Hodgkin’s disease (HD) is the most common non-AIDS<br />

defining tumour diagnosed in HIV setting. The introduction <strong>of</strong> highly active<br />

antiretroviral therapy (HAART) has opened a new prospective in the<br />

treatment <strong>of</strong> pts with HD-HIV as the better control <strong>of</strong> the underlying HIV<br />

infection allows the use <strong>of</strong> more aggressive chemotherapy regimens,<br />

including high dose chemotherapy. However, up to now prognostic factors<br />

on overall survival (OS) or time to treatment failure (TTF) have not yet been<br />

identified. Methods: In order to identify prognostic factors, we analyzed data<br />

on 596 pts with HD-HIV diagnosed and treated in 90 different Institution <strong>of</strong><br />

6 European countries from October 1983 to March 2010. All factors were<br />

analyzed for OS and TTF. Results: 86% <strong>of</strong> pts were male and the median<br />

CD4 cell count was 224/dl (range 3-1274); 52% <strong>of</strong> pts had mixed<br />

cellularity subtype, stages III-IV were diagnosed in 72% <strong>of</strong> cases and 55%<br />

<strong>of</strong> pts had extranodal involvement (bone marrow 35%, spleen 21%, liver<br />

14%). The table summarizes the results <strong>of</strong> multivariate analysis.<br />

Conclusions: We identified a new “European Score” for HD-HIV able to<br />

predict different outcomes in these patients. This score should be considered<br />

for future prospective studies.<br />

Factors Overall survival Time to treatment failure<br />

IPS < 2 1 1<br />

IPS > 2 2.33 (1.61-3.39) p�0.0001 1.57 (1.09-2.26) p�0.02<br />

CD4 > 200 1 1<br />

CD4 < 200 1.63 (1.16-2.29) p�0.005 1.43 (1.02-2.01) p�0.04<br />

European Score<br />

0 1 1<br />

1 2.06 (1.40 - 3.02) 1.64 (1.17 - 2.30)<br />

2 3.08 (2.13 - 4.45) p�0.001 2.31 (1.66 - 3.20) p�0.001<br />

8060 General Poster Session (Board #34A), Mon, 1:15 PM-5:15 PM<br />

Characteristics and outcomes <strong>of</strong> extranodal NK/t-cell lymphoma (ENKL): A<br />

North <strong>American</strong> (NA) multi-institutional experience. Presenting Author:<br />

Lauren Shizue Maeda, Stanford University Medical Center, Stanford, CA<br />

Background: ENKL is a rare and aggressive subtype <strong>of</strong> peripheral T-cell<br />

lymphoma. Due to its geographic predilection there is a paucity <strong>of</strong> data on<br />

clinical experiences from non-Asian countries. The purpose <strong>of</strong> this study<br />

was to analyze characteristics and outcomes <strong>of</strong> patients (pts) with ENKL<br />

identified from major academic centers in NA. Methods: Pts with newly<br />

diagnosed CD56� ENKL were retrospectively identified. Analyses included<br />

disease characteristics, ethnicity, therapy, and outcomes. Results: 115 pts<br />

(63.5% Caucasian, 20% Asian, 16.5% other) were identified across 10<br />

centers diagnosed between 5/1990-5/2011 (Era 1: pre-2000, n�16; Era<br />

2: 2000-2005, n�45; Era 3: post-2005, n�54). Median age was 52 years<br />

(19-88). 75 (65%) had stage I/II disease and were treated with combined<br />

modality therapy (CMT) n�48, chemotherapy (CT) n�14 or radiotherapy<br />

(RT) n�14. 40 pts had stage III/IV disease and were treated with CT<br />

(n�23), CMT (n�12) or RT (n�5). CT regimens used alone or in CMT were<br />

either anthracycline-based (n�68) or other (n�29). 63% <strong>of</strong> stage I/II pts<br />

and 40% with stage III/IV achieved complete remission (CR). 30 pts<br />

underwent a stem cell transplant (SCT); 14 in first CR and 16 at<br />

progression/relapse (autologous, n�21; allogeneic, n�9). Pts with stage<br />

I/II disease had a better progression-free survival (PFS) and overall survival<br />

(OS) compared with stage III/IV (12 vs 5.2 months (p�0.003) and 41.5 vs<br />

8.9 months (p�0.0001), respectively). For all stages, treatment with CMT<br />

compared with CT or RT alone was also associated with better PFS and OS,<br />

18.0 vs 3.9 months (p�0.0001), and 41.5 vs 10.2 months (p�0.002)<br />

respectively. Non-anthracycline-based regimens were associated with better<br />

PFS (p�0.001) and OS (p�0.045). No survival differences were seen<br />

between Asian and non-Asian pts. Conclusions: This series represents one<br />

<strong>of</strong> the largest experiences <strong>of</strong> ENKL in NA. Our data are consistent with<br />

Asian studies in: 1) majority <strong>of</strong> pts present with early stage disease; 2)<br />

overall poor outcome; 3) superiority <strong>of</strong> CMT and non-anthracycline regimens.<br />

Advances in understanding biology and international collaborative<br />

efforts are required to improve outcome in this rare entity.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


8061 General Poster Session (Board #34B), Mon, 1:15 PM-5:15 PM<br />

Determinants <strong>of</strong> and outcomes associated with chemoimmunotherapy dose<br />

density in elderly subjects with diffuse large B-cell lymphoma (DLBCL).<br />

Presenting Author: Usha S. Perepu, University <strong>of</strong> Iowa Hospitals and<br />

Clinics, Iowa City, IA<br />

Background: RCHOP is the standard first line therapy for DLBCL but elderly<br />

patients <strong>of</strong>ten receive suboptimal doses. Relative dose intensity (RDI) less<br />

than 70-90% has previously been associated with inferior survival in CHOP<br />

treated patients. We now investigate the clinical determinants <strong>of</strong> immunochemotherapy<br />

dose delivery and comparative effectiveness <strong>of</strong> dose intensity<br />

on lymphoma-specific outcomes in persons over the age <strong>of</strong> 60.<br />

Methods: SPORE MER (Molecular Epidemiology Resource) is a prospectively<br />

accrued observational study in which University <strong>of</strong> Iowa and Mayo<br />

Clinic patients with a diagnosis <strong>of</strong> Non Hodgkins lymphoma have been<br />

enrolled since 2003. We reviewed data from the Iowa patients along with<br />

medical records from DLBCL patients over the age <strong>of</strong> 60 treated with<br />

anthracycline based chemo immunotherapy consecutively enrolled up<br />

through to December 2009. Statistical tests <strong>of</strong> associations between RDI<br />

and clinicopathologic factors, as well as survival outcomes, were performed.<br />

Results: We identified 92 patients over the age <strong>of</strong> 60. The median<br />

age was 70. 47 subjects experienced at least 1 dose reduction, 26 subjects<br />

had dose delays, and 20 subjects were unable to complete therapy. RDI<br />

ranged from 0.30-1.70 (median 0.92) for doxorubicin and from 0.29-1.70<br />

(median 0.97) for cyclophosphamide. 24 and 19% <strong>of</strong> subjects received �<br />

80% <strong>of</strong> RDI for dox and cy respectively. Age (p� 0.001 for both dox and cy)<br />

and performance status (p�0.04 for dox) but not number <strong>of</strong> medications,<br />

ACE27 co-morbidity score, body surface area, nor need for acute care visits<br />

were associated with decreased RDI. Adjusted for IPI, RDI did NOT have a<br />

significant effect on event-free or lymphoma-specific survival. Conclusions:<br />

Among clinical variables, age is the dominant variable associated with<br />

delivery dose intensity for chemotherapy in elderly patients treated for<br />

DLBCL; and among elderly patients receiving chemo immunotherapy, we<br />

did not find an association between dose intensity and lymphoma-specific<br />

outcomes.<br />

Doxorubicin Cyclophosphamide<br />

Correlation p value Correlation p value<br />

Age at Rx -0.348 .0007 -0.358 .0006<br />

BMI 0.097 .359 0.205 .055<br />

#<strong>of</strong>meds 0.050 .636 -0.007 .942<br />

LVEF 0.096 .458 0.011 .933<br />

8063 General Poster Session (Board #34D), Mon, 1:15 PM-5:15 PM<br />

Progression-free survival for subsequent relapses in patients with peripheral<br />

T-cell lymphoma (PTCL). Presenting Author: Steven M. Horwitz, Memorial<br />

Sloan-Kettering Cancer Center, New York, NY<br />

Background: In B-cell lymphoma, overall response (ORR) is lower and progression<br />

free survival (PFS) is <strong>of</strong>ten shorter with each subsequent progression.<br />

However, this has not been described for PTCL. Methods: We undertook a<br />

retrospective analysis in two U.S. centers (MSKCC and UNMC) and in GELA<br />

studies. All data for PTCL were reviewed to collect data for each line <strong>of</strong> therapy:<br />

treatment, response, PFS, date <strong>of</strong> next treatment, and survival. When date <strong>of</strong><br />

progression could not be defined we calculated PFS as the time to next<br />

treatment. Pts who did not progress, died during first line, or were untreated at<br />

progression 1 were excluded. Data were collected for 4 lines <strong>of</strong> treatment, front<br />

line and 3 progressions. Results: 254 pts were identified. 205 were analyzed (49<br />

excluded for missing data): GELA 116, MSKCC 51, and UNMC 37. Diagnoses<br />

were PTCL NOS in 34% and 60%, AITL in 49% and 35%, ALCL Alk- in 3% and<br />

5%, and others in 15% and 18% for GELA and US centers respectively. 67%<br />

were male, median age was 51 vs 58 y. Advanced stage was more frequent in<br />

GELA (79% vs. 56% for US); baseline IPI was �2 in 90% vs 71%. Multidrug<br />

regimens were given in 99% and 80% for front line, 67% and 66% for 2nd line,<br />

61% and 50% for 3rd line, 53% and 54% for 4th line, for GELA and US.<br />

Significantly fewer pts received each subsequent line <strong>of</strong> therapy with 205 pts<br />

receiving 2nd line, 96 pts 3rd line and 38 pts 4th line. No pt �65 y received 4th<br />

line treatment. Responses and PFS are listed in the table. Conclusions: For pts<br />

with PTCL, similar to B-cell lymphomas, ORR and PFS decrease with each line <strong>of</strong><br />

therapy. When evaluating the activity <strong>of</strong> therapies in relapsed PTCL, line <strong>of</strong><br />

therapy is a consideration and this series provides a benchmark for comparison.<br />

Based on this dataset, it is possible that better responses will be seen as new<br />

agents are moved into earlier treatment paradigms.<br />

GELA data U.S. data All<br />

1st line (front line)<br />

- ORR<br />

-CR<br />

2nd line<br />

- ORR<br />

-CR<br />

- Median PFS<br />

- 6-m event-free<br />

3rd line<br />

- ORR<br />

-CR<br />

- Median PFS<br />

- 6-m event-free<br />

4th line<br />

- ORR<br />

-CR<br />

- Median PFS<br />

- 6-m event-free<br />

Abbreviation: NA: not available.<br />

.<br />

71%<br />

57%<br />

.<br />

44%<br />

35%<br />

13 m<br />

74%<br />

.<br />

23%<br />

13%<br />

Not reached<br />

63%<br />

.<br />

6%<br />

6%<br />

6m<br />

38%<br />

.<br />

64%<br />

43%<br />

.<br />

52%<br />

28%<br />

27 m<br />

66%<br />

.<br />

32%<br />

18%<br />

2m<br />

36%<br />

.<br />

38%<br />

9%<br />

NA<br />

NA<br />

Lymphoma and Plasma Cell Disorders<br />

.<br />

68%<br />

51%<br />

.<br />

47%<br />

32%<br />

15 m<br />

71%<br />

.<br />

27%<br />

15%<br />

8m<br />

50%<br />

.<br />

24%<br />

8%<br />

NA<br />

NA<br />

525s<br />

8062 General Poster Session (Board #34C), Mon, 1:15 PM-5:15 PM<br />

Biweekly regimen <strong>of</strong> nonpegylated liposomal doxorubicin with cyclophosphamide,<br />

vincristine, and prednisone plus rituximab (R-COMP-14) as primary<br />

treatment for diffuse large B-cell lymphoma (DLBCL): Long-term follow-up<br />

<strong>of</strong> a phase II study. Presenting Author: Joaquin Herrero, Hospital General<br />

de Alicante, Alicante, Spain<br />

Background: To evaluate the efficacy and toxicity <strong>of</strong> dose-dense biweekly<br />

schedule <strong>of</strong> (R-COMP-14) in patients newly diagnosed <strong>of</strong> agrressive diffuse<br />

B cell lymphomas (DLBCL). Methods: In this single-arm, open-label,<br />

multicenter trial, 60 pts were included between 2004 to 2008. Received<br />

rituximab (375 mg/m2 ) D1 � cyclophosphamide (750 mg/m2 )D1 �<br />

vincristine (1.4 mg/m2 ; max. 2 mg)D1 � prednisone (100 mg/d D1 and<br />

D5) and non-pegylated liposomal doxorubicin (50 mg/m2 ) every two weeks.<br />

Response was assessed at cycle 3, and patients with complete or partial<br />

response received 5 additional courses. Granulocyte colony-stimulating<br />

factor (G-CSF). Pegfilgrastim was administered on day 2. The primary<br />

efficacy endpoint was (CR) and objective response rate (ORR). Survival<br />

follow-up data were updated. Results: 59 evaluable patients with a median<br />

age <strong>of</strong> 50 years (21-65) were analyzed. <strong>Clinical</strong> Characteristics: 22 Pts<br />

stage I-II (aaIPI�1 (37%), 17 (29%) stage III, and 19 (32%) in stage IV.<br />

aaIPI�1 41(67%), aaIPI2/3� 18(33%).LVEF basal: 65,5[50-93]. Extranodal<br />

disease: 42%. B symptoms: 44.1%. The mean calculated dose<br />

intensities <strong>of</strong> cyclophosphamide, non-pegylated liposomal doxorubicin,<br />

vincristine and rituximab were 98,7%, 98,7%, 76,1% and 98,3% respectively.<br />

Among 60 eligible patients (96,7% completed six cycles and 74,4%<br />

completed all eight cycles. ORR was 81%, and CR rate <strong>of</strong> 54,2%. IC 95%<br />

[40,7-67,8]. The main toxicity was neutropenia Gr�III-IV/ and febrile<br />

neutropenia in 20% <strong>of</strong> patients. Neurotoxicity Gr�III-IV in 3,4%. No<br />

cardiac toxicity Gr: III-IV was reported. No toxic deaths. After a median<br />

follow-up <strong>of</strong> 25-64m(44m) the 5-year overall survival (OS), event-free<br />

survival, (EFS) and disease free survival (DFS) were 80%, 67%, and 77%<br />

respectively. Forty two patients (71%) had (�60 y) and med. OS(p�0,017)<br />

and med EFS(p�0,014) was 72,3% and 68,1 in aaIPI�1 and 50,4% and<br />

33,8 in aaIPI�1. Conclusions: Dose dense R-COMP-14 is an effective<br />

regimen in patients with (DLBCL) comparable o R- CHOP-14. Good<br />

tolerability pr<strong>of</strong>ile with no Gr: III-IV cardiac toxicity or reduction <strong>of</strong> LVEF.<br />

8064 General Poster Session (Board #34E), Mon, 1:15 PM-5:15 PM<br />

MLN9708, an investigational proteasome inhibitor, in patients (pts) with<br />

relapsed/refractory lymphoma: Emerging data from a phase I doseescalation<br />

study. Presenting Author: Peter Martin, Weill Cornell Medical<br />

College, New York, NY<br />

Background: MLN9708 is a reversible, orally bioavailable, specific 20S<br />

proteasome inhibitor. This study (NCT00893464) studied the safety and<br />

determined the MTD <strong>of</strong> IV MLN9708 in pts with relapsed/refractory<br />

lymphoma, and characterized pharmacokinetics (PK) and pharmacodynamics<br />

(PD). Methods: Pts aged �18 yrs who had failed �2 chemotherapeutic<br />

regimens received IV MLN9708 on days 1, 8, and 15 <strong>of</strong> 28-day cycles until<br />

disease progression or unacceptable toxicity. One pt was enrolled at the<br />

0.125 mg/m2 starting dose; dose doubling proceeded with 1 pt at each<br />

dose up to 1.0 mg/m2 . Dose escalation occurred in �40% increments<br />

using a standard 3�3 scheme based on DLT occurrence in cycle 1. Blood<br />

samples were collected at multiple time points after dosing on days 1 and<br />

15 <strong>of</strong> cycle 1 for PK/PD analyses. Results: At data cut-<strong>of</strong>f (Dec 1 2011), 21<br />

pts had been enrolled and treated: 1 each at 0.125, 0.25, 0.5 and 1<br />

mg/m2 , 4 at 1.4 mg/m2 , 7 at 1.76 mg/m2 , and 6 at 2.34 mg/m2 . Median<br />

age was 57 yrs (range 23–78); 57% were male. Median number <strong>of</strong> prior<br />

therapies was 5; 29% had prior radiation, 24% prior stem cell transplant.<br />

Histologies included T-cell lymphoma (n�5), Hodgkin lymphoma (n�3),<br />

follicular lymphoma (n�2), DLBCL (n�1) and other indolent B-cell<br />

lymphoma (n�7). Pts had received a median <strong>of</strong> 2 cycles (range 1–22); 2<br />

DLTs were seen (neutropenia at 1.76 and 2.34 mg/m2 ). All pts experienced<br />

drug-related AEs, including fatigue (48%), nausea (29%), diarrhea (29%),<br />

pyrexia, thrombocytopenia, and vomiting (each 24%). 43% had drugrelated<br />

grade �3 AEs, 1 pt discontinued due to drug-related grade 3<br />

neutropenia (2.34 mg/m2 ). Three pts had drug-related peripheral neuropathy<br />

(1 grade 1, 2 grade 2). There were no on-study deaths. Of 18<br />

response-evaluable pts, 3 achieved PR (including 2 who remain in<br />

response and on-study for �1 yr) and 4 SD. PK analyses showed linear PK<br />

(0.5–2.34 mg/m2 ) and a terminal half-life <strong>of</strong> ~6–9 days. There was a<br />

dose-dependent increase in maximal whole blood 20S proteasome inhibition.<br />

Conclusions: These data suggest IV MLN9708 is generally well<br />

tolerated, with infrequent PN, and is clinically active in pretreated<br />

lymphoma pts. The trial is ongoing and updated data will be presented.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


526s Lymphoma and Plasma Cell Disorders<br />

8065 General Poster Session (Board #34F), Mon, 1:15 PM-5:15 PM<br />

Phase I/II study <strong>of</strong> MEDI-551, a humanized monoclonal antibody targeting<br />

CD19, in subjects with relapsed or refractory advanced B-cell malignancies.<br />

Presenting Author: Trishna Goswami, MedImmune, Gaithersburg, MD<br />

Background: Novel B-cell targeting agents, including monoclonal antibodies<br />

such as rituximab, are among recent advances in treatment <strong>of</strong> B-cell malignancies.<br />

New approaches are needed for patients progressing after rituximab-based<br />

therapies. MEDI-551 is an afucosylated monoclonal antibody targeting CD-19, a<br />

B-cell restricted transmembrane protein with enhanced affinity and antibodydependent<br />

cellular cytotoxicity. Methods: Pts with relapsed or refractory follicular<br />

lymphoma (FL), diffuse large B-cell lymphoma (DLBCL), chronic lymphocytic<br />

leukemia, or multiple myeloma received single agent MEDI-551 at dosages<br />

ranging from 0.5 mg/kg to 12 mg/kg via intravenous infusion over 28-day cycles;<br />

cohorts 1-6 received 0.5, 1, 2, 4, 8, and 12 mg/kg, respectively. Results: 25 pts<br />

were enrolled in the phase I portion Jun 2010–Aug 2011. No maximum<br />

tolerated dose (MTD) was achieved. Most AEs were grade 1/2 with doseindependent<br />

frequency and severity (Table). Six pts had grade 3 toxicities<br />

including tumor lysis syndrome, infusion reaction, thrombocytopenia, and<br />

neutropenia, or grade 4 neutropenia. No grade 5 AEs were seen. All pts<br />

recovered. Three partial responses (PR) and 2 complete responses (CR) were<br />

seen in DLBCL and FL pts at 0.5, 4, and 8 mg/kg. Activity included a CR lasting<br />

9 mo. in a FL pt in cohort 1, who is currently being retreated with MEDI-551 on<br />

relapse. Conclusions: MEDI-551 demonstrated a safety pr<strong>of</strong>ile warranting further<br />

study and showed no MTD reached at the highest dose studied. Anti-tumor<br />

activity is suggested by the responses achieved across dose levels. Phase II is<br />

currently enrolling subjects. This study is funded by MedImmune, LLC.<br />

Frequency <strong>of</strong> treatment emergent Medi-551-related adverse events.<br />

Preferred term Total events (N�37)<br />

Infusion-related reaction 6 (16.2%)<br />

Anaemia 3 (8.1%)<br />

Nausea 3 (8.1%)<br />

Thrombocytopenia 3 (8.1%)<br />

Aspartate aminotransferase increased 2 (5.4%)<br />

Blood immunoglobulin M decreased 2 (5.4%)<br />

Chills 2 (5.4%)<br />

Fatigue 2 (5.4%)<br />

Headache 2 (5.4%)<br />

Hypertension 2 (5.4%)<br />

Hypotension 2 (5.4%)<br />

Neutropenia 2 (5.4%)<br />

Pyrexia 2 (5.4%)<br />

Alanine aminotransferase increased 1 (2.7%)<br />

8067 General Poster Session (Board #34H), Mon, 1:15 PM-5:15 PM<br />

Comparison <strong>of</strong> the international prognostic factors index (IPI) with the<br />

absolute monocyte and lymphocyte prognostic index (AMLPI) for patients<br />

(Pts) with diffuse large b-cell lymphoma (DLBCL) receiving R-CHOP.<br />

Presenting Author: Nicolas Batty, Roswell Park Cancer Institute, Buffalo,<br />

NY<br />

Background: We studied the value <strong>of</strong> a proposed prognostic index (PI)<br />

generated by baseline absolute monocyte (AMC) and lymphocyte (ALC)<br />

counts for pts with DLBCL, using values as previously reported (Leukemia<br />

25:1502-9, 2011). Methods: From 03/07 to 01/09, 245 consecutive pts<br />

with untreated DLBCL receiving standard R-CHOP from the MDACC<br />

database were evaluated. Baseline AMC and ALC were retrospectively<br />

recorded. High AMC (�610/uL) and a low ALC (�1000/uL) were examined<br />

as dichotomized variables for progression-free (PFS) and overall survival<br />

(OS). An AMLPI was generated, stratifying pts into 3 risk groups (RGs):<br />

low-(AMC �610/uL and ALC �1000/uL), intermediate-(AMC �610/uL or<br />

ALC �1000/uL), and high-risk(AMC �610/uL and ALC �1000/uL). The<br />

prognostic effect <strong>of</strong> the AMLPI and the IPI were examined by multivariate<br />

analysis (MVA). Results: Ninety (37%) had high AMC and 71 (29%) had low<br />

ALC. By univariate analysis, a high AMC was associated with inferior PFS<br />

(p�0.01) and OS (p�0.03). The frequencies <strong>of</strong> AMLPI RGs were: low-105<br />

pts (43%), intermediate-119 (48%), and high risk-21 (9%). With a median<br />

follow-up <strong>of</strong> 22 months (range �1-42), 3-year PFS and OS rates for these<br />

RGs were 80%, 61%, and 46% (p�0.007) and 92%, 76%, and 60%<br />

(p�0.006), respectively. Three-year PFS rates for IPI 0-2 and 3-5 RGs<br />

were 73% and 58%, respectively (p�0.0004); comparable OS rates were<br />

88% and 68%(p�0.0001). For pts with IPI 0-2, 1-year PFS rates for<br />

AMLPI low, intermediate, and high RGs were 92%, 89% and 80%<br />

(p�0.022); comparable 1-year OS rates were 96%, 95% and 80%<br />

(p�0.049). By MVA, AMLPI effect (low vs. high RGs) on PFS was<br />

significant (p�0.046) as was IPI effect (0-2 vs 3-5, p�0.005); similar<br />

results were observed for OS (p�0.052 and p�0.003, respectively).<br />

Conclusions: Baseline AMC and AMLPI are significant variables for PFS and<br />

OS for pts with DLBCL receiving R-CHOP. AMLPI can identify pts with low,<br />

intermediate, and high-risk disease for PFS and OS, particularly for those<br />

with IPI 0-2. AMLPI may also add prognostic value beyond that <strong>of</strong> the IPI.<br />

8066 General Poster Session (Board #34G), Mon, 1:15 PM-5:15 PM<br />

Dose- and time-intensified ABVD without radiotherapy (RT) for advancedstage<br />

Hodgkin lymphoma (HL) with mediastinal bulky disease (MBD).<br />

Presenting Author: Gaetano Corazzelli, Hematology-Oncology and Stem<br />

Cell Transplantation Unit, Istituto Nazionale Tumori, Fondazione �G.Pascale�,<br />

Naples, Italy<br />

Background: The role <strong>of</strong> consolidation RT on MBD after upfront chemotheraphy<br />

for advanced HL is debated, also given the supradditive iatrogenic<br />

risk. We present the results achieved in the subset <strong>of</strong> patients (pts) with<br />

MBD (max width � 1/3 <strong>of</strong> thoracic diameter) accrued in a phase II study <strong>of</strong><br />

an intensified ABVD program without RT. Methods: The current analysis<br />

derives from the final evaluation <strong>of</strong> our trial for advanced HL (stage IIXB-IV)<br />

conducted from 06/2004 to 03/2010 (Russo et al, ASH 2009 abst 715).<br />

Pts were scheduled to 6 cycles <strong>of</strong> a ‘time-densified’ ABVD (3-week<br />

intercycle, drugs on days 1 and 11) with the first 4 cycles being also<br />

‘dose-intensified’: doxorubicin (ADM) 35 mg/m2 , days 1 and 11 and G-CSF<br />

on days 6-8 and 17-19. Results: Of 82 accrued pts, 39 had BMD at<br />

presentation. Median age was 29yrs (r 16-58); male 46%; stage IIB 48%,<br />

III 8%, IV 43%; B-sympt 87%, E-disease 53%; IP Score �3 51%. All pts<br />

completed the intensified program. Median actual dose intensities for<br />

ADM, bleomycin, vinblastine and dacarbazine were 23.12, 6.69, 3.96 and<br />

245 mg/week, respectively; the increase over conventional ABVD was 85%<br />

for ADM and averaged 32% for the other agents. PET2 negativity was<br />

achieved in 36/39 (92%; 95% CI 79-98), complete responses (CR) in<br />

37/39 [94%; 95% CI 82-99]. At a median f.u. <strong>of</strong> 54 mo.s (r 20-91) all pts<br />

are alive with an event-free survival <strong>of</strong> 89% (95% CI, 80-98). Events were:<br />

�CR (n�1, CS IVB), progression (n�1, CS IIIA), relapse [n�2; at 10 (CS<br />

IVA ) and 15 (CS IIB) months after treatment]; all these pts had isolated<br />

mediastinal recurrence. CTCAE v3.0 toxicity: Grade (G) 2 nail changes<br />

(31%), G2-G3 hemorrhoids (12%-3%), G3 infection (13%) and constipation<br />

(5%), G3-G4 stomatitis (7%-2%). No acute or delayed G3-G4 cardiac<br />

events, nor G3-G4 decline in pulmonary function (FEV1, DLCO,FEF25-75) were seen. Conclusions: Intensified ABVD can achieve PET2 negativity in a<br />

very high proportion <strong>of</strong> pts with MBD and ensure a long-term disease-free<br />

status even without RT. While results need confirmation on a randomized<br />

basis, the low mediastinal failure rate seems in line with recent suggestions<br />

that RT could be omitted in MBD when CR is achieved upon intensified<br />

chemotheraphy.<br />

8068 General Poster Session (Board #35A), Mon, 1:15 PM-5:15 PM<br />

Prognostic value <strong>of</strong> etoposide area under the curve (AUC) in lymphoma<br />

patients treated with BEAM regimen and ASCT: Multicenter study <strong>of</strong><br />

Groupe d’Etudes des Lymphomes de l’Adulte (GELA). Presenting Author:<br />

Olivia Bally, Centre Leon Berard, Lyon, France<br />

Background: The prospective LYMPK study primary objective was to assess<br />

the impact <strong>of</strong> etoposide pharmacokinetic (PK) parameters on toxicity and<br />

efficacy in lymphoma patients receiving the BEAM regimen (carmustine,<br />

cytarabine, etoposide and melphalan) followed by autologous stem cell<br />

transplant (ASCT). We previously showed the high inter-individual variability<br />

in etoposide PKs, defined by area under the curve (AUC) and trough<br />

concentration (Cmin), among study patients treated with the same doses<br />

/m2 (You B et al, Proc. ASCO 2008). Methods: Ninety-six patients with<br />

malignant lymphoma at 1st line (n�52) or relapse (n�44) were enrolled in<br />

5 centers. All received BEAM regimen, including high-dose etoposide (100<br />

to 200 mg/m2 bid for 4 days), followed by ASCT. Individual etoposide AUC<br />

and Cmin were estimated by population PK approach using NONMEM<br />

program. The impact <strong>of</strong> PK parameters on toxicity and survival was<br />

assessed using linear regression and univariate/multivariate analyses.<br />

Results: Data from 90 patients were assessable after a 4.2-year median<br />

follow-up. The bi-compartment model previously reported was used to<br />

characterize PK parameters (You B et al, Proc. ASCO 2008). Etoposide<br />

AUC and Cmin correlated with mucositis duration, especially for grade 3-4<br />

toxicity (p� 0.05), but not with other toxicities. Cmin had significant<br />

prognostic value regarding 5 year progression free survival (p�0.03). Five<br />

year overall survival (OS) was longer in patients with higher AUC (76% vs<br />

56%, if AUC � median, p�0.04) as it was in patients with higher Cmin<br />

(78% vs 54%, if Cmin � median, p�0.02). When assessed with available<br />

IPI prognostic factors (age; performance status; LDH and stage) using Cox<br />

analysis, the only independent prognostic factors <strong>of</strong> OS and disease<br />

specific survival were etoposide AUC (HR� 0.39, 95% CI � 0.16-0.94)<br />

and Cmin (HR � 0.32, 95% CI � 0.12-0.80). Conclusions: LYMPK study<br />

results suggest that individual etoposide systemic exposure has a strong<br />

impact on survival in lymphoma patient receiving BEAM regimen and<br />

ASCT. Given the high variability in patient AUCs, plasma concentrationbased<br />

adjustment <strong>of</strong> etoposide dose may be considered in future studies.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


8069 General Poster Session (Board #35B), Mon, 1:15 PM-5:15 PM<br />

Central nervous system involvement in T-cell lymphomas: A single center<br />

experience. Presenting Author: Ronit Gurion, Rabin Medical Center,<br />

Petach Tikva, Israel<br />

Background: Large experiences have reviewed the risk <strong>of</strong> central nervous<br />

system (CNS) involvement in diffuse large B-cell lymphoma (DLBCL), but<br />

there are limited data on CNS involvement by peripheral T cell lymphomas<br />

(PTCL). We characterized the incidence <strong>of</strong> CNS involvement, risk factors<br />

and outcome in a large single institution dataset <strong>of</strong> PTCL. Methods:<br />

Retrospective review <strong>of</strong> the T-cell lymphoma database at Memorial Sloan<br />

Kettering Cancer Center. We identified 232 patients with any subtype <strong>of</strong><br />

PTCL between 1994-2011 with a minimum 6 months <strong>of</strong> follow-up or an<br />

event defined as relapse or death. We excluded indolent forms <strong>of</strong> cutaneous<br />

T cell lymphoma. Results: Histologies included PTCL-NOS (31%), angioimmunoblastic<br />

(16.8%), anaplastic (ALCL), ALK negative (12%), ALCL, ALK<br />

positive (6%), extranodal NK/T cell lymphoma (7.3%), adult T cell<br />

leukemia/lymphoma (ATLL) (7.3%), and transformed MF (8.6%). Median<br />

age was 58 years with 59.9% men. CNS disease was found in 17 patients<br />

(7.32%). 8 (47%) had pathologic confirmation and 7 (41.2%) were<br />

clinically diagnosed. Two had other diagnoses at biopsy: DLBCL and<br />

glioblastoma. Median time to CNS involvement was 2.33 months (range,<br />

0.16 to 103.1). CNS prophylaxis was given to 24 (10.34%), primarily<br />

intrathecal methotrexate. There was no difference in CNS involvement in<br />

patients who received prophylaxis vs. those who did not: 3/24 (12.5%) vs.<br />

12/208 (5.77%) (p�0.192) respectively. Univariate analysis identified:<br />

stage III-IV (p�0.03), bone marrow involvement (p�0.018), �1 extranodal<br />

site (p�0.001), and ATLL vs. all other subtypes, 23.5% vs. 6.4%<br />

(p�0.003) as risk factors for CNS disease. On multivariate analysis, �1<br />

extranodal site (p�0.004) and high intermediate (H-I) and high (H) IPI (IPI<br />

3-5 & 4-5) were predictive for CNS involvement (p�0.05). The median<br />

survival <strong>of</strong> patients with CNS involvement was 2.628 months. Conclusions:<br />

Despite high relapse rates, PTCL carries a low risk <strong>of</strong> CNS involvement<br />

other than the ATLL subtype. As with other aggressive lymphomas, survival<br />

<strong>of</strong> patients with CNS involvement is poor and risk factors include: �1 extra<br />

nodal site and H-I-H IPI. In this dataset, prophylactic intrathecal chemotherapy<br />

does not appear to reduce the risk <strong>of</strong> CNS disease.<br />

8071 General Poster Session (Board #35D), Mon, 1:15 PM-5:15 PM<br />

Ocular adnexal lymphoma (OAL)-outcomes for 82 patients (pts) treated at a<br />

single center. Presenting Author: Craig Anthony Portell, Cleveland Clinic<br />

Foundation, Cleveland, OH<br />

Background: OALs comprise 1-2% <strong>of</strong> NHL and are extranodal marginal zone<br />

(EMZL) in 80% <strong>of</strong> cases. We present a retrospective review <strong>of</strong> 82 pts with OAL<br />

managed at the Cleveland Clinic between January 2004 and November 2011.<br />

Methods: 82 pts with NHL <strong>of</strong> the OA were identified. All biopsies were<br />

performed/reviewed at the Cleveland Clinic. Survival and relapse were estimated<br />

using the Kaplan-Meier method. Risk factors for relapse and survival were<br />

identified using Cox proportional hazards analysis. Risk factors included age at<br />

diagnosis (dx), gender, prior autoimmune disease, prior lymphoma history (hx),<br />

hx <strong>of</strong> other malignancy, bilateral disease, and ocular disease site. Results: The<br />

table lists pt characteristics. Median follow-up was 25.8 months (range<br />

0.3-307.4). Age at dx (HR�1.45, CI 1.04-2.02) and prior lymphoma hx<br />

(HR�3.35, CI 1.33-8.46) were predictive <strong>of</strong> relapse in a multivariate analysis.<br />

There was no difference in relapse rates between pts with EMZL and other<br />

lymphoma types (p�0.82). Relapse occurred in 26 (31.7%) pts with most<br />

common sites being ipsilateral eye (n�8), contralateral eye (n�3), distant<br />

lymph node (n�11), and other organs (n�11). Most common organ was breast<br />

(n�4). Of the 10 pts who had eye-only relapses, 8 received rituximab (R) and 2<br />

were observed. Of the 16 with extraocular relapse, 9 received radiation (RT), 2<br />

received R, 3 received other therapies, and 2 were observed. 7 deaths were<br />

recorded with a 5 year overall survival <strong>of</strong> 84.2%. Survival was similar to a<br />

matched healthy population (p�0.69). Causes <strong>of</strong> death were lymphoma in 4,<br />

another cancer in 1, and unknown in 2. Conclusions: OAL relapse patterns differ<br />

depending on initial treatment. Initial RT was more likely to relapse at distant<br />

sites; where as initial R was more likely to relapse in the OA. RT should be used<br />

for localized OAL. With bilateral ocular or systemic disease, R results in a high<br />

rate <strong>of</strong> long-term disease control.<br />

Pt characteristics n(%)<br />

Median age at Dx (range) 64 (24-91)<br />

Male 45 (54.9)<br />

Bilateral involvement 22 (26.8)<br />

Stage IE (n�73) 48 (69.5)<br />

Pathologic Dx (n�80)<br />

ENMZ 56 (70.0)<br />

Follicular 9 (11.3)<br />

DLBCL 3 (3.8)<br />

Mantle 2 (2.5)<br />

Other lymphoma 7 (8.8)<br />

Non-dx 3 (3.8)<br />

Initial Rx (n�73)<br />

RT 43 (58.9)<br />

R 13 (17.8)<br />

Chemo 5 (6.8)<br />

Combination 8 (11.0)<br />

Observation 4 (5.5)<br />

Lymphoma and Plasma Cell Disorders<br />

527s<br />

8070 General Poster Session (Board #35C), Mon, 1:15 PM-5:15 PM<br />

Brentuximab vedotin for relapsed or refractory non-Hodgkin lymphoma:<br />

Preliminary results from a phase II study. Presenting Author: Ranjana<br />

Advani, Stanford University Medical Center, Stanford, CA<br />

Background: Brentuximab vedotin is a CD30-directed antibody-drug conjugate<br />

approved for the treatment <strong>of</strong> Hodgkin lymphoma and systemic<br />

anaplastic large cell lymphoma (ALCL) after failure <strong>of</strong> other therapies.<br />

Based on the high objective response rate observed in patients with<br />

systemic ALCL, a type <strong>of</strong> non-Hodgkin lymphoma that is characterized by<br />

homogeneous CD30 expression, a study was initiated in other non-Hodgkin<br />

lymphomas that express the CD30 target. Methods: A phase 2 open-label<br />

single-arm study is underway in patients with relapsed or refractory<br />

CD30-positive non-Hodgkin lymphoma, excluding ALCL (NCT01421667).<br />

Brentuximab vedotin is administered IV at 1.8 mg/kg every 3 weeks until<br />

disease progression or unacceptable toxicity. The primary endpoint is<br />

objective response rate assessed by the Revised Response Criteria for<br />

Malignant Lymphoma (Cheson 2007). Tumor specimens are assessed by<br />

central lab in order to characterize the relationship <strong>of</strong> CD30 expression with<br />

antitumor activity. Results: Ten patients (age range 28–83; 5 M, 5 F) have<br />

enrolled to date. Diagnoses include diffuse large B-cell lymphoma (DLBCL,<br />

n�2), EBV-positive DLBCL <strong>of</strong> the elderly (n�3), primary mediastinal B-cell<br />

lymphoma (n�2), peripheral T-cell lymphoma NOS (n�2), and angioimmunoblastic<br />

T-cell lymphoma (AITL). Patients had received 1–6 prior chemotherapy<br />

regimens; 3 patients had prior stem cell transplants. Of 6 patients<br />

who have completed the cycle 2 response assessment, 2 attained complete<br />

remission, 1 with DLBCL (90% CD30�) and 1 with AITL (8% CD30�), 1<br />

had stable disease, and 3 had progressive disease. Treatment-related<br />

serious adverse events observed to date were rash, febrile neutropenia, and<br />

mastoiditis. Conclusions: Preliminary results suggest that brentuximab<br />

vedotin may have antitumor activity in patients with relapsed or refractory<br />

CD30-expressing non-Hodgkin lymphomas, in addition to the efficacy<br />

previously observed in systemic ALCL. Updated study results will be<br />

presented.<br />

8072 General Poster Session (Board #35E), Mon, 1:15 PM-5:15 PM<br />

The impact <strong>of</strong> race, age, and sex in follicular lymphoma (FL): A comprehensive<br />

SEER analysis in the pre- and post-rituximab (R) eras. Presenting Author: Chadi<br />

Nabhan, Advocate Lutheran General Hospital and Oncology Specialists, S.C.,<br />

Park Ridge, IL<br />

Background: While racial disparity has been well documented in a number <strong>of</strong><br />

cancers, the impact <strong>of</strong> race in FL outcomes is not well defined. Further, the<br />

importance <strong>of</strong> gender in FL has not been fully explored. Methods: We examined<br />

population-based FL overall survival (OS) data from SEER 13 (1993-2008)<br />

regarding race, sex, age, and socioeconomic status (SES) over two consecutive<br />

8-year (yr) periods: Era 1 (1993-2000, n�7,409) and Era 2 (2001–2008,<br />

n�9,083). Results: We identified16,492 FL patients (pts) (white (W): n�13,441;<br />

Hispanic (H): n�1,417; Asian/Pacific Islander (A/PI): n�887; and Black (B):<br />

n�747). Median ages at diagnosis differed significantly according to: (in yrs, W:<br />

62.1, H: 57.3, A/PI: 60.5, B: 56.6; P�0.01 for each race vs. W). For all pts, OS<br />

was superior in Era 2 vs. Era 1 (5-yr OS: 77% vs. 68%, respectively, P�0.0001).<br />

Further, OS was significantly improved for all age groups (�50, 50-59, 60-69,<br />

and 70-79 yrs) as well as for males (P�0.0019) and females (P�0.0001)<br />

across eras. Interestingly, females had superior OS compared with males in Era 1<br />

(P�0.004), but not in Era 2 (P�0.83). We subsequently compared OS within<br />

and across races (Table). All races, except A/PI, had improved 5-yr OS rates (age<br />

adjusted) from Era 1 to Era 2 (W: �0.001, H: 0.049, A/PI: 0.15, B: 0.003).<br />

Notably, A/PIs had the highest OS in Era 1, while H had the poorest OS in Era 2.<br />

These differences were more evident in males compared with females within<br />

each race. Finally, pts with higher SES had better OS in both eras, although OS<br />

was improved across eras for lower and higher SES populations. Conclusions:<br />

Collectively, we identified improved OS across eras, which was apparent for all<br />

ages, both sexes, and all races. We did not find superior outcome for females in<br />

the modern era as has been recently noted. However, several racial disparities<br />

persist, including inferior OS for H and superior OS A/PIs in the contemporary<br />

era. The disproportionate improvement in outcomes for some, but not all races,<br />

warrants continued study <strong>of</strong> racial disparities in FL.<br />

OS across eras based on race.<br />

5-yr OS<br />

Era 1<br />

W vs. H<br />

P values<br />

W vs. A/PI W vs. B<br />

W B H A/PI<br />

68%<br />

Era 2<br />

64% 68% 73% 0.20 0.85 0.007<br />

W B H A/PI<br />

77% 75% 73% 79% �0.0001 0.019 0.11<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


528s Lymphoma and Plasma Cell Disorders<br />

8073 General Poster Session (Board #35F), Mon, 1:15 PM-5:15 PM<br />

Phase I trial <strong>of</strong> fenretinide (4-HPR) intravenous emulsion in hematologic<br />

malignancies: A California Cancer Consortium study (PhI-42). Presenting<br />

Author: Ann Mohrbacher, University <strong>of</strong> Southern California Keck School <strong>of</strong><br />

Medicine and Norris Comprehensive Cancer Center, Los Angeles, CA<br />

Background: Fenretinide (4-HPR) is a cytotoxic retinoid with broad anticancer<br />

activity in preclinical studies. Due to limited bioavailability <strong>of</strong> a capsule<br />

formulation an intravenous intralipid-like emulsion formulation (4-HPR<br />

ILE) was developed to increase systemic exposures. Methods: 4-HPR was<br />

administered as a continuous intravenous infusion for 120 hrs every 21<br />

days. Systemic toxicities, responses, and pharmacokinetics were assessed.<br />

Accelerated Simon design proceeded until moderate or dose-limiting<br />

toxicities (DLT) were scored on Course 1. Doses were 80 mg/m2 /day to<br />

1810 mg/m2 /day. Patients with asymptomatic hypertriglyceridemia were<br />

scored separately. All patients were heavily pretreated. Results: Toxicityevaluable<br />

patients � 25. At 1810 mg/m2 /day, two patients experienced<br />

DLT hypertriglyceridemia, one with transient Grade 2 pancreatitis; at 1280<br />

mg/m2 /day (8 pts), two had asymptomatic Grade 4 hypertriglyceridemia,<br />

one experienced DLT pleural effusions; at 905 mg/m2 /day (6 pts) 2<br />

experienced asymptomatic Grade 4 hypertriglyceridemia; All 5 pts at 640<br />

mg/m2/day tolerated treatment. Pharmacokinetics showed a dose-toplasma<br />

level relationship with mean steady-state 4-HPR levels <strong>of</strong> ~mid-<br />

20’s �M (640 mg/m2 ); ~mid-30’s �M (905 mg/m2/day); and ~mid-50’s<br />

�M (1280 mg/m2 ). Responses to date include a 64% CR�PR�SD<br />

response rate (36% CR�PR response rate) in 11 relapsed T-cell lymphomas<br />

which included histone deacetylase inhibitor-refractory patients, and a<br />

PRu response in a NHL B-cell lymphoma. Reversible hypertriglyceridemia<br />

related to the intralipid vehicle accounted for 6/7 DLTs. Conclusions: MTD<br />

� 1280 mg/m2 /day x 5 days, every three weeks. 4-HPR ILE was safely<br />

administered and obtained 4-HPR plasma levels 6 -7 times higher than<br />

previously obtained using oral capsules. Durable complete responses were<br />

observed in T-cell lymphomas from 905 – 1810 mg/m2 /day. An expanded<br />

cohort is accruing to a dosing schedule modified to decrease asymptomatic<br />

hypertriglyceridemia <strong>of</strong> 600 mg/m2 on Day 1 (to allow for induction <strong>of</strong><br />

serum lipases) followed by 1200 mg/m2 Days 2-5. Supported by NCI U01<br />

CA062505 and CPRIT RP10072.<br />

8075 General Poster Session (Board #35H), Mon, 1:15 PM-5:15 PM<br />

Phase I trial <strong>of</strong> temsirolimus and lenalidomide in pts with rel/ref lymphomas.<br />

Presenting Author: Sonali M. Smith, The University <strong>of</strong> Chicago,<br />

Chicago, IL<br />

Background: The PI3K/Akt/mTOR axis is deregulated in lymphomas and is<br />

an emerging therapeutic target. We previously reported activity <strong>of</strong> temsirolimus<br />

(TEM) in DLBCL and FL (JCO 2010 28(31); however, the response<br />

duration was short. Lenalidomide (LEN) is an immunomodulatory agent<br />

with multiple anti-tumoral and microenvironmental effects, with activity<br />

across lymphoma subtypes. We are thus conducting a phase I/II study <strong>of</strong><br />

TEM plus escalating doses <strong>of</strong> LEN. The phase I portion is completed.<br />

Methods: Patients (pts) had rel/ref lymphoma after �1 cytotoxic regimen.<br />

Other criteria: ANC � 1000/mL, platelets � 75,000/mL, nl renal and<br />

hepatic function, no VTE within 3 months, non-pregnant. A standard “3 �<br />

3” design was used with dose levels (DL) listed (Table). TEM was given IV<br />

weekly and LEN was dosed orally on D1-D21, q28 days. Dose-limiting<br />

toxicity (DLT) was defined as cycle 1 grade 3 or 4 non-hematologic toxicity<br />

not responsive to standard supportive care, grade 4 thrombocytopenia � 7<br />

days (or associated with bleeding or requiring more than 1 platelet<br />

transfusion), ANC � 500/mL � 7 days despite growth factors, or any<br />

thromboembolic event. Results: 18 pts (13M, 5F), med age 64 y (range,<br />

42-80 y) were enrolled. 3 pts are ineval for DLT evaluation: one withdrew<br />

consent before starting treatment, 1 withdrew consent after a single dose,<br />

and 1 died <strong>of</strong> rapid disease progression after 1 dose. There was 1 DLT at<br />

DL1 and 2 DLTs at DL3 (Table). Adverse effects that did not meet DLT<br />

criteria: hypokalemia, hypertriglyceridemia, vomiting, urinary tract infection,<br />

skin infection, nausea, hypoxia, hyponatremia, diarrhea, and hyperglycemia<br />

(each occurring in one pt). There are 5 partial responses, 4 stable<br />

disease, 3 progressive disease, 2 not adequately assessed, and 4 still on<br />

active treatment. Conclusions: The combination <strong>of</strong> weekly intravenous TEM<br />

plus oral LEN is well-tolerated in a heavily pretreated group <strong>of</strong> pts with<br />

rel/ref lymphomas. The recommended phase II doses are TEM 25mg<br />

weekly plus LEN 20mg (D1-D21, q28d).<br />

Dose level<br />

TEM<br />

(flat dose in mg)<br />

Dose<br />

LEN<br />

(flat dose in mg)<br />

No. Pts DLT<br />

-1 25 mg 10 mg n/a n/a<br />

1 25 mg 15 mg 8 (2 ineval for DLT) Grade 4 hypokalemia<br />

2 25 mg 20 mg 4 (1 ineval for DLT) No DLT<br />

3 25 mg 25 mg 6 Grade 3 diarrhea, grade 3<br />

HSV mucositis<br />

8074 General Poster Session (Board #35G), Mon, 1:15 PM-5:15 PM<br />

Use <strong>of</strong> exon-based transcriptome pr<strong>of</strong>iling to identify novel signaling<br />

pathways and survival-associated genes in diffuse large B-cell lymphoma.<br />

Presenting Author: Sirpa Leppa, Helsinki University Central Hospital,<br />

Helsinki, Finland<br />

Background: Identification <strong>of</strong> biological prognostic factors that could be<br />

used to define poor risk diffuse large B-cell lymphoma (DLBCL) patients is<br />

a main concern. Methods: Study population consisted <strong>of</strong> 38 de novo high<br />

risk DLBCL patients less than 65 years old. The patients were treated in the<br />

Nordic phase II protocol with six courses <strong>of</strong> R-CHOEP14 followed by<br />

systemic central nervous system prophylaxis with one course <strong>of</strong> high dose<br />

methotrexate and one course <strong>of</strong> high dose cytarabine. Exon array-based<br />

pr<strong>of</strong>iling was used to screen signaling pathways and differentially expressed<br />

genes between the clinically high risk patients, who had relapsed or<br />

remained in remission in response to dose dense chemoimmunotherapy. At<br />

the time <strong>of</strong> the analysis, median follow up was 34 months, progression free<br />

survival (PFS) 78% and overall survival (OS) 78%. Results: The screen<br />

between relapsed patients and the patients in remission using criteria <strong>of</strong> p<br />

� 0.05 and fold change � 1.6 revealed 566 differentially expressed genes<br />

(131 protein coding), <strong>of</strong> which 24 were likely to be involved in conventional<br />

signaling pathways, including those regulating antigen processing and<br />

presentation (CIITA, HLA-DQA2, HLA-DQB1, RFXAP), Jak-STAT signaling<br />

(SOCS3), Notch signaling (NOTCH1) and Toll-like receptor signalling<br />

(IRF5). In cox univariate analysis, 12 <strong>of</strong> 24 genes were found to associate<br />

with PFS (p�0.05). Of these, high expression <strong>of</strong> CIITA, DLL4, HLA-DQA2,<br />

HLA-DQB1, IRF5, NOTCH1, PER1, RFXAP, SEMA4D and ZFP36 had a<br />

favorable impact on PFS, whereas high levels <strong>of</strong> ENPP3 and PRKAR2B<br />

were associated with adverse outcome. Differential expression <strong>of</strong> four genes<br />

was confirmed by quantitative PCR, and prognostic value <strong>of</strong> six genes<br />

validated using Lymphoma/Leukemia Molecular Pr<strong>of</strong>iling Project microarray<br />

data set. Immunohistochemical validation <strong>of</strong> the findings in a larger<br />

patient cohort is ongoing. Germinal centre B-cell signature did not predict<br />

survival in this cohort. Conclusions: The results provide evidence that<br />

exon-based transcriptome pr<strong>of</strong>iling can identify biologically relevant signaling<br />

pathways and genes that discriminate the outcome <strong>of</strong> homogenously<br />

treated young high risk DLBCL patients.<br />

8076 General Poster Session (Board #36A), Mon, 1:15 PM-5:15 PM<br />

Efficacy and safety <strong>of</strong> bexarotene combined with psoralen/ultraviolet A light<br />

(PUVA) compared to PUVA treatment alone in stage IB-IIa mycosis<br />

fungoides (MF): Final results from EORTC cutaneous lymphoma task force<br />

(CLTF) phase III clinical trial 21011. Presenting Author: Sean Whittaker,<br />

Guy’s and St. Thomas’ Hospital, London, United Kingdom<br />

Background: Skin-directed treatment with methoxsalen (PUVA) is the<br />

current treatment standard in stage IB-IIA MF. A combination <strong>of</strong> PUVA and<br />

bexarotene might be <strong>of</strong> additional clinical benefit for MF stage I/II patients<br />

(pts). Methods: EORTC 21011 was a randomised, open label phase III<br />

study comparing combined bexarotene and PUVA versus PUVA alone in pts<br />

with stage IB and IIA MF. Study primary endpoint was response (complete<br />

clinical � partial response, CCR�PR) rate; secondary endpoints: cumulative<br />

dose <strong>of</strong> UVA and number <strong>of</strong> PUVA sessions necessary to achieve a CCR,<br />

duration <strong>of</strong> CCR, time to relapse, safety and percentage <strong>of</strong> drop-outs.<br />

Results: The study recruited stage IB/IIA MF pts and was prematurely<br />

closed due to low accrual after 93/145 required pts (65%) were randomized;<br />

45 to PUVA, 48 to PUVA�bexarotene. Median number <strong>of</strong> PUVA<br />

weeks were 12 (1-17) in PUVA vs. 10.5 (1-16) in combination arm. Total<br />

UVA doses were 107J/cm2 (1.4-489.9) in PUVA vs. 101.7J/cm2 (0.2-<br />

529.9) in combination arm. Few grade 3-4 toxicities were observed in both<br />

arms (liver enzyme elevation, neutropenia, anemia, increased cholesterol,<br />

photosensitivity, pruritus, rash, hypertriglyceridemia). Best overall response<br />

(CCR/PR) rate was 71.1% (33/45) for PUVA alone and 77.1%<br />

(37/48) for combination arm (p-value�0.57). The median <strong>of</strong> duration <strong>of</strong><br />

response was 9.6 for PUVA vs 5.8 months for combination arm (p<br />

value�0.33). CCR was seen in 25 pts, 10 in PUVA (CCR 24%) and 15 in<br />

combination therapy (CCR 33%) (pvalue�0.45). Similarily, a lower UVA<br />

dose was required to achieve a CCR in the combination arm (median <strong>of</strong><br />

55.8 J/cm2) compared to the PUVA arm (median <strong>of</strong> 117.58 J/cm2) (p<br />

value�0.5). Conclusions: No significant difference in response rate was<br />

observed in this study. There was a trend towards fewer PUVA sessions and<br />

lower UVA dose to achieve CCR in the PUVA/bexarotene combination arm<br />

(median <strong>of</strong> 27.5 vs. 22,p-value � 0.11) but this did not achieve statistical<br />

significance due to insufficient power. The safety pr<strong>of</strong>ile was acceptable, as<br />

there were only few grade 3-4 toxicities observed in both arms.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


8077 General Poster Session (Board #36B), Mon, 1:15 PM-5:15 PM<br />

Dual-point FDG-PET: A novel scanning technique in Hodgkin lymphoma<br />

with bulky disease. Presenting Author: Andrea Gallamini, Hematology<br />

Department, S. Croce General Hospital, Cuneo, Italy<br />

Background: Interim 18F-FDG-PET (iPET) is the most important prognosticator<br />

in advanced-stage ABVD-treated Hodgkin Lymphoma (HL), but in early<br />

stage w/ or w/o bulky lesion a low PPV <strong>of</strong> iPET was reported. Dual-point PET<br />

scan (2P-PET) has been used to discriminate unspecific inflammatory from<br />

neoplastic FDG uptake. At the Tx end, with a single FDG-avid mass (SFAM),<br />

the specificity <strong>of</strong> PET in Tx response evaluation was sub-optimal. We report<br />

here preliminary results from a cohort <strong>of</strong> HL patients (p) presenting with<br />

bulky lesions, scanned with 2P-PET with the aim to increase specificity and<br />

PPV. Methods: From 12.2008 till 01.2012 24 HL bulky p from Italian,<br />

French and Polish centers underwent 2P-PET at baseline (2P-PET-0), after<br />

2 ABVD (2P-PET-2) and at Tx end (2P-PET-end). 2P-PET scanning<br />

technique consisted in 2 consecutive image acquisitions �60’ (EaS) and<br />

�120’(LaS) after single, standard-dose FDG injection. Tx was ABVD (x4 or<br />

x6) � consolidation RxT. No Tx change was done based on iPET results.<br />

Scans were reviewed by 2 expert readers in a consensus session. Standardized-Uptake<br />

Value (SUV) MAX was calculated both in EaS and LaS using<br />

Volume <strong>of</strong> Interest Regions (VOIs) in sites <strong>of</strong> residual FDG uptake already<br />

recorded in PET-0. �SUVMAX and FDG retention index (RI) were calculated<br />

from SUVs in the same VOIs <strong>of</strong> both scans. Results: In 24 p (1 stage I,12 II,<br />

4 III, 7 IV), 34 2P-PET were done.10 p underwent 2P-PET-0: in 10/10<br />

SUVMAX increased from EaS to LaS (�SUVMAX 1-4.3). 15 p had a<br />

2P-PET-2: 12 with a mean-follow-up <strong>of</strong> 18.36 months were evaluable, 3<br />

too short ( �1, �5, �6 months). 7/12 p showed a RI reduction <strong>of</strong> 55%<br />

(200-14): all are in continuous CR (CCR). 5/12 p. showed a 20% (11-26)<br />

RI increase: all progressed �1 to�9 months after Tx end. Overall 3/12<br />

cases (1 false �, 2 false- with a Deauville score 4 and 2,3 respectively)<br />

were correctly classified after 2P-PET. 9 p with a SFAM had a 2P-PET-end:<br />

in the 5/9 with an increased RI <strong>of</strong> 23% (9-43) a biopsy proved HL relapse.<br />

In 4/9 a reduced RI <strong>of</strong> -29% (9-45) was found: in 2 biopsy disclosed<br />

presence <strong>of</strong> residual thymic along with inflammatory tissue and 2 were in<br />

CCR �2 to�8 after Tx end. Conclusions: DSUV Max increased at baseline<br />

and in all relapsing patients, and decreased in patients in CCR. The PPV<br />

and a PNV <strong>of</strong> 2P-PET were 100%.<br />

8080 General Poster Session (Board #36E), Mon, 1:15 PM-5:15 PM<br />

The incorporation <strong>of</strong> rituximab (R) and liposomal doxorubicin (LD) into<br />

CODOX-m/IVAC for untreated Burkitt lymphoma (BL): Final results <strong>of</strong> a<br />

prospective multicenter phase II study. Presenting Author: Andrew M.<br />

Evens, The University <strong>of</strong> Massachusetts Medical School, Worcester, MA<br />

Background: Two-year survival rates remain �65-70% for adult BL.<br />

Further, there is a paucity <strong>of</strong> data examining the addition <strong>of</strong> R with<br />

CODOX-M/IVAC. Methods: 25 patients (pts) with classic BL enrolled onto<br />

this phase II study (3/07-4/11). Pts were classified as low risk (LR) or high<br />

risk (HR); LR received 3 CODOX-M, while HR had 4 alternating CODOX-M/<br />

IVAC (Mead et al, Blood 2008). LD (40 mg/m2 ) was used in lieu <strong>of</strong><br />

doxorubicin, while intravenous R (500 mg/m2 ) was given days 0 � 8<strong>of</strong><br />

CODOX-M and days 0 � 6 <strong>of</strong> IVAC cycles. Ejection fraction (EF) was<br />

assessed at baseline, s/p 2 cycles and completion. Results: Median age was<br />

44 years (yrs) (23-70). There were 20 HR and 5 LR pts; 3 HR and 1 LR pt<br />

were HIV�. 15% <strong>of</strong> HR pts had � CNS disease. In addition, 35% <strong>of</strong> HR pts<br />

had bulk �10 cm and 40% had bone marrow involvement. 24/25 pts were<br />

evaluable for toxicity and response. Therapy was completed at a median <strong>of</strong><br />

13 weeks (11-20) for HR pts and 10 weeks for LR (9-12). Myelosuppression<br />

(62% grade 4 thrombocytopenia, 4% grade 4 anemia) and mucositis<br />

(33% grade 3, 13% grade 4) appeared comparable with prior CODOX-M/<br />

IVAC data. Other grade 3 toxicities were infection (38%), neutropenic fever<br />

(29%), transaminitis (33%), diarrhea (8%), creatinine (8%), seizure (4%),<br />

vomiting (4%). Notably, there was no grade 3 or 4 neuropathy. Two grade 2<br />

and three grade 3 cardiac events occurred (all depressed EF, no clinical<br />

CHF). Two <strong>of</strong> the three grade 3 cardiac events occurred in pts age �65 yrs.<br />

Among all pts, the median change in EF at baseline vs study end was -2%<br />

(-22% to �11%). The overall response rate (modified Cheson with<br />

FDG-PET) after 2 cycles was 100% with a 67% complete remission rate. At<br />

a median follow-up <strong>of</strong> 2.3 yrs, 2-year PFS and OS rates for all pts were 86%<br />

and 86%, respectively (LR 2-yr PFS and OS: both 100%; HR 2-yr PFS and<br />

OS: both 82%). Furthermore, 2-yr PFS and OS for HR, HIV-negative BL<br />

were 91% and 91%, respectively (disease-specific survival: 100%).<br />

Conclusions: The integration <strong>of</strong> R and LD into CODOX-M/IVAC for adult BL<br />

is feasible and associated with similar tolerability compared with prior<br />

reports. Moreover, this regimen was associated with excellent survival<br />

rates, especially for HIV-negative BL.<br />

Lymphoma and Plasma Cell Disorders<br />

529s<br />

8078 General Poster Session (Board #36C), Mon, 1:15 PM-5:15 PM<br />

Association <strong>of</strong> tumor-associated macrophages <strong>of</strong> M2 subtype with outcome<br />

in follicular lymphoma. Presenting Author: Clarinda Wei Ling Chua,<br />

National Cancer Centre Singapore, Singapore, Singapore<br />

Background: Current methods used to prognosticate patients with follicular<br />

lymphoma (FL) include the Follicular Lymphoma International Prognostic<br />

Index (FLIPI) and tumor grade. However, they do not provide information on<br />

the biological and molecular features <strong>of</strong> FL. Recent data suggest that high<br />

tumor-associated macrophages (TAM) content may be an adverse prognostic<br />

factor in FL. However, TAM consists <strong>of</strong> two main subtypes, M1 and M2,<br />

with the former related to pro-inflammatory properties while the latter,<br />

anti-inflammatory functions. Currently, the prognostic impact <strong>of</strong> each<br />

individual subtype has not been elucidated and this may be more important<br />

than looking at TAM alone. Methods: Tumor specimens <strong>of</strong> 98 patients with<br />

FL diagnosed between 2005 and 2009 were investigated using immunohistochemistry<br />

and fluorescence in-situ hybridization (FISH) using breakapart<br />

FISH probes targeting BCL2, BCL6, MYC and IgH genes. Tumor<br />

specimens expressing a higher proportion <strong>of</strong> CD68�/CD163- in one high<br />

power field were defined as M1 subtype and those that expressed a greater<br />

proportion <strong>of</strong> CD68�/CD163� were denoted as M2 subtype. Results:<br />

Amongst the 98 patients, 60 (61%) were <strong>of</strong> the M2 subtype, 59%<br />

presented with advanced disease, 47% were grade 3 and the median age<br />

was 59 years (21 – 88 years). Baseline characteristics including grade,<br />

stage and recurrent translocations did not significantly differ between the<br />

M1 and M2 groups. Similarly, there was also no difference in terms <strong>of</strong><br />

treatment received (both Rituximab and chemotherapy) between the two<br />

groups. Despite these similarities, with a median follow-up <strong>of</strong> 3.2 years, the<br />

3-year overall survival (OS) was significantly different (M1, 100% vs M2,<br />

85%; p � 0.01). All 11 patients who died were <strong>of</strong> the M2 subtype<br />

(p�0.009). Conclusions: Our findings show that despite similar patient<br />

characteristics and treatment, there was, however, a statistically significant<br />

difference in OS, with the M2 subtype demonstrating an inferior outcome.<br />

The M2 TAM subtype rather than TAM alone may be a more important<br />

prognostic marker in FL, which could possibly be explained by its known<br />

biological anti-inflammatory function.<br />

8081 General Poster Session (Board #36F), Mon, 1:15 PM-5:15 PM<br />

Efficacy <strong>of</strong> ocaratuzumab (AME-133v) in relapsed follicular lymphoma<br />

patients refractory to prior rituximab. Presenting Author: Jennifer L. Wayne,<br />

Mentrik Biotech, LLC, Dallas, TX<br />

Background: Ocaratzumab, previously known as AME-133v, is a humanized<br />

next-generation anti-CD20 monoclonal antibody. It has been optimized<br />

with a 13 to 20-fold increase in binding affinity to CD20 and improved<br />

binding to the low-affinity (F/F and F/V) polymorphisms <strong>of</strong> Fc�RIIIa (CD16),<br />

which are thought to predict lower response rates and shorter duration <strong>of</strong><br />

responses to rituximab. Methods: In a phase I dose escalation study in<br />

relapsed follicular lymphoma (FL) patients, ocaratuzumab was welltolerated<br />

at doses up to 375 mg/m2 (Forero-Torres et al. CCR 2012). In a<br />

follow-on phase II trial, 44 patients with relapsed FL following prior<br />

rituximab and the low-affinity Fc�RIIIa polymorphism (F-carriers) received<br />

375 mg/m2 <strong>of</strong> ocaratuzumab weekly for 4 doses. In this study, overall<br />

response rate (ORR) was 36% and median progression free survival (PFS)<br />

was 91 weeks (Ganjoo et al. Haematologica 2011). Results: Amongst the<br />

56 patients receiving 100 and 375 mg/m2 <strong>of</strong> ocaratuzumab, 8 patients had<br />

a previous time to progression <strong>of</strong> � 180 days following their last rituximab<br />

treatment. These patients had a median <strong>of</strong> 2 prior rituximab treatments,<br />

(range 1-6 treatments), and median PFS following last treatment <strong>of</strong> 159<br />

days. Five <strong>of</strong> the 8 patients showed a longer PFS after ocaratuzumab<br />

administration, compared with last rituximab treatment. All 5 patients<br />

expressed the homozygous low-affinity genotype <strong>of</strong> Fc�RIIIa (F/F). At the<br />

time <strong>of</strong> study closure, 3 <strong>of</strong> the patients were still in remission (indicated by<br />

* in the table). Conclusions: This retrospective analysis suggests that<br />

ocaratuzumab may be non-cross-resistant to rituximab in patients with the<br />

low-affinity Fc�RIIIa polymorphism. Prolonged PFS in selected patients<br />

following ocaratuzumab suggests that the increased binding affinity to<br />

CD16 and improved antibody-dependent cell-mediated cytotoxicity (ADCC)<br />

<strong>of</strong> this antibody is clinically relevant. As a single agent, ocaratuzumab may<br />

provide prolonged clinical benefit in relapsed FL patients and a clinical trial<br />

comparing ocaratuzumab to rituximab is in preparation.<br />

No. <strong>of</strong> prior RTX treatments PFS after RTX (days) PFS after ocaratuzumab (days)<br />

1 172 826*<br />

2 159 636<br />

1 97 434*<br />

2 163 357*<br />

3 103 257<br />

6 180 168<br />

2 116 119<br />

2 176 91<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


530s Lymphoma and Plasma Cell Disorders<br />

8082 General Poster Session (Board #36G), Mon, 1:15 PM-5:15 PM<br />

Disruption <strong>of</strong> the eIF4F translation initiation complex as a determinant <strong>of</strong><br />

diffuse large B-cell lymphoma responsiveness to enzastaurin<br />

(LY317615.HCl) and its primary metabolite (LY326020). Presenting<br />

Author: Jeremy Richard Graff, Lilly Research Labs Cancer Biology and<br />

Patient Tailoring Lilly, Indianapolis, IN<br />

Background: Enzastaurin (enza) is in ph 3 registration trials for DLBCL<br />

patients at high risk <strong>of</strong> relapse following R-CHOP therapy. In a phase 2<br />

DLBCL study, 4 <strong>of</strong> 55 treated patients were progression- free after<br />

prolonged, continuous oral enza therapy with 3 <strong>of</strong> these 4 confirmed as<br />

complete responders (Robertson et al., JCO, 2007). The molecular mechanism<br />

for this differential response is unclear. Methods: In clinical trials,<br />

Enza yields 2-4 �M total circulating drug, comprised <strong>of</strong> ~50% enza, ~50%<br />

primary metabolite, LY326020. We therefore evaluated the sensitivity <strong>of</strong> a<br />

DLBCL cell panel representing both Activated B Cell (ABC) and Germinal<br />

Center (GC) subtypes to enza and LY326020. Gene expression analyses,<br />

western blotting to explore intracellular signaling and mRNA cap analogue<br />

co-capture assays were used to identify the critical effectors <strong>of</strong> drug<br />

sensitivity. Results: For the first time, we show the pr<strong>of</strong>ound biological<br />

activity <strong>of</strong> LY326020, the primary metabolite that accounts for ~ 50% <strong>of</strong><br />

circulating drug in patients. Like Enza, though more potently, LY326020<br />

inhibits PKC and PI3K-AKT-TOR pathway signaling and robustly induces<br />

apoptosis in both ABC and GC DLBCL cells. In both sensitive and resistant<br />

cells, enza and LY326020 reduced phosphorylation <strong>of</strong> numerous proteins<br />

in the PI3K-AKT-TOR pathway (e.g. pGSK3�ser9 ) in a dose and timedependent<br />

manner. However, only sensitive DLBCL cells showed reduced<br />

4EBP1ser65 phosphorylation. Accordingly, we show a dose and timedependent<br />

increase in 4EBP1: eIF4E binding. This increase is most<br />

pronounced by LY326020. Moreover, cells selected for resistance to enza<br />

show reduced 4EBP1 expression and cells lacking 4EBP1 are insensitive to<br />

the pro-apoptotic effects <strong>of</strong> enza and LY326020. Conclusions: These data<br />

demonstrate that sensitivity <strong>of</strong> DLBCL to both enza and LY326020 is<br />

critically dependent upon 4EBP1 modulation and subsequent disruption <strong>of</strong><br />

the eIF4F translation complex. Moreover, these data are the first to show<br />

the potent biologic activity <strong>of</strong> LY326020, the primary metabolite <strong>of</strong> enza<br />

that accounts for ~50% <strong>of</strong> total circulating drug in patients and in<br />

preclinical models.<br />

8084 General Poster Session (Board #37A), Mon, 1:15 PM-5:15 PM<br />

An update on gemcitabine, rituximab, and oxaliplatin in combination for<br />

relapsed/refractory non-Hodgkin lymphomas. Presenting Author: Robert M.<br />

Crescentini, H. Lee M<strong>of</strong>fitt Cancer Center & Research Institute, Tampa, FL<br />

Background: Relapsed/refractory non-Hodgkin lymphomas (NHL) have no<br />

standard <strong>of</strong> care. A variety <strong>of</strong> salvage chemotherapy options are available.<br />

We previously reported results <strong>of</strong> our phase II trial using gemcitabine,<br />

rituximab and oxaliplatin (GROC) in the salvage setting for relapsed/<br />

refractory NHL in which we observed an overall response rate <strong>of</strong> 58% with<br />

an incidence <strong>of</strong> grade 3-4 thrombocytopenia <strong>of</strong> 9% and neutropenic fever<br />

<strong>of</strong> 3.5%, but no grade 3-4 non-hematologic toxicities. Here we update<br />

progression free survival (PFS) and overall survival (OS) data. Methods: This<br />

phase II, single-arm, multicenter study evaluated safety and efficacy <strong>of</strong><br />

GROC in patients with relapsed/refractory NHL. Patients were treated on a<br />

14 day cycle. On day 1, patients with CD20� NHL received rituximab (375<br />

mg/m2 ). On day 2, patients received gemcitabine (1000 mg/m2 ) and<br />

oxaliplatin (100 mg/m2 ). Granulocyte colony stimulating factor was given.<br />

Stem cell transplant (SCT) was considered after a minimum <strong>of</strong> 6 cycles.<br />

Results: A total <strong>of</strong> 58 patients were enrolled from the H. Lee M<strong>of</strong>fitt and the<br />

Auxilio Mutuo Cancer Centers. Ages ranged from 24 to 88 years (median 72<br />

years). The majority <strong>of</strong> patients had an ECOG performance status <strong>of</strong> 0-1<br />

(89%). Lymphoid neoplasms included large B-cell (79%), follicular (7%),<br />

lymphoblastic (1.8%), Burkitt (1.8%), primary mediastinal large B-cell<br />

(3.5%), and peripheral T-cell lymphoma (7%). Eighty-one percent <strong>of</strong><br />

patients had stage III-IV disease, median IPI was 3, 40% had B-symptoms,<br />

43% had bulky disease and 74% had an elevated LDH. Anthracyclinebased<br />

therapy had been used in 91% <strong>of</strong> patients and 66% had received<br />

rituximab. Median PFS was 134 days (95% CI 115-153) and median OS<br />

was 296 days (95% CI 164-428). No difference in response was observed<br />

based on age �60, IPI, LDH or albumin levels. Prior therapy with rituximab<br />

(p�0.02) and initial response to front-line therapy (p�0.04) appear to<br />

correlate with improved outcomes. Nine patients went on for SCT.<br />

Conclusions: GROC is a useful salvage regimen for relapsed/refractory NHL<br />

with minimal toxicities and good clinical efficacy. Several patients were<br />

able to be successfully mobilized, collected and transplanted post GROC<br />

therapy.<br />

8083 General Poster Session (Board #36H), Mon, 1:15 PM-5:15 PM<br />

Use <strong>of</strong> the cumulative amount <strong>of</strong> serum-free light chains (sFLC) at<br />

diagnosis and PET2 for the early identification <strong>of</strong> high risk <strong>of</strong> treatment<br />

failure in Hodgkin lymphoma (cHL). Presenting Author: Rosaria De Filippi,<br />

Hematology-Oncology and Stem Cell Transplantation Unit, Istituto Nazionale<br />

Tumori, Fondazione �G.Pascale�, Naples, Italy<br />

Background: Since early identification <strong>of</strong> patients (pts) at risk <strong>of</strong> failure is<br />

the mainstay <strong>of</strong> a risk-adapted therapy, we explored the prognostic impact<br />

<strong>of</strong> the sFLC assay in cHL, whose biology involves ongoing activation <strong>of</strong><br />

polyclonal B-cells. Methods: Serum samples from 248 untreated cHL pts<br />

were tested by the Freelite assay. Median age was 32 yrs (r 15-85), males<br />

47%, stages: I (5%), II (51%), III (17%), IV (27%); B-sympt. 60%,<br />

E-disease, 38%; bulky �10 cm, 44%; ESR � 65, 42%; IPS �3, 39%.<br />

Early unfavorable disease (GHLSG/ EORTC) was respectively found in 33%<br />

and 42% <strong>of</strong> cases. ABVD was given to 89% <strong>of</strong> pts. Results: Absolute FLC<br />

levels were summed into a sFLC(���) variable and ROC analysis indicated<br />

57.1 mg/mL as the threshold to discriminate outcomes. CR rates were 96%<br />

and 67% for pts below and above the cut<strong>of</strong>f, respectively (p�.0001). Cox<br />

univariate analysis disclosed a HR <strong>of</strong> 16.70 (95% CI, 8.5-32.9) <strong>of</strong> events<br />

for sFLC(���) � 57.1 mg/mL, by far higher than for PET2 positivity (HR<br />

10.8), PS �1 (HR 4.2), IPS �3 (HR 2.8) and all other predictors (HR<br />

0.54-2.4). In a multivariaye model only sFLC(���) and PET2 remained<br />

independent predictors. A dismal 8-yrs EFS characterized pts with<br />

sFLC(���) above threshold (20% vs 89%; �2 119, p�.0001). Pts with<br />

sFLC(���) below cut<strong>of</strong>f and a negative PET2 had an EFS <strong>of</strong> 93% as<br />

compared to 36% <strong>of</strong> those with sFLC(���) above cut<strong>of</strong>f or a positive PET2.<br />

Pts with sFLC(���) above cut<strong>of</strong>f and positive PET2, had the worse<br />

outcome with an EFS �10% and a median survival �12 mo.s (�2 65.4;<br />

p�.0001). sFLC assay was even more valuable in identifying poor risk pts<br />

within the early unfavorable category (5-yrs EFS �25%, �2 51 p�.0001).<br />

By immunoistochemistry small B cells and plasmacytoid lymphocytes were<br />

identified as the main source <strong>of</strong> sFLC in HL tissues while a strong sFLC<br />

uptake by mast cells was documented. Conclusions: A cumulative amount<br />

<strong>of</strong> sFLC �57.1 mg/mL, is the strongest independent predictor <strong>of</strong> failure in<br />

cHL. Combining sFLC(���) and PET2 outcomes can timely discriminate<br />

poor risk pts subsets, who may benefit from upfront treatment escalation or<br />

early salvage. Our data also support that sFLC might endorse immunobiologic<br />

activities relevant to cHL pathobiology.<br />

8085 General Poster Session (Board #37B), Mon, 1:15 PM-5:15 PM<br />

Prevention <strong>of</strong> adverse events during treatment <strong>of</strong> HIV-associated Hodgkin<br />

lymphoma with ritonavir and zidovudine. Presenting Author: Sonia Sandhu,<br />

John H. Stroger Jr. Hospital <strong>of</strong> Cook County, Chicago, IL<br />

Background: In response to very high rates <strong>of</strong> neurologic and hematologic<br />

adverse events (AE) when ABVD was used in combination with ritonavir<br />

(RTV) or zidovudine (AZT) in HIV-associated Hodgkin (HL) we instituted a<br />

policy to use alternative antiretroviral agents during HL therapy in our HIV<br />

patients. In this study, we examined all AE in HIV-HL since the exclusion <strong>of</strong><br />

RTV and AZT over 2 years ago. We also evaluated the AEs when HAART and<br />

chemotherapy for NHL were taken together in an expanded cohort <strong>of</strong> 52<br />

pts. Methods: A screen <strong>of</strong> pharmacy and hospital databases between<br />

1998-2012 identified all HIV-associated HL and NHL patients. Adverse<br />

events during chemotherapy were assessed by chart review and graded per<br />

the NCI Common Terminology Criteria for Adverse Events. Statistics:<br />

Fisher’s exact test was used to examine the differences in AE incidence<br />

associated with use <strong>of</strong> specific antiretrovirals. Results: HAART use during<br />

chemotherapy was identified in 35/36 (96%) pts with HL and 52/108<br />

(48%) <strong>of</strong> pts with NHL. Before RTV and AZT were prohibited, G3/4<br />

neuropathy, neutropenia, and anemia developed in 31, 68, and 57% <strong>of</strong> 23<br />

pts with HIV-HL respectively. Since then, 12 patients were treated with<br />

non-RTV and AZT based HAART. 0% neuropathy and only 20% G3/4<br />

neutropenia and 10% anemia was observed, each statistically significant<br />

(p�0.01). Of the 54 patients with NHL, 64% received CHOPR like,<br />

HYPERCVADR (18%), and daEPOCHR (11%). All AEs for each NHL<br />

regimen were similar to historical controls and no anti HIV medication was<br />

found to correlate with any AE, despite 28% <strong>of</strong> the HAART regimens<br />

containing RTV. Conclusions: No relationship between any AE and anti HIV<br />

medications was identified during treatment for NHL. But, excluding RTV<br />

or AZT-based HIV therapy during HL treatment, decreased neuropathy,<br />

neutropenia, and anemia by 100%, 38%, and 28% respectively. We<br />

suggest that exclusion <strong>of</strong> ritonavir and zidovudine from HAART regimens<br />

used with ABVD become the standard.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


8086 General Poster Session (Board #37C), Mon, 1:15 PM-5:15 PM<br />

Hematologic safety data from four phase II studies <strong>of</strong> single-agent<br />

carfilzomib in relapsed and/or refractory multiple myeloma Presenting<br />

Author: Ajay K. Nooka, Emory University Winship Cancer Institute, Atlanta,<br />

GA<br />

Background: Carfilzomib (CFZ) is a next-generation proteasome inhibitor<br />

that is currently being evaluated in patients (pts) with multiple myeloma<br />

(MM). Given that the majority <strong>of</strong> pts with MM exhibit either disease- or<br />

therapy-related myelosuppression sometime in the course <strong>of</strong> their disease,<br />

detailed evaluation <strong>of</strong> treatment-emergent hematologic safety data with<br />

CFZ is <strong>of</strong> significant interest. In the following analysis, we report the<br />

hematologic safety pr<strong>of</strong>ile for single-agent CFZ from 526 pts treated in four<br />

phase 2 studies. Methods: Pts treated with CFZ in trials 003-A0, 003-A1,<br />

004, and 005 were included in the analysis. Pts with preexisting hematologic<br />

abnormalities <strong>of</strong> Grade (G) 0–2 (G0–3 for 005) were eligible to enroll.<br />

CFZ was dosed in all studies on Days 1, 2, 8, 9, 15, and 16 <strong>of</strong> a 28-day<br />

cycle (C). Doses were 20 mg/m2 in C1 for all studies escalating to 27 mg/m2 in C2 per individual protocol, except 005 (15 mg/m2 in C1, 20 mg/m2 in<br />

C2, and 27 mg/m2 in C3). Incidence, frequency, and severity <strong>of</strong> thrombocytopenia,<br />

lymphopenia, neutropenia, and anemia were analyzed in terms <strong>of</strong><br />

adverse events (AEs) <strong>of</strong> interest. Laboratory data were analyzed for trends<br />

over time. Results: See table below for summary <strong>of</strong> hematologic events. In<br />

general, platelet counts trended down, reached a nadir by Day 8 <strong>of</strong> a 28-day<br />

treatment cycle, and normalized before the start <strong>of</strong> next cycle. There was no<br />

evidence <strong>of</strong> cumulative thrombocytopenia or clinically significant episodes<br />

<strong>of</strong> bleeding associated with thrombocytopenia. Febrile neutropenia was<br />

reported in 1% <strong>of</strong> pts. No mortality due to hematologic events was reported.<br />

Conclusions: Hematologic AEs, although common with CFZ treatment, were<br />

infrequently dose limiting. <strong>Clinical</strong>ly significant G3/4 cytopenias were both<br />

infrequent and transient. In summary, the hematological safety pr<strong>of</strong>ile <strong>of</strong><br />

CFZ was similar to or better than currently approved MM therapies,<br />

providing further evidence <strong>of</strong> its acceptable safety pr<strong>of</strong>ile in heavily<br />

pretreated MM pts. Summary <strong>of</strong> hematologic events.<br />

Total population<br />

N�526, n (%) Thrombocytopenia Lymphopenia Neutropenia Anemia<br />

Any AE 199 (37.8) 136 (25.9) 119 (22.6) 246 (46.8)<br />

G3/4 AE 131 (24.9) 104 (19.8) 63 (11.9) 118 (22.4)<br />

Dose reductions 6 (1.1) 0 6 (1.1) 2 (0.4)<br />

Discontinuations 5 (1.0) 0 1 (0.2) 3 (0.6)<br />

8088 General Poster Session (Board #37E), Mon, 1:15 PM-5:15 PM<br />

A prospective clinical study evaluating current models for risk <strong>of</strong> progression<br />

from smoldering multiple myeloma (SMM) to multiple myeloma (MM).<br />

Presenting Author: Benjamin M. Cherry, Multiple Myeloma Section, Metabolism<br />

Branch, National Cancer Institute, NIH, Bethesda, MD<br />

Background: The updated (2010) International Myeloma Working Group<br />

criteria emphasize individual follow-up and development <strong>of</strong> early treatment<br />

trials for high risk SMM patients. To facilitate these goals, it is important to<br />

have reliable models for risk <strong>of</strong> progression to MM derived from prospective<br />

clinical studies. Current clinical risk models by the Mayo Clinic and the<br />

Spanish PETHEMA group stratify SMM patients as low (LR), intermediate<br />

(IR), or high risk (HR) <strong>of</strong> progression using bone marrow plasma cell<br />

fraction and protein levels (Mayo) and abnormal plasma cell fraction on<br />

flow cytometry with or without immunoparesis (Spanish). We report the first<br />

comparison <strong>of</strong> these models in a prospective natural history study. Methods:<br />

We evaluated 70 patients with SMM and determined the risk classifications<br />

<strong>of</strong> each using the Mayo and Spanish models. P values <strong>of</strong> comparisons<br />

between risk groups were calculated using Fisher’s exact test. Results:<br />

Among SMM patients classified as HR by the Spanish model (n�31), 2<br />

patients (6%) were HR by the Mayo model; the remaining 17 (55%) and 12<br />

(39%) patients were classified as IR and LR by the Mayo model,<br />

respectively. Among 2 SMM patients classified as HR by the Mayo model,<br />

both patients (100%) were HR by the Spanish model. There was significant<br />

disagreement between the models in identifying HR patients (HR v.<br />

non-HR, p�0.0001). Similarly, <strong>of</strong> SMM patients classified as LR by the<br />

Spanish model (n�17), 11 patients (65%) were LR by the Mayo model; 6<br />

(35%) were IR and 0 (0%) were HR by the Mayo model. Among SMM<br />

patients classified as LR by the Mayo model (n�36), 11 (31%) patients<br />

were classified as LR by the Spanish model; 13 (36%) were IR and 12<br />

(33%) were HR by the Spanish model. There was significant disagreement<br />

between the models in identifying LR patients (LR v. non-LR, p�0.0016).<br />

Conclusions: The significant discordance between the Mayo Clinic and<br />

Spanish PETHEMA clinical risk models underscores the urgent need for<br />

biologically derived models that rely on molecular markers to predict<br />

disease progression. Such models are critical for counseling individual<br />

patients and for the development <strong>of</strong> early treatment studies that seek to<br />

prevent or delay progression to MM.<br />

Lymphoma and Plasma Cell Disorders<br />

8087 General Poster Session (Board #37D), Mon, 1:15 PM-5:15 PM<br />

Natural killer (NK) cell activation, cytokine production, and cytotoxicity in<br />

human PBMC/myeloma cell co-culture exposed to elotuzumab (Elo) alone<br />

or in combination with lenalidomide (Len). Presenting Author: Balaji<br />

Balasa, Abbott Biotherapeutics, Redwood City, CA<br />

Background: Elo is a monoclonal IgG1 antibody targeting CS1, a cell surface<br />

glycoprotein highly expressed on �95% <strong>of</strong> myeloma cells. In preclinical<br />

models Elo exerts anti-myeloma activity via NK cell-mediated antibodydependent<br />

cellular cytotoxicity. Len is an immunomodulatory agent that<br />

may activate NK cells. The combination <strong>of</strong> Elo � Len synergistically<br />

enhanced anti-tumor activity in myeloma xenograft models. We investigated<br />

the mechanism <strong>of</strong> enhancing NK cell activation and myeloma cell<br />

killing with Elo � Len. Methods: Human PBMC/OPM-2 co-cultures were<br />

treated for 24-72h with Elo, Len, or Elo � Len. Activation markers and<br />

adhesion receptors were evaluated by flow cytometry. Cytokines were<br />

measured by Luminex and ELISpot assays. Cytotoxicity was assessed by<br />

cell counting. Results: Elo � Len increased IFN-� secretion significantly<br />

more than Elo or Len alone. IFN-� elevates ICAM-1 expression, and ICAM-1<br />

surface expression on OPM-2 target cells increased synergistically with Elo<br />

� Len. Elo, Elo � Len but not Len increased expression <strong>of</strong> CD25 (IL-2R�)<br />

on NK cells. Len increased the levels <strong>of</strong> IL-2, but those were decreased in<br />

the presence <strong>of</strong> Elo due to increased consumption by CD25 expressing NK<br />

cells. Blocking uptake <strong>of</strong> IL-2 with anti-CD25 resulted in higher IL-2 levels<br />

than with Len. ELISpot assays confirmed that Elo � Len significantly<br />

increased the number <strong>of</strong> IL-2-producing cell colonies compared with Elo or<br />

Len. Elo induced NK dependent myeloma cell killing, and the effect was<br />

significantly higher with Elo � Len. Conclusions: Elo alone activated NK<br />

cells and mediated the killing <strong>of</strong> myeloma cells in PBMC/OPM-2 cocultures.<br />

Elo � Len synergistically enhanced myeloma cell killing and<br />

increased expression/production <strong>of</strong> IFN-�, ICAM-1, IL-2, and CD25.<br />

Treatment<br />

No blocking (control mAb) ave n�6 Anti-CD25 blocking mAb ave n�6<br />

IFN-� (pg/mL)<br />

ave n�8<br />

ICAM-1 on<br />

OPM-2 (MFI)<br />

n�4<br />

CD25 on<br />

NK cells<br />

(MFI) n�8<br />

IL-2 (pg/mL)<br />

(Luminex)<br />

# IL-2 positive<br />

colonies<br />

(ELISpot) n�9<br />

531s<br />

OPM-2<br />

killing (%)<br />

n�4<br />

*Control 48 427 155 21 - 30 1<br />

**Len 94 2040 131 50 - 71 15<br />

Elo � Len 386 42,259 1546 14 134 191 67<br />

Elo 65 3068 975 7.8 52 80 39<br />

*cIgG1 (isotype); **plus cIgG1.<br />

8089 General Poster Session (Board #37F), Mon, 1:15 PM-5:15 PM<br />

Costimulatory molecule pr<strong>of</strong>iles and NK cell recovery after autologous<br />

hematopoietic cell transplantation (HCT) in multiple myeloma (MM)<br />

patients. Presenting Author: Myo Htut, City <strong>of</strong> Hope, Duarte, CA<br />

Background: Increased immune responses post autologous HCT may be <strong>of</strong><br />

benefit in long term disease control. Responses may be mediated by NK<br />

cell function and possibly other alternate pathways, including costimulatory<br />

molecule pathways. This pilot study assesses the expression <strong>of</strong><br />

inhibitory (PD-1 and CTLA-4) and stimulatory (OX-40, ICOS, 4-1BB, and<br />

CD28) molecules on NK cells after auto-HCT in MM patients and evaluates<br />

the effect <strong>of</strong> lenalidomide treatment on these pathways. Methods: 17<br />

patients with MM undergoing HCT, median age 56.7 years (36 – 67), were<br />

included in the study. Peripheral blood samples were taken 3 days prior to<br />

HCT and 14, 30, 60, 90, 180 days after HCT. At d180 post-HCT, 13/17<br />

patients were receiving lenalidomide with d91 as median start date. NK<br />

cells and their costimulatory molecules were evaluated by flowcytometry<br />

using 2 six color panels <strong>of</strong> antibodies. One way ANOVA test and Kruskal-<br />

Wallis test (non-parametric) were applied to analyze the data using the<br />

Graphpad S<strong>of</strong>tware. Results: See table below. NK cell number was highest<br />

(median: 26% <strong>of</strong> total lymphocytes) at d14 (p: � 0.0001) compared to pre<br />

and post HCT levels. At d180, TNF-R OX40 expression was significantly<br />

increased in �PR group (n�5) (median: 9.5% <strong>of</strong> NK cells) compared to<br />

�VGPR (n�12) (0.8%; p�0.0084). In addition, NK cell number was<br />

higher in the lenalidomide group (n�13) (median: 15.15 % <strong>of</strong> total<br />

lymphocytes) compared to the no lenalidomide group (n�4) (6.74%;<br />

p�0.0108) at d180 post HCT. Significantly lower level <strong>of</strong> CTLA-4<br />

expression was also found in the lenalidomide group (0.33% vs. 2.54%;<br />

p�0.0362). Conclusions: We observed NK cell recovery to baseline values<br />

at 60 days after HCT. At d180 post-HCT, OX-40 expression in NK cells was<br />

higher in �PR group than �VGPR group. Lenalidomide treatment was<br />

associated with higher NK cells number and decreased expression <strong>of</strong><br />

CTLA-4. This observation could be a possible marker <strong>of</strong> enhanced host NK<br />

cell immune response against MM. Future clinical trials will explore<br />

therapies that increase NK cell responses.<br />

Pre-HCT d180<br />

Very good partial response or better (> VGPR) 6 12<br />

<strong>Part</strong>ial response or worse (< PR) 11 5<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


532s Lymphoma and Plasma Cell Disorders<br />

8090 General Poster Session (Board #37G), Mon, 1:15 PM-5:15 PM<br />

Retrospective analysis <strong>of</strong> second malignancies in patients with multiple<br />

myeloma. Presenting Author: Giampaolo Talamo, Penn State Hershey<br />

Cancer Institute, Hershey, PA<br />

Background: Recent data from patients with multiple myeloma (MM)<br />

enrolled in randomized clinical trials have shown an increased incidence <strong>of</strong><br />

second malignancies after treatment with lenalidomide, but the prevalence<br />

<strong>of</strong> second malignancies in the overall MM population is uncertain. Methods:<br />

We retrospectively analyzed the medical records <strong>of</strong> 320 consecutive MM<br />

patients followed at the Penn State Hershey Cancer Institute between<br />

2006 and 2010. We excluded from the analysis basocellular and squamocellular<br />

carcinomas <strong>of</strong> the skin. Results: Forty-three patients (13%) were<br />

found to have second malignancies, and 5 <strong>of</strong> them had a third cancer. One<br />

pt had 4 cancers. They included cancers <strong>of</strong> the prostate (8 pts), breast (8),<br />

MDS/leukemia (6), colon/rectum (5), melanoma (5), lung (4), uterus (4),<br />

bladder (3), kidneys (2), pancreas (2), testicle (1), myeloproliferative<br />

disorders (1), and sarcoma (1). Of 50 cancers, 36 (72%) developed before<br />

the diagnosis <strong>of</strong> MM, at a median <strong>of</strong> 65 months (range, 1-372), and 14<br />

after that, at a median <strong>of</strong> 37 months (range, 3-104). Lenalidomide was<br />

used in 239 (75%) patients, and in 9 <strong>of</strong> 14 cases <strong>of</strong> post-MM second<br />

malignancies. Conclusions: Second malignancies usually develop before<br />

the diagnosis <strong>of</strong> MM, i.e., MM is the second malignancy for the majority <strong>of</strong><br />

patients. The use <strong>of</strong> lenalidomide could not be indicated as a possible<br />

carcinogenic factor for the majority <strong>of</strong> MM patients with second malignancies.<br />

8092 General Poster Session (Board #38A), Mon, 1:15 PM-5:15 PM<br />

Second autologous stem cell transplantation as a strategy for management<br />

<strong>of</strong> relapsed multiple myeloma. Presenting Author: Wilson I. Gonsalves,<br />

Mayo Clinic, Rochester, MN<br />

Background: High dose therapy and autologous stem cell transplant (SCT)<br />

remain an integral part <strong>of</strong> the treatment <strong>of</strong> multiple myeloma (MM). Despite<br />

the introduction <strong>of</strong> several new drugs, disease relapse remains inevitable<br />

and a second SCT <strong>of</strong>ten allows continued disease control. Methods: We<br />

examined the outcomes in 105 patients (pts) undergoing a second SCT<br />

(SCT2) for relapsed MM, from among 1033 pts receiving initial transplant<br />

between 1994 and 2009. Patients receiving an allogeneic SCT post SCT2<br />

were not included in the survival analysis. Results: Median (range) age at<br />

SCT2 was 60 yrs (35-74) and the time between SCTs was 46 mos<br />

(10-130). The median follow-up was 5 yrs from SCT2. The median (range)<br />

number <strong>of</strong> regimens between the two SCTs and from diagnosis was 2 (0-7)<br />

and 3 (1-11), respectively. Conditioning regimen was Mel 200 in 85 (81%)<br />

pts. Median (range) CD34 cells infused was 4.6 (2.5-25 million/kg).<br />

Hospitalization was required for 53 pts (50%), median hospital stay was 7<br />

days.. Median time to ANC <strong>of</strong> 500 and platelets <strong>of</strong> 50,000 were 13 and 16<br />

days, respectively. Treatment related mortality was seen in 5 (5%) pts. A<br />

partial response or better was seen in 95 (90%) pts, including a stringent<br />

CR in 10%, CR in 19%, and VGPR in 21%. Stable disease or progression<br />

was seen in 7 pts and 3 pts died prior to disease assessment. The median<br />

(95% CI) PFS and OS after SCT2 were 10.4 (8, 14) and 33 (28, 51) mos,<br />

respectively. The median OS was not reached and 33 mos respectively, for<br />

those obtained a CR and those with �CR, P�0.03. FISH pre-SCT2 was<br />

available for 73 pts; the PFS and OS were not affected by the presence <strong>of</strong><br />

high-risk MM. In multivariate analyses, shorter duration <strong>of</strong> response to<br />

SCT1, higher plasma cell (PC) labeling index at SCT2, more regimens used<br />

prior to SCT2 all predicted shorter PFS and OS post SCT2. Lack <strong>of</strong> CR to<br />

SCT2 was also significantly associated with shorter PFS. Conclusions:<br />

Second SCT is an effective therapy for eligible pts relapsing after other<br />

treatments. It provides a meaningful duration <strong>of</strong> response and appears to be<br />

well tolerated. Pts with longer duration <strong>of</strong> response to first transplant and<br />

those achieving a CR appear to have maximum benefit.<br />

8091 General Poster Session (Board #37H), Mon, 1:15 PM-5:15 PM<br />

Predictive value <strong>of</strong> baseline serum MIP-1� and CRP on symptom burden<br />

and tumor response to induction therapy in patients with multiple myeloma.<br />

Presenting Author: Charles S. Cleeland, University <strong>of</strong> Texas M. D.<br />

Anderson Cancer Center, Houston, TX<br />

Background: Macrophage inflammatory protein-1�(MIP-1�) is a growth<br />

factor for human multiple myeloma (MM) cells. As an osteoclastic factor,<br />

its presence provides pathophysiological evidence <strong>of</strong> the development <strong>of</strong><br />

lytic bone lesions in MM. C-reactive protein (CRP) indicates systemic<br />

inflammation. The relationship between disease-driven inflammatory markers<br />

and both patient-experienced symptoms and tumor response to induction<br />

therapy is unknown. Methods: MM patients (N�39) who were either<br />

newly diagnosed or had received fewer than 2 cycles <strong>of</strong> chemotherapy, and<br />

who also were to receive induction therapy, were enrolled. To test<br />

concentrations <strong>of</strong> MIP-1� and CRP, serum samples were collected before<br />

and after induction and assayed by Luminex. Multiple symptoms were<br />

measured twice a week via the M. D. Anderson Symptom Inventory MM<br />

module (MDASI-MM) from -8 to �112 days <strong>of</strong> induction. The MM-specific<br />

items <strong>of</strong> the MDASI-MM are bone aches, constipation, muscle weakness,<br />

diarrhea, sore mouth or throat, rash, and difficulty paying attention.<br />

Correlation between symptom severity and inflammatory markers at baseline<br />

was examined by linear regression modeling. Kruskal-Wallis significance<br />

test and Wilcoxon test were used to examine association between<br />

MIP-1� and tumor response. Results: Patients received either bortezomibbased<br />

(89%) or lenalidomide-based induction therapy. Baseline MIP-1�<br />

and CRP were significantly inversely related to the mean severity component<br />

score <strong>of</strong> the 5 most-severe symptoms (fatigue, pain, bone aches, poor<br />

sleep, drowsiness) (p�.03; p�.02), and significantly inversely related to<br />

the severity <strong>of</strong> a component score <strong>of</strong> the module-specific symptoms<br />

(p�0.04; p�.002). Change over time in MIP-1� differed significantly by<br />

tumor response category (p�.04), with the partial response group having a<br />

higher median score than the complete response group (1.483 vs. 0.016,<br />

p�.01). Conclusions: Our data suggest that higher baseline levels <strong>of</strong> serum<br />

MIP-1� and CRP predict effective chemotherapy-induced reduction <strong>of</strong><br />

disease-related symptoms in MM patients. Higher serum MIP-1� expression<br />

after induction therapy was related to less-ideal tumor response.<br />

8093 General Poster Session (Board #38B), Mon, 1:15 PM-5:15 PM<br />

Prognostic value <strong>of</strong> serum lactate dehydrogenase in symptomatic multiple<br />

myeloma. Presenting Author: Krina K. Patel, University <strong>of</strong> Texas M. D.<br />

Anderson Cancer Center, Houston, TX<br />

Background: In patients with symptomatic multiple myeloma, the clinical<br />

features, responses to treatment, and survival times vary. Well established<br />

predictors <strong>of</strong> survival include the International Staging System (ISS),<br />

cytogenetic abnormalities, and response to therapy. Long recognized has<br />

been the association <strong>of</strong> high serum lactate dehydrogenase (LDH) with<br />

advanced disease and shorter survival. We focused here on the impact <strong>of</strong><br />

high LDH on staging and prognosis in order to guide the role <strong>of</strong> recent<br />

advances in therapy. Methods: We evaluated 1,247 patients with newly<br />

diagnosed, symptomatic myeloma from 10/74 to 7/11. Our goal was to<br />

determine the prognostic value <strong>of</strong> high LDH (�300 IU/L) in relation to ISS<br />

stage. We also compared the frequencies <strong>of</strong> anemia, hypercalcemia, and<br />

response to therapy in patients with high LDH with those <strong>of</strong> patients with<br />

Stage III disease and normal LDH values. Results: All 1,139 patients with<br />

normal LDH lived significantly longer than the 108 patients with elevated<br />

values (47 vs. 16 months, p �.01). LDH was elevated in 9% <strong>of</strong> all patients,<br />

but in 2%, 6%, and 18% <strong>of</strong> patients with ISS-I, II, and III disease,<br />

respectively. Their survival times were also significantly shorter than those<br />

<strong>of</strong> comparable patients in each stage with normal LDH (table). Among the<br />

108 patients with high LDH, the frequencies <strong>of</strong> hemoglobin �8.5 g/dl (54<br />

vs. 41%, p�.03), and serum calcium�11.5 mg/dl (41 vs. 27%, p�.01)<br />

were significantly higher than those <strong>of</strong> 292 patients with Stage III disease<br />

and normal LDH, and the frequency <strong>of</strong> response to therapy was less (40 vs.<br />

62%, p�.01). Conclusions: Serum lactate dehydrogenase provides a<br />

convenient and dependable prognostic indicator in patients with multiple<br />

myeloma. An elevated LDH value indicates a poor prognosis regardless <strong>of</strong><br />

ISS stage, confirming the report by Gkotzamanidou, Terpos, and Dimopoulos<br />

et al, and should be included in the definition <strong>of</strong> stage III disease. Such<br />

patients require rapid control <strong>of</strong> disease with sequential combinations <strong>of</strong><br />

effective drugs and intensive therapy in order to improve their outcome.<br />

Effect <strong>of</strong> elevated LDH on survival according to ISS stage (median months).<br />

ISS Stage < 300 IU/L >300 IU/L p<br />

I 56 16 .09<br />

II 49 21 �.01<br />

III 35 16 �.01<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


8094 General Poster Session (Board #38C), Mon, 1:15 PM-5:15 PM<br />

Predicting bortezomib-related severe neurologic adverse events by measuring<br />

proteasome activity in PBMCs. Presenting Author: Yukiko Cho, Ibaraki<br />

Prefectural Central Hospital, Ibaraki, Japan<br />

Background: Although a proteasome inhibitor bortezomib (BOR) is one <strong>of</strong><br />

the backbone drugs for the treatment <strong>of</strong> multiple myeloma, severe<br />

neurologic adverse event (sNE) is a major obstacle for continuing the<br />

treatment. As there is no clinically available biomarker for predicting the<br />

occurrence <strong>of</strong> sNAE, we measured the proteasome activity (PrsAtv) in<br />

peripheral blood mononuclear cells (PBMCs). Methods: Patients (Pts) were<br />

treated by either standard Q3wks or weekly (BOR 1.3mg/m2 : day 1, 8, 15,<br />

22; Q5wks) schedule depending on physician’s decision. PBMCs were<br />

collected from 21 Pts and 99 healthy volunteers. Pts’ samples were<br />

collected before treatment, 1 hour after the 1st injection <strong>of</strong> BOR, and at the<br />

end <strong>of</strong> the 1st course. PrsAtv in PBMCs was measured by using a specific<br />

substrate SUC-LLVY-AMC, which becomes fluorescent upon cleavage by<br />

proteasome. Results: Levels <strong>of</strong> PrsAtv among volunteers were highly<br />

variable with no certain correlation with age and sex. Pretreatment levels <strong>of</strong><br />

PrsAtv among Pts were also variable, and didn’t correlate with the<br />

occurrence <strong>of</strong> sNAE. After 1 hour <strong>of</strong> BOR injection, PrsAtv decreased to<br />

33.2�20.5% (mean � SD) <strong>of</strong> the pretreatment level, and it generally<br />

recovered (112.5�77.6%) at the end <strong>of</strong> the 1st course. However, in 12 out<br />

<strong>of</strong> 21 Pts, it didn’t recover to � 100%. Among these Pts, 5 manifested with<br />

sNAE (�G3) during the 2nd or 3rd course <strong>of</strong> the chemotherapy. In a sharp<br />

contrast, no patients whose PrsAtv recovered to �100% manifested with<br />

sNAE. Pts who manifested with sNAE (5 Pts) showed the lower levels <strong>of</strong><br />

PrsAtv at the end <strong>of</strong> the 1st course than those without sNAE (16 Pts)<br />

(67.4�11.8 vs 126.6�83.8%, p�0.075). It should be also strengthened<br />

that all the Pts who manifested with sNAE were treated by the standard<br />

Q3wks schedule, although levels <strong>of</strong> PrsAtv at the end <strong>of</strong> the 1st course were<br />

not significantly different between Q3 and Q5wks groups (109.0�68.3 vs<br />

121.1�96.4 %, p�0.38). Contrary, there was no significant difference <strong>of</strong><br />

the bortezomib response between these two treatment groups. Conclusions:<br />

Pts whose PrsAtv does not recover to �100% at the end <strong>of</strong> the 1st course<br />

are at high risk <strong>of</strong> manifesting with sNAE, and these Pts should not be<br />

treated by the standard Q3wks schedule in the subsequent courses.<br />

8096 General Poster Session (Board #38E), Mon, 1:15 PM-5:15 PM<br />

Long-term outcome with lenalidomide and dexamethasone therapy for<br />

newly diagnosed multiple myeloma. Presenting Author: Geetika Srivastava,<br />

Mayo Clinic, Rochester, MN<br />

Background: The combination <strong>of</strong> lenalidomide and dexamethasone (Len-<br />

Dex) is a commonly used initial therapy for newly diagnosed multiple<br />

myeloma. While the short-term outcomes with respect to response and<br />

toxicity is well-known, long-term outcome with this combination as initial<br />

therapy is not well described. Methods: We studied 286 consecutive<br />

patients with newly diagnosed MM seen at our institution, who received<br />

initial therapy with Len-Dex, and who had complete follow up records. Data<br />

regarding the clinical course was obtained from medical records. Results:<br />

The median (range) age at diagnosis was 63 (28-92) yrs; 166 (58% were �<br />

65 yrs and175 (61%) were male. The median estimated follow-up was 3.9<br />

yrs (95% CI, 3.4, 4.2) and 203 (71%) pts were alive at the time <strong>of</strong> last<br />

follow up. The median estimated duration on Len-Dex was 5.3 mos (95%<br />

CI, 4.6, 6.4). The best overall response (�PR) was 72%, including 26%<br />

with VGPR or better and 14 (5%) not being evaluable for a response. At last<br />

follow up, 41 (14%) patients were continuing on therapy. There were 93<br />

pts (32%) who stayed on therapy for 12 months or more. Among these<br />

patients, the ORR was 86%, including 45% with VGPR or better. The<br />

median overall survival (OS) for the entire cohort from diagnosis was 7.4 yrs<br />

(95% CI; 5.8, NR) and the estimated 5-yr survival was 67%. There were 16<br />

(5.5%) pts who died within a year <strong>of</strong> diagnosis. The median time to first<br />

disease progression, irrespective <strong>of</strong> transplant status, was 30.2 mos (95%<br />

CI, 25, 42). Overall, 143 (50%) <strong>of</strong> the patients have gone to stem cell<br />

transplant. Censoring those patients who proceeded to SCT prior to relapse<br />

at the time <strong>of</strong> BMT, the median TTP was 25.5 mos (95% CI, 22, 29). The<br />

median OS was 7.4 yrs for those �65 yrs, compared with 6.2 yrs for the<br />

older patients (P�0.01). The 5-yr OS estimate for patients in ISS stage 1,<br />

2 and 3 were 82, 65, and 44 months respectively. Conclusions: The current<br />

study provides long-term estimates <strong>of</strong> responses and survival in a series <strong>of</strong><br />

patients treated initially with lenalidomide and dexamethasone. The<br />

median survival <strong>of</strong> nearly 8 years reflects the efficacy <strong>of</strong> the novel agents<br />

both at diagnosis and at relapse and confirms the survival improvements<br />

seen in MM in the last decade.<br />

Lymphoma and Plasma Cell Disorders<br />

533s<br />

8095 General Poster Session (Board #38D), Mon, 1:15 PM-5:15 PM<br />

Regional differences in the treatment approaches for relapsed multiple<br />

myeloma: An IMF study. Presenting Author: Brian G. Durie, Cedars-Sinai<br />

Comprehensive Cancer Center, Los Angeles, CA<br />

Background: Multiple myeloma (MM) remains incurable and invariably<br />

relapses after initial therapy. There is no uniform approach to management<br />

<strong>of</strong> relapsed MM and is dictated by type <strong>of</strong> initial therapy and the response<br />

seen, and availability <strong>of</strong> new drugs. The outcomes associated with current<br />

approaches have not been systematically examined. Methods: We enrolled<br />

383 patients (pts) with myeloma who had a documented relapse between<br />

Jan 2007 and June 2010, including 220 from Europe, 106 from Asia, and<br />

31 from South America and 26 from North America. Time Zero (T0) was<br />

defined as date <strong>of</strong> treatment after first relapse. Results: Across the study,<br />

61% were male and 49% were over 65 years at T0. ISS stage distribution at<br />

diagnosis included 26, 40 and 33% <strong>of</strong> patients in stages 1, 2 and 3,<br />

respectively. Among regimens used at first relapse; bortezomib (Btz)<br />

containing regimens were most common (54%), followed by lenalidomide<br />

(Len, 25%) and cyclophosphamide (21%). The overall response rate<br />

(��PR) to first therapy after relapse was 58% including 14% with a CR.<br />

There was a progressive decrease in the response rates with successive<br />

regimen; 45%, 30% and 15% for regimens 2, 3 and 4 respectively. Len<br />

use was considerably lower in the Asian cohort, while Btz use was<br />

comparable across the regions. The median PFS from T0 was 13 mos and<br />

OS was 35 mos, for the entire cohort. The PFS to first salvage regimen was<br />

similar across the regions, while OS was shorter for the Asian and South<br />

<strong>American</strong> cohorts. In a univariate analysis, ISS stage 3, presence <strong>of</strong><br />

cytogenetic abnormalities, history <strong>of</strong> plasma cell leukemia or extramedullary<br />

disease (EMD), bone marrow PC% � 33% and presence <strong>of</strong> renal<br />

insufficiency were all associated with a shorter PFS as well as shorter OS<br />

(P��0.01). In a multivariate model, ISS stage 3 and presence <strong>of</strong> EMD<br />

were most associated with short OS. Conclusions: Median PFS for current<br />

second line regimens, typically containing Btz or Len appear to be 1 yr with<br />

an OS from first relapse <strong>of</strong> about 3 yrs. The OS from first relapse is nearly<br />

double that seen in a cohort <strong>of</strong> patients in first relapse prior to introduction<br />

<strong>of</strong> new drugs. Clear-cut regional differences can be seen in terms <strong>of</strong><br />

patterns <strong>of</strong> drug use and likely reflect drug availability and healthcare costs.<br />

8097 General Poster Session (Board #38F), Mon, 1:15 PM-5:15 PM<br />

<strong>Clinical</strong> pr<strong>of</strong>ile <strong>of</strong> multiple myeloma in Asia: An Asian Myeloma Network<br />

(AMN) study. Presenting Author: Jae Hoon Lee, Department <strong>of</strong> Internal<br />

Medicine, Gachon University Gil Hospital, Incheon, South Korea<br />

Background: The incidence <strong>of</strong> multiple myeloma (MM) is known to be<br />

variable according to ethnicity. However the difference <strong>of</strong> clinical characteristics<br />

between ethnic groups is not well-defined. In Asian countries the<br />

incidence <strong>of</strong> MM has been lower compared with Western countries.<br />

However, there are growing evidences that MM is increasing very rapidly in<br />

this region. Until now, only few data <strong>of</strong> Asian patients has been reported.<br />

Asian myeloma network (AMN) decided to analyze the first multinational<br />

project to explore clinical characteristics <strong>of</strong> Asian MM patients and clinical<br />

practice performed in Asian countries. Methods: Data were collected from<br />

21 centers from 7 countries and regions were collected retrospectively.<br />

<strong>Clinical</strong> characteristics <strong>of</strong> 2969 symptomatic MM patients at diagnosis<br />

were described. Overall survival (OS) and prognostic factors were analyzed<br />

for 2273 patients who have survival data. Results: Median OS was 54<br />

months (95% CI 48.0-60.0). Patients who were diagnosed before 2000<br />

were shorter survival. Transplantation was performed to 513 patients with<br />

better survival (84 vs. 45 months, p�0.001). First line treatment <strong>of</strong> 2339<br />

evaluable patients was analyzed. Overall response rate was 71% with 30%,<br />

VGPR or better. New drugs including bortezomib, thalidomide or lenalidomide<br />

were combined for 32.5% <strong>of</strong> all 2339 patients without difference in<br />

response rate or OS according to combination. Conclusions: We successfully<br />

described clinical characteristics <strong>of</strong> Asian MM patients and this<br />

project will be the base for future studies or clinical trials for Asian MM<br />

patients. Updated analyses and comparison with Western data will be<br />

presented. AMN, supported by the International Myeloma Foundation<br />

(IMF) IMWG initiative.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


534s Lymphoma and Plasma Cell Disorders<br />

8098 General Poster Session (Board #38G), Mon, 1:15 PM-5:15 PM<br />

A phase I/II study <strong>of</strong> carfilzomib (CFZ) as a replacement for bortezomib<br />

(BTZ) for multiple myeloma (MM) patients (Pts) progressing while receiving<br />

a BTZ-containing combination regimen. Presenting Author: James R.<br />

Berenson, Institute for Myeloma and Bone Cancer Research, West Hollywood,<br />

CA<br />

Background: Recent data has shown that single-agent CFZ can produce<br />

responses among MM pts refractory to previous treatment regimens<br />

including those containing BTZ. We conducted an intrapatient Phase 1/2<br />

trial investigating the safety and efficacy <strong>of</strong> CFZ as a replacement for BTZ in<br />

BTZ-containing regimens to which pts have progressed. Methods: Eligible<br />

pts had to have progressed while receiving their most recent BTZcontaining<br />

regimen after at least 4 doses <strong>of</strong> BTZ at � 1.0 mg/m² in � 4<br />

weeks per cycle. Combination regimens containing an alkylating agent,<br />

anthracycline, or a glucocorticosteroid were eligible. CFZ replaced BTZ in<br />

each regimen via intravenous administration over 30 min on days 1, 2, 8, 9,<br />

15, and 16 <strong>of</strong> each cycle. Treatment continued using the same dose(s) and<br />

schedule(s) <strong>of</strong> each drug administered in the BTZ-containing regimen. CFZ<br />

doses were escalated on each <strong>of</strong> the first 4 cycles from 20 to 27, 36, and 45<br />

mg/m² or until a maximum tolerated dose (MTD) was reached for that<br />

regimen. Results: Of 19 enrolled pts 13 are evaluable to date and 6 have<br />

recently started treatment. Pts received a median <strong>of</strong> 7 (range, 1-18) prior<br />

treatments and 5 (range, 1-5) different BTZ-containing regimens. Pts were<br />

treated with CFZ and the following different combinations: bendamustine<br />

(BEND) alone, BEND � methylprednisolone, dexamethasone (DEX) alone,<br />

DEX � pegylated liposomal doxorubicin, ascorbic acid � cyclophosphamide,<br />

and melphalan alone. Pts have completed a median <strong>of</strong> 3 cycles.<br />

<strong>Clinical</strong> benefit was seen in 10 (77%) pts (complete response � 8%; very<br />

good partial response � 8%; partial response � 31%; minor response �<br />

31%) with another 23% showing stable disease. The median time to<br />

progression (range: 2-8 months) has not been reached and only 2 pts have<br />

progressed. The most common grade 3/4 adverse events were thrombocytopenia<br />

occurring in 5 pts (all � grade 3 except 1 event) and fever occurring<br />

in two pts (grade 3). Four pts experienced a serious adverse event but no<br />

regimen has reached a MTD. Conclusions: These early results suggest that<br />

CFZ is an effective and tolerable replacement for BTZ for pts who are<br />

refractory to BTZ-containing combination regimens.<br />

8100 General Poster Session (Board #39A), Mon, 1:15 PM-5:15 PM<br />

Survival outcomes <strong>of</strong> early autologous stem cell transplant (ASCT) followed by<br />

lenalidomide, bortezomib, and dexamethasone (RVD) maintenance in patients<br />

with high-risk multiple myeloma (MM). Presenting Author: Jonathan L. Kaufman,<br />

Winship Cancer Institute, Emory University, Atlanta, GA<br />

Background: Despite markedly improved survival rates for MM pts in the last<br />

decade, 15-20% <strong>of</strong> pts with high risk genetics continue to have dismal outcomes<br />

with a median PFS <strong>of</strong> 18.5 months (Kapoor P et al). In this subset <strong>of</strong> pts, efforts<br />

are needed to improve response rates and prolong the response duration, but<br />

doing so without genotoxic therapy as this has been shown not to improve<br />

outcomes (Barlogie et al). Methods: We evaluated 37 pts with high risk features<br />

[del17p (n�16), t(4;14) (n�1), t(14;16) (n�4) by FISH/CTG; hypodiploidy<br />

(n�9), del 13 (n�16); complex karyotype (n�14) by CTG; PCL (n�7) and<br />

atypical presentation (n�4)]. After completing induction therapy, all pts<br />

underwent ASCT followed by RVD maintenance. 60 days after ASCT following<br />

hematological recovery, pts began maintenance [lenalidomide 10 mg/day<br />

(days1–21), bortezomib 1.3 mg/m2 and dexamethasone 40 mg once a week<br />

(days 1, 8, 15) every 28 days]. Results: The response rates are summarized in the<br />

table. 7/36 pts progressed while on RVD maintenance. The median PFS and OS<br />

for pts on RVD maintenance has not been reached. Pts with �VGPR pre-ASCT<br />

and with �VGPR on RVD maintenance have median PFS <strong>of</strong> 28 months and 11<br />

months, respectively. 4 pts with prior h/o DVT received anticoagulation, while all<br />

others received ASA for DVT prophylaxis. No thrombotic events were seen. There<br />

were no grade 3/4 toxicities or treatment-related mortality. The most common<br />

toxicities during maintenance schedule were: PN-40% (G1: 26%; G2:14%); G1<br />

rash-10% and G1 fatigue in 78% pts. Cytopenias were seen in 25% pts and dose<br />

reductions were made in 50% pts. There is no report <strong>of</strong> secondary malignancies.<br />

Conclusions: Early ASCT followed by RVD maintenance delivers superior response<br />

rates in this high risk segment achieving sCR in 47% and �VGPR in 73%<br />

pts and prevents early relapses. The median PFS and OS have not been reached.<br />

RVD maintenance regimen is well tolerated and promising.<br />

Response rates.<br />

Best response with<br />

Pre-ASCT response Post-ASCT response RVD maintenance<br />

n % n % n %<br />

sCR 3 8 8 22 17 47<br />

CR 3 8 8 22 5 14<br />

sCR � CR 6 16 16 44 22 51<br />

VGPR 13 35 16 43 8 22<br />

>VGPR 19 51 32 77 30 73<br />

PR 15 41 5 14 3 8<br />

>PR 34 92 37 91 33 81<br />

SD 1 3<br />

PD 2 6 3 8<br />

8099 General Poster Session (Board #38H), Mon, 1:15 PM-5:15 PM<br />

Effect <strong>of</strong> matrix metallopeptidase 13 (MMP13) on multiple myeloma (MM)<br />

cells, osteoclast (OCL) activity, and bone resorption. Presenting Author:<br />

Jing Fu, Columbia University Medical Center, Department <strong>of</strong> Medicine/<br />

Hematology-Oncology, New York, NY<br />

Background: MM cells produce OCL-activating factors that induce excessive<br />

bone resorption resulting in lytic lesions. The role <strong>of</strong> MMPs in invasion/<br />

progression <strong>of</strong> solid tumors is well-known, but its function in MM has not<br />

been well elucidated. Our group has shown that MMP13 is highly expressed<br />

in primary MM cells and in sera <strong>of</strong> MM patients. Levels <strong>of</strong> MMP13<br />

significantly correlate with the extent <strong>of</strong> bone disease. MMP13 is induced<br />

by IL-6 via AP-1 activation in MM cells and enhances fusion <strong>of</strong> OCL<br />

precursors resulting in excessive bone resorption. OCL formation using<br />

MNCs <strong>of</strong> mmp-13-/- mice resulted in a fusion defect, significantly decreased<br />

OCL size and activity, which could be reversed by exogenous<br />

MMP13 (ASH 2009, IMW 2011). Methods: Methods will be presented in<br />

the Results section. Results: RT-PCR and western blotting revealed that<br />

IL-6 treatment <strong>of</strong> MM cells induced MMP13 transcription (30-fold) and<br />

secretion (�1000-fold). Protein expression <strong>of</strong> the AP-1 members c-Jun<br />

and c-Fos was induced by IL-6, which correlated with MMP13 upregulation.<br />

Our data further indicate that the catalytic activity <strong>of</strong> MMP13 is not<br />

required to enhance OCL formation and bone resorption. To prove this, we<br />

generated the MMP13 activity-dead mutation MMP13-E223A construct by<br />

site-directed mutagenesis PCR-based cloning. The mutated protein was<br />

overexpressed in HEK293 cells and purified from the supernatant to<br />

confirm whether loss <strong>of</strong> catalytic activity blocks MMP13 function. To<br />

further investigate the in vivo role <strong>of</strong> MMP13 in MM bone disease, MMP13<br />

expression was knocked down (KD) in murine 5TGM1-MM cells by pKLO. 1<br />

puro lentiviral infection containing sh-RNA targeting mouse MMP13<br />

sequence. MMP13-KD 5TGM1-MM cells or WT-5TGM1-MM cells were<br />

intratibially injected into RAG2-/- mice. Development <strong>of</strong> lytic bone lesions<br />

are monitored by micro-QCT and data will be available at the time <strong>of</strong><br />

presentation. Conclusions: Our data suggest that MMP13, secreted by MM<br />

cells, plays a critical role in the development <strong>of</strong> lytic lesions. Targeting<br />

MMP13 represents a promising approach to treat or to prevent bone<br />

disease in MM.<br />

8101 General Poster Session (Board #39B), Mon, 1:15 PM-5:15 PM<br />

Efscalefect <strong>of</strong> lenalidomide, bortezomib, and dexamethasone (RVD) induction<br />

therapy in transplant-eligible patients (Pts) with newly diagnosed<br />

multiple myeloma (MM) on CR rates and survival. Presenting Author:<br />

Charise Gleason, Winship Cancer Instiute, Emory University, Atlanta, GA<br />

Background: Addition <strong>of</strong> lenalidomide to bortezomib and dexamethasone<br />

(RVD) demonstrated to be an effective and well tolerated regimen in phase<br />

II trials with overall response rate (ORR) �95% (Richardson P et al). Given<br />

the lack <strong>of</strong> phase III data, we have evaluated our institutional experience <strong>of</strong><br />

pts treated with RVD induction therapy, to support this triplet combination.<br />

Methods: 286 transplant-eligible pts with newly diagnosed MM were<br />

treated with RVD induction therapy [R - 25 mg/day (days1–14), V-1.3<br />

mg/m2 (days 1, 4 8, 11) andD-40mgonce/twice weekly as tolerated every<br />

21 days] from January 2008 until January 2012. 148 pts underwent ASCT<br />

and 138 pts opted for delayed transplant. Post-ASCT 56% pts are on<br />

maintenance therapy tailored to their risk (R-40%; RVD-10%; V-6%).<br />

Demographic and outcomes data for the pts that underwent ASCT were<br />

collected and responses were evaluated per IMWG Uniform Response<br />

Criteria. Results: Median age <strong>of</strong> the pts is 60 years (range 32-77). Other pt<br />

characteristics include: M/F 55%/45%; ISS I/II/III 40%/30%/30%; Isotype<br />

IgG/IgA/FLC/IgM 61%/20%/18%/1%; high risk/standard risk 13%/<br />

87%. Pts received a median <strong>of</strong> 4 cycles (2-9) <strong>of</strong> RVD. Median CD34� stem<br />

cell collection was 11.24 x 10 6 /kg. 18% pts required dose reductions<br />

(R/V/D-5%/9%/2%) and discontinuation in 2% pts for progressive disease.<br />

49%/8% pts had G1-2/G3-4 PN. Median estimated PFS is 47 months and<br />

median OS has not been reached. Response rates are included in the table.<br />

Conclusions: RVD is an active induction regimen with superior response<br />

rates <strong>of</strong> � 80% �VGPR rates post-ASCT and is well tolerated in newly<br />

diagnosed MM pts. Incorporation <strong>of</strong> lenalidomide did not impact the stem<br />

cell collection. Until phase III data are available, our institutional experience<br />

could provide a perspective in the choice <strong>of</strong> RVD as an effective<br />

induction regimen in improving ORR and prolonging survival.<br />

Post-induction response Post-ASCT response Best response<br />

N (148) % N (146) % N (142) %<br />

sCR 27 18 50 35 81 57<br />

CR 10 7 18 12 8 6<br />

nCR 15 10 12 8 10 7<br />

sCR � CR � nCR 52 35 80 55 99 70<br />

VGPR 40 27 36 25 26 18<br />

>VGPR 92 62 116 80 125 88<br />

PR 48 33 25 17 13 9<br />

>PR 140 95 141 97 138 97<br />

MR 3 2<br />

PD 5 3 5 3 4 3<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


8102 General Poster Session (Board #39C), Mon, 1:15 PM-5:15 PM<br />

Association <strong>of</strong> bone marrow plasma cell infiltration pre-auto transplant<br />

with adverse outcomes in multiple myeloma. Presenting Author: Qaiser<br />

Bashir, University <strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: Auto-stem cell transplantation (SCT) has become the standard<br />

<strong>of</strong> care for eligible patients (pts) with multiple myeloma (MM). However,<br />

the impact <strong>of</strong> bone marrow (BM) plasma cell (PC) percentage before SCT is<br />

not yet known. Methods: Retrospective review <strong>of</strong> 1489 MM pts who<br />

underwent auto-SCT from 7/8/98 – 12/31/2010 with post-induction,<br />

pre-SCT BM biopsy information available. Pts were divided into 2 groups:<br />

�10% PC infiltration (“PC low”) and �10% PC infiltration (“PC high”).<br />

Progression-free (PFS) and overall (OS) survivals were estimated by the<br />

Kaplan-Meier method. Log-rank test was performed to test differences in<br />

survival. Results: 1489 pts were studied. 1174 pts had �10% involvement<br />

<strong>of</strong> BM by PCs and 315 had � 10% involvement. For pts in the PC low<br />

group, 32% had a CR, 20% had a VGPR, 31% had a PR, 13% had �PR<br />

and 3% had progressive disease (PD) after SCT. For pts in the PC high<br />

group, 11% had a CR, 14% had a VGPR, 48% had a PR, 21% had �PR<br />

and 5% had PD after SCT. Median PFS was significantly shorter for the PC<br />

high group vs the PC low group (24.8 vs 29.5 months, p�0.05), as was<br />

median OS (52.5 vs 79.4 months respectively, p�0.001). When only pts<br />

who had a PR to induction were examined, there was a significant<br />

difference in both PFS (24.4 vs 33.2 months, p�0.04) and OS (58.3 vs<br />

81.2 months, p �0.002) for the PC high vs PC low groups, respectively. For<br />

the 1299 (87%) pts treated in the era <strong>of</strong> novel therapeutics (after 2000),<br />

the differences between the PC high and PC low groups were maintained for<br />

both PFS (24.4 vs 29.5 months respectively (p�0.029)) and OS (54.8 vs<br />

88.4 months respectively, p�0.001). Chemo-mobilization before SCT did<br />

not improve PFS or OS but this was done in only 44 (14%) <strong>of</strong> PC high pts.<br />

Conclusions: PC BM infiltration before auto-SCT is associated with a worse<br />

outcome. This finding persists in pts with a PR before SCT. Thus BM<br />

disease burden may further stratify pts with a PR. Though additional<br />

therapy did not significantly change the outcome for pts with high PC<br />

burden, this was done only in a minority <strong>of</strong> pts. Additionally, differences<br />

between PC high and PC low groups are maintained despite new salvage<br />

agents over the last 10 years. Further prospective study is warranted to<br />

determine the true impact <strong>of</strong> BM PC infiltration.<br />

8104 General Poster Session (Board #39E), Mon, 1:15 PM-5:15 PM<br />

Role <strong>of</strong> immune-related conditions in smoldering myeloma and MGUS.<br />

Presenting Author: Mary Kwok, Metabolism Branch, National Cancer<br />

Institute, National Institutes <strong>of</strong> Health, Bethesda, MD<br />

Background: Recent guidelines emphasize tailored follow-up and the need<br />

for clinical trials for high-risk smoldering myeloma (SMM). Emerging<br />

evidence from epidemiological studies suggests that immune-related<br />

conditions play a role in the causation <strong>of</strong> myeloma precursor disease (SMM<br />

and monoclonal gammopathy <strong>of</strong> undetermined significance; MGUS) and<br />

are <strong>of</strong> clinical importance for the risk <strong>of</strong> developing multiple myeloma. The<br />

aim <strong>of</strong> our study is to assess whether there is an altered biology in<br />

SMM/MGUS patients with preceding immune-related conditions. Methods:<br />

From our ongoing prospective SMM/MGUS natural history study, we<br />

evaluated 56 SMM and 60 MGUS patients. Information on autoimmunity<br />

was identified at baseline. All patients underwent extensive clinical and<br />

molecular characterization. At baseline, all patients underwent bone<br />

marrow biopsy evaluation using immunohistochemistry and multi-color<br />

flow cytometry <strong>of</strong> plasma cells. We assessed expression patterns <strong>of</strong> adverse<br />

plasma cell markers (CD56 and CD117), and applied risk models based on<br />

serum immune markers and bone marrow findings. Results: Among enrolled<br />

SMM and MGUS patients, 7 (12%) and 9 (15%) had a preceding<br />

autoimmune disorder. We found SMM patients with (vs. without) a<br />

preceding autoimmune disorder to have a substantially lower rate <strong>of</strong> CD56<br />

(28% vs. 61%) and CD117 (28% vs. 61%) expressing plasma cells. When<br />

we compared the same markers in MGUS patients, CD56 and CD117<br />

expression patterns were similar among patients with vs. without preceding<br />

autoimmunity (10% vs. 17%, and 50% vs. 48%). Using the Mayo Clinic<br />

risk model, none <strong>of</strong> the SMM patients with a preceding autoimmune<br />

disorder had high-risk features; in contrast, 3/41 (7%) <strong>of</strong> those without a<br />

preceding autoimmune disorder were high-risk SMM. Using the Mayo<br />

Clinic risk model, none <strong>of</strong> the MGUS patients were high-risk independent <strong>of</strong><br />

autoimmune status. Conclusions: Our prospective clinical study found<br />

SMM patients with preceding immune-related conditions to have less<br />

adverse biology, supportive <strong>of</strong> epidemiological studies suggesting the risk<br />

<strong>of</strong> developing multiple myeloma is substantially lower in these patients.<br />

Lymphoma and Plasma Cell Disorders<br />

535s<br />

8103 General Poster Session (Board #39D), Mon, 1:15 PM-5:15 PM<br />

Patterns <strong>of</strong> nonhematologic malignancies in a population <strong>of</strong> African<br />

<strong>American</strong> veterans with monoclonal gammopathy <strong>of</strong> unknown significance<br />

(MGUS). Presenting Author: Hady Ghanem, George Washington University,<br />

Washington, DC<br />

Background: Monoclonal Gammopathy <strong>of</strong> Unknown Significance (MGUS) is<br />

characterized by the presence <strong>of</strong> a monoclonal immunoglobulin in the<br />

serum or the urine with no evidence <strong>of</strong> hematologic malignancy. A possible<br />

relationship between MGUS and increased incidence <strong>of</strong> NHM has been<br />

suggested in Caucasian populations. However, data in African <strong>American</strong>s<br />

with MGUS are lacking. Methods: Non-MGUS controls were selected<br />

randomly from patients who did not have a paraprotein detected on<br />

electrophoresis (NMGUS) and were matched 2-to-1 to MGUS cases.<br />

Descriptive statistics and comparisons are presented to compare MGUS<br />

and NMGUS groups. Results: 492 male patients with MGUS patients were<br />

matched with 984 male NMGUS patients. 451 patients had abnormal<br />

serum protein studies (91.6%) and 40 had light chain disease (8.4%).<br />

Median age at diagnosis <strong>of</strong> MGUS was 68 years (28-81). 144 MGUS<br />

patients (29.2%) and 296 NMGUS patients (30%) had 1 or more NHM.<br />

The median age <strong>of</strong> diagnosis <strong>of</strong> 1st NHM was 70 (25-94) in the MGUS<br />

group and 68.4 in the NMGUS group (34.5-94.4). 19 MGUS patients<br />

(3.8%) and 27 NMGUS patients had 2 different types <strong>of</strong> NHM (2.7%). 1<br />

MGUS patient (0.2%) and 3 NMGUS patients (0.3%) had 3 NHM. 57<br />

patients had MGUS before NHM (11.5%) and 69 patients were diagnosed<br />

with MGUS after the diagnosis <strong>of</strong> NHM (14%), and median differences<br />

between diagnosis <strong>of</strong> MGUS and 1st NHM were 4 years (1-12 years) and 5<br />

years (1-38 years) respectively. Types <strong>of</strong> NHM were comparable, and<br />

prostate cancer was the most prevalent NHM in both groups (15% <strong>of</strong> MGUS<br />

patients and 17% <strong>of</strong> NMGUS patients). Median time <strong>of</strong> follow up was 49.3<br />

months for MGUS patients and 35.2 months for NMGUS patients 140 <strong>of</strong><br />

the MGUS patients (28.4%) and 214 non-MGUS patients (21.75%) had<br />

died at data cut-<strong>of</strong>f. Conclusions: Based on these observational data,<br />

prevalence and types <strong>of</strong> NHM appear to be comparable in MGUS and<br />

NMGUS African <strong>American</strong> patients. All cause mortality appears to be<br />

higher for NHM patients if they had MGUS. This pattern will need to be<br />

verified prospectively in a larger group <strong>of</strong> patients.<br />

8105 General Poster Session (Board #39F), Mon, 1:15 PM-5:15 PM<br />

Does stage migration exist in active multiple myeloma (MM)? Presenting<br />

Author: Prashant Kapoor, Mayo Clinc, Rochester, MN<br />

Background: Tumor stage migration can artifactually inflate cancer survival<br />

rates and overestimate benefits <strong>of</strong> newer therapies. We have previously<br />

shown a favorable impact <strong>of</strong> widely used novel agents in MM patients.<br />

However, it is not known whether use <strong>of</strong> well tolerated and easily<br />

administrable novel agents results in their introduction at lower levels <strong>of</strong><br />

MM burden (and therefore add to improved outcome). Our goal was to<br />

assess for stage migration in newly diagnosed active MM patients. Methods:<br />

We reviewed records <strong>of</strong> 1,467 patients with active MM at Mayo Clinic, at<br />

initiation <strong>of</strong> therapy from 3 consecutive 5-year intervals: 01/1996-12/<br />

2000 (Group 1), 01/2001-12/2005 (Group 2), 01/2006- 12/2010 (Group<br />

3). These intervals reflect the practice changing approaches at our center<br />

transitioning from the use <strong>of</strong> alkylator-based to novel agent-based initial<br />

therapy. Traditional parameters and staging systems were used to estimate<br />

tumor burden. We performed one way analysis <strong>of</strong> variance (ANOVA) to<br />

assess for differences in these parameters among the groups. A p-value <strong>of</strong><br />

�0.05 was considered significant. Results: Group 3 shows an upward trend<br />

for hemoglobin (Hgb), and lower creatinine and bone marrow plasma cell<br />

(BMPC) percent. Compared to other groups, a greater proportion <strong>of</strong> Group 3<br />

patients are assigned to lower Durie Salmon (DS) stages (Table) suggesting<br />

reduced tumor burden at therapy initiation. In contrast, the International<br />

Staging System (ISS) which is not used for decisions regarding therapy<br />

initiation divides cohorts in similar proportions. Conclusions: Stage migration<br />

is evident in our cohort <strong>of</strong> active MM patients presenting in the time<br />

periods <strong>of</strong> evolving initial therapeutic strategies. Future studies should take<br />

into account the bias introduced by this phenomenon in interpreting<br />

survival analysis <strong>of</strong> MM patients.<br />

Group 1 (N�432) Group 2 (N�480) Group 3 (N�555)<br />

1996-2000<br />

2001-2005<br />

2006-2010<br />

Parameters Median IQ range Median IQ range Median IQ range P value<br />

Hgb (g/dL) 10.9 9.6-12.1 10.9 9.5-12.1 11 9.8-12.6 0.01<br />

Creatinine (mg/dL) 1.3 1-1.6 1.2 1-1.6 1 0.8-1.3 �0.0001<br />

BMPC (%) 37 17-59 32 14-57 29 13-51 0.02<br />

Calcium (mg/dL) 9.5 9.1-10.2 9.6 9-10.1 9.6 9.1-10.1 NS<br />

DS stage (%)<br />

1 10 13 17 0.0003*<br />

2 06 14 12<br />

3 84 73 71<br />

ISS (%)<br />

1 25 32 24 NS*<br />

2 45 37 43<br />

3<br />

* Chi square.<br />

30 31 33<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


536s Lymphoma and Plasma Cell Disorders<br />

8106 General Poster Session (Board #39G), Mon, 1:15 PM-5:15 PM<br />

<strong>Part</strong>icipation <strong>of</strong> BTK in MYD88 signaling in malignant cells expressing the<br />

L265P mutation in Waldenstrom’s macroglobulinemia, and effect on<br />

tumor cells with BTK-inhibitor PCI-32765 in combination with MYD88<br />

pathway inhibitors. Presenting Author: Guang Yang, Dana-Farber Cancer<br />

Institute, Boston, MA<br />

Background: Bruton’s tyrosine kinase (BTK) promotes B-cell receptor<br />

signaling along with B-cell expansion and survival through NF-�B and<br />

MAPK. MYD88 L265P is a widely expressed somatic mutation in tumor<br />

cells from WM patients. MYD88 L265P promotes enhanced tumor cell<br />

survival through IRAK 1/4 mediated NF-�B and MAPK signaling. We<br />

therefore sought to clarify the role <strong>of</strong> BTK signaling in MYD88 L265P<br />

expressing WM cells, and the impact <strong>of</strong> BTK and MYD88/IRAK inhibition<br />

on WM cell signaling and survival. Methods: Western blot analysis was<br />

performed using total and phospho-specific BTK antibodies in MYD88<br />

L265P expressing primary WM patient cells, BCWM.1 and MCWL-1 WM<br />

cell lines following MYD88 knockdown by lentiviral transduction, and/or<br />

use <strong>of</strong> MYD88 or IRAK signal inhibitors. Cells were also treated with the<br />

BTK inhibitor PCI-32765, in the presence or absence <strong>of</strong> MYD88 homodimerization<br />

or IRAK1/4 inhibitors. Annexin V/PIstaining was used to<br />

assess cell survival, and synergism assessed with CalcuSyn s<strong>of</strong>tware.<br />

Results: BTK was phosphorylated in MYD88 L265P expressing WM cells.<br />

Knockdown <strong>of</strong> MYD88 by lentiviral transduction, and/or use <strong>of</strong> MYD88 or<br />

IRAK 1/4 kinase inhibitors led to decreased BTK phosphorylation. Phosphorylation<br />

<strong>of</strong> BTK, TIRAP, a major TLR adapter protein for MYD88 signaling,<br />

IRAK1, IKB�, ERK1/2 and STAT3 were significantly reduced following<br />

treatment with PCI-32765. Treatment with PCI-32765 also induced<br />

apoptosis <strong>of</strong> MYD88 L265P expressing WM cells, and showed synergistic<br />

tumor cell killing in the presence <strong>of</strong> either MYD88 homodimerization or<br />

IRAK 1/4 kinase inhibitors. Conclusions: BTK activation is facilitated by<br />

MYD88 pathway signaling in L265P expressing WM cells, and participates<br />

in MYD88 downstream signaling. Inhibition <strong>of</strong> BTK by PCI-327625 led to<br />

robust tumor cell killing <strong>of</strong> MYD88 L265P expressing WM,cells, which was<br />

potentiated by MYD88 pathway inhibitors. These studies provide the<br />

framework for the investigation <strong>of</strong> BTK inhibitors in WM, as single agents<br />

and in combination with MYD88 pathway inhibitors.<br />

8108 General Poster Session (Board #40A), Mon, 1:15 PM-5:15 PM<br />

miRNA expression pr<strong>of</strong>iling <strong>of</strong> CD20� plasma cell myeloma (PCM):<br />

Upregulation <strong>of</strong> miR-155 shedding new insight into disease biology and<br />

clinicopathologic behavior. Presenting Author: Ravindra Kolhe, Department<br />

<strong>of</strong> Pathology, Georgia Health Sciences University, Augusta, GA<br />

Background: Up to15-20% <strong>of</strong> patients with PCM have expression <strong>of</strong> CD20,<br />

although the significance <strong>of</strong> this remains unclear. The prognostic significance<br />

<strong>of</strong> CD20 expression in PCM is unclear. Recently, a class <strong>of</strong><br />

noncoding RNAs, miRNAs, were identified as critical gene regulators in cell<br />

growth, disease, and development. Our study investigates importance <strong>of</strong><br />

miRNAs in cases <strong>of</strong> CD20� PCM and correlates them with clinicopathological<br />

parameters. Methods: The miRNA expression pr<strong>of</strong>ile <strong>of</strong> CD20� PCM<br />

(n�6), diffuse large B cell lymphoma (DLBCL) (n�6), and CD20 negative<br />

PCM (n�8) were evaluated using the Affeymertrix miRNA microarray<br />

platform on GeneChip miRNA 2.0 array in paraffin-embedded samples.<br />

After hybridization and data acquisition, we used <strong>Part</strong>ek Genomics Suite<br />

s<strong>of</strong>tware for RMA normalization and to determine statistically significant<br />

differences in miRNA expression between experimental groups by ANOVA<br />

and pairwise comparisons (two-sided ��0.05). Results: miRNA expression<br />

pr<strong>of</strong>iles <strong>of</strong> CD20� PCM, show upto �4 times upregulation <strong>of</strong> 7 miRNAs<br />

and downregulation <strong>of</strong> 8 miRNAs. miR-155, the miRNA upregulated in<br />

various B cell lymphomas and plays a key role in the lymphomagenesis, was<br />

amongst the highest miRNAs that were upregulated. Conclusions: miR-155<br />

is known to repress SH2-domain containing inositol-5-phosphatase-1<br />

(SHIP-1), which is a critical phosphatase that negatively down modulates<br />

AKT pathway and has functions during normal B-cell development.<br />

Physiologically, miR-155 is upregulated during B-cell activation upon<br />

antigen stimulation and so plays a role in antibody class switching and<br />

plasma cell formation. We propose that this overexpression <strong>of</strong> miR-155 in<br />

CD20� PCM unblocks AKT activity, inducing cell proliferation and may<br />

explain some <strong>of</strong> the immunophenotypic behavior <strong>of</strong> CD20� PCM. This work<br />

is intriguing for the new information it provides about the role <strong>of</strong> miR-155<br />

in CD20� PCM. Furthermore, the phenotype <strong>of</strong> miR-155�ve CD20� PCM<br />

might ultimately provide new insights into regulation <strong>of</strong> poorly understood<br />

steps in B cell differentiation and maturation into plasma cell and more<br />

importantly the clinicopathological behavior <strong>of</strong> this entity.<br />

8107 General Poster Session (Board #39H), Mon, 1:15 PM-5:15 PM<br />

Use <strong>of</strong> whole genome sequencing to identify highly recurrent somatic<br />

mutations in Waldenström’s macroglobulinemia. Presenting Author: Zachary<br />

R Hunter, Dana-Farber Cancer Institute, Boston, MA<br />

Background: Waldenstrom’s Macroglobulinemia (WM) is an IgM secreting<br />

lymphoplasmacytic lymphoma. The genetic basis for this disease remains<br />

to be clarified. Methods: We performed whole genome sequencing (WGS)<br />

using CD19 � selected bone marrow lymphoplasmacytic cells (LPC) from<br />

30 WM patients. For 10 <strong>of</strong> these patients, paired CD19 � depleted<br />

peripheral blood samples were used for WGS as normal controls. Results:<br />

The most common somatic variants identified and validated by Sanger<br />

sequencing included MYD88 L265P, an activating mutation for IRAK/<br />

TRAF6/NFKB and MAPK signaling, which was observed in 27/30 (90%)<br />

patients; the N-terminal domain <strong>of</strong> CXCR4, which included mutations<br />

associated with WHIM syndrome and confer constitutive CXCR4 signaling<br />

resulting from dysfunctional receptor endocytosis, a finding observed in<br />

8/30 (27%) patients, and ARID1A (5/30; 17%), a tumor suppressor gene.<br />

Less common somatic variants were also identified in MUC16 (4/30; 13%),<br />

TRAF2 (3/30; 10%), TRRAP (3/30; 10%) and MYBBP1A (2/30; 7%).<br />

Conclusions: Using WGS and confirmatory Sanger sequencing, we have<br />

identified several somatic variants with oncogenic function, the most<br />

common <strong>of</strong> which include MYD88 L265P, the N-terminal domain <strong>of</strong><br />

CXCR4, and ARID1A.<br />

TPS8109 General Poster Session (Board #40B), Mon, 1:15 PM-5:15 PM<br />

Phase I/II study <strong>of</strong> investigational agent MLN8237 (alisertib) plus rituximab<br />

with or without vincristine in patients (pts) with relapsed/refractory<br />

(rel/ref) aggressive diffuse large B-cell lymphoma (DLBCL)/transformed<br />

follicular lymphoma (TFL). Presenting Author: Daniel Oscar Persky, University<br />

<strong>of</strong> Arizona Cancer Center, Tucson, AZ<br />

Background: DLBCL and TFL are aggressive subtypes <strong>of</strong> non-Hodgkin<br />

lymphoma (NHL), with poor prognosis in the rel/ref setting. Alternatives to<br />

standard rituximab-based treatments are needed. MLN8237 is an oral,<br />

selective inhibitor <strong>of</strong> Aurora A kinase – a key mitotic regulator overexpressed/<br />

amplified in various human cancers, including lymphomas (Hamada et al,<br />

2003; Yakushijin et al, 2004; Qi et al, 2011). Emerging data from a phase<br />

2 study suggest that MLN8237 has single agent activity in aggressive NHL<br />

(Friedberg et al, ASH 2011). MLN8237 � rituximab (MR) � vincristine<br />

(MRV) has also shown activity in preclinical B-cell NHL models (Mahadevan<br />

et al, ASH 2011). Further clinical evaluation <strong>of</strong> these regimens in<br />

rel/ref B-cell NHL is warranted. Methods: This single-arm phase 1/2 study<br />

(<strong>Clinical</strong>Trials.gov #NCT01397825) aims to assess the safety, efficacy,<br />

and pharmacokinetics <strong>of</strong>, and determine a recommended phase 2 dose<br />

(RP2D) and schedule for, MR and MRV in adults with CD20� rel/ref<br />

DLBCL/TFL after 1–4 prior regimens (including ASCT) and ECOG PS 0–2.<br />

Pts with other aggressive B-cell lymphomas may also enroll in phase 1.<br />

~100 pts will be recruited at 22 sites in the US, UK, Italy, and Spain, and<br />

study duration will be ~2 years. In part 1, a safety lead-in cohort will receive<br />

MR (table), with the RP2D determined as when �2 dose limiting toxicities<br />

(DLT) occur in 6 pts in cycle 1. MRV dose escalation (part 2) will then<br />

follow a 3�3 design. Phase 2 enrollment (part 3) will follow a Simon<br />

optimal 2-stage design, with pts receiving MRV at the RP2D determined in<br />

part 2. The primary phase 2 objective is overall response rate by IWG<br />

criteria. Responders may continue MLN8237 if clinical benefit is seen and<br />

if the regimen is tolerable. Enrollment as <strong>of</strong> 20 Jan 2012 is 6 evaluable pts.<br />

Starting doses: repeating 21-day cycles<br />

MLN8237 Rituximab Vincristine<br />

Phase 1 (part 1) 50 mg ECT BID, days 1–7* 375 mg/m 2 IV, day 1 –<br />

(part 2) ~50% <strong>of</strong> MR RP2D, days 1–7 †<br />

as above 1.4 mg/m 2 IV, days 1, 8 †<br />

Phase 2 (part 3) RP2D from part 2<br />

ECT, enteric coated tablet; BID, twice daily; *dose reduction permitted; † dose/schedule<br />

adjustments based on cycle 1 DLT.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


TPS8110 General Poster Session (Board #40C), Mon, 1:15 PM-5:15 PM<br />

Phase III study <strong>of</strong> investigational MLN8237 (alisertib) versus investigator’s<br />

choice in patients (pts) with relapsed/refractory (rel/ref) peripheral T-cell<br />

lymphoma (PTCL). Presenting Author: Owen O’Connor, Center for Lymphoid<br />

Malignancies, Columbia University Medical Center, New York, NY<br />

Background: PTCL is a rare form <strong>of</strong> aggressive non-Hodgkin lymphoma<br />

(NHL), accounting for 5–20% <strong>of</strong> NHL diagnoses. Standard NHL therapies<br />

developed primarily for B-cell lymphomas are not optimal for PTCL and<br />

early relapse is common. Current treatments for rel/ref PTCL include<br />

pralatrexate, romidepsin, and gemcitabine; however, outcomes remain<br />

poor. The oral, investigational drug, MLN8237, is a selective inhibitor <strong>of</strong><br />

Aurora A kinase (AAK) – a key mitotic regulator that is overexpressed or<br />

amplified in various human tumors. Emerging clinical data from a phase II<br />

study <strong>of</strong> single agent MLN8237 in rel/ref aggressive T-cell lymphoma<br />

(Friedberg et al, ASH 2011) support further clinical evaluation in this<br />

indication. Methods: In this open-label, randomized, phase III study, a<br />

maximum <strong>of</strong> 354 adults with rel/ref PTCL after �1 prior systemic cytotoxic<br />

therapies will be enrolled at approximately 140 centers worldwide. Pts will<br />

be randomized 1:1 to MLN8237 50 mg twice daily as an enteric coated<br />

tablet on days 1–7 <strong>of</strong> 21-day cycles, or to investigator’s choice <strong>of</strong>:<br />

pralatrexate 30 mg/m2 IV once weekly for 6 weeks in 7-week cycles;<br />

romidepsin 14 mg/m2 IV on days 1, 8, and 15 <strong>of</strong> 28-day cycles; or<br />

gemcitabine 1000 mg/m2 IV on days 1, 8, and 15 <strong>of</strong> 28-day cycles. Pts<br />

with disease response/stabilization will be able to continue treatment<br />

provided that clinical benefit is demonstrated and treatment is tolerable.<br />

The expected study duration is 44 months. Primary endpoints are overall<br />

response rate (complete response [CR] � partial response) and progression<br />

free survival by International Working Group criteria (Cheson et al, 2007).<br />

Secondary endpoints include CR rate, overall survival, time to progression,<br />

time to response, duration <strong>of</strong> response, safety, and quality <strong>of</strong> life.<br />

Exploratory endpoints include an evaluation <strong>of</strong> candidate biomarkers (such<br />

as AAK protein expression levels and gene amplification, and the tumor<br />

proliferative marker Ki-67) in tumor biopsies. This study is registered at<br />

<strong>Clinical</strong>Trials.gov: #NCT01482962.<br />

TPS8112 General Poster Session (Board #40E), Mon, 1:15 PM-5:15 PM<br />

ELOQUENT-2: A phase III, randomized, open-label trial <strong>of</strong> lenalidomide/<br />

dexamethasone (Len/Dex) with or without elotuzumab (Elo) in relapsed or<br />

refractory multiple myeloma (RR MM) (CA204-004). Presenting Author:<br />

Sagar Lonial, Multiple Myeloma Research Consortium, Norwalk, CT/<br />

Winship Cancer Institute <strong>of</strong> Emory University, Atlanta, GA<br />

Background: MM remains incurable and patients (pts) typically relapse or<br />

become refractory to current treatments. Novel regimens are needed to<br />

improve pt outcomes. Elo is a humanized monoclonal IgG1 antibody<br />

targeting the cell surface glycoprotein CS1, which is highly expressed on<br />

�95% <strong>of</strong> MM cells. Len/Dex is approved for treatment <strong>of</strong> relapsed MM and<br />

an objective response rate (ORR) <strong>of</strong> ~60% was reported in phase III trials <strong>of</strong><br />

this combination in RR MM. In a phase II study (N�73) <strong>of</strong> Elo (10 or 20<br />

mg/kg) in combination with Len/Dex in pts with RR MM, the 10 mg/kg<br />

group (n�36) demonstrated an ORR <strong>of</strong> 92% and median progression-free<br />

survival (PFS) that was not reached after a median follow-up <strong>of</strong> 14.1<br />

months. Encouraging activity was seen in patients with high-risk cytogenetics<br />

and/or stage 2-3 disease. Based on these data, a randomized,<br />

open-label phase III trial has been initiated to determine if the addition <strong>of</strong><br />

Elo to Len/Dex will improve PFS in patients with RR MM compared with<br />

Len/Dex alone. Methods: Pts (N�640) with RR MM and 1-3 prior therapies<br />

are eligible, including pts with mild or moderate renal impairment. Pts are<br />

randomized in a 1:1 ratio to receive 28-day cycles <strong>of</strong> Len 25 mg PO (days<br />

1-21) and Dex 40 mg PO (days 1, 8, 15 and 22) with or without Elo. Elo<br />

dose and schedule is 10 mg/kg IV on days 1, 8, 15, 22 in the first 2 cycles<br />

and on days 1 and 15 in subsequent cycles. Dex 8 mg IV � 28 mg PO is<br />

used during the weeks with Elo. Treatment will continue until disease<br />

progression, death, or withdrawal <strong>of</strong> consent. Patients will be followed for<br />

tumor response every 4 weeks until progressive disease and then survival<br />

every 12 weeks. The primary endpoint is PFS (90% power for a hazard ratio<br />

[experimental to control arm] <strong>of</strong> 0.74) and the secondary endpoints are<br />

ORR and overall survival. Exploratory endpoints are safety, time to<br />

response, duration <strong>of</strong> response, time to subsequent therapy, health-related<br />

quality <strong>of</strong> life, and pharmacokinetics and immunogenicity <strong>of</strong> Elo. Potential<br />

biomarkers will also be assessed. As <strong>of</strong> January 10th, 2012, 107 pts were<br />

enrolled and 68 pts were treated. NCT01239797.<br />

Lymphoma and Plasma Cell Disorders<br />

537s<br />

TPS8111 General Poster Session (Board #40D), Mon, 1:15 PM-5:15 PM<br />

Pilot study <strong>of</strong> MAGE-A3 protein vaccination and autologous stem cell<br />

transplantation (autoSCT) as consolidation therapy for multiple myeloma<br />

(MM). Presenting Author: Adam D. Cohen, Fox Chase Cancer Center,<br />

Philadelphia, PA<br />

Background: MAGE-A3 is a cancer-testis antigen (CT Ag) commonly<br />

expressed in MM but not in normal non-gonadal tissues. MAGE-A3<br />

inhibited p53-dependent and independent apoptosis in MM cells (Clin<br />

Cancer Res 2011;17:4309), and spontaneous immunity against CT Ags in<br />

MM patients was associated with favorable clinical outcomes (Blood<br />

2007;109:1103; Blood 2008;112:3362), making it a rational target for<br />

immunotherapy. In this study (NCT01380145), we are examining if a<br />

recombinant (rec) MAGE-A3 protein vaccine � adjuvant combined with<br />

vaccine-primed peripheral blood lymphocytes (PBL) can safely stimulate<br />

antigen-specific immunity in MM patients undergoing autoSCT for consolidation<br />

therapy. Methods: Patients meeting the following criteria are<br />

eligible: within 12 months <strong>of</strong> diagnosis; MAGE-A� MM cells by immunohistochemistry;<br />

achieving at least a very good partial response with induction<br />

therapy; and meeting institutional criteria for autoSCT. One patient <strong>of</strong> a<br />

planned cohort <strong>of</strong> sixteen has been enrolled. Six weeks before SCT,<br />

subjects will receive their first vaccination (300 �g IM) and three weeks<br />

later undergo leukopheresis to collect vaccine-primed PBL. They then<br />

undergo stem cell mobilization followed by a standard melphalanconditioned<br />

autoSCT. On day 3 after stem cell infusion, the unmanipulated,<br />

primed PBL will be re-infused followed by the second recMAGE-A3<br />

vaccination on day 10. An additional six vaccinations will be administered<br />

on days 31, 52, 73, 94, 180, and 270 after autoSCT. The primary<br />

objectives are safety and tolerability. The secondary objectives <strong>of</strong> cellular<br />

and humoral immune responses and lymphocyte reconstitution will be<br />

assessed by a validated series <strong>of</strong> quantitative assays using established<br />

response criteria (PNAS 2008;105:1650). Success criteria based on<br />

immune response have not been specified for this pilot study, though<br />

induction <strong>of</strong> MAGE-A3 immunity in at least 50% <strong>of</strong> patients would merit<br />

consideration for further evaluation <strong>of</strong> clinical efficacy.<br />

TPS8113 General Poster Session (Board #40F), Mon, 1:15 PM-5:15 PM<br />

ELOQUENT-1: A phase III, randomized, open-label trial <strong>of</strong> lenalidomide/<br />

dexamethasone with or without elotuzumab in subjects with previously<br />

untreated multiple myeloma (CA204-006). Presenting Author: Meletios A.<br />

Dimopoulos, Hellenic Cooperative Oncology Group, Athens, Greece<br />

Background: Elotuzumab (Elo) is a humanized monoclonal IgG1 antibody<br />

targeting the cell surface glycoprotein CS1, which is highly expressed on<br />

�95% <strong>of</strong> MM cells. In a MM xenograft mouse model, the combination <strong>of</strong><br />

Elo � lenalidomide (Len) significantly reduced tumor volume in a synergistic<br />

manner compared with either agent alone. In a phase 2 study (N�73) <strong>of</strong><br />

Elo (10 or 20 mg/kg) in combination with Len and low-dose-dexamethasone<br />

(Dex) in pts with RR MM, the 10 mg/kg dose group (n�36)<br />

demonstrated objective response rates (ORR) <strong>of</strong> 92% in all pts, and 100%<br />

in pts who had received only 1 prior therapy (n�16). The higher response<br />

rate in pts with fewer prior lines <strong>of</strong> therapy provides a rationale for<br />

investigating this combination earlier in the disease course. This randomized,<br />

open-label, phase 3 trial will determine if the addition <strong>of</strong> Elo to<br />

Len/Dex improves progression-free survival (PFS) in pts with newly diagnosed,<br />

untreated MM. Methods: Pts (N�750) with newly diagnosed<br />

symptomatic MM ineligible for stem cell transplant will be randomized in a<br />

1:1 ratio to receive 28-day cycles <strong>of</strong> Len 25 mg PO (days1-21) and Dex 40<br />

mg PO (days 1, 8, 15 and 22) with or without Elo. Elo dose and schedule is<br />

10 mg/kg IV on days 1, 8, 15, 22 in the first 2 cycles and on days 1 and 15<br />

<strong>of</strong> cycles 3-18 followed by 20 mg/kg on day 1 <strong>of</strong> cycle 19 onward. Dex 8 mg<br />

IV � 28 mg PO is used during the weeks with Elo. Treatment will continue<br />

until disease progression, death, or withdrawal <strong>of</strong> consent. Pts will be<br />

followed up for response every 4 weeks until progressive disease and for<br />

survival every 16 weeks. The primary endpoint is PFS (90% power for a<br />

hazard ratio [experimental to control arm] <strong>of</strong> 0.74) and the secondary<br />

endpoints are ORR and overall survival. Exploratory endpoints are safety,<br />

time to response, duration <strong>of</strong> response, time to subsequent therapy,<br />

health-related quality <strong>of</strong> life, and pharmacokinetics and immunogenicity <strong>of</strong><br />

Elo. Potential biomarkers will also be assessed. As <strong>of</strong> January 1, 2012, 13<br />

pts were enrolled and 9 pts were treated. NCT01335399.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


538s Lymphoma and Plasma Cell Disorders<br />

TPS8114 General Poster Session (Board #40G), Mon, 1:15 PM-5:15 PM<br />

A phase II randomized study <strong>of</strong> bortezomib/dexamethasone (Bort/Dex) with<br />

or without elotuzumab (Elo) in patients (pts) with relapsed/refractory<br />

multiple myeloma (RR MM) (CA204-009). Presenting Author: Andrzej J.<br />

Jakubowiak, University <strong>of</strong> Chicago Medical Center, Chicago, IL<br />

Background: MM is rarely curable and pts typically relapse or become<br />

refractory to current treatments. Elo is a humanized monoclonal IgG1<br />

antibody targeting the cell surface glycoprotein CS1, which is highly<br />

expressed on �95% <strong>of</strong> MM cells with little to no expression on normal<br />

tissues. The mechanism <strong>of</strong> action <strong>of</strong> Elo is primarily natural killer (NK)<br />

cell-mediated antibody-dependent cellular cytotoxicity (ADCC) against<br />

myeloma cells. Elo � Bort significantly enhanced antimyeloma activity in a<br />

mouse xenograft model vs either agent alone. The addition <strong>of</strong> Bort<br />

enhanced the ADCC activity <strong>of</strong> Elo in preclinical studies. In a phase I study<br />

<strong>of</strong> Elo � Bort in pts with RR MM, objective response rate (ORR) was 48%,<br />

median progression-free survival (PFS) was 9.5 months, and activity was<br />

observed in 2/3 patients (67%) refractory to Bort (Jakubowiak et al. J Clin<br />

Oncol, in press). This study will assess if the addition <strong>of</strong> Elo to Bort/Dex<br />

improves PFS and, if so, whether magnitude <strong>of</strong> the improvement is linked<br />

to Fc�RIIIa polymorphism. Methods: Pts (N�150) with RR MM after 1 or 2<br />

prior therapies will be randomized in a 1:1 ratio to receive Bort 1.3 mg/m2 IV or SQ (Cycles 1-8: days 1, 4, 8, and 11; Cycles �9: days 1, 8, and 15)<br />

and Dex with or without Elo. Elo dose and schedule is 10 mg/kg IV (Cycles<br />

1-2: days 1, 8, and 15 [21-day cycles]; Cycles 3-8: days 1 and 11 [21-day<br />

cycles]; Cycles �9: days 1 and 15 [28-day cycles]). In the arm without Elo,<br />

Dex 20 mg PO is scheduled for Cycles 1-8: days 1, 2, 4, 5, 8, 9, 11, and<br />

12; and Cycles �9: days 1, 2, 8, 9, 15, 16. In the arm with Elo, Dex 20 mg<br />

PO is scheduled for Cycles 1-2: days 2, 4, 5, 9, and 11; Cycles 3-8: days 2,<br />

4, 5, 8, 9, 12; Cycles �9: days 2, 8, 9, and 16) on weeks without Elo, and<br />

on weeks with Elo, Dex 8 mg PO and 8 mg IV is scheduled on the same day<br />

as Elo. Treatment will continue until disease progression, unacceptable<br />

toxicity, or withdrawal <strong>of</strong> consent. Patients refractory or intolerant to Bort<br />

will be excluded. Efficacy will be assessed on day 1 <strong>of</strong> each cycle by IMWG<br />

criteria. The primary endpoint is PFS. Secondary endpoints include ORR<br />

and PFS/ORR in pts with �1Fc�RIIIa V allele. As <strong>of</strong> February 1, 2012, 1 pt<br />

was enrolled. NCT01478048.<br />

TPS8116 General Poster Session (Board #41A), Mon, 1:15 PM-5:15 PM<br />

Phase I/II open-label, multiple-dose, dose-escalation study to evaluate the<br />

safety and tolerability <strong>of</strong> SNS01T administered by intravenous infusion in<br />

patients with relapsed or refractory multiple myeloma. Presenting Author:<br />

John Anthony Lust, Mayo Clinic, Rochester, MN<br />

Background: Eukaryotic translation initiation Factor 5A (eIF5A) has been<br />

implicated in the regulation <strong>of</strong> apoptosis and is the only known protein to be<br />

modified by hypusination. Hypusinated eIF5A is the predominant form <strong>of</strong><br />

eIF5A in cancer cells. However, in its unhypusinated form, eIF5A is<br />

pro-apoptotic. SNS01-T, designed to treat myeloma, consists <strong>of</strong> two<br />

components: a plasmid DNA expressing eIF5AK50R (human eIF5A containing<br />

a lysine to arginine substitution at position 50) which remains<br />

pro-apoptotic because it cannot be hypusinated, and an siRNA against an<br />

untranslated region <strong>of</strong> native eIF5A mRNA. When these two components<br />

are combined with linear polyethyleneimine (PEI), the nucleic acids are<br />

condensed into nanoparticles for protection from degradation in the blood<br />

and enhanced delivery to tissues. The eIF5AK50R transgene is under the<br />

control <strong>of</strong> the B29 promoter and enhancer, which restricts expression to B<br />

cells. The mode <strong>of</strong> action <strong>of</strong> SNS01-T is to use an eIF5A-specific siRNA to<br />

deplete the pool <strong>of</strong> hypusinated eIF5A in myeloma cells while simultaneously<br />

adding pro-apoptotic eIF5AK50R . In vitro cell studies and in vivo<br />

xenograft studies have demonstrated the efficacy <strong>of</strong> this approach. Methods:<br />

Eligible patients are enrolled sequentially into four cohorts <strong>of</strong> increasingly<br />

higher doses. Each cohort will receive SNS01-T by intravenous infusion<br />

twice weekly for 6 consecutive weeks and then be observed every 4 weeks<br />

during a 24-week follow-up period. Eligible patients must have been<br />

diagnosed with multiple myeloma according to IMWG criteria, have<br />

measurable disease, have relapsed disease after two or more prior treatment<br />

regimens, have a life expectancy <strong>of</strong> at least 3 months, and not be<br />

eligible to receive any other standard therapy known to extend life<br />

expectancy. The primary objective is to evaluate the safety and tolerability<br />

<strong>of</strong> multiple escalating doses <strong>of</strong> SNS01-T. Secondary objectives include<br />

pharmacokinetics, immunogenicity studies, proinflammatory cytokine quantitation,<br />

and therapeutic efficacy. Two <strong>of</strong> the planned 15 patients have<br />

been enrolled. (<strong>Clinical</strong> Trials.gov Identifier: NCT01435720.)<br />

TPS8115 General Poster Session (Board #40H), Mon, 1:15 PM-5:15 PM<br />

A single-arm, open-label, multicenter phase I/II study <strong>of</strong> the combination <strong>of</strong><br />

panobinostat (pan) and carfilzomib (cfz) in patients (pts) with relapsed/<br />

refractory multiple myeloma (RR MM). Presenting Author: Jesus G.<br />

Berdeja, Tennessee Oncology, PLLC/Sarah Cannon Research Institute,<br />

Nashville, TN<br />

Background: Despite novel therapies, MM remains an incurable disease.<br />

Changes in histone modification are commonly found in human cancers<br />

including MM. Preclinical studies demonstrate synergistic anti-MM activity<br />

with histone deacetylase inhibitors (HDACi) and proteasome inhibitors (PI)<br />

through the dual inhibition <strong>of</strong> the proteasome and aggresome pathways.<br />

Pan is an oral pan-HDACi which has shown synergy with bortezomib in<br />

clinical studies. Cfz is a 2nd generation PI which has shown marked<br />

anti-MM activity with an improved safety pr<strong>of</strong>ile. In this trial we evaluate<br />

the safety and efficacy <strong>of</strong> the combination <strong>of</strong> pan and cfz in pts with RR<br />

MM. Methods: This multi-center US study plans to enroll up to 52 adults<br />

with RR MM. The phase I study will determine the MTD <strong>of</strong> the combination<br />

<strong>of</strong> cfz and pan and follow a standard dose escalation design. Pan will be<br />

administered orally three times weekly during weeks 1 and 3 <strong>of</strong> each<br />

28-day cycle (Days 1, 3, 5, 15, 17, 19) at a starting dose <strong>of</strong> 20 mg. Cfz will<br />

be administered intravenously on Days 1, 2, 8, 9, 15, and 16 <strong>of</strong> each<br />

28-day cycle. Cfz starting dose will be 20mg on days 1,2 <strong>of</strong> cycle 1 with<br />

escalation to the corresponding dose level beginning at 27 mg , if well<br />

tolerated. Dose modifications will not be permitted during cycle 1 unless a<br />

pt experiences a DLT. A maximum <strong>of</strong> four dose levels will be evaluated.<br />

Approximately 24 pts will be enrolled during the phase I portion to<br />

establish the MTD. In the phase II portion <strong>of</strong> this study, pts with RR MM will<br />

receive treatment with the optimal dose <strong>of</strong> pan and cfz established during<br />

phase I. Pts will be assessed for response to treatment after each cycle (4<br />

weeks). Pts with objective response or stable disease will continue<br />

treatment until disease progression or unacceptable toxicity occurs. The<br />

primary endpoint is to establish the optimal doses <strong>of</strong> cfz and pan that can<br />

be administered to pts with RR MM (phase I) and to evaluate the overall<br />

response rate (phase II). Secondary endpoints include time-to-progression,<br />

progression-free survival, overall survival and safety. Approximately 25 pts<br />

are planned for the phase II portion. Current status: As <strong>of</strong> 1/27/12 3<br />

patients have been enrolled.<br />

TPS8117 General Poster Session (Board #41B), Mon, 1:15 PM-5:15 PM<br />

Recruitment in a randomized, double-blind, placebo-controlled study to<br />

assess siltuximab (CNTO 328), an anti-IL-6 monoclonal antibody, in<br />

patients with multicentric Castleman’s disease. Presenting Author: Frits<br />

Van Rhee, University <strong>of</strong> Arkansas for Medical Sciences, Little Rock, AR<br />

Background: Multicentric Castleman’s disease (MCD) is a rare lymphoproliferative<br />

disorder in which dysregulated production <strong>of</strong> interleukin (IL)-6<br />

results in lymph node enlargement and debilitating symptoms, including<br />

systemic inflammatory manifestations (eg, fever, fatigue, weight loss),<br />

autoimmune phenomena, and markedly abnormal laboratory findings (eg,<br />

anemia, hyper-�-globulinemia, hypoalbuminemia, thrombocytosis, increases<br />

in acute-phase proteins such as CRP, ESR, fibrinogen). There is<br />

currently no approved systemic treatment for MCD in the US or EU.<br />

Siltuximab (CNTO 328) is a chimeric mAb with high affinity for soluble<br />

IL-6. In a previous phase 1 study, high objective tumor response rate (64%)<br />

has been observed in patients with MCD (van Rhee F et al. Blood<br />

2008;112:1008), and the long-term safety pr<strong>of</strong>ile looks favourable (Kurzrock<br />

R et al. Blood 2011;118:3959). These results have prompted a<br />

randomized, double-blind, placebo-controlled study to definitively assess<br />

the efficacy and safety <strong>of</strong> siltuximab in combination with best supportive<br />

care (BSC) compared with BSC in patients with MCD. Methods: Eligibility<br />

includes HIV- and HHV-8-negative, symptomatic, measurable MCD patients.<br />

Patients with prior lymphoma or prior exposure to treatment<br />

targeting IL-6 or its receptor are excluded. Patients can be enrolled when<br />

receiving stable doses <strong>of</strong> corticosteroids (�1 mg/kg/d <strong>of</strong> prednisone or<br />

equivalent). Patients will be randomized in a 1:2 ratio to placebo � BSC or<br />

to 11 mg/kg siltuximab � BSC and will receive siltuximab/placebo in a<br />

1-hour IV infusion every 3 weeks. The primary objective is to evaluate<br />

durable tumor and symptomatic response in the intent-to-treat population.<br />

Secondary objectives include additional efficacy measures, safety, patient<br />

reported outcomes, and pharmacologic assessment. Status: 67/78 patients<br />

have been randomly assigned. This is the first randomized, placebocontrolled<br />

study to be conducted in MCD. The rarity <strong>of</strong> the disease has<br />

posed unique challenges, and various enrollment strategies have been<br />

successfully implemented, with projected completion <strong>of</strong> enrollment in<br />

March 2012.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


TPS8118 General Poster Session (Board #41C), Mon, 1:15 PM-5:15 PM<br />

PILLAR-2: A randomized, double-blind, placebo-controlled, phase III<br />

study <strong>of</strong> adjuvant everolimus in poor-risk diffuse large B-cell lymphoma<br />

(DLBCL). Presenting Author: Tongyu Lin, Department <strong>of</strong> Medical Oncology,<br />

Sun Yat-sen University Cancer Center, Guangzhou, China<br />

Background: Effective DLBCL adjuvant therapy after first-line chemotherapy<br />

is needed as high-risk DLBCL is associated with a poor 4-year<br />

overall survival (OS) rate (Sehn et al, Blood 2007;109:1857-61). In a<br />

phase II study <strong>of</strong> the oral mammalian target <strong>of</strong> rapamycin inhibitor<br />

everolimus in relapsed, aggressive non-Hodgkin lymphoma, DLBCL patients<br />

(n�47) had a 30% overall response rate (Witzig et al, Leukemia<br />

2011;25:341-7). Methods: PILLAR-2 is an ongoing international, randomized,<br />

double-blind, phase 3 study designed to compare everolimus efficacy<br />

and safety with that <strong>of</strong> placebo in poor-risk DLBCL patients who achieved<br />

complete response (CR) with first-line rituximab-based chemotherapy<br />

(R-chemo) (<strong>Clinical</strong>Trials.gov: NCT00790036; sponsor: Novartis Pharmaceuticals).<br />

Eligibility criteria include age �18 years; confirmed stage II<br />

bulky, III, or IV DLBCL and International Prognostic Index 3-5 at diagnosis;<br />

confirmed CR per revised International Workshop Response Criteria for<br />

malignant lymphoma (Cheson et al, J Clin Oncol 2007;25:579-86) after<br />

first-line R-chemo regimen completed 6-14 weeks before study drug start;<br />

Eastern Cooperative Oncology Group performance status �2; no ongoing or<br />

post–R-chemo radiation; and no myelosuppressive chemotherapy or biologic<br />

therapy within 3 weeks. Patients are randomized 1:1 to everolimus 10<br />

mg once daily or matching placebo and treated for 12 months or until<br />

disease relapse, unacceptable toxicity, or death. Radiologic tumor assessment<br />

is performed at baseline, every 12 weeks during years 1 and 2, every<br />

24 weeks during years 3 and 4, and annually thereafter until start <strong>of</strong> new<br />

anticancer therapy or 5 years after last patient randomization. The primary<br />

endpoint is disease-free survival (DFS). Secondary endpoints are OS,<br />

lymphoma-specific survival, and safety. Expected enrollment is 687<br />

patients. Final analysis will be performed when 279 DFS events occur;<br />

survival follow-up will continue until 338 deaths occur and the last<br />

randomized patient has been followed for �5 years. Currently, 422<br />

patients are enrolled. The data monitoring committee last reviewed the trial<br />

in July 2011 and recommended that it continue as planned.<br />

Lymphoma and Plasma Cell Disorders<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.<br />

539s


540s Melanoma/Skin Cancers<br />

LBA8500^ Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Phase III, randomized, open-label, multicenter trial (BREAK-3) comparing<br />

the BRAF kinase inhibitor dabrafenib (GSK2118436) with dacarbazine<br />

(DTIC) in patients with BRAFV600E-mutated melanoma. Presenting Author:<br />

Axel Hauschild, Universitaetsklinikum Schleswig-Holstein, Kiel Schleswig-<br />

Holstein, Germany<br />

The full, final text <strong>of</strong> this abstract will be available at<br />

abstract.asco.org at 12:01 AM (EDT) on Monday, June 4,<br />

2012, and in the <strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong> online<br />

supplement to the June 20, 2012, issue <strong>of</strong> Journal <strong>of</strong><br />

<strong>Clinical</strong> Oncology. Onsite at the <strong>Meeting</strong>, this abstract will<br />

be printed in the Monday edition <strong>of</strong> ASCO Daily News.<br />

8502^ Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Updated overall survival (OS) results for BRIM-3, a phase III randomized,<br />

open-label, multicenter trial comparing BRAF inhibitor vemurafenib (vem)<br />

with dacarbazine (DTIC) in previously untreated patients with BRAFV600E -<br />

mutated melanoma. Presenting Author: Paul B. Chapman, Memorial<br />

Sloan-Kettering Cancer Center, New York, NY<br />

Background: We previously reported results <strong>of</strong> the planned OS interim<br />

analysis for BRIM-3 (50% <strong>of</strong> the planned 196 deaths required for final<br />

analysis) at which time the independent Data Safety Monitoring Board<br />

recommended release <strong>of</strong> results due to compelling efficacy (hazard ratio<br />

[HR] for death, 0.37 [95% CI 0.26–0.55]); p�0.0001 and PFS HR 0.26<br />

[95% CI 0.20–0.33]; p�0.0001) and that DTIC-treated patients be<br />

permitted to cross over to receive vem. Median follow-up for vem patients<br />

was 3.75 months, and longer follow-up would estimate median OS more<br />

reliably. Updated OS with median 6.2 months follow-up and 199 total<br />

deaths showed HR for death 0.44 (95% CI 0.33–0.59) favoring vem and<br />

median OS for vem not reached. We report here the results <strong>of</strong> an updated<br />

OS analysis performed in Nov 2011 with ~10 months median follow-up on<br />

vem. Methods: 675 patients with previously untreated, unresectable Stage<br />

IIIC or IV melanoma that tested positive for BRAFV600E mutation by the<br />

cobas 4800 BRAF V600 Mutation Test were randomized (1:1) from Jan to<br />

Dec 2010 to vem (960 mg po bid) or DTIC (1000 mg/m2 IV q3w).<br />

Co-primary endpoints were OS and PFS. OS data for DTIC patients who<br />

crossed over to vem were censored at the time <strong>of</strong> crossover. Results: Median<br />

lengths <strong>of</strong> follow-up on vem and DTIC were 10.5 months (range 0.4–18.1)<br />

and 8.4 months (range �0.1–18.3), respectively. There were 334 deaths.<br />

Median OS rates with vem and DTIC were 13.2 months (95% CI<br />

12.0–15.0) and 9.6 months (95% CI 7.9–11.8), respectively. 12-month<br />

OS rates were 55% for vem and 43% for DTIC. HR for death was 0.62 (95%<br />

CI 0.49–0.77) in favor <strong>of</strong> vem. 81 DTIC patients crossed over to vem. 44<br />

(13%) vem and 65 (19%) DTIC patients received ipilimumab postprogression.<br />

Conclusions: With longer follow-up, vem treatment continues<br />

to be associated with improved OS in the BRIM-3 study. An updated<br />

analysis, with estimated median follow-up <strong>of</strong> ~13 months and including<br />

response data, will be conducted in Apr 2012 and presented at the<br />

meeting.<br />

8501 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

BREAK-MB: A phase II study assessing overall intracranial response rate<br />

(OIRR) to dabrafenib (GSK2118436) in patients (pts) with BRAF V600E/k<br />

mutation-positive melanoma with brain metastases (mets). Presenting<br />

Author: John M. Kirkwood, University <strong>of</strong> Pittsburgh Cancer Institute,<br />

Pittsburgh, PA<br />

Background: Melanoma brain mets carry a poor prognosis (median survival<br />

�4 months), for which more effective therapies are needed. Dabrafenib is a<br />

potent, selective oral inhibitor <strong>of</strong> mutated BRAF that has demonstrated<br />

clinical efficacy in pts with BRAF V600E/K mutant melanoma and<br />

previously untreated brain mets in a phase I study. Therefore, a phase II<br />

study (BREAK-MB) was initiated. Methods: Stage IV pts with � 1 intracranial<br />

met (0.5 cm–4 cm assessed by MRI) without prior brain therapy<br />

(Cohort A) or with progression following prior brain therapy (Cohort B) were<br />

eligible with V600E/K mutation. Pts received dabrafenib 150 mg BID. The<br />

primary endpoint was investigator-assessed OIRR among V600E mutationpositive<br />

pts. Results: Overall, 172 pts were recruited. At interim analysis, <strong>of</strong><br />

127 pts enrolled (safety population), 41 (Cohort A n�24; Cohort B n�17)<br />

had reached 8-week disease assessment. Unconfirmed OIRR in Cohort A<br />

was 10/19, (53%; 95% CI: 28.9–75.6%) for V600E and 1/5 (20%; 95%<br />

CI: 0.5–71.6%) for V600K pts. In Cohort B unconfirmed OIRR was 8/15<br />

(53%; 95% CI: 26.6%–78.7%) for V600E and 1/2 (50%; 95% CI:<br />

1.3–98.7%) for V600K pts. Unconfirmed overall response rate (ORR) in<br />

Cohort A was 10/19 (53%; 95% CI: 28.9–75.6%) for V600E and 1/5<br />

(20%; 95% CI: 0.5–71.6%) for V600K pts. In Cohort B unconfirmed ORR<br />

was 6/15 (40%; 95% CI: 16.3–67.7%) for V600E and 1/2 (50%; 95% CI:<br />

1.3–98.7%) for V600K pts. In the safety population, 12/63 pts (19%) in<br />

Cohort A reported an SAE; 1 (2%) was fatal (cerebral hemorrhage). In<br />

Cohort B 15/64 pts (25%) reported an SAE; 1 (2%) was fatal (seizure). In<br />

Cohort A, 47 pts (75%) experienced AEs: 9 grade 3 (14%) and 3 grade 4<br />

(5%). The most common AEs were headache (21%), hyperkeratosis and<br />

rash (17% each). In Cohort B, 47 pts (73%) experienced AEs: 12 grade 3<br />

(19%) and 3 grade 4 (5%). The most common AEs were fatigue or nausea<br />

(22% each) and pyrexia (17%). Conclusions: Dabrafenib shows high<br />

clinical activity both in pts with intra- and extracranial mets with acceptable<br />

toxicity. Final data will be presented.<br />

8503^ Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Analysis <strong>of</strong> molecular mechanisms <strong>of</strong> response and resistance to vemurafenib<br />

(vem) in BRAFV600E melanoma. Presenting Author: Jeffrey Alan<br />

Sosman, Vanderbilt-Ingram Cancer Center, Nashville, TN<br />

Background: Vem induces frequent clinical responses (RR �50%) and<br />

improved survival in patients (pts) with BRAF-mutated metastatic melanoma.<br />

Multiple mechanisms <strong>of</strong> escape from vem have been proposed. We<br />

performed a centralized analysis <strong>of</strong> pretreatment, cycle 1, day 15 (D15),<br />

and progression (DP) tumor samples collected during the phase II BRIM-2<br />

trial (Sosman et al, NEJM 2012). Methods: Of 132 pts enrolled, archival<br />

tumor samples, biopsies taken at baseline (BL), D15, and DP were<br />

obtained from 84, 38, 26, and 22 pts, respectively, and analyzed by<br />

immunohistochemistry (IHC) for signaling molecules in the MAPK, PI3K/<br />

AKT, and cell cycle pathways. Genetic analyses <strong>of</strong> signaling genes were<br />

performed using Sequenom MASSarray and direct DNA sequencing.<br />

Results: High levels <strong>of</strong> ERK phosphorylation were seen at BL indicating<br />

constitutive MAPK signaling due to the BRAF mutation. In 19/22 paired<br />

samples, pERK levels were reduced following vem (BL vs D15). Mean<br />

absolute pERK reduction in pts with clinical response (RECIST criteria) to<br />

vem (n�14) was significantly greater than in non-responders (n�8;<br />

p�0.013). Baseline cytoplasmic PTEN H-score was higher in responders<br />

vs non-responders (H-score�88 vs 68; p�0.042). At progression, upregulation<br />

<strong>of</strong> pERK (H-score�181) was frequently, but not uniformly found vs<br />

D15 tumors (H-score�48.5). At progression, increases in Cyclin D1 and<br />

Ki67 were seen, without obvious changes in PTEN or pAKT. NRAS<br />

mutations occurred in 3/13 DP; 2 had paired BL samples without NRAS<br />

mutations. Only 1/82 pts had a concomitant NRAS and BRAF mutation at<br />

BL, and did not respond to vem. MAP2K1 (MEK1) codon 124 mutations<br />

occurred in 7/92 BL and 1/20 DP samples. 2 pts with BL MEK1 mutations<br />

had partial responses. Conclusions: MAPK signaling was effectively inhibited<br />

by vem early in treatment, and in the subset <strong>of</strong> patients with matched<br />

tumor samples the degree <strong>of</strong> pathway inhibition correlated with clinical<br />

response. MAPK signaling is upregulated in many lesions at progression.<br />

NRAS mutations appear to be a mechanism <strong>of</strong> acquired resistance in a few<br />

tumors (3/13), while the role <strong>of</strong> MEK1 mutations in resistance is less clear.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


8504 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Phase III trial <strong>of</strong> high-dose interferon alpha-2b versus cisplatin, vinblastine,<br />

DTIC plus IL-2 and interferon in patients with high-risk melanoma<br />

(SWOG S0008): An intergroup study <strong>of</strong> CALGB, COG, ECOG, and SWOG.<br />

Presenting Author: Lawrence E. Flaherty, Wayne State University, Detroit,<br />

MI<br />

Background: High-dose interferon for one year (HDI) is the FDA approved<br />

adjuvant therapy for patients (pts) with high-risk melanoma (HRM). Efforts<br />

to modify IFN dose or schedule have not improved efficacy. A meta-analysis<br />

demonstrated that biochemotherapy (BCT) produced superior response<br />

rates compared with chemotherapy in pts with stage IV melanoma (Wheatley<br />

et al J Clin Oncol 25:5426, 2007). We sought to determine whether a<br />

short course <strong>of</strong> BCT would be more effective than HDI as adjuvant<br />

treatment in pts with HRM. Methods: S-0008 (an Intergroup Phase III trial)<br />

enrolled pts who were high risk (Stage III A-N2a thru Stage III C N3) and<br />

randomized them to receive either HDI or BCT consisting <strong>of</strong> dacarbazine<br />

800 mg/m2 day 1, cisplatin 20 mg/m2 / days 1-4, vinblastine 1.2 mg/m2 days 1-4, IL-2 9 MIU/m2 /day continuous IV days 1-4, IFN 5 MU/m2 /day sc<br />

days 1-4, 8,10,12, and G-CSF 5 ug/kg/day sc days 7-16. BCT cycles were<br />

given every 21 days x 3 cycles (9 weeks total). Pts were stratified for<br />

number <strong>of</strong> involved nodes (1-3 v �4), micro v macro metastasis, and<br />

ulceration <strong>of</strong> the primary. Co-primary endpoints were relapse free survival<br />

(RFS) and overall survival (OS) using a one-sided log rank test at p� 0.05.<br />

Results: 432 pts were enrolled between 8/2000 and 11/2007: 30 were<br />

ineligible or withdrew consent. Grade 3 and 4 adverse events occurred in<br />

57% and 7% respectively <strong>of</strong> HDI pts and 36% and 40% <strong>of</strong> BCT pts. At a<br />

median f/up <strong>of</strong> 6 yrs, BCT improved RFS (p � 0.02, HR 0.77 [90% CI:<br />

0.62 – 0.96]) with median RFS for BCT <strong>of</strong> 4.0 yrs (90% CI:1.9 – 5.9) v 1.9<br />

yrs (90% CI: 1.4 – 2.5) and 5 yr RFS <strong>of</strong> 47% v 39%. Median OS was not<br />

different between the two arms (p � 0.49 HR 1.0 [90% CI: 0.78 – 1.27])<br />

with median OS not yet reached for BCT v 8.4 yrs (90% CI: 4.5 – 9.3) for<br />

HDI and 5 yr survival 56% for both arms. Conclusions: In HRM pts, BCT<br />

provides a statistically significant improvement in RFS compared to HDI,<br />

but no discernable difference in OS and more grade IV toxicity. BCT<br />

represents a shorter, alternative treatment for pts with HRM, and a<br />

potential control arm and basis for future combinations in the adjuvant<br />

setting.<br />

8506 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Phase II randomized study <strong>of</strong> high-dose interferon alfa-2b (HDI) versus<br />

chemotherapy as adjuvant therapy in patients with resected mucosal<br />

melanoma. Presenting Author: Bin Lian, Peking University Cancer Hospital<br />

and Institute, Beijing, China<br />

Background: High-dose interferon alfa-2b regimen has been demonstrated<br />

as the standard <strong>of</strong> adjuvant therapy for resected melanoma. But the<br />

subgroup <strong>of</strong> mucosal melanoma seems more aggressive and insensitive to<br />

immunotherapy. So we conducted a phase II study (ChiCTR-TRC-<br />

11001798) to compare Interferon regimen with chemotherapy as adjuvant<br />

therapy for this particular subgroup pts. Methods: Resected mucosal<br />

melanoma pts were randomized to observation (Group A) or receive<br />

postoperative adjuvant Interferon (IFN) alfa2b 15 MU/m2 /day for 5<br />

days/week X 4 weeks followed by 9 MU/m2 three times each week for 48<br />

weeks (Group B), or chemotherapy with temozolomide 200 mg/m2 d1-5 �<br />

cisplatin 25 mg/m2 d1-3, repeated every 3 Weeks X 6 cycles (Group C).<br />

Results: 189 patients were enrolled and 184 patients were eligible for<br />

survival analysis. With a median follow-up <strong>of</strong> 26.8 months, the median RFS<br />

for Group A, B and C were 5.4, 9.4 and 20.8 months, respectively<br />

(P�0.001). Estimated median OS for Group A, B and C were 21.2, 41.1<br />

and 49.6 months (P�0.001), respectively. Toxicities were generally mild<br />

to moderate. Conclusions: Chemotherapy significantly improved RFS and<br />

OS as adjuvant therapy for mucosal melanoma pts over HDI regimen or<br />

observation. This trial suggests that different subgroup <strong>of</strong> melanoma may<br />

need different adjuvant therapy.<br />

Melanoma/Skin Cancers<br />

8505 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Intermittent high-dose intravenous interferon alpha 2b (IFNa2b) for adjuvant<br />

treatment <strong>of</strong> stage III malignant melanoma: Final analysis <strong>of</strong> a<br />

randomized phase III DeCOG-trial (NCT00226408). Presenting Author:<br />

Peter Mohr, Elbe-Klinikum Buxtehude, Buxtehude, Germany<br />

Background: Adjuvant high-dose interferon (HDI) treatment <strong>of</strong> patients (pts)<br />

with malignant melanoma (MM) usually consists <strong>of</strong> 4 weeks initial<br />

intravenous (iv) infusion <strong>of</strong> 20 MU/m² IFNa2b followed by 11 months <strong>of</strong> 10<br />

MU/m² subcutaneously (sc). It is still unclear, whether both components<br />

are necessary for the efficacy <strong>of</strong> HDI in high-risk MM pts. The adjuvant<br />

phase III DeCOG MM-ADJ-5 trial evaluates efficacy, safety, tolerability and<br />

quality <strong>of</strong> life (QoL) <strong>of</strong> an intermittent (pulsed) high-dose iv IFNa2b<br />

regimen as compared to standard HDI. Methods: Patients with stage III<br />

(AJCC 2002) resected intransit or lymph node metastasis from cutaneous<br />

malignant melanoma were randomly assigned to receive either standard<br />

HDI regimen (Arm A), or 3 courses <strong>of</strong> IFNa2b 20 MU/m² iv on 5 days a week<br />

for 4 weeks repeated every 4 months (Arm B). Distant metastasis free<br />

survival (DMFS) was the primary endpoint for efficacy analysis. The EORTC<br />

QLQ C30 questionnaire was used before and during the study in a<br />

standardized way (weeks 0, 4, 16, 24, 40). In addition, patients completed<br />

a diary containing global QoL measures every week. Results: Out <strong>of</strong> 649<br />

patients who were enrolled, 22 patients had to be excluded from the<br />

intent-to-treat analysis. The remaining 627 patients were well balanced<br />

between both arms according to sex, age, and stage. Median follow-up was<br />

similar in both groups (55.4 vs. 55.3 months). Distant metastasis occurred<br />

in 138 (Arm A; 44%) vs. 145 (Arm B; 47%) patients without statistical<br />

significance (p�0.46). Survival was not significantly different for DMFS<br />

(HR 1.1; p�0.39) or OS (HR�1.0; p�0.85). Termination <strong>of</strong> treatment due<br />

to adverse events or QoL related reasons occurred significantly more <strong>of</strong>ten<br />

in arm A than in arm B (26.0% vs. 14.8%; p�0.001). The pulsed schedule<br />

resulted in less cumulative toxicity as compared to the standard regimen, in<br />

particular regarding fatigue and neuropsychiatric side effects. Conclusions:<br />

The final analysis <strong>of</strong> pulsed adjuvant HDI treatment showed no significant<br />

DMFS or OS differences in comparison to conventional HDI. There is a<br />

clearly favorable safety and QoL pr<strong>of</strong>ile <strong>of</strong> the pulsed regimen.<br />

8507 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

<strong>Clinical</strong> activity and safety <strong>of</strong> anti-PD-1 (BMS-936558, MDX-1106) in<br />

patients with advanced melanoma (MEL). Presenting Author: F. Stephen<br />

Hodi, Dana-Farber Cancer Institute, Boston, MA<br />

Background: BMS-936558 is a fully human mAb that blocks the programmed<br />

death-1 (PD-1) co-inhibitory receptor expressed by activated T<br />

cells. This study describes the activity and safety <strong>of</strong> BMS-936558 in<br />

patients (pts) with previously treated advanced MEL and underscores the<br />

importance <strong>of</strong> the PD-1/PD-L1 pathway in MEL therapy. Methods: BMS-<br />

936558 was administered IV Q2WK to pts with various solid tumors at<br />

doses <strong>of</strong> 0.1 to 10 mg/kg during dose-escalation and/or cohort expansion.<br />

Pts received up to 12 cycles (4 doses/cycle) <strong>of</strong> treatment or until PD or CR.<br />

<strong>Clinical</strong> activity was assessed by RECIST 1.0. Results: Of 240 pts treated as<br />

<strong>of</strong> July 1, 2011, 95 MEL pts were treated with BMS-936558 at 0.1<br />

(n�13), 0.3 (n�17), 1 (n�28), 3 (n�17), or 10 mg/kg (n�20). ECOG<br />

performance status was 0/1/2 in 56/36/3 pts. The majority <strong>of</strong> pts (60/95)<br />

had received prior immunotherapy (IT), primarily interferon-alpha or IL-2<br />

(prior anti-CTLA-4 excluded). Prior B-raf inhibitor therapy was noted in<br />

7/95 pts. The number <strong>of</strong> prior therapies was 1 (n�35), 2 (n�34), or �3<br />

(n�26). Sites <strong>of</strong> metastatic disease included lymph node (n�60), liver<br />

(n�32), lung (n�55), and bone (n�10). Median duration <strong>of</strong> therapy was<br />

15 wk (max 120 wk), with 40 pts still receiving treatment. The incidence <strong>of</strong><br />

grade 3-4 related AEs was 19% and included gastrointestinal (4%),<br />

endocrine (2%), and hepatobiliary disorders (1%). There were no drugrelated<br />

deaths in MEL pts. <strong>Clinical</strong> activity was observed at all dose levels<br />

(Table). Of 20 pts with OR at the time <strong>of</strong> data lock, 12 had OR duration �1<br />

yr and 6 pts were on study with OR duration between 1.9 and 11.3 mo. OR<br />

were seen in pts with visceral or bone metastases. Several pts had<br />

prolonged SD. Some had a persistent decrease in overall tumor burden in<br />

the presence <strong>of</strong> new lesions and were not categorized as responders.<br />

Conclusions: BMS-936558 had durable clinical benefit in pts with advanced<br />

MEL, including those who had received prior IT. Further development<br />

<strong>of</strong> BMS-936558 in MEL is ongoing.<br />

Dose<br />

(mg/kg) n a OR b uPR c RR d (%)<br />

PFS e rate<br />

at 24 wk (%)<br />

0.1 10 2 - 20 TBD f<br />

0.3 15 0 3 20 TBD<br />

1 28 7 1 29 TBD<br />

3 17 7 - 41 55<br />

10 20 4 - 20 30<br />

541s<br />

a Response evaluable pts; b CR or PR; c Unconfirmed PR; d Response rate [(OR �<br />

uPR) ÷ n]; e Progression-free survival; f To be determined (follow up ongoing).<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


542s Melanoma/Skin Cancers<br />

8508 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Ipilimumab (Ipi) expanded access program (EAP) for patients (pts) with<br />

stage III/IV melanoma: 10 mg/kg cohort interim results. Presenting Author:<br />

Omid Hamid, The Angeles Clinic and Research Institute, Santa Monica, CA<br />

Background: The EAP (CA184-045), begun in 8/07, currently provides the<br />

largest safety database for Ipi outside <strong>of</strong> controlled clinical trials (CCTs)<br />

and included pts with brain metastases (BM) and primary ocular (OM) and<br />

mucosal (MM) melanoma. The EAP was amended in 10/08 to administer a<br />

3 mg/kg dose based on the current label (Hodi et al. NEJM 2010). We now<br />

report interim data from pts treated in the US at 10 mg/kg from<br />

8/07-10/08. Methods: Pts with unresectable Stage III or IV melanoma who<br />

progressed on at least 1 systemic therapy or had no alternate treatment<br />

options were eligible. Pts received 10 mg/kg Ipi i.v. q 3 wks up to 4 doses<br />

(induction) followed by 10 mg/kg q 12 wks (maintenance) until no longer<br />

clinically benefiting or unacceptable/unmanageable toxicity occurred. All<br />

serious adverse events (SAEs) and on-study deaths were collected; overall<br />

survival (OS) was assessed retrospectively. Results: Of 906 treated pts, 830<br />

were included in the analysis; 39 from prior Ipi trials � 37 from sites whose<br />

IRB did not agree to collect OS were excluded. Pts got a median <strong>of</strong> 4 doses;<br />

31% got �5 doses. ECOG 0/1/2 was 53%/39%/8% (1 pt was ECOG 3);<br />

27% had BM, 5% had OM, and 4% MM. Primary reasons for discontinuation<br />

were disease progression (67%) and drug toxicity (11%). Incidence <strong>of</strong><br />

drug-related SAEs was 27% with diarrhea 10%, colitis 8%, endocrinopathies<br />

4%, and dermatitis 0.8%. 2 pts (0.24%) had on-study hepatitis<br />

SAEs, both considered drug-related and resulted in discontinuation. There<br />

were 3 (0.36%) drug-related intestinal perforations. 2 deaths (0.24%)<br />

were the outcome <strong>of</strong> drug-related SAEs; 1 multi-organ failure and 1 acute<br />

respiratory distress syndrome (both �70 days from last induction dose). 72<br />

(9%) pts currently remain on treatment (i.e. �3 years). Available OS data<br />

showed that 138 pts (17%) were still at risk after 3 years. For 27% <strong>of</strong> pts<br />

the last known alive date was �30 days before data cut<strong>of</strong>f (with most �2<br />

yrs). Conclusions: Data from the US EAP must be interpreted with caution<br />

compared to CCTs. To date, incidence <strong>of</strong> SAEs for hepatitis and intestinal<br />

perforation, and drug-related death at 10 mg/kg Ipi monotherapy is<br />

consistent with that seen in BMS clinical trials. Durable OS (�3 yrs) was<br />

observed in at least 17% <strong>of</strong> pts.<br />

8510 <strong>Clinical</strong> Science Symposium, Mon, 3:00 PM-4:30 PM<br />

Updated safety and efficacy results from a phase I/II study <strong>of</strong> the oral BRAF<br />

inhibitor dabrafenib (GSK2118436) combined with the oral MEK 1/2<br />

inhibitor trametinib (GSK1120212) in patients with BRAFi-naive metastatic<br />

melanoma. Presenting Author: Jeffrey S. Weber, Comprehensive<br />

Melanoma Research Center, H. Lee M<strong>of</strong>fitt Cancer Center, Tampa, FL<br />

Background: In preclinical models, the BRAFi/MEKi combination has<br />

demonstrated enhanced activity against BRAF-mutant cancer cells compared<br />

with either drug alone, delayed emergence <strong>of</strong> BRAFi resistance, and<br />

prevented BRAFi-related proliferative skin lesions. A 3-part study investigating<br />

the dabrafenib/trametinib combination was conducted in patients (pts)<br />

with V600 BRAF mutant solid tumors. Interim data from the study were<br />

previously reported (Infante, ASCO 2011); updated safety and efficacy<br />

data are presented. Methods: In <strong>Part</strong> 2, 125 pts with V600 BRAF mutant<br />

solid tumors enrolled, including 77 melanoma pts with no prior BRAFi, and<br />

measurable disease according to RECIST 1.1. Pts were treated on 4<br />

escalating dose levels <strong>of</strong> dabrafenib/trametinib (mg BID/mg QD): 75/1,<br />

150/1, 150/1.5, 150/2. Demographic and efficacy data for the 77<br />

melanoma pts with no prior BRAFi and safety data for all 125 <strong>Part</strong> 2 pts are<br />

reported. Results: Among 77 melanoma pts, median age was 52 years, 61%<br />

male, 57% ECOG PS <strong>of</strong> 0, 91% V600E, 65% M1c stage, 26% prior brain<br />

metastases, and 52% LDH � ULN. Confirmed ORR was 56% (95% CI:<br />

44.1%-67.2%) with 4 CR, 39 PR, 29 SD and, 3 PD. Confirmed response<br />

rate for each dose level, respectively, was 67% (n�6), 64% (n�22), 48%<br />

(n�25), and 54% (n�24). Median PFS (months) for each dose level,<br />

respectively, was: 8.7, 8.3, 5.5; PFS is not mature for 150/2. Overall PFS<br />

was 7.4 (95% CI: 5.5-9.2). Among the 125 pts, there were 2 grade (G) 5<br />

adverse events (AEs), pneumonia and hyponatraemia. The most common<br />

G3/4 AEs were pyrexia (n�6, 5%), fatigue (n�6, 5%) and dehydration<br />

(n�6, 5%). Skin toxicity � G2 occurred in 17 (14%) pts. Cutaneous<br />

squamous cell carcinoma occurred in 3 (2%) pts and actinic keratoses in 2<br />

(2%). Conclusions: The combination <strong>of</strong> dabrafenib/trametinib has an<br />

acceptable safety pr<strong>of</strong>ile, with a lower incidence <strong>of</strong> MEKi-related rash and<br />

BRAFi-induced hyperproliferative skin lesions compared with the single<br />

agents. The clinical activity <strong>of</strong> dabrafenib/trametinib observed in pts with<br />

V600 BRAF mutant metastatic melanoma is encouraging and will be<br />

investigated further in a phase III trial.<br />

LBA8509 <strong>Clinical</strong> Science Symposium, Mon, 3:00 PM-4:30 PM<br />

METRIC phase III study: Efficacy <strong>of</strong> trametinib (T), a potent and selective<br />

MEK inhibitor (MEKi), in progression-free survival (PFS) and overall<br />

survival (OS) compared with chemotherapy (C) in patients (pts) with<br />

BRAF mutant advanced or metastatic melanoma (MM). Presenting<br />

V600/k<br />

Author: Caroline Robert, Institut Gustave Roussy, Villejuif, France<br />

The full, final text <strong>of</strong> this abstract will be available at<br />

abstract.asco.org at 12:01 AM (EDT) on Monday, June 4,<br />

2012, and in the <strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong> online<br />

supplement to the June 20, 2012, issue <strong>of</strong> Journal <strong>of</strong><br />

<strong>Clinical</strong> Oncology. Onsite at the <strong>Meeting</strong>, this abstract will<br />

be printed in the Monday edition <strong>of</strong> ASCO Daily News.<br />

8511 <strong>Clinical</strong> Science Symposium, Mon, 3:00 PM-4:30 PM<br />

Efficacy and safety <strong>of</strong> oral MEK162 in patients with locally advanced and<br />

unresectable or metastatic cutaneous melanoma harboring BRAFV600 or<br />

NRAS mutations. Presenting Author: Paolo Antonio Ascierto, Unit <strong>of</strong><br />

Medical Oncology and Innovative Therapy, Istituto Nazionale Tumori<br />

Fondazione Pascale, Napoli, Italy<br />

Background: BRAF and NRAS mutations occur in 50-60% and 15-20% <strong>of</strong><br />

cutaneous melanomas, respectively. MEK162, a selective inhibitor <strong>of</strong> the<br />

kinases MEK1 and MEK2, has shown pre-clinical activity in BRAF and<br />

NRAS mutant (mt) melanoma models. This open label, phase II study<br />

assessed the antitumor activity <strong>of</strong> MEK162 in patients (pts) with BRAFV600<br />

and NRAS mt advanced cutaneous melanoma. Methods: MEK162 was<br />

administered orally at a starting dose <strong>of</strong> 45 mg twice daily. Treatment was<br />

until unacceptable toxicity, disease progression (PD) or investigator or<br />

patient refusal. Tumor response was assessed by CT imaging every 8 weeks<br />

(RECIST 1.0) until PD. Results: As <strong>of</strong> 16 Sept 2011, the full analysis and<br />

safety populations comprised 66 pts: 42 BRAF mt and 24 NRAS mt.<br />

Median age 58.0 years; 57.6% male; 72.7% WHO performance status 0.<br />

All NRAS pts and all but 2 BRAF (1 each stage IIIB and IIIC) pts had stage<br />

IV disease, and 87.5% <strong>of</strong> NRAS pts and 66.7% <strong>of</strong> BRAF pts had received<br />

prior therapy at study entry. Median time from 1st diagnosis to 1st dose <strong>of</strong><br />

drug was 40.4 months. Median time on study was 10.4 and 8.5 weeks for<br />

the BRAF and NRAS arms. Relative dose intensities <strong>of</strong> 80–�100% were<br />

received by 71.4% and 70.8% <strong>of</strong> pts, respectively. Among 29 BRAF mt<br />

and 13 NRAS mt pts evaluable for efficacy, 1 confirmed and 6 unconfirmed<br />

partial responses (PRs) and 9 pts with stable disease (SD) were recorded in<br />

the BRAF arm and 2 confirmed PRs, 1 unconfirmed PR and 4 pts with SD<br />

recorded in the NRAS arm. Common treatment–related adverse events<br />

(AEs), all grades (Gs) and all pts, were rash (40.9%), diarrhea (33.3%),<br />

acneiform dermatitis (27.3%), creatine phosphokinase (CK) elevation<br />

(25.8%), fatigue (18.2%) and peripheral edema (21.2%). Central serous<br />

retinopathy-like retinal events (G 1/2 only) were reported in 8 (12.1%) pts<br />

(6 G1, 2 G2). All retinal events were reversible. G3/4 AEs in �1pt were<br />

diarrhea (4.5%) and CK elevation (15.2%). 5 pts discontinued due to<br />

toxicity. 34 pts are ongoing with more responses under current review.<br />

Conclusions: MEK162 showed clinical activity and good tolerability in pts<br />

with BRAF and NRAS mt advanced melanoma. This is the 1st targeted<br />

therapy to show activity in pts with NRAS mt melanoma.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


8512 Poster Discussion Session (Board #1), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Safety <strong>of</strong> ipilimumab in patients (pts) with untreated, advanced melanoma<br />

alive beyond 2 years: Results from a phase III study. Presenting Author: Luc<br />

Thomas, Lyon 1 University Centre Hospitalier Lyon Sud, Pierre Benite,<br />

France<br />

Background: Ipilimumab (IPI), a fully human monoclonal antibody, blocks<br />

cytotoxic T-lymphocyte antigen-4 to potentiate an antitumor T-cell response.<br />

In a phase 3 study (CA184-024) <strong>of</strong> previously untreated pts with<br />

stage III or IV melanoma, IPI � dacarbazine (DTIC) significantly improved<br />

overall survival (OS) vs. DTIC alone (Robert et al. NEJM 2011). We now<br />

report safety data <strong>of</strong> IPI in pts from this study alive � 2 yrs from study<br />

initiation. Methods: Pts with untreated advanced melanoma, were randomized<br />

to IPI (10 mg/kg) � DTIC (850 mg/m2 ) or placebo � DTIC (850<br />

mg/m2 ) given at Wks 1, 4, 7, 10 followed by DTIC q 3 wks through Wk 22.<br />

Eligible pts (stable disease or better) received IPI or placebo q 12 wks as<br />

maintenance. In the population <strong>of</strong> subjects alive �2 yrs, the appearance <strong>of</strong><br />

immune-related adverse events (irAEs) occurring after 2 yrs was evaluated.<br />

Within this group was a subset <strong>of</strong> subjects still receiving IPI dosing after 2<br />

yrs; safety for these pts was evaluated to assess the impact <strong>of</strong> prolonged IPI<br />

exposure. Results: In the IPI � DTIC group 68 (28%) pts survived � 2 yrs<br />

compared to 44 (18%) in the DTIC alone group; 11 <strong>of</strong> the 68 continued IPI<br />

dosing for � 2 yrs. Safety assessment beyond 2 yrs showed 3 <strong>of</strong> the 11 pts<br />

had any grade irAEs; 1 pt had grade 3/4 rash, pruritus while low grade<br />

events included rash, pruritus (n�2) and elevated ALT / AST (n�1). Overall<br />

among all 68 pts in the Ipi � DTIC group, there were 5 pts (7.4%) with any<br />

grade irAEs including grade 3/4 rash, pruritus (n�1) and low grade rash<br />

(n�3), pruritus (n�2), skin hypopigmentation, and elevated ALT / AST<br />

(n�1). No gastrointestinal or endocrine events (any grade) were observed.<br />

Conclusions: In Study 024, IPI � DTIC treatment improved OS pts with<br />

untreated, advanced melanoma with higher survival rates in the IPI � DTIC<br />

group at 1 yr (47.3% vs. 36.3%), 2 yrs (28.5% vs. 17.9%), and 3 yrs<br />

(20.8% vs. 12.2%) (HR 0.72, p�0.001). The safety pr<strong>of</strong>ile <strong>of</strong> pts alive<br />

after 2 yrs suggests that treatment with IPI � DTIC is associated with low<br />

rates <strong>of</strong> irAEs in these pts. Furthermore, in pts still receiving active IPI<br />

treatment beyond � 2 yrs, the safety pr<strong>of</strong>ile appears to be consistent and<br />

medically manageable using established safety guidelines (Weber, Oncologist<br />

2007).<br />

8514 Poster Discussion Session (Board #3), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Phase II study <strong>of</strong> the frontline combination <strong>of</strong> ipilimumab and temozolomide<br />

in patients with metastatic melanoma. Presenting Author: Sapna<br />

Pradyuman Patel, University <strong>of</strong> Texas M. D. Anderson Cancer Center,<br />

Houston, TX<br />

Background: Ipilimumab (Ipi) alters the immune system balance by<br />

inhibiting the suppression <strong>of</strong> T-cell function. In two phase III trials, Ipi has<br />

shown an overall survival benefit alone and in combination with dacarbazine<br />

in previously treated and treatment-naïve patients (pts) with metastatic<br />

melanoma (MM), respectively. We performed a single-institution,<br />

phase II clinical trial <strong>of</strong> Ipi plus temozolomide (Tem) in pts with MM.<br />

Methods: Pts between the ages <strong>of</strong> 18 and 75 with previously untreated<br />

unresectable stage III or stage IV MM and an ECOG Performance Status <strong>of</strong><br />

0 to 1 were enrolled in a phase II trial <strong>of</strong> Ipi plus Tem. Induction phase<br />

consisted <strong>of</strong> Ipi 10mg/kg intravenous on Day 1 and oral Tem 200 mg/m2 on<br />

Days1–4every 3 weeks for 4 doses. Maintenance consisted <strong>of</strong> Ipi 10<br />

mg/kg intravenous on Day 1 starting week 12 and repeated every 12 weeks<br />

and oral Tem 200 mg/m2 onDays1–5starting week 12 and repeated every<br />

4 weeks until disease progression or unacceptable toxicity occurred. The<br />

primary endpoint was progression-free survival (PFS) rate at 6 months.<br />

Responses were evaluated using immune-related response criteria. Results:<br />

Sixty-four pts were enrolled and received at least one dose <strong>of</strong> study drug. All<br />

pts were included in the analysis. With a median follow-up <strong>of</strong> 8.5 months,<br />

the PFS rate at 6 months was 43%, exceeding the proposed rate <strong>of</strong> 30%,<br />

and the median PFS was 5.1 months. There were 10 (15.6%) confirmed<br />

complete responses and 8 (12.5%) confirmed partial responses. At the<br />

time <strong>of</strong> this analysis, median overall survival has not been reached.<br />

Immune-related adverse events (irAEs) were experienced by 88% <strong>of</strong> pts,<br />

most commonly pruritus (88%), rash (83%), diarrhea (56%), transaminitis<br />

(45%), and colitis (11%). Grade 3/4 irAEs seen in more than one patient<br />

were skin rash (11%), diarrhea (9%), pruritus (6%), and transaminitis<br />

(5%). Constipation occurred in 70% <strong>of</strong> pts and was the most common<br />

gastrointestinal (GI) toxicity. There were no GI perforations or deaths on<br />

study due to treatment. Conclusions: At a median follow-up <strong>of</strong> 8.5 months,<br />

the best overall response rate in this study is 28%. Ipi at 10 mg/kg in<br />

combination with Tem given in an induction followed by maintenance<br />

fashion is safe, well-tolerated, and efficacious in MM.<br />

Melanoma/Skin Cancers<br />

543s<br />

8513 Poster Discussion Session (Board #2), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Phase II multicenter trial <strong>of</strong> ipilimumab combined with fotemustine in<br />

patients with metastatic melanoma: The Italian Network for Tumor Biotherapy<br />

(NIBIT)-M1 trial. Presenting Author: Anna Maria Di Giacomo,<br />

Medical Oncology and Immunotherapy, University Hospital <strong>of</strong> Siena,<br />

Siena, Italy<br />

Background: Ipilimumab (ipi), an antibody against cytotoxic T-lymphocyteassociated<br />

antigen-4, improves survival in patients (pts) with metastatic<br />

melanoma (MM); however, objective tumor responses are limited. NIBIT-M1<br />

aims to investigate the efficacy and safety <strong>of</strong> ipi plus fotemustine (FTM), a<br />

cytotoxic alkylating drug, in pts with MM. Methods: Eligible pts, with or<br />

without brain metastases, received induction therapy with ipi 10 mg/kg<br />

every 3 weeks (Q3W) for four doses and FTM 100 mg/m2 weekly for 3<br />

weeks. Ipi and FTM maintenance therapy was provided Q12W from Week<br />

24 and Q3W from Week 9, respectively. The primary objective was the<br />

immune-related (ir) disease control rate (irDCR: pts with complete response<br />

[CR], partial response [PR] or stable disease [SD] as determined<br />

using the ir response criteria). Secondary objectives included ir objective<br />

response rate (ORR), duration <strong>of</strong> response (DOR) and progression-free<br />

survival (PFS); overall survival (OS), and safety. Tumor assessments were<br />

performed Q8W from Week 12 to Week 36 and Q12W thereafter. Results:<br />

Among 86 pts with unresectable stage III (n�3) or stage IV (n�83) MM<br />

treated at 7 NIBIT centers, 42 were previously untreated, 44 had<br />

progressed following first-line treatment and 20 had asymptomatic brain<br />

metastases. As <strong>of</strong> December 2011, the irDCR was 46.5% (40/86; 95% CI,<br />

35.7–57.6%); the irORR was 29.1% (95% CI, 19.8–39.8%; 5 CRs and<br />

20 PRs) and with a median 8.3 months follow-up, median irPFS was 5.3<br />

months (95% CI, 3.5–7.1). The 1-year OS rate was 51.8% (95% CI,<br />

37.5–66.1%); median OS was not yet reached. Among all pts, 58.1% and<br />

87% completed ipi or FTM induction, respectively. The most common<br />

grade 3/4 drug-related adverse events (AEs) (reported in 54.6% pts overall)<br />

were myelotoxicity (43.5%), increased ALT/AST (14.1/10.6%), gastrointestinal<br />

(4.7%) and skin-related (2.3%). AEs were generally manageable and<br />

reversible per protocol guidance. Conclusions: The study reached its<br />

primary objective with 46.5% <strong>of</strong> pts achieving disease control. The<br />

combination <strong>of</strong> ipi plus FTM is safe; the irDCR, 1-year OS rate and median<br />

irPFS warrant its further investigation in MM pts.<br />

8515 Poster Discussion Session (Board #4), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Hypersensitivity skin reactions in melanoma patients treated with vemurafenib<br />

after ipilimumab therapy. Presenting Author: James J. Harding,<br />

Memorial Sloan-Kettering Cancer Center, New York, NY<br />

Background: Ipilimumab (IPI) and vemurafenib (VEM) each improve overall<br />

survival for patients (pts) with metastatic melanoma. Both are FDAapproved<br />

and are being used in pts with BRAFV600E-mutated metastatic<br />

melanoma. We previously described cases <strong>of</strong> prominent skin eruptions<br />

associated with VEM in pts who had previously received IPI. Methods: We<br />

have updated our experience <strong>of</strong> BRAFV600E-mutated melanoma pts treated<br />

with VEM who had previously received IPI. Pts were treated at our center<br />

from January 2007 to January 2012. Data were collected under an<br />

approved IRB waiver. Results: Sixteen melanoma pts were treated with VEM<br />

after having received IPI. The most common drug-related adverse event<br />

(AE) associated with VEM was rash, occurring in 13/16 patients (81.3%,<br />

95% CI 56.5-93.2). Four pts developed a severe, Grade 3, maculopapular<br />

rash within 8 days <strong>of</strong> starting VEM. Biopsies in 2 pts revealed spongiotic<br />

and perivascular dermatitis with eosinophils consistent with a drug hypersensitivity<br />

reaction. Hypersensitivity reactions did not progress to lifethreatening<br />

reactions such as anaphylaxis or Stevens-Johnson syndrome,<br />

nor did they result in VEM dose discontinuation. Reactions were managed<br />

with corticosteroids and dose modifications. Grade 3 rash strongly correlated<br />

with initiating VEM within one month <strong>of</strong> IPI (Fisher’s exact test, p �<br />

0.007) and was not associated with the dose <strong>of</strong> prior IPI, the number <strong>of</strong><br />

prior doses, or immune-related AEs. The incidence <strong>of</strong> Grade 3 rash in this<br />

pt cohort was significantly higher than in pts treated on the phase III trial <strong>of</strong><br />

VEM (4/16, 25% versus 28/336, 8%; �2 � 5.13, Df � 1, p � 0.02). The<br />

objective overall response for VEM was 50% (95% CI 26.5-73.4), which is<br />

similar to response rates seen on the phase II and III trials. Conclusions: In<br />

pts receiving VEM who have previously received IPI, dermatologic AEs<br />

appear to be more common. This effect seemed most pronounced if VEM<br />

was given within one month <strong>of</strong> completing IPI. Although more data are<br />

necessary to confirm this apparent association, we speculate that the<br />

release <strong>of</strong> immune checkpoint inhibition by IPI may predispose pts to<br />

hypersensitivity skin reactions to VEM.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


544s Melanoma/Skin Cancers<br />

8516 Poster Discussion Session (Board #5), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Predicting early relapse in patients with BRAFV600E melanoma with a highly<br />

sensitive blood BRAF assay. Presenting Author: Ryan J. Sullivan, Massachusetts<br />

General Hospital Cancer Center, Boston, MA<br />

Background: Many patients (pts) with metastatic melanoma (MM) who<br />

progress on BRAF inhibitors (BRAFi) develop rapidly progressive disease<br />

(PD) which is difficult to manage. Identification <strong>of</strong> an early marker <strong>of</strong> PD<br />

may allow for therapeutic intervention prior to clinical deterioration. We<br />

have developed a highly sensitive and inexpensive assay in our laboratory<br />

which can detect as little as 1 BRAFV600E mutant melanoma cell in<br />

400,000 peripheral blood lymphocytes (PBL). We aimed to determine the<br />

utility <strong>of</strong> our assay as a tool to follow pts with BRAF mutant MM who are<br />

being treated with a BRAFi. Methods: Every pt had a BRAFV600E mutation<br />

detected from a tumor sample in a CLIA-approved laboratory prior to<br />

commencing BRAFi therapy. 20 pts with stage IV BRAFV600E MM underwent<br />

serial venopunctures before and every 4 weeks during treatment with<br />

a BRAFi. BRAFV600E level was quantified using our proprietary assay (Panka<br />

et al Melanoma Research 2010). Serial BRAFV600E levels were generated<br />

for every pt and normalized to the pre-treatment value. BRAFV600E level was<br />

then correlated with response to therapy and PD. Results: BRAFV600E was<br />

detected in the PBL <strong>of</strong> each pt. To date, 10 pts have had BRAFV600E levels<br />

quantified at 3 or more time points (data for all 20 pts will be available at<br />

time <strong>of</strong> presentation). Relative BRAFV600E levels reduced by half following<br />

the first and second cycles <strong>of</strong> treatment in 7 pts, which correlated with<br />

disease regression in each pt. Following the third cycle <strong>of</strong> treatment, the<br />

relative BRAFV600E levels varied greatly with a subset <strong>of</strong> pts having<br />

continued suppression while others having a rise in their BRAFV600E levels.<br />

In each pt with PD, the BRAF assay showed an increase � 4 wks in advance<br />

<strong>of</strong> clinically or radiographically defined PD. Conclusions: Our assay is able to<br />

quantify changes in BRAFV600E levels in the blood <strong>of</strong> pts with BRAF mutant<br />

MM receiving BRAFi therapy. In these pts, the BRAFV600E levels are<br />

reduced in the setting <strong>of</strong> initial disease regression and increase well in<br />

advance <strong>of</strong> clinical or radiographic PD. While further development is<br />

necessary, such a test could be used to identify pts who may be selected to<br />

receive alternative therapy prior to clinical or radiographic PD.<br />

8518 Poster Discussion Session (Board #7), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Tumor-specific circulating cell-free DNA (cfDNA) BRAF mutations (muts)<br />

to predict clinical outcome in patients (pts) treated with the BRAF inhibitor<br />

dabrafenib (GSK2118436). Presenting Author: Georgina V. Long, Melanoma<br />

Institute Australia, Westmead Institute for Cancer Research and<br />

Westmead Hospital, The University <strong>of</strong> Sydney, Sydney, Australia<br />

Background: Tumor specific cfDNA levels in blood increase with tumor<br />

burden and decrease following treatment. cfDNA can harbor muts consistent<br />

with the tumor. Thus cfDNA could be a useful biomarker <strong>of</strong> therapeutic<br />

response. BREAK-2, an open label, single arm, Phase II study evaluated<br />

efficacy, safety and tolerability <strong>of</strong> dabrafenib in BRAF V600E/K mut�<br />

metastatic melanoma pts. Exploratory objectives <strong>of</strong> BREAK-2 were to<br />

evaluate whether (i) tumor and cfDNA BRAF muts are correlated; (ii) cfDNA<br />

levels correlate with baseline tumor burden and (iii) cfDNA muts predict<br />

clinical outcome with dabrafenib. Methods: BRAF mut status was established<br />

for 92pts using an allele-specific PCR assay in tumor samples.<br />

Baseline plasma samples were available for 91/92 pts. cfDNA BRAF mut<br />

status was evaluated by Inostics GmBH using the BEAMing technology.<br />

Spearman correlation coefficients (R) were used to determine the association<br />

between cfDNA fraction (mut DNA molecules � 0.01%) and estimated<br />

baseline tumor burden, calculated by the sum <strong>of</strong> RECIST measurements <strong>of</strong><br />

target lesions. Logistic regression and Cox proportional hazards models<br />

were used to assess the association between cfDNA mut status and<br />

objective response rate (ORR) and progression free survival (PFS), respectively.<br />

Results: The overall agreement between tumor and cfDNA BRAF<br />

V600E and V600K mut status was 83%, and 96% respectively. Higher<br />

cfDNA V600E mut fraction was associated with higher baseline tumor<br />

burden (R�0.73; p-value � 0.0001; n�60); lower ORR (O.R. � 0.83;<br />

95% CI � 0.72, 0.96; p-value�0.0134; n�46) and shorter PFS<br />

(H.R.�1.09; p-value�0.0006; n�46). Median PFS was 27.4 weeks in the<br />

overall V600E pt population (n�76) and 20.0 weeks in the cfDNA V600E<br />

pt population (n�46). Otherwise, the response endpoints were comparable<br />

between the two populations. There was no correlation between V600K mut<br />

fraction (n�14) and any efficacy endpoints. Conclusions: cfDNA was useful<br />

for detecting BRAF muts in pts treated with dabrafenib and increasing<br />

V600E mut fraction was associated with reduced ORR and shorter PFS,<br />

suggesting higher amounts <strong>of</strong> mut cfDNA in V600E mut� pts predicts<br />

poorer clinical outcome.<br />

8517 Poster Discussion Session (Board #6), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

An open-label, multicenter safety study <strong>of</strong> vemurafenib (PLX4032,<br />

RO5185426) in patients with metastatic melanoma. Presenting Author:<br />

James M. G. Larkin, Royal Marsden Hospital, London, United Kingdom<br />

Background: Vemurafenib, a BRAF inhibitor, is associated with improved<br />

PFS and OS in patients (pts) with BRAFV600-mutant metastatic melanoma<br />

(mM). We present preliminary safety and efficacy findings from a safety<br />

study <strong>of</strong> vemurafenib in pts with unresectable stage IIIC/IV mM with<br />

BRAFV600 mutations. Methods: Pts with untreated or previously treated<br />

stage IIIC/IV BRAFV600 mutation-positive (cobas 4800 BRAF V600 Mutation<br />

Test) melanoma were enrolled. Pts received continuous oral vemurafenib<br />

960 mg bid. Primary study endpoint was safety; efficacy (RECIST V<br />

1.1) was a secondary endpoint. Results: Of 1,964 screened pts between<br />

Mar and Sep 2011, 914 (47%) were enrolled and 834 were evaluable for<br />

safety. Median age was 53 (21–88 years), 55% males. Median time since<br />

first mM diagnosis was 7.6 months (0–18 years). At baseline, 80% <strong>of</strong> pts<br />

had ECOG PS 0–1, 11% ECOG PS 2 (missing 9%); 27% <strong>of</strong> pts had brain<br />

metastases, and 31% had elevated LDH. Most pts had received prior<br />

systemic therapy (70%) including ipilimumab (14%), MEK and BRAF<br />

inhibitors (2%). At data cut-<strong>of</strong>f (Sep 30, 2011), median treatment duration<br />

was 68 days (1–223 days) with 87% <strong>of</strong> pts still on treatment. Of 834 pts,<br />

553 (66%) to date have reported AEs. Of 553 pts reporting AEs, 88% were<br />

related to vemurafenib, 33% Grade 3, and 1.9% Grade 4. The most<br />

common (�1%) Grade 3/4 AEs were rash (3.6%), arthralgia (3.1%), and<br />

cutaneous squamous cell carcinoma/keratoacanthoma (4.3%). Most common<br />

AEs (�10%) <strong>of</strong> any grade were arthralgia (31%), rash (29%), fatigue<br />

(22%), photosensitivity (21%), nausea (15%), and were similar irrespective<br />

<strong>of</strong> brain metastases and ECOG PS. AEs caused treatment interruption<br />

in 141 (17%) pts. Of 109 pts who discontinued treatment (13%), main<br />

reasons for withdrawal were progressive disease (60%), death (20%), AEs<br />

(6%; most commonly arthritis and abdominal pain). Tumor assessments at<br />

Week 8 <strong>of</strong> treatment were available for 302/834 (36%) pts, 61% pts<br />

achieved CR or PR, and 29% had SD. Conclusions: In a setting representative<br />

<strong>of</strong> routine clinical practice, vemurafenib is seen to be well tolerated and<br />

both safety pr<strong>of</strong>ile and activity resemble the phase I–III data although this<br />

analysis is limited by the study duration.<br />

8519 Poster Discussion Session (Board #8), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Safety and efficacy <strong>of</strong> MET inhibitor tivantinib (ARQ 197) combined with<br />

sorafenib in patients (pts) with NRAS wild-type or mutant melanoma from a<br />

phase I study. Presenting Author: Julie A. Means, Vanderbilt University<br />

Medical Center, Nashville, TN<br />

Background: The MET receptor tyrosine kinase is implicated in tumor cell<br />

proliferation, invasion, and metastasis, and is activated in NRAS mutant<br />

melanoma. Tivantinib is an oral, selective MET inhibitor currently in phase<br />

II/III clinical trials. Tivantinib plus sorafenib exhibited synergistic antitumor<br />

activity vs single-agent activity in several tumor models. This phase I<br />

dose-escalation study assessed the safety <strong>of</strong> tivantinib plus sorafenib in pts<br />

with advanced solid tumors. Methods: Endpoints were safety, the recommended<br />

phase II dose (RP2D) <strong>of</strong> tivantinib plus sorafenib, and antitumor<br />

activity. Dose escalation previously established the RP2D as tivantinib 360<br />

mg twice daily (BID) plus sorafenib 400 mg BID. Extension cohorts<br />

enrolled � 20 pts each with melanoma or other tumors. Pts were treated<br />

until disease progression or unacceptable toxicity. Results: 16 pts with<br />

melanoma (median age, 66 yr) received treatment at the RP2D, and 3 pts<br />

are still on study. 12 pts received � 1 previous systemic anticancer<br />

treatment (median, 1.2; range, 0-5) including ipilimumab (2 pts) or MEK<br />

inhibitor (1 pt). Common adverse events (� 25%) were rash (50%),<br />

diarrhea and fatigue (44% each), anorexia (38%), stomatitis and nausea<br />

(31% each), and anemia, weight decrease, and hypophosphatemia (25%<br />

each). Best responses were complete response (CR) in 1 pt, partial<br />

response (PR) in 3 pts, and stable disease (SD) in 3 pts. 4 pts had<br />

progressive disease and 5 pts were not evaluable (3 pts had not reached<br />

first assessment time, 1 pt withdrew consent, and 1 pt had unacceptable<br />

toxicity). The overall response rate and disease control rate were 25% and<br />

44%, respectively. Median progression-free survival (mPFS) was 5.3 mo<br />

(95% CI, 1.6-12.9 mo). Among 8 pts with NRAS mutations, mPFS was 9.2<br />

mo (95% CI, 5.3-12.9 mo) and responses were 1 CR, 1 PR, and 2 SD.<br />

Conclusions: Tivantinib plus sorafenib combination therapy was well<br />

tolerated and exhibited preliminary anticancer activity in pts with melanoma.<br />

Dual inhibition <strong>of</strong> MET and angiogenesis may be an effective<br />

treatment strategy in NRAS-mutant melanoma.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


8520 Poster Discussion Session (Board #9), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

<strong>Clinical</strong> significance <strong>of</strong> genomic alterations <strong>of</strong> the CDK4-pathway and<br />

sensitivity to the CDK4 inhibitor PD 0332991 in melanoma. Presenting<br />

Author: Grant A. McArthur, Department <strong>of</strong> Medical Oncology, Peter<br />

MacCallum Cancer Centre, Melbourne, Australia<br />

Background: Activation <strong>of</strong> CDK4 by amplification, increased expression <strong>of</strong><br />

Cyclin D1 (CCND1) or reduced expression <strong>of</strong> the CDK inhibitor p16<br />

(CDKN2A) can contribute to transformation <strong>of</strong> melanocytes indicating that<br />

CDK4 can act as an oncogene in melanoma.To explore if CDK4 may be a<br />

viable target for the treatment <strong>of</strong> human melanoma we have analyzed the<br />

frequency and clinico-pathological associations <strong>of</strong> genomic alterations <strong>of</strong><br />

the CDK4 pathway in primary human melanoma and examined the genomic<br />

predictors <strong>of</strong> sensitivity to the highly selective CDK4/6 inhibitor PD<br />

0332991 (991) in a panel <strong>of</strong> melanoma cell lines. Methods: A series <strong>of</strong> 167<br />

primary melanomas with clinical, molecular and pathological annotation,<br />

including median follow up <strong>of</strong> 6.6 years, were analyzed for copy number<br />

variation (CNV)- gain <strong>of</strong> CDK4 or CCND1 (average gene copy �2.4) or loss<br />

<strong>of</strong> CDKN2A (average gene copy �1.4), by fluorescence in situ hybridization.<br />

A panel <strong>of</strong> 39 cell lines were treated with 991 in vitro and GI50s<br />

calculated. The mean GI50 <strong>of</strong> the melanoma cell lines was used to define<br />

sensitivity. Gene expression pr<strong>of</strong>iling and mutation or CNV in CDKN2A were<br />

used to identify predictors <strong>of</strong> sensitivity. Results: 75% <strong>of</strong> primary melanomas<br />

had at least one CNV (75%, 70% and 82% for BRAF, NRAS or<br />

wild-type BRAF/NRAS mutation status respectively). 55% showed loss <strong>of</strong><br />

CDKN2A. 28% <strong>of</strong> melanomas had two or more CNVs. Melanomas with two<br />

or more CNVs involving CCND1 had worse overall survival (HR 5.56,<br />

p�0.02). Low CDKN2A mRNA expression or mutation or loss <strong>of</strong> CDKN2A<br />

predicted sensitivity to 991 with 30/33 mutant/loss lines being sensitive<br />

compared to only 2/6 wild type lines (p�0.006). Expression <strong>of</strong> CDK4,<br />

CCND1 or other cyclins or CDK-inhibitors did not predict sensitivity to 991.<br />

Conclusions: Genomic alterations in the CDK4 pathway are frequent in<br />

melanoma and are associated with worse survival, particularly when<br />

melanomas harbor two or more CNVs involving CCND1. Mutation, loss or<br />

low expression <strong>of</strong> CDKN2A in melanoma cell lines predicted sensitivity to<br />

the CDK4 inhibitor 991. Taken together these data support evaluation <strong>of</strong><br />

CDK4 inhibitors in melanoma and suggest that CDKN2A maybe a genomic<br />

predictor <strong>of</strong> sensitivity to these agents.<br />

8522 Poster Discussion Session (Board #11), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

A phase II trial <strong>of</strong> dasatinib in patients with unresectable locally advanced<br />

or stage IV mucosal, acral, and solar melanomas: An Eastern Cooperative<br />

Oncology Group study (E2607). Presenting Author: Kevin Kalinsky, Columbia<br />

University Medical Center, New York, NY<br />

Background: Pre-clinical studies report a critical role <strong>of</strong> c-KIT mutations in<br />

mucosal, acral, and solar melanomas. <strong>Clinical</strong> trials <strong>of</strong> KIT-directed therapy<br />

with imatinib and sunitinib demonstrate activity in these subtypes. Unlike<br />

other TKIs, dasatinib inhibitory activity is seen in melanoma cell lines with<br />

the most common KIT mutation at exon 11L576P. E2607 tests the<br />

efficacy <strong>of</strong> dasatinib in treatment-naïve or previously treated patients (pts)<br />

with these unresectable subtypes. Methods: Pts receive dasatinib 70 mg PO<br />

twice daily for this two-stage phase II trial. The primary objective is<br />

response rate (RR: RECIST). Stage I was open to KIT mutated (KIT�) and<br />

wild-type (KIT-) tumors with these subtypes (n�56). Depending upon the<br />

interim analysis, stage II would pre-select for KIT�. If the overall response<br />

rate were � 5%, the study would be terminated. Secondary objectives<br />

include progression-free survival (PFS), overall survival (OS), safety, and<br />

KIT mutation status. Results: Between May 2009-Dec 2010, 57 pts have<br />

been accrued to stage I. Of 54 (2 no therapy, 1 ineligible), 26 have mucosal<br />

(48%), 13 acral (24%), and 15 solar melanomas (28%). As <strong>of</strong> Jan 2012,<br />

the median f/u is 15 months (mo, range: 4-24 mo). Best responses (n�52)<br />

are 1 complete (CR: 2%), 3 partial (PR: 6%), 13 stable disease (25%), 29<br />

progression (56%), and 6 unevaluable (11%). 51/52 have progressed<br />

(median PFS: 1.9 mo, 95% CI: 1.5-2.8) and 40/54 died (median OS: 7.4<br />

mo, 5.7-10.8). KIT status has been assessed in 42 pts: 39 KIT- (93%) and<br />

3 KIT� (7%). PR is seen in 1/39 KIT-. All KIT- have died: median PFS (1.8<br />

mo, 95% CI: 1.4-2.9) and OS (6.8 mo, 95% CI: 5.2-10.8). Of the 3 KIT�<br />

(1 acral, 2 mucosal), 2 have died (PFS 1.4-2.5 mo, OS: 1.4-10.5 mo), and<br />

1 refused f/u (OS: 13.3� mo). The pt with a CR is alive at 14.4� mo<br />

(unknown KIT status). Toxicities include 1 grade IV (elevated lipase) and<br />

grade III dyspnea (n�7), nausea (n�6), and vomiting (n�3). Conclusions:<br />

Further evaluation <strong>of</strong> dasatinib should be pursued in pts selected for KIT�<br />

and subtype, given the lack <strong>of</strong> promising results in Kit-(�5% RR). The<br />

current trial is revised to enroll only KIT� acral, mucosal, and vulvovaginal<br />

melanomas and available via CTSU.<br />

Melanoma/Skin Cancers<br />

545s<br />

8521 Poster Discussion Session (Board #10), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

SWOG S0826: A phase II trial <strong>of</strong> SCH 727965 (NSC 747135) in patients<br />

with stage IV melanoma. Presenting Author: Christopher D. Lao, University<br />

<strong>of</strong> Michigan, Ann Arbor, MI<br />

Background: Cyclin-dependent kinases (cdks) function to regulate cell cycle<br />

control and agents that can target cdks in malignant progression remain<br />

viable therapeutic strategies. Selective inhibition <strong>of</strong> cdk2, in particular,<br />

may be <strong>of</strong> therapeutic value in a subset <strong>of</strong> patients with melanoma.<br />

Methods: 60 patients with metastatic melanoma <strong>of</strong> cutaneous or mucosal<br />

origin were planned to be recruited to a multicenter, single-arm phase II<br />

trial <strong>of</strong> the cdk inhibitor, SCH 727965 (NSC747135). Patients were<br />

potentially eligible if they had 0-1 previous treatments, PS <strong>of</strong> 0-1, and<br />

adequate organ function. Ocular melanoma patients and patients with a<br />

history <strong>of</strong> brain metastases were excluded. SCH 727965 50 mg IV every 3<br />

weeks was given until progression with disease assessment occurring every<br />

2 cycles. Co-primary endpoints were 1-year overall survival (OS) and<br />

6-month progression free survival (PFS). Results: 72 patients were enrolled<br />

from July 1, 2009 to November 1, 2010 at 24 institutions. 68% <strong>of</strong><br />

patients had M1c disease and 43% had LDH elevation. 19% had prior<br />

therapy for metastatic disease. 28 patients (39%) experienced Grade 4<br />

adverse events, including 20 cases <strong>of</strong> neutropenia, one case each <strong>of</strong><br />

cardiac ischemia/infarction, cardiac troponin I elevation, dehydration,<br />

abdominal pain, leukopenia, muscle weakness, headache, syncope, and<br />

anterior ischemic optic neuropathy. 65 patients are currently evaluable for<br />

response. The response rate was 0/65 (95% C.I. (0-6%)). Stable disease<br />

was observed in 22%. The estimated median PFS was 1.5 months (95%<br />

CI: 1.4 – 1.5); 6-month PFS was 11% (5-20%). Median OS was 8 months<br />

(95% CI: 5-11 months); 1-year OS was 36% (95% CI: 24-48%). The null<br />

hypothesis <strong>of</strong> 1-year overall survival�25% was rejected (p�0.04) but<br />

6-month PFS�11% was not (p�0.8). Data Analysis will be updated when<br />

missing data are received. Correlative studies <strong>of</strong> Rb phosphorylation and<br />

cyclin expression will be pursued. Conclusions: SCH 727965 appears to be<br />

reasonably well tolerated although grade 4 events were relatively common,<br />

particularly near the time <strong>of</strong> infusion. There were no responses but few<br />

patients had prolonged disease stabilization that may have resulted in<br />

improvement in the 1-year OS rate.<br />

8523 Poster Discussion Session (Board #12), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

A Cancer Research UK two-stage multicenter phase II study <strong>of</strong> imatinib in<br />

the treatment <strong>of</strong> patients with c-kit positive metastatic uveal melanoma<br />

(ITEM). Presenting Author: Paul D. Nathan, Mount Vernon Cancer Centre,<br />

Middlesex, United Kingdom<br />

Background: The median overall survival (OS) for metastatic uveal melanoma<br />

is less than 6 months with a median progression free survival(PFS) <strong>of</strong><br />

3 months. No systemic or regional therapy has shown a survival advantage<br />

over best supportive care. Despite pre-clinical evidence suggesting antitumour<br />

activity for the KIT tyrosine kinase inhibitor imatinib, there are<br />

several negative phase II trials in unselected patients. Our primary aim was<br />

to test the efficacy <strong>of</strong> imatinib in patients with prospectively-tested c-kit<br />

immunopositive metastatic uveal melanoma. Secondary aims included<br />

assessment <strong>of</strong> toxicity and patient recruitment. Methods: A phase II UK<br />

multicentre single-arm, two-stage Gehan design recruited 25 evaluable<br />

patients receiving imatinib 400mg OD until progression/unacceptable<br />

toxicity. Primary efficacy outcome was PFS at 3 months. Secondary<br />

outcomes were OS, overall PFS, disease response (RECIST) and toxicity.<br />

Prospective sample collection for putative biomarkers was included.<br />

Results: After 16.6 months 37 patients were screened, with 25 were<br />

registered and included in final efficacy analyses. The sample included PS<br />

0-1 patients with a median age <strong>of</strong> 63 yrs and a median 9.3 months from<br />

diagnosis <strong>of</strong> metastatic disease. 82% had high LDH levels (�460IU/L),<br />

65% had no previous treatment for metastatic disease and 8% did not have<br />

liver involvement by metastasis. Preliminary final results indicate the<br />

estimated proportion <strong>of</strong> patients progression free at 3 months is 0.24 (95%<br />

CI, 0.09 to 0.45). Median PFS and OS were 12.0 weeks (95% CI,11.6 to<br />

14.3) and 29.6 weeks (95% CI, 19.3 to 61.0) respectively. Two patients<br />

had confirmed PR with response duration <strong>of</strong> 93 and 112 weeks, respectively,<br />

despite the absence <strong>of</strong> mutations in exons 11, 13, and 17 <strong>of</strong> the<br />

c-KIT gene. Conclusions: The trial successfully recruited to target in this<br />

rare disease area failing to convincingly show improved PFS in a selected<br />

cohort <strong>of</strong> c-kit immunopositive patients. Both patients with PR experienced<br />

long periods <strong>of</strong> disease control. Response was not dependent upon the<br />

presence <strong>of</strong> activating mutations in KIT. Further translational studies are<br />

ongoing to determine putative biomarkers <strong>of</strong> response.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


546s Melanoma/Skin Cancers<br />

8524 Poster Discussion Session (Board #13), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

A phase II single arm study <strong>of</strong> pazopanib and paclitaxel as first-line<br />

treatment for unresectable stage III and stage IV melanoma: Interim<br />

analysis. Presenting Author: John P. Fruehauf, UCI Comprehensive Cancer<br />

Center, Orange, CA<br />

Background: Metastatic melanoma lacks effective therapy. Pazopanib is a<br />

small-molecule inhibitor <strong>of</strong> VEGFR-1,2,3, PDGFR-B and c-KIT that has<br />

antiangiogenic activity in renal cell cancer as well as inhibition <strong>of</strong><br />

melanoma tumor xenografts. We designed a phase II single arm, open label<br />

clinical trial evaluating pazopanib in combination with metronomic paclitaxel<br />

as first line therapy for subjects with unresectable stage III and stage<br />

IV melanoma. Methods: This protocol utilizes a Simon 2-stage Minimax<br />

design, with a planned interim analysis to confirm �3 responders to move<br />

to the second stage. To date, 20 eligible patients have been enrolled with<br />

17 evaluable for response. All subjects were treatment naïve and received<br />

paclitaxel at 80mg/m2 weekly for three weeks in a 4 week cycle and<br />

pazopanib at 800mg as a continuous daily oral dose. The primary endpoint<br />

is 6 month progression free survival. Exploratory endpoints include biomarker<br />

analysis that may be associated with treatment outcomes (serum<br />

VEGF, soluble VEGF R2, serum HIF, serum TSP1 and BRAF mutation<br />

status). An additional exploratory endpoint includes the in vitro activity <strong>of</strong><br />

pazopanib and paclitaxel on patient biopsy material co-cultured with<br />

vascular endothelial cells. RECIST criteria were used to define treatment<br />

response. Results: For the 17 evaluable patients treated to date the<br />

following results were seen: 1 CR, 6 PR’s, 8 SD’s and 2 PD’s. The overall<br />

RR (CR�PR) was 40%. Total disease control rate was 80% (PR�SD). The<br />

most common AEs/lab abnormalities were nausea (71%), hypertension<br />

(57%), fatigue (57%) and vomiting (43%). Grade 3-4 AEs included<br />

hypertension (28%), transaminitis (21%) and neutropenia (14%). One<br />

patient discontinued for grade 4 transaminitis which subsequently resolved<br />

completely. Dose reductions were required for pazopanib in 5 patients and<br />

for paclitaxel in one patient. Conclusions: Planned interim analysis <strong>of</strong> this<br />

phase II study demonstrated that pazopanib in combination with paclitaxel<br />

was well tolerated and resulted in a 40% response rate, indicating that this<br />

combination is <strong>of</strong> further interest. Accrual will continue to reach a goal <strong>of</strong><br />

60 patients.<br />

8526 Poster Discussion Session (Board #15), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Targeting hyperactivation <strong>of</strong> the AKT survival pathway to overcome therapy<br />

resistance <strong>of</strong> melanoma brain metastases. Presenting Author: Friedegund<br />

Elke Meier, University <strong>of</strong> Tuebingen, Tuebingen, Germany<br />

Background: Brain metastases are a major contributor to mortality in stage<br />

IV melanoma patients. In melanoma, the RAF-MEK-ERK (MAPK) and<br />

PI3K-AKT-mTOR (AKT) signaling pathways play a major role in tumor<br />

progression and therapy resistance. On the basis <strong>of</strong> significant improvement<br />

in overall survival, the BRAF inhibitor vemurafenib recently gained<br />

FDA approval for the treatment <strong>of</strong> patients with metastatic BRAFV600E<br />

mutated melanoma. However, ongoing clinical studies suggest short-lived<br />

responses to BRAF inhibitors in melanoma brain metastases. Methods: We<br />

observed in several melanoma patients that chemotherapeutics or BRAF<br />

inhibitors yielded a significant regression <strong>of</strong> extracerebral metastases while<br />

brain metastases progressed or newly occurred. We therefore aimed at<br />

identifying factors that may contribute to treatment resistance <strong>of</strong> brain<br />

metastases. Results: Immunohistochemistry <strong>of</strong> matched brain and extracerebral<br />

melanoma metastases demonstrated an identical pattern <strong>of</strong> ERK,<br />

p-ERK and AKT staining but a different pattern <strong>of</strong> p-AKT and PTEN<br />

staining with hyperactivation <strong>of</strong> AKT and loss <strong>of</strong> PTEN expression in brain<br />

metastases. Mutation analysis revealed no difference in BRAF, NRAS or<br />

KIT mutation status in matched brain and extracerebral metastases. By<br />

contrast, in monolayer culture expression <strong>of</strong> ERK, p-ERK, AKT, p-AKT and<br />

PTEN was identical in cell lines derived from brain and extracerebral<br />

metastases. Furthermore, melanoma cells stimulated by astrocyteconditioned<br />

medium showed hyperactivation <strong>of</strong> AKT compared to melanoma<br />

cells stimulated by fibroblast-conditioned medium. When inhibiting<br />

the MAPK and AKT signaling pathways at different levels in cells freshly<br />

isolated from melanoma brain metastases, growth inhibition and apoptosis<br />

induction was most pronounced with novel PI3K inhibitors such as<br />

BKM120 and BEZ235. Conclusions: These data suggest that 1) hyperactivation<br />

<strong>of</strong> the AKT survival pathway is brain environment-induced and<br />

relevant for the survival <strong>of</strong> melanoma cells in the brain parenchyma and<br />

that 2) inhibition <strong>of</strong> AKT signaling may be a suitable strategy to enhance<br />

and/or prolong the antitumor effect <strong>of</strong> chemotherapeutics or BRAF inhibitors<br />

in melanoma brain metastases.<br />

8525 Poster Discussion Session (Board #14), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Phase II trial <strong>of</strong> RO4929097 Notch gamma-secretase inhibitor in metastatic<br />

melanoma: SWOG S0933. Presenting Author: Kim Allyson Margolin,<br />

University <strong>of</strong> Washington, Seattle, WA<br />

Background: The Notch pathway regulates expression <strong>of</strong> genes for cell<br />

cycle, tissue-specific differentiation, and vasculogenesis. Notch target<br />

genes affect melanomagenesis, and Notch levels can influence stemlike<br />

versus differentiated tumor cells. Gamma-secretase, which activates intracellular<br />

Notch, can be inhibited to kill melanoma cells. We designed this<br />

trial to test RO4929097 in pts with melanoma and its effects on T<br />

lymphocytes and tumor gene expression. Methods: To assess 6-month<br />

progression-free survival (PFS) and 1-year overall survival (OS) in advanced,<br />

untreated melanoma patients (pts), a 2-stage accrual design was<br />

used. Correlative studies: markers <strong>of</strong> Notch pathway activation in archived<br />

or fresh tumor and T cell functional assays pre-treatment (Rx) and at week<br />

3. Rx dose was 20 mg orally on 3 consecutive days, weekly. Results: 33 pts<br />

were Rx’d in stage 1 (median age 61 [range 32-85]; 70% male; 42%<br />

elevated LDH; 30% unknown primary; 24% bone mets; 36% liver; 55%<br />

lung; 55% lymph node, skin or s<strong>of</strong>t tissue). The clinical outcomes did not<br />

meet criteria for stage 2 accrual. One pt had a confirmed PR <strong>of</strong> 7 months’<br />

(mo) duration. The median PFS was 1.4 mo, [95% confidence interval, c.i.<br />

1.3-2.7], the 6-mo PFS was 11% [95% c.i 3%-33%], and the 1-year OS<br />

was 45% [95% c.i. 23%-90%]. Treatment was well-tolerated with no grade<br />

(gr) 4-5 tox. The most common gr 2 drug tox were fatigue and nausea in 4<br />

patients (12%) each, and only 4 <strong>of</strong> 7 gr 3 tox were considered drug-related<br />

(1 increased ALT, 1 QTc prolongation, 1 bradycardia, 1 lymphopenia). Preand<br />

week 3 on-Rx peripheral T cell samples assayed for IL-2 (23 pts) and<br />

IFN-� (22 pts) secretion to Staphylococcal enterotoxin A showed no<br />

significant change, in contrast to in vitro gamma-secretase inhibitors which<br />

blocked T cell activation. Pre- and on-Rx tumor biopsies in one pt showed<br />

no decrease in the Notch target Hey1. Conclusions: RO4929097 at this<br />

dose and schedule has limited activity in molecularly-unselected pts with<br />

melanoma. Lack <strong>of</strong> effect on T cell function and tumor Hey1 expression<br />

suggests that sustained target inhibition might not have been achieved.<br />

Supported in part by PHS Cooperative Agreements, NCI, DHHS CA32102<br />

and CA38926. <strong>Clinical</strong>Trials.gov identifier: NCT01120275.<br />

8527 Poster Discussion Session (Board #16), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

CNS metastases as a site <strong>of</strong> progression on SWOG intergroup study S0008:<br />

A phase III trial <strong>of</strong> high-dose interferon alpha-2b versus cisplatin, vinblastine,<br />

DTIC plus IL-2 (BCT) versus high-dose interferon (HDI) in patients<br />

with high-risk melanoma. Presenting Author: Wolfram E. Samlowski,<br />

Comprehensive Cancer Centers <strong>of</strong> Nevada, Las Vegas, NV<br />

Background: Central nervous system (CNS) metastases (mets) are common<br />

in stage IV melanoma patients (pts). However, the incidence <strong>of</strong> CNS mets in<br />

pts with high-risk regional melanoma (HRM; stages IIIA-N2a thru IIIC N3)<br />

is not well described. A recent large prospective S0008 trial provided an<br />

opportunity to evaluate the contribution <strong>of</strong> CNS mets to treatment failure<br />

and survival. Methods: All pts had HRM treated with wide excision and full<br />

regional lymphadenectomy. Pts were then randomized to receive treatment<br />

with either BCT or HDI. All eligible pts were included in the analysis.<br />

Relapse/progression in the CNS (PCNS) was retrospectively identified only<br />

if clearly documented in case report forms. The cumulative incidence <strong>of</strong><br />

PCNS in the presence <strong>of</strong> the competing hazard <strong>of</strong> death was estimated and<br />

potential risk factors were explored using the methods <strong>of</strong> Fine and Gray.<br />

Survival from PCNS was measured from date <strong>of</strong> PCNS to date <strong>of</strong> death.<br />

Results: 402 patients were evaluated (BCT: 200, HDI: 202), with median<br />

follow (if alive) <strong>of</strong> 6 years. The site <strong>of</strong> progression was identified in 162 (78<br />

%) <strong>of</strong> 208 pts relapsing on study. Clearly documented PCNS occurred in<br />

53/402 pts (13%). PCNS as a component <strong>of</strong> initial relapse/progression<br />

occurred in 34 patients (8%) and an additional 19 patients (5%) had<br />

delayed PCNS following initial systemic relapse. Most PCNS (85%)<br />

occurred within 3 years <strong>of</strong> initial surgery. Differences between arms were<br />

not significant (22 on BCT, and 31 on HDI)(p�0.21). Lymph node<br />

macromets demonstrated a strong correlation with development <strong>of</strong> PCNS<br />

(p�0.01). Neither primary tumor ulceration nor head and neck primary site<br />

were significant risk factors. Survival from diagnosis <strong>of</strong> brain mets was short<br />

(median 6 mo BCS, 5 mo HDI, p�0.93). Conclusions: Although the S0008<br />

trial was not specifically designed to evaluate PCNS, a retrospective<br />

analysis identified a high CNS failure rate (at least 13%) in HRM pts,<br />

including 8% as the initial site <strong>of</strong> relapse. Further studies are needed to<br />

evaluate if screening for CNS mets in high-risk pts is useful and whether<br />

early treatment improves survival.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


8528 Poster Discussion Session (Board #17), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Association <strong>of</strong> high T-cell immune infiltrate and low hemorrhage in<br />

melanoma brain metastases (MBMs) with prolonged survival. Presenting<br />

Author: Michal Tadeusz Krauze, University <strong>of</strong> Pittsburgh Melanoma Program,<br />

UPCI, Pittsburgh, PA<br />

Background: Despite the poor prognosis <strong>of</strong> patients (pts) with MBM several<br />

pts have prolonged survival. We hypothesized that the heterogeneity <strong>of</strong><br />

BrMM is determined by differences in melanoma biology and its brain<br />

microenvironment. Methods: We identified pts who have undergone craniotomy<br />

for MBMs. Evaluation entailed complete clinical information, acquisition<br />

<strong>of</strong> archived melanoma brain metastases, histopathologic analysis <strong>of</strong><br />

hematoxylin and eosin-stained sections (n�101), whole genome expression<br />

pr<strong>of</strong>iling (WGEP, Illumina DASL) in 29 archived tissues. Results were<br />

validated by immunohistochemistry (IHC) or in situ hybridization (ISH).<br />

Results: In univariate analysis (log-rank) factors significantly associated<br />

with prolonged survival were high immune infiltrate (HII) plus low hemorrhage<br />

(hazard ratio, HR, 2.71, p�0.001), present melanin (1.67, p�0.03),<br />

and recursive partitioning analysis (RPA) class 1 (0.37, p�0.0001). No<br />

association between HII and use <strong>of</strong> immunotherapy prior to craniotomy was<br />

noted. Only RPA class 1 (p�0.029) and HII plus low hemorrhage<br />

(p�0.002) remained significant in Cox proportional hazards model analysis.<br />

Gene set analysis <strong>of</strong> WGEP data confirmed that Encarta pathways<br />

related with T-cell activation and differentiation were significantly associated<br />

with prolonged survival whereas Y branching <strong>of</strong> actin filaments,<br />

presenilin action in Notch and Wnt signaling, G-protein signaling through<br />

tubby protein, lissencephaly gene in neuronal migration and development<br />

are among the gene categories associated with worse survival. IHC and ISH<br />

analysis <strong>of</strong> tumor sections for various markers (n�40) showed that high<br />

peritumoral CD3� (3.31, p�0.009), high peritumoral CD4� (4.41,<br />

p�0.014), and high peritumoral CD8� (3.02, p�0.030) are associated<br />

with prolonged survival whereas neither CD14� nor FoxP3� infiltrate, nor<br />

high melanoma expression <strong>of</strong> antigen presentation molecules are associated<br />

with survival. Conclusions: Our study is the first to document that high<br />

peritumoral T-cell infiltrates are associated with prolonged survival. High<br />

tumor hemorrhage, an adverse prognostic sign, reflects aggressive melanoma<br />

cells that migrate and invade in the brain.<br />

8530 Poster Discussion Session (Board #19), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

<strong>Clinical</strong> activity and safety <strong>of</strong> combination therapy with temsirolimus and<br />

bevacizumab for advanced melanoma: Phase II trial with correlative<br />

studies. Presenting Author: Craig L. Slingluff, University <strong>of</strong> Virginia,<br />

Charlottesville, VA<br />

Background: A CTEP-sponsored phase II trial was performed to evaluate<br />

safety and clinical activity <strong>of</strong> combination therapy with CCI-779 (temsirolimus)<br />

and bevacizumab in patients with advanced melanoma. Correlative<br />

studies assessed mTOR signaling in tumor biopsies and evidence <strong>of</strong><br />

induced immunologic dysfunction systemically. Methods: 17 patients with<br />

stage III or IV melanoma were enrolled and treated with temsirolimus (25<br />

mg IV weekly) and bevacizumab (10 mg/kg IV every 14d, starting d.8) for<br />

up to 13 months. <strong>Clinical</strong> response was determined by RECIST criteria.<br />

Adverse events were assessed (CTCAE v3.0). Blood was collected d.1, 2,<br />

and 23 (to assess immune function), and tumor biopsies were obtained (to<br />

assess protein kinase activity and melanoma cell proliferation). Results:<br />

Treatment-related grade 3 or 4 adverse events occurred in 5 and 1 patients,<br />

respectively; 1 patient developed reversible leukoencephalopathy. In 16<br />

patients evaluable for clinical response, best overall response was a partial<br />

response (PR) in 3 patients (19%), stable disease at 8 weeks (SD) in 9<br />

patients (56%), and progressive disease in 4 patients. Thus, disease<br />

control rate (DCR � PR � SD) was 75%. Ten <strong>of</strong> the patients had BRAF<br />

wild-type (BRAFwt ) melanomas: these accounted for the 3 PRs (30%), and<br />

a DCR <strong>of</strong> 100%. Maximal response duration has exceeded 3 years for a<br />

BRAFwt patient. mTOR signaling was inhibited in melanoma metastases,<br />

based on decreased phospho-S6 kinase after 24h temsirolimus. Ki67� melanoma cells in tumor biopsies decreased significantly by day 23 (p �<br />

0.007, F-test), most notably in clinical responders. There was no significant<br />

alteration <strong>of</strong> T cell and NK function with combination treatment, by<br />

ELIspot and cytotoxicity assays. Conclusions: Combination therapy with<br />

temsirolimus and bevacizumab is well-tolerated in patients with advanced<br />

melanoma and has intriguing clinical activity. The most notable responses<br />

were in patients with BRAFwt tumors, a population with no accepted<br />

effective targeted therapy. Decreases in Ki67� melanoma cells may be<br />

associated with clinical response. The lack <strong>of</strong> immunologic dysfunction<br />

supports future combination with immune therapies.<br />

Melanoma/Skin Cancers<br />

547s<br />

8529 Poster Discussion Session (Board #18), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

The NIBIT-M1 trial: Activity <strong>of</strong> ipilimumab plus fotemustine in patients<br />

with melanoma and brain metastases. Presenting Author: Michele Maio,<br />

Medical Oncology and Immunotherapy, University Hospital <strong>of</strong> Siena,<br />

Siena, Italy<br />

Background: Patients (pts) with metastatic melanoma (MM) <strong>of</strong>ten develop<br />

treatment-resistant brain metastases (mets). Treatment includes fotemustine<br />

(FTM), which crosses the blood-brain barrier. Ipilimumab (ipi) has<br />

shown activity in pts with MM and asymptomatic brain mets (Heller et al.<br />

ASCO 2011; abs 8581). In the phase II NIBIT-M1 trial, MM pts with<br />

asymptomatic brain mets were eligible for treatment with ipi plus FTM.<br />

Here, data from this pt subset are reported. Methods: Eligible pts received<br />

induction therapy with ipi 10 mg/kg every 3 wks (Q3W) x4 and FTM 100<br />

mg/m2 weekly for 3 wks, followed by ipi Q12W from Week (W) 24 and FTM<br />

Q3W from W9. The primary objective was the immune-related (ir) disease<br />

control rate (irDCR: complete/partial response [CR/PR] or stable disease<br />

[SD] using the ir response criteria). Secondary objectives included ir<br />

objective response rate (ORR) and progression-free survival (PFS); overall<br />

survival (OS), and safety. Tumor assessments were performed Q8W from<br />

W12 to W36 and Q12W thereafter. Results: Among 86 enrolled pts, 20 had<br />

brain mets. Of these, 7 had prior whole brain radiotherapy (n�4) or<br />

radiosurgery (n�3). As <strong>of</strong> December 2011, the irDCR was 50% (10/20;<br />

95% CI, 27.2–72.8%) with an irORR <strong>of</strong> 40% (95% CI, 19.1–63.9%: 2<br />

CRs and 6 PRs). Pts with irDC also had stability/reduction (n�5) or<br />

disappearance (n�5) <strong>of</strong> brain mets. Among pts with progressive disease, all<br />

but one had progression in the brain. With median follow-up <strong>of</strong> 8.3 months<br />

(range: 0.4–16.9), median irPFS was 4.6 months (95% CI, 0.7–12.3).<br />

The 1-year OS rate was 52.9% (95% CI, 26.6–79.2); median OS was not<br />

reached. Induction with ipi and FTM was completed by 55% and 85% pts,<br />

respectively. Grade 3/4 drug-related adverse events (AEs) occurred in 60%<br />

pts; most commonly myelotoxicity (50%), increased ALT/AST (5%) and<br />

gastrointestinal (5%). AEs were generally manageable and reversible per<br />

protocol guidance. CNS AEs <strong>of</strong> any grade (i.e., haemorrhage, headache and<br />

seizure) occurred in 25% pts (grade 3/4 in 2 pts) and were attributed to<br />

disease progression. Conclusions: The combination <strong>of</strong> ipi plus FTM is active<br />

and safe in pts with MM and brain mets, regardless <strong>of</strong> prior treatment, and<br />

will be further explored in the phase III NIBIT-M2 trial.<br />

8531 Poster Discussion Session (Board #20), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Activity <strong>of</strong> cabozantinib (XL184) in metastatic melanoma: Results from a<br />

phase II randomized discontinuation trial (RDT). Presenting Author:<br />

Michael S. Gordon, Pinnacle Oncology Hematology, Scottsdale, AZ<br />

Background: MET and VEGF signaling are implicated in angiogenesis,<br />

invasion, and metastasis. Cabozantinib (cabo) is an oral, potent inhibitor <strong>of</strong><br />

MET and VEGFR2. A RDT evaluated activity and safety in 9 tumor types.<br />

Here we report on the metastatic melanoma cohort, including the ocular<br />

subtype. Methods: Eligible patients (pts) were required to have progressive<br />

measurable disease per RECIST. Pts received cabo at 100 mg qd over a 12<br />

wk Lead-in stage. Tumor response (mRECIST) was assessed q6 wks.<br />

Treatment � wk 12 was based on response: pts with PR continued<br />

open-label cabo, pts with SD were randomized to cabo vs placebo, and pts<br />

with PD discontinued. Primary endpoint in the randomized phase was<br />

progression free survival (PFS). Results: Enrollment to this cohort is<br />

complete (n � 77); all pts are unblinded. Baseline characteristics: median<br />

age 66 years; melanoma subtype: cutaneous/mucosal 70% and ocular<br />

30%; known BRAF mutation 32%; LDH � 1.1 x upper limit normal 35%;<br />

bone metastases 19%; median prior lines <strong>of</strong> therapy 1 (range 0-5). Median<br />

follow-up was 2.8 months (range 0.3 - 25). 35 pts (45%) completed the<br />

open-label Lead-in stage with 25 pts randomized to continue cabo (n�12)<br />

or to placebo (n�13). Median PFS from randomization was 5.7 months for<br />

cabo vs. 3 months for placebo (HR�0.3, p �0.055). Median PFS from<br />

Study Day 1 was 4.4 months. The estimate <strong>of</strong> PFS at month 6 (PFS6) is<br />

44%. Evidence <strong>of</strong> objective tumor regression was observed in 39/65 pts<br />

(60%) with � 1 post-baseline tumor assessment including 11/23 pts<br />

(48%) with ocular melanoma. Two bone scan evaluable pts demonstrated<br />

partial resolution <strong>of</strong> bone lesions at wk 6 accompanied by pain relief. Most<br />

common Grade 3/4 AEs were fatigue (14%), HTN (9%), constipation (4%),<br />

and diarrhea (3%); one related Grade 5 AE <strong>of</strong> diverticular perforation and<br />

peritonitis reported during Lead-in stage. Conclusions: Cabo demonstrates<br />

activity in metastatic melanoma pts, regardless <strong>of</strong> subtypes or BRAF<br />

mutation status, with improvement in PFS relative to placebo, and high<br />

rates <strong>of</strong> PFS6 and objective tumor regression. The safety pr<strong>of</strong>ile <strong>of</strong> cabo was<br />

comparable to that <strong>of</strong> other VEGFR TKIs.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


548s Melanoma/Skin Cancers<br />

8532 Poster Discussion Session (Board #21), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Randomized phase III trial <strong>of</strong> intravenous (IV) versus hepatic intra-arterial<br />

(HIA) fotemustine in patients with liver metastases from uveal melanoma:<br />

Final results <strong>of</strong> the EORTC 18021 study. Presenting Author: Serge Leyvraz,<br />

Centre Pluridisciplinaire d’Oncologie, Lausanne, Switzerland<br />

Background: HIA fotemustine has shown promising results in Phase II<br />

studies that led to the EORTC randomized phase III trial (18021) in<br />

unpretreated patients (pts) with liver metastases from uveal melanoma.<br />

Methods: The treatment consisted in an induction cycle <strong>of</strong> either HIA<br />

(fotemustine 100 mg/m² over 4 hours, day 1, 8, 15, 22) vs IV control arm<br />

(fotemustine 100 mg/m² over 1 hour, day 1, 8, 15). After a 5-week break,<br />

maintenance cycles were given every 3 weeks. Randomization was stratified<br />

by PS (0 vs 1), LDH (normal vs abnormal) and center. Main endpoint<br />

was overall survival (OS). Required accrual per protocol was set to 262 pts,<br />

with final analysis planned after 220 deaths (hazard ratio (HR) �0.67,<br />

power�85%, 1-sided ��2.5%). Due to poor accrual an interim analysis<br />

was done after 134 deaths, in order to test futility (power�79%). Results:<br />

Between Feb-2005- Feb-2011, 171 pts were randomized (HIA: 86, IV:<br />

85). Characteristics: PS 1: 20%, abnormal LDH: 42%, male: 50%, median<br />

age: 59 y.; balanced between arms. In the HIA arm 20 (23%) pts never<br />

started treatment mainly due to catheter problems and 2 pts in the IV arm.<br />

In those who started the treatment, leucopenia grade 3-4 was 18% and<br />

thrombopenia grade 3-4: 21% in the HIA arm compared to 32% and 42%<br />

in the IV arm. Non-hematological grade 3-4 toxicities were minimal (GI<br />

toxicity, catheter complications). In May 2011, as the OS HR�1.097 was<br />

� critical value 0.87, the IDMC recommended stopping accrual for futility.<br />

The final results from Jan-2012 are presented in the table below.<br />

Treatment comparison adjusted by PS and LDH provided similar results.<br />

Conclusions: Even if HIA fotemustine administration could not start in 23%<br />

<strong>of</strong> pts, it led to a higher ORR and longer PFS compared to IV administration.<br />

HIA did not translate into an improvement in OS.<br />

All pts<br />

(N�171)<br />

Treatment started<br />

(N�149)<br />

Endpoint HIA (N�86) IV (N�85) HIA (N�66) IV (N�83)<br />

Response<br />

CR�PR�ORR, N (%) 9 (10.5) 2 (2.4) 9 (13.6) 2 (2.4)<br />

PFS<br />

Events, N 84 85 64 83<br />

Median (months) 4.5 3.7 5.4 3.7<br />

HR (95% CI), p value 0.62 (0.45 , 0.84), 0.002 0.53 (0.38 , 0.75), 0.0002<br />

OS<br />

Deaths, N 79 76 59 74<br />

Median (months) 14.6 13.0 14.6 13.7<br />

HR (95% CI), p value 1.09 (0.79 , 1.50), 0.59 1.00 (0.71 , 1.42), 0.98<br />

8534 Poster Discussion Session (Board #23), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Long-term cure after complete resection and adjuvant immunotherapy for<br />

distant melanoma metastases. Presenting Author: Donald L. Morton,<br />

Division <strong>of</strong> Surgical Oncology, John Wayne Cancer Institute, St. John’s<br />

Health Center, Santa Monica, CA<br />

Background: In phase II trials, postoperative therapy with Canvaxin allogeneic<br />

melanoma cell vaccine plus Bacillus Calmette-Guerin (BCG) improved<br />

the survival <strong>of</strong> patients with stage IV melanoma. A multicenter, phase III<br />

placebo-controlled study was undertaken to investigate the vaccine’s<br />

efficacy. Methods: After complete resection <strong>of</strong> melanoma involving up to 5<br />

distant sites, patients were randomized to treatment with BCG plus<br />

Canvaxin (BCG-Canvaxin) or BCG plus placebo (BCG-placebo). The primary<br />

endpoint was overall survival (OS); secondary endpoints were disease-free<br />

survival (DFS) and skin test responsiveness to the study agent. Results:<br />

Between May 1998 and April 2005, 496 patients were randomized. In<br />

April 2005, entry to the study was terminated due to low probability <strong>of</strong><br />

demonstrating treatment differences. However, 256 patients from sites<br />

enrolled in a follow-up study were monitored until March 2010. Median OS<br />

and 5-year and 10-year rates <strong>of</strong> OS were 39.1 months, 43.3% and 33.3%,<br />

respectively, in the BCG-placebo group, versus 34.9 months, 42.5% and<br />

36.4%, respectively, in the BCG-Canvaxin group (hazard ratio, 1.053;<br />

95% confidence interval, 0.81 to 1.36; p�0.6964). Median DFS, 5-year<br />

DFS, and 10-year DFS were 7.6 months, 23.8% and 21.7%, respectively,<br />

for the BCG-placebo group, versus 8.5 months, 30.0%, and 30.0%,<br />

respectively, for the BCG-Canvaxin group (hazard ratio, 0.882; 95%<br />

confidence interval, 0.708 to 1.097; p�0.2595). Positive skin test results<br />

correlated with improved survival. Conclusions: BCG-Canvaxin was not<br />

superior to BCG-placebo, but the highly favorable long-term survival for<br />

combined groups indicates that complete metastasectomy should be<br />

considered as initial therapy for patients with resectable stage IV melanoma<br />

(<strong>Clinical</strong>Trials.gov identifier: NCT00052156).<br />

8533 Poster Discussion Session (Board #22), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Neoadjuvant ipilimumab in locally/regionally advanced melanoma: <strong>Clinical</strong><br />

outcome and immune monitoring. Presenting Author: Ahmad A. Tarhini,<br />

University <strong>of</strong> Pittsburgh Cancer Institute, Pittsburgh, PA<br />

Background: Neoadjuvant ipilimumab (ipi) for stage IIIB-C melanoma may<br />

improve the clinical outcomes and provide access to pre/post ipi blood and<br />

tumor to gain insight into host effector and suppressor immune response<br />

mechanisms. Methods: Patients were treated with ipi (10 mg/kg IV q3weeks<br />

x 4doses total) bracketing definitive surgery. Tissue samples were obtained<br />

at baseline and at definitive surgery (week � 6) and serum/PBMC collected<br />

at baseline, 6 weeks, then at 3, 6, 9, 12 months and/or progression. Flow<br />

cytometry was used to monitor the host effector and suppressor immune<br />

response in blood and evaluable tumor. Results: Thirty pts (21 male, 9<br />

female), age 40-87 were enrolled (25 cutaneous primary, 1 unknown, 4<br />

mucosal). Six had AJCC stage IIIB (N2b, N2c) and 24 IIIC (N3) melanoma.<br />

Ninety-three cycles have been delivered (median 4). Worst toxicities<br />

included grade 3 diarrhea/colitis (5 patients; 17%), hepatic enzyme<br />

elevations (2; 7%), rash (2; 7%), lipase (1; 3%), all manageable. Median<br />

follow up is 14 months: among 29 evaluable pts 15 (52%) continue<br />

disease free. Median PFS is 15.5 months, 95% CI � (8.1,-). The<br />

probability <strong>of</strong> 6 and 12 month PFS is 82.4% (95% CI�0.63, 0.92) and<br />

53% (95% CI�0.31, 0.70) respectively. Peripherally, a significant increase<br />

in frequency <strong>of</strong> circulating T-regs (CD4�CD25hi� Foxp3�;p�0.02<br />

CD4�CD25hi�CD39�; p�0.001) from baseline to 6 weeks was observed.<br />

Greater increases in T-regs were associated with improved PFS<br />

(p�0.045; HR�0.54). Significant decreases in circulating MDSCs, were<br />

observed in monocytic HLA-DR� /low/CD14� MDSC subtype (p�0.0001).<br />

Spontaneous in vivo cross presentation was observed resulting in Th1 CD4<br />

and CD8 antigen specific T-cell immunity (gp-100, MART-1, NY-ESO-1<br />

peptides) with increase in frequency after ipi. Activated TIL in tumor<br />

increased after ipi (CD3�/CD4�/CD69�;p�0.06 and CD3�/CD8�/<br />

CD69�) with significant induction/potentiation <strong>of</strong> T-cell memory (CD8�/<br />

CD45RO�/TNF-��;p�0.03). Conclusions: Neoadjuvant ipi exhibits<br />

promising clinical activity and significantly modulates the host effector and<br />

suppressor immune response. Full analysis <strong>of</strong> this completed trial and its<br />

correlates will be presented. Support: BMS, P30CA047904 and<br />

P50CA121973.<br />

8535 Poster Discussion Session (Board #24), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Sensitivity rate <strong>of</strong> ultrasound (US)-guided fine-needle aspiration cytology<br />

(FNAC) using the Berlin morphology criteria for lymph node metastases to<br />

reduce the need for surgical sentinel node (SN) staging in melanoma.<br />

Presenting Author: Christiane A. Voit, Charité, University Medicine Berlin,<br />

Berlin, Germany<br />

Background: US-guided-FNAC prior to surgical SN staging is emerging as a<br />

possible cost-effective addition to the staging <strong>of</strong> melanoma patients (pts).<br />

Formerly, sensitivity (sens) rates <strong>of</strong> lymph node US in melanoma were<br />

disappointing (20–40%). The introduction <strong>of</strong> the Berlin Morphology<br />

Criteria has significantly improved sens rates for US-FNAC (J Clin Oncol<br />

2010;28(5):847-52). The aim <strong>of</strong> the current study was to report on 1000<br />

patients the sens, specificity (spec), positive (PPV) and negative (NPV)<br />

predictive value rates <strong>of</strong> US-FNAC from our prospective database with<br />

prolonged follow-up. Methods: Since 2001, �1000 stage I/IIconsecutive<br />

melanoma pts have undergone US-FNAC prior to SN. All patients underwent<br />

lymphoscintigraphy. Peripheral Perfusion (PP), Loss <strong>of</strong> Central<br />

Echoes (LCE), Balloon Shaped (BS) were the Berlin Morphology Criteria<br />

which were registered. FNAC was performed in case <strong>of</strong> presence <strong>of</strong> any <strong>of</strong><br />

these factors. SN tumor burden was measured according to the Rotterdam<br />

Criteria. All patients underwent SN or LND in case <strong>of</strong> positive FNAC.<br />

Results: Mean/median Breslow thickness was 2.56 / 1.57 mm (0.2 – 44<br />

mm).Mean/median follow-up was 39 / 32 months (0 – 115). Ulceration was<br />

present in 24 %. SN positivity rates were 20 % (202 / 1000). Sens was 51<br />

%. Spec, PPV and NPV were 99%, 91% and 89%. Sensitivity was highest<br />

for T4 tumors (77%). PP, LCE, BS had sens <strong>of</strong> 69%, 24%, 25%. SN tumor<br />

burden � 1 mm in largest diameter according to the Rotterdam Criteria was<br />

identified by US-FNAC in 86%. Threshold for positive FNAC was 0.4 mm in<br />

maximum diameter. Conclusions: The new criterion <strong>of</strong> Periferal Perfusion is<br />

<strong>of</strong> key importance to achieve the high sensitivity <strong>of</strong> US-FNAC according to<br />

the Berlin Morphology Criteria (J Clin Oncol 2010; 28:847-852) to identify<br />

lymph node metastases. Especially for T4 patients and in patients with<br />

advanced SN tumor burden it can reduce significantly the need for surgical<br />

SN staging. The EORTC Melanoma Group will launch the prospective<br />

validation study, USE FNAC, in 2012.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


8536 Poster Discussion Session (Board #25), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Impact <strong>of</strong> comorbidities on overall survival <strong>of</strong> high-risk and advanced<br />

melanoma. Presenting Author: Prashanth Peddi, University <strong>of</strong> Texas M. D.<br />

Anderson Cancer Center, Houston, TX<br />

Background: Comorbidities have been shown to adversely affect survival<br />

among patients with cancer. Currently, the most important prognostic<br />

factor in patients with melanoma is AJCC stage. However, little is known<br />

regarding the impact <strong>of</strong> comorbidities on melanoma prognosis. The aim <strong>of</strong><br />

our study was to determine the impact <strong>of</strong> the severity <strong>of</strong> concurrent<br />

comorbidities on the overall survival <strong>of</strong> patients with high-risk and advanced<br />

melanoma (defined as AJCC stages IIc, III and IV). Methods: We<br />

conducted a retrospective cohort study <strong>of</strong> eligible adult melanoma patients<br />

available in the MelCore prospective database at MD Anderson Cancer<br />

Center (MDACC) from 01-2003 to 12-2006 who were diagnosed and<br />

completed staging within 3 months <strong>of</strong> presentation to MDACC. Patients<br />

with ocular melanoma were excluded. Demographic, AJCC staging, and<br />

survival data were collected. The Adult Comorbidity Evaluation-27 (ACE-<br />

27) was utilized to collect comorbidity information and grade its severity. A<br />

Cox proportional hazards model was used. Results: Of 444 patients that met<br />

enrollment criteria, 176 (39.6%) had grade 0 (no comorbidities), 222<br />

(50%) had grade 1 or 2 (mild or moderate comorbidities) and 46 (10.4%)<br />

had grade 3 (severe comorbidities). The median age <strong>of</strong> the entire cohort was<br />

56.4 years (19.7-98.9), 141 (32%) were female, and 406 (91.4%) were<br />

white. The median overall survival after presentation to MDACC was 5.0<br />

years for the entire cohort. Adjusted hazard ratios are shown in the table.<br />

Comorbidity and AJCC Stage were significantly associated with survival.<br />

Age, gender and race did not have a significant impact on overall survival.<br />

Conclusions: In our study, the presence <strong>of</strong> comorbidities at presentation<br />

were independent predictors <strong>of</strong> decreased survival, even when adjusted for<br />

AJCC stage. Our findings suggest that comorbidity should be incorporated<br />

into prognostic models and therapeutic decision-making for patients with<br />

high-risk or advanced melanoma.<br />

Multivariate Cox model (adjusted for age, sex, gender and stage).<br />

Grade Hazard ratio 95% CI p value<br />

ACE-27 0<br />

1or2<br />

3<br />

AJCC stage IIC<br />

III<br />

IV<br />

1.0<br />

1.5<br />

1.7<br />

1.0<br />

0.7<br />

3.2<br />

----<br />

1.1-2.0<br />

1.1-2.7<br />

---<br />

0.4-1.2<br />

2.0-5.2<br />

---<br />

0.01<br />

0.02<br />

---<br />

0.27<br />

�0.001<br />

8538 General Poster Session (Board #31D), Sun, 8:00 AM-12:00 PM<br />

A prospective study investigating the impact <strong>of</strong> definitive chemoradiation in<br />

locoregionally advanced squamous cell carcinoma <strong>of</strong> the skin. Presenting<br />

Author: Michelle K. Nottage, Royal Brisbane Hospital, Brisbane, Australia<br />

Background: Our state, Queensland, Australia, has the highest rate <strong>of</strong><br />

cutaneous squamous cell cancer (SCC) in the world. Spread to regional<br />

lymph nodes or more distant sites occurs in 5-10%. A proportion <strong>of</strong><br />

patients can not undergo surgical resection but complete response rates<br />

with radiotherapy alone are low. This led to the hypothesis that combined<br />

chemoradiation (CRT) may be <strong>of</strong> benefit. We decided to document the<br />

outcomes <strong>of</strong> concurrent chemoradiation by means <strong>of</strong> a prospective trial.<br />

Methods: This was a single arm, phase II study with planned sample size 30<br />

patients. The primary endpoint was complete response rate (CRR), estimate<br />

60%. Patients with locally/regionally advanced (non-metastatic)<br />

cutaneous SCC deemed unresectable or unsuitable for surgery by consensus<br />

<strong>of</strong> the multidisciplinary Head and Neck Cancer Clinic, with measurable<br />

disease, aged over 18, performance status 0-2, received definitive radiotherapy<br />

(XRT) (70Gy in 35#) and concurrent weekly platinum based<br />

chemotherapy (CT) (cisplatin 40mg/m2 or carboplatin AUC 2). Results: 14<br />

patients were enrolled (Feb 2008-June 2011), median age 66 (48-84),<br />

64% ECOG PS 0, 64% stage IV, 57% nodal disease only. Cisplatin/<br />

carboplatin was administered in 64%/36% respectively. 42% received all<br />

planned CT while 58% had 1 or 2 weeks omitted. 2 patients had dose<br />

reductions. XRT was completed as planned in 93%. The CRR was 57%<br />

(8/14) at analysis in December 2011 (median follow-up 13.5m). 2 further<br />

patients with partial response (PR) achieved CR after undergoing salvage<br />

surgery. Six (43%) patients had a PR; 4(29%) did not receive surgery and<br />

later progressed. Median overall survival was not reached, with 3 year<br />

survival 54%. The most frequent toxicities were dermatitis, mucositis,<br />

thrombocytopenia, nausea, anaemia, dysphagia. 28% had grade 3/4<br />

toxicity, mainly cytopenias, infection, dehydration and nausea. Conclusions:<br />

This is the only prospective series <strong>of</strong> CRT for cutaneous squamous cell<br />

cancer. A high complete response rate was documented in patients with<br />

loco-regionally advanced disease and multiple co-morbidities, with acceptable<br />

toxicity, making this a reasonable alternative for patients unable to<br />

undergo surgery.<br />

Melanoma/Skin Cancers<br />

8537 General Poster Session (Board #31C), Sun, 8:00 AM-12:00 PM<br />

Suppression <strong>of</strong> apoptosis by BRAF inhibitors through <strong>of</strong>f-target inhibition<br />

<strong>of</strong> JNK signaling. Presenting Author: Kenneth Yee Tsai, University <strong>of</strong> Texas<br />

M. D. Anderson Cancer Center, Houston, TX<br />

Background: The advent <strong>of</strong> targeted therapy has revolutionized the treatment<br />

<strong>of</strong> cancer. The mutant BRAFV600E protein is found in over 50% <strong>of</strong><br />

melanomas and thyroid carcinomas, resulting in elevated kinase activity,<br />

increased mitogen-activated protein kinase (MAPK) pathway signaling, and<br />

cell proliferation. Vemurafenib and PLX4720 were designed to selectively<br />

inhibit the BRAF kinase, and clinical trials <strong>of</strong> vemurafenib in metastatic<br />

melanoma have demonstrated a response rate <strong>of</strong> over 50% and an overall<br />

survival advantage over standard dacarbazine therapy. Approximately<br />

20-30% <strong>of</strong> individuals treated with vemurafenib develop cutaneous squamous<br />

cell carcinoma (cSCC) highlighting the importance <strong>of</strong> understanding<br />

toxicities associated with this drug. Paradoxical ERK activation in BRAF<br />

wild-type, RAS-mutant cells is thought to be the major mechanism by<br />

which this occurs, as evidenced by the presence <strong>of</strong> RAS mutations in 60%<br />

<strong>of</strong> such lesions. Methods: Using a combination <strong>of</strong> BRAF-wild-type cSCC cell<br />

lines, primary human keratinocytes, as well as a UV mouse model <strong>of</strong> cSCC<br />

and human cSCC samples, we identified novel effects <strong>of</strong> BRAFi on<br />

apoptosis. Results: Here we show an unexpected and novel effect <strong>of</strong><br />

vemurafenib and PLX4720 in suppressing apoptosis through the inhibition<br />

<strong>of</strong> multiple <strong>of</strong>f-target kinases. JNK signaling and apoptosis are suppressed<br />

in cSCC lesions arising in vemurafenib-treated patients as well as in<br />

irradiated mouse skin. This occurs independently <strong>of</strong> paradoxical ERK<br />

signaling and in the presence <strong>of</strong> MEK inhibitor. Treatment with PLX4720<br />

greatly accelerates the development <strong>of</strong> UV-induced cSCC in mice without<br />

Ras mutations. Kinome screening identified ZAK and MKK4 (MEK4 /<br />

MAP2K4) kinases as inhibited by vemurafenib, leading to suppression <strong>of</strong><br />

MKK4 and MKK7 (MAP2K7) phosphorylation. Knockdown <strong>of</strong> inhibited<br />

<strong>of</strong>f-target kinases recapitulates these anti-apoptotic effects <strong>of</strong> vemurafenib.<br />

Conclusions: Our results implicate suppression <strong>of</strong> JNK signaling,<br />

independent <strong>of</strong> ERK activation, as an additional, complementary mechanism<br />

<strong>of</strong> adverse effects <strong>of</strong> vemurafenib. This has broad implications for<br />

combination therapies with other modalities that induce apoptosis and for<br />

the long-term use <strong>of</strong> vemurafenib in the adjuvant setting.<br />

8539 General Poster Session (Board #31E), Sun, 8:00 AM-12:00 PM<br />

Outcomes <strong>of</strong> ipilimumab treatment-related adverse events in patients with<br />

metastatic melanoma (MM) who received systemic corticosteroids in a<br />

phase III trial. Presenting Author: Jean-Francois Baurain, Oncologie<br />

Médicale Cliniques Universitaires Saint-Luc, Brussels, Belgium<br />

Background: Ipilimumab (ipi) is a fully human monoclonal antibody that<br />

blocks cytotoxic T-lymphocyte antigen-4 to augment antitumor immune<br />

responses. In a phase III trial, ipi at 10 mg/kg plus dacarbazine (DTIC)<br />

improved overall survival in previously untreated patients (pts) with MM. In<br />

ipi studies, the most common drug-related adverse events (AEs) were<br />

immune-related, which were generally reversible using treatment guidelines<br />

involving prompt recognition, intervention (corticosteroids, and rarely,<br />

alternative immunosuppressive agents), and possible discontinuation <strong>of</strong><br />

therapy. The purpose <strong>of</strong> this analysis is to evaluate outcomes <strong>of</strong> ipi<br />

treatment-related AEs with use <strong>of</strong> systemic corticosteroids. Methods: AEs<br />

were reported from the first ipi dose up to 70 days after the last dose.<br />

Immune-mediated adverse reactions (imARs) were used to characterize<br />

inflammatory AEs in previously untreated pts with MM who received ipi plus<br />

DTIC in a phase III trial. This analysis includes the imAR categories <strong>of</strong><br />

hepatitis, dermatitis, enterocolitis, and endocrinopathies. Results: More<br />

than 70% <strong>of</strong> pts who received corticosteroids had complete resolution <strong>of</strong><br />

the imAR, ie, last reported Grade (Gr) <strong>of</strong> 0 (Table). In pts for whom a<br />

high-grade imAR had not completely resolved at the last report, �50% had<br />

improved to Gr 1. Conclusions: In the majority <strong>of</strong> pts treated with ipi at 10<br />

mg/kg plus DTIC who received corticosteroids for the management <strong>of</strong><br />

imARs, as recommended per treatment guidelines, the most common<br />

imARs had completely resolved or improved to Gr 1.<br />

ImAR<br />

Resolution or<br />

last Gr reported<br />

Outcomes <strong>of</strong> imARs, N (%)<br />

Ipi � DTIC,<br />

worst Gr�2<br />

549s<br />

Ipi � DTIC,<br />

worst Gr >2<br />

Hepatitis N who received steroids N�10 N�63<br />

Outcomes Gr 0 8 (80.0) 45 (71.4)<br />

Gr 1 2 (20.0) 12 (19.0)<br />

Gr 2 0 (0.0) 2 (3.2)<br />

Gr 3 0 (0.0) 3 (4.8)<br />

Gr 4 0 (0.0) 1 (1.6)<br />

Dermatitis N who received steroids N�29 N�6<br />

Outcomes Gr 0 24 (82.8) 2 (33.3)<br />

Gr 1 2 (6.9) 2 (33.3)<br />

Gr 2 3 (10.3) 1 (16.7)<br />

Gr 3 0 (0.0) 1 (16.7)<br />

Enterocolitis N who received steroids N�5 N�10<br />

Outcomes Gr 0 4 (80.0) 7 (70.0)<br />

Gr 1 0 (0.0) 2 (20.0)<br />

Gr 2 1 (20.0) 0 (0.0)<br />

Gr 3 0 (0.0) 1 (10.0)<br />

Endocrinopathies N who received steroids N�3 N�0<br />

Outcomes Gr 0 0 (0.0) 0 (0.0)<br />

Gr 1 0 (0.0) 0 (0.0)<br />

Gr 2 3 (100.0) 0 (0.0)<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


550s Melanoma/Skin Cancers<br />

8540 General Poster Session (Board #31F), Sun, 8:00 AM-12:00 PM<br />

Prognostic value <strong>of</strong> BRAFV600 mutations in melanoma patients after<br />

resection <strong>of</strong> metastatic lymph nodes. Presenting Author: Philippe Saiag,<br />

Hospital Ambroise Pare, APHP, University Versailles-SQY, Boulogne-<br />

Billancourt, France<br />

Background: Prognosis <strong>of</strong> AJCC stage III melanoma is heterogeneous.<br />

BRAF V600 mutations are frequent in melanomas. BRAFV600-targeted therapy<br />

has dramatic, but <strong>of</strong>ten transitory, efficacy in stage IV patients (pts). We<br />

aimed to determine for the first time the prognostic value <strong>of</strong> BRAFV600 mutations and other known prognostic criteria in stage III pts with<br />

sufficient nodal invasion. Methods: We searched all pts with cutaneous<br />

melanoma who had radical lymphadenectomy in our institution between<br />

1/1/00 and 15/6/10 and included those with a nodal deposit �2 mm.<br />

BRAFV600 mutations were detected by DNA sequencing and pyrosequencing<br />

in formalin-fixed nodal samples containing �60% melanoma cells.<br />

Samples were considered mutated when �15% <strong>of</strong> DNA was positive.<br />

Endpoints were overall survival (OS) and distant metastasis free survival<br />

(DMFS). 92 patients had to be included to demonstrate a doubling <strong>of</strong> OS in<br />

patients without (40 months (m)) and with BRAFV600 mutation (20 m).<br />

Log-rank test and multivariate Cox proportional hazards regression model<br />

were used. Results: 105 consecutive pts were included, with 72% prospectively<br />

followed-up pts. BRAFV600 mutations (E: 83%; K: 14% <strong>of</strong> pts) were<br />

detected in 40% <strong>of</strong> pts. Median follow-up was 19 m (range: 3-139). Death<br />

occurred in 83% and 60% <strong>of</strong> pts with and without BRAF mutations,<br />

respectively, with median OS <strong>of</strong> 1.4 and 2.8 years. Pts’ age, primary<br />

melanoma ulceration, number <strong>of</strong> invaded nodes, AJCC staging, and BRAF<br />

status influenced OS and DMFS in the univariate analysis. The multivariate<br />

analysis showed the major prognostic role <strong>of</strong> BRAF status and <strong>of</strong> the<br />

number <strong>of</strong> invaded nodes (table). Conclusions: Provided our findings are<br />

independently replicated, BRAFV600 status should be used to stage<br />

melanoma pts with nodal metastasis. Our results also help to plan adjuvant<br />

trials with BRAFV600-targeted therapy in such patients, for whom the low<br />

tumor load may induce longer efficacy <strong>of</strong> BRAF-targeted therapies.<br />

OS DMFS<br />

P HR[IC95%] P HR[IC95%]<br />

N invaded nodes (1,2-3,4) 0.005 2.2 [1.3-3.9] 0.005 2.4 [1.3-4.3)<br />

BRAF mutated (Y/N) 0.005 1.9 [1.2-3.1] 0.002 2·1 [1·3-3·4]<br />

Ulceration <strong>of</strong> primary (Y/N) NS - 0.04 1.7 [1.0-2.8]<br />

AJCC staging, extracapsular spread, macro/micrometastasis, Breslow index: NS.<br />

8542 General Poster Session (Board #31H), Sun, 8:00 AM-12:00 PM<br />

Role <strong>of</strong> cytokine-induced plasticity in melanoma on invasive, therapyresistant<br />

cells that express EMT-related genes and pathways. Presenting<br />

Author: Jonathan S. Cebon, Ludwig Institute for Cancer Research, Melbourne,<br />

Australia<br />

Background: The acquisition <strong>of</strong> new mutations can explain resistance to<br />

targeted therapies; however resistance also develops without demonstrable<br />

new mutations. To better understand potential mechanisms for treatment<br />

failure we studied subpopulations <strong>of</strong> chemo-resistant cells that can be<br />

identified in early passage cell lines and the cellular plasticity that yields<br />

these. Methods: Melanoma subpopulations were identified and sorted on<br />

the basis <strong>of</strong> functional assays. Gene expression was evaluated with Illumina<br />

HT12 arrays and qPCR to identify potential mechanisms and targets.<br />

Sorted cells were evaluated in vitro for the induction <strong>of</strong> phenotype<br />

switching and effects <strong>of</strong> target inhibition. Results: A subset <strong>of</strong> slow<br />

proliferating label-retaining cells (LRC) was identified using a membrane<br />

dye. Direct isolation <strong>of</strong> LRC from a pt showed these were not an in vitro<br />

artefact. LRC isolated from multiple cell lines comprised the majority <strong>of</strong><br />

cells that resisted cytotoxic drugs and could invade through an artificial<br />

extracellular membrane. Gene expression pr<strong>of</strong>iling identified a network <strong>of</strong><br />

over-expressed genes related to epithelial-to-mesenchymal transition (EMT)<br />

notable for the expression <strong>of</strong> Thrombospondin-1 (TSP-1), Transforming-<br />

Growth Factor, Beta Induced, (TGFBI) and other extracellular matrix<br />

molecules. Initial studies show that LRC shared gene expression characteristics<br />

with PLX4032-resistant cells. Although interrogating LRC by qPCR<br />

showed up-regulation <strong>of</strong> the putative melanoma stem cell marker ABCB5<br />

and the lysine specific demethylase Jarid1B, cell sorting and serial<br />

re-labelling experiments revealed a dynamic and interchangeable phenotype<br />

for these cells, challenging the hierarchical stem cell model for<br />

melanoma. The observed slow-cycling and chemo-resistant phenotype<br />

could be induced in vitro through TGF beta-mediated phenotype switching<br />

and cellular invasion was blocked using an antibody against TSP-1.<br />

Conclusions: Cytokine-induced plasticity in melanoma yields invasive,<br />

therapy-resistant cells which express EMT-related genes and pathways. We<br />

hypothesize that these cells are a source <strong>of</strong> treatment resistance in vivo and<br />

blocking their emergence may prevent treatment failure.<br />

8541 General Poster Session (Board #31G), Sun, 8:00 AM-12:00 PM<br />

Characterization <strong>of</strong> ipilimumab exposure-efficacy/safety response relationships<br />

in subjects with previously treated or untreated advanced melanoma.<br />

Presenting Author: Yan Feng, Bristol-Myers Squibb, Princeton, NJ<br />

Background: Ipilimumab (IPI) is a fully human monoclonal antibody<br />

directed against CTLA-4 that is being developed as a novel immunotherapeutic<br />

agent against melanoma and other solid tumors. The objective <strong>of</strong><br />

this analysis was to retrospectively characterize the exposure-response<br />

(E-R) relationships for efficacy (overall survival, OS) in subjects with<br />

previously untreated advanced melanoma (pu-MEL), and safety (immune<br />

related adverse-events, irAEs) in subjects with previously treated advanced<br />

melanoma (pt-MEL) or pu-MEL. Methods: The E-R analysis <strong>of</strong> OS included<br />

498 pu-MEL from a phase III study (CA184024) <strong>of</strong> IPI administered at 10<br />

mg/kg � dacarbazine (DTIC) or DTIC � placebo; and the E-R analyses <strong>of</strong><br />

irAE included 1036 subjects from a phase I study (CA184078), 4 phase<br />

IIstudies (CA184004/007/008/022) and CA184024 <strong>of</strong> IPI administered<br />

at doses <strong>of</strong> 0.3, 3 or 10 mg/kg. The relationship between steady-state IPI<br />

trough concentration (Cminss) and OS was described using a Cox proportional<br />

hazards model, and the relationships between Cminss and incidence<br />

<strong>of</strong> irAEs were described by ordinal logistic regression models (separate<br />

models for any irAE, as well as skin, liver and GI irAE). Results: The risk <strong>of</strong><br />

death decreased with increasing Cminss, and is higher for subjects with<br />

elevated baseline LDH level. The risk <strong>of</strong> death was higher for subjects with<br />

baseline ECOG status � 0 and was higher with increasing stage <strong>of</strong> disease<br />

(M1C�M1B�M1A�M0). The hazard ratio for OS corresponding to the<br />

median Cminss <strong>of</strong> 49.99 �g/mL was 0.745 relative to subjects in DTIC �<br />

placebo group. The probability <strong>of</strong> Grade 2� or Grade 3� GI, skin, liver and<br />

any irAEs also increases with Cminss. Prior anti-cancer therapy (including<br />

prior DTIC) or concomitant DTIC were the only significant covariates in the<br />

E-R irAE models. Conclusions: These model-based analyses suggest that<br />

higher Cminss <strong>of</strong> IPI appears to be associated with improved survival in<br />

pu-MEL (based on data from CA184024) and higher Cminss also appears<br />

to be associated with increased probability <strong>of</strong> irAEs in pu-MEL or pt-MEL.<br />

8543 General Poster Session (Board #32A), Sun, 8:00 AM-12:00 PM<br />

Gene expression pr<strong>of</strong>ile signature (DecisionDx-Melanoma) to predict visceral<br />

metastatic risk in patients with stage I and stage II cutaneous<br />

melanoma. Presenting Author: Navneet Dhillon, Emory University , Atlanta,<br />

GA<br />

Background: The current AJCC TNM staging system has poor specificity for<br />

predicting visceral metastatic risk in patients diagnosed with stage I or<br />

stage II cutaneous melanoma. We, therefore, developed a gene expression<br />

pr<strong>of</strong>ile signature (GEP) following in silico investigation <strong>of</strong> previously<br />

published microarray analyses. Methods: 60 formalin fixed paraffin embedded<br />

primary cutaneous melanoma samples from patients with stage I or II<br />

cutaneous melanoma with at least a follow up period <strong>of</strong> at least 6 years were<br />

macrodissected and analyzed blindly. RNA was isolated, converted to<br />

cDNA and RT-PCR was performed to assess the expression <strong>of</strong> the gene set.<br />

Expression data and biostatistical analysis was performed using GeNorm<br />

and JMP Genomics (SAS) Predictive modeling included Radial Basis<br />

Machine (RBM) and <strong>Part</strong>ition Tree Analysis (PTA) Metastasis-free survival<br />

(MFS) was assessed using Kaplan-Meier analysis. The following clinical<br />

data was retrieved from medical records: survival, metastases, types <strong>of</strong><br />

metastases. 20 out <strong>of</strong> 60 patients had developed visceral metastases in the<br />

follow up period. Results: GEP was developed following multiple analytical<br />

approaches.Two types <strong>of</strong> signatures emerged: Low risk (Class 1) and High<br />

risk (Class 2). Without optimizing for sensitivity, the analyses <strong>of</strong> the 60<br />

sample cohort by radial basis machine (RBM) resulted in 92% ROC (met.<br />

accuracy � 90%, non-met. accuracy � 85%), while partition tree analysis<br />

(PTA) yielded 99% ROC (met. accuracy � 100%, non-met. accuracy �<br />

95%). RBM classification showed 6-year MFS rates <strong>of</strong> 97% for Class 1 and<br />

19% for predicted Class 2 <strong>of</strong> metastasis (median MFS � NR and 5.6 yrs,<br />

resp., P�0.0001 Log-Rank respectively). PTA showed 6-year MFS rates <strong>of</strong><br />

100% for predicted Class 1 and 14% for Class 2 <strong>of</strong> metastasis (median<br />

MFS � NR and 5.4 yrs, resp., P�0.0001 Log-Rank respectively).<br />

Conclusions: This study shows that DecisionDx-Melanoma GEP signature<br />

can provide excellent accuracy in predicting metastatic risk in stage I and II<br />

cutaneous melanoma.To our knowledge, the GEP provides the most<br />

accurate predictor to date for development <strong>of</strong> visceral metastases in<br />

patients with Stage I and II cutaneous melanoma.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


8544 General Poster Session (Board #32B), Sun, 8:00 AM-12:00 PM<br />

Elucidating distinct tumorigenic pathways in nodular versus superficial<br />

spreading melanoma. Presenting Author: Joshua Andrew Farhadian, New<br />

York University School <strong>of</strong> Medicine, New York, NY<br />

Background: Superficial spreading melanoma (SSM) and nodular melanoma<br />

(NM), which account for 70% and 20% <strong>of</strong> all melanoma diagnoses<br />

respectively, are believed to represent sequential phases <strong>of</strong> linear progression<br />

from radial to vertical growth. Recent evidence from several groups<br />

including ours challenges this linear progression model and suggests that<br />

SSM and NM are biologically distinct. In this study, we focused on<br />

identifying tumorigenic pathways that are differentially activated in melanoma<br />

subtypes and that can be therapeutically exploited. Methods: We<br />

performed Protein Pathway Array on SSM/radial growth phase (RGP) and<br />

NM/vertical growth phase (VGP) primary melanoma cell lines to determine<br />

expression levels <strong>of</strong> 141 tumorigenic proteins/phospho-proteins. Differential<br />

protein expression was determined using the Pavlidis Template<br />

Matching algorithm in TIGR Multi Experiment Viewer. Differentially expressed<br />

proteins were validated in an expanded panel <strong>of</strong> RGP, VGP, and<br />

metastatic melanoma cell lines using western blot. Constitutively active,<br />

overexpressed proteins in VGP melanoma were down-regulated in cell lines<br />

using several small molecule inhibitors and the effects on proliferation and<br />

migration were assessed. Results: 17 (12%) proteins/phospho-proteins are<br />

significantly overexpressed in VGP versus RGP melanoma (p�0.05).<br />

Western blot confirmed the results <strong>of</strong> 5 proteins/phospho-proteins. Each <strong>of</strong><br />

those 5 was equally overexpressed in VGP and metastatic melanoma cell<br />

lines. Phospho-p90 ribosomal s6 kinase (RSK) was prioritized for functional<br />

studies based on its role in both the MAPK and AKT signaling<br />

cascades, two tumorigenic pathways <strong>of</strong>ten dysregulated in melanoma.<br />

Constitutive phosphorylation <strong>of</strong> p90 RSK in VGP, but not RGP melanoma<br />

was observed in response to serum starvation. Small molecule inhibition <strong>of</strong><br />

phospho-p90 RSK attenuated proliferation and migration (p�0.05) in VGP<br />

melanoma. Conclusions: Data demonstrate discrete tumorigenic pathways<br />

are activated in nodular versus superficial spreading melanoma and<br />

suggest that phospho-p90 RSK might be a viable target against nodular<br />

melanoma.<br />

8546 General Poster Session (Board #32D), Sun, 8:00 AM-12:00 PM<br />

BEVATEM: Phase II single-center study <strong>of</strong> bevacizumab in combination<br />

with temozolomide in patients (pts) with first-line metastatic uveal melanoma<br />

(MUM): First-step results. Presenting Author: Sophie Piperno-<br />

Neumann, Institut Curie, Paris, France<br />

Background: Overall survival (OS) <strong>of</strong> MUM pts remains poor. In our<br />

retrospective series <strong>of</strong> 470 MUM pts, median OS was 13 months, ranging<br />

from 3 to 12 and 21 months for best supportive care, systemic treatment<br />

and surgery groups respectively, stressing the lack <strong>of</strong> effective therapies in<br />

this rare cancer. Targeting angiogenesis seems to be promising in uveal<br />

melanoma. Intravitreal application <strong>of</strong> antiangiogenic agents is used to treat<br />

neovascular ocular diseases such as age-related macular degeneration or<br />

proliferative diabetic retinopathy. Bevacizumab suppressed in vitro growth<br />

and in vivo hepatic establishment <strong>of</strong> micrometastases in experimental<br />

uveal melanoma. Preclinical data suggest a potential clinical benefit <strong>of</strong> the<br />

combination <strong>of</strong> dacarbazine and bevacizumab. Methods: Phase II study,<br />

modified 2-step Fleming plan; with expected 6-month progression-free<br />

survival (PFS) rates <strong>of</strong> 15% with chemotherapy and 40% with BEVATEM,<br />

35 pts are to be recruited for a power <strong>of</strong> 94% (� and � risk 3 and 6%). After<br />

the first 17 pts, the study will be continued and another 18 pts will be<br />

enrolled in the second step if �3 evaluable pts show stable disease or<br />

response. Primary endpoint: 6-month PFS according to RECIST. Secondary<br />

objectives: response and survival rates, safety; liver perfusion CT for<br />

functional imaging <strong>of</strong> response; impact <strong>of</strong> VEGF-A gene polymorphisms on<br />

bevacizumab pharmacodynamics. Treatment schedule: Temozolomide<br />

150mg/m2 d1-d7 and d15-d21 oral route, Bevacizumab 10 mg/kg d8 d22<br />

IV infusion, 6 cycles (d1�d28) then Bevacizumab maintenance until<br />

toxicity or progression. Results: From May 2010 to January 2011, 17 pts<br />

were enrolled according to the first step <strong>of</strong> the study: 3/17 achieved disease<br />

control at 6 months, second step is on going with 26/35 pts already<br />

recruited. One patient with minor response in liver and lung metastases is<br />

under Bevacizumab maintenance for 12 months. Safety <strong>of</strong> BEVATEM is as<br />

good as expected. Liver perfusion CT study showed decrease in blood flow<br />

and blood volume before lesion size decrease in one stable patient.<br />

Conclusions: BEVATEM study in first line MUM pts is on going, showing<br />

promising results in the first step <strong>of</strong> 17 pts.<br />

Melanoma/Skin Cancers<br />

551s<br />

8545 General Poster Session (Board #32C), Sun, 8:00 AM-12:00 PM<br />

Serum IL-6 as a prognostic biomarker in patients with stage IIB-III<br />

melanoma. Presenting Author: Lise Hoejberg, Department <strong>of</strong> Oncology,<br />

Odense University Hospital, Odense, Denmark<br />

Background: Interleukin (IL)-6 is an immunomodulatory cytokine produced<br />

by cancer cells and different immune cells. The specific function <strong>of</strong> IL-6 in<br />

cancer is unknown. Serum IL-6 is elevated in patients with different types<br />

<strong>of</strong> cancer. Elevated serum concentration <strong>of</strong> IL-6 is related to short survival<br />

in patients with stage IV melanoma. Methods: IL-6 was measured by ELISA<br />

(R&D) in pretreatment serum samples from 435 patients with stage IIB-III<br />

melanoma (151 women and 284 men, median age 51 years, range 18-77).<br />

After surgery patients were treated in a randomized study (The Nordic IFN<br />

trial) with either adjuvant interferon for 1 or 2 years, or observation. Results:<br />

Median serum concentration <strong>of</strong> IL-6 after surgery and before treatment was<br />

1.8 ng/l, range 0.23-24 ng/l. Patients with serum IL-6 above the median<br />

had shorter overall survival (OS) compared to patients with serum IL-6<br />

below the median (p�0.004). Median OS was 4.5 years in patients with<br />

serum IL-6 above the median (95% confidence interval (CI): 3.05-7.60).<br />

In patients with serum IL-6 below the median, median OS was not reached<br />

at the last survival-update. Multivariate Cox analysis including serum IL-6,<br />

ulceration in primary melanoma, LDH, white blood cells, lymph node<br />

metastases and Breslow thickness <strong>of</strong> primary melanoma showed that only<br />

serum IL-6 (hazard ratio (HR) � 1.48, 95% confidence interval (CI) :<br />

1.13-1.94, p�0.004) was an independent prognostic factor for OS. In<br />

subgroup analysis <strong>of</strong> the control group and the two treatment groups, serum<br />

IL-6 demonstrated a negative prognostic impact in the control group (HR �<br />

1.62, 95% CI: 1.02-2.57, p�0.04) and in the group treated with 2 years<br />

interferon HR � 1.69, 95% CI: 1.06-2.7, p�0.03). In the group treated<br />

with 1 year interferon this negative impact was not significant (HR � 1.15,<br />

95% CI: 0.71-1.85, p�0.58). An interaction between IL-6 and treatment<br />

arm was not significant (p�0.45), suggesting that the negative prognostic<br />

effect <strong>of</strong> elevated pretreatment serum IL-6 is independent <strong>of</strong> treatment.<br />

Conclusions: Serum IL-6 above the median is an independent prognostic<br />

biomarker <strong>of</strong> short OS in patients operated for stage IIB-III melanoma.<br />

8547 General Poster Session (Board #32E), Sun, 8:00 AM-12:00 PM<br />

Phase II study <strong>of</strong> the anti-ganglioside GD3 mouse/human chimeric antibody<br />

KW2871 combined with high dose interferon-a2b in patients with<br />

metastatic melanoma. Presenting Author: Stergios J. Moschos, University<br />

<strong>of</strong> Pittsburgh Medical Center, Pittsburgh, PA<br />

Background: Immunotherapy has demonstrated notable effects in metastatic<br />

melanoma (MM) with durable responses achieved by high-dose IL-2<br />

and IFN�2b, leading to approval <strong>of</strong> these therapies for treatment <strong>of</strong><br />

melanoma. However, complete responses occur in only a minority <strong>of</strong><br />

patients. KW2871 is a chimeric monoclonal antibody (mAb) targeting the<br />

GD3 ganglioside with demonstrated antitumor activity and enhancement <strong>of</strong><br />

antibody-dependent cell-mediated cytotoxicity (ADCC) and complementdependent<br />

cytotoxicity (CDC). IFN�2b has potent immunoregulatory, antiproliferative,<br />

differentiation-inducing, pro-apoptotic, and anti-angiogenic<br />

properties against a variety <strong>of</strong> malignancies including melanoma. Combining<br />

high dose IFN�2b (HDI) � KW2871 was hypothesized to have<br />

synergistic anti-tumor activity due to (1) the ability <strong>of</strong> HDI to enhance<br />

KW2871 induced ADCC in vitro (Liu, Cancer Immun 2002); (2) improved<br />

mAb targeting due to increased GD3 expression and induced inflammatory<br />

cytokines (TNF-�, IL-4 and IFN-�) (Hoon, Cancer Res, 1991); (3) increased<br />

tumor-infiltrating immune cells (Kirkwood, Cancer 2002; Moschos,<br />

J Clin Oncol 2006). Methods: This is an open label, dose-escalation, phase<br />

II study <strong>of</strong> KW2871 plus HDI in patients with measurable MM. Primary<br />

objectives are progression-free survival (PFS) and safety. Secondary objectives<br />

include assessment for tumor response by RECIST, ADCC, CDC,<br />

pharmacokinetics, human antichimeric antibodies (HACA), tumor-infiltrating<br />

immune cells, biomarkers and OS. Patients with measurable disease by<br />

RECIST, stable brain metastases, and performance status ECOG 0 or 1 are<br />

eligible. Patients with severe comorbidities or autoimmune disease or prior<br />

exposure to anti-GD3 antibodies are excluded. Sequential enrollment to<br />

cohorts <strong>of</strong> KW2871 at 5, 10 , 20 mg/m2 IV every 2 week in combination<br />

with HDI 20 MU/m2 IV once daily x 5 Days for 4 weeks, then 10 MU/m2 SC<br />

three times weekly until disease progression. Results: To date, Cohort 1 (5<br />

mg/m2 KW2871) and Cohort 2 (10 mg/m2 KW2871) have been completed<br />

safely. Cohort 3 (20 mg/m2 KW2871) has enrolled <strong>of</strong> 18 <strong>of</strong> 27 planned<br />

patients. Conclusions: Will be presented at study completion.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


552s Melanoma/Skin Cancers<br />

8548 General Poster Session (Board #32F), Sun, 8:00 AM-12:00 PM<br />

Correlations <strong>of</strong> molecular alterations in clinical stage III cutaneous melanoma<br />

with clinical-pathological features and patients outcome. Presenting<br />

Author: Piotr Rutkowski, Maria Sklodowska-Curie Memorial Cancer Center<br />

and Institute <strong>of</strong> Oncology, Warsaw, Poland<br />

Background: To evaluate frequency and type <strong>of</strong> oncogenic BRAF/NRAS<br />

mutations in cutaneous melanoma with clinically detected nodal metastases<br />

(stage IIIB,C) in relation to clinicopathologic features and outcome.<br />

Methods: We analyzed 221 patients after therapeutic lymphadenectomy-<br />

LND (1995-2010) not treated with tyrosine kinase inhibitors and performed<br />

molecular characterization <strong>of</strong> nodal metastases in terms <strong>of</strong> BRAF/<br />

NRAS genes (analyzed by sequencing <strong>of</strong> respective coding sequences).<br />

Median follow-up time was 53 months. Results: BRAF mutations were<br />

detected in 139 (63%) cases (127–V600E, 8–V600K, 4-others), mutually<br />

exclusive NRAS mutations in 35(15.8%) cases (mainly Q61R and Q61K).<br />

BRAF mutation presence correlated with patients’ younger age(median 52<br />

vs 60 years for BRAF� vs. BRAF-, p�0.05), metastases in axillary basin<br />

(p�0.05) and less involved nodes (median 3 vs. 4; p�0.05). 5-year overall<br />

survival (OS) was 35% and 45% (calculated from date <strong>of</strong> LND and primary<br />

tumor excision, respectively); 5-year recurrence-free survival RFS (from<br />

LND) – 29%. We have not found correlation between mutational status and<br />

RFS or OS (calculated from date <strong>of</strong> LND and primary tumor excision) – for<br />

BRAF mutated-melanomas prognosis was the same as wild-type melanoma<br />

patients(p�0.26) with even trend for better OS for non-V600E mutants.<br />

Negative prognostic factors (in univariate and multivariate analysis) for OS<br />

and RFS were: male gender (p�0.01), metastatic lymph nodes�1<br />

(p�0.001), nodal metastases extracapsular extension (p�0.001). The<br />

interval from diagnosis <strong>of</strong> first-ever melanoma to regional nodal metastasis<br />

(median-10 months) was not significantly different between BRAF-mutant<br />

and BRAF wild-type patients (p�0.29). Conclusions: BRAF/NRAS mutational<br />

status is not prognostic marker in stage III melanoma patients with<br />

macroscopic nodal involvement, what may have implication for potential<br />

adjuvant therapy. BRAF status had no impact on disease-free interval from<br />

diagnosis <strong>of</strong> primary melanoma to nodal metastases. Our first-ever comprehensive<br />

molecular analysis <strong>of</strong> clinical stage III melanomas revealed that<br />

BRAF-mutants show characteristic clinicopathologic features.<br />

8550 General Poster Session (Board #32H), Sun, 8:00 AM-12:00 PM<br />

Early alterations <strong>of</strong> microRNA expression to predict and modulate melanoma<br />

metastasis. Presenting Author: Eva Hernando, New York University<br />

School <strong>of</strong> Medicine, New York, NY<br />

Background: Melanoma is curable for most patients whose tumors are<br />

surgically removed early in disease progression; however, many primary<br />

melanomas recur and progress to metastasis. <strong>Clinical</strong> staging is insufficient<br />

to robustly classify patients at highest risk <strong>of</strong> recurrence, and prognostic<br />

molecular biomarkers have not yet been identified. Methods: We performed<br />

miRNA pr<strong>of</strong>iling <strong>of</strong> 92 FFPE primary melanomas to discover metastasis<br />

relevant miRNAs and develop predictive models <strong>of</strong> recurrence, which were<br />

then validated in an independent cohort. We identified miRNAs differentially<br />

expressed between primary tumors that did and did not recur (3-year<br />

minimum follow-up) and between thick and thin lesions. We selected<br />

candidate miRs for screening in a fluorescence-based in vitro invasion<br />

assay, and prioritized a subset for in vivo testing. Potential downstream<br />

mediators <strong>of</strong> these miRNAs were selected by mRNA array analysis and<br />

tested in a secondary invasion screen. mRNA candidates that mimicked the<br />

miR’s invasion-suppressive effect were tested in 3’UTR reporter assays to<br />

confirm them as direct targets. Results: Using the discovery cohort we<br />

identified a 20 miRNA signature that can distinguish Stage I/II primary<br />

tumors (n�70) that did from those did not recur with 3-year minimum<br />

follow-up with an AUC�94%, 95% CI: (0.88, 0.99). Applying this model<br />

to predict risk for recurrence in the independent validation cohort yielded<br />

an AUC � 96%, 95% CI: (0.90, 1) in discriminating recurrent versus<br />

non-recurrent stages I/ II patients (n�45). From the discovery cohort, 40<br />

candidates were selected for invasion assay screening, <strong>of</strong> which 5 miRNAs<br />

robustly inhibit in vitro invasion in 5 melanoma cell lines. Three miRs<br />

(miR-382, miR-516b, and miR-7) strongly suppressed metastasis in a<br />

mouse model. Moreover, multiple mRNAs tested as potential mediators<br />

mimicked the invasion-suppressive effects <strong>of</strong> candidate miRs. Of those,<br />

NCAPG2 and CDC42 were identified as miR-516b targets, CTTN as a<br />

miR-382 target, and PIK3CD as a miR-7 target. These genes have been<br />

linked to metastasis in melanoma or other tumors. Conclusions: Our data<br />

demonstrate that aberrant expression <strong>of</strong> specific miRNAs at diagnosis is<br />

predictive <strong>of</strong> and functionally impacts melanoma progression.<br />

8549 General Poster Session (Board #32G), Sun, 8:00 AM-12:00 PM<br />

Ipilimumab in the treatment <strong>of</strong> uveal melanoma: The Memorial Sloan-<br />

Kettering Cancer Center experience. Presenting Author: Shaheer A. Khan,<br />

Memorial Sloan-Kettering Cancer Center, New York, NY<br />

Background: Ipilimumab (ipi) is an antibody that blocks cytotoxic Tlymphocyte<br />

antigen-4 (CTLA-4) and improves overall survival in patients<br />

(pts) with metastatic melanoma. Uveal melanoma (UM) is a rare and<br />

biologically unique disease subtype with no known effective systemic<br />

therapy. Ipi has proven efficacy in cutaneous melanoma (CM), but limited<br />

data exists regarding its activity in UM. We reviewed our single-institutional<br />

experience with ipi in advanced UM. Methods: After IRB approval, the<br />

MSKCC melanoma database was queried for patients with metastatic UM<br />

treated with ipi between 03/08-01/12. Radiographic response by RECIST<br />

and immune-related response criteria (irRC) was assessed by a single<br />

radiologist blinded to clinical outcomes. Immune-related adverse events<br />

(irAEs), survival and absolute lymphocyte count (ALC) were also evaluated.<br />

Results: 20 pts were identified: the median age was 61yrs (range 46-83),<br />

55% were male, 85% had liver metastases, 60% had elevated LDH, and<br />

pts reported a median <strong>of</strong> 1 prior therapies (range 0-5). Pts received a<br />

median <strong>of</strong> 4 doses (range 1-16) <strong>of</strong> ipi. Response rates (RR) by irRC at 12<br />

and 24 wks are listed below. Among pts with stable disease (SD) at 12 wks,<br />

the median time to progression was 30.6 wks (range 19.6-83), with one<br />

partial response (PR) occurring after 24 wks (overall RR 10%). Responses<br />

were observed in lung, liver and peritoneal metastases. Pts with an ALC �<br />

1.0 at 7 wks had a trend toward a higher clinical benefit (CB� CR � PR �<br />

SD) than pts with ALC � 1.0 (5/12 [42%] vs 0/5 [0%]; p� .09), consistent<br />

with prior studies in CM. To date, median survival for the group is 8.6 mos<br />

(95% CI, 3.5-NR), with two ongoing PRs (3� yrs and 24� wks). Reported<br />

irAEs include rash/pruritis (10/20), hepatitis (1/20), colitis (1/20), pancreatitis<br />

(1/20) and uveitis (1/20). Conclusions: Ipi has potential for benefit in<br />

pts with advanced UM; RR and irAE rates are similar to those observed in<br />

pts with advanced CM. Further evaluation <strong>of</strong> ipi in the treatment <strong>of</strong> UM,<br />

including identification <strong>of</strong> potential biomarkers <strong>of</strong> CB, is warranted.<br />

Immune-related response criteria 12 wks 24 wks<br />

Stable disease 7/20 (35%) 4/20 (20%)<br />

<strong>Part</strong>ial response 1/20 (5%) 1/20 (5%)<br />

Complete response 0/20 0/20<br />

<strong>Clinical</strong> benefit 8/20 (40%) 5/20 (25%)<br />

8551 General Poster Session (Board #33A), Sun, 8:00 AM-12:00 PM<br />

Targeted agents matched with tumor molecular aberrations: Review <strong>of</strong> 160<br />

patients with advanced melanoma treated in a phase I clinic. Presenting<br />

Author: Haby Adel Henary, University <strong>of</strong> Texas M. D. Anderson Cancer<br />

Center, and Department <strong>of</strong> Investigational Cancer Therapeutics, Houston,<br />

TX<br />

Background: Identification <strong>of</strong> activating mutations in melanoma has increased<br />

the number <strong>of</strong> novel targeted agents for this disease. Methods:<br />

Weretrospectively reviewed clinical outcomes <strong>of</strong> 160 consecutive metastatic<br />

melanoma patients (pts) treated in the Dept <strong>of</strong> Investigational Cancer<br />

Therapeutics (Phase I program) at M. D. Anderson since 2008, and<br />

compared their median progression free survival (PFS) to their first and last<br />

standard systemic therapy PFS. In addition, we compared those pts’<br />

outcomes tested for tumor molecular aberrations on a phase I trial with a<br />

matched targeted agent with those <strong>of</strong> pts who were treated without regard<br />

for their molecular pr<strong>of</strong>iles. Results: Of 160 pts treated on 35 different<br />

phase 1 clinical trials, 110 pts (69%) had � 1 molecular aberration. Of<br />

those pts who had adequate tissue for molecular analysis, 63% (85/134)<br />

pts had BRAF mutation, 20% (22/109) NRAS mutation, 20% (1/5) GNAQ<br />

mutation, 11% (1/9) P53 mutation, 2.5% (1/39) PIK3CA and 1.3% (1/76)<br />

had KIT mutation. 77 (48%) pts were treated on a phase I trial with a<br />

matched targeted agent and 83 (52%) pts were treated on a non-matched<br />

phase 1 trial. The overall response rate was 39% (complete response [CR],<br />

9%; partial response [PR], 30%) in the 77 pts treated with matched<br />

therapy and 9% (all PRs) in the 83 pts treated without matched therapy (P<br />

� 0.0018). 139 (87%) pts received at least one systemic therapy before<br />

referral to phase I, median PFS was longer on phase 1 therapy than on last<br />

line standard therapy prior to referral to phase 1 (4.2 vs. 2.8 months, P �<br />

0.002). Median PFS was greater for pts on matched vs. non-matched<br />

therapy (5.3 vs. 3.7 months, log rank P � 0.004). Also, median PFS was<br />

longer on phase 1 matched trial than on first standard treatment (5.3 vs.<br />

3.9 months, log rank P � 0.045).PFS did not differ between first standard<br />

and non-matched phase 1 study. Univariate analyses with the log rank test<br />

revealed that matched therapy (P � 0.004) was positively associated with<br />

longer PFS on phase I clinical trials. Conclusions: Matching melanoma pts<br />

with targeted drugsbased on specific molecular aberrations in the phase I<br />

setting can be associated with superior outcomes compared to prior<br />

standard systemic therapies.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


8552 General Poster Session (Board #33B), Sun, 8:00 AM-12:00 PM<br />

Positron emission tomography/computed tomography to improve staging<br />

and restaging in Merkel cell carcinoma. Presenting Author: Elena B.<br />

Hawryluk, Department <strong>of</strong> Dermatology, Harvard Medical School, Boston,<br />

MA<br />

Background: Merkel cell carcinoma (MCC) is a rare (~1,500 cases per year)<br />

and highly aggressive (33% mortality) cutaneous neuroendocrine carcinoma<br />

that occurs in older white patients on the UV-exposed skin <strong>of</strong> the<br />

head, neck, and extremities. As a patient’s stage at presentation is a strong<br />

predictor <strong>of</strong> survival, and there is a high propensity for locoregional<br />

recurrence and distant progression, imaging remains crucial for initial and<br />

subsequent management. There is, however, no consensus on the timing or<br />

method <strong>of</strong> imaging for MCC. Methods: We retrospectively reviewed 270<br />

2-fluoro-[ 18F]-deoxy-2-D-glucose (FDG) positron emission tomography/<br />

computed tomography (PET/CT) scans performed in 97 patients at the<br />

Dana-Farber/Brigham and Women’s Cancer Center from August 2003 to<br />

December 2010. Results: The mean SUVmax was 6.5 for primary tumors,<br />

6.4 for regional lymph nodes, 7.2 for distant metastases (all sites), 8.0 for<br />

bone/bone marrow metastases, and 9.4 for non-regional metastases in<br />

those patients with no identified primary. PET/CT imaging performed for<br />

initial management tended to upstage patients with more advanced disease<br />

(50% <strong>of</strong> stage IIIB patients). Metastases to bone/bone marrow (12<br />

patients, 38%) was the 2nd most common site <strong>of</strong> distant spread after<br />

non-regional lymph nodes (19 patients, 59%), followed by skin (8 patients,<br />

25%), liver (6 patients, 19%), lung/pleura (5 patients, 16%), adrenal (3<br />

patients, 9%), muscle (3 patients, 9%), pancreas (2 patients, 6%), and<br />

peritoneum (1 patient, 3%). In 10 <strong>of</strong> 12 patients, PET identified bone/bone<br />

marrow metastases that were not seen on CT imaging, which resulted in<br />

either upstaging or initiation <strong>of</strong> more targeted palliative therapy. Conclusions:<br />

Added value <strong>of</strong> PET over CT, such as in the detection <strong>of</strong> bone/bone marrow<br />

metastases, may lead to more accurate staging, and thus prognostication,<br />

as well as earlier detection <strong>of</strong> relapse and initiation <strong>of</strong> salvage treatment. Its<br />

use should be considered in the staging and restaging <strong>of</strong> MCC.<br />

8554 General Poster Session (Board #33D), Sun, 8:00 AM-12:00 PM<br />

Randomized, double-blind, and multicenter phase II trial <strong>of</strong> rh-endostatin<br />

plus dacarbazine versus dacarbazine alone as first-line therapy for the<br />

patients with advanced melanoma. Presenting Author: Jun Guo, Peking<br />

University Cancer Hospital and Institute, Beijing, China<br />

Background: rh-endostatin (Endostar, water-soluble, rES) is an inhibitor <strong>of</strong><br />

angiogenesis, which is effective as single agent or in combination with<br />

chemotherapy for non-small cell lung cancer in phase I/II clinical trials.<br />

This study (NCT00813449) was a randomized, double-blind and placebocontrolled<br />

phase II trial, aimed to observe the efficacy and safety <strong>of</strong> rES<br />

with dacarbazine (DTIC) as the 1st line therapy for patients (pts) with<br />

advanced melanoma. Methods: Untreated pts with ECOG 0/1 and unresectable<br />

stage IIIC or IV melanoma (confirmed by histopathology) were<br />

enrolled, and randomized (1:1) into Arm A (DTIC 250 mg/m2 d1-5 �<br />

placebo d1-14) or Arm B (DTIC 250 mg/m2 d1-5 � rES 7.5 mg/m2 d1-14). Treatment was continued in 21-day cycle until progression or intolerable<br />

toxicity occurs. Primary endpoints were progression free survival (PFS) and<br />

overall survival (OS). Secondary endpoints included objective response rate<br />

(ORR) and safety, evaluated every 2 cycles. Results: From Dec. 2008, 120<br />

pts were enrolled and 110 pts evaluable. 30.9% <strong>of</strong> them were in M1a,<br />

39.1% in M1b, and 29.1% in M1c. Mean treatment cycles were 3.2<br />

(range: 1-10) in Arm A and 4.0 (range: 1-12) in Arm B. Until the last<br />

follow-up in Nov. 2011, 27 pts were still alive. The median PFS was 1.5<br />

months (95% CI: 1.35-1.65) in Arm A and 5.0 months in Arm B (95% CI:<br />

2.45-7.55, P�0.004, log-rank test). The median OS was 7.0 months in<br />

Arm A (95% CI: 4.41-9.59) versus 16.0 months in Arm B (95% CI:<br />

10.46-21.54, P�0.003, Breslow test). 1-year survival rate was 22.2%<br />

versus 51.0%, and 2-year survival rate 8.9% vs.10.2%. No statistical<br />

significance was found for both ORR and toxicities. The most common<br />

toxicities were increased transaminase (28.9% in Arm A versus 56.1% in<br />

Arm B) and leucopenia (14.4% versus 13.4%). The incidence <strong>of</strong> grade 3-4<br />

toxicity (increased transaminase and thrombocytopenia) was only 1.7% in<br />

Arm B. Conclusions: rES plus DTIC may obviously improve mPFS and mOS<br />

versus DTIC alone as the 1st line therapy for advanced melanoma. That<br />

combining therapy was well tolerated and could be recommended as a new,<br />

safe and effective regimen for untreated pts with advanced melanoma.<br />

Melanoma/Skin Cancers<br />

553s<br />

8553 General Poster Session (Board #33C), Sun, 8:00 AM-12:00 PM<br />

Effect <strong>of</strong> the BH3 mimetic ABT-737 on human melanoma cells to<br />

apoptosis induced by selective BRAF inhibitors. Presenting Author: Peter<br />

Hersey, Melanoma Institute Austrailia, Sydney, Australia<br />

Background: Although the introduction <strong>of</strong> selective BRAF inhibitors has<br />

been a major advance in treatment <strong>of</strong> metastatic melanoma, approximately<br />

50% <strong>of</strong> patients have limited responses including stabilisation <strong>of</strong> disease or<br />

no response at all. The present study aims to identify a novel means <strong>of</strong><br />

overcoming resistance <strong>of</strong> melanoma to killing by BRAF inhibitors. Methods:<br />

We examined the influence <strong>of</strong> the BH3 mimetic ABT-737 on induction <strong>of</strong><br />

apoptosis by the selective BRAF inhibitor PLX4720 on a panel <strong>of</strong><br />

melanoma cells with BRAF V600E mutations with or without mutations <strong>of</strong><br />

CDK4 and BRAF wildtype cells with or without NRAS mutations. Included<br />

in the studies were cell lines established from 4 patients before and during<br />

treatment with selective BRAF inhibitors. Results: Cell lines with no or low<br />

sensitivity to PLX 4720 underwent synergistic increases and increased<br />

rates <strong>of</strong> apoptosis when combined with ABT-737. This degree <strong>of</strong> synergism<br />

was not seen in cell lines without BRAF V600E mutations. Apoptosis was<br />

mediated through the mitochondrial pathway and was due in part to<br />

upregulation <strong>of</strong> Bim as shown by inhibition <strong>of</strong> apoptosis following siRNA<br />

knockdown <strong>of</strong> Bim. Similar effects were seen in cell lines established from<br />

patients prior to treatment but not in lines from patients clinically resistant<br />

to the selective BRAF inhibitors. Conclusions: These results suggest that<br />

combination <strong>of</strong> selective BRAF inhibitors with ABT-737 or the oral form<br />

ABT-263 may increase the degree and rate <strong>of</strong> responses in previously<br />

untreated patients with V600E melanoma but not in those with acquired<br />

resistance to these agents.<br />

8555 General Poster Session (Board #33E), Sun, 8:00 AM-12:00 PM<br />

BRAF mutation as a pejorative marker in metastatic melanoma. Presenting<br />

Author: Sophie Brissy, Hopital Timone, Aix-Marseille University, Marseille,<br />

France<br />

Background: BRAF mutation in melanoma has been shown to be associated<br />

with a trend in favour <strong>of</strong> a spontaneous worse outcome after metastases in a<br />

series <strong>of</strong> 197 patients in Australia. Objective: To correlate BRAF status in<br />

metastatic melanoma with clinicopathologic features and outcome. Methods:<br />

In our department in France 182 patients with metastatic melanoma have<br />

been tested for BRAF mutation between September 2009 and September<br />

2011. Survival was assessed by log-rank test. Multivariate analysis was<br />

performed with Cox model. Results: From 182 patients, 88 (48.3%) were<br />

B-RAF mutant; 77 (87.5%) V600E, 4 (4.5%) V600K, and 7 (8%) other<br />

mutation subtypes. BRAF-mutant patients were younger than BRAF wildtype<br />

patients at diagnosis <strong>of</strong> primary melanoma (median age 52.3 vs 60.7<br />

years, respectively, p�0.003), and at diagnosis <strong>of</strong> distant metastasis<br />

(median age 53.6 vs 64.1 years respectively, p�0.002). 34 patients were<br />

treated by B-RAF inhibitors. There was no significant difference in other<br />

demographic features <strong>of</strong> patients with metastatic melanoma by mutation<br />

status. Features <strong>of</strong> the primary melanoma significantly associated with a<br />

BRAF mutation (p�0.05) were histopathologic subtype (SSM), high<br />

mitotic rate (�1/mm2), lower Breslow thickness (median Breslow: 2.2 vs<br />

3.5 mm for BRAF mutant and BRAF-wild-type patients respectively,<br />

p�0.016), truncal location and location on occasionally exposed at sun<br />

site.The interval from diagnosis <strong>of</strong> first ever melanoma to first distant<br />

metastasis was not significantly different in BRAF-mutant and wild-type<br />

patients. The median overall survival (OS) from diagnosis <strong>of</strong> primary<br />

melanoma was 6.5 years for BRAF wild-type patients. Median OS was not<br />

reached in BRAF-mutant patients treated (34 <strong>of</strong> 88) with a BRAF inhibitor,<br />

but also in those not treated (p�0.24, and p�0.06 for treated BRAFmutant<br />

vs BRAF wild-type). The overall survival from diagnosis <strong>of</strong> first<br />

distant metastasis was not significantly different (p�0.75). These results<br />

remained unchanged in a multivariate analysis. Conclusions: Our results<br />

confirm the characteristics <strong>of</strong> BRAF-mutant metastatic patients, and the<br />

efficacy <strong>of</strong> B-RAF inhibitors, but not that the presence <strong>of</strong> mutant-BRAF is<br />

per se a pejorative predictive marker.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


554s Melanoma/Skin Cancers<br />

8556 General Poster Session (Board #33F), Sun, 8:00 AM-12:00 PM<br />

Everolimus in combination with paclitaxel and carboplatin in patients with<br />

advanced melanoma: A phase II trial <strong>of</strong> the Sarah Cannon Research<br />

Institute (SCRI). Presenting Author: Ralph J. Hauke, Nebraska Cancer<br />

Specialists, Omaha, NE<br />

Background: The PI3k/AKT pathway is activated in most metastatic melanomas;<br />

mTOR is a critical component <strong>of</strong> this pathway. Everolimus, an mTOR<br />

inhibitor, has demonstrated single-agent activity in patients with advanced<br />

melanoma. We evaluated the efficacy and toxicity <strong>of</strong> everolimus in<br />

combination with paclitaxel/carboplatin in patients with advanced melanoma.<br />

Methods: Eligible patients had stage IV or unresectable stage III<br />

melanoma, unselected for braf status, previously untreated with chemotherapy<br />

or targeted agents. Previous immunotherapy was allowed. Additional<br />

eligibility criteria: ECOG PS 0 or 1; measurable disease; no active<br />

brain metastases; adequate bone marrow, kidney, and liver function;<br />

informed consent. All patients received paclitaxel 175mg/m2 , 1-3 hour IV<br />

infusion, and carboplatin AUC 6.0 IV on day 1 <strong>of</strong> each 21-day cycle.<br />

Everolimus 5mg PO was given daily. Patients were evaluated for response<br />

every 6 weeks; treatment continued until progression or undue toxicity.<br />

Median progression-free survival (PFS) for paclitaxel/carboplatin treatment<br />

is 4 months; we looked for a median PFS <strong>of</strong> 6 months with this novel<br />

combination. Results: Seventy patients were treated between 2/2010 and<br />

2/2011; median age 63, 90% had stage IV melanoma. 91% <strong>of</strong> patients<br />

received at least 2 cycles <strong>of</strong> therapy; median cycles received: 4 (range:<br />

1-25�). Twelve patients (17%) had partial responses; an additional 42<br />

patients (60%) had stable disease at first reevaluation. After a median 13<br />

months <strong>of</strong> followup, the median PFS for the entire group was 4 months<br />

(95% CI: 2.8 – 5.0 months); 96% had progressed during the first 12<br />

months. Median survival was 10 months (95% CI: 7.3 – 10.9 months).<br />

Toxicity was as previously described with these agents; neutropenia was the<br />

most common grade 3/4 toxicity (27%). Only 3 patients stopped treatment<br />

due to toxicity. Conclusions: The addition <strong>of</strong> everolimus to paclitaxel/<br />

carboplatin was feasible and well-tolerated; however, efficacy results were<br />

similar to those reported with paclitaxel/carboplatin alone. Further development<br />

<strong>of</strong> this combination regimen for treatment <strong>of</strong> metastatic melanoma is<br />

not recommended.<br />

8558 General Poster Session (Board #33H), Sun, 8:00 AM-12:00 PM<br />

Patterns <strong>of</strong> progression in patients (pts) with V600 BRAF-mutated melanoma<br />

metastatic to the brain treated with dabrafenib (GSK2118436).<br />

Presenting Author: Mary W. F. Azer, The Crown Princess Mary Cancer<br />

Centre Westmead, Sydney, Australia<br />

Background: Dabrafenib has shown efficacy in pts with previously untreated<br />

brain metastases (BM) but most will progress. We report the pattern <strong>of</strong><br />

disease progression (PD) in pts with either previously treated (surgery (Sx),<br />

SRS, WBRT) or untreated BM on dabrafenib. Methods: Clinicopathologic<br />

parameters were collected on 23 pts enrolled in the brain cohort <strong>of</strong> the<br />

BRF112680 phase I and BRF113929 phase II BM study <strong>of</strong> dabrafenib at<br />

Westmead Hospital between Sept 2009 and June 2011. Pts with RECIST<br />

PD but ongoing clinical benefit were allowed to continue dabrafenib.<br />

Results: 12 pts (52%) had previously untreated BM and 11 (48%) were<br />

previously treated with evidence <strong>of</strong> progression prior to dabrafenib. Median<br />

OS from study entry (8.4 mo, 95% CI 5.0-11.8), diagnosis <strong>of</strong> first BM<br />

(11.0 mo, 95% CI 9.1-13.0), and stage IV diagnosis (17.5 mo, 95% CI<br />

12.1-23.0) were not different between the two groups. Similarly, PFS did<br />

not differ between groups. Of the 19 pts who had RECIST PD at datacut, 13<br />

pts had intracranial (IC) PD with no pts progressing in new brain lesions<br />

alone (see table). At IC PD, 3/13 underwent SRS/Sx, 5/13 WBRT and 5/13<br />

had no salvage local therapy to BM. 8 pts continued dabrafenib beyond IC<br />

PD, median 36 days; range 28-273. All measures <strong>of</strong> baseline disease<br />

burden correlated with worse OS; elevated LDH (HR 1.003, 95% CI<br />

1.0-1.007, P�0.044), increased number (no.) metastatic sites (HR 1.32,<br />

95% CI 0.97-1.81, P�0.08), increased no. <strong>of</strong> IC lesions (HR 1.04, 95%CI<br />

1.00-1.09, P�0.04), increased no. extracranial (EC) lesions (HR 1.07,<br />

95%CI 1.02-1.12, P�0.01) and increased RECIST sum <strong>of</strong> diameters<br />

(SoD) (HR 1.012, 95% CI 1.003-1.021, P�0.007). High baseline SoD<br />

was the only factor that predicted worse RECIST response. Conclusions:<br />

<strong>Clinical</strong> benefit from dabrafenib does not appear to be influenced by prior<br />

local therapies to BM. There was no dominant pattern <strong>of</strong> progression in pts<br />

with BM on dabrafenib, but PD due to new lesions alone is rare. Pts with IC<br />

PD may benefit from salvage local therapy and continued dabrafenib.<br />

Extent and burden <strong>of</strong> disease correlates with reduced OS, but not RECIST<br />

response.<br />

Sites <strong>of</strong> first RECIST PD<br />

IC only EC only IC and EC Total<br />

Existing lesions only 1 5 2 8<br />

New lesions only 0 0 0 0<br />

Existing and new 4 1 6 11<br />

Total 5 6 8 19<br />

8557 General Poster Session (Board #33G), Sun, 8:00 AM-12:00 PM<br />

The melanoma risk loci as determinants <strong>of</strong> melanoma prognosis. Presenting<br />

Author: Justin Rendleman, New York University Cancer Institute, New<br />

York, NY<br />

Background: Genetic risk factors <strong>of</strong> human cancer emerge as promising<br />

markers <strong>of</strong> clinical outcome. The recent melanoma genome-wide scans<br />

(GWAS) have identified loci associated with the disease risk, nevi or<br />

UV/pigmentation, but the prognostic potential <strong>of</strong> these variants is yet to be<br />

determined. In this study, we performed the first-to-date systematic<br />

evaluation <strong>of</strong> the association between established melanoma risk loci and<br />

melanoma progression. Methods: 891 melanoma patients prospectively<br />

accrued and followed up at NYU Medical Center were studied. We<br />

examined the association <strong>of</strong> 108 melanoma susceptibility single nucleotide<br />

polymorphisms (SNPs), selected or imputed from recent GWASs on<br />

melanoma, nevi or pigmentation, with recurrence-free survival (RFS) and<br />

overall survival (OS). The genotyping was performed using Sequenom<br />

I-plex. Cox PH model was used to test the association between each SNP<br />

and RFS and OS adjusted by age at diagnosis, gender, tumor stage and<br />

histological subtype. ROC curves were used to measure predictive utility <strong>of</strong><br />

SNPs in predicting 3-year recurrence. Results: The strong association was<br />

observed for rs7538876 (RCC2) with RFS (HR�2.445, 95% CI 1.57 –<br />

3.8, p�0.0006) and rs9960018 (DLGAP1) with both RFS and OS<br />

(HR�4.7, 95% CI�2.11-10.43, p�0.0061, HR�1.55, 95% CI�1.11-<br />

2.17, p�0.0094, respectively) using adjusted multivariate analysis. In<br />

addition, we identified the classifier with rs7538876 and rs9960018,<br />

stage and histological type at primary tumor diagnosis, achieving a higher<br />

area under the ROC curve (AUC�84%) compared to the baseline<br />

(AUC�78%) in predicting 3-year recurrence. Univariate survival analyses<br />

have identified associations <strong>of</strong> several SNPs with ulceration and/or tumor<br />

thickness. Conclusions: Our data revealed an association between specific<br />

melanoma susceptibility variants and worse clinicopathological variables at<br />

the time <strong>of</strong> diagnosis as well as worse disease outcome. The strength <strong>of</strong><br />

associations observed for rs7538876 and rs9960018 suggest biological<br />

implication <strong>of</strong> these loci in melanoma recurrence. The observed predictive<br />

patterns <strong>of</strong> associated variants with clinical outcome provide for the first<br />

time evidence for the potential utilization <strong>of</strong> genetic markers in melanoma<br />

prognostication.<br />

8559 General Poster Session (Board #34A), Sun, 8:00 AM-12:00 PM<br />

MAGE-A3 expression in patients screened for the DERMA trial: A phase III<br />

trial testing MAGE-A3 immunotherapeutic in the adjuvant setting for stage<br />

IIIB-C-Tx melanoma. Presenting Author: John F. Thompson, Melanoma<br />

Institute Australia, Sydney, Australia<br />

Background: Administration <strong>of</strong> MAGE-A3 immunotherapeutic involves the<br />

active immunization <strong>of</strong> patients (pts) with tumors expressing the MAGE-A3<br />

protein. This new investigational approach has been previously assessed in<br />

two phase II trials, one in pts with metastatic melanoma (NCT00086866)<br />

and another in pts with completely resected non-small cell lung cancer<br />

(NCT00290355). Based on the positive responses observed in both<br />

studies, a randomized phase III placebo controlled trial assessing MAGE-A3<br />

immunotherapeutic as adjuvant treatment in pts with resected, regionally<br />

advanced melanoma (stage IIIB-C-Tx AJCC/UICC 2010) is currently ongoing<br />

(DERMA Phase III trial, NCT00796445). Methods: Formalin-fixed,<br />

paraffin-embedded tumor tissues were prepared from surgically removed<br />

metastatic lymph nodes and tested for MAGE-A3 expression by qRT-PCR.<br />

Other baseline patient and tumor characteristics were collected during<br />

screening to further investigate factors that could potentially influence<br />

MAGE-A3 expression. Results: Between Dec 1, 2008 and Oct 25, 2011,<br />

3917 pts were screened. Of the 3,183 valid samples (excluding the 513<br />

inconclusive tests [66% for poor quality, 27% out-<strong>of</strong> range, 7% miscellaneous])<br />

and the 221 missing samples, 2,092 (65.7%) were positive for<br />

MAGE-A3 expression. In these stage III melanoma pts, no difference in<br />

MAGE-A3 expression levels were identified with regard to (1) disease stage<br />

- IIIB (62.6%), IIIC (66.5%) and IIITx (66.2%); (2) gender - male (64.2%),<br />

female (68.1%); (3) age group - 18 to 39 years (63.2%), 40 to 49 years<br />

(65.3%), 50 to 59 years (66.8%), 60 to 69 years (68.1%) and 70 years<br />

and over (63.4%) and (4) region: Europe (66.1%), North America (63.0%)<br />

and rest <strong>of</strong> the world including Argentina, Brazil, Australia, Mexico, Taiwan,<br />

Japan, Korea (67.9%). Conclusions: Expression <strong>of</strong> the MAGE-A3 gene in<br />

the DERMA trial population (stage IIIB-IIIC-IIITx) was 66%. It was not<br />

correlated with age, gender, disease stage or geographic region. This<br />

expression frequency is consistent with published data in metastatic<br />

melanoma (Van den Eynde, 1997; Vourc’h-Jourdain et al, 2009) and has<br />

potentially important clinical implications.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


8560 General Poster Session (Board #34B), Sun, 8:00 AM-12:00 PM<br />

Adjuvant sunitinib in high-risk patients with uveal melanoma: A pilot study.<br />

Presenting Author: Matias Emanuel Valsecchi, Department <strong>of</strong> Medical<br />

Oncology, Jefferson Medical College <strong>of</strong> Thomas Jefferson University,<br />

Philadelphia, PA<br />

Background: Uveal melanoma is the most common primary intraocular<br />

cancer in adults. Despite the successful treatments for primary tumors, up<br />

to 50% <strong>of</strong> the patients later die <strong>of</strong> distant metastases. Currently no<br />

effective adjuvant treatment is available for these patients. Our group<br />

previously reported that sunitinib stabilized systemic metastases in 65% <strong>of</strong><br />

uveal melanoma patients who failed prior treatments. In this pilot study, we<br />

tested sunitinib in an adjuvant setting in high-risk patients with primary<br />

uveal melanoma. Methods: Patients with estimated metastatic death rate �<br />

50% based on the following criteria were eligible: (1) monosomy 3 and 8q<br />

amplification; (2) large tumor (� 15 mm in diameter and �7 mmin<br />

thickness); (3) monosomy 3 and other risk factors (large tumor, epithelioid<br />

dominant cell type, local recurrence, or extra-scleral extension). Sunitinib<br />

was given at 25 mg PO daily for at least 6 months. Primary endpoint was<br />

disease-free survival (DFS) and overall survival (OS), estimated with<br />

Kaplan-Meier analysis (SPSS 17.0). Secondary endpoint was safety.<br />

Results: A total <strong>of</strong> 23 Caucasian patients (median, 54 years; range: 25 –<br />

77) were enrolled. All patients received sunitinib for at least 6 months<br />

(range: 6 – 12). Eighteen patients had confirmed monosomy 3, from whom<br />

13 (72%) also showed 8q amplification. The median follow-up was 24<br />

months (range 12 – 54 months). Only one patient died <strong>of</strong> unknown cause<br />

(OS: 30.4 months). A total <strong>of</strong> seven patients (30%) developed systemic<br />

metastases, all <strong>of</strong> which were liver metastases. The DFS and OS rates at 2<br />

years were 70% (95% CI: 47 – 86%) and 100%, respectively. The OS rate<br />

at 2 years was better than historical control with monosomy 3 patients<br />

(51%, 95% CI: 31 – 71%) (Invest Ophthalmol Vis Sci 2003; 44:1008). In<br />

those patients who relapsed, the median time to progression after finishing<br />

sunitinib was 2 months (range: 0 – 8). The most common adverse events<br />

were: diarrhea (47%), fatigue (39%), dermatitis (30%), stomatitis (13%),<br />

leucopenia (8%), and nausea (4%). Most were grade 1 or 2 (88%) and only<br />

one was considered grade 4 (diarrhea). Conclusions: In high-risk uveal<br />

melanoma patients with estimated metastatic death rate <strong>of</strong> � 50%,<br />

adjuvant sunitinib showed promising results and deserves further investigation.<br />

8562 General Poster Session (Board #34D), Sun, 8:00 AM-12:00 PM<br />

mTOR pathway activation in KIT-mutated melanoma with acquired imatinib<br />

resistance. Presenting Author: Lu Si, Peking University Cancer<br />

Hospital and Institute, Beijing, China<br />

Background: c-Kit mutation is a common genetic abnormality in certain<br />

melanoma subtypes, and is a target for treatment. However, the mechanisms<br />

<strong>of</strong> secondary (acquired) resistance and how to treat patients in the<br />

second line are unknown. Methods: Tissuesamples were obtained from<br />

c-Kit-mutated melanoma patients who failed <strong>of</strong> imatinib after having PR or<br />

SD for at least 6 months pre- and post-imatinib therapy. Somatic mutations<br />

in genes in MAP kinase (c-Kit, BRAF, NRAS, etc) and PI3K-AKT-mTOR<br />

(AKT1, TSC1, PTEN, etc) pathways were detected by DNA sequencing. The<br />

expression <strong>of</strong> pS6RP, p4E-BP1, pAKT and pERK1/2 were examined by<br />

immunohistochemistry (IHC) assays. Results: Four paired samples (formalinfixed,<br />

paraffin-embedded) were analyzed. 1) Laboratory results: No secondary<br />

mutations in addition to the c-Kit mutations identified at baseline were<br />

identified. However, IHC showed increased pS6RP and p4E-BP1 (two<br />

targets <strong>of</strong> mTOR activation) as well as increased pAKT and pERK1/2 in<br />

imatinib-resistant samples, suggested that mTOR signaling pathway was<br />

selectively activated in these secondary imatinib-resistant melanomas. 2)<br />

Treatment: Everolimus is an orally administered inhibitor <strong>of</strong> mTOR. The<br />

four patients received everolimus (10 mg/day) by continuous daily dosing.<br />

The tumor response revealed 1 PR and 3 SD (all �3 months) with grade 1-2<br />

toxicities consisting <strong>of</strong> myelosupression, hyperglycemia, hypercholesterolemia<br />

and hypertriglyceridemia, consistent with previously reported toxicities<br />

for everolimus. Conclusions: The case series provides an important clue<br />

that activation <strong>of</strong> the mTOR signal pathway may be one <strong>of</strong> the mechanisms<br />

for secondary imatinib resistance in c-Kit-mutated melanomas. The results<br />

also show that everolimus may be effective and safe for melanoma patients<br />

who develop resistance to imatinib and could be investigated in combination<br />

with c-Kit inhibitors in treatment-naive patients.<br />

Melanoma/Skin Cancers<br />

555s<br />

8561 General Poster Session (Board #34C), Sun, 8:00 AM-12:00 PM<br />

Association <strong>of</strong> the activation <strong>of</strong> the mTOR pathway with prognosis in<br />

Chinese melanoma patients. Presenting Author: Yan Kong, Peking University<br />

Cancer Hospital and Institute, Beijing, China<br />

Background: mTOR is a ubiquitously expressed protein kinase and a<br />

validated target in the treatment <strong>of</strong> cancer. However, the characterization<br />

<strong>of</strong> mTOR pathway in Asian melanoma populations has not been reported.<br />

Methods: This study involved primary tumor samples from 173 melanoma<br />

patients (79 acral melanomas, 57 mucosal melanomas, 17 melanomas on<br />

skin with chronic sun-induced damage, 20 melanomas on skin without<br />

chronic sun-induced damage) in Peking University Cancer Hospital &<br />

Institute. <strong>Clinical</strong> data were collected. Immunohistochemistry assays using<br />

antibodies against pmTOR, pS6RP, p4E-BP1 and pAKT were performed on<br />

formalin-fixed, paraffin-embedded specimens. Somatic mutations within<br />

the hotspot regions <strong>of</strong> frequently mutated genes in mTOR pathway (AKT1,<br />

TSC1, PI3K genes, etc) were analyzed by PCR amplification and Sanger<br />

sequencing. Results: Among the 173 samples, 26% <strong>of</strong> the cases (45/173)<br />

demonstrated positive staining with pmTOR. Expression <strong>of</strong> pS6RP was<br />

observed in 37% (64/173) <strong>of</strong> cases. Expression <strong>of</strong> p4E-BP1 was observed<br />

in 27% (46/173) <strong>of</strong> cases. pAKT was expressed by 22% (38/173) <strong>of</strong> cases.<br />

Somatic mutations in examined genes were detected. The median survival<br />

time for patients with expression <strong>of</strong> pmTOR was significantly shorter than<br />

patients with absence <strong>of</strong> pmTOR expression (25.3 vs 62.9 months, P<br />

�0.048). In addition, statistical differences were found for ulceration rates<br />

between melanomas with or without pS6RP (64.1%vs 35.9%, P� 0.049).<br />

Moreover, the median survival time for mucosal melanoma patients with<br />

pAKT expression was significantly shorter than for patients with absence <strong>of</strong><br />

pAKT expression (20.4 vs 52.1 months, P �0.022). Conclusions: These<br />

data demonstrate that activation <strong>of</strong> the mTOR signaling pathway carries<br />

prognostic significance in Asia patients with melanoma. Targeted therapies<br />

to mTOR pathway may <strong>of</strong>fer a therapeutic benefit for these melanoma<br />

patients.<br />

8563 General Poster Session (Board #34E), Sun, 8:00 AM-12:00 PM<br />

Analysis <strong>of</strong> KIT expression and mutations in sinonasal, genital, and<br />

acrolentiginous melanomas. Presenting Author: Christian R. F. Bull,<br />

Department <strong>of</strong> Dermatology, University Hospital Zurich, Zurich, Switzerland<br />

Background: Melanoma is subdivided into molecular defined groups. One<br />

major subtype is characterized by a mutation in the BRAF gene. BRAF is<br />

less commonly mutated in acrolentiginous (ALM) and mucosal (MM)<br />

melanomas, instead these subtypes predominantly show aberrations in the<br />

KIT/CD117 gene. KIT aberrations predict the outcome <strong>of</strong> targeted therapies<br />

in ALM and MM patients, however the particular role <strong>of</strong> KIT expression<br />

in these melanoma subtypes remains controversial. To explore the relationship<br />

between KIT expression, mutation, and tumor stages, we investigated<br />

immunohistochemical KIT expression and mutation status in primary<br />

ALM/MM lesions and their metastases. We also compared the frequency <strong>of</strong><br />

KIT mutations <strong>of</strong> MM in different anatomic locations. Methods: KIT<br />

immunoreactivity and mutation status was assessed in 41 ALM and 25 MM<br />

patients, using polyclonal rabbit anti-human KIT/CD117 antibody, and<br />

PCR was performed for KIT gene exons 9,11,13,17 and 18. Of these, 19<br />

ALM and 15 MM patients had matched primary and metastatic lesions.<br />

Phosphorylated extracellular regulated kinase (P-ERK) was investigated in<br />

a subset <strong>of</strong> 8 ALM and 8 MM matched primary/metastatic pairs by<br />

immunohistochemistry. Results: Heterogeneous KIT immunoreactivity was<br />

observed in both primary and metastatic lesions. Mutations were present in<br />

four <strong>of</strong> 41 ALM (10%) and five <strong>of</strong> 25 MM (20%) patients. Only vulvar<br />

mucosal samples carried KIT mutations in contrast to sinonasal lesions (p�<br />

0.0109). In KIT-mutated tumors, the mutations were present in KIT<br />

expressing as well as KIT negative cells, as shown by Laser Capture<br />

Microdissection (LCM). P-ERK expression was preferentially found in<br />

metastases. Conclusions: KIT expression is heterogeneous and shows no<br />

relationship with disease progression and mutation status, therefore KIT<br />

immunoreactivity is not a valuable marker for treatment decisions. In<br />

mucosal melanoma patients, KIT mutation frequency is significantly<br />

associated with anatomic location. Vulvar melanoma patients are prone to<br />

carry activating KIT mutations and thus may be more likely to benefit from<br />

KIT-targeted therapies than other subtypes. Therefore, especially vulvar<br />

melanoma patients should be screened for activating KIT mutations.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


556s Melanoma/Skin Cancers<br />

8564 General Poster Session (Board #34F), Sun, 8:00 AM-12:00 PM<br />

High-risk uveal melanoma: A prospective study evaluating the role <strong>of</strong><br />

magnetic resonance imaging <strong>of</strong> the liver. Presenting Author: Ernie Marshall,<br />

Clatterbridge Centre for Oncology, Wirral, United Kingdom<br />

Background: Almost 50% <strong>of</strong> uveal melanomas are fatal. Metastatic death<br />

occurs almost exclusively with tumours showing chromosome 3 loss and 8q<br />

gain. Metastases, which almost always involve the liver, are resectable in<br />

some patients. They are rarely detectable when the patient presents with<br />

the primary ocular tumour. Screening is therefore necessary, but there is no<br />

consensus as to who should be screened, how <strong>of</strong>ten, and for how long.<br />

Methods: Uveal melanoma patients with ECOG performance status 0-2<br />

were eligible if their risk <strong>of</strong> metastatic death at 5 years exceeded 50%.<br />

Survival probability was estimated by multivariate analysis <strong>of</strong> tumour stage,<br />

histological grade and genetic tumour type. Patients underwent screening<br />

6monthly, clinical examination, non-contrast liver MRI and liver function<br />

tests for at least five years. Results: Between Jan 2000 and November<br />

2010, 279 high-risk patients were referred for screening. Of these, 188<br />

(84 male, 104 female) accepted screening and underwent as least 1 MRI.<br />

The median age was 63 years (IQR 16.5). Median basal tumour diameter<br />

was 16.5mm (IQR 5.25). Chromosome 3 loss was detected in 175<br />

tumours. Median follow up time was 28.8 months (IQR 29.1). Median<br />

relapse-free survival was 33 months (95% CI 28-38) with a 35%<br />

relapse-free survival at 5 years. After a median <strong>of</strong> 18 months (IQR 20),<br />

screening detected metastases in 90/188 (48%), 83 <strong>of</strong> whom were<br />

asymptomatic. 12 patients underwent R0 liver resection, which increased<br />

the median survival from 10 (95% CI 8.1 - 11.9) to 24 (95% CI 20.2-<br />

27.8) months. The screening programme stimulated a UK NCRI portfolio <strong>of</strong><br />

clinical trials in which 23 <strong>of</strong> these patients were subsequently treated.<br />

Conclusions: Six-monthly liver MRI detects metastases from uveal melanoma<br />

at an early stage, thereby enhancing opportunities for surgical<br />

metastatectomy, clinical trial participation and prolonging life.<br />

8566 General Poster Session (Board #34H), Sun, 8:00 AM-12:00 PM<br />

Biomarker for benefit from ipilimumab: Correlation <strong>of</strong> breadth <strong>of</strong> humoral<br />

tumor-antigen-specific immunity with outcome. Presenting Author: Sarah<br />

Ellis, University Hospital Southampton NHS Foundation Trust, Southampton,<br />

United Kingdom<br />

Background: Checkpoint blockade through CTLA4 with ipilimumab has for<br />

the first time improved survival in patients with advanced melanoma.<br />

Immune-mediated toxicity and increase in humoral and T-cell anti-NY-<br />

ESO-1 immune responses after treatment are both linked with benefit.<br />

However there is currently no broadly relevant, immunological biomarker<br />

for predicting outcome prior to initiation <strong>of</strong> therapy. Methods: Using a novel<br />

immunological assay developed by Serametrix Corporation we analysed the<br />

presence <strong>of</strong> antibodies to a proprietary panel <strong>of</strong> tumour associated antigens<br />

(TAAs) in 34 patients with advanced melanoma, given ipilimumab at<br />

3mg/kg as second or subsequent line <strong>of</strong> treatment. Results: 34 patients<br />

were consented and analysed, 22 females, 13 males. With a median age <strong>of</strong><br />

63 years median overall survival was 24 weeks; 6/34 patients had an<br />

objective response to ipilimumab (17.6%, median survival not reached).<br />

Antibody (AB) responses were tested against the panel <strong>of</strong> 26 TAAs. This<br />

panel includes BRAF, Cancer/Testis and other described or novel TAAs. We<br />

observed a wide range in the incidence and levels <strong>of</strong> antibody response. 12<br />

patients had no detectable immunity, 6 an AB response to a single antigen,<br />

5 AB response to 2, and 11 patients to 3 or more TAA. Five patients had<br />

either pre-existing anti-BRAF antibodies or developed these after CTLA4blockade.<br />

Survival was significantly longer in those patients with preexisting<br />

antibodies to 2 or more TAA, compared to those with 0 or 1<br />

specificities (median survival 39.4 vs 16.4 weeks p�0.02). Conclusions:<br />

Patients with advanced melanoma had variable levels <strong>of</strong> humoral immunity<br />

to a large number <strong>of</strong> TAA, including to BRAF. The presence <strong>of</strong> antibody<br />

response to 2 or more TAA correlated with longer survival following<br />

treatment with ipilimumab and appears to provide a biomarker for identifying<br />

those patients that are most likely to respond to checkpoint blockade<br />

with ipilimumab.<br />

8565 General Poster Session (Board #34G), Sun, 8:00 AM-12:00 PM<br />

MicroRNA alterations associated with BRAF status in melanoma. Presenting<br />

Author: Michelle W. Ma, New York University School <strong>of</strong> Medicine, New<br />

York, NY<br />

Background: We hypothesize that BRAF mutations result in microRNA<br />

(miRNA) alterations which contribute to orchestrating the mutant BRAF’s<br />

oncogenic effects in melanoma. Our study is the first to examine the<br />

association between the BRAF mutation status in primary melanomas and<br />

the expression <strong>of</strong> miRNAs that target known tumor suppressors. Methods:<br />

84 prospectively accrued melanoma patients at New York University<br />

Langone Medical Center were studied. DNA and total RNA were extracted<br />

from consecutive sections <strong>of</strong> formalin-fixed paraffin-embedded primary<br />

tissues. BRAF mutation status was determined by DNA sequencing. RNA<br />

was hybridized to miRCURY miRNA microarrays containing 1314 probes.<br />

Normalized miRNA data were analyzed using the t-test (p�0.05) to<br />

identify differentially expressed miRNAs between BRAFmut vs. BRAFwt<br />

cases. Those with an average fold change (FC) � 2 were selected for<br />

predicted (TargetScan, PicTar) and validated (miRWalk) gene target<br />

analysis, and overlapping genes targeted by � 2 miRNAs were analyzed<br />

using pathway-mapping algorithms (KEGG, BioCarta, PANTHER). Results:<br />

48 (57%) primaries were BRAFwt and 36 (43%) were BRAFmut (26<br />

V600E, 4 V600K, 1 V600R, 1 V600D, 4 other). 30 miRNAs met the<br />

criteria for statistically significant differential expression and FC thresholding:<br />

let-7i, miR-23c, -26a/b, -27b, -34a, -98, -126*, -141, -148a, -181b,<br />

-195, -199a-3p, -199a/b-5p, -200a/b/c, -203, -205, -455-3p, -491-3p,<br />

-606, -641, -646, -1297, -4301; miRPlus-C1070, -C1110, -G1246-3p<br />

(average FC: 2.3-3.5, all increased in BRAFmut vs. BRAFwt). Predicted<br />

and validated target gene analysis revealed 317 genes, <strong>of</strong> which 110 (35%)<br />

were convergent targets <strong>of</strong> � 2 miRNAs. Pathway analyses <strong>of</strong> the predicted,<br />

validated, and convergent target genes pointed to the potential impact <strong>of</strong><br />

BRAFmut-associated miRNAs on known tumor suppressors FAS, PTEN,<br />

and TNF and the p53 pathway. Conclusions: Differentially expressed<br />

miRNAs in BRAFmut vs. BRAFwt primaries target genes with known roles<br />

in melanoma biology and/or treatment response. These miRNAs warrant<br />

further study as potential effectors <strong>of</strong> the BRAFmut oncogenic program.<br />

8567 General Poster Session (Board #35A), Sun, 8:00 AM-12:00 PM<br />

A single-arm, open-label, U.S. expanded access study <strong>of</strong> vemurafenib in<br />

patients with metastatic melanoma. Presenting Author: Lynn Mara Schuchter,<br />

University <strong>of</strong> Pennsylvania, Philadelphia, PA<br />

Background: Vemurafenib (vem) has been FDA approved for the treatment<br />

<strong>of</strong> unresectable or metastatic BRAFV600E mutated melanoma since August<br />

2011 based on results <strong>of</strong> a randomized phase III study (treatment-naive)<br />

and a single arm phase II study (previously treated). We report results <strong>of</strong> an<br />

expanded access study that allowed appropriate patients (pts) to receive<br />

vem until the drug was approved. Methods: Eligible pts had metastatic<br />

melanoma with a BRAFV600E mutation as detected by the cobas 4800<br />

BRAF V600<br />

Mutation Test. Enrolled pts received oral vem 960 mg b.i.d.<br />

Adverse events (AEs) were evaluated for vem-related toxicities; tumor<br />

responses were assessed using RECIST 1.1. Results: 29 US sites screened<br />

745 pts and enrolled 374 from December 2010 until October 2011. The<br />

following results are based on a median follow up time and treatment<br />

duration <strong>of</strong> 2 months. At baseline, mean age <strong>of</strong> pts was 54 y with 22% <strong>of</strong><br />

pts �65 y; 75% had stage M1c disease; 29% had received radiotherapy for<br />

brain metastases. 19% <strong>of</strong> pts were ECOG PS 2 or 3; 71% <strong>of</strong> pts had prior<br />

systemic therapy for metastatic melanoma (21% 1 regimen; 50% �2<br />

regimens). 50 pts had prior adjuvant treatment. At data cut-<strong>of</strong>f, 243 pts<br />

had sufficient follow-up time for tumor assessment. In this group, the<br />

unconfirmed overall response rate was 52% (95% CI, 46 to 59). The<br />

median time to response was 1.8 months. Based on 240 pts with available<br />

ECOG PS status at time <strong>of</strong> analysis, response rate was 53% for pts with<br />

ECOG PS 0 or 1 (n�209), and 45% for pts with ECOG PS 2 or 3 (n�31).<br />

370 pts were evaluable for safety analysis. The most common vem-related<br />

AEs were rash (36%), arthralgia (33%) and fatigue (21%) with the majority<br />

(~90%) <strong>of</strong> grade 1 or 2. 25 vem-related serious AEs were reported in 5.4%<br />

<strong>of</strong> pts with a slightly higher rate <strong>of</strong> pts with ECOG PS 2 or 3 (8.7%)<br />

compared to ECOG PS 0 or 1 (4.7%). 18% <strong>of</strong> pts missed at least one dose<br />

and 11% <strong>of</strong> pts required dose reduction <strong>of</strong> at least one level due to AEs.<br />

Conclusions: This expanded access study, with its limited follow-up time,<br />

confirms the established rapid and high tumor response rate with vem. No<br />

new safety signals were detected. Compared to the overall population, pts<br />

with an ECOG PS 2 or 3 demonstrated a similar benefit.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


8568 General Poster Session (Board #35B), Sun, 8:00 AM-12:00 PM<br />

Ipilimumab-induced hypophysitis in melanoma patients. Presenting Author:<br />

Typhanie Carré, Dermatology Department, Timone Hospital, Aix<br />

Marseille University, Marseille, France<br />

Background: Ipilimumab (Ipi) is a human monoclonal antibody directed<br />

against cytotoxic T-lymphocyte antigene-4 (CTLA-4) recently approved for<br />

the treatment <strong>of</strong> metastatic melanoma (MM) and currently under investigation<br />

in the adjuvant setting. Methods: Retrospective analysis <strong>of</strong> patients<br />

treated with ipi between June 2006 and September 2011 in our department<br />

in Marseille. As some patients are still blinded in trials, the exact<br />

number <strong>of</strong> patient under ipi is unknown. We present a minimal percentage<br />

(�%) assuming that the 120 patients received ipi. Results: A total <strong>of</strong> 120<br />

patients were treated: 76 stages IV MM, from which 16 in the BMS clinical<br />

trials (CA184-022, -024, and-025 and MDX 010-20) and 44 stages III<br />

MM (in the BMS CA184-029 trial). Stage IV MM were administered 0.3, 3<br />

or 10mg/kg IV dosage, while stages III MM were randomly assigned to<br />

receive 10 mg/ kg or placebo (1:1 ratio). Hypophysitis was diagnosed in 12<br />

patients (�10%): 2/76 patients with stage IV MM (�2. 6 %) and 10/44<br />

patients with stage III MM (�22.7). Diagnosis was performed at the 1st ,3rd and 4th administration in respectively 2 (1.6%), 6 (50%) and 4 patients<br />

(33.3%). <strong>Clinical</strong> symptoms included headaches (n�11; 91.6%), asthenia<br />

(n�7; 58.3%) and decreased libido (n�2; 1.6%). Adrenal, thyroidal<br />

and gonadal axis were affected in respectively 6 (50%), 9 (75%) and 7<br />

patients (58.3%). MRI changes were observed in 7 patients (58.3%):<br />

pituitary swelling in 5 patients (41.6%) and heterogeneous enhancement<br />

in 4 patients (33.3%) including 2 patients with normal biology. Corticosteroids<br />

supplementation was required in 11 patients and thyroidian supplementation<br />

in 4 patients. <strong>Clinical</strong> symptoms regressed within one week in 8<br />

patients (66.6%). Conclusions: Ipi-induced hypophysitis is detectable only<br />

if clinicians are aware <strong>of</strong> these unspecific signs. Only MRI can make<br />

diagnosis in some patients without clinical and/or biological signs. Our data<br />

suggest that it develops especially for 10mg/kg dosage, after the third<br />

administration, and that the rate could be higher in patients with a normal<br />

immune system (adjuvant treatment), than in metastatic ones. Hormonal<br />

supplementation usually controls the disease.<br />

8570 General Poster Session (Board #35D), Sun, 8:00 AM-12:00 PM<br />

Percutaneous hepatic perfusion (PHP or ChemoSat) with melphalan versus<br />

best alternative care (BAC) in patients (pts) with hepatic metastases from<br />

melanoma: A post hoc analysis <strong>of</strong> PHP-randomized versus BAC-to-PHP<br />

crossover versus BAC-only pts. Presenting Author: H. Richard Alexander,<br />

University <strong>of</strong> Maryland School <strong>of</strong> Medicine, Baltimore, MD<br />

Background: PHP (Chemosat, Delcath Systems Inc, New York, NY) allows<br />

repeated delivery <strong>of</strong> high dose chemotherapy to the liver while limiting<br />

unwanted systemic exposure by extracorporeal filtration <strong>of</strong> hepatic venous<br />

blood. A prospective randomized multicenter study compared melphalan<br />

PHP versus BAC in pts with unresectable hepatic metastases from ocular or<br />

cutaneous melanoma and showed a highly significant advantage for PHP in<br />

terms <strong>of</strong> the primary endpoint, hepatic PFS, and multiple secondary<br />

endpoints. We present an exploratory post-hoc analysis <strong>of</strong> pts assigned to<br />

BAC who crossed over to PHP after disease progression (BAC-to-PHP<br />

crossover) versus those who did not (BAC-only) versus PHP-randomized<br />

pts. Methods: 93 pts were randomized to PHP (n�44) or BAC (n�49). In<br />

the BAC group, crossover to PHP with melphalan was permitted after<br />

hepatic disease progression. Up to 6 PHP treatments with melphalan 3.0<br />

mg/kg ideal body weight were given every 4–8 weeks. Results: In the BAC<br />

group, 28 <strong>of</strong> 49 pts crossed over to PHP. Updated efficacy findings<br />

(investigator) as <strong>of</strong> 31 March 2011 are shown in the table. Independent<br />

Review Committee results were similar to investigator-assessed efficacy.<br />

Safety in BAC-to-PHP crossover pts was similar to that seen in PHPrandomized<br />

pts. The most common toxicities in BAC-to-PHP crossover pts<br />

were hematological: peri-procedural thrombocytopenia (71%) and anemia<br />

(57%), and melphalan-related neutropenia (82%) and thrombocytopenia<br />

(79%). Conclusions: Efficacy <strong>of</strong> melphalan PHP in BAC-to-PHP crossover<br />

pts was consistent with that seen in PHP-randomized pts with hepatic<br />

metastases from melanoma. Crossover from BAC to PHP after hepatic<br />

disease progression led to a median 11-month survival advantage vs BAC<br />

alone.<br />

Endpoint<br />

PHP-randomized<br />

(n�44)<br />

BAC-only<br />

(n�21)<br />

BAC-to-PHP crossover<br />

(n�28)<br />

Median hPFS, months 8.0 1.6 8.8<br />

HR (crossover vs BAC-only) 0.32<br />

Median overall survival, months 9.8 4.1 15.3<br />

HR (crossover vs BAC-only) 0.33<br />

No. alive (f/u 9.7–53.5 mos) 4 3* 7<br />

*One patient crossed over but never received PHP. BAC-only pts: chemoembolization, HAI<br />

nab-paclitaxel, temozolomide.<br />

Melanoma/Skin Cancers<br />

557s<br />

8569 General Poster Session (Board #35C), Sun, 8:00 AM-12:00 PM<br />

Outcomes <strong>of</strong> patients with malignant melanoma treated with immunotherapy<br />

prior to or after vemurafenib. Presenting Author: Allison Ackerman,<br />

Beth Israel Deaconess Medical Center, Boston, MA<br />

Background: Treatment <strong>of</strong> patients (pts) with BRAF mutant malignant<br />

melanoma (MM) with vemurafenib (vem), a BRAF inhibitor (BRAFi), is<br />

associated with a high response rate (RR) and improvement in progression<br />

free survival (PFS) and overall survival (OS) compared to standard chemotherapy.<br />

Responses to vem are typically not durable and most pts require<br />

additional therapy. However, there is little data to guide sequencing <strong>of</strong> vem<br />

with other standard therapies such as ipilimumab (ipi) or interleukin 2<br />

(IL-2). Methods: 43 pts treated with vem as part <strong>of</strong> clinical trials from<br />

2009-2012 were retrospectively identified. RR <strong>of</strong> vem was calculated for<br />

all pts as well as for pts treated with immunotherapy (IT) prior to vem. The<br />

OS from first systemic and vem treatment, duration <strong>of</strong> vem therapy, OS<br />

from when pts came <strong>of</strong>f vem, and PFS and OS for post-vem ipi was<br />

determined using Kaplan-Meier estimates. Results: Of the 43 pts, 11<br />

remain on vem and 32 are <strong>of</strong>f vem (19 deceased) with a median follow up <strong>of</strong><br />

19 mo. Pre-vem LDH was elevated in 46% (17/37 evaluable pts). The RR<br />

to vem was 52% (22/42 evaluable pts), the median duration <strong>of</strong> therapy 5.6<br />

months (mo), and the median OS 19.3 mo, similar to results in trials. The<br />

median OS from initiation <strong>of</strong> any treatment was 27 mo. 16 pts received IT<br />

(11 IL-2, 4 ipi, 1 IL-2 then ipi) prior to vem with a median OS <strong>of</strong> 31.2 mo;<br />

the RR <strong>of</strong> vem following IT was 75% (12/16). At time <strong>of</strong> vem discontinuation<br />

50% <strong>of</strong> pts (12/24 evaluable) had an elevated LDH, and the median<br />

OS <strong>of</strong> the 32 pts who stopped vem was 4 mo from last vem dose. 10 <strong>of</strong> these<br />

pts then received single-agent ipi; six <strong>of</strong> whom had an elevated LDH. Five<br />

pts died within 3 mo <strong>of</strong> last vem dose, and all 10 pts had disease<br />

progression (PD) at 6 months with a median PFS <strong>of</strong> 0.7 mo and OS <strong>of</strong> 2.2<br />

mo. The 3 pts who lived beyond 1 year following vem and then ipi were<br />

subsequently treated with a BRAFi following PD on ipi. Conclusions:<br />

Response to vem following IT appears similar to that seen in previously<br />

untreated pts. Median PFS and OS for pts receiving ipi after discontinuation<br />

<strong>of</strong> vem are poor, possibly due to rapid PD at the time <strong>of</strong> vem<br />

discontinuation. Prolonged OS is primarily seen with resumption <strong>of</strong> BRAF<br />

inhibition. Our data suggests that in appropriately selected pts, IT should<br />

be considered prior to BRAFi.<br />

8571 General Poster Session (Board #35E), Sun, 8:00 AM-12:00 PM<br />

Tim-3 expression and function in natural killer cells from advanced<br />

melanoma patients. Presenting Author: Ines Esteves Domingues Pires Da<br />

Silva, New York University Langone Medical Center, New York, NY<br />

Background: The concept <strong>of</strong> CD8 � T cell exhaustion in the context <strong>of</strong><br />

metastatic cancer has been reinforced by the recent success <strong>of</strong> immunotherapies<br />

targeting the exhaustion markers CTLA-4 and PD-1 in advanced<br />

melanoma. T-cell immunoglobulin 3 (Tim-3), another exhaustion marker,<br />

is also expressed in natural killer (NK) cells, however its role is still<br />

unknown. Recent reports have shown that NK cells, innate immune cells<br />

that eliminate tumors through cytotoxicity and IFN-g production, are<br />

functionally impaired in advanced melanoma patients, although no receptor<br />

has been linked with that phenotype so far. In this study, we<br />

characterize the role <strong>of</strong> Tim-3 in NK cells, particularly in the presence <strong>of</strong> its<br />

natural ligand, Galectin-9 (Gal-9), that is known to be expressed/secreted<br />

by some tumor cells including melanoma. Methods: We compared 20<br />

advanced melanoma donors NK cells with 40 healthy donors NK cells as it<br />

relates to Tim-3 expression (by flow cytometry) and function (cytotoxicity,<br />

IFN-� production and proliferation). NK cells cytotoxicity was measured by<br />

lamp-1 expression, and two different target cells were used: i) K562 cells<br />

(Gal-9 - ) and ii) Gmel Gal-9 � and Gmel Gal-9 - sorted melanoma cells.<br />

Proliferation was quantified by CFSE after 6 days in the presence <strong>of</strong> rhIL-2.<br />

Recombinant rhGal9 effect was tested in cytotoxicity and IFN-� production.<br />

Results: Melanoma patients NK cells express higher levels <strong>of</strong> Tim-3<br />

compared to healthy donors NK cells (p�0.05). Melanoma patients NK<br />

cells have a defect in cytotoxicity, proliferation and IFN-� production.<br />

Tim-3 expression by itself (without engagement <strong>of</strong> specific ligands) does<br />

not negatively affect NK cell functions (p�0.05). However, when rhGal9 is<br />

added to the system, a decrease in NK cell cytotoxicity and IFN-�<br />

production (p�0.05) was observed. Finally, the expression <strong>of</strong> Gal-9 by the<br />

target cells induces a defect in NK cell cytotoxicity (Gmel Gal-9 � vs Gmel<br />

Gal-9 - ). Conclusions: These data suggest that advanced melanoma patients<br />

NK cells are exhausted, although it still remains unclear if Tim-3 is involved<br />

in this phenotype. In addition,the expression/secretion <strong>of</strong> Galectin-9,<br />

immunosuppressive for NK cells, may be a possible mechanism for tumors<br />

to evade immune surveillance.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


558s Melanoma/Skin Cancers<br />

8572 General Poster Session (Board #35F), Sun, 8:00 AM-12:00 PM<br />

Use <strong>of</strong> Cobas 4800 BRAF mutation test for the analysis <strong>of</strong> BRAF V600<br />

mutations in cytological samples (CS) from metastatic melanoma (MM).<br />

Presenting Author: Salvador Martin-Algarra, Clínica Universidad de Navarra,<br />

Pamplona, Spain<br />

Background: Vemurafenib is currently the most active first-line treatment in<br />

MM patients that harbor BRAF-V600E mutations. The Cobas 4800 BRAF<br />

V600 Mutation Test is an FDA-approved, CE-marked test that identifies<br />

formalin-fixed-paraffin-embedded samples (FFPE) carrying the BRAF V600E<br />

mutation. Fine needle aspirates are frequently the only samples available in<br />

MM patients, but the use <strong>of</strong> CS has not been validated in this platform. We<br />

have evaluated the capability to identify BRAF-V600E mutations in CS <strong>of</strong><br />

MM with the Cobas 4800 BRAF Mutation Test, and results have been<br />

compared to the conventional Sanger direct sequencing method. Methods:<br />

BRAF mutations have been studied in 117 samples from 98 MM patients<br />

with the conventional Sanger method: 37 (32%) FFPE and 80 (68%) CS.<br />

DNA was extracted from 4-micron sections in FFPE and from stained<br />

smears in CS. All CS contained �50% <strong>of</strong> tumor cells. In a second step, 57<br />

out <strong>of</strong> the 117 samples have been studied using Cobas (25 CS and 32<br />

FFPE). In CS, Nucleospin Tissue (Macherey-Nagel) DNA extraction was<br />

used in 17 cases and the Cobas procedure in 8. FFPE and CS paired<br />

samples were available in 9 patients. Results were compared using the<br />

Kappa coefficient. Results: There was 93% (53/57) agreement between<br />

Cobas and direct sequencing. All V600E� cases were detected with both<br />

methods. Four V600R� cases were negative using Cobas. Mean DNA<br />

concentrations using Nucleospin and Cobas procedures were 55.20 ng/�l<br />

and 11.37 ng/�l respectively. Concordance using Cobas in FFPE and CS<br />

paired samples was 100%. The table shows the results with Cobas and<br />

direct sequencing in 25 CS. One V600R� case was not detected with<br />

Cobas. Conclusions: The Cobas 4800 BRAF Mutation Test results using CS<br />

containing �50% <strong>of</strong> tumor cells are identical to those obtained with FFPE.<br />

In our experience, DNA concentrations from CS with the Cobas method are<br />

lower than those obtained with Nucleospin procedure. These results<br />

provide a strong rationale for a larger validation study.<br />

Direct sequencing results<br />

WT V600E V600K V600R Total<br />

COBAS 4800 system results<br />

MND 4 0 0 1 5<br />

V600 0 16 4 0 20<br />

Total 4 16 4 1 25<br />

MND: Mutation not detected.<br />

8574 General Poster Session (Board #35H), Sun, 8:00 AM-12:00 PM<br />

Development <strong>of</strong> a melanoma risk prediction model incorporating MC1R<br />

genotype and indoor tanning exposure. Presenting Author: Lauren A.<br />

Smith, New York University Langone Medical Center, New York, NY<br />

Background: Invasive melanoma is the second most common cancer in<br />

young adults, yet they exhibit poor skin-protective behavior. We previously<br />

demonstrated a significant association between melanoma risk, melanocortin<br />

receptor (MC1R) polymorphisms, and indoor ultraviolet light (UV)<br />

exposure. As existing melanoma risk models do not take these factors into<br />

account, we investigated whether these variables would improve the<br />

predictive ability <strong>of</strong> a clinical risk model, especially in a younger population.<br />

Methods: We determined MC1R genotype and collected self-reported<br />

phenotypic and UV (indoor and outdoor) exposure variables from 923<br />

melanoma cases and 813 healthy controls between ages 25 – 59 from the<br />

Minnesota Skin Health Study. These data were initially used to develop a<br />

clinical melanoma risk model (Model A) with conventional risk factors (i.e.<br />

age; gender; hair, skin, and eye color; mole count, freckling, and family<br />

melanoma history). We then developed a second model (Model B) combining<br />

outdoor UV, indoor UV, and MC1R genotype variables with those in<br />

Model A to determine if the model’s predictive ability improved. Finally, we<br />

assessed the predictive ability <strong>of</strong> both models when confined to younger<br />

subjects (ages 25-35). Results: The clinical model, combining conventional<br />

melanoma risk factors (Model A), yielded an area under the receiver<br />

operating characteristic curve (AUC) <strong>of</strong> 0.72 (95% CI 0.70 – 0.75).<br />

Incorporating outdoor UV, indoor UV, and MC1R genotype variables (Model<br />

B) increased the AUC to 0.75 (p�0.001, 0.72 – 0.77). Confining the<br />

analyses to younger subjects substantially increased the AUC <strong>of</strong> Model A to<br />

0.78 (0.72 – 0.84) and Model B to 0.83 (p�0.007, 0.78-0.88).<br />

Conclusions: This preliminary risk model is the first in melanoma to<br />

demonstrate that the addition <strong>of</strong> genotypic data and indoor UV exposure<br />

results in a measurable increase in predictive ability when compared to<br />

models comprised only <strong>of</strong> clinical and outdoor UV exposure variables. The<br />

enhanced predictive ability <strong>of</strong> the model (AUC�0.80) when limited to<br />

younger individuals suggests the potential for developing tools to facilitate<br />

targeted screening and prevention strategies in melanoma.<br />

8573 General Poster Session (Board #35G), Sun, 8:00 AM-12:00 PM<br />

Immunological and biological changes during ipilimumab (Ipi) treatment<br />

and their correlation with clinical response and survival. Presenting Author:<br />

Ester Simeone, Unit <strong>of</strong> Medical Oncology and Innovative Therapy, Istituto<br />

Nazionale Tumori Fondazione Pascale, Napoli, Italy<br />

Background: Ipilimumab (Ipi) is approved in the US as first and second line<br />

therapy in patients with metastatic melanoma (MM) and in MM patients<br />

with previous therapy in the EU, based on an overall survival benefit shown<br />

in a phase III study (Hodi, NEJM 2010). To date, no clinical parameter has<br />

been found to be predictive for response to treatment and only few<br />

immunologic changes have been identified as potential candidates. Methods:<br />

From June 2010 to November 2011 we treated in the Expanded Access<br />

Program with Ipi at 3 mg/kg, 95 pre-treated metastatic melanoma patients.<br />

The median age was 58 yrs (range 17-84); 10 pts (10,5%) were stage IIIc<br />

inoperable, 2 pts (2,1%) stage M1a, 4 pts (4,2%) stage M1b, and 79 pts<br />

(83,2%) stage M1c. 30/95 pts had brain metastases and 1/95 spinal cord<br />

metastases. All 95 patients were evaluable for response (DCR �<br />

CR�PR�SD according the irRC), overall survival, safety, including changes<br />

in LDH, CRP (C-reactive protein) and lymphocyte populations<br />

(CD4�,CD4CD25�,FOXP3/T-Reg cells). PBMC and sera were collected at<br />

week 0, 4, 7, 10 and 12. Results: We found a statistical significant<br />

decrease <strong>of</strong> LDH, CRP and FOXP3/T-Reg cells (p�0.0001; �2 and<br />

Mann-Whitney), and an increase <strong>of</strong> lymphocyte count (p�.0001) in the<br />

responders group. These differences were also correlated to survival<br />

(log-rank test). No differences were observed for CD4� and CD4�CD25�<br />

between responders and non-responders (p�0.39;p�0.83; Mann-Whitney).<br />

An ORR <strong>of</strong> 22.1% (1CR�20PR; 95% CI 13.8-30.4) and a DCR at<br />

week 24 <strong>of</strong> 37.9% (36/95; 28.1-47.6, 95% CI) were observed. Median<br />

overall survival was estimated <strong>of</strong> 7.8 months (95%CI:5.0-10.6), with a p<br />

value not evaluable at the moment <strong>of</strong> the analysis due to insufficient<br />

follow-up because <strong>of</strong> long-term survival. Adverse events were registered in<br />

40% (38/95) <strong>of</strong> patients and the most frequent were <strong>of</strong> grade 1 and 2<br />

(pruritus 57.9%; rash 5.3%; thyroditis 5.3%). Conclusions: The decrease<br />

<strong>of</strong> LDH, CRP and T-Reg cells during Ipi treatment suggest these parameters<br />

should be further explored as potential predictive markers for response and<br />

survival. Given the potential clinical utility <strong>of</strong> these findings, these data<br />

warrants further prospective validation in a randomized trial.<br />

8575 General Poster Session (Board #36A), Sun, 8:00 AM-12:00 PM<br />

Evaluation <strong>of</strong> the absolute lymphocyte count as a biomarker for melanoma<br />

patients treated with the commercially available dose <strong>of</strong> ipilimumab<br />

(3mg/kg). Presenting Author: Michael Andrew Postow, Memorial Sloan-<br />

Kettering Cancer Center, New York, NY<br />

Background: Ipilimumab (ipi) has demonstrated an overall survival (OS)<br />

benefit in 2 phase III trials. Only ~30% <strong>of</strong> patients (pts) achieve clinical<br />

benefit, and factors that determine which pts benefit are unclear. For pts<br />

treated with 10mg/kg <strong>of</strong> ipi, we previously reported that an absolute<br />

lymphocyte count (ALC) �1000/�L prior to dose 3 [week (wk) 7] was<br />

associated with improved OS. Since the mean increase in ALC during ipi<br />

treatment correlates with dose, we investigated if ALC is also associated<br />

with improved OS at 3mg/kg, the currently FDA approved, commercially<br />

available dose. Methods: In an IRB-approved analysis, we evaluated<br />

landmark survival data from 137 pts treated with 3mg/kg <strong>of</strong> ipi at Memorial<br />

Sloan-Kettering Cancer Center. 67 pts were treated on an expanded access<br />

protocol (CA 184-045). 70 pts were treated per FDA approval (commercial<br />

ipi) with 4 standard induction doses. These 2 groups were analyzed<br />

separately because some pts in CA 184-045 received re-induction ipi. ALC<br />

was determined at first ipi dose (baseline, wk 1) and at subsequent doses<br />

(wks 4, 7, and 10). Results: Pts treated with 3mg/kg on CA 184-045 with a<br />

wk 7 (prior to dose 3) ALC �1000/�L had significantly improved OS<br />

compared to pts with an ALC at wk 7 �1000/�L (Median OS: not reached<br />

vs. 4.24 mos, p�0.001). This OS difference was also seen for pts treated<br />

with commercial ipi (Median OS: not reached vs. 4.44 mos, p�0.01). This<br />

difference remained significant in a multivariable model accounting for<br />

Karn<strong>of</strong>sky performance score, LDH, M-stage, and number <strong>of</strong> prior therapies<br />

for pts in the CA 184-045 group and commercial ipi group (p�0.01 and<br />

p�0.05, respectively). Baseline ALC �1000/�L was associated with<br />

improved OS (p�0.02) for pts in the commercial ipi group, though<br />

follow-up is limited. Conclusions: At the FDA approved dose <strong>of</strong> ipi, 3mg/kg,<br />

ALC at wk 7 remains significantly associated with improved OS. Our<br />

preliminary finding <strong>of</strong> improved OS for pts treated with commercial ipi<br />

whose pre-treatment baseline ALC �1000/�L deserves confirmation with<br />

longer follow-up and prospective validation. Baseline or on treatment ALC<br />

may be a marker <strong>of</strong> overall prognosis, regardless <strong>of</strong> therapy.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


8576 General Poster Session (Board #36B), Sun, 8:00 AM-12:00 PM<br />

Evaluation <strong>of</strong> chemokine-ligand pathways in pretreatment tumor biopsies<br />

as predictive biomarker <strong>of</strong> response to adoptive therapy in metastatic<br />

melanoma patients. Presenting Author: Davide Bedognetti, National Institutes<br />

<strong>of</strong> Health, Bethesda, MD<br />

Background: Adoptive therapy with tumor infiltrating lymphocytes (TILs)<br />

induces objective responses (OR) in approximately 50% <strong>of</strong> patients with<br />

metastatic melanoma. The recruitment <strong>of</strong> TILs through CXCR3/CCR5ligand<br />

chemokines is believed critical for immune-mediated rejection.<br />

Here, we investigated the predictive role <strong>of</strong> a gene-signature based on<br />

CXCR3/CCR5-ligand chemokine transcripts in pre-treatment melanoma<br />

biopsies, its biological role and its relation with CCR5-�32 polymorphism,<br />

which encodes a protein not expressed on cell surface. Methods: Expression<br />

<strong>of</strong> CXCR3/CXCR3-ligand transcripts (i.e CXCL9, 10, 11) and CCR5/CCR5ligand<br />

transcripts (i.e. CCL3, 4, 5) were assessed in 113 pre-treatment<br />

tumor biopsies from patients enrolled in adoptive therapy trials: 24<br />

patients achieved a complete remission (CR), 34 a partial remission (PR),<br />

and 55 did not respond (NR). Copy number variation and gene expression<br />

pr<strong>of</strong>ile <strong>of</strong> these target genes were assessed in 15 biopsy-derived cell lines.<br />

CCR5-�32 was assessed by sequencing germinal DNA. Results: CXCL9, 10<br />

and 11 and CCL5 clustered together and were selected for hierarchical<br />

clustering analysis based on the mean-centered gene expression values. A<br />

signature characterized by the over-expression <strong>of</strong> these genes was associated<br />

with the likelihood to achieve a clinical response (OR rate: PR�CR:<br />

65% vs 38%, High vs Low, respectively, P�0.015). Neither correlation<br />

between the copy number variation and the gene-expression <strong>of</strong> the<br />

corresponding genes, nor correlation between the transcripts <strong>of</strong> the<br />

investigated genes between tumor biopsies and the matched cell lines was<br />

detected. Transcript expression <strong>of</strong> the target genes did not differ between<br />

CCR5-�32 (n �20) and wild type patients (n�93). Conclusions: Coordinate<br />

over-expression <strong>of</strong> CXCR3/CCR5 ligands in pre-treatment tumor<br />

samples was associated with responsiveness to treatment. However, the<br />

lack <strong>of</strong> correlation between in vivo and ex vivo data suggest the inflammatory<br />

status characterized by the up-regulation <strong>of</strong> these inflammatory<br />

chemokine genes is an in vivo multifactorial phenomenon.<br />

8578 General Poster Session (Board #36D), Sun, 8:00 AM-12:00 PM<br />

Diagnosis <strong>of</strong> melanoma: Importance <strong>of</strong> comparative analysis and “ugly<br />

duckling” sign. Presenting Author: Jean Jacques Grob, Dermatology Department,<br />

Timone Hospital, Aix-Marseille University, Marseille, France<br />

Background: “Ugly duckling” (UD) sign is widely admitted as a major sign<br />

for melanoma (MM) detection. UD is based on the concept <strong>of</strong> intraindividual<br />

comparative analysis, which has never been validated. It is<br />

assumed that, in a given individual, variability <strong>of</strong> nevi is limited and<br />

represented by a few similarity clusters (SC) grouping all nevi sharing the<br />

same pattern, and that a nevus is suspicious when it does not fit with the<br />

SCs <strong>of</strong> this individual (UD). Objective: To validate the concept <strong>of</strong> UD and<br />

SC. Methods: 9 international experts (dermatologists) and 9 novices<br />

(untrained individuals) were blindly tested, using digital images <strong>of</strong> the nevi<br />

at clinical and dermoscospic scale successively in 80 patients, (2,089 nevi<br />

and 7 MM). Concordance was assessed by kappa statistics for UDs, and<br />

B-cubed metrics for SCs. Results: At clinical scale, experts had a better<br />

concordance for UDs (kappa� 0.53, [0.33 - 0.87]) and SCs (B-cubed �<br />

0.65, [0.55, 0.73]), than novices (0.31, [0.03, 0.51] and 0.56 [0.49,<br />

0.64], respectively). Experts identified a mean number <strong>of</strong> 1 UD and 2.7<br />

SCs per patient, and agreed on consensual UD and SCs, whatever their<br />

natural propensity to see a high or a low number <strong>of</strong> UDs and SCs: among the<br />

254 nevi considered as UDs by at least 1 expert, 54 were considered as<br />

such by at least 5, and from them, 20 by all 9 experts. The 20 consensual<br />

UDs included all the MMs (7/7) (sensitivity for MM: 100%). At dermoscopic<br />

scale, concordance tended to be lower for SCs than at clinical scale.<br />

Conclusions: We demonstrated the limited variability <strong>of</strong> nevi patterns in<br />

each patient, the consistency in the recognition <strong>of</strong> UD, and its clinical<br />

relevance, supporting evidence that it is useful for experts and to a lesser<br />

degree to general population to spot the skin lesions with the highest<br />

probability <strong>of</strong> being MM.<br />

Melanoma/Skin Cancers<br />

8577 General Poster Session (Board #36C), Sun, 8:00 AM-12:00 PM<br />

Rapid rational drug targeting <strong>of</strong> Merkel cell polyomavirus (MCV)-positive<br />

Merkel cell carcinoma (MCC) using the survivin inhibitor YM155. Presenting<br />

Author: Reety Arora, Cancer Virology Program, University <strong>of</strong> Pittsburgh<br />

Cancer Institute, Pittsburgh, PA<br />

Background: MCC is an aggressive, chemoresistant skin cancer causing<br />

more deaths each year than chronic myelogenous leukemia. We discovered<br />

a new virus, Merkel cell polyomavirus (MCV), clonally integrated into ~80%<br />

<strong>of</strong> primary and metastatic MCC in 2008. To find therapeutic targets for this<br />

cancer, we examined cellular genes perturbed by MCV infection. Methods:<br />

Digital transcriptome subtraction was used to discover MCV and also to<br />

reveal survivin gene (BIRC5) upregulation in virus-positive tumors. MCV T<br />

antigen knockdown studies in seven MCC lines and large T (LT) transduction<br />

into BJ fibroblasts were used to confirm this. Drug screening was<br />

performed in vitro using Cell-Titer Glo assays in a two stage analysis. In vivo<br />

screening used an MKL-1 (MCV�) MCC NOD-SCIDg mouse xenograft<br />

model with a single three-week treatment round. Results: MCV large T<br />

oncoprotein induces survivin transcription through retinoblastoma protein<br />

sequestration by the LT LXCXE motif. MCV T antigen knockdown results in<br />

nonapoptotic MCC cell death and loss <strong>of</strong> survivin expression. YM155, a<br />

phase II survivin transcription inhibitor, causes MCV� MCC cell necroptosis<br />

associated with autophagy at 1-12 nM EC50. Of 1359 other drugs from<br />

LOPAC and NCI Oncology Set II libraries, only bortezomib had in vitro<br />

potency comparable to YM155. In MKL-1 xenograft studies, mice were<br />

treated with saline, bortezomib or YM155 for three weeks using standard<br />

dosings. Bortezomib did not significantly improve mouse survival (33%)<br />

over saline (24%) during treatment. In contrast, all YM155-treated mice<br />

survived (100%, p�0.001) the 3 week treatment period. Tumors resumed<br />

growth once YM155 treatment was stopped suggesting that YM155 is<br />

cytostatic in vivo rather than cytotoxic. Conclusions: Survivin expression is<br />

induced by MCV LT and is critical to MCV� MCC survival. A survivin<br />

inhibitor, YM155 was nontoxic to mice and cytostatic for MCV� MCC<br />

xenografts. Using genomic technologies, in less than four years, the<br />

primary viral cause for most MCC was discovered, new diagnostic tests<br />

developed and a promising rational drug candidate identified. A cooperative<br />

group trial (E1611) for YM155 and bortezomib in MCC patients is<br />

currently planned.<br />

8579 General Poster Session (Board #36E), Sun, 8:00 AM-12:00 PM<br />

Efficacy and safety <strong>of</strong> the hedgehog pathway inhibitor vismodegib in<br />

patients with advanced basal cell carcinoma (BCC): ERIVANCE BCC study<br />

update. Presenting Author: Aleksandar Sekulic, Mayo Clinic, Scottsdale,<br />

AZ<br />

Background: The ERIVANCE BCC study is the pivotal trial <strong>of</strong> vismodegib<br />

(GDC-0449), a first-in-class small-molecule inhibitor <strong>of</strong> Hedgehog signaling,<br />

for treatment <strong>of</strong> locally advanced (laBCC) and metastatic BCC (mBCC),<br />

for whom there are no other effective therapy options. The study met the<br />

primary endpoint <strong>of</strong> overall response rate by independent review (Sekulic,<br />

Melanoma Res 2011). Here we report a 6-mo update <strong>of</strong> investigatorassessed<br />

(I-A) efficacy and safety endpoints as <strong>of</strong> May 26, 2011. Methods:<br />

This multicenter, international, nonrandomized 2-cohort study enrolled<br />

patients (pts) with laBCC (deemed inoperable or for whom surgery would be<br />

significantly disfiguring), and mBCC pts with RECIST-measurable disease.<br />

Pts received 150 mg oral vismodegib daily. Results: 104 pts (71 laBCC/33<br />

mBCC) enrolled at 31 sites in the US, Europe, and Australia. I-A efficacy<br />

endpoints are shown in the table. One-year survival rate was 77.3% (95%<br />

CI 62.48–92.09%) for mBCC and 93.1% (95% CI 86.49–99.63%) for<br />

laBCC. Adverse events (AEs) in �30% <strong>of</strong> pts were muscle spasms,<br />

alopecia, dysgeusia, weight decrease, fatigue, nausea, and amenorrhea<br />

(33.3%; 2/6 pts). Serious AEs were reported in 32 pts (31%). No<br />

additional fatal AEs were reported since the prior data cut (n�7, 7%; none<br />

considered related to vismodegib). Conclusions: This 6-mo update <strong>of</strong> I-A<br />

efficacy and safety endpoints from the ERIVANCE BCC study supports the<br />

significant clinical benefit <strong>of</strong> vismodegib in both laBCC and mBCC reported<br />

at the primary analysis. Median DOR and PFS increased numerically with<br />

follow-up. The AE pr<strong>of</strong>ile was consistent with the prior data cut. These<br />

results further support the efficacy <strong>of</strong> vismodegib for treatment <strong>of</strong> advanced<br />

BCC.<br />

Parameter<br />

Overall response rate<br />

(ORR), n (%) (95% CI)<br />

Median duration <strong>of</strong> response<br />

(DOR), mo (95% CI)<br />

Median progression-free survival<br />

(PFS), mo (95% CI)<br />

26 Nov 2010 data cut 26 May 2011 data cut<br />

mBCC<br />

(n�33)<br />

15 (45.5)<br />

(28.1–62.2)<br />

(n�15)<br />

12.9<br />

(5.55–12.91)<br />

9.2<br />

(7.39–NE)<br />

laBCC<br />

(n�63)<br />

38 (60.3)<br />

(47.2–71.7)<br />

(n�38)<br />

7.6<br />

(7.43–NE)<br />

11.3<br />

(9.46–16.82)<br />

mBCC<br />

(n�33)<br />

16 (48.5)<br />

(30.8–66.2)<br />

(n�16)<br />

12.9<br />

(5.55–NE)<br />

9.3<br />

(7.39–16.59)<br />

559s<br />

laBCC<br />

(n�63)<br />

38 (60.3)<br />

(47.2–71.7)<br />

(n�38)<br />

NE<br />

(7.62–NE)<br />

12.9<br />

(10.22–NE)<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


560s Melanoma/Skin Cancers<br />

8580 General Poster Session (Board #36F), Sun, 8:00 AM-12:00 PM<br />

A clinical and translational phase II trial <strong>of</strong> sequential axitinib and<br />

carboplatin/paclitaxel in advanced melanoma. Presenting Author: Alain<br />

Patrick Algazi, University <strong>of</strong> California, San Francisco, San Francisco, CA<br />

Background: Several clinical trials adding VEGF signaling inhibitors to<br />

chemotherapy have not demonstrated any benefit over chemotherapy alone<br />

in advanced melanoma potentially due to decreased tumor cell proliferation<br />

induced by VEGF blockade. We tested the hypothesis that sequential<br />

administration <strong>of</strong> axitinib followed by carboplatin and paclitaxel may be<br />

more effective than chemotherapy alone in metastatic melanoma. Methods:<br />

We conducted a prospective phase II trial <strong>of</strong> this combination in previously<br />

treated metastatic melanoma patients. Patients had an ECOG PS 0-1, and<br />

normal organ function. Axitinib 5 mg PO bid was taken on days 1 through<br />

14 <strong>of</strong> each 21-day treatment cycle, and carboplatin (AUC�5) with<br />

paclitaxel (175 mg/m2 ) was administered on day 1 starting with cycle 2.<br />

FLT-PET scans were performed on 6 patients to assess tumor cell<br />

proliferation on days 1, 14, 17, and 20 <strong>of</strong> cycle 1. Results: Treatment has<br />

been well tolerated. The most common grade 3 AEs have been neutropenia,<br />

hypertension, and gastrointestinal events. Grade 4 non-hematologic AEs<br />

have not been observed. 4 <strong>of</strong> 5 patients completing FLT-PET scans to date<br />

showed increases (23% to 92%) in SUV values during the axitinib holiday.<br />

The fifth patient had a single, minimally FLT avid lesion at baseline<br />

(SUV�1.9). In 30 evaluable patients, there have been 4 confirmed PRs, 2<br />

unconfirmed PRs, and 3 patients with 28.6, 29.3, and 29.5% decreases in<br />

tumor size by RECIST. Overall, 19 patients have had SD and 5 have had PD<br />

as the best response. With a median follow-up <strong>of</strong> 8.3 months and 17<br />

patients still active, the median PFS is 6.9 months, and 23 patients (77%)<br />

are still alive. 26 <strong>of</strong> 30 evaluable patients have been wild type for<br />

BRAF-V600E/K mutations. Conclusions: Axitinib followed by carboplatin<br />

and paclitaxel has been well tolerated and effective in a largely BRAF<br />

wild-type metastatic melanoma population. Given the urgent need for<br />

effective therapies in this population, this regimen warrants phase III<br />

testing.<br />

8582 General Poster Session (Board #36H), Sun, 8:00 AM-12:00 PM<br />

Phase I trial <strong>of</strong> extended-dose anti-PD-1 antibody BMS-936558 with a<br />

multipeptide vaccine for previously treated stage IV melanoma. Presenting<br />

Author: Ragini Reiney Kudchadkar, H. Lee M<strong>of</strong>fitt Cancer Center &<br />

Research Institute, Tampa, FL<br />

Background: BMS-936558, a fully human IgG4 anti-PD-1 antibody, has<br />

shown clinical activity in melanoma and renal cancer. Methods: 30 HLA<br />

A*0201 positive patients with unresectable, previously treated stage IV<br />

melanoma received MART-1/gp100/NY-ESO-1 peptides with adjuvant<br />

Montanide ISA 51 injected subcutaneously with BMS-936558 at 1, 3 or<br />

10 mg/kg intravenously every 2 weeks for 24 weeks, followed by BMS-<br />

936558 alone every 3 months. No patient had prior ipilimumab. Endpoints<br />

were toxicity, correlative studies <strong>of</strong> immunity measured by ELISPOT and<br />

T-cell phenotype assays, and response by RECIST. Results: Median age was<br />

62, with 14 men and 16 women. 73% <strong>of</strong> patients had M1c disease. One<br />

patient had dose limiting optic neuritis; one patient had dose limiting grade<br />

3 interstitial pneumonitis. Of 10 patients at 1 mg/kg, 2 had confirmed<br />

partial responses (PR), both one year from initiation <strong>of</strong> study. 13 patients<br />

were treated at the 3 mg/kg cohort; 3 patients did not complete more than 3<br />

doses secondary to progressive disease and one withdrew consent. Of 12<br />

evaluable patients, 3 had confirmed PRs and 2 unconfirmed PRs. In the 10<br />

mg/kg cohort, thus far 6 patients completed one cycle with 2 unconfirmed<br />

PRs and one patient stable. Only 2 <strong>of</strong> the 9 responders after one cycle have<br />

progressed with a median follow-up <strong>of</strong> 7 months. 2/6 patients failing<br />

BMS-936558 responded to ipilimumab. At week 12, patients in all cohorts<br />

had decreased PD-1 on CD4 and CD8 T-cells (p�0.0001), increased CD4<br />

CTLA-4 levels (p�0.01) and dose related decreases in CD8 T-cells whereas<br />

CD4 T-cells increased (both p�0.05). ELISPOT assays showed that<br />

patients at all doses had no increased responses in fresh PBMC to<br />

MART-1/gp100, but showed dose related decreased responses to viral<br />

(CMV, EBV, FLU) peptides over time (p�0.04). Conclusions: BMS-936558<br />

in combination with a peptide vaccine is well tolerated at 1 and 3mg/kg.<br />

Accural to the 10mg/kg cohort is on-going. Responses have been observed<br />

at all dose levels. PD-1 levels on T-cells decreased, and CTLA-4 levels<br />

increased in CD4 T cells in all cohorts. There was a dose-related decrease in<br />

CD8 T-cells and in viral-specific T-cells. Further study <strong>of</strong> BM-936558 is<br />

warranted to determine its optimal dose.<br />

8581 General Poster Session (Board #36G), Sun, 8:00 AM-12:00 PM<br />

Assessment <strong>of</strong> germ-line and somatic alterations in main candidate genes<br />

among patients with multiple primary melanoma. Presenting Author:<br />

Giuseppe Palmieri, Istituto di Chimica Biomolecolare, CNR, Sassari, Italy<br />

Background: We have studied a series <strong>of</strong> patients with multiple primary<br />

melanoma (MPM) for the involvement <strong>of</strong> the key-regulator genes in<br />

susceptibility (CDKN2A) and pathogenesis (BRAF, cKIT, CyclinD1) <strong>of</strong> such<br />

a disease. Methods: Genomic DNA from peripheral blood <strong>of</strong> 63 MPM<br />

patients (54 cases with two primary melanomas, 8 with three, and 1 with<br />

four) were screened for germline mutations in p16CDKN2A and p14CDKN2A genes by automated DNA sequencing. Melanoma families were identified<br />

according to standardized criteria: 9 (14%) patients were classified as<br />

familial cases. Paired synchronous and/or asynchronous MPM tissues<br />

(N�100) from same patients (N�46) were analyzed for somatic mutations<br />

in BRAF gene and FISH-based amplifications in cKIT and CyclynD1 genes.<br />

Results: Overall, 6 (10%) different CDKN2A germline mutations were<br />

identified: 5 in p16CDKN2A and1inp14CDKN2A . The age <strong>of</strong> onset was<br />

significantly lower and the number <strong>of</strong> primary melanomas higher in patients<br />

with mutations. CDKN2A mutations were significantly more frequent in<br />

patients with familial history <strong>of</strong> melanoma (5/9; 56%) compared with<br />

patients without (1/54; 2%) (P�0.001), and in patients with more than<br />

two melanomas (3/9; 33%) compared with patients with only two melanomas<br />

(3/54; 6%) (P�0.012). The debated A148T polymorphism was found<br />

at low level (2/54; 4%) in our series. Regarding genetic alterations at<br />

somatic level, BRAF mutations were identified in 36/100 (36%) primary<br />

melanoma tissues, whereas amplification <strong>of</strong> cKIT and CyclinD1 genes was<br />

observed in 2/88 (2%) and 10/88 (11%) analyzed tissue samples,<br />

respectively. Considering all types <strong>of</strong> genetic events, paired samples<br />

presented a poorly consistent distribution <strong>of</strong> somatic alterations in same<br />

patients (52% consistency). Conclusions: Coexistence <strong>of</strong> MPM and familial<br />

recurrence <strong>of</strong> melanoma as well as the presence <strong>of</strong> more than two<br />

melanomas seem to be strong indications to address patients to CDKN2A<br />

mutational screening. The low consistency in genetic patterns <strong>of</strong> primary<br />

tumors from the same patients provide additional evidence that pathogenetic<br />

mechanisms <strong>of</strong> melanomagenesis are heterogeneous and molecularly<br />

different cell types may be generated in multiple primary melanoma.<br />

8583 General Poster Session (Board #37A), Sun, 8:00 AM-12:00 PM<br />

Surgery for patients receiving ipilimumab: Safety pr<strong>of</strong>ile and immunological<br />

insights. Presenting Author: David E. Gyorki, Memorial Sloan-Kettering<br />

Cancer Center, New York, NY<br />

Background: Ipilimumab (ipi), a monoclonal antibody blocking CTLA-4,<br />

demonstrated survival benefit in metastatic melanoma in two randomized<br />

controlled trials. With its approval by the FDA, the use <strong>of</strong> this immunotherapy<br />

is increasing. The surgical safety <strong>of</strong> ipi has not been reported.<br />

Methods: From our prospective database, we identified patients undergoing<br />

surgery within 30 days <strong>of</strong> receiving a dose <strong>of</strong> ipi or on maintenance ipi<br />

between October 2007 and August 2011. Surgical toxicity was graded 1-5<br />

by the Clavien classification. In 10 patients matched blood and tumor<br />

infiltrating lymphocytes were harvested. Phenotype <strong>of</strong> T cell subsets were<br />

analyzed by flow cytometry. Results: 23 patients were identified who<br />

underwent 34 operations a median <strong>of</strong> 27 weeks after initiation <strong>of</strong> ipi<br />

(1-123 weeks). Indications for surgery were symptomatic, progressive, or<br />

isolated persistent disease in 52%, 39% and 9% respectively. Subcutaneous<br />

resections were the most frequent, followed by intra-abdominal and<br />

nodal procedures (Table). Grade 1 wound complications were seen in 17%<br />

<strong>of</strong> patients. One patient had a Grade 2 wound complication. No Grade 3-5<br />

complications were seen. With a median follow up from starting ipi <strong>of</strong> 86<br />

weeks, 3 patients have no evidence <strong>of</strong> disease, 10 are alive with disease<br />

and 10 have died <strong>of</strong> disease. Analysis <strong>of</strong> the T cell infiltrate and peripheral<br />

blood from 10 patients with progressive or symptomatic disease shows an<br />

elevated % <strong>of</strong> CD4 � FOXP3 � T regulatory cells and a 2.8 fold reduction in<br />

CD8 � / CD4 � FOXP3 � T regulatory ratio in the tumor compared to blood<br />

(p�0.02). Conclusions: Results from this single-center experience suggest<br />

that surgery is safe in patients receiving ipi. Immune modulation caused by<br />

CTLA-4 blockade does not appear to impact wound healing, even in the<br />

bowel. In carefully selected patients metastectomy may be appropriate for<br />

breakthrough metastases. The high percentage <strong>of</strong> T regulatory cells and low<br />

T effector cells in the progressive tumors suggests a mechanism <strong>of</strong> immune<br />

escape.<br />

Surgical indications.<br />

Metastectomy site n (%)<br />

Isolated<br />

disease<br />

Progressive<br />

disease<br />

Symptomatic<br />

disease<br />

Subcutaneous 12 (35%) 2 5 5<br />

Intra abdominal 11 (32%) 0 7 4<br />

Brain 5 (15%) 0 0 5<br />

Nodal 3 (9%) 1 2 0<br />

Other 3 (9%) 0 0 3<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


8584 General Poster Session (Board #37B), Sun, 8:00 AM-12:00 PM<br />

Correlation <strong>of</strong> BRAF and NRAS mutation status with tumor characteristics<br />

and treatment outcomes in melanoma patients with brain metastasis.<br />

Presenting Author: Eugene J. Koay, University <strong>of</strong> Texas M. D. Anderson<br />

Cancer Center, Houston, TX<br />

Background: The management <strong>of</strong> melanoma has evolved rapidly with the<br />

identification <strong>of</strong> activating mutations in the majority <strong>of</strong> patients (pts). We<br />

reviewed characteristics and outcomes <strong>of</strong> molecularly characterized melanoma<br />

pts with brain metastasis (BM) to help design and interpret future<br />

clinical trials. Methods: We reviewed clinical and pathologic features <strong>of</strong><br />

melanoma pts with known BRAF and NRAS mutation status and BM. Pt<br />

demographics, intra- (IC) and extra-cranial (EC) tumor characteristics, and<br />

IC/EC disease treatments were correlated to BM treatment outcomes (local<br />

and distant brain control [LC, DC]) and overall survival (OS). Univariate and<br />

multivariate analyses were performed to identify significant associations<br />

between mutation status and outcome. Results: We identified 296 pts with<br />

melanoma BM and known BRAF/NRAS mutation status diagnosed from<br />

2005 to 2011. Mutation prevalence was BRAF 57%, NRAS 17%, and<br />

wild-type for both genes (WT) 26%. The initial treatments given for BM<br />

were surgery/radiosurgery (SRS) 60%, surgery/SRS � whole brain radiation<br />

(WBRT) 7%, WBRT alone 19%, systemic therapy alone 9%, and no<br />

treatment 5%. Mutation status was not significantly associated with sex,<br />

performance status, EC disease status, number <strong>of</strong> brain lesions, or BM<br />

treatment. Pts with mutations were younger (p�0.0001) and more likely to<br />

have symptomatic BM (p�0.0003) than WT pts. In univariate analysis, BM<br />

treatment strategy (p�0.001) and mutation status predicted for poor LC,<br />

with 6 month LC rates <strong>of</strong> 70% for any mutation and 88% for WT<br />

(HR�1.86, p�0.048). Compared to systemic agents alone, use <strong>of</strong> radiation<br />

(SRS or WBRT) improved LC for pts with a mutation (HR 0.23,<br />

p�0.0006). Subsequent multivariate analysis identified mutation status<br />

and radiation treatment as independent predictors <strong>of</strong> LC (HR 2.5 and 0.25,<br />

respectively). Mutation status did not predict for DC or OS. Conclusions:<br />

The presence <strong>of</strong> BRAF and NRAS mutations predicts worse LC following<br />

conventional treatments for melanoma BM. Radiation may improve LC in<br />

pts with a mutation. This data suggest a role for combining radiation with<br />

targeted therapies in melanoma pts with activating BRAF or NRAS<br />

mutations in the treatment <strong>of</strong> BM.<br />

8586 General Poster Session (Board #37D), Sun, 8:00 AM-12:00 PM<br />

<strong>Clinical</strong> experience with atypical spitzoid tumors in patients younger than<br />

age 18: Does fluorescence in situ hybridization predict lymph node<br />

metastasis? Presenting Author: Eric J. Stanelle, Memorial Sloan-Kettering<br />

Cancer Center, New York, NY<br />

Background: Determining the malignant potential <strong>of</strong> atypical spitzoid<br />

melanocytic proliferations can be diagnostically challenging, and many<br />

patients are therefore managed as if they had melanoma. However, few<br />

studies focus specifically on atypical spitzoid tumors (AST). Further,<br />

cytogenetic analysis using fluorescence in situ hybridization (FISH) has<br />

been used to determine malignant potential. We reviewed our institutional<br />

experience to determine staging and clinical outcomes, and the correlation<br />

between FISH findings and regional nodal positivity in patients with AST.<br />

Methods: With IRB approval, we retrospectively reviewed all patients aged<br />

�18 years treated for AST from 1981-2011 to determine staging variables,<br />

outcome, and the results <strong>of</strong> 4-probe FISH assay. A total <strong>of</strong> 44 patients with<br />

a median age <strong>of</strong> 11.5 years were identified. All pathology was re-reviewed<br />

by a single dermatopathologist. Staging was based on 2010 AJCC criteria.<br />

Correlations were determined using either the Pearson or Spearman<br />

correlation coefficient. Results: Median lesion thickness was 2.8 mm<br />

(range: 0.55-12) and eight (18%) lesions met spitzoid melanoma criteria.<br />

Median followup was 4.1 years for all patients (5.5 years for spitzoid<br />

melanomas). Thirty-nine patients (89%) underwent sentinel lymph node<br />

biopsies (SLNB) and 15 (38%) were positive. Lymph node (LN) positivity<br />

correlated with tumor thickness (p�0.002), stage (p�0.001), and mitotic<br />

rate (p�0.05). FISH was available for 17/39 patients who had SLNB;<br />

sensitivity and specificity for LN metastases were 50% and 54%, respectively.<br />

Of 15 patients with a positive SLNB, 12 underwent completion LN<br />

dissection, three <strong>of</strong> which were positive for 1, 2, and 3 additional nodes,<br />

respectively. There were no recurrences or disease-related deaths.<br />

Conclusions: Traditional indicators <strong>of</strong> thickness and stage predicted nodal<br />

involvement in pediatric patients with AST. However, FISH results did not<br />

predict nodal status and should not be used to guide management. With<br />

100% disease specific survival and no recurrences, AST should be<br />

considered a separate entity from melanoma and complete excision may be<br />

sufficient therapy.<br />

Melanoma/Skin Cancers<br />

561s<br />

8585 General Poster Session (Board #37C), Sun, 8:00 AM-12:00 PM<br />

Metastatic basal cell carcinoma: Differences in survival by site <strong>of</strong> spread.<br />

Presenting Author: Margaret Elizabeth McCusker, Genentech, South San<br />

Francisco, CA<br />

Background: Basal cell carcinoma (BCC), the most common skin cancer,<br />

rarely metastasizes. Consequently, the epidemiology <strong>of</strong> metastatic BCC<br />

(mBCC) is poorly characterized. The largest published mBCC review<br />

includes 170 cases reported from 1894–1980 (von Domarus H, Stevens P.<br />

J Am Acad Dermatol 1984;10:1043-60). Targeted therapies with hedgehog<br />

signaling pathway inhibitors, such as vismodegib, are currently being<br />

developed to treat this rare disease. The objective <strong>of</strong> the current analysis<br />

was to provide an updated description <strong>of</strong> the epidemiology and survival<br />

outcomes for mBCC. Methods: A PubMed literature search was conducted<br />

for mBCC case reports published in English during 1981–2011. mBCC<br />

cases that met the following criteria were evaluated: primary BCC located<br />

on skin; primary tumor and metastasis confirmed by pathology, and<br />

metastasis was not the result <strong>of</strong> direct tumor spread. Only cases with<br />

survival data giving dates (month and year) or durations were included in<br />

the analysis. Differences between groups were assessed with t tests and �2 tests as appropriate. Survival was assessed with Kaplan–Meier (K-M)<br />

methods. Results: We identified 101 mBCC cases that met the selection<br />

criteria; 38 patients (38%) had lymph node-only disease (LD) and 63<br />

(62%) had distant metastases (DM). In both groups, males predominated.<br />

DM patients were younger at mBCC diagnosis (mean 59 vs 66 y for LD).<br />

Among 96 cases with treatment data for metastatic disease, more DM<br />

patients received chemotherapy (25% vs 9% for LD), but more LD patients<br />

underwent surgery (85% vs 52% for DM). Survival duration ranged from 0<br />

to 120� months in all patients. The overall 1-y probability <strong>of</strong> survival after<br />

mBCC diagnosis was 75.7% (95% CI 67.2–84.2%), with lower survival in<br />

DM patients (69.1%; 95% CI 57.5–80.7%) vs LD patients (86.5%; 95%<br />

CI 75.5–97.5%). Conclusions: Patients with LD and DM mBCC may have<br />

different clinical courses and outcomes. Based on published case reports,<br />

DM patients were younger at mBCC diagnosis and had a lower 1-y survival<br />

probability vs LD patients. To our knowledge, these are the first K-M<br />

survival estimates reported for an mBCC case series <strong>of</strong> this size. These data<br />

help to provide a historical context for novel mBCC therapies under<br />

development, such as vismodegib.<br />

8587 General Poster Session (Board #37E), Sun, 8:00 AM-12:00 PM<br />

A phase II, multicenter, open-label study <strong>of</strong> YM155 plus docetaxel in<br />

subjects with stage III (unresectable) or stage IV melanoma. Presenting<br />

Author: Joyce Leta Steinberg, Astellas Pharma Global Development,<br />

Deerfield, IL<br />

Background: Survivin is a microtubule-associated protein implicated in<br />

both preservation <strong>of</strong> cell viability and regulation <strong>of</strong> mitosis in tumor cells. It<br />

is over-expressed in melanoma, breast, and non-Hodgkin’s lymphoma.<br />

YM155 is a first in class survivin inhibitor. Methods: The study had 2 parts:<br />

<strong>Part</strong> 1 established the dose <strong>of</strong> docetaxel that was tolerable in combination<br />

withYM155 at 5 mg/m2 /day continuous infusion over 168 hours q 3 weeks.<br />

<strong>Part</strong> 2 utilized the dose <strong>of</strong> docetaxel established in <strong>Part</strong> 1 to further<br />

evaluate the tolerability and activity <strong>of</strong> the combination. The primary<br />

endpoint was 6-month progression-free survival (PFS). Secondary endpoints<br />

were overall response rate, 1-year overall survival (OS), time from<br />

first response to progression, clinical benefit rate, time to response, and<br />

safety. Results: 64 patients with metastatic melanoma were treated with<br />

docetaxel followed by continuous infusion YM155. 7 patients were treated<br />

with 100mg/m2 <strong>of</strong> docetaxel and 57 patients were treated with 75mg/m2 <strong>of</strong><br />

docetaxel. Median age was 59, with 44 men and 20 women treated.<br />

6-month PFS per Independent Review Committee (IRC) was 34.8% (95%<br />

CI 21.3 – 48.6%). Overall objective response rate per IRC was 12.5%, with<br />

no complete responses (CR) and 8 patients with partial responses (PR). The<br />

Stable disease (SD) rate was 51.6%, leading to a clinical benefit rate (CR �<br />

PR � SD) <strong>of</strong> 64.1%. Estimated 1-year overall survival is 50.5%. 87.5% <strong>of</strong><br />

patients experienced a Grade 3 (G3) or Grade 4 (G4) event attributable to<br />

either YM155 or docetaxel. The clinically pertinent G3 or 4 toxicities<br />

occurring in greater than 5% <strong>of</strong> patients treated included neutropenia<br />

(59.4%), febrile neutropenia (12.5%), mucositis (9.4%), fatigue (7.8%),<br />

diarrhea (6.3%), and dehydration (6.3%). There were 3 deaths on study, all<br />

attributable to disease progression. Conclusions: YM155 is a novel agent<br />

that shows modest activity when combined with docetaxel in patients with<br />

melanoma. YM155 was generally well tolerated, but the pre-determined<br />

primary endpoint for efficacy was not achieved.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


562s Melanoma/Skin Cancers<br />

8588 General Poster Session (Board #37F), Sun, 8:00 AM-12:00 PM<br />

KIT, NRAS, BRAF, and PTEN alterations in acral lentiginous melanomas.<br />

Presenting Author: Abdlsattar Zebary, Department <strong>of</strong> Oncology-Pathology,<br />

Karolinska Institute, Stockholm, Sweden<br />

Background: Acral lentiginous melanoma (ALM) accounts for ~5% <strong>of</strong> all<br />

melanomas. ALMs are <strong>of</strong>ten located on palms, soles and under nails.<br />

Patients with ALM are significantly older at diagnosis and have a poorer<br />

prognosis than patients with other types <strong>of</strong> cutaneous melanoma. We<br />

analyzed a large series <strong>of</strong> ALMs from Swedish patients to better define the<br />

frequency <strong>of</strong> KIT, BRAF, NRAS, and PTEN mutations in ALM. The effect <strong>of</strong><br />

mutation status on tumor and patients characteristics was also studied.<br />

Methods: A total <strong>of</strong> 77 primary ALMs and 8 paired metastases were<br />

analyzed. Laser capture microdissection was used to dissect tumor cells<br />

from formalin-fixed paraffin-embedded tissue. Tumors were screened for<br />

mutations in the KIT (exons 11, 13, 17 and 18), NRAS (exons 1 and 2),<br />

BRAF (exons 11 and 15) and PTEN (exons 1, 3-6, 10, 11 and 12) genes by<br />

direct sequencing. Results: There were 42 females and 35 males with a<br />

median age at diagnosis <strong>of</strong> 71 years. The most common location was the<br />

feet (60.5%), followed by subungual sites (34.2%). Overall, mutations in<br />

KIT, NRAS and BRAF were detected in 11.8%, 14.5% and 16.0% <strong>of</strong> the<br />

ALMs, respectively. In no case were KIT, NRAS and BRAF mutations<br />

detected in the same tumor. For 8 primary ALMs the corresponding<br />

metastases were available for analysis. In five metastases the same KIT<br />

(n�3) or BRAF (n�2) mutation was detected as in the primary ALMs,<br />

suggesting that the KIT and BRAF mutations had occurred before metastasis.<br />

BRAF and NRAS mutations were significantly more common in females<br />

(P�0.044). ALMs with NRAS and BRAF mutations were more commonly<br />

located on the feet compared with KIT mutated ALMs which were more<br />

frequent on subungual sites (P�0.041). Other clinicopathological features<br />

such as age at diagnosis, thickness, ulceration, histological subtype and<br />

Clark’s level showed no significant correlation with the mutation status. A<br />

subset <strong>of</strong> 25 tumors was evaluated for mutations in the PTEN tumor<br />

suppressor gene. One tumor was found to carry a nonsense mutation<br />

(W111X). To our knowledge, this is the first description <strong>of</strong> a PTEN mutation<br />

in ALM. Conclusions: Our results confirm the presence <strong>of</strong> KIT, NRAS and<br />

BRAF mutations in ALM. KIT, NRAS and BRAF mutation status associated<br />

significantly with anatomical site. PTEN mutations seem to be rare in<br />

ALMs.<br />

8590 General Poster Session (Board #37H), Sun, 8:00 AM-12:00 PM<br />

The impact <strong>of</strong> immune status on clinical outcomes in patients with Merkel<br />

cell carcinoma. Presenting Author: Sean Donovan, Mayo Clinic, Jacksonville,<br />

FL<br />

Background: Merkel Cell Carcinoma (MCC) is an aggressive cutaneous<br />

malignancy <strong>of</strong> neuroendocrine origin associated with immunosuppression<br />

presumably through infection with Merkel Cell Polyomavirus present in<br />

�80% <strong>of</strong> patients (pts). The impact <strong>of</strong> immune status on clinical outcomes<br />

in pts with MCC is unknown. The primary objective <strong>of</strong> this study was to<br />

compare clinical characteristics and outcomes <strong>of</strong> pts with MCC who are<br />

immunosuppressed (ISP) versus non-immunosuppressed (non-ISP).<br />

Methods: We performed a retrospective chart review on pts with MCC<br />

diagnosed at the Mayo Clinic between 1981 and 2009. A dermatopathologist<br />

confirmed all cases. ISP was defined as pts diagnosed with chronic<br />

lymphocytic leukemia, HIV, solid organ transplant recipients, or chronic<br />

immunosuppressive medication. The association between ISP status and<br />

overall survival was summarized using the hazard ratio (HR) and 95%<br />

confidence interval (CI) estimated from a Cox regression model. Results: Of<br />

the 268 pts identified and included in the study, 38 (14%) were ISP. We<br />

found no differences in age, tumor size, tumor location, stage <strong>of</strong> disease, or<br />

recurrence rate in ISP vs Non-ISP. Among pts who had Stage 3-4 disease,<br />

there was no difference in the size <strong>of</strong> the primary between groups. Among<br />

pts with Stage 1-2 disease, ISP status was not significantly associated with<br />

poorer survival (HR 1.5, 95% CI 0.7-3.3). However, among pts with Stage<br />

3-4 disease, ISP pts had significantly poorer survival compared to non-ISP<br />

pts (HR 2.7, 95% CI 1.2 - 6.2). Conclusions: Baseline clinical characteristics<br />

<strong>of</strong> pts with MCC do not differ based on immunosuppression, and<br />

outcomes do not differ in pts in regards to immunosuppression in early<br />

stage MCC (Stage 1-2). However, in pts with Stage III-IV MCC, ISP pts have<br />

a worse clinical outcome suggesting that metastatic MCC either behaves<br />

more aggressively in ISP pts either through intrinsic differences in the<br />

biology <strong>of</strong> the tumor or improved immune evasion in ISP pts. These results<br />

should be cautiously interpreted given the small number (n�12) <strong>of</strong><br />

immunosuppressed pts with advanced stage MCC. The authors will update<br />

their data with an additional 58 patients by the date <strong>of</strong> presentation.<br />

8589 General Poster Session (Board #37G), Sun, 8:00 AM-12:00 PM<br />

TILs in metastatic melanoma tumors: A biomarker for immunotherapy?<br />

Presenting Author: Sunandana Chandra, New York University School <strong>of</strong><br />

Medicine, New York, NY<br />

Background: Increased tumor infiltrating lymphocytes (TILs) in primary (P)<br />

and locoregional melanoma tissue correlate with improved clinical outcome.<br />

Our recent data have suggested that matrix metalloproteinase 23<br />

(MMP 23) expression (exp) in P result in lower prevalence <strong>of</strong> TILs and<br />

correlate with poor clinical outcome. On this basis, we examined P and<br />

metastatic (M) melanoma tissues to assess for concordance between the<br />

presence <strong>of</strong> TILs, MMP 23 protein levels and clinical response(resp) to<br />

anti-cytotoxic T-lymphocyte antigen 4 (CTLA4) therapy (tx). Methods: 21<br />

melanoma patients (pts) with M specimens were analyzed. 17 matched P<br />

specimens were also evaluated. Immunohistochemical (IHC) staining for<br />

TILs <strong>of</strong> the pre-anti-CTLA4 specimens were conducted and confirmed by 2<br />

pathologists. IHC TILs were graded- 2�: �10% TILs present in multiple<br />

foci in both peri- and through the tumor; 1�: 1-10% TILs present in one or<br />

more foci in the tumor and predominantly peri-tumor; 0: no TILs were<br />

present or if the lymphocytes did not infiltrate the tumor. TILs in P and M<br />

were analyzed for concordance and potential for predictability <strong>of</strong> resp to<br />

anti-CTLA4 tx. Staining to identify lymphocyte subtypes and MMP 23 exp<br />

in M is being completed. Results: 20 pts received anti-CTLA4 tx. M<br />

analysis- 6 pts with 0 TILs in M (5 no response [NR], 1 partial response[PR]);<br />

8 pts with 1-2� TILs in M (1 complete response [CR], 5 PR, 2<br />

progressive disease [PD]); 6 pts with 2� TILs inM(3CR,2PR,1PD). 1 pt<br />

with 2� TILs in M resected, no tx and 4 years disease free. TILs present in<br />

13 P, absent in 4 P and not evaluable in 4 pts with unknown P melanoma.<br />

MMP 23 protein scores in P (range 2-4) correlated with melanoma<br />

recurrence. MMP 23 exp in M will be reported. Conclusions: TILs in P do not<br />

appear to correlate with TILs in M or predict for resp to anti-CTLA4 tx. TILs<br />

in M may be an indicator <strong>of</strong> responsiveness to anti-CTLA4 tx. Identification<br />

<strong>of</strong> the type <strong>of</strong> M TIL subsets may further refine tx recommendations.<br />

8591 General Poster Session (Board #38A), Sun, 8:00 AM-12:00 PM<br />

Vemurafenib (V) in BRAF V600E metastatic melanoma (mM): Analysis <strong>of</strong> 507<br />

patients (pts) enrolled in the French temporary authorization for use (ATU).<br />

Presenting Author: Celeste Lebbe, Saint-Louis Hospital, Paris, France<br />

Background: V, a selective BRAF inhibitor, significantly improves OS in BRAF<br />

mutated mM. ATU is an exceptional measure making available drugs that have<br />

not yet been granted a Marketing Authorisation. We provide demographic data <strong>of</strong><br />

pts treated by V in the ATU. Methods: V 960 mg BID was given to pts with<br />

unresectable stage IIIC or IV BRAF V600E mM. Genotyping was done on the<br />

national network <strong>of</strong> molecular genetics platforms funded by the Institut National<br />

du Cancer. Data were prospectively collected. Results: From Apr 2011 to Jan<br />

2012, 83 sites enrolled 507 pts. 80% were treated by oncodermatologists. Pts<br />

characteristics at baseline are summarized below. Safety and efficacy data are<br />

being evaluated. Conclusions: Around 2 out <strong>of</strong> 3 patients with a BRAF mutated<br />

mM were enrolled in France in the ATU highlighting a large access to BRAF<br />

genotyping and to V. Demographic data differs from literature and clinical trials<br />

for: site <strong>of</strong> primary melanoma (reverse trunk/extremity ratio) and inclusion <strong>of</strong> pts<br />

with brain metastasis or PS�2 (excluded in CT). A high number <strong>of</strong> pts had risk<br />

factors for NMSC emphasizing the importance <strong>of</strong> the communication between<br />

oncologists and dermatologists for managing V therapy.<br />

n� 507<br />

Males (%) 54.8<br />

Median age (years), range 57 (22-92)<br />

ECOG PS (%)<br />

0-1<br />

2-3<br />

4<br />

Site on primary melanoma (%)<br />

Extremity<br />

Trunk<br />

Head /neck<br />

Occult<br />

Others<br />

Mucosal<br />

86.4<br />

9.7<br />

4<br />

44<br />

34.7<br />

11.5<br />

8.5<br />

1<br />

0.2<br />

Current AJCC stage IV (%) 97<br />

Mean time from diagnosis <strong>of</strong> stage IV or unresectable stage III C (months) (range) 4 (0-261)<br />

Median number <strong>of</strong> metastatic sites, (range) 3 (1-7)<br />

Brain metastasis (%) 31.9<br />

Number <strong>of</strong> previous therapies for mM ( %)<br />

0<br />

1<br />

>2<br />

Main previous therapies (%)<br />

Dacarbazine<br />

Fotemustine<br />

Ipilimumab<br />

Temozolamide<br />

Previous tyrosine kinase inhibitors (n)<br />

BRAF inh<br />

MEK inh<br />

Epithelial proliferations at baseline (%)<br />

Actinic keratosis<br />

Basal cell carcinoma<br />

Squamous cell carcinoma<br />

Risk factors for nonmelanoma skin cancer (NMSC) (%)<br />

Previous radiation<br />

Past history <strong>of</strong> NMSC<br />

BRAF V600E identification method (%)<br />

Sanger sequencing<br />

Pyrosequencing<br />

Allele-specific oligonucleotide PCR<br />

High-resolution melt<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.<br />

31.7<br />

61.8<br />

6.5<br />

45.1<br />

17.9<br />

10.6<br />

6.7<br />

4<br />

7<br />

13.8<br />

3.1<br />

1.5<br />

12.4<br />

14.4<br />

43.4<br />

14.8<br />

12.8<br />

12.4


8592 General Poster Session (Board #38B), Sun, 8:00 AM-12:00 PM<br />

Molecular pr<strong>of</strong>iling and comparative genomic hybridization analysis in<br />

human melanoma cell lines and identification <strong>of</strong> BRAF amplification in a<br />

PLX4032 acquired resistance cell line. Presenting Author: Erika Maria Von<br />

Euw, University <strong>of</strong> California, Los Angeles School <strong>of</strong> Medicine/Translational<br />

Oncology Research Laboratory, Los Angeles, CA<br />

Background: Melanoma is the most aggressive form <strong>of</strong> skin cancer. Its<br />

management is evolving rapidly due to an improved understanding <strong>of</strong> the<br />

molecular heterogeneity and the development <strong>of</strong> effective, personalized<br />

targeted therapies. PLX4032 is a BRAF inhibitor that induces tumor<br />

response in patients with BRAF mutation whose response is limited due to<br />

acquired resistance. We used comprehensive microarray and genomic<br />

analysis to molecularly characterize a panel <strong>of</strong> melanoma cell lines with the<br />

goal <strong>of</strong> identifying new molecular subgroups. To better understand the<br />

mechanisms <strong>of</strong> acquired resistance to PLX4032, we included two cell lines<br />

that were conditioned in vitro to acquire resistance. Methods: Using<br />

microarray, we studied 52 melanoma cell lines for mRNA expression and<br />

performed array CGH using genome microarrays. Data analysis was done<br />

using Rosetta Resolver and Agilent Analytics 4.0 s<strong>of</strong>tware. Results: Microarray<br />

analysis suggests melanoma is comprised <strong>of</strong> at least two distinct<br />

molecular groups. One group has a differentiated melanocyte phenotype<br />

and the other has an expression signature characteristic <strong>of</strong> progenitor-like<br />

cells. The progenitor-like group expresses SOX9, WNT5A and WNT5B and<br />

also has the lowest relative expression <strong>of</strong> MITF. The most differentiated<br />

group had the highest MITF and SOX5 levels, while the progenitor-like cell<br />

lines have decreased expression. MITF appears to be a strong candidate<br />

marker for this group. Positive correlation exists between MITF expression<br />

levels and gene copy number, as almost all <strong>of</strong> the MITF samples amplified<br />

by CHG analysis are also overexpressed. A third group shows strong<br />

expression <strong>of</strong> SOX5, SOX10 and Nestin. One <strong>of</strong> the most important findings<br />

is M14-R cell line, conditioned to acquire PLX4032 resistance, has a focal,<br />

high level amplification <strong>of</strong> BRAF (Log2 ratio � 3, 8-fold amplification)<br />

when compared with the parental cell line, which is extremely sensitive to<br />

PLX4032. Conclusions: These results afford new insight into the potential<br />

pathogenesis and classification and provide a greater understanding <strong>of</strong><br />

melanoma. BRAF amplification could be a novel mechanism <strong>of</strong> resistance<br />

to PLX4032.<br />

8594 General Poster Session (Board #38D), Sun, 8:00 AM-12:00 PM<br />

A phase IB study <strong>of</strong> lenvatinib (E7080) in combination with temozolomide<br />

for treatment <strong>of</strong> advanced melanoma. Presenting Author: David S. Hong,<br />

University <strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: Lenvatinib is an oral tyrosine kinase inhibitor targeting<br />

VEGFR1-3, FGFR1-4, RET, KIT and PDGFR�. In phase I studies <strong>of</strong><br />

lenvatinib partial responses were observed in melanoma as well as thyroid,<br />

endometrial, and renal cancers. Methods: Patients (pts) with stage 4 or<br />

unresectable stage 3 melanoma were treated in sequential cohorts <strong>of</strong><br />

escalating doses <strong>of</strong> lenvatinib by continuous once daily dosing and<br />

temozolomide (TMZ) days 1-5 every 28 days. Cohort 1: lenvatinib 20mg,<br />

TMZ 100 mg; cohort 2: lenvatinib 24 mg, TMZ 100 mg; cohort 3:<br />

lenvatinib 24 mg, TMZ 150 mg. Adverse events were recorded according to<br />

CTCAE v3.0 and tumor response assessed according to RECIST 1.0.<br />

Results: 32 pts were treated: cohort 1: 6 pts, cohort 2: 4 pts, cohort 3: 22<br />

pts. Demographics: median age: 58; males: 63%. Dose limiting toxicities<br />

occurred in 0/6 pts in cohort 3 leading to cohort expansion to a total <strong>of</strong> 22<br />

pts to enable more extensive assessment <strong>of</strong> toxicity and a preliminary<br />

estimate <strong>of</strong> efficacy. The most common treatment related adverse events in<br />

the cohort 3 were fatigue: 50% (gr3: 9%), hypertension: 50% (gr3: 9%),<br />

proteinuria: 50% (no gr3), vomiting: 46% (no gr3), hypothyroidism: 46%<br />

(no gr3), anorexia: 41% (no gr3), nausea: 41% (no gr3), diarrhea: 36%<br />

(gr3: 5%), thrombocytopenia: 32% (no gr3). No treatment-related gr4<br />

events were reported in these categories. The best overall responses per<br />

RECIST were partial responses: 5/22 (23%) for cohort 3 pts and 6/32<br />

(19%) for all pts treated at all dose levels. Six-month progression free<br />

survival (PFS) rate was 37% and median PFS was 5.4 months. PK/PD<br />

analysis and correlation <strong>of</strong> response with genetic mutational status (BRAF<br />

mutations in 31% pts; NRAS mutations in 19% pts) will be presented.<br />

Conclusions: Lenvatinib 24 mg once daily in combination with TMZ 150 mg<br />

days1-5 every 28 days was given without apparent worsening <strong>of</strong> either<br />

lenvatinib or TMZ related toxicities. Modest clinical efficacy for this<br />

combination was observed.<br />

Melanoma/Skin Cancers<br />

563s<br />

8593 General Poster Session (Board #38C), Sun, 8:00 AM-12:00 PM<br />

Gene expression pr<strong>of</strong>iling <strong>of</strong> whole blood in ipilimumab-treated patients for<br />

identification <strong>of</strong> potential biomarkers <strong>of</strong> immune-mediate gastrointestinal<br />

adverse events. Presenting Author: Vafa Shahabi, Bristol-Myers Squibb,<br />

Princeton, NJ<br />

Background: Treatment with ipilimumab, a fully human anti-CTLA-4<br />

antibody approved for treatment <strong>of</strong> metastatic melanoma (MM), is associated<br />

with a number <strong>of</strong> immune-mediated Adverse Events (imAEs) such as<br />

colitis and skin rash. Predictive biomarkers that can help identify patients<br />

(pts) who might experience these imAEs could enhance the management <strong>of</strong><br />

these toxicities. Methods: Gene expression pr<strong>of</strong>iling (using Affymetrix gene<br />

chip HT-HG-U133A) was performed on the whole blood samples from 162<br />

MM pts at baseline, 3 and 11 weeks after the start <strong>of</strong> ipilimumab treatment<br />

in two phase II clinical trials (CA184004 and -007). Overall, 49 pts<br />

experienced Grade 2 or higher GI-imAE (G2�) during the course <strong>of</strong><br />

treatment. A repeated measures ANOVA was used to evaluate the differences<br />

in mean expression levels between the two groups and at the three<br />

time points. Uncorrected p-value <strong>of</strong> 0.05 was used as a cut<strong>of</strong>f for this<br />

analysis. Results: In baseline samples, 27 probe sets showed differential<br />

mean expression (� 1.5 fold, p � 0.05) between G2� pts and others. Most<br />

<strong>of</strong> these genes belonged to three functional categories: immune system,<br />

cell cycle or intracellular trafficking. Changes in gene expression over time<br />

were also characterized. In the G2� pts, 58 and 247 genes had a � 1.5<br />

fold (p � 0.05) change in expression from baseline to week 3 and 11<br />

post-treatment, respectively, compared to 17 and 73 in other pts. In<br />

particular, the on-treatment increases <strong>of</strong> the expression <strong>of</strong> CD177 and<br />

CEACAM1, two neutrophil activation markers, were closely associated with<br />

G2� GI-imAE, suggesting a possible role <strong>of</strong> neutrophils in ipilimumab<br />

associated GI-imAEs. In addition, the expression <strong>of</strong> several Ig genes<br />

increased over time, with higher increases in the G2� pts. These observations<br />

were reproduced in another ipilimumab monotherapy study in MM<br />

(CA184078). Conclusions: Gene expression pr<strong>of</strong>iling <strong>of</strong> peripheral blood<br />

resulted in the identification <strong>of</strong> a set <strong>of</strong> potential biomarkers that may be<br />

predictive <strong>of</strong> severe GI-imAEs before, or early in the course <strong>of</strong> treatment<br />

with ipilimumab. Further validation <strong>of</strong> these biomarkers in a larger patient<br />

cohort is warranted.<br />

8595 General Poster Session (Board #38E), Sun, 8:00 AM-12:00 PM<br />

Patterns <strong>of</strong> care in adjuvant systemic therapy <strong>of</strong> patients with stage III<br />

melanoma: A U.S. population-based study. Presenting Author: Boyu Hu,<br />

Case Western Reserve University School <strong>of</strong> Medicine, Cleveland, OH<br />

Background: Use <strong>of</strong> adjuvant systemic therapy in patients with stage III<br />

melanoma is widely known to be variable based upon multiple factors such<br />

as patient age and comorbidities as well as the preference and even<br />

geographic location <strong>of</strong> the oncologist and patient. The purpose <strong>of</strong> this study<br />

was to compare the use <strong>of</strong> adjuvant therapy among patients treated in<br />

teaching hospitals and community hospitals. Methods: The study population<br />

consisted <strong>of</strong> patients with stage III melanoma enrolled into the<br />

National Cancer Database (NCDB) between 2000-2008. Patients were<br />

selected based upon surgery as the first course <strong>of</strong> therapy which resulted in<br />

a total <strong>of</strong> 27,353 eligible for analysis. The study population was then<br />

categorized into those who were treated at Teaching Hospitals (TH)<br />

including National Cancer Institute-designated cancer centers or Community<br />

Hospitals (CH). Multiple variables including age, median household<br />

income, insurance status, race and overall survival were compared between<br />

patients in the two hospital groups. Results: The overall proportion <strong>of</strong> stage<br />

III patients who received adjuvant systemic therapy was approximately<br />

30%. There was no difference in the proportion <strong>of</strong> patients receiving<br />

adjuvant systemic therapy between patients treated in TH as compared to<br />

CH, and there was no obvious trend towards increased use over time. Of<br />

interest was that the cohort <strong>of</strong> patients designated as being treated at TH<br />

had a higher proportion <strong>of</strong> patients less than 70 years old as compared to<br />

CH. Median household income was found to be higher in patients treated at<br />

TH. Finally, despite the observation that the proportion <strong>of</strong> patients who<br />

received adjuvant therapy was not different, there a significantly higher<br />

5-year overall survival in patients treated at TH as compared to CH.<br />

Conclusions: Although the proportion <strong>of</strong> patients who received adjuvant<br />

systemic therapy was comparable in TH and CH, there was a significant<br />

increase in 5-year overall survival within TH. Additional factors such as age,<br />

lesser comorbidities, more favorable socioeconomic factors or other unmeasured<br />

factors such as type <strong>of</strong> adjuvant therapy or whether adjuvant therapy<br />

was completed may have contributed to the improved survival in patients<br />

treated at TH.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


564s Melanoma/Skin Cancers<br />

8596 General Poster Session (Board #38F), Sun, 8:00 AM-12:00 PM<br />

Detection <strong>of</strong> BRAF mutations in melanoma: Rate <strong>of</strong> mutation detection at<br />

codon 600 using Sanger sequencing as compared to the cobas 4800<br />

method. Presenting Author: Kevin Z. Qu, Nichols Institute, San Juan<br />

Capistrano, CA<br />

Background: Detection <strong>of</strong> BRAF V600 mutations is currently a prerequisite<br />

for approved use <strong>of</strong> vemurafenib in patients with metastatic melanoma. The<br />

cobas 4800 BRAF V600 Mutation Test (Roche Molecular Diagnostics), a<br />

PCR-based assay approved to aid in selecting patients for vemurafenib<br />

therapy, primarily detects V600E. It is also reported to detect V600K,<br />

which has been associated with vemurafenib response as well. We<br />

compared the mutation detection rate <strong>of</strong> the cobas assay with that <strong>of</strong><br />

Sanger sequencing. Methods: 125 de-identified FFPE tissues submitted for<br />

BRAF mutation analysis that all showed histologically-confirmed melanoma<br />

were tested. BRAF mutations were detected using both the cobas kit<br />

and bidirectional Sanger sequencing using BigDye kits (Applied Biosystems).<br />

DNA was extracted from 5-um sections without macrodissection<br />

using the cobas DNA extraction kit (for the cobas test) or from 5-10-um<br />

sections using Agencourt extraction kits (Beckman Coulter) following<br />

macrodissection. Results: The two methods showed agreement in 104/125<br />

(83.2%) <strong>of</strong> cases (Table). Sanger sequencing detected V600 dinucleotide<br />

mutations in 9 samples that were negative by the cobas assay. Sanger<br />

sequencing produced no results in 10 cases owing to suboptimal PCR,<br />

including 2 that were positive by the cobas assay. The cobas assay<br />

produced 2 invalid results, including 1 that was positive for V600E by<br />

Sanger.The cobas assay detected 7/11 V600K mutations. Conclusions:<br />

Overall agreement between cobas and Sanger sequencing was 83.2%. The<br />

Sanger method had higher analytic sensitivity, resulting in nine additional<br />

V600 mutations not called by cobas compared to the two seen by cobas but<br />

not Sanger sequencing. Thus, 16% (9/57) more patients would be<br />

identified as candidates for vemurafenib therapy using the Sanger method.<br />

Sanger<br />

sequencing<br />

Not detected V600E V600K V600R Non-600 No result Total<br />

Not detected 56 3 a<br />

4 b<br />

2 c<br />

2 d<br />

7 74<br />

cobas detected 40 7 2 49<br />

Invalid 1 1 2<br />

Total 56 44 11 2 2 10 125<br />

a 2/3 had dinucleotide mutations (GTG�GAA) and 1 had V600_K601del. b All had<br />

dinucleotide mutations (GTG�AAG). c Both had GTG�AGG dinucleotide mutations. d 1)<br />

G469R; 2) E501G.<br />

8598 General Poster Session (Board #38H), Sun, 8:00 AM-12:00 PM<br />

Pharmacodynamic activity <strong>of</strong> selumetinib to predict radiographic response<br />

in advanced uveal melanoma. Presenting Author: Richard D. Carvajal,<br />

Memorial Sloan-Kettering Cancer Center, New York, NY<br />

Background: Functionally activating mutations (mut) in Gnaq or Gna11,<br />

genes that encode for widely expressed G-protein alpha subunits, are early<br />

oncogenic events in uveal melanoma (UM) development and result in<br />

activation <strong>of</strong> the MAPK pathway. We previously demonstrated effective<br />

pathway inhibition with selumetinib (AZD6244, ARRY-142866) in UM cell<br />

lines, with decreased viability associated with pERK and cyclinD1 suppression<br />

(Ambrosini, AACR 2010). Methods: Using paired metastatic tumor<br />

biopsies from patients (pts) with radiographically progressing UM treated<br />

with selumetinib 75 mg BID on a phase II trial (NCT01143402), we<br />

correlated MAPK pathway inhibition with radiographic tumor regression<br />

and clinical benefit. Biopsies were performed at baseline and after 14 �/-1<br />

days <strong>of</strong> treatment. Western blotting was performed for pERK and cyclinD1,<br />

and quantitated by densitometry. Response (RECIST 1.1) was assessed at<br />

baseline, week (wk) 4, wk 8, and q8wks subsequently. Radiographic<br />

regression was defined as greatest percentage shrinkage from baseline.<br />

<strong>Clinical</strong> benefit was defined as RECIST response or stable disease �16wks.<br />

Results: Paired tumor biopsies were assayed from 18 pts: median age 60<br />

(range 47-81), M:F 11:7, median 1 prior therapy (range 0-2), 17 with liver<br />

involvement, Gnaq mut:Gna11 mut:wild-type 8:9:1. Radiographic regression<br />

was observed in 5 pts, with 2 achieving partial responses. 4 pts were<br />

on study �16wks (16�, 20, 25, 31 wks), with one currently on study at<br />

11� wks. Median pERK and cyclinD1 as measured by densitometry<br />

decreased by 48% (p�.03) and 76% (p�.03), respectively. Radiographic<br />

regression correlated with suppression <strong>of</strong> pERK (Spearmen’s rank correlation;<br />

p�0.04) but not cyclinD1 (p�0.38). A trend towards pERK suppression<br />

correlating with clinical benefit was observed (p�.07) with each <strong>of</strong> the<br />

5 pts achieving PR or SD �16wks having a decrease <strong>of</strong> �30% in pERK<br />

from baseline. Conclusions: Selumetinib can inhibit pERK and cyclinD1 in<br />

UM and can result in tumor shrinkage. Sustained inhibition <strong>of</strong> pERK<br />

inhibition at day 14 may be predictive <strong>of</strong> benefit. Further evaluation <strong>of</strong> MEK<br />

inhibition in this disease is warranted.<br />

8597 General Poster Session (Board #38G), Sun, 8:00 AM-12:00 PM<br />

MicroRNAs induced in melanoma treated with combination targeted<br />

therapy <strong>of</strong> temsirolimus and bevacizumab. Presenting Author: Aubrey G.<br />

Wagenseller, University <strong>of</strong> Virginia, Charlottesville, VA<br />

Background: Targeted therapies directed at commonly overexpressed pathways<br />

in melanoma have clinical activity in numerous trials. Little is known<br />

about how these therapies influence microRNA expression, particularly<br />

with combination regimens. A better understanding <strong>of</strong> how microRNAs are<br />

altered with treatment may contribute to understanding mechanisms <strong>of</strong><br />

therapeutic effects as well as mechanisms <strong>of</strong> tumor escape from therapy.<br />

Methods: Using microRNA arrays, we analyzed microRNA expression levels<br />

in melanoma samples from a Cancer Therapy Evaluation Programsponsored<br />

phase II trial <strong>of</strong> combination temsirolimus and bevacizumab in<br />

stage III or IV melanoma, which elicited clinical benefit in a subset <strong>of</strong><br />

patients. Seventeen patients were treated with temsirolimus for one week,<br />

then combination <strong>of</strong> both temsirolimus and bevacizumab. Metastatic<br />

melanoma biopsies were evaluated days 1, 2, and 23. Tumor samples were<br />

evaluated from 12 patients. Results: microRNA expression remained<br />

unchanged with temsirolimus alone; however, expression <strong>of</strong> 15 microRNAs<br />

was significantly upregulated (1.4 to 2.5-fold) with combination treatment,<br />

compared to pre-treatment levels. Interestingly, twelve <strong>of</strong> these fifteen<br />

microRNAs have been reported to possess tumor suppressor capabilities in<br />

various cancer types, including melanoma. We identified 15 putative<br />

oncogenes and B7-H3, IGF-1, and IGF-1R as potential targets <strong>of</strong> the 12<br />

tumor suppressor microRNAs, based on published experimental evidence.<br />

For 18 <strong>of</strong> 25 pairings <strong>of</strong> microRNA and target-mRNA, changes in gene<br />

expression from pretreatment to post-combination treatment samples were<br />

inversely correlated with changes in microRNA expression, suggesting a<br />

functional effect <strong>of</strong> the microRNA changes induced by combination<br />

therapy. Clustering analysis based on selected microRNAs revealed signatures<br />

characteristic <strong>of</strong> clinical response to combination treatment and <strong>of</strong><br />

tumor BRAF mutational status. Conclusions: We have identified microRNAs<br />

that may be involved in the mechanism <strong>of</strong> action <strong>of</strong> combination temsirolimus<br />

and bevacizumab in metastatic melanoma, possibly through inhibition<br />

<strong>of</strong> oncogenic pathways.<br />

8599 General Poster Session (Board #39A), Sun, 8:00 AM-12:00 PM<br />

Exhibition <strong>of</strong> a stem-cell like phenotype with the expression <strong>of</strong> CD271 in<br />

drug-resistant melanoma cells. Presenting Author: Elizabeth Ann Teresa<br />

Lieser, Mayo Clinic, Rochester, MN<br />

Background: Chemotherapy resistance is a common, difficult problem for<br />

melanoma patients needing long term care and those suffering from<br />

recurrence. Neural growth factor receptor (NGFR), also known as CD271, is<br />

commonly expressed on mesenchymal stem cells and is important for<br />

development, survival and differentiation <strong>of</strong> cells. This has previously<br />

shown to be expressed on melanoma stem cells. We hypothesized that<br />

treatment with carboplatin enriches the population <strong>of</strong> tumor cells for<br />

cancer stem cells (CSC) as a mechanism <strong>of</strong> chemotherapy resistance.<br />

Methods: Core tumor samples from 118 patients with metastatic melanoma<br />

were obtained from formalin-fixed paraffin embedded specimens. Slides<br />

were stained using an anti-CD271 antibody for IHC. A375 melanoma cells<br />

were treated with increasing concentrations <strong>of</strong> carboplatin for approximately<br />

1 year. WT, 6 mg/mL and 10 mg/mL resistant cells were tagged with<br />

CD271-PE antibody and run on a flow cytometer. These cells were then<br />

tested for gene expression by microarray using an Affymetrix human 133<br />

plus 2.0 chip, and analyzed on <strong>Part</strong>ek s<strong>of</strong>tware. Pathway analysis <strong>of</strong> these<br />

carboplatin resistant cells was preformed using Ingenuity. Results: Virtually<br />

all melanoma tumor samples stained positively for CD271 by IHC, with a<br />

median <strong>of</strong> 50% cells within tumor samples expressing the cytoplasmic<br />

marker. Flow cytometry demonstrated an increase in the percentage <strong>of</strong> cells<br />

expressing CD271 as resistance to carboplatin increased. Wild type A375<br />

cells contained 30% positive CD271, A375 6mg/mL resistant contained<br />

42% positive and A375 10mg/mL resistant showed 66% positive. Microarray<br />

also exhibited a direct correlation between CD271 gene expression with<br />

increasing carboplatin resistance. Functional ontology enrichment revealed<br />

development and regulation <strong>of</strong> the EMT pathway as an important<br />

process impacted by our resistant cell line. Conclusions: The data suggests<br />

that as melanoma cells become resistant to certain chemotherapy drugs<br />

they essentially loose their differentiated phenotype and become more<br />

stem cell-like. Monitoring melanoma expression <strong>of</strong> CD271 could help<br />

individualize therapy and anticipate chemotherapeutic resistance in this<br />

high-risk group <strong>of</strong> patients.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


8600 General Poster Session (Board #39B), Sun, 8:00 AM-12:00 PM<br />

Preoperative peripheral blood lymphocyte/monocyte ratio and survival in<br />

patients with resected stage IV melanoma. Presenting Author: Nicole M.<br />

Rochet, Mayo Clinic College <strong>of</strong> Medicine, Rochester, MN<br />

Background: The prognosis <strong>of</strong> stage IV melanoma patients remains poor.<br />

Published results have suggested that components <strong>of</strong> the complete blood<br />

count have significant prognostic value in several malignancies. Among the<br />

most studied were the absolute lymphocyte count (ALC), and absolute<br />

monocyte count (AMC) on clinical outcomes <strong>of</strong> patients with lymphoid<br />

malignancies. Thus, we sought to investigate if the pre-operative ALC<br />

(ALC-PO), AMC (AMC-PO) and ALC/AMC ratio (ALC/AMC-PO) affects the<br />

risk <strong>of</strong> disease recurrence and survival after complete surgical resection <strong>of</strong><br />

metastatic melanoma. Methods: We studied 66 stage IV, resected melanoma<br />

patients followed at Mayo Clinic from 2000 to 2010. Log rank<br />

chi-square analysis was used to determine the best cut-<strong>of</strong>f values for each<br />

pre-operative variable, while proportional hazards models were used to<br />

compared survival. Results: The median follow-up <strong>of</strong> the cohort was 24<br />

months (range: 2.3 – 117 months). ALC-PO, AMC-PO and ALC/AMC-PO, as<br />

continuous variables, were all identified as prognostic factors for both<br />

relapse-free survival (RFS) and overall survival (OS). The best cut-<strong>of</strong>f values<br />

for ALC-PO, AMC-PO and ALC/AMC-PO were 1.9; 0.62; and 2.05,<br />

respectively. Using Kaplan-Meier analysis, patients with an ALC-PO � 1.9<br />

x109 /L experienced superior OS and RFS compared with ALC-PO � 1.9 x<br />

109 /L patients [median OS <strong>of</strong> 58 months vs. 34 months, p � 0.04; median<br />

RFS <strong>of</strong> 14 months vs. 5 months, p � 0.009]. Conversely, a low AMC-PO<br />

(�0.62 x 109 /L) was associated with better OS and RFS compared with<br />

higher AMC-PO (� 0.62 x 109 /L): [median OS <strong>of</strong> 47 months vs. 14 months,<br />

p � 0.007; median RFS <strong>of</strong> 9 months vs. 5 months, p � 0.02]. When the<br />

ALC-PO and AMC-PO were combined as an ALC/AMC ratio, the group with<br />

an ALC/AMC-PO � 2.05 experienced a superior OS and RFS compared to<br />

patients with ALC/AMC-PO � 2.05: [median OS <strong>of</strong> 49 months vs. 12<br />

months, p � 0.0001; median RFS <strong>of</strong> 10 months vs. 4 months, p �<br />

0.0001]. Multivariate analysis showed ALC/AMC-PO to be an independent<br />

prognostic factor for RFS and OS (HR � 0.32, p � 0.003; HR � 0.23, p �<br />

0.002). Conclusions: Our study showed, that ALC/AMC-PO ratio is an<br />

independent prognostic factor for RFS and OS in patients undergoing<br />

resection <strong>of</strong> metastatic (stage IV) melanoma.<br />

TPS8602 General Poster Session (Board #39D), Sun, 8:00 AM-12:00 PM<br />

A phase I/II trial <strong>of</strong> BKM120 combined with vemurafenib (PLX4032) in<br />

BRAFV600E/k mutant advanced melanoma. Presenting Author: Andrea<br />

Kantor, University <strong>of</strong> California, San Francisco, San Francisco, CA<br />

Background: Vemurafenib induces transient objective responses in half <strong>of</strong><br />

BRAFV600E mutant melanoma patients and a median PFS <strong>of</strong> 5.3 months<br />

(NEJM. 2011;364:2507-2516). BRAF mutations are not sufficient to<br />

cause melanoma, but the combination <strong>of</strong> BRAFV600E mutation and PTEN<br />

loss is sufficient to recapitulate the malignant melanoma phenotype in vivo<br />

(Nat. Genet. 2009;41:544-552). PTEN loss and PI3K activation are<br />

common in BRAF mutant metastatic melanoma, and PI3K activation has<br />

been implicated as a cause acquired resistance to BRAF inhibitors (Cancer<br />

Cell. 2010;18:683-695). This phase I/II study is the first trial to test the<br />

safety and efficacy <strong>of</strong> combining a potent BRAF inhibitor, vemurafenib,<br />

with a potent PI3K inhibitor, BKM120, in patients with metastatic BRAF<br />

mutant melanoma. Methods: Design: Phase I patients receive a single dose<br />

<strong>of</strong> oral BKM120 (d -7) then vemurafenib twice daily with BKM120 daily<br />

(starting on c1d1). PK analysis is performed for BKM120 alone and for<br />

both drugs in combination. Doses <strong>of</strong> both drugs will be escalated in 3�3<br />

scheme. Phase II patients receive continuous dosing <strong>of</strong> vemurafenib twice<br />

daily and BKM120 daily. Serial biopsies for PD and mRNA expression<br />

analyses are required for patients with visible or palpable tumors. Reimaging<br />

will be performed every 8 weeks.Eligibility: This study is enrolling<br />

BRAFV600E/K mutant metastatic melanoma patients with no prior exposure<br />

to BRAF inhibitors or PI3K inhibitors, ECOG PS � 2 and adequate organ<br />

function. Endpoints: The primary endpoint for phase 1 is the recommended<br />

phase 2 dose <strong>of</strong> the combination. The primary endpoint for phase II is the 6<br />

month PFS rate. Secondary endpoints include median PFS and OS.<br />

Baseline PTEN expression and changes is pS6 protein levels will be<br />

examined as predictors <strong>of</strong> efficacy, and changes in gene expression pr<strong>of</strong>iles<br />

will be assessed. Summary: This is the first trial examining the safety and<br />

efficacy <strong>of</strong> combined BRAF/PI3K inhibition in BRAF mutant melanoma<br />

patients.<br />

Melanoma/Skin Cancers<br />

565s<br />

8601 General Poster Session (Board #39C), Sun, 8:00 AM-12:00 PM<br />

Comparison <strong>of</strong> sun protection modalities in parents and children. Presenting<br />

Author: Laurent Mortier, Clinique de Dermatologie, CHRU de Lille,<br />

Lille, France<br />

Background: Routine sun protection is recommended to prevent skin<br />

cancer. Our aims were to examine and compare, in an observational survey,<br />

modalities <strong>of</strong> sun protection in parents and their children. Methods: This<br />

French nationwide observational survey, EDIFICE melanoma, was conducted<br />

through phone interviews among a representative sample <strong>of</strong> 1502<br />

subjects aged � 18 years old, using the quota method. The survey took<br />

place shortly after summer, from 28 Sep 2011 to 20 Oct 2011. Results:<br />

1502 subjects were interviewed. Among them 1067 reported that they<br />

were sun-exposed (SE) at least ten days per year, 748 were parents, and<br />

319 had no children. Sun protection measures seemed well done both in<br />

“parents” and “nonparents” groups: 74% used clothing; 43% used<br />

sunscreen, which was regularly renewed in 57% <strong>of</strong> cases. Sun protection<br />

measures used by SE parents for SE children were superior, both qualitatively<br />

and quantitatively to those used for themselves, ie 50% <strong>of</strong> parents<br />

reported using clothing, sunglasses and hat for their children vs 23% for<br />

themselves. In 87% <strong>of</strong> cases, parents reported regular re-application <strong>of</strong><br />

sunscreen for their children vs 44% for themselves. The sunscreen SPF<br />

(Sun Protection Factor) was significantly lower for parents than for their<br />

children. Such data are in accordance with Buller et al. (Oncol Nurs Forum<br />

1995), but contrast with the findings <strong>of</strong> Riordan et al. (J Pediatr 2003) with<br />

children being significantly less likely to wear sunglasses and to adopt sun<br />

avoidance habits than their parents. Conclusions: Sun protection awareness<br />

seems globally satisfying in the French population, with no difference<br />

between adult parents and nonparents, and is similar to that reported in<br />

Germany (Gambicher JEADV 2010), but seems better than in the US<br />

(Buller et al. JAAD 2011). Furthermore, French parents use sun protective<br />

measures qualitatively and quantitatively more efficiently for their children<br />

than for themselves.<br />

TPS8603 General Poster Session (Board #39E), Sun, 8:00 AM-12:00 PM<br />

CA184-161: A phase I/II trial <strong>of</strong> vemurafenib and ipilimumab in patients<br />

with BRAF V600 mutation-positive metastatic melanoma. Presenting<br />

Author: Antoni Ribas, University <strong>of</strong> California, Los Angeles, Los Angeles,<br />

CA<br />

Background: Ipilimumab (ipi) is a fully human monoclonal antibody that<br />

blocks cytotoxic T-lymphocyte antigen-4 (CTLA-4) to augment antitumor<br />

immune responses. Ipi significantly improved overall survival (OS) in two<br />

separate phase III trials <strong>of</strong> metastatic melanoma (MM): (1) at 3 mg/kg as<br />

monotherapy in previously treated patients and (2) at 10 mg/kg combined<br />

with dacarbazine in previously untreated patients. The most common<br />

drug-related adverse events with ipi were immune-related, and were<br />

generally reversible using treatment guidelines. Vemurafenib (vem) is a<br />

potent inhibitor <strong>of</strong> BRAF V600E-mutated kinase, which occurs in ~50% <strong>of</strong><br />

MM. In clinical studies, vem produced objective response rates <strong>of</strong> ~50%<br />

with a significant improvement in the rate <strong>of</strong> OS in patients with previously<br />

untreated BRAF V600-positive MM. The purpose <strong>of</strong> this phase I/II study is<br />

to determine the safety and feasibility <strong>of</strong> ipi plus vem in patients with BRAF<br />

V600-positive MM, and to evaluate the efficacy <strong>of</strong> the combination in<br />

comparison to historical results observed with each agent alone. Methods:<br />

In phase I, an escalating dose design is used to assess safety/tolerability<br />

and to determine the maximum tolerated dose (MTD) <strong>of</strong> each agent. Vem<br />

will initially be given alone for 28 days at 960 mg p.o. BID, followed by vem<br />

in combination with ipi i.v. at 3 mg/kg [induction (q3w x 4) and<br />

maintenance (q12w)]. The next cohort will receive vem at 960 mg plus ipi<br />

increased to a dose <strong>of</strong> 10 mg/kg. Phase I will enroll an estimated 20<br />

patients who have not previously received a CTLA-4 antagonist or BRAF<br />

inhibitor. If new or heightened frequency or severity <strong>of</strong> toxicities is not<br />

observed, a phase II single-arm extension at the MTD <strong>of</strong> each agent will be<br />

conducted in patients with previously untreated MM. The primary objectives<br />

<strong>of</strong> phase II (estimated enrollment <strong>of</strong> 30 patients) are to obtain<br />

preliminary evidence <strong>of</strong> efficacy (OS, 1- and 2-year survival rates) and to<br />

evaluate safety <strong>of</strong> the combination. Major inclusion criteria are � 18 years<br />

<strong>of</strong> age, BRAF V600-positive MM, measurable tumor, no active brain<br />

metastasis, and an ECOG PS <strong>of</strong> 0-1. The first patient was enrolled in<br />

November 2011 and enrollment is ongoing (<strong>Clinical</strong>Trials.gov identifier:<br />

NCT01400451).<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


566s Melanoma/Skin Cancers<br />

TPS8604 General Poster Session (Board #39F), Sun, 8:00 AM-12:00 PM<br />

OPTiM: A randomized phase III trial to evaluate the efficacy and safety <strong>of</strong><br />

talimogene laherparepvec (T-VEC) compared with subcutaneously (sc)<br />

administered GM-CSF for the treatment (tx) <strong>of</strong> unresectable stage IIIb, IIIc,<br />

and IV melanoma. Presenting Author: Howard Kaufman, Rush University<br />

Medical Center, Chicago, IL<br />

Background: T-VEC (formerly OncoVEXGM-CSF ) is an oncolytic HSV1 that<br />

selectively replicates in tumors. The proposed MOA includes lytic destruction<br />

<strong>of</strong> injected tumors and induction <strong>of</strong> a systemic anti-tumor immune<br />

response enhanced by local GM-CSF expression. Intratumoral T-VEC tx in a<br />

50-patient (pt) ph II study in advanced melanoma (Stage IIIc-IVM1c) was<br />

well tolerated and achieved a high rate and duration <strong>of</strong> response, including<br />

20% CR (Senzer et al., JCO 2009; 27: 5763-71). As immune effects may<br />

be delayed, progressive disease (PD) <strong>of</strong>ten occurred before response.<br />

Based on the ph II data, a randomized ph III trial <strong>of</strong> T-VEC in unresectable<br />

melanoma (the OPTiM study; clinical trials registry NCT00769704) was<br />

designed taking into account the response patterns seen with T-VEC and<br />

other immunotherapeutic agents. T-VEC is the first �armed� oncolytic agent<br />

to enter pivotal testing worldwide. Methods: OPTiM compares the efficacy<br />

and safety <strong>of</strong> intratumoral T-VEC to sc GM-CSF in 430 pts with treated or<br />

untreated unresectable Stage IIIb-IVM1c melanoma stratified by typical<br />

prognostic factors. Pts are randomized 2:1 to T-VEC (priming dose <strong>of</strong> up to<br />

4x106pfu intratumorally then 3 wks later by up to 4x108pfu Q2W) or<br />

GM-CSF 125 �g/m2 qd sc x 14 days every 28 days. Key eligibility criteria<br />

are � 18 yrs old, ECOG 0-1, and at least 1 injectable cutaneous, sc, or<br />

nodal tumor. The primary endpoint is durable response rate (DRR: CR or PR<br />

continuously maintained for � 6 mo initiating within 12 mo <strong>of</strong> starting tx;<br />

secondary endpoints include OS. Responses are subject to independent<br />

review. Pts are treated until wk 24, even with PD. Thereafter pts are treated<br />

until clinically significant PD, CR, or for 1 yr. The study has 90% power to<br />

show a 10% difference in DRR with 2-sided Fisher Exact Test (� � 5%).<br />

Enrollment closed in July 2011 with 439 pts randomized in the US, UK, S<br />

Africa, and Canada. Interim analysis in Dec 2010 on 75 pts on study � 9<br />

mo concluded that the study should continue. Results are expected during<br />

2012. Conclusions: T-VEC provides a novel potential tx for melanoma. This<br />

pivotal ph III study is expected to report during 2012.<br />

TPS8606 General Poster Session (Board #39H), Sun, 8:00 AM-12:00 PM<br />

Pilot evaluation <strong>of</strong> sulforaphane in melanoma patients with multiple<br />

atypical nevi: Tissue STAT1 and STAT3 as risk markers. Presenting Author:<br />

David Lobur, University <strong>of</strong> Pittsburgh Medical Center, Pittsburgh, PA<br />

Background: Increasing incidence and the poor prognosis <strong>of</strong> advanced<br />

melanoma argue for prevention efforts. Primary prevention and ultraviolet<br />

radiation (UVR) reduction have failed to gain traction in the population,<br />

making chemoprevention increasingly attractive. Sulforaphanes (SFN) are<br />

bioactive cruciferous compounds rich in the Brassica family, especially<br />

broccoli sprouts. Topical broccoli sprout extracts (BSE) decrease UVR<br />

erythema response in human skin, and may protect against UVR DNA<br />

damage. We have shown SFN inhibition <strong>of</strong> STAT3, which is constitutively<br />

activated in melanoma. We have therefore initiated a pilot study to evaluate<br />

BSE SFN in relation to melanoma progression biomarkers, and expression<br />

<strong>of</strong> STAT proteins <strong>of</strong> atypical melanocytic nevi. Methods: Melanoma patients<br />

with �2 atypical nevi are eligible for this trial, which aims to define the<br />

safety and clinico-pathological response to oral BSE-SFN. Secondary<br />

objectives are documentation <strong>of</strong> pharmacokinetics and pharmacodynamics<br />

<strong>of</strong> BSE-SFN, and the modulation <strong>of</strong> STAT 1/3 expression in atypical nevi<br />

and normal skin. Baseline photo-documentation and atypical nevus biopsy<br />

is performed to assess histopathological atypia and STAT 1/3 expression.<br />

Adjacent normal skin biopsies will establish baseline skin SFN levels. Six<br />

subjects per group will be accrued at daily dosages <strong>of</strong> 50, 100, and 200<br />

�M. Subjects must abstain from dietary glucosinolates and isothiocyanates<br />

for the study duration and maintain food diaries. Following 27 days <strong>of</strong><br />

BSE-SFN, blood samples, photography <strong>of</strong> index atypical nevi, and biopsies<br />

<strong>of</strong> selected atypical nevi and normal skin will be obtained. The three dosage<br />

groups will be analyzed for changes in atypia, SFN localization histopathology,<br />

and STAT 1/3 expression in the nevi and skin harvested at baseline and<br />

at study completion. This trial, UPCI 10-114, has received an IND and is<br />

IRB-approved. Accrual is planned to be completed during 2012-13.<br />

TPS8605 General Poster Session (Board #39G), Sun, 8:00 AM-12:00 PM<br />

A randomized phase II study <strong>of</strong> sunitinib versus dacarbazine in the<br />

treatment <strong>of</strong> patients with metastatic uveal melanoma. Presenting Author:<br />

Ernie Marshall, Clatterbridge Centre for Oncology, Wirral, United Kingdom<br />

Background: Metastatic uveal melanoma represents an orphan disease area<br />

with a median survival <strong>of</strong> less than 6 months. There is currently no effective<br />

systemic therapy for metastatic uveal melanoma and few clinical trials have<br />

been conducted. In the absence <strong>of</strong> phase III data, many patients in the UK<br />

continue to receive single agent dacarbazine or best supportive care outside<br />

<strong>of</strong> the context <strong>of</strong> clinical trials. Uveal melanoma is characterized by<br />

activation <strong>of</strong> the MAP kinase pathway via functionally activating mutations<br />

in Gnaq/11. Evidence also suggests that dysfunctional c-Kit signalling and<br />

angiogenesis may both play a role in disease progression and a small<br />

single-arm phase II trial recently reported preliminary activity using the<br />

multi-targeted receptor tyrosine kinase inhibitor, sunitinib (Tijani et al,<br />

ASCO 2010). Methods: The SUAVE trial aims to evaluate the Progression<br />

Free Survival (PFS) <strong>of</strong> good performance status patients treated with<br />

sunitinib or dacarbazine. Secondary objectives include: Overall Survival<br />

(OS), Overall Response Rate (ORR), safety, crossover PFS and response<br />

and biomarker analyses. SUAVE is a CR-UK-funded, open-label, randomised,<br />

phase II trial that will include 124 patients. Patients will be<br />

stratified according to their Helsinki Prognostic Index (ALP, largest<br />

diameter <strong>of</strong> largest metastasis, ECOG). Inclusion Criteria: good performance<br />

status patients with confirmed unresectable metastatic uveal<br />

melanoma, with at least one target lesion measurable by RECIST 1.1.<br />

Patients must not have received previous systemic therapy. At confirmed<br />

progression, good performance status patients may crossover to the other<br />

study treatment. The trial opened in Oct 2010 and as <strong>of</strong> 23 January 2012,<br />

had randomized 49 patients with at least 1 recruited from each <strong>of</strong> the 12<br />

recruiting sites. There will be a total <strong>of</strong> 13 sites. Completion <strong>of</strong> the<br />

recruitment phase is expected within 36 months. Prospective tissue and<br />

blood sample collection for translational biomarker analyses is also<br />

ongoing. The SUAVE trial represents one <strong>of</strong> the largest randomised trials in<br />

this rare disease area (<strong>Clinical</strong> trial registry number: 2008-008794-55).<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


9000 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Randomized trial <strong>of</strong> vitamin D3 to prevent worsening <strong>of</strong> musculoskeletal<br />

symptoms and fatigue in women with breast cancer starting adjuvant<br />

letrozole: The VITAL trial. Presenting Author: Qamar J. Khan, University <strong>of</strong><br />

Kansas Medical Center, Kansas City, KS<br />

Background: Musculoskeletal (MS) pain and fatigue are common in women<br />

on adjuvant Aromatase Inhibitors (AIs) and lead to reduced compliance.<br />

Pilot studies suggest a positive impact <strong>of</strong> vit D on MS pain and disability<br />

from AIs. We conducted a bi-institutional, double blind, placebo controlled<br />

randomized trial to study the impact <strong>of</strong> 30,000 I.U <strong>of</strong> vit D3 in preventing<br />

worsening <strong>of</strong> MS pain and fatigue in women starting letrozole. Methods:<br />

Women with stage I-III breast cancer starting adjuvant AI and a 25(OH)D<br />

level <strong>of</strong> 40 ng/ml or less were eligible. Women with prior renal stones or<br />

hypercalcemia were excluded. All subjects received letrozole, plus standard<br />

dose vitD3 (600 IU) and calcium (1200 mg) daily, all provided by the<br />

study. Women were randomly assigned to 30,000 IU <strong>of</strong> oral vitD3 wkly (vitD<br />

arm) or matched placebos (PL arm) for 24 weeks. The following were<br />

assessed at baseline, 12 wks, and 24 wks (end <strong>of</strong> study): 1) 25OHD levels,<br />

2) symptoms tools (BFI – Brief Fatigue Inventory, HAQII - Health Assessment<br />

Questionnaire II, Qualitative Joint Pain (none, mild, moderate,<br />

severe), BPI – Brief Pain Inventory) and 3) hand grip strength with a<br />

dynamometer. Results: 160 women (80/arm) were enrolled from 4/09 to<br />

7/10. There were no differences between the two arms in demographics/<br />

tumor characteristics. Median age was 61, median BMI was 29.8 kg/m2.<br />

43% had adjuvant chemotherapy. Median 25OHD (ng/ml) was 25 at<br />

baseline, 32 at 12 wks and 31 at 24 wks in the PL arm and 22, 53 and 57<br />

in vitD arm. One patient in the PL arm developed mild hypercalcemia.<br />

There were no SAEs. 147 subjects were evaluable for efficacy. 3 subjects,<br />

all in the PL arm discontinued early due to a MS adverse event. At wk 24, a<br />

higher proportion <strong>of</strong> women in PL (51%) vs vitD arm (37%) had a protocol<br />

defined MS event (worsening <strong>of</strong> joint pain, disability from joint pain or<br />

discontinuation <strong>of</strong> Letrozole due to MS symptoms) (p�0.069). A significantly<br />

higher proportion <strong>of</strong> women in PL (72%) vs vitD arm (42%) had an<br />

adverse QOL event (worsening <strong>of</strong> pain, disability or fatigue) (p��0.001).<br />

Conclusions: 30,000 IU/week <strong>of</strong> vitamin D3 is safe and results in decreased<br />

adverse QOL events from adjuvant aromatase inhibitors in women with<br />

breast cancer.<br />

9002 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Dexamethasone (DM) for cancer-related fatigue: A double-blinded, randomized,<br />

placebo-controlled trial. Presenting Author: Sriram Yennurajalingam,<br />

University <strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: Cancer-related-fatigue (CRF) is the most common and distressing<br />

symptoms in patients with advanced cancer. Currently, there is no<br />

standard treatment for CRF. Although corticosteroids have been used in the<br />

treatment <strong>of</strong> CRF, there are no well-powered placebo-controlled trials that<br />

used a validated CRF outcome measure. The primary objective <strong>of</strong> this<br />

prospective, randomized, double-blind, placebo-controlled study is to<br />

compare the effect <strong>of</strong> DM versus placebo on CRF. Methods: Advanced<br />

cancer patients with fatigue � 4/10 on the Edmonton Symptom Assessment<br />

Scale (ESAS) and at least 2 other CRF-related symptoms (pain,<br />

nausea, appetite, depression, anxiety or sleep disturbance � 4/10), normal<br />

cognition, no infections and hemoglobin � 9 g/L were eligible for<br />

enrollment. Patients were randomized to either receive dexamethasone 4<br />

mg orally twice a day for 15 days (primary end point) or matching placebo.<br />

The primary outcome was the day 15 change in Functional Assessment <strong>of</strong><br />

Chronic Illness-Fatigue (FACIT-F) subscale scores. Differences in the group<br />

means (normal distribution) were analyzed using the two-sample t-test.<br />

Results: In 83 evaluable patients (43 DM and 40 placebo), median age was<br />

60 years, 61% were white, and 53% were female. There was no difference<br />

in the demographics and fatigue (FACIT-F subscale) between DM and<br />

placebo groups except for sex (p�0.02). The mean (SD) FACIT-F subscores<br />

at baseline and at day 15 for DM were 18 (11) and 27 (11) (p�0.001) and<br />

for placebo were 21 (9) and 24 (12) (p�0.06), respectively. Mean<br />

improvement in FACIT-F subscale was significantly higher in the DM group<br />

compared to placebo (9.6 (11) vs. 3.1 (9.7), p�0.005). We found a<br />

significant difference in ESAS physical distress (p�0.02), but no differences<br />

in ESAS overall symptom distress (p�0.11) and ESAS psychological<br />

distress (P�0.88) between DM and placebo. There were insignificantly<br />

higher numbers <strong>of</strong> grade �3 toxicities in patients who received DM than in<br />

patients who received placebo (20/42 vs. 18/47, p�0.37). Conclusions:<br />

Dexamethasone was more effective than placebo in reducing CRF in<br />

patients with advanced cancer. Long-term safety studies are needed.<br />

Patient and Survivor Care<br />

9001 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Phase III evaluation <strong>of</strong> <strong>American</strong> ginseng (panax quinquefolius) to improve<br />

cancer-related fatigue: NCCTG trial N07C2. Presenting Author: Debra L.<br />

Barton, Mayo Clinic, Rochester, MN<br />

Background: Ginseng is popularly used as a treatment for fatigue, one <strong>of</strong> the<br />

most common and disabling symptoms in people diagnosed with cancer. It<br />

is termed an “adaptogen”, thought to help the body combat negative<br />

effects <strong>of</strong> stress. This trial was to evaluate 2,000 mg <strong>American</strong> Ginseng<br />

versus placebo for cancer-related fatigue (CRF). Methods: Patients with<br />

cancer undergoing or having completed curative intent treatment and<br />

experiencing fatigue, rated at least 4 on a numeric analogue fatigue scale<br />

(1-10) for �1 month, were eligible. Exclusion criteria included CNS<br />

lymphoma, brain malignancies, or prior use <strong>of</strong> ginseng or chronic systemic<br />

steroids. Other etiologies for fatigue, such as pain and sleep, were also<br />

excluded. Patients were randomized to receive, in a double blind manner,<br />

2,000 mg/d <strong>of</strong> <strong>American</strong> Ginseng or placebo in BID dosing for 8 weeks. The<br />

primary endpoint was change from baseline in the general subscale <strong>of</strong> the<br />

Multidimensional Fatigue Symptom Inventory (MFSI) at 4 weeks. Other<br />

MFSI subscales and the fatigue-inertia subscale <strong>of</strong> the Pr<strong>of</strong>ile <strong>of</strong> Mood<br />

States (POMS) were also analyzed. Data were transformed to a 0-100 scale.<br />

Results: 364 patients were enrolled from 10/2008 to 07/2011. Data at 4<br />

and 8 weeks are provided for several fatigue endpoints in the table below;<br />

higher numbers are better. Mental, emotional and vigor subscales <strong>of</strong> the<br />

MFSI were not significantly different between arms. There were no<br />

statistically significant differences in any grade <strong>of</strong> toxicity or self reported<br />

side effects between ginseng and placebo. Conclusions: This trial provides<br />

data to support that <strong>American</strong> Ginseng reduces general and physical CRF<br />

over 8 weeks without side effects. The treatment did not provide significant<br />

reductions in fatigue at 4 weeks and did not impact mental, emotional, and<br />

vigor dimensions <strong>of</strong> fatigue.<br />

Fatigue measure Weeks<br />

Change scores (SD)<br />

Ginseng<br />

n�147<br />

567s<br />

Placebo<br />

n�152 p value<br />

MFSI - general 4 14.4 (27.1) 8.2 (24.8) 0.0737<br />

8 20.0 (27.0) 10.3 (26.1) 0.0029<br />

MFSI - physical 4 1.6 (15.9) -0.4 (14.7) 0.3942<br />

8 3.0 (17.9) -1.7 (18.2) 0.0043<br />

MFSI – total 4 4.1 (13.4) 2.1 (12.9) 0.2061<br />

8 6.7 (14.0) 3.7 (14.6) 0.0193<br />

POMS – fatigue/inertia 4 14.5 (25.0) 7.7 (23.6) 0.0795<br />

8 18.6 (24.8) 10.2 (26.1) 0.0083<br />

9003 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Cluster-randomized trial <strong>of</strong> early palliative care for patients with metastatic<br />

cancer. Presenting Author: Camilla Zimmermann, Princess Margaret Hospital,<br />

University Health Network, Toronto, ON, Canada<br />

Background: Patients with metastatic cancer have compromised quality <strong>of</strong><br />

life (QOL), which tends to worsen towards the end <strong>of</strong> life. We conducted a<br />

cluster-randomized trial <strong>of</strong> early versus routine palliative care in patients<br />

with metastatic cancer, to assess impact on QOL, symptom control and<br />

satisfaction with care. Methods: Twenty-four medical oncology clinics were<br />

randomized, stratified by tumour site (4 lung clinics, 8 gastrointestinal, 4<br />

genito-urinary, 6 breast, 2 gynecological), to intervention and follow-up (at<br />

least monthly) by a palliative care team, or to routine cancer care. Eligible<br />

patients had ECOG performance status 0-2 and a clinical prognosis <strong>of</strong> 6<br />

months to 2 years. Patients completed measures <strong>of</strong> QOL (FACIT-Sp,<br />

including physical, social, emotional, functional and spiritual well-being;<br />

range 0-156, with higher scores indicating better QOL), symptom severity<br />

(Edmonton Symptom Assessment System; range 0-90, with higher scores<br />

indicating worse symptom severity), and satisfaction with care (FAMCARE-<br />

P16; score range 16-80) at baseline and monthly for 4 months. The<br />

primary outcome was the change in FACIT-Sp at 3 months. The planned<br />

sample size was 225 patients per arm, assuming 80% power and a 2-sided<br />

significance level <strong>of</strong> 0.05. Results: From December 2006 to September<br />

2010, 461 patients completed baseline measures (228 intervention, 233<br />

control); 442 patients completed at least one follow-up assessment (mean<br />

patients/cluster 18.8�11.6 control, 18.1�12.6 intervention). At 3 months,<br />

patients in the intervention group had marginally improved QOL (mean<br />

change in intervention vs. control, 1.6�14.5 vs. -2.0�13.6, p�0.07) but<br />

not symptom severity (0.1�16.9 vs. 2.1�13.9, p�0.34). At 4 months the<br />

change in QOL was more marked (2.5�15.5 vs. -4.0�14.2, p�0.008)<br />

and symptom severity was marginally better (-1.3�16.0 vs. 3.2�13.9,<br />

p�0.05). Improvement in satisfaction with care was evident at one month<br />

(p�0.001) and remained significant at both 3 months (2.3�9.1 vs.-<br />

1.8�8.2, p�0.001) and 4 months (3.7�8.6 vs. -2.4�8.3, p�0.001).<br />

Conclusions: In patients with metastatic cancer, early palliative care<br />

intervention immediately improved satisfaction with care, while QOL and<br />

symptom control improved later.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


568s Patient and Survivor Care<br />

9004 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

A video decision support tool for CPR in advanced cancer: A randomized<br />

controlled trial. Presenting Author: Angelo E. Volandes, Massachusetts<br />

General Hospital, Boston, MA<br />

Background: End-<strong>of</strong>-life decision-making is important to patients, including<br />

whether or not to attempt cardiopulmonary resuscitation (CPR). Doctors<br />

<strong>of</strong>ten rely solely on verbal descriptions to communicate information<br />

regarding CPR. Video decision support tools have the potential to improve<br />

patients’ understanding <strong>of</strong> CPR by providing visual images <strong>of</strong> what this<br />

intervention entails. The objective <strong>of</strong> this study was to examine the effect <strong>of</strong><br />

a CPR video among patients with advanced cancer on their preferences and<br />

knowledge <strong>of</strong> CPR. Methods: A randomized controlled trial <strong>of</strong> 150 subjects<br />

with diverse advanced cancers (� 1-year prognosis) recruited from 4<br />

cancer centers in the United States. Subjects were randomized to either a<br />

verbal narrative describing CPR, or to a video with verbal narrative. The<br />

video depicts CPR and reviews the success rate in advanced cancer. Study<br />

endpoints were subjects’ CPR preferences, knowledge <strong>of</strong> CPR (knowledge<br />

scores ranged from 0-4, higher score indicating more knowledge), and<br />

perceived value <strong>of</strong> the video. Chi-square tests were used to compare the<br />

distributions <strong>of</strong> categorical outcomes and two-sample t-tests were used to<br />

compare the means between the two groups. Results: A total <strong>of</strong> 150<br />

subjects were randomized to a verbal narrative (n�80) or video with verbal<br />

narrative (n�70). Mean age was 62, 49% were women, 47% White, 34%<br />

Black, and 47% had lung or colon cancer. Among subjects receiving the<br />

verbal narrative, 38 (47.5%) preferred to have CPR attempted; 41 (51.2%)<br />

chose not to have CPR; and 1 (1.3%) was uncertain. In the video group, 14<br />

(20%) preferred to have CPR attempted; 55 (78.6%) chose not to have<br />

CPR; and 1 (1.4%) was uncertain (P�0.001). The mean knowledge score<br />

was higher in the video group than in the verbal group (3.3 vs. 2.6<br />

respectively; P�0.001). Of the subjects who viewed the video, 94.1%<br />

stated they were comfortable watching the video, 97.1% found the video<br />

helpful, and 100% would recommend the video. Conclusions: Compared to<br />

subjects who only heard a verbal description, subjects with advanced<br />

cancer who viewed a CPR video were more likely to prefer not having CPR,<br />

and were more knowledgeable about CPR. The majority <strong>of</strong> subjects found<br />

the video helpful, comfortable to view, and would recommend it to others.<br />

9006 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Long-term protective effects <strong>of</strong> the angiotensin receptor blocker telmisartan<br />

on epirubicin-induced inflammation, oxidative stress, and myocardial<br />

dysfunction. Presenting Author: Giovanni Mantovani, Department <strong>of</strong> Internal<br />

Medical Sciences, Service <strong>of</strong> Medical Oncology, University <strong>of</strong> Cagliari,<br />

Cagliari, Italy<br />

Background: Chronic inflammation, oxidative stress and renin-angiotensin<br />

system (RAS) play a significant role in chemotherapy-induced cardiotoxicity<br />

(CTX). Telmisartan (TEL), an antagonist <strong>of</strong> the angiotensin II type-1<br />

receptor, reduces anthracycline (ANT)-induced CTX. Methods: A phase II<br />

placebo (PLA)-controlled randomized trial was carried out to assess the role<br />

<strong>of</strong> TEL in the prevention <strong>of</strong> cardiac subclinical damage induced by<br />

epirubicin (EPI). Forty-nine patients (mean age � SD, 53.0�8 years),<br />

cardiovascular disease-free with cancer at different sites and eligible for<br />

EPI-based treatment, were randomized to one <strong>of</strong> two arms: TEL n�25; PLA<br />

n�24. A conventional echocardiography equipped with Tissue Doppler<br />

imaging, strain and strain rate (SR) was performed, and serum levels <strong>of</strong><br />

proinflammatory cytokines, IL-6 and TNF-�, and oxidative stress parameters,<br />

reactive oxygen species (ROS) and glutathione peroxidase were<br />

assessed. All assessments were carried out at baseline, after every 100<br />

mg/m2 EPI dose and at 6, 12 and 18-month follow-up (FU). Results: A<br />

significant reduction in the SR peak in both arms was observed at t2 (cumulative dose <strong>of</strong> 200 mg/m2 EPI) in comparison to t0. Conversely, at t3, t4 (300, 400 mg/m2 EPI), 6, 12 and 18-month FU, the SR increased<br />

reaching the normal range only in TEL arm, while in PLA arm SR remained<br />

significantly lower as compared to t0. The differences between SR changes<br />

in PLA and TEL arms were significant from 300 mg/m2 EPI (t3)upto18 month-FU. IL-6 increased significantly in PLA arm at t2 compared to t0, but<br />

remained unchanged in the TEL arm. ROS levels also increased significantly<br />

at t2 vs. baseline in PLA arm, while remained unchanged in TEL arm.<br />

The mean change in ROS and IL-6 at t2 was significantly different between<br />

the two arms. At 3, 6, 12 and 18 month-FU, ROS and IL-6 decreased in<br />

comparison to t2 in PLA arm, while remained low in TEL arm. Conclusions:<br />

Our results suggest that TEL is able to reverse acute EPI-induced<br />

myocardial dysfunction and maintain a normal systolic function up to 18<br />

month-FU. These effects are likely due to two different mechanisms, RAS<br />

blockade and prevention <strong>of</strong> chronic inflammation/oxidative stress.<br />

9005 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

ZOOM: A prospective, randomized trial <strong>of</strong> zoledronic acid (ZOL;q4wkvsq<br />

12 wk) for long-term treatment in patients with bone-metastatic breast<br />

cancer (BC) after 1 yr <strong>of</strong> standard ZOL treatment. Presenting Author: Dino<br />

Amadori, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori,<br />

Meldola, Italy<br />

Background: ZOL (4 mg q 4 wk) is an established therapy for reducing the<br />

risk <strong>of</strong> debilitating skeletal-related events (SREs) in patients (pts) with<br />

bone metastases (mets) from BC. As BC treatments continue to improve pt<br />

survival, long-term SRE-reduction is increasingly important, and evaluation<br />

<strong>of</strong> modified ZOL dosing to retain efficacy with reduced adverse events (AEs)<br />

is warranted. Methods: ZOOM, a phase III prospective, randomized,<br />

open-label, multicenter study, assessed the safety and efficacy <strong>of</strong> quarterly<br />

(4 mg q 12 wk; Arm 1) vs monthly (4 mg q 4 wk; Arm 2) ZOL for ~1 yr in pts<br />

with BC who have � 1 bone met, and have received ~1 yr <strong>of</strong> prior ZOL<br />

treatment (9 to 12 doses over � 15 months; last dose � 3 months prior).<br />

The primary endpoint was skeletal morbidity rate (SMR; number <strong>of</strong><br />

SREs/pt/yr). Sample size to detect non-inferiority with 80% power (1-sided<br />

a � 0.025) was 420 pts. Secondary endpoints included time to first SRE,<br />

bone pain, bone marker (N-telopeptide <strong>of</strong> type I collagen; NTX) levels, and<br />

safety. Results: 425 pts were enrolled (209 in Arm 1; 216 in Arm 2); arms<br />

were well balanced for pt and disease characteristics, and anticancer<br />

therapies. SMR was similar between arms: 0.26 (95% confidence interval<br />

[CI] � 0.15, 0.37) in Arm 1 vs 0.22 (95% CI � 0.14, 0.29) in Arm 2;<br />

between-arms difference � 0.04 (upper limit <strong>of</strong> 1-tailed 97.5% CI �<br />

0.17). Despite the proximity <strong>of</strong> 0.17 to the adjusted non-inferiority margin<br />

(0.19), the non-inferiority <strong>of</strong> Arm 1 vs 2 remains statistically significant.<br />

Safety analyses showed that ZOL was well tolerated. Renal AEs were<br />

reported in similar proportions <strong>of</strong> pts in both arms; 7 cases <strong>of</strong> osteonecrosis<br />

<strong>of</strong> the jaw were reported (1.65% overall; 4 cases in Arm 1 vs 3 in Arm 2).<br />

Conclusions: ZOOM is the first randomized trial to compare ZOL q 12 wk vs<br />

ZOLq4wkinBCptsafter 1 yr <strong>of</strong> standard ZOL therapy. SMR was similar<br />

between arms. Limitations in study design suggest the need to confirm<br />

non-inferiority <strong>of</strong> ZOL q 12 wk in other ongoing phase III trials.<br />

9007 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Post diagnosis weight gain and the role <strong>of</strong> exercise in breast cancer<br />

survivors. Presenting Author: Tara Beth Sanft, Yale University, New Haven,<br />

CT<br />

Background: Obesity is linked to poor breast cancer (Br Ca) outcomes<br />

including higher risk <strong>of</strong> recurrence and death. No study has examined if<br />

post-diagnosis weight (wt) gain modifies the effect <strong>of</strong> exercise interventions<br />

on body composition. We examined wt change from diagnosis (diag) to<br />

enrollment (enroll) into a 6-mo randomized trial <strong>of</strong> exercise v. usual care to<br />

explore if post-diagnosis wt gain modified the effect <strong>of</strong> exercise on body wt,<br />

body fat, lean body mass (LBM) and bone mineral density (BMD) in breast<br />

cancer survivors. Methods: 75 inactive Br Ca survivors were recruited and<br />

randomized to exercise (n � 37) or usual care (UC) (n � 38). At baseline,<br />

women completed a questionnaire assessing self-report wt at diag and at<br />

the time <strong>of</strong> enroll into the study (~3 yr). The exercise group participated in<br />

150 min/wk <strong>of</strong> supervised exercise. The UC group maintained their<br />

physical activity level. Body composition was assessed at enroll and 6-mo<br />

via dual-energy X-ray absorptiometry (DXA) by one radiologist blinded to<br />

intervention group. Results: Wt and DXA data were available for 68 / 75<br />

participants. On average, women gained 3.3�8.2 kg from diag to enroll,<br />

with 53% gaining wt (mean � 8.9 � 6.8 kg) and 47% maintaining or losing<br />

wt (mean change � -2.9 �3.9 kg). The majority <strong>of</strong> wt gain occurred among<br />

women who were non-obese at diag (BMI � 30), with 60% <strong>of</strong> non-obese<br />

women gaining wt (mean � 8.0 � 6.8 kg) compared with 38% <strong>of</strong> obese<br />

women gaining wt (mean � 12.2 � 6.4 kg). Those randomized to exercise<br />

lost body fat including women who gained wt since diag (mean % fat loss<br />

for exercisers (-0.92 � 0.31) vs. UC (0.36 �0.30), p � .0063) and women<br />

who did not gain wt since diag (mean % fat loss for exercisers (-0.65 �<br />

0.39) vs. UC (0.45�0.47), p � .08) even after adjusting for obesity status<br />

at diag. Similar improvements in LBM and BMD were observed with<br />

exercise in women who did or did not gain wt since diag. Conclusions: Wt<br />

gain in Br Ca survivors is common, the majority <strong>of</strong> weight gain in this study<br />

occurred among women who were not obese at diag. Exercise led to body fat<br />

loss and other favorable body composition changes among both non-obese<br />

and obese women. Exercise and other wt management programs should be<br />

<strong>of</strong>fered to all Br Ca survivors regardless <strong>of</strong> BMI at diagnosis as a strategy<br />

towards prevention <strong>of</strong> wt gain.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


9008 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Electronic self-report assessment for cancer: Results <strong>of</strong> a multisite randomized<br />

trial. Presenting Author: Donna Lynn Berry, Dana-Farber Cancer<br />

Institute, Boston, MA<br />

Background: Attending to symptoms and side effects promotes safe and<br />

effective delivery <strong>of</strong> cancer therapies. Providing focus to the patient’s<br />

report in the clinic can efficiently address the most important concerns,<br />

without extending visit times. The web-based electronic self report assessment<br />

for cancer (ESRA-C) and clinician summary has been shown to<br />

improve patient-clinician communication about symptoms and quality <strong>of</strong><br />

life issues (SQI). The purpose <strong>of</strong> this trial was to evaluate the impact <strong>of</strong> an<br />

enhanced ESRA-C intervention (ESRA-C INT) on symptom outcomes.<br />

Methods: Patients (planned N�702) with all cancer types treated in stem<br />

cell transplant, medical oncology and radiation oncology at two comprehensive<br />

cancer centers used ESRA-C to self-report SQI during and after new<br />

anti-cancer therapy, with summary reports delivered to clinicians. Patients<br />

were randomized to standard ESRA-C (control) or ESRA-C INT adding the<br />

opportunity to self-monitor SQI between clinic visits and receive self-care<br />

SQI management education, plus custom coaching on how to report SQI to<br />

clinicians, all delivered via a secure web sites. We analyzed the intervention<br />

effect on Symptom Distress Scale (SDS) scores using a two-sided t-test and<br />

multivariate linear regression, adjusting for pre-selected covariates, including<br />

baseline SDS, age, service, and work status. Results: Among 757<br />

patients (increased N due to involuntary attrition), demographic variables<br />

were balanced between groups except age (t�2.13; p�.03) with a younger<br />

INT group (mean 1.97 year difference). Of the 562 patients who completed<br />

final reports, the control group reported significantly higher SDS scores (t�<br />

1.98; p � .048). A significant interaction between study group and age was<br />

observed; significantly lower SDS scores were found in the ESRA-C INT<br />

group among patients � 50 years (-1.95; p�.002), and no significant<br />

difference among patients � 50. Conclusions: Adding self-care and<br />

communication coaching to ESRA-C, a system to allow self-report and<br />

clinician notification <strong>of</strong> SQI, reduced symptom distress in a large sample <strong>of</strong><br />

patients during and after active cancer treatment compared with ESRA-C<br />

alone. Patients over the age <strong>of</strong> 50, in particular, may benefit from the<br />

intervention.<br />

9010 <strong>Clinical</strong> Science Symposium, Sat, 3:00 PM-4:30 PM<br />

EXCAP exercise to improve fatigue, cardiopulmonary function, and strength:<br />

A phase II RCT among older prostate cancer patients receiving radiation<br />

and androgen deprivation therapy. Presenting Author: Karen Michelle<br />

Mustian, University <strong>of</strong> Rochester Medical Center, Rochester, NY<br />

Background: Radiation therapy (RT) and androgen deprivation therapy<br />

(ADT) result in cancer-related fatigue (CRF), decreased cardiopulmonary<br />

function (CPF) and decreased strength. Research suggests exercise can<br />

improve CRF during RT and ADT, through physical conditioning responses<br />

that improve CPF and strength. We explored the influence <strong>of</strong> an individuallytailored,<br />

home-based exercise intervention (EXCAP), including progressive<br />

resistance and aerobic training, on CRF, CPF and strength. Methods: Older<br />

prostate cancer patients (N�58; mean age�67), receiving RT (47%) or<br />

ADT (53%), were randomized to 6 wks <strong>of</strong> EXCAP (7 days/wk) or standard<br />

care (RT or ADT with no exercise). CPF (VO2 max) was assessed via graded<br />

exercise testing (GXT) or a 6-minute walk test when GXT was contraindicated.<br />

Muscular strength was assessed using multiple repetition maximum<br />

testing (chest press and leg extension). CRF was assessed via valid<br />

self-report questionnaires (BFI, POMS-FI, MFSI). All assessments were<br />

pre- and post-intervention. Results: ANCOVAs, controlling for baseline,<br />

revealed significant differences between groups in mean levels <strong>of</strong> CRF on<br />

the BFI and POMS-FI (all p�0.05), and a trend toward differences on the<br />

MFSI (p�0.10) with significant baseline interactions (all p�0.05) postintervention:<br />

exercisers decreased CRF while controls increased. ANCOVAs<br />

revealed a trend toward differences between groups in mean levels <strong>of</strong> CPF<br />

(VO2 max) and strength (all p�0.10): exercisers improved while controls<br />

declined in performance. Pearson correlations revealed significant inverse<br />

associations between changes in CRF (BFI) and CPF (p�0.05;r�-0.0.36),<br />

and CRF and strength (p�0.05;r�-0.0.31). MANOVA revealed that changes<br />

in CPF and strength significantly predicted changes in CRF (p�0.05,<br />

r�0.67) and accounted for 45% <strong>of</strong> the variance. Conclusions: Exercise<br />

improves CRF and these improvements may be mediated, in part, by<br />

improvements in CPF and strength. Future phase III RCTs with prostate<br />

cancer patients receiving RT and ADT are needed to confirm these<br />

relationships. Funding: DOD W81XWH-07-1-0341, NCI K07CA120025,<br />

NCI 1R25CA102618.<br />

Patient and Survivor Care<br />

569s<br />

9009 <strong>Clinical</strong> Science Symposium, Sat, 3:00 PM-4:30 PM<br />

Physical activity participation and functional limitations in geriatric cancer<br />

survivors. Presenting Author: Lisa Sprod, University <strong>of</strong> Rochester Medical<br />

Center, Rochester, NY<br />

Background: Functional limitations (FL) increase with age, as does cancer<br />

incidence. Treatments for cancer may exacerbate age-related FL. Physical<br />

activity (PA) reduces the risk <strong>of</strong> recurrence <strong>of</strong> some cancers and may<br />

improve survival. FL may reduce PA participation in geriatric cancer<br />

survivors (�65 yrs.) which could increase the risk <strong>of</strong> recurrence and reduce<br />

survival. This investigation describes and compares patterns <strong>of</strong> PA and FL<br />

in geriatric cancer survivors versus those without a cancer history. Methods:<br />

Using a national sample <strong>of</strong> community-dwelling elders (� 65 yrs.) from the<br />

2003 Medicare Current Beneficiary Survey (N�14,887), we characterized<br />

the differences between cancer survivors and those without a cancer history<br />

in FL, current amount <strong>of</strong> PA, and current amount <strong>of</strong> PA compared to PA one<br />

year prior. Respondents rated FL on a 1-5 scale (1�no difficulty, 5�can’t<br />

do): stooping, crouching, or kneeling (stoop), carrying objects up to 10 lbs<br />

(lift), extending arms above shoulder level (reach), grasping small objects<br />

(grasp), and walking ¼ <strong>of</strong> a mile (walk). Frequency <strong>of</strong> walking for a least 10<br />

minutes (1-5 rating scale; 1�daily, 5�never), weekly participation in PA,<br />

exercise, or sports (yes/no), and time spent doing moderate or vigorous PA<br />

(hrs/wk) were reported. Multivariate logistic regression was used to determine<br />

associations. Results: Of the 14,887 participants, 2,603 (6%)<br />

reported a history <strong>of</strong> cancer. Compared to those without a cancer history, a<br />

greater proportion <strong>of</strong> cancer survivors reported having difficulty or being<br />

unable to stoop, lift, reach, grasp or walk (all p�0.01). Cancer survivors<br />

who had more FL were less likely to engage in PA (all p�0.01). Cancer<br />

survivors reported a lower frequency <strong>of</strong> walking at least 10 minutes at a<br />

time (p�0.01). Cancer survivors were more likely to decrease PA from the<br />

previous year (p�0.01) and spent less time doing moderate (p�0.01) or<br />

vigorous activity (p�0.01) than those without a cancer history. Conclusions:<br />

Older cancer survivors engage in less PA and are at greater risk <strong>of</strong> FL than<br />

those without a history <strong>of</strong> cancer. This may lead to reduced independence,<br />

a greater risk <strong>of</strong> cancer recurrence, and reduced survival. Therefore, PA<br />

interventions are important in this population.<br />

9011 <strong>Clinical</strong> Science Symposium, Sat, 3:00 PM-4:30 PM<br />

Correlation <strong>of</strong> short telomeres (ST) with vulnerability, toxicity, and early<br />

death in elderly AOC patients receiving carboplatin: A multicenter GINECO<br />

trial. Presenting Author: Claire Falandry, Centre Hospitalier Lyon Sud,<br />

Lyon, France<br />

Background: Age induces a progressive decline in the functional reserve and<br />

interferes with cancer treatments. As aging is heterogeneous, this decline<br />

has to be assessed individually. Telomere attrition leads to tissue senescence.<br />

We tested the hypothesis that telomere lenght (TL) could predict pts<br />

vulnerability and outcome during cancer treatment. Methods: This study<br />

was performed in the “Elderly women” GINECO trial designed to evaluate<br />

the impact <strong>of</strong> geriatric covariates on survival in AOC pts over 70 receiving 6<br />

courses <strong>of</strong> carboplatin. TL was estimated in duplicate using standard<br />

Terminal Restriction Fragment analysis from peripheral blood cells at<br />

inclusion and tested for its correlation with geriatric covariates and pts<br />

outcomes (TC and tolerance, overall survival: OS). Results: TL (in base pair)<br />

was estimated for 109/111 pts (median 5997; extremes [4517-8333]).<br />

No significant correlation was found with any pts characteristics. With a<br />

cut<strong>of</strong>f <strong>of</strong> 5770 bp, TL discriminated two groups with significantly different<br />

Treatment Completion (TC) rates: 0.80 (95CI[0.71-0.89]) and 0.59<br />

(95CI[0.41-0.76]), OR�2.8, p�0.02 for long telomere (LT) and short<br />

telomere groups, respectively . ST pts were at higher risk <strong>of</strong> severe adverse<br />

events (SAE, OR�2.7; p�0.02) and tended to have more unplanned<br />

hospital admissions (OR�2.1; p�0.08). Considering OS, after adjustment<br />

on FIGO stage, TL shorter than the median was a nearly significant risk<br />

factor <strong>of</strong> premature death (HR�1.57; p�0.06. Finally, we addressed if TL<br />

correlated with our previously validated geriatric vulnerability score (GVS)c<br />

including ADL score�6, IADL score�25, albuminemia�35g/l,<br />

lymphopenia�1G/L, HADS score£15 as risk factors <strong>of</strong> poorer survival.<br />

Despite no significant correlation with any <strong>of</strong> these factors, GVS³3) and ST<br />

tended to be correlated (OR�2.1; p�0.08). Conclusions: This exploratory<br />

study identifies TL as predictive factor <strong>of</strong> decreased TC, SAE risk,<br />

unplanned hospital admissions and OS after adjustment on FIGO stage.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


570s Patient and Survivor Care<br />

9012 <strong>Clinical</strong> Science Symposium, Sat, 3:00 PM-4:30 PM<br />

Prognostic factors <strong>of</strong> the loss <strong>of</strong> autonomy (LoA) in older patients with<br />

cancer receiving first-line chemotherapy. Presenting Author: Pierre-Louis<br />

Soubeyran, Institut Bergonie, Bordeaux, France<br />

Background: Actual data on prognostic geriatric factors <strong>of</strong> health outcomes<br />

are lacking. We aimed to examine whether a decrease in autonomy for<br />

activities <strong>of</strong> daily living (ADL) after a first cycle <strong>of</strong> chemotherapy influences<br />

elderly cancer pts prognosis, and to determine prognostic factors <strong>of</strong> this<br />

LoA from a range <strong>of</strong> biological and geriatric evaluation factors. Methods:<br />

Pts� 70 years receiving 1st-line chemotherapy for a range <strong>of</strong> cancers were<br />

included in this multicentre prospective study. A LoA was defined as a<br />

decrease <strong>of</strong> 0.5 or more points on the ADL scale between the beginning <strong>of</strong><br />

treatment and the 2nd cycle (ADL scored 6 to 0). The association between<br />

a LoA and OS was examined and prognostic factors were sought from the<br />

pre-treatment Geriatric Assessment data (CIRS-G, IADL, MNA, MMSE,<br />

GDS et Get up and Go) and from baseline biological and clinical information<br />

(age, sex, tumor extension and localization, performance status, BMI,<br />

weight loss, albumin, CRP, haemoglobin levels, leucocyte and platelet<br />

count, creatinine clearance). Pts completely dependent at baseline (ADL<br />

score 0) were excluded as a LoA was not possible. Results: Among 364 pts<br />

included, 299 pts were evaluable and 50 experienced a LoA. A LoA was<br />

significantly associated with an increased risk <strong>of</strong> death (RR 1.518,<br />

95%CI[1.079-2.135]). Biological and clinical factors were not associated<br />

with LoA but pre-treatment low GDS15, dependencies on the IADL, low<br />

MMS, slow Get Up and Go, low ECOG-PS and poor MNA were found to be<br />

prognostic <strong>of</strong> a LoA in univariate analyses. In the multivariate model, low<br />

GDS (OR 2.4, p�0.01, 95%CI [1.23-4.66]) and dependencies on the<br />

IADL (OR 3.0, p� 0.027, 95%CI � [1.13-9.09]) were independently<br />

associated with an increased risk <strong>of</strong> LoA. Conclusions: Our study underlines<br />

the close link between LoA during treatment and poorer prognosis <strong>of</strong> elderly<br />

pts with cancer. Pts with a pre-treatment low GDS15 and IADL-dependent<br />

were the most likely to experience LoA after the 1st chemotherapy cycle.<br />

This research suggests that the GDS15 and IADL should be included as<br />

part <strong>of</strong> any screening for elderly pts before treatment.<br />

9014 <strong>Clinical</strong> Science Symposium, Tue, 9:45 AM-11:15 AM<br />

Falls, physical performance deficits, and functional losses in cancer<br />

survivors with chemotherapy-induced neuropathy (CIPN): A University <strong>of</strong><br />

Rochester CCOP study. Presenting Author: Supriya Gupta Mohile, University<br />

<strong>of</strong> Rochester Medical Center, Rochester, NY<br />

Background: CIPN impairs quality <strong>of</strong> life in cancer survivors. Little is known<br />

about the prevalence <strong>of</strong> falls, physical performance (PP) deficits, and<br />

functional losses or their association with CIPN toxicities in cancer<br />

survivors. Methods: We conducted an analysis <strong>of</strong> baseline assessments from<br />

a phase III randomized clinical trial in cancer survivors with CIPN self<br />

reported pain scores <strong>of</strong> � 4 reflecting leg and foot pain from neuropathy<br />

over the past 24 hours on a scale from 0 (“no pain”) to 10 (“pain as bad as<br />

you can imagine”). Patients also completed EORTC QLQ-CIPN-20 sensory<br />

and motor scales for neuropathy toxicities and self reported falls in the<br />

previous 3 months. A PP deficit was defined as “a lot <strong>of</strong> difficulty” or<br />

“unable to do” any <strong>of</strong> 6 physical tasks (e.g., lifting objects, walking a<br />

quarter <strong>of</strong> a mile). Functional losses were defined as “a lot <strong>of</strong> difficulty” or<br />

“unable to do” any <strong>of</strong> 5 functional tasks (e.g., managing money, bathing).<br />

We examined the association <strong>of</strong> baseline characteristics and CIPN toxicities<br />

with falls, PP deficits and functional losses using logistic regression.<br />

Results: Of 421 patients, 11.9% experienced recent falls, 58.6% reported<br />

a PP deficit, and 26.6% reported a functional loss. Patients with falls<br />

and/or PP deficits were more likely to be older (mean age 60.9 vs 58.9,<br />

p�0.02), female (75.3% vs 65.2%, p�0.03) and have less education<br />

(�high school: 7.1% vs 0.6%, p�.01). Cancer and chemotherapy history<br />

were not different between groups. Patients with falls and/or PP deficits<br />

reported higher severity <strong>of</strong> CIPN toxicities: pain (6.82 vs 6.05, p�0.01),<br />

sensory (23.3 vs 19.6, p�0.01), and motor (17.4 vs 12.7, p�0.01). In<br />

multivariable analysis, factors associated with having a fall and/or PP<br />

deficit included: older age (OR 1.03, p�0.04), low education (OR 9.34,<br />

p�0.04), and motor toxicity (OR 1.21, p�0.01). Factors associated with<br />

functional losses included: non-white race (OR 3.16, p�0.01), Hispanic<br />

ethnicity (OR 5.32, p�0.05), motor toxicity (OR 1.19, p�0.01), and PP<br />

deficit (OR 4.94, p�.01). Conclusions: CIPN toxicities, primarily motor, are<br />

significantly associated with falls, physical performance deficits, and<br />

functional losses.<br />

CRA9013 <strong>Clinical</strong> Science Symposium, Tue, 9:45 AM-11:15 AM<br />

CALGB 170601: A phase III double blind trial <strong>of</strong> duloxetine to treat painful<br />

chemotherapy-induced peripheral neuropathy (CIPN). Presenting Author:<br />

Ellen M. Lavoie Smith, University <strong>of</strong> Michigan, Ann Arbor, MI<br />

The full, final text <strong>of</strong> this abstract will be available at<br />

abstract.asco.org at 12:01 AM (EDT) on Monday, June 4,<br />

2012, and in the <strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong> online<br />

supplement to the June 20, 2012, issue <strong>of</strong> Journal <strong>of</strong><br />

<strong>Clinical</strong> Oncology. Onsite at the <strong>Meeting</strong>, this abstract will<br />

be printed in the Monday edition <strong>of</strong> ASCO Daily News.<br />

9015 <strong>Clinical</strong> Science Symposium, Tue, 9:45 AM-11:15 AM<br />

Rates and risks <strong>of</strong> suicidality in young adults (YAs) with advanced cancer.<br />

Presenting Author: Kelly Marie Trevino, Dana-Farber Cancer Institute,<br />

Boston, MA<br />

Background: Suicide rates in YA cancer patients are higher than in the<br />

general population. Although cancer is associated with a four-fold increase<br />

in the likelihood <strong>of</strong> a suicide attempt, little is known about suicidality in<br />

YAs with cancer. This study examined rates and clinical risk factors<br />

associated with suicidality in a sample <strong>of</strong> YAs with advanced cancer.<br />

Methods: Structured interviews were conducted between 4/2010 and<br />

9/2011 with 70 YA advanced cancer patients (range 20-40 yrs, M�33.97,<br />

SD�5.61) receiving care at the Dana-Farber Cancer Institute. Validated<br />

measures assessed suicidality (i.e., Yale Evaluation <strong>of</strong> Suicidality), quality<br />

<strong>of</strong> life, major depressive disorder, grief over cancer-related losses, and<br />

social support. Scores on the suicidality measure were dichotomized into<br />

positive screen � 1 and negative screen � 0. Chi-square, t-test, and<br />

logistic regression analyses evaluated the relationship between suicidality<br />

and participant characteristics and psychosocial variables, controlling for<br />

confounding variables. Results: Over one-fifth (21.4%) <strong>of</strong> the sample<br />

screened positive for suicidality. Female gender �2 (1, N � 70) � 4.95, p �<br />

.026), breast compared with other cancer diagnosis �2 (1, N � 70) � 5.66,<br />

p � .017), and better performance status (t(68) � 3.13, p � .01) were<br />

associated with lower rates <strong>of</strong> suicidality. <strong>Part</strong>icipants who met criteria for<br />

current (OR [95% CI] 8.67 [1.78, 42.22]) or lifetime major depressive<br />

disorder (5.38 [1.60, 18.12]) endorsed higher rates <strong>of</strong> suicidality. Better<br />

overall (.97 [.94, .99]), psychological (.93 [.87, .94]), and existential<br />

quality <strong>of</strong> life (.91 [.85, .98]) were associated with reduced suicidality risk.<br />

More severe grief was associated with greater risk (1.15 [1.04, 1.28])<br />

whereas greater social support was associated with lower suicidality risk<br />

(.85 [.74, .97]). Conclusions: YAs with advanced cancer reported higher<br />

rates <strong>of</strong> suicidality than observed in other age groups. Developmentally<br />

targeted interventions that promote physical function, effectively treat<br />

depression, improve quality <strong>of</strong> life and reduce grief, and provide opportunities<br />

for social support may reduce rates <strong>of</strong> and risk for suicidality in this<br />

population.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


9016 <strong>Clinical</strong> Science Symposium, Tue, 9:45 AM-11:15 AM<br />

Patterns <strong>of</strong> antidepressant use in cancer patients (pts): An analysis from<br />

SOAPP (ECOG E2Z02: Symptom Outcomes and Practice Patterns). Presenting<br />

Author: Judith Manola, Dana-Farber Cancer Institute, Boston, MA<br />

Background: Antidepressant (AD) use is common in outpatient oncology,<br />

but the pattern and determinants <strong>of</strong> prescribing for commonly used AD are<br />

unknown. Methods: 3106 pts with cancer <strong>of</strong> the breast, prostate, colon/<br />

rectum, or lung were enrolled from multiple sites in a study <strong>of</strong> symptoms.<br />

Five depression case-finding methods were explored : 3 based on MDASI<br />

items reported by patients (1) sadness/ depression��4, (2) distress��4,<br />

(3) interference with mood��7 or enjoyment��7; 2 based on clinician’s<br />

report (4) presence <strong>of</strong> psychological distress, (5) depression being listed as<br />

one <strong>of</strong> top 3 symptoms. AD use (excluding tricyclic antidepressants and<br />

psychostimulants) was examined by depression status. Logistic regression<br />

models were used to examine the effect <strong>of</strong> demographic and clinical<br />

characteristics on AD use. Results: Rates <strong>of</strong> depressive symptoms varied by<br />

casefinding method (1�29%, 2�28%, 3�14%, 4�24%, 5�11%); 47%<br />

(1457) pts were defined as having depressive symptoms by at least one<br />

method. AD were prescribed in 25% <strong>of</strong> depressed pts compared to 14% <strong>of</strong><br />

non-depressed pts. After adjusting for other covariates, factors associated<br />

with greater use <strong>of</strong> AD included depression (OR�1.7, P�0.01), family<br />

history <strong>of</strong> depression (OR�2.2, P�0.01), female sex (OR�1.8, P�0.01),<br />

younger age (OR�1.2, P�0.04), non-Hispanic White race (OR�2.0,<br />

P�0.01), prior chemo/immune/ hormonal treatment (OR�1.5, P�0.01),<br />

more concurrent medication use (OR�3.3, P�0.01), anxiolytics use<br />

(OR�2.0, P�0.01), sedative use (OR�2.1, P�0.01), receiving counseling<br />

(OR�1.6, P�0.04), patient’s perception <strong>of</strong> poor QOL (OR�1.3,<br />

P�0.02), duration <strong>of</strong> current treatment �1 year (OR�1.6, P�0.01), and<br />

being enrolled by a CCOP (OR�1.8, P�0.01). These significant associations<br />

remained regardless <strong>of</strong> the case-finding method used for depression,<br />

with only slight changes in the magnitude <strong>of</strong> ORs. Conclusions: Depressive<br />

symptoms are common in outpatients with cancer. One-fourth <strong>of</strong> solid<br />

tumor pts with depressive symptoms are taking an AD. Antidepressant<br />

prescribing varies by type <strong>of</strong> institution, race/ethnicity, age, concomitant<br />

medication exposure and several other clinical factors.<br />

9018 Poster Discussion Session (Board #2), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

SWOG S0715: Randomized placebo-controlled trial <strong>of</strong> acetyl-L-carnitine<br />

for the prevention <strong>of</strong> taxane-induced neuropathy during adjuvant breast<br />

cancer therapy. Presenting Author: Dawn L. Hershman, Columbia University<br />

Medical Center, New York, NY<br />

Background: Chemotherapy-induced peripheral neuropathy (CIPN) is a<br />

common, disabling side effect <strong>of</strong> taxanes that leads to suboptimal<br />

treatment and diminished quality <strong>of</strong> life. Acetyl-L-Carnitine (ALC) is a<br />

natural compound involved in tubulin acetylation and neuronal protection.<br />

Pre-clinical and phase II studies suggest ALC may be effective for the<br />

prevention and treatment <strong>of</strong> CIPN. Methods: A randomized, double-blind,<br />

multi-center, phase III trial comparing ALC (1000 mg TID) vs placebo for<br />

24 wks in women with stage I-III breast cancer undergoing adjuvant taxane<br />

therapy was conducted. The primary objective was to determine if ALC<br />

prevents CIPN as measured by the 11-item neurotoxicity (NTX) component<br />

<strong>of</strong> the FACT-Taxane scale (low score � worse NTX) at 12 wks. Secondary<br />

objectives included change at 24 wks, change in the Trial Outcome Index<br />

(TOI), fatigue (FACIT Fatigue) and NTX grade. Patients were stratified by<br />

planned treatment, and age (�60, �60). Results: 409 patients were<br />

evaluable (208 ALC, 201 placebo). No imbalances were observed by age,<br />

ethnicity, race, planned taxane treatment, performance status, or stage.<br />

The mean FACT-NTX score was 5.2 points lower at 12 wks on ALC and 4.5<br />

points lower on placebo. In a linear regression adjusting for baseline score,<br />

planned treatment and age, wk 12 scores were 0.9 points lower on ALC<br />

than placebo (95% CI: -2.2 to 0.4, p�.17). At 24 wks, the mean observed<br />

FACT-NTX score was lower for ALC (5.3 vs. 3.6). In the multivariate model,<br />

wk 24 scores were 1.8 points lower on ALC (95% CI: -3.2 to -0.4, p�.01),<br />

representing more self-reported NTX. Grade 3/4 NTX was more frequent in<br />

the ALC arm (p�.04). Also at 24 wks, 38% on ALC had �5 point decrease<br />

score compared to 28% on placebo (OR�1.57, p�.05), and FACT-TOI<br />

scores were 3.5 points lower on ALC (95% CI: -6.5 to -0.4, p�.03). No<br />

differences between arms were observed for the FACIT-Fatigue scale or for<br />

other toxicities. Conclusions: There is no evidence that ALC has a positive<br />

impact on CIPN at 12 wks. However, ALC increases CIPN by 24 wks.<br />

Correlative studies are ongoing to explain this unexpected finding. Patients<br />

should be discouraged from using ALC and other supplements without<br />

proven efficacy.<br />

Patient and Survivor Care<br />

571s<br />

9017 Poster Discussion Session (Board #1), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

A prospective study to evaluate the efficacy and safety <strong>of</strong> oral acetyl-Lcarnitine<br />

(ALC) in treatment <strong>of</strong> chemotherapy-induced peripheral neuropathy<br />

(CPIN). Presenting Author: Yuanjue Sun, Sixth Affiliated Hospital <strong>of</strong><br />

Shanghai Jiaotong University, Shanghai, China<br />

Background: Chemotherapy-induced peripheral neuropathy (CIPN) is a<br />

major side effect <strong>of</strong> many commonly used chemotherapeutic. This side<br />

effect <strong>of</strong> chemotherapy can be debilitating and effective treatment for CIPN<br />

remains elusive. Previous studies demonstrated that Acetyl-L-Carnitine<br />

(ALC) is effective in attenuating CIPN, controlled study is needed to<br />

substantiate ALC’s effect in treatment <strong>of</strong> CIPN. Methods: This study was<br />

designed to evaluate the efficacy and safety <strong>of</strong> Acetyl-L-Carnitine (ALC)<br />

Hydrochloride Enteric-coated Tablet (oral administration) in the treatment<br />

<strong>of</strong> CIPN. It was a prospective, randomized, double-blinded, placebocontrolled<br />

and paralleled clinical study (registration No. 2007L03540). Of<br />

239 subjects enrolled in the study (NCI grade 2 or above), 118 subjects<br />

received 3g/day ALC orally for 8 weeks and 121 received placebo. Primary<br />

endpoint was set as improvement <strong>of</strong> peripheral neuropathy at least 1 grade<br />

and assessment was made in week 4, 8 and 12 after enrollment. Results: In<br />

full analyses set (FAS) and per-proposal set (PPS), the peripheral sensory<br />

neuropathy was significantly ameliorated in ALC group with 50.5% and<br />

51.6% patients meeting the primary endpoint at week 8 and 12 respectively<br />

while only 24.1% and 23.1% <strong>of</strong> patients in the placebo group at week<br />

8 and 12 respectively (p�0.001 in both sets). Secondary endpoint such as<br />

nerve electrophysiological test and Physical Condition Score (Karn<strong>of</strong>sky<br />

performance status, KPS) were also significantly improved in patients with<br />

ALC treatment (in FAS, P�0.0463 and P�0.022; in PPS, P�0.0076and<br />

P�0.0064, respectively). Cancer-related fatigue was significantly alleviated<br />

after ALC treatment in PPS (P�0.0135). Safety: 236 subjects were<br />

included in safety assessment and 41 patients experienced 62 adverse<br />

events during the course <strong>of</strong> study. There was no significant difference in<br />

AE/SAE incidence between the two groups (P�0.3903). Conclusions: Oral<br />

administration <strong>of</strong> ALC is effective in attenuating CIPN as well as in<br />

reducing cancer-related fatigue and improving physical conditions in<br />

cancer patients. The treatment <strong>of</strong> oral ALC is safe and well tolerated.<br />

9019 Poster Discussion Session (Board #3), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Low-level laser therapy for chemotherapy-induced peripheral neuropathy.<br />

Presenting Author: John Muzi Lee, Legacy Health Systems, Portland, OR<br />

Background: Chemotherapy induced peripheral neuropathy (CIPN) is a<br />

common and serious side effect from chemotherapy agents. Low-level laser<br />

light therapy (LLLT) devices were approved in 2002 for pain management.<br />

Studies suggest a local release <strong>of</strong> serotonin, increased mitochondrial ATP<br />

production, or anti-inflammatory effects as a mechanism <strong>of</strong> action. We then<br />

questioned whether LLLT would show efficacy in mitigating symptoms<br />

caused by CIPN. Methods: In a single center prospective randomized trial,<br />

participants were randomized to receive either treatment with LLLT twice a<br />

week for 8 weeks or placebo LLLT twice a week for 4 weeks followed by<br />

actual treatment twice a week for 4 weeks. FACT/GOG-NTX, Brief Pain<br />

Inventory (BPI -Severity and Interference), SF-36 Quality <strong>of</strong> Life (Physical<br />

and Mental Score), mon<strong>of</strong>ilament and function testing (buttoning, coin use<br />

and walking) were conducted prior to initiation <strong>of</strong> therapy, during, at<br />

completion, and 2 months after treatment. 20 participants, 16 women and<br />

4 men (average age 58 and 63 respectively), were enrolled between<br />

October 2009 and June 2010. 10 patients were randomized to each group.<br />

Average time from end <strong>of</strong> chemotherapy to enrollment was 32.6 months<br />

(range 2 - 120 mo). All patients had neuropathic involvement <strong>of</strong> the feet.<br />

14 patients had additional involvement <strong>of</strong> the hands. Results: Compared to<br />

baseline, patients receiving any amount <strong>of</strong> active treatment showed<br />

significant improvement at 8 weeks in NTX, BPI, function testing, and<br />

SF36 Mental score. Those receiving 4 weeks <strong>of</strong> placebo treatment showed<br />

improvement in only BPI, NTX and SF36 Mental score. At 2-month follow<br />

up, all 20 patients showed a significant improvement in walking and SF 36<br />

mental score, suggesting 4 weeks <strong>of</strong> active treatment improved function.<br />

No significant difference in mon<strong>of</strong>ilament testing was observed throughout<br />

the study in either group. Direct comparison between 4 or 8 weeks <strong>of</strong><br />

treatment vs. placebo showed a statistically significant difference in<br />

walking function at 2-months. All patients tolerated therapy well without<br />

side effects. Conclusions: Low-level laser light therapy improved functional<br />

test over placebo and may be a viable option for non-medical management<br />

<strong>of</strong> CIPN. Further study <strong>of</strong> LLLT in CIPN is warranted.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


572s Patient and Survivor Care<br />

9020 Poster Discussion Session (Board #4), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Patient-reported cognitive impairments among women with breast cancer<br />

randomly assigned to hormonal therapy (HT) alone versus chemotherapy<br />

followed by hormonal therapy (C�HT): Results from the Trial Assigning<br />

Individualized Options for Treatment (TAILORx). Presenting Author: Lynne<br />

I. Wagner, The Robert H. Lurie Comprehensive Cancer Center <strong>of</strong> Northwestern<br />

University, Chicago, IL<br />

Background: Cognitive impairment is a complication <strong>of</strong> chemotherapy.<br />

Perceived cognitive impairments (PCI) were prospectively assessed among<br />

TAILORx participants randomized to HT alone versus chemotherapy followed<br />

by HT (C�HT). Methods: TAILORx participants with an OncoType DX<br />

Recurrence Score 11-25 were randomly assigned to HT or C�HT. PCI,<br />

fatigue, endocrine symptoms and health-related quality <strong>of</strong> life (HRQL) were<br />

assessed at baseline, 3, 6, 12, 24, and 36 months, using the Functional<br />

Assessment <strong>of</strong> Cancer Therapy (FACT) in 455 patients enrolled after<br />

1/15/10. PCI change scores � 4.5 from baseline were defined a priori as<br />

clinically meaningful. Linear regression (LR) was used to model PCI scores<br />

on baseline PCI, treatment and other factors. Results: PCI scores were<br />

significantly worse at 3, 6, and 12 months compared to baseline for both<br />

groups (Table). The decline was greater for C�HT than HT at 3 months, but<br />

scores were similar at 12 months. Tests <strong>of</strong> an interaction between<br />

menopausal status and treatment were non-significant. PCI correlated with<br />

fatigue (r � 0.57-0.64) but not FACT Emotional well-being (EWB; r �<br />

0.28-0.38); controlling for EWB did not account for differences in PCI<br />

change scores between treatment arms. Conclusions: Our study is the first<br />

to examine PCI among breast cancer patients randomized to receive C�HT<br />

vs. HT alone. C�HT was associated with greater declines in PCI at 3<br />

months, but at 12 months PCI was similar in the C�HT and HT groups. PCI<br />

was associated with fatigue but not EWB. Pre- and post-menopausal groups<br />

demonstrated the same pattern <strong>of</strong> change. Since this study did not include<br />

a control group <strong>of</strong> patients not treated with HT, further study is required to<br />

determine if and to what extent HT contributes to PCI.<br />

Mean change in PCI score from baseline.<br />

3 months 6 months 12 months<br />

Change<br />

scores Difference<br />

(LR) P value Change<br />

scores Difference<br />

(LR) P value Change<br />

scores Difference<br />

(LR) P value<br />

HT -2.78 -3.61 � 0.001 -3.27 -2.28 � 0.05 -4.67 1.20 n.s.<br />

C�HT -6.40 -5.66 -6.08<br />

9022 Poster Discussion Session (Board #6), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Subjective cognitive complaints one year after ceasing adjuvant endocrine<br />

therapy for early-stage breast cancer: Findings from the Breast International<br />

Group (BIG) 1-98 trial. Presenting Author: Kelly-Anne Phillips, Peter<br />

MacCallum Cancer Centre, Melbourne, Australia<br />

Background: We have previously reported that, in the BIG 1-98 trial,<br />

objective cognitive function improved in postmenopausal women one year<br />

after cessation <strong>of</strong> adjuvant endocrine therapy for breast cancer. Here we<br />

evaluate changes in subjective cognitive function (SCF). Methods: One<br />

hundred postmenopausal women, randomized to receive five years <strong>of</strong><br />

adjuvant tamoxifen, letrozole, or sequences <strong>of</strong> both, completed selfreported<br />

measures on SCF, psychological distress, fatigue and quality <strong>of</strong><br />

life during the fifth year <strong>of</strong> trial treatment (year 5) and one year after<br />

treatment completion (year 6). Changes between years 5 and 6 were<br />

evaluated using the Wilcoxon signed-rank test. SCF and its correlates were<br />

explored. Results: Mean age <strong>of</strong> participants was 63.9 years [SD�7.1 years].<br />

SCF and the other patient-reported outcomes did not change significantly<br />

after cessation <strong>of</strong> endocrine therapy with the exception <strong>of</strong> improvement in<br />

hot flushes (p�0.0005). No difference in changes was found between<br />

women who were taking tamoxifen or letrozole at year 5. SCF was the only<br />

psychosocial outcome with a substantial correlation between year 5 and 6<br />

(Spearman’s R�0.80). Correlations between SCF and the other patientreported<br />

outcomes were generally low. Conclusions: Although objective<br />

cognitive function improved after cessation <strong>of</strong> adjuvant endocrine therapy<br />

in the BIG 1-98 trial, improvement in SCF was not evident. The underlying<br />

reason for the clear disconnect between objective and subjective cognitive<br />

function seen in this and most other studies, is a crucial issue. It should be<br />

a research priority in order to effectively tackle the concerns <strong>of</strong> women<br />

about their cognition during and after breast cancer treatment.<br />

9021 Poster Discussion Session (Board #5), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Cognitive function and fatigue in colorectal cancer (CRC) patients: A<br />

prospective longitudinal controlled study. Presenting Author: Janette L.<br />

Vardy, Sydney Cancer Centre, University <strong>of</strong> Sydney, Sydney, Australia<br />

Background: A subset <strong>of</strong> cancer patients has cognitive impairment and<br />

fatigue after chemotherapy (CTh). We evaluated these symptoms and<br />

potential mechanisms in CRC patients and healthy controls (HC). Methods:<br />

Cognitive function was evaluated in CRC patients and HC at baseline<br />

(pre-CTh), 6 and 12 months. Group 1A (Stage II/III) received CTh and Gr<br />

1B (Stage I/II) no CTh. Gr 2 had limited metastatic CRC. All subjects<br />

completed cognitive assessment and questionnaires for fatigue, QOL,<br />

anxiety/depression, and perceived cognitive function. Blood tests evaluated:<br />

10 cytokines, clotting factors, sex hormones, CEA, CBC and apolipoprotein<br />

genotyping as potential causal factors. Primary endpoints: cognitive<br />

function and fatigue (Gr. 1A & 1B) at 12 months. Associations between<br />

results and demographic and disease-related factors were sought. Results:<br />

359 CRC patients (173 Gr 1A, 116 Gr 1B, 70 Gr. 2) and 72 HC were<br />

assessed. Median age 58 (23-75 years); 58% male. Cognitive impairment:<br />

baseline Gr 1A 34% vs 1B 32.5% (p�.9), Gr 1 33% vs HC 10%<br />

(p�0.001); 12 months Gr 1A 21% vs 1B 16% (p�0.43), Gr 1 19% vs HC<br />

2% (p�0.001). Cognitive domains most affected: verbal & visual memory,<br />

processing speed. Men had greater impairment than women (p�0.009).<br />

Cognitive decline from 0-12 months: Gr 1A 30%, Gr 1B 19%, Gr 2 24%,<br />

and HC 16%. Perceived cognitive impairment at 12 months: Gr 1A 19% vs<br />

1B 7% (p�.04), Gr 1 14% vs HC 0% (p�.009). Fatigue was greatest in Gr<br />

1A (70%) at 6 months; at 12 months Gr. 1A 46% vs 1B 31% (p�0.056),<br />

Gr 2 60%, HC 36%. Cytokines were elevated in CRC patients compared to<br />

HC. No association was found with cognitive function and: fatigue, QOL,<br />

anxiety/depression, cytokines, sex hormones, clotting factors, CEA or apoE<br />

genotype. Objective cognitive function was associated with perceived<br />

cognitive function at baseline only. Fatigue at baseline was associated with<br />

hemoglobin and at 12 months with neutrophil count. Conclusions: Compared<br />

to HC, CRC patients had more cognitive impairment at baseline, 6<br />

and 12 months; but impairment was not significantly different between<br />

those who did and did not receive CTh. The mechanisms <strong>of</strong> cognitive<br />

impairment remain unknown. Fatigue improved with time.<br />

9023 Poster Discussion Session (Board #7), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Methylphenidate (MP) and nursing telephone intervention (NTI) for cancerrelated<br />

fatigue (CRF) in advanced cancer patients: A double-blind randomized<br />

phase II trial. Presenting Author: Eduardo Bruera, University <strong>of</strong> Texas<br />

M. D. Anderson Cancer Center, Houston, TX<br />

Background: CRF is the most common and distressing symptom in advanced<br />

cancer patients. Preliminary studies support MP and NTI for CRF<br />

(Bruera et al. JCO 2006). The primary objective <strong>of</strong> our study was to<br />

determine the effect <strong>of</strong> MP as compared to placebo (P). A secondary<br />

objective was to investigate the role <strong>of</strong> NTI as compared to control<br />

telephone intervention (CTI). Methods: Advanced cancer patients with<br />

fatigue �4/10 on the Edmonton Symptom Assessment Scale (ESAS),<br />

normal cognition, no evidence <strong>of</strong> major depression and hemoglobin �8<br />

were eligible. Patients were randomized to 4 groups in a 2x2 factorial<br />

design (MP�NTI, P�NTI, MP�CTI and P�CTI). Primary endpoint was<br />

Functional Assessment <strong>of</strong> Chronic Illness-Fatigue (FACIT-F) subscale<br />

scores between day 15 and baseline. The dose and duration <strong>of</strong> methylphenidate<br />

was 5 mg every two hours, as needed, up to 20 mg/day. We tested the<br />

median difference in FACIT-F subscale scores between the groups using<br />

the Kruskal Wallis test and Wilcoxon signed rank test. Longitudinal<br />

regression analysis was conducted with a mixed model. Results: Total<br />

accrual was 197. Mean (SD) age was 58 (12), female 67% (N�148), white<br />

72% (N�136), gastrointestinal cancers were the most common 22%<br />

(N�41). Baseline FACIT-F subscores were similar among the 4 groups. The<br />

median FACIT-F subscores showed significant improvement between Day<br />

15 and baseline for all four groups except for P�CTI (Table): MP�NTI<br />

(4.5, P�0.004), P�NTI (8, P�0.001), MP�CTI (7, P�0.001), and<br />

P�CTI (5, P�0.06), with no statistically significant difference between MP<br />

and P (6 vs. 6, P�0.89). Longitudinal regression analysis showed a time<br />

effect (P�0.001) and group differences for NTI vs. CTI with FACIT-F<br />

(P�0.13) and ESAS (P�0.03). Grade 3 toxicities were similar between the<br />

MP and P arms (34/93 vs. 24/97, P�0.09). Conclusions: MP was not<br />

effective as compared to P for CRF in advanced cancer patients. NTI may<br />

be effective and should be further studied.<br />

FACIT-F at baseline, day 8 and day 15 group.<br />

Median (interquartile range)<br />

Group Baseline Day 8 Day 15<br />

MP�NTI 21 (12) 31 (14) 25 (16)<br />

P�NTI 20 (13) 32 (14) 28 (13)<br />

MP�CTI 20 (14) 27 (14) 27 (14)<br />

P�CTI 22 (14) 31 (18) 28 (23)<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


9024 Poster Discussion Session (Board #8), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Phase Ib, multicenter, single-blinded, placebo-controlled, sequential doseescalation<br />

study to assess the safety and tolerability <strong>of</strong> topically applied<br />

AG013 in subjects with locally advanced head and neck cancer (LAHNC)<br />

receiving induction chemotherapy (ICT). Presenting Author: Sewanti Atul<br />

Limaye, Dana-Farber Cancer Institute/Harvard Medical School, Boston, MA<br />

Background: Oral mucositis (OM) is a significant toxicity <strong>of</strong> ICT in LAHNC.<br />

AG013, a mouth rinse composed <strong>of</strong> a recombinant Lactococcus lactis<br />

(sAGX0085) engineered to secrete human Trefoil Factor 1 (hTFF1) and<br />

deficient in the gene encoding thymidylate synthase, was investigated in<br />

patients receiving ICT (cisplatin, 5-fluorouracil �/-docetaxel:PF�/-T).<br />

TFFs have wound-healing properties and are protective <strong>of</strong> mucosal tissues.<br />

Methods: To evaluate the safety, tolerability, PK pr<strong>of</strong>ile and efficacy <strong>of</strong><br />

AG013 in attenuating ICT-associated OM, 52 patients (pts) with newly<br />

diagnosed LAHNC receiving ICT (24TPF, 1PF) were screened in ICT cycle<br />

(Cy) 1 for symptomatic OM (SOM). 25 pts who developed SOM in Cy1 were<br />

enrolled into 3 successive groups <strong>of</strong> at least 7 pts each in the active phase<br />

and randomly assigned at the Baseline visit (d1 <strong>of</strong> ICT Cy2) in a 5:2 ratio to<br />

receive AG013 or placebo during ICT Cy2. Dose was escalated in successive<br />

groups from 1 x/day (qd) to 3 x/day (tid) and to 6 x/day, resulting in<br />

doses <strong>of</strong>2x1011 ,6x1011 , and 1.2 x 1012 colony-forming units (CFU)/day<br />

respectively. Pts were assessed daily from Cy 2 d1 to d14 for OM using<br />

WHO criteria. Safety endpoints and PK (mucosal, salivary and blood<br />

samples) were assessed. Results: AG013-sAGX0085 could not be detected<br />

in blood. Recovered oral live bacterial levels were high immediately after<br />

dosing, decreased by 90 minutes post dose, and undetectable at study end.<br />

No differences in hTFF1 serum levels were seen. Pts in the placebo group<br />

had ulcerative OM (UOM) on nearly 60% <strong>of</strong> the days vs. 35-40% days in the<br />

AG013 group. 29 % pts who received AG013 had 0 or 1 day <strong>of</strong> UOM vs. at<br />

least 2 days <strong>of</strong> UOM in the placebo group. Pts who received AG013 on any<br />

dosing schedule had a lower % <strong>of</strong> days with OM, fewer unplanned <strong>of</strong>fice and<br />

emergency room visits compared to pts who received placebo. No differences<br />

were noted in mouth and throat soreness, opioid use, or gastrostomy<br />

tube placement. Conclusions: Topical administration <strong>of</strong> AG013 appears<br />

safe, well tolerated and effective in reducing the severity and course <strong>of</strong> OM<br />

in patients receiving ICT for HNC.<br />

9026^ Poster Discussion Session (Board #10), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Effect <strong>of</strong> cancer cachexia on treatment toxicity in metastatic colorectal<br />

cancer patients: Results <strong>of</strong> a prospective multicenter study. Presenting<br />

Author: Maximilien Barret, Hôpital Européen Georges Pompidou, Department<br />

<strong>of</strong> Gastroenterology, Paris, France<br />

Background: Malnutrition reduces tolerance to treatment and survival in<br />

numerous cancers, including metastatic colorectal cancer (mCRC). Previous<br />

studies have shown that chemotherapy toxicity may be linked to<br />

sarcopenia. We evaluated the effect <strong>of</strong> sarcopenia on chemotherapy toxicity<br />

among mCRC patients. Methods: In this prospective, cross-sectional,<br />

multicenter study, oncological and nutritional data were collected in all<br />

consecutive mCRC patients between March 7th and March 20th 2005 in<br />

three hospitals. Computed tomography (CT) images were analysed using<br />

Slice-O-Matic s<strong>of</strong>tware V4.3 (Tomovision) to evaluate cross-sectional areas<br />

(cm2) <strong>of</strong> muscle tissue (MT), visceral and subcutaneous adipose tissue<br />

(VAT and SAT). The 3rd lumbar vertebra (L3) was chosen as a reference,<br />

since L3 and whole-body measurements are linearly related. Indexed on<br />

height, MT, VAT and SAT (cm2/m2), were computed and described, after<br />

stratification on sex. In accord with the literature, sarcopenia was defined<br />

as MT �55 cm2/m2 for men and �39 cm2/m2 for women. Images were<br />

obtained within one month <strong>of</strong> clinical evaluation. Toxicities were evaluated<br />

according to the NCI-CTC, version 3.0, in the two months following clinical<br />

evaluation. Results: 53 mCRC patients (72% men (M)), participated in the<br />

study. According to body mass index (BMI) only 7% <strong>of</strong> female patients (F)<br />

and 3% M were malnourished (BMI�17 kg/m²), and 93% F and 97% M<br />

were <strong>of</strong> normal weight or overweight (BMI� 25kg/m²). These results are to<br />

be compared to the 38% (F) and 82% (M) <strong>of</strong> patients with sarcopenia.<br />

Grade 3-4 toxicities were observed in 28% <strong>of</strong> the cases, with neurotoxicity<br />

in 6%, diarrhea 2%, anemia 2%, neutropenia 9%, and nausea and<br />

vomiting 6%. In multivariate analysis including age, sex, BMI, sarcopenia,<br />

SAT and VAT, the only factor associated with grade 3-4 toxicity was<br />

sarcopenia (OR� 13.55 95% CI [1.08;169.31], p�0.043). Conclusions:<br />

In mCRC patients undergoing chemotherapy, low muscle tissue was much<br />

more frequently observed (68%) than “visible” malnutrition (4%). Despite<br />

the small number <strong>of</strong> patients included in our study, we showed that<br />

sarcopenia was associated with severe chemotherapy toxicity in mCRC.<br />

Patient and Survivor Care<br />

573s<br />

9025 Poster Discussion Session (Board #9), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Parenteral hydration (PH) in advanced cancer patients: A multi-center,<br />

double-blind, placebo-controlled randomized trial. Presenting Author:<br />

Shalini Dalal, University <strong>of</strong> Texas M. D. Anderson Cancer Center, Houston,<br />

TX<br />

Background: The vast majority <strong>of</strong> cancer patients at the end <strong>of</strong> life receive<br />

PH in hospitals and no PH in hospice. There is limited evidence supporting<br />

either practice. Our preliminary study suggested that PH (1L/day) improved<br />

dehydration symptoms (Bruera et al. JCO 2005). In this randomized<br />

controlled trial, we determined the effect <strong>of</strong> PH on symptoms associated<br />

with dehydration, quality <strong>of</strong> life and survival for patients with advanced<br />

cancer. Methods: We randomly assigned 129 cancer patients from 6<br />

hospices to receive subcutaneous PH (normal saline 1 L/day) or placebo<br />

(normal saline 100 mL/day) daily over 4 hours. The primary outcome was<br />

change in the sum <strong>of</strong> 4 dehydration symptoms (fatigue, myoclonus,<br />

sedation and hallucinations, 0�best and 40�worst possible) between day<br />

4 and baseline. Secondary outcomes included Edmonton Symptom Assessment<br />

Scale (ESAS), Memorial delirium assessment scale [MDAS], Nursing<br />

delirium screening scale [NuDESC], unified myoclonus rating scale [UMRS],<br />

FACIT-F, creatinine, urea, and overall survival. Intention-to-treat analysis<br />

was conducted to examine the change in each variable between baseline<br />

and day 4 or day 7. Log rank test was used for survival analysis. Results:<br />

Mean age was 67 (range 43-92), female 61%, Caucasians 60%, gastrointestinal,<br />

genitourinary and lung cancers 70%, performance status 3-4 113<br />

(88%), with no baseline differences between the hydration (N�63) and<br />

placebo (N�66) groups. No significant differences were found between the<br />

hydration and placebo groups for the sum <strong>of</strong> 4 dehydration symptoms (-3.3<br />

v. -2.8, p�0.77), ESAS (all non-significant), MDAS (1 vs. 3.5, p�0.084),<br />

NuDESC (0 v. 0, p�0.13) and UMRS (0 vs. 0, p�0.54). We also did not<br />

identify any group differences for day 7, except for a decrease in urea level<br />

in the hydration group (-2 vs. 2, P�0.02). Median overall survival was 21<br />

days for PH and 15 days for placebo (p�0.83). Conclusions: PH at 1 L/day<br />

did not improve symptoms, quality <strong>of</strong> life or survival compared to placebo.<br />

Further studies are required to determine if any subgroups would benefit<br />

from PH.<br />

9027 Poster Discussion Session (Board #11), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Curcumin for radiation dermatitis: A randomized, double-blind, placebocontrolled<br />

clinical trial <strong>of</strong> 30 breast cancer patients. Presenting Author:<br />

Julie L. Ryan, University <strong>of</strong> Rochester Medical Center, Rochester, NY<br />

Background: Radiation dermatitis occurs in approximately 95% <strong>of</strong> patients<br />

receiving radiation therapy for cancer and <strong>of</strong>ten leads to pain and treatment<br />

delays. There is no standard treatment with demonstrated effectiveness for<br />

the prevention <strong>of</strong> radiation dermatitis. We conducted a randomized,<br />

double-blind, placebo-control clinical trial to assess the efficacy <strong>of</strong> curcumin,<br />

a potent antioxidant and anti-inflammatory component <strong>of</strong> turmeric,<br />

to reduce radiation dermatitis in 30 breast cancer patients. Methods:<br />

Eligible patients included adult females with non-inflammatory breast<br />

cancer or carcinoma in situ prescribed radiation therapy without concurrent<br />

chemotherapy. After randomization, patients took four 500 mg capsules <strong>of</strong><br />

curcumin or placebo three times daily throughout their course <strong>of</strong> radiation<br />

therapy (total daily dose � 6.0 g). Weekly assessments included Radiation<br />

Dermatitis Severity (RDS) Score, presence/absence <strong>of</strong> moist desquamation,<br />

erythema measure, and McGill Pain and Symptom Inventory (SI)<br />

questionnaires. Results: The 30 evaluable patients were white (90%; mean<br />

age � 58.1 years) with ER�PR� breast cancer (76.7%) who did not have<br />

total mastectomy (90%) or chemotherapy prior to start <strong>of</strong> radiation therapy<br />

(56.7%). No significant differences were observed between arms for<br />

demographics, compliance, erythema, pain, symptoms, or radiation skin<br />

dose. Standard pooled variances t-test showed that curcumin reduced RDS<br />

at end <strong>of</strong> treatment compared to placebo (mean RDS � 2.6 vs 3.4;<br />

p�0.008). Fisher’s exact test showed that curcumin significantly reduced<br />

the presence <strong>of</strong> moist desquamation at the end <strong>of</strong> radiation therapy (28.6%<br />

vs. 87.5 %; p�0.002). Repeated measures analysis confirmed divergence<br />

<strong>of</strong> RDS between curcumin and placebo arms at Week 5. Conclusions: Oral<br />

curcumin, 6.0 g daily during radiation therapy, reduced radiation dermatitis<br />

severity and moist desquamation in breast cancer patients. A multisite<br />

CCOP trial (N�700) is underway to confirm the effectiveness <strong>of</strong> curcumin<br />

to reduce radiation dermatitis severity during various radiation therapy<br />

regimens for breast cancer. Support: IND 75,444, Dermatology Foundation,<br />

KL2 RR024136, NCI PHS 1R25CA10618.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


574s Patient and Survivor Care<br />

9028 Poster Discussion Session (Board #12), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

YOCAS yoga for musculoskeletal symptoms in breast cancer patients<br />

receiving aromatase inhibitors: A URCC CCOP randomized, controlled<br />

clinical trial. Presenting Author: Luke Joseph Peppone, University <strong>of</strong><br />

Rochester Medical Center, Rochester, NY<br />

Background: Up to 50% <strong>of</strong> breast cancer patients on aromatase inhibitor<br />

therapy report significant musculoskeletal symptoms such as joint and<br />

muscle pain, which decreases treatment adherence. We conducted a<br />

secondary data analysis <strong>of</strong> a multi-site, phase III randomized, controlled,<br />

clinical trial examining the efficacy <strong>of</strong> yoga for improving musculoskeletal<br />

symptoms among breast cancer patients currently receiving hormone<br />

therapy (aromatase inhibitors [AI] or tamoxifen [TAM]) through the University<br />

<strong>of</strong> Rochester Cancer Center Community <strong>Clinical</strong> Oncology Program<br />

(CCOP). Methods: The original RCT randomized patients with any type <strong>of</strong><br />

non-metastatic cancer without previous yoga participation into 2 arms: 1)<br />

standard care monitoring [controls] or 2) 4-week yoga intervention (2x/wk;<br />

75 min/session) plus standard care. The yoga intervention utilized the UR<br />

Yoga for Cancer Survivors (YOCAS) program consisting <strong>of</strong> breathing<br />

exercises, 18 Hatha and Restorative yoga postures, and meditation. Only<br />

breast cancer patients currently receiving AI (N�95) or TAM (N�72) were<br />

included in this secondary analysis. Changes in musculoskeletal symptoms<br />

were assessed using ANCOVA with baseline values as covariates between<br />

the yoga and control groups. Results: Compared to TAM users at baseline,<br />

AI users reported higher levels <strong>of</strong> general pain (1-5 score: AI�2.65 vs.<br />

TAM�2.17; p�0.01), muscle aches (0-4 score: AI�2.14 vs. TAM�1.65;<br />

p�0.01), and total physical discomfort (0-24: AI�8.03 vs. TAM�5.92;<br />

p�0.01). Among AI users only, participants in the yoga group demonstrated<br />

greater reductions in general pain (CS�change score; Yoga CS�<br />

-0.37 vs. Control CS� �0.02; p�0.02), muscle aches (Yoga CS� -0.58<br />

vs. Control CS� -0.15; p�0.03) and total physical discomfort (Yoga CS�<br />

-2.07 vs. Control CS� -0.58; p�0.04) from pre- to post-intervention than<br />

the control group. Conclusions: The severity <strong>of</strong> musculoskeletal symptoms<br />

was higher for AI users than for TAM users. Among breast cancer patients<br />

on AI therapy, the community-based YOCAS intervention significantly<br />

reduced general pain, muscle aches, and physical discomfort. Funding:<br />

NCI U10CA37420, KL2RR024136-05, K07CA120025.<br />

9030 Poster Discussion Session (Board #14), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Association <strong>of</strong> screening for psychosocial distress in patients with newly<br />

diagnosed stage IV NSCLC and survival. Presenting Author: William F. Pirl,<br />

Massachusetts General Hospital, Boston, MA<br />

Background: While screening for psychosocial distress has become an<br />

ASCO QOPI indicator <strong>of</strong> quality cancer care, data has yet to show an<br />

association with improved cancer outcomes. We examined the relationship<br />

between adherence to the QOPI measure for assessment <strong>of</strong> emotional<br />

well-being and survival in a sample <strong>of</strong> patients with newly diagnosed stage<br />

IV NSCLC. Methods: From 8/07 to 9/10, we recruited consecutive new<br />

patients in a multidisciplinary thoracic oncology clinic to participate in a<br />

research database for which patients completed self-report instruments for<br />

depression (PHQ-9) and anxiety (GAD-7) at their first oncology visit. We<br />

then performed a QOPI chart audit for patients with stage IV NSCLC who<br />

received care at our institution. To adhere to the QOPI measure, oncologists<br />

were required to document their own assessment <strong>of</strong> patient emotional<br />

well-being (without access to patient self-report data) within one month <strong>of</strong><br />

the first <strong>of</strong>fice visit. Survival was calculated from first oncology visit to<br />

death. Cox proportional hazards models were used to test associations with<br />

survival. Results: 243 patients completed baseline assessments and had<br />

follow up. Emotional well-being was assessed in 90 (37%). Median survival<br />

<strong>of</strong> those with an emotional assessment was 12 months vs. 10 months for<br />

those without (p�.03). Yet, emotional assessment was associated with<br />

younger age (p�.01) and higher depression (p�.05) and anxiety scores<br />

(p�.03). Controlling for age, sex, performance status, smoking, primary<br />

oncology provider, PHQ-9, and GAD-7, having an assessment <strong>of</strong> emotional<br />

well-being was associated with improved survival (HR .68, 95% CI<br />

.49-.93, p�.02). Although higher PHQ-9 scores were independently<br />

associated with worse survival (HR 1.05, 95% CI 1.02-1.08, p�.004),<br />

neither depression nor anxiety moderated the relationship between clinician<br />

screening and survival. Conclusions: In this large prospective study <strong>of</strong><br />

patients with newly diagnosed stage IV NSCLC, adherence to the QOPI<br />

measure for assessment <strong>of</strong> emotional well-being was associated with a<br />

survival benefit. This benefit appears to extend to all patients, not just those<br />

with depression and anxiety. Randomized trials are needed to confirm these<br />

findings.<br />

9029 Poster Discussion Session (Board #13), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

A randomized controlled trial <strong>of</strong> expressive writing for patients with renal<br />

cell carcinoma (RCC). Presenting Author: Timothy Burnight, University <strong>of</strong><br />

Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: Most previous research examining the efficacy <strong>of</strong> brief expressive<br />

writing interventions have used small sample sizes and followed people<br />

for no more than 3 months. We conducted a large randomized trial to<br />

examine an expressive writing intervention for patients with renal cell<br />

carcinoma and followed them for 10 months after the end <strong>of</strong> the writing<br />

sessions. Methods: Two hundred patients with RCC were randomly assigned<br />

to either write their deepest thoughts and feelings about their cancer (EW)<br />

or to write about neutral topics (NW) on four separate occasions over 10<br />

days for a maximum <strong>of</strong> 20 minutes at each writing session. Patients<br />

completed the MD Anderson Symptom Inventory (MDASI), Brief Fatigue<br />

Inventory, SF-36, IES, CES-D, and PSQI at baseline and then again 1, 4,<br />

and 10 months after the writing sessions. Results: The mean age <strong>of</strong> the<br />

participants was 58 (range 34-82 years), 41% were women, and staging<br />

data were 39% stage I, 15% stage II, 19% stage III, and 27% stage IV. The<br />

groups were well balanced on all demographic and medical characteristics.<br />

Examination <strong>of</strong> group differences 1 month after the writing sessions,<br />

controlling for the respective baseline measure, revealed decreased IES<br />

scores for the EW group (intrusive thoughts: EW, 5.0 v NW, 7.2; p�.02;<br />

avoidance behaviors: EW, 6.3 v NW, 8.7; p�.07). By 4 months after the<br />

intervention, the EW group reported higher levels <strong>of</strong> SF-36 Social Functioning<br />

scores (EW: 52.6 v NW: 49.7; p�.04). At the 10 month time point, the<br />

EW group reported fewer cancer-related symptoms (EW: 20.8 v NW: 30.8;<br />

p�.04), higher levels <strong>of</strong> SF-36 Role Physical scores (EW: 69.6 v NW: 54.0;<br />

p�.02), and fewer sleep disturbances (subscale <strong>of</strong> the PSQI; EW: 1.4 v<br />

NW: 1.6; p�.05). Means for the other SF-36 subscales at 10 months were<br />

in the expected, but did not reach statistical significance. There were no<br />

group differences for CES-D or fatigue scores at any time point. Mediation<br />

analyses revealed that IES scores at 1 month mediated the effects <strong>of</strong> EW on<br />

cancer-related symptoms (F� 1.85, p�.06) at the 10 month follow up.<br />

Conclusions: These findings indicate expressive writing leads to short-term<br />

reduction in intrusive thoughts about the cancer experience and results in<br />

long-term improvement in aspects <strong>of</strong> quality <strong>of</strong> life.<br />

9031 Poster Discussion Session (Board #15), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

The effect <strong>of</strong> pretreatment psychosocial factors on immune function and<br />

quality <strong>of</strong> life after brief presurgical stress management. Presenting Author:<br />

Chelsea D. Gilts, University <strong>of</strong> Houston, Houston, TX<br />

Background: It is important to identify factors that predict who will benefit<br />

the most from psychosocial interventions in cancer populations. Methods:<br />

We examined the association between pretreatment psychosocial factors<br />

and subjective and objective outcomes from a brief cognitive-behavioral<br />

stress management program. This three-arm randomized clinical trial<br />

investigated the effects <strong>of</strong> a presurgical stress management (SM; n�53)<br />

compared to supportive attention (SA; n�54) and standard care (SC;<br />

n�52) on quality <strong>of</strong> life (QOL) and immune outcomes in men with prostate<br />

cancer scheduled for radical prostatectomy. We previously reported improved<br />

QOL 1 year after surgery in the SM and SA groups compared to the<br />

SC group and higher pro-inflammatory cytokines 48h after surgery in the<br />

SM group. We investigated the moderating effects <strong>of</strong> baseline psychosocial<br />

factors (social support (SS), dyadic adjustment (DA), distress (BSIGSI),<br />

and impact <strong>of</strong> event (IES)), on QOL (SF-36 physical (PCS) and mental<br />

(MCS) component scores) 1 year after surgery and immune response 48h<br />

after surgery. Results: Baseline SS and BSIGSI interacted with group in<br />

predicting PCS 1 year after surgery (p’s �.05). Men who reported low<br />

baseline SS and were in SM had increased PCS 1 year after surgery<br />

compared to men with low SS in the SC group (b�.36, p�.01). Conversely,<br />

men who reported high BSIGSI at baseline and were in SA had increased<br />

PCS 1 year after surgery compared to those in SM (b�11.14, p�.03), with<br />

SC having an intermediate nonsignificant effect. All four psychosocial<br />

factors (SS, DA, BSIGSI, IES) interacted with group in predicting changes<br />

in pro-inflammatory cytokines (interleukin (IL)-1, IL-6, IL-8, IL-10, tumor<br />

necrosis factor, and interferon) 48h post-surgery (all p’s �.04). In all<br />

significant interactions, men who reported greater psychosocial vulnerability<br />

at baseline and were in SM had increased immune response 48h after<br />

surgery compared to psychosocially vulnerable men in either SC (all p’s<br />

�.02) or SA (all p’s �.02). Conclusions: These findings suggest that<br />

baseline psychosocial factors are important indicators <strong>of</strong> who may benefit<br />

most from a SM intervention on both subjective and objective outcomes.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


9032 Poster Discussion Session (Board #16), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Social environment as a predictor <strong>of</strong> smoking cessation and recidivism in<br />

lung cancer survivors. Presenting Author: Lawson Eng, Princess Margaret<br />

Hospital, University <strong>of</strong> Toronto, Toronto, ON, Canada<br />

Background: Smoking during cancer treatment negatively impacts treatment,<br />

survival and quality <strong>of</strong> life. Lung cancer patients with a smoking<br />

history <strong>of</strong>ten continue to smoke; some ex-smokers re-start after diagnosis.<br />

Social environment impacts cessation and recidivism rates in non-cancer<br />

patients. We assessed whether the same influences occur among lung<br />

cancer patients. Methods: Lung cancer patients, recruited from Princess<br />

Margaret Hospital, completed a baseline questionnaire about their demographics<br />

and smoking history (at diagnosis). A follow-up questionnaire was<br />

administered at a median <strong>of</strong> two years, assessing changes in smoking<br />

habits, exposure at home/work/among friends, healthcare use, social<br />

support and alcohol use since diagnosis. The relationship between each<br />

variable with cessation/recidivism was analyzed. Odds ratios (OR) and 95%<br />

confidence intervals (95% CI) were calculated. Results: 478 patients<br />

completed both questionnaires. Of the 100 current smokers at diagnosis;<br />

52 quit by the time <strong>of</strong> the follow-up questionnaire. Among 294 ex-smokers,<br />

15 started to smoke after diagnosis. None <strong>of</strong> the 84 never smokers at<br />

baseline started to smoke after diagnosis. Exposure to smoking at home was<br />

associated with continued smoking and relapse (OR�5.1, 95% CI:<br />

1.8–14.3, p�0.001; and OR�3.9, 95% CI: 0.8–14.4, p�0.04, respectively).<br />

Specifically, spousal smoking was associated with both continued<br />

smoking (OR�7.3, 95% CI: 2.4–21.7, p�2.0E-04) and recidivism<br />

(OR�3.7, 95% CI: 0.6–16.6, p�0.08). Having more than a few friends<br />

who smoke is associated with continued smoking (OR�3.5, 95% CI:<br />

1.4–8.7, p�0.005) and relapse (OR�4.8, 95% CI: 1.5–15.0, p�0.004).<br />

Not completing high school was also associated with continued smoking<br />

(OR�3.0, 95% CI: 1.2–7.6, p�0.02). Multivariate analysis identified<br />

spousal smoking as the major single predictor <strong>of</strong> continued smoking<br />

(OR�8.8, 95% CI: 2.2–34.8, p�0.002). Conclusions: Smoking cessation<br />

programs for lung cancer patients should not only target the patient but<br />

also include the immediate family, consider a patient’s peers and be<br />

tailored to the patient’s education level. Involvement <strong>of</strong> the immediate<br />

family and consideration <strong>of</strong> peers may help prevent smoking relapse.<br />

9034 Poster Discussion Session (Board #19), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Factors associated with life expectancy (LE) < 3 months (mo) among older<br />

adults receiving palliative chemotherapy (chemo). Presenting Author: Arti<br />

Hurria, City <strong>of</strong> Hope, Duarte, CA<br />

Background: Optimal treatment decision-making for older adults with<br />

advanced cancer requires a better understanding <strong>of</strong> risk factors associated<br />

with limited LE. Our objective was to evaluate the factors associated with<br />

LE� 3mo among patients (pts) age � 65 who were beginning a new chemo<br />

regimen. Methods: We conducted a secondary analysis <strong>of</strong> a multi-site cohort<br />

study <strong>of</strong> pts �65 years receiving chemo (Hurria et al, JCO 2011). This<br />

analysis included only pts receiving palliative chemo. Bivariate analysis<br />

and multivariate logistic regression were utilized to identify factors associated<br />

with LE � 3 mo including: sociodemographics, labs [hemoglobin<br />

(Hb), albumin, liver function, creatinine clearance], and geriatric assessment<br />

(GA) variables (functional status, social support, comorbidity, psychological,<br />

cognitive, and nutritional status). Results: Among 290 pts (median<br />

age 72 [range 65-91], 52% female) with advanced cancer (gastrointestinal<br />

28%, lung 31%, breast/gyn 22%, other 19%), 13.4% died within 3 mo <strong>of</strong><br />

chemo initiation. In bivariate analysis, pts with LE � 3mo were more likely<br />

(p�0.05 for each variable) to have lower albumin and Hb, unintentional<br />

weight loss, and poorer physical function [defined as need for assistance<br />

with instrumental activities <strong>of</strong> daily living (IADL), lower score on Medical<br />

Outcomes Survey (MOS) Physical Health, MD-rated and patient-rated<br />

Karn<strong>of</strong>sky performance status (KPS), and MOS Social Activity score].<br />

Measures <strong>of</strong> functional status were highly correlated with one another and<br />

therefore one functional status measure (in addition to measures significant<br />

in bivariate analysis) was included in each multivariate logistic<br />

regression. In multivariate analyses, (controlling for age, comorbidity, and<br />

line <strong>of</strong> chemo) poorer physical function (as evaluated by need for assistance<br />

with IADLs, MOS Physical �70, or MD-rated KPS �70) and unintentional<br />

weight loss were independently associated with LE�3mo (p�0.05).<br />

Conclusions: Among older pts with advanced cancer who were prescribed a<br />

new chemo regimen for palliative intent, physical function measures (as<br />

evaluated by geriatric assessment and MD report) and unintentional weight<br />

loss were associated with LE � 3months.<br />

Patient and Survivor Care<br />

575s<br />

9033 Poster Discussion Session (Board #18), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Strength and endurance training in the treatment <strong>of</strong> lung cancer patients<br />

staged IIIA/IIIB/IV. Presenting Author: Corinna C. Henke, Department <strong>of</strong><br />

Oncology and Hematology, Vivantes Klinikum Neukölln, Berlin, Germany<br />

Background: This RCT tested the effect <strong>of</strong> a specially designed strength and<br />

endurance training on the independence in activities <strong>of</strong> daily living and<br />

quality <strong>of</strong> life in lung cancer patients staged IIIA/IIIB/IV while receiving<br />

palliative chemotherapy. The aim was to break the vicious circle created<br />

through the connection <strong>of</strong> physical inactivity and the worsening <strong>of</strong> symptoms<br />

and side effects. Methods: Between August 2010 and December<br />

2011 lung cancer patients staged IIIA/IIIB/IV with a good performance<br />

status receiving an inpatient palliative chemotherapy treatment at the<br />

Vivantes Hospital Neukölln/Berlin, were randomized into an intervention<br />

and control group. The Barthel Index and the EORTC QLQ-C30/ LC13<br />

questionnaire were used for evaluation. The Six-Minute-Walk-Test and stair<br />

walking in combination with the Modified Borg Scale have been used to test<br />

the patient´s endurance capacity. Furthermore muscle strength was examined.<br />

Non-parametrical data were statistically analyzed with the Wilcoxon<br />

and Mann-Whitney-U test. For parametric data student t- tests were used. A<br />

significance level <strong>of</strong> p� .05 was accepted. Results: Out <strong>of</strong> 46 patients, who<br />

signed the informed consent, 29 patients completed the trial (18�<br />

Intervention group, 11� Control group). Significant differences between<br />

the groups were detectable in the Barthel Index (IGmean(SD)�92.08<br />

(15.15); CGmean�81.67 (14.98); p�.041), and in single scores <strong>of</strong> the<br />

EORTC QLQ C-30/LC-13 questionnaire (Physical Functioning p�.025;<br />

Haemoptysis p�.019, Pain in Arms or Shoulder p�.048, Peripheral<br />

Neuropathy p�.050, Cognitive functioning p�.050). Significant differences<br />

were found between the groups concerning the 6MWT, stair walking<br />

and strength capacity (IG�CG). Additionally the level <strong>of</strong> dyspnoea decreased<br />

significantly in the IG while performing submaximal walking<br />

activities. Conclusions: The training program has a positive impact on the<br />

patient´s independence in carrying out activities <strong>of</strong> daily living. Although it<br />

does not have a significant impact on the patient´s quality <strong>of</strong> life, single<br />

factors can be significantly improved. Moreover it has a positive effect on<br />

the patient´s endurance and strength capacity. The dyspnoea perception is<br />

improved.<br />

9035 Poster Discussion Session (Board #20), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

The influence <strong>of</strong> age plus standard clinical approach with or without<br />

comprehensive geriatric assessment (CGA) on treatment decisions in older<br />

cancer patients: Final results. Presenting Author: Lore Decoster, Department<br />

<strong>of</strong> Medical Oncology, Oncologisch Centrum, Universitair Ziekenhuis<br />

Brussel, Brussel, Belgium<br />

Background: The aim <strong>of</strong> this prospective study was to examine how age and<br />

standard clinical approach with and without CGA results determine<br />

treatment decisions in older cancer patients (pts). Methods: This study,conducted<br />

in 2 Belgian university hospitals, included pts � 70 years with a<br />

malignant tumor (breast, colorectal, ovarian, lung, prostate and hematological)<br />

if a new cancer therapy was considered. All pts underwent a uniform<br />

CGA. Results were communicated to the treating physician. After the<br />

treatment decision, an interview with the treating physician was performed,<br />

using a predefined questionnaire: 1/ What would be your oncological<br />

treatment proposal in case the pt was 55y without other comorbidity? 2/ Is<br />

this different from your treatment proposal for this pt according to age and<br />

standard clinical approach without CGA results? 3/ Is this different from<br />

your treatment proposal for this pt with CGA results? Results: From October<br />

2009 till July 2011, 937 pts were included in the study. Median age was<br />

76y (range 70-95) and 63.5% was female. A total <strong>of</strong> 902 (96.3%)<br />

questionnaires were completed and 56.2% <strong>of</strong> the physicians were aware <strong>of</strong><br />

the CGA results at treatment decision. In 381 pts (42.2%; 95%CI<br />

39.0-45.5) age and standard clinical approach led to a different treatment<br />

decision compared to younger pts without comorbidity. This influence was<br />

most prominent for chemotherapy decisions: 309 patients did not receive<br />

standard chemotherapy (reduced dose (13), less toxic regimen (163), less<br />

toxic regimen at reduced dose (5) or no chemotherapy (128)). When the<br />

physician was aware <strong>of</strong> the CGA, these results influenced their treatment in<br />

6.7% (95%CI: 4.5-8.9), mostly concerning chemotherapy. In 8 pts CGA<br />

results encouraged the treating physician to choose a more aggressive<br />

chemotherapy regimen and in 11 pts CGA results led to a decision <strong>of</strong><br />

palliative care. Conclusions: Based on this prospective trial, we conclude<br />

that physicians use adapted treatment regimens in older versus younger<br />

pts, only based on age and standard clinical approach. CGA results change<br />

the treatment decision in 6.7% and sometimes trigger the use <strong>of</strong> a more<br />

aggressive treatment.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


576s Patient and Survivor Care<br />

9036 Poster Discussion Session (Board #21), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Patient information desire in actual decision making for advanced cancer<br />

treatment: Do doctors know their patients? Presenting Author: Linda J. M.<br />

Oostendorp, Radboud University Nijmegen Medical Center, Nijmegen,<br />

Netherlands<br />

Background: Providing information is an important element <strong>of</strong> cancer care.<br />

While several studies have investigated patients’ information desire, to our<br />

knowledge, this study is the first to involve patients facing an actual<br />

decision. We examined the information desire <strong>of</strong> patients at the point <strong>of</strong><br />

decision making, the ability <strong>of</strong> doctors to judge this desire, and the<br />

information provided by the doctor. Methods: This prospective multicenter<br />

study included patients with advanced colorectal or breast cancer faced<br />

with the decision whether or not to pursue second-line chemotherapy.<br />

Patients received the usual treatment-related information from the doctor<br />

plus a decision aid from a nurse. The aid contained information on adverse<br />

events, tumor response, and survival. For each item, the nurse asked the<br />

patient whether the information was desired and whether it had been<br />

disclosed by the doctor and the doctor made a substitute judgment <strong>of</strong> the<br />

patient’s information desire on the inclusion form. The match between<br />

patient’s desire and doctor’s judgment was expressed in percentage<br />

agreement and agreement corrected for chance (�). Results: By 01/2012,<br />

71 patients had received the decision aid. Median age was 62 years (range<br />

39-80), 38% were male, and 28% had college education or higher.<br />

Information on adverse events, tumor response, and survival was desired by<br />

94%, 90%, and 73% <strong>of</strong> patients. The doctors judged that information<br />

desire would be 100%, 97%, and 81%, respectively. There was a poor<br />

match between doctor’s judgment and actual information desire for<br />

adverse events (94%, � not applicable), tumor response (87%, � �<br />

-0.049), and survival (61%, � � -0.104). When asked whether the<br />

information was previously disclosed by the doctor, 73%, 57%, and 30% <strong>of</strong><br />

patients answered affirmatively. Conclusions: Patients expressed a high<br />

information desire, which was accurately judged by the doctors. However,<br />

doctors were unable to judge an individual patient’s information desire<br />

beyond chance. According to the patients, doctors did not disclose all<br />

desired information, especially on survival. Decision aids, similar to those<br />

used here, have been shown to help doctors provide safe, effective, and<br />

timely information to patients.<br />

9038 General Poster Session (Board #38A), Sat, 8:00 AM-12:00 PM<br />

Difficult conversations with terminal patients: Palliative care and death at<br />

home. Presenting Author: Eduardo L. Morgenfeld, Instituto Oncológico<br />

Henry Moore, Buenos Aires, Argentina<br />

Background: More than half the total number <strong>of</strong> oncological patients may<br />

die due to the disease’s progression, and at some point in time the<br />

oncologist must communicate that the disease is terminal. This is the<br />

hardest conversation (HC) <strong>of</strong> a clinical oncologist. The following paper’s<br />

objective is to determine whether or not the treatments applied to patients,<br />

and their evolution, are affected by having had the HC. Methods: All<br />

patients who died due to progression <strong>of</strong> the disease in 2011 were selected<br />

for this study. Both medical histories and applied treatments were<br />

evaluated. All doctors were interviewed to establish when the HC had taken<br />

place. The distribution <strong>of</strong> variables was analyzed and both populations<br />

compared. Results: Between January 2011 and December 2011, 110<br />

patients who met the previous criteria were studied. Population characteristics:<br />

Sex (Female: 52 pts; Male: 58 pts). Median age at the time <strong>of</strong> death:<br />

61 years (Range: 27-88). Patients were divided in two groups: A (terminal<br />

status <strong>of</strong> the disease had been discussed; 70 pts) and B (terminal status <strong>of</strong><br />

the disease had not been discussed; 40 pts). Both groups were similar in<br />

diagnosis, age and previous treatments. During the post HC evolution, three<br />

elements were categorized according to the table below. Conclusions: 1–<br />

The conviction, ethical sense and strength <strong>of</strong> will <strong>of</strong> the doctor are<br />

determining factors when related to his or her capacity to talk with the<br />

patient about the terminal aspect <strong>of</strong> the disease. 2 – The chemotherapy<br />

treatments applied during the last two months <strong>of</strong> life are similar in both<br />

groups. 3 – The percentage <strong>of</strong> patients that died at their homes under<br />

palliative care was remarkably higher amongst those who had been<br />

informed than those who had not been informed.<br />

Group A<br />

N�70 (%)<br />

Group B<br />

N�40 (%) p value<br />

Condition<br />

Recieved chemotherapy in the<br />

two months prior to death<br />

24 (34%) 17 (42%) 0.391<br />

Received palliative care 55 (79% 15 (37%) 0.0001<br />

Died at home 44 (63%) 6 (15%) 0.0001<br />

9037 Poster Discussion Session (Board #22), Mon, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Women know, and men wish they knew, prognostic information in advanced<br />

cancer. Presenting Author: Kalen M. Fletcher, Dana-Farber Cancer Institute,<br />

Boston, MA<br />

Background: Little is known about gender differences in advanced cancer<br />

patient communication with oncologists. The few studies conducted have<br />

explored differences in preferences for prognostic disclosure. Our data<br />

allow us to test for gender differences in actual rates <strong>of</strong> audio-recorded,<br />

patient and oncologist reported, prognostic disclosures. We studied a group<br />

<strong>of</strong> advanced cancer patients to determine whether gender disparities exist<br />

in: a.) reported rates <strong>of</strong> prognostic disclosures from physicians and b.)<br />

willingness to estimate (versus not) one’s prognosis (i.e., amount <strong>of</strong> time<br />

left to live). Among patients who report never receiving a prognosis from<br />

their physician, we also tested for gender difference in wishing that this had<br />

been discussed. Methods: Coping with Cancer II is an NCI -funded<br />

multi-site, prospective longitudinal study <strong>of</strong> advanced cancer patients.<br />

Patients were interviewed after receiving scan results and asked if they<br />

have received a prognosis from their oncologist either at their most recent<br />

visit or at any time in the course <strong>of</strong> their disease. They are also asked if they<br />

would be willing to estimate their prognosis. Patients who state that they<br />

have not received a prognosis are asked if they wish that they had. Results:<br />

Among the advanced cancer patients studied (N�51; men�23,<br />

women�28), male cancer patients were significantly more likely to state<br />

never receiving a prognosis from their physician than female patients<br />

(OR�3.5; �2�4.49, df�1, p�0.034). Male cancer patients were also<br />

significantly less willing to provide a life-expectancy estimate (OR�5.6;<br />

�2�5.06, df�1, p�0.025). Among patients who stated never receiving a<br />

prognosis (N�27; men�16, women�11), male patients tended to be more<br />

likely than female patients to wish that their prognosis had been discussed<br />

(OR�7.8; �2�3.11, df�1, p�0.078). Conclusions: Male advanced cancer<br />

patients are less likely than female cancer patients to state that they have<br />

received prognostic information and less willing to provide a lifeexpectancy<br />

estimate. Although male patients receive less open prognostic<br />

disclosure than female patients, male patients tend to be more likely than<br />

female patients to want prognostic information.<br />

9039 General Poster Session (Board #38B), Sat, 8:00 AM-12:00 PM<br />

In-hospital cancer care: Are we missing an opportunity for end <strong>of</strong> life care?<br />

Presenting Author: Gabrielle Betty Rocque, University <strong>of</strong> Wisconsin Carbone<br />

Cancer Center, Madison, WI<br />

Background: Little data exists on the estimated survival <strong>of</strong> patients with<br />

metastatic cancer after hospitalization. As part <strong>of</strong> a prospective quality<br />

improvement project, we characterized the population <strong>of</strong> patients admitted<br />

to an inpatient oncology service in an academic medical center with<br />

emphasis on the disposition at discharge and overall survival. Methods: We<br />

collected data over a 4 month period (9/1/10-12/23/10) representing 149<br />

admissions <strong>of</strong> 119 unique patients. We measured patient characteristics,<br />

disease evaluation, procedures, consults, imaging studies performed,<br />

disposition, length <strong>of</strong> stay, and overall survival. These data were compared<br />

to a similar study conducted in our center in 2000. Results: Uncontrolled<br />

symptoms were the most common reason for admission (pain 28%,<br />

dyspnea 9%, nausea 9%). Imaging studies were more common than in<br />

2000 (415 vs. 196 total procedures). Eighty-five percent <strong>of</strong> patients had<br />

progressive disease. Seventy percent <strong>of</strong> patients were discharged home<br />

without additional services such as home health or hospice. The overall<br />

median survival was poor in 2000 and in 2010, 100 days and 60 days from<br />

discharge, respectively. Despite an overall poor prognosis, palliative care<br />

consultation was obtained only 13 times (8% <strong>of</strong> admissions) and 18% <strong>of</strong><br />

patients were enrolled in hospice at discharge. Conclusions: An unscheduled<br />

hospital admission portends a poor prognosis. Cancer patients in the<br />

hospital are nearing the end-<strong>of</strong>-life with a median life expectancy <strong>of</strong><br />

approximately 3 months and could be considered hospice-eligible and<br />

appropriate for a palliative care consult. We believe that hospital admission<br />

represents a missed opportunity to provide palliative care services and<br />

end-<strong>of</strong>-life counseling to this patient population.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


9040 General Poster Session (Board #38C), Sat, 8:00 AM-12:00 PM<br />

Palliative cancer therapy during end <strong>of</strong> life at a regional cancer center in<br />

Norway in 2005 and 2009. Presenting Author: Mari Aas Gynnild, Department<br />

<strong>of</strong> Cancer Research and Molecular Medicine, Norwegian University <strong>of</strong><br />

Science and Technology and The Cancer Clinic, St. Olavs Hospital,<br />

Trondheim University Hospital, Trondheim, Norway<br />

Background: With increasing number <strong>of</strong> available therapies, there is a risk<br />

that patients (pts) are overtreated. Palliative cancer therapy is mostly<br />

recommended for pts with good Performance Status (PS). In one study, 42<br />

% <strong>of</strong> pts received chemotherapy (CTx) during the last 30 days <strong>of</strong> life –<br />

suggesting that this may not always be the case. Methods: All pts who,<br />

according to the national registry, died from cancer in our region in 2005<br />

and 2009 were analyzed. Data were collected from individual medical<br />

records. Endpoints: Time from the end <strong>of</strong> palliative cancer therapy until<br />

death. Whether there were differences depending on age; type <strong>of</strong> cancer;<br />

year <strong>of</strong> death or if they were seen at a palliative care unit (PCU). PS when<br />

the last cancer therapy was initiated. Results: 616 pts died in 2005; 599 in<br />

2009. We excluded 495 pts: No cancer therapy (n�260); no information<br />

<strong>of</strong> cancer (n�101); last therapy with curative intention (n�83); hematological<br />

malignancy (n�51). Median age 71 (6 - 99); 49 % men; median overall<br />

survival from diagnosis: 16.9 mos. Last therapy was radiotherapy (RT): 31<br />

%; CTx: 40 %; hormonal: 15 %; surgery: 11 %. 4 % died from treatment<br />

complications. Median time from start <strong>of</strong> last CTx or RT until death: 100<br />

days; from end <strong>of</strong> last CTx or RT: 63 days. Younger pts received more CTx<br />

and RT in the last 30 days: Age � 60: 28 %; 60-70: 23 % and 70�:12%<br />

(p�.001). The table shows the use <strong>of</strong> CTx and RT the last 30 and 14 days<br />

for the most common cancers. Among those who got CTx in the last 30 days<br />

(n�74); 54 % had PS 2; 14 % PS 3-4. Among those who got RT in the last<br />

30 days (n�61), 31 % had PS 2; 54 % PS 3-4. Of the 49 % referred to the<br />

PCU, fewer received CTx or RT in the last 30 days (PCU: 14 %, no PCU: 22<br />

%; p�.002) and 14 days (PCU: 5 %, no PCU: 12 %; p�.001). Conclusions:<br />

Many pts received cancer therapy the last month <strong>of</strong> life. The percentage<br />

varies with age, cancer type and was lower in 2009 than in 2005. Pts seen<br />

at the PCU received less CTx and RT. Many pts had a poorer PS at the start<br />

<strong>of</strong> last cancer therapy than recommended.<br />

(n in 2005/2009)<br />

Lung<br />

(n�74/81)<br />

Colon<br />

(n�57/52)<br />

Breast<br />

(n�34/33)<br />

Prostate<br />

(n�45/46)<br />

All<br />

(n�352/369)<br />

CTx last 30 d 19%/20% 4%/10% 18%/6% 0%/0% 11%/10%<br />

CTx last 14 d 12%/4% 0%/4% 12%/3% 0%/0% 6%/3%<br />

RT last 30 d 22%/11% 11%/4% 9%/6% 0%/11% 10%/7%<br />

RT last 14 d 12%/6% 5%/0% 6%/3% 0%/7% 5%/3%<br />

9042 General Poster Session (Board #38E), Sat, 8:00 AM-12:00 PM<br />

Feasibility and acceptability <strong>of</strong> patient-reported outcomes data collection<br />

for clinical care following breast reconstruction. Presenting Author: Andrea<br />

Pusic, Memorial Sloan-Kettering Cancer Center, New York, NY<br />

Background: To date, systematic measurement <strong>of</strong> patient-reported outcomes<br />

(PROs) has played an important role in cancer research, but not in<br />

routine clinical care. Our objective was to evaluate the feasibility <strong>of</strong><br />

developing and piloting an electronic PRO data collection in clinical care<br />

among breast reconstruction patients using the BREAST-Q, a previously<br />

developed condition-specific PRO measure for breast surgery patients that<br />

measures quality <strong>of</strong> life (e.g. psychosocial, physical and sexual well-being)<br />

as well as patient satisfaction (e.g. satisfaction with breasts, with information,<br />

with surgeon). Methods: The BREAST-Q was loaded to the MSKCC<br />

WebCore, a generic electronic patient-reporting platform adhering to strict<br />

privacy and security standards. Patients attending visits at the MSKCC<br />

Breast Reconstruction Clinic were asked to complete the BREAST-Q<br />

electronically prior to scheduled visits. For patients with email addresses, a<br />

reminder with web-link to the questionnaire was emailed automatically<br />

prior to the visit. Results: Over a 9 month start-up period, BREAST-Q<br />

surveys were completed by 1442 patients. Patients completed the questionnaire<br />

at set time points before and after surgery. A total <strong>of</strong> 2340 BREAST-Q<br />

surveys were completed overall. Mean completion time was 5:53 minutes.<br />

Acceptability was high with both patients and clinical staff contributing<br />

positive comments along with suggestions for improvement via email.<br />

Conclusions: This pilot experience suggests that ePRO data can be<br />

efficiently collected among outpatient breast surgery patients with high<br />

acceptability. In the next phase <strong>of</strong> this project, we will introduce real-time<br />

individual patient reports to the clinical team and evaluate the impact <strong>of</strong><br />

this information on clinical care and quality improvement.<br />

Patient and Survivor Care<br />

577s<br />

9041 General Poster Session (Board #38D), Sat, 8:00 AM-12:00 PM<br />

Effect <strong>of</strong> chemotherapy and psychosocial distress parameters on perceived<br />

cognitive disturbances in Asian breast cancer patients. Presenting Author:<br />

Yin Ting Cheung, National University <strong>of</strong> Singapore, Singapore<br />

Background: A qualitative study has revealed that Asian breast cancer<br />

patients attributed their post-chemotherapy cognitive disturbances to<br />

psychosocial distress. To validate this claim, we aim to examine perceived<br />

cognitive disturbances, anxiety and quality <strong>of</strong> life (QoL) in Asian breast<br />

cancer patients and to identify clinical and psychosocial factors associated<br />

with perceived cognitive disturbances. Methods: A prospective, observational<br />

study was held at the largest cancer center in Singapore. Chemotherapy<br />

(CT) and non-chemotherapy (non-CT) receiving breast cancer<br />

patients completed self- reported questionnaires to assess the following<br />

domains: patients’ perceived impact <strong>of</strong> chemotherapy on cognitive disturbances<br />

(FACT-Cog), health-related QoL (EORTC QLQ-C30) and anxiety<br />

(Beck Anxiety Inventory). Multiple regression was conducted to delineate<br />

factors associated with perceived cognitive impairment. Results: A total <strong>of</strong><br />

166 (1:1 CT/non-CT) patients were recruited (age: 54.1�10.2 years;<br />

78.9% Chinese; 53.6% post-menopausal). Most <strong>of</strong> the CT patients<br />

received anthracycline-based chemotherapy (93.1%) and anti-hormonal<br />

therapy (69.4%). Comparing to non-CT patients, CT patients experienced<br />

more fatigue (QLQ-C30 fatigue scores: 22.2 vs 33.3 points; p�0.005),<br />

more significant anxiety (8.6% vs 21.9%; p�0.002), and more cognitive<br />

disturbances (FACT-Cog scores: 110 vs 124 points; p�0.0001). Regression<br />

model identified chemotherapy, anti-hormonal therapy, emotional<br />

functioning and global health status to be strongly associated with<br />

cognitive disturbances in Asian breast cancer patients. The interacting<br />

effect between anxiety and fatigue, comparing to fatigue alone, was more<br />

associated with cognitive disturbances (��-0.212; p�0.032 vs ��-0.07;<br />

p�0.25, respectively). Conclusions: This is the first study to demonstrate<br />

that Asian breast cancer patients experiencing both fatigue and anxiety are<br />

more predisposed to cognitive disturbances. Post-chemotherapy cognitive<br />

changes are observed in our patients, and our results suggest that<br />

psychosocial factors are impactful to identify cancer patients who are more<br />

susceptible to cognitive disturbances.<br />

9043 General Poster Session (Board #38F), Sat, 8:00 AM-12:00 PM<br />

Patient-reported satisfaction with reconstructed breasts in the long-term<br />

survivorship period: Comparison <strong>of</strong> autologous and nonautologous breast<br />

reconstruction. Presenting Author: Amie Scott, Memorial Sloan-Kettering<br />

Cancer Center, New York, NY<br />

Background: Breast cancer patients undergoing mastectomy may choose to<br />

have reconstruction performed using either their own tissue or an implant.<br />

As many patients are candidates for both, valid and reliable patientcentered<br />

outcomes data are crucial to shared medical decision-making.<br />

The objective <strong>of</strong> this study is to determine whether patient-reported<br />

satisfaction with their reconstructed breasts is dependent on type <strong>of</strong><br />

reconstructive surgery and length <strong>of</strong> time from reconstruction. Methods:<br />

<strong>Part</strong>icipants were recruited from Memorial Sloan-Kettering Cancer, NY and<br />

the University <strong>of</strong> British Columbia, Canada. Patients completed the<br />

BREAST-Q, a new patient-reported outcome measure for breast surgery<br />

patients. The dependent variable was the BREAST-Q Satisfaction with<br />

Breast score, a 16-item scale scored from 0-100. Procedure type, time<br />

since surgery, and patient characteristics were independent variables.<br />

Univariate analysis and clinical judgment were used to identify variables<br />

included in the model, and multivariate linear regression models were<br />

constructed to control for confounders. Results: The study sample consisted<br />

<strong>of</strong> 510 women (response rate 62%). The sample was on average aged 54.3<br />

� 9.3 (range 21-81), surveyed 3.71 years � 1.55 (range 1-9) after<br />

surgery, 66% were reconstructed using an implant. Type <strong>of</strong> surgery and<br />

laterality were found to be variables that predicted higher patient satisfaction<br />

with their breasts after controlling for radiation therapy, follow-up time,<br />

timing <strong>of</strong> surgery, age, body mass index, and major complications (surgery<br />

type p�0.001; laterality p�0.001, R-square�0.17). Conclusions: As there<br />

is a growing population <strong>of</strong> breast cancer survivors, understanding how a<br />

woman’s satisfaction with her reconstructed breasts changes over time is<br />

essential. This study suggests that patient satisfaction with breast reconstruction<br />

depends on the type <strong>of</strong> reconstruction a woman undergoes. This<br />

patient-centered outcome data can be used to enhance shared medical<br />

decision-making by providing patients with information about realistic<br />

expectations for satisfaction with breasts related to type <strong>of</strong> surgery chosen.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


578s Patient and Survivor Care<br />

9044 General Poster Session (Board #38G), Sat, 8:00 AM-12:00 PM<br />

Predictors <strong>of</strong> resilience in women treated for breast cancer: A prospective<br />

study. Presenting Author: Barbara Kaye Bennett, University <strong>of</strong> New South<br />

Wales, Sydney, Australia<br />

Background: Because a cancer diagnosis may be regarded as a potentially<br />

traumatic event with untoward consequences, much research has focussed<br />

on negative aspects <strong>of</strong> cancer diagnosis and treatment, emphasising<br />

psychological outcomes. A recent paradigm shift recognises that such a<br />

psycho-pathological approach discounts the human capacity for resilience<br />

�the ability to maintain relatively stable functioning following an aversive<br />

life event� (Bonanno GA Curr Direct in Psychol Science 2005 14(3) 135).<br />

Predictors <strong>of</strong> resilience in women who recovered uneventfully from surgical<br />

and adjuvant treatment for early stage breast cancer were investigated in a<br />

prospective study. Methods: Validated self-report measures <strong>of</strong> mood,<br />

somatic symptoms, temperament, illness attitudes, and social support<br />

were completed post-surgery. <strong>Clinical</strong>, tumor and treatment details were<br />

recorded. At end treatment and subsequently at 1, 3, 6, 9 and 12 months,<br />

self-report measures <strong>of</strong> mood and somatic symptoms were completed.<br />

Resilience was defined as: no evidence <strong>of</strong> protracted psychological or<br />

somatic distress in the 12 months following treatment completion. Identified<br />

resilient and non-resilient groups were compared and predictors sought<br />

by logistic regression. Results: Of 218 women evaluated, 106 (49%) were<br />

classified as �resilient.� They either reported no clinically significant<br />

psychological or other distress post-surgery (34%) and 12months following<br />

adjuvant treatment or recovered promptly (15%) and remained well. There<br />

were no significant differences in age (52 years); marital status; tumor size;<br />

treatment; treatment toxicity (nadir hemoglobin and neutrophil count) or<br />

mortality at 5 years post treatment (8 confirmed deaths in each group).<br />

Logistic regression identified low neuroticism (temperament or personality<br />

trait) as the most significant predictor <strong>of</strong> resilience. Conclusions: These<br />

findings suggest that without any intervention almost half the women<br />

treated for breast cancer adapt well to diagnosis and treatment. These<br />

outcomes warrant further research, to provide further insight into how<br />

individuals cope with major illness and to facilitate development <strong>of</strong><br />

programs to aid those in whom outcomes are protracted.<br />

9046 General Poster Session (Board #39A), Sat, 8:00 AM-12:00 PM<br />

Patient-related factors influencing time between first signs <strong>of</strong> breast cancer<br />

(BC) and appointment for medical visit (AMV): An international survey.<br />

Presenting Author: Jacek Jassem, Klinika Onkologii i Radioterapii Gdanski<br />

Uniwersytet Medyczny, Gdansk, Poland<br />

Background: Reducing diagnostic delays may improve treatment outcomes<br />

in BC. We investigated in various countries patient-related factors influencing<br />

time to seeking medical advice for signs <strong>of</strong> BC. Methods: A total <strong>of</strong><br />

4,816 female BC patients from 10 countries were surveyed using a uniform<br />

questionnaire translated into local languages. Time between first patientdetected<br />

signs <strong>of</strong> BC and AMV was measured using categorized time scales.<br />

Out <strong>of</strong> 14 original items on a multiple scale measuring BC-related attitudes<br />

and behaviors, 5 factors were extracted and used for further analysis:<br />

distrust in medical system and success <strong>of</strong> therapy, disregard <strong>of</strong> signs, fear<br />

<strong>of</strong> BC, practicing regular breast self-examination and support from friends<br />

and family. Results: In the subset <strong>of</strong> 2,870 patients with self-detected BC<br />

who provided complete answers to relevant variables, the mean time to<br />

AMV varied in particular countries from 3.4 to 6.2 weeks (grand mean <strong>of</strong><br />

4.7 weeks), with 39% <strong>of</strong> cases with a delay <strong>of</strong> �4 weeks. Overall, patient<br />

attributes that significantly influenced time to AMV were: distrust (p�1E-<br />

36), disregard (p�1.26E-30), fear (p�2.65E-16), self-examination<br />

(p�1.31E-21), place <strong>of</strong> living (p�3.5E-3) and education (p�2.73E-3).<br />

Multilevel analysis indicated that significant differences among particular<br />

countries were only due to slopes <strong>of</strong> distrust and disregard included in<br />

general regression model. The model enhanced with the two abovementioned<br />

random effects provided significant improvement in predicting time<br />

to AMV. Re-estimation <strong>of</strong> the model based solely on data from individual<br />

countries produced 10 significant equations with varied coefficients for<br />

distrust and disregard (see table). Conclusions: Patient-related factors<br />

contribute considerably to delay in the diagnosis <strong>of</strong> BC. Differences<br />

between particular countries call for country-specific approaches.<br />

Country Distrust Country Disregard<br />

Romania 2.3 Turkey 1.5<br />

Serbia 1.7 Serbia 1.5<br />

Slovakia 1.5 Latvia 1.4<br />

Hungary 1.4 Slovakia 1.1<br />

Latvia 1.2 Poland 1.0<br />

Poland 1.1 Russian Fed. 1.0<br />

Russian Fed. 0.9 Bulgaria 1.0<br />

Lithuania 0.9 Lithuania 0.6<br />

Bulgaria 0.7 Hungary 0.4<br />

Turkey 0.03 Romania -0.9<br />

9045 General Poster Session (Board #38H), Sat, 8:00 AM-12:00 PM<br />

Predictors <strong>of</strong> satisfaction with decision in patients with advanced cancer.<br />

Presenting Author: Felicitas Hitz, Department <strong>of</strong> Medical Oncology, Kantonsspital<br />

St.Gallen, St. Gallen, Switzerland<br />

Background: Treatment decisions in palliative cancer care depend on<br />

various patient-, treatment, and disease-related factors. This study investigated<br />

patients’ satisfaction with decision and decision-making preferences<br />

in a Swiss oncology network and evaluated factors that predict satisfaction<br />

with decision (SWD). Methods: Patients receiving a new line <strong>of</strong> palliative<br />

cancer treatment completed a multi-dimensional questionnaire 4-6 weeks<br />

after treatment decision. Patient-reports comprised socio-demographic<br />

information, satisfaction with decision (primary endpoint), subjective<br />

treatment goal, preferences for decision-making, health, locus <strong>of</strong> control,<br />

and several quality <strong>of</strong> life (QoL) domains. Information on diagnosis, line <strong>of</strong><br />

therapy, metastases, prognosis, performance status and duration <strong>of</strong> consultation<br />

were provided by the attending physician. Predictors <strong>of</strong> SWD (range:<br />

0�worst; 30�best) were evaluated by uni- and multivariate (primary<br />

analysis, 80% power, 5% significance level) regression models. We<br />

consider �24 points as high SWD. Results: 445 out <strong>of</strong> 480 patients treated<br />

in 8 hospitals and 2 private practices completed all questions regarding the<br />

primary endpoint. 44% <strong>of</strong> patients preferred shared-, 42% physiciancentered<br />

decision making. Median duration <strong>of</strong> consultation was 30 minutes<br />

(range 10-200 minutes). Overall, 326 (73%) patients reported high SWD.<br />

In univariate analyses various factors (e.g. global and physical QoL,<br />

performance status, treatment goal, locus <strong>of</strong> control, prognosis and<br />

duration <strong>of</strong> consultation) were significant predictors <strong>of</strong> SWD. The only<br />

significant predictor in the multivariate analysis was duration <strong>of</strong> consultation,<br />

which was positively associated with SWD (p�0.01). Conclusions:<br />

SWD is generally high in patients with advanced cancer treated in an<br />

oncology network in Switzerland. Our results indicate that duration <strong>of</strong><br />

consultation is a very important factor among many others in predicting<br />

SWD. In busy clinical practice, taking your time with patients may be a<br />

simple measure to enhance patients’ SWD.<br />

9047 General Poster Session (Board #39B), Sat, 8:00 AM-12:00 PM<br />

Validity and reliability <strong>of</strong> the patient-reported outcomes version <strong>of</strong> the<br />

common terminology criteria for adverse events (PRO-CTCAE). Presenting<br />

Author: Amylou C. Dueck, Mayo Clinic, Scottsdale, AZ<br />

Background: Symptomatic adverse events (AE) in cancer trials are reported<br />

by clinicians using the National Cancer Institute’s (NCI) Common Terminology<br />

Criteria for Adverse Events (CTCAE). To integrate the patient perspective<br />

into AE reporting, NCI contracted (HHSN261201000043C) to create<br />

a patient-reported outcomes companion tool (PRO-CTCAE). We report the<br />

validity and reliability <strong>of</strong> PRO-CTCAE’s 124 items reflecting 78 symptomatic<br />

AEs. Methods: English-speaking subjects (n�869; 44% male; median<br />

[mdn] age 59; 32% racial/ethnic minority; 34% high school or less; 17%<br />

ECOG Performance Status [PS] 2-4) receiving treatment for a range <strong>of</strong><br />

cancers at 4 NCI-designated cancer centers and 5 sites in NCI’s Community<br />

Cancer Centers Program completed a web-based survey in clinic<br />

including PRO-CTCAE and EORTC QLQ-C30. Pearson correlations were<br />

computed between PRO-CTCAE items and QLQ-C30 scales. Differences in<br />

PRO-CTCAE item scores between clinician-reported ECOG PS (0-1 vs 2-4)<br />

groups were computed. Test-retest reliability <strong>of</strong> 48 prespecified items was<br />

evaluated in a subset (n�79). Results: Correlations in the expected<br />

direction were observed for 116/124 PRO-CTCAE items with the QLQ-C30<br />

global health scale (mdn r�-.21; range .08 to -.57). Stronger correlations<br />

were seen for PRO-CTCAE items with conceptually related QLQ-C30<br />

domains: fatigue with physical, role and social functioning (-.54 to -.66);<br />

anxiety and depression with emotional functioning (-.54 to -.70); and<br />

concentration and memory problems with cognitive functioning (-.62 to<br />

-.72) [all p �0.001]. Fatigue, nausea, vomiting, pain, dyspnea, insomnia,<br />

appetite loss, constipation and diarrhea items strongly correlated with<br />

corresponding QLQ-C30 symptom scales (.69 to .79, all p �0.001).<br />

Scores for 98/124 PRO-CTCAE items were higher in the ECOG PS 2-4 vs<br />

0-1 group (mdn effect size .3). Test-retest reliability was observed across<br />

all tested items (mdn intraclass correlation coeff .77; range .53 to .96).<br />

Conclusions: Results demonstrate favorable validity and reliability <strong>of</strong><br />

PRO-CTCAE in a large, heterogeneous sample <strong>of</strong> patients undergoing<br />

cancer treatment. Further testing in multicenter treatment trials is underway.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


9049 General Poster Session (Board #39D), Sat, 8:00 AM-12:00 PM<br />

Patient-physician communication about code status preferences: A randomized<br />

controlled trial. Presenting Author: Wadih Rhondali, University <strong>of</strong><br />

Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: Code status discussions are important in cancer care. The best<br />

modality for such discussions has not been established. Our objective was<br />

to determine the impact <strong>of</strong> a physician ending a code status discussion with<br />

a question (autonomy approach) versus a recommendation (beneficence<br />

approach) on patients’ do-not-resuscitate (DNR) preference. Methods:<br />

Patients in a supportive care clinic watched two videos showing a<br />

physician-patient discussion regarding code status. Both videos were<br />

identical except for the ending: one ended with the physician asking for the<br />

patient’s code status preference and the other with the physician recommending<br />

DNR. Patients were randomly assigned to watch the videos in<br />

different sequences. The main outcome was the proportion <strong>of</strong> patients<br />

choosing DNR for the video patient. Results: 78 patients completed the<br />

study. 74% chose DNR after the question video, 73% after the recommendation<br />

video (p�NS). Median physician compassion score was very high<br />

and not different for both videos (p�0.73). 30/30 patients who had chosen<br />

DNR for themselves and 30/48 patients who had not chosen DNR for<br />

themselves chose DNR for the video patient (100% v/s 62%, p�0.001).<br />

Age (OR�1.1/year, p�0.01) and white ethnicity (OR�9.43, p�0.004)<br />

predicted DNR choice for the video patient. Conclusions: Ending DNR<br />

discussions with a question or a recommendation did not impact DNR<br />

choice or perception <strong>of</strong> physician compassion. Therefore, both approaches<br />

are clinically appropriate. All patients who chose DNR for themselves and<br />

most patients who did not choose DNR for themselves chose DNR for the<br />

video patient. Age and race predicted DNR choice.<br />

9051 General Poster Session (Board #39F), Sat, 8:00 AM-12:00 PM<br />

Psychosocial correlates <strong>of</strong> sleep architecture in women with advanced<br />

breast cancer. Presenting Author: Arianna Aldridge Gerry, Stanford University<br />

School <strong>of</strong> Medicine, Stanford, CA<br />

Background: Sleep disturbance is prevalent among metastatic breast<br />

cancer (MBC) patients and few studies have objectively evaluated this<br />

problem using polysomnography (PSG). Disturbed sleep negatively affects<br />

quality <strong>of</strong> life. The current study examines the relationship <strong>of</strong> mood and<br />

family structure with sleep parameters assessed over three nights <strong>of</strong> PSG (2<br />

at-home, 1 in-lab). Methods: MBC patients (n � 103) and healthy controls<br />

(n � 27) were recruited. Patients were 57.8 years (SD � 7.7), non-<br />

Hispanic white (86.3%), and had Karn<strong>of</strong>sky ratings <strong>of</strong> at least 70%.<br />

Multiple regression analyses assessed relationships among depression,<br />

cohabitation, and sleep parameters. Results: Among patients, sleep architecture<br />

was significantly related to depression and marital status/<br />

cohabitation. Women reporting more depressive symptoms had less deep<br />

sleep: lower percentage (B � -.26, p � .01) and fewer minutes <strong>of</strong> REM (B<br />

� -.23, p � .02), lower % (B � -.25, p � .02) and fewer minutes <strong>of</strong> stage 2<br />

sleep (B � -.28, p � .01), and more % stage 1 sleep (B � .28, p � .01).<br />

Marriage/cohabitation was positively related to sleep quality indicators:<br />

less wake time after sleep onset (B � -.20, p � .05), better sleep efficiency<br />

(B � .28, p � .01), more total sleep time (B � .30, p � .01), more minutes<br />

<strong>of</strong> REM (B � .23, p � .02), more stage 2 sleep (B � .27, p � .01), and less<br />

% stage 1 sleep (B � -.27, p � .01). Relative to healthy women, patients<br />

had more sleep stage transitions/hour (B � .21, p � .03), more awakenings<br />

(B � .18, p � .04), and more time awake after sleep onset (B � .21, p �<br />

.02). Slow wave (deep, non-REM) sleep was shorter for patients as<br />

measured by both minutes (B � -.20, p � .04) and % <strong>of</strong> stage 4 sleep (B �<br />

-.23, p � .02). Conclusions: Depressed patients or those living alone have<br />

greater reductions in deep sleep. Relative to healthy women, patients’ sleep<br />

disturbance and quality was worse. These findings demonstrate that the<br />

architecture <strong>of</strong> deep sleep is linked to depression and family structure.<br />

Patient and Survivor Care<br />

579s<br />

9050 General Poster Session (Board #39E), Sat, 8:00 AM-12:00 PM<br />

Smoking habits <strong>of</strong> relatives <strong>of</strong> patients (pts) with cancer. Presenting<br />

Author: Mutlu Hayran, Hacettepe University Institute <strong>of</strong> Oncology, Ankara,<br />

Turkey<br />

Background: We aimed to determine the rate and habitual patterns <strong>of</strong><br />

smoking, intentions <strong>of</strong> cessation, dependence levels and sociodemographic<br />

characteristics <strong>of</strong> relatives <strong>of</strong> cancer patients. Methods: The<br />

smoking habits <strong>of</strong> relatives <strong>of</strong> cancer pts were evaluated with a questionnaire<br />

and Fagerstrom test (FT) <strong>of</strong> nicotine dependence. FT and a questionnaire<br />

form were filled in by the volunteers. The quit rate <strong>of</strong> the relatives <strong>of</strong><br />

the cancer pts was compared with the rates declared in the literature.<br />

Results: A total <strong>of</strong> 560 subjects who have relatives <strong>of</strong> pts with cancer. The<br />

median age <strong>of</strong> those with lower and higher Fagerstrom scores (FS) were 40<br />

years and 42 years, respectively (p�0.001). We found no association<br />

between FS and sociodemographic variables, such as the subject’s medical<br />

status, gender, living in the same house, educational status, family income,<br />

closeness and time spent with the cancer pts. Only 2% <strong>of</strong> the subjects<br />

started smoking after cancer was diagnosed in their relative and almost<br />

20% <strong>of</strong> subjects had quit smoking within the year. Spontaneous quit rates<br />

<strong>of</strong> these subgroup <strong>of</strong> smokers (20%) was significantly higher than reported<br />

self-quit rates (max. ~10%) in the literature (p�0.001). Conclusions: The<br />

FS is known to be helpful in determining who would benefit from a cigarette<br />

smoking cessation program. We also show here that the cancer diagnosis in<br />

a smoker’s family motivates this individual to quit smoking and may create<br />

a time window for any involved health staff to intervene for help. This study<br />

was designed by the Turkish Oncology Group, Epidemiology and Prevention<br />

Subgroup.<br />

Individual characteristics by Fagerstrom score.<br />

Low FS High FS<br />

n % n % p<br />

Sex F 33 27 18 21 0.358<br />

M 89 73 66 79<br />

Education Literate 4 3 4 5 0.183<br />

Primary 45 37 33 39<br />

High 34 28 30 35<br />

University 39 32 18 21<br />

Income �1000 TL 58 49 31 39 0.266<br />

1001-3000 TL 43 36 38 47<br />

�3000 TL 18 15 11 14<br />

Finds smoking bans effective No 44 36 44 48 0.028<br />

Yes 77 64 41 52<br />

Wants pr<strong>of</strong>essional help No 55 46 37 45 0.860<br />

Yes 65 54 46 55<br />

Obeys smoking bans No 10 8 17 20 0.012<br />

Yes 112 92 67 80<br />

Contemplates quitting No 38 31 31 36 0.001<br />

Indecisive 6 5 5 6<br />

Yes 77 64 49 58<br />

9052 General Poster Session (Board #39G), Sat, 8:00 AM-12:00 PM<br />

Mistletoe as complementary treatment in patients with advanced non-small<br />

cell lung cancer treated with carboplatin-based combinations: A randomized<br />

phase II study. Presenting Author: Gil Bar-Sela, Division <strong>of</strong> Oncology,<br />

Rambam Health Care Campus, Haifa, Israel<br />

Background: The mistletoe preparation, iscador, is commonly used in<br />

complementary medicine for improving the quality <strong>of</strong> life (QoL) <strong>of</strong> cancer<br />

patients, but its efficacy needs to be further proved. This randomized phase<br />

II study was aimed at assessing the value <strong>of</strong> iscador as a modifier <strong>of</strong><br />

chemotherapy-induced toxicity in chemotherapy-naïve non-small-cell lung<br />

cancer (NSCLC) patients treated with carboplatin containing regimens.<br />

Methods: Patients with stage IIIA or IIIB/IV NSCLC, ECOG performance<br />

status 0-2, and no history <strong>of</strong> brain metastasis were randomized to receive<br />

either chemotherapy alone (arm-1) or chemotherapy plus iscador Q 10mg<br />

SC injections 3 times weekly until tumor progression (arm-2). Chemotherapy<br />

consisted <strong>of</strong> 21-day cycles <strong>of</strong> IV carboplatin area under curve 5.0,<br />

day 1, combined with IV gemcitabine 1000mg/m2 , days 1, 8, or with IV<br />

pemetrexed 500mg/m2 , day 1. Results: Seventy-two patients were enrolled,<br />

39 to arm-1 and 33 to arm-2. The arms were well balanced except that<br />

more patients with PS-0 were in arm-1 (p�0.041). Most patients (65%)<br />

were in stage IV and 62% had squamous histology. Median overall survival<br />

was 11 months in both arms. Median TTP was 4.8 months (arm-1) and 6<br />

months (arm-2) (p�0.07). Carboplatin dose reductions and delays in<br />

gemcitabine on day 8 were more frequent in arm-1 (44% vs. 13% in arm-2,<br />

p�0.005; 31% vs. 13% in arm-2, p� 0.013). The difference in grade 3-4<br />

hematological toxicity was not significant. More non-hematological toxicity<br />

was observed in arm-1 (41% compared to 16% in arm-2, p�0.043). The<br />

number <strong>of</strong> hospitalizations was higher in arm-1, 54%, compared to 24% in<br />

arm-2 (p�0.016). No significant difference was seen in QoL using the<br />

EORTC QLQ-C30 questionnaire; However, significant improvement in<br />

arm-2 was seen in coughing (p�0.013) and peripheral neuropathy<br />

(p�0.019). Conclusions: Chemotherapy dose reductions, severe nonhematological<br />

side effects, and hospitalizations were less frequent in<br />

patients treated with iscador. Further investigation <strong>of</strong> iscador as a modifier<br />

<strong>of</strong> chemotherapy-related toxicity is warranted.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


580s Patient and Survivor Care<br />

9053 General Poster Session (Board #39H), Sat, 8:00 AM-12:00 PM<br />

3D index calculated from duration and severity <strong>of</strong> neutropenia and a degree<br />

<strong>of</strong> fever as prognostic factors predicting mortality <strong>of</strong> chemotherapy-induced<br />

febrile neutropenia in hematologic malignancies. Presenting Author: Ryosuke<br />

Shirasaki, Department <strong>of</strong> Hematology/Oncology, Teikyo University<br />

School <strong>of</strong> Medicine, Tokyo, Japan<br />

Background: Risk stratification <strong>of</strong> patients with febrile neutropenia (FN) is<br />

important to select the suitable therapeutic option. Although the MASCC<br />

scoring system is used as a reliable risk-discriminator, the objective<br />

evaluation <strong>of</strong> “burden <strong>of</strong> illness” has been difficult to be pointed-out. The<br />

latest IDSA Guidelines on FN proposes a set <strong>of</strong> risk factors based on the<br />

expert’s recommendations. The present study demonstrates usefulness <strong>of</strong><br />

newly proposed 3D-index on predicting mortality <strong>of</strong> chemotherapy-induced<br />

FN in hematologic malignancies (HM). Methods: Patients with HM admitted<br />

to our hospital between Jan 2008 and Dec 2011 were enrolled, and<br />

data from 282 FN episodes in 129 FN patients including 20 infectionassociated<br />

deaths were retrospectively analyzed to determine useful<br />

prognostic factors on the mortality <strong>of</strong> FN. Correlation between mortality and<br />

59 characteristics including 3D-index were examined by univariate analysis<br />

and receiver operating characteristic curve analysis. 3D-index is a<br />

duration and severity <strong>of</strong> neutropenia and degree <strong>of</strong> fever. Total duration <strong>of</strong><br />

days during neutropenia was not calculate at the time <strong>of</strong> risk evaluation.<br />

3D-indexes after three and five days from the start <strong>of</strong> FN were also<br />

analyzed. Results: 32 out <strong>of</strong> 59 characteristics were significantly correlated<br />

with mortality by univariate analysis. Among them, 3D-index and 3Dindexes<br />

after three and five days were demonstrated as more useful factors<br />

(3D-index, p�0.0015; 3D-index after three days, p�0.0030; 3D-index<br />

after five days, p�0.0044). And 3D-index after three days over 4000<br />

indicates patients’ mortality above 20%, index 2600-4000 15-20% ,<br />

index 900-2600 10-15%, and index below 900 below 10%, respectively.<br />

Conclusions: 3D-indexes after three days were suggested as useful predictors<br />

for infection-related mortality during FN. It was suggested that FN<br />

patients were categorized into four risk groups according to the value.<br />

9055 General Poster Session (Board #40B), Sat, 8:00 AM-12:00 PM<br />

Comparison <strong>of</strong> three different radiation fractionation schedules in the<br />

palliation <strong>of</strong> painful bone metastasis. Presenting Author: Monica Malik,<br />

Nizam’s Institute <strong>of</strong> Medical Sciences, Hyderabad, India<br />

Background: The purpose <strong>of</strong> this study was to compare the efficacy <strong>of</strong> a<br />

single fraction versus two multifractionated regimens in the palliation <strong>of</strong><br />

painful bone metastases. Methods: Patients with painful bone metastases<br />

were randomized into three groups. Group I received a dose <strong>of</strong> 8 Gy in a<br />

single fraction, Group II received 20 Gy in five fractions and Group III<br />

patients received 30 Gy in 10 fractions. Pain score, ECOG performance<br />

status and analgesic requirement were recorded at baseline and at 1, 4, 8<br />

and 12 weeks following treatment. Pain score was recorded on a 5-point<br />

verbal rating scale from 0 (no pain) to 4 (extremely severe pain). Overall<br />

response was defined as decrease in pain score by at least one point.<br />

Complete response was defined as achieving a pain score <strong>of</strong> zero at any<br />

point during follow-up. Duration <strong>of</strong> overall response was defined as the time<br />

from initial response till return <strong>of</strong> pain to its baseline value. Results: 45<br />

patients were included with 15 in each group. Median age was 55 years<br />

(range 29-78 years). 17(37.7%) had metastasis in pelvic bones; 17(37.7%)<br />

in the spine while the remainder in the appendicular bones. Overall<br />

response rates in Groups I, II and III at week 1 were 60%, 53.3% and 60%<br />

respectively (p�0.71). At 1 month, overall response rates were 71.4%,<br />

73.3% and 73.3% (p�0.84) and at 3 months; 78.5%, 80% and 80%<br />

respectively (p�0.86). The rate <strong>of</strong> complete response in all the three<br />

groups was 20%. Improvement in performance status in Groups I, II and III<br />

was seen in 60%, 66% and 80% respectively (p�0.69). Analgesic usage<br />

decreased in 86%, 87% and 80% patients in groups I, II and III<br />

respectively (p�0.66). Out <strong>of</strong> the nine complete responders, two sustained<br />

the response for less than four weeks, four patients up to eight weeks and<br />

remaining three till the end <strong>of</strong> follow-up. There was no statistically<br />

significant difference in between the three arms among all the variables<br />

compared. Conclusions: All three groups showed equal efficacy in pain<br />

palliation, analgesic requirement, improvement in performance status and<br />

duration <strong>of</strong> response. In patients with very advanced disease and short life<br />

expectancy, where the treatment goal is to decrease pain, 8 Gy in single<br />

fraction is a convenient and cost effective schedule.<br />

9054 General Poster Session (Board #40A), Sat, 8:00 AM-12:00 PM<br />

A positron emission tomography (PET) study to assess the degree <strong>of</strong><br />

neurokinin-1 (NK1) receptor occupancy in the human brain after single<br />

doses <strong>of</strong> netupitant to healthy male subjects. Presenting Author: Giorgia<br />

Rossi, Helsinn Healthcare SA, Lugano, Switzerland<br />

Background: Netupitant (NETU) is a highly selective neurokinin 1 (NK1) receptor antagonist for the prevention <strong>of</strong> nausea and vomiting associated<br />

with emetogenic chemotherapy. In this study, PET imaging with the NK1 receptor-binding–selective tracer 11C-GR205171 was used to determine<br />

the levels and the duration <strong>of</strong> central NK1 receptor occupancy (RO)<br />

achieved by therapeutic doses <strong>of</strong> NETU. Methods: This was a single dose,<br />

randomized, open-label, PET study investigating the degree <strong>of</strong> NK1 RO in<br />

human brain after single oral doses <strong>of</strong> NETU (100, 300 or 450 mg) in 6<br />

healthy male subjects. PET scans and blood samples for NETU determination<br />

were obtained up to 96 hrs post dose. The Patlak model was used to<br />

define the net uptake rate <strong>of</strong> 11C-GR205171 in the brain regions <strong>of</strong> interest<br />

while the percent <strong>of</strong> NK1 RO was calculated as the relative difference<br />

between the uptake rate at baseline and post-treatment administration.<br />

Results: NETU plasma concentrations over the 100 mg to 450 mg dose<br />

range were comparable with those obtained in previous single dose studies.<br />

ANK1RO <strong>of</strong> 90% or higher was achieved with all tested single oral doses in<br />

the majority <strong>of</strong> the outlined brain regions 6 hrs after dosing (corresponding<br />

to netupitant Cmax). All doses had a long duration <strong>of</strong> blockade <strong>of</strong> NK1 receptors with the 300 mg dose showing moderate (62%) to high (94%)<br />

NK1 RO for all investigated brain regions at 96 hrs post dose. The<br />

relationship between NETU concentration and NK1 RO, assessed using a<br />

sigmoid Emax model, indicated that in the striatum, the reference brain<br />

area with the highest NK1 receptor expression, a concentration <strong>of</strong> 225 �g/L<br />

<strong>of</strong> NETU corresponded to an NK1 RO <strong>of</strong> 90%. This data suggests that a<br />

single oral dose between 100 and 300 mg NETU would be needed to reach<br />

90% NK1 RO levels in human brain. Conclusions: PET results demonstrate<br />

that NETU is a potent agent targeting NK1 receptors with a high degree <strong>of</strong><br />

occupancy for a long duration. NETU, given as single doses <strong>of</strong> 100 to 450<br />

mg was well tolerated.<br />

9056 General Poster Session (Board #40C), Sat, 8:00 AM-12:00 PM<br />

Pulmonary symptoms and their importance to patients with non-small cell<br />

lung cancer undergoing second- and third-line chemotherapy. Presenting<br />

Author: Susan Magasi, Northwestern University Feinberg School <strong>of</strong> Medicine,<br />

Chicago, IL<br />

Background: As new cancer therapies are developed, it is important to<br />

evaluate their efficacy based not only on survival outcomes but also<br />

meaningful patient benefit in terms <strong>of</strong> symptoms and concerns that are<br />

important to patients. The purpose <strong>of</strong> this study <strong>of</strong> patients undergoing<br />

2nd /3rd line chemotherapy for non-small cell lung cancer (NSCLC) was to<br />

characterize pulmonary symptoms and the importance patients place on<br />

these symptoms, as a part <strong>of</strong> assessing the content validity <strong>of</strong> the<br />

Pulmonary Symptom Index (PSI) <strong>of</strong> the Functional Assessment <strong>of</strong> Cancer<br />

Therapy – Lung (FACT-L). Methods: We conducted semi-structured thematic<br />

interviews with 20 stage III/IV NSCLC patients undergoing 2nd or 3rd line treatment. Interviews included open elicitation <strong>of</strong> NSCLC symptoms<br />

and their functional impact, and participant ratings <strong>of</strong> the relative importance<br />

<strong>of</strong> pulmonary symptoms. Results: Mean age was 62 years (range<br />

30-79); 80% had non-squamous histology, and 30% had co-morbid COPD.<br />

While participants described a range <strong>of</strong> symptom experiences and severity,<br />

a core set <strong>of</strong> pulmonary symptoms emerged - shortness <strong>of</strong> breath (reported<br />

by 16/20, 13/20 rated as very important), cough (reported by 14/20, 10/20<br />

rated as very important), and chest tightness (reported by 15/20, 9/20<br />

rated as very important); these matched key symptoms in the PSI.<br />

Symptom importance ratings were influenced by the functional impact they<br />

had upon patient ability to perform valued roles and responsibilities.<br />

Patients described diverse coping strategies including breaking down<br />

activities into manageable tasks, priority setting, physical assistance,<br />

emotional support, and health promoting changes in diet and exercise.<br />

Ninety percent (18/20) <strong>of</strong> participants reported tiredness or fatigue; most<br />

said it was “very important.” A quarter (5/20) <strong>of</strong> the participants reported<br />

having no pulmonary symptoms. Conclusions: These results in NSCLC<br />

patients undergoing 2nd /3rd line treatment support the importance, relevance,<br />

and impact <strong>of</strong> core pulmonary symptoms included in the PSI and<br />

<strong>of</strong>fer initial evidence that its content may constitute a key element <strong>of</strong><br />

patient benefit in evaluating the efficacy <strong>of</strong> new cancer therapies.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


9057 General Poster Session (Board #40D), Sat, 8:00 AM-12:00 PM<br />

Widespread use <strong>of</strong> complementary and alternative medicine (CAM) among<br />

non-Hodgkin lymphoma (NHL) survivors. Presenting Author: Alexis D. Leal,<br />

Mayo Clinic, Rochester, MN<br />

Background: The incidence <strong>of</strong> CAM use among patients with cancer is<br />

higher when compared to the general population. However, there are few<br />

studies examining CAM use in NHL survivors, and limited data are available<br />

regarding beliefs in CAM. This study was conducted to examine the<br />

prevalence <strong>of</strong> CAM use in NHL, define CAM beliefs among NHL survivors,<br />

and explore differences between patients with indolent and aggressive<br />

lymphoma. Methods: Newly diagnosed lymphoma patients were prospectively<br />

enrolled within 9 months <strong>of</strong> diagnosis in the University <strong>of</strong> Iowa/Mayo<br />

Clinic SPORE Molecular Epidemiology Resource from 2002-2008. NHL<br />

patients who completed the 3-year post diagnosis questionnaire, which<br />

includes questions regarding CAM use and beliefs, were included in this<br />

study. Chi-squared tests and Wilcoxon rank-sum tests were used to assess<br />

the association <strong>of</strong> CAM use with prognostic and demographic factors.<br />

Results: 719 patients were included with a median age <strong>of</strong> 63 years (range<br />

22-92). 53% were male. Overall, 636 (89%) reported ever using CAM.<br />

78% <strong>of</strong> patients used vitamins and 54% alternative therapies (chiropractic<br />

(36%) and massage therapy (24%)). Among CAM users, 141 (22%)<br />

believe CAM can assist the body to heal, 123 (19%) believe CAM can<br />

relieve cancer symptoms, 115 (18%) believe CAM use gives a feeling <strong>of</strong><br />

control, 106 (17%) believe CAM can boost immunity, 24 (4%) believe<br />

CAM can cure cancer, and 35 (6%) believe CAM can prevent the spread <strong>of</strong><br />

cancer. Female gender was associated with increased overall CAM use<br />

(p�0.0001) as well as use <strong>of</strong> vitamins (p�0.0001), herbal supplements<br />

(p�0.006) and alternative therapy (p�0.0002) specifically for cancer.<br />

Older age was also associated with increased vitamin use (p�0.005) and<br />

decreased herbal supplements use (p�0.008). There was no significant<br />

difference in overall CAM use between those with follicular lymphoma<br />

grades I-II (n�195, 91%) and non-relapsed diffuse large B-cell lymphoma<br />

(n�151, 87%), although massage therapy was utilized more <strong>of</strong>ten by FL<br />

survivors (29% versus 18%, p�0.005). Conclusions: CAM modalities are<br />

used by the majority <strong>of</strong> NHL survivors (89%). The assessment <strong>of</strong> CAM use<br />

and education regarding potential harms is imperative for the NHL survivor.<br />

9059 General Poster Session (Board #40F), Sat, 8:00 AM-12:00 PM<br />

Prevention <strong>of</strong> palmoplantar erythrodysesthesia (PPE) with an antiperspirant<br />

in breast cancer patients treated with pegylated liposomal doxorubicin<br />

(PLD), a placebo-controlled, double blinded, phase lll trial (SAKK 92/08).<br />

Presenting Author: Thomas Ruhstaller, Swiss Group for <strong>Clinical</strong> Research,<br />

Berne, Switzerland<br />

Background: PPE, also known as hand-foot syndrome, is a distinctive<br />

adverse drug reaction <strong>of</strong> PLD treatment. PLD has been detected in elevated<br />

concentrations in eccrine sweat glands in palms and soles. We postulated<br />

that prophylactic administration <strong>of</strong> an antiperspirant (F511 cream) prior<br />

and during treatment with PLD could decrease the incidence <strong>of</strong> PPE.<br />

Methods: Patients (pts) with metastatic breast cancer treated with PLD<br />

monotherapy �10mg/m2 per week applied an antiperspirant to the left or<br />

right hand and foot and a corresponding placebo to the opposite site with<br />

double-blinding for the content <strong>of</strong> the cream applied to either side<br />

(intra-patient randomization). The creams were applied once daily during<br />

the first week, then three times per week. The primary endpoint was the rate<br />

<strong>of</strong> PPE grade (G) � 2 in the antiperspirant or placebo treated side. Pts were<br />

evaluable if they developed PPE G � 2 or had received cumulatively at least<br />

160mg/m2 PLD. Patient-reported extent <strong>of</strong> symptom burden was a secondary<br />

endpoint. Using McNemar’s matched pairs design 53 pts were needed<br />

to detect a difference <strong>of</strong> 20% between the sides with a significance level <strong>of</strong><br />

5% and power <strong>of</strong> 90%. Results: 52 <strong>of</strong> 90 pts from 11 Swiss centers<br />

included were evaluable. Median age was 64.5 years; median duration <strong>of</strong><br />

PLD treatment was 12 weeks. 30 pts developed PPE G � 2. In 3 pts PPE G<br />

� 2 occurred on the placebo side but not on the antiperspirant side<br />

(p�0.097; table). PPE G � 2 was borderline significantly more frequent in<br />

placebo foot than antiperspirant foot (p�0.048). Patient-reported extent <strong>of</strong><br />

symptom burden showed a trend in favor <strong>of</strong> the antiperspirant side for skin<br />

problems (peeling, blistering, bleeding) in the group <strong>of</strong> pts with PPE G � 2<br />

(p�0.051). Conclusions: In this double-blind trial with intra-patient randomization<br />

we observed a trend towards less PPE G � 2 with application <strong>of</strong> the<br />

antiperspirant cream F511 in pts treated with PLD as determined by the<br />

treating physician and reported by the pts.<br />

Antiperspirant side<br />

PPE grade > 2 toxicity<br />

No Yes Total<br />

Placebo side<br />

No 22 (42%) 0 (0%) 22 (42%)<br />

Yes 3 (6%) 27 (52%) 30 (58%)<br />

Total 25 (48%) 27 (52%) 52 (100%)<br />

Patient and Survivor Care<br />

581s<br />

9058 General Poster Session (Board #40E), Sat, 8:00 AM-12:00 PM<br />

Prospective survey study regarding the implementation <strong>of</strong> new communication<br />

skills curriculum for medical oncology trainees. Presenting Author:<br />

Nathan M. Shumway, San Antonio Military Medical Center, Ft. Sam<br />

Houston, TX<br />

Background: After a diagnosis <strong>of</strong> cancer, many patients suffer from anxiety<br />

and distress from uncertainty <strong>of</strong> symptoms, treatments, and prognosis.<br />

Clinicians must recognize opportunities to explore concerns. This study<br />

assesses oncology trainees’ views about communication with cancer<br />

patients before (PRE) and after (POST) a 12 month curriculum. Methods:<br />

Medical oncology fellows were surveyed PRE and POST a communication<br />

curriculum consisting <strong>of</strong> case studies and 6 core lectures which included<br />

fundamentals, breaking bad news, transitions to palliative care, advanced<br />

care planning, conducting family conferences, and discussing treatment<br />

options and informed consent. A 5 point Likert-scale was used to measure<br />

fellows’ attitudes and comfort regarding communication PRE and POST<br />

with questions grouped according to core topics. (�2 � trainee disagreement,<br />

3�neutral opinion, and � 4 indicated agreement). Results: PRE and<br />

POST surveys were completed by 11 and 8 trainees respectively. In PRE<br />

100% felt communication skills were important and 63% believed these<br />

skills could be taught. 82% felt there was not enough time during most<br />

visits to address emotion. This decreased to 27% in POST. 18% PRE<br />

agreed they were comfortable recognizing coping mechanisms versus<br />

100% POST. 45% felt comfortable eliciting values in PRE versus 87.5%<br />

POST. Only 1 fellow (9%) felt comfortable addressing futility in PRE versus<br />

25% in POST. The median grouped score for fundamentals increased from<br />

15 to 17 (p�0.016) and median grouped score for advance care plans and<br />

DNR increased from 11 to 13(p�0.026). Conclusions: There is a need to<br />

improve oncology communication skills. Our curriculum is just one approach.<br />

After intervention, the majority <strong>of</strong> fellows agreed there was enough<br />

time to address emotion and felt more comfortable recognizing coping<br />

mechanisms and eliciting values. Discussing futility remains difficult for<br />

our fellows. All fellows were more comfortable with fundamental communication<br />

skills and advance care plans after the curriculum. The extent the<br />

curriculum contributed to the change in survey results is unclear. Further<br />

research is needed to guide communication education <strong>of</strong> oncologists.<br />

9060 General Poster Session (Board #40G), Sat, 8:00 AM-12:00 PM<br />

Pharmacokinetics, pharmacodynamics, and tolerability <strong>of</strong> BCD-017, a<br />

novel pegylated filgrastim: Results <strong>of</strong> open-label controlled phase I study<br />

with dose escalation in healthy volunteers. Presenting Author: Kirill D.<br />

Nikitin, BIOCAD, Moscow, Russia<br />

Background: Pegfilgrastim (conjugate <strong>of</strong> filgrastim and 20 kDa PEG) is<br />

approved for treatment <strong>of</strong> chemotherapy-associated neutropenia. BCD-017<br />

is a covalent conjugate <strong>of</strong> filgrastim with 30 kDa PEG. Increased molecular<br />

weight <strong>of</strong> PEG molecule may provide additional benefits compared to<br />

pegfilgrastim. We have conducted this open-label phase I study to assess<br />

the PK, PD and tolerability <strong>of</strong> BCD-017. Methods: 24 healthy male<br />

volunteers signed the informed consent and were sequentially assigned to<br />

receive 1, 3 or 9 mg <strong>of</strong> BCD-017 or 5 mcg/kg/day <strong>of</strong> filgrastim for 7 days, 6<br />

volunteers per group. Outcome measures included absolute neutrophil<br />

count (ANC) and �D34� cell count, PK parameters and adverse events<br />

(AEs). Results: BCD-017 induced a fast and significant increase <strong>of</strong> ANC.<br />

Median maximum ANC (ANCmax) for BCD-017 1, 3, 9 mg and filgrastim<br />

was 18.68 (10.62-21.02), 25.92 (15.43-28.07), 32.22 (18.22-45.79),<br />

and 28.21 (21-31.95) �103 cells/mm3 , respectively; median time to<br />

ANCmax was 24 (12-24); 48 (24-72); 72 (48-72); and 132,5 (12-169) h,<br />

respectively; median increase in �D34� cells number 96 h post dose was<br />

4.7 (1.2-6.5), 6.5 (1.7-12.3), 40.9 (24.5-102), and 17.8 (3.3-35.2)<br />

times, respectively. Filgrastim serum concentration was analyzed using<br />

ELISA. Median Cmax for BCD-017 3 and 9 mg and filgrastim was 45<br />

(31-65), 446 (191-649), and 40 (20-54) ng/mL, respectively; median<br />

Tmax was 48 (24-72), 36 (24-48), and 8 (6-8) h respectively; median T1/2 was 65 (51-70), 46 (41-57), and 6.7 (6.2-7.6) h, respectively. BCD-017<br />

was well tolerated. No dose-limiting AEs were observed. AEs included<br />

headache, back pain, myalgia, arthralgia, thrombocytopenia, hyperuricemia,<br />

alkaline phosphatase/LDH increased. All AEs were <strong>of</strong> grade 1-2.<br />

Compared to filgrastim, the best tolerability was observed in 3 mg group.<br />

Conclusions: BCD-017 is shown to be a potent stimulator <strong>of</strong> granulopoiesis.<br />

BCD-017 3 mg did not differ from filgrastim in terms <strong>of</strong> ANC increase and<br />

its safety was shown in healthy volunteers. Further phase II study <strong>of</strong><br />

BCD-017 for treatment and prophylaxis <strong>of</strong> neutropenia in patients receiving<br />

cytotoxic chemotherapy is necessary.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


582s Patient and Survivor Care<br />

9061 General Poster Session (Board #40H), Sat, 8:00 AM-12:00 PM<br />

Randomized double-blind evaluation <strong>of</strong> dronabinol for the prevention <strong>of</strong><br />

chemotherapy-induced nausea. Presenting Author: Steven M. Grunberg,<br />

Fletcher Allen Health Care, Burlington, VT<br />

Background: Although great progress has been made in the control <strong>of</strong><br />

chemotherapy-induced vomiting (CIV), prevention <strong>of</strong> chemotherapyinduced<br />

nausea (CIN) has been less successful. Preliminary data suggests<br />

that some families <strong>of</strong> lesser used antiemetic agents, such as the cannabinoids,<br />

may have greater efficacy against nausea than against vomiting.<br />

Methods: Adult solid tumor patients (pts) receiving cyclophosphamide<br />

� 1500 mg/m2 (C) and/or doxorubicin � 40 mg/m2 (A) were eligible. Pts<br />

could have received prior mildly emetogenic chemotherapy (EC). Pts were<br />

not eligible who were receiving other moderately or highly EC, were<br />

receiving cranial, abdominal or pelvic radiotherapy, had CIV/CIN with<br />

previous chemotherapy, had other causes for nausea/vomiting besides<br />

chemotherapy, or were scheduled to receive other antiemetics. Pts with<br />

habitual cannabinoid use were not eligible. All pts received palonosetron<br />

0.25 mg (PALO) and dexamethasone 10 mg (DXM) IV before chemotherapy.<br />

Patients were randomized double-blind to receive dronabinol 5 mg<br />

(D) or matching placebo (P) 3 times a day for 5 days. Nausea, vomiting and<br />

toxicity data was collected daily for 5 days. Results: 62 pts were entered on<br />

study – female/male 61/1, White/Black/Hispanic/Other 45/14/2/1, median<br />

age (range) 58 (29-76). No significant difference was noted in CIVdependent<br />

endpoints (including No Vomiting, Complete Response, or<br />

Complete Protection) or in rescue medication use. However pts receiving D<br />

had a shorter duration <strong>of</strong> nausea – Mean number <strong>of</strong> days <strong>of</strong> nausea (D vs P)<br />

1.86 days vs 3.10 days (p�0.027) – and a trend toward greater frequency<br />

<strong>of</strong> No Nausea (D vs P) 37% vs 17% (p�0.143). Common toxicities<br />

included fatigue (D/P 17/11), headache (D/P 16/16), dizziness (D/P 14/7),<br />

constipation (D/P 14/11), and diarrhea (D/P 13/6) No pt discontinued<br />

therapy due to mood changes. Conclusions: Low-dose D decreased the<br />

duration <strong>of</strong> CIN and increased the frequency <strong>of</strong> No Nausea in pts receiving<br />

C and/or A. Agents to prevent CIV (such as PALO and DXM) and to prevent<br />

CIN (such as D) have complementary activity and result in improved overall<br />

control when used together.<br />

9063 General Poster Session (Board #41B), Sat, 8:00 AM-12:00 PM<br />

Decision-making preferences <strong>of</strong> advanced cancer Hispanic patients from the<br />

United States and Latin America. Presenting Author: Henrique Afonseca<br />

Parsons, University <strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: To determine whether preferences in frequency <strong>of</strong> passive decision<br />

making differ between Hispanic patients from Latin America (HLA) and<br />

Hispanic-<strong>American</strong> (HA) patients. Methods: We conducted a survey <strong>of</strong> advanced<br />

cancer Hispanic patients referred to outpatient palliative care clinics in the U.S,<br />

Chile, Argentina, and Guatemala. Information on demographic variables, PS,and-<br />

Marin Acculturation Assessment Tool (only U.S. patients) was collected.<br />

Decision-making preference was evaluated by the decision-making assessment<br />

tool. Results: A total <strong>of</strong> 387 patients with advanced cancer were surveyed: 91<br />

(24%) in the US, 100 (26%) in Chile, 94 (25%) in Guatemala, and 99 (26%) in<br />

Argentina. Median age was 59 years, and 61% were female. HLA preferred<br />

passive decision-making strategies significantly more frequently with regard to<br />

involvement <strong>of</strong> the family (24% versus 10%, p�0.009) or the physician (35%<br />

versus 26%, p�0.001), even after controlling for age and education (OR 3.8,<br />

p�0.001 for physician and 2.4, p�0.03 for family) (Table 1). 76/91 HA<br />

(83.5%), and 242/293 HLA (82%) preferred family involvement in decisionmaking<br />

(p�NS). No differences were found in decision-making preferences<br />

between low- and highly acculturated U.S. Hispanics. Conclusions: HA prefer<br />

more active decision-making as compared to HLA. Among HA, acculturation did<br />

not seem to play a role in decision-making preference determination. Our<br />

findings in this study confirm the importance <strong>of</strong> family participation in decision<br />

making in both HA and HLA. However, HA patients were much less likely to want<br />

family members or physicians to make decisions on their behalf.<br />

Distribution <strong>of</strong> decision-making preferences according to the dyadic independent<br />

relationships between patient and family.<br />

USA<br />

(N�90)<br />

Latin America<br />

(N�296)<br />

n (%) n (%) p<br />

Passive 9 (10%) 69 (24%) �0.001<br />

Shared 45 (50%) 144 (49%)<br />

Active 32 (36%) 64 (22%)<br />

Didn’t know/preferred not to answer 4 (4%) 17 (6%)<br />

Distribution <strong>of</strong> decision-making preferences according to the triadic simultaneous<br />

relationships between patient, physician, and family.<br />

USA<br />

(N�91)<br />

Latin America<br />

(N�293)<br />

n (%) n (%)<br />

p<br />

Passive 13 (14%) 69 (23.5%) �0.001<br />

Shared 31 (34%) 151 (51.5%)<br />

Active 47 (52%) 73 (25%)<br />

9062 General Poster Session (Board #41A), Sat, 8:00 AM-12:00 PM<br />

The effect <strong>of</strong> melatonin on appetite and other symptoms in patients with<br />

advanced cancer and cachexia: A double-blind placebo-controlled trial.<br />

Presenting Author: Egidio Del Fabbro, University <strong>of</strong> Texas M. D. Anderson<br />

Cancer Center, Houston, TX<br />

Background: Patients with advanced cancer experience anorexia and weight<br />

loss which impairs their quality <strong>of</strong> life. Prior studies suggest melatonin, a<br />

frequently used integrative medicine may attenuate weight loss, anorexia,<br />

fatigue, and depression. These studies were limited by a lack <strong>of</strong> blinding<br />

and absence <strong>of</strong> placebo controls. The primary objective <strong>of</strong> this study was to<br />

compare melatonin to placebo for appetite in patients with cachexia.<br />

Methods: A randomized, double-blind, 28 day trial <strong>of</strong> melatonin 20mg vs.<br />

placebo in patients with advanced lung or gastrointestinal cancer, appetite<br />

scores �3 ona0to10scale (10 � worst appetite) and a history <strong>of</strong> weight<br />

loss � 5% within 6 months. Patients unable to maintain oral intake, thyroid<br />

or adrenal dysfunction, or with a karn<strong>of</strong>sky �40 were excluded from the<br />

study. The assessments included weight, symptom severity by Edmonton<br />

Symptom Assessment Scale (ESAS) and quality <strong>of</strong> life by the Functional<br />

Assessment <strong>of</strong> Anorexia/Cachexia Therapy (FAACT).Differences between<br />

groups from baseline to day 28 were analyzed using one-sided two sample t<br />

tests (appetite, pain and well-being) or Wilcoxon two-sample tests for the<br />

other variables. Interim analysis at half point had a Lan-DeMets monitoring<br />

boundary with an O’Brien-Fleming stopping rule. The decision boundaries<br />

for the interim test was to accept the null hypothesis <strong>of</strong> no treatment<br />

difference (futility) if the test statistic Z � 0.39 (p � 0.348). Results: After<br />

interim analysis <strong>of</strong> 48 patients, the study was closed by the Data Safety<br />

Monitoring Board for futility. There were no significant differences between<br />

groups in appetite (p�0.78), weight (p� 0.17), FAACT score (p�0.95),<br />

insomnia (p�0.62) or other symptoms measured by the ESAS from<br />

baseline to day 28.No significant toxicities were observed. Conclusions: In<br />

cachectic patients with advanced cancer, 20mg oral Melatonin at night<br />

does not improve appetite, weight or quality <strong>of</strong> life compared to placebo.<br />

9064 General Poster Session (Board #41C), Sat, 8:00 AM-12:00 PM<br />

The use <strong>of</strong> olanzapine versus metoclopramide for the treatment <strong>of</strong> breakthrough<br />

chemotherapy-induced nausea and vomiting (CINV) in patients<br />

receiving highly emetogenic chemotherapy. Presenting Author: Rudolph M.<br />

Navari, Indiana University School <strong>of</strong> Medicine South Bend, South Bend, IN<br />

Background: Olanzapine (OLN) has been shown to be a safe and effective<br />

agent for the prevention <strong>of</strong> CINV. OLN may also be an effective rescue<br />

medication for patients who develop breakthrough CINV despite having<br />

received guideline directed CINV prophylaxis. Methods: A double blind,<br />

randomized phase III trial was performed for the treatment <strong>of</strong> breakthrough<br />

CINV in chemotherapy naïve patients receiving highly emetogenic chemotherapy<br />

(HEC) (cisplatin, �70 mg/m2 , or doxorubicin, �50 mg/m2 and<br />

cyclophosphamide, � 600mg/m2 ) comparing OLN to Metoclopramide<br />

(METO). Patients who developed breakthrough emesis or nausea despite<br />

prophylactic dexamethasone (12 mg IV), palonosetron (0.25 mg IV), and<br />

fosaprepitant (150 mg IV) pre chemotherapy and dexamethasone (8 mg<br />

p.o. daily, days 2-4) post chemotherapy were randomized to receive OLN,<br />

10 mg orally daily for three days or METO, 10 mg orally TID for three days.<br />

Patients were monitored for emesis and nausea for the 72 hours after taking<br />

OLN or METO. Eighty patients (median age 56 yrs, range 38-79; 43<br />

females; ECOG PS 0,1) consented to the protocol and all were evaluable.<br />

Results: During the 72 hour observation period, 30 <strong>of</strong> 42 (71%) patients<br />

receiving OLN had no emesis compared to 12 <strong>of</strong> 38 (32%) patients with no<br />

emesis for patients receiving METO (p�0.01). Patients without nausea (0,<br />

scale 0-10, M.D. Anderson Symptom Inventory) during the 72 hour<br />

observation period was: OLN: 67% (28 <strong>of</strong> 42); METO 24% (9 <strong>of</strong> 38)<br />

(p�0.01). There were no Grade 3 or 4 toxicities. Conclusions: OLN was<br />

significantly better than METO in the control <strong>of</strong> breakthrough emesis and<br />

nausea in patients receiving HEC.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


9065 General Poster Session (Board #41D), Sat, 8:00 AM-12:00 PM<br />

<strong>Clinical</strong> needs <strong>of</strong> patients referred early to supportive and palliative care.<br />

Presenting Author: Jung Hye Kwon, Department <strong>of</strong> Internal Medicine,<br />

Hallym University Medical Center, Hallym University College <strong>of</strong> Medicine,<br />

Seoul, South Korea<br />

Background: Palliative care is evolving from delivering care to patients at<br />

the end-<strong>of</strong>-life to those earlier in the disease trajectory. We evaluated the<br />

differences in clinical characteristics, symptoms burdens, and service<br />

utilization between traditional palliative care patients (late referrals, LR)<br />

and the new patients group who are in earlier in their course <strong>of</strong> disease<br />

(early referrals, ER). Methods: We reviewed consecutive cancer patients<br />

referred to the Supportive Care Clinic with follow up visit within 30 days<br />

between August 2008 and October 2010. Patients were divided into two<br />

groups: ER (defined as patients with expected survival � 2 years or<br />

receiving treatment for curative intent) and LR (all others). We compared<br />

clinical characteristics, symptoms and service utilization between both<br />

groups using chi-square test and Wilcoxon rank sum test. Results: 58%<br />

(695/1208) patients had a 2nd visit within 30 days. Among them, 100<br />

patients were classified as ER (14.4%) and 100/595 LR were randomly<br />

selected as the comparison group. ER patients were younger (median age<br />

54 years vs 60 years, p�0.009), more likely to have head and neck cancer<br />

(67% vs 6%, p�0.001) and to have a shorter interval between cancer<br />

diagnosis and palliative care consultation (3.8 m vs 16.2 m, �0.001). ER<br />

patients were also more likely to be CAGE positive (15% vs 4%, p�.014),<br />

referred from radiation oncology (49% vs 3%, p�0.001), referred for<br />

treatment related side effects (70% vs 9%, p�0.001), and receiving active<br />

anti-cancer treatment at the time <strong>of</strong> consultation (74% vs 48%, p�0.0002).<br />

Baseline symptoms (Edmonton Symptom Assessment Scale) were similar<br />

between ER and LR except for insomnia (1.8 vs 2, p�0.004). LR patients<br />

experienced greater improvement in the symptom distress score (-5.5 vs -3,<br />

p�0.007). Overall median number <strong>of</strong> medical visits was 24 for ER vs 10.5<br />

for LR (p�0.001); however, median visit per month was 4.3 for LR and 2.1<br />

for ER (p�0.001). Conclusions: ER was associated with different patient<br />

characteristics; patients have similar distress but different needs and<br />

outcomes as compared to traditional LR patients.<br />

9067 General Poster Session (Board #41F), Sat, 8:00 AM-12:00 PM<br />

Clostridium difficile-associated diarrhea in cancer patients treated with<br />

fidaxomicin or vancomycin. Presenting Author: Oliver A. Cornely, 1st<br />

Department <strong>of</strong> Internal Medicine, and Zentrum für Klinische Studien–ZKS<br />

Köln (BMBF 01KN070), University Hospital <strong>of</strong> Cologne, Koeln, Germany<br />

Background: Oncology patients are at risk for infections, including C<br />

difficile infection (CDI). Fidaxomicin (FDX) is a novel antibiotic highly<br />

specific for C difficile with improved sustained response compared with<br />

vancomycin (VAN). Another important clinical outcome <strong>of</strong> CDI is time to<br />

resolution <strong>of</strong> diarrhea (TTROD), corresponding to the duration <strong>of</strong> passing<br />

unformed bowel movements. Methods: 183 patients with a current diagnosis<br />

<strong>of</strong> cancer were compared to non-cancer patients from a combined<br />

population <strong>of</strong> 1105 subjects with CDI in two phase 3 trials comparing 10<br />

days <strong>of</strong> treatment with FDX (400 mg/day) or VAN (500 mg/day). Number <strong>of</strong><br />

unformed bowel movements (UBM) per 24 h was monitored. Response was<br />

considered as �3 UBM/24 h. Time to resolution <strong>of</strong> diarrhea was defined as<br />

time in hours from the first dose <strong>of</strong> study drug to the last UBM on the day<br />

preceding two days <strong>of</strong> �3 UBM/24 h. Kaplan-Meier survival analysis (K-M)<br />

used the log rank test for significance (p�0.05). Results: The response rate<br />

at end <strong>of</strong> treatment was 79.2% for 183 patients with cancer, compared to<br />

88.6% for 922 patients without cancer (p�0.001). Median TTROD was<br />

longer by 45 h in patients with cancer than those without. In the VAN<br />

treatment group, median TTROD was 65 h longer in patients with cancer vs<br />

those without and significant by K-M. In the FDX group, although median<br />

TTROD was longer by 20 h in patients with cancer vs those without, the<br />

difference was not significant by K-M. For patients with cancer, TTROD was<br />

significantly shorter during treatment with FDX than with VAN. Conclusions:<br />

Patients with cancer responded more slowly to treatment for CDI than did<br />

non-cancer patients; however, resolution <strong>of</strong> diarrhea was more rapid by 49<br />

h during treatment with FDX than with VAN.<br />

Median TTROD, h (95% CI)<br />

Subgroup<br />

No cancer Cancer K-M log rank p<br />

All patients (N�1105) 55 (53-58) 100 (71-119) �0.001<br />

Treated with FDX (N�539) 54 (48-58) 74 (54-103) 0.145<br />

Treated with VAN (N�566) 58 (54-72) 123 (78-142) �0.001<br />

Fidaxomicin Vancomycin<br />

Patients with cancer (N�183) 74 (54-103) 123 (78-142) 0.045<br />

Patient and Survivor Care<br />

583s<br />

9066 General Poster Session (Board #41E), Sat, 8:00 AM-12:00 PM<br />

Effect <strong>of</strong> complementary and alternative medicine on the survival and<br />

health-related quality <strong>of</strong> life among terminally ill cancer patients: A<br />

prospective cohort study. Presenting Author: Young Ho Yun, Seoul National<br />

University College <strong>of</strong> Medicine, Seoul, South Korea<br />

Background: Few have examined the effect <strong>of</strong> complementary and alternative<br />

medicine (CAM) use on survival and health-related quality <strong>of</strong> life<br />

(HRQOL) in cancer patients. Methods: From July 2005 to October 2006, we<br />

conducted a prospective cohort study <strong>of</strong> 481 terminally ill cancer patients<br />

at 11 university hospitals and the National Cancer Center in Korea. We<br />

assessed how the use <strong>of</strong> CAM affected HRQOL and survival. Results: In a<br />

follow-up <strong>of</strong> 481 patients and 163.8 person-years, we identified 466<br />

deceased cases. On multivariate analyses, CAM users did not have better<br />

survival than nonusers (adjusted hazard ratio (aHR), 0.91; 95% confidence<br />

interval (CI), 0.74-1.10). Among biologically based therapies, dietary<br />

supplements (aHR, 0.70; 95% CI, 0.48–1.01) and ginseng (aHR, 0.73;<br />

95% CI, 0.52–1.04) did not show significantly better survival. Among<br />

mind-body interventions, prayer showed significantly worse survival<br />

(aHR,1.56; 95% CI, 1.00–2.43). <strong>Clinical</strong>ly, CAM users reported significantly<br />

worse cognitive functioning (-11.6 v -1.3; p � 0.05) and fatigue<br />

(9.9 v -1.0; p � 0.05) than non-users. Compared with non-users in<br />

subgroup analysis, users <strong>of</strong> alternative medical treatments, prayer, vitamin<br />

supplements, mushrooms, or rice and cereal reported clinically significant<br />

worse changes in some HRQOL subscales. Conclusions: While CAM did not<br />

provide any definite survival benefit, CAM users reported clinically significant<br />

worse HRQOLs.<br />

9068 General Poster Session (Board #41G), Sat, 8:00 AM-12:00 PM<br />

Interdisciplinary palliative care in metastatic non-small-cell lung cancer.<br />

Presenting Author: Marianna Koczywas, City <strong>of</strong> Hope, Duarte, CA<br />

Background: Palliative cancer care is the integration into cancer care <strong>of</strong><br />

therapies that address multiple issues that cause suffering for patients and<br />

their families and impact their life quality. Challenges and barriers<br />

continue to hinder the integration <strong>of</strong> palliative care into comprehensive,<br />

ambulatory oncology care. This paper aims to describe how symptoms,<br />

distress, and quality <strong>of</strong> life (QOL) data from the usual care phase <strong>of</strong> a<br />

NCI-supported Program Project (P01) informed the development <strong>of</strong> an<br />

interdisciplinary palliative care intervention for patients with metastatic<br />

non-small-cell lung cancer (NSCLC). Methods: Patients receiving usual<br />

care for metastatic NSCLC were recruited into this prospective longitudinal<br />

study. A total <strong>of</strong> 130 patients with stage IV NSCLC were accrued, and 114<br />

patients had evaluable data. Upon informed consent, patients completed<br />

outcome measures that assessed physical function/cognitive status, social<br />

activities and support, symptom characteristics, psychological distress,<br />

and overall QOL. Patient-reported outcomes were completed at baseline, 6,<br />

12, and 24 weeks post-accrual. Results: Subjects were primarily female<br />

(64%), ranged in age from 40-84 years, and 39% were ethnic minorities.<br />

The majority were former smokers (66%). KPS, IADL, and Cognitive scores<br />

deteriorated significantly over time (p�.001, .009, and .042, respectively).<br />

Patients reported higher severity scores with symptoms such as<br />

dyspnea, fatigue, insomnia, lack <strong>of</strong> appetite, peripheral neuropathy, pain,<br />

dry skin, nail changes, and problems with sexual interest in all four time<br />

periods. Global Symptom Distress Index and Total Symptom score both<br />

significantly worsened at 24 weeks (p�.003 and .017, respectively).<br />

Physical Well-Being worsened significantly (p�.036), while overall QOL,<br />

spiritual well-being, and psychological distress scores remained statistically<br />

stable over time. Conclusions: Patients with metastatic NSCLC<br />

continue to experience high symptom burden and diminished physical<br />

well-being over time while receiving treatments. An interdisciplinary<br />

palliative care intervention is currently being tested to improve symptom<br />

burden and overall QOL.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


584s Patient and Survivor Care<br />

9069 General Poster Session (Board #41H), Sat, 8:00 AM-12:00 PM<br />

Attrition rates, reasons, and predictive factors in supportive/palliative<br />

oncology clinical trials at a comprehensive cancer center. Presenting<br />

Author: Isabella Claudia Glitza, University <strong>of</strong> Texas M. D. Anderson Cancer<br />

Center, Houston, TX<br />

Background: Attrition is common among supportive/palliative oncology<br />

clinical trials. Few studies have documented the reasons and predictors for<br />

dropout. We aimed to determine the rate, reasons and factors associated<br />

with attrition both before reaching the primary endpoint (PE) and the end <strong>of</strong><br />

study (EOS). Methods: We conducted a review <strong>of</strong> all prospective interventional<br />

supportive/palliative oncology trials by our department between<br />

1999-2010. Patient and study characteristics and attrition data were<br />

extracted. We determined factors associated with attrition using multivariate<br />

logistic regression analysis. Results: 15 blinded randomized trials and 3<br />

single arm trials were included. 16 <strong>of</strong> 18 studies did not reach accrual<br />

target. Baseline demographics for the 1214 patients were: median age 60<br />

(range 23-93 years), female 56%, Caucasians 69%, ECOG performance<br />

status �3 41%, gastrointestinal malignancies (23%), median fatigue<br />

7/10, appetite 5/10 and pain 4/10. Attrition rate was 26% (N�311) for PE<br />

and 44% (N�535) for EOS. Common reasons for EOS dropout were patient<br />

preference (N�93, 17%), symptom burden (N�87, 16%), death (N�45,<br />

8%) and hospital admission (N�43, 8%), and were similar for PE<br />

dropouts. No predictors were identified for PE attrition. The Table shows<br />

poor performance status, anorexia and dyspnea are associated with EOS<br />

attrition in multivariate analysis. Conclusions: We found that attrition rate<br />

was high amongsupportive/palliative oncology clinical trials, and was<br />

associated with poor function and high baseline symptom burden. These<br />

findings have implications for future study designs including eligibility<br />

criteria and sample size calculation.<br />

Factors associated with EOS attrition in multivariate analysis.<br />

Characteristics Odds ratio (95% confidence interval) p value<br />

ECOG performance status �0.001<br />

0 0.35 (0.11-1.16) 0.09<br />

1 0.26 (0.12-0.58) �0.001<br />

2 0.34 (0.16-0.72) 0.01<br />

3 0.56 (0.26-1.2) 0.14<br />

4 1.0 Ref<br />

Median ESAS (interquartile range)<br />

Appetite (per point) 1.08 (1.01-1.14) 0.02<br />

Dyspnea (per point) 1.1 (1.03-1.18) �0.001<br />

9071 General Poster Session (Board #42B), Sat, 8:00 AM-12:00 PM<br />

Biomarkers <strong>of</strong> amenorrhea and ovarian function in breast cancer survivors.<br />

Presenting Author: Kathryn Jean Ruddy, Dana-Farber Cancer Institute,<br />

Boston, MA<br />

Background: Ovarian function after treatment <strong>of</strong> early breast cancer may<br />

affect fertility, menopausal symptoms, endocrine therapy decision-making,<br />

and subsequent health and psychosocial concerns among young survivors.<br />

We aim to improve understanding <strong>of</strong> ovarian reserve in women with a history<br />

<strong>of</strong> breast cancer by comparing anti-mullerian hormone (AMH), estradiol<br />

(E2), and follicle-stimulating hormone (FSH) levels in survivors with and<br />

without amenorrhea, a surrogate for ovarian dysfunction and menopause.<br />

Methods: As part <strong>of</strong> an ongoing prospective multi-center cohort study,<br />

women diagnosed with breast cancer at age �41 have blood and surveys<br />

collected one year after diagnosis. Women who reported that they were<br />

receiving ovarian suppression or had undergone hysterectomy or bilateral<br />

oophorectomy/ovarian ablation were excluded. Amenorrhea was defined as<br />

�6 months between blood draw and last menstrual period. AMH, E2, and<br />

FSH levels were compared between those with and without amenorrhea<br />

using a Wilcoxon rank sum test with 2-sided p-values. Results: The first 199<br />

eligible women with blood and survey data at one year were included in this<br />

analysis. Median age was 37 years (range 22-40). Seventy-two(36%) had<br />

received chemotherapy alone, 75 (38%) had received chemotherapy<br />

followed by tamoxifen (TAM), 25 (13%) had received TAM alone, and 10<br />

(5%) reported TAM use but did not respond to chemotherapy items. Median<br />

AMH and E2 were lower and FSH was higher in women with amenorrhea,<br />

and these differences remained significant when those on and <strong>of</strong>f TAM were<br />

analyzed separately (Table). Conclusions: AMH, E2, and FSH all are<br />

promising biomarkers for amenorrhea and residual ovarian function in<br />

young breast cancer survivors. Future research will evaluate whether AMH,<br />

E2, and FSH at baseline and early in the survivorship period predict ovarian<br />

function later.<br />

N<br />

Median<br />

AMH<br />

AMH<br />

p value<br />

Median<br />

E2<br />

E2<br />

p value<br />

Median<br />

FSH<br />

FSH<br />

p value<br />

Total patients 199 0.06 85.0 7.91<br />

No amenorrhea 134 0.22 �0.0001 111.5 �0.0001 5.94 �0.0001<br />

Amenorrhea 65 0.01* 25.0 31.48<br />

No TAM 0.001 0.02 0.001<br />

No amenorrhea 66 0.09 81.5 5.61<br />

Amenorrhea 23 0.01* 30.0 47.09<br />

TAM �0.0001 0.001 �0.0001<br />

No amenorrhea 68 0.48 183.0 6.18<br />

Amenorrhea 42 0.01* 16.5 29.04<br />

* Values reported to be �0.02 were analyzed as 0.01<br />

9070 General Poster Session (Board #42A), Sat, 8:00 AM-12:00 PM<br />

Humoral and cellular immune response after influenza vaccination in<br />

patients with postcancer fatigue and patients with chronic fatigue syndrome.<br />

Presenting Author: Hetty Prinsen, Department <strong>of</strong> Medical Oncology,<br />

Radboud University Nijmegen Medical Centre, Nijmegen, Netherlands<br />

Background: Postcancer fatigue (PCF) is a frequently occurring problem,<br />

impairing quality <strong>of</strong> life. Patients with chronic fatigue syndrome (CFS) also<br />

suffer from severe fatigue symptoms. We hypothesized that in fatigued<br />

patients (PCF and CFS) alterations in immune response could explain<br />

fatigue symptoms. Therefore, we examined whether the humoral and/or<br />

cellular immune response after influenza vaccination differed between<br />

fatigued patients and non-fatigued individuals and between PCF and CFS<br />

patients. Methods: PCF (n�15) and CFS patients (n�22) were vaccinated<br />

against influenza. Age and gender matched non-fatigued cancer survivors<br />

(n�12) and healthy controls (n�23) were included for comparison.<br />

Antibody responses were measured at baseline and at day 21 by a<br />

hemagglutination inhibition test. T cell responses were measured at<br />

baseline and at day 7 by a lymphocyte proliferation and activation assay.<br />

Results: Both patient groups developed seroprotection rates comparable to<br />

the accompanying control groups. Functional T cell reactivity was observed<br />

in all groups. Proliferation at baseline was significantly lower in fatigued<br />

patients compared to non-fatigued individuals. A significant increase in<br />

proliferation from baseline to day 7 was observed in fatigued patients, but<br />

not in controls. At day 7, proliferation was not significantly different<br />

between fatigued patients and non-fatigued individuals. CD4�CD127-<br />

FoxP3� expression was significantly higher in PCF patients compared to<br />

non-fatigued cancer survivors. Conclusions: We observed a lower T cell<br />

proliferation at baseline in fatigued patients compared to non-fatigued<br />

individuals, suggesting a difference in the baseline state <strong>of</strong> the immune<br />

system between fatigued patients and non-fatigued individuals. Furthermore,<br />

the difference in CD4�CD127-FoxP3� expression between PCF and<br />

CFS patients suggests subtle differences in immune state between these<br />

two fatigued patient groups. However, since humoral and cellular immune<br />

responses after vaccination did not differ significantly between fatigued<br />

patients and non-fatigued individuals, vaccination <strong>of</strong> fatigued patients<br />

(PCF and CFS) can be effective.<br />

9072 General Poster Session (Board #42C), Sat, 8:00 AM-12:00 PM<br />

A randomized, double blind, placebo-controlled crossover trial <strong>of</strong> a sustained<br />

release methylphenidate in cancer-related fatigue. Presenting<br />

Author: Carmelita P. Escalante, University <strong>of</strong> Texas M. D. Anderson Cancer<br />

Center, Houston, TX<br />

Background: Cancer-related fatigue (CRF) is common and distressing. This<br />

study assessed the efficacy <strong>of</strong> OROS methylphenidate 18 mg daily (OM) vs.<br />

placebo (P) for CRF reduction. Other objectives were to compare the effects<br />

<strong>of</strong> OM vs. P on other symptoms, cognitive function, work yield, patient (pt)<br />

perceptions and preferences, and an exploratory analysis <strong>of</strong> cytokines.<br />

Methods: Initially, pts were randomly assigned (1:1) to receive OM-P or<br />

P-OM for 4 weeks (wks). Pts were crossed to the other treatment after 2<br />

wks. Assessments were done at baseline, 2 and 4 wks. Continuous and<br />

categorical variables were assessed using Wilcoxon signed rank tests and<br />

McNemar tests, respectively. The primary efficacy end point was the<br />

change <strong>of</strong> “fatigue worst” on the Brief Fatigue Inventory (BFI) at the end <strong>of</strong><br />

each 2 wk period. Results: 42 female breast cancer pts were enrolled (3<br />

ineligible, 6 unevaluable); 33 pts were studied. The mean age was 58<br />

(range, 32-79), 30% had metastasis, 82% were receiving chemotherapy<br />

(ctx), 9% hormonal therapy (ht), and 9% both ctx and ht. 94% were ECOG<br />

�1 and 52% were employed. 45% were on pain medicines and none on<br />

antidepressants. The mean baseline BFI was 5.7 (range 4.1-8.6). Fatigue<br />

worst did not differ between OM and P (p� 0.54) or in other symptoms.<br />

There was significance in WAIS-III Digit Symbol between OM and P<br />

(p�0.01), and Hopkins Verbal Learning Test with BFI interfere and BFI<br />

active (p�0.04, p�0.03, respectively). Hours (hrs) missed due to health<br />

(p�0.03) and hrs worked (p�0.04) differed in OM vs. P. At study end, pts<br />

were asked if OM improved CRF and if they wanted to stay on OM. 64% felt<br />

improved. Of these, 58% wanted to continue. There were no serious<br />

adverse events due to OM. There were differences in serum IL6 (p�0.03),<br />

IL10 (p�0.0004), and TNF� (p�0.02) among OM and P. Conclusions: Low<br />

dose OM did not show improvement in CRF on fatigue worst <strong>of</strong> BFI. Pts<br />

taking OM had better cognition and less work absences. Pts tolerated OM<br />

well and a majority felt better and wanted to continue OM. Future studies<br />

examining higher doses or longer treatment duration with OM, pt preferences<br />

and impact on productivity are necessary. Changes in serum cytokine<br />

levels should be further explored.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


9073 General Poster Session (Board #42D), Sat, 8:00 AM-12:00 PM<br />

Genetic polymorphisms <strong>of</strong> SCNA9 as predictive markers <strong>of</strong> oxaliplatininduced<br />

neuropathy. Presenting Author: Maria Sereno, Oncology Department,<br />

Hospital Infanta S<strong>of</strong>ía, Madrid, Spain<br />

Background: Oxaliplatin (OXL) is an active drug in digestive tumors.<br />

Oxaliplatin induced peripheral neuropathy (OIN) is the main dose limiting<br />

toxicity. Around 60-80% <strong>of</strong> patients developed a mild cold-induced<br />

neurotoxicity that used to disappear few days after but in 15% became a<br />

persistent neuropathy. The pathophysiology <strong>of</strong> oxaliplatin-induced neuropathy<br />

(OIN) remains unclear, preclinical studies suggest involvement <strong>of</strong><br />

voltage Na channels. SCN9A gene codifies to Na channels � subunit highly<br />

expressed in nociceptive neurons. Mutations in SCN9A are involved in<br />

alterations in neuropatic pain perception. This study tried to identify if<br />

variants in SCN9A could be associated to a higher risk to OIN. Methods:<br />

Serum were obtained from 100 patients with digestive cancer (colorectal,<br />

gastric, pancreatic and bile duct) treated with OXL (adjuvant or advanced<br />

setting) in Infanta S<strong>of</strong>ía University Hospital. No diabetes or hypotiroidism<br />

were detected. Patients were divided in two groups according to the<br />

development <strong>of</strong> OIN: Arm A 50 patients diagnosed <strong>of</strong> grade 2-3 OIN after 6<br />

courses and Arm B 50 with no OIN. The evaluation <strong>of</strong> severity <strong>of</strong> the OIN by<br />

a neurologist included a classification according to NCI-CTC and OSNS<br />

scales and conduction studies. Genomic DNA was isolated from serum and<br />

variants <strong>of</strong> SCN9A were analyzed by PCR-RFLP. The relation between<br />

SCN9A genotype and the development <strong>of</strong> severe OIN was the primary aim<br />

Results: One hundred patients were enrolled between May 2010-November<br />

2011. 56 (56%) females and 44 (44%) males; mean age was 62; mean<br />

ECOG was 1; primary tumor were colorectal 81 pts (81%), gastric 10<br />

(10%); 8 (8%) pancreto-biliar and anus 1(1%). Stage were classified in<br />

localized 65 (65%) and advanced (35%). The chemotherapy regimen<br />

included: 94 (94%) XELOX, 1 (1%) XELOX-Herceptin and 5 (5%) EOX<br />

regimen. The mean <strong>of</strong> cycles were 7 (1-14). Mean OXL dose intensity was<br />

100mg/m2.The mean PFS was 23 months. High expression <strong>of</strong> SCN9A<br />

variants were detected in patients 26/50, 52% arm A and in 5/50, 10%<br />

arm B patients, suggesting a possible relationship beetwen the development<br />

<strong>of</strong> OIN and SCN9A genotype (p�0.04). Conclusions: SCN9A polimorphysms<br />

may help to identify those patients with a higher risk to develop<br />

oxaliplatin induced neuropathy.<br />

9075 General Poster Session (Board #42F), Sat, 8:00 AM-12:00 PM<br />

Pilot study <strong>of</strong> Scrambler therapy for the treatment <strong>of</strong> chemotherapyinduced<br />

peripheral neuropathy. Presenting Author: Deirdre R. Pachman,<br />

Mayo Clinic, Rochester, MN<br />

Background: Chemotherapy-induced peripheral neuropathy (CIPN), a common<br />

dose-limiting side effect <strong>of</strong> chemotherapy, remains without known<br />

effective interventions. Preliminary data support that Scrambler therapy, a<br />

device which treats pain via non-invasive cutaneous electrostimulation, is<br />

beneficial for the treatment <strong>of</strong> CIPN. This pilot trial was performed to<br />

investigate the effect <strong>of</strong> Scrambler therapy for the treatment <strong>of</strong> CIPN.<br />

Methods: Eligible patients included those age �18 years, ECOG PS �2, life<br />

expectancy �3 months, with pain or CIPN symptoms <strong>of</strong> �1 month duration<br />

and tingling or pain �4/10 during the prior week. Patients were treated<br />

with Scrambler therapy to the affected area for up to 10 daily 30 minute<br />

sessions. Symptoms were monitored daily during therapy using a questionnaire<br />

to measure symptoms <strong>of</strong> neuropathy with a numerical analogue scale.<br />

Results: We report on the first 11 CIPN patients, enrolled between<br />

7/18/2011 and 12/12/2011, 3 men and 8 women; mean age 57 years.<br />

Patients had history <strong>of</strong> exposure to various chemotherapeutic agents and<br />

the majority had symptoms �2 years. The table portrays data at baseline, at<br />

the end <strong>of</strong> the 10 planned days <strong>of</strong> therapy, and the percent changes from<br />

baseline to the end <strong>of</strong> treatment, regarding patient reported pain, tingling,<br />

and numbness over the preceding 24 hours. There were no adverse events.<br />

Persistent benefit out to 5 weeks was seen in some patients; maturing data<br />

will be available by May 2012. Descriptive summary statistics formed the<br />

basis <strong>of</strong> data analysis. Further patients are being entered on this trial.<br />

Conclusions: Scrambler therapy appears to be beneficial in the treatment <strong>of</strong><br />

CIPN. A prospective placebo-controlled clinical trial should be performed<br />

to confirm these preliminary findings.<br />

Effect <strong>of</strong> Scrambler therapy on CIPN score.<br />

Mean baseline score Mean final score Percent change<br />

Symptom Mean Worst Mean Worst Mean Worst<br />

Pain 5.7 (2.20) 6.0 (2.10) 3.0 (2.65) 3.8 (3.06) -48% (2.65) -36% (3.54)<br />

Tingling 6.5 (1.81) 7.0 (1.84) 3.8 (2.82) 4.0 (2.65) -41% (2.34) -43% (1.95)<br />

Numbness 6.5 (1.57) 7.2 (1.78) 5.1 (2.43) 5.2 (2.48) -21% (1.75) -28% (2.14)<br />

Numbers in parentheses represent standard deviations.<br />

Patient and Survivor Care<br />

585s<br />

9074 General Poster Session (Board #42E), Sat, 8:00 AM-12:00 PM<br />

Psychological impact <strong>of</strong> exercise in women with breast cancer receiving<br />

adjuvant therapy: What is the optimal dose needed? Presenting Author:<br />

Marion Carayol, Laboratory Epsylon EA 4556 Dynamics <strong>of</strong> Human Abilities<br />

and Health Behaviors, Department <strong>of</strong> Medicine, Human, <strong>Society</strong> and Sport<br />

Sciences, University <strong>of</strong> Montpellier and St-Etienne, Montpellier, France<br />

Background: Several meta-analyses have examined the role <strong>of</strong> exercise<br />

interventions in improving psychological outcomes in cancer survivors but<br />

most did not focus on adjuvant therapy period and did not investigate the<br />

optimal dose <strong>of</strong> exercise needed. Methods: The present meta-analysis<br />

examines the impact <strong>of</strong> exercise interventions delivered at this particular<br />

period on fatigue, anxiety, depression and quality-<strong>of</strong>-life (QoL) as well as<br />

dose-response relationships between volume <strong>of</strong> prescribed exercise and<br />

these psychological outcomes. Randomized controlled trials that proposed<br />

an exercise intervention to breast cancer patients undergoing chemotherapy<br />

and/or radiotherapy were systematically identified and coded.<br />

Standardized mean differences (SMDs) <strong>of</strong> psychological outcomes were<br />

weighted by the inverse <strong>of</strong> their variances to obtain a pooled estimate using<br />

random effects model. Linear and quadratic regressions were carried out to<br />

explore dose-response relationships. Results: In total, 17 studies involving<br />

1380 participants and 20 exercise interventions were included. Intervention<br />

subjects significantly reduced their fatigue and depression levels<br />

showing pooled effect sizes (EF) and their associated 95% confidence<br />

interval (95%CI) <strong>of</strong> -0.28 [95%CI: -0.54; -0.03] and -0.28 [95%CI:<br />

-0.46; -0.09] respectively. Levels <strong>of</strong> anxiety also appeared to be reduced<br />

but pooled estimate did not reach significance (p�0.06). Significantly<br />

increased QoL was observed: EF�0.34 [95%CI: 0.07; 0.62] favouring<br />

intervention. Consistent and significant inverse associations <strong>of</strong> weekly and<br />

total volume <strong>of</strong> prescribed exercise were observed with fatigue (F test:<br />

p�0.04, R²�0.19 and p�0.009, R²�0.26 respectively) and QoL (F test:<br />

p�0.01, R²�0.14 and p�0.02, R²�0.29 respectively), implying that<br />

SMDs magnitude decreased as exercise dose increased. Conclusions:<br />

Exercise intervention enhanced fatigue, depression and QoL in breast<br />

cancer patients undergoing adjuvant therapy. Prescription <strong>of</strong> relatively low<br />

doses <strong>of</strong> exercise (�12 MET.h per week) consisting in approximately<br />

90-120 min <strong>of</strong> weekly moderate physical exercise seems more efficacious<br />

in improving fatigue and QoL than higher doses.<br />

9076 General Poster Session (Board #42G), Sat, 8:00 AM-12:00 PM<br />

Paucity <strong>of</strong> core palliative care elements available to children with cancer at<br />

end-<strong>of</strong>-life: Perspectives <strong>of</strong> bereaved parents and clinicians. Presenting<br />

Author: Alisha Kassam, The Hospital for Sick Children, Toronto, ON,<br />

Canada<br />

Background: Recognition <strong>of</strong> serious deficits in palliative care for children<br />

has prompted hospitals to develop specialized pediatric palliative care<br />

programs. However, only a minority <strong>of</strong> children who die from advanced<br />

cancer receive services from these dedicated programs. We aimed to<br />

determine which elements <strong>of</strong> palliative care are considered important<br />

according to bereaved parents and pediatric oncology clinicians and to<br />

determine availability <strong>of</strong> these elements. Methods: We administered questionnaires<br />

to 75 bereaved parents (response rate 53%) and 48 pediatric<br />

oncology clinicians (response rate 91%) at the Hospital for Sick Children in<br />

Toronto, Canada. The main outcome measures were importance ratings and<br />

availability <strong>of</strong> core elements <strong>of</strong> palliative care delivery based on the<br />

National Quality Forum clinical practice guidelines for quality palliative<br />

care. Results: Fifteen <strong>of</strong> the twenty-one core elements were highly valued by<br />

both parents and clinicians (defined as � 60% <strong>of</strong> parents and clinicians<br />

reporting the item as important). When compared to clinicians, parents<br />

gave higher importance ratings to receiving cancer-directed therapy during<br />

the last month <strong>of</strong> life (p � 0.007) and involvement <strong>of</strong> a religious or spiritual<br />

mentor (p � 0.03). Clinicians gave higher importance ratings to involvement<br />

<strong>of</strong> a social worker (p � 0.0001). Notably, <strong>of</strong> the valued elements, only<br />

three <strong>of</strong> them were available greater than 60% <strong>of</strong> the time according to<br />

clinicians and parents. Valued elements least likely to be available<br />

included a direct admission policy to hospital, involvement <strong>of</strong> a religious or<br />

spiritual mentor, sibling support and parent preparation for medical<br />

aspects surrounding death. Conclusions: Children with advanced cancer are<br />

not receiving key elements <strong>of</strong> palliative care despite both parents and<br />

clinicians recognizing them as important. Evaluation <strong>of</strong> barriers to provision<br />

<strong>of</strong> quality palliative care and strategies for overcoming them is critical.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


586s Patient and Survivor Care<br />

9077 General Poster Session (Board #42H), Sat, 8:00 AM-12:00 PM<br />

Efficacy and safety <strong>of</strong> rolapitant, a novel NK-1 receptor antagonist, for the<br />

prevention <strong>of</strong> chemotherapy-induced nausea and vomiting in subjects<br />

receiving highly emetogenic chemotherapy. Presenting Author: Luis Enrique<br />

Fein, Centro Oncologico de Rosario, Rosario, Argentina<br />

Background: Management <strong>of</strong> chemotherapy-induced nausea and vomiting<br />

(CINV) improves quality <strong>of</strong> life and increases the likelihood that patients<br />

will continue to receive appropriate treatment. The objective <strong>of</strong> this dose<br />

finding study was to evaluate rolapitant for the prevention <strong>of</strong> CINV in<br />

subjects receiving highly emetogenic chemotherapy (HEC). Methods: A<br />

phase II, double blind study in which 454 subjects receiving HEC<br />

(�70mg/m2 cisplatin-based chemotherapy) were randomized in equal<br />

fashion prior to chemotherapy to receive ondansetron � dexamethasone �<br />

either placebo or 10, 25, 100 or 200mg <strong>of</strong> rolapitant. Subjects recorded<br />

episodes <strong>of</strong> emesis, severity <strong>of</strong> nausea, and use <strong>of</strong> rescue medication(s)<br />

daily within a subject diary from Days 1 through 6 <strong>of</strong> Cycle 1. Results: The<br />

rolapitant 200mg group had significantly greater complete response rates<br />

(no emesis and no use <strong>of</strong> rescue medication) in the overall (0 to 120 hours),<br />

acute (0 to �24 hours) and delayed (�24 to 120 hours) phases compared<br />

to the placebo group (62.5% vs. 46.7%, p�0.032; 87.6% vs. 66.7%,<br />

p�0.001 and 63.6% vs. 48.9%, p�0.045, respectively). Moreover, the<br />

200mg group had significantly greater rates <strong>of</strong> no emesis and no significant<br />

nausea in the overall, acute, and delayed phases and achieved statistically<br />

significant better QoL scores (FLIE questionnaire) compared to the placebo<br />

group. Rates for no emesis and no significant nausea for the 200mg dose<br />

group in Cycles 2 to 6 continued to demonstrate superior treatment effect<br />

vs. placebo. Treatment-related adverse events were mild and included<br />

constipation, headache, fatigue and dizziness. Overall, serious adverse<br />

events (SAEs) occurred with similar incidences across all treatment groups<br />

(9% - 14%). Most common SAEs were febrile neutropenia, neutropenia,<br />

vomiting, dehydration, nausea and pneumonia and were considered related<br />

to chemotherapy or underlying cancer and not to rolapitant. Conclusions:<br />

Administration <strong>of</strong> rolapitant 200mg with ondansetron and dexamethasone<br />

is safe and effective at preventing CINV in subjects receiving HEC.<br />

9079 General Poster Session (Board #43B), Sat, 8:00 AM-12:00 PM<br />

Development <strong>of</strong> a geriatric vulnerability score (GVS) in elderly advanced<br />

ovarian cancer (AOC) patients (pts) treated in first line: A prospective<br />

GINECO trial. Presenting Author: Gilles Freyer, Oncologie Médicale, Centre<br />

Hospitalier Lyon-Sud, Lyon, France<br />

Background: Two previous prospective GINECO studies in elderly patients<br />

have underlined the prognostic value <strong>of</strong> geriatric covariates (Co) on overall<br />

survival (OS) (Freyer G., et al. Ann Oncol. 2005 and Tredan O, et al Ann<br />

Oncol 2007). Methods: This open prospective trial was designed to confirm<br />

the impact <strong>of</strong> geriatric Co including psycho-geriatric Co on OS <strong>of</strong> elderly pts<br />

(�70) with stage III-IV AOC treated in first line with 6 courses <strong>of</strong><br />

carboplatin AUC5/3weeks. Geriatric Co were tested for their impact on OS<br />

in uni- and multivariate analyses. The best fitting proportional hazard<br />

model (GVS) was developed with the inclusion <strong>of</strong> major (MaC) and minor Co<br />

(miC). Results: From 08/2007 to 01/2010, 111 pts were included in 21<br />

centres. A majority <strong>of</strong> pts displayed characteristics <strong>of</strong> vulnerability: age<br />

(median:78, range: 70-93, �80:41%), PS�2: 43%, �3 major comorbidities:<br />

27%, �4 comedications: 69%, ADL score�6: 55%, IADL score�25:<br />

75%, HADS�15:37%. A total <strong>of</strong> 74% <strong>of</strong> pts, however, completed planned<br />

chemotherapy. Gr3-4 haematological toxicity was observed in 50% <strong>of</strong> pts<br />

(thrombocytopenia [27%], anaemia [19%], and neutropenia [30%]) and<br />

gr3-4 non-haematological toxicity was fatigue (16%), anorexia (13%) and<br />

infection (10%). Median OS was 16.2 months (95%CI[14-21]). MaC were:<br />

albuminemia�35g/L; ADL score �6; IADL score �25 and miC:<br />

lymphopenia�1G/L; HADS£14. The survival score�exp(0.320*Number<br />

[MaC] �0.354*Number[miC]) was validated upon a bootstrap analysis.<br />

Using a cut<strong>of</strong>f <strong>of</strong> 3, the simplified GVS score � Number[MaC] �<br />

Number[miC] discriminated two groups with significantly different OS<br />

:11.5 vs 21.7 months; HR�2.94; p�10-4 , but also treatment completion<br />

rates: 65.4% vs 82.1%; OR�0.41; p�0.04; severe adverse events (SAE):<br />

52.7% vs 28.6%; OR�2.8; p�0.009 and unplanned hospital admissions:<br />

52.8% vs 30.3% OR�2.6; p�0.02. Conclusions: The GVS identified a<br />

group <strong>of</strong> pts at high risk <strong>of</strong> severe toxicity, early treatment stopping,<br />

unplanned hospitalization and poor outcome. GVS provides a useful tool to<br />

identify vulnerable pts in future elderly AOC trials.<br />

9078 General Poster Session (Board #43A), Sat, 8:00 AM-12:00 PM<br />

Effect <strong>of</strong> aprepitant on adherence to high-dose cisplatin-based chemotherapy.<br />

Presenting Author: Serge Makarenko, British Columbia Cancer<br />

Agency, Vancouver, BC, Canada<br />

Background: Treatment <strong>of</strong> locally advanced head and neck cancers (HNC)<br />

and gastroesophageal cancers (GEC) frequently consists <strong>of</strong> high-dose<br />

cisplatin, which is highly emetogenic. Our aims were to 1) explore the<br />

impact <strong>of</strong> aprepitant for improving adherence to cisplatin-based chemotherapy<br />

in HNC and GEC, 2) examine its effect on reducing chemotherapyinduced<br />

nausea and vomiting (CINV) and 3) determine if use <strong>of</strong> aprepitant<br />

changed after introduction <strong>of</strong> insurance coverage for this drug. Methods:<br />

Patients diagnosed with HNC or GEC in British Columbia, Canada from Jan<br />

2008 and June 2011 and prescribed high-dose cisplatin were reviewed.<br />

Using regression models that adjusted for confounders, we evaluated the<br />

relationship between aprepitant use and treatment and outcome characteristics,<br />

such as number <strong>of</strong> chemotherapy cycles, prevalence <strong>of</strong> CINV, and<br />

recurrence and survival. Results: A total <strong>of</strong> 333 patients were identified:<br />

162 HNC and 171 GEC patients <strong>of</strong> whom 80% were men, 44% were aged<br />

�/�60 years, 35% were smokers, and 42% were alcohol users. Aprepitant<br />

was prescribed in 49%, nausea and vomiting occurred in 64 and 24%,<br />

respectively, and completion <strong>of</strong> all planned cisplatin was 52%. Younger<br />

patients (55 vs 41%, p�0.01) and those with less tumor burden (64 vs<br />

38%, p�0.01) were more likely to be given aprepitant. Individuals who<br />

received aprepitant were significantly less likely to experience CINV<br />

(p�0.01). Use <strong>of</strong> aprepitant differed between HNC and GEC patients<br />

(p�0.01); however, its use did not increase when insurance coverage <strong>of</strong><br />

this agent was introduced (p�0.16). In multivariate analyses, aprepitant<br />

use was significantly associated with adherence to all planned cisplatin<br />

treatments (OR 2.33, 95% CI 1.27-4.25, p�0.01). In Cox regression,<br />

completion <strong>of</strong> all cisplatin cycles was significantly correlated with a lower<br />

risk <strong>of</strong> recurrence (HR 0.56, 95%CI 0.32-0.97 p�0.04) and a trend<br />

towards decreased death (HR 0.56, 95%CI 0.31-1.10, p�0.10).<br />

Conclusions: Aprepitant was associated with a reduction in CINV in both<br />

HNC and GEC patients and correlated with better adherence to high-dose<br />

cisplatin-based chemotherapy. Individuals who completed all planned<br />

cisplatin had improved outcomes, specifically a lower risk <strong>of</strong> recurrence<br />

from HNC and GEC.<br />

9080 General Poster Session (Board #43C), Sat, 8:00 AM-12:00 PM<br />

Correlations between depression according DSM-IV-TR (DSM) criteria,<br />

three validated scales, oncologist assessment, and clinical psychiatric<br />

interview in elderly advanced ovarian cancer (AOC) patients (pts): A<br />

GINECO study. Presenting Author: Marilène Filbet, Hospices Civils de Lyon<br />

Lyon, France<br />

Background: Depression is a major outcome in cancer pts. Clinicians<br />

typically rely on their clinical impression <strong>of</strong> depression rather than pts<br />

self-reports. Our aim was to explore the association between patientreported<br />

depression, oncologist assessment (OA) and a clinical psychiatric<br />

interview (CPI) in elderly AOC pts. Methods: This analysis was a secondary<br />

endpoint <strong>of</strong> theElderly Women AOC trial, designed to assess the impact <strong>of</strong><br />

geriatric covariates, notably depression, on survival in pts over 70 receiving<br />

6 courses <strong>of</strong> carboplatin. Depression was assessed using the Geriatric<br />

Depression Scale-30 (GDS; cut <strong>of</strong>f score <strong>of</strong> 10/30), the Hospital Anxiety<br />

Depression Scale (HADS; cut <strong>of</strong>f score <strong>of</strong> 15/42), the distress thermometer<br />

(DT; cut <strong>of</strong>f score <strong>of</strong> 4/10) and OA (yes/no). CPI was conducted by<br />

psychologists within 10 days after inclusion. The interview guide for CPI<br />

(yes/no) was constructed and adapted from three validated scale: GDS,<br />

Hamilton Depression Rating Scale Hamilton (HDRS), Montgomery Asberg<br />

Depression Rating Scale (MADRS) and the DSM criteria. DSM was<br />

considered as the gold standard. Results: Out <strong>of</strong> 111 pts, 100 (90.1 %)<br />

completed all the assessment (OA, GDS, HADS, DT, CPI). Patients<br />

characteristics were: mean age 78, performance status �2: 48 (55%).<br />

Thirty six pts (36%) were identified as depressed by the CPI versus 17<br />

(17%) by OA, 32 (32%) by the GDS, 36 (36%) by the HADS, 58 (58%) by<br />

the DT and 16 (16%) according to DSM. We found a significant correlation<br />

between DSM and GDS (r�0.58; p�0.001), DSM and CPI (r�0.53;<br />

p�0.001). We did not find any significant correlation between DSM and OA<br />

(r�-0.05;p�0.733), DSM and DT (r�-0.07;p�0.583), and between DSM<br />

and HADS (r�0.127;p�0.258). Identification according to OA (yes/no)<br />

resulted in 87% false negatives and 18% false positives rates. The best<br />

sensitivity and specificity as a screening tool was found for GDS, 94% and<br />

80% respectively. Conclusions: The use <strong>of</strong> validated tools such as GDS and<br />

a collaboration between psychologists and oncologists are warranted to<br />

better identify emotional disorders in elderly women with AOC.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


9081 General Poster Session (Board #43D), Sat, 8:00 AM-12:00 PM<br />

Inflammatory markers and symptom burden in patients with multiple<br />

myeloma undergoing autologous stem cell transplantation. Presenting<br />

Author: Loretta A. Williams, University <strong>of</strong> Texas M. D. Anderson Cancer<br />

Center, Houston, TX<br />

Background: We explored the association between activation <strong>of</strong> inflammatory<br />

networks and development <strong>of</strong> severe symptoms during mobilization<br />

and the acute phase <strong>of</strong> autologous hematopoietic stem cell transplantation<br />

(ASCT) for multiple myeloma (MM). Methods: From mobilization through<br />

the first 3 months after ASCT, multiple symptoms were measured repeatedly<br />

via the myeloma module <strong>of</strong> the M. D. Anderson Symptom Inventory<br />

(MDASI-MM). Serum levels <strong>of</strong> interleukin (IL)-6, IL-10, tumor necrosis<br />

factor (TNF)-a, soluble TNF receptors 1 and 2 (sTNF-R1, sTNF-R2), IL-1<br />

receptor antagonist (IL-1RA), vascular endothelial growth factor (VEGF),<br />

macrophage inflammatory protein-1 (MIP-1a), monocyte chemoattractant<br />

protein (MCP)-1 and C-reactive protein (CRP) were collected up to 11 times<br />

per patient and assayed by Luminex. Ordinal regression modeling was used<br />

to analyze repeated-measures data. Results: Among50 MM patients, the<br />

most severe symptoms reported by werefatigue, pain, muscle weakness,<br />

disturbed sleep, drowsiness, numbness, poor appetite, and bone aches.<br />

Symptoms worsened rapidly from conditioning therapy and peaked at WBC<br />

nadir. Over time, controlling for age, sex, and MM stage, increased serum<br />

IL-6 (P�.0007) and MCP-1 (P�.0005) were significantly associated with<br />

worsening <strong>of</strong> the most severe symptoms; MIP-1a (P�.005) and VEGF<br />

(P�.002) were inversely associated with these symptoms. Increased CRP<br />

was significantly associated with worsening pain (P�.01), fatigue (P�.007),<br />

and bone aches (P�.006). Conclusions: This longitudinal study indicates a<br />

strong association between dynamic changes in circulating inflammatory<br />

molecules and the most severe symptoms in MM patients during the acute<br />

phase <strong>of</strong> ASCT. This observation, similar to IL-6-related multisymptom<br />

development around WBC nadir <strong>of</strong> allogeneic SCT (Wang et al, 2008),<br />

provides evidence <strong>of</strong> potential inflammation-induced behavioral changes in<br />

humans. Testing <strong>of</strong> anti-inflammatory interventions is warranted to further<br />

confirm the role <strong>of</strong> inflammation in the development <strong>of</strong> symptoms and<br />

identify effective mechanism-driven symptom management during ASCT.<br />

9083 General Poster Session (Board #43F), Sat, 8:00 AM-12:00 PM<br />

Longitudinal relationship between inflammatory markers and patientreported<br />

symptom severity during induction therapy for multiple myeloma.<br />

Presenting Author: Mary H. Sailors, University <strong>of</strong> Texas M. D. Anderson<br />

Cancer Center, Houston, TX<br />

Background: Multiple myeloma (MM) patients undergoing induction therapy<br />

experience both disease- and therapy-related symptoms. We investigated<br />

the association between the trajectory <strong>of</strong> symptom severity and changes in<br />

levels <strong>of</strong> inflammatory markers. Methods: MM patients (N�62) rated<br />

symptoms via the M. D. Anderson Symptom Inventory (MDASI) twice a<br />

week during induction therapy. Patients contributed serum samples before<br />

the start <strong>of</strong> every chemotherapy cycle. A panel <strong>of</strong> pro- and antiinflammatory<br />

cytokines and markers was evaluated by Luminex. Ordinal<br />

regression analyses were used to describe the relationship between<br />

cytokines and symptom outcomes across time. These analyses were<br />

adjusted for patient and clinical factors (age, sex, diabetes diagnosis,<br />

anemia, BMI, comorbidity, staging, ECOG PS, prior treatment status,<br />

tumor response, opioid use, and chemo regimen). Results: Bortezomibbased<br />

induction therapy was received by 89% <strong>of</strong> the sample. Fatigue was<br />

persistently the most severe symptom during induction therapy, followed by<br />

disturbed sleep, muscle weakness, pain, drowsiness, and bone aches.<br />

Numbness, which is representative <strong>of</strong> chemotherapy-induced peripheral<br />

neuropathy, significantly worsened from baseline (p�0.01). We observed<br />

significant longitudinal associations between sIL-1R1 and distress and<br />

sadness (both p�0.02); between sIL-6R and disturbed sleep (p�0.001),<br />

poor appetite (p�0.04), and sore mouth (p�0.006). IL-6 was significantly<br />

associated with pain, fatigue, nausea, and sore mouth (all p�.05). A<br />

negative association between sTNF-RII and pain, sleep, distress, remembering,<br />

poor appetite, and nausea (all p�0.05) was also observed. MCP-1 was<br />

positively associated with numbness (p�0.04); while MIP-1� was negatively<br />

associated with sleep, numbness, constipation, poor attention (all<br />

p�0.01), and bone aches (p�0.0006). IL-10 was negatively associated<br />

with mood interference (p�0.04). Conclusions: Frequent assessment can<br />

document the longitudinal course <strong>of</strong> multiple symptoms during induction<br />

and provides opportunity to evaluate systemic inflammation as a potential<br />

source <strong>of</strong> symptom burden during induction therapy for MM.<br />

Patient and Survivor Care<br />

587s<br />

9082 General Poster Session (Board #43E), Sat, 8:00 AM-12:00 PM<br />

Baseline subclinical sensory deficits and the development <strong>of</strong> pain and<br />

numbness in patients with multiple myeloma (MM) being treated with<br />

chemotherapy. Presenting Author: Elisabeth G. Vichaya, University <strong>of</strong><br />

Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: Chemotherapy-induced peripheral neuropathy (CIPN) is a<br />

dose-limiting toxicity experienced by patients with MM. Patients may<br />

develop painful and non-painful (e.g., numbness) symptoms that impair<br />

function and <strong>of</strong>ten persist after therapy is terminated. This study examined<br />

the predictive value <strong>of</strong> baseline subclinical sensory deficits on the development<br />

<strong>of</strong> treatment-related pain and numbness. Methods: Patients (N�56)<br />

who had two or fewer cycles <strong>of</strong> induction therapy and no obvious<br />

neuropathy were assessed for sensory deficits using quantitative sensory<br />

testing (QST). Patients reported the severity <strong>of</strong> pain and numbness (on a<br />

0-10 scale) at least weekly via the MD Anderson Symptom Inventory during<br />

induction (up to 16 weeks); 15 <strong>of</strong> 56 patients provided data for up to 16<br />

additional weeks <strong>of</strong> maintenance therapy. These values correlate to patient<br />

reported pain and numbness in the hands/feet. Based on data collected<br />

from healthy controls, patients were classified as being with or without a<br />

sensory deficit at baseline on each QST domain. We fit linear mixed models<br />

for pain and numbness with each sensory deficit and time, controlling for<br />

cumulative cycles, diabetes, age, and treatment agent. Data is shown as<br />

mean � standard deviation. Results: Patients who showed baseline deficits<br />

in sharpness detection (20%) reported significantly lower levels <strong>of</strong> pain<br />

(1.2�1.9 vs 3.1�2.9, p�.004) and numbness (0.4�0.8 vs 2.4�2.6,<br />

p�.001) during induction therapy and less numbness (0.0 vs 3.5�2.8,<br />

p�.001) during maintenance therapy. Further, those who showed deficits<br />

in warmth detection (40%) reported higher levels <strong>of</strong> pain (5.2�2.8 vs<br />

1.7�2.3, p�.001) and numbness (5.8�2.2 vs 1.6�1.8, p�.001) during<br />

maintenance therapy. Finally, those with lower skin temperature (47%)<br />

reported higher levels <strong>of</strong> pain (4.5�2.3 vs 1.7�2.9, p�.005) during<br />

maintenance therapy. Conclusions: Our results suggest that subclinical<br />

sensory deficits may be useful in determining a patient’s risk for developing<br />

CIPN prior to initiation <strong>of</strong> induction therapy. This information could be used<br />

to provide patients with more-personalized chemotherapy plans that take<br />

into account their neuropathy risk.<br />

9084 General Poster Session (Board #43G), Sat, 8:00 AM-12:00 PM<br />

Randomized trial <strong>of</strong> a physical activity intervention in patients with<br />

metastatic breast cancer. Presenting Author: Jennifer A. Ligibel, Dana-<br />

Farber Cancer Institute, Boston, MA<br />

Background: Physical activity (PA) interventions improve fitness, functional<br />

capacity, and quality <strong>of</strong> life in patients with early-stage breast cancer.<br />

There are almost no data regarding the feasibility or potential benefits <strong>of</strong> PA<br />

in women with metastatic breast cancer (MBC). Our study evaluated the<br />

impact <strong>of</strong> a moderate-intensity PA intervention upon functional measures<br />

in patients with MBC. Methods: <strong>Part</strong>icipants were randomized 1:1 to a<br />

16-week PA intervention or usual care control group. Eligibility included<br />

diagnosis <strong>of</strong> MBC, ECOG 0-1, and no active brain metastases. The<br />

intervention consisted <strong>of</strong> 4 weekly meetings with an exercise physiologist,<br />

followed by monthly in-person and weekly telephone-based meetings. The<br />

PA goal was 150 minutes per week. Baseline and 16-week evaluation<br />

included: modified Bruce treadmill test, 7-Day Physical Activity Recall<br />

Interview, and EORTC QLQ C-30 questionnaire. Results: 101 women<br />

enrolled between 9/06 and 3/11. Median age was 49 years, median time<br />

since diagnosis <strong>of</strong> MBC was 1.1 years, 48% <strong>of</strong> participants were on<br />

hormonal therapy and 42% on chemotherapy/biologic therapy. 69%<br />

completed all study measures (an additional 9% all but the final treadmill<br />

test). Attrition was higher in the intervention arm (30% vs. 16%, p�0.15).<br />

Outcome measures are shown in table below. Both groups experienced<br />

improvements in treadmill times. Intervention participants also reported<br />

improvements in physical functioning and increased minutes <strong>of</strong> weekly PA,<br />

with a trend toward significant differences between groups. Conclusions:<br />

Women with MBC participating in a PA intervention experienced consistent,<br />

but non-significant, trends toward improvements in functional measures.<br />

Our sample size was small, limiting power to evaluate the potential<br />

benefits <strong>of</strong> PA in women with MBC. Additional work is warranted,<br />

particularly since PA interventions appear feasible in this patient population.<br />

Baseline Change over 16 weeks<br />

Exercise Control Exercise Control p value<br />

Treadmill (min)* 5.6 � 1.8 5.3 � 1.7 .61 � .19** .37 � .16** .35<br />

Physical functioning* 84.2 � 14.4 83.9 � 13.2 4.79 � 2.40** .93 � 2.07 .23<br />

Exercise min/wk* 86.3 � 217.8 74.7 � 80.6 71.8 � 185.6** 53.8 � 183.7 .14<br />

*Data presented as means � SD. **Within-group comparison �0.05.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


588s Patient and Survivor Care<br />

9085 General Poster Session (Board #43H), Sat, 8:00 AM-12:00 PM<br />

A randomized trial to determine if vitamin B6 can prevent hand and foot<br />

syndrome in cancer patients treated with capecitabine chemotherapy.<br />

Presenting Author: Tareq Braik, John H. Stroger Jr. Hospital <strong>of</strong> Cook<br />

County, Chicago, IL<br />

Background: Capecitabine is an oral fluoropyrimidine that is used to treat<br />

various malignancies. Hand and Foot Syndrome (HFS) is a dose limiting<br />

toxicity <strong>of</strong> capecitabine and can limit the use <strong>of</strong> this agent in some patients.<br />

Some investigators have observed that pyridoxine (vitamin B6) can ameliorate<br />

HFS caused by capecitabine. We designed a prospective trial to<br />

determine if pyridoxine can prevent HFS in patients receiving capecitabine.<br />

Methods: In our double-blind, placebo controlled trial, we randomly<br />

assigned eligible patients treated with capecitabine to receive either daily<br />

pyridoxine 100 mg or placebo along with their capecitabine-containing<br />

chemotherapy regimen. Patients were observed during the first four cycles<br />

<strong>of</strong> capecitabine treatment. The primary end point was the incidence and<br />

grade <strong>of</strong> HFS that occurred in both arms. Results: Between 2008 and<br />

2011, 77 patients were randomly assigned to receive either pyridoxine<br />

(n�38) or placebo (n�39). Both arms were matched in baseline characteristics.<br />

The median age was 53.5 years. The ethnic composition <strong>of</strong> the study<br />

population was African <strong>American</strong> 53%, Caucasian 22%, Hispanic 18%,<br />

and Asian 7%. The daily doses <strong>of</strong> capecitabine were: 2000 mg/m2 (69<br />

pts), 1650 mg/m2 (5 pts), 1400 mg/m2 (2 pts) and 1000 mg/m2 (1 pt).<br />

Dosages <strong>of</strong> capecitabine were equally matched between the two arms <strong>of</strong> the<br />

study. HFS occurred after a median <strong>of</strong> 2 chemotherapy cycles in both<br />

groups. HFS developed in 10/38 (26%) patients in the pyridoxine group<br />

and in 8/39 (20%) patients in the placebo group (p�0.547). Therefore, the<br />

risk <strong>of</strong> HFS was 6 percentage points higher in pyridoxine group (95% CI for<br />

difference: -13 percentage points to �25 percentage points). Given our<br />

study results, we can be confident <strong>of</strong> excluding a true benefit from<br />

pyridoxine larger than 13 percentage points. No difference in HFS grades<br />

was observed. Conclusions: Prophylactic pyridoxine (vitamin B6), given<br />

concomitantly with capecitabine-containing chemotherapy, was not effective<br />

for the prevention <strong>of</strong> HFS.<br />

Grades HFS I HFS II HFS III No HFS<br />

Pyridoxine 4 (11%) 2 (5%) 4 (11%) 28 (73%)<br />

Placebo 2 (5%) 3 (8%) 3 (8%) 31 (79%)<br />

9087 General Poster Session (Board #44B), Sat, 8:00 AM-12:00 PM<br />

Usefulness <strong>of</strong> medical oncologists’ estimates <strong>of</strong> survival time in people with<br />

advanced cancer. Presenting Author: Belinda E. Kiely, NHMRC <strong>Clinical</strong><br />

Trials Centre, University <strong>of</strong> Sydney, Sydney, Australia<br />

Background: We sought to determine the calibration, precision, prognostic<br />

significance and suitability <strong>of</strong> estimated survival time (EST) as a basis for<br />

estimating and explaining prognosis in advanced cancer. Methods: Medical<br />

oncologists recorded their EST as the “median survival <strong>of</strong> a group <strong>of</strong><br />

identical patients” in patients with advanced cancer and a life expectancy<br />

�3 months recruited to a randomized trial <strong>of</strong> sertraline (Lancet Oncol<br />

2007; 8: 603). Calibration, precision and suitability were defined by the<br />

proportions <strong>of</strong> patients whose observed survival times (OST) were bounded<br />

by simple multiples <strong>of</strong> their EST (based on our previous studies), i.e. 50%<br />

expected to live longer (or shorter) than their EST; 30% expected to live<br />

from 0.75 to 1.33 times their EST (arbitrary criterion for precision); 50%<br />

expected to live from half to double their EST (range for typical scenario);<br />

10% expected to live �3 times their EST (best case scenario), or �¼ <strong>of</strong><br />

their EST (worst case scenario). Results: Characteristics <strong>of</strong> the 114 patients<br />

were: median age 63 years, Karn<strong>of</strong>sky performance status (KPS) �70 in<br />

25%, and a median <strong>of</strong> 8.5 months since diagnosis <strong>of</strong> advanced cancer.<br />

Primary cancer sites included breast (18%), colorectal (16%), lung (15%),<br />

prostate (12%) and ovary (10%). Median survival was 10.6 months after a<br />

median follow-up <strong>of</strong> 14 months and 68 deaths. EST were well-calibrated:<br />

54% <strong>of</strong> patients lived longer than their EST and 46% lived shorter than<br />

their EST. EST were imprecise (21% within 0.75 to 1.33 times OST) but<br />

equally likely to be over-optimistic (34% �1.33 x OST) or over-pessimistic<br />

(39% �0.75 x OST). 6% <strong>of</strong> patients lived �¼ <strong>of</strong> their EST; 62% lived from<br />

half to double their EST and 9% lived �3 times their EST. Independently<br />

significant predictors <strong>of</strong> OST in a multivariable Cox model included EST<br />

(HR�0.92, p�0.004), dry mouth (HR�5.07, p�0.0001), alkaline phosphatase<br />

�101U/L (HR�2.80, p�0.0002), KPS �70 (HR�2.30,<br />

p�0.007), prostate primary (HR�0.23, p�0.002), and steroid use<br />

(HR�2.35, p�0.02). Conclusions: Medicaloncologists’ EST were wellcalibrated,<br />

imprecise, independently associated with OST, and useful for<br />

estimating and explaining best case, worse case, and typical scenarios for<br />

survival time in patients with advanced cancer.<br />

9086 General Poster Session (Board #44A), Sat, 8:00 AM-12:00 PM<br />

In-hospital and long-term outcomes in patients with malignancy undergoing<br />

percutaneous endoscopic gastrostomy (PEG). Presenting Author: Emily<br />

Keung, Department <strong>of</strong> Surgery, Brigham and Women’s Hospital, Boston,<br />

MA<br />

Background: PEG is widely performed in cancer patients as a means <strong>of</strong><br />

providing nutrition or palliation. Although considered safe, PEG-associated<br />

outcomes in these patients remain poorly described. We examined the<br />

safety and benefits <strong>of</strong> PEG placement in this patient population. Methods: A<br />

five year retrospective review <strong>of</strong> all patients with malignancy (excluding<br />

head/neck, thoracic) who underwent attempted PEG at our institution was<br />

performed. Results: PEG was placed in 187 <strong>of</strong> 189 patients; 64 with<br />

hematologic malignancy (H-M), 125 non-hematologic malignancy (NH-M).<br />

Median age at time <strong>of</strong> PEG was 60.8 years. Indication was nutritional<br />

support (100%) in H-M, enteral access (59.2%) and management <strong>of</strong><br />

obstructive symptoms (38.4%) in NH-M. A minority were able to return<br />

home (27.5%), discontinue parenteral nutrition (22%), advance diet for<br />

nutrition (24.6%) or comfort (17.1%) with the rest remaining NPO. Overall<br />

rates <strong>of</strong> PEG-related major (aspiration, tube dislodgement/leakage, bleeding,<br />

visceral injury, respiratory failure after procedure, cardiac arrest) and<br />

minor (superficial infection, ileus) complications were 21.4% and 11.3%,<br />

respectively, with higher rates in H-M (34.4% and 20.3% vs 14.4% and<br />

6.4%). All cause in-hospital mortality was high: 31.3% H-M, 13.6%<br />

NH-M. Median time from PEG placement to death was 54 days. Leading<br />

cause <strong>of</strong> death differed by malignancy: respiratory failure and sepsis in H-M<br />

(31% and 21%), primary malignancy in NH-M (75%). Overall one year<br />

mortality was 56%. Code status was changed in 21% <strong>of</strong> patients after PEG<br />

(“Full Code” to “Do Not Resuscitate/Do No Intubate” or “Comfort Measures<br />

Only”). Multivariate/subgroup analyses will be presented at time <strong>of</strong> meeting.<br />

Conclusions: PEG placement in this study population was associated<br />

with significant procedure-related complications and failed to achieve TPN<br />

independence or advancement <strong>of</strong> diet. Nearly 25% <strong>of</strong> patients declined<br />

aggressive resuscitation strategies after undergoing surgery for PEG. Thus,<br />

higher burden <strong>of</strong> counseling is needed to carefully weigh the risk and<br />

benefit <strong>of</strong> PEG placement in these patients. Further studies are needed to<br />

elucidate the factors affecting the decision process and patient selection.<br />

9088 General Poster Session (Board #44C), Sat, 8:00 AM-12:00 PM<br />

Risk <strong>of</strong> rash with nilotinib: A systematic review <strong>of</strong> the literature and<br />

meta-analysis. Presenting Author: Aaron Mark Drucker, University <strong>of</strong><br />

Toronto, Toronto, ON, Canada<br />

Background: Nilotinib is indicated for the treatment <strong>of</strong> chronic myelogenous<br />

leukemia (CML). The reported incidence and risk <strong>of</strong> rash from this<br />

medication vary widely and have been inconsistently reported in published<br />

trials. Therefore we conducted a systematic review and meta-analysis <strong>of</strong> the<br />

literature to determine the incidence and risk <strong>of</strong> developing rash. Methods:<br />

Relevant studies were identified from the PubMed database (1998-2012),<br />

abstracts presented at ASCO and ASH Conferences (2004-2011) and Web<br />

<strong>of</strong> Science database (1998-2012). Eligible studies were limited to prospective<br />

Phase II-III clinical trials in which patients received nilotinib at doses<br />

<strong>of</strong> either 300 mg or 400 mg twice daily. Incidence, relative risk (RR), and<br />

95% confidence intervals (CI) were calculated using random-effects or<br />

fixed-effects models based on heterogeneity <strong>of</strong> included studies. Results:<br />

Data from a total <strong>of</strong> 3,186 patients receiving nilotinib in 16 clinical trials<br />

were available for analysis. The overall incidence <strong>of</strong> all-grade and highgrade<br />

(grade �3) rash were 33.1% (95% CI: 27.7-39.1) and 2.6% (95%<br />

CI: 2.1-3.4), respectively. Incidence <strong>of</strong> all-grade rash for patients with<br />

CML, gastrointestinal stromal tumor (GIST) and systemic mastocytosis<br />

were 33.2% (95% CI: 27.2-39.9), 25.7% (95% CI: 14.0-42.5) and<br />

25.0% (95% CI: 15.7-37.4), respectively. Nilotinib was associated with<br />

increased risk <strong>of</strong> all-grade rash (RR�2.891, 95% CI: 2.079-4.020;<br />

P�0.001) when compared to patients treated with imatinib. Risk <strong>of</strong><br />

high-grade rash was increased compared to imatinib (RR�1.823, 95% CI:<br />

0.670-4.957), but this was not statistically significant (P�0.24).<br />

Conclusions: Patients with hematologic malignancies and GIST who are<br />

treated with nilotinib are at significant risk for developing a rash. Further<br />

studies for characterization, prevention and treatment <strong>of</strong> this untoward<br />

toxicity are needed in order to maintain patients’ quality <strong>of</strong> life and<br />

minimize the need for dose modification, which may impact clinical<br />

outcome.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


9089 General Poster Session (Board #44D), Sat, 8:00 AM-12:00 PM<br />

The chemotherapy-induced peripheral neuropathy (CIPN) outcome measure<br />

standardization (CI-PeriNomS) study: From consensus to validity and<br />

reliability in CIPN assessment. Presenting Author: Guido Cavaletti, University<br />

<strong>of</strong> Milan-Bicocca, Monza, Italy<br />

Background: Chemotherapy-Induced Peripheral Neuropathy (CIPN) is a<br />

debilitating and dose limiting complication <strong>of</strong> cancer treatment. However,<br />

the impact <strong>of</strong> CIPN has never been studied in a clinimetric manner, in its<br />

impact on quality <strong>of</strong> life. This study was performed to select appropriate<br />

outcome measures and to establish, for the first time with a clinimetric<br />

approach, their validity and reproducibility. Methods: After literature review<br />

and a consensus meeting, face/content validity were obtained for the<br />

following scales: the National Cancer Institute Common-Toxicity-Criteria<br />

(NCI-CTC), the Total Neuropathy Score (TNSc), the modified INCAT<br />

sensory sumscore (mISS), the European Organization for Research and<br />

Treatment <strong>of</strong> Cancer (EORTC) QLQ-C30 and CIPN20 quality <strong>of</strong> life<br />

measures. 281 patients with stable CIPN were examined through an EU/US<br />

collaboration involving 20 oncology/neurology centers. For each scale the<br />

inter- and intra-rater agreement was evaluated by means <strong>of</strong> weighted<br />

K-Cohen coefficients and 95% confidence intervals, when analyzing<br />

qualitative ordinal scales and by means <strong>of</strong> Spearman’s rank correlation<br />

coefficient and t-test when analyzing quantitative ordinal scales. The<br />

adopted weights for the estimate <strong>of</strong> K-Cohen coefficients were the Fleiss-<br />

Cohen weights. For validity purposes, the Kruskal-Wallis equality-<strong>of</strong>populations<br />

rank test was performed relating the mISS and TNSc to the<br />

NCI-CTC grades. Results: Proper, although slightly different, inter-/intraobserver<br />

scores (i.e. r � 0.7) were obtained for the TNSc, mISS, and<br />

NCI-CTC sensory/motor subscales. Test-retest values were also high for the<br />

EORTC QLQ-C30 and for the recently developed and never formally tested<br />

CIPN20. Acceptable validity scores were obtained through correlation<br />

between the the mISS and TNSc to the NCI-CTC scores (p values ranging<br />

from 0.04 to � 0.001). Conclusions: Proper validity and reliability scores<br />

were demonstrated for the set <strong>of</strong> selected outcome measures in CIPN.<br />

These results will allow to base new trials on a solid methodological<br />

background, although future studies are warranted to investigate the<br />

responsiveness issues.<br />

9091 General Poster Session (Board #44F), Sat, 8:00 AM-12:00 PM<br />

Gender difference in vulnerability and geriatric syndromes among elderly<br />

cancer survivors. Presenting Author: Lin Fan, University <strong>of</strong> Rochester<br />

Medical Center, Rochester, NY<br />

Background: Few studies have evaluated the gender difference <strong>of</strong> vulnerability<br />

and geriatric syndromes (GS) among cancer survivors. Methods: Using<br />

2003 Medicare Current Beneficiary Survey, we applied multivariable<br />

logistic regression to evaluate the association <strong>of</strong> gender with vulnerability<br />

and GS among cancer survivors. Vulnerability is measured by Vulnerable<br />

Elders Survey-13 (VES-13), which is calculated from age, self-rated health<br />

status, difficulty with physical activities and functional activities. Vulnerability<br />

is defined by a VES-13 score <strong>of</strong> 3 or higher. 8 common geriatric<br />

syndromes were assessed: sight trouble, hearing trouble, nutrition problem,<br />

incontinence, falls, depression, memory loss and osteoporosis. Cancer<br />

survivors are defined as patients with cancer diagnosed more than one year<br />

ago. Results: Among the 1,883 cancer survivors, 749 (40.0%) were male;<br />

1,134 (60.0%) were female. Female survivors had a higher prevalence <strong>of</strong><br />

vulnerability (47.8% vs 35.7%), and GS (65.5% vs 44.6%) than male<br />

survivors. Mean GS number was 1.19 for female survivors, while 0.77 for<br />

male survivors (p�0.001). More specifically, female survivors had significantly<br />

higher prevalence <strong>of</strong> sight trouble (7.73% vs 5.0%), incontinence<br />

(14.3% vs 7.1%), falls (28.9% vs 22.1%), depression (13.8% vs 7.7%)<br />

and osteoporosis (38.2% vs 6.7%). After adjusting for confounders, female<br />

survivors were more likely to be vulnerable (adjusted OR (AOR): 1.62; CI:<br />

1.21-2.17; p�0.001); have a geriatric syndrome (AOR: 2.39; CI: 1.91-<br />

3.00; p�0.001); have sight trouble (AOR: 1.70; CI: 1.00-2.87; p�0.049);<br />

have incontinence (AOR: 2.48; CI: 1.67-3.69; p�0.001); have depression<br />

(AOR: 1.87; CI: 1.29-2.71; p�0.001); and have osteoporosis (AOR: 8.48;<br />

CI: 6.07-11.84; p�0.001). Conclusions: Female cancer survivors experience<br />

a higher prevalence <strong>of</strong> vulnerability and overall geriatric syndromes.<br />

Patient and Survivor Care<br />

589s<br />

9090 General Poster Session (Board #44E), Sat, 8:00 AM-12:00 PM<br />

Incidence, characteristics, and associations <strong>of</strong> oxaliplatin-induced peripheral<br />

neuropathy in colorectal cancer patients: Results <strong>of</strong> a prospective,<br />

multicenter, international study. Presenting Author: Andreas Argyriou,<br />

Department <strong>of</strong> Neurology, “Saint Andrew’s” State General Hospital <strong>of</strong><br />

Patras, Patras, Greece<br />

Background: We sought to trace the incidence and severity <strong>of</strong> oxaliplatininduced<br />

peripheral neuropathy (OXLIPN) and to determine its clinical<br />

pattern. Among other associations, we also specifically aimed at testing<br />

whether the degree <strong>of</strong> acute neuropathy is clinically related to the<br />

development and severity <strong>of</strong> chronic OXLIPN. Methods: 170 patients (mean<br />

age 64y), scheduled to be treated with either FOLFOX or XELOX for<br />

metastatic colorectal cancer were studied. Patients were prospectively<br />

monitored at baseline and followed-up during chemotherapy in four<br />

European sites. The motor/neurosensory NCI-CTCv3 criteria and the<br />

clinical version <strong>of</strong> the Total Neuropathy Score were applied to clinically<br />

grade the severity <strong>of</strong> OXLIPN. Nerve conduction studies were also longitudinally<br />

performed. Results: Evidence <strong>of</strong> acute OXLIPN was disclosed in<br />

146/170 patients (85.9%). The vast majority <strong>of</strong> patients manifested<br />

cold-induced perioral (95.2%) or pharyngolaryngeal dysesthesias (91.8%).<br />

Other uncommon symptoms, such as jaw spasm were also present. Severe<br />

acute OXLIPN, requiring prolongation <strong>of</strong> OXL infusion from 2 to 4-6 hours<br />

was evident in 32/146 patients (21.9%). Overall, 123/170 patients<br />

(72.4%) experienced chronic OXLIPN. Severities were grade I: 39 (22.9%);<br />

grade II: 52 (30.6%); grade III: 33 patients (19.4%). Worst severities were<br />

related to cumulative oxaliplatin dose (r:0.269;p�0.001). Follow-up<br />

assessments revealed significant decrease in all sensory action potentials<br />

examined. The severity <strong>of</strong> the acute OXLIPN was significantly correlated<br />

with both the development (r:0.450;p�0.001) and degree <strong>of</strong> the chronic<br />

form (r:0.703;p�0.001). Conclusions: Most patients treated with oxaliplatin-based<br />

chemotherapy would manifest OXLIPN, mainly <strong>of</strong> moderate<br />

degree. The severity <strong>of</strong> the acute syndrome appears to clinically correlate<br />

with the degree <strong>of</strong> the chronic form <strong>of</strong> OXLIPN. Our data suggest that acute<br />

phenomena, related to axonal hyperexcitability, could contribute to the<br />

development <strong>of</strong> peripheral neuropathy; thus it could be advisable to test<br />

agents against acute OXLIPN in order to verify their effects on the chronic<br />

form.<br />

9092 General Poster Session (Board #44G), Sat, 8:00 AM-12:00 PM<br />

Risk <strong>of</strong> skin rash with the proteasome inhibitor bortezomib: Updated<br />

systematic review and meta-analysis. Presenting Author: Emily Christine<br />

Case, Columbia University, College <strong>of</strong> Physicians and Surgeons, New York,<br />

NY<br />

Background: Rash is a common, adverse event to the novel proteasome<br />

inhibitor bortezomib (Velcade). Indicated for the treatment <strong>of</strong> multiple<br />

myeloma and mantle cell lymphoma, bortezomib is the first proteasome<br />

inhibitor approved by regulatory agencies. Because the incidence <strong>of</strong><br />

bortezomib-induced skin rash varies widely in published manuscripts, we<br />

performed a systematic literature review and meta-analysis to determine<br />

the incidence and overall risk. Methods: We searched PubMed and Web <strong>of</strong><br />

Science databases and abstracts presented at the <strong>American</strong> <strong>Society</strong> <strong>of</strong><br />

<strong>Clinical</strong> Oncology and The <strong>American</strong> <strong>Society</strong> <strong>of</strong> Hematology annual meetings<br />

(1998 to July 2011) to identify relevant clinical studies. Eligible<br />

studies included prospective clinical phase II and phase III trials, with data<br />

on the incidence <strong>of</strong> rash in patients taking 1.3mg/m2 , 1.5mg/m2 ,or1.6<br />

mg/m2 <strong>of</strong> bortezomib intravenously either weekly or twice weekly. The<br />

incidence <strong>of</strong> rash and relative risk (RR) were calculated using randomeffects<br />

or fixed-effects model, depending on the heterogeneity <strong>of</strong> included<br />

studies. Results: A total <strong>of</strong> 2,616 patients with various hematologic and<br />

solid malignancies from 35 clinical trials were included for analysis.<br />

Among patients receiving twice weekly bortezomib, the summary incidence<br />

<strong>of</strong> all-grade and high-grade rash were 18.8 % (95% CI: 14.9% to 23.5%)<br />

and 3.6 % (95% CI: 2.3% to 5.7%), respectively. We found no significant<br />

increase in all grade rash incidence with higher doses <strong>of</strong> bortezomib: 19.3<br />

% (95% CI: 15% to 24.5%) and 20.8% (95% CI: 11.6% to 34.4%) for<br />

doses <strong>of</strong> 1.3 mg/m2 and 1.5 mg/m2 , respectively. In addition, bortezomib<br />

was associated with an increased risk in both all grade (RR: 19.70, 95% CI:<br />

8.73 to 44.44, p�0.001) and high-grade rash (RR: 5.35, 95% CI: 2.16 to<br />

13.29, p�0.001), compared to controls. Weekly bortezomib is associated<br />

with lower risk <strong>of</strong> rash compared to twice weekly dosing (incidence 3.9%<br />

versus 18.8%, p�0.001). Conclusions: Bortezomib is associated with a<br />

significant risk <strong>of</strong> developing rash with a higher risk among patients<br />

receiving twice weekly dosage. Management <strong>of</strong> rash to bortezomib is<br />

critical to prevent a negative effect on quality <strong>of</strong> life and dose modifications,<br />

both <strong>of</strong> which affect clinical outcome.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


590s Patient and Survivor Care<br />

9094 General Poster Session (Board #44H), Sat, 8:00 AM-12:00 PM<br />

Feasibility <strong>of</strong> screening hospitalized cancer patients for palliative care.<br />

Presenting Author: Paul A. Glare, Memorial Sloan-Kettering Cancer Center,<br />

New York, NY<br />

Background: The National Comprehensive Cancer Network Palliative Care<br />

(PC) Guideline recommends screening all oncology patients for PC needs,<br />

and to call a PC consult when referral criteria are met. There are no data on<br />

the feasibility or impact <strong>of</strong> this approach. The aim <strong>of</strong> this pilot study was to<br />

assess the feasibility <strong>of</strong> PC screening in patients admitted to a comprehensive<br />

cancer center (CCC). Methods: Design: Observational study. From<br />

11/1/10 to 1/31/11, floor nurses screened all patients the day after<br />

admission under the two teams (“Team A” and “Team B”) <strong>of</strong> the<br />

Gastrointestinal Oncology Service at Memorial Sloan-Kettering Cancer<br />

Center. Team A patients were also evaluated by the referral criteria.<br />

Endpoints: Patients screened ‘positive’ if they had advanced disease and<br />

any <strong>of</strong> the clinical situations nominated in the Guideline. The referral<br />

criteria triggered PC consults in Team A patients; clinical judgment<br />

triggered Team B consults. Outcomes: Screening rates, nursing satisfaction<br />

survey, clinical and operational metrics. Results: Ninety percent (229<br />

<strong>of</strong> 254) <strong>of</strong> admissions were screened. Both Teams’ patients were seriously<br />

ill (see Table), and it was no surprise that 63% (145 <strong>of</strong> 229) screened<br />

positive. Survey respondents (response rate 50%) rated screening as<br />

simple, quick and helpful, although nurses scored the extent <strong>of</strong> disease<br />

wrong in 16%. Sixty eight percent (55 <strong>of</strong> 780) <strong>of</strong> Team A patients who<br />

screened positive met the referral criteria. This generated more consults on<br />

Team A, but the effect on key outcomes was not significant (n.s.).<br />

Conclusions: Screening for PC was feasible in this setting, but is a<br />

challenging concept in terms <strong>of</strong> reliability, validity and timing. The value to<br />

a CCC <strong>of</strong> increasing PC access via referral criteria needs evaluation in<br />

well-designed trials.<br />

Screened patients Team A (n�113) Team B (n�116) p value<br />

Status on admission<br />

Time since diagnosis (months) 13 15 n.s.<br />

Advanced disease 92% 97% n.s.<br />

Best supportive care 36% 39% n.s.<br />

Died within next 3 months 33% 45% n.s.<br />

Pain score > 7/10 33% 34% n.s.<br />

Screen positive 71% 56% 0.028<br />

Outcomes<br />

Consults 42% 15% �0.0001<br />

Pain score > 7/10, day 4* 13% 38% n.s<br />

Median length <strong>of</strong> stay (days) 4 4 n.s.<br />

Died in hospital 4% 7% n.s.<br />

Hospice referral 19% 21% n.s.<br />


9098 General Poster Session (Board #45D), Sat, 8:00 AM-12:00 PM<br />

Medical oncologist’s commitment in the end <strong>of</strong> life (EoL) care <strong>of</strong> cancer<br />

patients: The caregiver’s perspective. Presenting Author: Romeo Bascioni,<br />

Ospedale Murri, Fermo, Italy<br />

Background: Optimizing the impact <strong>of</strong> EoL care on cancer patients (pts) and<br />

their caregivers should be a primary goal <strong>of</strong> an oncology unit. In this study<br />

we evaluated satisfaction <strong>of</strong> family caregivers when the medical oncology<br />

team assisted pts until death. Methods: Two oncology units were reorganized<br />

to ensure continuity <strong>of</strong> care; oncologists trained in palliative care<br />

medicine assisted pts until EoL. The model assumes that the medical<br />

oncologist (MO) is the physician in charge throughout the entire disease<br />

trajectory. Relatives <strong>of</strong> pts assisted at home or at an inpatient hospice<br />

underwent a semistructured phone interview conducted by a psychologist<br />

or a social worker � 1 month after pts’ death. Satisfaction was evaluated for<br />

symptoms control, communication, psychological support, overall quality<br />

<strong>of</strong> care and continuity <strong>of</strong> therapeutic relationship with the MO. A final<br />

open-ended question was included for any additional comment. Satisfaction<br />

was measured using a five-point Likert scale ranging from very<br />

dissatisfied to very satisfied and converted to a 0-to-100 scale. Results:<br />

Relatives <strong>of</strong> 65 pts were contacted, 55 accepted the interview (27 spouses,<br />

22 sons, 5 in-laws, 1 parent); 50/55 were the leader caregiver. Patients<br />

were followed at home (41) or at an inpatient hospice (14), for 1-24 wks.<br />

Satisfaction mean scores were: symptoms control 76/100, communication<br />

85, psychological support 82 and overall quality <strong>of</strong> care 87; a specific<br />

question on the relevance <strong>of</strong> the MO in EoL care produced a score <strong>of</strong> 87,<br />

with no negative or neutral responses recorded for this item. Of note, a<br />

common perception among caregivers was the appreciation <strong>of</strong> the MO’s<br />

commitment during EoL in addition to the technical quality <strong>of</strong> the<br />

intervention. The overall satisfaction score was higher than in our previous<br />

study in which a continuity <strong>of</strong> care model was not formally adopted, with a<br />

score improvement from 55 to 87/100. Conclusions: A care pathway where<br />

the MO is involved in EoL management <strong>of</strong> cancer pts improved satisfaction<br />

<strong>of</strong> caregivers.When a longstanding and trusting relationship has been<br />

established, the pts-MO connection should not be lost to prevent feelings <strong>of</strong><br />

abandonment.<br />

9100 General Poster Session (Board #45F), Sat, 8:00 AM-12:00 PM<br />

Sexual functioning in young women with breast cancer. Presenting Author:<br />

Shoshana Rosenberg, Harvard School <strong>of</strong> Public Health, Boston, MA<br />

Background: Sexual dysfunction is a known complication <strong>of</strong> adjuvant<br />

therapy for breast cancer and an important determinant <strong>of</strong> quality <strong>of</strong> life.<br />

Little is known about the frequency and magnitude <strong>of</strong> this problem among<br />

very young women with breast cancer during the year following diagnosis.<br />

Methods: 298 sexually-active women enrolled in an ongoing multi-center<br />

cohort study with Stage 0-III breast cancer at or before age 40 were<br />

included in this analysis. Treatment data was self-reported on a survey<br />

mailed to participants at enrollment. Sexual functioning was assessed<br />

using the sexual interest and dysfunction subscales from the Cancer<br />

Rehabilitation Evaluation System (CARES). Scores range from 0-4, with<br />

higher scores indicative <strong>of</strong> poorer function. The survey included a measure<br />

<strong>of</strong> anxiety and depression (Hospital Anxiety and Depression Scale), <strong>of</strong> body<br />

image (CARES) and <strong>of</strong> physical and menopausal symptoms (Breast Cancer<br />

Prevention Trial Symptom Checklist). Mean differences in CARES scores<br />

between treatment groups (chemotherapy vs. none; hormone therapy vs.<br />

none; lumpectomy vs. mastectomy vs. mastectomy � reconstruction vs. no<br />

surgery; radiation vs. none) were compared using T-tests and ANOVA.<br />

Multiple regression models were fit to assess symptoms thought to be<br />

mediators <strong>of</strong> the treatment-sexual functioning association. Results: Mean<br />

age at diagnosis was 36 (range: 22-40) years and mean time from diagnosis<br />

to survey completion was 5 months (range: 1-16). Mean CARES sexual<br />

interest and dysfunction scores were higher in women who received<br />

chemotherapy compared to those who did not (p�0.0001). In the<br />

multivariate analysis, chemotherapy was no longer associated with decreased<br />

sexual interest or function. Anxiety, depression, musculoskeletal<br />

pain, and poorer body image were predictive <strong>of</strong> both decreased sexual<br />

interest and function. Vaginal pain symptoms were associated with greater<br />

dysfunction, while unhappiness with appearance was associated with<br />

decreased interest. Conclusions: Young women who receive chemotherapy<br />

are at risk for problems related to sexual functioning early in the survivorship<br />

period. This effect appears to be mediated through several physiologic<br />

and psychologic mechanisms, underscoring the need for interventions that<br />

target both.<br />

Patient and Survivor Care<br />

591s<br />

9099 General Poster Session (Board #45E), Sat, 8:00 AM-12:00 PM<br />

Defining mild, moderate, and severe fatigue in cancer patients and<br />

survivors: E2Z02, a trial <strong>of</strong> the Eastern Cooperative Oncology Group.<br />

Presenting Author: Xin Shelley Wang, University <strong>of</strong> Texas M. D. Anderson<br />

Cancer Center, Houston, TX<br />

Background: Although mild, moderate and severe categories have been<br />

used in clinical guidelines for fatigue management in cancer patients, the<br />

optimal cutpoints on a 0-10 scale for delineating these categories have not<br />

been replicated. Methods: A multicenter ECOG study (E2Z02) enrolled<br />

breast, lung, prostate, or colorectal cancer patients with any treatment<br />

status. Fatigue and symptom interference were measured on the 0-10<br />

numerical rating scale <strong>of</strong> the M. D. Anderson Symptom Inventory (MDASI).<br />

The optimal boundaries for categorizing fatigue severity were determined<br />

by the largest F ratios from MANOVA (Serlin’s criteria, 1995). Logistic<br />

regression with robust standard errors was used to identify risk factors for<br />

moderate/severe fatigue for cancer survivors (defined as patients with no<br />

evidence <strong>of</strong> disease and receiving no cancer treatment). Results: The<br />

optimal cutpoints that identified 3 distinct levels <strong>of</strong> fatigue severity for the<br />

2341 patients were: ratings <strong>of</strong> 1-3 as mild, 4-6 as moderate, and 7-10 as<br />

severe. Known-group validity for these cutpoints was established by<br />

significant differences <strong>of</strong> fatigue severity by ECOG performance status and<br />

patient-reported quality <strong>of</strong> life (all P�0.001). Using these cutpoints, 45%<br />

(983/2177) <strong>of</strong> patients undergoing active therapy had moderate/severe<br />

fatigue, with significant more mild fatigue in breast and colorectal cancer<br />

patients, while more severe fatigue in lung cancer patients (p�.001).<br />

Among cancer survivors, 29% (150/515) had moderate/severe fatigue<br />

(breast 31%, colorectal 27%, prostate 22%, lung 33%). Younger age<br />

(OR�0.97, 95% CI�0.95-0.99) and poor performance status (OR�4.21,<br />

95% CI�2.36-7.51) were associated with more moderate/severe fatigue in<br />

cancer survivors. Survivor time was also associated with moderate/severe<br />

fatigue in breast and colorectal cancer survivors (��5yrs vs. �5yrs:<br />

OR�0.23, P�0.01 for breast, OR�9.3, P�0.03 for colorectal).<br />

Conclusions: This multicenter study confirmed the standard cutpoints for<br />

fatigue severity used in NCCN fatigue management guidelines. It also<br />

provides a pr<strong>of</strong>ile <strong>of</strong> moderate to severe fatigue prevalence for actively<br />

treated cancer patients and for cancer survivors.<br />

9101 General Poster Session (Board #45G), Sat, 8:00 AM-12:00 PM<br />

Early outpatient palliative care in patients with metastatic NSCLC: A<br />

qualitative study. Presenting Author: Jaclyn Yoong, Massachusetts General<br />

Hospital Cancer Center, Boston, MA<br />

Background: Early outpatient palliative care (PC) is an emerging practice<br />

and its key components have not been defined. We conducted a qualitative<br />

analysis <strong>of</strong> data from a randomized controlled trial which demonstrated<br />

improved quality <strong>of</strong> life, mood and survival in outpatients who received<br />

early PC integrated with standard oncologic care versus standard oncologic<br />

care alone. Our objectives were to 1) identify the content and key<br />

components <strong>of</strong> early PC clinic visits, 2) explore timing <strong>of</strong> key components,<br />

and 3) compare the content <strong>of</strong> PC and oncology visit notes at the critical<br />

time points <strong>of</strong> clinical deterioration and radiographic disease progression.<br />

Methods: We chose a random selection <strong>of</strong> 20 patients with newly diagnosed<br />

metastatic NSCLC who received early PC who survived within 4 time<br />

frames: less than 3 months (n�5), 3 to 6 months (n�5), 6 to 12 months<br />

(n�5) and 12 to 24 months (n�5). We performed content analysis on PC<br />

and oncology electronic health record notes <strong>of</strong> these patients using NVivo 9<br />

s<strong>of</strong>tware. Results: Addressing symptoms and coping ability were the most<br />

prevalent components <strong>of</strong> the PC clinic visits. The content <strong>of</strong> initial visits<br />

focused on building relationships and rapport with patients and their<br />

families and illness understanding. Discussions about prognostic awareness<br />

occurred during earlier visits while goals <strong>of</strong> care and hospice<br />

discussions occurred in later visits. The frequency <strong>of</strong> addressing these<br />

components was similar regardless <strong>of</strong> patients’ length <strong>of</strong> life. Comparing PC<br />

and oncology visits around critical time points, both included discussions<br />

about current illness status and goals <strong>of</strong> care; however PC visits emphasized<br />

symptoms, coping, and implications <strong>of</strong> anti-cancer treatments, while<br />

oncology visits focused on medical discussions about treatment plans.<br />

Conclusions: Early PC clinic visits emphasize managing symptoms, strengthening<br />

coping, and cultivating illness understanding and prognostic awareness<br />

in a structured, responsive and time-sensitive model. Around critical<br />

clinical time points PC and oncology visits have distinct features that<br />

suggest a key role for PC involvement while enabling oncologists to focus on<br />

anti-cancer management.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


592s Patient and Survivor Care<br />

9102 General Poster Session (Board #45H), Sat, 8:00 AM-12:00 PM<br />

<strong>Clinical</strong> trial participation as part <strong>of</strong> end-<strong>of</strong>-life (EOL) cancer care:<br />

Associations with medical care near death and bereaved caregivers’ mental<br />

health. Presenting Author: Andrea Catherine Phelps, Dana-Farber Cancer<br />

Institute, Boston, MA<br />

Background: <strong>Clinical</strong> trial participation is necessary to improve existing<br />

therapies, encouraged by national guidelines, and common among advanced<br />

cancer patients. The relationships between trial participation and<br />

important EOL outcomes such as aggressive care are unknown. Methods:<br />

Coping with Cancer Study, an NCI-funded multicenter prospective cohort<br />

study <strong>of</strong> advanced cancer patients and their caregivers, enrolled September<br />

2002 – February 2008. Patients were interviewed at baseline, and clinical<br />

trial participation was documented by chart review. Patients were followed<br />

to death, (median 4.4 months from baseline). Medical care and quality <strong>of</strong><br />

life (QOL) in the last week <strong>of</strong> life was assessed by caregiver interview and<br />

chart review. Caregiver interview 6 months post-bereavement assessed<br />

QOL and mental health (Structured <strong>Clinical</strong> Interview for the DSM-IV). The<br />

primary outcome was aggressive EOL care (ventilation, resuscitation, or<br />

chemotherapy in the last week <strong>of</strong> life). Secondary outcomes were bereaved<br />

caregivers’ mental health and QOL. Propensity-score weighting balanced<br />

patient characteristics (e.g. clinical variables, EOL preferences) that<br />

differed by trial participation. Propensity-score weighted regression models<br />

estimated the effect <strong>of</strong> trial participation on outcomes. Results: Of 246<br />

patients followed to death with non-missing propensity scores, 27 were<br />

clinical trial participants. In propensity-score weighted analyses, trial<br />

participation was significantly associated with aggressive EOL care [29.6%<br />

v 9.1%; adjusted OR (AOR), 12.14; 95% CI, 3.65-40.36], ICU admission,<br />

mechanical ventilation, chemotherapy, and a trend toward inferior QOL<br />

near death (p � 0.069). Of 180 matched caregivers, trial participation<br />

predicted less mental illness [AOR, 0.15; 95% CI 0.04-0.57], major<br />

depression [AOR, 0.25; 95% CI, 0.08-0.80], but was unassociated with<br />

QOL (p � 0.15) in adjusted analyses. Conclusions: <strong>Clinical</strong> trial participation<br />

is associated with increased risk <strong>of</strong> aggressive EOL care for advanced<br />

cancer patients, but better mental health for bereaved caregivers.<br />

9104 General Poster Session (Board #46B), Sat, 8:00 AM-12:00 PM<br />

Comparative patient-centered outcomes (health state and adverse sexual<br />

symptoms) between adjuvant brachytherapy versus no adjuvant brachytherapy<br />

in early-stage endometrial cancer. Presenting Author: Shari Damast,<br />

Yale University School <strong>of</strong> Medicine, New Haven, CT<br />

Background: We performed a cross-sectional analysis <strong>of</strong> survivors <strong>of</strong> stage I<br />

endometrial cancer (EC) to examine the relationship between receipt <strong>of</strong><br />

adjuvant intra-vaginal radiation therapy (IVRT), health state (HS), and<br />

sexual functioning (SF). Methods: Survivors were administered anonymous<br />

questionnaires containing demographic and treatment-related items, the<br />

EQ5D to measure current HS, and the Female Sexual Function Index (FSFI)<br />

to measure current SF. All were disease-free and had undergone simple<br />

hysterectomy (SH) with or without adjuvant IVRT at least one year prior to<br />

questionnaire completion. None had received chemotherapy or hormonal<br />

therapy. The study was IRB-approved under a waiver <strong>of</strong> consent. The<br />

primary endpoint was a comparison <strong>of</strong> EQ5D-health states and FSFI scores<br />

between survivors who had received SH alone and those who had received<br />

SH and adjuvant IVRT. Fisher’s exact test was used to compare IVRT and<br />

no-IVRT groups and multivariate regression was applied to find associations<br />

between factors and the outcome measures. Results: 136 (66.3%) participants<br />

had undergone SH alone and 69 (33.7%) had undergone SH and<br />

adjuvant IVRT. The SH alone group was younger; otherwise, the two groups<br />

were balanced with respect to ethnicity, marital status, education level,<br />

medical co-morbidities (diabetes, hypertension, anxiety), type <strong>of</strong> SH<br />

(laparotomy vs minimally invasive surgery-MIS), and baseline sexual<br />

activity. In the IVRT and no-IVRT groups, the median EQ5D-health states<br />

were 88 (range 40-100) and 85 (range 10-100) respectively, and the<br />

median FSFI scores were 14.1 (range 1.2-35.4) and 16.5 (range 1.2-34.8)<br />

respectively. Controlling for age and type <strong>of</strong> SH, receipt <strong>of</strong> IVRT was<br />

associated with better HS (p�0.018) and not associated with poorer SF<br />

(p�0.1399). Receipt <strong>of</strong> laparotomy (vs MIS) was associated with poorer<br />

HS and worse SF (p�0.0156 and p�0.0115, respectively), and its<br />

detrimental effect on FSFI score was more pronounced in the IVRT<br />

compared to no-IVRT group (p�0.0486). Conclusions: Compared to SH<br />

alone, receipt <strong>of</strong> adjuvant IVRT was generally not associated with poorer SF<br />

or HS.<br />

9103 General Poster Session (Board #46A), Sat, 8:00 AM-12:00 PM<br />

Changes in patient-reported outcomes in women with breast cancer in a<br />

multicenter double-blind randomized controlled trial assessing the effect<br />

<strong>of</strong> acupuncture in reducing aromatase inhibitor-induced musculoskeletal<br />

symptoms (AIMSS). Presenting Author: Ting Bao, University <strong>of</strong> Maryland<br />

Greenebaum Cancer Center, Baltimore, MD<br />

Background: Aromatase Inhibitors (AIs) have been associated with worsening<br />

<strong>of</strong> patient related outcomes (PROs) such as AIMSS, menopausal<br />

symptoms and depression. Acupuncture has been reported to alleviate<br />

such symptoms. We hypothesized that real acupuncture (RA) would<br />

improve PROs more than sham acupuncture (SA). Methods: We collected<br />

PROs at baseline, 4, 8, and 12 weeks (wks), from women enrolled in a<br />

multi-center double blind RCT designed to assess the effect <strong>of</strong> acupuncture<br />

in reducing PROs. Patients were randomized to 8 wkly RA or SA. PROs were<br />

measured by the revised National Surgical Adjuvant Breast and Bowel<br />

Project (NSABP) menopausal symptoms questionnaire, Center for Epidemiological<br />

Studies Depression Scale (CESD), Hospital Anxiety and Depression<br />

Scale (HADS), Pittsburgh Sleep Quality Index (PSQI), Hot Flash Daily<br />

Diary, Hot Flash Related Daily Interference Scale (HFRDI) and EuroQoL<br />

survey. We measured estrogen and cytokines concentrations at baseline<br />

and wk 8. We used Wilcoxon rank sum and signed-rank tests to make<br />

comparisons between and within group, respectively. Results: We included<br />

23 patients from RA and 24 from SA arms in the intent-to-treat analysis.<br />

We have previously reported no significant difference in reduction <strong>of</strong> AIMSS<br />

between two arms. RA caused reduction <strong>of</strong> CESD scores compared to SA<br />

(median: -2 vs 0, p � 0.057). When compared to baseline, there were<br />

statistically significant improvements at wk 8 in hot flash severity score<br />

(p�0.006), hot flash frequency (p�0.011), HFRDI (p�0.014) and NSABP<br />

menopausal symptoms (p�0.022) scores in RA arm; for EuroQoL<br />

(p�0.022), HFRDI (p�0.043) and NSABP menopausal symptoms<br />

(p�0.005) scores in SA arm. The majority <strong>of</strong> patients’ estradiol concentrations<br />

were undetectable at baseline and wk 8. Changes in other time points,<br />

data and analysis <strong>of</strong> cytokines changes will be presented at the meeting.<br />

Conclusions: Real and sham acupuncture were both associated with<br />

improvement in PROs in breast cancer patients taking AIs. We detected no<br />

significant difference in the change <strong>of</strong> PROs between real and sham<br />

acupuncture, except for CESD.<br />

9105 General Poster Session (Board #46C), Sat, 8:00 AM-12:00 PM<br />

A randomized trial <strong>of</strong> a cardiopulmonary resuscitation (CPR) video (V) in<br />

advance care planning (ACP) for progressive hepatopancreaticobiliary<br />

cancer patients. Presenting Author: Andrew S. Epstein, Mount Sinai, New<br />

York, NY<br />

Background: CPR is an important advance directive topic in patients with<br />

progressive cancer; however such discussions are challenging. Previous<br />

work by Volandes et al suggests that V images help, however whether V<br />

educational information about CPR engenders broader ACP discourse is<br />

unknown. Methods: A randomized trial <strong>of</strong> an educational V or narrative (N)<br />

in patients with progressive hepatopancreaticobiliary cancers was conducted.<br />

The V and N were 3 minutes long and contained an identical<br />

description <strong>of</strong> CPR. Primary endpoint: ACP documentation 1-month<br />

post-test (either a formal advance directive or a documented ACP discussion),<br />

80% powered with 0.01 type-I error rate for estimated advance<br />

directive completion rates <strong>of</strong> 70% V arm vs. 25% N arm with a sample <strong>of</strong><br />

56 subjects. Secondary endpoints (exploratory): impressions <strong>of</strong> the study<br />

information; pre and post knowledge <strong>of</strong> and preferences for CPR and<br />

mechanical ventilation; and follow-up longitudinally (to death or 6 months<br />

post-test). Results: 56 subjects consented and analyzed. 1-month post-test<br />

ACP documentation was not significantly higher in the V arm (12/30,<br />

40.0%) vs. the N arm (4/26, 15.4%); Fisher’s exact test, OR � 3.583<br />

[95% CI: 0.886 – 17.937], p � 0.07. Knowledge increased in both arms<br />

after the intervention. Preferences for CPR changed in V arm but not in N<br />

arm; for mechanical ventilation, there was no change in either arm. The<br />

majority <strong>of</strong> subjects in both arms reported the information as helpful,<br />

comfortable to discuss, and recommended to others. Longitudinally,<br />

outcomes were similar between V & N arms, respectively: % advance<br />

directives completed (63% & 58%); median days from test to advance<br />

directive (60 & 72) and advance directive to death (21 & 20); median ACP<br />

documentation after 1 month (0 & 1); median hospital admissions (1 & 1),<br />

days <strong>of</strong> stay (5 & 7), ICU stays (0 & 3) and CPR/mechanical ventilation (1 &<br />

3). 52% <strong>of</strong> all subjects died by study end, the majority in hospice care.<br />

Conclusions: This first randomized trial addressing downstream ACP effects<br />

<strong>of</strong> V vs. N decision tools demonstrated a trend towards more ACP<br />

documentation in V patients. Further research into these types <strong>of</strong> educational<br />

media is needed.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


9106 General Poster Session (Board #46D), Sat, 8:00 AM-12:00 PM<br />

Understanding the symptom experience <strong>of</strong> breast cancer outpatients: The<br />

validity and utility <strong>of</strong> the M.D. Anderson Symptom Inventory (MDASI).<br />

Presenting Author: Tito R. Mendoza, University <strong>of</strong> Texas M. D. Anderson<br />

Cancer Center, Houston, TX<br />

Background: Breast cancer and its treatments can produce multiple<br />

symptoms that cause distress and impair function. The MDASI, a patientreported<br />

outcome measure <strong>of</strong> symptoms (sxs) and functional interference<br />

has been validated in general oncology and has the potential to inform<br />

symptom experience and guide treatment specific to breast cancer patients<br />

(pts). Methods: The Eastern Cooperative Oncology Group (ECOG) conducted<br />

the Symptom Outcomes and Practice Patterns (SOAPP) study at academic<br />

and community medical oncology clinics and included pts with breast<br />

cancer <strong>of</strong> all stages and phases <strong>of</strong> care. At baseline and 4 weeks, pts<br />

completed the MDASI. Symptom experiences and psychometric properties<br />

<strong>of</strong> the MDASI in breast cancer pts (n � 1544) were analyzed. Results: The<br />

median age was 58 years, and the race/ethnicity included 11% black and<br />

8% hispanic. 5% had ECOG performance status (PS) �2. The 5 most<br />

prevalent moderate/severe sxs reported at baseline were fatigue (31%),<br />

disturbed sleep (27%), drowsiness (21%), hair loss (22%) and dry mouth<br />

(19%); moderate/severe skin rash (6%) and vomiting (4%) were the least<br />

prevalent. At follow-up, about 1/3 <strong>of</strong> patients had moderate/severe fatigue<br />

and 19% had moderate/severe pain and distress. Internal consistency and<br />

test-retest reliability were good, with Cronbach alphas <strong>of</strong> �0.85 and<br />

intraclass correlations <strong>of</strong> �0.76 for all subscales. Among those whose PS<br />

was stable or improving, the change scores in sx severity improved most for<br />

sleep disturbance, numbness/tingling, and difficulty remembering things.<br />

Significantly higher scores and moderately large effects for the severity<br />

scale were reported by pts with poorer PS (ES�.61) and those with<br />

local/regional/metastatic disease (ES�.67). Results were similar for the<br />

interference scale. Pts whose quality <strong>of</strong> life (QOL) declined showed greater<br />

increase in severity (1.1 vs .07, p�.001) from baseline to follow-up than<br />

pts whose QOL was unchanged, demonstrating sensitivity to change.<br />

Conclusions: Breast cancer pts have significant sx burden despite wellpreserved<br />

PS. The MDASI is a valid, reliable, and sensitive sx assessment<br />

method for research and patient care in breast cancer outpatients.<br />

9108 General Poster Session (Board #46F), Sat, 8:00 AM-12:00 PM<br />

Comparing three validated methods <strong>of</strong> patient self-reported fatigue in a<br />

prospective randomized clinical trial. Presenting Author: Katherine Sams,<br />

Wake Forest University, Winston Salem, NC<br />

Background: Fatigue can be measured with different validated assessment<br />

instruments in symptom management trials. Methods: Between 2004-<br />

2009, the Wake Forest CCOP Research Base protocol 97202 randomized<br />

236 women receiving adjuvant chemotherapy for newly diagnosed breast<br />

cancer to Coenzyme Q10 supplementation vs placebo. The primary<br />

endpoint was change in self-reported fatigue. Patients (pt) were assessed at<br />

baseline, 8, 16, and 24 weeks (wk) with 3 instruments: 1) Pr<strong>of</strong>ile <strong>of</strong> Mood<br />

States – Fatigue (POMS; 7 questions; scored 0-4); 2) Functional Assessment<br />

<strong>of</strong> Cancer Treatment – Fatigue (FACIT; 13 questions; scored 0-4);<br />

and 3) Visual Analog Scale (VAS; one scale; scored 0-10). For comparison,<br />

each instrument was rescaled from 0 to 100; higher numbers indicate<br />

worse fatigue. Results: CoQ10 did not significantly affect fatigue (Lesser G<br />

et al, ASCO Proc., 2010). All 3 measures demonstrated an increase in<br />

fatigue after chemotherapy initiation at 8&16wkswith trend towards<br />

baseline levels at 24 wks. The fatigue measures were highly correlated: r �<br />

0.8 for all pairwise associations at all times. However, their scores varied.<br />

The Table shows mean rescaled scores for pt below/above the median<br />

fatigue level, calculated by taking mean <strong>of</strong> the three scores averaged across<br />

the four time periods. In general, POMS tended to give the lowest and VAS<br />

the highest scores, but differences between POMS and FACIT and FACIT<br />

and VAS depended on mean fatigue level. For pt experiencing lower<br />

(�median) fatigue, FACIT and VAS scores were similar, while POMS scores<br />

were significantly lower. For higher (�median) fatigue, POMS and FACIT<br />

scores were similar, while VAS was significantly higher. Conclusions: While<br />

POMS, FACIT, and VAS scores were highly correlated, their scale scores<br />

varied depending on the level <strong>of</strong> fatigue experienced by the pt. Fatigue<br />

assessment methods in clinical trials should be selected carefully as they<br />

do not always give equivalent results. Supported by NCI/DCP grant U10<br />

CA81851.<br />

Means (standard deviations) by average level <strong>of</strong> fatigue across all time<br />

points.<br />

Instrument < Median fatigue (N�116) > Median fatigue (N�116)<br />

POMS 10.8 (7.6) 41.6 (17.9)<br />

FACIT 14.1 (9.0) 41.4 (14.3)<br />

VAS-F 15.3 (9.8) 48.2 (14.9)<br />

Patient and Survivor Care<br />

593s<br />

9107 General Poster Session (Board #46E), Sat, 8:00 AM-12:00 PM<br />

A randomized trial evaluating the integration <strong>of</strong> online questionnaires into<br />

follow-up (FU) care for early-stage breast cancer (ESBC). Presenting<br />

Author: Alyse Wheelock, University <strong>of</strong> California, San Francisco Helen<br />

Diller Family Comprehensive Cancer Center, San Francisco, CA<br />

Background: Large numbers <strong>of</strong> cancer survivors have led to the need for FU<br />

care addressing lasting effects <strong>of</strong> BC and its treatment. We conducted a<br />

trial evaluating formats <strong>of</strong> FU care to determine if integration <strong>of</strong> remote<br />

electronic questionnaire FU provides for timely symptom management and<br />

more efficient care than routine clinic visits alone. Methods: Patients (pts)<br />

with ESBC were randomized to either usual care (UC, frequency <strong>of</strong> visits<br />

determined by oncology providers) with completion <strong>of</strong> an optional online<br />

health and symptom questionnaire (Q) before each clinic visit, or to<br />

SIS.NET FU care, in which pts were scheduled for up to 3 routine oncology<br />

related clinic visits over 18 mos and completed the online Q every 3 mos.<br />

Qs were reviewed by a survivorship nurse practitioner (NP) with pt phone<br />

contact for symptoms requiring urgent attention. We recorded the time<br />

between electronic symptom reporting and FU for pts in the SIS.NET arm.<br />

Data included the number <strong>of</strong> oncology related clinic visits, total number <strong>of</strong><br />

physician visits, and number <strong>of</strong> medical tests including labs and imaging<br />

studies ordered. Kolmogorov-Smirnov tests for equal distributions were<br />

used to determine if there were any significant differences between<br />

SIS.NET and UC. Results: 100 pts were enrolled, 75 completed the 18<br />

month study and 25 pts remain in FU. For the 75 pts, 85% received<br />

chemotherapy and 77% received hormone therapy. Pts in the SIS.NET arm<br />

completed an average <strong>of</strong> 3.8 out <strong>of</strong> 7.2 emailed surveys (52.6%), and pts in<br />

the UC arm completed an average <strong>of</strong> 2.2 out <strong>of</strong> 3.4 (69.1%) routine<br />

pre-clinic surveys. For pts in the SIS.NET arm, 75% <strong>of</strong> reported symptoms<br />

were reviewed by a NP in � 3 days. There was no significant difference<br />

between SIS.NET and UC FU for oncology related clinic appointments,<br />

total number <strong>of</strong> physician visits, or number <strong>of</strong> medical tests performed.<br />

Conclusions: Use <strong>of</strong> on-line health and symptom surveys with remote NP<br />

review provided timely symptom FU but did not reduce clinic visits or<br />

medical testing in pts with ESBC. Further analyses <strong>of</strong> these data as well as<br />

additional studies are necessary to understand the barriers to integration <strong>of</strong><br />

web-based tools to achieve more efficient FU care for BC survivors.<br />

9109 General Poster Session (Board #46G), Sat, 8:00 AM-12:00 PM<br />

Anemia and functional disability in older adults with cancer. Presenting<br />

Author: Cynthia Owusu, Case Western Reserve University, Cleveland, OH<br />

Background: While anemia is associated with functional disability in older<br />

adults in general, this relationship has not been well characterized in older<br />

adults with cancer. We sought to examine the association between anemia<br />

and functional disability and to identify other factors associated with<br />

functional disability in patients (pts) age �65 with cancer. Methods: We<br />

conducted a secondary analysis <strong>of</strong> a multi-center prospective study <strong>of</strong> 500<br />

pts � age 65 that identified predictors <strong>of</strong> chemotherapy (chemo) toxicity<br />

(Hurria et al, JCO, 2011). The primary outcome <strong>of</strong> this analysis was<br />

functional disability, defined as need for assistance with �1 instrumental<br />

activities <strong>of</strong> daily living. Data collected prior to initiation <strong>of</strong> a new chemo<br />

regimen included: age, tumor/treatment variables, labs [including hemoglobin<br />

(Hb)], and geriatric assessment (functional status, comorbidity, social<br />

support, psychological, cognitive, and nutritional status). Anemia (World<br />

Health Organization criteria) was defined as Hb �12g/dl (women) and Hb<br />

�13g/dl (men). Bivariate analysis and logistic regression were used to<br />

examine the association between functional disability, anemia, and other<br />

pre-treatment variables. Results: Among 500 pts [median age 72 years<br />

(range 65-91), 56% female, 61% stage IV], the prevalence <strong>of</strong> functional<br />

disability and anemia was 43% and 51%, respectively. The mean Hb was<br />

11.7g/dl and 12.6g/dl among pts with and without functional disability.<br />

Pts with anemia were more likely to report functional disability (53% vs.<br />

46%, p�0.01). On multivariate analysis, factors associated with functional<br />

disability included anemia [Odds Ratio (OR)� 2.06, 95% confidence<br />

interval (CI)� 1.39-3.05], increased age (OR� 1.04, 95% CI �<br />

1.00-1.07), increased comorbidity (OR� 1.23, 95% CI � 1.09-1.39),<br />

advanced stage (OR� 1.85, 95% CI � 1.22-2.80), and unintentional<br />

weight loss (OR� 2.02, 95% CI � 1.37-3.00). Conclusions: Anemia was<br />

highly prevalent and was associated with functional disability. Prospective<br />

studies that further examine the relationship between anemia and functional<br />

disability and randomized-controlled trials that evaluate the correction<br />

<strong>of</strong> anemia as a modifiable strategy to improve functional status in older<br />

pts with cancer are warranted.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


594s Patient and Survivor Care<br />

9110 General Poster Session (Board #46H), Sat, 8:00 AM-12:00 PM<br />

Efficacy and safety <strong>of</strong> half-dose pegfilgrastim in cancer patients receiving<br />

cytotoxic chemotherapy. Presenting Author: Ryan C. Ramaekers, Saint<br />

Francis Cancer Treatment Center, Grand Island, NE<br />

Background: Pegfilgrastim reduces neutropenia risk in patients (pts) on<br />

cytotoxic chemotherapy. A single 6 mg dose per chemotherapy cycle<br />

commonly causes bone pain and leukocytosis. While half-dose pegfilgrastim<br />

has been shown to be safe and effective in breast cancer pts, there is no<br />

data on half-dose pegfilgrastim in general oncology pts. Methods: We<br />

evaluated the efficacy and safety <strong>of</strong> half-dose pegfilgrastim in general<br />

oncology pts on cytotoxic chemotherapy at St Francis Cancer Center. Pts<br />

who received at least one dose <strong>of</strong> 6 mg pegfilgrastim and developed<br />

NCI-CTCAE grade 2 or more bone pain and/or leukocytosis (WBC�10,000/<br />

ml) were given 3 mg dose pegfilgrastim on their following chemotherapy<br />

cycles. Pt demographics, type/number <strong>of</strong> chemotherapy regimens, efficacy,<br />

side effects and complications were evaluated. McNemar’s test, logistic<br />

regression analysis and ANOVA were used for statistical analysis. Results:<br />

Twenty-six pts (3 males, 23 females, all Caucasian) with median age 55<br />

(range 34-86) received a total <strong>of</strong> eighty-three 3 mg doses <strong>of</strong> pegfilgrastim.<br />

Cancers treated were breast (N�16), colorectal (N�7), head and neck<br />

(N�1), non-Hodgkin lymphoma (N�1) and non-small cell lung cancer<br />

(N�1). Chemotherapy received was anthracycline (N�9), taxane (N�7),<br />

5-FU/oxaliplatin (N�7), 5-FU/cisplatin (N�1), gemcitabine/oxaliplatin<br />

(N�1), pemetrexed/carboplatin (N�1). No neutropenia or chemotherapy<br />

dose modifications occurred on half-dose pegfilgrastim. In the 26 pts we<br />

report, bone pain occurred in 23 pts at 6 mg and 14 pts at 3 mg dose.<br />

Leukocytosis occurred in 23 pts at 6 mg and only 2 pts at 3 mg dose<br />

(McNemar’s test, P�0.01). While older age and full-dose pegfilgrastim<br />

were predictive <strong>of</strong> bone pain, more than one line <strong>of</strong> chemotherapy and<br />

half-dose pegfilgrastim were predictive <strong>of</strong> lack <strong>of</strong> leukocytosis (logistic<br />

regression analysis/ANOVA, P�0.01). Conclusions: Half-dose pegfilgrastim<br />

in general oncology pts receiving cytotoxic chemotherapy seems to be safe<br />

and effective with less bone pain and leukocytosis without resultant<br />

neutropenia or need for chemo modification. Larger prospective studies <strong>of</strong><br />

half-dose pegfilgrastim in this setting are needed to further understand the<br />

feasibility <strong>of</strong> this approach.<br />

9112 General Poster Session (Board #47B), Sat, 8:00 AM-12:00 PM<br />

Determinants <strong>of</strong> fatigue improvement in outpatient oncology according to<br />

baseline categories <strong>of</strong> fatigue severity. Presenting Author: Michael Fisch,<br />

University <strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: Understanding the determinants <strong>of</strong> fatigue response may help<br />

distinguish between different fatigue phenotypes and inform trial designs.<br />

Methods: Patients with invasive cancer <strong>of</strong> the breast, prostate, colon/<br />

rectum, or lung were enrolled from multiple sites. At baseline and 4-5<br />

weeks later patients rated their symptoms on a 0-10 numerical rating scale.<br />

A 2-point change was considered clinically significant for fatigue change.<br />

Logistic regression models and ordinal logistic regression models were used<br />

to examine the effects <strong>of</strong> demographic and clinical factors on fatigue<br />

change. Separate models were constructed for fatigue deterioration in<br />

patients with mild fatigue at baseline and lack <strong>of</strong> improvement for those<br />

with severe fatigue at baseline or any significant change for those with<br />

moderate baseline fatigue. Results: 3,106 pts were enrolled at baseline and<br />

3,032 were analyzable for fatigue change. At baseline, 23% had no<br />

fatigue, 35% mild, 25% moderate, and 17% severe. Improvement varied<br />

by baseline fatigue score with response in 54% with severe fatigue, 31%<br />

with moderate, and 9% with mild. Overall, 13 different parameters were<br />

significant in these 3 models <strong>of</strong> fatigue change, but no single parameter<br />

was common to all 3 categories <strong>of</strong> fatigue. Exposure to anxiolytics and<br />

duration <strong>of</strong> current treatment were significant in models <strong>of</strong> mild and severe<br />

fatigue; antidepressant exposure was significant for mild and moderate<br />

fatigue, and baseline fatigue level was a significant parameter in moderate<br />

and severe fatigue. In colorectal and lung cancer patients, gender was not<br />

significant for fatigue change in patients with mild or moderate fatigue, but<br />

gender was the strongest predictor fatigue improvement in patients with<br />

severe fatigue with men most likely to significantly improve (OR�2.49,<br />

95% CI 1.15-5.41, p�0.021). Conclusions: <strong>Clinical</strong>ly significant improvement<br />

in fatigue varies between 9% and 54%. Predictors <strong>of</strong> fatigue change<br />

vary depending on categories <strong>of</strong> baseline fatigue severity. Gender is the<br />

strongest predictor for fatigue improvement among lung and colorectal<br />

outpatients with severe baseline fatigue. Future trial designs should<br />

account for gender and baseline fatigue severity.<br />

9111 General Poster Session (Board #47A), Sat, 8:00 AM-12:00 PM<br />

Continued suppression <strong>of</strong> bone turnover following a single dose <strong>of</strong> zoledronic<br />

acid: Time to re-think dosing intervals in the management <strong>of</strong> bone<br />

metastases? Presenting Author: Christine E. Simmons, St. Michael’s<br />

Hospital, University <strong>of</strong> Toronto, Toronto, ON, Canada<br />

Background: The management <strong>of</strong> patients (pts) with bone metastases has<br />

changed significantly over the past decade, with the advent <strong>of</strong> newer, more<br />

potent anti-osteoclastic agents. However, the dosing interval in which these<br />

agents are given has not changed; pts are still dosed at a frequency <strong>of</strong><br />

monthly or q3weekly injections. It is not known if this is the optimal dosing<br />

frequency or due to convention. The purpose <strong>of</strong> this study was to determine<br />

the duration <strong>of</strong> suppression <strong>of</strong> bone turnover by Zoledronic Acid (ZA) in a<br />

population <strong>of</strong> bisphosphonate naive metastatic breast cancer (MBC) pts.<br />

Methods: This prospective single arm phase II study enrolled 26 pts with<br />

MBC to bone who had not received bisphosphonates in the metastatic<br />

setting. A single dose <strong>of</strong> ZA was given at week 0, and biomarkers <strong>of</strong> bone<br />

turnover (serum CTX) were collected every 2 weeks subsequently. Pain<br />

scores and quality <strong>of</strong> life data were collected every 4 weeks. Pts remained<br />

on study if their biomarkers remained suppressed, but came <strong>of</strong>f study as<br />

soon as escape from suppression was noted, (defined as a rise � 50% <strong>of</strong><br />

baseline value). Results: At 12 weeks, 73% (95% CI 56%-90%) <strong>of</strong> pts had<br />

continued suppression <strong>of</strong> bone turnover demonstrated by serum CTX. The<br />

pts that escaped suppression did so at a median <strong>of</strong> 8 weeks after first<br />

infusion <strong>of</strong> ZA (range 8-10 weeks). The magnitude <strong>of</strong> suppression (proportion<br />

decrease from baseline) <strong>of</strong> serum CTX at week 4 was significantly<br />

greater in those who had continued suppression compared to those who<br />

escaped suppression before 12 weeks (75% vs 58%, p � 0.02). The<br />

absolute baseline value <strong>of</strong> CTX was significantly lower in pts who maintained<br />

suppression (507 vs. 745 ng/L, p � 0.04). Quality <strong>of</strong> life scores and<br />

pain medication use did not change appreciably during this period.<br />

Conclusions: This study suggests that ZA may not need to be given at<br />

conventional intervals in the majority <strong>of</strong> pts, and may safely be withheld<br />

without increased risk to morbidity or decline in quality <strong>of</strong> life over a 12<br />

week period. Baseline CTX and the magnitude <strong>of</strong> suppression <strong>of</strong> bone<br />

turnover at 4 weeks may predict for the optimal dosing frequency.<br />

Confirmation in a randomized trial is needed.<br />

9113 General Poster Session (Board #47C), Sat, 8:00 AM-12:00 PM<br />

Pain, sleep disturbance, and fatigue symptom cluster in patients suffering<br />

from chronic chemotherapy-induced neuropathic pain (CINP). Presenting<br />

Author: Jennifer S. Gewandter, University <strong>of</strong> Rochester Medical Center,<br />

Rochester, NY<br />

Background: Various symptom cluster combinations <strong>of</strong> pain, fatigue, sleep<br />

disturbance (SD), and depression have been identified in cancer patients.<br />

The presence <strong>of</strong> symptom clusters has not been assessed in patients with<br />

persistent CINP. This exploratory analysis aimed to determine which<br />

symptoms statistically clustered with pain in cancer survivors with CINP.<br />

Methods: The University <strong>of</strong> Rochester Cancer Center Community <strong>Clinical</strong><br />

Oncology Program recruited 461 patients with CINP � 4 (on a 0-10 scale)<br />

who had completed chemotherapy (median 7 months ago) for a pain<br />

intervention trial. At baseline, groups <strong>of</strong> highly associated symptoms that<br />

included pain were identified empirically using factor and cluster analyses<br />

<strong>of</strong> the 11 symptoms in the University <strong>of</strong> Rochester Symptom Inventory plus<br />

the Hospital Anxiety and Depression Scale (HADS) scores. To investigate if<br />

associated symptoms track together over time, multiple linear regression<br />

(MLR) analysis was performed using changes in symptom severity between<br />

baseline and week 6, controlling for gender, age, education, race, and<br />

marital status. Results: Subjects were 88% white and 71% female, and on<br />

average 61 years old. Mean (standard deviation) baseline pain, fatigue, and<br />

SD were 5.7 (2.8), 5.0 (2.7), and 4.2 (3.1), respectively; 26% <strong>of</strong> subjects<br />

had borderline or abnormal HADS scores. Factor analysis identified 3<br />

factors that accounted for 88% <strong>of</strong> the variance. One factor included pain,<br />

fatigue, SD, but not HADS, and accounted for 37% <strong>of</strong> the variance.<br />

Variable clustering also identified pain, SD, and fatigue as 1 symptom<br />

cluster. Changes in severity <strong>of</strong> SD and fatigue (p � 0.0001), but not HADS,<br />

were associated with changes in pain (adjusted R2 � 0.168) in MLR<br />

analysis. Conclusions: Pain, fatigue, and SD were identified as a symptom<br />

cluster by factor and cluster analyses, and were found to track together over<br />

time by MLR. Since these data suggest that pain is associated with sleep<br />

quality and fatigue in patients with persistent CINP, targeting one <strong>of</strong> these<br />

symptoms may lead to reductions in the others. Future research should<br />

investigate interventions that target pain, fatigue, and SD concurrently in<br />

cancer survivors suffering from CINP.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


9114 General Poster Session (Board #47D), Sat, 8:00 AM-12:00 PM<br />

Treatment and mortality rates <strong>of</strong> older cancer patients with a body mass<br />

index Grade 3 2 (50) 3 (30) 2 (67) 6 (67)<br />

Liver AEs 0 1 (10) 0 2 (22)<br />

Renal AEs<br />

Thromboembolic events<br />

2 (50) 0 0 3 (33)<br />

b<br />

0 1 (10) 0 2 (22)<br />

SAEs 1 (25) 2 (20) 1 (33) 5 (56)<br />

Deaths, n (%) c<br />

595s<br />

2 (50) 4 (40)� 1 (33) 3 (33)<br />

Plts: Cycles 2-6, mean (n)<br />

Pre ctx Day 1 309 (4) 443 (9) 314 (3) 442 (9)<br />

Pre ctx Day 8 204 (4) 355 (10) 239 (2) 270 (9)<br />

Pre ctx Day 15 75 (3) 164 (10) - -<br />

Nadir 103 (4) 143 (10) 53 (2) 113 (9)<br />

Grade 3/4 thrombocytopenia: Cycle 2-6, n (%) 1 (25) 0 2 (67) 3 (33)<br />

a 7/33 randomized pts never received epag/pbo. b None were considered related to epag and all<br />

resolved. One event occurred after stopping epag and following disease progression (PD). c All<br />

deaths were from PD; three additional patients died prior to receiving epag or pbo.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


596s Patient and Survivor Care<br />

9118 General Poster Session (Board #47H), Sat, 8:00 AM-12:00 PM<br />

Disease-driven symptoms and response to chemotherapy in treatmentnaive<br />

patients with advanced non-small cell lung cancer (NSCLC). Presenting<br />

Author: Desiree Jones, University <strong>of</strong> Texas M. D. Anderson Cancer<br />

Center, Houston, TX<br />

Background: The disease-driven symptoms <strong>of</strong> treatment-naïve patients with<br />

advanced NSCLC have not been the primary focus <strong>of</strong> previous studies. One<br />

reason for this gap in the literature is that randomized trials typically lack<br />

an opportunity to capture the symptom pr<strong>of</strong>iles <strong>of</strong> patients who are truly<br />

treatment-naïve. To address this need, this study sought to: (i) characterize<br />

the symptom pr<strong>of</strong>ile <strong>of</strong> treatment-naïve patients with advanced NSCLC, and<br />

(ii) describe patients’ response to chemotherapy. Methods: The study<br />

sampleconsisted <strong>of</strong>43 treatment-naïve patients (no prior treatment by<br />

surgery, radiation, or chemotherapy) with a primary diagnosis <strong>of</strong> advanced<br />

NSCLC from the MD Anderson Cancer Center, and was derived from a larger<br />

study (Cleeland et al. J Clin Oncol 29:2859-65, 2011). Patients’ symptoms<br />

were assessed pre-chemotherapy, and then prospectively at 6, 12,<br />

and 18 weeks from initiation <strong>of</strong> chemotherapy using the MD Anderson<br />

Symptom Inventory Lung Cancer module. Results: The top symptoms<br />

reported by patients, in order <strong>of</strong> severity, were shortness <strong>of</strong> breath, fatigue,<br />

distress, sadness, drowsiness, and pain. Chemotherapy did not significantly<br />

improve shortness <strong>of</strong> breath, drowsiness, pain, disturbed sleep, lack<br />

<strong>of</strong> appetite, coughing, and constipation in this sample <strong>of</strong> patients, and was<br />

associated with a significant increase in levels <strong>of</strong> fatigue and numbness<br />

(mean difference in fatigue at 12 weeks from baseline � 2.8; 95% CI,<br />

0.5–2.9; p � .01; and in numbness from baseline � 1.1; 95% CI,<br />

0.2–2.0; p � .02). Conclusions: This study characterizes the disease-driven<br />

symptoms <strong>of</strong> truly treatment-naïve patients with advanced NSCLC. Knowledge<br />

<strong>of</strong> these symptoms is <strong>of</strong> intrinsic value in the clinical management <strong>of</strong><br />

advanced NSCLC as it provides the critical reference point against which<br />

symptom palliation must be measured. Patients’ lack <strong>of</strong> symptomatic<br />

improvement from chemotherapy in this study suggests that this sub-set <strong>of</strong><br />

patients may have had rapidly progressing disease and that chemotherapy<br />

may have prevented symptoms levels from worsening. Consistent with<br />

literature, transient but significant increase in both fatigue and numbness<br />

are chemotherapy-related toxicities.<br />

9120 General Poster Session (Board #48B), Sat, 8:00 AM-12:00 PM<br />

Final results <strong>of</strong> a large collaborative, hospital-based quality improvement<br />

study aimed at the implementation <strong>of</strong> interventions for the psychosocial<br />

care <strong>of</strong> adult cancer patients (HUCARE project). Presenting Author:<br />

Rodolfo Passalacqua, Istituti Ospitalieri di Cremona, Cremona, Italy<br />

Background: Incorporating psychosocial research into practice is challenging.<br />

Aim <strong>of</strong> this study is to assess the feasibility and effectiveness in real life<br />

<strong>of</strong> interventions which have been tested in RCTs and have demonstrated to<br />

improve pts psychosocial conditions Methods: This is an implementation<br />

study <strong>of</strong> five EBM interventions conducted in 28 centers. We adopted the<br />

model <strong>of</strong> Pronovost (BMJ 2008), which includes context analysis to<br />

identify local barriers; introduction <strong>of</strong> the interventions, and evaluation <strong>of</strong><br />

how many pts receive the recommended interventions. Primary EPs: degree<br />

<strong>of</strong> implementation detected in a blinded fashion by external personnel.<br />

Interventions included: 1) EBM communication courses for doctors and<br />

nurses; 2) use <strong>of</strong> a question prompt list (QPL) for all pts; 3) creation <strong>of</strong> the<br />

Point <strong>of</strong> Information and Support (PIS) in the ward (J Clin Oncol<br />

27:1794-99,2009); 4) identification <strong>of</strong> a referring nurse (RN) for informing<br />

and educating pts; 5) screening <strong>of</strong> distress and social needs. Results: In<br />

December 2008, 28 eligible centers applied to participate. 27 are<br />

evaluable and 1 is too early. 156 oncologists and 401 nurses attended the<br />

3 days training course. A QPL was subjected to cross-cultural adaptation,<br />

yielding the first validated Italian QPL (BMC Health Serv Res 2010). 24<br />

centers (89%) have successfully implemented the intervention; 3 centers<br />

failed for various reasons: internal conflicts <strong>of</strong> the staff, poor motivation,<br />

etc. Main results for each intervention are shown in the table. Conclusions:<br />

A successful implementation <strong>of</strong> EBM measure was obtained in the vast<br />

majority <strong>of</strong> oncology centers and yields to significant changes in the<br />

delivery <strong>of</strong> psychosocial care. Project funded by the Italian Ministry <strong>of</strong><br />

Health and Lombardia Region.<br />

EBM training<br />

courses<br />

(No. <strong>of</strong><br />

attendee, oncologists<br />

plus nurses)<br />

Referring<br />

nurse (RN)<br />

(% <strong>of</strong> pts<br />

withaRN)<br />

PIS<br />

(No. <strong>of</strong><br />

centers<br />

with a PIS)<br />

Use <strong>of</strong><br />

the QPL<br />

(% <strong>of</strong> pts<br />

who receive<br />

the QPL)<br />

Psychosocial<br />

evalutaion<br />

(% <strong>of</strong> screened<br />

patients)<br />

Pre-implementation 0/598 0% 5 0% 0%<br />

Post-implementation 557/598 (93%) 86% 22 73% 83%<br />

9119 General Poster Session (Board #48A), Sat, 8:00 AM-12:00 PM<br />

Evaluation <strong>of</strong> a non-peptidic mimic <strong>of</strong> host defense proteins, PMX30063,<br />

in an animal model <strong>of</strong> oral mucositis. Presenting Author: Richard W Scott,<br />

PolyMedix, Inc., Radnor, PA<br />

Background: Oral ulcerative mucositis (OM) is a common, painful, doselimiting<br />

toxicity <strong>of</strong> cancer therapy. The care for OM is largely palliative and<br />

there is an urgent need for new and effective therapies. Host defense<br />

proteins (HDPs) are amphiphilic components <strong>of</strong> the innate immune system<br />

that serve as a primary response to infection. Exclusive <strong>of</strong> their direct<br />

antimicrobial activity, HDPs have been shown to possess immune modulatory<br />

and anti-inflammatory activities. We are developing non-peptidic<br />

mimics <strong>of</strong> HDPs, capturing the structural and biological properties <strong>of</strong> HDPs<br />

within a framework <strong>of</strong> smaller fully synthetic oligomers. The lead compound,<br />

PMX30063, is in Phase 2 clinical trials for iv treatment <strong>of</strong> systemic<br />

staphylococcal infections. Recently, we observed that HDP mimics attenuate<br />

anti-inflammatory pathways and reasoned that these activities could be<br />

leveraged into a mucositis intervention. Methods: In an acute radiation<br />

hamster model, PMX30063 was applied as a topical rinse, 3 times daily at<br />

1, 3 or 10 mg/ml over a 28 day treatment regimen ( n � 10), following a<br />

single radiation dose to the buccal cheek pouch (40 Gy). In a fractionated<br />

radiation model, radiation (7.5 Gy/dose) was administered on days 0, 1, 2,<br />

3, 6, 7, 8 and 9, targeting the left buccal pouch mucosa. PMX30063 was<br />

given topically 3 times daily at 3 mg/ml over a 35 day regimen beginning on<br />

day0(n�10). OM was scored using an established blinded method.<br />

Results: In the acute model, PMX30063 reduced the number <strong>of</strong> animal<br />

days with ulceration from 43% in vehicle-treated hamsters to � 5% in all 3<br />

PMX30063 dose groups (�2 test; p�0.001). In the fractionated model,<br />

PMX30063 significantly reduced the daily mucositis scores prior to peak<br />

mucositis and throughout the remaining course <strong>of</strong> treatment (Mann-<br />

Whitney Rank-sum analysis; p�0.001). PMX30063 reduced the number<br />

<strong>of</strong> animal days with ulceration from 55% in vehicle-treated hamsters to<br />

3.3% (�2 test; p�0.001). The time courses <strong>of</strong> efficacy argue that direct<br />

antimicrobial action is not the primary driver <strong>of</strong> efficacy. Conclusions: The<br />

safe and highly efficacious activity <strong>of</strong> PMX30063 in animals supports its<br />

further development as a topical therapeutic to prevent OM in cancer<br />

patients.<br />

9121 General Poster Session (Board #48C), Sat, 8:00 AM-12:00 PM<br />

<strong>Clinical</strong> ethics consultation in oncology. Presenting Author: Andrew G.<br />

Shuman, Memorial Sloan-Kettering Cancer Center, New York, NY<br />

Background: There is limited empirical research exploring the nature <strong>of</strong><br />

clinical ethical consultations within the oncology population. Our objective<br />

is to review, describe and compare clinical ethics consultations at two<br />

NCI-designated comprehensive cancer centers, in order to identify opportunities<br />

for systems improvement in clinical care. Methods: This case series is<br />

derived from prospectively-maintained clinical ethics consultation databases<br />

at each institution. All adult oncology patients receiving ethics<br />

consultation from 2007 through 2011 were included as eligible cases.<br />

Both qualitative and quantitative analyses were undertaken. Demographic<br />

and clinical information were obtained from the databases for all patients,<br />

and verified via chart abstraction. Additional variables studied included the<br />

reason for and context <strong>of</strong> the ethical consultation, the patient’s code status<br />

before and after consultation, and involvement <strong>of</strong> palliative care or other<br />

adjuvant services. Opportunities for systems-level improvements and/or<br />

educational initiatives were identified. Results: A total <strong>of</strong> 207 eligible cases<br />

were identified. The most common primary issues leading to ethics<br />

consultation were code status and advance directives (25%), surrogate<br />

decision-making (17%), and medical futility (13%). Communication lapses<br />

were identified in 41%, and interpersonal conflict arose in 51%. Prior to<br />

ethics consultation, 26% <strong>of</strong> patients were DNR; 60% were DNR after<br />

ethics consultation. Palliative care consultation occurred in 41% <strong>of</strong> cases.<br />

Opportunities for systems improvement and pr<strong>of</strong>essional education related<br />

to goals <strong>of</strong> care at the end <strong>of</strong> life, the role <strong>of</strong> palliative care involvement, and<br />

improved communication. Conclusions: Ethics consultations among cancer<br />

patients reflect the realities inherent to their clinical management. Appropriately<br />

addressing advance directives within the context <strong>of</strong> overall goals <strong>of</strong><br />

care is crucial. Thoughtful consideration <strong>of</strong> communication barriers,<br />

sources <strong>of</strong> interpersonal conflict, symptom control, and end-<strong>of</strong>-life care are<br />

paramount to optimal management strategies in this patient population.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


9122 General Poster Session (Board #48D), Sat, 8:00 AM-12:00 PM<br />

A prospective longitudinal study <strong>of</strong> cancer-related fatigue in patients undergoing<br />

breast-conserving surgery and radiation with or without chemotherapy<br />

for breast cancer. Presenting Author: Mylin Ann Torres, Department<br />

<strong>of</strong> Radiation Oncology, Emory University, Atlanta, GA<br />

Background: We prospectively evaluated risk factors for persistent cancerrelated<br />

fatigue in women with breast cancer undergoing lumpectomy with<br />

or without chemotherapy (CTX) prior to whole breast radiotherapy (XRT).<br />

We assessed the potential role <strong>of</strong> inflammatory mediators, demographic<br />

characteristics, and treatment history including CTX. Methods: Following<br />

lumpectomy, 60 women received a definitive course <strong>of</strong> whole breast XRT<br />

(50 Gy plus a 10 Gy boost). Prior to XRT, at week 6 <strong>of</strong> XRT, and 6 weeks<br />

post XRT, subjects completed the Multidimensional Fatigue Inventory<br />

(MFI) and underwent blood draws for inflammatory mediators (protein and<br />

mRNA). Results: Independent multivariate analyses <strong>of</strong> clinical and demographic<br />

factors revealed that CTX (p�.001) , given neoadjuvantly or<br />

adjuvantly, and age �50 (p�.03) were significant predictors <strong>of</strong> higher<br />

fatigue scores post XRT. Mean MFI scores in patients treated with CTX<br />

(n�24) were 20 points higher than patients not treated with CTX (p�.001)<br />

with a clinically meaningful difference in scores being 10 points on the<br />

MFI. Gene ontology analysis <strong>of</strong> differentially expressed genes indicated<br />

increased activation <strong>of</strong> genes involved in immune and inflammatory<br />

responses in fatigued vs. non-fatigued patients (p�.001). Of the inflammatory<br />

mediators, plasma IL-6 prior to XRT was the strongest predictor <strong>of</strong> post<br />

XRT fatigue (p�.02). Moreover, plasma IL-6 concentrations prior to XRT<br />

were significantly higher in patients who received CTX (mean 4.96 vs.<br />

2.53, p�.01). Patients who received CTX also had significantly higher<br />

levels <strong>of</strong> NF Kappa B DNA binding 6 weeks post XRT (p�.001), and<br />

transcription factor binding analysis revealed a greater representation <strong>of</strong><br />

genes with the NF Kappa B DNA binding motif in fatigued vs. non-fatigued<br />

patients (p �.05). Conclusions: Collectively, these data suggest an interaction<br />

between CTX and XRT leading to inflammation and fatigue several<br />

weeks post XRT. This relationship was independent <strong>of</strong> whether CTX was<br />

given pre or post-operatively. Treatments targeting inflammation before<br />

XRT may reduce fatigue post therapy, particularly in patients previously<br />

treated with CTX.<br />

9124 General Poster Session (Board #48F), Sat, 8:00 AM-12:00 PM<br />

A national survey <strong>of</strong> support and information needs <strong>of</strong> Australian women<br />

living with advanced breast cancer. Presenting Author: Linda R. Mileshkin,<br />

Peter MacCallum Cancer Centre, Melbourne, Australia<br />

Background: Improved treatments mean women with advanced breast<br />

cancer (ABC) are living longer, sometimes for many years. As a result, these<br />

women may experience chronic needs that are different from those with<br />

early breast cancer. We aimed to assess the support and information needs<br />

<strong>of</strong> women living with ABC. Methods: A national postal survey was sent to<br />

2,345 women with ABC registered as members <strong>of</strong> the consumer organizations<br />

BCNA and/or BreaCan. Women were asked about their disease,<br />

treatment, experiences <strong>of</strong> care, and to complete the Supportive Care Needs<br />

Survey. Results: The response rate was 34% (792 valid responses). Mean<br />

age was 57 (range 25-99) with 21% living alone and 27% working. 18%<br />

reported living with ABC for �7 years. 656 (85%) were having current<br />

treatment: chemotherapy (43%), hormonal therapy (46%), bisphosphonates<br />

(49%), and/or trastuzumab (Herceptin) (19%) respectively. 49% <strong>of</strong><br />

women reported having access to a Breast Care Nurse (BCN) since<br />

diagnosis <strong>of</strong> ABC but only 3% cited a BCN or cancer nurse as their main<br />

contact. The majority (76%) cited their medical oncologist as their main<br />

contact, 8% cited their family doctor. Women wanted information about:<br />

treatment options (84.2%), new treatments (79.2%), symptoms/side<br />

effects (78.9%), clinical trials (60.8%), managing pain (57.2%) and<br />

financial assistance (38%). Women � 65 had significantly higher levels <strong>of</strong><br />

unmet needs than those �65 for the 7 items below, including all items<br />

from the sexual domain. Conclusions: Women with ABC have many unmet<br />

needs with younger women particularly needing more support with sexual<br />

needs and anxiety. Women are heavily reliant on their medical oncologist,<br />

who may not be equipped or best placed to meet these needs. Models <strong>of</strong><br />

care need to be developed to address the unmet supportive care needs <strong>of</strong><br />

women living with ABC.<br />

Moderate-high Moderate-high<br />

Item<br />

need < 65<br />

need > 65 P value<br />

Uncertainty about the future 50% 39% 0.009<br />

Concerns about the worries <strong>of</strong><br />

those close to you<br />

48% 35% 0.002<br />

Worry that the results <strong>of</strong><br />

treatment are beyond your control<br />

37% 27% 0.026<br />

Changes in sexual relationships 28% 15% 0.001<br />

Changes in sexual feelings 27% 15% 0.001<br />

Being given information about<br />

sexual relationships<br />

21% 9% �0.0001<br />

More choice about cancer specialists 17% 8% 0.002<br />

Patient and Survivor Care<br />

597s<br />

9123 General Poster Session (Board #48E), Sat, 8:00 AM-12:00 PM<br />

The relationship among age, anxiety, and depression in older adults with<br />

cancer. Presenting Author: Talia Weiss, Memorial Sloan-Kettering Cancer<br />

Center, New York, NY<br />

Background: Depression and anxiety are common psychological sequelae <strong>of</strong><br />

cancer, resulting in decreased adherence to treatment regimens and longer<br />

hospital stays. In men with prostate cancer, aging is associated with<br />

reduced anxiety and increased depression. The overall goal <strong>of</strong> this study<br />

was to examine the association among age, anxiety, and depression in a<br />

cohort <strong>of</strong> older adults receiving chemotherapy (chemo). Methods: This is a<br />

secondary analysis <strong>of</strong> a prospective longitudinal study investigating chemotherapy<br />

toxicity in older adults with cancer. Eligibility included: age � 65,<br />

diagnosis <strong>of</strong> cancer, and scheduled to receive a new chemo regimen.<br />

Baseline data (pre-chemotherapy) included: age, sociodemographics, tumor<br />

and treatment factors (including tumor type and stage), geriatric<br />

assessment parameters (functional status, comorbidities, psychological<br />

state, nutritional status, social support). Anxiety and depression were<br />

measured by the Hospital Anxiety and Depression Scale (HADS). Univariate<br />

and multiple regression analyses were conducted to test the relationship<br />

between age, anxiety, and depression. Results: The average age <strong>of</strong> the 500<br />

patients (56% females) was 73.1 (range 65-91, SD�6.18) with 5% Stage<br />

I, 12% Stage II, 22% Stage III and 61% stage IV. Mean depression and<br />

anxiety scores were: 3.6�3.17; 4.7�3.60. <strong>Clinical</strong>ly significant depression<br />

was reported in 12.6% (n�62). <strong>Clinical</strong>ly significant anxiety was<br />

reported in 20.9% (n�103). In univariate analyses, there was no association<br />

between anxiety and age, or depression and age. In multivariable<br />

analyses, older age (beta� -0.07, p � 0.05) was associated with decreased<br />

anxiety, as well as lack <strong>of</strong> social support (p � 0.01) and increased number<br />

<strong>of</strong> comorbidities (p � 0.01). In multivariable analysis, depression was<br />

associated with lack <strong>of</strong> social support (p � 0.01), increased number <strong>of</strong><br />

comorbidities (p � 0.01), and advanced stage (p � 0.01). Conclusions:<br />

This study supports previous research that anxiety decreases with age in<br />

older adults with cancer. However, depression remained constant with<br />

increasing age. Greater resources and attention to identifying and treating<br />

the psychological sequelae <strong>of</strong> cancer in older adults are warranted.<br />

9125 General Poster Session (Board #48G), Sat, 8:00 AM-12:00 PM<br />

Efficacy and safety <strong>of</strong> balugrastim compared with pegfilgrastim in patients<br />

with breast cancer who are receiving chemotherapy. Presenting Author:<br />

Constantin D. Volovat, Centrul de Oncologie Medicala, Iasi, Romania<br />

Background: Patients receiving cancer chemotherapy are at an increased<br />

risk <strong>of</strong> neutropenia. Recombinant granulocyte colony stimulating factors<br />

(G-CSFs) have been developed to stimulate proliferation and differentiation<br />

<strong>of</strong> neutrophils. Pegfilgrastim is a pegylated recombinant G-CSF that allows<br />

for once-per-cycle dosing. Balugrastim is a long-acting G-CSF composed <strong>of</strong><br />

a genetic fusion between recombinant human serum albumin and G-CSF.<br />

The objective <strong>of</strong> this study was to compare the efficacy and safety <strong>of</strong><br />

balugrastim and pegfilgrastim in patients with histologically or cytologically<br />

confirmed breast cancer who were scheduled to receive doxorubicin and<br />

docetaxel. Methods: In this double-blind, randomized, active-comparator,<br />

noninferiority trial, patients with �1.5x109 neutrophils/L, and �100x109 platelets/L were randomly assigned to subcutaneous injections <strong>of</strong> balugrastim<br />

40 mg (n�153) or pegfilgrastim 6 mg (n�151) with stratifications for<br />

weight, prior chemotherapy exposure, and global location. The primary<br />

efficacy endpoint was the duration <strong>of</strong> severe neutropenia (days with an<br />

absolute neutrophil count �0.5x109 cells/L) during the cycle 1 for the<br />

population <strong>of</strong> patients who did not have major protocol violations. Results:<br />

Mean duration <strong>of</strong> severe neutropenia in cycle 1 was 1.1 days in the<br />

balugrastim group and 1.0 days in the pegfilgrastim group (95% CI for<br />

difference between groups -0.13 to 0.37). Fifty-eight percent <strong>of</strong> patients in<br />

the balugrastim group and 59% in the pegfilgrastim group had severe<br />

neutropenia during cycle 1 (95% CI for difference between groups<br />

-11.98% to 10.41%). Two and 4 patients, respectively, had febrile<br />

neutropenia during cycle 1; no patients in either group had febrile<br />

neutropenia during cycles 2-4. Twenty percent <strong>of</strong> patients in the balugrastim<br />

group and 19% in the pegfilgrastim group had adverse events that the<br />

investigator considered to be related to study medication. Six and 7<br />

patients, respectively, had serious adverse events. Conclusions: The results<br />

<strong>of</strong> this study support the noninferiority <strong>of</strong> balugrastim versus pegfilgrastim,<br />

demonstrating that both compounds have comparable efficacy. There were<br />

no unexpected safety events.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


598s Patient and Survivor Care<br />

9126 General Poster Session (Board #48H), Sat, 8:00 AM-12:00 PM<br />

Using scenarios to explain survival time: Attitudes <strong>of</strong> people with a cancer<br />

experience. Presenting Author: Martin R. Stockler, NHMRC <strong>Clinical</strong> Trials<br />

Centre, University <strong>of</strong> Sydney, Sydney, Australia<br />

Background: Most people with advanced cancer want some information on<br />

their life expectancy. We sought the attitudes <strong>of</strong> people with a cancer<br />

experience to using three scenarios for survival (best-case, worst-case, and<br />

typical) to explain life expectancy to people with advanced cancer.<br />

Methods: Oncology clinic attendees and Breast Cancer Network Australia<br />

(BCNA) members completed a survey describing two formats for explaining<br />

life expectancy to a hypothetical patient with incurable cancer - providing<br />

either three scenarios for survival or just the median survival time.<br />

Associations between respondent attitudes and their demographic and<br />

tumour characteristics were explored. Results: Characteristics <strong>of</strong> the505<br />

respondents from outpatient clinics (n�251) and BCNA (n�254) were:<br />

median age 58 years; female 74%; completed high school education 80%;<br />

breast primary 64%; self-report <strong>of</strong> cancer spread to other body parts 23%<br />

and; a median <strong>of</strong> 4 years since diagnosis <strong>of</strong> cancer. More respondents<br />

agreed that explaining three scenarios (vs. just the median survival time)<br />

would: make sense (93% vs. 75%), be helpful (93% vs. 69%), convey hope<br />

(68% vs. 44%), reassure (60% vs. 40%) and help family members (91%<br />

vs. 71%); all p-values �0.001. Fewer respondents agreed that explaining<br />

three scenarios (vs. median survival) would upset people (24% v 36%);<br />

p�0.001. The proportions <strong>of</strong> respondents agreeing that each <strong>of</strong> the 3<br />

scenarios should be presented were: best-case 89%, worst-case 82%, and<br />

typical 92%. Most respondents (85%) agreed it would be helpful to receive<br />

a printed summary <strong>of</strong> the scenarios for survival. For information about their<br />

own prognosis, 88% preferred all three scenarios and 5% a single estimate<br />

<strong>of</strong> the median. Respondents with higher education were more likely to agree<br />

that presenting three scenarios would be helpful (95% v 90%, p�0.05).<br />

Respondents with breast cancer were more likely to agree that explaining<br />

three scenarios would upset people (31% v 13%, p�0.001). Conclusions:<br />

Most respondents judged presentation <strong>of</strong> best-case, worst-case and typical<br />

scenarios preferable and more helpful and reassuring than presentation <strong>of</strong><br />

just the median survival time when explaining life expectancy to patients<br />

with advanced cancer.<br />

9128 General Poster Session (Board #49B), Sat, 8:00 AM-12:00 PM<br />

Caring for people at the end <strong>of</strong> life: How do cancer caregivers differ from<br />

other caregivers? Presenting Author: Afaf Girgis, University <strong>of</strong> New South<br />

Wales, Sydney, Australia<br />

Background: The size <strong>of</strong> the population in receipt <strong>of</strong> cancer care is growing<br />

as the population ages and grows, with many receiving outpatient care,<br />

living for longer and wishing to be cared for at home. Hence, cancer is now<br />

one <strong>of</strong> the most common health conditions in receipt <strong>of</strong> informal care<br />

giving. Methods: The South Australian Health Omnibus is an annual,<br />

face-to-face, cross-sectional, whole-<strong>of</strong>-population, multi-stage, systematic<br />

area sampling survey which seeks a minimum <strong>of</strong> 3,000 respondents each<br />

year state-wide. From 2000-2007, questions about end <strong>of</strong> life were asked.<br />

This study compares people who cared for someone with cancer until death<br />

(cancer caregivers) with caregivers <strong>of</strong> people with other life limiting<br />

illnesses (non-cancer caregivers). Results: 4,665/14,624 (31.9%; participation<br />

rate 71.6%) had someone close to them die from an “expected”<br />

death in the 5 years prior to being surveyed. One in 10 (10%; 1,504/<br />

14,624) provided hands-on (day-to-day or intermittent) care; the majority<br />

(79.5%) having cared for someone with cancer. Compared to non-cancer<br />

caregivers, cancer caregivers cared for someone who was significantly<br />

younger (mean age 66 vs 74 years; p�0.0001), and to have had a palliative<br />

care service involved in the care <strong>of</strong> that person (64.9% vs 39%; p�0.000).<br />

Whilst not statistically significant, cancer caregivers were somewhat less<br />

likely to be the spouse <strong>of</strong> the person they cared for (11.8% vs 16.8%); more<br />

likely to be <strong>of</strong> non-English speaking background (11% vs 7.5%); more<br />

likely to report that the deceased was comfortable in the last 2 weeks <strong>of</strong> life<br />

(44.1% vs 31.7%); and prepared to care again for someone with a<br />

life-limiting illness (81.3% vs 71.4%). Conclusions: Caring for someone<br />

with cancer at the end <strong>of</strong> life appears to be fundamentally different to other<br />

caregiver populations, particularly in relation to age <strong>of</strong>, and relationship to,<br />

the patient, which may contribute to their substantially greater utilisation<br />

<strong>of</strong> palliative care services. Being a younger caregiver increases the<br />

likelihood <strong>of</strong> them caring again in the future.<br />

9127 General Poster Session (Board #49A), Sat, 8:00 AM-12:00 PM<br />

A prospective study <strong>of</strong> family conferences (FCs) in a palliative care unit<br />

(PCU) at a comprehensive cancer center. Presenting Author: Rony Dev,<br />

University <strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: TheFC provides the family information and emotional support<br />

in the transition to end-<strong>of</strong>-life care. There are a few, mainly qualitative,<br />

studies <strong>of</strong> FCs in a palliative care setting. A retrospective report suggested<br />

increased emotional expression by family members when patients were not<br />

present in the FC. The purpose <strong>of</strong> our study was to determine the<br />

characteristics <strong>of</strong> FCs and the effect <strong>of</strong> patient participation on emotional<br />

expression by family members. Methods: A prospective study was conducted<br />

during 18-month period with140 consecutive FCs conducted by an<br />

interdisciplinary team (palliative care physician, social worker, and others).<br />

Data collected included demographics, discharge disposition, number <strong>of</strong><br />

participants, caregiver characteristics, expressions <strong>of</strong> emotional distress,<br />

conflict with healthcare providers, and topics discussed. Descriptive<br />

statistics and frequencies were calculated. Analysis was performed using a<br />

chi-square test. Results: 70 patients (50%) were female, 64 (46%) white,<br />

127 (91%) with solid tumors and a median age <strong>of</strong> 59. In 68/140 family<br />

meetings (49%), patients actively participated. The primary caregivers<br />

were predominantly female (66%), white (49%), and either the spouse or<br />

partner (59%). A median number <strong>of</strong> 4 family members and 4 healthcare<br />

providers attended the FCs with median duration <strong>of</strong> 1 hour and median <strong>of</strong> 2<br />

days prior to discharge. Questions concerning advanced directives, symptoms<br />

anticipated at death and caregiver well-being were infrequent.<br />

Patients verbalized distress frequently, (73.1%). Primary caregivers emotional<br />

expression <strong>of</strong> verbal distress were high but not significantly affected<br />

by patient presence (82% vs 82%, P:NS); however, verbal expressions <strong>of</strong><br />

emotional distress by family were more common when patients were absent<br />

(87%) than when present (73%), p � 0.037. Conclusions: There was a high<br />

frequency <strong>of</strong> expression <strong>of</strong> emotional distress by patients and family<br />

members. Patient participation was associated with decreased verbal<br />

emotional expression by family members but not the primary caregiver.<br />

Further studies are needed on the benefits <strong>of</strong> allocating additional time to<br />

meet with family members without patient presence during a FC.<br />

9129 General Poster Session (Board #49C), Sat, 8:00 AM-12:00 PM<br />

Polymorphisms in genes OPRM1, �-arrestin 2, STAT6, and COMT and<br />

clinical outcome to opioid therapy. Presenting Author: Sara Cros, Catalan<br />

Institute <strong>of</strong> Oncology, Hospital Germans Trias i Pujol, Barcelona, Spain<br />

Background: Morphine sulfate is the most widely used opioid in oncologic<br />

pain. There is a high variability in response between individuals. The<br />

objectives are to correlate the variability in response (R) and tolerability<br />

with polymorphisms within OPRM1, �-arrestin 2, STAT6 and COMT gene<br />

sequences. Methods: DNA from 44 oncologic patients (pts) with an EVA<br />

score � 6 was prospectively analyzed using allelic discrimination techniques<br />

and automatic sequencing. Pts (never treated with opioids) received<br />

morphine sulfate 10 mg per day. Pain was assessed by EVA score and<br />

toxicity was evaluated according to the most common opioid side effects.<br />

Symptomatology was reassessed after 72 hours. Chi-squared and Fisher’s<br />

tests where used to analyze data. Results: 17 pts (38.6%) responded with<br />

an EVA � 3. 6 pts (13.6%) did not experience toxicity, 36 pts (81.8%) had<br />

mild side effects and 2 pts (4.5%) had severe side effects. There was no<br />

relation between R rate (RR) and tolerance. 76.5% <strong>of</strong> pts responding to<br />

treatment had a CC/CT �-arrestin 2 genotype and this was associated with a<br />

better RR (CC/CT R � 46.4% vs. TT R � 25% p � 0.16). The 2 pts having<br />

severe side effects belonged to CC/CT group. COMT genotypes AA/GA were<br />

associated with a higher RR (83.3%pts) and better tolerance (100% pts<br />

with no toxicities). OPRM1 genotype GG was associated with both treatment<br />

failure (0% responded) and worse tolerance (any toxicity � 100%;<br />

severe side effects � 33%). Only 2,4% pts in the AA/GA genotype<br />

experienced some toxicity (p � 0.041). Although STAT6 genotypes were<br />

not associated with RR, tolerance was better in those pts with an AA or GA<br />

genotype with 97.5% pts showing either mild or no side effects (p �<br />

0.007). Conclusions: Common variants in �-arrestin 2, COMT, OPRM1 and<br />

STAT6 genes have shown to predict for opioid-associated response and side<br />

effects in oncologic patients. Further studies are needed to assess factors<br />

determining the variability in response and tolerance to opioid treatment.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


9130 General Poster Session (Board #49D), Sat, 8:00 AM-12:00 PM<br />

Cognitive symptoms in non-Hodgkin lymphoma survivors: Prevalence and<br />

discussion with doctors. Presenting Author: Neeraj K Arora, National<br />

Cancer Institute, Bethesda, MD<br />

Background: Several studies have documented the negative impact <strong>of</strong><br />

chemotherapy on patients’ cognitive functioning. However, this concern<br />

has been understudied among non-Hodgkin Lymphoma (NHL) survivors.<br />

Methods: We analyzed survey data from a population-based study <strong>of</strong><br />

survivors diagnosed with aggressive NHL in Los Angeles County 2-5 years<br />

prior to the study; data were collected between 2003-2005. We assessed<br />

prevalence <strong>of</strong> cognitive symptoms (CS) by a single item screener that asked<br />

if in the past 6 months survivors experienced problems with memory,<br />

attention, or concentration; if yes, we asked if they discussed the problem<br />

with a doctor. Frequency <strong>of</strong> CS was assessed by a 4 item scale that<br />

measured how <strong>of</strong>ten survivors experienced different CS in the past 4 weeks<br />

(none, a little, some, most, all the time). Logistic regression analyses<br />

identified correlates <strong>of</strong> reporting CS (yes/no) as well as discussion with a<br />

doctor (yes/no). Analyses were based on data from 358 survivors who had<br />

received chemotherapy and saw a doctor for follow-up care in the past year.<br />

Results: 134/358 survivors (37%) reported CS, many reporting difficulty<br />

some, most, or all the time with memory (64%), attention (58%),<br />

concentration (57%), and problem solving (46%). Survivors with higher<br />

education, who were not married/partnered, and who had more comorbidities<br />

were more likely to report CS (p�0.05 for all). Report <strong>of</strong> CS was not<br />

associated with age, NHL grade, time since completion <strong>of</strong> chemotherapy,<br />

or having been treated with radiation or bone marrow/stem cell transplant.<br />

Of the 134 survivors who reported CS, only 43% discussed them with a<br />

doctor. Survivors who reported being given information about late effects <strong>of</strong><br />

treatment were more likely to discuss CS than those who were not (OR: 3.4,<br />

95% CI 1.4-8.5, p�0.01). Conclusions: One in three NHL survivors<br />

reported CS following chemotherapy; however, a majority did not discuss<br />

these with a doctor. Periodic screening for CS in NHL survivors may be<br />

needed and would provide an opportunity to improve patient-clinician<br />

communication about this issue. Additionally, research that identifies<br />

mechanisms that cause CS in this population is warranted in order to<br />

develop targeted behavioral and medical interventions.<br />

9132 General Poster Session (Board #49F), Sat, 8:00 AM-12:00 PM<br />

Addressing spirituality within the care <strong>of</strong> patients at the end <strong>of</strong> life:<br />

Perspectives <strong>of</strong> advanced cancer patients, oncologists, and oncology<br />

nurses. Presenting Author: Andrea C. Phelps, Dana-Farber Cancer Institute,<br />

Department <strong>of</strong> Medical Oncology, Boston, MA<br />

Background: Attention to patients’ religious and spiritual needs is included<br />

in national guidelines for quality end-<strong>of</strong>-life care, but little data exists to<br />

guide spiritual care. Methods: The Religion and Spirituality in Cancer Care<br />

Study is a multi-institution, quantitative-qualitative study <strong>of</strong> 75 advanced<br />

cancer patients and 339 cancer doctors and nurses. Patients underwent<br />

semi-structured interviews and providers completed a web-based survey<br />

exploring their perspectives on the routine provision <strong>of</strong> spiritual care by<br />

doctors and nurses. Theme extraction was performed following triangulated<br />

procedures <strong>of</strong> interdisciplinary analysis. Multivariable ordinal logistic<br />

regression models assessed relationships between participant characteristics<br />

and attitudes toward spiritual care. Results: The majority <strong>of</strong> patients<br />

(77.9%), physicians (71.6%), and nurses (85.1%) believed that routine<br />

spiritual care would positively impact patients. Only 25% <strong>of</strong> patients had<br />

previously received spiritual care. Among patients, prior spiritual care<br />

(adjusted odds ratio [AOR], 14.65; 95% CI, 1.51-142.23), increasing<br />

education (AOR, 1.26; 95% CI, 1.06-1.49) and religious coping (AOR,<br />

4.79; 95% CI, 1.40-16.42) were associated with favorable perceptions <strong>of</strong><br />

spiritual care. Physicians held more negative perceptions <strong>of</strong> spiritual care<br />

than patients (p � 0.001) and nurses (p � 0.008). Qualitative analysis<br />

identified benefits <strong>of</strong> spiritual care, including supporting patients’ emotional<br />

wellbeing and strengthening patient-provider relationships. Objections<br />

to spiritual care frequently related to pr<strong>of</strong>essional role conflicts.<br />

<strong>Part</strong>icipants described ideal spiritual care to be individualized, voluntary,<br />

inclusive <strong>of</strong> chaplains/clergy, and based upon assessing and supporting<br />

patient spirituality. Conclusions: Most advanced cancer patients, oncologists,<br />

and oncology nurses value spiritual care. Themes described provide<br />

an empiric basis for engaging spiritual issues within clinical care.<br />

Patient and Survivor Care<br />

599s<br />

9131 General Poster Session (Board #49E), Sat, 8:00 AM-12:00 PM<br />

Prevalence <strong>of</strong> cancer-related fatigue in a population-based sample <strong>of</strong><br />

colorectal, breast, and prostate cancer survivors. Presenting Author:<br />

Jennifer M. Jones, University Health Network, Princess Margaret Hospital,<br />

Toronto, ON, Canada<br />

Background: Cancer-related fatigue (CRF) is the most prevalent and<br />

distressing cancer-related symptom and has a greater negative impact on<br />

patients’ daily activities and quality <strong>of</strong> life than other cancer-related<br />

symptoms, including pain and depression. However, the prevalence and<br />

severity <strong>of</strong> persistent CRF and related disability in the post-treatment<br />

survivorship period has seldom been examined in populations other than<br />

breast cancer. The primary objective <strong>of</strong> the study was to describe the<br />

prevalence <strong>of</strong> significant CRF and associated levels <strong>of</strong> disability in a mixed<br />

cancer population sample at 3 time points in the post-treatment survivorship<br />

trajectory. Methods: Based on cancer registry data, a self-administered<br />

mail based questionnaire using Dillman’s Tailored Design Method was sent<br />

to 3 cohorts <strong>of</strong> disease-free cancer survivors (6-18 months; 2-3 years; and<br />

5-6 years post-treatment) previously treated for non-metastatic breast,<br />

prostate or colorectal cancer. Fatigue was measured using the FACT-F and<br />

disability was measured with the WHO-Disability Assessment Schedule.<br />

<strong>Clinical</strong> information was extracted from chart review. Frequencies <strong>of</strong><br />

significant fatigue by disease sites and time points were studied and<br />

compared using chi-square test. Disability between those with and without<br />

CRF was also compared using Cochran-Armitage trend test. Results: 1294<br />

questionnaire packages were completed (63% response rate). The FACT-F<br />

score was 39.1�10.9; 29% (95% CI: [27%, 32%]) <strong>of</strong> the sample reported<br />

significant fatigue (FACT-F�34) and this was associated with much higher<br />

levels <strong>of</strong> disability (p�0.0001). Breast (40% [35%, 44%]) and colorectal<br />

(33% [27%, 38%]) survivors had significantly higher rates <strong>of</strong> fatigue (�34)<br />

compared to the prostate group (17% [14%, 21%]) (p�0.0001). Fatigue<br />

levels remained relatively stable across the 3 time points. Conclusions: CRF<br />

was a significant and debilitating symptom for a substantial minority <strong>of</strong> the<br />

respondents across all 3 time points. Effective CRF management strategies<br />

are needed and have the potential to significantly reduce morbidity<br />

associated with cancer and its treatments and to improve quality <strong>of</strong> life for<br />

the growing population <strong>of</strong> cancer survivors.<br />

9133 General Poster Session (Board #49G), Sat, 8:00 AM-12:00 PM<br />

Palonosetron (PALO) plus 1-day versus 3-day dexamethasone (DEX) with or<br />

without aprepitant against delayed nausea (DN): Pooled analysis for 554<br />

women receiving highly emetogenic chemotherapy (HEC). Presenting<br />

Author: Luigi Celio, Department <strong>of</strong> Medical Oncology, Fondazione IRCCS<br />

Istituto Nazionale Tumori, Milan, Italy<br />

Background: Nausea is more difficult to control than vomiting, because the<br />

incidence <strong>of</strong> the symptom continues to rise over the days after chemotherapy.<br />

This post-hoc analysis addressed two questions: 1) Does PALO<br />

plus single-dose DEX result in suboptimal control <strong>of</strong> DN when compared to<br />

the same regimen with additional DEX doses? 2) Is the combination <strong>of</strong><br />

PALO, DEX, and aprepitant a more effective coverage against DN? Methods:<br />

The analysis was based upon outcomes in chemo-naïve women (median age<br />

�52) with solid tumors (89% breast) enrolled in two Phase III and two<br />

Phase II trials <strong>of</strong> HEC (AC combination or cisplatin). Patients received the<br />

following regimens: 1) PALO 0.25 mg plus DEX 8 mg IV on day 1 <strong>of</strong><br />

chemotherapy (1-day regimen, n�243); 2) the same regimen on the day 1<br />

followed by DEX 8 mg orally on days 2 and 3 (3-day regimen, n�207); or<br />

PALO plus DEX plus aprepitant 125 mg on day 1 followed by aprepitant 80<br />

mg on days 2 and 3 (triple regimen, n�104). The pre-specified primary<br />

endpoint was the proportion <strong>of</strong> patients with no DN (days 2-5 after the first<br />

cycle <strong>of</strong> chemotherapy). The two primary comparisons were 1-day vs. 3-day<br />

regimen, and triple vs. 3-day regimen, with statistical significance set to<br />

the 0.025 level. Results: The 1-day to 3-day regimen comparison was not<br />

statistically significant (44% vs. 47.8%; risk difference (RD) � -3.8%,<br />

95% CI (-5.4 to 12.9%), P�0.421 (two-sided chi-square test). The triple<br />

to 3-day regimen comparison was also not significant (57.7% vs. 47.8%),<br />

RD��9.9%, 95% CI (-1.9 to 21.3%), P�0.101. More patients receiving<br />

triple regimen experienced no acute-onset nausea compared with those<br />

administered only DEX (97.1% vs. 51.2%, P�0.0001). Conclusions: This<br />

analysis provides evidence that PALO plus 1-day or 3-day DEX yield similar<br />

prevention <strong>of</strong> DN. The addition <strong>of</strong> aprepitant does not improve the control <strong>of</strong><br />

DN. Complete results utilizing a multivariable model accounting for risk<br />

factors and trial heterogeneity will also be presented.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


600s Patient and Survivor Care<br />

9134 General Poster Session (Board #49H), Sat, 8:00 AM-12:00 PM<br />

Longitudinal study <strong>of</strong> hope, meaning/peace (M/P), and quality <strong>of</strong> life (QOL)<br />

in patients (pts) with ovarian cancer (OC). Presenting Author: Lois M.<br />

Ramondetta, University <strong>of</strong> Texas M. D. Anderson Cancer Center, Houston,<br />

TX<br />

Background: M/P may be the most valued end point <strong>of</strong> life. How hope, faith,<br />

physical and psychological factors impact M/P is unknown. We evaluated<br />

factors affecting M/P in pts with OC. Methods: OC pts at a cancer center<br />

(CC), academic hospital (AH) and county hospital (CH for primarily<br />

uninsured) participated. Surveys completed at initiation <strong>of</strong> chemotherapy<br />

(CTX); completion <strong>of</strong> CTX; and 1 yr later. Surveys included FACT-O, -SP,<br />

Herth Hope Index, Hospital Anxiety and Depression Scale (HADS), ESAS,<br />

and Locus <strong>of</strong> Control (LOC). Results: N�115. Median age�55 yrs, married<br />

64%, Christian 96%. CH had more AA/Hispanics pts (p�.001) and<br />

unmarried pts (p�.001). QOL and symptoms improved for all sites over<br />

time (p �.03); CH pts had the worst scores (p� �.001). CC pts expressed<br />

more hope, less anxiety and depression (A/D) compared to CH and AC pts<br />

for all time points (p�.03). CH pts had higher and increasing A/D over time<br />

while CC pts had the least (p�.02). LOC scores differed by site (p�.01).<br />

CH pts held strongest belief that life was controlled by chance and<br />

“others”; CC pts had the least. There was no association between site/time<br />

for belief <strong>of</strong> internal control over one’s life. CH pts consistently had the<br />

lowest M/P scores (p�004). Adjusting for site, disease status and time,<br />

higher M/P associated with higher hope, better QOL, symptoms and faith<br />

(p� �.0001). Lower M/P associated with increased A/D (p�.003) and<br />

symptoms (p�.0001). Poorer M/P over time correlated with belief that life<br />

was controlled by chance (p�.01) and �powerful others�(p�.02). Level <strong>of</strong><br />

M/P did not correlate with belief <strong>of</strong> internal control over one’s life.<br />

Conclusions: M/P did not change over time. CC pts had highest M/P. Higher<br />

M/P associated with higher hope and faith, better QOL, less symptoms and<br />

A/D. Lower M/P associated with sense that life is controlled by chance and<br />

powerful others. Data show medically underserved pts have poorer QOL,<br />

more symptoms and A/D and may believe the future is determined by<br />

luck/chance and by “others”. Triaging for spiritual crisis may be important<br />

in these pts. Interventions to decrease A/D and symptoms may improve pts’<br />

sense <strong>of</strong> M/P over the cancer journey.<br />

9136 General Poster Session (Board #50B), Sat, 8:00 AM-12:00 PM<br />

Associations between receipt <strong>of</strong> a treatment summary, emotional concerns,<br />

and patterns <strong>of</strong> care among post-treatment cancer survivors. Presenting<br />

Author: Ruth Rechis, LIVESTRONG, Austin, TX<br />

Background: Treatment summaries (TS), a critical component <strong>of</strong> survivorship<br />

care plans, were identified as a tool to improve long-term outcomes for<br />

the 12 million cancer survivors alive in the US. Methods: In 2010, the<br />

Lance Armstrong Foundation fielded the LIVESTRONG Survey for People<br />

Affected by Cancer. Respondents were recruited through several channels<br />

including partnerships with national organizations such as ASCO. Over a 9<br />

month period, more than people completed the survey, including 3,682<br />

post-treatment cancer survivors (PTCS). The survey addressed posttreatment<br />

concerns including receipt <strong>of</strong> TS. Full survey results were<br />

presented at the 2011 ASCO Conference. Results: Receipt <strong>of</strong> TS data was<br />

available for 3042 PTCS: average age (50); female (65%); average time<br />

since diagnosis (6 years); received a TS (34%). PTCS who received TS<br />

reported that they were: Closer to time since diagnosis or since treatment<br />

ended (p�0.01); more likely to have received chemotherapy (p�0.01);<br />

more <strong>of</strong>ten receiving the majority <strong>of</strong> their health care from a medical<br />

oncologist (p�0.05); experiencing significantly fewer (p�0.05) posttreatment<br />

emotional concerns (including emotional distress; fears <strong>of</strong><br />

recurrence; concerns about family risk; and appearance concerns) and<br />

were more likely to have received care; significantly less likely to say that<br />

they had “learned to live with” their concerns (p�0.05) – the most<br />

common reason among participants for not receiving care. Finally, receipt<br />

<strong>of</strong> a TS was related to higher information efficacy (p�0.01; which appeared<br />

to mediate the relationship between receipt <strong>of</strong> a TS and fewer emotional<br />

concerns). PTCS who received a TS more <strong>of</strong>ten reported that their needs<br />

were met including information received about possible late-effects; care<br />

they got during treatment; and care they received after treatment.<br />

Conclusions: These results support the provision <strong>of</strong> TS to PTCS. Receipt <strong>of</strong><br />

TS was associated with a variety <strong>of</strong> positive outcomes; however, only about<br />

one-third <strong>of</strong> PTCS received one. Future studies focused on patient<br />

perspectives on care planning tools, such as treatment summaries and care<br />

plans, can help to improve optimal survivorship care delivery.<br />

9135 General Poster Session (Board #50A), Sat, 8:00 AM-12:00 PM<br />

Prospective cohort study <strong>of</strong> chemotherapy-associated toxicity and supportive<br />

care in oncology practice. Presenting Author: Eva Culakova, Duke<br />

University, Durham, NC<br />

Background: Neutropenic complications remain important dose-limiting<br />

toxicities <strong>of</strong> cancer chemotherapy associated with considerable morbidity,<br />

mortality and cost. The risk <strong>of</strong> the initial neutropenic event is greatest in the<br />

first cycle when most patients are receiving full dose chemotherapy.<br />

Methods: A prospective cohort study <strong>of</strong> adult patients with solid tumors or<br />

lymphoma receiving a new chemotherapy regimen was conducted at 115<br />

U.S. practice sites between 2002 and 2006. Chemotherapy-associated<br />

toxicities were captured in up to 4 cycles including severe neutropenia<br />

(SN), febrile neutropenia (FN), and infection. Documented interventions<br />

included colony-stimulating factor (CSF) and antibiotics use and reductions<br />

in chemotherapy relative dose intensity (RDI). Results: Results are<br />

available on 3,638 patients starting chemotherapy <strong>of</strong> which 3,301, 2,937,<br />

and 2,199 went on to cycles 2, 3, and 4, respectively. The majority <strong>of</strong><br />

neutropenic and infection events occurred in cycle 1. A significant inverse<br />

relationship was observed between subsequent reductions in neutropenic<br />

and infectious events and efforts to reduce these complications (Table).<br />

Most patients with stage 4 solid tumors underwent reductions in chemotherapy<br />

RDI. Patients with lymphoma and stage 1-3 solid tumors were less<br />

likely to undergo dose reductions with half or more receiving prophylactic<br />

CSFs during treatment. Approximately 15% <strong>of</strong> patients also received<br />

prophylactic antibiotics. Conclusions: While the risk <strong>of</strong> neutropenic complications<br />

remains greatest during the initial cycle <strong>of</strong> chemotherapy, clinician<br />

efforts to reduce the risk <strong>of</strong> these events vary with cancer type and stage.<br />

Reduced dose intensity is evident in two-thirds <strong>of</strong> patients with advanced<br />

solid tumors whereas the CSFs are employed in half to two-thirds <strong>of</strong><br />

patients with early stage solid tumors or lymphoma.<br />

Percent <strong>of</strong> patients (%)<br />

Cycle-specific events Cumulative events<br />

Solid tumor Solid tumor<br />

Measure<br />

1 2 3 4 Lymphoma stage 1-3 stage 4<br />

FN 6.4 3.8 2.9 0.8 13.7 11.9 7.8<br />

SN or FN 20.0 14.4 13.5 14.3 37.3 33.4 20.6<br />

Infection 12.6 9.4 8.6 1.7 27.5 23.6 21.1<br />

Proph CSF 21.2 38.9 47.9 53.7 63.5 49.5 35.2<br />

Proph antibiotics 4.8 9.5 12.7 15.6 18.2 14.4 11.7<br />

RDI < 85% 25.0 34.6 34.3 36.8 48.4 43.3 60.3<br />

9137 General Poster Session (Board #50C), Sat, 8:00 AM-12:00 PM<br />

Outpatient chemotherapy supportive care: Trial <strong>of</strong> an IT-integrated, NPdelivered<br />

system for unrelieved symptoms. Presenting Author: Kathi<br />

Mooney, University <strong>of</strong> Utah, Salt Lake City, UT<br />

Background: We tested an automated computer based remote monitoring<br />

system paired with nurse practitioner (NP) follow up using a case<br />

management system to address unrelieved symptoms. Methods: Prospectively<br />

336 patients beginning a course <strong>of</strong> chemotherapy were randomized<br />

to the Telephone Care NP (TC) intervention (n 174) or usual care (UC) (n<br />

162). All called daily reporting presence, severity, and distress (0-10 scale)<br />

for 11 common symptoms. Those in the TC intervention also received<br />

automated tailored symptom self-care messages and, based on automated<br />

alerts for unrelieved symptoms at moderate or higher levels, NP calls to<br />

further treat symptoms utilizing national guidelines. Results: There were no<br />

differences between groups on any demographics: 84% White, 56 years<br />

old, female (77%), breast (45%) or lung (17%) cancer. Average study days<br />

were 73 with 87% call completion. Prevalence <strong>of</strong> participants reporting<br />

moderate to severe symptoms were fatigue (86%), pain (80%), sleep<br />

(78%), nausea (60%), depressed mood (52%), anxious (49%), trouble<br />

thinking (48%), numbness (43%), sore mouth (38%), diarrhea (38%), and<br />

appearance concerns (35%). Mixed modeling with intention to treat was<br />

used to compare overall symptom scores by treatment condition (TC/UC)<br />

while accounting for individuals. Results indicate the TC group mean<br />

symptom score was significantly lower than UC (mean difference � .30, p<br />

� .001). Also, each symptom was significantly lower for the TC group<br />

except for diarrhea. Poisson regression showed TC had lower Severe days<br />

than UC (est. means and SE) 3.16 (0.44) vs 10.24 (1.84), p � .001; and<br />

lower Moderate days 8.91 (1.04) vs 19.06 (2.22), p � .001. TC had<br />

somewhat higher Mild days than UC 19.85 (2.81) vs 13.75 (1.85), p �<br />

.06; and more No symptom days 66.06 (3.82) vs 52.02 (4.15), p � .01.<br />

Mixed modeling was used to explore TC intervention impact following NP<br />

calls for alerts. TC reduced symptom scores compared to UC over a4day<br />

period (mean difference � 1.28, p� .001). Conclusions: Remote telephone<br />

monitoring <strong>of</strong> symptoms after chemotherapy with nurse practitioner follow<br />

up on moderate and severe symptoms results in decreased symptom<br />

severity, distress, fewer severe and moderate days and more no symptom<br />

days.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


9138 General Poster Session (Board #50D), Sat, 8:00 AM-12:00 PM<br />

Prospective cohort study <strong>of</strong> chemotherapy-induced alopecia with or without<br />

scalp cooling. Presenting Author: Julie Lemieux, URESP, Centre de<br />

Recherche FRSQ du CHA Universitaire de Québec, Quebec City, QC,<br />

Canada<br />

Background: Scalp cooling can prevent chemotherapy-induced alopecia.<br />

Success varies according to the type <strong>of</strong> chemotherapy. A controversy exists<br />

regarding the use <strong>of</strong> scalp cooling because <strong>of</strong> the lack <strong>of</strong> efficacy data with<br />

modern chemotherapy regimen and safety data. We present a prospective<br />

cohort study design to measure alopecia. Methods: The prospective study<br />

was conducted at the Centre des Maladies du Sein Deschênes-Fabia (CMS)<br />

in Quebec City (where scalp cooling is <strong>of</strong>fered routinely and 85% <strong>of</strong> women<br />

use it) and at the Centre Hospitalier Universitaire de Montreal (CHUM), in<br />

Montreal (where scalp cooling is not available). Women were eligible if they<br />

were going to receive neoadjuvant or adjuvant chemotherapy for breast<br />

cancer. The study involved completion <strong>of</strong> questionnaires (on degree <strong>of</strong><br />

alopecia, hair type, hair care, use <strong>of</strong> head accessories, tolerance to scalp<br />

cooling and questions related to sick leave from work) and having pictures<br />

taken at baseline, cycle 3 and at the last cycle <strong>of</strong> chemotherapy. For the last<br />

43 patients, the EORTC QLQ-C30/BR23 and an adaptation for Hairdex<br />

were added. Results: A total <strong>of</strong> 136 patients were recruited (110 at CMS<br />

over 2 years and 26 at the CHUM over a 9-month period). Preliminary<br />

efficacy results are shown in the Table (data on quality <strong>of</strong> life have not yet<br />

been analyzed). Hair preservation was defined as a SUCCESS for hair loss<br />

(since the beginning <strong>of</strong> chemotherapy) characterized as “not at all”, “a<br />

little” or “moderate” and “FAILURE” when characterized as “a lot” or “all”<br />

or “hair shaved”. Overall, in the scalp cooling group, 34% were considered<br />

a “success” using hairdresser evaluation and 49% using patient evaluation;<br />

for the non scalp-cooling group, these rates were 9% and 4%<br />

respectively. Conclusions: Scalp cooling appears to be efficacious for<br />

preventing chemotherapy-induced alopecia in this exploratory cohort study.<br />

Success <strong>of</strong> scalp cooling as assessed at last cycle <strong>of</strong> chemotherapy.<br />

CMS<br />

CHUM<br />

(n�99 evaluable)<br />

(n�23 evaluable)<br />

Hairdresser evaluation<br />

AC 7 (32%) 1 (25%)<br />

TC 14 (45%) 0 (0%)<br />

FEC-D 6 (20%) 0 (0%)<br />

Other 7 (44%) 1 (9%)<br />

Total 34 (34%) 2 (9%)<br />

Patient evaluation N(%) N(%)<br />

AC 8 (36%) 0<br />

TC 19 (61%) 0<br />

FEC-D 12 (40%) 0<br />

Other 9 (56%) 1 (9%)<br />

Total 48 (49%) 1 (4%)<br />

9140 General Poster Session (Board #50F), Sat, 8:00 AM-12:00 PM<br />

Validation <strong>of</strong> the chemotherapy-induced neuropathy assessment scale.<br />

Presenting Author: Sheeba K. Thomas, University <strong>of</strong> Texas M. D. Anderson<br />

Cancer Center, Houston, TX<br />

Background: Many <strong>of</strong> the assessment tools used to assess peripheral<br />

neuropathy in clinical trials focus on painful neuropathy. However, some<br />

chemotherapy agents cause both painful and non-painful peripheral<br />

neuropathy. The Chemotherapy-Induced Neuropathy Assessment Scale<br />

(CINAS) was designed to evaluate the multi-dimensional nature <strong>of</strong> chemotherapy-induced<br />

neuropathy, including non-painful sensory and motor<br />

deficits. This study evaluated the psychometric properties <strong>of</strong> the CINAS in<br />

patients with multiple myeloma (MM) receiving induction chemotherapy<br />

and/or autologous hematopoietic stem cell transplantation. Methods: We<br />

collected demographic and clinical data and administered 2 items measuring<br />

pain and numbness/tingling at its worst (0-10 scale), and the CINAS. A<br />

total CINAS score was computed by summing all 11 CINAS items. For<br />

reliability and validity analyses, we used the highest total CINAS score for<br />

each patient. Validity was assessed by factor analysis and by correlating<br />

CINAS subscales with the pain and numbness/tingling items. Cronbach<br />

alpha reliability coefficients were computed. To test sensitivity, we compared<br />

change in total CINAS score during induction from baseline to last<br />

total CINAS score in a subset <strong>of</strong> patients (N�20) receiving bortezomib or<br />

thalidomide. We hypothesized that such patients would develop neuropathy.<br />

Results: Ninety patients were enrolled. <strong>Part</strong>icipants were primarily<br />

white (90%) and male (63%). Factor analysis revealed 3 underlying<br />

constructs for the CINAS - sensory, motor functioning, and allodynia -<br />

explaining 74% <strong>of</strong> the common variance. Cronbach alphas were 0.84,<br />

0.85, and 0.83 for the sensory, motor functioning, and allodynia subscales,<br />

respectively, while subscale correlations with the numbness/<br />

tingling item were 0.74, 0.56, and 0.52, respectively. Pain was only<br />

modestly correlated with these subscales (0.21, 0.36, 0.29). Total CINAS<br />

scores <strong>of</strong> patients receiving bortezomib or thalidomide induction worsened<br />

significantly (from 3.4 to 11.9, effect size�0.55, p�.02). Conclusions:<br />

The CINAS demonstrated validity, reliability and sensitivity in patients with<br />

MM during and after chemotherapy and can be used to assess the<br />

multidimensional aspects <strong>of</strong> chemotherapy-induced neuropathy.<br />

Patient and Survivor Care<br />

601s<br />

9139 General Poster Session (Board #50E), Sat, 8:00 AM-12:00 PM<br />

Advance care planning (ACP) among hematopoietic cell transplant (HCT)<br />

patients and bereaved caregivers. Presenting Author: Elizabeth T. Loggers,<br />

Fred Hutchinson Cancer Research Center, Seattle, WA<br />

Background: HCT remains a high risk, potentially curative treatment<br />

typically <strong>of</strong>fered to young, otherwise healthy patients with hematologic<br />

malignancies. Given this, patients and caregivers may not have considered<br />

end-<strong>of</strong>-life ACP. This study investigated the effect <strong>of</strong> pre-transplant ACP in<br />

surviving patients or bereaved caregivers. Methods: A retrospective, mixedmethods,<br />

audio-taped telephone survey adapted from validated instruments<br />

was conducted by a trained interviewer with English-speaking HCT<br />

survivors (n�18) and bereaved caregivers (n�11), 6-12 months post-HCT.<br />

Subjects were identified via HCT databases at two high-volume, tertiary<br />

centers between 2001-2003. Analysis included percentages from quantitative<br />

sections and qualitative transcripts coding by 2 investigators, with<br />

differences resolved by consensus. Results: HCT survivors [median age 47<br />

years (range 33-67); 50% college-educated; all white] were interviewed a<br />

median <strong>of</strong> 13 months after HCT for acute leukemia (7), lymphoma (5), or<br />

other (6). Twelve (67%) had discussed mortality risk pre-HCT with the<br />

medical team; <strong>of</strong> these, 83% felt hope was increased or unchanged (I/U)<br />

and 100% felt clinician commitment to transplant was I/U by the<br />

conversation. Regardless <strong>of</strong> whether mortality was discussed, 50% had<br />

living wills and �75% had a formal proxy, yet 67% didn’t discuss mortality<br />

with family members pre-HCT. “Ignorance is bliss” was expressed by 6<br />

survivors while 2 survivors regretted not discussing mortality risk. Bereaved<br />

caregivers [73% spouses; all W] were interviewed a median <strong>of</strong> 10 months<br />

after their loved one’s death, which occurred a median <strong>of</strong> 31 days (range<br />

13-152) post-HCT. 100% were the patient’s proxy. Nine (82%) had<br />

discussed mortality risk pre-HCT with the medical team; <strong>of</strong> these, 78% felt<br />

hope was I/U, 100% felt clinician commitment to transplant was I/U, and<br />

78% discussed EOL preferences with the patient pre-HCT, with 67%<br />

feeling ACP reduced burden. Four expressed appreciation for “the straight<br />

story.” Conclusions: Discussions <strong>of</strong> potential mortality with patients and<br />

caregivers before HCT did not affect hope, and supported confidence in<br />

medical teams.<br />

9141 General Poster Session (Board #50G), Sat, 8:00 AM-12:00 PM<br />

Physical and functional well-being in women with breast cancer taking<br />

gabapentin for hot flashes. Presenting Author: Kavita Dayal Chandwani,<br />

University <strong>of</strong> Rochester Medical Center, Rochester, NY<br />

Background: Hot flashes reduce quality <strong>of</strong> life (QOL) in women with breast<br />

cancer. In a randomized, double-blind, placebo-controlled trial <strong>of</strong> gabapentin<br />

for hot flashes, 900 mg <strong>of</strong> the drug was found to reduce hot flashes in<br />

women with breast cancer; however, their QOL <strong>of</strong> has not been studied. We<br />

conducted secondary analyses to study two domains <strong>of</strong> QOL: physical<br />

(PWB) and functional well-being (FWB) in women in this trial. Methods: A<br />

nationwide sample <strong>of</strong> women with breast cancer and hot flashes � 2/day<br />

was studied at baseline (T1), 4 weeks (T2), and 8 weeks (T3) <strong>of</strong> treatment<br />

with gabapentin 300mg/day (G300) and 900mg/day (G900) in divided<br />

doses, and a matching placebo (PL). PWB and FWB were assessed via<br />

subscales <strong>of</strong> the Functional Assessment <strong>of</strong> Cancer Therapy-Breast. Linear<br />

mixed model analyses <strong>of</strong> PWB and FWB were conducted, adjusted for<br />

demographic and treatment variables. Results: The mean age <strong>of</strong> women<br />

(N�384) was 54.9 years � 8.07 (range: 31-81, 72% �50 years); 76%<br />

were married; 71% had more than high school education; 95% were<br />

Caucasian and 3% African-<strong>American</strong>; 10% were undergoing chemotherapy,<br />

and 9% radiotherapy (RT). PWB showed significant time effect<br />

(p�.0001) and interaction <strong>of</strong> time and treatment (p�.0001). There was an<br />

improvement in PWB in the G300 by T2 with no change at T3, while the<br />

improvement in PL and G900 was more modest at T2 and continued at T3<br />

(PL - T1 20.7, T2 21.2, T3 21.6; G300 - T1 20.9, T2 22.2, T3 22.2; and<br />

G900 - T1 21.2, T2 21.5, T3 21.7). FWB also showed a significant time<br />

effect (p�.001) and interaction <strong>of</strong> time and treatment (p�.002). FWB in<br />

G300 improved by T2, with no additional improvement at T3; it was<br />

unchanged in G900 at T2 with a slight reduction at T3; PL showed a<br />

modest improvement at T2 followed by a modest worsening at T3 (PL - T1<br />

17.8, T2 18.1, T3 17.9; G300 - T1 17.9, T2 18.6, T3 18.6; G900 - T1<br />

18.6, T2 18.6, T3 18.5). Conclusions: As reported previously, G900 is<br />

effective in reducing hot flashes. In this analysis, however, PWB and FWB<br />

showed different patterns <strong>of</strong> change over time between the two gabapentin<br />

and placebo groups with G900 showing minimal effects on PWB and FWB.<br />

Future trials to further study the changes in physical and functional<br />

well-being <strong>of</strong> women with breast cancer taking gabapentin for hot flashes<br />

are needed.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


602s Patient and Survivor Care<br />

9142 General Poster Session (Board #50H), Sat, 8:00 AM-12:00 PM<br />

YOCAS yoga, fatigue, memory difficulty, and quality <strong>of</strong> life: Results from a<br />

URCC CCOP randomized, controlled clinical trial among 358 cancer<br />

survivors. Presenting Author: Michelle Christine Janelsins, University <strong>of</strong><br />

Rochester Medical Center, Rochester, NY<br />

Background: Memory difficulty is a serious problem for many cancer<br />

survivors and can negatively affect quality <strong>of</strong> life (QOL). Little evidence<br />

exists on the effects <strong>of</strong> physical activity (PA) interventions, such as yoga, on<br />

memory in cancer survivors. We previously reported in a nationwide, Phase<br />

III RCT that YOCAS Yoga, implemented over 4 weeks (2 x/wk; 75<br />

min/session), significantly improved fatigue and QOL in cancer survivors.<br />

The purpose <strong>of</strong> this study was to assess the effects <strong>of</strong> YOCAS on memory<br />

and to identify moderating and possible mediating effects <strong>of</strong> memory in<br />

YOCAS improvements <strong>of</strong> fatigue and QOL. Methods: Cancer survivors<br />

between 2-24 months post adjuvant therapy with no participation in yoga<br />

during the previous 3 months were randomized to standard care (SC) or SC<br />

with YOCAS. YOCAS consists <strong>of</strong> breathing exercises, gentle Hatha and<br />

Restorative yoga postures and meditation. This secondary analysis included<br />

participants (N�358, mean age � 54, 96% female, 75% breast<br />

cancer) who provided pre- and post-intervention data on Difficulty Remembering<br />

Things (modified MD Anderson Symptom Inventory with 0 � “Did<br />

not Interfere” to 10 � “Interfered Completely”), fatigue and QOL (FACIT-<br />

F). ANCOVA was used to assess the effects <strong>of</strong> YOCAS on memory at<br />

post-intervention incorporating baseline memory score. Path Modeling was<br />

conducted to assess the moderating and mediating effects <strong>of</strong> memory on<br />

YOCAS changes in fatigue and QOL. Results: ANCOVA revealed a significant<br />

group effect <strong>of</strong> YOCAS on memory at post intervention (mean change: Yoga<br />

- Control � -0.44; Cohen’s D� -0.24; p �0.05). Moderation analyses<br />

revealed that baseline memory difficulty did not affect how much YOCAS<br />

would improve fatigue (p�0.05) or QOL (p�0.05). Mediation analyses<br />

identified changes in memory as a significant partial mediator for improved<br />

fatigue (p�0.05) and improved QOL (p�0.05). Conclusions: YOCAS yoga<br />

significantly reduces perceived memory difficulty in cancer survivors.<br />

Improvements in memory due to YOCAS may be a possible mediator for<br />

improving fatigue and QOL. More elaborate PA RCTs are needed to confirm<br />

these results. Funding: NCIR25CA10618, K07CA120025, and<br />

U10CA37420.<br />

9144 General Poster Session (Board #51B), Sat, 8:00 AM-12:00 PM<br />

Validation <strong>of</strong> the NCI patient-reported outcomes version <strong>of</strong> the common<br />

terminology criteria for adverse events (PRO-CTCAE) in women receiving<br />

treatment for metastatic breast cancer. Presenting Author: Sandra A.<br />

Mitchell, National Cancer Institute, Bethesda, MD<br />

Background: NCI PRO-CTCAE is designed to enhance adverse event<br />

reporting by integrating patients’ self-report <strong>of</strong> the frequency (F), severity<br />

(S) and interference (I) with usual activities <strong>of</strong> 78 symptomatic treatment<br />

toxicities. This study examined the construct validity <strong>of</strong> a subset <strong>of</strong><br />

PRO-CTCAE items in women with metastatic breast cancer (MBC). Methods:<br />

207 women (70% aged 45-59 years; 94% White;71% college-educated)<br />

with HER2� MBC who had received treatment in the past month were<br />

recruited from 6 U.S. breast cancer support groups and completed a web<br />

survey that collected 18 PRO-CTCAE symptoms, and the Rotterdam<br />

Symptom Checklist (RSC). Pairwise concordance among PRO-CTCAE<br />

symptom dimensions was examined using weighted Kappa and Bowker’s<br />

test for symmetry. Results: Respondents were a median <strong>of</strong> 47 months since<br />

MBC diagnosis and 61% rated their health-related quality <strong>of</strong> life (HRQL) as<br />

good to excellent. Symptom prevalence was similar for PRO-CTCAE and<br />

RSC, with respondents more likely to endorse mood disturbance on<br />

PRO-CTCAE (Anxiety/Worry 90%; Sad/Unhappy Feelings 86%) vs. RSC<br />

(Anxiety 63%; Depressed Mood 61%). There was parallel rank-ordering <strong>of</strong><br />

fatigue, anxiety, insomnia, depression, difficulty concentrating and neuropathy<br />

as the symptoms that were most severe, interfered most and caused<br />

the greatest bother. Within PRO-CTCAE, pairwise agreement among F, S<br />

and I was moderate for most symptoms (�w�.42 to .54). Agreement<br />

between F and S was highest for pain, nausea and arm/leg swelling<br />

(�w�.61 to .80), and lowest for anxiety/worry and sad/unhappy feelings<br />

(�w� .27-.37). Except for arm/leg swelling, endorsement patterns by the<br />

dimensions <strong>of</strong> F, S and I were distinct (Bowker’s p all �.002). Across<br />

PRO-CTCAE symptoms, S was consistently higher than I (mean differences<br />

.12 to .72, all p�.01]). Means for S were consistently and significantly<br />

higher for those with impaired versus preserved HRQL, providing evidence<br />

<strong>of</strong> construct validity. Conclusions: This study supports the construct validity<br />

<strong>of</strong> PRO-CTCAE, and suggests that F, S and I dimensions <strong>of</strong>fer nonoverlapping<br />

information relative to 17 /18 PRO-CTCAE symptomatic<br />

toxicities.<br />

9143 General Poster Session (Board #51A), Sat, 8:00 AM-12:00 PM<br />

Changes in the use <strong>of</strong> eythropoetin-stimulating agents (ESAs) in cancer<br />

patients: Before and after FDA safety advisories. Presenting Author: Dana<br />

Stafkey-Mailey, South Carolina College <strong>of</strong> Pharmacy, Columbia, SC<br />

Background: Benefits <strong>of</strong> ESAs in the treatment <strong>of</strong> cancer-induced anemia<br />

have been well documented, most importantly reducing the likelihood <strong>of</strong><br />

patients needing red blood cell transfusions. However, in early 2007, the<br />

FDA issued a series <strong>of</strong> cautions and Black Box Warning (BBW), that ESAs<br />

may enhance tumor progression, increase likelihood <strong>of</strong> a VTE and decrease<br />

patient survival. The objective <strong>of</strong> this study is to elucidate population<br />

patterns and trends in ESA use for cancer patients pre- and post- the BBW<br />

and examine their associated outcomes. Methods: Data for this analysis was<br />

derived from South Carolina Medicaid claims. Patients with a diagnosis <strong>of</strong><br />

Lung Cancer (162.x), Breast Cancer (174.x) or Non-Hodgkin’s Lymphoma<br />

(200.x or 202.x) receiving chemotherapy between January 1, 1994 and<br />

December 31, 2010 were eligible for inclusion. The main outcome<br />

measures were ESA utilization, blood transfusion, and venous thromboembolism.<br />

The percentage <strong>of</strong> patients experiencing these outcomes were<br />

plotted over time to allow for visual examination <strong>of</strong> changes due to the<br />

BBW. In addition, we used multivariable logistic regression models to<br />

analyze each <strong>of</strong> the associated outcomes with clinical and demographic<br />

variables. Results: Among the 9,280 patients with one <strong>of</strong> the study cancers,<br />

an average <strong>of</strong> 25.4% received an ESA. The proportion <strong>of</strong> patients receiving<br />

an ESA increased from 6.2% in 1994 to a high <strong>of</strong> 47.1% in 2005 at which<br />

time utilization began to decline. In 2010, only 7.8% <strong>of</strong> these patients<br />

received an ESA. Use was associated with older age, female sex, and<br />

receipt <strong>of</strong> radiation therapy. The rate <strong>of</strong> blood transfusion varied sporadically<br />

throughout the study period from 5.2% (2008) to 11.7% (1994)<br />

(p�0.0001). Venous thromboembolisms developed in 12.3% and 8.5% <strong>of</strong><br />

patients receiving and not receiving an ESA, respectively (OR� 1.31;<br />

p�0.001). Conclusions: As the FDA advisory cautioned, we find an<br />

increased risk <strong>of</strong> venous thromboembolisms associated with ESA use. In<br />

contrast to our a priori beliefs initial decline in utilization <strong>of</strong> ESAs began, in<br />

2005, prior to the BBW and was likely due to changes in reimbursement<br />

rates. However, the BBW resulted in the largest decline in ESA use, 19.1<br />

percentage points, between 2007 and 2008.<br />

9145 General Poster Session (Board #51C), Sat, 8:00 AM-12:00 PM<br />

Health behaviors among testicular cancer survivors. Presenting Author:<br />

Steven C. Palmer, University <strong>of</strong> Pennsylvania, Philadelphia, PA<br />

Background: Testicular cancer survivors (TCS) experience elevated risk for<br />

cardiovascular issues including coronary artery disease that present a<br />

greater threat to their long-term health than recurrence <strong>of</strong> cancer. These<br />

risks may be moderated by health behaviors (HBs), yet little is known about<br />

HBs or their barriers in this population. This is unfortunate, as these<br />

barriers may serve as intervention targets. This study examined HBs and<br />

their barriers in a population-based sample <strong>of</strong> TCS. Methods: TCS were<br />

recruited through the PA Cancer Registry. Eligible individuals were diagnosed<br />

with testicular cancer between 1990 - 2007 and provided informed<br />

consent. <strong>Part</strong>icipants completed measures <strong>of</strong> tobacco use, alcohol use,<br />

dietary habits, physical activity, BMI, quality <strong>of</strong> life (QOL), and barriers to<br />

performing HBs, as well as self-reported disease and treatment information.<br />

Results: 189 participants provided data. <strong>Part</strong>icipants were middleaged<br />

(M�44 yrs. R�19-59), predominately white (95%), married (72%),<br />

parents (69%), 6.8 years post-diagnosis (R�1-19), and at normative levels<br />

for physical and psychosocial QOL. A substantial minority (31%) was<br />

unaware <strong>of</strong> their disease stage or histopathological type (27%), although<br />

89% reported surgery, 32% chemotherapy, and 53% XRT. Almost 25%<br />

reported current tobacco use, and 35% reported “risky” alcohol use.<br />

Adequate aerobic activity was reported by half, but only 28% reported<br />

adequate strength and flexibility activities. 84% reported above normal<br />

BMI, with 35% in the obese range. Barriers to HBs included cancer-related<br />

problems (e.g., neuropathy, pain, hernia risk) and competing demands<br />

(e.g., work responsibility, time constraints). Cancer-related barriers were<br />

related to worse physical QOL and higher BMI. Competing demands<br />

predicted worse psychosocial QOL, unhealthy eating, and inadequate<br />

physical activities. Conclusions: Given the negative HBs in this sample and<br />

their potential cardiovascular risk, interventions aimed at reducing tobacco<br />

and risky alcohol use, as well as improving dietary and physical activity<br />

levels are needed. These interventions will likely need to overcome<br />

perceived barriers to adoption <strong>of</strong> HBs in order to be successful.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


9146 General Poster Session (Board #51D), Sat, 8:00 AM-12:00 PM<br />

Lifetime traumas and their influence on advanced cancer patients’ illness<br />

understanding and likelihood <strong>of</strong> end-<strong>of</strong>-life discussions. Presenting Author:<br />

Elizabeth L. Kacel, Dana-Farber Cancer Institute, Boston, MA<br />

Background: Research on posttraumatic stress in cancer patients has<br />

focused on patient reactions to their cancer diagnosis. Few studies have<br />

examined the impact <strong>of</strong> lifetime traumatic events prior to diagnosis on how<br />

advanced cancer patients understand the life-threatening nature <strong>of</strong> their<br />

illness and how likely they are to discuss their end-<strong>of</strong>-life (EOL) wishes with<br />

their oncology providers. Methods: The Coping with Cancer (CwC) is an<br />

NCI-funded prospective, multi-institutional cohort study <strong>of</strong> advanced<br />

cancer patients and their caregivers. <strong>Part</strong>icipants were recruited from<br />

September 2002 to February 2008 from six comprehensive cancer centers<br />

across the United States. Lifetime traumas were captured by the number <strong>of</strong><br />

events reported in response to the Structured <strong>Clinical</strong> Interview for the<br />

DSM-IV (SCID) Lifetime Posttraumatic Stress Disorder question that<br />

probed for “extremely upsetting” life events. Associations between patient<br />

characteristics and number <strong>of</strong> lifetime traumas were estimated as odds<br />

ratios using ordinal logistic regression. Associations between number <strong>of</strong><br />

lifetime traumatic exposures, controlling for confounding patient characteristics,<br />

and patient Terminal Illness Acknowledgement (TIA) and reports <strong>of</strong><br />

EOL discussions were estimated as odds ratios using multiple logistic<br />

regression. Results: After adjusting for race, education, and recruitment<br />

site, the number <strong>of</strong> lifetime traumas patients reported remained significantly<br />

associated with TIA (OR � 1.25, p � .034) and discussion <strong>of</strong> EOL<br />

wishes (OR � 1.29, p � .013). Conclusions: The greater the number <strong>of</strong><br />

traumatic experiences reported by cancer patients the more likely they are<br />

to acknowledge that they are terminally ill and discuss their end-<strong>of</strong>-life<br />

wishes with their oncology providers. The impact <strong>of</strong> witnessing or experiencing<br />

serious or life-threatening events in the past appears to improve<br />

advanced cancer patients’ abilities to understand the seriousness <strong>of</strong> their<br />

condition and increases the likelihood that they will discuss their preferences<br />

for care at the end <strong>of</strong> life with their doctors.<br />

TPS9148 General Poster Session (Board #51F), Sat, 8:00 AM-12:00 PM<br />

Prospective and working memory decline as a result <strong>of</strong> impaired sleep<br />

and/or impared blood glucose in breast cancer patients during chemotherapy<br />

treatment. Presenting Author: Liliana Moyers-Ruiz, Bournemouth<br />

University/ Royal Bournemouth Hospital, Bournemouth, United Kingdom<br />

Background: Breast cancer patients might develop impaired sleep patterns<br />

during chemotherapy, and impaired cognitive function has been associated<br />

with sleep patterns disruption. It has been suggested that sleepiness (sleep<br />

tendency) in cancer patients is prevalent and causes distress in the<br />

patient’s quality <strong>of</strong> life, that and their cognitive abilities are also affected.<br />

Furthermore, increased blood glucose levels among women undergoing<br />

chemotherapy may contrubute to memory deficits in breast cancer patients.<br />

It is proposed that Prospective and Working Memory are being<br />

affected by the disturbance <strong>of</strong> these biological factors over the course <strong>of</strong><br />

chemotherapy. Methods: Ninety participants will be invited to take part in<br />

the study. Thirty will be breast cancer patients receiving chemotherapy<br />

treatment, thirty breast cancer patients receiving treatment other than<br />

chemotheray, and thirty healthy controls. Measures <strong>of</strong> sleep and sleepiness,<br />

will be administered at 4 testing sessions during a 9-month period,<br />

along with a neuropsychological battery. Additionally, blood samples will<br />

be analysed. Eight <strong>of</strong> planned 60 cancer patients have been enrolled, and<br />

four <strong>of</strong> them have completed the second testing session. For the assessment<br />

<strong>of</strong> sleep/wake activity, participants will be required to wear a sleeping<br />

monitor (sensewear armband) for 7 days. Pupil dilatation to observe<br />

sleepiness will be meassured by using a Pupillometry developed at Poole<br />

hospital in the UK. Prospective Memory will be assessed using CAMPROMT<br />

test to analyse if this is a more sensitive measure for mild cognitive<br />

impairment. Discussion: Impairment on variables such as sleep, sleepiness<br />

and glucose have not been studied within the context <strong>of</strong> Chemo-Brain on<br />

breast cancer patients.Therefore we want to observe whether impaired<br />

sleep and sleepiness, and impaired glucose levels are at the core <strong>of</strong><br />

Prospective and Working Memory decline in Chemo-Brain patients in order<br />

to find a cause-and-effect relationship as a consequence <strong>of</strong> chemotherapy.<br />

Patient and Survivor Care<br />

603s<br />

TPS9147 General Poster Session (Board #51E), Sat, 8:00 AM-12:00 PM<br />

Using cancer patient stories to “power” communication modules. Presenting<br />

Author: Forrest Lang, East Tennessee State University, Johnson City,<br />

TN<br />

Background: Caring for patients with cancer poses great challenges to<br />

doctors’ communication skills. This project, funded by NIH-NCI, represents<br />

an innovative collaboration between ETSU faculty in medical oncology,<br />

family medicine, and storytelling. The team secures patients’ and<br />

caregivers’ cooperation in recording stories <strong>of</strong> their journeys with illness.<br />

These are the focus <strong>of</strong> a set <strong>of</strong> cancer communication modules, to be<br />

<strong>of</strong>fered as educational experiences for medical students, residents, and<br />

oncology fellows. The modules in development address 1) breaking bad<br />

news, 2) living through treatment, 3) transitioning from curative to<br />

palliative care, 4) communicating with family, and 5) sensitivity to issues <strong>of</strong><br />

religion and spirituality. Methods: A collaborative inter-pr<strong>of</strong>essional team<br />

developed an interview protocol to facilitate sharing <strong>of</strong> cancer-themed<br />

narratives. Video records are transcribed and coded using N-Vivo 8. The<br />

rating team meets to identify video clips that speak powerfully to positive,<br />

negative, or ambivalent aspects <strong>of</strong> cancer communication. Selected patients<br />

are brought together into “story circles,” where additional narratives<br />

are gathered. The module development team uses these stories to create<br />

empathic involvement in viewers, and to sensitize them to effective and<br />

ineffective communication strategies and challenges surrounding critical<br />

moments in patients’ lived experience <strong>of</strong> cancer. Modules effectiveness is<br />

tested with 1) Family Medicine and Internal Medicine residents, 2) medical<br />

oncology fellows and 3) multi-pr<strong>of</strong>essionals health students just completing<br />

a communication courses. Assessment includes a pre-test and post-test<br />

OSCE addressing a number <strong>of</strong> the challenging cancer communication<br />

moments. Eighty-four <strong>of</strong> a projected 100 patient interviews and twelve<br />

physician//faculty interviews have been recorded, and all have agreed to the<br />

use <strong>of</strong> their interview materials. Representative examples <strong>of</strong> recorded<br />

cancer stories will be presented to demonstrate their evocative and<br />

pedagogical value. Opportunities will be discussed for further uses <strong>of</strong> these<br />

and similar stories in collaborations between medicine and the arts and<br />

humanities.<br />

TPS9149^ General Poster Session (Board #51G), Sat, 8:00 AM-12:00 PM<br />

CATCH: A randomized trial comparing tinzaparin versus warfarin for<br />

treatment <strong>of</strong> acute venous thromboembolism (VTE) in cancer patients.<br />

Presenting Author: Agnes Y. Lee, University <strong>of</strong> British Columbia, Vancouver,<br />

BC, Canada<br />

Background: VTE is a major cause <strong>of</strong> morbidity and mortality in cancer<br />

patients. LMWHs have been shown to be superior to warfarin in one<br />

randomized study, but adequately powered confirmatory studies have not<br />

been conducted and warfarin continues to be widely used for treatment <strong>of</strong><br />

cancer-associated VTE. Methods: We are conducting an open-label, randomized<br />

trial <strong>of</strong> tinzaparin versus warfarin in 900 patients with active cancer<br />

and symptomatic proximal deep vein thrombosis (DVT) and/or pulmonary<br />

embolism (PE). Tinzaparin is given at full treatment doses (175 IU/kg once<br />

daily) for 6 months in the experimental arm and initial tinzaparin treatment<br />

for 5-10 days followed by dose-adjusted warfarin (target INR 2.0-3.0) is<br />

given for 6 months in the control arm. The primary composite outcome is<br />

time to recurrent VTE event, including incidentally diagnosed VTE and fatal<br />

PE. Baseline characteristics will be analysed for their ability to predict the<br />

risk for recurrent VTE or bleeding. In particular, the parameters <strong>of</strong> the<br />

Khorana scale and Wells rule will be tested for their usefulness in<br />

predicting recurrent VTE. Predictive biomarkers will be tested including<br />

D-dimer and Tissue Factor. Assessment <strong>of</strong> post-thrombotic syndrome<br />

(PTS), quality <strong>of</strong> life and healthcare resource utilization will also be<br />

performed. The trial is recruiting in � 160 sites in �25 countries in 4<br />

continents (NCT01130025). As <strong>of</strong> Jan 2012, 135 sites were activated for<br />

study enrolment and 228 patients have been enrolled. We anticipate<br />

completion <strong>of</strong> enrolment in Jan 2013. The results obtained from this study<br />

will add significantly to the knowledge on the efficacy, safety and<br />

cost-effectiveness <strong>of</strong> LMWH to prevent recurrent VTE. Important prospective<br />

data on the clinical significance <strong>of</strong> incidental VTE in patients with<br />

active cancer will be generated, and analyses <strong>of</strong> risk stratification parameters<br />

will add important information that may help to further tailor therapy.<br />

The study <strong>of</strong> PTS, which has not previously been done in this selected<br />

patient population, will add to the evidence that tinzaparin significantly<br />

reduces the incidence <strong>of</strong> PTS and leg ulcers (Hull et al, Am J Med<br />

2009;122:762-9).<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


604s Patient and Survivor Care<br />

TPS9150 General Poster Session (Board #51H), Sat, 8:00 AM-12:00 PM<br />

Randomized phase III trial to evaluate radiopharmaceuticals and zoledronic<br />

acid in the palliation <strong>of</strong> osteoblastic metastases from lung, breast,<br />

and prostate cancer: Report <strong>of</strong> RTOG 0517. Presenting Author: Michael J.<br />

Seider, Akron City Hospital, Akron, OH<br />

Background: Skeletal related events (SREs) diminish quality <strong>of</strong> life (QOL) as<br />

well as overall survival (OS) in patients with bone metastases, a common<br />

event in breast, lung and prostate cancer. SREs can be reduced or delayed<br />

by the use <strong>of</strong> bisphosphonates. It is postulated that the radiopharmaceuticals,<br />

Strontium-89 (Sr89) and Samarium-153 (Sm153), when added to a<br />

bisphosphonate can decrease the incidence <strong>of</strong> SREs. Methods: RTOG 0517<br />

randomized patients with breast, lung and prostate cancer and blastic bone<br />

metastases to either Zoledronic acid (ZA) alone or ZA plus a single standard<br />

dose <strong>of</strong> either Sr89 or Sm153. No limitations were placed on additional<br />

therapy such as chemotherapy or hormonal treatment. The projected<br />

median time to SRE [pathological bone fracture, spinal cord compression,<br />

surgery to bone, or radiation to bone] for the ZA arm was 10.4 months<br />

requiring 257 SRE events to detect a 33% relative reduction for the<br />

radiopharmaceutical arm in the time to development <strong>of</strong> an SRE with 90%<br />

power. Other study objectives included quality <strong>of</strong> life, pain control, OS and<br />

toxicity. Results: 261 patients (median age 68; 62% male; 55% prostate,<br />

35% breast, 10% lung) were accrued from July 2006 through February<br />

2011 (4.6 patients/month). Due to a lower than expected rate <strong>of</strong> SREs in<br />

the control (ZA) arm, the study was closed early and therefore did not reach<br />

the targeted accrual. 28 (17.4%) patients in the ZA arm and 27 (16.8%) in<br />

the radiopharmaceutical arm experienced an SRE. Median time to development<br />

<strong>of</strong> an SRE in the ZA and radiopharmaceutical arms was 11.60 and<br />

16.74 months, respectively (p�.47). Median OS in the ZA arm and<br />

radiopharmaceutical arm was 15.95 and 11.18 months, respectively<br />

(p�0.12). Cox proportional hazards regression model showed that baseline<br />

characteristics, including gender, race, ethnicity, primary disease site or<br />

number <strong>of</strong> bone metastases, had no significant impact on OS. There was no<br />

difference in QOL parameters or toxicities between the two arms. Conclusion:<br />

Patients receiving ZA only experienced a much lower SRE rate than<br />

was hypothesized. The addition <strong>of</strong> Sr89 or Sm153 did not result in a<br />

difference in SREs, OS, or QOL<br />

TPS9152 General Poster Session (Board #52B), Sat, 8:00 AM-12:00 PM<br />

Radioprotection study <strong>of</strong> topical norepinephrine in postsurgical breast<br />

cancer patients. Presenting Author: James F. Cleary, University <strong>of</strong> Wisconsin,<br />

Madison, WI<br />

Background: This study derives from radiodermatitis studies in a rat model<br />

evaluating radioprotection from topical norepinephrine application. In the<br />

model, increasing radiation doses induce dermatitis <strong>of</strong> increasing severity,<br />

ranging from mild erythema to wet desquamation. The model was initially<br />

designed to identify topical radioprotectants that function as oxygen free<br />

radical scavengers. In an experiment designed to test the hypothesis that<br />

the co-application <strong>of</strong> a topical vasoconstrictor and an aminothiol scavenger<br />

would limit the systemic uptake <strong>of</strong> the scavenger, it was observed that the<br />

combination was synergistic and that the topical vasoconstrictor was active<br />

when administered alone as a control. Subsequent experiments demonstrated<br />

that topical a-adrenergic receptor agonists are active in preventing<br />

radiodermatitis. The data are consistent with a mechanism based on local<br />

and transient vasoconstriction within the skin, which reduces local tissue<br />

oxygenation. This subsequently limits the formation <strong>of</strong> radiation-induced<br />

reactive oxygen species and protects skin stem cells from radiation<br />

damage. Methods: Two Phase 1 safety studies have been conducted, one in<br />

normal volunteers and one in cancer patients receiving palliative radiation<br />

therapy for bone metastases. This study is a nonrandomized, open-label<br />

safety and exploratory study in post-surgical breast cancer patients treated<br />

with 45 to 50 Gy to the whole breast and axilla. A 10-16 Gy boost to the<br />

lumpectomy region is permitted. Eligible patients are age 18 years or older<br />

with a diagnosis <strong>of</strong> Stage Ia (T1, N0, M0), Stage Ib (T0 or 1, N1mic, M0) or<br />

Stage IIa (T�3cm, N0, M0) infiltrating ductal or lobular carcinoma <strong>of</strong> the<br />

breast or ductal carcinoma in situ (DCIS). The dose is a single daily topical<br />

application <strong>of</strong> 3.0 mL <strong>of</strong> norepinephrine HCl (82.3 mg/mL in 70% ethanol)<br />

applied to the same 50 cm2 treatment site in the axilla about 20 minutes<br />

prior to each radiotherapy fraction. The radiodermatitis severity at the<br />

treatment site will be compared to surrounding (untreated) control areas. 5<br />

<strong>of</strong> planned 12 patients have been enrolled. <strong>Clinical</strong> trial registry number<br />

NCT01263366.<br />

TPS9151 General Poster Session (Board #52A), Sat, 8:00 AM-12:00 PM<br />

Do we need to perform pulmonary function tests in non-small cell lung<br />

cancer patients never diagnosed as COPD? Presenting Author: Narongwit<br />

Nakwan, Prince <strong>of</strong> Songkla University, Hatyai, Thailand<br />

Background: Although chronic obstructive pulmonary disease (COPD) and<br />

lung cancer share a common risk factor, namely smoking, health care<br />

providers usually overlook the symptom <strong>of</strong> COPD in the management <strong>of</strong><br />

lung cancer. Should, then, lung cancer patients undergo pulmonary<br />

function tests (PFT) to identify the presence <strong>of</strong> COPD? Our study was<br />

performed to describe the results <strong>of</strong> pulmonary function tests and define<br />

risk factors for COPD in non-small cell lung cancer (NSCLC) patients.<br />

Methods: A total <strong>of</strong> 31 eligible patients with NSCLC but no obvious<br />

symptoms <strong>of</strong> COPD participated. We collected baseline characteristics and<br />

conducted a detailed assessment <strong>of</strong> pulmonary function, particularly<br />

spirometry, lung volume measurement, and diffusing capacity <strong>of</strong> carbon<br />

monoxide (DLCO). Dyspnea was assessed using modified Borg (mBorg) and<br />

modified Medical Research Council (mMRC) scores. Results: Twelve<br />

patients had airflow limitation (FEV1/FVC�0.7). These patients had mean<br />

percent predicted FEV1, RV and DLCO <strong>of</strong> 52%, 143% and 66% respectively,<br />

and a mean RV/TLC <strong>of</strong> 0.55. Being male, and having a smoking<br />

history, low body mass index and squamous cell carcinoma were significantly<br />

associated with obstruction in univariate analysis. However, obstruction<br />

was not more common in advanced stage than in locally advanced<br />

NSCLC. Neither mBorg nor mMRC differed between obstructive and<br />

non-obstructive groups. Discussion: COPD was found in patients with<br />

NSCLC who had never been diagnosed as, or showed symptoms <strong>of</strong>, COPD.<br />

Male, smoking history, low BMI and squamous cell carcinoma were<br />

significantly associated with obstruction.<br />

TPS9153 General Poster Session (Board #52C), Sat, 8:00 AM-12:00 PM<br />

A multi-site, fixed dose, parallel arm, double-blind, placebo controlled,<br />

block randomised trial <strong>of</strong> the addition <strong>of</strong> infusional octreotide or placebo to<br />

regular ranitidine and dexamethasone for the evaluation <strong>of</strong> vomiting<br />

associated with bowel obstruction at the end <strong>of</strong> life. Presenting Author:<br />

David C. Currow, Flinders University, Adelaide, Australia<br />

Background: Bowel obstruction due to advanced cancer that is surgically<br />

inoperable is a major management problem. Studies to date have either<br />

been underpowered or have used comparators that may not draw on the<br />

best available evidence. Methods: This double-blind, block randomised,<br />

placebo controlled, set dose, parallel arm study was conducted across 12<br />

sites in Australia. Eligibility included inoperable bowel obstruction secondary<br />

to cancer or its treatments. The intervention was the addition <strong>of</strong><br />

infusional octreotide or placebo in addition to 200mg ranitidine per 24<br />

hours parenterally and 4mg per 24 hours parenterally <strong>of</strong> dexamethasone.<br />

The primary outcome measure was the numbers <strong>of</strong> days free <strong>of</strong> vomiting up<br />

to 72 hours after all medications were administered the first time.<br />

<strong>Part</strong>icipants were also administered between 10-20mls per hour <strong>of</strong><br />

subcutaneous isotonic fluid over the 72 hour period. Results: This study<br />

will close to recruitment in March 2012. To date 89 <strong>of</strong> 92 required<br />

participants have been randomised. Conclusions: This adequately powered<br />

study will define the additional net clinical benefit derived from octreotide<br />

over placebo in people who have an anti-secretary agent (ranitidine) and<br />

glucocorticoids (dexamethasone).<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


TPS9154 General Poster Session (Board #52D), Sat, 8:00 AM-12:00 PM<br />

The European InCASA telecare-telehealth electronic platform (ICT-FP7) for<br />

the daily assessment <strong>of</strong> symptoms, weight, and activity in cancer patients<br />

on chronotherapy at home. Presenting Author: Francis Levi, INSERM<br />

U776, Paul Brousse Hospital, Villejuif, France<br />

Background: Self-rated symptoms contribute to the Quality <strong>of</strong> Life (QoL) <strong>of</strong><br />

cancer patients (pts) and impact on prognosis. Behavioural functions are<br />

controlled by the circadian timing system (CTS), through a network <strong>of</strong><br />

molecular clocks that time cellular proliferation and drug metabolism (Lévi<br />

et al. ARPT 2010). Chronotherapy aims at the reduction <strong>of</strong> treatmentrelated<br />

symptoms through the adjustment <strong>of</strong> chemotherapy delivery to the<br />

CTS. Cancer chronotherapy is delivered at home using programmable<br />

pumps, and avoids familial and social disruption. Telemedicine tools can<br />

provide continuous information on symptoms, behavior, QoL and CTS from<br />

non hospitalized pts. Methods: The inCASA platform enables telemonitoring<br />

<strong>of</strong> multiple functions in order to help physicians to detect early warning<br />

signals in pt condition at home, and prompt adequate and timely interventions.<br />

Our pilot site investigates the clinical relevance <strong>of</strong> daily teletransmitted<br />

self assessed symptoms, body weight and rest-activity circadian rhythm<br />

in cancer pts. This system was first well understood and accepted by 14 pts<br />

(9 male, 5 female, 43-77 years) at the outpt clinic, then used by 5 pts (4<br />

male, 1 female, 61�13 years) at home daily for 6� weeks, while receiving<br />

chronotherapy. The pilot phase will accrue 30 pts receiving chronotherapy<br />

at home from 03/01/2012. Pts will fill in the M.D. Anderson Symptom<br />

Inventory scale on a touch-screen device, measure their body weight using<br />

a Bluetooth scale and record their circadian rest-activity rhythm using an<br />

infrared wrist-watch accelerometer over 6-weeks. The data are transmitted<br />

to the platform, then to a web portal with secured access to be daily<br />

inspected by the oncology nursing and medical team. The modelling <strong>of</strong><br />

these continuous records will define alert thresholds for nurse-controlled<br />

graded intervention decisions. Conclusion: The assessment criteria set<br />

forth for the inCASA solution will address the issues <strong>of</strong> pt autonomy, CTS<br />

entrainment in the pt usual environment, safety <strong>of</strong> chronotherapy delivery<br />

at home and treatment costs. Its use should induce important organisational<br />

changes and perception in healthcare pr<strong>of</strong>essionals.<br />

Patient and Survivor Care<br />

605s<br />

TPS9155 General Poster Session (Board #52E), Sat, 8:00 AM-12:00 PM<br />

Assessing clinical appropriateness on a population basis within a regional<br />

cancer network. Presenting Author: Paolo Giovanni Casali, Fondazione<br />

IRCCS Istituto Nazionale dei Tumori, Milan, Italy<br />

Background: The Lombardia Cancer Network (ROL: “Rete Oncologica<br />

Lombarda”) connects all cancer resources <strong>of</strong> the Lombardia region, with<br />

9,000,000-plus citizens, to improve quality <strong>of</strong> cancer care and patient<br />

data sharing. Assessment <strong>of</strong> appropriateness <strong>of</strong> cancer care on a population<br />

basis is an aim. Implementation <strong>of</strong> electronic records is partial and<br />

heterogeneous. However, clinical discharge reports for both inpatients and<br />

oupatients are released as “pdf” files onto a secure regional health care<br />

information system, accessible by physicians. Methods: ROL includes 58<br />

oncology premises. <strong>Clinical</strong> practice guidelines are annually updated<br />

within this oncology community. For each solid cancer, guidelines enlist all<br />

main clinical presentations (“disease phases”), along with their “treatment<br />

options” (either “standard”, “individualized”, or “investigational”). ROL<br />

upgraded the existing non-structured electronic clinical discharge report<br />

into a field-based resource, with both free-text (17) and codified (25) fields.<br />

Two codified fields enlist, respectively, disease phases as per guidelines,<br />

and the corresponding treatment options. If they match, the strategic<br />

medical decision is considered tentatively “appropriate”. Even the report <strong>of</strong><br />

a single encounter within a disease phase is enough to make this work.<br />

Results: Currently, the instrument is being incorporated stepwisely within<br />

the information systems <strong>of</strong> all oncology facilities, though a central<br />

web-based tool is also available. More than 36,000 reports were released in<br />

2011. Appropriateness assessment proved feasible on a pilot set <strong>of</strong><br />

patients. A research project is underway to detect causes <strong>of</strong> mistakes and<br />

mismatches, by automatically analyzing free-text fields. A training effort is<br />

ongoing to improve clinicians’ learning curve on semantics. Conclusions:<br />

To assess clinical appropriateness on a population basis in a large region,<br />

we exploited two key bottlenecks: 1) the electronic discharge report, within<br />

patient information flows, to cope with a variety <strong>of</strong> local information<br />

systems; 2) the disease phase, within the cancer-specific clinical decisionmaking<br />

process, to cope with a likely lack <strong>of</strong> reports from a substantial<br />

proportion <strong>of</strong> encounters.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


606s Pediatric Oncology<br />

9500 Oral Abstract Session, Sat, 1:15 PM-4:15 PM<br />

Efficacy <strong>of</strong> crizotinib in children with relapsed/refractory ALK-driven<br />

tumors including anaplastic large cell lymphoma and neuroblastoma: A<br />

Children’s Oncology Group phase I consortium study. Presenting Author:<br />

Yael P. Mosse, The Children’s Hospital <strong>of</strong> Philadelphia, Philadelphia, PA<br />

Background: Genetic aberrations in the anaplastic lymphoma kinase (ALK)<br />

gene are found in anaplastic large cell lymphoma (ALCL), neuroblastoma<br />

(NB) and other tumors.Crizotinib,a small molecule inhibitor <strong>of</strong> ALK and<br />

c-Met, is active in non-small cell lung cancers (NSCLC) harboring an ALK<br />

translocation. We performed a phase 1 dose-escalation and pharmacokinetic<br />

(PK) trial <strong>of</strong> crizotinib in patients (pts) with refractory solid tumors<br />

and ALCL. Methods: Crizotinib was administered bid without interruption in<br />

28 day cycles using the rolling-six design. Six dose levels (100, 130, 165,<br />

215, 280, 365 mg/m2 /dose) have been evaluated (A1). Pts with confirmed<br />

ALK fusion proteins, mutations or amplification (A2) could enroll at one<br />

dose level lower than part A1 and those with NB could enroll on a separate<br />

stratum (A3). PK studies were performed on day 1 and at steady state (SS).<br />

ALK genomic status in tumor tissue was evaluated and qPCR was used to<br />

measure NPM-ALK fusion transcript in bone marrow and blood samples <strong>of</strong><br />

ALCL pts. Results: 70 pts were enrolled, 57 fully evaluable for toxicity,<br />

[median (range) age 9.9 yrs (1.1–21.3)]: 29 on A1, 18 on A2, and 10 on<br />

A3. In A1, 2/7 pts developed DLT (grade 3 dizziness, grade 5 intra-tumoral<br />

hemorrhage) at 215 mg/m2 and 1/6 pts developed DLT (grade 4 liver<br />

enzyme elevation) at 365 mg/m2 . In A2, 1 grade 4 DLT (neutropenia)<br />

occurred at 165 mg/m2 ; in A3, no DLTs occurred. Mean (�SD) Cave (�AUC0-12h/12h) <strong>of</strong> crizotinib at SS was 466�114 ng/mL at 215 mg/m2 /<br />

dose (n�5), 443�121 ng/mL at 280 mg/m2 /dose (n�8), and 720�230<br />

ng/mL at 365mg/m2 /dose (n�4). Response data for pts with ALCL (six at<br />

165 mg/m2 , two at 280 mg/m2 ) approved for release by the Data Safety<br />

Monitoring Committee demonstrates 7/8 (88%) complete response (CR)<br />

rate to date. RT-PCR data for 6 <strong>of</strong> these pts at 57 time points was obtained<br />

and will be described. In addition, 2 pts with NB have had CRs, one with a<br />

documented ALK mutation. One patient with an inflammatory my<strong>of</strong>ibroblastic<br />

tumor and one with NSCLC had PRs. Conclusions: Inhibition <strong>of</strong> ALK in<br />

pediatric pts with ALK-driven tumors occurs with minimal toxicity and is<br />

associated with objective anti-tumor activity.<br />

9502 Oral Abstract Session, Sat, 1:15 PM-4:15 PM<br />

Stanford V chemotherapy and involved field radiotherapy for children and<br />

adolescents with unfavorable risk Hodgkin lymphoma: Results <strong>of</strong> a multiinstitutional<br />

prospective clinical trial. Presenting Author: Monika Metzger,<br />

University <strong>of</strong> Tennessee Health Science Center, Memphis, TN<br />

Background: To evaluate the efficacy <strong>of</strong> 12 weeks <strong>of</strong> Stanford V chemotherapy<br />

(prednisone, vinblastine, doxorubicine, nitrogen mustard, etoposide,<br />

vincristine, and bleomycin) without routine growth factor support plus<br />

response-adapted low-dose, involved-field radiotherapy (IFRT) in children<br />

and adolescents with unfavorable risk Hodgkin lymphoma (HL). Methods:<br />

Multi-institutional (St. Jude Children’s Research Hospital, Stanford University,<br />

Children’s Hospital Boston, Massachusetts General Hospital and<br />

Maine Children’s Hospital) clinical trial. One hundred forty-one patients<br />

with clinical stages IIB (n�43), IIIB (n�19), IVA (n�27), and IVB (n�52)<br />

HL were treated with 12 weeks <strong>of</strong> Stanford V chemotherapy and low dose<br />

IFRT between June 2002 and May 2011. Involved nodal sites in complete<br />

remission (CR, defined as � 75% shrinkage <strong>of</strong> the original tumor and PET<br />

negative) after 8 weeks <strong>of</strong> Stanford V received 15 Gy IFRT; those sites that<br />

achieved only partial response received 25.5 Gy IFRT after completion <strong>of</strong><br />

all 12 weeks <strong>of</strong> chemotherapy. Results: With a median follow-up <strong>of</strong> 4.6<br />

years, the 3-year overall and event-free survival (EFS) are 97% (SE�2%)<br />

and 79% (SE�4%) respectively. There was no significant difference in EFS<br />

by stage (IIB vs. IIIB vs. IV; P�0.84). Ten patients developed progessive<br />

disease and 18 relapsed, while 5 have died (1 after relapse in an accident<br />

and 4 <strong>of</strong> refractory disease). Most common toxicities were grade 3<br />

hematologic with 234 episodes <strong>of</strong> neutropenia in 101 patients (72%) and<br />

85 episodes <strong>of</strong> anemia in 52 patients (37%); Fever and neutropenia<br />

occurred 13 times in 12 patients (9%). Conclusions: Risk-adapted,<br />

combined-modality therapy using 12 weeks <strong>of</strong> Stanford V chemotherapy<br />

plus IFRT is well tolerated in this population with manageable acute<br />

toxicities. Overall survival is comparable to other more intense chemotherapy<br />

regimens. Future high-risk front line therapies may consider a<br />

Stanford V backbone with targeted intensification and further tailoring <strong>of</strong><br />

radiation therapy.<br />

9501 Oral Abstract Session, Sat, 1:15 PM-4:15 PM<br />

Efficacy <strong>of</strong> rituximab plus FAB group C chemotherapy without CNS<br />

radiation in CNS-positive pediatric Burkitt lymphoma/leukemia: A report<br />

from the Children’s Oncology Group. Presenting Author: J. Kimble Frazer,<br />

University <strong>of</strong> Utah, Salt Lake City, UT<br />

Background: Historically, CNS-positive (CNS�) mature B-NHL in children<br />

and adolescents has been associated with a dismal outcome (Miles/Cairo,<br />

Br J Haematol, 2012). Adding CNS radiation and high-dose MTX and<br />

cytarabine to group C therapy for children with CNS� B-NHL yielded a<br />

77% 5-yr EFS in LMB89 (Patte et al., Blood, 2001). An ensuing<br />

FAB/LMB96 trial replaced cranial radiation with further HD-MTX and<br />

intrathecal therapy with similar results (75% 4-yr EFS) (Cairo et al., Blood,<br />

2007). Here, patients with marrow and CNS� disease fared worse than<br />

isolated CNS� (61% vs. 83% 4-yr EFS, p�0.001), with �50% <strong>of</strong> CNS�<br />

patients who progressed or recurred having systemic, but not CNS, relapse.<br />

Rituximab, a chimeric anti-CD20 antibody, improved EFS, PFS, and OS<br />

when added to chemotherapy for adults with DLBCL (Coiffier et al., N Engl J<br />

Med, 2002; Pfreundschuh et al., Lancet Oncol, 2006). We tested whether<br />

adding rituximab to FAB group C chemotherapy in pediatric patients with<br />

CNS� B-NHL was safe and efficacious. Methods: Children and adolescents<br />

(�21 yrs.) with CNS� B-NHL received FAB group C1 therapy (Cairo et al.,<br />

Blood, 2007). Rituximab (375 mg/m2 /dose) was given twice in COPADM<br />

courses 1&2andonce in CYVE courses (Cairo et al., ASCO, 2010). CSF<br />

blasts (�1), cranial nerve palsy (CNP), intra-cerebral mass (ICM), and/or<br />

parameningeal extension (PME) defined cases as CNS�. Results: Of 40<br />

eligible Group C patients,15 (38%) were CNS�; all 15 had Burkitt<br />

morphology. Eight CNS� patients were CSF� [WBC median 35 (range<br />

1-1104)] with 6 cases CNP�, 4 PME�, and 1 ICM�. No adverse events<br />

were attributed to rituximab. Fourteen <strong>of</strong> 15 CNS� patients (93%) are alive<br />

and disease-free. Of 7 BM- /CNS� patients, 100% are NED. In BM�/CNS�<br />

cases, 7/8 (88%) are NED, with one patient progressing with systemic and<br />

CNS disease. Conclusions: Addition <strong>of</strong> rituximab to group C FAB therapy<br />

was well tolerated. Outcomes in this small cohort <strong>of</strong> 15 children and<br />

adolescents with CNS� BL (93%) suggest that adding rituximab to FAB<br />

group C therapy without CNS radiation may reduce systemic relapse. Large<br />

randomized studies are warranted to test this hypothesis in this previously<br />

high-risk clinical subgroup.<br />

9503 Oral Abstract Session, Sat, 1:15 PM-4:15 PM<br />

The effects <strong>of</strong> dexrazoxane on cardiac status and second malignant<br />

neoplasms (SMN) in doxorubicin-treated patients with osteosarcoma (OS).<br />

Presenting Author: Lisa M. Kopp, The University <strong>of</strong> Arizona, Tucson, AZ<br />

Background: Anthracyclines are highly efficacious in OS but associated with<br />

risk <strong>of</strong> cardiotoxicity. We conducted two studies using dexrazoxane as a<br />

cardioprotectant: i) AOST0121, a phase II study for metastatic OS and ii)<br />

P9754, a series <strong>of</strong> pilot studies including doxorubicin dose intensification<br />

in patients with localized OS. Methods: Patients on AOST0121 received<br />

methotrexate, doxorubicin (375 mg/m2 ), cisplatin, ifosfamide and etoposide<br />

(MAPIE). Those with HER2-positive tumors also received trastuzumab<br />

(4 mg/kg loading dose, 2 mg/kg weekly). On P9754, patients received MAP<br />

in Pilot 1, MAPI or MAPIE in Pilots 2 and 3. Total doxorubicin was 450<br />

mg/m2 or 600 mg/m2 with each dose preceded by dexrazoxane (10:1 ratio).<br />

Etoposide was never given simultaneously with dexrazoxane. Measurements<br />

<strong>of</strong> left ventricular systolic function by echocardiography, serum<br />

troponin-T (cTnT, a myocardial injury marker) and N-terminal pro-brain<br />

natriuretic peptide (NT-proBNP, a cardiomyopathy marker) were obtained<br />

at baseline and repeated during therapy. Secondary malignancies were<br />

reported to the NCI. Results: None <strong>of</strong> the 47 patients (17 receiving<br />

trastuzumab) evaluated for cardiac toxicity on AOST0121 had significant<br />

changes in left ventricular fractional shortening, measurable cTnT, elevation<br />

<strong>of</strong> NT-proBNP or clinical evidence <strong>of</strong> cardiotoxicity (CTCv2.0). In<br />

P9754, 242 patients were evaluated for cardiotoxicity. Only 1 patient had<br />

CTCv2.0 grade 3 toxicity and 1 patient had a measurable cTnT. Three <strong>of</strong> 96<br />

patients treated on AOST0121 and two <strong>of</strong> 272 patients on P9754<br />

developed AML. Combining both studies: 5/398 or 1.4%. Conclusions: This<br />

large group <strong>of</strong> OS patients demonstrates that dexrazoxane is an effective<br />

cardioprotectant for doxorubicin alone (375-600 mg/m2 ), and in combination<br />

with trastuzumab. Dexrazoxane did not lead to an increase in<br />

secondary malignancies in OS patients treated with these regimens as<br />

compared to historical rates <strong>of</strong> 1-2%. Our results do not support the<br />

findings <strong>of</strong> a recent European Medicines Agency safety review that<br />

concluded dexrazoxane was unsafe for use in children and adolescents due<br />

to risk <strong>of</strong> SMN.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


9504 Oral Abstract Session, Sat, 1:15 PM-4:15 PM<br />

Cardioprotection and safety <strong>of</strong> dexrazoxane (DRZ) in children treated for<br />

newly diagnosed T-cell acute lymphoblastic leukemia (T-ALL) or advanced<br />

stage lymphoblastic leukemia (T-LL). Presenting Author: Barbara Asselin,<br />

University <strong>of</strong> Rochester, Rochester, NY<br />

Background: POG 9404 evaluated the efficacy <strong>of</strong> DRZ as a cardioprotectant<br />

and its side effects in children receiving doxorubicin (DOX) as part <strong>of</strong><br />

chemotherapy for T-ALL/T-LL. Methods: Between 6/1996 and 9/2001, 537<br />

T-ALL/T-LL patients were treated with multi-agent chemotherapy that<br />

included 12 DOX doses <strong>of</strong> 30 mg/m2 . Patients were randomized at<br />

diagnosis to treatment with or without DRZ (300 mg/m2 IV immediately<br />

prior to each DOX dose). Cardiac effects were assessed by serial echocardiograph<br />

measurements <strong>of</strong> left ventricular (LV) function (fractional shortening)<br />

and structure (end-diastolic dimension, wall thickness, and mass). Adverse<br />

events, grades 3-5 were recorded using CTCAE version 2.0. Results:<br />

Five-year EFS was similar for the DRZ (0.77�0.03%) and no-DRZ<br />

(0.76�0.03%) groups (p�0.9). Acute cardiac toxicity occurred in 1<br />

patient (arrhythmia) on DRZ and 4 patients treated without DRZ (1arrhythmia,<br />

3-decreased LV fractional shortening). At each <strong>of</strong> the three<br />

time-points post-baseline (end-induction, after the last DOX dose and two<br />

years post study entry) LV dimensions were larger from baseline for patients<br />

who did not receive DRZ. In univariate analyses, change in LV end-diastolic<br />

dimension from baseline was the only variable that was significantly<br />

different between DRZ and no-DRZ arms. This was true at each <strong>of</strong> the three<br />

time points (p�0.005-0.02). The frequencies <strong>of</strong> grade 3 or 4 toxicity<br />

(hematologic, infection, CNS events, mucositis) or death were similar in<br />

groups with or without DRZ (p�0.24). There were 13 second malignancies,<br />

10 patients had received DRZ and 3 had not (p�0.07). Conclusions: Our LV<br />

dimension measurements suggest that DRZ may protect against the early<br />

LV remodeling and enlargement that is seen with DOX cardiotoxicity.<br />

Addition <strong>of</strong> DRZ did not interfere with the anti-tumor efficacy <strong>of</strong> this<br />

DOX-containing, multi-agent regimen, and there was no significant increase<br />

in toxicities. An increased rate <strong>of</strong> second malignancies with DRZ did<br />

not reach conventional levels <strong>of</strong> statistical significance. The study was not<br />

powered to assess second malignancy rates and this trend is <strong>of</strong> unknown<br />

clinical significance.<br />

9506 Oral Abstract Session, Sat, 1:15 PM-4:15 PM<br />

In vivo monitoring <strong>of</strong> JAK/STAT and PI3K/mTOR signal transduction<br />

inhibition in pediatric CRLF2-rearranged acute lymphoblastic leukemia<br />

(ALL). Presenting Author: Sarah Kathleen Tasian, The Children’s Hospital<br />

<strong>of</strong> Philadelphia, Philadelphia, PA<br />

Background: Therapy intensification for children with B-precursor ALL with<br />

high-risk genetic lesions has improved relapse-free survival. CRLF2 rearrangements<br />

and JAK2 and IL7RA mutations occur in 10-15% <strong>of</strong> adult and<br />

pediatric ALL patients, most <strong>of</strong> whom relapse. We and others identified<br />

aberrant kinase signatures and perturbed JAK/STAT and PI3K/mTOR signal<br />

transduction via in vitro studies <strong>of</strong> CRLF2-rearranged (CRLF2r) ALLs,<br />

suggesting the therapeutic relevance <strong>of</strong> signal transduction inhibitors<br />

(STIs). Our creation <strong>of</strong> CRLF2r ALL xenograft models has enabled rapid<br />

preclinical testing <strong>of</strong> STIs and measurement <strong>of</strong> in vivo target inhibition. We<br />

hypothesized that inhibition <strong>of</strong> JAK/STAT and PI3K/mTOR phosphosignaling<br />

correlates with therapeutic responses in these models. Methods:<br />

NOD/SCID/�c-null (NSG) mice well-engrafted with pediatric ALL samples<br />

were treated with the JAK inhibitor ruxolitinib, the mTOR inhibitor<br />

sirolimus, or vehicle for 72 hours (for signaling response) or 4 weeks (for<br />

therapeutic response). Splenocytes were briefly stimulated ex vivo with<br />

thymic stromal lymphopoietin (ligand for CRLF2) and stained with humanspecific<br />

surface and intracellular phosphoantibodies for multi-parameter<br />

phosph<strong>of</strong>low cytometry analysis. Results: Ruxolitinib-induced inhibition <strong>of</strong><br />

phospho (p)-JAK2 and pSTAT5 was most pronounced in non-CRLF2r ALLs<br />

with novel JAK2-activating BCR-JAK2 and IL7RA/LNK mutations. Sirolimus<br />

potently inhibited pS6 and other PI3K/mTOR pathway phosphoproteins<br />

in the CRLF2r r ALLs. PSTAT5 and pS6 inhibition correlated with<br />

longer-term ruxolitinib- and sirolimus-induced decreases in ALL cell<br />

burden, demonstrating therapeutic responses to STIs. Conclusions: Ruxolitinib<br />

inhibited JAK/STAT phosphosignaling and markedly decreased<br />

leukemic burden in the JAK2-activating BCR-JAK2 and IL7RA/LNK mutant<br />

ALL xenografts. Sirolimus potently inhibited PI3K/mTOR (as well as some<br />

JAK/STAT) phosphosignaling and had greater therapeutic efficacy than<br />

ruxolitinib in the CRLF2r ALLs. The safety <strong>of</strong> ruxolitinib and <strong>of</strong> temsirolimus<br />

with cytotoxic chemotherapy are currently being established in Children’s<br />

Oncology Group Phase I trials.<br />

Pediatric Oncology<br />

9505 Oral Abstract Session, Sat, 1:15 PM-4:15 PM<br />

Efficacy <strong>of</strong> the Children’s Oncology Group (COG) long-term followup<br />

(LTFU) guidelines in reducing risk <strong>of</strong> congestive heart failure<br />

(CHF) in childhood cancer survivors (CCS). Presenting Author: F.<br />

Lennie Wong, City <strong>of</strong> Hope, Duarte, CA<br />

Background: CCS are at risk for left ventricular dysfunction (LVD) and<br />

subsequent CHF due to exposure to anthracyclines and chest radiation<br />

(RT). COG LTFU guidelines recommend screening for LVD using echocardiograms<br />

(ECHOs) every 1-5y depending on anthracycline dose, RT, and<br />

age at cancer diagnosis. The relevance and cost-effectiveness <strong>of</strong> these<br />

consensus-based guidelines are unknown. Methods: Life expectancy and<br />

age at onset <strong>of</strong> CHF were projected in a simulated cohort (�1 million) <strong>of</strong><br />

CCS undergoing screening ECHO per COG guidelines. Intervention for LVD<br />

was modeled to reduce annual CHF risk by 30%. Quality-adjusted life-years<br />

(QALYs) and lifetime costs with and without ECHO screening were<br />

calculated. Non-CHF mortality was estimated from the Childhood Cancer<br />

Survivor Study and US population rates. Costs and QOL adjustments were<br />

obtained from the Healthcare Cost and Utilization Project and medical<br />

literature. Screening was considered cost-effective if it resulted in a �6<br />

month delay in onset <strong>of</strong> CHF and �$50,000/QALY gained. Results:<br />

Recommended screening strategies (Table) were cost-effective in: i) CCS<br />

exposed to �300mg/m² <strong>of</strong> anthracycline regardless <strong>of</strong> RT or age; and ii)<br />

CCS diagnosed at age 1-4y, exposed to RT and �300mg/m² <strong>of</strong> anthracycline.<br />

Screening was most cost-effective for CCS diagnosed at age 1-4y<br />

exposed to RT � �300 mg/m² <strong>of</strong> anthracycline (1.4y delay in CHF onset;<br />

$15,821/QALY gained). Screening as currently proposed was not costeffective<br />

for other age/anthracycline/RT combinations. Conclusions: Recommended<br />

ECHO screening strategies are cost-effective for all CCS exposed to<br />

�300 mg/m² <strong>of</strong> anthracycline; screening was also cost-effective for those<br />

1-4y at diagnosis, exposed to anthracycline �300 mg/m² � RT. Alternate<br />

screening strategies are needed for CCS with other exposure conditions.<br />

Age Dx<br />

(years) RT<br />

Anthracycline<br />

dose<br />

(mg/m²)<br />

Recommended<br />

ECHO<br />

interval<br />

(years)<br />

Delay in<br />

CHF onset age<br />

(years)<br />

607s<br />

Cost / QALY<br />

gained<br />

(US$)<br />

1-4 Yes �300 1 0.8 $49,750<br />

�300 1 1.4 $15,821<br />

No �100 5 0.3 $17,798<br />

100-299 2 0.4 $29,415<br />

�300 1 1.2 $25,065<br />

>5 Yes �300 2 0.4 $36,874<br />

�300 1 0.8 $28,093<br />

No � 200 5 0.1 $50,750<br />

200-299 2 0.2 $86,867<br />

�300 1 0.7 $36,401<br />

9507 Oral Abstract Session, Sat, 1:15 PM-4:15 PM<br />

Relapse-specific mutations in cytosolic 5’-nucleotidase II in childhood<br />

acute lymphoblastic leukemia. Presenting Author: Julia Meyer, New York<br />

University Langone Medical Center, New York, NY<br />

Background: Relapsed childhood acute lymphoblastic leukemia (ALL)<br />

carries a very poor prognosis despite intensive retreatment that <strong>of</strong>ten<br />

includes allogeneic stem cell transplantation, due to intrinsic drug resistance.<br />

Novel therapeutic approaches are urgently needed and identification<br />

<strong>of</strong> the biological pathways that mediate resistance might provide targets for<br />

the effective prevention and treatment <strong>of</strong> relapsed ALL. However, the<br />

spectrum <strong>of</strong> somatic mutations responsible for ALL relapse is not yet<br />

known. Methods: We pr<strong>of</strong>iled the transcriptome <strong>of</strong> matched diagnosis and<br />

relapse bone marrow specimens from 10 pediatric B lymphoblastic<br />

leukemia patients using massively parallel sequencing technology (RNAseq)<br />

to identify novel mutations specific at disease recurrence. Results:<br />

Transcriptome sequencing identified 20 newly acquired novel nonsynonymous<br />

mutations in 10 relapse specimens not present at initial<br />

diagnosis. Two patients harbored relapsed specific mutations in the same<br />

gene, NT5C2, which codes for the Cytosolic 5’-nucleotidase II protein at<br />

different sites in the protein sequence. Mutations were validated as relapse<br />

specific after Sanger resequencing <strong>of</strong> germline, diagnosis and relapse DNA.<br />

RNA-seq coverage <strong>of</strong> each mutation was �100X, indicating that if a<br />

relapsed clone were present at diagnosis it made up � 1% <strong>of</strong> the bulk<br />

leukemia. Full exon sequencing <strong>of</strong> NT5C2 was completed in 61 additional<br />

relapse specimens, identifying 5 additional mutations which were also<br />

confirmed as relapse specific. Structural modeling <strong>of</strong> the relapseassociated<br />

mutations in the encoded protein Cytosolic 5’-nucleotidase II<br />

suggests alteration <strong>of</strong> enzyme subunit association/dissociation. <strong>Clinical</strong>ly,<br />

patients who harbored NT5C2 mutations were more likely to relapse earlier<br />

following initial diagnosis than those patients without mutations (p�0.03).<br />

Conclusions: Mutations in NT5C2 are associated with the outgrowth <strong>of</strong> drug<br />

resistant cells in childhood ALL.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


608s Pediatric Oncology<br />

CRA9508 Oral Abstract Session, Sat, 1:15 PM-4:15 PM<br />

Outcome in adolescent and young adult (AYA) patients compared with<br />

younger patients treated for high-risk B-precursor acute lymphoblastic<br />

leukemia (HR-ALL): A report from the Children’s Oncology Group study<br />

AALL0232. Presenting Author: Eric Larsen, Maine Children’s Cancer<br />

Program, Scarborough, ME<br />

The full, final text <strong>of</strong> this abstract will be available at<br />

abstract.asco.org at 12:01 AM (EDT) on Saturday, June 2,<br />

2012, and in the <strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong> online<br />

supplement to the June 20, 2012, issue <strong>of</strong> Journal <strong>of</strong><br />

<strong>Clinical</strong> Oncology. Onsite at the <strong>Meeting</strong>, this abstract will<br />

be printed in the Saturday edition <strong>of</strong> ASCO Daily News.<br />

9510 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Development <strong>of</strong> prognostic molecular markers in pediatric rhabdomyosarcoma<br />

based on gene expression and copy number variations. Presenting<br />

Author: Edoardo Missiaglia, Swiss Institute <strong>of</strong> Bioinformatics, Lausanne,<br />

Switzerland<br />

Background: Rhabdomyosarcoma (RMS) is the most common pediatric s<strong>of</strong>t<br />

tissue sarcoma and comprises two major histological subtypes: alveolar and<br />

embryonal. The majority <strong>of</strong> alveolar tumors harbor PAX/FOXO1 fusion<br />

genes. Current patient risk stratification, unlike other pediatric embryonal<br />

tumors, does not utilize any molecular data. Therefore, we aimed to<br />

improve the risk stratification <strong>of</strong> RMS patients through the use <strong>of</strong> molecular<br />

biological data. Methods: Two independent data sets <strong>of</strong> gene expression<br />

pr<strong>of</strong>iling for 124 and 101 RMS were used to derive prognostic gene<br />

signatures by meta-analysis. Genomic array CGH data for 109 RMS was<br />

also evaluated to develop a prognostic marker based on copy number<br />

variations (CNVs). The performance and usefulness <strong>of</strong> these derived<br />

metagenes and CNVs as well as a previously published metagene signature<br />

were evaluated using rigorous leave-one-out cross-validation analyses.<br />

Results: The new prognostic gene expression signature, MG15, and one<br />

previously published (MG34) (Davicioni. JCO. 2010) performed well with<br />

reproducible and significant effects (HR 3.2 [1.7-5.9] p � 0.001 and HR<br />

2.5 [1.5-4.3] p � 0.001, respectively). However, they did not significantly<br />

add new prognostic information over the fusion gene status (PAX3/FOXO1,<br />

PAX7/FOXO1 and Negative). Similarly, a prognostic CNV marker, although<br />

showing HR 2.9 [1.5-5.6] p � 0.01, was also not improving models with<br />

fusion gene status. Within fusion negative RMS, the analysis identified<br />

prognostic markers based on either gene expression or CNVs and showed<br />

significant association with patients outcome (HR 6.3 [1.5-26.3] p �<br />

0.016 and HR 11.2 [2.5-50.7] p � 0.010, respectively). Moreover, these<br />

were able to identify distinct risk groups within the COG (Children’s<br />

Oncology Group) risk categories, which is currently used to guide treatment.<br />

Conclusions: Molecular signatures derived using all RMS effectively<br />

stratify patients by their risk, but most <strong>of</strong> their prognostic information is<br />

contained in the PAX/FOXO1 fusion gene status which is simpler to assay.<br />

New markers developed within the fusion negative population seem<br />

improving current RMS risk classifier and should be tested in follow-up<br />

studies.<br />

9509 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Vincristine (V), dactinomycin (A), and lower doses <strong>of</strong> cyclophosphamide (C)<br />

with or without radiation therapy for patients with newly diagnosed low-risk<br />

embryonal rhabdomyosarcoma (ERMS): A report from the Children’s<br />

Oncology Group (COG). Presenting Author: David Walterhouse, Children’s<br />

Memorial Hospital, Chicago, IL<br />

Background: Intergroup Rhabdomyosarcoma Study (IRS) trials showed<br />

improved survival with VAC compared with VA for patients with Stage 1<br />

Group III (non-orbit) or Stage 3 Group I/II ERMS (see table). In COG<br />

ARST0331, we hypothesized that VA in combination with lower doses <strong>of</strong> C<br />

(total cumulative dose�4.8 g/m2 ) would produce the benefit <strong>of</strong> IRS-IV VAC<br />

with less toxicity for patients with Stage 1 Group III (non-orbit) or Stage 3<br />

Group I/II low-risk ERMS. Methods: This single arm, non-inferiority, phase<br />

III study enrolled newly diagnosed patients with Stage 1 Group III<br />

(non-orbit) ERMS or Stage 3 Group I/II ERMS onto Subset 2. Therapy was 4<br />

cycles <strong>of</strong> VAC followed by 12 cycles <strong>of</strong> VA over 46 weeks (total cumulative<br />

doses: V�54 mg/m2 ,A�21.6 mg/m2 ,C�4.8 g/m2 ). The radiation therapy<br />

dose was 36 Gy for Group IIA patients, 41.4 Gy for Group IIB/C patients,<br />

and 50.4 Gy for Group III patients. From 2004–2008 girls with Group III<br />

vaginal RMS did not receive radiotherapy if a complete response was<br />

achieved with chemotherapy with or without delayed resection. The primary<br />

endpoint was failure-free survival (FFS), and results were compared with a<br />

fixed expected outcome. Results: With a median follow-up <strong>of</strong> 3.0 yrs, we<br />

observed 16 failures vs. 7.8 expected failures. Estimated 3-yr FFS was<br />

63% (95% CI: 46%, 75%) (n�60), and overall survival (OS) was 84%<br />

(95% CI: 68%, 93%). Estimated 3-yr FFS was 46% (95% CI: 23%, 67%)<br />

for girls with non-bladder genitourinary tract ERMS (n�21) and 75% (95%<br />

CI: 53%, 88%) for all other Subset 2 patients (n�39). Conclusions: We<br />

observed suboptimal FFS <strong>of</strong> patients with Subset 2 low-risk RMS using<br />

reduced total cyclophosphamide (4.8 g/m2 ). Results were complicated by<br />

the choice <strong>of</strong> no radiation therapy for girls with vaginal tumors. Future<br />

studies for low-risk RMS Subset 2 patients could investigate a dose <strong>of</strong> C<br />

between 4.8 and 26.4 g/m2 with VA and local radiotherapy.<br />

Chemotherapy cumulative dose<br />

Study<br />

Duration<br />

(weeks) V (mg/m<br />

FFS<br />

(5-yr)<br />

OS<br />

(5-yr)<br />

2 ) A (mg/m2 ) C (g/m2 )<br />

IRS-III 45 72 13.5 - 70% 79%<br />

IRS-IV 45 48 22.5 26.4 84%<br />

98%<br />

p�0.008 p�0.004<br />

9511 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Identification <strong>of</strong> TNK2 as a critical kinase in rhabdomyosarcoma through a<br />

loss <strong>of</strong> function shRNA screen. Presenting Author: Fernanda Irene Arnaldez,<br />

Pediatric Oncology Branch, National Cancer Institute, Bethesda, MD<br />

Background: Rhabdomyosarcoma is the most common pediatric s<strong>of</strong>t tissue<br />

sarcoma. Embryonal rhabdomyosarcomas (ERMS) are characterized by<br />

11p15 LOH while alveolar rhabdomyosarcomas harbor a translocation<br />

between PAX3 or PAX7 and FOXO1. Relapsed or metastatic disease has a<br />

5-year survival rate <strong>of</strong> 25%. Methods: We sought to identify critical genes<br />

for rhabdomyosarcoma cell growth and survival. We performed a loss-<strong>of</strong>function<br />

shRNA screen where a library <strong>of</strong> 15,000 shRNAs was introduced<br />

in RH30 (ARMS) and RD (ERMS) cells engineered to express the bacterial<br />

tetracycline repressor in a tet-on system. Relative abundance <strong>of</strong> cells<br />

containing a certain shRNA was determined using specific barcodes. We<br />

subtracted “hits” that are common to 4 lymphoma cell-lines to eliminate<br />

common survival pathways. Forty genes were associated with rhabdomyosaroma<br />

cell growth, <strong>of</strong> which 15 cause greater suppression in ARMS. We<br />

performed a subsequent validation <strong>of</strong> these 15 genes in multiple ARMS<br />

and ERMS cell lines using three different shRNA sequences using lentiviral<br />

constructs. In addition, we validated our results in an independent siRNA<br />

screen <strong>of</strong> the protein kinome in RMS cells. Results: TNK2 (tyrosine kinase,<br />

non-receptor 2) was identified as tyrosine kinase involved in rhabdomyosarcoma<br />

cell growth independently in the inducible shRNA screen and in the<br />

non-inducible siRNA platform. This molecule has been involved in downstream<br />

signaling from EGFR and integrins. Amplification <strong>of</strong> the TNK2 gene,<br />

located at 3q29, has been described in breast, prostate and lung cancer.<br />

More recently, a siRNA screen identified it as a potential therapeutic target<br />

in Ewing’s sarcoma. We confirmed TNK2 expression across patient tumor<br />

samples in a publically available database; where patients with high TNK2<br />

expression showed lower overall survival. TNK2 knock down resulted in<br />

decreased rhabdomyosarcoma cell growth in vitro and in orthotopic<br />

xenografts. We confirmed specificity <strong>of</strong> our findings with a rescue experiment.<br />

Conclusions: We identified TNK2 as a potential therapeutic target in<br />

rhabdomyosarcoma using a loss-<strong>of</strong>-function shRNA screen and subsequent<br />

validation. Further work is ongoing to fully characterize the molecular<br />

mechanisms involved in these findings.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


9512 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Evaluation <strong>of</strong> the effect <strong>of</strong> care at NCI comprehensive cancer centers<br />

(NCICCCs) on disparities in outcome within adolescents and young adults<br />

(AYAs) with cancer. Presenting Author: Julie Anna Wolfson, City <strong>of</strong> Hope,<br />

Duarte, CA<br />

Background: AYAs (15-39y at diagnosis) with cancer have not seen the<br />

survival improvement evidenced by younger and older age groups with<br />

similar diagnoses, leaving an AYA Gap. While treatment on pediatric<br />

protocols is associated with superior survival in 15-21 year-olds, the<br />

impact <strong>of</strong> site <strong>of</strong> care on survival for vulnerable AYA subpopulations (age at<br />

diagnosis or race/ ethnicity) between 22-and 39y at diagnosis remains<br />

unstudied. Methods: We constructed a cohort <strong>of</strong> 10,727 patients newly<br />

diagnosed between the ages <strong>of</strong> 22- and 39y with lymphoma, leukemia,<br />

brain tumors, melanoma, thyroid and GU cancers, and reported to the LA<br />

County cancer registry between 1998 and 2008. Multivariable Cox<br />

regression analysis was conducted, and included race/ethnicity, age at<br />

diagnosis, SES, insurance status, primary cancer diagnosis and diagnosis<br />

year in the model; the analysis was stratified by site <strong>of</strong> care (NCICCC vs.<br />

non-NCICCC). Results: A total <strong>of</strong> 928 (9%) patients received treatment at<br />

the 3 NCICCCs (City <strong>of</strong> Hope, Jonsson Cancer Center and Norris Cancer<br />

Center) in LA County, and 9,799 received care elsewhere. Five-year overall<br />

survival (5y OS) was significantly worse for patients treated at non-NCICCC<br />

(87%) when compared with those treated at NCICCC (84%, p�0.02). In<br />

addition, 5y OS was worse for African <strong>American</strong>s (71%) vs. non-Hispanic<br />

whites (89%, p�0.0001) and for older patients (31-39yo: 84%, vs.<br />

22-30yo: 86%, p�0.0004). Multivariable analysis adjusting for SES,<br />

insurance status, diagnosis and diagnosis year revealed that African<br />

<strong>American</strong>s (HR�1.4, p�0.0002) and older AYAs (31-39y: HR�1.24,<br />

p�0.0001) were at an increased risk <strong>of</strong> death. Among patients treated at<br />

NCICCC, the difference in risk <strong>of</strong> death due to race (African <strong>American</strong>s:<br />

HR�0.8, p�0.7) and age (31-39yo: HR�1.1, p�0.6) was abrogated. On<br />

the other hand, among patients treated at non-NCICCC, these differences<br />

in outcome persisted (African <strong>American</strong>s: HR�1.45, p �0.0002; 31-<br />

39yo: HR�1.25, p�.0001). Conclusions: Population-based data reveal<br />

that receipt <strong>of</strong> care at an NCICCC abrogates the effects <strong>of</strong> race and older<br />

age on mortality in AYAs with cancer. Barriers to accessing care at NCICCCs<br />

are being explored.<br />

9514 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Increasing risk <strong>of</strong> chronic health conditions in aging survivors <strong>of</strong> childhood<br />

cancer: A report from the Childhood Cancer Survivor Study. Presenting<br />

Author: Gregory T. Armstrong, St. Jude Children’s Research Hospital,<br />

Memphis, TN<br />

Background: Survivors <strong>of</strong> childhood cancer are at an increased risk for<br />

treatment-related chronic health conditions during early adulthood. However,<br />

the incidence, severity, and spectrum <strong>of</strong> chronic health conditions in<br />

the fourth and fifth decades <strong>of</strong> life have not been well studied. Methods:<br />

Analyses included 14,358 � 5 yr survivors <strong>of</strong> childhood cancer (median<br />

age at last follow-up 32.3 yrs, range 8.0-58.0; 21.4% � 40 years) and a<br />

sibling comparison group (n � 4,031). Self-reported health conditions<br />

were classified using NCI CTCAE 4.0 grading system. Analyses focused on<br />

two primary outcomes: severe/life-threatening/fatal conditions (grades<br />

3-5), and multiple (� 2) conditions. Cumulative incidence <strong>of</strong> a new chronic<br />

health condition was calculated from age 26 yrs. Cox proportional hazards<br />

models adjusted for gender and race, were evaluated using age as the time<br />

scale. Results: Among survivors with no previous health conditions through<br />

age 25, the cumulative incidence for a new grade 3-5 condition by age 50<br />

compared to siblings was 45.9% (95% CI 45.9-45.9) vs. 13.9%, (95% CI<br />

13.9-14.0) and for new onset <strong>of</strong> � 2 conditions 33.0% (95% CI<br />

33.0-33.1) vs. 24.9% (95% CI 24.8-24.9) . Survivors � 40 yrs <strong>of</strong> age had<br />

a 5.8-fold (95% CI 5.3 – 6.5) increased risk <strong>of</strong> a grade 3-5 condition<br />

compared to same age siblings, in contrast to those � 40 years <strong>of</strong> age (HR<br />

2.7, 95% CI 2.5-3.0). A similar magnitude <strong>of</strong> difference was present for<br />

risk <strong>of</strong> �2 conditions (HR 2.7 vs. 1.2). In comparison to siblings, survivors<br />

� 40 years <strong>of</strong> age had a significantly increased risk for: congestive heart<br />

failure (HR 15.7, 95% CI 9.2-26.7), myocardial infarction (HR 8.8, 95%<br />

CI 6.0-12.9), stroke (HR 8.6, 95% CI 5.6-13.2),joint replacement (HR<br />

6.8, 95% CI 4.1-11.4), renal failure (HR 5.1, 95% CI 2.2-11.9) among<br />

other serious conditions. Conclusions: As they age, adult survivors <strong>of</strong><br />

childhood cancer continue to develop new and serious health conditions at<br />

substantially higher rates than siblings. These data emphasize the importance<br />

<strong>of</strong> placing a greater focus on investigations <strong>of</strong> premature aging and<br />

organ senescence in this high risk population.<br />

Pediatric Oncology<br />

609s<br />

CRA9513 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

New insights into the risk <strong>of</strong> breast cancer in childhood cancer survivors<br />

treated with chest radiation: A report from the Childhood Cancer Survivor<br />

Study (CCSS) and the Women’s Environmental Cancer and Radiation<br />

Epidemiology (WECARE) Study. Presenting Author: Chaya S. Moskowitz,<br />

Memorial Sloan-Kettering Cancer Center, New York, NY<br />

The full, final text <strong>of</strong> this abstract will be available at<br />

abstract.asco.org at 12:01 AM (EDT) on Monday, June 4,<br />

2012, and in the <strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong> online<br />

supplement to the June 20, 2012, issue <strong>of</strong> Journal <strong>of</strong><br />

<strong>Clinical</strong> Oncology. Onsite at the <strong>Meeting</strong>, this abstract will<br />

be printed in the Monday edition <strong>of</strong> ASCO Daily News.<br />

9515 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

A large prospective trial <strong>of</strong> children with unilateral retinoblastoma with and<br />

without histopathologic high-risk features and the role <strong>of</strong> adjuvant chemotherapy:<br />

A Children’s Oncology Group (COG) study. Presenting Author:<br />

Murali M. Chintagumpala, Baylor College <strong>of</strong> Medicine, Houston, TX<br />

Background: The definition <strong>of</strong> histopathologic high-risk features (HRF) in<br />

enucleated eyes <strong>of</strong> children with unilateral retinoblastoma and their<br />

contribution to metastases is controversial. The COG completed a large,<br />

prospective international study to determine the prevalence <strong>of</strong> strictly<br />

defined histopathologic HRF that are predictors <strong>of</strong> recurrence and the role<br />

<strong>of</strong> chemotherapy to prevent recurrences. Methods: All patients who underwent<br />

enucleation for unilateral retinoblastoma were eligible for the study.<br />

Pathology slides were submitted for central review within 21 days <strong>of</strong><br />

enucleation. Patients with evidence <strong>of</strong> one or more high-risk features<br />

(posterior uveal invasion grades IIC and D, concurrent optic nerve and<br />

choroid involvement and post-lamina optic nerve involvement) as determined<br />

by central review, received 6 cycles <strong>of</strong> chemotherapy consisting <strong>of</strong><br />

carboplatin, vincristine and etoposide. All others were observed. All<br />

patients were followed for extraocular recurrences. Results: Patients were<br />

enrolled from February <strong>of</strong> 2005 until May 2010. As <strong>of</strong> July 2011, the<br />

median follow-up from enrollment was 1.9 years (max�5.3 years). Of 312<br />

patients with central histopathology review, 49 patients had their risk<br />

classification changed (13% with no HRF had HRF, 24% with HRF had no<br />

HRF). Two patients developed extraocular disease and one patient died <strong>of</strong><br />

unknown cause. The death and one <strong>of</strong> the extraocular relapses occurred<br />

among the 93 (2/93�2.2%, upper 95% CI 3.4%) patients assigned by the<br />

central review to receive chemotherapy, while one patient experienced<br />

extraocular relapse among the 209 (1/209�0.5%, upper 95% CI 0.6%)<br />

assigned to observation only. There is no evidence <strong>of</strong> a difference in the<br />

EFS and OS in these two groups. Conclusions: Preliminary results strongly<br />

suggest that a central review <strong>of</strong> pathology can spare a significant number <strong>of</strong><br />

patients from exposure to chemotherapy. Chemotherapy may have contributed<br />

to fewer relapses in patients with high-risk features as defined in this<br />

study. The preliminary results from this study indicate an excellent<br />

outcome with this approach.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


610s Pediatric Oncology<br />

9516 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Relationship <strong>of</strong> divergent ancestral genetic variation on chromosome 6p22<br />

and racial disparities in survival in neuroblastoma. Presenting Author:<br />

Navin R. Pinto, The University <strong>of</strong> Chicago, Chicago, IL<br />

Background: An increased prevalence <strong>of</strong> high-risk disease and worse<br />

outcome are observed in children with neuroblastoma who self-report as<br />

black versus white. We sought to determine whether genetic variation<br />

would explain this racial disparity. Methods: After quality control, we<br />

analyzed 511,836 germline genetic variants in 2,709 ethnically diverse<br />

children with neuroblastoma enrolled on Children’s Oncology Group study<br />

ANBL00B1 from 2001 to 2009. Genetic variation was summarized by<br />

conducting principal components analysis. The first principal component<br />

(PC1) separated patients with African ancestry from all others. PC1 was<br />

used as a continuous variable for ordinal regression with risk group and a<br />

Cox proportional hazard model <strong>of</strong> EFS. To identify genetic mechanisms for<br />

the observed disparities, we developed a method using genome-wide<br />

variation data applied to high-risk versus non-high risk samples. We<br />

identified a comprehensive list <strong>of</strong> loci with significant divergence between<br />

the ancestral populations. We then tested each such locus for association<br />

with high-risk phenotype using logistic regression with the proportion <strong>of</strong><br />

African ancestry estimated by ADMIXTURE as covariate. Finally, top SNPs<br />

were added to multivariate models <strong>of</strong> both risk and event-free survival to<br />

determine if any top associations could abrogate observed disparities.<br />

Results: PC1 was associated with both risk (p � 0.007) and EFS (p �<br />

0.037). We identified 72 population-divergent SNPs nominally associated<br />

with high-risk disease (p � 0.001). The risk allele for one <strong>of</strong> the top SNPs:<br />

rs9295536 (p � 9.2x10-8 ) was more common in the African ancestral<br />

population, was associated with high-risk phenotype and poor outcome in<br />

all patients and validated in a Caucasian-only subanalysis. In multivariate<br />

testing, this SNP abrogated the PC1 association with EFS (p � 0.18).<br />

Conclusions: A SNP with high divergence between ancestral populations on<br />

chromosome 6p22 accounts for the observed racial disparity in survival and<br />

is also a common genetic variant associated with survival in patients<br />

derived from either European or African ancestry (Bonferroni adjusted p �<br />

0.05). Studies to elucidate the function <strong>of</strong> this SNP are underway.<br />

9518 <strong>Clinical</strong> Science Symposium, Mon, 11:30 AM-1:00 PM<br />

Use <strong>of</strong> whole genome sequencing to identify novel mutations in distinct<br />

subgroups <strong>of</strong> medulloblastoma. Presenting Author: Giles W. Robinson, St.<br />

Jude Children’s Research Hospital, Memphis, TN<br />

Background: Medulloblastoma is a malignant childhood brain tumor comprising<br />

four discrete subgroups (SHH-subgroup, WNT-subgroup, subgroup-3<br />

and subgroup-4). The genetic alterations that drive these subgroups<br />

and that might serve as treatment targets are largely unknown. Methods: We<br />

sequenced entire genomes <strong>of</strong> 37 tumors and matched normal blood. 136<br />

somatically mutated genes identified in this discovery cohort were sequenced<br />

in an additional 56 medulloblastomas. All tumors were classified<br />

into the 4 subgroups by expression pr<strong>of</strong>iling and immunohistochemistry. All<br />

mutations were validated by custom capture, 454, or Sanger sequencing.<br />

Results: Recurrent mutations were detected in 49 genes: 41 are not yet<br />

implicated in medulloblastoma. Several target distinct components <strong>of</strong> the<br />

epigenetic machinery in different disease subgroups, e.g., regulators <strong>of</strong><br />

H3K27 and H3K4 trimethylation in subgroup-3 and 4 (e.g., KDM6A and<br />

ZMYM3), and CTNNB1-associated chromatin remodellers in WNTsubgroup<br />

tumors (e.g., SMARCA4 and CREBBP). Modelling <strong>of</strong> mutations in<br />

mouse lower rhombic lip progenitors that generate WNT-subgroup tumours,<br />

identified genes that maintain this cell lineage (DDX3X) as well as mutated<br />

genes that initiate (CDH1) or cooperate (PIK3CA) in tumourigenesis.<br />

Conclusions: We have identified several new recurrent somatic mutations<br />

that are enriched in specific subgroups <strong>of</strong> medulloblastoma. Alterations<br />

affecting subgroup-3 and 4 tumors appear to disrupt chromatin marking,<br />

most notably H3K27me3 , potentially preserving a stem cell-like state in<br />

tumor cells. Mutations in WNT subgroup tumors affect binding partners <strong>of</strong><br />

CTNNB1 that regulate WNT-response gene transcription. These data<br />

provide important new insights into the pathogenesis <strong>of</strong> medulloblastoma<br />

subgroups and highlight targets for therapeutic development.<br />

9517 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Phase II study <strong>of</strong> temozolomide in combination with topotecan (TOTEM) in<br />

relapsed or refractory neuroblastoma and other pediatric solid malignancies:<br />

A European ITCC study. Presenting Author: Angela Di Giannatale,<br />

Institute Gustave Roussy, Villejuif, France<br />

Background: Temozolomide and topotecan have shown activity in several<br />

pediatric cancers, including neuroblastoma. Resistance to alkylating agents<br />

due to MGMT expression, MMR deficiency or microsatellite instability may<br />

be overcome through the combination with topoisomerase I inhibitors. The<br />

combination <strong>of</strong> temozolomide and topotecan (TOTEM) was well tolerated<br />

and showed preliminary activity in children with neuroblastoma and glioma<br />

(Rubie et al, 2010). Methods: This multicenter, non-randomized, multicohort<br />

Phase II study included children with neuroblastoma according to a<br />

2-stage Simon design, and patients with central nervous system (CNS) and<br />

extra-cranial solid tumors in a descriptive design. Temozolomide was<br />

administered orally at 150 mg/m2 followed by topotecan at 0.75 mg/m2<br />

intravenously for 5 consecutive days every 28 days. The main endpoint was<br />

objective response (OR), i.e., Complete or <strong>Part</strong>ial Response (CR�PR),<br />

evaluated after 2 cycles according to WHO criteria, or INRC criteria for<br />

neuroblastoma patients with mIBG-positive lesions, by an independent<br />

radiological review. Independent review <strong>of</strong> mIBG imaging is pending.<br />

Results: 103 patients, median age 9.4 years (range 1-21), were treated<br />

between June 2009 and May 2011 in 18 centers: 38 neuroblastoma, 33<br />

CNS tumors and 32 other solid tumors. Overall 420 cycles were administered<br />

(median 3 per patient; range 1-12). Grade 3 or 4 neutropenia was<br />

frequent (55% courses), though only 6% <strong>of</strong> patients developed febrile<br />

neutropenia. In the neuroblastoma cohort, 1 CR and 7 PR were observed,<br />

leading to an estimated OR rate <strong>of</strong> 21% (95%CI, 10-37%). Additionally 22<br />

patients had disease stabilization (SD), leading to an overall tumor control<br />

(CR�PR�SD) <strong>of</strong> 79% (63-90%), and a 12-month progression-free survival<br />

rate <strong>of</strong> 47% (31-64%). Overall, 17/102 evaluable patients achieved<br />

an OR (17%, 10-25%), with 1 CR and 3 PR in 9 medulloblastoma (44%,<br />

14-79%), 2 PR in 4 PNET, 1 PR in 12 malignant glioma, and 2 PR in 9<br />

RMS. Conclusions: Temozolomide-topotecan combination results in significant<br />

tumor control in children with neuroblastoma and medulloblastoma/<br />

PNET with favorable toxicity pr<strong>of</strong>ile.<br />

9519 <strong>Clinical</strong> Science Symposium, Mon, 11:30 AM-1:00 PM<br />

A phase I/II study <strong>of</strong> LDE225, a smoothened (Smo) antagonist, in pediatric<br />

patients with recurrent medulloblastoma (MB) or other solid tumors.<br />

Presenting Author: Birgit Geoerger, Institut Gustave Roussy, Villejuif,<br />

France<br />

Background: Hedgehog (Hh) signaling is crucial in the development and<br />

homeostasis <strong>of</strong> many human organs and tissues. Aberrant Hh signaling is<br />

involved in tumorigenesis through promotion <strong>of</strong> cell proliferation, survival,<br />

and differentiation in wide range <strong>of</strong> human cancers, including approximately<br />

30% <strong>of</strong> MBs. LDE225 is a potent and selective inhibitor <strong>of</strong> Smo, a<br />

key positive regulator <strong>of</strong> Hh signaling. The phase I is exploring the safety<br />

and pharmacokinetics <strong>of</strong> LDE225 in pediatric patients with advanced solid<br />

tumors that are potentially dependent on Hh signaling. Preliminary data<br />

from the ongoing phase I are presented. Methods: Dose-escalation was<br />

performed according to a Bayesian design starting at 372 mg/m2 <strong>of</strong><br />

continuous once daily oral LDE225. Safety and preliminary efficacy <strong>of</strong><br />

LDE225 at the maximum tolerated dose will be assessed in pediatric and<br />

adult patients with recurrent MB in a phase II expansion part. Pharmacokinetic<br />

pr<strong>of</strong>iles were performed at Day 1 and 21. Tumor samples were<br />

analyzed for Hh pathway activation status using a 5-gene Hh signature<br />

assay. Results: Thirty-three patients (24 MB, 3 rhabdomyoscarcoma [RMS],<br />

3 osteosarcoma, and 1 each <strong>of</strong> neuroblastoma, gliomatosis and oligoastrocytoma)<br />

with a median age <strong>of</strong> 13 years (range, 4–17 y) have enrolled.<br />

Dose-limiting toxicity <strong>of</strong> Grade 4 creatine phosphokinase elevation occurred<br />

in 1 RMS patient out <strong>of</strong> 7 patients treated at 372 mg/m2 .No<br />

dose-limiting toxicity was observed at 233 and 425 mg /m2 . LDE225 at<br />

233 and 372 mg/m2 was absorbed with a median Tmax <strong>of</strong> 2 h (range, 1–24<br />

h). Systemic exposures were comparable with adults. Two MB patients<br />

achieved a confirmed complete response (CR) at doses <strong>of</strong> 372 and 425<br />

mg/m2 . Analysis <strong>of</strong> 14 available MB tumor samples using the 5-gene Hh<br />

signature assay showed that the 2 CR patients have Hh-activated tumor.<br />

The remaining 12 tumor samples were from patients who did not achieve<br />

response and were determined to be Hh pathway non-activated. Conclusions:<br />

LDE225 is well tolerated in pediatric patients with advanced malignancies.<br />

Preliminary data show promising efficacy in medulloblastoma patients and<br />

support the use <strong>of</strong> the 5-gene Hh signature assay as a pre-selection tool in<br />

future trials.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


9520 <strong>Clinical</strong> Science Symposium, Mon, 11:30 AM-1:00 PM<br />

Real-time dynamic and sequential tracking <strong>of</strong> tumor propagation and<br />

associated immune responses in the CNS microenvironment. Presenting<br />

Author: Alex Yee-Chen Huang, Case Western Reserve University, Cleveland,<br />

OH<br />

Background: Ex vivo experimental systems are <strong>of</strong>ten unable to fully capture<br />

complex intercellular communication between tumor cells and surrounding<br />

tissues - a critical feature in understanding cancer development and<br />

immune evasion. Imaging modality such as bioluminescence lacks the<br />

resolution necessary to discern subtle structural differences and heterogeneity<br />

in the tumor niche. Microscopic examination <strong>of</strong> fixed specimens is<br />

devoid <strong>of</strong> the 3-dimensional context or evolution <strong>of</strong> tumor progression<br />

within the same host. Methods: New insights have come from studies<br />

involving the use <strong>of</strong> intravital 2-photon laser scanning microscopy (2P-<br />

LSM), which allows deep visualization (�300um) with single-cell resolution<br />

(�1um), thus enables direct observation <strong>of</strong> cellular behavior in intact<br />

tissues at a suitable dynamic spatial-time resolution. We study the role <strong>of</strong><br />

tumor niche in shaping immune repertoire and develop strategies to modify<br />

tumor niche to enhance anti-tumor immunity. Results: One example is our<br />

study <strong>of</strong> glioblastoma multiforme (GBM), which contains a cellular hierarchy<br />

with a CD133� sub-population representing self-renewing and tumorigenic<br />

GBM stem cells (GSCs). In a xeno-transplant model, GSC was<br />

capable <strong>of</strong> tumor initiation in the mouse brain. To directly test the relative<br />

tumorigenic potential <strong>of</strong> GSCs (CD133�) and non-GSCs (CD133- ), we<br />

inoculated paired tumor populations from the same primary GBM tumor<br />

cells and monitored tumor competition by serial 2P-LSM through implanted<br />

cranial windows. Serial 2P-LSM imaging shows that after 35 days,<br />

GBM formation was driven exclusively by GSCs but not non-GSCs. To<br />

interrogate tumor-associated immune responses, we inoculated syngeneic<br />

mouse glioma tumors into C57BL/6 mice. Using this and CNS-inflammatory<br />

models, we have begun to undercover the role <strong>of</strong> perivascular<br />

antigen-presenting cells and microglia in guiding the recruitment <strong>of</strong><br />

CNS-bound lymphocytes. Conclusions: Our data provide the first direct<br />

functional evidence that CSCs are responsible for tumor propagation in<br />

GBM, and represent an in vivo experimental platform to monitor immunotherapeutic<br />

interventions.<br />

9522 Poster Discussion Session (Board #2), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Outcome <strong>of</strong> 116 cases <strong>of</strong> pleuropulmonary blastoma type I and type Ir<br />

(regressed): A report from the International PPB Registry (IPPBR). Presenting<br />

Author: Yoav H. Messinger, Children’s Hospitals and Clinics <strong>of</strong><br />

Minnesota, Minneapolis, MN<br />

Background: Pleuropulmonary blastoma (PPB) is a rare dysembryonic lung<br />

neoplasm <strong>of</strong> early childhood with progression from a purely cystic Type I<br />

(T-I) lesion to cystic solid and solid high grade sarcoma (Type II and Type<br />

III). A regressed form <strong>of</strong> PPB (T-Ir) has been recognized pathologically. The<br />

outcome <strong>of</strong> both T-I and T-Ir has been only partially described. Methods:<br />

Retrospective analysis <strong>of</strong> 345 IPPBR cases showed 116 T-I or T-Ir. In all<br />

cases the PPB diagnosis was made on surgically removed cysts. The<br />

treating physician decided whether to use chemotherapy after surgery.<br />

Results: The pathologic diagnosis <strong>of</strong> the 91 PPB T-I and 25 T-Ir is now<br />

confirmed by central review (LPD and DAH). Patients with T-I were younger<br />

than T-Ir (median: 8 months vs. 48 months).Diagnosis after age 6 years<br />

included only one T-I compared to 10 T-Ir patients. Therapy is not known<br />

for 28 T-I and 2 T-Ir. Surgery was followed by chemotherapy in 31 T-I and 2<br />

T-Ir. Six (5%) recurred with the same type, all were alive at last follow-up: 5<br />

(5.5%) T-I, 1 (4%) T-Ir. Progression to high-grade Type II or III occurred in<br />

9/91 (10%) T-I and 2/25 (8%) T-Ir. The addition <strong>of</strong> chemotherapy did not<br />

significantly reduce progressions (Fisher’s exact test). All <strong>of</strong> the tumor<br />

progressions were seen by 75 months <strong>of</strong> age; this finding is similar to<br />

broader IPPBR data: � 95% <strong>of</strong> patients are diagnosed with Type II/III by<br />

72 months <strong>of</strong> age. Of the 9 patients with T-I who progressed, 5 ultimately<br />

died, whereas the 2 T-Ir who progressed were alive. At last follow-up<br />

111/116 (95.6%) were alive. Conclusions: A cyst in an older individual<br />

most likely will be Type Ir. Type I and Type Ir are clinically similar with a<br />

small risk <strong>of</strong> progression to the advanced Type II/III up to 6 years <strong>of</strong> age.<br />

Outcome for those whose cystic PPBs progressed is poor. The role <strong>of</strong><br />

chemotherapy remains uncertain for the prevention <strong>of</strong> progression in the<br />

pure cystic PPB Type I or Ir.<br />

Upfront chemotherapy and progression.<br />

Progression No progression Total<br />

Type I:<br />

Surgery � chemotherapy 2 29 31*<br />

Surgery only 6 25 31<br />

Unknown<br />

Type Ir:<br />

1 27 28<br />

Surgery � chemotherapy 1 1 2**<br />

Surgery only 1 19 20<br />

Unknown 0 2 2<br />

Excluding 2 chemotherapy treated patients: for 2nd malignancy*, and for concurrent rhabdomyosarcoma**<br />

Pediatric Oncology<br />

611s<br />

9521 Poster Discussion Session (Board #1), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Effect <strong>of</strong> radiation on outcome <strong>of</strong> types II and III PPB: A report from the<br />

International Pleuropulmonary Blastoma Registry. Presenting Author:<br />

Gretchen M. Williams, Children’s Hospitals and Clinics <strong>of</strong> Minnesota,<br />

Minneapolis, MN<br />

Background: Pleuropulmonary blastoma (PPB) is a rare dysembryonic lung<br />

neoplasm <strong>of</strong> early childhood with progression from a purely cystic (Type I)<br />

lesion to a solid high-grade sarcoma Type III (T-III), or a mixed solid/cystic<br />

stage Type II (T-II). Surgery and chemotherapy are required for T-II and<br />

T-III PPB, but the benefit <strong>of</strong> radiation therapy for T-II and T-III PPB is<br />

debated and is being evaluated in this study. Methods: This is a retrospective<br />

analysis <strong>of</strong> central pathology-reviewed Types II and III PPB from the<br />

IPPBR. Treatments were chosen by physician, were not randomized, and<br />

included surgery and chemotherapy. The outcome <strong>of</strong> patients treated with<br />

upfront radiation (before progression or relapse) was compared to patients<br />

who did not receive radiation. Event-free survival (EFS) and overall survival<br />

(OS) were determined to last follow-up, using the Kaplan-Meier analysis,<br />

with log-rank test. Results: Outcome for 212 patients (117 Type-II and 95<br />

Type III) was significantly better for T-II than T-III: EFS for T-II was 68.1%<br />

vs. T-III 45.7% (P�0.002), and OS for T-II was 75.2% vs. T-III 57.9%<br />

(p�0.01). Excluding patients with incomplete therapy information, 174<br />

were treated with surgery and chemotherapy. Of these 44 (25%) also<br />

received radiation therapy; 130 patients did not. The table below shows<br />

comparison <strong>of</strong> radiated vs. non-radiated. Radiation provided no additional<br />

survival benefit, overall or by tumor Type. Multivariate analysis showed that<br />

gender and extent <strong>of</strong> primary tumor resection (biopsy vs. gross total<br />

resection) had no effect on outcome, however T-II vs. T-III retained<br />

prognostic significance. Conclusions: For this collection <strong>of</strong> advanced-type<br />

PPB, outcome is significantly better for T-II than T-III. For patients treated<br />

initially with surgery and chemotherapy, adding radiation therapy <strong>of</strong>fered<br />

no improvement in event-free or overall survival. Limitations <strong>of</strong> this study<br />

include its retrospective nature and non-uniform treatment regimens.<br />

Non-radiated Radiated P (log-rank)<br />

All patients (n�174) EFS 55.5% 46.1% 0.312<br />

OS 63.1% 63.6% 0.952<br />

Type II (n�90) EFS 65.8% 46.7% 0.096<br />

OS 70.7% 66.7% 0.651<br />

Type III (n�84) EFS 41.8% 46.4% 0.452<br />

OS 52.7% 62.1% 0.405<br />

9523 Poster Discussion Session (Board #3), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Increase in the incidence <strong>of</strong> differentiated thyroid carcinoma in children,<br />

adolescents, and young adults (AYA): A population-based study. Presenting<br />

Author: Lucas B. Vergamini, Faculdade de Ciencias Medicas da Santa Casa<br />

de Sao Paulo, Sao Paulo, Brazil<br />

Background: Studies have shown an increase in thyroid cancer incidence<br />

among adults since the 1990s. However, few studies have evaluated this<br />

occurrence among children and AYA. Increases resulting from enhanced<br />

detection are most likely to involve small tumors. The objective <strong>of</strong> this study<br />

is to investigate trends in incidence <strong>of</strong> differentiated thyroid carcinomas in<br />

children and AYA by size and sex. Methods: This is an ecological time-trend<br />

study. Cases <strong>of</strong> differentiated thyroid cancer (1984-2008) in patients<br />

younger than 30 years old were selected from SEER 9 cancer registries<br />

using International Classification <strong>of</strong> Diseases for Oncology 3rd edition<br />

(ICD-O-3) codes for papillary and follicular cancers (codes 8050/3,<br />

8052/3, 8130/3, 8260/3, 8290/3, 8330-8332/3, 8335/3, 8340-<br />

8344/3, 8450/3 and 8452/3). Patients who had multiple primary tumors<br />

were excluded from the study. SEER*Stat s<strong>of</strong>tware was used to calculate<br />

age-standardized rates (estimated per 1,000,000 persons; World Standard<br />

Population) and annual percentage changes (APC) were calculated using<br />

Joinpoint model. Results: Rates ranged from 2.77 (1990) to 7.45 (2002)<br />

for males and from 17.19 (1987) to 41.3 (2008) for females. Overall, a<br />

significant increasing trend in incidence was observed for females<br />

(APC�3.20, 95%CI 2.80, 3.60). When a stratified analysis based on<br />

tumor size was performed, significant increasing trends were noted for the<br />

following categories: 0.5-0.9 cm (Males: APC�3.50, 95%CI 1.50, 5.40;<br />

Females: APC�7.30, 95%CI 5.90, 8.80), 1.0-1.9 cm (Males: APC�3.20,<br />

95%CI 1.00, 5.40; Females: APC�2.90, 95%CI 2.20, 3.70), and � 2cm<br />

(Males: APC�1.30, 95%CI 0.30, 2.40; Females: APC�2.50, 95%CI<br />

1.70, 3.20). However, no statistically significant trends were noted for<br />

tumors �0.5 cm (Males: APC�2.50, 95%CI -0.30, 5.40; Females:<br />

APC�2.0, 95%CI -6.90, 11.80). Conclusions: Incidence rates for differentiated<br />

thyroid carcinoma are also increasing among children and AYA in the<br />

United States. The absence <strong>of</strong> increasing trends for small tumors (� 0.5cm)<br />

rules out diagnostic scrutiny as the only explanation for the observed<br />

results. Environmental, dietary and genetic factors should be investigated.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


612s Pediatric Oncology<br />

9524 Poster Discussion Session (Board #4), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Treatment <strong>of</strong> pediatric stage IA lymphocyte-predominant Hodgkin lymphoma<br />

with surgical resection alone: A report from the Children’s Oncology<br />

Group. Presenting Author: Burton Appel, Hackensack University Medical<br />

Center, Hackensack, NJ<br />

Background: Lymphocyte-predominant Hodgkin lymphoma (LPHL) is an<br />

uncommon histologic subtype comprising up to 10% <strong>of</strong> cases in pediatric<br />

series. Patients with LPHL typically present with low stage disease and are<br />

responsive to regimens used for classical HL. Recurrence is uncommon;<br />

secondary malignant neoplasms or evolution to non-Hodgkin lymphoma are<br />

more common events. Small retrospective studies have suggested that<br />

some patients with stage I LPHL can be cured with surgery alone. We report<br />

the results <strong>of</strong> a large prospective study using a treatment algorithm in which<br />

a selected subset <strong>of</strong> patients received surgery alone. Methods: Patients ages<br />

0-21 years with newly-diagnosed, low risk LPHL were eligible for<br />

AHOD03P1. Low risk was defined as clinical Stage IA and IIA in the<br />

absence <strong>of</strong> bulk disease (a mediastinal mass � 1/3 <strong>of</strong> the thoracic diameter<br />

or a nodal aggregate � 6 cm). Central pathology review was performed to<br />

confirm LPHL histology. Baseline evaluations included CT and FDG-PET.<br />

Patients with Stage IA disease in a single node that was completely<br />

resected were observed without further therapy. Total resection (TR) was<br />

confirmed by rapid central review <strong>of</strong> CT and FDG-PET. In some cases the<br />

extent <strong>of</strong> resection was equivocal; repeat imaging 6-7 weeks after initial<br />

evaluation was used to allocate such patients to either observation or<br />

chemotherapy. Patients who recurred were assigned to treatment with 3<br />

cycles <strong>of</strong> AV-PC (doxorubicin/vincristine/prednisone/cyclophosphamide).<br />

Results: Between January 2006 and November 2010, 52 patients with<br />

Stage IA, single node LPHL were enrolled with a confirmed TR. Nine<br />

patients have experienced a relapse; 8 were Stage I and 1 was Stage II at<br />

relapse. The median time to relapse was 10 (range 1-17) months. The<br />

median follow up among the 43 remaining patients is 26 (range 4-60)<br />

months. The current 2 year EFS estimate among these patients is 80.3%<br />

(95% CI: 65.3% -89.3%). Overall survival for the 52 patients is 100%.<br />

Conclusions: With this strategy <strong>of</strong> surgical resection followed by observation<br />

in a highly select cohort <strong>of</strong> pediatric LPHL, up to 80% <strong>of</strong> patients may be<br />

spared chemotherapy. Follow up is limited but overall survival to date is<br />

excellent.<br />

9526 Poster Discussion Session (Board #6), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Dietary intake and metabolic syndrome in adult survivors <strong>of</strong> childhood<br />

cancer. Presenting Author: Webb A. Smith, St. Jude Children’s Research<br />

Hospital, Memphis, TN<br />

Background: Childhood cancer survivors (CCS) are at increased risk for<br />

metabolic syndrome (MetSyn) and increased morbidity and mortality from<br />

cardiovascular disease. Following a heart healthy diet may decrease this<br />

risk. The purpose <strong>of</strong> this investigation was to characterize dietary patterns<br />

and to evaluate the association between diet and MetSyn among CCS.<br />

Methods: CCS who were 10� year survivors <strong>of</strong> childhood cancer, who were<br />

older than 18 years <strong>of</strong> age, and participating in the St. Jude Lifetime Cohort<br />

Study (SJLIFE). They completed a medical assessment based on Children’s<br />

Oncology Group screening guidelines, and a food frequency questionnaire<br />

(FFQ). Laboratory and physical measures were obtained to determine<br />

MetSyn status using the NCEP-ATPIII criteria. <strong>Part</strong>icipants were classified<br />

as having MetSyn if they met the criteria for �3 components <strong>of</strong> MetSyn.<br />

Data from the FFQ were used to score dietary patterns according to<br />

WCRF/AICR recommendations. Those who met �4 <strong>of</strong> the 7 recommendations<br />

were classified as following a “healthy diet.” A stratified analysis by<br />

sex using log-binomial regression models was undertaken to evaluate<br />

associations between diet and MetSyn, adjusted for age, age at diagnosis,<br />

cranial radiation, education, household income, and smoking status.<br />

Results: Among 1421 participating CCS (49.3% male, median age 33.0<br />

years, range, 18.9-60.0 years), 31.5% met the criteria for MetSyn and<br />

25.8 % followed a healthy diet according to the WCRF/AICR recommendations.<br />

In multiple regression models stratified by sex, females who followed<br />

a “healthy diet” were 2.1 (95% CI 1.5-2.9) times less likely and males who<br />

followed a “healthy diet” were 2.2 (95% CI 1.5-3.1) times less likely to<br />

have MetSyn than those who reported following a “healthy diet”. Conclusions:<br />

Heart healthy diets in CCS are associated with decreased risk for MetSyn.<br />

Dietary interventions may be warranted in CCS with MetSyn.<br />

9525 Poster Discussion Session (Board #5), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Family history <strong>of</strong> cancer and clinical outcome in pediatric oncology.<br />

Presenting Author: Joshua David Schiffman, Huntsman Cancer Institute,<br />

University <strong>of</strong> Utah, Salt Lake City, UT<br />

Background: We previously reported in a small study (n�363) that children<br />

diagnosed with cancer who have a family history <strong>of</strong> cancer (FHC) may have<br />

worse prognosis than those without FHC, specifically children diagnosed<br />

with Hodgkin Disease (HD) and acute lymphoblastic leukemia (ALL)<br />

(Eichstadt et al., ASCO 2009). The Utah Population Database (UPDB), a<br />

unique resource, includes a Surveillance Epidemiology and End Results<br />

(SEER) registry (the Utah Cancer Registry) record-linked to extensive<br />

genealogies for 6.5 million people in which most Utah families are<br />

represented. A majority <strong>of</strong> pedigrees include �3 generations and several<br />

span 7� generations. The UPDB provides an accurate, objective assessment<br />

<strong>of</strong> FHC. We expanded our original study by examining cancer<br />

mortality <strong>of</strong> 4,482 pediatric cases (age �18) diagnosed from 1966-2009<br />

with adequate follow-up (median 9.5 years) and family information.<br />

Methods: We compared survival from cancer-caused mortality for pediatric<br />

cases diagnosed from 1966-2009 with FHC <strong>of</strong> any cancer diagnosed �80<br />

yo in 1st-degree, 2nd-degree, or 3rd-degree relatives to pediatric cases<br />

without FHC. Nonparametric estimates <strong>of</strong> survivor function were determined<br />

by the life-table method and a log-rank test <strong>of</strong> homogeneity was used<br />

to test for differences between pediatric probands with FHC compared to no<br />

FHC. Results: Overall survival was significantly lower in children with<br />

positive FHC (n�1,128) than in children with no FHC (n�3,354) independent<br />

<strong>of</strong> stage (p�0.0001; 10yr survival, 69% with FHC vs.78% with no<br />

FHC). We observed lower survival in children with any leukemia and FHC<br />

(n�279) compared to children with leukemia and no FHC (n�871;<br />

p�0.0001). In particular, cases with ALL and FHC had lower survival<br />

(p�0.0001; 10yr survival, 61% vs. 75%). Pediatric lymphoma cases had<br />

lower survival when positive for FHC (n�150) vs. no FHC (n�428;<br />

p�0.0001), specifically for an HD diagnosis (p�0.0004; 10yr survival,<br />

83% vs. 94%). Conclusions: Pediatric cancer cases with FHC <strong>of</strong> any cancer<br />

(children or adults) have decreased survival compared to cases with no<br />

FHC, specifically in children diagnosed with ALL or HD, thus validating our<br />

earlier findings. Familial factors likely contribute to increased pediatric<br />

cancer mortality.<br />

9527 Poster Discussion Session (Board #7), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Association <strong>of</strong> bone mineral density with incidental renal stone in long-term<br />

survivors <strong>of</strong> childhood acute lymphoblastic leukemia. Presenting Author:<br />

Prasad Laxman Gewade, St. Jude Children’s Research Hospital, Memphis,<br />

TN<br />

Background: With 5-year survival <strong>of</strong> childhood acute lymphoblastic leukemia<br />

(ALL) now exceeding 90%, long term morbidities are <strong>of</strong> growing<br />

concern. Both low bone mineral density (BMD) and renal dysfunction have<br />

been reported in ALL survivors. Our objective was to evaluate the association<br />

between low BMD and incidental renal stones, a known predictor <strong>of</strong><br />

renal dysfunction. Methods: Adult participants who were 10� year survivors<br />

<strong>of</strong> childhood ALL and members <strong>of</strong> St. Jude Lifetime Cohort study were<br />

recruited between 12/2007 and 3/2011. During their risk-based medical<br />

evaluations they underwent quantitative computed tomography (QCT) to<br />

evaluate BMD. Incidental renal stones were identified by radiologists’<br />

review <strong>of</strong> axial QCT source images. Demographic information was abstracted<br />

from responses to health surveys and dietary intake from a Block<br />

food frequency questionnaire. Association between BMD and renal stones<br />

was evaluated in a multivariable logistic regression model. Confounding<br />

variables were selected using directed acyclic graphs and change in effect<br />

estimates strategy. Results: At a median <strong>of</strong> 26.1 years from diagnosis, BMD<br />

Z score <strong>of</strong> � 1 standard deviation (SD) was detected in 77/662 (11.6%)<br />

and renal stones in 73/662 (11%) participants. In a multivariable model<br />

adjusted for age, dietary vitamin D and renal radiation, when compared to<br />

BMD Z score � 1 SD, the risk <strong>of</strong> renal stones increased with a decrease in<br />

BMD Z-score; 1 to 0 SD (Odds Ratio (OR), 1.93; 95% confidence interval<br />

(CI), 0.69 to 5.41), 0 to -1 SD (OR, 1.46; 95% CI, 0.52 to 4.05), -1 to -2<br />

SD (OR, 2.72; 95% CI, 0.81 to 6.41), and � -2 SD (OR, 4.96; 95% CI,<br />

1.43 to 17.13). Older age (45-54 vs.18-24 y; OR, 3.66; 95% CI, 1.10 to<br />

12.15), renal radiation (OR, 1.97; 95% CI, 0.59 to 6.50) and � 141.5 IU<br />

intake <strong>of</strong> vitamin D (OR, 1.65; 95% CI, 0.98 to 2.77) were also associated<br />

with renal stone formation. Conclusions: Our results not only help us<br />

recognize survivors at risk, but also informs radiologists to be vigilant <strong>of</strong><br />

incidental renal stones among older ALL survivors with low BMD.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


9528 Poster Discussion Session (Board #8), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Renal carcinoma following therapy for cancer in childhood: A report from<br />

the Childhood Cancer Survivor Study. Presenting Author: Carmen Louise<br />

Wilson, St. Jude Children’s Research Hospital, Memphis, TN<br />

Background: Limited data exists describing the incidence <strong>of</strong> and risk factors<br />

for subsequent renal carcinoma among long-term survivors <strong>of</strong> childhood<br />

cancer. Methods: The study included 14,351 five-year survivors <strong>of</strong> childhood<br />

cancer diagnosed between 1970 and 1986 who participated in the<br />

Childhood Cancer Survivor Study. Chemotherapy and radiotherapy exposures<br />

were abstracted from medical records; total dose <strong>of</strong> radiation to the<br />

renal beds was estimated by a radiation physicist. Standardized incidence<br />

ratios (SIRs) were calculated using age-, sex-, and calendar-specific<br />

incidence data from the Surveillance, Epidemiology and End Results<br />

(SEER) program. Cumulative incidence was calculated treating death as a<br />

competing risk. Poisson regression analyses were used to assess associations<br />

between diagnosis and treatment characteristics and the risk <strong>of</strong><br />

subsequent renal carcinoma while adjusting for changes in risk due to age.<br />

Results: Twenty-six survivors were diagnosed with a renal carcinoma at a<br />

median follow-up <strong>of</strong> 19.3 years (range: 1 month to 34.3 years) from study<br />

entry at 5 years post diagnosis. Eight patients received �5Gy radiotherapy<br />

to a renal bed, 16 received chemotherapy, seven <strong>of</strong> whom seven received<br />

both radiotherapy �5Gy and chemotherapy. Cumulative incidence <strong>of</strong> renal<br />

carcinoma at 20 years was 0.16% (95% CI 0.12-0.20). The SIR was 8.1<br />

(95% CI 5.3-11.8) comparing survivors to the general population. Highest<br />

risk for renal carcinoma was observed among survivors <strong>of</strong> neuroblastoma<br />

(SIR 87.1, 95% CI 38.4-175.2), non-Hodgkin lymphoma (SIR 9.3, 95%<br />

CI 1.9-27.4), and bone tumors (SIR 7.0, 95% CI 1.4-20.4). In multivariable<br />

analyses, the risk <strong>of</strong> subsequent renal carcinoma was elevated among<br />

survivors exposed to platinum-based chemotherapy (Rate Ratio 3.1, 95%<br />

CI 0.9-10.6) or renal bed radiotherapy �5Gy (RR 3.6, 95% CI 1.5-8.4).<br />

Conclusions: While cumulative incidence is low, survivors <strong>of</strong> childhood<br />

cancer are at an eight-fold increased risk for subsequent renal carcinoma<br />

compared to the general population. In addition to a primary diagnosis <strong>of</strong><br />

neuroblastoma, radiotherapy directed to the renal bed increased the risk for<br />

renal carcinoma.<br />

9530 Poster Discussion Session (Board #10), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Effect <strong>of</strong> dexraoxane on impaired mitochondrial structure and function in<br />

doxorubicin-treated childhood ALL survivors. Presenting Author: Steven E.<br />

Lipshultz, Department <strong>of</strong> Pediatrics, University <strong>of</strong> Miami Miller School <strong>of</strong><br />

Medicine, Miami, FL<br />

Background: Cardiotoxicity in doxorubicin (DOX)-treated long-term childhood<br />

cancer survivors is characterized by impaired cardiac function which<br />

is partly mediated by mitochondrial (mt) energy production. Preclinical<br />

studies show irreversible DOX-associated cardiac mt dysfunction. A response<br />

to impaired mt function is to increase the mtDNA copies/cell<br />

(mtDNA). We examined mtDNA and function in peripheral blood mononuclear<br />

cells (PBMCs) in childhood survivors <strong>of</strong> high-risk ALL who received<br />

DOX alone or DOX plus dexrazoxane (DOX/DEX). Methods: Patients with HR<br />

ALL treated on DFCI Childhood ALL protocols from 1987-2005 were<br />

eligible for this study. PBMCs mtDNA and oxidative phosphorylation<br />

(OXPHOS) NADH dehydrogenase (CI) and cytochrome c oxidase (CIV)<br />

activities were measured by RT-PCR and immunoassays respectively. A<br />

Wilcoxon rank-sum test was used to compare continuous measures between<br />

groups. Models for mitochondrial parameters were based on maximum<br />

likelihood variance component estimation and were adjusted for<br />

patient characteristics. Results: 64 patients provided samples at a median<br />

follow-up <strong>of</strong> 7.8 (2.9-30.2) years. 58% received DOX/DEX. Median<br />

cumulative DOX dose was 300 mg/m2 . A significant difference was<br />

detected between groups for mtDNA (p�0.001, adjusted p�0.015).<br />

MtDNA medians for the DOX and DOX/DEX groups were 1106.3 and 310.5.<br />

No significant differences were found between groups for CI or CIV<br />

activities. Conclusions: DOX-treated survivors have increased mtDNA consistent<br />

with impaired mt function, which is abrogated by concurrent use <strong>of</strong><br />

DEX. The use <strong>of</strong> DOX/DEX is associated with lower mtDNA. Due to a<br />

compensatory increase in mtDNA to augment mt function, overall mt<br />

function is not different between groups. This compensatory mechanism in<br />

7.8-year survivors at risk for cardiac dysfunction is concerning over their<br />

subsequent lifespan.<br />

N<br />

DOX<br />

Median (range) N<br />

DOX/DEX<br />

Median (range) P<br />

mtDNA 27 1106.3 (144.2-6746.8) 35 310.5 (15.3-1859.2) 0.001<br />

CI activity (OD/mg x 10 3 ) 25 10.5 (5.0-31.3) 33 11.7 (5.0-41.4) 0.97<br />

CIV activity (OD/mg x 10 3 ) 25 9.8 (5.0-20.1) 34 8.1 (4.7-23.4) 0.40<br />

Pediatric Oncology<br />

613s<br />

9529 Poster Discussion Session (Board #9), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

A longitudinal study <strong>of</strong> anthracycline cardiotoxicity in pediatric Ewing<br />

sarcoma. Presenting Author: Tanya Renae Brown, British Columbia Children’s<br />

Hospital, Vancouver, BC, Canada<br />

Background: Trends in cardiac function over time have not been previously<br />

described in Ewing sarcoma. Our cohort is unique as all the cardiac<br />

evaluations occurred systematically in one tertiary cardiology unit during a<br />

28 year period. Objectives: 1.To evaluate the cardiac functional trends in<br />

the cohort at baseline, during treatment and in surveillance. 2.To evaluate<br />

the risk factors associated with cardiac toxicity. Design: Received ethics<br />

approval for a retrospective chart review <strong>of</strong> all patients less than 17 years,<br />

diagnosed with Ewing sarcoma from 1978-2006, treated initially at British<br />

Columbia Children’s Hospital . Methods: There was complete data on 71/79<br />

eligible patients. Echocardiograms were recorded at 5 time points: pre<br />

treatment, worst function during treatment, on therapy completion, worst<br />

function during surveillance and the most recent echocardiogram. Cardiac<br />

toxicity was graded using CTCAE v 4.0. The statistical analysis with SAS<br />

s<strong>of</strong>tware v 9.2 calculated the chi square and odds ratios to explore a<br />

possible association between exposure and outcome. Results: Median age<br />

at diagnosis 11.1 yrs (2.2-16.1 yrs), 52% female, metastatic in 17, overall<br />

survival 74% at 10yrs. A total <strong>of</strong> 397 echocardiograms were performed with<br />

244 (61%) being normal and 153 (39%) being abnormal. The median<br />

cumulative dose <strong>of</strong> anthracyclines was 365mg/m2 . Grade 1-5 cardiomyopathy<br />

occurred in 42 (59%) patients. Grade 3-5 toxicity occurred in 11<br />

(15%) patients on chemotherapy completion increasing to 23 (32%) 120<br />

months post diagnosis. The median time to the worst cardiac function was<br />

51months (2-183). Four <strong>of</strong> 19 (20%) deaths were cardiac related<br />

including 2/3 cardiac transplants. Conclusions: Symptomatic cardiac<br />

toxicity increased from 20% noted in our earlier Ewings cohort (Kakader et<br />

al. 1997) to 32% with longitudinal evaluation assessments. Significant<br />

and progressive cardiac toxicity occurred after completion <strong>of</strong> chemotherapy.<br />

9531 Poster Discussion Session (Board #11), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Tumor location and neurocognitive impairment in adult survivors <strong>of</strong><br />

pediatric brain tumors: A report from the St. Jude Lifetime Cohort (SJLIFE).<br />

Presenting Author: Tara M. Brinkman, St. Jude Children’s Research<br />

Hospital, Memphis, TN<br />

Background: Follow-up guidelines identify supratentorial tumor location as<br />

a risk factor for poor neurocognitive outcomes during childhood; yet few<br />

studies have systematically compared long-term cognitive outcomes between<br />

adult survivors <strong>of</strong> childhood infratentorial and supratentorial brain<br />

tumors. Methods: Neurocognitive functions were evaluated in 130 adult<br />

survivors <strong>of</strong> pediatric brain tumors (58 supratentorial and 72 infratentorial,<br />

mean [SD] current age � 27.4 years [5.2], age at diagnosis � 8.6 years<br />

[4.6], and time since diagnosis � 18.8 years [4.8]) participating in the<br />

SJLIFE long-term follow-up protocol. Age-adjusted standard scores for<br />

measures <strong>of</strong> intelligence, attention, memory, processing speed, and executive<br />

functioning were calculated, with clinical impairment defined as scores<br />

�10th percentile. Odds ratios (OR) and 95% confidence intervals (CI) were<br />

calculated using multivariable logistic regression models to examine<br />

associations between neurocognitive functions and tumor location. Results:<br />

As a group, survivors performed below average across multiple neurocognitive<br />

domains, including full scale IQ (mean�88.1; SD�18.2), with 34%<br />

demonstrating impaired IQ. Survivors <strong>of</strong> infratentorial tumors were more<br />

likely to be impaired on measures <strong>of</strong> focused attention (OR�2.19, 95%<br />

CI�1.03-4.65) and fine motor dexterity (OR�2.62, 95% CI�1.21-5.66)<br />

compared to survivors <strong>of</strong> supratentorial tumors. After adjusting for sex, age<br />

at diagnosis, shunt placement and cranial radiation (yes/no), infratentorial<br />

tumor location was only associated with reduced performance on a task <strong>of</strong><br />

visual abstract reasoning (OR�3.76, 95% CI�1.40-10.1). Cranial radiation<br />

therapy was independently associated with impaired short-term<br />

memory (OR�15.6, 95% CI�1.64-147.8) and processing speed<br />

(OR�3.86, 95% CI�1.15-13.0). Conclusions: Tumor location was not<br />

associated with neurocognitive impairment after adjusting for treatment<br />

exposures. To further delineate potential differences associated with tumor<br />

location, future studies will examine factors including radiation dose/<br />

volume, extent <strong>of</strong> surgical resection, and medical complications.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


614s Pediatric Oncology<br />

9532 Poster Discussion Session (Board #12), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Parent-reported cognition and its clinical applications in pediatric oncology.<br />

Presenting Author: Jin-Shei Lai, Northwestern University, Chicago, IL<br />

Background: Parent-reported cognitive function (PCF) <strong>of</strong> their child’s<br />

cognitive abilities is <strong>of</strong>ten used to trigger referral for comprehensive<br />

neuropsychological evaluation. The purpose <strong>of</strong> this study was to evaluate<br />

the clinical utility <strong>of</strong> PCF to predict impairment as measured by neuropsychological<br />

assessment and to identify factors associated with PCF. Methods:<br />

565 patients (53% brain tumor, BT; 47% other types <strong>of</strong> cancer, non-BT)<br />

aged 7-21 (mean�14 yrs; 56% males) and their parents were recruited.<br />

34% received radiation therapy, 72% chemotherapy and 71% surgery.<br />

Mean years since diagnosis� 5.7 yrs. PCF was measured using a 43-item<br />

pediatric perceived cognitive function item bank (pedsPCF). Parents<br />

completed the pedsPCF and a single item to rate their child’s quality <strong>of</strong> life<br />

(QOL). Patients completed NINDS-NeuroQOL Depression, PedsQL Fatigue<br />

scales and neuropsychological tests (NPT) <strong>of</strong> psychomotor function,<br />

attention, learning and working memory using the CogState. K-Means<br />

clustering was used to group patients based on scores <strong>of</strong> depression,<br />

fatigue and pedsPCF. Results: PedsPCF significantly differentiated BT from<br />

non-BT, t�5.65, p�.01. Correlations between pedsPCF and NPT ranged<br />

from 0-0.66 (BT, � 1 yr diagnosis, psychomotor), depending on BT (vs<br />

non-BT), yrs since diagnosis and treatment, and NPT. Three clusters were<br />

identified with its own unique characteristics. Specifically, pedsPCF was<br />

significantly correlated w/ depression & fatigue for cluster 1; correlated w/<br />

NPT scores for non-BT for cluster 2; and correlated w/ NPT for BT for<br />

cluster 3. Cluster membership <strong>of</strong> patients were significantly differentiated<br />

by Karn<strong>of</strong>sky rating, surgery (yes/no), QOL, parent education, child age,<br />

and child gender, but not types <strong>of</strong> treatment, and grade repetition (yes/no).<br />

Conclusions: This is one <strong>of</strong> the first studies to report an association between<br />

perceived cognitive function, using parent ratings, and neuropsychological<br />

performance. PedsPCF demonstrated clinical utility in differentiating<br />

between children with a brain tumor from children with other cancer types.<br />

PedsPCF has the potential to serve as a screening tool to facilitate efficient<br />

referral for comprehensive neurocognitive assessment.<br />

9534 Poster Discussion Session (Board #14), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Genome-based outcome prediction in MYCN nonamplified high-risk neuroblastoma.<br />

Presenting Author: Andres Eduardo Morales La Madrid, University<br />

<strong>of</strong> Chicago, Chicago, IL<br />

Background: Less than 40% <strong>of</strong> children with high-risk neuroblastoma<br />

achieve long-term survival, and at diagnosis, it is not possible to identify<br />

patients who will be cured. Microarray studies have proposed expression<br />

signatures associated with outcome within high-risk cohorts. However,<br />

integrating this technology as a clinical test has been difficult, in part due<br />

to the lack <strong>of</strong> available frozen tissue and high quality RNA. The nCounter<br />

overcomes this obstacle, using formalin-fixed paraffin embedded tissue<br />

(FFPE). Our objective is to test the correlation <strong>of</strong> a previously published<br />

“ultra high-risk” microarray gene signature developed in the MYCN<br />

nonamplified high-risk population (Asgharzadeh et al, J Natl Cancer Inst,<br />

2006) with the gene expression signature obtained with the nCounter<br />

(NanoString Technologies) using RNA isolated from FFPE MYCN nonamplified<br />

high-risk neuroblastoma samples. Methods: FFPE tumor samples<br />

linked to clinical outcome data were obtained from 6 collaborative<br />

institutions. Tumor content <strong>of</strong> each sample was assessed morphologically.<br />

RNA was isolated using the RNeasy FFPE-kit. Customized probes corresponding<br />

to the candidate genes were designed by NanoString. Hybridization<br />

reactions were performed in duplicate using 100 ng <strong>of</strong> RNA. Positive<br />

and negative control probes and housekeeping probes were included in<br />

every reaction and were used to normalize data for differences in purification,<br />

hybridization, and capture efficiencies Results: Forty-two MYCN<br />

nonamplified high-risk neuroblastoma samples were analyzed by the<br />

nCounter. The cohort 5-year event-free survival and overall survival were<br />

44.6 �/- 8.0% and 53.8 �/- 8.4% respectively. Highly degraded RNA<br />

(RIN ~ 1.5-2.8) was obtained. Unsupervised clustering and principle<br />

components analysis on normalized expression data showed differential<br />

expression <strong>of</strong> most <strong>of</strong> the genes with clustering <strong>of</strong> cases depending upon<br />

outcome (FDR � 0.05). Conclusions: Our results demonstrate that the<br />

nCounter can yield gene expression pr<strong>of</strong>iles that are similar to microarray<br />

gene signatures. Further investigation <strong>of</strong> the clinical utility the nCounter<br />

technology to prognosticate outcome in patients with high-risk neuroblastoma<br />

is warranted.<br />

9533 Poster Discussion Session (Board #13), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Outcome analysis <strong>of</strong> non-high-risk neuroblastoma patients enrolled on<br />

Children’s Oncology Group trials P9641 and A3961. Presenting Author:<br />

Holly Jane Meany, Children’s National Medical Center, Washington, DC<br />

Background: Patients with non-high-risk neuroblastoma (low- and intermediate-risk)<br />

generally have an excellent event free (EFS) and overall (OS)<br />

survival with current therapy. However, within this heterogeneous patient<br />

population, there are patients that may benefit from further therapy<br />

reduction and patients that may benefit from augmented therapy. Methods:<br />

Survival tree regression analysis was performed in patients enrolled on<br />

P9641 and A3961 with OS the primary endpoint. Univariate Cox proportional<br />

hazards models determined statistically significant prognostic factors.<br />

Secondary analysis <strong>of</strong> cases with MYCN non-amplified, non-high-risk<br />

neuroblastoma classified patients by age, INSS stage, Shimada histology<br />

and genomic features to determine 5-year EFS and OS. Favorable genomics<br />

were defined as hyperdiploid tumors without 1p or 11q loss <strong>of</strong> heterozygosity<br />

(LOH). Those with LOH at 1p or 11q or with a diploid DNA index were<br />

considered unfavorable. Patients without genomic data were excluded.<br />

Results: In the survival tree analysis, ploidy and genomic features were<br />

found to be statistically significant. In the secondary analysis, patients<br />

�18 months <strong>of</strong> age with stage 2 or 3, favorable histology tumors with<br />

favorable genomic pr<strong>of</strong>ile had 5-year EFS and OS rates <strong>of</strong> 92.9�3.9% and<br />

100% respectively. Patients with stage 2 and 3 tumors, unfavorable<br />

histology and unfavorable genomic features had EFS <strong>of</strong> 68.4�8.0% and<br />

OS <strong>of</strong> 78.4�7.0%. Patients �12 months <strong>of</strong> age with stage 4 disease and<br />

either unfavorable histologic or genomic features had EFS <strong>of</strong> 69.1�5.4%<br />

and OS <strong>of</strong> 88.5�3.8%. Conclusions: Excellent outcomes in non-high-risk<br />

neuroblastoma patients with favorable histologic and genomic features<br />

suggest further reduction in therapy is possible in this cohort while<br />

maintaining survival and decreasing side effects. Select patients with<br />

unfavorable features have suboptimal OS and would benefit from modifications<br />

in therapy. Histologic and genomic criteria can be used to identify<br />

patients with non-high-risk neuroblastoma that may benefit from modifications<br />

in therapy. These data support the conduct <strong>of</strong> a cooperative group<br />

clinical trial to refine therapy for patients with non-high-risk disease.<br />

9535 Poster Discussion Session (Board #15), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Relationship <strong>of</strong> fusion protein status and outcome for children with<br />

intermediate-risk rhabdomyosarcoma: A Children’s Oncology Group report.<br />

Presenting Author: Stephen Skapek, University <strong>of</strong> Texas Southwestern<br />

Medical Center, Dallas, TX<br />

Background: Rhabdomyosarcoma (RMS) includes both alveolar (ARMS)<br />

and embryonal (ERMS) subtypes, with ARMS generally having a worse<br />

prognosis. Most ARMS express one <strong>of</strong> two fusion genes generated by<br />

balanced translocations fusing DNA binding domains <strong>of</strong> PAX3 or PAX7 with<br />

FOXO1 (P3F and P7F, respectively). The prognostic value <strong>of</strong> fusion gene<br />

versus histological subtype is not clear. We carried out a multi-institutional<br />

clinical trial through the Children’s Oncology Group (COG) to evaluate this<br />

molecular feature. Methods: All participants were enrolled on COG D9803<br />

from 1999-2005 for children and young adults with intermediate risk<br />

ARMS or ERMS, and were treated with vincristine, dactinomycin, and<br />

cyclophosphamide (�/- topotecan), radiotherapy, and surgery. Diagnostic<br />

specimens were reviewed centrally and fusion gene was assessed by<br />

fluorescence in situ hybridization (FISH), RT-PCR, or both. Event-free<br />

(EFS) and overall survival (OAS) at 5 years (yr) was correlated with histology<br />

and fusion gene status. Results: Of 327 cases included in a re-review <strong>of</strong> the<br />

pathology, 295 could be classified biologically. Cases with ARMS but<br />

unknown trans status (N�23) and those with mixed or RMS-NOS histology<br />

without trans (N�17) and 12 with inadequate clinical data were excluded.<br />

ARMS cases with detectable fusion gene were defined as ARMS P3F�,<br />

ARMS P7F�, while ARMSn was reserved for those without detectable<br />

fusion. P3F and P7F were not observed in ERMS. An additional 189 pts<br />

with ERMS tumors not re-reviewed were also included in the survival<br />

analysis. There was a trend toward better EFS for those with ARMSn than<br />

for ERMS (p�0.15); EFS for ARMS P3F� and P7F� were worse than that<br />

for ERMS (p�0.001). Only ALV P3F� cases had poorer OAS (p�0.004).<br />

Conclusions: ARMSn has an outcome similar to ERMS and a superior EFS<br />

compared to ARMS with P3F or P7F, when given therapy designed for<br />

intermediate risk RMS. This analysis, from a single, prospective trial<br />

restricted to those with non-metastatic disease and with near universal<br />

molecular evaluation, supports incorporating fusion status into treatment<br />

allocation.<br />

ERMS ARMSn ARMS P3F� ARMS P7F�<br />

N <strong>of</strong> patients 305 21 85 23<br />

5-yr EFS 77% 90% 54% 65%<br />

5-yr OAS 82% 89% 64% 87%<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


9536 Poster Discussion Session (Board #16), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Association <strong>of</strong> chromosome complexity with age and metastasis in synovial<br />

sarcomas: Validation <strong>of</strong> expression and genomic prognostic signatures.<br />

Presenting Author: Fred Chibon, Bergonie Cancer Institute, Bordeaux,<br />

France<br />

Background: Synovial sarcoma (SS) is one <strong>of</strong> the rare sarcomas that occurs<br />

in adolescents as well as in adults. Nevertheless, metastasis occurs with a<br />

lowest frequency in the former, 10 vs 50% respectively. In almost all cases,<br />

a characteristic translocation t(X;18)(p11.2;q11.2) exists and the importance<br />

<strong>of</strong> this translocation in SS oncogenesis is well established, but the<br />

genetic basis <strong>of</strong> SS metastasis is still poorly understood. We recently<br />

published expression (CINSARC) and genomic (GI) signatures related to<br />

chromosome integrity control that predict outcome in undifferentiated<br />

sarcomas and GIST and ask whether these signatures could also predict<br />

outcome in SS. Methods: To asses this issue we selected in the European<br />

sarcoma database CONTICABASE 92 primary untreated SS for expression<br />

and genomic pr<strong>of</strong>iling. Results: As demonstrated by metastasis-free survival,<br />

CINSARC and GI have strong and independent prognostic values (p �<br />

1.6x10-5 and p � 4x10-6 , respectively). Comparing expression pr<strong>of</strong>iles <strong>of</strong><br />

tumors with or without metastasis in a training series <strong>of</strong> 58 SS, a 52-genes<br />

signature was identified and validated in an independent series <strong>of</strong> 34 SS.<br />

Fourteen <strong>of</strong> these genes are common with CINSARC and these 52 genes are<br />

involved the same pathways than CINSARC, mitosis checkpoints and<br />

chromosome integrity. Comparing genomic pr<strong>of</strong>iles <strong>of</strong> adult versus pediatric<br />

SS we show that in both situations metastasis is associated to genome<br />

complexity and that the adult genome is highly more frequently rearranged.<br />

In line with this, the pediatric good-prognosis patients, according to GI, do<br />

not develop metastasis. Conclusions: Results clearly indicate that SS<br />

metastasis development is strongly associated with chromosome complexity<br />

and that CINSARC and GI are powerful prognostic factors. Data also<br />

mean that the metastasis frequency difference between adult and children<br />

likely is associated to the genome instability which is highly more frequent<br />

in adults. Finally, among these tree signatures, GI is potentially the best<br />

overall and clearly the most clinically relevant considering that CGH from<br />

FFPE samples is already used in the daily practice <strong>of</strong> pathology.<br />

9538 Poster Discussion Session (Board #18), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Conservative treatment <strong>of</strong> intraocular retinoblastoma: A prospective phase<br />

II randomized trial <strong>of</strong> neoadjuvant chemotherapy followed by local treatments<br />

and chemothermotherapy. Presenting Author: Isabelle Aerts, Institut<br />

Curie, Pediatric Oncology Department, Paris, France<br />

Background: intraocular retinoblastoma treatments <strong>of</strong>ten associate chemotherapy<br />

and focal treatments. The protocols vary and may combine two or<br />

three drugs, and different number <strong>of</strong> cycles associated to the local adjuvant<br />

treatments. A first prospective protocol <strong>of</strong> conservative treatments in our<br />

institution showed the efficacy <strong>of</strong> the use <strong>of</strong> two courses <strong>of</strong> chemoreduction<br />

with etoposide and carboplatin, followed by chemothermotherapy using<br />

carboplatin, as a single agent. In order to decrease the possible long term<br />

toxicity <strong>of</strong> chemotherapy due to etoposide, a prospective randomized phase<br />

II chemo reduction protocol was conducted, using vincristine and carboplatin<br />

vs. etoposide carboplatin. Methods: the study was proposed when initial<br />

tumour size or location did not allow the use <strong>of</strong> front-line local treatments.<br />

The phase II randomized study <strong>of</strong> reduction chemotherapy used vincristin<br />

carboplatin or etoposide carboplatin, followed by local treatment including<br />

chemothermotherapy using the combination carboplatin and laser diode<br />

hyperthermia. Primary endpoint was the need for secondary enucleation or<br />

EBRT not exceeding 40% at 2 years. The new treatment was considered<br />

sufficiently effective if 10 events or less were observed among 33 eyes.<br />

Results: 55 children, 65 eyes were included in the study (May 2004-<br />

August 2009).32 eyes (27 children) were treated conservatively in the arm<br />

etoposide-carboplatin and 33 (28 children) eyes in the arm vincristin<br />

carboplatin.At two years after treatment 23/33 (69.7%) eyes were treated<br />

and salvaged without EBRT or enucleation in the arm vincristin-carboplatin<br />

and 26/32 (81.3%) in the arm etoposide and carboplatin. Conclusions:<br />

neoadjuvant chemotherapy by two cycles <strong>of</strong> vincristine and carboplatin<br />

followed by chemothermotherapy (using the combination carboplatin, as a<br />

single drug, and laser diode hyperthermia) does not seem to <strong>of</strong>fer an<br />

optimal local control.<br />

Pediatric Oncology<br />

615s<br />

9537 Poster Discussion Session (Board #17), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Evaluation <strong>of</strong> local control strategies in patients with localized Ewing<br />

sarcoma <strong>of</strong> bone: A report from the Children’s Oncology Group. Presenting<br />

Author: Steven G. DuBois, University <strong>of</strong> California, San Francisco, San<br />

Francisco, CA<br />

Background: Patients (pts) with Ewing sarcoma (EWS) require local control,<br />

either with surgery alone (S), radiation alone (R), or a combination <strong>of</strong><br />

surgery � radiation (S�R). Optimal choice <strong>of</strong> local control for disease<br />

control remains unclear. Our primary aim was to determine the mode <strong>of</strong><br />

local control associated with the highest event-free survival (EFS). Methods:<br />

Pts with localized EWS <strong>of</strong> bone treated on INT0091, INT0154, or<br />

AEWS0031 phase III trials were included if they had complete local control<br />

data, did not have cranial tumors, received local control starting 2-6<br />

months after enrollment, and were randomized to receive standard dose<br />

5-drug chemotherapy every 3 weeks. We used propensity scores to control<br />

for differences in age, tumor site, and year <strong>of</strong> diagnosis between local<br />

control groups. We constructed Cox models controlling for local control<br />

propensity scores to assess the impact <strong>of</strong> local control type on EFS and<br />

overall survival (OS) from the start <strong>of</strong> local control. Results: 465 pts were<br />

included. Pts selected for S were treated more recently (p � 0.001), more<br />

likely to have appendicular tumors (p � 0.001), and younger (p � 0.02).<br />

Pts treated with R, compared to S, had higher unadjusted risk <strong>of</strong> any event<br />

(HR 1.70; 95% CI 1.18 - 2.44; p � 0.004) or death (HR 1.84; 95% CI<br />

1.18 – 2.85; p � 0.006). Pts treated with S�R, compared to S, had higher<br />

unadjusted risk <strong>of</strong> death (HR 1.75; 95% CI 1.10 – 2.76; p � 0.02). After<br />

adjusting for propensity scores, there was a trend <strong>of</strong> higher risk <strong>of</strong> any event<br />

for pts treated with R (HR 1.42; 95% CI 0.94 – 2.14; p � 0.10) compared<br />

to S, though this was not statistically significant. No other differences in<br />

adjusted risk <strong>of</strong> event or death between local control groups were statistically<br />

significant. We confirmed these results with standard Cox models<br />

using age, tumor site, and year <strong>of</strong> diagnosis as covariates. Conclusions: In<br />

this large group <strong>of</strong> uniformly treated pts, investigator choice <strong>of</strong> local control<br />

approach was not significantly related to EFS. These data support current<br />

practice <strong>of</strong> surgical resection when feasible, while validating radiotherapy<br />

as a reasonable alternative in selected pts.<br />

9539 Poster Discussion Session (Board #19), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Carboplatin (CBP) versus cisplatin (CP) within prognostic groups in<br />

pediatric extracranial malignant germ cell tumors (MGCTs). Presenting<br />

Author: Juliet Hale, Newcastle upon Tyne Hospitals Trust, Newcastle upon<br />

Tyne, United Kingdom<br />

Background: It is established that CBP is inferior to CP in adult MGCTs<br />

when used at AUC 5 or 400mg/m2 . There are no randomised data for<br />

paediatric MGCTs, so conclusions from adult MGCTs have been generalised<br />

despite differences in site, histology and biology. A database, pooling<br />

patients from US and UK paediatric MGCT studies, was established to<br />

determine prognostic factors in paediatric MGCTs. We examined the effect<br />

<strong>of</strong> CBP and CP in identified prognostic groups. Methods: A dataset <strong>of</strong> 1110<br />

paediatric MGCTs was created, treated 1983 to 2009. Treatment was CBP<br />

(JEB, AUC 7.9) in the UK and CP (PEB) in the US. After excluding patients<br />

with surgery only, pure germinoma or immature teratoma , 697 patients<br />

remained. The cure model was used to assess the prognostic significance <strong>of</strong><br />

chemotherapy regimen after adjustment for patient characteristics. Results:<br />

Analysis stratified prognosis according to age (0-10 v 11�yrs) and stage<br />

(1-3 v 4) at diagnosis by site (testis: ovary: extragonadal). Outcome for CBP<br />

and CP in these prognostic groups is shown in the table. Conclusions:<br />

Interpretation <strong>of</strong> the results <strong>of</strong> this nonrandomised comparison requires<br />

caution. However, after adjustment for other prognostic factors, the risks <strong>of</strong><br />

failure for JEB and PEB were not statistically different . This is particularly<br />

true for 0-10yrs. At AUC7.9 CBP is tolerable in paediatric patients, with<br />

potentially fewer late toxicities than CP.<br />

Prognostic group 4-year EFS JEB (95% CI) 4-year EFS PEB (95% CI) P value<br />

Testis 0-10 stage 1-3 0.94 (0.63-0.99) 0.92 (0.83-0.96) 0.79<br />

Testis 0 –10 stage 4 0.82 (0.45-0.95) 0.94 (0.67-0.99) 0.31<br />

Ovary 0-10 stage 1-3 1.00 (0.80-1.00) 1.00 (0.92-1.00) n/a<br />

Ovary 0 -10 stage 4 0.67 (0.05-0.95) 0.80 (0.20-0.97) 0.78<br />

Extragonadal 0-10 stage 1-3 0.92 (0.81-0.97) 0.90 (0.82-0.94) 0.90<br />

Extragonadal 0-10 Stage 4 0.76 (0.60-0.86) 0.83 (0.74-0.90) 0.38<br />

Testis 11� stage 1-3 1.00 (0.54-1.00) 0.95 (0.68-0.99) 0.57<br />

Testis 11� stage 4 1.00 (0.16-1.00) 0.81 (0.59-0.91) 0.52<br />

Ovary 11� stage 1-3 0.82 (0.62-0.92) 0.90 (0.80-0.95) 0.25<br />

Ovary 11� stage 4 0.71 (0.26-0.92) 1.00 (0.29-1.00) 0.85<br />

Extragonadal 11� stage 1-3 0.50 (0.01-0.91) 0.68 (0.44-0.83) 0.55<br />

Extragonadal 11� stage 4 Not assessable 0.45 (0.17-0.71) 0.45<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


616s Pediatric Oncology<br />

9540 Poster Discussion Session (Board #20), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Phase I trial <strong>of</strong> temsirolimus (TEM), irinotecan (IRN), and temozolomide<br />

(TMZ) in children with refractory solid tumors: A Children’s Oncology<br />

Group study. Presenting Author: Rochelle Bagatell, The Children’s Hospital<br />

<strong>of</strong> Philadelphia, Philadelphia, PA<br />

Background: Inhibitors <strong>of</strong> mTOR have demonstrated activity in preclinical<br />

pediatric solid tumor models. A phase I trial to define the dose limiting<br />

toxicities (DLTs) associated with the mTOR inhibitor TEM in combination<br />

with IRN and TMZ was conducted in patients (pts) with refractory solid<br />

tumors. Methods: Escalating doses <strong>of</strong> TEM were administered intravenously<br />

on days (d) 1 and 8 <strong>of</strong> a 21-d cycle for a maximum <strong>of</strong> 1 year (y). IRN (50<br />

mg/m2 /dose) was administered orally on d1-5. TMZ (100 mg/m2 /dose) was<br />

administered orally on d1-5. When the maximum planned dose <strong>of</strong> TEM was<br />

reached (35 mg/m2 /dose), IRN was escalated stepwise from 50 to 90<br />

mg/m2 /dose. Pts were enrolled on 6 dose levels using the rolling-six design.<br />

Results: 46 eligible pts (30 male, median age 11y, range 1 – 21) were<br />

enrolled; 37 were fully evaluable for toxicity [neuroblastoma (9), osteosarcoma<br />

(4), Ewing sarcoma (3), rhabdomyosarcoma (3), CNS (10) or other<br />

(8) tumors]. 173 cycles, median 2 (range 1 – 17) have been delivered.<br />

Dose-limiting hyperlipidemia was observed during cycle 1 in 2 pts at dose<br />

level 3 (TEM 25 mg/m2 , IRN 50 mg/m2 , TMZ 100 mg/m2 ); both pts were on<br />

chronic corticosteroids. The protocol was amended to preclude chronic<br />

systemic steroid use and modify hyperlipidemia management. Doselimiting<br />

hyperlipidemia was not observed in subsequent pts. Cycle 1 DLT<br />

(elevated GGT) was observed in 1 pt treated with TEM 35 mg/m2 , IRN 65<br />

mg/m2 , TMZ 100 mg/m2 . DLT has not been observed in 4 <strong>of</strong> the first 6 pts<br />

treated at the highest planned dose level (TEM 35 mg/m2 , IRN 90 mg/m2 ,<br />

TMZ 100 mg/m2 ). Additional �Grade 3 regimen-related toxicities occurring<br />

in �1 evaluable pt include neutropenia (12), lymphopenia (10),<br />

leukopenia (6), thrombocytopenia (4), anemia (2), nausea or vomiting (5),<br />

hypokalemia (4), hypophosphatemia (2), diarrhea (2), elevated transaminases<br />

(2), and infection (2). 1 pt had a Grade 3 allergic reaction to TEM. 1<br />

pt had a confirmed partial response and 4 have remained on protocol<br />

therapy for �1 year. Conclusions: The combination <strong>of</strong> TEM (35 mg/m2 /<br />

dose) d 1 and 8, IRN (90 mg/m2 /dose) d 1-5, and TMZ (100 mg/m2 /dose) d<br />

1-5 <strong>of</strong> a 21-d cycle appears to be well tolerated in children with refractory<br />

solid tumors.<br />

9542 Poster Discussion Session (Board #22), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

A phase I trial <strong>of</strong> AT9283 (a selective inhibitor <strong>of</strong> Aurora kinases) given for<br />

72 hours every 21 days via intravenous infusion in children and adolescents<br />

with relapsed and refractory solid tumours. Presenting Author: Darren<br />

R Hargrave, Great Ormond Street Hospital, London, United Kingdom<br />

Background: AT9283, is a multi-targeted inhibitor, against Aurora A and B,<br />

JAK & ABL kinases. Aurora kinases are potential therapeutic targets in<br />

paediatric solid cancers. Methods: A phase I dose escalation study was<br />

performed using a 72 hour intravenous infusion repeated 3 weekly using a<br />

rolling 6 design for patients aged �2to�19 years with relapsed/ refractory<br />

solid tumours. Results: Eighteen patients treated with a median age <strong>of</strong> 10<br />

(range 3 to 16) years. Four dose cohorts <strong>of</strong> 7, 9, 11.5 and 14.5 mg/m2 /day.<br />

The diagnoses included; 5 high grade glioma, 4 rhabdoid tumours, 3<br />

neuroblastomas, 3 sarcomas & 3 others. There has been only one dose<br />

limiting toxicity; Grade 3 febrile neutropenia at 11.5 mg/m2 /day. The<br />

majority <strong>of</strong> adverse events (AEs) have been grade 1/2 & considered<br />

unrelated/ unlikely related to study drug. Two patients have experienced<br />

Grade 3 or 4 AEs considered at least possibly related to study drug: Grade 3<br />

haemoglobin and Grade 4 neutrophils in a patient treated at 9 mg/m 2 /day &<br />

Grade 3 lymphopenia, neutrophils, infection with normal neutrophil count<br />

and aspartate transaminase in a patient treated at 11.5 mg/m2 /day.<br />

Pharmacokinetics <strong>of</strong> AT9283 in this population are largely in keeping with<br />

those seen in adult patients at similar doses (Arkenau et al., 2011)<br />

although there may be greater variability. Pharmacodynamic evidence <strong>of</strong><br />

aurora B inhibition, as manifested by a reduction in histone H3 phosphorylation<br />

in normal skin biopsies pre & post infusion, has been documented at<br />

all dose levels tested. Stable disease (up to 6 cycles) has been observed in<br />

3 patients. Conclusions: This paediatric phase I study has demonstrated<br />

AT9283 administered as a 72 hour continuous infusion can be given at a<br />

dose level <strong>of</strong> 11.5 mg/m2 /day which is higher than the maximum tolerated<br />

dose observed in adult patients (9 mg/m2 /day) with advanced solid<br />

tumours. Myelosuppresion is the main toxicity but the regimen is well<br />

tolerated with preliminary anticancer activity seen in heavily pre-treated<br />

paediatric patients.<br />

Dose level mg/m2 /day<br />

(N) Cmax (ng/mL) AUC (ng/mL.hr) Half life (hr) Tmax (hr)<br />

7(3) 14.4 698 5.4 56<br />

9(3) 32.0 1818 4.7 56<br />

11.5(6) 39.4 2081 5.1 56<br />

9541 Poster Discussion Session (Board #21), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

A phase I trial <strong>of</strong> IMC A12 and temsirolimus in children with refractory solid<br />

tumors: A Children’s Oncology Group study. Presenting Author: Maryam<br />

Fouladi, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH<br />

Background: IMC-A12, a fully recombinant IgG1 monoclonal antibody that<br />

targets the human IGF-IR and inhibits downstream signaling <strong>of</strong> MAPK and<br />

PI3K/AKT, and temsirolimus, an mTOR inhibitor, have demonstrated<br />

preclinical single agent, antitumor activity against many pediatric tumors.<br />

A phase I and pharmacokinetic (PK) trial evaluating the combination <strong>of</strong><br />

these agents was conducted in children with refractory solid and CNS<br />

tumors. Methods: Using a modified 3�3 design, IMC-A12 and temsirolimus<br />

were administered intravenously once every 7 days in 28 day cycles.<br />

Serial PK studies were obtained on day 1, cycle 1 and trough samples were<br />

obtained on days 15 and 28. Biology studies included assessment <strong>of</strong> IGFR<br />

and insulin receptor expression and phosphorylation in archival tumor<br />

samples and phosphorylation <strong>of</strong> IGFR and mTOR downstream targets in<br />

peripheral blood mononuclear cells (PBMC) pre and post therapy. Results:<br />

Thirty-nine patients [20 male, median age 11.8 years (range 1-21.5)] with<br />

rhabdomyosarcoma (n�10), osteosarcoma (n�6), Ewings sarcoma (n�5),<br />

astrocytoma (n�5), or other tumors (n�13) were enrolled; 33 were<br />

evaluable for toxicity. At dose level 1 [IMC-A12 (6 mg/kg); temsirolimus<br />

(15 mg/m2 )], 2 DLTs (grade 3 hypercholestrolemia, mucositis) occurred<br />

among 6 evaluable patients. Six more patients were accrued and 3<br />

additional DLTs were observed (grade 3 ALT, fatigue and prolonged<br />

thrombocytopenia � 14 days). At dose level 0 [IMC-A12 (6 mg/kg);<br />

temsirolimus (10 mg/m2 )], dose-limiting mucositis occured in 2/6 patients.<br />

At dose level -1 [IMC-A12, (4 mg/kg) and temsirolimus at (8<br />

mg/m2 )], 0 <strong>of</strong> 3 patients experienced a DLT. At an intermediate dose level<br />

[IMC-A12 (6 mg/kg) and temsirolimus (8 mg/m2 )], 1 <strong>of</strong> 6 patients<br />

experienced mucositis; among an expanded PK cohort for children �12<br />

years, none <strong>of</strong> 6 patients experienced a DLT. Target inhibition (decreased<br />

S6K1 and PAkt) in PBMCS were noted at all dose levels. At 8 mg/m2 , the<br />

median sirolimus AUC was 225 �g/ml · h (159-226), and median temsirolimus<br />

AUC was 3340 �g/ml · h (1894-4387), and median Cmax 1176<br />

�g/ml (480-2231). Conclusions: The recommended pediatric phase II<br />

doses for the combination <strong>of</strong> IMC-A12 and temsorolimus are 6 mg/kg and 8<br />

mg/m2 , respectively.<br />

9543 Poster Discussion Session (Board #23), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Pharmacokinetic (PK) and pharmacodynamics (PD) properties <strong>of</strong> SC-PEG<br />

e. coli L-asparaginase (EZN-2285) in the treatment <strong>of</strong> patients with acute<br />

lymphoblastic leukemia (ALL): Results from Children’s Oncology Group<br />

(COG) study AALL07P4. Presenting Author: Anne L. Angiolillo, Children’s<br />

National Medical Center, Washington, DC<br />

Background: Asparaginase is a critical therapeutic agent in the treatment <strong>of</strong><br />

childhood ALL, and pegaspargase (Oncospar, ONC) is now the preferred<br />

product in COG frontline ALL trials. EZN-2285 replaces the succinimidyl<br />

succinate linker in ONC with a succinimidyl carbamate linker creating a<br />

more stable molecule. AALL07P4 was designed to determine the PK<br />

comparability <strong>of</strong> EZN-2285 to ONC when given intravenously in newly<br />

diagnosed patients with NCI high-risk (HR) B-precursor ALL. Methods: 162<br />

eligible patients were randomized to receive EZN-2285 at 2100 (LD,<br />

n�66) or 2500 (HD, n�42) IU/m 2 vs. 2500 IU/m2 <strong>of</strong> ONC (n�51) in a 2:1<br />

manner based on the prednisone/Capizzi methotrexate arm <strong>of</strong> COG<br />

AALL0232. All groups were similar demographically with the exception <strong>of</strong><br />

number <strong>of</strong> patients over the age <strong>of</strong> 16 years were 6, 5, and 19 in the ONC,<br />

HD and LD, respectively. Results: PK, PD and targeted AEs (Gr 3-5) after<br />

the first doses <strong>of</strong> study drug are presented in the table below. Adverse<br />

events in induction were not significantly different between arms with the<br />

exception <strong>of</strong> a higher incidence <strong>of</strong> hyperglycemia (p�0.042). Conclusions:<br />

Based on Cmax , AUC0-25d , and asparaginase activity 25 days post drug,<br />

EZN-2285 2500 IU/m2 appears to have an improved PK/PD pr<strong>of</strong>ile<br />

compared to pegaspargase 2500 IU/m2 and a comparable toxicity pr<strong>of</strong>ile in<br />

children with HR ALL.<br />

PK/PD (mean�SD) and AEs post induction dose.<br />

Pegaspargase<br />

2500 IU/m 2 (n�43)<br />

EZN-2285<br />

2500 IU/m 2 (n�40)<br />

EZN-2285<br />

2100 IU/m 2 (n�62)<br />

Cmax (mIU/mL) 1,647�474 1,655�366 1,291�379<br />

t½ (hrs) 127�51 322�118 305�98<br />

AUC0-25d (mIU*hr/mL) 359,781�80,309 470,742�123,584 369,998�122,013<br />

AUC0-inf (mIU*hr/mL) 387,015�85,753 574,091�158,574 454,394�144,348<br />

Asparaginase activity (mIU/mL) 25 D<br />

post drug<br />

72.8�47.9 339.6�126.8 271.6�118.2<br />

Asparaginase activity >100/>400 mIU/mL 25 D<br />

post drug (%)<br />

67/0 98/28 94/14<br />

Plasma asparagine depletion 25/42 D<br />

post drug (%)<br />

88/8 92/72.4 93/75<br />

Safety population (n�54) (n�42) (n�69)<br />

Allergy (%) 7 2 1<br />

Coagulopathy (%) 0 7 3<br />

Hyperbilirubinemia (%) 7 12 9<br />

Hyperglycemia (%) 15 26 35<br />

Hyperlipidemia (%) 6 5 1<br />

Pancreatitis (%) 4 5 6<br />

Thrombosis (%) 0 0 6<br />

CNS (%) 0 0 1<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


9544 Poster Discussion Session (Board #24), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

The application <strong>of</strong> radiotherapy to the pediatric preclinical testing program:<br />

Results <strong>of</strong> a pilot study. Presenting Author: Christopher E. Pelloski, The<br />

Ohio State University Arthur G. James Comprehensive Cancer Center and<br />

Richard L. Solove Research Institute, Columbus, OH<br />

Background: The Pediatric Preclinical Testing Program (PPTP) has been<br />

successfully utilized to determine the efficacy <strong>of</strong> novel agents by testing via<br />

its mouse-flank in vivo model. We report on the feasibility and biologic<br />

outcomes <strong>of</strong> a pilot study using rhabdomyosarcoma (RMS) xenograft lines<br />

treated with radiotherapy (RT) alone and concurrently with the mTOR<br />

tyrosine kinase inhibitor, AZD8055, using the PPTP model. Methods: We<br />

developed a mouse flank irradiation device for daily delivery <strong>of</strong> RT in<br />

clinically relevant doses (2 Gy per fraction up to 40 Gy).Two RMS xenograft<br />

lines <strong>of</strong> the PPTP, Rh30 (alveolar) and Rh18 (embryonal), were implanted<br />

into SCID mice, grown to appropriate volumes and were subjected to<br />

fractionated RT. In a second study, daily co-administration <strong>of</strong> AZD8055<br />

(5-20 mg/Kg, gavage) with RT was performed. Cure rates (durable complete<br />

response �12 weeks post-treatment) and RT dose densities (given dose /<br />

initial xenograft volume, Gy/cc) were compared between groups. Results:<br />

With RT alone at mean dose-densities <strong>of</strong> 59-60 Gy/cc, cure was achieved in<br />

only 4/18 (22%) <strong>of</strong> the Rh30-bearing mice and 9/12 (75%) <strong>of</strong> the<br />

Rh18-bearing mice (p�0.006). Pr<strong>of</strong>iling data revealed higher levels <strong>of</strong><br />

Fanconi anemia pathway gene expression in Rh30 compared to the more<br />

sensitive Rh18. Since recent data showed conditional knockout <strong>of</strong> mTOR<br />

resulted in the loss <strong>of</strong> FANCD2 gene expression, we postulated that<br />

blockade <strong>of</strong> TORC1/TORC2 with AZD8055 would reduce FANCD2 and<br />

increase the RT-sensitivity <strong>of</strong> Rh30. The addition <strong>of</strong> AZD8055 to RT<br />

resulted in a selective sensitization <strong>of</strong> the Rh30 line. With a mean RT<br />

dose-density <strong>of</strong> 27 Gy/cc, the cure rate in Rh30-bearing mice improved to<br />

11/15 (73%). For the Rh18 group, the cure rate was 7/15 (46%) at a mean<br />

dose density <strong>of</strong> 44 Gy/cc. Western blot analysis showed the coadministration<br />

<strong>of</strong> AZD8055 abrogated the brisk increase in mTOR signaling<br />

and FANCD2 expression after the first several 2 Gy fractions <strong>of</strong> RT; most<br />

strikingly in Rh30. Conclusions: This study demonstrates the feasibility <strong>of</strong><br />

applying RT to the PPTP model. It recapitulated the expected clinical<br />

radiobiology and demonstrated its utility in preclinical testing and the<br />

discovery <strong>of</strong> novel mechanisms <strong>of</strong> RT resistance in pediatric tumors.<br />

9546 General Poster Session (Board #40A), Sun, 8:00 AM-12:00 PM<br />

Trends in body mass index (BMI) during and after treatment for standard<br />

risk (SR) acute lymphoblastic leukemia (ALL): A report from the Children’s<br />

Oncology Group (COG). Presenting Author: Susan Joy Lindemulder, OHSU,<br />

Portland, OR<br />

Background: Obesity is a potential complication in children treated for ALL.<br />

Limited data exist regarding timing <strong>of</strong> BMI changes and risk in long-term<br />

survivors <strong>of</strong> ALL treated without cranial radiation (CRT). This study<br />

describes temporal trends in BMI during and after therapy for SR-ALL and<br />

identifies associated factors. Methods: We conducted a retrospective cohort<br />

study <strong>of</strong> children with SR-ALL enrolled on two sequential clinical trials<br />

between 1993 and 2000 and on the COG ALTE02C2 follow-up study.<br />

Therapy included prednisone or dexamethasone during induction, the<br />

same steroid in maintenance phases, and no CRT. Standing height and<br />

weight was ascertained at diagnosis (dx), start <strong>of</strong> consolidation, start <strong>of</strong><br />

maintenance, start <strong>of</strong> the last cycle <strong>of</strong> maintenance, and at least one year<br />

<strong>of</strong>f therapy. Age and gender-specific BMI percentiles (BMI%) were calculated<br />

using 2000 CDC growth charts for patients 2-20 years. Results: The<br />

269 subjects were a median <strong>of</strong> 3.5 years at dx, 46.7% female, 82.3%<br />

white, and a median <strong>of</strong> 9.1 years since dx at the last timepoint. The BMI%<br />

was associated with elapsed time since dx: BMI% increased between dx<br />

and consolidation (50.9%-68.3%, p � 0.0001), remained stably elevated<br />

until the end <strong>of</strong> maintenance, and then decreased somewhat from the end<br />

<strong>of</strong> maintenance to the last timepoint (74.1%-70.6%, p � 0.03). After<br />

therapy, 18.1% <strong>of</strong> survivors were overweight (BMI% 85-95) and 20.9%<br />

were obese (BMI% � 95). By unadjusted linear regression, higher dx BMI%<br />

was positively associated with BMI% post-therapy (p � 0.0001). There was<br />

an interaction with steroid and dx BMI% such that the association between<br />

dx BMI% and post therapy BMI% was significantly greater for the<br />

dexamethasone group than the prednisone group (p � 0.01). There was no<br />

association with age at dx, gender, or race. Conclusions: In this retrospective<br />

study <strong>of</strong> SR-ALL survivors treated without CRT, we found that BMI%<br />

increased significantly in the month after dx and remained substantially<br />

elevated even several years after the end <strong>of</strong> therapy. Dx BMI% was highly<br />

associated with <strong>of</strong>f-therapy BMI%, particularly in patients who had<br />

received dexamethasone rather than prednisone.<br />

Pediatric Oncology<br />

617s<br />

9545 Poster Discussion Session (Board #25), Sun, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Ipilimumab: First results <strong>of</strong> a phase I trial in pediatric patients with<br />

advanced solid tumors. Presenting Author: Melinda S. Merchant, National<br />

Cancer Institute, Bethesda, MD<br />

Background: Ipilimumab is a fully-human IgG1 monoclonal anti-CTLA-4<br />

antibody which has been approved by the FDA for adult patients with<br />

metastatic melanoma . Although the approved dose is 3mg/kg every 21<br />

days, the dose <strong>of</strong> 10mg/kg IV every 21 days for 4 doses in combination with<br />

chemotherapy was well tolerated in a phase III trial enrolling adult patients<br />

with melanoma. Methods: This pediatric phase I, dose escalation study<br />

examined the safety, tolerability, pharmacokinetics, and immunogenicity<br />

<strong>of</strong> ipilimumab administered to patients �21yo with recurrent or progressive<br />

solid tumors. Ipilimumab was administered at 1, 3, 5, and 10mg/kg IV<br />

in a standard 3 � 3 design with 4 doses <strong>of</strong> induction therapy q3 weeks<br />

followed by maintenance q3 months until disease progression or unacceptable<br />

toxicity. Tumors were measured after 6 weeks, 12 weeks, and then<br />

every 3 months. Results: Twenty one patients (melanoma n�6, osteosarcoma<br />

n�6, s<strong>of</strong>t tissue sarcomas n�7, neuroblastoma, and renal cell<br />

carcinoma) received a total <strong>of</strong> 57 doses <strong>of</strong> Ipilimumab to date. One <strong>of</strong> 6<br />

patients treated at 5mg/kg developed a dose-limiting grade 3 pancreatitis.<br />

.At the highest dose level <strong>of</strong> 10mg/kg, two <strong>of</strong> three patients under 12yo<br />

developed DLT: grade 3 colitis and concurrent norovirus in one patient, and<br />

self-resolving grade 3 transaminitis in a second patient. Three <strong>of</strong> three<br />

patients between the ages <strong>of</strong> 12 and 21yo tolerated the 10mg/kg dosing<br />

schedule without DLT. Across all dose levels, grade 2 or 3 adverse events<br />

included colitis (n�1), transaminitis (n�3), pancreatitis (n�1), autoimmune<br />

thyroiditis (n�2), and hypophysitis (n�1) observed between C1D8<br />

and C4D21. A single grade 1 rash was observed. Stable disease was the<br />

best response in 5 patients with a duration <strong>of</strong> 3 to 18 months. Conclusions:<br />

Ipilimumab can be safely administered to pediatric patients. Adolescents<br />

are able to tolerate the highest dose <strong>of</strong> 10mg/kg. Younger children (�12yo)<br />

had 2 DLTs in 3 patients at 10 mg/kg . Expansion <strong>of</strong> the 10 mg/kg cohort for<br />

adolescents and <strong>of</strong> the 5 mg/kg dose for the �12yo for further safety and<br />

pharmacokinetic information is underway. The spectrum <strong>of</strong> adverse events<br />

appears similar to those described in the adult trials, although there were<br />

no grade 2 or higher incidents <strong>of</strong> rash.<br />

9547 General Poster Session (Board #40B), Sun, 8:00 AM-12:00 PM<br />

Pharmacokinetics (PK) <strong>of</strong> bendamustine in pediatric patients with relapsed/<br />

refractory acute leukemia. Presenting Author: Mona Darwish, Teva Pharmaceutical<br />

Industries Ltd., Frazer, PA<br />

Background: The PK pr<strong>of</strong>ile <strong>of</strong> bendamustine in adult patients is well<br />

characterized. The objective <strong>of</strong> this analysis was to describe the pediatric<br />

PK pr<strong>of</strong>ile <strong>of</strong> bendamustine relative to the adult PK pr<strong>of</strong>ile and correlate the<br />

systemic exposure <strong>of</strong> bendamustine to efficacy and safety parameters.<br />

Methods: Samples were obtained after a single dose from patients aged<br />

1-19 years with relapsed/refractory acute leukemia who were enrolled in an<br />

open-label, nonrandomized study <strong>of</strong> bendamustine (90–120 mg/m2 ,<br />

infused over 60 minutes). Samples were obtained prior to bendamustine<br />

infusion and preselected time points through 24 hours after start <strong>of</strong><br />

infusion on day 1. Population PK modeling was performed using plasma<br />

concentration data from these patients. PK data from adults were used for<br />

comparison. Results: Systemic exposure <strong>of</strong> pediatric patients to bendamustine<br />

was similar to that obtained previously in adult patients. Mean Cmax was 6806 ng/mL and mean AUC0-24 was 8240 ng*hr/mL in pediatric<br />

patients, compared with a mean Cmax <strong>of</strong> 5746 ng/mL and AUC0-24 <strong>of</strong> 7121<br />

ng*hr/mL in adults. Similarity in exposure despite the large range <strong>of</strong> body<br />

surface area across the pediatric and adult populations confirms appropriateness<br />

<strong>of</strong> the body surface area–based dosing scheme. In pediatric<br />

patients, age, race, sex, or disease state had no statistically significant<br />

effect on systemic exposure to bendamustine. No changes in systemic<br />

exposure to bendamustine in the presence <strong>of</strong> a CYP1A2 inhibitor/inducer<br />

were observed. Differences in PK were not observed in pediatric patients<br />

with mild renal impairment as compared with patients with normal renal<br />

function. Exposure in 2 pediatric patients with moderate hepatic dysfunction<br />

appeared to be higher. No clear exposure-response relationship was<br />

observed. Infection was the only adverse event for which the probability <strong>of</strong><br />

occurrence increased with increase in exposure to bendamustine.<br />

Conclusions: The PK pr<strong>of</strong>ile <strong>of</strong> bendamustine in pediatric patients was<br />

similar to the known PK pr<strong>of</strong>ile in adults, demonstrating that exposures<br />

reflective <strong>of</strong> the therapeutic range in adults were attained following<br />

administration <strong>of</strong> 120 mg/m2 to pediatric patients. Support: Teva Pharmaceutical<br />

Industries Ltd., Frazer, PA.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


618s Pediatric Oncology<br />

9548 General Poster Session (Board #40C), Sun, 8:00 AM-12:00 PM<br />

Safety and biological activity <strong>of</strong> the FLT3 inhibitor lestaurtinib in infant<br />

MLL-rearranged (MLL-r) ALL: Children’s Oncology Group protocol<br />

AALL0631. Presenting Author: Di Sun, Oncology and Pediatrics, Johns<br />

Hopkins University School <strong>of</strong> Medicine, Baltimore, MD<br />

Background: MLL-r infant ALL overexpresses activated FLT3. Lestaurtinib<br />

potentiates chemotherapy(chemo)-induced cytotoxicity in MLL-r ALL.<br />

AALL0631 therapy is stratified based on age and MLL status: high risk<br />

(HR) �90 days, MLL-r; intermediate risk (IR) � 90 days, MLL-r; standard<br />

risk any age, not MLL-r. IR and HR patients (pts) get intensive chemo and<br />

are eligible for lestaurtinib. A safety/activity (S/A) phase to determine a safe<br />

and biologically active dose <strong>of</strong> lestaurtinib is followed by an efficacy phase<br />

randomizing MLL-r patients to chemo �/- lestaurtinib. The S/A phase<br />

proceeds independently for IR & HR pts, as optimal lestaurtinib dosing may<br />

differ between neonates and older infants. We previously reported successful<br />

completion <strong>of</strong> the S/A phase for IR pts; here, we report results <strong>of</strong> the S/A<br />

phase for HR pts. Methods: Lestaurtinib-related dose limiting toxicities<br />

(DLTs) were defined as grade � 3 non-heme toxicities excluding those<br />

expected with chemo alone (e.g., F&N/infection); a dose <strong>of</strong> lestaurtinib was<br />

“safe” if 0 or 1 DLTs were seen in 5 evaluable pts. FLT3 inhibition was<br />

assessed using a plasma inhibitory activity (PIA) assay that measures %<br />

inhibition <strong>of</strong> phospho-FLT3 at 5 trough time points relative to pretreatment<br />

baseline. A dose <strong>of</strong> lestaurtinib was “biologically active” if PIA<br />

was � 90% for the majority (�3 <strong>of</strong> 5) <strong>of</strong> troughs in at least 3 <strong>of</strong> 5 pts.<br />

Results: 11 eligible HR pts received lestaurtinib, 6 at dose level 1 (DL1, 3.5<br />

mg/kg/day) and 5 at DL2 (4.25 mg/kg/day). There were no DLTs in 5<br />

evaluable pts at DL1. The inevaluable pt discontinued lestaurtinib due to<br />

parental refusal, not toxicity. There was 1 DLT in 5 evaluable pts at DL2.<br />

The DLT was grade 4 intestinal perforation secondary to severe typhlitis<br />

during neutropenia following reinduction, possibly related to lestaurtinib.<br />

The pt was taken <strong>of</strong>f protocol and chemo and died <strong>of</strong> progressive disease.<br />

By PIA assay, 2 <strong>of</strong> 5 pts treated at DL1, and 4 <strong>of</strong> 5 at DL2 demonstrated<br />

FLT3 inhibitory biologic activity. Conclusions: DL2 is safe and biologically<br />

active in HR pts. The efficacy phase <strong>of</strong> AALL0631 is expanding such that<br />

HR pts will join IR pts in being randomized (1:1) post-induction to chemo<br />

�/- lestaurtinib at DL2.<br />

9550 General Poster Session (Board #40E), Sun, 8:00 AM-12:00 PM<br />

Diminished asparaginase turnover due to a germ-line mutation in cathepsin<br />

B. Presenting Author: Dunja Maroeslea W.M. Te Loo, Radboud University<br />

Medical Centre, Nijmegen, Netherlands<br />

Background: ASNase is a key component <strong>of</strong> multi-agent chemotherapy<br />

protocols used in the treatment <strong>of</strong> pediatric as well as adult acute<br />

lymphoblastic leukemia (ALL). Patients that are treated according to fixed<br />

shedules show a strong inter individual variation in serum ASNase levels.<br />

While underexposure may compromise therapeutic benefits, strongly elevated<br />

serum levels may lead to serious adverse effects. In at least 10% <strong>of</strong><br />

the patients the desired ASNase levels are either not reached or exceeded.<br />

Therefore our understanding <strong>of</strong> ASNase dynamics in vivo needs to be<br />

improved. Here we describe the identification <strong>of</strong> a novel mutation in the<br />

gene encoding Cathepsin B to diminished ASNase turnover in a pediatric<br />

patient with ALL. Methods: Sequencing, biochemical experiments and<br />

immun<strong>of</strong>luorescence. Results: A 3-year-old girl diagnosed with ALL experienced<br />

serious toxicity in the form <strong>of</strong> hyperammonemic encephalopathy<br />

during treatment with ASNase derived from Erwinia chrysantemi. Pharmacokinetic<br />

data showed a strongly delayed clearance <strong>of</strong> the ASNase, which<br />

with the standard treatment protocol resulted in extremely high serum<br />

ASNase levels. Plasmapheresis was performed and dosage frequencies<br />

were adjusted to improve the clinical status <strong>of</strong> the patient. No underlying<br />

metabolic disorder could be diagnosed. Based on a previous report<br />

describing the role <strong>of</strong> proteases in degradation <strong>of</strong> ASNase in vitro, we<br />

hypothesized that the cysteine protease Cathepsin B might be implicated in<br />

the strongly reduced clearance <strong>of</strong> ASNase in this patient. Sequencing <strong>of</strong> the<br />

open reading frame <strong>of</strong> the Cathepsin B gene (CTSB) revealed a single codon<br />

deletion in the germline <strong>of</strong> the patient, affecting a lysine residue in the<br />

carboxy terminus <strong>of</strong> the protein. Immun<strong>of</strong>luorescence and biochemical<br />

experiments show that this single amino acid deletion leads to a protein<br />

product that is retained in the endoplasmic reticulum and is inefficiently<br />

processed. Conclusions: ASNase degradation assays show that the mutant<br />

Cathepsin B has lost its ability to efficiently degrade ASNase which could<br />

explain the high levels <strong>of</strong> ASNase as observed in our patient.Together,<br />

these findings suggest that variations in Cathepsin B activity may influence<br />

serum ASNase levels in the patients.<br />

9549 General Poster Session (Board #40D), Sun, 8:00 AM-12:00 PM<br />

Serial assessment <strong>of</strong> regulatory T cells (Tregs) in pediatric AML: A<br />

prospective study. Presenting Author: Anuj Kumar Bansal, All India<br />

Institute <strong>of</strong> Medical Sciences, New Delhi, India<br />

Background: Data <strong>of</strong> Tregs in pediatric AML is lacking. The study objectives<br />

were determining Tregs in pediatric AML at diagnosis and follow up; and<br />

correlating with outcome. Methods: From Nov 2010-May 2011, 30 consecutive<br />

AML patients � 18 years were prospectively enrolled with 6 healthy<br />

controls. All patients received daunorubicin / cytarabine induction and 3<br />

courses <strong>of</strong> cytarabine. Tregs (CD4�CD25�FoxP3�) were assessed at<br />

diagnosis, post-induction, post-consolidation, 3 and 6 months follow up<br />

and relapse. Results: 30 cases with median age 9.5 years; male/female ratio<br />

14:16 had significantly higher baseline Tregs than healthy controls<br />

(12.36�4.65% vs 3.16�1.49%; p�0.0001). Patients with high Tregs<br />

frequency were females (p�0.044), WBC�50,000/mm3 (p�0.023), hypoalbuminemia<br />

(p�0.002), absence <strong>of</strong> lymphadenopathy (p�0.04) and<br />

linear correlation with peripheral blood blast% (r�0.55, p�0.0014). CR<br />

rate was 83.3% (25/30); EFS 38% and OS 73% at 14 months follow up.<br />

When Tregs were categorized as high and low based on median value, CR<br />

rate (86.6% vs 80%, p�NS), EFS [17% vs 60%, HR 1.46 (0.51-4.14)<br />

p�0.46] and OS (66% vs 80%, HR 0.58 (0.13-2.44) p�0.45) were not<br />

different. Amongst those who achieved CR, there were 7/13 relapses in<br />

those with high Tregs versus 3/12 in those with low Tregs (p�0.226).Tregs<br />

reduced post-induction (12.1�4.6 vs 6.32�2.8, p�0.000) from baseline.<br />

Using generalized estimating equation regression model, average<br />

Tregs reduction from post-induction till 3 month follow up was 0.785<br />

units/visit (p�0.005) in those with continuous CR(n�15) and 1.25<br />

units/visit (p�0.001) in those whom relapsed (n�10). Difference between<br />

these two groups was not significant. Tregs significantly increased at<br />

relapse as compared to post consolidation value (15.3�5.2 vs 4.5�1.9,<br />

p�0.003). Conclusions: This first study in pediatric AML demonstrates that<br />

Tregs is increased at diagnosis and is significantly associated with females,<br />

high WBC count, hypooalbuminemia, absence <strong>of</strong> lymphadenopathy and<br />

high peripheral blood blast%. Tregs significantly reduced post-induction on<br />

follow up; however, it increased at relapse. Baseline Tregs did not predict<br />

CR, EFS, or OS. However, there were more relapses in those with high<br />

baseline Tregs.<br />

9551 General Poster Session (Board #40F), Sun, 8:00 AM-12:00 PM<br />

Serial assessment <strong>of</strong> humoral immunity in pediatric AML: A prospective<br />

study. Presenting Author: Sameer Bakhshi, Dr. B.R. Ambedkar Institute<br />

Rotary Cancer Hospital, All India Institute <strong>of</strong> Medical Sciences, New Delhi,<br />

India<br />

Background: Humoral immunity has been evaluated in pediatric ALL; data<br />

in pediatric AML is lacking. The objectives <strong>of</strong> this study were to assess<br />

humoral immunity on follow-up in pediatric AML patients. Methods: From<br />

April 2010-May 2011, 45 consecutive AML patients 1-18 years old were<br />

prospectively enrolled with 7 healthy controls. Immunoglobulin (Ig) levels<br />

in 45 patients and B-lymphocytes (CD19�) in 29 patients were assessed at<br />

diagnosis, post-induction, post-consolidation, 3 and 6 months follow-up<br />

and relapse. Results: At diagnosis, Ig levels were higher in patients as<br />

compared to healthy controls (IgG, p�0.041; IgA, p�0.07; IgM, p�0.16)<br />

while B-cells were lower (p�0.001). Patients with gum hypertrophy had<br />

low Ig levels (IgG, p�0.007; IgA, p�0.003; IgM, p�0.06). CR rate was<br />

84.4% (38/45); EFS 40% and OS 58% at 20 months follow-up. Postinduction,<br />

there was reduction in IgM (p�0.001) and IgA (p�0.048) levels<br />

and increase in B cells (p�0.001). Using generalized estimating equation<br />

regression model, average reduction from post-induction till 3 month<br />

follow-up was 36.9 units/visit (IgG); 10.3 units/visit (IgA); 0.32 units/visit<br />

(IgM) in those with continuous CR (group-1, n�24) and 91 units/visit<br />

(IgG); 8.9 units/visit (IgA); 1.5 units/visit (IgM) in those who relapsed<br />

(group-2, n�14). Difference between these two groups was significant for<br />

IgA (p�0.017). There was increase in IgG (p�0.52) and IgA levels (0.21)<br />

at relapse when compared with post-consolidation values. B-cells had an<br />

average increase <strong>of</strong> 3.6 units/visit (p�0.012) in group 1 and decrease <strong>of</strong><br />

0.58 units/visit (p�0.82) in group 2; (group-1 vs group-2, p�0.10). There<br />

was no significant correlation <strong>of</strong> baseline Ig and B cells with CR rate, EFS<br />

and OS. Conclusions: This is the first study to evaluate humoral immunity<br />

serially in pediatric AML. AML patients with gum hypertrophy have low IgG<br />

and IgA at diagnosis. On serial monitoring, B cells show an increase in<br />

patients who are in continuous CR while those who relapse tend to show a<br />

reduction on follow-up. Further, on follow-up, although all Ig decrease, but<br />

the reduction in IgA is significantly lower in patients who relapse suggesting<br />

their possible role in disease biology.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


9552 General Poster Session (Board #40G), Sun, 8:00 AM-12:00 PM<br />

Contribution <strong>of</strong> diet and physical activity to metabolic parameters among<br />

survivors <strong>of</strong> childhood leukemia. Presenting Author: Emily S. Tonorezos,<br />

Memorial Sloan-Kettering Cancer Center, New York, NY<br />

Background: Survivors <strong>of</strong> childhood acute lymphoblastic leukemia (ALL) are<br />

at increased risk for obesity, insulin resistance and increased visceral<br />

adiposity. A history <strong>of</strong> cranial radiation therapy (CRT) worsens this risk. In<br />

non-cancer populations, adherence to a Mediterranean Diet has been<br />

shown to improve metabolic parameters and risk <strong>of</strong> diabetes mellitus.<br />

Whether diet may contribute to insulin resistance and increased adiposity<br />

in ALL survivors is not known. Methods: We surveyed 117 adult survivors <strong>of</strong><br />

childhood ALL using the Harvard Food Frequency Questionnaire. Physical<br />

activity energy expenditure (PAEE) was measured with the SenseWear Pro2<br />

Armband. Fasting blood was obtained on all subjects and insulin resistance<br />

was estimated using the Homeostasis Model for Insulin Resistance (HOMA-<br />

IR). Visceral adiposity was measured by abdominal CT. Adherence to a<br />

Mediterranean Diet pattern was calculated using the index developed by<br />

Trichopoulou. Subjects were compared using univariate analysis. Results:<br />

Subjects were majority female (56%); 25% were minority or Hispanic<br />

white. A history <strong>of</strong> CRT was present in 15 (29%) men and 25 (38%)<br />

women. Among all subjects, greater adherence to a Mediterranean diet<br />

pattern was associated with improved HOMA-IR (p�0.14), visceral adiposity<br />

(p�0.03), subcutaneous adiposity (p�0.005), waist circumference<br />

(p�0.02), and body mass index (p�0.03) (all unadjusted analyses). The<br />

effect <strong>of</strong> adherence to a Mediterranean diet on insulin resistance was<br />

especially notable in men who did not receive CRT (p�0.06). Higher dairy<br />

intake was found to worsen HOMA-IR (p�0.014), but other individual<br />

components <strong>of</strong> the Mediterranean diet, such as low intake <strong>of</strong> meat and high<br />

intake <strong>of</strong> fruits and vegetables, were not significant. Inclusion <strong>of</strong> PAEE did<br />

not alter our findings, although higher PAEE was associated with lower body<br />

mass index. Conclusions: Adherence to a Mediterranean diet pattern,<br />

especially low consumption <strong>of</strong> dairy products, may improve insulin resistance<br />

in survivors <strong>of</strong> childhood ALL. Further study is warranted.<br />

9554 General Poster Session (Board #41A), Sun, 8:00 AM-12:00 PM<br />

Minimal residual disease by flow cytometry at the end <strong>of</strong> chemotherapy for<br />

remission induction in pediatric patients with acute lymphoblastic leukemia:<br />

Experience from a center in a low-income country. Presenting Author:<br />

Eloy Perez, Hospital Infantil de Morelia Eva Samano de Lopez Mateos,<br />

Morelia, Mexico<br />

Background: The presence <strong>of</strong> minimal residual disease (MDR) following<br />

therapy for acute lymphoblastic leukemia (ALL) has been shown to be an<br />

important prognostic marker in many studies. MRD is typically detected<br />

either by polymerase chain reaction amplification or by flow cytometry.<br />

Flow-based MRD assessment has the potential for rapidly identifying<br />

patients at increased risk <strong>of</strong> relapsed, allowing for prompt changes in<br />

therapy, including earlier intensification. There are not many information<br />

about the response by MRD in countries with limited resources. Methods:<br />

The patients included were 90 ALL patients treated at the Hospital Infantil<br />

de Morelia from June 1, 2009 to January 5, 2012. MRD positivity (�) was<br />

defined as �0.01% <strong>of</strong> the gated population. Results: MRD was obtained in<br />

90 patients, 38 males and 36 females. The median age was 7 years (10<br />

months to 15 years). The levels <strong>of</strong> MRD were: �0.01, 74 (82.2%),<br />

0.01-1%, 9 (10%), �1%, 7 (7.7%). There was not a statistically<br />

significant association between the most important ALL prognostic factors<br />

(Gender, Age at diagnosis, White blood cell count at diagnosis, Central<br />

Nervous System disease, Prednisone response, DNA Index, Immunophenotype).<br />

Conclusions: The good response found is similar to that reported by<br />

international groups, a situation which suggests that the response to<br />

chemotherapy is appropriate. However, cure rates are still not equal making<br />

it necessary to review institutional treatment protocols and social characteristics<br />

<strong>of</strong> the population to achieve cure rates reported by international<br />

groups.<br />

Pediatric Oncology<br />

619s<br />

9553 General Poster Session (Board #40H), Sun, 8:00 AM-12:00 PM<br />

Fever at diagnosis <strong>of</strong> pediatric acute lymphoblastic leukemia (ALL): Are<br />

antibiotics really necessary? Presenting Author: Monica Khurana, Children’s<br />

Hospital Oakland Research Institute, Oakland, CA<br />

Background: Great variability exists in the management <strong>of</strong> suspected<br />

bacteremia in febrile, neutropenic pediatric ALL patients during chemotherapy.<br />

The National Comprehensive Cancer Network and Infectious<br />

Diseases <strong>Society</strong> <strong>of</strong> America encourage immediate, empiric antibiotics in<br />

patients with chemotherapy-induced fever and neutropenia to reduce<br />

infection-related mortality. No standard recommendation exists for patients<br />

with isolated fever at initial presentation <strong>of</strong> their ALL. This study<br />

evaluates bacteremic episodes in this subpopulation <strong>of</strong> pediatric patients.<br />

Methods: We retrospectively analyzed 245 consecutive patients with ALL at<br />

Children’s Hospital Oakland from 2000 through 2011. Using electronic<br />

medical records, we investigated each patient’s history and outcome. We<br />

surveyed bacteremic episodes up to 60 days after presentation per National<br />

Healthcare Safety Network’s guidelines. Inclusion criteria were patients<br />

with fever at presentation, which prompts a blood culture, and were started<br />

on antibiotics. We stratified bacteremic episodes into community-acquired<br />

(up to three days from admission) and nosocomial (four to 60 days).<br />

Results: Seventy-seven patients met the inclusion criteria, five <strong>of</strong> whom had<br />

positive cultures – four were contaminants (three coagulase-negative<br />

Staphylococcus and one non-anthracis Bacillus) and one was a true<br />

nosocomial bactermic episode (E coli). There were no infection-related<br />

deaths in the first 60 days <strong>of</strong> diagnosis in this cohort. Conclusions: Given<br />

our institution’s rarity <strong>of</strong> bacteremic episodes, we contemplate a more<br />

judicious use <strong>of</strong> antibiotics, including quicker narrowing <strong>of</strong> broad-spectrum<br />

antibiotics coverage and discontinuing all antibiotics sooner. These modifications<br />

may decrease bacterial resistance to antibiotics, reduce costs, and<br />

shorten patients’ hospitalization. We encourage other institutions to<br />

conduct a similar investigation.<br />

9555 General Poster Session (Board #41B), Sun, 8:00 AM-12:00 PM<br />

YM155 sensitivity in pediatric acute lymphoblastic leukemia. Presenting<br />

Author: Bill H. Chang, Doernbecher Children’s Hospital, Portland, OR<br />

Background: Despite advances in treatment and outcomes in pediatric<br />

acute lymphoblastic leukemia, there continue to be subsets <strong>of</strong> patients that<br />

are refractory to standard intensive chemotherapy. Therefore, novel agents<br />

are needed to further advance treatment for this disease. YM155 (Astellas)<br />

is a selective suppressant <strong>of</strong> survivin expression. Survivin has been shown<br />

to be over-expressed in malignant cells and in relapsed ALL. Early adult<br />

phase I/II studies show promise in both tolerability and possible efficacy in<br />

B-cell malignancies. Methods: Fresh diagnostic leukemic patient samples,<br />

ALL cell lines, and xenograft samples were obtained and subjected to<br />

YM155 at doses ranging from 1nM to 10�M. Samples were then tested for<br />

viability using standard methane-thiosulfate, apoptosis using Guava nexin<br />

Annexin V binding, protein expression using immunoblot, and P53 activation<br />

with phospho-flow cytometry. Results: The median IC50 for viability and<br />

apoptosis for the samples are: NCI standard risk ALL 5.3nM �3.47, NCI<br />

high risk ALL 91.76 nM �93.42, T-ALL 336.6nM �147.7, AML 333.6nM<br />

�489, Ph�ALL xenografts 3.8nM �1.04, and ALL cell lines 18.5nM<br />

�135. Ph�ALL samples, including primary patient, xenograft, and dasatinib<br />

(BMS) resistant samples remain significantly sensitive to YM155. For<br />

dasatinib sensitive samples, combination therapy suggests an additive<br />

effect by isobologram analysis. Immunoblot and RT2-PCR Arrays (Qiagen)<br />

identify that multiple protein expression are decreased with YM155<br />

treatment. Conclusions: Our data supports the model <strong>of</strong> the use <strong>of</strong> YM155<br />

as a treatment for ALL including an added benefit in combination with<br />

dasatinib in Ph�ALL. Although initial studies suggested that YM155 is a<br />

selective suppressant <strong>of</strong> survivin, subsequent studies are beginning to<br />

identify <strong>of</strong>f target effects. These <strong>of</strong>f target effects may enhance the drugs<br />

potential for activating cell death. Finally, we show that screening primary<br />

samples with YM155 has the potential to select a target population that is<br />

more sensitive to YM155 treatment. Future studies will be designed to<br />

develop YM155 as an additional therapeutic agent for pediatric acute<br />

lymphoblastic leukemia.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


620s Pediatric Oncology<br />

9557 General Poster Session (Board #41D), Sun, 8:00 AM-12:00 PM<br />

Diagnostic and treatment challenges <strong>of</strong> adolescent and young adult<br />

oncology patients. Presenting Author: Yanqing Xu, McGill Adolescent and<br />

Young Adult Oncology Program, Montreal, QC, Canada<br />

Background: Adolescent and young adult (AYA) cancer patients are faced<br />

with obstacles and challenges related to their diagnosis and treatment<br />

compared to children and older adults. The aim <strong>of</strong> this study was to explore<br />

the patient and health care system-related delays in the interval from<br />

cancer symptom onset to diagnosis and treatment as well as to identify the<br />

possible contributing factors to these delays in the AYA group. Methods:<br />

This study was based on a questionnaire conducted in 2010-2011<br />

completed by patients diagnosed with a malignancy between the ages <strong>of</strong> 16<br />

and 39 in addition to older patients diagnosed with a pediatric type<br />

malignancy. Four categories <strong>of</strong> delays: patient delay (time from patient<br />

symptom onset until first health care contact date), health care system<br />

delay (time from first health care contact until diagnosis date), treatment<br />

delay (time from diagnosis date until first treatment) and oncologist delay<br />

(time from first health care contact until first medical oncologist meeting)<br />

were calculated. Median delay (in days) with interquartile interval (IQI) was<br />

the main outcome measure. Median time for each category <strong>of</strong> delay was<br />

further analysed to explore how they vary with different patient characteristics.<br />

Results: We identified a median patient delay <strong>of</strong> 30 days (IQI 1-131), a<br />

median health care system delay <strong>of</strong> 53 days (IQI 1-213), a median<br />

treatment delay <strong>of</strong> 36 days (IQI 5-92) and a median oncologist delay <strong>of</strong> 77<br />

days (IQI 30-281). Patient delay was affected by patient gender, age at<br />

diagnosis and type <strong>of</strong> first health care contact. Health care system delay<br />

was associated with patient marital status, financial situation and attitude<br />

<strong>of</strong> first health care pr<strong>of</strong>essional. Treatment delay was related to type <strong>of</strong><br />

cancer. Conclusions: The health care system delay (including oncologist<br />

delay) accounts for much <strong>of</strong> the delay from symptom onset to first<br />

treatment. Pr<strong>of</strong>essional characteristics <strong>of</strong> frontline medical personnel as<br />

well as socioeconomic and biological characteristics <strong>of</strong> the patients may<br />

contribute to delay. Healthcare pr<strong>of</strong>essionals and the general community as<br />

a whole need to be aware <strong>of</strong> the factors contributing to delay in diagnosis<br />

and treatment in the underserved patient population.<br />

9559 General Poster Session (Board #41F), Sun, 8:00 AM-12:00 PM<br />

Results <strong>of</strong> autologous transplants for children from a tertiary cancer center<br />

in India. Presenting Author: Amol Dongre, Tata Memorial Centre, Mumbai,<br />

India<br />

Background: Autologous transplantation is a curative option for children<br />

with various malignancies. We report the results <strong>of</strong> our center with the aim<br />

<strong>of</strong> evaluating toxicities and prognostic factors that may influence decision<br />

making to use our limited resources judiciously. Methods: Forty-six patients<br />

diagnosed below 18 years from 1st April 1994- 28th February 2011 were<br />

included in this retrospective study. Twenty-four patients had lymphoma,<br />

12- neuroblastoma and 10-others ( 5- acute leukemia, 4- Ewing sarcoma<br />

and 1 - rhabdomyosarcoma). Twenty-eight patients received peripheral<br />

blood stem cells, 12 – marrow and 6-both. Prognostic factors evaluated for<br />

overall survival (OS) and progression-free survival (PFS) were remission<br />

status at transplant, baseline and pre-transplant serum albumin, time<br />

interval between diagnosis and transplant, type <strong>of</strong> malignancy and number<br />

<strong>of</strong> lines <strong>of</strong> chemotherapy pre transplant. Results: Median age at diagnosis<br />

and transplant were 9 and 12 years respectively. Median baseline and pre<br />

transplant serum albumin were 3.6 and 3.9 g/dl. Median number <strong>of</strong> lines <strong>of</strong><br />

chemotherapy was 2. Median time from diagnosis to transplant was 1.1<br />

years. Incidence <strong>of</strong> grade 3 and 4 oral mucositis and diarrhea were 46%<br />

and 25% respectively. At the time <strong>of</strong> transplant, 45% were in complete<br />

remission (CR), 41% in partial remission (PR) and 13% in refractory state.<br />

Total Parenteral Nutrition (TPN) was used in 63% patients with median<br />

duration <strong>of</strong> 11 days. No patient developed sinusoidal obstruction syndrome-<br />

.Median days to neutrophil and platelet engraftment were 11 and 15 days<br />

respectively. Neutropenic sepsis leading to death was seen in 17%. Median<br />

follow up was 1.5 years. At 2 years, the probability <strong>of</strong> OS and PFS were<br />

39% and 35% for all patients while OS and PFS for lymphoma, neuroblastoma<br />

and others were 45% and 32%, 17% and 0% and 50% and 40%<br />

respectively. CR at transplant was the most important determinant <strong>of</strong> better<br />

OS (59 % vs 28%; p � 0.000004) and PFS (53% vs 8%; p �<br />

0.0000018). Conclusions: CR at transplant is the most important prognostic<br />

factor. Patients with neuroblastoma have dismal outcome which<br />

requires reevaluation <strong>of</strong> transplant strategies for this group.<br />

9558 General Poster Session (Board #41E), Sun, 8:00 AM-12:00 PM<br />

Treatment outcomes <strong>of</strong> pediatric oncology patients in Zambia. Presenting<br />

Author: Jeremy Slone, Vanderbilt University School <strong>of</strong> Medicine, Nashville,<br />

TN<br />

Background: Due to challenges in the delivery <strong>of</strong> pediatric oncology care in<br />

low-middle income countries (LMIC), diagnosis and treatment remains<br />

inadequate for the majority <strong>of</strong> patients. The University <strong>of</strong> Zambia School <strong>of</strong><br />

Medicine/University Teaching Hospital (UTH) and Vanderbilt University<br />

School <strong>of</strong> Medicine/Vanderbilt Institute for Global Health established a<br />

partnership to investigate treatment outcomes at UTH, the only institution<br />

providing pediatric oncology care in Zambia, and assess risk factors<br />

associated with treatment abandonment. Methods: A retrospective study<br />

was conducted in a cohort <strong>of</strong> patients, presenting from July 2008 – June<br />

2010, using an established database and medical record review. Results:<br />

Of the 230 children enrolled in the database, 162 met the inclusion<br />

criteria. The average age at diagnosis was 6.0 years; males comprised<br />

55.6% <strong>of</strong> the cohort; 51.6% had a histopathological diagnosis and 10.5%<br />

<strong>of</strong> the cohort was HIV positive. The most common diagnoses were<br />

lymphoma (25.9%), Wilms tumor (22.8%), and retinoblastoma (17.9%).<br />

Leukemia and Kaposi sarcoma accounted for 7.4% each. Death (46.3%)<br />

and abandonment <strong>of</strong> treatment (45.7%) were the most common outcomes<br />

with only 8.0% having completed treatment or currently undergoing<br />

treatment, including palliative regimens, at the time <strong>of</strong> data acquisition.<br />

Residence in Lusaka or Central provinces, closest in proximity to UTH, was<br />

associated with a decreased risk for abandonment <strong>of</strong> treatment (Odds Ratio<br />

(OR) 0.41 (95% CI 0.21-0.81, p � 0.009) while maternal education less<br />

than secondary school (OR 2.73 95% CI 1.2-6.6, p � 0.03) was<br />

associated with an increased risk for abandonment <strong>of</strong> treatment. Conclusions:<br />

At the only pediatric cancer center in Zambia, treatment outcomes are dire<br />

with the majority <strong>of</strong> the cohort abandoning treatment or dying during<br />

therapy. Challenges include access to cancer chemotherapy, logistical<br />

facilitation, fiscal support <strong>of</strong> radiotherapy, and community engagement.<br />

Further investigation is required to inform effective intervention strategies<br />

to improve outcomes.<br />

9560 General Poster Session (Board #41G), Sun, 8:00 AM-12:00 PM<br />

Evaluation <strong>of</strong> renal function in children with solid tumors: Comparison<br />

between estimation with Schwartz formula and EDTA clearance. Presenting<br />

Author: Carole Serrano, Institut Gustave Roussy-<strong>Clinical</strong> Pharmacy<br />

department, Villejuif, France<br />

Background: Using the [51Cr]-ethylenediamine tetra acetic acid ([51Cr]-<br />

EDTA) method provides an accurate measure <strong>of</strong> glomerular filtration rate<br />

(GFR) for each patient. However, this method is not always feasible in<br />

routine. Thus, several mathematical equations have been developed to<br />

predict creatinine clearance. The most widely used in pediatric patients is<br />

the Schwartz formula, based on patient height and serum creatinine.<br />

However, this method has not been validated in pediatric cancer patients.<br />

The aim <strong>of</strong> the present study was to compare GFR measured with the<br />

51Cr-EDTA method to GFR estimated with the Schwartz formula in<br />

patients younger than 18 years <strong>of</strong> age and treated for solid tumors. Methods:<br />

Data have been retrospectively collected on consecutive children treated<br />

between 2001 and 2011 at Institut Gustave Roussy. All <strong>of</strong> the patients had<br />

undergone assessment <strong>of</strong> GFR via 51Cr-EDTA clearance and estimation <strong>of</strong><br />

renal function by the Schwartz formula. Exclusion criteria were serum<br />

creatinine under 25�mol/L or no recent dosage available. GFR analysis was<br />

realized using the slope intercept method after a single injection <strong>of</strong><br />

51Cr-EDTA solution.. Values <strong>of</strong> the GFR were calculated by multiplying the<br />

volume <strong>of</strong> dilution by the slope <strong>of</strong> the single exponential fit to the three data<br />

points and then expressed in ml/min/1.73m². Student paired t-test (alpha<br />

� 0.05) was used to compare results obtained for each patient with the two<br />

methods. Results: 196 patients (120 males, 76 females), treated with<br />

various types <strong>of</strong> cancer (neuroblastoma, osteosarcoma, Ewing sarcoma,<br />

lymphoma. . .) Mean age was 6.7 years. Mean GFR was 127.5 ml/min/<br />

1.73m² with the 51Cr-EDTA method versus 148.1 ml/min/1.73m² with the<br />

Schwartz formula. Mean <strong>of</strong> difference was 20.7 ml/min/1.73m² with a<br />

confidence interval [-25.7 ; -15.6] (p � 0.0001). Conclusions: GFR<br />

estimated with the Schwartz formula is statistically different from the<br />

isotopic method. In most cases, GFR estimated with the Schwartz formula<br />

is overestimated. This study highlights the lack <strong>of</strong> accuracy <strong>of</strong> this equation<br />

in pediatric population with cancer. The 51Cr-EDTA method must be<br />

preferred when a reliable and accurate evaluation <strong>of</strong> renal function is<br />

needed.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


9561 General Poster Session (Board #41H), Sun, 8:00 AM-12:00 PM<br />

Familial risk in pediatric cancer and implications for practice. Presenting<br />

Author: Karen Curtin, University <strong>of</strong> Utah and Huntsman Cancer Institute,<br />

Salt Lake City, UT<br />

Background: Using a unique genealogical resource, the Utah Population<br />

Database, we examined risk <strong>of</strong> cancer in 1st- through 3rd-degree relatives<br />

<strong>of</strong> 4,482 pediatric cancer cases �18 years old diagnosed between 1966<br />

and 2009. We quantified cancer risk in relatives <strong>of</strong> children with cancer in<br />

order to determine evidence <strong>of</strong> a familial aggregation <strong>of</strong> cancer, identify<br />

high-risk pedigrees, and inform counseling protocols for genetic testing and<br />

screening. Methods: We estimated cancer risk in relatives <strong>of</strong> pediatric cases<br />

compared to random population controls matched 5:1 on sex, birth year,<br />

and birthplace. Odds ratios were calculated using conditional logistic<br />

regression, adjusting for number <strong>of</strong> biological relatives, their degree <strong>of</strong><br />

relatedness, and their person-years at risk. We included all relatives <strong>of</strong><br />

cases and controls with followup who linked to a pedigree <strong>of</strong> �2 generations;<br />

this approach has been shown to lead to unbiased risk estimates. As<br />

observations within families are non-independent, a robust variance<br />

estimator for cluster-correlated data was incorporated. Results: Mostly<br />

siblings, 1st-degree relatives (N�49 affected) <strong>of</strong> pediatric cases were at<br />

2-fold increased risk <strong>of</strong> being diagnosed at age�19 with cancer themselves<br />

(p�10-4 ); siblings <strong>of</strong> cases diagnosed at age�5 had a 4-fold risk <strong>of</strong><br />

pediatric cancer (p�10-7 ). Likewise, 2nd-degree relatives (N�61 affected)<br />

and cousins (N�105 affected) <strong>of</strong> cases had a ~2-fold risk <strong>of</strong> childhood<br />

cancer (p�10-4 ). Proband siblings were at increased risk for leukemia,<br />

brain tumor, epithelial neoplasia, and bone cancer. While 1st-degree<br />

relatives <strong>of</strong> pediatric cases had a slight increased risk <strong>of</strong> adult-onset cancer,<br />

when they do develop cancer they exhibit a 50% increased risk <strong>of</strong> a<br />

Li-Fraumeni Syndrome-related tumor diagnosis (N�142 affected relatives;<br />

p�10-4 ). Conclusions: Our findings provide evidence <strong>of</strong> familial aggregation<br />

<strong>of</strong> cancer and suggest a higher percent <strong>of</strong> pediatric cancers are related to<br />

hereditary syndromes than are adult cancers. We encourage a 3-generation<br />

family history be collected and routinely updated for all pediatric cancer<br />

patients, and those children with family histories <strong>of</strong> early-onset cancer be<br />

referred for genetic counseling and early surveillance when appropriate.<br />

9563 General Poster Session (Board #42B), Sun, 8:00 AM-12:00 PM<br />

Preclinical study <strong>of</strong> a Bcl-2 inhibitor in neuroblastoma. Presenting Author:<br />

Assila Belounis, Research Center CHU Sainte Justine, Montréal, QC,<br />

Canada<br />

Background: Neurblastoma (NB) is the most common and deadly extracranial<br />

solid tumor <strong>of</strong> childhood. This malignant tumor exhibits a broad<br />

spectrum <strong>of</strong> clinical features, including spontaneous regression or maturation<br />

without any treatment or progression to metastisis leading to death.<br />

However, in spite <strong>of</strong> many therapeutic improvements, only 60% survive<br />

long term. In fact, 40% <strong>of</strong> patients with high-risk NB still relapse and<br />

eventually die <strong>of</strong> the disease despite aggressive combinations <strong>of</strong> multiagent<br />

chemotherapy. In those cases, new therapeutic strategies must be<br />

developed. Studies have shown that BCl-2, a central anti-apoptotic protein,<br />

is over-expressed in NB. Although Bcl-2 is not a significant prognostic<br />

factor in NB, its increased expression would contribute to chemotherapy<br />

resistance. BCl-2 is also shown to be involved in the modulation <strong>of</strong><br />

autophagy by inhibiting Beclin-1. Therefore, BCl-2 protein represents an<br />

attractive target for new therapeutic strategies in NB.The objective <strong>of</strong> this<br />

study is to determine the efficacy in vitro <strong>of</strong> Obatoclax (OB), a BCl-2<br />

inhibitor, used in monotherapy or in combination to cisplatine (CP), a<br />

classical chemotherapeutic agent used in NB treatment, on NB cell<br />

survival. Methods: Six NB cell lines (SK-N-DZ, SK-N-FI, SK-N-SH, N91,<br />

NB8 and NB10) were treated with OB alone or in association to CP. Cell<br />

survival was measured by MTT test. Autophagy was measured by MDC test.<br />

Western Blots (WB) were done to evaluate the modulation <strong>of</strong> apoptotic and<br />

autophagic pathways in treated cells. Results: OB used in monotherapy<br />

shows promising results on NB cell lines with an average IC50 <strong>of</strong> 0.12�M.<br />

Also, OB demonstrates synergistic effects when associated to CP. The IC50<br />

<strong>of</strong> CP treated cells varied from 3.183�M to 6.837�M but dropped from<br />

0.003�M to 0.008�M when combined with 0.5�M <strong>of</strong> OB. Moreover, our<br />

WB show an increase in cleaved Caspase-3 and PARP-1 expression,<br />

suggesting an upregulation <strong>of</strong> apoptosis in treated cells. Autophagy is also<br />

upregulated confirmed by an increase in autophagic vacuoles and cleaved<br />

LC3 II protein. Conclusions: OB used in monotherapy or in combination at<br />

potentially therapeutic doses shows promising results in NB. These results<br />

will allow for the improvement <strong>of</strong> NB treatments by introducing a new<br />

therapeutic strategy.<br />

Pediatric Oncology<br />

621s<br />

9562 General Poster Session (Board #42A), Sun, 8:00 AM-12:00 PM<br />

Mitigating smoothened inhibitor-induced growth plate closure with parathyroid<br />

hormone. Presenting Author: Yoav E. Timsit, Novartis Institutes for<br />

BioMedical Research Inc., Cambridge, MA<br />

Background: The Hedgehog pathway plays an important role in regulating<br />

growth plate patency by activating PTH/PTHrP signaling, and loss <strong>of</strong><br />

PTH/PTHrP signaling is an expected pharmacological effect <strong>of</strong> smoothened<br />

(Smo) inhibition. LDE225, a Smo inhibitor currently in phase I/II trials for<br />

the treatment <strong>of</strong> basal cell carcinoma (BCC) and medulloblastoma (MB),<br />

causes closure <strong>of</strong> the epiphyseal (growth) plate in rodents and dogs. As a<br />

therapeutic strategy to mitigate these effects, we investigated whether<br />

administration <strong>of</strong> human parathyroid hormone (PTH amino acids 1-34 or<br />

PTH1-34) could prevent premature growth plate closure caused by LDE225.<br />

Methods: Oral LDE225 was given once daily to rapidly growing male rats<br />

(starting at 4 or 6 weeks <strong>of</strong> age) in combination with hPTH1-34 administered<br />

subcutaneously as a continuous infusion (to mimic signaling by locallyproduced<br />

PTHrP) or as once-daily injections. Femur and tibial growth<br />

plates were scanned by microCT and growth plate volumes were calculated.<br />

Growth plates were also assessed histologically. Results: Continuous<br />

delivery <strong>of</strong> hPTH1-34 provided the greatest effect for maintaining patent<br />

growth plates compared to once-daily subcutaneous injections. However,<br />

the dose <strong>of</strong> continuous hPTH1-34 giving the greatest protection was not<br />

well-tolerated beyond 1 week due to PTH-induced hyperparathyroidism,<br />

confirmed by clinical chemistry and histological assessment. Reduction in<br />

the dose <strong>of</strong> continuous hPTH1-34 improved tolerability, but provided less<br />

protection to the growth plate. Once-daily hPTH1-34 injection for two weeks<br />

was well-tolerated, with anabolic effects clearly observed by histology and<br />

microCT. The extent <strong>of</strong> closure in animals co-treated with LDE225 and<br />

once-daily hPTH1-34 injections was less compared to that caused by<br />

LDE225 treatment alone. Conclusions: As shown in this preclinical model,<br />

hPTH1-34 administration mitigates LDE225-induced growth plate closure.<br />

Although hPTH1-34 shows promise experimentally, current use is restricted<br />

in pediatric patients and further study will be needed before considering<br />

hPTH1-34 treatment concurrent with LDE225 therapy in children or<br />

adolescents.<br />

9564 General Poster Session (Board #42C), Sun, 8:00 AM-12:00 PM<br />

Effect <strong>of</strong> insulin-like growth factor 2 gene expression on pleuropulmonary<br />

blastoma and embryonal rhabdomyosarcoma. Presenting Author: Rajkumar<br />

Venkatramani, Children’s Hospital Los Angeles, Los Angeles, CA<br />

Background: The sarcomatous element in pleuropulmonary blastoma (PPB)<br />

<strong>of</strong>ten cannot be differentiated from embryonal rhabdomyosarcoma (ERMS)<br />

by histology, particularly when characteristic elements like cartilage and<br />

respiratory mucosa are inapparent. The initial diagnosis is <strong>of</strong>ten ERMS. A<br />

diagnosis <strong>of</strong> PPB is based on a combination <strong>of</strong> clinical (age, location) and<br />

pathologic features. We hypothesized that the sarcomatous portions <strong>of</strong> PPB<br />

are a form <strong>of</strong> ERMS indistinguishable from typical ERMS. We reasoned that<br />

a detailed comparison <strong>of</strong> RNA expression could clarify this hypothesis.<br />

Methods: Samples from 7 PPB patients were obtained from the rhabdomyosarcomatous<br />

portion <strong>of</strong> the tumor by macro-dissection after pathologic<br />

review <strong>of</strong> all material. Representative ERMS tumor tissue was selected from<br />

21 ERMS cases. FFPE tissue scrolls from each case were analyzed using<br />

Affymetrix Human Exon arrays after RNA extraction with Agincourt Formapure<br />

extraction kit and amplification using the NuGEN Ovation FFPE WTA<br />

kit and labeling with the Encore Biotin Module. Data analysis utilized<br />

<strong>Part</strong>ek and Genetrix s<strong>of</strong>tware. Results: All PPB cases and 7 <strong>of</strong> 21 ERMS<br />

cases were � 4 years old. 20 transcripts (10 annotated, 10 non-coding<br />

RNAs) were significantly differentially expressed in ERMS when compared<br />

to PPB patients. Insulin-like growth factor 2 (IGF2) was uniformly overexpressed<br />

in ERMS (19/21 � 200) but was expressed at low levels in PPB<br />

(p�0.001). 2 ERMS cases which had low level IGF2 expression were � 4<br />

years <strong>of</strong> age. Expression in the other 5 ERMS aged �4 years was similar to<br />

ERMS overall (range, 500-2,000). No other differences between the two<br />

approached this degree <strong>of</strong> significance, despite a common rhabdomyogenic<br />

phenotype in the sarcomatous areas <strong>of</strong> PPB. Conclusions: We conclude that<br />

PPB is a distinct entity from most ERMS, and is not driven by IGF2<br />

signaling. Rare cases <strong>of</strong> ERMS in the very young are more similar to PPB<br />

than common ERMS. This observation is highly significant, as IGF2 is<br />

known to drive growth in ERMS, generally related to demethylation<br />

secondary to LOH <strong>of</strong> 11p.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


622s Pediatric Oncology<br />

9565 General Poster Session (Board #42D), Sun, 8:00 AM-12:00 PM<br />

Circulating myeloid-derived suppressor cells in pediatric solid tumor<br />

patients. Presenting Author: John M. Goldberg, University <strong>of</strong> Miami Miller<br />

School <strong>of</strong> Medicine, Miami, FL<br />

Background: Cure rates have reached a plateau for many pediatric solid<br />

tumors. Identifying new therapeutic targets, biomarkers <strong>of</strong> response and<br />

prognostic indicators should improve clinical outcomes. Accumulation <strong>of</strong><br />

myeloid derived suppressor cells (MDSCs) is an important mechanism <strong>of</strong><br />

tumor mediated immune evasion. Increased levels <strong>of</strong> MDSCs in adult<br />

cancer patients correlate with a worse prognosis, and decrease in levels<br />

with treatment is associated with benefit. Little is known about MDSCs in<br />

childhood, or in children with cancer. This pilot measured levels <strong>of</strong> MDSCs<br />

in pediatric patients with cancer and healthy children. Methods: We<br />

enrolled subjects using an Institutional Review Board approved protocol<br />

after obtaining informed consent. Blood was obtained from 14 children<br />

with newly diagnosed or recurrent solid tumors at start <strong>of</strong> therapy. In 10 <strong>of</strong><br />

these patients, levels were also drawn after therapy. Blood was obtained<br />

once from 6 healthy children in a primary care <strong>of</strong>fice. Samples were<br />

obtained concurrently with complete blood counts. MDSCs were measured<br />

on fresh whole blood and were defined as Lin-1�/HLADR-/CD 33�/<br />

CD11b�/ by flow. Total MDSC numbers were then calculated. Results: The<br />

mean total number <strong>of</strong> MDSCs was 596 cells/ul at diagnosis for the 14<br />

children with cancer and 170 cells/ul for the 6 healthy children (t(18) �<br />

3.02, p � .0073). For the 10 children with cancer who had repeat values<br />

measured after treatment, MDSCs decreased in 4 and increased in 6. The<br />

mean initial MDSC count for these children was 494 cells/ul, and the mean<br />

post treatment count was 1716 cells/ul (t(9) � 1.81, p � .1036). Larger<br />

change scores tended to be associated with children treated with alkylator<br />

therapy followed by G-CSF. The results for percent MDSC in white cells<br />

mirrored those <strong>of</strong> total number. Conclusions: Circulating MDSCs were<br />

higher in pediatric cancer patients than healthy controls. Cancer treatment<br />

did not reliably reduce MDSC levels. After some treatments, the levels<br />

increased, potentially increasing immune tolerance. Further research is<br />

needed to determine if circulating MDSCs are a reliable predictive or<br />

prognostic marker in pediatric cancer, and whether they represent a<br />

potential target for therapeutic intervention.<br />

9567 General Poster Session (Board #42F), Sun, 8:00 AM-12:00 PM<br />

Diffuse pontine gliomas in children: Do changing strategies change results?<br />

Presenting Author: Rejin Kebudi, Istanbul University, Cerrahpasa Medical<br />

Faculty and Oncology Institute, Istanbul, Turkey<br />

Background: The prognosis <strong>of</strong> children with diffuse intrinsic pontine<br />

gliomas (DIPG) is dismal. Despite various studies undertaken to improve<br />

outcome, radiotherapy (RT) remains the standard treatment, which is<br />

mostly palliative. This study aims to evaluate characteristics and treatment<br />

outcome <strong>of</strong> children with DIPG in a single center. Methods: We retrospectively<br />

reviewed the demographic, clinical characteristics and treatment<br />

outcome <strong>of</strong> children with DIPG treated at Istanbul University, Oncology<br />

Institute from 1999 to 2011. We also evaluated the group that prospectively<br />

recieved RT with concurrent and adjuvant temozolamide after 2004.<br />

Results: 47 children (24 female, 23 male) with the median age <strong>of</strong> 7 years (6<br />

months-16 years) were analyzed. The median duration <strong>of</strong> symptoms was 30<br />

days (2-630 days). The frequent clinical findings were ataxia, strabismus<br />

and motor weakness. All patients received RT, 54-60 Gy to the tumor site.<br />

12 recieved only RT. 35 had concomitant and/or adjuvant chemotherapy<br />

with RT. 8 recieved cisplatinum, 7 vincristine. Since 2004, 20 patients<br />

recieved the institutional protocol consisting <strong>of</strong> temozolomide (TMZ) (75<br />

mg/m2/day) for 6 weeks concurrent with RT, followed by TMZ (200<br />

mg/m2/day) for 5 days every 28 days for 12 cycles or until progression.<br />

There was no major side effect due to TMZ, thrombocytopenia being the<br />

most frequent, but managable side effect. The median overall survival after<br />

diagnosis was 13 months (3-132 mo.) for the whole group. The median<br />

overall survival in 20 patients that received RT and TMZ [ 17 months<br />

(3-132 months)], was significantly superior than that in 12 patients that<br />

recieved only RT [ 12 months (3-20 months)] ( (p�0.03). Nimotuzumab<br />

was given to 4 patients that progressed after RT and TMZ. There was no<br />

major side effect due to nimotuzumab. One was stable for 1 year with<br />

significant clinical improvement, the others were stable for 5, 2 and 2<br />

months after nimotuzumab. Conclusions: In our series, the median survival<br />

was significantly superior in patients who received RT with concurrent and<br />

adjuvant temozolamide in comparison to patients that recieved RT alone.<br />

Nimotuzumab may be promising in some progressive patients, its role as<br />

upfront treatment needs further investigation.<br />

9566 General Poster Session (Board #42E), Sun, 8:00 AM-12:00 PM<br />

Intensive alternating 6-drug chemotherapy for high-risk nonmetastatic<br />

rhabdomyosarcoma in children and adolescents: Impact on survival.<br />

Presenting Author: Gerges Attia Demian, National Cancer Institute, Cairo<br />

University, Cairo, Egypt<br />

Background: Both 5-year overall survival (OS) and event free survival (EFS)<br />

for pediatric rhabdomyosarcoma (RMS) has increased in the last 3 decades<br />

through multimodality, risk-adapted management. The reported EFS for<br />

high risk RMS in children treated at the NCI in Egypt during the 1990s was<br />

50%. Using an intensive 6 drug alternating chemotherapy regimen in<br />

addition to local control measures was our aim to improve the outcome for<br />

this group <strong>of</strong> patients. Methods: Forty-six previously untreated patients,<br />

younger than 21 years <strong>of</strong> age, with localized high risk RMS received this<br />

regimen. High risk criteria included: (1) Localized tumors (T1) biopsied or<br />

incompletely resected, ortumors extending beyond the tissue or organ <strong>of</strong><br />

origin (T2) completely or incompletely resected at any site (excluding orbit,<br />

uterus, vagina, and paratestis); (2)All node positive patients with primary<br />

tumor at any site; and (3) All RMS with alveolar histology at any site.<br />

Chemotherapy regimen comprised 27 weeks <strong>of</strong> alternating 6 drugs (carboplatin,<br />

doxorubicin hydrochloride, ifosfamide, actinomycin D, etoposide,<br />

vincristine). Local therapy (surgery, radiotherapy, or both) was <strong>of</strong>fered at<br />

week 9. Results: Forty-six patients meeting high-risk criteria were recruited<br />

from September 2000 to November 2005. Median follow-up <strong>of</strong> survivors<br />

was 62 months. The 5-year OS and EFS for the whole group was 64% �<br />

10% and 47% � 8% respectively. The EFS was significantly affected by:<br />

the size <strong>of</strong> the tumor (�5 cmvs.� 5 cm, p� 0.03), SIOP UICC clinical<br />

stage (p � 0.004), IRS stage (p � 0.01), lymph node status (p � 0.02),<br />

surgery vs. incisional biopsy (p�0.01) and overall duration <strong>of</strong> time in which<br />

therapy was delivered (p � 0.04). There was significant toxicity, mainly<br />

hematologic, but only one treatment related fatality. Conclusions: The use<br />

<strong>of</strong> intensified alternating 6-drug CT did not improve the EFS compared with<br />

historical control although it was feasible to be delivered safely in a variety<br />

<strong>of</strong> outpatient settings. Surgical resection <strong>of</strong> the tumor is essential.<br />

Delivering therapy in a timely fashion appears to impact outcome and<br />

future investigations will focus on impediments to administering chemotherapy<br />

as scheduled.<br />

9568 General Poster Session (Board #42G), Sun, 8:00 AM-12:00 PM<br />

Demographics, treatment, and outcome <strong>of</strong> retinoblastoma: A large series<br />

with protocol-based management. Presenting Author: Vijay Anand Palkonda<br />

Reddy, Apollo Hospitals, Hyderabad, India<br />

Background: Retinoblastoma is the most common primary intraocular<br />

malignancy <strong>of</strong> childhood with incidence <strong>of</strong> 1 in 20,000 and the third most<br />

common pediatric cancer. In the previous decades, early diagnosis and<br />

treatment <strong>of</strong> retinoblastoma have improved survival rates in developed<br />

countries. Nevertheless, protocol based management help reduce the<br />

mortality rate associated with advanced tumors. The purpose <strong>of</strong> the study is<br />

to report demographics, management, and outcome <strong>of</strong> retinoblastoma in<br />

the Ocular oncology centre <strong>of</strong> South India. Methods: Consecutive case<br />

series <strong>of</strong> 1,543 eyes <strong>of</strong> 1,067 patients with 22 years <strong>of</strong> follow-up.<br />

Demographics, clinical features, treatment and outcome (survival, organ<br />

salvage and function salvage) were analyzed. Management comprised <strong>of</strong><br />

focal therapy for small tumors, standard triple drug chemoreduction for<br />

larger (�4mm) tumors, high dose chemoreduction and periocular chemotherapy<br />

for vitreous seeds, and enucleation for advanced tumors. Adjuvant<br />

therapy was provided in patients with histopathologic high-risk factors for<br />

systemic metastasis. Orbital retinoblastoma was managed with a multimodal<br />

treatment protocol. Results: Sixty-six percent were �3 years <strong>of</strong> age.<br />

Forty-five percent were bilateral. Symptoms were leucocoria (33%), vision<br />

loss (10%), proptosis (6%), and squint (6%). The tumor was intraocular in<br />

94% and orbital in 6%. Management was by protocol and included<br />

enucleation (52%), chemoreduction (26%), focal therapy (11%), and<br />

external beam radiation (5%). Fifty-five percent <strong>of</strong> enucleated patients had<br />

histopathologic risk factors for metastasis and received adjuvant chemotherapy.<br />

Chemoreduction with focal therapy resulted in eye and vision<br />

salvage in 92%. Overall, 94% survived. Conclusions: Protocol-based<br />

management <strong>of</strong> retinoblastoma provides excellent prognosis for survival,<br />

eye salvage and vision salvage.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


9569 General Poster Session (Board #42H), Sun, 8:00 AM-12:00 PM<br />

Long-term outcomes <strong>of</strong> children with Xp11.2 translocation renal cell<br />

carcinoma. Presenting Author: Hiroshi Asanuma, Department <strong>of</strong> Urology,<br />

Keio University School <strong>of</strong> Medicine, Tokyo, Japan<br />

Background: We aimed to assess the clinico-pathological characteristics<br />

and the long-term prognosis <strong>of</strong> Xp11.2 translocation renal cell carcinoma<br />

(Xp-RCC) in children. Methods: A total <strong>of</strong> 52 Japanese children with renal<br />

tumors were presented to our institutions. Of these, 5 (10%) had RCC, and<br />

all RCCs were Xp11.2 translocation subtype with positive nuclear transcription<br />

factor E3 immunostaining. We retrospectively reviewed the pathological<br />

and hospital records <strong>of</strong> these 5 patients. Results: In the 1 boy and 4 girls<br />

with an average age <strong>of</strong> 9 years 7 months at diagnosis, the most common<br />

presenting complains were gross hematuria (60%) and palpable abdominal<br />

mass (40%). All patients had unilateral disease and there was no special<br />

family or medical history in any children. Computerized tomography<br />

revealed characteristic calcification within the tumor in 4 <strong>of</strong> the 5 patients<br />

(80%). In the remaining case, the lesion had high density areas with<br />

microcalcification, as confirmed by histopathological study. In 3 patients<br />

with regional lymph node metastasis, calcification was also observed in the<br />

metastatic lesions. Stage <strong>of</strong> the disease were stage I in 1 patient, II in 1, IV<br />

without and with distant metastasis in 2 and 1, respectively. All patients<br />

underwent transabdominal nephrectomy with regional lymphadenectomy.<br />

One patient with stage I disease had multiple metastases 15 months after<br />

surgery and died <strong>of</strong> disease in spite <strong>of</strong> interferon and chemotherapy 4 years<br />

postoperatively. Three patients received adjuvant interferon therapy and 2<br />

<strong>of</strong> them are without evidence <strong>of</strong> recurrence postoperative 19 and 14 years,<br />

respectively. One <strong>of</strong> them had lymph node and lung metastases 12 years<br />

postoperatively. The remaining one patient had multiple lymph node, lung,<br />

bone, and dura mater metastases at diagnosis. The latter 2 patients have<br />

received targeted therapy (sunitinib, temsirolimus) without their progression<br />

for 6 and 12 months, respectively. Conclusions: Calcification within<br />

the tumor and/or metastatic lesions is characteristic findings suggestive <strong>of</strong><br />

Xp-RCC in children. Regional lymphadenectomy or targeted therapy may<br />

provide some benefit in selected pediatric patients. Long-term follow-up is<br />

essential in pediatric patients with RCC.<br />

9571 General Poster Session (Board #43B), Sun, 8:00 AM-12:00 PM<br />

Substitution <strong>of</strong> cisplatin for etoposide in 2 <strong>of</strong> 3 induction cycles <strong>of</strong> CAV/IE<br />

with subsequent irinotecan and vincristine for relapse in adults with Ewing<br />

sarcoma (ES) and primitive neuroectodermal tumor (PNET). Presenting<br />

Author: Daniel A. Rushing, Indiana University Melvin and Bren Simon<br />

Cancer Center, Indianapolis, IN<br />

Background: Treatment for adults with cyclophosphamide, doxorubicin and<br />

vincristine alternating with ifosfamide and etoposide (CAV/IE) for ES/PNET<br />

is based on the success reported in children however the same treatment in<br />

adults is less effective. 50% <strong>of</strong> relapsed disease is expected to occur in the<br />

lungs with median survival <strong>of</strong> 12.5 months (mo) after relapse. Since<br />

Cisplatin is used for relapse, we initially substituted cisplatin for etoposide<br />

in courses#2and#3<strong>of</strong>IEfor2patients (pts) who were not responding to<br />

IE as determined by serial CT scanning and then used it for all pts.<br />

Relapsed pts were treated with irinotecan � vincristine (IRI-V) for at least 2<br />

cycles as salvage therapy. We report the effect <strong>of</strong> cisplatin on influencing<br />

the relapse pattern and the effect <strong>of</strong> IRI-V on survival for pts after relapse<br />

Methods: A retrospective chart review <strong>of</strong> 19 consecutive adult pts (July<br />

2004 to June 2011), with newly diagnosed ES /PNET were treated with<br />

CAV/IE as per H. Grier NEJM 2003 and modified as a 14 day cycle.<br />

Cisplatin 20 mg/m2 I.V.qdX4wassubstituted for etoposide for courses #<br />

2 and 3 <strong>of</strong> IE and combined with ifosfamide as above (IC) followed by<br />

surgery or RT. For pts with relapse Irinotecan was given I.V. 20mg/m2 days<br />

1-5 & 8-12 � vcr I.V. 2mg days1&8q21days for 2 or more cycles. Repeat<br />

treatment with CAV, IE or IC was also utilized for relapse. We evaluated the<br />

toxicities, the pattern <strong>of</strong> relapse and the survival after relapse with these<br />

treatments Results: Grade IV hematologic toxicity occurred in 11 <strong>of</strong> 19 pts<br />

treated with CAV vs. 7<strong>of</strong> 19 pts treated with IE vs. 7 <strong>of</strong> 19 pts treated with<br />

IC and 1 <strong>of</strong> 11 pts treated with IRI-V but 3 <strong>of</strong> those 11 pts had Grade IV<br />

diarrhea. Median survival was 44.8� mo for 10 pts with non- metastatic<br />

and 23 mo for 9 pts with metastatic disease respectively. More importantly<br />

median survival from the time <strong>of</strong> relapse was 22.5� mo, (range 12.7 to<br />

31� mo). 11 pts relapsed: 2 pts only in bone, 9 pts Local only, 0 pts had<br />

distant lung recurrence Conclusions: Cisplatin appears to markedly alter the<br />

relapse pattern in adults with ES/PNET and Irinotecan �vcr appears to<br />

prolong survival in relapsed pts previously treated with cisplatin.<br />

Pediatric Oncology<br />

623s<br />

9570 General Poster Session (Board #43A), Sun, 8:00 AM-12:00 PM<br />

Preclinical study <strong>of</strong> a PARP inhibitor in neuroblastoma. Presenting Author:<br />

Anissa Addioui, Research Center CHU Sainte Justine, Montreal, QC,<br />

Canada<br />

Background: Neuroblastoma (NB) is the most common extracranial solid<br />

tumor <strong>of</strong> childhood. In spite <strong>of</strong> many therapeutic improvements, only 60%<br />

survive long term despite aggressive combinations <strong>of</strong> multi-agent chemotherapy.<br />

In previous studies, we have demonstrated that tumor initiating<br />

cells (TIC) expressing CD133 (CD133high ) in NB are more resistant to<br />

chemotherapy. Moreover, these cells express higher levels <strong>of</strong> PARP-1, a<br />

central protein involved in DNA repair. PARP-1 expression is significantly<br />

lower in NB usually showing spontaneous regression than in standard NB,<br />

suggesting an implication <strong>of</strong> PARP-1 in NB progression. The objective <strong>of</strong><br />

this study is to determine the efficacy in vitro <strong>of</strong> AG-014699 (AG), a PARPinhibitor,<br />

used in monotherapy or in combination to cisplatine (CP) and<br />

doxorubicine (DR), classical chemotherapeutic agents used in NB treatment,<br />

on NB cell survival. Methods: Six NB cell lines (parental or CD133high purified by flow cytometry (FACS)) were treated with AG alone or in<br />

association to CP or DR. PARP-1 ELISA protein assay was used to<br />

determine the optimal drug concentration needed to inhibit the protein.<br />

Cell survival was measured by MTT test. Western Blots were done to<br />

evaluate any apoptotic or autophagic pathway modulations. Quantification<br />

<strong>of</strong> DNA damage in treated cell was done by immun<strong>of</strong>luorescence <strong>of</strong> H2A-X<br />

protein. Results: We showed that a 4�M concentration <strong>of</strong> AG is sufficient for<br />

PARP-1 inhibition. One third <strong>of</strong> celllines presented a sensitivity to AG when<br />

used in monotherapy with an IC50 lower than 5�M. However, AG<br />

demonstrated synergistic effects when associated to DR, decreasing the<br />

IC50 by half, although none is observed when combined to CP. Sentitivity<br />

<strong>of</strong> the TIC did not appear to be more important than the bulk cells. With<br />

increasing concentration <strong>of</strong> AG, our WB showed no increase in cleaved<br />

Caspase-3 suggesting no modulation <strong>of</strong> the apoptotic pathway. However,<br />

autophagy seemed to be upregulated confirmed by an increase in cleaved<br />

LC3 II protein. Double strand breaks increased 2.5 folds when 4�M AGis<br />

added to the IC50 <strong>of</strong> DR. Conclusions: AG used in combination at<br />

potentially therapeutic doses shows promising results in NB. These results<br />

will allow for the improvement <strong>of</strong> NB treatments by introducing a new<br />

therapeutic strategy.<br />

9572 General Poster Session (Board #43C), Sun, 8:00 AM-12:00 PM<br />

High-dose thiotepa: Neurotoxicity and risk factors in children with solid<br />

tumors. Presenting Author: Christophe Maritaz, Institut Gustave Roussy,<br />

<strong>Clinical</strong> Pharmacy Department, Villejuif, France<br />

Background: Thiotepa (TTP) is a cytotoxic agent used in children with solid<br />

tumors at high doses followed by autologous stem cell transplantation<br />

(ASCT). During these treatments, neurological adverse events (NAE) were<br />

observed. Causal relationships and risk factors were investigated. Methods:<br />

Patients with solid tumors treated with high-dose thiotepa with ASCT<br />

between May 1987 and March 2011 were retrospectively identified.<br />

Clinico-biological data were collected and NAE were identified from<br />

medical and nursing records. Toxicity was graded according to the NCI<br />

CTCAE v4.03 classification. Imputability <strong>of</strong> thiotepa was assessed according<br />

to Begaud et al. method. Analysis <strong>of</strong> risk factors was assessed with<br />

Fisher’s exact test (alpha 0.05) and relative risk was estimated by<br />

calculating odds ratios with their 95% confidence interval. Results: 251<br />

patients received 307 courses (56 patients received twice) <strong>of</strong> high-dose<br />

thiotepa (600 mg/m²: 82 courses; 720 mg/m²: 76 courses; 900 mg/m²:<br />

149 courses) with ASCT. The median age was 8.33 years (range, 1 - 31.2<br />

years). 81 NAE (26.4%) were described. NAE were considered “possibly”<br />

related to TTP in 9 courses, “likely” related to TTP in 35 courses and “very<br />

likely” related to TTP in 13 courses. Other NAE were assessed “doubtful”<br />

or “incompatible”. In these 57 NAE, neurological symptoms appeared at a<br />

median time <strong>of</strong> 2 days (range, 0-4days) after the introduction <strong>of</strong> TTP.<br />

Symptoms were headache, tremor, dizziness and confusion, blurred vision,<br />

seizure, pyramidal tract syndrome, cerebellar syndrome, opsoclonusmyoclonus<br />

syndrome and coma. These events disappeared without sequelea<br />

in a median time <strong>of</strong> 3 days (range, 1-8days). TTP was reintroduced in<br />

21 cases. NAE reappeared in 12 cases. For 3 patients with seizure during<br />

the first course, premedication with clonazepam prevented NAE during the<br />

second course. The use <strong>of</strong> analgesic for moderate to severe pain, such as<br />

tramadol or codeine, increased the probabiblity to have NAE (OR 5.6467;<br />

95CI [1.5523-21.2786]; p 0.037). Conclusions: In our study, the incidence<br />

<strong>of</strong> NAE related to TTP was 18.6%. The outcome was favorable<br />

without sequelea in all cases. TTP could be reintroduced after NAE. The<br />

association <strong>of</strong> TTP with tramadol increases the risk <strong>of</strong> neurotoxicity.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


624s Pediatric Oncology<br />

9573 General Poster Session (Board #43D), Sun, 8:00 AM-12:00 PM<br />

Do children, adolescents, and young adults with s<strong>of</strong>t tissue sarcoma benefit<br />

from early detection <strong>of</strong> recurrences? Results <strong>of</strong> a population-based study.<br />

Presenting Author: Tobias M. Dantonello, Olgahospital, Pediatrics 5,<br />

Klinikum Stuttgart, Stuttgart, Germany<br />

Background: Most patients with pediatric s<strong>of</strong>t tissue sarcoma (PSTS)<br />

achieve a 1st remission with primary therapy, but recurrences are frequent<br />

and drive mortality. Relapse detection (RD) is the foundation <strong>of</strong> posttreatment<br />

surveillance. The rationale for frequent follow-up investigations<br />

is RD in an early stage with better salvage options. There is however little<br />

evidence regarding the value <strong>of</strong> early asymptomatic RD and it was not<br />

investigated by any larger or population-based study in PSTS. We determined<br />

whether early radiographic RD in asymptomatic patients improves<br />

post-relapse outcome. Methods: Patients aged � 18 years at first referencereviewed<br />

PSTS-diagnosis registered with CWS were enrolled if a 1st relapse<br />

occurred between 1/2003 and 7/2010. The methods <strong>of</strong> RD and their<br />

impact on outcome were evaluated. The data were correlated with the<br />

German Childhood Cancer Registry (GCCR), which receives all information<br />

about cancer patients � 18 years. Results: 235 patients from Germany (n �<br />

212) and Sweden/Switzerland (n � 23) were eligible. In 2010 all German<br />

PSTS-patients were registered both with the GCCR and CWS. The most<br />

common PSTS-histiotypes were embryonal (n � 76) and alveolar (n � 74)<br />

rhabdomyosarcoma. 156 patients had primary nonmetastatic PSTS. The<br />

median time to relapse diagnosis (TTRD) was 1.4 years. 118 recurrences<br />

were locoregional, 38 combined and 79 metastatic. In 229 patients, the<br />

methods leading to RD could be evaluated. 139 recurrences were detected<br />

because <strong>of</strong> clinical signs and symptoms, primarily observed by patients<br />

and/or parents in 72%. 90 RDs were radiographic in asymptomatic<br />

patients. TTRD did not differ between symptomatic and asymptomatic<br />

patients. As <strong>of</strong> 12/2011, 77/235 patients are currently alive with a median<br />

follow-up <strong>of</strong> 5.4 years. Overall survival from primary surgery at 3-, 5-, and<br />

10-years for all 235 patients was 49%, 34% and 22%. It was 43%, 29%<br />

and 23% for the symptomatic and 59%, 40% and 21% for the asymptomatic<br />

group (p � 0.05). Conclusions: In this series, early radiographic RD in<br />

asymptomatic PSTS-patients had no significant impact on TTRD or<br />

post-relapse survival, although asymptomatic patients tended to survive<br />

longer.<br />

9575 General Poster Session (Board #43F), Sun, 8:00 AM-12:00 PM<br />

Use <strong>of</strong> pharmacokinetic modeling to optimize direct inhibition <strong>of</strong> the<br />

�undruggable� target EWS-FLI1 <strong>of</strong> Ewing sarcoma. Presenting Author:<br />

Jeffrey Toretsky, Georgetown Lombardi Comprehensive Cancer Center,<br />

Washington, DC<br />

Background: Chimeric transcription factors are ideal anti-cancer targets;<br />

however, they lack enzymatic activity thus rendering them ‘undruggable’<br />

proteins. The EWS-FLI1 fusion protein <strong>of</strong> Ewing sarcoma (ES) has been<br />

validated as an anticancer target both alone and as a partner <strong>of</strong> RNA<br />

Helicase A (RHA). Our prior work identified the small molecule YK-4-279<br />

as a specific inhibitor that blocks RHA from binding to EWS-FLI1. Blocking<br />

this interaction leads to apoptosis. We have modeled YK-4-279 treatment<br />

using ES cells to establish the duration <strong>of</strong> YK-4-279 exposure that leads to<br />

apoptosis. Methods: Apoptosis was measured by caspase-3 cleavage. A<br />

validated plasma detection method allows for HPLC measurement <strong>of</strong><br />

YK-4-279. Pharmacokinetic (PK) models <strong>of</strong> YK-4-279 for both intraperitoneal<br />

(IP) and intravenous (IV) administration were developed in SD rats and<br />

BL6 mice. Results: ES cell lines were exposed to YK-4-279 over a<br />

time-course, followed by a caspase-3 activity assay that demonstrated a<br />

minimum <strong>of</strong> 16 hours exposure was necessary to achieve maximal<br />

apoptosis using either 3 or 10 microM compound. A dose-dependency<br />

assay demonstrated that while apoptosis could be achieved with 30 – 80<br />

microM YK-4-279 after 4 hours <strong>of</strong> treatment, caspase-3 activity was less<br />

than or equal to 3 microM for 16 hours. Rat PK modeling demonstrated a<br />

t1/2 <strong>of</strong> 30 minutes following IV administration. BL6 mice demonstrated<br />

similar kinetics to the rat. SCID/bg mice, which are necessary for tumor<br />

efficacy studies, demonstrated approximately 50% faster clearance than<br />

either rat or BL6 mice. Absolute bioavailability for IP administration was<br />

41%. Conclusions: Models will be created to optimize IP dosing regimen.<br />

The optimized IP dosage and dosing intervals will be evaluated in tumor<br />

bearing animals in order to advance development <strong>of</strong> YK-4-279. The results<br />

<strong>of</strong> these studies will be used to guide toxicology studies in animals. In<br />

addition, pharmacodynamics models are being developed to compare<br />

YK-4-279 levels with functional activity. The combined results <strong>of</strong> these<br />

investigations will lead to human clinical trials for this first-in-chemical<br />

class, first-in-mechanism drug candidate.<br />

9574 General Poster Session (Board #43E), Sun, 8:00 AM-12:00 PM<br />

A multidisciplinary treatment comprising standard chemotherapy with<br />

vincristine, doxorubicin, cyclophosphamide, ifosfamide, and etoposide<br />

for Ewing sarcoma family <strong>of</strong> tumor (ESFT) in Japan: Results <strong>of</strong> the first<br />

Japan Ewing Sarcoma Study (JESS) 04. Presenting Author: Atsushi<br />

Makimoto, National Cancer Center Hospital, Tokyo, Japan<br />

Background: Survival rate <strong>of</strong> ESFT in Japan was reportedly less than 50% in<br />

1990s. The JESS started in order to improve this condition by conducting<br />

clinical trials for ESFT in Japan. Methods: This is a phase II, nonrandomized<br />

trial to test safety and efficacy <strong>of</strong> multidisciplinary treatment<br />

for non-metastatic ESFT. Age under 30 is eligible. Chemotherapy regimens<br />

are alternating vincristine 1.5 mg/m2 on day 1, doxorubicin 37.5 mg/m2 on<br />

days 1 and 2, cyclophosphamide 1200 mg/m2 on day 1 (VDC) and<br />

ifosfamide 1800 mg/m2 on days 1 through 5 and etoposide 100 mg/m2 on<br />

days 1 through 5 (IE) repeating every 21 days for 52 weeks. Local<br />

treatment includes surgical resection and/or radiation therapy, <strong>of</strong> which<br />

dosage varies from 0 to 55.8 Gy based on margin <strong>of</strong> the resection and<br />

pathological response. Primary endpoint is 3 year progression free survival<br />

(PFS). Statistical design was made to test whether the lower limit <strong>of</strong> 90%<br />

confidence interval (CI) <strong>of</strong> PFS will exceed the threshold <strong>of</strong> 60%. Planned<br />

sample size is 53 patients including 10% ineligible patients. Results:<br />

Fifty-three patients were registered from December 2004 to May 2008.<br />

Among the 53 patients, 7 patients were judged ineligible after the<br />

registration (metastatic disease 1, changed diagnoses 6). Forty six patients<br />

were considered as per protocol set and used for efficacy analyses. Three<br />

year PFS is 71.7% (90% CI: 0.59 – 0.81). Estimated 5 year PFS and OS<br />

are both 69.6%. Radiographic response rate (RR) in evaluable 38 patients<br />

is 71.1% and pathological RR in 25 is 52.0%. In safety data, hematological<br />

toxicities are significant such as grade 4 neutropenia observed in 98%.<br />

Although there is no previously unknown adverse event reported, there are<br />

three patients who experienced secondary malignancies (ALL 1, MDS 1,<br />

osteosarcoma 1). Conclusions: A multi-disciplinary treatment comprising<br />

standard multi-agent chemotherapy with vincristine, doxorubicin, cyclophosphamide,<br />

ifosfamide and etoposide improved outcome <strong>of</strong> Ewing sarcoma<br />

family <strong>of</strong> tumor in Japan although statistical confirmation <strong>of</strong> absolute<br />

efficacy was not successful.<br />

9576 General Poster Session (Board #43G), Sun, 8:00 AM-12:00 PM<br />

Pharmacokinetics (PK) <strong>of</strong> the chimeric anti-GD2 antibody, ch14.18, in<br />

children with high-risk neuroblastoma. Presenting Author: Ami V. Desai,<br />

The Children’s Hospital <strong>of</strong> Philadelphia, Philadelphia, PA<br />

Background: The PK <strong>of</strong> ch14.18, a chimeric monoclonal antibody (cMoAb)<br />

that significantly improves survival in high-risk neuroblastoma, was previously<br />

reported to be highly variable in children, and its clearance (CL, 130<br />

mL/h · m2 ) and volume <strong>of</strong> distribution (Vdss, 32 L/m2 ) were �10-fold<br />

higher than the CL and Vdss <strong>of</strong> other cMoAbs. Characterizing the PK <strong>of</strong><br />

ch14.18 and the factors responsible for its variability could lead to<br />

alternative dosing strategies that reduce toxicity. Methods: Detailed PK<br />

sampling was performed prior to, during and for 25 d after 4 daily 10 h<br />

infusions <strong>of</strong> 25 mg/m2 <strong>of</strong> ch14.18 administered with sargramostim<br />

(ANBL0032) in children enrolled on an IRB-approved institutional correlative<br />

protocol supported by United Therapeutics Corp. Ch14.18 concentrations<br />

were measured with a validated ELISA with a lower limit <strong>of</strong> detection<br />

<strong>of</strong> 0.44 mcg/mL. PK parameters were derived using non-compartmental<br />

methods and reported as median (range). Results: 5 males and 4 females,<br />

median (range) age 3.5 (1-7) yrs, have been enrolled. Six were studied on<br />

cycle 1 (C1), 2 on C5 and 1 on C3; 3 <strong>of</strong> 6 patients studied on C1 were<br />

re-studied on C3. Data from 3 patients are not included because pretreatment<br />

plasma samples contained a substance that interfered with the<br />

ELISA. The Cmax after the 4th daily infusion was 14 (10-24) mcg/mL, and<br />

ch14.8 was undetectable at day 25 in 3/6 patients. The half-life was 220<br />

(120-390) h, and the CL was 11 (3.8-16) mL/h · m2 , which is similar to<br />

the average CL <strong>of</strong> 4 other cMoAbs (11 mL/h · m2 ). The Vdss <strong>of</strong> 2.8 (1.2-3.9)<br />

L/m2 approximated blood volume and was similar to the Vdss <strong>of</strong> other<br />

cMoAbs (1.7 L/m2 ) and humanized MoAbs (2.8 L/m2 ). AUC0-� in 2 patients<br />

studied twice were 3440 and 1540 mcg · h/mL on C1 and 2760 and 2690<br />

mcg · h/mL on C3. Conclusions: Ch14.18 CL, Vdss and half-life in children<br />

are similar to those in adults receiving ch14.18 at the same dose and<br />

similar to other cMoAbs. Ch14.18 PK in children was less variable than<br />

previously reported. The identity <strong>of</strong> the interfering substance in the plasma<br />

<strong>of</strong> some patients requires further investigation. A limited sampling strategy<br />

will be developed from these data for use in larger multi-institutional PK<br />

studies <strong>of</strong> ch14.18.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


9577 General Poster Session (Board #43H), Sun, 8:00 AM-12:00 PM<br />

Ifomide/pirarubicin/etoposide/carboplatin (ITEC) as second-line chemotherapy<br />

for hepatoblastoma. Presenting Author: Eiso Hiyama, Hiroshima<br />

Univerisity, Natural Science Center for Basic Research and Development,<br />

Hiroshima, Japan<br />

Background: The Japanese Study Group for Pediatric Liver Tumor is running<br />

cooperative treatment studies on hepatoblastoma (HBL) since 1991. The<br />

main aim in JPLT-1 study was to evaluate the efficacy <strong>of</strong> cisplatin/<br />

pirarubicin (CITA). A total <strong>of</strong> 145 cases were registered and their 5-year<br />

overall survival (OS) and event-free survival (EFS) were 73.4% and 66%,<br />

respectively (J Pediatr Surg 37: 851-6, 2002). Then, JPLT-2 protocol, in<br />

which CITA is kept as the first-line treatment, and ifomide, etoposide,<br />

pirarubicin, and carboplatin (ITEC) is the second-line regimen for CITAresistant<br />

cases, was launched to evaluate the cure rate <strong>of</strong> risk-stratified<br />

HBL: standard risk HBL (a tumor involving three or fewer sectors <strong>of</strong> the<br />

liver) and high risk HBL (a tumor involving all sectors <strong>of</strong> the liver or with<br />

metastasis). Methods: In JPLT-2, 281 HBL cases were registered in JPLT-2<br />

between 1999 and 2010, and 69 cases underwent ITEC protocol due to<br />

poor response to the CITA regimen. To evaluate the efficacy <strong>of</strong> the ITEC<br />

regimen, surgical resectability and outcome <strong>of</strong> these 69 patients who<br />

underwent this treatment. Results: These 69 cases were divided into 53<br />

high-risk (metastatic, involvement <strong>of</strong> all sectors <strong>of</strong> the liver, vascular<br />

invasion and/or extra-hepatic intra-abdominal disease) and 16 standardrisk<br />

HBL (all others). All cases were initially treated with the CITA regimen<br />

and then underwent the ITEC regimen due to poor response to CITA.<br />

Complete resection <strong>of</strong> the liver tumor could be achieved in 48 patients<br />

(69.6%) consisting <strong>of</strong> 13 (81%) standard, 35 (66%) high- risk cases. The<br />

5-year OS and EFS <strong>of</strong> the cases with standard risk HB were 96% and 76%,<br />

while that <strong>of</strong> the cases with high risk HB was 54% and 34%. The late phase<br />

complications in 281 cases were 3 cases with maldevelopment, 13 with<br />

cardiac complications, 20 with ototoxicity and 5 with second malignancies.<br />

Conclusions: As compared with the CITA-sensitive cases, ITEC regimen<br />

achieved similar rates <strong>of</strong> survival for CITA-resistant cases both in standard<br />

and high-risk HBL cases, indicating ITEC is effective as a second-line<br />

chemotherapeutic regimen for HBL.<br />

9579 General Poster Session (Board #44B), Sun, 8:00 AM-12:00 PM<br />

A phase II trial <strong>of</strong> docetaxel and irinotecan (DI) in children and young adults<br />

with recurrent or refractory Ewing sarcoma family <strong>of</strong> tumor (ESFT).<br />

Presenting Author: Jong Hyung Yoon, Center for Pediatric Oncology,<br />

National Cancer Center, Goyang, South Korea<br />

Background: Patients with ESFT resistant to second-line therapy, such as<br />

topotecan plus cyclophosphamide, have a dismal prognosis and few<br />

therapeutic options. Docetaxel (D), a microtubule inhibitor, demonstrated<br />

activity in patients with ESFT (Zwerdling T et al. Cancer 2006:106:1821-<br />

1828). Irinotecan (I), a toposiomerase I inhibitor, is being evaluated in<br />

clinical trials for ESFT. Despite the different mechanisms <strong>of</strong> action <strong>of</strong> D and<br />

I, and their activities as a single-agent against ESFT, DI combination has<br />

not been previously evaluated in ESFT. Thus, we prospectively performed a<br />

single-arm, phase II trial <strong>of</strong> DI in a single center in Korea. Methods: Patients<br />

�30 years with ESFT, who failed second-line therapy, were eligible for the<br />

study. D was administered IV over 60 minutes at a dose <strong>of</strong> 100 mg/m2 on<br />

Day 1, and I at a dose <strong>of</strong> 80 mg/m2 over 90 minutes on Days 1 and 8 <strong>of</strong> a<br />

21-day cycle until disease progression. The primary end-point was objective<br />

response assessed by RECIST; the secondary end-point was safety. All<br />

toxicities were graded according to the National Cancer Institute’s Common<br />

Toxicity Criteria (version 4.0). Results: Of seven eligible patients (4 males;<br />

median age, 17 years (range 5-21)), 4 were recurrent/refractory cases, and<br />

3 refractory cases. Median number <strong>of</strong> previous regimens was 6 (range 3-6).<br />

One CR, 1 PR, and 5 PD were obtained by DI combination (median number<br />

<strong>of</strong> cycles, 2 (range 1-15)), with objective response (CR�PR) rate <strong>of</strong> 2/7<br />

(28.6%). One patient with PR achieved CR with subsequent surgery. CR <strong>of</strong><br />

these two patients lasted 8.9 months and 10.6 months. Of all seven<br />

patients fully evaluable for toxicity, grade 4 neutropenia occurred in 7<br />

(100%); grade 3 and 4 thrombocytopenia in 1 (14%) and 6 (86%),<br />

respectively; grade 3 pericardial effusion, paresthesia, motor weakness,<br />

oral mucositis, each in one (14%). Two patients required �1 dose delay; 2<br />

required �1 dose reduction due to adverse events, but no one experienced<br />

treatment-related mortality. Conclusions: DI combination demonstrated<br />

activity in children and young adults with ESFT who were heavily pretreated,<br />

and was feasible.<br />

Pediatric Oncology<br />

625s<br />

9578 General Poster Session (Board #44A), Sun, 8:00 AM-12:00 PM<br />

Cell cycle and apoptosis regulatory protein (CARP)-1 expression in neuroblastoma.<br />

Presenting Author: Santosh S. Hanmod, Children’s Hospital <strong>of</strong><br />

Michigan, Detroit, MI<br />

Background: Neuroblastoma (NB) is the most common extra-cranial solid<br />

tumor in children. The prognosis for patients with high risk NB is still poor.<br />

Study <strong>of</strong> additional prognostic factors will enhance current understanding<br />

<strong>of</strong> the tumor biology and risk stratification. CARP-1 is a recently identified<br />

molecule mediating apoptosis signaling by agents like doxorubicin and<br />

etoposide. Since these agents are used in the front line treatment <strong>of</strong> NB, we<br />

tested whether CARP-1 expression could be another prognostic factor in<br />

NB. Methods: CARP-1 expression was examined by Western blot in a pair <strong>of</strong><br />

NB cell lines, SK-N-SH and SK-N-SH Dox. We reviewed medical records <strong>of</strong><br />

patients with NB at Children’s Hospital <strong>of</strong> Michigan from 2000-2009 and<br />

examined CARP-1 expression by immune-histochemistry in the archived,<br />

formalin fixed paraffin embedded NB tissues. CARP-1 expression was<br />

recorded as positive or negative. Correlation <strong>of</strong> CARP-1 expression and<br />

progression free survival (PFS) was calculated. Results: CARP-1 expression<br />

was detected in SK-N-SH but not SK-N-SH Dox, a doxorubicin-resistant<br />

derivative <strong>of</strong> SK-N-SH, suggesting that resistance to doxorubicin is associated<br />

with decreased level <strong>of</strong> CARP-1 expression in NB cells. Of the 30<br />

cases studied, 53 % (16/30) expressed CARP-1. There was no significant<br />

difference in N-myc amplification status (13% vs.14%, p�0.3), or<br />

proportion <strong>of</strong> unfavorable Shimada histology (60% vs. 64%, p�0.5)<br />

between CARP-1 positive vs. negative groups. Twenty three percent (7/22)<br />

patients progressed or relapsed, including 2/16 (13%) in CARP-1 positive<br />

group vs. 5/14 (36%) in CARP-1 negative group (p�0.1). There was no<br />

significant difference in the PFS at 1 year (94% vs.79%, p�0.2) and 3<br />

year (86% vs.71%, p�0.2) between CARP-1 positive vs. negative groups.<br />

Conclusions: CARP-1 expression was decreased in doxorubicin resistant NB<br />

cell line. CARP-1 expression was detected in approximately half (53%) <strong>of</strong><br />

NB tumors. Patients with NB who expressed CARP-1 showed trend towards<br />

better 1- & 3-year PFS as compared to those who did not express CARP-1,<br />

however, the results are not statistically significant which could be due to<br />

the small sample size. Further study with a larger number <strong>of</strong> patients is<br />

warranted to clarify these findings.<br />

9580 General Poster Session (Board #44C), Sun, 8:00 AM-12:00 PM<br />

Assessment <strong>of</strong> peripheral blood T regulatory cells (Tregs) in PNET/Ewing<br />

sarcoma: A prospective study. Presenting Author: Tilak Tvsvgk, Dr. B.R.A.,<br />

IRCH, New Delhi, India<br />

Background: Tregs in bone marrow have been previously evaluated in PNET<br />

patients; however, data on peripheral blood ccirculating Tregs is lacking.<br />

The objective <strong>of</strong> our study was to determine baseline Treg frequency in<br />

PNET patients and correlate the same with patient characteristics and<br />

outcome. Methods: Samples <strong>of</strong> 5ml venous blood were obtained from 38<br />

newly diagnosed PNET patients at diagnosis along with six healthy controls.<br />

Flow cytometric analysis was done for detecting Treg cells<br />

[CD4�CD25�FoxP3�]. Results: Thirty-eight patients with median age 17<br />

years; male/female ratio <strong>of</strong> 5.5:1 had significantly higher baseline Tregs<br />

than healthy controls [9.17%�.3.08 vs 3.16�1.49%; p��0.0001].<br />

Eight patients (21.1%) had fever at baseline presentation. The disease was<br />

extra-skeletal in one and metastatic at baseline in 11 (28.9%) patients.<br />

Ten patients relapsed on standard protocol <strong>of</strong> therapy and seven died. The<br />

median Treg frequency was 8.84% (Range: 2.49-16.31). When the Tregs<br />

were categorized as high and low based on the median value, patients with<br />

fever had a significantly higher Tregs than those without fever<br />

[11.3%�.3.5% vs 8.6% �. 2.7%; p�0.02]. No significant association <strong>of</strong><br />

peripheral blood Treg cells frequency was noted with other factors like age,<br />

sex, metastatic disease, relapse or death. The EFS was 55% and OS 70%<br />

<strong>of</strong> the entire cohort at a median follow up <strong>of</strong> 14 months. There was no<br />

significant difference in the EFS or OS between the high and low Treg cell<br />

groups [EFS- 52% vs 64%; p�0.99 and OS-75% vs 70%; p�0.26].<br />

Conclusions: This is the first study on circulating Tregs in PNET, and it<br />

shows that the peripheral blood Treg frequencies are higher in these<br />

patients as compared to healthy controls. Further, PNET patients with fever<br />

had significantly higher Treg frequency. However, Tregs did not differ with<br />

respect to metastatic disease at presentation, EFS or OS.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


626s Pediatric Oncology<br />

9581 General Poster Session (Board #44D), Sun, 8:00 AM-12:00 PM<br />

A phase I trial <strong>of</strong> MK 2206 in children with refractory solid tumors: A<br />

Children’s Oncology Group study. Presenting Author: Christine L. Phillips,<br />

Cincinnati Children’s Hospital Medical Center, Cincinnati, OH<br />

Background: AKT, a serine threonine protein kinase, is activated downstream<br />

<strong>of</strong> phosphatidylinositol 3-kinase (PI3K) which transmits signals<br />

from cytokines, growth factors and oncoproteins to multiple targets.<br />

Activated AKT regulates survival, proliferation, and growth. The PI3K/AKT<br />

pathway is downstream <strong>of</strong> most <strong>of</strong> the common growth factor tyrosine<br />

kinase receptors in cancer, e.g., EGFR, HER2, IGFR, etc., and is a driver <strong>of</strong><br />

tumor progression in many cancers. AKT protein kinase is activated in many<br />

pediatric solid tumors, including glioblastoma, malignant rhabdoid tumors,<br />

neuroblastoma, synovial sarcoma, rhabdomyosarcoma and medulloblastoma.<br />

MK2206, an oral allosteric AKT1, 2,3 inhibitor, has demonstrated<br />

antitumor activity in in vitro and in vivo cancer models.A phase I trial<br />

evaluating MK2206 was conducted in children with refractory solid<br />

tumors. Methods: Using a rolling-6 design, MK2206 was administered<br />

either once every 7 days (schedule 1), or once every other day (schedule 2)<br />

in a 28-day cycle. Serial PK studies were obtained on day 1, cycle 1 and<br />

trough samples were obtained on days 7, 14, 21 and 28. Biological studies<br />

included analysis <strong>of</strong> PI3K/PTEN/AKT-cell signaling pathway in pre and<br />

post-therapy in PBMC and in tumors at diagnosis or recurrence. Results:<br />

Forty-five patients [23 males, median age 13.6 years (range 3.1-21.9)]<br />

with malignant glioma (14), ependymoma (4), hepatocellular carcinoma<br />

(3), gliomatosis cereberi (2) or other tumors (22) were enrolled; 34 were<br />

fully evaluable for toxicity (schedule 1 n�17; schedule 2 n�17). Schedule<br />

1 DLTs included: grade 3 dehydration in 1/6 patients at 28 mg/m2 ; grade 4<br />

hyperglycemia and neutropenia in 1/6 patients at 45 mg/m2 . There were no<br />

DLTs at 35 mg/m2 and dose level 4 (58 mg/m2 ) is currently open for patient<br />

accrual with one enrollment. Schedule 2 DLTs included: grade 3 alkaline<br />

phosphatase in 1/6 patients at 90 mg/m2 ; grade 3 rash in 1/6 patients at<br />

120 mg/m2 ), and grade 3 rash in 2/6 patients at 155 mg/m2. Conclusions:<br />

The recommended pediatric phase II dose <strong>of</strong> weekly MK2206 is 120<br />

mg/m2 and the last cohort <strong>of</strong> patients to the every other day dosing schedule<br />

<strong>of</strong> MMK206 is enrolling. PK and PD data are currently being analyzed and<br />

will be presented.<br />

9584 General Poster Session (Board #44F), Sun, 8:00 AM-12:00 PM<br />

Integration <strong>of</strong> genomic and proteomic data to identify MYCN-regulated<br />

genes, proteins, and interaction networks in neuroblastoma cells. Presenting<br />

Author: Kolbrun Kristjansdottir, University <strong>of</strong> Chicago, Chicago, IL<br />

Background: Neuroblastoma is the most common extracranial solid tumor<br />

found in children. The most common marker prognostic <strong>of</strong> poor outcome is<br />

amplification <strong>of</strong> the MYCN oncogene, yet the biological programs by which<br />

MYCN affects its aggressive phenotype are largely unknown. In order to<br />

identify biological pathways affected by MYCN amplification, we performed<br />

global analysis <strong>of</strong> genes and proteins in the Tet 21/N neuroblastoma cell<br />

line. Methods: MYCN expression in Tet21/N cells is regulated by tetracycline.<br />

Gene arrays were performed on MYCN-high and MYCN-low Tet21/N<br />

cells and SH-EP parental cells using Affymetrix GeneChip Human Exon 1.0<br />

ST. For proteomic analysis, the cells were labeled with stable isotopes for<br />

relative quantitation, enriched for phosphoproteins to enhance detection <strong>of</strong><br />

signaling proteins, and analyzed by GeLC-MS/MS analysis. Integrative<br />

pathway analysis was performed on the genomic and proteomic datasets<br />

using DAVE and GeneGo. Results: Integrating genomic and proteomic data<br />

in MYCN-high and MYCN-low expressing cells identified 88 proteins and<br />

300 genes that change in abundance. We compare these results with<br />

previous studies and find both novel and previously identified genes and<br />

proteins. Integrating proteomic and genomic data identified over 50 gene<br />

products that are present in both analyses and are altered in abundance in<br />

response to modulating MYCN expression. The majority <strong>of</strong> these have<br />

discordant abundance changes, with protein levels more frequently altered<br />

with no change in gene expression. We have validated changes in protein<br />

levels using Western blots including NPM1 and MATR3. We present<br />

interaction networks and pathways that correlate with MYCN expression.<br />

Conclusions: We showed a significant discordance between gene and<br />

protein expression, underscoring the need for integrative genomic and<br />

proteomic analysis to describe complex systems. Our analysis identified<br />

previously reported and novel genes and proteins and networks regulated by<br />

MYCN expression that may provide new insights into the biology <strong>of</strong><br />

MYCN-amplified tumors.<br />

9583 General Poster Session (Board #44E), Sun, 8:00 AM-12:00 PM<br />

Development <strong>of</strong> an open-source, flexible framework for interinstitutional<br />

data sharing and collaboration. Presenting Author: Chaim Kirby, University<br />

<strong>of</strong> Chicago, Chicago, IL<br />

Background: <strong>Clinical</strong> information, “-omic” datasets, and tissue samples are<br />

becoming more difficult to harmonize and manage for advanced data<br />

mining. We believe that clinical research data can be centralized and<br />

provide direct access to sample availability and associated data from a<br />

variety <strong>of</strong> information stores. Methods: We obtained a standardized set <strong>of</strong><br />

anonymized patient data from the International Neuroblastoma Risk<br />

Group. The cohort consists <strong>of</strong> more than 11,000 children diagnosed<br />

worldwide between 1974 and 2002. The data consist <strong>of</strong> 34 metrics, such<br />

as age at diagnosis, stage <strong>of</strong> tumor, and other clinical and biological<br />

markers. We instantiated the dataset into a Postgres database, and using<br />

the Django web framework, created a data model for rapid development <strong>of</strong><br />

tools and views and built a front-end interface for generating complex<br />

queries. To test the feasibility <strong>of</strong> accessing information on disparate and<br />

geographically distinct data samples, we have a formal agreement with the<br />

Children’s Oncology Group Tumor Bank at The Research Institute at<br />

Nationwide Children’s Hospital. Based on query results, we consume the<br />

Tumor Bank tissue inventory data through a web-facing application<br />

programming interface. The end-user is presented only with the number <strong>of</strong><br />

patients who match their query search terms and for whom tissue samples<br />

are available. Results: We have completed our initial implementation and<br />

have agreements for collaboration with other international consortium<br />

groups. We have created a paradigm for statisticians to securely update and<br />

add data, and a verification system checks for internal validity and provides<br />

a report <strong>of</strong> the transaction. Our system can initiate queries and accept<br />

results in a variety <strong>of</strong> standards-compliant formats, and will be available in<br />

demonstration form by May 2012. Conclusions: Querying patient data while<br />

interrogating external sources allows researchers to observe which ancillary<br />

data and samples are available and to quickly download data or request any<br />

samples. While designed around a neuroblastoma dataset, our system can<br />

be applied to a variety <strong>of</strong> clinical scenarios and will be made available<br />

through an open-source license.<br />

9585 General Poster Session (Board #44G), Sun, 8:00 AM-12:00 PM<br />

Proton radiotherapy for rhabomyosarcoma: Preliminary results from a<br />

multicenter prospective study. Presenting Author: Torunn I. Yock, Massachusetts<br />

General Hospital, Boston, MA<br />

Background: Pediatric rhabdomyosarcoma (RMS) is commonly cured with<br />

chemotherapy and radiation. However, late effects <strong>of</strong> radiotherapy (RT) can<br />

be disabling. Proton RT irradiates less normal tissue, which should result in<br />

fewer late side effects <strong>of</strong> treatment. The purpose <strong>of</strong> this study was to<br />

describe the disease control and side effect pr<strong>of</strong>ile <strong>of</strong> proton RT in RMS<br />

patients (pts). Methods: Eligible pts included those with localized disease<br />

and metastatic embryonal RMS if age 2-10. All pts were treated with VAC<br />

(vincristine, actinomycin, cyclophosphamide) based chemotherapy and<br />

proton RT, median dose 50.4 Gy (36-50.4 GyRBE). Concurrent enrollment<br />

in COG protocols was allowed. All pathology/imaging was reviewed at the<br />

treating institution. Results: 47 pts with RMS were prospectively enrolled<br />

from January 2005 to June 2011 and evaluable for analysis. Median age<br />

was 3.1 yrs, (range 0.6-15.6 years; M/F ratio 23:24). There were 1, 7, 37,<br />

and 2 Group I, II, III and IV and 14, 12, 19 and 2, Stage I-IV pts<br />

respectively, and 33 with embryonal 14 with alveolar/other. Most common<br />

sites included PM (48.9%), orbital (23.4%) bladder/prostate (6.4%), H&N<br />

non-PM (6.4%), extremities (4.3%), trunk/abdomen 2.1%), perineal/anal<br />

region (2.1%) and other (6.4%). Median follow-up is 15.2 months.<br />

One/two-year overall survival (OS) and progression-free survival (PFS) for<br />

the entire group is 94/81% (OS) and 79/73% (PFS). 2-year OS for stage<br />

I,II/III,IV pts is 91/66% (OS, p�0.114) and two-year PFS for these pts is<br />

86/57%, respectively, (p�0.083. 16 (34%) had grade 3/4 acute toxicities<br />

attributable to the radiation during treatment, the most common <strong>of</strong> which<br />

was mucositis/oral pain (12.8%), anorexia (4.3%), and erythema (4.3%).<br />

Among the 24 pts analyzable for late toxicities with at least 2 yrs <strong>of</strong> follow<br />

up, there were no grade 3 or 4 late toxicities attributable to radiation.<br />

Conclusions: Early results <strong>of</strong> this prospective trial demonstrate comparable<br />

disease outcomes and thus far limited late effects in a young pediatric RMS<br />

population. However, additional follow up is needed to determine if protons<br />

truly reduce rates and severity <strong>of</strong> late effects compared with photon cohorts<br />

published in the literature.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


9586 General Poster Session (Board #44H), Sun, 8:00 AM-12:00 PM<br />

A national survey <strong>of</strong> general internists’ preferences and knowledge gaps<br />

regarding the care <strong>of</strong> childhood cancer survivors. Presenting Author: Tara<br />

O. Henderson, University <strong>of</strong> Chicago Pritzker School <strong>of</strong> Medicine, Chicago,<br />

IL<br />

Background: Although most childhood cancer survivors (CCS) report obtaining<br />

health care in the community, primary care physicians’ views and<br />

knowledge regarding the long-term follow-up (LTFU) care <strong>of</strong> CCS are largely<br />

unknown. Methods: Surveys were mailed to a random sample <strong>of</strong> 1,907<br />

general internists under age 65 years from the <strong>American</strong> Medical Association<br />

Physician Masterfile in November 2011. A second mailing to nonresponders<br />

is ongoing. Results: 892 (47%) physicians have responded.<br />

Respondents have practiced a median <strong>of</strong> 10 years (range: 3-20), and see a<br />

median <strong>of</strong> 70 patients/week (range: 40-100). 46% are in solo/group<br />

practice, 17% in multi-specialty practice, and 14% in academic practice.<br />

In the last five years, 53% have seen at least one CCS, 71% <strong>of</strong> whom have<br />

never received a treatment summary. 85% prefer to care for CCS in<br />

consultation with a cancer center based physician. A vignette <strong>of</strong> a<br />

29-year-old female treated for Hodgkin lymphoma with mantle radiation at<br />

age 16 was provided and participants were asked a series <strong>of</strong> questions<br />

regarding monitoring for late effects. The percentage <strong>of</strong> responses that were<br />

concordant with available LTFU Surveillance Guidelines were: breast<br />

cancer, 29%; cardiac, 15%; thyroid, 77%; and all three recommendations,<br />

only 5%. By logistic regression, greater likelihood <strong>of</strong> concordance with at<br />

least one surveillance recommendation was associated with being female<br />

(OR�2.0 95% CI 1.3-3.1), seeing more patients/week (OR�1.4 per SD<br />

increase, 95% CI 1.1-1.7) and more years in practice (OR�1.3 per SD<br />

increase, 95% CI 1.0-1.6). The two modalities felt to be most useful for<br />

independent care <strong>of</strong> CCS by internists were access to LTFU guidelines and<br />

receiving a patient-specific care plan from the cancer center. Conclusions:<br />

Although the majority <strong>of</strong> internists are willing to follow CCS, they appear<br />

unfamiliar with the available LTFU guidelines and prefer to care for<br />

patients in collaboration with a cancer center based physician.<br />

9588 General Poster Session (Board #45B), Sun, 8:00 AM-12:00 PM<br />

Insurance retention in adolescent and young adult survivors <strong>of</strong> malignancies<br />

in a vertically integrated health care system. Presenting Author: Robert<br />

Michael Cooper, Kaiser Permenante, Los Angeles, CA<br />

Background: Adolescents and Young Adults (AYA) with cancer are a unique<br />

population <strong>of</strong> patients. This group is afflicted with a wide range <strong>of</strong> cancer<br />

types. AYA patients <strong>of</strong>ten have delays in diagnosis due to issues related to<br />

insurance and continuity <strong>of</strong> care and have less participation clinical trials<br />

related to other age groups. Survivors are at risk <strong>of</strong> late effects <strong>of</strong> treatment.<br />

A vertically integrated health care system with a comprehensive electronic<br />

medical record provides an ideal opportunity to care for adolescent and<br />

young adult (AYA) cancer survivors and conduct survivorship research. In<br />

this system, patients who complete oncology care return to primary medical<br />

care but can be tracked in a systematic fashion. Methods: We evaluated the<br />

insurance retention <strong>of</strong> survivors <strong>of</strong> AYA malignancies by querying the<br />

Cancer Registry and identified 13,240 known survivors <strong>of</strong> malignancies<br />

diagnosed within the system between 1990 and 2005. We looked at the<br />

percentage <strong>of</strong> survivors who were still cared for in our system at certain time<br />

points after diagnosis. We then reanalyzed the group by ethnicity, disease,<br />

and age at diagnosis. Results: The retention rate for all patients was 88% at<br />

1 year, 64 % at 5 years, and 52% at 10 years. The retention rates were<br />

similar regardless <strong>of</strong> ethnicity. When we looked at the influence <strong>of</strong> age at<br />

time <strong>of</strong> diagnosis we noted that the younger the patients were at diagnosis<br />

the lower the percentage <strong>of</strong> those retaining insurance at the 10 year time<br />

point. We analyzed the group by the more common cancers and noted that<br />

patients diagnosed with breast cancer had greatest retention at 10 years <strong>of</strong><br />

68%. Conclusions: The lengthy insurance retention <strong>of</strong> adolescent and<br />

young adult cancer survivors makes a vertically integrated medical care<br />

system an ideal population laboratory for cancer survivorship research in<br />

this understudied population <strong>of</strong> cancer survivors.<br />

Pediatric Oncology<br />

627s<br />

9587 General Poster Session (Board #45A), Sun, 8:00 AM-12:00 PM<br />

Patient-perceived facilitators in the transition <strong>of</strong> survivorship care from the<br />

pediatric to adult care setting. Presenting Author: Karim Thomas Sadak,<br />

Children’s National Medical Center, Washington, DC<br />

Background: Survival rates in childhood cancer continue to rise. A new<br />

challenge has emerged in optimizing the transition <strong>of</strong> pediatric survivorship<br />

care to similarly focused programs that are age-appropriate. The purpose <strong>of</strong><br />

this study is to identify components <strong>of</strong> a clinical survivorship program that<br />

facilitate the transition <strong>of</strong> care for adult survivors <strong>of</strong> childhood cancer from<br />

the pediatric to adult care-setting. Methods: A descriptive study <strong>of</strong> childhood<br />

cancer survivors was conducted using a cross-sectional study design.<br />

A questionnaire was used to identify which clinical components <strong>of</strong> a<br />

survivorship program most influenced the decision to transition care to an<br />

adult medical center. Primary data collection occurred through the mail.<br />

Secondary mechanisms included an online version <strong>of</strong> the questionnaire<br />

and in-person enrollment during survivorship clinic visits. 140 survivors<br />

were eligible but only 129 had valid addresses and received the questionnaire.<br />

The study endpoint was achieved when the response rate reached<br />

80% (n�103). A descriptive review <strong>of</strong> clinical program components was<br />

performed using a frequency analysis. Results: Of 129 invited survivors,<br />

103 participated (80%). Mean and median age-range was 20-24 years.<br />

There were 49 males (48%) and 54 females (52%). The most common<br />

diagnoses were leukemia (40/103), lymphoma (20/103), and CNS tumors<br />

(9/103). When asked if the participant was willing to transition their<br />

survivorship care to an adult facility, 97 (95%) responded affirmatively.<br />

The clinical components most frequently rated “Very Important” in the<br />

decision to transition survivorship care were the acceptance <strong>of</strong> insurance<br />

(80/103, 78%) followed by the presence <strong>of</strong> providers knowledgeable in<br />

childhood cancer (68/103, 66%). The clinical components most frequently<br />

rated “Very Important” or “Important” were flexible scheduling<br />

(102/103, 99%) followed by comprehensive care being <strong>of</strong>fered (101/103,<br />

98%). Conclusions: The decision to transition survivorship care to ageappropriate<br />

care-settings is complex and not well understood. Issues<br />

related to insurance, clinical team composition, and scheduling appear to<br />

be most important for young adult survivors making this decision.<br />

9589 General Poster Session (Board #45C), Sun, 8:00 AM-12:00 PM<br />

Psychological study <strong>of</strong> adolescent and young adult (AYA) cancer patients<br />

and their parents throughout and beyond their cancer treatment: A pilot<br />

study. Presenting Author: Helen Hatcher, University <strong>of</strong> Cambridge, Department<br />

<strong>of</strong> Oncology, Cambridge, United Kingdom<br />

Background: The cancer journey for an AYA is <strong>of</strong>ten complicated by<br />

diagnostic delays, poor prognosis and negative physical and psycho-social<br />

consequences. Distress <strong>of</strong> young patients and survivors is <strong>of</strong>ten overlooked<br />

and the real incidence <strong>of</strong> psychopathology remaining hidden. We aimed to<br />

understand the psychological impact and identify which factors most affect<br />

the patient experience. Methods: 172 patients on the Cambridge AYA<br />

register were invited to take part. Patients were asked to complete 2<br />

questionnaires, SCL90 and RSQ, measuring psychological symptoms and<br />

response styles respectively, and to take part in a semi-structured interview.<br />

Parents were asked to complete the York Survey and be interviewed.<br />

Results: 33 patients and 45 parents/others were recruited. Questionnaires<br />

completed: 33 SCL90 and RSQ; 42 York Survey. Interview data from: 23<br />

patients, 26 parents, 3 others. Patient sample: 16 male, 17 female; mean<br />

age (recruitment) � 20.76 (16-26); age (diagnosis) � 17.26 (12-24).<br />

Diagnoses evenly distributed across solid and haematological malignancies.<br />

All patients had experienced significant levels <strong>of</strong> psychological<br />

distress at some point in their cancer journey. 51% reached caseness on<br />

the SCL90 (showed a symptom pr<strong>of</strong>ile consistent with a high risk <strong>of</strong> a<br />

disorder). Caseness increased with age and was correlated with higher<br />

rumination scores. Positive influences included family, friend and peer<br />

support, good communication and support from the AYA service and<br />

specialist nurses. Negative influences included delayed diagnosis, poor<br />

communication styles, anxiety about long term effects and lack <strong>of</strong> appropriate<br />

psychological support. Conclusions: We have shown evidence <strong>of</strong><br />

elevated psychological symptoms in this population. Patients and their<br />

families have prioritized clear needs; early diagnosis, high quality <strong>of</strong><br />

information and communication, access to psychological help at each<br />

stage and post treatment help in gaining independence and planning for a<br />

positive future. We should work towards meeting these needs for this<br />

vulnerable group.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


628s Pediatric Oncology<br />

9590 General Poster Session (Board #45D), Sun, 8:00 AM-12:00 PM<br />

Trends in survival <strong>of</strong> childhood solid tumors diagnosed 1985-2008 in the<br />

Nordic countries. Presenting Author: Milada Cvancarova, Cancer Registry<br />

<strong>of</strong> Norway and University <strong>of</strong> Oslo, Oslo, Norway<br />

Background: Classification <strong>of</strong> childhood solid tumors differs greatly from<br />

tumors diagnosed in adults. Thus establishment <strong>of</strong> childhood cancer<br />

registries is essential to monitor changes and progress in cancer care.<br />

Methods: All individuals born in the Nordic countries are allocated a unique<br />

ID number and have access to comprehensive health care services. Further,<br />

it is required by law that all new cases <strong>of</strong> cancer are reported to cancer<br />

registries. This makes it possible to achieve almost complete registration <strong>of</strong><br />

all cancer cases and their status together with information regarding the<br />

cancer tumour and its treatment. Status registrations were made partly<br />

from national population registries and partly from death registries only, as<br />

conditions differ between countries. Crude survival was modelled using the<br />

Kaplan-Meier method stratified by selected diagnostic groups, age groups<br />

and diagnostic period. The crude estimates were compared with log-rank<br />

tests. Results: In total, 12,343 children �15 yrs <strong>of</strong> age were diagnosed with<br />

cancer during 1986-2010 in the Nordic regions (Denmark, Finland,<br />

Iceland, Norway, Sweden) and classified according to Birch & Marsden<br />

classification. Data were collected at the NOPHO solid tumour registry<br />

located at the Cancer Registry <strong>of</strong> Norway. The overall incidence for the<br />

whole period was 11.2 per 100,000 children per year. The overall 5-year<br />

survival for all solid tumours reached 80.0 %, 95% CI [79.3 to 80.7] %.<br />

There was a significant increase in survival for those diagnosed after 1991,<br />

p � 0.001. Children diagnosed when older than 10 years reached higher<br />

5-yrs survival than those diagnosed �5 yrs, 82.4% and 78.3%, respectively.<br />

The highest 5-yrs survival was seen for retinoblastoma (97.0%),<br />

germ.cell neoplasm (90.8%), renal tumours (88.3%) and lymphomas<br />

(88.2%). The lowest 5-yrs survival was observed for sympathetic nervous<br />

system (65.4%). The highest increase in survival was seen for non-Hodgkin<br />

lymphoma pts, 77.2% diagnosed 1986-1990 vs. 85.9% diagnosed<br />

2001-05. Conclusions: There has been some improvement in survival in<br />

recent time periods, especially for some diagnoses. However, there were no<br />

changes in overall survival wrt sex and only very limited changes in overall<br />

survival by age groups.<br />

9592 General Poster Session (Board #45F), Sun, 8:00 AM-12:00 PM<br />

Effectiveness <strong>of</strong> using cardiac biomarkers to detect late anthracycline<br />

cardiotoxicity in childhood leukemia survivors. Presenting Author: Beata<br />

Mladosievicova, Comenius University, School <strong>of</strong> Medicine, Bratislava,<br />

Slovakia<br />

Background: Cardiotoxicity is usually detected only when clinical symptoms<br />

or progressive cardiac dysfunction has already occurred. Cardiac biomarkers<br />

(troponin T and N-terminal fragment brain natriuretic peptide precursor)<br />

have been hypothesized to reflect subclinical anthracycline<br />

cardiotoxicity earlier than echocardiography. This study aims to assess the<br />

effectiveness <strong>of</strong> using cTNT and NTproBNP in asymptomatic long-term<br />

survivors <strong>of</strong> childhood leukemia treated with and without antracyclines<br />

(ANT). Methods: Sixty-nine childhood leukemia survivors 5-25years after<br />

completion <strong>of</strong> therapy were evaluated with immunochemical analysis <strong>of</strong><br />

cTnT and NT-proBNP and echocardiography. Patients from group I (n �<br />

36) received combined therapy with anthracyclines (ANT) with total<br />

cumulative dose 95-600 (median 221) mg/m2 , patients from group II (n �<br />

33) received therapy without anthracyclines (nonANT). Control group<br />

consisted from 44 age- and gender-matched apparently healthy subjects.<br />

Results: Levels <strong>of</strong> NTproBNP were significantly higher in ANT group than in<br />

controls (median 51,52 vs 17,37 pg/ml; p�0.0026). Patients treated with<br />

ANT had significantly increased median values <strong>of</strong> NTproBNP compared<br />

with patients in non ANT group (51,52 vs 12,24 pg/ml; p�0.0002). CTnT<br />

levels remained below the diagnostic cut-<strong>of</strong>f levels in all groups. No patient<br />

had echocardiographic abnormalities, but significant differences were<br />

found in mean values <strong>of</strong> left ventricular ejection fraction and deceleration<br />

time between patients treated with and without ANT. Conclusions: Assessment<br />

<strong>of</strong> plasma NTproBNP concentrations may be an effective tool for<br />

detection <strong>of</strong> late subclinical cardiac damage in survivors <strong>of</strong> childhood<br />

leukemia previously treated with low ANT doses. Higher NTproBNP levels<br />

in patients after ANT therapy might reflect an initial stage <strong>of</strong> cardiotoxicity<br />

before the development <strong>of</strong> echocardiographic abnormalities. This study<br />

was supported by a grant from Ministry <strong>of</strong> Health 2007/42-UK-18,<br />

Slovakia.<br />

9591 General Poster Session (Board #45E), Sun, 8:00 AM-12:00 PM<br />

Bleomycin-associated lung toxicity in childhood cancer survivors. Presenting<br />

Author: Alexandra Patricia Zorzi, The Hospital for Sick Children,<br />

Toronto, ON, Canada<br />

Background: Bleomycin has been established as a pulmonary toxin, but the<br />

risk for toxicity in survivors <strong>of</strong> childhood cancer is poorly characterized.<br />

Methods: We conducted a cross-sectional study <strong>of</strong> lung function in survivors<br />

<strong>of</strong> childhood Hodgkin lymphoma and germ cell tumor treated with bleomycin<br />

at our institution between 1997 and 2010. We assessed their most<br />

recent post-therapy pulmonary function test (PFT). Spirometry and lung<br />

volumes were categorized as normal, restrictive, obstructive or mixed.<br />

Diffusing capacity <strong>of</strong> carbon monoxide (DLCO) was categorized as normal or<br />

abnormal. Results: 195 patients were treated with bleomycin. Ten died <strong>of</strong><br />

non-pulmonary causes. Of 185 survivors, 143 (77%) had complete data<br />

available for analysis. Median cumulative bleomycin dose was 60U/m2 (IQR 30-60). Three patients (2%) had a history <strong>of</strong> acute bleomycin toxicity.<br />

PFTs were performed a median <strong>of</strong> 2.3 years (IQR 1.4-4.9) from completion<br />

<strong>of</strong> therapy. Spirometry was abnormal in 58 patients (41%); <strong>of</strong> whom 5 (9%)<br />

had respiratory symptoms. 42 (70%) had obstructive, 11 (18%) restrictive<br />

and 5 (9%) mixed ventilatory defects. Abnormalities were mild in 53<br />

(91%), moderate in 3 (5%) and severe in 2 (4%). DLCO was abnormal in<br />

27 patients, 26 (96%) <strong>of</strong> whom had mildly reduced DLCO and were<br />

asymptomatic. Univariate analysis did not demonstrate a significant<br />

association between gender, smoking, lung metastases, lung radiation,<br />

chemotherapy regimen, or autologous transplant and abnormal lung<br />

function. Disease relapse was associated with abnormal lung function<br />

(p�0.01). Smoking (p�0.04) and relapse (p�0.03) were associated with<br />

abnormal DLCO. The odds ratio <strong>of</strong> developing abnormal spirometry for each<br />

1unit/m2 increase in bleomycin was 1.01 (95% CI 1.00-1.02, p�0.07).<br />

Conclusions: Childhood cancer survivors treated with bleomycin frequently<br />

have evidence <strong>of</strong> asymptomatic abnormalities on PFT. The current recommendation<br />

for pulmonary function testing in childhood cancer survivors<br />

appears justified.<br />

TPS9594 General Poster Session (Board #45G), Sun, 8:00 AM-12:00 PM<br />

A phase II study <strong>of</strong> dasatinib therapy in children and adolescents with<br />

newly diagnosed chronic phase chronic myelogenous leukemia (CML-CP)<br />

or Philadelphia chromosome-positive (Ph�) leukemias resistant or intolerant<br />

to imatinib. Presenting Author: Michel Zwaan, Erasmus University<br />

Medical Center-Sophia Children’s Hospital, Rotterdam, Netherlands<br />

Background: Dasatinib is a BCR-ABL inhibitor approved for treatment in<br />

adult patients (pts) with newly diagnosed Ph� CML-CP; CML resistant/<br />

intolerant to prior therapy, including imatinib; and Ph� acute lymphoblastic<br />

leukemia (ALL). There are no established dasatinib treatment regimens<br />

for children/adolescents with relapsed/refractory leukemia, but pediatric<br />

trials are underway. A phase I dose-escalation study <strong>of</strong> dasatinib in<br />

pediatric pts with refractory solid tumors (n�28) and imatinib-refractory,<br />

Ph� leukemia (n�11) reported a maximum tolerated dose <strong>of</strong> 85 mg/m2 twice daily in solid-tumor pts and at least a partial cytogenetic response<br />

(CyR) in all evaluable CML pts (n�9) (Aplenc, J Clin Oncol 2011).<br />

Preliminary results from a phase I dose-escalation study in pediatric pts<br />

with subtypes <strong>of</strong> relapsed/refractory leukemia (NCT00306202) indicate<br />

that dasatinib was well tolerated up to 120 mg/m2 (Zwaan, Blood 2010<br />

[abstr 2265]). Further study <strong>of</strong> dasatinib in pediatric pts is warranted.<br />

Methods: To evaluate the safety and efficacy <strong>of</strong> dasatinib monotherapy in<br />

children/adolescents with newly diagnosed CML-CP or Ph� leukemias<br />

resistant/intolerant to imatinib, a phase II nonrandomized, global study <strong>of</strong><br />

dasatinib in pts birth to �18 y is ongoing (NCT00777036): Cohort 1 (C1),<br />

Ph� CML-CP pts resistant/intolerant to imatinib; Cohort 2 (C2), Ph� ALL,<br />

accelerated or blast phase CML pts resistant/intolerant to or relapsed after<br />

imatinib therapy; or Cohort 3 (C3), newly diagnosed, treatment-naïve Ph�<br />

CML-CP pts. Treatments are once daily with dasatinib 60 mg/m2 (C1/C3) or<br />

80 mg/m2 (C2) for �24 months. Primary endpoints are major CyR (C1),<br />

complete hematologic response (C2), and complete CyR (C3). Secondary<br />

endpoints include safety, tolerability, best response, time to/duration <strong>of</strong><br />

response, survival, and molecular response rates. BCR-ABL mutations are<br />

evaluated. First patient first visit was March 2009; estimated trial completion<br />

is September 2016. As <strong>of</strong> January 2012, 63 pts (n�27 aged �12 y;<br />

n�36 aged �12 y) have been treated in C1/C2 (n�41) and C3 (n�22).<br />

Enrollment is ongoing at 79 sites.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


TPS9595 General Poster Session (Board #45H), Sun, 8:00 AM-12:00 PM<br />

Intraperitoneal radioimmunotherapy (RIT) for desmoplastic small round<br />

cell tumor (DSRCT). Presenting Author: Shakeel Modak, Memorial Sloan-<br />

Kettering Cancer Center, New York, NY<br />

Background: DSRCT, a rare sarcoma <strong>of</strong> adolescents and young adults arises<br />

from serosal surfaces typically the peritoneum. It is characterized by<br />

t(11;22)(p13;q12) chromosomal translocation; and is lethal in �80% <strong>of</strong><br />

patients despite aggressive surgery and chemoradiotherapy. Disease recurrence<br />

<strong>of</strong>ten presents as multifocal peritoneal implants, making it uniquely<br />

suited for intraperitoneal (IP) targeting. Since DSRCT is radiosensitive, we<br />

hypothesize that improving local control with targeted radiotherapy may<br />

reduce relapse and enhance survival. IP RIT, by virtue <strong>of</strong> prolonged<br />

residence time, and slow/incomplete transfer to the circulation, may<br />

selectively target IP disease while minimizing organ toxicity. The anti-4Ig-<br />

B7H3 murine monoclonal antibody 8H9 binds to 96% <strong>of</strong> primary DSRCT<br />

with restricted reactivity to normal tissue (Med Pediatr Oncol 39:547).<br />

131I-8H9 targets DSRCT xenografts and has proven safe at 80mCi when<br />

injected intrathecally (J Neurooncol 97:409). 124I-8H9, a novel positronemitting<br />

radioimmunoconjugate is ideally suited for quantitative imaging<br />

and pharmacokinetic (PK) studies. Methods: We have initiated a phase I<br />

study to test the safety <strong>of</strong> IP RIT using 131I-8H9 in patients with DSRCT<br />

(clinicaltrials.gov NCT01099644). Cohorts <strong>of</strong> 3-6 patients are treated with<br />

131 2 2 I-8H9 at escalated doses from 30mCi/m -60mCi/m as a single IP<br />

injection. An IP tracer dose <strong>of</strong> 2mCi124I-8H9 is given prior to 131I-8H9 to<br />

acquire high-resolution PET images and data on 8H9 tumor targeting and<br />

biodistribution. PK is also studied using serial blood draws. Patients are<br />

monitored for toxicity clinically and biochemically. Response to treatment<br />

is assessed using RECIST criteria and via a follow up 124I-8H9 PET scan if<br />

tumor targeting is noted on initial imaging. Hematopoietic stem cells are<br />

harvested a priori to reverse myelosuppression if any. Three cohorts <strong>of</strong><br />

patients (n�9) up to 50mCi/m2 have completed therapy thus far without<br />

dose-limiting toxicity or myelosuppression. This is the first study <strong>of</strong> IP RIT<br />

in pediatrics, and the first to use PET for IP RIT. Data obtained will be<br />

critical in establishing safety <strong>of</strong> IP 131I-8H9 in children and young adults,<br />

and in designing phase II trials to study efficacy <strong>of</strong> IP RIT for DSRCT.<br />

TPS9597^ General Poster Session (Board #46B), Sun, 8:00 AM-12:00 PM<br />

The BERNIE study: A phase II study evaluating addition <strong>of</strong> bevacizumab<br />

(Bv) to chemotherapy in children and adolescents with metastatic rhabdomyosarcoma<br />

(mRMS) and non-rhabdomyosarcoma s<strong>of</strong>t tissue sarcoma<br />

(mNRSTS). Presenting Author: Julia C. Chisholm, The Royal Marsden NHS<br />

Foundation Trust, Sutton, United Kingdom<br />

Background: Despite therapeutic advances, patient outcomes in mRMS and<br />

mNRSTS remain poor. The phase I study (Glade-Bender et al., J Clin Oncol.<br />

2008) indicated that Bv is well tolerated in children with refractory solid<br />

tumors and yielded pharmacokinetic (PK) data that support further studies<br />

<strong>of</strong> Bv in childhood cancer. Reports <strong>of</strong> Bv used in children with solid tumors<br />

showed safety pr<strong>of</strong>iles consistent with data from adults. Methods: In this<br />

phase II trial, 150 patients aged 6 months to 18 years who present with<br />

mRMS or mNRSTS are randomized to receive 18 months <strong>of</strong> standard<br />

combined modality therapy as per EpSSG guidelines, either alone or with<br />

Bv. Treatment consists <strong>of</strong> 2 phases: induction therapy [9 three-weekly<br />

cycles including 4 cycles <strong>of</strong> IVADo (ifosfamide, vincristine, actinomycin D,<br />

and doxorubicin), followed by 5 cycles <strong>of</strong> IVA (i.e., without doxorubicin)]<br />

and maintenance therapy (12 four-weekly cycles <strong>of</strong> vinorelbine and<br />

cyclophosphamide). Local therapy is considered after the 6th induction<br />

cycle. Primary endpoint is event-free survival (EFS). PK sampling is<br />

performed on all patients randomized to the experimental arm during the<br />

first 4 cycles <strong>of</strong> induction. After the primary efficacy and safety analysis, all<br />

patients who have not met the primary endpoint are followed for at least 47<br />

months for survival and long-term effects <strong>of</strong> treatment. The study enrolled<br />

75 patients between July 2008 and January 2012; 37 patients discontinued<br />

study treatment (including 17 patients who died, all due to disease<br />

progression) and 9 patients have completed study treatment.<br />

Pediatric Oncology<br />

629s<br />

TPS9596 General Poster Session (Board #46A), Sun, 8:00 AM-12:00 PM<br />

The HERBY study: A phase II open label, randomized, multicenter,<br />

comparative study <strong>of</strong> bevacizumab (Bv)-based therapy in pediatric patients<br />

with newly diagnosed supratentorial high-grade glioma (HGG). Presenting<br />

Author: Jacques Grill, Institut Gustave Roussy, Villejuif, France<br />

Background: Despite therapeutic advances, outcomes in pediatric HGG<br />

remain poor. The phase I study (Glade-Bender et al., J Clin Oncol. 2008)<br />

indicated that Bv is well tolerated in children with refractory solid tumors<br />

and yielded pharmacokinetic (PK) data that support further studies <strong>of</strong> Bv in<br />

childhood cancer. Reports <strong>of</strong> Bv used in children with solid tumors showed<br />

safety pr<strong>of</strong>iles consistent with data from adults. Methods: 120 evaluable<br />

patients aged 3-18 years with newly diagnosed histologically confirmed<br />

WHO grade 3or 4 HGG are randomized to receive standard combined<br />

modality therapy as currently adopted worldwide by the pediatric neurooncology<br />

community with or without Bv. Treatment consists <strong>of</strong> 6 weeks <strong>of</strong><br />

concomitant TMZ and local radiotherapy, followed by a 4-week TMZ<br />

treatment break and 48 weeks <strong>of</strong> adjuvant TMZ with or without Bv every<br />

other week. Primary endpoint is event-free survival (EFS). Progression is<br />

based on RANO criteria. Secondary endpoints are overall survival (OS),<br />

safety, feasibility, and tolerability. PK sampling is performed during cycles<br />

1-4 <strong>of</strong> the adjuvant TMZ phase on all patients randomized to receive Bv.<br />

Health-related quality <strong>of</strong> life, neurocognitive functions, MGMT methylation<br />

status, functional changes in tumor based on magnetic resonance diffusion<br />

& perfusion imaging and spectroscopy are explored as well as the<br />

correlation <strong>of</strong> biomarkers with clinical activity and adverse events. All<br />

randomized patients will be followed for at least 3 years. Analysis <strong>of</strong> EFS<br />

and secondary endpoints will be performed after the 120 patients evaluable<br />

for EFS have been followed for 1 year. Multimodal imaging will provide<br />

a platform to develop new imaging criteria for pediatric neuro-oncological<br />

treatment response. An updated OS and safety analysis will be performed 3<br />

years after the last patient has been randomized. The first patient was<br />

randomized in October 2011; completion <strong>of</strong> the study is expected in 2016.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


630s Sarcoma<br />

10000 Oral Abstract Session, Mon, 3:00 PM-6:00 PM<br />

Predicting survival in primary resected retroperitoneal s<strong>of</strong>t tissue sarcoma:<br />

A specific nomogram built on three major sarcoma center data sets.<br />

Presenting Author: Alessandro Gronchi, Fondazione IRCCS Istituto Nazionale<br />

Tumori, Milano, Italy<br />

Background: To build a specific nomogram for predicting postoperative<br />

overall survival (OS) in primary retroperitoneal s<strong>of</strong>t tissue sarcoma (RPS). A<br />

few single institutions attempts are already available, but due to the rarity<br />

<strong>of</strong> the disease they are based on limited number <strong>of</strong> cases with inherent<br />

limitations. In an attempt to improve the predictive capability <strong>of</strong> such a<br />

prognostic model we utilized series from 3 major sarcoma centers. Methods:<br />

We included patients with primary localized RPS resected with curative<br />

intent between 1999 and 2009 at 3 institutions (1 European and 2 North<br />

<strong>American</strong>s). Univariable (Kaplan Meier plots) and multivariable (Cox<br />

model) analyses were carried out. Prognostic covariates were: Age (modeled<br />

as continuous covariate using 3 knot restricted cubic spline transformation);<br />

Tumor size (modeled as continuous covariate using 3 knot<br />

restricted cubic spline transformation); Grade (I,II,III); Histological subtype<br />

(Leiomyosarcoma, DD Liposarcoma, WD Liposarcoma, MPNST, Pleomorphic<br />

Sa, SFT, Other); Multifocality (no,yes); Quality <strong>of</strong> surgery<br />

(macroscopically complete, incomplete); radiation therapy (RT, done/not<br />

done). A backward selection procedure, based on the Akaike Information<br />

Criterion (AIC), was applied to select the Cox model covariates. The model<br />

discriminative ability was estimated by means <strong>of</strong> bootstrap-corrected<br />

Harrel C statistic. Results: 526 patients were identified. At a median<br />

follow-up <strong>of</strong> 45 months (interquartile range: 22-72 months) 174 deaths<br />

were recorded. 5-yr and 7yr OS (95% confidence interval) were 56.4%<br />

(51.1,62.2%) and 50.1% (44.1,57.0%). The backward selection procedure<br />

by AIC criterion lead to exclude RT from the covariates set. All other<br />

covariates proved to be statistically significant in the final multivariable Cox<br />

model. The Bootstrap-corrected Harrell C statistic was 0.74. Of interest,<br />

size <strong>of</strong> the tumor and age <strong>of</strong> the patient had a bimodal contribution to the<br />

risk <strong>of</strong> death and the combination <strong>of</strong> histological subtype and grade proved<br />

significant. Conclusions: A new multi-institution RPS specific nomogram is<br />

now available for prognostication in the clinic and patient selection in<br />

clinical trials.<br />

10002 Oral Abstract Session, Mon, 3:00 PM-6:00 PM<br />

Phase II trial <strong>of</strong> the CDK4 inhibitor PD0332991 in CDK4-amplified<br />

liposarcoma. Presenting Author: Mark Andrew Dickson, Memorial Sloan-<br />

Kettering Cancer Center, New York, NY<br />

Background: CDK4 is amplified in approximately 90% <strong>of</strong> well-differentiated/<br />

de-differentiated liposarcomas (WD/DDLS). The selective CDK4/CDK6<br />

inhibitor PD0332991 (PD) inhibits growth and induces senescence in<br />

liposarcoma cell lines and xenografts. In a phase I trial <strong>of</strong> PD, several<br />

patients with progressive WD/DDLS had prolonged stable disease for<br />

several years. To determine the safety and efficacy <strong>of</strong> PD, a phase II study<br />

was performed. Methods: <strong>Part</strong>icipants were patients with advanced WD/<br />

DDLS. Eligibility criteria were age�18 years, measurable WD/DDLS<br />

(RECIST 1.1), documented progression on at least one systemic therapy<br />

directly before enrollment, CDK4 amplification by fluorescence in situ<br />

hybridization and retinoblastoma protein (RB) expression by immunohistochemistry<br />

(�1�). Pts received oral PD 200mg daily for 14 consecutive<br />

days in 21-day cycles. The primary endpoint was progression-free survival<br />

(PFS) at 12 weeks. Based on historical data, a promising result was defined<br />

as a 12-week PFS <strong>of</strong> �40% and not promising as �20%. The sample size<br />

was up to 28 evaluable patients. If 9 patients were progression free at 12<br />

weeks, then PD would be considered to have activity in WD/DDLS. Results:<br />

Of 44 patients screened (42/44 CDK4 amplified; 41/44 RB�), 29 were<br />

enrolled and 27 were evaluable for the primary endpoint. Median age was<br />

65 (range 37-83); 52% were male; ECOG scores were 0 (69%) or 1 (31%),<br />

and the median number <strong>of</strong> prior regimens was 1 (range 1-5). PFS at 12<br />

weeks was 70% (19/27 patients; 90% CI 56-100%), and thus the study<br />

significantly exceeded its primary endpoint. At the data cut<strong>of</strong>f, the median<br />

PFS was 18 weeks. Seven patients remain on study with stable disease at<br />

18-48 weeks <strong>of</strong> followup. Grade 3 and 4 events included anemia (grade 3,<br />

14%), thrombocytopenia (grade 3, 17%; grade 4, 14%), neutropenia<br />

(grade 3, 41%; grade 4, 7%) and febrile neutropenia (3%). Dose reductions<br />

were required in 24% <strong>of</strong> patients. Conclusions: Among patients with<br />

WD/DDLS with CDK4 amplification and RB expression who had actively<br />

progressing disease despite prior systemic therapy, treatment with the<br />

CDK4 inhibitor PD0332991 was associated with improved PFS. A randomized<br />

phase 3 trial is planned.<br />

10001 Oral Abstract Session, Mon, 3:00 PM-6:00 PM<br />

How well do we communicate risk? An evaluation <strong>of</strong> AJCC version 6 and 7<br />

staging systems for s<strong>of</strong>t tissue sarcomas. Presenting Author: Robert G.<br />

Maki, Department <strong>of</strong> Medicine, Memorial Sloan-Kettering Cancer Center,<br />

Present address: Departments <strong>of</strong> Medicine, Pediatrics, and Orthopaedics,<br />

Mount Sinai School <strong>of</strong> Medicine, New York, NY<br />

Background: Cancer staging systems allow physicians to communicate risks<br />

<strong>of</strong> a particular clinical situation with one another and with patients. Few<br />

data have been presented to support the update <strong>of</strong> a commonly employed<br />

staging system for s<strong>of</strong>t tissue sarcoma (STS). We examined <strong>American</strong> Joint<br />

Committee on Cancer (AJCC) versions 6 (2002) and 7 (2010) staging<br />

systems for patients with primary STS using a single institution clinically<br />

annotated prospective database. Methods: Subsets <strong>of</strong> the prospectively<br />

collected Memorial Sloan-Kettering Cancer Center STS database <strong>of</strong> 8647<br />

patients from 1982 to 2010 were examined with respect to criteria <strong>of</strong> the<br />

AJCC versions 6 and 7 staging systems. Results: Tumor size does not appear<br />

to be adequately addressed in version 6 or 7. Relapse-free survival was<br />

statistically worse for increasing size <strong>of</strong> primary STS �5, 5-10, 10-15, and<br />

�15 cm; in comparison, overall survival decreased over three size categories<br />

(�5, 5-10, �10 cm). Tumor depth, a statistically significant factor in<br />

patient outcomes that is included in version 6, is functionally omitted in<br />

version 7. Patients with node involvement without other metastases fare<br />

statistically worse than patients with large, high grade tumors without nodal<br />

metastasis, as shown previously. Version 6 and 7 criteria do not address<br />

effects <strong>of</strong> primary anatomic site and histology, even for tumors with same<br />

FNCLCC grade. Sarcoma subtypes defined after publication <strong>of</strong> FNCLCC<br />

criteria are difficult to incorporate into existing guidelines. Conclusions:<br />

Improved prognostication <strong>of</strong> STS outcomes is better achieved by staging<br />

according to anatomic primary site, depth, and a larger number <strong>of</strong> size<br />

categories. Histology-specific nomograms improve prognostic accuracy,<br />

such as those for liposarcoma, GIST, or rhabdomyosarcoma. Staging<br />

accuracy increases at the cost <strong>of</strong> portability and simplicity. The increasing<br />

difficulties with use <strong>of</strong> a single STS staging system highlight the potential<br />

benefit <strong>of</strong> newer techniques, such as patient-specific nomograms or<br />

Bayesian networks. Staging systems will require significant modifications<br />

to incorporate increasingly sophisticated molecular diagnostics.<br />

10003 Oral Abstract Session, Mon, 3:00 PM-6:00 PM<br />

A phase II multicenter study <strong>of</strong> the IGF-1 receptor antibody cixutumumab<br />

(A12) and the mTOR inhibitor temsirolimus (TEM) in patients (pts) with<br />

refractory IGF-1R positive (�) and negative (-) bone and s<strong>of</strong>t tissue<br />

sarcomas (STS). Presenting Author: Gary K. Schwartz, Memorial Sloan-<br />

Kettering Cancer Center, New York, NY<br />

Background: Preclinical studies demonstrate synergistic anti-tumor activity<br />

by inhibiting mTOR and IGF-1R. This study evaluated the safety and<br />

efficacy <strong>of</strong> A12 and TEM in 3 chemo-refractory cohorts: IGF-1R (�) STS<br />

(Arm A), IGF-1R (�) bone (Arm B), and IGF-1R (-) bone and STS (Arm C).<br />

Methods: An optimal Simon 2 stage design was used for each arm. The<br />

primary end-point was progression free survival (PFS) at 12 weeks. Based<br />

on historical data, a 40% PFS rate was considered promising and a 20%<br />

PFS non-promising (type I/ II error, 0.05/0.10). �5 PFSs at 12 weeks were<br />

required in the first 19 and �16 PFSs were required in a total <strong>of</strong> 54 pts to<br />

consider each arm positive. Key eligibility: measurable disease (RECIST<br />

1.1), age �18, 1- 4 priors, ECOG status 0-1. A12 (6 mg/kg) and TEM (25<br />

mg) were administered IV weekly. Pre and post treatment tumor biopsies<br />

and plasma for IGF-1 and IGFBP3 were obtained. 20 Sarcoma Centers<br />

participated. Results: Starting in 02/2010, 383 pts were tested for IGF-1R<br />

by immunohistochemistry (IHC; 54% �) and 171 were treated: mean age<br />

47 (range: 18-80), mean # priors 2.2 (range: 1-4). Grade 3/4 toxicities<br />

�10%: lymphopenia (12%), mucositis (10%). By intent to treat (ITT)<br />

analysis, each arm <strong>of</strong> the study achieved its primary 12 week PFS end-point<br />

(�16 PFSs): Arm A: 18/56 (32%), Arm B: 19/50 (38%) (4 too early), Arm<br />

C: 27/63 (43%). The one-sided 95% CI for 12-week PFS is (0.22, 1),<br />

(0.27, 1), and (0.32, 1), respectively. By ITT the median PFS (95% CI) in<br />

weeks for each arm is 6.3 (5.9, 12.0), 11.0 (8.0, 18.0) and 11.6 (9.0,<br />

17.9), respectively, and for chondro (n�18), Ewing’s (n�24), and osteo<br />

(n�23) only it is 22.6 (5.7, 40.9), 10.4 (5.7, 17.9) and 6.0 (6.0, 18.0),<br />

respectively. Westerns on 32 matched pair tumor biopsies indicate<br />

inhibition <strong>of</strong> IGF-1R, pAKT and pS6. Of biopsies on 7 IGF-1R (-) IHC pts<br />

(Arm C), 5 were IGF-1R (�) by western. Plasma biomarkers did not<br />

correlate with PFS. Conclusions: A12 and TEM met its primary end-point <strong>of</strong><br />

improved PFS in pts with metastatic sarcoma. This effect was independent<br />

<strong>of</strong> IGF-1R status by IHC. These results support further clinical development<br />

<strong>of</strong> this combination in bone and STS.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


10004 Oral Abstract Session, Mon, 3:00 PM-6:00 PM<br />

A first-in-human, phase Ib combination study to assess safety, pharmacokinetics<br />

(PK), and pharmacodynamics (PD) <strong>of</strong> a hedgehog inhibitor, GDC-0449, with a<br />

Notch inhibitor, RO4929097, in patients with advanced sarcoma. Presenting<br />

Author: Mrinal M. Gounder, Memorial Sloan-Kettering Cancer Center, New York,<br />

NY<br />

Background: Aberrant Hedgehog and Notch signaling is seen in sarcoma and<br />

anti-tumor activity is enhanced by inhibiting both pathways. This first in man<br />

Phase Ib study evaluated safety and efficacy <strong>of</strong> the combination <strong>of</strong> GDC-0449<br />

(G), a smoothened inhibitor with RO4929097 (R), a gamma-secretase/notch<br />

inhibitor. Methods: The study evaluated fixed dose G (150 mg qd) for 21 days<br />

followed by G � R at either 10 mg (level 1) or 15 mg (level 2) QD. At MTD pts<br />

were evaluated with G � R (without “lead in” G) or single agent R. Pre and post<br />

treatment tumor biopsies were obtained to assess notch and hedgehog inhibition.<br />

PK was assessed for each drug. Key eligibility: advanced sarcoma, ECOG<br />

� 2, age � 18 and prior therapies � 4. RECIST response was assessed Q6 wks.<br />

Results: 34 pts had a median age <strong>of</strong> 53 yrs (23-78), median priors 3 and various<br />

histologies [liposarcoma (7), chondrosarcoma (7), leiomyosarcoma (4), osteosarcoma<br />

(2), GIST (2) and other (12)]. No DLT was seen. Common (�10 %) grade<br />

�3 toxicity was hypophosphatemia (18%). Systemic exposure (AUC0-24 and<br />

Cmax) <strong>of</strong> G was similar to established studies (not shown). AUC0-24 and Cmax <strong>of</strong> R<br />

was significantly lower (~70%) when administered with G (Table). However,<br />

measurement <strong>of</strong> unbound, free drug (Cfree) <strong>of</strong> R showed comparable levels<br />

between single agent R and G � R, but not when G was a “lead in” (Table).<br />

Target inhibition <strong>of</strong> the notch pathway (cleaved notch) and pAkt was observed in<br />

matched tumor biopsies with both arms. Durable SD was seen in both arms:<br />

myxoid chondrosarcoma (56 wks), liposarcoma (24� wks), clear cell (21� wks),<br />

desmoid (17� wks), spindle cell (15� wks) and chondrosarcoma (13� wks).<br />

Conclusions: The combination <strong>of</strong> G�R is well tolerated and the RP2D is G 150<br />

mg qd and R 15 mg qd without a “lead in” G. Despite reduction in total levels <strong>of</strong><br />

R in the combination, free drug was similar and inhibits target. Disease stability<br />

was seen with R and R � G. Further clinical development <strong>of</strong> notch and hedgehog<br />

inhibitors in sarcomas is warranted.<br />

PK <strong>of</strong> R �Lead in G� then G � R G�R R<br />

AUC0-24 714 3150 4477<br />

Half life hr 21 17.5 28<br />

Tmax hr 2.75 2 2<br />

Cmax ng/mL 87.6 382 535<br />

Trough total ng/mL 28 38 103<br />

Trough free 0.5 0.9 1<br />

10006 Oral Abstract Session, Mon, 3:00 PM-6:00 PM<br />

An open-label international multicentric phase II study <strong>of</strong> nilotinib in<br />

progressive pigmented villo-nodular synovitis (PVNS) not amenable to a<br />

conservative surgical treatment. Presenting Author: Isabelle Ray-Coquard,<br />

GINECO and Centre Leon Berard, Lyon, France<br />

Background: PVNS, also known as tenosynovial giant cell tumor (TGCT), is a<br />

rare pathological entity affecting the synovium in young adults. This<br />

neoplastic disease is driven by a t(1;2) translocation which results in the<br />

fusion <strong>of</strong> COL6A3 and M-CSF genes. Nilotinib has inhibitory properties on<br />

M-CSF receptor pathway which could be <strong>of</strong> therapeutic interest in pts with<br />

non resectable PVNS. Methods: In this open-label International, multicenter,<br />

non-randomized, phase II study the primary objective is to evaluate<br />

the efficacy <strong>of</strong> nilotinib treatment in patients (pts) with progressive or<br />

relapsing PVNS not amenable to surgery or in whom surgery would be<br />

mutilating, as measured by the 12-week progression-free rate (12-w PFR)<br />

validated by an independent committee. Using a Bayesian design with a<br />

stopping rule for futility, interim analyses were planned after the inclusion<br />

<strong>of</strong> 10 pts and then every 5 pts. Results: From December 2010 to November<br />

2011, 33 pts with progressive disease from 17 institutions from Europe<br />

and Australia were enrolled. 14 pts (median age 37 y (range: 19-68), 7<br />

males) were analyzed in the 2nd interim analysis. Median time since<br />

diagnosis was 4.4 years (range: 0.2-26). Primary tumour was located on a<br />

knee for 10 pts (71%) and on hip (1 pt, 7%), finger (7%), foot (7%), hand<br />

(7%), respectively. 69% <strong>of</strong> pts had a inoperable PVNS and 31% had a<br />

resectable tumour but requiring mutilating surgery. All pts started nilotinib<br />

at the planned dose <strong>of</strong> 800 mg/day. At a median follow-up <strong>of</strong> 8.9 months<br />

(range: 0.6-11), 2 pts had a dose reduction and 1 pt had discontinued<br />

nilotinib after 14 days due to toxicity. Nilotinib was well tolerated, with only<br />

2 pts experiencing grade 3 adverse events (anorexia: 1 pt; hepatic failure: 1<br />

pt).The 12-w PFR was 85.7% (95%CI, 57.2%-98.2%), in favour <strong>of</strong> the<br />

study continuation. No OR was observed at w-12; 1 pt (7%) was in PR at<br />

w-30. Updated results will be presented after the 3rd interim analysis.<br />

Conclusions: Nilotinib treatment induces disease stabilisation in a large<br />

proportion <strong>of</strong> PVNS patients with non resectable disease or in whom<br />

resection would be mutilating.<br />

Sarcoma<br />

631s<br />

10005 Oral Abstract Session, Mon, 3:00 PM-6:00 PM<br />

GDC-0449 in patients with advanced chondrosarcomas: A French Sarcoma<br />

Group (FSG)/French and U.S. National Cancer Institutes phase II collaborative<br />

study. Presenting Author: Antoine Italiano, Institut Bergonié, Bordeaux,<br />

France<br />

Background: Chondrosarcomas (CS) exhibit a strong activation <strong>of</strong> hedgehog<br />

(Hh) signaling which plays a crucial role in cartilage tumorigenesis.<br />

Preclinical data show that hedgehog blockade reduces strongly chondrosarcoma<br />

cell proliferation and tumour size. GDC-0449 is a small-molecule<br />

antagonist <strong>of</strong> the Hh pathway. Methods: This is a multicentric single-arm<br />

phase 2 clinical trial based on two-stage Simon’s design which assesses<br />

safety and efficacy <strong>of</strong> GDC-0449 in patients (pts) with advanced CS. All pts<br />

had to have documented progressive disease (PD) as per RECIST 1.1 before<br />

study entry. Pts receive GDC-0449 150mg (oral route), daily until PD or<br />

unacceptable toxicity. The primary endpoint is the 6-month non-PD rate<br />

according to RECIST. Based on the following hypotheses: 20% 6-month<br />

non-PD rate (H0), 40% acceptable 6-month non-PD rate (H1), 10% type I<br />

error rate, 90% power, a total <strong>of</strong> 37 assessable pts are necessary (17 for the<br />

first stage � 20 for the second stage). Following the inclusion <strong>of</strong> the first 17<br />

pts, if � four pts are progression-free at 6 months, the accrual will<br />

continue. In order to account for not assessable pts (�/- 10%), 41 pts will<br />

be recruited. Accrual started in February 2011 in six centers <strong>of</strong> the FSG.<br />

Results: As <strong>of</strong> January 24 2012, 40 pts (28 males, 12 females) have been<br />

included in the study. Median age is 59 years (30-86). The most frequent<br />

histological subtype is conventional CS (n�32). Twenty eight pts (70%)<br />

had grade 1 or 2 adverse events (AE) possibly related to the drug whereas 3<br />

pts (7.5%) had grade 3 or 4 AE. The planned interim statistical analysis<br />

performed after central histological and radiological review showed that<br />

four patients out <strong>of</strong> the first 17 evaluable patients had stable disease<br />

indicating that GDC-0449 had reached the primary endpoint to justify<br />

continuing accrual after the 1st step <strong>of</strong> the study. 15 pts are still on<br />

treatment. Molecular analyses are available for 21 pts. No mutation <strong>of</strong> SMO<br />

was detected. qRT-PCR experiments and PTCH sequencing are ongoing.<br />

Conclusions: GDC-0449 is well tolerated and shows some activity in a<br />

subset <strong>of</strong> pts with advanced CS. Final clinical and molecular data will be<br />

presented at the meeting.<br />

10007 Oral Abstract Session, Mon, 3:00 PM-6:00 PM<br />

Masitinib mesylate in imatinib-resistant advanced GIST: A randomized<br />

phase II trial. Presenting Author: Antoine Adenis, Centre Oscar Lambret,<br />

Lille, France<br />

Background: Masitinib is an oral tyrosine kinase inhibitor with greater in<br />

vitro activity and selectivity than imatinib against wild-type and juxtamembrane<br />

mutations <strong>of</strong> KIT (Dubreuil, 2009, PLoSONE). This trial studied<br />

efficacy and safety <strong>of</strong> masitinib at 12 mg/kg/day and sunitinib at 50 mg/day<br />

for the treatment <strong>of</strong> imatinib-resistant GIST. Methods: Patients with<br />

inoperable, locally advanced or metastatic GIST and showing disease<br />

progression while treated with imatinib 400 mg/day received either<br />

masitinib or sunitinib until progression. Primary endpoint was PFS evaluated<br />

with RECIST with OS as a secondary endpoint. Based upon an 80%<br />

power to detect with a Fleming design a median PFS �3 months (alpha<br />

10%, unilateral), a population <strong>of</strong> 44 patients (22 per arm) was required.<br />

Stratification was applied based on mutation status. Results: 44 patients<br />

(exon 11 [66%], exon 9 [11%], wild-type or other [5%], not done at<br />

baseline [18%]) were randomized into masitinib (n�23) or sunitinib<br />

(n�21) treatment-arms from 9 centers between 02/2009 and 09/2011.<br />

After a median follow-up <strong>of</strong> 14 months, median PFS was 3.9 months<br />

(95%CI [2.6;8.1]) for masitinib vs 3.8 months [1.9;11] for sunitinib. Of<br />

those patients progressing under masitinib, 88% received sunitinib in third<br />

line. Median OS was not reached (NR) (95%CI [21;NR]) for masitinib vs 15<br />

months [9.4;22] for sunitinib. The 18-month and 24-month OS rates for<br />

masitinib vs sunitinib were 79% (95%CI [53;92]) vs 20% [1.5;55], and<br />

53% (95%CI [9.5;84]) vs 0%, respectively. Similar results were seen in<br />

the exon 11 subpopulation with median OS for masitinib NR (95%CI<br />

[NR;NR]) vs 15 months [9.6;22] for sunitinib. The safety pr<strong>of</strong>ile <strong>of</strong><br />

masitinib was better than sunitinib with a lower occurrence <strong>of</strong> severe<br />

related adverse events (AEs) (17% vs 52%) and related AEs leading to<br />

discontinuation (0% vs 9.5%). In masitinib treated patients, nausea,<br />

diarrhea and asthenia were the most common related AEs. Conclusions:<br />

Although PFS curves looked similar, this study apparently showed a longer<br />

survival in the masitinib treatment-arm with a better safety pr<strong>of</strong>ile than<br />

sunitinib. Masitinib may potentially <strong>of</strong>fer patients a new active compound<br />

for advanced GIST in the second-line setting.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


632s Sarcoma<br />

LBA10008 Oral Abstract Session, Mon, 3:00 PM-6:00 PM<br />

Randomized phase III trial <strong>of</strong> regorafenib in patients (pts) with metastatic<br />

and/or unresectable gastrointestinal stromal tumor (GIST) progressing<br />

despite prior treatment with at least imatinib (IM) and sunitinib (SU): GRID<br />

trial. Presenting Author: George D. Demetri, Dana-Farber Cancer Institute,<br />

Boston, MA<br />

The full, final text <strong>of</strong> this abstract will be available at<br />

abstract.asco.org at 12:01 AM (EDT) on Monday, June 4,<br />

2012, and in the <strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong> online<br />

supplement to the June 20, 2012, issue <strong>of</strong> Journal <strong>of</strong><br />

<strong>Clinical</strong> Oncology. Onsite at the <strong>Meeting</strong>, this abstract will<br />

be printed in the Monday edition <strong>of</strong> ASCO Daily News.<br />

10010 Poster Discussion Session (Board #2), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Phase III, placebo-controlled trial (SUCCEED) evaluating ridaforolimus as<br />

maintenance therapy in advanced sarcoma patients following clinical<br />

benefit from prior standard cytotoxic chemotherapy: Long-term (> 24<br />

months) overall survival results. Presenting Author: Jean-Yves Blay, University<br />

Claude Bernard Lyon I, Centre Léon Bérard, Lyon, France<br />

Background: The mammalian target <strong>of</strong> rapamycin (mTOR) regulates cell<br />

growth and proliferation and is abnormally activated in many sarcomas.<br />

Ridaforolimus, an oral mTOR inhibitor, demonstrated clinical activity in<br />

previous nonrandomized trials in advanced sarcomas following failure <strong>of</strong><br />

prior chemotherapy. Methods: An international, multicenter, placebocontrolled,<br />

phase 3 trial was conducted to evaluate maintenance therapy<br />

with ridaforolimus in patients with metastatic s<strong>of</strong>t-tissue or bone sarcomas<br />

who achieved disease control from prior chemotherapy. Patients were<br />

randomized (1:1) to receive oral ridaforolimus (40 mg) or placebo once<br />

daily for 5 days each week. The primary endpoint was progression-free<br />

survival (PFS); secondary endpoints included overall survival (OS) and<br />

safety and tolerability. For OS, patients were to be followed at 3-month<br />

intervals for at least 24 months and up to 60 months after randomization.<br />

Results: 702 <strong>of</strong> 711 randomized patients received treatment. At the time <strong>of</strong><br />

the data cut<strong>of</strong>f for OS (386 deaths), patients in the study population had<br />

been followed for at least 15 months. Median OS was 93.3 weeks with<br />

ridaforolimus vs 83.4 weeks with placebo (hazard ratio [HR]�0.88; 95%<br />

confidence interval [CI]: 0.72, 1.08; P�0.23). Ridaforolimus significantly<br />

improved PFS vs placebo (HR�0.72; 95% CI: 0.61, 0.85; P�0.0001;<br />

median PFS: 17.7 weeks vs 14.6 weeks); PFS improved across all<br />

prespecified baseline characteristics. As expected from the class <strong>of</strong> mTOR<br />

inhibitors, the most common adverse events with ridaforolimus were<br />

stomatitis, thrombocytopenia, noninfectious pneumonitis, hypertriglyceridemia,<br />

hyperglycemia, infections, and rash. Conclusions: Oral ridaforolimus<br />

was generally well-tolerated and significantly improved PFS in<br />

metastatic sarcoma patients with benefit from prior chemotherapy, <strong>of</strong>fering<br />

an effective treatment alternative to surveillance alone. Results <strong>of</strong> a<br />

long-term OS analysis (prespecified to occur at 67% mortality, 24 months<br />

minimum follow-up) in the intent-to-treat population will be available in<br />

early 2012.<br />

10009 Poster Discussion Session (Board #1), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

PALETTE: Final overall survival (OS) data and predictive factors for OS <strong>of</strong><br />

EORTC 62072/GSK VEG110727, a randomized double-blind phase III<br />

trial <strong>of</strong> pazopanib versus placebo in advanced s<strong>of</strong>t tissue sarcoma (STS)<br />

patients. Presenting Author: Winette T.A. Van Der Graaf, Department <strong>of</strong><br />

Medical Oncology, Radboud University Nijmegen Medical Centre, Nijmegen,<br />

Netherlands<br />

Background: At ASCO 2011 we presented an increase in median progressionfree<br />

survival (PFS) <strong>of</strong> pazopanib <strong>of</strong> 13 weeks (median 7 to 20 weeks; HR<br />

0.31) in a randomized double-blind, placebo-controlled phase III trial in<br />

advanced non-adipocytic STS patients pretreated with chemotherapy. Now<br />

we present final OS data and predictive factors for OS, such as ethnicity,<br />

ancestry and well-known prognostic and predictive factors in STS patients<br />

treated with chemotherapy. Methods: At clinical cut-<strong>of</strong>f median follow-up<br />

was 25 months. Comparison <strong>of</strong> both treatments for OS was performed using<br />

a two-sided log rank test stratified for number <strong>of</strong> prior lines <strong>of</strong> systemic<br />

therapy for advanced disease and WHO performance status (PS). HR was<br />

calculated with a 2-sided CI. A Kaplan-Meier OS curve was used. Baseline<br />

characteristics as potential predictive factors for OS were: PS (0 vs 1),<br />

number <strong>of</strong> lines <strong>of</strong> prior therapy for advanced disease (0-1 vs 2�), age<br />

(�55 yrs vs � 55 yrs), gender, ethnicity (Hispano or Latino vs other),<br />

geographic ancestry (White Caucasian, Asian, other), tumour grade (1-2 vs<br />

3), histology (leiomyosarcoma vs synovial sarcoma vs other). A Cox model<br />

was fitted with the investigated factor, treatment arm and associated<br />

interaction factor. Data <strong>of</strong> all 369 randomized patients (123 placebo, 246<br />

pazopanib) were used for the analyses. Results: Median OS (95% CI) was<br />

10.7 (8.7-12.8) in the placebo versus 12.5 (10.6-14.8) months in the<br />

pazopanib group, HR: 0.86 (0.67-1.1). Post study protocol systemic<br />

treatment was administered in 63% <strong>of</strong> placebo, and in 49% <strong>of</strong> pazopanib<br />

treated patients. None <strong>of</strong> the investigated factors showed any statistical<br />

significant predictive value for OS. Conclusions: Although pazopanib<br />

demonstrated a statically significant increase in PFS compared to placebo,<br />

final OS analysis, which showed a trend in favor <strong>of</strong> pazopanib in patients<br />

with metastatic non-adipocytic STS, did not reach statistical significance.<br />

The factors studied were not predictive for OS.<br />

10011^ Poster Discussion Session (Board #3), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Phase I study <strong>of</strong> neoadjuvant chemoradiation (CRT) plus sorafenib (S) for<br />

high-risk extremity s<strong>of</strong>t tissue sarcomas (STS). Presenting Author: Janelle Marie<br />

Meyer, Oregon Health & Science University Knight Cancer Institute, Portland,<br />

OR<br />

Background: We previously reported a regimen <strong>of</strong> epirubicin/ifosfamide (E/I) and<br />

hyp<strong>of</strong>ractionated radiation (RT) followed by surgery and postoperative E/I. We<br />

hypothesize that S � CRT is safe and augments activity through anti-angiogenic<br />

mechanisms. We performed dynamic contrast-enhanced MRI using shutter<br />

speed pharmacokinetic analysis (SSM DCE-MRI) to assess preoperative therapy<br />

effect. Methods: Subjects with �5 cm, grade 2-3 extremity STS were treated in<br />

cohorts <strong>of</strong> escalating S dose using a 3�3 design based on dose-limiting toxicity<br />

(DLT) during the first 8 weeks. Daily S was initiated 14 days prior to E/I (E 30<br />

mg/m2 /day days 1-3 and I 2.5 g/m2 /day days 1-3 every 21 days for 3 pre- and 3<br />

postoperative cycles). 28 Gy preoperative RT was administered in 8 fractions<br />

during cycle 2 (E omitted). Primary objective was to determine maximum<br />

tolerated dose (MTD). DCE-MRI was performed at baseline, after 2 weeks S, and<br />

prior to surgery. Results: 16 subjects were enrolled. Histologies included<br />

pleomorphic (5), synovial (5), liposarcoma (3), other (3). All were extremity (12<br />

lower, 4 upper) tumors. Median tumor size was 11.7 cm. The MTD <strong>of</strong> S was 400<br />

mg daily (see table). Most common grade 3/4 adverse events among all dose<br />

levels were neutropenia (94%), anemia (75%), hypophosphatemia (69%),<br />

thrombocytopenia (56%), neutropenic fever/infection (50%), and hypokalemia<br />

(31%). 38% developed wound complications requiring surgical intervention<br />

including amputation in 1 subject. 44% had �95% histologic necrosis at<br />

surgery. No subjects have recurred with median follow-up <strong>of</strong> 15 (6-27) months.<br />

8 subjects underwent DCE-MRI. Changes in SSM DCE-MRI biomarkers after 2<br />

weeks S correlated with histologic response (R2�0.71). Conclusions: The<br />

addition <strong>of</strong> S to CRT is feasible with promising activity that warrants further<br />

investigation. Rate <strong>of</strong> wound complications is similar to other reports <strong>of</strong><br />

preoperative radiation. SSM DCE-MRI detected changes in tumor perfusion after<br />

only 2 weeks <strong>of</strong> S and may prove useful to assess early drug effect in STS.<br />

S (mg) #DLTs/subjects DLT Grade<br />

200 1/6 Mucositis 3<br />

400 1/7 Syncope 3<br />

800 3/3 -Neutropenic sepsis, thrombosis, HFS<br />

-Rash, HFS<br />

-Rash<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.<br />

4,3,3<br />

3,3<br />

3


10012 Poster Discussion Session (Board #4), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

18f-FDG-PET imaging as an early survival predictor in patients with<br />

high-grade s<strong>of</strong>t tissue sarcomas undergoing neoadjuvant therapy. Presenting<br />

Author: Ken Herrmann, UCLA, Los Angeles, CA<br />

Background: Neoadjuvant therapy is associated with considerable toxicity<br />

and limited survival benefits in patients with s<strong>of</strong>t tissue sarcoma (STS). We<br />

prospectively evaluated whether 18F-FDG PET/CT (PET) imaging after the<br />

initial cycle and after end <strong>of</strong> neoadjuvant therapy could predict overall<br />

survival in these patients. Methods: 76 patients (primary STS: n�57;<br />

metastatic disease: n�19) with high grade STS were included in this study.<br />

PET was performed prior to (n�76), after one cycle (n�52) and after the<br />

end <strong>of</strong> neoadjuvant therapy (n�74). Overall survival was correlated with<br />

changes <strong>of</strong> SUVpeak, RECIST, histopathological response and other<br />

parameters predictive <strong>of</strong> STS survival. Results: One-, two- and five- year<br />

survival rates were 95�3.0%, 86�4.6% and 68�6.6% for primary STS.<br />

Corresponding one- and two- year survival rates for recurrent/metastatic<br />

STS were 77�10.0% and 47�12.1%. Optimal cut-<strong>of</strong>f for early decreases<br />

in SUV peak were significant predictors <strong>of</strong> survival in log-rank test<br />

(p�0.027 and p�0.043). However, late decreases in SUV peak were only<br />

predictive in primary STS (SUV peak decrease 57%; p�0.035) but not in<br />

recurrent/metastatic STS (SUV peak decrease 52%; p�0.057). In primary<br />

STS, 7/15 early PET non-responders but only 4/24 early PET responders<br />

died during follow up (p�0.068). Conclusions: 18F-FDG-PET seems feasible<br />

to predict survival after the initial cycle <strong>of</strong> neoadjuvant chemotherapy<br />

in both patients with primary STS and recurrent/metastatic STS and can<br />

potentially serve as an intermediate endpoint biomarker in clinical research<br />

and patient care.<br />

10014 Poster Discussion Session (Board #6), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Growth modulation index and RECIST-based STBSG-EORTC criteria for the<br />

assessment <strong>of</strong> drug activity in advanced s<strong>of</strong>t tissue sarcoma (ASTS)<br />

patients (pts). Presenting Author: Sophie Cousin, Centre Oscar Lambret,<br />

Lille, France<br />

Background: Despite promising phase II trial results, most <strong>of</strong> new drugs<br />

failed to improve the overall survival (OS) in ASTS pts. The choice <strong>of</strong> the<br />

endpoint in early trials remains crucial. We have evaluated the Growth<br />

Modulation Index (GMI) as potential endpoint. The GMI is defined as the<br />

Time To Progression with the second (or n�1) line <strong>of</strong> chemotherapy (TTP2)<br />

divided by the TTP with the first (or n) line (TTP1). Methods: We have<br />

carried out a retrospective multicenter study in pts receiving second-line<br />

chemotherapy after failure/intolerance to doxorubicin-based regimens.<br />

Data collected included best response(s), TTP1 & TTP2 and OS. Treatments<br />

have been classified as �active� treatment according to the EORTC-<br />

STBSG criteria (3-month Progression-free rate �40% or 6-month<br />

PFR�14%: including trabectedin, ifosfamide, gemcitabine�docetaxel for<br />

all subtypes and weekly paclitaxel for angiosarcoma) versus non-active<br />

drugs. Comparisons used chi-2 tests and Log-rank tests. We performed a<br />

logistic regression analysis for identifying factors associated with longer<br />

GMI. Results: The study population consisted in 106 men and 121 women,<br />

the median age was 57 years. 110 pts (48.4%) have received �active<br />

drugs�. The median TTP1, TTP2 and GMI were 197 days, 134 days and<br />

0.75, respectively. The median OS was 446 days. 70 pts experienced<br />

GMI�1.33 (30.6%). There was a strong relation between best objective<br />

response and GMI (p�0.0001). The treatment with �active drug� was not<br />

associated with an improvement <strong>of</strong> the OS: 490 days 407 versus days<br />

(p�0.524). The median OS <strong>of</strong> pts with GMI�1, GMI�[1.00-1.33] and<br />

GMI�1.33 were 324, 302 and 710 days, respectively (p�0.0001). None<br />

<strong>of</strong> the following factors were associated with GMI�1.33 in logistic<br />

regression analysis: age, gender, histological subtypes, grade, metastasis<br />

locations, interval between diagnosis & metastasis, association with ifosfamide<br />

or dacarbazine in 1st-line regimen or treatment with �active drug� in<br />

second-line. Conclusions: GMI seems to be an interesting endpoint providing<br />

additional information compared to classical criteria. GMI�1.33 is<br />

associated with significant improvement <strong>of</strong> the OS.<br />

Sarcoma<br />

633s<br />

10013 Poster Discussion Session (Board #5), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Growth modulation index (GMI) as a metric <strong>of</strong> clinical benefit assessment<br />

among advanced s<strong>of</strong>t tissue sarcoma (ASTS) patients receiving trabectedin<br />

as salvage therapy. Presenting Author: Nicolas Penel, Centre Oscar<br />

Lambret, Lille, France<br />

Background: Von H<strong>of</strong>f has proposed GMI �1.33 in cancer clinical trials as a<br />

sign <strong>of</strong> drug activity. GMI is the ratio between time to progression (TTP)<br />

with the nth line (TTPn) <strong>of</strong> therapy divided by the TTPn-1 with the n-1th line<br />

<strong>of</strong> therapy. With this endpoint, each patient is his/her own control. Methods:<br />

We have carried out a retrospective analysis <strong>of</strong> 279 pretreated ASTS<br />

patients treated in four consecutive phase II trials with trabectedin 1.5<br />

mg/m², given as a 24-hour infusion every 3 weeks. Results: The study<br />

population consisted <strong>of</strong> 170 women and 109 men with a median age <strong>of</strong> 52.<br />

The two most common histologies were leiomyosarcoma (145 patients,<br />

52%) and liposarcoma (59, 21%). 142 (51%) patients received one prior<br />

line and 137 (49%) �2 lines. The median TTPn was 2.8 months (range:<br />

0.2-26.8), whereas the median TTPn-1 was 4.0 months (range: 0.3-79.5).<br />

The Spearman correlation coefficient between TTPn-1 and TTPn was 0.07<br />

(p�0.23). The median GMI was 0.6 (range: 0.0-14.4). 177 patients<br />

(63%) had a GMI �1, 21 (8%) a GMI � 1-1.33 and 81 (29%) a GMI<br />

�1.33. The median overall survival (OS) strongly correlated with GMI<br />

groups: OS was 9.1, 13.9 and 23.8 months, respectively (p�0.0005,<br />

log-rank test). There were also a strong correlations between response rate<br />

and GMI (p�0.0001, Fisher exact test), and between GMI and progressionfree<br />

survival (�0.0001, log-rank test). The logistic regression analysis<br />

retained only performance status (PS) [OR�1.76 if PS�0; p�0.04]<br />

associated with GMI�1.33. Sarcoma grade (p�0.21), histological subtype<br />

(p�0.16), and number <strong>of</strong> prior chemotherapy lines (p�0.95) were not<br />

associated with GMI�1.33. Conclusions: Overall,�30% <strong>of</strong> patients experienced<br />

GMI�1.33 regardless <strong>of</strong> histological subtype or grade, and number<br />

<strong>of</strong> prior lines <strong>of</strong> chemotherapy. Patients with PS�0 benefit more from<br />

trabectedin. GMI�1.33 is associated with longer OS (median �24<br />

months). GMI is better defined than previously described parameter such<br />

as �Tumor Growth Rate� (Lopez-Martin, Abstract 3293, ASCO 2003). GMI<br />

as an indicator <strong>of</strong> drug activity merits further exploration in this setting.<br />

10015 Poster Discussion Session (Board #7), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

A prognostic nomogram for prediction <strong>of</strong> recurrence following surgical resection<br />

<strong>of</strong> desmoid tumors. Presenting Author: Aimee Marie Crago, Department <strong>of</strong><br />

Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY<br />

Background: Desmoid tumors can respond to novel chemotherapeutics (e.g.,<br />

sorafenib). We sought to construct a postoperative nomogram identifying<br />

desmoid patients who are at high-risk for local recurrence and potential<br />

candidates for systemic therapy. Methods: Desmoid patients undergoing resection<br />

from 1982-2011 were identified from a single-institution prospective<br />

database. Cox regression analysis was used to create a desmoid-specific<br />

recurrence nomogram integrating clinical risk factors. Results: Desmoids were<br />

treated surgically in 495 patients (median follow-up 60 months). Of 439<br />

patients undergoing complete gross resection, 100 recurred (92 within 5 years<br />

<strong>of</strong> operation). Five-year recurrence-free survival (RFS) was 71%. Only 8 patients<br />

died <strong>of</strong> disease, all after R2 resection (6 with intraabdominal desmoids).<br />

Radiation was associated with worse RFS (p�0.001). Multivariate analysis<br />

suggested associations between recurrence and extremity location, young age,<br />

and large tumors, but not margin (Table). Abdominal wall tumors had the best<br />

outcome (5-year RFS 92% vs. 34% in patients �25y.o. with large, extremity<br />

tumors). Age, site and size were used to construct an internally-validated<br />

nomogram (concordance index 0.703). Integration <strong>of</strong> margin, gender, depth,<br />

and presentation status (primary vs. recurrent disease) did not improve concordance<br />

significantly (0.707). Conclusions: A postoperative nomogram including<br />

only size, site and age predicts local recurrence and aids in counseling patients.<br />

Systemic therapies may be tested in young patients with large, extremity<br />

desmoids, but surgery alone is curative for most abdominal wall lesions.<br />

Multivariate analysis<br />

factor Hazard ratio (95% C.I.) P value<br />

Margin status (R1 vs. R0) 0.99 (0.65, 1.52) 0.97<br />

Presentation (recurrent vs. primary) 1.16 (0.70, 1.90) 0.57<br />

Depth (deep vs. superficial) 1.37 (0.54, 3.45) 0.51<br />

Gender (female vs. male) 1.32 (0.82, 2.10) 0.25<br />

Site (vs. abdominal wall)<br />

Extremity 5.02 (2.14, 8.42) �0.001<br />

Chest wall 3.12 (1.20, 8.42) 0.020<br />

Intraabdominal 2.73 (0.98, 7.59) 0.054<br />

Other 1.54 (0.31, 7.63) 0.60<br />

Size (>10 cm vs. 65y.o.)<br />


634s Sarcoma<br />

10016 Poster Discussion Session (Board #8), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Is CTNNB1 mutational analysis predictive for progression in patients with<br />

sporadic desmoid tumors primarily observed? Presenting Author: Chiara<br />

Colombo, Fondazione IRCCS Istituto Nazionale Tumori, Milano, Italy<br />

Background: Recently, a wait and see (W&S) approach has been proposed<br />

for patients affected by desmoid tumor (DT). Specific CTNNB1 mutation<br />

has been correlated with higher risk <strong>of</strong> recurrence after surgical resection.<br />

Aim <strong>of</strong> this study was to evaluate the correlation between CTNNB1<br />

mutation type and time to progression in patients primarily observed (TTP).<br />

Methods: We included all consecutive patients:(1) primarily observed at the<br />

Fondazione IRCSS Istituto Nazionale Tumori (Milano, Italy) or Institut<br />

Gustave Roussy (Paris, France) for primary sporadic DTs (2) with available<br />

FFPE preserved tissue for CTNNB1 mutational analysis, (3) with measurable<br />

disease and adequate follow-up. TTP from date <strong>of</strong> diagnosis to date <strong>of</strong><br />

radiological (PRO-R) or symptomatic (PRO-S) progression were conducted<br />

by Kaplan-Meier method and log-rank test to compare strata. Results: A<br />

total <strong>of</strong> 79 patients (August 2002- July 2011) were included (81% female,<br />

19% male); median age was 34 (IQ, 10-77); sites distribution: abdominal/<br />

chest wall (75%), extremity (21%), intra-abdominal (4%). CTNNB1<br />

mutations were observed in 76% <strong>of</strong> DT samples: 41A (48%), 45F (25%),<br />

45P (2%); 24% were WT. Median follow-up was 19 mo (IQ, 11-28).<br />

Thirty-six patients experienced progression (86% PRO-R, 14% PRO-S):<br />

41A (47%), 45F (55%), 45P (100%), WT (26%). An inferior TTP at 36<br />

months was observed in mutated patients vs WT, while no difference was<br />

detected for specific subtype mutated patients (WT 68%, 45F 24%,<br />

41/45P 35%, p� 0.045). A variety <strong>of</strong> different treatments including<br />

surgery, hormonal therapy, chemotherapy, antiCOX2 or persistent W&S<br />

approach were proposed at progression. Forty-two patients did not receive<br />

any treatment. Conclusions: A clear trend towards a lower risk <strong>of</strong> progression<br />

was observed in WT patients, but no difference between specific mutated<br />

patients (45F vs 41/45P) was observed. Prospective studies to eventually<br />

clarify the potential role <strong>of</strong> CTNNB1 as prognostic tool in tailoring desmoids<br />

treatment are presently underway.<br />

10018 Poster Discussion Session (Board #10), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Localized angiosarcomas (AS): Identification <strong>of</strong> prognostic factors (PF) and<br />

analysis <strong>of</strong> treatment impact—Series from the French Sarcoma Group<br />

(GSF/GETO). Presenting Author: Clothilde Lindet, Centre Oscar Lambret,<br />

Lille, France<br />

Background: AS represents less than 2% <strong>of</strong> all adult s<strong>of</strong>t tissue sarcoma. PF<br />

and role <strong>of</strong> respective (neo)-adjuvant treatments in management <strong>of</strong> localized<br />

AS remain badly established. Methods: We have conducted a retrospective<br />

multicentre study (June 1980-October 2009) <strong>of</strong> 107 patients with<br />

localized AS. All cases have been centrally reviewed by pathologist expert.<br />

Univariate and multivariate analysis were conducted to identify independent<br />

PF. Overall survival (OS) and Local Recurrence Free Survival (LRFS)<br />

were estimated using the Kaplan-Meier method. Impact <strong>of</strong> treatments had<br />

been explored with Cox Model after adjustment to the PF. Results: The sex<br />

ratio was 18/89 (0.2). Median age was 71 years. 22% (24/107) and 62%<br />

(67/107) <strong>of</strong> patients developed AS in preexisting lymphoedema and in<br />

irradiated field, respectively. 71 cases (66%) were superficial. The most<br />

frequent primary locations were: limbs (35, 33%), chest wall (32, 29%)<br />

and breast (22, 20%). Grade was 1 in 21/103 (20%), 2 in 35/103 (34%)<br />

and 3 in 47/103 cases (46%). Treatments consisted in surgery, adjuvant<br />

radiotherapy and (neo)-adjuvant chemotherapy in, respectively, 95, 27 and<br />

27 cases. R0 margin was obtained in 49 cases. The median OS and LRFS<br />

were, respectively, 38.8 months and 27 months. In multivariate analysis,<br />

the following PF influenced the OS: lymphedema (HR�2.02 [1.15-3.55])<br />

and size�5cm(HR�1.48 [1.01-2.45]). After adjustment to these PF, R0<br />

margins was the sole treatment parameter improving the OS (HR�0.19<br />

[0.05-0.73]). In multivariate analysis, the following PF influence the<br />

LRFS: Age�70 (HR�1.68 [1.02-2.76]) and pre-existing lymphedema<br />

(HR�2.07 [1.15-3.75]). After adjustment to these prognostic factors, R0<br />

margins (HR�0.48 [0.38-0.69]) and adjuvant radiotherapy (HR�0.29<br />

[0.10-0.83]) improved the LRFS. Conclusions: Pre-existing lymphedema,<br />

tumour size and age�70 are the major PF in patients with localized AS.<br />

The achievement <strong>of</strong> R0 margins is <strong>of</strong> major importance for improving the<br />

outcome whatever the endpoint (OS, LRFS) Adjuvant radiotherapy decreased<br />

the local recurrence. (Neo)-adjuvant chemotherapy does not<br />

influence survivals.<br />

10017 Poster Discussion Session (Board #9), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Desmoid fibromatosis and pregnancy: A multi-institutional analysis <strong>of</strong> recurrence<br />

and obstetric risk. Presenting Author: Marco Fiore, Fondazione IRCCS<br />

Istituto Nazionale dei Tumori, Milan, Italy<br />

Background: Desmoid fibromatosis (DF) may be diagnosed during or after<br />

pregnancy (P). However, the risk <strong>of</strong> progression during P, or in women <strong>of</strong><br />

child-bearing age with DF prior to P, is unknown. Furthermore, obstetric risks<br />

have not been well described. Methods: Institutional databases were reviewed at<br />

3 sarcoma referral centers in Europe and US for women with sporadic DF from<br />

1985 to 2011. Pregnancy and treatment data, outcomes, and obstetric<br />

complications were recorded. Results: Overall 75 women were identified. DF was<br />

diagnosed during P in 17 women (Group A) or within 6 mo after P in 10 (Group<br />

B), was in situ at the time <strong>of</strong> P in 29 women (Group C), or had been resected prior<br />

to P in 19 (Group D). Anatomic site, outcomes, and treatment for each group are<br />

in the Table. Among patients operated at diagnosis, 2/11 (18%) recurred (Group<br />

A�B). Among the entire cohort, 15 women (20%) recurred after definitive<br />

treatment and only 6 (8%) needed multiple treatments after P. Ten spontaneous<br />

regressions occurred after P (13%). Twelve women had further P following the<br />

DF-related one, and 3 (25%) needed treatment after the subsequent P. At a<br />

median follow up <strong>of</strong> 35 mo from P, 17 women did not receive any treatment<br />

(23%), and 39 remain disease-free (52%). Caesarean section was needed in 14<br />

cases (19%), but only in 1 expressly due to DF. DF-related P was associated with<br />

abortion in 6 cases (4 spontaneous, 2 voluntary); in no case was it caused by the<br />

presence <strong>of</strong> DF. Conclusions: DF developing prior to or during P may progress<br />

during the course <strong>of</strong> P or thereafter. Spontaneous regression after P was also<br />

observed. When resected, P-related DF rarely recurs. Wait & see is an option as<br />

well. DF history is not an indication for therapeutic abortion nor a contraindication<br />

against subsequent P.<br />

A B C D<br />

Total number 17 10 29 19<br />

Abdominal wall 11 (64%) 6 (60%) 14 (48%) 14 (73%)<br />

Limbs 1 (6%) 2 (20%) 11 (38%) 2 (11%)<br />

Visceral 4 (24%) 2 (20%) 1 (4%) 2 (11%)<br />

Other 1 (6%) 0 3 (10%) 1 (5%)<br />

Primary / recurrent 17/0 10/0 19/10 17/2<br />

DF progression during or after P 12 (70%) - 16 (55%) 4 (21%)<br />

Treatment after progression 9 (53%) - 8 (28%) 3 (16%)<br />

Surgery (*1 ILP) 5 - 6* 2<br />

Medical therapy 4 - 2 1<br />

DF progression after definitive treatment 3 (18%) 1 (10%) 8 (28%) 3 (16%)<br />

Spontaneous regression 1 (5%) 1 (10%) 7 (24%) 1 (5%)<br />

10019 Poster Discussion Session (Board #11), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Activity <strong>of</strong> sorafenib in radiation-associated breast angiosarcomas harboring<br />

MYC and FLT4 amplifications. Presenting Author: Sandra P. D’Angelo,<br />

Memorial Sloan-Kettering Cancer Center, New York, NY<br />

Background: Angiosarcomas (ASs) are rare, aggressive vascular malignancies<br />

<strong>of</strong> endothelial cell differentiation which arise de novo or secondary to<br />

radiation therapy (RAS.) A subset <strong>of</strong> patients(pts) with AS harbor mutations<br />

in KDR (VEGFR-2) or overexpression <strong>of</strong> MYC and/or FLT4(VEGFR-3). MYC<br />

& FLT4 gene amplifications occur almost exclusively in RAS. These<br />

alterations provide a rationale for investigating agents that target angiogenesis<br />

in RAS. In a phase II trial <strong>of</strong> sorafenib, AS pts had a partial response<br />

(PR) <strong>of</strong> 14%; targeting the appropriate pt population may be essential. We<br />

hypothesize that co-amplification <strong>of</strong> MYC & FLT4 may be predictive <strong>of</strong><br />

response to sorafenib. Methods: Using our institutional sarcoma database &<br />

data query system, we identified AS patients treated with sorafenib at<br />

Memorial Sloan-Kettering Cancer Center between 1992-2011. Molecular<br />

analysis performed on available tissue. With IRB approval, clinical information<br />

was collected. Results: We identified 10 women with RAS that received<br />

sorafenib. Average age 68 (range 51-84.) 7 pts had distant metastatic<br />

disease. Median lines <strong>of</strong> systemic therapy prior to sorafenib: 2 (range 1-4.)<br />

Sorafenib was first line in 60% <strong>of</strong> pts, administered at 400mg daily and<br />

adjusted for toxicity. Best responses included: complete response (CR)<br />

2/9(22%), PR 1/9(11%), stable disease (SD) 5/9(56%) range 5-23m,<br />

progressive disease 1/9(11%) for a clinical benefit rate <strong>of</strong> (CR�PR�SD)<br />

89%. Responses were seen in patients with lung metastases (n�2) and<br />

locally advanced disease (n�1) and were durable for 17, 42 and 26<br />

months. Median duration on therapy was 15 months (range 0-42 months.)<br />

7 pts underwent molecular analysis: co-amplification <strong>of</strong> MYC & FLT4 3 pts<br />

(30%); MYC amplification 4 pts (40%); KDR mutation 0 patients. Of the 3<br />

responders, MYC & FLT4 co-amplification were observed in 2 patients and<br />

not evaluated in one. Conclusions: Sorafenib is active against RAS <strong>of</strong> the<br />

breast. In this small series, complete and partial responses were seen in<br />

patients with co-amplification <strong>of</strong> MYC & FLT4. This observation requires<br />

further study. Performing molecular studies on all AS pts will help define<br />

the pathophysiology <strong>of</strong> this deadly disease to guide rationale clinical trials.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


10020 Poster Discussion Session (Board #12), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Phase II study <strong>of</strong> sorafenib mesylate (So) in patients (pts) with evolutive<br />

and advanced epithelioid hemangioendothelioma (EHE) or hemangiopericytoma/solitary<br />

fibrous tumor (SFT). Presenting Author: Christine Chevreau,<br />

Institut Claudius Regaud, Toulouse, France<br />

Background: There is no standard <strong>of</strong> care for both rare sarcomas. Regarding,<br />

the important vascularization <strong>of</strong> EHE and SFT, we explored the activity/<br />

toxicity <strong>of</strong> So in pts with these sarcomas. Methods: We conducted a<br />

multicenter one-step phase II trial <strong>of</strong> So (800 mg/d). The primary endpoint<br />

was the 9-months progression-free rate (9-PFR). According to EORTC<br />

criteria, So is considered as promising drug if 6-PFR�14%. All pts have<br />

had documented progressive disease at entry. Results: 20 pts (15 EHE & 5<br />

SFT) were enrolled from June 2009 to February 2011 in the 8 participating<br />

institutions. 12 men and 8 women. Median age was 57. The most common<br />

primaries were superficial trunk (8 cases) and liver (4 cases). PS were 0 in<br />

10 cases, 1 in 7 cases and 2 in 3 cases. 16 pts had metastasic disease ,<br />

especially in lung (15), liver (6) and bone (4). Eight pts received prior<br />

chemotherapy (Doxorubicin : n� 8 cases and taxane : n �3). The median<br />

So treatment duration was 124 days. 9 pts experienced grade-3 toxicities;<br />

the most frequent grade-3 toxic events were hand foot syndrome (5 pts),<br />

myalgia (1), stomatitis (1), anorexia (1), diarrhea (1) and arterial hypertension<br />

(1).Because <strong>of</strong> this toxicity, treatment discontinuation was necessary<br />

in 6 cases and dose reduction was necessary in 5 pts. The 9-PFR was 6/18<br />

(33.3% [11.5-55.1]). The 2, 4 and 6 PFR were 15/18 (83.3%), 8/18<br />

(44.4%) and 7/18 (38.8) respectively We observed 2 partial responses<br />

lasting 2 and 9 months in 2 pts with EHE. Analysis <strong>of</strong> the predictive value <strong>of</strong><br />

circulating pre-angiogenetic biomarkers is ongoing. Conclusions: According<br />

to the STBSG-EORTC criteria (3-PFR�40% & 6-PFR�14%), So is a<br />

promising drug for EHE and SFT pts. Further trials is needed, especially a<br />

discontinuation randomized trial.<br />

10022 Poster Discussion Session (Board #14), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Tenosynovial giant cell tumor (TGCT)/pigmented villonodular synovitis<br />

(PVNS): Outcome <strong>of</strong> 313 patients before the era <strong>of</strong> kinase inhibitors.<br />

Presenting Author: Emanuela Palmerini, Istituto Ortopedico Rizzoli, Bologna,<br />

Italy<br />

Background: Tenosynovial giant cell tumor (TGCT) is a rare, usually benign<br />

neoplasm <strong>of</strong> synovium and tendon sheath. TGCT is classified as localized or<br />

diffuse according to the extent <strong>of</strong> synovial involvement. Surgery is the<br />

primary treatment, but the recurrence rate is high, with possible multiple<br />

recurrences, joint function deterioration and decline in quality <strong>of</strong> life.<br />

Recent data suggest a role for TKIs in advanced disease. In order to identify<br />

prognostic factors for recurrence, a retrospective pooled analysis was<br />

carried out in three institutions (Istituto Ortopedico Rizzoli, Bologna, Italy;<br />

Istituto Nazionale Tumori, Milano, Italy; Memorial Sloan Kettering Cancer<br />

Center, New York, USA). Methods: <strong>Clinical</strong> charts and pathology reports <strong>of</strong><br />

patients (pts) treated in the period 1998-2008 were examined. Results:<br />

The study included 313 pts, 177 F and 136 M; median age: 36 years<br />

(range: 11-89 years). Most (64%) pts had tumors in the knee (15% ankle,<br />

11% hip, 10% other). Tumor size was: �2 cm in 24% <strong>of</strong> pts, 2-5 cm in<br />

44%, �5 cm in 32%. A diffuse pattern was reported in 69% <strong>of</strong> pts. The<br />

resection status was available in 289 pts: 51% had R0 surgery, 28% R1<br />

and 21% R2. No metastases were documented. Local recurrence was<br />

reported in 76 pts (median time to recurrence: 15.7 months). With a<br />

median follow-up <strong>of</strong> 4.2 years, 5-year local recurrence-free survival (LRFS)<br />

was 66% (95% CI: 59 - 73). Size (� 2 cm 80% vs. 2-5 cm 67% vs. �5cm<br />

62%, p�0.04), gender (F 73% vs. M 56%, p�0.02), type (localized 78%<br />

vs. diffuse 61%, p�0.02), and resection status (R0 76% vs. R1 55%, vs.<br />

R2 57%, p�0.002) influenced 5-year LRFS, whereas age, tumor location<br />

and bone involvement did not. The 5-year 2nd LRFS was 43% (95% CI: 28 -<br />

59). Multiple (2 to 5) local recurrences were observed in 39% <strong>of</strong> relapsed<br />

patients. Conclusions: The study confirms TGCT propensity to multiple local<br />

recurrences. Diffuse type, suboptimal surgery, male gender and larger<br />

tumors increase the recurrence risk. In order to improve the probability <strong>of</strong><br />

local control, studies addressing the role <strong>of</strong> TKIs could be considered in<br />

subsets <strong>of</strong> patients.<br />

Sarcoma<br />

635s<br />

10021 Poster Discussion Session (Board #13), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Optimizing the therapy <strong>of</strong> desmoplastic small round cell tumor: Combined<br />

experience from the two major cancer centers. Presenting Author: Vivek<br />

Subbiah, Division <strong>of</strong> Cancer Medicine, University <strong>of</strong> Texas M. D. Anderson<br />

Cancer Center, Houston, TX<br />

Background: Desmoplastic small round cell tumor (DSRCT) is a rare<br />

translocation-positive (EWS-WT1)sarcoma subtype that <strong>of</strong>ten presents as<br />

diffuse peritoneal sarcomatosis in male adolescents and young adults. The<br />

objective <strong>of</strong> this study was to assess the survival <strong>of</strong> patients with DSRCT<br />

from the two major cancer centers. Methods: A multi-center retrospective<br />

review was performed <strong>of</strong> patients diagnosed with DSRCT treated at The<br />

University <strong>of</strong> Texas MD Anderson Cancer Center (MDACC) and Memorial<br />

Sloan-Kettering Cancer Center (MSKCC). Results: Of the 197 pts evaluated<br />

at both cancer centers (109 from MSKCC and 88 from MDACC) 87 % were<br />

male. The mean age was 25 � 11 years; 139 (70.6%) underwent surgery;<br />

38 ( 19.6%) had debulking surgery, 30 received radiation therapy, and 27<br />

underwent hyperthermic chemotherapy after debulking (CHPP), 11 had a<br />

stem cell transplant. 112 patients were seen after 2003 in both the centers<br />

vs 85 before 2003. Neoadjuvant chemotherapy included Ewing’s-like<br />

chemotherapy with MSKCC’s P6 regimen (�18 yrs <strong>of</strong> age) and VAI in<br />

adults. We further analyzed the MDACC cohort. In univariate analysis<br />

radiotherapy, surgery, CHPP, removal <strong>of</strong> primary mass � metastases, age �<br />

30 and patients treated after 2003 were associated with significantly<br />

improved overall survival. Multi-variate analysis showed that patients<br />

treated after 2003 had 59% lesser hazard (HR�0.41) than those treated<br />

earlier (p�0.04). Comparing 1990-2003 vs. 2004-2010 survival �3 yr<br />

with DSRCT was �25 % and is now �50 %, a significant improvement.<br />

Conclusions: We present long-term outcomes for the largest series <strong>of</strong><br />

patients diagnosed with DSRCT. A multidisciplinary approach to DSRCT<br />

treatment is required to prolong 3- and 5-yr overall survival. Our recent<br />

improved outcomes required not only aggressive chemotherapy but also<br />

regional local control measures. Best adjuvant therapy remains to be<br />

determined. A treatment algorithm will be proposed.<br />

10023 Poster Discussion Session (Board #15), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Advanced chondrosarcomas: Role <strong>of</strong> chemotherapy and survival. Presenting<br />

Author: Binh Bui Nguyen, Institut Bergonie, Bordeaux, France<br />

Background: There are no data about the role <strong>of</strong> chemotherapy in patients<br />

with advanced chondrosarcomas. Methods: From 2000 to 2011, 98<br />

patients with a confirmed diagnosis <strong>of</strong> advanced chondrosarcomas were<br />

referred to one <strong>of</strong> the 8 participating institutions, and their medical records<br />

reviewed. Results: Median age was 46 years (range 16-82). The most<br />

frequent histological subtype was conventional chondrosarcoma (n�60,<br />

61%). 83 patients (85%) had metastatic disease ( lung n�71, bone n�23,<br />

liver n�6) and 15 patients (15%) had locoregional unresectable disease.<br />

30 patients (31%) had � 2 metastatic sites. 63 patients (64%) received<br />

1st-line combination agent chemotherapy. 70 patients (71%) received a<br />

1st-line anthracycline-containing regimen (doxorubicin � cisplatin �/ifosfamide:<br />

n�30, doxorubicin or pegylated liposomal doxorubicin: n�18,<br />

doxorubicin � ifosfamide �/- dacarbazine: n�16, others� 6). RECIST<br />

objective response was observed in 14 patients (14%), all but two treated<br />

with anthracyclines. 30 patients (31%) had stable disease � 6 months.<br />

Median progression-free survival (PFS) and overall survival (OS) were 5.3<br />

months (95% CI: 2.8-7.7) and 19 months (95% CI: 14-24) respectively.<br />

Performance status (PS) � 2 was the sole factor significantly associated<br />

with OS. Conclusions: Chemotherapy is associated with a 6-month nonprogression<br />

rate <strong>of</strong> about 45% in patients with advanced chondrosarcoma.<br />

Best supportive care should be considered in patients with poor PS. These<br />

data should be used as a reference for response and outcome in the<br />

assessment <strong>of</strong> investigational drugs in advanced chondrosarcoma.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


636s Sarcoma<br />

10024 Poster Discussion Session (Board #16), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Cetuximab and EGFR inhibitors in patients with neurological symptoms<br />

due to chordoma. Presenting Author: Ola Linden, Department <strong>of</strong> Oncology,<br />

Lund University Hospital, Lund, Sweden<br />

Background: Chordomas are rare tumours located along the neuraxis,<br />

showing limited sensitivity to radiotherapy and lack <strong>of</strong> response to chemotherapy.<br />

However, tyrosine kinase inhibitors (TKIs) have been reported to<br />

have an effect. Following the failure <strong>of</strong> single-agent treatment in a young<br />

patient using cetuximab, we decided to treat patients (pts) with neurological<br />

symptoms with dual targeting <strong>of</strong> the epidermal growth factor receptor<br />

(EGFR), based on a case report by H<strong>of</strong> et al., who successfully used dual<br />

targeting <strong>of</strong> EGFR, and supporting in vitro data. Methods: Eight consecutive<br />

pts with neurological symptoms at referral were treated with cetuximab<br />

(250 mg/m2 weekly) and an EGFR signalling inhibitor (250 mg gefitinib or<br />

150 mg erlotinib daily). The duration <strong>of</strong> treatment was not decided in<br />

advance. Results: EGFR expression on tumour cells was variable but<br />

present in all pts. Six pts were evaluable. One tetraplaegic pt died <strong>of</strong> an<br />

unknown cause and one pt on lithium carbonate discontinued treatment<br />

before the first follow-up due to renal toxicity. Four pts showed neurological<br />

improvement. Two <strong>of</strong> these regained and one improved the ability to walk,<br />

and a fourth, with a sacral tumour, improved his voiding ability. The fifth pt<br />

had clinically stable disease and, although the PET scan showed improvement,<br />

the treatment had to be discontinued due to skin toxicity. One pt<br />

showed progressive disease. In the four pts responding neurologically,<br />

improvement was noted already by the end <strong>of</strong> the third week. Three pts were<br />

retreated without failure while on treatment. The first pt who regained the<br />

ability to walk is still walking at 52� months. This pt, while still responsive<br />

to cetuximab and erlotinib, has failed to respond to single-agent treatment<br />

with imatinib, sorafenib or everolimus. The second pt who regained walking<br />

ability had severe alcoholism and died while being treated. The third is still<br />

walking at 16� months. All pts exhibited significant cutaneous side<br />

effects, in some cases necessitating intermittent interruption <strong>of</strong> therapy.<br />

Conclusions: Treatment with cetuximab and TKIs can thus achieve meaningful<br />

and long-lasting clinical improvement in pts with paresis due to<br />

chordoma, and is feasible in pts with limited comorbidities.<br />

10026 Poster Discussion Session (Board #18), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Lapatinib in advanced chordoma: A phase II study. Presenting Author:<br />

Silvia Stacchiotti, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan,<br />

Italy<br />

Background: To report on a prospective Phase II, investigator-driven,<br />

collaborative study to explore the activity <strong>of</strong> lapatinib, a dual EGFR and<br />

ErbB2 inhibitor, in EGFR-positive advanced chordomas. Methods: From<br />

December 2009 to December 2011, 19 advanced progressing chordoma<br />

patients entered this study (mean age: 59 yrs – range � 35-75; PS: 0-3 -<br />

�2 � 52%; site: sacrum � 68%, spine � 21%, skull base � 11%; disease<br />

extent: metastatic � 68%, locally advanced � 32%). EGFR expression was<br />

evaluated by immunohistochemistry in all patients. EGFR activation was<br />

detected in 15 <strong>of</strong> 17 evaluable cases by phospho-arrays and/or real-time<br />

PCR and/or fluorescence immunostaining. EGFR FISH analysis showed<br />

high level <strong>of</strong> gene gain in 4. HerB2 status is under evaluation. Patients<br />

received lapatinib 1500 mg/day (dose intensity in excess <strong>of</strong> 1250 mg/day),<br />

until progression or toxicity. The primary study end-point was response rate<br />

(RR) as for Choi criteria extended to MRI. Secondary end-points were RR by<br />

RECIST, PFS by Choi, OS, clinical benefit rate (CBR) (CR�PR�SD�6mos),<br />

correlation between EGFR status and response. Results: 16 pts are<br />

evaluable for response (3 too early); 4 patients are still on treatment. 3<br />

patients (19%) had a partial response (PR) and 8 (50%) a stable disease<br />

(SD) as their best Choi response, corresponding to RECIST SD in all cases.<br />

The 3 cases with a PR by Choi had a minor decrease in size; another patient<br />

had a mixed response. The CBR was 19%. Median PFS as for Choi was 5.5<br />

(range � 2-12�) months, with 1 patient being progression-free at 18<br />

months. Toxicity was as expected. In one case treatment was interrupted<br />

due to side effects. No correlation was observed between EGFR expression/<br />

activation and response. Conclusions: This Phase II study showed a modest<br />

activity <strong>of</strong> lapatinib in chordoma. EGFR expression was not predictive <strong>of</strong><br />

response. The clinical relevance <strong>of</strong> EGFR targeting in chordoma needs to be<br />

further investigated.<br />

10025 Poster Discussion Session (Board #17), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Establishment <strong>of</strong> a chordoma xenograft and in vivo testing <strong>of</strong> compounds<br />

identified by high-throughput screening. Presenting Author: Matteo Maria<br />

Trucco, The Johns Hopkins University, Baltimore, MD<br />

Background: Chordoma is a rare primary bone malignancy that arises in the<br />

skull base, spine and sacrum and originates from remnants <strong>of</strong> the<br />

notochord. Therapy primarily consists <strong>of</strong> surgical resection and radiation.<br />

These tumors are typically resistant to conventional chemotherapy, and to<br />

date there are no FDA-approved agents for chordoma. The lack <strong>of</strong> in vivo<br />

models <strong>of</strong> chordoma has impeded the development <strong>of</strong> new therapies for this<br />

tumor. Methods: Primary tumor from a classic sacral chordoma was<br />

obtained, immediately processed into a single cell suspension and injected<br />

in to the parasacral area <strong>of</strong> a NOD/SCID/IL-2R gamma-null mouse, and<br />

tumor grew after 3 months. The NIH Chemical Genomics Center performed<br />

high-throughput screening <strong>of</strong> 2,816 compounds. Two established chordoma<br />

cell lines, U-CH1 and UCH2B, were treated and cell viability<br />

measured by CellTiter-Glo assay. Cells were incubated for 48 hours with<br />

drug concentrations ranging from 0.5nM to 46uM. The screen yielded<br />

several compounds that showed activity and two were tested in the<br />

xenograft. Results: We have established a xenograft model <strong>of</strong> dedifferentiated<br />

chordoma. High-throughput screening <strong>of</strong> compounds identified compounds<br />

that show activity against chordoma cell lines. In vivo testing <strong>of</strong> two<br />

identified compounds showed a dramatic reduction <strong>of</strong> tumor growth.<br />

Conclusions: We have established a xenograft model <strong>of</strong> dedifferentiated<br />

chordoma. High-throughput screening <strong>of</strong> compounds identified compounds<br />

that show activity against chordoma cell lines. In vivo testing <strong>of</strong> two<br />

identified compounds showed a dramatic reduction <strong>of</strong> tumor growth.<br />

10027 Poster Discussion Session (Board #19), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Efficacy and safety <strong>of</strong> panobinostat (LBH-589), a histone-deacetylase<br />

inhibitor (HDACi), in patients (pts) with advanced, previously treated s<strong>of</strong>t<br />

tissue sarcoma (STS) and sex cord tumors (SCT): A study from the French<br />

Sarcoma Group. Presenting Author: Philippe Alexandre Cassier, Centre<br />

Léon Bérard, Lyon, France<br />

Background: Treatment options are limited for pts with advanced pretreated<br />

STS. HDACi have shown activity in preclinical models <strong>of</strong> STS and SCT<br />

Methods: Pts with advanced pretreated STS and SCT were enrolled in this<br />

open label, single arm, multicenter phase II study. Panobinostat was given<br />

orally, 40 mg thrice per week. The primary end-point was 3-month<br />

progression-free rate according to RECIST 1.1 using central review.<br />

Fleming-A’Hern single-stage design for phase II trials was used, and<br />

assuming a type I error alpha <strong>of</strong> 5% with 90% power, 15/47 pts were<br />

needed to be progression free at 3 months to reject a rate <strong>of</strong> 20%. Results:<br />

Fifty three pts were enrolled between January 2010 and January 2011.<br />

Median age was 59 (range 21-79) years, and 22 (42%) pts were males, 13<br />

had translocation-related sarcoma (TRS) and 4 had SCT. All but 5 pts had<br />

metastatic disease. The most common sites were the lung and the liver. All<br />

but one pt had documented disease progression prior to study entry. The<br />

median number <strong>of</strong> prior lines <strong>of</strong> therapy was 2 (range 1-7). Panobinostat<br />

dose was lowered to 20 mg thrice a week after 11 pts were enrolled based<br />

on the recommendation <strong>of</strong> an independent safety committee. Fifty-two pts<br />

were evaluable for toxicity (1 pt never received treatment), 48 pts (92%)<br />

reported at least one adverse event (AE) and 22 (42%) reported at least one<br />

treatment-related grade 3 or 4 AE. The most common grade 3/4 AEs were<br />

thrombocytopenia, fatigue and anemia. Fifty pts were evaluable for the<br />

primary end-point, <strong>of</strong> these, 12 pts (24%, 95%CI[13-38%]) were progression-free<br />

at 3 months, including 4/13 pts (31%, 95%CI[9-61%]) with TRS<br />

and 2/4 pts (50%, 95%CI[7-93%]) with SCT. No CR was seen, two<br />

patients (4%) with SCT had a PR and 19 pts (37%) had SD as their best<br />

response. Seven pts were progression-free at 6 months: 2 pts with SCT, 2<br />

with TRS, 1 with liposarcoma, 1 with malignant solitary fibrous tumor and 1<br />

with leiomyosarcoma. Conclusions: Panobinostat was poorly tolerated at 40<br />

mg thrice a week. Efficacy in unselected advanced STS was limited<br />

although some patients had prolonged SD. Activity in pts with SCT warrants<br />

further investigation.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


10028 Poster Discussion Session (Board #20), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Phase I study <strong>of</strong> doxorubicin (D) plus anti-insulin-like growth factor 1 receptor<br />

(IGF-1R) antibody cixutumumab (IMC-A12) in advanced s<strong>of</strong>t tissue sarcoma<br />

(STS). Presenting Author: Rashmi Chugh, University <strong>of</strong> Michigan, Ann Arbor, MI<br />

Background: IGF1R is overexpressed in many STS, but its exact role in the biology<br />

<strong>of</strong> the disease is unclear. Anti-IGF1R antibody cixutumumab (C) has shown<br />

acceptable toxicity in a single-agent phase 2 study <strong>of</strong> STS. Here we performed a<br />

dose escalation study <strong>of</strong> C with D in STS. Methods: Eligible pts had advanced<br />

STS, ECOG �2, age�16, �1 prior chemotherapy, fasting glucose�120 mg/dL<br />

and acceptable organ function. C was administered IV over 60 min on D1,8,15 at<br />

one <strong>of</strong> three dose levels and D as a 48 hr infusion on D1 <strong>of</strong> a 21-day cycle. After 6<br />

cycles <strong>of</strong> combination rx, pts with SD or better continued on single agent C. The<br />

Time-to-Event Continual Reassessment Method (TITE-CRM) was used to estimate<br />

the probability <strong>of</strong> DLT at each dose level and to assign patients to the dose<br />

with the estimated probability <strong>of</strong> DLT closest to but not exceeding 30%. Results:<br />

30 pts with STS were enrolled between 9/08-1/12. Median age was 64 (range<br />

29-80); 17M/13 F, 7 pts received prior chemo. One pt withdrew prior to rx; 5 pts<br />

are on active rx. Reasons for discontinuation were: progression (18), toxicity (3),<br />

death (1), pt decision (2). DLTs observed were hyperglycemia and mucositis.<br />

Grade 3 decrease in cardiac left ventricular ejection fraction was observed in 2<br />

pts at dose level 2 after 4 or more cycles. Common G1/2 nonhematologic AEs:<br />

nausea (73%), fatigue (68%), pain (59%), diarrhea (41%), constipation (36%),<br />

hyperglycemia (32%), mucositis (32%), nail changes (27%), weight loss (27%).<br />

Gr3/4 toxicities in �10%: hyperglycemia (18%), lymphopenia (18%), anemia<br />

(14%), neutropenia (14%), thrombocytopenia (14%). Median PFS was 5.3<br />

months, 95% CI 2.7 -7.9 months. Four <strong>of</strong> 22 pts (18%) evaluable for response<br />

achieved a PR. Conclusions: C and D is tolerable and the recommended phase II<br />

dosing is dose level 3. The potential <strong>of</strong> cardiotoxicity should be monitored and<br />

assessed further in ongoing studies <strong>of</strong> this regimen.<br />

Dose<br />

level<br />

1: C: 1 mg/kg<br />

D: 75 mg/m2 2: C: 3 mg/kg<br />

D: 75 mg/m2 3: C: 6 mg/kg<br />

D: 75 mg/m2 Pts enrolled<br />

(evaluable)<br />

DLTs<br />

observed<br />

TITE-CRM estimates<br />

Probability <strong>of</strong> DLT 95% Credible interval<br />

Objective<br />

responses<br />

4 (4) 0 4.7 1.1 – 14.7 1<br />

13 (13) 2 6.5 1.6 – 18.4 2<br />

13 (10) 0 9.7 4.8 – 24.3 1<br />

10030 Poster Discussion Session (Board #22), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Efficacy <strong>of</strong> a phosphoinositol 3 kinase (PI3K) inhibitor in gastrointestinal<br />

stromal tumor (GIST) models. Presenting Author: Thomas Van Looy,<br />

Laboratory <strong>of</strong> Experimental Oncology and Department <strong>of</strong> General Medical<br />

Oncology, KU Leuven and University Hospitals, Leuven, Belgium<br />

Background: PI3K signaling is crucial for GIST proliferation and survival.<br />

We assessed the efficacy <strong>of</strong> the PI3K inhibitor NVP-BEZ235 (BEZ), alone<br />

or in combination with imatinib (IM), in GIST xenografts. Methods: Nude<br />

mice were grafted bilaterally with human GIST, carrying either KIT exon 9<br />

(GIST-BOE, dose-dependent IM resistant) or exon 11 (GIST-DFR, IM<br />

sensitive) mutations. Animals, randomized into four groups (n�8/group)<br />

were dosed orally for 2 weeks with either vehicle, IM (50mg/kg/bid), BEZ<br />

(10mg/kg/qd), or IM�BEZ. Treatment efficacy was assessed by tumor<br />

volume, histopathology and Western immunoblotting. Moreover tumor<br />

regrowth was evaluated for 3 weeks after treatment cessation. Results: As a<br />

single agent IM and BEZ stabilized tumor growth <strong>of</strong> both GIST-BOE and<br />

DFR. Moderate to significant tumor regression was observed in GIST-BOE<br />

under BEZ (by 27%), and IM�BEZ (66%), and also in GIST-DFR under IM<br />

(75%) and IM�BEZ (75%). In GIST-BOE significant reduction in mitotic<br />

index was observed under BEZ (8.5 fold) and IM�BEZ (8.5 fold) as<br />

compared to control. In GIST-DFR mitotic activity was virtually absent<br />

under all regimens. Apoptotic activity increased significantly after treatment<br />

with IM (5.5 fold) and IM�BEZ (14.0 fold) in GIST-DFR, whereas it<br />

was almost unaffected by BEZ as single agent, as well as in all treatment<br />

groups in GIST-BOE. By Western, PI3K signaling was incompletely inhibited<br />

in all groups in GIST-DFR, and after BEZ in GIST-BOE. Complete<br />

inhibition <strong>of</strong> PI3K signaling was observed only after combination treatment.<br />

After treatment cessation long-lasting growth-inhibition was observed<br />

in IM�BEZ treated GIST-DFR. Moreover, mitotic index after BEZ<br />

and BEZ�IM in GIST-DFR was lower than in control even after treatment<br />

withdrawal. Conclusions: BEZ shows significant efficacy in GIST xenografts.<br />

Furthermore, combination with IM shows synergistic and long-lasting<br />

effects even after treatment withdrawal, which is not the case with drugs<br />

routinely used for GIST treatment.<br />

Sarcoma<br />

637s<br />

10029 Poster Discussion Session (Board #21), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Immunohistochemical identification <strong>of</strong> SDHA-mutant gastrointestinal stromal<br />

tumors (GISTs). Presenting Author: Andrew J. Wagner, Dana-Farber<br />

Cancer Institute, Boston, MA<br />

Background: GISTs are most commonly driven by activating mutations in<br />

KIT or PDGFRA. However, 15% <strong>of</strong> GISTs in adults and �90% in children<br />

lack such mutations. A subset <strong>of</strong> KIT/PDGFRA wild-type GISTs shows<br />

distinctive morphologic and clinical features and loss <strong>of</strong> expression <strong>of</strong><br />

succinate dehydrogenase (SDH) B. Only a fraction <strong>of</strong> SDHB-deficient<br />

GISTs carry loss-<strong>of</strong>-function mutations in SDHB or SDHC. Due to the<br />

complexity <strong>of</strong> its locus and the presence <strong>of</strong> several pseudogenes, SDHA is<br />

rarely analyzed. Recently, mutations in SDHA were shown to lead to loss <strong>of</strong><br />

expression <strong>of</strong> SDHA and SDHB in paraganglionomas. We sought to<br />

determine whether SDHA IHC could identify GISTs with SDHA mutations.<br />

Methods: Tumors (n�11) with features <strong>of</strong> SDH-deficient GIST (gastric<br />

origin, epithelioid morphology, multinodular/plexiform architecture) were<br />

selected from pathology archives under an IRB-approved protocol. Expression<br />

<strong>of</strong> SDHA and SDHB was determined on tumor sections by IHC.<br />

Genomic DNA was isolated from SDHA-negative tumors and amplified<br />

using primers specific to introns flanking each <strong>of</strong> the 15 SDHA exons.<br />

Amplicons were bidirectionally sequenced and compared to genomic<br />

repository data. Exons with somatic mutations were also examined in DNA<br />

from corresponding normal tissue to determine germline status. Results: All<br />

tumors (100%) were deficient for SDHB expression by IHC. Four <strong>of</strong> 11<br />

(36%) SDHB-deficient GISTs also lacked expression <strong>of</strong> SDHA. SDHA<br />

expression was intact in the 7 remaining tumors, including 3 with known<br />

SDHB (n�2) or SDHC (n�1) mutations. Nonsense mutations in SDHA<br />

were identified in all 4 SDHA-deficient tumors, caused by a single base pair<br />

(bp) substitution (n�3) or a single bp deletion (n�1), and heterozygous<br />

mutations were also found in DNA from normal tissue <strong>of</strong> all 4 patients.<br />

Somatic loss <strong>of</strong> the 2nd allele has thus far been found in 3 <strong>of</strong> 4 tumors; 2 by<br />

loss <strong>of</strong> heterozygosity and 1 by a 13-bp deletion. Further analysis <strong>of</strong> the 4th specimen is ongoing. Conclusions: SDHA mutations are a common cause <strong>of</strong><br />

SDH-deficient GISTs and result in combined loss <strong>of</strong> expression <strong>of</strong> both<br />

SDHA and SDHB. Loss <strong>of</strong> SDHA expression by IHC reliably predicts SDHA<br />

mutations and can be used to select cases for SDHA genetic testing.<br />

10031 Poster Discussion Session (Board #23), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Neoadjuvant treatment <strong>of</strong> locally advanced GIST: Results <strong>of</strong> APOLLON, a<br />

prospective, open label phase II study in KIT- or PDGFRA-positive tumors.<br />

Presenting Author: Peter Hohenberger, Department <strong>of</strong> Surgery, Mannheim<br />

University Medical Center, Mannheim, Germany<br />

Background: Imatinib may significantly downstage the metastatic GIST.<br />

This prospective, phase II, open-label trial <strong>of</strong> neoadjuvant imatinib evaluated<br />

the overall tumor response and progression rate <strong>of</strong> disease in locally<br />

advanced, non-metastic patients. Methods: Inclusion criteria were: KIT<br />

(n�39) or PDFRA (n�6) positive GIST, proven by biopsy and IHC. Tumors<br />

had to be locally advanced, potentially resectable, M0. Patients received<br />

400 mg <strong>of</strong> imatinib daily (KIT exon 9 mutations: twice daily) for 6 months,<br />

in the absence <strong>of</strong> disease progression or unacceptable toxicity. At two<br />

months 18F-FDG-PET examination was performed to assess response in<br />

paralle to CT imaging. Adjuvant treatment postoperatively was not part <strong>of</strong><br />

the study protocol (CST1571-BDE43, NCT00112632). Median follow-up<br />

is 36 months in 39 patients. Results: From 7/2005 to 10/ 2009, 41<br />

patients (16f, 25 m, mean age 54 yrs) were recruited to the trial with an<br />

average tumor size <strong>of</strong> 10.8 cm. One patient died from myocardial infarction<br />

3 weeks after start <strong>of</strong> treatment, all other patients completed the protocol.<br />

In two patients dose reduction/interruption due to toxicity was required.<br />

34/41 patients underwent resection <strong>of</strong> the tumor after a median <strong>of</strong> 200 d<br />

(range 57-394), while two patients had to be operated early due to disease<br />

progression. R0 resections were performed in 30/34 patients, two patients<br />

showed M1 disease at resection. Five patients showed developed progression<br />

postop., resulting in a PFS rate at 3 years <strong>of</strong> 85.2%. No patient has<br />

died so far from the disease. Conclusions: Neoadjuvant treatment with<br />

imatinib for six months is a safe treatment in patients with locally advanced<br />

disease. The extent <strong>of</strong> the operation can be significantly downsized after<br />

pretreatment. Despite the fact that no adjuvant treatment was foreseen, the<br />

progression-free rate at 3 years postoperatively is promising.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


638s Sarcoma<br />

10032^ Poster Discussion Session (Board #24), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Phase I trial <strong>of</strong> panobinostat (P) and imatinib (IM) in patients with<br />

treatment-refractory gastrointestinal stromal tumors (GIST). Presenting<br />

Author: Sebastian Bauer, Sarcoma Center, West German Cancer Center,<br />

Essen, Germany<br />

Background: Panobinostat (LBH589; P) is a pan-deacetylase-inhibitor that<br />

has preclinical activity in combination with IM in GIST models in vitro and<br />

in vivo. Aim <strong>of</strong> this study was to determine the maximum tolerated dose<br />

(MTD) and dose-limiting toxicities (DLT) <strong>of</strong> escalating doses <strong>of</strong> P in<br />

combination with IM in patients with GIST who have failed IM and sunitinib<br />

treatment. Methods: This was a two-center phase I study using a 3�3<br />

design with a prespecified expansion <strong>of</strong> the MTD cohort. IM was administered<br />

at a dose <strong>of</strong> 400mg qd. Following a 7 day run-in phase, escalating<br />

doses <strong>of</strong> P were added. The starting dose for P was 20 mg given as a<br />

three-times-per-week (MWF schedule) oral dose for 3 out <strong>of</strong> 4 weeks. Doses<br />

were increased by 10 mg if no dose limiting toxicities emerged. Blood<br />

samples were drawn for PK and biomarker assessments <strong>of</strong> IM, its main<br />

metabolite N-desmethyl-IM, and P using a validated RP-HPLC method.<br />

Acetylation <strong>of</strong> histone A3 was evaluated in peripheral blood mononuclear<br />

cells (PBMNC) as pharmacodynamic marker for P activity. Metabolic<br />

response using PET (EORTC-PET study criteria) was assessed on day 7 <strong>of</strong><br />

IM run-in and after 3 weeks <strong>of</strong> combined treatment with IM and P. Results:<br />

In total 12 extensively pretreated (median 5 pretreatments) pts (4 f, 8 m;<br />

median age 56 y, 34-75 y) received study treatment at 2 dose levels (DL). 2<br />

dose-limiting toxicities (grade 4 thrombocytopenia) occurred at DL 2 (30<br />

mg). Most common AEs were thrombocytopenia, anemia, fatigue, nausea,<br />

emesis, diarrhea, creatinine elevation, abdominal cramping, and weight<br />

loss. DL 1 (20mg) was declared MTD, and 5 additional pts were enrolled at<br />

DL1. Analysis <strong>of</strong> P and IM PK revealed mean peak concentration <strong>of</strong> 14.8<br />

�/- 9.5 ng/ml for P (20 mg). IM plasma concentrations with 400 mg<br />

once-daily administration were 2.8 � 1.1 �g/mL at peak and 1.2 � 0.4<br />

�g/mL at trough. Histone A3 acetylation was demonstrated in PBMNC from<br />

pts treated at DL 1. 11 pts were evaluable for PET response: 1 had mPR, 7<br />

had mSD and 3 had mPD. Longest treatment duration was 17 weeks<br />

(median: 6wks). Conclusions: P in combination with IM is moderately<br />

tolerated. Evidence <strong>of</strong> target inhibition at the MTD was associated with<br />

limited clinical activity in heavily pretreated pts with GIST.<br />

10034 General Poster Session (Board #46C), Sun, 8:00 AM-12:00 PM<br />

Characteristics <strong>of</strong> sarcoma patients with long-term response to gemcitabine<br />

and paclitaxel. Presenting Author: Daniela Katz, Sharette Institute <strong>of</strong><br />

Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel<br />

Background: The combination <strong>of</strong> fixed-dose-rate gemcitabine and docetaxel<br />

has demonstrated efficacy in advanced uterine leiomyosarcoma and nonleiomyosarcoma<br />

sarcomas. We evaluated the tolerability and efficacy <strong>of</strong><br />

modified taxol-gemcitabine (GemTax) protocol for patient with advanced<br />

sarcoma. The protocol was designed on a weekly basis to reduce toxicity.<br />

The purpose <strong>of</strong> this retrospective study was to evaluate the characteristics<br />

<strong>of</strong> long term-responders (�12 treatments) to weekly GemTax, among<br />

pre-treated advanced high-grade sarcoma patients. Methods: From May<br />

2004 to October 2011, 48 advanced sarcoma patients were treated with<br />

combination paclitaxel (60-90 mg/m2) and gemcitabine (600-900 mg/m2<br />

over 100 min), on days 1,8 <strong>of</strong> every 3-week cycle or on days 1, 8 and 15 <strong>of</strong><br />

every 4-week cycle at a wide-range treatment lines. Two patients are still on<br />

the protocol. Results: Fifteen patients (6 males and 9 females) completed<br />

12 weekly treatments or more (mean 22.3 range 12-37) <strong>of</strong> which 4<br />

completed �30 treatments. These include 5 leiomyosarcomas (one retroperitoneal),<br />

2 uterine leiomyosarcomas, 3 MFH, 2 spindle sarcoma, 1<br />

osteosarcoma, 1 adamantimoma and 1 pleomorphic rhabdomyosarcoma.<br />

GemTax was administered as 1st or 2nd line treatment in 2 and 8 patients,<br />

respectively, and as 3rd -6th line in 5. Of these patients, none achieved<br />

complete response (CR), 7/15 (47%) achieved a partial response (PR) and<br />

8/15 (53%) had stable disease (SD). Median treatment duration was 10.3<br />

months (range 4-22) Median progression-free survival was 28.85 months<br />

(range 5-84 months), and median overall survival was 51.2 months (range<br />

16-95 months). A lower starting dose <strong>of</strong> paclitaxel (60-70 mg/m2) and<br />

gemcitabine (600-700 mg/m2) characterized the 4 patients with responses<br />

�30 cycles. Only five patients required dose reduction ranging<br />

between 10-40% due to grade 3 hematological toxicity. None <strong>of</strong> these<br />

patients, however, had febrile neutropenia, or bleeding events, and all<br />

non-hematologic toxicities including neurotoxicity were manageable.<br />

Conclusions: GemTax is a reasonable treatment option for patients with<br />

advanced pre-treated high-grade sarcomas. The regimen is well tolerated<br />

and dose adjustments do not seem to impact response.<br />

10033 Poster Discussion Session (Board #25), Sat, 1:15 PM-5:15 PM and<br />

4:45 PM-5:45 PM<br />

Dasatinib first-line treatment in gastrointestinal stromal tumors: A multicenter<br />

phase II trial <strong>of</strong> the SAKK (SAKK 56/07). Presenting Author:<br />

Michael Montemurro, Centre Pluridisciplinaire d’Oncologie, University<br />

Hospital Lausanne, Lausanne, Switzerland<br />

Background: First line imatinib has improved the outcome <strong>of</strong> patients (pts)<br />

with gastrointestinal stromal tumor (GIST), but primary and secondary<br />

resistance remain a problem. Dasatinib is a second generation tyrosine<br />

kinase inhibitor (TKI) and inhibits the activity <strong>of</strong> bcr-abl, src-family kinases<br />

along with a number <strong>of</strong> other oncogenic kinases including kit. In vitro,<br />

dasatinib has shown activity against imatinib-resistant cell lines. Routinely,<br />

pts diagnosed with GIST and treated with TKIs are monitored by<br />

computed tomography (CT). 18F-fluorodeoxyglucose positron-emissiontomography<br />

(FDG-PET) measures tumor metabolic activity for early response<br />

assessment, which might be <strong>of</strong> particular use in the clinical trial<br />

setting. Methods: This two-stage phase II trial investigated dasatinib in pts<br />

with TKI-naïve GIST. Dasatinib starting dose was 2x70mg/day in pts with<br />

histologically proven, FDG-PET positive GIST. Response evaluation was<br />

done by serial CT and FDG-PET using EORTC PET response criteria (Young<br />

et al. 1999). Elective surgery was allowed after 6 months <strong>of</strong> trial treatment.<br />

Primary endpoint was response (CR�PR) by FDG-PET after one month <strong>of</strong><br />

dasatinib. Results: 47 <strong>of</strong> planned 52 pts have been enrolled from December<br />

2007 to November 2011, when the trial was terminated due to slow<br />

accrual. 43 pts were eligible. Median age was 61 years, 24 pts were male,<br />

19 female and 41 had a performance status <strong>of</strong> 0 or 1. At a median<br />

follow-up <strong>of</strong> 11.9 months, 20 pts were still on treatment. Pts went <strong>of</strong>f trial<br />

for elective surgery (n�6), progression (n�11), toxicity (n�3), and three<br />

patients died (one on-drug). 5% <strong>of</strong> pts experienced G4, and 38% <strong>of</strong> pts G3<br />

toxicity, which were most <strong>of</strong>ten gastrointestinal or pulmonary. 28% had<br />

their dose reduced or interrupted. The primary endpoint, FDG-PET response<br />

rate (CR�PR) at 4 weeks was 67% (13 CR, 16 PR, 7 SD, 3 PD, 4<br />

not evaluable). Median progression-free survival is 11.1 months and<br />

median overall survival not yet reached. Conclusions: Dasatinib shows<br />

promising efficacy in TKI-naïve pts with FDG-PET positive GIST. Mutational<br />

data will be presented at the meeting.<br />

10035 General Poster Session (Board #46D), Sun, 8:00 AM-12:00 PM<br />

Effect <strong>of</strong> metastasis surgery on overall survival in patients (pts) with<br />

advanced s<strong>of</strong>t tissue sarcoma (ASTS): A subanalysis <strong>of</strong> the PALSAR trials.<br />

Presenting Author: Nuria Kotecki, Centre Oscar Lambret, Lille, France<br />

Background: The role <strong>of</strong> surgery in pts with ASTS remains controversial. We<br />

have conducted an exploratory retrospective analysis <strong>of</strong> the role <strong>of</strong> metastasis<br />

surgery in ASTS pts receiving 1st-line chemotherapy (CT). Methods: The<br />

database includes all pts enrolled in PALSAR-1 and PALSAR-2 trials<br />

[Fayette 2009; Bui-Nguyen 2011], treated with dose-intensified MAID or<br />

high-dose CT with peripheral blood stem cells support as 1st-line treatment<br />

<strong>of</strong> ASTS. In our analysis, the primary endpoint is overall survival (OS).<br />

Log-ranks are used for univariate analysis and Cox model for multivariate<br />

analysis. Impact <strong>of</strong> treatments had been evaluated after adjustment to<br />

confounders (Cox Model). Confounders were defined as parameters with<br />

significantly different distribution in pts who underwent metastasis surgery<br />

and those who did not (p�0.05) and significantly associated with OS<br />

(p�0.05). Results: The database consists <strong>of</strong> 410 pts (160 in PALSAR-1<br />

and 248 in PALSAR-2) with a median age <strong>of</strong> 43. Among them, 77 patients<br />

(18%) underwent metastasis surgery. At the end <strong>of</strong> the treatment, 61 pts<br />

experienced complete response (CR), 45 with CT alone and 16 with<br />

metastasis surgery and CT. The median follow-up was 29 months. The<br />

median OS was 35.7 months (29.9-41.5). The following parameters are<br />

associated with longer OS in univariate analysis: primary location<br />

(p�0.0001), performance status (p�0.010) and absence <strong>of</strong> liver metastasis<br />

(p�0.001). We identified 4 factors associated with metastasis surgery<br />

(n�76): limb/trunk primaries, young age, absence <strong>of</strong> liver metastasis and<br />

absence <strong>of</strong> progression <strong>of</strong> the target lesions after 4 cycles. The sole<br />

identified confounder was primary location. In multivariate analysis the 2<br />

categories <strong>of</strong> patients experiencing significant longer OS are those with CR<br />

without surgery (HR�2.2, [1.7-5.8], p�0.0001) and those with CR<br />

following metastasis surgery (HR�3.8, [2.3-6.2], p�0.0001). Conclusions:<br />

Among the pts with ASTS receiving poly-CT as 1st-line treatment, the OS <strong>of</strong><br />

pts experiencing CR with or without metastasis surgery appears similar.<br />

Metastatis surgery without CR does not <strong>of</strong>fer significant OS advantage.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


10036 General Poster Session (Board #46E), Sun, 8:00 AM-12:00 PM<br />

Phase Ib/II study <strong>of</strong> INNO-206 (EMCH-doxorubicin) in patients with s<strong>of</strong>t<br />

tissue sarcoma. Presenting Author: Sant P. Chawla, Sarcoma Oncology<br />

Center, Santa Monica, CA<br />

Background: The safety and preliminary tumor response <strong>of</strong> a doxorubicin<br />

conjugate, INNO-206, was evaluated primarily in patients with metastatic<br />

STS who progressed on prior chemotherpy. INNO-206 consists <strong>of</strong> doxorubicin<br />

attached to an acid-sensitive linker that binds covalently to cysteine-34<br />

in circulating albumin. Methods: INNO-206 was administered IV at doses <strong>of</strong><br />

either 230, 350 and 450 mg/m2 (165, 260 and 325 mg/m2 dox. eq.) every<br />

21 days for up to 8 consecutive cycles. Subsequent dose levels were<br />

administered if � 2/5 or 4/8 patients experienced a non-hematological<br />

dose-limiting toxicity during Cycle 1. Tumor response was monitored every<br />

other month and treatment continued until tumor progression or unacceptable<br />

toxicity. Standard safety monitoring was performed and cardiac<br />

function was followed periodically using MUGA or cardiac ultrasound.<br />

Results: As <strong>of</strong> January 11, 2012, 25 patients were entered in the study.<br />

21/25 patients had STS <strong>of</strong> various types. Of the 5 patients treated at 230<br />

mg/m2 INNO-206, 1 subject had grade 3 fatigue and acid reflux. Of the<br />

initial 5 patients entered at the 350 mg/m2 dose, no individuals experienced<br />

a grade 3 or 4 non-hematological toxicity during cycle 1. 2 patients<br />

treated at the 450 mg/m2 dose developed grade 3 oral mucositis during<br />

cycle 1. No patient exhibited cardiotoxicity as determined by MUGA or<br />

cardiac ultrasound. The MTD was determined to be 350 mg/m2 INNO-206<br />

(260 mg/m2 dox.eq.). 15 more patients were entered at this dose (total <strong>of</strong><br />

20 patients). Of the 16 patients with STS, 3 objective partial responses<br />

(one <strong>of</strong> whom received prior doxorubicin) are ongoing as well as 10 patients<br />

with stable disease (range 2-7 months). 1 patient had progressive disease<br />

at the first evaluation. 2 patients died within the first cycle, one due to<br />

progressive disease and the other due to sepsis. Conclusions: INNO-206 is<br />

an active drug for the treatment <strong>of</strong> patients with advanced STS who have<br />

failed prior chemotherapy. Cumulative doses <strong>of</strong> 2000 mg/m2 <strong>of</strong> doxorubicin<br />

equivalents have been achieved, which is over 3 1/2 x the peak<br />

cumulative dose <strong>of</strong> standard doxorubicin. Adverse events are primarily<br />

hematological and no cardiotoxicity has been observed.<br />

10038 General Poster Session (Board #46G), Sun, 8:00 AM-12:00 PM<br />

A retrospective study from the Royal Marsden Hospital (RMH) <strong>of</strong> patients<br />

with malignant perivascular epithelioid cell tumors (PEComa) receiving<br />

treatment with sirolimus (SI) or temsirolimus (TSI). Presenting Author:<br />

Joanna Vitfell Pedersen, Sarcoma Unit, The Royal Marsden Hospital,<br />

London, United Kingdom<br />

Background: Perivascular epithelioid cell tumors (PEComas) are rare tumors<br />

driven by tuberous sclerosis complex gene mutations causing upregulation<br />

<strong>of</strong> mTOR. Two studies reporting a total <strong>of</strong> five patients (pts) have described<br />

the treatment <strong>of</strong> PEComa by mTOR inhibition. We report the outcome <strong>of</strong> 7<br />

pts with PEComa treated with SI or TSI at RMH. Methods: A retrospective<br />

analysis was performed on all pts with PEComa referred to the Sarcoma unit<br />

at RMH between 2000 and 2011. Data collected included gender,<br />

performance status, site <strong>of</strong> primary tumour, tumour size, surgery, SI/TSI<br />

and SI/TSI dose, toxicity according to common toxicity criteria (CTC)<br />

version 2.0 and response by RECIST. Results: Five (71%) pts were female,<br />

median age was 46 years. Two (29%) had metastatic disease at baseline.<br />

Most common primary site was the gastrointestinal tract (29%). Six pts<br />

received SI, one TSI. Median treatment duration was 131 days (7-1135).<br />

TSI was given at 25 mg IV weekly. Median starting dose <strong>of</strong> SI was 3 mg daily<br />

(1-4), median highest dose was 4 mg daily (1-5). Best responses by<br />

computed tomography (CT) scan by RECIST criteria were partial response:<br />

3 (43%), stable disease: 2 (29%) and progressive disease (PD): 1 (14%).<br />

One pt only received 7 days <strong>of</strong> SI and was therefore not evaluable. Two pts<br />

continue on treatment, 3 stopped due to PD, one stopped due to grade (gr)<br />

3 thrombocytopenia and one due to gr 3 cough, but two weeks later had<br />

confirmed PD on CT. Other toxicities reported were generally mild (gr 1/2)<br />

and included mucositis (n:3), rash (n:2), fatigue (n:1), anaemia (n:1),<br />

tooth pain (n:1) and oedema (n:1). One pt experienced a grade 3 trigeminal<br />

nerve pain leading to dose reduction in SI. Therapeutic drug monitoring was<br />

not performed. Conclusions: Our study confirms that SI and TSI are well<br />

tolerated with good clinical response and supports the continuous administration<br />

<strong>of</strong> SI or TSI for PEComas. A median treatment time <strong>of</strong> only 131 days<br />

indicates that most pts have a good response to treatment, but responses<br />

are sometimes short-lived and further treatment options are needed,<br />

justifying further research into inhibitors <strong>of</strong> this signalling pathway.<br />

Sarcoma<br />

639s<br />

10037 General Poster Session (Board #46F), Sun, 8:00 AM-12:00 PM<br />

Analysis <strong>of</strong> a neoadjuvant regimen with a fixed dose <strong>of</strong> doxorubicin followed<br />

by a 10-day course <strong>of</strong> 27 Gy radiation in the treatment <strong>of</strong> s<strong>of</strong>t tissue<br />

sarcoma (STS) <strong>of</strong> the extremity/trunk. Presenting Author: John Mathews,<br />

Scott and White Memorial Hospital, Temple, TX<br />

Background: Local tumor control and preservation <strong>of</strong> limb function are<br />

major goals <strong>of</strong> treatment <strong>of</strong> STS. With current standard <strong>of</strong> combining<br />

limb-sparing surgery and 5-6 weeks <strong>of</strong> neoadjuvant or adjuvant 50-65 Gy<br />

radiation, reducing local recurrence has been a challenge with rates <strong>of</strong><br />

15-30%. Eilber et al. (Cancer Treatment Symposia 1985; 3:49-57) used a<br />

fixed daily dose <strong>of</strong> doxorubicin for three days with radiation <strong>of</strong> 35Gy. This<br />

yielded a 5% local recurrence, while wound complication rate was 35 %.<br />

We reduced the radiation dose to lower the wound complications rate. We<br />

report a cohort <strong>of</strong> patients treated in this manner at our institution.<br />

Methods: After a baseline echocardiogram, patients received a radiosensitizing<br />

dose <strong>of</strong> intravenous doxorubicin (30mg/day) for 3 consecutive days,<br />

then radiation with 2.7 Gy in 10 fractions, followed by limb sparing surgery.<br />

After IRB approval, we retrospectively analyzed records <strong>of</strong> 191 patients<br />

during the treatment period from 1991 to 2010. Data extracted included<br />

local tumor recurrences and wound complications defined as non-healed<br />

wound at 1 month, number <strong>of</strong> limb salvage surgeries and involvement <strong>of</strong><br />

surgical margins. Results: Of the 191 patients, median age was 53 (range,<br />

13-89). Stages at presentation included I, II, III and IV, with 2%, 33%,<br />

61%, 4% respectively. The median follow up was 5 years. Of the 24 types<br />

<strong>of</strong> sarcomas treated, pleomorphic sarcoma was the most common at 21%.<br />

The local recurrence rate was 5% (95% CI, 2.7-8.6%) and local wound<br />

complications were 21% (95% CI, 15-28%). Most (172 or 90%) <strong>of</strong><br />

patients were able to undergo limb salvage surgery while 19 (10%) required<br />

amputation. Of the 172 patients that had limb salvage, 97% (95% CI,<br />

93-98%) had wide surgical margins, three had � 2 cm margins and two<br />

had involved margins. Conclusions: To our knowledge, this is the largest<br />

study showing that this short two week course <strong>of</strong> chemoradiation is<br />

effective in achieving high rate <strong>of</strong> local control, acceptable wound complication<br />

rate, wide surgical margins and limb salvage in the treatment <strong>of</strong> STS.<br />

Randomized trials are warranted to confirm results.<br />

10039 General Poster Session (Board #46H), Sun, 8:00 AM-12:00 PM<br />

Palliative ambulatory 14-day infusional ifosfamide in the outpatient setting<br />

for treatment <strong>of</strong> advanced s<strong>of</strong>t tissue sarcomas (STS): “Old wine in a new<br />

bottle.” Presenting Author: Salma Moona Alam, Sarcoma Unit,The Royal<br />

Marsden Hospital NHS Foundation Trust, London, United Kingdom<br />

Background: Ifosfamide (IFO) has recognised activity in s<strong>of</strong>t tissue sarcomas.<br />

There is evidence for improved cytotoxicity and tolerability utilising a<br />

prolonged 14 day infusional outpatient regimen compared with the<br />

standard 3-day regimen (Loeffler et al 1990). We undertook a retrospective<br />

review <strong>of</strong> all patients treated with this regimen from September 2008 -<br />

December 2011 to determine toxicity and treatment response. Methods:<br />

Pts. were identified from our database and demographics, histological<br />

subtype, toxicity and survival outcomes were collected. IFO was given via<br />

central venous access using 2x7daypumps at 1g/m2/day with mesna for<br />

14 days q 4 weekly. Oral sodium bicarbonate was used to maintain alkaline<br />

urine with oral mesna for haematuria, and thiamine for symptoms <strong>of</strong><br />

encephalopathy as required. Results: 29 patients (pts), with advanced<br />

sarcoma, median age 56 years (27-76 yrs), 14F 15 M, median number <strong>of</strong><br />

cycles � 1 ( 1-9). At baseline: PS 0�1, 1�24, 2�4. 18 pts had<br />

de-differentiated liposarcoma (D-LPS), 6 synovial sarcoma, 5 had other<br />

subtypes. 12 pts received only 1 cycle, with 5 stopping due to encephalopathy.<br />

Ten patients developed encephalopathy, 9 occurring in cycle 1, other<br />

toxicity was tolerable, with Gr 2-3 nausea (15 pts) and vomiting (9pts) and<br />

only 2 Gr 3 episodes <strong>of</strong> myelotoxicity for all cycles. 4 patients achieved PR,<br />

(2 with D-LPS and 2 with Synovial sarcoma) 11 pts. had SD. Median PFS<br />

was 19 months and OS was 12.5 months. 2 pts. with D-LPS had a<br />

sustained prolonged response <strong>of</strong> 15 and 16 months. Conclusions: Infusional<br />

IFO is generally well tolerated; however a significant incidence <strong>of</strong><br />

neurotoxicity was seen. Pts. with D-LPS demonstrated best response to<br />

treatment, a sarcoma subtype that has previously failed to respond to<br />

systemic therapy. This regime warrants further investigation.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


640s Sarcoma<br />

10040 General Poster Session (Board #47A), Sun, 8:00 AM-12:00 PM<br />

Gemcitabine/docetaxel for metastatic or locally advanced s<strong>of</strong>t tissue<br />

sarcoma: A retrospective single-center experience. Presenting Author:<br />

Gerlinde Egerer, Department <strong>of</strong> Hematology, Oncology, and Rheumatology,<br />

Heidelberg University Hospital, Heidelberg, Germany<br />

Background: S<strong>of</strong>t tissue sarcoma (STS) represent a group <strong>of</strong> rare malignant<br />

tumors. Survival for patients with disseminated disease is dismal. Gemcitabine/docetaxel<br />

(GD) is a commonly used systemic 2°-line regime in this<br />

setting. Methods: Here we report on our single center experience <strong>of</strong> GD (G<br />

900 mg/m² or 675 mg/m² d1�8 and D 100 mg/m² d8) in STS. Patients<br />

were retrospectively analysed. Restaging according to RECIST criteria <strong>of</strong><br />

primary tumor site and metastases was performed every 3 cycles or on<br />

clinical progression. Progression-free (PFS) and overall survival (OS) were<br />

estimated using the method <strong>of</strong> Kaplan and Meier. Results: Between 2005<br />

and 2011, 34 STS patients (male�20, female�14, median age 59 years<br />

[range 32-75]) were treated with GD at our institution. Histological<br />

subtypes comprised leiomyo- (n�13), lipo- (n�7), pleomorphic- (n�6),<br />

rhabdomyo- (n�3), synovial sarcoma (n�2), and other subtypes (n�3)<br />

with tumor grades being G1 (n�1), G2 (n�9) and G3 (n�24). Primary<br />

tumor sites included extremities (n�19), abdomen/retroperitoneum (n�10),<br />

trunk (n�3), and others (n�2). Metastases were found in lung (n�26), s<strong>of</strong>t<br />

tissue/solid organs (n�12), bone (n�8) and lymph nodes (n�5). Patients<br />

had received no (n�2),1(n�22), 2 (n�9) or 3 (n�1) previous lines <strong>of</strong><br />

CTX. A total number <strong>of</strong> 158 cycles was administered with a median number<br />

<strong>of</strong> 6 cycles (range 1-6) per patient. Best response by RECIST criteria after 3<br />

cycles <strong>of</strong> treatment was PR (n�2, 6%). Disease stabilisation (SD) was<br />

achieved in further 22 subjects, resulting in a clinical benefit rate (CBR) <strong>of</strong><br />

70%. Median PFS and OS for the whole population were 7.7 and 15.3<br />

months, respectively. PFS at 6 months was 62%. Patients responding to<br />

therapy had a significantly longer median PFS with 8.6 months (p<br />

�0.0001; HR 33.1) and OS with 22.5 months (p�0.0001; HR 20.2).<br />

Major toxicities included hand-foot syndrome (n�10), febrile neutropenia<br />

(n�7), mucositis (n�7), oedema (n�5), hematological toxicity (n�4) and<br />

polyneuropathy (n�3), leading to dose reductions in 10 patients.<br />

Conclusions: GD is an active regimen in STS with tolerable side effects.<br />

CBR was 70% after 3 cycles. Patients achieving at least SD had a<br />

significantly prolonged PFS and OS.<br />

10042 General Poster Session (Board #47C), Sun, 8:00 AM-12:00 PM<br />

Sarcoligo: Impact <strong>of</strong> local ablative treatment <strong>of</strong> oligometastatic sarcomas<br />

on overall survival. Presenting Author: Juliette Thariat, Centre Antoine-<br />

Lacassagne, Nice, France<br />

Background: 40-50% <strong>of</strong> sarcomas become metastatic. Median survival <strong>of</strong><br />

metastatic patients has improved over time. The probably multifactorial<br />

reasons for such improvement are not fully clear. Noteworthy, for patients<br />

with a controlled primary and a limited number <strong>of</strong> lung metastases,<br />

complete resection <strong>of</strong> their metastases yields survival rates <strong>of</strong> up to 40% at<br />

three years. Advances in surgery, radiotherapy and radi<strong>of</strong>requency have<br />

fostered the use <strong>of</strong> local treatments for various metastatic sites (lung, liver,<br />

spine...). Methods: A multicentric retrospective study <strong>of</strong> the Groupe<br />

Sarcome Francais (GSF-GETO); approved by the nationally-review board<br />

and ethical committee, was conducted to assess the impact <strong>of</strong> local<br />

ablative treatment on overall survival. Patients who had had oligometastases<br />

(any site, 1-5 synchronous metastases) at diagnostic or during the<br />

course <strong>of</strong> disease between 2000 and 2010 were included. Results: Median<br />

age <strong>of</strong> the 243 oligometastatic sarcoma patients was 53 years-old (11-86).<br />

Patients had grade I, II and III in 7.5%, 29.6% and 63.3% <strong>of</strong> cases,<br />

respectively with various histologies. 69% <strong>of</strong> patients underwent local<br />

ablative treatment <strong>of</strong> metastases. Median follow-up was 59 months<br />

(4-212) for living patients. Median overall survival was 51 months (1-348).<br />

On univariate analysis, grade, histology, absence <strong>of</strong> chemotherapy, local<br />

ablative treatment (surgery, irradiation, radi<strong>of</strong>requency or chemoembolisation)<br />

correlated with survival but not age or site <strong>of</strong> oligometastasis. On<br />

multivariate analyses, grade (hazard ratio HR 0.12 [CI95 0.3-0.6]) and<br />

local ablative treatment (HR 3.8 [CI95 2.1-7.1]) remained significant.<br />

Conclusions: Local ablative treatment <strong>of</strong> metastases is associated with<br />

better survival in sarcoma patients with oligometastatic disease. The role <strong>of</strong><br />

the locoregional treatment <strong>of</strong> metastases and its impact on quality <strong>of</strong> life<br />

should be assessed prospectively.<br />

10041^ General Poster Session (Board #47B), Sun, 8:00 AM-12:00 PM<br />

Pharmacokinetic analyses <strong>of</strong> vorinostat in patients with metastatic s<strong>of</strong>t<br />

tissue sarcoma. Presenting Author: Thomas Schmitt, Department <strong>of</strong><br />

Hematology, Oncology, and Rheumatology, Heidelberg University Hospital,<br />

Heidelberg, Germany<br />

Background: New treatment options for patients with metastatic s<strong>of</strong>t tissue<br />

sarcoma (STS) are urgently needed. Preclinical studies suggest activity <strong>of</strong><br />

vorinostat (V), a histone desacetylase (HDAC) inhibitor, in STS. Methods:<br />

We initiated a multicenter, open-label, non-randomized phase II trial<br />

(SAHA-I, NCT00918489) to investigate efficacy and safety <strong>of</strong> V in patients<br />

with metastatic STS failing 1°-line anthracycline-based CTX. Patients were<br />

treated with V 400 mg po QD for 28 days followed by a therapy-free period<br />

<strong>of</strong> 7 days. Blood samples for pharmacokinetic (PK) analyses were collected<br />

on day 7 <strong>of</strong> treatment cycle 1. Samples were acquired before and 30, 60,<br />

90, 120, 240, 360, and 480 minutes after oral administration <strong>of</strong> V.<br />

Plasma- and intracellular concentration <strong>of</strong> V in peripheral blood mononuclear<br />

cells (PBMCs) was determined by mass spectrometry. Statistical<br />

differences were assessed by Wilcoxon matched-pairs signed rank test.<br />

This trial is ongoing. Results: Data on PK was available for n�8 subjects<br />

(male�4, female�4, median age�62 years). In plasma samples, mean<br />

Cmax (maximum concentration), tmax (time to reach max. concentration),<br />

AUC (area under the plasma-concentration time curve), t1/2 (elimination<br />

half-life) and Cl/F (apparent total clearance) were 350 ng/mL, 101 min,<br />

71.1 min*�g/mL, 103 min and 5903 mL/min. The corresponding parameters<br />

in PBMCs were 558 ng/mL, 97.5 min, 208.4 min*�g/mL, 286 min<br />

and 2475 mL/min, respectively.The AUC plasma/PBMC ratio was 2.93,<br />

indicating accumulation <strong>of</strong> V in PBMCs. Differences in AUC (p�.008) and<br />

t1/2 (p�.01) reached statistical significance. Conclusions: Interestingly,<br />

significantly higher concentrations <strong>of</strong> V were achieved in PBMCs andelimination<br />

half-life was prolonged compared to plasma. These results suggest<br />

potent intracellular activity <strong>of</strong> V.<br />

10043 General Poster Session (Board #47D), Sun, 8:00 AM-12:00 PM<br />

<strong>Clinical</strong> activity <strong>of</strong> mTOR inhibition in combination with cyclophosphamide<br />

in the treatment <strong>of</strong> recurrent nonresectable chondrosarcomas. Presenting<br />

Author: Ofer Merimsky, Sourasky Medical Center, Tel Aviv, Israel<br />

Background: Chondrosarcomas (CS) represent a heterogeneous group <strong>of</strong><br />

rare sarcomas, poorly responsive to chemotherapy or radiotherapy. When<br />

local therapies in recurrent or metastatic disease are exhausted, chemotherapy<br />

plays a marginal role. Different molecular pathways have been<br />

shown to be activated in CS. In this retrospective study we summarize our<br />

experience in treating a cohort <strong>of</strong> patients with recurrent nonresectable CS<br />

with a combination <strong>of</strong> sirolimus and cyclophosphamide. Methods: Nine<br />

consecutive patients with nonresectable CS were <strong>of</strong>fered <strong>of</strong>f-label treatment<br />

with sirolimus and cyclophosphamide between 2007-2011. Tumor<br />

response, progression-free survival (PFS), adverse events and other relevant<br />

clinical data were analyzed. Results: The median patients’ age was 52<br />

(range, 35-68). Median disease-free interval (DFI) since the primary<br />

diagnosis was 24 months. Median time from the disease recurrence to<br />

initiation <strong>of</strong> sirolimus and cyclophosphamide treatment was 20.9 months<br />

due to additional local surgical treatments/excision <strong>of</strong> metastases/slow and<br />

asymptomatic progression. One (11%) objective response was observed<br />

and five (56%) patients had stabilization <strong>of</strong> disease for at least 6 months.<br />

One patient has been treated for only 3.5 months and reported symptomatic<br />

improvement at this stage, two patients had progressive disease.<br />

Median time to treatment failure (TTF) was 15 months (range, 2.8-30.3).<br />

No significant adverse events were observed. Conclusions: Although advanced<br />

CS remains an incurable disease, our experience suggests that a<br />

combination <strong>of</strong> sirolimus and cyclophosphamide is well-tolerated and has<br />

meaningful clinical activity with clinical benefit rate <strong>of</strong> 67%. Further<br />

prospective studies are warranted.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


10044 General Poster Session (Board #47E), Sun, 8:00 AM-12:00 PM<br />

High-dose ifosfamide (HDI) in metastatic synovial sarcoma: The Institut<br />

Gustave Roussy experience. Presenting Author: Arslane Skander Rahal,<br />

Institut Gustave Roussy, Villejuif, France<br />

Background: Synovial sarcoma (SS) is one <strong>of</strong> the most chemo-sensitive<br />

histological subtypes <strong>of</strong> sarcoma but with a dismal prognosis. In metastatic<br />

setting, Anthracyclin/ifosfamide-containing chemotherapy (CT) regimen is<br />

the gold standard <strong>of</strong> treatment in front-line therapy. Patients (pts) with<br />

resistant diseases are candidates to investigational drugs or higher dose<br />

ifosfamide regimen in selected pts. Methods: Pts treated in our institution,<br />

with histological confirmation <strong>of</strong> advanced SS, adequate organ function<br />

and in good condition (PS�2) were included in this retrospective analysis.<br />

Pts who progressed after 1–2 chemotherapy lines received High-dose<br />

ifosfamide (HDI) in the form <strong>of</strong> 4 g/m2 daily (over 24 hours CI) for 3 days for<br />

a total dose <strong>of</strong> 12 g/m2 ; cycles were given with growth factor support and<br />

were repeated every 3 weeks. The primary endpoints were PFS and overall<br />

response rate (ORR); the secondary endpoints OS and toxicity pr<strong>of</strong>ile. AE<br />

were assessed by NCI-CTC v. 3.0. Results: 34 pts (21 males) with a median<br />

age <strong>of</strong> 39 years (range 19-66) received this HDI regimen. PS was 0-1 in<br />

90%. 95% <strong>of</strong> pts received prior doxorubicin, ifosfamide or both, 7 patients<br />

had 2 prior lines. Grade III monophasic pattern was seen in 21 patients,<br />

grade II in 13. 171 cycles were delivered, median number <strong>of</strong> cycles was 4<br />

(2-10). The objective responses were CR 0; PR 15; SD 14; PD 2; and NE 3,<br />

resulting in an ORR <strong>of</strong> 44%. The clinical benefit rate was 85%. Median<br />

PFS: 11.6 months (95% CI: 9-14). Eleven pts were amenable to a<br />

subsequent resection <strong>of</strong> residual disease. Common adverse events included<br />

grade III/IV neutropenia: 47%, febrile neutropenia: 12%, thrombocytopenia:<br />

12%, cystitis: 3%, neurologic: 3%. There was no treatment<br />

related death. Conclusions: HDI demonstrated clinically meaningful efficacy<br />

with manageable toxicity in pts who failed previous standard Ifosfamide<br />

dose. Ifosfamide remains the most active agents in SS and high<br />

dose regimen have to be considered in pts previously treated with<br />

ifosfamide.<br />

10046 General Poster Session (Board #47G), Sun, 8:00 AM-12:00 PM<br />

Rechallenge with ifosfamide-containing regimen as salvage option for<br />

patients with metastatic s<strong>of</strong>t tissue sarcomas progressing after multiple<br />

pretreatments. Presenting Author: Mathias Hoiczyk, Sarcoma Center, West<br />

German Cancer Center, Essen, Germany<br />

Background: Ifosfamide (I) is commonly used in first or second-line<br />

treatment <strong>of</strong> locally advanced or metastatic s<strong>of</strong>t tissue sarcomas (STS). The<br />

clinical efficacy <strong>of</strong> a rechallenge with ifosfamide (IR) or I-containing<br />

regimen (ICR) is unclear. We conducted a retrospective analysis <strong>of</strong> our<br />

institutional database (n�1354) <strong>of</strong> pts who received IR or ICR. Methods:<br />

66 STS pts were identified, who had received more than one I-based<br />

regimen. Ifosfamide retreatment was given in neoadjuvant (n�26), adjuvant<br />

(n�18) or metastatic settings (n�22) and most patient were pretreated<br />

with doxorubicin (D; 94%). PFS was determined from first<br />

administration <strong>of</strong> IR until disease progression, OS from R until last<br />

follow-up or death. ORR (RECIST) was assessed to determine the clinical<br />

benefit rate (CBR) defined as CR, PR plus SD. Covariates were age, gender,<br />

histology, pretreatment, dose intensity, response and toxicity. Results:<br />

Median age at time <strong>of</strong> first I was 45y (range: 17-74, 44% f 56% m). Median<br />

time from diagnosis until first ICR treatment was 4mo. Median time from<br />

first I until IR, which was given as median 3rd-line treatment (2-9 lines),<br />

was 17 mo. Histologies were 12 leiomyo-, 11 lipo- 13 pleomorphic,14<br />

synovial, 4 rhabdomyosarcomas and 12 other subtypes. Pts received a<br />

median <strong>of</strong> I 9.6 g/m²/cycle and a median <strong>of</strong> 4 cycles. I was given as<br />

monotherapy (38%) or in combination (D: 45%, other 17%). Overall<br />

median PFS was 5mo with PFS-rates <strong>of</strong> 63% at 12 and 25% at 24 wks.<br />

Median PFS was 6 mo for ICR and 2 mo for IR. The median OS was 36 mo<br />

from time <strong>of</strong> first diagnosis <strong>of</strong> metastases or locally advanced disease. ORR<br />

was 29% for I/D combination (CBR 70%) and 12 % for I monotherapy<br />

(CBR: 52%). 22 pts received a second IR with a median <strong>of</strong> 2 cycles (27%<br />

PR; CBR:77%; 69% combinations). Most common toxicities for IR/ICR<br />

were neutropenia (42%), encephalopathy (21%) and neutropenic fever<br />

(18%). Dose reductions were necessary in 18% <strong>of</strong> pts and treatment was<br />

stopped in 8% due to toxicity. No significant cardiotoxicity was observed.<br />

Conclusions: Patients with s<strong>of</strong>t tissue sarcomas who had at least disease<br />

stabilization after initial ifosfamide-based therapy may derive substantial<br />

clinical benefit from a rechallenge.<br />

Sarcoma<br />

641s<br />

10045 General Poster Session (Board #47F), Sun, 8:00 AM-12:00 PM<br />

<strong>Clinical</strong> effectiveness <strong>of</strong> recombinant adenovirus human p53 gene combined<br />

with hyperthermia in treatment <strong>of</strong> advanced s<strong>of</strong>t tissue sarcoma.<br />

Presenting Author: Shaowen Xiao, Peking University Cancer Hospital and<br />

Institute, Beijing, China<br />

Background: S<strong>of</strong>t tissue sarcoma in advanced stage usually resist to both<br />

chemo- and radiotherapy. Several studies indicated that rAdp53 can<br />

reverse this resistance and enhance the effectiveness <strong>of</strong> radiotherapy,<br />

chemotherapy and hyperthermia. This study is to evaluate the efficacy and<br />

safety <strong>of</strong> recombinant adenovirus human p53 gene (rAdp53) combined<br />

with hyperthermia and radiation in treatment <strong>of</strong> advanced s<strong>of</strong>t tissue<br />

sarcoma. Methods: From Nov. 2001 to July 2011, 30 patients with<br />

advanced s<strong>of</strong>t sarcoma were enrolled in this study. All cases received<br />

hyperthermia treatment, once a week for total <strong>of</strong> 9 times, and intratumoral<br />

injection <strong>of</strong> rAdp53 at dose <strong>of</strong> 1x1012 virus particles (vp) for 8 times.<br />

Radiation were given 48 hours after the first rAdp53 injection at a dose <strong>of</strong><br />

2Gy/f, five fractions a week for a total dose 16~70Gy /8~35f/2~8w, at an<br />

average <strong>of</strong> 56.3�5.3 Gy. Patients were monitored for response and adverse<br />

events. Results: The median follow-up time was 96 months. Among these<br />

patients, one case (3.3%) achieved complete response (CR), 9 (30%)<br />

cases had a partial response (PR), and 18 (60.0%) cases had a stable<br />

disease (SD). The overall clinical effectiveness rate (CR�PR�SD<br />

� 6months) was 83.3.0% (25/30). One-year survival rate is 56.6%<br />

(17/30), 2-year survival rate is 23.3% (7/30), 3-year survival rate is 10.0%<br />

(3/30), and 5-year survival rate is 6.7% (2/30). Overall median survival<br />

time was 18.1 months and median TTP was 13 months. No significant<br />

adverse events were noted, except for transient fever associated with<br />

rAdp53 administration. Conclusions: rAdp53 combined with hyperthermia<br />

and radiation is effective and safe modalities in treatment <strong>of</strong> advanced s<strong>of</strong>t<br />

tissue sarcoma.<br />

10047 General Poster Session (Board #47H), Sun, 8:00 AM-12:00 PM<br />

Association between body weight and efficacy outcomes during trabectedin<br />

therapy for recurrent advanced s<strong>of</strong>t tissue sarcoma (STS). Presenting<br />

Author: Maurizio D’Incalci, Istituto di Ricerche Farmacologiche, Milan,<br />

Italy<br />

Background: Trabectedin is the first marine-derived antineoplastic drug<br />

approved in Europe for the treatment <strong>of</strong> patients with recurrent advanced<br />

STS. Different studies have implicated tumor-derived cytokines as mediators<br />

<strong>of</strong> cachexia in cancer patients. Preclinical studies have demonstrated<br />

that trabectedin acts on tumor microenvironment particularly reducing<br />

IL-6, which is involved in cancer-induced cachexia (Germano et al. 2011,<br />

Ligouri et al. 2011). Methods: This exploratory retrospective analysis<br />

evaluated the association between body weight increase during treatment<br />

with respect to baseline and efficacy outcomes in 319 adult patients with<br />

recurrent STS. Patients were treated in four phase II trials with trabectedin<br />

at the approved dose <strong>of</strong> 1.5 mg/m2 , given as a 24-hour infusion every 3<br />

weeks. An analysis <strong>of</strong> the concordance between weight increase and<br />

objective response (OR) rate, tumor control (TC) rate (OR�stable disease<br />

lasting �3 months), progression-free survival (PFS) and overall survival<br />

(OS) was performed. Results: Correlation between weight increase and<br />

responses was observed: 68% <strong>of</strong> patients who achieved OR and 70% <strong>of</strong><br />

patients with TC increased their weight for a median <strong>of</strong> 1.2 kg. Remarkably,<br />

PFS (5.1 vs. 1.9 months; p�0.0001, log rank) and OS (21.0 vs. 8.2<br />

months; p�0.0001, log rank) were significantly prolonged in patients who<br />

gained weight during treatment. Six and 12-months Kaplan-Meier PFS<br />

estimates and survival rates at 12, 24 and 36 months were also better in<br />

those patients. Additionally, in Cycle 1-2, a significant relationship<br />

between weight increase and improved median PFS (4.2 vs. 2.2 months;<br />

p�0.0001) and OS (19.4 vs. 9.3 months; p�0.0002) was observed. This<br />

is in concordance with the tendency to gain weight starting from the first<br />

cycle <strong>of</strong> treatment. Conclusions: This analysis suggests that weight gain is<br />

associated with favorable efficacy outcomes when patients are treated with<br />

trabectedin. Further research is needed to assess the prognostic value <strong>of</strong><br />

weight changes as a complementary source <strong>of</strong> evaluation on the long-term<br />

clinical outcomes and impact <strong>of</strong> trabectedin on tumor microenvironment.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


642s Sarcoma<br />

10048 General Poster Session (Board #48A), Sun, 8:00 AM-12:00 PM<br />

Differential gene expression in liposarcoma: Insight into pathways for<br />

dedifferentiation? Presenting Author: Dale Han, H. Lee M<strong>of</strong>fitt Cancer<br />

Canter & Research Institute, Tampa, FL<br />

Background: Liposarcoma (LPS) dedifferentiation signifies conversion to a<br />

clinically aggressive phenotype, but the biologic processes required for this<br />

change have not been determined. We describe differential gene expression<br />

patterns between well-differentiated (WD) and dedifferentiated (DD)<br />

tumors to determine pathways involved in LPS dedifferentiation. Methods:<br />

From 1999 to 2006, 121 fatty tumors were resected at a single institution.<br />

Twenty tumors, consisting <strong>of</strong> atypical lipomatous tumors (ALT), WD LPS or<br />

DD LPS, were randomly selected and clinicopathologic characteristics were<br />

retrospectively reviewed. Gene expression pr<strong>of</strong>iling was performed on<br />

extracted RNA using the Affymetrix GeneChip platform. Differentially<br />

expressed genes were obtained and gene network analysis was done using<br />

GeneGO by MetaCore. Results: Median age was 59 years and 70% <strong>of</strong> cases<br />

were male. WD tumors, consisting <strong>of</strong> 3 ALT and 6 WD LPS, were compared<br />

with 11 DD LPS. After a median follow-up <strong>of</strong> 64 months, 7 patients had<br />

died <strong>of</strong> whom 6 had DD LPS. DD histology was associated with lower overall<br />

survival (p�0.05). Significance Analysis <strong>of</strong> Microarrays for WD tumors vs.<br />

DD LPS using a 0% false discovery rate showed differential expression <strong>of</strong><br />

188 genes. Network analysis <strong>of</strong> genes from WD tumors vs. DD LPS showed<br />

significant (p�0.001) differential regulation <strong>of</strong> glucose-activated transcription<br />

factor ChREBP (carbohydrate response element binding protein), a key<br />

element involved in lipogenesis, gluconeogenesis and glycolysis. There was<br />

also significant differential regulation <strong>of</strong> insulin signaling, PI3K-dependent<br />

and PKA signal transduction pathways and <strong>of</strong> amino acid, fatty acid and<br />

glucose metabolism pathways (p�0.05). These pathways, based on Gene<br />

Ontology cellular processes, mapped to gene networks primarily involved in<br />

lipid metabolism (p�0.05). Conclusions: Differential expression <strong>of</strong> genes<br />

involved in lipid metabolism networks is seen in DD LPS and changes in<br />

lipid metabolism may be associated with dedifferentiation. These differential<br />

gene expression patterns may help identify fatty tumors potentially at<br />

risk for progressing to a malignant or DD state and provide prognostic<br />

factors and therapeutic targets for patients with LPS.<br />

10050 General Poster Session (Board #48C), Sun, 8:00 AM-12:00 PM<br />

Does combined dose intensification radiotherapy improve disease control<br />

in resectable retroperitoneal sarcoma? Long-term results <strong>of</strong> a phase I/II<br />

trial. Presenting Author: Myles J. F. Smith, Department <strong>of</strong> Surgical<br />

Oncology , Toronto, ON, Canada<br />

Background: Late failure is a challenging problem in retroperitoneal<br />

sarcoma (RPS) and reported 10 yr overall survival (OS) rates range from<br />

20-30%. Use <strong>of</strong> preoperative external beam radiotherapy (XRT) in the<br />

management <strong>of</strong> RPS remains controversial. No RCT and very few prospective<br />

trials <strong>of</strong> any type have been completed. We investigated the effects <strong>of</strong><br />

preop XRT plus dose escalation with early postop brachytherapy (BT) on<br />

long term survival and recurrence in RPS. Methods: From 06/96 to 10/00,<br />

40 patients (25 female) with resectable RPS were entered onto a phase I/II<br />

trial <strong>of</strong> preop XRT (50 Gy) plus postop BT (20-25 Gy). As previously<br />

reported, BT to the upper abdomen was associated with significant grade<br />

III-V postop toxicity, and from 12/98 on, BT was applied only to cases<br />

where the “field at risk” excluded the upper abdomen. Kaplan Meier<br />

survival curves were constructed; OS and recurrence free survival (RFS)<br />

were compared by log rank (SPSS 19.0). Results: Median age at study entry<br />

was 58 (38-70) yrs. Twenty nine patients presented to our center with<br />

primary disease (73%), and 22 (55%) had high grade tumors. All patients<br />

had preop XRT and total gross resection, while half (n�19) received BT. As<br />

<strong>of</strong> 12/2011, median follow-up time is 108 mos. For the entire study cohort,<br />

OS at 5 and 10 yrs were 70% and 65%, respectively; RFS at 5 and 10 yrs<br />

were 65% and 58%, respectively. RFS at 5 yrs was reduced in high vs. low<br />

grade RPS (50% vs. 83%, p�0.028), but by 10 yrs. was similar in high and<br />

low grade tumors (50% vs. 67%, p�ns). RFS was reduced in patients who<br />

presented with recurrent vs. primary disease (27% vs. 69% at 10 yrs.,<br />

p�0.018), as was OS (36% vs. 76% at 10 yrs., p�0.034). Neither OS nor<br />

RFS was improved in the cohort <strong>of</strong> patients who received BT compared to<br />

the cohort who did not: at 10 yrs. RFS was 53% �BT and 62% -BT, while<br />

OS was 53% and 76%, respectively, p�ns. Conclusions: In this prospective<br />

study with mature follow-up, long term OS and RFS in patients who<br />

underwent combined preop XRT plus resection <strong>of</strong> RPS compare favorably<br />

with those reported in retrospective institutional and population-based<br />

series. Postoperative BT did not contribute to disease control.<br />

10049 General Poster Session (Board #48B), Sun, 8:00 AM-12:00 PM<br />

Interest <strong>of</strong> tumor to liver ratio 18F-FDG uptake in positron emission<br />

tomography for detection <strong>of</strong> neur<strong>of</strong>ibrosarcoma in neur<strong>of</strong>ibromatosis 1<br />

patients. Presenting Author: Patrick Combemale, Centre Leon Berard,<br />

Lyon, France<br />

Background: Malignant peripheral nerve sheath tumors (MPNSTs) are<br />

difficult to detect in neur<strong>of</strong>ibromatosis 1 (NF1) individuals. The 18F FDG<br />

positron emission tomoscintigraphy (FDG PET) is a useful tool. The<br />

Standard Uptake Value (SUV) is the gold standard but it is subject to<br />

individual variation among patients and medical centers. So we assessed<br />

the interest <strong>of</strong> FDG PET based on a specific tumor to liver uptake ratio (T/L)<br />

instead <strong>of</strong> SUV to find a reproducible and constant cut-<strong>of</strong>f for detecting<br />

malignant tumors. Methods: From 2000 to 2010, we conduct a multi<br />

centric study: all patients with NF1 and presenting clinical signs <strong>of</strong> MPNST<br />

suspicion (increased tumor size, induration or pain) underwent a FDG PET.<br />

Preliminary study estimated a T/L ratio <strong>of</strong> 1.5 as cut-<strong>of</strong>f for malignity.<br />

Based on semi-quantitative analysis <strong>of</strong> FDG PET images, patients were<br />

compared with this cut-<strong>of</strong>f value. Results: 98 patients with 125 tumors<br />

were included. FDG PET evaluation identified 42/125 suspected lesions<br />

with T/L � 1.5; among these 30 MPNST and 12 benign lesions were found.<br />

83/125 tumors were classified as non-suspicious and 82 were actually<br />

benign according to histology or long term follow up. The 1.5 T/L cut <strong>of</strong>f<br />

corresponds to 99 % Negative Predictive Value (NPV) and 71% Positive<br />

(PPV). The positive likelihood ratio (LR) was thus equal to 7, 69 and the<br />

Negative LR to 0, with 97% sensitivity and 87% specificity. When T/L ratio<br />

is � 1.5, MPNST is eliminated with 99% NPV, thus avoiding useless<br />

surgery. When T/L ratio is above 1.5, there is a strong suspicion <strong>of</strong><br />

malignancy. But there is a risk <strong>of</strong> false positivity, which requires discussion<br />

before any therapeutic decision. On the over hand we found no significant<br />

correlation between SUVmax and malignancy. Conclusions: This study<br />

confirms the FDG PET in the detection <strong>of</strong> NF1-associated MPNST. The<br />

tumor/liver ratio with a cut <strong>of</strong>f 1.5 has a very sensitive and specific value to<br />

sort out malignant from benign tumors and thus provides adequate surgery.<br />

More this semi-quantitative analysis method is just as easy as SUV, more<br />

sensitive and more reproducible.<br />

10051 General Poster Session (Board #48D), Sun, 8:00 AM-12:00 PM<br />

Geriatric high-grade s<strong>of</strong>t tissue sarcoma (G-HG STS): An analysis <strong>of</strong> 116<br />

patients (pts) evaluating prognostic factors and clinical outcomes stratified<br />

by histology. Presenting Author: Richard Hong Hui Quek, National Cancer<br />

Centre, Singapore<br />

Background: STS in geriatric pts is not well studied. We evaluated<br />

prognostic factors and clinical outcomes in elderly pts with HG STS.<br />

Methods: Single centre retrospective study. G-HG STS pts defined as age<br />

� 65 yrs seen in our centre from 2002 - 2011 with complete medical<br />

records were identified. Charlson age-comorbidity score was assessed for<br />

each pt. Results: 116 pts from 4 most common HG STS histo-types<br />

representing 69% <strong>of</strong> pts in the geriatric STS cohort were analysed;<br />

leiomyosarcoma (LMS, 14%), non well-differentiated liposarcoma (nWD-<br />

LPS, 9%), angiosarcoma (AS, 30%), and undifferentiated pleomorphic<br />

sarcoma (UPS, 47%). Median age was 72 yrs, 81% presented with<br />

localised disease; <strong>of</strong> 78% <strong>of</strong> these localised pts who had curative surgery,<br />

49% received adjuvant therapy, <strong>of</strong> whom 92% had radiotherapy (RT) only.<br />

AS arises more commonly from the head/neck region (p�0.001) and fewer<br />

receive curative surgery (p�0.006). In 43 pts who had metastases either at<br />

diagnosis or relapse, 33% received first-line palliative chemotherapy with a<br />

response rate <strong>of</strong> 27% in evaluable pts. At a median follow-up <strong>of</strong> 15.8 mths,<br />

overall survival (OS) for the entire cohort was 25.1 mths, 30.5 vs 3.9 mths<br />

in pts presenting with localised vs metastatic disease respectively<br />

(p�0.0001). In pts who had curative surgery for localised disease, overall<br />

relapse-free survival (RFS) was 17.7mths; 26.8 mths vs 16.0 mths vs 7.3<br />

mths vs 12.5 mths in LMS, nWD-LPS, AS and UPS respectively. In<br />

univariate analysis, adjuvant RT, non-head/neck primary and sarcoma<br />

subtype were associated with improved RFS. In multivariate analysis,<br />

adjuvant RT (p�0.001), sarcoma subtypes AS (p�0.011) and UPS<br />

(0.012) vs LMS remained significant. In pts with metastatic HG STS either<br />

at diagnosis or relapse, overall median OS was 5.9 mths; 5.9 mths (LMS),<br />

30.5 mths (nWD-LPS), 6.4 mths (AS) and 4.3 mths (UPS). In univariate<br />

analysis, presence <strong>of</strong> bone metastases was significantly associated with<br />

inferior OS (p�0.0029). Charlson score did not correlate with RFS or OS.<br />

Conclusions: Prognosis <strong>of</strong> G-HG STS appears poor particularly in AS and<br />

UPS. Adjuvant RT improves outcomes in this group <strong>of</strong> pts and should not be<br />

omitted based on age alone.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


10052 General Poster Session (Board #48E), Sun, 8:00 AM-12:00 PM<br />

Phase II study <strong>of</strong> neoadjuvant high-dose ifosfamide with concurrent<br />

radiotherapy followed by surgical resection in high-risk s<strong>of</strong>t tissue sarcoma:<br />

A Spanish Group for Research on Sarcomas (GEIS) study. Presenting<br />

Author: Xavier Garcia del Muro, Instituto Catalan Oncologia, Barcelona,<br />

Spain<br />

Background: High-dose ifosfamide (HDI) has shown promising activity as<br />

single agent in first-line chemotherapy for advanced s<strong>of</strong>t tissue sarcoma<br />

(STS). The purpose <strong>of</strong> this study was to assess the activity and toxicity <strong>of</strong> a<br />

preoperative doxorubicin-free regimen <strong>of</strong> HDI given concurrently with<br />

radiotherapy (RT) and followed by surgery in patients (pts) with localized<br />

high-risk STS. Methods: Pts with localized � 5 cm grade 2-3 and deep STS<br />

<strong>of</strong> the extremities and trunk wall, �65 years, no prior chemotherapy and<br />

ECOG PS 0-1 were enrolled in this multicenter phase II study. Pts received<br />

3 cycles <strong>of</strong> preoperative HDI at a dose <strong>of</strong> 12 gr/m2 by continuous infusion<br />

over 5 days every 3 weeks, with mesna and prophylactic GCSF support, and<br />

concomitant external beam RT to a total dose <strong>of</strong> 50 Gy, followed by surgical<br />

resection. Postoperatively, pts with pathological response received 2 cycles<br />

<strong>of</strong> HDI and those with positive surgical margins 16 Gy <strong>of</strong> RT. The primary<br />

study endpoint was pathologic response (� 95% pathologic necrosis). A<br />

Simon 2-stage design (response rate P0�15%, P1�35%, ��0.10,<br />

��0.10) required at least 2 responses in the first 17 pts to expand to a<br />

second cohort, and 7/32 responses to be considered <strong>of</strong> positive. Results:<br />

From March 08 to December 10, 34 pts were included. Two pts were<br />

ineligible. Median age was 54 (18-65). Tumor location was extremity (28<br />

pts) and trunk wall (4). Preoperative planned treatment was completed in<br />

87.5% pts. HDI was completed in 87.5% pts while RT in 94% pts. Grade<br />

3-4 toxicities included neutropenic fever (3 pts), anemia (4), asthenia (2),<br />

infection (2) and radiation dermatitis (2). 31 pts underwent surgery: 27<br />

were R0 resections, <strong>of</strong> which 2 were amputations, and 4 were R1<br />

resections. Pathologic response was � 95% necrosis in 9 pts (28%) and<br />

50%-94% necrosis in 12 pts. After a median follow-up <strong>of</strong> 21 months,<br />

estimated 2-year rates for disease-free survival and overall survival were<br />

58% (95%CI, 40%-77%) and 77% (95%CI, 61%-93%), respectively.<br />

Conclusions: Preoperative treatment with HDI given concurrently with RT in<br />

pts with high-risk STS is feasible and safe, yielding promising pathologic<br />

response rates.<br />

10054 General Poster Session (Board #48G), Sun, 8:00 AM-12:00 PM<br />

Early response to neoadjuvant chemotherapy (NAC) in combination with<br />

regional hyperthermia (RHT) to predict long-term survival for adult-type<br />

high-risk s<strong>of</strong>t tissue sarcoma (HR-STS): Results <strong>of</strong> the EORTC-ESHO<br />

intergroup phase III study. Presenting Author: Rolf D. Issels, Universitaet<br />

München, Grosshadern, Munich, Germany<br />

Background: A randomized phase III completed trial showed that RHT<br />

added to NAC was beneficial in terms <strong>of</strong> local progression-free (LPFS), and<br />

disease-free (DFS) survival. Overall survival (OS) was improved in patients<br />

(pts) who completed the preoperative induction therapy with RHT (Lancet<br />

Oncol 2010). Here we analyzed both the radiographic (RR) and histopathologic<br />

(HR) response as early predictors for survival. Methods: 341 pts were<br />

randomized to receive 4 cycles <strong>of</strong> NAC � RHT (169 pts) or NAC alone (172<br />

pts) as induction therapy. RR (CR/PR vs NC/PD) and HR (�75% vs �75%<br />

necrosis) were used to define responder vs non-responder. Predictive<br />

impact <strong>of</strong> response on LPFS, DFS, DDFS (distant disease-free survival) and<br />

OS was evaluated by intention-to-treat using Kaplan-Meier estimates and<br />

the log-rank test. Stratified (surgery before study entry yes/no; extremity vs<br />

non-extremity tumors) multivariate analyses were carried out by Cox<br />

regression. Results: Early response in pts with measurable disease was<br />

performed in 238 pts (103 pts not evaluable because <strong>of</strong> surgery before<br />

study entry). In the NAC�RHT group (114 pts) response rate was 49.1%<br />

(56 responders: RR: 18; HR: 22; RR � HR: 16) and substantially higher<br />

(p�0.001) compared to the NAC alone group (124 pts) which was 26,6%<br />

(33 responders: RR: 7; HR: 17; RR � HR: 9). In the NAC � RHT group,<br />

tumor response was associated with improved DFS (HR 0.58 CI 0.36-0.95;<br />

p�0.031) and OS (HR 0.50 CI 0.27-0.90; p�0.020) but not LPFS (HR<br />

0.91 CI 0.49-1.71; p�0.78). For responders, OS median time was � 120<br />

months vs 33 months for non-responders. For the entire NAC � RHT group<br />

(169 pts, including pts with surgery before study entry) response remained<br />

predictive for better OS (HR 0.54 CI 0.32-0.94; p�0.028) and was also<br />

associated with better DDFS (HR 0.47 CI 0.27-0.83; p�0.009).<br />

Conclusions: Adding RHT to NAC as induction therapy compared to NAC<br />

alone leads to significantly higher early response in almost half <strong>of</strong> the pts<br />

classified as responders which translates in better OS and prevention <strong>of</strong><br />

distant metastases.<br />

Sarcoma<br />

643s<br />

10053 General Poster Session (Board #48F), Sun, 8:00 AM-12:00 PM<br />

The Italian Rare Cancer Network. Presenting Author: Roberta Sanfilippo,<br />

Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy<br />

Background: Rare cancers (RC) are a challenge in terms <strong>of</strong> quality <strong>of</strong> care,<br />

access to health resources and clinical research. The Italian Rare Cancer<br />

Network (RTR: “Rete Tumori Rari”) is a clinical collaborative effort to<br />

improve quality <strong>of</strong> care in adult rare solid cancers in Italy. RTR enables<br />

institutions to share clinical cases and to rationalize access to distant<br />

reference centers minimizing patient migration. It indirectly promotes<br />

collaborative clinical research by encouraging accrual into clinical trials<br />

and supporting observational studies. Methods: RTR includes 150 oncology<br />

institutions across Italy. <strong>Clinical</strong> cases are shared asynchronously over a<br />

secure Web resource. Data, images and transactions are stored in an online<br />

clinical record. Patients are shared: 1. �logically”, when they are dealt with<br />

following common clinical practice guidelines; 2. �virtually”, when they are<br />

discussed over the network between two or more centers; 3: �physically�,<br />

when they are referred to an excellence center for a specific treatment<br />

modality. Pathology review is arranged through transferal <strong>of</strong> paraffinembedded<br />

specimens and upload <strong>of</strong> consultations. While it was chosen not<br />

to implement telepathology facilities, a teleradiology resource is now<br />

available. Results: From 2003 to 2011, more than 5,000 rare cancers<br />

cases (mostly sarcomas) have been uploaded. More than 1,300 teleconsultations<br />

have been delivered, while more than 1,000 patients moved across<br />

the network during their experience <strong>of</strong> disease. 700 cases were reviewed<br />

pathologically: amongst 365 cases originally diagnosed as s<strong>of</strong>t tissues<br />

sarcomas up to 2010, treatment-relevant discordances were recorded in<br />

more than one third. An observational prospective study on gastrointestinal<br />

stromal tumors was done, enrolling 800 patients. An original paper<br />

documenting the activity <strong>of</strong> a drug in a highly specific sarcoma subgroup<br />

was published. Conclusions: <strong>Clinical</strong> asynchronous online collaboration on<br />

RC is feasible through a Web-based secure environment and proved the<br />

most practical way <strong>of</strong> clinical distant sharing. Pathologic review was a<br />

crucial network service, with a special added value in RC. Retrospective<br />

and prospective observational studies, and unplanned observations in very<br />

rare cases, were an interesting by-product.<br />

10055 General Poster Session (Board #48H), Sun, 8:00 AM-12:00 PM<br />

Delay in diagnosis and treatment <strong>of</strong> s<strong>of</strong>t tissue sarcomas (STS): Causes <strong>of</strong><br />

late intervention and their role in prognosis—A prospective, multidisciplinary<br />

group study. Presenting Author: Alessandro Comandone, Piedmontese<br />

Group for Sarcomas/Italian Sarcoma Group, Torino, Italy<br />

Background: STS are 1% <strong>of</strong> malignant tumors in adults. Rarity, heterogeneity<br />

in presentation, low expertise in primary care physicians (PCP) or in<br />

general hospitals, organisation problems in specialized centers may cause<br />

a delay in both diagnosis and treatment. Aim <strong>of</strong> this study is to acknowledge<br />

the barriers to optimal care and the consequences <strong>of</strong> the delay on<br />

prognosis. Methods: Patients with STS <strong>of</strong> the extremities, trunk, retroperitoneum<br />

treated and followed from 1999 to 2011 by the same multidisciplinary<br />

group were included. Time and pattern <strong>of</strong> symptoms onset,<br />

anatomic site, tumor volume, patients’ age, gender and home, interval<br />

between diagnosis and surgical treatment or neoadjuvant chemotherapy;<br />

time to start adjuvant RT or CT were considered in a univariate -<br />

multivariate analysis. Results: 449 adult patient (53% F, 47% M, median<br />

age 55 years) were followed for a median time <strong>of</strong> 116.38 months. 65.7% <strong>of</strong><br />

STS were at the extremities, 17.6% retroperitoneal, 16.7% at the trunk<br />

wall. Median volume at diagnosis was 8 cm for trunk and extremities; 15<br />

cm for retroperitoneum. Commonest histologies: liposarcoma. 18.2%;<br />

leiomyo 16.8%; mix<strong>of</strong>ibro 13.6%. Increasing mass, pain, and abdominal<br />

disconfort were the main revealing signs <strong>of</strong> diseases. Median time <strong>of</strong> delay<br />

were: from onset <strong>of</strong> symptom to first medical visit 68 days for trunk and<br />

extremities, 82 for retroperitoneum; 104 days from symptoms to histological<br />

diagnosis; 129 days from symptoms to start <strong>of</strong> therapy. Time to surgery<br />

after definitive diagnosis was 12 days in extremities and 21 in abdomen.<br />

Adjuvant CT started 22 days after surgery for extremities, 25 in trunk, 35 in<br />

retroperitoneum. RT initiated after 78 days. Longer delay in treatment lead<br />

to worse prognosis: MS 89.95 months if delay was � 3 months; 190.40<br />

months if wait was � 3 months (p 0.007). Conclusions: Low self<br />

consciousness <strong>of</strong> the patient; misdiagnosis or inadequate approach in<br />

general hospitals; late referral to specialized centres are 75% <strong>of</strong> the cause<br />

<strong>of</strong> wasted time. Organization problems at the referral Centre concur for<br />

25% <strong>of</strong> delay. Guidelines implementation and educational programme<br />

among general population and PCP are necessary.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


644s Sarcoma<br />

10056 General Poster Session (Board #49A), Sun, 8:00 AM-12:00 PM<br />

Prospective web-based collection <strong>of</strong> sarcoma cases diagnosed and treated<br />

in France: Experience <strong>of</strong> the NetSarc network <strong>of</strong> the French Sarcoma Tumor<br />

Boards. Presenting Author: Francoise Ducimetiere, Centre Léon Berard,<br />

Lyon, France<br />

Background: The management <strong>of</strong> sarcomas, as for all rare tumors, is<br />

hampered by the limited dissemination <strong>of</strong> medical knowledge. Management<br />

within a specialized multidisciplinary sarcoma tumor board (STB)<br />

improves compliance to clinical practice guidelines. Since 2010, a<br />

national network with a dedicated system for referral, gathering the 26<br />

regional STBs, is supported by the French NCI. Its objectives are to monitor<br />

the management <strong>of</strong> sarcomas and give equal access to expertise and<br />

innovative treatments to all patients with sarcoma. We report here the first<br />

experience <strong>of</strong> a web-based data collection from these STBs. Methods: The<br />

expected incidence <strong>of</strong> sarcoma in France is 6.1/105 /year, ie 4000 new<br />

cases per year. A website collecting all files discussed in the 26 STBs was<br />

generated from Jan 2010 (www.netsarc.org). The prospective collection <strong>of</strong><br />

clinical data, STB decision and patient follow-up was implemented in the<br />

national database (40 items). Results: 7472 patients were accrued during<br />

2 years, 6202 (83%) had sarcoma, 2860 (46%) were new cases,<br />

representing 36% <strong>of</strong> expected incident cases. The number <strong>of</strong> new cases<br />

discussed in STB increased between 2010 and 2011 (�38%). Patients<br />

were referred to the STB (2010-2011; % <strong>of</strong> cases) prior 1) to biopsy<br />

(9%-15%), 2) to first surgery (34%-39%), 3) to adjuvant treatment<br />

(35%-34%), after treatment (14%-8%) and at time <strong>of</strong> relapse (8%-4%).<br />

15% had a metastases at initial diagnosis. The R0�R1 resection rates for<br />

primary surgery were 55% vs 42% within vs outside the STB centers <strong>of</strong><br />

NetSarc. 8% <strong>of</strong> declared patients were proposed to inclusion into a clinical<br />

trial. Conclusions: This web-based tool enabled the description <strong>of</strong> the<br />

activity <strong>of</strong> the STB at the national level. <strong>Clinical</strong> practices improved<br />

between 2010 and 2011, but 64% <strong>of</strong> new cases <strong>of</strong> sarcoma were not<br />

discussed in the referent STB prior to surgery. The yearly follow-up <strong>of</strong> the<br />

database will help to monitor evolution <strong>of</strong> clinical practices.<br />

10058 General Poster Session (Board #49C), Sun, 8:00 AM-12:00 PM<br />

Neoadjuvant chemoradiotherapy for patients with high-risk extremity and truncal<br />

sarcomas: A 10-year follow-up study. Presenting Author: Nicole J. Look Hong,<br />

Massachusetts General Hospital, Boston, MA<br />

Background: Patients with high-risk extremity and truncal s<strong>of</strong>t tissue sarcomas<br />

(STS) are at considerable risk for recurrence. A regimen <strong>of</strong> preoperative mesna,<br />

doxorubucin hydrochloride (Adriamycin), ifosfamide, and dacarbazine (MAID),<br />

interdigitated with radiotherapy (RT), followed by resection and postoperative<br />

chemotherapy with or without RT, has demonstrated high rates <strong>of</strong> local and<br />

distant control. The goal <strong>of</strong> this series is to study outcomes <strong>of</strong> our most recent<br />

cohort <strong>of</strong> patients treated on this regimen. Methods: We retrospectively reviewed<br />

records <strong>of</strong> 55 consecutive patients with STS <strong>of</strong> the extremity or superficial trunk<br />

who completed all phases <strong>of</strong> treatment at our institution from January 2000–<br />

July 2011. Clinicopathologic characteristics and patient outcomes were analyzed.<br />

Results: Fifty-five patients were analyzed and had a median age <strong>of</strong> 53 years<br />

(range 18-73). The median tumor size was 10.1cm (range 2.5-35.5 cm).<br />

Twenty-seven (49%) and 28 (51%) patients had grade 2 and 3 tumors,<br />

respectively. Margins were negative in 49 (89%) patients, and positive in 6<br />

(11%) patients. At a median follow-up <strong>of</strong> 43 months, there were 7 (13%)<br />

locoregional, and 17 (31%) distant recurrences. The local and distant 5-year<br />

recurrence-free survival (RFS) rates were 92% and 64%, respectively. The<br />

5-year overall (OS) and disease-specific survival rates were 86% and 89%,<br />

respectively. There were no treatment-related deaths or secondary myelodysplasias.<br />

There were no significant predictors <strong>of</strong> OS or RFS. Conclusions: Outcomes <strong>of</strong><br />

a contemporary cohort <strong>of</strong> patients with extremity and truncal STS treated with an<br />

intense regimen <strong>of</strong> neoadjuvant chemoradiotherapy and surgery are consistent<br />

with previous reports from our institution and better than reports <strong>of</strong> chemotherapy<br />

alone.<br />

Comparison <strong>of</strong> interdigitated chemoradiotherapy with adjuvant/neoadjuvant<br />

chemotherapy.<br />

N 5-year OS 5-year distant RFS<br />

Current (2012) 1<br />

55 86 % 64%<br />

Gotzak et al. (2001) 2<br />

134 65% -<br />

DeLaney et al. (2003) 1<br />

48 87% 75%<br />

Sarcoma Meta-Analysis<br />

Collaboration (2007) 3<br />

1568 ~65% ~ 60%<br />

Radiation Therapy Oncology Group trial 9514,<br />

Kraybill et al. (2010) 1<br />

64 71% 64%<br />

Mullen et al. (2011) 1<br />

48 84% 80%<br />

1 2 3<br />

MAID chemoradiotherapy. Neoadjuvant chemotherapy. Adjuvant chemotherapy.<br />

10057 General Poster Session (Board #49B), Sun, 8:00 AM-12:00 PM<br />

Advanced s<strong>of</strong>t-tissue sarcoma in patients over age 75: Patterns <strong>of</strong> care and<br />

survival. Presenting Author: Delphine Garbay Decoopman, Institut Bergonié,<br />

Bordeaux, France<br />

Background: There are no data regarding the management and the outcome<br />

<strong>of</strong> elderly patients (pts) diagnosed with advanced s<strong>of</strong>t-tissue sarcoma<br />

(STS). Methods: The charts <strong>of</strong> pts aged � 75 years and diagnosed with<br />

metastatic and/or unresectable STS between 1990 and 2011 at Memorial<br />

Sloan Kettering (New York) and Institut Bergonié (Bordeaux, France) were<br />

reviewed. Results: 189pts were included.Median age was 79 years (range<br />

75-93).160 pts (84.5%) had metastatic disease. The most frequent<br />

histological subtype was leiomyosarcoma (n�57, 30%). The median<br />

age-adjusted Charlson comorbidity score was 10 (range 5-17). 120 pts<br />

(63.5%) received systemic therapy whereas 69 pts (36.5%) were managed<br />

with best supportive care (BSC) only. Pts who received BSC only were more<br />

likely � 80 years old (58% versus 38%, p�0.01) and with performance<br />

status (PS) � 2 (35% versus 14%, p�0.01). Single agent and combination<br />

therapy were delivered in 87 (72.5%) and 33 (27.5%) cases respectively.<br />

67 pts (56%) received a 1st-line anthracycline-containing regimen. The<br />

median progression-free survival <strong>of</strong> pts treated with systemic therapy was 4<br />

months (95% CI: 2.5-5.5). 21 pts (17.5%) had to stop 1st line treatment<br />

because <strong>of</strong> toxicity. On multivariate analysis, age � 80 years, PS � 2, and<br />

single-agent therapy were significantly associated with poor PFS. 51 pts<br />

received one or more further lines <strong>of</strong> therapies. At the time <strong>of</strong> analysis, 170<br />

pts (90%) had died. 96% <strong>of</strong> deaths were related to the disease. The median<br />

OS for the entire cohort <strong>of</strong> pts was 9.6 months (95% CI: 7.3-12). The<br />

1-year and 2-year OS rates were 43% (95% CI: 36–50), and 22% (95% CI:<br />

16–28), respectively. The median OS <strong>of</strong> pts managed with systemic<br />

therapy and BSC were 12.5 months (95% CI: 8.4-16.6) and 5.1 months<br />

(95% CI: 3.7-6.5), respectively (p�0.02). However, on multivariate<br />

analysis, PS � 2 and age-adjusted Charlson score � 10 were the sole<br />

independent factors significantly associated with OS. Conclusions: A high<br />

proportion <strong>of</strong> elderly pts with advanced STS were considered unfit for<br />

chemotherapy. The OS <strong>of</strong> STS pts � 75 years who were managed with<br />

systemic therapy seems similar to that <strong>of</strong> younger pts. Further efforts are<br />

needed to better define the optimal care for fit and unfit elderly pts with<br />

advanced STS.<br />

10059 General Poster Session (Board #49D), Sun, 8:00 AM-12:00 PM<br />

Risk factors leading to grading changes within 300 low-grade s<strong>of</strong>t tissue<br />

sarcomas. Presenting Author: Tanja Koshrawipour, BG University Hospital<br />

Plastic Surgery, Bochum, Germany<br />

Background: S<strong>of</strong>t tissue sarcomas (STS) are rare, mesenchymal derived<br />

tumors. Based on malignancy, three grades are distinguished with G1<br />

equaling low grade, G2 intermediate grade and G3 high grade, respectively.<br />

Studies have shown that some G1 tumors which underwent complete<br />

surgical resection reoccurred as lesions with a grading shift from G1 to<br />

higher malignancy. This grading change is referred to as up grading.<br />

Patients with grading changes were thoroughly examined in our study. Our<br />

aim was to find possible risk factors for up gradings, such as age,<br />

localization <strong>of</strong> tumor and tumor type. Methods: This retrospective case<br />

control study evaluated 333 Patients, who, between 1996 to 2011 were<br />

treated for G1 STS at Bergmannsheil Bochum, university clinic. These<br />

patients received complete initial surgical resection. Among our 333<br />

patients, 9.9% (n�33) developed up gradings <strong>of</strong> their STS. These were<br />

then reviewed more closely in the aim <strong>of</strong> finding possible risk factors. We<br />

did not exclude any patients from our data or deliberately pick any grading<br />

changers and regard our collective as randomly sampled. The processed<br />

data includes age, gender, tumor type, tumor localization, occurrence <strong>of</strong><br />

recidive and grading change. Results: Patients with up gradings were found<br />

to have a higher mean age <strong>of</strong> 4, 5 years than reference collective. The tumor<br />

type has a strong, statistically significant effect on grading change as<br />

patients with fibrosarcomas have a threefold risk <strong>of</strong> developing up gradings<br />

when compared to patients with other G1 STS. Conclusions: Our results<br />

indicate that age and tumor type play the key role in the development <strong>of</strong> up<br />

gradings in low malignant STS .Patients aged 60 and above diagnosed with<br />

fibrosarcomas are at a 3 times higher risk <strong>of</strong> developing a grading change as<br />

opposed to patients younger than 60 with other low grade STS than<br />

fibrosarcoma. We discussed the significance <strong>of</strong> these risk factors and<br />

argued whether, in addition to wide tumor resection, a short term<br />

postoperative radiotherapy should be applied for these patients to improve<br />

therapeutical outcome.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


10060 General Poster Session (Board #49E), Sun, 8:00 AM-12:00 PM<br />

Treatment outcomes <strong>of</strong> primary mediastinal and intrathoracic great vessels<br />

sarcoma: Royal Marsden Hospital experience. Presenting Author: Shir<br />

Kiong Lu, Sarcoma Unit, Royal Marsden NHS Foundation Trust, London,<br />

United Kingdom<br />

Background: Sarcoma <strong>of</strong> the mediastinum and great vessels remain a rare<br />

subgroup <strong>of</strong> s<strong>of</strong>t tissue sarcoma and prognosis remains poor with patients<br />

(pts) <strong>of</strong>ten presenting with advanced disease. There is no standard<br />

treatment due to paucity <strong>of</strong> data hence its management relies on case<br />

series. To date this is the largest series <strong>of</strong> pts reported. Methods:<br />

Retrospective analysis <strong>of</strong> pts identified anonymously from a prospectively<br />

maintained database <strong>of</strong> the sarcoma unit. Results: 46 pts were included in<br />

the analysis. The median age at diagnosis was 45.9 with a male: female<br />

ratio <strong>of</strong> 1.2: 1. 60% <strong>of</strong> pts presented with localised disease. The most<br />

common histological subtype was spindle cell sarcoma followed by angiosarcoma.<br />

The heart (n�21), majority in the atria, was the commonest<br />

primary site. 28 pts presented with localised disease <strong>of</strong> which 21 had<br />

surgery and 16 received chemotherapy (CT). 4 pts received multi-modality<br />

treatment. 83% <strong>of</strong> pts with metastases had CT. 6 <strong>of</strong> them attained stable<br />

disease, 1 had partial response and 4 (Ewing’s, rhabdomyosarcoma and<br />

angiosarcoma) had complete response. 31 pts developed disease recurrence<br />

<strong>of</strong> which 58% had initial localised disease. 6 pts had metastectomy,<br />

19 received palliative CT and 11 had palliative radiotherapy (RT). RT <strong>of</strong><br />

50-60Gy to the heart or mediastinum was delivered to 61% <strong>of</strong> pts with<br />

initial localized disease and 33% <strong>of</strong> pts with metastases. 2 pts had<br />

documented post-treatment cardiac complication. After a median follow up<br />

<strong>of</strong> 2.16 years, the median event-free survival was 0.97 years (95%CI<br />

0.65-1.30), median overall survival (OS) was 1.7 years (95% CI 0.70-<br />

2.71) and 5 year OS was 17.9%. Conclusions: The optimal management in<br />

this disease is unclear. In this study, surgery was frequently implemented<br />

in both localised and metastatic settings but challenges remain in<br />

achieving clear margins. Chemotherapy has a role and it is key to identify<br />

chemosensitive subtypes. RT is <strong>of</strong>fered as an adjunct following suboptimal<br />

resection. Despite aggressive treatments, this group <strong>of</strong> young pts had high<br />

recurrence rate and poor prognosis. Multi-modality treatment may have a<br />

role in selected pts but its efficacy and long-term cardiac sequelae need to<br />

be evaluated prospectively.<br />

10062 General Poster Session (Board #49G), Sun, 8:00 AM-12:00 PM<br />

Rechallenge with trabectedin in patients with locally advanced or metastatic<br />

s<strong>of</strong>t tissue sarcoma following drug holiday: The experience <strong>of</strong> the<br />

French Sarcoma Group (FSG). Presenting Author: Esma Saada, Centre<br />

Antoine Lacassagne, Nice, France<br />

Background: Trabectedin (T) is a marine-derived alkaloid used to treat<br />

advanced s<strong>of</strong>t tissue sarcomas (STS) after ifosfamide and/or anthracyclins<br />

failure. Since then, the FSG evaluated the clinical benefit in readministrating<br />

T after an initial hold, either medically indicated or upon<br />

patient’s request. Methods: Following an online request, clinical and<br />

histopathological data were collected from six centers <strong>of</strong> the FSG who<br />

declared to have rechallenged patients. Baseline data were collected and<br />

analyze will be used. Results: From 1999 to 2011, 49 pts with T drug<br />

holiday have been identified (26 male/ 23 female), with a median age <strong>of</strong> 50<br />

y [23-75]. Most frequent histotypes were: myxoid liposarcoma (18,<br />

36.7%), leiomyosarcomas (13, 26.5 %) and well-differentiated/dedifferentiated<br />

liposarcoma (9, 18%). WHO grade were 1 in 14 (29%), 2 in 19<br />

(39%) and 3 in 5 (10%) pts respectively. Patients who had a maximum <strong>of</strong><br />

2, 3 or 4 therapeutic sequences (TS) with T (drug-holiday and rechallenge)<br />

were 41/49 ,7/49 and 1/49 respectively. Median number <strong>of</strong> cycles for 1, 2,<br />

3 and 4 TS were 7 [3-21], 6 [2-30], 6 [2-9] and 6. Median total number <strong>of</strong><br />

cycles was 15 [6-43]. Median duration <strong>of</strong> drug-holiday for 1, 2 and 3 TS<br />

were 11 [3-91], 7 [2-29] and 4 months [1-5]. Grade 3-4 toxicities<br />

incidence decreased with the number <strong>of</strong> TS (occurred in 36%, 29%, 14%<br />

and 0% <strong>of</strong> pts with 1, 2, 3 and 4 TS) as well as mean T dose per cycle (1.3<br />

mg/m², 1.2 mg/m², 1.1 mg/m² and 1.1 mg/m² for TS 1, 2, 3, 4). Efficacy<br />

decreased with number <strong>of</strong> TS (Number <strong>of</strong> CR/PR/SD/PD were 1 (2%)/15<br />

(31%)/33 (67%)/0 for TS1; 0/4 (8%)/29 (59%)/16 (3%) for TS2; 0/1<br />

(14%)/2 (29%)/4 (57%) for TS3 and 0/0/0/1(100%) for TS4). Median<br />

overall survival was 5.0 y [2.7-7.3] since T introduction, and 1.5 y<br />

[0.1-4.8], 0.8 y [0.5-1.3] and 0.6 y following 2nd , 3rd and 4th T<br />

reintroduction respectively. Objective response after TS2 were seen in 4<br />

cases <strong>of</strong> grade 1 sarcomas. Conclusions: Due to the lack <strong>of</strong> cumulative<br />

toxicities over time with T, its rechallenging in responding patients to T (no<br />

progression under T) have to be considered in advanced STS.<br />

Sarcoma<br />

645s<br />

10061 General Poster Session (Board #49F), Sun, 8:00 AM-12:00 PM<br />

Drug treatment (tx) patterns and survival among patients (pts) with metastatic or<br />

recurrent s<strong>of</strong>t tissue sarcoma (m/rSTS) refractory to at least one prior therapy.<br />

Presenting Author: Michael Wagner, Dana-Farber Cancer Institute/Harvard<br />

Medical School, Boston, MA<br />

Background: Limited clinical data on real-world practice patterns are available for<br />

care <strong>of</strong> pts with m/rSTS. This study evaluated drug tx patterns and outcomes in<br />

m/rSTS pts following failure <strong>of</strong> prior chemotherapy based on data from a U.S.<br />

referral academic cancer center. Methods: Data from medical records consented<br />

for research use were retrospectively reviewed at Dana-Farber Cancer Institute<br />

for pts with m/rSTS who were �18 years, commenced 2nd-line systemic therapy<br />

between 1/1/2000 and 2/4/2011, had confirmed disease progression on or after<br />

1st-line therapy and had �3 months <strong>of</strong> observation. Pts were excluded if they<br />

had been diagnosed with gastrointestinal stromal tumor, bone sarcoma or<br />

dermat<strong>of</strong>ibrosarcoma protuberans, or had ever been treated with pazopanib.<br />

Histologic subtype, tx type and duration, tx modifications (e.g., discontinuation)<br />

and reasons for modifications were examined. Overall survival, time to progression<br />

and clinician-assessed tumor response were collected. Results: Study pts<br />

had leiomyosarcoma (n�50), synovial cell sarcoma (n�7), liposarcoma (n�5)<br />

and other histologic subtypes (n�39). These 101 pts received an average 4 lines<br />

<strong>of</strong> tx (maximum�10 lines). Wide variations were noted in each line <strong>of</strong> tx (Table)<br />

with no consistent nor dominant regimens. Median 2nd-line tx duration was 4.1<br />

months (95%CI 2.9-5.0). Among 98 pts who discontinued 2nd-line tx, 72<br />

(73%) did so due to progressive disease, 10 due to “completion” <strong>of</strong> planned tx,<br />

and 6 to adverse events. Median progression-free survival from initiation <strong>of</strong><br />

2nd-line tx varied across regimens from 2.0 to 6.5 months (overall median 5.5<br />

months). Conclusions: The wide variation in �2nd-line tx confirms there is no<br />

single “standard” <strong>of</strong> care for m/rSTS. The majority <strong>of</strong> pts discontinued 2nd-line<br />

tx due to progressive disease and <strong>of</strong>ten received additional lines <strong>of</strong> tx, indicating<br />

frequent physician and pt switching <strong>of</strong> tx.<br />

Number pts receiving<br />

Line <strong>of</strong> AnthracyclineGemcitabine- Alkylating Angio-genesis<br />

therapy basedbased agent inhibitor Trabectedin Total<br />

Taxanebased<br />

Other<br />

txt<br />

1st 44 29 9 5 4 0 10 101<br />

2nd 26 28 12 7 13 3 12 101<br />

3rd 12 12 15 12 16 1 9 77<br />

4th 7 5 13 10 9 1 3 48<br />

5th 0 5 12 5 8 0 2 32<br />

6th 2 2 8 3 2 0 1 18<br />

10063 General Poster Session (Board #49H), Sun, 8:00 AM-12:00 PM<br />

Bone metastases in s<strong>of</strong>t tissue sarcoma patients: A survey <strong>of</strong> natural,<br />

prognostic value, and treatment. Presenting Author: Bruno Vincenzi,<br />

Università Campus Bio-Medico, Rome, Italy<br />

Background: Given the limited data currently available, we surveyed the<br />

natural history <strong>of</strong> bone metastases in patients affected by s<strong>of</strong>t tissue<br />

sarcoma (STS). Methods: This retrospective, multicenter, observational<br />

study evaluated data from 135 patients with STS metastatic to the bone<br />

who presented between 2001 and 2011. For all patients, we recorded the<br />

primary tumor histological subtype, bone metastases characteristics (onset,<br />

site), type <strong>of</strong> treatment received (surgery, radiotherapy, zoledronic<br />

acid), type and frequency <strong>of</strong> skeletal related events (SRE) and disease<br />

outcome. Results: The most represented histological subtypes among the<br />

enrolled patients were leiomyosarcoma (27%), angiosarcoma (13%) and<br />

spindle cell sarcoma (8%). The spine was the most common site for bone<br />

involvement (51%), followed by hip/pelvis (20%), long bones (15%) and<br />

other sites (14%). In 27% <strong>of</strong> cases, bone metastases were present at the<br />

time <strong>of</strong> diagnosis. Fiftyfour patients (40%) developed SREs and the<br />

median time to first SRE (if developed) was 4 months. The most common<br />

SRE was the need for radiotherapy occurring in 28% <strong>of</strong> patients followed by<br />

pathologic fracture (22%). Patients survived for a median <strong>of</strong> 6 months after<br />

bone metastases diagnosis. The occurrence <strong>of</strong> a SRE was associated with a<br />

decreased overall survival (P�0.04). A subgroup analysis revealed that<br />

zoledronic acid significantly prolonged median time to first SRE (5 versus 2<br />

months; P � 0.002). Conversely, It did not determine an improvement in<br />

terms <strong>of</strong> overall survival, even if favorable trend was identified (median, 7<br />

versus 5 months, respectively; P � 0.105). Conclusions: This study<br />

illustrates the burden <strong>of</strong> bone disease from STS and supports the use <strong>of</strong><br />

zoledronic acid in this setting.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


646s Sarcoma<br />

10064 General Poster Session (Board #50A), Sun, 8:00 AM-12:00 PM<br />

Head and neck sarcomas: A comprehensive cancer center experience. Presenting<br />

Author: Mohamedtaki Abdulaziz Tejani, University <strong>of</strong> Rochester Medical<br />

Center, Rochester, NY<br />

Background: Head/neck sarcomas are rare, accounting for 1% <strong>of</strong> head/neck<br />

malignancies and 5% <strong>of</strong> sarcomas. There are about 1000 cases/year in the U.S.<br />

comprising <strong>of</strong> many histologic subtypes making rigorous study <strong>of</strong> these tumors<br />

difficult. Outcomes have historically been worse in this anatomic group due to<br />

unique obstacles encountered during their treatment. Methods: We retrospectively<br />

analyzed cases <strong>of</strong> head/neck s<strong>of</strong>t tissue sarcomas treated at Fox Chase<br />

Cancer Center (1999–2009). Study objectives were to describe clinicopathologic<br />

characteristics, treatment and outcomes <strong>of</strong> adult head/neck sarcoma<br />

patients and identify prognostic factors for disease control and survival. Cox<br />

proportional hazards regression analysis was performed to identify predictors <strong>of</strong><br />

DFS and OS. The HR and 95% CI for Cox model and median DFS/OS from<br />

Kaplan-Meier curves were calculated. Results: 31 patients were identified.<br />

Median age was 50 (range 15–91). 23 patients were male. Most tumors were<br />

high grade (25) and almost all were localized at presentation (29). Common<br />

histologies were synovial cell (6), rhabdomyosarcoma (5), angiosarcoma (4),<br />

lipsarcoma (4) and leiomyosarcoma (3). 25 patients with localized stage<br />

underwent excision: 9 had surgical resection only, 10 had surgical resection with<br />

neo/adjuvant radiation and 6 had surgical resection as well as neo/adjuvant<br />

chemotherapy and radiation. 7 patients required re-excision to achieve negative<br />

margins. Median DFS was 1.1 years (95% CI: 0.7–12.1) and median OS was 3.3<br />

years (95% CI: 2.1-5.4). Of 16 patients with recurrent disease, 12 were local.<br />

Patients with positive margins had worse DFS (p�0.04) and OS (p�0.03).<br />

Tumors greater than 5cm were associated with worse DFS (p�0.07). Age and<br />

grade did not correlate with outcome. Conclusions: Head/neck sarcomas are<br />

difficult to completely excise due to anatomic constraints and local recurrence<br />

rates are high. Achieving a negative margin is critical in improving outcome.<br />

Median DFS and OS are worse than those achieved with other subsites.<br />

Head/neck sarcomas are best managed in a multi-disciplinary setting.<br />

Univariate association<br />

DFS OS<br />

HR 95% CI p HR 95% CI p<br />

Neg vs. pos margins 4.03 1.05-15.39 0.04 4.47 1.14-17.58 0.03<br />

Size 5cm 3.00 0.91-9.91 0.07 1.66 0.55-4.99 0.37<br />

10066 General Poster Session (Board #50C), Sun, 8:00 AM-12:00 PM<br />

Preclinical study <strong>of</strong> trabectedin (TR) and poly(ADP-ribose)polymerase 1<br />

(PARP-1) inhibitor combination in s<strong>of</strong>t tissue sarcoma (STS). Presenting<br />

Author: Ymera Pignochino, Institute for Cancer Research and Treatment,<br />

Candiolo, Italy<br />

Background: TR is an alkylating agent approved in Europe for the treatment<br />

<strong>of</strong> advanced STS as second/third line therapy. TR binds to the minor groove<br />

<strong>of</strong> DNA and interferes with gene transcription and nucleotide excision<br />

repair mechanism, inducing DNA double strand breaks (DSBs), and S/G2<br />

cell cycle arrest. There is a strong clinical interest to increase TR activity<br />

combining it with other anti-cancer drugs. PARP-1 inhibitors disable DNA<br />

base-excision repair mechanism causing the accumulation <strong>of</strong> DSBs and<br />

look like a reasonable TR partner to be explored. We focused our in vitro<br />

studies on the effects <strong>of</strong> the combination <strong>of</strong> TR with the PARP-1 inhibitor<br />

Olaparib (OL). Methods: We explored the activity <strong>of</strong> TR-OL combination<br />

against a panel <strong>of</strong> different histotypes <strong>of</strong> STS cell lines, evaluating cell<br />

viability after 72h treatment with escalating doses <strong>of</strong> TR (0-2 nM), OL<br />

(0-20 �M), and their constant combination. Following colony formation,<br />

cell cycle, apoptosis (annexin V�/PI�) and DNA damage (phospho-histone<br />

H2AX - Ser139) were checked. Results: The TR-OL combination strongly<br />

affects STS cell viability, showing synergism (Combination Index � 1,<br />

based on Chou–Talalay method) on 8 out <strong>of</strong> 13 cell lines tested. We<br />

observed a strong synergism, as a massive reduction <strong>of</strong> colony growth<br />

(402.91, MES-SA, and DMR-SN-8.4.98 lines) induced by the combination<br />

if compared with each single agent (78% vs. ~27% : p�0.05). OL<br />

potentiates the S/G2 cell-cycle arrest caused by TR at 48h (Control� 29%,<br />

TR� 33.4%, OL� 31.5%, TR-OL� 80.9%), and induces a strong increase<br />

<strong>of</strong> the apoptotic cells at 72h (Control� 17.4%, TR� 28.3%, OL� 33.5%,<br />

TR-OL� 56.8%). Furthermore, the TR-OL synergism on DNA damage is<br />

confirmed by a significant increase <strong>of</strong> the DSBs marker (Control� 8.7%,<br />

TR� 59.5%, OL� 23.8%, TR-OL� 76.5%). Conclusions: These results<br />

validate the biological rationale to combine TR and PARP-1 inhibitors in<br />

STS and suggest assessing this drug combination in the clinical setting.<br />

10065 General Poster Session (Board #50B), Sun, 8:00 AM-12:00 PM<br />

Response to radiation therapy in solitary fibrous tumor/hemangiopericytoma.<br />

Presenting Author: Giacomo Baldi, Fondazione IRCCS Istituto<br />

Nazione dei Tumori, Milan, Italy<br />

Background: To reporton the activity <strong>of</strong> radiotherapy (RT) in a retrospective<br />

series <strong>of</strong> solitary fibrous tumors (SFT)/hemagiopericytoma (HCP), a rare s<strong>of</strong>t<br />

tissue sarcoma subtype whose sensitivity to RT is poorly known. Methods:<br />

We retrospectively reviewed all patients with a diagnosis <strong>of</strong> SFT/HCP whom<br />

we saw from 2005 to 2011, focusing on cases treated with RT in<br />

neoadjuvant, adjuvant, exclusive, palliative settings. Patients who received<br />

concomitant chemotherapy were excluded. We confirmed diagnosis by<br />

review in all cases. We evaluated the overall response rate (RR) by RECIST.<br />

Results: Nineteen patients were identified (males � 9, females � 10;<br />

median age � 61 years, range 39-85; site <strong>of</strong> primary� 6 meninges, 4<br />

pleura, 3 pelvis, 3 retroperitoneum, 2 limbs, 1 abdominal wall; site <strong>of</strong> RT�<br />

5 bone and s<strong>of</strong>t tissue, 5 pelvis, 3 s<strong>of</strong>t tissue, 2 meninges, 2 retroperitoneum,<br />

1 abdominal wall, 1 pleura; reason for RT� 3 neoadjuvant, 2<br />

adjuvant, 2 exclusive and 12 palliative setting). Median RT dose was 42 Gy<br />

(range 12-60). Seventeen <strong>of</strong> 19 pts are evaluable for response (2 adjuvant).<br />

The RR was: 29% (4 pts) partial response, 65% (12 pts) stable disease<br />

(SD), 6% (1 pt) progressive disease (PD). All patients treated with<br />

neoadjuvant RT had a RECIST SD as best response to pre-operative<br />

treatment with evidence <strong>of</strong> pathologic response in 2 <strong>of</strong> 3 cases. Conclusions:<br />

Contrary to what is widely thought,this retrospective analysis suggests that<br />

SFT/HCP is sensitive to radiation therapy. Prospective studies are needed.<br />

10067 General Poster Session (Board #50D), Sun, 8:00 AM-12:00 PM<br />

Global differences in chemotherapeutic regimens <strong>of</strong> s<strong>of</strong>t tissue sarcoma<br />

(STS): Results <strong>of</strong> PALETTE, an EORTC 62072/GSK VEG110727 global<br />

network phase III trial <strong>of</strong> pazopanib versus placebo. Presenting Author:<br />

Sandrine Marreaud, EORTC Headquarters, Brussels, Belgium<br />

Background: The global PALETTE EORTC/GSK network study on advanced<br />

STS patients (pts) randomized 223 European (EU), 43 <strong>American</strong> (US), 81<br />

Asian, and 22 Australasian (AUS) pts between pazopanib and placebo.<br />

Prior to study entry pts should have received at least one anthracycline<br />

containing regimen, and preferably, if pts’condition allowed, all available<br />

standard chemotherapy treatments for STS in their countries. We investigated<br />

the differences in chemotherapy pts received prior- and post-protocol<br />

at a global level. Methods: Pts were grouped by continent. Pre- and<br />

post-protocol chemotherapeutic treatments (txs) were analyzed. The number<br />

<strong>of</strong> prior lines <strong>of</strong> systemic tx for advanced STS was divided between 0-1<br />

vs 2 or more. Chemotherapy was studied in detail between Asian and other<br />

countries. Fisher’s exact test was used for statistical analysis. Results: Data<br />

from all 369 pts were analyzed. (Neo-) adjuvant systemic tx was given in<br />

21% <strong>of</strong> EU, 23% <strong>of</strong> US, 36 % <strong>of</strong> Asian and 41% <strong>of</strong> AUS pts. In advanced<br />

STS: 2 or more lines <strong>of</strong> prior systemic txs were administered in 60% <strong>of</strong> EU,<br />

84% <strong>of</strong> US, 42% <strong>of</strong> Asian, 22% <strong>of</strong> AUS pts before study entry. Prior to<br />

study entry 68% <strong>of</strong> EU, 65% <strong>of</strong> US, 84% <strong>of</strong> Asian and 68% <strong>of</strong> AUS pts<br />

received ifosfamide(or analogs); after protocol 21 % <strong>of</strong> EU, 14% <strong>of</strong> US,<br />

13% <strong>of</strong> Asian and 5% <strong>of</strong> AU pts. There were significant differences in<br />

chemotherapy between Asian (n�81) and other (n�288) pts: prior to<br />

protocol trabectedin was administered in 1 vs 21%; gemcitabine 19 vs 39<br />

%; cisplatin: 36 vs 5%; etoposide 32 vs 4% (all p�0.001), and docetaxel:<br />

17 vs 31 % (p�0.004) <strong>of</strong> pts. Post-protocol trabectedin was given to 4 vs<br />

34% (p�0.001), gemcitabine both in 19%; cisplatin 10 vs 4% (p�0.01);<br />

etoposide 12 vs 6%, and docetaxel 20 vs 12 % (both, p�0.02) <strong>of</strong> pts.<br />

Conclusions: The PALETTE study showed considerable intercontinental<br />

differences in chemotherapeutic txs for advanced STS pre- and post study<br />

drug. These differences might be due to availability and reimbursement <strong>of</strong><br />

drugs, clinical studies and the practice patterns <strong>of</strong> sarcoma specialists<br />

world-wide. This is <strong>of</strong> particular interest for the design <strong>of</strong> future randomised<br />

global clinical trials in advanced STS.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


10068 General Poster Session (Board #50E), Sun, 8:00 AM-12:00 PM<br />

Whole-body PET/MR in s<strong>of</strong>t tissue sarcoma (STS) patients. Presenting<br />

Author: Stephan Richter, Medizinische Klinik und Poliklinik I, University<br />

Hospital Carl Gustav Carus Dresden, Dresden, Germany<br />

Background: PET/MR (positron emission tomography/magnetic resonance<br />

imaging) is a new whole-body hybrid imaging technique that combines<br />

metabolic and cross-sectional diagnostic imaging. To date the only available<br />

clinical data are drawn from feasibility studies in small series <strong>of</strong> head<br />

and neck cancers and intracranial tumors. Imaging by combined PET and<br />

computed tomography (PET/CT) has been investigated in STS for biopsy<br />

guidance, response assessment and grading. Methods: This exploratory<br />

analysis evaluated the outcomes <strong>of</strong> PET/MRI in 21 patients with STS in<br />

different treatment settings: (a) neoadjuvant setting, (b) metabolic-driven<br />

local therapy in metastatic sarcoma, and (c) palliative treatment. For<br />

examination we used an Ingenuity PET/MR system (Philips Healthcare). It<br />

combines a 3-Tesla MRI and a PET scanner with time-<strong>of</strong>-flight technology.<br />

Results: PET/MR shows a high contrast imaging without significant artifacts<br />

or distortions. (a) Four patients with high-risk sarcoma (3 rhabdo, 1<br />

pleomorphic) completed the planned neoadjuvant therapy. Change in<br />

tumor size did not correlat with pathologic response, whereas surgical<br />

outcome was well predicted by metabolic changes. Due to this finding the<br />

preplanned course <strong>of</strong> chemotherapy for one patient was changed. (b) To<br />

prolong disease stabilization <strong>of</strong> 3 patients with a remnant metabolic activity<br />

in a single spot they underwent a surgical resection <strong>of</strong> a single metastatic<br />

lesion or had a local radiotherapeutic approach. (c) In 3 patients with stable<br />

disease after first-line treatment with combination <strong>of</strong> anthracyline and<br />

ifosfamide persisting metabolic activity indicated a switch to another<br />

alternative 2nd line regime. Trabectedin as 2nd line therapy decreased<br />

metabolic activity that finally resulted in a tumor regression. Conclusions:<br />

To our knowledge this is the first report on patients with STS examined with<br />

whole-body-PET/MR. PET/MR is feasible in STS and may provide valuable<br />

information in treatment, monitoring and prognosis <strong>of</strong> patients with STS.<br />

This technique may help to choose accurate and on-time treatment option<br />

without unnecessary time delay. Further prospective studies to evaluate<br />

PET/MR in STS are warranted.<br />

10070 General Poster Session (Board #50G), Sun, 8:00 AM-12:00 PM<br />

Multimodality therapy for pulmonary metastasis in sarcoma patients:<br />

Results <strong>of</strong> a single institution. Presenting Author: Nasreen A Vohra, H. Lee<br />

M<strong>of</strong>fitt Cancer Canter & Research Institute, Tampa, FL<br />

Background: Pulmonary metastasectomy (PM) for sarcoma can result in<br />

significant long term survival in carefully selected patients (pts). We report<br />

our experience with pulmonary metastasectomy. Methods: After IRB approval,<br />

120 pts who underwent PM for sarcoma at M<strong>of</strong>fitt Cancer Center<br />

from 1999 to 2011 were reviewed. Survival was calculated according to the<br />

Kaplan-Meier method with Cox proportional hazard univariate (UV) and<br />

multivariate (MV) models used to analyze relationships between clinicopathologic<br />

variables and overall survival (OS). P-value � 0.05 was<br />

considered significant. Results: Median age was 51 yrs, 95(79%) pts had<br />

s<strong>of</strong>t tissue sarcomas with pleomorphic being most common. Of the<br />

25(21%) osseous sarcomas, osteosarcoma was most common. 20(15%)<br />

had synchronous metastasis (mets); <strong>of</strong> the pts with local disease only,<br />

median time to recurrence from primary resection was 13 months (mo). OS<br />

from initial resection was 55 mo for pts with local disease only vs. 27 mo for<br />

pts with synchronous mets. On UV analysis, use <strong>of</strong> radiation, number <strong>of</strong><br />

index lesions, presence <strong>of</strong> synchronous mets and time to recurrence<br />

achieved significance. On MV analysis, only shorter time to recurrence<br />

(p�.03) and presence <strong>of</strong> synchronous mets (p�.04) were independent<br />

predictors <strong>of</strong> poor survival. 63(52%) pts [26(22%) neoadjuvant, 19(16%)<br />

adjuvant, 18(15%) both] received chemotherapy for the primary tumor,<br />

while 57(48%) did not. There was a trend towards poor survival in pts<br />

receiving chemotherapy compared to pts receiving no chemotherapy<br />

(p�0.06, HR 1.59). In addition 40(34%) pts received chemotherapy prior<br />

to PM with no difference in OS compared to pts who did not get<br />

chemotherapy (p�0.1, HR 1.55). 71(59%) had 1 PM, 32(27%) had 2 and<br />

17(14%) had 3 or more PM with OS being 17 mo, 37 mo and 34 mo in<br />

each group, respectively. A higher number <strong>of</strong> PM was associated with<br />

improved survival (p�.01). Conclusions: Pts with a shorter disease free<br />

interval have a decreased OS as do pts with synchronous mets. Pts<br />

undergoing multiple PM have a survival benefit likely resulting from<br />

favorable disease biology. Failure <strong>of</strong> chemotherapy to show a survival<br />

benefit may be a result <strong>of</strong> selection bias for patients with aggressive disease<br />

being treated with chemotherapy.<br />

Sarcoma<br />

647s<br />

10069 General Poster Session (Board #50F), Sun, 8:00 AM-12:00 PM<br />

Neoadjuvant chemotherapy for adult s<strong>of</strong>t tissue sarcomas: A phase II trial.<br />

Presenting Author: Samuel Aguiar, A.C. Camargo Cancer Hospital, Sao<br />

Paulo, Brazil<br />

Background: Treatment <strong>of</strong> s<strong>of</strong>t tissue sarcomas (STS) is characterized by<br />

high rates <strong>of</strong> local control, but poor overall survival because <strong>of</strong> distant<br />

relapses and high rates <strong>of</strong> wound complications, when preoperative<br />

radiation is used. The objective <strong>of</strong> this study was to test the effectiveness <strong>of</strong><br />

a protocol with neoadjuvant chemotherapy for STS. Methods: A phase II<br />

single-arm prospective trial was carried out. Only adult patients with high<br />

grade extremity lesions and tumors deep and larger than 5 cm were<br />

included. A total <strong>of</strong> four cycles <strong>of</strong> chemotherapy was administered<br />

pre-operatively. The chemotherapeutic regimen was: ifosfamide – total <strong>of</strong><br />

9.0 g/m2 per cycle, infused in 2 hours from Day 1 to Day 5 (1.8<br />

mg/m2/day). Half <strong>of</strong> the equivalent dose <strong>of</strong> mesna was infused 15 min<br />

pre-ifosfamide and 4 hours post-ifosfamide. Doxorubicin – total <strong>of</strong> 60mg/m2<br />

per cycle, was infused in bolus on Day 1. Filgrastima 300 mcg, SC, was<br />

administered after the last dose <strong>of</strong> chemotherapy for 5 days. Radiation was<br />

given after surgery. Toxicity was classified by the NIH Toxicity Criteria and<br />

response was determined by the RECIST criteria. The others endpoints<br />

were the amputation and the wound-related complication rates. Results:<br />

Between January, 2005 and May, 2011, 42 patients were included.<br />

21(50%) patients have completed the 4 cycles. Nineteen patients (45.2%)<br />

have grade 3 or 4 toxicity, and one (2.3%) death related to treatment had<br />

occurred. Between severe complications, febrile neutropenia was the most<br />

frequent. By using the RECIST criteria, we observed 10(24.5%) cases <strong>of</strong><br />

progression, 24(58.5%) cases <strong>of</strong> stable disease, and 7(17%) partial<br />

responses. No complete clinical or radiological response was observed. In<br />

the pathological analysis <strong>of</strong> the surgical specimens, 4(9.7%) cases showed<br />

no residual disease (complete pathological response), and a total <strong>of</strong> 6<br />

(14.6%) showed � 5% <strong>of</strong> viable residual cells. The amputation rate was<br />

4.8% (2 cases) and complications related to the wound were observed in 9<br />

patients (21.9%). Conclusions: The protocol showed a good rate <strong>of</strong><br />

objective and pathological response, low rate <strong>of</strong> complications related to<br />

the operative wound, and maintained an acceptable amputation rate. On<br />

the other hand, we observed high rate <strong>of</strong> progression, by RECIST criteria.<br />

10071 General Poster Session (Board #50H), Sun, 8:00 AM-12:00 PM<br />

Atypical vascular neoplasms and risk <strong>of</strong> development <strong>of</strong> angiosarcoma.<br />

Presenting Author: Vinod Ravi, University <strong>of</strong> Texas M. D. Anderson Cancer<br />

Center, Houston, TX<br />

Background: Atypical vascular neoplasms are rare cutaneous lesions that<br />

develop following radiation therapy to the breast that have evidence <strong>of</strong><br />

atypia but do not meet all criteria for a diagnosis <strong>of</strong> angiosarcoma. They<br />

typically have a benign course and are managed by surgical resection.<br />

Histopathological changes consistent with atypical vascular neoplasm are<br />

commonly found adjacent to angiosarcoma and have lead some investigators<br />

to believe that these may be precursor lesions to development <strong>of</strong><br />

angiosarcoma. Methods: We performed a retrospective review <strong>of</strong> all patients<br />

with a diagnosis <strong>of</strong> atypical vascular neoplasm from 2002 to 2012. Patient<br />

with a prior/concurrent history <strong>of</strong> angiosarcoma were excluded from the<br />

analysis. Primary objective <strong>of</strong> the study was to determine the percentage <strong>of</strong><br />

patients with atypical vascular neoplasms that progressed to angiosarcoma<br />

on long-term follow-up. Results: 37 patients were identified in the tumor<br />

registry at MD Anderson cancer center with a diagnosis <strong>of</strong> atypical vascular<br />

neoplasm between 2002 and 2012 with a median follow-up <strong>of</strong> 24 months.<br />

The median age <strong>of</strong> the study population was 53. Females represented 89%<br />

<strong>of</strong> the study population. Atypical vascular neoplasms occurred most<br />

frequently in the breast/chest (70%) and 68% <strong>of</strong> patients had a prior<br />

history <strong>of</strong> breast cancer. 54% <strong>of</strong> patients had prior radiation therapy. The<br />

median time to development <strong>of</strong> atypical vascular neoplasms from radiation<br />

therapy was 52 months. Among patients who underwent surgical resection<br />

with negative margins, there were no recurrences. We failed to observe any<br />

progression to angiosarcoma in the study population. Conclusions: Patients<br />

with atypical vascular neoplasms may not progress to angiosarcoma and are<br />

best managed with surgical resection.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


648s Sarcoma<br />

10072 General Poster Session (Board #51A), Sun, 8:00 AM-12:00 PM<br />

Effect <strong>of</strong> anti-�1 integrin antibody on lung seeding <strong>of</strong> osteosarcoma cells in<br />

live mice visualized by single-cell in vivo imaging. Presenting Author:<br />

Hiroaki Kimura, Department <strong>of</strong> Orthopaedic Surgery, Graduate School <strong>of</strong><br />

Medical Science, Kanazawa University, Kanazawa, Japan<br />

Background: Integrins play a role in tumor growth and metastasis (Int. J.<br />

Cancer 129, 2905-2915, 2011). However, the effect <strong>of</strong> integrin inhibition<br />

has not been visualized on single cancer cells in vivo. In this study, we used<br />

a powerful subcellular in vivo imaging model to demonstrate how an<br />

anti-integrin antibody affects seeding and growth <strong>of</strong> osteosarcoma cells on<br />

the lung. Methods: The 143B human osteosarcoma cell line expressing red<br />

fluorescent protein (RFP) in the cytoplasm and green fluorescent protein<br />

(GFP) in the nucleus was established. Using the double-labeled osteosarcoma<br />

cells, single cancer-cell seeding in the lung after i.v. injection <strong>of</strong><br />

osteosarcoma cells was imaged in live mice. Results: The anti-b1 integrin<br />

monoclonal antibody, AIIB2, greatly inhibited the seeding <strong>of</strong> cancer cells<br />

on the lung while a control antibody had no effect. To image the efficacy <strong>of</strong><br />

the anti-integrin antibody on spontaneous metastasis, mice with orthotopically-growing<br />

143B-RFP cells in the tibia were also treated with AIIB2 or<br />

control anti-rat IgG1 antibody. After 3 weeks treatment, mice were<br />

sacrificed and primary tumors and lung metastases were evaluated with<br />

fluorescence imaging. AIIB2 significantly inhibited spontaneous lung<br />

metastasis but not primary tumor growth. In a separate experiment, the<br />

anti-�1 integrin antibody increased survival in the orthothopic osteosarcoma<br />

model. Conclusions: The efficacy <strong>of</strong> the anti-�1 integrin antibody<br />

against metastasis may be due to inhibition <strong>of</strong> lung seeding <strong>of</strong> the cancer<br />

cells. The increased survival <strong>of</strong> mice with orthotopically-growing 143B-RFP<br />

treated with AIIB2 may be due to inhibition <strong>of</strong> metastasis, which in turn<br />

may be inhibited by effect <strong>of</strong> the anti-�1 integrin on cancer-cell seeding in<br />

the lung.<br />

10074 General Poster Session (Board #51C), Sun, 8:00 AM-12:00 PM<br />

Age as a prognostic factor in osteosarcoma: Survival analysis. Presenting<br />

Author: Maria Pallotta Guadalupe, Hospital Italiano de Buenos Aires,<br />

Ciudad Autonoma de Buenos Aires, Argentina<br />

Background: Studies comparing survival between adult and paediatric<br />

population with osteosarcoma are scarce and contradictory. Generally<br />

adults were excluded from analysis in historical series. End Point: evaluate<br />

age as prognostic factor in ostosarcomas IIb treated exclusively by a<br />

multidisciplinary group in a single institution. Methods: 132/278 patients<br />

with histological diagnosis <strong>of</strong> osteosarcoma IIb were selected. All were<br />

treated exclusively in our hospital from July 1988 until December 2010.<br />

Patients 17 years old or younger were considered paediatric. They all<br />

received presurgical chemotherapy with the same scheme: ifosfamide �<br />

doxorrubicin � high dose methotrexate. Univariate analysis was made<br />

(Fischer exact test). Survival was calculated wih Kaplan-Meier actuarial<br />

method. Curves were compared with log rank test. Multivariate Cox analysis<br />

was made. Results: Median age was 19.6 years (std: 9,1; range 5-58).<br />

Adults: 77, children: 55. No differences were detected between the two<br />

age groups regarding: elevated alkaline phosphatase, kind <strong>of</strong> surgery<br />

(amputation vs. limb sparing), relapse site (lung, local, other), necrosis<br />

greater than 90% or number <strong>of</strong> lung resections. There was a tendency<br />

towards axial localization in adults (p�0.05). No paediatric patient had<br />

inadequate medical intervention, but it was present in 14.3% <strong>of</strong> adults<br />

(p�0.002). 5 year Overall survival (5y OS) in children was 85,2%<br />

compared with 61,8% in adults (p�0.005); Disease free survival (DFS)<br />

had a non significant tendency to be better in children (69.6% vs. 51,3%).<br />

Variables associated with worse OS were: axial location (p�0.01), elevated<br />

alkaline phosphatase (0.003), amputation (p�0.008), local relapse or<br />

systemic non-lung metastasis (p� 0.001), necrosis less than 90% (0.001).<br />

Multivariate Cox analysis showed association between OS and paediatric<br />

population (p�0.01) and necrosis greater than 90% (p�0.001), while<br />

DFS was associated with necrosis greater than 90% (p�0.01) and elevated<br />

alkaline phosphatase (p�0.05). Conclusions: Paediatric population presents<br />

better survival compared to adults in our institution. Differences in<br />

tumour biology and in the mode <strong>of</strong> presentation related to age may be the<br />

explanation.<br />

10073 General Poster Session (Board #51B), Sun, 8:00 AM-12:00 PM<br />

Navigation-assisted surgery for bone and s<strong>of</strong>t tissue tumors with bony<br />

extension. Presenting Author: Makoto Ieguchi, Department <strong>of</strong> Orthopedic<br />

Surgery, Yodogawa Christian Hospital, Osaka, Japan<br />

Background: In recent times,minimally invasive surgery, such as endoscopic<br />

surgery, has gained a lot <strong>of</strong> popularity.The navigation system was<br />

introduced to orthopedic surgery in the 1990s. These days, computed<br />

tomography (CT)-based navigation systems are commonly used in spine<br />

and joint replacement surgery because <strong>of</strong> their precision. The aim <strong>of</strong> our<br />

study was to evaluate the accuracy and efficacy <strong>of</strong> navigation-assisted<br />

excision <strong>of</strong> bone and s<strong>of</strong>t tissue tumors. Methods: From January 2006 to<br />

December 2009, we performed navigation-assisted surgery in 16 patients<br />

(11 men and 5 women; mean age, 39 years; range, 13-70 years). We<br />

diagnosed 9 benign bone tumors and 7 malignant bone and s<strong>of</strong>t tissue<br />

tumors. In 2 patients, the malignant s<strong>of</strong>t tissue tumors infiltrated the<br />

adjacent bones. We performed excisional biopsies for the benign tumors<br />

and en bloc excisions for the malignant tumors. In all the cases, the point<br />

registration method was used with 10 skin markers that were placed around<br />

the tumor. Each excisional difference between the preoperative and<br />

postoperative plans was evaluated histologically or by postoperative CT.<br />

Results: The mean preoperative registration matching time was 12.8 min<br />

(range, 8-25 min). The total mean preparation time was 24 min (range,<br />

16-35 min). The mean accuracy <strong>of</strong> this system, which was determined<br />

using the skin markers, was 0.93 mm (range, 0.6-1.5 mm). All biopsied<br />

and excised specimens were evaluated by pathologic examination and<br />

postoperative CT imaging. The mean difference between the planned<br />

margin and postoperative CT or the excised histological specimen was 0 to<br />

4 mm. The mean follow-up period was 53.2 months (range, 10-70<br />

months). There were no local recurrences, except for a case <strong>of</strong> chordoma<br />

that required excision <strong>of</strong> skip metastases and a case <strong>of</strong> extraskeletal<br />

osteosarcoma, in which the patient died from the disease. Conclusions: We<br />

report our experience with navigation-assisted surgery for bone and s<strong>of</strong>t<br />

tissue tumors performed using skin markers. Navigation-assisted surgery<br />

was indicated in the case <strong>of</strong> sufficiently reliable, accurate, and minimally<br />

invasive resections.<br />

10075 General Poster Session (Board #51D), Sun, 8:00 AM-12:00 PM<br />

Efficacy <strong>of</strong> newly developed platinum complexes against osteosarcoma,<br />

bone-targeting platinum, and proteasome inhibitory platinum. Presenting<br />

Author: Kentaro Igarashi, Department <strong>of</strong> Orthopaedic Surgery, Kanazawa<br />

University, Kanazawa, Japan<br />

Background: Cisplatin is one <strong>of</strong> the most effective anti-cancer drugs<br />

available for the treatment <strong>of</strong> human solid tumors including osteosarcoma.<br />

As we had already reported, we have utilized caffeine in our chemotherapy<br />

protocol. And we achieved excellent clinical results. But effectiveness <strong>of</strong><br />

cisplatin has been limited by side effects, and resistance. Here we<br />

developed two novel platinum compounds. 3Pt is trinuclear platinum<br />

complex bearing geminal bisphosphonate moieties, 1Pt is mononuclear<br />

platinum complex which has proteasome inhibitory activity. We performed<br />

comparative studies <strong>of</strong> our novel platinum compounds with osteosarcoma.<br />

Methods: Two novel platinum complexes were synthesized by Pr<strong>of</strong>. Odani at<br />

school <strong>of</strong> pharmaceutical sciences <strong>of</strong> our university and cisplatin and<br />

caffeine were obtained from constructor. Three cell lines (MG63, 143B,<br />

and LM8) were used. Cell survival after a 72 hrs exposure to these<br />

compounds was assessed by WST-8 assay, and IC50 value was calculated<br />

for each compound. Apoptosis was assessed by DNA fragmentation and<br />

Annexin V-FITC/propidium iodine assay. Results: Each compound strongly<br />

caused concentration-dependent cytocidal effect. IC50 value <strong>of</strong> trinuclear<br />

compound is superior to cisplatin, and both complexes showed caffeine<br />

potentiation. Apoptosis induction and acetylation <strong>of</strong> histon H2AX were<br />

observed. In vivo, 1Pt showed almost same, 3Pt showed strong antitumor<br />

effect compared to cisplatin. Conclusions: Two novel platinum compounds<br />

that we developed showed strong ant-tumor effect in osteosarcoma in vitro<br />

and in vivo. As bisphosphonate has high affinity to calcium ions, 3Pt targets<br />

bone tissue and expected to reduce side effects at extraskeletal sites and to<br />

overcome the drug resistance. Proteasome inhibitory platinum compound<br />

has never been reported before, we will investigate its anti-tumor mechanism<br />

precisely.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


10076 General Poster Session (Board #51E), Sun, 8:00 AM-12:00 PM<br />

Study <strong>of</strong> the expression <strong>of</strong> integrin pathway’s proteins as predictive markers<br />

<strong>of</strong> response to neoadjuvant chemotherapy in pretreatment biopsies <strong>of</strong><br />

patients with high-grade osteosarcoma. Presenting Author: Elizabeth Laurence<br />

Cohen-Jonathan Moyal, Institut Claudiusr Regaud-INSERM 1037,<br />

Toulouse, France<br />

Background: Up to now, chemosensitivity to neoadjuvant chemotherapy<br />

performed for patients with osteosarcoma is evaluated on the surgical<br />

resection by the evaluation <strong>of</strong> the percentage <strong>of</strong> necrotic cells. No<br />

predictive pr<strong>of</strong>ile <strong>of</strong> chemotherapy has been described yet. Because we<br />

have previously shown that integrin pathway controls genotoxic-induced<br />

cell death and hypoxia via ILK, FAK and RhoB, we hypothesized that the<br />

expression <strong>of</strong> the proteins involved in this pathway in pre-treatment<br />

biopsies could be associated with sensitivity to chemotherapy in osteosarcoma.<br />

Methods: We studied by immunohistochemistry �1, �3, �5, integrins,<br />

ILK, FAK, GSK-3�, RhoB and Ang-2 expression in 36 osteosarcomas<br />

biopsies <strong>of</strong> patients obtained before treatment. All the patients were<br />

treated by the SFOP OS94 chemotherapy protocol and underwent wide<br />

conservative surgery. Response to preoperative chemotherapy was assessed<br />

on the surgical resection by the Rosen’s protocol. An immunoreactive<br />

score (IRS) was assessed, combining the percentage <strong>of</strong> positive tumour<br />

cells and staining intensity. We evaluated the correlation <strong>of</strong> the selected<br />

markers expression with response to preoperative chemotherapy and<br />

clinical outcome. Results: FAK expression was statistically linked with ILK<br />

expression (rho�0.43; p�0.020), �1integrin expression with FAK expression<br />

(rho�0.51); p�0.003), RhoB with ILK expression (rho�0.55;<br />

p�0.002), and �1 with �3integrin expression (rho�0.70; p�.001),<br />

suggesting that a regulation <strong>of</strong> the integrin pathway that we studied might<br />

exist in high grade osteosarcoma. Moreover, the combination <strong>of</strong> �5<br />

integrin, FAK and GSK-3� discriminated good and poor responder to<br />

chemotherapy (89.9%; 95% CI� [77.4;1.00], yielding a sensitivity <strong>of</strong><br />

94.1%, a specificity <strong>of</strong> 85.7% and a diagnostic accuracy <strong>of</strong> 90.3%.<br />

Conclusions: We report for the first time a protein expression pr<strong>of</strong>ile on<br />

pre-treatment biopsies associating �5 integrin FAK and GSK-3� related<br />

with a poor response to neoadjuvant chemotherapy. This pr<strong>of</strong>ile could help<br />

to select patients for clinical trials using inhibitors <strong>of</strong> this pathway in<br />

association with chemotherapy.<br />

10078 General Poster Session (Board #51G), Sun, 8:00 AM-12:00 PM<br />

Primary localized gastrointestinal stromal tumors (GIST) <strong>of</strong> the duodenum:<br />

Final results <strong>of</strong> a French Sarcoma Group (FSG) retrospective review <strong>of</strong> 110<br />

patients (pts). Presenting Author: Thanh Khoa Huynh, University Hospital<br />

La Timone, Marseille, France<br />

Background: Duodenal GISTs represent 3-5% <strong>of</strong> all GISTs with limited<br />

understanding <strong>of</strong> patient outcomes from small series. We conducted a<br />

retrospective analysis <strong>of</strong> primary localized duodenal GISTs over the past 18<br />

years. Methods: Pts were identified in two ways: a group <strong>of</strong> 101 pts reported<br />

via survey from 20 FSG centers, and a group <strong>of</strong> 9 pts enrolled in the BFR14<br />

trial. Results: Pts were:55 females, 55 males, with a median age <strong>of</strong> 57 years<br />

(30-84), and median ECOG 0 (0-3). Abdominal pain, anemia, and GI<br />

bleeding were the most common symptoms. Tumors (T) originated mainly<br />

in D2 (41%), or D3 (27%), with a median size <strong>of</strong> 5 cm (1.5-30). All pts<br />

except four had resection <strong>of</strong> the primary T. Surgical procedures were: local<br />

resection (LR) [segmental duodenectomy (n�31), wedge local resection<br />

(n�31), local excision (n�6)], and duodenopancreatectomy (DP, n�20).<br />

Resections were R0 in 84 pts (79%), R1 in 6 pts (6%). T characteristics<br />

included: KIT� (n�100), CD34 � (n� 54), mitoses/50 HPF � 5(n�70), or � 5(n�24), Miettinen low-risk (n�37), and high-risk (n�19), necrosis<br />

(n�29), spindle cell (n�84). Genotype was evaluated in 36 cases: KIT<br />

exon 11 mutant (n�30), no mutation (n�4), and KIT exon 9 mutant<br />

(n�2). 12 pts received neoadjuvant imatinib (IM) therapy resulting in 6<br />

PR, 3 SD, 1 PD. 17 pts received adjuvant IM therapy. With a median FU <strong>of</strong><br />

32 months (1-250), 95 pts (86%) are alive. Twenty-eight (26%) pts<br />

relapsed: 6 LR, and 26 metastases. The 4-year OS and EFS rates were<br />

89.5% and 68.2 %. The 6-year OS and EFS rates were 89.5% and 36.5%.<br />

Univariate analysis showed that: age and ECOG PS have an impact on OS<br />

(p� 0.008, p �0.001), necrosis, spindle-cell type, T size, mitoses/50<br />

HPF, and Miettinen risk were predictive <strong>of</strong> relapse (p� 0.001). In<br />

multivariate analysis tumor size and mitoses/ 50 HPF only were predictive<br />

<strong>of</strong> relapse (p� 0.001). Conclusions: Pts with resected duodenal GIST have<br />

a reasonably favourable prognosis. LR rather than DP should be pursued if<br />

possible to preserve optimal pancreas function. Neoadjuvant IM may<br />

potentially allow a proportion <strong>of</strong> patients requiring DP to undergo LR.<br />

Adjuvant IM should be systematically discussed with a patient based on the<br />

individual-risk <strong>of</strong> recurrence.<br />

Sarcoma<br />

649s<br />

10077 General Poster Session (Board #51F), Sun, 8:00 AM-12:00 PM<br />

Association <strong>of</strong> the genetic variants in the nucleotide excision repair genes<br />

XPA and XPC with cisplatin-induced hearing loss in patients with osteosarcoma.<br />

Presenting Author: Melanie M. Hagleitner, Radboud University<br />

Medical Centre, Nijmegen, Netherlands<br />

Background: Cisplatin is a widely used and effective chemotherapeutic<br />

agent in the treatment <strong>of</strong> osteosarcoma. However, cisplatin-induced<br />

ototoxicity is a serious problem, affecting more than 60% <strong>of</strong> patients and<br />

compromising language and cognitive development. Unfortunately, individuals<br />

at risk to develop ototoxicity cannot be identified upfront. Genetic<br />

variants in genes involved in the metabolism <strong>of</strong> cisplatin may predispose to<br />

cisplatin-induced hearing loss and help to identify patients at risk.<br />

Methods: In a candidate gene pathway approach, we selected 224 SNPs in<br />

30 candidate genes related to Platinum-DNA repair pathways and genotyped<br />

for a discovery group <strong>of</strong> 105 patients with osteosarcoma for these<br />

variants. Cisplatin-induced ototoxicity (n � 47), defined as the development<br />

<strong>of</strong> grade 2–4 hearing impairment using Common Terminology Criteria<br />

for Adverse Events (CTCAE version 3), showing a hearing loss <strong>of</strong> �25 dB at<br />

frequencies <strong>of</strong> 4–8 kHz, was associated with genetic variation. A replication<br />

study was performed in a independent cohort <strong>of</strong> 51 patients with<br />

osteosarcoma. Genotyping was performed using the Illumina GoldenGate<br />

assay. Association analysis and meta-analysis were performed using the<br />

whole genome association analysis toolset PLINK. Results: In the discovery<br />

cohort a total <strong>of</strong> 13 SNPs were significantly (p value � 0.05) associated<br />

with ototoxicity. Upon meta-analysis, addition <strong>of</strong> the replication set<br />

resulted in lower p-values for 2 SNPs. The two SNPs showing a strong<br />

association with hearing loss in patients with osteosarcoma were rs2805835<br />

in the gene XPA (p-value 0.01, OR�2.7 (95%CI: 1.20-6.15) and<br />

rs2227999 in XPC (p-value 0.02; OR�3.2 (95% CI:1.19-8.80).<br />

Conclusions: The Nucleotide Excision Repair (NER) genes XPA and XPC<br />

form an important molecular mechanism by which cisplatin DNA adducts<br />

can be repaired and it has recently been shown that these genes have a high<br />

expression in the cochlea. Our data suggest that genetic variants in these<br />

genes, may contribute to cisplatin ototoxicity. This study should be viewed<br />

as the first step in the development <strong>of</strong> genetic markers to predict<br />

cisplatin-induced ototoxicity in individual patients.<br />

10079 General Poster Session (Board #51H), Sun, 8:00 AM-12:00 PM<br />

Relationship <strong>of</strong> CUL4A gene underexpression and prognosis in localized<br />

high-risk s<strong>of</strong>t tissue sarcoma (STS) patients <strong>of</strong> limbs or trunk wall.<br />

Presenting Author: Javier Martin Broto, Hospital Universitario Son Espases,<br />

Palma de Mallorca, Spain<br />

Background: Genes involved in cell cycle checkpoints and chromosome<br />

stability seem to be relevant in distant metastases appearance in STS.<br />

CUL4A is an E3 ubiquitin ligase that has been related to both p16<br />

activation and maintenance <strong>of</strong> genomic stability. Expression <strong>of</strong> CUL4A has<br />

been scarcely described in some tumors with divergent outcome and there<br />

are no publications regarding its expression in STS patients. We previously<br />

showed that protein underexpression <strong>of</strong> CUL4A in metastatic STS was<br />

related to poor survival. Methods: The expression level <strong>of</strong> CUL4A was<br />

determined by quantitative qPCR and was retrospectively performed in a<br />

subset <strong>of</strong> 39 high-risk (grade 3, deep tumours and � than 5 cm) localized<br />

STS <strong>of</strong> limbs or trunk wall patients for which histological material and<br />

clinical data were available. Patients <strong>of</strong> this series were enrolled prospectively<br />

into a randomized clinical trial conducted by the Italian and Spanish<br />

sarcoma groups (ISG-GEIS 0101) comparing 3 vs 5 cycles <strong>of</strong> epirrubicin<br />

and ifosfamide. Differences in PFS, RFS, and OS were calculated using<br />

log-rank test Results: The median age was 51 years with a median follow-up<br />

<strong>of</strong> 52 months. The pathologic subtypes were undifferentiated pleomorphic<br />

sarcoma (35.3%), synovial sarcoma (23.5%) and mixed subtypes (41.2%).<br />

Location was distributed as inferior limbs (66.6%), upper limbs (25.5%)<br />

and trunk wall (7.9%). No relationship was seen between CUL4A expression<br />

and clinical variables (histologic types, location, size or response to<br />

neoadjuvant treatment). The expression level <strong>of</strong> CUL4A was significantly<br />

associated with events <strong>of</strong> progression, 65% vs 32%, p �0.037, for<br />

expression values under and over the median respectively. Regarding 4 y<br />

actuarial survival, there was a significant worse PFS and RFS for patients<br />

underexpressing CUL4A (p�0.041 and 0.009 respectively) and a trend<br />

towards worse OS (p�0.056). Conclusions: Expression <strong>of</strong> CUL4A gene<br />

shows a prognostic role in sarcomas within this high risk localized subset <strong>of</strong><br />

STS patients. The outcome <strong>of</strong> this exploratory analysis is aligned with our<br />

hypothesis that genes implicated in cell cycle regulation and chromosome<br />

stability could play a relevant prognostic role in STS.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


650s Sarcoma<br />

10080 General Poster Session (Board #52A), Sun, 8:00 AM-12:00 PM<br />

Results <strong>of</strong> pulmonary metastasectomy for osteosarcoma in the pediatric<br />

age group. Presenting Author: Bassam Redwan, Division <strong>of</strong> Thoracic<br />

Surgery, University Hospital <strong>of</strong> Muenster, Muenster, Germany<br />

Background: Despite multimodal treatment concepts and complete surgical<br />

resection, prognosis in pediatric patients with pulmonary metastases from<br />

osteosarcoma has remained limited due to frequent relapse <strong>of</strong> disease. We<br />

investigated the results <strong>of</strong> an aggressive surgical approach. Methods: In a<br />

retrospective study, procedures and outcomes <strong>of</strong> pulmonary metastasectomy<br />

in the pediatric age group (up to 18 years) at our institution were<br />

analyzed over a period <strong>of</strong> 10 years (1999-2009). Resection <strong>of</strong> the primary<br />

osteogenic tumor and chemotherapy (CROSS-96-protocol and EURAMOS-<br />

1-protocol) were performed prior to thoracic surgery. Results: Forty-five<br />

pediatric patients (20 females) underwent pulmonary metastasectomies<br />

via sternotomy or sequential anterolateral thoracotomy at a mean age <strong>of</strong> 14<br />

(6 -18) years. At primary surgery, a mean number <strong>of</strong> 7.9 (1 – 53) palpable<br />

suspicious lesions were resected per patient. Histo-pathological evaluation<br />

revealed 3.7 (0 – 40) metastases per patient. Mean total duration <strong>of</strong> surgery<br />

was 152 (46–323) minutes. Mean hospital stay was 10 days (3 – 33).<br />

In-hospital and 30-day mortality was 0%. The overall survival at 1 and 5<br />

years was 97.8% and 77.3%, respectively. Mean disease-free-survival was<br />

12.2 (3.2-38.0) months. In 19 (42.2 %) patients recurrent pulmonary<br />

metastases were detected and re-thoracotomies were required. Up to 7<br />

procedures per patient were performed. Overall survival for patients<br />

undergoing more than one surgical procedure for recurrent lung metastases<br />

was not statistically different from survival in patients without relapse (p �<br />

0.05). Survival was significantly better in patients initially presenting with<br />

less than 10 metastases (85.3 % vs. 54.1 % at 5 years, p � 0.028).<br />

Conclusions: Complete pulmonary metastasectomies are essential in pediatric<br />

osteosarcoma patients with lung metastasis. Repeated resections for<br />

recurrent relapses improve survival and may allow for long-term event-freesurvival.<br />

10082 General Poster Session (Board #52C), Sun, 8:00 AM-12:00 PM<br />

Multimodality treatment <strong>of</strong> pediatric and adult patients with Ewing sarcoma:<br />

A single-institution experience. Presenting Author: David Lorente,<br />

Medical Oncology Department, Valencia, Spain<br />

Background: Treatment <strong>of</strong> Ewing sarcoma pts. usually follows pediatric<br />

protocols, both in children and in adults. However, older patients fare<br />

poorly in most series. We analyze our experience with the 2001 protocol <strong>of</strong><br />

the Spanish <strong>Society</strong> <strong>of</strong> Pediatric Oncology. Methods: Retrospective analysis.<br />

Schema: 6 cycles (cy) <strong>of</strong> VIDE chemotherapy (CT: vincristine, ifosfamide,<br />

etoposide, doxorrubicin). If no progression, local treatment (surgery<br />

or RT) and consolidation adjusted to risk: VACx8 (vincristine, dactinomycin,<br />

ciclophosphamyde) in standard-risk pts; if increased risk (axial,<br />

complete response in lung metastases or non-pulmonary metastases)<br />

VACx1, high-dose CT (busulphan-melphalan) and autologous transplant<br />

(ATSP). Analysis: induction CT toxicity, pathological response rates,<br />

consolidation treatment, disease-free (DFS) and overall survival (OS)<br />

(Kaplan- Meier). Log-rank and Cox regression analysis <strong>of</strong> prognostic factors<br />

in OS. Results: 35 patients (01.2003-05.2011). 60% male. Median age<br />

16 y (r 7-57). Axial (43%), extremities (34%), extra-osseous (18%) and<br />

ribs (9%). Metastases: 54% (lung 58%, bone 26%, others 12%). � 1<br />

location: 29%. Induction CT: 83% received 6 cy. 6% early progressions<br />

and 3% toxic deaths. 196 cycles <strong>of</strong> CT. Dose reduction (etoposide) in 60%.<br />

Grade 3-4 toxicity: neutropenia 13%, anemia 14%, neutropenic fever<br />

13%, diarrhoea-stomatitis 7%.Local treatment: surgery (49%), radiotherapy<br />

(29%), none (22%). In 17 resections, � 90% necrosis in 53%.<br />

Consolidation: VACx8 29%; VACx1-ATSP in 34%; 37% other treatments<br />

(progression). No ATSP-related mortality. Median follow-up: 36 m ( 5-101<br />

m). Median DFS 25 m (16-34 m). Median OS 28 m (15-41 m), 3-year OS<br />

40%. Median time to progression 7 m (0.4-15 m). Median OS from<br />

progression 7 m (0.4-15 m). Age � 15 years, a non-axial primary and no<br />

extra-pulmonary metastases were favourable prognostic factors in the<br />

univariate analysis. Conclusions: Induction CT with the VIDE regimen is<br />

feasible in most patients, with a low risk <strong>of</strong> early progression. Hematological<br />

toxicity is substantial but manageable. Adults patients have a worse<br />

prognosis compared to pediatric patients. Unfortunately, survival after<br />

progression is dismal.<br />

10081 General Poster Session (Board #52B), Sun, 8:00 AM-12:00 PM<br />

EURAMOS-1 study: Recruitment, characteristics, and initial treatment <strong>of</strong><br />

more than 2,000 patients (pts) with high-grade osteosarcoma. Presenting<br />

Author: Jeremy Whelan, University College Hospital, London, United<br />

Kingdom<br />

Background: EURAMOS is a transAtlantic collaboration formed to improve<br />

survival in osteosarcoma by conducting RCTs in a clinically relevant<br />

timeframe. EURAMOS-1, the largest study conducted in this rare cancer,<br />

has completed accrual. It includes two randomized comparisons investigating<br />

treatment optimization on the basis <strong>of</strong> histological response to<br />

neoadjuvant chemotherapy. Methods: Pts �40yrs with resectable, high<br />

grade extremity or axial osteosarcoma were eligible for registration. All were<br />

planned for 2 cycles <strong>of</strong> neoadjuvant methotrexate, doxorubicin, cisplatin<br />

(MAP) then surgical resection <strong>of</strong> the primary tumour. Pts with complete<br />

macroscopic resection and no disease progression were eligible for randomization:<br />

[i] “good responders”, �10% viable tumor, MAP �/- 18m<br />

maintenance pegylated interferon; [ii] “poor responders”, �10% viable<br />

tumor, MAP vs MAPIE (MAP � ifosfamide, etoposide). Target sample size<br />

was ~1,260 pts randomized requiring ~2000 pts registered estimating a<br />

non-randomization rate <strong>of</strong> 30-35%. Results: From Apr 2005 to Dec 2011,<br />

2,260 pts were registered and 1,332 randomized from 17 countries, 320<br />

sites: median age 14yrs (IQR 11-17); 59% male; 50% femoral site; 23%<br />

definite/possible metastases; 92% conventional osteosarcoma (diagnostic<br />

biopsy). At Sep 2011 planned IDMC review, neoadjuvant treatment data<br />

were known for 2024 pts; 1926 (95%) completed 2 cycles pre-operative<br />

MAP with 3 treatment related deaths. 59% pts were randomized; nonconsent<br />

was the most frequent reason for non-randomization. Relative to<br />

expected age-specific incidence (UK NCIN 1979-2007) there was apparent<br />

under recruiting <strong>of</strong> older pts: females �15yrs and all pts �19yrs, with<br />

greater under recruitment <strong>of</strong> females than males �35yrs. Pts 20-29yrs<br />

were less likely to be randomized than those aged 5-19 and �30 yrs, 52%<br />

vs 57-62%. Conclusions: EURAMOS-1 demonstrates that large RCTs are<br />

feasible in rare cancers with inter-continental collaboration and is a model<br />

for future trials. Neoadjuvant MAP was safe in a geographically diverse<br />

cohort. Improving clinical trial access and randomization rates for young<br />

people is still required. Multiple funders detailed at bit.ly/ymUR9w.<br />

10083 General Poster Session (Board #52D), Sun, 8:00 AM-12:00 PM<br />

A retrospective safety analysis <strong>of</strong> adult patients treated with high-dose<br />

methotrexate for osteosarcoma in Stockholm, Sweden. Presenting Author:<br />

Daniel Alm, Oncology, Stockholm, Sweden<br />

Background: Patients with osteosarcoma are routinely treated with pre- and<br />

post-operative chemotherapy that includes high-dose methotrexate. The<br />

treatment is associated with a risk <strong>of</strong> severe renal and hepatic toxicity.<br />

Methods: All patients with osteosarcoma that had received at least one cycle<br />

<strong>of</strong> high-dose methotrexate at the adult oncology department, Karolinska<br />

University Hospital were retrospectively identified. Treatment toxicity,<br />

including hematologic, renal, hepatic toxicity, infections and oral mucositis<br />

were registered and graded according to CTCAE v 4.0. A possible<br />

relationship between methotrexate blood concentration and toxicity was<br />

investigated. Results: Sixteen eligible patients that had received a total <strong>of</strong><br />

103 cycles <strong>of</strong> high-dose methotrexate were identified. Ten patients<br />

experienced a severe hepatic toxicity, (Grade 3, n�5 and Grade 4, n�5).<br />

Grade 3 renal toxicity was seen in one patient and although reversible, it led<br />

to treatment interruption. Reversible, grade 2 elavation <strong>of</strong> serum creatinine<br />

occured in 5 cases. Four grade 3 infections were seen in 2 patients and 8<br />

patients had at least one occurrence <strong>of</strong> Grade 3 oral mucositis. Thrombocytopenia<br />

was a common event (Grade 3, n�5 and Grade 4, n�2) but no<br />

severe bleeding complications were observed. One patient died as a result<br />

<strong>of</strong> multi-organ failure two days after methotrexate administration. Methotrexate<br />

blood concentration at 24 hours from administration could predict<br />

for renal toxicity (p�0.005, by chi-square test), but not for other toxicity.<br />

Conclusions: High-dose methotrexate in adult patients with osteosarcoma<br />

was frequently associated with severe, however reversible toxicity.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


10084 General Poster Session (Board #52E), Sun, 8:00 AM-12:00 PM<br />

Activity <strong>of</strong> MK1775, a selective Wee1 inhibitor, alone or in combination<br />

with gemcitabine, in sarcomas. Presenting Author: Jenny Kreahling, H. Lee<br />

M<strong>of</strong>fitt Cancer Center & Research Institute, Tampa, FL<br />

Background: Sarcomas consist <strong>of</strong> more than 50 subtypes <strong>of</strong> mesenchymal<br />

tumors. Doxorubicin alone or in combination has been the primary therapy<br />

for treatment <strong>of</strong> sarcomas; however, the response rates are suboptimal in<br />

many <strong>of</strong> the more common adult subtypes <strong>of</strong> s<strong>of</strong>t tissue sarcoma.<br />

Accordingly, new agents are needed for the treatment <strong>of</strong> this heterogeneous<br />

group <strong>of</strong> diseases. Wee1 is a critical component <strong>of</strong> the G2/M cell cycle<br />

checkpoint control and mediates cell cycle arrest by regulating the<br />

phosphorylation <strong>of</strong> CDC2. Methods: MK1775 treatment <strong>of</strong> multiple sarcoma<br />

preclinical models at clinically relevant concentrations leads to<br />

unscheduled entry into mitosis and initiation <strong>of</strong> apoptotic cell death. In our<br />

current study we have investigated the therapeutic efficacy <strong>of</strong> MK1775 in<br />

sarcoma cell lines, patient-derived tumor explants ex vivo and in vivo in a<br />

xenograft model <strong>of</strong> osteosarcoma both alone and in combination with<br />

gemcitabine. Results: In patient-derived bone and s<strong>of</strong>t tissue sarcoma<br />

samples ex vivo treatments show MK1775 in combination with gemcitabine<br />

causes significant apoptotic cell death suggesting that this treatment<br />

may represent a novel approach in the treatment <strong>of</strong> sarcomas. The cytotoxic<br />

effect <strong>of</strong> Wee1 inhibition on sarcoma cells appears to be independent <strong>of</strong><br />

p53 mutational status. Furthermore, in a patient-derived osteosarcoma<br />

xenograft mouse model we show the therapeutic efficacy <strong>of</strong> MK1775 in vivo<br />

by utilizing magnetic resonance imaging (MRI) and diffusion MRI methods.<br />

Our data shows MK1775 in combination with gemcitabine dramatically<br />

slows tumor growth, increases apoptotic cell death and increases CDC2<br />

activity. Cell viability, a clinically established prognostic indicator <strong>of</strong><br />

survival, was lowest with the combination and very low in animals treated<br />

with MK1775 alone. This was mainly due to increased mineralization <strong>of</strong> the<br />

tumors. Caspase-3 was increased in MK1775 treated animals by immunohistochemistry<br />

as well. Conclusions: These results together with the<br />

promising safety pr<strong>of</strong>ile <strong>of</strong> MK1775 strongly suggest that this drug can be<br />

used as a potential therapeutic agent alone or in combination with<br />

gemcitabine in the treatment <strong>of</strong> both adult as well as pediatric sarcoma<br />

patients.<br />

10086 General Poster Session (Board #52G), Sun, 8:00 AM-12:00 PM<br />

Early assessment <strong>of</strong> MCV to predict clinical outcome in patients with<br />

advanced gastrointestinal stromal tumors (GIST) receiving imatinib. Presenting<br />

Author: Anastasia Constantinidou, The Royal Marsden Hospital NHS<br />

Foundation Trust and the Institute <strong>of</strong> Cancer Research, London, United<br />

Kingdom<br />

Background: In patients with advanced GIST, imatinib trough levels have<br />

been associated with a lower rate <strong>of</strong> clinical benefit. Anecdotal observations<br />

have suggested that commencing treatment with imatinib may be associated<br />

with an increase in red blood cell size as measured by MCV and MCH,<br />

indicating the effect <strong>of</strong> imatinib on KIT signalling in the bone marrow.<br />

Methods: In this retrospective review we examined a possible connection<br />

between changes in MCV and MCH after the first 3 months (mo) <strong>of</strong><br />

treatment with imatinib and progression-free survival (PFS). Data for<br />

patients with inoperable advanced or metastatic GIST treated between<br />

2000 and 2011 were available for analysis. Patients were categorised in<br />

quartiles according to the distribution <strong>of</strong> the percentage <strong>of</strong> change <strong>of</strong> MCV<br />

and MCH between start <strong>of</strong> imatinib (baseline) and 3 (�/- 0.5) mo (ratio<br />

%�MCV change at 3 mo/MCV at baseline). PFS curves were plotted using<br />

the Kaplan-Meier method and compared using the log rank test. Results:<br />

One hundred and thirty patients were eligible for analysis and the<br />

demographics were: male:female 84:46, median age at presentation: 60.5<br />

(23.5-86.5) years, commonest primary tumour sites: stomach (72/130)<br />

and small bowel (44/130), commonest metastatic site: liver (76/130). In<br />

the patients with a 10% or over increase in MCV (p75-p100) at the 3 mo<br />

(�/-0.5) time point a significantly longer PFS was observed compared to<br />

those with a smaller magnitude <strong>of</strong> change (PFS <strong>of</strong> 37.1 mo vs 24.3 mo,<br />

p�0.032). A similar trend although not statistically significant was<br />

observed in patients with MCH <strong>of</strong> 15% or over (p75-p100) with PFS <strong>of</strong><br />

34.0 mo vs 25.8 (p�0.125). The two parameters (MCV and MCH) showed<br />

a high correlation <strong>of</strong> percentage change (r2�0.85). The patients with a<br />

10% MCV increase and /or 15% MCH increase (total�40) showed a<br />

significantly longer PFS (34.0 mo) compared to those with lower percentage<br />

change (PFS� 24.3) p�0.035. Conclusions: Simple laboratory parameters<br />

may be indicators <strong>of</strong> imatinib pharmacokinetics and may therefore be<br />

used as surrogate markers <strong>of</strong> clinical outcome. Larger prospective studies<br />

are required to confirm the results <strong>of</strong> this study.<br />

Sarcoma<br />

651s<br />

10085 General Poster Session (Board #52F), Sun, 8:00 AM-12:00 PM<br />

Pharmacokinetics <strong>of</strong> escalated dose <strong>of</strong> imatinib in patients with advanced<br />

gastrointestinal stromal tumors. Presenting Author: Changhoon Yoo, Department<br />

<strong>of</strong> Oncology, Asan Medical Center, University <strong>of</strong> Ulsan College <strong>of</strong><br />

Medicine, Seoul, South Korea<br />

Background: Although escalated dose (ED, 800 mg daily) <strong>of</strong> imatinib (IM) is<br />

one <strong>of</strong> important options in resistant gastrointestinal stromal tumors (GIST)<br />

on standard dose (SD, 400mg daily), there has been lack <strong>of</strong> pharmacokinetic<br />

data at ED. We therefore explore the potential relationship between<br />

IM plasma trough level (Cmin), and clinical outcomes or toxicities in<br />

patients treated with ED. Methods: Between 2008 and 2011, steady-state<br />

IM Cmin was measured in 66 patients with GIST who received ED <strong>of</strong> IM<br />

using liquid chromatography-tandem mass spectrometry. Percent change<br />

from IM Cmin at SD was calculated in 43 patients that both SD and ED IM<br />

Cmin were measured. The association with efficacy and toxicity was<br />

analyzed with grouping patients into quartiles according to IM Cmin at ED<br />

and percent change. Results: Median age was 59 years (range, 36–77) and<br />

40 patients were male. KIT exon 11 and 9 mutation was detected in 32 and<br />

18 patients, respectively. At ED, partial response was achieved in 4<br />

patients (6%) and stable disease was in 32 patients (48%). The median<br />

progression-free survival was 4.2 months (95% CI, 1.8-6.6) and overall<br />

survival was 38.5 months (95% CI, 7.6-69.3). The mean (� standard<br />

deviation) IM Cmin at ED was 3552 (� 1540) ng/mL. IM Cmin was<br />

significantly increased after dose escalation (p �0.001), and mean percent<br />

change from IM Cmin at SD was 162% (� 100). Body surface area<br />

(p�0.01), hemoglobin (p�0.001), and neutrophil count (p�0.006) were<br />

significant covariates for IM Cmin at ED in multivariate analysis. The group<br />

<strong>of</strong> quartiles <strong>of</strong> IM Cmin at ED and percent change was not associated with<br />

response and survival outcomes. However, grade 3-4 hematologic or grade<br />

2-4 non-hematologic toxicity was observed in 9% (1/11) <strong>of</strong> patients in the<br />

lowest percent change quartile (Q1, �90%) compared with 56% (18/32)<br />

in Q2 to Q4 (p�0.012), although absolute values were not associated with<br />

toxicity. Conclusions: In GIST patients treated with ED <strong>of</strong> IM, IM Cmin was<br />

not associated with response and survival. <strong>Clinical</strong>ly significant toxicity<br />

following dose escalation was correlated with percent change <strong>of</strong> IM Cmin, rather than absolute values. Monitoring <strong>of</strong> IM Cmin might help to predict or<br />

manage ED <strong>of</strong> IM induced toxicity.<br />

10087 General Poster Session (Board #52H), Sun, 8:00 AM-12:00 PM<br />

SDHA and SDHB mutations in KIT/PDGFRA WT gastrointestinal stromal<br />

tumors. Presenting Author: Margherita Nannini, Seragnoli Department and<br />

GIST Study Group, University <strong>of</strong> Bologna, Bologna, Italy<br />

Background: KIT/PDGFRA wild-type (WT) GISTs harbour mutations on<br />

SDHB and SDHC and, more recently, we described mutations on SDHA<br />

using massively parallel sequencing approach. We sequenced SDHA and<br />

SDHB genes in a larger series in order to validate the data. Methods: SDHA<br />

gene (1-15 exons) and SDHB gene (1-8 exons) (even not all exons in all<br />

samples) were sequenced on tumor (T) and/or peripheral blood (PB) <strong>of</strong> WT<br />

GIST patients by Sanger Sequencing method. DNA was extracted from<br />

tumor specimens by the QIAmp DNA Mini kit (Qiagen, Milan, Italy) and<br />

amplified with specific primer pairs designed to amplify exons but not<br />

SDHA pseudo-genes located on chromosomes 3 and 5. Then, PCR products<br />

were purified with the Qiaquick PCR purification kit (Qiagen, Milan, Italy)<br />

and sequenced on both strands using the Big Dye Terminator v1.1 Cycle<br />

Sequencing kit (Applied Biosystems). Sanger sequencing was performed<br />

on ABI 3730 Genetic Analyzer (Applied Biosystems). Results: SDHA gene<br />

exons were sequenced on a total <strong>of</strong> 27 WT GIST patients, in particular on T,<br />

PB and both from 12, 6 and 9 patients respectively. SDHB gene exons were<br />

sequenced on a total <strong>of</strong> 18 out <strong>of</strong> 27 patients, in particular on T, PB and<br />

both from 7, 8 and 3 patients respectively. 8 SDHA mutations were found<br />

in 5 samples (18.5%). Besides those previously identified, 5 new SDHA<br />

mutations were found in other 3 samples: one sample harboured R171C<br />

and R589Q heterozygous missense mutation in exons 5 and 13 respectively.<br />

The other one harboured G419R and E564K heterozygous missense<br />

mutations in exons 9 and 13 respectively. The third sample harboured a<br />

delCAG immediately upstream <strong>of</strong> exon 5, in heterozygosis on PB and in<br />

homozygosis on T. A SDHB heterozygous mutation (301delT) in exon 4 was<br />

found on 1 PB sample. Conclusions: the presence <strong>of</strong> SDHA mutations has<br />

been confirmed in a subgroup <strong>of</strong> WT GIST patients. All subunits <strong>of</strong> SDH<br />

complex should be sequenced on WT GIST patients in order to explore the<br />

frequency and any linkage between each other and the pathogenetic and<br />

clinical significance.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


652s Sarcoma<br />

10088 General Poster Session (Board #53A), Sun, 8:00 AM-12:00 PM<br />

Diagnosis and initial evaluation <strong>of</strong> patients with gastrointestinal stromal<br />

tumor (GIST): An observational study <strong>of</strong> 1,226 patients. Presenting Author:<br />

Jonathan C. Trent, University <strong>of</strong> Miami Sylvester Comprehensive Cancer<br />

Center, Miami, FL<br />

Background: The GIST registry (reGISTry) is an observational database<br />

designed to determine diagnostic method and evaluation <strong>of</strong> GIST patients<br />

outside <strong>of</strong> clinical trials. We previously described differences in treatment<br />

<strong>of</strong> patients in community versus academic centers (Pisters, Ann Oncol<br />

2011;22:2523-9). Herein we present distinct differences in the diagnosis<br />

and initial evaluation <strong>of</strong> patients with GIST. Methods: The analysis cut<strong>of</strong>f<br />

date was May 2010. Patient demographics, presenting symptoms, diagnostic<br />

methodology, and KIT and PDFGRA mutations were described. For<br />

localized tumors, size and mitotic index (MI) were collected. Results: 1226<br />

patients were included in this analysis. Initial diagnosis was most commonly<br />

made by surgeons (54.6%) and gastroenterologists (15.7%). Primary<br />

management decisions were made at time <strong>of</strong> diagnosis by medical<br />

oncologists (53%) and surgeons (53%) (sum �100% due to �1 responses/<br />

site). 84% were symptomatic at diagnosis, while 16% were diagnosed<br />

incidentally. Diagnostic procedure was performed during surgery (75%)<br />

and/or by endoscopy (22%) or interventional radiology (11%) using<br />

percutaneous core biopsy or fine needle aspirate. Initial diagnostic imaging<br />

was by CT scan (76%), endoscopy (25%), ultrasound (12%), or a<br />

combination. Most primary GISTs were located in the stomach (55%) or<br />

small intestine (28%). At diagnosis, localized and metastatic disease was<br />

seen in 83% and 17% <strong>of</strong> patients, respectively, with metastases mainly in<br />

the liver (62%) and/or peritoneum (36%). KIT immunostaining was<br />

performed in 93% <strong>of</strong> patients, with 98% positive. Mutation analysis was<br />

carried out in 8% <strong>of</strong> cases (57% KIT exon 11 and 12% exon 9 mutations).<br />

Of localized primary tumors, 52% were �5 cm in size and 23% had �5<br />

mitoses per 50 high-power fields. Conclusions: We found that initial<br />

diagnosis is made by several different modalities. The majority <strong>of</strong> GIST<br />

patients are managed by medical and surgical oncologists. Most patients<br />

with localized tumor were at intermediate risk <strong>of</strong> recurrence and would be<br />

considered for adjuvant imatinib therapy.<br />

10090 General Poster Session (Board #53C), Sun, 8:00 AM-12:00 PM<br />

Outcomes <strong>of</strong> multivisceral resection <strong>of</strong> gastric gastrointestinal stromal<br />

tumors. Presenting Author: Sabha Ganai, University <strong>of</strong> Chicago Medical<br />

Center, Chicago, IL<br />

Background: Despite the recent introduction <strong>of</strong> imatinib and laparoendoscopic<br />

techniques to the management <strong>of</strong> gastric gastrointestinal stromal<br />

tumors (GISTs), outcomes remain uncertain in the setting <strong>of</strong> multivisceral<br />

involvement. Methods: We conducted a retrospective review <strong>of</strong> 73 consecutive<br />

patients who underwent resection <strong>of</strong> gastric GISTs from October 2002<br />

through December 2011. Median follow-up was 22 months (interquartile<br />

range [IQR] 6-37). Results: Patients were 51% female, with a mean age <strong>of</strong><br />

65 � 12 years and BMI <strong>of</strong> 30 � 8 kg/m2 . Patients undergoing multivisceral<br />

resection (n�14) had a longer interval from diagnosis to surgery (7.3 [IQR<br />

1.9 – 15.0] vs. 1.3 [IQR 0.7-4.2] months, p�0.01), greater use <strong>of</strong><br />

neoadjuvant imatinib (64% vs. 3%, p�0.0001), and greater preoperative<br />

tumor size (12 � 8 vs. 4 � 3 cm, p�0.0001) in comparison to gastric-only<br />

resections (n�59). Patients were less likely to be managed laparoscopically<br />

(7% vs. 71%, p�0.0001), had a longer operative time (310 � 117<br />

vs. 145 � 62 min, p�0.0001), and were less likely to be R0 (71% vs.<br />

98%, p�0.001). While patients undergoing multivisceral resection were<br />

more likely to have a pathological complete response to therapy (29% vs. 0,<br />

p�0.001), they were also more likely to have metastatic disease present<br />

(29% vs. 0, p�0.001). Hospital length <strong>of</strong> stay was greater (median 8 [IQR<br />

6-9] vs. 3 [IQR 2-6] days, p�0.0001). There were no significant differences<br />

in grade or mitotic index between groups. There was greater use <strong>of</strong><br />

adjuvant imatinib (64% vs. 25%, p�0.05). Overall survival was less in<br />

patients undergoing multivisceral resection (64% vs. 87% at 3 years,<br />

p�0.05), as was disease-free survival (52% vs. 71% at 3 years, p�0.05).<br />

Median overall and disease-free survival were 43 and 22 months after<br />

multivisceral resection for gastric GISTs. Controlling for tumor size, grade,<br />

resection status, and the use <strong>of</strong> neoadjuvant imatinib, multivisceral<br />

resection and use <strong>of</strong> adjuvant imatinib were both independent predictors <strong>of</strong><br />

disease-free survival (p�0.05). Conclusions: Multivisceral involvement is<br />

associated with tumors <strong>of</strong> greater size and, despite an increased use <strong>of</strong><br />

neoadjuvant imatinib, it is associated with poor outcome for patients with<br />

gastric GISTs.<br />

10089 General Poster Session (Board #53B), Sun, 8:00 AM-12:00 PM<br />

Masitinib in imatinib-naive advanced gastrointestinal stromal tumor (GIST):<br />

Five-year follow-up <strong>of</strong> the French Sarcoma Group phase II trial. Presenting<br />

Author: Axel Le Cesne, Institut Gustave Roussy, Villejuif, France<br />

Background: Masitinib is a tyrosine kinase inhibitor that in vitro has greater<br />

activity and selectivity than imatinib against KIT. This multicenter, open<br />

label, phase 2 study evaluated efficacy and safety <strong>of</strong> masitinib as a first-line<br />

treatment <strong>of</strong> advanced GIST. Initial results with a median follow-up <strong>of</strong> 34<br />

months, were previously reported in EJC 2010. We present here 5-year<br />

follow-up data from the same series with updated safety and survival data.<br />

Methods: Imatinib-naïve patients (pts) with inoperable, advanced GIST<br />

received oral masitinib (7.5 mg/kg/day) until progression, refusal or<br />

toxicity. Results: Thirty pts with a median age <strong>of</strong> 58 years (60% <strong>of</strong> males)<br />

were included from 06/2005 to 04/2007 in 5 institutions. At the cut-<strong>of</strong>f<br />

date <strong>of</strong> 13/09/2011, 7/30 pts (23%) were still under treatment with<br />

median follow-up <strong>of</strong> 65 months and the median overall survival (OS) had<br />

not yet been reached (NR). The 5-year OS rate was 61.5% (95% CI<br />

[41.1;76.6]) (see Table). Among patients with confirmed KIT exon 11<br />

mutation, 8/9 pts (89%) were still alive with one pt having died from<br />

non-treatment related causes (surgical complication) following a complete<br />

response while receiving masitinib. No additional progressions have been<br />

reported giving a total <strong>of</strong> 14 events (13 progressions and 1 death). Updated<br />

median PFS was 41.3 months (95% CI [17.5;53.8]).No additional grade<br />

3/4 adverse events have been reported. Conclusions: Long term results <strong>of</strong><br />

masitinib confirm an interesting activity with prolonged PFS and OS. The<br />

median PFS rate in masitinib compares very favorably to that <strong>of</strong> imatinib,<br />

which is reported as 18 months (JCO 26:626, 2008). The 5-year OS rates<br />

for masitinib in the overall and exon 11 populations also compare favorably<br />

to imatinib, both <strong>of</strong> which are reported at �50% for imatinib (JCO 26:626,<br />

2008; JCO 28:1247, 2010). These results support the head to head<br />

comparison with imatinib in the currently ongoing phase 3 randomized<br />

clinical trial in first line advanced GIST pts.<br />

All (N�30) Exon 11 (n�9)<br />

Median OS (months) [95%CI] NR [53;NR] NR [65;NR]<br />

Month-36 86.5% [68.0;94.7] 88.9% [43.3;98.4]<br />

48 76.2% [56.4;87.9] 88.9%<br />

60 61.5% [41.1;76.6] 88.9%<br />

72 55.9% [34.7;72.6] 71.1% [23.3;92.3]<br />

10091 General Poster Session (Board #53D), Sun, 8:00 AM-12:00 PM<br />

Single-dimension CT measurements with RECIST 1.1 to evaluate liver<br />

metastases in GIST patients on imatinib. Presenting Author: Gaia Schiavon,<br />

Department <strong>of</strong> Medical Oncology, Erasmus University Medical Center -<br />

Daniel den Hoed Cancer Center, Rotterdam, Netherlands<br />

Background: Tumor response assessment to therapy is crucial in oncology,<br />

both in daily practice and research.We analyzed the morphology <strong>of</strong> liver<br />

metastases (LM) in gastrointestinal stromal tumors (GIST). Aims were to 1)<br />

determine whether uni-dimensional measurements <strong>of</strong> tumor lesions (according<br />

to RECIST 1.1) are accurate reflections <strong>of</strong> their volumes and 2)<br />

estimate eventual bias in volume change calculations, as introduced by<br />

using only the longest diameter. Methods: 78 GIST patients with LM were<br />

evaluated at baseline (n�139 lesions), 3 (n�129), 6 (n�128), and 12<br />

months (n�108) after imatinib therapy. LM were segmented semiautomatically<br />

on standard CT scans. All measurements at baseline were<br />

obtained by 2 independent investigators and agreement between observers<br />

was assessed with Bland-Altman plots. Values for segmented volume (Vs;<br />

mL), maximum transaxial diameter (MTD; mm), and elongation value (EV,<br />

defined as 1 - [width/length], where EV <strong>of</strong> 1 � fully ellipsoidal and 0 � fully<br />

spherical) were reported for each lesion at several time-points. A calculated<br />

volume (Vc) <strong>of</strong> each LM was also generated using the MTD, with the<br />

hypothesis that metastases were fully spherical. Results: At baseline, 44%<br />

(61/139) <strong>of</strong> the LM was spherical and 56% (78/139) was ellipsoidal. Their<br />

EV ranged from 0.08 to 0.94 (mean, 0.53�0.17). During treatment,<br />

overall 42% <strong>of</strong> the lesions kept their morphology (spherical or ellipsoidal)<br />

and the other half changed from spherical to ellipsoidal or vice versa. At<br />

baseline, there was strong inter-observer agreement in the determination <strong>of</strong><br />

Vs, with mean inter-observer variability <strong>of</strong> 1.5% (95% CI, -14.5% to<br />

17.5%). The difference between Vs and Vc was highly significant<br />

(P�0.0001). Vc overestimates the actual volume <strong>of</strong> 35% (95% CI, -26%<br />

to 95%). Conclusions: LM are not always spherical and can change their<br />

morphology during imatinib therapy. Therefore, a lesion’s single largest<br />

trans-axial dimension, as used in RECIST 1.1, is not an accurate surrogate<br />

<strong>of</strong> its actual volume. Further studies are warranted to fully understand the<br />

clinical impact <strong>of</strong> these findings and to assess whether we should apply<br />

different criteria for response assessment to therapies in solid tumors.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


10092 General Poster Session (Board #53E), Sun, 8:00 AM-12:00 PM<br />

Explored prognostic factors for survival in patients with advanced GIST<br />

treated with standard dose imatinib (IM): Results from the BFR14 phase III<br />

trial <strong>of</strong> the French Sarcoma Group. Presenting Author: David Pérol, Centre<br />

Léon Berard, Lyon, France<br />

Background: Factors predicting progression free survival (PFS) and overall<br />

survival (OS) <strong>of</strong> patients (pts) with advanced GIST treated with 400 mg<br />

daily dose IM included in the BFR14 study with a median follow up <strong>of</strong> 54<br />

months (CI95%:47-61) was investigated evaluating added prognostic<br />

factors. Methods: 434 pts were included in this prospective multicenter<br />

trial from June 2002 to July 2009. After 1, 3 and 5 yrs <strong>of</strong> IM 400mg/day,<br />

pts free from progression were randomly <strong>of</strong>fered to continue or interrupt (I<br />

arm) IM. Prognostic factors for overall survival were investigated in the<br />

entire cohort (randomized and not randomized pts) included in the BFR14.<br />

Survival was defined from the date <strong>of</strong> inclusion to the date <strong>of</strong> death for OS or<br />

to the first occurrence <strong>of</strong> disease progression under imatinib or death for<br />

PFS. A multivariate cox model including statistical significant baseline<br />

characteristics tested in univariate were included in a backward procedure<br />

d to identify independent prognostic factors for PFS then OS. Results: As <strong>of</strong><br />

January 2012, there were 285 progressions (65%) and 161 deaths (37%).<br />

The median PFS <strong>of</strong> the entire cohort was 29 months (CI95%: 24-33) and<br />

the 4 and 5-yrs PFS were 31% and 24% respectively. A low tumour volume<br />

at inclusion, PS (0), sex (female), CD34 positivity on tumor cells were the<br />

four independent prognostic factors <strong>of</strong> a higher PFS. The 5-yrs OS was 54%<br />

(CI95%: 48-60). A higher OS was independently predicted by gender<br />

(female), PS (0), platelets count (�400 giga/L) and CD34 expression on<br />

GIST cells. Median PFS and OS were slightly superior in this more recent<br />

series as compared to B2222 consistently with the improved outcome <strong>of</strong><br />

patients with low tumor volume. Conclusions: OS and PFS are predicted by<br />

gender, PS and CD34 expression in this large series <strong>of</strong> pts treated with<br />

standard dose IM. The biological significance <strong>of</strong> CD34 expression on tumor<br />

cells has to be explored in GIST.<br />

10094 General Poster Session (Board #53G), Sun, 8:00 AM-12:00 PM<br />

Characteristics <strong>of</strong> gastrointestinal stromal tumor (GIST) patients receiving<br />

short-term versus long-term imatinib (IM) adjuvant therapy: A chart review<br />

analysis. Presenting Author: Annie Guerin, Analysis Group, Inc., Boston, MA<br />

Background: In clinical practice, significant variability is seen in duration <strong>of</strong><br />

adjuvant IM use. The objective <strong>of</strong> this study is to compare characteristics <strong>of</strong> GIST<br />

pts receiving adjuvant IM for a short (6-12 months) vs extended period (�24<br />

months) to better understand factors that may influence treatment (trt) duration<br />

decisions. Methods: Physician prescribing patterns and clinical information on<br />

adult pts with primary resectable Kit positive GIST initiating IM �84 days<br />

post-surgery was collected from 248 U.S. oncologists using online data<br />

collection forms. In addition to physicians’ perception <strong>of</strong> short- vs long-term use,<br />

pts’ risk assessment, trt, demographics, and comorbidities were collected for<br />

246 short-term and 395 long-term IM pts. Characteristics were compared using<br />

Wilcoxon and Chi-square tests. Results: While pts were similar in age [59.0 vs.<br />

58.1, P �.23], ethnicity, and region <strong>of</strong> residence, the short-term group included<br />

fewer males (57.7% vs 69.6%, P �.01) and had a higher prevalence <strong>of</strong><br />

cardiovascular (11.4% vs 5.8%, P � .01) and ischemic heart diseases (5.3% vs<br />

1.5%, P�.01). Differences were also observed in indicators <strong>of</strong> pre-treatment risk<br />

pr<strong>of</strong>ile (tumor size, location, and rupture during surgery, mitotic count, and<br />

Miettinen score) (Table). Findings were consistent with main reasons reported by<br />

physicians for prescribing adjuvant IM over longer duration; in addition to pt risk<br />

pr<strong>of</strong>ile (76.6%), tolerability (70.6%), younger pts (59.7%), safety (39.1%), trt<br />

response (29.8%), and economic reasons (26.2%) were other reasons impacting<br />

trt decisions. Conclusions: Pt risk is an important factor in physicians’ decisions<br />

to prescribe adjuvant IM for extended duration. However, age, tolerability, and<br />

comorbidities, also play an important role.<br />

Long-term Short-term<br />

Tumor characteristics<br />

Size (mean � SD)<br />

Rupture during surgery, %<br />

8.2 � 7.0 7.2 � 7.6 *<br />

Yes 2.5 5.7 *<br />

No 91.9 92.3<br />

Unknown<br />

Location, %<br />

5.6 2.0 *<br />

Stomach 53.9 44.3 *<br />

Small intestine 29.6 27.6<br />

Rectum 1.3 2.0<br />

Other 15.2 26.0 *<br />

Miettinen risk (mean � SD)<br />

Mitotic rate (/HPF; %)<br />

0.4 � 0.3 0.3 � 0.3 *<br />

< 5 9.6 16.7 *<br />

5 30.9 33.3<br />

6–10 33.7 31.3<br />

>10 17.7 11.8 *<br />

N/A<br />

* Significant at 5% level.<br />

8.1 6.9<br />

Sarcoma<br />

653s<br />

10093 General Poster Session (Board #53F), Sun, 8:00 AM-12:00 PM<br />

The role <strong>of</strong> surgical cytoreduction before imatinib therapy in patients with<br />

advanced GIST. Presenting Author: Hojung An, Division <strong>of</strong> Medical<br />

Oncology, Asan Medical Center, Seoul, South Korea<br />

Background: Despite dramatic improvement <strong>of</strong> survival with introduction <strong>of</strong><br />

imatinib mesylate, resistance to imatinib eventually develops in most <strong>of</strong><br />

patients with advanced gastrointestinal stromal tumor (GIST). It is well<br />

known that baseline tumor size is an important factor related to imatinib<br />

resistance. We hypothesize that decreasing tumor size by surgery before<br />

starting imatinib may delay the emergence <strong>of</strong> resistance and improve<br />

prognosis in patients with advanced GIST. Methods: From 2001 to 2010,<br />

102 patients with initially metastatic GIST and 147 patients with recurrent<br />

GIST were enrolled in this analysis. Patients with local relapse only were<br />

excluded because they were potentially curable by surgery alone. Patients<br />

were categorized into two groups according to the extent <strong>of</strong> cytoreduction;<br />

i.e., patients whose initial tumor bulk reduced �75% surgically before<br />

starting imatinib (group A) and the others (group B). Results: Among total<br />

249 patients, 62 patients received initial surgery. 35 (14%) patients<br />

whose tumor bulk reduced � 75% were categorized into group A, and the<br />

remaining 214 (86%) were in group B. In group A, the median age was<br />

younger; more patients were initially metastatic; peritoneal metastases<br />

were more frequent; but liver metastases were less. The total tumor size was<br />

significantly reduced from median 122 mm before cytoreduction to 0 mm<br />

after on CT scans in group A. With a median follow-up <strong>of</strong> 42.7 months,<br />

progression-free survival (PFS) tended to be better in group A than in group<br />

B in univariate analysis (HR�0.60; 95% CI, 0.36-1.02; p�0.061), but<br />

PFS was not statistically different between the two groups in multivariate<br />

analysis (p�0.488). Meanwhile, absence <strong>of</strong> KIT exon 11 mutation<br />

(p�0.007), baseline total tumor size � 150mm (p�0.043), and granulocyte<br />

count � 5000/mm3 (p�0.009) remained independent poor prognostic<br />

factors. Conclusions: Despite marked decrease <strong>of</strong> total tumor size, the<br />

outcomes were not significantly improved in patients receiving initial<br />

cytoreduction more than 75% <strong>of</strong> baseline tumor bulk before starting<br />

imatinib. These results suggest that initial cytoreduction does not have<br />

beneficial role in advanced GIST, so imatinib should be still the first<br />

treatment <strong>of</strong> choice in this population.<br />

10095 General Poster Session (Board #53H), Sun, 8:00 AM-12:00 PM<br />

Effect <strong>of</strong> five years <strong>of</strong> imatinib on cure for patients with advanced GIST:<br />

Updated survival results from the prospective randomized phase III BFR14<br />

trial. Presenting Author: François Bertucci, Institut Paoli-Calmettes, Marseille,<br />

France<br />

Background: We previously demonstrated that interruption <strong>of</strong> imatinib (IM)<br />

after 1, 3, and 5 yrs in responding patients (pts) with advanced GIST is<br />

associated with rapid relapse but reintroduction <strong>of</strong> IM allowed tumor<br />

control in almost all pts. Here, we examined the outcome <strong>of</strong> patients<br />

randomized in the interruption arm (I arm), and notably the characteristics<br />

<strong>of</strong> those not yet progressing. Methods: This prospective multicenter BFR14<br />

study was initiated in June 2002 and closed for inclusion in July 2009.<br />

Seventy-one non-progressing patients were randomized in the I arm after 1<br />

(N�32), 3 (N�25) and 5 years (N�14) <strong>of</strong> IM 400 mg/day. IM (same dose)<br />

was reintroduced in case <strong>of</strong> progressive disease (PD). Results: The median<br />

follow-up (FU) from randomization was 74, 48 and 22 months for pts<br />

randomized at 1, 3 and 5 yrs <strong>of</strong> IM treatment respectively. Updated survival<br />

data (January 2012) are summarized in the table. Out <strong>of</strong> the 3 pts not<br />

progressing in the I arm at 1 yr, 1 pt had refused to stop IM and 1 pt had a<br />

localized GIST with small residual disease at inclusion. Out <strong>of</strong> the 3 pts not<br />

progressing in the I arm at 3 yrs, 1 pt had refused to stop IM and 2 pts were<br />

included after complete resection <strong>of</strong> both primary tumor and metastases<br />

with a small residual disease. The FU <strong>of</strong> pts randomized at 5 yrs was short<br />

but out <strong>of</strong> the 5 non progressive pts, 2 are considered in PD on functional<br />

imaging (January 2012), 2 had a locally advanced GIST subsequently<br />

operated during IM and before randomization, and 1 had no target lesion at<br />

inclusion (resection <strong>of</strong> synchronous metastases). Conclusions: All but one<br />

pts with residual masses under IM and randomized in the I arm have<br />

relapsed. Six out <strong>of</strong> 7 patients not yet progressing in the I arm had reached<br />

complete remission following surgery at inclusion or before randomization<br />

(initial resection/debulking <strong>of</strong> metastases).<br />

Parameter 1 yr 3 yrs 5 yrs<br />

Randomized pts (I/continuous [C] arms) 58 (32/28) 50 (25/25) 27 (14/13)<br />

Median PFS (months) I/C arms<br />

7/29<br />

9/49<br />

13/not reached<br />

(p)<br />

p�0.0001<br />

p�0.0001<br />

p�0.017<br />

2-y PFS (%) : I/C arms 13 [4-26] p / 11[2-27]p/80[58-91] 28[6-57]p/<br />

62 [40-77]<br />

92 [54-99]<br />

No. pts non-progressive (I arm) (%) 3/32(9) 3/25(12) 5/14(35)<br />

Post-randomization FU (months) 64, 73, 90 23, 46, 54 2, 3, 17, 18, 25<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


654s Sarcoma<br />

10096 General Poster Session (Board #54A), Sun, 8:00 AM-12:00 PM<br />

Analysis <strong>of</strong> the quality <strong>of</strong> reporting surgical procedures in patients undergoing<br />

resection for primary gastrointestinal stromal tumors: A reporting tool<br />

derived from the EORTC–STBSG 62024 trial. Presenting Author: Peter<br />

Hohenberger, Universitätsklinikum Mannheim, Mannheim, Germany<br />

Background: Features <strong>of</strong> surgery for the primary tumor (rupture, extent and<br />

completeness <strong>of</strong> resection) contribute significantly on the indication for<br />

adjuvant treatment in patients with GIST. No reporting tool for surgery for a<br />

primary GIST has been established. EORTC 62024 is a phase III trial<br />

evaluating the use <strong>of</strong> adjuvant imatinib in GIST and was used for an<br />

analysis <strong>of</strong> the data <strong>of</strong> surgical procedures. Methods: 908 pts. from 11<br />

countries were recruited. At study entry, a copy <strong>of</strong> the surgical record and<br />

pathology report had to be submitted in addition to the case record forms<br />

(CRFs) which included a surgical questionnaire on 19 items on the<br />

circumstances <strong>of</strong> the resection (elective/emergency, GIST diagnosis established<br />

preop., tumor rupture, R-status, concordance <strong>of</strong> preop. and intraop.<br />

staging, surgical technique, complications). We then reviewed the original<br />

documents and compared them with the documentation on the CRFs. The<br />

full set <strong>of</strong> data was available from 793/906 patients (87.3%). Results: The<br />

diagnosis <strong>of</strong> GIST was known preop. in 58% <strong>of</strong> the cases, and 12% <strong>of</strong> the<br />

pts. had been treated as an emergency. The R0 resection rate was 87%.<br />

The incidence <strong>of</strong> tumor rupture was 9% and 24% <strong>of</strong> the pts. had undergone<br />

lymphadenectomy. The consistency between pre- and intraoperative findings<br />

was 82%. The concordance between CRFs and original documents<br />

was 77%. Concordance <strong>of</strong> the original documents with the surgical<br />

questionnaire (CRFs) was critical in 4/19 items. Discrepancies and<br />

misinterpretation rates were highest when data managers from medical<br />

oncologists had completed the CRFs based on letters <strong>of</strong> patient discharge.<br />

Based on the evaluation, the surgical reporting tool can be refined and<br />

shortened to 13 items for future applications. Conclusions: Findings at<br />

surgery for the primary tumor not properly reported might significantly<br />

influence the decision <strong>of</strong> whether or not to treat patients with adjuvant<br />

imatinib. The refined surgical reporting tool could be helpful to gather all<br />

relevant information about the intraoperative elements <strong>of</strong> surgery for the<br />

primary GIST. Surgeons are to be asked to report those data themselves.<br />

10098 General Poster Session (Board #54C), Sun, 8:00 AM-12:00 PM<br />

Doxorubicin, cisplatin, and ifosfamide (API) as first-line therapy for<br />

relapsed or metastatic uterine leiomyosarcoma. Presenting Author: Julien<br />

Hadoux, Institut Gustave Roussy, Villejuif, France<br />

Background: Uterine leiomyosarcomas (ULMS) are rare gynecologic malignancies<br />

characterized by a poor prognosis due to a high rate <strong>of</strong> local and<br />

metastatic recurrences. Chemotherapy (CT) with doxorubicin or ifosfamide<br />

or both is associated with a 10 to 30% objective response rate (ORR) and a<br />

cisplatin-based multiCT approach achieved a good response rate (DECAV<br />

therapy: API � dacarbazine � vindesine, 54% ORR in uterine sarcomas),<br />

though toxic. We aimed to determine efficacy and toxicity <strong>of</strong> doxorubicin,<br />

cisplatin and ifosfamide (API) combination as first line treatment <strong>of</strong><br />

metastatic or relapsed ULMS (MRULMS). Methods: This monocentric study<br />

included MRULMS pts with a physiological age � 65 y. CT consisted in<br />

doxorubicin 50 mg/m² d1, ifosfamide 3 g/m²/d d1d2 � mesna, cisplatin 75<br />

mg/m² d3, � G-CSF; q 3 weeks. Results: Results in 38 pts with MRULMS<br />

were analyzed; median age was 51 (40-64), median cycles <strong>of</strong> CT was 5; 8<br />

(21%) pts were treated for local relapse, 21 (55.3%) for metastatic disease<br />

and 9 (23.7%) for both. Metastatic sites were: lungs in 16 pts (42.1%),<br />

pelvis in 7 pts (18.4%), liver in 7 pts (18.4%), peritoneum in 6 pts<br />

(15.8%) and bone in 5 pts (13.2%); 14 pts (36.8%) had a multisite<br />

metastatic disease. Main grade 3-4 toxicities in 38 pts were neutropenia<br />

(74%), thrombopenia (60%), anemia (55%), fatigue (18%) and vomiting<br />

(13%). Febrile neutropenia was observed in 35% <strong>of</strong> pts and 1 patient died<br />

<strong>of</strong> septic shock after cycle 1. Thirty four pts were evaluable for response (4<br />

pts had complete surgery at relapse) and 16 pts responded (4 CR � 12 PR)<br />

(ORR: 47%); 23.5% and 29.4% <strong>of</strong> the pts had respectively stable and<br />

progressive disease. For all pts (38) and evaluable pts (34), median PFS<br />

were 9.8 and 9.5 months and OS 27 and 25.3 months respectively.<br />

Conclusions: Despite toxicity observed, API is an effective treatment which<br />

compares favorably with other first line therapies for MRULMS pts.<br />

10097 General Poster Session (Board #54B), Sun, 8:00 AM-12:00 PM<br />

Genome-wide analysis and characterization <strong>of</strong> an online sarcoma cohort.<br />

Presenting Author: Kimberly E. Barnholt, 23andMe, Inc., Mountain View,<br />

CA<br />

Background: Recruitment <strong>of</strong> an adequately sized cohort for genome-wide<br />

studies presents a serious challenge for rare diseases such as sarcoma.<br />

Traditional barriers to participation include proximity <strong>of</strong> clinical centers<br />

and motivation or ability to travel. 23andMe’s web-based platform provides<br />

increased accessibility to research participation, facilitating rapid recruitment<br />

<strong>of</strong> patients (pts) and enabling a large-scale genome-wide association<br />

study (GWAS) <strong>of</strong> sarcoma. Methods: Sarcoma pts were recruited through<br />

web and email campaigns, patient advocacy groups, physician <strong>of</strong>fices, and<br />

events. Pts provide IRB-approved consent, complete surveys, and receive<br />

updates about research progress through an online account. In collaboration<br />

with an uncompensated panel <strong>of</strong> academic experts, an online survey<br />

was developed to collect patient-reported data on diagnosis, family history,<br />

symptoms and treatment. Results: This web-based approach has accrued<br />

the largest genotyped, recontactable sarcoma cohort to date. In 20 months,<br />

772 sarcoma pts have enrolled, 683 have been genotyped and 611 have<br />

provided data online. The cohort is primarily <strong>of</strong> European ancestry (92%),<br />

disproportionately female (72%), with an average age <strong>of</strong> 51 (� 15 years).<br />

More than 88% <strong>of</strong> pts indicated a s<strong>of</strong>t tissue sarcoma diagnosis, with<br />

leiomyosarcoma, liposarcoma and “malignant fibrous histiocytoma” being<br />

the most commonly reported subtypes. Over 36% <strong>of</strong> pts report undergoing<br />

active treatment <strong>of</strong> some type. Association scans were conducted across a<br />

set <strong>of</strong> 8,058,452 imputed SNPs, using 568 unrelated sarcoma cases <strong>of</strong><br />

European ancestry and �70,000 unrelated population controls from the<br />

23andMe database. Initial results have identified no significant genomewide<br />

associations for general sarcoma risk, despite having �90% power to<br />

detect risk variants with �5% minor allele frequency and odds ratio �2.5,<br />

suggesting the absence <strong>of</strong> common variants with strong shared effects<br />

across sarcoma subtypes. Conclusions: This pilot study demonstrates<br />

feasibility <strong>of</strong> rapid recruitment and longitudinal engagement <strong>of</strong> pts through<br />

online technology. Such techniques may significantly accelerate, and in<br />

some cases fully enable, large-scale genomic studies <strong>of</strong> sarcoma and other<br />

rare diseases.<br />

TPS10099 General Poster Session (Board #54D), Sun, 8:00 AM-12:00 PM<br />

A phase Ib/II study evaluating the efficacy <strong>of</strong> doxorubicin (D) with or<br />

without a human anti-PDGFR� monoclonal antibody olaratumab (IMC-<br />

3G3) in the treatment <strong>of</strong> advanced s<strong>of</strong>t tissue sarcoma (STS). Presenting<br />

Author: William D. Tap, Memorial Sloan-Kettering Cancer Center, New<br />

York, NY<br />

Background: PDGFR� is a receptor commonly overexpressed in STS.<br />

Olaratumab is a fully human IgG1 MAb which specifically binds to human<br />

PDGFR� and blocks PDGFR-mediated signaling pathways. Olaratumab<br />

demonstrated significant tumor inhibition as a monotherapy and in combination<br />

with standard chemotherapy in preclinical human sarcoma xenograft<br />

models. A Phase 1b/2 clinical trial for patients with advanced or<br />

metastatic STS is currently enrolling patients in 9 sites across the United<br />

States; approximately 45 patients have been randomized. Methods: In<br />

Phase 1b, patients received olaratumab (15 mg/kg) via intravenous<br />

infusion on Days 1 and 8 <strong>of</strong> each 21-day cycle and D (75 mg/m²) 1 hour<br />

after olaratumab on Day 1. Phase 1b was completed without encountering<br />

dose-limiting toxicities; enrollment into Phase 2 has begun. The primary<br />

endpoint <strong>of</strong> the Phase 2 portion is to compare progression-free survival in<br />

patients with advanced STS when treated with D plus olaratumab versus D<br />

alone. Planned enrollment goal for Phase 2 is 130 patients. Patients are<br />

required to be � 18 years old with ECOG 0-2, have appropriate organ<br />

function and histologically confirmed, measurable, and advanced STS.<br />

There is no restriction on the number <strong>of</strong> prior therapies. Patients are<br />

randomized 1:1 to either D plus olaratumab (Arm A) at same dose/schedule<br />

as in Phase 1b or D alone (Arm B). All patients can receive dexrazoxane.<br />

Upon completion <strong>of</strong> 8 cycles, patients in Arm A continue with olaratumab<br />

monotherapy. Patients in Arm B who develop disease progression during or<br />

after treatment can subsequently receive olaratumab monotherapy. Patients<br />

are stratified to treatment arms according to PDGFR� expression<br />

(positive vs. negative), number <strong>of</strong> previous lines <strong>of</strong> treatment, sarcoma<br />

subtype, and ECOG performance status. Response assessment occurs<br />

every 6 weeks. Exploratory analyses include biomarkers <strong>of</strong> olaratumab<br />

pharmacodynamic activity including PDGF ligands, PDGFR downstream<br />

molecules, VEGF, and changes in vascularity <strong>of</strong> tumor specimens.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


TPS10100 General Poster Session (Board #54E), Sun, 8:00 AM-12:00 PM<br />

Randomized phase III, multicenter, open-label study comparing TH-302 in<br />

combination with doxorubicin versus doxorubicin alone in subjects with<br />

locally advanced unresectable or metastatic s<strong>of</strong>t tissue sarcoma. Presenting<br />

Author: Sant P. Chawla, Sarcoma Oncology Center, Santa Monica, CA<br />

Background: A hypoxic microenvironment characterizes solid tumors including<br />

s<strong>of</strong>t tissue sarcoma (STS) and is associated with chemotherapy<br />

resistance. TH-302, a prodrug <strong>of</strong> the alkylating agent bromo-isophosphoramide<br />

mustard (BR-IPM) is reduced in hypoxic environments, releasing<br />

Br-IPM. Combining D with TH-302 should effectively target both the<br />

normoxic and hypoxic regions <strong>of</strong> STS. TH-302 was investigated with<br />

full-dose D (75 mg/m2 ). The regimen was well tolerated, �60% completed<br />

6 cycles. DLTs at higher doses were infection with neutropenia and grade 4<br />

thrombocytopenia. The efficacy was higher than generally reported with<br />

single agent D (Sleijfer, The Oncologist, 2005). At MTD (75 mg/m2 D and<br />

300 mg/m2 TH-302) best response were 2 (2%) CR, 30 (34%) PR, 43<br />

(48%) SD. Median PFS and OS were 6.7 and 17.5 months, respectively.<br />

Based upon these data, a study (NCT01440088) is ongoing to assess the<br />

benefit <strong>of</strong> adding TH-302 to the standard D as first-line therapy <strong>of</strong> STS.<br />

Methods: The randomized (1:1) phase III study <strong>of</strong> TH-302 (300 mg/m2 )<br />

with D versus D alone was initiated September 2011. Target goal is 450<br />

patients. TH-302 is administered IV over 30-60 minutes on Days 1 and 8<br />

(21-day cycle). D (75 mg/m2 , bolus or continuous) is administered Day 1<br />

starting 2-4 hrs after TH-302 when used in combination. Patients receiving<br />

TH-302 plus D without progression after 6 cycles may continue on TH-302<br />

alone. Key eligibility criteria: locally advanced unresectable or metastatic<br />

STS untreated with chemotherapy (adjuvant allowed), ECOG 0/1, measurable<br />

disease (RECIST 1.1) and adequate hematologic, hepatic, renal<br />

function. A FDA Special Protocol Agreement was reached on study design<br />

and analyses. Primary efficacy endpoint is overall survival (OS) comparison<br />

<strong>of</strong> the combination vs D. The study is designed to detect a 40%<br />

improvement in OS with 85% power and one-sided alpha <strong>of</strong> 2.5%. An<br />

interim futility PFS analysis is scheduled after 113 PFS events. Significant<br />

improvement in PFS (one-sided p � 0.10) is required to continue. A further<br />

analysis for early efficacy stoppage is scheduled after 175 OS events. IDMC<br />

will monitor safety and efficacy.<br />

TPS10102^ General Poster Session (Board #54G), Sun, 8:00 AM-12:00 PM<br />

Masitinib in comparison to imatinib as first-line therapy <strong>of</strong> patients with<br />

advanced gastrointestinal stromal tumor (GIST): A randomized phase III<br />

trial. Presenting Author: Antoine Adenis, Centre Oscar Lambret, Lille,<br />

France<br />

Background: Masitinib is an oral tyrosine kinase inhibitor (TKI) that has<br />

greater in vitro kinase activity and/or selectivity than imatinib against KIT<br />

and PDGFRA/B. A phase 2 study has previously reported that masitinib<br />

treatment produced clinically relevant activity in imatinib-naïve patients<br />

with advanced GIST. Considering the promising long term response<br />

observed in overall survival (OS) (see Table) there is compelling evidence to<br />

compare masitinib against imatinib (the current standard <strong>of</strong> care) in the<br />

first-line setting. Methods: A multicenter, randomized, open label, phase 3,<br />

1:1 study to compare efficacy and safety <strong>of</strong> masitinib (7.5 mg/kg/day) to the<br />

active control <strong>of</strong> imatinib (400 or 600 mg/day) in first-line treatment <strong>of</strong><br />

patients with advanced GIST. Primary endpoint is progression-free survival<br />

(PFS), with secondary endpoints including OS, time to progression (TTP),<br />

and clinical response rates (RECIST). Based on a planned sample size <strong>of</strong><br />

222 patients (111/arm) this 15% non-inferiority study was designed to<br />

have an 85% power using a 95% two-sided CI <strong>of</strong> the hazard ratio. Patient<br />

eligibility criteria include: histologically proven, metastatic or locally<br />

advanced nonresectable, or recurrent post-surgery GIST; TKI naïve patient<br />

or patient previously receiving imatinib only as a neoadjuvant or adjuvant<br />

therapy; and confirmed KIT-positive or PDGFRA-positive tumors. Recruitment<br />

is ongoing. On 09/2011, the DMC recommended continuation <strong>of</strong> this<br />

study. <strong>Clinical</strong>trial.gov: NCT00812240.<br />

Masitinib phase 2 a<br />

Imatinib phase 3 b<br />

{Meta-analysis data} c<br />

Dose 7.5 mg/kg/d 400 mg/d 800 mg/d<br />

Population, N 30 345 {818} 349 {822}<br />

Median follow-up (months) 65 54<br />

Median OS (months) [95%CI] Not reached [53; -] 55 {49} 51 {49}<br />

Month-12 97% 86% {90%} 86% {90%}<br />

24 90% 76% {80%} 72% {80%}<br />

36 87% 64% {60%} 62% {61%}<br />

48 76% 56% {56%} 56% {56%}<br />

60 62% 48% {45%} 49% {45%}<br />

Confirmed exon 11, n 9/30 (30%) 283/397 (71%) d<br />

Median OS(months) [95%CI] Not reached [65; -] 60<br />

Month-12 100% 93%<br />

24 89% 80%<br />

36 89% 70%<br />

48 89% 63%<br />

60 89% 51%<br />

a Blay, JCO 29: 2011 (suppl 4; abst 85); b Blanke, JCO 2008, 26:626; c GSTMA Group. JCO 2010, 28:1247; d<br />

Heinrich, JCO 2008, 26:5360.<br />

Sarcoma<br />

655s<br />

TPS10101 General Poster Session (Board #54F), Sun, 8:00 AM-12:00 PM<br />

Randomized multicenter phase III trial <strong>of</strong> trabectedin (T) versus doxorubicinbased<br />

chemotherapy as first-line therapy in patients with translocationrelated<br />

sarcoma (TRS). Presenting Author: Jean-Yves Blay, Centre Léon<br />

Bérard, Lyon, France<br />

Background: One third <strong>of</strong> sarcoma histotypes have specific chromosomal<br />

translocations which result in abnormal transcription factors, integral to the<br />

change to a malignant phenotype. T is the first <strong>of</strong> a new class <strong>of</strong> anti-tumor<br />

agents with a transcription-targeted mechanism <strong>of</strong> action. In vitro evidence<br />

exists <strong>of</strong> the ability <strong>of</strong> T to interfere with the aberrant transcription factor’s<br />

binding to DNA promoters in TRS. Patients with TRS, such as myxoid/round<br />

cell liposarcomas (MRCL) have benefited from long-lasting tumor control in<br />

response to T. Methods: A randomized, phase III study <strong>of</strong> T (1.5 mg/m2 in<br />

24-h intravenous infusion every 3 weeks [q3wk]) vs doxorubicin 75 mg/m2 q3wk, or doxorubicin 60 mg/m2 with ifosfamide 6-9 g/m2 q3wk) as first line<br />

treatment in patients with advanced TRS. Primary aim: to determine the<br />

efficacy <strong>of</strong> T vs doxorubicin-based therapy by comparing progression-free<br />

survival (PFS) in each arm. Secondary aims: comparison <strong>of</strong> PFS rates at six<br />

months, response rate (RR), PFS and RR by histological type (MRCL vs<br />

other subtypes), overall survival, safety, exploratory response evaluation by<br />

Choi criteria and pharmacogenomic analyses. Patients with confirmed TRS<br />

<strong>of</strong> the following subtypes: MRCL, alveolar s<strong>of</strong>t part sarcoma,angiomatoid<br />

fibrous histiocytoma, clear cell sarcoma, desmoplastic small round cell<br />

tumor, low grade endometrial stromal sarcoma, low grade fibromyxoid<br />

sarcoma,myxoid chondrosarcoma and synovial sarcoma, are stratified by<br />

performance status (PS 0 vs 1-2) and sarcoma subtype (MRCL vs other<br />

subtypes) and randomized (1:1 ratio) to T or doxorubicin � ifosfamide.<br />

Confirmation <strong>of</strong> the translocation by fluorescence in situ hybridization is<br />

compulsory for inclusion in the primary efficacy analysis. An adaptive<br />

design was chosen to test the reduction in relative risk <strong>of</strong> progression or<br />

death with T, an interim analysis will be conducted with 45 PFS events<br />

from 80 evaluable patients. To date 117 patients have been enrolled from<br />

six countries and 29 centers. An independent data monitoring committee<br />

met on 15 November 2011 and concluded that the trial should continue as<br />

planned.<br />

TPS10103 General Poster Session (Board #54H), Sun, 8:00 AM-12:00 PM<br />

Randomized multicenter double-blind phase II trial: The immunological<br />

adjuvant OPT-821 with or without a trivalent ganglioside vaccine in<br />

metastatic sarcoma patients following metastasectomy. Presenting Author:<br />

Richard D. Carvajal, Memorial Sloan-Kettering Cancer Center, New York,<br />

NY<br />

Background: The gangliosides GM2, GD2, and GD3 are strongly expressed<br />

in sarcoma and represent novel antitumor targets. We have demonstrated<br />

the safety and immunogenicity <strong>of</strong> individual monovalent ganglioside<br />

conjugate vaccines when administered with OPT821, a synthetic saponin<br />

adjuvant that augments the T-cell response. Antitumor effects are observed<br />

with anti-ganglioside antibodies in preclinical models, with greatest benefit<br />

seen in the micrometastatic setting. Patients (pts) with metastatic sarcoma<br />

rendered disease-free by surgery are at high risk for disease recurrence,<br />

with no therapy demonstrated to improve outcomes. Therefore, this<br />

population is ideally suited to test the efficacy <strong>of</strong> vaccine induced<br />

anti-ganglioside antibodies. We thus initiated this randomized doubleblind<br />

phase II study <strong>of</strong> OPT821 with or without a trivalent ganglioside<br />

vaccine composed <strong>of</strong> GM2, GD2 lactone and GD3 lactone conjugated to<br />

KLH, an immunogenic carrier protein. Methods: A total <strong>of</strong> 134 pts will be<br />

randomized on a 1:1 basis stratified by disease state (relapsed disease vs<br />

metastasis at time <strong>of</strong> diagnosis), disease-free interval and number <strong>of</strong><br />

relapses. Key inclusion criteria include stage IV sarcoma (locoregional<br />

recurrence is not sufficient) rendered disease-free following surgery or<br />

multi-modality therapy, no more than 8 weeks since surgery, and no<br />

continuous use <strong>of</strong> anti-inflammatory medications. Pts with Ewings sarcoma,<br />

GIST and rhabdomyosarcoma (except pleomorphic/anaplastic) are<br />

ineligible. Eligible pts will receive 10 subcutaneous injections in the<br />

outpatient setting over 84 weeks. The primary endpoint is progression-free<br />

survival (PFS). Secondary objectives include overall survival (OS) and 1and<br />

3-year PFS rate. Exploratory objectives include correlation <strong>of</strong> serologic<br />

response to outcome, comparison <strong>of</strong> tumor ganglioside expression at<br />

baseline and at recurrence, and analysis <strong>of</strong> circulating tumor cells.<br />

Enrollment began in July 2010. As <strong>of</strong> January 1, 2012, 83 patients had<br />

been recruited from 12 participating sites. <strong>Clinical</strong>trials.gov#:<br />

NCT01141491. Funded by Mabvax Therapeutics and NIH (R21CA13386).<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


656s Tumor Biology<br />

10500 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Gene expression-based predictors <strong>of</strong> chemotherapy response in basal-like<br />

breast cancer. Presenting Author: Aleix Prat, Lineberger Comprehensive<br />

Cancer Center, University <strong>of</strong> North Carolina at Chapel Hill, Chapel Hill, NC<br />

Background: The Basal-like subtype is generally associated with high<br />

chemo-sensitivity, but not all tumors respond and/or benefit to the same<br />

extend. In this study, we sought to identify gene expression predictors <strong>of</strong><br />

neoadjuvant chemotherapy sensitivity in Basal-like breast cancer. Methods:<br />

Expression <strong>of</strong> 542 genes was measured using the Nanostring nCounter<br />

platform from 69 FFPE pre-treated samples <strong>of</strong> the GEICAM/2006-03<br />

phase II trial, which were treated with epirrubicin/cyclophosphamide<br />

followed by docetaxel�/-carboplatin. Research-based PAM50 and Claudinlow<br />

predictors were also evaluated. The association between response<br />

(Miller-Payne criteria) and gene/signature expression was assessed by<br />

multivariable ordinal logistic regression. Significant findings were evaluated<br />

in 109 independent triple-negative and Basal-like tumors treated with<br />

anthracycline/taxane-based chemotherapy (Hatzis et al.). Finally, interaction<br />

tests were performed to identify genes/signatures associated with<br />

carboplatin response. Results: In GEICAM/2006-03, 61/69 (88%) tumors<br />

were identified as Basal-like by PAM50. High correlation to the Basal-like<br />

centroid, or high expression <strong>of</strong> proliferation-related genes (i.e. FANCA),<br />

were found to be significantly associated with high chemo-sensitivity,<br />

whereas high expression <strong>of</strong> genes associated with mesenchymal/stem cell<br />

biological processes (i.e. SNAI1 and IL6) and/or luminal differentiation<br />

(i.e. MUC1 and FOXA1) were significantly associated with chemoresistance;<br />

similar findings were observed in Hatzis et al. Finally, high<br />

expression <strong>of</strong> genes associated with proliferation/DNA-repair (i.e. ATR) and<br />

tight junctions (i.e. CLDN3/4/7) were found associated with carboplatin<br />

response, whereas expression <strong>of</strong> the Claudin-low signature was found<br />

associated with carboplatin resistance. Conclusions: High expression <strong>of</strong><br />

Basal-like and/or proliferation-related genes and low expression <strong>of</strong> luminal/<br />

mesenchymal/stem cell-like biological processes were consistently identified<br />

as predictive <strong>of</strong> chemotherapy response. Our data suggests that gene<br />

expression pr<strong>of</strong>iling might help shed light into the biological and clinical<br />

heterogeneity <strong>of</strong> Basal-like breast cancer.<br />

10502 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

A heuristic platform for clinical interpretation <strong>of</strong> cancer genome sequencing<br />

data. Presenting Author: Eliezer Mendel Van Allen, Dana-Farber Cancer<br />

Institute, Boston, MA<br />

Background: The ability to identify and effectively sort the full spectrum <strong>of</strong><br />

biologically and therapeutically relevant genetic alterations identified by<br />

massively parallel sequencing may improve cancer care. A major challenge<br />

involves rapid and rational categorization <strong>of</strong> data-intensive output, including<br />

somatic mutations, insertions/deletions, copy number alterations, and<br />

rearrangements into ranked categories for clinician review. Methods: A<br />

database <strong>of</strong> clinically actionable alterations was created, consisting <strong>of</strong> over<br />

100 annotated genes known to undergo somatic genomic alterations in<br />

cancer that may impact clinical decision-making. A heuristic algorithm was<br />

developed, which selectively identifies somatic alterations based on the<br />

clinically actionable alterations database. Remaining variants are sorted<br />

based on additional heuristics, including high priority alterations based on<br />

presence in the Cancer Gene Census, biologically significant cancer genes<br />

based on presence in COSMIC or MSigDB, and low priority alterations in the<br />

same gene family as biologically significant cancer genes. The heuristic<br />

algorithm was applied to whole exome sequencing data <strong>of</strong> clinical samples<br />

and whole genome sequencing data from a cohort <strong>of</strong> prostate cancer<br />

samples processed using established Broad Institute pipelines. Results:<br />

Application <strong>of</strong> the heuristic algorithm to the prostate cancer whole genome<br />

rearrangement data identified 172 (out <strong>of</strong> 5978) rearrangements involving<br />

actionable genes (averaging 2-3 events per tumor). Furthermore, two<br />

clinical samples processed prospectively were analyzed, yielding three<br />

potentially actionable alterations for clinical review. Conclusions: The<br />

heuristic model for clinical interpretation <strong>of</strong> next generation sequencing<br />

data may facilitate rapid analysis <strong>of</strong> tumor genomic information for<br />

clinician review by identifying and prioritizing alterations that can directly<br />

impact care. Our platform can also be applied to research data to<br />

prospectively explore clinically relevant findings from existing cohorts.<br />

Future analytical approaches using heuristic or probabilistic algorithms<br />

should underpin a robust prospective assessment <strong>of</strong> clinical cancer<br />

genome data.<br />

10501 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Estrogen receptor alpha (ESR1) gene amplification status and clinical<br />

outcome in tamoxifen-treated postmenopausal patients with endocrineresponsive<br />

early breast cancer: An analysis <strong>of</strong> the prospective ABCSG-6<br />

trial. Presenting Author: Christian F. Singer, Medical University <strong>of</strong> Vienna,<br />

Department <strong>of</strong> OB/GYN, Vienna, Austria<br />

Background: Estrogen receptor alpha (ER�) expression is a prognostic<br />

parameter in breast cancer and predicts response to endocrine therapy.<br />

One <strong>of</strong> the factors important for protein expression is amplification <strong>of</strong> its<br />

encoding gene ESR1. We have investigated the value <strong>of</strong> ESR1 amplification<br />

in predicting the long-term clinical outcome in tamoxifen-treated<br />

postmenopausal women with endocrine-responsive breast cancer. Methods:<br />

394 patients who had been randomized into the tamoxifen-only arm <strong>of</strong> the<br />

prospectively designed endocrine ABCSG-06 trial and in whom FFPE tumor<br />

tissue was available were included in this analysis. Immunohistochemical<br />

ER� expression was evaluated both locally and centrally using the Allred<br />

score, while ESR1 gene amplification status was evaluated by FISH<br />

analysis using the ESR1/CEN6 ratio. Results: ESR1 copy number gains<br />

were detected in 187 <strong>of</strong> 394 (47%) tumor specimen and was associated<br />

with favorable clinical outcome. At a median follow-up <strong>of</strong> 10 years, women<br />

with intratumoral ESR1 copy number gains had a significantly longer<br />

distant recurrence-free survival (adjusted HR for relapse 0.48; 95% CI<br />

0.28-0.83; p�0.009) and breast cancer-specific survival (adjusted HR for<br />

death 0.46; CI 0.46-0.71; p�0.006) when compared to women with<br />

normal ESR1 copy numbers. Immunohistochemical ER� protein expression,<br />

evaluated by Allred score, was significantly correlated with ESR1 copy<br />

number alterations (p�0.0001; Chi-Square test), but did itself not allow to<br />

discriminate between patients with poor and good prognosis. Conclusions:<br />

ESR1 amplification status is an independent and powerful predictor for<br />

long-term distant recurrence-free and breast cancer-specific survival in<br />

postmenopausal women with endocrine-responsive early-stage breast cancer<br />

who received 5 years <strong>of</strong> tamoxifen.<br />

10503 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Circulating tumor cell counts (CTC) as prognostic <strong>of</strong> overall survival (OS) in<br />

SWOG S0421-docetaxel with or without atrasentan for metastatic castration<br />

resistant prostate cancer (mCRPC). Presenting Author: Nicholas J.<br />

Vogelzang, Comprehensive Cancer Centers <strong>of</strong> Nevada and US Oncology<br />

Research, Las Vegas, NV<br />

Background: CTC are promising biomarkers in mCRPC but have not been<br />

prospectively validated for docetaxel treatment (Rx). Using CellSearch<br />

technology (J&J), we enumerated CTC in this Phase 3 trial & assessed<br />

prognostic value for OS. The aim <strong>of</strong> this correlative study nested within<br />

0421 was to compare CTC via CellSearch technology to newer micr<strong>of</strong>ilter<br />

technologies (Cote & Goldkorn PIs RO1 CA141077). Comparative data<br />

analysis is ongoing. Methods: CTC were drawn at baseline (d1) & pre-cycle 2<br />

(d21) <strong>of</strong> Rx & shipped overnight to a central site for enumeration (CTC/7.5<br />

ml). Cox regression evaluated the association between OS and (i) baseline<br />

CTC counts & (ii) CTC dynamics (d1 to d21) in pts with good (�5) vs. poor<br />

(��5) baseline CTC counts. Receiver operator characteristic (ROC) analysis<br />

and Characteristics & Regression Trees (CART) were used to explore<br />

further prognostic CTC cutpoints for 2-yr survival. Results: Of 263 patients<br />

(pts) consented, 238 were evaluable at d1 & 232 at d21. At d1 median<br />

CTC was 5 (range 0-5916) & d1 CTC � vs. �� 5 was associated with<br />

baseline PSA (mean 99 vs. 320 ng/ml, p�0.004) and worse bone pain<br />

(36% vs. 51%, p�0.03). There was a significant difference in OS for d1<br />

CTC � vs. ��5, with a hazard ratio (HR) <strong>of</strong> 2.92 (95% CI 1.92-4.43,<br />

p�0.001) after adjustment for PSA & other factors. In pts with low d1 CTC<br />

(� 5), an increase in CTC was associated with shorter OS, HR 4.04 (95%CI<br />

1.56-10.44, p�0.004); in pts with high d1 CTC (��5), a ��2-fold<br />

decrease in CTC was associated with longer OS, HR 0.45 (95%CI<br />

0.24-0.84, p�0.012); adjusting for risk factors. D1 CTC and 2-year<br />

survival had ROC AUC <strong>of</strong> 0.781. CART analysis identified prognostic<br />

subgroups based on CTC <strong>of</strong> 0, 1-5, 6-53, and �53: (HR 0.36, 0.77,1.3<br />

and 2.8). Conclusions: In this phase 3 trial, d1 CTC was prognostic <strong>of</strong> OS<br />

after risk factor adjustment. CTC dynamics from d1 to d21 were also<br />

prognostic <strong>of</strong> OS. These data are an exploratory subset analysis <strong>of</strong> the<br />

overall study. Yet, they comprise the largest docetaxel-based prospective<br />

cohort to date, which validates a 5 CTC prognostic threshold for OS &<br />

identifies new potential prognostic subgroups that may extend the clinical<br />

utility <strong>of</strong> CTC enumeration in mCRPC.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


10504 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Gene expression pr<strong>of</strong>iles <strong>of</strong> circulating tumor cells in metastatic breast<br />

cancer patients. Presenting Author: Bianca Mostert, Department <strong>of</strong> Medical<br />

Oncology, Erasmus University Medical Center, Daniel den Hoed Cancer<br />

Center, Rotterdam, Netherlands<br />

Background: A circulating tumor cell (CTC) count is an established<br />

prognostic factor in metastatic breast cancer. Besides enumeration, CTC<br />

characterization promises to further improve outcome prediction and<br />

treatment guidance. We have previously shown the feasibility <strong>of</strong> measuring<br />

the expression <strong>of</strong> a panel <strong>of</strong> 96 clinically relevant genes in CTCs in a<br />

leukocyte background, and in the current study, we determined the<br />

prognostic value <strong>of</strong> CTC gene expression pr<strong>of</strong>iling in metastatic breast<br />

cancer. Methods: CTCs were isolated and enumerated from blood <strong>of</strong> 130<br />

metastatic breast cancer patients prior to start <strong>of</strong> first-line systemic,<br />

endocrine or chemotherapeutic, therapy. Of these, 103 were evaluable for<br />

mRNA gene expression levels measured by quantitative RT-PCR in relation<br />

to time to treatment switch (TTS). Separate prognostic CTC gene pr<strong>of</strong>iles<br />

were generated by leave-one-out cross validation for all patients and for<br />

patients with �5 CTCs per 7.5 mL blood, and cut-<strong>of</strong>fs were chosen to<br />

ensure optimal prediction <strong>of</strong> patients who might benefit from an early<br />

therapy switch. Results: In the total cohort, <strong>of</strong> whom 56% received<br />

chemotherapeutic and 44% endocrine therapy, baseline CTC count (�5<br />

versus �5 CTCs/7.5 mL blood) predicted for TTS (Hazard Ratio (HR) 2.92<br />

[95% Confidence Interval (CI) 1.71 – 4.95] P �0.0001). A 16-gene CTC<br />

pr<strong>of</strong>ile for all patients and a separate 9-gene CTC pr<strong>of</strong>ile applicable for<br />

patients with �5 CTCs were identified, which distinguished those patients<br />

with TTS or death within 9 months from those with a more favorable<br />

outcome. Test performance for both pr<strong>of</strong>iles was favorable; the 16-gene<br />

pr<strong>of</strong>ile had 90% sensitivity, 38% specificity, 50% positive predictive value<br />

(PPV) and 85% negative predictive value (NPV), and the 9-gene pr<strong>of</strong>ile<br />

performed slightly better at 92% sensitivity, 52% specificity, 66% PPV and<br />

87% NPV. In multivariate Cox regression analysis, the 16-gene pr<strong>of</strong>ile was<br />

the only factor independently associated with TTS (HR 3.15 [95%CI 1.35<br />

– 7.33] P 0.008). Conclusions: Two CTC gene expression pr<strong>of</strong>iles were<br />

discovered, which provide prognostic value in metastatic breast cancer<br />

patients. This study further underscores the potential <strong>of</strong> molecular characterization<br />

<strong>of</strong> CTCs.<br />

10506 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

DEAR1, a novel tumor suppressor, negative regulator <strong>of</strong> epithelialmesenchymal<br />

transition, and prognostic factor in non-small cell lung<br />

cancer. Presenting Author: Alfonso Quintas-Cardama, University <strong>of</strong> Texas<br />

M. D. Anderson Cancer Center, Houston, TX<br />

Background: DEAR1, also known as TRIM62, maps to 1p.35, a genomic<br />

interval within which loss <strong>of</strong> heterozygosity occurs at high frequency in<br />

carcinomas. DEAR1 has been proposed as a regulator <strong>of</strong> cell polarity in<br />

vitro. Methods: We genetically engineered mice lacking DEAR1 alleles at<br />

murine chromosome 4. To study the impact <strong>of</strong> DEAR1 loss in lung cancer,<br />

DEAR1 deficient mice were crossed to mice carrying the latent mutant<br />

K-ras G12D allele (K-ras LA1 ). Results: No survival difference was observed<br />

between DEAR1 �/� and DEAR1 �/� mice (729 vs 699 days, p�0.98) but<br />

they were markedly shorter than that <strong>of</strong> DEAR1 wild-type littermates (803<br />

days, p�0.01). Over 90% <strong>of</strong> mice developed tumors, mainly adenocarcinomas,<br />

including invasive and/or metastatic lung cancer in 23% <strong>of</strong> cases.<br />

DEAR1-deficient K-ras LA1 mice had reduced life span compared with<br />

DEAR1 �/� :K-ras �/LA1 littermates (184 vs 291 days, p�0.0001). The<br />

survival <strong>of</strong> DEAR1 �/� :K-ras �/LA1 and DEAR1 �/� :K-ras �/LA1 mice were<br />

similar (180 vs 153 days, p�0.38) and qPCR and IHC analyses suggested<br />

loss <strong>of</strong> the wild-type DEAR1 allele. The survival <strong>of</strong> DEAR1-deficient<br />

K-ras LA1<br />

mice was also shorter than that <strong>of</strong> control p53�/� :K-ras�/LA1 mice<br />

(240 days; p�0.01). The metastasis incompetent K-ras LA1 –positive LKR13<br />

cell line after shRNA DEAR1 knock-down and cell lines derived from<br />

DEAR1-deficient K-ras LA1 tumors recapitulated an invasive phenotype in<br />

transwell migration and 3D culture assays and a metastatic phenotype on<br />

SC or IV injection into immunocompetent wild-type littermates. The latter<br />

was associated with epithelial-mesenchymal transition involving loss <strong>of</strong><br />

E-cadherin and DEAR1 expression and upregulation <strong>of</strong> vimentin, Twist,<br />

and CD44. DEAR1 downregulation was very prevalent in a non-small cell<br />

lung cancer (NSCLC) tissue array with 214 samples. Patients with early<br />

stage NSCLC and loss <strong>of</strong> DEAR1 expression had a markedly shorter time to<br />

relapse compared to those retaining DEAR1 expression (5.1 vs 2.87 years,<br />

p�0.049) and worse 5-year relapse-free and overall survival rates.<br />

Conclusions: DEAR1 is a novel tumor suppressor that negatively regulates<br />

EMT and promotes lung cancer metastasis. DEAR1 loss is a poor prognostic<br />

factor in NSCLC.<br />

Tumor Biology<br />

657s<br />

10505 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Circulating DNA analysis and concordance with tumor section analysis in<br />

the detection <strong>of</strong> KRAS and BRAF point mutations from metastatic<br />

colorectal cancer. Presenting Author: Alain R. Thierry, Sysdiag UMR3145 -<br />

CNRS, Montpellier, France<br />

Background: We developed a specific method for circulating cell-free DNA<br />

(ctDNA) enabling the detection <strong>of</strong> point mutations. CtDNA exists at high<br />

level in patients with different types <strong>of</strong> cancer, and presents great potential<br />

in regards, in particular, to its low invasiveness, rapid data turnaround and<br />

cost effectiveness. We described here the first blinded prospective study on<br />

the comparison <strong>of</strong> KRAS and BRAF mutational status data obtained from<br />

the analysis <strong>of</strong> ctDNA and tumor section. Methods: Intplex is a refined<br />

Q-PCR-based method specifically designed to analyze ctDNA. IntPlex<br />

allows not only the detection <strong>of</strong> point mutations, but also the determination<br />

<strong>of</strong> tumor-ctDNA concentration and fragmentation index simultaneously.<br />

Intplex sensitivity is 0.01% (mutant to WT ratio) and is unprecedented<br />

among Q-PCR-based methods. The study was conducted from a multicenter<br />

cohort <strong>of</strong> 79 metastatic CRC patients not having received chemo- or<br />

radio-therapy within the month prior to blood sample collection. Results:<br />

Mutational status could not be determined in 9 samples with one <strong>of</strong> the two<br />

methods. CtDNA analysis showed 100% specificity and 87% sensitivity for<br />

KRAS detection (only three samples are misclassified) as compared to<br />

tumor section analysis with 23/70 (33%) positive samples, the concordance<br />

value being 96%. For BRAF mutation, the method exhibited a<br />

specificity and a sensibility <strong>of</strong> 100% with 5/70 (7%) positive samples<br />

(concordance <strong>of</strong> 100%). When combining KRAS and BRAF mutations,<br />

100% specificity, 89% sensibility and 98% concordance were determined.<br />

The three discordant samples may be explained by the malignant genotype<br />

<strong>of</strong> primary tumor versus metastasis, or by the sensitivity <strong>of</strong> our detection<br />

method. The mutation load (median, 12.4%) expressed as the proportion<br />

<strong>of</strong> mutant allele in ctDNA is highly variable (0.037% to 69%) among<br />

mutated samples showing a very high inter-individual heterogeneity.<br />

Conclusions: Our blinded prospective multicenter study clearly showed for<br />

the first time that tumor section analysis might be advantageously replaced<br />

by ctDNA analysis, enlarging personalized medicine power for cancer<br />

patients.<br />

10507 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Rare epidermal growth factor receptor (EGFR) mutations in 10,117<br />

patients with non-small cell lung cancer (NSCLC) evaluated by the French<br />

ERMETIC IFCT network: <strong>Clinical</strong>, molecular, and survival data. Presenting<br />

Author: Michele Beau-Faller, CHU Strasbourg, Strasbourg, France<br />

Background: The EGFR exon 19 deletion and exon 21 L858R mutation are<br />

associated with response to EGFR-TKI. However, the correlation with<br />

response to EGFR-TKI remains unclear for rare exon 18 and 20 EGFR<br />

mutations. We investigated the clinical features <strong>of</strong> NSCLC patients with<br />

exon 18 and exon 20 EGFR mutations, analyzed response to EGFR-TKI and<br />

patient’s survival. Methods: Between 2005 and 2011, all rare exon 18 and<br />

20 EGFR mutations were collected from 10117 cases <strong>of</strong> NSCLC in 15 <strong>of</strong><br />

28 French NCI molecular genetic laboratories. Results: During the study<br />

period, EGFR mutations were identified in 1048 (10.35%) out <strong>of</strong> the<br />

10117 performed tests and 108 rare mutations (10.3%) were identified in<br />

103 patients, including 25 never identified mutations. Among them, 48<br />

mutations were localized at exon 18 and 60 at exon 20, with 3 out <strong>of</strong> the 4<br />

identified T790M exon 20 mutations associated with L858R mutation.<br />

Only 5 other rare mutations were associated with EGFR 19 or 21 mutations.<br />

38 patients received EGFR-TKI (erlotinib, n�32; gefitinib, n�6). Among<br />

the 36 evaluable patients, best response on TKI was progression in 18<br />

patients (50%), stabilization in 11 (30.5%) and partial response in 7<br />

(19.4%). Tumor control rate observed in exon 18 mutations was 61%<br />

(8/13) and 50% (13/26) in exon 20 mutations. For the EGFR-TKI treated<br />

patients, the median PFS was 6 months (2-53). Median OS was 15 months<br />

(1-92), with no difference between the EGFR-TKI treated patients or not.<br />

Conclusions: Prevalence <strong>of</strong> rare EGFR mutations was 1.06% <strong>of</strong> all tested<br />

NSCLC and 10.35% <strong>of</strong> all EGFR mutations. Tumor control on TKI was<br />

observed in half <strong>of</strong> the patients. Reports <strong>of</strong> NSCLC harboring rare mutations<br />

are important to help the decision-making process in this subset <strong>of</strong><br />

patients.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


658s Tumor Biology<br />

10508 Poster Discussion Session (Board #1), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Next-generation RNA sequencing reveals transcriptomic changes after<br />

brief exposure to preoperative nab-paclitaxel, bevacizumab, and trastuzumab.<br />

Presenting Author: Vinay Varadan, Philips Research North America,<br />

Briarcliff Manor, NY<br />

Background: Identification <strong>of</strong> differentially expressed transcripts after brief<br />

exposure to preoperative therapy can help determine likely response<br />

markers. We quantify and compare differential gene and is<strong>of</strong>orm expression<br />

using RNA-seq on patient samples with 10 day exposure to one dose <strong>of</strong><br />

trastuzumab, bevacizumab or nab-paclitaxel. Methods: We sequenced<br />

transcriptomes <strong>of</strong> 23 pairs <strong>of</strong> core biopsy RNA from breast cancers pre/post<br />

10 day exposure to therapy. Paired-end sequencing was done on the<br />

Illumina GAII platform using amplified total RNA with 74bp read length,<br />

yielding data on transcript abundance for a total <strong>of</strong> 22,160 genes and<br />

34,449 transcripts. Differential expression <strong>of</strong> transcripts between pre/post<br />

samples was estimated assuming Poisson-distributed read-counts, followed<br />

by multiple testing correction and enrichment analysis <strong>of</strong> 185 KEGG<br />

pathways. Results: PAM50-based clustering showed individual samples<br />

cluster together, demonstrating that tumor subtypes do not change over the<br />

10-day treatment (SABCS 2011). We identified genes that were significantly<br />

differentially expressed (p�0.05; FDR�0.1) in at least 60% <strong>of</strong><br />

samples within each therapy arm: 780 genes in trastuzumab, 302 in<br />

bevacizumab, and 176 in nab-paclitaxel. Surprisingly, only THAP11 and<br />

TINF2 were common amongst them. THAP11 is involved in stem cell<br />

maintenance and TINF2 is important for regulation <strong>of</strong> telomere length.<br />

Immune system and metabolism-related pathways were commonly affected<br />

(p�0.05) across all arms. The bevacizumab arm showed significant<br />

down-regulation <strong>of</strong> angiogenesis-associated genes: ESM1 and VEGFR2 in<br />

� 80% <strong>of</strong> samples. The nab-paclitaxel arm exhibited changes in TGF-beta<br />

signaling, Nod-like receptor and Wnt signaling. The trastuzumab arm<br />

exhibited consistent alteration <strong>of</strong> ErbB2 and mTOR pathways, with SOX11<br />

and TOP2B downregulated in every sample. Conclusions: This is the first<br />

study to compare gene expression with brief exposure across therapies<br />

using RNA-seq technology. The unique aspects <strong>of</strong> transcriptional response<br />

to each treatment underscore the need for specific markers <strong>of</strong> therapeutic<br />

response to nab-paclitaxel, bevacizumab and trastuzumab.<br />

10510 Poster Discussion Session (Board #3), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

PTEN assessment and PI3K/mTOR inhibitors: Importance <strong>of</strong> simultaneous<br />

assessment <strong>of</strong> MAPK pathway aberrations. Presenting Author: Filip Janku,<br />

Department <strong>of</strong> Investigational Cancer Therapeutics (Phase I Program),<br />

University <strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: Loss <strong>of</strong> PTEN function increases PI3K/mTOR signaling.<br />

Preclinical data suggest that PTEN aberrations can predict sensitivity to<br />

drugs targeting the PI3K/mTOR pathway. Methods: Tumors from 461<br />

patients with diverse cancers referred to the <strong>Clinical</strong> Center for Targeted<br />

Therapy were tested for cytoplasmic immunohistochemical expression <strong>of</strong><br />

PTEN using the following scoring system: negative (no staining); positive<br />

(intact staining); reduced staining, with stroma serving as an internal<br />

positive control. Whenever possible, tumors were also tested for mutations<br />

in the MAPK pathway (KRAS, NRAS, and BRAF). We analyzed outcomes <strong>of</strong><br />

patients treated with PI3K /mTOR with respect to their PTEN status.<br />

Results: Of 461 patients, 47 (10%) had negative PTEN expression, 168<br />

(36.5%) had positive PTEN expression, and 246 (53.5%) had reduced<br />

PTEN. In tumor types with �10 patients tested, negative PTEN was most<br />

frequent in uterine (10/33, 33%), renal (3/11, 27%), salivary gland (2/10,<br />

20%), colorectal (15/82, 18%), breast (2/15, 13%), pancreatobiliary<br />

(3/24, 13%), and prostate cancers (2/19, 11%). Of 206 patients tested for<br />

MAPK (KRAS, NRAS, and BRAF) pathway mutations, patients with either<br />

negative or reduced PTEN had more MAPK mutations than patients with<br />

positive PTEN expression (72/144, 50% vs. 20/62, 32%, p�0.02). A total<br />

<strong>of</strong> 153 (33%) patients received therapies with PI3K /mTOR inhibitors and<br />

17 (11%, 95% CI 0.07-0.17) achieved a partial response (PR). Proportions<br />

<strong>of</strong> PRs were 2/13 (15%) for PTEN negative, 3/50 (6%) for PTEN<br />

positive, and 12/90 (13%) for reduced PTEN. There was no significant<br />

difference in PR rate amongst patients with positive PTEN expression<br />

compared to patients with negative or reduced PTEN (3/50, 6% vs.<br />

14/103, 14%; p�0.27). Conclusions: Complete loss <strong>of</strong> PTEN expression<br />

was found in 10% <strong>of</strong> patients. Positive PTEN expression was found in<br />

36.5% <strong>of</strong> patients. PTEN status did not predict response to PI3K/mTOR<br />

inhibitors, perhaps because patients with negative and reduced PTEN<br />

expression had a higher incidence <strong>of</strong> simultaneous MAPK (KRAS, NRAS,<br />

BRAF) mutations.<br />

10509 Poster Discussion Session (Board #2), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Early change in 18-fluorodeoxyglucose (FDG) uptake on positron emission<br />

tomography (PET) to predict response to preoperative systemic therapy<br />

(PST) in HER2-negative primary operable breast cancer: Translational<br />

breast cancer research consortium (TBCRC008). Presenting Author: Roisin<br />

M. Connolly, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins<br />

School <strong>of</strong> Medicine, Baltimore, MD<br />

Background: PST allows for improved surgical outcomes and response<br />

assessment without compromising long term outcomes. PST is an attractive<br />

model for assessing surrogate markers <strong>of</strong> response to therapy. We<br />

hypothesized that changes in tumor standardized uptake values corrected<br />

for lean body mass (SUL) max on FDG- PET by cycle 1 day 15 (C1D15) <strong>of</strong><br />

therapy would predict pathological complete response (pCR) to PST in<br />

women with stage 2-3, grade 2-3, HER2-negative breast cancer. Methods:<br />

TBCRC008 is a multicenter placebo-controlled trial that investigates pCR<br />

following 12 weeks <strong>of</strong> preoperative carboplatin and albumin-bound paclitaxel<br />

with or without vorinostat. FDG-PET followed by tumor biopsies were<br />

performed at baseline and C1D15. We correlated % reduction in SULmax<br />

on FDG-PET (PERCIST 1.0; Wahl RL, J Nuc Med 2009) with pCR (no<br />

invasive cancer in breast/axilla). We compared % reduction in SULmax<br />

between responders (pCR) and non responders (no pCR) using nonparametric<br />

Wilcoxon rank sum test. We explored association <strong>of</strong> % reduction in<br />

SULmax at pre-specified cut<strong>of</strong>f with response using Fisher’s exact test and<br />

logistic regression. We correlated baseline, C1D15, and % change in Ki67<br />

at C1D15 with pCR. Results: Accrual is complete. Of 62 women enrolled<br />

(10/2009-11/2011), 40 have completed study PST and surgery (median<br />

age 47.5 [range 30-68], ER-positive 67%). Overall pCR was 26%. In an<br />

intent to treat analysis (n�39), we observed a significant difference in<br />

median % reduction in SULmax between responders vs not (66.6% vs<br />

32.4%, p �0.001). We observed a higher proportion <strong>of</strong> reduction in<br />

SULmax � 60% in responders vs not (80% vs 3.5%, p �0.001). The<br />

differences in baseline, C1D15 and % change in Ki67 were not significant<br />

between responders and non-responders. Conclusions: Change in SULmax<br />

on FDG-PET 15 days after initiating PST was significantly greater in<br />

patients with pCR versus no pCR. Future studies will determine whether<br />

altering therapy based on early changes in SULmax will improve pCR.<br />

Unblinded data from all participants will be presented at the meeting.<br />

10511 Poster Discussion Session (Board #4), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Analysis <strong>of</strong> the intratumoral heterogeneity <strong>of</strong> PIK3CA mutant alleles in<br />

breast cancer (BC): Implications for the luminal (LUM) phenotype. Presenting<br />

Author: Leticia De Mattos-Arruda, Medical Oncology Department, Vall<br />

d’Hebron University Hospital, Barcelona, Spain<br />

Background: The hyperactivation <strong>of</strong> the phosphatidylinositol 3-kinase<br />

(PI3K) pathway may confer endocrine therapy resistance and is an<br />

attractive target for LUM patients (pts). However, PI3KCA mutations could<br />

be heterogeneously distributed within the tumor as different genetic clones<br />

and this could have therapeutic implications. Our aim was to assess the<br />

frequency <strong>of</strong> PIK3CA mutant alleles in different BC phenotypes. Methods:<br />

DNA was obtained from 75 consecutive BC FFPE samples and was pr<strong>of</strong>iled<br />

with the OncoCarta Panel v1.0 (Sequenom). Frequencies <strong>of</strong> mutant alleles<br />

(% mutant allele, mA) <strong>of</strong> PIK3CA mutations were extracted from the<br />

MassARRAY spectrum data. The viable tumor area (TA) was scored by H&E.<br />

Pts were stratified by BC phenotypes: ER�/HER2- (LUM); HER2� (HER2);<br />

triple negative (TN). Results: 25.3% <strong>of</strong> pts had PIK3CA mutations. The<br />

mean PIK3CA mA (p�0.04) and mean TA (p�0.07) differed among<br />

phenotypes. LUM tumors demonstrated greater frequencies <strong>of</strong> PIK3CA<br />

mutation (38% vs.15%, p�0.05) and mean PIK3CA mA (31% vs.17%,<br />

p�0.01) than non-LUM.There was a significant linear correlation between<br />

mA and TAfor LUM tumors (r�0.91); when HER2 and TN tumors were also<br />

considered this relation was less pronounced (r�0.66). Overall, LUM pts,<br />

median age 55 (42-84), had significantly better clinical outcomes<br />

(p�0.00024), whilst analyses <strong>of</strong> prognosis did not differ among LUM pts<br />

(PIK3CA mutant vs. wild-type, p�0.68) nor mutant pts (LUM vs. non-<br />

LUM, p�0.36). Outcomes will be presented. To assess the intratumoral<br />

heterogeneity <strong>of</strong> PIK3CA mutations, we determined the mA/TA ratio. The<br />

mA/TA ratio should be 0.5 for a homogeneous distribution <strong>of</strong> the heterozygous<br />

PIK3CA mutation. The ratio <strong>of</strong> LUM patients was close to 0.5, while<br />

for non-LUM it was lower, suggesting a non-homogeneous distribution <strong>of</strong><br />

PIK3CA mutant alleles in non-LUM tumors. Conclusions: Our analysis<br />

indicates that LUM tumors tend to be homogeneous regarding PIK3CA<br />

mutation as compared to HER2 and TN. This suggests that PIK3CA<br />

oncogenic activation could be an early hit for tumor initiation in LUM<br />

tumors, which could be more specifically targetable; thus, pts would derive<br />

greater benefit from PI3K-inhibitors plus hormonal therapy.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


10512 Poster Discussion Session (Board #5), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Dynamic contrast-enhanced MRI versus 18F-misonidazol-PET/CT to predict<br />

pathologic response in bevacizumab-based neoadjuvant therapy in<br />

breast cancer. Presenting Author: Jesus Garcia-Foncillas, Oncology Department,<br />

Hospital Universitario Fundación Jiménez Díaz, Universidad Autónoma<br />

de Madrid, Madrid, Spain<br />

Background: To investigate the role <strong>of</strong> DCE-MRI versus 18F-Misonidazole<br />

(FMISO) positron emission tomography (PET/CT) in the prediction <strong>of</strong><br />

pathological response to bevacizumab-based neodajuvant therapy. Methods:<br />

73 chemotherapy naïve, stage II and III breast cancer (BC) patients (pts)<br />

were enrolled in a phase II, single-arm, multicenter, open-label and<br />

prospective clinical trial. Pts received single infusion <strong>of</strong> bevacizumab (15<br />

mg/ kg) (C1) 3 weeks prior to the beginning <strong>of</strong> neoadjuvant chemotherapy<br />

(NAC) consisting <strong>of</strong> 4 cycles <strong>of</strong> docetaxel (60 mg/mq), doxorubicin (50<br />

mg/mq) and bevacizumab (15 mg/ kg) every 21 days (C2-C5), followed by<br />

surgery. Tumor proliferation, hypoxia and perfusion were evaluated respectively<br />

using 18F-Fluorothymidine (FLT) and 18F-Misonidazole (FMISO)<br />

positron emission tomography (PET/CT) and dynamic contrast enhancement<br />

magnetic resonance (DCE-MR). Serial imaging studies were performed<br />

in parallel at several time points including baseline (BL) and 14-21<br />

days after bevacizumab alone (C1). Results: After only one administration <strong>of</strong><br />

bev, tumor proliferation and perfusion assessed using FLT-PET and<br />

DCE-MRI significantly decrease (-26% and -46%, p�0.001) but these<br />

changes were not found to be associated with final response. Most<br />

important, changes in tumor hypoxia induced by bevacizumab was significantly<br />

associated with pathological response (p� 0.004) and was an<br />

independent predictor <strong>of</strong> response in multivariate analysis (RR�0.95, IC<br />

95% 0.92-0.99, p�0.02). Decrease in FMISO uptake �10% yielded a<br />

ROC curve area <strong>of</strong> 0.7 (95% CI: 0.56 - 0.85) with high specificity (94%).<br />

Conclusions: Our findings suggest a significant value <strong>of</strong> early changes in<br />

tumor hypoxia assessed by FMISO-PET as a biomarker <strong>of</strong> pathological<br />

response in bevacizumab-based neoadjuvant therapy in breast cancer.<br />

10514 Poster Discussion Session (Board #7), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

A translational study to investigate the association between smoking-induced<br />

lung inflammation and lung metastases (LM) from breast cancer (BC). Presenting<br />

Author: Patrick Glyn Morris, Memorial Sloan-Kettering Cancer Center, New<br />

York, NY<br />

Background: Retrospective and preclinical studies suggest that smokers are at<br />

increased risk for LM from BC, possibly mediated by lung inflammation and the<br />

bioactive lipid, PGE2. To investigate this link in pts, we examined urinary<br />

PGE-M, a stable end metabolite <strong>of</strong> PGE2. Methods: A translational study was<br />

conducted, consisting <strong>of</strong> pts with LM (n�100), pts with mets but no known lung<br />

mets (NKL, n�100) and controls (CTRLS) with a history <strong>of</strong> BC (n�200). Pts<br />

receiving steroids and radiotherapy were excluded. Pts gave urine (PGE-M),<br />

blood (biomarkers), had BMI measured, and completed a validated questionnaire<br />

(smoking, NSAIDs, confounders). PGE-M was measured by mass spectrometry,<br />

normalized to urinary creatinine and compared between grps in both<br />

univariate and multivariate models, correcting for covariates. Results: From<br />

09/2010 to 06/2011, 400 pts, med age 58 yrs (range 24-88) enrolled. There<br />

were no significant differences between grps in smoking and NSAID exposure<br />

(table). PGE-M (med; range) was highest among pts with LM (6.7; 0.7 - 43.4)<br />

compared to pts with NKL (4.6; 0.7 - 26.8) and CTRLS (4.2; 0.9 – 62.6,<br />

P�0.001). In univariate analysis, smoking pack year was associated with PGE-M<br />

(��0.13, P�0.01), and ever smokers with LM had the highest PGE-M (med 7.4;<br />

range 0.8-28.9). In a multivariable model ever smokers with LM had higher<br />

PGE-M than never smoking CTRLS (P�0.03). Pts with �24 pack year smoking<br />

history and LM had the highest co-variate-adjusted log PGE-M (P�0.03). In this<br />

model, age (P�0.001), pack year smoking hx (P�0.02), receipt <strong>of</strong> chemoRx �1<br />

mth (P�0.003) and BMI (P�0.002) were all associated with higher PGE-M.<br />

Conversely, receipt <strong>of</strong> NSAIDs �7 days was associated with lower PGE-M<br />

(P�0.001). Conclusions: Increased lung inflammation, as characterized by<br />

PGE-M, occurs in ever smokers with LM from BC, supporting the hypothesis that<br />

changes in the lung microenvironment (“soil”) predispose to metastasis.<br />

Baseline characteristics<br />

LM<br />

N�100<br />

NKL<br />

N�100<br />

CTRLS<br />

N�200 P<br />

N(%) N(%) N(%)<br />

Ever smokers 44 (44) 48 (48) 104 (52) 0.3<br />

Pack year smoking Hx, med (range) 24 (1-126) 20 (1-150) 20 (1-150) 0.5<br />

NSAIDs < 7 days 48 (48) 45(45) 92 (46) 0.9<br />

ChemoRx


660s Tumor Biology<br />

10516 Poster Discussion Session (Board #9), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Mutational analysis <strong>of</strong> circulating tumor cells in breast cancer patients by<br />

targeted clonal sequencing. Presenting Author: Xunhai Xu, New York<br />

Presbyterian-Columbia, New York, NY<br />

Background: The molecular signature <strong>of</strong> circulating tumor cells (CTCs) may<br />

serve as a surrogate marker for accurate description <strong>of</strong> the metastatic tumor<br />

<strong>of</strong> interest, especially in the setting <strong>of</strong> treatment response and selection.<br />

We present a method for mutational analysis <strong>of</strong> CTCs in metastatic breast<br />

cancer (MBC) patients by using an emulsion-formulated, semiconductorbased,<br />

targeted clonal sequencing platform. Methods: CTCs in MBC<br />

patients were enriched by a micr<strong>of</strong>luidic OncoCEE device using antibodies<br />

against both epithelial and mesenchymal markers. Genomic DNA was<br />

extracted from enriched CTC samples. Emulsion-based multiplex-PCR<br />

targeted for various cancer genes was performed, after which semiconductorbased<br />

deep sequencing was completed. The read error rates were analyzed<br />

based on quality score and context <strong>of</strong> sequence including homopolymers.<br />

Statistical significance for each mutational analysis was assessed using a<br />

method based on beta-binomial distribution. Results: Of the 17 patients<br />

samples obtained, we were able to enrich CTC samples in 9 <strong>of</strong> them (CTC<br />

range 1-1063, median�12). Multiplex targeted sequencing was performed<br />

on DNA from the enriched CTC patient samples (purity range<br />

0.3-6%). Greater than 3000-fold coverage was accomplished. Missense<br />

mutations at E545D on PIK3CA (p� 2.0e-07), F354L on STK11 (p�2.0e-<br />

04), and Q61R on NRAS (p�2.0e-07) were detected. Novel mutations <strong>of</strong><br />

L540F and Q1033K within the hot spot regions <strong>of</strong> PIK3CA were observed<br />

(p�1.3e-04). Genomic DNA from WBCs from a healthy female was<br />

analyzed concurrently as a negative control, in which none <strong>of</strong> the mutations<br />

were observed. Conclusions: Mutational analysis <strong>of</strong> CTCs in MBC patients<br />

can be accomplished by deep sequencing. We developed a de novo protocol<br />

for clonal mutation analysis on CTCs in MBC and detected various<br />

significant and novel mutations. We anticipate reporting sequencing<br />

results on CTCs and matched WBCs, as negative controls, from 40 MBC<br />

patients. This will provide the foundation for the future studies in which we<br />

will compare the mutational pr<strong>of</strong>ile between CTCs and primary/metastatic<br />

tumors. We intend to validate clonal mutational analysis <strong>of</strong> CTCs as a<br />

predictive blood-based biomarker in subsequent trials.<br />

10518 Poster Discussion Session (Board #11), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Genetic variation in CYP19A1 and response to exemestane: Survival in early<br />

breast cancer in the Dutch TEAM trial. Presenting Author: Daniel Houtsma,<br />

Department <strong>of</strong> <strong>Clinical</strong> Oncology, Leiden University Medical Center, Leiden,<br />

Netherlands<br />

Background: In patients with endocrine-sensitive breast cancer treated with<br />

adjuvant aromatase inhibitors (AI) it is unclear which patients will develop a<br />

recurrence and who will benefit from AI’s. Variations in the aromatase gene<br />

(CYP19A1) are associated with altered estrogen levels and altered aromatase<br />

activity. The aim <strong>of</strong> this study was to examine the effect <strong>of</strong> SNPs in the CYP19A1<br />

gene on survival in a prospective cohort <strong>of</strong> breast cancer patients treated with<br />

adjuvant exemestane. Methods: Patients <strong>of</strong> whom tissue was available and who<br />

were treated with five years <strong>of</strong> exemestane were selected from the Tamoxifen<br />

Exemestane Adjuvant Multinational (TEAM) trial. DNA was isolated from tumor<br />

samples and 30 SNPs were identified using a tagging SNP approach, aiming for<br />

80% coverage <strong>of</strong> CYP19A1. Genotypes were determined with taqman assays.<br />

Primary endpoint <strong>of</strong> the study was relapse-free survival (RFS) and secondary<br />

endpoint was overall survival (OS). A Kaplan-Meier analysis was performed and<br />

Cox proportional hazards models assessed survival differences. Analyses were<br />

adjusted for age at diagnosis, tumor size, nodal status, histological grade,<br />

surgery, adjuvant radiotherapy and chemotherapy. Results: 807 patients were<br />

included in the analyses and genotypes were obtained in 722 cases. A significant<br />

association with worse RFS was found with two SNPs: rs7176005 and<br />

rs16964211, showing hazard ratios (HR) <strong>of</strong> 3.48 and 5.42 for the homozygeous<br />

variant types respectively. These SNPs, as well as a third SNP, rs6493497, were<br />

also significantly associated with OS (HR 5.87, 5.3 and 3.36 respectively).<br />

Conclusions: Germline variations in the CYP19A1 gene are related to a worse<br />

outcome in early breast cancer patients treated with exemestane. These findings<br />

may contribute to the individualization <strong>of</strong> hormonal therapy in breast cancer. The<br />

relation between RFS and SNP’s in CYP19A1.<br />

Number <strong>of</strong><br />

genotype<br />

Univariate HR<br />

(95% CI) P value<br />

Multivariable HR<br />

(95% CI) P value<br />

rs7176005 0.015 0.033<br />

CC 578 1 (ref) 1 (ref)<br />

CT 130 1.83 (1.15-2.91) 1.75 (1.05-2.92)<br />

TT 8 2.78 (0.68-11.4) 3.48 (0.83-14.6)<br />

rs16964211 0.021 0.019<br />

GG 629 1 (ref) 1 (ref)<br />

AG 67 1.04 (0.52-2.06) 0.52 (0.23-1.17)<br />

AA 4 5.82 (1.43-23.7) 5.42 (1.25-23.4)<br />

10517 Poster Discussion Session (Board #10), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Intraoperative detection <strong>of</strong> lymph node metastasis using one-step nucleic<br />

acid amplification (OSNA) in breast cancer patients: Effect on second<br />

surgery rate and delay for adjuvant therapy. Presenting Author: Sophie<br />

Klingler, Centre Alexis Vautrin, Vandoeuvre-lès-Nancy, France<br />

Background: Sentinel lymph node (SLN) analysis is conventionally analysed<br />

using HES and CK19 immunohistochemistry. In case <strong>of</strong> SLN involvement,<br />

a second surgery is required for axillary lymph node (ALN) resection thus<br />

delaying the initiation <strong>of</strong> adjuvant therapies. Methods: 381 pts with invasive<br />

breast cancer were considered in this retrospective study. SLN were<br />

detected using combined radio-isotope and color detection. SLN involvement<br />

was analysed using OSNA for CK19 mRNA, in 100 pts (group 1) and<br />

compared to conventional histopathology in 281 pts (reference population,<br />

group 2). In all cases, control cytology was performed. Results: No<br />

significant difference was found between group 1 and 2 regarding patients<br />

characteristics, tumor localization, size, grade, steroid receptors and HER2<br />

expression and the mean number <strong>of</strong> SLN analysed per pt, 2.4 (range 1-7)<br />

and 2.5 (range 1-8), respectively. Considering positive SLN as “��”<br />

(CK19 mRNA copy number�5000), “�” (250 � CK19 mRNA copy<br />

number � 5000) and positive by inhibition in group 1 and macro-,<br />

micrometastases and isolated tumor cells in group 2, no difference in<br />

lymph node involvement rate was found between the two groups with 29.0<br />

and 29.9% <strong>of</strong> positive SLN, respectively. Only one discordance case was<br />

observed with negative OSNA analysis and positive cytology with isolated<br />

tumor cells. Using OSNA intraoperatively, the mean time to process SLN<br />

was 42 min (range 10-104) allowing immediate ALN resection. Significant<br />

(P�.01) reduction <strong>of</strong> re-intervention rate was observed when OSNA was<br />

used (9 vs 39%), including margins insufficiency (5 vs 13%), ALN<br />

resection (3 vs 15%) or both (1 vs 11%). Delay before adjuvant therapy was<br />

also significantly (P �.01) reduced when OSNA was used with 43 (range<br />

20-82) vs 59 (range 14-212) days. Conclusions: Results achieved with<br />

OSNA are fully consistent with those achieved using conventional immunohistochemistry<br />

analysis. SLN analysis using OSNA avoids a second operation<br />

in most patients for ALN resection and shortens the delay for adjuvant<br />

therapy.<br />

10519 Poster Discussion Session (Board #12), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

64Cu-DOTA-trastuzumab-PET imaging in patients with HER2-positive breast<br />

cancer. Presenting Author: Kenji Tamura, Department <strong>of</strong> Breast and<br />

Medical Oncology, National Cancer Center Hospital, Tokyo, Japan<br />

Background: Targeting <strong>of</strong> HER2 with trastuzumab (T) is a well-established<br />

strategy in the metastatic and adjuvant setting in HER2 positive breast<br />

cancer (HER2-BC). Although HER2 status is routinely determined using<br />

immunohistochemistry or fluorescence in situ hybridization, technical<br />

problem can arise when lesions are poorly accessible. HER2 expression can<br />

vary during the course <strong>of</strong> the disease, and there can be discordance in<br />

HER2 expression across tumor lesion even in the same patients. Noninvasive<br />

HER2 imaging is crucial needed to solve these problems. Previous<br />

imaging using 111In or 89Zn- trastuzumab produced high radiation exposure<br />

to patients by their long half life (�67.9and 78.4h) and low resolution<br />

image. Half life <strong>of</strong> 64Cu is 12.7h. We performed a feasibility study <strong>of</strong> the<br />

64Cu-1, 4, 7, 10- tetraazacylododecane-N,N’,N�,N�’-tetraacetic acid<br />

(DOTA)-T to perform PET imaging in patients with HER2-BC. Methods:<br />

Patients with HER2-BC received 150 MBq <strong>of</strong> 64Cu-DOTA-T and underwent<br />

PET scan 1, 24 and 48h after the injection. Six patients were evaluated<br />

internal dosimetry by collecting radioactivity data <strong>of</strong> blood and normal<br />

tissue in each time point from PET study, and radiation exposure by<br />

collecting clothes, linen and urine to test feasibility for outpatients. Results:<br />

Fifteen patients who received T therapy were enrolled in the “first-inhuman”<br />

trial. All patients had no severe toxicity. Radiation excretion<br />

evaluating clothes, linen were under background level. Radiation exposure<br />

<strong>of</strong> 64Cu-DOTA-T was equivalent to that <strong>of</strong> conventional 18F-FDG-PET. Distribution <strong>of</strong> liver, kidney, spleen, and blood vessel was 2-8 SUV, and<br />

uptake in other normal tissue was low. At visual examination, in two<br />

patients brain metastases were clearly visualized by 64Cu-DOTA-T-PET, suggesting that there are disruptions <strong>of</strong> the blood-brain barrier at the site <strong>of</strong><br />

the brain metastases. The sternum bone metastasis was well visualized and<br />

quantitatively monitored according to the response by T. Primary breast<br />

cancers, lymph node metastases and lung metastases could also be<br />

visualized at the lesion indentified by CT. Conclusions: 64Cu-DOTA-T-PET was feasible test even for outpatient, and provides specific and high<br />

resolution image in HER2 positive lesion.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


10520 Poster Discussion Session (Board #13), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Reproducibility, performance, and clinical utility <strong>of</strong> a genetic risk prediction<br />

model for prostate cancer in Japanese patients. Presenting Author:<br />

Shusuke Akamatsu, Department <strong>of</strong> Urology, Kyoto University Graduate<br />

School <strong>of</strong> Medicine, Kyoto, Japan<br />

Background: Prostate specific antigen (PSA) is widely used as a diagnostic<br />

biomarker for prostate cancer (PC). However, due to its low predictive<br />

performance, many patients without PC suffer from the harms <strong>of</strong> unnecessary<br />

prostate needle biopsies. The present study aims to evaluate the<br />

reproducibility and performance <strong>of</strong> a genetic risk prediction model and<br />

estimate its utility as a diagnostic biomarker in a clinical scenario. Methods:<br />

We created a logistic regression model incorporating 16 SNPs that were<br />

significantly associated with PC in a genome-wide association study <strong>of</strong> the<br />

Japanese. The model was validated by two independent sets <strong>of</strong> samples<br />

comprising 3,294 cases and 6,281 controls. Various cut <strong>of</strong>fs were<br />

evaluated to be used in a clinical scenario. Results: The area under a curve<br />

(AUC) <strong>of</strong> the model was 0.679, 0.655, and 0.661 for the samples used to<br />

create the model and those used for validation respectively. The AUC <strong>of</strong> the<br />

model was not significantly altered in samples with PSA 1-10 ng/ml.<br />

24.2% and 9.7% <strong>of</strong> the patients had odds ratio �0.5 (low risk) or �2 (high<br />

risk) in the model, and assuming the overall positive rate <strong>of</strong> prostate needle<br />

biopsies to be 20% in PSA gray zone (PSA 2-10 ng/ml), the positive biopsy<br />

rates were 10.7% and 42.4% respectively for the two genetic risk groups.<br />

Conclusions: The genetic risk prediction model was highly reproducible,<br />

and its predictive performance was not influenced by PSA. The model could<br />

have a potential to affect clinical decision when it is applied to patients<br />

with gray-zone PSA, which should be confirmed in future clinical studies.<br />

10522 Poster Discussion Session (Board #15), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

The effect <strong>of</strong> two BRM promoter variants on the risk <strong>of</strong> stage I/II upper<br />

aerodigestive tract cancers. Presenting Author: Kit Man Wong, University <strong>of</strong><br />

Toronto, Toronto, ON, Canada<br />

Background: BRM is a key subunit <strong>of</strong> the chromatin remodeling complex<br />

SWI/SNF and a putative tumor suppressor gene that is silenced in 15-20%<br />

<strong>of</strong> many solid tumors (PMID 15722796). Evidence suggests that it is<br />

epigenetically regulated, as two BRM promoter insertion variants (BRM-<br />

741 and BRM-1321) may lead to gene silencing by recruiting histone<br />

deacetylases. The presence <strong>of</strong> both homozygous BRM-741 and BRM-1321<br />

highly correlate with loss <strong>of</strong> BRM expression and function in lung tumors,<br />

while increasing smoking-related lung cancer by two-fold (PMID<br />

21478907). Also, the pharmacologic reversal <strong>of</strong> epigenetic changes <strong>of</strong><br />

BRM <strong>of</strong>fers a potential novel therapeutic approach (PMID 21478905). We<br />

assessed whether these BRM variants are associated with the risk <strong>of</strong> upper<br />

aerodigestive tract cancers, focusing on Stage I/II tumors that would most<br />

benefit from new screening and prevention strategies. Methods: BRM was<br />

genotyped by qPCR using TaqMan probes. 1,008 controls were matched to<br />

595 cases by frequency distribution based on age, gender and smoking<br />

status. Multivariate logistic regression generated adjusted odds ratios<br />

(aOR). Results: The 595 cases were: 115 esophageal, 278 lung, and 202<br />

head and neck cancers. 51% were adenocarcinomas; 60% were Stage I.<br />

The frequency <strong>of</strong> homozygozity was: BRM-741, 26%; BRM-1321, 23%;<br />

both variants, 15%. In the combined analysis, there was significant<br />

correlation between malignancy and homozygous BRM-741 (aOR 1.91<br />

(95%CI 1.3-2.4); p�0.001) or BRM-1321 (aOR 1.94 (95%CI 1.4-2.7;<br />

p�3x10E-4). Being homozygous for both BRM variants carried an even<br />

greater risk (aOR 2.45 (95%CI 1.6-3.9); p�1x10E-5). This correlation was<br />

similar for adenocarcinomas (aOR 2.53 (95%CI 1.4-4.2); p�6x10E-4)<br />

and squamous cell carcinomas (aOR 2.33 (95%CI 1.3-4.4); p�8x10E-4).<br />

The increased cancer risk was also similar between these subgroups: head<br />

and neck, esophageal and lung cancers; Stage I and II patients; smokers<br />

and non-smokers. Conclusions: The two homozygous BRM variants increase<br />

the risk <strong>of</strong> early stage upper aerodigestive tumors by more than two-fold<br />

independent <strong>of</strong> smoking status. BRM promoter variants and their potential<br />

epigenetic effect may be early events in the evolution <strong>of</strong> these cancers.<br />

Tumor Biology<br />

661s<br />

10521 Poster Discussion Session (Board #14), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

BAP1: The first mutated gene causing familial uveal melanoma. Presenting<br />

Author: Johan Hansson, Department <strong>of</strong> Oncology-Pathology, Stockholm,<br />

Sweden<br />

Background: Uveal melanoma (UM) is a rare malignancy with a poor<br />

prognosis. Familial predisposition to UM is rare and accounts for only a few<br />

percent <strong>of</strong> all cases. The genetic background <strong>of</strong> hereditary UM is unknown<br />

and the aim <strong>of</strong> our project was to identify susceptibility gene(s) for UM.<br />

Methods: We identified a family with hereditary predisposition for UM – the<br />

proband <strong>of</strong> which is a young female diagnosed with UM at age 16 who<br />

within 6 months developed liver metastases. We also identified two older<br />

paternal relatives who were diagnosed with UM at 39 and 44 years <strong>of</strong> age,<br />

respectively. We performed massively parallel sequencing using the Illumina<br />

Hiseq2000 technology on germline DNA from the proband, her<br />

parents and a healthy sibling. After QC and mapping against the human<br />

reference genome the average coverage across the exome was between 35<br />

and 86 for the four sequenced samples. Results: Out <strong>of</strong> more than 260,000<br />

single nucleotide variants (SNVs) and small insertion / deletion variants<br />

(indels), 51 gene variants were filtered out by being novel, shared by the<br />

affected proband and her father (considered an obligate mutation carrier),<br />

but not by the healthy mother, <strong>of</strong> predicted functional importance and /or<br />

located within strongly conserved regions. The strongest candidate among<br />

these was a loss <strong>of</strong> function-variant in the BAP1 gene, since BAP1 has been<br />

suggested as a tumor suppressor in several cancer-related syndromes,<br />

including cases <strong>of</strong> UM. The sequence data indicated an insertion <strong>of</strong> one<br />

base-pair in exon 3 <strong>of</strong> the BAP1 genecausing a frame-shift and subsequently<br />

a truncated protein lacking all its functional domains. The<br />

mutation was also present in UM tumor tissue from the two deceased<br />

paternal relatives and was found to segregate with the UM phenotype in the<br />

family. We also detected loss <strong>of</strong> heterozygosity in the tumor <strong>of</strong> the proband,<br />

supporting BAP1 as the causative gene in this family. Conclusions: The<br />

identification <strong>of</strong> BAP1 as the gene responsible for this syndrome is the first<br />

demonstration <strong>of</strong> a germline mutation causing UM. This enables us to<br />

identify and monitor risk individuals belonging to mutation positive<br />

families with predisposition to UM, and possibly other cancer syndromes.<br />

We are continuously screening other cases <strong>of</strong> familial UM for mutations in<br />

BAP1.<br />

10523 Poster Discussion Session (Board #16), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Prognostic significance <strong>of</strong> LINE-1 methylation level in the upper gastrointestinal<br />

cancer. Presenting Author: Yoshifumi Baba, Kumamoto University,<br />

Kumamoto, Japan<br />

Background: Genome-wide DNA hypomethylation plays a role in genomic<br />

instability and carcinogenesis. DNA methylation in long interspersed<br />

nucleotide element-1 (LINE-1) is a good indicator <strong>of</strong> global DNA methylation<br />

level. Although LINE-1 methylation level is attracting interest as a<br />

useful marker for predicting cancer prognosis, the prognostic significance<br />

<strong>of</strong> LINE-1 hypomethylaiton in the upper gastrointestinal cancer [i.e.,<br />

esophageal squamous cell carcinoma (ESCC) and gastric cancer (GC)]<br />

remains unclear. Methods: Using 217 ESCC and 207 GC specimens, we<br />

quantified the LINE-1 methylation using bisulfite-pyrosequencing technology.<br />

During the follow-up, there were a total <strong>of</strong> 63 ESCC recurrences, 51<br />

ESCC deaths and 56 GC deaths. The median follow-up time for censored<br />

patients was 2.8 years. A Cox proportional hazards model was used to<br />

calculate the hazard ratio (HR), adjusted for the clinical, epidemiological,<br />

and pathological variables. The term “prognostic marker” is used throughout<br />

this study according to the REMARK Guidelines. Results: ESCCs and<br />

GCs showed significantly lower LINE-1 methylation levels compared to<br />

matched normal mucosa (p�0.0001). In ESCC, LINE-1 hypomethylation<br />

was significantly associated with disease-free survival [log-rank p�0.0008;<br />

univariate HR� 2.32, 95% confidence interval (CI) 1.38-3.84, p�0.0017;<br />

multivariate HR�1.81, 95% CI 1.06-3.05, p�0.031] and cancer-specific<br />

survival (log-rank p�0.0020; univariate HR�2.21, 95% CI 1.33-3.60,<br />

p�0.0026; multivariate HR�1.87, 95% CI 1.12-3.08, p�0.018]. We<br />

found a significant modifying effect <strong>of</strong> the tumor stage on the relationship<br />

between LINE-1 methylation and the recurrence rate (P for interaction �<br />

0.031). In GC, LINE-1 hypomethylation was significantly associated with<br />

cancer-specific survival (log-rank p�0.029; univariate HR�2.01, 95% CI<br />

1.09-3.99). Conclusions: LINE-1 hypomethylation is associated with shorter<br />

survival in both ESCC and GC, suggesting that it has potential for use as a<br />

prognostic biomarker.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


662s Tumor Biology<br />

10524 Poster Discussion Session (Board #17), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Next-generation sequencing <strong>of</strong> FFPE solid tumor specimens for clinical<br />

use. Presenting Author: Roman Yelensky, Foundation Medicine, Cambridge,<br />

MA<br />

Background: As more therapies targeting genomic alterations become<br />

available, genotyping (e.g., by Sequenom) is increasingly performed in<br />

tumor types where mutational status may drive treatment choice. Nextgeneration<br />

sequencing (NGS) technology can expand on genotyping <strong>of</strong><br />

individual base pairs because it can detect mutations across entire exons,<br />

copy changes and fusion genes. However, for NGS to be clinically viable, it<br />

must be made compatible with FFPE tissues and shown concordant with<br />

best current diagnostic methods. Methods: To explore a potential clinical<br />

role for NGS, we selected 120 FFPE specimens (68 NSCLC, 32 CRC, 20<br />

melanoma) previously tested for 97 oncogenic mutations in 8 oncogenes by<br />

Sequenom, and sequenced all exons <strong>of</strong> 182 cancer genes. Genotyping and<br />

sequencing were both performed in CLIA compliant labs. DNA was<br />

extracted from 4x 10� unstained sections from the diagnostic FFPE block,<br />

followed by library construction and hybridization capture <strong>of</strong> 3230 exons<br />

and 37 commonly rearranged introns. Average coverage <strong>of</strong> �900X uniquelymapping<br />

reads was obtained. Sequence data were analyzed for all genomic<br />

alterations and examined for potentially actionable mutations. Results:<br />

High concordance was noted between Sequenom and NGS: 103 and 105<br />

mutations were called by the two technologies, respectively, at mutually<br />

tested sites, with 97 mutation calls in common. Notably, mutant allele<br />

frequencies in concordant calls ranged as low as 2% by NGS, highlighting<br />

the sensitivity <strong>of</strong> detection enabled by both approaches. Furthermore, in<br />

45/120 (38%) specimens, NGS revealed additional alterations that may<br />

confer sensitivity or resistance to approved or experimental targeted<br />

therapies and thus plausibly influence treatment decisions. These included<br />

39 copy changes or loss-<strong>of</strong>-function variants best assayed by an NGS<br />

approach. Conclusions: Our results demonstrate technical feasibility and<br />

highlight potential benefits <strong>of</strong> comprehensive cancer gene characterization<br />

through next-generation sequencing <strong>of</strong> clinical FFPE specimens. As NGS is<br />

the most practical means to detect all classes <strong>of</strong> somatic alteration in a<br />

small, clinically relevant sample, we suggest that this type <strong>of</strong> testing will<br />

become an essential component <strong>of</strong> patient care.<br />

10526 Poster Discussion Session (Board #19), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Frequency and clinical characterization <strong>of</strong> NSCLC patients harboring<br />

PIK3CA mutations identified within a regional screening network. Presenting<br />

Author: Masyar Gardizi, Lung Cancer Group Cologne, Center for<br />

Integrated Oncology, University Hospital Cologne, Cologne, Germany<br />

Background: PIK3CA mutations are a rare oncogenic event <strong>of</strong> potential<br />

therapeutic relevance in NSCLC. Here we report frequency and characteristics<br />

<strong>of</strong> patients with PIK3CA mutated lung tumors. Methods: Patients with<br />

NSCLC and PIK3CA mutations were identified within our regional Network<br />

for Molecular Screening in Lung Cancer. We further analyzed the presence<br />

<strong>of</strong> BRAF, KRAS, EGFR mutations as well as ALK translocation, ERBB2 and<br />

FGFR1 amplifications in PIK3CA mutated samples. <strong>Clinical</strong> data on age,<br />

sex, TNM classification and tumor stage, histological type, grading, overall<br />

survival, smoking status, comorbidity, BMI and secondary malignancies<br />

were retrieved from clinical charts in accordance with the local ethics<br />

committee. Results: PIK3CA mutations were detected with a frequency <strong>of</strong><br />

3.7% (24% exon 20,76% exon 9) in 1000 patients. Histologically 32%<br />

were defined as squamous cell carcinoma, 48% as adenocarcinoma and<br />

18% other histological subtypes or NSCLC-NOS. Exon 9 mutations were<br />

present in the acinar and lepidic subtype, whereas exon 20 mutations were<br />

seen in the papillary and solid subtype. Cooccuring genetic lesions were<br />

observed in 16% (mutations in KRAS�2, EGFR�1, BRAF�1; FGFR1<br />

amplification�2). 14 were female, 23 male with a mean age <strong>of</strong> 69 years.<br />

21 <strong>of</strong> these patients were further clinically annotated. 11 patients<br />

presented with stage IIIb/IV eligible for palliative treatment and 10 stage I –<br />

IIIa eligible for surgical therapy �/- adjuvant therapy. All but 1 patient were<br />

smokers with an average BMI <strong>of</strong> 26,2kg/m2 with a typical high load <strong>of</strong><br />

comorbidity mainly <strong>of</strong> cardiovascular diseases, 8 <strong>of</strong> 21 patients showed<br />

prior malignancies in their medical history. The median overall survival<br />

within this population has not been reached yet. Conclusions: Screening for<br />

PIK3CA mutations is feasible. A high proportion (38%) <strong>of</strong> patients with<br />

PIK3CA mutated lung cancer have prior malignancies and show a high load<br />

<strong>of</strong> comorbidity. Furthermore PIK3CA mutations are not exclusive to KRAS,<br />

EGFR or BRAF mutations or FGFR1 amplifications. Successful identification<br />

<strong>of</strong> patients with oncogenic lesions in lung cancer in a screening<br />

network might allow future personalized treatment <strong>of</strong> these patients.<br />

10525 Poster Discussion Session (Board #18), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Prospective study <strong>of</strong> oncogenic mutations in circulating cell-free DNA<br />

(cfDNA) using a multiplex sequencing platform for patient (pt) allocation to<br />

phase I clinical trials. Presenting Author: Michael Ong, Royal Marsden<br />

Hospital and Institute <strong>of</strong> Cancer Research, Sutton, United Kingdom<br />

Background: Studies suggest that tumoral cfDNA can be isolated from<br />

plasma and used as a biomarker. Methods: We conducted a prospective<br />

study in adult pts with advanced cancer referred for phase I clinical trials<br />

Sep09-Dec10 (A: Pilot Phase n�104) and Jan11-Sep11 (B: Expansion<br />

Phase n�176). cfDNA and tumor DNA from formalin-fixed paraffinembedded<br />

(FFPE) tissue were isolated, quantified, and analyzed for 238<br />

mutations in 19 oncogenes by the Sequenom OncoCarta Panel 1.0.<br />

Mutation data were used for trial allocation when available. Pt data and<br />

outcomes were collected. Statistical inferences used Wilcoxon rank-sum<br />

and Pearson’s Chi-squared. Results: 280 pts enrolled including breast (60),<br />

colorectal (42), ovarian (37), lung (31), and prostate (20); median age<br />

60yrs [23-81]; 93% ECOG PS 0-1. cfDNA in 95% and FFPE in 60% were<br />

analyzed. Median time to cfDNA result, trial allocation and consent were 7<br />

[5-14], 6 and 23 d. Median cancer pt cfDNA concentration (ng/ml) (A: 18<br />

[1-1600]; B: 34 [16-1657]) was higher than 20 healthy volunteers (6<br />

[5-13] A: p�0.0001) and higher in metastatic vs locally advanced cancer<br />

(A: 21 vs 4 p�0.0006; B: 42 vs 24 p�0.066). Pts with higher than<br />

median [cfDNA] had shorter median survival (MST) than below (A: 217 vs<br />

274 d, HR 1.72 95% CI 1.07-2.65, p�0.025; B: MST not reached yet).<br />

Mutations were found in 89 pts (32%), including KRAS (11%), PIK3CA<br />

(5%), MET (5%), BRAF (4%), and AKT (2%). cfDNA mutations (57 pts,<br />

21.5%) were mainly found above the median [cfDNA] (A: 41 vs 17%<br />

�2�6.9, p�0.009; B: 24 vs 11% �2�5.0, p�0.025). Of 155 pts with<br />

both cfDNA and FFPE available, 68 had mutations detected: 30 identical<br />

in cfDNA and FFPE; 28 in FFPE alone and 10 in cfDNA alone. Overall, 153<br />

pts were treated on a phase I trial; 23 pts had relevant mutation data and<br />

received a trial targeting the PI3-Akt-mTOR axis. Conclusions: Higher<br />

cfDNA is associated with greater disease burden, worse prognosis, and<br />

higher mutation detection rate, suggesting cfDNA is tumorally derived.<br />

Moreover, cfDNA can be analyzed for oncogenic mutations in a time-frame<br />

suitable for clinical decision making, making it a potentially useful<br />

non-invasive adjunct to tumour DNA analysis.<br />

10527 Poster Discussion Session (Board #20), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Association <strong>of</strong> EGFR mutation or ALK rearrangement with expression <strong>of</strong><br />

DNA repair and synthesis genes in never-smoker females with pulmonary<br />

adenocarcinoma. Presenting Author: Xiaoxia Chen, Department <strong>of</strong> Oncology,<br />

Shanghai Pulmonary Hospital, Tongji University; Tongji University<br />

Medical School Cancer Institute, Shanghai, China<br />

Background: Epidermal growth factor receptor (EGFR) mutation or anaplastic<br />

lymphoma kinase (ALK) rearrangement was found not only predict the<br />

efficacy <strong>of</strong> targeted drugs, but also associate with the efficacy <strong>of</strong> chemotherapy<br />

drugs in non-small cell lung cancer (NSCLC) patients. We investigated<br />

the relationship <strong>of</strong> EGFR mutation status or ALK rearrangement and<br />

DNA repair or synthesis genes, such as excision repair cross-complementing<br />

1 (ERCC1), ribonucleotide reductase subunit M1 (RRM1), thymidylate<br />

synthetase (TS) and breast cancer gene one (BRCA1) gene expression, as a<br />

potential explanation for these observations. Methods: In this surgical<br />

series, 104 resected lung adenocarcinomas from nonsmoker females were<br />

analyzed concurrently for the EGFR mutation, ALK rearrangement status<br />

and mRNA expression <strong>of</strong> ERCC1, RRM1, TS and BRCA1 genes. EGFR<br />

mutation detection was performed by the method <strong>of</strong> ADx-ARMS, ALK<br />

rearrangement was detected by PCR and the mRNA expression <strong>of</strong> different<br />

genes were tested using the method <strong>of</strong> real-time PCR. Results: 73 (70.2%)<br />

patients harbored EGFR mutations and 10 (9.6%) had ALK rearrangement.<br />

The ERCC1 mRNA level in patients with EGFR mutation was<br />

3.44�1.94�10 -3<br />

, which is significantly lower than in the patients with<br />

ALK positive and both negative(4.60�1.95�10 -3 and 4.95�2.33�10 -3<br />

respectively, P�0.010). While the TS mRNA levels were significantly lower<br />

in the patients with EGFR mutation(1.15�1.38�10 -3 VS<br />

2.69�3.97�10 -3 , P�0.006) or ALK positive (1.21�0.78�10 -3 VS<br />

2.69�3.97�10 -3 , P�0.020) than in patients with both negative.<br />

Conclusions: NSCLC specimens harboring activating EGFR mutations are<br />

more likely to express low ERCC1 and TS mRNA levels, while NSCLC<br />

patients with ALK rearrangement are more likely to express low TS mRNA<br />

levels, which could be helpful to select a proper chemotherapy regimen for<br />

NSCLC patients with known EGFR mutation or ALK fusion status.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


10528 Poster Discussion Session (Board #21), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

New microRNA-based diagnostic test for lung cancer classification. Presenting<br />

Author: Ranit Aharonov, Rosetta Genomics, Rehovot, Israel<br />

Background: Lung cancer is the leading cause <strong>of</strong> cancer deaths in the US.<br />

Treatment options are determined by tumor subtyping, for which there is<br />

lack <strong>of</strong> standardized, objective, and highly accurate techniques. In 20%-<br />

30% <strong>of</strong> cases significant limitations <strong>of</strong> tumor quantity and quality prevent<br />

full classification <strong>of</strong> the tumor using traditional diagnostic methods. Using<br />

microRNA microarray data generated from over a hundred formalin-fixed,<br />

paraffin-embedded (FFPE) primary lung cancer samples, we have identified<br />

microRNA expression pr<strong>of</strong>iles that differ significantly for the main lung<br />

cancer types. Based on these findings, we have developed and validated a<br />

microRNA-based qRT-PCR assay that differentiates primary lung cancers<br />

into four types: squamous cell carcinoma, non-squamous non-small cell<br />

lung cancer, carcinoid and small cell carcinoma. Methods: Over 700<br />

primary tumor samples from different histological types <strong>of</strong> lung cancer were<br />

collected. Samples included FFPE blocks from resection or biopsies and<br />

cell blocks from cytology specimens including fine needle aspiration,<br />

bronchial brushing and bronchial washing. High-quality RNA was extracted<br />

from the samples using proprietary protocols. Expression levels <strong>of</strong> potential<br />

microRNA biomarkers were pr<strong>of</strong>iled using microarrays followed by a<br />

sensitive and specific qRT-PCR platform. An assay for lung tumors<br />

classification using 8 microRNAs on qRT-PCR was developed based on<br />

data from 261 samples. This assay was validated on an independent<br />

blinded set <strong>of</strong> 451 cytological and pathological samples. Results: Using the<br />

expression levels <strong>of</strong> 8 microRNAs measured in qRT-PCR, accurate classification<br />

<strong>of</strong> the primary lung tumors into the four main cancer types is<br />

obtained. The microRNA-based assay reached an accuracy <strong>of</strong> 94%.<br />

Moreover, cytological samples composed over 50% <strong>of</strong> the validation set<br />

and reached an accuracy <strong>of</strong> 95%. Conclusions: We present here a new<br />

microRNA-based assay for the classification <strong>of</strong> the four main types <strong>of</strong> lung<br />

cancer based only on the expression <strong>of</strong> 8 microRNAs. This assay displays<br />

very high levels <strong>of</strong> accuracy for both pathological and cytological samples.<br />

The assay comprises a standardized, well-tested and objective tool which<br />

can assist physicians in the diagnosis <strong>of</strong> lung cancer.<br />

10530 Poster Discussion Session (Board #23), Tue, 8:00 AM-12:00 PM and<br />

11:30 AM-12:30 PM<br />

Molecular gene expression pr<strong>of</strong>iling to predict the tissue <strong>of</strong> origin and direct<br />

site-specific therapy in patients (pts) with carcinoma <strong>of</strong> unknown primary<br />

site (CUP): Results <strong>of</strong> a prospective Sarah Cannon Research Institute<br />

(SCRI) trial. Presenting Author: Frank A. Greco, SCRI/Tennessee Oncology,<br />

PLLC, Nashville, TN<br />

Background: Tumor pr<strong>of</strong>iling is an emergent technique to determine tissue<br />

<strong>of</strong> origin in CUP patients. However, the value <strong>of</strong> these predictions in<br />

improving treatment efficacy is unknown. In this prospective trial, we used<br />

tumor pr<strong>of</strong>iling results to direct site-specific therapy for CUP pts. Methods:<br />

A 92-gene RT-PCR assay (CancerTYPE ID; bioTheranostics, Inc.) was<br />

performed on tumor biopsies from previously untreated CUP pts who<br />

consented. When a tissue <strong>of</strong> origin was predicted, pts who were treatment<br />

candidates were assigned standard site-specific first-line therapy. Results:<br />

Between 10/08 and 12/11, 289 pts were enrolled, 252 had successful<br />

assays performed, and 247 (98%) had a tissue <strong>of</strong> origin predicted. 224 pts<br />

were eligible for treatment; 197 pts received assay-directed treatment.<br />

120 <strong>of</strong> 224 treated pts (54%) had assay diagnoses <strong>of</strong> tumor types known to<br />

derive substantial benefit from standard site-specific treatment (bladder<br />

27, colorectal 26, NSCLC 24, breast 10, ovary 10, kidney 9, prostate 4,<br />

germ cell 4, others 6 (3 sites), while 104 pts (46%) had assay diagnoses <strong>of</strong><br />

relatively resistant tumors (biliary tract 45, pancreas 12, gastroesophageal<br />

10, liver 7, sarcoma 5, cervix 5, others 20 (8 sites). Median OS for all<br />

treated pts was 10.8 months (mos); OS for 197 pts with assay-directed<br />

treatment was 12.2 mos (versus 6.0 mos for 27 pts receiving empiric<br />

therapy). Median OS was better in the 120 pts with assay diagnoses <strong>of</strong> more<br />

responsive tumor types (12.8 vs 7.4 mos; p � .027). Median OS (mos) in<br />

specific subgroups: pancreas 9, kidney 12, colon 12, NSCLC 16, ovary 30.<br />

Conclusions: This is the first prospective trial in which molecular pr<strong>of</strong>iling<br />

has directed site-specific therapy in CUP pts. Assay-directed therapy in<br />

197 pts produced a median OS (12.2 mos) that compares favorably with<br />

previous empiric CUP therapy. CUP pts predicted to have more responsive<br />

tumor types had longer survival compared to less responsive types,<br />

suggesting accurate identification by the assay. These results strengthen<br />

the rationale for molecular pr<strong>of</strong>iling in CUP management.<br />

Tumor Biology<br />

663s<br />

CRA10529 Poster Discussion Session (Board #22), Tue, 8:00 AM-12:00 PM<br />

and 11:30 AM-12:30 PM<br />

Regional screening network for characterization <strong>of</strong> the molecular epidemiology<br />

<strong>of</strong> non-small cell lung cancer (NSCLC) and implementation <strong>of</strong><br />

personalized treatment. Presenting Author: Thomas Zander, Lung Cancer<br />

Group Cologne, Center for Integrated Oncology, University Hospital Cologne,<br />

Cologne, Germany<br />

The full, final text <strong>of</strong> this abstract will be available at<br />

abstract.asco.org at 12:01 AM (EDT) on Monday, June 4,<br />

2012, and in the <strong>Annual</strong> <strong>Meeting</strong> <strong>Proceedings</strong> online<br />

supplement to the June 20, 2012, issue <strong>of</strong> Journal <strong>of</strong><br />

<strong>Clinical</strong> Oncology. Onsite at the <strong>Meeting</strong>, this abstract will<br />

be printed in the Monday edition <strong>of</strong> ASCO Daily News.<br />

10531 General Poster Session (Board #41D), Mon, 1:15 PM-5:15 PM<br />

Homeobox B9: A potential surrogate marker for bevacizumab in combination<br />

with chemotherapy in colorectal cancer. Presenting Author: Yoshinori<br />

Hoshino, Department <strong>of</strong> Surgery, Keio University School <strong>of</strong> Medicine,<br />

Tokyo, Japan<br />

Background: Homeobox B9 (HOXB9) is known to be overexpressed in<br />

human breast cancer and pr<strong>of</strong>oundly related to tumorigenicity, lung<br />

metastasis and radio-resistance. (Hayashida, PNAS 2010, and Chiba,<br />

PNAS 2011). However, little is known about the relation between the<br />

expression <strong>of</strong> HOXB9 and angiogenesis in colorectal cancer (CRC). We<br />

aimed to clarify the impact <strong>of</strong> HOXB9 in CRC and evaluate the importance<br />

for bevacizumab treatment. Methods: The expression <strong>of</strong> HOXB9 in human<br />

CRC specimens was analyzed. Then, we introduced HOXB9 construct into<br />

human CRC cell lines and examined TGF� signaling and angiogenic<br />

factors. Xenograft model was established by these cell lines either with or<br />

without the administration <strong>of</strong> bevacizumab (5mg/kg, weekly) intraperitoneally.<br />

Finally, we examined the mRNA levels <strong>of</strong> consecutive patients who<br />

were treated by chemotherapy with bevacizumab in our institute and<br />

calculated the Kaplan- Meier curve with log-rank test. Results: 47 <strong>of</strong> 69<br />

surgical specimens (67%) showed positive expression <strong>of</strong> HOXB9 mRNA.<br />

The high HOXB9 mRNA levels significantly correlated with poor differentiation<br />

and liver metastasis. The HOXB9-overexpressed cell lines showed<br />

significantly higher expression <strong>of</strong> TGF� signaling target genes and angiogenic<br />

factors. HOXB9 overexpression significantly increased tumor volume<br />

and burden with higher microvessel density in vivo, even though the cell<br />

proliferation decreased in vitro. Notably, HOXB9-overexpressed tumor was<br />

dramatically shrunk by administration <strong>of</strong> bevacizumab (tumor shrinkage<br />

rate; 93% vs. 42% in HT29, 83% vs. 27% in HCT116). Patients with high<br />

expression <strong>of</strong> HOXB9 in tumor showed significantly longer progression free<br />

and overall survival periods (n�39). Conclusions: Our results demonstrated<br />

that patients with high expression <strong>of</strong> HOXB9 in tumor had better prognosis<br />

with bevacizumab treatment but worse without. In vivo and in vitro<br />

experiments revealed that HOXB9 might orchestrate angiogenesis and<br />

establish positive feedback between cancer cells and microenvironment.<br />

Bevacizumab might inhibit the feedback to reduce tumor growth dramatically.<br />

Therefore, HOXB9 may work as a potential surrogate marker <strong>of</strong><br />

bevacizumab treatment in CRC.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


664s Tumor Biology<br />

10532 General Poster Session (Board #41E), Mon, 1:15 PM-5:15 PM<br />

VEGF polymorphism may be associated with overall survival in lapatinibtreated<br />

breast cancer patients with brain metastases. Presenting Author:<br />

Colin F. Spraggs, GlaxoSmithKline, Stevenage, United Kingdom<br />

Background: Lapatinib is a dual HER2/EGFR tyrosine kinase inhibitor (TKI)<br />

approved in combination with capecitabine or letrozole for patients with<br />

HER2� metastatic breast cancer (MBC). Consistent with TKI therapies,<br />

patient response is variable and suggestive <strong>of</strong> additional determinants <strong>of</strong><br />

sensitivity and resistance. This exploratory pharmacogenetic study sought<br />

to identify germline genetic variants that associate with lapatinib treatment<br />

outcomes in HER2� MBC patients. Methods: Fifty five functional single<br />

nucleotide polymorphisms (SNPs) in 24 candidate genes were evaluated in<br />

a subset <strong>of</strong> MBC patients participating in two clinical trials: EGF105084:<br />

lapatinib monotherapy in HER2� MBC patients with recurrent brain<br />

metastases following trastuzumab and cranial radiotherapy (n�120) and<br />

EGF104900: lapatinib plus trastuzumab (n�92) and lapatinib monotherapy<br />

(n�103) in HER2� MBC patients with disease progression<br />

following trastuzumab. Testing for associations <strong>of</strong> SNPs with progression<br />

free survival (PFS) and overall survival (OS) during lapatinib treatment was<br />

performed using Cox proportional hazards methods, with covariate adjustment.<br />

Markers were considered to be significantly associated if they<br />

achieved a predefined multiple testing threshold <strong>of</strong> p�0.0003. Results: No<br />

SNPs were significantly associated with PFS in either study. A SNP in<br />

VEGFA (rs3025039, 936C�T) was significantly associated with improved<br />

OS (p�0.0002) for the T allele carriers, with an allelic hazard ratio <strong>of</strong> 0.21<br />

(0.08-0.52) in EGF105084. The association was not seen in EGF104900.<br />

This SNP is located in the 3’ UTR gene region, the T allele is associated<br />

with lower serum VEGFA levels and reduced breast cancer risk [Krippl et al,<br />

2003, Int J Cancer 106: 468; Kataoka et al, 2006, Cancer Epidemiol<br />

Biomarkers Prev, 15:1148]. Conclusions: A germline variant in VEGFA may<br />

be associated with survival outcome in MBC patients with brain metastases<br />

who are treated with lapatinib. This may represent activation <strong>of</strong> VEGF<br />

angiogenic pathways to overcome HER2 inhibition in patients carrying the<br />

higher expression genotype. These associations are considered exploratory<br />

and require confirmation in an independent dataset.<br />

10534 General Poster Session (Board #41G), Mon, 1:15 PM-5:15 PM<br />

Molecular characterization <strong>of</strong> circulating tumor cells in human metastatic<br />

colorectal cancer. Presenting Author: Jorge Barbazan, Translational Oncology<br />

Laboratory, Medical Oncology Department, Santiago de Compostela,<br />

Spain<br />

Background: Metastatic colorectal cancer (mCRC) relies on the detachment<br />

<strong>of</strong> aggressive malignant cells from the primary tumor into the bloodstream,<br />

being this Circulating Tumor Cells (CTC) the principal source <strong>of</strong> the further<br />

metastasis. <strong>Clinical</strong>ly, the presence <strong>of</strong> CTC is associated with poor<br />

prognosis and there exists a clear necessity for more specific and efficient<br />

chemotherapies in the treatment <strong>of</strong> mCRC. Our aim was to overcome this<br />

adverse scenario through the massive molecular pr<strong>of</strong>iling <strong>of</strong> the CTC<br />

population isolated from mCRC patients. Methods: CTC’s were magnetically<br />

isolated by using anti-EpCAM coupled magnetic beads from 6 mCRC<br />

patients and 3 healthy controls. The presence <strong>of</strong> CTCs was evaluated by<br />

citokeratins 8, 18 and 19 staining. RNA from CTCs was amplified by a<br />

whole transcriptome amplification system (WTA) and resulting material was<br />

hybridized onto gene expression arrays. Bioinformatics were used to array<br />

analysis. Selected genes were validated by RT-qPCR. Results: 410 transcripts<br />

were found to specifically characterise the CTC population after<br />

array signals comparison between mCRC patients and controls. Gene-gene<br />

interaction analysis generated networks related with cell migration, adhesion<br />

or apoptosis resistance. Gene ontology revealed similar functions.<br />

Signalling pathways such as RhoA, PKA, ILK, integrins or actin cytoskeleton<br />

signalling were found as relevant in CTCs. Eleven genes (TGFB1, APP,<br />

TIMP1, CD9, CLU, ITGB5, LIMS1, RSU1, VCL, BMP6 and TLN1) were<br />

validated by real-time PCR in 20 mCRC patient and 10 control samples. All<br />

genes showed a significantly higher expression in patients (p�0.0001).<br />

Except from TLN1 and CD9, all genes had a prognostic value in terms <strong>of</strong><br />

progression free survival (p�0.05). Conclusions: We described the molecular<br />

pr<strong>of</strong>iling <strong>of</strong> CTCs in stage IV CRC patients, characterized by a migratory<br />

and invasive phenotype. Specific and sensitive diagnostic/prognostic markers<br />

were obtained. Our strategy represents an innovative and promising<br />

approach in the clinical management <strong>of</strong> mCRC patients.<br />

10533 General Poster Session (Board #41F), Mon, 1:15 PM-5:15 PM<br />

Isolation <strong>of</strong> circulating tumor cells using a novel EMT-based capture<br />

method. Presenting Author: Rhonda Lynn Bitting, Duke Cancer Institute,<br />

Durham, NC<br />

Background: Circulating tumor cells (CTCs) have potential prognostic,<br />

predictive and surrogate implications in oncology. In patients with metastatic<br />

castration-resistant prostate cancer (CRPC) and breast cancer (BC),<br />

we have shown that CTCs isolated using epithelial cell adhesion molecule<br />

(EpCAM) ferromagnetic capture express mesenchymal markers, including<br />

N- and O-cadherin, suggesting phenotypic plasticity and the presence <strong>of</strong><br />

epithelial-mesenchymal transitions (EMT). Therefore, we postulate that<br />

during metastasis, tumor cells exist as a spectrum <strong>of</strong> epithelial to<br />

mesenchymal phenotypes and may not be captured with existing EpCAMbased<br />

CTC technology. The goal <strong>of</strong> this study is to identify CTCs in CRPC<br />

and BC patients using a novel mesenchymal-based capture method.<br />

Methods: In patients with advanced CRPC and BC, two EDTA and one<br />

CellSave tube <strong>of</strong> blood are collected. Using the CellSearch system (Veridex,<br />

USA), CTCs are captured with either anti-N-cadherin (N-cad) or anti-Ocadherin<br />

(O-cad) ferr<strong>of</strong>luid and detected cells (events) are defined as<br />

beta-catenin and DAPI positive, CD45 negative intact cells. We evaluated<br />

the performance <strong>of</strong> these EMT-based ferr<strong>of</strong>luids in healthy volunteers,<br />

control cells, and in a pilot study <strong>of</strong> patients with CRPC, and compared<br />

enumeration <strong>of</strong> cells using novel vs. standard EpCAM-based capture<br />

methods. Results: In healthy volunteers, rare events were detected using<br />

the novel capture methods. In CRPC patients, O-cad capture detected more<br />

events in 3 <strong>of</strong> 5 subjects than EpCAM-based capture, and the majority <strong>of</strong><br />

captured cells were cytokeratin negative. See table below. Conclusions:<br />

These preliminary results suggest that a novel CTC phenotype exists in<br />

CRPC based on EMT properties, particularly overexpression <strong>of</strong> O-cadherin.<br />

Further characterization <strong>of</strong> these cells from patients with advanced PC, BC,<br />

and other solid tumors will provide insight into EMT and metastasis biology.<br />

O-cad capture, beta-catenin� N-cad capture, beta-catenin� EpCAM capture, cytokeratin�<br />

Healthy volunteers 0-51 events (mean 5.95) n�21 0-4 events (mean 0.28) n�25 NA<br />

CRPC patients 0-465 events (mean 138.4) n�5 0-2 events (mean 1.2) n�5 1-123 CTCs (mean 50.8) n�5<br />

10535 General Poster Session (Board #41H), Mon, 1:15 PM-5:15 PM<br />

Pr<strong>of</strong>iling <strong>of</strong> circulating tumor cells isolated from 105 metastatic gastric<br />

cancer patients revealed HER2 overexpression/activation for potential use<br />

in clinical setting. Presenting Author: Saswati Hazra, Prometheus Laboratories<br />

Inc., San Diego, CA<br />

Background: Gastric cancer (GCA) is the second leading cause <strong>of</strong> cancer<br />

mortality in the world. Survival <strong>of</strong> patients with advanced GCA treated with<br />

chemotherapy remains low. New targeted therapies are urgently needed.<br />

There is mounting evidence <strong>of</strong> the role <strong>of</strong> HER2 overexpression in patients<br />

with GCA, and it has been highly correlated to poor outcomes with more<br />

aggressive disease. The ability to accurately determine HER2 status by<br />

testing circulating tumor cells (CTCs) may improve patient treatment by<br />

allowing ongoing assessment <strong>of</strong> HER2 status during treatment and/or<br />

identifying additional patients who could potentially benefit from HER2targeted<br />

therapy. Methods: The Collaborative Enzyme Enhanced Reactiveimmunoassay<br />

(CEER) was utilized to determine the expression and activation<br />

(phosphorylation) levels <strong>of</strong> HER2 in CTCs isolated from blood specimens<br />

obtained from 105 metastatic GCA patients. Results: Utilizing the CEER<br />

platform, the levels <strong>of</strong> HER2 expression and phosphorylation were determined<br />

for CTCs isolated from metastatic GCA patients. Evaluable CTCs<br />

were found in 33% (35/105) <strong>of</strong> enrolled patients. Out <strong>of</strong> 35 patients, 7<br />

patients (20%) have high HER2 over expression, 6 patients (17%) have<br />

moderate HER2 expression and 11 patients (31%) have HER2 activation<br />

(phospho positive) with no HER2 over-expression. Conclusions: When CTCs<br />

were present, the CEER assay identified varying levels <strong>of</strong> HER2 involvements<br />

in 68% <strong>of</strong> metastatic GCA patients. HER2 positive CTCs could serve<br />

as a prognostic and/or predictive marker in patients with advanced GCA and<br />

CTC-HER2 pr<strong>of</strong>ile shifts can be utilized to monitor the treatment efficacy.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


10536 General Poster Session (Board #42A), Mon, 1:15 PM-5:15 PM<br />

In vivo isolation <strong>of</strong> circulating tumor cells in non-small cell lung cancer<br />

(NSCLC) patients by a medical device. Presenting Author: Lukasz Gasiorowski,<br />

Medical University Poznan, Poznan, Poland<br />

Background: Currently, circulating tumor cells (CTCs) are isolated in vitro<br />

from small limited volumes <strong>of</strong> blood samples. Furthermore, CTC results for<br />

NSCLC as a prognostic and stratification biomarker are scarce. The aim <strong>of</strong><br />

the study was to assess a functionalized and structured medical wire<br />

(FSMW) for in vivo isolation <strong>of</strong> CTCs directly from the blood <strong>of</strong> NSCLC<br />

patients, and to compare it to the Cell Search method. Methods: The device<br />

was inserted in a cubital vein through a standard cannula for thirty minutes.<br />

The interaction <strong>of</strong> target CTCs with the FSMW was mediated by an antibody<br />

directed against the epithelial cell adhesion molecule (EpCAM). To confirm<br />

the CTCs binding to the wire, the immunohistochemical staining against<br />

EpCAM as well as CD45 for negative cell selection was performed. There<br />

were 72 applications <strong>of</strong> the wire in 48 stage I-IIIB NSCLC patients (12<br />

double applications) and 12 non cancer patients. Enumeration data was<br />

available for 34 NSCLC patients and 8 non cancer patients. For 34<br />

patients, samples were also tested in the Cell Search system. Results: The<br />

device was well tolerated in all 72 applications without side effects. We<br />

obtained in vivo isolation <strong>of</strong> CTCs in 32 <strong>of</strong> 34 NSCLC patients (94.1%) with<br />

a median (range) <strong>of</strong> 13 (0-300) CTCs. The sensitivity was similar for early<br />

and late stage NSCLC patients. In the non-cancer patients, no CTCs were<br />

detected (100 % specificity). In 2 <strong>of</strong> 34 samples (5.8%), CTCs were<br />

detected by the Cell Search method. In all matched pairs, the FSMW<br />

detected the same number or more CTCs compared to the Cell Search<br />

method. Conclusions: Whilst well tolerated without side effects, the CTC<br />

detection rate <strong>of</strong> the FSMW in NSCLC patients was �90%. In contrast,<br />

�10% detection was obtained using the Cell Search analysis. No CTCs<br />

were detected in non-cancer patients. This pro<strong>of</strong> <strong>of</strong> concept study may have<br />

important clinical implications, as the implementation <strong>of</strong> the device into<br />

clinical practice may improve early detection, prognosis and therapy<br />

monitoring <strong>of</strong> NSCLC patients. The method may also allow the molecular<br />

analysis <strong>of</strong> the CTCs, with the possibility <strong>of</strong> establishing more personalized<br />

treatment regiments.<br />

10538 General Poster Session (Board #42C), Mon, 1:15 PM-5:15 PM<br />

Prognostic and diagnostic value <strong>of</strong> Ewing family <strong>of</strong> tumors (EFTs)associated<br />

fusion transcripts detected in peripheral blood specimens.<br />

Presenting Author: Joanna Przybyl, Department <strong>of</strong> Molecular Biology,<br />

Maria Sklodowska-Curie Memorial Cancer Center and Institute <strong>of</strong> Oncology,<br />

Warsaw, Poland<br />

Background: EFTs are characterized by chromosomal translocations leading<br />

to formation <strong>of</strong> oncogenic EWSR1-FLI1 fusion gene in 85-90% <strong>of</strong> cases.<br />

The aim <strong>of</strong> the study was to detect circulating tumor cells (CTCs) carrying<br />

EWSR1-FLI1 fusion transcript in peripheral blood and assess their added<br />

value to standard diagnostic procedures and utility as a prognostic marker<br />

in EFTs. Methods: 10mL <strong>of</strong> whole blood was collected from 35 untreated<br />

adult EFTs patients at the diagnosis (period: 2008-2011, median age 27<br />

years) and 13 healthy controls. 13 patients presented metastatic disease<br />

(M1) at the diagnosis. Nested RT-PCR was applied in triplicate for the<br />

detection <strong>of</strong> EWSR1-FLI1 transcript. Blood specimen was regarded CTCpositive<br />

when at least 2 out <strong>of</strong> 3 nested RT-PCR assays were positive and<br />

results were confirmed by sequencing. FISH assay for EWSR1 rearrangement<br />

was performed on FFPE tumor tissue in 28 available cases. Median<br />

follow-up was 16 months. Results: EWSR1-FLI1 transcript was detected in<br />

peripheral blood <strong>of</strong> 71.4% (n�25) <strong>of</strong> patients. FISH assay was positive for<br />

EWRS1 rearrangement in 58% <strong>of</strong> cases. In 10 patients, where FISH assay<br />

could not be performed due to insufficient quality or lack <strong>of</strong> material,<br />

nested RT-PCR provided confirmation <strong>of</strong> immunopathological diagnosis.<br />

Specificity <strong>of</strong> RT-PCR blood test in healthy control was 91.4% (12/13<br />

negative; p�0.0001). Median overall survival (OS) was 18 months.<br />

CTC-positive patients showed the trend for longer OS than CTC-negative<br />

patients (1-year OS: 89% vs. 58%; p�0.07), but longer follow-up is<br />

needed. Conclusions: Our results show that the fusion transcript detection<br />

in peripheral blood specimens may be a useful additional test to the<br />

standard clinicopathological diagnosis <strong>of</strong> EFTs. High detection rate <strong>of</strong><br />

EWSR1-FLI1 transcript in peripheral blood <strong>of</strong> EFTs patients at the<br />

diagnosis may imply EFTs as a systemic disease with clinically evident<br />

metastases or micrometastases at presentation. Prognostic significance <strong>of</strong><br />

CTCs in EFTs warrants further studies.<br />

Tumor Biology<br />

665s<br />

10537 General Poster Session (Board #42B), Mon, 1:15 PM-5:15 PM<br />

A new medical device for in vivo capturing <strong>of</strong> circulating tumor cells in<br />

breast cancer (BC) patients. Presenting Author: Dawid Murawa, Wielkopolska<br />

Cancer Centre, Poznan, Poland<br />

Background: In BC, the number <strong>of</strong> circulating tumor cells (CTCs) is<br />

discussed as a prognostic and stratification biomarker, and could also<br />

reflect treatment efficacy. Currently, CTCs are isolated ex vivo from a small<br />

volume <strong>of</strong> blood. Results from a larger volume <strong>of</strong> blood are scarce. The aim<br />

<strong>of</strong> the study was to assess a functionalized and structured medical wire<br />

(FSMW) for in vivo capturing <strong>of</strong> CTCs directly from the blood stream <strong>of</strong> BC<br />

patients. Methods: The device was inserted in a cubital vein through a<br />

standard cannula for thirty minutes. The interaction <strong>of</strong> target CTCs with the<br />

FSMW was mediated by antibodies directed against the epithelial cell<br />

adhesion molecule (EpCAM). To confirm binding <strong>of</strong> CTCs to the wire, the<br />

immunohistochemical positive staining against EpCAM as well as negative<br />

staining for CD45 was performed. There were 54 applications <strong>of</strong> the wire in<br />

42 stage I-IV BC patients (12 double applications). Enumeration data from<br />

37 BC patients with 49 applications (5 failed subsequent analyses) were<br />

assessed. CTC counts on 23 devices were directly compared to counts by<br />

CellSearch. Results: The device was well tolerated in all 54 applications<br />

without side effects. We obtained in vivo isolation <strong>of</strong> CTCs in 44 <strong>of</strong> 49<br />

applications to BC patients (89.7 %). The sensitivity was similar for early<br />

and late stage BC patients. The median (range) <strong>of</strong> isolated EpCAM-positive<br />

CTCs was 5 (0-515). The CellSearch method reached a sensitivity <strong>of</strong><br />

18.5%. In all paired samples the number <strong>of</strong> CTCs detected with the FSMW<br />

was higher or equal to CellSearch, regardless <strong>of</strong> the disease stage. Linear<br />

regression <strong>of</strong> the data <strong>of</strong> the double application <strong>of</strong> the FSMW showed a very<br />

good concordance (r2 � 0.97, p�0.0001). Conclusions: Whilst well<br />

tolerated without side effects, the CTC detection rate <strong>of</strong> the FSMW in BC<br />

patients was nearly 90 %. CTC detection was obtained in 18.5% by the<br />

CellSearch. Double application <strong>of</strong> FSMWs in the same patient indicates<br />

ample precision. This pro<strong>of</strong> <strong>of</strong> concept study may have important clinical<br />

implications, as the device may improve early detection, prognosis and<br />

therapy monitoring <strong>of</strong> BC patients. The molecular analysis <strong>of</strong> the captured<br />

CTCs could become a breakthrough in personalized medicine.<br />

10539 General Poster Session (Board #42D), Mon, 1:15 PM-5:15 PM<br />

Circulating melanoma cells isolated from clinical blood samples and<br />

characterized by full-length mRNA sequencing at single-cell level. Presenting<br />

Author: Yu-Chieh Wang, Department <strong>of</strong> Chemical Physiology, The<br />

Scripps Research Institute, La Jolla, CA<br />

Background: Melanoma is the most aggressive type <strong>of</strong> skin cancer. Latestage<br />

melanoma is highly metastatic and currently lacks effective treatment.<br />

This discouraging clinical observation highlights the need for a better<br />

understanding <strong>of</strong> the molecular mechanisms underlying melanoma initiation<br />

and progression and for developing new therapeutic approaches based<br />

on novel targets. Although genome-wide transcriptome analyses have been<br />

frequently used to study molecular alterations in clinical samples, it has<br />

been technically challenging to obtain the transcriptomic pr<strong>of</strong>iles at<br />

single-cell level. Methods: Using antibody-mediated magnetic activated<br />

cell separation (MACS), we isolated and individualized putative circulating<br />

melanoma cells (CMCs) from the blood samples <strong>of</strong> the melanoma patients<br />

at advance stages. The transcriptomic analysis based on a novel and robust<br />

mRNA-Seq protocol (Smart-Seq) was established and applied to the<br />

putative CMCs for single-cell pr<strong>of</strong>iling. Results: We have discovered distinct<br />

gene expression patterns, including new putative markers for CMCs.<br />

Meanwhile, the gene expression pr<strong>of</strong>iles derived <strong>of</strong> the CMC candidates<br />

isolated from the patient’s blood samples are closely-related to the<br />

expression pr<strong>of</strong>iles <strong>of</strong> other cells originated from human melanocytes,<br />

including normal melanocytes in primary culture and melanoma cell lines.<br />

Compared with existing methods, Smart-Seq has improved read coverage<br />

across transcripts, which provides advantage for better analyzing transcript<br />

is<strong>of</strong>orms and SNPs. Conclusions: Our results suggest that the techniques<br />

developed in this research for cell isolation and transcriptomic analyses can<br />

potentially be used for addressing many biological and clinical questions<br />

requiring genomewide transcriptome pr<strong>of</strong>iling in rare cells.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


666s Tumor Biology<br />

10540 General Poster Session (Board #42E), Mon, 1:15 PM-5:15 PM<br />

HER2 expression on circulating tumor cells (CTC) in patients with early<br />

HER2-positive breast cancer: Results <strong>of</strong> the German SUCCESS B trial.<br />

Presenting Author: Bernadette Anna Sophia Jaeger, Department <strong>of</strong> Gynecology<br />

and Obstetrics, Ludwig-Maximilians-University, Munich, Germany<br />

Background: Recent reports showed a discrepancy in the Her2-status <strong>of</strong><br />

metastases or minimal residual disease in blood and bone marrow compared<br />

to the primary tumor in patients with breast cancer. The Her2-status<br />

<strong>of</strong> CTC was prospectively evaluated in the German multicenter SUCCESS B<br />

study. Methods: The SUCCESS B trial is a randomized Phase III study<br />

comparing FEC-Docetaxel (Doc) vs. FEC-Docetaxel-Gemcitabine (Doc-G)<br />

as well as Her2 targeted therapy with Trastuzumab as adjuvant treatment in<br />

patients with early, Her2 positive, node positive or high risk node negative<br />

primary breast cancer. As part <strong>of</strong> the translational research program, 23ml<br />

<strong>of</strong> peripheral blood were drawn before adjuvant chemotherapy. In 638<br />

samples CTC and Her2-status were assessed using the CellSearch System<br />

(Veridex, USA). After immunomagnetic enrichment with an anti-Epcamantibody,<br />

cells were labeled with anti-CK8/18/19, anti-CD45 antibodies as<br />

well as a fluorescein conjugate antibody for Her2 phenotyping. Cut<strong>of</strong>f for<br />

CTC-positivity was �1 CTC and for Her2 �1 CTC with strong Her2-staining<br />

(���). Results: 40.2% <strong>of</strong> pts (n�257) were positive for CTC (mean 4.52;<br />

range 0-1689). The number <strong>of</strong> detected CTC was distributed as follows: 1<br />

CTC (n�112; 43.6%), 2 CTC (n�65; 25.3%), 3 CTC (n�36; 14.0%), 4<br />

CTC (n�12; 4.7%) and �5 CTC (n�31; 12.1%). Her2 status on CTC was<br />

negative or weak in 12.5% (n�32) and 8.9% (n�23) <strong>of</strong> CTC positive<br />

patients respectively and therefore categorized as Her2 negative. In 21.4%<br />

(n�55) we detected moderate and in 57.2% (n�147) strong Her2staining<br />

<strong>of</strong> �1 CTC per sample. Therefore 57.2% <strong>of</strong> CTC-positive samples<br />

were diagnosed as Her2 positive and 21.4% as questionable. No association<br />

was found between the detection <strong>of</strong> CTC or the Her2-status on CTC<br />

with tumor size, histopathological grading, hormone receptor status or<br />

axillary lymph node involvement. Conclusions: The data <strong>of</strong> this trial<br />

confirms the frequent discordance <strong>of</strong> Her2 expression on CTC compared to<br />

the primary tumor. Survival data within the Success B trial will give further<br />

insight into the tumor biology <strong>of</strong> Her2 positive disease and the role <strong>of</strong> the<br />

Her2-status on CTC to predict treatment efficacy.<br />

10542 General Poster Session (Board #42G), Mon, 1:15 PM-5:15 PM<br />

Serum hepatocyte growth factor and interleukin-6 as prognostic markers<br />

for stage III non-small cell lung cancer. Presenting Author: Hideki Ujiie,<br />

Department <strong>of</strong> Thoracic Surgery, Saitama Cancer Center, Saitama, Japan<br />

Background: We surveyed prognostic biomarkers for resectable non-small<br />

cell lung cancer (NSCLC). Methods: We obtained preoperative serum from<br />

109 patients admitted to our facility, and measured the levels <strong>of</strong> hepatocyte<br />

growth factor (HGF), interleukin-6 (IL-6), and nicotinamide<br />

N–methytransferase (NNMT) in the sera by the ELISA method. We also<br />

examined the clinical backgrounds <strong>of</strong> the patients. Results: The median<br />

HGF and IL-6 contents in sera from 109 patients were 860 pg/ml and 2.7<br />

pg/ml, respectively. Analysis <strong>of</strong> survival curves indicated that HGF or IL-6<br />

levels higher than the median were associated with poor overall survival<br />

(HGF, P � 0.019; IL-6, P � 0.002). On the other hand, carcinoembryonic<br />

antigen (CEA), a known tumor marker, and NNMT, which we found to be a<br />

candidate tumor marker, exhibited no correlation with the prognosis. The<br />

tumor status (pT factor) and stage were strong prognostic indicators (P �<br />

0.001). In addition, we analyzed stage III lung cancer alone. Higher HGF or<br />

IL-6 levels were associated with poor overall survival (HGF, P � 0.016;<br />

IL-6, P � 0.013). Disease-free survival was also affected by these cytokine<br />

contents, but not statistically significantly. The prognosis <strong>of</strong> patients with<br />

adenocarcinomas (ADC) was better than that <strong>of</strong> patients with other<br />

histological types. However, the pT factor and nodal status (pN factor) were<br />

not associated with the survival <strong>of</strong> stage III patients. Conclusions: The levels<br />

<strong>of</strong> HGF and IL-6 in serum could be useful prognostic indicator <strong>of</strong> the<br />

survival <strong>of</strong> stage III NSCLC patients undergoing surgery and chemotherapy.<br />

10541 General Poster Session (Board #42F), Mon, 1:15 PM-5:15 PM<br />

Detection and HER2 expression <strong>of</strong> circulating tumor cells in advanced<br />

gastric cancer patients. Presenting Author: Satoshi Matsusaka, Department<br />

<strong>of</strong> Gastroenterology, Gastroenterology Center, Cancer Institute Hospital;<br />

<strong>Clinical</strong> Chemotherapy, Cancer Chemotherapy Center, Japanese<br />

Foundation for Cancer Research, Tokyo, Japan<br />

Background: This study was aimed at detecting and HER2-expressiong<br />

circulating tumor cells (CTC) in peripheral blood <strong>of</strong> chemo-naïve or<br />

chemo-resistant patients with advanced gastric cancer. Methods: All<br />

patients were enrolled using institutional review board-approved protocols<br />

at the Cancer Institute Hospital in the Japanese Foundation for Cancer<br />

Research and provided informed consent. The study population consisted<br />

<strong>of</strong> patients <strong>of</strong> age 18 years or older with histologically proven advanced<br />

gastric cancer. A total <strong>of</strong> 140 patients with advanced gastric cancer were<br />

enrolled into a prospective study. We used CellSearch system for detection<br />

and HER2 expression for CTC. Results: We detected �1 CTC/7.5ml in 80 <strong>of</strong><br />

140 patients (57.1%). HER2-positive CTC were observed in 19 <strong>of</strong> 80<br />

CTC-positive patients (23.8%), including 6 patients with HSR2-negative<br />

primary tumors. 21 patients with HER2-positive primary tumors administered<br />

trastuzumab in combination with chemotherapy (Xeloda�cisplatin;<br />

15, weekly Paclitaxel; 5). We detected �1 CTC/7.5ml at the baseline in 11<br />

<strong>of</strong> 20 patients with trastuzumab treatment. HER2-positive CTCs were<br />

observed in 6 <strong>of</strong> 11 CTC-positive patients (54.5%). Patients with HER2positive<br />

CTC at the baseline were shorter PFS than those with HER2negative<br />

CTC at the baseline. Conclusions: HER2 expression on CTC was<br />

associated with chemo-resistance. Information on the HER2 status <strong>of</strong> CTC<br />

might be helpful for stratification <strong>of</strong> HER2-directed therapies.<br />

10543 General Poster Session (Board #42H), Mon, 1:15 PM-5:15 PM<br />

Promoter methylation <strong>of</strong> MGMT in paired primary and recurrent glioblastomas.<br />

Presenting Author: Li Bie, The First Hospital <strong>of</strong> Jilin Universtiy,<br />

Changchun, China<br />

Background: Epigenetic sliencing <strong>of</strong> MGMT gene promoter in primary<br />

glioblastomas (GBM) <strong>of</strong> pts subsequently treated with temozolomide (TMZ)<br />

is associated with prolonged survival. Further, several studies have observed<br />

a change in MGMT silencing in paired primary and recurrent GBM.<br />

However, the relationship between this “MGMT switch” and patients<br />

outcome is largely unknown. Methods: The study involved primary and<br />

recurrent tumor tissue samples from 53 GBM pts diagnosed and treated<br />

within the First Hospital <strong>of</strong> Jilin University from Jan 2003 to Nov 2010.<br />

After surgical treatment, all pts were subjected to radiotherapy with<br />

concomitant administration <strong>of</strong> TMZ (75 mg/m2 /d), followed by 5 cycles <strong>of</strong><br />

adjuvant TMZ given at 4-weeks intervals. Pts that experienced recurrent<br />

tumors received TMZ at a dose <strong>of</strong> 200 mg/m2 /5 days for 4 weeks. 53 pts<br />

underwent 58 further operations after recurrence (5 pts received a third<br />

surgery). MGMT promoter methylation levels were determined using qMSP.<br />

The relationship between “MGMT switch” and clinical outcome was<br />

investigated. Results: Fifty three (M/F�33/20; median age: 49.2�3.6,<br />

range: 19.5-72.3 ys) underwent a first operation for GBM. qMSP analysis<br />

revealed MGMT promoter methylation in 19 pts (35.8%, group A, OS 31.5<br />

ms); no methylation in 34 pts (64.2%, group B, OS 7.9 ms). In the<br />

recurrent tumors, MGMT promoter methylation was detected in 25 pts<br />

(47.2%); and no methylation in 28 pts (52.8%). Comparison <strong>of</strong> individual<br />

pairs <strong>of</strong> primary and recurrent GBMs revealed a changed methylation status<br />

in 10 (18.9%), including eight changed from unmethylated to methylated<br />

tumors (15.1%, group C, OS 29.4 ms), two changed from methylated to<br />

unmethylated tumors (3.8%, group D, OS 10.5 ms). Median overall<br />

survival was 12.1 ms. MGMT promoter methylation was significantly<br />

associated with a favorable clinical outcome (group A vs group B,<br />

p�0.0027). The outcome <strong>of</strong> pts were not significant different between A<br />

and C (p�0.01). Conclusions: The methylation status <strong>of</strong> the MGMT<br />

promoter was altered in 10 (18.9%) <strong>of</strong> 53 recurrent GBM after chemoradiotherapy.<br />

Eight pts changed from unmethylated to methylated who have a<br />

median overall survival similar to methylated pts. The pts changed from<br />

methylated to unmethylated who have poor outcome.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


10544 General Poster Session (Board #43A), Mon, 1:15 PM-5:15 PM<br />

DJ-1 as a predictor <strong>of</strong> pathologic complete remission <strong>of</strong> neoadjuvant<br />

chemotherapy with breast cancer patients. Presenting Author: Takahiko<br />

Kawate, Basic Pathology, National Defence Medical Colleage, Saitamaken,<br />

Japan<br />

Background: DJ-1 is a multifunctional protein which is encoded by the<br />

causative gene <strong>of</strong> familial Parkinson disease (i.e., PARK7), and is associated<br />

with carcinogenesis. DJ-1 is also a candidate marker for the presence<br />

<strong>of</strong> breast cancer cells, in which it is secreted in nipple fluid and serum. We<br />

previously reported that a DJ-1 expression pattern that is low at the protein<br />

level and high at the mRNA level is associated with DJ-1 secretion. This<br />

secretory expression pattern correlated with a negative hormone receptor<br />

status and with unfavorable clinical outcome in breast cancer patients.<br />

However, the association <strong>of</strong> the DJ-1 secretion pattern with therapeutic<br />

effect has yet to be determined. Methods: A total <strong>of</strong> 299 patients received<br />

neoadjuvant chemotherapy and surgery from 2002 to 2010 at our<br />

institution. We performed immunohistochemistry and in-situ hybridization<br />

<strong>of</strong> DJ-1 in both the needle biopsy and operative specimens <strong>of</strong> 147 cases<br />

with a follow-up time <strong>of</strong> 3.1 years. The average degree <strong>of</strong> staining intensity<br />

was evaluated semi-quantitatively using an image analyzer. Univariate and<br />

multivariate analyses were used to evaluate the predictive value <strong>of</strong> the<br />

therapeutic effect <strong>of</strong> neoadjuvant chemotherapy. Results: A low expression<br />

<strong>of</strong> DJ-1 protein was detected in the needle biopsy and operative specimens<br />

<strong>of</strong> 89 <strong>of</strong> the 147 cases, regardless <strong>of</strong> a high or maintained mRNA level. The<br />

tumor response was evaluated as pathological complete remission (pCR) in<br />

39 cases and as non-pCR in the remaining 108 cases. A low expression <strong>of</strong><br />

DJ-1 protein, which was detected in 34 (87.2%) pCR cases, was a<br />

significant predictor on univariate analysis (P �0.001). On multivariate<br />

analysis, a low expression <strong>of</strong> DJ-1 was shown to be a significant independent<br />

predictor similar to established predictors such as estrogen receptor<br />

status and human epidermal growth factor receptor 2 score. Conclusions: A<br />

low expression <strong>of</strong> DJ-1 protein can serve as a novel and important predictor<br />

<strong>of</strong> a neoadjuvant chemotherapeutic effect, as well as a promising indicator<br />

for serologic diagnosis in breast cancer patients.<br />

10546 General Poster Session (Board #43C), Mon, 1:15 PM-5:15 PM<br />

Evaluation <strong>of</strong> CYP3A5, VEGF-a, and VEGFR2 polymorphisms as markers <strong>of</strong><br />

sunitinib toxicity. Presenting Author: Cristina Rodriguez de Antona, Spanish<br />

National Cancer Research Center, Madrid, Spain<br />

Background: Sunitinib (Sun) is a multitargeted tyrosine-kinase inhibitor<br />

approved for the treatment <strong>of</strong> renal cell carcinoma (RCC), GIST and<br />

pancreatic neuroendocrine tumors (pNETs). It is known that a polymorphism<br />

in Sun metabolizing gene CYP3A5, was associated with an increased<br />

risk <strong>of</strong> dose reductions due to toxicity (tox). Additionally, polymorphisms in<br />

VEGF-A and VEGFR2 were suggested to increase the risk <strong>of</strong> hypertension<br />

(HT) during treatment (García-Donas, et al. Lancet Oncol 2011). These<br />

data have not been validated so far. We aim to assess the value <strong>of</strong> these<br />

polymorphisms as markers <strong>of</strong> tox using a wide range <strong>of</strong> solid tumors treated<br />

with Sun. Methods: In this non-interventional and retrospective study, DNA<br />

was collected from 28 patients with different pathologies (16 pNETs, 5<br />

medullary thyroid carcinomas, 2 NETs <strong>of</strong> the lung, 2 undifferentiated<br />

follicular carcinomas, 1 undifferentiated papillar carcinoma, 1 GIST, and 1<br />

carcinoid <strong>of</strong> the rectum) treated with Sun in a daily practice setting.<br />

Genotyping for CYP3A5*1 allele (rs776746), VEGF-A -2578C�A<br />

(rs699947) and VEGFR2 Q472H (rs1870377) was performed. Associations<br />

between the genotypes and Sun dose reductions and HT were<br />

performed using univariable analyses. Results: In agreement with previous<br />

reports, we found that VEGF-A rs699947 conferred a statistically significant<br />

increased risk <strong>of</strong> developing HT during treatment (HR�9.8,<br />

95%CI�1.2-82.0, P�0.034). CYP3A5*1 high metabolizing allele showed<br />

a trend towards the increase in the risk <strong>of</strong> Sun dose reductions due to tox<br />

(HR�2.4, 95%CI�0.8-6.8, P�0.11) with a median time to dose reduction<br />

<strong>of</strong> 7.0 months for wild type homozgygous patients and 4.6 months for<br />

heterozygous patients. A trend was also found on the relation between<br />

VEGF-A rs699947 A/A patients (HR�3.8, 95%CI�0.8-16.8, P�0.080)<br />

and dose reduction risk. No significant associations were found for VEGFR2<br />

rs1870377. Conclusions: The present study suggests that VEGF-A rs699947<br />

is a risk factor for Sun HT and has a role in other tox leading to dose<br />

reductions. Similarly, CYP3A5*1 shows a trend towards an increased risk<br />

<strong>of</strong> Sun dose reductions. If confirmed, these markers could be used to<br />

identify a subset <strong>of</strong> patients with an increased risk <strong>of</strong> Sun toxicity.<br />

Tumor Biology<br />

667s<br />

10545 General Poster Session (Board #43B), Mon, 1:15 PM-5:15 PM<br />

Correlation between miRNA (miR) and gene expression pr<strong>of</strong>iles (GEP) and<br />

response to neoadjuvant chemotherapy (NT) in patients with locally<br />

advanced and inflammatory breast cancer (BC). Presenting Author: Sierra<br />

Mi Li, City <strong>of</strong> Hope, Duarte, CA<br />

Background: GEP may predict for pathologic complete response (pCR).<br />

Correlation between miRs, GEP, and pCR may reveal novel targets.<br />

Methods: Patients (pts) with HER2- BC were randomized to receive<br />

docetaxel, doxorubicin, cyclophosphamide (TAC, arm A) or A and C given<br />

every 2 weeks x 4, followed by carboplatin and nab-paclitaxel (arm B). Pts<br />

with HER2� BC received NT per arm B, with trastuzumab added (arm C).<br />

Core biopsies were snap-frozen prior to NT, and RNA extracted for gene<br />

array analysis (Agilent 44K microarray) and deep sequence miRNA analysis<br />

(Solexa/Illumina platform). HER2 testing by IHC [3�] or FISH and/or gene<br />

array was carried out (Blueprint). MiRs were measured in 72 <strong>of</strong> 119<br />

enrolled pts. Of 882 miRNAs reported by Solexa sequencing, 487 were<br />

assessed (observed at raw counts � 10 in at least 3 pts). TargetScan was<br />

used to obtain miRNA targets and negative correlation between miR and<br />

mRNA expression was applied. Results: In 44 HER2- cases, we found 17<br />

miRs differentially expressed between pts who achieved pCR vs. pts who<br />

did not (� pCR), 13 with predicted targets by TargetScan (TS). We found<br />

28 canonical pathways (including Embryonic Cell, EGF signaling, Estrogendependent<br />

signaling) affected by the identified miRs, and inclusive <strong>of</strong> 74<br />

target mRNAs identified (Agilent) and correlating with pCR. The top<br />

overexpressed miRs are 31 [gene targets: FZD4]; 18a [IGF1, ALDH5A1];<br />

19a [PIK3R, IGF1]) and 3 lowest expressors are miR-190b [MMP1]; 29c<br />

[FOS, WNT5B]; 342-3p [BMP7]. Among the 28 HER2� cases there were<br />

14 miRNAs differentially expressed between BCs with pCR vs �pCR, and 8<br />

had predicted targets from TS. We found 10 canonical pathways (including<br />

p53, HER-2, PTEN) affected by miRs, which included 44 target mRNAs<br />

(Agilent) correlating with pCR. The most overexpressed miRs are miR-708<br />

[gene target: RRM2B; 193a-p5 [BMPR1B, RPRM] ; 92b [CCNE2, DKK2].<br />

The lowest expressors are: miR-30a [FZD1, SMAD1, TP63]; miR-450-5p<br />

[FOXO1, WINT5A, ERBB2]; miR-497 [PIK3R1, FGFR1, FOXO1].<br />

Conclusions: Combined assessment <strong>of</strong> miRNA and gene expression patterns<br />

is feasible, and may yield information leading to individualized and<br />

improved NT.<br />

10547 General Poster Session (Board #43D), Mon, 1:15 PM-5:15 PM<br />

Pharmacogenetic predictors <strong>of</strong> adverse events in stage II-III colon cancer<br />

(CC) patients treated with oxaliplatin and fluoropyrimidines-based adjuvant<br />

chemotherapy (CT): A GEMCAD study. Presenting Author: Ana Custodio,<br />

Department <strong>of</strong> Medical Oncology, La Paz University Hospital, Madrid,<br />

Spain<br />

Background: Although the benefit <strong>of</strong> oxaliplatin-based adjuvant CT has been<br />

demonstrated in patients with resected stage II-III CC, the recommendation<br />

to administer postoperative treatment must consider its potential<br />

adverse events. Predicting individual patients´risk <strong>of</strong> severe toxicity could<br />

potentially improve the quality <strong>of</strong> care by allowing individualization <strong>of</strong><br />

treatment. We investigate the utility <strong>of</strong> determining single nucleotide<br />

polymorphisms (SNPs) in genes involved in oxaliplatin and fluoropyrimidines<br />

metabolisms to predict the toxicity <strong>of</strong> adjuvant CT in stage II-III CC<br />

patients. Methods: DNA was extracted from formalin-fixed paraffinembedded<br />

samples from 379 surgically treated high-risk stage II (27.71%)<br />

and stage III (72.29%) CC patients receiving adjuvant CT (54.35%<br />

FOLFOX, 45.65% XELOX) from January 2004 to December 2008. Genotyping<br />

was performed for 35 SNP in 18 genes using the MassARRAY<br />

(SEQUENOM) technology. Results: A total <strong>of</strong> 89 (23.4%) patients experienced<br />

at least one grade 3-4 adverse event. The most common grade 3-4<br />

toxicities were neutropenia (15.3%), diarrhea (8.17%) and neurotoxicity<br />

(7.65%). The MTHFR rs1801133 C�T C/C (30.1% C/C, 17.5% C/T,<br />

21.7% T/T; p�0.043) and XRCC2 rs3218408 T�G T/T (28.3% T/T,<br />

16.2% G/T and 10% G/G; p�0.007) genotypes were associated with<br />

higher risk <strong>of</strong> any grade 3-4 toxicities, whereas the incidence <strong>of</strong> severe<br />

toxicity was lower in patients with the UMPS rs3772807 G�C C/C (14%<br />

C/C, 21% C/G, 28.9% G/G; p�0.021) and DPYD rs970337 G�A A/A<br />

(16.7% A/A, 21.1% A/G, 30.1% G/G; p�0.028) genotypes. In addition,<br />

the DPYD rs970337 G�A A/A genotype was associated with a lower rate <strong>of</strong><br />

grade 3-4 diarrhea (5% A/A, 9.4% A/G, 8.1% G/G; p�0.030), the ABCG2<br />

rs3114018 A�C C/C genotype with an increased rate <strong>of</strong> grade 3-4<br />

neutropenia (15.2% C/C, 6.3% A/C, 4.5% A/A; p�0.034) and the ERCC1<br />

rs11615 T�C T/T genotype with a lower rate <strong>of</strong> grade 3-4 neurotoxicity<br />

(5.4% T/T, 9.1% C/T, 10.2% C/C; p�0.032). Conclusions: Our data<br />

suggest that SNPs in genes involved in oxaliplatin and fluoropyrimidines<br />

metabolisms can potentially predict severe toxicity in stage II-III CC<br />

patients treated with adjuvant CT.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


668s Tumor Biology<br />

10548 General Poster Session (Board #43E), Mon, 1:15 PM-5:15 PM<br />

Different EGFR/KRAS mutation spectrums in lung cancer between never<br />

smokers and heavy smokers. Presenting Author: Kazuya Takamochi,<br />

Juntendo University School <strong>of</strong> Medicine, Tokyo, Japan<br />

Background: EGFR mutations are more commonly found in lung adenocarcinomas<br />

in never smokers, while KRAS mutations are more frequently<br />

present in those in heavy smokers. Although the incidence is low, EGFR<br />

mutations are also found in tumors in heavy smokers and KRAS mutations<br />

also occur in tumors in never smokers. Therefore, there may be different<br />

biological characteristics in EGFR/ KRAS mutated lung cancers according<br />

to the smoking status. Methods: This was a retrospective review <strong>of</strong> 382<br />

patients with surgically resected lung cancers between February 2009 and<br />

March 2011. The clinicopathological factors (age, gender, histology, serum<br />

CEA level, pathological nodal status, lymphatic permeation, and blood<br />

vessel invasion) and EGFR/KRAS mutation spectrums were compared<br />

between never smokers (pack-year � 0, n � 161) and heavy smokers<br />

(pack-year � 20, n � 167). Results: EGFR mutations were detected in 88<br />

(55%) never smokers and in 33 (20%) heavy smokers. In contrast, K-ras<br />

mutations were detected in 7 (4%) never smokers and in 31 (19%) heavy<br />

smokers. Both EGFR and KRAS mutations were more frequently observed<br />

in female never smokers and in male heavy smokers. The spectrum <strong>of</strong> both<br />

EGFR and KRAS mutation differed according to the smoking status. Minor<br />

EGFR mutations other than exon 21 L858R and exon 19 deletions that are<br />

activating mutations indicating the efficacy <strong>of</strong> EGFR tyrosine kinase<br />

inhibitors (TKIs) were more frequently found in heavy smokers than in never<br />

smokers. G¡A transitions were more common KRAS mutations in never<br />

smokers and G¡T transversions that are thought to be related to exposure<br />

to the polycyclic aromatic hydrocarbons found in cigarette smoke were<br />

more common KRAS mutations in heavy smokers. Conclusions: The<br />

EGFR/KRAS mutation spectrums in lung cancer were quite different in<br />

never smokers and heavy smokers. Further study is needed to evaluate the<br />

relationship between the efficacy <strong>of</strong> such molecular targeting agents as<br />

EGFR TKIs and the EGFR/KRAS mutation spectrums.<br />

10551 General Poster Session (Board #43H), Mon, 1:15 PM-5:15 PM<br />

Prediction <strong>of</strong> late metastasis in node-negative breast cancer. Presenting<br />

Author: Marcus Schmidt, Department <strong>of</strong> Obstetrics and Gynecology,<br />

Johannes Gutenberg University, Mainz, Germany<br />

Background: Prediction <strong>of</strong> late metastasis is <strong>of</strong> clinical relevance in breast<br />

cancer. However, systematic genome-wide studies to identify genes associated<br />

with increased risk <strong>of</strong> metastasis 5 or more years after surgery are<br />

scarce. Methods: We examined the natural course <strong>of</strong> disease in three<br />

previously published cohorts (Mainz, Rotterdam, Transbig) including 766<br />

node-negative breast cancer patients with gene array data who did not<br />

receive systemic chemotherapy in the adjuvant setting. We established a<br />

Cox regression based method adjusted for multiple testing that identified<br />

genes predicting late metastasis (5 or more years after surgery). Only those<br />

genes were accepted that showed similar results in all three cohorts.<br />

Metastasis-free survival (MFS) was analyzed with univariate and multivariate<br />

Cox regression. Results: We identified 9 genes [ABCC5 (Hazard Ratio<br />

(HR) 2.19, p�0.003), EDDM3B (HR 3.58, p�0.044), RAD23B (HR<br />

0.37, p�0.001), XYLT2 (HR 2.19, p�0.027), DDX18 (HR 0.35,<br />

p�0.006), GPBP1L1 (HR 0.20, p�0.018), UBB (HR 9.73, p�0.025),<br />

RPS24 (HR 0.20, p�0.050), GPC1 (HR 2.36, p�0.013)] predicting late<br />

metastasis. These genes retained their independent prognostic significance<br />

after adjustment for established clinical factors (age, tumor size,<br />

grade, hormone receptor status, HER2) and biological motives like estrogen<br />

receptor, proliferation, B or T cells. These late-type genes are largely<br />

associated with resistance to hypoxia, apoptosis and DNA damage, suggesting<br />

that they might contribute to persistence <strong>of</strong> disseminated tumor cells.<br />

Conclusions: Genes associated with late metastasis <strong>of</strong>fer a perspective to<br />

identify breast cancer patients suitable for additional and prolonged<br />

therapies.<br />

10549 General Poster Session (Board #43F), Mon, 1:15 PM-5:15 PM<br />

Combined targeting <strong>of</strong> LSD1 (KDM1A) and histone deacetylases exerts<br />

superior efficacy against human AML. Presenting Author: Warren Fiskus,<br />

University <strong>of</strong> Kansas Medical Center, Kansas City, KS<br />

Background: LSD1 (KDM1A) is FAD-dependent histone H3K4Me2 demethylase.<br />

Inhibition <strong>of</strong> LSD1 increases H3K4Me3-a permissive mark for gene<br />

expression, and inhibits growth <strong>of</strong> pluripotent cancer cells. We have<br />

previously noted that treatment with the histone deacetylase (HDAC)<br />

inhibitor panobinostat (PS) depletes EZH2 (the catalytic subunit <strong>of</strong> the<br />

polycomb repressive complex 2, PRC2) and disrupts its interaction with the<br />

other PRC2 proteins, attenuates LSD1, and de-represses growth inhibitory<br />

and pro-apoptosis genes. Methods: In the present studies, we determined<br />

the chromatin effects and the pre-clinical in vitro and in vivo anti-AML<br />

activity <strong>of</strong> the novel, non-monoamine oxidase, reversible inhibitor <strong>of</strong> LSD1,<br />

HCI2509 (100 to 500 nM), alone and in combination with PS, utilizing<br />

cultured (OCI-AML3 and HL-60) and primary human AML blast progenitor<br />

cells. Results: Treatment with HCI2509 dose-dependently increased the<br />

levels <strong>of</strong> H3K4Me3, p16, p27, and CEBP� in cultured AML cells. This<br />

correlated with inhibition <strong>of</strong> cell growth and induction <strong>of</strong> morphologic<br />

differentiation in AML cells. Exposure to HCI2509 disrupted the binding <strong>of</strong><br />

LSD1 with the co-repressor CoREST and HDAC1. Treatment with PS (10 to<br />

50 nM) dose-dependently depleted not only LSD1 but also EZH2, SUZ12<br />

and BMI1 in AML cells. Co-treatment with PS enhanced the chromatin<br />

modifying effects <strong>of</strong> HCI2509. The combination also synergistically induced<br />

loss <strong>of</strong> viability <strong>of</strong> cultured and primary AML cells (combination<br />

indices, CI �1.0). Following tail vein infusion and establishment <strong>of</strong> AML by<br />

OCI-AML3 cells in NOD-SCID mice, treatment with HCI2509 (25 mg/kg,<br />

b.i.w, IP, for 3 weeks) improved survival, as compared to the control mice<br />

(p �0.001). HCI2509 treatment (15 mg/kg IP) also dramatically improved<br />

survival <strong>of</strong> NSG mice with established human AML following tail-vein<br />

injection <strong>of</strong> primary AML blasts expressing FLT3-ITD. Survival was further<br />

improved upon co-treatment with HCI2509 and PS (5 mg/kg IP, MWF).<br />

Mice did not experience any toxicity or weight loss. Conclusions: Combined<br />

epigenetic therapy with HCI2509 and PS exerts superior pre-clinical<br />

activity, warranting further in vivo development and testing <strong>of</strong> HCI2509<br />

against human AML.<br />

10552 General Poster Session (Board #44A), Mon, 1:15 PM-5:15 PM<br />

Prognostic role <strong>of</strong> microRNA polymorphisms in advanced gastric cancer<br />

patients. Presenting Author: Linnea Stenholm, University Hospital Aachen,<br />

Aachen, Germany<br />

Background: As little is known about the impact <strong>of</strong> microRNA (mir)<br />

polymorphisms on prognosis <strong>of</strong> advanced gastric cancer (AGC) we sought to<br />

determine their impact on overall survival (OS) in these patients (pts).<br />

Methods: Pts were treated with either 5-fluorouracil, leucovorin and<br />

oxaliplatin (FLO) or with additional docetaxel (FLOT) in frame <strong>of</strong> a phase III<br />

or 3 phase II trials <strong>of</strong> the Arbeitsgemeinschaft Internistische Onkologie. All<br />

pts consented genetic analyses. DNA was extracted from whole blood<br />

samples (n�515) and polymorphisms <strong>of</strong> mir26a1 (rs7372209), mir27a<br />

(rs895819), mir100 (rs1834306), mir125a (rs12975333), mir146a<br />

(rs2910164), mir196a2 (rs11614913), mir219-1 (rs107822) and mir423<br />

(rs6505162) were genotyped using PCR-based methods. Hetero- and<br />

homozygous variant genotypes were grouped versus homozygous wild type<br />

genotypes. Selection <strong>of</strong> clinical and genetic parameters for the final<br />

multivariate model was based on univariate and multivariate Cox-regression<br />

analyses with a cut-<strong>of</strong>f value <strong>of</strong> p � 0.25 and included 498 pts. Results:<br />

Median age was 67 (range 23 to 86) years. 42% were treated with FLO and<br />

58% with FLOT with a median OS <strong>of</strong> 14 months. Multivariate analyses<br />

revealed following associations: Genetic factors significantly associated<br />

with OS were mir26a1 variant genotypes (HR 1.3 [95%CI (1.01;1.6),<br />

p�.025]), mir27a variant genotypes (HR 1.3 [95%CI (1.01;1.6), p�.036])<br />

and mir196a2 variant genotypes (HR 0.8 [95%CI (0.6;0.99), p�.046]).<br />

<strong>Clinical</strong> factors with significant impact on OS were ECOG 2 performance<br />

status (HR 1.9 [95%CI (1.3;2.8), p�.002]), curative surgery <strong>of</strong> advanced<br />

disease (HR 0.3 [95%CI (0.1;0.5), p�.001]) and locally advanced disease<br />

(HR 0.5 [95%CI (0.3;0.7), p�.001]). Combined analyses <strong>of</strong> the identified<br />

adverse genotypes <strong>of</strong> mir26a1, mir27a and mir196a2 resulted in a median<br />

OS <strong>of</strong> 9, 13, 14 and 17 months for pts with 0 (n�72), 1 (n�193), 2<br />

(n�171) and 3 (n�54) adverse genotypes (p�.029), respectively.<br />

Conclusions: In addition to established clinical factors, mir26a1, mir27a<br />

and mir196a2 polymorphisms were significantly associated with OS. Our<br />

data suggest a strong impact <strong>of</strong> these mir polymorphisms on prognosis in<br />

AGC and might be <strong>of</strong> help for a better guidance <strong>of</strong> treatment decisions.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


10553 General Poster Session (Board #44B), Mon, 1:15 PM-5:15 PM<br />

Activation <strong>of</strong> and dependence on ataxia telangiectasia mutated kinase in<br />

PTEN-deficient tumor cells. Presenting Author: Nuala McCabe, Almac<br />

Diagnostics, Craigavon, United Kingdom<br />

Background: Loss <strong>of</strong> PTEN function has been widely reported to cause<br />

up-regulation <strong>of</strong> the PI3K/AKT signalling pathway resulting in increased<br />

cell growth, proliferation and survival. More recently it has been reported<br />

that PTEN null cells demonstrate genomic instability and have increased<br />

production <strong>of</strong> reactive oxygen species (ROS) and oxidative stress induced<br />

DNA damage. Ataxia Telangiectasia Mutated (ATM) is the primary response<br />

kinase, which responds to stalled DNA replication and DNA double strand<br />

breaks due to oxidative DNA damage. Methods: A metagene representing<br />

ATM activation was generated from cell line data and used to perform<br />

hierarchical clustering analysis <strong>of</strong> public DNA microarray pr<strong>of</strong>iling datasets<br />

<strong>of</strong> breast cancer, ovarian cancer and glioblastoma with known PTEN<br />

IHC/mutation status. Furthermore, we ask if ATM activation may be<br />

therapeutically exploited in PTEN null tumours using ATM specific siRNA<br />

and compounds in 2 PTEN isogenic cell line model systems. Results: We<br />

show that PTEN null cells have elevated levels <strong>of</strong> ROS, DNA damage and<br />

have endogenous activation <strong>of</strong> ATM, an enzyme important in responding to<br />

DNA damage resulting from oxidative stress. We hypothesised that PTEN<br />

deficient tumours may rely on ATM enzyme for survival. To investigate this<br />

we generated a 189-gene list representing ATM activation and used this to<br />

perform hierarchical clustering analysis <strong>of</strong> a breast cancer DNA microarray<br />

dataset. This list was able to significantly cluster tumours with known loss<br />

<strong>of</strong> PTEN expression (p�0.004). Furthermore, this gene list was able to<br />

segregate PTEN null/mutant tumours from PTEN wild-type tumours in 2<br />

independent datasets <strong>of</strong> glioblastoma and ovarian cancer (p�0.015 and<br />

p�0.012). In addition, we found that inhibition <strong>of</strong> ATM using the selective<br />

inhibitor KU-55933 caused DNA damage, cell cycle arrest and apoptosis<br />

specifically in PTEN deficient cells when compared to PTEN wild-type<br />

cells. Conclusions: These observations suggest that ATM may represent a<br />

therapeutic target in PTEN deficient tumours and furthermore ATM<br />

activation may be the basis <strong>of</strong> a biomarker <strong>of</strong> PTEN status in human<br />

cancers.<br />

10555 General Poster Session (Board #44D), Mon, 1:15 PM-5:15 PM<br />

Topoisomerase II alpha gene status, HER2, and microtubule-associated<br />

protein tau as predictors <strong>of</strong> pathologic complete response after neoadjuvant<br />

chemotherapy. Presenting Author: Agust Barnadas, Hospital de la Santa<br />

Creu i Sant Pau, Medical Oncology Department, Barcelona, Spain<br />

Background: There is growing evidence that topoisomerase II-alpha<br />

(TOPOII�) is a marker for anthracycline-, and microtubule-associated<br />

protein tau (MAPT) for taxane sensitivity. HER2 has been described as a<br />

marker <strong>of</strong> both anthracycline and taxane sensitivity.The goal <strong>of</strong> our study<br />

was to examine the predictive and prognostic value <strong>of</strong> TOPOII�, MAPT and<br />

HER2 expression in breast cancer patients (pts) who received neoadjuvant<br />

chemotherapy (NAC) based on anthracycline-taxane regimens. We analyzed<br />

the relationship between these biomarkers and pathological complete<br />

response (pCR), disease-free survival (DFS) and overall survival (OS).<br />

Methods: Between November 1993-2009, 140 pts (mean age 52 years)<br />

with locally advanced breast cancer (T1-4, N0-3, M0) were retrospectively<br />

studied. The study contained 2 groups: group A included 85 pts who<br />

received NAC with an anthracycline-taxane regimen and group B included<br />

the last 55 pts who received NAC with only an anthracycline-based<br />

regimen. HER2 was tested by immunohistochemistry (IH) or FISH, TOPOII�<br />

by CISH and MAPT by IH. Expression <strong>of</strong> these proteins was evaluated in<br />

core needle breast biopsy. Results: Overall, 12 pts (8.5%) had a pCR.<br />

TOPOII� was amplified in 6 (4%) <strong>of</strong> the tumors. Among pts without<br />

amplification, 6 (4%) had deletion <strong>of</strong> the TOPOII�, and 10 (7%) polysomia<br />

<strong>of</strong> chromosome 17. TOPOII� gene aberrations (amplification, deletion,<br />

polysomia), which was present in 22 (25%) <strong>of</strong> the tumors, was a strong<br />

predictive factor for pCR (p�0.018) but showed no direct association with<br />

prognostic outcome in multivariate analysis. HER2 amplification, which<br />

was present in 16% (21) <strong>of</strong> the tumors, show no direct association with<br />

pCR, DFS or OS. Finally, differences by treatment arm or pCR in low versus<br />

high MAPT expression groups were not observed, indicating that MAPT is<br />

not a useful predictive marker for taxane-based chemotherapy. In multivariate<br />

analysis high MAPT expression was associated with longer DFS<br />

(p�0.002). Conclusions: TOPOII� gene aberrations but not HER2 are<br />

highly predictive <strong>of</strong> pCR for anthracycline-based regimen. MAPT is not<br />

associated with response to taxanes but has a prognostic value.<br />

Tumor Biology<br />

669s<br />

10554 General Poster Session (Board #44C), Mon, 1:15 PM-5:15 PM<br />

Central review <strong>of</strong> discordant samples for microarray based on ER, PR, and<br />

HER2 and local IHC/FISH assessment worldwide from 827 patients.<br />

Presenting Author: Jelle Wesseling, Netherlands Cancer Institute - Antoni<br />

van Leeuwenhoek Hospital, Amsterdam, Netherlands<br />

Background: Differences in fixation and IHC and subjective interpretation<br />

can substantially affect the accuracy and reproducibility <strong>of</strong> estrogen<br />

receptor (ER), progesterone receptor (PR) and HER2 expression. The<br />

commercially available TargetPrint test measures the mRNA expression<br />

level <strong>of</strong> ER, PR and HER2 and is 98% concordant with centrally assessed<br />

ER as presented by Viale et al, SABCS 2011. This study compares results<br />

from the microarray-based TargetPrint with IHC and FISH (for HER2<br />

IHC2�) generated by local standard procedures. Methods: Fresh tumor<br />

samples (core needle biopsies or surgical) were collected for 831 patients<br />

diagnosed with breast cancer stage I to IV (Feb 2008 - Jan 2011) from 22<br />

hospitals from Europe, New Zealand, Japan and US. The results <strong>of</strong> the<br />

IHC/FISH assessments performed according to the local standards at the<br />

hospitals were compared to the quantitative gene expression readouts with<br />

TargetPrint. Discordant cases were centrally reviewed for IHC/FISH assessment.<br />

Results: Of the 831 samples, IHC assessment was unknown for 4 ER/<br />

PR samples; HER2 was unknown for 12 samples. Comparison <strong>of</strong> IHC and<br />

gene expression read out by TargetPrint showed a concordance <strong>of</strong> 95% for<br />

ER; 83% for PR and 94% for HER2. In this study, 3% <strong>of</strong> all IHC ER positive<br />

samples were classified negative by microarray, and 11% <strong>of</strong> IHC PR<br />

positive samples were classified negative by microarray. For HER2, 4% <strong>of</strong><br />

IHC/FISH HER2 positive samples were classified negative by microarray<br />

and 2% <strong>of</strong> IHC/FISH HER2 negative samples were classified positive by<br />

microarray. Most notably, all available 5 ER IHC negative/TargetPrint<br />

positive samples turned out to be positive with central re-assessment.<br />

HER2 IHC2� samples with discordant classifications for TargetPrint and<br />

local assessment are currently being reviewed for FISH/SISH assessment.<br />

Conclusions: Microarray based readout <strong>of</strong> ER, PR and HER2 status using<br />

TargetPrint is fairly comparable to local IHC and FISH analysis in 827<br />

analyzed samples in various hospitals worldwide. However, re-assessment<br />

<strong>of</strong> discordant cases –especially IHC ER-/TargetPrint ER� cases- confirms<br />

TargetPrint to be a useful high quality second opinion for local IHC/FISH<br />

assessment.<br />

10556 General Poster Session (Board #44E), Mon, 1:15 PM-5:15 PM<br />

ALK amplification and crizotinib sensitivity in non-small cell lung cancer<br />

cell lines and patients report. Presenting Author: Kalai Khadija, Institut<br />

Gustave Roussy, Villejuif, France<br />

Background: ALK high copy number (HCN) seems to be a frequent event,<br />

described in 13-17% <strong>of</strong> Non-small cell lung cancer (NSCLC). The goal <strong>of</strong><br />

this study was to describe ALK genomic aberrations on NSCLC patients and<br />

cell lines, to explore the ALK HCN response to crizotinib through in vitro<br />

assays and to report three patients case. Methods: 191 Paraffin embedded<br />

specimens from advanced NSCLC patients and 27 NSCLC cancer cell lines<br />

were screened for ALK copy number by fluorescent in situ hybridization<br />

(FISH). Crizotinib sensitivity was evaluated in 9 cell lines through WST1<br />

assays and clonogenic tests. Three patients exhibiting ALK HCN were<br />

assessed for response to crizotinib. Results: EML4-ALK translocation was<br />

present in 22 pts (11.5%). 21 pts (11%) exhibited over 6 copies <strong>of</strong> ALK.6<br />

(22%) cell lines displayed more than 5 copies <strong>of</strong> ALK, 19 (70%) presented<br />

a gain <strong>of</strong> 3 or 4 ALK copy number, only one cell line exhibited normal ALK<br />

copies and one harbored EML4-ALK translocation. FISH with CEP2<br />

revealed a polysomy <strong>of</strong> chromosome 2 in cases with ALK HCN.Out <strong>of</strong> the 9<br />

cell lines tested, 4 ALK HCN cell lines (H661, A427, BEN, H1299)<br />

exhibited increased sensitivity for crizotinib vs. 3 low ALK copy number<br />

(LCN) cell lines (H1975, H1651, H1650) with a low sensitivity. Median<br />

IC50 with crizotinib values was1750 nM [300-2800nM] in ALK HCN cell<br />

lines vs 4500 nM [800-8000nM] in ALK LCN cell lines, p�0.35. 3<br />

patients with ALK HCN tumor received crizotinib ( in 4th , 5th and 6th -line<br />

therapy) for 2, 3 and 5 months with stable disease as best response and<br />

clinical benefit in 2 pts. Conclusions: ALK HCN may predict sensitivity to<br />

crizotinib. A clinical study is planned in ALK HCN pts.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


670s Tumor Biology<br />

10557 General Poster Session (Board #44F), Mon, 1:15 PM-5:15 PM<br />

Exosomal-miRNA pr<strong>of</strong>iles as diagnostic biomarkers in cervical cancer.<br />

Presenting Author: Hirotaka Nishi, Department <strong>of</strong> Obstetrics and Gynecology,<br />

Tokyo Medical University, Tokyo, Japan<br />

Background: MicroRNA (miRNA) expression is altered in cancer cells and<br />

associated with the development and progression <strong>of</strong> various types <strong>of</strong><br />

cancer. miRNA could serve as diagnostic or prognostic biomarker for<br />

cancer patients. Our study was designed to analyze circulating exosomalmiRNA<br />

in patients with cervical cancer. Methods: Total RNA was extracted<br />

from serum in healthy women and patients with cervical intraepithelial<br />

neoplasia (CIN) and cervical cancer. We first explored miRNA expression<br />

pr<strong>of</strong>iles using miRCURY LNA microRNA Array (Exiqon) in each six<br />

malignant and healthy serum samples. miRNAs with significant differences<br />

in expression were validated in larger sample sets by a quantitative<br />

real-time RT-PCR using TaqMan gene expression assays (Applied Biosystems).<br />

Results: Six <strong>of</strong> 1223 miRNAs compared in serum samples from<br />

cervical cancer and normal control were found to have a �3.0-fold change<br />

with a P value �0.01 using miRCURY LNA microRNA Array. In a validation<br />

set (n�101), expression <strong>of</strong> 3 <strong>of</strong> the 6 miRNAs, miR-483-5p, miR-1246<br />

and miR-1275, was significantly different between the cervical cancer<br />

(n�40) and control (n�20) samples. We also found that the median levels<br />

<strong>of</strong> these miRNAs were significantly higher in patients with cervical cancer<br />

(n�40) than those in CIN (n�41). Circulating miRNAs were not correlated<br />

with clinical-pathological parameters except for miR-1246. Compared to<br />

patients with squamous cell carcinoma, the miR-1246 level was significantly<br />

higher in those with adenocarcinoma. Whereas, receiver-operator<br />

characteristic (ROC) curve analyses suggest that these serum miRNAs may<br />

be useful markers for discriminating patients with cervical cancer from<br />

healthy controls. Conclusions: Expression <strong>of</strong> circulating miR-483-5p,<br />

miR-1246 and miR-1275 is significantly higher in the blood <strong>of</strong> cervical<br />

cancer patients compared to controls and may potentially serve as a useful<br />

biomarker for cervical cancer diagnosis. Especially, miR-1246 may be a<br />

candidate for early detection <strong>of</strong> cervical adenocarcinoma. Larger-scaled<br />

studies are warranted to fully explore the role <strong>of</strong> circulating exosomalmiRNAs<br />

in cervical cancer.<br />

10559 General Poster Session (Board #44H), Mon, 1:15 PM-5:15 PM<br />

Use <strong>of</strong> next-generation sequencing (NGS) to detect high frequency <strong>of</strong><br />

targetable alterations in primary and metastatic breast cancer (MBC).<br />

Presenting Author: Lajos Pusztai, University <strong>of</strong> Texas M. D. Anderson<br />

Cancer Center, Houston, TX<br />

Background: The aim <strong>of</strong> this study was to assess the frequency <strong>of</strong> genomic<br />

alterations in breast cancer potentially treatable with approved targeted<br />

agents or investigational drugs in clinical trials. NGS was performed in a<br />

CLIA setting (Foundation Medicine). Methods: DNA was extracted from<br />

needle biopsies <strong>of</strong> 33 pre-therapy primary and 17 MBCs (mean age 52 yrs;<br />

58% ER�, 20% HER2�, 30% triple negative) obtained prospectively for<br />

biomarker discovery and preserved in RNAlater. Patients with MBC<br />

received an average <strong>of</strong> 7 drugs (range 5-17) including adjuvant therapy<br />

before biopsy for this research; 13 biopsies were from s<strong>of</strong>t tissues, 3 from<br />

liver and 1 from bone. Sequencing was targeted to 3230 exons in 182<br />

cancer-related genes and 37 introns in 14 genes <strong>of</strong>ten rearranged in<br />

cancer. Average median depth was �1200x. Results: All biopsies yielded<br />

sufficient DNA. NGS revealed a total <strong>of</strong> 117 known driver mutations across<br />

36 genes (per-tumor average�2.5, range 1-6), including 37 base substitutions<br />

(32%), 28 indels (24%), 42 amplifications (36%) and 10 homozygous<br />

deletions (9%). NGS identified HER2 gene amplification in 6/7 cases<br />

scored HER2� by FISH. The average number <strong>of</strong> functionally important<br />

alterations was surprisingly similar, 2.3 in primaries vs 2.8 in heavily<br />

treated MBCs (p�0.32). Remarkably, 25/33 (76%) <strong>of</strong> primary and 14/17<br />

(82%) <strong>of</strong> MBCs had at least 1 genomic alteration targetable with an FDA<br />

approved drug or novel agent in clinical trials. These included: ERBB2<br />

alterations (n�9), PIK3CA mutations (n�8), NF1 mutations (n�4, candidate<br />

for PI3K/MEK inhibitors), AKT1-3 mutations (n�5, PI3K inhibitors),<br />

BRCA1/2, (n�6, PARP inhibitors), and CCND2 (n�3)/CDKN2A (n�3)<br />

mutations (CDK inhibitors). Numerous other alterations with less apparent<br />

therapeutic implications were also observed. Conclusions: Comprehensive<br />

NGS pr<strong>of</strong>iling in breast cancer needle biopsies showed high frequency <strong>of</strong><br />

genomic alterations linked to a clinical treatment option or clinical trials <strong>of</strong><br />

targeted therapies. These results demonstrate it is feasible to use NGS to<br />

guide targeted therapy. Prospective testing <strong>of</strong> the diagnostic/predictive<br />

value <strong>of</strong> this patient selection approach is currently under way.<br />

10558 General Poster Session (Board #44G), Mon, 1:15 PM-5:15 PM<br />

Evaluation <strong>of</strong> gene expression by RNA-seq after single dose <strong>of</strong> trastuzumab<br />

(T) reveals predictors <strong>of</strong> pathologic complete response (pCR) in HER2positive<br />

early breast cancer. Presenting Author: Natalie Galanina, Yale<br />

University School <strong>of</strong> Medicine, Yale Comprehensive Cancer Center, New<br />

Haven, CT<br />

Background: The use <strong>of</strong> a ‘brief exposure’ to single agent T allows the<br />

measurement <strong>of</strong> dynamic changes in the transcriptome that may predict<br />

response to T-based combinations. We have shown that most gene<br />

expression changes in HER2� tumors treated with T occur in tumors that<br />

ultimately achieve a pCR. Our further analysis suggests several patterns <strong>of</strong><br />

transcriptional change in pCR tumors suggesting different mechanisms <strong>of</strong><br />

action <strong>of</strong> T. RNA-seq analysis provides more in-depth annotation <strong>of</strong> these<br />

mechanisms. Methods: Fresh tumor core biopsies were taken at a 2 week<br />

time point after a single dose <strong>of</strong> T (8mg/m2) from 80 HER2� early breast<br />

cancer patients enrolled on a clinical trial <strong>of</strong> T�T�C. Nucleic acids were<br />

extracted using Qiagen AllPrep and were analyzed with Illumina HT12v3<br />

Beadchip and Illumina 610 QUAD V1 SNP arrays. RNA was also processed<br />

for sequencing using the Ovation RNA-Seq System and paired-end sequenced<br />

using an Illumina Genome Analyzer IIxRNA-seq data was analyzed<br />

with Tophat/Cufflinks. Network analysis was performed with Metacore.<br />

Results: Among pCR tumors, distinct patterns <strong>of</strong> differential expression<br />

pre/post T were observed, in both microarray and RNA-seq data. ERBB2<br />

down-regulation was characteristic <strong>of</strong> pCR in one subgroup by microarray.<br />

In this group, differentially expressed genes belonged to interaction<br />

networks involved in apoptosis and cell cycle regulation. In contrast,<br />

tumors with no change in ErbB2 showed differentially expressed genes that<br />

belonged to networks related to chromatin assembly and regulation <strong>of</strong><br />

immune pathways. NOLC1, RPL41, ZCHHC17, and B2M had altered<br />

alternative splicing product distributions in both groups. In the ERBB2<br />

down-regulated group, genes with changed expression were enriched for<br />

targets <strong>of</strong> STAT3 and YY1. Conclusions: RNA-seq and microarray reveal<br />

distinct responses in tumors that achieve pCR to T-containing regimens.<br />

These methods provide predictive markers for validation in subsequent<br />

clinical trials.<br />

10560 General Poster Session (Board #45A), Mon, 1:15 PM-5:15 PM<br />

EGFR exon 20 insertion mutations: Incidence and clinicopathologic<br />

characteristics in U.S. patients with lung adenocarcinoma. Presenting<br />

Author: Maria E. Arcila, Memorial Sloan-Kettering Cancer Center, New<br />

York, NY<br />

Background: Activating insertion mutations in exon 20 <strong>of</strong> EGFR are reported<br />

in a small subset <strong>of</strong> lung adenocarcinomas (ADC). In contrast to the classic<br />

EGFR mutations, they appear to confer primary resistance to currently<br />

approved EGFR tyrosine kinase inhibitors. Their incidence and clinicopathologic<br />

features are not well established. Methods: Lung ADCs (n�1500)<br />

were screened for major activating mutations in EGFR (exons 19 and 21)<br />

and KRAS (exon 2). Negative cases were tested for EGFR exon 20<br />

insertions by a PCR-based sizing assay. Extended testing for additional<br />

recurrent point mutations in EGFR, KRAS, BRAF, NRAS, PIK3CA, MEK1<br />

and AKT was performed in all cases by Sequenom mass spectrometry. A<br />

subset <strong>of</strong> cases was also tested for ALK rearrangements by FISH. Results:<br />

We identified 32cases withEGFRexon 20 insertions, accounting for 11% <strong>of</strong><br />

all EGFR mutations. EGFRexon 20 insertions were mutually exclusive with<br />

the other genetic alterations tested except for PIK3CA mutations. The<br />

incidence was higher among never-smokers (p�0.0001) but there was no<br />

association with sex, ethnic origin or stage at diagnosis. Insertions were 3,<br />

6, 9 or 12bp; 9bp insertions were most common (50%, 16/32). Morphologically,<br />

90% <strong>of</strong> tumors were moderate to poorly differentiated with a<br />

predominant mixed ADC phenotype. Conclusions: EGFR exon 20 testing<br />

may identify a unique subset <strong>of</strong> EGFR mutant lung ADCs which is<br />

significantly larger than previously reported, making this the third most<br />

common type <strong>of</strong> EGFR mutation after exon 19 deletions and L858R. This<br />

population could potentially benefit from alternate targeted therapies,<br />

many <strong>of</strong> which are currently in clinical development.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


10561 General Poster Session (Board #45B), Mon, 1:15 PM-5:15 PM<br />

Independent validation <strong>of</strong> prognostic value <strong>of</strong> 22 micrornas (miRs) in stage<br />

I-II squamous cell lung cancer (SqCLC). Presenting Author: Marcin Tomasz<br />

Skrzypski, Medical University <strong>of</strong> Gdansk, Gdansk, Poland<br />

Background: About 50% <strong>of</strong> NSCLC patients (pts) will develop distant<br />

metastases following pulmonary resection. Currently, apart from clinical<br />

stage at diagnosis, there are no reliable factors to select high risk pts for<br />

adjuvant chemotherapy. We previously demonstrated prognostic value <strong>of</strong><br />

22 miRs in frozen tissue samples <strong>of</strong> early stage SqCLC, and the feasibility<br />

<strong>of</strong> their expression assessment in formalin fixed paraffin embedded (FFPE)<br />

samples (Skrzypski et. al. J Clin Oncol 2010;28;15s). In this study, we<br />

validated the prognostic value <strong>of</strong> these miRs in an independent cohort <strong>of</strong><br />

early SqCLC pts. Methods: FFPE tumor samples were obtained from 132<br />

stage I-II SqCLC pts who underwent radical pulmonary resection, 42% <strong>of</strong><br />

whom developed distant metastases. Median follow-up <strong>of</strong> pts who did not<br />

develop metastases was 5.6 years (range, 4.1-10.0 years). miRs were<br />

isolated from tumor tissue with RecoverAll kit (Ambion). Expression <strong>of</strong> 22<br />

miRs previously found to be related to the risk <strong>of</strong> metastases was analyzed<br />

by RT-PCR assay (Appliedbiosystems). Raw data were normalized vs. the<br />

expression <strong>of</strong> U6. Individual miRs and the risk score comprising 5 most<br />

predictive miRs were correlated with distant metastases-free survival<br />

(MFS). Results: Eight miRs were significantly relatedtoMFS. MiR-192 was<br />

significantly correlated with MFS (p�0,0007; p�0,02 after correction for<br />

multiple comparisons). The 5-miR risk score was an independent prognostic<br />

factor (p�1.70E-06) in a multivariate analysis comprising stage<br />

(p�1,10E-03) and histological grade (p�0,46). Using the median <strong>of</strong> the<br />

risk score as a cut-<strong>of</strong>f value, the median MFS was 1.73 years in the high risk<br />

group, and not reached in the low risk group (HR�2.11). The sensitivity<br />

and specificity <strong>of</strong> the risk score to predict the occurrence <strong>of</strong> distant<br />

metastases were 66% and 64%, respectively. Conclusions: Risk score<br />

based on expression <strong>of</strong> 5 miRs is a strong and independent predictor <strong>of</strong><br />

distant relapse in operable early SqCLC.<br />

10563 General Poster Session (Board #45D), Mon, 1:15 PM-5:15 PM<br />

Effect <strong>of</strong> the loss <strong>of</strong> the type III TGF� receptor during tumor progression on<br />

tumor microenvironment: Preclinical development <strong>of</strong> TGF� inhibition and<br />

TGF�-related biomarkers to enhance immunotherapy efficacy. Presenting<br />

Author: Brent Allen Hanks, Duke University Medical Center, Durham, NC<br />

Background: Overall, the clinical efficacy <strong>of</strong> tumor immunotherapy has been<br />

limited. Our incomplete understanding <strong>of</strong> the complex interplay between<br />

tumors and the immune microenvironment has contributed to these<br />

modest outcomes. Our work has revealed that several tumors downregulate<br />

the expression <strong>of</strong> the type III TGF� receptor (T�RIII) with progression.<br />

T�RIII is shed from the cell surface to generate soluble T�RIII (sT�RIII)<br />

which is capable <strong>of</strong> sequestering TGF�. Methods and Results: Using both<br />

breast cancer and melanoma tumor models we have demonstrated that the<br />

loss <strong>of</strong> T�RIII expression is associated with diminished tumor infiltrating<br />

CD8� T cells and increased regulatory T cells (Tregs) within the tumor<br />

microenvironment. Our data implies that these alterations correlate with<br />

suppressed tumor antigen-specific T cell responses and more rapid disease<br />

progression. We show that these changes are due to enhanced TGF�<br />

signaling within the immune compartment <strong>of</strong> the tumor microenvironment<br />

resulting in enhanced expression <strong>of</strong> the indoleamine 2,3-dioxygenase<br />

immunoregulatory enzyme by local plasmacytoid dendritic cells (DCs) as<br />

well as increased expression <strong>of</strong> the Treg-recruiting CCL22 chemokine by<br />

local myeloid DCs. Microarray analysis indicates that these same gene<br />

expression associations also exist in human breast cancers. Consistent with<br />

these studies, we have demonstrated that TGF-� inhibition synergistically<br />

enhances the efficacy <strong>of</strong> a Her2/neu vaccine in a breast cancer model and<br />

that plasma levels <strong>of</strong> sT�RIII correlate with clinical response and overall<br />

survival in stage III melanoma patients. Conclusions: We have elucidated a<br />

novel mechanism that tumors utilize to suppress the generation <strong>of</strong><br />

anti-tumor immunity by establishing a link between the loss <strong>of</strong> an<br />

endogenous suppressor <strong>of</strong> tumor metastasis, T�RIII, and the generation <strong>of</strong><br />

an immunotolerant tumor microenvironment. We are pursuing a phase I<br />

clinical trial to investigate the efficacy <strong>of</strong> combining a TGF-� inhibitor with<br />

a tumor vaccine while also determining if sT�RIII may function as a<br />

predictive biomarker for this approach.<br />

Tumor Biology<br />

671s<br />

10562 General Poster Session (Board #45C), Mon, 1:15 PM-5:15 PM<br />

Lymphopenia combined with low TCR diversity to predict overall survival in<br />

metastatic breast cancer patients. Presenting Author: Manuarii Manuel,<br />

Centre de Recherche en Cancérologie de Lyon, Inserm U1052 CNRS<br />

5286, Centre Léon Bérard, Lyon, France<br />

Background: Lymphopenia (�1 Giga/L) before initiation <strong>of</strong> chemotherapy is<br />

a predictive factor for toxicity and death in metastatic phase for cancer<br />

patients. Combinatorial diversity <strong>of</strong> T Cell Receptor beta chain (TCR-ß), as<br />

a measure <strong>of</strong> T cell repertoire diversity, was investigated and tested either<br />

alone or in combination with lymphopenia as a prognostic factor for overall<br />

survival (OS) in first line treatment <strong>of</strong> metastatic breast cancer (MBC)<br />

patients. Methods: Using semi quantitative multiplex PCR, the V-D-J<br />

combinatorial diversity <strong>of</strong> the TCR was measured on cryo-preserved blood<br />

samples from 2 cohorts <strong>of</strong> MBC patients before the initiation <strong>of</strong> the first line<br />

chemotherapy: in an experimental cohort (cohort A, n�66) and in a<br />

validation series (cohort B, n�67). A prognostic score, defined NDL<br />

(Number & Diversity <strong>of</strong> Lymphocytes) combining lymphocyte count and<br />

TCR diversity was delineated. Univariate and multivariate analysis <strong>of</strong><br />

prognostic factors for OS were performed in both cohorts. Results: Lymphopenia<br />

(�1 Giga/L) was associated with shorter OS for cohort B while TCR<br />

diversity �33% (called divpenia) was associated with a reduced OS in<br />

cohort A. The combination <strong>of</strong> lymphopenia with low TCR diversity (called<br />

lympho-divpenia) was associated with poor OS compared to patients with<br />

either lymphocyte count �1 Giga/L or diversity �33% or both, in cohort A<br />

(median OS: 7.6 vs 24.5 months, p.value �0.0006) and cohort B (median<br />

OS 10.6 vs 22.9 months, p.value �0.0035). In a multivariate analysis,<br />

including all significant clinical factors from the univariate analysis (PS,<br />

liver metastasis, hemoglobin) lympho-divpenia was found to be an independent<br />

prognostic factor in the pooled cohort (A�B) (p�0.005) along with<br />

triple negative tumors (p�0.011) and hemoglobin level (11.5 g/dL)<br />

(p�0.009). Conclusions: NDL score combining lymphopenia and reduced<br />

TCR diversity seems to be a strong prognostic factor for OS and could be<br />

use to improve care quality <strong>of</strong> MBC patients.<br />

10565 General Poster Session (Board #45F), Mon, 1:15 PM-5:15 PM<br />

Involvement <strong>of</strong> NK cell molecular signatures in favorable prognosis <strong>of</strong><br />

breast cancer patients. Presenting Author: Maria Libera Ascierto, National<br />

Institutes <strong>of</strong> Health, Bethesda, MD<br />

Background: Tumor cell recognition by NK cells is mediated by the<br />

interaction <strong>of</strong> activating and inhibitory NK cell receptors with their ligands<br />

expressed on tumor cells. In addition, NK cells express adhesion molecules<br />

that facilitate formation <strong>of</strong> the immunological synapse with the tumor<br />

targets. Here, we investigated whether the coordinate expression <strong>of</strong> NK<br />

activating receptors and adhesion molecules could provide a signature to<br />

segregate breast cancer patients into relapse and relapse-free outcomes.<br />

Methods: Gene expression pr<strong>of</strong>iling, RT-PCR screening and survival analysis<br />

were performed on RNA extracted from primary breast cancers. Tumors<br />

were obtained from patients experiencing either 5-8 years relapse-free<br />

survival or tumor relapse within 1-3 years following initial treatment.<br />

Results: Tumors from patients with a favorable prognosis were characterized<br />

by increased expression <strong>of</strong> genes involved in NK cell interaction with<br />

tumor cells and its activation signaling. In particular, up-regulation <strong>of</strong><br />

Natural Cytotoxicity Receptors (NCRs), leukocyte function-associated antigen<br />

1 (LFA-1), CD226 (DNAM-1) and CD96 was observed in relapse-free<br />

patients. Thus, the expression <strong>of</strong> the NK activating receptors and relevant<br />

adhesion molecules involved in NK cell:target interactions can predict<br />

relapse free survival in breast cancer patients. Conclusions: Results from<br />

the present study, highlighted the effector cooperation between the innate<br />

and adaptive immune components within the tumor microenvironment.<br />

The NK cells parameters identified in this study, together with the<br />

prognostic B and T cell signatures previously reported by us, represent a<br />

powerful tool for predicting breast cancer outcome which might be easily<br />

introduced in clinical practice.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


672s Tumor Biology<br />

10566 General Poster Session (Board #45G), Mon, 1:15 PM-5:15 PM<br />

Comparative analysis <strong>of</strong> myeloid-derived suppressor cell (MDSC) subsets in<br />

patients with gastrointestinal (GI) malignancies. Presenting Author: Austin G.<br />

Duffy, National Cancer Institute/National Institutes <strong>of</strong> Health, Bethesda, MD<br />

Background: Immunotherapy represents a potential new treatment option for<br />

patients with cancer. Myeloid derived suppressor cells (MDSCs) constitute a<br />

heterogenous cell population, which inhibit the host anti-tumor immune<br />

response, limiting the effectiveness <strong>of</strong> both immune and non-immune treatments.<br />

MDSC are known to be increased in patients with GI cancer. Characterization<br />

<strong>of</strong> MDSC in humans has been difficult with heterogeneous phenotype<br />

classification. Differences in processing are also potentially important. The aim<br />

<strong>of</strong> this study was to perform a comparative analysis <strong>of</strong> MDSC subsets in patients<br />

with GI cancer. Methods: Fluorescence activated cell sorting (FACS) analysis was<br />

performed on PBMC and whole blood (WB) from healthy adults (N�44) and<br />

patients with GI cancer (N�41). MDSC subsets were enumerated using the<br />

following antibodies: anti-CD14; anti-CD15; anti-HLA-DR; anti-CD33; anti-<br />

CD11b; 7-AAD. Suppressor function <strong>of</strong> MDSC subsets was tested using FACS<br />

sorted MDSC subpopulations co-cultured with CD3/CD28-stimulated autologous<br />

effector cells. Proliferation and IFN-g were measured. Results: The frequencies<br />

<strong>of</strong> Monocytic (Def: CD14 � , HLA-DR -/low ) and Granulocytic (Def: CD15 � ,CD14 -<br />

CD33 � CD11b � ) MDSC are shown in the table below. Monocytic MDSC were<br />

increased in the blood <strong>of</strong> patients (vs control; P�.0001) and frozen (vs fresh<br />

PBMC; P�.05). While CD15� cells were increased in cancer patients we did not<br />

find an increase in granulocytic MDSC. Functional studies will also be presented.<br />

Conclusions: Monocytic MDSC are more frequent in patients with advanced GI<br />

cancer compared to healthy volunteers. An increase in this population was also<br />

found in WB and frozen PBMC compared to fresh PBMC reflecting the need for<br />

homogenous preparation in studies evaluating this population in humans. While<br />

CD15�CD14- cells were also increased in cancer patients we did not find an<br />

increase in CD14-CD15�CD33�CD11b� cells.<br />

CD14�ve, HLA-DR -/low* CD15�ve,CD14-veCD33�CD11b�*<br />

Patient PBMC 5.5 3.1<br />

Healthy PBMC 1.2 4.55<br />

Frozen PBMC (healthy) 4.16 3.3<br />

Frozen PBMC (patient) 10 1.36<br />

Patient WB 2.5 15<br />

Healthy WB<br />

* Rel frequency.<br />

0.8 17.65<br />

10568 General Poster Session (Board #46A), Mon, 1:15 PM-5:15 PM<br />

Extracellular matrix and breast cancer cells’ response to trastuzumab: The<br />

role <strong>of</strong> glycosaminoglycans, heparanases, and syndecan-1. Presenting<br />

Author: Maria Aparecida Silva Pinhal, Universidade Federal de São Paulo,<br />

Sao Paulo, Brazil<br />

Background: Trastuzumab is an antibody anti-epidermal growth factor 2<br />

receptor (HER2), which improves disease-free and overall survival in HER2<br />

positive breast cancer. Nevertheless, many patients become resistant to<br />

this treatment. Heparanase (HPSE) is an enzyme that is responsible for<br />

removal <strong>of</strong> heparan sulfate (HS) chains from proteoglycans, generating free<br />

oligosaccharides that modulate many physiopathological functions, including<br />

tumor developing. We have analyzed whether some extracellular matrix<br />

components influence trastuzumab efficacy. Methods: Heparanase-1<br />

(HPSE-1) overexpression effect was analyzed using MCF7 cells stable<br />

transfected with HPSE-1 cDNA (MCF7-HPSE-1). HPSE-1, HPSE-2, Syndecan-1<br />

(Syn-1) and HER2 expression, HPSE-1 activity and cell viability were<br />

evaluated in different breast cancer cells treated or not with trastuzumab.<br />

The glycosaminoglycans synthesis and shedding were also evaluated.<br />

Trastuzumab and HS binding were analyzed by confocal microscopy and<br />

Fluorescence Resonance Energy Transfer (FRET). Results: MCF7 transfected<br />

with HPSE-1 cDNA becomes completely resistant to trastuzumab.<br />

HS affinity by Trastuzumab was then tested, showing that they bind in high<br />

levels and this binding is necessary to antibody activity. In MCF7 cells,<br />

trastuzumab decreases HPSE-1, HPSE-2, HER2 and Syn-1 mRNA expression,<br />

while in MCF7-HPSE-1 the antibody increases the expression <strong>of</strong> these<br />

molecules. Conclusions: Our results have demonstrated that an ideal<br />

concentration <strong>of</strong> HS in cell surface, regulated by trastuzumab, is necessary<br />

to its action, beyond HER2 high levels. High HS concentration at cell<br />

surface enhances the antibody amount disposable to interact with HER2 in<br />

cell surface, determining breast cancer cells susceptibility to trastuzumab.<br />

These new insights could be useful when devising strategies for overcoming<br />

trastuzumab resistance in HER2 positive cancers. Supported by FAPESP,<br />

CNPq, CAPES.<br />

10567 General Poster Session (Board #45H), Mon, 1:15 PM-5:15 PM<br />

Correlation <strong>of</strong> the maximum standardized uptake value for f-18 fluorodeoxyglucose<br />

with molecular subtype and survival in advanced breast cancer.<br />

Presenting Author: Zhen Jia, Fudan University Cancer Center, Shanghai,<br />

China<br />

Background: Tumor glucose metabolism correlates with tumor biology and<br />

clinical outcome <strong>of</strong> breast cancer patients. This prospective cohort study<br />

was to explore correlations <strong>of</strong> 18F-FDG uptake, a surrogate for glucose<br />

metabolism, with molecular subtypes in women with advanced breast<br />

cancer. Methods: Patients diagnosed as advanced breast cancer were<br />

enrolled in the study. PET/CT was performed and the Maximum Standardized<br />

Uptake Value (SUVmax) <strong>of</strong> each lesion was documented as baseline,<br />

so was clinical and treatment information. Diagnosis <strong>of</strong> malignant nature <strong>of</strong><br />

a lesion was confirmed by pathology or further follow-up. Patients who had<br />

got a progressive disease in the efficacy assessment were assigned new<br />

treatments as per-institutional guidelines. Results: 244 patients met the<br />

criteria and were all put into this analysis. Independent factors for<br />

influencing SUVmax value included luminal subtype (p� 0.002), triplenegative<br />

subtype (p�0.001), ER status (p� 0.028), Her-2 status<br />

(p�0.001), and CEA level (p�0.001). A higher SUVmax was significantly<br />

associated with poorer median PFS (6.9 vs. 8.1 months, p�0.040) and OS<br />

(13.8 vs. 16.9 months, p�0.003). Cox Regression analysis showed that<br />

SUVmax value, previous treatments, subsequent treatments and treatment<br />

efficacy were four independent prognostic factors for PFS, while age,<br />

menopausal status, disease free interval, 4 subtypes, previous treatments,<br />

number <strong>of</strong> metastatic sites, SUVmax value and subsequent treatment<br />

efficacy were independent prognostic factors for OS. Conclusions: 18F-FDG uptake correlates with molecular subtypes in women with advanced breast<br />

cancer. Baseline SUVmax is an independent prognostic factor for survival<br />

<strong>of</strong> patients with advanced breast cancer, especially those with luminal<br />

subtypes.<br />

10569 General Poster Session (Board #46B), Mon, 1:15 PM-5:15 PM<br />

Detection <strong>of</strong> EML4-ALK in serum RNA from lung cancer patients using<br />

MassARRAY platform. Presenting Author: Keita Kudo, Thoracic Oncology<br />

Center, Cancer Institute Hospital, Japanese Foundation for Cancer Research,<br />

Tokyo, Japan<br />

Background: The presence <strong>of</strong> the transforming fusion gene EML4-ALK in<br />

non–small cell lung cancer (NSCLC) is a predictive marker for the efficacy<br />

<strong>of</strong> ALK kinase inhibitors. The break apart fluorescence in situ hybridization<br />

(FISH) assay has served as the standard test for identification <strong>of</strong> NSCLC<br />

patients with ALK gene rearrangement. Circulating RNA in plasma/serum is<br />

an emerging field for noninvasive molecular diagnosis. We previously<br />

developed a MassARRAY assay to detect nine types <strong>of</strong> EML4-ALK fusion<br />

gene (variants 1, 2, 3a, 3b, 4, 5a, 5b, 6, and 7). The assay is based upon<br />

region-specific PCR followed by specific single-base primer extension<br />

reactions; the extended products are resolved using MALDI-TOF mass<br />

spectrometry. In this study, we evaluated whether EML4-ALK fusion RNA<br />

could be detected in serum obtained from NSCLC patients. Methods: The<br />

RNA was extracted from serum obtained from 12 lung cancer patients with<br />

ALK rearrangements by FISH analysis. The extracted RNA was reverse<br />

transcribed into cDNA, and the resultant cDNA was amplified by nested<br />

PCR. The PCR product was subjected to MassARRAY assay to identify<br />

EML4-ALK variants. Results: The EML4-ALK RNA (variant 1 and 3) was<br />

detected in serum from 2 <strong>of</strong> 12 patients. These are confirmed by<br />

subcloning and sequencing. Conclusions: The detection <strong>of</strong> EML4-ALK in<br />

serum RNA samples by MassARRAY assay is feasible. This noninvasive<br />

assay will be useful in patients with insufficient or unavailable tumor<br />

specimens. Additional experiments will reveal the sensitivity and specificity<br />

<strong>of</strong> this detection system.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


10570 General Poster Session (Board #46C), Mon, 1:15 PM-5:15 PM<br />

Hybrid PET-MRI <strong>of</strong> the breast: A promising tool for the characterization <strong>of</strong><br />

breast tumors. Presenting Author: Katja Pinker-Domenig, Medical University<br />

Vienna, Department <strong>of</strong> Radiology, Division <strong>of</strong> Molecular and Gender<br />

Imaging, Vienna, Austria<br />

Background: To evaluate if PET-MRI <strong>of</strong> breast tumors improves staging <strong>of</strong><br />

breast cancer and obviates unnecessary breast biopsies. Methods: 106<br />

patients with breast tumors detected by mammography or ultrasound and<br />

classified as BIRADS 3-5 were included in this IRB approved prospective<br />

study. All patients were examined with dedicated 18FDG-PET-CT and 3T<br />

multiparametric MRI <strong>of</strong> the breast. Examinations were scheduled no longer<br />

than 7 days apart. MRI protocol included: a diffusion-weighted sequence<br />

(DWI), a T2-w sequence and a contrast-enhanced combined high temporal<br />

and spatial resolution 3D-T1-w sequence before and after application <strong>of</strong> a<br />

standard dose Gd-DOTA. For PET-CT patients fasted at least 6 h before<br />

injection <strong>of</strong> approx. 300 MBq 18F-FDG based on the patients weight.<br />

Scanning was started 45 min after injection. Blood glucose levels were<br />

�150 mg/dl. A prone PET dataset over the breasts was acquired using a<br />

positioning device allowing the same patient geometry as the breast MRI<br />

coil. CT data was used for attenuation correction. Co-registration <strong>of</strong> imaging<br />

data and image fusion were performed. PET-MRI was assessed for lesion<br />

morphology and EH-kinetics according to BI-RADS and restricted diffusivity<br />

with an ADC threshold 1.25 x10-3mm2/s set as the cut-<strong>of</strong>f for<br />

malignancy. Lesions were assed for 18F-FDG–avidity and classified as<br />

positive when 18F-FDG-uptake was greater than blood-pool activity. Additionally,<br />

nodal status was recorded for each technique and patient.<br />

Histopathology was used as the standard <strong>of</strong> reference. Results: PET-MRI<br />

achieved an excellent sensitivity <strong>of</strong> 100%. Specificity was increased from<br />

68% to 80% as compared to routinely used MRI. Diagnostic accuracy <strong>of</strong><br />

PET-MRI for diagnosis <strong>of</strong> breast cancer was 95%. PET-MRI would have<br />

obviated unnecessary breast biopsies in 80% <strong>of</strong> benign breast lesions<br />

without missing any cancers. Additionally, PET-MRI increased sensitivity<br />

in the detection <strong>of</strong> lymphnode metastases from 70% to 87% compared to<br />

MRI alone. Conclusions: With PET-MRI unnecessary breast biopsies can be<br />

obviated without missing any cancers. PET-MRI increases overall diagnostic<br />

accuracy in the diagnosis <strong>of</strong> breast cancer and lymphnode metastases<br />

for an accurate staging.<br />

10572 General Poster Session (Board #46E), Mon, 1:15 PM-5:15 PM<br />

The effect on detection rate <strong>of</strong> second cancers after the addition <strong>of</strong> MRI to<br />

conventional screening mammography in patients with a history <strong>of</strong> breast<br />

cancer. Presenting Author: Chana Weinstock, University <strong>of</strong> Maryland<br />

Marlene and Stewart Greenebaum Cancer Center, Baltimore, MD<br />

Background: Mammography is currently used in the surveillance <strong>of</strong> breast<br />

cancer survivors, who are at increased risk <strong>of</strong> developing ipsilateral and<br />

contralateral breast cancers regardless <strong>of</strong> age at diagnosis or time since<br />

diagnosis. Several prospective studies have shown the utility <strong>of</strong> breast MRI<br />

in other high risk populations; however, little data exists on the use <strong>of</strong> MRI<br />

for surveillance <strong>of</strong> breast cancer survivors. We aimed to compare the<br />

outcome <strong>of</strong> surveillance breast MRI vs. mammography in this population.<br />

Methods: We identified women �65 with non-metastatic breast cancer or<br />

DCIS with at least one MRI performed at our center �11 months after<br />

initial diagnosis, along with a mammogram done within 6 months <strong>of</strong> the<br />

MRI. We compared the outcome <strong>of</strong> MRI and mammography in terms <strong>of</strong><br />

biopsies performed as well as in detection <strong>of</strong> new cancers. Results: Of 512<br />

consecutive charts reviewed, 204 patients met inclusion criteria, 105<br />

(51.4%) <strong>of</strong> whom were African-<strong>American</strong>. The average number <strong>of</strong> MRIs per<br />

patient was 2.3 (range 2-7), with a total <strong>of</strong> 474 MRIs performed between<br />

2005 and 2011. MRI resulted in BIRADS scores <strong>of</strong> 1 or 2 in 73.5% <strong>of</strong><br />

studies vs. 84.4% for mammography. There were 19 biopsies performed<br />

due to MRI findings alone, 7 done due to mammographic findings alone,<br />

and 6 biopsies done based on abnormalities seen on both MRI and<br />

mammography. There were 7 malignancies identified based on an abnormal<br />

MRI, 3 seen on both MRI and mammography, and none identified via<br />

mammography alone. The malignancies identified via MRI alone included<br />

1 patient with DCIS, 5 with stage I disease, and 1 with isolated lung<br />

metastases. Of the 10 recurrences detected, 5 (50%) were in African<br />

<strong>American</strong>s. Two patients developed interval cancers within 6 months <strong>of</strong><br />

normal screening MRI and mammography. Sensitivity and specificity for<br />

MRI were 83.3% (95% CI 0.51-0.97) and 92.2% (95% CI 0.87-0.95), vs.<br />

25% (95% CI 0.05-0.57) and 94.8% (95% CI 0.90-0.97) for mammography.<br />

Positive and negative predictive values were 40% and 98.9% for MRI<br />

vs. 25% and 95.2% for mammography. Conclusions: Gadoliniumenhanced<br />

breast MRI is a useful surveillance modality in breast cancer<br />

survivors � age 65. Prospective studies are needed in this population.<br />

Tumor Biology<br />

673s<br />

10571 General Poster Session (Board #46D), Mon, 1:15 PM-5:15 PM<br />

Online biomarker validation <strong>of</strong> survival-associated biomarkers in breast and<br />

ovarian cancer using microarray data <strong>of</strong> 3,862 patients. Presenting Author:<br />

Balazs Gyorffy, Joint Research Laboratory <strong>of</strong> the Hungarian Academy <strong>of</strong><br />

Sciences and the Semmelweis University, Budapest, Hungary<br />

Background: The pre-clinical validation <strong>of</strong> prognostic gene candidates in<br />

large independent patient cohorts is a pre-requisite for the development <strong>of</strong><br />

robust biomarkers. Today, curated microarray cohorts combined with<br />

appropriate clinical data <strong>of</strong>fer a cost effective tool to prescreen potential<br />

new biomarkers. In present study we expanded our online Kaplan-Meier<br />

plotter tool to assess the effect <strong>of</strong> genes on ovarian cancer prognosis.<br />

Methods: Gene expression data and survival information <strong>of</strong> breast and<br />

ovarian cancer patients were downloaded from GEO and TCGA. To analyze<br />

the prognostic value <strong>of</strong> the selected gene in the various cohorts the patients<br />

are divided into two groups according to the quantile expression <strong>of</strong> the<br />

gene. Filtering is implemented for stage, grade, and histology subtypes.<br />

Follow-up threshold is implemented to exclude long-term effects. A<br />

Kaplan-Meier survival plot is generated and significance is computed in the<br />

R statistical environment using Bioconductor packages. The combination<br />

<strong>of</strong> several probe sets can be employed to assess the mean <strong>of</strong> their<br />

expression as a multigene predictor <strong>of</strong> survival. Results: All together 1,390<br />

ovarian cancer patients and 2,472 breast cancer patients were entered into<br />

the database. These groups can be compared using relapse free survival<br />

(n�2,324 in breast cancer and 1,090 in ovarian cancer) or overall survival<br />

(n�464 and n�1,287). We used this integrative data analysis tool to<br />

validate the prognostic power <strong>of</strong> 37 ovarian cancer associated biomarkers<br />

identified in the literature. Of these, CA125 (p�3.7e-5, HR�1.4), CDKN1B<br />

(p�5.4e-5, HR�1.4), KLK6 (p�0.002, HR�0.79), IFNG (p�0.004,<br />

HR�0.81), P16 (p�0.02, HR�0.66) and BIRC5 (p�0.00017, HR�0.75)<br />

were associated with survival. Conclusions: We set up a global online<br />

biomarker validation platform to mine all available microarray data to<br />

assess the prognostic power <strong>of</strong> 22,277 genes in 2,472 breast and 1,390<br />

ovarian cancer patients. Registration-free online access at: http://<br />

www.kmplot.com/.<br />

10573 General Poster Session (Board #46F), Mon, 1:15 PM-5:15 PM<br />

FDG-PET and CT as early predictors <strong>of</strong> outcome in NSCLC patients treated<br />

with erlotinib. Presenting Author: Robert Iannone, Merck, North Wales, PA<br />

Background: Tumor metabolism as assessed by FDG-PET is an early<br />

biomarker for clinical benefit in oncology. This multi-national study<br />

evaluated associations <strong>of</strong> changes in mean standardized uptake values<br />

(SUVmean) <strong>of</strong> FDG and tumor burden with disease control rates (DCR:<br />

CR�PR�SD), PFS and OS in 48 NSCLC patients (EGFR mutants or<br />

favorable clinical/demographic characteristics) treated with erlotinib. The<br />

primary hypothesis was that changes in SUVmean at day 8 predicted DCR.<br />

Methods: Patients were scanned by FDG-PET (twice pre-therapy, 1 and 3<br />

weeks post-therapy) and CT (same as FDG-PET and every 6 weeks<br />

post-therapy until progression or death). Early response biomarkers included<br />

the decrease in SUVmean (DiSUV), sum <strong>of</strong> diameters (DiSOD) and<br />

sum <strong>of</strong> volumes (DiSOV). For the primary hypothesis, metabolic response<br />

(MR) was defined as a decrease <strong>of</strong> �2SD the variability between baseline<br />

PET scans (30%). Exploratory analyses <strong>of</strong> the biomarkers at various<br />

thresholds were performed. Results: The primary hypothesis was met: DCR<br />

was 34.6% (80% CI: 17.5%, 48.2%) higher in patients with a MR at day 8<br />

(p � 0.033); the association was stronger for day 22. The table compares<br />

hazard ratios derived from the �optimal� % decrease (smallest upper 80%<br />

confidence limit) over the 10% to 50% (by 10%) range for all biomarkers<br />

and time points, by PFS and OS. Conclusions: FDG-PET was predictive for<br />

clinical responses after 8 days <strong>of</strong> treatment. Comparing all biomarkers,<br />

DiSOV had stronger associations than both DiSUV and DiSOD with PFS or<br />

OS. For NSCLC treated with erlotinib in this clinical context, the decrease<br />

in CT-based tumor volume may be a better early predictor <strong>of</strong> clinical<br />

outcome than FDG-based SUVmean. Hazard ratio and the 80% confidence interval by change in biomarker and<br />

time.<br />

Time Outcome DiSOD DiSOV DiSUV<br />

Day 8 OS 0.41 (0.21,0.80) 0.18 (0.07,0.47) 0.47 (0.23,0.93)<br />

PFS 0.40 (0.25,0.65) 0.24 (0.14,0.39) 0.57 (0.34,0.93)<br />

Day 22 OS 0.39 (0.21,0.75) 0.23 (0.09,0.60) 0.45 (0.23,0.88)<br />

PFS 0.17 (0.10,0.30) 0.19 (0.12,0.32) 0.21 (0.09,0.46)<br />

Day 43 OS 0.13 (0.05,0.36) 0.10 (0.04,0.28)<br />

PFS 0.22 (0.12,0.41) 0.12 (0.06,0.24)<br />

Based on the �optimal� % decrease, over the 10% to 50% (by 10%) range, in<br />

biomarker which yielded the smallest, upper 80% confidence interval.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


674s Tumor Biology<br />

10574 General Poster Session (Board #46G), Mon, 1:15 PM-5:15 PM<br />

Correlation <strong>of</strong> ERCC1 expression on circulating tumor cells with progressionfree<br />

survival in metastatic non-small cell lung cancer patients treated with<br />

platinum-based chemotherapy. Presenting Author: Jonathan Riess, Stanford<br />

University School <strong>of</strong> Medicine, Stanford, CA<br />

Background: Biomarkers predicting efficacy <strong>of</strong> chemotherapy are highly<br />

desirable. Fiber array scanning technology (FAST) is a novel method <strong>of</strong><br />

detecting circulating tumor cells (CTCs) that does not employ an EpCam<br />

enrichment step. The purpose <strong>of</strong> this study was to use FAST to evaluate<br />

ERCC1 expression on CTCs to determine whether ERCC1 expression<br />

correlates with progression-free survival (PFS) in patients who received<br />

platinum-based chemotherapy for metastatic non-small-cell lung cancer<br />

(NSCLC). Methods: Peripheral blood from one hundred enrolled patients<br />

with metastatic NSCLC was collected by two institutions (Stanford Cancer<br />

Institute and Billings Clinic Cancer Center). FAST was used to identify<br />

individual CTCs on immun<strong>of</strong>luorescence by pancytokeratin antibodies.<br />

Nuclear localization <strong>of</strong> ERCC1 expression by immun<strong>of</strong>luorescence was<br />

quantified on individual CTCs. Total patient ERCC1 levels were determined<br />

from the average expression <strong>of</strong> all the CTCs in each patient sample.<br />

Fifty-seven <strong>of</strong> the one hundred patients enrolled received platinum chemotherapy.<br />

Seventeen <strong>of</strong> those fifty-seven patients (30%) had � 2 evaluable<br />

intact CTCs and were analyzed retrospectively. Linear regression (F-test)<br />

was used to evaluate the correlation between ERCC1 expression and PFS.<br />

Kaplan-Meier survival analysis (log-rank test) was used to compare PFS in<br />

patients with CTCs with no detectable ERCC1 expression versus patients<br />

with CTCs that expressed any level <strong>of</strong> ERCC1. Results: PFS decreased with<br />

increasing ERCC1 expression (P�0.04 F-test, linear regression). Lack <strong>of</strong><br />

ERCC1 expression was associated with longer PFS (266 days vs. 172 days,<br />

log-rank test P�0.02). The difference in survival was statistically significant<br />

with a hazard ratio <strong>of</strong> 4.20 (95% CI 1.25-14.1, p�0.02, log-rank<br />

test). Conclusions: In this small study, using FAST to isolate CTCs, low<br />

expression <strong>of</strong> ERCC1 on evaluable CTCs correlated with increased PFS in<br />

patients with metastatic NSCLC who received platinum chemotherapy. A<br />

larger, prospective study to validate these retrospective results is warranted.<br />

10576 General Poster Session (Board #47A), Mon, 1:15 PM-5:15 PM<br />

A gene expression pr<strong>of</strong>ile test that distinguishes ovarian from endometrial<br />

cancers. Presenting Author: Anita Lal, Pathwork Diagnostics, Redwood<br />

City, CA<br />

Background: The differential diagnosis between primary epithelial ovarian<br />

and endometrial cancers is <strong>of</strong>ten unresolved because the histologic<br />

subtypes <strong>of</strong> these two tumor types can have very similar histology and<br />

immunohistochemical appearance. Here we report the development and<br />

validation <strong>of</strong> a gene expression pr<strong>of</strong>ile (GEP) diagnostic test (Pathwork<br />

Tissue <strong>of</strong> Origin Endometrial Test) that distinguishes ovarian and endometrial<br />

cancers in formalin-fixed, paraffin-embedded (FFPE) specimens using<br />

a 316–gene classification model. Methods: The test was clinically validated<br />

in a blinded study using a pre-specified algorithm and microarray<br />

data files for 75 metastatic, poorly differentiated or undifferentiated<br />

primary FFPE tumor specimens that had either a known clinical diagnosis<br />

<strong>of</strong> ovarian or endometrial cancer and belonged to one <strong>of</strong> the 14 ovarian and<br />

endometrial WHO histologic subtypes on the test panel. Results: Classification<br />

biomarkers, empirically selected by machine learning, included<br />

several genes such as homeobox transcription factors and kallikrein<br />

peptidases with known function in the biology <strong>of</strong> ovarian and endometrial<br />

cancers. The Tissue <strong>of</strong> Origin Endometrial Test accurately identified the<br />

primary site for 94.7% (95% CI, 87% to 99%) <strong>of</strong> ovarian and endometrial<br />

cancers. Other measures <strong>of</strong> test performance include an area under the<br />

ROC curve <strong>of</strong> 0.997 and a diagnostic odds ratio <strong>of</strong> 406. Test performance<br />

did not change significantly when stratified by specimens from metastases<br />

(90.5%) or by poorly differentiated and undifferentiated primary tumors<br />

(96.3%). All stages <strong>of</strong> ovarian and endometrial cancers were included in<br />

the validation study and had agreements <strong>of</strong> 85% to 100% with the clinical<br />

diagnosis. In a precision study, concordance in test results was 100%.<br />

Reproducibility in test results between three laboratories had a concordance<br />

<strong>of</strong> 94.3%. Conclusions: The GEP test identified specific ovarian and<br />

endometrial cancer morphologies even when a clear distinction could not<br />

be made by histologic criteria. The GEP test can aid in resolving an<br />

important differential diagnostic question in gynecologic oncology and may<br />

impact clinical management <strong>of</strong> these patients as well as entry opportunities<br />

into clinical trials.<br />

10575 General Poster Session (Board #46H), Mon, 1:15 PM-5:15 PM<br />

Accuracy <strong>of</strong> microRNA-based classification <strong>of</strong> metastases <strong>of</strong> unknown<br />

primary. Presenting Author: Nicholas Pavlidis, Ioannina University Hospital,<br />

Ioannina, Greece<br />

Background: Identification <strong>of</strong> the tissue <strong>of</strong> origin <strong>of</strong> metastatic tumors is<br />

important for estimation <strong>of</strong> prognosis and patient management. Carcinoma<br />

<strong>of</strong> unknown primary (CUP) is not uncommon in oncology, representing<br />

3-5% <strong>of</strong> all newly found malignancies. We used a microarray-based test<br />

that uses the expression <strong>of</strong> 64 microRNAs to retrospectively evaluate<br />

tumors from CUP patients. Methods: A cohort <strong>of</strong> resected metastatic lesions<br />

from patients diagnosed with CUP was studied blindly on the microRNAbased<br />

test. The cohort included 93 samples (from 92 patients) with tissue<br />

adequate for the test. Eight samples failed due to inadequate RNA quality;<br />

85 samples (84 patients) were processed successfully. Test results were<br />

compared with clinical presentation including imaging, pathological data<br />

(histology and IHC) at initial CUP diagnosis and with clinical management<br />

and outcome data at the completion <strong>of</strong> each patient�s follow up. Results:<br />

The test results were fully concordant with the diagnosis based on all the<br />

clinical and pathological information available including follow-up and<br />

outcome in 92% <strong>of</strong> patients compared to ~70% agreement with the<br />

patients’ diagnosis at initial presentation, before additional data gathered<br />

throughout patient management. Two different metastases from the same<br />

patient yielded the same result. The microRNA test assigned a single tissue<br />

<strong>of</strong> origin for 51 patients and two tissues <strong>of</strong> origin in 33 patients, with the<br />

first being the more likely diagnosis. When comparing only the first (or<br />

single) diagnosis, a concordant level <strong>of</strong> 88% was achieved. Conclusions: In<br />

a retrospective, well studied cohort <strong>of</strong> metastases from CUP patients, a<br />

previously developed test based on the expression pr<strong>of</strong>ile <strong>of</strong> 64 microRNAs<br />

allowed accurate identification <strong>of</strong> tissue <strong>of</strong> origin in 92% <strong>of</strong> the cases. This<br />

study validates the high accuracy <strong>of</strong> the test on real CUP patients. The high<br />

concordance <strong>of</strong> the test results to the final tissue <strong>of</strong> origin diagnosis <strong>of</strong> the<br />

patient, even in cases where the initial diagnosis at CUP presentation was<br />

discordant, demonstrates the importance <strong>of</strong> the test in yielding additional<br />

data valuable for patient management at an early stage <strong>of</strong> the patient’s<br />

treatment plan.<br />

10577 General Poster Session (Board #47B), Mon, 1:15 PM-5:15 PM<br />

Use <strong>of</strong> early 18f-fluorodeoxyglucose-positron emission tomography to<br />

predict clinical outcome <strong>of</strong> patients with advanced non-small cell lung<br />

cancer on gefitinib treatment versus carboplatin plus paclitaxel. Presenting<br />

Author: Masaki Kanazu, National Hospital Organization Kinki-Chuo Chest<br />

Medical Center, Sakai, Japan<br />

Background: Early prediction <strong>of</strong> clinical efficacy is <strong>of</strong> great value in cancer<br />

patients in avoiding unnecessary toxicities and giving them another chance<br />

for different treatments. This study aimed to assess the 18F-fluorodeoxyglu cose-positron emission tomography (FDG-PET) at 3 days on treatment as an<br />

early predictor <strong>of</strong> clinical outcome in patients with advanced non-small cell<br />

lung cancer (NSCLC) treated with gefitinib and in those treated with<br />

cytotoxic chemotherapy. Methods: This study comprised two groups:<br />

patients with stage IIIB or IV NSCLC were treated with gefitinib (250mg)<br />

once daily (gefitinib group) or with carboplatin (AUC 6, day 1) plus<br />

paclitaxel (200mg/m2 , day 1) q21 days (CP group) according to the<br />

physicians’ choice. FDG-PET was performed before, 3 days on and 28 days<br />

on each treatment. Reduction <strong>of</strong> tumor FDG uptake was assessed by using<br />

standardized uptake value (SUV). Metabolic response was defined as<br />

reduction <strong>of</strong> FDG uptake in the tumor � 25% according to the criteria <strong>of</strong><br />

the European Organization for Research and Treatment <strong>of</strong> Cancer. Metabolic<br />

response was correlated with clinical outcomes. Results: This study<br />

included 38 patients: 19 in gefitinib group and 19 in CP group. Reduction<br />

<strong>of</strong> SUV at 3days on treatment preceded tumor shrinkage more closely in<br />

gefitinib group. Metabolic response was significantly correlated with longer<br />

progression-free survival (PFS) in gefitinib cohort (median PFS 15.8<br />

months [95% CI 13.2-18.4] vs. 3.7 months [95% CI 0.1-8.0], p �<br />

0.001), but not in CP group (median PFS 5.7 months vs. 2.9 months, p �<br />

0.054). Furthermore, metabolic response was significantly correlated with<br />

longer overall survival (OS) in gefitinib group (median OS 28.7 months<br />

[95%CI 23.5-33.9] vs. 9.8 months [95% CI 2.1-17.5], p � 0.009), but<br />

not in CP group (median OS 13.9 months vs. 10.5 months, p � 0.56). In<br />

multivariate analysis using Cox hazards models, metabolic response was a<br />

significant predictive factor <strong>of</strong> PFS and OS. Conclusions: Reduction <strong>of</strong> SUV<br />

levels on FDG-PET at 3 days on treatment may predict response and<br />

survival in gefitinib-treated patients with advanced NSCLC.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


10578 General Poster Session (Board #47C), Mon, 1:15 PM-5:15 PM<br />

Preoperative detection <strong>of</strong> circulating tumor cells in patients with colorectal<br />

carcinoma: Correlation with clinicopathologic variables and recurrence.<br />

Presenting Author: Hideyasu Sakihama, Gastroenterological Surgery 1,<br />

Hokkaido University Graduate School <strong>of</strong> Medicine, Sapporo, Japan<br />

Background: The role <strong>of</strong> circulating tumor cells (CTC) in the management <strong>of</strong><br />

colorectal cancer patients has not fully established. The aims <strong>of</strong> this study<br />

are to investigate the relationship between the presence <strong>of</strong> CTC with<br />

clinicopathological variables and recurrence by magnetic activated cell<br />

sorting (MACS) system. Methods: Peripheral blood samples were collected<br />

from 80 patients. Enrichment <strong>of</strong> CTC was performed by direct immunomagnetic<br />

labeling <strong>of</strong> EpCAM positive cells in peripheral blood. Subsequently,<br />

double immun<strong>of</strong>luorescence for cytokeratin and CD45 was performed to<br />

detect CTC. Peripheral blood samples from twenty healthy volunteers were<br />

used as controls. Results: Preoperative positive rate <strong>of</strong> CTC was 35% while<br />

specificity was 100%. No CTC was found in peripheral blood from healthy<br />

volunteers. No correlation was found between the presence <strong>of</strong> CTC and<br />

location <strong>of</strong> tumors, grade <strong>of</strong> differentiation, vessel invasion, lymph node<br />

metastasis or TNM stages. On the other hand, the depth <strong>of</strong> invasion (0% in<br />

Tis, 11.1% in T1, 18.2% in T2 and 34.7% in T3�T4, P�0.05) and tumor<br />

recurrence (28.2% for initial operation and 88.9% for reoperative surgery<br />

for tumor recurrence, P�0.001) closely correlated with the presence <strong>of</strong><br />

CTC. Preoperative positive rate <strong>of</strong> CTC among patients who have recurred<br />

postoperatively was 75%. Conclusions: Our results indicate that detections<br />

<strong>of</strong> CTCs correlate with the depth <strong>of</strong> invasion and tumor recurrence.<br />

Preoperative presence <strong>of</strong> CTCs might be a strong predictor for tumor<br />

recurrence.<br />

10580 General Poster Session (Board #47E), Mon, 1:15 PM-5:15 PM<br />

Defining a FISH chromosomal aneusomy panel for predicting lung cancer in<br />

the setting <strong>of</strong> CT-detected lung nodules. Presenting Author: Wilbur A.<br />

Franklin, University <strong>of</strong> Colorado Denver, Denver, CO<br />

Background: Early detection <strong>of</strong> lung cancer by CT is supported by the<br />

National Lung Screening Trial; but while sensitivity <strong>of</strong> CT is high, specificity<br />

is low. Biomarkers to predict the malignant nature <strong>of</strong> CT detected lung<br />

nodules would have great clinical utility. We previously found in a nested<br />

case-control study that a 4-target chromosomal aneusomy (CA) FISH panel<br />

exhibited sensitivity/specificity <strong>of</strong> 76%/88% in sputum samples <strong>of</strong> heavy<br />

smokers obtained within 18 months <strong>of</strong> lung cancer diagnosis. We hypothesized<br />

that CA-FISH may be an effective biomarker to assist with clinical<br />

decisions in the setting <strong>of</strong> CT detected lung nodules <strong>of</strong> indeterminate<br />

etiology and attempted to identify a more effective reagent. Methods:<br />

Homebrew probes encompassing genomic sequences <strong>of</strong> EGFR, NXK2-1,<br />

PIK3CA, MYC, BRF2, SOX2, PPMID, FGFR1 and the commercial reagent<br />

LSI D5S721/D5S23 (Abbott Molecular) were combined in 2-4-target FISH<br />

assays to investigate tissue copy number in early stage lung squamous cell<br />

carcinoma (SCC, N�19) and adenocarcinoma (AC, N�20). Logistic<br />

regression models were used to estimate predictive discrimination [sensitivity,<br />

specificity, area under the ROC curve (AUC)]. Results: Copy number<br />

gain was largely detected for all markers (mean range 3.39 - PPMID to 4.67<br />

- MYC). Mean copy number <strong>of</strong> PI3CA, BRF2, SOX2 and FGFR1 were<br />

significantly higher in SCC than AC, while NKX2 and MYC were marginally<br />

higher in AC than SCC. Gene amplification was detected for all 9 markers,<br />

most frequently for SOX2 and FGFR1 (6 and 5 cases) with significant<br />

overlap between FGFR1/BRF2 and SOX2/PIK3CA. Based on the AUC<br />

results and the existence <strong>of</strong> targeted inhibitors, the probe set EGFR/FGFR1/<br />

MYC/PIK3CA was selected as a candidate for further development as an<br />

adjunct to CT screening for early detection. For this selected set,the<br />

optimal cut<strong>of</strong>f based in the linear predictor from logistic regression yields a<br />

sensitivity and specificity <strong>of</strong> 0.85. Conclusions: A highly sensitive/specific<br />

CA-FISH probe set was identified which may well complement CT screening<br />

in the early diagnosis <strong>of</strong> lung cancer. This probe set will be tested in the<br />

setting <strong>of</strong> CT detected lung nodules. (Supported by LUNGevity and<br />

NCI-Lung Cancer SPORE grants).<br />

Tumor Biology<br />

675s<br />

10579 General Poster Session (Board #47D), Mon, 1:15 PM-5:15 PM<br />

Retrospective analysis <strong>of</strong> gene expression pr<strong>of</strong>iling and TTF-1 staining in<br />

cancer <strong>of</strong> unknown primary (CUP). Presenting Author: Wendy M. Chiang,<br />

Tufts Medical Center, Boston, MA<br />

Background: CUP involves extensive use <strong>of</strong> immunohistochemical (IHC)<br />

stains because no individual marker is highly both site specific or sensitive.<br />

IHC algorithms lack standardization and may even eliminate actual primary<br />

sites. Extensive validation studies with gene expression pr<strong>of</strong>iling (GEP) has<br />

shown it be a new diagnostic technique that further contributes tumor site<br />

location in CUP. Thyroid transcription factor-1 (TTF-1) IHC stain is<br />

commonly used to identify the pulmonary origin <strong>of</strong> CUP (particularly<br />

adenocarcinoma) and is <strong>of</strong>ten used to exclude lung primary in CUP<br />

patients. This study evaluates the utility <strong>of</strong> TTF-1 staining in lung primaries<br />

(specifically non-small cell lung carcinoma - NSCLC) <strong>of</strong> CUP to GEP<br />

testing. Methods: This retrospective study contains data obtained from a<br />

registry <strong>of</strong> physicians who received the GEP-based Tissue <strong>of</strong> Origin (TOO)<br />

test (Pathwork Diagnostic, Sunnyvale, CA) between 07/2009 and 12/2009<br />

on CUP cases. Sixty-six physicians contributed 111 TOO test cases. Only<br />

cases that had TTF-1 done were included for analysis and these were<br />

compared to TOO NSCLC results. Results: Out <strong>of</strong> 111 cases, there were 73<br />

analyzable TTF-1 results with 12 cases <strong>of</strong> NSCLC and 61 cases <strong>of</strong><br />

non-NSCLC by TOO (see TABLE). Assuming that the results <strong>of</strong> TOO testing<br />

accurately indicated the true primary site, the sensitivity and specificity <strong>of</strong><br />

TTF-1 was 50% and 90%, respectively. The false negative rate was 50%,<br />

indicating that half <strong>of</strong> the identified NSCLC cases in this series had<br />

negative TTF-1. On the other hand, 10% <strong>of</strong> the 61 cases with primaries<br />

other than NSCLC (none <strong>of</strong> whom had thyroid cancer), had positive TTF-1.<br />

The “false positive” TTF-1 cases comprised 4 ovarian, 1 breast and 1<br />

colorectal as the site <strong>of</strong> origin. Conclusions: TTF-1 has limited utility in<br />

identifying NSCLC in the setting <strong>of</strong> CUP. Half <strong>of</strong> NSCLCs identified by TOO<br />

testing had negative TTF-1, and 10% <strong>of</strong> non-lung primaries were TTF-1<br />

positive. Negative TTF-1 should not be used to exclude NSCLC in the<br />

workup <strong>of</strong> CUP.<br />

NSCLC by TOO Non- NSCLC by TOO Total<br />

TTF-1 stain positive 6 (TP) 6 (FP) 12<br />

TTF-1 stain negative 6 (FN) 55 (TN) 61<br />

Total 12 61 73<br />

Sensitivity 0.50<br />

Specificity 0.90<br />

False negative rate (1-sensitivity) 0.50<br />

False positive rate (1-specificity) 0.10<br />

10581 General Poster Session (Board #47F), Mon, 1:15 PM-5:15 PM<br />

89 Zr-bevacizumab PET imaging in metastatic renal cell carcinoma patients<br />

before and during antiangiogenic treatment. Presenting Author: Sjoukje<br />

Oosting, Department <strong>of</strong> Medical Oncology, University Medical Center<br />

Groningen, Groningen, Netherlands<br />

Background: Renal cell cancer (RCC) is characterized by high VEGF<br />

production leading to excessive angiogenesis. To visualize VEGF, we<br />

performed serial 89Zr-bevacizumab-PET scans before and during antiangiogenic<br />

treatment in RCC patients. Methods: Metastatic (m) RCC patients<br />

who received sunitinib (50 mg once daily, 4 out <strong>of</strong> 6 weeks) or bevacizumab<br />

(10 mg/kg every 2 weeks) plus interferon (IFN 3-9 MU 3x/week), underwent<br />

89Zr-bevacizumab-PET scans at baseline and after 2 and 6 weeks, and CT<br />

scans at baseline and every 3 months. Tracer uptake in tumor lesions was<br />

quantified with maximum Standardized Uptake Value (SUVmax). Relationship<br />

between baseline and � SUVmax and time to progression (TTP) was<br />

analyzed. Wilcoxon test was used to compare scans, Kaplan-Meier method<br />

for survival analysis. Results: 22 out <strong>of</strong> 26 patients were evaluable, 11 per<br />

treatment. On 89Zr-bevacizumab-PET, 131 out <strong>of</strong> 231 lesions � 10 mm<br />

(detection limit) were visible and 125 quantifiable. Mean SUVmax at<br />

baseline was 10.1 (SD 8.4; range 2.3 - 46.9). During bevacizumab/IFN<br />

treatment, SUVmax consistently decreased (mean decrease 47.0% 95% CI<br />

39.1-54.9, P�0.0001) at 2 weeks with a further decrease <strong>of</strong> 9.7% (95%<br />

CI 0.86-18.5, P�0.016) at 6 weeks. After 2 weeks sunitinib, there was<br />

only a modest decrease in mean SUVmax (14.6%, 95% CI 1.57-27.63,<br />

P�0.0064) with a wide range (-80.4% to �269.9%) and an overshoot <strong>of</strong><br />

84.4% (95% CI 47.8-120.9, P�0.0001) after 2 drug free weeks. TTP was<br />

longer in (n�15) patients with baseline SUVmax � 11.1 (highest normal<br />

tissue uptake) in the 3 most intense lesions than in those with a lower value<br />

(median 89.7 vs 22.8 weeks, HR 0.16, 95% CI 0.04 - 0.70). TTP was<br />

longer in patients (n�11) with an absolute � SUVmax �6.00 in the most<br />

intense lesion at 2 weeks (HR 0.25, 95% CI 0.06-0.98). Conclusions:<br />

89Zr-bevacizumab-PET visualizes tumor lesions in mRCC patients. Different<br />

changes in tumor tracer uptake after start <strong>of</strong> bevacizumab/IFN versus<br />

sunitinib indicate that these drugs induce different angiogenic responses.<br />

High baseline SUVmax and large change in SUVmax corresponded with<br />

longer TTP, suggesting that 89Zr-bevacizumab-PET may help to identify<br />

patients who benefit the most from antiangiogenic treatment.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


676s Tumor Biology<br />

10582 General Poster Session (Board #47G), Mon, 1:15 PM-5:15 PM<br />

Antigens diagnoses in paraneoplastic neurological syndromes. Presenting<br />

Author: Halina Rudnicka, Maria Sklodowska-Curie Memorial Cancer Center<br />

and Institute <strong>of</strong> Oncology, Warsaw, Poland<br />

Background: At present, paraneoplasmatic neurological syndroms (PNS)<br />

are thought to be autoimmunological disorders. Neurological symptoms in<br />

PNS result from the disturbance <strong>of</strong> the nervous system, with a participation<br />

<strong>of</strong> the immune response triggered by the aberrant expression <strong>of</strong> the<br />

so-called onconeuronal antigens <strong>of</strong> a tumour. Very important for PNS<br />

diagnostics is the investigation <strong>of</strong> antineuronal antibodies (ANA) in the<br />

serum. Different types <strong>of</strong> ANA are frequently associated with specific<br />

tumors and with specific neurological syndromes.Investigations <strong>of</strong> serum<br />

ANA in patients with breast cancer with the symptoms <strong>of</strong> PNS. Methods:<br />

Investigations <strong>of</strong> ANA in serum were performed with the indirect immun<strong>of</strong>luorescence<br />

test but the type <strong>of</strong> ANA was analyzed by the Western Blot<br />

(WB) method . The presence <strong>of</strong> these antibodies which are included to the<br />

group <strong>of</strong> “the so called well characterized” is believed to be a definite (95 –<br />

100%) determinant <strong>of</strong> the PNS diagnosis. Results: We examined 147<br />

patients with breast cancer suspected to have PNS, hospitalized at Breast<br />

Cancer Clinic <strong>of</strong> the Institute <strong>of</strong> Oncology in Warsaw. The immun<strong>of</strong>luorescence<br />

test was positive in 92 patients, however in 24 patients the WB<br />

occurrence <strong>of</strong> “the well characterized” antibodies was found. In this group<br />

we observed: in 4 patients the cerebellar degeneration (anti-Yo, anti-Ma2);<br />

in 6 patients brainstem encephalitis and encephalomyelitis combined with<br />

sensory neuropathy (anti-Ma2, anti-CV2, anti-Yo, anti-Hu, anti-Ri); 4<br />

myastenic syndrome (anti-Hu,anti-Ri,anti-Ma2/Ta); 5 sensory neuropathy<br />

(anti-Hu, anti-Yo, anti-Ma2, anti-Ri); and in single cases we also observed<br />

polyneuropathy: Guillain-Barre (Ma2): Miller-Fisher syndrom (anti-Ma2/<br />

Ta); cancer-associated retinopathy (anti-Yo); dermatomyositis (anti-Ri) and<br />

scleroderma (anti-Yo). Conclusions: Our analysis indicates that the investigation<br />

<strong>of</strong> the presence <strong>of</strong> ANA in the serum is the important in the diagnosis<br />

<strong>of</strong> PNS in patients with breast cancer. The types <strong>of</strong> ANA correspond to the<br />

individual paraneoplastic neurological syndromes which opens the proper<br />

methods <strong>of</strong> treatment in PNS.<br />

10584 General Poster Session (Board #48A), Mon, 1:15 PM-5:15 PM<br />

Estimation <strong>of</strong> expected survival time using gene expression pr<strong>of</strong>iling for<br />

tumor site origin. Presenting Author: William David Henner, Pathwork<br />

Diagnostics, Redwood City, CA<br />

Background: Expected survival is an important factor in treatment selection<br />

and patient decision making. Gene expression pr<strong>of</strong>iling for tissue <strong>of</strong> origin<br />

(GEP TOO) can be used to classify metastatic and poorly-differentiated<br />

tumors according to the most likely primary site for difficult-to-diagnose<br />

tumors. The Pathwork Tissue <strong>of</strong> Origin Test, (Redwood City, CA) generates<br />

Similarity Scores (SS), one for each <strong>of</strong> the 15 tissues on the Tissue <strong>of</strong> Origin<br />

Test Panel and sum to 100. The highest SS has been validated as the most<br />

likely tissue <strong>of</strong> origin in a large validation study (Pillai et al. 2010). This<br />

abstract reports the relationship between the highest SS for a sample and<br />

patient survival. Methods: Two cohorts were analyzed in IRB-approved<br />

studies. Cohort 1 consisted <strong>of</strong> 45 patients with carcinoma <strong>of</strong> unknown<br />

primary treated empirically with therapy for carcinoma <strong>of</strong> unknown primary<br />

with known outcome data including survival. Cohort 2 consisted <strong>of</strong> 107<br />

patients receiving the TOO test in routine clinical practice and included in a<br />

decision impact registry study. The relationship between the highest SS<br />

generated by the TOO test was examined using parametric (Gamma) and<br />

non-parametric (Cox Proportional Hazards) regression models with covariates<br />

<strong>of</strong> age, gender and median survival <strong>of</strong> the cancer type indicated.<br />

Logrank tests were performed to compare survival in patients with SS above<br />

or below 50 in each cohort. Results: The highest SS generated by the TOO<br />

Test was strongly and positively correlated with patient survival in both<br />

Cohort 1 (p � 0.0093, Cox PH) and in Cohort 2 (p�0.0045, Gamma). In<br />

Cohort 1, survival for patients with a SS � 50 was 5.9 mos and for patients<br />

with a SS � 50 was 9.7 mos (p�0.03, logrank). In Cohort 2, survival for<br />

patients with a SS � 50 was 10.6 mos and 22.6 mos for patients with a SS<br />

� 50 (p�0.0073, logrank). Conclusions: The Similarity Scores generated<br />

by GEP TOO Test have been validated as an aid to diagnosis for<br />

difficult-to-diagnose malignancies. Two independent studies <strong>of</strong> this GEP<br />

TOO Test demonstrated a strong positive relationship between the highest<br />

SS and survival. This has the potential to expand the clinical utility <strong>of</strong> this<br />

test beyond diagnosis to include prognosis. Further studies to examine this<br />

question are underway.<br />

10583 General Poster Session (Board #47H), Mon, 1:15 PM-5:15 PM<br />

Antagonistic activity <strong>of</strong> acolbifene, fulvestrant, tamoxifen, and raloxifene<br />

on cancer-associated genes in the mouse mammary gland. Presenting<br />

Author: Ezequiel Calvo, Molecular Endocrinology, Oncology and Human<br />

Genomics Research Center, Laval University and Laval University Hospital<br />

Research Center, Quebec City, QC, Canada<br />

Background: The efficacy and exceptionally good tolerance <strong>of</strong> estrogen<br />

blockade in the treatment <strong>of</strong> breast cancer is well recognized. Acolbifene<br />

(ACOL) is a novel and unique SERM completely free <strong>of</strong> estrogen-like<br />

activity in both the mammary gland and uterus. To better understand the<br />

specificity <strong>of</strong> ACOL, we have investigated its effect on the expression <strong>of</strong> a<br />

set <strong>of</strong> genes modulated by estradiol (E2) in the mouse mammary gland.<br />

Methods: ACOL, tamoxifen (TAM), raloxifene (RALOX) and fulvestrant<br />

(FULV) were administered (0.01 mg/mouse; sc) to ovariectomized (OVX)<br />

mice or to OVX mice simultaneously treated with E2 (0.05 �g/mouse; single<br />

sc injection). Microarray screening followed by Q_RTPCR was used to<br />

identify a reproducible set <strong>of</strong> E2 responsive genes. Results: From 128 genes<br />

significantly modulated by E2, 108 genes were up-regulated and 20 were<br />

down-regulated. Forty-nine <strong>of</strong> these genes were associated with tumorigenesis<br />

while 22 are known to be associated with breast cancer. This set <strong>of</strong> 49<br />

genes were used to determine the specificity <strong>of</strong> ACOL compared to another<br />

pure antiestrogen in the mammary gland and uterus, namely FULV, as well<br />

as to the mixed estrogen antagonists/agonists TAM and RALOX, in their<br />

ability to block the effect <strong>of</strong> E2. Efficacy <strong>of</strong> reversal <strong>of</strong> the effect <strong>of</strong> E2 was<br />

94%, 63%, 45% and 90% for ACOL, FULV, TAM and RALOX, respectively.<br />

The overlap between all treatments was 30.6% (15/49). ACOL reversed the<br />

effect <strong>of</strong> E2 on 42 <strong>of</strong> the 49 (85.7%) cancer-related genes. Between the<br />

genes up-regulated by E2 and reversed by ACOL, seven are considered as<br />

prognostic markers in breast cancer, namely Fgfr3, Fos12, Junb, Jdp2,<br />

Gdf15, Greb1 and Tgm2. On the other hand, two genes down-regulated by<br />

E2, namely Foxa1 and Fgfr2, were restored by ACOL. Conclusions: Taken<br />

together, these data <strong>of</strong>fer new information for a better understanding <strong>of</strong> the<br />

previously demonstrated potent tumoricidal action <strong>of</strong> ACOL in human<br />

breast cancer xenografts. The data also suggest, under the tested conditions,<br />

superiority <strong>of</strong> ACOL over TAM and the other compounds to reverse the<br />

effect <strong>of</strong> E2 on specific gene expression, thus supporting the interest <strong>of</strong> this<br />

new 3rd generation SERM for the hormonal therapy and prevention <strong>of</strong> breast<br />

cancer.<br />

10585 General Poster Session (Board #48B), Mon, 1:15 PM-5:15 PM<br />

Novel malignant-mesothelioma-associated antigens (Gene-X and THBS-2)<br />

in the diagnosis <strong>of</strong> malignant pleural mesothelioma (MPM). Presenting<br />

Author: Fumihiro Tanaka, University <strong>of</strong> Occupational and Environmental<br />

Health, Kitakyusyu, Japan<br />

Background: Malignant pleural mesothelioma (MPM) is a highly aggressive<br />

malignant tumor <strong>of</strong> the pleura associated with asbestos exposure, and its<br />

diagnosis is usually difficult at early stage. We identified novel mesothelioma-related<br />

antigens, Gene-X and thrombospondin-2 (THBS-2), recognized<br />

by tumor-infiltrating B cells (Cancer Sci 2009), but the clinical<br />

significance in the diagnosis <strong>of</strong> MPM remains unclear. Methods: A total <strong>of</strong><br />

120 patients, who presented with a suspicion <strong>of</strong> MPM and received pleural<br />

biopsy, were reviewed; 97 patients were finally diagnosed with MPM and<br />

27 were with non-malignant diseases (NM). The antibody-titers against<br />

Gene-X and THBS-2 in the sera were measured by ELISA method. Results:<br />

The serum antibody-titer against THBS-2 was significantly higher in MPM<br />

patients than in NM (P�0.01), but there was no difference in the serum<br />

antibody-titer against Gene-X (Table). The receiver operating characteristic<br />

(ROC) curve analysis showed a significant diagnostic value <strong>of</strong> serum<br />

antibody-titer against THBS-2 with the area-under curve <strong>of</strong> 0.886 (95% CI,<br />

0.797 - 0.975; P�0.001) in discrimination <strong>of</strong> MPM from NM diseases; the<br />

sensitivity and specificity, when the cut-<strong>of</strong>f value was 0.08, were 72.2%<br />

and 95.5%, respectively. Conclusions: The serum antibody-titer against<br />

THBS-2 can be a useful non-invasive marker in the diagnosis <strong>of</strong> MPM.<br />

Antigens<br />

MPM<br />

(n�97)<br />

Non-malignant<br />

(n�23)<br />

p value<br />

(Mann-Whitney)<br />

Gene-X<br />

(mean) 0.49 0.48 p�0.997<br />

(median) 0.39 0.36<br />

THBS-2<br />

(mean) 0.11 0.02 p�0.001<br />

(median) 0.10 0.00<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


10586 General Poster Session (Board #48C), Mon, 1:15 PM-5:15 PM<br />

Multiplex TaqMan assays for a 7-gene prognostic immune response score to<br />

differentiate risk among women with ER-negative breast cancer. Presenting<br />

Author: John J. Sninsky, Celera Corporation, Alameda, CA<br />

Background: Teschendorff et al. (Breast Cancer Res 2008) reported a<br />

7-gene, immune response-enriched gene expression classifier using microarrays<br />

that was prognostic in women with ER-negative breast cancer. To<br />

facilitate further evaluation <strong>of</strong> the prognostic value <strong>of</strong> a classifier in archival<br />

fixed tumor tissue, we developed two multiplex TaqMan assays to quantify<br />

the expression levels <strong>of</strong> these genes, then trained and validated a 7-gene<br />

immune response score (IRS). Methods: RNA was extracted from FFPE<br />

sections <strong>of</strong> 185 ER-negative breast cancer patient samples from Mayo<br />

Clinic (n�81), Guy’s Hospital (n�50) and California Pacific Medical<br />

Center (CPMC) (n�27). Patients (N� and N-) did not receive adjuvant<br />

therapy and were followed for a minimum <strong>of</strong> 5 years. Amplification <strong>of</strong> i)<br />

LY9, TNFRSF17, HLA-F, and IGLC2 and ii) XCL2, SPP1, C1QA and two<br />

reference genes NUP214 and PPIG were carried out in two multiplex<br />

RT-PCR TaqMan reactions and relative expression levels were determined<br />

using the DDCT method. The training set, comprised <strong>of</strong> the Mayo Clinic<br />

samples, was used to develop an IRS by using a summation <strong>of</strong> expression<br />

levels <strong>of</strong> the 7 genes. Association <strong>of</strong> the IRS with distant metastasis was<br />

assessed using a Cox proportional hazards model. The score was subsequently<br />

tested in a validation set (Guy’s � CPMC). Results: The amplification<br />

efficiency <strong>of</strong> each gene within the multiplex reactions was comparable<br />

to singleplex reactions. Duplicate amplifications showed excellent assay<br />

reproducibility and sensitivity (R2�0.98). The hazard ratio for a single<br />

standard deviation decrease in the IRS was 1.45 (95% CI 0.99-2.12,<br />

p�0.056) in the training set and 2.18 (1.32-3.61, p�0.002) in the<br />

validation set. ROC curves <strong>of</strong> continuous IRS to predict distant metastasis<br />

within 5 years had AUC <strong>of</strong> 0.74, 95% CI � 0.60-0.88. Conclusions: Robust<br />

multiplex TaqMan assays for FFPE tissue extracted RNA were developed for<br />

a 7-gene IRS. Increases in risk are associated with decreases in the IRS.<br />

Immune related gene expression clearly plays an important role in the<br />

spectrum <strong>of</strong> risk <strong>of</strong> ER-negative tumors. Additional signatures are in<br />

development that further support this conclusion.<br />

10588 General Poster Session (Board #48E), Mon, 1:15 PM-5:15 PM<br />

Multisite validation <strong>of</strong> a 92-gene molecular classifier: Diagnostic accuracy<br />

and clinical utility in metastatic cancer. Presenting Author: Catherine A.<br />

Schnabel, bioTheranostics, Inc., San Diego, CA<br />

Background: Accurate tumor classification is increasingly critical to individualized<br />

care as patient outcomes improve with use <strong>of</strong> targeted cancer<br />

therapies and predictive biomarkers. In metastatic cancers <strong>of</strong> uncertain or<br />

unknown origin, identification <strong>of</strong> a primary site remains equivocal in a<br />

significant number <strong>of</strong> cases. Gene expression signatures may improve<br />

accuracy and specificity <strong>of</strong> tumor classification, however, large-scale,<br />

blinded validation studies that demonstrate stable performance in metastatic<br />

and poorly differentiated cases are currently lacking. Methods: Seven<br />

hundred and ninety cases (51% female, 44% metastatic, 63% grade 2 and<br />

3, 14% limited tissue specimens) representing 28 tumor types and 50<br />

subtypes were processed and adjudicated between Mayo Clinic, UCLA and<br />

Massachusetts General Hospital. Blinded FFPE tumor sections were<br />

submitted and tested using a 92-gene RT-PCR assay (CancerTYPE ID,<br />

bioTheranostics Inc.). Molecular predictions were evaluated for concordance<br />

with the reference diagnosis; diagnostic accuracy between clinical<br />

subsets was compared. Results: The 92-gene assay demonstrated overall<br />

sensitivities <strong>of</strong> 87% for tumor classification and 82% for subtyping.<br />

Forty-seven (5.9%) cases were considered unclassifiable. Comparative<br />

analysis <strong>of</strong> performance between metastatic (n�329) and primary (n�414)<br />

tumors showed no statistically significant difference in accuracy (85% vs<br />

88%, p�0.16). Similarly, no significant decreases in performance were<br />

observed across histological grades (p�0.58) and when comparing limited<br />

tissue to excisional biopsy specimens (p�0.16). Strong precision was<br />

demonstrated for accurate identification <strong>of</strong> a primary tumor in tissues<br />

biopsied from common metastatic sites, reported as positive predictive<br />

values <strong>of</strong> 100% for lung, brain and peritoneum, 92% for ovary and 80% for<br />

liver. Conclusions: Results from this multisite, blinded study validate the<br />

diagnostic accuracy <strong>of</strong> the 92-gene assay for classifying a diverse set <strong>of</strong><br />

tumors, and support its clinical utility in the diagnosis <strong>of</strong> metastatic tumors<br />

to determine tissue <strong>of</strong> origin, and in the differential diagnosis <strong>of</strong> metastatic<br />

vs new primary disease.<br />

Tumor Biology<br />

677s<br />

10587 General Poster Session (Board #48D), Mon, 1:15 PM-5:15 PM<br />

Specific and sensitive detection <strong>of</strong> BRAF and KRAS mutations in clinical<br />

samples. Presenting Author: Vanessa Kelchner, Swift Biosciences, Inc,<br />

Ann Arbor, MI<br />

Background: The development <strong>of</strong> highly sensitive and specific genotyping<br />

assays that are suitable for clinical research and molecular diagnostics<br />

opens new opportunities for the detection, assessment, and research <strong>of</strong><br />

cancer. Swift Biosciences has developed myT Primer qPCR technology<br />

which has unique structural and thermodynamic properties that enables<br />

highly sensitive mismatch discrimination. The myT Primer reagents can<br />

detect 1% mutant BRAF or KRAS present in a background <strong>of</strong> 103 wild-type<br />

genomic DNA copies with no background signal from wild-type. A separate<br />

ultrasensitive BRAF format has been developed which enables the detection<br />

<strong>of</strong> a single mutant BRAF copy in � 104 wild-type genomic DNA copies,<br />

representing 0.01% detection. Methods: To evaluate the performance on<br />

clinical samples, DNA was prepared from 26 melanoma, 40 colorectal, and<br />

49 lung tumor fresh frozen and FFPE samples, and their corresponding<br />

normal adjacent samples. qPCR was performed using standard cycling<br />

conditions in single tube format with allele-specific myT primers to assess<br />

two BRAF alleles or seven KRAS alleles. Performance <strong>of</strong> the assay was<br />

evaluated on the ABI 7500, CFX96, and Roche LightCycler. A subset <strong>of</strong><br />

samples were selected for validation by Sanger sequencing. Results:<br />

Mutations <strong>of</strong> BRAF in melanoma, colorectal, and lung tumors were<br />

observed to be 46%, 5%, and 2%, in concordance with published data for<br />

the frequency <strong>of</strong> BRAF mutations. Mutations <strong>of</strong> KRAS in colorectal cancer<br />

were observed to be in 17/40, or 42% <strong>of</strong> samples, consistent with<br />

published reports <strong>of</strong> KRAS in colorectal cancer. 34% <strong>of</strong> lung cancer<br />

samples had a KRAS mutation, also consistent with available statistics.<br />

Mutation status was confirmed with Sanger sequencing in the subset <strong>of</strong><br />

selected samples. Conclusions: The extreme selectivity <strong>of</strong> the myT BRAF<br />

Primers makes them well suited for genotyping <strong>of</strong> conventional FFPE and<br />

frozen tissue samples, whereas the ultrasensitive format is ideal for use<br />

with difficult samples such as needle biopsies, CTCs, and serum. The<br />

extreme selectivity <strong>of</strong> these primers results in a definitive Yes/No answer<br />

and eliminates the need to use a delta Ct method.<br />

10589 General Poster Session (Board #48F), Mon, 1:15 PM-5:15 PM<br />

A pilot study on the clinical value <strong>of</strong> 18F-sodium fluoride PET/CT in<br />

advanced prostate cancer. Presenting Author: Joseph W. Kim, Medical<br />

Oncology Branch, National Cancer Institute, National Institutes <strong>of</strong> Health,<br />

Bethesda, MD<br />

Background: We evaluated the clinical utility <strong>of</strong> 18F-sodium fluoride PET/CT<br />

bone scan ( 18F-NaF) in the detection <strong>of</strong> bone metastases in patients (pts)<br />

with prostate cancer in comparison with Technetium-99m MDP bone scan<br />

(TcBS). Methods: In a prospective study, from October 2010-December<br />

2011, 30 prostate cancer pts (ages 51-79), 21 with known bone metastases<br />

and 9 without known bone metastases, had18F-NaF and a TcBS<br />

performed. Abnormal foci <strong>of</strong> uptake on both TcBS and 18F-NaFwere classified as benign, malignant or indeterminate. Benign lesions included<br />

uptake in the joints and linear uptake at the endplates <strong>of</strong> the vertebral<br />

bodies consistent with degenerative changes. Malignant uptake on 18F-NaF scans was confirmed by characteristic osteoblastic features on CT. All TcBS<br />

and 18F-NaF were reviewed by an experienced nuclear medicine physician.<br />

For the patient-based analysis, scan results were categorized as positive<br />

(POS) � any malignant lesion; indeterminate (IND) � not distinctly<br />

malignant or benign; negative (NEG) � benign lesions only. Results: In the<br />

lesion-based analysis, 21 <strong>of</strong> 30 (70%) pts had more malignant lesions<br />

identified on 18F-NaF than on TcBS. The mean number <strong>of</strong> additional<br />

malignant lesions per patient on 18F-NaF vs TcBS was 4. Eight <strong>of</strong> the 30 pts<br />

had same number <strong>of</strong> malignant lesions identified in both studies. One <strong>of</strong> 30<br />

pts had one less malignant lesion identified on 18F-NaF than on TcBS. CT<br />

correlation by 18F-NaF PET/CT <strong>of</strong> this particular lesion did not confirm<br />

osteoblastic feature. Malignant lesion distribution on 18F-NaF included:<br />

spine (28%), thorax (26%), pelvis (24%), long bones (13%) and skull<br />

(10%). In the patient-based analysis, 24 pts (80%) were POS by 18F-NaF, <strong>of</strong> whom 14 pts were POS, 8 were IND, and 2 were NEG by corresponding<br />

TcBS; in the 4 pts with NEG 18F-NaF, zero were POS, 2 were IND and 2<br />

were NEG by corresponding TcBS. Conclusions: 18F-NaF identified more<br />

malignant lesions than TcBS. 18F-NaF may also add useful information in<br />

the management <strong>of</strong> advanced prostate cancer pts with and without known<br />

bone metastases.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


678s Tumor Biology<br />

10590 General Poster Session (Board #48G), Mon, 1:15 PM-5:15 PM<br />

Next-generation sequencing (NGS) to identify actionable genomic changes<br />

in common and rare solid tumors: The FMI experience with the initial 50<br />

consecutive patients. Presenting Author: Gary A. Palmer, Foundation<br />

Medicine, Cambridge, MA<br />

Background: Solid tumor (ST) oncology has been transformed by the linkage<br />

<strong>of</strong> genomic changes with targeted therapeutics. Unfortunately, most STs<br />

still have no alteration detected with currently available assays. Thus, more<br />

informative testing platforms are needed to analyze genomic changes in<br />

FFPE biopsy samples <strong>of</strong>ten limited by tumor size, heterogeneity and ploidy.<br />

Methods: We reviewed the genomic pr<strong>of</strong>iles <strong>of</strong> the first 50 clinical<br />

specimens received by our CLIA lab (Foundation Medicine) and analyzed by<br />

our NGS assay (Ross J. ASCO 2011). 182 genes known altered in STs and<br />

14 genes known rearranged in STs were included. Genomic alterations<br />

were categorized as “actionable” if linked to an approved therapy in the ST<br />

under study or another ST (e.g. ALK rearrangement in lung or breast cancer,<br />

respectively), a known or suspected contraindication to a given therapy<br />

(e.g. KRAS G12D in colorectal cancer) or a clinical trial linked to the<br />

alteration (e.g. mTOR inhibition in a tumor with STK11 loss). Results: Fifty<br />

tumor specimens were received from 50 patients (median age 54,<br />

F/M�30/20) representing 35 sites. Two samples failed analysis and 6 had<br />

no detectable genomic alteration. Among 16 primary tumor types, lung<br />

(n�15), colorectal (n�7), sarcoma (n�5), breast and esophageal/gastric<br />

(n�4 each) were most common. We reported 135 genomic alterations<br />

(mean 2.8, range 0-6) unequivocally involved in oncogenesis and 69<br />

actionable alterations. At most, 29 <strong>of</strong> these (42%) would have been<br />

detected by current assays (138% increase with NGS). Thirty seven<br />

patients had one or more actionable alterations (range 1-4) detected by<br />

NGS (74%, 95% CI 60-85%) versus only 21 (42%, 95% CI 28-57%) in<br />

whom changes would have been detected by current assays. Conclusions:<br />

Use <strong>of</strong> a broad, comprehensive NGS assay identifies an unprecedented<br />

number <strong>of</strong> actionable genomic alterations across a variety <strong>of</strong> STs from<br />

routine FFPE samples. This assay can serve as a paradigm for improving<br />

access to approved therapies in STs, minimizing use <strong>of</strong> ineffective<br />

therapies and enhancing enrollment in rationally chosen trials. Ongoing<br />

and planned trials will study the impact on several efficacy endpoints.<br />

10592 General Poster Session (Board #49A), Mon, 1:15 PM-5:15 PM<br />

Genomic pr<strong>of</strong>iling <strong>of</strong> non-small cell lung cancer (NSCLC) for personalized<br />

targeted therapy using CT-guided transthoracic needle biopsy (TTNB).<br />

Presenting Author: Ritu R. Gill, Brigham and Women’s Hospital, Harvard<br />

Medical School, Boston, MA<br />

Background: Genomic pr<strong>of</strong>iling for personalized targeted therapy is emerging<br />

for NSCLC. DFCI introduced systematic testing for mutations in BRAF,<br />

HER2, PIK3CA and ALK translocations in addition to EGFR and KRAS in<br />

July 2009 as part <strong>of</strong> a prospective study. We report the utility, efficacy and<br />

safety <strong>of</strong> CT guided TTNB in this cohort. Methods: Patients with stage IV or<br />

relapsed NSCLC seen at the DFCI were referred to BWH for CT guided TTNB<br />

<strong>of</strong> their tumors to identify driver mutations prior to starting therapy.<br />

Pathology specimens were dissected and analyzed by PCR-Sanger sequencing<br />

for mutations in selected exons <strong>of</strong> EGFR, KRAS, BRAF, PIK3CA and<br />

HER2. ALK rearrangements were detected with fluorescence in-situ hybridization<br />

(FISH). Testing was performed after the pathologist deemed that<br />

the tissue was adequate. Complications such as pneumothorax and<br />

hemorrhage were recorded. Admission rates were also recorded. Results:<br />

Between 7/1/2009 and 1/09/2011, 81 patients underwent TTNB. The<br />

median age was 63 years. 54 (67%) were female, 66 (88%) were<br />

former/current smokers and 58(72%) had stage IIIB/IV disease. 64(79%)<br />

patients had sufficient tissue on core biopsies for genomic pr<strong>of</strong>iling, 4 (6%)<br />

<strong>of</strong> the 64 patients failed analysis for ALK rearrangements due to less than<br />

50 tumor cells on the hybridized slide. The number <strong>of</strong> samples obtained<br />

ranged from 1-5 (2 cm 18-20 (G)). Lesions biopsied ranged in size from<br />

1.2–8.9 cm. Mutations were identified in 38/81 (46.9%) patients (EGFR:<br />

18; KRAS: 17; ALK: 2;PIK3CA: 1). 23(28.3%) had pneumothoraces<br />

15(�10%), 5 (10-30%) and 3(�30%). 6 (7%) patients needed chest<br />

tubes. 9 (11%) were admitted post procedure for observation (8 for (24hrs)<br />

and 1 (72hrs). 19(23%) (18 grade1; 1 grade 2) had intra-parenchymal<br />

hemorrhage. A higher rate <strong>of</strong> pneumothorax was observed with the 18<br />

gauge needles (p �.05). 15 <strong>of</strong> 20 (75%) patients with EGFR, HER2, BRAF<br />

or ALK alterations were treated with molecularly targeted therapy based on<br />

their genetic alteration. Conclusions: CT guided TTNB is a feasible, safe and<br />

efficacious technique for genomic pr<strong>of</strong>iling for targeted therapy and<br />

enables the acquisition <strong>of</strong> sufficient tissue for gene mutation analyses.<br />

10591 General Poster Session (Board #48H), Mon, 1:15 PM-5:15 PM<br />

Combining semiconductor-based sequencing with amplicon-based cancer<br />

gene panels: A rapid next-gen approach to clinical cancer genotyping.<br />

Presenting Author: Christopher L. Corless, Knight Diagnostic Laboratories,<br />

Oregon Health & Science University, Portland, OR<br />

Background: Bringing next-gen sequencing into clinical (CLIA-licensed)<br />

laboratories is an important step in the advancement <strong>of</strong> personalized<br />

cancer care. We have validated a new sequencing approach on the Ion<br />

Torrent (IT) PGM using the AmpliSeq Cancer Panel, which covers hotspot<br />

regions across 46 commonly mutated cancer genes. Methods: The AmpliSeq<br />

panel is comprised <strong>of</strong> 190 primer pairs that are co-amplified in a<br />

single tube to generate amplicons for sequencing. In our testing only 10ng<br />

<strong>of</strong> input DNA was used. Initial PCR was for 20 cycles, after which the<br />

amplicons were ligated with sequencing/barcode adapters, amplified for an<br />

additional 7 cycles, and then subjected to emulsion PCR. The resulting<br />

nanospheres were sequenced on an IT 316 chip. Results: We sequenced 44<br />

samples <strong>of</strong> FFPE-derived tumor DNA that were previously genotyped on a<br />

mass spectroscopy (MS)-based panel. Samples were barcoded and sequenced<br />

in batches <strong>of</strong> 4, yielding an average <strong>of</strong> 2034 reads per amplicon<br />

(range: 95-5162) and an average read length <strong>of</strong> 76bp. Overall, 95.4% <strong>of</strong><br />

reads were on target, and 79% <strong>of</strong> reads were AQ20 or better; 95% <strong>of</strong> the<br />

190 amplicons had over 500 reads. All 42 known point mutations were<br />

accurately identified by the variant caller s<strong>of</strong>tware. Seventeen in/dels from<br />

4 to 63 bp in length were at least partially visible upon manual inspection<br />

<strong>of</strong> the read alignments, but some were not accurately called by the<br />

s<strong>of</strong>tware. In addition, 22 new mutations were identified in gene regions not<br />

covered on our MS-based panel. We demonstrated sensitivity to the level <strong>of</strong><br />

5% mutant allele, and the correlation between allelic ratios measured by<br />

MS and IT sequencing was excellent (r2�0.81). Two DNA samples from<br />

laser-captured tumor worked well with the AmpliSeq Panel. Conclusions:<br />

Combining solid-state sequencing with a highly multiplexed PCR method<br />

for library construction is a rapid (48 hr) approach for next-gen sequencing<br />

<strong>of</strong> clinical cancer samples. The process is highly scalable and larger,<br />

cancer-specific amplicon panels are in development. Automated identification<br />

<strong>of</strong> in/dels remains a challenge in next-gen sequencing output.<br />

10593 General Poster Session (Board #49B), Mon, 1:15 PM-5:15 PM<br />

Prevalence <strong>of</strong> TP53 p.R337H mutation in children with cancer from a<br />

public hospital in southern Brazil. Presenting Author: Juliana Giacomazzi,<br />

Programa de Pós Graduação em Medicina: Ciências Médicas, Universidade<br />

Federal do Rio Grande do Sul (UFRGS) e Laboratório de Medicina<br />

Genômica, Centro de Pesquisa Experimental, Hospital de Clinicas de Porto<br />

Alegre (HCPA), Porto Alegre, Brazil<br />

Background: Childhood cancers are a common feature in Li-Fraumeni/Li<br />

Fraumeni-Like Syndromes (LFS/LFL), associated with the inheritance <strong>of</strong> a<br />

germline TP53 mutation. Recently, a specific germline mutation in exon 10<br />

<strong>of</strong> the TP53 gene, p.R337H, has been reported at a high prevalence in<br />

Southeastern Brazil. Initial studies on this mutation claimed that the main,<br />

if not exclusive, cancer risk in carriers is childhood adrenocortical carcinoma<br />

(ADR). However, others recent reports identified p.R377H carriers<br />

among LFL families and among a larger spectrum <strong>of</strong> tumors, such as<br />

choroid plexus carcinoma (CPC) and osteosarcoma. The aim <strong>of</strong> this study<br />

was to assess the frequency <strong>of</strong> the p.R337H mutation in children<br />

diagnosed with tumors <strong>of</strong> the LFS/LFL spectrum: ADR, sarcoma, central<br />

nervous system tumors, leukemia, germline cell tumors and Wilm´s tumor)<br />

at the Children´s Cancer Institute <strong>of</strong> Rio Grande do Sul (Brazil) between<br />

1998 and 2011. Methods: Family history (FH) was recorded in pedigrees,<br />

and DNA was extracted from peripheral blood and FFPE samples by<br />

standard methods. Screening for the p.R337H mutation was performed by<br />

allelic discrimination (TaqMan assay) and by TP53 exon 10 sequencing.<br />

Results: Analysis <strong>of</strong> 295 children diagnosed with tumors <strong>of</strong> the LFS/LFL<br />

spectrum identified the p.R337H mutation in 9/11 (81,8%) children with<br />

ADR (including a homozygous p.R337H individual) and in 2/2 (100%)<br />

children with CPC. All individuals had loss <strong>of</strong> heterozygosity in the tumor<br />

and in all cases the mutant allele occurred on the same founder haplotype.<br />

One hundred fourth-six (49,5%) patients had a cancer FH in first or second<br />

degree relatives, 47 (16,0%) had a FH <strong>of</strong> breast cancer and 49 (16,6%)<br />

had FH <strong>of</strong> LFL consistent with Chompret and/or Birch criteria. Conclusions:<br />

These results confirm the strong association <strong>of</strong> p.R337H with ADR and<br />

CPC. A FH consistent with LFL syndrome is present in a high proportion <strong>of</strong><br />

children diagnosed with tumors <strong>of</strong> the LFS/LFL spectrum in Southern<br />

Brazil. Financial support: FIPE-HCPA, CAPES, FAPERGS and Glaxo Smith<br />

Kline.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


10594 General Poster Session (Board #49C), Mon, 1:15 PM-5:15 PM<br />

Prognostic value <strong>of</strong> “angiogenic” risk score in early-stage NSCLC. Presenting<br />

Author: Elena Sanmartin, Fundación para la Investigación del Hospital<br />

General Universitario de Valencia, Valencia, Spain<br />

Background: Angiogenesis is a key mechanism in tumor growth and<br />

dissemination mainly regulated by VEGF family members. We analyze the<br />

expression <strong>of</strong> 11 angiogenic genes in a cohort <strong>of</strong> resectable NSCLC patients<br />

and correlate them with clinico-pathological variables and prognosis.<br />

Methods: RNA was obtained from tumor and normal lung specimens from<br />

175 NSCLC patients. RT-PCR was performed to assess the expression <strong>of</strong><br />

HIF1-A, PIGF, VEGFA, VEGFB, VEGFC, VEGFD, VEGFR1, VEGFR2,<br />

VEGFR3, NRP1 and NRP2. Relative expression was normalized by an<br />

endogenous gene (GUS) using the Pfaffl formulae. Differences were<br />

considered statistically significant at p�0.05. Results: We found that<br />

tumor samples had a significant higher expression <strong>of</strong> PIGF and lower<br />

expression <strong>of</strong> VEGFD, VEGFR2, and VEGFR3 compared with normal tissue<br />

(2.76X, 0.035X, 0.417X and 0.426X, respectively). The group <strong>of</strong> patients<br />

with higher expression levels <strong>of</strong> VEGFA or PlGF had a significantly reduced<br />

TTP (p�0.024 and p�0.027, respectively) and OS (p�0.055 and<br />

p�0.048, respectively) whereas those patients with values <strong>of</strong> VEGFB or<br />

VEGFD below the median had reduced TTP (p�0.020 and p�0.135,<br />

respectively) and OS (p�0.003 and p�0.089, respectively). The multivariate<br />

Cox regression analysis revealed that VEGFA, VEGFB and PlGF were<br />

independent prognostic markers for TTP and OS and based on these results<br />

we generated an “angiogenic” risk score model. TTP and OS were<br />

significantly different among the low, medium and high risk score patients<br />

(Table). Conclusions: VEGF family members are master control genes <strong>of</strong> the<br />

angiogenic process and have a crucial role in the prognostic <strong>of</strong> the disease.<br />

We found differences in the expression <strong>of</strong> four genes between tumor and<br />

normal lung samples. An “angiogenic” risk score (based on the expression<br />

<strong>of</strong> PlGF, VEGF-A and VEGF-B) significantly predicts OS and TTP in our<br />

cohort <strong>of</strong> early-stage NSCLC patients. Validation in an independent cohort<br />

is needed. Supported by grants PS09-01149 and RD06/0020/1024 from<br />

ISCIII.<br />

TTP OS<br />

Risk score N Median 95% CI p N Median 95% CI p<br />

Low 45 NR - 0.001 47 NR - �0.0001<br />

Intermediate 47 NR - 50 NR -<br />

High 41 12.7 5.1-20.3 42 18.8 13.9-23.6<br />

10596 General Poster Session (Board #49E), Mon, 1:15 PM-5:15 PM<br />

Retrospective EGFR mutation testing <strong>of</strong> clinical specimens from the<br />

EURTAC trial <strong>of</strong> erlotinib in non-small cell lung cancer (NSCLC) using a<br />

novel allele-specific PCR (AS-PCR) assay. Presenting Author: Susana<br />

Benlloch, Pangaea Biotech, USP Dexeus University Institute, Barcelona,<br />

Spain<br />

Background: Anti-EGFR inhibitors are superior to chemotherapy in first-line<br />

therapy <strong>of</strong> advanced EGFR-mutant NSCLC. The EURTAC trial was a<br />

randomized Phase III trial <strong>of</strong> erlotinib vs. chemotherapy in patients with<br />

EGFR-mutant NSCLC. Interim results showed significant improvement in<br />

progression-free survival (R. Rosell, ASCO 2011). An accurate rapid in vitro<br />

diagnostic for EGFR mutations is needed to select patients for this therapy.<br />

Methods: Prospective EGFR mutation testing for the trial was performed on<br />

laser-capture microdissected tumor cells using a combination <strong>of</strong> 3 labdeveloped<br />

tests (LDTs), including a Length Analysis <strong>of</strong> Fluorescentlylabeled<br />

PCR (Genescan) method for exon 19 deletions, a Taqman-based<br />

PCR assay for exon 21 mutation with laser-capture macrodissected tumor<br />

cells, and secondary Sanger sequencing. A subset <strong>of</strong> samples from the trial<br />

was retrospectively tested with an AS-PCR assay (cobas EGFR mutation<br />

test) which detects L858R and � 29 exon 19 deletions. The test provides<br />

automated results within 8 h; the DNA required can be isolated from one<br />

5-micron tissue section. Four methods were compared: AS-PCR assay,<br />

LDT, direct Sanger sequencing and massively parallel sequencing (MPS;<br />

454, Branford, CT). Results: LDT results were obtained for 1044 screened<br />

patients. Residual tumor blocks were available for 487 patients (47%),<br />

including 303 wild-type, 172 mutant (135 enrolled on the trial) and 12<br />

inconclusive cases by the LDT. Comparison <strong>of</strong> AS-PCR and LDT results<br />

showed a positive percent agreement (PPA) – 93.7% (CI 88.8%, 96.5%),<br />

and negative percent agreement (NPA) – 97.5% (94.9%, 98.8%). Comparison<br />

<strong>of</strong> AS-PCR and Sanger results showed a PPA <strong>of</strong> 96.6% (91.7%,<br />

98.7%) but an NPA <strong>of</strong> 88.3% (84.1%, 91.5%). Among 34 AS-PCR �<br />

/<br />

Sanger- case, MPS confirmed the presence <strong>of</strong> exon 19 deletions in 25 cases<br />

and L858R mutations in 7. Direct comparison <strong>of</strong> AS-PCR and MPS results<br />

showed a PPA <strong>of</strong> 93.1% (88.1%, 96.1%) and NPA <strong>of</strong> 97.7% (95.0%,<br />

98.9%). <strong>Clinical</strong> outcomes for cases with mutations detected by the<br />

AS-PCR test will be presented. Conclusions: The AS-PCR assay was highly<br />

concordant with the LDT and MPS, and more sensitive than Sanger<br />

sequencing.<br />

Tumor Biology<br />

679s<br />

10595 General Poster Session (Board #49D), Mon, 1:15 PM-5:15 PM<br />

Activation <strong>of</strong> angiogenic pathway in the prediction <strong>of</strong> pathologic response to<br />

bevacizumab-based neoadjuvant therapy in breast cancer. Presenting<br />

Author: Jose Manuel Lopez-Vega, Hospital Universitario Marques de<br />

Valdecilla, Cantabria, Spain<br />

Background: To evaluate potential biomarkers <strong>of</strong> pathological response to<br />

bevacizumab-based neoadjuvant therapy in untreated breast cancers (BC)<br />

patients recruited in a phase II, multicenter clinical trial. Methods: Patients<br />

received a single infusion <strong>of</strong> bevacizumab (15 mg/ kg) (C1) 3 weeks prior to<br />

the beginning <strong>of</strong> neoadjuvant chemotherapy (NAC) consisting in 4 cycles <strong>of</strong><br />

docetaxel (60 mg/mq), doxorubicin (50 mg/mq) and bevacizumab (15 mg/<br />

kg) every 21 days (C2-C5) following by surgery. Biomarker expression was<br />

assessed by immunohistochemistry (Ki67, CD31, CD31/Ki67, VEGFR2,<br />

pVEGFR2 [Y951]) before and after bevacizumab infusion (C1). Gene<br />

expression was analyzed using Affimetrix Human Gene ST 1.0. Results: This<br />

analysis was performed on 73 patients (49 yr, range 29-70). Twenty (27%)<br />

patients obtained best response (G4-G5) whether 50 (68%) were considered<br />

as no responder (G1-G2-G3). Response was associated with negative<br />

estrogen receptors expression (p�0.02) and high Ki67 basal and after C1<br />

expression (p�0.009 and p�0.01). Six (54%) <strong>of</strong> the triple negative<br />

tumors were responders (p�0.05). Interestingly, change in pVEGFR2<br />

[Y951] staining induced by bevacizumab administration was found significantly<br />

associated with response (p�0.0). Decrease in the phosphorilation<br />

status <strong>of</strong> VEGFR2 (Y951) �70% yielded a receiver operating characteristic<br />

(ROC) curve area <strong>of</strong> 0.681 (95% CI: 0.536 - 0.825) with 84% sensitivity<br />

and 95% specificity. The positive and negative predictive values for this<br />

marker were 60% and 64%, respectively. The change in phosphorilation<br />

status <strong>of</strong> VEGFR2p remains a significant predictor biomarker <strong>of</strong> response in<br />

multivariate analysis (OR�0.9, IC%95 0.96-0.99, p�0.04) after adjusting<br />

for clinical-pathological characteristics. Conclusions: These findings<br />

suggest the role <strong>of</strong> the phosphorilation status <strong>of</strong> VEGFR2 as predictive<br />

biomarkers <strong>of</strong> pathological response to bevacizumab in neoadjuvant setting<br />

in breast cancer.<br />

10597 General Poster Session (Board #49F), Mon, 1:15 PM-5:15 PM<br />

Response and long-term outcomes after neoadjuvant chemotherapy: Pooled<br />

dataset <strong>of</strong> patients stratified by molecular subtyping by MammaPrint and<br />

BluePrint. Presenting Author: Stefan Gluck, University <strong>of</strong> Miami/Sylvester<br />

Comprehensive Cancer Center, Miami, FL<br />

Background: Classification <strong>of</strong> breast cancers into molecular subtypes may<br />

be important for the proper selection <strong>of</strong> therapy for patients with early<br />

breast cancer. Previous analyses had shown that breast cancer subtypes<br />

have distinct clinical outcome (Sorlie, PNAS, 2001; Esserman, BCRT,<br />

2011). Herein, we analyze using MammaPrint together with an 80-gene<br />

molecular subtyping pr<strong>of</strong>ile (BluePrint) the response to neo-adjuvant<br />

chemotherapy and long term outcomes. Methods: This study was carried out<br />

on data from 144 patients from the I-SPY I trial; 232 patients from<br />

biomarker discovery program at MD Anderson (133 and 99 respectively;<br />

Hess, 2006, JCO; Iwamoto, 2011, BCRT); and 68 patients from City <strong>of</strong><br />

Hope (Somlo, ASCO, 2010). All patients were treated in the neo-adjuvant<br />

setting with standard chemotherapy. MammaPrint and BluePrint were<br />

determined on 44K Agilent arrays run at Agendia or available through the<br />

I-SPY 1 data portal, or from Affymetrix U133A arrays. MammaPrint and<br />

BluePrint resulted in 4 distinct molecular groups: Luminal A (MammaPrint<br />

Low-risk/Luminal-type), Luminal B (MammaPrint High-risk/Luminal-type),<br />

Basal-type and HER2-type. Results: The overall pCR <strong>of</strong> this patient cohort<br />

was 22% but differed substantially among the subgroups. pCR was<br />

observed in 5% <strong>of</strong> the Luminal-A samples and 10% <strong>of</strong> Luminal-B, in 39%<br />

<strong>of</strong> the HER2-type samples and in 33% <strong>of</strong> the Basal-type samples. Patients<br />

with Basal-type tumors had a 5-year DFS <strong>of</strong> 71%; HER2-type had a 5-year<br />

DFS <strong>of</strong> 67%(n�71); 69% in HER2-type subgroup not treated with<br />

HER2-targeted therapy (n�45); Luminal-B type had a 5-year DFS <strong>of</strong> 77%<br />

and Luminal-A type showed 5-year DFS <strong>of</strong> 95%. Conclusions: We observed<br />

marked differences in response and DFS to neo-adjuvant treatment in<br />

groups stratified by MammaPrint and BluePrint. These findings confirm<br />

differences in chemotherapy response among molecular subgroups and<br />

indicate that the BluePrint and MP pr<strong>of</strong>ile used for this analysis helps to<br />

further establish a clinical correlation between molecular subtyping and<br />

treatment outcomes.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


680s Tumor Biology<br />

10598 General Poster Session (Board #49G), Mon, 1:15 PM-5:15 PM<br />

The efficacy <strong>of</strong> targeted next-generation sequencing for detection <strong>of</strong><br />

clinically actionable mutations in cancer. Presenting Author: Yu-Ye Wen,<br />

Baylor College <strong>of</strong> Medicine, Houston, TX<br />

Background: The emergence <strong>of</strong> next-generation sequencing (NGS) technologies<br />

has significantly accelerated the identification <strong>of</strong> cancer-causing<br />

mutations and the development <strong>of</strong> personalized cancer care. However, the<br />

clinical application <strong>of</strong> these technologies to detect cancer gene mutations<br />

has been extremely limited due to the long turn around time, the high cost,<br />

and large amount <strong>of</strong> input DNA required by existing NGS-based tests.<br />

Methods: We have assessed the performance <strong>of</strong> a novel NGS technology that<br />

merges multiplex PCR with ion semiconductor sequencing (AmpliSeq, Life<br />

Technologies, Inc.) in our clinical diagnostic laboratory. The test interrogates<br />

739 common mutations in 46 cancer genes including many clinically<br />

actionable mutations concurrently. First, we studied 12 tumor samples<br />

including 4 archived FFPE, 4 blood/bone marrow, and 4 cell line samples<br />

with known mutations to evaluate the sensitivity and specificity <strong>of</strong> the test.<br />

We then studied 34 de-identified, archived FFPE tumor samples <strong>of</strong><br />

unknown genotype to further evaluate the efficacy <strong>of</strong> the test. Results: We<br />

successfully identified all known mutations previously detected by Pyposequencing<br />

or Sanger sequencing technologies. Multiple serial dilution<br />

studies showed that the test could detect mutations at frequencies as low<br />

as 5% with 99% confidence. For the samples <strong>of</strong> unknown genotype, we<br />

detected 23 COSMIC mutations in 16 samples including HRAS, BRAF,<br />

MET, TP53 mutations in lung cancer, KRAS, PIK3CA, TP53, APC, BRAF,<br />

ERBB2 mutations in colon cancer, TP53 and KRAS mutations in breast<br />

cancer, and KIT and PDGFRA mutations in GIST. Analysis <strong>of</strong> the variant<br />

call data showed that a minimum <strong>of</strong> 100X coverage is required in order to<br />

detect mutations at 10% frequency or above; a minimum 300K final library<br />

reads are necessary in order to minimize/eliminate amplicon dropout.<br />

Conclusions: The targeted NGS test can effectively detect cancer gene<br />

mutation with input DNA as low as a few nanograms, turn around time can<br />

be as short as two days, and can significantly lower cost compared to<br />

traditional Sanger sequencing. Our experience demonstrates that this<br />

technology holds great potential for clinical use, including diagnostic and<br />

therapeutic applications.<br />

10600 General Poster Session (Board #50A), Mon, 1:15 PM-5:15 PM<br />

A circulating epithelial cell type merging stem cell, EMT, and hypoxic<br />

stress markers in early and advanced breast cancer. Presenting Author: Leo<br />

Habets, Metares e.V, Aachen, Germany<br />

Background: MAINTRAC as developed by our coworkers from Jena finds<br />

more CETC. They found CETC in 90% <strong>of</strong> high risk patients. Circulating cells<br />

in epithelial mesenchymal transition (EMT�) should be detectable in early<br />

disease. Methods: 2 colours (7AAD, EP-FITC) detected living and dead<br />

cells. Cells in EMT (EP-FITC, Vim-PE) were analysed in paralell. Complex<br />

marker expression urged us to introduce a three colour technique. 1.<br />

EPCAM-FITC, Vimentin-PE and DAPI , marking living or dead cells in EMT.<br />

2. EP-FITC, Vim-PE and CD44 PB for stemness. ACA9 (hypoxia) and<br />

PARP-1 expression (genotox) were used too. Results: In the 2 colour phase<br />

we examined CEC in 110 patients with early disease and 44 patients with<br />

metastasis. Cells were detectable in both disease situations in 50 and 60%<br />

<strong>of</strong> patients, in a control population only in few patients low numbers were<br />

found. In non cancer situations e.g liver disease high numbers <strong>of</strong> EMT�cells<br />

were found.The results <strong>of</strong> 3 colour analysis are shown in the table. No clear<br />

differences between the classical risk groups are found, other subgroups<br />

are too small yet to be considered. Conclusions: Our findings show that in<br />

both disease situations cells merging EMT, stemcells ( EP�,Vim�.CD44�)<br />

and other markers are detectable. This cell type however is found also in<br />

non cancer conditions.We hypothesize that hypoxia is the common driver.<br />

Metastasis gives different patterns. Highly aggressive cancers show <strong>of</strong>ten<br />

none or few cells. In less agressive disease high cell numbers are found. We<br />

need to characterize the �benign� EMT phenomenon, as it could mingle<br />

with �real� CTC. Definitely liver is the source <strong>of</strong> these benign cells. Multi<br />

colour analysis will reveal the differences. Nevertheless monitoring <strong>of</strong> this<br />

special cell type could give new insights in the metastatic process in early<br />

and late disease.<br />

Cell type EMT living EMT total EMT�CD44<br />

CEC per ml �250 �250 �250 �250 �250 �250<br />

ALL 56 44 21 79 25 75 n�60<br />

N0 80 20 41 59 57 43 n�36<br />

N� 64 26 51 49 34 66 n�24<br />

Lum A 48 52 20 80 34 66 n�30<br />

Lum B 48 52 31 69 23 77 n�29<br />

Advanced 39 61 27 73 37 63 n�40<br />

Controls 98 2 91 9 81 19 n�112<br />

�Liver � 10 90 22 78 n�84<br />

10599 General Poster Session (Board #49H), Mon, 1:15 PM-5:15 PM<br />

Serum HER2 in the context <strong>of</strong> circulating tumor cells in patients with<br />

metastatic breast cancer. Presenting Author: Volkmar Mueller, University<br />

Medical Center Hamburg, Gynecology, Hamburg, Germany<br />

Background: Circulating tumor cells (CTC) reflect an aggressive tumor<br />

behavior by hematogenous tumor cell dissemination. Overexpression <strong>of</strong><br />

HER2 in breast cancer (BC) is associated with increased angiogenesis and<br />

therefore potentially linked to increased hematogenous tumor cell spread.<br />

The aim <strong>of</strong> the analysis was to investigate whether concentrations <strong>of</strong> serum<br />

HER2 (sHER2) deliver prognostic information in the context <strong>of</strong> CTC<br />

detection in metastatic BC patients. Methods: Blood was obtained in a<br />

prospective multicenter setting from 254 patients with metastatic BC at<br />

the time <strong>of</strong> disease progression. sHER2 was determined using a commercial<br />

ELISA-kit (Wilex). CTC were detected with the CellSearch system<br />

(Veridex). Patients received systemic treatment according to national and<br />

international guidelines including HER2-targeted treatment. Results: Five<br />

or more CTC were detected in 122 <strong>of</strong> 245 evaluable patients (49.8%).119<br />

<strong>of</strong> 251 (47%) metastatic patients had serum sHER2 levels above 15ng/<br />

mL. Median PFS was 9.2 months (95%-CI: 9.9 – 13.0 mths) with elevated<br />

sHER2 versus 11.4 mths (9.9 – 13.0 mths) with non-elevated levels<br />

(p�0.07). OS was 17.4 mths (14.6 – 20.3 mths) vs. 26.5 mths<br />

(23.1-29.8 mths; p�0.01). In patients with 5 or more CTC, serum levels<br />

were above the cut-<strong>of</strong>f for sHER2 in 61% vs. 33% in those with less than 5<br />

CTC (p� 0.01). Patients with elevated sHER and 5 or more CTC hat a PFS<br />

<strong>of</strong> 9.1 mths (7.2 – 11.1 mths) and a OS <strong>of</strong> 14.5 mths (11.8 – 17.2 mths),<br />

those with non-elevated sHER2 and less than 5 CTC a PFS <strong>of</strong> 12.1 (10.1 –<br />

14.1 mths) and a OS <strong>of</strong> 29.5 month (25.4 – 33.6 mths) (p�0.15 for PFS<br />

and p� 0.01 for OS). Including sHER2, CTC and established prognostic<br />

factors in the multivariate analysis, the presence <strong>of</strong> CTC, line <strong>of</strong> therapy, ER<br />

and HER2 status <strong>of</strong> the primary tumor remained independent predictors <strong>of</strong><br />

OS. Conclusions: Elevated serum levels <strong>of</strong> sHER2 are associated with the<br />

presence <strong>of</strong> CTC and indicate poor clinical outcome. However, sHER2 has<br />

no independent prognostic value when presence <strong>of</strong> CTC were taken into<br />

account.<br />

10601 General Poster Session (Board #50B), Mon, 1:15 PM-5:15 PM<br />

Exploratory study <strong>of</strong> histopathological and biomolecular characteristics <strong>of</strong><br />

breast cancer in two different populations: African (Tanzania) and Caucasian<br />

(Italy). Presenting Author: Patrizia Serra, Unit <strong>of</strong> Biostatistics and<br />

<strong>Clinical</strong> Trials, IRST, Meldola, Italy<br />

Background: There is little information available on the clinical and<br />

molecular epidemiology <strong>of</strong> breast cancer in Sub-Saharan African women. In<br />

Tanzania, breast cancer mortality is the second cause <strong>of</strong> death after cancer<br />

<strong>of</strong> the uterine cervix, with the majority <strong>of</strong> tumors diagnosed at a very<br />

advanced stage. We aimed to compare the distribution <strong>of</strong> histopathological<br />

and biomolecular characteristics (ER, PgR, Ki67, HER2, grading) <strong>of</strong> breast<br />

cancer in African (Tanzania) and Caucasian (Italy) patients. Methods: 142<br />

women with invasive breast cancer, 71 from Tanzania and 71 from Italy,<br />

were considered. Histopathological classification and biomolecular determinations<br />

were performed at the Biosciences Laboratory <strong>of</strong> the Cancer<br />

Institute <strong>of</strong> Romagna (I.R.S.T., Meldola, Italy) as part <strong>of</strong> a joint Cancer<br />

Control Project between I.R.S.T and the Bugando Medical Center in<br />

Mwanza, Tanzania. All cases were evaluable for histopatological characterization,<br />

while biomolecular determination was possible in only 61 <strong>of</strong> the<br />

African patients. Hormone receptor status and Ki67 were determined by<br />

IHC; HER2 was evaluated by IHC for the African population and by FISH for<br />

the Italian one. Results: Whilst histopathological patterns were similar in<br />

the two populations, some biomolecular characteristics differed substantially.<br />

Of note, 64% <strong>of</strong> African breast cancer patients had ER negative<br />

tumors compared to only 17% <strong>of</strong> the Caucasian population. Important<br />

differences were observed in high Ki67 (�15%) values: 76% in Tanzanian<br />

women vs 58% in Italian breast cancer cases. HER2 positivity rates also<br />

differed in the two populations (25% in Tanzanian and 17% in Italian<br />

patients), as did the incidence <strong>of</strong> triple negative disease (13% and 7% in<br />

African and Caucasian populations, respectively). Conclusions: This preliminary<br />

study shows that breast cancer in African (Tanzanian) women is<br />

characterized by a high frequency <strong>of</strong> HER2 positive, ER negative, highly<br />

proliferating tumors. These biological patterns, together with very advanced<br />

stage at diagnosis, could be considered the main prognostic factors related<br />

to the high mortality rates for breast cancer in this African population.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


10602 General Poster Session (Board #50C), Mon, 1:15 PM-5:15 PM<br />

The components <strong>of</strong> progression as explanatory variables for overall survival<br />

in the RECIST database. Presenting Author: Saskia Litière, European<br />

Organisation for Research and Treatment <strong>of</strong> Cancer, Brussels, Belgium<br />

Background: Progressive disease (PD) according to RECIST 1.1 (Eisenhauer<br />

et al, 2009) is defined as one or more <strong>of</strong> (1) PD <strong>of</strong> measurable target<br />

lesions, (2) the presence <strong>of</strong> a new lesion (NL) or (3) the unequivocal PD <strong>of</strong><br />

non-target disease (NT-PD). We explored the impact <strong>of</strong> these components,<br />

varying over time, on predicting overall survival (OS) in the RECIST<br />

database residing at EORTC (Bogaerts et al, 2009). Methods: Data was<br />

selected from 12 randomized clinical trials (3530 patients with breast,<br />

lung or colorectal cancer). A maximum <strong>of</strong> 5 target lesions was used to<br />

determine the sum <strong>of</strong> diameters. The following were calculated or assigned<br />

at each measurement time t: best response (BR) was best % improvement<br />

from baseline up to t (0% if no improvement - 100% if complete response<br />

(CR)); tumor growth (TG) was the weekly rate <strong>of</strong> increase from nadir to t (0 if<br />

no increase; irrespective <strong>of</strong> prior shrinkage), presence <strong>of</strong> NL (yes/no), and<br />

occurrence <strong>of</strong> NT-PD (yes/no); categories were not mutually exclusive. OS<br />

was analyzed by tumor type using a Cox regression model, adjusting for<br />

baseline sum, and including BR, TG, presence <strong>of</strong> NL and NT-PD as time<br />

dependent covariates. Results: Thirty-six percent <strong>of</strong> patients had NL, 26%<br />

had NT-PD, 11% achieved CR and 14% did not have shrinkage <strong>of</strong> target<br />

lesions, while 46% experienced TG (median strongest growth per patient <strong>of</strong><br />

0.5 mm/week). Regardless <strong>of</strong> tumor type, the presence <strong>of</strong> NL (Hazard Ratio<br />

(HR) ranging 1.4-2.5), NT-PD (HR 1.2-2.5) and TG (per 1mm/week<br />

increase; HR 1.1-1.4) were associated with worse OS, while achieving CR<br />

was associated with a longer OS (HR 0.2-0.8). Further analyses exploring<br />

the functional shape <strong>of</strong> the association between BR and TG, and OS are<br />

ongoing. This includes putting TG in contrast with the more usual % cut<strong>of</strong>f<br />

defining target PD. Conclusions: All three components <strong>of</strong> PD according to<br />

RECIST are independently strongly associated with OS. Quantification<br />

such as this will enable a better understanding <strong>of</strong> the role <strong>of</strong> each<br />

component (e.g. clinical aspect <strong>of</strong> NT-PD assessment) in PD evaluation.<br />

Work is ongoing to incorporate this information into an updated RECIST<br />

with enhanced prediction <strong>of</strong> subsequent survival.<br />

10604 General Poster Session (Board #50E), Mon, 1:15 PM-5:15 PM<br />

Monitoring <strong>of</strong> plasma pro-GRP level during EGFR-TKI treatment. Presenting<br />

Author: Yuko Kawano, Cancer Institute Hospital <strong>of</strong> the Japanese<br />

Foundation for Cancer Research, Tokyo, Japan<br />

Background: Lung cancers harboring mutations in the epidermal growth<br />

factor receptor gene (EGFR) respond to EGFR tyrosine kinase inhibitors<br />

(EGFR-TKI), but drug resistance invariably emerges. The major acquired<br />

mechanisms <strong>of</strong> resistance are the EGFR T790M mutation or MET gene<br />

amplification. Transformation from NSCLC into small-cell lung cancer<br />

(SCLC) has been recently identified in acquired resistance to EGFR-TKI.<br />

However, it is difficult to predict the transformation during EGFR-TKI<br />

treatment because obtaining serial and sufficient specimens for biopsy is<br />

difficult. Pro-gastrin-releasing peptide (Pro-GRP) is a specific and sensitive<br />

tumor marker for SCLC. We evaluated the plasma Pro-GRP levels in<br />

EGFR-mutant NSCLCs and determined whether plasma Pro-GRP levels<br />

could predict SCLC transformation in resistance to EGFR-TKI. Methods:<br />

From July 2008 to December 2011, 49 patients with EGFR-mutant NSCLC<br />

who received EGFR-TKI treatment were enrolled. Plasma was obtained<br />

from these patients before EGFR-TKI treatment and when EGFR-TKI<br />

treatment failed. Pro-GRP and CEA levels were measured and compared<br />

before and after treatment. Results: Patient characteristics for 49 patients<br />

(15 men, 34 women) were as follows: median age, 62 years (41–81 years);<br />

histology, 46 adenocarcinomas (AD) and 3 non-AD tumors; and EGFR<br />

mutation type, 25 exon 19 deletions and 24 exon 21 L858R. All 49<br />

patients had received EGFR-TKI treatment (45 with gefitinib and 4 with<br />

erlotinib); the response to EGFR-TKI treatment was PR in 39 patients, SD<br />

in 7, PD in 2, and NE in 1. Positive rate <strong>of</strong> ProGRP and CEA at<br />

pre-EGFR-TKI treatment was 2.0% and 57.2% and that at post-EGFR-TKI<br />

treatment was 6.1% and 69.4%, respectively. In 3 <strong>of</strong> 49 patients, the<br />

Pro-GRP levels had increased after treatment, but the CEA level did not<br />

increase. Objective responses to cytotoxic chemotherapy were noted in all 3<br />

patients after EGFR-TKI treatment. Conclusions: Monitoring <strong>of</strong> plasma<br />

Pro-GRP during EGFR-TKI treatment may be useful for early detection <strong>of</strong><br />

SCLC transformation in resistance to EGFR-TKI.<br />

Tumor Biology<br />

681s<br />

10603 General Poster Session (Board #50D), Mon, 1:15 PM-5:15 PM<br />

pAKT expression in paraffin-embedded xenograft tumors after fixation<br />

delays and human breast cancer by optimized immunohistochemistry.<br />

Presenting Author: Sherry X. Yang, Division <strong>of</strong> Cancer Treatment and<br />

Diagnosis, National Cancer Institute, Bethesda, MD<br />

Background: With progress in targeting PI3K/AKT pathway in the treatment<br />

<strong>of</strong> a wide range <strong>of</strong> cancers, it is getting more important than ever for<br />

optimization <strong>of</strong> detection method <strong>of</strong> AKT kinase, a central effector <strong>of</strong> the<br />

pathway, in archived tissues. Recently, we found that pAKT-S473 (pAKT)<br />

significantly predicts paclitaxel benefit in breast cancer (JCO 28: 2974,<br />

2010), in which a good analytical and clinical performance <strong>of</strong> quantitative<br />

pAKT immunohistochemistry (IHC) was demonstrated. The current study<br />

evaluates pAKT stability in human breast cancer xenograft tumors after<br />

delays in fixation, and assesses a detection window following fixation delays<br />

with an established cut<strong>of</strong>f [staining index (SI) � 2]. Methods: Xenograft<br />

tumors with high (MDA-MB-468) and intermediate levels (MDA-MB-231)<br />

<strong>of</strong> pAKT were fixed or snapped frozen in 10% neutral-buffered formalin or<br />

liquid nitrogen after delays post-excision. pAKT staining was performed in<br />

xenograft tumors and 96 cases <strong>of</strong> human surgical breast tumors by our<br />

optimized and traditional IHCs, and Western blot. Results: The mean SIs<br />

were 133, 121, 112, 94, 84, and 66 using optimized, and in contrast were<br />

51, 44, 29, 14, 12, and 6.4 on traditional method at 0, 15, 30, 60, 120<br />

and 180 min (2.6-fold by comparing the optimized with the traditional at<br />

baseline) in MDA-MB-468 xenograft tumors. pAKT level was ½ by the<br />

optimized, similar to the level detected by Western blot, relative to 1/8 <strong>of</strong><br />

the baseline by the traditional (10.6-fold) at 180 min. The logarithmic<br />

decline rate by the optimal was 3.1 times (95% CI, 2.4 - 3.7) less than that<br />

<strong>of</strong> the traditional (normal approximation; 2-sided P � 0.0001). pAKT<br />

expression was observed in 38.5% (37/96 ) <strong>of</strong> surgical breast tumors,<br />

comparable to 38% (606/1581) in a large cohort derived from the NSABP<br />

B-28 trial. There was little loss <strong>of</strong> pAKT in MDA-MB-231 xenograft tumors<br />

up to 180 min by IHC and Western blot. Conclusions: The optimized pAKT<br />

IHC significantly increases the sensitivity <strong>of</strong> detection with a large window<br />

for positivity. It is suitable for use in archived human specimens although it<br />

warrants further standardization and validation among research laboratories<br />

and perhaps diagnostic laboratories in the future.<br />

10605 General Poster Session (Board #50F), Mon, 1:15 PM-5:15 PM<br />

Anti-AML activity <strong>of</strong> a novel beta-catenin antagonist BC2059. Presenting<br />

Author: Kapil N. Bhalla, University <strong>of</strong> Kansas Medical Center, Kansas City,<br />

KS<br />

Background: The canonical WNT-�-catenin pathway is essential for selfrenewal,<br />

growth and survival<strong>of</strong> AML stem and progenitor cells. Deregulated<br />

WNT signaling inhibits degradation <strong>of</strong> �-catenin, causing increased nuclear<br />

translocation and interaction <strong>of</strong> �-catenin with the TCF/LEF transcription<br />

factor, which up regulates cyclin D1, Myc and survivin expression in AML<br />

progenitor cells. BC2059 (�-Cat Pharmaceuticals) is a potent, small<br />

molecule, anthraquinone oxime-analog, which inhibits WNT-� catenin<br />

pathway by promoting the degradation and attenuation <strong>of</strong> �-catenin levels.<br />

Methods: We determined the in vitro anti-AML activity <strong>of</strong> BC2059 (BC)<br />

(250 to 1000 nM) against cultured and primary human AML blast<br />

progenitors, as well as evaluated the in vivo anti-AML efficacy <strong>of</strong> BC in<br />

NOD-SCID and NOD-SCID-IL2� receptor deficient (NSG) mice. Results: BC<br />

induced cell cycle G1 phase accumulation and apoptosis (40%) <strong>of</strong> the<br />

cultured OCI-AML3, HL-60 and HEL92.1.7 (HEL) AML cells. BC dosedependently<br />

also induced apoptosis <strong>of</strong> primary AML versus normal progenitors.<br />

Treatment with BC resulted in proteasomal degradation and decline in<br />

the nuclear levels <strong>of</strong> �-catenin, which led to decreased activity <strong>of</strong> the<br />

LEF1/TCF4 transcription factor highlighted by reduced TOP-FLASH luciferase<br />

activity in the AML cells. This was associated with reduced protein<br />

levels <strong>of</strong> cyclin D1, MYC and survivin. Co-treatment with BC and the<br />

histone deacetylase inhibitor panobinostat (PS) (10 to 20 nM) synergistically<br />

induced apoptosis <strong>of</strong> cultured and primary AML blasts. Following tail<br />

vein infusion and establishment <strong>of</strong> AML by OCI-AML3 or HEL cells in<br />

NOD-SCID mice, treatment with BC (5, 10 or 15 mg/kg b.i.w, IV) for three<br />

weeks demonstrated improved survival, as compared to the control mice (p<br />

�0. 001). Survival was further improved upon co-treatment with BC and<br />

PS (5 mg/kg IP, MWF). BC treatment (5 or 10 mg/kg IV) also dramatically<br />

improved survival <strong>of</strong> NSG mice with established human AML following<br />

tail-vein injection <strong>of</strong> primary AML blasts expressing FLT3 ITD. Mice did not<br />

experience any toxicity or weight loss. Conclusions: These findings highlight<br />

the notable pre-clinical in vitro and in vivo activity and warrant further<br />

development and in vivo testing <strong>of</strong> BC against human AML.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


682s Tumor Biology<br />

10606 General Poster Session (Board #50G), Mon, 1:15 PM-5:15 PM<br />

Evaluation <strong>of</strong> the clinical utility <strong>of</strong> kallikrein-related peptidase 6 gene<br />

(KLK6) downregulation in breast cancer. Presenting Author: Kleita Michaelidou,<br />

Department <strong>of</strong> Biochemistry and Molecular Biology, University <strong>of</strong><br />

Athens, Panepistimiopolis, Athens, Greece<br />

Background: Breast cancer (BC), the most common malignancy among the<br />

female population, remains a crucial public health problem. The identification<br />

and exploitation <strong>of</strong> novel molecular biomarkers can contribute in the<br />

multidimensional approach which is required for optimal management <strong>of</strong><br />

BC patients. The abnormal expression <strong>of</strong> several KLK members has been<br />

documented for breast cancer. Interestingly, many <strong>of</strong> these genes are found<br />

to be potential prognostic markers for this malignancy. KLK6 expression is<br />

positively associated with tumor progression in various human malignancies;<br />

however in breast cancer, it can restrain tumor progression. We<br />

therefore sought to investigate the possible clinical value <strong>of</strong> KLK6 as a<br />

breast cancer biomarker. Methods: Total RNA was isolated from 107 breast<br />

tumors and their matched normal compartments. After testing the quality<br />

<strong>of</strong> the extracted RNA, cDNA was prepared by reverse transcription.<br />

Quantitative Real-time PCR was performed, for KLK6 mRNA quantification,<br />

using the SYBR Green chemistry. Relative quantification analysis was<br />

made using the comparative Ct (2-��C T) method. HPRT1 was used as an<br />

endogenous control gene and BT-474 breast cancer cell line served as a<br />

calibrator. Results: KLK6 mRNA levels were significantly downregulated in<br />

the cancerous breast tissue specimens compared to their normal counterparts<br />

(p�0.001). Additionally, a negative association was observed between<br />

KLK6 expression status and tumor stage, given the fact that 54.2%<br />

<strong>of</strong> less advanced breast tumors (TNM stage� T2a) were KLK6-positive<br />

compared to the 34.6% <strong>of</strong> more advanced-stage tumors (TNM �T2a)<br />

(p�0.034). Moreover, a statistically significant negative correlation was<br />

documented between KLK6 mRNA levels and estrogen (p�0.001) and<br />

progesterone receptor (p�0.003) statuses. Conclusions: The downregulation<br />

<strong>of</strong> KLK6 in cancerous compared to normal sections <strong>of</strong> breast tissue<br />

samples reflects the reported tumor suppressor properties <strong>of</strong> KLK6 gene in<br />

breast malignancies. Furthermore, the negative association <strong>of</strong> KLK6 mRNA<br />

expression with tumor stage, ER and PR statuses reveals the putative role <strong>of</strong><br />

KLK6 as a promising prognostic breast cancer biomarker.<br />

10608 General Poster Session (Board #51A), Mon, 1:15 PM-5:15 PM<br />

Study <strong>of</strong> the usefulness <strong>of</strong> angiotensin type 1 receptor (AGTR1) as a<br />

possible response predictor in patients with metastatic breast cancer<br />

(mBC) subjected to chemotherapy and treatment with bevacizumab<br />

(AVALUZ Study). Presenting Author: Juan De la Haba- Rodriguez, Hospital<br />

Universitario Reina S<strong>of</strong>ía, Cordoba, Spain<br />

Background: Angiotensin type 1 receptor (AGTR1) is a membrane receptor<br />

implicated in the regulation <strong>of</strong> arterial pressure. It has also been related to<br />

carcinogenesis and tumor spread, and participates in neoangiogenesis.<br />

AGTR1 is expressed in a number <strong>of</strong> neoplasms as BC. In a previous<br />

neoadjuvant study, a significant relationship was observed between AGTR1<br />

expression and CR to a regimen <strong>of</strong> chemo and bevacizumab (B). To study<br />

and confirm the relationship between tumor expression <strong>of</strong> AGTR1 and<br />

response to a regimen <strong>of</strong> chemo and B in pts with mBC. Methods: AGTR1<br />

expression was subjected to immun<strong>of</strong>luorescence (monoclonal AGTR1<br />

sc1173 antibody, Santa Cruz Biotechnology, CA, USA Dilution 1:50) in 50<br />

samples <strong>of</strong> mBC pts from the AVALUZ study treated with first line<br />

bevacizumab 10 mg/kg, paclitaxel 150 mg/m2 and gemcitabine 2000<br />

mg/m2 d 1 and 15 c/28 d until PD, unacceptable toxicity or medical<br />

decision. Immunoreactivity was reported as negative (0), low positive (1�)<br />

and high positive (2�). RNA was extracted from paraffin blocks, using the<br />

QIAamp DNA FFPE Tissue Kit and Deparaffinization Solution (QIAGEN)<br />

and expression <strong>of</strong> AGTR1 candidate gene was quantified using reverse<br />

transcription-PCR (RT-PCR). Results: 60% <strong>of</strong> the samples expressed<br />

AGTR1 (26% weak and 34% intense) – expression being more frequent in<br />

HR � tumors (65% vs 45%). Among the 50 samples analyzed, measurable<br />

disease was present in 39 pts. Treatment activity was significantly greater<br />

in the tumors AGTR1 expression 2� (p�0.009) (Table). With a median<br />

follow-up <strong>of</strong> 19.30 months, the progression-free interval was longer in the<br />

tumors with intense AGTR1 expression (17.7 vs 10.8 months, p�0.132).<br />

More consolidated PFS and OS will be presented. Conclusions: AGTR1<br />

expression may be useful as a predictor <strong>of</strong> response to chemo and<br />

bevacizumab. Prospective studies are needed to confirm these findings.<br />

AGTR1 expression/objective response CR PR SD PD<br />

Intense AGTR1 expression (N:14 ) 35.7% (5) 64.3% (9) 0% 0%<br />

Negative or weak AGTR1 expression (N:25) 4% (1) 56.0% (14) 28% (7) 12 (3%)<br />

CR: complete response, PR: partial response, SD: stable disease, PD: progression disease<br />

10607 General Poster Session (Board #50H), Mon, 1:15 PM-5:15 PM<br />

p53 mutations in advanced cancers: <strong>Clinical</strong> characteristics and outcomes<br />

in a phase I setting. Presenting Author: Rabih Said, Department <strong>of</strong><br />

Investigational Cancer Therapeutics (Phase I Program), University <strong>of</strong> Texas<br />

M. D. Anderson Cancer Center, Houston, TX<br />

Background: p53 mutation is one <strong>of</strong> the most common mutations in human<br />

tumors and plays an important role in apoptosis, genomic stability and<br />

inhibition <strong>of</strong> angiogenesis. Methods: We retrospectively reviewed 121<br />

consecutive patients (pts) with solid tumors referred to the <strong>Clinical</strong> Center<br />

for Targeted Therapy (Phase I program) who were tested for p53 mutation,<br />

and compared clinical characteristics and outcomes in p53 positive vs.<br />

wild-type (wt) pts. Results: Of the 121 tested pts, 65 (53.7%) were p53<br />

mutation-positive. The presence <strong>of</strong> p53 mutation was not associated with<br />

sex and race. Tumors with p53 mutation metastasized more <strong>of</strong>ten to the<br />

liver (44 pts (67.7%) with mutated p53 vs. 26 pts (46.4%) with wt p53,<br />

p�0.0264); there was no difference in metastasis to the bones, lungs,<br />

brain, s<strong>of</strong>t tissues and adrenals. p53 mutation also showed a trend toward<br />

an association with PTEN-loss (13 out <strong>of</strong> 43 (30%) with p53 mutated vs. 4<br />

out <strong>of</strong> 37 (11%) with wt p53, p�0.053). Among pts with the p53<br />

mutation, the best median progression-free survival (PFS) prior to phase I<br />

trial entry was 10.97 months (range 0.85–29.04 months) when the<br />

treatment regimen included bevacizumab and 4.37 months (range 0.92–<br />

14.98 months) when the treatment did not include bevacizumab<br />

(P�0.0046). Among patients with wt p53, there was no significant<br />

difference in PFS between bavacizumab-containing regimens and regimens<br />

without bevacizumab. The median OS from diagnosis was 7.7 years<br />

(95% CI 6.32–9.84 years) vs. 11.8 years (95% CI 10.37 years, not<br />

attained) for p53 mutant vs. wild-type disease (p � 0.03). Univariate<br />

analysis for OS from diagnosis showed that mutated p53, Hispanic race (vs.<br />

Caucasians) and the shorter time from diagnosis to metastasis were<br />

associated with a worse prognosis. Conclusions: Tumors with p53 mutation<br />

have a more aggressive clinical behavior, metastasize more to the liver and<br />

may have a higher rate <strong>of</strong> PTEN loss compared to tumors with wt p53; In<br />

addition, anti-angiogenic agents may be <strong>of</strong> therapeutic value in p53mutated<br />

pts.<br />

10609 General Poster Session (Board #51B), Mon, 1:15 PM-5:15 PM<br />

The lipid metabolome <strong>of</strong> clear cell renal cell carcinoma (CCRCC). Presenting<br />

Author: Saby George, Roswell Park Cancer Institute, Buffalo, NY<br />

Background: The commonest type <strong>of</strong> kidney cancer is CCRCC. Treatment<br />

approaches mostly target aberrant vasculature. However, kidney cancer is<br />

also known to accumulate lipids and a detailed knowledge <strong>of</strong> the lipid<br />

species present in these tumors could lead to a better understanding <strong>of</strong> the<br />

underlying aberrant metabolic pathways and suggest possible treatment<br />

strategies. Lipidomics is an emerging field driven by rapid advances in<br />

mass spectrometry (MS), and is widely used to discover biomarkers. We<br />

attempt to identify the lipidomic pr<strong>of</strong>ile <strong>of</strong> CCRCC using a liquid chromatography<br />

MS-based approach (LC-MS). Methods: We utilized 6 fresh frozen<br />

representative samples <strong>of</strong> CCRCC and matching non-tumor areas <strong>of</strong> kidney<br />

from nephrectomy samples. Lipids and other non-polar cellular constituents<br />

were extracted from both CCRCC and control tissues by methyl-t-butyl<br />

ether /methanol. LC-MS based lipid pr<strong>of</strong>iling was performed on a Waters<br />

Q-ToF Premier MS coupled with Ultra Performance LC. The peak detection<br />

and alignment across all chromatograms were performed using the XCMS<br />

s<strong>of</strong>tware (v 1.14.1, Scripps Center for Metabolomics). Statistical comparisons<br />

<strong>of</strong> the intensities <strong>of</strong> aligned peaks were performed using the XCMSbuilt-in<br />

Welch’s t-test. Results: The outcome <strong>of</strong> XCMS was converted to a<br />

table that contains fold change, p-value and mass to charge ratio (m/z) for<br />

each peak, its corresponding retention time, and the integrated peak<br />

intensities from all samples. 224 peaks out <strong>of</strong> 1419 differed between<br />

CCRCC and the control group, with p �0.05, calculated by XCMS. About an<br />

equal number <strong>of</strong> analytes increased or decreased in CCRCC compared with<br />

control samples. Preliminary attempts to identify the analytes included use<br />

<strong>of</strong> METLIN (Scripps Center for Metabolomics) and HMDB (Human metabolome<br />

database, Genome Alberta & Genome Canada) databases. Many <strong>of</strong> the<br />

hits identified phosphatidylcholines, phosphatidylethanolamines, triacylglycerols<br />

and diacylglycerols, as well as other lipid species. Conclusions:<br />

The lipid metabolomic pr<strong>of</strong>ile varied significantly between CCRCC and<br />

control. Further studies are required to confirm the identities <strong>of</strong> the lipid<br />

species contributing to this variation by obtaining structural information<br />

using tandem MS (LC-MS/MS).<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


10610 General Poster Session (Board #51C), Mon, 1:15 PM-5:15 PM<br />

Cytokeratin 19 fragment (CYFRA21-1) to predict the efficacy <strong>of</strong> epidermal<br />

growth factor receptor tyrosine kinase inhibitor (EGFR-TKI) in non-small<br />

cell lung cancer (NSCLC) harboring EGFR mutation. Presenting Author:<br />

Kosuke Tanaka, Division <strong>of</strong> Integrated Oncology, Institute <strong>of</strong> Biomedical<br />

Research and Innovation, Kobe, Japan<br />

Background: EGFR mutation is independently associated with a favorable<br />

response in NSCLC patients receiving EGFR-TKIs, regardless <strong>of</strong> gender or<br />

smoking history. However, recent reports have indicated that squamous<br />

cell carcinoma patients harboring EGFR mutations show a worse response<br />

to EGFR-TKIs than adenocarcinoma patients. We hypothesized that serum<br />

CYFRA21-1 is a predictive marker in EGFR mutated patients treated with<br />

EGFR-TKIs. Methods: We retrospectively screened 160 NSCLC patients<br />

harboring EGFR mutations (exon 19 deletions, L858R in exon 21, or other<br />

minor mutations) who received either gefitinib or erlotinib between 1992<br />

and 2011. Patients were screened for histology, sex, age, smoking status,<br />

efficacy <strong>of</strong> EGFR-TKI and tumor markers (CEA/CYFRA21-1) at initial<br />

diagnosis. Results: Out <strong>of</strong> 160 eligible patients treated with EGFR-TKIs, 77<br />

patients with high CYFRA21-1 level (�2 ng/ml) showed statistically shorter<br />

progression-free survival (PFS) than 83 patients with normal CYFRA21-1<br />

level (median PFS 7.5 vs 14.0 months, p�0.006). No significant difference<br />

in PFS was observed between high CEA group (�5 ng/ml) and normal<br />

CEA group (median PFS 8.6 vs 11.2 months, p�0.2423). Multivariate<br />

analysis revealed that high CYFRA21-1 level is independently associated<br />

with PFS (HR 1.35; p�0.002) as well as squamous cell carcinoma (HR<br />

1.40; p�0.020) and performance status 2-4 (HR 2.63; p�0.003). No<br />

statistically significant difference in overall survival (OS) was observed<br />

between high CYFRA21-1 group and normal group (median OS 24.8 vs<br />

39.1 months, p�0.104). Conclusions: High CYFRA level patients have<br />

significantly shorter PFS, which may indicate that this subgroup has a<br />

larger squamous component and thus less response to EGFR-TKIs. Serum<br />

CYFRA21-1 level is a predictive marker <strong>of</strong> EGFR-TKIs efficacy and EGFR<br />

mutated patients can be divided into two subgroups according to CY-<br />

FRA21-1 level at initial diagnosis.<br />

10612 General Poster Session (Board #51E), Mon, 1:15 PM-5:15 PM<br />

Use <strong>of</strong> proteomic analysis <strong>of</strong> LKB1/AMPK/mTOR pathways to identify<br />

IGF-1R pathway upregulation with LKB1 loss or mTOR inhibition in<br />

NSCLC: Implications for targeted combinations. Presenting Author: Emrullah<br />

Yilmaz, Baylor College <strong>of</strong> Medicine, Houston, TX<br />

Background: LKB1 is a serine/threonine kinase which is mutated in<br />

20-30% <strong>of</strong> non-small cell lung cancers (NSCLC) and functions as a tumor<br />

suppressor by activating AMPK. Loss <strong>of</strong> LKB1 by point mutation or deletion<br />

suppresses AMPK, leading to increased mTOR signaling. We investigated<br />

the signaling pathways modulated by LKB1 mutations and by mTOR<br />

inhibition in NSCLC. Methods: Protein expression in cell lines was measured<br />

by reverse phase protein array. Differences in protein expression at<br />

baseline in LKB1 wild-type versus mutant cell lines and the effects <strong>of</strong><br />

protein modulation by treatment were assessed by ANOVA. Results: LKB1<br />

mutant cell lines had lower expression <strong>of</strong> phosphorylated AMPK and TSC<br />

(p�0.01 for both) consistent with prior observations. In addition, mutant<br />

cell lines expressed higher levels <strong>of</strong> proteins in the IGF1R pathway<br />

including IGFR1b (p�0.0001); AIB1, which is known to upregulate IGF1<br />

(p�0.0001); and IGFBP2 (p�0.016). LKB1 mutant cell lines (n�11/25)<br />

were 1.5-fold more sensitive to the AMPK activator metformin, although<br />

this did not reach statistical significance (p�0.10). Expression levels <strong>of</strong><br />

IGF1R pathway proteins increased significantly after 48h treatment with<br />

metformin, the mTOR inhibitor temsirolimus, and the dual PI3K/mTOR<br />

inhibitor PI103. For example, IGFBP2 and AIB1 were elevated after<br />

metformin treatment (p�0.02 and 0.005, respectively); IRS1 and IGFR1<br />

were elevated after temsirolimus or PI103 (p�0.05 for both). Following<br />

treatment with metformin and temsirolimus, there was also increased pAkt,<br />

a downstream target <strong>of</strong> IGF1R and activator <strong>of</strong> mTOR (p�0.01 for both).<br />

Modulation <strong>of</strong> the IGF1R pathway by these drugs was independent <strong>of</strong> LKB1<br />

mutation status. Conclusions: LKB1 mutant cell lines have increased IGFR<br />

activity with higher baseline IGFR1, IGFB2 and AIB1, suggesting that IGFR<br />

may be a potential therapeutic target in LKB1 mutant tumors. In addition,<br />

inhibition <strong>of</strong> the mTOR pathway upregulates the IGFR pathway possibly as<br />

a feedback mechanism. These results support the investigation <strong>of</strong> IGFR<br />

inhibitors in combination with drugs targeting the mTOR pathway, particularly<br />

for tumors bearing LKB1 mutations.<br />

Tumor Biology<br />

683s<br />

10611 General Poster Session (Board #51D), Mon, 1:15 PM-5:15 PM<br />

Evaluation <strong>of</strong> EZH2 SNPs in cholangiocarcinoma patients. Presenting<br />

Author: Elisa Paolicchi, Department <strong>of</strong> Internal Medicine, University <strong>of</strong><br />

Pisa, Pisa, Italy<br />

Background: Cholangiocarcinoma (CCA) is an aggressive tumor arising from<br />

biliary tract epithelium.CCA is the second most common primary hepatic<br />

malignancy, with a progressive increasing incidence in western countries.<br />

Polycomb group protein Enhancer <strong>of</strong> Zeste homolog 2 (EZH2) is overexpressed<br />

in several human carcinomas, including CCA, where EZH2 overexpression<br />

is associated with tumor progression. The aim <strong>of</strong> this study is to<br />

evaluate the correlation between candidate EZH2 Single Nucleotide<br />

Polymorphisms (SNPs) with clinical outcome in CCA patients. Methods:<br />

Genomic DNA was extracted from blood samples <strong>of</strong> 75 patients [44 male<br />

and 31 female, with average age <strong>of</strong> 62.3 (range, 26-80 years)] affected by<br />

hystologically confirmed advanced CCA, treated with the epirubicincisplatin-xeloda<br />

(ECX) regimen. We performed an in silico characterization<br />

to select EZH2 SNPs (rs2302427 C/G, rs6464926 C/T, rs17171119 T/G<br />

and rs887569 C/T) from 26 EZH2 SNPs described previously. Genotyping<br />

was performed through Taqman PCR. Prognostic value <strong>of</strong> selected EZH2<br />

SNPs was assesses by correlation with time to progression (TTP) and overall<br />

survival (OS). OS and TTP curves were obtained through Kaplan-Meier<br />

method, and comparison with survival distribution was evaluated with<br />

logrank test. Results: Through specific s<strong>of</strong>tware (PROMO 3.0, MicroSNiper<br />

and Gene Card) we performed an in silico analysis based on functional<br />

characterization criteria (transcription factor binding-TFB, miRNA binding<br />

and missense mutations). We selected 4 EZH2 SNP alleles showing<br />

specific relevance in CCA because <strong>of</strong> their differential TFB affinity<br />

(PPAR�/RXR�, E2F-1, Pax-5 and p53). The rs887569 C/T SNP was<br />

significantly associated with clinical outcome. The TT genotype predicted a<br />

significantly longer OS in CCA patients (TT vs CT-CC p�0.026). Moreover,<br />

the TT genotype showed a trend-like association with reduced risk <strong>of</strong> death<br />

(HR�0.59, 95%CI�0.33-1.05, p�0.075), at multivariate analysis.<br />

Conclusions: These results suggest the role <strong>of</strong> rs887569 EZH2 SNP as a<br />

possible predictive marker <strong>of</strong> OS in advanced CCA patients treated with<br />

ECX regimen, and <strong>of</strong>fer a potential new tool for treatment optimization.<br />

10613 General Poster Session (Board #51F), Mon, 1:15 PM-5:15 PM<br />

Evaluation <strong>of</strong> circulating biomarkers <strong>of</strong> bone metastasis in early-stage<br />

breast cancer. Presenting Author: Windy Marie Dean-Colomb, University <strong>of</strong><br />

Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: Bone is the preferred site for metastasis <strong>of</strong> breast cancer,<br />

affecting approximately 70% <strong>of</strong> women with advanced disease. N-terminal<br />

<strong>of</strong> procollagen type 1 (P1NP), c-terminal peptide crosslinks (CTX), Osteocalcin<br />

(OC) and interleukin-6 (IL-6) are markers <strong>of</strong> bone turnover that may<br />

have clinical utility as predictors <strong>of</strong> breast cancer recurrence in the bone.<br />

Methods: Serum was collected from 168 patients with stage I/II/III breast<br />

cancer prior to treatment from 09/2001 to 12/2008 and stored at -80 C.<br />

Serum levels <strong>of</strong> P1NP, CTX, OC and IL-6 were determined using the<br />

Roche’s Elecsys 2010 automated immunoassay system. Correlations <strong>of</strong><br />

biomarker levels with time to bone metastasis (BM) development were<br />

assessed with Cox proportional hazards regression analysis and the Kaplan-<br />

Meier method. Results: Among the 168 patients analyzed, 60 patients<br />

subsequently developed BM. The biomarkers all had skewed distributions<br />

with long right tails and thus were all analyzed on the log scale. Residual<br />

analysis suggested non-linear relationships between each biomarker and<br />

risk <strong>of</strong> developing BM during follow-up. Thus, we fit Cox proportional<br />

hazards regression models for each biomarker with quadratic polynomials<br />

(on the log scale). On univariate analysis, these analyses generated p �<br />

0.33 for IL-6, 0.26 for osteocalcin, 0.40 for CTX, and 0.032 for P1NP.<br />

Adjusting for clinical factors (stage, age, race, post menopausal, ER/PR<br />

status, HER2 status, nuclear grade) yielded p � 0.0035 for the quadratic<br />

polynomial for log P1NP. A cut-point <strong>of</strong> 75 ng/mL identified patients with a<br />

short time to development <strong>of</strong> BM. The 1, 3, and 5-year freedom-from-BM<br />

probabilities were 96%, 77% and 66% for the 150 patients with P1NP<br />

values � 75 ng/mL and 88%, 45%, and 36% for the 16 patients with<br />

P1NP values � 75 ng/mL. The hazard ratio comparing patients with P1NP<br />

values � 75 ng/mL to patients with P1NP values � 75 ng/mL was 3.0<br />

(95% CI, 1.5 - 6.2) and p � 0.0075 ng/mL. After adjustment for clinical<br />

factors, the hazard ratio was 3.4 (1.5, 7.6) with p � 0.0026. Conclusions:<br />

Serum P1NP levels �75 ng/mL correlate with a shorter time to development<br />

<strong>of</strong> BM in patients with stage I-III breast cancer.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


684s Tumor Biology<br />

10614 General Poster Session (Board #51G), Mon, 1:15 PM-5:15 PM<br />

Isolation and expansion <strong>of</strong> circulating tumor cells (CTC) from melanoma<br />

patients using a novel cell culture technique. Presenting Author: John R.<br />

McGregor, TrueCells, LLC, Las Vegas, NV<br />

Background: Identification <strong>of</strong> rare (�2-5) circulating tumor cells (CTC) in<br />

7.5 ml blood by immun<strong>of</strong>luorescence assay (IFA) correlates with a poor<br />

prognosis in colon, breast, prostate and lung cancer. Changes in CTC count<br />

during treatment also predict the eventual patient progression and survival<br />

in these cancers. Existing assays do not detect melanoma CTC, however. In<br />

addition, isolation <strong>of</strong> viable CTC remains problematic. To overcome these<br />

limitations we attempted to develop novel melanoma CTC assays, using IFA<br />

and cell culture approaches. Methods: Blood samples were obtained from<br />

patients and controls following informed consent. The buffy coat (white<br />

cells � tumor) was isolated by Ficoll/Hypaque centrifugation, and split into<br />

6 replicate cultures in proprietary TrueCells medium. After 21 days in<br />

culture, tumor colonies were counted, and stained for melanoma and<br />

leukocyte markers. Buffy coat cells from parallel blood samples were<br />

stained with a panel <strong>of</strong> CSPG4-specific mAb (a pan-melanoma marker) on<br />

ultraclean glass slides for analysis by immun<strong>of</strong>luorescence microscopy.<br />

Results: Blood samples were obtained from 16 melanoma patients, ages<br />

28-87. Eight patients were men and 8 were women. CSPG4 � events (�2)<br />

were detected in 8/16 patients by IFA (range 0-52). In contrast, tumor cell<br />

colonies <strong>of</strong> �50 cells grew in 12 out <strong>of</strong> 16 patients with Stage 3 or 4<br />

melanoma (range 0-1054), shown in Table. Cells isolated from CTC<br />

colonies produced melanin, stained for CSPG4 and other melanoma<br />

markers, but not for leukocyte markers. Control cultures grew no tumor<br />

colonies. Conclusions: Our pilot study shows that melanoma CTC can be<br />

identified by both IFA and cultured from blood in many patients with stage<br />

3 or 4 melanoma. These CTC exhibited cytologic characteristics diagnostic<br />

<strong>of</strong> melanoma. The culture assay may represent a useful means <strong>of</strong> enumerating,<br />

isolating, and expanding viable melanoma CTC for further molecular<br />

study.<br />

Melanoma patients<br />

IFA Culture<br />

Pos assay Median Mean St dev Pos culture Median Mean St dev<br />

Control 0 <strong>of</strong> 5 0.0 0.0 0.0 0 <strong>of</strong> 5 0.4* 0.4* 0.8<br />

Stage 1-2 0 <strong>of</strong> 1 0.0 0.0 0.0 0 <strong>of</strong> 1 0.0 0.0 0.0<br />

Stage 3 1 <strong>of</strong> 4 1.0 1.3 1.3 3 <strong>of</strong> 4 320.5 285.3 209.6<br />

Stage 4 7 <strong>of</strong> 11 3.0 8.3 15.0 9 <strong>of</strong> 11 500.0 343.4 221.8<br />

*Non-tumor.<br />

10616 General Poster Session (Board #52A), Mon, 1:15 PM-5:15 PM<br />

Role <strong>of</strong> proliferation in response to neoadjuvant chemotherapy in GEICAM/<br />

2006-03 and GEICAM/2006-14 breast cancer patients. Presenting Author:<br />

E. Alba, Hospital Clínico Universitario Virgen de la Victoria, Málaga,<br />

Spain<br />

Background: Ki67 proliferation biomarker determined by immunohistochemistry<br />

(IHC) has been studied as a prognostic and predictive factor in<br />

Operable Breast Cancer (OBC). Ki67 modifications after neoadjuvant<br />

endocrine therapy have been correlated with long term outcome. However,<br />

there is no robust data about its predictive role in Neoadjuvant Chemotherapy<br />

(NC). In this study, we investigated Ki67 value as predictor <strong>of</strong> NC<br />

efficacy. Methods: 193 patients (pts) from 2 GEICAM phase II randomized<br />

trials (2006-03 and 2006-14) were included: 78 (40%) received epirubicine<br />

plus cyclophosphamide followed by docetaxel (EC-D), 41 (21%) EC-D<br />

plus carboplatin, and out <strong>of</strong> the 74 HER2� pts, 37 (19%) received EC-D<br />

plus tratuzumab and 37 (19%) EC-D plus lapatinib. Median age was 49<br />

years. From series, 87% were invasive ductal carcinoma, 58% premenopausal,<br />

50% grade III, 23% luminal , 39% basal and 38% HER2�. Ki67<br />

was centrally assessed by IHC (MIB1 clone) and median score was 40%<br />

(range 1-100%). Pathological Complete Response (pCR), defined as<br />

absence <strong>of</strong> invasive cells in breast and lymph nodes, was achieved in 56 pts<br />

(29%). Univariate and multivariate logistic regression models were used to<br />

study the association <strong>of</strong> each clinical-pathological variable with pCR. ROC<br />

curves were used to determine the most accurate ki67 cut-<strong>of</strong>f for predicting<br />

NC response. Results: Ki67�50% was defined as the most accurate<br />

threshold to select patients obtaining benefit from NC. In the univariate<br />

analysis, histological grade (p�0.01), treatment (P�0.006), ER<br />

(p�0.0001), PR (p�0.0001), HER2 (p�0.01), and Ki67�50%<br />

(p�0.0003) were statistically associated with pCR. A multivariate logistic<br />

regression showed that only Ki67� 50% (p�0.0003; OR�5.4 CI95%<br />

2.1-13.4), ER (p�0.0001; OR�0.2 CI95% 0.1-0.4), and HER2 status<br />

(p�0.0001; OR�8.8 CI95% 3.3-23.6) were predictive for pCR<br />

(AUC�0.7812). Conclusions: These results suggest that a high proliferation<br />

in breast cancer measured by Ki67 marker is an independent<br />

predictive factor for pCR in an unclassified HER2 population <strong>of</strong> OBC<br />

patients treated with NC.<br />

10615 General Poster Session (Board #51H), Mon, 1:15 PM-5:15 PM<br />

Zoledronic acid effect on circulating RANK/RANK-l/OPG axis in cancer<br />

patients. Presenting Author: Toni Ibrahim, Osteoncology Center, Istituto<br />

Scientifico Romagnolo per lo Studio e la Cura dei Tumori, Meldola, Italy<br />

Background: Bone metastasis disrupts bone integrity through loss <strong>of</strong> the<br />

balance between osteoclastic-mediated osteolysis and osteoblastic osteogenesis.<br />

The RANK/RANK-L/OPG axis governs osteoclastogenesis and bone<br />

resorption. We evaluated the trend <strong>of</strong> markers over time. Secondary aims<br />

were to study the predictive role <strong>of</strong> different circulating markers in the<br />

response to Zoledronic Acid (ZA) with respect to objective response and to<br />

skeletal-related events (SREs). Methods: The study prospectively evaluated<br />

levels <strong>of</strong> RANK, RANKL and OPG transcripts by Real-Time PCR and VEGF<br />

and NTX expression by ELISA in the peripheral blood <strong>of</strong> 49 consecutive<br />

patients with advanced breast, prostate or lung cancer (36, 7 and 6,<br />

respectively). Enrolled patients were at first diagnosis <strong>of</strong> bone metastases<br />

and had not previously undergone treatment with bisphosphonates. Patients<br />

received the standard schedule <strong>of</strong> ZA (4 mg infusion every 28 days)<br />

for about 12 months, undergoing blood tests and instrumental evaluation<br />

before the first infusion <strong>of</strong> ZA and every 4 months thereafter. Results:<br />

Median RANKL values after 12 infusions <strong>of</strong> ZA had decreased by 22% with<br />

respect to baseline whereas OPG, had increased by about 96%, with a 56%<br />

decrease in the RANKL/OPG ratio. NTX decreased over time (p�0.0001).<br />

On the basis <strong>of</strong> ROC curve analysis, RANKL was the most accurate marker<br />

for bone response with an AUC <strong>of</strong> 0.74 (95% CI 0.54-0.93). No<br />

correlations were found between circulating markers and SREs. Conclusions:<br />

The present work is one <strong>of</strong> the few prospective studies carried out on the<br />

circulating markers that are potentially associated with bone metastases.<br />

Our findings would seem to indicate that ZA decreases osteoclast activity<br />

through RANK/RANKL/OPG pathway modulation and that RANKL may play<br />

a role in the prediction <strong>of</strong> objective response to ZA. Confirmation <strong>of</strong> results<br />

is needed in a larger case series.<br />

10617 General Poster Session (Board #52B), Mon, 1:15 PM-5:15 PM<br />

Predictive impact <strong>of</strong> RRM1 protein expression on vinorelbine efficacy in<br />

NSCLC patients randomized in a chemotherapy phase III trial. Presenting<br />

Author: Adam Vilmar, Department <strong>of</strong> Oncology, Finsen Centre, National<br />

University Hospital, Copenhagen, Denmark<br />

Background: Platinum based doublets remain the cornerstone <strong>of</strong> treatment<br />

in non-small cell lung cancer (NSCLC) and <strong>of</strong>ten includes gemcitabine. A<br />

biomarker predicting sensitivity to this antimetabolite would represent a<br />

major step forward in the management <strong>of</strong> advanced disease. Ribonucleotide<br />

reductase subunit M1 (RRM1) has been <strong>of</strong> some value in NSCLC but<br />

evidence is not uniform. Accordingly, we explored the predictive role <strong>of</strong><br />

RRM1 in patients with advanced NSCLC. Methods: 443 patients with<br />

advanced NSCLC were randomized in a phase III trial to regimen A<br />

(paclitaxel and cisplatin with gemcitabine) or regimen B (cisplatin and<br />

vinorelbine). Immunohistochemical evaluation <strong>of</strong> RRM1 was performed on<br />

mainly bioptic material and correlated to response rates, progression free<br />

survival (PFS) and overall survival (OS). Results: 261 (58.9%) patients had<br />

representative tissue samples for RRM1 evaluation. Disease control rate,<br />

PFS and OS were significantly improved in patients with RRM-negative<br />

tumors receiving regimen B as compared to patients with RRM-positive<br />

tumors (68.8% vs. 31.2%, P � 0.046 , 6.90 months (mths) (95% CI<br />

5.83-7.96) vs. 3.93 mths (95% CI 3.11-4.76), P � 0.000 and 11.57<br />

mths (95% CI 9.65-13.48) vs. 7.4 mths (95% CI 5.37-9.43), P � 0.002,<br />

respectively). However, this was not the case for patients receiving regimen<br />

A (67.6% vs. 32.4%, P � 0.366, 6.73 mths vs. 7.6 mths, P � 0.207,<br />

11.07 mths vs. 12.37 mths, P � 0.103, respectively ). Together with<br />

histological subtype, RRM1-positivity emerged as the only other significant<br />

prognostic variable with a hazard ratio <strong>of</strong> 1.56 (95% CI 1.38-2.35, P �<br />

0.033) in multivariate analysis for patients treated with regimen B.<br />

Conclusions: RRM1 protein expression was without predictive impact in<br />

patients treated with cisplatin, paclitaxel and gemcitabine. Surprisingly,<br />

the predictive power proved robust in the cisplatin and vinorelbine arm<br />

across classic endpoints confirmed by multivariate analysis. These findings<br />

may suggest that RRM1 is involved in vinorelbine sensitivity and warrant<br />

further research which will be presented shortly.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


10618 General Poster Session (Board #52C), Mon, 1:15 PM-5:15 PM<br />

Biomarkers that predict sensitivity to heat shock protein 90 inhibitors<br />

(HSP90i). Presenting Author: Komal L. Jhaveri, Memorial Sloan-Kettering<br />

Cancer Center, New York, NY<br />

Background: HSP90 is an attractive target in tumors as it mediates the<br />

maturation and stabilization <strong>of</strong> client oncoproteins. HSP90i are potentially<br />

active in a variety <strong>of</strong> tumors, but therapeutic benefit is confirmed in only a<br />

small subset to date. Predictive biomarkers could identify patients (pts)<br />

and/or tumors with sensitivity to HSP90i improving the therapeutic index.<br />

We explored potential biomarkers across multiple studies <strong>of</strong> HSP90i.<br />

Methods: Archived pre-treatment tumor specimens from pts treated with<br />

any <strong>of</strong> several HSP90i (17-AAG, 17-DMAG, CNF2024, retaspimycin &<br />

ganetespib) on 8 phase I/II trials at MSKCC from 1999 to 2011 were<br />

identified. The following antibodies were validated at MSKCC and tumor<br />

tissue was tested by immunohistochemistry (IHC) with results defined for<br />

each as: ER, PR & AR: �1% pos & �1% neg; HSP90 & HSP70: 0, 1� neg<br />

&2�, 3� pos; PTEN: 0, 1� neg&2�pos; HER2: 0, 1� neg, 2�<br />

equivocal, 3� pos; EGFR: 0 neg & 1�,2�,3�pos. <strong>Clinical</strong> response was<br />

correlated with IHC using Fisher’s exact test. Results: Of the 164 pts<br />

identified, adequate tissue was available for 51, including 11 different<br />

solid tumors & 1 CML. Breast (N�31), melanoma & prostate (N�4 each)<br />

were most frequent. The mean age at primary diagnosis was 50 yrs (24-81).<br />

The median no. <strong>of</strong> lines <strong>of</strong> prior chemotherapy for metastatic disease was 2<br />

(0-8). The mean duration <strong>of</strong> HSP90i therapy was 79 days (0-411). There<br />

were 19 responses (5 PR; 14 SD); 14/19 responses strongly correlated with<br />

HER2� status (p � 0.001); 1 pt with ER�/HER2-/EGFR- & 4 with EGFR<br />

�/HER2-/ER- disease also responded. A correlation for ER & EGFR with<br />

response was not excluded (p�0.07 & p�0.086 respectively). Conclusions:<br />

Our findings confirm HER2 as a sensitive client and an effective biomarker<br />

for sensitivity to HSP90 inhibitors. ER & EGFR did not meet statistical<br />

significance but may warrant further exploration in prospective settings.<br />

10620 General Poster Session (Board #52E), Mon, 1:15 PM-5:15 PM<br />

Serum activinA and TGF-� as biomarkers <strong>of</strong> breast cancer bone metastasis<br />

behavior. Presenting Author: Christina L. Addison, Ottawa Hospital Research<br />

Institute, University <strong>of</strong> Ottawa, Ottawa, ON, Canada<br />

Background: Bisphosphonates (BP) are prescribed to pts with metastatic<br />

bone disease every 3-4 weeks regardless <strong>of</strong> individual risk for skeletal<br />

related events (SREs). In an era <strong>of</strong> personalized medicine this “one size fits<br />

all” approach is not appropriate and novel markers <strong>of</strong> SRE risk are required.<br />

TRIUMPH is an ongoing clinical trial evaluating 12 weekly IV BP therapy<br />

for 1 year in women with low risk bone metastases from breast cancer (BC)<br />

as defined by the bone resorption marker C-telopeptide (CTx,) levels �600<br />

ng/L. This sub-study evaluated the utility <strong>of</strong> novel biomarkers in better<br />

predicting the risk <strong>of</strong> developing SREs. Methods: Serum obtained from pts<br />

at baseline and 6 weeks post-entry were analyzed for tumor growth factor-�<br />

(TGF-�) and activinA levels by ELISA (sensitivity ~15-30 pg/ml). Levels<br />

were correlated with pt parameters including time to development <strong>of</strong> bone<br />

metastasis, and number <strong>of</strong> previous SREs using linear regression analysis.<br />

Changes in levels <strong>of</strong> biomarkers from baseline to 6 weeks were used to<br />

calculate odds ratios using logistic regression analysis. Results: Baseline<br />

activinA correlated with baseline CTx and bone specific alkaline phosphatase<br />

(p�0.004 and p�0.0001 respectively). Baseline activinA also correlated<br />

with weight (p�0.02), BMI (p�0.007) and trended towards total<br />

number <strong>of</strong> prior SREs (p�0.07). Baseline TGF-� correlated with pt age<br />

(p�0.02), weight (0.006), BMI (p�0.0005) and duration <strong>of</strong> metastatic<br />

bone disease (p�0.004), but did not correlate with any other biomarker.<br />

Change in activinA (baseline to week 6) was the only biomarker that<br />

trended to predict coming <strong>of</strong>f study early (p�0.053) as per protocol (i.e.<br />

CTx�600 ng/ml, SREs or pt/physician choice). Conclusions: Baseline levels<br />

<strong>of</strong> activinA trended to predict incidence <strong>of</strong> SREs in patients with bone<br />

metastases, and changes in levels from baseline to 6 weeks trended to<br />

predict coming <strong>of</strong>f study early. These findings warrant future studies in BC<br />

pts assessing activinA as a predictor <strong>of</strong> risk associated with breast cancer<br />

bone metastases. This study was conducted with the support <strong>of</strong> the Ontario<br />

Institute for Cancer Research through funding provided by the Government<br />

<strong>of</strong> Ontario, and with funding from the Ontario Chapter <strong>of</strong> the Canadian<br />

Breast Cancer Foundation.<br />

Tumor Biology<br />

685s<br />

10619 General Poster Session (Board #52D), Mon, 1:15 PM-5:15 PM<br />

Effect <strong>of</strong> everolimus on angiogenic biomarkers in patients with tuberous<br />

sclerosis complex (TSC): Results from EXIST-1 and EXIST-2. Presenting<br />

Author: David Neal Franz, Cincinnati Children’s Hospital Medical Center,<br />

Cincinnati, OH<br />

Background: The efficacy and safety <strong>of</strong> everolimus, an oral mTOR inhibitor,<br />

was assessed in two randomized, double-blind, placebo-controlled, phase<br />

3 trials: EXIST-1 (NCT00789828) and EXIST-2 (NCT00790400). EX-<br />

IST-1 examined everolimus for the treatment <strong>of</strong> subependymal giant cell<br />

astrocytoma (SEGA) associated with TSC and EXIST-2 for the treatment <strong>of</strong><br />

renal angiomyolipoma (AML) associated with either TSC or sporadic<br />

lymphangioleiomyomatosis. In each instance, everolimus was superior to<br />

placebo for the primary endpoints, SEGA and renal AML response rates.<br />

Inhibitors <strong>of</strong> mTOR have antiangiogenic effects on tumor growth in vitro<br />

and in vivo. Methods: Patients were randomized to receive everolimus<br />

(n�78) starting at 4.5 mg/m2 /day (target trough, 5-15 ng/mL) or placebo<br />

(n�39) in EXIST-1 and 10 mg/day everolimus (n�79) or placebo (n�39)<br />

in EXIST-2. Plasma samples were taken at baseline and pre-dosing on day<br />

1 <strong>of</strong> weeks 4, 12, 24, 36, and 48 <strong>of</strong> treatment. Angiogenic markers <strong>of</strong><br />

interest were vascular endothelial growth factor (VEGF)-A and -D, placental<br />

growth factor (PlGF), soluble VEGF receptor-1 (sVEGFR1), soluble VEGF<br />

receptor 2 (sVEGFR2), c-Kit, and collagen type IV. Results: Compared with<br />

placebo, a sustained ~30% and ~60% increase in VEGF-A was observed in<br />

the everolimus arm <strong>of</strong> EXIST-1 and EXIST-2, respectively. A concomitant<br />

decrease in collagen type IV (~25% EXIST-1; ~45% EXIST-2) and<br />

sVEGFR2 (~25% both trials) was also observed in the everolimus arm. A<br />

sustained decrease (~60%) in VEGF-D was observed in the everolimus arm<br />

<strong>of</strong> EXIST-2, but not EXIST-1. In both studies, no change was observed in<br />

PlGF, sVEGFR1, or c-Kit plasma concentrations in the everolimus arm or<br />

any biomarkers evaluated in the placebo arm. Baseline sVEGFR2 and<br />

VEGF-D were ~40% and ~4-fold higher, respectively, while VEGF-A was<br />

~50% lower in EXIST-2 compared with EXIST-1. A similar baseline plasma<br />

concentration for the other biomarkers was noted in both studies.<br />

Conclusions: Patients presenting with SEGA or renal AML associated with<br />

TSC had a reduction in plasma concentrations <strong>of</strong> sVEGFR2, collagen type<br />

IV, and VEGF-D (AML only) and an increase in VEGF-A. Everolimus may<br />

have antiangiogenic properties in TSC patients.<br />

10621 General Poster Session (Board #52F), Mon, 1:15 PM-5:15 PM<br />

Identification and validation <strong>of</strong> plasma protein biomarker panels for breast<br />

cancer diagnosis by using multiple reaction monitoring-based mass spectrometry.<br />

Presenting Author: Hyeong-Gon Moon, Department <strong>of</strong> Surgery,<br />

Seoul National University Hospital, Seoul, South Korea<br />

Background: Multiple reaction monitoring-based mass spectrometry (MRM-<br />

MS) has the ability to perform a wide range <strong>of</strong> proteome analysis in a single<br />

experiment using a small volume <strong>of</strong> specimen. We aimed to develop a<br />

plasma protein signature for breast cancer diagnosis using the MRM-MS<br />

technology. Methods: Previously, we have identified lists <strong>of</strong> breast cancerrelated<br />

proteins from various models <strong>of</strong> proteomic discovery including<br />

cancer plasma vs healthy plasma, cancer cell line secretome vs nontumorigenic<br />

cell line secretome, cancer tissue vs normal tissue, and<br />

literature search. Based on these protein panels, total <strong>of</strong> 29 proteins were<br />

selected for further experiments. We verified and validated the protein<br />

signature in two independent cohorts <strong>of</strong> breast cancer patients and healthy<br />

women. Results: In the verification cohort <strong>of</strong> 80 breast cancer patients and<br />

80 healthy women, MRM-MS showed significant differences in plasma<br />

concentration for 11 proteins. Among them, the difference was not<br />

significant for 4 proteins when the cases were limited to stage I and II<br />

patients. Based on p values and consistent expression level along the AJCC<br />

stages, we have created a plasma protein signature comprised <strong>of</strong> 3 plasma<br />

proteins. The 3 plasma protein signature effectively discriminated cancer<br />

and healthy cases with the AUC <strong>of</strong> 0.831 (sensitivity 78.7%, specificity<br />

78.7%). The performance <strong>of</strong> the 3 plasma protein signature was validated<br />

in the cohort <strong>of</strong> 100 cancer patients and 100 healthy women. The accuracy<br />

<strong>of</strong> the 3 protein signature was still meaningful with the AUC <strong>of</strong> 0.746 and<br />

0.797 for all stages and stage I or II patients, respectively. Conclusions: The<br />

3 plasma protein signature for breast cancer diagnosis, developed by the<br />

MRM-MS technology, showed promising results in the present study.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


686s Tumor Biology<br />

10622 General Poster Session (Board #52G), Mon, 1:15 PM-5:15 PM<br />

Diurnal proteomics and biomarker discovery in indolent versus aggressive<br />

chronic lymphocytic leukemia patients. Presenting Author: Georg A.<br />

Bjarnason, Sunnybrook Odette Cancer Centre, University <strong>of</strong> Toronto,<br />

Toronto, ON, Canada<br />

Background: In rodents, 10-20% <strong>of</strong> the genome has a 24-hour (h) rhythm in<br />

RNA expression. A molecular clock consisting <strong>of</strong> transcription / translation<br />

feedback loops <strong>of</strong> clock-genes controls this rhythmicity. In a microarray<br />

study (Affymetrix HG_U133_Plus2 chip) on RNA extracted from CLL cells<br />

sampled every 4 hours over 24 hours (6 samples) from 3 male (M) and 6<br />

female (F) patients (pts) with chronic lymphocytic leukemia (CLL) we found<br />

15,094 and 13,415 rhythmic transcripts (Cosinor analysis) in M and F<br />

respectively, only 6,629 <strong>of</strong> which were common to both M and F. We<br />

hypothesized that these gender differences in rhythmic RNA expression<br />

might have clinical implication for biomarker discovery with some important<br />

biomarkers only found at a certain times <strong>of</strong> day and with gender<br />

differences. Methods: Sampling was performed again at 6 time points in M<br />

and F CLL patients with indolent CLL (10pts, 5 M, 5F) and aggressive CLL<br />

(10 pts, 5M, 5F). An 8-plex iTRAQ kit was employed for mass spectrometry<br />

(MS) based relative quantification. Each iTRAQ block comprised all<br />

time-points for a single patient including a universal control. Results: At<br />

least 300 proteins were identified at a 95% confidence level in each iTRAQ<br />

experiment. Of these, 74 proteins displayed significant change between<br />

aggressive and indolent pts in 24-hour average or single time point<br />

normalized expression based on a non-parametric U-test (p�0.05). The<br />

JTK_CYCLE algorithm was used to detect 24-hour rhythmic patterns in the<br />

proteomic datasets. Significant rhythmic expression was found for 44<br />

proteins (p�0.10) identified by iTRAQ. Data for each time point were<br />

independently analyzed using an in-house Butterfly clustering algorithm<br />

based on discrete dynamical systems. The 4 PM time point was found to be<br />

optimal for stratifying aggressive and indolent disease. Cellular pathways<br />

with significant association to differentially expressed proteins included<br />

PPAR signaling, fatty acid metabolism, and granzyme-A signaling. Confirmation<br />

by selected reaction monitoring (SRM) MS is ongoing. Conclusions:<br />

Rhythmic protein expression may have clinical implications for biomarker<br />

discovery.<br />

10624 General Poster Session (Board #53A), Mon, 1:15 PM-5:15 PM<br />

Synthetic lethality <strong>of</strong> EGFR blockade in the context <strong>of</strong> tumor hypoxia.<br />

Presenting Author: Haruhiko Makino, University <strong>of</strong> Texas SouthWestern<br />

Medical Center Department <strong>of</strong> Radiation Oncology, Dallas, TX<br />

Background: Hypoxia induces a wide spectrum <strong>of</strong> biological responses in<br />

tumors that alter tumor radio-sensitivity. In several tumors, including<br />

non-small cell lung carcinoma (NSCLC), the epidermal growth factor<br />

receptor (EGFR) is frequently over-expressed and activated in response to<br />

hypoxia, although the precise role <strong>of</strong> EGFR in hypoxia-associated radioresitance<br />

is unclear. Activating mutations in EGFR have been clinically linked<br />

to dramatic responses to EGFR tyrosine kinase inhibitors. We previously<br />

demonstrated that NSCLCs harboring activating EGFR mutations are<br />

radiosensitive with severe deficits in the non homologous end-joining<br />

(NHEJ) repair <strong>of</strong> DNA damage. Methods: We investigated hypoxia associated<br />

radiation responses in a panel <strong>of</strong> NSCLC cell lines and immortalized<br />

human bronchial epithelial cells expressing wild type EGFR (WT) or<br />

expressed EGFR forms with somatic activating L858R or DE746-E750<br />

mutations (MT). Results: (1) Relative to WT EGFR, MT EGFR expression is<br />

associated with significantly higher radiosensitivity under chronic hypoxia<br />

(2) Under hypoxic conditions, MT EGFR expression is associated with a<br />

unique spectrum <strong>of</strong> DNA damage responses (DDR) that significantly<br />

deviated from canonical response <strong>of</strong> WT EGFR expressing NSCLCs (3)<br />

Genome wide gene expression analysis identified a dramatic hypoxiaassociated<br />

DDR shift to selective down-regulation <strong>of</strong> homologous recombination<br />

(HR). Moreover, MT EGFR deficits in NHEJ caused a synthetic<br />

lethality effect in the context <strong>of</strong> hypoxia through a simultaneous blockade<br />

<strong>of</strong> HR and NHEJ. (4) Blockade <strong>of</strong> WT EGFR by the anti-EGFR monoclonal<br />

antibody, cetuximab perfectly recapitulated the contextual synthetic lethality<br />

<strong>of</strong> MT EGFR expression. (5) Cetuximab had a dramatic synergistic effect<br />

on survival <strong>of</strong> rat lung orthotopic tumors where hypo-fractionated radiotherapy<br />

or cetuximab alone had marginally controlled tumors. Conclusions:<br />

The data support a critical role for EGFR-mediated DNA repair in hypoxia<br />

associated tumor radio-resistance. Furthermore, this role could be a target<br />

for refractory hypoxic EGFR-WT tumor especially in the context <strong>of</strong> hyp<strong>of</strong>ractionated<br />

radiotherapy which, in its current form, is potentially counterproductive<br />

for hypoxic tumors.<br />

10623 General Poster Session (Board #52H), Mon, 1:15 PM-5:15 PM<br />

New mechanistic insights into possible radiobiologic and pathophysiologic<br />

explanations for unexpectedly impressive outcomes data observed for<br />

hormone-refractary metastatic prostate cancer patients treated with radium<br />

223. Presenting Author: Roger M. Macklis, Cleveland Clinic Foundation<br />

and Taussig Cancer Center, Cleveland, OH<br />

Background: The ALSYMPCA Trial <strong>of</strong> 223Ra (“Alpharadin”) for men with<br />

metastatic castrate-resistant prostate cancer was a Phase III randomized<br />

trial comparing 223Ra plus SOC vs best SOC. This trial was stopped early<br />

after 922 patients enrolled at over 100 centers achieved primary (overall<br />

survival) and all secondary endpoints including median survived (14 vs<br />

11.2 m) time to first SRE (13.6 m vs 8.4 m) and time to PSA<br />

progression.These unexpectedly positive results prompted us to review<br />

some <strong>of</strong> the unique aspects <strong>of</strong> alpha-particle radiobiology and ultra-short<br />

range molecular targeting capabilities. Methods: Literature analysis. Results:<br />

The highly energetic alpha particles such as those released during decay <strong>of</strong><br />

223Ra (T1/2 � 11.4 mins) display unique radiobiologic and dosimetric<br />

aspects. The nuclear dose required in vitro for approximately 90% target<br />

cell kill is less than 1 Gy and electron micrographs shortly after exposure<br />

confirm bizarre blebbing and condensation <strong>of</strong> chromosomal material<br />

characteristic <strong>of</strong> apoptosis. Bovine aortic endothelial cells exposed to alpha<br />

particle radiation indicate that the radiobiologic effects depend heavily on<br />

cell culture condition with nonuniform DNA strand break damage observed<br />

in isolated detached cells. The subcellular alpha particle path length<br />

appears to allow specific target cell populations to be killed within bony<br />

stroma without excessive damage to nearby sensitive hematopoietic cells.<br />

223Ra treatment in rodent models showed that this family <strong>of</strong> Ca �� analogs<br />

delivers extremely high doses to stromal elements such as bone-resorbing<br />

osteoclasts and this selectivity may aid in decreasing SREs. Conclusions:<br />

We highlight the fact that some basic radiobiologic and cell compartmentspecific<br />

stromal localization principles known to be effective in alpha<br />

particle radiopharmaceutical therapy may contribute to the very positive<br />

clinical outcomes observed while allowing subcellular damage accumulation<br />

to target cell groups.<br />

10625 General Poster Session (Board #53B), Mon, 1:15 PM-5:15 PM<br />

Using CHFR expression to predict response and survival after first line<br />

treatment with carboplatin-paclitaxel in NSCLC. Presenting Author: Rathi<br />

Narayana Pillai, Winship Cancer Institute, Emory University, Atlanta, GA<br />

Background: The identification <strong>of</strong> resistance mechanisms for conventional<br />

chemotherapy in lung cancer is <strong>of</strong> fundamental importance not only for<br />

personalization <strong>of</strong> chemotherapy but also for the subsequent development<br />

<strong>of</strong> novel targeted approaches to overcome this resistance. Currently, there<br />

is no clinically validated test for the prediction <strong>of</strong> response to tubulintargeted<br />

agents in NSCLC. Our previous preclinical work identified Checkpoint<br />

with Forkhead and Ringfinger domain” (CHFR) as a predictor <strong>of</strong><br />

taxane cytotoxicity in in vitro models. The current work assessed the<br />

translational significance <strong>of</strong> this finding in a cohort <strong>of</strong> US veterans treated<br />

at a single-institution. Methods: We studied a cohort <strong>of</strong> patients with<br />

advanced NSCLC treated with taxane-containing frontline regimens at the<br />

Atlanta VA Medical Center between 2000 and 2009. Archived paraffinembedded<br />

tissue was retrieved and stained for CHFR expression using<br />

standard immunohistochemical techniques. Level <strong>of</strong> protein expression<br />

was assessed by light microscopy and scored for intensity <strong>of</strong> CHFR staining.<br />

Intensity <strong>of</strong> staining was correlated with clinical outcome including<br />

objective response and median overall survival using Chi-Square test and<br />

Cox proportional models. Results: We analyzed tumor samples from 45<br />

eligible patients with median age 62.6 years, M/F (44/1). In this cohort,<br />

high expression <strong>of</strong> CHFR is strongly associated with adverse outcomes: the<br />

risk for progressive disease (PD) after first-line chemotherapy with carboplatin-paclitaxel<br />

was 54% in patients with CHFR-high vs. only 18% in those<br />

with CHFR-low tumors (HR 3.1; 95% CI 1.09-9.04; p�0.02). Median<br />

overall survival was strongly correlated with response to first-line therapy<br />

(clinical benefit: 9.24 months; PD: 4.7 months; p�0.001) and with CHFR<br />

expression status (CHFR low: 10 months; CHFR high: 5.6 months; p<br />

�0.01). Conclusions: CHFR expression is a novel predictive marker <strong>of</strong><br />

response and overall survival in NSCLC patients treated with taxanecontaining<br />

chemotherapy.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


10626 General Poster Session (Board #53C), Mon, 1:15 PM-5:15 PM<br />

DNA damage responses in relation to late effects following radiotherapy for<br />

early breast cancer. Presenting Author: Melvin Chua, University College<br />

London Cancer Institute, London, United Kingdom<br />

Background: In this study, we assessed if induction <strong>of</strong> apoptosis in G0 blood<br />

lymphocytes following ex vivo irradiation correlated with clinical radiosensitivity<br />

and DNA double-strand (DSB) repair in breast radiotherapy (RT)<br />

patients. Using small molecule inhibitors, we examined the relationship<br />

between DSB repair and radiation-induced apoptosis. Methods: Breast<br />

cancer patients with minimal (controls) or marked late RT changes (cases)<br />

were selected. DSB were quantified by �H2AX/53BP1 immunostaining 0.5<br />

and 24 h after exposure <strong>of</strong> lymphocytes to 0.5 and 4 Gy, respectively.<br />

Induction <strong>of</strong> apoptosis was assessed 48 h after 8 Gy using a fluorochrome<br />

inhibitor <strong>of</strong> caspases (FLICA) assay. Apoptotic cells were defined by small<br />

cell size and positive FLICA signal, measured using flow cytometry. Small<br />

molecule DNA-PK (Nu7441) and ATM (Ku55933) inhibitors were used at<br />

concentrations <strong>of</strong> 1 �M and 10 �M, respectively. Results: Despite similar<br />

foci induction at 0.5 h, residual foci levels 24 h after 4 Gy differed<br />

significantly between cases and controls (Foci per cell � 12.7 in cases, n �<br />

8 vs 10.3 in controls, n � 8, p � 0.002). In contrast to residual foci levels,<br />

comparable levels <strong>of</strong> apoptosis were observed between cases and controls<br />

48 h after 8 Gy. Mean proportion <strong>of</strong> apoptotic cells was 37.2% in cases and<br />

34.7% in controls (p � 0.413). Residual foci levels were not correlated<br />

with levels <strong>of</strong> apoptosis in lymphocytes <strong>of</strong> the same patients (R � -0.059, p<br />

� 0.785). To determine if apoptosis is modulated by DSB repair, apoptosis<br />

measurements were repeated in lymphocytes treated with Nu7441 to<br />

inhibit non-homologous end-joining. We observed that Nu7441-treated<br />

cells had higher levels <strong>of</strong> residual foci and apoptosis compared to<br />

mock-treated cells, 48 h after 1 Gy. This effect was wholly dependent on an<br />

active ATM function as cells treated simultaneously with Nu7441 and<br />

Ku55933 had extremely low levels <strong>of</strong> apoptosis, comparable to levels<br />

observed in cells treated with Ku55933 alone. Conclusions: Higher levels <strong>of</strong><br />

residual DSB observed among radiosensitive patients suggest a role for<br />

DSB repair in the pathogenesis <strong>of</strong> late radiation-induced effects. Induction<br />

<strong>of</strong> apoptosis appears to be dependent on DSB end-joining following<br />

exposure to ionising radiation.<br />

10628 General Poster Session (Board #53E), Mon, 1:15 PM-5:15 PM<br />

In silico identification <strong>of</strong> an epithelial core signature in human tumors.<br />

Presenting Author: Chirayu Pankaj Goswami, Center for Computational<br />

Biology and Bioinformatics, Indiana University School <strong>of</strong> Medicine, Indianapolis,<br />

IN<br />

Background: Gene expression analysis is performed on grossly selected<br />

specimens <strong>of</strong>ten without any microscopic analysis <strong>of</strong> tumor content. In<br />

studies where histological analyses have been performed, cases having<br />

80% or more tumor content are used for microarray analysis. The variability<br />

in amount <strong>of</strong> epithelial and stromal cells may generate to misleading<br />

differential expression analysis and selection for wrong targets for therapeutics.<br />

It is also <strong>of</strong>ten unclear, whether the genes identified are stromal or<br />

epithelial in origin. The goal <strong>of</strong> this study was to identify genes that define<br />

core epithelial phenotype; these genes could provide means <strong>of</strong> normalization<br />

<strong>of</strong> expression data. Methods: The CABIG GSK microarray (HG-<br />

U133_plus_2) data consisting <strong>of</strong> 950 cell lines from carcinoma (n�562),<br />

non-carcinoma (n�385) and normal tissue (n�3) was analyzed to identify<br />

epithelial specific genes. 10 carcinomas each from 11 sites (n�110) and<br />

an equal number <strong>of</strong> non-carcinomas were randomly selected. In silico<br />

analyses were performed by 1) identifying genes differentially expressed<br />

between carcinoma and non-carcinoma samples using a one way ANOVA;<br />

2) identifying gene signature associated with carcinoma using Predictive<br />

Analysis <strong>of</strong> Microarrays (PAM) and 3) a weighted gene coexpression<br />

network analysis (WGCNA) was performed to identify co-expression modules.<br />

A similar analysis was also performed on tissue samples (E-GEOD-<br />

12360) from carcinomas and non-carcinomas. Venn-diagram was generated<br />

to identify intersecting set. Results: Comparison <strong>of</strong> the carcinoma and<br />

non-carcinoma samples using ANOVA identified 1455 differential expressed<br />

gene probes in cell lines and 540 gene probes in tissues<br />

(FDR�1E-10). The cell lines analysis identified 5 modules and a 65-gene<br />

signature (43 core and 22 accessory set) that was specific for epithelial<br />

cells. In the tissue analysis a 188-gene signature was similarly identified.<br />

Cross-comparison identified a smaller 31 gene intersecting set; this was<br />

not associated with loss <strong>of</strong> discriminatory power. Conclusions: A 31 geneset<br />

which can be used to determine the epithelial content <strong>of</strong> heterogeneous<br />

tumors, was identified. This study has the potential to significantly impact<br />

the use <strong>of</strong> microarray based gene expression data.<br />

Tumor Biology<br />

687s<br />

10627 General Poster Session (Board #53D), Mon, 1:15 PM-5:15 PM<br />

Correlation between in vitro chemotherapy sensitivity and PARP-1 expression<br />

in patients with breast cancer. Presenting Author: Cornelia Liedtke,<br />

University <strong>of</strong> Muenster, Muenster, Germany<br />

Background: Based on its potential use as a therapeutic target through<br />

PARP inhibitors expression <strong>of</strong> Poly-A-Ribose-Polymerase-1 (PARP-1) has<br />

come into scientific focus. Furthermore, it could be demonstrated that<br />

cytoplasmic PARP-1 expression varies depending on molecular breast<br />

cancer subtypes and that it correlates with an increased response to<br />

neoadjuvant taxane-anthrazykline containing chemotherapy (von Minckwitz<br />

et al., J Clin Oncol 2011). In-vitro-chemotherapy sensitivity and<br />

resistance assays (CSRAs) are a means to directly measure chemotherapy<br />

sensitivity in a given tumor uninfluenced by host factors. Methods: We<br />

conducted a systematic immunohistochemical tissue-microarray (TMA)based<br />

analysis <strong>of</strong> 550 samples <strong>of</strong> invasive breast cancers to evaluate the<br />

expression <strong>of</strong> a set <strong>of</strong> molecular markers including estrogen receptor (ER),<br />

progesterone receptor (PR) and HER2 as well as PARP-1. Triple negative<br />

breast cancers (TNBC) were identified through lack <strong>of</strong> (over-)expression <strong>of</strong><br />

ER, PR and HER2. All carcinomas were analyzed in an in vitro CSRA<br />

protocol for epirubicin/docetaxel (ED) and epirubicin/cyclophosphamide<br />

(EC). In-vitro-chemotherapy sensitivity was analyzed using an adenosine<br />

triphosphate (ATP) bioluminescence assay. Results: Moderate/high expression<br />

<strong>of</strong> PARP-1 was found in 48 and 33% <strong>of</strong> cases with TNBC and<br />

non-TNBC, respectively (p�0.015). No correlation between TNBC phenotype<br />

and cytoplasmic expression was observed. However, increased expression<br />

<strong>of</strong> both cytoplasmic and nuclear PARP-1 was correlated with an<br />

increased in-vitro sensitivity against ED (p�0.012 and 0.025, respectively)<br />

but not EC (p�0.27 and 0.62, respectively). Conclusions: Our<br />

results support previous observations demonstrating a significant correlation<br />

between expression <strong>of</strong> PARP-1 and an increased sensitivity against<br />

taxane-anthracycline chemotherapy independent <strong>of</strong> tumor phenotype.<br />

10629 General Poster Session (Board #53F), Mon, 1:15 PM-5:15 PM<br />

Intestinal HIF-2 to protect against radiation-induced gastrointestinal syndrome.<br />

Presenting Author: Cullen M. Taniguchi, Stanford University<br />

Medical Center, Stanford, CA<br />

Background: Gastrointestinal (GI) toxicity accounts for significant morbidity<br />

during systemic chemotherapy or abdominopelvic radiation therapy. Unfortunately,<br />

there are no effective preventative treatments. Hypoxic signaling<br />

through hypoxia-inducible factor-1 (HIF-1) and -2 (HIF-2) is critical for<br />

intestinal homeostasis during inflammatory challenges and was posited to<br />

play a role in radioprotection. Methods: We stabilized both HIF-1 and HIF-2<br />

pharmacologically with the prolyl hydroxylase inhibitor DMOG and with<br />

transgenic mice. Male C57/Bl6 mice eight weeks <strong>of</strong> age were pretreated<br />

with saline or 8mg DMOG 16hrs and 4hrs prior to being irradiated with a<br />

single fraction <strong>of</strong> 20Gy to the abdomen using a modified TLI jig that shields<br />

the upper body <strong>of</strong> the mouse to reduce hematopoietic toxicity. After XRT,<br />

the mice got daily doses <strong>of</strong> saline or DMOG for 5 days. Results: Mice treated<br />

with DMOG had a 1.5-fold enrichment <strong>of</strong> crypt regeneration in the colon as<br />

determined by microcolony assay (13.3�3.4 vs 21.2�1.7 crypts/section,<br />

saline vs DMOG, n�10/group, p�0.005). Death from GI radiotoxicity can<br />

result from compromised epithelial integrity. We tested epithelial and<br />

barrier function by gavaging irradiated mice with FITC-dextran, which<br />

undetectable in the blood unless the GI epithelia is compromised.<br />

Treatment with DMOG decreased FITC-dextran uptake by 4-fold over<br />

controls (52.6�14.7 vs 12.4�3.4 mcg/ml, n�6/group, p�0.02). These<br />

physiologic improvements translated to improved overall survival with<br />

DMOG treatment after being exposed to 20Gy <strong>of</strong> abdominal radiation<br />

(p�0.03). To determine if the radioprotective effect <strong>of</strong> DMOG was specific<br />

to HIF-1 or HIF-2, we overexpressed stabilized HIF1 or HIF2 in the<br />

intestinal epithelia using a lox-stop-lox (LSL) system and Villin-Cre. The<br />

overexpression <strong>of</strong> HIF1 had no effect on survival (p�0.22), while mice with<br />

intestinal-specific HIF2-overexpression had significantly improved survival<br />

after receiving 20Gy <strong>of</strong> abdominal radiation (p�0.001). Conclusions:<br />

Activation <strong>of</strong> HIF-2 is sufficient to protect mice from lethal doses <strong>of</strong><br />

abdominal radiation may provide a novel class <strong>of</strong> treatments to protect the<br />

GI epithelium from the deleterious side effects <strong>of</strong> chemotherapy and<br />

radiation.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


688s Tumor Biology<br />

10630 General Poster Session (Board #53G), Mon, 1:15 PM-5:15 PM<br />

Molecular pr<strong>of</strong>iling <strong>of</strong> uveal melanoma patients. Presenting Author: Zoran<br />

Gatalica, Caris Life Sciences, Phoenix, AZ<br />

Background: Although uveal melanoma represents only 5% <strong>of</strong> all melanomas,<br />

it is the most common primary intraocular malignancy <strong>of</strong> the adult<br />

eye. Approximately 50% <strong>of</strong> patients will develop metastases which are<br />

resistant to medical interventions. There is a great need for improved<br />

therapy as the prognosis is poor for advanced stages. Our study was<br />

undertaken to investigate the presence <strong>of</strong> novel therapeutic targets.<br />

Methods: We analyzed 49 uveal melanoma patients with immunohistochemistry<br />

for 16 markers including cKIT, PDGFR, cMET, PTEN and IGF1R.<br />

Further, microarray analysis was performed on 29 samples using the<br />

Illumina platform. We also investigated amplification <strong>of</strong> EGFR and mutational<br />

analysis <strong>of</strong> cKIT, BRAF, KRAS and NRAS on a smaller patient subset.<br />

Results: Overexpression <strong>of</strong> KIT at the protein and RNA level was 74% (28<br />

out <strong>of</strong> 38) and 45% (13 out <strong>of</strong> 29), respectively. Expression <strong>of</strong> cKIT did not<br />

correlate with gain-<strong>of</strong>-function cKIT mutations in any <strong>of</strong> the 34 samples<br />

tested. In our study, MET was overexpressed in 15 out <strong>of</strong> 17 cases at the<br />

RNA level and IGF1R was high in 4 out <strong>of</strong> 6 patients indicating poor<br />

prognosis. PTEN expression by IHC was present in 90% (36 out <strong>of</strong> 40)<br />

patients indicating the PI3K pathway is not activated in the majority <strong>of</strong><br />

uveal melanoma patients. BRAF was wildtype in all 42 patients tested.<br />

Similarly, no KRAS or NRAS mutations were detected. Protein and RNA<br />

expression <strong>of</strong> PDGFR were low in our patients. MGMT was lost in 16 out <strong>of</strong><br />

40 patients at the protein level and 10 out <strong>of</strong> 29 patients at the RNA level.<br />

EGFR expression, copy number and protein levels were low in the patients<br />

tested. Conclusions: Our data on cKIT suggests that it is a promising target<br />

in uveal melanoma. Low expression <strong>of</strong> MGMT in about a third <strong>of</strong> our<br />

patients may indicate the likelihood <strong>of</strong> favorable response to dacarbazine<br />

and temozolomide. There are currently several clinical trials investigating<br />

various cKIT inhibitors, as well as temozolomide in advanced uveal<br />

melanoma patients. Our findings highlight the importance <strong>of</strong> molecular<br />

pr<strong>of</strong>iling uveal melanoma patients.<br />

TPS10632 General Poster Session (Board #54A), Mon, 1:15 PM-5:15 PM<br />

The PARSC trial, a prospective study for the assessment <strong>of</strong> recurrence risk<br />

in stage II colon cancer patients using ColoPrint. Presenting Author: Ramon<br />

Salazar, Institut Catala d’Oncologia, Barcelona, Spain<br />

Background: An 18-gene expression pr<strong>of</strong>ile, ColoPrint, has been developed<br />

for identifying colon cancer patients more likely to develop recurrent<br />

disease and who would be candidates for adjuvant chemotherapy. The gene<br />

signature was validated in public datasets and independent patient cohorts<br />

(stage II and III patients). Uni-and multivariate analysis was performed on<br />

the pooled stage II patient set (n�320) (median follow-up 70 months).<br />

ColoPrint classifies 65% <strong>of</strong> stage II patients as Low Risk. The 3-year RFS<br />

was 91% for Low Risk and 74% for High Risk patients with a HR <strong>of</strong> 2.9<br />

(p�0.001). ColoPrint was the only significant prognostic marker in the<br />

subgroup <strong>of</strong> patients with T3-MSS phenotype (Tabernero, ASCO GI 2012).<br />

Methods: A blinded prospective trial, PARSC (Prospective study for the<br />

Assessment <strong>of</strong> Recurrence risk in Stage II colon cancer patients) using<br />

ColoPrint has been initiated. Objectives are: (1)To validate the performance<br />

<strong>of</strong> ColoPrint in estimating the 3-year relapse rate in patients with<br />

stage II colon cancer. (2) To compare the risk assessment in stage II<br />

patients using the ColoPrint pr<strong>of</strong>ile vs a clinical risk assessment based on<br />

Investigator’s assessment <strong>of</strong> risk and ASCO high-risk recommendations. (3)<br />

To investigate therapy as a potential confounding factor for ColoPrint<br />

results. (4) To assess the performance <strong>of</strong> ColoPrint in estimating the 3-year<br />

relapse rate in patients with stage III colon cancer. Inclusion criteria: age �<br />

18 years, adenocarcinoma <strong>of</strong> the colon, stage II and III, no prior neoadjuvant<br />

therapy, no synchronous tumors, fresh tumor sample, and written<br />

informed consent. The treatment <strong>of</strong> the patient is at the discretion <strong>of</strong> the<br />

physician adhering to National Comprehensive Cancer Network (NCCN)approved<br />

regimens or a recognized alternative (blinded for ColoPrint<br />

result). The trial started in Sept. 2008 with currently 32 participating sites<br />

in 11 countries. Thus far, 340 eligible stage II and 280 stage III patients<br />

have been enrolled. The aim is to enroll 575 stage II patients. <strong>Clinical</strong> trial<br />

registry number: NCT00903565.<br />

TPS10631 General Poster Session (Board #53H), Mon, 1:15 PM-5:15 PM<br />

Detection <strong>of</strong> discordant HER2 status by FISH in circulating tumor cells and<br />

disseminated tumor cells in early-stage breast cancer using a micr<strong>of</strong>luidicbased<br />

cell enrichment and extraction platform (OncoCEE). Presenting<br />

Author: Savitri Krishnamurthy, University <strong>of</strong> Texas M. D. Anderson Cancer<br />

Center, Houston, TX<br />

Background: Evaluation <strong>of</strong> HER2 in circulating (CTCs) and disseminated<br />

(DTC) tumor cells may aid therapy in breast cancer. We report here the<br />

discordance in HER2 status in CTCs and DTCs in early stage breast cancer<br />

by fluorescence in situ hybridization (FISH) using a micr<strong>of</strong>luidic cell<br />

platform (OncoCEE). Methods: Blood (10ml) and BM (1-2ml) from patients<br />

with Stage T1 and T2 breast cancer was collected in OncoCEE-Sure<br />

collection tubes. Mononuclear cells were recovered using a Percoll density<br />

gradient method, incubated with a mixture <strong>of</strong> 10 primary capture antibodies<br />

(Abs), and introduced into streptavidin coated OncoCEE microchannels<br />

for tumor cell capture. For blood samples, captured cells were stained with<br />

anti cytokeratin (CK) and CD45 Abs for CTC enumeration followed by FISH<br />

using probes specific to centromere 17 and HER2. For BM samples,<br />

captured cells were subjected to HER2 FISH analysis. The ratio <strong>of</strong><br />

HER2:CEP17�2.0 in any CK�/ CD45- and CK-/CD45- cell was regarded<br />

as positive for HER2. Results: Blood and BM from 68 patients (68 Blood,<br />

54 BM; 54 matched blood and BM) with stage T1N0 (41), T1N1 (6), T2N0<br />

(11), T2N1(2), T2N2 (1), T2N3 (2) with HER2� (n�7) and HER2-<br />

(n�61) breast cancers were studied. The 7 patients with HER2 � primary<br />

tumor had HER2� DTCs in 3/7 (43 %) and HER2 � CTCs in 1/6 (17 %)<br />

patients. HER2 � DTCs and HER2 � CTCs occurred in 10/47 (21%) and in<br />

4/57 (7%) patients with HER2- primary breast tumors. The discordance <strong>of</strong><br />

HER2 status was observed in 14% in CTCs and in 22% in DTCs.<br />

Conclusion: 1. The cell enrichment and extraction micr<strong>of</strong>luidic platform<br />

(OncoCEE) provides a sensitive approach for evaluation <strong>of</strong> HER2 in CTCs<br />

and DTCs. 2. CTCs and DTCs acquired HER2 gene amplification in 21%<br />

and 7% <strong>of</strong> patients with HER2 negative early stage primary breast cancer.<br />

3. CTCs and DTCs lost HER2 gene amplified status in 57% and 83% <strong>of</strong><br />

patients with HER2 positive early stage primary breast cancer. 4. The<br />

clinical significance <strong>of</strong> alterations in HER2 status among CTCs and DTCs in<br />

early stage breast cancer needs further investigation.<br />

TPS10633^ General Poster Session (Board #54B), Mon, 1:15 PM-5:15 PM<br />

Emerging findings in the Cancer Research UK stratified medicine program.<br />

Presenting Author: Emily Shaw, Cancer Research UK, London, United<br />

Kingdom<br />

Background: Molecular analysis <strong>of</strong> tumours may be used to identify those<br />

predicted to benefit from novel targeted therapies. The Cancer Research<br />

UK programme is piloting plans to apply such testing broadly across the UK<br />

healthcare system, linking molecular phenotype to clinical outcomes.<br />

Methods: The Stratified Medicine Programme (SMP) aims to develop a<br />

model for high quality, large-scale molecular characterization <strong>of</strong> cancer<br />

specimens through an initiative developed in partnership with AstraZeneca,<br />

Pfizer, the UK Department <strong>of</strong> Health and academic researchers. Phase<br />

One <strong>of</strong> the SMP is a two year feasibility study. It aims to demonstrate the<br />

submission <strong>of</strong> consented blood samples and sections <strong>of</strong> surplus diagnostic<br />

formalin-fixed paraffin-embedded tumour tissue from 9,000 patients at<br />

centres across the UK to one <strong>of</strong> three ‘technology hubs’ for mutation testing<br />

<strong>of</strong> genes <strong>of</strong> potential clinical interest (KRAS, BRAF, NRAS, PIK3CA, TP53,<br />

PTEN, TMPRSS2-ERG, EGFR, EML4-ALK and KIT) in six selected tumour<br />

types. The tests are technically validated and will be completed in clinically<br />

relevant timescales. Data including pathological and treatment information<br />

and clinical outcome is also collected for the recruited patients, linked to<br />

the genetic data and stored in a central data repository hosted within the<br />

National Cancer Registration Service. The study opened in September<br />

2011 at 7 sites across the UK and by the end <strong>of</strong> 2011, 760 patientswith<br />

breast, lung, prostate, colorectal, ovarian cancer or metastatic malignant<br />

melanoma had consented to participate. 142 sets <strong>of</strong> molecular results had<br />

been returned to clinical teams. Updated figures will be presented at the<br />

meeting, by which time the programme is projected to have accrued 4000<br />

subjects. By 2013, we hope to have developed a scalable model for routine,<br />

high quality, prospective molecular characterisation <strong>of</strong> tumours for NHS<br />

cancer patients, with consent for the collection, storage and research use <strong>of</strong><br />

population-scale genetic and clinical outcome data. We will report the<br />

emerging results from the Stratified Medicines Programme and early<br />

insights into implications for wider implementation across the UK healthcare<br />

system.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


TPS10634 General Poster Session (Board #54C), Mon, 1:15 PM-5:15 PM<br />

Highly sensitive determination <strong>of</strong> PIK3CA exon 9 and 20 hotspot mutations<br />

in breast tumors. Presenting Author: Alexandre Harle, Centre Alexis<br />

Vautrin, Pathology and Tumor Biology Dept, EA4421 SiGReTO Nancy<br />

University, Vandoeuvre-lès-Nancy, France<br />

Background: Hotspot mutations in exons 9 (E542K and E545K) and 20<br />

(H1047L and H1047R) <strong>of</strong> PIK3CA gene encoding for the p110� catalytic<br />

subunit <strong>of</strong> Phosphatidylinositol 3-kinases (PI3K) are found in approximately<br />

25% <strong>of</strong> breast carcinomas. PIK3CA mutations cause the overactivation<br />

<strong>of</strong> PI3K/AKT/mTOR pathway and lead to resistance to anti-HER2<br />

targeted therapies in breast cancers, are involved in response to anti-mTOR<br />

agents and are proposed as molecular diagnosis marker for PI3K inhibitors.<br />

A highly sensitive method is required to detect PIK3CA mutations in<br />

paraffin-embedded tumor tissues. Methods: We developed a real-time PCR<br />

assay using allele-specific scorpion primers and amplification refractory<br />

mutation system (PCR-ARMS) to detect hotspot mutations in exons 9<br />

(E542K and E545K) and 20 (H1047L and H1047R) <strong>of</strong> PIK3CA <strong>of</strong> PI3K.<br />

Results: PIK3CA mutations were analyzed in 102 paraffin embedded breast<br />

tumors (88 invasive ductal carcinomas and 14 lobular ductal carcinomas).<br />

All specimens were validated by pathologist examination and processed for<br />

DNA extraction and PCR. PIK3CA exon 9 and 20 mutations were found in<br />

22.5% <strong>of</strong> the specimens, 39.1% in exon 9 (26.1% for E542K, 13.0% for<br />

E545K) and 60.9% in exon 20 (17.4% for H1047L and 43.5% for<br />

H1047R). These results were comparable to the literature data (�²�0.327<br />

;p�0.05). PCR ARMS was found to be highly sensitive, able to detect 0.5<br />

% mutated DNA. Conclusion: PCR ARMS assay is highly sensitive for<br />

PIK3CA exon 9 and exon 20 hotspot mutations analysis in breast<br />

carcinomas as molecular marker for anti-HER2 and PI3K inhibitors<br />

response prediction.<br />

Tumor Biology<br />

689s<br />

TPS10635 General Poster Session (Board #54D), Mon, 1:15 PM-5:15 PM<br />

ACRIN 6684 assessment <strong>of</strong> tumor hypoxia in glioblastoma using 18Ffluoromisonidazole<br />

with PET and MRI. Presenting Author: Elizabeth Robins<br />

Gerstner, Massachusetts General Hospital, Boston, MA<br />

Background: Glioblastoma (GBM) is an aggressive type <strong>of</strong> primary malignant<br />

brain tumor and despite treatment with surgery, radiation, and temozolomide<br />

(TMZ) chemotherapy, median overall survival (OS) remains poor. A<br />

pathologic hallmark <strong>of</strong> GBM is tumor necrosis, a suspected result from<br />

endogenous tumor hypoxia. Angiogenesis is stimulated by hypoxia-driven<br />

signaling cascades and is required for tumor proliferation. The inefficient<br />

blood supply <strong>of</strong> these hypoxic tumors also limits the efficacy <strong>of</strong> chemotherapy<br />

and radiotherapy. Surviving hypoxic tumor cells may be selected<br />

out and proliferate as a more aggressive tumor subtype, therefore hindering<br />

OS. 18F-Fluoromisonidazole (FMISO) is a PET radiotracer whose uptake in<br />

hypoxic tissues can be measured radiographically. The degree <strong>of</strong> tumor<br />

hypoxia has been negatively associated with time to tumor progression and<br />

survival (Spence et al, 2008). Knowledge <strong>of</strong> the degree/distribution <strong>of</strong><br />

tumor hypoxia by PET uptake and perfusion MRI parameters may provide<br />

prognostic information and help guide therapy for patients with GBM.<br />

Methods: In this phase II prospective single arm multi-institution study,<br />

patients will undergo baseline FMISO PET and MR imaging two weeks prior<br />

to chemoradiotherapy (CRT). A subset <strong>of</strong> patients will receive a second<br />

FMISO PET one week prior to CRT to assess reproducibility. <strong>Clinical</strong><br />

outcomes <strong>of</strong> OS and 6-month progression-free survival (PFS-6) will be<br />

correlated to PET and MRI parameters. Eligibility: Pathologically confirmed<br />

GBM with residual tumor after surgery (including T2/FLAIR hyperintensity<br />

consistent with tumor) scheduled to receive standard fractionated radiation<br />

therapy and temozolomide alone or with an anti-VEGF agent or PARP<br />

inhibitor. Current enrollment: 22 patients <strong>of</strong> 50 sample size Contact<br />

information: Please contact the PI, Elizabeth R. Gerstner, MD<br />

(egerstner@partners.org) for additional information. Significance: With a<br />

better understanding <strong>of</strong> the extent <strong>of</strong> tumor hypoxia and changes in hypoxia<br />

levels from treatment, more effective therapies could be developed to<br />

inhibit GBM growth, target hypoxic areas and individualize patient care.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.


Publication-Only Abstracts<br />

Publication-only abstracts, which are selected to be published in conjunction with the 2012 <strong>Annual</strong> <strong>Meeting</strong>,<br />

but not to be presented at the <strong>Meeting</strong>, can be found online in full-text, fully searchable versions at<br />

abstract.asco.org and JCO.org.<br />

The publication-only abstracts are not included in the print volume, but are citable to this Journal <strong>of</strong> <strong>Clinical</strong><br />

Oncology supplement. Please refer to the following example when citing publication-only abstracts:<br />

J Clin Oncol 30, 2012 (suppl; abstr e12000)


Author Index<br />

Note: Numerals refer to abstract number. Accepted abstracts not presented at the meeting (designated by �e�) are available in full-text versions on ASCO.org.<br />

A<br />

A’Hern, Roger 2540<br />

Aabakken, Lars e14637<br />

Aaitel, Andrea e15083<br />

Aapro, Matti S. 9133<br />

Aaseboe, Ulf 7001<br />

Aasebø, Ulf 7027<br />

Aaseem, Aarifah e11053<br />

Abacioglu, Ufuk e16015<br />

Abad, Albert 9129<br />

Abad, Leslie W TPS4675^<br />

Abaid, Lisa e15500<br />

Abakar-mahamat, Abakar<br />

5075<br />

Abal, Miguel 10534<br />

Abali, Huseyin 9050<br />

Abalo, Alicia 10534<br />

Abbadessa, Giovanni 4006<br />

Abbas, Ghulam 7074<br />

Abbas, Haider LBA4001,<br />

e13007<br />

Abbas, Kanza S e17018<br />

Abbas, Laura 9589<br />

Abbasi, Fatima 6581<br />

Abbott, David H e15167<br />

Abbotts, Rachel 1014<br />

Abboud, Christelle e15524<br />

Abboud, Miguel e20004<br />

Abboud, Steven F. 6081<br />

Abbruzzese, Bonnie e21066<br />

Abbruzzese, James L. 4051,<br />

4054, 4060, 4116, 4130,<br />

4131<br />

Abda, Naima e18124<br />

Abdalla, Iman A. e16031<br />

Abdel Halim, Inas Ibraheim<br />

e15129<br />

Abdel Karim, Nagla TPS3116<br />

Abdel-Fatah, Tarek M. A.<br />

1013, 1014, 1098<br />

Abdel-Rasoul, Mahmoud 609<br />

Abdel-Wahab, Omar Ibrahim<br />

6606<br />

Abdelhafez, Mohamed F<br />

e19021<br />

Abdelrahman, Fadwa e15024<br />

AbdelRehem, Rania Naguib<br />

4116<br />

Abdelsalam, Mahmoud<br />

e18001<br />

Abdelsalam, Mohamed<br />

e14664<br />

Abdi, Ehtesham A. 9087,<br />

e15152<br />

Abdolmohammadi, Alireza<br />

e16545<br />

Abdool, Adam 5510<br />

Abdous, Belkacem 9138<br />

Abdul Rahman, Rozana<br />

e14705<br />

Abdul Razak, Albiruni Ryan<br />

5587<br />

Abdulkader, Ihab 10534<br />

Abdullaev, Zied TPS7609<br />

Abdullah, Nik Azim Nik 4106<br />

Abe, Hajime e11564<br />

Abe, Hiroaki e11007<br />

Abe, Seiji e13029<br />

ABE, Tatsuya e19508<br />

Abe, Tetsuya 6112<br />

Abedin, Sameem 1085<br />

Abel, Gregory Alan 6012,<br />

6103, e18530<br />

Abenhardt, Wolfgang 3588<br />

Abeni, Chiara e14018<br />

Aberki, Clemence 6594<br />

Abern, Michael 4670<br />

Abernethy, Amy Pickar 2021,<br />

6041, 6047, 6051, 6102,<br />

9128, TPS9153, e15061,<br />

e16556<br />

Abgrall-Barbry, Gaelle 9080<br />

Abhyankar, Dhiraj J. e13548<br />

Abhyankar, Sunil H. 10605<br />

Abid, Arifa 6620<br />

Abidi, Muneer Hyder 6549<br />

Abidoye, Oyewale O. 1526<br />

Aboagye, Eric e21093<br />

Abonour, Rafat 8037,<br />

e18556<br />

Abou-Alfa, Ghassan K. 4057,<br />

9105<br />

Aboukameel, Amro e21057<br />

Aboulafia, David Michael<br />

4030<br />

Abraham, Anasooya A 6016<br />

Abraham, Christine e15131<br />

Abraham, Deepak T e19562<br />

Abraham, Jame 611<br />

Abraham, Joseph 6067<br />

Abraham, Kathryn 7022<br />

Abraira, Victor e21104<br />

Abramovici, Olivia 5565<br />

Abrams, Judith 6549<br />

Abrams, Thomas Adam 3537,<br />

4027, 4112, 4125,<br />

e19586<br />

Abramson, David H. 8549,<br />

8598<br />

Abramson, Jeremy S. 8060,<br />

e18520<br />

Abramson, Vandana Gupta<br />

510<br />

Abrantes, Fernanda L. 9523<br />

Abravaya, Klara e21111<br />

Abreu, Andre Luis de Castro<br />

e15060, e15171, e15212<br />

Abrey, Lauren E. 2006, 2008<br />

Abrial, Catherine 1051<br />

Abruzzese, Elisabetta 6503<br />

Abu Awwad, Raed e12019<br />

Abu Hazeem, Mahmoud<br />

e14641<br />

Abu Hejleh, Taher 6034<br />

Abu sabaa, Amal e15530<br />

Abu Salah, Jehad e14025<br />

Abu-Khalaf, Maysa M. 508,<br />

1045, 10508<br />

Abujamra, Ana Lucia e13546<br />

Abukhdeir, Abde e18147<br />

Abuodeh, Yazan Asad e15024<br />

Acerbi, Irene e11096<br />

Acevedo, Agustin 1111<br />

Acevedo, Andres e12039<br />

Acevedo-Gadea, Carlos<br />

Rodrigo 5529, 5532<br />

Acharya, Sandip TPS6638<br />

Acharya, Shivani Anil 3011<br />

Achatz, Maria Isabel 1522<br />

Achour, Nacredine e14148<br />

Acikalin, Arbil e11088<br />

Acikgoz, Ozgur e14618<br />

Acin, Yolene TPS10102^<br />

Ackerl, Michael 2071<br />

Ackerman, Allison 8569<br />

Ackermann, Elizabeth J.<br />

e13562<br />

Ackland, Stephen P. e14166<br />

Ackloo, Suzanne 10622<br />

Acoba, Jared David 1599<br />

Acosta, Daniel e20513<br />

Acosta, Soraida 2527<br />

Acquaviva, Jaime 3090<br />

Acton, Gary TPS6637<br />

Adab, Kalid e18534<br />

Adachi, Mitsuru e13029<br />

Adai, Alex 3630<br />

Adair, Beth 4526<br />

Adair, Courtney A 7072^<br />

Adam, Barbara e15081<br />

Adam, Clovis 2023<br />

Adam, Kiraely 10051,<br />

e20509<br />

Adam, Paul J. 3092<br />

Adam, Rene 3547<br />

ADAM, Virginie 9080<br />

Adamia, Sophia 6606<br />

Adamo, Barbara TPS656,<br />

e11056<br />

Adamo, Vincenzo 4627,<br />

e11056<br />

Adamoli, Laura 546, 1049,<br />

1115<br />

Adamowicz, Krzyszt<strong>of</strong> e21099<br />

Adams, Bonne J. 3034,<br />

3042^<br />

Adams, Daniel e21079<br />

Adams, Jacqueline e19513<br />

Adams, John 1581<br />

Adams, Judith A. 4021<br />

Adams, Laurel M. e13596<br />

Adams, Ramon 5102<br />

Adams, Richard A. 2522,<br />

3516, 3530<br />

Adams, Rosemary Frances<br />

1066<br />

Adams, Shari TPS3112<br />

Adams, Sylvia 2589, e11064,<br />

e11529<br />

Adams-Campbell, Lucile L.<br />

e14005<br />

Adamson, Douglas 1094<br />

Adamson, Laura M 611<br />

Adamson, Peter C. 9500<br />

Adavikolanu, Kesava<br />

Ramgopal e12501<br />

Addeo, Raffaele e14135<br />

Addioui, Anissa 9563, 9570<br />

Addison, Christina L. 10620<br />

Adel, Nelly G. 4057, 6613<br />

Adelberg, David E. 4569<br />

Adelberg, David 3043<br />

Adelson, Kerin B. e21032<br />

Adelstein, David J. e14611,<br />

e14665<br />

Adelusola, Kayode e11554<br />

Ademuyiwa, Foluso Olabisi<br />

e11563, e19568<br />

Adenis, Antoine 3010, 3502,<br />

3507, LBA4003, 4091,<br />

10007, 10089, 10092,<br />

10095, TPS10102^,<br />

e14560, e14650<br />

Aderka, Dan e14143<br />

Adesunkanmi, Abdulrusheed<br />

Kayode e11554, e12022<br />

Adesunloye, Bamidele 4569,<br />

e13122<br />

Adetchessi, Tarek e19539<br />

Adetiloye, Adebayo e11554<br />

Adhami, Faisal e21064<br />

Aditi, Bhatt e14721<br />

Adjei, Alex A. 3029, 4117,<br />

4545, 7073, 7541, 7555,<br />

7605, 8519, e13006,<br />

e17535<br />

Adjei, Araba 7555, e18118<br />

Adjei, Brenda A. 6086<br />

Adjiri-Awere, Alfred 3086<br />

Adkins, Douglas 5501, 10003<br />

Adolf, Guenther R. 3092<br />

Adra, Thais Rodrigues<br />

e15562<br />

Adsay, Volkan e14170,<br />

e14692<br />

Adua, Daniela e14096<br />

Advani, Anjali S. 6501, 6521<br />

Advani, Ranjana 8060, 8070,<br />

e13079<br />

Adzovic, Sonja 4633<br />

Aebi, Stefan Paul e19648<br />

Aegerter, Philippe 8540<br />

Aerts, Cindy 2507<br />

Aerts, Isabelle 9517, 9519,<br />

9538<br />

Aerts, Joachim 7019<br />

Affronti, Mary Lou 2074^<br />

Afonso Afonso, Francisco J.<br />

e15076, e15077, e18040<br />

Afonso Gomez, Ruth e11081<br />

Afonso, Francisco Javier<br />

e15067<br />

Afonso, Noemia e15532<br />

Afshari, Ashkan 1132<br />

Aft, Rebecca 2525<br />

Aftimos, Philippe TPS652<br />

Agafitei, Raluca Dana 4096<br />

Agajanian, Richy 4550<br />

Agami, Reuven e13515<br />

Agaoglu, Fulya Yaman 9567<br />

Agar, Meera 2604, TPS9153<br />

Agarwal, Amit e16023<br />

Agarwal, Gaurav e16023<br />

Agarwal, Jaiprakash e16021<br />

Agarwal, Neeraj 4506, 4511,<br />

4525, 4536, 4538, 4663,<br />

10503, e13596<br />

Agarwal, Roshan 3022<br />

Agarwal, Vijay e13554<br />

Agarwala, Sandeep 9580<br />

Agarwala, Sanjiv S. e15115<br />

Agarwala, Sanjiv S 8511,<br />

e13088<br />

Agbakwuru, Augustine<br />

e11554, e12022<br />

Agbarya, Abed 9052<br />

Agbo, Felix 3045<br />

Agelaki, S<strong>of</strong>ia 7564<br />

Agelidou, Athina 7564<br />

Agelidou, Maria 7564<br />

Agerholm-Larsen, Birgit 2069<br />

Aggarwal, Ashutosh N e18009<br />

Aggarwal, Charu 7092, 7595<br />

Aggarwal, Rahul Raj 4664<br />

Aggarwal, Shyam e18041<br />

Agha, Mounzer E. 6563<br />

Aghajanian, Carol 5030,<br />

5043, 5054<br />

Aghi, Manish K. 2101<br />

Aglietta, Massimo 530, 4110,<br />

9005, 10009, 10066,<br />

10067, e15073, e15133,<br />

e15192, e20512<br />

Agloria, Maliha e11541<br />

Agnelli, Giancarlo e19612<br />

Agnese, Doreen Marie 1125<br />

692s Numerals refer to abstract number


Agrawal, Kamal e14186<br />

Agrawal, Kireet e14186<br />

Agrawal, Neeraj R. 4574<br />

Agrawal, Nishant e16061<br />

Agrawal, Priya e18523<br />

Agrawal, Rajiv 511<br />

Agrawal, Ritesh e16023<br />

Agrawal, Shruti 2510,<br />

TPS2622<br />

Agre, Pat 9105<br />

Agresta, Samuel V. TPS3111<br />

Aguado Loi, Claudia X 1608<br />

Aguayo, Cristina 1570<br />

Aguayo-Hiraldo, Paibel I.<br />

6568, 6608<br />

Aguggini, Sergio e11546<br />

Aguiar Bujanda, David 7011<br />

Aguiar, Samuel 10069,<br />

e14119<br />

Aguila, Christian e12027<br />

Aguilar Marin, Ivan Carlos<br />

e15036<br />

Aguilar, Alfredo e19636<br />

Aguilar, José Luis 1607<br />

Aguilar-Mahecha, Adriana<br />

TPS1139<br />

Aguin Losada, Santiago<br />

e14733, e19505<br />

Aguirre, Lina 1558<br />

Aguirre, Mercedes 8572<br />

Agulnik, Jason Scott 6058,<br />

e18061, e18098, e19626<br />

Agulnik, Mark 5500, 10003,<br />

TPS10099, TPS10103<br />

Agus, David B. 4656,<br />

TPS4693<br />

Agustoni, Francesco 9133<br />

Aharonov, Ranit T. e14067<br />

Aharonov, Ranit 10528,<br />

e21008<br />

Ahern, Charlotte H 9500,<br />

9540, 9581<br />

Ahluwalia, Manmeet Singh<br />

2014, 2076, 2092, 2093,<br />

2096, 2100<br />

Ahmad, Aamir e21057<br />

Ahmad, Ahmad e15042<br />

Ahmad, Ateeq 3089^<br />

Ahmad, Hilal e16057<br />

Ahmad, I. 3089^<br />

Ahmad, Javed e20508<br />

Ahmad, Saif e14100<br />

Ahmad, Shakil e13039<br />

Ahmad, Syed A e14714<br />

Ahmadi, Hamed 4589,<br />

e15060<br />

Ahmadi, Tahamtan e18529,<br />

e18540<br />

Ahmadzadehfar, Hojjat<br />

e14565<br />

Ahmed, Alaa Abdelhalim<br />

e19568<br />

Ahmed, Asra Z. 8011<br />

Ahmed, Eliyaz e11053<br />

Ahmed, Gul e14111<br />

Ahmed, Mashrafi e11539<br />

Ahmed, Mohamed e15530<br />

Ahmed, Rakib Uddin e11533<br />

Ahmed, Sairah 8040<br />

Ahmed, Samreen 530<br />

Ahmed, Shabina Roohi 3020,<br />

TPS3117<br />

Ahn, Hanjong 4628, e15080<br />

Ahn, Hee Kyung 644, 7104,<br />

e16003, e18148<br />

Ahn, Ji Yeong e14504,<br />

e14652<br />

Numerals refer to abstract number<br />

Ahn, Jin Seok 644, 1003,<br />

2542, 7004, 7104,<br />

e14580, e18148<br />

Ahn, Jin-Hee TPS649, 1003,<br />

2542, 4544^, e11504,<br />

e11508, e12029, e13104,<br />

e13560, e15064, e15080<br />

Ahn, Joong Bae 3606,<br />

e14081, e14084<br />

Ahn, Myung-ju 7004, 7104,<br />

7533, TPS7614, e16003,<br />

e18148<br />

Ahn, Peter H. e19507<br />

Ahn, Sei-Hyun 578, e11504,<br />

e11508<br />

Ahn, Seung Do e18501<br />

Ahn, Soo Kyung 1133,<br />

e21160<br />

Ahn, Yong Chan 7004<br />

Ahn, Yongchel e15080<br />

Ahnen, Dennis J. 3567<br />

Ahrendt, Gretchen M. 1107<br />

Ahrendt, Steven Arthur<br />

e14184<br />

Ahrens, Marit 10032^,<br />

10046<br />

Ährlund-Richter, Lars 4561,<br />

e13518<br />

Ahsan, Habibul e15086<br />

Aiello, Rosa Anna 623<br />

Aigner, Julia 1096<br />

Aihara, Kazuyuki e15201<br />

Aihara, Taichi e21031<br />

Aihara, Tomohiko 1106<br />

Aiken, Christine TPS3643<br />

Aiken, Robert TPS2107<br />

Aikou, Takashi 1106<br />

Ailawadhi, Sikander 6605<br />

Ain, Kenneth B. e16035<br />

Airoldi, Mario 5546, 5548,<br />

e11524<br />

Aisner, Dara L 7504, 7534<br />

Aisner, Joseph TPS3112<br />

Aissat-Daudigny, Louiza 3052<br />

Ait-Oudhia, Sihem e14690<br />

Aitini, Enrico LBA5003<br />

Aizpurua, Miren e21021<br />

Ajaikumar, Basavalinga S.<br />

1093, e11511, e12030,<br />

e14675, e14721, e19619<br />

Ajaikumar, Basavalinga S<br />

e11027, e11043<br />

Ajani, Jaffer A. 4069, 4077,<br />

4078, 4081, 4086,<br />

e14547, e14548<br />

Ajavon, Yves 3547<br />

Ajiki, Tetsuo e14518<br />

Akagi, Yoshito 3558, e14077<br />

Akaike, Makoto 3524<br />

Akakura, Koichiro e15201<br />

Akakura, Nobuaki 3593<br />

Akamatsu, Hiroaki 7088,<br />

e21007<br />

Akamatsu, Hiroki 3607<br />

Akamatsu, Shusuke 10520<br />

Akard, Luke Paul 6513,<br />

6596, 6604, 6605<br />

Akay, Catherine 1102<br />

Akaza, Hideyuki e15004,<br />

e15098, e15106<br />

Akazawa, Kohei 1119,<br />

TPS1141<br />

Akbulut, Hakan 1572, 9050,<br />

e19024<br />

Akcakanat, Argun 1054<br />

Akcali, Zafer e14698<br />

Akdas, Atif 4<br />

Akdeniz, Aydan e11515<br />

Akewanlop, Charuwan e14051<br />

Akgun, Zuleyha e16015<br />

Akhtar, Tanveer 6612<br />

Akhurst, Timothy J. 3563<br />

Akimoto, Tetsuo 5542,<br />

e17554<br />

Akimov, Mikhail 530, 7543<br />

Akin, Sema 10552<br />

Akin, Yildiz 5578<br />

Akinkuolie, Akinbolaju Andrew<br />

e11554<br />

Akintayo, Adebowale O 2576<br />

Akita, Hirotoshi 7571,<br />

e17520, e18135<br />

Akiyama, Hirohiko 10542<br />

Akiyama, Hirotoshi e14047,<br />

e14050, e14624<br />

Akiyama, Nobu 9053<br />

Akiyama, Shoko e19611<br />

Akiyoshi, Kohei e14126<br />

Aklilu, Mebea 2561<br />

Akmaev, Slava 3015<br />

Akmaev, Viacheslav R e14593<br />

Akpek, Gorgun e19565<br />

Akpeng, Belinda 5506<br />

Akram, Madeeha e14658<br />

Akram, Muzaffar 10618<br />

Aksamit, Inga e14118<br />

Aksnes, Anne-Kirsti TPS4694<br />

Aksnessæther, Bjørg Yksnøy<br />

7027<br />

Aksoy, Sercan e11009,<br />

e11011, e11033, e11093,<br />

e13031, e14526<br />

Aktas, Bahriye 552, 556,<br />

624, 1077, TPS1146,<br />

10599<br />

Akula, Komala 1097<br />

Al Ali, Najla 6586, 6588<br />

Al Banna, Amr e18061<br />

AL Basmy, Amany e16002<br />

Al Husaini, Hamed Homoud<br />

e15530<br />

Al mubarak, Mustafa e15530<br />

Al Nasrallah, Nawar 4534<br />

Al Saleh, Khalid e16002<br />

Al Sayyad, Said e18564<br />

al Zoubi, Mazhar 629<br />

Al-Ahmadie, Hikmat 4527<br />

Al-Ali, Haifa Kathrin 6514^<br />

Al-Ali, Haifa-Kathrin<br />

TPS6642^<br />

Al-Ameri, Ali e17004<br />

Al-badawi, Ismail e15530<br />

Al-Batran, Salah-Eddin 4065,<br />

4072, 4080, 4083, 4090,<br />

TPS4138, 10552, e14557,<br />

e21063<br />

Al-Dalee, Abdelmuniem<br />

e19524, e19525, e19526<br />

Al-Dayel, Fouad e20508<br />

Al-Kali, Aref 6614<br />

Al-Matwari, Majid e11046<br />

Al-Mousa, Abdelatif e15024<br />

Al-muderis, Omar 10039<br />

Al-Muhaderis, Omar 10060<br />

Al-Nahhas, Adil e21093<br />

Al-Refaie, Waddah B. 6016<br />

Al-Rifai, Taha e18510<br />

Al-Shamsi, Humaid Obaid<br />

e14001, e17532<br />

Al-Tourah, Abdulwahab J.<br />

8049<br />

Al-Zoubi, Ammar 8011<br />

Alabek, Michelle 1512<br />

Alabiso, Oscar LBA7501<br />

Alalawi, Raed e21105<br />

Alam, Farida e18068<br />

Alam, Salma Moona 10039<br />

Alam, Salma 9589<br />

Alanee, Shaheen e15022<br />

Alarcon Rozas, Ashley 606<br />

Alarcon-Rozas, Ashley E.<br />

2531<br />

Alashwah, Ahmed 5557<br />

Alatise, Olusegun Isaac<br />

e12022<br />

Alattar, Mona Lisa 6558<br />

Alba, E. 4630, 10500,<br />

10616, e14115<br />

Alba, Emilio 530<br />

Albain, Kathy S. CRA6009,<br />

9018<br />

Albanell, Joan 10500, 10616<br />

Albanese, Christopher 3095<br />

Albani, Fiorenzo 2057<br />

Albany, Costantine 4534,<br />

e15213<br />

Albarel, Frédérique 8568<br />

Albelda, Steven 7077, 7092<br />

Alberghini, Marco 10022<br />

Alberola, Vicente 1531<br />

Albers, Peter 4522, 4525,<br />

4530, 4590, 4601,<br />

e15112, e15195<br />

Albert, Daniel 9515<br />

Albertelli, Manuela 1604,<br />

e19038<br />

Alberti, Antonio Maria<br />

e11039, e12510<br />

Alberti, Paola 9090<br />

Albertini, Mark R. e19047<br />

Alberts, David Samuel 5015<br />

Alberts, Steven R. 3514,<br />

3526, 3564, 3617,<br />

e13052<br />

Albertson, Tina Marie 3069<br />

Albiges, Laurence 601, 4540,<br />

4554, 4621, e15049<br />

Albino, Miguel E. 1060<br />

Albino, Vittorio e14130<br />

Albright, Frederick S. 6583<br />

Albus, Kerstin 10526<br />

Alcaide, Julia e14116<br />

Alcantara, Jose 3093<br />

Alcindor, Thierry 10010,<br />

e14020<br />

Alcorn, Sara 9132<br />

Aldabbagh, Kais 10042<br />

Aldape, Kenneth D. 2001,<br />

2002, 2003, 2019<br />

Alderson, Derek TPS4143<br />

Aldrich, Melinda C 7008<br />

Aldridge, Julie 8516, 9022<br />

Aldridge, Lynley 6111<br />

Aldrighetti, Luca 4110<br />

Aleem, Aamer 6612<br />

Aleixo, Sabina B. 606<br />

Alekseev, Boris TPS4692^<br />

Alektiar, Kaled M. 9104<br />

Aleman, Berthe M 8039<br />

Alemar, Barbara 10593,<br />

e13546<br />

Alert, Jose 2515<br />

Alessandria, Sebastian<br />

e15507<br />

Alessandroni, Paolo 4084<br />

Alessi, Alessandra e21118<br />

Alessi, Barbara 9005<br />

Alexander, Brian Michael<br />

2005<br />

Alexander, Gwen e14160<br />

Alexander, H. Richard 8570<br />

Alexander, Joseph e16545<br />

Alexander, Maurice 6042<br />

Alexander, Sarah 9076<br />

Alexandre, Jerome 9011<br />

Alexandrow, Mark e21155<br />

693s


Alexanian, Raymond 8093<br />

Alexeev, Boris Y. 4501<br />

Alfaro, Iván E. 3072<br />

Alfaro, Maricarmen e19636<br />

Alfonsi, Marc e16044,<br />

e16055<br />

Alfonsi, Simona e14086,<br />

e21070<br />

Algazi, Alain Patrick 3102,<br />

8510, 8580, TPS8602,<br />

e19046<br />

Algazy, Kenneth M. e16062<br />

Alger, Beverly 8524<br />

Algret, Nathalie 9538<br />

Alguire, Kathryn B. 4127<br />

Alhan, Nurcan e11023<br />

Ali, Asma 1542, e19545<br />

Ali, Haider e14073<br />

Ali, Haris e15066<br />

Ali, Haythem Y. 5508<br />

Ali, Khaled B. 2081,<br />

TPS4684<br />

Ali, Maria e19527<br />

Ali, Mohammed Faisal<br />

e13053<br />

Ali, Mohammed Javed 9515,<br />

9568<br />

Ali, Mohammed e14132<br />

Ali, Sana e13053<br />

Ali, Sayed Sahanawaz 550<br />

Ali, Shadan e21057<br />

Ali, Shihab e18520<br />

Ali, Suhail M. 501, 607, 614,<br />

7054, 7055, e14520,<br />

e15062, e15121, e15170,<br />

e17560<br />

Aliaga, Luis Alberto e19576<br />

Aliberti, Sandra e15073,<br />

e20512<br />

Alibhai, Shabbir M.H. 6005<br />

Alici, Suleyman 638<br />

Alidzanovic, Lejla e14099<br />

Alikhan, Mir Asif e16513<br />

Alila, Hector 3060<br />

Alimoghaddam, Kamran 6556<br />

Alipour, Sina 3560<br />

Alistar, Angela Tatiana<br />

e21109<br />

Alizadeh, Arash A. e21071<br />

Aljadir, David Naji e13126<br />

Aljitawi, Omar Salah 10605,<br />

e13568<br />

Aljubran, Ali Husain e19524,<br />

e19525, e19526<br />

Aljumaily, Raid e17547,<br />

e17550<br />

Alkaied, Homam e14704,<br />

e18574<br />

Alkhasawneh, Ahmad e14612<br />

Alkhateeb, Ahmed e14520<br />

Alkhazna, Ammar Ali e21085<br />

Alkis, Necati e14526<br />

All-Eriksson, Charlotta 10521<br />

Allam, Emad S. 6079, 6081,<br />

e19593<br />

Allan, Suzanne 3022<br />

Allanore, Laurence 10049<br />

Allard, Aurore TPS5112<br />

Allegra, Carmen Joseph 3505,<br />

3579, 3602, TPS4136<br />

Allegrini, Giacomo 629,<br />

3546, e18074<br />

Allen, Ace e13551<br />

Allen, Andrew R. 4041,<br />

e13028<br />

Allen, Christian e19598<br />

Allen, Deborah 3031, 3087,<br />

3529, 5020<br />

Allen, Ian M. 4658, 6032,<br />

6037<br />

Allen, Jeffrey Warren<br />

TPS1137, 3032, 7005<br />

Allen, Jill N. 4021, 4027,<br />

e14615<br />

Allen, Joanna e14015<br />

Allen, Joshua e11544<br />

Allen, Pamela e14669<br />

Allen, Peter J 3577, 3620<br />

Allen, Richard Read e16541,<br />

e16550<br />

Allendorf, John e14708<br />

Allevi, Giovanni e11546<br />

Allgeier, Anouk 2<br />

Allison, Roger 5518, 5591<br />

Allmer, Cristine 9057<br />

Allo, Ghassan 1036, 5105<br />

Allo, Julio 9023, 9025, 9096<br />

Alloisio, Marco 7061<br />

Allred, Alicia J 3528<br />

Allred, Donald Craig 503<br />

Allred, Jacob B e13501<br />

Allum, William H. TPS4143<br />

Alm, Daniel 10083<br />

Almagro, Elena e11527,<br />

e12015, e17502, e17515<br />

Almanaseer, Imad e12002<br />

Alnajjar, Abdulsalam e14154<br />

Alnajjar, Hussain M 4640,<br />

e15100<br />

Aloia, Thomas A. 4116<br />

Aloisi, Alessia 5093, 5094,<br />

e15547, e15550, e15551,<br />

e15553<br />

Aloj, Luigi e19034<br />

Alonso Bermejo, Miguel<br />

e18040<br />

Alonso Martinez, Milagros<br />

5595<br />

Alonso Soler(1), Sonia<br />

e11543, e19596<br />

Alonso, Lorenzo e14115<br />

Alonso, M Carmen 1120,<br />

10555<br />

Alonso, Maria Carmen<br />

e19576<br />

Alonso, Paloma e11517<br />

Alonso, Teresa e20513<br />

Alonso, Vicente 3553, 3571,<br />

3586, e14041, e14097,<br />

e14671<br />

Alonso-Alconada, Lorena<br />

10534<br />

Alonso-Basanta, Michelle<br />

e12508<br />

Alonso-Espinaco, Virginia<br />

3553<br />

Alonso-Guadalajara, Juan<br />

Domingo e14671<br />

Alonso-Jaudenes Curbera,<br />

Guillermo e21002<br />

Alonso-Nocelo, Marta 10534<br />

Aloshin, Vladimir A. e19012<br />

Alpaugh, R. Katherine 605<br />

Alran, Severine e15524,<br />

e15535<br />

Alrwas, Anas e19009<br />

Alsamarai, Susan 6033<br />

Alsfeld, Leonard C e19008<br />

Alsibai, Khaled e16019<br />

Alsina, Maria 3014, 3062<br />

Alsina, Melissa 8012, 8035<br />

Altavilla, Giuseppe e18039<br />

Altena, Renske 4528,<br />

e15035<br />

Alter, Blanche P 5563<br />

Althaus, Betsy 528<br />

Altieri, Dario C. e13504<br />

Altintas, Sevilay 1129<br />

Altiok, Soner 10084<br />

Altman, Jessica K. 6520<br />

Altmannsberger, Hans-Michael<br />

e14557<br />

Altshuler, David 2081<br />

Altundag, Kadri 1572,<br />

e11009, e11020, e11033,<br />

e11080, e11092, e11093,<br />

e11509<br />

Altundag, Mustafa Kadri<br />

e11011, e11026<br />

Altundag, Ozden e11011,<br />

e11023, e11026, e11045,<br />

e11515, e14174, e15572,<br />

e15573<br />

Altwairgi, Abdullah Khalaf<br />

500, e11005, e19541<br />

Alumkal, Joshi J. 4549,<br />

TPS4697<br />

Aluri, Jagadeesh 3016<br />

Alva, Ajjai Shivaram 4586<br />

Alvarado Miranda, Alberto<br />

1607, e11016, e12027<br />

Alvarado-luna, Gabriela<br />

e11098<br />

Alvarez Bernardi, Julio<br />

e11028<br />

Alvarez Gomez, Elena e18040<br />

Alvarez, Carlos e18044<br />

Alvarez, Delia 3034, 3042^<br />

Álvarez, Elena e18070<br />

Alvarez, Inaki e11074<br />

Alvarez, Isabel 10512,<br />

10595, e11081<br />

Alvarez, Lucia e15521<br />

Alvarez, Martina e14115<br />

Alvarez, Rene J 3019<br />

Alvarez, Ricardo H. 594,<br />

1102, 3010, 9072<br />

Alvarez, Ronald David 5036,<br />

5060, 5062^, e13087<br />

Alvarez, Ruth e11545<br />

Alvarez, Sonsoles e14058<br />

Alvarez-Gallego, Mario 1570<br />

Alvarez-Gallego, Rafael 4040<br />

Alvarez-Reeves, Marty 9007<br />

Alve, Abigail e19048^<br />

Alvear, Maria 5078<br />

Alvelo-Rivera, Miguel e17528<br />

Alverdy, John C. 10090<br />

Alves, Beatriz da Costa Aguiar<br />

e11059<br />

Alves, Marclesson S. e18078<br />

Alves, Marcos Pantarotto<br />

5567<br />

Alvfors, Carina 7539<br />

Alyasiry, Amer 2525<br />

Alyasova, Anna 4501<br />

Amaadour, Lamiae e18124<br />

Amadio, Giulia 5059<br />

Amadio, Placido e15544<br />

Amador, Rosa Mari-a 2527<br />

Amadori, Dino 9005, 10601,<br />

10615, e11054, e17546,<br />

e18045<br />

Amakye, Dereck Dokyi 9519,<br />

9562<br />

Amalfitano, Andrea 2585<br />

Amalraj, Jerrin e19619<br />

Amant, Frederic 5071<br />

Amarantidis, Kiriakos e14567<br />

Amaravadi, Ravi K. 3029,<br />

3102, e13604<br />

Amarti, Affaf e11502<br />

Amato, Robert J. e15124<br />

Amatruda, Thomas 2569<br />

Amatu, Alessio 3570<br />

Amaya De Carrillo, Carla 5096<br />

Amaya, Alex 2555, TPS3110<br />

Ambady, Prakash e11085,<br />

e12509<br />

Ambinder, Richard F. 8003<br />

Ambros, Peter F 9583<br />

Ambros, Tadeu Frantz 595,<br />

e11038<br />

Ambrose, Robert e15138<br />

Ambrosini, Grazia 8598<br />

Ambrosone, Christine B.<br />

e11563<br />

Amdur, Richard 8103<br />

Amela, Eric 10027, e14560<br />

Ameri, Pietro e19038<br />

Ames, Matthew M. 9581,<br />

e13086<br />

Ames, Patricia 4052<br />

Ameye, Lieveke e18006<br />

Amiel, Gilad TPS4678<br />

Amillano, Kepa 1011<br />

Amin, Asim e13088<br />

Amin, Hesham M. 3107^<br />

Amin, Mahul 10584<br />

Amin, Shahla e17009<br />

Amin, Shubarna 6087<br />

Amini, Arya 7043, e14669<br />

Amir, Eitan 558, 1597, 4535,<br />

6043, e13001<br />

Amir, M I 7009<br />

Amiri-Kordestani, Laleh 2548,<br />

e13122<br />

Amler, Lukas C. 2568, 5575,<br />

e13048<br />

Amling, Christopher L 2564<br />

Ammendola, Michele e21113,<br />

e21134<br />

Amodu, Leo 10061<br />

Amonkar, Mayur 604<br />

Amoroso, Domenico e18074<br />

Amoruso, Alessia e14156<br />

Amos, Christopher I. 1520<br />

Ampil, Fred e11541, e16009<br />

Ampollini, Luca e17546<br />

Amrane, Selma e16546<br />

Amrein, Philip C. 6526,<br />

6606, 6617, 6618, 6621,<br />

6629<br />

Amstutz, Platte T 6581,<br />

e21079<br />

Amthauer, Holger TPS4148<br />

Amtmann, Dagmar 9008<br />

An, Hojung 10093, e11504<br />

An, Shejuan 7039, e18090<br />

An, Sungwhan 3606<br />

An, Tongtong 7537, 7545,<br />

e18022, e18035<br />

Anagnostopoulos, Ioannis<br />

8030<br />

Anaissie, Elias J. e18555,<br />

e18576<br />

Anak, Ozlem 3099<br />

Anaka, Matthew 8542<br />

Anan, Keisei 588<br />

Anand, Aseem 4516, 4562<br />

Anand, Banmeet 4568<br />

Anastasopoulou, Eleftheria<br />

625, 2508, e15125<br />

Anastassopoulos, Kathryn P.<br />

e18559<br />

Anaya, Daniel A. 10000<br />

Anchineyan, Pichandi e19619<br />

Anchisi, Sandro 9059,<br />

e11048<br />

Ancukiewicz, Marek 1026,<br />

2009, 2010, 4112<br />

Anderes, Kenna Lynn<br />

e13562, e13575, e13577,<br />

e21058<br />

694s Numerals refer to abstract number


Andergassen, Ulrich 1072,<br />

1600^, 10540<br />

Anders, Carey K. TPS656,<br />

1524<br />

Anders, Robert e14538<br />

Andersen, Mads Hald 2565,<br />

2574<br />

Andersen, Peter e16045<br />

Andersen, Rikke Fredslund<br />

3573<br />

Anderson, Abraham 4005<br />

Anderson, Clay M. 8534<br />

Anderson, Garnet L 1501,<br />

5015<br />

Anderson, James Robert 9535<br />

Anderson, John 4509<br />

Anderson, Kenneth Carl 8016,<br />

8043, 8106, TPS8112,<br />

TPS8113<br />

Anderson, Lee 8592<br />

Anderson, Lynn 9530<br />

Anderson, Mark Daniel 2030<br />

Anderson, Matthew L. 2558<br />

Anderson, Michelle A e14578<br />

Anderson, Peter Meade<br />

10021<br />

Anderson, Richard 1094<br />

Anderson, Roger T. 6052<br />

Anderson, S. Keith 2015<br />

Anderson, Sonya 9134<br />

Anderson, Stewart J. TPS657<br />

Anderson, Tara B. 8011<br />

Anderson, William F. 1603<br />

Andersson, Michael TPS653<br />

Andersson, Roland 4041<br />

Ando, Kiyoshi 6527, 8023,<br />

8029<br />

Ando, Koji e14033, e14601<br />

Ando, Masahiko 6112, 7003,<br />

7000, 10577, e18128<br />

Ando, Masahiro 7565<br />

Ando, Masashi 10519,<br />

e19535<br />

Andrade, Felipe e11513<br />

Andrade, Gabriela Maria 7008<br />

Andre, Charles 2070<br />

Andre, Elisabeth 10031<br />

Andre, Fabrice 515, 543,<br />

TPS648, TPS667, 1091,<br />

TPS1148, 1601<br />

Andre, Maria do Rosário<br />

e13119<br />

Andre, Thierry 2537^,<br />

LBA3500^, CRA3503,<br />

3506, 3535, e14650<br />

Andreas, Karen 7556<br />

Andreas, Stefan 7001<br />

Andreason, Molly 555, 563,<br />

6063<br />

Andreeff, Michael 6576,<br />

6585, TPS6635<br />

Andreetta, Claudia 1044,<br />

e13058, e13070<br />

Andreis, Daniele e11546<br />

Andreis, Federica e14018<br />

Andreoli, Claudio 1500<br />

Andreopoulou, Eleni 515,<br />

e11010<br />

Andreou, Cal 4550<br />

Andres, Bianca 10041^<br />

Andres, Christian 8563<br />

Andres, Raquel 1001, 10052<br />

Andresen, Corina TPS4682,<br />

5591, 8594<br />

Andreu, Monica e11028<br />

Andrews, Barry 8056<br />

Andrews, Bethany Michelle<br />

e16038, e19534<br />

Andrews, James 1608<br />

Numerals refer to abstract number<br />

Andrezalova, Iveta e19020<br />

Andria, Michael L e14028<br />

Andriole, Gerald L. 4646<br />

Andritsos, Leslie A. 6508<br />

Andrulis, Irene L. 1502<br />

Andrulis, Mindaugas 6519<br />

Andrykowski, Michael A.<br />

e16564<br />

Ang, Celina 3577<br />

Ang, Joo Ern 5022<br />

Ang, K. Kian 5530, 5561,<br />

5583, 5589<br />

Ang, Mei-Kim 3098, 5551,<br />

5580, e16024<br />

Ang, Siok Hoon 5551, 5580<br />

Angelico, Mario TPS4151<br />

Angelini, Francesco e18065<br />

Angelov, Lilyana 2018<br />

Angelsen, Anders 4508<br />

Angelucci, Michela 5093,<br />

5094, e15547, e15550,<br />

e15553<br />

Angevin, Eric 2583<br />

Anghileri, Elena 2083<br />

Angioli, Roberto 5093, 5094,<br />

e15547, e15549, e15550,<br />

e15551, e15553<br />

Angiolillo, Anne L. 9543<br />

Angiuoli, Samuel V. 3068<br />

Anglaret, Bruno 8014<br />

Angleitner-Boubenizek, Lukas<br />

5080<br />

Angrilli, Francesco 8006<br />

Anguera, Georgia e15159<br />

Anguille, Sébastien 2506,<br />

e13051<br />

Angulo, Patricia e19618<br />

Anido, Urbano e14733,<br />

e15067, e15076, e15077,<br />

e15079, e19505<br />

Ann, Mi Sun e18519<br />

Anne, Pramila R. 3621,<br />

e11505, e19507<br />

Annecke, Katja 1072, 1081<br />

Annels, Nicola E. 4653,<br />

e15517<br />

Annesi, Diego 8513<br />

Annetta, Alessandro e19002<br />

Annunziata, Christina M.<br />

2545<br />

Annunziata, Mario 6603,<br />

9120<br />

Anract, Philippe 10056<br />

Ansa, Benjamin 5570<br />

Ansari, Aziz e19588<br />

Ansari, Daniel 4041<br />

Ansari, Rafat H. 7555<br />

Anschuetz, Julia e15566<br />

Ansell, Peter J e13583<br />

Ansell, Stephen Maxted 8043,<br />

8053, 8060<br />

Anselmetti, Giovanni Carlo<br />

e20512<br />

Anshushaug, Malin 9040<br />

Anson-Cartwright, Lynn 4535<br />

Antelo, Maria L e21086<br />

Anthoney, David Alan 2552<br />

Anthony, Lowell Brian<br />

e14610<br />

Anthony, Stephen Patrick<br />

TPS1143, TPS3111<br />

Antic, Vladan TPS7615<br />

Antoine, Eric-Charles e11019<br />

Antoine, Martine e18075,<br />

e18089<br />

Antolino, Giusy e17006,<br />

e18563, e18566<br />

Antón Aparicio, Luis M.<br />

e15076, e15077, e15079,<br />

e15112<br />

Anton, Antonio 1001, 10512,<br />

10595<br />

Antón-Aparicio, Luis e14595,<br />

e21002<br />

Antonarakis, Emmanuel S.<br />

4559, 4654, TPS4697,<br />

e15188<br />

Antonelli, Giovanna e18002<br />

Antonescu, Cristina R.<br />

10001, 10019<br />

Antonescu, Cristina 10002,<br />

10003<br />

Antoni, Giorgia 9006, e19634<br />

Antonia, Scott 2501,<br />

CRA2509, 2559, TPS2620,<br />

7065, 7509, e17559<br />

Antonio, Anna Liza M 6105<br />

Antonio, Heriton MR 1075<br />

Antoniotti, Carlotta 3518,<br />

3540, 3546, 3549, 3585<br />

Antoniou, Antonis C. 1545<br />

Antoun, Sami 4621, 5569,<br />

9026^, e13118<br />

Antsaklis, Aris 5033<br />

Antunes, Ana e18111<br />

Antunes, Luis Carlos Moreira<br />

e14597<br />

Antunez de Mayolo, Jorge<br />

7546<br />

Anwar, Muhammad Suhail<br />

e15534<br />

Anyang, Bean N. 1070<br />

Anyang, Bean 7022<br />

Aoe, Keisuke 7042<br />

Aoki, Daisuke 5003<br />

Aoki, Yoichi 5086<br />

Aparicio Gallego, Guadalupe<br />

e15076, e15077, e15079<br />

Aparicio, Ana 4533, e15092<br />

Aparicio, Jorge 3586, 3618,<br />

10082, 10547, e14097<br />

Aparicio, Luis Anton e14671,<br />

e15067, e15149<br />

Aparicio, Thomas 4018, 4091<br />

Aparisi, Francisco e17545,<br />

e18049, e18053<br />

Aparo, Santiago e14019,<br />

e14682<br />

Apelian, David TPS3638,<br />

e14501<br />

Apisarnthanarax, Smith 7098,<br />

e18032<br />

Aplenc, Richard 1504<br />

Apolikhin, Oleg TPS4692^<br />

Apollinari, Serena e13098<br />

Apollo, Arlyn J. 4057<br />

Apolo, Andrea Borghese<br />

3043, 4569, 4581^, 4592,<br />

10589<br />

Aponte, Luis 10074<br />

Apostolikas, Nikiforos e11519<br />

Appel, Burton 9524<br />

Appelbaum, Frederick R.<br />

6502<br />

Apperley, Jane 6503, 10550<br />

Appignanesi, Remo 9098<br />

Appleman, Leonard Joseph<br />

TPS1138, 2553, 3019,<br />

3037, 3049, 3054,<br />

e13576<br />

Aprile, Giuseppe 3518, 3546,<br />

3549, 3555, 3612,<br />

TPS3637, e14040<br />

Aqua, Keith A. e15546<br />

Aquilina, Michele e11054<br />

Aquino, Luciana Carla Martins<br />

de e14027<br />

Aragaki, Aaron K 1501<br />

Aragon-Franco, Raul e15584<br />

Arai, Daisuke e18058<br />

Arai, Roberto J. e11513<br />

Arai, Yasuyuki e15065,<br />

e15069<br />

Araki, Nobuhito 10009,<br />

10067<br />

Araki, Toshimitsu e21053<br />

Arance, Ana M 8517<br />

Arance, Ana Maria e19023<br />

Aranda, Enrique 3571<br />

Aranda, Sanchia 9124<br />

Arantes, Heloisa 606<br />

Arastu, Nabeel H. 6093<br />

Araten, David J. e21082<br />

Araujo, Bertha Catherine B.<br />

e16046<br />

Araujo, Dejka M. 10071,<br />

e19520<br />

Araujo, John C. e15151<br />

Arauz, Gabrielle 4548,<br />

TPS4697<br />

Aravantinos, Gerasimos 5033<br />

Araya, Tomoyuki e17540<br />

Arbani, Tammy TPS661<br />

Arbaud, Alexandre TPS9154<br />

Arber, Daniel 6072<br />

Arber, Nadir 1507, 1564<br />

Arcaini, Luca 8006<br />

Arcamone, Manuela 8066<br />

Arcasoy, Murat O. 6624<br />

Arce, Claudia e11016<br />

Archila, Pilar e18114<br />

Arcila, Giovanna e19604<br />

Arcila, Maria E. 7532, 7578,<br />

10560<br />

Arcos, Rebecca 3001<br />

Arcury, Justin T. e14724<br />

Arcusa Lanza, Angels 10500,<br />

10616<br />

Ardavanis, Alexandros 625,<br />

2508, 10606, e11073,<br />

e21098<br />

Ardenkjaer-Larsen, Jan Henrik<br />

4660<br />

Ardizzone, Antonino e11001<br />

Ardizzoni, Andrea e18021,<br />

e18096<br />

Arellano, Martha Lucia 6520<br />

Arena, Francis P. 559,<br />

TPS4134<br />

Areses Manrique, Carmen<br />

e18040, e18146<br />

Aresti, Unai e17522, e21009<br />

Argiles, Guillem 3005, 3014<br />

Argiris, Athanassios 2610,<br />

3049, 3073, 5541<br />

Arguello, David 1040, 10630<br />

Argyriou, Andreas 9090<br />

Arias, Fernando e14057<br />

Arias-Pulido, Hugo 5549<br />

Arican, Ali e15104<br />

Arifi, Samia e11502,<br />

e11516, e14725, e14726,<br />

e18124<br />

Ariga, Takuro e15533<br />

Arik, Zafer e11009, e11033,<br />

e11093, e13031<br />

Arima, Yoshimi 635<br />

Arin-Silasi, Dan 5099,<br />

e15581<br />

Aristarco, Valentina 519<br />

Arita, Shuji 3045<br />

Ariyan, Charlotte Eielson<br />

8583<br />

695s


Arkenau, Hendrik-Tobias<br />

2540, 3000, 3041, 3097,<br />

e13585<br />

Arkhiri, Peter e20501<br />

Arlen, Aimee Ginsburg<br />

e16536<br />

Arlen, Philip M. 2517, 3043,<br />

4569<br />

Armaghany, Tannaz e14667,<br />

e16009<br />

Armakolas, Athanasios 583<br />

Armand, Corine e15109<br />

Armendariz, Pedro e14057<br />

Armengol Alonso, Alejandra<br />

e15567<br />

Armenian, Saro 9505<br />

Armitage, James O. 8047<br />

Armstrong, Andrew J. 4515,<br />

4519^, 4550, 10533<br />

Armstrong, Anne 530<br />

Armstrong, Dawn Elizabeth<br />

e14073<br />

Armstrong, Deborah Kay<br />

5062^, 9103<br />

Armstrong, Elizabeth<br />

TPS4134<br />

Armstrong, Graham 4121<br />

Armstrong, Gregory T. 6030,<br />

9505, 9514, 9528, 9531<br />

Armstrong, Joanne 1608<br />

Arnal, Francisco e21002<br />

Arnaldez, Fernanda Irene<br />

9511<br />

Arndt, Diane 3013<br />

Arnedos, Monica 1091, 1601<br />

Arneson, Thomas J. 4659,<br />

e15169<br />

Arnold, Andrew Michael 7511<br />

Arnold, Angela G. 5043<br />

Arnold, Dirk CRA3503, 4090,<br />

4095^, e14162<br />

Arnold, Julie 3567<br />

Arnold, Susanne M. e16035,<br />

e21000<br />

Aroch, Ilan 1564<br />

Aron, Monish e15060,<br />

e15171<br />

Aronow, Mary E 8071<br />

Arons, Evgeny 2503^<br />

Aronson, Melyssa 5026<br />

Arora, Amit 3545<br />

Arora, Madan L. 3619,<br />

e14045, e15554<br />

Arora, Neeraj K 9130<br />

Arora, Rajender Singh<br />

TPS649<br />

Arora, Reety 8577<br />

Arora, Sameer e14694<br />

Arowolo, Olukayode Adeolu<br />

e11554<br />

Arpaci, Erkan e14526<br />

Arpaci, Fikret 6550<br />

Arpino, Grazia TPS654^,<br />

1554<br />

Arqueros, Cristina e15159<br />

Arra, Claudio e11052,<br />

e13539<br />

Arrabal, Rafael 7011<br />

Arranz Arija, Jose Angel<br />

TPS4672, TPS4674<br />

Arranz, Jose Angel TPS4685,<br />

e15041<br />

Arranz, Juan Luis 5520,<br />

e15094, e18069, e18106,<br />

e21015<br />

Arriaga, Yull Edwin e15066<br />

Arrichiello, Cecilia e14130<br />

Arrieta, Oscar 1607, 7068,<br />

e18028, e18114, e19594,<br />

e19618<br />

Arrighi, Giada 629<br />

Arrigo, Carmela e18039<br />

Arrigoni, Pierre Paul 2082<br />

Arrington, Amanda Kathleen<br />

1117<br />

Arriola, Edurne 7554<br />

Arrossi, A. Valeria e18085<br />

Arrowsmith, Edward 8556<br />

Arroyo, Gerardo F. e14521<br />

Arsenault, Daniel M. e16518<br />

Arsenault, Youri e13027<br />

Arslan, Bulent e14610<br />

Arslan, Cagatay e11011,<br />

e11020<br />

Arslan, Deniz e14070<br />

Arsov, Christian e15195<br />

Artac, Mehmet e11008<br />

Artal, Angel e12000,<br />

e12012, e18011<br />

Arteaga, Carlos L. 510, 605,<br />

1024, 1054<br />

Arteta-Bulos, Rafael e21077<br />

Arthur, Chris 6559, 6627<br />

Arthur, Diane Carol e18568<br />

Arthur, Douglas W. TPS657<br />

Artigiani-Neto, Ricardo<br />

e14657<br />

Artignan, Xavier e15131<br />

Artioli, Fabrizio e11071<br />

Artioli, Grazia e12037<br />

Artru, Pascal 4018, e14129<br />

Artz, Andrew S. 6535^,<br />

6562, e13120^<br />

Arun, Banu 1518, 1546,<br />

1549, e11090<br />

Arya, Monica 6561<br />

Arya, Nikita 8518<br />

AS, Kirthi Koushik e11536<br />

Asaad, Razan e14704<br />

Asad, Sultan 6612<br />

Asada, Yukinori e16020<br />

Asahina, Hajime 7086,<br />

e18135<br />

Asami, Kazuhiro 7521,<br />

10577<br />

Asano, Michio TPS10632<br />

Asanuma, Hiroshi 9569<br />

Asanuma, Taku e14011<br />

Asatiani, Ekatherine TPS3118<br />

Asch, Adam Steven e17008<br />

Asch, Steven M 6105<br />

Asche, Carl V e14106,<br />

e14141, e14146, e15183,<br />

e18097<br />

Asche, Julia 4115<br />

Aschebrook-Kilfoy, Briseis<br />

8072<br />

Aschele, Carlo e17533<br />

Ascherman, Jeffrey 6118<br />

Ascierto, Maria Libera 8576,<br />

10565<br />

Ascierto, Paolo Antonio 8501,<br />

8502^, 8511, 8513, 8517,<br />

8518, 8529, 8539, 8573,<br />

8581, 10565, e19034<br />

Asero, Salvatore e15544<br />

Ashcr<strong>of</strong>t, John 8015<br />

Ashfaq, Raheela 1040,<br />

10630, e13571<br />

Ashkenazi, Avi 3543, 3552<br />

Ashkenazi, Karin 10528,<br />

10575, e21008<br />

Ashley, David M 9519<br />

Ashley, Pandora e11069<br />

Ashley, Sue 10086<br />

Ashman, Jonathon 6108<br />

Ashraf, Noman e14715<br />

Ashton-Prolla, Patricia 1522,<br />

10593, e13546, e21042<br />

Ashur-Fabian, Osnat e19573<br />

Ashworth, Alan 3050, 5035<br />

Asin, Gemma e14057<br />

Askenazi, Manor 6136<br />

Askin, Dudu e11008<br />

Asmis, Timothy R. e14154<br />

Asnacios, Amani 4018<br />

Asrari, Fariba 1128<br />

Assadourian, Sylvie 3579<br />

Assaker, Richard 2070<br />

Asselain, Bernard 8546<br />

Asselin, Barbara 9504<br />

Assenat, Eric 3633, 4032<br />

Assikis, Vasily J. 4550<br />

Assis, Joana e15532<br />

Assmann, Gunter 1523<br />

Assmus, Joerg 4015<br />

Assouline, Sarit E. 2547,<br />

8064<br />

Astl, Dorothea e13040<br />

Astolfi, Annalisa 10087<br />

Astone, Antonio e14156<br />

Astudillo, Horacio e11565,<br />

e15584<br />

Astudillo, Julio 7011<br />

Ata, Alper e15104<br />

Ataee, Mari 6556<br />

Ataergin, Selmin 6550<br />

Atagi, Shinji 7003, 7017,<br />

10577, e19556<br />

Atallah, Ehab L. 6582<br />

Ataman, Ozlem e14015<br />

Atanackovic, Djordje 2573^,<br />

4095^, 7013<br />

Atasever Akkas, Ebru e16005,<br />

e19575<br />

Atasoy, Beste Melek e16015<br />

Atassi, Bassel e16019<br />

Atefi, Mohammad e13045<br />

Atencio, Liezl 1592<br />

Ates, Can e20000<br />

Athanatou, Evangelia e19610<br />

Atherton, Pamela J. 6108,<br />

6133, e16572<br />

Atias, Dikla e14646<br />

Atif, Hesham 5557<br />

Atilgan, Alev e11023<br />

Atkins, Barbara McCormick<br />

7531<br />

Atkins, James Norman<br />

LBA506, TPS657,<br />

LBA1000, 3545<br />

Atkins, James LBA4007<br />

Atkins, Michael B. CRA2509,<br />

4500, 4505, 4543, 4606,<br />

4635, 8504, 8516, 8527,<br />

8569<br />

Atkinson, Bradley J. 4615<br />

Atkinson, Thomas Michael<br />

9144, e19633<br />

Atluri, Prashanti e13109<br />

Atmaca, Akin e14557<br />

Atmadini, Maizaton e15091<br />

Atreya, Chloe Evelyn 3517<br />

Attal, Michel 8009<br />

Attar, Eyal C. 6526, 6606,<br />

6617, 6618, 6621, 6629<br />

Attard, Gerhardt 4553,<br />

e15134, e15136<br />

Attassi, Mared e14565<br />

Attiyeh, Edward F. 9533<br />

Attner, Per e16048<br />

Attwood, Kristopher e11563,<br />

e15042, e15204, e19568<br />

Atwal, Siminder e21122<br />

Atwan, Manal e14117<br />

Atwood, Todd 10629<br />

Atzema, Clare 6039<br />

Au, Joseph Siu-kie 1534<br />

Au-Yeung, George e15541<br />

Aubry, Regis e19623<br />

Auby, Dominique 3506<br />

Auchus, Richard J 4645<br />

Aucoin, Nathalie LBA3500^<br />

Audet, Robert e21099<br />

Audhuy, Bruno 6633<br />

Audigier-Valette, Clarisse<br />

TPS7112<br />

Audretsch, Werner e11556<br />

Auerbach, Martin 10012<br />

Auerbach, Michael e19609<br />

Auger, Nathalie 10556<br />

Augustin, Doris 552, 1072,<br />

1081<br />

Augustine, Christina K.<br />

10563<br />

Augustine, Titto A e14019<br />

Auliac, Jean Baptiste e16560<br />

Auliac, Jean-Bernard 1574<br />

Ault, Patricia e19589<br />

Aumont, Maud 2082<br />

Aung, Fleur M 6622<br />

Aung, LeLe e20003<br />

Aung, Thinzar 3589, e14155<br />

Aura, Claudia 509, 566,<br />

1011, 3014, 7041, 10511<br />

Aurer, Igor 8002<br />

Auriemma, Alessandra<br />

e14661<br />

Aurilio, Gaetano 546, 1049,<br />

1115<br />

Aussilhou, Beatrice 3609<br />

Aust, Daniela Ellen 4053<br />

Austin, Frances e14184<br />

Austin, J. Max e13087<br />

Austin, Melissa 5515<br />

Austin, TaShara e14617<br />

Autier, Philippe 1509<br />

Autio, Karen A. 4517<br />

Autran, Didier 2060<br />

Auziere, Sebastien e15198<br />

Avallone, Antonio e14040<br />

Avancha, Kiran Kumar<br />

Venkata Raja e15107,<br />

e16525<br />

Avella, Antoni e11036<br />

Avenia, Bettine TPS6638<br />

Averbook, Bruce Jeffrey 8522<br />

Avery, William 4056<br />

Avet-Loiseau, Herve 8014<br />

Avgerinos, Costas e14625<br />

Aviles-Salas, Alejandro<br />

e18028<br />

Avino, Robert J. 6079<br />

Avisar, Noa 9125<br />

Avivi, Camila e14646<br />

Avril, Marie-Françoise 8578,<br />

e21014, e21046<br />

Avritscher, Rony 10559,<br />

e14664<br />

Avsar, Emin 7543<br />

Avuzzi, Barbara e21118<br />

Awad, Ola 10025<br />

Awada, Ahmad 535, TPS652,<br />

TPS1140^, 3018^,<br />

e13594, e13605<br />

Awais, Omar 7074<br />

Awan, Farrukh Tauseef 6573,<br />

8108<br />

Awan, Tashfin 6612<br />

Awane, Hiroaki e14518<br />

Awasthi, Sanjay e13557<br />

Axelrod, Rita 7512, TPS7619<br />

Axelson, Magnus 7539<br />

696s Numerals refer to abstract number


Axiotis, Constantine A<br />

e18504<br />

Ayala, Francisco 4587,<br />

e11024<br />

Ayala-Ramirez, Montserrat<br />

4641<br />

Ayan, Inci 9567<br />

Ayanian, John 6006<br />

Aydin, Kubra e14698<br />

Aydogan, Fatma e14628<br />

Ayello, Janet e13022,<br />

e21150<br />

Ayers, Mark 3531, 3587,<br />

4054<br />

Ayerza, MIguel Angel 10074<br />

Ayhan, Ali e15572, e15573<br />

Aykan, Faruk e14628<br />

Ayuso Sacido, Angel 3601,<br />

e14093, e14144<br />

Ayuso, Juan Ramón 3536,<br />

e14097<br />

Ayyildiz, Veysel e11509<br />

Azad, Abul Kalam 1535,<br />

6005, 7586, 10522<br />

Azad, Nil<strong>of</strong>er Saba 2517,<br />

2545, 3009, 3016, 3020,<br />

TPS3117, 3616<br />

Azada, Michele e18140<br />

Azam, Faisal e11053<br />

Azar, Catherine A. LBA506,<br />

LBA1000<br />

Azar, Riad e14584<br />

Azaro, Analia 2042, 3005,<br />

3014<br />

Azcona, Eider e18031<br />

Azer, Mary W. F. 8558<br />

Azevedo, Carolina e14183<br />

Azevedo, Isabel 5567<br />

Azevedo, Renata Guise Soares<br />

e19552<br />

Azevedo, Sergio Jobim<br />

TPS4135<br />

Azim, Hamdy A. e18570<br />

Azinovic, Ignacio e16000<br />

Aziz, Noreen 6006, 6008,<br />

9130<br />

Aziza, Richard 4618<br />

Azizi, Amedeo A. TPS9596<br />

Azizi, Michel 1095, 5067<br />

Azkona, Eider e11525,<br />

e21009<br />

Aznab, Mozaffar 6556<br />

Aznar, Marianne Camille<br />

e18527<br />

Azodi, Masoud 5099,<br />

e13125, e15526, e15581<br />

Azuma, Haruhito e15000,<br />

e15048<br />

Azuma, Koichi e18060<br />

Azuma, Mizutomo 4026,<br />

4088<br />

Azzarello, Giuseppe 5513<br />

Azzoli, Christopher G. 7010,<br />

7014, 7052, 7066, 7527<br />

Alvarez Gallego, Jose Valero<br />

e11081<br />

B<br />

Baar, Joseph TPS655<br />

Baas, Paul 7081<br />

Baba, Hideo 3558, 10523,<br />

e13553, e21084<br />

Baba, Tomohisa e19583<br />

Baba, Yoshifumi 10523,<br />

e13553<br />

Babacan, Nalan Akgul<br />

e16005, e19575<br />

Babacan, Taner e11026,<br />

e11080<br />

Numerals refer to abstract number<br />

Babakoohi, Shahab 7087<br />

Babal, Pavel e15027<br />

Babb, Sheri 1520<br />

Babich, John W. e13592<br />

Babiera, Gildy 594, 1028,<br />

1102<br />

Babigumira, Joseph e14525<br />

Babirak, Loren 6617, 6618<br />

Babu, Thambidurai Ganesh<br />

e13002<br />

Bacanu, Florin 9046<br />

Baccarani, Michele 6503,<br />

6504, 6513, 6531, 6596,<br />

6604<br />

Baccelli, Irene 1090<br />

Bach, Bruce A. 5504^<br />

Bach, Ferdinand 2033, 2515<br />

Bach, Peter 6101<br />

Bacha, Jeffrey A. e13123<br />

Bachelot, Thomas Denis<br />

TPS646<br />

Bachelot, Thomas 10562<br />

Bachet, Jean Baptiste 10078<br />

Bachet, Jean-Baptiste 2537^,<br />

e14059<br />

Bachir, Jeanette TPS9596<br />

Bachleitner-H<strong>of</strong>mann, Thomas<br />

3510, TPS10632<br />

Baciewicz, Frank e17528<br />

Back, Anthony 9008, 9101,<br />

9139<br />

Back, Michael e12507<br />

Back, Ronald R e14505<br />

Backen, Alison e14015<br />

Bacon, Ludwing e12027<br />

Bacus, Sarah S. e21120<br />

Badalamenti, Giuseppe<br />

LBA10008, 10063<br />

Badarinath, Suprith<br />

TPS3635^<br />

Badger, Heath D. TPS1136<br />

Badgwell, Brian D. e19563<br />

Bading, James R 639<br />

Badoual, Cecile 5554,<br />

e13578<br />

Badoux, Xavier C. 6516^<br />

Badovinac Crnjevic, Tanja<br />

e21126<br />

Badreldin, Waleed 4529<br />

Badri, Nadia 10047<br />

Badros, Ashraf Z. 8086,<br />

e19565<br />

Badurak, Pawel e12011<br />

Badve, Sunil S. 505, 1005,<br />

1021, 7032, 7106, 7107,<br />

7108, 10628<br />

Badwe, Rajendra A. 1108,<br />

e11552<br />

Bae, Jeong Mo 3624<br />

Bae, Kyun Seop 3592<br />

Bae, Kyun-Seop e13104,<br />

e13560<br />

Bae, Sang Byung e19554<br />

Bae, Sang-Byung e14688<br />

Bae, So Hyun 2041<br />

Bae, Sung Hwa e14570<br />

Bae, Susie e16516<br />

Bae, Woo Kyun e14649<br />

Bae-Jump, Victoria Lin 2591,<br />

5009, 5050, 5092,<br />

e15511<br />

Bae-Jump, Victoria e13063<br />

Baehner, Frederick L. 1005,<br />

1021<br />

Baek, HeeJong e17523<br />

Baek, Jeong-Heum e14137<br />

Baek, Ji Yeon e14072<br />

Baek, Jin Ho 6536, 6538,<br />

e18533<br />

Baek, Sun Kyung e14128<br />

Baena, Jose Manuel 10608,<br />

e11535, e21088<br />

Baer, Maria R. 6611,<br />

e16519, e16537<br />

Baerlocher, Gabriela M 6510<br />

Baertschi, Daniela 3595<br />

Baez-Diaz, Luis LBA506<br />

Baeza, Mario R. e14002<br />

Baffert, Sandrine TPS667<br />

Bagal, Bhausaheb Pandurang<br />

9559, e17003<br />

Bagalà, Cinzia e14156<br />

Bagatell, Rochelle 9500,<br />

9540<br />

Bagga, Dilprit 595<br />

Baggerly, Keith 1047<br />

Baghmar, Saphalta e18573<br />

Baglin, Anne Catherine<br />

e16013<br />

Baglioni, Michele 3061<br />

Bagnardi, Vincenzo 546,<br />

1049, 1115<br />

Bago-Horvath, Zsuzsanna<br />

10570, e21033<br />

Bague, Silvia 10052<br />

Bahary, Nathan 3545, 4022<br />

Bahassi, El Mustapha e16017<br />

Bahl, Ankur 5540<br />

Bahleda, Rastilav 3080, 4614<br />

Bahleda, Rastislav 3000,<br />

3012, 3104<br />

Bahlis, Nizar J. 6548, 8035<br />

Bahn, Duke e15212<br />

Bahrami, Armita e13544<br />

Bai, Hua 7537, 7545,<br />

e18022, e18035<br />

Bai, Li-Yuan e17014<br />

Bai, Shuang 2567, e13000<br />

Bai, Wei e14009<br />

Bai, Yuxian 4008<br />

Baichwal, Vijay R. e13581<br />

Baig, Hana e19015<br />

Baiget, Montserrat e14104,<br />

e18057<br />

Bailador, Gonzalo 1570<br />

Baile, Walter F. 9049<br />

Bailey, Clyde 8041<br />

Bailey, Howard Harry 520,<br />

9039<br />

Bailey, Leighann 2090^,<br />

2094^<br />

Bailey, Stuart 7543<br />

Bainbridge, Daryl 6035<br />

Bains, Manjinder e15189<br />

Bains, Manjit S. 4061<br />

Bains, Puneet 6572<br />

Bains, Sarina e19641<br />

Baiocchi, Robert A. e18508<br />

Bair, A H. 4503<br />

Baird, Kristin 9545<br />

Baird, Richard D 3100<br />

Bajetta, Emilio LBA4001<br />

Bajor, David e18529<br />

Bajorin, Dean F. 4527,<br />

4581^, 4592, TPS4673,<br />

e15022<br />

Bajpai, Jyoti e13068<br />

Bajrami, Ilirjana 5035<br />

Bak, Sharon 6097<br />

Baka, Margarita e20002<br />

Bakacs, Tibor e13102<br />

Bakalarski, Pamela 4098<br />

Bakalian, Silvin e13017<br />

Baker Neblett, Katherine<br />

e15085<br />

Baker, Dave 9533<br />

Baker, Jackie S. 4077<br />

Baker, Kevin Scott e16502<br />

Baker, Marshall e14642<br />

Baker, Sharyn D. 2549<br />

Bakhru, Arvind 5055<br />

Bakhshi, Sameer 9549,<br />

9551, 9580<br />

Bakkar, Rania 10510<br />

Bakkum-Gamez, Jamie<br />

Nadine 5004<br />

Bal, Wieslaw e14159,<br />

e21049<br />

Balaa, Fady e14154<br />

Balabanov, Stefan 4590<br />

Balakrishnar, Bavanthi 550<br />

Balana, Carmen 2045,<br />

10079, e12504<br />

Balar, Arjun Vasant 4581^<br />

Balasa, Balaji 8087<br />

Balasubramaniam, Sanjeeve<br />

2548, 2553<br />

Balasubramanian, Sohail<br />

3533<br />

Balasubramaniyan,<br />

Veerakumar 2002<br />

Balboni, Michael J. 9116,<br />

9132<br />

Balboni, Tracy A. 5579,<br />

9116, 9132<br />

Balcaitis, Stephanie 2585<br />

Balch, Glen C. e14165<br />

Balchin, Katelyn 6083,<br />

e19527<br />

Balci, Derya e18552<br />

Balci, Pinar e11507<br />

Balcueva, Ernie P. 9001<br />

Baldassarre, Randall L 7043<br />

Baldassarri, Paolo e14096<br />

Baldazzi, Valentina e15185<br />

Baldelli, Anna Maria 4084<br />

Baldelli, Elisa e18101,<br />

e21094, e21096<br />

Baldeos, Raquel e19636<br />

Balderi, Segio e18096<br />

Baldi, Giacomo G. 10063<br />

Baldi, Giacomo 10053,<br />

10065<br />

Baldini, Capucine e11070<br />

Baldini, Federica e19035,<br />

e19036<br />

Baldino, Giovanni e14094<br />

Baldotto, Clarissa Serodio Da<br />

Rocha 7506<br />

Baldotto, Clarissa Serodio<br />

e17548<br />

Baldueva, Irina Alexandrovma<br />

e13059<br />

Baldus, Stephan e11556,<br />

e14527<br />

Baldwin, Gouri e13507<br />

Baldwin, Graham e15117<br />

Bale, Allen E 1540<br />

Balen, Enrique e14057<br />

Balint, Joseph 2585<br />

Balis, Frank M. 9500, 9576<br />

Balk, Steven P. 4520, 4521<br />

Balko, Justin M. 510, 1024<br />

Ball, David 3590, e16538<br />

Ball, Douglas Wilmot 3020,<br />

TPS3117, 5508, 5518,<br />

5591<br />

Ball, Graham 1098<br />

Ball, S e13522<br />

Ballal, Shaila TPS3634<br />

Ballard, David 1046<br />

Ballatore, Valentina e15073,<br />

e15133<br />

Ballatore, Zelmira e21070<br />

Balleine, Rosemary L 550<br />

Balleisen, Leopold 6610<br />

697s


Ballen, Karen K. 6606, 6617,<br />

6618, 6621, 6629<br />

Ballester Navarro, Inmaculada<br />

4587<br />

Balleyguier, Corinne 5028,<br />

10098<br />

Ballman, Karla V. 4565,<br />

6015, 7008<br />

Ballo, Harald 3<br />

Ballot, Josephine 615<br />

Bally, Olivia 8068<br />

Balmaña, Judith 509, 566,<br />

619, 1009, 1011<br />

Balogh, Agnes 8508<br />

Balsells, Josep e21035<br />

Balser, Christina 3<br />

Balusu, Ramesh 10549<br />

Balvers, Melan 3059<br />

Balzarini, Luca 2580<br />

Bamberg, Michael e16033<br />

Bambury, Ian Garfield 5032<br />

Bambury, Richard Martin<br />

2062, e11569, e15078,<br />

e16506<br />

Bambury, Richard e19059<br />

Bamia, Christina e15040<br />

Bamias, Aristotelis 4524,<br />

4577, LBA5002^, 5033,<br />

e15040<br />

Bamlet, William R e14594<br />

Bampo, Chiara e21118<br />

Banasiak, James 6088<br />

Banat, Andre G. 3<br />

Banavali, Shripad 9559<br />

Bancr<strong>of</strong>t, Elizabeth 1545,<br />

4653<br />

Banda, Deliya e16543<br />

Bandali, Nesan e15177^<br />

Bandekar, Rajesh 2583,<br />

8018^<br />

Bandman, Olga TPS4699<br />

Bando, Etsuro e14635<br />

Bando, Hideaki 3079,<br />

e14038<br />

Bando, Inmaculada e14147<br />

Bandos, Hanna LBA506,<br />

TPS1615<br />

Bandres, Eva e14057<br />

Banek, Severine e15195<br />

Baneke, Alex e14696<br />

Banerjee, Mousumi 6056<br />

Banerjee, Nila 10508<br />

Banerjee, Prajna 10513<br />

Banerjee, Susana N. 3048,<br />

5012, 5022<br />

Banerji, Aleena e18520<br />

Banerji, Udai 3004, 3022,<br />

10525<br />

Banez, Lionel Lloyds 4670<br />

Bang, Bhavesh Amarnath<br />

e12014<br />

Bang, Hyun J e15081<br />

Bang, Yung-Jue 3076, 3624,<br />

4081, 4118, 4124, 7508,<br />

e14136, e14572<br />

Bangerter, Markus TPS3639<br />

Banks, Phillip Lee 4085,<br />

e14564<br />

Bannan, Michael 8589<br />

Banni, Sebastiano e19634<br />

Bannykh, Serguei I. 2080<br />

Bansal, Anuj Kumar 9549,<br />

9551<br />

Banwait, Ranjit 8043<br />

Banys, Malgorzata 5042,<br />

e21003<br />

Banzet, Michelle 1562<br />

Bao, Ting 1039, 9103,<br />

10572<br />

Bao, Weichao 2543<br />

Bapsy, P P e21132<br />

Bar Yehuda, Sara e14731<br />

Bar, Jair e13541, e17509<br />

Bar-Ad, Voichita e19507<br />

Bar-Hanna, Micha e18511<br />

Bar-Joseph, Hadas e13080<br />

Bar-Sela, Gil 9052<br />

Barabe, Frederic 634<br />

Baracos, Vickie E. 4621,<br />

5569<br />

Baracos, Vickie E e13118<br />

Barahona, Nancy 4006<br />

Baranda, Joaquina Celebre<br />

4010<br />

Baranger, Bernard e15524<br />

Baranzelli, Marie-Christine<br />

2070<br />

Barash, Steven 9125<br />

Barath, Sandor e21140<br />

Barbachano, Yolanda<br />

LBA4000, e14088<br />

Barbareschi, Mattia e21102<br />

Barbas, Coral e15111<br />

Barbazan, Jorge 10534<br />

Barbee, Meagan Spencer<br />

e18553<br />

Barber, Beth e19043,<br />

e19044, e19045, e19060<br />

Barber, Emma L. 5039<br />

Barber, Jim 4509<br />

Barber, Kenya TPS4698^<br />

Barbera, Lisa Catherine 6039<br />

Barberi, Simona e11514<br />

Barberis, Massimo 4120,<br />

7023<br />

Barbi, Stefano 4076<br />

Barbieri, Antonio e11052,<br />

e13539<br />

Barbieri, Elena 614, 631<br />

Barbieux, Hubert 1574,<br />

TPS7111<br />

Barboriak, Daniel TPS10635<br />

Barbour, Andrew TPS4145<br />

Barboza Quintana, Oralia<br />

e15525<br />

Barbui, Tiziano 6514^,<br />

6625^, 6626^<br />

Barbuto, Anna Maria 3559<br />

Barcenas, Carlos Hernando<br />

612<br />

Bardia, Aditya 1122, 9103<br />

Bareiss, Petra M. e15577<br />

Barfield, Michael E. e14724<br />

Barg, Frances K e19531<br />

Bargallo Rocha, Enrique<br />

e11016, e12027<br />

Bargfrede, Michael 4656<br />

Barginear, Myra F. 1046<br />

Bargou, Ralph 6500^<br />

Barham, Ruth e19543<br />

Bariani, Giovanni Mendonca<br />

6055, e20502<br />

Barile, Carmen e15160<br />

Barin<strong>of</strong>f, Jana 556, 1023<br />

Barista, Ibrahim 6550<br />

Barkan, Guliz e15014<br />

Barkauskas, Deborah Sim<br />

9520<br />

Barker, Christopher Andrew<br />

8549<br />

Barker, Peter 3055<br />

Barlesi, Fabrice TPS7111,<br />

7543, 7574, e13113<br />

Barletta, Giulia 7577,<br />

e21065<br />

Barlev, Arie e16512<br />

Barlogie, Bart 8041, e18548,<br />

e18576<br />

Barlow, William e16524<br />

Barnadas, Agust 1001, 1120,<br />

4079, 5590, 5595, 10555,<br />

10616, e11545, e14104,<br />

e14552, e15159, e18057<br />

Barnato, Sara E. TPS1134<br />

Barnes, Edward K. 7035<br />

Barnes, Jeffrey A. e18520<br />

Barnes, Tanganyika e14124,<br />

e14125, e14727<br />

Barnett, Anne Elizabeth 9039<br />

Barnett, Gene H. 2018,<br />

2076, 2100<br />

Barnett, Stephen e17539<br />

Barnett-Griness, Ofra 1578<br />

Barney, Brandon M. e14507<br />

Barnhart, Kerry M. 3060<br />

Barnholt, Kimberly E 10097<br />

Barnholtz-Sloan, Jill 1603<br />

Barnhurst, Kelly 2555<br />

Barni, Sandro 1073, 4627,<br />

7550, e19612<br />

Barnosky, Andrew R 9121<br />

Baron Toaldo, Marco 3061<br />

Baron, Andre T. e21000<br />

Barón, Anna E. 7601<br />

Barón, Anna E 10580<br />

Baron, Ari David e15115<br />

Barón, Francisco J. e19505<br />

Barone, Carlo 3502, e14113,<br />

e14156, e16553<br />

Barone, Julie 10554<br />

Barosi, Giovanni 6514^,<br />

6625^, 6626^, TPS6642^<br />

Barr, Frederick G. 9535<br />

Barr, John e19635<br />

Barr, Paul M. 6515^, 8032,<br />

8080<br />

Barr, Ronald 6011<br />

Barrack, Evelyn R e15174<br />

Barrajon, Enrique 4587,<br />

e16025<br />

Barranco, Charles TPS8116<br />

Barrellier, marie-Therese<br />

1580<br />

Barrera, Juan 1015<br />

Barret, Maximilien 9026^<br />

Barretina Ginesta, Maria Pilar<br />

1588<br />

Barrett, Cris e19522<br />

Barrett, John A e13040<br />

Barrett, Olivia Claire e15502<br />

Barrie, Maryline 2060<br />

Barrientos, Jacqueline Claudia<br />

6515^, 6518^<br />

Barriere, Jerome 2600<br />

Barrios, Carlos H. TPS670,<br />

7503, 7506, 7512,<br />

e11087<br />

Barriuso, Jorge 3601, 3618,<br />

10546, 10547, e13085,<br />

e14093<br />

Barron, Thomas Ian 521,<br />

2599, e13096<br />

Barroso, Ana e18111<br />

Barrow, David A. e13063<br />

Barry, Michael J 9004<br />

Barry, William Thomas<br />

CRA1002<br />

Barshack, Iris 10528,<br />

e14646<br />

Barta, Stefan K. 8005<br />

Barth, Juergen 3<br />

Barth, Matthew John e18537,<br />

e18539<br />

Barth, Neil M. 2569<br />

Barthelemy, Philippe 636<br />

Bartido, Shirley TPS2619,<br />

TPS4700<br />

Bartlett, Cynthia Huang 7596,<br />

7598, 7599, 7600<br />

Bartlett, David L. e14184<br />

Bartlett, John M. S. 516,<br />

517, TPS665<br />

Bartlett, John M.S. TPS1144<br />

Bartlett, John 6102<br />

Bartlett, Nancy 8000, 8027,<br />

8060, 8070<br />

Bartoli, Claudia 5546<br />

Bartolini, Stefania 2057,<br />

2061<br />

Bartolome, Adela e15033<br />

Bartolotti, Marco 2061<br />

Bartolozzi, Carlo TPS4148<br />

Barton, Debra L. 1557, 9001,<br />

9075<br />

Barton, John H. 1086<br />

Bartos, Claudia I. 5592<br />

Bartsch, Rupert 2053,<br />

e21033<br />

Bartzi, Valentina 5574<br />

Baruzzi, Agostino 2057<br />

Baryshnikov, Anatoly Y. 2524<br />

Barzi, Afsaneh 1514, 4088,<br />

e14031<br />

Bas, Carlos Arturo e15570<br />

Basak, Jayasri e18043<br />

Basaran, Gul e16015<br />

Basaria, Shehzad e15139<br />

Basch, Ethan M. 9042,<br />

9047, 9104, 9144,<br />

e19633, e19647<br />

Bascioni, Romeo 9098<br />

Baselga, José LBA1, 508,<br />

509, 512, 533^, 539, 540,<br />

551, 559, 566, 597^,<br />

598^, 1009, 1011, 2042,<br />

2567, 2568, TPS3118,<br />

e11055^, e13048<br />

Basen-Engquist, Karen<br />

e11090<br />

Bashir, Omer e15140<br />

Bashir, Qaiser 6546, 8040,<br />

8102<br />

Basic Koretic, Martina<br />

e21126<br />

Basik, Mark TPS1139<br />

Basile, Assunta e13098<br />

Basirat, Farnoosh 9098<br />

Basiricò, Marco 10066<br />

Baskin-Bey, Edwina S.<br />

TPS4698^<br />

Bass, J. David 1018<br />

Bass, Joyce 6528<br />

Bassett, Mitchell 4670<br />

Bassett, Roland 4533, 6540,<br />

e19009, e19014<br />

Bassi, Claudio e14625<br />

Bassi, Fabio 519<br />

Bassim, Carol 4569<br />

Basso, Enrico e11039,<br />

e11514, e12510, e19620<br />

Basso, Michele e14156,<br />

e16553<br />

Basso, Umberto e15160<br />

Bassot, Karine 636<br />

Bast, Martin 8047<br />

Baste, Neus 1588<br />

Basterretxea, Laura TPS4685<br />

Bastholt, Lars 8545<br />

Bastian, Boris 10524<br />

Bastie, Anne e14129<br />

Bastie, jean Noel 6523<br />

Bastien, Berangere TPS7610<br />

Bastos-Rodrigues, Luciana<br />

e13008<br />

Basu, Bristi 3004<br />

698s Numerals refer to abstract number


Basu, Gargi D. 1040, 10630,<br />

e13571, e15209<br />

Basu, <strong>Part</strong>ha 5106, e15546<br />

Basu, Sanjib 7067, e16503,<br />

e18117, e21069<br />

Bataille, Vincent e19019<br />

Batchelor, Tracy 2005, 2009,<br />

2010, 2012, 2025, 3036<br />

Bates, Andrew T. 8056<br />

Bates, Gleneara e13067<br />

Bates, Susan Elaine 2548,<br />

2553, e13122<br />

Batist, Gerald 1099,<br />

TPS1139, e14020<br />

Batista, Norberto e15149<br />

Batista, Ranyell Spencer<br />

Sobreira 10069<br />

Batistatou, Anna 592<br />

Batra, Surinder e17549<br />

Bats, Anne-Sophie 5098,<br />

e15558<br />

Battaglia, Leslie 5523<br />

Battaglia, Luigi e21118<br />

Battaglia, Tracy Ann 6097<br />

Battista, Kristina 3082<br />

Battista, Marco Johannes<br />

5087<br />

Battista, Michle e12514<br />

Battley, Jodie E 2062,<br />

e11569, e14666, e14687,<br />

e15078, e19059<br />

Battolla, Enrico e17533<br />

Batty, Nicolas 8067, e15042,<br />

e18538<br />

Batus, Marta 7067, e18117<br />

Baudin, Eric 4014, 4071,<br />

5569<br />

Baudrin, Laurence e18089<br />

Baudry, Elodie e18505<br />

Bauer, Hillevi 6583<br />

Bauer, Joseph E. 1608<br />

Bauer, Juergen 8526<br />

Bauer, Kenneth S e13075<br />

Bauer, Maret 1084<br />

Bauer, Sebastian 10032^,<br />

10033, 10046<br />

Bauerfeind, Ingo 1114<br />

Bauersachs, Rupert<br />

TPS9149^<br />

Bauerschlag, Dirk Olaf<br />

e13503<br />

Bauerschlag, Dirk 556<br />

Baujat, Bertrand e16013<br />

Bauknecht, Thomas 1059<br />

Baulieu, Jean-Louis TPS646<br />

Baum, George e11090<br />

Bauman, Glenn 6130<br />

Bauman, Jessica R e16062<br />

Bauman, John W. 5065<br />

Bauman, Julie E. 3009,<br />

5503, 5515, 5549, 5566,<br />

TPS5602, 7029, e13101<br />

Baumann, Klaus H. 5005,<br />

5007<br />

Baumann, Sybille e19530<br />

Baumgart, Megan Ann 5529,<br />

5532<br />

Baurain, Jean-Francois 5038,<br />

8532, 8539<br />

Bausela, Cristina e14147<br />

Bautista, Dolores e14116<br />

Bavbek, Sevil E. e15112<br />

Baxevanis, Constantin N.<br />

e15125<br />

Baxi, Shrujal S. 5514, 6001<br />

Baxley, Allison Ashley e17001<br />

Baxter, Nancy N. 6027<br />

Baxter-Lowe, Lee Ann 6537<br />

Numerals refer to abstract number<br />

Bay, Jacques-Olivier 4625,<br />

6527, 6542, 10020,<br />

10035, 10042, 10078<br />

Bayer, Charlene W. 7048<br />

Baylin, Stephen 7006,<br />

e18147<br />

Baylon, Sheley M e16523<br />

Bayman, Neil e18095<br />

Bayo Calero, Juan Lucas<br />

e11081<br />

Bayo, Juan L. 10608,<br />

e11075, e11535, e21088<br />

Baz, Rachid C. 8016, 8017<br />

Bazeos, Alexandra 10550<br />

Bazhenova, Lyudmila 2052,<br />

7030, e16535, e21074<br />

Bazin, Igor e14623<br />

Bazolli, Barbara 1115<br />

Bazzi, Mouhamad e13075<br />

Bazzola, Letizia e11546<br />

Beach, Douglas Francis<br />

e12008<br />

Beadle, Beth Michelle 5561,<br />

5589<br />

Beadling, Carol 10591<br />

Beale, Philip James 3531,<br />

9021, 9087, 9126<br />

Beaney, M. P. e14022<br />

Bear, Harry Douglas LBA506,<br />

10565<br />

Bearden, James D. 9047<br />

Beardslee, Brian e11529<br />

Beare, Sandra 4004, 4029<br />

Bearelly, Smitha 6561<br />

Bearss, David 10549<br />

Beart, Robert W. TPS10632<br />

Bearzi, Italo e14561, e21070<br />

Beato, Carlos Augusto de<br />

Mendonça 7506<br />

Beatson, Melony 4569<br />

Beatty, Gregory Lawrence<br />

2500^<br />

Beau-Faller, Michele 10507<br />

Beaubien, Ron 10587<br />

Beauchemin, Michel 634<br />

Beauchene, Suzanne e15206<br />

Beaumont, Hubert e13547<br />

Beaumont, Jennifer L 6092<br />

Beaunoyer, Mona 9563, 9570<br />

Beausoleil, Michel S 4535<br />

Beaussant, Yvan e19623<br />

Bebb, D. Gwyn e14073,<br />

e18110, e21083<br />

Bebek, Gurkan 5573<br />

Becchimanzi, Cristina 8066<br />

Becerra Ramirez, Henry<br />

Alberto e18114<br />

Becerra, Carlos 508, 2516,<br />

3023, TPS3111<br />

Becerra, Henry A. e12039<br />

Bechade, Dominique e14017,<br />

e14023<br />

Bechstein, Wolf 3508,<br />

TPS3641^<br />

Bechter, Oliver Edgar e14719<br />

Beck, Christian 6584<br />

Beck, J Robert 6021<br />

Beck, J. Thaddeus LBA4,<br />

530, 539, 8511<br />

Beck, Mo Youl 10085<br />

Beck, Robert J e14021<br />

Beck, Rodney A. 516<br />

Beck, Susan L. 9137<br />

Beck, Thomas 1600^<br />

Becker, Daniel Jacob 6114,<br />

e18139<br />

Becker, Dorothea 8528<br />

Becker, Gerd 5512<br />

Becker, Heinz 3600, e14161,<br />

e14167<br />

Becker, Jürgen C. 8517<br />

Becker, Robert 3074<br />

Becker, Shawn 10584<br />

Becker-Hapak, Michelle 2525<br />

Beckers, Marielle e13062<br />

Beckett, Brooke 10011^<br />

Beckett, Laurel 7582, 7594,<br />

e13550<br />

Beckjord, Ellen Burke 9136<br />

Beckman, Robert A. 7536,<br />

e14617<br />

Beckmann, Matthias W. 554,<br />

1600^, 1602, 10540<br />

Becouarn, Yves e14017,<br />

e14023<br />

Becq, Jennifer 544<br />

Bedair, Ahmed e16002<br />

Bedard, Philippe 3003,<br />

3082, 4535, e13001,<br />

e19616<br />

Bedenne, Laurent LBA4003<br />

Bedetti, Benedetta 10080<br />

Bedikian, Agop Y. 8514,<br />

8587, e19009, e19014,<br />

e19039<br />

Bedke, Jens 4530<br />

Bedognetti, Davide 8576,<br />

10565<br />

Bedoya, Ivan Dario e14714<br />

Bedrosian, Isabelle 1102<br />

Beebe-Dimmer, Jennifer<br />

e14609, e15147<br />

Beecham, Kwamena e15163<br />

Beeker, Aart TPS4692^<br />

Beelen, Karin J. 562<br />

Beer, David G. 10587<br />

Beer, Tomasz M. 4522,<br />

4525, TPS4691<br />

Beeram, Muralidhar 2512,<br />

2555, 3001, 3019,<br />

e13025, e13602, e13604<br />

Beg, Muhammad Shaalan<br />

TPS3116, e14165<br />

Begbie, Stephen e19642<br />

Begg, Colin B. CRA9513<br />

Beghelli, Stefania 4076<br />

Begin, Marie Josee 2102,<br />

e18120<br />

Begna, Kebede 6614<br />

Begnami, Maria D. e14728<br />

Beguin, Yves 6534<br />

Begum, Rubina 4004, 4029<br />

Begum, Tahmina e11539<br />

Beham-Schmid, Christine<br />

e18525<br />

Behera, Digambar e18009<br />

Behera, Madhusmita 7059<br />

Behera, Manoj Kumar e11082<br />

Behl, Ajay S e14160<br />

Behlendorf, Timo e19579<br />

Behr, Spencer 8580<br />

Behren, Andreas 8542<br />

Behrendt, Carolyn E. e19644<br />

Behrens, Carmen 7023<br />

Behrens, Robert J. TPS657<br />

Behringer, Dirk M. 4083<br />

Behrns, Kevin TPS4136<br />

Beijert, Max L 8039<br />

Beijnen, Jos H. 2550, 2596,<br />

e13028<br />

Beiko, Jason 2019<br />

Beissbarth, Tim 3600,<br />

e14161, e14162<br />

Beitler, Jonathan Jay 5560,<br />

5570, 7059, e21161<br />

Bejanyan, Nelli 8055<br />

Bekaii-Saab, Tanios B 4099<br />

Bekaii-Saab, Tanios S. 4039,<br />

4043, TPS10102^,<br />

e14169, e14170<br />

Bekers, Dave J. 10091<br />

Beksac, Meral 8095,<br />

e18572^, e21006<br />

Bekyashev, Ali H. e19012<br />

Belanger, Karl 2102<br />

Belani, Chandra Prakash<br />

TPS1138, 2553, 3049,<br />

3054, 7040, 7054, 7055,<br />

7512, 7551, e17560,<br />

e18138<br />

Belau, Antje 522, 624, 5031<br />

Belda-Iniesta, Cristobal 3068,<br />

3601, e13085, e14093,<br />

e14144<br />

Belderbos, Jose 7019<br />

Belghiti, Jacques 3609,<br />

e14553<br />

Belitsis, Kenneth 1540<br />

Belizna, Cristina 1580<br />

Belka, Claus TPS7617,<br />

e16033<br />

Belknap, Steven M 2532,<br />

4063, 6623<br />

Belk<strong>of</strong>f, Laurence 4550<br />

Bell, Melanie 6111, e16507<br />

Bell, Richard 502, 513,<br />

TPS670<br />

Bell, Ronny A. 6127<br />

Bell, Sue E 8015<br />

Bella, Santiago Rafael<br />

e21025<br />

Bellati, Filippo 5070<br />

Belldegrun, Arie S. e13045<br />

Belleannee, Genevieve 4129<br />

Bellera, Carine 9012<br />

Bellet, Meritxell 509, 566,<br />

619<br />

Belletier, Christine e11542<br />

Bellevicine, Claudio e19038<br />

Bellew, Kevin M. 3003, 3023<br />

Bellezza, Guido e21094,<br />

e21096<br />

Belli, Carmen 4017<br />

Belli, Susana e14598<br />

Bellin, Lisa Sheri e11550<br />

Belliveau, James e13561<br />

Bellizzi, Keith M 9130<br />

Bellmunt, Joaquim TPS4674,<br />

TPS4693, e15007, e15111<br />

Bello, Carlo e18093<br />

Bello, Celeste M. 2099,<br />

6568, 8065, 8084,<br />

e18503, e18506, e18509,<br />

e21123<br />

Bellon, Jennifer Ruth 6022<br />

Belloni, Benedetta 8563,<br />

e19017<br />

Belloni, Carlo 1500<br />

Belluco, Claudio e14151<br />

Belmans, Ann 1020<br />

Belmar, Nicole 8087<br />

Belmont, Laure e18075,<br />

e18102<br />

Belotti, Alessandro 3570<br />

Belounis, Assila 9563, 9570<br />

Belousova, Elena 4632,<br />

10619<br />

Beltran, Himisha 4649<br />

Beltran, Michel e16044,<br />

e16055<br />

Beltran, Miguel 1588,<br />

e15144<br />

Belvederesi, Laura e14561,<br />

e15074<br />

Belvin, Marcia 2566<br />

Ben, Yong LBA4537<br />

699s


Ben-Aharon, Irit 548, 2556,<br />

e13080<br />

Ben-David, Merav A. e14143<br />

Ben-Josef, Edgar e14603<br />

Ben-Or, Sharon e17550<br />

Benaim, Ely 2597, 3010,<br />

5021, TPS8109, TPS8110<br />

Benakanakere, Indira 6561,<br />

e11031<br />

Benali-Furet, Naoual e21014,<br />

e21046<br />

Benassi, Maria Serena 10066<br />

Benazzouna, Rana 9560<br />

Benboubker, Lotfi 8009,<br />

8014, 8020<br />

Benbrahim, Zineb e11502<br />

Bencardino, Katia 3570,<br />

TPS3637<br />

Benchimol, Daniel 5075<br />

Bendahmane, Belguendouz<br />

CRA3503<br />

Bendell, Johanna C. 2566,<br />

3006^, 3008, 3021, 3033,<br />

3041, 3067, 3097,<br />

TPS3118, LBA3501, 3543,<br />

3631, TPS3640, 4027,<br />

4127, e13076<br />

Bender, James 2087<br />

Bender, Ryan P 10630,<br />

e15209<br />

Bendszus, Martin 2034<br />

Bene, Marie C. 8026<br />

Benecchi, Sara e18039<br />

Benedetti Panici, Pierluigi<br />

LBA5003, 5070, 5093,<br />

5094, e15547, e15550,<br />

e15551, e15553<br />

Benedetti, Antonio e14561<br />

Benedetti, Fabio M. 3631<br />

Benedetti, Jacqueline K.<br />

4019<br />

Benedetti, Jacqueline 4010<br />

Benedikova, Andrea e14556<br />

Benes, Cyril 1055<br />

Benevelli, Carlotta 627<br />

Benevides, Carlos Frederico<br />

Lopes 643<br />

Benezery, Karen 5521,<br />

e16044, e16051, e16055<br />

Benfante, Antonina e21095<br />

Bengrine-Lefevre, Leila 4071<br />

Benhadji, Karim Adnane<br />

2554, 3005<br />

Benhaim, Leonor 3584,<br />

3597, 3604, 3615, 3622,<br />

4026, 4088, e11018,<br />

e14034, e14052<br />

Benhammane, Hafida e11516<br />

Benhar, Itai e13018<br />

Benitez, Julio 4620<br />

Benito, Juliana M. 6585<br />

Benizri, Emmanuel 5075<br />

Benjamin, Hila 10528,<br />

10575, e21008<br />

Benjamin, Kamlakshi Esther<br />

e19622<br />

Benjamin, Robert S. 10021,<br />

10071, e19520<br />

Benjaminov, Ofer e14731<br />

Benlloch, Susana 4068,<br />

10596, e14521, e18130,<br />

e18131, e18137, e18143<br />

Bennati, Chiara e18023,<br />

e18101<br />

Bennett, Antonia V 9144<br />

Bennett, Barbara Kaye 9044<br />

Bennett, Cathryn M.<br />

TPS10103<br />

Bennett, Charles L. 6044,<br />

9143, e19639<br />

Bennett, Dimitri e12001<br />

Bennett, Kathleen 521,<br />

2599, e13096<br />

Bennett, Kristi L 5573<br />

Bennett, Lee 539<br />

Bennett, Michael WIlliam<br />

e14666, e14687<br />

Bennis, Robyn 10574<br />

Bennouna, Jaafar 3010,<br />

CRA3503, LBA7500, 7557,<br />

7575, TPS7612, e13009,<br />

e13010, e18005<br />

Bensaid, Benoit 10049<br />

Bensaid, Cherazade 5098,<br />

e15558<br />

Bensfia, Samira e15112<br />

Bensinger, William 8034<br />

Bensink, Mark Eliot 6077<br />

Benson, Al B. e14542,<br />

e14551, e14610<br />

Benson, Al Bowen 3605,<br />

4030, 4102, 4126<br />

Benson, Brad e16502<br />

Benson, Charlotte 10038,<br />

10039, 10060<br />

Benson, Mitchell C. 2516,<br />

e15021<br />

Benteyn, Daphné 2507<br />

Bentley, David 544<br />

Bentley, John e16527<br />

Bentley, Tanya G. 3626,<br />

6057<br />

Benton, Christopher Brent<br />

6576<br />

Bentrem, David J. e14642<br />

Bentrem, David J 3608<br />

Benyunes, Mark 533^,<br />

e11055^<br />

Benz, Christopher 10586<br />

Benz, Matthias R. 10012<br />

Benzel, Heather 9110<br />

Bepler, Gerold 2559, 7063,<br />

7593, e15005<br />

Beppu, Yasuo 10009, 10067<br />

Berahovich, Robert D. 2043<br />

Berano-Teh, Jennifer 1592,<br />

8038<br />

Berard, Henri e16560<br />

Berard, Paul e18558<br />

Berardi, Rossana 4627,<br />

10063, e21070<br />

Berardi, Simona 9059<br />

Berchmans, Emmanuel 6575<br />

Berdeja, Jesus G. 8033,<br />

TPS8115<br />

Berdel, Wolfgang E. 6610,<br />

e15026<br />

Bereder, Jean Marc 5075,<br />

e15574<br />

Berek, Jonathan S. 5065,<br />

TPS5113<br />

Berenguel, Maria e19636<br />

Berenson, James R. 8034,<br />

8098, e18549, e18558<br />

Beretta, Giordano D. 623,<br />

7082, TPS9155<br />

Berg, Deborah 8017, 8033,<br />

8034, 8064<br />

Bergagnini, Isabella 7033<br />

Bergamini, Cristiana 5555<br />

Bergamo, Francesca 3555,<br />

3585, 4017, 9090<br />

Berge, Eamon M 7504<br />

Berge, Eamon 7601, e21120<br />

Berge, Victor e15194<br />

Berge, Yann 3026, 3104,<br />

5522, 10027<br />

Bergelson, Ilana e20008<br />

Berger, Allison 3077<br />

Berger, Andre K e15060<br />

Berger, Andreas TPS1612,<br />

TPS3636<br />

Berger, Lars Arne 4597<br />

Berger, Markus 5047<br />

Berger, Michael F. 4604,<br />

5030, 10524<br />

Berger, Michael F 4527<br />

Berger, Raanan 535, 3027,<br />

4619, e14143, e14646,<br />

e15058<br />

Bergerot, Cristiane Decat<br />

e19504<br />

Bergethon, Kristin 7508<br />

Bergfeldt, Kjell 5052<br />

Bergh, Jonas 10083, e11510<br />

Berghmans, Thierry e18006<br />

Bergh<strong>of</strong>f, Anna Sophie 2053,<br />

e21033<br />

Berghout, Koralee 10620<br />

Bergman, Bengt 7001<br />

Bergmann, Lothar e15044<br />

Bergnolo, Paola 10055<br />

Bergqvist, Michael 7539<br />

Bergqvist, Peter e15199^<br />

Bergsagel, Leif 8043<br />

Bergsagel, P. Leif 8010<br />

Bergsland, Emily K. 4085,<br />

4126, e13000, e14542,<br />

e14551<br />

Bergstralh, Eric J. 4565<br />

Bergstrom, Donald Alan<br />

10573<br />

Bergstrom, Kimberly A.<br />

e14118<br />

Bergström, Stefan 7539<br />

Berigaud, Sylvie e18505<br />

Berille, Jocelyne LBA4003<br />

Berinstein, Neil Lorne 2588<br />

Berk, Lawrence B. TPS9150<br />

Berk, Veli e14089, e14670<br />

Berkenblit, Anna 4501<br />

Berking, Carola 8511<br />

Berkovcova, Jitka e19020<br />

Berkowitz, Alison e19647<br />

Berkowitz, Noah C. 1017<br />

Berlin, Jordan 2595, 3532,<br />

4052, e13606, e14503<br />

Berloco, Pasquale 4110<br />

Berman, Arlene W 7033,<br />

7103<br />

Berman, David M. 4577,<br />

4580, 4582<br />

Berman, David Mark 8541,<br />

8593<br />

Berman, David Monty 4585<br />

Berman, Ellin 6613, 9088<br />

Berman, Russell S. 8550,<br />

8557, 8565, 8589,<br />

e19003<br />

Berman, Tara 5078<br />

Bermejo, Begoña 1074,<br />

10500, 10616, e11523<br />

Bermejo, Justo Lorenzo 1096<br />

Bermingham, Niamh 2062,<br />

e11569<br />

Bernabé, Reyes 1531,<br />

e11075<br />

Bernales, Sebastián 3072<br />

Bernard, jean-Louis 5075<br />

Bernard, Laurence 3539,<br />

7090<br />

Bernard, Paquito 9074<br />

Bernard, Paula J e14075<br />

Bernard, Philip Seth 1008<br />

Bernard, Stephen A. 6042<br />

Bernardi, Daniele 586<br />

Bernardini, Marcus 5026<br />

Bernardo, Antonio 589<br />

Bernardo, Giovanni 589<br />

Bernards, Karen e13542<br />

Bernards, Rene’ 3065<br />

Berneman, Zwi N. 2506,<br />

e13051<br />

Bernemann, Christ<strong>of</strong> e11534<br />

Bernhard, Helga 2573^<br />

Bernhard, Juerg 9022<br />

Bernhardt, Christiane 3550<br />

Berno, Elisa 10055<br />

Berns, Els M. 526<br />

Bernsen, Hans J.J.B. 2<br />

Bernshaw, David e15541<br />

Bernstein, Jonine L. CRA9513<br />

Bernstein, Lori J 5587<br />

Bernstein, Mark L. 9503,<br />

9537, 10081<br />

Bernstein, Melanie 4552<br />

Bernstein, Sarah 2577<br />

Bernstein, Steven H. e18536<br />

Bernstein-Molho, Rinat 10043<br />

Beroukhim, Rameen 2020<br />

Berreho, Mohamed Amine<br />

e11502<br />

Berremili, Toufik 1580<br />

Berretta, Massimiliano<br />

e14040<br />

Berriolo-Riedinger, Alina<br />

TPS646<br />

Berrocal, Alfonso e19023<br />

Berros, Jose Pablo e18044,<br />

e19023<br />

Berruti, Alfredo 4627<br />

Berry, Allen 7072^<br />

Berry, Donna Lynn 4571,<br />

9008, e19586, e19637<br />

Berry, John S. 625<br />

Berry, Kristin 6077<br />

Berry, Lynne D 7589<br />

Berry, Mark Francis e14562<br />

Berry, Scott R. e14171,<br />

e15177^, e16558<br />

Berry, William R. TPS4697<br />

Berryman, Robert B. e13079<br />

Bersani, Maurizio TPS9155<br />

Bersani, Samantha 4076<br />

Bertagnolli, Monica M. 3517,<br />

10017<br />

Bertelli, Gianfilippo 1094<br />

Bertelsen, Caitlin 1065<br />

Bertero, Luca 2037, 2075<br />

Berthelet, Eric 7020<br />

Berthet, Pascaline e12502<br />

Berthod, Grégoire e19050<br />

Bertolaso, Laura e13019<br />

Bertoldo, Francesco 9005,<br />

10063<br />

Bertolini, Francesco 2083<br />

Bertolini, Stephane V.<br />

TPS8111<br />

Berton-Rigaud, Dominique<br />

5018, e13095^, e13097^,<br />

e13108^, e15535<br />

Bertoni, Ramona e11546<br />

Bertorelle, Roberta 2049<br />

Bertossi, Monica 2580<br />

Bertran-Alamillo, Jordi 7522,<br />

10596, e14521, e18131,<br />

e18143<br />

Bertrand, Claude e14148<br />

Bertrand, Yves TPS9594<br />

Bertschinger-Ehrler, Julian<br />

2575<br />

700s Numerals refer to abstract number


Bertucci, François 1047,<br />

10014, 10023, 10042,<br />

10056, 10078, 10092,<br />

10095, TPS10102^,<br />

e21010<br />

Bertulli, Rossella TPS9155,<br />

10053, 10065<br />

Berube, Jayne 549^<br />

Berwick, Marianne 8574<br />

Berz, David 4098<br />

Besse, Benjamin 7543,<br />

7599, 10507, 10556,<br />

e18071<br />

Besson, Caroline 8046<br />

Besson, Nadine 6511<br />

Bessudo, Alberto 2579,<br />

e13101<br />

Best, Kate e17519<br />

Bestani, Claudia e14598<br />

Besterman, Jeffrey M. 3039,<br />

e13602, e13604<br />

Bestoso, Elena e13098<br />

Betancur, Carlos Alberto<br />

e19604<br />

Betensley, Alan e12019<br />

Beteta, Carmen Rosa e19597<br />

Bethe, Ullrich 5516^<br />

Bethel, Kelly e21074<br />

Bethke, Kevin P. TPS1134<br />

Betsuyaku, Tomoko e18058<br />

Betta, Pier Giacomo 7084<br />

Bettelli, Stefania Raffaella<br />

631<br />

Betticher, Daniel C. e19648,<br />

e18012<br />

Bettini, Anna LBA7501<br />

Bettis, Claudette 8524<br />

Betts, Kelly 9103<br />

Beumer, Jan H. TPS1138<br />

Beumer, Jan Hendrik 1070,<br />

2533, 2553, 3031, 3037,<br />

3049, 3054, 7022,<br />

TPS8606<br />

Beuthien-Baumann, Bettina<br />

10068<br />

Beutler, Thomas e14045<br />

Beuzeboc, Philippe 1095,<br />

LBA4567^, 4583, 5067,<br />

e15535<br />

Bevan, Catherine e15501<br />

Bevan, Paul 1092<br />

Beveridge, Roy A. 6109,<br />

e16536<br />

Bevers, Michael W. 9134<br />

Bevilacqua, Generoso 629<br />

Bevis, Kerri S. 5060<br />

Bex, Axel 4611<br />

Bexon, Alice Susannah<br />

e18047<br />

Beyeler, Michael 9059<br />

Beyer, Joerg 4597, 4600,<br />

e15026<br />

Beygi, Hooman e13081<br />

Beyne-Rauzy, Odile 6523<br />

Beyssac, Richard e14023<br />

Bezerra, Stephania 643<br />

Bhakta, Radhe S. 2581<br />

Bhalla, Kapil N. 10549,<br />

10605, e13568<br />

Bhalla, Megha e21047<br />

Bhamidipati, Pavan Kumar<br />

6597<br />

Bhangu, Aneel e14132<br />

Bhardwaj, Nina 2570, 2589,<br />

8571, e21081<br />

Bhardwaj, Vikas 7096<br />

Bhargava, Pankaj 4112, 4132<br />

Bhargava, Ranjana S. 1039<br />

Numerals refer to abstract number<br />

Bhargava, Rohit 1038, 1085,<br />

e15560<br />

Bharthuar, Anubha e19607<br />

Bhat, Ashwini 10037<br />

Bhat, Krishna 2002<br />

Bhat, M L e19516<br />

Bhat, Neal 5088<br />

Bhathena, Anahita e13583<br />

Bhatia, Jasmine 5043<br />

Bhatia, Ravi 1592<br />

Bhatia, Smita 1592, 8038,<br />

9505, 9512, CRA9513<br />

Bhatla, Neerja 5104<br />

Bhatnagar, Aseem Rai<br />

e16012<br />

Bhatnagar, Bhavana 6611,<br />

e19565<br />

Bhatt, Aashish D. e21072,<br />

e21144, e21149<br />

Bhatt, Astha e14689<br />

Bhatt, Geetika e21072,<br />

e21144, e21149<br />

Bhatt, Niraj 2592<br />

Bhattacharya, Sudin 2581<br />

Bhattacharyya, Anirban<br />

TPS10631<br />

Bhattacharyya, Helen 6092<br />

Bhattacharyya, Pritish e18507<br />

Bhimani, Chandar 7059<br />

Bhor, Menaka e15063<br />

Bhora, Faiz Y. e18139<br />

Bhosale, Priya 4054<br />

Bhutani, Divaya 6549<br />

Bhutani, Manisha 8088,<br />

8104, e18568<br />

Bhutani, Manoop S. 4069,<br />

4078, 4086, e14547,<br />

e14548, e14669<br />

Bhutta, Usman e17012<br />

Bi, Feng LBA4537, 4638<br />

Bi, Nan e14511<br />

Biagetti, Simona e14086,<br />

e14561, e21070<br />

Biaggi, Christine 10033<br />

Biagi, James Joseph 3632,<br />

5019<br />

Biagini, Matteo 4015<br />

Biagioni, Francesca 3521<br />

Biakhov, Mikhail 7090<br />

Bian, John 6044<br />

Bianca, Ralph e19609<br />

Bianchetti, Sara e14082,<br />

e18065<br />

Bianchi, Andrea 8077<br />

Bianchi, Maria Paola e17006,<br />

e18563, e18566<br />

Bianchini, Carlo e19612<br />

Bianchini, Diletta 4553,<br />

e15134, e15136<br />

Bianco, Roberto e13509<br />

Biancon, Antonella e19595<br />

Bianconi, Fortunato e21094,<br />

e21096<br />

Bianconi, Maria Ines e15521<br />

Bianconi, Maristella e14086,<br />

e14561, e14663, e15074<br />

Biankin, Andrew Victor<br />

TPS4137<br />

Bias, Peter 9125, e19587<br />

Biasco, Guido 10087,<br />

e14647<br />

Bibbo, Marluce 10528<br />

Bibby, David C. 4603, 7079^<br />

Bibeau, Frédéric 10505,<br />

TPS10632, e14059<br />

Bible, Keith Christopher 5544<br />

Bichev, Dmitry TPS4138<br />

Bickel, Hubert 10570<br />

Bickell, Nina e11004,<br />

e15513, e19621<br />

Bickerstaff, Matthew 8523<br />

Bickley, Thomas e15536<br />

Bidard, Francois-Clement<br />

8546<br />

Biddle, Andrea K 6096,<br />

e15184<br />

Bidoli, Paolo LBA4001,<br />

LBA7507, 7557<br />

Bie, Li 10543<br />

Bieber, Carolyn 5544<br />

Bieche, Yvan 10507<br />

Bielack, Stefan S. 9573,<br />

10081<br />

Bierman, Philip Jay 8047<br />

Biernat, Wojciech 505, 603,<br />

10561<br />

Biersack, Hans-Juergen<br />

e14565<br />

Biesma, Bonne 7554<br />

Biganzoli, Laura 530<br />

Bigbee, William L e14689<br />

Bigelow, Elaine e14585<br />

Biggi, Alberto 8077, e13093<br />

Biggs, David D. LBA1000<br />

Biggs, David e18122<br />

Biggs, Heather 544<br />

Biglia, Nicoletta 627<br />

Bignon, Emmanuelle e14168<br />

Bignotti, Eliana e15549<br />

Bihrle, Richard 4586<br />

Bijwaard, Karen 3074<br />

Bikov, Kaloyan A e14035,<br />

e14056<br />

Bilen, Mehmet Asim 6544,<br />

e13506<br />

Bilici, Mehmet e14526<br />

Bilim, Vladimir e15057,<br />

e15088<br />

Bilimaga, Ramesh s e19619<br />

Bilimoria, Karl Y. 3608<br />

Bilker, Warren B. 1503<br />

Bill, Matthew A e13549<br />

Billett, Amy 9502<br />

Billiter, David 9583<br />

Billmire, Deborah 9539<br />

Billups, Catherine 9502<br />

Bilotto, Anthony 2511<br />

Bilusic, Marijo 2526,<br />

TPS2617, TPS4701<br />

Binder, Claudia e15026<br />

Binder, Gary e18560,<br />

e18561<br />

Bines, Jose e11087<br />

Bingman, Anissa 4039<br />

Bingoel, Cigdem 4601<br />

Biniecka, Monika e21024<br />

Binmoeller, Kenneth e14546<br />

Binns, Shemeica N. 561<br />

Bintner, Marc 2048<br />

Biondani, Pamela e21118<br />

Biondi-Zoccai, Giuseppe<br />

e15192<br />

Biondo, Andrea 10525<br />

Biondo, Sebastiano 3571<br />

Biran, Haim e17509<br />

Birch, Kate E. 1502<br />

Bird, Brian Richard e14111<br />

Bird, Deborah TPS4150<br />

Bird, Ian M e15167<br />

Birgisson, Helgi 3576<br />

Birkemeyer, Elke 4015<br />

Birkhäuser, Frédéric D.<br />

e13045<br />

Birkmann, Josef 8031<br />

Birks, Peter Crawford 6085<br />

Birnbaum, Ariel E. e13124<br />

Birner, Peter 2053, e21033<br />

Bironzo, Paolo e15192<br />

Birrer, Michael J. 5029,<br />

TPS5112<br />

Birt, Michael 6077<br />

Birtwisle-Peyrottes, Isabelle<br />

10018<br />

Bisch<strong>of</strong>f, Farideh Z. 584,<br />

10516, TPS10631<br />

Bisch<strong>of</strong>f, Joachim 556, 624<br />

Bisch<strong>of</strong>f, Sven 4044<br />

Biscotti, Tommasina e21070<br />

Bisen, Ajit e15121<br />

Bisgaier, Charles L. TPS3110<br />

Bishnoi, Sarwan K. e13007<br />

Bishop, Justin A. 5549<br />

Biskup, Karina 5047<br />

Bisovskaya, Yulia e14681,<br />

e14723<br />

Bissler, John 4632, 10619,<br />

e12042, e15096<br />

Biswas, Jaydip 2581<br />

Biswas, Tithi e17547,<br />

e17550<br />

Bitas, Christiana 10003<br />

Bitker, Marc-Olivier e15105<br />

Bitoun, Laurence 4036, 8591<br />

Bitran, Jacob D. e12002<br />

Bitting, Rhonda Lynn 10533<br />

Bittoni, Alessandro e14086,<br />

e14561, e14663, e15074<br />

Bitzer, Michael 4115<br />

Biville-Hedouin, Fabienne<br />

7575<br />

Bivona, Trever Grant 7522<br />

Bizieux-Thaminy, Acya<br />

e16560<br />

Bjarnason, Georg A. 4536,<br />

4538, 4613, 4633,<br />

TPS4683, 10622<br />

Bjelic-Radisic, Vesna 514<br />

Bjerregaard, Jon Kroll<br />

e14517, e14633<br />

Bjordal, Kristin 6002<br />

Bjorkholm, Magnus 8074<br />

Bjurlin, Marc e15119<br />

Blachar, Arye 1564<br />

Black, Amanda 4646<br />

Black, Amber A. e21090<br />

Black, Keith L. 2080, 2084,<br />

2087<br />

Black, Peter C. 4565<br />

Blackford, Amanda L. 6026<br />

Blackhall, Fiona Hellen<br />

e18095<br />

Blackstein, Martin E.<br />

LBA10008<br />

Blackwell, Kimberly L. LBA1,<br />

1006<br />

Blackwell, Kimberly TPS1148<br />

Blade, Joan 8018^<br />

Blaes, Anne Hudson 4659,<br />

e15169, e16502<br />

Blagden, Sarah Patricia 2608,<br />

3000, 3022, e13033<br />

Blais, Normand e17507,<br />

e18120, e18136, e19591<br />

Blaise, Didier e18526<br />

Blakaj, Dukagjin e16058<br />

Blake, Monika 7068<br />

Blake, Robert L. e19617<br />

Blakeley, Jaishri O’Neill 2042<br />

Blakemore, Stephen 3077<br />

Blakesley, Rick E. 6509^<br />

Blana, Andreas e15194<br />

Blanc, Ellen 4614, 4625<br />

Blanc, Jean-Frédéric 4032,<br />

TPS4146, 9012<br />

Blanc-Bisson, Christele 9012<br />

701s


Blancas, Isabel e11535,<br />

e11545, e21088<br />

Blanchard, Pierre 4554<br />

Blanchard, Raymond K<br />

e21011<br />

Blanchard, Rita A. e21076<br />

Blanchard, Suzette 1070<br />

Blanchard, Veronique 5047<br />

Blanchet, Benoit e13056<br />

Blanchon, Francois 1574<br />

Blanco Codeisido, Montse<br />

e14058<br />

Blanco, Esperanza e11074<br />

Blanco, Monsterrat 3022<br />

Blanco, Remei 1531, e18053<br />

Blanco-Calvo, Moisés e14595,<br />

e21002<br />

Blanco-Codesido, Montserrat<br />

3030<br />

Blandino, Giovanni 3521<br />

Blaney, Susan 9500, 9540,<br />

9541, 9581<br />

Blank, Christian U. 4611,<br />

LBA8500^, 8511, 8517<br />

Blank, Sima e14583, e14656<br />

Blank, Stephanie V. 5016,<br />

5054<br />

Blanke, C. D. 4010, 4019,<br />

4113, 10088<br />

Blasco, Ana 7058, 7060,<br />

10594, e12012<br />

Blaszkowsky, Lawrence Scott<br />

4021, 4112, e14615<br />

Blau, Gary E e18555<br />

Blau, Igor Wolfgang e18552<br />

Blay, Jean-Yves 1567, 1568,<br />

10005, 10006, 10007,<br />

LBA10008, 10009, 10010,<br />

10013, 10014, 10020,<br />

10027, 10035, 10049,<br />

10056, 10062, 10067,<br />

10078, 10089, 10092,<br />

10095, TPS10101, 10562,<br />

e13519, e13600^<br />

Blaydorn, Lisa e19048^<br />

Blazar, Bruce R. 6539<br />

Blazeby, Jane M TPS4143<br />

Blazer, Dan G. e14724<br />

Bleckmann, Annalen 3600,<br />

e14167<br />

Bleeg, Mary Lou e19588<br />

Bleicher, Jamie F. e21048<br />

Bleicher, Richard J. 6061<br />

Bleif, Martin 5512<br />

Bleijenberg, Gijs 9070<br />

Blenkinsop, C TPS1144<br />

Blesius, Aurore 4554, 10007,<br />

10033, 10044<br />

Blettner, Maria 1082,<br />

e11040^<br />

Bleuse, jean-Pierre 3633<br />

Blevins, Richard 1040, 5523,<br />

e13571<br />

Blieden, Clifford 595<br />

Bligny, Dominique e15131<br />

Blin, Nicolas 8009<br />

Blinder, Victoria Susana 6073<br />

Blinman, Prunella Louise<br />

e16501<br />

BLISS, R e13522<br />

Bliss, Robin L. e16502<br />

Blobe, Gerard C. 2585,<br />

10563<br />

Block, Andreas e13095^<br />

Block, Anne Marie W. 6565<br />

Block, Matthew Stephen<br />

5076<br />

Block, Norman L. e15153<br />

Block, Susan 9139<br />

Bloecher, Andrew 3587<br />

Blohmer, Jens U. 541, 1023<br />

Bloma, Marianne TPS4694<br />

Blondin, Beth A e21111<br />

Blons, Helene 2537^, 5554,<br />

10507<br />

Bloom, Gregory C 10048<br />

Bloom, Kenneth J. 516<br />

Bloomfield, Clara D. 6526<br />

Bloomston, Mark 4126,<br />

e14542, e14551<br />

Blough, David K 6007,<br />

e14068, e16524<br />

Blue, Michael e21066<br />

Bluemn, Eric Gregory 4669<br />

Blum, David e19648<br />

Blum, Joanne Lorraine 547<br />

Blum, Kristie A. 6507, 6508,<br />

8000, e18508<br />

Blum, Mariela A. 4069,<br />

4078, 4086, e14547,<br />

e14669<br />

Blum, Ronald H. 6114<br />

Blum, William G. 2533, 6526<br />

Blumberg, Joelle TPS4153<br />

Blumenschein, George R.<br />

2567, 2568, 3023, 5592,<br />

e13048<br />

Blumenthal, Susan e21022<br />

Blumetti, Jennifer e14590<br />

Bluth, Mark J. 8549, 8598<br />

Boachie-Adjei, Kwadwo<br />

e11057<br />

Boame, Nana e14154<br />

Bobadilla, Luz 8591<br />

Bobba, Ravi Kiran 1586,<br />

6561<br />

Bobek, Vladimir e21051<br />

Bobek-Billewicz, Barbara<br />

e21049<br />

Bobiak, Sarah 4126, e14542,<br />

e14551<br />

Bobos, Mattheos 573, 592<br />

Boc, Marko e14083<br />

Boccardi, Lidia 645<br />

Boccardo, Francesco e15185<br />

Bocchia, Monica TPS6640<br />

Bocci, Guido 3518, e11061<br />

Boccia, Ralph V. 6518^,<br />

8098, 10530, e18558,<br />

e21023<br />

Boccone, Paola e15192,<br />

e20512<br />

Bochicchio, Anna Maria<br />

LBA4001<br />

Bochner, Bernard 4527,<br />

4581^<br />

Bociek, Gregory 8047<br />

Bock, Meredith 9107<br />

Bockorny, Bruno 6555,<br />

e18543<br />

Boddy, Alan V. 9542<br />

Bodenner, Donald 5518,<br />

5591<br />

Bodmann, Joanna e14611,<br />

e14665<br />

Bodmer, Alexandre 9059,<br />

e11048<br />

Bodnar, Carolyn 6077<br />

Bodoky, Gyorgy 4042<br />

Bodrogi, Istvan TPS4683<br />

Bodurka, Diane C. 5012,<br />

9134<br />

Bodurtha, Joann 1563<br />

Body, Jean-Jacques e19514<br />

Boeck, Stefan Hubert 4023,<br />

4041, 4072<br />

Boegemann, Martin 4590<br />

Boehler, Geir 4066<br />

Boehlke, Brittani e21022<br />

Boehm, Andreas e16016<br />

Boehm, Daniel 10551<br />

Boehme, Michael 624<br />

Boehrer, Simone 6523<br />

Boelke, Edwin e11556,<br />

e14527, e16033<br />

Boellaard, Ronald 7603<br />

Boer, Hink 4528, e15035<br />

Boerman, Otto C. 3035,<br />

e13111<br />

Boers-Sonderen, Marye 2593,<br />

5061<br />

Boezen, H. Marike 1502<br />

Bogaerts, Jan 1022, 10602<br />

Bogale, Solomon Desalegne<br />

580<br />

Bogani, Paola 9005<br />

Bogart, Jeffrey e16047<br />

Bogatch, Kita 8575<br />

Bogenrieder, Thomas 3092<br />

Boggio, Gaston Federico<br />

10074<br />

Boggio, Lisa N e16503<br />

Boglione, Antonella 10055<br />

Bogner, Paul e17535<br />

Bogner, Wolfgang 10570<br />

Bohanes, Pierre Oliver 3584,<br />

3597, 3604, 3615, 3622,<br />

4026, 4088, e11018,<br />

e14031, e14034, e14052<br />

Bohnen, Daniela TPS4138<br />

Bohr, David 8598<br />

Boice, John D. CRA9513<br />

Boiche, Julie 9074<br />

Boige, Valerie 3547,<br />

LBA4003<br />

Boileau, Jean-Francois 616<br />

Boise, Lawrence 8100, 8101<br />

Boisselier, Blandine 2023<br />

Boitier, Francoise e21014,<br />

e21046<br />

Bojar, Hans e11556<br />

Bojic, Marija e15090<br />

Bok, Robert A. 4660<br />

Bokemeyer, Carsten 3539,<br />

3562, 3574, 3591, 4095^,<br />

4597, 9114, e13097^,<br />

e13108^, e15026, e15168<br />

Boku, Narikazu 4002, 4031,<br />

4035, 4046, 4082,<br />

e14510<br />

Boland, Amy L 4531,<br />

TPS4681<br />

Boland, William e18110<br />

Boles-Ponto, Laura e15143<br />

Boleti, Ekaterini 4611<br />

Bolgeri, Marco e15100<br />

Bolina, Marina Borges<br />

e13008<br />

Bollag, David T. LBA5002^,<br />

5017^<br />

Bollag, Gideon 8503^<br />

Bolli, Geremia 1509, 1510<br />

Bollig-Fischer, Aliccia 7063<br />

Bolling, Joan e18550<br />

Bologna, Serge 6633, 8002<br />

Bolondi, Luigi 3061,<br />

TPS4151<br />

Bolt-de Vries, Joan 10504<br />

Bolten, Wolfgang W. e11040^<br />

Bolton, Damien M e15117<br />

Bolton, Susan 1560<br />

Bolwell, Brian James 8055<br />

Bomalaski, John S. e17558<br />

Boman, Bruce M. TPS1615<br />

Bomba, Darrin e13077<br />

Bombard, Yvonne e11000<br />

Bombí, Josep Antoni 3536<br />

Bompas, Emmanuelle 10010,<br />

10014, 10020, 10056,<br />

10092, 10095<br />

Bomprezzi, Chiara 2037<br />

Bona, Eleonora 629<br />

Bona, Francesco e20512<br />

Bonamino, Martin H e14101<br />

Bonanni, Bernardo 519, 1500<br />

Bonanno, Laura e18130<br />

Bonapace, Ian Marc 4661<br />

Bonaventura, Antonino 3572,<br />

e14166, e15152<br />

Bonavida, Benjamin e18549<br />

Bond, Mary 9547, e18522<br />

Bondar, Vladimir e13095^,<br />

e13097^, e13108^<br />

Bondarde, Shailesh Arjun<br />

2592, e13127<br />

Bondarenko, Igor N. 4501,<br />

7558, e15037<br />

Bondarenko, Igor 7551,<br />

8512, 9125, e19587<br />

Bondra, Kathryn 9544<br />

Bonebrake, Albert J. 5015<br />

Bonehill, Aude 2507<br />

Bonekamp, Susanne 4114<br />

Bonetti, Andrea 5513<br />

Bonfill, Teresa TPS4674<br />

Bonfils, Claire 3039<br />

Bong, Chin Wei 4106<br />

Bongaerts, A. H.H. 10581<br />

Bongero, Danielle e13569<br />

Bongu, Navneeth Rao 8047<br />

Boni, Corrado LBA5003<br />

Boni, Ermenegilda e11546<br />

Boni, Luca e18074<br />

Boni, Valentina 3005, 10512,<br />

10595<br />

Bonifacio, Cristina 2580<br />

Bonini, Chiara 6541<br />

Boniol, Mathieu 1509, 1510<br />

Bonizzoni, Erminio e19612<br />

Bonnefoi, Herve R. TPS667,<br />

1011<br />

Bonner, James A. 621<br />

Bonner, William 2553, 3031<br />

Bonner-Ferraby, Phoebe 8566<br />

Bonnet, Fabrice 8046<br />

Bonnet, Isabelle 1580<br />

Bonnet, Nathalie 8555<br />

Bonnetain, Franck TPS667,<br />

1584, LBA3500^, 3506,<br />

4018, 4053, 4091,<br />

e18005<br />

Bonneterre, Jacques 2070,<br />

e11012, e11049, e11070,<br />

e11500, e12506<br />

Bonneviale, Paul 10076<br />

Bonnin, Nathalie e15009<br />

Bono, Petri 507, CRA4502<br />

Bonomi, Maria 630<br />

Bonomi, Philip D. 7067,<br />

e18117, e21069<br />

Bonomi, Philip LBA4<br />

Bonomo, Guido e14607<br />

Bononi, Antonio e13019<br />

Bonotto, Marta 1044,<br />

e13058, e13070<br />

Bonsing, Bert A 4053<br />

Bonta, Dacian e21076<br />

Bonta, Ioana e21076<br />

Bontempo, Kelly M. 9584<br />

Bonthapally, Vijayveer 570,<br />

e12515, e16571<br />

Bonura, Salvatore 586<br />

Bonvalot, Sylvie 10016,<br />

10017, 10044, 10096<br />

Bonzano, Alessandro e15073<br />

702s Numerals refer to abstract number


Bookman, Michael A. 5041,<br />

5056, 5057<br />

Boolbol, Susan K. e11057<br />

Boominathan, Rengasamy<br />

10533<br />

Boone, Jonathan David 5060<br />

Boonstra, Philip S e12031<br />

Boonyaphiphat, Pleumjit<br />

e14639<br />

Boorjian, Stephen A. 4526,<br />

4657<br />

Booser, Daniel J. 515, 527,<br />

10613<br />

Booth, Christopher M. 1583,<br />

3632, e14003, e15177^<br />

Boraas, Marcia 6061<br />

Borad, Mitesh J. 2579,<br />

e13052<br />

Borah, Bijan J 5004<br />

Boral, Anthony 3007, 9562<br />

Borbath, Ivan 4006, 4118<br />

Borchiellini, Delphine<br />

e16044, e16055<br />

Borczuk, Alain C. e13067<br />

Bordeaux, Jennifer 573<br />

Bordignon, Claudio 2580,<br />

5059, 6541, 7076, 7085,<br />

7581<br />

Bordogna, Walter 10596<br />

Bordonaro, Roberto e14040,<br />

e15544, e18026, e18096<br />

Bordonaro, Sebastiano<br />

e18002<br />

Bordoni, Rodolfo TPS7619<br />

Boren, Todd Patrtick e15501<br />

Borg, Martin e17544<br />

Borgato, Lucia 5078, e12037<br />

Borger, Darrell R. 4021,<br />

5029, 6606, 8522,<br />

e14615<br />

Borges, Eric 3092<br />

Borges, Virginia F. 9100<br />

Borget, Isabelle 5569, 9560,<br />

e16560<br />

Borghaei, Hossein 2595,<br />

e13606<br />

Borgia, Jeffrey Allen 7067,<br />

e21069<br />

Borgia, Jeffrey A e18117<br />

Borgida, Ayelet 4058<br />

Borker, Rohit e16573,<br />

e20510<br />

Bornemann, Antje 8526<br />

Borner, Markus 9059<br />

Borowski, Laurel e16532<br />

Borquez-Ojeda, Oriana<br />

TPS2619, TPS4700<br />

Borra, Anna 8077<br />

Borreani, Claudia 9120<br />

Borrello, Ivan e18561<br />

Borriello, Maria Rosaria 9098<br />

Borsatti, Eugenio 8046<br />

Borsu, Laetitia 10524<br />

Bortesi, Beatrice e14042<br />

Borthakur, Gautam 6528,<br />

6558, 6585, 6587, 6589,<br />

6592, 6597, 6603,<br />

TPS6635<br />

Bortolin, Michela e19595<br />

Bortoluzzi, Stephanie 8528<br />

Boruban, Melih Cem e11008<br />

Boruta, David M. 5029,<br />

e15545, e15568<br />

Borzelleca, Joseph 1563<br />

Bos, Marc Christiaan Allardt<br />

1533, 3044, TPS3115,<br />

7603, 10526, CRA10529<br />

Bos, Monique M.E.M. 4539<br />

Bosa, Chiara 2075<br />

Numerals refer to abstract number<br />

Bosch, Ana 1074<br />

Bosch, Joaquim 7522,<br />

e18137, e18143<br />

Bosco, Jaclyn Lee Fong 9008<br />

Bose, Ron 503<br />

Bosh<strong>of</strong>f, Chris 7538, 7562<br />

Bosl, George J. 4532<br />

Bosly, Andre 8021<br />

Bosman, Fred 3509, 3511,<br />

3575<br />

Bosomworth, Mike e13083<br />

Bosquee, Lionel LBA7000,<br />

7013, 7544<br />

Bosserman, Linda D. 1018,<br />

e11562, e13003<br />

Bosset, Daphne 8540<br />

Bossevot, Rachel TPS9154<br />

Bossi, Alberto 4554,<br />

TPS4689, TPS4691<br />

Bossi, Paolo 5555<br />

Bosson, Jean-Luc 1580<br />

Bossou, Ayoko R 5544<br />

Bossuyt, Veerle 1045, 10508,<br />

10558<br />

Bostrom, Bruce C. 9546<br />

Boswell, Erica 8043<br />

Bote, Josiah T. 3594<br />

Botero, Maria Luisa 10596<br />

Bott, Matthew Joseph 1541<br />

Botta, Cirino e13098,<br />

e14577<br />

Botta, Mario 7082, 7084<br />

Botteman, Marc 553, e15189<br />

Botteri, Edoardo 1049, 1115,<br />

e19036<br />

Botti, Gerardo 8581<br />

Bottini, Alberto 579, 1059,<br />

e11546<br />

Botto, Henry e15145<br />

Bottomley, Andrew 6002,<br />

6090, 7607<br />

Bottomley, David LBA4512,<br />

4551<br />

Bouabdallah, Kamal 8026,<br />

TPS8118<br />

Bouabdallah, Reda TPS1613,<br />

e18526<br />

Bouattour, Mohamed 2557,<br />

e14553, e14660<br />

Boucard, Celine 2060<br />

Bouchahda, Mohamed 3547<br />

Bouchal, Jan 1087<br />

Bouchalova, Katerina 1087<br />

Bouche, Olivier 3502, 3507,<br />

LBA4003, 10007, 10078,<br />

10089, TPS10102^,<br />

e14059<br />

Boucher, Eveline 4032,<br />

10078, e14148<br />

Boucher, Jean e19637<br />

Boucher, Kenneth M. 4525,<br />

e14177<br />

Boucher, Yves 1026<br />

Bouda, Damien 3063,<br />

e18071<br />

Boudahna, Lamie e11516<br />

Boudou-Rouquette, Pascaline<br />

10042, e13056<br />

Boue, Francois 8005<br />

Bouffet, Eric 9519<br />

Bouganim, Nathaniel 10620<br />

Boughan, Kirsten Marie 6590<br />

Boughey, Judy Caroline 1107<br />

Bougie, Olga 5032<br />

Bougnoux, Philippe TPS646<br />

Bouleuc, Carole e19623<br />

Boumedien, Feriel e13027<br />

Bounous, Valentina Elisabetta<br />

627<br />

Bouquier, Julie 5098<br />

Bourahla, Khalil TPS646<br />

Bourgeois, Hugues Pierre<br />

5018<br />

Bourgier, Céline 3063<br />

Bourgouin, Patrick P e17507<br />

Bourhis, Jean TPS5599<br />

Bourne Branchu, Veronique<br />

10006<br />

Bouros, Demosthenes e17553<br />

Bourquelot, Priscille M.<br />

e18572^<br />

Bourre, Ludovic e15161<br />

Bourredjem, Abderrahmane<br />

3507, 7007<br />

Boursi, Ben 1507, 1564,<br />

4619, e15058<br />

Boussion, Helene 6110<br />

Boussioutas, Alex TPS4141<br />

Bouteille, Catherine e21056<br />

Boutros, Paul C e17539<br />

Boutsicaris, Christina e15585<br />

Bouvet, Michael 2044,<br />

10072, e14014<br />

Bouyahia, Nezar e11502,<br />

e11516, e14725, e14726<br />

Bover, Isabel 5031, 5068,<br />

7095, e12012, e15575<br />

Bowen, Deborah 1563,<br />

e13517<br />

Bowen, Kristina E. 571<br />

Bower, Mark 8046, e14574<br />

Bowes, Barbara e15118<br />

Bowles, Daniel W. 3017<br />

Bowling, Mark e17547,<br />

e17550<br />

Bowman, Doug 3077<br />

Bowman, Jill 2500^<br />

Bowman, Kevin 2500^<br />

Bowman, Lee 1068, 7075,<br />

e18138<br />

Bowser, Jessica e13516<br />

Bowtell, David 5073<br />

Bowyer, Samantha Elizabeth<br />

e16004<br />

Boy, Davide 1073<br />

Boy, Sandra 7105<br />

Boyar, Michelle S. 4057<br />

Boychak, Oleksandr<br />

Volodymyrovych e14687,<br />

e19059<br />

Boyd, Kevin 1535, 7586,<br />

8015<br />

Boyd, Leslie R. 5016,<br />

e15548<br />

Boyd, Thomas E. 3069,<br />

6518^, e18047<br />

Boye, Mark e18138<br />

Boyer, Michael J. 7079^,<br />

LBA7500, 7511, TPS7615,<br />

e19642<br />

Boyer-Chammard, Agnes<br />

TPS1613<br />

Boyette-Davis, Jessica A.<br />

9082<br />

Boyiadzis, Michael 6563<br />

Boyko-Fabian, Mariya e16018<br />

Boyle, Frances M. TPS1136<br />

Boyle, Frances 596, 604<br />

Boyle, Helen Jane 4614,<br />

e15009<br />

Boyle, Peter 1509, 1510<br />

Boyle, Terence 1543, e12038<br />

Bozcuk, Hakan Sat e11008,<br />

e14070<br />

Bozec Le Moal, Laurence<br />

5505<br />

Bozec, Alexandre 5521,<br />

e16044, e16055<br />

Bozkurt, Mustafa e14698<br />

Bozzarelli, Silvia e14672<br />

Bracarda, Sergio 4541, 4627,<br />

e15112, e15185<br />

Brach del Prever, Elena Maria<br />

10055<br />

Brachman, David 2008b,<br />

2047<br />

Brachtel, Elena F 10588<br />

Brack, Simon 2575<br />

Bradbear, Joseph e19598<br />

Bradbury, Angela R. 6095,<br />

6128<br />

Bradbury, Penelope Ann<br />

7093, 7511, e18079<br />

Brade, Anthony M. 2013<br />

Bradford, Carol Rossier<br />

e16014<br />

Bradford, Chad e13581<br />

Bradford, Leslie e15545,<br />

e15568<br />

Bradley, Cori e13584<br />

Bradley, Victoria e14574<br />

Bradley-Garelik, M. Brigid<br />

6504, TPS9594<br />

Brady, Anna K. 7595<br />

Brady, Claire e11569,<br />

e15078<br />

Brady, John C e21038<br />

Brady, Mary Susan 8583<br />

Brady, Rachel 4125<br />

Braghiroli, Maria Ignez Freitas<br />

Melro e12513, e14702<br />

Brahmbhatt, Himanshu<br />

e13054<br />

Brahmer, Julie R. CRA2509,<br />

2510, TPS2615, 7509,<br />

7531, 7541<br />

Brahmi, Sami Aziz e11502,<br />

e11516, e14725, e14726<br />

Braicu, Elena Ioana 5070<br />

Braicu, Elena Ioana 5047,<br />

5080<br />

Braicu, Ioana 5034, 5053,<br />

5058, 5071, e15520,<br />

e15531<br />

Braik, Tareq 8085, 9085,<br />

e18534<br />

Brain, Etienne TPS662,<br />

TPS667<br />

Braiteh, Fadi S. 3023, 3069,<br />

TPS3111<br />

Brakchi, Zahir 3052<br />

Brakemeier, Susan 4632<br />

Braly, Patricia S. 5012<br />

Brambilla, Christian 7529<br />

Brambilla, Elisabeth 7007,<br />

7529<br />

Brames, Mary J. 4534, 4598<br />

Bramhall, Simon R e14625<br />

Brammer, Melissa G. 1037,<br />

e11076, e19633<br />

Brammer, Melissa e11063,<br />

e11077<br />

Bramuglia, Guillermo Federico<br />

e18114<br />

Bramuglia, Guillermo e14109<br />

Brana, Irene 2042, 3014,<br />

3027, 3059, e13001<br />

Brand, Andrew e21091<br />

Brand, Lana 1608<br />

Brandao, Guilherme e18061<br />

Brandeis, Alison Emily<br />

e15576<br />

Brandenburg, Nancy A.<br />

e18559<br />

Brandes, Alba Ariela 2, 2057,<br />

2061, TPS2104, 7550<br />

703s


Brandes, Johann Christoph<br />

7048, 10625<br />

Brandi, Mario e21113,<br />

e21134<br />

Brandt, Amanda C. 1513,<br />

1518, 1520, 1546<br />

Brandt, Mark 6528<br />

Branham, Donna 5015<br />

Brannigan, Robert E 2532<br />

Branscum, Adam J. e21000<br />

Brar, Amarpali e15097<br />

Brase, Jan C. 542<br />

Brasnu, Daniel 5554<br />

Brastianos, Priscilla Kaliopi<br />

2020<br />

Brat, Daniel 3055<br />

Bratslavsky, Gennady<br />

TPS4680<br />

Bratthall, Charlotte e14531<br />

Braun, Ada H. TPS670,<br />

e16528<br />

Braun, Christian 2000<br />

Braun, Denis TPS7111<br />

Braun, Donald Peter 5049,<br />

e14012<br />

Braun, Eduardo 572, e21069<br />

Braun, Jonathan 1052<br />

Brautigan, David L 8530<br />

Bravaccini, Sara 10601<br />

Braverman, Albert S. e18504<br />

Bray, Emma 10550<br />

Braylan, Raul 8088<br />

Braziel, Rita M 8001<br />

Brazil, Kevin 6035<br />

Breathnach, Oscar S. e19059<br />

Breda, Enrico LBA5003<br />

Bredel, Markus 2001, 2035<br />

Bredella, Miriam A. 6136<br />

Breder, Valeriy Vladimirovich<br />

e14723<br />

Breen, Laura e15126<br />

Breen, Stephen TPS5600<br />

Breen, Timothy 4586<br />

Brega, Nicoletta 2610<br />

Brehar, Oana 8057<br />

Breitbach, Caroline TPS4152,<br />

e13044, e14566<br />

Breithaupt, Kirstin TPS4138<br />

Brekken, Rolf A. e13563<br />

Bremnes, Roy M. 7027<br />

Breneman, John C. 9509<br />

Brenin, Christiana 3047<br />

Brennan, Cameron W 2028<br />

Brennan, Michael 9024<br />

Brennan, Murray F. 10001,<br />

10015<br />

Brennan, Rachel Christine<br />

e13544, e13584<br />

Brennan, Tina 3533<br />

Brenner, Adrienne A 3528<br />

Brenner, Andrew Jacob<br />

TPS10099<br />

Brenner, Arnold e14704,<br />

e18574<br />

Brenner, Baruch e14067,<br />

e14571<br />

Brenner, David J. e15162,<br />

e17562<br />

Brenner, Dean E. e18118<br />

Brenner, Malcolm K. 2558<br />

Brentjens, Renier J. 2586,<br />

TPS2619, 6613<br />

Breslin, Susan 617<br />

Breslin, Tara M. 1125<br />

Bressan, Alexsander Kurowa<br />

10069, e14119<br />

Bressel, Mathias 3629<br />

Bressler, Linda R. 2073<br />

Bretcha, Pere 3064<br />

Breuer, F. e13100^<br />

Breunig, Ian Michael e14522,<br />

e14626, e14638<br />

Brew, Christine Taylor<br />

e13577<br />

Brewer, Cathy J. 2018<br />

Brewer, Janice e12507<br />

Brewer, Jerry D 6571<br />

Brewer, Katherine e15046<br />

Brewer, Molly e15563<br />

Brewer, Takae 1047<br />

Brewster, Michael 6591<br />

Brewster, Wendy R. 2591,<br />

5050, e13063<br />

Brezden, Christine B. 9111<br />

Bria, Emilio 630, 4076,<br />

4541, TPS7109, e14661<br />

Briani, Chiara 9090<br />

Briasoulis, Evangelos C.<br />

e18567<br />

Brice, Pauline 8002, 8027<br />

Brichard, Vincent G. 7013,<br />

7056<br />

Brichkova, O. e11528<br />

Brickhouse, Alise 2027,<br />

2074^<br />

Bridge, Julia Ann 9535<br />

Bridgewater, John A.<br />

TPS3637<br />

Brienza, Silvano 3026<br />

Briere, Josette 8048<br />

Briggs, Kathryn Jane<br />

TPS4140<br />

Brighenti, Matteo 4097,<br />

e19612<br />

Bright, Mary Anne 6086<br />

Briley, Linda P. 10532<br />

Brill, Yolanda e16035<br />

Brilliant, Kate E. e21116<br />

Brindle, James 5102<br />

Brinkman, Tara M. 9531<br />

Brisbin, Lori e15209<br />

Briscoe, Karen P. e19642<br />

Brissy, Sophie 8555<br />

Britan, Laura e15187<br />

Brites, Patricia Chaves<br />

e14597<br />

Brito, Rose- Marie 9563,<br />

9570<br />

Britten, Carolyn D. 508, 3003<br />

Britton, David J. e21091<br />

Brixner, Diana I. 6583<br />

Broadbridge, Vy e19513<br />

Broaddus, Russell 1513,<br />

10510, e13516<br />

Brocard, Anabelle e19019<br />

Brocia, Christine 8557<br />

Brock, Malcolm 1573,<br />

10625, e17552, e18147<br />

Brockmann, Michael 1533,<br />

10526, CRA10529,<br />

e18000<br />

Brockmeyer, Norbert 8505<br />

Brockstein, Bruce 5500,<br />

10003, 10028<br />

Broder, Michael S. 3626,<br />

6057, e14610<br />

Brodeur, Garrett M. e20008<br />

Brodie, Seth 10625<br />

Brodin, Patrik Nils e18527<br />

Brodowicz, Thomas 3574<br />

Brodsky, Jeffrey 8577<br />

Brody, Ed e21012, e21142<br />

Brody, Joshua 2513<br />

Broggini, Massimo LBA7501<br />

Brogi, Edi 557<br />

Bromberg, Michael e16022<br />

Bron, Dominique 8048<br />

Brondfield, Samuel Craig<br />

e15137<br />

Brooks, Barry Don 6109<br />

Brooks, Gabriel 6003<br />

Brooks, James D 10513<br />

Brooks, Robert J. 547<br />

Brooks, Roserika 3081<br />

Brooks, Sandra Elane 1512<br />

Broom, Reuben James<br />

e15050, e15054<br />

Brose, Marcia S. 5508, 5547<br />

Brossoie, Susan M 5577<br />

Brostjan, Christine e14095,<br />

e14099<br />

Brouchet, Anne 10076<br />

Brough, Rachel 3050, 5035<br />

Brouste, Veronique 9012,<br />

10035, e14017<br />

Brouwers, Adrienne H. 10581<br />

Brouwers, Barbara 1020<br />

Brown, Andrew M. K. e13107<br />

Brown, Brita e13581<br />

Brown, Camille D. 9565<br />

Brown, Chris 7079^, e16501,<br />

e19642<br />

Brown, Christopher 5517<br />

Brown, Damien e21037<br />

Brown, Dennis e13123<br />

Brown, Eric e14642<br />

Brown, Erika N. 537<br />

Brown, Gina 3586, e14088,<br />

e14097, e14132<br />

Brown, Holly M. 3531, 3587<br />

Brown, Janet Elizabeth 4642,<br />

e16528, e19514<br />

Brown, Jennifer R. 6515^,<br />

8032<br />

Brown, Jennifer 3000<br />

Brown, Jillian e15012<br />

Brown, John V. e15500<br />

Brown, Jubilee 5027, 9134<br />

Brown, Laura E. 9095<br />

Brown, Martin e13580<br />

Brown, Michael Paul 8517<br />

Brown, Monica 4607<br />

Brown, Myles 576<br />

Brown, Patrick Andrew 9548<br />

Brown, Paul D. 2031, 8584<br />

Brown, Peter D. 3052, 6513,<br />

6596, 6604<br />

Brown, Robert S 4101<br />

Brown, Stephen CW e15194<br />

Brown, Tanya Renae 9529<br />

Brown, Tiffany 2052<br />

Browne, Brigid 617, 633<br />

Browne, Sarah K 7033<br />

Brozos Vazquez, Elena Maria<br />

e14733<br />

Bruce, Can 10558<br />

Bruce, Justine Yang 4549,<br />

4568, 4654<br />

Bruce, Richard 10574<br />

Bruchovsky, Nicholas e15201<br />

Bruckner, Howard e14729,<br />

e14732, e15576<br />

Brue, Thierry 8568<br />

Brueckl, Wolfgang M. e17536<br />

Bruegl, Amanda S. 1513<br />

Brueilly, Kevin E e17002<br />

Bruera, Eduardo 9002, 9023,<br />

9025, 9049, 9062, 9063,<br />

9065, 9069, 9096, 9127,<br />

e19528, e19584, e19599,<br />

e19603, e19613, e19643<br />

Bruera, Gemma e14010<br />

Brufsky, Adam LBA506,<br />

LBA1000, 1085, 1526,<br />

1589<br />

Brugarolas, A. 3064, e16000<br />

Brugarolas, James 4616,<br />

TPS4678, e15066<br />

Brugel Ribere, Lydia 5505<br />

Brugger, Wolfram 3<br />

Brugiatelli, Maura 8006,<br />

e18564<br />

Brugnara, Sonia e14711<br />

Brugnatelli, Silvia e13103<br />

Brugni, Manuela 9098<br />

Bruins, Harman Maxim 4588,<br />

e15060<br />

Brulard, Celine 9536<br />

Brummendorf, Tim H. 5516^,<br />

6511, 10552, e19610<br />

Bruna, Jordi 9090<br />

Brundage, Michael Donald<br />

4509<br />

Brune, Frederique 5069<br />

Brunelle, Serge e15155,<br />

e19539<br />

Brunelli, Alessandro e21070<br />

Brunello, Antonella 586,<br />

e13019<br />

Brunet, Joan 1588<br />

Brunet, Rene e14023<br />

Brunetto, Algemir Lunardi<br />

10593<br />

Brunetto, Andre 3030<br />

Bruneu, Yvane 8578<br />

Bruneval, Patrick 5554,<br />

e13578<br />

Brunner, Andrew Mark 6617,<br />

6618<br />

Bruno Da Costa, Ligia e14673<br />

Bruno, Margarita 1060,<br />

e11015, e18547<br />

Bruno, Onorina e14553<br />

Bruns, Rolf TPS4139<br />

Brunson, Ann 3566<br />

Brunsvig, Paal 7027<br />

Brusa, Federica e15192<br />

Brusquant, David LBA3500^,<br />

3506<br />

Brustugun, Odd Terje 7027<br />

Brutcher, Edith e14614,<br />

e14692<br />

Bruzzi, Paolo 1073<br />

Bryant, Donna M. 9047,<br />

e16532, e16561<br />

Bryant-Comstock, Lynda 6060<br />

Bryla, Christina 2553<br />

Brünner, Nils 3576<br />

Bshara, Wiam e11563<br />

Buadi, Francis 8010, 8035,<br />

8092, 8096, 8105,<br />

TPS8116<br />

Buanes, Trond e14637<br />

Bubenheim, Michael 8026<br />

Bubis, Jeffrey Alan e14514<br />

Bubis, Jeffrey TPS4696^<br />

Bubley, Glenn J. 4521<br />

Buchanan, Adam 1520<br />

Buchbinder, Aby 1017<br />

Bucheit, Amanda Dawn 8584<br />

Buchholz, Thomas A. 594,<br />

600, 1029, 1032, 1104,<br />

7062<br />

Buchler, Markus W e14625<br />

Buchner, Anton 9125,<br />

e19587<br />

Buchner, Thomas 6610<br />

Buchschacher, Gary L.<br />

e16031<br />

Buck, Martin LBA5000<br />

Buck, Steven e13089<br />

Buckley, Julliette M 1122<br />

Buckley, Sarah A e16052<br />

Buckner, Jan C. 2008b,<br />

2004, 2031, 2032, 6133<br />

704s Numerals refer to abstract number


Buclin, Thierry e11048<br />

Budach, Volker 5512,<br />

e16018, e16033<br />

Budach, Wilfried 5512,<br />

e11556, e14527, e16033<br />

Budakoglu, Burcin e11540<br />

Budd, G. Thomas 620,<br />

TPS655, TPS10103<br />

Buddavarapu, Kalyan 3630<br />

Budde, Klemens 4632,<br />

10619<br />

Budha, Nageshwar e13114<br />

Budihardjo-Welim, Henry<br />

7056<br />

Budinska, Eva 3511, 3575<br />

Budman, Daniel R. 1046<br />

Budrukkar, Ashwini<br />

Narsingrao e16021<br />

Buduhan, Gordon e14695<br />

Buehring, Hans-Joerg e15577<br />

Buekers, Thomas E. e15512<br />

Buelga, Paula e11517<br />

Buening, Barbara 5082^<br />

Buerger, Horst 10627<br />

Buerki, Christine 4565<br />

Buettner, Reinhard 1533,<br />

10526, CRA10529<br />

Bufalino, Rosaria TPS9155<br />

Buffoli, Alberto 5513<br />

Buffon, Rosana e19036<br />

Bugat, Roland e19640<br />

Bugdahn, Tim e18565<br />

Buges, Cristina 3093, 7522,<br />

7540, 7542, 9129,<br />

e18137<br />

Buglioni, Simonetta 1050<br />

Bui Nguyen, Binh 9536,<br />

10005, 10006, 10007,<br />

10009, 10010, 10014,<br />

10020, 10023, 10027,<br />

10033, 10035, 10056,<br />

10057, 10067, 10089,<br />

10092, 10095, TPS10101,<br />

TPS10102^, e13600^<br />

Bui, Binh 10062<br />

Bui, Marilyn M 10048,<br />

10070<br />

Bui, Son 5543<br />

Buisan, Oscar 4579<br />

Bujosa, Juan Ramon e12000<br />

Bulanov, Anatoly e15025<br />

Bulgrin, Angela e16549<br />

Bull, Christian R F 8563<br />

Bull, Janet 9002<br />

Buller, Harry e13062<br />

Bulley, Sue 7562<br />

Bulone, Linda 9004<br />

Bulotta, Alessandra 7076<br />

Bumb, Caroline 1048<br />

Bumgardner, William Mark<br />

3005<br />

Bunge, S<strong>of</strong>ia 9063<br />

Bunn, Paul A. 7526, 7532,<br />

7589, e18077<br />

Bunnell, Craig A. e16515,<br />

e16559<br />

Bunskoek, Sophie 4528<br />

Bunworasate, Udomsak<br />

6509^<br />

Buonamici, Silvia 9562<br />

Buosi Silva, Thiago e19528,<br />

e19584<br />

Bupathi, Manojkumar 6521<br />

Buque, Aitziber e17522,<br />

e21009<br />

Burandt, E C 10501<br />

Burattini, Luciano e15074<br />

Burch, Patrick A. e14594<br />

Numerals refer to abstract number<br />

Burel-Vandenbos, Fanny<br />

e12502<br />

Burg, Kenda CRA6009<br />

Burger, Jan Andreas 6507,<br />

6515^, 6517, 6558, 6598,<br />

TPS6635<br />

Burger, Renate e18565<br />

Burgett, Monica 2014<br />

Burgio, Marco Angelo e17546<br />

Burgio, Salvatore Luca<br />

e17546<br />

Burgos, Emilio 3618, 4089,<br />

10547<br />

Burgues, Octavio 1015,<br />

10500, 10616<br />

Burhenne, Juergen 10041^<br />

Burillon, Jean-Philippe<br />

e13036<br />

Burke, James M. TPS4152,<br />

e13044<br />

Burke, James 4593, e14566<br />

Burke, John M. 3069,<br />

e18522<br />

Burke, Joseph Malachy 7526<br />

Burke, Lillian P. e13012<br />

Burke, Meghan 6606<br />

Burke, Rachel e14172<br />

Burke, Thomas W. e16574<br />

Burke, Wendy 3004, 3048,<br />

3051<br />

Burke, William M. 6078<br />

Burkey, Brian 5573<br />

Burkhard, Roger 10033<br />

Burkhardt, Carmen 8059,<br />

e18532<br />

Burkholder, Iris 3541<br />

Burma, Sandeep 10624<br />

Burmeister, Bryan TPS4145<br />

Burmeister, Lynn A. 5591<br />

Burmester, James e14008<br />

Burn, Timothy 6514^<br />

Burnett, Bruce K. 2585<br />

Burnett, Omer L. e15502<br />

Burnett, Terrilea 3567<br />

Burnight, Timothy 9029<br />

Burningham, Zachary 9525<br />

Burns, Karen Cristly e20007<br />

Burns, Kathleen H. 6629<br />

Burns, Lisa 1559<br />

Burns, Matthew 7107, 7108<br />

Burr, Laura 3057<br />

Burrafato, Giovanni e15544<br />

Burri, Ryan J. e15162<br />

Burris, Howard A. 508, 512,<br />

535, 539, 540, 551, 559,<br />

618, TPS648, 1018, 1086,<br />

TPS2621, 3008, 3023,<br />

3033, 3041, 3067, 3097,<br />

3102, TPS3118, 7035,<br />

7100, 7567, 7587, 8510,<br />

e13076, e13077<br />

Burris, Howard 3062<br />

Burris, Patricia 6108<br />

Burrows, Francis 6577<br />

Burrows, Jon e21068<br />

Burstein, Harold J. 6089,<br />

e16515<br />

Burt, Michael e14632<br />

Burt, Paul e18095<br />

Burtness, Barbara 5566,<br />

5576, TPS5599, e13504<br />

Burton, Allen W, e19578<br />

Burton, Elizabeth M 6587<br />

Burton, Eric Christopher<br />

e17019<br />

Burton, Gary Von e11541<br />

Burton, Jill K. 5544<br />

Burton, Jon N 5564<br />

Bury, Martin J. 6054, 8521<br />

Bury, Martin Joseph 1025,<br />

7517, 9113<br />

Burzawa, Jennifer K. e13516<br />

Burzykowski, Tomasz<br />

TPS7610<br />

Busam, Klaus J. 8586<br />

Busby, Leslie T. 6109<br />

Buscariollo, Daniela L. 6019<br />

Busch, Steffi e19540<br />

Bush, Nigel 9008<br />

Bushmakin, Andrew G. 6092,<br />

e16530<br />

Bushnell, David L e14600<br />

Bushunow, Peter Walter<br />

e17535<br />

Buso, Marco Murilo e19504<br />

Busowski, Mary T e16030<br />

Busque, Lambert 6605<br />

Busse, Paul M. 5579, 5594,<br />

e16059<br />

Bussink, Jan e13111<br />

Bustami, Rami e19057<br />

Busteros, Jose Ignacio 4089<br />

Buta, Eugenia 5532<br />

Butaney, Mohit 7547, 7588,<br />

10592<br />

Butcher, Eugene e21071<br />

Butera, Alfredo e18002<br />

Buti, Sebastiano e15160<br />

Butler, Rachel TPS10633^<br />

Butler, Robert W. 2040<br />

Butler, Steven M 1005<br />

Butori, Catherine 7591<br />

Butow, Phyllis Noemi 6111,<br />

e16507<br />

Butowski, Nicholas A. 2026<br />

Butrynski, James E. 1547<br />

Butt, Nadeem e14703<br />

Butterfass-Bahloul, Trude<br />

10081<br />

Butterfield, Lisa H. 8533<br />

Button, Anna M 8061,<br />

e14620, e15580<br />

Butts, Charles Andrew<br />

LBA7000<br />

Buturovic, Ljubomir J 10576<br />

Buxbaum, James e14679<br />

Buxh<strong>of</strong>er-Ausch, Veronika<br />

e14087<br />

Buxo, Maria 1588<br />

Buys, Saundra S. 1502, 1521<br />

Buyse, Marc E. 611, TPS648,<br />

1025, 3557, e13055<br />

Buyse, Marion e13056<br />

Buyukafsar, Kansu e15104<br />

Buyukberber, Suleyman 3565,<br />

e14526, e15047, e19024,<br />

e20514<br />

Buyukunal, Evin 3565,<br />

e21143<br />

Buzaglo, Joanne S. 9048<br />

Buzdar, Aman 596, 604<br />

Buzoianu, Manuela e18073<br />

Bychkov, Mark B. e19012<br />

Büchler, Markus W. 4095^<br />

Bye, Tyler e13537<br />

Byer, Jennifer e14519<br />

Byers, Lauren Averett 7096,<br />

10612<br />

Bylicki, Olivier 7574<br />

Bylow, Kathryn A. 4603<br />

Byrd, John C. 6507, 6508,<br />

e13568, e18508<br />

Byrne, Clare 3613<br />

Byrne, Jennifer 6512, 8015<br />

Byrum, Stephanie D. e21039<br />

Byström, Styrbjörn 6557<br />

Bytautas, Jessica Peace<br />

e19616<br />

Büttner, Reinhard 3550,<br />

7603<br />

Böckenh<strong>of</strong>f, Annette 4628<br />

Böning, L. e14140<br />

C<br />

C. Roach, Emir e11509<br />

Caballero, Cristina 5068,<br />

e15575<br />

Caballero, Hector M e15527<br />

Caballero, Rosalia 10500,<br />

10616<br />

Cabanillas, Fernando 1060,<br />

8084, e11015, e18547<br />

Cabanillas, Maria E. 5518,<br />

Cabanne, Ana<br />

5547, 5591<br />

e14598<br />

Cabel, Luc 6110<br />

Cabeza Rodriguez, Maria<br />

Angeles e15033<br />

Cabibbo, Giuseppe TPS4151<br />

Cabrera, Antonio 1111<br />

Cabrera, Paula e15122<br />

Cabri, Jan 9033<br />

Cabrita, Fernanda e14673<br />

Cabuk, Devrim e14618,<br />

e16015<br />

Caceres, Jaime e12041<br />

Cachin, Florent TPS646<br />

Cadeddu, Christian 9006<br />

Cadeddu, Jeffrey 4616<br />

Cadenas, Christina 10551<br />

Cadiot, Guilaume 4071<br />

Cadoo, Karen Anne 1543<br />

Cadore, Anne Claire 10027<br />

Cadranel, Jacques 10507,<br />

e18075, e18089, e18102<br />

Cadron, Isabelle 5034, 5071<br />

Cafà , Ester Valentina 5093,<br />

e15547, e15551, e15553<br />

Caffo, Orazio 7550, e14711,<br />

e15160<br />

Caggiano, Vincent 1582,<br />

1606<br />

Caglio, Silvia 4122<br />

Cagney, Jennifer M. e18029<br />

Cagnon, Laurene e19050<br />

Cagossi, Katia e11071<br />

Cahana, Ayelet 559<br />

Cahill, Daniel P. 2019<br />

Cahova, Dana e14556<br />

Cai, Fan e13538<br />

Cai, Guoxiang 3551<br />

Cai, Hongyan 4055<br />

Cai, Jian-Chun e14629<br />

Cai, Jie 4576, 4588, 4589,<br />

e13531<br />

Cai, Ju-Fen e17512<br />

Cai, Ling 9103, e18121<br />

Cai, Ruigang e11025<br />

Cai, SanJun 3551<br />

Cai, Shirong 3047<br />

Cai, Wenli 6136<br />

Cai, Xiaohong e18033<br />

Cai, Xun e14508<br />

Cai, Xuwei e21115<br />

Caillot, Denis 8002, 8009<br />

Caimi, Paolo Fabrizio 8080<br />

Cain, Suzanne e19009<br />

Cainap, Calin 3532<br />

Caires, Inacelli Queiroz de<br />

Souza e15562<br />

Caires-Lima, Rafael e15562<br />

Cairncross, J. Gregory 2008b,<br />

TPS2104<br />

Cairo, Mitchell S. 6537,<br />

9501, e13022, e21150<br />

Cajal, Rosana e12000<br />

Cajfinger, Francis 1580<br />

705s


Cakir, Betul 9567<br />

Calabek, Bernadette 2071<br />

CALABRESE, BERNADETTE<br />

e12511<br />

Calabrese, Giuseppe e14042<br />

Calabrese, Pasquale 2040<br />

Calabro, Fabio e15003<br />

Calabuig, Silvia 10079<br />

Calatrava, Ana M 3618,<br />

10547<br />

Calcagnile, Selma e19532,<br />

e19533<br />

Calcedo, Maria Celia e12033<br />

Caldara, Alessia 586,<br />

e14711, e21102<br />

Caldas, Carlos 544, TPS1144<br />

Calderero, Veronica e15144<br />

Caldes, Trinidad e14147<br />

Caldito, Gloria e11541<br />

Caldwell, Imogen Rose<br />

e15050<br />

Caleffi, Maira 606, 1522<br />

Calemma, Rosa 8083<br />

Califano, Joseph A 5528<br />

Califf, Robert M 6047<br />

Caligiuri, Philip e13548<br />

Caliogna, Laura e13103<br />

Caliolo, Chiara 630<br />

Calise, Fulvio 4110<br />

Calistri, Daniele e18045<br />

Call, Jason Andrew e14555<br />

Call, Timothy e13517<br />

Callacondo, David e15036<br />

Callado, Rodrigo Barbosa<br />

e14183<br />

Callahan, Margaret 2521,<br />

8515, 8549, 8575<br />

Calleri, Angelica 2083<br />

Calles, Antonio 2567, 2568,<br />

3059, 3066, 3068,<br />

e13048, e18055<br />

Callies, Sophie 2554<br />

Callister, Matthew 6108<br />

Calmon, Raphael 9517<br />

Calogero, Raffaele A e13527<br />

Calvani, Nicola e11001,<br />

e11547<br />

Calvert, Paula 2609<br />

Calvetti, Lorenzo 3555,<br />

e14040<br />

Calvo, Alejandro R. e17506<br />

Calvo, Alfonso e15041<br />

Calvo, Begoña e17522,<br />

e21009<br />

Calvo, Elena Galve 10512,<br />

10595<br />

Calvo, Emiliano 3005, 3059,<br />

3066, e15111<br />

Calvo, Ezequiel 10583<br />

Calvo, Felipe A. e12003<br />

Calvo, Isabel 602, e11006,<br />

e11517<br />

Calvo, Katherine R. 8088,<br />

8104, e18568<br />

Calvo, Lourdes 1001, 10500,<br />

10616<br />

Calvo, Ovidio Fernández<br />

e15067, e15076, e15077<br />

Camacho, Ignacio e15171<br />

Camacho, Luis H. 2510,<br />

e13040<br />

Camacho, Mercedes 5590<br />

Camara, Oumar e15566<br />

Camargo, Veridiana Pires De<br />

e20502<br />

Cambois, Anne 3052<br />

Cambon, Alex e21144<br />

Camci, Celalettin 3565<br />

Camerini, Andrea e18074<br />

Cameron, David A. 502, 513,<br />

TPS660, TPS665,<br />

TPS1144, 6091<br />

Cameron, Silke e13039,<br />

e14707, e21128<br />

Camey, Suzi Alves 1522,<br />

10593<br />

Camidge, D Ross 7508, 7601<br />

Camidge, D. Ross 3007,<br />

7504, 7526, 7532, 7534,<br />

7589, 7596, e21120<br />

Caminiti, Caterina 9120<br />

Camitta, Bruce M. 9504<br />

Camoriano, John Kelly 8043,<br />

8053<br />

Camp, Aaron C 4108<br />

Camp, Melissa 1122<br />

Campagnolo, Marta 9090<br />

Campana, Davide e14647<br />

Campanella, Delia e20512<br />

Campanha, Daniel e14680<br />

Campassi, Cristina 10572<br />

Campbell, Alicyn TPS7615<br />

Campbell, Fiona e14625<br />

Campbell, Jenny 4061<br />

Campbell, Michelle M. 587<br />

Campbell, Nancy 9084<br />

Campbell, Olivia M. 10563<br />

Campbell, Steven C. e15095<br />

Campbell, Toby Christopher<br />

9039<br />

Campbell-Baird, Cynthia<br />

e14520<br />

Campbell-Hewson, Quentin<br />

9542<br />

Campelo, Rosario Garcia<br />

1531<br />

Campenni, Giuseppe e18065<br />

Campian, Jian Li e17552<br />

Campion, Loic e11551<br />

Campitelli, Maura e15535<br />

Campone, Mario 530, 532,<br />

540, 551, 559, 2082,<br />

3052<br />

Camporeale, Jayne 6093<br />

Campos Balea, Begoña<br />

e15067, e18040, e18146<br />

Campos, Angel 5520,<br />

e14147, e15094, e18069,<br />

e18106, e21015<br />

Campos, Arinilda Silva<br />

e16568<br />

Campos, Begoña e18070<br />

Campos, Conceicao Souza<br />

e15509<br />

Campos, France 636<br />

Campos, Marta Carmona<br />

e14733, e19505<br />

Campos, Tiffany 4125<br />

Campos-Parra, Alma Delia<br />

e18114<br />

Camps, Carlos 7058, 7060,<br />

10594, e18131, e18137,<br />

e18143<br />

Camuset, Juliette 1580<br />

Canadas, Carmen 5520,<br />

e15094, e18069, e18106,<br />

e21015<br />

Canal, Barbara 7585, e18104<br />

Canamares, Irene 4620<br />

Canar, Jeff 572<br />

Cancian, Maurizio e19592<br />

Candamio, Sonia 10534,<br />

e14733, e19505<br />

Candelaria, Myrna 6599<br />

Candeloro, Giampiero e11041<br />

Canela, Mercedes 7041<br />

Canessa, Pier Aldo e17533<br />

Canet, Ramon e11036<br />

Cañete, Adela TPS9596,<br />

10082<br />

Canfield, Angela C. e15179<br />

Cangiarella, Joan e11010<br />

Cangir, Ayten Kayi 1596<br />

Canil, Christina M. e15537<br />

Cannell, Amanda 10050<br />

Canney, Peter TPS665<br />

Cannita, Katia e14010<br />

Cannon, Donald Maurice<br />

e13537<br />

Cannon, George M. TPS9152,<br />

e13537<br />

Cano, Oscar D. 10628<br />

Cano, Stefan 9042, 9043<br />

Canon, Jean-Luc e14044,<br />

e14060, e19511<br />

Cañon, Rosa e16000<br />

Canonici, Alexandra 632,<br />

e13520<br />

Canter, Daniel e15006<br />

Canter, Robert J. e19013<br />

Cantin, Guy 8002<br />

Cantore, Maurizio 10611<br />

Cantos, Blanca e11035,<br />

e12012, e18521<br />

Cany, Laurent 9012, e14023<br />

Canzler, Ulrich 5005, 5031<br />

Cao, Gabriel e19569<br />

Cao, Junning 3038<br />

Cao, Liang 7033<br />

Cao, Lulu e21116<br />

Cao, Yabing e18517<br />

Cao, Yang 8042, 8106, 8107<br />

Cao, Yen Thi Kim Hong<br />

e13588<br />

Capaldi, Antonio e15133<br />

Capanu, Marinela 3577,<br />

4061<br />

Capasso, Immacolata e11052,<br />

e13539<br />

Capdevila, Enric 1120<br />

Capdevila, Jaume 3014,<br />

3571, 4119, 4122, 10546,<br />

TPS10632, e14088,<br />

e14671, e21021<br />

Capdevila, Laia 7540, 9129,<br />

e12504, e15001, e18131,<br />

e18143<br />

Capellades, Jaume 2045<br />

Capelletti, Marzia 7510<br />

Capelletto, Enrica e18103<br />

Capelli, Francesca e14094<br />

Capelli, Laura e18045<br />

Caplen, Natasha 9511<br />

Caplin, Martyn E 4123,<br />

TPS4153, e14696<br />

Caplin, Martyn 4121,<br />

e17543, e19581<br />

Capo, Adolfo Miguel e11087<br />

Capobianco, Gaetana 8066,<br />

8083<br />

Capozza, Scott TPS669<br />

Capozzi, Federica 10066<br />

Cappelleri, Joseph C 6092,<br />

e16530<br />

Cappelletti, Mariarosa e11546<br />

Capper, David 6519<br />

Cappetta, Alessandro e12037<br />

Cappuzzo, Federico 3521,<br />

e21018, e21095<br />

Capri, Giuseppe 3080<br />

Capriglione, Stella 5093,<br />

5094, e15547, e15550,<br />

e15551, e15553<br />

Caprino, Massimo TPS9154<br />

Caprio, Giuliana e11514<br />

Caprio, Kimberly Anne 1109<br />

Capuano, Loreto Giovanni<br />

e19002<br />

Capussotti, Lorenzo 4110,<br />

e14113<br />

Caraco, Corradina e14130,<br />

e19034<br />

Caraco, Corrado 8573, 8581,<br />

e19034<br />

Caramanti, Miriam e21070<br />

Carames, Cristina 5520,<br />

e14147, e15094, e18069,<br />

e18106, e21015<br />

Carapella, Carmine Maria<br />

2037, 2085<br />

Carasi, Stefania 1007<br />

Caratu, Ginevra 10511<br />

Carayol, Marion 9074<br />

Carballido, Estrella M. 5564,<br />

6568, 6608<br />

Carballido, Marcela e14521<br />

Carballo, Mar e11028<br />

Carberry, Mary 4111<br />

Carbone, Antonino 7585,<br />

e18104<br />

Carbone, David Paul 6038,<br />

7094<br />

Carbone, David P 7568<br />

Carbone, Giuseppina 4661<br />

Carbone, Salvatore Francesco<br />

e13098<br />

Carboni, Joan M. e15151<br />

Carcas, Antonio e13085<br />

Carcereny Costa, Enric 7540,<br />

7542, e18131, e18137,<br />

e18143<br />

Carcereny, Enric 1531, 3093,<br />

7522, 9129<br />

Carde, Patrice P. 8002<br />

Cardellino, Giovanni 3612<br />

Cardenal, Felipe 7522,<br />

e18137, e18143<br />

Cardenas, Andres e21000<br />

Cardenas, Jessica C e14505<br />

Cardenas, Joel e15556<br />

Cardenas, Jose M e11517<br />

Cardenes, Higinia Rosa<br />

e14683<br />

Cardillo, Giuseppe e19033<br />

Cardillo, Thomas M. 3091<br />

Cardin, Dana Backlund<br />

e14503<br />

Cardona Zorrilla, Andres<br />

Felipe e12039, e18114<br />

Cardona, Jose Vicente<br />

e18031, e18055<br />

Cardonick, Elyce 1127<br />

Cardoso, Fatima TPS661,<br />

1022, 9022<br />

Cardoso, Mauro 9557<br />

Cardozo, Timothy 9507<br />

Carducci, Michael Anthony<br />

2042, 3009, 3020,<br />

TPS3117, 4511, 4550,<br />

4559, 4564, 4619, 4642,<br />

4654, 4655, 4663, 4667,<br />

TPS4687, 6026, 10503,<br />

e15058, e15188<br />

Carella, Angelo Michele 2539,<br />

8006<br />

Carelli, Bruno e19552<br />

Carew, Jennifer S. 3073<br />

Carey, Brittany 10588<br />

Carey, George e17549<br />

Carey, Lisa A. TPS656,<br />

CRA1002, 1006, 1008,<br />

1524, 10515<br />

Carey, Thomas E e16014<br />

Carinelli, Silvestro 5090<br />

Caristi, Nicola e11056<br />

706s Numerals refer to abstract number


Carles, Joan LBA4518, 4574,<br />

TPS4692^, e15001,<br />

e15041, e21021<br />

Carlile, David 7544<br />

Carlo, Victor 1060, e11015<br />

Carlomagno, Chiara e13509<br />

Carlotti, Agnes e21014,<br />

e21046<br />

Carlotti, Carlos Gilberto<br />

e12512<br />

Carlson, Dawn M. 3532,<br />

7512, e13583<br />

Carlson, Heather R. 4130,<br />

4131<br />

Carlson, Josh John 560<br />

Carlson, Julie R. e19515<br />

Carlson, Rachel 4565,<br />

e15132<br />

Carlson, Robert O. e13581<br />

Carlson, Robert W. 2514<br />

Carmack, Condie Edwin<br />

10598<br />

Carmichael, Courtney e15084,<br />

e15178, e16563<br />

Carmichael, Lakeesha 3101<br />

Carmona, Maria Soledad<br />

e11035<br />

Carnaghi, Carlo 623<br />

Carneiro, Bruno Gustavo<br />

Muzzi Carvalho e12018<br />

Carnero Moya, Amancio<br />

e16037<br />

Carneseca, Estela C. e14599<br />

Carney, Desmond e19632<br />

Carney, Walter P. e15062,<br />

e15121, e21146<br />

Carnio, Simona 5546, 5548<br />

Carolan, Hannah 7020<br />

Caroli, Manuela 2037<br />

Caron, Huib 9517<br />

Caron, Philip 4057<br />

Caron, Whitney Paige 2591,<br />

5050<br />

Carpanese, Livio e14654<br />

Carpenito, Jennifer 4662<br />

Carpenter, Christopher L.<br />

e13601<br />

Carpenter, Christopher 4605,<br />

e15059<br />

Carpenter, Janet S 525<br />

Carpenter, John T. 10509<br />

Carpenter, William Ruffin<br />

5539, 6104, e15184<br />

Carr, Brian I. e14720<br />

Carr, Ruth Elizabeth e13522<br />

Carrasco, Eva Maria 1001,<br />

10500, 10616<br />

Carrasco, Javier e14044,<br />

e14060<br />

Carrasquillo, Jorge A.<br />

TPS4694, TPS9595<br />

Carrato, Alfredo TPS4135,<br />

e12033, e16029, e19590,<br />

e21104<br />

Carré, Typhanie 8568<br />

Carreca, Ignazio Ugo e14676<br />

Carreno, Beatriz M 2525<br />

Carreño, MariCruz e12015<br />

Carrera, Sergio e11525,<br />

e17522, e18031, e21009<br />

Carrere, Sebastien 3633<br />

Carrigan, Angela e16561<br />

Carrillo-de Santa Pau, Enrique<br />

e15191<br />

Carroll, Andrew J 9548<br />

Carroll, June e12034<br />

Carroll, Mary I. 639, 3108<br />

Carroll, Paul A. 1511<br />

Carroll, Peter 2564<br />

Numerals refer to abstract number<br />

Carroll, William L. 9507,<br />

CRA9508, 9543, 9548<br />

Carson, Kenneth Robert<br />

4594, 8080<br />

Carteni, Giacomo TPS4151,<br />

4627, TPS4683, e15185<br />

Carter, Corey Allan 7033,<br />

7103<br />

Carter, David 2552<br />

Carter, Ebony B. 5063<br />

Carter, Jeanne 9104<br />

Carter, John A. e15189<br />

Carter, Kirsten 3003<br />

Carthon, Bradley e15006<br />

Cartmel, Brenda TPS669,<br />

TPS1614<br />

Cartmill, John TPS9153<br />

Cartron, Guillaume 8026<br />

Cartwright, Thomas H. 3522,<br />

3523, 3525, 3626, 6109,<br />

e16536<br />

Caruntu, Irina Draga e15032<br />

Carus, Andreas e21110<br />

Caruso, Melania e15544<br />

Caruso, Michele 623, e18002<br />

Carvajal, Lucas E. 4660<br />

Carvajal, Richard D. 3016,<br />

8507, 8515, 8549, 8575,<br />

8598, 10004, 10019,<br />

TPS10103<br />

Carvalho, Adriana Camargo<br />

e14150<br />

Carvalho, Andre Lopes<br />

e16046<br />

Carvalho, Benilton e16041<br />

Carvalho, Carlos 3547<br />

Carvalho, Heloisa de Andrade<br />

e15562<br />

Carvalho, Rafael Augusto<br />

Ribeiro e12018<br />

Casadei, Riccardo e14647<br />

Casadei, Virginia 4084<br />

Casadio, Rita 10087<br />

Casado, Antonio LBA5000,<br />

5038, 5087, 10052,<br />

10079, e20513<br />

Casado, Enrique 1570, 9073,<br />

e11028<br />

Casado, Esther 4079<br />

Casado, Luis Felipe 6512<br />

Casado, Victoria 5520,<br />

e15094, e18069, e18106,<br />

e21015<br />

Casagrande, Mariaelena 3612<br />

Casal Rubio, Joaquin e17545,<br />

e18040, e18146<br />

Casali, Paolo Giovanni<br />

TPS9155, LBA10008,<br />

10009, 10013, 10017,<br />

10022, 10026, 10053,<br />

10065, 10067, 10079,<br />

10096<br />

Casali-da-Rocha, Jose Claudio<br />

e14027<br />

Casalonga, François e19539<br />

Casanova, Claudia 5513<br />

Casanova, Michela 9517,<br />

9519, TPS9597^<br />

Casares, Carolina 1608<br />

Casares, Maria de los Angeles<br />

10534<br />

Casaretti, Rossanna LBA4001<br />

Casas, Ana Maria 1011<br />

Casas, Maria Isabel 1001,<br />

10616<br />

Casasnovas, Olivier 8068<br />

Casassus, Philippe 8014<br />

Casbourne, Daniela 8101<br />

Cascales, Almudena e15033<br />

Casciano, Roman e14525,<br />

e14627, e15043<br />

Cascinu, Stefano e14040,<br />

e14086, e14561, e14663,<br />

e15074, e21070<br />

Cascione, Luciano 1007<br />

Case, Ashley Stephens 5101<br />

Case, Emily Christine 9092,<br />

e19624<br />

Case, L. Doug 9108<br />

Casey, Michelle LBA8509<br />

Casey, Rory e13570<br />

Casher, Jennifer e11531<br />

Cashy, John 2532<br />

Casillas, Jacqueline N. 6030<br />

Casini, Beatrice 1050<br />

Caspar, Clemens B. e19648,<br />

9059<br />

Cassano, Alessandra e14156,<br />

e16553<br />

Cassar, Alexia 10013, 10062<br />

Cassells, Lucinda Jane<br />

e19543<br />

Cassier, Philippe Alexandre<br />

3022, 10027<br />

Cassier, Philippe A 4614,<br />

10005, 10006, 10033,<br />

10078<br />

Cassinello, Javier e11074<br />

Cassingena, Andrea 3570<br />

Cassivi, Stephen David 4062<br />

Casso, Deborah 1590, 1591<br />

Castagneris, Nicolas e11044,<br />

e11501, e18125<br />

Castagneto, Bruno e14676<br />

Castaing, Denis 3547<br />

Castaldo, Noemi e11052<br />

Castaneda Altamirano, Carlos<br />

2097<br />

Castañeda, Carlos 642<br />

Castaño, Patricia e11024<br />

Castelao, Jose E 4575<br />

Castellana, Ramon 4122<br />

Castellano, Daniel E. 4118,<br />

4119, 4584, 4620,<br />

TPS4674, TPS4685,<br />

10546, e15001, e15041,<br />

e15149<br />

Castells, Antoni 3536, 3553<br />

Castelo, Beatriz 10546<br />

Castelvetere, Marina e21092<br />

Castera, Laurent e14553<br />

Castex, Marie Pierre 10076<br />

Castiglione, Monica M. 504<br />

Castillo, Javier Omar e15570<br />

Castillo, Jorge J. 8045<br />

Castillo-Martin, Mireia<br />

e14552<br />

Castillo-Tong, Dan Cacsire<br />

5034, 5071<br />

Castonguay, Vincent 5022<br />

Castro Gomez, Enrique<br />

e18040<br />

Castro Junior, Dalvaro Oliveira<br />

e16046<br />

Castro, Carlos e12039<br />

Castro, Elena 1545, 4653<br />

Castro, Gilberto 5588,<br />

e20502, e21040<br />

Castro, Javier e18011<br />

Castro, Kathleen M. 6086,<br />

9047, 9144, e16532,<br />

e19633<br />

Castro, Rafael amaral de<br />

e14173<br />

Catala, Mauricio 2515<br />

Catalan, Gustavo e11074<br />

Catalano, John V. 6624<br />

Catalano, Paul J. 4030<br />

Catalano, Vincenzo 4084,<br />

e14135<br />

Catalan, Gustavo e11081<br />

Cataldo, Francesco e19036<br />

Catane, Raphael e14143<br />

Catapano, Carlo 4661<br />

Catchatourian, Rosalind<br />

e18534<br />

Catenacci, Daniel Virgil<br />

Thomas 2561, 4022<br />

Cates, Joe e14632<br />

Cathomas, Richard 3595,<br />

e19648<br />

Catoira, Isabel e11523<br />

Caton, John Robert 7502<br />

Catot, Silvia 5068, e12012,<br />

e15575, e18053<br />

Cattaneo II, Richard e15512<br />

Cattani, Fabio 2085<br />

Catton, Charles N 9131,<br />

10050<br />

Catton, Pamela 9131<br />

Cau, Maria Chiara e19634<br />

Cauchy, Francois 3609<br />

Caudle, Abigail Suzanne 1130<br />

Caudwell, Beth e15050<br />

Caulin-Glaser, Teresa 9115<br />

Causeret, Sylvain 1584<br />

Cauvin, Cecile e19539<br />

Caux, Christophe 10562<br />

Cavalcante, Ludimila 7049<br />

Cavaletti, Guido 9089, 9090<br />

Cavallero, Erica e15192<br />

Cavallo, Federica e13527<br />

Cavallo, Michele 2057<br />

Cavanagh Podesta, Mercedes<br />

e12003<br />

Cavanna, Luigi 2057<br />

Cavenagh, James D. 8018^<br />

Cavic, Milena e12020<br />

Cavicchiolo, Tina e15056<br />

Cavina, Raffaele 7085, 7585,<br />

e18104<br />

Cawkwell, Lynn e13554<br />

Cayir, Kerim e14526<br />

Cayre, Anne 1051, 10507<br />

Cayre, Yvon E e21014,<br />

e21046<br />

Cazaly, Angelica 8566<br />

Cazap, Eduardo L. e16505<br />

Cazes, Aurelie e18102<br />

Cazin, Lionel e21156<br />

Cazzaniga, Marina Elena<br />

9090<br />

Cazzaniga, Massimiliano 519,<br />

1500<br />

Ceacareanu, Alice C. e15042<br />

Cease, Kemp Bailey e18118<br />

Cebon, Jonathan S. 8542,<br />

e17539<br />

Cebrian, Juan Luis e20513<br />

Ceccaldi, Joel 9012<br />

Ceccarelli, Matteo 1073<br />

Cecconetto, Lorenzo e11054<br />

Cecere, Fabiana Letizia<br />

e18074<br />

Cechini, Luis 4579<br />

Cedres, Susana 7041<br />

Cehelsky, Jeffrey 3062<br />

Cejas, Paloma 3618, 10547<br />

Celik, H.Eray e21050<br />

Celik, Ismail e19024<br />

Celio, Luigi 9133<br />

Celis, Aneska e20502<br />

Celiz, Pamela 4579<br />

Cella, David CRA4502, 6092,<br />

6117, 8054, 9020, 9056,<br />

9099, 9112, 9532,<br />

e16530<br />

707s


Ceraulo, Domenica 4017<br />

Cerbone, Linda e15003<br />

Cercek, Andrea 3577,<br />

e14112<br />

Cereda, Stefano 4017<br />

Cerello, Piergiorgio e13093<br />

Ceresoli, Giovanni Luca 7082<br />

Cerfolio, Robert 7008<br />

Cerhan, James R. 6571,<br />

8061, 9057<br />

Cerillo, Ivana 1554<br />

Cermak, Jaroslav 6559<br />

Cerny, Jan 6569<br />

Cerny, Thomas 9045, e14544<br />

Cerrato, Sebastian e12511<br />

Cerullo, Vincenzo e13035<br />

Cervantes, Francisco 6505^,<br />

6514^, 6625^, 6626^,<br />

TPS6639<br />

Cervantes-Ruiperez, Andres<br />

2567, 2568, 3021, 3062,<br />

3535, TPS3637, e13048,<br />

e13095^, e13097^,<br />

e13114, e14088, e14589<br />

Cervera Grau, J. M. 4587<br />

Cervera Grau, Jose Manuel<br />

e15144, e16025<br />

Cervio, Andres e12511<br />

Cesar, Isabela Costa e13008<br />

Cesaretti, Jamie A. e15162<br />

Cescutti, Jessica 3018^<br />

Cesne, Axel Le 10010,<br />

10095<br />

Cesta Incani, Ursula e13512,<br />

e13572<br />

Cesta, Alisia e11041<br />

Cetin, Bulent e14526<br />

Cetin, Karynsa 1581, 4659,<br />

e15147, e15169<br />

Cetina, Lucely e15569<br />

Cevik, Duygu 3565<br />

Ceyssens, Sarah 3559<br />

Cha, Edward 8580, e19046<br />

Cha, Joo Hee e11508<br />

Chabaud, Sylvie 10027,<br />

10092, 10095, 10562,<br />

e16036, e16560<br />

Chabolle, Frederic e16013<br />

Chabot, John A e14708<br />

Chachoua, Abraham e18139<br />

Chacko, Celin 1126<br />

Chacko, Raju Titus e15082<br />

Chacon Lopez-Muniz, Jose<br />

Ignacio 10500, 10616,<br />

e11074<br />

Chacon, Jose Ignacio e11543,<br />

e19596<br />

Chacon, Matias e14598<br />

Chad, Amine Mohammed<br />

e11516, e14725, e14726<br />

Chadha, Manpreet 3060<br />

Chadjaa, Mustapha<br />

TPS4692^, e15115<br />

Chae, Jung Min e14128<br />

Chae, Yee Soo 3554, 6536,<br />

6538, 8023, e14074,<br />

e14570, e18533<br />

Chae, Young Kwang 537<br />

Chae, Young K 536<br />

Chaft, Jamie E. 7014, 7580,<br />

10560<br />

Chagnon, Sophie 8540<br />

Chagpar, Anees B. 1109,<br />

1121, 1571, 1595,<br />

e16511<br />

Chai, Akiko 6591<br />

Chai, Christy Yoon-Hee 10070<br />

Chai, Feng 4117, 4545,<br />

8519<br />

Chai, Li e13505<br />

Chaib, Iman 4579<br />

Chaïbi, Pascal 6110, e18505<br />

Chaigneau, Loic 10014,<br />

10020, 10023, 10078,<br />

TPS10102^<br />

Chaix, Marie 10014<br />

Chakraborty, Amitava 2581<br />

Chakraborty, Nitya e21135<br />

Chakravarti, Arnab 2001,<br />

e19040, e21044<br />

Chakravartty, Arunava e13583<br />

Chalabi, Nassera 1051<br />

Chalas, Eva e15504<br />

Chalela, Patricia e16566<br />

Chaleteix, Carine 8014<br />

Challapalli, Amarnath e21093<br />

Chalmers, Jeffrey J. e21124<br />

Chamberlain, Marc C. 2046<br />

Chambers, Glenda 3025<br />

Chambers, Mark Steven<br />

e19520<br />

Chamorey, Emmanuel e16051<br />

Champ, Colin E. e11505<br />

Champeny, Torie L 6063<br />

Champion, Laurence TPS646<br />

Champlin, Richard E. 4533,<br />

6501, 6529, 6540, 6546,<br />

6551, 8040, 8102<br />

Chan, Adrien M. 6049<br />

Chan, Alexandre 9041,<br />

e13023, e18546<br />

Chan, Amy e12017<br />

Chan, Annie W 5579, 5594,<br />

e16059<br />

Chan, Anthony T. C. 5502,<br />

5511, e14085, e14528,<br />

e15110<br />

Chan, Ching-Wan 1064<br />

Chan, Dan e18077<br />

Chan, Daniel Boon Yeow<br />

2551, e13002<br />

Chan, Edward Michael 3001,<br />

3008<br />

Chan, Emily 3535, e14503<br />

Chan, Eric Chun Yong<br />

e13023<br />

Chan, Esther 6130<br />

Chan, Ge<strong>of</strong>frey 6584<br />

Chan, Godfrey Chi Fung<br />

e12006<br />

Chan, Iris Tan-Chi 2566<br />

Chan, Jennifer A. 4027,<br />

4125, e19586<br />

Chan, John K. 3058, e13078<br />

Chan, Keith S. e15002<br />

Chan, Kelvin K. e14003,<br />

e14532, e16558<br />

Chan, Kelvin 5587, e14171<br />

Chan, Kenjey e14020<br />

Chan, Matthew 6018<br />

Chan, Megan 2525<br />

Chan, Pierre e14545<br />

Chan, Sabrina e13583<br />

Chan, Stephen E 2564<br />

Chan, Stephen Lam e14528<br />

Chan, Stephen 1013, 1014,<br />

1098<br />

Chan, Susanne M. 4613,<br />

4633<br />

Chan, Timothy An-thy 7052<br />

Chan, Yiong Huak e18528<br />

Chanan-Khan, Asher Alban<br />

Akmal 8035<br />

Chance, Lacey 4574<br />

Chand, Gyan e16023<br />

Chand, Vikram K. 7557,<br />

7558, e13011<br />

Chandarlapaty, Sarat 10618<br />

Chander, Sarat 3629<br />

Chandiwana, David e15189<br />

Chandler, G. Scott e13000,<br />

e13113<br />

Chandler, James 2035,<br />

TPS2107<br />

Chandler, Jason Claud<br />

TPS1143<br />

Chandra, Anita e19518<br />

Chandra, Subhash 5104<br />

Chandra, Sunandana 8589<br />

Chandramouleeswaran,<br />

Sindhu 5577<br />

Chandramouli, Gadisetti V.R.<br />

e13045<br />

Chandramouli, Nitin B.<br />

TPS2103<br />

Chandran, Raj e13583<br />

Chandrashekar, A e15003<br />

Chandrinos, Vassilis 7564<br />

Chandwani, Kavita Dayal<br />

9009, 9010, 9028, 9141,<br />

9142<br />

Chang, Andrew C. 7037<br />

Chang, Anne L. S. e19048^<br />

Chang, Bill H. 9555<br />

Chang, Chien-Hsing e13005,<br />

e18513<br />

Chang, Daniel Tandel 4045<br />

Chang, Ellen T. 6050<br />

Chang, Eunice 3626, 6057<br />

Chang, Gee-Chen TPS7614<br />

Chang, George J. 3556,<br />

3598, 4060, TPS10632<br />

Chang, Heung-Moon e13104<br />

Chang, Howard T. 2056<br />

Chang, Hyun e14559,<br />

e18092<br />

Chang, Ilsung e13000,<br />

e13113<br />

Chang, Joe Y. 7062<br />

Chang, Jose 4660<br />

Chang, Julie E. 8064<br />

Chang, Kay W 5525<br />

Chang, Kenneth J. 4055,<br />

e14546<br />

Chang, King-Jen 1043<br />

Chang, Martin 1019<br />

Chang, Mike e15117<br />

Chang, Monica 10586<br />

Chang, Sei-Kyung e18501<br />

Chang, Susan Marina 2026,<br />

2101<br />

Chang, Tung-Chieh e16050<br />

Chang, Twan Ying 1061<br />

Chang, Victor Tsu-Shih<br />

e14727, e19638, e19639<br />

Chang, Victor T e14124,<br />

e14125, e18516, e19606<br />

Chang, Yao-Yin 1043<br />

Chang, Yen-Hwa e15064<br />

Chang, Yu-Lin 8035<br />

Chang, Yuan 8577<br />

Chang, Yuchiao 9004<br />

Chanthawong, Suthan e13023<br />

Chantrathammachart, Pichika<br />

e14505<br />

Chao, Angel 5109<br />

Chao, Bo H. e13537<br />

Chao, Calvin 549^, 3626<br />

Chao, David 7562<br />

Chao, Herta H. 6033<br />

Chao, Jeff 6615<br />

Chao, Joseph e13116,<br />

e14182<br />

Chao, Ju-Hsien e16533<br />

Chao, K. S. Clifford 7050,<br />

e15162, e17562, e21097<br />

Chao, Richard C. 4118<br />

Chao, Samuel T. 2018, 2076,<br />

2092, 2100<br />

Chao, Samuel 4106<br />

Chao, Ta-Chung 1079<br />

Chao, Yee 4108, e14605<br />

Chapman, Judy-Anne W. 500,<br />

501, 538, 561, e11005<br />

Chapman, Paul B. LBA8500^,<br />

8501, 8502^, 8515, 8549,<br />

8598<br />

Chapman, Robert e12019<br />

Chapotot, Louis 4618<br />

Chappell, Bridget e15056<br />

Charalambous, Charis e15030<br />

Charamis, Helen 1099<br />

Chari, Ajai e21006<br />

Charif, Mahmoud TPS3116<br />

Charlot, David e21047<br />

Charlot, Marjory 6097<br />

Charlson, John A. 555, 563,<br />

6063<br />

Charnsangavej, Chusilp 3544<br />

Charpentier, Danielle 3504<br />

Charpentier, Eric 1011<br />

Charpentier, Julie e15161<br />

Charpentier, Kevin 4098<br />

Charpentier, Monica e21079<br />

Chasalow, Scott D. 8593<br />

Chase, Dana Meredith 6132<br />

Chastek, Benjamin e14013,<br />

e14016<br />

Chateau, Marie-Christine<br />

10018<br />

Chatelut, Etienne 2549, 4614<br />

Chatfield, Mark D. 4531,<br />

TPS4681, 5081<br />

Chatman, David 3015<br />

Chatta, Gurkamal S. 2588<br />

Chatterjee, Anirban LBA4001<br />

Chatterjee, Aradeep 2581<br />

Chatterjee, Ashim Kumar<br />

2581<br />

Chatterjee, Nilanjan 1521<br />

Chatterton, Robert T. 518<br />

Chaturvedi, Pankaj 5585,<br />

e16021<br />

Chau, Ian LBA4000,<br />

TPS4146, e14088<br />

Chau, Noan Minh 3098<br />

Chau, Noan-Minh e16024<br />

Chau, Quincy 8028<br />

Chau, Zeling 4059, e14691<br />

Chaubet Houdu, Marie<br />

e18071<br />

Chaudhary, Imran 3093,<br />

e14019<br />

Chaudhary, Rekha T.<br />

TPS3116<br />

Chaudhri, Shalini e11062<br />

Chaudhry, Arvind 2585<br />

Chaudhry, Muhammad Ali<br />

4045<br />

Chaugule, Shraddha e18097<br />

Chauhan, Bharat 5585<br />

Chauhan, Cynthia 6133<br />

Chauhan, Lata e13568<br />

Chauhan, Nikhil 6530<br />

Chauhan, Sanket e15171<br />

Chauhan, Shailendra<br />

TPS4136<br />

Chaukar, D. 5585<br />

Chaussy, Christian G e15194<br />

Chauvie, Stephane 8077,<br />

e13093<br />

Chavarri-Guerra, Yanin 524,<br />

596<br />

Chaves-Conde, Manuel<br />

e16037<br />

Chavez, Jose Fortino e11565<br />

708s Numerals refer to abstract number


Chavez, Julio C. 6568, 6575,<br />

6588, 6608, 8084<br />

Chavez-Gallegos, Silvia 9554<br />

Chavez-Mac Gregor, Mariana<br />

537, 600, 1104<br />

Chawla, Sant P. 3015,<br />

10009, 10010, 10013,<br />

10036, 10067, TPS10100,<br />

TPS10101<br />

Chay, Christopher H. e13109<br />

Chay, Wen Yee e16024<br />

Chayahara, Naoko e16034<br />

Che, Yebin e14716<br />

Cheadle, Jeremy 3516<br />

Cheang, Maggie Chon U.<br />

1008, 3628<br />

Chebib, Amale 6110, e18505<br />

Checka, Cristina e11064<br />

Cheedella, Naga K Sucharita<br />

4086<br />

Cheema, Faisal N. e11541<br />

Cheerva, Alexandra C 6537<br />

Chehab, Nabil 4577<br />

Chekerov, Radoslav 5031,<br />

5034, 5044, 5053, 5064,<br />

5070, 5071, 5080,<br />

e15520, e19558<br />

Chekmasova, Alena A. 2586<br />

Chelis, Leonidas e14567<br />

Chellini, Federico e13098<br />

Chemler, Joseph A. e12026<br />

Chen, Albert 4660<br />

Chen, Alice P. 1010,<br />

TPS1138, 3049, 3054,<br />

3087, 5020, e19633<br />

Chen, Alice 3031, 9144<br />

Chen, Allen C. 7584<br />

Chen, Amy Y. 5560, 5570<br />

Chen, Bang-Bin 1079<br />

Chen, Beiyun 1083<br />

Chen, Bingshu E 2547,<br />

4509, TPS5600<br />

Chen, Bo e14511<br />

Chen, Caroline 8587<br />

Chen, Charles 3032<br />

Chen, Chun e18013<br />

Chen, Chun-yan 5535<br />

Chen, Clara A 6097<br />

Chen, Clara 2526, 4569,<br />

e20510<br />

Chen, Clark 2101, 10553<br />

Chen, Cong 2512<br />

Chen, Connie e15061<br />

Chen, Daniel S. e13000,<br />

e13113<br />

Chen, David 3578, 4526,<br />

10596, 10619<br />

Chen, Diana M e19048^<br />

Chen, Dongfu e14511<br />

Chen, Eric Xueyu 3082,<br />

5587, 7511, e14524<br />

Chen, Frank 5007<br />

Chen, Gong e13583, e14674<br />

Chen, Gongyan 7520, 7548<br />

Chen, Haiming e18549<br />

Chen, Han Yang e19030<br />

Chen, Hao 6066<br />

Chen, Heidi 7008<br />

Chen, Helen X. 534, 3020,<br />

7033<br />

Chen, Helen 4047, 4639<br />

Chen, Herbert e14554<br />

Chen, Homer e13555<br />

Chen, Hong 2520^<br />

Chen, Hongbin 7541, e17535<br />

Chen, Hui-Ling e14516<br />

Chen, Huiqin 1054<br />

Chen, Isan 4548, 4644,<br />

TPS4697<br />

Numerals refer to abstract number<br />

Chen, James Lin 4617<br />

Chen, James e13123<br />

Chen, Jen-Shi 4118<br />

Chen, Jianhua 7559<br />

Chen, Julianne 6529<br />

Chen, JUN e17514<br />

Chen, Ken 503<br />

Chen, Kezhong e21103<br />

Chen, Kuen-Feng e14516<br />

Chen, Kuo-Hsing e18034<br />

Chen, Lee-may 5022<br />

Chen, Lei 6583, 6594, 6600,<br />

7520<br />

Chen, Leo 6069<br />

Chen, Li-Tzong TPS4149<br />

Chen, Lieping 2514, 5506<br />

Chen, Ling Y. 9105<br />

Chen, Ling 6079, 6081,<br />

e19593<br />

Chen, Lu 9524<br />

Chen, Min Hua e14701<br />

Chen, Minshan 4033<br />

Chen, P.C. e11561<br />

Chen, Pei Xian 1538<br />

Chen, Pei-Jer 4033, 4108,<br />

e14516<br />

Chen, Peng-Yu e11526<br />

Chen, Ping 7091, 8074<br />

Chen, Robert W. 8027,<br />

e13079<br />

Chen, Ronald C. 4647, 4658,<br />

6032, 6037, 6093, 6107,<br />

e15184<br />

Chen, Shang-Hung TPS7614<br />

Chen, Shiuan e15114<br />

Chen, Siyu e13526<br />

Chen, Stephanie 3058<br />

Chen, Steven L. 1035, 1117<br />

Chen, Sting e20507<br />

Chen, Susan 3077<br />

Chen, Suzie TPS3112<br />

Chen, Tai-Tsang 8539<br />

Chen, Wallace Jian Jun<br />

4100^, 5551<br />

Chen, Wallace 5580<br />

Chen, Wei e17528<br />

Chen, Wei-Wu 1061, 1079,<br />

e14576<br />

Chen, Weidong 4092,<br />

e14575<br />

Chen, Wengen e13075<br />

Chen, Wenming 8095, 8097<br />

Chen, Xi 7532<br />

Chen, Xiang 9518<br />

Chen, Xiangai 2598<br />

Chen, Xiaoxia 7535, 10527,<br />

e18123<br />

Chen, Ya-Fang e16050<br />

Chen, Yao-Tseng e17539<br />

Chen, Yen-Lin 10058<br />

Chen, Yi-Bin Albert 6606,<br />

6617, 6618<br />

Chen, Ying 4503<br />

Chen, Yingbei 4604<br />

Chen, Yinpu 4006<br />

Chen, Yongshun e14645<br />

Chen, Yuh-Min 7519^, 7557,<br />

e18132<br />

Chen, Zhengjia 2576, 5560,<br />

7059, 7097, e14169,<br />

e14170, e14614<br />

Chen, Zhengyi 1569<br />

Chen, Zhi-Yu 3598<br />

Chen, Zhihong 7039,<br />

e13510, e18090<br />

Chen, Zhiwei 7091<br />

Chen, Zhong-pin e18121<br />

Chen, Zhong-ping e12503<br />

Chen, Zhuo Georgia e13110,<br />

e21161<br />

Chen, Zhuo 1535, 7586,<br />

10522<br />

Chen-Kiang, Selina 8036<br />

Cheng, Ann-Lii 1061, 1079,<br />

4035, TPS4147, e14516,<br />

e14576, e14617<br />

Cheng, Ashley Chi Kin 4105,<br />

5511, TPS7614<br />

Cheng, Catherine e14586<br />

Cheng, Dangxiao 1535, 7586,<br />

10522<br />

Cheng, HC 5511<br />

Cheng, Hsiao-Wang 8592<br />

Cheng, Hua e13510<br />

Cheng, Huan 573, e21106<br />

Cheng, Jason C. e14576<br />

Cheng, Jonathan D. 3027,<br />

e13598<br />

Cheng, Jushun e13542<br />

Cheng, Ke 4638<br />

Cheng, Kit 1046<br />

Cheng, Lee e16574<br />

Cheng, Liang 4586<br />

Cheng, Lucy e14727<br />

Cheng, Michael 8575<br />

Cheng, Phillip e13084<br />

Cheng, Qiao-Yuan e17512<br />

Cheng, Shih-min 5510<br />

Cheng, Shin-June e16050<br />

Cheng, Simon 7050, e17562,<br />

e21097<br />

Cheng, Skye H Hong-Chun<br />

e11526<br />

Cheng, Steven Kunyuan<br />

6047, 6051<br />

Cheng, Ying LBA4537, 7559<br />

Chenna, Ahmed 608<br />

Chenna, Sumana 1587<br />

Cheong, David 10048, 10070<br />

Cheong, Dennis 1064<br />

Cheong, Jae-Ho e14504,<br />

e14652<br />

Cheong, Pei Fen e13002<br />

Chepeha, Douglas Brian<br />

e16014<br />

Cher, Michael L. 1560, 4571<br />

Chera, Bhishamjit S. 5539<br />

Cherbavaz, Diana B. 4560<br />

Chereguini, Maria F e11517<br />

Cherepanov, Dasha 6057,<br />

e14610<br />

Cherian, Anish Jacob e19562<br />

Cherif, Amani 9572<br />

Cherkasova, Marina Valerievna<br />

e15543<br />

Chernick, Michael R e19519<br />

Chernovskaya, Tatiana V.<br />

9060<br />

Cherry, Benjamin M. 8088,<br />

8104<br />

Cherry, Mohamad Ali e17012<br />

Chervenick, Paul A. e18503,<br />

e18506<br />

Cheshire, Sprague 2074^<br />

Cheson, Bruce D. 8000,<br />

8001, 8065<br />

Chester, John D. 4531<br />

Chetaille, Bruno e18526<br />

Chetty, Runjan e13587<br />

Cheung, Foon Yiu 4105<br />

Cheung, Nai-Kong V.<br />

TPS9595<br />

Cheung, Veronica e21047<br />

Cheung, Wing Kui 3099<br />

Cheung, Winson Y. 3527,<br />

3560, 3610, 6006, 6014,<br />

6018, 6064, 6066, 6085,<br />

9078, e14048, e14532,<br />

e14537, e14588, e19547<br />

Cheung, Yin Ting 9041<br />

Chevalier, Soazig 3579<br />

Chevez-Barrios, Patricia 9515<br />

Chevreau, Christine 4625,<br />

10006, 10020, 10027,<br />

10035, 10056, 10062,<br />

10076, 10092, 10095<br />

Chew, Helen K. 1010, 3054<br />

Chew, Jenny e18063^<br />

Chew, Sin Chi 2598<br />

Cheze, Stephane 6523<br />

Chhabra, Akansha e11522<br />

Chi, Andrew S. 2009<br />

Chi, David 576<br />

Chi, Dennis 5056, 5057,<br />

5108<br />

Chi, Kim N. 4514, 4519^,<br />

4558, TPS4675^<br />

Chi, Yihebali e14606,<br />

e14668<br />

Chi, Zhi Hong 8506, 8561,<br />

8562, e15051, e15052<br />

Chia, Stephen K. L. 549^,<br />

582, 1008, 1036<br />

Chia, Yen Lin 5048<br />

Chiado’ Cutin, Simona 10055<br />

Chianese-Bullock, Kimberly A.<br />

8530, e19028<br />

Chiang, C L 4105<br />

Chiang, ChienWei 2601<br />

Chiang, Wendy M. 4117,<br />

4545, 10579<br />

Chiao, Judy H. 3053, 6520<br />

Chiappori, Alberto 2559,<br />

7065, e17559<br />

Chiara-Bruneaud, Claire<br />

e15535<br />

Chiarella, Michael 6601<br />

Chiarelli, Anna M. e12034<br />

Chiari, Rita e18023, e18101,<br />

e21094<br />

Chibaudel, Benoist 2537^,<br />

LBA3500^, 3506<br />

Chibon, Fred 9536<br />

Chibout, Salah-Dine 9562<br />

Chicata, Victor e14057<br />

Chidambaramurthy, K N<br />

e11511<br />

Chidiac, Tarek 8525<br />

Chieco, Pasquale 3061<br />

Chigurupati, Sravanthi<br />

e13516<br />

Child, J. Anthony 8015<br />

Childs, Barrett H 1021<br />

Childs, Jennifer 5082^<br />

Childs, Timothy 5032<br />

Chilson, Kate 9139<br />

Chim, James 8097<br />

Chin, Francis 10051, e20509<br />

Chin, Keisho 10541<br />

Chin, Kevin M. TPS4689,<br />

TPS4691, 8512, 8539,<br />

e19010<br />

Chin, Lynda 1055, e15200<br />

Chin, Melvin T.M. e13585<br />

Chin, Motoaki 9574<br />

Chin, Ng Soo e18528<br />

Chin, Suet-Feung 544<br />

Chinchon Espino, David<br />

e16037<br />

Chindavijak, Somjin e16040<br />

Chinen, Ludmilla T. D.<br />

e14680<br />

709s


Chinen, Ludmilla T.D.<br />

e16046<br />

Chinen, Ludmilla Thome<br />

e21073<br />

Chinn, Danielle C. e17537<br />

Chinn, Steven Bennett<br />

e16014<br />

Chinnaiyan, Arul e15205<br />

Chinot, Oliver L. 2023, 2060<br />

Chintagumpala, Murali M.<br />

9515<br />

Chintu, Chifumbe 9558<br />

Chiorean, E. Gabriela 3034,<br />

4049<br />

Chiorrini, Silvia e21070<br />

Chiparus, Ovidiu 2603<br />

Chipeta, James 9558<br />

Chiriboga, Luis 2589<br />

Chirikov, Viktor e14522,<br />

e14626, e14638<br />

Chiriva-Internati, Maurizio<br />

e21105<br />

Chirivella, Isabel 1074,<br />

e11523<br />

Chirpaz, Emmanuel 2048<br />

Chiruzzi, Chiara e21118<br />

Chirwa, Thabiso W. 10530,<br />

e21023<br />

Chisesi, Teodoro 8006<br />

Chishima, Takashi 2044,<br />

e14014<br />

Chisholm, Gary B. 1102,<br />

9049, 9065, 9069, 9096,<br />

e19528, e19584, e19603,<br />

e19613<br />

Chisholm, Julia C. 9510,<br />

9519, TPS9597^<br />

Chisti, Mohammad Muhsin<br />

628, e17015<br />

Chitale, Dhananjay 1560<br />

Chitsazzadeh, Vida 8537<br />

Chittawatanarat, Kaweesak<br />

e18538<br />

Chittenden, Eva 9049<br />

Chittoor, Sreenivas 7502<br />

Chiu, Brian C-H 8072<br />

Chiu, Chang-Fang e17014<br />

Chiu, Connie Ging Ting 2519<br />

Chiu, Joanne e14545<br />

Chiu, Ken e14182<br />

Chiu, Kung Y. e11068<br />

Chiu, Sophia e21016<br />

Chiuri, Vincenzo Emanuele<br />

e18048<br />

Chkadua, George Z. 2524<br />

Chlebowski, Rowan T. 1501,<br />

e14005<br />

Chlenski, Alexandre 9534<br />

Chmielik, Ewa e21049<br />

Chmielowski, Bartosz 3015,<br />

8587, 10003, TPS10099,<br />

TPS10103<br />

Chmura, Steven J. e11512<br />

Chng, Wee Joo 8097<br />

Chng, WeeJoo 8095<br />

Cho, Byoung Chul e14559,<br />

e18080, e18092<br />

Cho, Chie-Hee 5512<br />

Cho, Daniel C. TPS4686,<br />

8516, 8569<br />

Cho, Eun Kyung TPS7614,<br />

e14137, e14685, e18050<br />

Cho, Hearn J. TPS8111<br />

Cho, Hirotomi e11564<br />

Cho, Jae Yong e14580<br />

Cho, Kyung Sam e14128<br />

Cho, Kyung-Ja 4093, 5586<br />

Cho, Michael e19639<br />

Cho, Mong e14566<br />

Cho, Myung Mi 1587<br />

Cho, Nam-Yun 3624<br />

Cho, Nariya e21160<br />

Cho, Sang-Hee e14649<br />

Cho, Soo-Jeong 4028<br />

Cho, Steve Y. 4654, e13116<br />

Cho, Su-Hee e16003,<br />

e18148<br />

Cho, Yoon Young 6536<br />

Cho, Young-Seok e14634<br />

Cho, Yukiko 8094<br />

Chodacki, Ales LBA4512,<br />

4551<br />

Chodak, Gerald 4648<br />

Chodon, Thinle e13045<br />

Choe, Kyuran Ann e14714<br />

Choi, Audrey 1031<br />

Choi, Chang-Min e13104,<br />

e18052<br />

Choi, Eugene A. 10090<br />

Choi, Eun Kyung 7004,<br />

e18501<br />

Choi, Hye Jin 4103<br />

Choi, Il Ju 4028<br />

Choi, Jin-Hyuk e11559,<br />

e18519<br />

Choi, Jun Young 3554, 6536,<br />

e18533<br />

Choi, Kee Don 4093<br />

Choi, Kyoung-Soo 10609<br />

Choi, Mehee 1069<br />

Choi, Mihee 4028<br />

Choi, Minsig e14163,<br />

e14519, e14609<br />

Choi, Moon Ki e13094<br />

Choi, Seung-Ho 5586<br />

Choi, So-Jung 7053, e18083,<br />

e21062<br />

Choi, Walter e18139<br />

Choi, Young 574, e11029<br />

Choi, Yunsuk 6564<br />

Choksi, Janak K. 7002<br />

Chollet, Philippe J. M. 1051<br />

Chomat-Neyraud, Muriel 9080<br />

Chomy, François 9012<br />

Chon, Hye Sook TPS3114<br />

Chone, Carlos Takahiro<br />

e16041<br />

Chong, Dawn Qingqing 3098,<br />

e14155<br />

Chong, Kelly 2519<br />

Chong, Marc 4105, e14706<br />

Choo, Richard 4595<br />

Choo, Su Pin 4100^<br />

Choong, Nicholas W. e17555<br />

Chopra, Akhil e15045,<br />

e15107<br />

Chopra, Mona 8564<br />

Chopra, Rajesh 3006^<br />

Chopra, Shamit e19607<br />

Choquet, Sylvain 2023, 8030<br />

Choti, Michael A. 3596,<br />

3616, 4126, e14542,<br />

e14551<br />

Chotirosniramit, Anon e13061<br />

Chou, Joanne F. 3577,<br />

4581^, 8598, CRA9513<br />

Chou, Takaaki 8097<br />

Chouahnia, Kader e18003,<br />

e18020<br />

Chouaid, Christos 6058,<br />

7574, e16560, e18005,<br />

e18102<br />

Chouaki, Nadia LBA7507<br />

Choucair, Ali K. 2058<br />

Choudhary, Vidhu e16549<br />

Choudry, Haroon e14184<br />

Choueiri, Toni K. TPS2613,<br />

4504, 4505, 4522, 4525,<br />

4536, 4538, 4543, 4577,<br />

4582, 4585, 4635,<br />

TPS4686, e15007, e21016<br />

Choufi, B 6523, 8026<br />

Choussy, Olivier 5565<br />

Chovanec, Michal TPS4677,<br />

e15027, e15028<br />

Chow Maneval, Edna 4548<br />

Chow, Annabelle e15177^<br />

Chow, Diana 2098<br />

Chow, Laura Q. 2510,<br />

TPS2615<br />

Chow, Oliver e14656<br />

Chow, Warren Allen 3108<br />

Chowbay, Balram 2598, 3098<br />

Chowdhary, Rajesh 2532<br />

Chowdhary, Sajeel A. e12500<br />

Chowdhury, Simon 4529,<br />

4611<br />

Chowdry, Rajasree Pia 7097<br />

Choy, Edwin 10058<br />

Choy, Hak 7002<br />

Choyke, Peter L. TPS4701,<br />

10589, e18568<br />

Chrétien, Anne-Sophie 10517<br />

Chrischilles, Elizabeth A.<br />

6034<br />

Chris<strong>of</strong>os, Michael e15040<br />

Christakidis, Evagelos e14567<br />

Christensen, Ib Jarle 3576,<br />

8545<br />

Christensen, James G. 8520<br />

Christensen, Kevin e15119<br />

Christensen, Rene dePont<br />

3513, 3573<br />

Christgen, Matthias 552<br />

Christian, Beth e18508<br />

Christiani, David C. 3594<br />

Christians, Kathleen K.<br />

e14613<br />

Christiansen, Hans e13039,<br />

e21128<br />

Christiansen, Jason 517<br />

Christman, Lori e21066<br />

Christmas, Timothy e16501<br />

Christner, Susan M. 2533<br />

Christodouleas, John Paul<br />

7098, e17534<br />

Christodoulou, Marianna<br />

e21027<br />

Christodoulou, Sophie 9126<br />

Christopherson, Cindy 10586<br />

Christopherson, Kent W. 6552<br />

Christophi, Christopher<br />

TPS3643<br />

Christophyllakis, Charalampos<br />

e18105<br />

Christos, Paul J. e18029<br />

Chronowski, Christopher 9544<br />

Chu, Alice e19058<br />

Chu, Chia Lin 4056<br />

Chu, Christina 5065<br />

Chu, Chun Hung e12006<br />

Chu, CM e15110<br />

Chu, Edward TPS1138,<br />

2553, 3037, 3049, 3054<br />

Chu, Franklin 4665<br />

Chu, Hui-May 8541<br />

Chu, Kyung e14708<br />

Chu, Laura 1590, 1591,<br />

4092, e14575<br />

Chu, Luis TPS657<br />

Chu, Peiguo e21019<br />

Chu, Quincy 3013, e13011<br />

Chu, Quyen e11541<br />

Chu, Ray M 2084, 2087<br />

Chu, Tianqing e18088<br />

Chu, Yaya e13022<br />

Chua, Clarinda Wei Ling 8078<br />

Chua, Mei-Sze e14037<br />

Chua, Melvin 10626<br />

Chua, Victoria S 3015,<br />

10036<br />

Chua, Yu Jo TPS4137,<br />

e14088, e14703<br />

Chuah, Benjamin 2551,<br />

3002, e13002<br />

Chuah, Charles 6503<br />

Chuang, Eric Y 1043<br />

Chuang, Linus T. e15556<br />

Chuang, Tony 8524<br />

Chudzik, Malgorzata 603<br />

Chugh, Rashmi 3025, 10028,<br />

TPS10103<br />

Chui, Stephen Y. e11042<br />

Chui, Wai Keung 9041<br />

Chulikavit, Maruit e14525,<br />

e14627<br />

Chuma, Stacey 8043<br />

Chumsri, Saranya 1039,<br />

9103, 10572<br />

Chunda-Liyoka, Catherine<br />

9558<br />

Chung, Cathie T. 1105<br />

Chung, Christine H. 5549,<br />

5566, e16061<br />

Chung, Chung Taik e16047<br />

Chung, Doo Hyun 7566<br />

Chung, Gina 1045<br />

Chung, Harold M. e18550<br />

Chung, Hsingwen e15089<br />

Chung, Hyun Cheol TPS4142<br />

Chung, Hyun Cheol 3606,<br />

e14084, e14652<br />

Chung, Ik-Joo e14649,<br />

e15064<br />

Chung, Jo L e14037<br />

Chung, Jooseop e15064<br />

Chung, Karen e12024,<br />

e15172, e15197, e16512,<br />

e16528, e18107, e19630<br />

Chung, Peter W. M. 4535<br />

Chung, Vincent M. 3078,<br />

3108, e14182<br />

Chupreta, Sergey 10587<br />

Church, David N. 4599<br />

Church, Frank C e14505<br />

Church, Terry 1124, e19506<br />

Churilla, Thomas M. 6031<br />

Churilova, L. e11528<br />

Chyou, Po-Huang e14008<br />

Ciafone, Denise 6108<br />

Ciancola, Fabrizio e14082<br />

Cianfrocca, Mary E. 610<br />

Cianni, Roberto e14654<br />

Cianniello, Daniela 630<br />

Ciardiello, Fortunato 7550<br />

Ciarlo, Andrea LBA4001<br />

Cibula, David 5001<br />

Cibull, Michael e15578<br />

Ciceri, Fabio 6541<br />

Cicetti, Moreno 4084<br />

Cicolin, Alessandro e11524<br />

Cid, J.I. 4630<br />

Cierna, Zuzana e15027<br />

Cigler, Tessa 1010<br />

Cihon, Frank 3502<br />

Ciliberto, Domenico e14577<br />

Cillan, Elena e15159<br />

Cillero, Lourdes e14147<br />

Cilli, Fiona 9024<br />

Cillo, Umberto 4115, e14563<br />

Cinefra, Margherita e11001,<br />

e11547, e18048<br />

710s Numerals refer to abstract number


Cinieri, Saverio LBA5003,<br />

e11001, e11547, e18026,<br />

e18048<br />

Ci<strong>of</strong>fi Lavina, Maureen 608<br />

Ci<strong>of</strong>fi, Angela 10022, 10023,<br />

10044, 10057, 10089<br />

Ci<strong>of</strong>fi, Gianni 645<br />

Ciombor, Kristen Keon<br />

e14503<br />

Cione, Charlotte 4526<br />

Ciosek, Stephanie L. 1539<br />

Cipolla, Bernard G e15131<br />

Cipone, Mario 3061<br />

Cipriano, Toni 6137<br />

Ciprotti, Marika e15056<br />

Cirkel, Geert A. 2596<br />

Cirrincione, Constance<br />

CRA1002, CRA9013<br />

Ciruelos Gil, Eva Maria<br />

e11081<br />

Ciruelos Gil, Eva Maria 642<br />

Ciruelos, Eva M. 10608,<br />

e11535, e15552, e21088<br />

Citron, Marc L. 532<br />

Cittadini, Antonio e11052<br />

Cittelly, Diana M. 564<br />

Citterio, Giovanni 7581<br />

Ciuffreda, Libero e15192<br />

Ciuffreda, Ludovica e13512,<br />

e13572<br />

Ciuleanu, Tudor-Eliade<br />

LBA7000<br />

Ciuraru, Noa e14731<br />

Ciurea, Ana 8537<br />

Ciurea, Stefan O. 6546<br />

Civelek, Ali Cahid e21072,<br />

e21144, e21149<br />

Civelek, Burak e13031<br />

Clack, Glen e21037<br />

Claes, Bart 507<br />

Clapisson, Gilles 10562<br />

Clark, Amy Sanders 6000<br />

Clark, Ann Lau e16027<br />

Clark, Emma 533^, 597^,<br />

598^, e11055^<br />

Clark, Jeffrey W. 3053, 4021,<br />

7508, e14615<br />

Clark, John Ross 5579, 5594,<br />

e16059<br />

Clark, Joseph 4546, 5501,<br />

e16027<br />

Clark, Karen L 9048<br />

Clark, Katherine TPS9153<br />

Clark, Leslie Horn 5009,<br />

5092, e15511<br />

Clark, Matthew M. 1610<br />

Clark, Melanie e15008<br />

Clark, Rachel Marie e15545,<br />

e15568<br />

Clark, Sarah Victoria 6623<br />

Clark, William B. e18550<br />

Clark, William R. 4550, 4656<br />

Clark-Langone, Kim 3512<br />

Clarke, Blaise A 5019<br />

Clarke, Blaise 5010, 5026,<br />

5105<br />

Clarke, Jennifer Leigh 2026<br />

Clarke, Michael F. 2514<br />

Clarke, Noel W e15141<br />

Clarke, R. M. 1543<br />

Clarke, Roisin e12038<br />

Clarke, Stephen John 4644,<br />

9021<br />

Clarke-Pearson, Daniel L.<br />

2591, 5050, e13063<br />

Clarkson, James E. TPS9150<br />

Classe, jean Marc e11551<br />

Claudio, Martin MARCELO<br />

e18114<br />

Numerals refer to abstract number<br />

Clausen, Cathy L TPS9150<br />

Clauser, Eric e21014<br />

Clauser, Steven B. 6046,<br />

6086, 9047, 9144,<br />

e16532, e19633<br />

Claussen, Christina e19502<br />

Clay, Timothy M. 2585<br />

Cleary, James F. 9039,<br />

TPS9152, e16010<br />

Cleary, James M. 3021,<br />

3028, 3039, 3053<br />

Cleary, Sean F TPS9150<br />

Cleary, Sean P 4109, e14592<br />

Cleeland, Charles S. 5561,<br />

5589, 6002, 6551, 8091,<br />

9047, 9072, 9081, 9082,<br />

9083, 9099, 9106, 9112,<br />

9118, 9140, e11051,<br />

e15173, e19589<br />

Clegg, Eericca e15536<br />

Clemens, Michael R. 552<br />

Clemens, Michael 4023,<br />

10540<br />

Clement, Paul M. 5516^<br />

Clemente, Claudio e14042,<br />

e18021<br />

Clemente, Gemma e11036<br />

Clementi, Regina 9547<br />

Clements, Arthur e19011<br />

Clements, June 4041<br />

Clemenzi-allen, Angelo 8072<br />

Clemons, Mark J. 587<br />

Clemons, Mark 6083, 9111,<br />

10620<br />

Clerici, Thierry 8540<br />

Clerkin, Barbara M. e14582<br />

Clerkin, Kara M. e14582<br />

Cleveland, Barbara 9085,<br />

e14186<br />

Cleverly, Ann 2042<br />

Cliby, William 5076<br />

Cliffe, Bettina e19648<br />

Clifton, Guy T. 625, 2508,<br />

2529<br />

Climent Duran, Miguel Angel<br />

TPS4672, TPS4674,<br />

TPS4688, e15041<br />

Climent, Miguel Angel<br />

TPS4685<br />

Cline, Kathryn J. 6049<br />

Clingan, Philip 7511<br />

Clisant, Stephanie 10020,<br />

e11012, e14560<br />

Clive, Sally 2583<br />

Clotet, Montserrat 10555<br />

Cloughesy, Timothy Francis<br />

2005, 2101<br />

Cloutier, Martin e16571<br />

Clynes, Martin 2536, e15126<br />

Cmelak, Anthony 5502, 5566<br />

Co, Chloe e13516<br />

Coakley, Fergus V. 8580<br />

Coan, April D. 2027, 2090^,<br />

2094^<br />

Coate, Linda 2063<br />

Coates, Alan Stuart 504,<br />

9022<br />

Cobleigh, Melody A. 572,<br />

TPS657<br />

Cobo, Manuel 7011, 7540,<br />

7558, e18015, e18131<br />

Cobos, Everardo e15103,<br />

e21105<br />

Coca, Pragnya 6554, e18544,<br />

e18545<br />

Coche, Jean-Charles e14060<br />

Coche, Thierry 7056, e13061<br />

Cochet, Alexandre TPS646<br />

Cochrane, Dawn R. 564<br />

Cocker, David j e13073<br />

Cocorocchio, Emilia e19036<br />

Codacci Pisanelli, Giovanni<br />

e12510<br />

Codes, Manuel 10608,<br />

e11535, e21088<br />

Codignola, Claudio e14018<br />

Codina, Gomez 6633<br />

Codreanu, Ion 6555, e18543<br />

Codrington, Henk 7019<br />

Coeffic, David Emmanuel<br />

TPS646<br />

Coelho, Ana e15532<br />

Coelho, Luiz Gonzaga e13008<br />

Coelle, Celine 7591<br />

Coenen, Marieke J.H. 10077<br />

Coens, Corneel LBA5000,<br />

6002, 6090, 7607<br />

Coeshott, Claire TPS3638,<br />

e14501<br />

C<strong>of</strong>fin, Robert TPS8604,<br />

e14546<br />

Cogan, Chad 5088<br />

Cognetti, Francesco 630,<br />

1073, 4541, e13512,<br />

e13572, e14113, e14661<br />

Cogswell, Janet e14727,<br />

e18516, e19638<br />

Cohen, Adam D. TPS8111,<br />

e19580<br />

Cohen, Alice J. e16565<br />

Cohen, Deirdre Jill 4022<br />

Cohen, Edward P. e13034<br />

Cohen, Evan N. 8091, 9081,<br />

9083, e13582<br />

Cohen, Ezra E. W. 5500,<br />

5508, 5517, TPS5598,<br />

TPS5599, e13106<br />

Cohen, Harvey Jay 6015<br />

Cohen, Jack A. e18502<br />

Cohen, Jonathan M. e13034<br />

Cohen, Kenneth Stuart 8075<br />

Cohen, Lorenzo 6054, 9029,<br />

9031<br />

Cohen, Marlene Z. 9096<br />

Cohen, Orit 10034<br />

Cohen, Pamela TPS6639<br />

Cohen, Richard F. e16052<br />

Cohen, Roger B. 2595, 7092,<br />

7595, e13606<br />

Cohen, Samantha 5079<br />

Cohen, Shira e14731<br />

Cohen, Stephen M TPS10632<br />

Cohen, Steven J. 2583,<br />

3590, 3605, 4052<br />

Cohen, Victor e18061,<br />

e18098, e19626<br />

Cohen, Yoram e18511<br />

Cohen, Zvi R 2078<br />

Cohen-Jonathan Moyal,<br />

Elizabeth Laurence 10076<br />

Cohn, Allen Lee TPS3111,<br />

LBA3501, 4007, 7502,<br />

e18558<br />

Cohn, Allen TPS3638,<br />

e14501<br />

Cohn, David E. 1519, 5101,<br />

e15585<br />

Cohn, Michael 3590<br />

Cohn, Susan Lerner 9516,<br />

9533, 9534, 9583<br />

Cohn-Cedermark, Gabriella<br />

4508<br />

Coiffier, Bertrand 8002,<br />

8057, 8063<br />

Coimbra, Felipe José<br />

Fernandez e14728<br />

Coindre, Jean-Michel 9536,<br />

10018, 10035, 10056<br />

Coit, Daniel G. 8583, 8586<br />

Cojocarasu, Oana 9011<br />

Cok, Jaime e12041<br />

Colak, Havva e14557<br />

Colantonio, Ida TPS7109<br />

Colantuoni, Giuseppe 1073<br />

Colao, Annamaria 1604,<br />

e19038<br />

Colbeck, Margaret e19513<br />

Colcher, David 639<br />

Colchon, Paulo Henrique<br />

e12035<br />

Colditz, Graham A. e18551<br />

Coldwell, Douglas M. 3590<br />

Cole, John T. 6580<br />

Cole, Sarah 4096, e13084<br />

Coleman, Morton 8036,<br />

e21006<br />

Coleman, R. Edward e13592<br />

Coleman, Robert Edward 502,<br />

511, 513, TPS670, 1094,<br />

LBA4512, 4551<br />

Coleman, Robert L. 5015,<br />

5021, 5027, TPS5114,<br />

e15578<br />

Coletti, Gino e14010<br />

Colevas, A. Dimitrios 2514,<br />

5525, 5566, 9024<br />

Colevas, Electra A 5525<br />

Colevas, Kara 2028<br />

Colicchio, Lynn e16515<br />

Collard, Olivier 10020,<br />

10078<br />

Collecchi, Paola 629<br />

Colleoni, Marco 504<br />

Collette, Laurence 4053<br />

Collichio, Frances A. 6093<br />

Collignon, Marie-Anne<br />

e15558<br />

Collin, Christine 3520<br />

Collin, Francoise 10018<br />

Collins, Brian Timothy 2091<br />

Collins, Connie TPS4687<br />

Collins, Denis 637, 2536<br />

Collins, Helen e19511<br />

Collins, Ian M. 1502<br />

Collins, Jerry M. 3087<br />

Collins, LaTonya R 6505^<br />

Collins, Sara 2534<br />

Collins, V. Peter TPS10633^<br />

Collinson, Fiona Jane e13083<br />

Collisson, Eric Andrew 3058<br />

Colman, Howard 2002, 2003<br />

Colmenero, Maria e15033<br />

Colo, Anna Estela Luiza<br />

e11059<br />

Colom, Bartomeu e11036<br />

Colom, Helena 3096<br />

Colombini, Sonia e19549<br />

Colombino, Maria 8581,<br />

e14094<br />

Colombo, Chiara 10000,<br />

10016, 10017<br />

Colombo, Massimo 4033<br />

Colombo, Nicoletta LBA5000,<br />

5001, 5090, e13095^,<br />

e13097^<br />

Colombo, Renata e15562<br />

Colomer, Anna e14552<br />

Colon-Otero, Gerardo 2015<br />

Colorito, Maria Luisa e21095<br />

Colucci, Silvia e13525<br />

Columbo, Laurie Ann 4045<br />

Comandone, Alessandro<br />

10055<br />

Comas, Carmen 1570<br />

Combemale, Patrick 10049<br />

Comber, Harry 2609, 6036,<br />

e16514<br />

711s


Come, Steven E. 9071, 9100<br />

Comis, Silvia 4122<br />

Commo, Frederic 10556<br />

Company, Maria 4089<br />

Compton, Carolyn C. 3517<br />

Compton, Kathryn 3103<br />

Conan, Anne-Hélène 9563<br />

Conces, Madison 7032,<br />

7107, 7108<br />

Concha, Angel 4089<br />

Concin, Nicole 5071, 5111<br />

Conde, Esther 3068<br />

Conde, Sara e18111<br />

Condello, Caterina 1554,<br />

e15034<br />

Condemi, Giovanni e14676<br />

Condori, Dhiossett e15159<br />

Condy, Mercedes M 10002,<br />

10004<br />

Cone, Christina 2074^<br />

Conforti, Rosa 1091<br />

Cong, Xiangyu e14534<br />

Cong, Xiuyu Julie TPS5598<br />

Cong, Ze e15172, e16528<br />

Conill, Carles 3536<br />

Conings, Godelieve 9035<br />

Conklin, James J e13082<br />

Conkling, Paul TPS3111,<br />

e18047<br />

Conkright, William A. e15115<br />

Conley, Anthony Paul 10048,<br />

10070, 10094, e20511<br />

Conley, Barbara A. 5020<br />

Conley, Christoher R 8053<br />

Conley, Jennifer e13592<br />

Conlon, Susan e21147<br />

Connault, Jerome 1580<br />

Connell, Jacqueline e14015<br />

Connell, Louise Catherine<br />

e19632<br />

Connelly, Mark C. 7092,<br />

10533<br />

Conner, Kathy 8075<br />

Conner, Michael G. 5036,<br />

e15502<br />

Conner, Thomas 1502<br />

Connery, Cliff P. e18139<br />

Connolly, E. M. 1511, 1543<br />

Connolly, Elizabeth M.<br />

e12038<br />

Connolly, Elizabeth e18095<br />

Connolly, Gregory Clayton<br />

6059<br />

Connolly, John 3098<br />

Connolly, Roisin M. 10509<br />

Connor, James e14520<br />

Connor, Michael e14632<br />

Connors, Joseph M. 8049<br />

Conort, Pierre e15105<br />

Conrad, Charles A. 2003,<br />

2029^, 2036, 2064<br />

Conradi, Lena-Christin 3600,<br />

e14161, e14162, e14167<br />

Conrads, Thomas P e15560<br />

Conroy, John D. e19544,<br />

e19567<br />

Conroy, Thierry 1584,<br />

LBA4003, 4053<br />

Considine, Michael 5549<br />

Constantinidou, Anastasia<br />

10086<br />

Constenla, Manuel e18011<br />

Constine, Louis S. 9524<br />

Conte, Pier Franco 631<br />

Conte, Pierfranco 614<br />

Contel, Estela e11523<br />

Contentin, Nathalie 6542<br />

Conter, Henry Jacob 6529<br />

Conti, Alessandro e15074<br />

Conti, Laura e13527<br />

Conti, Peter 639<br />

Contreras, Ana Cecilia<br />

e11517<br />

Contreras, Ana C e11006<br />

Contu, Antonio Silverio<br />

e14094<br />

Cook, Gordon 8015<br />

Cook, Michele 5073<br />

Cook, Robert W. 7106, 8543<br />

Cooley, Timothy Patrick 4030<br />

Cools, Nathalie 2506<br />

Coombes, Charles e21093<br />

Coombes, Kevin R. 7096,<br />

10612<br />

Coombes, R. Charles e21137<br />

Coombs, John e15043<br />

Cooney, Kathleen A. e15205<br />

Cooney, Matthew M.<br />

TPS10099, e15208<br />

Cooper, Gregory 1569<br />

Cooper, Joseph 569, 571,<br />

6098, e11002, e16540,<br />

e16552<br />

Cooper, Laurence 6540<br />

Cooper, Matthew 6091,<br />

e16007<br />

Cooper, Robert Michael 9588<br />

Cooper, Tiffiny 621<br />

Coopey, Suzanne 1122<br />

Cooray, Prasad 3629,<br />

e14644, e14718<br />

Cope, Frederick Oliver<br />

e21066<br />

Cope, Reed e14632<br />

Copelan, Edward Alan 8055<br />

Copeland, Larry J. e15585<br />

Copetti, Massimiliano e21092<br />

Copley-Merriman, Kati 6626^<br />

Coppola, Carmela e11052,<br />

e13539<br />

Coppola, Sara 10017<br />

Copreni, Elena LBA7501<br />

Copur, Mehmet Sitki 9110<br />

Corallo, Salvatore e16553<br />

Corazzelli, Gaetano 8066,<br />

8083<br />

Corben, Adriana 10618<br />

Corbishley, Catherine M 4640<br />

Corcoran, Claire 617<br />

Corcoran, Ryan Bruce 3528<br />

Cordeiro De Lima, Vladmir<br />

Claudio 643, e21073<br />

Cordeiro, Peter 9043<br />

Cordero Franco(1), Nazaret<br />

e11543, e19596<br />

Cordio, Stefano Sergio<br />

LBA4001, e14040,<br />

e15544, e18026<br />

Cordova, Sharon e15094,<br />

e18069, e18106, e21015<br />

Coriat, Romain 2557<br />

Corless, Christopher L. 4586,<br />

TPS7609, 10591<br />

Corman, John M 2564<br />

Cormio, Gennaro LBA5003<br />

Corn, Paul G. 4513, 4523,<br />

4549, 4615, e15092<br />

Cornelio, Gerardo H. 1534<br />

Cornelison, Terri L 520<br />

Cornelius, Lynn 2525<br />

Cornely, Oliver A. 9067<br />

Cornillon, Jerome 6534<br />

Cornu, Jean-Nicolas e15105<br />

Corona Villalobos, Celia<br />

Pamela 4114<br />

Corona, Rosalie 1563<br />

Coronado, Cinthya 3093,<br />

e15001<br />

Coronado, Monica e13085<br />

Corpening, Jennifer C.<br />

e16561<br />

Corradetti, Michael Nino<br />

7050, 7098, e17534<br />

Corradini, Nadege 9517,<br />

10014<br />

Corradini, Paolo e18572^<br />

Corradino, Irene e19549<br />

Corrado, Claudia TPS8118<br />

Corral Jaime, Jesus 2530,<br />

LBA7507, 10525<br />

Corral, Mitra 5533, e16053<br />

corrales-Alfaro, Carmen 6599<br />

Corrales-Rodriguez, Luis<br />

e18136<br />

Corre, Romain 7574<br />

Correa, Arlene M 4078,<br />

4086, 7043<br />

Correa, Denise 2006, 2008<br />

Correa-Gallego, Camilo 3620<br />

Correale, Pierpaolo e13098,<br />

e14577<br />

Correll, Judy e20007<br />

Corrie, Philippa 4121,<br />

TPS8605, e14625<br />

Corsi, Domenico C. e14082<br />

Cortes, Javier 509, 566,<br />

597^, 598^, 619, TPS652,<br />

1032, TPS1140^, 10511<br />

Cortes, Jorge E. 6501, 6503,<br />

6504, 6506, 6511, 6513,<br />

6525, 6528, 6544, 6558,<br />

6578, 6585, 6587, 6589,<br />

6596, 6597, 6601, 6603,<br />

6604, 6605, 6607,<br />

TPS6635, TPS6636,<br />

TPS6639, TPS6641,<br />

e19589<br />

Cortes, Matias Nicolas<br />

e11044, e11501, e18125<br />

Cortes-Funes, Hernan 642,<br />

4620, e11074, e15033,<br />

e15552<br />

Cortes-Gonzalez, Ruben<br />

e11098<br />

Cortesao, Emi-lia e12040<br />

Cortesi, Enrico e18065<br />

Cortessis, Victoria K 4575<br />

Cortez, Brandon 8580<br />

Cortez, Czrina 2518<br />

Corthals, Jurgen 2507<br />

Cortinovis, Diego Luigi 9090<br />

Cortot, Alexis B. e18006,<br />

e18066<br />

Cosaert, Jan TPS4675^<br />

Coscas, Yvan 1567, 1568<br />

Cosentino, Sebastiano<br />

e18096<br />

Cosgrove, David 3020,<br />

TPS3117, 3596, 4114<br />

Cosimelli, Maurizio e14082<br />

Coskinas, Xanthi 7079^<br />

Coskun, Hasan Senol e14070<br />

Coskun, Ugur 638<br />

Coso, Diane TPS1613, 6633,<br />

e18526, e19539<br />

Cossor, Furha Iram e14005<br />

Cossu, Antonio 8581,<br />

e14042, e14094<br />

Costa, Bruno M 2026<br />

Costa, Carlota 4068, e14521,<br />

e18130, e18131, e18137,<br />

e18143<br />

Costa, Daniel Botelho 7523,<br />

7579<br />

Costa, Felipe D’Almeida 643<br />

Costa, Fernando Ferreira<br />

e16041<br />

Costa, Luciano 8010<br />

Costa, Luis 607<br />

Costa, Serban Dan 541<br />

Costa, Wilson Luiz e14728<br />

Costantini, Chiara 4081<br />

Costantino, Joseph P.<br />

LBA506, 538, LBA1000,<br />

TPS1142<br />

Costello, Brian Addis 4657,<br />

5077, e13052<br />

Costello, Joseph F 2026<br />

Costello, Rene 8088, 8104,<br />

e18568<br />

Costes Martineau, Valerie<br />

e16013<br />

Cote, Marc-Andre 634<br />

Cote, Michele L 7063, 7593<br />

Cote, Richard J 4575, 4663,<br />

10503<br />

Cotreau, Monette M. 4132,<br />

e13041<br />

Cotrell, Aaron e17015<br />

Cotter, Maura B. 1042<br />

Cotter, Ryan 5578<br />

Cottini, Silvia e14661<br />

Cottler-Fox, Michele e18576<br />

Cotto, Elise e18505<br />

Cottu, Paul H. 601, e15524<br />

Cottura, Ewa 10020<br />

Coudert, Bruno P. TPS646<br />

Coughlin, Christina Marie<br />

3016<br />

Coukos, George 5065<br />

Coulson, Luke C 9127<br />

Coulter, Sally 550<br />

Counago, Felipe 1111<br />

Counsell, Nicholas 7538<br />

Countouriotis, Athena Maria<br />

6512<br />

Coupkova, Helena e18062<br />

Coupland, Sarah 8523, 8564,<br />

TPS8605<br />

Courbon, Frederic 9517<br />

Couriel, Daniel R. 8011<br />

Cournoyer, Sonia 9563, 9570<br />

Courtier, Anais 10562<br />

Cousin, Sophie 10013,<br />

10014, e13118<br />

Cousson-Gelie, Florence 9074<br />

Couto, Rosario e15532<br />

Coutre, Steven E. 6502,<br />

6507, 6518^, 8012<br />

Couture, Felix 3504<br />

Couvelard, Anne 4129, 4133<br />

Couvillon, Anna TPS4701<br />

Cova, Agata 5555<br />

Cova, Dario e14676<br />

Covello, Kelly L. 5533<br />

Cowan, Kenneth H. 1538<br />

Cowan, Richard 4509<br />

Cowey, C. Lance TPS4686<br />

Cowey, Charles Lance 4603,<br />

8503^, 8567<br />

Cowie, Julie e19617<br />

Cox, David TPS1145<br />

Cox, Donna S 3023, 4108<br />

Cox, James D. e14548<br />

Cox, Nancy J 9516<br />

Cox, Trevor F e14625<br />

Coxon, Fareeda Y LBA4000<br />

Coxon, Katy Marie 6115<br />

Coyne, James C. 9145<br />

Crabtree, Carol 6113<br />

Craft, Paul Stanley e15152<br />

Crago, Aimee Marie 10015<br />

Craig, Adam 6513, 6596,<br />

6604, TPS6637<br />

Craig, Michael TPS6637<br />

Craighead, Peter S TPS5116<br />

712s Numerals refer to abstract number


Cramer, Laura D. 4651<br />

Crane, Christopher H. 4051<br />

Crane, Gemma Louise 4599<br />

Crane, Heidi TPS3634<br />

Cranmer, Lee D. 10010<br />

Crawford, Beth e16510<br />

Crawford, Brooke S. e18508<br />

Crawford, E. David 4, e15176<br />

Crawford, Jeffrey 1062, 6129,<br />

9135<br />

Crawford, S. Michael 5046<br />

Crawley, Freya 587<br />

Craxi, Antonio TPS4151<br />

Crea, Francesco 10611<br />

Creekmore, Allison N 3047<br />

Creelan, Ben C. 2559<br />

Cremolini, Chiara 3518,<br />

3540, 3546, 3549, 3555,<br />

3585, 3597<br />

Crequit, Jacky 7574<br />

Crescentini, Robert M. 6568,<br />

6575, 6588, 6608, 8084<br />

Crespo Massieu, Carmen<br />

e11081<br />

Crespo, Aurora e16000<br />

Cress, Rosemary 6052<br />

Cresti, Nicola 3100<br />

Cretella, Elisabetta e13019<br />

Cretin, Elodie e19623<br />

Cretin, Jacques 9011<br />

Crevenna, Richard 2071<br />

Creveuil, Christian TPS7112<br />

Crew, Katherine D. 9018,<br />

10516<br />

Criado, Ernesto 1570<br />

Crida, Benedetta e18103<br />

Crighton, Emilia M 3599<br />

Crino, Lucio 1604, 3521,<br />

5513, 7503, 7531, 7533,<br />

7550, 7600, e18021,<br />

e18023, e18101, e19038,<br />

e21018, e21094, e21096<br />

Cripe, Timothy P 9509<br />

Crippa, Flavio 4507, e21118<br />

Cripps, M. Christine e19527<br />

Crisan, Anamaria 4565<br />

Crisci, Stefania 8083<br />

Crisi, Girolamo 2057<br />

Crispens, Marta A. e15582<br />

Crispo, Anna 1554<br />

Crist, Wendy A LBA8509<br />

Cristea, Mihaela C. 3108,<br />

5025, 7518, 9068<br />

Cristin, Antonio 3612<br />

Crist<strong>of</strong>anilli, Massimo 531,<br />

577, 594, e13582<br />

Critchfield, Jeffrey J e14609<br />

Crivello, Margaret Lord 6061<br />

Crockard, M. A. e14022<br />

Crocker, Theresa e21155<br />

Crombet Ramos, Ofelia 6580<br />

Crombet Ramos, Tania 2515,<br />

2527<br />

Crombie, Catherine e16501<br />

Cromer, Jamie TPS650<br />

Cromwell, Ian 549^<br />

Crona, Daniel James e15086<br />

Cronin, Beth 5100<br />

Cronin, Maureen T. 1015,<br />

3533, 4649, 7510, 7529,<br />

10524, 10559, 10590,<br />

e17541, e19004<br />

Cronk, Michelle F. 3572,<br />

TPS4137<br />

Crook, Tim e18567<br />

Cropet, Claire 10006<br />

Cros, Jerome F 5554<br />

Cros, Jerome 4133<br />

Numerals refer to abstract number<br />

Cros, Sara 7540, 9129,<br />

e12504, e18131, e18143<br />

Crosara, Marcela Alves<br />

Teixeira 2038<br />

Crosby, Thomas TPS4143<br />

Crosby, Tom David Lewis<br />

LBA4000<br />

Crosby-Thompson, Allison<br />

e18530<br />

Crouzet, Sebastien e15194<br />

Crow, Jaime 1028<br />

Crowley, John CRA6009,<br />

8041<br />

Crowley, Michael J e21011<br />

Crown, John 581, 615, 617,<br />

622, 632, 633, 637, 1042,<br />

2536, e13520, e19059<br />

Cruciani, Giorgio 7550, 7584<br />

Cruickshank, Scott 567<br />

Crump, Barbara e14124,<br />

e18516, e19606, e19638<br />

Cruz Mora(1), Miguel Angel<br />

e11543, e19596<br />

Cruz, Cristina 3014, e13605<br />

Cruz, Crystal M 2589<br />

Cruz, Felipe Melo e16568,<br />

e19537, e19552<br />

Cruz, Josefina 10079<br />

Cruz, Marcelo Rocha De<br />

Sousa e11058, e18078<br />

Cruz-Borja, Primo 9554<br />

Cruz-Munoz, William e11061<br />

Cryer, Sarah K 7579<br />

Csajka, Chantal e11048<br />

Csapo, Kimberly A. e16561<br />

Cseh, Jozsef 2522, 3530<br />

CsToth, Ingrid e18006<br />

Cuadra, Jose Luis 4579,<br />

e12504<br />

Cuartero, Julie 5105<br />

Cuatrecasas, Miriam 3536,<br />

3553, e14041<br />

Cubedo, Ricardo 10079,<br />

e21088<br />

Cubelli, Marta e13057<br />

Cubero, Daniel de Iracema<br />

Gomes 5588<br />

Cubero, Daniel Iracema<br />

e16568, e19537, e19552<br />

Cubillo, Antonio 3066,<br />

TPS3637, 4040, e15111<br />

Cucchi, Silvia e13105<br />

Cuccuini, Wendy 8048<br />

Cuéllar Ponce de león, Luis<br />

Ernesto e18524<br />

Cueva Banuelos, Juan<br />

Fernando e14733<br />

Cufer, Tanja 558, 7081,<br />

e16505<br />

Cuff, Justin 6072<br />

Cuffe, Sinead 1535, 6005,<br />

7586, 9032<br />

Cui, Chuan Liang 8506,<br />

8554, 8561, 8562,<br />

e15051, e15052<br />

Cui, Kemi 2098<br />

Cui, Yonggang e21026<br />

Cuillerot, Jean-Marie<br />

TPS4689, TPS4691,<br />

TPS7113, TPS7611<br />

Cuinet, Marie 4618<br />

Cuisenier, Jean 1584<br />

Culakova, Eva 6129, 9135<br />

Culbertson, Richard e15130<br />

Culine, Stephane 4583<br />

Culliney, Bruce 5531, 5559,<br />

5581<br />

Cultrera, Jennifer L. 6568,<br />

6608, 8084, e18503,<br />

e18506, e18509, e21123<br />

Cummings, Francis J. e13126<br />

Cummings, Francis e13124<br />

Cummings, K. Michael 1529<br />

Cummins, Michelle M 7079^<br />

Cunanan, Josephine e13117,<br />

e15008<br />

Cunha, Geraldo Felicio<br />

e13008<br />

Cunha, Isabela Werneck<br />

e11058, e20506<br />

Cunningham, David 3531,<br />

3587, LBA4000, 4004,<br />

4029, 4121, TPS4139,<br />

TPS4143, e13541,<br />

e14088, e14132, e14625<br />

Cuorvo, Lucia Veronica<br />

e21102<br />

Cupissol, Didier 10035,<br />

10092, 10095<br />

Cuppini, Lucia 2083<br />

Curiel, David T e13087<br />

Curiel, Tyler J. 3073<br />

Curigliano, Giuseppe 546<br />

Curley, Steven A. 4116<br />

Curley, Tracy 4548, TPS4697<br />

Currà, Maria Francesca<br />

e18023, e18101<br />

Curran, Emily 6582<br />

Curran, John 10590<br />

Curran, Walter J. 2001,<br />

2008b, 2013, 5583, 7059<br />

Curran, Walter John 7097<br />

Currin, Erin-Siobhain R. 1088<br />

Currow, David C 2604, 9128,<br />

TPS9153<br />

Curry, Jonathan 8537<br />

Curry, Richard Charles 2006<br />

Curry, Saundra e15021<br />

Cursio, Elisabetta e14156<br />

Curti, Alessandra 4097<br />

Curti, Brendan D. TPS2613,<br />

3032<br />

Curtin, John Patrick 5016<br />

Curtin, Karen 9525, 9561<br />

Curtis, Alexandra e15203,<br />

e15204<br />

Curtis, Kelly Kevelin 3060,<br />

8053<br />

Curve, Herve LBA5000<br />

Curvietto, Marcello 8573,<br />

e19034<br />

Cushen, Samantha 1559<br />

Cusi, Maria Grazia e13098<br />

Cusnir, Mike e19646<br />

Cussenot, Olivier e15105<br />

Custodio Cabello, Sara<br />

e12003, e14058<br />

Custodio, Ana 3618, 10547,<br />

e12033, e13085<br />

Cusumano, Pino 10554<br />

Cuynet, Patrice e19623<br />

Cuyun Carter, Gebra 5533,<br />

e16053<br />

Cvancarova, Milada 4066,<br />

4637, 9590, e14053<br />

Cwiertka, Karel 1087<br />

Cynober, Luc e13056<br />

Czapiewski, Piotr 10561<br />

Czapski, Natalie e12024,<br />

e15197, e18107, e19630<br />

Czarkowski, Andrea e19028<br />

Czartoryska-Arlukowicz,<br />

Bogumila 505, 603<br />

Czechura, Tomasz 1033,<br />

6040<br />

Czerkawski, Barbara 549^<br />

Czerlanis, Cheryl M. e13121<br />

Czernin, Johannes 10012<br />

Czerwinski, Debra 2514<br />

Czito, Brian G. e14724<br />

Czok, Sarah e15548<br />

Czuczman, Myron Stefan<br />

8001, e18530, e18537,<br />

e18538, e18539<br />

Czuczman, Natalie e18537<br />

D<br />

D Arlhac, Michel 1574<br />

D’Achille, Anna Maria e19002<br />

D’Adamo, Stefania 3061<br />

D’Addetta, Carmela e21092<br />

D’Agostino, Diego 7056,<br />

e13061<br />

D’Aiuto, Giuseppe 1500<br />

d’Amato, Claudia e13509<br />

D’Ambrosi, Rafael e15033<br />

D’Ambrosio, Consuelo 5513<br />

D’Ambrosio, Lorenzo 10066,<br />

e15192, e20512<br />

D’Ambrosio, Paul 4662<br />

D’Amico, Anthony Victor 6121<br />

D’Amico, Thomas A. 7008,<br />

e14562, e18018<br />

D’Amore, Francesco 8051<br />

D’Andrea, Gabriella 1016,<br />

10514<br />

D’Angelica, Michael Ian<br />

3577, 3620<br />

D’Angelo, Sandra P. 8549,<br />

8575, 8598, 10004,<br />

10019<br />

D’Arcangelo, Manolo e21095<br />

d’Ario, Giovanni 3575<br />

D’Ascenzo, Fabrizio e15192<br />

D’Auria, Kevin 8597<br />

D’Avella, Domenico 2049<br />

D’Cunha, Nicholas C. e21105<br />

D’Cunha, Ruth e21105<br />

D’Emidio, Silvia 4084<br />

D’haens, Jean 2050<br />

D’hondt, Veronique e13594<br />

D’Incalci, Maurizio 10047<br />

D’Incecco, Armida e21018<br />

d’Journo, Xavier 4091<br />

D’Souza, David 6130<br />

D, Madhusudan e19619<br />

Da Corte, Donatella e13019<br />

da Costa, Alexandre Andre<br />

Balieiro 643<br />

DA Silva, Guedes e14673<br />

da Silva, Joao Santana<br />

da Silva, Marcelo Gurgel<br />

1075<br />

Daaboul, Hamid Elia<br />

e15509<br />

e20500<br />

Dabakuyo, Tienhan Sandrine<br />

1584<br />

Dabney, Raetasha Sheavette<br />

625, 2508, 2529<br />

Dabrow, Michael B. e19519<br />

DaCosta Byfield, Stacey<br />

e16539<br />

Dad, Shakeela e14079<br />

Dadaev, Tokhir 1545<br />

Dadduzio, Enzo e14156<br />

Dadras, Soheil Sam e21135<br />

Dafni, Urania 592<br />

Daga, Haruko e18036,<br />

e18042<br />

Dagoglu, Nergis 9567<br />

Dahan, Laetitia 4087, 4091<br />

Dahiya, Saurabh e19543<br />

Dahl, Olav 4508<br />

Dahlberg, Suzanne Eleanor<br />

7516, 7553<br />

Dahle-Smith, Asa LBA4001<br />

713s


Dahlheimer, Tambra 2546<br />

Dahmane, Elyes e11048<br />

Dahut, William L. 2526,<br />

3043, 4569, TPS4701,<br />

10589<br />

Dai, David 3007<br />

Dai, Jie 8561<br />

Dai, Luyan TPS5599<br />

Dai, Qun e18574<br />

Dai, Shuqin e11091<br />

Dai, Wei 5582<br />

Daignault, Stephanie 2602,<br />

4506<br />

Daignault-Newton, Stephanie<br />

4591, e18118<br />

Daigo, Yataro e13552,<br />

e13565<br />

Dailey, Seth H e16010<br />

Daily, Maureen 6054<br />

Dakheel, Omar 2577<br />

Dakhil, Shaker R. 551, 1083,<br />

6591, 7002, 7590, 7592,<br />

TPS7618, 9001, 9014,<br />

e15115<br />

Dal Bello, Maria Giovanna<br />

7577, e21065<br />

Dal Canton, Orietta 10055<br />

Dal Mas, Antonella e14010<br />

Dal, Pinar e11045<br />

Dalac-Rat, Sophie 8591<br />

Dalal, Jay S. e16027<br />

Dalal, Mehul 6059<br />

Dalal, Shalini 9002, 9025,<br />

9049, e19584<br />

Dalasanur Nagaprashantha,<br />

Lokesh Prasad Gowda<br />

e13557<br />

Dalbagni, Guido 4581^<br />

Dalban, Cecile 3506, 9026^<br />

Dale, David C. 1062, 6129,<br />

9135<br />

Dalen, Egbert J. van e19560<br />

Dalgleish, Angus George 8534<br />

Dalia, Samir 6568, 6575,<br />

6588, 6608, 8084<br />

Dalkin, Bruce L. 4521<br />

Dall’Occa, Patrizia 2061<br />

Dalla Palma, Maurizia<br />

e12037<br />

Dalla Pria, Alessia e14574<br />

Dallas, Nikolaos Andreas<br />

1130<br />

Dallinger, Claudia e15037<br />

Daly, Andrew 6548<br />

Daly, Corinne 6027<br />

Daly, Mary Beryl 1502, 1550<br />

Daly, Peter e12038<br />

Damaj, Gandhi Laurent 6534,<br />

6542, 8026<br />

Damast, Shari 9104<br />

Damato, Bertil 8523, 8564,<br />

TPS8605<br />

Damber, Jan-Erik 4550<br />

Damian, Silvia 3080<br />

Damiani, Patrizio 5093,<br />

5094, e15547, e15550,<br />

e15551, e15553<br />

Damiano, Vincenzo e13509,<br />

e15034<br />

Damião, Ronaldo 4510, 4642<br />

Damjanov, Nevena 4111<br />

Damjanovic, Svetozar e21136<br />

Damm, Robert e14630<br />

Danaee, Hadi 2597<br />

Danaei, Mahmoud 10600<br />

Dancey, Janet 3047, e13049,<br />

e13107, e19616<br />

Dane, Faysal 3565, e14526,<br />

e16015<br />

Danenberg, Kathleen D. 7582<br />

Danenberg, Kathleen D 1040,<br />

10630<br />

Danenberg, Kathleen 7594<br />

Danenberg, Peter V. 7066<br />

Danese, Mark e15172<br />

Danesh Manesh, Amir<br />

Hossein 6557<br />

Daneshmand, Manijeh 3515<br />

Daneshmand, Siamak 4576,<br />

4578, 4586, 4588, 4589<br />

Danesi, Hassan 1016, 3085,<br />

4077<br />

Danesi, Romano 10611,<br />

e13057<br />

Dang, Chau T. 532, 6025,<br />

10514<br />

Dang, Ha 9539<br />

Dang, Long H. TPS4136,<br />

4603, TPS10099<br />

Dang, Nam H. 8081<br />

Dangi-Garimella, Surabhi<br />

e14515<br />

Daniel, Catherine 1095, 5067<br />

Daniel, Davey B. 7035, 7100,<br />

7567<br />

Daniel, Hubert D 3616<br />

Daniele, Bruno 4006,<br />

TPS4151<br />

Daniele, Gennaro 3030,<br />

TPS4151, e14130<br />

Daniels, Dianne 2078<br />

Daniels, Gregory A. 10539<br />

Daniels, Molly S 1513, 1518,<br />

1520<br />

Danila, Daniel Costin 4548,<br />

4562, TPS4697<br />

Danilov, Alexey Olegovich<br />

e13059<br />

Danilova, Anna Borisovna<br />

e13059<br />

Danjoux, Marie 4129<br />

Dannenberg, Andrew J 10514<br />

Danova, Marco 4017<br />

Dansey, Roger D. 4510<br />

Dansin, Eric 7575<br />

Danso, Michael A. TPS658<br />

Danson, Sarah 6113, 7081,<br />

TPS8605<br />

Dantonello, Tobias M. 9573<br />

Dantzer, Jessica 525<br />

Dao, Fanny 5030<br />

Daoudi, Khaoula e11502,<br />

e11516, e14725, e14726<br />

Daponte, Antonio 8573<br />

Dar, Lalit 5540<br />

Darb-Esfahani, Silvia 522,<br />

523, e15531<br />

Darby, Charles Harris e13588<br />

Darcourt, Jacques TPS646,<br />

8077<br />

Darendeliler, Emin 9567<br />

Darfler, Marlene e21068<br />

Darkow, Theodore e17009<br />

Darling, Gail Elizabeth 1583,<br />

10522<br />

Darloy, Franck e16051<br />

Darnell, Colleen Marie<br />

e16017<br />

Daroqui, Cecilia e14019<br />

Darvin, Maxim E. 5064,<br />

e19558<br />

Darvishian, Farbod 2589,<br />

8550, 8589, e19003<br />

Darwish, Alaa Eldin e15530<br />

Darwish, Mona 9547, e18522<br />

Das Gupta, Soumyasri<br />

e13568<br />

Das, Asha 1591<br />

Das, Mayukh 7583<br />

Das, Millie Snigdha 10574<br />

Das, Prajnan 4060, 4641<br />

Dasanu, Constantin 6555,<br />

e18543<br />

Dascalu, Bogdan e19547<br />

Dascola, Isabella 2057<br />

Dasgupta, Roshni e20007<br />

Dasgupta, Swati e13030,<br />

e18043<br />

Dashk<strong>of</strong>f, Cyd 9019<br />

Daskalova, Anastassia 3594<br />

Dasoula, Aggeliki e18567<br />

Dassonville, Olivier 5521,<br />

e16044, e16051, e16055<br />

Datar, Ram H 4575, 10503<br />

Dauba, Jérôme LBA3500^,<br />

9012, e14023<br />

Daud, Adil 3058, 3102,<br />

8510, 8531, 8580,<br />

TPS8602, e13596, e19046<br />

Daugaard, Gedske e15115<br />

Daugherty, Christopher 6117,<br />

9586, e13026<br />

Daugherty, Claire 6530<br />

Dauplat, Marie-Melanie 1051<br />

Davar, Diwakar TPS8606<br />

Dave, Manish J e18117<br />

Daver, Naval Guastad 6544,<br />

6558, 6578, 6592, 6597,<br />

6607<br />

Daver, Roshni 6597<br />

Davey, Monica e19580<br />

Davi, Maria Vittoria 1604<br />

Davicioni, Elai 4565<br />

David, Alice K. TPS658<br />

David, Philippe 7057<br />

Davidenko, Irina 5504^<br />

David<strong>of</strong>f, Amy J. 6028, 6060,<br />

6075, 6131, e16519,<br />

e16537<br />

Davidson, Ashley 582<br />

Davidson, Christian 2010<br />

Davidson, Cynthia 3057<br />

Davidson, Dirk C. e11562,<br />

e13003<br />

Davidson, James Ashley 549^<br />

Davidson, John Andrew<br />

e16004, e19642<br />

Davidson, Nancy E. 1005,<br />

1021, 1027<br />

Davidson, Wendy Louise<br />

e19550<br />

Davies, Andrew John 8056<br />

Davies, Angela M. 4052<br />

Davies, Anthony 637<br />

Davies, Claire 502<br />

Davies, Faith 8015<br />

Davies, Janine Marie e14073<br />

Davies, Marianne 5529, 5532<br />

Davies, Michael A. 8501,<br />

8584, e19009, e19039<br />

Davila, Jessica 3542<br />

Davila, Marco L. TPS2619<br />

Davis, Aretha Delight 9004<br />

Davis, Brian 4595<br />

Davis, Darren W. e13500,<br />

e13562, e13575, e13577,<br />

e15124, e21028, e21029<br />

Davis, Darren e21058<br />

Davis, Gerard e13583<br />

Davis, Ian D. TPS4681,<br />

TPS4690, 8542, e15056<br />

Davis, James W. e19051<br />

Davis, John D 2567<br />

Davis, John W. 4556, e15181<br />

Davis, Keith L. 7101, e15165<br />

Davis, Mellar P. 6092<br />

Davis, Rihan 2039<br />

Davis, Samantha e11529<br />

Davis, Thomas A. TPS2103<br />

Davis, Thomas W. e13588<br />

Davis, Warren e15203<br />

Davis, Yolanda M. 4055<br />

Davison, Jon M. e14689<br />

Davol, Pam e13124<br />

Davos, Marine 2060<br />

Davoudianfar, Mina LBA4001<br />

Daw, Hamed e15023,<br />

e18046<br />

Dawar, Richa e18538,<br />

e18539<br />

Dawe, David e17518<br />

Dawood, Shaheenah S. 1032<br />

Dawsey, Scott 4506<br />

Dawson, Laura A. 4109,<br />

e14592<br />

Dawson, Nancy Ann 4, 4569<br />

Dawson, Sarah-Jane 544<br />

Day, Bann-Mo 8567<br />

Day, Fiona Lee 3623<br />

Day, Kathleen C. 4506, 4573<br />

Day, Mark L. 4506, 4573<br />

Day, Rena e15181<br />

Dayao, Zoneddy Ruiz 1558<br />

Dayes, Ian S. 6049<br />

Dayyani, Farshid e15151<br />

Dazzi, Claudio 2057<br />

Dcruz, A. K. 5585, e16021<br />

de Aguirre, Itziar 7540,<br />

10596<br />

De Albuquerque Ribeiro,<br />

Ronaldo 7506, e14183<br />

de Almeida Leite, Raphael<br />

e19528, e19584<br />

de Andrade, Jurandyr Moreira<br />

1075<br />

de Andrade, Mariza 1610<br />

de Angelis Nascimento, Maria<br />

Salete e19528, e19584<br />

De Angelis, Carmine 1554<br />

De Angelis, Verena e18101<br />

De Azambuja, Evandro<br />

e12035<br />

De Baere, Thierry 3547<br />

De Benedittis, Caterina 6531<br />

De Benedittis, Daniela<br />

e17006, e18563, e18566<br />

De Boer, Carla 3059<br />

De Boer, J. P. 8039<br />

De Bono, Johann Sebastian<br />

2530, 4513, 4515,<br />

LBA4518, 4519^, 4553,<br />

TPS4692^, TPS4693,<br />

5022, 10525, e13599,<br />

e13601, e15134, e15136<br />

De Braud, Filippo G. 3578,<br />

4507, 4629, e21118<br />

De Braud, Filippo 2595,<br />

e13606<br />

De Buck, Stefan S. 3003<br />

De Camargo Cancela,<br />

Marianna 6036, e16514<br />

De Carvalho Bittencourt,<br />

Marcelo e12506<br />

De Castro, Javier 3618,<br />

7522, 7542, TPS7612,<br />

10547, e12000, e12012,<br />

e13085, e18015, e18055<br />

De Celis Ferrari, Anezka<br />

Carvalho Rubin 6055<br />

De Censi, Andrea 519, 1500<br />

De Cicco Nardone, Carlo<br />

e15547, e15551, e15553<br />

De Conti, Ronald C. 5564,<br />

8534<br />

De Correia Pereira, Marta<br />

Isabel e12040<br />

714s Numerals refer to abstract number


De Coster, Sandra 4622<br />

De Cre, Mireille e13073<br />

De Cremoux, Hubert e18102<br />

DE Cremoux, Patricia 10507<br />

de Crespigny, Alex 2566<br />

De Faverge, Ge<strong>of</strong>froy 1574<br />

De Filippi, Rosaria 8066,<br />

8083<br />

De Filippis, Lucilla e14096<br />

DE Fraipont, Florence 10507<br />

De Geeter, Patrick 4601<br />

de Geus- Oei, Lioe-Fee<br />

e13111<br />

De Giorgi, Ugo CRA4502,<br />

TPS4683<br />

De Giorgi, Valeria 8576<br />

De Giorgi, Vincenzo 8581<br />

De Gramont, Aimery 2537^,<br />

LBA3500^, 3506, 4053<br />

De Gregorio, Nikolaus 5005,<br />

5007<br />

De Greve, Jacques 5022,<br />

9035<br />

De Groot, John Frederick<br />

2003, 2011, 2029^, 2036,<br />

2064<br />

de Groot, Steffen 1080<br />

de Gruijl, Tanja 2562<br />

de Haas, Sanne 10581<br />

de Herder, W.W. 4122<br />

De Jong, Floris Aart e16526<br />

de Jong, Igle J. 10581<br />

De Jong, Johan R 10581<br />

De Jonge, Maja J. 2593<br />

de Jongh, Felix E 10504<br />

De Juan, Ana 10079, 10512,<br />

10595<br />

de La Bourdonnaye,<br />

Guillaume 5516^, e18047<br />

de la Cruz, Juan J e11006<br />

De la Cruz, Maxine e19613<br />

de la Fouchardière, Arnaud<br />

e14730<br />

DE LA Fouchardiere,<br />

Christelle 4032, e14730<br />

De La Garza, Jaime G. 1607,<br />

e18028<br />

De la Haba Rodriguez, Juan<br />

R. TPS654^<br />

De la Haba- Rodriguez, Juan<br />

10608, 10616, e11535,<br />

e21088<br />

de la Hoya, Miguel e14147<br />

De la Mata, Dolores 7068,<br />

e19618<br />

de la Morena, Pilar e11024<br />

de la Motte Rouge, Thibault<br />

641, 6110<br />

De La Orden, Margarita<br />

e16526<br />

de la Riba Alvarez, Ines<br />

10047<br />

De la Rochefordiere, Anne<br />

e15535<br />

de la Rosa, Federico 4584,<br />

4620<br />

de la Torre, Martha e19594<br />

De Las Casas, Luis e14098<br />

de las Heras, Begona 3030<br />

de las Penas, Ramon 1531<br />

De Laurentiis, Michele 630,<br />

1073<br />

de Lavallade, Hugues 10550<br />

de Leon, Cristina 2569<br />

de Leon, Maria e15581<br />

de Liguori Carino, Nicola<br />

e21037<br />

de Lima Araujo, Luiz<br />

Henrique 7506, e17548<br />

Numerals refer to abstract number<br />

De Lima, Marcos J.G. 6529,<br />

6540, 6544<br />

De Lisa, Maria Grazia e21070<br />

De Lorenzo, Claudia e11052,<br />

e13539<br />

De Lorenzo, Francesco<br />

e19592<br />

De Los Santos, Jennifer F.<br />

e15502<br />

de Luque, V. 4630, e14115<br />

De Lutio, Elisabetta e14130<br />

De Maglio, Giovanna 3612,<br />

e18021<br />

de Maio, Ana Paula e13509<br />

De Manzoni, Giovanni 4076<br />

de Marco, Luiz e13008<br />

De Maria, Andrea 10565<br />

De Maria, Ruggero e13512<br />

De Marinis, Filippo LBA7507,<br />

7522, 7540, 7542, 7550,<br />

7557, 7558<br />

de Martino Lee, Maria Lucia<br />

TPS9594<br />

De Mattos -Arruda, Leticia<br />

10511<br />

De Mattos-Arruda, Leticia<br />

509, 566, 619<br />

De Mello, Ramon Andrade<br />

e15532<br />

De Mesmaeker, Peggy<br />

e13605<br />

de Miguel Luken, Maria Jose<br />

e16037<br />

de Miguel Luken, Veronica<br />

e16037<br />

De Miguel, Bernardo 2539,<br />

3093<br />

De Minicis, Samulele e14561<br />

de Muga, Silvia 4580<br />

De Nictolis, Michele 4084<br />

De Oronzo, Maria Antonietta<br />

5094, e15550, e15553<br />

De Pas, Tommaso M 7581<br />

De Pas, Tommaso Martino<br />

3007, 7013, 7533, 7596<br />

De Pascale, Antonella e19549<br />

De Pasquale, Maria e11001<br />

De Piaggio, Marina e16025<br />

de Pinho, Marcos Barcelos<br />

e14101<br />

De Placido, Sabino 1073,<br />

1554, e13509, e15034<br />

de Pril, Veerle TPS7113<br />

de Rink, Iris 3065<br />

de Romeuf, Christophe 5069<br />

de Roquancourt, Anne 568^<br />

De Rossi, Costanza e19595<br />

De Sanctis, Rita e16025<br />

De Santis, Maria 4539<br />

De Sarro, Giovambattista<br />

e21113<br />

De Schaetzen, Gaetan 8532<br />

De Signoribus, Giorgio 9098<br />

De Snoo, Femke 577, 1022,<br />

10545, 10554, 10597,<br />

e21013<br />

de Sousa Alves, Pedro 7013<br />

De Souza Lawrence, Lana<br />

Maria 1128<br />

De Souza, Bradley 6005<br />

De Souza, Caio Silverio<br />

e12021<br />

De Souza, Carmino 6505^,<br />

TPS9594<br />

De Souza, Paul L. LBA4518,<br />

e13585, e15152<br />

De Stefano, Alfonso e13509<br />

de Torres, Ines e21021<br />

De Troia, Beatrice 10026,<br />

10053, 10065<br />

De Vega, Josep M. 5590,<br />

5595<br />

De Velasco, Guillermo 4620,<br />

e15033, e15149, e15552<br />

De Vicente, Emilio 4040<br />

De Vincenzo, Fabio 2580,<br />

e21139<br />

De Vincenzo, Rosa 5059<br />

De Vita, Fernando 546, 3546<br />

De Vos, Sven 6518^, 8081<br />

de Vries, Elisabeth G. E.<br />

e15035<br />

De Vries, Elisabeth 4528,<br />

10581, 10602<br />

de Vries, Jolanda 9070<br />

de Vries, Petrus 4632<br />

de Waard, Jan Willem<br />

TPS10632<br />

De Wit, Maike 9033<br />

De Wit, Ronald 4570<br />

De, Pradip 626, e13579<br />

De-Montserrat, Helene 3104,<br />

e13009, e13010<br />

Deady, Sandra 2609<br />

Deal, Allison Mary TPS656,<br />

1524, 6042, 6107<br />

Deal, Ken e11000<br />

Deal, Meredith 6042<br />

Dealis, Cristina 2037<br />

Dean, Andrew Peter e14644,<br />

e14718<br />

Dean, Cheryl A. e21090<br />

Dean, Emma Jane 3004,<br />

3030, 3051<br />

Dean, James P. e15206<br />

Dean, James T 563, 6063<br />

Dean, Robert M. 8055<br />

Dean-Colomb, Windy Marie<br />

10613<br />

Deandreis, Desiree 5083,<br />

5509^<br />

DeAngelis, Carlo e15180<br />

DeAngelis, Lisa Marie 2006,<br />

2008<br />

DeAngelo, Daniel J. 6526,<br />

6621<br />

DeAngelo, Daniel J 6503,<br />

8043<br />

Debernardi, Felicino e20512<br />

DeBernardo, Robert 5102<br />

Debiec-Rychter, Maria 9536,<br />

10030, 10538<br />

Debieuvre, Didier 1574,<br />

TPS7111<br />

Deblasio, Weiping Xu 8547<br />

Debled, Marc 601, TPS667<br />

Debois, Muriel 7013<br />

Debourdeau, Philippe 1580,<br />

e13062<br />

Debra, Rowett 2604<br />

Debruyne, Philip R. 4622<br />

Debska, Sylwia e21099<br />

Deburr, Tawania L 8514,<br />

e19009, e19014<br />

Debus, Jürgen 2034, e16033<br />

DeCamp, Malcolm M. 7040,<br />

7054, 7055<br />

Decano, Julius L e13574<br />

Decarli, Nicola L e21102<br />

DeCastro, Guarionex e15018,<br />

e15019, e15021<br />

Decatris, Marios P. e15030<br />

Decaux, Olivier 8014<br />

Deck, Kenneth B. 10554<br />

Decker, Brian 9095, e12500<br />

Decker, David A. 9095<br />

Decker, Paul A. 4062, 7008<br />

Decker, Paul D e12500<br />

Decker, Roy H. 5529, 5532<br />

Decker, Thomas 3588<br />

Deconinck, Eric 6542,<br />

e19623<br />

Decordova, Shaun 3022<br />

Decoster, Lore 9035<br />

Decosterd, Laurent e11048<br />

DeCourtney, Christine 6126<br />

Dedieu, Jean-Francois<br />

e15116<br />

Dediu, Mircea LBA7507<br />

Deeb, George 6565<br />

Deeg, H Joachim 6522<br />

Deeken, John F. 2590, 3095<br />

Dees, Elizabeth Claire<br />

TPS656, 9020, 10515,<br />

e13596<br />

DeFord-Watts, Laura M 6524^<br />

Defrein, Anne Marie 615<br />

Deftereos, Savas e14567<br />

Degendorfer, Pamela 3013<br />

Degenhardt, Tom 552<br />

Degheidy, Heba A 6581<br />

Degiovanni, Daniela 7084<br />

Degnim, Amy C. 10586<br />

Degos, Francoise 4033<br />

Deguiral, Philippe e14148<br />

DeGuzman, Angel e13583<br />

Dehesdin, Daniele 5565<br />

Dehghan-Paz, Irene 572<br />

Dehle, Francis 7604<br />

Dehner, Louis P 9521, 9522<br />

Dei Tos, Angelo Paolo 10009,<br />

10053, 10063<br />

Deighan, Clayton e21124<br />

Deininger, Michael W.N.<br />

6624<br />

Deitel, Wayne 6027<br />

Dejardin, David 6504, 6506<br />

DeJesus, Yvette e16574,<br />

e19625, e19629<br />

Dejoie, Thomas e21117<br />

DeJoubner, Nutan Jyothi<br />

e21045<br />

Dekeyser, Josh 3580<br />

DeKoven, Mitch 570<br />

del Barco, Sonia e11545,<br />

e19542<br />

Del Bene, Gabriella e14096<br />

del Carmen, Marcela G 5029,<br />

e15545, e15568<br />

del Cerro, Elia 1111<br />

Del Conte, Gianluca 3080<br />

Del Curatolo, Anais e13512,<br />

e13572<br />

Del Fabbro, Egidio 9062,<br />

9127, e19528, e19584,<br />

e19643<br />

Del Giglio, Adriana Braz<br />

e19552, e21151<br />

Del Giglio, Auro 5588,<br />

10568, e11059, e13016,<br />

e16568, e19537, e19552,<br />

e21151<br />

Del Marmol, Veronique<br />

e19026<br />

Del Mastro, Lucia TPS653,<br />

1073<br />

Del Prete, Michela e14086,<br />

e14561, e14663, e15074<br />

Del Priore, Giuseppe e15514,<br />

e15561<br />

Del Re, Marzia e13057<br />

Del Rio, Elisabeth e18057<br />

Del rio, Maguy 10505<br />

Del Vecchio, Michele 8513,<br />

8517<br />

Delabie, Jan 8074<br />

715s


DeLacure, Mark D. e16052<br />

Delage, Judith e21156<br />

Delaloge, Suzette TPS652,<br />

1011, 1091, 1601<br />

Delande, Stephanie 4622<br />

Delaney, Colleen 6631<br />

Delaney, Joseph e21159<br />

DeLaney, Thomas F. 4021,<br />

10058, e16059<br />

Delarochefordiere, Anne<br />

e15524<br />

Delarue, Richard 8021, 8063<br />

Delasalle, Kay 8093<br />

Delattre, Jean-Yves 2<br />

Delattre, Olivier 9510<br />

Delaunay, Jacques 6523,<br />

6559, 6632<br />

Delaunoit, Thierry 3559,<br />

e14060<br />

Delavault, Patrick TPS4153<br />

Delbecque, Laure e12515<br />

Delbeuck, Xavier e11012<br />

Delea, Thomas E. e11067<br />

Delee, Maxon 7601<br />

deLeon, Maria 5099<br />

Deleporte, Amélie 3559<br />

Delfanti, Sara e13103<br />

Delgado, Francois-Michel<br />

6110<br />

Delgado, Juan Ramon 1041<br />

Delgado, Lucia Beatriz<br />

e16505, e19007<br />

Delgado, Salvadora 3536,<br />

3586<br />

Delgado-Guay, Marvin Omar<br />

9002, 9063<br />

Delios, Anna Maria 2028<br />

Delioukina, Maria L. 6545<br />

Delisle, Marie-Bernadette<br />

10076<br />

Deliveliotis, Charalambos<br />

e15199^<br />

Dell’ Aglio, Carmine 10066<br />

Dell’Orto, Patrizia 1022<br />

Dell, Sharon 9591<br />

Della Puppa, Alessandro 2049<br />

Dellinger, Andrew e13589,<br />

e21038<br />

Delman, Keith A. 8543<br />

Delmar, Paul 5522<br />

Delmore, James Edwin 5015<br />

DeLongis, Susan e19588<br />

Delord, Jean-Pierre 2605,<br />

3026, 3104, 5516^, 5522,<br />

e13009, e13010, e13594<br />

Delorenzi, Mauro 3509,<br />

3511, 3575, 9510<br />

Delourme, Julie e18066<br />

Delpech, Yann 612<br />

DelPriore, Guiseppe 5107<br />

Delrio, Paolo e14130<br />

Deluca, Jill 7040<br />

Delva, Remy LBA4567^,<br />

4625<br />

Delwail, Vincent 2023<br />

Demanse, David 508, 3003,<br />

e13605<br />

Demarchi, Martin LBA4007<br />

Demas, William F. TPS9150<br />

DeMatteo, Ronald P. 3577,<br />

3620<br />

DeMers, April e14503<br />

Demers, Brigitte 3080<br />

Demers, Eric 634<br />

Demetri, George D. 1547,<br />

LBA10008, 10010, 10013,<br />

10061, 10067, 10097,<br />

TPS10101<br />

Demeure, Michael J. 4013<br />

Demian, Gerges Attia 9566<br />

Demicco, Elizabeth G 10071<br />

DeMichele, Angela e19531<br />

Demidov, Lev V. 2524,<br />

LBA8500^, LBA8509,<br />

e19005<br />

DeMie, Lynn e13581<br />

Deming, Dustin A. 3103<br />

Deming, Richard L. e16572<br />

Demir, Gokhan e14698,<br />

e19024<br />

Demir, Lutfiye 638<br />

Demirag, Gà1 ⁄4zin e14107<br />

Demirdjian, Sylvie 9572<br />

Demirelli, Fuat e21143<br />

Demiri, Stamatina e11519<br />

Demirkan, Hatice Mirac<br />

Binnaz e11507<br />

Demirkazik, Ahmet 1572,<br />

1596<br />

Demkow, Tomasz 4606<br />

Demmy, Todd L. 1529,<br />

e17535<br />

Demolin, Gauthier 3559<br />

DeMoor, Patricia A e16535<br />

Demura, Yoshiki 7561<br />

Demuth, Tim e13598<br />

den Hollander, Martha<br />

Willemine 4132<br />

Den, Robert Benjamin<br />

e15012, e19507<br />

Denda, Tadamichi TPS3642,<br />

e14510<br />

Denduluri, Neelima TPS1148<br />

Denehy, Linda e19605<br />

Deng, Jie e16038<br />

Deng, Li e16017<br />

Deng, Qiaolin 10539<br />

Deng, Qiqi e13011<br />

Deng, Wei 5110<br />

Deng, Xingming 7059, 7097<br />

Deng, Yanhong 6033,<br />

e14153, e21106<br />

Deng, Yongchuan 9017<br />

Deng, Zhuoxia 5558<br />

Dengel, Lynn Thacher e19062<br />

Dengler, Jolanta 6510<br />

Denham, Fiona e19008<br />

Denholm, Katie Ann 2530,<br />

10525<br />

Denic<strong>of</strong>f, Andrea 9047,<br />

9144, e16561, e16569,<br />

e19633<br />

Denis, Marc G. 2082,<br />

e21117<br />

Denisova, Elena e15025<br />

DeNittis, Albert S. e14157<br />

Denitz, Jordan e16026<br />

Deniz, Ferhat 4641<br />

Deniz, Kemal e14089,<br />

e14670<br />

Denkert, Carsten 522, 523,<br />

1023, 4020, 10627,<br />

e15531<br />

Denlinger, Crystal Shereen<br />

TPS3111<br />

Denmeade, Samuel R. 4550,<br />

4559<br />

Denoux, Yves 543<br />

Denslow, Sheri 5063<br />

Dent, Louise 8056<br />

Dent, Rebecca Alexandra<br />

1032, 9041<br />

Dent, Susan Faye 587<br />

Dent, Susan 1036<br />

Denton, Brian 10015<br />

Denzlinger, Claudio TPS3639<br />

Deo, S. V. S e11013, e17505<br />

Deonarain, Mahendra e13525<br />

Depenni, Roberta 2057<br />

Depierre, Alain 7057<br />

Deplanque, Gael LBA4007,<br />

LBA4567^, 4583, 9011<br />

Deppen, Stephen A 7008<br />

DePriest, Carol 2569<br />

Der, Sandy 10522<br />

Deray, Gilbert 1095, 5067<br />

Derbel, Olfa e14730<br />

Derdel, Jerry TPS9150<br />

Derenne, Laurent e14148<br />

Dering, Judy 8592<br />

Derks, Maarten e15166<br />

Derollez, Marc TPS7111<br />

Deroma, Laura e15091<br />

DeRoos, Anneclaire 1527,<br />

6560<br />

Derr, Alan G 1057<br />

Dervenis, Christos e14625<br />

Des Guetz, Gaetan e18003,<br />

e18020<br />

Desai, Ami V. 9576<br />

Desai, Amishi e18554,<br />

e21114<br />

Desai, Anant e13507<br />

Desai, Arati Suvas e16062<br />

Desai, Bhardwaj 2013<br />

Desai, Jayesh 3623, e13054,<br />

e16516<br />

Desai, Mihir e15060, e15171<br />

Desai, Neelam Vijay<br />

TPS4136, e14612<br />

Desai, Palak 572<br />

Desai, Pankaj B TPS3116<br />

Desai, Sangeeta 1108<br />

Desai, Sonal 7582, 7594,<br />

e13526<br />

Desal, Hubert 2082<br />

Desantis, Mariangela e15574<br />

Desantis, Mariangella 5075<br />

Desar, Ingrid 5061<br />

Desbiens, Christine TPS1139<br />

Deschler, Daniel G. e16059<br />

Deshmukh, Chetan Dilip<br />

2592, e13127<br />

Deshmukh, Sanjay P e11506<br />

Deshpande, Hari Anant 5529,<br />

5532<br />

Deshpande, Rahul e21037<br />

Deshpande, Vikram 4021<br />

Desideri, Giovambattista<br />

e11041<br />

DeSilva, Ashley A 2026<br />

DeSilvio, Michelle TPS658<br />

Desjardins, Annick 2027,<br />

2074^, 2090^, 2094^,<br />

2095^, TPS2103<br />

Desjardins, Laurence 8546,<br />

9538, e13017<br />

Desmedt, Christine 515,<br />

e21010<br />

Desmoulins, Isabelle 10062<br />

Desolneux, Gregoire e14017<br />

DeSon, Amanda 4102<br />

Desruennes, Eric 1580<br />

Dessanti, Paolo e17533<br />

Desseigne, Françoise e14730<br />

Dessi’, Mariele 9006<br />

Dessureault, Sophie 2559<br />

Dessypris, Nick e20002<br />

Desta, Zeruesenay 525<br />

Destaillats, Alice 3026<br />

Destro, Anna e18104<br />

Destro, Annarita 7061, 7585<br />

Desutter, Petra 1097<br />

Detjen, Katharina M 4128<br />

Dettino, Aldo A. e14728,<br />

e18078<br />

Detweiler-Short, Kristen 8011<br />

Detzler, Taylor 4591<br />

Deutsch, Alexander Josef<br />

e18525<br />

Deutsch, Eric 3063<br />

Deutsch, Jeremy Michael<br />

3056<br />

Dev, Rony 9062, 9127,<br />

e19528, e19584, e19643<br />

Deva, Sanjeev e15054<br />

Devanarayan, Viswanath<br />

e13583<br />

Devarajan, Karthik 1550<br />

Devery, Aoife 2536<br />

Devesa, Manuel e21104<br />

Devgan, Vikram e21011<br />

Devi, Gayathri 2585<br />

Devidas, Meenakshi 9504,<br />

CRA9508, 9537, 9543,<br />

9546, 9548<br />

Deville, Anne e20006<br />

Devin, Etienne TPS7111<br />

Devine, Katie 9113<br />

Devine, Steven M 2533<br />

Devins, Gerald M. 2063<br />

Devito, Vikki 7078<br />

Devitt, Bianca Alix e16562<br />

Devlin, Sean 6532<br />

Devlin, Susan 6508<br />

Devore, Nathan 1569<br />

Devos, Annick 9517<br />

Devos, Patrick 2070<br />

DeVries, Todd e15120<br />

Dewas, Sylvain e14560<br />

Dewdney, Alice e14088<br />

Dewhirst, Mark W e21119<br />

Dewi, Sagita e11022<br />

Dewitt, William 10516<br />

Dewji, Mohamedraza 10009,<br />

10067<br />

Dey, Jyotirmoy 9519<br />

Dey, Nandini 626, e13579<br />

DeYoung, Barry e15580<br />

Dezentje, Vincent O. 526<br />

Dezerlle, Martine 9080<br />

Dhage, Shubhada e11064<br />

Dhakal, Binod e17555<br />

Dhalluin, Christophe<br />

TPS9597^<br />

Dhalluin, Xavier e18066<br />

Dhanda, Rahul e15063,<br />

e15089, e20510<br />

Dhangada Majhi, Prabin<br />

e17549<br />

Dhani, Neesha C. e14535<br />

Dhanvijay, Sushant e15011<br />

Dhar, Arindam TPS2614<br />

Dharsee, Moyez 10622<br />

Dhermain, Frederic 3063<br />

Dhesy-Thind, Sukhbinder K.<br />

6049, e14001<br />

Dhillon, Haryana M. 9021<br />

Dhillon, Navneet 8543<br />

Dhinakaran, Mullai e19622<br />

Dhindsa, Navreet TPS663<br />

Dhobe, Poornima 3011, 3013<br />

Di Bacco, Alessandra 3077,<br />

8017, 8033, 8034, 8064,<br />

e13603<br />

Di Bartolomeo, Maria<br />

LBA4001, e21118<br />

Di Battista, Monica 2061<br />

Di Biasi, Brunella e14018<br />

Di Blasi, Concetta e15544<br />

Di Cocco, Barbara e19501<br />

Di Cosimo, Serena 619, 1011<br />

Di Costanzo, Francesco<br />

LBA4001, e18074<br />

Di Costanzo, Giovan Giuseppe<br />

TPS4151<br />

716s Numerals refer to abstract number


Di Emma, Veronica e19002<br />

Di Fabio, Francesca LBA4001<br />

Di Filippo, Franco 1050<br />

Di Filippo, Simona 1050<br />

Di Giacomo, Anna Maria<br />

8513, 8529<br />

Di Giacomo, Daniela e14010<br />

Di Giannatale, Angela 9517<br />

Di Lascio, Simona e14156<br />

Di Lecce, Valentina e21113<br />

di Legge, Alessia 5059<br />

Di Leo, Angelo 1008, 1012<br />

Di Lorenzo, Giuseppe<br />

e15034, e15043, e15112,<br />

e15185<br />

Di Loreto, Carla e15091<br />

Di Lucca, Giuseppe 4017<br />

Di Lullo, Gloria A. 6135<br />

Di Maio, Massimo TPS4151,<br />

e18021, e19592<br />

Di Marco, Marco e14096<br />

Di Marco, Mariacristina<br />

e14647<br />

Di Narzo, Antonio Fabio<br />

3509, 3575<br />

Di Pace, Raffaella 1500<br />

Di Palma, Mario 4554, 4621,<br />

e15049<br />

Di Raimondo, Francesco 8006<br />

Di Salvatore, Mariantonietta<br />

e14156<br />

Di Sanza, Cristina e13572<br />

Di Stasi, Savino Mauro 4572<br />

Di Stefano, Mirella 5059<br />

Di Tommaso, Luca 623<br />

Di, Genhong e11567<br />

Di, Yang e14651<br />

Diaconu, Iulia e13035<br />

Diamond, Jennifer Robinson<br />

3017<br />

Diane, Goere e14158<br />

Diao, Lixia 7096, 10612<br />

Dias, Lauren Elizabeth<br />

e14615<br />

Dias, Nathalie 9517<br />

Dias-Santagata, Dora 7532<br />

Diasio, Robert B. 3564,<br />

3617, 4009, 4075<br />

Diaz Paniagua(1), Laura<br />

e11543, e19596<br />

Diaz Pena, Elena e16037<br />

Diaz Rubio, Eduardo 10564<br />

Diaz, Angelica 2587<br />

Diaz, Enrique Gallardo<br />

TPS4672<br />

Diaz, Gema e17503<br />

Diaz, Jose 10061<br />

Diaz, Jr., Luis A. 3068<br />

Diaz, Luis A. 2517, 3016,<br />

3596, 3616, 4045, 4114<br />

Diaz, Nieves e11545, e15144<br />

Diaz, Robert 10082<br />

Diaz, Zuanel TPS1139,<br />

e14108<br />

Diaz-Flores, Ernesto 6582<br />

Diaz-Gavela, Ana 1111<br />

Diaz-Montero, C Marcela<br />

9565<br />

Diaz-Padilla, Ivan 5019<br />

Diaz-Rubio, Eduardo e14147,<br />

e20513<br />

Dib-Faria, Luiza e14702<br />

DiBenedetto, Joseph e19615<br />

DiCarlo, John e21011<br />

Dicato, Mario-A e14180<br />

Dichmann, Robert 8098<br />

Dicker, Adam P. 4041,<br />

e11505<br />

Numerals refer to abstract number<br />

Dicker, Adam e15012,<br />

e19507<br />

Dicker, David e11544<br />

Dickinson, Kim e21111<br />

Dickinson, Kimberly M. 5040<br />

Dickler, Maura N. 575,<br />

10514<br />

Dickman, Eileen D. e13568<br />

Dickow, Brenda e15081<br />

Dickson, Mark Andrew 8549,<br />

8575, 8598, 10002,<br />

10004, 10019, TPS10103<br />

Dickson, Mike 9143<br />

Die Trill, Javier e21104<br />

Diebold, Marie Danielle<br />

e14059<br />

Diebolder, Herbert e15566<br />

Dieci, Maria Vittoria 631<br />

Dieckmann, Karin 2071<br />

Dieleman, Edith 7019<br />

Dienstmann, Rodrigo 509,<br />

566, 2567, 2568, 3014,<br />

e13048<br />

Dieras, Veronique LBA1,<br />

TPS662, e13017<br />

Dierckx, Rudi A 10581<br />

Dierickx, Daan 8030<br />

Dietel, Manfred e18016^<br />

Dieterle, Nelli 10054<br />

Dietlein, Markus 3044, 7603,<br />

8079<br />

Dietrich, Daniel 10033<br />

Dietrich, Mary S. e16038,<br />

e19559<br />

Dietrich, Sascha 6519,<br />

10040<br />

Dietz, Allen 2546<br />

Dietz, Andreas e16016<br />

Dietzfelbinger, Hermann F.<br />

3588<br />

Dieudonné, Anne-Sophie<br />

1020<br />

Diez, Blanca D. e12511<br />

DiGianni, Lisa 1547<br />

DiGiovanna, Michael 1045<br />

Dignan, Mark e11097,<br />

e12004<br />

Digumarthy, Subba 7524<br />

Digumarti, Raghunadharao<br />

6554, e16505, e18544,<br />

e18545<br />

Dikalioti, Stavroula e20002<br />

Dilawari, Asma Ali e11010<br />

Dileo, Palma 10063<br />

Diller, Lisa CRA9513<br />

Dillies, Anne-Francoise 5505<br />

Dilling, Thomas J. 7065,<br />

e17559<br />

Dillinger, Jennifer 9132<br />

Dillman, Robert Owen 2569<br />

Dillon, Patrick Michael<br />

e19028<br />

Dilts, David M. 6047, 6051,<br />

e16569<br />

Dimitriadis, George e21027<br />

Dimitrov, Todor e13505<br />

Dimitrova, Desislava 5053,<br />

5070<br />

Dimitrova, Nevenka 10508<br />

Dimond, Eileen P. 6086,<br />

e16561<br />

Dimopoulos, Meletios A.<br />

5033, 8018^, TPS8112,<br />

TPS8113, e15040,<br />

e18572^<br />

Dimopoulos, Prokopios<br />

e14567<br />

Dinan, Michaela A 1525<br />

Dinardo, Courtney Denton<br />

6579<br />

Dincaslan, Handan e20000<br />

Dincer, Murat e21050<br />

Ding, Hao e18088<br />

Ding, Ke-Feng e14024<br />

Ding, Keyue TPS2104, 7511<br />

Ding, Li 503, 9518<br />

Ding, Michelle e13600^<br />

Ding, Robert Phooi Huat<br />

4106<br />

Ding, Wei e13517<br />

Ding, Xian e14642<br />

Ding, Xiaoyan e11538<br />

Ding, Yan 4646<br />

Ding, Yi tao e14509<br />

Ding, Yi 8589<br />

Ding, Zhenyu e18086<br />

Dingemans, Anne-Marie C.<br />

TPS7612<br />

Dingli, David 8092, 8096,<br />

8105, TPS8116, e21006<br />

Dinh, Yvonne 8040, 8102<br />

Dinh<strong>of</strong>, Carina 2053<br />

Diniz, Alessandro Landskron<br />

e14728<br />

Dinjens, Winand N.M. 2<br />

Dinkel, Andreas e15157<br />

Dinkel, Carina 554<br />

Dinney, Colin P.N. 4523<br />

DiN<strong>of</strong>ia, Amanda 9587<br />

Diodati, Francesca 9120<br />

Diong, Colin e18528<br />

Dionigi, Paolo e13103<br />

Diorio, Caroline 634<br />

Diorio, Dawn TPS655<br />

Dip Borunda, Abdel Karim<br />

e15567<br />

DiPaola, Robert S. 2526,<br />

4500<br />

DiPasquale, MariaChiara<br />

e14711, e21102<br />

DiPersio, John F. 6624<br />

DiPersio, John F 6503<br />

Dirix, Luc Yves 516, 517,<br />

e13605<br />

Dirix, Luc 532, 2583, 8579,<br />

10504<br />

Dirksen, Uta 10080<br />

Dirlea, Mihaela 9131<br />

Dirven, Hubert 6619^<br />

Disalvatore, Davide 546<br />

Disci, Rian e14628<br />

DiSiena, Michael Ross<br />

e13561<br />

Disilvestro, Paul 1519<br />

DiSipio, Theresa C. 9576<br />

Disis, Mary L. 5082^<br />

Diskin, Sharon 9516<br />

Dispenzieri, Angela 8043,<br />

8092, 8096, 8105,<br />

TPS8116<br />

Dispersyn, Garance TPS9154,<br />

e14006<br />

Dito, Elizabeth 4048<br />

Ditsch, Nina 1114<br />

Ditto, Antonino 5096<br />

Dive, Caroline e14015,<br />

e15141, e21037<br />

Divekar, Ganesh 2592,<br />

e13127<br />

Diwakar, Ravi B 1093,<br />

e11511, e12030, e14721<br />

Dixon, Margie e13026<br />

Dizdar, Omer e11011,<br />

e11023, e11026, e11045,<br />

e11515, e14174, e15572,<br />

e15573<br />

Dizdar, Yavuz 9567<br />

Dizon, Don S. 5100<br />

Djaladat, Hooman 4576,<br />

4589<br />

Djanani, Angela e14719<br />

Djordjevic, Bojana 1513<br />

Dmitriev, Igor e13087<br />

Dmitrovsky, Ethan 7022,<br />

e13567<br />

Do, An 5575<br />

Do, Chuong B 10097<br />

Do, Ingu e13094<br />

Do, Khanh Tu 3087, 3529<br />

Do, Kim-Anh 1054, e15092<br />

Do, Young Rok e14570<br />

Doat, Solene e14065<br />

Dobbin, Zachary C. 5036<br />

Dobi, Albert e15207<br />

Dobi, Erion e11019<br />

Dobrilla-Dintinjana, Renata<br />

9097<br />

Dobrovic, Alexander 8520<br />

Dobs, Adrian e19582<br />

Dobson, Lisa TPS8605<br />

Dobson, Nicky 9589<br />

Dockx, Yanina 1129<br />

Dodd, Anna 9021<br />

Dodion, Pierre F. 10010<br />

Dodson, Shontelle e19582,<br />

e19585<br />

Dodwell, David John 502<br />

Doebele, Robert Charles<br />

7504, 7526, 7531, 7534,<br />

7601, e21120<br />

Doehn, Christian e15044<br />

Doerfel, Steffen TPS3641^,<br />

5031<br />

Dogan, Semih 1601<br />

Dogan, Snjezana 10524<br />

Dogan, Yasemin TPS4138<br />

Doger de Speville, Bernard<br />

Gaston e11035, e11527,<br />

e12015, e17502, e17503,<br />

e17515, e18521<br />

Dogliotti, Luigi e15185<br />

Doherty, Gerard M 6056<br />

Doherty, J. E. e14022<br />

Doherty, Mark e17561<br />

Dohner, Hartmut 6567<br />

Dohner, Konstanze 6567<br />

Dohollou, Nadine 5018<br />

Dohrenwend, Eric 10002<br />

Doi, Ryuichiro e14506<br />

Doi, Seiji 7569<br />

Doi, Toshihiko 3079<br />

Doihara, Hiroyoshi 1119,<br />

TPS1147<br />

Doimi, Franco F. 1024<br />

Doimi, Franco e11066<br />

Dokmak, Safi 3609<br />

Dolan, James e15138<br />

Dolan, M. Eileen 9516<br />

Dolinsky, Jozef e19020<br />

Doll, Donald C. e17525<br />

Dollinger, Matthias M. 4115<br />

Doll<strong>of</strong>f, Nathan G 8090<br />

Domanska-Czyz, Katarzyna<br />

e18512<br />

Domas, Jean TPS7111<br />

Domchek, Susan M. 1506<br />

Domenech, Montserrat<br />

TPS4674, e15144<br />

Domenichini, Enzo e14598<br />

Domerg, Caroline 7007<br />

Domine, Manuel 1531, 5520,<br />

7095, e15094, e18011,<br />

e18015, e18055, e18069,<br />

e18106, e21015<br />

Domingo, Gelenis C. 6608<br />

717s


Domingo, Gelenis 6568,<br />

6575, 6588<br />

Domingues, Pedro Masson<br />

e17548<br />

Dominguez Franjo,<br />

Purificacion e11028<br />

Dominguez Hormaetxe, Saioa<br />

e21089<br />

Dominguez, Rafael 2515<br />

Dominici, Fernando e19569<br />

Dominioni, Lorenzo 1532<br />

Domino, Marisa E 6029<br />

Domling, Alexander 8577<br />

Dommermuth, Alena e14131<br />

Domont, Julien 3063, 10005,<br />

10007, 10016, 10020,<br />

10027, 10033, 10044,<br />

10089, 10092, 10095<br />

Donachie, Paul 9539<br />

Donahue, Amy 3533<br />

Donahue, Timothy R. e14582<br />

Donald, Emma 1548<br />

Donaldson, Kirsteen 3100<br />

Donaldson, Sarah S. 9502,<br />

9537<br />

Dondero, Richard TPS8116<br />

Dondi, Alessandra 8006<br />

Done, Susan 1019, 1123<br />

Donehower, Ross C. 2535,<br />

2594, 3020, 3616, 4005,<br />

4055<br />

Donepudi, Sreekanth 10614<br />

Dong, Bin 538<br />

Dong, Hongshan e21080<br />

Dong, Mei e14662<br />

Dong, Song e13510<br />

Dong, Wei 1590, 1591,<br />

4092, e14575<br />

Dong, Yu e18038<br />

Dongre, Amol 9559, e17003<br />

Donia, Marco 2565, 2574<br />

Donini, Maddalena 4097<br />

Donnadieu, Anne e11070<br />

Donnellan, Paul P. LBA1000,<br />

e19059<br />

Donnelly, Bryan J 4<br />

Donnelly, Kerri e13588<br />

Donnelly, Patrick E. 6031<br />

Donner, Allan 9003<br />

Donnini, Alessia e15011<br />

Donovan, Jenny TPS665<br />

Donovan, Sean 8590<br />

Donskov, Frede 4536, 4538,<br />

e21110<br />

Doolin, Elizabeth E. 4603,<br />

7079^<br />

Doolittle, Nancy Diane 2040,<br />

2077<br />

Doot, Robert K 1088<br />

Dorantes, Yuzmiren e19618<br />

Doratotaj, Behzad e21082<br />

Dorca Ribugent, Joan 1001<br />

Dorent, Richard 1095, 5067<br />

Dorff, Tanya B. e13084,<br />

e13531, e15005, e15153<br />

Dorff, Tanya B TPS4679<br />

Dorin, Ryan Paul 4588<br />

Dorkhom, Stephan Joseph<br />

6544, 6607<br />

Dorkin, Maurizio e19002<br />

Dorlhiac-Llacer, Pedro E<br />

6505^<br />

Dormans, John P. 9537<br />

Dorn, Paige L. e11512<br />

Doros, Leslie Ann 9521,<br />

9522<br />

Dorosario, Andrew 8552<br />

Doroshow, James H. 3031,<br />

3087, 3529, 5020, 10603<br />

Dorta, Javier 1531<br />

Dorta, Miriam e15033<br />

Dorvault, Nicolas 10556<br />

dos Santos, Renata e19528,<br />

e19584<br />

Dosa, Edit 2040, 2077<br />

Doshi, Sameer 2535, 2594<br />

Doshi, Simit e15181<br />

Dotan, Efrat e14021<br />

Dotson, Emily K. e18508<br />

Dotti, Katia F e21118<br />

Doubet, Ellen e15012<br />

Douce, Thomas B e13581<br />

Douek, Michael 1131<br />

Douer, Dan 6532<br />

Dougherty, Patrick M. 9082,<br />

9140<br />

Douglas, Jeremy 9019<br />

Douillard, Jean Yves e14148<br />

Douillard, Jean-Yves 3507,<br />

3581, LBA4003, 7007,<br />

7013<br />

Doung, Yee-Cheen 10011^<br />

Dourthe, Louis-Marie e11019<br />

Doussau, Adelaide 4053<br />

Doval, Dinesh e15082<br />

Dove, Lorna 4101<br />

Dowden, Scot D. e14073<br />

Dowdy, Sean Christopher<br />

5004, 5087<br />

Dowell, Barry e13583<br />

Dowell, Jonathan 7518<br />

Dowlati, Afshin 7087,<br />

e17563, e18046<br />

Dowling, Ryan JO 1019<br />

Downing, James 9518<br />

Downing, Sean 3533, 4649<br />

Downs, Melinda 3020,<br />

TPS3117<br />

Dowsett, Mitchell 9103<br />

Doyle, Catherine 3572<br />

Doyle, L. Austin 4639, 6582,<br />

8598, 9541, 9581,<br />

e14586<br />

Doyle, Laurence A. 8075<br />

Doyle, Laurence A 3020,<br />

3103, 3529<br />

Doyle, Yvonne e15534<br />

Doz, Francois P. 9538<br />

Drabick, Joseph J. 8090<br />

Dracopoli, Nicholas J e17541<br />

Draetta, Giulio Francesco<br />

e15200<br />

Dragnev, Konstantin H. 7022,<br />

e14514<br />

Drake, Catherine 2062,<br />

e16506<br />

Drake, Charles G. CRA2509,<br />

TPS2613, 4505, TPS4689,<br />

5506<br />

Drakes, Maureen e13121<br />

Dranitsaris, George e18562,<br />

e18567<br />

Dran<strong>of</strong>f, Glenn 2502<br />

Drape, Jacqueline 8060<br />

Drapkin, Benjamin Jacob<br />

6617, 6618<br />

Drappatz, Jan 2009<br />

Drayson, Mark 8015<br />

Dreger, Peter 6543, 8004<br />

Dreher, Cara 1512<br />

Dreicer, Robert 4609, 4650,<br />

4662, TPS4684, TPS4693,<br />

e15095, e15208<br />

Drengler, Ronald L. e13025<br />

Drengler, Ronald 2512, 2555<br />

Dreno, Brigitte 8559, 8591,<br />

e19019<br />

Dresser, Mark J. e13114<br />

Dressler, Lynn G. 6042<br />

Drevs, Joachim e13508,<br />

e13597<br />

Drew, Yvette 3048, 3051<br />

Dreyer, Chantal 2557,<br />

e14553, e14660<br />

Dreyer, Nancy A. e11051<br />

Dreyer, ZoAnn Eckert 9548<br />

Dreyfus, Brigitte 6584<br />

Dreyfus, Francois 6523<br />

Dreyling, Martin H. 8030,<br />

8031<br />

Drilon, Alexander Edward Dela<br />

Cruz 7052, 7066, 7527,<br />

7578, 7580<br />

Driol, Pamela 1044, e13058,<br />

e13070<br />

Drobish, Amy 10515<br />

Drobniene, Monika 9046<br />

Droege, Cornelia M. e18012<br />

Drolsum, Anders e14637<br />

Drooz, Alain 3590<br />

Dropulic, Boro TPS1616<br />

Drosik, Kazimierz 4606<br />

Drouet, Alain 10049<br />

Drouet, Ludovic e21156<br />

Drouin, Michel A. 3039,<br />

e13602, e13604<br />

Drouin, Sarah e15105<br />

Drovetto, Nicholas Alan<br />

e21148<br />

Droz, Jean Pierre 5028,<br />

e15009<br />

Droz, Jean-Pierre e15122<br />

Drozdowskyj, Ana 7522,<br />

7540, 7542<br />

Drucker, Aaron Mark 9088<br />

Druker, Brian J. 9555<br />

Drullinsky, Pamela 8054<br />

Drumm, William e13542<br />

Dry, Sarah M 10012, 10588<br />

Du Bois, Andreas 5005,<br />

5007, 5051<br />

Du Four, Stephanie 2050,<br />

e19026, e19027<br />

du Manoir, Jeanne Michelle<br />

e15020<br />

Du, Feng e14606<br />

Du, Min e14593<br />

Du, Nan 7036<br />

Du, Weidong e18088<br />

Du, Xiang 3551<br />

Du, Yuhong 3094<br />

Dua, Rajesh 2585<br />

Duan, Jianchun 7537,<br />

e18022, e18035<br />

Duan, Qiuli 6548<br />

Duan, Xiuzhen e15538<br />

Duan, Zhigang 3542<br />

Duarte, Jose 4584<br />

Duarte, Lupe 8089<br />

Dubel, Gregory 4098<br />

Dubey, Sarita 2535, 2594,<br />

4005<br />

Dubinett, Steven M. 7518,<br />

7532<br />

Dubois, François 2070<br />

DuBois, Kara S e14566<br />

DuBois, Steven G. 3058,<br />

9537<br />

Dubost, Emilie e19555<br />

Dubovsky, Jason A. 6575<br />

Dubowy, Ronald L. 3019,<br />

e13576<br />

Dubreuil, Olivier 4018<br />

Dubrow, Robert 1591<br />

Dubsky, Peter Christian 514,<br />

542, 10570<br />

Ducassou, Marie 10042<br />

Duch, Joan 1120<br />

Ducharme, Murray P 6619^<br />

Duchesne, Gillian M.<br />

TPS4690<br />

Duchnowska, Renata 505,<br />

603, e21099<br />

Ducimetiere, Francoise 10056<br />

Duckworth, Lizette Vila<br />

e14612<br />

Ducolone, Alain 7057<br />

Ducreux, Michel 3507, 3535,<br />

3547, 4032, 4036, 4053,<br />

4071, e14129<br />

Duda, Dan G. 1026, 2010,<br />

4112<br />

Duddalwar, Vinay 4096<br />

Dudgeon, Deborah Jane 6039<br />

Dudley, Mark E. 8576,<br />

e14179<br />

Dudnik, Elizabeth 9052<br />

Dudnitchenko, Alexander Serg<br />

e13095^, e13097^,<br />

e13108^<br />

Dueck, Amylou C. 6108,<br />

6133, 8010, 8053, 9047<br />

Duehrsen, Ulrich 8030<br />

Dueland, Svein 4015<br />

Dueñas Porto, Marta 4584<br />

Duerinck, Johnny 2050,<br />

e19027<br />

Duerk, Heinz A. 3<br />

Duffaud, Florence 10005,<br />

10006, 10023, 10027,<br />

10056, 10078, 10092,<br />

10095<br />

Duffey, Steven e15054<br />

Duffield, Amy 6629<br />

Duffner, Jay 4056<br />

Duffy, Alison P 6611<br />

Duffy, Austin G. 3103,<br />

10566, e14064<br />

Duffy, Deirdre 6537<br />

Duffy, Mary e16538<br />

Duffy, Michael J. 622, 1042<br />

Duffy, Regan M. TPS7609<br />

Duffy, Steven 6572<br />

Dufour, Christelle 9560,<br />

9572<br />

Dufour, Jean-Francois 4107<br />

Dufour, Patrick Regis 636<br />

Dugan, Margaret Han 2603<br />

Duggal-Beri, Priya 6111<br />

Duggan, Peter 6548<br />

Dugo, Nella 5094, e15547,<br />

e15550<br />

Dugué, Pierre-Antoine e14006<br />

Duh, Mei Sheng 10061<br />

Duhamel, Alain 2540, 6534,<br />

6542<br />

Duhamel, Jean-Paul<br />

TPS7112, e18089<br />

Dujon, Cecile e16560<br />

Dukelow, Karen e17558<br />

Duman, Berna Bozkurt<br />

e11088, e13523<br />

Dumez, Herlinde 3018^<br />

Dummer, Reinhard 8076,<br />

8502^, 8505, LBA8509,<br />

8511, 8526, 8563,<br />

e19017<br />

Dumont, Amaury Gerard<br />

e13519<br />

Dumont, Frédéric e14158<br />

Dumont, Patrick TPS7111<br />

Dumont, Sarah N. e13519<br />

Dumstorf, Chad A 8082<br />

Dunbar, Erin M. TPS2103<br />

Duncan, Gail 4526<br />

Dunet, Vincent 10033<br />

718s Numerals refer to abstract number


Dunleavy, Kieron 2528,<br />

8005, 8051<br />

Dunlop, Alison 10039, 10060<br />

Dunlop, Joanna 1094<br />

Dunn, Ian F. 2020<br />

Dunn, Janet A TPS660,<br />

TPS665, TPS1144, 5046,<br />

e16042<br />

Dunn, Sandra E. e13123<br />

Dunne, Grainne 2536<br />

Dunning, Bernadine<br />

TPS5114, TPS10635<br />

Dunning, Mark 544<br />

Dunnwald, Lisa K 1088<br />

Dunphy, Frank e13032<br />

Dunson, William A. 9137<br />

Duong, Vu 2099<br />

Dupont, Jakob 3025<br />

Dupont-Gossard, Anne Claire<br />

4018<br />

Dupre, Pierre-Francois<br />

TPS646<br />

Duprez, Thierry 5519<br />

Dupuis, Charlotte e16044,<br />

e16051, e16055<br />

Dupuis, Jehan 8057<br />

Dupuis, Lee 9556<br />

Dupuis, Olivier LBA3500^,<br />

e14148<br />

Dupuy, Danielle 1553, 6120,<br />

e16531<br />

Dupuy, Evelyne e21156<br />

Duque, Cristiano Guedes<br />

e14027<br />

Duque, Lourdes e14727,<br />

e18516, e19638<br />

Durak, Merih e11507<br />

Duran, Hipolito 602<br />

Duran, Ignacio 3005, 3066,<br />

4119, TPS4672, e15111<br />

Duran, Jose e11036<br />

Durand, Jean-Bernard e13506<br />

Durand, Jean-Philippe<br />

e13056<br />

Durando, Antonio 1073<br />

Durando, Xavier 8532,<br />

e13108^<br />

Durant, Cecile 1580<br />

Durante, Emilia e13019<br />

Durante, Michael e13596<br />

Duranti, Simona e18023,<br />

e18101<br />

Durastante, Vittorio 3612<br />

Durci, Michael e17524<br />

Durie, Brian G. 8037, 8095,<br />

8097, e18556, e18561<br />

Durosinmin, Muheez e11554<br />

Dursun, Polat e15572,<br />

e15573<br />

Duruisseaux, Michael e18075,<br />

e18102<br />

Dutcher, Janice P. 4500,<br />

4504<br />

Dutel, Jean Luc 3506<br />

Dutt, Neelam e15554<br />

Dutta, Anindya 8597<br />

Dutton, Susan LBA4001<br />

Duval, Vincent e13605<br />

Duval-Modeste, Anne<br />

Benedicte 8591<br />

Duvic, Madeleine 8537<br />

Duvillard, Pierre 4129, 5028,<br />

10098<br />

Duvillie, Ladan 6512<br />

Duvvuri, Seshagiri TPS6635<br />

Dwivedi, Alok kumar e14714<br />

Dworacki, Grzegorz 10536<br />

Dwyer, David 8537<br />

Numerals refer to abstract number<br />

Dy, Grace K. 4117, 4545,<br />

7073, 7555, 7605, 8519,<br />

e13006, e13101, e17535,<br />

e18118<br />

Dy, Irene Ang 6595<br />

Dy, Philip 6582<br />

Dyer, Martin J. S. 6584<br />

Dyer, Michael A. e13544,<br />

e13584<br />

Dyer, Richard e11066<br />

Dykas, Dan 1540<br />

Dykema, Karl 4624<br />

Dyla, Alicja e21049<br />

Düran, Andreas 2560<br />

Dyson, Greg e14163<br />

Dyson, Gregory e14609,<br />

e16019<br />

Dyster, Lyn M. 4654<br />

Dyszkiewicz, Wojciech 10536<br />

Dytfeld, Dominik 8011<br />

Dzhabarov, Farkhad e19049<br />

Dziadziuszko, Rafal 7597<br />

Dziewirski, Wirginiusz 8548<br />

Dzodic, Radan e21136<br />

Dörken, Bernd 4020, 4044,<br />

e14533, e15053<br />

E<br />

E, Mingyan e14511<br />

Ea, Roth TPS8602<br />

Eaby-Sandy, Beth e18032<br />

Eagle, Ralph 9515<br />

Eakle, Janice F. 512, 3545<br />

Eap, Chin-Bin e11048<br />

Eapen, Ann e21076<br />

Earashi, Mitsuharu 1119<br />

Earl, Helena Margaret<br />

TPS660, TPS665,<br />

TPS1144<br />

Earl, Julie e12033<br />

Earle, Craig 6006, 6026,<br />

6039, 7606, e11050<br />

Early, Dayna S e14584<br />

Earnest, Arul 5553<br />

Earwood, Chris 4127<br />

Eary, Janet F. TPS3109<br />

Easaw, Jacob C. e14073,<br />

e21044<br />

Eastham, James Andrew 4552<br />

Easton, Doug 1545<br />

Easton, John 9518<br />

Eastwood, Daniel e17555<br />

Eaton, Anne 557, 575, 9104,<br />

e11531<br />

Eaton, Keith D. 2610, 5503,<br />

5515<br />

Ebb, David 9503<br />

Ebbinghaus, Scot 7531,<br />

10010<br />

Ebeid, Emad Nabil 9566<br />

Ebeling, P. e13100^<br />

Eberhardt, Wilfried Ernst<br />

Erich e18016^, e18100<br />

Eberhardt-Wetherington, Berit<br />

e19502<br />

Ebert Motara, Maja e14083<br />

Ebos, John M. e15042<br />

Ebrahim, Jalal 6084, 9111<br />

Ebsworth, Karen e13580<br />

Eccher, Claudio e21102<br />

Echaburu, Juan Virizuela<br />

TPS4672, TPS4674<br />

Echalier, Ben R. 520<br />

Echarri, Mari-a Eugenia 8572<br />

Echenique, Miguel M. 1060,<br />

e11015<br />

Echeveste, Jose Ignacio 8572,<br />

e18008<br />

Echt, Katharina V e15187<br />

Eckardt, Jeff J. 10012<br />

Ecke, Thorsten 4524<br />

Eckel, Robert H e15192<br />

Eckermann, Simon 2604,<br />

TPS9153<br />

Eckert, Robert 7572<br />

Eckhardt, S. Gail 3017,<br />

e13006, e14071<br />

Economopoulos, The<strong>of</strong>anis<br />

e21027<br />

Ecsedy, Jeffrey 2597<br />

Ecstein-Fraïssé, Evelyne B<br />

4570, e15112<br />

Eddy, Sean e14164<br />

Edelman, Gerald 7514, 8531<br />

Edelman, Martin J. 4022,<br />

6131, 7546, TPS7616<br />

Edelmann, David 10034<br />

Edelsberg, John e12024,<br />

e15197, e18107, e19630<br />

Edelstein, Kim 2063<br />

Edenfield, Jeff TPS3111<br />

Edesa, Wael e15530<br />

Edge, Stephen B. 1033,<br />

1125, 6088, e19568<br />

Edge, Stephen B e11563<br />

Edgerton, Susan e21099<br />

Ediebah, Divine Ewane 6002,<br />

6090, 7607<br />

Edil, Barish H 3596, 4045,<br />

e13559, e14585<br />

Edington, Howard 8533,<br />

TPS8606<br />

Edmonston, Tina B 10528<br />

Edmundowicz, Steven<br />

Alphonse e14584<br />

Edsgard, Daniel 10521<br />

Edwards, Beatrice J. 2532,<br />

4063, 6623<br />

Edwards, Connla 637<br />

Edwards, Joanne E. e14054,<br />

e14110<br />

Edwards, Susan e13084<br />

Eeles, Ros A. 1545, 4653<br />

Efremidis, Anna P. e11519<br />

Efstathiou, Eleni 4556,<br />

e15122<br />

Egan, Barbara 9094<br />

Egawa, Shin e15127<br />

Egawa, Shinichi 4035,<br />

e14011<br />

Egberts, Friederike e19031<br />

Egerer, Gerlinde 10040,<br />

10041^<br />

Egger, Matthias 4083, 4090<br />

Eggermond, Anja M 8039<br />

Eggermont, Alexander M.<br />

8535, e19022<br />

Eggers, Carrie 5560<br />

Eggmann, Nina e19017<br />

Eghan, Naa 6021<br />

Egleston, Brian 6021, 6061,<br />

6095, 6128<br />

Eglitis, Janis 9046<br />

Egorin, Merrill J. 2533<br />

Egorova, Natalia e15513<br />

Egrot, Christophe e15105<br />

Eguchi, Kenji 1530, e19556<br />

Ehinger, Mats 10024<br />

Ehlers, Shawna L. 9057<br />

Ehmann, W. Christopher 8090<br />

Ehninger, Gerhard 10068,<br />

10552<br />

Ehrenstein, Vera 1579,<br />

e12023<br />

Ehrenwald, Edward 3590<br />

Ehrlich, Peter 9524<br />

Ehrnrooth, Eva TPS5598,<br />

TPS5599<br />

EI-Khoueiry, Anthony e14031<br />

EI-Khoueiry, Rita e14031<br />

Eichelberg, Christian 4539<br />

Eichenauer, Dennis A. 8079<br />

Eichhorn, Christoph e19031<br />

Eichler, April F. 2009, 9004<br />

Eichler, Katrin e14131<br />

Eickh<strong>of</strong>f, Jens C. 3101,<br />

3103, TPS9152, e19047<br />

Eid, Toufic e20004<br />

Eidtmann, Holger 522, 541,<br />

1023<br />

Eiermann, WOLFGANG 1023<br />

Eigentler, Thomas K. 8526,<br />

e19031<br />

Eilber, Frederick C. 10000,<br />

10012<br />

Eilber, Frederick R. 10000<br />

Eilender, David S. 3619,<br />

e15554<br />

Eimermacher, Hartmut 6610<br />

Ein-Gal, Shlomit Y. 8524<br />

Einefors, Rickard 1594<br />

Einhorn, Jan e14125,<br />

e19606, e19638<br />

Einhorn, Lawrence H. 4534,<br />

4596, 4598<br />

Einsele, Hermann 6500^,<br />

e18572^<br />

Einstein, Mark H. 5025<br />

Eisbruch, Avraham e16014<br />

Eiseman, Julie 2553, 3031<br />

Eisen, Andrea 1555, e12034<br />

Eisen, Tim 4501, e15037,<br />

e18100<br />

Eisenberg, Marcia 614<br />

Eisenberg, Rosana 6038<br />

Eisenberger, Mario A. 4506,<br />

4559, 4564, 4568, 4570,<br />

4619, 4665, TPS4692^,<br />

e15058, e15188<br />

Eisenbruch, Maurice 6111,<br />

e16507<br />

Eisenhauer, Anne 5019<br />

Eisenhauer, Elizabeth A.<br />

1036, 2051, 5010<br />

Eisenhauer, Eric Lawernce<br />

5101, e15585<br />

Eiser, Christine 6113<br />

Eisinger, Francois 1567,<br />

1568, TPS1613<br />

Eisner, Florian e14066<br />

Eisner, Joel Robert 4671,<br />

e15167<br />

Eisterer, Wolfgang 4074,<br />

e14103, e14719<br />

Eito, Clara e14057<br />

Ejaz, Shamim 4641<br />

Ejlertsen, Bent 596, 604<br />

Ekenel, Meltem e14628<br />

Ekinci, Ahmet Siyar e14526<br />

Eklund, Martin e16504<br />

Ekman, Simon 7539, 7597<br />

Eksborg, Staffan 7539<br />

El Ashry, Mohamed S e15129<br />

El Cheikh, Jean 6534, 6542<br />

El Ghali, Ilham 10600<br />

el Ghandour, Ashraf e18572^<br />

El Ghazaly, Hesham e17556<br />

EL Haoudi, Khalid e14726<br />

El Mekkeoui, Zineb<br />

Benbrahim e18124<br />

El Mesbahi, Omar e11502,<br />

e11516, e14725, e14726,<br />

e18124<br />

El Messaoudi, Safia 10505<br />

El mrabet, Fatima Zahra<br />

e11516<br />

El Sadda, Wael M e15129<br />

719s


El Sherbeiny, Esam e15129<br />

El-Deiry, Mark 5570<br />

El-Deiry, Wafik e11544<br />

El-Gehani, Faraj e15540<br />

El-Hariry, Iman TPS7613^<br />

El-Hashimy, Mona 512, 2543<br />

El-Jawahri, Areej 6106, 9004<br />

El-Khoueiry, Anthony B.<br />

3078, 3584, 3604, 3615,<br />

3622, 4096, 4099, 4113,<br />

e13084, e14581, e14679<br />

El-Khoueiry, Rita 3540,<br />

3546, 3549, 3584, 3597,<br />

3604, 3622, 4026, 4088,<br />

4096, e11018<br />

El-Modir, Ahmed e15534<br />

El-Naggar, Adel K. 5524,<br />

5592<br />

El-Osta, Hazem Edmond<br />

3106<br />

El-Rayes, Bassel F. 3094,<br />

e14169, e14692, e14736<br />

El-Rayes, Bassel F 2576,<br />

e14170, e14512, e14614<br />

El-Serag, Hashem 3542<br />

El-Shikh, Wafaa 4116<br />

El-Shinawi, Mohamed e21055<br />

El-Tamer, Mahmoud 557,<br />

1118<br />

Elaesser, Reiner e19587<br />

Elagoz, Sahande e11083<br />

Elaidi, Reza-Thierry e15040<br />

Elash<strong>of</strong>f, David 2519<br />

Elash<strong>of</strong>f, Robert M. 7518<br />

Elash<strong>of</strong>f, Robert 8534<br />

Elatre, Wafaa 4010<br />

Elbadawy, Samy 9566<br />

Eldon, Michael A. 5048<br />

Eleftheraki, Anastasia G 5033<br />

Elena Vaja, Elena Vaja 5574<br />

Elez, Elena 2583, 3539<br />

Elfakharany, Mohamed 3619<br />

Elfring, Gary L. e13588<br />

Elia, Loredana 6531<br />

Elias, Anthony D. 528, 564,<br />

TPS668, TPS10099,<br />

TPS10103<br />

Elias, Dominique 3507,<br />

e14158<br />

Elias, E. George e13129<br />

Eliott, Jaklin A. e19553<br />

Elisei, Rossella 5508, 5591<br />

Elkin, Elena B. 6001, 6073,<br />

6101<br />

Elkiran, Emin Tamer 638<br />

Elkiran, Tamer 9050<br />

Ellard, Susan 549^, 582,<br />

1036, 4514<br />

Ellberg, Carolina 1594<br />

Ellebaek, Eva 2565, 2574<br />

Ellezam, Benjamin 2022<br />

Ellimoottil, Chandy e16544<br />

Elling, Dirk 624<br />

Elliott, Brian M e21006<br />

Elliott, Martin 9542<br />

Elliott, Michelle A. 6614<br />

Elliott, P. Anthony e15141<br />

Elliott, Richard 3050<br />

Ellis, Alison 7030<br />

Ellis, Catherine Elizabeth<br />

TPS658<br />

Ellis, Ian O. 1013, 1014,<br />

1098<br />

Ellis, Matthew J. TPS664,<br />

1048, 3047<br />

Ellis, Matthew James 503,<br />

534, 1008<br />

Ellis, Peter Michael 7093,<br />

7511, e17532<br />

Ellis, Robin Anne 2569<br />

Ellis, Sarah Gabrielle 8566<br />

Ellis, Steve 1545<br />

Ellis, Sue 6113<br />

Ellis, William J 4520<br />

Ellison, Christie e16561<br />

Ellison, David W. 9518<br />

Ellsworth, Eric M. e21048,<br />

e21121<br />

Ellwanger, Kristina 8059<br />

Elmesidy, Salah Eldeen<br />

e18001<br />

Elmishad, Amira e13121<br />

Elramsisy, Adam 2080<br />

Elsass, Karen E. 9575<br />

Elsayed, Yusri A. 3059<br />

Elsayegh, Nisreen 1546,<br />

1549<br />

Elsayem, Ahmed F. 6125<br />

Elserafy, Mostafa M. e20514<br />

Elshaikh, Mohamed A. 5088,<br />

e15512<br />

Elson, Paul 2018, 2076,<br />

2096, 4609, TPS4679,<br />

TPS4684, 6521, e15095,<br />

e15208, e18046<br />

Elston-Lafata, Jennifer<br />

e18144<br />

Elstrom, Rebecca L. 8054<br />

Elter, Thomas 7603, 8031<br />

Elting, Linda S. 600, 6124<br />

Elvin, Paul 3561<br />

Elving, Lammy 9070<br />

Ely, Benjamin 4547, 4571,<br />

4663, 10503<br />

Ely, Scott 8036<br />

Elyan, Firas 10034<br />

Emadi, Ashkan 6629<br />

Emami, Marjan 1535, 7586<br />

Emanuelsson, Ol<strong>of</strong> 10521<br />

Emblem, Kyrre E. 2009<br />

Embree, Elizabeth 10533<br />

Embree, Heather TPS1616<br />

Emerick, Kevin S. e16059<br />

Emerson, Ryan 6631<br />

Emi, Manabu e14636<br />

Emi, Yasunori 3558<br />

Emile, Jean-François 8540,<br />

e14059<br />

Emmanuel, Romala 4063,<br />

e18571<br />

Emmel, Ann E. 6074<br />

Emmenegger, Urban<br />

e15177^, e15180<br />

Emmons, Gregory Scott<br />

e19543<br />

Emond, Jean 4101<br />

Emoto, Shigenobu e14530<br />

Endell, Jan 6574<br />

Enderica Gonzalez, Sergio<br />

5077<br />

Endlicher, Esther 4034<br />

Endo, Itaru 2044, e14014,<br />

e14047, e14050, e14624<br />

Endo, Kojin e14011<br />

Endo, Masahiro 7088,<br />

e21007<br />

Endo, Takashi 10531<br />

Eng, Cathy LBA3501, 3544,<br />

3556, 3598, 4060<br />

Eng, Charis 1508, 1516,<br />

5573<br />

Eng, Dawn Fun TPS6638<br />

Eng, Lawson 1597, 9032<br />

Eng, Yoko 4098, e16058<br />

Engel, Christoph 3550<br />

Engel, Erik e14138<br />

Engel, Jessica Marie 555,<br />

563, 6063, e14160<br />

Engel, Juergen e15153<br />

Engel, Jutta 1114<br />

Engel-Riedel, Walburga 1533,<br />

7001, 7603, CRA10529,<br />

e18000<br />

Engelen, Kristel 1022<br />

Engell-Noerregaard, Lotte<br />

2574<br />

Engellau, Jakob 10024<br />

Engelman, Eric Steven<br />

e14600, e14620<br />

Engelman, Jeffrey A. 7508<br />

Engels, Eric A. 6070<br />

Engelstaedter, Verena e15529<br />

Engenhart-Cabillic, Rita 5512<br />

Engert, Andreas 8079,<br />

e13079<br />

Engle, Robert e13081<br />

English, Patricia A e13606<br />

Engracia, Ruth Ginger<br />

e15571<br />

Enis, Marguerite 1019<br />

Enk, Alexander 4034, 8505<br />

Enke, Aaron S. TPS7613^<br />

Enkemann, Steven 10048<br />

Ennis, Marguerite 2572<br />

Enomoto, Takahiro e19583<br />

Enos, Rebecca A. e16561<br />

Enrech, Santos e11074<br />

Enright, Katherine e14003<br />

Ensminger, William D.<br />

e14603<br />

Ensor, Joe 594, e14664<br />

Enting, Roelien H. 2<br />

Enzinger, Peter C. 4027,<br />

e19586<br />

Eoli, Marica 2083<br />

Eom, Hyeon Seok 8023<br />

Epenetos, Agamemnon A.<br />

e13525<br />

Epner, Elliot E 6616, e18500<br />

Epstein, Andrew S. 9004,<br />

9105<br />

Epstein, Joel Brian 5528<br />

Epstein, Jonathan I 4570<br />

Epstein-Peterson, Zachary<br />

9116<br />

Eramo, Adriana e13512<br />

Erasmus, Jeremy J. 4069,<br />

4078<br />

Erba, Harry Paul 6502, 6582,<br />

TPS6637<br />

Erben, Philipp 6510<br />

Erbs, Philippe 2605<br />

Erbstoesser, Erhard 1023<br />

Ercan, Dalia 7510<br />

Ercolano, Elizabeth TPS669,<br />

TPS1614<br />

Ercoli, Alfredo e12037<br />

Erdag, Gulsun 8530<br />

Erdem, Dilek e14107<br />

Erdmann, Michael e19032<br />

Erdozain, Jose Carlos 1570<br />

Eremin, Dmitry 6088<br />

Eren, Mehmet Fuat e16005,<br />

e19575<br />

Eren, Onder Orhan e11008<br />

Eren, Tulay e11011<br />

Erfán, Jozsef 5516^<br />

Ergin, Melek e11088<br />

Ergun, Aydin e11026<br />

Erho, Nicholas 4565<br />

Erickson, Beth e14613<br />

Erickson, Julie e21129<br />

Ericson, Solveig 6594, 6605,<br />

TPS6636, TPS6638<br />

Eriksson, Andre Huss e19056<br />

Eriksson, Jennifer 6058<br />

Eriksson, Mikael 8074,<br />

10081<br />

Erill, Nadine e14552<br />

Erin, Nuray e21130<br />

Erinjeri, Joseph Patrick<br />

10004<br />

Erismis, Betul e14174<br />

Erkmen, Cherie Parungo 7022<br />

Erlander, Mark G. 561,<br />

10530, 10588, e21019<br />

Erler, Brian 1540<br />

Erlichman, Charles 534,<br />

3001, 4047, 4099, 5020,<br />

5025, 5544, 6570, 8013,<br />

e13052, e13517<br />

Ermacora, Paola 3612<br />

Ermani, Mario 2057, 2061<br />

Ernest, Mary Lou 9107<br />

Ernst, Brenda e16030<br />

Ernst, Christine 2584<br />

Ernst, D. Scott 8587, e19616<br />

Ernst<strong>of</strong>f, Marc S. TPS2614<br />

Eroglu, Celalettin e14526,<br />

e14670<br />

Ersahin, Cagatay e15538<br />

Erten, Mujde Z. 6060, 6075<br />

Ertl, Linda S e13580<br />

Ervin, Thomas J. LBA3501,<br />

TPS3640, e13113<br />

Esaki, Taito 3045, 4002<br />

Escalante, Carmelita P. 9072<br />

Escallon, Jaime 1019<br />

Escalon, Juliet 2589<br />

Escande, Fabienne 10507<br />

Escano, Crystal 2589<br />

Escarzaga, Diana e18117<br />

Escriu, Carles 8523<br />

Escudero, M. Pilar 3571<br />

Escudero, P. 4119<br />

Escudero, Pilar 3553<br />

Escudier, Bernard J.<br />

CRA4502, 4540, 4546,<br />

4554, 4614, 4621, 4625,<br />

TPS4683, e15049<br />

Escuin, Daniel 10555<br />

Eser, Markus 8059<br />

Eskens, Ferry 2593, 4132<br />

Esmay, Paula 5523<br />

Espadana, Ana e12040<br />

Esparaz, Benjamin 9028<br />

Espat, Joseph 4098<br />

Esperide, Barbara 9098<br />

Espie, Marc TPS652<br />

Espino, Guillermo e13604<br />

Espinosa, Enrique 8517,<br />

e19023<br />

Espinosa, Iñigo e14104<br />

Espinoza Venegas, Macarena<br />

Paz 4587, e16025<br />

Espinoza, Anne Marie 4038<br />

Espinoza, Ramiro 6599<br />

Espinoza-Delgado, Igor<br />

TPS4687<br />

Espirito, Janet L. 6109<br />

Esplen, Mary-Jane 3567<br />

Esposito, Angela 1049, 1115<br />

Esposito, Assunta 8573,<br />

e19034<br />

Esquerdo, Gaspar 4587,<br />

e16025, e18049, e18053<br />

Esser, Regina 3574<br />

Esserman, Laura 9107,<br />

10586, e16504<br />

Esteban Esteban(1), Carmen<br />

e11543, e19596<br />

Esteban, Emilio TPS4683,<br />

TPS4685, e15041,<br />

e15067, e15149, e18044<br />

Esteban, Isabel e11028<br />

720s Numerals refer to abstract number


Esteller, Manel 3570<br />

Esterni, Benjamin LBA4567^,<br />

e18526, e19539<br />

Estes, Jacob Michael 5062^,<br />

e13087, e15502<br />

Esteva, Francisco J. 527,<br />

612, 10613<br />

Estevan, Rafael 3586<br />

Esteves, Brooke 4132, 4501,<br />

e15082<br />

Esteves, Jorge e14673<br />

Esteves, Susana e13119<br />

Estevez-Diz, Maria Del Pilar<br />

e15562<br />

Estorch, Montserrat 1120<br />

Estrella, Heather 3509<br />

Estrella, Timothy e21155<br />

Estrov, Zeev 6517, 6528,<br />

6589<br />

Esumi, Hiroyasu 1552<br />

Eswarvee, Chinnamani<br />

e15128, e15135, e18068<br />

Etchin, Julia e13586<br />

Etemadi, Hooman e17521<br />

Etemadmoghadam, Dariush<br />

5073<br />

Etienne, Pierre-Luc LBA4003<br />

Etienne-Grimaldi,<br />

Marie-Christine 2537^,<br />

2600<br />

Etiz, Durmus e21050<br />

Eto, Hidehiro e15533<br />

Eto, Masatoshi e15048<br />

Etoh, Tsuyoshi 3569<br />

Ettl, Johannes 556<br />

Ettore, Giuseppe e15544<br />

Ettrich, Thomas TPS1612,<br />

TPS3636<br />

Etxaniz, Olatz 4579, e12504,<br />

e15001<br />

Etzioni, Ruth D 4557<br />

Eubanks, Susan e17547,<br />

e17550<br />

Eucker, Jan 8031<br />

Eulenburg, Christine 5058<br />

Eustace, Alex J. 633<br />

Eustaquio, Joy e19570<br />

Eustermann, Heidi 640<br />

Evans, Andrew 4633,<br />

e15118, e18139<br />

Evans, Arthur T 9085<br />

Evans, Douglas B. 4013,<br />

4051, e14613<br />

Evans, Erica 8032<br />

Evans, Kenneth R. 10622<br />

Evans, Kenneth 7020<br />

Evans, Laura 9019<br />

Evans, Linda TPS4143<br />

Evans, Steven e14075<br />

Evans, T.R. Jeffry 2552,<br />

3000, TPS4134, TPS8605,<br />

TPS10633^<br />

Evans, Tracey L. 7092, 7098,<br />

7533, 7546, 7595, 7596,<br />

e18032<br />

Evans, William K. 6049<br />

Evaul, Kristen 4645<br />

Evelhoch, Jeffrey L. 10573<br />

Even, Caroline 5028<br />

Even-Shoshan, Orit 6000<br />

Evens, Andrew M. 8060,<br />

8072, 8080<br />

Everaert, Hendrik 2050<br />

Evers, Kathryn 6061<br />

Evoy, Denis 1042<br />

Evrard, Serge e14017<br />

Evren, Sevan e15118<br />

Ewald-Riegler, Nina 5005<br />

Ewend, Matthew G. 2091<br />

Numerals refer to abstract number<br />

Ewer, Michael 533^, e13506<br />

Ewesuedo, Reginald 2534,<br />

e13074<br />

Extermann, Martine 6100,<br />

7065, e17559<br />

Extremera, Sonia TPS652,<br />

2539<br />

Ey, Frederick S. e11042<br />

Eymard, Jean-Christophe<br />

e15142<br />

Ezaoui, Sarah e21046<br />

Ezeife, Doreen 6082<br />

Ezeome, Emmanuel e19572<br />

Ezewuiro, Obiageli 9571<br />

Ezhilarasan, Ravesanker 2002<br />

Ezziddin, Samer e14565,<br />

e14654<br />

F<br />

F. Blaine Hollinger, F. Blaine<br />

e17016<br />

Fabarius, Alice 6510<br />

Fabbri, Francesca 10615<br />

Fabbri, Maria Agnese e12514<br />

Fabbri, Maria Agnese<br />

LBA7501, e14082, e18065<br />

Fabbro, Michel 5018,<br />

e15535<br />

Fabi, Alessandra 1073<br />

Fabian, Carol J. 520, 9000<br />

Fabian, Thomas 7024<br />

Facanha, Adriana e14183<br />

Facchini, Gaetano e15160<br />

Facchini, Thomas TPS4692^<br />

Fackler, Mary Jo 518<br />

Fackrell, David Gareth<br />

e15534<br />

Facon, Thierry 8009, 8020,<br />

TPS8112, TPS8113,<br />

TPS8114<br />

Fadare, Oluwole e15582<br />

Faderl, Stefan 6501, 6517,<br />

6528, 6544, 6558, 6578,<br />

6585, 6589, 6592, 6597,<br />

6603, 6607<br />

Fadhel, Ehab 6084, 9032<br />

Fadul, Camilo E. CRA9013<br />

Faedi, Marina 2037, 2057,<br />

e11054<br />

Faehling, Martin 7572<br />

Fagan, Dedra H. e13590<br />

Faggiano, Antongiulio 1604,<br />

e19038<br />

Fagin, James A. 5509^, 5514<br />

Fagnani, Daniele 9120<br />

Fagniez, Nathalie 8057<br />

Fagundes, Renato Borges<br />

e14597<br />

Fahey, Thomas J e21011<br />

Fahl, William E TPS9152<br />

Fahrig, Antje e17516<br />

Fainstein, Igor e15029<br />

Fairbairn, Elliann 6115<br />

Fairchild, Justin P. TPS4691,<br />

TPS7113<br />

Fairey, Adrian Stuart 4563,<br />

4576, e15171<br />

Fairooz, Puthiyapurayil M<br />

9568<br />

Faivre, Sandrine J. 2554,<br />

2557, 3609, 4032, 5504^,<br />

e13594, e14553, e14660<br />

Faivre, Théa TPS3118<br />

Faivre-Finn, Corinne 7538,<br />

e18095<br />

Faivre-Pierret, Matthieu 2070<br />

Fajardo, Julieta e19528,<br />

e19584<br />

Fakhro, Summer A 3036<br />

Fakhry, Nicolas e16055<br />

Fakih, Marwan 3582<br />

Fakiris, Achilles 7040<br />

Falandry, Claire TPS667,<br />

8068, 9011, 9079, 9080<br />

Falchero, Lionel e18005<br />

Falchi, Lorenzo 6516^<br />

Falchook, Gerald Steven<br />

3102, 3106, 3107^, 3528,<br />

5021, 8510, 8551, 8594,<br />

10510, 10607, e13020,<br />

e13506, e13595, e13596,<br />

e19004<br />

Falci, Cristina e13019<br />

Falcone, Alfredo 629, 3502,<br />

3518, 3540, 3546, 3549,<br />

3555, 3585, 3597,<br />

e13057, e14040, e14113<br />

Falcone, Italia e13512,<br />

e13572<br />

Falcone, Katherine e15565<br />

Faletti, Carlo 10055<br />

Falge, Christiane 6510<br />

Falk, Martin H. TPS4701,<br />

LBA7000<br />

Falk, Roni 1521<br />

Falk, Stephen LBA4000,<br />

LBA4001, TPS4143, 7562<br />

Falkowski, Slawomir 8548,<br />

10538<br />

Fallai, Carlo 5555<br />

Faller, Douglas V. e14591<br />

Fallowfield, Lesley 5527<br />

Faloppi, Luca e14086,<br />

e14561, e14663, e15074<br />

Falusi, Adeyinka e11554<br />

Faluyi, Olusola Olusesan<br />

4058<br />

Falzarano, Sara Moscovita<br />

4560<br />

Falzone, Rick 3062<br />

Fan, Alice C. 10513<br />

Fan, Jia e14538<br />

Fan, Kang-Hsien e14503<br />

Fan, Lin 9009, 9014, 9091<br />

Fan, Lu e13002<br />

Fan, Qingxia e18033<br />

Fan, Sheung Tat e14545<br />

Fan, Songhua 3038<br />

Fan, Tiffany 7049<br />

Fan, Timothy M. e21145<br />

Fan, Xuemo 2087<br />

Fan, Ying e11025, e11538<br />

Fan, You Hong 5524, 10612<br />

Fan, Yunhua 1576<br />

Fan, Zhongyi 7036<br />

Fanale, Michelle A. 6116,<br />

8027, 8065<br />

Fanciulli, Giuseppe 1604<br />

Fanelli, Marcello Ferretti 643,<br />

e14680, e14728, e18078<br />

Fanelli, Marcelo e11087<br />

Fang, Cheng 10500<br />

Fang, Erica 10598<br />

Fang, Hui e14511<br />

Fang, Liang LBA1<br />

Fang, Min e15207<br />

Fang, Qin 7034<br />

Fang, Xiang e16554<br />

Fang, Xue-Feng e14026<br />

Fang, Yong 4092, e14575<br />

Fanica, Daniela 4018<br />

Fann, Jesse R 9008<br />

Fanta, Paul Timothy 2052,<br />

e14069, e14078, e14079<br />

Fantasia, John e16026<br />

Fanti, Stefano e21006<br />

Fantin, Bruno 9067<br />

Fantini, Simona e21118<br />

Farace, Françoise 3080<br />

Faraggi, Marc e15558<br />

Farah, José Francisco de<br />

Mattos e14657<br />

Farah, Michelle de lima<br />

e21151<br />

Farah, Naim e15024<br />

Farassati, Faris e13535<br />

Farber, Charles Michael 6122<br />

Farbstein, Motti e14731<br />

Fareed, Jawed e13115,<br />

e13117, e15008<br />

Farfalli, German 10074<br />

Farfan, Carlos Alberto 4620<br />

Farge, Dominique 1580,<br />

e13062<br />

Farges, Olivier 3609<br />

Farhadian, Joshua Andrew<br />

8544, 8565<br />

Farhat, Rawad e20004<br />

Faria, Claudio e16521<br />

Farias, Caroline Brunetto de<br />

e13546<br />

Faricy-Anderson, Katherine E.<br />

4098<br />

Fariello, Anna Maria e12514<br />

Faries, Mark B. 8534,<br />

e19029<br />

Farina, Gabriella LBA7501,<br />

9005<br />

Farina, Patrizia 2049<br />

Faris, Jason Edward e14615<br />

Farmer, Pat 1036<br />

Farol, Len 8089<br />

Farolfi, Alberto e11054<br />

Faron, Matthieu 3507<br />

Farooq, Aamer e17535<br />

Farouk, Fatma e15129<br />

Farragher, Susan 3516<br />

Farrar, John T e19531<br />

Farrar, William Blair LBA506<br />

Farre, Jose 3064<br />

Farre, Nuria 5590, 5595<br />

Farrell, Brian T. 2077<br />

Farrell, Michael P. 1543,<br />

e12031, e12038<br />

Farsi, Fadila e19640<br />

Fartoux, Laetitia 4032<br />

Farver, Carol e18085<br />

Fasching, Peter A. 541,<br />

TPS1146, 10599<br />

Fasciano, Karen 9015<br />

Fasola, Giampiero 1604<br />

Fasola, Gianpiero 1044,<br />

3612, 7013, 7550,<br />

e13058, e13070, e14040,<br />

e15091<br />

Fassunke, Jana CRA10529<br />

Fast, Loren e21116<br />

Fatato, Penny Marie 7030<br />

Fathallah, Hassana TPS3116<br />

Fathi, Amir Tahmasb 6606,<br />

6617, 6618, 6621, 6629<br />

Fauci, Janelle M. 5060<br />

Faure, Gilbert e12506<br />

Fausel, Christopher A.<br />

e18555<br />

Fautrel, Alain e19037<br />

Favaretto, Adolfo G. 7082,<br />

7550, 7575, e18130<br />

Favier, Bertrand e16036<br />

Favier, Laure LBA5000<br />

Favret, Anne 1018<br />

Fawcett, Jonathan TPS3643<br />

Fawole, Adewale Alade 2076,<br />

2093<br />

Fawzy, Mahmoud 5557<br />

Fay, Angela Musial 1540<br />

Fay, Michael F. TPS2104<br />

721s


Fayad, Luis 6501, 8067<br />

Fayette, Jerome 5505,<br />

e16036<br />

Fazal, Salman 6620<br />

Fazekas, Belinda 2604<br />

Fazio, Nicola 4120<br />

Fazio, Vito Michele e21092<br />

Fecher, Leslie Anne 8518<br />

Fedarko, Neal e13042<br />

Fedele, Palma e11001,<br />

e11547<br />

Fedele, Roberta e18564<br />

Fedeli, Anna e11054<br />

Fedeli, Franco e17533<br />

Federico, Massimo 8006<br />

Federico, Piera e15034,<br />

e19038<br />

Federico, Sara Michele<br />

e13584<br />

Federico, Victoria 9144,<br />

e19633<br />

Federspiel, Birgitte 4015<br />

Federspiel, Jerome J e15184<br />

Fedorenko, Catherine R. 6007<br />

Fedyanin, Mikhail e15025,<br />

e15029, e15082<br />

Fee-Schroeder, Kelliann C.<br />

9075<br />

Feeback, Jennifer Wanpen<br />

e14632<br />

Feely, Homira 2052<br />

Feeney, Kendra J. e19016<br />

Fegely, Maryellen e15565<br />

Feghaly, Julien e20004<br />

Fegueux, Nathalie 6534<br />

Feher, Olavo 2038, e12513,<br />

e16046<br />

Fehm, Tanja N. 1023, 1067,<br />

1090, TPS1146, 1602,<br />

5042, 10599, e15577,<br />

e21003<br />

Fehrenbacher, Louis LBA506,<br />

LBA1000, 1025, TPS1142,<br />

7517<br />

Fehrmann, Rudolf S. N.<br />

e15035<br />

Fei, Kezhen e11004, e19621<br />

Fei, Shi-Jiang e13510<br />

Feigenberg, Steven J. 10572<br />

Feigin, Kimberly 1006<br />

Feiglelson, Heather S e14160<br />

Feilotter, Harriet 5032<br />

Fein, Francine 4018<br />

Fein, Luis Enrique 9077<br />

Fein, Luis 7029, e11087<br />

Feinberg, Bruce A. 569, 571,<br />

6098, e11002, e16540,<br />

e16552<br />

Feinberg, Zachery e14185<br />

Feinmesser, Meora 10528<br />

Feinstein, Stuart e19571<br />

Feit, Kevie e18063^<br />

Feith, Marcus e14527<br />

Fekrazad, M Houman 5549<br />

Feld, Ronald 1535, 7029,<br />

7586, e14524<br />

Feldberg, Edit e19529<br />

Feldman, Darren Richard<br />

4532<br />

Feldman, Eric Jay 6525,<br />

6601, TPS6637<br />

Feldman, Lawrence Eric 5528<br />

Feldman, Mark 10037<br />

Feldman, Max e14579<br />

Feldman, Rebecca A e15209<br />

Feldman, Tatyana e18507,<br />

e19641<br />

Feldman-Moreno, Rebecca<br />

Anne 5523<br />

Felger, Jennifer 9122<br />

Felici, Alessandra 4541<br />

Feliciano, Josephine Louella<br />

6131<br />

Felip, Enriqueta 1531, 3007,<br />

7041, 7522, 7542, 7543,<br />

7544, e12000, e12012<br />

Feliu, Jaime 1570, 3586,<br />

3618, 4079, 10547,<br />

e13085, e14097<br />

Féliz, Luis Roberto 3536,<br />

e13540<br />

Fellague-Chebra, Rafik<br />

TPS8111<br />

Fellenz, Lori 6054<br />

Fellman, Bryan 6054<br />

Felsberg, Joerg 2067<br />

Felsher, Dean W. 10513<br />

Fenaux, Pierre 6522, 6523,<br />

6534<br />

Fencl, Pavel e14631<br />

Fend, Falko e15577<br />

Feng, Gansheng e14605<br />

Feng, Guosheng e16054<br />

Feng, Janine D. 4041<br />

Feng, Ji Feng 7520, 7557,<br />

7559<br />

Feng, Lei 4641, 5524, 5592,<br />

8067<br />

Feng, Mary Uan-Sian e14603<br />

Feng, Ping bo 4073<br />

Feng, Rentian 8099<br />

Feng, Shibao 1526,<br />

TPS3635^, e19048^<br />

Feng, Yan TPS2622, 8541,<br />

e18085<br />

Feng, Yang 3605<br />

Fengler, Ruediger 9573<br />

Fenig, Eyal e14571<br />

Fennell, Dean Anthony 7583,<br />

TPS7613^<br />

Fennell, Mary 6086<br />

Fennell, Thomas e17015<br />

Fennelly, David William<br />

e13570<br />

Fennelly, David e21024<br />

Fenner, Martin 4631, e15190<br />

Fenton, Dave 7511<br />

Fenton, Mary Anne 1045<br />

Fenwick, Stephen W 3613<br />

Fergus, Karen e19545<br />

Ferguson, Maree e19550<br />

Ferguson, Mark K. 4062<br />

Ferguson, Sarah E 5026<br />

Ferhanoglu, Burhan 6550<br />

Ferland, Emery e14020<br />

Ferlay, Celine 4614, 4625,<br />

7574<br />

Ferme, Christophe 8002<br />

Fern, Lorna Anne 6115<br />

Fernandes, Annemarie 7098,<br />

e17534, e18032<br />

Fernandes, Donald Jerald<br />

5526<br />

Fernandes, Gabriela 1522<br />

Fernandes, Kimberly A. 6087<br />

Fernandes, Recidia Rayane<br />

Reboucas e12512<br />

Fernandez Abad, Maria 602,<br />

e11006, e11517<br />

Fernandez Calvo, Ovidio<br />

e18040, e18146<br />

Fernandez Franco(1), Lourdes<br />

e11543, e19596<br />

Fernández Gabriel, Elena<br />

e15067<br />

Fernandez Montes, Ana<br />

e18040<br />

Fernandez Parra, Eva e11075<br />

Fernandez, Antonio e19576<br />

Fernandez, Beatriz 10534<br />

Fernandez, C. 4630<br />

Fernandez, Carmen Gomez<br />

595, 608<br />

Fernandez, Cristian 7029<br />

Fernandez, Lita 3015<br />

Fernandez, Maria Elena Elez<br />

3014<br />

Fernandez-Acenero, Maria<br />

Jesus e15094, e18069,<br />

e18106, e21015<br />

Fernandez-cruz, Laureano<br />

e21035<br />

Fernandez-del Castillo, Carlos<br />

4021<br />

Fernández-Esparrach, Gloria<br />

3536<br />

Fernandez-Garcia, Eva<br />

TPS652, 2539<br />

Fernandez-Martos, Carlos<br />

3586, 3618, 10547,<br />

e14041, e14097<br />

Fernandez-Morejon, Francisco<br />

Jose 3064<br />

Fernando, Indrajit Nalinika<br />

5046, e15534<br />

Ferolla, Piero 1604, e19038<br />

Ferone, Diego 1604, e19038<br />

Ferrajoli, Alessandra 6516^,<br />

6517, 6587, 6598<br />

Ferrandez, Dolores e14147<br />

Ferrandina, Gabriella<br />

LBA5003<br />

Ferrandino, Michael 4670<br />

Ferraresi, Virginia 10006,<br />

e14661<br />

Ferrari, Alessia e11071<br />

Ferrari, Anna C. e15191<br />

Ferrari, Antonella e17006<br />

Ferrari, Daris 5513<br />

Ferrari, Laura 3612, 4017<br />

Ferrari, Stefano 10022,<br />

10079<br />

Ferrarini, Ilaria 629<br />

Ferrario, Cristiano 1099<br />

Ferraro, Giuseppa e11056<br />

Ferraro, John TPS3638,<br />

e14501<br />

Ferrarotto, Renata 3544<br />

Ferraù, Francesco 7550<br />

Ferraz, Marcio R. e11513<br />

Ferre, Malena e14552<br />

Ferre, Pierre e13036<br />

Ferreira, Ana Carolina e21073<br />

Ferreira, Carlos G. M. 7506,<br />

e14101<br />

Ferreira, Fabio Oliveira<br />

10069, e14119<br />

Ferreiro, Jose Luis e21158<br />

Ferreiro, Josefa e11525<br />

Ferrell, Betty R. 9068,<br />

e16563, e19563<br />

Ferrer Lopez, Pablo 1574<br />

Ferrer, Ana Isabel e19576<br />

Ferrer, Milagros 10564<br />

Ferrer-Lozano, Jaime 1015<br />

Ferreri, Andres J. 8058<br />

Ferreri, Andres 8006<br />

Ferrero, Annamaria LBA5000<br />

Ferrero, Jean Marc LBA4567^<br />

Ferrero, Jean-Marc TPS646,<br />

TPS654^<br />

Ferretti, Gabriele 7084<br />

Ferretti, Gianluigi 1050<br />

Ferretti, Vincent e13107<br />

Ferreyros, Greenlandia<br />

e11518<br />

Ferri, Ivana e21094<br />

Ferri, Marco e14018<br />

Ferris, Robert L. 5528, 5566<br />

Ferro, Alfredo 1007<br />

Ferro, Antonella 586,<br />

e14711, e21102<br />

Ferro, Paola e17533<br />

Ferrone, Cristina 4021<br />

Ferrone, Marcus 4660<br />

Ferrone, Soldano 8528,<br />

10614<br />

Ferru, Aurelie 3520<br />

Ferrucci, Pier Francesco<br />

8513, 8529, e19035,<br />

e19036<br />

Ferrusi, Ilia L e11050<br />

Ferry, David Raymond<br />

LBA4000, e13007<br />

Ferry, David R 3505,<br />

LBA4001<br />

Fersching, Deborah e21067<br />

Fersis, Nikos 624<br />

Ferte, Charles 4540<br />

Fertig, Elana J. 5549<br />

Fertil, Bernard 8578<br />

Feser, William 10580<br />

Fesl, Christian 514<br />

Festiccia, Claudio e15200<br />

Fetsch, Patricia e14179<br />

Fetterly, Gerald J. 3029,<br />

4117, 4545, 4654, 8519,<br />

e13006, e14690<br />

Fetting, John H. 1128,<br />

10509<br />

Feusner, James Henry 9553<br />

Feyer, Petra C. 5512, 9033,<br />

e19540<br />

Feyerabend, Susan e15195<br />

Feyler, Sylvia 8015<br />

Fhied, Cristina e21069<br />

Ficarra, Guido 631<br />

Ficker, Joachim H e17536<br />

Ficorella, Corrado e14010<br />

Fidias, Panos 4027, 7099<br />

Fidler, Mary J. 7067, 7546,<br />

e18117, e21069<br />

Fiduccia, Pasquale 2049<br />

Fiedler, Walter M. 2504<br />

Fiel, M. Isabel e14583<br />

Field, Jeff e21152<br />

Field, Kathryn Maree e16516<br />

Fiellin, Martha TPS669<br />

Fietkau, Rainer 7001<br />

Figarella, Dominique 2060<br />

Figarella-Branger, Dominique<br />

2023<br />

Figg, William Douglas 3042^,<br />

3043, 4569, 4671<br />

Figlin, Robert A. 7518,<br />

e15059, e15084<br />

Figueira, Alicardo Cesar 606<br />

Figueiredo-Pontes, Lorena<br />

Lobo de 7523<br />

Figueras, Joan e14120<br />

Figueredo, Javier 2515<br />

Figueroa, Angelica e14595,<br />

e21002<br />

Figueroa, Salvador 1531,<br />

e12012<br />

Figueroa, Santiago 7058,<br />

10594<br />

Figurin, Konstantin e15025,<br />

e15029<br />

Filanovsky, Kalman e19529<br />

Filbet, Marilene 9049, 9080,<br />

e19578<br />

Filiberti, Silvia 9120<br />

Filicko-O’Hara, Joanne E.<br />

e16548<br />

Filidei, Mario 629<br />

722s Numerals refer to abstract number


Filipazzi, Paola 5555<br />

Filipits, Martin 542, 7007,<br />

10501<br />

Filippi, Luca e19002<br />

Filippi, Marie-Helene e11019<br />

Filleron, Thomas 10076<br />

Filleul, Bertrand 3559<br />

Filleur, Stephanie e15103<br />

Finas, Dominique e13100^<br />

Findlay, Brian Peter 3572<br />

Findlay, Michael P. N.<br />

e15054<br />

Fine, Barbara e16515<br />

Fine, Robert e14708<br />

Finelli, Antonio e15118<br />

Fingert, Howard 2597<br />

Fink, James R TPS10635<br />

Fink, Karen L. 2008b,<br />

TPS2107<br />

Fink, Louis M. 10503<br />

Fink, Stephanie 4565<br />

Finkelstein, Daniel TPS3117<br />

Finkelstein, Dianne M 596,<br />

604, TPS670<br />

Finkelstein, Julia e15021<br />

Finkelstein, Marsha J 9521<br />

Finkelstein, Sydney D.<br />

e21048, e21121<br />

Finley, David J. 7024<br />

Finley, Richard 7020<br />

Finn, Kathleen T. 6054<br />

Finn, Richard S. 8592<br />

Finnern, Henrik W. 6058<br />

Finney, Lindsey H. 618<br />

Finocchiaro, Gaetano 2083<br />

Finocchiaro, Giovanna 7061,<br />

7585, e18104<br />

Fins, Joseph J 9121<br />

Fintak, Patricia A 5082^<br />

Fioravanti, Antonella e13098<br />

Fioravanti, Suzanne 3103<br />

Fioraventi, Suzanne 10566<br />

Fiore, James 8547<br />

Fiore, John Joseph 7052<br />

Fiore, Marco 10016, 10017,<br />

10053<br />

Fiorentini, Giammaria 4084<br />

Fiorentino, Stefania e15075<br />

Fioretti, Agnese Maria 645<br />

Fiorica, James TPS5113<br />

Fiorio, Elena 586<br />

Firdaus, Irfan LBA3501,<br />

4127<br />

Firvida Perez, Jose Luis<br />

e18146<br />

Firvida Perez, Jose Luis<br />

e15076, e15077, e18040<br />

Firvida, Jose-luis e15067,<br />

e17545<br />

Fiscella, Kevin 9091<br />

Fisch, Michael 2003, 6054,<br />

6076, 6080, 9016, 9061,<br />

9099, 9106, 9112,<br />

e15173, e19631<br />

Fischbach, Wolfgang 4090<br />

Fischer von Weikersthal,<br />

Ludwig 3519^, 3541,<br />

3588, TPS3639, e17516<br />

Fischer, Anna e15577<br />

Fischer, Christian 4128<br />

Fischer, Johannes C. e21017,<br />

e21020<br />

Fischer, Lars 8031<br />

Fischer, Mary 3620<br />

Fischer, Patricia 4532<br />

Fischer, Peter 2024<br />

Fischer, Thomas 6511<br />

Fischer, Victoria 6620<br />

Numerals refer to abstract number<br />

Fischer-Valuck, Benjamin W<br />

e17524<br />

Fishel, Jean 3016<br />

Fisher, Barbara 6130<br />

Fisher, Brian 1504<br />

Fisher, Cyril 10038, 10060<br />

Fisher, David 3516, 3530<br />

Fisher, Deborah 6044<br />

Fisher, Douglass 3567<br />

Fisher, George A. e14610<br />

Fisher, Maxine e11063<br />

Fisher, Richard I. 8001,<br />

e18536<br />

Fisher, Richard TPS4690,<br />

5571<br />

Fisher, Rosalie Anne<br />

TPS4679<br />

Fisher, Sheila e16007<br />

Fisher, William E. e14501<br />

Fishman, David 5016<br />

Fishman, Mayer N. TPS2613,<br />

4503, e15010<br />

Fishman, Peter 553, e15189<br />

Fishman, Pnina e14731<br />

Fishman, Sari e14731<br />

Fiskus, Warren 10549,<br />

10605<br />

Fithian, Andrew 10535<br />

Fitilev, Sergei B. 9060<br />

Fitoussi, Olivier 8014<br />

Fitzgerald, Barbara e19522<br />

Fitzgerald, Catherine A. 6071<br />

FitzGerald, David JP 2503^<br />

Fitzgerald, Kristina e12515<br />

Fitzgerald, Meghan 569, 571,<br />

e11002<br />

FitzGerald, S. P. e14022<br />

Fitzmaurice, Thomas F.<br />

TPS4689<br />

Fizazi, Karim 4510,<br />

LBA4518, 4519^, 4540,<br />

4554, 4558, LBA4567^,<br />

4574, 4621, 4642,<br />

TPS4689, TPS4691,<br />

TPS4693, 5028, e15049,<br />

e19514<br />

Flacco, Antonella e18023,<br />

e18101, e21094, e21096<br />

Flahavan, Evelyn e13096<br />

Flaherty, Keith T. 4056,<br />

4500, 8502^, 8503^,<br />

LBA8509, 8510, 8516,<br />

8526, 8561, 8562, 8567,<br />

8569<br />

Flaherty, Lawrence E. 8504,<br />

8521, 8525, 8527, 8567<br />

Flamaing, Johan 9035<br />

Flamen, Patrick 3559<br />

Flammiger, Anna e15168<br />

Flanagan, Louise 1042<br />

Flanigan, Robert Charles<br />

e15014<br />

Flavin, Aileen e14666<br />

Flechl, Birgit 2071<br />

Flechon, Aude e15009<br />

Flechtner, Henning 6002,<br />

6090, 7607<br />

Fleisher, Martin 4516, 4548,<br />

4562<br />

Fleishman, Stewart Barry<br />

CRA9013<br />

Fleming, Deborah L. e16561<br />

Fleming, Gini F. 534, 1010,<br />

5012, 5019, 5020<br />

Fleming, Mark T. 4586<br />

Fleming, Natanya e21022<br />

Fleming, Ronald Alan 3000<br />

Fleming, Saroj e16546<br />

Fleming, Steven 6052<br />

Fleming, Stewart e15078<br />

Flentje, Michael 5512, 7001<br />

Fleshner, Neil Eric 9131,<br />

e15118<br />

Fletcher, Julie 5046<br />

Fletcher, Kalen M 9037<br />

Fletcher, Sarah e15582<br />

Fleury, Joel e11019<br />

Flickinger, John Charles<br />

7040, 7054, 7055<br />

Flinn, Ian W. 6122, 6507,<br />

6509^, 6515^, 6518^,<br />

TPS6636, 8081<br />

Flinois, Alain e15198<br />

Flockhart, David A. 525<br />

Flohr, Penelope 10525<br />

Floquet, Anne 5018, 5028,<br />

9012, e15535<br />

Flor, Maria Jose 10052<br />

Flora, Douglas B. 4127,<br />

7100, 7567<br />

Florance, Allison M. TPS661,<br />

10532<br />

Florero, Marilyn e11032<br />

Flores, Aurea M. 5065<br />

Flores, Elsa 8537<br />

Flores, Jacklyn J e19625,<br />

e19629<br />

Flores, Lucia Edith e15569<br />

Flores, Maria Regina C.<br />

e18047<br />

Flores, Natasha 505<br />

Flores, Rafael 6088<br />

Flores, Ronald A. e13591<br />

Flores-Estrada, Diana 7068<br />

Florescu, Anca e21092<br />

Florescu, Marie 2102,<br />

e17507, e18120, e18136<br />

Florez, Maria del Pino 4089<br />

Floriani, Irene LBA4001,<br />

5513, LBA7501<br />

Floris, Giuseppe 10030<br />

Flowers, Christopher 6122<br />

Flowers-Poole, Dava 10508<br />

Floyd, Leroy Cordero e18516,<br />

e19638<br />

Fluge, Oystein 8074<br />

Flugelman, Anath 1578<br />

Flynn, Jayme e14503<br />

Flynn, Joseph M. 6508,<br />

e18508<br />

Flynn, Patrick J. 1025, 2031,<br />

LBA4007, 6570, e14501<br />

Flörcken, Anne 4539,<br />

e15053<br />

Fløtten, Øystein 7027<br />

Foa, Paolo 5504^, 5513,<br />

7550, e14676<br />

Foà, Robin 6531<br />

Focan, C. N. J. 3547,<br />

e14006<br />

Fode, Kirsten 8545<br />

Foekens, John A 10504<br />

Foerster, Frank Gerhard 1077<br />

Fogelman, David R. 4038,<br />

4054<br />

Fogli, Laura K e13569<br />

Fojo, Antonio Tito 2548,<br />

e13122, e14064<br />

Fokt, Izabela e18557<br />

Folin, Annika 10083<br />

Folkerd, Elizabeth 9103<br />

Follana, Philippe LBA5002^<br />

Folprecht, Gunnar 3530,<br />

3562, 3574, 3591<br />

Foltran, Luisa 3612, e14040<br />

Foltz Boklage, Susan H<br />

e14106, e14141, e14146,<br />

e15183, e18097<br />

Fomin, Svetalna 1540<br />

Fonck, Marianne 9012,<br />

e14017, e14023<br />

Fong, Alan M. 2591<br />

Fong, Erika e13571<br />

Fong, Lawrence 2563, 2564,<br />

4593<br />

Fong, Mei Ka e14712<br />

Fong, Peter C.C. 5022,<br />

e15050, e15054<br />

Fong, Yuman 3577, 3620<br />

Fonkem, Ekokobe e18541<br />

Fonsatti, Ester 8513<br />

Fonseca, Eileen e16512<br />

Fonseca, Fernando L. A.<br />

e11059<br />

Fonseca, Rafael 8037,<br />

e18556<br />

Font Pous, Albert 4579,<br />

TPS4672, TPS4674,<br />

e15001<br />

Font, Albert 7095<br />

Fontaine, Jacques 10070<br />

Fontana, Andrea 629, 4627<br />

Fontana, Vincenzo e17533<br />

Fontanella, Caterina e13058,<br />

e13070<br />

Fontanelli, Rosanna 5096<br />

Fontanini, Gabriella 3521,<br />

3585, e14042<br />

Fontayne, Alexandre 5069<br />

Fontein, Duveken 10518<br />

Fontela, Antoinette e13585<br />

Foo, Leon Siang Shen 10051,<br />

e20509<br />

Fora, Ahmad Ali 3582<br />

Fora, Gianluca 5546, 5548<br />

Foran, James M. 8032<br />

Forastiere, Arlene A. 5576,<br />

e13504, e16061<br />

Forbes, John F. TPS1136<br />

Ford, Charles N e16010<br />

Ford, Daniel TPS4692^<br />

Ford, Eric C 2017<br />

Ford, Hugo e14100<br />

Ford, James 10619<br />

Ford, Jennifer 9586<br />

Ford, Kathy e13592<br />

Ford, Kendra e13568<br />

Ford, Laurie Ann 6565, 6602<br />

Foreman, Trudi e21142<br />

Forero, Andres 8065<br />

Forero-Torres, Andres 510,<br />

1006, 3069, 8027,<br />

e13079<br />

Forest, Valerie e15202<br />

Forestieri, Valeria 1554<br />

Forgeson, Garry e15054<br />

Forget, Frederic 9117<br />

Forgey, Robert TPS3110<br />

Formaker, Carl 2564, 4664<br />

Forman, Lora W. e14591<br />

Forman, Stephen J. 1592,<br />

6545, 6547, 8038, 8089,<br />

e18542<br />

Formenti, Silvia e11010,<br />

e11064, e11529<br />

Formento, Jean-Louis 2537^<br />

Formento, Patricia 2537^<br />

Formica, Serena 10087<br />

Formiga, Maria Nirvana<br />

e14728<br />

Formisano, Barbara e21118<br />

Formisano, Luigi e13509,<br />

e15034<br />

Fornarini, Giuseppe e15185<br />

Fornaro, Lorenzo 3555, 3585<br />

Forner, Dirk 5005<br />

Fornier, Monica Nancy 10514<br />

723s


Foroni, Chiara e11546<br />

Foroni, Letizia 10550<br />

Foroutan, Parastou 10084<br />

Forsberg, Goran 4550<br />

Forschner, Andrea 8526<br />

Forse, Armour R. e16554<br />

Forshew, Tim 544<br />

Forstbauer, Helmut 1072,<br />

1602<br />

Forster, Lourdes 9565<br />

Forstpointner, Roswitha 4072<br />

Forsyth, Michael T. e15089<br />

Forsyth, Peter A. J. 2099<br />

Forsythe, Laura Pence 9130<br />

Fort, Pauline e19555<br />

Fortin, Andre 5502, 5530<br />

Fortpied, Catherine 8002<br />

Fossa, Sophie D. 1536,<br />

LBA4512, 4551, 4637<br />

Foster, Julia 6606, 6621<br />

Foster, Matthew Charles<br />

e13074<br />

Foster, Nathan R. 7073,<br />

7605<br />

Foster, Richard 4586<br />

Foszczynska-Kloda,<br />

Malgorzata e21099<br />

Fotheringham, Lori e13581<br />

Fotis, Michael A 2532<br />

Fotopoulou, Christina 5005,<br />

5034, 5053, 5070, 5071,<br />

5080, 5087, e15520,<br />

e15531, e19597<br />

Fotovati, Abbas e13123<br />

Foucher, Pascal TPS7111<br />

Fougeray, Ronan 4522, 4525,<br />

e15007<br />

Foukakis, Theodoros 10083,<br />

e11510<br />

Fouladi, Maryam 9541, 9581<br />

Foulis, Alan K e14117<br />

Foulon, Veerle 4622<br />

Fountzilas, George 573, 592,<br />

5033, 10575, e15040,<br />

e21018<br />

Fourchotte, Virginie e15524,<br />

e15535<br />

Fourme, Emmanuelle TPS662<br />

Fournel, Marielle e13602,<br />

e13604<br />

Fournel, Pierre 7574, e16560<br />

Fournel-Frederico, Cécile<br />

2605<br />

Fournier, Charles 10020,<br />

e11012, e11049, e14560<br />

Fournier, Marion e13007<br />

Fournier, Nathalie 5069<br />

Fourrier-Réglat, Annie e14168<br />

Fouyssac, Fanny 9517<br />

Fowler, Jeffrey e15585<br />

Fowler, Nathan Hale 6518^<br />

Fox, Catharine e19522<br />

Fox, Elizabeth 9500, 9576<br />

Fox, Floyd E. TPS4675^<br />

Fox, Josef J. 4517, 4548<br />

Fox, Judith Ann TPS6637<br />

Fox, Kevin R. 6000<br />

Fox, Paul e14574<br />

Fox, Richard TPS4150<br />

Fox, Stephen B. TPS1136,<br />

8520, e21111<br />

Fox, Tara 7536<br />

Foxhall, Lewis E e19625,<br />

e19629<br />

Foxwell, Tyler e14689<br />

Fra, Joaquin 10052<br />

Fracasso, Paula M. 3047<br />

Fraccon, Anna Paola e13019<br />

Fradique, Antonio CALDEIRA<br />

e14673, e14713<br />

Fragandrea, Ioanna e20002<br />

Fram, Robert J 3539<br />

Frame, Sheelagh 3053<br />

Frampton, Garrett 1015,<br />

4649, 7529, 10524<br />

Franasiak, Jason 5009, 5092<br />

Franc, Benjamin L. e21026<br />

Franceschi, Enrico 2057,<br />

2061, 4017<br />

Franceschini, Maria Cristiana<br />

e17533<br />

Francesconi, Alessandra<br />

Bastian 8538<br />

Francescutti, Valerie e14172<br />

Francese, Valentina e11524<br />

Franchi, Giulia Maria 1604<br />

Franchina, Tindara e11056<br />

Francia, Giulio e11061<br />

Francis, Adele TPS665,<br />

e11062<br />

Francis, Prudence A.<br />

TPS1136<br />

Francis, Stephen 8512,<br />

8539, TPS8603<br />

Francisco, Federico e14075<br />

Francisco, Liton 1592, 9505<br />

Franck, Anne 8089<br />

Franco, Maria Isete Fares<br />

e14657<br />

Franco, Rebeca e11004,<br />

e15513, e19621<br />

Franco, Vito 8006<br />

Franco, Vivian I 9530<br />

Franco-Lie, Isabel e14637<br />

François, Eric e14168<br />

Francois, Sylvie 6534<br />

Franekova, Veronica e18077<br />

Frangou-Plemenou, M e13556<br />

Frank, Douglas TPS5601,<br />

e16026<br />

Frank, Konrad 1533, 10526,<br />

CRA10529<br />

Frank, Steven J. 5561, 5589<br />

Franke, Fabio A. 7503, 7506<br />

Frankel, Arthur E. 2505<br />

Frankel, Paul Henry 1010,<br />

2538, 3108, 10545,<br />

10574, e14182, e15114,<br />

e16563<br />

Frankel, Stanley 2504<br />

Franklin, Wilbur A. 7504,<br />

7532, 7534, 7552, 7589,<br />

10580<br />

Frankum, Jessica 3050<br />

Frantziyantz, Elena e19049<br />

Franz, Bettina 2502<br />

Franz, David Neal 10619<br />

Franzem, Melanie e14138<br />

Franzen, Lars e14531<br />

Fraser, Alison M 9561<br />

Fraser, Christopher 9547<br />

Fraser-Browne, Carol<br />

TPS4690<br />

Frassica, Deborah Anne 1128<br />

Frassineti, Luca LBA4001<br />

Frassoldati, Antonio TPS4151<br />

Frati, Luigi e14096<br />

Frattini, Mark G. TPS2619,<br />

6613<br />

Frattini, Milo e21018<br />

Frattini, Silvia 645<br />

Frazer, J. Kimble 9501<br />

Frazier, A. Lindsay 9523,<br />

9539<br />

Frazier, Kenneth 4085,<br />

e14564<br />

Frear, Meghan e15154<br />

Freas, Elizabeth 3017<br />

Frébourg, Thierry e14010<br />

Fredeking, Arden e19000<br />

Frederick, Mitchell J. 5524<br />

Fredrickson, Jill O. e13000<br />

Freedland, Stephen J. 4670<br />

Freedman, Matthew L 4543<br />

Freedman, Matthew 4635<br />

Freedman, Rachel A. 1100<br />

Freeman, Burgess B. e13584<br />

Freeman, Carol e14098<br />

Freeman, Daniel J. 3580<br />

Freeman, Marc 6049<br />

Freemantle, Sarah J. e13567<br />

Frees, Melanie 3056<br />

Fregnani, José Humberto<br />

e20506<br />

Freiberg-Richter, Jens<br />

TPS3639<br />

Freier, Werner 4539, e14138<br />

Freilich, Bradley L e14632<br />

Freilone, Roberto 8006<br />

Freiman, Joel 4085, e14564<br />

Freitas, Paulo Henrique Aires<br />

643, e18078<br />

Freixenet, Jorge 7011<br />

Freixinos, Víctor e15575<br />

Frenay, Marc 2, e12502,<br />

e20006<br />

French, Audrey L. 1611,<br />

8085<br />

French, Pim 2<br />

Frenel, Jean-Sebastien 601,<br />

2082<br />

Frenkel, Eugene P. 2077,<br />

3100<br />

Frenken, Bettina 10600<br />

Fresno-Vara, Juan Angel<br />

e11081<br />

Freter, Carl 1586, 6561,<br />

e11031<br />

Fretwell, Jo 9589<br />

Freund, Karen M 6097<br />

Frey, Noelle V. 6579, e18540<br />

Freyer, Craig e18509<br />

Freyer, Gilles 529, 636,<br />

8068, 9011, 9079, 9080,<br />

e15182<br />

Frezza, Anna Maria 10063,<br />

e14135<br />

Friard, Sylvie TPS7112,<br />

e18089<br />

Frias, Jesus e13085<br />

Friberg, Gregory R. 3009<br />

Friboulet, Luc 10556<br />

Frick, Kevin D. 6026<br />

Fricke, Harald 2034<br />

Fricke, Lothar 9033<br />

Frickh<strong>of</strong>en, Norbert e15026<br />

Fridrik, Michael A. 4074<br />

Fried, Daniel B. e17550<br />

Fried, Daniel B e17547<br />

Friedberg, Jonathan W.<br />

6515^, 8001, TPS8109,<br />

e18530<br />

Friedbichler, Katrin 3092<br />

Friedland, Julie C 3090<br />

Friedlander, Michael 3048,<br />

4531, 5001, 5022, 5081,<br />

TPS5116, 9044<br />

Friedlander, Terence W. 4593<br />

Friedman, Allan H. 2090^,<br />

2094^, 2095^<br />

Friedman, Danielle Novetsky<br />

CRA9513<br />

Friedman, Debra L. 9558<br />

Friedman, Erica Brooke 8565,<br />

e19018<br />

Friedman, Henry S. 2027,<br />

2074^, 2090^, 2094^,<br />

2095^<br />

Friedman, Kent P e11522<br />

Friedman, Sue 1608<br />

Friedmann, Alison M. 9502,<br />

9585<br />

Friedrich, Matthias 2504<br />

Friend, Julia C. TPS4680<br />

Friese, Klaus 554, 1072,<br />

1081, 1114, e15529<br />

Friesland, Signe e14531<br />

Friess, Helmut e14625<br />

Frigeri, Ferdinando 8066,<br />

8083<br />

Frimodt-Moller, Bente 7554<br />

Frisbee-Hume, Susan 9002,<br />

9023, 9025, e19599<br />

Frisinghelli, Michela e14711<br />

Frizziero, Melissa 4076,<br />

e14661<br />

Froehling, Stefan 6519<br />

Frohlich, Mark Walter e15120<br />

Froim, Doriana 1055<br />

Frost, Debra 1545<br />

Frost, Gregory I 2579<br />

Frost, Michael 4632, 10619<br />

Fruchart, Christophe 6633<br />

Fruehauf, John P. 8521,<br />

8524<br />

Fruehauf, S. e13100^<br />

Fruth, Briant 5025, 8013<br />

Frutiger, Yvette M. e18534<br />

Fryar, Beverly B. 3085<br />

Frydenberg, Mark TPS4690<br />

Frye, Sasha TPS3111<br />

Fryer, Christopher 9529<br />

Frödin, Jan-Erik 7539<br />

Fu, Cheng 4628<br />

Fu, De Liang e14651<br />

Fu, Haian 3094<br />

Fu, Jack Brian e19509<br />

Fu, Jing 8099<br />

Fu, Ling 5575<br />

Fu, Ma<strong>of</strong>u e13545<br />

Fu, Maoyong 1052<br />

Fu, PingFu TPS655,<br />

TPS1616, 7087<br />

Fu, Qiang e14716<br />

Fu, Rochelle 2040<br />

Fu, Shi-bo e13558<br />

Fu, Siqing 3106, 3107^,<br />

4639, 8551, 10510,<br />

10607, e13020, e13101,<br />

e13506, e13595<br />

Fu, Zhen-fu e13558<br />

Fucek, Ivica 8059, e18532<br />

Fuchs, Barry e19641<br />

Fuchs, Charles S. 3537,<br />

4027, 4042, 4112, 4125,<br />

TPS4135, e19586<br />

Fuchs, Eva-Maria 607<br />

Fuehrer, Kim e16549<br />

Fueki, Naoto e18037<br />

Fuentes Pradera, Jose<br />

e11075<br />

Fuentes, Eloisa 10576<br />

Fuentes, M Josefa 10555<br />

Fuentes, Stephane e19539<br />

Fuerhauser-Rappaport,<br />

Christine 1537<br />

Fuerst, Sophie 5007<br />

Fugazza, Clara 4017<br />

Fuhr, Uwe 3044<br />

Fujii, Hir<strong>of</strong>umi 5542<br />

Fujii, Kazuhiko e18059<br />

Fujii, Masato 5542<br />

Fujii, Satoshi 585, e14038<br />

Fujii, Shinji e13072<br />

724s Numerals refer to abstract number


Fujii, Shoichi 3569, 3607,<br />

e14047, e14050, e14624<br />

Fujii, Yasuhisa e15038<br />

Fujiki, Yoshitaka e11017<br />

Fujimori, Minoru e13037<br />

Fujimori, Tamaki e14635<br />

Fujimoto, Chinatsu 3045<br />

Fujimoto, Hiroyuki e15098<br />

Fujimoto, Junya 7023, 7078<br />

Fujimoto, Yasushi e16032<br />

Fujimoto, Yoshiya e14145<br />

Fujimoto-Ouchi, Kaori<br />

e13502, e18064, e18091<br />

Fujino, Katsuki 3079<br />

Fujioka, Tomoaki 10520<br />

Fujisawa, Masato e15048<br />

Fujisawa, Tomomi TPS1147<br />

Fujita, Shiro 7528, 10610,<br />

e18025, e18134<br />

Fujita, Tomoyuki e13037<br />

Fujita, Yuka 7561, 7571,<br />

e18135<br />

Fujitani, Kazumasa 4002<br />

Fujiwara, Keiichi 3084,<br />

5087, 5103<br />

Fujiwara, Toshiyoshi 1063<br />

Fujiwara, Yasuhiro 3084,<br />

10519, e11065, e19535<br />

Fujiwara, Yoshiro e18027<br />

Fujiwara, Yutaka e16034<br />

Fujiyama, Shigetoshi e14622<br />

Fuke, Satoshi e18135<br />

Fuks, David 3609<br />

Fukuda, Haruhiko 3524,<br />

5542, 6112, 7003, 7017<br />

Fukuda, Masaaki 7515<br />

Fukuda, Minoru 7569<br />

Fukuda, Satoko e15065<br />

Fukuda, Shunichi 7080<br />

Fukuda, Takahiro 6533<br />

Fukui, Iwao e15038<br />

Fukumura, Dai 1026<br />

Fukunaga, Yosuke 3625,<br />

e14145<br />

Fukuoka, Junya TPS659,<br />

e17540<br />

Fukuoka, Kazuya 7031, 7080<br />

Fukuoka, Masahiro 7521<br />

Fukushima, Hiroki e19583<br />

Fukushima, Julia Tizue<br />

e15562<br />

Fukushima, Masao e21031<br />

Fukutomi, Akira 4031, 4035<br />

Fuller, Clifton David 5561,<br />

5589<br />

Fuller, Courtney 9110<br />

Fuller, Greg 2011<br />

Fulp, William J 6100, 7024<br />

Fumagalli, Caterina 4120<br />

Fumagalli, Debora 507,<br />

e21010<br />

Fumagalli, Elena 10053<br />

Fumoleau, Pierre 596, 604,<br />

3026<br />

Funahashi, Yasuhiro 5518,<br />

5591, e13511, e19055<br />

Funakoshi, Akihiro 4035<br />

Funakoshi, Tohru 10578<br />

Funakoshi, Yohei e16034<br />

Funchain, Pauline 5573<br />

Fung, Chunkit 1536<br />

Fung, Henry C. 6552,<br />

e16503<br />

Fung, Hua TPS1616<br />

Funke, Roel Peter TPS2616,<br />

e13000, e13113, e13114<br />

Furge, Kyle A. 4624<br />

Furini, Lara e12037<br />

Furlanetto, Jenny 630<br />

Numerals refer to abstract number<br />

Furman, Richard R. 6507,<br />

6518^<br />

Furman, Wayne Lee 9534<br />

Fursse, Jo TPS9154<br />

Furugaki, Koh e18091<br />

Furukawa, Masayuki 4031<br />

Furuse, Junji 4031, 4035,<br />

4046<br />

Furuya, Kenichi e15579<br />

Furuya, Luis e12043<br />

Furuyama, Hiroaki e14518<br />

Fury, Matthew G. 5509^,<br />

5514, 5537, 5545^<br />

Fusco, Anne 8598<br />

Fuse, Nozomu 3079, e14510<br />

Fusi, Alberto 5572, e18010<br />

Fuss, Martin 5596^<br />

Fuste, Pere e13540<br />

Fuster, Jose 4119<br />

Futagawa, Shunji e14004<br />

Futran, Neal 5503<br />

Fuwa, Nobukazu 5556<br />

Fuxius, Stefan e15520<br />

Fyfe, David Walter LBA4001<br />

Färnegårdh, Katarina e13518<br />

G<br />

G, Janaki M e11536<br />

G, Rajeev A e11536<br />

Gaafar, Rabab Mohamed<br />

7081<br />

Gaal, Karl e18542<br />

Gabarre, Jean 8058<br />

Gabbanini, Massimo 5096<br />

Gaber, Germaine 1069<br />

Gabitzsch, Elizabeth 2585<br />

Gabow, Aaron e19647<br />

Gabrail, Nashat Y. e15557<br />

Gabram, Sheryl G. A. 7048<br />

Gabriele, Pietro e15133<br />

Gabrielson, Edward 10509<br />

Gabril, Manal Y. 4613, 4633<br />

Gabrilove, Janice Lynn 8032<br />

Gadaleta, Caldarola Gennaro<br />

e21113<br />

Gadaleta, Cosmo Damiano<br />

e21113, e21134<br />

Gadaleta, Gabriella e19641<br />

Gadaleta-Caldarola, Gennaro<br />

e21134<br />

Gadashova, Ayten e13535<br />

Gadbaw, Brian Christopher<br />

e19047<br />

Gadd, Michele 1122<br />

Gadellaa-van Hooijdonk,<br />

Christa G.M. 2596<br />

Gadgeel, Shirish M. 2081,<br />

7005, 7063, 7593,<br />

e17528, e21057<br />

Gadi, Vijayakrishna K.<br />

TPS3109<br />

Gadisseur, Alain P. 2506<br />

Gafford, Philip e14045,<br />

e14046<br />

Gagliato, Debora De Melo<br />

643, e18078<br />

Gagnier, Paul TPS4689,<br />

TPS4691<br />

Gagnon, Bruno e18098<br />

Gagnon, Patrick James 5596^<br />

Gagnon, Robert C. 4605,<br />

8518<br />

Gago, Jose Luis 4579<br />

Gagua, Irina Rezo e15506<br />

Gahramanov, Seymur 2077<br />

Gaiger, Alexander 2071<br />

Gaillard, Isabelle 8002<br />

Gaines, Natalie e15103<br />

Gaion, Fernando 9005<br />

Gaita, Fiorenzo e15192<br />

Gajate, Pablo e20513<br />

Gajavelli, Srikanth e14019<br />

Gajdosik, Martina Srajer<br />

e21116<br />

Gajendra, Smeeta 6593<br />

Gajewski, Thomas 2500^,<br />

2561, 2582, 8525, 8547<br />

Gajjar, Amar J. 9518, 9531<br />

Gajra, Ajeet 6572, 9034,<br />

9109, 9123, e16047,<br />

e18133, e18535<br />

Gakis, Georgios e19021<br />

Gal, Anthony A 7097<br />

Gal, Jocelyn 6633, e16044<br />

Gal, Tamas e21000<br />

Gal, Thomas e16035<br />

Gala, Marianna e11052,<br />

e13539<br />

Galais, Marie-Pierre LBA4003<br />

Galamaga, Robert W. e12002<br />

Galan, Antonio 7095, 10608,<br />

e11535, e21088<br />

Galanina, Natalie 10558<br />

Galanis, Evanthia 2004,<br />

2031, 2032<br />

Galant, Christine e21010<br />

Galazan, Lior 1564<br />

Galcheva-Gargova, Zoya 4056<br />

Gale, Davina 544<br />

Galetta, Domenico 7023,<br />

7550<br />

Galiauskas, Raimundas<br />

e14111<br />

Galibert, Marie-Dominique<br />

e19037<br />

Galicia, Ignacio e13085<br />

Galicier, Lionel 8005<br />

Galimberti, Fabrizio 7022<br />

Galimberti, Sara 8006<br />

Galizia, Danilo 10066,<br />

e15073, e20512<br />

Galizia, Eva e14663<br />

Gall, Valerie e19502<br />

Gallach, Sandra 7058, 7060,<br />

10594<br />

Gallagher, David James<br />

e19601<br />

Gallagher, David J 1543,<br />

e12031, e12038, e15022<br />

Gallagher, Emily R. 6004,<br />

6048, 6062, 6106, 9030,<br />

9101<br />

Gallagher, Fergal e12038<br />

Gallagher, Maryann 4596<br />

Gallagher, Robert E e15191<br />

Gallamini, Andrea 8077,<br />

e13093<br />

Gallarati, Chiara 3003<br />

Gallardo Rincon, Dolores<br />

e15527<br />

Gallardo, Elena e14733<br />

Gallardo, Enrique 4580,<br />

4582, 4585, e15041<br />

Gallardo, Humilidad F. 2518,<br />

8583<br />

Galle, Peter Robert TPS3641^<br />

Gallego Jimenez, Inmaculada<br />

e11075<br />

Gallego Plazas, Javier 3571,<br />

10547<br />

Gallego Rubio, Oscar 5590,<br />

5595<br />

Gallego, Javier 3618, e14041<br />

Gallego, Oscar 2045, 10052<br />

Gallego, Rosa 3536, 3553,<br />

4079<br />

Gallegos Sancho, Isabel<br />

e11074<br />

Gallegos, Jessica 9134<br />

Gallen, Manuel 3586,<br />

e14097<br />

Gallenstein, Donna 8582<br />

Galleta, Laura 5073<br />

Gallez-Hawkins, Ghislaine<br />

8089<br />

Gallick, Gary E. 3561,<br />

e15151<br />

Galligioni, Enzo LBA5003,<br />

e14711, e15160, e21102<br />

Gallinger, Steve 5026<br />

Gallinger, Steven 3567,<br />

4058, 4109, e14535,<br />

e14592<br />

Gallinson, David H. TPS3640<br />

Gallo, Aly e11066<br />

Gallo, Ciro TPS4151,<br />

LBA5003<br />

Gallo, Enzo 1050<br />

Gallo, Giuseppe Alberto<br />

e18564<br />

Gallois, Anne 2570, 8571,<br />

e21081<br />

Gallop-Evans, Eve 8056<br />

Gallot, Lillia 8080<br />

Galloway, Thomas J 10064<br />

Galmarini, Carlos 10047<br />

Galmarini, Felipe e13105<br />

Galmiche, Marie e11048<br />

Galsky, Matt D. 4516, 4522,<br />

4524, 4525, 4586, 4603,<br />

4623, TPS4676, 6065,<br />

e21016, e21032<br />

Galtes, Susana e15111<br />

Galve Calvo, Elena e11074<br />

Galvez, Jose 2531<br />

Gamazon, Eric R 9516<br />

Gambacorti-Passerini, Carlo<br />

6503, 6511, 6512<br />

Gambara, Guido e15549<br />

Gambardella, Antonio e14676<br />

Gambaro, Alessandra 3570<br />

Gamble, Mary 4101<br />

Gamble, S. Ellen 9575<br />

Gambol, Teresa E 4557<br />

Gamerman, Victoria e19054<br />

Gámez-Pozo, Angelo e11081<br />

Gammaitoni, Loretta 10066<br />

Gammoh, Emily 2578<br />

Gample, Tina LBA4001<br />

Gamucci, Teresa e12514,<br />

e14082, e18065, e18072<br />

Gamulin, Marija e15031<br />

Gan, Christine M. e15514,<br />

e15578<br />

Gan, Gregory 7526<br />

Gan, Hui Kong 3000, 5587<br />

Gan, Jacek e19047<br />

Ganai, Sabha 10090<br />

Ganapathi, Ram N. 5034,<br />

5070<br />

Gandara, David R. 1010,<br />

3023, 3078, 5020, 7005,<br />

7018, 7083, 7517, 7552,<br />

7570, 7582, 7594, 7599,<br />

TPS7619, e17537<br />

Gandhi, Ajeet 2072<br />

Gandhi, Arpita e14124,<br />

e14727<br />

Gandhi, Jitendra G. 4656<br />

Gandhi, Leena 3028, 3053,<br />

TPS3118, 7099, 7509,<br />

7543<br />

Gandhi, Varsha 6528<br />

Gandikota, Neetha 5578<br />

Gandini, Sara 519, e19035,<br />

e19036<br />

Gane, Nicolas 9080<br />

725s


Gang, Wu 7091, e14511<br />

Gangaram, Anjali e21069<br />

Gangopadhyay, Sudeshna<br />

e13030, e13533, e18043<br />

Ganguli, Arijit e12515,<br />

e13038<br />

Ganguli, Sushila 10060<br />

Ganguly, Siddhartha 10549,<br />

10605, e13568<br />

Ganjoo, Kristen N. 8081,<br />

10036<br />

Ganly, Ian 5545^, 5562<br />

Gannon, Philippe O. e15202<br />

Gano, Katherine 8053<br />

Ganschow, Pamela 1553<br />

Ganser, Arnold e15190<br />

Ganten, Tom M 4115<br />

Ganti, Apar Kishor e17549<br />

Ganz, Patricia A. TPS1142,<br />

3545, 6006, e11003<br />

Ganzer, Roman e15194<br />

Ganzinelli, Monica LBA7501<br />

Gao, Bo 550<br />

Gao, Chong e13505<br />

Gao, Feng 1048, 3077,<br />

5013, 5101, e14584,<br />

e15528<br />

Gao, Guanghui e18123<br />

Gao, Harry 10545<br />

Gao, Hui e13582<br />

Gao, Jianfei 9017<br />

Gao, Jianjun 2520^<br />

Gao, Jing e14122, e14604<br />

Gao, Li 5535<br />

Gara, Michelle TPS4134<br />

Garambois, Veronique 5069<br />

Garassino, Isabella M.G. 7585<br />

Garassino, Isabella e18104<br />

Garassino, Marina Chiara<br />

LBA7501<br />

Garay, Carlos A. 4608<br />

Garbay Decoopman, Delphine<br />

10057<br />

Garbe, Claus 4034, 8502^,<br />

LBA8509, 8512, 8517,<br />

8526, e19031<br />

Garber, Judy Ellen 576,<br />

1009, 1506, 1547, 10097<br />

Garbo, Lawrence E. 3023,<br />

TPS3111, 7502<br />

Garcia Alfonso, Pilar e14058,<br />

e14671<br />

Garcia Alonso, Mirta TPS4672<br />

Garcia Aranda, Mariola 602<br />

Garcia Arroyo, Francisco<br />

Ramon 8062<br />

Garcia Carbonero, Iciar<br />

e11545, e15144<br />

Garcia de Santiago, Clara<br />

e11028<br />

Garcia del Muro, Xavier 3096,<br />

TPS4672, TPS4674,<br />

TPS4685, 10052<br />

Garcia Escudero, Ramon<br />

4584<br />

Garcia Gerardi, Carlos<br />

e15507, e15508<br />

Garcia Gonzalez, Maria<br />

10512, 10595<br />

Garcia Lopez, Maria Jose<br />

e11074<br />

Garcia Nieto, Sandra e19576<br />

Garcia Palomo, Andres<br />

e11074<br />

Garcia Sanchez, Jose e17545,<br />

e18049, e18053<br />

Garcia Valdecasas, Barbara<br />

10555<br />

Garcia, Agustin 1010, 1065,<br />

3054, TPS3111, e13084,<br />

e13531<br />

Garcia, Alicia e18030<br />

Garcia, Antonia e15111<br />

Garcia, Beatriz 2527<br />

Garcia, Camilo 3559<br />

Garcia, Damian 1570, 3586<br />

Garcia, Helena 10608<br />

Garcia, Jorge A. 4609,<br />

TPS4684, e15095, e15208<br />

Garcia, Jose e19643<br />

Garcia, Katherine e15571<br />

García, Lina 4040, e11517<br />

Garcia, Olga 8005<br />

Garcia, Reynaldo e13074<br />

García, Yolanda 5068,<br />

e15575<br />

Garcia-Albeniz, Xabier 3536,<br />

3553, 4079, e14041<br />

Garcia-Alonso, Angel<br />

LBA4001, e14587<br />

Garcia-Blanco, Mariano<br />

10533<br />

Garcia-Bueno, Jose Maria<br />

e18015<br />

Garcia-Caballero , Tomas<br />

4089<br />

Garcia-Campelo, M. Rosario<br />

e12012, e18131<br />

Garcia-Carbonero, Rocio<br />

TPS3637, 4119<br />

Garcia-Donas, Jesus<br />

TPS4674, TPS4685,<br />

TPS4688, 5068, 10546,<br />

e15041, e15575<br />

García-Estevez, Laura 602,<br />

e11006, e11517<br />

Garcia-Foncillas, Jesus 4079,<br />

5520, 10512, 10595,<br />

e14057, e14147, e15094,<br />

e18069, e18106, e21015<br />

García-García, Elena 4040<br />

Garcia-Garre, Elisa e11024<br />

Garcia-Gomez, Ramon<br />

e18011, e18015<br />

Garcia-Gonzalez, Araceli<br />

e19589<br />

Garcia-Grossman, Ilana<br />

Rebecca 4581^, e15022<br />

Garcia-Hernandez, Carmen<br />

TPS6640<br />

Garcia-Hernandez, Jorge<br />

4123, e19581<br />

Garcia-Manero, Guillermo<br />

6520, 6544, 6558, 6578,<br />

6587, 6597, 6603, 6607<br />

Garcia-Martinez, Elena<br />

e11024, e21088<br />

Garcia-Pinon, Francisco<br />

e18053<br />

Garcia-Ribas, Ignacio 1011<br />

Garcia-Vargas, Jose E.<br />

LBA4512, 4551<br />

Garcias-Espana, Carmen<br />

e11036<br />

Garde, Javier 4587, e18049,<br />

e18053<br />

Garden, Adam S. 5561, 5589<br />

Garden, Benjamin C e18571<br />

Garderet, Laurent 8014<br />

Gardiman, Marina Paola 2049<br />

Gardin, Claude 6523<br />

Gardiner, Stuart K 9019<br />

Gardizi, Masyar 1533, 3044,<br />

TPS3115, 10526<br />

Gardner, Brian 7518<br />

Gardner, Ginger J. 5108<br />

Gardner, Lesa R. LBA3501<br />

Gardner, Lisa 6131<br />

Gardner, Olivia S. 3023,<br />

5065<br />

Gardner, Thomas 4650<br />

Garfield, David H. e18123,<br />

e19573<br />

Garg, Karuna 5030, 5043<br />

Garg, Madhur 4030, e16058<br />

Garg, Mandeep K e18009<br />

Garg, Naveen e19645<br />

Garg, Vishvas 2532, 4063<br />

Garibaldi, Elisabetta e15133<br />

Garin, Avgust e15025,<br />

e15029<br />

Garin, M. e11528<br />

Garino, Astrid 9108<br />

Garje, Rohan e15023<br />

Garland, Linda L. 7083<br />

Garlipp, Benjamin e14630<br />

Garmire, Lana 3630<br />

Garner, Elizabeth e14625<br />

Garnier, Alice 6534, 6542<br />

Garnier, Frederico 6534<br />

Gar<strong>of</strong>alo, Amanda TPS658<br />

Gar<strong>of</strong>alo, Rosemary 1132<br />

Garon, Edward B. LBA4,<br />

7002, 7543, 7589<br />

Garonzik, Samira Merali 2603<br />

Garosi, Vittorio Luigi 4050<br />

Garralda, Elena 3068<br />

Garraway, Levi A. 10502<br />

Garrett, Chris R. 3544,<br />

e13599, e19645<br />

Garrett, Karen 9019<br />

Garrett, Leslie A. e15545,<br />

e15568<br />

Garrett, William M.<br />

TPS6643^<br />

Garrido López, Pilar 1531,<br />

7540, e12000, e12012,<br />

e16029, e18055<br />

Garrido-Laguna, Ignacio 3057<br />

Garrison, Louis P e14525<br />

Gartrell, Benjamin Adam<br />

4623, TPS4676, 6065<br />

Garufi, Carlo e14661<br />

Gary, Kristen 9105<br />

Garza, Miguel e13500<br />

Garzaro, Massimiliano 5546,<br />

5548<br />

Garzon, Felix Tomas e18063^<br />

Garzotto, Mark 4511, 4663,<br />

10503<br />

Gasbarra, Rita e19033<br />

Gasbarrini, Antonio 4006,<br />

TPS4151<br />

Gasco, Amaya 7540, 7542,<br />

e18137, e18143<br />

Gascon, Pedro e13540<br />

Gascoyne, Randy D. 8003,<br />

8007, 8049<br />

Gasiorowski, Lukasz 10536<br />

Gaska, Tobias 8022<br />

Gaspar, Laurie E. 7018, 7526<br />

Gasparetto, Cristina 8012,<br />

8037, e18556<br />

Gasparini, Daniele e14654<br />

Gasparini, Giampietro e19612<br />

Gasparini, Mauro e15192<br />

Gasparini, Pierluigi 1007<br />

Gasparri, Mario e17555<br />

Gasparro, Donatello e15185<br />

Gass, Jennifer S. 1045<br />

Gass, Margery 1501<br />

Gastier-Foster, Julie M 9535<br />

Gat-Charlap, Amira 1564<br />

Gatalica, Zoran 1040, 4013,<br />

5523, 10630, e20507<br />

Gately, Kathy 637<br />

Gates, Mary R. 2566<br />

Gatica, Gabriela e15570<br />

Gato-Weis, Cara e14098<br />

Gatto, Lidia 10087<br />

Gattoni, Elisabetta 7084<br />

Gattuso, Paolo 572<br />

Gaucher, Alison 8530<br />

Gaucher, Christine 5069<br />

Gauchez, Philippe 2048<br />

Gaudet, Mia e15022<br />

Gaudy-Marqueste, Caroline<br />

8555, 8568, 8578<br />

Gaughan, Elizabeth Mary<br />

7579<br />

Gaughan, John P. e16022<br />

Gaulard, Philippe 8048<br />

Gauler, Thomas C. 4023,<br />

5516^, e15026<br />

Gaur, Rakesh 4019<br />

Gautam, Ajay 5526<br />

Gautam, Shiva e18541<br />

Gauthier, Eric 10020<br />

Gauthier, Genevieve 10094,<br />

e11076, e16571, e20511<br />

Gauthier, Marie-Pierre 8532<br />

Gauthier, Mélanie 1584,<br />

4018<br />

Gautschi, Oliver 2068, 3595<br />

Gava, Alessandro 5513<br />

Gavila, Joaquin 1011<br />

Gavina, Belen e14147<br />

Gavine, Paul 4124<br />

Gavrilovic, Dusica e21030<br />

Gavrilovic, Igor T. 2008<br />

Gawade, Prasad Laxman 9527<br />

Gawande, Jayant Shankar<br />

9559<br />

Gawel, Susan e13583<br />

Gay, E. Greer 6046, 6088<br />

Gaylor, Shari TPS3110<br />

Gazawi, Nidal e19540<br />

Gazial-Sovran, Avital e19015<br />

Gazzah, Anas 3012<br />

Gazzaniga, Paola e14096<br />

Geary, David e13106<br />

Geater, Sarayut Lucien<br />

LBA7500, TPS7614,<br />

TPS9151, e18063^,<br />

e19608<br />

Gebauer, Gerhard e21003<br />

Gebbia, Vittorio 7554,<br />

e18002<br />

Geberth, Matthias 1077,<br />

e19540<br />

Gebhardt, F e13597<br />

Gebhardt, Mark C. 9537<br />

Geboers, Inge 1129<br />

Gebski, Val 550, TPS1136,<br />

TPS3643, TPS4137,<br />

TPS4141, 4531, TPS4681,<br />

TPS5116, e13007<br />

Geddings, Julia E e14505<br />

Gee, Adrian P. 4643<br />

Geentjens, Krist<strong>of</strong> e13073<br />

Geffken, Mareike 10046<br />

Geffriaud-Ricouard, Christine<br />

e15161<br />

Gehl, Julie 2069<br />

Geho, David e13600^<br />

Gehrig, Paola A. 2591, 5009,<br />

5050, 5063, 5092,<br />

e13063, e15511<br />

Gehrke, Flavia de Sousa<br />

e11059<br />

Gehrmann, Mathias 10551<br />

Geiger, Jessica L. 8060<br />

Geisel, Juergen 4107<br />

Geiss, Romain 6110<br />

726s Numerals refer to abstract number


Geissler, Benjamin e15109,<br />

e21056<br />

Geissler, Michael 4023,<br />

4072, TPS7109<br />

Gelber, Richard D. 504, 9022<br />

Gelber, Shari I. 9100<br />

Gelber, Shari 9071<br />

Gelderblom, Hans 526,<br />

10006, LBA10008, 10009,<br />

10067, 10077, 10081,<br />

10518, e16526<br />

Gelet, Albert e15194<br />

Gelibter, Alain e14661<br />

Gellert, Klaus 4020<br />

Gellert, Lan L. 8003<br />

Gellhaus, Katharina 5053<br />

Gellrich, Sylke 8076<br />

Gelmon, Karen A. 538, 549^,<br />

582<br />

Gemba, Kenichi 7560<br />

Gemignani, Mary 575,<br />

e11531<br />

Gemma, Akihiko 7515, 7561,<br />

7563, 7565, e18129<br />

Gemma, Donatello e14082<br />

Gencer, Deniz 3541<br />

Genden, Eric 5578<br />

Genega, Elizabeth M. 4521<br />

Generali, Daniele Giulio 579,<br />

e11546<br />

Genestie, Catherine 641,<br />

e11551<br />

Genestreti, Giovenzio 7550,<br />

e17546<br />

Genin, Pascal 10517,<br />

TPS10634<br />

Gennari, Alessandra 519<br />

Gennari, Paolo e15550,<br />

e15553<br />

Gennatas, Konstantinos<br />

e13556<br />

Genova, Carlo 7577, e21065<br />

Gentilcore, Giusy 8573<br />

Gentile, Michelle Sara<br />

e18523<br />

Gentles, Andrew e21071<br />

Gentzler, Ryan D. e16548<br />

Genyk, Yuri e14679<br />

Geoerger, Birgit 9517, 9519,<br />

TPS9597^<br />

Ge<strong>of</strong>frois, Lionnel 4625<br />

Ge<strong>of</strong>froy, Francois J. 2032<br />

George, Ben e14613<br />

George, Binsah 3544<br />

George, Claude Paul e13588<br />

George, Daniel J. 4513,<br />

4549, 4550, 4612, 4655,<br />

4667, 10533, e15061,<br />

e15120<br />

George, Goldy 8551<br />

George, James N e17001<br />

George, Saby TPS4687,<br />

10609, e15042<br />

George, Suzanne 1547,<br />

5090, 10017, TPS10103<br />

George, Thomas J. TPS4136,<br />

e14612<br />

George, Timothy e18509<br />

Georgoulias, Vassilis 7564,<br />

e18105<br />

Geraci, Joseph 10622<br />

Geraldes, Catarina e12040<br />

Gerber, Bernd 541, 1023<br />

Gerber, David E. e13563,<br />

e18076<br />

Gerber, Linda 6123<br />

Gerber, Naamit Kurshan 9104<br />

Gerber, Peter Arne e11556<br />

Gercheva, Liana 8018^<br />

Numerals refer to abstract number<br />

Gercovich, Daniela 9038<br />

Gercovich, Felipe G. 9038,<br />

e11037, e15507, e15508<br />

Gercovich, Natasha e15508<br />

Gerdemann, Ulrike 2558<br />

Gerecitano, John F. 8054,<br />

9092<br />

Geredeli, Caglayan 638<br />

Geretti, Elena TPS663<br />

Gerger, Armin 3584, 3597,<br />

3604, 3622, 4026, 4088,<br />

e11018, e14034, e14052<br />

Gergich, Kevin 2512<br />

Gerigk, Ulrich 1533, 10526,<br />

CRA10529<br />

Gerken, Guido 4115<br />

Gerl, Arthur 4602<br />

Germano, Serena 617<br />

Germanos, Peter 7604<br />

Germaschewski, Fiona 5065<br />

Germenis, Anastassios<br />

e20002<br />

Gernone, Angela e15160<br />

Gerotzafias, Grigoris 1580<br />

Gerrard, Gareth 10550<br />

Gerratana, Lorenzo 1044,<br />

e13058, e13070<br />

Gerritsen, Winald R. 2562,<br />

TPS4689, TPS4691,<br />

e15112<br />

Gerry, Arianna Aldridge 9051,<br />

e16564<br />

Gerschenson, Mariana 9530<br />

Gershenson, David Marc<br />

1518, e15583<br />

Gershenwald, Jeffrey E. 8530,<br />

8536<br />

Gerson, Stanton TPS1616<br />

Gerstenblith, Meg R. 1603<br />

Gerstner, Elizabeth Robins<br />

2009, 2012, 2025, 2059,<br />

3036, TPS10635<br />

Gerstner, Gregory James<br />

e17021<br />

Gertz, Erik 4547<br />

Gertz, Morie A. 8043, 8092,<br />

8096, 8105, TPS8116<br />

Gerunda, Giorgio Enrico 4110<br />

Gervais, Radj 7522, 7542,<br />

7574, e18089<br />

Geschwind, Jean-Francois<br />

4114, e14581<br />

Gessert, David e18502<br />

Gettinger, Scott N. CRA2509,<br />

TPS2615, 7509, 7531,<br />

7533, 7599, e21106<br />

Getz, Gad 2020, 10502<br />

Geubels, Barbara M. 4528<br />

Geva, Ravit 1507, e21018<br />

Gewandter, Jennifer S. 9010,<br />

9014, 9028, 9113,<br />

e16056<br />

Gewirtz, Alexandra N. 3577<br />

Geye, Heather M e16010<br />

Geyer, Charles E. LBA506,<br />

LBA1000, 1025, TPS1142,<br />

9020<br />

Geyer, Mark B 6537<br />

Geyer, Susan Michelle 4039,<br />

6508<br />

Geynisman, Daniel M. 2561<br />

Gezgen, Gamze e11509<br />

Ghadessi, Mercedeh 4565<br />

Ghadimi, B. Michael 3600,<br />

e14161, e14167<br />

Ghai, Vikas e18500<br />

Ghalib, Mohammad Haroon<br />

3093, e14019<br />

Ghaneh, Paula 8564<br />

Ghanem, Hady 8103<br />

Ghanem, Ismael 642,<br />

e15001, e15552<br />

Ghazal, Hassan e18558<br />

Ghazalpour, Anatole 1040,<br />

10630, e15209, e20507<br />

Ghebremichael, Musie S.<br />

e13504<br />

Gherardi, Giorgio LBA7501<br />

Ghesquieres, Herve 2023<br />

Ghi, Maria Grazia 5513<br />

Ghim, Byoung-Chul 7053<br />

Ghinea, Nicolae e21138<br />

Ghio, Domenico 7076<br />

Ghiotto, Cristina e12037<br />

Ghnassia, Jean-Pierre e11542<br />

Ghobrial, Irene M. 8043<br />

Gholam, Pierre Michel<br />

e14581<br />

Ghonimi, Elham 8067<br />

Ghosh, Jaya e13068<br />

Ghosh, Sarbani Laskar<br />

e16021<br />

Ghosh, Sunita e15540<br />

Ghossein, Ronald A. 5514<br />

Ghushchyan, Vahram e16550<br />

Giacchetti, Sylvie e14006<br />

Giaccia, Amato 10629<br />

Giaccone, Giuseppe 7033,<br />

7103, TPS7609<br />

Giacobbe, Felicia 3570<br />

Giacomantonio, Carman A.<br />

e21090<br />

Giacomazzi, Juliana 1522,<br />

10593, e21042<br />

Giacomi, Nora e14598<br />

Giagini, Athina 5574<br />

Giagounidis, Aristoteles 6522,<br />

6527, 6610<br />

Giaj Levra, Matteo e18103<br />

Giamas, Georgios 3546,<br />

3597, 4088, e11018<br />

Giammarile, Fransesco 10049<br />

Giammicchio, Karen e16510<br />

Giampaglia, Marianna e18048<br />

Giampaolo, Bianchini 545,<br />

1012<br />

Giampieri, Riccardo e14040,<br />

e14086, e14561, e14663,<br />

e15074<br />

Gian, Victor 7035, 7567,<br />

7587, TPS8115<br />

Giani, Jorge e19569<br />

Giannarelli, Diana 630, 4541,<br />

8513<br />

Giannatempo, Patrizia 4507<br />

Gianni, Alessandro M. 4507<br />

Gianni, Lorenzo 9005<br />

Gianni, Luca LBA1, 532,<br />

545, e18096<br />

Giannini, Caterina 2031,<br />

2032<br />

Giannini, Edoardo G 4115<br />

Giannini, Giuseppe e14042<br />

Giannitto-Giorgio, Carmelo<br />

e18026<br />

Gianoncelli, Letizia 7061,<br />

7585, e18104<br />

Giantonio, Bruce J. 3614,<br />

4111, 6000<br />

Giardina, C 10554<br />

Giardini, Roberto e11546<br />

Gibbon, Darlene 3054,<br />

TPS3112<br />

Gibbons, Jay 3041<br />

Gibbs, John F. e14610<br />

Gibbs, Peter 3515, 3623,<br />

e16516<br />

Gibori, Hadas e14067<br />

Gibson, Diana 3000<br />

Gibson, Jeffrey B e17531^<br />

Gibson, Laura TPS655<br />

Gibson, Michael K. 5528,<br />

5541, e14689<br />

Gich, Ignasi 10555, e14104,<br />

e14552<br />

Gichuru, Gladwell Wantiru<br />

e20504<br />

Gierszewska, Magdalena 4562<br />

Giese, K. e13597<br />

Giessen, Clemens Albrecht<br />

3519^, 3603, TPS3639<br />

Gietema, Jourik A. 4132,<br />

4528, 10581, e15035<br />

Gietema, Joyce Q. 4528<br />

Gifoni, Markus e14183<br />

Giglio, Pierre 2003<br />

Gignac, Gretchen e15139<br />

Gigot, Jean Francois e14044<br />

Giguere, Jeffrey Kent 9061,<br />

9108<br />

Gil Deza, Ernesto 9038,<br />

e11037, e15507, e15508<br />

Gil Gil, Miguel J. 502, 2045<br />

Gil, I e19023<br />

Gil, Marta 3096<br />

Gil, Miquel 10608, e11535,<br />

e21088<br />

Gil, Roberto de Almeida<br />

e14027<br />

Gil, Thierry 3018^,<br />

TPS4692^<br />

Giladi, Moshe e14616<br />

Gilbert, Jill 5566<br />

Gilbert, Judith A. e13086<br />

Gilbert, Mark R. 2001, 2003,<br />

2019, 2029^, 2030, 2036,<br />

2064<br />

Gilbert, Ralph William<br />

TPS5600<br />

Gilbertson, Richard J. 9518<br />

Gilcrease, Glynn Weldon<br />

e14090<br />

Giles, Francis J. 3073,<br />

6619^, e15126<br />

Gill, Amandeep 3566<br />

Gill, Inderbir S e15060,<br />

e15171, e15212<br />

Gill, Ritu R. 10592<br />

Gill, Rosalynn D e21012,<br />

e21142<br />

Gill, Sandeep 5046<br />

Gill, Sharlene 6014, e14568,<br />

e14588<br />

Gillen, Jessica e15103<br />

Gillenwater, Heidi H. e13077<br />

Gillespie, Marion Boyd 5528<br />

Gillespie, Theresa Wicklin<br />

e15187<br />

Gillett, Cheryl 10586<br />

Gillies, Richard e13587<br />

Gillies, Robert 10084<br />

Gillies, Stephen e19047<br />

Gilligan, Timothy D. TPS4684<br />

Gillio, Alfred P 6537<br />

Gillis, Theresa A. 9047<br />

Gillison, Maura L. 5530,<br />

5566<br />

Gilman, Paul CRA9013<br />

Gilmore, James W. 569, 571,<br />

e11002, e14106, e14141,<br />

e14146, e15183, e18097<br />

Gilmore, Katherine R. 8091,<br />

9081<br />

Gilmore, Steven e19565<br />

Gilson, Eric 9011<br />

Gilts, Chelsea D. 9031<br />

727s


Gimenez Capitan, Ana 4068,<br />

10596, e14521, e18130,<br />

e18131, e18137, e18143<br />

Gimenez, Alejandra 10082<br />

Gimotty, Phyllis A. e19054<br />

Gimpel, Petra 10563<br />

Gimsing, Peter 8019<br />

Giner, Vicente e17545,<br />

e18049<br />

Ginés, Alba 9129<br />

Ginés, Àngels 3536<br />

Gingerich, Joel Roger 4514<br />

Gingrich, Jeffrey R. 4550<br />

Gino, Giancarlo 10055<br />

Ginsberg, Lawrence E. 5592<br />

Ginsberg, Michelle S. 3563,<br />

7052, e19647<br />

Ginsburg, Elizabeth S 9071<br />

Ginther, Charles 8592<br />

Giobbie-Hurder, Anita 8569,<br />

9084<br />

Giommoni, Elisa e18074<br />

Giordani, Erika e11039,<br />

e11514, e12510, e19620<br />

Giordani, Paolo 4084<br />

Giordano, Alessandra e11524<br />

Giordano, Antonio e13582<br />

Giordano, Carlo 5546, 5548<br />

Giordano, Laura 7061, 7585,<br />

e14672, e18104, e21139<br />

Giordano, Pasqualina e14130<br />

Giordano, Sharon Hermes<br />

537, 600, 612<br />

Giorgi, Francesca 9098,<br />

e14086<br />

Giotis, Angie e15180<br />

Giotta, Francesco 645<br />

Gioulbasanis, Ioannis e18098<br />

Giovannetti, Elisa 10611<br />

Giovanni, Antoine e16044,<br />

e16055<br />

Giovannini, Catia 3061<br />

Giovannini, Monica 7581<br />

Giovannoni, Mariella 3612<br />

Giovannucci, Edward 4555<br />

Giovinazzo, Hugh 5050<br />

Giralt, Jordi 5502<br />

Giralt, Sergio 6529, 6544,<br />

6546<br />

Giranda, Vincent L. TPS1138,<br />

2013, 3049, 3054<br />

Girard, Luc 7096, 10612,<br />

e18077<br />

Girard, Philippe TPS7111<br />

Girardi, Fabio e19595<br />

Girgis, Afaf 9128, e16507<br />

Giri, Dilip D. 1006<br />

Giri, Nagdeep e18093<br />

Giri, Neelam 5563<br />

Giri, Uma 10612<br />

Giri, Veda N. 1517<br />

Girodet, Didier e16036<br />

Girones, Regina e15144,<br />

e17545<br />

Giroux Leprieur, Etienne<br />

e18075, e18102<br />

Girre, Veronique TPS667<br />

Gisleskog, Per Olsson 2535,<br />

2594<br />

Gisselbrecht, Christian 8048<br />

Gisslinger, Heinz 6514^,<br />

6625^, 6626^, TPS6640,<br />

TPS6642^<br />

Gitlitz, Barbara Jennifer<br />

TPS7619, e13084<br />

Giubellino, Elena 10055<br />

Giudice, Aldo e11052,<br />

e13539<br />

Giuffrida, Dario 1604<br />

Giuliani, Costanza e18074<br />

Giuliano, Armando E. 1105<br />

Giulini, Stefano Maria 4110<br />

Giurescu, Marius 4050<br />

Giusca, Simona Eliza e15032<br />

Giustini, Lucio 9098, e14086<br />

Given, Barbara Ann e12500<br />

Given, Charles W e12500<br />

Gizzi, Marco e14044<br />

Gkiozos, Ioannis e17508,<br />

e17557<br />

Gladieff, Laurence 5017^<br />

Gladkov, Oleg 531, 7090,<br />

9125, e19587<br />

Gladson, Candece 2014<br />

Glantz, Michael J. TPS2107,<br />

e11544, e12005<br />

Glanz, Karen 1503<br />

Glare, Paul A. 9087, 9094,<br />

e19566<br />

Glas, Annuska 3065<br />

Glasberg, Joao e16568<br />

Glasgow, Amanda L. 9087<br />

Glaspy, John A. e19511<br />

Glass, Andrew e12024,<br />

e15197, e18107, e19630<br />

Glass, Bertram 6543, 8004<br />

Gleason, Charise 8100, 8101<br />

Gleave, Martin Edwin 4514,<br />

TPS4695<br />

Gleissner, Jochen e15195<br />

Glenjen, Nils e14531<br />

Gligorov, Joseph 568^, 641,<br />

1051, 1095, 5067<br />

Glimelius, Bengt 3508, 3538,<br />

3548, 3576, 4053, 8002,<br />

TPS10632, e14053,<br />

e14088<br />

Glimm, Hanno 6519<br />

Glisson, Bonnie S. 5592,<br />

7096, 7597<br />

Glitza, Isabella Claudia 9069<br />

Globig, Cordula e19579<br />

Glogowska, Iwona 10582<br />

Glogowski, Maciej e12011<br />

Gloor, Beat e14544<br />

Glover, Curtis L e15565<br />

Gluck, Iris e14143<br />

Gluck, Stefan 10597,<br />

e15107, e16525<br />

Gluz, Oleg 552<br />

Glynn, Sharon A. e15126<br />

Glynne-Jones, Robert 4004,<br />

4029<br />

Gnad-Vogt, Ulrike Senta<br />

2573^<br />

Gnani, Alessandra 6531<br />

Gnant, Michael 512, 514,<br />

539, 540, 542, 551, 559,<br />

10501<br />

Gnjatic, Sacha 2589,<br />

TPS8111, e11563<br />

Gnocchi, Chiara 10087<br />

Go, Mylene Sy e15522<br />

Go, Ronald S. 6074, e16555,<br />

e16561<br />

Gobet-Bourcelot, Sophie 9080<br />

Goda, Hiroyuki 5584<br />

Godard, Aurelie e19623<br />

Godbold, James H. 4524,<br />

6065, e21032<br />

Goddard, Karen 7020<br />

Goddard, Katrina e14160<br />

Godefroy, Emmanuelle 2570,<br />

e21081<br />

Godette, Karen D. 9122<br />

Godfrey, CJ 8541<br />

Godfrin, Yann e14650<br />

Godinez-Puig, Victoria 2532,<br />

4063, 6623<br />

Godley, Lucy A. 6562<br />

Godley, Paul Alphonso 4647,<br />

4658<br />

Godoy-Scripes, Paola e16058<br />

Godwin, John E. 6502<br />

Goebeler, Marie-Elisabeth<br />

2504, 6500^<br />

Goebell, Peter J. 4539,<br />

e15044<br />

Goehler, Thomas 640<br />

Goehring, Gudrun 6510<br />

Goekbuget, Nicola 6500^<br />

Goekkurt, Eray 2573^, 10552<br />

Goel, Ajay e14029<br />

Goel, Rakesh 1025<br />

Goel, Sanjay 3093, e14019,<br />

e14682<br />

Goel, Shom 1026<br />

Goeminne, Jean-Charles<br />

4622, e14060<br />

Goessl, Carsten Dietrich<br />

4510, 4642, e15172,<br />

e19514<br />

Goettel, Rainer 6566<br />

Goetz, Marlies 6567<br />

Goetz, Matthew P. 534,<br />

3001, 10509, e13086<br />

Goez, David 2078<br />

G<strong>of</strong>f, Barbara Ann 5021,<br />

5054, 5082^<br />

G<strong>of</strong>f, Laura Williams 2595,<br />

4117, 4545, 8519,<br />

e13606, e14503<br />

G<strong>of</strong>fard, Nicolas 10553<br />

G<strong>of</strong>fin, John R. e18079<br />

G<strong>of</strong>fredo, Veronica e21113,<br />

e21134<br />

Gogas, Helen 592, 8505<br />

Gogia, Ajay 6593, e11013<br />

Gogoi, Radhika e15536<br />

Goh, Alvin e15060, e15171<br />

Goh, Boon C. 1064, 2551,<br />

3002, 4100^, 5544,<br />

e13002<br />

Goh, Chee Leng 1545<br />

Goh, Jeffrey C. TPS5113<br />

Goh, Wee Lee e18528<br />

Goiffon, Reece J. 503<br />

Gok Durnali, Ayse 638<br />

Gokmen, Erhan e19024<br />

Gokmen-Polar, Yesim 505,<br />

7106, 10628<br />

Goksu, Sema Sezgin e14070<br />

Golan, Talia 3027, e14143,<br />

e14646<br />

Golant, Mitch 9048<br />

Gold, Anne TPS4682<br />

Gold, David V. 3091, 4043<br />

Gold, Heather Taffet 6628<br />

Gold, Kathryn A. 7047<br />

Gold, Michael e15582<br />

Goldberg, Jeffrey M. 1512<br />

Goldberg, John M. 9565<br />

Goldberg, Judith D. 5016,<br />

e11010, e11064<br />

Goldberg, Richard M. 3502,<br />

3514, 3517, 3532, 3617<br />

Goldberg, Sarah B. 7524,<br />

7525<br />

Goldberg, Stacie 2510<br />

Goldberg, Stuart 6509^,<br />

6520, 6525, TPS6636,<br />

e19641<br />

Goldberg, Zelanna 7530<br />

Goldemberg, Alberto e14657<br />

Goldenberg, Alec e14581<br />

Goldenberg, David M. 3091,<br />

4043, e13005, e18513<br />

Goldfarb, Paul M. TPS10632<br />

Goldfarb, Shari Beth 575,<br />

9104, 10514<br />

Goldgar, David 1538<br />

Goldhirsch, Aron 504, 546,<br />

1049, 1115, 9022<br />

Goldim, Jose Roberto 1522,<br />

10593<br />

Goldin, Jonathan G. 4566<br />

Golding, Sophie 5522, 7544<br />

Goldinger, Simone M. e19017<br />

Goldkorn, Amir 4511, 4563,<br />

4663, 10503<br />

Goldmacher, Gregory V.<br />

e13082<br />

Goldman, Bryan H. 4019,<br />

e18536<br />

Goldman, Cecilia e19552<br />

Goldman, John M. 10550<br />

Goldman, Jonathan Wade<br />

3032, 3034, 3042^,<br />

e13000<br />

Goldman, Stanton 9501<br />

Goldman, Stewart 9532<br />

Goldschmidt, Jerome H. 3069<br />

Goldschmidt, Pablo e13105<br />

Goldsmith, Stanley J. e13592<br />

Goldsmith, Tessa A. e16059<br />

Goldstein, Bram e15500<br />

Goldstein, Daniel A. 10514<br />

Goldstein, David P. 9032<br />

Goldstein, David P 10522<br />

Goldstein, David TPS3643,<br />

TPS4137, 6111, 9044,<br />

9087, e16507<br />

Goldstein, Lori J. 1021, 1092<br />

Goldstein, Matthew 2514<br />

Goldstein, Scott 3621<br />

Goldstein, Steven C. e18540<br />

Goldstein, Theodore C. 503<br />

Goldsweig, Howard TPS8604,<br />

e14546<br />

Goldwasser, Francois 1095,<br />

2557, 4583, 5067,<br />

e13056<br />

Goldwasser, Meredith 3007<br />

Golfieri, Rita e14654<br />

Golfinos, John e19015<br />

Gollerkeri, Ashwin 3032<br />

Gollins, Simon e14587<br />

Gollob, Jared 3062<br />

Gollub, Marc 3563, 3583<br />

Golomar, Wilson e13105<br />

Goloubeva, Olga G. 10572,<br />

e19565<br />

Gombos, Andrea 3018^<br />

Gomella, Leonard G. e15012,<br />

e15120<br />

Gomes, Monica e15532<br />

Gomez de Liaño, Alfonso<br />

e15159<br />

Gomez Dorronsoro, Maria<br />

Luisa e14057<br />

Gomez Dorronsoro, Marisa<br />

e14120<br />

gomez Lopez, Cesar 9073<br />

Gomez Ordonez, Silvia<br />

e15033<br />

Gomez Raposo, Cesar 1570,<br />

e11028<br />

Gomez, Alexandra e12039<br />

Gomez, Ana 10564<br />

Gomez, Auxiliadora 3571<br />

Gomez, Daniel Richard 6124,<br />

7062<br />

Gomez, David e17503<br />

Gómez, Francisco 3072<br />

728s Numerals refer to abstract number


Gomez, Henry Leonidas 531,<br />

1024, 2097, e11087,<br />

e11518<br />

Gomez, Henry e11066,<br />

e15036<br />

Gomez, Javier 10013<br />

Gomez, Jorge E. e17564<br />

Gomez, Lourdes 4089<br />

Gomez, Nerea 8572<br />

Gomez, Patricia 619<br />

Gomez, Rosa Maria Sanchez<br />

10512, 10595<br />

Gomez, Scarlett Lin 6050<br />

Gomez-Abuin, Gonzalo<br />

e15570<br />

Gómez-Codina, Jose 7011,<br />

7095, 8062<br />

Gomez-Martin, Carlos 3068<br />

Gómez-Pardo, Patricia 509,<br />

566<br />

Gomez-Pinillos, Alejandro<br />

e15191<br />

Gomez-Roca, Carlos Alberto<br />

2539<br />

Gomez-Roca, Carlos 5522<br />

Gomez-Tato, Antonio 10534<br />

Gon�aves de Freitas<br />

Seriaco, Fabiola de Lourdes<br />

e19528, e19584<br />

Goncalves, Anthony 529,<br />

e19539<br />

Goncalves, Margarida Brito<br />

e13119<br />

Goncalves, Marina Sahade<br />

e20502<br />

Goncalves, Priscila Hermont<br />

e14519<br />

Gondek, Lukasz 6521<br />

Gonen, Mithat 3583, 3620,<br />

10015<br />

Gong, Cynthia e15154<br />

Gong, Gyungyub e11504,<br />

e11508<br />

Gong, Jifang 3040, e14604<br />

Gong, Jing TPS3117<br />

Gong, Ke-Wei 8592<br />

Gong, Yun 515, 594<br />

Gongora, Celine 10505<br />

Gonsalves, Wilson I. 8092<br />

Gonschior, Ann-Katrin<br />

e13095^<br />

Gonzague, Laurence<br />

TPS1613, e19539<br />

Gonzales, Alicia 1592<br />

Gonzalez de Castro, David<br />

LBA4000, TPS10633^,<br />

e14088<br />

Gonzalez del Alba, Aranzazu<br />

TPS4672, TPS4674,<br />

e15041, e15144<br />

Gonzalez Gordaliza, Cristina<br />

e12033<br />

Gonzalez Griego, Martha 2515<br />

Gonzalez Guerrero, Juan<br />

Francisco e15525<br />

Gonzalez Larriba, Jose Luis<br />

10564, e18055<br />

Gonzalez Vela, Jose Luis<br />

e15525<br />

Gonzalez, Adriana 6038<br />

González, Alan e14104<br />

Gonzalez, Alberto 2023<br />

Gonzalez, Arancha TPS4685<br />

Gonzalez, Carmen E. 6125<br />

Gonzalez, Encarnacion<br />

10608, e11535, e21088<br />

Gonzalez, Federico 1111<br />

Gonzalez, Gisela 2527, 2531<br />

Numerals refer to abstract number<br />

Gonzalez, Joseph 1006,<br />

TPS1143<br />

Gonzalez, Manuel Sureda<br />

3064, e16000<br />

Gonzalez, Maria Magdalena<br />

e16561<br />

Gonzalez, Melanie L. e14124,<br />

e14125, e14727, e18516,<br />

e19606, e19638<br />

Gonzalez, Michael 3004<br />

Gonzalez, Ramon 3064<br />

Gonzalez, Rene 3102, 8510,<br />

8567<br />

Gonzalez, Ricardo Jorge<br />

10048, 10070<br />

Gonzalez, Sonia 1001<br />

Gonzalez, Xavier e11081<br />

Gonzalez-Angulo, Ana M. 537,<br />

1015, 1029, 1032, 1054,<br />

1104, 9072<br />

Gonzalez-Billalabeitia,<br />

Enrique 4587, 7095<br />

Gonzalez-Carrero, Joaquin<br />

4089<br />

Gonzalez-Cortijo, Lucia 1111<br />

Gonzalez-Huarriz, Marisol<br />

e14057<br />

Gonzalez-Larriba, Jose-Luis<br />

7011, e12012<br />

González-Martín, Antonio<br />

5017^, 5031, 5068,<br />

e15575<br />

Gonzalez-Santiago, Santiago<br />

1011<br />

Gonzalo, Nuria 3096<br />

Gonzalvez, Maria Luisa<br />

e11545, e15144<br />

Goodall, Shannon e15180<br />

Goode, Ellen L. 5077<br />

Goodhand, James e19581<br />

Goodheart, Michael J e15580<br />

Goodman, Aaron 7097<br />

Goodman, Andrew TPS8605<br />

Goodman, Annekathryn 5029,<br />

e15545, e15568<br />

Goodman, Karyn A. 3583,<br />

4045, 4061, e14555<br />

Goodman, Michael 5570,<br />

e14512, e15187<br />

Goodman, Oscar B. 4558,<br />

e15210<br />

Goodman, Simon L. 2055<br />

Goodman, Vicki L.<br />

LBA8500^, 8501, 8518<br />

Goodwin, Neal TPS3110<br />

Goodwin, Pamela Jean 1019,<br />

9111<br />

Goodwin, Rachel Anne 1036<br />

Goodyer, Ian 9589<br />

Gooley, Theodore 5082^<br />

Gooskens, Saskia 8535<br />

Gopal, Ajay K. 8027<br />

Gopal, Arun e15140<br />

Gopalakrishna, Prashanth<br />

2552, e18063^<br />

Gopaluni, Srivalli e11089,<br />

e18535<br />

Gopinath, Kodaganur<br />

Srinivasachar 1093,<br />

e11511, e12030, e14721<br />

Goranova-Marinova, Vesselina<br />

8018^<br />

Gorbach, Sherwood 9067<br />

Gorbunova, Vera A. 3532,<br />

7512, e15543, e19012<br />

Gorbunova, Vera LBA7500,<br />

e14723<br />

Gordon, Leo I. 8003, 8080,<br />

e18530<br />

Gordon, Lynn K 1052<br />

Gordon, Michael Alexander<br />

4010<br />

Gordon, Michael S. e13077<br />

Gordon, Michael S. 3023,<br />

3032, 3034, 3042^, 4513,<br />

7514, 8531, e13000<br />

Gordon, Nicolaus e19040<br />

Gordon, Ryan Robert 4669,<br />

e15207<br />

Gordon, Stephen e13007<br />

Gordon, Stuart R. e14514<br />

Gore, Lia 3017, TPS9594,<br />

e13600^<br />

Gore, Martin Eric 5012,<br />

5017^, 5022<br />

Gore, Steven e16519,<br />

e16537<br />

Gorey, Tom 1113<br />

Gorgun, Omer 9567<br />

Gori Savellini, Gianni e13098<br />

Gori, Stefania e19592<br />

Gorin, Sherri Sheinfeld 3567<br />

Gorlia, Thierry 2<br />

Gorlick, Richard Greg 9503<br />

Gorman, Greg e15515,<br />

e19006<br />

Gornet, Terrie 10613<br />

Gornick, Michele Caroline<br />

1515<br />

Gorritz-Kindu, Magdaliz<br />

e14028, e14030<br />

Goryca, Krzyszt<strong>of</strong> 10561<br />

Gos, Aleksandra 8548,<br />

e19022<br />

Gosiengfiao, Yasmin C. 9534,<br />

e20001<br />

Gospodarowicz, Mary K. 4509<br />

Goss, Glenwood D. 7093,<br />

7511, TPS7613^, 10620,<br />

e13541, e18079<br />

Goss, Paul Edward 500, 501,<br />

524, 561, 596, 604,<br />

TPS670, e11005<br />

Goswami, Chirayu Pankaj<br />

505, 7106, 10628<br />

Goswami, Trishna 8065<br />

Gotay, Carolyn 6002, 7607<br />

Goteke, Vamshi Krishna<br />

Reddy e18545<br />

Gotlib, Jason R. 6624<br />

Goto, Ayumu e14047,<br />

e14050, e14123<br />

Goto, Hidemi e14596<br />

Goto, Koichi 1552, 7044,<br />

e17513, e17526, e17554,<br />

e18054, e18064<br />

Goto, Tomoko 5037, e15579<br />

Gotoh, Masahiro 3088^<br />

Gotoh, Mitsukazu e13015<br />

Gottardis, Marco M. e15151<br />

Gotte, Martin e21041<br />

Gottfried, Maya 4564, 4619,<br />

e15058, e18108<br />

Gottlieb, Linda TPS1614<br />

Gotto, Ge<strong>of</strong>frey 4552<br />

Gou, Hongfeng 4638<br />

Goubar, Aicha 543, 1091,<br />

1601<br />

Gough, Karla 9124, e19605<br />

Gouin, Francois 10056<br />

Goulart, Bernardo H.L. 6007<br />

Gould, Jeff 9103<br />

Gouldesbrough, David e16042<br />

Gounant, Valérie TPS7112,<br />

e18075<br />

Gounder, Mrinal M. 3016,<br />

3062, 10004, 10019<br />

Gourdin, Theodore Stewart<br />

6611<br />

Gourevitch, David e13507<br />

Gourgou-Bourgade, Sophie<br />

LBA4003<br />

Gourin, Christine Gail e16061<br />

Gourley, Charlie 3048, 5022<br />

Gousia, Anna 10575<br />

Gouy, Sebastien 5083,<br />

10098<br />

Govil, Sushama 1540<br />

Govindan, Ramaswamy LBA4,<br />

2610, 3046, 7006, 7010,<br />

7002<br />

Govindan, Serengulam V.<br />

3091<br />

Govindarajan, Mallarajapatna<br />

e11027, e14675, e14721,<br />

e19619<br />

Govindarajan, Rangaswamy<br />

6099<br />

Govindasami, Koveela 1545<br />

Gowdar, Indraneel e12004<br />

Gowdra Halappa, Vivek 4114<br />

Goy, Andre e18507, e19641<br />

Goyal, Abhishek 4101<br />

Goyal, Gautam 9559<br />

Goyal, Ravi K. 6017, 6029,<br />

e15184<br />

Goyal, Shikha 2072<br />

Goydos, James S TPS3112<br />

Graaf, Winette T.A. Van der<br />

10067<br />

Grabellus, Florian 10032^,<br />

10046<br />

Grabsch, Heike 4124,<br />

TPS4143<br />

Grabska, Joanna 6555,<br />

e18543<br />

Grabulovski, Dragan 2575<br />

Gracia, Jose Manuel e18044<br />

Gracien, Edith 7558<br />

Gradishar, William John 596,<br />

604, 610, TPS1134, 2532,<br />

6623<br />

Gradishar, William TPS661<br />

Grados Sanchez, Oswaldo<br />

Abelardo e12041<br />

Graeber, Stefan 1523<br />

Graesslin, Olivier 1584<br />

Graf von der Schulenburg,<br />

J.-Mathias e18016^<br />

Graff, Jeremy Richard 8082<br />

Graff, John 6023<br />

Graham, Donna M. e16506<br />

Graham, Mark e21022<br />

Graham, Martin 3029<br />

Graham, Richard A. e13106<br />

Graham, Roger e11548<br />

Graham, Tracy 1555<br />

Gralla, Richard J. e19635<br />

Gralow, Julie TPS3109,<br />

e16524<br />

Gramantieri, Laura 3061<br />

Gramatzki, Martin 6543,<br />

8004, e18565<br />

Grambach, Jana 1084<br />

Graña, Begoña 1009<br />

Granados-Garcia, Martin 1607<br />

Granata, Roberta 5555<br />

Grande Pulido, Enrique 4119,<br />

10546<br />

Grande Ventura, Carlos<br />

e18040<br />

Grande, Enrique e14671<br />

Grande, Roberta e14082<br />

Grange, Florent 1566, 8601<br />

Granger, Catherine e19605<br />

Granito, Alessandro TPS4151<br />

729s


Granowetter, Linda 9537<br />

Grant, Barbara W. 2006,<br />

6507<br />

Grant, Cliona 2528, 8051<br />

Grant, Florence 3620<br />

Grant, Janice 6069<br />

Grant-Huerta, Yanina 1581<br />

grant-Kels, Jane e21135<br />

Grasela, Thaddeus 9547<br />

Graser, Anno 3603, e14043<br />

Grasic Kuhar, Cvetka 558<br />

Grasso, Luigi 3016<br />

Gratama, Jan W 10504<br />

Graudenz, Marcia S 1522<br />

Grauer, Matthias e15577<br />

Gravalos Castro, Cristina 3571<br />

Gravenor, Donald 7100<br />

Gravina, Giovanni Luca<br />

e15200<br />

Gravis, Gwenaelle LBA4567^,<br />

4583, 4606, 4625,<br />

e15155, e19539<br />

Gray, Angel e13115, e13117<br />

Gray, Heidi J. 5021,<br />

TPS5113<br />

Gray, Jhanelle Elaine 3027,<br />

7065, e17559, e21155<br />

Gray, Kathryn P. 4543, 4555,<br />

4635<br />

Gray, Robert James 1005,<br />

1021, 1027, 9020<br />

Gray, Sarah E 9064<br />

Graziano, Francesco 3518,<br />

3540, 4026, 4084,<br />

e14135<br />

Graziano, Joseph e13066,<br />

e13067<br />

Graziano, Paolo e18021<br />

Graziano, Stephen L. 7007<br />

Graziano, Steve e16047<br />

Grebennik, Dmitri O.<br />

TPS4675^<br />

Greco, Frank A. 3097, 4130,<br />

7035, 7100, 7567, 7587,<br />

10530, 10584, e13076,<br />

e21023<br />

Greco, Luigi e19002<br />

Green, Andrew R. 1013,<br />

1014, 1098<br />

Green, Dan e11098, e15569,<br />

e19594<br />

Green, Daniel M. 9526<br />

Green, Jennifer A TPS2103<br />

Green, John LBA5000<br />

Green, Margaret 6582<br />

Green, Marjorie C. 527<br />

Green, Michael J e13026<br />

Green, Rebecca L. 6093<br />

Green, Sarah G. e14118<br />

Green, Stephen 3004<br />

Greenan, Caroline 10553<br />

Greenberg, James e18507<br />

Greenberg, Jonathan 7536,<br />

e14617<br />

Greenberg, Mark 6030<br />

Greenberg, Peter 6522<br />

Greenberg, Philip D 2523<br />

Greenberg, Richard Evan<br />

4526<br />

Greenberg, Richard H.<br />

TPS3635^<br />

Greene, Jessica 6602<br />

Greene, John e19059<br />

Greene, Joshua B. e16027<br />

Greene, Mark H. 1519<br />

Greenhalf, William e14625<br />

Greeno, Edward e14610<br />

Greenup, Rachel Adams 1122<br />

Greer, Joseph A. 6004, 6048,<br />

6062, 6106, 9030, 9101<br />

Gregorc, Vanesa 7076, 7085,<br />

7544, 7581, TPS7619,<br />

e18096<br />

Gregori, Josep 10511<br />

Gregory, Stephanie A. 6552,<br />

8080, e16503, e18502,<br />

e18510<br />

Gregory, Walter 502, 513,<br />

8015, e13083<br />

Greig, Paul David 4109,<br />

e14592<br />

Greil, Richard 514, TPS1148,<br />

CRA3503, TPS3641^,<br />

4074, 10501<br />

Greillier, Laurent e16560<br />

Greimel, Elfriede 7607<br />

Greimel, Eva 6002, 6090<br />

Greiner, Jochen 6567<br />

Greisinger, Anthony 8543<br />

Grell, Peter e11072<br />

Gremillet, Eric e15109,<br />

e21056<br />

Gresham, Gillian e14154<br />

Greshock, Joel e13596<br />

Gressin, Remy 2023, 8026<br />

Greten, Tim F 4023, 4032<br />

Greten, Tim 3103, 10566<br />

Greulich, Heidi 4580<br />

Grever, Michael R. 6508<br />

Grewal, Jai TPS2107<br />

Grewal, Ravinder K. 5509^<br />

Grey, Andrew e15054<br />

Greystoke, Alastair P 3004<br />

Greystoke, Alastair 3030<br />

Grgic, Mislav e15031<br />

Gribbin, Thomas Edward<br />

e16561<br />

Gribk<strong>of</strong>f, Greta TPS7611<br />

Gridelli, Cesare LBA7507,<br />

7550, TPS7612<br />

Griebel, Lis-Femke 5007<br />

Grier, Charles E. 3627,<br />

e13047<br />

Grier, Holcombe E. 9503,<br />

9537<br />

Griesinger, Frank TPS7612<br />

Griffin, Melissa e13596<br />

Griffin, Michael TPS4143<br />

Griffin, Nicole 10630<br />

Griffin, Thomas W. LBA4518<br />

Griffith, Kent A. 8011, 10028<br />

Griffiths, Elizabeth A. 6565,<br />

6602<br />

Griffiths, Michael TPS10633^<br />

Griggs, Jennifer J. 6024,<br />

6056<br />

Grigiene, Ruta TPS661<br />

Grignani, Giovanni 10066,<br />

e15073, e15192, e20512<br />

Grigorieva, Julia e18096,<br />

e18100<br />

Grigsby, Perry Wayne e15514<br />

Grigull, Joerg 4633<br />

Grijuela, Barbara 5096<br />

Grill, Jacques TPS9596<br />

Grilley-Olson, Juneko E. 3046<br />

Grillo, Federica e19038<br />

Grillo, Tarin 4021<br />

Grimaldi, Antonio M 8573,<br />

e19034<br />

Grimaldi, Franco 1604,<br />

e19038<br />

Grimison, Peter S. 4531,<br />

TPS4681<br />

Grimison, Peter 9126<br />

Grimm, Christoph 5111<br />

Grimm, Sean Aaron 2006,<br />

2035<br />

Grimminger, Peter Philipp<br />

e12007<br />

Grinblatt, David L. 6122<br />

Griner, Paula L. 1018<br />

Grinsted, Lynda 3004, 7503<br />

Grisham, Rachel Nicole 5030,<br />

5043<br />

Grisold, Wolfgang 2<br />

Grissom, Daniel e14177<br />

Grist, William 5560, 5570<br />

Grivas, Petros 4506, 4573<br />

Grivaux, Michel 1574<br />

Grives, Audrey 10562<br />

Grob, Jean Jacques 8518,<br />

8568, 8578, 8591<br />

Grob, Jean Jacques<br />

LBA8500^, 8555, 8559<br />

Grober, Yuval 2078<br />

Groen, Harry J.M. 7543<br />

Groeschel, Andreas 2573^<br />

Grogan, Eric L. 7008<br />

Grohe, Christian TPS7109<br />

Grollier, Chrystelle e14148<br />

Groman, Adrienne E. 3582,<br />

e14690, e19041<br />

Grommes, Christian 2008<br />

Gronbaek, Henning 4015<br />

Gronchi, Alessandro 10000,<br />

10016, 10017, 10022,<br />

10026, 10065, 10079,<br />

10096<br />

Gros, Alena e14179<br />

Grosch, Kai 3099<br />

Grosh, William W. 8530<br />

Groshen, Susan G. 2538,<br />

4563, 4575, 8073,<br />

e13531, e15153<br />

Gross, Anne e16515, e16559<br />

Gross, Cary P. TPS669,<br />

1030, 1595, 4651, 6019,<br />

6628, 9034, 9109, 9123,<br />

e15150, e18133<br />

Gross, David 4014<br />

Gross, Gerald G. 1083<br />

Gross, Neil D. e16045<br />

Grossbard, Michael L. 6114,<br />

e18139<br />

Grosser, Susanne 4034<br />

Grosshans, David 9585<br />

Grossi, Francesco 7081,<br />

7085, 7550, 7558, 7577,<br />

7581, TPS7612, e21065<br />

Grossman, H. Barton 4523<br />

Grossman, Mary e19626<br />

Grossman, Stuart A. 2017,<br />

e17552<br />

Grosso, Federica 7082, 7084,<br />

10053, e19549<br />

Grosso, Marco 8529<br />

Grothey, Axel 1557, 3502,<br />

TPS3634<br />

Grotz, Travis Edward 8600<br />

Grotzinger, Kelly LBA8500^,<br />

e12001<br />

Grove, Laurie E. 8027, 8070<br />

Grover, Surbhi 7098,<br />

e12009, e12508, e17534,<br />

e18032<br />

Groves, Morris D. 2003,<br />

2029^, 2036, 2064<br />

Growcott, Jim W e15141<br />

Growdon, Whitfield Board<br />

5029, e15545, e15568<br />

Grubb, Robert L. 4594<br />

Grubbs, Elizabeth Gardner<br />

4641<br />

Grubbs, Stephen S. e18122<br />

Gruber, Harry E. 2101<br />

Gruber, Michael L. TPS2107<br />

Gruber, Stephan 10570<br />

Gruber, Stephen B. 1515,<br />

1578, e12026<br />

Grude, Francoise e14148<br />

Gruenberger, Birgit 4074,<br />

e14095, e14099<br />

Gruenberger, Thomas 3508,<br />

3613, e14095, e14099<br />

Grueneisen, Andreas 6610<br />

Gruenhage, Frank 1523<br />

Gruenwald, Frank e14565<br />

Gruenwald, Viktor 4631,<br />

e19502<br />

Grunberg, Steven M. 9061<br />

Grundy, Anna e17519<br />

Grupp, Stephan A. 9506<br />

Gruselle, Olivier 7056,<br />

TPS8111, e13061<br />

Gruttadauria, Salvatore 4110<br />

Grybowicz, L TPS1144<br />

Grycz, Krystyna 4660<br />

Grünwald, Viktor 4503, 4636,<br />

e15190<br />

Grzegorzewski, Kris e14525,<br />

e14627<br />

Grzesiczek, Anja 3595<br />

Gröpper, Stefanie 6527<br />

Grønberg, Bjørn Henning<br />

7027, 9040<br />

Gschwend, Juergen 4590<br />

Gschwend, Jürgen E. e15112,<br />

e15157, e15195<br />

Gu, Aiqin e18088<br />

Gu, Chu Shu e14001<br />

Gu, Chu-Shu 6049<br />

GU, Kangsheng 8554<br />

Gu, Xiaoling e18082<br />

Guan, Bin e14538<br />

Guan, Changjiang e14716<br />

Guan, Jilin e18090<br />

Guan, Ran e19030<br />

Guan, Wen xian e14509,<br />

e21034<br />

Guan, Zhongzhen 3614<br />

Guancial, Elizabeth Ann<br />

4577, 4582<br />

Guaraldi, Monica 5513<br />

Guardado, Sandra e15033<br />

Guardino, Ellie LBA1, 528,<br />

1037<br />

Guarducci, Cristina 1012<br />

Guarino, Lorena e11052<br />

Guarino, Michael J. 4043,<br />

e18122<br />

Guarneri, Valentina 614, 631<br />

Guastafierro, Anna 8577<br />

Guastalla, Jean Paul<br />

e13095^, e13097^<br />

Guba, Susan C. LBA4<br />

Gucalp, Ayca 1006<br />

Guchelaar, Henk-jan 526,<br />

10077, 10518<br />

Guckert, Mary E. LBA8500^,<br />

8501<br />

Guderian, Gernot e15044<br />

Gudikote, Jayanthi 10612<br />

Guelfucci, Bruno e16044,<br />

e16051, e16055<br />

Guenther, Georg 6566<br />

Gueorguieva, Ivelina 2042<br />

Guerci-Bresler, Agnes 6505^,<br />

6522, 6523<br />

Guerif, Stephane e15199^<br />

Guerin, Annie 1037, 6594,<br />

10094, e11076, e16571,<br />

e20511<br />

730s Numerals refer to abstract number


Guerin-Meyer, Veronique<br />

e14148<br />

Guerra Alia, Eva e15575<br />

Guerra, Eva 5068<br />

Guerrero, Carmen T e12033<br />

Guerrero, David e14057<br />

Guerrero, Javier 3072<br />

Guerrero, Lisa 9534<br />

Guerrieri-Gonzaga, Aliana 519<br />

Guerry, DuPont e19054<br />

Guglielmi, Alfredo 4110<br />

Guglin, Maya 2612<br />

Guha, Chandan e16058<br />

Guha, Udayan 7103<br />

Guhlke, Stefan e14565<br />

Gui, Jiang e14514<br />

Guidelli, Giacomo e13098<br />

Guidoboni, Massimo 8513<br />

Guiducci, Graziano 2057<br />

Guigay, Joel 5505<br />

Guijarro, Ricardo 7058,<br />

7060, 10594<br />

Guiles, Fran 3621<br />

Guilhot, Francois 6506<br />

Guillem, Jose G. 3563, 3583<br />

Guillemaut, Severine 10006<br />

Guillemet, Cecile 10035,<br />

10056<br />

Guillemin, Françis 1584<br />

Guillemot, Didier e14065<br />

Guillen, Carmen 1531<br />

Guillen-Ponce, Carmen<br />

e12033, e19590<br />

Guillen-Rodriguez, Jose<br />

e18534<br />

Guillot, Bernard 8591<br />

Guillot, Eugenie e11551<br />

Guimaraes, Alexander R<br />

2012, e14615<br />

Guimbaud, Rosine 3547,<br />

4091<br />

Guinan, Emer M. 1511<br />

Guinan, Patrick e13513,<br />

e13532, e15119<br />

Guindalini, Rodrigo Santa<br />

Cruz 2038<br />

Guinebretiere, Jean-Marc<br />

10554<br />

Guitart, Joan e18523<br />

Guiu, Michel e14129<br />

Guivarch, Laurent e14148<br />

Guix, Marta 4582, TPS4688<br />

Gujja, Swetha 6099<br />

Gul, Naheed 10025<br />

Gulati, Roman 4557<br />

Gulaya, Sachin 6121<br />

Gulbas, Zafer e21050<br />

Gulec, Seza A 4043<br />

Gulenchyn, Karen Y. 6049<br />

Guller, Ulrich e14544<br />

Gulley, James L. 2526,<br />

TPS2617, 3043, TPS3638,<br />

4569, TPS4701, 10589<br />

Gullo, Giuseppe 615, 623,<br />

e13520<br />

Guminski, Alexander David<br />

e19042<br />

Gumus, Mahmut e14618<br />

Gumusay, Ozge 638<br />

Gunaldi, Meral e13523<br />

Gunchenko, Alexandra 3027<br />

Gundacker, Holly 4010, 6502<br />

Gundeti, Sadashivudu 6554,<br />

e18544, e18545<br />

Gunduz, Seyda e14070<br />

Gunn, Gary Brandon 5561,<br />

5589<br />

Gunn, Shelly e13025<br />

Gunnarsson, Orvar e15139<br />

Numerals refer to abstract number<br />

Gunson, Diane E 9562<br />

Gunter, Marc J e14005<br />

Gunturi, Anasuya 8569<br />

Gunturu, Krishna Soujanya<br />

e14534<br />

Gunzer, Katharina TPS662<br />

Guo, C.H. e11561<br />

Guo, Chengye e18033<br />

Guo, Fuchun e18051<br />

Guo, Huiqin e18087<br />

Guo, Jun LBA4537, 4628,<br />

8506, 8554, 8561, 8562,<br />

e15051, e15052<br />

Guo, Min Amy 6583, 6600<br />

Guo, Rong e19588<br />

Guo, Shuliang 7091<br />

Guo, Wei 5593, e14645<br />

Guo, Xiang e13021<br />

Guo, Xiaotao e13064<br />

Guo, Yantong e13526<br />

Guo, Ye 8552, e16039<br />

Guo, Ying 3055<br />

Guo, Yongli e13567<br />

Guo, Zhanfang 534, 3047<br />

Gupta, Ashok Kumar<br />

CRA2509, 2510, 2521,<br />

TPS2613, TPS2622, 4505,<br />

7509, 8507<br />

Gupta, Avinash 3051<br />

Gupta, Charu 4666<br />

Gupta, Digant 5049, e14012,<br />

e14697<br />

Gupta, Gopal Nand e15014,<br />

e16544<br />

Gupta, Ira 6584<br />

Gupta, Monica e19619<br />

Gupta, Neeraj 3077, 8017,<br />

8033, 8034, 8064,<br />

e13603<br />

Gupta, Nilesh 1560, e15174<br />

Gupta, Pankaj e18513<br />

Gupta, Rajeev e19516<br />

Gupta, Rajesh 3083<br />

Gupta, Rajnish K. e16551<br />

Gupta, Ritu 6593, 9549,<br />

9551, 9580<br />

Gupta, Rohan 10071<br />

Gupta, Shachi e15123<br />

Gupta, Shivani e16012<br />

Gupta, Shweta 1611, 8085,<br />

e14590<br />

Gupta, Sudeep 1032, 3011,<br />

e11552, e13068<br />

Gupta, Tejpal e16021<br />

Gupta, Vikas 6624, TPS6640<br />

Gupta, Vishal e13500,<br />

e13562, e21028, e21029<br />

Gur, Eyal 1564<br />

Gurbuz, Yesim e14618<br />

Guren, Marianne Gronlie<br />

4015<br />

Guren, Tormod Kyrre 3531,<br />

3548<br />

Gurevich, Pavel 2567<br />

Gurion, Ronit 8069<br />

Gurkan, Emel 6550<br />

Gurney, Howard 550, 4531<br />

Gurney, James G. 9526,<br />

9531<br />

Guron, Gunwant K. e12025<br />

Gurpide, Alfonso 7095, 8572,<br />

e15041, e18008<br />

Gurski, Carol Ann 4566<br />

Guru, Khurshid e15204<br />

Gury, Jean-Pierre 1574<br />

Gusella, Milena e13019<br />

Guset, Valentin 6123<br />

Gustafson, Eric e13561<br />

Gustafson, Michael 2546<br />

Gustafsson, Goran 9590<br />

Gustavson, Mark 517<br />

Gusyatin, Oleg e21146<br />

Guth, Amber Azniv e11010<br />

Guth, Dagmar e19540<br />

Guth<strong>of</strong>f, Rainer 2054<br />

Guthrie, Graeme JK e14054<br />

Guthrie, Katherine 6631<br />

Gutierrez Restrepo, Eduardo<br />

e18044<br />

Gutierrez, Antonio 10079<br />

Gutierrez, Benjamin e15165<br />

Gutierrez, Martin 2610, 8073<br />

Gutierrez, Maya TPS662<br />

Gutierrez-Barrera, Angelica M.<br />

1546, 1549, e11090<br />

Gutierrez-Gutierrez, Gerardo<br />

9073<br />

gutierrez-Hernandez, Olga<br />

6599<br />

Gutin, Alexander 7023<br />

Gutin, Philip H. 8583<br />

Gutkind, Jorge Silvio 5541<br />

Gutkind, Silvio e16061<br />

Gutzmer, Ralf LBA8500^,<br />

8505, 8559, e19031<br />

Guy, Michelle 1545<br />

Guyetant, Serge 4129<br />

Guyon, Frederic e15535<br />

Guzzo, Federica 5093, 5094,<br />

e15547, e15550, e15551,<br />

e15553<br />

Gwede, Clement 6100<br />

Gyan, Emmanuel 2023,<br />

2539, 8026<br />

Gyimesi, Edit e21140<br />

Gynnild, Mari Aas 9040<br />

Günther, Andreas e18565,<br />

e18572^<br />

Gyorffy, Balazs 10571,<br />

e21005<br />

Gyorffy, Steve e13011<br />

Gyorki, David E. 8583<br />

Gyotoku, Hiroshi 7569<br />

Gyure, Maria 1563<br />

H<br />

Ha, Caroline Ann 9065<br />

Ha, Jeslin 8078, e18515<br />

Ha, Tam Cam 1593, e18546<br />

Haab, Francois e15105<br />

Haag, Georg Martin 4083<br />

Haagsma, Ben e15517<br />

Haaland, Benjamin 5580,<br />

e15045<br />

Haalck, Thomas 8505<br />

Haanen, John B. A. G. 4611,<br />

8502^<br />

Haas, Jonathan A. e15504,<br />

e21114<br />

Haas, Lindsey R 5004<br />

Haas, Michael 4023, 4072<br />

Haas, Naomi B. 4500, 4513,<br />

e13604<br />

Haasbeek, Cornelis J 7009<br />

Haasler, George e17555<br />

Haass, Nikolas 8553<br />

Habbous, Steven 1597, 9032<br />

Haber, Daniel A. 4566<br />

Haberal, Nihan e15572,<br />

e15573<br />

Haberl, Christopher 3588<br />

Habermann, Elizabeth Butzer<br />

6016<br />

Habermann, Thomas Matthew<br />

9057<br />

Habertheuer, Karlheinz<br />

e14087<br />

Habets, Leo 10600<br />

Habib, Nabil e20500<br />

Habib, Sharifa 9076<br />

Habib, Stefanie e18066<br />

Habibian, Muriel LBA4567^<br />

Habr, Dany 2543<br />

Habra, Mouhammed Amir<br />

4639, 4641<br />

Habuchi, Tomonori 10520<br />

Hack, Stephen Paul TPS3640<br />

Hacker-Prietz, Amy 4045<br />

Hacking, Nigel TPS4150<br />

Hackl, Monika 2053<br />

Hackl, Wolfgang 508<br />

Hackshaw, Allan 7538, 7562<br />

Haddad, Abdo S. e18046<br />

Haddad, Fabio Jose e21073<br />

Haddad, Housam 4609,<br />

TPS4684<br />

Haddad, Mojgan 603, 608,<br />

614<br />

Haddad, Riad 1578<br />

Haddad, Robert I. 5501,<br />

5579, 5582, TPS5598,<br />

9024, e16060<br />

Haddad, Vincent 4042,<br />

TPS4135<br />

Haddad, Zaid 4565<br />

Hadeiba, Husein e21071<br />

Hadenfeldt, Rebecca 9110<br />

Hadengue, Alexandra 3506<br />

Hadji, Peyman e11040^<br />

Hadler, Dietrich 7536<br />

Hadoux, Julien 10098<br />

Hadziahmetovic, Mersiha<br />

9544<br />

Haefner, Mark Dieter e19648<br />

Haenel, Mathias 8025, 8031<br />

Haeusler, Christa e14103<br />

Haferlach, Claudia 6510,<br />

6610<br />

Haffty, Bruce George e13543<br />

Haga, Shunsuke 1112<br />

Hage, Georges e20500<br />

Hagemann, Andrea R. 5013<br />

Hagemeister, Erin e16515<br />

Hagemeister, Fredrick B.<br />

8067<br />

Hagen, Steinar 6619^<br />

Hagenbeck, Carsten<br />

TPS1146, 1602<br />

Hager, Gudrun 5058<br />

Hager, Henrik e21110<br />

Haggman, Michael 4550<br />

Hagiwara, Eri e19583<br />

Hagiwara, Koichi 7563,<br />

e18129<br />

Hagiwara, May e16573<br />

Hagiwara, Sachiko 10604,<br />

e18004, e18014, e18017<br />

Hagleitner, Melanie M. 10077<br />

Hah, Jeong-Hun 5552<br />

Hahn, Astrid e15157<br />

Hahn, Elizabeth A 6092<br />

Hahn, Lars 1077<br />

Hahn, Markus 1067, 1090,<br />

e21003<br />

Hahn, Noah M. 4524, 4586,<br />

4603, TPS4676, e15179,<br />

e15213, e21016<br />

Hahn, Seung Shin e16047<br />

Hahn, William C 2020, 4585,<br />

5011<br />

Haidau, Augustin e18534<br />

Haie Meder, Christine 10098<br />

Haie-Meder, Christine 5083<br />

Haigentz, Missak e16058<br />

Haim, Nissim 9052<br />

Hainaut, Pierre 1522, 7007,<br />

10593, e13546<br />

731s


Hainsworth, John D. 618,<br />

1018, 1086, 4014, 4130,<br />

7035, 7100, 7567, 7587,<br />

8556, 8567, 10530,<br />

10584, e21023<br />

Hainsworth, John D 8579,<br />

e19048^<br />

Haioun, Corinne 8021, 8057<br />

Haislip, Sally 569, 571,<br />

e11002, e14106, e14141,<br />

e14146, e15183, e18097<br />

Haitao, Ma e18013<br />

Haiying, Nan 1094<br />

Hajage, David 9538<br />

Hajdenberg, Julio e15010,<br />

e15102<br />

Hajduch, Marian 1087,<br />

e14556<br />

Hajek, Roman 8095<br />

Hakamada, Kenichi e14709<br />

Hakansson, Ulf 4508<br />

Hakim, Khaldoun 1574<br />

Hakimi, Abraham A. 4604<br />

Halabi, Susan 4515, 4592,<br />

4655, 4667<br />

Haldeman, Jennifer e19047<br />

Hale, Christopher e19003<br />

Hale, Diane F 625, 2508,<br />

2529<br />

Hale, Juliet 9539<br />

Hale, Katherine Stemke<br />

1015, 4131<br />

Halene, Stephanie 6628<br />

Haley, Sherri 2517<br />

Halfdanarson, Thorvardur<br />

Ragnar 6034, e14594,<br />

e14600, e14620, e15580<br />

Halford, Sarah E. R. 9542<br />

Hall, Brett 2583<br />

Hall, Carolyn S. TPS10631,<br />

e21107<br />

Hall, Chad A 8082<br />

Hall, Ge<strong>of</strong>f e13083<br />

Hall, Junior 9506<br />

Hall, MacLean S 2511<br />

Hall, Marcia LBA5000<br />

Hall, Michael J. 1544, 1550,<br />

e14021<br />

Hall, Peter TPS665<br />

Hall, Philip S 4610<br />

Hall, Robert 7029<br />

Hall, Simon 4623<br />

Hall, Stacy e19528, e19584<br />

Hall, Suzanne P 4557,<br />

e15206<br />

Hallberg, Kenth e13518<br />

Hallemeier, Christopher Leigh<br />

4595<br />

Haller, Daniel G. 3522,<br />

3523, 3525, 3578<br />

Haller, Nairmeen Awad<br />

e17004<br />

Halling, Kevin C 1083<br />

Hallmeyer, Sigrun 8567<br />

Hallock, Abhirami 2081<br />

Hallquist, Allan e11562,<br />

e13003<br />

Halluard, Céline 2605<br />

Halm, Melissa e15089<br />

Halmos, Balazs 3024,<br />

e17563<br />

Halpenny, Barbara 9008<br />

Halperin, Marc e15172<br />

Halpern, Allan C. e19052<br />

Halpern, Anna 6562<br />

Halpern, Marisa e14067<br />

Halpern, Michael T. 6046<br />

Halpern, Wendy 2567<br />

Hals, Petter-Arnt 6619^<br />

Halton, Elizabeth 6613<br />

Haluska, Frank G. 6503,<br />

8504, 10010<br />

Haluska, Paul 534, TPS2618,<br />

3001, 3027, 5012, 5073,<br />

5077<br />

Halvorsen, Tarje Onsoien<br />

7027<br />

Halyard, Michele Y. 6108,<br />

e16572<br />

Ham Guo, Maung Shwe 9041<br />

Ham, Hye Seon e14136<br />

Hamada, Akinobu e13072<br />

Hamada, Chikuma 3607<br />

Hamadani, Mehdi 8065<br />

Hamaguchi, Masanari e18056<br />

Hamaguchi, Tetsuya 3524,<br />

e14126<br />

Hamakawa, Hiroyuki 5584<br />

Hamaker, Marije 1080<br />

Hamalidou, Eleni Kyrillos<br />

e14567<br />

Hamamoto, Yasuo 4002,<br />

4031, e14123, e14510<br />

Hamatake, Motoharu 3045<br />

Hamberg, Paul 2593, 10504<br />

Hamdan, Shadi E. e15123<br />

Hameed, Meera 10001,<br />

10015<br />

Hamid, Omid 2510, 3528,<br />

8502^, 8508, 8510, 8518,<br />

8567, e13101, e19048^<br />

Hamidou, Zeinab 1584<br />

Hamilton, Ann S. 6023, 9130<br />

Hamilton, Audrey M. 8054<br />

Hamilton, Bronwyn E. 2077<br />

Hamilton, Mark e21044<br />

Hamilton, Michael 2577<br />

Hamilton, Olivia 6021<br />

Hamilton, Robert James<br />

e15022<br />

Hamilton, Ronald H. 8528<br />

Hamilton, Stephanie A. 3627,<br />

e13047<br />

Hamlin, Paul A. 8054<br />

Hammel, Pascal LBA4007,<br />

4032, 4053, 4133,<br />

e14660<br />

Hammer, Liat 9052<br />

Hammerer, Peter e15195<br />

Hammerman, Peter S. 7006<br />

Hammers, Hans J. TPS2614,<br />

4549, 4564, 4619,<br />

TPS4694, e15058<br />

Hammers, Hans TPS4687<br />

Hammond, Sue CRA9513,<br />

9528<br />

Hamoir, Marc 5519<br />

Hampshire, Margaret K. 6135<br />

Hampson, Grace 8056<br />

Hamza, Mohamed Ali 2030,<br />

2064<br />

Han, Baohui 1551, e12010,<br />

e12013, e13528, e17551,<br />

e18013, e18038, e18088,<br />

e21001<br />

Han, Chen 1066<br />

Han, Chun 4073<br />

Han, Dale 10048<br />

Han, Dolly 6084, 9111<br />

Han, Gang 2544, 10048<br />

Han, Guohong e14605<br />

Han, Heather TPS2620<br />

Han, Hye-Suk e19538<br />

Han, Hyo S. 1585, 2534<br />

Han, Jae Ho e18519<br />

Han, Ji-Youn 7089, 7533,<br />

7596, e18119<br />

Han, Jung Woo 3606<br />

Han, Lei 500, 561<br />

Han, Leona C. 4657<br />

Han, Mark e21080<br />

Han, Misop e15214<br />

Han, Myung Woul 5586<br />

Han, Qinghong 1063<br />

Han, Sae-Won 1003, 3076,<br />

3624, e14136<br />

Han, Tae H. e13079<br />

Han, Wei e14716<br />

Han, Wonshik 1056, 1133,<br />

10621, e11532, e21160<br />

Hanafi, Widayana 10051,<br />

e20509<br />

Hanagiri, Takeshi 10585<br />

Hanai, Nobuhiro e16032<br />

Hanash, Alan M. 6539<br />

Hanby, Andrew TPS10633^<br />

Hancock, John F. e13516<br />

Hancock, Michael L. e19582,<br />

e19585<br />

Hande, Kenneth R. 10003<br />

Handgretinger, Rupert 9573<br />

Handisides, Damian 6585<br />

Handorf, Elizabeth e18032<br />

Handy, Beverly Carol 10613<br />

Haney, Patricia LBA8500^<br />

Haney, Sophie 2552<br />

Hanfstein, Benjamin 6510<br />

Hangauer, David G. 4654<br />

Hanin, Francois-Xavier 5519<br />

Hank, Jacquelyn A e19047<br />

Hanker, Lars Christian 5044,<br />

e15520<br />

Hanks, Brent Allen 10563<br />

Hanmod, Santosh S. 9578<br />

Hanna, Ehab Y. 5561, 5589<br />

Hanna, Glenn 5582<br />

Hanna, Michelle 6130<br />

Hanna, Nader 1039, e14035,<br />

e14056, e14522, e14626,<br />

e14638<br />

Hanna, Nasser H. 4534,<br />

7516<br />

Hanna, Wedad 1123<br />

Hanna, William e21047<br />

Hannah, Alison L. TPS4695<br />

Hanneken, Jan M 7608<br />

Hanniford, Douglas 8550,<br />

8565<br />

Hannig, Carla 4065<br />

Hanning, Fritha J. e15054<br />

Hannon, Breffni e16567<br />

Hanrahan, Aphrothiti 4527<br />

Hans, Stephane 5554,<br />

e13578<br />

Hansen, Lisa 9018<br />

Hansen, Morten 2565<br />

Hansen, Nora M. TPS1134<br />

Hansen, Olfred 7001<br />

Hansen, Torben 3513, 3573<br />

Hanson, Jennifer 3515<br />

Hanson, Sven e13503<br />

Hansra, Damien Mikael<br />

e18109<br />

Hansson, Johan 8517, 8588,<br />

10521<br />

Hao, Desiree 3013, e18110<br />

Hao, Si Jie e14651<br />

Hao, W-H e14699<br />

Haq, Rashida 3515, 9111<br />

Haqq, Christopher M. 4521,<br />

4558<br />

Haque, S<strong>of</strong>ia 5514, 5545^<br />

Har-Noy, Michael e13013<br />

Hara, Hiroki 4031<br />

Hara, Kazuo e14540<br />

Harada, Masao 7017, 7028,<br />

7070, 7086, 7561<br />

Harada, Naoki e13502<br />

Harada, Toshiyuki 7563,<br />

7565, 7571, e18135,<br />

e19556<br />

Haraf, Daniel J. 5500<br />

Haralambakis, Nick 5574<br />

Haraldsdottir, Sigurdis<br />

e14178<br />

Harari, Paul M. e16010<br />

Harasymczuk, Malgorzata<br />

8528<br />

Harb, Wael A. 7556, 8032,<br />

e13040, e18558<br />

Harbeck, Nadia 552, 556,<br />

TPS649, 1072, 1081,<br />

1092, 1114, e11040^,<br />

e13605<br />

Harbison, Christopher 3615<br />

Harbom, Kirsten e18147<br />

Hardacre, Jeffrey M 4049<br />

Hardenbergh, Patricia H.<br />

6094<br />

Hardesty, Rosemarie A. 2590,<br />

3095<br />

Hardikar, S. 4107<br />

Harding, Gale e18556<br />

Harding, James J. 8515,<br />

8575<br />

Hardingham, Jenny 3534<br />

Hardj, Marjie e13120^<br />

Hardt, Molly 9034, 9109,<br />

9123, e18133<br />

Hardwicke, Fred L. e21105<br />

Hardy, Janet 2604<br />

Hardy, Richard 4121<br />

Hardy-Bessard, Anne-Claire<br />

TPS4692^, 5018<br />

Hare, Elethea e13064,<br />

e13067<br />

Hare, Ryan B e19041<br />

Haregewoin, Abebe 3627,<br />

e13047<br />

Hargrave, Darren R 9519,<br />

9542, TPS9596<br />

Hari, Parameswaran 8033<br />

Harich, Hanns-Detlev 640<br />

Hariprasad, Gururao e15510<br />

Hariprasad, Roopa e15510<br />

Harita, Shingo 7042<br />

Harkin, D. Paul 10553<br />

Harlacker, Kathleen e13602,<br />

e13604<br />

Harland, Luke 9032<br />

Harland, Stephen John 4529,<br />

4558<br />

Harle, Alexandre TPS10634<br />

Harman, Dennis 6088<br />

Harmenberg, Johan 7539<br />

Harmenberg, Ulrika 4561<br />

Harmon, David C. 10058<br />

Harms, JAN-Dirk 2048<br />

Harness, Jay K. 6086, 9047<br />

Harnicar, Stephen 6613<br />

Haro, Akira e13090<br />

Harold, Michael e16516<br />

Harold, Nancy 2571<br />

Haroun, Iman 1555<br />

Harpaz, Zivit e14731<br />

Harpole, David 7008<br />

Harr, Jodi L. e16561<br />

Harrell, Frank E e14501<br />

Harrelson, Robin 6524^<br />

Harrington, Elizabeth 3004<br />

Harrington, Kevin J. 2530<br />

Harris, Adrian L. 1066<br />

Harris, Anne 9521, 9522<br />

Harris, Brianna 8043<br />

Harris, Dean Laurence<br />

e15054<br />

732s Numerals refer to abstract number


Harris, Ella 1113<br />

Harris, Gordon J 6136<br />

Harris, Jonathan 5530, 5583<br />

Harris, Lyndsay 1045, 10508,<br />

10558<br />

Harris, Pamela Jo 2606,<br />

5544<br />

Harris, Wayne A C e21045<br />

Harris, Wayne 2576, e15006<br />

Harrison, Claire N 6514^,<br />

6625^, 6626^, TPS6639,<br />

TPS6642^<br />

Harrison, Evan 5055<br />

Harrison, Lauren 6537, 9501<br />

Harrison, Louis Benjamin<br />

5531, 5559, 5581, 6114,<br />

e15138<br />

Harrison, Marilynn e19008<br />

Harrison, Mark LBA4001<br />

Harrison, Michael Roger<br />

4654, e15061<br />

Harrison, Timothy 10553<br />

Harrow, Kimberly 3041<br />

Harshman, Lauren Christine<br />

4536, 4538, 4610, 10513,<br />

e15005<br />

Hart, Eric M 3083<br />

Hart, Lowell L. 512,<br />

TPS8115<br />

Harter, Philipp 5007, 5031,<br />

5051, e15520<br />

Hartford, Alan 4568<br />

Hartig, Gregory K e16010<br />

Hartkopf, Andreas D. 1067,<br />

1090, 5042, e21003<br />

Hartley, John Anthony 4123<br />

Hartley, Lynne 5073<br />

Hartmann, Arndt 4590<br />

Hartmann, Holger e14138<br />

Hartmann, Joerg Thomas<br />

4090, e15026<br />

Hartmann, Lynn C. 5076<br />

Hartmann, Michael 4530<br />

Hartner, Lee 9117<br />

Hartshorn, Kevan L. 9004<br />

Harun, Nusrat 7078<br />

Harunal Rashid, Mohamad<br />

Farid Rin 10051, e20509<br />

Harustiak, Tomas e14631<br />

Harvey, Christopher 2502<br />

Harvey, Jimmie Huling 6624<br />

Harvey, R Donald 2576,<br />

3049, 3094, e13026,<br />

e18569<br />

Harvey, R. Donald 3055,<br />

e18553<br />

Harvey, Vernon J. 9022<br />

Harwin, William N. 1086,<br />

7567, e18560, e19615<br />

Harza, Mihai 4501<br />

Harzstark, Andrea Lynne<br />

4513, 4660, e15137<br />

Hasan, Baktiar 7081, 8076<br />

Hasan, Farah 2589<br />

Hasan, Yasmin 1101, 1116,<br />

e11512<br />

Hasanali, Zainul 6616,<br />

e18500<br />

Hasani, Ekta 6005<br />

Hasanovic, Adnan 7505,<br />

7578<br />

Hase, Kazuo 3625<br />

Hasegawa, Dave e13500,<br />

e21029<br />

Hasegawa, Hirotoshi 10531,<br />

e14091<br />

Hasegawa, Junichi 3607<br />

Hasegawa, Kiyoshi 4104<br />

Hasegawa, Koki 10519<br />

Numerals refer to abstract number<br />

Hasegawa, Seiki 7031, 7080,<br />

10585<br />

Hasegawa, Takeo e13015<br />

Hasegawa, Yasuhisa 5542,<br />

e16032<br />

Hasegawa, Yoshie TPS1141<br />

Hasegawa, Yoshihiro e15098<br />

Hasegawa, Yoshikazu 7000,<br />

e18128<br />

Hasegawa, YUkihiro 7086<br />

Hasenbach, Kathy 2055<br />

Hasenburg, Annette 516, 517<br />

Hasenkamp, Justin 6543,<br />

8004<br />

Hasford, Joerg 6510<br />

Hashemi Sadraei, Nooshin<br />

2014<br />

Hashemi-Sadraei, Neda 2076,<br />

2093, 2096, 2100<br />

Hashiguchi, Yojiro 3625<br />

Hashimoto, Kenji e19535<br />

Hashimoto, Kenkichi e14722<br />

Hashimoto, Kiyoshi e21053<br />

Hashimoto, Kohei 7038<br />

Hashimoto, Masaki 7080,<br />

10585<br />

Hashimoto, Yutaka e13072<br />

Hashine, Katsuyoshi e15127<br />

Hasija, Nalini 2099<br />

Haslinger, Michelle e14172<br />

Hasrouni, Edy 6521<br />

Hass, Holger 3519^, 3588<br />

Hass, Tomas e14621<br />

Hassan Hamed, Ali e15561<br />

Hassan, Andrew Bassim<br />

10006<br />

Hassan, Manal 4116<br />

Hassan, Muhammad Radzi<br />

Abu 4106<br />

Hassan, Raffit 7030<br />

Hassel, Jessica C. LBA8509,<br />

e19031<br />

Hassell, Robert 569, 571,<br />

e11002<br />

Hassett, Edel e14111<br />

Hastings, B Taylor 6103<br />

Hata, Akito 7528, 10610,<br />

e18025, e18134<br />

Hata, Hiroyuki 8097<br />

Hata, Taishi TPS3642<br />

Hata, Yoshinobu e18127<br />

Hatabu, Hiroto 7553<br />

Hatake, Kiyohiko 10541,<br />

e19512<br />

Hatanaka, Kazuteru 3593,<br />

e14062<br />

Hatano, Koji e15065<br />

Hatch, Stephanie e13587<br />

Hatcher, Helen 3100, 9589<br />

Hatfield, James 9578<br />

Hatlevoll, Ingunn e14531<br />

Hatori, Masahito 9574<br />

Hatori, Takashi 4035<br />

Hatse, Sigrid 1020<br />

Hatsuse, Kazuo 3625<br />

Hattermann, Valerie 1067<br />

Hatton, Matthew 6113, 7562<br />

Hattori, Michiyo e18036,<br />

e18042<br />

Hattori, Minoru e14091<br />

Hattori, Yoshihiro e18060,<br />

e18134<br />

Hatzidaki, Dora 7564<br />

Hatzimichael, Eleftheria<br />

e18567<br />

Hatzimouratidis, Constantinos<br />

e15040<br />

Hatzis, Christos 515<br />

Hauber, A. Brett 4608<br />

Hauck, Adina 4602<br />

Hauge, Petra Weber e14637<br />

Hauge, Truls e14637<br />

Haugnes, Hege Sagstuen<br />

4508<br />

Hauke, Ralph J. 4603,<br />

TPS4676, TPS4697, 8556<br />

Hauns, Bernhard 4115<br />

Haupt, Eric C. 4517<br />

Hauptmann, Steffen 5005<br />

Haura, Eric B. 2544, 2559,<br />

7065, 7589, e17559,<br />

e21155<br />

Hauschild, Axel LBA8500^,<br />

8501, 8502^, 8505, 8511,<br />

8517, 8579, 8585<br />

Hauser, Stefan 4636<br />

Hausman, Diana Felice 3024<br />

Hauss, Pierre-Alexandre 1574<br />

Haustermans, Karin 4053,<br />

TPS4141<br />

Hautaniemi, Sampsa 8074<br />

Hautefeuille, Agnes e13546<br />

Hautmann, Matthias 5512<br />

Havel, Libor 7575<br />

Havelange, Violaine TPS6637<br />

Havlik, Roman e14556<br />

Havsteen, Hanne e13108^<br />

Hawaldar, Rohini W 1108,<br />

e11552<br />

Haward, Robert 6091<br />

Hawes, Debra 4575, e15005,<br />

e15153<br />

Hawk, Natalyn Nicole 2576,<br />

e14614<br />

Hawker, Gillian e19522<br />

Hawkes, Claire 1014<br />

Hawkes, Eliza Anne 3587<br />

Hawkins, Douglas S. 1576,<br />

9509, 9535<br />

Hawkins, Elma S. 2087,<br />

TPS2107<br />

Hawkins, Robert E. CRA4502<br />

Hawks, Geri TPS4680<br />

Hawks, Laura 10514<br />

Hawley, Sarah T. 6023, 6024<br />

Haworth, Annette TPS4690<br />

Hawryluk, Elena B. 8552<br />

Hawthorne, Ben e17529<br />

Hay, John TPS5600<br />

Hayakawa, Kazushige e15533<br />

Hayashi, Hidetoshi e18060<br />

Hayashi, Katsuhiro 10072,<br />

10075, e20505<br />

Hayashi, Kazuhiko e21108<br />

Hayashi, Michio 10610<br />

Hayashi, Mina e18081<br />

Hayashi, Mitsuhiro TPS1141<br />

Hayashi, Naoki 590, 1012<br />

Hayashi, Ryuichi 5542<br />

Hayashi, Tatsuro e17517<br />

Hayashi, Yuji 613<br />

Hayashi, Yuki e14547<br />

Hayashida, Tetsu 10531,<br />

e11060<br />

Hayat, Henry 4619, e15058<br />

Hayden, James B. 10011^<br />

Haydon, Andrew Mark 3504<br />

Haydu, Lauren 8558, e19011<br />

Hayeems, Robin Zoe e19616<br />

Hayenga, Jon e17529<br />

Hayes, Daniel F. 525, 9020<br />

Hayes, David N. 5503, 5515,<br />

5533, 5539, 5577, 7006<br />

Hayes, Malcolm 549^<br />

Hayes, Monica Prasad<br />

e15513<br />

Hayes, Teresa Gray 4643<br />

Hayes-Jordan, Andrea Anita<br />

9509, 10021<br />

Hayes-Lattin, Brandon M.<br />

9136, e19503<br />

Haylock, Brian e14587<br />

Hayman, Suzanne R. 8013,<br />

8043, 8092, 8096, 8105,<br />

TPS8116<br />

Haymart, Megan Rist 6056<br />

Hayn, Birgit 10526<br />

Hayn, Matthew H. e15046,<br />

e15203<br />

Haynes, Hilda e16058<br />

Haynes, Kevin 1503<br />

Hayran, Mutlu 9050<br />

Hays, John L. 2545<br />

Hays, Rita 9110<br />

Hayter, Charles 4509<br />

Hayward, Larry 511,<br />

TPS1144<br />

Hayward, Michele C 5577<br />

Haz-Conde, Mar e14595,<br />

e21002<br />

Hazarika, Diganta e11043<br />

Hazlett, Allison e18550<br />

Hazra, Saswati 10535<br />

He, Aiwu Ruth 2590, 3095,<br />

TPS3638, e14009,<br />

e14569, e14658<br />

He, Bing e14523<br />

HE, Chengcheng 5558<br />

He, Chunyu e14645<br />

He, Hang e14651<br />

He, Housheng Hansen 576<br />

He, Kuanglin e15565<br />

He, Miaoling e15178<br />

He, Ren 6105<br />

He, Shui TPS6643^<br />

He, Suqin 3090<br />

He, Xi C e14153<br />

He, Xin 1508<br />

He, Yulei 1605<br />

He, Zijing 9082<br />

Headly, Anna e14175<br />

Heale, Esti e12034<br />

Healey, Diane I. 6506<br />

Healey, John H. 9537<br />

Healy, Laura 1511, 1559<br />

Healy, Patrick N. 2602, 4591<br />

Heaney, Mark L. 6609, 6613<br />

Heath, Elisabeth I. 1560,<br />

4654, 4656<br />

Heaton, Simon 3022<br />

Hebbar, Mohamed 3506,<br />

3547, LBA4007<br />

Hebden, Tony e17009<br />

Hebert, Christophe 5521<br />

Heck, Matthias e15195<br />

Heckel, Martha L. TPS10635<br />

Heckler, Charles E. 9028<br />

Heckler, Charles E 9010,<br />

9014, 9027, 9113, 9141,<br />

9142, e19510<br />

Hedley, David W 4109,<br />

e14524, e14535, e14592<br />

Hedley, Mary Lynne 9077<br />

Hedlund, Åsa 7539<br />

Hedrick, Eric 6507, 6508,<br />

6515^<br />

Hedrick, Nancy A. 2040<br />

Hedvat, Cyrus 10524<br />

Hee, Kim-Hor e13002<br />

Heeger, Steffen 3591<br />

Heemers, Hannelore e15204<br />

Heenan, Sue 4640<br />

Heerema, Nyla A. 6507,<br />

6508, 9548<br />

Heeren, Timothy C 6097<br />

Heeroma, Dr Karin 1534<br />

733s


Heerschap, Arend e13111<br />

Heersink, Mila J e11505<br />

Heery, Christopher Ryan<br />

2526, TPS2617, TPS3638,<br />

TPS4701<br />

Heese, Oliver 2034<br />

Heffner, Leonard T. 8100,<br />

8101<br />

Heflin, Jon e17506<br />

Hegarty, Sarah e15163<br />

Hegde , Aparna 4037<br />

Hegde, Priti e13000, e13113<br />

Hegde, Upendra P. e21135<br />

Hege, Kristen 2562, 3006^<br />

Hegewisch-Becker, Susanna<br />

TPS3641^, 4023, 4065<br />

Hegi, Monika 2001<br />

Heguy, Adriana TPS4144,<br />

4527, 7052<br />

Hehlmann, Rudiger 6510<br />

Hei, Tom K e21097<br />

Hei, Yong Jiang 7549<br />

Heiba, Sherif 5578<br />

Heidecke, Cordula 7542<br />

Heidenreich, Axel 4530,<br />

TPS4698^, e15112,<br />

e15122, e15195<br />

Heider, Ulrike e18552<br />

Heigener, David Felix e18100<br />

Heijmen, Linda e13111<br />

Heike, Michael TPS3641^,<br />

8022<br />

Heil, Gerhard 3<br />

Heilbrun, Lance K. 1560,<br />

5534, e15081<br />

Heilbrun, Marta Elise e13548<br />

Heimberger, Amy B. 2011<br />

Heimburger, Douglas C. 9558<br />

Heinecke, Achim 6610<br />

Heinemann, Volker 3519^,<br />

3541, 3588, 3603,<br />

TPS3639, TPS3641^,<br />

4023, 4041, 4050, 4072,<br />

e11528, e14043<br />

Heinrich, Bernhard 1092,<br />

e11528<br />

Heinrich, Daniel LBA4512,<br />

4551<br />

Heinrich, Georg 1602,<br />

10540, e15520<br />

Heinrich, Michael C. 10591<br />

Heirman, Carlo 2507<br />

Heiskanen, Raita e13035<br />

Heist, Rebecca Suk TPS3118,<br />

7010, 7099, 7513<br />

Heitjan, Daniel 7077<br />

Hejduk, Karel e18062<br />

Hejna, Matthew e13115<br />

Hejna, Michael 4074<br />

Hekmat, Khosro 1533,<br />

10526, CRA10529<br />

Helal, Rafaela Cordeiro<br />

e14101<br />

Helbich, Thomas 10570<br />

Helbling, Daniel 3595<br />

Held, Gerhard 8022, 8024,<br />

8025<br />

Held, Swantje 4072<br />

Held-Warmkessel, Jeanne<br />

e19580<br />

Helenowski, Irene B. 1561<br />

Helfrich, Barbara 7534<br />

Helft, Paul R. 6095, 6128<br />

Helgadottir, Hildur 10521<br />

Helias-Rodzewicz, Z<strong>of</strong>ia 8540<br />

Hellborg, Henrik 4561<br />

Heller, Glenn 4517<br />

Heller, Michael J e21047<br />

Hellerstedt, Beth A. 3069,<br />

TPS3111, 7514<br />

Hellman, Andrzej 6584<br />

Hellriegel, Edward 9547,<br />

e18522<br />

Hellstrom, Anita e18551<br />

Hellwig, Birte 10551<br />

Helman, Lee J. 9511<br />

Helms, Hans-Joachim e14167<br />

Helvaci, Kaan e11540<br />

Hembrough, Todd A. e21068<br />

Hemminki, Akseli e13035<br />

Henary, Haby Adel 8551<br />

Hendershott, Karen 1127<br />

Henderson, Ben e11541<br />

Henderson, David Andrew<br />

3012, 3046<br />

Henderson, Les 4022<br />

Henderson, Tara O. CRA9513,<br />

9586<br />

Hendifar, Andrew Eugene<br />

3015<br />

Hendifar, Andrew 10036<br />

Hendlisz, Alain 3559<br />

Hendrick, Franklin e16519,<br />

e16537<br />

Hendrickx, Tine 4622<br />

Hendriks, Bart TPS663<br />

Hendriks, T 3576<br />

Hendrikx, Jeroen 2550<br />

Hendrix, Laura H 4647,<br />

4658, 6037<br />

Heng, Daniel Yick Chin 4536,<br />

4538, 6082, e14073,<br />

e21083<br />

Hengstler, Jan G 10551<br />

Henin, Emilie e15182<br />

Henke, Corinna C. 9033<br />

Henke, Michael 5502,<br />

e16033<br />

Henner, William David<br />

10576, 10584<br />

Hennessy, Sean e19531<br />

Henning, Jan-Willem 6548<br />

Henriksson, Anders e19532<br />

Henriquez Cooper, Hoover<br />

Rodyl e15556<br />

Henriquez, Alexandra 1111<br />

Henry, David H. 1577, 4030<br />

Henry, Elizabeth e19588<br />

Henry, Norah Lynn 525,<br />

e16524<br />

Henry, Stefanie 5505<br />

Henschen, Stephan 552<br />

Hense, Joerg 2086<br />

Hensing, Thomas A. e18117<br />

Hensley, Kenneth e14098<br />

Hensley, Martee Leigh 5090,<br />

TPS10103<br />

Hentic, Olivia 4133, e14660<br />

Hentrich, Marcus 4602, 8046<br />

Hentsch, Bernd 4115<br />

Heo, Dae Seog 5552, 7566<br />

Heo, Jeong 4103, e14566<br />

Heon, Stephanie 7525, 7547,<br />

7553, 10592<br />

Hepburne-Scott, Henry 8566<br />

Hepp, Philip Gm 1072,<br />

e21020<br />

Heras, Javier e11028<br />

Heras, Lucia e18030<br />

Herbert, Robert J. 6026<br />

Herbst, Roy S. 5529, 5532<br />

Hercbergs, Aleck A e19573<br />

Herder, Judith 6058<br />

Heriot, Alexander Graham<br />

3629<br />

Herkommer, Kathleen e15157<br />

Herman, Christine e21060,<br />

e21111<br />

Herman, Dominique 1574<br />

Herman, Gary A. 10573<br />

Herman, James Gordon<br />

10625, e18147<br />

Herman, Joseph M. 3596,<br />

4045, 4055, e14585<br />

Hermann, Jacques TPS7111<br />

Hermann, Robert C. LBA4,<br />

e13113<br />

Hermans, Chris A.M. e19560<br />

Hermine, Olivier LBA4007,<br />

10007, 10089,<br />

TPS10102^<br />

Hernan, Carranza e12039<br />

Hernandez, Claudia 6580<br />

Hernandez, Elvin e16523<br />

Hernandez, Irene e14057,<br />

e14120, e21086<br />

Hernandez, Liza e13088,<br />

e15010<br />

Hernandez, Rohini Khorana<br />

1579, 1581, e12023<br />

Hernandez, Susana 10564<br />

Hernandez-Cortes, Gines<br />

1111<br />

Hernandez-Ilizaliturri,<br />

Francisco J. e18537,<br />

e18538, e18539<br />

Hernandez-Losa, Javier 566,<br />

3014, 7041<br />

Hernando, Blanca 10512,<br />

10595, e11074<br />

Hernando, Cristina 7058,<br />

10594<br />

Hernando, Eva 8550, 8565,<br />

e19015, e19018<br />

Hernando-Trancho, Florentino<br />

7011, 10564<br />

Herndon II, James E 2074^<br />

Herndon, Betty e21085<br />

Herndon, James Emmett<br />

2027, 2090^, 2094^,<br />

2095^<br />

Herold, Stefanie 10536,<br />

10537<br />

Herr, Harry W. 4581^,<br />

e15022<br />

Herrel, Lindsey Allison<br />

e15187<br />

Herremans, Catherine e13605<br />

Herrera Gomez, Angel 1607<br />

Herrera, Alex Francisco<br />

e18530<br />

Herrera, Javier e21086<br />

Herrera, Jorge Eduardo<br />

e11565<br />

Herrera, Luis A e12027<br />

Herrera, MaryTere e15527<br />

Herrera, Victoria L.M. e13574<br />

Herrero, Ana 1570<br />

Herrero, Joaquin 8062<br />

Herrero, Mercedes 602,<br />

e11517<br />

Herrin, Jeph 1030, 4651<br />

Herrmann, Edwin e15044<br />

Herrmann, Ken 10012<br />

Herrmann, Richard e19648<br />

Herschbach, Peter e15157<br />

Herschberger, Amber B<br />

TPS3114<br />

Hersey, Peter 8503^,<br />

LBA8509, 8553<br />

Hersh, Evan 8534<br />

Hershman, Dawn L. TPS669,<br />

CRA6009, 6078, 6118,<br />

9018, 10516<br />

Herskovic, Arnold M. e18502<br />

Hertenstein, Bernd 6543<br />

Hertz, Daniel Louis 10515<br />

Hertz-Eichenrode, Martin<br />

Michael 1077<br />

Hertzog, Antoine e19555<br />

Herve, Christian e19640<br />

Herwig, Martina 2054<br />

Herzog, Thomas J. 5087,<br />

6078, e15578<br />

Heskamp, Sandra 3035<br />

Hesketh, Paul Joseph 7570<br />

Heslop, Helen E. 2558<br />

Hess, Dagmar e13605<br />

Hess, Georg 4128, 6543,<br />

6584<br />

Hess, Gregory P. e16512<br />

Hess, Kenneth R. 527, 612,<br />

2003, 2029^, 2036,<br />

10613, 10627, e13020<br />

Hesse, Sylvie 8555<br />

Hessel, Colin 5508<br />

Hetherington, John 4509<br />

Heublein, Sabine e15529<br />

Heuck, Christoph J. e18548<br />

Heuck, Christoph Johann<br />

8041<br />

Heudel, Pierre 10562,<br />

e14730<br />

Heukamp, Lukas Carl 1533,<br />

7603, 10526, CRA10529<br />

Heussen, Nicole 10552<br />

Hevessy, Zsuzsanna e21140<br />

Hew, Karina E 5055<br />

Hexner, Elizabeth O. 6579,<br />

6624<br />

Heyburn, John 7030<br />

Heymach, John 3598, 7096,<br />

7567, 10612, e15075,<br />

e15085, e15087<br />

Hibler, Lauren Marie 1571<br />

Hickish, Tamas TPS651,<br />

TPS9148<br />

Hickman, Theresa TPS4152,<br />

e14566<br />

Hickson, Ian e15166<br />

Hida, Naoya e18099<br />

Hida, Toyoaki 7003, 7028,<br />

e17510, e18145<br />

Hidaka, Noriaki 10073<br />

Hidalgo, Manuel 602, 2567,<br />

2568, 3066, 3068, 4040,<br />

e11006, e11517, e13048,<br />

e15111<br />

Hidalgo, Rosa 4101<br />

Hiddemann, Wolfgang 6610<br />

Hieken, Tina J. e19061<br />

Higaki, Kenji 588<br />

Higano, Celestia S. 4, 4511,<br />

4513, 4557, 4571, 4663,<br />

10503, e15201, e15206<br />

Higgins, Brian Patrick<br />

e19570<br />

Higgins, Doreen 5082^<br />

Higgins, Helen B TPS660,<br />

TPS665, TPS1144, 5046<br />

Higgins, Kristin 7059<br />

Higgins, Linda C TPS4678<br />

High, Hilda Anne 5022<br />

Highsmith, W Edward 10588<br />

Higuchi, Mitsunori e13015<br />

Hijazi, Hussam 10042<br />

Hijazi, Youssef 2504<br />

Hijikata, Yasuki e13014<br />

Hijioka, Susumu e14540<br />

Hijiya, Nobuko TPS9594<br />

Hijri, Fatima Zahra e11502,<br />

e11516, e14725, e14726<br />

Hilden, Joanne M. 9548<br />

734s Numerals refer to abstract number


Hilf, Norbert 3530<br />

Hilfenhaus, Georg 4128<br />

Hilfrich, Joern 1023<br />

Hilger, James D. 8098<br />

Hilger, Ralf A. 2086, 10032^<br />

Hill, Andrew e15152<br />

Hill, Brian Thomas 8055<br />

Hill, D Ashley 9521, 9522<br />

Hill, Dawn 3025<br />

Hill, Jamie 3062<br />

Hill, Jerrold W. e16512<br />

Hill, Kimberley 1542<br />

Hill, Mark 3578<br />

Hill, Richard P e14535<br />

Hill-Kayser, Christine E. 6135<br />

Hille, Elysee T.M. 526<br />

Hillemanns, Peter 5007<br />

Hiller, Louise TPS660,<br />

TPS665, TPS1144<br />

Hillmen, Peter 6584<br />

Hilpert, Felix LBA5002^,<br />

5005, 5007, 5031<br />

Hils, Rita 5051<br />

Hilton, Christie 6620<br />

Hilton, Nancy e16515<br />

Hiltz, Andrea Clare 4509<br />

Himmetoglu Ussakli, Cigdem<br />

e15207<br />

Himsl, Isabelle Katrin 1114<br />

Hindenburg, Hans-Joachim<br />

5044, e11040^, e19540<br />

Hinder, Vicky e15054<br />

Hindi, Nadia 3618, e13085<br />

Hines, Oscar J e14582<br />

Hinke, Axel 640, 3<br />

Hino, Masayuki TPS8118<br />

Hinoi, Takao e14091<br />

Hinotsu, Shiro e15004,<br />

e15106<br />

Hiotis, Spiros P. e14583,<br />

e14656<br />

Hirai, Fumihiko 3045<br />

Hirai, Takashi 3607<br />

Hirakawa, Akihiro e19535<br />

Hirakawa, Hitoshi e16032<br />

Hirakawa, Masakazu e13014<br />

Hirama, Hiromi e15127<br />

Hirashima, Tomonori 7521,<br />

e18036, e18042, e18141<br />

Hirashita, Teijiro e14722<br />

Hirata, Akira 10531<br />

Hirata, Kazuto e17538<br />

Hirata, Yoshito e15201<br />

Hirchaud, Edouard 8048<br />

Hiret, Sandrine e21117<br />

Hirmand, Mohammad 4519^<br />

Hiro, Junichiro e14039,<br />

e14541, e21053<br />

Hironaka, Shuichi 4002<br />

Hirose, Kenzo 3596, 3616<br />

Hirose, Shigemichi e11060<br />

Hirose, Takashi e13029<br />

Hiroshige, Shoji e14722<br />

Hirota, Makoto 5556<br />

Hirsch, Bradford Richard<br />

6041, 6047, 6051<br />

Hirsch, Fred R. 7516, 7517,<br />

7552, 7597, e18077<br />

Hirsch, Jennifer e15103<br />

Hirsch, Michelle S. 4543,<br />

4582, 4635, 5011<br />

Hirschfeld, Azriel e14729,<br />

e14732, e15576<br />

Hirsh, Vera LBA7500, 7511,<br />

7590, 7592, TPS7618<br />

Hirte, Hal W. 3082, 5001,<br />

5019, e13011, e19616<br />

Hisamatsu, Kazufumi e11503<br />

Numerals refer to abstract number<br />

Hisamoto, Akiko 2607, 7045,<br />

7576, e18027<br />

Hishida, Tomoyuki e17554<br />

Hishiki, Tomoro 9577<br />

Hitre, Erika 2522, 3530<br />

Hitz, Felicitas 9045<br />

Hixson, Douglas C. e21116<br />

Hiyama, Eiso 9577<br />

Hjelm-Eriksson, Marie 4561,<br />

TPS4696^<br />

Hlavata, Zuzana e19020<br />

Hlubocky, Fay J. 6117,<br />

e13026<br />

Ho, Adelyn 9043<br />

Ho, Alan Loh 5509^, 5514,<br />

5545^<br />

Ho, Albert 8596<br />

Ho, Alice Y. 1118<br />

Ho, Anthony D. 6519<br />

Ho, Anthony D 10040,<br />

10041^<br />

Ho, Cheryl 7020, e18079<br />

Ho, Gui Yi e19523<br />

Ho, Gwo Yaw e19598<br />

Ho, Han Kiat e13023<br />

Ho, Linus 4051, e14548<br />

Ho, Maria Yi 3610, e14588<br />

Ho, Seong-Hyun e19574<br />

Ho, Steffan Nicholas 2610<br />

Ho, Thai Huu 4624<br />

Ho, Xuan Dung 641<br />

Hoang, Anh 4556, 4624<br />

Hoang, Tien e16010<br />

Hoang-Xuan, Khê 2, 2023<br />

Hobbs, Adele e15164<br />

Hobday, Timothy J. 4047,<br />

e14594<br />

Hobeika, Amy 2585<br />

Hoch, Ute 5048, e13577<br />

Hochberg, Ephraim P.<br />

e18520<br />

Hochberg, Fred 2009<br />

Hochberg, Jessica e13022<br />

Hochhaus, Andreas 3541,<br />

6504, 6506, 6509^, 6510,<br />

6511, TPS6641, e21063<br />

Hochman, Jimena e19007<br />

Hochman, Tsivia e11010,<br />

e11064<br />

Hochster, Howard S. e14534<br />

Hochwald, Steven N.<br />

TPS4136<br />

Hodge, James W. 2526<br />

Hodge, Rachel 10009<br />

Hodges, David e11069<br />

Hodgson, Darren R e15141<br />

Hodgson, David C. 9524<br />

Hodgson, John Graeme 3509,<br />

3511, 3575<br />

Hodgson, Lydia 7513<br />

Hodi, F. Stephen 2502,<br />

CRA2509, 8507, 8508,<br />

8516, 8569, TPS8603<br />

Hoefler, Gerald e14066<br />

Hoehler, Thomas 2522,<br />

3530, 3539<br />

Hoejberg, Lise 8545<br />

Hoelscher, Arnulf e14527<br />

Hoelzer, Dieter 6500^<br />

Hoenigschnabl, Selma<br />

e14087<br />

Hoering, Antje 3078, 8041<br />

Hoerring, Antje 8095<br />

Hoersch, Dieter 4118<br />

Hoersch, Silke 3578<br />

Hoeyer-Hansen, Maria Helena<br />

e19056<br />

H<strong>of</strong>er, Silvia 2067<br />

H<strong>of</strong>f, Paulo M. e15562,<br />

e20502<br />

H<strong>of</strong>f, Paulo Marcelo 6055<br />

H<strong>of</strong>f, Paulo Marcelo 2038,<br />

e11513, e12513, e14702,<br />

e21040<br />

H<strong>of</strong>f, Paulo 3544, 3598<br />

H<strong>of</strong>fman, Arnold e19565<br />

H<strong>of</strong>fman, Eric W. 8547,<br />

e17558<br />

H<strong>of</strong>fman, Karen Elizabeth<br />

e15181<br />

H<strong>of</strong>fman, Robert M. 1063,<br />

2044, 10072, e14014,<br />

e14700, e21051<br />

H<strong>of</strong>fman-Censits, Jean H.<br />

4526, e15012<br />

H<strong>of</strong>fmann, Andreas-Claudius<br />

2086<br />

H<strong>of</strong>fmann, Chen 2078<br />

H<strong>of</strong>fmann, Isabell 1082<br />

H<strong>of</strong>heinz, Ralf 3541, 4083,<br />

4090<br />

H<strong>of</strong>man, Paul 7591<br />

H<strong>of</strong>man, Veronique 7591<br />

H<strong>of</strong>mann, Hans-Stefan 7105<br />

H<strong>of</strong>mann, Lawrence V<br />

e21153<br />

H<strong>of</strong>mann, Marco H. 3092<br />

H<strong>of</strong>mann, Susanne 6567<br />

H<strong>of</strong>meister, Craig C. e16027<br />

H<strong>of</strong>sli, Eva 4015<br />

H<strong>of</strong>stetter, Gerda 5111<br />

H<strong>of</strong>stetter, Wayne Lewis<br />

4069, 4078, 4086,<br />

e14547, e14548, e14669<br />

H<strong>of</strong>städter, Ferdinand e16520<br />

Hogan, Katherine e14179<br />

Hogan, Laura E. 9507<br />

Hogan, William J. 6614,<br />

8092<br />

Hogarth, Linda 3100<br />

Hogendoorn, Pancras C. W.<br />

10081<br />

Hogeveen, Sophie 6084,<br />

9111<br />

Hogg, David 8502^, 8517<br />

Hogge, Donna 6601<br />

Hohenberger, Peter<br />

LBA10008, 10009, 10031,<br />

10067, 10096, TPS10101<br />

Hohenberger, Werner 10031<br />

Hohl, Raymond J. 3056<br />

Hohloch, Karin 8052<br />

Hoiczyk, Mathias 10032^,<br />

10046<br />

Hoiom, Veronica 8588,<br />

10521<br />

Hojat Farsangi, Mohammad<br />

6557<br />

Holbrechts, Stephane 3559<br />

Holbrook, Jaimee S 4063,<br />

6623<br />

Holcombe, Randall F. e21109<br />

Holcr<strong>of</strong>t, Christina e14020<br />

Holden, Debra J. 6046<br />

Holden, Scott N. 2567<br />

Holder, Chad A 3055<br />

Holder, Dara Denise e15018,<br />

e15019<br />

Holdh<strong>of</strong>f, Matthias 2042<br />

Holen, Kyle D. 3101,<br />

e14554, e16571<br />

Holes, Leanne 6518^<br />

Holinski-Feder, Elke 3550<br />

Holla, Soumya e14710<br />

Holland, Jaymes 4504<br />

Holland, John M 5596^,<br />

e16045<br />

Holland, William S. e17537<br />

Hollander, Thomas e14584<br />

Hollebecque, Antoine 3000,<br />

3021, 3026, 3104, 4614,<br />

e13118<br />

Hollema, Harry 10581<br />

Holleran, Julianne L. 2533,<br />

3031<br />

Hollowell, Courtney M.P.<br />

e13513, e13532<br />

Holman, Allison e16059<br />

Holman, Laura L. 1518<br />

Holman, Rose Marie 2589<br />

Holmberg, Leona 4533<br />

Holmes, Frankie Ann 547,<br />

1017<br />

Holmes, Holly Michelle 9072<br />

Holmström, Benny 4668<br />

Holodny, Andrei 7052<br />

Holst, Frederik 10501<br />

Holt, Abby E 6099<br />

Holt, Cheryl L e16543<br />

Holt, Nanna 4015<br />

Holtan, Shernan G. 8599<br />

Holte, Harald 8074<br />

Holten-Andersen, Mads 3576<br />

Holter, Jennifer L. e17001,<br />

e17010<br />

Holter, Spring 3567, 4058<br />

Holtorf, Megan L. 10011^<br />

Holtzhausen, Alisha 10563<br />

Holtzman, Matthew Peter<br />

e14184<br />

Holzer, Alison K e21071,<br />

e21153<br />

Holzhausen, Hans-Juergen<br />

522<br />

Holzmann, David 8563<br />

Homann, Nils 4080, 4083,<br />

4090<br />

Homayounfar, Kia 3600,<br />

e14161, e14167<br />

Homerin, Michel 2605,<br />

TPS4152<br />

Homma, Akihiro 5542<br />

Homma, Sakae e18127<br />

Homma, Shigenori 10578<br />

Hommura, Fumihiro 7571<br />

Hon, Henrique 6005, 6084,<br />

9032<br />

Hon, Jeremy K. 7590, 7592,<br />

TPS7618<br />

Honarpishe, Helen 1109<br />

Honavar, Santosh 9515,<br />

9568<br />

Honda, Yoshihiro e19521<br />

Honecker, Friedemann 4590,<br />

4597, 9114<br />

Honegger, Juergen Bernd<br />

8526<br />

Honey, R. John 4633<br />

Hong, Dae Sik e14688,<br />

e19554<br />

Hong, David S. 3106, 3107^,<br />

3598, 4639, 8551, 8594,<br />

10510, 10607, e13020,<br />

e13506, e13595, e19004,<br />

e19055<br />

Hong, Dr Shengyan e18073<br />

Hong, Fangxin 8007, 9008<br />

Hong, Ji Hyung e14558,<br />

e19614<br />

Hong, Jun Hyuk e15080<br />

Hong, Jung Yong e13094<br />

Hong, Junshik e14137<br />

Hong, Junsik e14685<br />

Hong, Ruey-Long e16050<br />

Hong, S. Peter 9575<br />

Hong, Seung-Hyun 7089<br />

735s


Hong, Seung-Mo 3568<br />

Hong, Sook Hee e14558,<br />

e18050<br />

Hong, Sung Joon 4628<br />

Hong, Sung-Hyeok 9575<br />

Hong, Theodore S. 4021,<br />

e14615<br />

Hong, Waun Ki 5524, 7078<br />

Hong, Yong Sang 3568,<br />

3592, e13104, e13560,<br />

e14072<br />

Hong, Young Seon e14558<br />

Hongo, Fumiya e15055,<br />

e15099<br />

Honisch, Ellen 5097, e21020<br />

Honma, Rio e17520<br />

Honma, Yoshitaka e14126<br />

Honoré, Charles e14158<br />

Honoré, Stéphane 2060<br />

Honovic, Lorena 9097<br />

Hood, Brian L e15560<br />

Hood, Deborah D. 6086<br />

Hood, Nicky 1019<br />

Hooda, H.S. e16002<br />

Hoog, Jeremy 3047<br />

Hoogerbrugge, Peter 9550,<br />

10077<br />

Hook, Jane 10081<br />

Hooker, Craig 1573, e17552<br />

Hooley, Regina 1109<br />

Hoon, Dave S. B. 2519<br />

Hooper, Catherine L. e19637<br />

Hooshmand, Susanne M.<br />

3594<br />

Hope, Andrew J. 5587<br />

Hopkins, Jenny R e16569<br />

Hopkins, Judith O. 9014<br />

Hopkins, Wendie e15056<br />

Hopman, Wilma e19541<br />

Hoppe, Stephanie 9012<br />

Hoppenreys, Laurent 9012<br />

Hoppensteadt, Debra e13115,<br />

e13117, e15008, e15538<br />

Hopper, John L. 1502, 3567<br />

Hoppo, Toshitaka e14689<br />

Hoque, Ehteshamul e11533<br />

Hor, Pooja e13030<br />

Horai, Takeshi 10604,<br />

e18004, e18014, e18017<br />

Horak, Christine E. e13504<br />

Horak, Mary 8533<br />

Horard, Béatrice 9011<br />

Horchani, Ali e15122<br />

Horenblas, Simon 4611<br />

Horgan, Anne 4109, e14524,<br />

e14592<br />

Horgan, Meaghan e12034<br />

Horgan, Paul G 3599, 3611,<br />

e14054, e14092, e14110,<br />

e14117<br />

Horger, Marius 4115<br />

Hori, Mitsuo 8094<br />

Horie, Hisanaga e14091<br />

Horiguchi, Jun 540, 613,<br />

TPS1141<br />

Horiike, Atsushi 10569,<br />

10604, e18004, e18014,<br />

e18017<br />

Horinouchi, Hidehito 7070<br />

Horinouchi, Takashi 1103<br />

Horio, Yoshitsugu e17510,<br />

e18145<br />

Horn, Leora 6068, 7094,<br />

7509, 7568, 7589<br />

Hornberger, John C. 10584<br />

Horndler, Carlos 3553,<br />

e14041<br />

Horne, Heather 4043<br />

Hornedo, Javier 1111<br />

Horneffer, Yvonne 3529<br />

Hornicek, Francis J 10058<br />

Hornick, Jason L. 10029<br />

Horning, Sandra J. 8003,<br />

8007<br />

Hornslien, Kjersti 7027<br />

Horowitz, David Paul 7050,<br />

e15162, e17562<br />

Horowitz, Laurie 10528,<br />

e21008<br />

Horowitz, Neil S. 5063<br />

Horowitz, Nina Ruth 1109<br />

Horowitz, Peleg 2020<br />

Horrigan, Stephen 10605,<br />

e14090<br />

Horsman, Janet M 6113<br />

Horst, Heinz-August 6500^<br />

Hortobagyi, Gabriel N. 512,<br />

515, 527, 537, 539, 540,<br />

551, 559, 600, 612, 1012,<br />

1029, 1047, 1102, 1104,<br />

9018, 10613, e13582<br />

Horwitz, Eric M. 5530, 5583<br />

Horwitz, Steven M. 8060,<br />

8063, 8069<br />

Hoshi, Manabu 10073<br />

Hoshino, Mika e13015<br />

Hoshino, Mirei 2519<br />

Hoshino, Yoshinori 10531<br />

Hosing, Chitra 6546, 6622,<br />

8040<br />

Hoskin, Peter LBA4512,<br />

4551<br />

Hosnii, Shama 1098<br />

Hosokawa, Ayumu e14062<br />

Hosokawa, Masahito e21007<br />

Hosomi, Yukio 7012, e18099<br />

Hosonaga, Mari 635<br />

Hospital, Valerie 8591<br />

Hossain, Akm Mosharraf 1586<br />

Hossain, Mohammed S 2576<br />

Hotko, Yevhen 3535<br />

Hotta, Katsuyuki 2607, 7045,<br />

7576, e18027<br />

Hotta, Tomomitsu 8050<br />

Hotte, Sebastien J. 3082,<br />

4514, 5502, e13011,<br />

e19616<br />

Hou, Jeannie 8579, 8585<br />

Hou, Jian 8097, e18572^<br />

Hou, Jing-Zhou 6563<br />

Hou, Mei e18033<br />

Hou, Ming-Feng 1043<br />

Hou, Ningqi 1101, 1116<br />

Hou, Shengcai e18013<br />

Hou, Steve j 10528<br />

Hou, Wei TPS4136, e14612<br />

Hou, Yijuan 3093<br />

Houbiers, Ghislain 3559<br />

Houck, William A. TPS658<br />

Houede, Nadine 9012,<br />

e13594, e15142<br />

Hough, Holly 6054<br />

Houghton, Peter 9541, 9544,<br />

9581<br />

Houk, Alice 6103<br />

Houlgatte, Remi 8048<br />

Hountis, Panagiotis e17553<br />

Houot, Roch 2514, 8057<br />

Hourdequin, Kathryn<br />

Cunningham 6013<br />

House, Larry 2611<br />

Housset, Martin e13578<br />

Houston, Kathleen Emilie<br />

Mary 8538<br />

Houston, Nicole D. 2545<br />

Houston, Stephen 511, 8539<br />

Houston, Teresa 1529<br />

Houts, Arthur C. 5014<br />

Houtsma, Daniel 10518<br />

Houweling, Leanne e16526<br />

Hoverman, J. Russell 6109,<br />

e16536<br />

Hovey, Elizabeth J. e15152<br />

Howard, Dianna S. 6502<br />

Howard, Jason 5549<br />

Howard, Leslie M. e19543<br />

Howard, Margaret e16543<br />

Howard, Scott C. 6530, 9502<br />

Howard-McNatt, Marissa<br />

Michelle 6127<br />

Howe, Erin N. 564<br />

Howell, Doris 6035, 6039,<br />

9131<br />

Howell, Elizabeth A. e15513<br />

Howitt, Brooke E 5011<br />

Howitt, John 614<br />

Hoyle, Martin 3602<br />

Hoyos, Sergio 642<br />

Hozaeel, Wael 4083, 4090<br />

Hrinczenko, Borys e21133,<br />

e21154<br />

Hristova, Gergana 2547<br />

Hromas, Robert TPS5602<br />

Hrstic, Irena 9097<br />

Hruby, Gregory e15019,<br />

e15021<br />

Hsia, Te-Chun 7549,<br />

TPS7614<br />

Hsiao, Chao e15542<br />

Hsiao, Wendy C. 6057<br />

Hsieh, H Ben 10574<br />

Hsieh, James 4604<br />

Hsieh, Susie 6132<br />

Hsieh, Wen Chuan e11068<br />

Hsieh, Wen Son 4103, 8003<br />

Hsing, Ann W 4646<br />

Hsu, Cheng-Lung e16050<br />

Hsu, Chih-Hung e14576<br />

Hsu, Chiun 1061, 4103<br />

Hsu, CS e14699<br />

Hsu, Feng-Ming e14576<br />

Hsu, Frank James 7077<br />

Hsu, Jack e16047<br />

Hsu, Joann e15084<br />

Hsu, Karl TPS3118,<br />

TPS6641<br />

Hsu, Limin 1029, 1104<br />

Hsu, Shiaowen David 2585<br />

Hsu, Tina e16558<br />

Htut, Myo 8038, 8089<br />

Hu, Adriana e12017<br />

Hu, Beihong 8593<br />

Hu, Boyu 8595<br />

Hu, Chao-su 5535<br />

Hu, Chaosu e16039<br />

Hu, Cheng-ping 7091, 7520<br />

Hu, Chunhong 7091, 7559<br />

Hu, Eddie e12017<br />

Hu, Jethro Lisien 2080, 2084<br />

Hu, Jing TPS6642^<br />

Hu, Kai e16054<br />

Hu, Kaiji e13123<br />

Hu, Kenneth 5531, 5559,<br />

5581, 9024<br />

Hu, Leijun 2516<br />

Hu, Qiong 10527, e18123<br />

Hu, Xi-Chun 10567, e11567,<br />

e11568<br />

Hu, Xioaping P 3055<br />

Hu, Yi e13521<br />

Hu, Zhihong e15538<br />

Hu, Zhihuang e13021<br />

Hu, Zhiquan 4628<br />

Hu, Zhiyuan 1027<br />

Hua, Emily 505<br />

Hua, Guoqiang 6539<br />

Hua, Zhaolai e14539<br />

Huang, Alex Yee-Chen 9520<br />

Huang, Alexander 2525<br />

Huang, Baisong 4033<br />

Huang, Bee-Yau e13088,<br />

e15010<br />

Huang, Bin 5095<br />

Huang, Bo 5558<br />

Huang, Chao Hui e13551<br />

Huang, Chao H 5500,<br />

e13535<br />

Huang, Cheng 3040, 7091,<br />

7548<br />

Huang, Chiun-Sheng TPS649,<br />

TPS651<br />

Huang, Chu-Chun 5109<br />

Huang, Chunzi 2576<br />

Huang, Eric 7066, 7582,<br />

7594, e12007<br />

Huang, James 7043, e17015<br />

Huang, Jianan 7520<br />

Huang, Jing e13110,<br />

e14502, e14734<br />

Huang, Kun e19053<br />

Huang, Lily 1569<br />

Huang, Lingkang e15059<br />

Huang, Lyen C. 6010<br />

Huang, Meijuan e14648,<br />

e18033<br />

Huang, Pei-Ming e14576<br />

Huang, Peng 518, e15188<br />

Huang, Ruili 10025<br />

Huang, Shang-Yi 8097<br />

Huang, Sharon 2519<br />

Huang, Sherry e21148<br />

Huang, Shiang-Fu e16050<br />

Huang, Shu-Min 1061, 1079<br />

Huang, Susie Yi 2059<br />

Huang, Ta-Chen e14576<br />

Huang, Wei 10011^<br />

Huang, Weidong 603, 608,<br />

614<br />

Huang, Wenmei TPS2618,<br />

8065<br />

Huang, Xuan 4569<br />

Huang, Yajue 10528, e21008<br />

Huang, Yan e13021<br />

Huang, Ying e18090, e18517<br />

Huang, Yingjie 604, TPS661<br />

Huang, Yiran LBA4537, 4628<br />

Huang, Yong 4102<br />

Huang, Yuan-Shung 1504<br />

Huang, Yuhui 2010<br />

Huang, Zhimin e18090<br />

Huber, Rudolf M. 7001,<br />

TPS7617<br />

Huberman, Kety H 7052<br />

Hubert, Catherine e14044<br />

Hubner, Richard LBA4001<br />

Huchot, Eric 1574<br />

Hucke, Christian 8079<br />

Hudes, Gary R. TPS2613,<br />

TPS2614, 4526, 4605,<br />

TPS4686, e15070, e19580<br />

Hudis, Clifford 557, TPS668,<br />

CRA1002, 1006, 1016,<br />

TPS1143, TPS1145, 6015,<br />

10514, 10618<br />

Hudson, Christopher 8542<br />

Hudson, Lori L. 2021,<br />

e15061<br />

Hudson, M’Liss A. 4594<br />

Hudson, Malcolm e16501<br />

Hudson, Melissa M. 6030,<br />

9502, 9505, CRA9513,<br />

9526, 9527, 9531<br />

Huebner, Alexander e21044<br />

Huebner, Kay 1007<br />

Huellein, Jennifer 6519<br />

Huelsewig, Carolin e11534<br />

736s Numerals refer to abstract number


Hueltenschmidt, Beatrix 5512<br />

Huelves, Miriam e11035,<br />

e11527, e12015, e17502,<br />

e17503, e17515, e18011,<br />

e18521<br />

Huether, Robert 9518<br />

Huettmann, Andreas 8030<br />

Huges, Elisha 7023<br />

Huggins-Puhalla, Shannon<br />

Leigh TPS1138, 3054<br />

Hughes, Andrew M. e15141,<br />

e21037<br />

Hughes, Brett Gordon Maxwell<br />

7079^, 8538, e16501,<br />

e19642<br />

Hughes, David J. e12031<br />

Hughes, Kevin S. CRA1505<br />

Hughes, Lorie L. 1005<br />

Hughes, Lucinda 1517<br />

Hughes, Melissa E 599, 1125<br />

Hughes, Nicholas Peter 1066<br />

Hughes, Steven TPS4136<br />

Hughes, Timothy P. 6505^,<br />

6509^<br />

Hughes-Davies, Luke TPS665<br />

Hugosson, Jonas 4668<br />

Huh, Joo Ryung e18501<br />

Huh, Sang Y. TPS3635^,<br />

7035<br />

Huh, Warner King TPS5115,<br />

e13087<br />

Hui, Ai-Min 8017, 8033,<br />

8034, 8064<br />

Hui, David 9002, 9023,<br />

9025, 9049, 9062, 9065,<br />

9069, e19528, e19584,<br />

e19602, e19603, e19613<br />

Hui, Edwin P. e14528<br />

Hui, Edwin Pun 5511<br />

Hui, Joyce e14528<br />

Hui, Pei 5099, e15581<br />

Hui, Rina 550<br />

Hui, Siu e16557<br />

Hui, Zhouguang e14511<br />

Huidobro Vence, Gerardo<br />

e18040, e18146<br />

Huijbers, Anouck e14151<br />

Huillard, Olivier e13017<br />

Huitema, Alwin D. R. e13028<br />

Huitema, Alwin D.R. 2550,<br />

2596<br />

Huizing, Manon Thirza 1129,<br />

e13051<br />

Hulin, Cyrille 8009, 8014<br />

Hulkki Wilson, Sanna e14088<br />

Hulsh<strong>of</strong>, Maarten C. 4094<br />

Humblet, Yves 3502,<br />

e14044, e14060<br />

Humm, John 4517<br />

Hummel, Horst 8079<br />

Hummelen, Paul Van 5011<br />

Humphray, Sean 544<br />

Humphrey, TIM e13587<br />

Hundemer, Michael 6519<br />

Hunder, Naomi N. H. e13079<br />

Hung, Arthur 10011^<br />

Hung, Mien-Chie 4624<br />

Hung, Shih-Kai e16008<br />

Hunger, Stephen 9506,<br />

9507, CRA9508, 9543,<br />

9548<br />

Hungria, Vânia Tietsche de<br />

Moraes 8095, e18572^<br />

Hunt, Joanne 4553<br />

Hunt, Kelly 503, 1028,<br />

1102, 1107, 1130<br />

Hunt, Wendy 3039, e13604<br />

Hunter, Deborah S. 6514^<br />

Hunter, Krystal 1127<br />

Numerals refer to abstract number<br />

Hunter, Robert F. e14102<br />

Hunter, Terri B. 2559<br />

Hunter, Zachary R 8042,<br />

8106, 8107, e18575<br />

Hunter-Merrill, Rachel 4557<br />

Huntsman, David e14568<br />

Huo, Dezheng 1033, 1101,<br />

1116, 6040<br />

Huo, Lei 594, 1102<br />

Huober, Jens Bodo 523,<br />

1023<br />

Huppert, Nelly e11064<br />

Hur, Hyuk e14084<br />

Hur, Min Hee 1053<br />

Hurd, David Duane 6524^<br />

Hurd, Ralph E. 4660<br />

Hurh, Eunju 9519<br />

Hurley, Judith 595, 608,<br />

1071, e11038<br />

Hurley, Laura 614<br />

Hurria, Arti 1010, 1070,<br />

6015, 9034, 9109, 9123,<br />

e18133, e19586<br />

Hurst, Deborah 6591<br />

Hurtubise, Brenda e19641<br />

Hurvitz, Sara A. TPS648,<br />

TPS649, 1037, 9144<br />

Hurwitz, Herbert 3039,<br />

3042^, 3614, e13589,<br />

e21038<br />

Husain, Amna 6039<br />

Husain, Amreen 5054<br />

Husain, Juhi e18535<br />

Husband, David J. e18068<br />

Huschens, Susanne 9114<br />

Huse, Jason T. 2028<br />

Hussain, Arif e13075,<br />

e15108, e15146, e15148<br />

Hussain, Maha 4, 4506,<br />

4511, 4513, 4547, 4549,<br />

4571, 4573, 4663, 10503<br />

Hussain, Raza e16043<br />

Hussein, Hany 9566<br />

Hussein, Hussein Soudy<br />

e15530<br />

Hussein, Lily 9085<br />

Hussein, Mohamad A. 8011,<br />

8016<br />

Husseini, Faress 2605<br />

Hussey, Juliette M. 1511<br />

Husson, Marie TPS10634<br />

Hutchison, Anne Smith 7094<br />

Hutchison, Robert E. 9524<br />

Huth, James F. 8534<br />

Hutson, Thomas E. 4501,<br />

4603, TPS4676, TPS4682,<br />

TPS4686, e15063,<br />

e15070, e15089<br />

Hutterer, Markus 2053<br />

Hutzschenreuter, Ulrich<br />

e14138<br />

Huyghe, Ivan 1129<br />

Huynh, Anne 6534<br />

Huynh, Hung The 4100^<br />

Huynh, Thanh Khoa 10078<br />

Hwang, Clara 1560, e14160,<br />

e15174<br />

Hwang, Deok Won 7104,<br />

e16003, e18148<br />

Hwang, Eu-Chang e14649<br />

Hwang, In Gyu 4064<br />

Hwang, Jessica 9072,<br />

e19631<br />

Hwang, Jimmy J. 2590,<br />

3095, TPS3638, e14009,<br />

e14569, e14658<br />

Hwang, Jimmy 4102, e19046<br />

Hwang, Jin Wook e17523<br />

Hwang, Jun-Eul e14649<br />

Hwang, Larn e15542<br />

Hwang, Michael e14069,<br />

e14078<br />

Hwang, Rosa F. 1028<br />

Hwang, Scott N 3055<br />

Hwang, Seong-Geun e14504<br />

Hwang, Siok Gek Jacqueline<br />

5551, 5580<br />

Hwang, Tae-Ho e14566<br />

Hwang, Tsann-Long e20514<br />

Hwang, Wei-Ting 4596<br />

Hwang, Wenke 6052<br />

Hwang, William e18528<br />

Hwu, Patrick 2510, 8514,<br />

8551, e19009, e19014,<br />

e19039<br />

Hwu, Wen-Jen 2510, 8508,<br />

8514, e19009, e19014,<br />

e19039<br />

Hyland, Andrew 1529<br />

Hyland, John e13570,<br />

e21024<br />

Hylands, Lucy e13601<br />

Hyman, David Michael 5030,<br />

5043<br />

Hymes, Kenneth B. e21082<br />

Hyodo, Ichinosuke 4002<br />

Hyoju, Sanjiv Kumar e11094<br />

Hyseni, Entela 5093, 5094,<br />

e15551<br />

Hysert, Pat 1529<br />

Hyslop, Terry e15163<br />

Hyun, Eric D. 8584<br />

Hyung, Woo Jin e14504,<br />

e14652<br />

Hölscher, Arnulf H 4090<br />

Hölzle, Erhard 8505<br />

Hörsch, Dieter 4122, e14513<br />

I<br />

Iaccarino, Giancarla 8083<br />

Iacobucci, Ilaria 6531<br />

Iacono, Rosanne e11505<br />

Iacovelli, Roberto 4621, 4629<br />

Iadanza, Manlio e11071<br />

Iaffaioli, Rosario Vincenzo<br />

e14130<br />

Iaffaioli, Rosario Vincenz<br />

e11052, e13539<br />

Iafrate, A. John 6606, 7508,<br />

7589, 8522<br />

Iafrate, Franco e14096<br />

Iaiza, Emiliana 3612<br />

Iannacone, Claudio 7550<br />

Iannessi, Antoine e13547<br />

Iannone, Robert 10573,<br />

e13598, e13601<br />

Iannotti, Nicholas e13113,<br />

e18063^<br />

Iasonos, Alexia 5030, 5090,<br />

5108<br />

Iau, Philip 1064<br />

IbañezdeCáceres,<br />

Inmaculada 3601, e14093,<br />

e14144<br />

Ibanez Martinez, Jose 4089<br />

Ibanez, Carlos E. 2101<br />

Ibarz, Luis 4579<br />

Ibeas, Patricia e11035,<br />

e11527, e12015, e17502,<br />

e17503, e17515, e18521<br />

Iberico, Carlos 2531<br />

Iborra, Severine 5007<br />

Ibrahim, Azman e18068<br />

Ibrahim, Fady 2098<br />

Ibrahim, Nageatte 8569<br />

Ibrahim, Nuhad K. 10613<br />

Ibrahim, Ramy A. 2503^<br />

Ibrahim, Sherif Abdelaziz<br />

e21041<br />

Ibrahim, Toni 4627, 9005,<br />

10615, e11054<br />

Ibrahimi, Adil e14726<br />

Ibsen, Per 2069<br />

Ibusuki, Mutsuko e11017<br />

Ichida, Takafumi 4104<br />

Ichihara, Eiki 2607, 7576,<br />

e18027<br />

Ichikawa, Kimihisa e14516<br />

Ichikawa, Wataru 4012<br />

Ichikawa, Yasushi e14047,<br />

e14050<br />

Ichinose, Yukito 6112, 7003,<br />

7045, TPS7110, 7549<br />

Ichiyanagi, Osamu e15057,<br />

e15088<br />

Ichiyasu, Hidenori e18059<br />

Icli, Fikri 1572, 1596<br />

Ictech, Sandra 2029^, 2036<br />

Ida, Kohmei 9577<br />

Ida, Satoshi e13553<br />

Idbaih, Ahmed 2023<br />

Idelevich, Efraim e14571,<br />

e19529<br />

Idoate, Miguel A. 8572,<br />

e18008<br />

Idowu, Michael O 10565<br />

Idrees, Kamran e14184<br />

Ieguchi, Makoto 10073<br />

Iganej, Shawn e16031<br />

Igarashi, Kentaro 10075,<br />

e20505<br />

Igarashi, Seiji 7021<br />

Iglesias Dios, Jorge Luis 3093<br />

Iglesias, David A. e13516<br />

Iglesias, Lara 5516^<br />

Iglesias, Mar 4089<br />

Ignatiadis, Michail 515<br />

Iguchi, Haruo 4031<br />

Iizasa, Toshihiko e21054<br />

Ijichi, Kei e16032<br />

Ikai, Iwao e14518<br />

Ikeda, Hideko e17540<br />

Ikeda, Masafumi 4031, 4042<br />

Ikeda, Norihiko 7012<br />

Ikeda, Satoshi e19583<br />

Ikeda, Takaya 7569<br />

Ikeda, Tetsuo e14536<br />

Ikeda, Yuji 5037, e15579<br />

Ikegame, Kazuhiro 6533<br />

Ikegami, Toru e14536,<br />

e14602<br />

Ikemura, Shinnosuke e18058<br />

Ikezawa, Kenji e14518<br />

Ikhlaque, Nadeem 4085<br />

Ikpeazu, Chukwuemeka<br />

e16001<br />

Ilagan, Venus M 8091, 9081<br />

Ileana, Ecaterina 4554<br />

Ilhan-Mutlu, Aysegul 2024<br />

Ilie, Marius 7591<br />

Illarramendi, Jose J. 10512,<br />

10595<br />

Illerhaus, Gerald 2040, 4090,<br />

8031<br />

Illidge, Timothy M 8056<br />

Ilson, David 4057, 4061,<br />

TPS4144<br />

Im, Annie P. 6563<br />

Im, Hae Kyung 9516,<br />

e11554<br />

Im, Ra Joo 1053<br />

Im, Seock-Ah 597^, TPS649,<br />

1003, 2542, 3076, 3624,<br />

4124, 10621, e11532,<br />

e14136, e21160<br />

737s


Im, Young-Hyuck 598^, 644,<br />

TPS649, 1003, 2542,<br />

e11055^<br />

Imada, Toshio 4070<br />

Imai, Yukihiro 7528<br />

Imamura, Fumio 7028,<br />

e18007, e18025, e18134,<br />

e18141, e19556<br />

Imamura, Hiroshi 4012<br />

Imamura, Jun e14622<br />

Imamura, Takahisa e21084<br />

Imamura, Yoshinori e16034<br />

Imaoka, Hiroshi e14540<br />

Imbeaud, Sandrine e14059<br />

Imbert, Yves 9012<br />

Imbevaro, Silvia e15072<br />

Imoto, Shigeru 1106, 1119<br />

In, Reika 590<br />

Ina, Kenji TPS3642<br />

Inaba, Masaaki 1106<br />

Inaji, Hideo 540<br />

Inal, Ali 638, e14526<br />

Inamine, Morihiko 5086<br />

Inanc, Mevlude 638, e14089,<br />

e14526, e14670<br />

Inatani, Hiroyuki 10075<br />

Inatani, Michiyasu 3022<br />

Inauen, Roman Ignaz 3595,<br />

10033<br />

Inazawa, Johji 10520<br />

Incio, Jaoa 1026<br />

Inclan, Alejandro A. 1086<br />

Inculet, Richard I. 6049<br />

Indio, Valentina 10087<br />

Indresh, Desai e14675<br />

Indushekar, Subbanna<br />

e14721<br />

Infante, Jeffrey R. 2579,<br />

TPS2621, 3006^, 3008,<br />

3023, 3033, 3041, 3062,<br />

3067, 3077, 3097, 3102,<br />

TPS3118, 3528, 8510,<br />

8556, e13076, e13077,<br />

e13603<br />

Infante, Maurizio 7061<br />

Infanzon, Erick e12043<br />

Ingaramo, Maria Clara<br />

e13042<br />

Ingle, Ashish M 9500, 9540,<br />

9541, 9581<br />

Ingle, James N. 1005, 1006,<br />

10586, e13501<br />

Ingrosso, Antonella 7551<br />

Ingui’, Manuela 10055<br />

Inic, Momcilo e21136<br />

Inic, Zorka Momcilo e21136<br />

Inman, Brant Allen 4670<br />

Innocenti, Federico e13120^,<br />

e15086<br />

Innominato, Pasquale F.<br />

3547, TPS9154, e14006<br />

Inomata, Masafumi 3569<br />

Inou, Takashi e14004<br />

Inoue, Akira 7086, 7515,<br />

7563, 7565, 7571,<br />

e18129<br />

Inoue, Hiromi 4634<br />

Inoue, Hiroyuki e13014<br />

Inoue, Junichiro e17020<br />

Inoue, Kenichi 613<br />

Inoue, Masanori 7038<br />

Inoue, Takahiro 10520<br />

Inoue, Takayo 3086<br />

Inoue, Tomio 4626<br />

Inoue, Yasuhiro e14029,<br />

e14039, e14541, e21053<br />

Insa, Amelia 1074<br />

Intra, Mattia 1115<br />

Inukai, Michio TPS3642<br />

Inwards, David James 9057<br />

Inzerillo, John J. e13109<br />

Ioakimidis, Leukothea I<br />

e18575<br />

I<strong>of</strong>fe, Olga B. 1519, 10572<br />

Ioka, Tatsuya 4031, 4035,<br />

e14518<br />

Ionescu, Diana 549^, 7020<br />

Ionta, Maria Teresa e14094<br />

Iop, Aldo 586, e11071<br />

Ippolito, Massimo e18096<br />

Iqbal, Hassan e16043<br />

Iqbal, Mudassar 6612<br />

Iqbal, Omer M. e15538<br />

Iqbal, Sheikh Usman 3602,<br />

e14028, e14030, e14061<br />

Iqbal, Syma 3584, 3622,<br />

4019, 4096, 4113,<br />

e13084, e14031, e14679<br />

Iqbal, Zafar 6612<br />

Irmscher, Romy e14630<br />

Iro, Heinrich 5512<br />

Irrera, Giuseppe e18564<br />

Iruarrizaga, Eluska e11525,<br />

e17522, e18031<br />

Irvin, William Johnson<br />

TPS656<br />

Irwig, Michael 2532<br />

Irwin, Blair Billings 6041<br />

Irwin, Melinda TPS669,<br />

TPS1614, 9007<br />

Isa, Luciano 9120<br />

Isa, Shunichi 7000, e18126,<br />

e18128<br />

Isaacs, Claudine 1521<br />

Isaacson, Jeffrey D. 4041,<br />

e13028<br />

Isabel Gil Garcia, Maria<br />

e14097<br />

Isac<strong>of</strong>f, William H. e14582<br />

Isada, Akira e18135<br />

Isaka, Keiichi 10557<br />

Isak<strong>of</strong>f, Steven J. 508, 510,<br />

1006, 1009, 1026, 3021<br />

Isambert, Nicolas 3026,<br />

10014, 10020, 10042,<br />

10056, 10062, 10078,<br />

TPS10102^, e13009,<br />

e13010<br />

Isdale, Debora 4098<br />

Isenberg, Gerald 3621<br />

Isenring, Elizabeth e19550<br />

Ish-Shalom, Maya 4564,<br />

4619, e15058<br />

Isham, Crescent R 5544<br />

Ishibashi, Olivia 9557<br />

Ishida, Hiroo e13029<br />

Ishida, Tadashi 7528,<br />

e18025<br />

Ishida, Takanori e11530<br />

Ishida, Tsuyoshi 9574<br />

Ishido, Kenji 4046<br />

Ishigami, Hironori e14530<br />

Ishiguro, Hiroshi e21052<br />

Ishihara, Shinichi e18037<br />

Ishii, Genichiro 1552, 7044,<br />

e17554, e18064<br />

Ishii, Hiroshi 4031, e14506,<br />

e14677<br />

Ishii, Mari 10577<br />

Ishii, Takeshi 9574<br />

Ishii, Yoshiki 7561<br />

Ishii, Yoshiyuki 10531<br />

Ishikawa, Takashi TPS1141,<br />

e14047<br />

Ishikawa, Toshiaki e14127<br />

Ishikawa, Yuichi 3625,<br />

10541, e18017<br />

Ishikura, Satoshi 7017,<br />

7028, 8050<br />

Ishimori, Ayako e18019<br />

Ishimoto, Osamu 7086,<br />

7561, 7565, e19521<br />

Ishimoto, Takatsugu 10523,<br />

e13553<br />

Ishioka, Chikashi e19611<br />

Ishioka, Kouta e18058<br />

Ishioka, Tatsunari e13076<br />

Ishitobi, Makoto 1103<br />

Ishizawa, Kenichi 8029,<br />

8050<br />

Ishizuka, Naoki 8050,<br />

e15004<br />

Isidoro, Miguel e13119<br />

Isikdogan, Abdurrahman 638,<br />

e14526<br />

Isla, Dolores 1531, 7095,<br />

e12000, e12012, e18055,<br />

e18131<br />

Islam, Nahida 8080<br />

Islam, Rezwan e14008<br />

Ismaeel, Fakher e19597<br />

Ismael, Gustavo e11087<br />

Ismail, Jeffri R. M. e16506<br />

Ismail-Khan, Roohi 1585,<br />

TPS2620<br />

Isobe, Hiroshi 7070, 7515,<br />

7571, e18129, e18135<br />

Isobe, Kazutoshi e18127<br />

Isobe, Takeshi e13072<br />

Isobe, Yasushi 8050<br />

Isogai, Pierre K e15180<br />

Isonishi, Seiji 5003<br />

Israel, Robert Joseph 4662<br />

Issa, Amalia M. 6005<br />

Issa, Djamila e16526<br />

Issa, Jean-Pierre 5524<br />

Issels, Rolf D. 10054<br />

Istomin, Yuriy e20514<br />

Isu, Kazuo 9574<br />

Itakura, Haruka e21153<br />

Itakura, Meiji e21054<br />

Italiano, Antoine 9536,<br />

10005, 10006, 10014,<br />

10023, 10027, 10035,<br />

10042, 10057, 10062,<br />

10078, e13594<br />

Itano, Osamu e14080<br />

Itano, Satoshi e14080<br />

Ito, Kenichiro 7571<br />

Ito, Tatsuo 7578<br />

Ito, Tomoaki e14139<br />

Ito, Toshikazu 1119<br />

Ito, Yoshinori 540, TPS659,<br />

e19512<br />

Itoga, Sakae 7046<br />

Itoh, Kuniaki 585, 8050<br />

Itoh, Kyogo e13046, e13050<br />

Itoh, Norimasa e18126<br />

Itokazu, Gail 8085<br />

Itri, Loretta 1016, 2555,<br />

4077<br />

Itsura, Peter 5107<br />

Ittmann, Michael M. 4643<br />

Iturbe, Julian e11021<br />

Itwaru, Sandy Ann e15140<br />

Itzhaki, Aviran e14616<br />

Ivancic, David 518<br />

Ivanov, Krasimir e21100<br />

Ivanov, Roman A. 9060<br />

Ivanov, Sergey TPS4692^<br />

Ivanova, Anastasia e13074<br />

Ivanova, Ivayla 10521<br />

Ivanovic, Marija 5050<br />

Ivanyi, Philipp e15190<br />

Iversen, Helle K. 2069<br />

Iversen, Trine Zeeberg 2565,<br />

2574<br />

Ives, Denise I. e14611,<br />

e14665<br />

Iveson, Timothy 2535, 2594,<br />

LBA4000, 4005<br />

Ivey, Alison TPS4136,<br />

e14612<br />

Ivy, S. Percy 2009, 2606,<br />

3078, 3082, 3101, 5019,<br />

10004<br />

Iwae, Shigemichi 5542<br />

Iwagami, Shiro 10523,<br />

e13553<br />

Iwai, Toshiki e18091<br />

Iwai, Toshinori 5556<br />

Iwakuma, Nobutaka e13046,<br />

e13050<br />

Iwamoto, Shigeyoshi TPS3642<br />

Iwamoto, Takayuki 545,<br />

1012, 1047<br />

Iwanaga, Ichiro e14062<br />

Iwasa, Satoru e14126<br />

Iwasaki, Masashi e21004<br />

Iwasaku, Masahiro 7528,<br />

e18025, e18134<br />

Iwasashi, Hajime e19508<br />

Iwase, Hiroaki e14596<br />

Iwase, Hirotaka 540, e11017<br />

Iwata, Hiroji 512, 540,<br />

TPS661, TPS670<br />

Iwatani, Tsuguo 591<br />

Iwatsuki, Masaaki 10523,<br />

e13553<br />

Iyengar, Neil M. 6582,<br />

10618<br />

Iyer, Gopa 4527, 4581^,<br />

5030<br />

Iyer, Renuka V. 4038,<br />

e14610, e14659, e14690<br />

Iyer, Shrividya 7533, 7596,<br />

e16570<br />

Iza, Estibaliz e17522<br />

Izarzugaza, Yann 5520,<br />

e15094, e18069, e18106,<br />

e21015<br />

Izetti, Patricia e13546<br />

Iznaga, Normando 2515<br />

Izquierdo, Angel 1588,<br />

e12012<br />

Izrailov, Roman e14717<br />

Izumi, Namiki 4104<br />

Izumi, Tetsuta 1103<br />

Izumi, Yotaro 7038<br />

Izzo, Francesco e14130,<br />

e14654<br />

J<br />

Jabbour, Abdo e20500<br />

Jabbour, Elias 6501, 6527,<br />

6544, 6577, 6585, 6587,<br />

6589, 6592, 6594, 6597,<br />

6603, 6607, TPS6635<br />

Jabbour, Melhem Salim 9110<br />

Jabbour, Nicola e11097<br />

Jabbour, Salma e14555<br />

Jabel, Nicole e16565<br />

Jac, Jaroslaw TPS1143,<br />

e13040<br />

Jaccard, Arnaud 8026<br />

Jacene, Heather A. 8552<br />

Jackisch, Christian 640,<br />

1023, e11040^<br />

Jackman, David Michael<br />

7524, 7525, 7547, 7553,<br />

7556, 7579, 7588, 10592<br />

Jackson, Alan e14015<br />

Jackson, Erica TPS2620<br />

Jackson, Gilchrist L. 8543<br />

738s Numerals refer to abstract number


Jackson, Graham H. 8015<br />

Jackson, Jessica 4085,<br />

e14564<br />

Jackson, Marianne 6093<br />

Jackson, Nadine 4027<br />

Jackson, Vicki A. 6004,<br />

6048, 6106, 9004, 9101<br />

Jacob, Linu Abraham TPS649<br />

Jacob, Rojymon e14555<br />

Jacob, Simon 634<br />

Jacobi, Oded e13018<br />

Jacobs, Andrew 9113<br />

Jacobs, Benjamin e16022<br />

Jacobs, John Robert 5534,<br />

e16019<br />

Jacobs, Linda A. 9145<br />

Jacobs, Lisa K. 518, 1128<br />

Jacobs, Paula M. 2077<br />

Jacobs, Samuel A. 611, 1025<br />

Jacobs-Small, Mona 4111,<br />

7077<br />

Jacobsen, Anne-Birgitte<br />

e14531<br />

Jacobsen, Britta M. 564<br />

Jacobsen, Tove Flem 6619^<br />

Jacobson, Kristine B e21060,<br />

e21111<br />

Jacobus, Laura S. 8061<br />

Jacobus, Susanna J. e15170<br />

Jácome, Alexandre A.A.<br />

e14599<br />

Jacot, William 568^<br />

Jacquemier, Jocelyne 543<br />

Jacquet, Alexandra 5069<br />

Jadhav, Nitin 3011<br />

Jaeckle, Kurt A. 2004, 2015,<br />

2031, 2032, e16569<br />

Jaeger, Bernadette Anna<br />

Sophia 554, TPS1146,<br />

1602, 10540<br />

Jaeger, Ulrich 8058<br />

Jaekel, Nadja TPS6642^<br />

Jaen, Ana 10608, e11535,<br />

e21088<br />

Jaen, Juan C. 2043, e13580<br />

Jaffa, Ariel J. 1564<br />

Jaffa, Zachary 5582<br />

Jaffe, Elaine S. 2528, 8051<br />

Jaffee, Elizabeth M. e14585<br />

Jafferji, Insiya e13500,<br />

e21029<br />

Jafri, Syed Faisal e14632<br />

Jafri, Syed Hasan Raza<br />

e16009<br />

Jagannath, Sundar 8011,<br />

8016, 8020, 8035,<br />

e21006<br />

Jagdt, Bjoern e14103<br />

Jager, A. 10504, e16526<br />

Jagiello Gruszfeld, Agnieszka<br />

I. 10582<br />

Jaglal, Michael 2099,<br />

e21123<br />

Jaglowski, Samantha Mary<br />

6508<br />

Jagtap, Deepa e17528<br />

Jahagirdar, Balkrishna N.<br />

5566<br />

Jahan, Thierry Marie 3058,<br />

7030<br />

Jahanzeb, Mohammad 605,<br />

TPS1137<br />

Jahn, Franziska e19579<br />

Jahn, Thomas Michael 6518^<br />

Jahng, Patrick e16523<br />

Jahnke, Kristoph 2040, 2054<br />

Jaimes, Natalia e19052<br />

Jain, Alka e13042<br />

Jain, Amit 4598<br />

Numerals refer to abstract number<br />

Jain, Kunal e19550, e19622<br />

Jain, Minish Mahendra 2592,<br />

3011, e13127<br />

Jain, Nitin 6535^, 6582,<br />

e14609<br />

Jain, Pooja e18068<br />

Jain, Prantesh 1611<br />

Jain, Rakesh K. 1026, 2009,<br />

2010, 2012, 2025, 4112<br />

Jain, Salvia Sanjay 5016<br />

Jain, Shivi 8085, e14186,<br />

e14590<br />

Jain, Supriya K. e11512<br />

Jain, Vinay Kumar 8081<br />

Jaiyesimi, Ishmael A. 628,<br />

e14608, e17015<br />

Jajac Brucic, Lana e15031<br />

Jajeh, Ahmad e18510<br />

Jakesz, Raimund 514, 10501<br />

Jakic Razumovic, Jasminka<br />

e21126<br />

Jakkoju, Rakesh 8536,<br />

e15181<br />

Jakob, Andreas e15044<br />

Jakob, Augustinus TPS4148<br />

Jakob, John Andrew 8584<br />

Jakobsen, Anders Kristian<br />

Moeller 3513, 3573, 5052,<br />

e14134<br />

Jakobsen, Erik 7026<br />

Jakobsen, Jan Nyrop 7001<br />

Jakovljevic, Ksenija e12020<br />

Jakowatz, James G. 8524<br />

Jakubowiak, Andrzej J. 8011,<br />

8020, 8035, TPS8114<br />

Jakubowski, Loni 3057<br />

Jalal, Prasun 4116<br />

Jamal, Rahima 2547<br />

Jamal-Hanjani, Mariam 3100<br />

Jamaluddin, Muhammad<br />

Faisal e14687, e19059<br />

Jamaluddin, Nimah e15528<br />

James, Danelle Frances 6507,<br />

6508, 6515^<br />

James, Iain 10553<br />

James, Katherine 6126<br />

James, Michelle D 3516<br />

James, Nicholas David<br />

e15091<br />

James, Ralph e21026<br />

James, Roger 4004, 4029<br />

James, Samuel e15582<br />

Jameson, Gayle S. 2516<br />

Jameson, Michael B. e15054<br />

Jamieson, Rebekah 10563<br />

Jamoul, Corinne 8559<br />

Jamshed, Arif e16043<br />

Jamshed, Saad e18558<br />

Janakiraman, Manickam 4527<br />

Janas, Mette S. TPS9149^<br />

Janelsins, Michelle Christine<br />

9009, 9010, 9028, 9141,<br />

9142<br />

Janevski, Angel 10508<br />

Janeway, Katherine A. 9503<br />

Jang, Geundoo 4093<br />

Jang, Hee-Jin e16534<br />

Jang, Raymond Woo-Jun<br />

e14003<br />

Jang, Se Jin 3568<br />

Jang, Sekwon e19000,<br />

e19025<br />

Jani, Ashesh B. e15006<br />

Janik, John E. 8051<br />

Janjigian, Yelena Yuriy 4057,<br />

4061, TPS4144, 7580,<br />

9105, e14586<br />

Jankilevich, Gustavo e15521<br />

Jankovic, Radmila e12020<br />

Jankowitz, Rachel Catherine<br />

611<br />

Jankowski, Janusz LBA4001<br />

Jankowsky, Rüdiger 4115<br />

Janku, Filip 3023, 3106,<br />

3107^, TPS3117, 8551,<br />

10510, 10607, e13020,<br />

e13534, e13595<br />

Janne, Pasi A. 7010, 7503,<br />

7547, 7588, TPS7615,<br />

e18093<br />

Janne, Pasi Antero 7007,<br />

7510, 7530, 7553, 10592<br />

Janni, Johann Wolfgang<br />

TPS1146, 5097, e11556<br />

Janni, Wolfgang 554, 1072,<br />

1081, 1090, 1600^, 1602,<br />

10540, 10599, e21020<br />

Janot, Francois e16013<br />

Jansen, Jeroen P. e19010<br />

Janson, Eva Tiensuu 4015<br />

Janssen, Eva e19031<br />

Janssen, Jan e15044<br />

Janssen, Marc e15166<br />

Jantus-Lewintre, Eloisa 7058,<br />

7060, 10594<br />

Janus, Cecile P 8039<br />

Janus, Nicolas 1095, 5067<br />

Janz, Nancy K. 6023, 6024<br />

Jara-Sanchez, Carlos e11535<br />

Jarabo, Jose Ramon 10564<br />

Jarboe, Jamie 4117, 4545<br />

Jardel, Henry 8014<br />

Jardine, Victoria L e18100<br />

Jares, Pedro 3553, e14041<br />

Jariwala, Krutika e16527<br />

Jarkowski, Anthony e19041<br />

Jarlenski, Donna 4056<br />

Jarnagin, William R. 3577,<br />

3620<br />

Jarner, Mikala F. TPS9149^<br />

Jarosz, Bozena 603<br />

Jarosz, Mirna 3533<br />

Jarrar, Doraid 7051<br />

Jarrett, Lee Ann e16038<br />

Jarzab, Barbara 5508, 5518,<br />

5591, e14159<br />

Jarzab, Michal e14159,<br />

e21049<br />

Jaskowiak, Nora T. 1101,<br />

1116<br />

Jaspan, Tim 9517, TPS9596<br />

Jasra, Sakshi e16565<br />

Jassem, Ewa 10561<br />

Jassem, Jacek 505, 603,<br />

TPS649, 4050, 9046,<br />

10561, e21099<br />

Jasso Mosqueda, Juan<br />

Guillermo e14061<br />

Jatoi, Aminah 5076, 7605<br />

Java, James 5056, 5057<br />

Javed, Yasir Ahmed TPS1137<br />

Javle, Milind M. 4038, 4054,<br />

e19645<br />

Javvadi, Prashanthi 10624<br />

Jaw-Tsai, Sarah e13028<br />

Jawed, Irfan e14064<br />

Jawlekar, Gajendra 3013<br />

Jaworska, Magdalena e21099<br />

Jayabalan, David 8036<br />

Jayachandran, Aparna 8542<br />

Jayakumar, Arumugam<br />

e18557<br />

Jayanti, Venkata K e14640<br />

Jayaprakash, Nalini 6530<br />

Jayaram, Anuradha e14687,<br />

e19059<br />

Jayasekera, Jinani e15146<br />

Jayasuriya, Chami e16567<br />

Jayson, Gordon C. e14015<br />

Jean, Annie 564<br />

Jeannet, Laëtitia 8089<br />

Jeannin, Gaelle 7503<br />

Jeannon, Jean-Pierre 5522<br />

Jedrzejczak, Wieslaw W.<br />

e18572^<br />

Jeffers, Michael 3019,<br />

e13576, e18100<br />

Jefford, Michael 3504, 6111,<br />

9087, e16507<br />

Jego, Virginie 3107^<br />

Jehn, Christian e14140<br />

Jelaca-Maxwell, Kathy 4568<br />

Jelinek, Jaroslav 5524<br />

Jelovac, Danijela 5062^<br />

Jemal, Abdusebur 9555<br />

Jemming, Chloé 2605<br />

Jenkins, Emily Oldham 1524<br />

Jenkins, Marjorie e21105<br />

Jenkins, Robert B. 2008b,<br />

4009, 4565<br />

Jenkins, Valerie A 5527<br />

Jennings, Dominique 2009,<br />

2025, 3036<br />

Jenny, Matthias e13508<br />

Jensen, Anni Ravnsbæk<br />

e14063, e14105<br />

Jensen, Brett T e15124<br />

Jensen, Gail e18144<br />

Jensen, Helle Anita e14517<br />

Jensen, Kristian e21129<br />

Jensen, Kurt S. 2069<br />

Jensen, Lindsay G e14555<br />

Jensen, Roxanne E 9130<br />

Jensen, Steen Lindkaer<br />

e11022<br />

Jeon, Eun Kyoung e14634,<br />

e18050<br />

Jeon, Justin Y e14114<br />

Jeon, Mijin 6564<br />

Jeon, Ung Bae e14566<br />

Jeon, Yoon-Kyung 7566<br />

Jeon, Young Woo e19554<br />

Jeong, Deok Hyun e14114<br />

Jeong, Jae-Heon e14128<br />

Jeong, Jong-Hyeon TPS1142<br />

Jeong, Seong Hyun e18519<br />

Jeong, Seung-Yong 3624<br />

Jephcott, Catherine Rachel<br />

e14100<br />

Jerin, Lems 9097<br />

Jerkeman, Mats 8074<br />

Jeruss, Jacqueline Sara<br />

TPS1134<br />

Jeschke, Udo e15529<br />

Jeter, Stacie 518, 9103,<br />

10509<br />

Jethwa, Alexander 6519<br />

Jett, James R. 7605<br />

Jeung, Hei-Cheul e14652<br />

Jewell, Andrea e15548<br />

Jewett, Michael A. S. 4500,<br />

4633, e15118<br />

Jewett, Michael A 4535<br />

Jewitt, Natalie C. 2063<br />

Jeyabalan, Neera e14001<br />

Jeziorski, Arkadiusz e19022<br />

Jha, Gautam Gopalji e15211<br />

Jhaveri, Komal L. 10618<br />

Ji, Fei 2503^, TPS2621<br />

Ji, Jianfei 2087<br />

Ji, Jiuping Jay 3031, 5020<br />

Ji, Jun Ho e14072, e16003,<br />

e18148<br />

Ji, Lingyun 1065, 1124,<br />

e13084, e19506<br />

Ji, Qinghai e16039<br />

Ji, Rui-Ru 8593<br />

739s


Ji, Yongling e17527<br />

Jia, Jingquan e13589<br />

Jia, Wang Xiao e17512<br />

Jia, Xiaoyu 4517, 4532<br />

Jia, Zhen 10567, e11567,<br />

e11568<br />

Jian, Hou 8095<br />

Jian, Weiguo e15002<br />

Jiang, Bin 9017<br />

Jiang, Brittany 5575<br />

Jiang, Chen 2533<br />

Jiang, Chunsheng 1091,<br />

e21146<br />

Jiang, Dingfeng 6034<br />

Jiang, Guanchao e17504,<br />

e21103<br />

Jiang, Haiyan 9131<br />

Jiang, Haoyuan 7548<br />

Jiang, Jing 7062<br />

Jiang, Ming e18514<br />

Jiang, Ping 2579<br />

Jiang, Qin 3090<br />

Jiang, Su 9017<br />

Jiang, Wen-chang e14668<br />

Jiang, Xuejuan 4575<br />

Jiang, Yixing TPS1138, 2553,<br />

3049, 3054<br />

Jiang, Yizhou 2535, 2594,<br />

4005<br />

Jiang, Yong Jian e14651<br />

Jiang, Yuqiu 10504<br />

Jiang, Zefei e13036<br />

Jiang, Zhi-Qin 3561, 3598,<br />

e14071<br />

Jiao, Wei e13107<br />

Jiao, Xiaolong 570<br />

Jibara, Ghalib e14656<br />

Jibb, Lindsay 9556<br />

Jie, Fei 8587<br />

Jie, He e14502, e18013<br />

Jilla, Swapna 9055<br />

Jillella, Anand P. 6573, 8108<br />

Jimenez, Camilo 4641,<br />

e13592<br />

Jimenez, Connie R. e18094<br />

Jimenez, Joaquin J e14593<br />

Jimenez, Jose 7041<br />

Jimenez, Laura 3096<br />

Jimenez, Marta 10035<br />

Jimenez, Paula TPS4685,<br />

TPS4688, 10546, e14671,<br />

e15144<br />

Jimenez, Rachel e16500<br />

Jimenez, Ulpiano 7011<br />

Jimenez, Valerie 8089<br />

Jimenez-Baranda, Sonia 8571<br />

Jimeno, Antonio 2567, 2568,<br />

3017, 3024, LBA5000,<br />

e13048<br />

Jin, Bo e18038<br />

Jin, Chen e14651<br />

Jin, Denise 2567<br />

Jin, Feng 5535<br />

Jin, Kate 7519^<br />

Jin, Mei Hua 8099<br />

Jinno, Hiromitsu 1106,<br />

10531, e11060, e14080<br />

Jirillo, Antonio e15072<br />

Jitawatanarat, Potjana<br />

e18538<br />

Jo, Booil 9051<br />

Jo, Deog Yeon e14529<br />

Jo, Jungmin e11508<br />

Jobe, Blair A e14689<br />

Jobkar, Terri L. 8011<br />

Jobo, Toshiko 5003<br />

Jodrell, Duncan Ian 3100<br />

Joe, Andrew K. 8502^,<br />

8503^, TPS8603<br />

Joe, Teresa 4643<br />

Joens, Thomas e15531<br />

Joensuu, Heikki 507,<br />

TPS647, LBA10008<br />

Joensuu, Timo K. e13035<br />

Joergensen, Jens Christian<br />

Riis e14134<br />

Joergensen, Stine 3513<br />

J<strong>of</strong>fe, Steven 1605<br />

Johal, Balvindar Singh<br />

e14176<br />

Johannessen, Dag Clement<br />

LBA4512, 4551<br />

Johansen, Cathrine e14637<br />

Johansen, Julia S. 3548,<br />

8545<br />

Johansson, Carolina 8588<br />

Johansson, Harriet Ann 519<br />

John, Esther M 1502<br />

John, Sonia e17010<br />

John, Tom e17539<br />

John, William J. LBA4, 1068,<br />

7075, LBA7507, e18138<br />

Johne, Andreas 3107^<br />

Johns, Amanda L. 5014<br />

Johnson, Amy J 6507, 6508<br />

Johnson, Ana TPS5600<br />

Johnson, Brendan Mark 9117<br />

Johnson, Bruce E. 7525,<br />

7588, 7589, 10592<br />

Johnson, Bruce E 7099,<br />

7547, 7553<br />

Johnson, Candace S. e18118<br />

Johnson, Catherine 1013,<br />

1098<br />

Johnson, Dave 6518^<br />

Johnson, David B. 6133<br />

Johnson, David H. 6068,<br />

7094, 7568<br />

Johnson, Derek Richard 2065<br />

Johnson, Doug 6102<br />

Johnson, Frank E. 6079,<br />

6081, e15528, e19593<br />

Johnson, Jeffrey L. 6526,<br />

8000<br />

Johnson, John R 3074<br />

Johnson, Jonas Talmadge<br />

8533<br />

Johnson, Karen 1501,<br />

e14005<br />

Johnson, Laureen 3013<br />

Johnson, Maureen 6086<br />

Johnson, Melissa Lynne 3083,<br />

7580<br />

Johnson, Nathalie 9019<br />

Johnson, Peter W. M. 8056,<br />

TPS10633^<br />

Johnson, Philip James 4033,<br />

TPS4150, e13507<br />

Johnson, Russell F. e16513<br />

Johnson, S e13522<br />

Johnson, Valen E. 8091,<br />

9081, 9083<br />

Johnson, William 593<br />

Johnsson, Anders 3573<br />

Johnston, Claire 2552, 3030<br />

Johnston, Curtis 5523<br />

Johnston, Donna 9556<br />

Johnston, Gregory 3619,<br />

e14046<br />

Johnston, Mary Ann e19582,<br />

e19585<br />

Johnston, Patrick B. 8028<br />

Johnston, Richard 4669,<br />

e15207<br />

Johnston, Stephen R. D. 531,<br />

TPS661, TPS666,<br />

TPS10633^<br />

Johnston, Tammie e21026<br />

Johnston, Taren A 5027<br />

Johnstone, David 7022<br />

Jolivet, Jacques 4105,<br />

e14706<br />

Jolly, Damien 1584<br />

Jolly, Douglas J. 2101<br />

Joly, Bertrand 8026<br />

Joly, Florence 636, 2583,<br />

LBA4567^, LBA5000,<br />

e14650, e15535<br />

Joly, Katelle e11019<br />

Jonak, Zdenka 5065<br />

Jonasch, Eric 4503, 4609,<br />

4615, 4624, 9029,<br />

e15075, e15092<br />

Jonat, Walter 1084, e13503<br />

Jones, Cheryl 2566<br />

Jones, Chris TPS9596<br />

Jones, Daniel 8596<br />

Jones, David R 7008<br />

Jones, David 3057<br />

Jones, Dennie V. 5549<br />

Jones, Desiree 6551, 9118<br />

Jones, Emma 3008<br />

Jones, Frank 2585<br />

Jones, Glenn e19570<br />

Jones, Heather L e14184<br />

Jones, Howard e15582<br />

Jones, Jeffrey Alan 6508,<br />

e18508<br />

Jones, Jennifer M 9131<br />

Jones, Jeremy e15084,<br />

e15178<br />

Jones, Julia J 10050<br />

Jones, Keith 3022<br />

Jones, Kimberly 4048<br />

Jones, Louise K. TPS10633^<br />

Jones, Mark M. 6559, 6627,<br />

6632<br />

Jones, Mary Elizabeth 3027<br />

Jones, Natalie Beckman 1125<br />

Jones, Richard 7562<br />

Jones, Robert J. 4558<br />

Jones, Robert Peter 3613<br />

Jones, Robert T. 2020<br />

Jones, Robin Lewis 10011^,<br />

10039, 10086, TPS10099<br />

Jones, Roy B. 4533, 8102<br />

Jones, Sarah TPS8605<br />

Jones, Siân 3068<br />

Jones, Suzanne Fields 3008,<br />

3033, 3041, 3067, 3097,<br />

e13076<br />

Jones, Suzanne 3006^, 3021<br />

Jones, Tamekia 9548<br />

Jones, Terence 5543<br />

Joniau, Steven e13061<br />

Jonker, Derek J. 3504, 3515,<br />

3572, e14154<br />

Jonnalagadda, Sreenivasa<br />

e14584<br />

Jonsson, Håkan 4668<br />

Jonsson, Olafur G. 9590<br />

Jonstam, Gustaf 4561<br />

Joo, Jungnam e16534,<br />

e18119<br />

Joo, Sam e17009<br />

Jooss, Karin 2562<br />

Joosten, J J.A. 7009<br />

Jordan, Berit e19579<br />

Jordan, Emmet 1543,<br />

e21147<br />

Jordan, Karin e19579<br />

Jordan, Mary Ann e19571<br />

Jorgensen, Jeffrey L. 6501<br />

Jorissen, Robert N 3623<br />

Jose, Chakiath TPS4690<br />

Joseph, David John TPS4690<br />

Joseph, Kathie-Ann P.<br />

e11004<br />

Joseph, Kurian e15540<br />

Joseph, Lindsay J. TPS2621<br />

Joseph, Loren 6562<br />

Joseph, Mathew John 7035<br />

Joseph, Richard Wayne 8590<br />

Joseph, Vijai e15022<br />

Joshi, Alaknanda 10532<br />

Joshi, Amit 5585, e16028,<br />

e17500<br />

Joshi, Brijen L 6067<br />

Joshi, Monika e17560<br />

Joshi, Nikhil Purushottam<br />

2072<br />

Joshi, Rohit e19513<br />

Joshua, Anthony Michael<br />

4514, e15118<br />

Joshua, Anthony TPS4695<br />

Jotereau, Francine 2570<br />

Jotte, Robert M. LBA4, 3023,<br />

7541<br />

Jouannaud, Christelle 1051,<br />

1095, 5067<br />

Jouary, Thomas LBA8500^,<br />

8532, 8559, 8591<br />

Joubert, Warren Lance 3572<br />

Joulain, Florence 3602<br />

Jouravleva, Elena e13562,<br />

e21058<br />

Jouret-Mourin, Anne e14044,<br />

e14060<br />

Jouvet, Anne 2023<br />

Jouybari, Touraj 6556<br />

Jovanovic, Borko 8080<br />

Jove, Jeremy e14168<br />

Jovic, Gordana 10081<br />

Jozefara, Jolanta 1127<br />

Jozwiak, Sergiusz 10619<br />

Ju, Gui-zhi e13558<br />

Ju, Melody e12009<br />

Ju, Xiaoming e13545<br />

Ju, Yan 10543<br />

Juan Vidal, Oscar 4587,<br />

e15144, e17545, e18049<br />

Juan, Gloria 3580<br />

Juan, Oscar e18015, e18053<br />

Juarez, Eva e19594<br />

Jubb, Adrian M. 1066<br />

Juckett, Mark 6570<br />

Judde, Jean-Gabriel e15161<br />

Judge, Joshua Marshall<br />

e19062<br />

Judson, Benjamin 5529,<br />

5532<br />

Judson, Ian Robert 10009,<br />

10013, 10038, 10039,<br />

10060, 10067, 10086<br />

Judson, Patricia Lynn 5054<br />

Judy, Brendan 7595<br />

Jueckstock, Julia Kathrin<br />

1081, 5007<br />

Juergens, Herbert 10080<br />

Juergens, Rosalyn A. e18147<br />

Juez, Ignacio 4079<br />

Juhasz-Boess, Ingolf 1523<br />

Juhn, Frank 3533<br />

Julian, Jim A LBA1031, 6049<br />

Julian, Thomas B. TPS657,<br />

1025<br />

Julier, Patrick LBA4001<br />

Julka, Pramod Kumar 2072,<br />

e11082, e15518<br />

Juma, Matthews TPS6638<br />

Jumonville, Alcee 6074,<br />

6570<br />

Jumper, Cynthia A. e21105<br />

Junecko, Beth Fallert 8528<br />

Jung, Ah Rang 6564<br />

740s Numerals refer to abstract number


Jung, Andreas 3519^<br />

Jung, Eun-Jung 4124<br />

Jung, Hwoon-Yong 4093<br />

Jung, Hyun Ae e16003<br />

Jung, Kyung Hae 1003,<br />

2542, e11504, e11508,<br />

e13104<br />

Jung, Minkyu TPS4142,<br />

e14137, e14652, e14685<br />

Jung, Sin-Ho 8000<br />

Jung, Su Yon 1085, 1589<br />

Jung, Yun Hwa e14558<br />

Jungberg, Peter e19540<br />

Jungbluth, Achim A 2589,<br />

TPS8111, e17558<br />

Junker, Niels 2574<br />

Junqueira, Manuela Jacobsen<br />

575<br />

Junqueira, Maribel e15114<br />

Jurado, Josefina Cruz 10052<br />

Jurcic, Joseph G. 6609, 6613<br />

Jure-Kunkel, Maria CRA2509,<br />

2511, 2521, TPS2613,<br />

8593<br />

Juretic, Antonio e21126<br />

Juretic, Manuela 3039<br />

Juric, Dejan 2567, 2568,<br />

e13048<br />

Jurkowska, Monika 8548,<br />

e19022<br />

Jusko, William J e13006<br />

Just-Landi, Sylvaine e15155<br />

Justus, Jacob 1562<br />

Juvekar, S 5585, e16028<br />

Juwara, Lamin 3529<br />

Jürgensmeier, Juliane M<br />

e13541<br />

Jäger, Elke 4080, 4083,<br />

4090, e14557<br />

Jäger, Michael 2584<br />

Jänicke, Martina e14138<br />

Jöhrens, Korinna e15053<br />

Jönsson, Mattias e13518<br />

K<br />

K, Jagannathrao Naidu<br />

e12501<br />

Kaasa, Stein e19648, 9040<br />

Kaatsch, Peter 9573<br />

Kabata, Hiroki e18058<br />

Kabbinavar, Fairooz F.<br />

e13045<br />

Kabbinavar, Fairooz 3614,<br />

TPS4683, TPS4686<br />

Kaburagi, Takayuki 7012<br />

Kaburaki, Kyohei 10569,<br />

10604, e18004, e18014,<br />

e18017<br />

Kacan, Turgut e16005,<br />

e19575<br />

Kacel, Elizabeth L 9146<br />

Kachnic, Lisa A. 4030,<br />

TPS9150<br />

Kadalayil, Latha 4004, 4029<br />

Kadambi, Ananth 6553<br />

Kadan-Lottick, Nina 9546<br />

Kadar, Lianna e14531<br />

Kadia, Tapan M. 6517, 6528,<br />

6558, 6587, 6589, 6592,<br />

6597, 6607, TPS6635<br />

Kadlec, Adam e16544<br />

Kadlec, Bohdan e18062<br />

Kadlubek, Pamela CRA1505,<br />

e16532<br />

Kadowaki, Shigenori e14038<br />

Kadowaki, Tadashi 5518,<br />

e13511, e19055<br />

Kaechele, Volker 4065<br />

Kaeding, Allison 9548<br />

Numerals refer to abstract number<br />

Kaehler, Katharina K. 4034<br />

Kaempgen, Eckhart 8532,<br />

e19032<br />

Kaen, Diego e14521<br />

Kaen, Luis Alberto e14521<br />

Kafka-Ritsch, Reinhold<br />

e14719<br />

Kafri, Zyad 5534<br />

Kaga, Kichizo e17520<br />

Kagami, Yoshikazu 8050<br />

Kager, Leo 10081<br />

Kageyama, Shinichi e19546<br />

Kahan, Zsuzsanna TPS661,<br />

9046<br />

Kahatt, Carmen Maria<br />

TPS652, 2539, e15001<br />

Kahl, Brad S. 8003, 8007<br />

Kahlert, Steffen 1114<br />

Kahn, Daniel e13117,<br />

e15008<br />

Kahraman, Deniz 3044, 7603<br />

Kai, Kazuharu 1047<br />

Kai, Yuki e13072<br />

Kaidar-Person, Orit e18511<br />

Kaira, Kyoichi e18037<br />

Kaiser, Brianne e13025<br />

Kaiser, Karen 9056<br />

Kaiser, Ulrich 3<br />

Kaji, Reiko 7528, 10610<br />

Kajiura, Shinya e14038<br />

Kajiura, Yuka 590<br />

Kakehi, Yoshiyuki e15127<br />

Kakeji, Yoshihiro 3558,<br />

e14033, e14601<br />

Kakizume, Tomoyuki 3088^<br />

Kaklamani, Virginia G. 610,<br />

TPS1134, 1561<br />

Kako, Severine e21018<br />

Kakolyris, Stylianos e14567,<br />

e17553<br />

Kakudo, Yuichi e19611<br />

Kakumanu, Ankineedu<br />

Saranya 2051<br />

Kakuske, Jennifer C e16010<br />

Kalac, Matko e13569<br />

Kalachand, Roshni Deepa<br />

e15016<br />

Kalambakas, Stacey 9519<br />

Kalanovic, Daniel 4631,<br />

e19502<br />

Kalaw, Emarene 8078,<br />

e18515<br />

Kalaycio, Matt E. 6509^,<br />

8055<br />

Kalbacher, Elsa 10042<br />

Kalbakis, Kostas e18105<br />

Kaldate, Rajesh R. 1550,<br />

3627, e13047<br />

Kale, P. 3089^<br />

Kale, Sule e21130<br />

Kalebic, Thea 3077, e13603<br />

Kalemkerian, Gregory Peter<br />

e18018, e18118<br />

Kaley, Thomas Joseph 2008<br />

Kalfin, Michael e16515,<br />

e16559<br />

Kalfoglou, Creton 10619<br />

Kalhor, Neda 7023<br />

Kalil, Karime 3544<br />

Kalinka-Warzocha, Ewa 603<br />

Kalinowski, Katherine H<br />

2074^<br />

Kalinsky, Kevin 584, 8522,<br />

10516<br />

Kalirai, Helen 8523,<br />

TPS8605<br />

Kalkavan, Halime 2086<br />

Kalle, Christ<strong>of</strong> V. 6519<br />

Kallen, Karl-Josef 2573^<br />

Kallenberg, Kai 2079<br />

Kallender, Howard 4108<br />

Kalli, Kimberly 5076<br />

Kallur, Kumar G e11027,<br />

e12030, e14675, e14721<br />

Kalmadi, Sujith R. e18046<br />

Kalmanti, Maria e20002<br />

Kalnicki, Shalom e16058<br />

Kal<strong>of</strong>onos, Haralabos 9090<br />

Kalogeras, Konstantine T.<br />

592, e21018<br />

Kalpathy-Cramer, Jayashree<br />

2012, 2059<br />

Kaltman, Rebecca Davidson<br />

e11084<br />

Kam, Michael K. 5511,<br />

e15110<br />

Kamada, Yoshihiko e11530<br />

Kamai, Takao e15048<br />

Kamal, Arif e16556<br />

Kamal, Muna e15540<br />

Kamalakaran, Sitharthan<br />

10508<br />

Kamat, Ashish M. 4523<br />

Kamata, Minoru e14500<br />

Kamatani, Naoyuki 10520<br />

Kamba, Tomomi e15048<br />

Kambhampati, Suman<br />

e13568<br />

Kamdar, Manali K. e17008<br />

Kamei, Haruhito 7560<br />

Kamel, Ihab R. 4114<br />

Kamel-Reid, Suzanne 2051,<br />

5010, 5105<br />

Kamen, Charles e16564<br />

Kami, Kenjiro e14635<br />

Kamijo, Izumi e13508<br />

Kamimatsuse, Arata 9577<br />

Kaminski, Mark Stefan 8001,<br />

8011, e18530<br />

Kaminsky, Anna 1038<br />

Kaminsky, Michael S 1037<br />

Kamiyama, Toshiya 10578<br />

Kamiyama, Yukari 7021<br />

Kammerer, Klaus Peter<br />

e19533<br />

Kamp, Torsten G. 7572<br />

Kamphuisen, Pieter W<br />

TPS9149^<br />

Kampletsas, Eleftherios 592<br />

Kampmann, Eric 10054<br />

Kamsu-Kom, Nyam e19022<br />

Kamura, Toshiharu 5006,<br />

5085<br />

Kanaan, Mohammed Nawaf<br />

3619, e14045, e14046,<br />

e15554<br />

Kanaan, Zeyad e17004<br />

Kanagavel, Dheepak e14623,<br />

e15029<br />

Kanai, Fumihiko e14622<br />

Kanai, Masashi e14518<br />

Kanakry, Jennifer Ann 8003<br />

Kanan, Sadhana e17003<br />

Kanani, Samir 3590<br />

Kanarek, Norma 1573<br />

Kanayama, Hiro-Omi e15068<br />

Kanazu, Masaki 10577<br />

Kanbara, Hisashige e21007<br />

Kanda, Jossi Ledo 5588<br />

Kandalaft, Lana 5023<br />

Kandasamy, Ravichandran<br />

e19524, e19526<br />

Kandilakis, Giannis 9033<br />

Kandrnal, Vit e11047,<br />

e11072<br />

Kane, Christopher J 4500<br />

Kane, John Michael e14172,<br />

e19041<br />

Kane, Michael P. TPS3112<br />

Kaneda, Hiroyasu e18060<br />

Kaneko, Masahiro 1530<br />

Kanemitsu, Yukihide 3524<br />

Kanerva, Anna e13035<br />

Kaneta, Toshikado 3088^<br />

Kanetsky, Peter A. 1539<br />

Kang, Byung Woog 3554,<br />

6536, 6538, e14074,<br />

e14580, e18533<br />

Kang, Eunyoung 1056<br />

Kang, Gyeong Hoon 3624<br />

Kang, Haiyan 4008<br />

Kang, Hye Jin e14072,<br />

e14580<br />

Kang, Hyunseok e13110,<br />

e21045<br />

Kang, Jin Hyoung TPS7614,<br />

e13061, e14558, e14580,<br />

e18050<br />

Kang, Jung Hun 4064, 9065,<br />

e19603, e19613<br />

Kang, Min H 8073<br />

Kang, Moon Chul e17523<br />

Kang, Myoung Joo e14543<br />

Kang, Sanghee 2036<br />

Kang, Seok Yun 1003, 2542,<br />

TPS4142, e11559, e18519<br />

Kang, Soonmo Peter 2512,<br />

e13106<br />

Kang, Su-yi e14668<br />

Kang, Sung-Soo 1053<br />

Kang, Tae Ho 1053<br />

Kang, Tongjun 1592, 8038,<br />

9512<br />

Kang, Un-Beom 10621<br />

Kang, Won Ki 4011, 4064<br />

Kang, WonKi 10535, e13094<br />

Kang, Xinmei e11095<br />

Kang, Yoon-Koo 4082,<br />

LBA10008, 10085, 10093,<br />

e13092, e13104, e14543,<br />

e14617<br />

Kang, Young-Ah 6564<br />

Kangasniemi, Lotta e13035<br />

Kanji, Zaheer 4058<br />

Kankasa, Chipepo 9558<br />

Kankesan, Janarthanan 1583<br />

Kann, Lisa M. 3068<br />

Kannan, Sadhna 9559<br />

Kanner, Andrew TPS2106<br />

Kanno, Hitoshi e21108<br />

Kannourakis, George<br />

TPS4681<br />

Kano, Jessica e19570<br />

Kantarjian, Hagop 6501,<br />

6503, 6504, 6506, 6509^,<br />

6513, 6516^, 6517, 6520,<br />

6525, 6527, 6528, 6544,<br />

6558, 6559, 6576, 6578,<br />

6585, 6587, 6589, 6592,<br />

6596, 6597, 6603, 6604,<br />

6607, 6624, 6627, 6632,<br />

TPS6635<br />

Kantelhardt, Eva Johanna 580<br />

Kant<strong>of</strong>f, Emily 3105, 4048<br />

Kant<strong>of</strong>f, Philip W. 4543,<br />

4585, 4635, 4648, 4655<br />

Kant<strong>of</strong>f, Philip 4516, 4521,<br />

4582, 4662, 4667,<br />

e15170<br />

Kantor, Andrea TPS8602,<br />

e19046<br />

Kantor, Evan e14621<br />

Kanujia, Arti e13068<br />

Kanz, Lothar e15577<br />

Kanzler, Stephan TPS3641^<br />

Kao, Johnny e15162<br />

Kapadia, Nirav S. 7037<br />

741s


Kaparelou, Maria 583<br />

Kapaun, Christine 3519^<br />

Kaphan, Regis 6633<br />

Kaplan, Ellen B 4515<br />

Kaplan, Henry G. 6560<br />

Kaplan, Joseph 7587<br />

Kaplan, Lawrence D. 8005<br />

Kaplan, Muhammed Ali<br />

e14526<br />

Kaplan, Muhammet Ali 638<br />

Kaplan, Richard S. 3516,<br />

6091<br />

Kaplon, Rita 9544<br />

Kapoor, Nimmi S. 1105<br />

Kapoor, Prashant 8105<br />

Kapoor, Rahul e12014<br />

Kapoun, Ann 3025<br />

Kapp, Amy V. 3543, 3552<br />

Kapp, Daniel Stuart 10576<br />

Kapp, Kerstin 4636<br />

Kappauf, Herbert W. 3588<br />

Kappeler, Christian 4103<br />

Kappelmayer, Janos e21140<br />

Kapur, Payal 4616<br />

Kar, Niladri 2014<br />

Kar, Siddhartha P. 4054<br />

Kara, Ismail Oguz e13523<br />

Karaboué, Abdoulaye 3547,<br />

e14006<br />

Karaca, Halit e14089,<br />

e14670<br />

Karachaliou, Niki e18137<br />

Karagkounis, Georgios 3616<br />

Karahan, Vladislav B. e19012<br />

Karakasis, Katherine 5010<br />

Karakitsos, Petros e17553<br />

Karakousis, C. P. 8534<br />

Karakousis, Giorgos<br />

Constantine e19054<br />

Karam, Andre 3039, e13602,<br />

e13604<br />

Karamchandani, Jason 2043<br />

Karameris, Andreas e21098<br />

Karamlou, Kasra e11042<br />

Karampeazis, Athanasios<br />

7564<br />

Karanes, Chatchada 8038,<br />

8089<br />

Karanth, Siddharth 4130,<br />

4131<br />

Karaoglu, Aziz e15006<br />

Karapanegiotou, Lena 2530<br />

Karapetis, Christos Stelios<br />

3504, 3515, 3532,<br />

TPS4137, e13007,<br />

e17544, e19550<br />

Karaszewska, Boguslawa 9117<br />

Karavasilis, Vasilios e15040<br />

Karayan-Tapon, Lucie 3520<br />

Karayiannakis, Anastasios<br />

e14567<br />

Karcaaltincaba, Musturay<br />

e11509<br />

Kardinal, Carl G. 1586<br />

Kareb, Jacalyn e19588<br />

Karginova, Olga TPS656<br />

Karim, Shanaz e11533<br />

Karimi, Misagh e11562,<br />

e13003<br />

Karjalainen-Lindsberg,<br />

Marja-Liisa 8074<br />

Karkada, Mohan 2588<br />

Karl, Alexander 4530<br />

Karlan, Beth 5072<br />

Karlberg, Mia 3538<br />

Karlin, Nina J. 5544<br />

Karlov, Petr A 4503<br />

Karmakar, Parimal e13533<br />

Karmali, Rashida A. e15515,<br />

e19006<br />

Karmali, Reem e21069<br />

Karne, Sheetal 6615<br />

Karnes, R. Jeffrey 4565,<br />

4657<br />

Karol, Michael D. 3094<br />

Karp, Daniel D. 7608<br />

Karpov, Andrei 6064<br />

Karr, George e11569<br />

Karra Gurunath, Ravichandra<br />

8532<br />

Karrasch, Matthias 8002,<br />

8076<br />

Karrison, Theodore 2561,<br />

4022, 5500, 8075,<br />

e13106<br />

Karsh, Lawrence Ivan 4642<br />

Kartashov, Alex e11067<br />

Karthaus, Meinolf e11528<br />

Karthikeyan, Venkat 5540<br />

Karuturi, Meghan Sri e19603,<br />

e19613<br />

Karve, Sudeep 7101<br />

Karyakin, Oleg TPS4692^<br />

Karzai, Fatima H 3043<br />

Kasahara, Kazuo e17540<br />

Kasahara, Takashi e15071<br />

Kasai, Takashi 7021<br />

Kasamatsu, Takahiro 5085<br />

Kasatkin, Vadim e20503<br />

Kaseb, Ahmed Omar 4116,<br />

e13020, e14664<br />

Kashani-Sabet, Mohammed<br />

8534<br />

Kashigar, Aidin 9032<br />

Kashiwa, Miyuki e17020<br />

Kashtan, Hanoch e14571<br />

Kashyap, Trinayan 1055,<br />

e13549<br />

Kasiborski, Fabrice 3052<br />

Kasimir-Bauer, Sabine 5042,<br />

10599<br />

Kasimis, Basil e14124,<br />

e14125, e14727, e18516,<br />

e19606, e19638<br />

Kasper, Hallie Bieber 9585<br />

Kasper, Stefan TPS3641^<br />

Kasperzyk, Julie 4555<br />

Kasprowicz, Nikola S. 1072,<br />

1602, e21020<br />

Kass, Samantha Lindsay<br />

e19647<br />

Kassam, Alisha 9076<br />

Kassam, Zahra 6084<br />

Kassarjian, Ara 6136<br />

Kassem, Mohammed Azher<br />

9085<br />

Kassem, Nader e20500<br />

Kassem, Neemat e18570<br />

Kasseroler, Marie-Therese<br />

e14719<br />

Kaste, Sue 9527<br />

Kasubhai, Saifuddin M. 3543<br />

Kasuga, Akiyoshi 4046<br />

Kasymjanova, Goulnar<br />

e18061, e18098, e19626<br />

Katabi, Nora 5545^<br />

Katagiri, Katsunori e16020<br />

Katai, Hitoshi 4012<br />

Kataja, Vesa V 507,<br />

TPS4696^<br />

Katakami, Nobuyuki 7515,<br />

7521, 7528, 10610,<br />

e18025, e18134, e19556<br />

Kataoka, Akihiko 10578<br />

Kataoka, Tomoko e16034<br />

Katashiba, Yuichi e18081<br />

Katato, Khalil 7102<br />

Katay, Ildiko 3044<br />

Katayama, Hiroshi 3569,<br />

6112<br />

Katayama, Naoyuki e19546<br />

Katayose, Yu e14011<br />

Kates, Max 4623<br />

Kates, Ronald E. 552<br />

Katheria, Vani 9034, 9109,<br />

9123, e18133<br />

Kathman, Steven 2535, 2594<br />

Kathpal, Madeera 2039<br />

Kato, Jason e13573<br />

Kato, Ken e14126<br />

Kato, Kengo e16020<br />

Kato, Koji 6533<br />

Kato, Shunsuke e19611<br />

Kato, Takashi 3593, e14062<br />

Kato, Takeshi 3524<br />

Kato, Terufumi 7012,<br />

LBA7500, e19583<br />

Kato, Tomoaki 4101<br />

Kato, Tomoyuki e15057,<br />

e15088<br />

Kato, Yuka 7576, e18099<br />

Katre, Ashwini A. 5036<br />

Katsanis, Emmanual e13013<br />

Katsaounis, Panagiotis 5574<br />

Katsaros, Dionyssios<br />

LBA5000, LBA5003<br />

Katsumata, Noriyuki 3084,<br />

5003, 5006, e19535,<br />

e19557<br />

Katsura, Yukitaka 8094<br />

Katsuura, Yutaka 7571<br />

Kattan, Michael W. 1514,<br />

4552, 4570, 4652, 5562,<br />

e15070<br />

Katurakes, Nora C. 6086<br />

Katz, Artur e11014, e12513<br />

Katz, Daniela 10034<br />

Katz, Deborah e19543<br />

Katz, Jessica 2521<br />

Katz, Leah M. 4516<br />

Katz, Sanford R. e17524<br />

Katz, Seth 1016<br />

Katz, Steven J. 6023, 6024<br />

Katz, Terry 7502<br />

Kaubisch, Andreas 4022,<br />

4047, 4098, 4099, 5025,<br />

e14019, e14682<br />

Kaubitzsch, Sabine 2504<br />

Kauff, Noah D. 1519, 5043<br />

Kauffman, Michael 1055,<br />

4634, e13549, e13586,<br />

e15200<br />

Kauffman, Tia N e14160<br />

Kaufman, Bella 5022<br />

Kaufman, Howard 2588,<br />

TPS8604<br />

Kaufman, Jonathan L. 8086,<br />

8100, 8101, e18553,<br />

e18569<br />

Kaufman, Peter Andrew 529,<br />

1526, e11051<br />

Kaufmann, J. e13597<br />

Kaufmann, Manfred 596,<br />

604, 1023<br />

Kaufmann, Scott H 5073<br />

Kauh, John S. 3077, 3094,<br />

4043, e13603, e14614,<br />

e14692<br />

Kauh, John S 2510, 2576<br />

Kaul, Karen 577<br />

Kaun, Stephan 2007<br />

Kauppinen, Sakari e13567<br />

Kaur, Harmeet e19645<br />

Kaur, Jyotdeep e18009<br />

Kaur, Paramjit e13028<br />

Kaur, Sukhwinder e17549<br />

Kaura, Satyin e18562<br />

Kaushal, Aradhana TPS4701<br />

Kaushal, Sunil e14046<br />

Kaushik, Amit e15510<br />

Kaushik, Rucha Vishal 1108,<br />

e11552<br />

Kavan, Petr 9557, e14020<br />

Kavanagh, Brian D. 7526<br />

Kavcic, Marko 1504<br />

Kavianimoghadam, Kaveh<br />

6556<br />

Kawabata, Hidetaka 591<br />

Kawabata, Hugh e17009<br />

Kawabata, Izumi 3100<br />

Kawabe, Takumi 7029<br />

Kawachi, Mark H. e15178<br />

Kawaguchi, Koji e16049<br />

Kawaguchi, Tomoya 7000,<br />

10577, e18126, e18128<br />

Kawaguchi, Yasuo e14091<br />

Kawahara, Masaaki 7017,<br />

7028<br />

Kawai, Hiroki e14510<br />

Kawai, Norisuke e15098<br />

Kawai, Yasutaka e18135<br />

Kawai, Yuki e11564<br />

Kawakami, Yukikiyo e11560<br />

Kawamoto, Hiroshi 9574<br />

Kawamura, Masafumi 7038<br />

Kawamura, Mikio e14541,<br />

e21053<br />

Kawamura, Shigeyuki e13076<br />

Kawamura, Taiichi e14635<br />

Kawano, Hiroyuki e14601<br />

Kawano, Yoshifumi 9574<br />

Kawano, Yuko 10604,<br />

e18004, e18014, e18017<br />

Kawasaki, Hitoshi e14709<br />

Kawase, Akikazu 1552<br />

Kawase, Ichiro e18056,<br />

e18141<br />

Kawashima, Atsunari e15065,<br />

e15069<br />

Kawashima, Osamu 7012<br />

Kawashima, Toana 9514<br />

Kawashima, Yousuke e19521<br />

Kawasugi, Kazuo 9053<br />

Kawate, Takahiko 10544<br />

Kawazoe, Hisashi e15057,<br />

e15088<br />

Kay, Neil E 6122, e13517<br />

Kaya, Serap e14618<br />

Kaya, Yalcin e11083<br />

Kayaleh, Omar 4113<br />

Kaye, James A. e15165<br />

Kaye, Stanley B. 2540, 3048,<br />

3051, 5022, 5035, 10525,<br />

e13601<br />

Kayitalire, Louis 3052<br />

Kayser, Clarissa e19610<br />

Kayser, Jacques e14180<br />

Kazama, Toshifumi e18037<br />

Kazanjian, Kevork 1587<br />

Kazanov, Diana 1564<br />

Kazanov, Dina 1507<br />

Kazeem, Gbenga 1059, 1073<br />

Kazim, Abdul Latif 10609<br />

Kazumoto, Tomoko e15533<br />

Ke, Chunlei TPS670<br />

Ke, Xuehua 6028, 6075<br />

Keam, Bhumsuk 5552, 7566<br />

Kean, Yin 9067<br />

Keane, Maccon M. 502<br />

Keane, Maccon TPS661<br />

Kearney, Jill TPS666<br />

Kearney, Nora e19617<br />

Kearns, Pamela TPS9594<br />

Keating, Anne Therese 8587<br />

Keating, Karen E 10553<br />

742s Numerals refer to abstract number


Keating, Michael J. 6122,<br />

6516^, 6517, 6598<br />

Keating, Nancy Lynn 1100,<br />

6045<br />

Kebenko, Maxim 2504<br />

Kebriaei, <strong>Part</strong>ow 6501, 6540<br />

Kebudi, Rejin 9567<br />

Keck, Michaela K. 5517<br />

Keedy, Vicki Leigh 7094,<br />

7568<br />

Keegan, Patricia 3074<br />

Keeler, Elizabeth 1518<br />

Keeler, Emmett B. 6057<br />

Keeling, Peter e21158<br />

Keenan, Kathleen 10514<br />

Keenan, Robert 7008<br />

Keeney, Gary L. 5004<br />

Keessen, Marianne 2550<br />

Kefford, Richard 550, 3102,<br />

8501, 8510, 8518, 8558,<br />

e13594, e19011<br />

Kehoe, Siobhan Marie<br />

e15501<br />

Kehrer, Diederik F. S. 10504<br />

Keikavoussi, Petra e19032<br />

Keil, Felix 4074<br />

Keil, O. e13597<br />

Keilani, Mohammad 2071<br />

Keilholz, Ulrich 2523,<br />

5516^, 5572, 8532,<br />

e16018, e18010<br />

Keir, Christopher Hunt<br />

10088, 10094, e20511<br />

Keizman, Daniel 4564, 4619,<br />

e15058, e18108<br />

Kelchner, Vanessa 10587<br />

Keldsen, Nina 3548, 5052<br />

Kell, M. R. 1543<br />

Kell, Malcolm R. 1113,<br />

e12038<br />

Kelleher, Fergal C 8520<br />

Kelleher, William Peter 9103<br />

Keller, Jason 8041<br />

Keller, Ulrich 8025, e19031<br />

Kelley, Mark C. 8534<br />

Kelley, Robin Katie 3006^,<br />

3105, 4007, 4102<br />

Kelley, Stacey 6126<br />

Kellman, Robert e16047<br />

Kellock, Trenton Tyler<br />

e14048<br />

Kellokumpu-Lehtinen,<br />

Pirkko-Liisa 507<br />

Kelly, Anne e16515<br />

Kelly, Catherine M. 581,<br />

e21147<br />

Kelly, Catherine Margaret<br />

e19632<br />

Kelly, Catherine M 545<br />

Kelly, Ciara Marie 581<br />

Kelly, John 8520<br />

Kelly, Karen 7005, 7018,<br />

e13551, e17537<br />

Kelly, Kevin R. 8032,<br />

TPS8109<br />

Kelly, Maria e16514<br />

Kelly, Mary A TPS6635<br />

Kelly, Tracy R. e14613<br />

Kelly, Virginia 6511<br />

Kelly, William Kevin 4526,<br />

4655, 4667, e15012<br />

Kelsen, David Paul 4061,<br />

TPS4144, 9094, 9105,<br />

e14586<br />

Kelvin, Joanne Frankel 575<br />

Kemal, Yasemin e14107<br />

Kemeny, Gabor 10533<br />

Kemeny, Margaret 529,<br />

6073, 9004<br />

Numerals refer to abstract number<br />

Kemeny, Nancy E. 3577,<br />

3620<br />

Kemkes, Ariane C. 5523<br />

Kemp, Charisse N. 6509^<br />

Kendle, Ryan F. 8575<br />

Kendra, Kari Lynn e13549<br />

Kenis, Cindy 9035<br />

Kenmotsu, Hirotsugu 7088,<br />

e21007<br />

Kennecke, Hagen F. 3527,<br />

3628, 4118, 6014, 6069,<br />

e14176, e14588, e19547<br />

Kennedy, Andrew S. 3590<br />

Kennedy, Breandan e13570<br />

Kennedy, John 1511, e13520<br />

Kennedy, Kelly Marie e21119<br />

Kennedy, M. John 1543,<br />

e12038<br />

Kennedy, Richard D. 10553<br />

Kennedy, Victoria 9048<br />

Kenney, Kara 518<br />

Kenney, Richard T. e19058<br />

Kenny, Lizbeth M. 5571,<br />

8538<br />

Kenny, Ursula 1559<br />

Kenouchi, Ouarda 10517<br />

Kent, Debra A. 9552, e20007<br />

Kentepozidis, Nikolaos K.<br />

e18105<br />

Kentsis, Alex e13586<br />

Keogh, Louise A. 3567<br />

Keogh, Mary 1046<br />

Keohan, Mary Louise 10002,<br />

10004, 10019, 10057,<br />

TPS10103<br />

Keohane, Catherine 2062,<br />

e11569<br />

Kerbel, Robert S. e15177^,<br />

e19015<br />

Kerbel, Robert S e11061<br />

Kerbrat, Pierre 5028, 10056,<br />

e11012<br />

Kereakoglow, Sandra 9100<br />

Keresztes, Roger 7546<br />

Kerger, Joseph N. 5505<br />

Kerkhoven, Ron M 3065<br />

Kerns, William D e14731<br />

Kerr, Christine e15535<br />

Kerr, Keith M 7583<br />

Kerr, Phil e21135<br />

Kerr, Sarah E 10588<br />

Kerrou, Khaldoun TPS646<br />

Kershenbaum, Anne 1578<br />

Kersten, Christian 3538,<br />

4066<br />

Kersual, Nathalie 5069<br />

Kesari, Santosh 2052, 2101,<br />

TPS2106, TPS2107<br />

Keshari, Rajiv Prasad e14674<br />

Keskin, Muge e21050<br />

Keskin, Serkan e14628<br />

Keskin, Özge e11020<br />

Kesler, Kenneth 7032, 7106,<br />

7108<br />

Kesmodel, Susan 10572<br />

Kessler, Joerg 8079<br />

Kessler, Luisa e11510<br />

Ketchens, Charlean e13531<br />

Ketchum, Steven B TPS6637<br />

Ketterling, Rhett 6614<br />

Kettlety, Timothy R e15565<br />

Kettner, Erika 4023<br />

Keung, Emily 5090, 9086<br />

Keung, Yi-Kong e12017<br />

Key, Nigel e14505<br />

Keyver-Paik, Mignon-Denise<br />

5005<br />

Khabele, Dineo e15582<br />

Khabra, Komel 10060<br />

Khader, Jamal e15024<br />

Khadija, Kalai 10556<br />

Khair, Tina Ashley e11057<br />

Khajavi, Mehrdad e15075<br />

Khaled, Hussein Mustafa<br />

8002<br />

Khalid, Sanaullah 6620<br />

Khalifa, Rania H e18570<br />

Khalife, Riad e20500<br />

Khalil, Ahmed LBA3500^,<br />

3506<br />

Khalil, Iya G. e19055<br />

Khammari, Amir e19019<br />

Khan Manji, Samira 6615,<br />

e14185<br />

Khan, Atif J. e13543<br />

Khan, Awais M. 6586<br />

Khan, Cyrus 6620<br />

Khan, Ghulam 6123<br />

Khan, Hussein e13117<br />

Khan, Iftekhar 7562<br />

Khan, Jamal Anono e13071^<br />

Khan, Khurum Hayat e13599<br />

Khan, Mohammad K. e15006<br />

Khan, Mohid Shakil 4123<br />

Khan, Muhammad Atif 6099<br />

Khan, Qamar J. 9000<br />

Khan, Sadya CRA4502<br />

Khan, Saira 9584, e20008<br />

Khan, Salam 6557<br />

Khan, Seema Ahsan 518,<br />

TPS1134, 2532<br />

Khan, Shah Alam 9580<br />

Khan, Shaheer A. 8549<br />

Khan, Zeba M. 6122,<br />

e18556, e18560, e18561<br />

Khan, Zubair 5528<br />

Khandekar, Alim e17531^<br />

Khanin, Raya 7578<br />

Khanna, Anchit e13566<br />

Khanna, Rahul e16527<br />

Khanna, Samir e15101<br />

Khanna, Sarika e19025<br />

Khanshour, Ammar e12019<br />

Kharaishvili, Gvantsa 1087<br />

Kharfan-Dabaja, Mohamed A.<br />

6586<br />

Kharkevitch, Dmitri D.<br />

e19058<br />

Khar<strong>of</strong>a, Jordan e14613<br />

Khatcheressian, James L.<br />

8567<br />

Khater, Leticia e16041<br />

Khatkov, Igor e14717<br />

Khatri, Vijay P. 3566<br />

Khattri, Arun 5517<br />

Khattry, Navin 9559, e17003<br />

Khawaja, Osmaan e16513<br />

Khayat, David 641, 6110,<br />

e11551<br />

Khazan, Mikhail e12016<br />

Khazanov, R. e11528<br />

Khder, Yasser e15037<br />

Khelif, David Hamid 2048<br />

Khella, Heba WZ 4633<br />

Kheoh, Thian San LBA4518,<br />

4521, 4558<br />

Khetan, Vikas 9515<br />

Khillan, Ratesh e15097<br />

Khin, Sonny 3087, 3529<br />

Khleif, Samir 2577, 2578<br />

Khoa, Mai Trong 1534<br />

Khoja, Leila e21037<br />

Khokhlova, Svetlana<br />

Victorovna e14723,<br />

e15543<br />

Khong, Hoa e11050<br />

Khoo, Lay Poh 8078, e18515<br />

Khoo, Stella 2538<br />

Khoo, Vincent e15199^<br />

Khorana, Alok A. 6059,<br />

TPS9149^<br />

Khouri, Issa F. 6529, 6544<br />

Khouri, Nagi 518, 10509<br />

Khoury, Hanna Jean 6503,<br />

6511, 6577<br />

Khoury, Nabil e20004<br />

Khozin, Sean 7033, 7103<br />

Khrebtukova, Irina 10539<br />

Khurana, Monica 9553<br />

Khuri, Fadlo Raja 2576,<br />

3094, 5501, 5560, 5570,<br />

7048, 7059, 7097, 7589,<br />

10625, e13026, e21045,<br />

e21161<br />

Khushalani, Nikhil I. e19041<br />

Kiatsimkul, Porntip 7534<br />

Kibel, Adam Stuart 4570<br />

Kichenadasse, Ganessan<br />

e19550<br />

Kida, Hiroshi e18056<br />

Kidner, Travis Brad e19029<br />

Kieback, Dirk Guenter 516,<br />

517<br />

Kiechle, Marion 1072, 1081<br />

Kiefer, Amy K 10097<br />

Kiehl, M. e13100^<br />

Kiel, Krystyna D. 1069<br />

Kiel, Nancy e19588<br />

Kiel, Patrick e18555<br />

Kiely, Belinda E. TPS1136,<br />

9087, 9126<br />

Kier, Peter e14087<br />

Kieran, Mark W. 9519<br />

Kies, Merrill S. 5583, 5592<br />

Kiesel, Brian 3049, 3054<br />

Kiesel, Ludwig 10627,<br />

e11534, e21041<br />

Kiessling, Peter 4034<br />

Kieszkowska-Grudny, Anna<br />

e19561<br />

Kiewe, Philipp M. 2007<br />

Kigawa, Junzo 5103<br />

Kigin, Mackenzie 9586<br />

Kii, Takayuki 3088^<br />

Kijima, Takashi e18007,<br />

e18056, e18141<br />

Kikuchi, Eiji 9569<br />

Kikuchi, Eiki 7571<br />

Kikuchi, Ryoko e15579<br />

Kikuchi, Yoshihiro 5037,<br />

e15579<br />

Kikuhara, Yoshihito e21007<br />

Kikuta, Atsushi 9574<br />

Kiladjian, Jean-Jacques<br />

6514^, 6625^, 6626^,<br />

TPS6641, TPS6642^,<br />

TPS6643^<br />

Kilgore, Kelly L. e19528,<br />

e19584<br />

Kilgour, Elaine 4124<br />

Kilic, Diclehan 9050<br />

Kilic, Leyla e14628<br />

Kilickap, Saadettin 3565,<br />

9050, e11083, e16005,<br />

e19575<br />

Killelea, Brigid K. 1030,<br />

1109, 1595<br />

Killian, Keith TPS7609<br />

Killick, Emma 4653<br />

Kilpatrick, Deborah 4526<br />

Kim, Benjamin 6105<br />

Kim, Bong-Seog TPS4142<br />

Kim, Bonglee e13514<br />

Kim, Byung Chul 7053<br />

Kim, Byung Sik 10093<br />

Kim, Chae-yong 2041<br />

Kim, Chan Gyoo 4028<br />

743s


Kim, Chan Kyu e14688<br />

Kim, Chang Won e14566<br />

Kim, Choung-Soo e15080,<br />

e15122<br />

Kim, Da Rae e18092<br />

Kim, Dae Joon e14559<br />

Kim, Dae-Eun e14649<br />

Kim, Dae-Young 6564<br />

Kim, Dong Kwan e14573<br />

Kim, Dong-Sik e19574<br />

Kim, Dong-Wan 3007, 3076,<br />

5552, 7530, 7533, 7543,<br />

7566, 7596, 7599, 7600,<br />

10009, e12029<br />

Kim, Dong-Wook 6503, 6506<br />

Kim, Edward S. 5524, 7023,<br />

7047, 7502<br />

Kim, Eun-Hye 7053<br />

Kim, George P. 4038,<br />

e13052<br />

Kim, Gun Min 3606, e18092<br />

Kim, Gyu IM e14580<br />

Kim, Gyunji 4628<br />

Kim, Hadong 574, e11029<br />

Kim, Hae Rin e14537<br />

Kim, Hak-Hee e11508<br />

Kim, Han Jo e19554<br />

Kim, Han Sang 3606,<br />

e14559, e18080<br />

Kim, Hanjo e14688<br />

Kim, Hark K. e21043<br />

Kim, Harold 5530, 5534,<br />

e16019<br />

Kim, Hee Jeong 578, e11508<br />

Kim, Hee Young e13094<br />

Kim, Heeyoung 9512<br />

Kim, Ho Young e12029<br />

Kim, Ho-Sook 3592<br />

Kim, Hwa Jung 3568<br />

Kim, hye Jin 578<br />

Kim, Hye Ryun 2542,<br />

e14652<br />

Kim, Hyeon Chang e16529<br />

Kim, Hyeong Ryul 4093,<br />

e14573<br />

Kim, Hyeong Ryul 7510<br />

Kim, Hyo Song e12029,<br />

e14652<br />

Kim, Hyo-Jin e14686<br />

Kim, Hyoung-Il e14504,<br />

e14652<br />

Kim, Hyun Jeong e14136<br />

Kim, Hyun Jung e14688,<br />

e19554<br />

Kim, Hyun Ki e14559<br />

Kim, Hyun Sik e14734,<br />

e14736<br />

Kim, Hyun Sue 1017<br />

Kim, Hyun-Jung e13065<br />

Kim, Hyung Lae e15046,<br />

e15203<br />

Kim, Hyunsoo e14164<br />

Kim, Hyunsung e14181<br />

Kim, Isaac Yi e15214<br />

Kim, Ja Young e18501<br />

Kim, Jae Hoon e15503<br />

Kim, Jae Hyun e17523<br />

Kim, Jae Weon e15503<br />

Kim, Jang Hee e18519<br />

Kim, Jee Hyun 1003, 3076<br />

Kim, Jenny J. TPS2613<br />

Kim, Jenny e19570<br />

Kim, Jeong Eun 3568,<br />

e11504<br />

Kim, Jeong-Eun e11508<br />

Kim, Jeong-Oh e18050<br />

Kim, Jeri e15181<br />

Kim, Jhingook 7053, 7510,<br />

e18083, e21062<br />

Kim, Ji sun 1133, e21160<br />

Kim, Ji Yeon 4544^<br />

Kim, Jin Cheon 3568<br />

Kim, Jin Seok 8097,<br />

TPS8118<br />

Kim, Jin Young 7089,<br />

e14570<br />

KIM, Jiyoun 4093, 5586<br />

Kim, Jong Gwang 3554,<br />

6536, 6538, e14074,<br />

e14570, e18533<br />

Kim, Jong Hoon 4093,<br />

e18501<br />

Kim, Jong-Hyeok e15559<br />

Kim, Jong-Mook e19574<br />

Kim, Jongphil 2544, 7065,<br />

10070, e17559<br />

Kim, Joo-Hang 7549, 7557,<br />

7558, e14559, e18092<br />

Kim, Joon-Hyung J. 5587<br />

Kim, Joseph W. 2526,<br />

TPS2617, TPS4701,<br />

10589<br />

Kim, Ju Young e14580<br />

Kim, Julian 1031, 8595<br />

Kim, June e16052<br />

Kim, Jung-AH e16047<br />

Kim, Jung-Young 2074^<br />

Kim, Kenneth H. 5062^,<br />

e13087<br />

Kim, Kevin B. 8503^, 8510,<br />

8514, 8536, 8537, 8551,<br />

e19009, e19014, e19039<br />

Kim, Ki Hyang TPS4142<br />

Kim, Ki Won e14583,<br />

e14656<br />

Kim, Ki-Han e14686<br />

Kim, Ki-Hun 10093<br />

Kim, Kihyun 8097<br />

Kim, Koen Kook e14685<br />

Kim, Ku Sang 578, 2542,<br />

e11559<br />

Kim, Kwang Hyun 5552<br />

Kim, Kwang Pyo e21043<br />

Kim, Kyoung Ha e19554<br />

Kim, Kyoung-Mee 4011,<br />

10535<br />

Kim, Kyoungha e14688<br />

Kim, Kyu-Pyo 3568, 3592,<br />

e11504, e13104, e13560,<br />

e14072<br />

Kim, Kyu-Rae e15559<br />

Kim, Kyung Min e14504<br />

Kim, KyungMann 555, 563,<br />

6063, e13537, e19047<br />

Kim, M. K. e14610<br />

Kim, Mi-Jung e14136<br />

Kim, Michelle e21109<br />

Kim, Min Chan e14686<br />

Kim, Min Kyoung e14570,<br />

e19538<br />

Kim, Miranda 7098, e17534<br />

Kim, Nam Kyu 3606,<br />

e14081, e14084, e14114<br />

Kim, Noe Kyeong 4028<br />

Kim, Philip 4563<br />

Kim, Phillip Sangwook 4011,<br />

10535, 10536, 10537,<br />

e14584, e21017<br />

Kim, Richard D. e14519,<br />

e14715<br />

Kim, Roger Hoon e11541<br />

Kim, Ryan 2052<br />

Kim, Samyong e14529<br />

Kim, Sang Yoon 5586<br />

Kim, Sang-We 7004, 7543,<br />

7549, TPS7614, e13104,<br />

e18052<br />

Kim, Se Hyun e14081,<br />

e14084, e18092<br />

Kim, Se Hyung e14136,<br />

e14688, e19554<br />

Kim, Se Won 1053<br />

Kim, Seok Jin 8023<br />

Kim, Seung Il 2542<br />

Kim, Seung Jin TPS1141<br />

Kim, Sharon 1031, e19580<br />

Kim, Si-Young e14128<br />

Kim, Simon P. 4657, 6126<br />

Kim, Sinil 4503, LBA4537,<br />

4546, 7551<br />

Kim, So Yeon 1577<br />

Kim, Sook-Young 7089<br />

Kim, Stefano Chong Hun<br />

e14007<br />

Kim, Sun Hyun e19613<br />

Kim, Sun Young 3531, 3587,<br />

e14072<br />

Kim, Sung Hoon e13514<br />

Kim, Sung Hyun 4028<br />

Kim, Sung 4011<br />

Kim, Sung-Bae 578, 1003,<br />

2542, 4093, 5586,<br />

e11504, e11508, e13104<br />

Kim, Sung-Doo 6564<br />

Kim, Sung-Hyun e14686,<br />

e18083, e21062<br />

Kim, Sung-Won 1056<br />

Kim, Sungjin 5560, 7059,<br />

7097, e14614<br />

Kim, Sunyoung e19574<br />

Kim, Tae IL e14081, e14084<br />

Kim, Tae Min 2041, 3076,<br />

4544^, 5552, 7566,<br />

e12029<br />

Kim, Tae Soo 3606<br />

Kim, Tae Won 3531, 3532,<br />

3568, 3587, 3592,<br />

e13104, e13560, e14072<br />

Kim, Tae Yong e14580<br />

Kim, Tae-Hoon 574, e11029<br />

Kim, Tae-Yong e14136<br />

Kim, Tae-You 3076, 3531,<br />

3587, 3624, e11532,<br />

e14136, e14580, e21160<br />

Kim, Won Seog 8023, 8029,<br />

e18528<br />

Kim, Woo Ho 4124<br />

Kim, Yeong 1538<br />

Kim, Yeul Hong e14580<br />

Kim, Yong Hee 4093<br />

Kim, Yong-Hee e14573<br />

Kim, Yong-Man e13104<br />

Kim, Yookyung e16500<br />

Kim, Young Ae 9066<br />

Kim, Young Hwan e21043<br />

Kim, Young Woo 4028<br />

Kim, Young-Hoon 2041<br />

Kim, Yu Jung 2041, 3076<br />

Kim, Yu Min e14504<br />

Kim, YuJung 4544^<br />

Kimball, Amy Sarah 8075<br />

Kimberg, Cara I. 9531<br />

Kimbler, Kimberly D. e21000<br />

Kimbro, Lisa e16510<br />

Kimler, Bruce F. 520, 9000<br />

Kimlin, Michael e19550<br />

Kimmick, Gretchen Genevieve<br />

6015, 6052<br />

Kimmig, Rainer 5042<br />

Kimura, Eizo 5003<br />

Kimura, Hideharu TPS659<br />

Kimura, Hiroaki 2044,<br />

10072, 10075, e20505<br />

Kimura, Takayuki e13511<br />

Kimura, Tatsuo 7003,<br />

e17538<br />

Kimura, Yasunori e15055,<br />

e15099<br />

Kinarsky, Yulia 1538<br />

Kinders, Robert J. 3031,<br />

3087, 3529<br />

Kindler, Hedy Lee 2561,<br />

3105, 4022, 4047, 7029,<br />

7030, 7077<br />

Kindler, Manfred e15044<br />

Kindler, Thomas 6511<br />

King, Dennis e21066<br />

King, Erin Rebecca e15583<br />

King, Gentry T 536<br />

King, Karen 10532<br />

King, Madeleine TPS5116,<br />

6002, 6111, 7607,<br />

e16507<br />

King, Robin 1520<br />

King, Stephanie e15522<br />

King, Tari A. 10618<br />

King, Thomas 6530<br />

Kingham, T. Peter 3577,<br />

3620<br />

Kingsley, Edwin e18558<br />

Kingswood, Christopher 4632,<br />

10619<br />

Kinhult, Sara 10024<br />

Kink, John A. TPS3113<br />

Kinoshita, Hidefumi e15071<br />

Kinoshita, Ichiro 7571,<br />

e17520, e18135<br />

Kinoshita, Rumiko e15533<br />

Kinoshita, Takayuki TPS1147<br />

Kinoshita, Tomohiro 8050<br />

Kintzinger, Clement 2048<br />

Kinugasa, Yusuke 3569,<br />

3607<br />

Kinzig, Martina 3044<br />

Kioi, Mitomu 5556<br />

Kipps, Thomas J. 6122, 8032<br />

Kira, Yukimi e17538<br />

Kirby, Chaim 9583<br />

Kirchh<strong>of</strong>f, Tomas 8557<br />

Kirchner, Sandra 9019<br />

Kirchner, Thomas 3519^,<br />

4041, 10054<br />

Kirichenko, Alexander V.<br />

e21036<br />

Kirita, Keisuke e17526<br />

Kirk, Dylan T e18549<br />

Kirkbride, Peter 4509<br />

Kirkpatrick, John 2090^,<br />

2094^<br />

Kirkpatrick, Nathaniel D.<br />

2010<br />

Kirkwood, John M. 8501,<br />

8504, 8522, 8527, 8528,<br />

8533, 8547, 8559,<br />

TPS8606<br />

Kirn, David H. TPS4152,<br />

e13044, e14566<br />

Kirsa, Sue e16538<br />

Kirschbaum, Mark H. 6582<br />

Kirshner, Jeffrey J. 9141,<br />

9142, e19510<br />

Kish, Julie Ann 5564, 6100<br />

Kishi, Hiroto e13072<br />

Kishi, Kazuma 7012<br />

Kishimoto, Hiroyuki e14014<br />

Kishimoto, Junji 7045,<br />

e13014<br />

Kishimoto, Takashi K 4056<br />

Kishine, Kennji e14004<br />

Kishino, Daizo 7042<br />

Kiss, Alex e19545<br />

Kissane, David W 9121<br />

Kistangari, Gaurav e15023<br />

Kit, Oleg I e11558, e20503,<br />

e21101<br />

744s Numerals refer to abstract number


Kita, Aya e21059<br />

Kita, Tsunekazu 5037,<br />

e15579<br />

Kitada, Koji 4012<br />

Kitagawa, Ryo 5006<br />

Kitagawa, Yuko 10531,<br />

e11060, e14045, e14080<br />

Kitaichi, Masanori e18126<br />

Kitajima, Masaki 1106<br />

Kitajima, Masayuki e14004<br />

Kitamura, Hideya e19583<br />

Kitano, Atsuko 590<br />

Kitano, Seigo 3569<br />

Kitano, Shigehisa 2518<br />

Kitao, Akihito 5045, e19548<br />

Kitayama, Joji e14142,<br />

e14530<br />

Kitsios, Petros e15030<br />

Kitsukawa, Shinichi e15038<br />

Kittas, Christos e21098<br />

Kittel, Kornelia e19540<br />

Kitto, Alex e18549<br />

Kitzweger, Elvira e14087<br />

Kiura, Katsuyuki 2607, 7042,<br />

7045, 7560, 7576, 7602,<br />

e18027<br />

Kivett, Valerie A 2026<br />

Kiyici, Aysel e11008<br />

Kiyohara, Meagan 1052<br />

Kiyota, Hidemi e18060<br />

Kiyota, Naomi 5542, e16034,<br />

e17020<br />

Kizilarslanoglu, Cemal<br />

e11080, e11509<br />

Kizilbash, Sani Haider<br />

e14608<br />

Kjaer-Frifeldt, Sanne 3513<br />

Kjellén, Elisabeth 10024<br />

Kjellman, Anders 4508<br />

Klaase, Joost TPS10632<br />

Klaassen, Robert 6103<br />

Klaisuban, Wipawi e14051<br />

Klamt, Fábio e21042<br />

Klapman, Jason 4055<br />

Klappers, Petra 6500^<br />

Klare, Peter 5031, e15520,<br />

e19540<br />

Klasen, Hermann A. 2007<br />

Klassen, Anne 9042, 9043<br />

Klautke, Gunther 7001<br />

Klee, George G. 4565<br />

Kleha, Joseph Francis<br />

e13596<br />

Klein Connolly, Hannah<br />

e11096<br />

Klein, Eric A. 4560<br />

Klein, Pamela 567<br />

Klein, Paula e11057<br />

Klein, Peter e11040^<br />

Klein, Stefan 3588, 4023<br />

Kleinberg, Lawrence 2013,<br />

2017<br />

Kleinfield, Robert 2597<br />

Kleinman, Shlomi 1564<br />

Klem, Robert E. e15113<br />

Klemp, Jennifer R. 9000<br />

Klener, Pavel e18539<br />

Klepin, Heidi D. 6015, 9034,<br />

9109, 9123, e18133<br />

Kleski, Amy 9115<br />

Klevesath, Manfred B 3107^<br />

Kliesch, Sabine 4530<br />

Klimberg, V. Suzanne e19563<br />

Klimovsky, Judith 4014,<br />

4632, e14621<br />

Klimowicz, Alexander C.<br />

2001, e21083<br />

Klimstra, David 10524<br />

Kline, Justin Paul 8075<br />

Numerals refer to abstract number<br />

Kline, Zoe e18077<br />

Klingbiel, Dirk 3595,<br />

e19648, 9045<br />

Klingebiel, Thomas 9573<br />

Klinghammer, Konrad<br />

Friedrich 5572<br />

Klingler, Christoph e15090<br />

Klingler, Sophie 10517<br />

Klinkhammer-Schalke, Monika<br />

e16520<br />

Kloecker, Goetz H. e16517,<br />

e16533, e16545, e21072,<br />

e21149<br />

Klos, Dusan e14556<br />

Kloss, Susanne e15044<br />

Kloth, Jacqueline S.L. 2596,<br />

10091<br />

Klouvas, George 5033<br />

Kluetz, Paul Gustav 4569<br />

Klug, Evelyn 542<br />

Kluger, Harriet M. 7514,<br />

8531<br />

Klughammer, Barbara 10596<br />

Klumpen, Heinz-Josef<br />

TPS4148<br />

Klumpp, Bernhard 8526<br />

Kluschke, Franziska 5064,<br />

e19558<br />

Knackmuss, Stefan 8059,<br />

e18532<br />

Knapen, Marie Claire 9035<br />

Kneebone, Andrew TPS4690<br />

Knezevic, Dejan 4560<br />

Knigge, Ulrich 4015<br />

Knight, Alastair D. e19544,<br />

e19567, e19578<br />

Knight, Julie A 1502<br />

Knight, Robert D. 607,<br />

e15170, e18559<br />

Knight, Simon e17539<br />

Knight, Tyler e18559<br />

Knobler, Robert 8076<br />

Knoedler, Maren 5572<br />

Knoefel, Wolfram Trudo<br />

e14527, e21020<br />

Knol, Anne-Chantal e19019<br />

Knol, Jaco C e18094<br />

Knoops, Laurent 6514^,<br />

6625^, 6626^<br />

Knop, Stefan 8095<br />

Knopf, Kevin B. 6553,<br />

e13081<br />

Knott, Adam 597^, 598^,<br />

e11055^<br />

Knox, Chetaye CRA9013<br />

Knox, Jennifer J. 4099,<br />

4109, 4536, 4538,<br />

TPS4686, 10522, e14524,<br />

e14592<br />

Knudsen, Kraig J. 1598<br />

Knusli, Claudio 10033<br />

Knuth, Alexander 2573^<br />

Knutsen, Raymond 1587<br />

Ko, Andrew H. 3105, 4038,<br />

4048, 4102<br />

Ko, Clifford Y. 3545, 3608,<br />

6057<br />

Ko, Emily Meichun 5009,<br />

e15511<br />

Ko, Emily 5092<br />

Ko, Jennifer TPS648<br />

Ko, Jenny J 6014<br />

Ko, Ryo e18019<br />

Ko, Sanghwa e14121,<br />

e14181<br />

Ko, Seung Sang 1053<br />

Ko, Wan-Seng e11561<br />

Ko, Yon-Dschun 1533,<br />

10526, CRA10529<br />

Ko, Yoo-Joung 4522, 4525,<br />

e14171, e15177^<br />

Ko, Yoon-Ho e14634<br />

Koay, Eugene Jon 8584<br />

Koba, Hayato e17540<br />

Kobayashi, Kazuma 3558<br />

Kobayashi, Kunihiko 7515,<br />

7561, 7563, e18129<br />

Kobayashi, Masashi e18036,<br />

e18042<br />

Kobayashi, Nozomi 10578<br />

Kobayashi, Susumu 7523<br />

Kobayashi, Takao e19521<br />

Kobayashi, Toshimitsu<br />

e16020<br />

Kobayashi, Yasuyuki e15068<br />

Kobayashi, Yukio 8050<br />

Kobe, Carsten 3044, 7603<br />

Kobs, Christina L. 9575<br />

Kobyakov, Grigory 2034<br />

Koc, Basak 9567<br />

Koca, Dogan 638<br />

Kocak, Ivo TPS4696^<br />

Kocak, Necmettin 6550<br />

Kocáková, Ilona 3539<br />

Kocaman, Gokhan 1596<br />

Kocaturk, Celalettin 7024<br />

Kocer, Murat 638<br />

Koch, Michael O. 4586<br />

Koch, Sven D. 2573^<br />

Kocherginsky, Masha 5500<br />

Kockx, Mark 8503^<br />

Kocs, Darren M. 1017<br />

Koczwara, Bogda e19550,<br />

e19553<br />

Koczywas, Marianna 3108,<br />

7029, 7518, 9068,<br />

e18018<br />

Kodaira, Makoto 10519,<br />

e19535<br />

Kodaira, Takeshi e14500,<br />

e15533<br />

Kodali, Swati e16060<br />

Kodama, Kotaro e13511<br />

Kodama, Shoji 5003<br />

Kodama, Tetsuro 7021<br />

Kodera, Yasuhiro 3558<br />

Koeberle, Dieter 3595,<br />

e19648, 9045, 10033,<br />

e14544<br />

Koechlin, Alice 1509, 1510<br />

Koehler, Astrid 8503^<br />

Koehler-Santos, Patricia 1522<br />

Koelbl, Heinz 10551<br />

Koeller, Jim M. e16522<br />

Koenig, Kimberly<br />

Higginbotham 527<br />

Koenig, Nelly e18505<br />

Koenig, Ronald J 6056<br />

Koenigsberg, Tova 1126<br />

Koenigsmann, Michael 4083<br />

Koepfgen, Kathryn M. e15137<br />

Koepl, Lisel 6007, e14068<br />

Koepnick, Sven 3550<br />

Koeppel, Marie-Christine<br />

8555<br />

Koeppen, Susanne 2086<br />

Koerber, Wolfgang 10600<br />

Koestler, Wolfgang 607<br />

K<strong>of</strong>ahl-Krause, Dorothea 3<br />

Koga, Hir<strong>of</strong>umi e15127<br />

Koga, Rintaro e14506<br />

Kogan, Mikhail 4501<br />

Kogler, Ulrika 1594<br />

Kogut, Neil Martin 6545<br />

Koh, Sung Ae e19538<br />

Koh, Tong San 1064, 4100^<br />

Koh, Wee Yao 2016, 5553<br />

Koh, Yasuhiro e21007<br />

Koh, Yin Ling e18546<br />

Koh, Youngil 4544^, 5552<br />

Kohen, Nelson e15507<br />

Kohler, David R. 3043<br />

Kohler, Racquel Elizabeth<br />

6017, 6029<br />

Kohli, Antia e13042<br />

Kohli, Manish e15132<br />

Kohlmann, Wendy 9525,<br />

9561<br />

Kohn, Elise C. 2545<br />

Kohne, Claus-Henning 3562,<br />

3591<br />

Kohno, Mikihiro e13090<br />

Kohno, Norio 635, TPS1141<br />

Kohno, Shigeru 7569<br />

Kohnoe, Shunji e14033<br />

Kohrman, Michael 1609,<br />

e12042, e15096<br />

Kohrogi, Hirotsugu e13072,<br />

e18059<br />

Kohrt, Holbrook Edwin 2043,<br />

2513, 2514, e13555,<br />

e21071<br />

Koie, Takuya e15068<br />

Koike, Teruaki 7012<br />

Koike, Yukihiro e14622<br />

Koivula, Satu 8074<br />

Koizumi, Fumiaki 10519,<br />

e11065<br />

Koizumi, Tatsuhiko e19521<br />

Koizumi, Toshiyuki 5556<br />

Koizumi, Wasaburo 4026,<br />

4088, e14510<br />

Kojima, Hiroshi 8094<br />

Kojima, Hiroyuki e14500<br />

Kojima, Tetsuya e18135<br />

Kojouhar<strong>of</strong>f, Georgi 4023<br />

Kok, Victor C. e11561<br />

Kokorsch, Philipp e15577<br />

Kokudo, Norihiro 4104<br />

Kokx, Patricia 6054<br />

Kolar, Zdenek 1087<br />

Kolarevic, Daniela e21030<br />

Kolatkar, Anand e21074<br />

Kolb, Brigitte 8009, 8014<br />

Kolek, Vitezslav e18062<br />

Kolesar, Jill 3101, 3103,<br />

7516<br />

Kolesnick, Richard 6539<br />

Kolev, Nikola e21100<br />

Kolhe, Ravindra 6573, 8108<br />

Kolitz, Jonathan E. 2533,<br />

6525, 6526, 6601<br />

Kollar, Attila 3595<br />

Kollender, Yehuda 10043<br />

Koller, Josef 8505<br />

Koller, Michael 6090,<br />

e16520<br />

Kollia, Georgia CRA2509,<br />

TPS2622, 4505, 7509,<br />

8507<br />

Kolligs, Frank T 4006,<br />

e14654<br />

Kollmannsberger, Christian K.<br />

TPS2614, 3039, 4536,<br />

4538, e14537, e14568<br />

Kollmeier, Jens 7001, 7575<br />

Kollmeyer, Thomas M. 4565<br />

Kolman, Petr 7512<br />

Kolodziej, Ilka TPS5116<br />

Kolodziej, Michael A. 6109<br />

Kolodzieyski, Lev 3086<br />

Kolostova, Katarina e21051<br />

Komaki, Ritsuko e14548,<br />

e14669<br />

Komarnitsky, Philip B 7090<br />

Komarova, Ekaterina e19049<br />

Komatsu, Shigeru e19583<br />

745s


Komatsu, Yoshihiro e14689<br />

Komatsu, Yoshito 3593,<br />

e14062<br />

Komeno, Takuya 8094<br />

Kometani, Takuro 3045<br />

Kommoss, Friedrich 5005<br />

Kommoss, Stefan 5005<br />

Komori, Takahide e16034<br />

Komrokji, Rami S. 3081,<br />

6568, 6575, 6586, 6588,<br />

6608<br />

Komuta, Kiyoshi e18007,<br />

e18056, e18141<br />

Kondadasula, Sri Vidya<br />

e13089<br />

Kondalsamy-Chennakesavan,<br />

Srinivas e15541<br />

Kondi-Pafiti, Agathi e13556<br />

Kondo, Chiaki e17510,<br />

e18145<br />

Kondo, Nobuyuki 7080,<br />

10585<br />

Kondo, Satoshi 9577<br />

Kondo, Shinya e14540<br />

Kondo, Tsunenori e15068,<br />

e15071<br />

Kond<strong>of</strong>f, Matt R 1608<br />

Kondoh, Chihiro e14500<br />

Kondziolka, Douglas 8528<br />

Konety, Badrinath e15211<br />

Kong, Anthony 530<br />

Kong, Feng-Ming (Spring)<br />

7037, e17530, e21115<br />

Kong, Li-Ren 3002<br />

Kong, Ling-Yuan 2011<br />

Kong, Sun-Young 4028<br />

Kong, Weidong 1583<br />

Kong, Yan 8561, 8562<br />

Kong, Yana e11091<br />

Konias, Marina e19540<br />

Konicek, Bruce W 8082<br />

Konigsberg, Robert 514<br />

Konishi, Fumio 3569<br />

Konishi, Tsuyoshi 3625<br />

Konkankit, Veerauo V 8592<br />

Konkashbaev, Anuar 9516<br />

Konno, Satoshi e18135<br />

Kono, Hidaka 9569<br />

Kono, Scott A. 5560, 5570,<br />

e21045<br />

Kono, Scott Arthur 7048<br />

Konopleva, Marina 6558,<br />

6576, 6585, TPS6635<br />

Konopski, Zbigniew e14637<br />

Konski, Andre A. e17528<br />

Konstantinopoulos, Panagiotis<br />

8528<br />

Konto, Cyril 8512, 8539,<br />

TPS8603<br />

Kontopodis, Emmanouil<br />

e18105<br />

Kontoyiannis, Dimitrios P<br />

e17016<br />

Koo, Yang Seo e14685<br />

Kooby, David A. e14692<br />

Koock, Ulrike 4083<br />

Koolen, Stijn 2550<br />

Koon, Henry B. 8595<br />

Koong, Albert 4045<br />

Koornstra, Rutger H.T. 562<br />

Kopcewicz, Katarzyna 3055<br />

Kopetz, Scott 3528, 3544,<br />

3556, 3561, 3598,<br />

e14071<br />

Kopit, Justin TPS8112,<br />

TPS8113, TPS8114<br />

Kopp, Joachim e15053<br />

Kopp, Lisa M. 9503<br />

Koppe, Fredirike 7019<br />

Koraa, Emad e17556<br />

Korc-Grodzicki, Beatriz 5108<br />

Korcum, Aylin Fidan e21130<br />

Korczak, Bozena 9119<br />

Korde, Neha 8088, 8104,<br />

e18568<br />

Kordes, Sil 4094<br />

Korfel, Agnieszka 2007,<br />

2040, 2054, 8031<br />

Koritzinsky, Marianne e15118<br />

Korman, Alan CRA2509<br />

Korn, Michael 3006^<br />

Korn, Wolfgang Michael 4102<br />

Kornacker, Martin 3012,<br />

3046<br />

Kornblith, Alice B. 6015<br />

Korogi, Yohei e18025,<br />

e18134<br />

Koroukian, Siran M. 1598<br />

Korphaisarn, Krittiya e14051<br />

Korte, Susan H. 5514, 5545^<br />

Korth, Christopher Charles<br />

e13040<br />

Kortylewski, Marcin e15178<br />

Koru-Sengul, Tulay 1572<br />

Korves, Caroline 10061<br />

Korytowsky, Beata 4542,<br />

6092, e15061, e16530<br />

Kos, Fahriye Tugba e13031<br />

Kosaka, Takashi e14624<br />

Koscielniak, Ewa 9573<br />

Koscielny, Serge 4618,<br />

e13118<br />

Kose, Kenan e20000<br />

Kosel, Matthew L. e13086<br />

Kosela, Hanna 10538<br />

Koshikawa, Kayoko e13037<br />

Koshiol, Jill e18128<br />

Koshrawipour, Tanja 10059<br />

Kosik, Kelly e13121<br />

Koski, Anniina e13035<br />

Kosl<strong>of</strong>f, Rebecca A. 7587<br />

Kosmider, Suzanne e16516<br />

Kosovec, Juliann e14689<br />

Koss, Juliane 1084<br />

Koss<strong>of</strong>f, Ellen e15042,<br />

e19568<br />

Kostakis, Ioannis e21098<br />

Kostakoglu, Lale 5578,<br />

e13592, e21006<br />

Kostashuk, Edward 4509<br />

Kosten, Scott e14501<br />

Kostjakova, Ljudmila e15029<br />

Kostlevy, Jordan e19047<br />

Kostouros, Efthimios e15040<br />

Kosuge, Tomoo e14506<br />

Kosugi, Hiroshi 8097<br />

Kosztyla, Daniel 9573<br />

Kota, Vamsi 6573, 8108<br />

Kotani, Yoshikazu 7003,<br />

e18134<br />

Kotapati, Srividya e19010<br />

Kotasek, Dusan e19511<br />

Kotchko, Nancy 1608<br />

Kote-Jarai, Zs<strong>of</strong>ia 1545, 4653<br />

Kotecki, Nuria 2070, 10035,<br />

e11049<br />

Kotkova, Tatjana N. e11086<br />

Kotoula, Vassiliki 573, 592,<br />

10575<br />

Kotowski, Adam S. e14659<br />

Kotsakis, Athanasios<br />

Panayotis 5541, e18105<br />

Kottschade, Lisa A. 1557,<br />

8600<br />

Kotzerke, Joerg 10068<br />

Kouakam, Claude e11049<br />

Koubkova, Leona e18062<br />

Kouchner, Pierre e11551<br />

Koudelakova, Vladimira 1087<br />

Koufopoulos, Nektarios<br />

e11519<br />

Koumakis, George e11519<br />

Koumarianou, Anna e21027<br />

Koussis, Haralabos 5513,<br />

e12037<br />

Koustenis, Andrew 7002<br />

Kouta, Hiroko 5037, e15579<br />

Koutcher, Lawrence 5537<br />

Koutras, Angelos 573<br />

Kouwenhoven, Mathilde C.M.<br />

2<br />

Kovac, Suzana e15117<br />

Kovach, Barbara 10596<br />

Kovacsovics, Tibor 6601<br />

Koval, Greg 6582<br />

Kovalenko, Nadezhda<br />

TPS651, 5017^<br />

Kovel, Svetlana 4564, 4619,<br />

e15058<br />

Kowal, Monika e21049<br />

Kowalczyk, Anna 603<br />

Kowalska, Malgorzata e21049<br />

Kowalski, Dariusz M e12011<br />

Kowalski, Luiz Paulo e16046<br />

Kowalsky, Richard J. 5050<br />

Kowalyszyn, Ruben Dario<br />

e14521<br />

Koya, Richard C. e13045<br />

Koyama, Hiroki 1103<br />

Koyanagi, Nozomu 3084<br />

Kozl<strong>of</strong>f, Mark 3543, 3552,<br />

TPS3637, 6122<br />

Kozlowski, Piotr 6584<br />

Kozma, Sara TPS3116<br />

Kozuki, Toshiyuki 7576,<br />

e18099<br />

Kozyreva, Olga N. e15123<br />

Kraan, Jaco 10504<br />

Krabisch, Petra 552<br />

Kracht, Karolyn 4606<br />

Kraemer, Dale F. 2040<br />

Kraemer, Sandrine 4071<br />

Kraetschell, Robert W. 5087<br />

Kraft, Andrew S. 10003<br />

Kraft, Walter K e16548<br />

Krahn, Thomas 10536,<br />

10537, e21017, e21020<br />

Krailo, Mark D. 9503, 9537,<br />

9539, 10081<br />

Krajcsi, Peter e21140<br />

Krakobsky, Vanesa e11555<br />

Krakowski, Ivan LBA4567^,<br />

e19640<br />

Kramar, Andrew 2540,<br />

e11070<br />

Kramer, Daniel 7090<br />

Kramer, Gero e15090<br />

Kramer, Melissa 4562<br />

Kranenburg, Britte 8018^<br />

Kranenburg, Tanya 9518<br />

Krasin, Matthew J. 9502<br />

Krasna, Mark 1540<br />

Krasner, Carolyn N. 5029<br />

Krasnik, MARK 7026<br />

Krasnozhon, Dmitriy e19609<br />

Kratz, Christian 9521<br />

Kraus, Dennis 5537, 5545^<br />

Kraus, Dietmar e17536<br />

Kraus, Sarah 1507, 1564<br />

Krause, Stefan W. 6510<br />

Krauss, John C. 8007<br />

Krauss, Juergen 5516^<br />

Krauze, Michal Tadeusz 8528<br />

Krawczyk, Natalia 5042,<br />

e21003<br />

Kreahling, Jenny 10084<br />

Krebs, Matthew 3004, 3030,<br />

3048<br />

Krege, Susanne 4524, 4530<br />

Kreienberg, Rolf 1078, 1082,<br />

e11040^<br />

Kreilick, Charles e14106,<br />

e14141, e14146, e15183,<br />

e18097<br />

Kreipe, Hans Heinrich 552<br />

Kreischer, Nathan e13601<br />

Kreissl, Michael 5508<br />

Kreitman, Robert J. 2503^<br />

Krekow, Lea 547<br />

Krell, Jonathan e21137<br />

Krell, Robert Wallace 6020<br />

Kremmidiotis, Gabriel 4603,<br />

7079^<br />

Krenn, Markus e14103<br />

Krestin, Gabriel P. 10091<br />

Kreutz, Werner e13508<br />

Kreys, Eugene e16522<br />

Kridel, Steven 6524^<br />

Kriegel, Irene 1580<br />

Krikelis, Dimitrios 10086,<br />

10575<br />

Krikov, Maxim e14152<br />

Kris, Mark G. 3083, 7014,<br />

7052, 7066, 7505, 7527,<br />

7530, 7532, 7580, 7589,<br />

10560, e19647<br />

Krishna, Sai Sakktee 5551,<br />

5580<br />

Krishnamoorthy, Saravanan<br />

e13064<br />

Krishnamurthy, Savitri<br />

TPS10631, e13582<br />

Krishnamurthy, Vikram<br />

e19061<br />

Krishnan, Amrita Y. 6547,<br />

8038, 8089, e18542<br />

Krishnan, Monica Shalini<br />

6022<br />

Krishnan, Rajaram e21047<br />

Krishnan, S. 3089^<br />

Krishnan, Shobha e12009<br />

Krishnan, Sunil 4116<br />

Krishnasamy, Meinir e19605<br />

Krishnatry, Rahul e16021<br />

Krissel, Heiko 4103<br />

Krist, Alex 1563<br />

Kristensen, Gunnar<br />

LBA5002^, 5008<br />

Kristjansdottir, Kolbrun 9584,<br />

e20008<br />

Kristopaitis, Theresa e19588<br />

Kristopher, Attwood e15039<br />

Krivak, Thomas C. e15560,<br />

e15578<br />

Krivokuca, Ana e12020<br />

Krizman, David e21068<br />

Krocker, Jutta 624<br />

Kroeger, Nils e13045<br />

Kroep, Judith R. 10006<br />

Kroepil, Feride e14527<br />

Krogh, Merete 8545<br />

Krogsgaard, Michelle 8589<br />

Kroiss, Regina 1537<br />

Krol, Augustinus 8039<br />

Kroll, Stew TPS10100<br />

Kroll, Stewart 6585<br />

Kroll, Torsten 10600<br />

Kronborg, Camilla J S<br />

e14063, e14105<br />

Kronenwett, Ralf 522, 523,<br />

542<br />

Kroog, Glenn Scott 8020,<br />

TPS8112, TPS8114<br />

Krop, Ian E. 508, TPS663,<br />

1026<br />

746s Numerals refer to abstract number


Krop, Ian E LBA1, 528, 532,<br />

10558<br />

Kros, Johan M 2<br />

Kroschinsky, Frank 10068<br />

Krouse, Robert S. e19563<br />

Krucker, Thomas 9562<br />

Kruczek, Kimberly R. 8072<br />

Krueger, Colin 4095^<br />

Krueger, Darcy A. 9541<br />

Kruer, James A e15179<br />

Krug, Lee M. 1541, 7014,<br />

7029, 7052, 7527,<br />

e17558<br />

Krug, Utz 6610<br />

Kruger, Darcy 1609<br />

Krukemeyer, Manfred Georg<br />

e13091<br />

Krull, Kevin R. 9531, 9532<br />

Kruper, Laura 1035, 1117<br />

Kruse, Edward J. e14694<br />

Kruse, Janice 6108<br />

Kryukov, Gregory 10502<br />

Krzakowski, Maciej Jerzy<br />

LBA7000, 7090, 7536,<br />

7575, e12011<br />

Krzyzanowska, Monika K.<br />

2572, 3632, 6087, 9003,<br />

9131, e14003, e14524,<br />

e14592<br />

Kröning, Hendrik 4065,<br />

e14140, e14152<br />

Ku, Yonson 4104<br />

Kuang, Peng 10527<br />

Kuang, Qin 1535, 7586<br />

Kuang, Yukang e18013<br />

Kuba, Maria Gabriela 510,<br />

1024<br />

Kubal, Timothy Edward<br />

e18503, e18506<br />

Kube, Stefanie 9573<br />

Kube, Ulrich e15044<br />

Kubicka, Stefan CRA3503<br />

Kubin, Thomas 4072<br />

Kubo, Akihito 7000, 10577,<br />

e18126, e18128<br />

Kubo, Kazuyuki 613<br />

Kubo, Michiaki 10520<br />

Kubo, Yoshiro 3524<br />

Kubota, Akira 5542<br />

Kubota, Kaoru 7028, 7515,<br />

7549<br />

Kubota, Yoshihiro e11564<br />

Kubota, Yoshinobu 4626<br />

Kucera, Stephen e14714<br />

Kuchimanchi, Mita 5072<br />

Kuchroo, Vijay 8571<br />

Kucuk, Omer 4656, 5534,<br />

e15006, e16019, e20000<br />

Kucuk, Ozlem e21006<br />

Kucukoner, Mehmet 638,<br />

e14526<br />

Kucukoztas, Nadire e11011,<br />

e11023, e11026, e11045,<br />

e11515, e14174, e15572,<br />

e15573<br />

Kuczyk, Markus 4530,<br />

e15195<br />

Kuda, Tomoya 7571, e19521<br />

Kudaka, Wataru 5086<br />

Kudchadkar, Ragini Reiney<br />

3102, 8510, 8582, 8587<br />

Kuderer, Nicole Maria 6129,<br />

9135<br />

Kudo, Daisuke 8094<br />

Kudo, Keita 10569, 10604,<br />

e18004, e18014, e18017<br />

Kudo, Kenichiro 7576<br />

Kudo, Masatoshi 4033, 4104,<br />

e14617<br />

Numerals refer to abstract number<br />

Kudo, Mineo 3593<br />

Kudo, Toshihiro e18060<br />

Kudoh, Kazuya 5037,<br />

e15579<br />

Kudoh, Shinzoh 7521,<br />

e17538<br />

Kudoh, Shoji 7515<br />

Kudrik, Fred J. 8567<br />

Kudrimoti, Mahesh R.<br />

e16035<br />

Kuebler, J. Phillip LBA506<br />

Kuehne, Reto 3595<br />

Kuemmel, Sherko 556, 624<br />

Kuemmerlen, Verena e18010<br />

Kuenzel, Heike TPS7617<br />

Kuerer, Henry Mark 1028,<br />

1102, 1130, 1549,<br />

TPS10631<br />

Kufer, Peter 2504<br />

Kugler, John William 7073<br />

Kuhn, Christina e15529<br />

Kuhn, Peter 4102, e21074<br />

Kukla, Andrea K. 5544<br />

Kukreti, Vishal 8035<br />

Kukura, Vlastimir e12505<br />

Kulakodlu, Mahesh e14013,<br />

e14016<br />

Kulanthaivel, Palaniappan<br />

3001<br />

Kulig, Kimary 2021<br />

Kulikov, Evgeny TPS4696^<br />

Kulita, Patrycja e19561<br />

Kulkarni, Raghav e16042<br />

Kulkarni, Sujay 3011, 3013<br />

Kulkarny, Vibhati 1558<br />

Kulke, Matthew 3594, 4085,<br />

4125, 4126, e14542,<br />

e14551<br />

Kuller, Anne 4011<br />

Kuller, Lewis H 1501<br />

Kullmann, Frank TPS3641^,<br />

4072<br />

Kullmann, Verena 5051<br />

Kulma-Kreft, Monika e21099<br />

Kumagai, Toru e18025<br />

Kumaki, Nobue 565<br />

Kumanogoh, Atsushi e18007,<br />

e18141<br />

Kumar, Aalok 3527, e14568<br />

Kumar, Chandan 4591<br />

Kumar, K. Sunesh 5104<br />

Kumar, Kirushna 9117<br />

Kumar, Lalit 5104, 6593,<br />

e15510, e18518, e18573<br />

Kumar, Mukesh 8101<br />

Kumar, Nagi B. e21155<br />

Kumar, Pankaj 5540<br />

Kumar, Parag 2591, 5050,<br />

e13063, e13074<br />

Kumar, Pavan e19055<br />

Kumar, Priya 5500, 5544<br />

Kumar, Rachit e19025<br />

Kumar, Sanjeev e19516<br />

Kumar, Satish TPS8605<br />

Kumar, Shaji 8033, 8034,<br />

8092, 8095, 8096, 8105,<br />

TPS8116, e13517<br />

Kumar, Sunesh e15518<br />

Kumatani, Youji 1103<br />

Kumbhaj, Prashant e16012<br />

Kume, Makoto e14518<br />

Kummar, Shivaani 3031,<br />

3087, 3529, 5020, 8073<br />

Kumosa, Lucas e21047<br />

Kun, Larry E. 9531<br />

Kundel, Yulia e14067,<br />

e14571<br />

Kung, Andrew L. e13586<br />

Kung, Ling e16523<br />

Kunieda, Katsuyuki 3607,<br />

TPS3642<br />

Kunimasa, Kei 7528,<br />

e18025, e18134<br />

Kunisaki, Chikara 4070,<br />

e14624<br />

Kunitoh, Hideo 7012, 7515<br />

Kuniyoshi, Renata Kelly<br />

e11059<br />

Kunju, Priya 4591<br />

Kunkel, Lori A. 8035<br />

Kunnavakkam, Rangesh<br />

2561, 6582<br />

Kunnavakkum, Rangesh 6562<br />

Kunos, Charles 5089, 5102<br />

Kunst, Peter W.E. 7019<br />

Kunst, Tricia 7103<br />

Kuntz, Karen M 6016<br />

Kunz, Claudia 2034<br />

Kunzmann, Volker 4072,<br />

e13100^<br />

Kuo, Han Pin TPS7614<br />

Kuo, Kevin F 4557<br />

Kuo, R-Y e14699<br />

Kuo, Wen-Hung 1043<br />

Kuom, Sabine 7572<br />

Kupershmidt, Ilya e21159<br />

Kupersztoch, Yankel M<br />

e16500<br />

Kupets, Rachel 6134<br />

Kupka, Markus 554<br />

Kurachi, Yoshitaka TPS3642<br />

Kurahashi, Issei 4012<br />

Kuramochi, Hidekazu e21108<br />

Kuraoka, Kazuya 5084<br />

Kurata, Takayasu 7000,<br />

e18060, e18128<br />

Kurbacher, Christian M.<br />

e13100^<br />

Kurdziel, Karen A 10589,<br />

e18568<br />

Kure, Elin 3548<br />

Kurek, Ralf e15191<br />

Kurhanewicz, John V. 4660<br />

Kurian, Seira 9505<br />

Kurihara, Hiroaki 10519<br />

Kurihara, Laurie 10587<br />

Kurkjian, Carla 3008, 3041,<br />

3067, 3097, e17012<br />

Kurkure, Purna Arun 9559<br />

Kurland, Brenda F 1088,<br />

TPS3109, 5503, 5515<br />

Kurland, John F. TPS8603<br />

Kurmasheva, Raushan 9544<br />

Kuroda, Ken e19577<br />

Kuroda, Nobukazu e14518<br />

Kuroi, Katsumasa 540<br />

Kurokawa, Koji e17540<br />

Kurokawa, Mineo 6533<br />

Kurosaka, Masahiro e17020<br />

Kurosumi, Masafumi 613<br />

Kurozumi, Sasagu 613<br />

Kursar, Jonathan D. 3008<br />

Kurt, Hakan e15104<br />

Kurtagic, Elma 4056<br />

Kurtin, Sandra E 3028<br />

Kurtz, Jean Emmanuel 636<br />

Kurtz, Jean-Emmanuel 2605,<br />

4071, 10056<br />

Kurzeder, Christian 5031<br />

Kurzrock, Razelle 535, 2583,<br />

3003, 3078, 3106, 3107^,<br />

TPS3117, 3528, 4639,<br />

5021, 8531, 8551, 8594,<br />

10510, 10607, e13020,<br />

e13506, e13534, e13595,<br />

e13596, e13600^,<br />

e19004, e19055<br />

Kusche, Manfred 10600<br />

Kuschel, Petra 10046<br />

Kushi, Lawrence H e14160<br />

Kushnir, Michal e14067<br />

Kuss, Iris LBA10008<br />

Kussman, Ashleigh M.<br />

e14098<br />

Kusuhara, Masatoshi e14635<br />

Kusumoto, Masahiko 1530<br />

Kusumoto, Sojiro e13029<br />

Kusumoto, Tetsuya e14722<br />

Kusunoki, Masato e14029,<br />

e14039, e14541, e21053<br />

Kusunoki, Yoko 10577<br />

Kutateladze, Tatiana G. 7504<br />

Kuten, Abraham e18511<br />

Kutikov, Alexander 4526<br />

Kutluk Cenik, Bercin e13563,<br />

e18076<br />

Kutscheidt, Andreas 1077<br />

Kutscher, Sarah 2522<br />

Kuttruff, Sabrina 2522, 3530<br />

Kutwak, Flavia 7049<br />

Kuvshin<strong>of</strong>f, Boris W. e14659<br />

Kuykendal, Adam Rea 4647<br />

Kuzel, Timothy e13088,<br />

e18523<br />

Kuzman, James A e15580<br />

Kuznetsov, Galina 3030<br />

Kvale, Elizabeth 6080<br />

Kvikstad, Anne 9040<br />

Kvols, Larry 4125, e14579<br />

Kwa, Maryann e11010<br />

Kwak, Eunice Lee 2566,<br />

3053, 3528, 4021, 4027,<br />

7508, e14615<br />

Kwak, Larry W. 2528, 8067<br />

Kwan, Min-Fun Rudolf 4654<br />

Kwan, Sherry e15154<br />

Kwari, Monica 6501<br />

Kwiatkowski, David J. 7589<br />

Kwiatkowski, Fabrice 1051<br />

Kwo, Paul e14683<br />

Kwoh, T. Jesse e13562<br />

Kwok, Chloe 4105, e14706<br />

Kwok, Mary 8088, 8104,<br />

e18568<br />

Kwok, Shirley 10586<br />

Kwon, Deukwoo e16001<br />

Kwon, Hyuk-Chan e14686<br />

Kwon, Jung Hye 9065,<br />

e19599, e19603, e19613<br />

Kwon, Tack-Kyun 5552<br />

Kwon, Woo Sun 3606<br />

Kwong, Philip 4105<br />

Ky, Bonnie 4500<br />

Kühne, Thomas 10081<br />

Kyle, Robert A. 8105<br />

Kyriakopoulos, Christos 1071,<br />

e11038<br />

Kyuichi, Kadota 7052<br />

König, Jochem 1082<br />

König, Klaus e11040^<br />

L<br />

L’Heureux, Darryl TPS5114,<br />

TPS10635<br />

La Delfa, Anthony 9032<br />

La Quaglia, Michael P. 8586,<br />

TPS9595<br />

La Russa, Francesca 4541<br />

La Spina, Chiara Maria 7082<br />

La Torre, Ignazia 1073<br />

La Verde, Giacinto e17006,<br />

e18563, e18566<br />

La-Beck, Ninh My 2591,<br />

5050<br />

La-Touche, Susannah 4640<br />

Laack, Eckart LBA7507<br />

747s


Laack, Nadia N. 2032,<br />

e14507<br />

Laadem, Abderrahmane 607,<br />

e15170, e19609<br />

Labadie, Julia 1100<br />

labardini-Mendez, Juan 6599<br />

Laberge, Francis 7511<br />

Labianca, Roberto LBA4001,<br />

4053, LBA7501, TPS9155,<br />

e19549, e19612<br />

Labiano, Tania 8572, e18008<br />

Labidi-Galy, Sana Intidhar<br />

10562<br />

Labonte, Melissa Janae 3540,<br />

3546, 3584, 3604, 3622,<br />

4026, 4088, e11018,<br />

e14034, e14052<br />

Labouba, Ingrid e15202<br />

Labow, Daniel M. e14656<br />

Labrie, Fernand 10583<br />

LaCasce, Ann S. e18530<br />

Lacave, Roger e18075,<br />

e18089<br />

Lachance, Daniel Honore<br />

9075<br />

Lack, Carole 9589<br />

Lackey, Jacqueline 4568<br />

Lackner, Mark e21122<br />

Lacombe, Denis A. 2, 2540<br />

Lacombe, Sophie 4018<br />

Lacoste, Gisele TPS7610<br />

Lacour, Valerie 4622<br />

Lacouture, Mario E. 8515,<br />

9088, 9092, e13591,<br />

e18571, e19052, e19624<br />

Lacroix, Joëlle 4618<br />

Lacroix, Ludovic 10556<br />

Lacroix, Magali TPS667<br />

Lacroix-Triki, Magali 543<br />

Lacy, Antonio 3536<br />

Lacy, Cindy 2077<br />

Lacy, Jill e14534<br />

Lacy, Martha 8043, 8092,<br />

8096, 8105, TPS8116<br />

Lad, Thomas E. 9085<br />

Ladabaum, Uri 6077<br />

Ladanyi, Marc 1541, 7505,<br />

7532, 7578, 7580, 7589,<br />

10524, 10560<br />

Ladekarl, Morten e21110<br />

Lademann, Juergen 5064,<br />

e19558<br />

Ladha, Abdullah e19627<br />

Ladner, Robert D. 3584,<br />

3622<br />

Ladrera, Guia Elena 7519^<br />

Lae, Marick 10018<br />

Lafitte, Jean-Jacques<br />

TPS7111, e18006, e18066<br />

Laflamme, Christian 634<br />

Lafleur, Josiane TPS1139<br />

LaFrance, Norman David<br />

e13592<br />

Lagarde, Pauline 9536<br />

Lage, Agustin 2527<br />

Lagerwaard, Frank J 7009<br />

Lagier, Robert 10586<br />

Lagrone, Lore 2003<br />

Laguerre, Brigitte LBA4567^<br />

Laheru, Dan 3596, 4045,<br />

4055, e14585<br />

Lahn, Michael M. F. 2042<br />

Lahogue, Annick TPS649<br />

Lahuerta, Ainhara 10512,<br />

10595<br />

Lahuerta, Juan José 8095<br />

Lai, Cheng e13101<br />

Lai, Chyong-Huey 5109<br />

Lai, Dominic W. e16052<br />

Lai, Jin-Shei 9532<br />

Lai, Lily L. e14182<br />

Lai, Maria 4105, e14706<br />

Lai, Pamela 6083<br />

Lai, Rose 2006<br />

Lai, Ruay-Sheng TPS7614<br />

Lai, Siang Hui 2066<br />

Lai, Wei-Chu V 5078<br />

Lai, Xin e14706<br />

Lainas, Ioannis TPS6634,<br />

TPS8603<br />

Laine, Fabrice 8057<br />

Lainez Milagro, Nuria<br />

TPS4672, TPS4674,<br />

TPS4688<br />

Lainez, Jose Miguel e18555<br />

Laird, A. Douglas 8531<br />

Laird, Glen 7584<br />

Lakdawalla, Darius 4666,<br />

e15186<br />

Lake, Diana 10514<br />

Lake, Jessica 7054, 7055<br />

Lakomy, Radek 3505, 3579<br />

Lakshmanan, Imayavarambam<br />

e17549<br />

Lal, Anita 10576<br />

Lal, Priti 4596<br />

Lal, Rohit 7562<br />

Lalet, Caroline e14023<br />

Laliberte, Julie 9500, 10587<br />

Laliberte, Robert e13101<br />

Lall, Terrance e21026<br />

Lalla, Deepa 1037, 9144,<br />

e11063, e11076, e11077,<br />

e19633<br />

Lallas, Costas D. 4526,<br />

e15012<br />

Lallemant, Benjamin e16044,<br />

e16051, e16055<br />

Lally, Brian E. e16518<br />

LaLonde, Michelle 9115<br />

Lam, Elaine Tat 3017<br />

Lam, Paula 2066<br />

Lam, Raymond 10573,<br />

e13598<br />

Lam, Wayne 4640, e15100<br />

Lamanna, Nicole 6122,<br />

6609, 6613<br />

Lamar, Maria 4006, 4117,<br />

4545, 8519<br />

Lamar, Ruth E. 618,<br />

TPS8115<br />

Lamarca, Angela 10547,<br />

e13085<br />

Lambert, John M. 8057<br />

Lambiase, Antonio 2580,<br />

5059, 6541, 7076, 7085,<br />

7581<br />

Lambrechts, Diether 507,<br />

1020<br />

Lambrechts, Sandrina 5058<br />

Lamerato, Lois e12024,<br />

e15147, e15197, e18107,<br />

e19630<br />

Lamm, Donald L. 4593<br />

Lammers, Philip Edward<br />

6038<br />

Lammertsma, Adriaan A.<br />

7603<br />

Lamont, Elizabeth B. 1605<br />

Lamont, J. V. e14022<br />

Lamparella, Nicholas E.<br />

e15062<br />

Lampe, Dieter 624<br />

Lampron, Megan 4543, 4635<br />

Lamy, Aude 5565, e14010<br />

Lamy, Pierre-Jean 10505<br />

Lamy, Thierry 8026<br />

Lan, Guiping 5558<br />

Lan, Lan 1605<br />

Lan, Yun 4005<br />

Lancaster, Johnathan M.<br />

1585<br />

Lance, Ray 4586<br />

Lancelotta, Mary Pat 10590<br />

Lancet, Jeffery 6588<br />

Lancet, Jeffrey E. 3081,<br />

6568, 6586, 6601, 6608,<br />

TPS6637<br />

Lanchbury, Jerry S. 7023<br />

Lanctot, Jennifer 9526<br />

Lanczky, Andras 10571<br />

Land, Stephanie R. TPS1142<br />

Land, Susan 7063<br />

Landa, Jonathan 4548,<br />

10002<br />

Landau, Danny e15010,<br />

e15102<br />

Landaverde, Denis e16547<br />

Landeiro, Luciana Garcia<br />

5588<br />

Landen, Charles N. 5036<br />

Lander, Thomas 2573^<br />

Landers, Mark 1058<br />

Landesman, Yosef 1055,<br />

4634, e13549<br />

Landgren, Ola 8088, 8104,<br />

e18568<br />

Landi, Bruno 10078<br />

Landi, Lorenza 3521,<br />

LBA4001, e21018, e21095<br />

Landier, Wendy 9505<br />

Landolfi, Joseph C. TPS2107<br />

Landolfi, Stefania 4089,<br />

e21021<br />

Landreneau, Joshua P 7071<br />

Landreneau, Rodney Jerome<br />

7071, 7074<br />

Landry, Jerome Carl 3605<br />

Landsman, Keren 1578<br />

Landsman-Blumberg, Pamela<br />

593<br />

Lane, Alex R. e15116<br />

Lane, Ashley R. 9543<br />

Lane, Dorothy S 1501<br />

Lane, Kathleen A. e16557<br />

Lane, Sharon e16515,<br />

e16559<br />

Lang, Amy 2555<br />

Lang, Evan Z. 4127<br />

Lang, Forrest TPS9147<br />

Lang, Frederick F. 2019<br />

Lang, Hauke TPS3641^<br />

Lang, Jin Yi 5597<br />

Lang, Jin-yi 5535<br />

Lang, Katherine 1563<br />

Lang, Kathy 9144, e19633<br />

Lang, Melanie e19502<br />

Lang, Roland 8563<br />

Lang, Wenhua 5524<br />

Langdon, Robert M. 4085,<br />

TPS7619<br />

Lange, Georgia 1028<br />

Lange, Joan E e16053<br />

Lange, Julie R. 1128<br />

Lange, Paul H. 4520<br />

Langer, Christiane TPS3635^<br />

Langer, Claus 10031<br />

Langer, Corey J. 5583, 7077,<br />

7092, 7098, 7590, 7595,<br />

TPS7612, TPS9150,<br />

e16062, e18032<br />

Langer, Seppo W. 4015<br />

Langer, Stefan 10059<br />

Langholz, Bryan 9515<br />

Langleben, Adrian 1099<br />

Langley, Emma J. e14584<br />

Langley, Ruth E TPS4143<br />

Langridge, Carolyn I 5527<br />

Langston, Amelia A. 8101<br />

Lani, Elisa e21095<br />

Laniado, Michael 10068<br />

Lanier, Keith S. e16561<br />

Lanigan, Christopher 620,<br />

e18085<br />

Lankeit, Henrike Katharina<br />

8052<br />

Lankheet, Nienke A.G. 2596<br />

Lann, Danielle 1540<br />

Lannin, Donald R. 1045,<br />

1109, 1121<br />

Lannoy, Valerie e14044<br />

Lannutti, Brian Joseph 6518^<br />

Lanoy, Emilie 4621, 10044,<br />

e18071<br />

Lantzsch, Tilmann 624<br />

Lanuti, Michael 7010<br />

Lanza, Giovanni 3510<br />

Lanzalone, Silvana 7533<br />

Lanzardo, Stefania e13527<br />

Lanzetta, Gaetano 4627<br />

Lanzirotti, Antonio e13067<br />

Lanzos Gonzalez, Eduardo<br />

e15033<br />

Lanzotti, Victor J. 6054<br />

Lao Romera, Juan e11081<br />

Lao, Christopher D. 8521<br />

Lao, Juan 10512, 10595<br />

Lao-Sirieix, Si-houy 4121<br />

Lapadula, Vittoria 9133,<br />

e14096<br />

Laperriere, Normand 2008b,<br />

TPS2104<br />

Lapique, Nicolas 3511<br />

LaPlant, Betsy 6570, 8013,<br />

8043<br />

Lapolla, Assunta Maria<br />

e11001<br />

Laporta, Rosalia e12015<br />

Lapunzina, Pablo 3618,<br />

10547<br />

Lara, Gisell Anaid 1607<br />

Lara, Jonathan F. 614<br />

Lara, Miguel Angel e11074<br />

Lara, Primo 3566, 4511,<br />

4547, 4607, 4663, 7514,<br />

10503, e17537<br />

Lara-Medina, Fernando<br />

e12027<br />

Larbi, Emmanuel Douglas<br />

e15517<br />

Lardelli, Pilar TPS652,<br />

TPS10101<br />

Largillier, Remy 568^,<br />

e11019<br />

Lariou, Maria Stella e20002<br />

Larkin, James M. G.<br />

TPS4679, TPS4682,<br />

8502^, LBA8509, 8517,<br />

TPS8605, 10038<br />

Larkins, Gail 8016<br />

Larocca, Luigi Maria e14042,<br />

e18021<br />

LaRocca, Renato V. TPS2107<br />

Larry, Wang 9564<br />

Larsen, Eric 9502, CRA9508<br />

Larsen, Julie S. TPS666<br />

Larsen, Linda 1124<br />

Larsen, Stig Einride e11022<br />

Larsimont, Denis 507,<br />

e21010<br />

Larson, Melissa L. e18510<br />

Larson, Peder E. 4660<br />

Larson, Richard A. 2533,<br />

6509^, 6526, 6562, 6582<br />

Larson, Robin Joyce 6013<br />

748s Numerals refer to abstract number


Larson, Steven M. 3563,<br />

4517, 4548, 5509^<br />

Larsson, Olle 7539<br />

Larue, Christine 10098<br />

Lasalvia-Galante, Eduardo E.<br />

e13105<br />

Lasalvia-Prisco, Eduardo M.<br />

e13105<br />

Lasch, Kathryn Eilene<br />

e12515<br />

Laser, Benjamin S. 5088<br />

Lash, Alex e19647<br />

Laskin, Janessa J. 7020<br />

Lasley, Foster D e14683<br />

Lassau, Nathalie 3080, 4618<br />

Lasso, Roberto 10079<br />

Last, David 2078<br />

Lastiri, Jose Maria 10074<br />

Lastoria, Secondino e14130<br />

Lastoria, Secondo e19034<br />

Lata, Suman 531<br />

Lathan, Christopher S.<br />

e18067<br />

Lathia, Chetan D. e13576,<br />

e18100<br />

Latiano, Tiziana Pia e21092<br />

Latiano, Tiziana e19035<br />

Latif, Asma e21032<br />

Latif, Marcia e19647<br />

Latif, Rabia e18504<br />

Latko, Ewa 9026^<br />

Latorre, Claudia Grandino<br />

e19552<br />

Latorzeff, Igor LBA4567^<br />

Latouche, Aurelien TPS667<br />

Latreille, Jean LBA3500^<br />

Latronico, Antuono 1500<br />

Lattanzio, Laura 2600<br />

Latten, M. J. e14022<br />

Latteri, Fiorenza e18026,<br />

e18096<br />

Lau, Anthea 7606<br />

Lau, Clayton e15178<br />

Lau, Derick 7517<br />

Laubacher, Barbara A.<br />

e21107<br />

Laubender, Ruediger Paul<br />

3541, 3588, 3603, 4023,<br />

e14043<br />

Lauderdale, Katharine E 9132<br />

Lauer, Richard C. 4603<br />

Lauer, Ulrich M 4115<br />

Laughlin, Anne 3082<br />

Launay-Vacher, Vincent 1095,<br />

5067<br />

Launchbury, Francesca 4653<br />

Laura, Perelli e11044,<br />

e11501, e18125<br />

Lauratet, Betty e11551<br />

Laurent, Dirk 3502,<br />

LBA10008<br />

Laurent, Louise C. 10539<br />

Laurent-Puig, Pierre 2537^,<br />

5098, 5554, e14059,<br />

e14129<br />

Lauria, Rossella 1554,<br />

LBA5003<br />

Lauricella, Calogero 3570,<br />

LBA7501<br />

Laurie, Scott Andrew<br />

TPS2615, 7093, 7511<br />

Laurino, Mercy 3567<br />

Lauseker, Michael 6510<br />

Lausoontornsiri, Wirote 4108,<br />

e13013, e16040<br />

Lautenschlaeger, Tim e21044<br />

Laux, Douglas Earl 3056<br />

Lauzier, Sophie 9138<br />

LaVallee, Theresa TPS2618<br />

Numerals refer to abstract number<br />

Lavelle, Aonghus e19601<br />

Lavenia, Giuseppe e15544<br />

Laver, Joseph H. 9526<br />

Lavergne, Emile e15009<br />

Laverman, Peter e13111<br />

Lavery, Hugh J. e15164<br />

Lavey, Robert S 10048<br />

Lavole, Armelle e16560,<br />

e18075, e18102<br />

Lavolle, Armelle 7574<br />

Lavori, Philip W. 5525<br />

Lavranos, Tina C. 4603,<br />

7079^<br />

Law, Linda 4085, e14564<br />

Law, Wai Lun TPS10632,<br />

e14032<br />

Lawal, Olukayode O e11554,<br />

e12022<br />

Lawal, Taoreed O e14734,<br />

e14736<br />

Lawrence, Donald P.<br />

CRA2509, 8503^, 8507,<br />

8510, 8516, 8522, 8569<br />

Lawrence, H. Jeffrey 10596<br />

Lawrence, Scott M 3031<br />

Lawrence, Theodore Steven<br />

e14603<br />

Lawrence, Yaacov Richard<br />

e14143<br />

Lawson, David H. 2576,<br />

8543, 8567, e13088<br />

Lawton, Thomas J. 516<br />

Lax, Sigurd e14066<br />

Lay, Harry H 2074^<br />

Lay, John C. 2591<br />

Layman, Rachel M. CRA1002<br />

Layne, Tracy Melissa e16511<br />

Lazar, Vladimir 7529<br />

Lazaridis, George 592,<br />

e15040<br />

Lazaro Quintela, Martin<br />

TPS4672, e12012,<br />

e15067, e15076, e15077,<br />

e18040, e18146<br />

Lazarus, Hillard M. TPS1616<br />

Lazaryan, Aleksandr 8055<br />

Lazovich, DeAnn 8574<br />

Lazzarelli, Silvia 4097<br />

Lazzaretti, Maria Grazia<br />

e11071<br />

Lazzari, Chiara e18096<br />

Lazzeroni, Matteo 519, 1500<br />

Le Beau, Michelle M. 6562<br />

Le Boulicaut, Julie 10089<br />

le Caer, Herve 7574, e16051<br />

Le Cesne, Axel 10005,<br />

10006, 10007, LBA10008,<br />

10009, 10013, 10016,<br />

10017, 10020, 10023,<br />

10027, 10033, 10044,<br />

10056, 10062, 10067,<br />

10078, 10089, 10092<br />

Le Chevalier, Thierry 7007<br />

Le Corre, Delphine e14059<br />

le Coutre, Philipp D. 6503,<br />

6512, TPS6640<br />

Le Deley, Marie-Cécile 9517,<br />

TPS9596<br />

le Frere-Belda, marie-Aude<br />

5098, e15558<br />

Le Gall, Fabrice e18532<br />

Le Gall, Francois e19037<br />

Le Gouill, Steven 8026<br />

Le Guellec, Sophie 10076<br />

Le Loupp, Anne-Gaelle 2082,<br />

e21117<br />

Le Maignan, Christine 10042,<br />

e11019<br />

Le Monies, Alise e14168<br />

Le Page, Cécile e15202<br />

Le Pechoux, Cécile 10016,<br />

10044, e18071<br />

Le Rhun, Emilie TPS662,<br />

2070, e11012, e12506<br />

Le Teuff, Gwenael 3063,<br />

7007<br />

Le Tourneau, Christophe<br />

e13017<br />

Le, Anh T 7504<br />

Le, Dung Thi 4045<br />

Le, Lisa W. 7606<br />

Le, Lisa e16567<br />

Le, Lyly H. 6071<br />

Le, Ngocdiep T. 3003<br />

Le-Petross, Huong T. 594,<br />

1107<br />

Lea, Jayanthi e13078,<br />

e15501<br />

Leach, Laura M. 9115<br />

Leahey, Ann 9515<br />

Leahy, Michael Gordon<br />

LBA10008, TPS10101<br />

Leahy, Terri 2013<br />

Leak, Ashley Nicole 6119<br />

Leal, Alessandro e11513<br />

Leal, Alexis D. 9057<br />

Leal, Jeffrey P 4045, 10509<br />

Leandro-Garcia, Lj 10546<br />

Leary, Colleen e14549,<br />

e14550<br />

Leary, Sarah 9519, 9581<br />

Leasure, Justin 9544<br />

Leasure, Nick C. TPS657<br />

Leath, Charles A. e15578<br />

Lebbe, Celeste 1566, 8512,<br />

8518, 8591, 8601, 10056,<br />

e13594<br />

Lebedinsky, Claudia<br />

TPS6639, TPS6641<br />

Leber, Brian 6505^<br />

Lebitasy, Marie Paule<br />

TPS7111<br />

Leblanc, Eric 5083<br />

LeBlanc, Michael Leo 8001<br />

Leblond, Veronique 8030<br />

Leboeuf, Rebecca 5509^<br />

Lebot, Marie-Annick e14148<br />

Leboulleux, Sophie 5569<br />

Lebourgeois, Paul A. 3630<br />

Lebovic, Daniel 8011<br />

Lebowicz, Yehuda Z.<br />

TPS5601<br />

Lebrec, Jeremie 10619<br />

Lebret, Thierry e15145<br />

Lebrun, Christine e12502<br />

Lebrun-Jezekova, Daniela<br />

9079<br />

Lebrun-Ly, Valerie 10078<br />

Lebwohl, David Edward 512,<br />

559<br />

Leccia, Marie Thérèse 8591<br />

Lechuga, Carmen e15033<br />

Lechuga, Mariajose 4644<br />

Leclercq, Nathalie e18006<br />

Lecomte, Thierry 3520, 4018<br />

Lecumberri, Ramon 7095<br />

Lecuru, Fabrice 5098,<br />

e15558<br />

Ledermann, Jonathan A.<br />

4004, 4029<br />

Ledesma, Wendy M. 555,<br />

563, 6063<br />

Lee, Adam 3564, 3617<br />

Lee, Adrian Pak Siu e12507<br />

Lee, Adrian e19042<br />

Lee, Agnes Y. TPS9149^<br />

Lee, Ann Sing 4105, e14706<br />

Lee, Annette 1046<br />

Lee, Belinda e21137<br />

Lee, Bong-Jae e18501<br />

Lee, Bonnie e16523<br />

Lee, Brian 9553<br />

Lee, Byeong IL e14580<br />

Lee, Byung-In 1058, e14164<br />

Lee, Changsuk e14543<br />

Lee, Charlene e15042<br />

Lee, Chee 3534, 5081<br />

Lee, Ching-Chih e16008<br />

Lee, Christina R. e15177^<br />

Lee, Christopher W. 7511<br />

Lee, Clara Inkyung e19011<br />

Lee, Conrad 4105, 5511<br />

Lee, Dae Ho 7004, e13104,<br />

e15080, e18052<br />

Lee, Dana e16533<br />

Lee, Dennis J e15060<br />

Lee, Donghoon e18119<br />

Lee, Edmund Jon Deoon<br />

2598<br />

Lee, Edward James 6591<br />

Lee, Eudocia Quant 2005<br />

Lee, Fook Thean e15056<br />

Lee, Geon Kook e21043<br />

Lee, Geun Dong e14573<br />

Lee, Gil Yeon e14128<br />

Lee, Gin-Hyug 4093<br />

Lee, Grace Lauren e13084<br />

Lee, Guek Eng 3098, 3589<br />

Lee, Guk Jin e14558,<br />

e19614<br />

Lee, Gwo-Shu Mary 4516,<br />

4543, 4635<br />

Lee, Hae Won e17523<br />

Lee, Haekyung 1053<br />

Lee, Hee Jin e11504<br />

Lee, Hee Seok e21043<br />

Lee, Hee Yeon e14558,<br />

e19614<br />

Lee, How-Sung e13002<br />

Lee, Hun Ju 6598<br />

Lee, Hwang-Jae e19574<br />

Lee, Hye Yoon 2542<br />

Lee, Hyo Jin e12029,<br />

e14529<br />

Lee, Hyun Woo e14580,<br />

e18519<br />

Lee, Hyun-Ju 2541<br />

Lee, Hyun-Jung 3624,<br />

e14136<br />

Lee, Hyun-Sung e16534<br />

Lee, J. Jack 5524, 7047,<br />

7078<br />

Lee, Jae Cheol e18052<br />

Lee, Jae Hoon 8095, 8097,<br />

e14137, e14685, e18572^<br />

Lee, Jae Seok 6564<br />

Lee, Jae-Lyun 3568, 3592,<br />

4536, 4538, 4544^, 4628,<br />

e13104, e13560, e15080<br />

Lee, Jang-Ming e14576<br />

Lee, Jay Ted e19509<br />

Lee, Je-Hwan 6564<br />

Lee, Jeanette Y 4030, 8005<br />

Lee, Jeeyun 4011, 4064,<br />

10067, 10535, e12029,<br />

e13044, e13094, e14072<br />

Lee, Jeffrey Edwin 4051,<br />

8534, e14548<br />

Lee, Jeffrey H. 4051<br />

Lee, Jeffrey H 4069, 4078,<br />

4086, e14547, e14669<br />

Lee, Jennifer TPS4134<br />

Lee, Jeong-Ok 7566<br />

Lee, Jerry 1058<br />

Lee, Ji Hyun 1053<br />

Lee, Ji Hyun e14686<br />

Lee, Ji Yean e16003, e18148<br />

749s


Lee, Ji-Hyun TPS3114, 6100<br />

Lee, Jihye e13044<br />

Lee, Jill Lunsford e16502<br />

Lee, Jin Soo 7089, 7519^,<br />

e18119<br />

Lee, Jin e19611<br />

Lee, Jinseon 7053, e18083,<br />

e21062<br />

Lee, John Muzi 9019<br />

Lee, John e19580<br />

Lee, Jong Won e11508<br />

Lee, Jong Yeul 4028<br />

Lee, Jong-Eun e13065<br />

Lee, Jong-Seok 3076,<br />

TPS7614<br />

Lee, Ju Ruey-Jiuan 10596<br />

Lee, Ju-Whei 5566, 5576<br />

Lee, Jun Ho 4028<br />

Lee, Jun Woo e14566<br />

Lee, Jung Shin 3592, 7004,<br />

e18052<br />

Lee, Jung-Hee 6564<br />

Lee, Jung-min 2545<br />

Lee, Jungsin e13061,<br />

e13104<br />

Lee, Kang Young e14081,<br />

e14084<br />

Lee, Keun Seok 2542<br />

Lee, Keun Wook e14580<br />

Lee, Keun-Seok TPS649,<br />

1003<br />

Lee, Keun-Wook 3076,<br />

e14572<br />

Lee, Khai Mun 5553<br />

Lee, Ki Hyeong LBA7500<br />

Lee, Kichun Sky 7048<br />

Lee, Kyoo-Hyung 6564<br />

Lee, Kyu Seop e14529<br />

Lee, Kyu Taek e14688,<br />

e19538, e19554<br />

Lee, Kyung Hee TPS4142,<br />

e14570, e14572, e19538<br />

Lee, Kyung-Hun 2542, 3624,<br />

5552, e14136<br />

Lee, Kyung-Min 1056<br />

Lee, Lawrence Soon-U<br />

e13002<br />

Lee, Lip e18095<br />

Lee, Louis e15110<br />

Lee, Luen F. 3522, 3523,<br />

3525<br />

Lee, Lynette e14155<br />

Lee, Mark 3512, 4560<br />

Lee, Michele 1118<br />

Lee, Michelle Mei Lin 4106<br />

Lee, Min Mi 2041<br />

Lee, Min-Jung 3087, 7033<br />

Lee, Moon Hee 1003,<br />

TPS4142<br />

Lee, Myung Ah e14558,<br />

e14634, e19614<br />

Lee, Myung Kyung 9066<br />

Lee, Namsu e14688, e19554<br />

Lee, Nancy Y. 5537, 5545^<br />

Lee, Patrick W. e21090<br />

Lee, Peter 3010, TPS8109<br />

Lee, Po-Huang e13061,<br />

e20514<br />

Lee, Richard J. 4566<br />

Lee, S. Ming 7538<br />

Lee, Sandra J. 8522<br />

Lee, Sang Cheol e19554<br />

Lee, Sang-Cheol e14688<br />

Lee, Sang-Joon 5549,<br />

TPS5602<br />

Lee, Sang-wook 5586,<br />

e18501<br />

Lee, Se-Hoon 4544^, 5552,<br />

7004, 7566, e15047,<br />

e15064<br />

Lee, Shih-Yuan 4549<br />

Lee, Shing Mirn 2541<br />

Lee, Simon Craddock e19536<br />

Lee, Siow-Ming 7562<br />

Lee, Soo Jung 3554, 6536,<br />

6538, e14074, e18533<br />

Lee, Soo-Chin 530, 1064,<br />

1575, 2551, 3002,<br />

e13002, e19523<br />

Lee, Soohyeon 1003, 2542,<br />

e16529<br />

Lee, Soonil 4064<br />

Lee, Stephanie 6103, 9139<br />

Lee, Su Jin 644, 7104<br />

Lee, Suee e14686<br />

Lee, Sun Ah e14570<br />

Lee, Sunmin 3087<br />

Lee, Susanna TPS5114<br />

Lee, Sze Ting e15056<br />

Lee, Sze Xian e21078<br />

Lee, Tani Ann T. 4011<br />

Lee, Theresa 1521<br />

Lee, Ting Yim TPS5114<br />

Lee, Victor 5511, 7519^,<br />

e18063^<br />

Lee, William 3086<br />

Lee, Won Chan 570<br />

Lee, Won-Suk e14137<br />

Lee, WY e15110<br />

Lee, Yeon-Su 7089<br />

Lee, Yihua 4007, 4504,<br />

4513, 5547<br />

Lee, Yin Leng 1593<br />

Lee, Yong Chan e14559<br />

Lee, Yoo Jin 6536, e18533<br />

Lee, Young Joo e18119<br />

Lee, Young-Shin 6564<br />

Lee, Yu-Chin e18132<br />

Lee, Zi-jia e11068<br />

Leen, Ann M. 2558<br />

Leer, Jan Willem 9070<br />

Lees, Michael e19010<br />

Leeuwen, Frank van 9550<br />

Lefebvre, Caroline 530<br />

Lefesvre, Pierre e14044<br />

Lefeuvre, Delphine 3063<br />

Lefeuvre-Plesse, Claudia<br />

e11012<br />

Lefevre, Arthur e12506<br />

Lefranc, Anne 10027<br />

Lefranc, Jean-Pierre 641,<br />

e11551<br />

Legare, Robert Duffy 1045<br />

Legouffe, Eric 4625,<br />

TPS4692^, 9011<br />

Legoux, Jean-Louis 3506<br />

Legrier, Marie-Emmanuelle<br />

e15161<br />

Legros, Laurence 6523<br />

Lehman, Robert TPS1145<br />

Lehmann, Frederic 7013,<br />

7056<br />

Lehmann, Karl M 5051<br />

Lehmann, Sylvia 10552<br />

Lehmkuhl, Hans 8030<br />

Lehner, Julia e21067<br />

Lehners, Nicola 6519<br />

Lehnert, Manfred 2512<br />

Lehtimaki, Terho e15564<br />

Lei, Junying 1508<br />

Lei, Lei e17512<br />

Lei, Xiudong 537, 1029,<br />

1104<br />

Leibold, Tobias 3563<br />

Leibovich, Bradley C. 4657<br />

Leiby, Benjamin E e16548<br />

Leichman, Lawrence P. 4052<br />

Leigh, Bryan R. 3034,<br />

3042^, e21038<br />

Leigh, Meggan TPS661<br />

Leighl, Natasha B. 1535,<br />

6005, 7541, 7586, 7606,<br />

9003, 9032, 10522,<br />

e11000, e11050, e16505,<br />

e18079<br />

Leighton, John Charles<br />

e16561<br />

Leijen, Suzanne e13598<br />

Leinwand, Joshua e13064,<br />

e13066, e13067<br />

Leinweber, Clinton 2032<br />

Leip, Eric 6511<br />

Leis, Jose Francisco 8053<br />

Leisen, Margarete 10041^<br />

Leisenring, Wendy M. 6030,<br />

9505, CRA9513, 9514,<br />

9528<br />

Leitao, Mario M. 9104<br />

Leitch, A. Marilyn 503, 1107<br />

Leitzel, Kim 501, 607, 614,<br />

e14520, e15062, e15121,<br />

e15170, e21039<br />

Lekakis, Lazaros e11519<br />

Leleu, Xavier 8009, 8014,<br />

8095<br />

Lema, Mauricio e19604<br />

Lemare, Francois 9560, 9572<br />

Lemarié, Etienne e18100<br />

Lemas, M. Victor 8003<br />

Lemech, Charlotte Rose<br />

e13585<br />

Lemech, Charlotte 3000<br />

Lemieux, Julie 634, 9138<br />

Lemke, Klaus 6026<br />

Lemke, Sheila M. e11089<br />

Lemoine, Antoinette e14521<br />

Lemonnier, Christine 1574<br />

Lena, Hervé TPS7112, 7584<br />

Lenain, Pascal 8014<br />

Lencioni, Monica 4006<br />

Lencioni, Riccardo 4108,<br />

TPS4152<br />

Lendinez, Alberto e14147<br />

Lendvai, Nikoletta TPS8111<br />

Lengerke, Claudia e15577<br />

Lengfelder, Eva 6610, 8024<br />

Lenhard, Miriam Susanne<br />

1114<br />

Lennes, Inga Tolin 6048,<br />

6062, 6106, 7524, 9030<br />

Lensing, Janet 3001<br />

Lentzsch, Suzanne 8099<br />

Lenz, Cosima 3584<br />

Lenz, Felicitas 3584<br />

Lenz, Guido e13546<br />

Lenz, Heinz-Josef 3078,<br />

3502, 3518, 3540, 3546,<br />

3549, 3584, 3597, 3604,<br />

3615, 3622, TPS3635^,<br />

4010, 4019, 4022, 4026,<br />

4047, 4049, 4088, 4096,<br />

4113, e11018, e14031,<br />

e14034, e14052, e14679<br />

Lenzi, Renato 4130, 4131<br />

Leo, Ermanno e21118<br />

Leo, Silvana Assunta e18048<br />

Leon Mateos, Luis TPS4674,<br />

TPS4688, e15067,<br />

e15076, e15077<br />

Leon Rodriguez, Eucario<br />

e15567<br />

Leon, Ana 5520, e14147,<br />

e15094, e18069, e18106,<br />

e21015<br />

Leon, Larry 7573<br />

Leon, Luis TPS4685, e19505<br />

Leon, M. 4630<br />

Leon, Paola Beatriz 4089<br />

Leon, Xavier 5590, 5595<br />

Leon-Ferre, Roberto e14620<br />

Leonard, David e19536<br />

Leonard, E. Jane TPS8109,<br />

TPS8110<br />

Leonard, Gregory e14687<br />

Leonard, John 6518^, 8000<br />

Leonard, Robert C. F. 1094<br />

Leone, Alvaro e19033<br />

Leone, Bernardo Amadeo<br />

e11021<br />

Leone, Francesco 4110<br />

Leone, Jose Pablo e11021<br />

Leone, Julieta e11021<br />

Leong, Lambert T 1599<br />

Leong, Lucille A. 3108<br />

Leong, Stephen 3017<br />

Leong, Swan e16024<br />

Leong, Trevor 3629,<br />

TPS4141<br />

Leong, Wey 1019<br />

Leongamornlert, Daniel 1545<br />

Leontovich, Alexey A 8599<br />

Leopold, Lance 2500^<br />

Lepage, Come 4129<br />

Lepère, Céline 3547, 4036<br />

Lepeu, Gerard 6633<br />

Leporati, Meri e11071<br />

Leppa, Sirpa 8074<br />

Leppert, John 10513<br />

Lera, Andrea Thaumaturgo<br />

e19552<br />

Lerchenmuller, Christian A.<br />

556, 4065, 4539<br />

Lerda, Daniel e21025<br />

Lerma, Enrique 1120, 10555<br />

Lerner, Seth P. 4586,<br />

TPS4673, TPS4678,<br />

e15002, e15003<br />

Lerner, Susan 6122, 6517,<br />

6598<br />

Lerner, Tatiana Goberstein<br />

e19552<br />

Leroux, Agnès 10517<br />

Lerro, Catherine C. 6008,<br />

6011<br />

Lerut, Evelyne e13061<br />

Lerzo, Guillermo Luis e11087<br />

Leschinger, Monika Iris 7554<br />

Lesho, Patricia 8075<br />

Lesimple, Thierry 8591,<br />

e13594, e19037<br />

Lesinski, Gregory B. e13549<br />

Leslie, Scott 4589, e15060,<br />

e15212<br />

Lesniak, Maciej S. 2014<br />

Lesnick, Connie E e13517<br />

Lesnikoski, Beth-Ann 1030<br />

Lesnock, Jamie L e15560<br />

Lesoin, Anne LBA5000, 5018<br />

Lesokhin, Alexander M. 2518<br />

Lesovoy, Vladmir 4501<br />

Lespagnol, Alexandra e19037<br />

Lessa, Rafael Costa 643,<br />

e18078<br />

Lessard, Laurent e15202<br />

Lesser, Glenn Jay TPS2107,<br />

9108<br />

Lester, Jason 6058<br />

Lestuzzi, Chiara 645<br />

LeTarte, Nathalie 2073,<br />

e13027, e19591<br />

Letendre, Louis 6614<br />

Leteurtre, Emmanuelle 4129<br />

Lethert, Kristine H. e11042<br />

Letocha, Henry e14531<br />

750s Numerals refer to abstract number


Letrent, Stephen P. 7530,<br />

TPS7615<br />

Letson, Douglas D 10048,<br />

10070<br />

Leunen, Karin 3048<br />

Leung, Abraham C. F. 5048<br />

Leung, CM e15110<br />

Leung, K C 4105<br />

Leung, Marco 8537<br />

Leung, Mova e19570<br />

Leung, Roland Ching-Yu<br />

e14545<br />

Leung, Sing Fai 5511,<br />

e15110<br />

Leuschner, Carola 3060<br />

Leuschner, Ivo 9573<br />

Leux, Christophe 2082<br />

Lev, Dina 10000<br />

Levchenko, Eugeny 8559<br />

Levenback, Charles 5027<br />

Levers, Marloes 9550<br />

Leverson, Glen e14554<br />

Levi, Edi 9578<br />

Levi, Francis 3547,<br />

TPS9154, e14006<br />

Levigne, Frederic e15109<br />

Levillain, Pierre 3520<br />

Levin, Glenn N e15192<br />

Levin, Tomer 9004, 9105<br />

Levin, Victor A. 2003, 2036<br />

Levin, Wendy J. 4010<br />

Levine, Douglas A. 5030,<br />

5043<br />

Levine, Edward A. LBA1000<br />

Levine, Edward Allen 6127,<br />

TPS10632<br />

Levine, Ellis Glenn 4506<br />

Levine, Mark Norman<br />

LBA1031, 6049<br />

Levine, Ross L. 6606, 9507<br />

Levinson, Adam W. e15164<br />

Levit, Claudio e15507<br />

Levitan, Denise A. 6524^<br />

Levitsky, Hyam I. 2513<br />

Levitt, Amanda e18061<br />

Levitt, Daniel 10036<br />

Levitt, Jonathan M. e15002<br />

Levra, Matteo G. 7085<br />

Levy, Antonin e15049<br />

Levy, Benjamin 3024, 6114,<br />

e18139<br />

Levy, Christelle e11019<br />

Levy, Richard S. 6626^<br />

Levy, Robert M. 2035<br />

Levy, Ronald 2513, 2514,<br />

e13555<br />

Levy-Gabriel, Christine 9538,<br />

e13017<br />

Lew, Danika 9018<br />

Lewandowski, Jennifer Ann<br />

6115<br />

Lewanski, Conrad R. 7538,<br />

7562<br />

Lewin, Sharyn Nan 6078,<br />

e13066<br />

Lewinshtein, Daniel 4669,<br />

e15207<br />

Lewis, Andrea L. 1512<br />

Lewis, Colleen Margaret<br />

2576, 3094<br />

Lewis, Jacqueline S e21137<br />

Lewis, James D. 1503<br />

Lewis, Jason S. TPS9595<br />

Lewis, Jen Kate 10525<br />

Lewis, Joe e15126<br />

Lewis, John D. e21090<br />

Lewis, Karl D 3102, 8503^,<br />

8510, 8579<br />

Lewis, Lionel D. 2533<br />

Numerals refer to abstract number<br />

Lewis, Marina T 10580<br />

Lewis, Mark 4075<br />

Lewis, Samantha 3028<br />

Leyland-Jones, Brian 626,<br />

1027, e13579, e21099<br />

Leynia, Pierre e14148<br />

Leyvraz, Loredana e19050<br />

Leyvraz, Serge 8532, 10033,<br />

e11048<br />

Lezama -Valle, Pablo e20005<br />

Lezon-Geyda, Kimberly 10508<br />

Lhomel, Christine 1566,<br />

1567, 8601<br />

Lhomme, Catherine 5028,<br />

10098<br />

Lhommel, Renaud 3559,<br />

5519<br />

Li Destri, Marta 5093, 5094<br />

Li, Aiwu e18123<br />

Li, Ang e17018<br />

Li, Baoping e13069<br />

Li, Benjamin Xiaoyi 9017<br />

Li, Benjamin e11541<br />

Li, Bing 7535<br />

Li, Chenghong 9526<br />

Li, Chunde 4561<br />

Li, Chung-I 7094, 7568<br />

Li, Claire 2601<br />

Li, Dan e18051<br />

Li, Daneng 5108<br />

Li, Dechuan e14049<br />

LI, Fang 1097<br />

Li, Gang e13045<br />

Li, Gavin 1583, 3632<br />

Li, Ge 10625<br />

Li, Gu e21060, e21111<br />

Li, Guisheng e16054<br />

Li, Hailun 8003<br />

Li, Hairong 8596<br />

Li, Haiyan e13589<br />

Li, Hanzhong 4628<br />

Li, Haojie 4092, e14575<br />

Li, Hong e21156<br />

Li, Jennifer 5019, e18549<br />

Li, Ji e14651<br />

Li, Jian e14604<br />

Li, Jianbin e11520, e11521,<br />

e13524, e13529, e13530<br />

Li, Jiayu 7535, 10527<br />

Li, Jie e14604<br />

Li, Jieran 10622<br />

Li, Jin 3038<br />

Li, Jing 1560<br />

Li, Jingjin TPS6643^<br />

Li, Jinhui 4558<br />

Li, Jun 1515, 1561, e14024<br />

LI, Kai 7548<br />

Li, Lang 505, 525, 2601<br />

Li, Li 1569, 10532<br />

Li, Ling 6003<br />

Li, Lingheng e14153<br />

Li, Longjiang 5507<br />

Li, Lu 7091, e14648,<br />

e18033<br />

Li, Marilyn M. 10598<br />

Li, Mingjie e18549<br />

Li, Mo-Dan e14026<br />

Li, Ning 9544, e14511,<br />

e14539<br />

Li, Qiang 7559, e18013<br />

Li, Qing e11025, e11538<br />

Li, Qiyu 9045<br />

Li, Qunna e21045<br />

Li, Robin 10539<br />

Li, Rong 1551, e12010,<br />

e12013, e17551<br />

Li, Rubi 7549<br />

Li, Rui 4645<br />

Li, Sandra e13585<br />

Li, Shan 3623<br />

Li, Sheng 9507<br />

Li, Shirley e18542<br />

Li, Shirong 8099<br />

Li, Shuhong 6581, e21079<br />

Li, Shukuan 1063<br />

Li, Shuling 4659, e15169<br />

Li, Si Ming 8506, 8562,<br />

e15051, e15052<br />

Li, Sierra Mi 10545<br />

Li, Sonia Patricia 1066<br />

Li, Szu-Chin e16008<br />

Li, Tao e14511<br />

Li, Tianhong 7582, 7594,<br />

e13526<br />

Li, Tianyu e14021<br />

Li, Wei 7091<br />

Li, Wen e21004<br />

Li, Wenhui e18033<br />

Li, Xi e18142<br />

Li, Xia e14511<br />

Li, Xiang 4638<br />

Li, Xiao e17504<br />

Li, Xiao-Feng e21072,<br />

e21144, e21149<br />

Li, Xiaochun 5016<br />

Li, Xiaosong 7036<br />

Li, Xiaoyu e14508<br />

Li, Xiaoyun 7531<br />

Li, Xiuli 8089<br />

Li, Xuan e16521<br />

Li, Xuefei 7535, 10527<br />

Li, Yan e14604<br />

Li, Yanchun 10598<br />

Li, Yi 5507<br />

Li, Yimei 1504<br />

Li, Yisheng 510<br />

Li, Yuelin 9105<br />

Li, Yufeng e15502<br />

Li, Zejian e18087<br />

Li, Zhenghong 9527<br />

Li, Zhiyong e13538<br />

Li, Zhong-Qian e15565<br />

Li, Zujun 5531, 5559, 5581<br />

Liaaen, Erik Dyb 7027<br />

Lian, Bin 8506<br />

Lianes, Pilar e12012<br />

Liang, Chris 3041<br />

Liang, Hong e16549<br />

Liang, Jane Q TPS7615<br />

Liang, Ji 2011<br />

Liang, Jun e14511<br />

Liang, Long 8561<br />

Liang, Meina TPS2621<br />

Liang, Min Rui e14651<br />

Liang, Raymond 8097<br />

Liao, Chun-Ta e16050<br />

Liao, Jason e18500<br />

Liao, Jay Justin 5503, 5515<br />

Liao, Kai-Ping Eric 6124<br />

Liao, Laura e15193<br />

Liao, Meilin 7519^<br />

Liao, Riqiang 7039<br />

Liao, Wei-Li e21068<br />

Liao, Winston W. P. e21097<br />

Liao, Yuqian e11538<br />

Liao, Zhongxing X. 4073,<br />

e14548, e14669<br />

Liau, Linda M. 2101<br />

Liauw, Winston S. 3504,<br />

e13585<br />

Libby, Edward N. 8010<br />

Libener, Roberta 7084<br />

Liberati, Emanuele 4572<br />

Liberman, Eliezer 1507,<br />

1564<br />

Libertini, Michela 10065<br />

Libertino, John A e15196<br />

Liboy, Idalia 8084<br />

LiButti, Daniel 9530<br />

Lich, Kristen Hassmiller<br />

6017, 6029<br />

Licht, Thomas 10031<br />

Lichtenegger, Werner 1600^,<br />

1602, 5070, 5087, 10540<br />

Lichter, Peter 1096<br />

Lichtiger, Benjamin 6622<br />

Lichtman, Stuart M. 5056,<br />

5057, 5108, 9034, 9109,<br />

9123, e18133<br />

Licitra, Lisa F. 5504^, 5508,<br />

5518, 5555, 5591,<br />

TPS5598, TPS9155<br />

Liddle, Chris 550<br />

Lie, Wen-rong 2525, e21069<br />

Lie, Yolanda 603, 608, 614<br />

Liebaert, Francois e14059<br />

Liebert, Monica 4591<br />

Liebman, Hannah M. 3594<br />

Liebner, David A. e19053<br />

Liebrich, Clemens 5044<br />

Liede, Alexander 1579, 1581,<br />

4659, e12023, e15147,<br />

e15169<br />

Liederer, Bianca M. e13114<br />

Liedke, Pedro E.R. 596<br />

Liedke, Pedro Emanuel<br />

Rubini 524<br />

Liedtke, Bernd 552<br />

Liedtke, Cornelia 552,<br />

10627, e11534<br />

Liem, Giok Siong e14085<br />

Lienard, Danielle e19026<br />

Liens, David e14650<br />

Liersch, Torsten 3600,<br />

e14161, e14162, e14167<br />

Lieser, Elizabeth Ann Teresa<br />

8599<br />

Lieu, Christopher Hanyoung<br />

3017, 3598, e14071<br />

Lievens, Johanna Caroline<br />

e13073<br />

Lièvre, Astrid e14129<br />

Liew, Choong-Chin 4106<br />

Ligeza-poisson, Catherine<br />

TPS7112<br />

Light, Emily e16014<br />

Lightsey, Judith L. TPS4136<br />

Ligibel, Jennifer A. TPS669,<br />

1026, TPS1614, 9084,<br />

e14114<br />

Ligon, Keith 2020<br />

Liguigli, Wanda 4097<br />

Lih, Chih Jian 3529<br />

Lih, Chih-Jian 5020<br />

Lilenbaum, Rogerio 7506<br />

Liliac, Ludmila e15032<br />

Lilja, Hans 4668, e15113<br />

Lillie, Sarah E 6023<br />

Liloglou, Triantafilos 5543<br />

Lim, Adrian K. e21137<br />

Lim, Allison 9576<br />

Lim, Chun Ren 4106<br />

Lim, Dean 3108, e14182<br />

Lim, Do Hyoung 4064<br />

Lim, Elgene 576<br />

Lim, Ho Yeong 4064, 4103,<br />

TPS4147, 4544^, e13044,<br />

e14617, e15064<br />

Lim, Howard John 549^,<br />

3527, 3610, 6014,<br />

e14176, e14532, e14537,<br />

e14568, e14588, e19547<br />

Lim, Hoyeong e13094<br />

Lim, Hyeong-Seok e13092<br />

Lim, Joline Sijing e13505<br />

Lim, Joo Han e13065<br />

Lim, Keith Hsiu Chin 2016<br />

751s


Lim, Kian-Huat e14584<br />

Lim, Kiat Hon 3589<br />

Lim, Megan S 9500<br />

Lim, Siew Eng 2551<br />

Lim, Soon Thye 1593, 8078,<br />

10051, e18515, e18528,<br />

e18546, e20509<br />

Lim, Sun Min e14559,<br />

e16529<br />

Lim, Tee TPS4690<br />

Lim, Wan-Teck 2598, 3098,<br />

5544, 5551, 5580, 7549,<br />

e16024, e18063^<br />

Lim, Xn-Yii 2551<br />

Lim, Yoojoo 5552<br />

Lima, Aldo Angelo Moreira<br />

e14183<br />

Lima, Alessandro e14119<br />

Lima, Carmen Silvia Passos<br />

e16041<br />

Lima, Geraldine Eltz 643,<br />

e18078<br />

Lima, Roberto Cesar e14183<br />

Lima, Vladmir C. e11058,<br />

e18078<br />

Limacher, Jean-Marc 2605,<br />

TPS4152, e19019<br />

Limareva, Svetlana<br />

Vladimirovna e15543<br />

Limaye, Maneesha R 4045<br />

Limaye, Sewanti Atul 5501,<br />

5582, 9024, e16060<br />

Limburg, Paul J. 3526<br />

Limentani, Steven A. 611<br />

Limesand, Dianne L e16513<br />

Limmroth, Christina 8024<br />

Limon, Jesus e11016<br />

Limonero, Joaquin T. e19542<br />

Limor, Sora 6525<br />

Limwongse, Chanin e14051<br />

Lin, Albert Y. e14037<br />

Lin, Alexander e16062<br />

Lin, Boris K. 2516<br />

Lin, Charles 8538<br />

Lin, Cheng-Yao 4103<br />

Lin, Chia-Chi 4524, e14576<br />

Lin, Chiao-Yun 5109<br />

Lin, Chien-Yu e16050<br />

Lin, Chihche TPS4134<br />

Lin, Chii-Dean e14037<br />

Lin, Ching-Hung 1061, 1079<br />

Lin, Daniel W. 4520, 4650<br />

Lin, Daniel e14682<br />

Lin, Derrick T. e16059<br />

Lin, Edward H. 6077,<br />

e14153<br />

Lin, Felice 6605<br />

Lin, Heather 1546, 1549,<br />

7047<br />

Lin, Ho-Sheng e16019<br />

Lin, James e15018, e15019<br />

Lin, Jay 6059<br />

Lin, Jenny J e11004<br />

Lin, Jianqing 4526, 4656,<br />

e15012<br />

Lin, Jin-Ching e16006<br />

Lin, Lilie L. e12009, e17534<br />

Lin, Lilie e18032<br />

Lin, Meng-Chih TPS7614<br />

Lin, Nancy U. 599, 605,<br />

TPS658, 3036, 6089,<br />

9084<br />

Lin, Pin-Wen e20514<br />

Lin, Po-Han e17014<br />

Lin, Simon M 2532<br />

Lin, Steven H. 4069, 4078,<br />

4086, 7043, e14547,<br />

e14548<br />

Lin, T-H e14699<br />

Lin, Tasha e16014<br />

Lin, Tongyu 5535, TPS8118,<br />

8554, e18517<br />

Lin, Xun 4542<br />

Lin, Yan 3037, 3049, 3054,<br />

6563, 8533, 8547<br />

Lin, Ying e15085, e15087<br />

Lin, Yvonne Gail e13531,<br />

e19506<br />

Lin, Z-Ping e13125<br />

Linard, Pauline 7574<br />

Linardou, Helena 573,<br />

e15040<br />

Linares, Susana 1111<br />

Linari, Alessandra 10055<br />

Linciano, Francesca e15547,<br />

e15550<br />

Lincy, Jeremie 4632<br />

Lind, Charlotte TPS5599<br />

Lind, Joline S. e18094<br />

Lind, Michael J. e13554<br />

Lind, Pehr A. 5066<br />

Lindberg, Patricia e16520<br />

Lindebjerg, Jan 3513,<br />

e14134<br />

Lindeman, Neal Ian 7588,<br />

8042, 10592<br />

Lindemann, Kristina 5008<br />

Lindemulder, Susan Joy 9546<br />

Linden, Diana 8588<br />

Linden, Hannah M. TPS3109<br />

Linden, Ola 10024<br />

Lindenberg, Maria Liza 10589<br />

Linder Stragliotto, Christina<br />

10083<br />

Linder, Albert 7056, e13061<br />

Lindet, Clothilde 10018<br />

Lindh, Birgitta 3538<br />

Lindh<strong>of</strong>er, Horst 2584, 5080,<br />

e13108^<br />

Lindner, Juha 2522<br />

Lindner, Lars 10054<br />

Lindor, Noralane M. 3567<br />

Lindquist, Deborah 547, 596,<br />

604<br />

Lindsay, Dianne Frances<br />

TPS1136<br />

Linehan, David e14584<br />

Linehan, W. Marston<br />

TPS4680<br />

Linette, Gerald P. 2525,<br />

8567<br />

Ling, Huichung Tina e11010,<br />

e11522<br />

Ling, Leona E. 10563<br />

Ling, Marilyn N 9027<br />

Ling, Winnie Hui Yee 1575,<br />

e13002, e19523<br />

Ling, Yi-He e18113<br />

Ling, Zhiqiang e21080<br />

Linga, Vijay Gandhi 6554,<br />

e18544, e18545<br />

Lingen, Mark W. 5517<br />

Lingle, Wilma L. 1083<br />

Lingua, Alejo e21025<br />

Link, Brian K. 8061, 9057<br />

Link, Charles J. 2501, 2571,<br />

4049, e19008<br />

Link, Jeanne TPS3109<br />

Link, Michael Paul 9502<br />

Link, Richard E TPS4678<br />

Linke, Rolf Gerhard 8567<br />

Linker, Charles A. 2533<br />

Linklater, Karen 7026<br />

Linn, Sabine C. 562<br />

Linnert, Mette 2069<br />

Linquist, Breanna M. 9001,<br />

9075<br />

Lins- Almeida, Thiago e12512<br />

Lion, Maeva TPS10634<br />

Liotta, Lance A. e14151,<br />

e15549<br />

Lioure, Bruno 6534, 6542<br />

Lipatov, Oleg N. 4501,<br />

e15082<br />

Lipitz Snyderman, Allison<br />

Nicole 6101<br />

Lipman, Andrew J. 7567<br />

Lipp, Eric S 2027, 2074^<br />

Lippert, Hans e14630<br />

Lippman, Scott Michael<br />

5524, 5592, 7047, 7096<br />

Lippmann, Lynne T. 5101<br />

Lipscomb, Joseph 6044,<br />

e14608, e15187<br />

Lipshultz, Steven E. 9503,<br />

9504, 9505, 9530<br />

Lipsky, Michael e15021<br />

Lipson, Doron 3533, 4649,<br />

7510, 10524, 10559,<br />

10590<br />

Lipton, Allan 501, 607, 614,<br />

7054, 7055, e14520,<br />

e15062, e15121, e15170,<br />

e17560, e19514, e21039<br />

Lipton, Jeffrey H. 6513,<br />

6596, 6604<br />

Lipton, Jeffrey Howard<br />

6505^, 6512, 6605<br />

Lipton, Lara Rachel TPS4135<br />

Lira, Cynthia 6607<br />

Lira, Ruth R 5525<br />

Lis, Christopher G. e14697<br />

Lis, Rosina 4577, 4580<br />

Lisa, Donna M. 5545^<br />

Lisano, Julie M. 7541<br />

Lisby, Steen 6584, 8019<br />

Liskova, Stefania TPS4677,<br />

e15028<br />

Lisovsky, Mikhail e14514<br />

Liss, Adam 7040<br />

Lissia, Amelia 8581<br />

List, Alan F. 3081, 6522,<br />

6586<br />

Lister, John 6620<br />

Lithwick Yanai, Gila 10528<br />

Lithwick-Yanai, Gila e14067<br />

Litière, Saskia 10602<br />

Litman, Thomas e19056<br />

Litrup, Peter e15081<br />

Litsas, Georgia 6089<br />

Litta, Pietro e12037<br />

Little, Anthony 9540<br />

Little, Melvyn 8059, e18532<br />

Little, Richard F. 8005<br />

Little, Shonda M 3528,<br />

8510, e19046<br />

Littler, John e18068<br />

Littman, Catherine 2568<br />

Litton, Jennifer Keating 537,<br />

1032, 1130, 1549, 6116,<br />

9002<br />

Litwin, Samuel e19580<br />

Litwiniuk, Maria Malgorzata<br />

e21099<br />

Litz Curry, Christine<br />

TPS3635^<br />

Litzow, Mark Robert 6614<br />

Liu, Annie Y e15582<br />

Liu, Baorui 4068, 9017,<br />

e14509, e21034<br />

Liu, Biao 3029<br />

Liu, Cameron S. 4521<br />

Liu, Chen TPS4136<br />

Liu, Chun-Yu e14516<br />

Liu, Donglin e13005<br />

Liu, Fang 3551<br />

Liu, Fengjun e14643<br />

Liu, Fenlin 4092, e14575<br />

Liu, Ge<strong>of</strong>frey 1535, 1597,<br />

3007, TPS5600, 6005,<br />

6084, 7586, 9032, 10522<br />

Liu, Glenn 4, 3101, 3103,<br />

4656, TPS4697<br />

Liu, Guohui 8017, 8033,<br />

8034, 8064, e13603<br />

Liu, Han 4604<br />

Liu, Heshan 1083, 1557,<br />

9001, 9075<br />

Liu, Hongbing e18082,<br />

e18084, e18116<br />

Liu, Hongtao 6535^, 6562<br />

Liu, Hongyu e17514<br />

Liu, Hua 5021<br />

Liu, Huan 6104<br />

Liu, Jijun 8084<br />

Liu, Jinan 6600, e15130<br />

Liu, Jingyi LBA4<br />

Liu, Jinsong e14645<br />

Liu, Jiyan 4628, 4638<br />

Liu, Li 3023<br />

Liu, Limin 10535<br />

Liu, Liming 7541<br />

Liu, Lu 10041^<br />

Liu, Luying e14049<br />

Liu, Meilian e16054<br />

Liu, Meng-zhong e18121<br />

Liu, Minetta C. 1006<br />

Liu, Mitchell 7020<br />

Liu, Nathan S. 4612<br />

Liu, Ping 8091, 8514, 9017,<br />

9081, 9083<br />

Liu, Qi 6030<br />

Liu, Ray 3077<br />

Liu, Shih-An e16006<br />

Liu, Songling 8580<br />

Liu, Stephen V. e15153<br />

Liu, Suiyang 6606<br />

Liu, Tian 9122<br />

Liu, Wanli e11091, e13021<br />

Liu, Wei 7548, 9531<br />

Liu, Wen e15124<br />

Liu, Wenchao 7548<br />

Liu, Wendy 8520<br />

Liu, Wenli 9072<br />

Liu, Xia 8042, 8106, 8107<br />

Liu, Xiangdong 2500^<br />

Liu, Xiaohe 10574<br />

Liu, XiaoMei e15191<br />

Liu, Xiaoqing 7520, 7533,<br />

7548, 7557, 7559<br />

Liu, Xiaoxia 9086<br />

Liu, Xin 6616<br />

Liu, Xinjun 4011<br />

Liu, Xueli 8089, e15084<br />

Liu, Yi 626<br />

Liu, Ying 6039<br />

Liu, Yingmiao e21038<br />

Liu, Yong 3006^<br />

Liu, Yongyu e18013<br />

Liu, Yuan 3086, 4605,<br />

e13026, e15085, e15087<br />

Liu, Zhanqi 8547<br />

Liu, Zhaoyu e14605<br />

Liu, Zhimei 1609, 4608,<br />

4612, e12042, e14525,<br />

e14627, e15063, e15096<br />

Liu, Ziyue 4586, e21016<br />

Livaudais, Jennifer C. e15513<br />

Liverani, Chiara 10615<br />

Livingston, Michael B.<br />

10003, TPS10099<br />

Livrati, Philippe e15155<br />

Liyange, Nilani TPS4153<br />

Lizard, Sarab 10507<br />

Ljungman, Gustaf 9573<br />

752s Numerals refer to abstract number


Ljungman, Mats 4591<br />

Llacer-Moscardo, Carmen<br />

LBA4003<br />

Llanos, Marta 8062<br />

Lledo, Gérard LBA3500^<br />

Llombart Cussac, Antonio<br />

10512, 10595, e11545<br />

Llombart, Antonio 1011<br />

Llorca, Cristina e11074<br />

Llorca, Laurence 2537^<br />

Llorente, Rosa M. e11074<br />

Lloreta Trull, Josep 4580,<br />

4585<br />

Lloyd, Andrew R. 9044,<br />

e15152<br />

Lloyd, Arwel e14587<br />

Lloyd, Shane 6019<br />

Lluch, Ana 1001, 1011,<br />

1015, 1074, 10500,<br />

10616, e11523<br />

Llugdar, Jose Roberto e21025<br />

Lo Nigro, Cristiana 2600<br />

Lo Re, Giovanni e15160<br />

Lo Russo, Chiara 7585,<br />

e18104<br />

Lo Russo, Giuseppe e11039,<br />

e11514, e12510, e19620<br />

Lo, Dennis YM 5511<br />

Lo, Nangi 5046<br />

Lo, Peter 7547<br />

Lo, Pu-Han 8542<br />

Lo, Shelly S. e19588<br />

Lo, Simon S. TPS1616<br />

Lo, Soo Kien 9041<br />

Lo, Yungtai e14682<br />

Loaiza, Sandra 10550<br />

Lobelle, Jean Pierre 9035<br />

Lober, William B 9008<br />

Loberiza, Fausto R. 9139<br />

Lobo, Christopher F. 7035<br />

Lobo, Francisco 5520, 8062,<br />

e15094, e18069, e18106,<br />

e21015<br />

Loboda, Andrey 3531, 3587<br />

Lobry, Celine 4018<br />

LoBuglio, Albert F. 621<br />

Lobur, David TPS8606<br />

Locati, Laura 5555<br />

Loch, Carolyn Sue 2029^<br />

Loch, Michelle Marie e21076<br />

Locher, Chrystele 1574<br />

Lock, Victoria 3028, 9542<br />

Lockart, Barbara e20001<br />

Locke, Susan C. 2021<br />

Lockhart, A. Craig 3047,<br />

LBA3501, 4062<br />

Lockhart, Albert C. e15116<br />

Lockley, Michelle 4056<br />

LoConte, Noelle K. 3101,<br />

3103, 3105, e14554<br />

Loda, Massimo F. 4521,<br />

4555, 4577, 4582<br />

Loder, Charisse e15191<br />

Lodhi, Ashutosh TPS10631,<br />

e13582<br />

Loeb, David Mark 10025<br />

Loeb, Keith 2523<br />

Loeffler, Markus 8022<br />

Loehrer, Patrick J. 7032,<br />

7106, 7107, 7108,<br />

e16557<br />

Loembe, Arsene Bienvenu<br />

e15037<br />

Loertzer, Hagen e15158<br />

Loevendorf, Marianne Begtson<br />

e19056<br />

L<strong>of</strong>freda, Massimiliano<br />

e12514<br />

Logan, Theodore 4603<br />

Numerals refer to abstract number<br />

Loggers, Elizabeth T 9139<br />

Loghin, Monica Elena 2003,<br />

2036<br />

Loghman-Adham, Mahmoud<br />

1590<br />

Logothetis, Christopher 4513,<br />

LBA4518, 4521, 4556,<br />

4558, TPS4691, e15151<br />

Logue, John P. LBA4512,<br />

4551<br />

Loh, Elwyn 2535, 2594,<br />

4005<br />

Loh, Marie 2551<br />

Loh, Mignon L. 9506, 9548<br />

Lohneis, Philipp e18552<br />

Lohrisch, Caroline A. 549^,<br />

582<br />

Loi, Sherene 507, 515<br />

Loi, Shrushma TPS660<br />

Loibl, Sibylle 522, 523, 541,<br />

556, 1023<br />

Loizos, Nick TPS10099<br />

Lok, Anna e19631<br />

Lok, Edwin e18541<br />

Lokhorst, Henk M 8019<br />

Lolkema, Martijn P.J.K. 2596,<br />

4056, e13028, e13596<br />

Lolli, Cristian 10087<br />

Lombardi, Francesca 7023<br />

Lombardi, Giuseppe 2049,<br />

e14563<br />

Lombardo, Marco 8006<br />

Lombo, Liliana 5567<br />

Lonardi, Sara 3518, 3549,<br />

3555, 3585, e14040,<br />

e14563<br />

London, Karin 1117<br />

London, Wendy B. 9516,<br />

9533, 9534, 9583<br />

Long, Alison 4056<br />

Long, Elodie 7591<br />

Long, Georgina V. 3102,<br />

8501, 8503^, 8510, 8518,<br />

8558, e19011<br />

Long, Jessica B. 1030, 1595,<br />

6628<br />

Long, Mary Ann 1529<br />

Longauer Banholzer, Maria<br />

7544<br />

Longhi, Alessandra 10026<br />

Longhitano, Laura e15544<br />

Longmate, Jeff 2538<br />

Longo, Federico 10546<br />

Longo, Flavia LBA7501,<br />

9133, e14082, e14096<br />

Longo, Marina e18103<br />

Longo, Valter e13531<br />

Longpre, Lara TPS4152,<br />

e14566<br />

Longvert, Christine 8540<br />

Lonial, Sagar 2576, 8012,<br />

8017, 8020, 8033, 8035,<br />

8086, 8100, 8101,<br />

TPS8112, TPS8113,<br />

e18553, e18569, e18572^<br />

Lonneux, Max 5519<br />

Look Hong, Nicole Jannelle<br />

10058<br />

Look, A. Thomas e13586<br />

Look, Maxime P 10504<br />

Look, Regan M. 9019<br />

Loong, Herbert e14085<br />

Loos, Walter J. 2593, 4132<br />

Lopa, Samia H. 3545<br />

Lopatin, Margarita 3512,<br />

3626<br />

Lopes, Ademar 10069,<br />

e14119<br />

Lopes, Antonio de Barros<br />

e14597<br />

Lopes, Gilberto de Lima<br />

e15045, e16525<br />

Lopes, Gilberto e15107<br />

Lopes, Vrishali 5100<br />

Lopez Ben, Santiago e14120<br />

Lopez Calderero, Iker 7544<br />

Lopez Criado, M Pilar<br />

TPS4672, e19023<br />

Lopez de Ceballos, Maria<br />

Helena e11545<br />

Lopez Gomez, Miriam 9073<br />

López Jiménez, Antonio<br />

e18049<br />

Lopez Ladron, Amelia e11075<br />

Lopez Lincuez, Maria Emilia<br />

10074<br />

López López, Rafael 530<br />

Lopez Ruitti, Paula e14109<br />

Lopez Vilaro, Laura 1120,<br />

10555<br />

Lopez Vivanco, Guillermo M<br />

7095<br />

Lopez, Antonio 4587<br />

Lopez, Christian TPS668,<br />

TPS4695<br />

Lopez, Ines e14057<br />

López, José Luis e14002<br />

Lopez, Jose e15103<br />

Lopez, Katrina e15500<br />

Lopez, Lenny 9004<br />

López, Maria e16029<br />

Lopez, Rafael 10534,<br />

e14733, e19505<br />

Lopez, Roberto e11035,<br />

e11527<br />

Lopez, Salvatore 5093,<br />

e15547, e15551, e15553<br />

López-Basave, Horacio Noe<br />

e15527<br />

López-Brea, Marta F.<br />

TPS4672<br />

López-Casas, Pedro P. 3066,<br />

3068<br />

Lopez-Chavez, Ariel TPS7609<br />

Lopez-Crapez, Evelyne 10505<br />

Lopez-Diaz, Christine 547<br />

Lopez-Gomez, Miriam 1570,<br />

e11028<br />

Lopez-Gonzalez, Ana e11035,<br />

e11527, e12015, e17515,<br />

e18521<br />

Lopez-Guerrero, Jose Antonio<br />

10079, e15575<br />

Lopez-Ilasaca, Marco e11066<br />

López-López, Carlos 3571<br />

Lopez-Macias, Constantino<br />

e15584<br />

Lopez-Martin, Jose A. 2583,<br />

10052, e19023<br />

Lopez-Mozos, Fernando<br />

e14589<br />

Lopez-Peñalver, Jesus 1041<br />

López-Picazo, José María<br />

e15041, e18008<br />

Lopez-Pousa, Antonio 5590,<br />

5595, 10052, 10079<br />

López-Ríos, Fernando 602,<br />

3066, 3068, 4040,<br />

e11006<br />

Lopez-Trabada Ataz, Daniel<br />

e12003<br />

Lopez-Vega, Jose Manuel<br />

TPS653, 1001, 10512,<br />

10595<br />

Lopez-Vivanco, Guillermo<br />

7011, e11525, e17522,<br />

e18031, e21009<br />

Loprinzi, Charles L. 1557,<br />

9001, 9075<br />

Loquai, Carmen 8505, 8517,<br />

e19031<br />

Lora, David 642<br />

Lorch, Anja 4597, 4600,<br />

e15026<br />

Lorch, Jochen H. 5501,<br />

5579, 5582, e16060<br />

Lord, Christopher J. 3050,<br />

5035<br />

Lord, Raymond S. 9141<br />

Lord, Sally 5081<br />

Lordick, Florian 3535,<br />

e13095^, e13097^,<br />

e13108^, e14167<br />

Loren, Alison W. 6579,<br />

e18529, e18540<br />

Lorence, Robert M. LBA7500<br />

Lorente, David 10082<br />

Lorente, Gustavo A. 6585<br />

Lorenz, Mario TPS4138<br />

Lorenz, Ralf 1602, 5044<br />

Lorenz, Wilfried e16520<br />

Lorenzen, Sylvie 4080<br />

Lorenzi, Elena 2580, e21139<br />

Lorenzo-Patiño, Maria J<br />

e21002<br />

Loretelli, Cristian e14086,<br />

e14561, e15074<br />

Lorf, Thomas e14167<br />

Lorigan, Paul 8502^,<br />

TPS8605, e19010<br />

Lorimer, Ian 3515<br />

Loring, Jeanne F. 10539<br />

Loriot, Virginie 3633, 10505<br />

Loriot, Yohann 4540, 4554,<br />

4574, 4621<br />

Lorsbach, Michael 8052<br />

Lortholary, Alain 5018<br />

Lorusso, Domenica LBA5003,<br />

5059, 5087, 5096<br />

LoRusso, Patricia 528,<br />

TPS663, TPS668, 2534,<br />

2566, 2579, TPS2618,<br />

3029, 3059, 3062, 4639,<br />

e19048^<br />

Lorusso, Vito e18048<br />

Los, Maartje 4539<br />

Losa, Ferran 3571, e18030<br />

Losa, Raquel e18044<br />

Loscalzo, Matthew 9048<br />

Losem, Christoph 3<br />

Losonczy, Gyorgy e13113<br />

Losurdo, Agnese 623<br />

Lotan, Yair 4591, 4616,<br />

TPS4678<br />

Lotia, Mitesh e17019<br />

Lotz, Jean-Pierre 10035<br />

Lotze, Michael T. 3019<br />

Lou, CaiJin e17512<br />

Lou, Feiran 7043<br />

Lou, Pei-Jen e16050<br />

Lou, Yuqing 1551, e17551<br />

Louahed, Jamila 7013, 7056,<br />

e13061<br />

Loucaides, Eileen 6115<br />

Loucks, Aimee e15154<br />

Loud, Peter e19041<br />

Loughran, Thomas P. 6590,<br />

6616<br />

Louhimo, Riku 8074<br />

Louie, Alexander V. 6130<br />

Louie, Arthur Chin 6525,<br />

6601<br />

Loundou, Anderson 8555<br />

Lounsbury, Heather 8032<br />

753s


Loupakis, Fotios 3518, 3540,<br />

3546, 3549, 3555, 3585,<br />

3597, 3604, 3622, 4026,<br />

4088, 4096, e11018,<br />

e13057, e14031, e14034,<br />

e14040, e14052<br />

Lourenco, Gustavo Jacob<br />

e16041<br />

Loussouarn, Delphine 2082<br />

Louvet, Christophe<br />

LBA3500^, 3506, 4032<br />

Louwman, Marieke J. 8039<br />

Lovat, Francesca 1007<br />

Lovat, Katherine e19598<br />

Lovecchio, John L. 5056,<br />

5057<br />

Lovecek, Martin e14556<br />

Lovly, Christine Marie 7094,<br />

7532<br />

Low, Eng Joo 4106<br />

Low, Li shan 4100^<br />

Low, Walter 2546, 3070<br />

Lowe, Ann M. 6508<br />

Lowe, Robert 10550<br />

Lowe, Val J. e21006<br />

Lowery, Jan 3567<br />

Lowery, Maeve A 4057<br />

Lowy, Andrew M. 4019,<br />

e14069<br />

Lowy, Israel 2562<br />

Loya, Asif e16043<br />

Loyer, Evelyn 3544<br />

Lozada, Claudia Patricia 2531<br />

Lozano, Guillermina 10506<br />

Lozano, Juan Jose 3553<br />

Lozano, Maria D. 8572,<br />

e15041, e18008<br />

Lozano, Nicolas TPS10634<br />

Lozanski, Gerard 6526<br />

Lu, Charles 5589, 5592,<br />

9118, 9518<br />

Lu, Dongrui (Ray) 4118<br />

Lu, Gary 8040<br />

Lu, Haolan TPS7611<br />

Lu, He e21156<br />

Lu, Hongyang e17512<br />

Lu, Jerry e21047<br />

Lu, Jiade Jay 2016<br />

Lu, Jianfeng 5072<br />

Lu, Jiangyang e18087<br />

LU, Jinlong 5558<br />

Lu, Karen H. CRA1505,<br />

1513, 1518, 1519, 1520,<br />

1546, 5027, e13516<br />

Lu, Kelley 1562<br />

Lu, Lanchun 9544<br />

Lu, Ligong 4008, e14605<br />

Lu, Lingeng 5099, e15581<br />

Lu, Michael W LBA1<br />

Lu, Ming e14604<br />

Lu, Minqiang 4008<br />

Lu, Ning e14511<br />

Lu, Shir Kiong 10060<br />

Lu, Shun 7091, 7520<br />

Lu, Sunny e11068<br />

Lu, Tai-xiang 5535<br />

Lu, Xian 7601<br />

Lu, Xiang e11003<br />

Lu, Xiaomin 9546<br />

Lu, Xuyang e13045<br />

Lu, Yen-Shen 1061, 1079,<br />

e11022<br />

Lu, You 4073, e14648,<br />

e18033, e18086<br />

Lu, Yu-Shiang 6078<br />

Lu, Yunhua e11567, e11568<br />

Lu, Zhen-Yi e17512<br />

Lu, Zhihao e14604<br />

Lu, Zhiqiang Kevin e19639<br />

Lu-Emerson, Christine 2010<br />

Luaces, Patrcia Lorenzo 2515<br />

Luan, Jennifer 3528, e19046<br />

Luan, Shen 3015, e14593<br />

Luan, Wei e14583<br />

Lub-de Hooge, Marjolijn N.<br />

10581<br />

Lubbe, Dirk 5007<br />

Lubben, Jade Marie 8010<br />

Lubner, Sam Joseph 3101,<br />

3103, e14554<br />

Lubow, Meredith A. 2040<br />

Lucas, Jared 4669, e15207<br />

Lucca, Joan Vern 7099,<br />

e19637<br />

Lucchesi, Sara 629, 3518,<br />

3546, 3549<br />

Lucchini, Eleonora e14661<br />

Lucci, Anthony 594, 1102,<br />

TPS10631, e13582,<br />

e21107<br />

Lucci, Joseph A. 5065<br />

Lucey, James e19601<br />

Luciano, Antonio e11052,<br />

e13539<br />

Luciano, Jason e14184<br />

Ludkovski, Olga 1036<br />

Ludovini, Vienna 3521,<br />

e14042, e18021, e18023,<br />

e18101, e21018, e21094,<br />

e21096<br />

Ludwig, David A. 9565<br />

Ludwig, Elizabeth e18522<br />

Ludwig, Joerg 2522, 3530<br />

Ludwig, Joseph A. 10003,<br />

10021, 10071, e19520<br />

Ludwig, Justin 6000<br />

Luebbe, Kristina 556,<br />

TPS661<br />

Luebke, Aaron Lynn e13548<br />

Lueck, Hans-Joachim 556<br />

Luecke, Klaus 10536, 10537<br />

Lueftner, Diana e14140<br />

Lueke, Matthias 2054<br />

Luengo, Maria Isabel e11024<br />

Luengo-Gil, Gines e11024<br />

Lueza, Beranger 10556<br />

Luger, Selina M. 6520, 6579<br />

Luger, Thomas 4034<br />

Lugowska, Iwona A. 10538<br />

Lugtenburg, Pieternella 8002,<br />

8039, e16526<br />

Luini, Alberto 519<br />

Luis, Ines Maria Vaz Duarte<br />

599, 6089<br />

Luisser, Irmard 542<br />

Lujan, Ramona 4096<br />

Lukawy, Jacek TPS6643^<br />

Luke, Jason John 2606,<br />

8549, e13588<br />

Luketich, James D. 7071,<br />

7074, e14689<br />

Luknic, Alice Marie TPS657<br />

Luley, Kim Barbara 4083,<br />

4090<br />

Lum, Christopher A. 1599,<br />

e19051<br />

Lum, Lawrence G. e13089<br />

Lum, Lawrence G e13124<br />

Lumba, Sumit e19042<br />

Lumbroso, Jean 9560<br />

Lumbroso, Livia e13017<br />

Lumbroso-Le Rouic, Livia<br />

9538<br />

Lumgair, Heather e17543<br />

Luminari, Stefano 8006<br />

Luna Fra, Pablo 10079<br />

Luna, Hermenigilda 6108<br />

Luna, Yesenia L 4666,<br />

e15186<br />

Lunceford, Jared 3531<br />

Lund, Bente 5052<br />

Lund, Mary Jo B. 6052<br />

Lund, Peder 2514<br />

Lundeberg, Joakim 10521<br />

Lundgren, Lotta 10024<br />

Lungershausen, Juliane 6058<br />

Lunning, Matthew Alexander<br />

8060, 8063<br />

Luo, Jialing e14049<br />

Luo, Jingqin 503, TPS664<br />

Luo, Rongcheng e18033<br />

Luo, Shujun 10539<br />

Luo, Suhong 4594<br />

Luo, Yang e11025, e11538<br />

Luo, Yi 7091, 7520<br />

Luo, Yuling 5543<br />

Luo, Zhongyao e11557<br />

Luong, Richard 2513<br />

Luong, Tu Vinh 4121<br />

Lupi, Cristiana 3521, 3585<br />

Lupichuk, Sasha M. 1036,<br />

e14073<br />

Lupinacci, Lorena 10074<br />

Luppe, Denise 4098<br />

Luppi, Mario 6531<br />

Luptrawan, Anne 2084<br />

Luque Caro, Raquel TPS4672<br />

Luque, Manuel 2539,<br />

e15001<br />

Lurain, John Robert e15523<br />

Lusa, Amanda 7050<br />

Lusebrink, Jessica e18000<br />

Lusky, Monika TPS4152<br />

Lust, John Anthony 8096,<br />

8105, TPS8116<br />

Lustberg, Maryam B. e21124<br />

Lustig, Robert A. e12508<br />

Luthra, Rajyalakshmi 3106,<br />

10510, e13595<br />

Luthra, Rakesh e11063<br />

Lutke Holzik, Martijn 10554<br />

Lutrino, Stefania Eufemia<br />

3612, TPS3637, e14040<br />

Lutsyk, Miroslav e18511<br />

Luttgen, Madelyn 4102,<br />

e21074<br />

Lutz, Manfred P. 4053,<br />

TPS4140<br />

Luu, Dai Chu Nguyen e18018<br />

Luu, Thehang H. 1010,<br />

1035, 1070, 3108<br />

Luu-The, Van 10583<br />

Luukkaa, Marjaana TPS4696^<br />

Luvero, Daniela 5093, 5094,<br />

e15547, e15550, e15551,<br />

e15553<br />

Luzi Fedeli, Stefano 4084<br />

Lv, Chentao e14575<br />

Lv, Fangfang e11568<br />

Lv, Jima e14511<br />

Lv, Meijun 7091<br />

Lv, Yanling e18116<br />

Lwin, Thuya Win 3002<br />

Lwin, Zarnie 6087<br />

Ly, Janey 10574<br />

Lybeert, Marnix L. 8039<br />

Lüchtenborg, Margreet 7026<br />

Lyerly, H. Kim 2585, 10563<br />

Lyerly, Susan 6524^<br />

Lyford-Pike, S<strong>of</strong>ia 5506<br />

Lyle, Stephen 8543<br />

Lyman, Gary H. 1062, 1525,<br />

6129, 9135<br />

Lynch, Caroline Dorothy 5107<br />

Lynch, Henry T. 1538<br />

Lynch, Julie Ann e18067<br />

Lynch, Kristian 1608<br />

Lynch, Siobhan P. 1029,<br />

1104<br />

Lynch, Thomas James 7010,<br />

TPS7611<br />

Lynn, Audrey 1569<br />

Lyons, John F. 3028, 9542<br />

Lyons, Roger M. 6624<br />

Lyons, Tomas e19059<br />

Lypas, Georgios 1547<br />

Lüpfert, Christian e13594<br />

Lyss, Alan P. CRA1002<br />

Lyulko, Alexey A. 4501<br />

M<br />

M. Tirado, Oscar 3096<br />

Ma, Ai-Hong e13526<br />

Ma, Brigette 5511, e14085,<br />

e14528<br />

Ma, Cynthia X. 534, TPS664,<br />

1010, 1048, 3047<br />

Ma, Daniel 2031<br />

Ma, Fei e11025, e11538<br />

Ma, Jianhui 4628<br />

Ma, Junxun 7036<br />

Ma, Meili e12013<br />

Ma, Michelle W. 8557, 8565,<br />

8589, e19003<br />

Ma, Patrick C. e17563,<br />

e18046, e18085<br />

Ma, Shenglin e17527<br />

Ma, Shuo 8032<br />

Ma, Suyong e19574<br />

Ma, Tian 7022, e13567<br />

Ma, Wen Wee 3024, 3582,<br />

4117, 4545, 8519,<br />

e13006, e14659<br />

Ma, Xiao-Jun 5543<br />

Ma, Xiaomei 1595, 6628<br />

Ma, Xingqun e18116<br />

Ma, Yifei 6010<br />

Ma, Yu e18051<br />

Ma, ZhenLu e14716<br />

Maale, Gerhard 10037<br />

Maartense, Eduard 1080<br />

Maas, Paige 1521<br />

Maass, Nicolai 556, e13503<br />

Maazaz, Khalid e14726<br />

Maazen, RWM Van Der 8039<br />

Mabjeesh, Nicola J. 1564<br />

Mabro, May 2537^,<br />

LBA3500^, 3506<br />

Mac Mathuna, Padraic M.<br />

e12031<br />

Macak, Dusan e15027<br />

Macarulla, Teresa 3014,<br />

4053<br />

Maccario, Rita e13103<br />

Maccaroni, Elena e14086,<br />

e14561, e14663, e15074<br />

Macchini, Marina e14647<br />

Macciò, Antonio e19634<br />

MacConaill, Laura E. 5011<br />

MacDiarmid, Jennifer e13054<br />

Macdonald, David R. 2051<br />

Macdonald, John S. 4010<br />

MacDonald, Lisa 2588<br />

MacDonald, Neil e18098<br />

Macdonald, Orlan Kenneth<br />

e14507<br />

MacDonald, Shannon M.<br />

9585<br />

Macdonald, Theresa 4649<br />

MacDonald, Tobey 9519<br />

Mace, Joseph TPS8115<br />

Macedo, Gabriel de Souza<br />

e13546, e21042<br />

Macedo, Omar e19618<br />

Macfarlane, David e13128<br />

754s Numerals refer to abstract number


Macgregor, Tom e13587<br />

Machado Frascari, Luciana<br />

e19528, e19584<br />

Machado, Isidro e14041<br />

Machado, Karime Kalil<br />

e15562<br />

Machalekova, Katarina<br />

e15027<br />

Machavoine, Jean-Luc 9080<br />

Machida, Nozomu 4046<br />

Machiels, Jean-Pascal H.<br />

4583, 4622, 5505, 5519,<br />

TPS5598, e13605,<br />

e14060, e15115<br />

Machiels, Jean-Pascal 2583<br />

Machiorlatti, Michael e17012<br />

Machtay, Mitchell TPS1616,<br />

5530<br />

Maciá, Sonia 4587,<br />

TPS4674, e18053<br />

Macias, Ismael e15159<br />

Maciejewski, Jaroslaw P.<br />

6521<br />

Maciejewski, John Joseph<br />

6552<br />

Maciejewski, Paul K 9037,<br />

9146<br />

Macis, Debora 519<br />

Mack, Philip C. 4511, 4547,<br />

4663, 7018, 7552, 7582,<br />

10503<br />

Mack, Philip C 7005, 7083,<br />

7517, e13550, e17537<br />

Mackall, Crystal 9545<br />

Mackay, Helen 1597, 5001,<br />

5010, 5019, 5026, 5105,<br />

e14524<br />

MacKenzie, Mary J. 3013,<br />

4536, 4538, TPS4683<br />

Mackey, John Robert e14625<br />

Mackillop, William J. 1583,<br />

3632<br />

Mackley, Heath B. 7055<br />

Macklin, Eric e18520<br />

Macklis, Roger M. 10623<br />

Mackman, Nigel e14505<br />

MacLean, Anthony e14073<br />

MacLeod, Nicholas James<br />

e15037<br />

MacNeil, Mary Valeria 2051<br />

Macon, William R 9057<br />

MacVicar, Gary R. 4506<br />

Madadi, Anusha Reddy<br />

e16555<br />

Madajewicz, Stephan e14019<br />

Madan, Ravi A. 2526,<br />

TPS2617, 3043, 4569,<br />

TPS4701, 10589, e13122<br />

Maddala, Tara 4560<br />

Madden, Erik e18549<br />

Madden, Stephen F. 617,<br />

633<br />

Maddi, Rahul Narayan 6554,<br />

e18544, e18545<br />

Maddocks, Kami J. 6508,<br />

e13568<br />

Madeddu, Clelia 9006,<br />

e19634<br />

Madero, Rosario 3618,<br />

10547<br />

Madhuri, Thumuluru Kavitha<br />

e15517<br />

Madhusudan, Srinivasan 1014<br />

Madhusudanannair, Vinu<br />

3106<br />

Madi, Ayman 2552<br />

Madkour, Ashraf e17556<br />

Madoz, Juan 5520, e15094,<br />

e18069, e18106, e21015<br />

Numerals refer to abstract number<br />

Madroszyk, Anne e19539<br />

Maeda, Atsuyuki 3607<br />

Maeda, Lauren Shizue 8060<br />

Maeda, Patricia Hikari<br />

e11059<br />

Maehara, Yoshihiko 3558,<br />

e13090, e14033, e14536,<br />

e14601, e14602, e14722<br />

Maekawa, Hiroshi e14139<br />

Maemondo, Makoto 7086,<br />

7561, 7563, 7565, 7571,<br />

e18129<br />

Maeng, Chi Hoon 644, 7104,<br />

e13094, e16003, e18148<br />

Maenpaa, Johanna e15564<br />

Maerten, Angela 4023<br />

Maertens, Johan TPS6637<br />

Maestri, Marcello e13103<br />

Maestu, Inmaculada 1531<br />

Maetzold, Derek 7106, 8543<br />

Magagna, Silvia e13057<br />

Magargal, Wells Wrisley<br />

e21131<br />

Magasi, Susan 9056<br />

Mager, Donald E. e14690<br />

Maggard Gibbons, Melinda<br />

6057<br />

Maggioni, Angelo e19036<br />

Maggiori, Leon e14158<br />

Maggiorotto, Furio 627<br />

Magi-Galluzzi, Cristina 4560<br />

Magidson, Jay 4516<br />

Magley, Andrea 10088<br />

Magliocca, Kelly R. 5560<br />

Magliocco, Anthony M. 2001,<br />

e21083<br />

Magliocco, Tony e21044<br />

Magneux, Celine 641<br />

Magnin, Valerie Florence<br />

2048<br />

Magometschnigg, Heinrich<br />

10570<br />

Magoon, Daniel 9548<br />

Magoski, Nadine M 3504<br />

Magremanne, Michele 5519<br />

Magro, Ana 1074<br />

Maguire, Aoife e21024<br />

Maguire, Roma e19617<br />

Mahadevan, Daruka 3028,<br />

8032<br />

Mahajan, Anita 2001, 9585<br />

Mahal, Lara K e19018<br />

Mahale, Parag e17016<br />

Mahalingam, Devalingam<br />

3073, 3078, e15116<br />

Mahamat, Abakar e15574<br />

Mahan, Meredith e15174,<br />

e15512<br />

Mahapatra, Manoranjan<br />

e17000<br />

Mahaseth, Hemchandra<br />

e14512, e14614<br />

Mahdavi, Khosrow 2569<br />

Mahe, Isabelle 1580<br />

Maher, Michael A. 1543,<br />

e19601<br />

Maher, Richard e19042<br />

Mahesh, Bandemegal e11027<br />

Maheshwari, Neelabh 7040<br />

Maheswaran, Shyamala 4566<br />

Mahlberg, Rolf 4083<br />

Mahler-Araujo, Betania 544<br />

Mahmud, Nadiya 10550<br />

Mahner, Sven 5001, 5005,<br />

5007, 5031, 5034, 5058,<br />

5071, 5087, e15520<br />

Mahon, Brett 7067, e18117<br />

Mahon, Garry e14180<br />

Mahoney Shannon, Kristen<br />

e16510<br />

Mahoney, Douglas W. e15132<br />

Mahoney, John Francis 4655,<br />

4667<br />

Mahoney, Martin 1529<br />

Mahoney, Michelle R. 3514,<br />

3617<br />

Mahoney, Sandy 8514,<br />

e19014, e19039<br />

Mai, Phuong L. 1519<br />

Maia, Joyce Maria L. e14680,<br />

e14728<br />

Maiello, Evaristo e21092<br />

Maier, Gal e15136<br />

Maier, Sabine TPS7113,<br />

TPS7611<br />

Maier, Thomas e14099<br />

Maillard, Natacha 6542<br />

Maillard, Sophie e15535<br />

Maillet, Denis e15182<br />

Mailliez, Audrey e11012,<br />

e11049, e11500<br />

Maimon, Natalie 4564, 4619,<br />

e15058, e18108<br />

Maimonis, Peter J. e21091<br />

Maindrault-Goebel, Frederique<br />

e19623<br />

Maines, Francesca 4541<br />

Mainwaring, Paul N.<br />

LBA4518, 4558, TPS4683,<br />

TPS4692^, TPS4696^<br />

Maio, Michele 8502^, 8513,<br />

8529<br />

Maiolino, Angelo 8018^,<br />

8095<br />

Maiolino, Piera e14130<br />

Maire, Frederique 4133<br />

Mais, Anna 4115<br />

Maison-Blanche, Pierre<br />

e15115<br />

Maisonneuve, Patrick 1500<br />

Maitah, Main e21057<br />

Maithel, Shishir Kumar<br />

e14169, e14170, e14692<br />

Maiti, Baidehi e15095<br />

Maitland, Michael L. e13106<br />

Maitra, Radhashree e14019<br />

Maiya, Arun 5526<br />

Maizel, Abby e13124<br />

Majem, Margarita 1531,<br />

7522, 7542, e12000,<br />

e12012, e18057, e18130,<br />

e18131<br />

Majeske, Cindy e12500<br />

Majeti, Ravindra 2523<br />

Mak, Milena Perez e21040<br />

Makarenko, Serge 9078<br />

Makarov, Danil V. 6019<br />

Makarov, Vladimir 10587<br />

Makarova, Olga V e21079<br />

Makatsoris, Thomas 5033<br />

Makeyev, Yan G. 5016<br />

Makhson, Anatoly 7512<br />

Maki, Robert G. 10001,<br />

10003, 10004, LBA10008,<br />

10013, 10021, 10022,<br />

10057, 10097, TPS10103,<br />

e13600^<br />

Makimoto, Atsushi 9574<br />

Makin, Guy 9542<br />

Makino, Haruhiko 10624<br />

Makino, Hirochika e14624<br />

Makiura, Daisuke e17020<br />

Makiya, Monica 10074<br />

Makiyama, Akitaka 3045<br />

Makrilia, Nektaria e17508<br />

Makris, Andreas TPS665,<br />

1066<br />

Makuuchi, Masatoshi 4104<br />

Mala, Carola 1092<br />

Malafa, Mokenge Peter<br />

e14715<br />

Malagola, Michele 6531<br />

Malamos, Nikolaos 583<br />

Malamou-Mitsi, Vassiliki<br />

10575<br />

Malangone, Elisabetta<br />

e14028, e14030<br />

Malapelle, Umberto e13509<br />

Malasse, Stephanie 7575<br />

Malatesta, Theresa Maria<br />

e19507<br />

Malats, Nuria e12033<br />

Malaurie, Emmanuelle<br />

LBA5003<br />

Malchiodi, Michael e21122<br />

Maldonado, Antonio 1111<br />

Maldonado, Xavier e15199^<br />

Maleddu, Alessandra 10087<br />

Malek, Kamel TPS6643^<br />

Malekzadeh, Katty 9517<br />

Malfertheiner, Peter TPS4148<br />

Malhi, Vikram e13114<br />

Malhotra, Jyoti CRA9513<br />

Malhotra, Usha e14712<br />

Malik, Hassan Zakria 3613<br />

Malik, Laeeq e14703<br />

Malik, Monica 9055, e12501<br />

Malik, Prabhat Singh e17505,<br />

e17511<br />

Malik, Shakun M. 3074<br />

Malik, Zafar I e15112,<br />

e15128, e15135, e18068<br />

Malin, Jennifer L. e14610<br />

Malin, Jennifer 6105<br />

Malin, Jessica e14175<br />

Malinoski, Frank J e16053<br />

Malinowski, Paige 4027<br />

Malisic, Emina e12020<br />

Malka, David 4018, 4032,<br />

9026^, e14158<br />

Malkowski, Bogdan 8077<br />

Mallet, Stéphanie 8555,<br />

8568<br />

Mallick, Atrayee Basu e14019<br />

Mallick, Rajiv 9056<br />

Mallick, Ranjeeta 10620<br />

Mallick, Supriya 2072<br />

Mallick, Ujjal e13522<br />

Mallikarjun, Priya e11097,<br />

e12004<br />

Mallmann, Peter 3044<br />

Mallon, Robert e13576<br />

Malmgren, Judith April 6560<br />

Malmstrom, Robert e15154<br />

Malone, Michael D e15141<br />

Maloney, David G. 8001<br />

Maltese, Mariangela 4097<br />

Maltoni, Roberta e11054<br />

Maluccio, Mary A. e14683<br />

Malvehy, Joseph 8578<br />

Maly, Joseph 609<br />

Malysa, Agnes 7593<br />

Malysz, Jozef 8090<br />

Mambo, Elizabeth 3630<br />

Mambrini, Andrea LBA4001,<br />

4017, 10611<br />

Mamedov, Alexandre e15180<br />

Mamet, Rizvan e18018<br />

Mamon, Harvey J. 4021<br />

Mamorska-Dyga, Aleksandra<br />

6123<br />

Mamounas, Eleftherios P.<br />

LBA506, LBA1000<br />

Mamounas, Eleftherios P 616,<br />

TPS657, 1025, TPS1142<br />

Mamtani, Ronac 1503<br />

755s


Man, Shan e11061, e15177^<br />

Manaloor, Elizabeth 8108<br />

Manasanch, Elisabet E. 8088,<br />

8104, e18568<br />

Manca, Antonio e20512<br />

Mancao, Christoph TPS3637<br />

Mancari, Rosanna 5090<br />

Manceau, Gilles e14059<br />

Mancenido, Noemi 1570<br />

Manches, Olivier 2570,<br />

e21081<br />

Mancini, Andrea e15200<br />

Mancini, Julien 10078<br />

Mancini, Maria Laura e18072<br />

Mancini, Maria 5096<br />

Mancini, Raffaello e14082<br />

Mancuso, Andrea Petricca<br />

e19033<br />

Mancuso, Maurizio 7084<br />

Mancuso, Patrizia 2083<br />

Mandadi, Mounika e16517<br />

Mandal, Rakesh e17005<br />

Mandala, Mario e19549,<br />

e19612<br />

Mandalia, Amar e16001<br />

Mandat, Tomasz 603<br />

Mandel, Nil Molinas e19024<br />

Mandeli, John P e15191<br />

Mandolesi, Alessandra<br />

e14086, e14561, e21070<br />

Mandrekar, Sumithra J. 6133,<br />

7555, 7605<br />

Mandrioli, Anna 10087<br />

Mane, Anupama Dutt e11506<br />

Manegold, Christian<br />

TPS7109, TPS7613^<br />

Manente, Liborio e19033<br />

Manente, Paolo e19595<br />

Manenti, Luigi TPS3637,<br />

5522, 7544<br />

Manetsch, Gabriela e19648<br />

Maneval, Daniel C. 2579<br />

Maneval, Edna Chow 4644,<br />

TPS4697<br />

Maney, Todd 4013<br />

Mangardich, Antranik 6123<br />

Mangili, Giorgia 5059<br />

Maniar, Manoj 3081<br />

Maniar, Tapan TPS670<br />

Manieh, Moustafa e20514<br />

Manikhas, Alexey 531, 1059,<br />

e11086, e19609<br />

Manji, Arif e13001<br />

Manji, Gulam Abbas 6615,<br />

e14185<br />

Manjili, Masoud 10565<br />

Manka, Lukas e15195<br />

Mank<strong>of</strong>f, David A. 1088,<br />

TPS3109, TPS10635<br />

Manley, Paul W. 10030<br />

Mann, Bhupinder Singh<br />

e16569<br />

Mann, Elaina e13028<br />

Mann, Susan e16515,<br />

e16559<br />

Manneh, Ray e15033,<br />

e15552<br />

Mannel, Robert S. TPS5114<br />

Manning Duus, Elizabeth<br />

e19577<br />

Manning, Robert J 8042,<br />

8107, e18575<br />

Mano, Max S. e11014,<br />

e11087, e11513<br />

Mano, Yohei e14536<br />

Manoharan, Prakash e14015<br />

Manola, Judith 4500, 6080,<br />

9016<br />

Manolis, Evangelos e21098<br />

Manrique, Gonzalo e19007<br />

Mansfield, Aaron Scott<br />

e21112<br />

Mansfield, David 2530<br />

Mansi, Janine 1094<br />

Mansmann, Ulrich Robert<br />

3588, 3603, 4023, 10054,<br />

e14043<br />

Manso, Luis 642, 1011,<br />

e11074, e11081, e11535,<br />

e15552, e21088<br />

Manson, JoAnn E 1501<br />

Manson, Stephanie 10061<br />

Mansoor, Atiya 10011^<br />

Mansoor, Maryah e18574<br />

Mansoor, Wasat LBA4000,<br />

LBA4001, TPS4143<br />

Mansour, Marc 2588<br />

Mansourbakht, Touraj 9026^<br />

Mansouri, David 3599<br />

Mansouri, Kambiz 1059<br />

Mansouri, Ladan 6557<br />

Mansouriah, Merad 5569<br />

Mansueto, Giovanni 9133,<br />

e12514, e18065, e18072<br />

Mansutti, Mauro 1044, 1073,<br />

e13058, e13070<br />

Manta, Carmen e17533<br />

Mantovani, Giovanni 5513,<br />

9006, e13098, e14577,<br />

e19634<br />

Mantovani, Lorenzo e14113<br />

Mantripragada, Kalyan C<br />

4098<br />

Mantry, Parvez e14581<br />

Manu, Michell 7103<br />

Manuaba, Tjakra e11022<br />

Manuel, Manuarii 10562<br />

Manuel, Megan 6524^<br />

Manuguerra, Giovanna 623<br />

Manuyakorn, Ananya e14051<br />

Manyam, Madhavi e14045<br />

Manzano, Hermini e14041<br />

Manzano, Jose Luis 3571,<br />

e14671<br />

Manzoni, Marco 1604<br />

Manzoni, Mariangela e13103<br />

Manzullo, Ellen F. 9072<br />

Mao, Hui e13110, e14734<br />

Mao, Jun e19531<br />

Mao, Li Li 8506, e15051,<br />

e15052<br />

Mao, Li 5524<br />

Mao, Mao 3509<br />

Mao, Weimin e17512,<br />

e18013, e21080<br />

Mao, You-Sheng e14502<br />

Mapara, Markus Y. 8081<br />

Mapelli, Sergio 10022<br />

Maples, W. e14610<br />

Maples, William James 5544<br />

Maquilan, Genevieve e12508<br />

Mar, Victoria 8520<br />

Marabelle, Aurelien 2513,<br />

e13555<br />

Marabese, Mirko LBA7501<br />

Maragkos, Charilaos e17557<br />

Maragulia, Jocelyn 8069<br />

Maraldo, Maja Vestmo<br />

e18527<br />

Marandino, Ferdinando 1050<br />

Marasco, Antonella 8513<br />

Maraval-Gaget, Raymonde<br />

9011<br />

Marban, Patrick 3002<br />

Marcacci, Giampaolo 8066<br />

Marcaillou, Charles e14059<br />

Marcato, Paola e21090<br />

Marcello, Norina 2057<br />

Marchal, Frederic 10078,<br />

10517<br />

Marchello, Benjamin T. 3545<br />

Marchetti, Antonio e14042,<br />

e18021<br />

Marchetti, Paolo e15185<br />

Marchetti, Serena 2550<br />

Marchi, Enrica e13569<br />

Marchi, Paolo 1073<br />

Marchionni, Luigi 4585<br />

Marcial, Luisa Vanessa 1060<br />

Marciano, Roberta e13509<br />

Marciscano, Ariel e16052<br />

Marck, Brett 4520<br />

Marcom, P. Kelly 599<br />

Marcomigni, Luca e18023<br />

Marcos, Jean-Michel 1574<br />

Marcos, Manuel e11517<br />

Marcos, Rafael 1588<br />

Marcott, Valerie D. e15092<br />

Marcoux, J Paul 7588,<br />

e19637<br />

Marcucci, Guido 6526<br />

Marcucci, Lorenzo 629<br />

Marcus, Adam 5560<br />

Marcus, Elizabeth A. e16531<br />

Marcus, Karen Chayt 9502,<br />

9537, 9585<br />

Mardiak, Jozef TPS4677,<br />

9046, e15027, e15028<br />

Mardis, Elaine 503, 9518<br />

Mardjuadi, Feby I e14060<br />

Mardor, Yael 2078<br />

Maréchal, Raphael 3559,<br />

4050<br />

Marek, Billie J. 4085<br />

Marelli, Laura e17543<br />

Margalit, Danielle Nina<br />

e16060<br />

Margeli Vila, Mireia 1001,<br />

e11081<br />

Margeli, Mireia e11545<br />

Margenthaler, Julie A. 6079,<br />

6081, e19593<br />

Margery, Jacques TPS7112,<br />

e16560<br />

Marghoob, Ashfaq A. e19052<br />

Marginean, Celia 3515<br />

Margolese, Richard G.<br />

LBA506, 538<br />

Margolin, Kim Allyson 4533,<br />

8525, 8567, e16563<br />

Margolis, Jeffrey e17015<br />

Margono, Benjamin 7519^<br />

Margulis, Vitaly 4616,<br />

TPS4678<br />

Mari, Ettore 7550<br />

Mari, Véronique 5018,<br />

e12502, e20006<br />

Mariadason, John e14019<br />

Mariani, Andrea 5004, 5076<br />

Mariani, Luca 627<br />

Mariani, Luigi 4507<br />

Mariani, Pascale 8546,<br />

e15524<br />

Mariani, Rachel e12002<br />

Maric, Irina 8088, 8104,<br />

e18568<br />

Mariconti, Luisa 7544<br />

Marien, Ann e15166<br />

Marienhagen, Joerg 7105<br />

Mariette, Christophe 4091,<br />

e14560<br />

Marin, Cristi 8540<br />

Marin, David 10550<br />

Marin, Jan e15195<br />

Marina, Neyssa 9503, 9537,<br />

10081<br />

Marinca, Andreea e15032<br />

Marinca, Mihai Vasile e15032<br />

Marincola, Francesco 8576<br />

Marincola, Franco 10565<br />

Marinello, Jorge Juan 2515<br />

Marineo, Giuseppe e19564^<br />

Maringwa, John T 6002,<br />

7607<br />

Marinho, Simone e11513<br />

Marinko, Tanja 558<br />

Marino, Ana Luisa e19007<br />

Marino, Antonella e11001<br />

Marino, Gregory 6126<br />

Marinucci, Dena e21074<br />

Maris, John M. 9516, 9533,<br />

9576<br />

Maritaz, Christophe 9560,<br />

9572<br />

Mariucci, Sara e13103<br />

Marjanovic, Mira 10576<br />

Mark, Tomer Martin 8036,<br />

e21006<br />

Markert, James TPS2107<br />

Markey, Janell 6042<br />

Marki-Zay, Janos e21140<br />

Markin, Abraham 6016<br />

Markman, Ben TPS3637<br />

Markman, Maurie 5015,<br />

5049, e14697<br />

Markmann, Susanne 552<br />

Markopoulos, Christos 516,<br />

517, 583<br />

Markovic, Svetomir 8599,<br />

8600, e14507, e21112<br />

Markovich, Alla Anatolievna<br />

e19005<br />

Markovits, Judit E 9562<br />

Markow, Justin M. e18554<br />

Markowitz, Avi B. 5502<br />

Markowitz, Sanford D. 1569<br />

Marks, Charles e11569<br />

Marks, Gerald J. e14157<br />

Marks, John e14157<br />

Marks, Lawrence Bruce 6093<br />

Marks, Reinhard 6500^<br />

Markulin-Grgic, Lijerka<br />

e12505<br />

Markus, Susan TPS3117<br />

Marley, Sarah E e15141<br />

Marme, Frederik 1090, 1096<br />

Marmissolle, Fabiana e14598<br />

Marmol, Maribel 3553<br />

Marolleau, Jean pierre 6534,<br />

8026<br />

Maron, Leeza M. 2040<br />

Marosi, Christine 2024,<br />

2053, 2071<br />

Maroto, José Pablo e15001,<br />

e15159<br />

Maroto, Pablo TPS4685<br />

Maroun, Christiane R. 3039,<br />

e13602, e13604<br />

Maroun, Jean Alfred 3578<br />

Marples, Maria 8523<br />

Marques, Dania S<strong>of</strong>ia e15532<br />

Marques, Daniel Fernandes<br />

2038<br />

Marques, Gilberto P e12040<br />

Marques, Mateus e14673<br />

Marquette, Charles Hugo<br />

7591<br />

Marquez, Cristina e11517<br />

Marquez, Diana 6132<br />

Marquez, I e19023<br />

Marquez, Mirari e12033<br />

Marquez, Raul 5068, e15575<br />

Marquez-Medina, Diego 7554<br />

Marquez-Rodas, Ivan e12003<br />

Marquina, Isabel 1570<br />

Marr, Brian 8598<br />

756s Numerals refer to abstract number


Marr, Tiffany e19570<br />

Marrapese, Giovanna 3570<br />

Marrari, Andrea 10026,<br />

10065<br />

Marreaud, Sandrine 10009,<br />

10067<br />

Marrero, Jorge A. e14581<br />

Marrinucci, Dena e21122<br />

Marrodan, Ines e11525,<br />

e17522, e18031, e21009<br />

Marsault, Blandine e19555<br />

Marschner, Ian 5081<br />

Marschner, Norbert e14138<br />

Marsh, Brian e19632<br />

Marsh, Robert de Wilton<br />

4022, e14642<br />

Marsh, Sharon 7586<br />

Marshall, Amy 7077<br />

Marshall, Andrea 5046<br />

Marshall, Ariela 1539<br />

Marshall, Deborah e11000,<br />

e11050<br />

Marshall, Ernest 7562, 8523,<br />

8532, 8564, TPS8605<br />

Marshall, Helen 502, 511,<br />

513<br />

Marshall, James Roger<br />

e15203<br />

Marshall, John 2590, 3095,<br />

TPS3638, TPS10632,<br />

e13116, e14009, e14569,<br />

e14658<br />

Marsiglia, Hugo e14002<br />

Marsoni, Silvia 2540,<br />

LBA7501, e19549<br />

Martel, Céline 10583<br />

Martel, Diane 4098<br />

Martel, Guillaume e14154<br />

Martel-Lafay, Isabelle<br />

LBA4003<br />

Martel-Planche, Ghyslaine<br />

1522<br />

Martell, Robert E. 4117,<br />

4545, e14005<br />

Martella, Francesca e18074<br />

Martelli, Olga LBA7501,<br />

e18065<br />

Marten, Angela TPS3636<br />

Marten-Mittag, Birgitt e15157<br />

Martens, John W.M 10504<br />

Martens, Tobias 2034<br />

Martens, Uwe Marc 3539,<br />

4050, 4090<br />

Martenson, James A. e16572<br />

Marth, Christian 514, 515,<br />

5031, 5042, 5052, 5071,<br />

5087, 5111<br />

Marthey, Lysiane 4018<br />

Marti, Gerald E 6581<br />

Marti, Roberto e14589<br />

Marti-Ciriquian, Juan Luis<br />

7095<br />

Marti-Marti, Francisca Elena<br />

e14015<br />

Martin Broto, Javier 10079<br />

Martin del Yerro, Jose Luis<br />

1111<br />

Martin Liberal, Juan 3096<br />

Martin Lorente, Cristina<br />

TPS4672, e15001, e15159<br />

Martin Reinas, Gaston 9038<br />

Martin Valades, Jose Ignacio<br />

5520<br />

Martin, Andrew J. 9087<br />

Martin, Anne-Laure 543<br />

Martin, Anne-Marie 3102,<br />

LBA8500^, 8501, 8518,<br />

e15085, e15087<br />

Martin, Antoine 8026<br />

Numerals refer to abstract number<br />

Martin, Daniel 5541<br />

Martin, Dena e16047<br />

Martin, Eric 3509<br />

Martin, Francis 1574<br />

Martin, Jan 5004<br />

Martin, Jeremiah Thomas<br />

e14562<br />

Martin, John 1026<br />

Martin, Lainie P. 3029,<br />

5021, 8530<br />

Martin, Ludmila Katherine<br />

4039, e14169, e14170<br />

Martin, Marta e14041<br />

Martin, Michael Gary 3085<br />

Martin, Miguel 596, 604,<br />

TPS652, TPS670, 1001,<br />

1008, e12003, e14058<br />

Martin, Montse e18030<br />

Martin, Paloma 1074<br />

Martin, Peter 8064<br />

Martin, Petra e21024<br />

Martin, Robert C. G. e14581<br />

Martin, Stephane 2082<br />

Martin, Stuart S e21079<br />

Martin, Sue Ellen 4563<br />

Martin, Thomas 8035<br />

Martin-Algarra, Salvador<br />

8572, e18008<br />

Martin-Richard, Marta 3586,<br />

4079, e14097, e14104,<br />

e14552<br />

Martin-Valades, Jose Ignacio<br />

e14147, e15094, e18069,<br />

e18106, e21015<br />

Martinelli, Fabio 5096<br />

Martinelli, Francesca 6002<br />

Martinelli, Giovanni 6527,<br />

6531<br />

Martinez Banaclocha,<br />

Natividad 7011<br />

Martinez Cardús, Anna 9129<br />

Martinez Carrasco (1), Begona<br />

e11543, e19596<br />

Martinez de Vega, Vicente<br />

1111<br />

Martinez Garza, Laura Elia<br />

e15525<br />

Martinez Lago, Nieves<br />

e14733, e19505<br />

Martinez Marín, Virginia<br />

3618, 10547, e13085<br />

Martinez Marti, Alex 7041,<br />

7542<br />

Martinez Rodriguez, Jorge<br />

Luis e15525<br />

Martinez Sesmero (2), Jose<br />

Manuel e19596<br />

Martinez Villacampa,<br />

Mercedes 3571<br />

Martinez, Alberto J 8582<br />

Martinez, Alejandra 5083<br />

Martinez, Carmen 4089<br />

Martínez, Esther e15041<br />

Martinez, Gary 10084<br />

Martinez, Itzel Anahi 1607<br />

Martinez, Juan Carlos e21104<br />

Martinez, Manuel e12016<br />

Martínez, Miguel Angel<br />

e19007<br />

Martinez, Mireia e21009<br />

Martinez, Nieves 7058,<br />

10594<br />

Martinez, Noelia 602<br />

Martinez, Pablo 7041, 7544<br />

Martinez, Patricia e11523<br />

Martinez, Purificación 10512,<br />

10595<br />

Martinez, Raquel 3068<br />

Martinez, Steve R. e19001,<br />

e19013<br />

Martinez, Victor 4584<br />

Martinez-Amores, Brezo 3601,<br />

e14093, e14144<br />

Martinez-Balibrea, Eva 9129<br />

Martinez-Bueno, Alejandro<br />

e18137, e18143<br />

Martinez-Diaz, Francisco<br />

e11024<br />

Martinez-Fernandez, Monica<br />

4584<br />

Martínez-Galan, Joaquina<br />

1041<br />

Martini, Chiara 1604<br />

Martini, Thomas e15160<br />

Martinisi, Julia e19048^<br />

Martinius, Holger e13108^<br />

Martino, Massimo Vincenzo<br />

e18564<br />

Martino, Rosemary TPS5600<br />

Martino, Silvana 1021<br />

Martino, Tami 10622<br />

Martins Ferreira Castro, Ana<br />

Filipa 5567<br />

Martins, Flavia Linhares<br />

e13008<br />

Martins, Renato 5503, 5515,<br />

5518, 5591, 7530, 7600<br />

Martins, Sandro José e14657,<br />

e16046, e18078<br />

Martins, Suelen Patricia dos<br />

Santos e16568<br />

Martins, Yolanda 6103<br />

Martinsson, Jessica e13518<br />

Martoni, Andrea e15185<br />

Martorell, Miguel 7058,<br />

7060, 10594<br />

Martschick, Anja 5064,<br />

e19558<br />

Marttila, Timo e15112<br />

Martus, Peter 2007, 8031<br />

Martyn, Julie TPS5116<br />

Maru, Anish 3011<br />

Maru, Dipen M 3561, 4069,<br />

4078, 4086, e14071,<br />

e14547, e14548<br />

Marucci, Gianluca 2061<br />

Marulappa, Shashidhara Y.<br />

8081<br />

Marumoto, Tomotoshi e13014<br />

Marur, Shanthi 5566,<br />

e16061<br />

Maruyama, Kaoru 10577<br />

Maruyama, Satoru e15127<br />

Marven, Michelle 9124<br />

Marvin, Monica L. e12026<br />

Marx, Alexander 7105<br />

Marx, Gavin M. 4510,<br />

e15152<br />

Marynchenko, Maryna 6600<br />

Mas Estelles, Fernando<br />

e14097<br />

Mas Lopez, Luis Alberto 7531<br />

Más, Luís e15036<br />

Masalu, Nestory Andrew<br />

10601<br />

Mascarenhas, Leo 9564<br />

Mascaux, Celine e18006<br />

Maschek, Birgit J 4056<br />

Maschmeyer, Georg 3<br />

Masci, Giovanna 623<br />

Masciari, Serena 1547<br />

Masciullo, Valeria 5059<br />

Mase, Takahiro 588<br />

Masedu, Francesco 4572<br />

Maseki, Nobuo 8050<br />

Masi, Gianluca 3518, 3540,<br />

3549, 3555, 3585<br />

Masiero, Elena 3612<br />

Maskiell, Judith A 3567<br />

Maslak, Peter George 6609,<br />

6613<br />

Masny, Agnes e16510<br />

Mason, Cathleen 1046<br />

Mason, Christopher 9507<br />

Mason, James R. TPS5113,<br />

e21061<br />

Mason, Malcolm David 4509,<br />

TPS10633^<br />

Mason, Malcom e15199^<br />

Mason, Robert TPS4143<br />

Mason, Teresa e16045<br />

Mason, Warren P. 2051,<br />

2063, TPS2104<br />

Masood, Asif e16043<br />

Masroor, Sohaib 6005<br />

Masrorpour, Fatemeh 7096<br />

Mass, Robert D. 3067<br />

Massacesi, Cristian TPS648<br />

Massad, L. Stewart 5013,<br />

5101<br />

Massard, Christophe 3104,<br />

4554, 4642<br />

Massari, Francesco 630,<br />

4541, e14661<br />

Massett, Holly e16569<br />

Massey, Christine 2063<br />

Massey, Dan LBA7500<br />

Massey, Woody C 2027,<br />

2074^<br />

Massi, Daniela 8581<br />

Massicotte, Marie-Helene<br />

5569<br />

Massidda, Bruno e14094<br />

Massie, Lisa 9002<br />

Massimino, Maura TPS9596<br />

Massin, Frederic e12506<br />

Massmanian, Lisa e13041<br />

Masson, Eric TPS2622, 8541<br />

Massopust, Kristy e13531<br />

Massuti, Bartomeu 7011,<br />

7095, 7522, 7540, 7542,<br />

e18055, e18130, e18131,<br />

e18137, e18143<br />

Masszi, Tamás 8018^<br />

Master, Viraj A. e15006<br />

Masters, Gregg 2521<br />

Masters, Gregory A. e18122<br />

Masterson, Timothy A. 4586<br />

Mastrangelo, Michael J.<br />

8560, e19016<br />

Masuda, Aya 9569<br />

Masuda, Hiroko 1047<br />

Masuda, Munetaka 4070<br />

Masuda, Norikazu 540, 1106,<br />

TPS1147<br />

Masuda, Shinobu 565<br />

Masullo, Pietro 1500<br />

Masumori, Naoya e15071<br />

Matano, Alessandro 530<br />

Matano, Elide e15034<br />

Matasar, Matthew J. 2008,<br />

2089, 8054<br />

Matczak, Ewa 4542, 6092,<br />

e16530<br />

Mateescu, Cristian e14553,<br />

e14660<br />

Mateo, Joaquin 2530, 3021,<br />

3048, 10525<br />

Mates, Mihaela e19541<br />

Mathenge, Edward G. e21090<br />

Matheson, Alastair 3557<br />

Mathew, Josy e19627<br />

Mathew, Sherry 6613<br />

Mathews, John 10037<br />

Mathiason, Michelle A. 6074<br />

Mathieu, Luckson Noe 1573<br />

757s


Mathieu-Nicot, Florence<br />

e19623<br />

Mathijssen, Ron H.J. 2549,<br />

2596, 5001, 10091<br />

Mathios, Assefa 580<br />

Mathiot, Claire 8014<br />

Mathiot, Nathalie e18089<br />

Mathisen, Michael 6544,<br />

6597, 6607<br />

Mathonnet, Muriel 10505<br />

Mathoulin-Pelissier, Simone<br />

4053, 9012, 10035,<br />

e14023<br />

Matijevic, Mark 3030, 5518<br />

Matijevich, Karen 2582,<br />

8547<br />

Matin, Surena F. 4523,<br />

9029, e15075, e15092<br />

Mato, Anthony R. e18507,<br />

e19641<br />

Matono, Rumi e14536<br />

Matos Neto, Joao Nunes 606<br />

Matos, Leandro Luongo 5588,<br />

e16568, e21151<br />

Matous, Jeffrey 8027<br />

Matrana, Marc Ryan 4615<br />

Matro, Jennifer Madeline<br />

1544<br />

Matsubara, Akio e15048<br />

Matsubara, Nobuaki 585<br />

Matsubara, Nobumichi 7565<br />

Matsuda, Eisuke e17517<br />

Matsuda, Yoshinobu 10577<br />

Matsueda, Satoko e13046,<br />

e13050<br />

Matsuguma, Haruhisa 7021<br />

Matsui, Junji e13511,<br />

e19055<br />

Matsui, Kaoru 7028<br />

Matsui, Osamu 4104<br />

Matsui, Takanori e14510<br />

Matsui, Zenichi 9569<br />

Matsumoto, Hiroshi 613<br />

Matsumoto, Koji 5045,<br />

e19548<br />

Matsumoto, Seiji 7080,<br />

10585<br />

Matsumoto, Takeshi 7528,<br />

10610<br />

Matsumoto, Toshifumi<br />

e14722<br />

Matsumura, Akihide e18126<br />

Matsunaga, Tadashi e21007<br />

Matsuno, Yoshihiro e17520<br />

Matsuo, Keitaro 4067, 7560<br />

Matsuo, Takuji 9053<br />

Matsuoka, Lea e14679<br />

Matsusaka, Satoshi 10541,<br />

e14145<br />

Matsushita, Kazuyuki 7046<br />

Matsushita, Kohei e21053<br />

Matsuura, Hiroshi e14722<br />

Matsuura, Kazuto 5542<br />

Matsuura, Kentaro 3079<br />

Matsuura, Kuniomi e17538<br />

Matsuura, Yuka e18036,<br />

e18042<br />

Matsuyama, Yutaka 4104,<br />

e14506<br />

Matsuzaki, Akinobu 9574<br />

Matsuzaki, Hiroumi 5574<br />

Matta, Mona 2060<br />

Matta-Castro, Thalita e14027,<br />

e17548<br />

Matthaios, Dimitrios e17553<br />

Matthay, Katherine K. 9515,<br />

9533<br />

Matthews, John TPS4690<br />

Matthews-Smith, Velmalia<br />

6038<br />

Mattina, Marco e15544<br />

Mattioli, Sandro e18045<br />

Mattonet, Christian 3044,<br />

TPS3115, 7603,<br />

CRA10529<br />

Matulewicz, Theodore 1540<br />

Matulonis, Ursula A 5011<br />

Matulonis, Ursula 5012<br />

Matuschek, Christiane<br />

e11556, e14527, e16033<br />

Matushek, Paul E e21060<br />

Matveev, Vsevolod B. e15025<br />

Matza, Louis S. e16528<br />

Mau, Christine 541<br />

Mauch, Cornelia 8505<br />

Maude, Shannon L. 9506<br />

Mauer, Murielle E. 3508,<br />

4053, TPS4140<br />

Maughan, Benjamin 4525<br />

Maughan, Tim 2522, 3516,<br />

3530, 3574<br />

Maunsell, Elizabeth 9138<br />

Maurea, Nicola 645, e11052,<br />

e13539<br />

Maurel, Joan 3553, 4079,<br />

e14041, e21035<br />

Maurel, Juan 3536, 3586,<br />

3618, 10052, 10547,<br />

e14097<br />

Maurer, Barry 8073<br />

Maurer, Dominik 2522<br />

Maurer, Matthew A. 10516<br />

Maurer, Matthew J. 5077,<br />

9057<br />

Mauri, Davide 5066<br />

Mauri, Simonetta e19648<br />

Mauro, David J. 3531, 3587,<br />

4054<br />

Mauro, Lucia M e14593<br />

Mauro, Michael J. 6605,<br />

TPS6636<br />

Mauro, Virginie 7591<br />

Maus, Martin Karl Herbert<br />

3518, 3546, 3549, 7582,<br />

7594, e12007<br />

Mautner, Victor-Felix 6136<br />

Maverakis, Emanual e19001,<br />

e19013<br />

Mavis, Cory e18537, e18539<br />

Mavridis, Konstantinos 10606<br />

Mavroudis, Dimitrios e18105<br />

Maxim, Peter 10629<br />

Maximiano, Constanza<br />

e17502, e17503, e17515,<br />

e18521<br />

Maxson, DeLee 7534<br />

Maxuitenko, Yulia e15515,<br />

e19006<br />

May, Christoph 4122, 7105,<br />

e13503<br />

May, Kenneth F. 2502<br />

Mayer, Bhabita 9117<br />

Mayer, Deborah K. 6119<br />

Mayer, Erica L. CRA1002<br />

Mayer, Françoise 1051<br />

Mayer, Frank 2522, 2573^,<br />

3530, 4083, 7554,<br />

e14152, e15026<br />

Mayer, Ingrid A. 510, 605<br />

Mayer, Jiri 6559, 6584, 6627<br />

Mayer, Julie Ann 584,<br />

TPS10631<br />

Mayer, Musa 1526, e11051,<br />

e19633<br />

Mayer, Nicholas J. e15078<br />

Mayer, Robert J. 3517, 3631<br />

Mayer, Tina M. TPS2107<br />

Mayer, Wesley A TPS4678<br />

Mayer-Mokler, Andrea 2522,<br />

3530<br />

Mayerle, Julia TPS3639<br />

Mayne, Susan T e16511<br />

Mayo, Angela 8010<br />

Mayo, Clara 4068, 7522,<br />

10596, e14521, e18130,<br />

e18131, e18143<br />

Mayorga, Cheryl Ann 2569<br />

Mayr, Doris e15529<br />

Mays, Theresa A. e13025<br />

Mazard, Thibault 4087<br />

Mazer, Mia 2084, 2087<br />

Mazeron, Renaud 10098<br />

Mazhar, Danish 4529, 4531<br />

Maziere, Camille e14017<br />

Mazieres, Julien TPS7112,<br />

7584, e18089<br />

Mazorra, Zaima 2527<br />

Mazuir, Florent e15116<br />

Mazumdar, Jolly 3000, 8518<br />

Mazur, Grzegorz 6559, 6632<br />

Mazurenko, Natalia N.<br />

e20501<br />

Mazzanti, Chiara 629<br />

Mazzanti, Paola e21070<br />

Mazzanti, Roberto e14577<br />

Mazzara, Calogero 4627<br />

Mazzarello, Sasha 10620<br />

Mazzei, Maria Eugenia<br />

e19007<br />

Mazzer, Micol 3612<br />

Mazzocchi, Maria e14018<br />

Mazzola, Giuseppe e19595<br />

Mazzoni, Enrica e11547<br />

Mazzoni, Francesca e18074<br />

Mc Hugh, Kieran 9517<br />

McAfee, Steven L 6617,<br />

6618, 6621<br />

McAleer, Cecilia TPS4679<br />

McAleese, Fionnuala 8059<br />

McArdle, Stephanie 1013,<br />

1098<br />

McArthur, Grant A. 8502^,<br />

8503^, 8517, 8520,<br />

e13054<br />

McBride, Sean M 5594<br />

McBride, William H. e13045<br />

McCabe, Christopher TPS665<br />

McCabe, Mary S. 6073, 9121<br />

McCabe, Nuala 10553<br />

McCaffrey, John 581, 1543,<br />

e12038, e19601, e19632,<br />

e21147<br />

McCaffrey, Judith C 5564,<br />

5591<br />

McCaffrey, Thomas V 5564<br />

McCall, Linda Mackie 1107<br />

McCann, Georgia Anne-Lee<br />

e15585<br />

McCann, Lauren CRA4502,<br />

e15059<br />

McCanna, James e21047<br />

McCarter, Martin TPS10632<br />

McCarthy, Aoife e15078<br />

McCarthy, Bridget J. e12036<br />

McCarthy, Damian L. 3095<br />

McCarthy, Joy S. e11079<br />

McCarthy, Philip L. 6565<br />

McCarty, John e18550<br />

McCaskill-Stevens, Worta J.<br />

TPS657<br />

McCaskill-Stevens, Worta<br />

6086<br />

McCaughan, Georgia J. 9126<br />

McCaul, James Anthony<br />

e16042<br />

McCauley, Dilara 1055,<br />

e13586, e15200<br />

McClain, Jonathan e12005<br />

McClanahan, Pamela e14503<br />

McClay, Edward Francis 2569<br />

McCleary, Nadine Jackson<br />

4112, e19586<br />

McClelland, Michael 10543<br />

McCleod, Michael 4127<br />

McClish, Donna 1563<br />

McCloud, Philip 7604<br />

McClure, Bev 3629<br />

McClure, Candace e13078,<br />

e15578<br />

McCombie, William R. 4562<br />

McCook, John 3015, e14593<br />

McCormack, Peter 3048,<br />

3051<br />

McCormick, Beryl 1004<br />

McCormick, Kristen N. 7071<br />

McCormick, Lynn 6126<br />

McCready, David R. 1019,<br />

9131<br />

McCulloch, Leanne 8086,<br />

e18569<br />

McCulloch, Timothy M<br />

e16010<br />

McCulloch, William 567<br />

McCully, Katen C. 1544,<br />

1550<br />

McCusker, Margaret Elizabeth<br />

8585<br />

McCutcheon, Ian E. 2019<br />

McDade, Theodore P. 4059,<br />

e14691<br />

McDaniel, Kelly e16528<br />

McDermed, Jonathan E.<br />

e15113<br />

McDermott, David F.<br />

CRA2509, 4504, 4505,<br />

4543, 4617, 4635, 8507,<br />

8508, 8516, 8567, 8569<br />

McDermott, Enda 1042<br />

McDermott, Martina 617, 633<br />

McDermott, Raymond S.<br />

e14705, e15093, e17561<br />

McDermott, Raymond e15016<br />

McDermott, Shaunagh<br />

e14615<br />

McDevitt, Jennifer T.<br />

TPS2621<br />

McDonagh, Charlotte<br />

TPS3111<br />

McDonagh, Paul D. e19055<br />

McDonald, Alicia 6595<br />

McDonald, Angus 4531<br />

McDonald, Dan CRA2509,<br />

TPS2613, TPS2622, 4505,<br />

7509, 8507<br />

McDonnell, Felicity e14705,<br />

e15016<br />

McDonnell, Kevin e12026<br />

McDunn, Susan H. 9085<br />

McDyer, Fionnuala A 10553<br />

McElduff, Patrick e14166<br />

McElhinny, Abigail 4041<br />

McFarlane, Graham 6005<br />

McGaughey, Dean S. e13032<br />

McGee, Sharon F. 581<br />

McGinn, Travis W. 9001<br />

McGinniss, Matthew Jerome<br />

1040, 4013, 10630,<br />

e15209, e20507<br />

McGlone, Thomas e16513<br />

McGoldrick, Trevor 3048<br />

McGovern, Rachel TPS7611<br />

McGovern, Renee M. 9581<br />

McGowan, Margit e21075<br />

758s Numerals refer to abstract number


McGowan, Patricia M. 622,<br />

1042<br />

McGrath, Sophie e15517<br />

McGraw, Steven 2031<br />

McGree, Michaela 5004<br />

McGregor, John R. 10614<br />

McGregor, Kimberly 3057<br />

McGreivy, Jesse S. 4042,<br />

TPS4135<br />

McGuire, Brendan e14581<br />

McGuire, Kandace P 1038<br />

McGuire, William P. 5012<br />

McGuirk, Joseph 10605<br />

McHenry, Brent TPS4689,<br />

TPS4691<br />

McHugh, Deaglan e15016<br />

McHugh, Kelsey 9544<br />

McHUGH, Peter J e13587<br />

McIntosh, Christine e13569<br />

McIntosh, Lynn e18079<br />

McIntyre, John B. e21044<br />

McIntyre, Kristi 547, 3069<br />

McKay, Marie 8564<br />

McKee, Kathryn S. e15075<br />

McKee, Mark D. 3532, 7512,<br />

e13583<br />

McKeegan, Evelyn Mary<br />

e13583<br />

McKeever, Elizabeth 4109,<br />

e14524, e14592<br />

McKendrick, Joseph James<br />

3629, TPS3637, e16516<br />

McKendrick, Joseph J 3505<br />

McKenna, Aaron 2020<br />

McKenna, David 2546<br />

McKenna, Dorothy R 6013<br />

McKenna, Edward 3522,<br />

3523, 3525, e19048^<br />

McKenney, Jesse e15005<br />

McKenzie, Meghan 2566<br />

McKeown, S. e14022<br />

McKibbin, Trevor e18553<br />

McKiernan, James M. 4593,<br />

e15018, e15019, e15021<br />

McKinlay, Mark A 3029<br />

McKinley, Marti 4052<br />

McKinney, Yolanda 10589<br />

Mckoy, June M. 4063, 6623<br />

McKoy, June M 2532<br />

McLachlan, Sue-Anne<br />

e16501, e16562<br />

McLaughlin, Brian Thomas<br />

6563<br />

Mclaughlin, Laurie e19598<br />

McLaughlin, Peter 8067<br />

McLean, Estelle 10553<br />

McLemore, Elisabeth Paul<br />

e14069<br />

McLendon, Roger E 2021<br />

Mcleod, David G. e15207<br />

McLeod, Howard L. 10515,<br />

e13109<br />

McLinden, Michelle 1094<br />

McMahon, Jeanne A 3582<br />

McMahon, Jeremy McMahon<br />

e16042<br />

McMahon, Pamela M 6004<br />

McMasters, Kelly M. 8534<br />

McMeekin, D. Scott 3008,<br />

3041, 3097<br />

McMenemin, Rhona e17519<br />

McMillan, Alex 8060<br />

Mcmillan, Donald C. 3599,<br />

3611, e14054, e14092,<br />

e14110, e14117<br />

McMillan, Robert 1118, 1541<br />

McMullin, Mary Frances<br />

TPS6642^<br />

Numerals refer to abstract number<br />

McNally, Diane L. e15146,<br />

e15148<br />

McNally, Orla 5073<br />

McNamara, Erica J. 6046,<br />

6088<br />

McNamara, Mairead Geraldine<br />

4109, e14524<br />

McNamara, Mairead e14592<br />

McNamara, Michael J.<br />

e14611, e14665<br />

McNeel, Douglas G.<br />

TPS2613, 4650<br />

McNeill, Jonathan D 7595<br />

McNulty, Ann M 8082<br />

McPherson, Christopher<br />

TPS3116<br />

McPherson, Eugene e17022<br />

McPherson, Monica e14124,<br />

e18516, e19638<br />

McQuade, Jennifer e18540<br />

McQueen, Robert Brett<br />

e16550<br />

McQueen, Teresa J TPS6635<br />

McSherry, Frances 2027,<br />

2074^<br />

McSwain, Anita P. e11084<br />

McTiernan, Anne e14005<br />

McWalter, Gael 1548<br />

McWhirter, Elaine 3082<br />

McWilliams, Robert R. 3102,<br />

e14594<br />

Meade, Angela M. 3516<br />

Meadows, Anna T. 9515<br />

Meadows, Helen 4004, 4029<br />

Meagher, Alison e19568<br />

Meani, Francesco e15549<br />

Means-Powell, Julie Ann<br />

4117, 4545, 8519<br />

Meany, Holly Jane 9533<br />

Mebis, Jeroen 4622<br />

Medalia, Gal e14571,<br />

e14731<br />

Medeiros, Michelle 7546,<br />

9103<br />

Medeiros, Rui e15532<br />

Medford-Davis, Laura Nyshel<br />

7040<br />

Medina Sanson, Aurora<br />

e20005<br />

Medina Villaamil, Vanessa<br />

e14595, e15076, e15077,<br />

e15079, e21002<br />

Medina, Fernando Lara 1607,<br />

e11016<br />

Medlin, Stephen Charles<br />

8041<br />

Medlin, Stephen e18576<br />

Medvedev, Viktor 5508<br />

Meer, Laurens van der 9550<br />

Meershoek – Klein<br />

Kranenbarg, Elma 526<br />

Meert, Anne-Pascale e18006<br />

Meert, Nicolas e14060<br />

Meeus, Marie-Anne 3018^<br />

Meeus, Pierre e14730<br />

Mefti, Fawzia TPS662<br />

Mega, Anthony E. 4662<br />

Megason, Gail C. 9547<br />

Mego, Michal TPS4677,<br />

e15027, e15028<br />

Mehan, Mike e21012,<br />

e21142<br />

Mehanna, Hisham e16042<br />

Mehedint, Diana e15039,<br />

e15046, e15204<br />

Mehmood, Tahir e16043<br />

Mehmud, Faisal CRA4502<br />

Mehnert, Janice M. TPS3112<br />

Mehra, Ranee 3007, 5500<br />

Mehran, Mariam 3039,<br />

e13602, e13604<br />

Mehran, Reza J. 4069, 4086,<br />

7078<br />

Mehring, Elmar 6567<br />

Mehrishi, Jitendra e13102<br />

Mehrotra, Bhoomi TPS5601,<br />

e16026<br />

Mehta, Ajay O. TPS649,<br />

TPS651, 3011<br />

Mehta, Cyrus R. TPS6637<br />

Mehta, Jayesh 6623, 8037,<br />

e18556<br />

Mehta, Keyur 522, 523<br />

Mehta, Maitrik e15505<br />

Mehta, Minesh P. 2008b,<br />

2001, 2013, 2047<br />

Mehta, Monal e13543<br />

Mehta, Shaveta 1066<br />

Mei, Baigen TPS3113<br />

Mei, Matthew 8038<br />

Mei, Qi 5097<br />

Meier, Friedegund Elke 8526<br />

Meier, Werner 5007, 5031<br />

Meijer, Coby 4528, e15035<br />

Meijer, Sybren L. 4094<br />

Meiler, Johannes 4083<br />

Mein, Charles 10550<br />

Meiri, Eti 10575, e21008<br />

Meisner, Angela W. 1558<br />

Meisner, Christoph 1067,<br />

2000<br />

Mekan, Sabeen Fatima 1046<br />

Mekata, Eiji e11564<br />

Mekhail, Tarek 7100, e13113<br />

Mel Lorenzo, Jose Manuel<br />

e18040<br />

Mel, Jose Ramón e11535,<br />

e18070, e21088<br />

Melamed, Jonathan e15191<br />

Melanson, Anne Marie<br />

e11548<br />

Melcher, Carola Anna<br />

TPS1146, 1602, 10540<br />

Meldgaard, Peter 7001<br />

Melegari, Elisabetta e11054<br />

Melemed, Allen S. 1068<br />

Melemed, Symantha<br />

LBA7507<br />

Melendez Asensio, Barbara<br />

10500<br />

Melhem-Bertrandt, Amal 527,<br />

537<br />

Melichar, Bohuslav 559,<br />

3010, LBA4007, 4503,<br />

e14556<br />

Melisi, Davide e14661<br />

Melisko, Michelle E. 9107<br />

Mell, Loren K. 6121, e14555<br />

Mellado, Begoña TPS4674,<br />

e15041<br />

Mellemgaard, Anders 7026<br />

Mello, Celso Lopes 10069,<br />

e14119, e14680, e14728<br />

Mellor, Dan e16538<br />

Mellstedt, Hakan 6557,<br />

e13518, e15531<br />

Melnikova, Vladislava O.<br />

e13500, e13562, e13575,<br />

e13577, e15124, e21028,<br />

e21029<br />

Melnikova, Vladislava e21058<br />

Melnychuk, David e14020<br />

Melo Mendes, Ana Valeria<br />

e12021<br />

Melosky, Barbara L. 7020<br />

Meltzer, Paul S. TPS7609<br />

Melville, Heather 3095<br />

Melville, Sharon K. 6070<br />

Melvin, Cathy 6017, 6029<br />

Memoli, Vincent 7022<br />

Memon, Muhammad e20508<br />

Men, Taoyan 7608<br />

Menard, Jean e15049<br />

Menasherov, Nikolai e14571<br />

Menda, Yusuf e15143<br />

Mendelson, David S. 3032,<br />

3034, 3042^, e13077<br />

Mendelson, Estella 6626^<br />

Mendez Herrera, Maria del<br />

Carmen e15527<br />

Mendez Vidal, Maria Jose<br />

TPS4672, TPS4688<br />

Mendez, Eduardo 5503, 5515<br />

Mendez, Guillermo Ariel<br />

e14598<br />

Mendez, Jose Carlos e14041<br />

Mendez, Maria Jose TPS4685<br />

Mendez, Miriam e17502,<br />

e17503, e17515, e18521<br />

Mendez, Pedro 4579<br />

Mendiola Fernandez, Cesar<br />

1001<br />

Mendiola, Cesar 642,<br />

e11081, e15552<br />

Mendivil, Alberto e15500<br />

Mendonca, Eneida 9583<br />

Mendoza, Jose Francisco<br />

e15571<br />

Mendoza, Silvia 2527<br />

Mendoza, Tito R. 5561,<br />

5589, 6551, 8091, 9047,<br />

9072, 9081, 9082, 9083,<br />

9099, 9106, 9118, 9140,<br />

e15173<br />

Menefee, Michael E.<br />

TPS2618, 5544<br />

Menen, Rhiana e21051<br />

Menendez, Xiomara e13084<br />

Meneses, Abelardo 1607<br />

Menetrier-Caux, Christine<br />

10562<br />

Meng, Fanliang e15563<br />

Meng, Maxwell V. 4593<br />

Meng, Raymond D. TPS2616,<br />

9105, e13114<br />

Meng, Xia 3551<br />

Meng, Xue 4073<br />

Meng, Xuli e11566<br />

Menon, Hari e17003<br />

Menon, Jarayam 4106<br />

Menon, Mani 1560<br />

Menoyo Bueno, Alicia e16037<br />

Menten, Johan TPS2104<br />

Menzies, Alexander M. 550,<br />

8558, e19011<br />

Menzies, Alexander M 3102<br />

Menzin, Andrew 5056, 5057<br />

Meoni, Giulia e18074<br />

Merad, Mansouriah 4621<br />

Merante, Serena 6531<br />

Mercado, Gabriela e18028<br />

Mercaldo, Nathaniel D.<br />

e14501<br />

Mercatali, Laura 10615<br />

Mercer, Carol A TPS3116<br />

Merchant, Melinda S. 9545<br />

Merchant, Thomas E. 9531<br />

Mercier, Betty 9074<br />

Mercier, Mariette 1584<br />

Mercier-Blas, Anne e11019<br />

Mercill, Sharon L e13086<br />

Mercuro, Giuseppe 9006<br />

Merdek, Keith D. e21146<br />

Meredith, Judy e14098<br />

Meredith, Rhonda 1058<br />

Mergen, Noemi 6500^<br />

759s


Mergenthaler, Hans-Guenther<br />

e15026<br />

Merghoub, Taha 2521<br />

Mergui-Roelvink, M. 2550,<br />

e13028<br />

Meria, Paul e15524<br />

Meric-Bernstam, Funda 1015,<br />

1029, 1054, 1104, 1130,<br />

e14662<br />

Merican, Ismail 4106<br />

Meriggi, Fausto e14018<br />

Merimsky, Ofer 10034,<br />

10043<br />

Merino, Maria 1570, 9073,<br />

e11028<br />

Merkelbach-Bruse, Sabine<br />

CRA10529<br />

Merker, Vanessa L. 6136<br />

Merkow, Ryan P 3608<br />

Merks, Johannes H.<br />

TPS9597^<br />

Merla, Amartej e14019<br />

Merlano, Marco Carlo 2600,<br />

5502<br />

Merle, Patrick e18089<br />

Merlin, Jean-Louis 10517,<br />

TPS10634, e14129<br />

Merlo, Domenico Franco<br />

1073, e21065<br />

Mernaugh, Ray 628<br />

Meropol, Neal J. 605, 1514,<br />

6021, e14021<br />

Merrick, Daniel 10580<br />

Merriewether, A. Rene 2576<br />

Merriman, Kelly W. 6125<br />

Merseburger, Axel S. e15195<br />

Mertens, Ann 9505<br />

Mertens, Cecile 9012<br />

Mertens, Wilson C. e19543<br />

Mes-Masson, Anne-Marie<br />

e15202<br />

Mesa, Ruben A. 6624,<br />

TPS6641<br />

Meschino, Wendy e12034<br />

Meservey, Caitlin 610,<br />

TPS1134<br />

Meshref, Mohamed M.<br />

e16505<br />

Mesia, Ricard 5502, 5516^<br />

Mesker, Wilma E e14151<br />

Mesnard, Nathalie TPS646,<br />

e11019<br />

Messa, Francesca 2600<br />

Messahel, Souad e15091<br />

Messam, Conrad A. 9117<br />

Messersmith, Wells A. 2567,<br />

2568, 3017, 3528, 3552,<br />

e13006, e13048, e14071<br />

Messina, Antonella 10026<br />

Messina, Caterina e15185<br />

Messina, Giuseppe e18564<br />

Messinger, Yoav H. 9521,<br />

9522<br />

Mester, Jessica 1508, 1516<br />

Mesti, Tanja e14083<br />

Met Domestici, Marie Amelie<br />

e12502, e20006<br />

Metges, Jean Philippe<br />

e14148<br />

Metheny, Trina 520<br />

Metrakos, Peter 4118,<br />

e14621<br />

Metran Nascente, Cristiane<br />

e20506<br />

Metro, Giulio e18023,<br />

e18101<br />

Metwalli, Adam R. TPS4680<br />

Metz, James M. 6135<br />

Metz, Wendy e21066<br />

Metzger, Brigitte e14180<br />

Metzger, Monika 9502<br />

Metzger, Otto e21010<br />

Metzger, Severine 4614<br />

Metzinger, Daniel 5095<br />

Metzner, Bernd 8031,<br />

e15026<br />

Metzner, Cornelia 4122<br />

Metzner-Sadurski, Joanna K.<br />

9061<br />

Meulemans, Bart 8076<br />

Meunier, Jérome 9011<br />

Meunier, Katy Luce e19500<br />

Mewawalla, Prerna 6555,<br />

e18543<br />

Meyer, Anne-Marie 5539,<br />

6104, e15184<br />

Meyer, Christian e17536<br />

Meyer, Christiane e19050<br />

Meyer, Frank e14630<br />

Meyer, Gary 3537, e14584<br />

Meyer, Guy TPS9149^<br />

Meyer, James S. 9537<br />

Meyer, Janelle Marie 10011^<br />

Meyer, Jeffrey John e14165<br />

Meyer, Julia 9507<br />

Meyer, Meghan E 9071<br />

Meyer, Michael e17529<br />

Meyer, Nicolas 8591, e13594<br />

Meyer, Tim 4121, 4123,<br />

TPS4150, e14696, e17543<br />

Meyer, William H. 9509<br />

Meyer-Sabellek, W e13597<br />

Meyer-ter-Vehn, Tobias 2054<br />

Meyerhardt, Jeffrey A. 3537,<br />

4112, 4125, e14114,<br />

e19586<br />

Meyers, Brandon M e14001<br />

Meyers, Bryan F 4062, 7008<br />

Meyers, Christina A. 9072<br />

Meyers, Jeffrey P 7555<br />

Meyers, Marlene e11529<br />

Meyers, Paul A. 9503<br />

Meyerson, Howard TPS655<br />

Meyerson, Matthew 7006<br />

Meyskens, Frank L. 9018<br />

Meza, Jane L 9509<br />

Mezhir, James J. 10015<br />

Mezquita, Laura 3601,<br />

e14093, e14144<br />

Mezynski, Janusz 4553,<br />

e13599<br />

Mhawech-fauceglia, Paulette<br />

5091<br />

Mhlanga, Joyce 10509<br />

Miah, Aisha 10060<br />

Mianowska, Marta e14159<br />

Miao, Sara 4632<br />

Micallef, Sandrine 3080<br />

Miceli, Rosalba 5555,<br />

10000, 10016<br />

Micetich, Kenneth C. 4113<br />

Micewicz, Ewa D. e13045<br />

Micha, John P. e15500<br />

Michael, Agnieszka e15517<br />

Michael, Michael 3629,<br />

TPS4141<br />

Michael, Shaunita A. 4117,<br />

4545, 8519<br />

Michaelides, Ioannis e15030<br />

Michaelidou, Kleita 10606<br />

Michaelson, James e18520<br />

Michaelson, M. Dror 4546,<br />

4549, 4566, 4644,<br />

TPS4682<br />

Michaelson, Richard 614<br />

Michailichenko, Tatyana<br />

e13059<br />

Michailidis, Prodromos<br />

e14567<br />

Michalaki, Vasiliki e13556<br />

Michallet, Anne-Sophie 8068<br />

Michallet, Mauricette 6534,<br />

6542<br />

Micheau-Bonnier, Douglas<br />

9011<br />

Michej, Wanda 8548, e19022<br />

Michel, Maurice Stephan<br />

4539<br />

Michel, Pierre LBA4003,<br />

TPS10102^<br />

Michele, Antonio e15152<br />

Micheli, Rodrigo 1522<br />

Michelin, Odair Carlito<br />

e14173<br />

Michels, Jean-Jacques 10018<br />

Michelucci, Angela e13057<br />

Michener, Tracy 8508<br />

Michielin, Olivier e19050<br />

Michiels, Stefan 507, e21010<br />

Michimae, Hir<strong>of</strong>umi 5003,<br />

7515<br />

Michina, Zoya P. e19012<br />

Michl, Patrick 4050<br />

Michor, Franziska 4577,<br />

4585<br />

Michotte, Alex 2050<br />

Michoux, Nicolas 5519<br />

Middel, Peter 3600, e14162<br />

Middleton, Gary William<br />

LBA4000, TPS4143<br />

Middleton, Mark R. 3051,<br />

e13587, e14625<br />

Middleton, Steven e13600^<br />

Midgley, Rachel A TPS10632,<br />

e13007<br />

Midgley, Rachel LBA4001<br />

Midthun, David E. 1610<br />

Miele, Evelina e12510<br />

Mielgo Rubio, Xabier e11074<br />

Mier, James Walter 8516<br />

Mierzwa, Michelle Lynn<br />

e14714<br />

Mietlowski, William Leonard<br />

2543<br />

Mietzel, Melissa A 8011<br />

Migdady, Yazan 1034<br />

Migden, Michael R 8579<br />

Miglianico, Laurent e15131<br />

Miguel, Ana 10608<br />

Mihalcioiu, Catalin Liviu<br />

Dragos TPS1139<br />

Mihalis, Eva 9107<br />

Mihic, Daniela 8563<br />

Mijatov, Branka 8553<br />

Mikami, Kazuya e15055,<br />

e15099<br />

Mikami, Tomomi 3002<br />

Mikhael, Joseph 8010<br />

Mikhail, Sameh e14009<br />

Mikhaylova, Irina N. 2524<br />

Mikheykin, Andrey 10587<br />

Miki, Izumi e19557<br />

Miki, Jun e15127<br />

Miki, Tsuneharu e15055,<br />

e15098, e15099<br />

Mikkelsen, Tom 2018<br />

Mikula, Lynn A 10050<br />

Mikus, Gerd 10041^<br />

Mikuskova, Eva 9592<br />

Milam, Michael R. 5095<br />

Milandri, Carlo e18045<br />

Milani, Manuela e11546<br />

Milani, Richard V e15192<br />

Milano, Filippo 6631<br />

Milano, Gerard A. 2537^,<br />

2600<br />

Milano, Michael T 1536<br />

Milanowski, Janusz 7090<br />

Milas, Mira 1508<br />

Milbourne, Andrea 6116<br />

Milburn, Christy 8533<br />

Milecki, Piotr 4642<br />

Milella, Michele 4017, 4541,<br />

e13512, e13572, e14661<br />

Miles, Dale 4504, 5547<br />

Miles, David LBA1, TPS660,<br />

e11055^, e16547<br />

Miles, Joshua 639<br />

Miles, Lisa e14015<br />

Mileshkin, Linda R. TPS5116,<br />

9124, e15541, e16538,<br />

e16562, e19605<br />

Miletello, Gerald 1086<br />

Milhem, Mohammed M.<br />

3056, LBA8509, 10010,<br />

TPS10103, e13088,<br />

e15010<br />

Milione, Massimo e21118<br />

Miliotto, Anthony e11563<br />

Milisen, Koen 9035<br />

Millán, Isabel e18521<br />

Millar, Ewan K.A 504<br />

Millar, Jeremy TPS4690<br />

Millard, Frederick E. e15115<br />

Millenson, Michael Mark<br />

e18530<br />

Millenson, Michael M e19580<br />

Miller, Andrew H. 3055,<br />

6076, 9016, 9122<br />

Miller, Austin 5041<br />

Miller, Barry 2528<br />

Miller, Brandon L. e21124<br />

Miller, Carole Brennan 6605,<br />

6624, TPS6642^<br />

Miller, Claire 3083, e19647<br />

Miller, Danielle TPS4141<br />

Miller, David Scott 5087,<br />

e15501<br />

Miller, Dustyn e15042<br />

Miller, Ehud 1564<br />

Miller, Elizabeth 2074^<br />

Miller, Fiona Alice e19616<br />

Miller, Jessica 6054, 9014<br />

Miller, Kathy TPS663, 1027,<br />

6025, e21099<br />

Miller, Kenneth David 6067<br />

Miller, Kurt 4510, 4519^<br />

Miller, Langdon L. 6518^<br />

Miller, Laura Elizabeth<br />

7072^, e17531^<br />

Miller, Lisa e13067<br />

Miller, Mark 9067<br />

Miller, Melissa F 9048<br />

Miller, Meredith Halks 10576<br />

Miller, Naomi 1123<br />

Miller, Paul J. E. e21077<br />

Miller, Paul 4056<br />

Miller, Robert C. e14507,<br />

e14555, e16572<br />

Miller, Robert Stephen 10509<br />

Miller, Rowan 5035<br />

Miller, Thomas P. 8001,<br />

e18534, e18536<br />

Miller, Todd W 1054<br />

Miller, Tracie L. 9530<br />

Miller, Vincent A. 3083,<br />

3533, 7527, 7529, 10590,<br />

e17541<br />

Miller, Wilson H. 1099<br />

Miller, York E 10580<br />

Milleron, Bernard 7057,<br />

e18075<br />

Millholland, Robert 563,<br />

6063<br />

760s Numerals refer to abstract number


Millikan, Randall E. 4523,<br />

4615<br />

Mills, David R. e21116<br />

Mills, Glenn Morris e14667,<br />

e16009<br />

Mills, Gordon B. 1015, 1054,<br />

4131<br />

Millward, Carl 3512, 4560<br />

Millward, Michael 3000,<br />

7079^, LBA8500^,<br />

e16501, e19642<br />

Milne, Ginger L 7518, 10514<br />

Milne, Roger L 1502<br />

Milner, Alvin e13594<br />

Milner, Ruth 9529<br />

Milojkovic Kerklaan, Bojana<br />

e13594<br />

Milosevic, Michael 5105<br />

Milowsky, Matthew I. 4527,<br />

4581^, 4592, e15022<br />

Milton, Ashley 2597<br />

Milutinovic, Aladin e21146<br />

Mimaki, Sachiyo 1552<br />

Mimidis, Kostantinos e14567<br />

Mims, Martha P. 4643<br />

Min, Byung Soh e14084<br />

Min, Chang Ki 8097<br />

Min, Trisha 4109, e14524,<br />

e14592<br />

Minami, Hironobu e16034,<br />

e16505<br />

Minami, Seigo e18007,<br />

e18056, e18141<br />

Minamimoto, Ryogo 4626<br />

Minardi, Daniele e15074<br />

Minasian, Lori M. 9018,<br />

9047, 9144, e19633<br />

Minato, Koichi 7003, 7012,<br />

7515, e18037, e19556<br />

Minato, Nagahiro e21004<br />

Minca, Eugen C. 620<br />

Minden, Mark D 6627, 6632<br />

Minegishi, Yuji 7563, 7565,<br />

e18129<br />

Miner, Thomas J. 4098<br />

Mineur, Laurent 3502, 3506,<br />

TPS10102^<br />

Ming, Zhao e17501<br />

Minhans, Sachin e18041<br />

Minichillo, Santino e13057<br />

Minisini, Alessandro 1044,<br />

e13058, e13070<br />

Mink, Jennifer 8594<br />

Minn, Heikki 5502<br />

Minna, John D. 7096, 7589,<br />

10612, e18077<br />

Minni, Francesco e14647<br />

Mino, Emilio Araujo 1611<br />

Minomo, Shojiro 10577<br />

Minor, David R. 8508, 8518<br />

Minoshima, Yukinori e13511<br />

Minotti, Vincenzo e18023,<br />

e18101, e21094<br />

Minozzi, Marina e12510<br />

Minsat, Mathieu e15535<br />

Minton, Neil 8037, e18559<br />

Minton, Susan E. 1585,<br />

TPS2620<br />

Mintz, Arlan H. 8528<br />

Minuto, Francesco e19038<br />

Miquel, Catherine 2023<br />

Miquel, Rosa 3536<br />

Mir, Olivier 1095, 2557,<br />

5067, e13056<br />

Mirabel, Xavier e14560<br />

Mirabile, Aurora 5555<br />

Miraglio, Emanuela 2600<br />

Mirakhur, Beloo LBA8500^<br />

Miralles, Ambrosio 9073<br />

Numerals refer to abstract number<br />

Miramon Lopez, Jose e11081<br />

Miranda Rosales, Luis Miguel<br />

e18524<br />

Miranda, Andrea 5093, 5094,<br />

e15547, e15551, e15553<br />

Miranda, Christine e14102<br />

Miranda, Gus 4576, 4578,<br />

4588, 4589<br />

Miranda, Raelson Rodrigues<br />

e12021<br />

Miranda, Susana e13601<br />

Miranda, Vanessa Costa<br />

e14702<br />

Mirandola, Leonardo e21105<br />

Miro, Cristina e11517<br />

Miron, Lucian e15032<br />

Miron, Patricia 6569<br />

Miroshnichenko, Gennady<br />

6134<br />

Mirshahidi, Hamid R. e16523<br />

Mirshams, Maryam 10522<br />

Mirza, Mansoor Raza 1055,<br />

LBA5002^<br />

Miscoria, Manuela e15091<br />

Misek, Glen I. e14640<br />

Mishima, Hideyuki 3524,<br />

3607, TPS3642<br />

Mishima, Yuji 10541<br />

Mishra, Anand Kumar e19516<br />

Mishra, Anjali 10587,<br />

e16023<br />

Mishra, Mark Vikas e11505<br />

Mishra, Pravas Chandra<br />

e17000<br />

Mishra, Priyankana 527<br />

Mishra, Saroj Kanta e16023<br />

Misino, Andrea e21113,<br />

e21134<br />

Misitzis, Ioannis 10606,<br />

e11073, e11519<br />

Miskovska, Viera TPS4677,<br />

e15027, e15028<br />

Misleh, Jamal Ghazi e18122<br />

Misra, Sumathi 9004<br />

Misra, Vivek e14015<br />

Missale, Gabriele TPS4151<br />

Misset, Jean-Louis 568^,<br />

5018<br />

Missiaglia, Edoardo 9510<br />

Mister, Donna 9122<br />

Mistry, Bipin 3030<br />

Mita, Alain C. 2610, 3073,<br />

3078, 7029, e15116,<br />

e18047<br />

Mita, Monica M. 3060, 3073,<br />

3078, e15116<br />

Mitchell, Aaron 6051<br />

Mitchell, Alan 8041<br />

Mitchell, Carmen 4586,<br />

e15003<br />

Mitchell, Edith P. 3621,<br />

4043<br />

Mitchell, Heather e21008<br />

Mitchell, Lada TPS653,<br />

TPS654^, 3614, 8517<br />

Mitchell, Paul 6058,<br />

LBA7000, 7079^, e17539<br />

Mitchell, Sandra A. 9047,<br />

9130, 9144, e19633<br />

Mitchell, Scott 10038,<br />

10039<br />

Mitchell, Susan L. 9004<br />

Mitchinson, Christy e21037<br />

Mitha, Nathalie e18505<br />

Mitikiri, Nirupama Devi<br />

e15108<br />

Mitin, Andrey e15029<br />

Mitra, Anirban Pradip 4565,<br />

4575, 4578<br />

Mitra, Dipendrea Kumar<br />

e17511<br />

Mitra, Nandita 1539, e18032<br />

Mitry, Emmanuel 4036,<br />

4071, 4087, e14065,<br />

e14168<br />

Mitsiades, Nicholas e17018<br />

Mitsudo, Kenji 5556<br />

Mitsudomi, Tetsuya TPS7110,<br />

7521, e18145<br />

Mitsuoka, Keisuke e21059<br />

Mitsuoka, Shigeki 6112,<br />

e17538<br />

Mitsuyama, Shoshu 1119<br />

Mitsuyasu, Ronald T. 4030<br />

Mittal, Bharat Bhushan 5500,<br />

e18523<br />

Mittar, Priyanka e11057<br />

Mittempergher, Lorenza 3065<br />

Mittendorf, Elizabeth Ann<br />

625, 1028, 1032, 1107,<br />

1130, 2508, 2529<br />

Mittermueller, Johann 3588<br />

Mittra, Arjun e19506<br />

Miura, Daishu 591, 1106,<br />

TPS1141<br />

Miura, Satoru 7561<br />

Miura, Yasushi e17020<br />

Miura, Yousuke e18037<br />

Miwa, Keisuke 3084<br />

Miwa, Shinji 10075<br />

Miyagishima, Takuto 3593<br />

Miyajima, Akira 9569<br />

Miyakawa, Hiroyuki 4031<br />

Miyake, Hideaki e15048<br />

Miyamoto, Atsushi e14518<br />

Miyamoto, David Tomoaki<br />

4566<br />

Miyamoto, Natsuko e18036,<br />

e18042<br />

Miyamoto, Yuji TPS3642,<br />

e13553<br />

Miyamura, Koichi 6533<br />

Miyashita, Minoru e11530<br />

Miyata, Jun e18058<br />

Miyata, Yoshihiro 7031<br />

Miyazaki, Jun e15004<br />

Miyazaki, Masaki e18058,<br />

e18060<br />

Miyoshi, Eiji e17520<br />

Miyoshi, Shinichiro e17517<br />

Miyoshi, Sosuke e21059<br />

Miyoshi, Yasuo e13037<br />

Mizoguchi, Akira e21053<br />

Mizoguchi, Kosuke 7569<br />

Mizumoto, Kazuhiro 4035<br />

Mizuno, Nobumasa 4042,<br />

e14540<br />

Mizuno, Ryuichi 9569<br />

Mizuno, Toshiro e19546<br />

Mizuno, Yoshio e11007<br />

Mizunoe, Tomoya 5084<br />

Mizunuma, Nobuyuki 10541,<br />

e14145<br />

Mizusawa, Junki 3524, 3569,<br />

4031, 5542, 6112, 7028,<br />

e14510<br />

Mizutani, Yoichi e15000<br />

Mjaaland, Odd 4066<br />

Mjaaland, Svein 4066<br />

Mladosievicova, Beata 9592<br />

Mlineritsch, Brigitte 514<br />

Mo, Frankie 5511, e14085,<br />

e14528<br />

Mo, Jinggang e14523<br />

Moachon, Gilbert 3052<br />

Mobley, Gary M. 9118<br />

Mocci, Evelina e12033<br />

Moccia, Alden A 8049<br />

Mocellin, Simone e12037<br />

Moch, Holger 2055<br />

Mochizuki, Hitoshi e14622<br />

Mocquery, Magali TPS9154<br />

Modak, Anil S e13086<br />

Modak, Shakeel TPS9595<br />

Modena, Yasmina e13019<br />

Modest, Dominik Paul 3519^,<br />

3588, TPS3639, 4023,<br />

e14043<br />

Modi, Shanu 528, 10514,<br />

10618<br />

Modi, Tejas e12025<br />

Modi, Yashpal e12025<br />

Modiano, Manuel R. 7541,<br />

7556<br />

Modonesi, Caterina e13019<br />

Moehle, Robert 2007, 8031<br />

Moehler, Markus Hermann<br />

TPS3641^, 4090,<br />

TPS4138, TPS4140<br />

Moenig, Stefan Paul 4090<br />

Moertel, Christopher L. 2546<br />

Moes, Johannes 2550<br />

Moeslein, Fred M 3590<br />

Mogal, Harveshp D. e19593<br />

Mogg, Robin 3531, 3587<br />

Mogha, Ariane e19037<br />

Moghadamfalahi, Mana 5095<br />

Moghaddamjou, Ali 6085<br />

Mognetti, Thomas TPS646,<br />

10049<br />

Mogushi, Kaoru e11065<br />

Mohamed, Amin Eltigani<br />

e15530<br />

Mohamed, Ateesha F 4608<br />

Mohamed, Hesham 6506<br />

Mohamed, Mohamed K. 7005<br />

Mohamed, Tarek Yakout<br />

e18570<br />

Mohammad, Helai P e18147<br />

Mohammad, Waleed e14154<br />

Mohammed, Tamer e17556<br />

Mohammed, Zahra MA<br />

e14054<br />

Mohan, Anant e17505<br />

Mohanti, B. K 5104, e11013,<br />

e17505<br />

Mohanti, BK 5540<br />

Mohar, Alejandro 1607,<br />

e12027<br />

Mohd Sharial, Mohd Syahizul<br />

Nuhairy e17561<br />

Mohile, Supriya Gupta 9009,<br />

9010, 9014, 9028, 9034,<br />

9091, 9109, 9113, 9123,<br />

9141, 9142, e18133<br />

Mohler, James e15203,<br />

e15204<br />

Mohr, Peter 8505, LBA8509<br />

Mohr, Raphael e12015<br />

Mohrbacher, Ann 8073<br />

Mohri, Yasuhiko e14029,<br />

e14039, e14541, e21053<br />

Mohrmann, Svjetlana e21020<br />

Mohty, Mohamad 6534, 6542<br />

Moinpour, Carol 4571,<br />

CRA6009, 9018<br />

Moiseyenko, Vladimir<br />

Mikhailovich 3505<br />

Moiseyenko, Vladimir e13059<br />

Mojtahedi, Golnessa 5026<br />

Mok, Tony LBA7500, 7519^,<br />

7530, TPS7614, TPS7615,<br />

TPS7616, e18063^<br />

Mokhtari, Karima 2023<br />

Molchanova-Cook, Olga<br />

e13075<br />

Molderez, Christoph 1129<br />

761s


Molero, Xavier e21035<br />

Moles-Moreau, Marie-Pierre<br />

8026<br />

Moley, Jeffrey F. 2525<br />

Molhoek, Kerrington R. 8530,<br />

8597<br />

Molica, Carmen e18023,<br />

e18101<br />

Molica, Stefano e18564<br />

Molife, L Rhoda 2530, 3021,<br />

3030, 3048, 3051, 3080,<br />

10525<br />

Molina, Ana M. 4542,<br />

TPS4682<br />

Molina, Arturo TPS666,<br />

LBA4518, 4521, 4556,<br />

4558, 4664, e15137<br />

Molina, Ashley 1608<br />

Molina, Franck 10505<br />

Molina, Julian R. 1565,<br />

3001, 5544, 7073, 7555,<br />

7605<br />

Molina, Matias e11044,<br />

e11501, e18125<br />

Molina, Thierry Jo 8021<br />

Molina-Garrido, MJ e12033,<br />

e19590<br />

Molina-Vila, Miguel Angel<br />

4068, 7522, e14521,<br />

e18130, e18131, e18143<br />

Molinaro, Annette M 2026<br />

Moline, Teresa e21021<br />

Molinier, Oliver 7057,<br />

TPS7112, LBA7507<br />

Molino, Annamaria 586<br />

Molinolo, Alfredo e16061<br />

Molins, Carmen 4587<br />

Molins, Joaquim Bellmunt<br />

4522, 4525, 4577, 4580,<br />

4582, 4585, e15001,<br />

e15041<br />

Molkenboer-Kuenen, Janneke<br />

D.M. 3035<br />

Mollee, Peter 8030<br />

Mollema, R 7009<br />

Moller, Jakob M. 2069<br />

Mollevi, Caroline 10505<br />

Molnar, Erin 6508<br />

Moloney, Julie 3537<br />

Molteni, Agostino e21085<br />

Momen, Lamia 1006<br />

Momiyama, Masashi 2044,<br />

10072, e14014<br />

Mon, Yin Yin e11078<br />

Monaco, Francesco e18039<br />

Monarca, Bruno e17006,<br />

e18563, e18566<br />

Monasterolo, Gabriella 10055<br />

Monclair, Tom 9583<br />

Mondejar Solís, Rebeca<br />

e14058<br />

Monden, Kazuya 7528,<br />

10610, e18134<br />

Monden, Nobuya 5542<br />

Monestier, Sandrine 8555,<br />

8568, 8578<br />

Monfardini, Lorenzo e14607<br />

Monfardini, Silvio e14607<br />

Monforte, Joseph 1058,<br />

e14164<br />

Monga, Dulabh K. e21036<br />

Monges, Genevieve 4129<br />

Mongort, Cristina e14589<br />

Mongoue-Tchokote, Solange<br />

e16045<br />

Monica, Valentina TPS7109<br />

Monika, Brueggemann 6500^<br />

Monjazeb, Arta 3566<br />

Monk, J. P. 4511, 4655,<br />

4663, 4667, 10503<br />

Monnereau, Alain e14168<br />

Monnet, Elisabeth 7057<br />

Monnet, Isabelle TPS7112,<br />

7574, e18089, e18102<br />

Monnier, Justin e21071<br />

Monreal, Mayte 7095<br />

Monroe, Philip J. 9575<br />

Monson, Kathryn 5527<br />

Montagna, Daniela e13103<br />

Montagut, Clara e15111<br />

Montalar, Joaquin 10082<br />

Montañana, Myriam 8572<br />

Montanari, Bruno 1115<br />

Montandon, Nicole e19050<br />

Montaner, David 7060<br />

Montas, Sacha M 9121<br />

Montazeri, Amir H e15128,<br />

e15135, e18068<br />

Montbarbon, Xavier e16036<br />

Montcuquet, Philippe e11019<br />

Montefusco, Mary 2521,<br />

8549, 8598<br />

Montemurro, Michael 10033<br />

Montemurro, Severino e21134<br />

Montenegro, Alexander<br />

TPS5600, 7511<br />

Montera, Roberto 5093,<br />

5094, e15547, e15550,<br />

e15551, e15553<br />

Montero, Alberto J. 4043,<br />

9565, e11038, e15107,<br />

e16525<br />

Montero, Maria Angeles 7041<br />

Montero, Pablo H. 5562<br />

Montes, Ana 7544<br />

Montes, Marta e14057<br />

Montesa, A. 4630<br />

Montesarchio, Vincenzo 4115<br />

Montestruc, Francois 10007<br />

Monteverde, Martino 2600<br />

Montgomery, Robert B. 4520,<br />

4521, 4665, TPS4695<br />

Monti, Maria Gaia e11052<br />

Monti, Nadia 1500<br />

Montini, Enrica e13103<br />

Montironi, Rodolfo e15074<br />

Montone, Eva 5093, e15551,<br />

e15553<br />

Montoto, Silvia 8058<br />

Monzo, Mariano e13540<br />

Monzon, Federico A. 4624<br />

Monzon, Francisco e14057<br />

Monzon, Jose Gerard e13049<br />

Moo, Tracy-Ann 1118<br />

Mookerjee, Bijoyesh<br />

TPS6643^<br />

Moon, Hyeong Gon 1133,<br />

e21160<br />

Moon, Hyeong-Gon 1056,<br />

10621, e11532<br />

Moon, James 5015, 7005,<br />

7018, 7083, 7517, 7552,<br />

7570, 8504, 8521, 8525,<br />

8527<br />

Moon, Joon Ho 3554, 6536,<br />

6538, e18533<br />

Moon, Woo Kyung e11532,<br />

e21160<br />

Mooney, Kathi 9137<br />

Moorahrend, Enno 4065<br />

Moore, Alexis 6017, 6029<br />

Moore, Amy Patricia 3058,<br />

e13000<br />

Moore, Andrew Jacob e19559<br />

Moore, Dan H. 9107, 10586<br />

Moore, David J e15141<br />

Moore, Dominic T 6107<br />

Moore, Halle C. F. 1076<br />

Moore, Joanne e19519<br />

Moore, Kathleen N. 3008,<br />

3041, 3067, 3097, 5012,<br />

TPS5116<br />

Moore, Malcolm J. 3082,<br />

3504, 3515, 3572, 4047,<br />

4058, TPS4134, TPS4135,<br />

4535, 9003, e14524,<br />

e14592, e19522<br />

Moore, Marietta L. e15179<br />

Moore, Nicholas e14168<br />

Moore, Nicola TPS9597^<br />

Moore, Patrick S. 8577<br />

Moore, Rebekah 1516<br />

Moore, Sara 4058<br />

Moore, Theodore 6537<br />

Moore, Timothy David<br />

TPS3110<br />

Moore, William R 4671,<br />

e15167<br />

Moorman, Meredith e17001<br />

Moorthy, Ganesh TPS3116<br />

Mora, Serena 519<br />

Morabito, Fortunato 8083,<br />

e18564<br />

Moraco, Nicole H. 10001,<br />

10019<br />

Moral, Antonio 1120<br />

Morales La Madrid, Andres<br />

Eduardo 9534<br />

Morales Pancorbo, David<br />

e11075<br />

Morales, Rafael 2540,<br />

e15001, e21021<br />

Morales, Rita 10004<br />

Morales, Serafin 1011,<br />

10512, 10595, e11545<br />

Morales-Barrera, Rafael<br />

TPS4674<br />

Morales-Espinosa, Daniela<br />

e11098, e15569<br />

Morales-Vasquez, Flavia<br />

e15527<br />

Moran, Ann Marie e13574<br />

Moran, Cesar 7023<br />

Moran, Diarmuid M e21120<br />

Moran, Jon e17550<br />

Moran, Kirsty 10525<br />

Moran, Susan 4549, 4656<br />

Moran, Teresa 3093, 7011,<br />

7522, 7532, 7542, 7543,<br />

e18131, e18137<br />

Mordechovich, Daniel e14616<br />

Mordenti, Joyce TPS668<br />

Moreau, Michel 3559<br />

Moreau, Philippe 8009,<br />

8014, 8020, 8095,<br />

TPS8112, TPS8113,<br />

TPS8114, e18572^<br />

Moreau, Sandrine e13605<br />

Moreau, Stephanie 8540<br />

Moreau, Thierry e14006<br />

Moreira, António e13119<br />

Moreira, Rubem e14027<br />

Morelli, Emanuela 8066<br />

Morelli, Franco 5513<br />

Morelli, María Pia 4040<br />

Moreno, Antonio e16000<br />

Moreno, Carolina 2612<br />

Moreno, Jesus Zamora 7068<br />

Moreno, Juan 3618, 10547<br />

Moreno, Lucas 9542<br />

Moreno, Ofir 3067<br />

Moreno, Victor 3051, 3080,<br />

3510, 3601, 3618, 10547,<br />

e13085, e13599, e13601,<br />

e14093<br />

Moreno-Aspitia, Alvaro<br />

CRA1002, 1083, 6570,<br />

e13501<br />

Moreno-Eutimio, Mario Adan<br />

e15584<br />

Moreno-Fernandez, Debora<br />

3014<br />

Morere, Jean F. 1095, 1567,<br />

1568, 5067, e18003,<br />

e18020<br />

Morero, Jose Luis 7029<br />

Moretto, Patricia e15537<br />

Morgades, Mireia 8005<br />

Morgan, Adrienne TPS665<br />

Morgan, Gareth J 8015, 8095<br />

Morgan, Jeffrey A. 1547<br />

Morgan, Jessica 575<br />

Morgan, John 1587<br />

Morgan, Michael A. e15190<br />

Morgan, Paul TPS9596<br />

Morgan, Richard 4653<br />

Morgan, Robert 3054, 3108,<br />

5020, 5025<br />

Morgan, Shethah 4124,<br />

e13541<br />

Morgan, Susan Kay 1018<br />

Morgans, Alicia Katherine<br />

6045<br />

Morgenfeld, Eduardo L. 9038,<br />

e11037, e15507, e15508<br />

Morgenstern, Sara e14067<br />

Morgensztern, Daniel 5529,<br />

5532<br />

Morgenthaler, Nils G. 10536,<br />

10537<br />

Mori, Kiyoshi 7017, 7021,<br />

7070<br />

Mori, Masahide e18007,<br />

e18025, e18134<br />

Mori, Tsuyoshi e11564,<br />

e13559<br />

Mori, Yoshiaki 7565<br />

Morice, Philippe 5028, 5083,<br />

10098<br />

Morikawa, Naoto 7086, 7561,<br />

7563, 7565, e19508<br />

Morimoto, Masami e11560<br />

Morimoto, Yuhki e21053<br />

Morin, Franck TPS7111,<br />

TPS7112, 10507, e18089<br />

Morioka, Junko e18059<br />

Morioka, Toru 565<br />

Morishima, Yasuo 6533<br />

Morishita, Naoko e18036,<br />

e18042<br />

Morita, Craig T e21004<br />

Morita, Kazutoyo e14602<br />

Morita, Masaru e14601<br />

Morita, Satoshi 1106, 4002,<br />

7521, 9574, e14123,<br />

e18007, e18025, e18060,<br />

e18129<br />

Morita, Shane Young e19051<br />

Morita, Yoshitaka TPS3642<br />

Moriwaki, Toshikazu 4002<br />

Moriya, Yoichiro e18091<br />

Moriya, Yoshihiro 3524, 3569<br />

Morizane, Chigusa 4031<br />

Morland, Bruce 9542<br />

Moro-Sibilot, Denis TPS7112,<br />

7533, e18089<br />

Morosi, Carlo 4507, 10026<br />

Morosky, Anne e13599<br />

Moroso, Stefano 1044<br />

Morote, Juan 4510<br />

Morozov, Alexei 5012<br />

Morrell, Glen 3057<br />

Morris, Carol 10022<br />

Morris, David E. 2091<br />

762s Numerals refer to abstract number


Morris, David e18098<br />

Morris, Donald e18110<br />

Morris, Elizabeth Ann e11531<br />

Morris, G. Stephen e17002<br />

Morris, Jackie e14587<br />

Morris, Jeffrey 3598, 4116<br />

Morris, John Charles 2571,<br />

e21064<br />

Morris, Karen e21037<br />

Morris, Michael J. 4517,<br />

4548, 4568, 4655, 4667,<br />

TPS4694<br />

Morris, Patrick Glyn 2006,<br />

10514<br />

Morris, Shannon Renae<br />

e13596<br />

Morris, Stephan 3090<br />

Morris, Van Karlyle 3561<br />

Morrison, Carl D e11563,<br />

e15203<br />

Morrison, Debra J. 9507<br />

Morrison, Devi 6573<br />

Morrison, Jodie 4665<br />

Morrison, P. J. 1543<br />

Morrison, Rosemary 2552<br />

Morrison, Tara e11085,<br />

e12509<br />

Morrison, William H. 5561,<br />

5589<br />

Morrissey, Stephanie 4504<br />

Morrone, Robert j 2587<br />

Morrow, Gary R. 9009, 9010,<br />

9014, 9027, 9028, 9113,<br />

9141, 9142, e16056,<br />

e19510<br />

Morrow, Kevin 6580<br />

Morrow, Phuong Khanh H.<br />

9061, 9072<br />

Morschhauser, Franck 8002,<br />

8030, 8057<br />

Morse, Linda K. 7099<br />

Morse, Michael 2517, 2585,<br />

2588, 10563<br />

Morsi, Amr M. e19015<br />

Mortarotti, Joan 6108<br />

Mortensen, Deborah 3006^<br />

Mortensen, Michael Bau<br />

e14633<br />

Mortier, Laurent 1566, 8559,<br />

8591, 8601<br />

Mortimer, Joanne E. 639,<br />

1010, 1070, e13116,<br />

e19644<br />

Mortimer, Peter 4549<br />

Morton, Donald L. 2519,<br />

8534, e19029<br />

Morton, Ronald A. e19582<br />

Morvan, Francois e16044<br />

Morvillo, Manfredi 3555<br />

Moscetti, Alessandro e17006,<br />

e18563, e18566<br />

Moscetti, Luca e12514,<br />

e18065<br />

Moschos, Stergios J. 8533,<br />

8547<br />

Moschos, Stergios J 8503^,<br />

8528<br />

Moschovi, Maria e20002<br />

Moscinski, Lynn e18503,<br />

e18506<br />

Moscol, Giancarlo 7051<br />

Mosconi, Massimo e19035,<br />

e19036<br />

Moseley, Jennifer L. e15116<br />

Moseley, Paul 1013, 1098<br />

Mosenkis, Jeffrey 9104<br />

Moser, Barry K 6526<br />

Moser, Lutz 5512<br />

Moser, Rachel 6091, e16007<br />

Numerals refer to abstract number<br />

Moshfegh, Ali 6557<br />

Moshier, Erin L. 4524, 6065,<br />

e21032, e21109<br />

Moshkowitz, Menachem 1564<br />

Moskowitz, Chaya S.<br />

CRA9513, 9552<br />

Moskowitz, Craig 2008, 2089<br />

Moskvina, Ekaterina A.<br />

e19012<br />

Mosley, Jane e19522<br />

Mosquera, Juan Miguel 4649<br />

Moss, Ralph W e13102<br />

Moss, Rebecca Anne 2534,<br />

TPS3112<br />

Moss, Wendy 9589<br />

Mosse, Yael P. 9500<br />

Mostafa Kamel, Yasser<br />

Mostafa 9117<br />

Mostafa, Magda 5557<br />

Mostafa, Nael 2013<br />

Mostaghel, Elahe A. 4520<br />

Mostert, Bianca 10504<br />

Motamed, Kouros e15542<br />

Motamed-Khorasani, Afsaneh<br />

e17521<br />

Motamedinia, Piruz e15021<br />

Motley, Christine 6025<br />

Motoi, Noriko e18017<br />

Motola, Daniel e11098<br />

Motomura, Kazuyoshi 1103,<br />

1106<br />

Motomura, Takashi e14536<br />

Motoshima, Kohei 7569<br />

Motsinger-Reif, Alison A.<br />

10515<br />

Mott, Judith H. e13522<br />

Motta, Leonardo e21042<br />

Mottolese, Marcella 1050,<br />

e14042, e18021<br />

Motzer, Robert John 4501,<br />

4532, 4542, 4546, 4604,<br />

TPS4682, TPS4683, 6092,<br />

e15070<br />

Mouarrawy, Doraid 10540<br />

Mouchet, Nicolas e19037<br />

Mouillet, Guillaume 7057<br />

Moul, Judd W. 4670, e15113<br />

Mould, Diane R. 2597<br />

Moulder, Stacy L. 515, 527,<br />

10510, 10559, 10613<br />

Mouliere, Florent 10505<br />

Moulinoux, Jacques Philippe<br />

e15131<br />

Moulton, Brian e13520<br />

Mounier, Nicolas 5075,<br />

8002, 8005, 8021, 8046,<br />

8048, 8077, e15574<br />

Mountz, James M 3019<br />

Moura, Shari e19522<br />

Mourad, Waleed Fouad 5531,<br />

5559, 5581, e15138<br />

Mouradov, Dmitri 3623<br />

Mourah, Samia 8048<br />

Moureau-Zabotto, Laurence<br />

10042<br />

Mouret, Jean-Francois 10562<br />

Mouret-Reynier, Marie-Ange<br />

1051, 5018<br />

Mourey, Loic LBA4567^,<br />

TPS4692^<br />

Mourlane, Frederic 2575<br />

Mouro, Larissa R 1075<br />

Mouroux, Jerome 7591<br />

Mourouzis, Iordanis e11073<br />

Moussa, Ali H. 8531<br />

Moussy, Alain LBA4007,<br />

10007, 10089,<br />

TPS10102^<br />

Moutinho, Catia 3570<br />

Movassaghi, Kamran e15053<br />

Movva, Sujana TPS10103<br />

Mowat, Rex Bradford 1083<br />

Moxhon, Anne e14060<br />

Moy, Beverly 596, 599, 604,<br />

6095, 6128<br />

Moya Gomez(2), Paloma<br />

e11543, e19596<br />

Moya, Beatriz e12003<br />

Moya, Irene TPS3637<br />

Moye, Virginia Ann 5577<br />

Moyer, Ann M. 5077<br />

Moyers-Ruiz, Liliana TPS9148<br />

Moylan, Eugene J. 2062,<br />

e11569, e16506<br />

Moynihan, Timothy Jerome<br />

534<br />

Moyo, Victor M. TPS663,<br />

TPS3111, 7556<br />

Mozayen, Mohammad 7102,<br />

e14046<br />

Mozer, Pierre e15105<br />

Mozley, P. David 10573<br />

Mozzillo, Nicola 8534, 8573,<br />

8581, e19034<br />

Mpasi, Priscilla e18550<br />

MU, Hua 3038, 3040<br />

Mu, Zhenyu 8583<br />

Mubdi, Nidha Z. CRA9513<br />

Mucci, Lorelei 4555<br />

Mucciarini, Claudia 2057<br />

Muche, Rainer TPS1612<br />

Mudad, Raja e17564<br />

Muddu, Vamshi 5585,<br />

e16028, e17500<br />

Mudunuru, Uma 10605<br />

Muehlenbein, Catherine E.<br />

5533, e16053<br />

Mueller, Andreas 9059<br />

Mueller, Berit 1023, 10627<br />

Mueller, Carsten 8024, 8025<br />

Mueller, Stefan e15112<br />

Mueller, Volkmar 1090,<br />

TPS1146, 10599<br />

Mueller-Huebenthal, Jonas<br />

e13508<br />

Muffly, Lori S. 8060<br />

Muggeri, Alejandro Daniel<br />

e12511<br />

Muggia, Franco 5016, 5041,<br />

5078, e11010, e11529<br />

Mugishima, Hideo 9574<br />

Mugiya, Soichi e15098<br />

Muhr, Daniela 1537<br />

Muindi, Josephia e18118<br />

Muinelo-Romay, Laura 10534<br />

Muiruri, Charles 6102<br />

Mukai, Harumi 8094<br />

Mukai, Hir<strong>of</strong>umi 512, 540,<br />

585<br />

Mukai, Masatoshi e15065,<br />

e15069<br />

Mukherjee, Bhramar e12031<br />

Mukherjee, Debraj 2080<br />

Mukherjee, Srijita e14632<br />

Mukherji, Bijay e21135<br />

Mukherji, Deborah 4553,<br />

e15134, e15136<br />

Mukhopadhyay, Ashis<br />

e13030, e18043<br />

Mukhopadhyay, Pabak 551,<br />

TPS648<br />

Mukhopadhyay, Soma<br />

e13030, e18043<br />

Mukhopadhyay, Sutapa 8012<br />

Mukohara, Toru e16034<br />

Mukonje, Terence 1089<br />

Mulcahy, Hugh e13570,<br />

e21024<br />

Mulcahy, Mary Frances 3608,<br />

4049, 4077, 4102<br />

Mulder, Ina 8039<br />

Mulder, Sasja 9070<br />

Mulders, Peter LBA4518,<br />

4519^<br />

Muldoon, C. 1543<br />

Muldoon, Leslie L. 2040<br />

Mulet-Margalef, Nuria 619<br />

Mulick Cassidy, Amy 4553,<br />

10525, e15134, e15136<br />

Mulkerin, Daniel 3101, 3103,<br />

e14554<br />

Mull, Ruslan e15576<br />

Mullady, Daniel e14584<br />

Mullan, Mohmaduvesh<br />

e19581<br />

Mullan, Paul B 10553<br />

Mullane, Kathleen M. 9067<br />

Mullane, Michael Russell<br />

9085, e14186, e14590<br />

Mullany, Lisa K e15583<br />

Mulle, Lynette 3001<br />

Mullen, John Thomas 10058<br />

Muller, Leandro Bizarro<br />

e14597<br />

Muller, Stephanie 4562<br />

Muller, Susan 5560<br />

Muller-Greven, Ga‘lle 2014<br />

Mullighan, Charles Grenfell<br />

9506<br />

Mullins, C. Daniel e14035,<br />

e14056, e14522, e14626,<br />

e14638, e15108, e15146,<br />

e15148<br />

Mullooly, Maeve 622<br />

Mulquin, Marcia 4569, 8088,<br />

8104, e18568<br />

Mulrenan, Heather a 5534<br />

Mulrooney, Daniel A. e16502<br />

Muluneh, Benyam 6042<br />

Mulvenna, Paula M e17519<br />

Mulvihill, Erica e14666<br />

Mummy, David G 6007,<br />

e14068<br />

Mun, Yong e11051<br />

Munck af Rosenschold, Per<br />

e18527<br />

Mundhenke, Christoph 556,<br />

1084, e13503<br />

Mundia, Abdul G. e13053<br />

Mundia, Mubasir e13053<br />

Munemoto, Yoshinori<br />

TPS3642<br />

Munhoz, Rodrigo Ramella<br />

e12513<br />

Munive, Carlos e11066<br />

Munkarah, Adnan 5088<br />

Munn, David 2501<br />

Munoz Llarena, Alberto<br />

e11525<br />

Muñoz Martin, Andrés Jesús<br />

e14058<br />

Munoz Platon(3), Enriqueta<br />

e19596<br />

Muñoz, Alberto e17522,<br />

e18031, e21009<br />

Munoz, Carmen 3096,<br />

e13540<br />

Munoz, Edgar e16566<br />

Muñoz, Eva e19023<br />

Munoz, Francisco e16500<br />

Munoz, Silvia 1588<br />

Muñoz-Couselo, Eva 509,<br />

566, 619<br />

Muñoz-Langa, José e17545,<br />

e18049<br />

Munoz-Mateu, Montserrat<br />

1001<br />

763s


Munoz-Sanchez, MM e19590<br />

Munsell, Mark F. 9061<br />

Munshi, Hidayatullah G.<br />

e14515<br />

Munster, Pamela N. TPS663,<br />

3006^, 3058, 4102, 4660,<br />

TPS8602, e13000, e13596<br />

Munzert, Gerd Michael<br />

3018^, 7557<br />

Munzone, Elisabetta 546,<br />

1049, 1115<br />

Muppidi, Samatha 8100,<br />

8101<br />

Mura, Donna 5027<br />

Murad, Andre 7506<br />

Murad, Faris e14584<br />

Murahashi, Mutsunori e13014<br />

Murakami, Haruyasu 7088,<br />

e21007<br />

Murakami, Keiichi e11017<br />

Murakami, Yoshihiro e21059<br />

Murali, Sujatha 3094<br />

Murata, Akihiko TPS3642<br />

Murata, Kohei e14091<br />

Murata, Paola 5595, e14104,<br />

e15159<br />

Murata-Hirai, Kaoru e21004<br />

Murawa, Dawid 10537<br />

Murawa, Pawel 10537<br />

Murawska, Magdalena<br />

e11553<br />

Murawski, Niels 8022, 8024,<br />

8025<br />

Muren, Caroline 2589<br />

Murgia, Viviana e14711<br />

Murgo, Anthony J. 8073<br />

Murgo, Roberto e21092<br />

Murias, Adolfo 10608,<br />

e11535, e21088<br />

Murillo, Raquel 1111<br />

Murli, Sumati e13114<br />

Murnane, Andrew e19605<br />

Muro, Kei 4067, 4082,<br />

e14500<br />

Muro, Kenji 2035<br />

Murone, Carmel 3534<br />

Murphy, Adrian Gerard<br />

e13570<br />

Murphy, Anne Marie 1553,<br />

6120, e16531<br />

Murphy, Barbara A. 9024,<br />

e16038, e19559<br />

Murphy, Cheryl F. 8530,<br />

8597<br />

Murphy, Christi 4102<br />

Murphy, Conleth G. e14111<br />

Murphy, Erin Sennett 2076,<br />

2100<br />

Murphy, James e13022<br />

Murphy, Janet E. 3594,<br />

e14615<br />

Murphy, Joan 5105<br />

Murphy, Kate e14111<br />

Murphy, Kevin C. 6071<br />

Murphy, Leigh TPS1136<br />

Murphy, Patrick Brian 618,<br />

TPS8115<br />

Murphy, Thomas J e14666<br />

Murphy, Valerie 7593<br />

Murray, H. A. e14022<br />

Murray, James L. 527,<br />

e11090<br />

Murray, Jonathan 4660<br />

Murray, Nevin 7020, e18079<br />

Murray, Nicholas TPS1144<br />

Murray, Paul Gerard e15091<br />

Murray, Samuel e21087,<br />

e21141<br />

Murray, Sharon Cornell 3000,<br />

5065<br />

Murray, Susan 2062, e11569<br />

Murray, Tom e15169<br />

Murtaza, Muhammed 544<br />

Murthy, Rashmi Krishna 527<br />

Murthy, Vedang e16021<br />

Murtra-Garrell, Nuria 7041<br />

Musa, Zahra 6077<br />

Muscarella, Peter e14501<br />

Muscato, Joseph J. TPS658<br />

Muschen, Markus 9555<br />

Muse, Kimberly I. 1549<br />

Musgrove, Elizabeth A. 504<br />

Musib, Luna C. 2566,<br />

TPS2616, 3021, e13114<br />

Muslimani, Alaa 628, e17015<br />

Musolino, Caterina e18564<br />

Muss, Hyman Bernard<br />

TPS656, 1524, 6015,<br />

6017<br />

Muss, Hyman 5577<br />

Mussari, Salvatore e14711,<br />

e21102<br />

Mustacchi, Giorgio 579, 586<br />

Mustafa, Rashid 1032<br />

Mustea, Alexander 5007<br />

Mustian, Karen Michelle<br />

9009, 9010, 9014, 9028,<br />

9141, 9142<br />

Musulen, Eva 4089<br />

Musuuza, Jackson S. 1598<br />

Mutale, Faith e16062<br />

Mutalima, Nora 9558<br />

Mutch, David Gardner 1519,<br />

1520, 5013, 5101,<br />

e15528<br />

Mutchler, Jan e16500<br />

Muth, Mathias e13503<br />

Muthen, Noemi 3044<br />

Muthukaruppan, Raman 4106<br />

Muto, Giovanni e15133<br />

Muto, Hiroki e13511<br />

Muto, Jun e14536, e14602<br />

Muto, Osamu e14062<br />

Muto, Tetsuichiro 3625<br />

Muto, Yoichi e14722<br />

Muus, Petra 6522<br />

Muwakkit, Samar e20004<br />

Muzaffar, Mahvish e14693<br />

Muzi, Mark TPS10635<br />

Muzii, Ludovico e15547,<br />

e15550<br />

Muzikansky, Alona 6136,<br />

7010, 7524<br />

Mwamburi, Mkaya 6553<br />

Mwandoro, Tibu N. 3047<br />

Myers, Jay T. 9520<br />

Myers, Jeffrey 5524<br />

Myers, Lee 1128<br />

Myers, Robert Edward<br />

e19570<br />

Mühlenberg, Thomas 10032^<br />

Mykletun, Andrew 10061<br />

Mykulowycz, Kristine 4111<br />

Myles, Bevan H e14548<br />

Müller, Lothar 556, e15520<br />

Müller, Martin C 6503, 6510<br />

Müller, Stefan 5508<br />

Münstedt, Karsten 5005,<br />

5044<br />

Myo, Aung e13061<br />

Myrand, Scott 7554, 7583<br />

Myskowski, Patricia L.<br />

e19052<br />

Myssiorek, David e16052<br />

Mytilineos, Joannis 6567<br />

Mægbæk, Merete Lund 1579,<br />

e12023<br />

Möbs, Markus e15053<br />

Møller, Henrik 7026<br />

N<br />

N, Soumitra e18041<br />

Nabell, Lisle 1006<br />

Nabhan, Chadi 8072, 8080,<br />

e15120<br />

Nabholtz, Jean-Marc A.<br />

TPS646<br />

Nabholtz, Jean-Marc 1051<br />

Nabid, Abdenour TPS5600<br />

Nacci, Angelo e11001,<br />

e11547, e18048<br />

Nachman, James B. CRA9508<br />

Nacul, Mario Javier e11037<br />

Nadeau, Laura e17015<br />

Nadeau-Larochelle, Corinne<br />

634<br />

Nademanee, Auayporn 6545,<br />

e18530<br />

Nader, Helena Bonciani<br />

10568, e13016<br />

Nadig, Juerg e19648<br />

Nadler, Eric e20510<br />

Naermann, Regina 4600<br />

Nafa, Khedoudja 10560<br />

Nafissi, Julieta e14109<br />

Nag, Shona Milon 3011<br />

Nagai, Kanji 1552, 7044,<br />

e17526, e17554, e18054,<br />

e18064<br />

Nagai, Shigenori E 613<br />

Nagai, Yohei e13553<br />

Nagai, Yutaka 5086<br />

Nagalla, Balakrishna 6554<br />

Nagamatsu, Hiroaki e14080<br />

Nagamine, Shinji 1119<br />

Nagamitsu, Yuzo 10557<br />

Nagano, Hiroaki e14518<br />

Nagao, Shoji 5103<br />

Nagaoka, Akira e15057,<br />

e15088<br />

Nagaoka, Isao e14004<br />

Nagaraj, Gayathri<br />

Nagaraj, Kanakapura<br />

TPS664<br />

Ramanna e11027, e14675,<br />

e14721<br />

Nagarajah, James 10032^<br />

Nagarajan, Rajaram e20007<br />

Nagarkar, R. 3089^<br />

Nagarwala, Yasir M. TPS658<br />

Nagasaka, Tsuneki 5037<br />

Nagase, Michitaka 4002,<br />

4031<br />

Nagashima, Takeshi 1106<br />

Nagata, Hirohiko 9569<br />

Nagata, Kazuma 7528,<br />

10610<br />

Nagata, Misato e17538<br />

Nagata, Naoki TPS3642<br />

Nagata, Takuya e12028<br />

Nagatomo, Izumi e18056<br />

Nagayama, Satoshi 3625,<br />

e14145<br />

Nagel, Christa Irene e15501<br />

Nagengast, Wouter B.<br />

Naghashpour, Mojdeh<br />

10581<br />

e18503, e18506<br />

Nagourney, Robert Alan<br />

e14075<br />

Nagpal, Sunil 3619, e14046,<br />

e15554<br />

Nagre, Sandeep 3015, 10036<br />

Nagura, Naoya e13037<br />

Nagy, Cindy K 9064<br />

Nahi, Hareth 8019<br />

Nahleh, Zeina A. e11549<br />

Naidoo, Jarushka 2609<br />

Naidu, K. V. Jagannath Rao<br />

9055<br />

Naing, Aung 3106, 3107^,<br />

4639, 8551, 10510,<br />

10607, e13020, e13506,<br />

e13534, e13595<br />

Nair, Bijay P. e18548<br />

Nair, Bijay 8041, e18576<br />

Nair, Deepa e16021<br />

Nair, Kavita V. e16541,<br />

e16550<br />

Nair, Nita S. 1108, e11552<br />

Nair, Reena e17003<br />

Nair, Sudhir Vasudevan<br />

e16021<br />

Nair, Suresh G e16561<br />

Nair, Vidya 5582<br />

Naito, Sei e15057, e15068,<br />

e15071, e15088<br />

Naito, Seiji e15048, e15071,<br />

e15098<br />

Naito, Tateaki 7088, e21007<br />

Naito, Yoichi 585<br />

Najdi, Adil e11502<br />

Najera, Jose Eugenio 6546<br />

Nakabayashi, Mari e15170<br />

Nakagawa, Hidewaki 10520<br />

Nakagawa, Kazuhiko 6112,<br />

7003, 7017, 7028,<br />

TPS7110, 7521, 7602,<br />

e18060<br />

Nakagawa, Kei e14011<br />

Nakagawa, Ken 9569<br />

Nakagawa, Misako e11560<br />

Nakagawa, Wilson Toshihiko<br />

e14119<br />

Nakahama, Hiroko e19557<br />

Nakahara, Rie 7021<br />

Nakahara, Takahito e21059<br />

Nakai, Toshiyuki e17538<br />

Nakai, Yasutomo e15065,<br />

e15069<br />

Nakaigawa, Noboru 4626<br />

Nakajima, Go e21108<br />

Nakajima, Takako Eguchi<br />

e14123<br />

Nakajima, Takashi 7088<br />

Nakajima, Yuki 10542<br />

Nakakura, Eric K. 4126,<br />

e14542, e14551<br />

Nakamae, Hirohisa 6509^<br />

Nakamori, Shoji 4031<br />

Nakamoto, Tomoko 5086<br />

Nakamura, Asako 2553<br />

Nakamura, Hiroaki 10073<br />

Nakamura, Hiroko 5084<br />

Nakamura, Kenichi 3524,<br />

5542, 6112, e14510<br />

Nakamura, Masato TPS3642<br />

Nakamura, Michio 3593,<br />

e14062<br />

Nakamura, Ryuji e14011,<br />

e19508<br />

Nakamura, Seigo 590,<br />

e13037<br />

Nakamura, Shinichiro<br />

TPS659, 6112<br />

Nakamura, Terukazu e15055,<br />

e15099<br />

Nakamura, Tsuyoshi e15048<br />

Nakamura, Yoichi 7515,<br />

7569<br />

Nakamura, Yukiko 7088,<br />

e21007<br />

Nakamura, Yusuke 10520,<br />

e13014, e13015, e13029,<br />

e13037, e13552, e13565<br />

Nakanishi, Katsuyuki 1103<br />

Nakanishi, Ryota e14033<br />

764s Numerals refer to abstract number


Nakanishi, Toru 5006<br />

Nakanishi, Yoichi TPS659,<br />

TPS7110, e13014<br />

Nakano, Hir<strong>of</strong>umi 7569<br />

Nakano, Takashi 7031, 7080<br />

Nakanoko, Tomonori e14601<br />

Nakao, Akimasa e14506<br />

Nakashima, Masanao e13029<br />

Nakashima, Osamu 4104<br />

Nakashiro, Koh-ichi 5584<br />

Nakatani, Akinori e14004<br />

Nakatani, Takeshi e18056<br />

Nakatomi, Katsumi 7569<br />

Nakatsuka, Seishi 7038<br />

Nakayama, Norisuke e14123<br />

Nakayama, Shoko e15057<br />

Nakayama, Souhei e18058<br />

Nakayama, Yoshie e19512<br />

Nakazawa, Atsuhito e19583<br />

Nakazawa, Yukie e18007<br />

Nakwan, Narongwit TPS9151<br />

Nam, Byung-Ho 4028<br />

Nam, Hae-Seong e13065<br />

Nam, Joo-Hyun e15559<br />

Nam, Seung-Hyun e19538<br />

Nam, Soon Yuhl 5586<br />

Namakydoust, Azadeh 2561<br />

Namal, Esat e14698<br />

Naman, Herve L. 568^<br />

Namba, Yoshinobu e18007,<br />

e18025, e18134<br />

Namba, Yukiko e18019<br />

Namburi, Swathi 8103<br />

Namendys-Silva, Silvio A<br />

e15527<br />

Namer, Moise e19640<br />

Namiki, Masayuki 3084<br />

Namjoshi, Madhav 593,<br />

e19615<br />

Nanayakkara, Nuwan 529<br />

Nand, Sucha 6502<br />

Nanda, Nisha 6513, 6596<br />

Nanda, Rita 534, 1010<br />

Nandini, Marysingh e11043<br />

Nandini, P K 1093<br />

Nanji, Sulaiman 3632<br />

Nanjo, Shigeki 7528, 10610<br />

Nanni, Cristina e21006<br />

Nanni, Isabelle 8555, 10507<br />

Nanni, Oriana 1099, e11054<br />

Nannini, Margherita 10087<br />

Nanus, David M. 4649<br />

Naoki, Katsuhiko e18058<br />

Naoshy, Sarah e14028,<br />

e14030, e14061<br />

Nappi, Lucia e13509,<br />

e15034<br />

Naraev, Boris G. e14620,<br />

e15580<br />

Naraev, Boris e14600<br />

Narain, Niven R 3015,<br />

e14593<br />

Narala, Neeharika 2558<br />

Narang, Mohit 8037, e18556<br />

Naranjo, Arlene 9533<br />

Narayan, Kailash TPS5116,<br />

e15541<br />

Narayanan, Ramkishen<br />

e15039, e15046<br />

Nardelli, Giovanni B. e12037<br />

Nardone, Beatrice 2532,<br />

4063, 6623, e18571<br />

Narducci, Fabrice 5083<br />

Narducci, Filomena e18065,<br />

e18072<br />

Narimanov, Mekhty e14623<br />

Narine, Nadira e15011<br />

Narisawa, Takafumi e15057<br />

Narod, Steven 1123<br />

Numerals refer to abstract number<br />

Narumi, Sodai 7563<br />

Narvaez, Jose Antonio 10052<br />

Nascene, David 2546<br />

Nascimento, Ellen Caroline<br />

e11014<br />

Nash, Anthony F e15141<br />

Nash, Garrett Michael 3583<br />

Nash, John e15536<br />

Nasim, Muhammad Yaser<br />

2552<br />

Nasioulas, George e21087,<br />

e21141<br />

Naskhletashvili, David R.<br />

e19012<br />

Naso, Virginia e17006,<br />

e18563, e18566<br />

Nason, Katie Sue e14689<br />

Nass, Dvora 2078<br />

Nasseri, Morad 2077<br />

Nassif, George e14157<br />

Nassir, Youram 8098,<br />

e18558<br />

Nasta, Sunita e18529,<br />

e18540<br />

Nasti, Guglielmo e14130<br />

Natale, Romain 5521<br />

Natale, Ronald B. TPS2621<br />

Natali, Maurilio e19002<br />

Nath, Rajneesh 6569<br />

Nathan, Cherie-Ann e16009<br />

Nathan, Hari 3596<br />

Nathan, Paul C. 6027, 6030,<br />

9556, 9586, 9591<br />

Nathan, Paul D. LBA8509,<br />

8523, TPS8605<br />

Nathan, Sunita 6552<br />

Nathanson, Katharine 1539<br />

Nathanson, Katherine L.<br />

1506, 3102<br />

Natrajan, Kavita 6573, 8108<br />

Natsugoe, Shoji 3558<br />

Nattam, Sreenivasa R. 4022<br />

Naucler, Gisela 3538<br />

Naudts, Bart e13073<br />

Naughton, Michael CRA1002,<br />

3047<br />

Naughton, Michelle Joy 9108<br />

Nauman, Deirdre J. e11042<br />

Naumov, George 3587<br />

Naumov, Inna 1564<br />

Nava, Hector R. e14712<br />

Navabi, Jafar 6556<br />

Navale, Lynn 5072<br />

Navaratnam, Sri e17518<br />

Navari, Rudolph M. 9064<br />

Navarrete, Alicia e15111<br />

Navarro Coy, Nuria 8015<br />

Navarro, Alejandro 509, 566,<br />

7041, 10511<br />

Navarro, Jose-Tomas 8005<br />

Navarro, Samuel e14589<br />

Navarro, Tasheda 6507,<br />

6508, 6515^<br />

Navarro-Garcia, Lilian Monica<br />

e11098<br />

Navez, Benoit e14044<br />

Nawarawong, Weerasak 8097<br />

Nawfel, Mellas e11502,<br />

e11516, e14725, e14726,<br />

e18124<br />

Nawras, Ali e14098<br />

Nawrocki, Sergiusz LBA7000,<br />

e19561<br />

Nawrocki, Steffan T 3073<br />

Nayak, Asha e14694, e18024<br />

Naylor, Keith e16509<br />

Nazha, Aziz 6544, 6578,<br />

6589, 6597<br />

Neal, Christopher e13577,<br />

e21028, e21029<br />

Neal, Jennifer 6042<br />

Neal, Joel W. 7010, 7541<br />

Neaume, Julien e18005<br />

Nebot, Noelia 3000<br />

Nebozhyn, Michael 3531,<br />

3587<br />

Necchi, Andrea 4507<br />

Nechushtan, Hovav 535,<br />

10034<br />

Neciosup, Silvia P. e11066,<br />

e11087, e11518, e15036<br />

Necozione, Stefano e11041<br />

Nedelman, Jerry 2603<br />

Neelapu, Sattva Swarup 2528<br />

Neery, Maureen P. 10061<br />

Neff, Tanaya 10591<br />

Negassa, Abdissa e14019<br />

Neglia, Joseph Philip<br />

CRA9513, 9528, 9546,<br />

e16502<br />

Negoro, Shunichi 5045,<br />

7028, 7521, e18060,<br />

e18134, e19548<br />

Negri, Federica 4097<br />

Negri, Tiziana 10026<br />

Negrier, Sylvie CRA4502,<br />

4614, 4625, 8532<br />

Negron, Viviana e11015<br />

Nehaeva, Tatyana Leonidovna<br />

e13059<br />

Neidhardt, Eve Marie e16036<br />

Neif, Joao Antonio Junior<br />

e12021<br />

Nejjari, Chakib e11502,<br />

e18124<br />

Nekhlyudov, Larissa 6008<br />

Nekljudova, Valentina 556<br />

Nelius, Thomas e15103<br />

Nelkin, Barry 3020,<br />

TPS3117, 5508<br />

Nelli, Fabrizio e18065<br />

Nelson, Aaron L 9562<br />

Nelson, Alan e17529<br />

Nelson, Betty J 8502^<br />

Nelson, Brian e13542<br />

Nelson, Christian J 9123<br />

Nelson, David e21122<br />

Nelson, Deborah 9110<br />

Nelson, Edward L. 6132<br />

Nelson, Garth D. 3514, 3526<br />

Nelson, Greg C e16536<br />

Nelson, Marisa 10097<br />

Nelson, Mark e14574<br />

Nelson, Peter 4520, 4521,<br />

4669, e15207<br />

Nelson, Rebecca A. 1117<br />

Nelson, Robert P 7107, 7108<br />

Nelson, Sarah J. 4660<br />

Nelson, Zenith e17547,<br />

e17550<br />

Nemec, Cheryl TPS4679<br />

Nemunaitis, John J. 2588,<br />

4593, TPS7610, 8594,<br />

e13101, e18047, e19055<br />

Neninger, Elia 2527, 2531<br />

Neogi, Tuhina TPS669<br />

Neoptolemos, John P. 4053,<br />

e14625<br />

Nepple, Kenneth Gerard 4594<br />

Nerenz, David R 1591<br />

Neri, Eric 9051<br />

Neri, Santo e18564<br />

Nerodo, Ekaterina e15519,<br />

e21127<br />

Nerodo, Galina A. e15519,<br />

e21127<br />

Neron do Nascimento, Yeni<br />

7506, e11087<br />

Neskovic-Konstantinovic, Zora<br />

TPS661<br />

Ness, Daniel 6574<br />

Ness, Kirsten K. 6030, 9526,<br />

9527, 9528<br />

Nessim, Carolyn 616<br />

Nestico, Jill e19041<br />

Nestle-Kraemling, Carolin<br />

e11556<br />

Neto, Hugo Sterman 2038<br />

Netto, Cristina 10593,<br />

e21042<br />

Neubauer, Andreas 6510<br />

Neubauer, Hans 5042,<br />

e21003<br />

Neubauer, Marcus A. 6109,<br />

7502<br />

Neubauer, Nikki Lynn 5039<br />

Neuberg, Donna S. 6606,<br />

6618, 6621, 9530<br />

Neubert, Sylvia 5044<br />

Neufeld, Caleb e13045<br />

Neugebauer, Julia Katharina<br />

554, 1600^, 10540<br />

Neuger, Anthony 2501,<br />

TPS2620<br />

Neugut, Alfred I. 4101,<br />

6078, 6118<br />

Neuhaus, Peter 4020<br />

Neuhaus, Ruth 8030<br />

Neumaier, Bernd 7603<br />

Neumann, Avivit 4564, 4619<br />

Neumann, Frank TPS6639,<br />

TPS6641<br />

Neumann, Svenja 6500^<br />

Neumann, Tabea 4128<br />

Neumann, Thomas e17529<br />

Neumeister, Peter e18525<br />

Neumeister, Veronique 573,<br />

1121<br />

Neupane, Prakash e13551<br />

Neuss, Michael N. CRA1505<br />

Neuville, Agnes 10018<br />

Neuwelt, Edward A. 2040,<br />

2077<br />

Neuzillet, Yann e15145<br />

Nevala, Wendy Kay 8599,<br />

e21112<br />

Neven, Patrick 1020<br />

Neves, S<strong>of</strong>ia e18111<br />

Neveux, Nathalie e13056<br />

Neville, Alan John e16558<br />

Neville, Bridget A. 6026<br />

New, Pamela Z. 2098<br />

Newbold, Kate 5518, 5591<br />

Newcomb, Polly A. 3526<br />

Newman, Edward M. 2538,<br />

8073<br />

Newman, Nancy 6572,<br />

e18535<br />

Newman, Suzanne E. 2591,<br />

5050, e13063<br />

Newsom-Davis, Thomas<br />

e14574<br />

Newton, Robert Charles<br />

2500^<br />

Newton, Robert 9558<br />

Ney, Gina 4591<br />

Ney, Jasmin Teresa 1523<br />

Neyns, Bart 2050, 2507,<br />

8517, e19026, e19027<br />

Nezu, Masahiko 585<br />

Ng Tang, Derek 2520^<br />

Ng, Alice Wan-ying 5511<br />

Ng, Chaan S. TPS5114<br />

Ng, Daniel B 4666, e14028,<br />

e14030, e15186<br />

765s


Ng, Daniel e15050<br />

Ng, Dawn Marie e11050<br />

Ng, Elise 8574<br />

Ng, John e15162, e17562<br />

Ng, Kimmie e19586<br />

Ng, Patrick e13555<br />

Ng, Quan Sing 3098, 4100^,<br />

e16024<br />

Ng, Raymond C.H. 9041<br />

Ng, Sing Chau e14691<br />

Ng, Siobhan LBA4518,<br />

TPS4692^, e15112<br />

Ng, Su Ann 5578<br />

Ng, Tat Ming e18546<br />

Ng, Thian C 1064<br />

Ng, Wai Hoe 2066<br />

Ngai, Yenting 7562<br />

Ngamphaiboon, Nuttapong<br />

e19568<br />

Ngan, Roger K.C. 5511<br />

Ngan, Sam 3629<br />

Ngeow, Joanne Y. Y. 1508<br />

Ngo, Charlotte e15524<br />

Ngo, Justine V 6010<br />

Nguyen, Anne T 525<br />

Nguyen, Bichlien 10554<br />

Nguyen, Diana 10603<br />

Nguyen, Diane Duyen Thuy<br />

e21105<br />

Nguyen, Hoa 5012<br />

Nguyen, Hoang 6124<br />

NGuyen, Jean-Michel e19019<br />

Nguyen, Ly Minh 8594<br />

Nguyen, Lyly 1132<br />

Nguyen, Minh-Tri 1569<br />

Nguyen, Nhu-Nhu 6529<br />

Nguyen, Nicole e15154<br />

Nguyen, Paul Linh 4657<br />

Nguyen, Phuong T.B. e13542<br />

Nguyen, Phuoung 9544<br />

Nguyen, Rosa 6136<br />

Nguyen, Susan 2585<br />

Nguyen, Tan Dat 4091<br />

Nguyen, Teresa T. 7061<br />

Nguyen, Tina e15114<br />

Nguyen, Tuan 7554<br />

Nguyen, TX Quyen e15115<br />

Nguyen, Vinh e16026<br />

Nguyen-Tan, Felix 5530<br />

Niazi, Faiz 3003, 8511<br />

Niazi, Tamim e14020<br />

Nibu, Kenichi e16034<br />

Nicacio, Leonardo 4666,<br />

e15186<br />

Niccoli, Patricia 4118<br />

Nicholas, Garth Andrew<br />

e18079<br />

Nicholas, Richard W. 9537<br />

Nicholl, Michael Bruce 2519<br />

Nichols, Christine 9144,<br />

e19633<br />

Nichols, Craig R. 9001<br />

Nichols, Gwen L. e13600^<br />

Nichols, Laura L 3516<br />

Nicholson, James 9539<br />

Nicholson, Steve TPS8605<br />

Nickolich, Myles 7087<br />

Nicol, Andrew John e13128<br />

Nicolai, Nicola 4507, 4629<br />

Nicolaizeau, Cyrielle<br />

TPS10634<br />

Nicolardi, Linda e13019<br />

Nicolas, Patrick e18003,<br />

e18020<br />

Nicolau, Ulisses Ribaldo<br />

e16046, e18078<br />

Nicoletti, Stefania Vittoria,<br />

Luisa 8529, 10615<br />

Nicoletto, Maria Ornella<br />

e12037<br />

Nicolini, Franck E. 6503,<br />

6513, 6596, 6604<br />

Nicolle, Delphine e15161<br />

Nicolson, Marianne 7583<br />

Niculescu, Liviu 4542<br />

Nicum, Shibani 3048, 3051<br />

Nie, Qiang 7039<br />

Nie, Shuming e21161<br />

Nieda, Mie e13128<br />

Niederacher, Dieter 5097,<br />

e21017, e21020<br />

Niedergethmann, Marco 4020<br />

Niederle, Bruno 5508<br />

Niederle, Norbert 3<br />

Niederwieser, Dietger 6626^,<br />

TPS6640, e16016<br />

Niedzwiecki, Donna 2585,<br />

3517<br />

Niegisch, Guenter 4522,<br />

4525, 4590<br />

Nieh, Peter e15006<br />

Niehr, Franziska e16018<br />

Nielander, Aldrik 4622<br />

Nielsen, Boye Schnack 3513<br />

Nielsen, G. Petur 10058<br />

Nielsen, Hans J. 3576<br />

Nielsen, Kirsten Marie<br />

e14517<br />

Nielsen, Patricia Switten<br />

e21110<br />

Nielsen, Torsten O. 1008<br />

Nielsen, Ulrik TPS663<br />

Niemierko, Andrzej 1122<br />

Nierodzik, Mary Lynn R.<br />

e16052<br />

Niessner, Heike 8526<br />

Niesvizky, Ruben 8033,<br />

8034, 8036, e18569<br />

Niethammer, Andreas G.<br />

7551<br />

Nieto, Antonio 10013<br />

Nieto, Yago 4533, 6540,<br />

8102<br />

Nieuwland, Marja 3065<br />

Nieva, Jorge J. 10574,<br />

e21074<br />

Nigam, Rita 2588<br />

Niggli, Felix 9573<br />

Niho, Seiji 1552, 7012,<br />

7044, 7070, e17513,<br />

e17526, e17554, e18054,<br />

e18064<br />

Niibe, Yuzuru e15533<br />

Niikura, Naoki 565<br />

Niitsu, Nozomi 8023<br />

Nijem, Ihsan 2567<br />

Nijs, Griet 2506, e13051<br />

Nikipelova, Elena A e21101<br />

Nikitin, Kirill D. 9060<br />

Nikkhah, Guido 2000<br />

Niknam, Bijan 6000<br />

Nikrad, Malti e21142<br />

Nikulin, Maxim P. e20501<br />

Nilakantan, Vani e19030<br />

Niland, Joyce C. 599, 1125,<br />

1562, e18018, e18530<br />

Nilsson, Monique B. 7096<br />

Nilsson, Sten LBA4512,<br />

4551, 4561<br />

Nimeiri, Halla Sayed 3605,<br />

4102<br />

Nimmannit, Akarin e14051<br />

Nin, Mikio e15065, e15069<br />

Ning, Yan 3540, 3549, 3584,<br />

3597, 3604, 3615, 3622,<br />

4026, 4088, 4096,<br />

e11018, e14031, e14034,<br />

e14052<br />

Ning, Yangmin M. 4569<br />

Ning, Yi 6611<br />

Ninomiya, Jun 613<br />

Ninot, Gregory 9074<br />

Nippgen, Johannes TPS10099<br />

Nir, Raphael 1055<br />

Niraula, Saroj 1019<br />

Nirodi, Chaitanya 10624<br />

Nishi, Hirotaka 10557<br />

Nishida, Hayato e15057,<br />

e15088<br />

Nishida, Hideji 10075<br />

Nishida, Toshirou LBA10008<br />

Nishigaki, Yutaka e18135<br />

Nishihira, Ryuichi e19583<br />

Nishikawa, Ryo TPS2104<br />

Nishikawa, Shingo e17540<br />

Nishimura, Hajime e13037<br />

Nishimura, Hideki e16034<br />

Nishimura, Jennifer 1031<br />

Nishimura, Kazuhiko e14004<br />

Nishimura, Kazuo e15048,<br />

e15065, e15069<br />

Nishimura, Kenji e15065<br />

Nishimura, Masaharu 7571,<br />

e18135<br />

Nishimura, Meiko e16034<br />

Nishimura, Reiki 588<br />

Nishimura, Ryuichiro 5103<br />

Nishimura, Sadako 5006<br />

Nishimura, Shintaro e21059<br />

Nishimura, Takashi e18134<br />

Nishina, Tomohiro 4002,<br />

4046, 4082, e14038,<br />

e14123, e14510<br />

Nishino, Kazumi e18007,<br />

e18025<br />

Nishino, Mizuki 7525, 7553<br />

Nishio, Kazuto TPS659,<br />

10569<br />

Nishio, Makoto 7003, 7028,<br />

7515, 7602, 10569,<br />

10604, e18004, e18014,<br />

e18017<br />

Nishio, Shin 5085<br />

Nishioka, Yuki 3079<br />

Nishiwaki, Morie 5084,<br />

e18064<br />

Nishiwaki, Satoshi 6533<br />

Nishiyama, Akihiro 7528,<br />

e18025, e18134<br />

Nishiyama, Hiroyuki e15004<br />

Nishizawa, Yusuke 3569<br />

Nissim, Ouzi 2078<br />

Nistér, Monica 4561<br />

Nitiruangjaras, Anupong<br />

e14639<br />

Nitsche, Ulrich 3510<br />

Nitta, Hidetoshi e21084<br />

Nitz, Ulrike 552<br />

Niu, Jiangong 600<br />

Niv, Yaron e14067<br />

Niwa, Yasumasa e14540<br />

Niwakawa, Masashi e15098<br />

Niwinska, Anna e11553<br />

Nixon, Andrew B. e13589,<br />

e21038<br />

Niyikiza, Clet TPS663<br />

Njiaju, Uchenna O e11554<br />

Njoh, Roland Fokwen e17011<br />

Njuguna, Festus 2601<br />

Noberasco, Cristina 2595,<br />

e13606<br />

Nobile, Francesco e18564<br />

Nock, Charles John TPS1616<br />

Noda, Akihiro e21059<br />

Noda, Hidehiro e13029<br />

Noe, Jeanne TPS656<br />

Noeding, Stefanie 554<br />

N<strong>of</strong>ech-Mozes, Sharon 1123<br />

Noga, Stephen J. e18558<br />

Nogami, Naoyuki 7042,<br />

7576, e18099<br />

Nogova, Lucia 1533, 3044,<br />

TPS3115, 7603<br />

Noguchi, Atsushi 1103<br />

Noguchi, Kohei 3022<br />

Noguchi, Shinzaburo 512,<br />

539, 540, 551, 559<br />

Noh, Dong Young 1133,<br />

e21160<br />

Noh, Dong-Young 1056,<br />

10621, e11532<br />

Noh, Sung Hoon TPS4142,<br />

e14504, e14652<br />

Noh, Yook-Hwan e13092<br />

Noize, Pernelle e14168<br />

Nokisalmi, Petri e13035<br />

Nolan, C. 1543<br />

Nolan, Carmel e12038<br />

Nolan, Craig 2008<br />

Nolan, Vikki G 9526<br />

Nole, Franco 546, 1049,<br />

1115, 5513<br />

Nolop, Keith B. 8502^<br />

Nomori, Hiroaki 7038<br />

Nomura, Fumio 7046<br />

Nomura, Hironori e15065,<br />

e15069<br />

Nomura, Motoo e14500<br />

Nomura, Shosaku e18081<br />

Nonnenmacher, Anika 5572<br />

Nonomura, Norio 4632,<br />

e15065, e15069, e15098<br />

Nooka, Ajay K. 8086, 8100,<br />

8101<br />

Noonan, Anne M. 2545<br />

Noonan, Krista e11079<br />

Noonan, Sinead e21024<br />

Noone, Anne Michelle 6006<br />

Norden, Andrew David 2009<br />

Nordgren, Brian K. 8011<br />

Nordle, Orjan 4550<br />

Nordlinger, Bernard 3508<br />

Nordstrom, Beth L e13081<br />

Noriega-Iriondo, Maria<br />

Fernanda e15525<br />

Normanno, Nicola e14042,<br />

e14113, e18021<br />

Normolle, Daniel Paul 7071,<br />

8577<br />

Noronha, Vanita 5585,<br />

e13068, e16028, e17500<br />

Norris, LeAnn B. 9143<br />

Norris, Robin Elizabeth 9540<br />

Norris, Sarah 7604<br />

Norström, Carina e13518<br />

North, Marie- Odile e21014<br />

North, Richard T. e15054<br />

North, Scott A. 4536, 4538,<br />

4558, e16558<br />

North, Scott TPS4683<br />

Northfelt, Donald W.<br />

TPS2618, 3060, 6025,<br />

7073, e13052, e13086,<br />

e13501<br />

Northover, John 4004, 4029<br />

Northrup, Robert e19577<br />

Nortier, J. W. R. 526, 1080,<br />

10518, e16526<br />

Nortilli, Rolando 630<br />

Norton, Larry TPS1143<br />

Norwood, Thomas 6502<br />

766s Numerals refer to abstract number


Nos, Claude 5098, e15558<br />

Nosaki, Kaname 3045<br />

Nosov, Aleander TPS4696^<br />

Nosov, Dmitry 4501, e15082<br />

Nosova, Kristin 2080<br />

Noto, Richard e13592<br />

Nott, Louise M. 3504,<br />

TPS4137<br />

Nottage, Kerri 9526, 9527<br />

Nottage, Michelle K. 8538<br />

Nourbakhsh, Ali e18560<br />

Novaes, Paulo Eduardo<br />

e15509<br />

Novarino, Anna 4017<br />

Novello, Silvia 7085,<br />

TPS7109, 7550, TPS7619,<br />

e18103<br />

Novetsky, Akiva Pesach 5013<br />

Novik, Alexey Viktorovich<br />

e13059<br />

Novik, Yelena e11529<br />

Novopashenny, Igor 1078<br />

Novotny, William F. 4560<br />

Nowacki, Amy S 2014<br />

Nowaczyk, Piotr 10537<br />

Nowadly, Cynthia e17535<br />

Nowak, Anna K. 7079^,<br />

9087<br />

Nowakowski, Grzegorz S.<br />

8053, 9057<br />

Nowara, Elzbieta 2522,<br />

3530, e14159<br />

Nowshad, Gul TPS6635<br />

Nowsheen, Somaira 621<br />

Noy, Ariela 6609, 8054,<br />

9004<br />

Noyes, David 2501, 2559<br />

Noyes, Katia 9091<br />

Noyes, R Dirk TPS657<br />

Nozawa, Hiroaki e14142<br />

Ntabaye, Moshi 6102<br />

Nteliopoulos, Georgios 10550<br />

Nuara, Michael TPS5602<br />

Nuber, Ulrike TPS4139<br />

Nucci, Marisa 5090<br />

Nuevo, Gema e17515<br />

Nugent, William 7022<br />

Nuguyen, Tuan Stevon 7583<br />

Nuijen, Bastiaan 2550<br />

Nuin, Paulo S 10622<br />

Nukiwa, Toshihiro 7086,<br />

7563, 7565, e18129<br />

Nukui, Tomoko 8528<br />

Numico, Gianmauro e19592<br />

Nunes, Joao Soares e14599<br />

Nuñez, Isaac TPS4679,<br />

e21021<br />

Núñez, M. Isabel 1041<br />

Nunez-Valencia, Carolina<br />

e19618<br />

Nuno, Miriam 2080, 2084,<br />

2087<br />

Nurkin, Steven J. e14712<br />

Nutt, Stephanie 9136,<br />

e19503<br />

Nutting, Charles W. 3590<br />

Nutting, Christopher e16007<br />

Nuver, Janine 4528<br />

Nuzhad, Afrin 9086<br />

Nuzzo, Carmen e14661<br />

Nuzzo, Gennaro 4110<br />

Nwankwo, Kenneth Chima<br />

e19572<br />

Nwe, Khin Khin e14055<br />

Nwogu, Chukwumere E.<br />

e17535<br />

Nyalendo, Carine 9563, 9570<br />

Nycum, Lawrence R. 5054<br />

Nyman, Jan LBA7000<br />

Numerals refer to abstract number<br />

Nystrom, J. Scott 10579,<br />

10584<br />

O<br />

O Suilleabhain, Criostoir<br />

e14666<br />

O’ Keefe, Christine 6521<br />

O’ Sullivan, Michael GJ<br />

e11569<br />

O’Brien, Donna M. 6086<br />

O’Brien, James P. 5518,<br />

5591, 8594, e19055<br />

O’Brien, Joan 9515<br />

O’Brien, Mary 7081<br />

O’Brien, Maureen Megan<br />

e20007<br />

O’Brien, Neil A. 617, 633<br />

O’Brien, Robert 4577, 4582,<br />

4585<br />

O’Brien, Susan Mary 6501,<br />

6507, 6515^, 6516^,<br />

6517, 6528, 6578, 6587,<br />

6598, 6607, 6619^, 8032<br />

O’Brien, Thomas 7072^,<br />

e17531^<br />

O’Bryan-Tear, C. Gillies<br />

LBA4512, 4551, TPS4694<br />

O’Bryant, Cindy L. e16541,<br />

e16550<br />

O’Byrne, Kenneth John 637,<br />

LBA7500, TPS7615,<br />

TPS7616, e19059<br />

O’Callaghan, Christopher J.<br />

3504, 3515<br />

O’Callaghan, Christopher J<br />

TPS2104, 3572<br />

O’Cathail, Sean Micheal<br />

2608, e13033<br />

O’Cearbhaill, Roisin Eilish<br />

5041<br />

O’Connell, Joseph P.<br />

TPS7615, e18093<br />

O’Connell, Michael J.<br />

TPS1615<br />

O’Connell, Michael 3512,<br />

3522, 3523, 3525<br />

O’Connor, Ashley TPS3117<br />

O’Connor, Juan Manuel<br />

e14598<br />

O’Connor, Kate e14687<br />

O’Connor, Miriam 581<br />

O’Connor, Owen TPS8110,<br />

e13569<br />

O’Connor, Robert 2536<br />

O’Day, Steven e13088<br />

O’Dea, Pauline 2062<br />

O’Donnell, Anne e15054<br />

O’Donnell, Colin e15176<br />

O’Donnell, Margaret R. 1592<br />

O’Donnell, Patrick 10596<br />

O’Donnell, Peter H. 4522,<br />

e13120^<br />

O’Donnell, Robert e13573<br />

O’Donoghue, Diarmuid<br />

e13570, e21024<br />

O’Donovan, Norma 617, 622,<br />

632, 633, 637, e13520<br />

O’Dorisio, Thomas M. 4085,<br />

e14600, e14620, e15580<br />

O’Driscoll, Lorraine 617, 633<br />

O’Dwyer, Peter J. 4111,<br />

TPS4134, 4596, e13602,<br />

e13604<br />

O’Dwyer, Peter J 2500^<br />

O’Grady, Elizabeth 3613<br />

O’Keefe, Graeme Joseph<br />

e15056<br />

O’Keefe, Margaret e16506<br />

O’Keefe, Sylvia e12038<br />

O’Keeffe, Margaret e14666<br />

O’Mahar, Shannon Eileen<br />

6570<br />

O’Malley, A. James 6045<br />

O’Malley, David M. e15585<br />

O’Malley, Meaghan Working<br />

2602<br />

O’Malley, Rebecca Leigh<br />

e15046<br />

O’Malley, Rebecca e15039<br />

O’Mara, Ann M. 9047, 9099,<br />

9112, 9144, e19633<br />

O’Neil, Bert H. 3532, 4052,<br />

e13109<br />

O’Neill, Anne M. 605, 6025<br />

O’Neill, Anne 1021, 5501<br />

O’Regan, Kevin N. 8552<br />

O’Reilly, Adrienne M. 8081<br />

O’Reilly, Derek e21037<br />

O’Reilly, Eileen Mary 4057,<br />

9105<br />

O’Reilly, Kathryn E. 8544<br />

O’Reilly, Seamus 1559,<br />

2062, e11569, e14666,<br />

e16506<br />

O’Rourke, Donald TPS2107<br />

O’Rourke, Pat 3078<br />

O’Shannessy, Daniel 3016,<br />

e21028<br />

O’Shaughnessy, Joyce 547,<br />

596, 604, TPS666, 1016,<br />

1017, 1018, TPS1140^<br />

O’Shaughnessy, Matthew<br />

e15211<br />

O’Shea, Alan e16506<br />

O’Shea, Tanya e13520<br />

O’Sullivan, Brendan e13507,<br />

e13554<br />

O’Sullivan, Brian 5571,<br />

TPS5600, 10050<br />

O’Sullivan, Conor 4526<br />

O’Sullivan, Jacintha e13570,<br />

e21024<br />

O’Sullivan, Joe M. LBA4512,<br />

4551<br />

O’Toole, Dermot 4036<br />

O’Toole, Sandra 504<br />

Oades, Kahuku e14164<br />

Oaknin, Ana 5001,<br />

LBA5002^, 5017^, 5038,<br />

5068, e15575<br />

Oates, Jacqueline Rose<br />

e14088<br />

Oba, Hanako 613<br />

Oba, Koji TPS3642<br />

Obad, Susanna e13567<br />

Obajimi, Millicent e11554<br />

Obara, Wataru e15068<br />

Obasaju, Coleman K. LBA4,<br />

7002, e18138<br />

Obasaju, Coleman 5533<br />

Obayashi, Tomohiko e14540<br />

Obel, Jennifer Carrie 4049<br />

Oberg, Ann L. e15132<br />

Oberg, Kjell E. 4122<br />

Oberholtzer, Carl TPS7609<br />

Oberlin, Odile 9510,<br />

TPS9597^<br />

Obermiller, Angela Mae 9110<br />

Oberndorfer, Stefan 2071<br />

Oberthur Johnson, Laura<br />

e17017<br />

Obertin, Stephanie e14180<br />

Obertova, Jana TPS4677,<br />

e15028<br />

Obi, Shuntaro e14622<br />

Obi-Tabot, Eliot e15193,<br />

e15198<br />

Oborski, Matthew J 3019<br />

Oboyle, Erin e19635<br />

Obrador, Antonia 10079<br />

Ocak Arikan, Ozlem e15047<br />

Ocana, Alberto 558, 6043<br />

Ocean, Allyson J. 4043<br />

Ochi, Takanori e14004<br />

Ochiai, Atsushi 1552, 4012<br />

Ochiai, Hiroki 10531<br />

Ochiai, Kazunori 5003<br />

Ochiai, Takumi e14004<br />

Ochiai, Toshiya e14518<br />

Ochoa de Olza, Maria 3096<br />

Ochoa, Roberto Enrique<br />

1071, e18109<br />

Ochsenreither, Sebastian<br />

2523<br />

Ockene, Judith 1501<br />

Ocvirk, Janja e14083<br />

Oczko-Wojciechowska,<br />

Malgorzata e14159,<br />

e21049<br />

Oda, Hiroyasu e19546<br />

Oda, Katsutoshi 5037<br />

Oda, Tsuneyuki e19583<br />

Odani, Akira 10075<br />

Oddoux, Carole e15191<br />

Odenike, Olatoyosi 6562,<br />

6582<br />

Odintsova, Anastasiya<br />

Sergeevna e15543<br />

Odunsi, Kunle 2510, 2588,<br />

e11563<br />

Oechsle, Karin 4597<br />

Oeffinger, Kevin C. 6001,<br />

6030, 6073, CRA9513,<br />

9514, 9552, 9586<br />

Oei, Paul e15050<br />

Oelmann, Elisabeth 3004<br />

Oelschlager, Kristen M. 7106,<br />

8543<br />

Oeltjen, Amy R 4062<br />

Oermann, Eric Karl 2091<br />

Oesterreich, Steffi 525<br />

Oestreicher, Judith e13511<br />

Oettle, Helmut 4020, 4034,<br />

4044<br />

Offit, Kenneth e15022<br />

Offutt, Julie e19030<br />

Ogan, Ken e15006<br />

Ogata, Ryo e19583<br />

Ogata, Yoshitaka e18007,<br />

e18141<br />

Ogata, Yutaka 3558, e14077<br />

Ogawa, Carolina<br />

MATSUBAYASHI e21151<br />

Ogawa, Masao e14518<br />

Ogawa, Osamu 10520,<br />

e15048<br />

Ogawa, Takenori e16020<br />

Ogawara, Daiki 7569<br />

Ogbagabriel, Selam 3580<br />

Ogbata, Obiageli Uchenna<br />

7025<br />

Ogburn, Emma TPS660<br />

Ogden, Angela 3033<br />

Ogdie, Alexis R 6579<br />

Oger, Emmanuel e19037<br />

Ogino, Atsuko e18099<br />

Ognjanovic, Miodrag 1576<br />

Ognjanovic, Simona 1576<br />

Oguchi, Masahiko 8050<br />

Ogundiran, Temidayo O<br />

e11554<br />

Ogura, Kaoru 9577<br />

Ogura, Mariko 10541<br />

Ogura, Michinori 8023, 8029,<br />

TPS8118<br />

Ogura, Takashi e19583<br />

Ogura, Takeshi e14540<br />

767s


Oguri, Senri 5556<br />

Oh, Do Youn 1003<br />

Oh, Do-Youn LBA1, 3076,<br />

3624, e14136, e14617<br />

Oh, Jeong Hoon 8536,<br />

e15181<br />

Oh, Ji-Eun e18050<br />

Oh, Nahmgun e14121,<br />

e14181<br />

Oh, Shiaki 10548<br />

Oh, Sung Tae 10093<br />

Oh, Sung Yong 4064, 8029,<br />

e14686<br />

Oh, William K. 4516, 4524,<br />

4623, 4656, 6065,<br />

e21016, e21032<br />

Ohanian, Maro 6587<br />

Ohashi, Kadoaki 7532<br />

Ohashi, Kazuteru 6533<br />

Ohashi, Yasuo 4035, e19556<br />

Ohashi, Yoshiaki e14635<br />

Ohdan, Hideki e14091<br />

Ohe, Yuichiro 1552, 6112,<br />

7003, 7017, 7044,<br />

TPS7612, e17513,<br />

e17526, e17554, e18054,<br />

e18064<br />

Ohga, Takefumi e14033,<br />

e14601<br />

Ohgino, Keiko e18058<br />

Ohi, Masaki e14541<br />

Ohinata, Aki 4060<br />

Ohkawa, Shinichi 4035, 4042<br />

Ohki, Masahiro 3079<br />

Ohlfest, John 2546<br />

Ohlmann, Carsten Henning<br />

4590, e15195<br />

Ohmachi, Ken 8023<br />

Ohmann, Christian 4590,<br />

e16033<br />

Ohmatsu, Hironobu 1552,<br />

7044, 7515, e17513,<br />

e17526, e17554, e18054,<br />

e18064<br />

Ohmori, Tohru e13029<br />

Ohnmacht, Ute 3005<br />

Ohno, Shinji 588<br />

Ohrling, Katarina 4015<br />

Ohsugi, Jun e13015<br />

Ohsumi, Shozo 540<br />

Ohta, Hideaki 9574<br />

Ohta, Naohiro 3607<br />

Ohtsu, Atsushi 3079, 4067,<br />

4081, TPS4139<br />

Ohtsu, Hajime e14601<br />

Ohuchi, Kohta e19611<br />

Ohue, Masayuki 3524, 3607<br />

Ohyanagi, Fumiyoshi 10569,<br />

10604, e18004, e18014,<br />

e18017<br />

Oikawa, Masaya e14011<br />

Oikonomopoulos, Georgios<br />

e11073, e11519<br />

Oing, Christoph 4597<br />

Oishi, Takayuki e19611<br />

Oizumi, Satoshi 7086, 7571,<br />

e18129, e18135<br />

Ojeda, Belen 1120, 10555<br />

Ojeda, Juan 6599<br />

Ojeda, Sandra 8537<br />

Ojong-Ntui, Martin e11084<br />

Oka, Daizo e15065<br />

Oka, Ryota 5584<br />

Oka, Yoko 9053<br />

Okabayashi, Koji 10531<br />

Okabe, Kazunori e17517<br />

Okabe, Takafumi e18060<br />

Okada, Daisuke 10542<br />

Okada, Hideaki e18036,<br />

e18042<br />

Okada, Morihito 7031<br />

Okada, Toshiaki 7560<br />

Okada, Yoshinari e19611<br />

Okafuji, Kouhei e18141<br />

Okajima, Masazumi e14091<br />

Okamoto, Hiroaki 6112,<br />

7012, 7017, 7028, 7515,<br />

e18099<br />

Okamoto, Isamu 4002,<br />

TPS7110, 7521, 7590,<br />

7592, TPS7618, e18060<br />

Okamoto, Norio e18036,<br />

e18042<br />

Okamoto, Tatsuro e13090<br />

Okamura, Atsuo e17020<br />

Okamura, Haruki e21004<br />

Okamura, Takuho 565<br />

OKane, Patricia 9018<br />

Okano, Shinji e13014<br />

Okazaki, Keito e19521<br />

Okazaki, Toshihiko e13014<br />

Okhovat, Jean-Phillip e18549<br />

Oki, Eiji 3558, e14033,<br />

e14601, e14722<br />

Oki, Yasuhiro 8070<br />

Okigami, Masato e21053<br />

Okinaga, Shoji 7563<br />

Okines, Alicia Frances Clare<br />

LBA4000<br />

Okita, Hajime 9574<br />

Okoshi, Yasushi 8094<br />

Oksanen, Minna e13035<br />

Oksuzoglu, Omur Berna 1596,<br />

e11540<br />

oktay Tanyildiz, Gulsah<br />

e20000<br />

Oktay, Kutluk 1097<br />

Okten, Ilker 1596<br />

Okubo, Satoshi e14004<br />

Okuda, Kentaro e13029<br />

Okuda, Ryo e19583<br />

Okudera, Koichi 7086, 7561<br />

Okugawa, Yoshinaga e14029,<br />

e14039, e14541, e21053<br />

Okumura, Yoshitomo 7080,<br />

10585<br />

Okunnu, Marita 3015<br />

Okuno, Keiko e18060<br />

Okuno, Scott H. 10003,<br />

TPS10099<br />

Okur, Yillar e16005, e19575<br />

Okusaka, Takuji 4031, 4035,<br />

4042, TPS4135, TPS4146<br />

Okutur, Kerem e14698<br />

Okuyama, Takako e18025<br />

Olatoye, Dare 4559<br />

Olaussen, Ken 10556<br />

Olaverri Hernandez, A.<br />

e19590<br />

Olbara, Gilbert 2601<br />

Olcese, Cristina 575<br />

Old, Lloyd J. e17558<br />

Oldenburg, Jan 4508, 4637<br />

Oldenhuis, Corina N. 4132<br />

Olek, Sven 5053, 5070<br />

Olijade, Oludamilola e13109<br />

Olin, Michael 2546<br />

Oliner, Kelly S. 2535, 2594,<br />

4005, 5502, 5504^<br />

Olino, Kelly e13559<br />

Oliva, Claudia 2531<br />

Oliva, Cristiano 10055<br />

Oliva, Stefano 645<br />

Olivari Tilola, Silvia 9054,<br />

e19530<br />

Olive, Daniel e15155<br />

Oliveira, Celia Tosello 1092<br />

Oliveira, Harley F. e12512<br />

Oliveira, Harley Francisco<br />

e19628<br />

Oliveira, Ivanir Martins<br />

e14027<br />

Oliveira, Joao e13119<br />

Oliveira, Mafalda 509, 566,<br />

619, TPS652, 10511<br />

Oliveira, Mario e14673<br />

Oliveira, Thiago Bueno<br />

e16046<br />

Oliveria, Susan A. 1590,<br />

1591<br />

Olivier, Kenneth R e14507<br />

Olivo, Martin Sebastian 7093<br />

Olivotto, Ivo A. LBA1031<br />

Ollari, Maria Grazia 9120<br />

Olmedo Garcia, Maria Eugenia<br />

e16029<br />

Olmos, David 1545, 2540,<br />

4553, 5022, 10039,<br />

10086, e15136<br />

Olocco, Ricardo e21025<br />

Olopade, Olufunmilayo I. 534<br />

Olopade, Olufunmilayo I<br />

e11554<br />

Olopade, Olusola C e11554<br />

Olowokure, Olugbenga<br />

Olanrele TPS3116, e14714<br />

Olpin, Jeffrey 3057<br />

Olsen, Brian e21133<br />

Olsen, Ingrid Holst 4015<br />

Olsen, Jon 9110<br />

Olsen, Mark R. LBA4<br />

Olsen, Steven R. LBA1<br />

Olshinka, Liran e14571<br />

Olson, Erin Macrae 609,<br />

6089<br />

Olson, Garth T 5549,<br />

TPS5602<br />

Olson, Jeffrey J. 3055<br />

Olson, Jillian 6108<br />

Olson, John A. 503<br />

Olson, Karin Lou 9131<br />

Olson, Kristin 3566<br />

Olson, Lawrence Karl 2056<br />

Olson, Thomas Arthur 9539<br />

Olson, Tim e16540<br />

Olson, Walter C. e19028<br />

Olson, William C. e21131<br />

Olsson, Hakan Lars 1594<br />

Olszanski, Anthony J. 2554,<br />

8530<br />

Olszewska, Malgorzata<br />

TPS2619, TPS4700<br />

Olszewski, Adam J. 1034,<br />

8044, 8045<br />

Olszewski, Wlodzimierz<br />

Tadeusz 8548<br />

Olungu, Christine 10096<br />

Olusanya, Abimbola O<br />

e19013<br />

Oluwasola, Abideen Olayiwola<br />

e11554<br />

Olver, Ian N. e19553<br />

Olvera-Caraza, Daniela 1607<br />

Omar, Haniza 4106<br />

Omarini, Claudia 631<br />

Omata, Masao e14622<br />

Omene, Coral Oghenerukevwe<br />

e11529<br />

Omenge, Elkanah 5107<br />

Omholt, Katarina 8588<br />

Omlin, Aurelius Gabriel 3022,<br />

4553, e13601, e15134,<br />

e15136<br />

Omollo, Raymond e20504<br />

Omoniyi-Esan, Ganiyat<br />

Oluwatoyin e12022<br />

Omuro, Antonio Marcilio<br />

Padula 2006, 2008, 2028,<br />

2089<br />

Onar-Thomas, Arzu 9518<br />

Onate-Ocana, Luis F. e19618<br />

Oncel, Mufide e11008<br />

Ondovik, Michael S. 8012<br />

Ondrey, Frank G. 5500<br />

Oneko, Olola 6102<br />

Ong, Anna 4048<br />

Ong, Eugene 1064<br />

Ong, Michael 3080, 10525,<br />

e13599<br />

Ong, Teng Jin e18100<br />

Ong, Whee Sze e18515<br />

Ongarello, Stefano e13594<br />

Onitilo, Adedayo A. 555, 563,<br />

6063, 9061, e14160<br />

Ono, Akira 7088, e21007<br />

Ono, Hidetaka e14624<br />

Ono, Mayumi 7540<br />

Ono, Rei e17020<br />

Ono, Sachiyo e21007<br />

Ono, Yoko 6567<br />

Onoe, Takuma 5045<br />

On<strong>of</strong>ri, Azzurra e21070<br />

Onozawa, Yusuke 5542<br />

Onsrud, Mathias 5008<br />

Onukwugha, Ebere e14035,<br />

e14056, e15108, e15146,<br />

e15148<br />

Onur, Handan 1572<br />

Ooi, Boon Phoe 4106<br />

Ooi, Edie Jian Jiek 4106<br />

Oommen, Nikhil Babu 4553<br />

Oostendorp, Linda JM 9036<br />

Oosterhuis, J W 7009<br />

Oosting, Sjoukje 4528,<br />

10581<br />

Ooyama, Takuma 5086<br />

Opdam, Mark 562<br />

Opocher, Enrico 4110<br />

Oppenheim, David 5522<br />

Oppido, Piero Andrea 2085<br />

Oppong, Bridget A. 575<br />

Oprea, Corina 3080<br />

Oprea, Tudor I. TPS5602<br />

Or, Reuven e13013<br />

Orange, Clare e14110<br />

Orange, Clélia 2605<br />

Orbach, Daniel 9536<br />

Orbay, Ozge e11507<br />

Orcun, Seza e18555<br />

Ord, Celine B e14555<br />

Ordentlich, Peter 567<br />

Orel, Nadezhda Fedorovna<br />

e19005<br />

Orera Clemente, Maria<br />

e12003<br />

Orfeuvre, Hubert e11019<br />

Orgiano, Laura 9006<br />

Orita, Hajime e14139<br />

Orlandi, Armando e14156,<br />

e16553<br />

Orlandi, Ester 5555<br />

Orlandi, Massimo 10611<br />

Orlandini, Cinzia 629<br />

Orlando, Laura e11001,<br />

e11547<br />

Orlov, Sergey V 7029,<br />

LBA7500, 7551<br />

Orlova, Kristina<br />

Vyacheslavovna e19005<br />

Orlowski, Robert Z. 8018^,<br />

8035, 8040, 8093,<br />

TPS8112, TPS8113<br />

Ormanns, Steffen 4041<br />

Ormerod, Anne e16557<br />

Ormianer, Mati 1564<br />

768s Numerals refer to abstract number


Oro, Anthony E 8579,<br />

e19048^<br />

Oronzi, Irma 5093, 5094,<br />

e15550<br />

Orr, Maria 1548<br />

Orsi, Franco e14607<br />

Orsi, Jennifer M. 6120,<br />

e16531<br />

Orsini, Christine TPS667<br />

Orsini, James M. 7590<br />

Orsini, James 7592,<br />

TPS7618<br />

Orsini, Lucinda S. 4033<br />

Ortega Ruiperez, Carolina<br />

e19590<br />

Ortega, Alette e18028<br />

Ortega, Cinzia 4627, e15073,<br />

e15133<br />

Ortega, Luis e20513<br />

Ortega, Vanesa 566, 619<br />

Ortega, Vanessa 509<br />

Ortego, Javier 3553<br />

Ortiz Duran, Rosa Maria<br />

4089, e14120<br />

Ortiz, Marcos e14521<br />

Ortiz-Romero, Pablo L. 8076<br />

Ortmann, Uta 554, TPS1146,<br />

1600^, 1602<br />

Ortner, Petra Angelika<br />

e19540<br />

Ortuño-Pacheco, Guzman<br />

e11024<br />

Oruezabal, Mauro e11074<br />

Orui, Hayato 7080<br />

Oruzio, Daniel V. 3588<br />

Osada, Takuya 2585, 10563<br />

Osann, Kathryn 6132<br />

Osarogiagbon, Raymond U<br />

7025, 7069, 7072^,<br />

e17531^<br />

Osborne, C. Kent 606<br />

Osborne, Cynthia R. C. 1017,<br />

e19609<br />

Osborne, Karen 551<br />

Osborne, Raymond 5110<br />

Osborne, Richard 5046<br />

Osei-Boateng, Kwabena<br />

e14611<br />

Oshima, Takashi 4070,<br />

e14624<br />

Oshimi, Kazuo 8050<br />

Oshiro, Takashi TPS3642<br />

Oshita, Fumihiro 7012<br />

Oshitanai, Risa 565<br />

Oskay-Özcelik, Gülten 5044<br />

Osman, Iman 8544, 8550,<br />

8557, 8565, 8571, 8589,<br />

e19003, e19018<br />

Osoba, David 6002<br />

Osorio, Cynthia ABT 1522<br />

Osowski, Susanne 7001<br />

Ospovat, Inna 10034<br />

Ostap<strong>of</strong>f, Katherine T e13563<br />

Oster, Gerry e12024,<br />

e15197, e16573, e18107,<br />

e19630<br />

Oster, Jean-Philippe e16560,<br />

e18089<br />

Osterlund, Pia J. CRA3503,<br />

4015<br />

Ostr<strong>of</strong>f, Rachel e21012,<br />

e21142<br />

Ostrovnaya, Irina 4581^,<br />

4592, e15022<br />

Ostrowska, Beata e18512<br />

Osunkoya, Adeboya O.<br />

e15006<br />

Osuorji, Ikenna e14163<br />

Numerals refer to abstract number<br />

Osvaldt, Alessandro Bersch<br />

e13546<br />

Oswald, Michaela 1046<br />

Ota, David M. 503<br />

Ota, Ikuyo 8094<br />

Ota, Mitsuyoshi e14047,<br />

e14050<br />

Oteiza, Katharine 4045<br />

Otero, Jorge M. e12039<br />

Otero, Silvina e15521<br />

Othieno-Abinya, Nicolas<br />

Anthony e20504<br />

Othman, Fauzi e19632<br />

Othman, Youmna 9520<br />

Othus, Megan 6502, 8525,<br />

8527<br />

Otley, Clark 8590<br />

Oto, Yuji e13029<br />

Otsuka, Hiroko e13046,<br />

e13050<br />

Otsuka, Kojiro 10610,<br />

e18025, e18134<br />

Otsuka, Kyoko 7528, 10610<br />

Otsuka, Shannon e18110<br />

Ott, Marion Gabriele 7544<br />

Ott, Patrick Alexander 2589<br />

Ottaiano, Alessandro e14130<br />

Ottaviani, Davide 4627<br />

Ottensmeier, Christian H.H<br />

7081, 8566<br />

Ottensmeier, Christian 2583,<br />

TPS8605<br />

Otterson, Gregory Alan 2610,<br />

7513, 7546, 7600,<br />

TPS7619, e18018<br />

Ottesen, Lone Harild 4108<br />

Ottesen, Rebecca A 599,<br />

1125, 1562<br />

Ottevanger, P. B. 5061<br />

Ottevanger, Petronella B 9036<br />

Otto, Ge<strong>of</strong>f 3533, 7510<br />

Ou, Sai-Hong Ignatius 2610,<br />

7000, 7508, 7530, 7533,<br />

7557, 7596, 7599, 7600,<br />

e18126, e18128, e18140<br />

Ou, Sai-Hong 7083<br />

Ou, Wei 7034<br />

Ou, Yen-Chuan e15047,<br />

e15064<br />

Ouafik, L’houcine 8555<br />

Ouali, Monia 10009, 10067,<br />

10096<br />

Ouatas, Taoufik TPS4698^<br />

Oubel, Estanislao e13547<br />

Oudard, Stephane 1095,<br />

4510, LBA4567^, 4583,<br />

4642, TPS4696^, 5067,<br />

e15040, e15161, e19640<br />

Oude Munnink, Thijs H.<br />

10581<br />

Ouellet, Danielle LBA8500^<br />

Ouellette Freve,<br />

Johann-Francois e19591<br />

Ould Kaci, Mahmoud TPS647<br />

Oulla, Karima e11516<br />

Oura, Shoji 1119<br />

Ouyang, Xuenong 9017,<br />

e18033<br />

Ovalle, Valentina e14002<br />

Ovechkina, Yulia e13542<br />

Overbury, Rebecca S. e21077<br />

Overkamp, Friedrich<br />

TPS3641^, e15044<br />

Overman, Michael J. 3544,<br />

3556, 3561, 3598, 4037,<br />

4054, 4116, 9072<br />

Owen, Kate 6527<br />

Owen, Roger G. 8015<br />

Owens, Brian L e14160<br />

Owens, Pamela e18551<br />

Owonikoko, Ta<strong>of</strong>eek Kunle<br />

2576, 3037, 3049, 3094,<br />

5560, 5570, 7048, 7059,<br />

7097, 7512, 10625,<br />

e13026, e21045<br />

Owugah, Tina TPS6638<br />

Owusu, Cynthia 9034, 9109,<br />

9123, e18133<br />

Owzar, Kouros 2533<br />

Oxnard, Ge<strong>of</strong>frey R. 7524,<br />

7547<br />

Oya, Mototsugu 9569,<br />

e15068<br />

Oyan, Basak 6550<br />

Oyen, Wim J.G. 3035,<br />

e13111<br />

Oza, Amit M. 3013, 3082,<br />

5001, 5010, 5019, 5020,<br />

5022, 5025, 5026, 5110,<br />

9003, e13001, e13598,<br />

e19616<br />

Ozaka, Masato e14677<br />

Ozaki, Michitaka 10578<br />

Ozaki, Toshifumi 9574<br />

Ozanne, Elissa e16504<br />

Ozawa, Taijirou e16032<br />

Ozaydin, Sukru 6550<br />

Ozbalak, Murat 6550<br />

Ozcimen, Ata e15104<br />

Ozdemir, Handan e11023<br />

Ozdemir, Nuriye 638,<br />

e11033, e11093, e14526<br />

Özdemir, Nuriye 1596,<br />

e13031<br />

Ozdener, Fatih 3565<br />

Ozdogan, Mustafa e14070,<br />

e19024<br />

Ozen, Haluk e15122<br />

Ozen, Ozlem e15572,<br />

e15573<br />

Ozenne, Gervais TPS7111<br />

Ozer Stillman, Ipek 6553<br />

Ozet, Ahmet 6550<br />

Ozguroglu, Mustafa e15112,<br />

e21143<br />

Ozisik, Yavuz Yasin e11020<br />

Ozisik, Yavuz e11092<br />

Ozkan, Elgin e21006<br />

Ozkan, Metin e14089,<br />

e14670<br />

Ozkaya, Enis 1097<br />

Ozkaynak, Mehmet Fevzi<br />

6537<br />

Ozmen, Vahit 9046<br />

Ozono, Seiichiro e15071,<br />

e15098<br />

Oztop, Ilhan 638<br />

Oztuna, Derya Gokmen 1596<br />

Ozturk, Akin e14698<br />

Ozturk, Mehmet Akif e21143<br />

Ozturk, Mustafa 6550<br />

Ozyilkan, Ozgur e11011,<br />

e11023, e11026, e11045,<br />

e11515, e14174<br />

Ozyoruk, Derya e20000<br />

P<br />

Paasche-Orlow, Michael 9004<br />

Pabla, Maninder e14632<br />

Pac, Joaquin 7011<br />

Paccagnella, Adriano 5513<br />

Pacchiarotti, Alberto e19002<br />

Pace, Andrea 2037, 2085<br />

Pace, Kenneth T 4633<br />

Pace, Margaret e13562<br />

Pace, Thaddeus 9122<br />

Pacetti, Paola 10611<br />

Pacheco, Jose 1048<br />

Pacheco, Luisa Silva e14597<br />

Pachman, Deirdre R. 9075<br />

Pachmann, Katharina 10600,<br />

e21063<br />

Pachon Ibanez, Jeronimo<br />

e16037<br />

Pachter, Jonathan A 1057<br />

Pachynski, Russell Kent<br />

e21071<br />

Pacini, Furio 5591<br />

Pack, Svetlana TPS7609<br />

Packeisen, Jens 10627<br />

Padbury, Rob TPS3643<br />

Padhani, Anwar R. 1066<br />

Padhya, Tapan 5564<br />

Padilla, Juan e12041<br />

Padrini, Robeto e13019<br />

Padua, Fernando Vidigal 643,<br />

e14680, e14728, e18078<br />

Paesmans, Marianne 3559,<br />

e18006<br />

Paetzold, Sylvie e19031<br />

Páez, David 3597, 3604,<br />

4026, 4096, e11018,<br />

e14034, e14052, e14104,<br />

e18057<br />

Pagani, Gianmatteo 1115<br />

Pagani, Olivia 9022<br />

Paganin, Fabrice 1574<br />

Page, David L 1005<br />

Page, Elizabeth 1545, 4653<br />

Page, Ray D. 7590, 7592,<br />

TPS7618<br />

Pagès, Cécile e13594<br />

Pages, jean-Christophe 3520<br />

Pagés, Mario 3536<br />

Paglialunga, Luca e18101<br />

Pagliaro, Lance C. 4533,<br />

e15092<br />

Pagliuchi, Marco e13098<br />

Pahernik, Sascha 4539<br />

Pai, Ashok P. 4607<br />

Pai, P. S. 5585<br />

Pai, Sara I 5506<br />

Paik, Paul K. 7014, 7505<br />

Paik, Seung-Woon TPS4149<br />

Paik, Soonmyung LBA506,<br />

LBA1000, 1025, TPS1142,<br />

3512<br />

Paintaud, Gilles TPS662<br />

Pairet, Geraldine e14044<br />

Pais-Costa, Sérgio Renato<br />

e14657<br />

Paiusco, Paola e19595<br />

Paiva Jr, Tadeu Ferreira<br />

e14680<br />

Paiva, Bianca Sakamoto<br />

Ribeiro e19602<br />

Paiva, Carlos Eduardo<br />

e14173, e19528, e19584,<br />

e19602<br />

Paiva, Henrique Soares<br />

e19552<br />

Paiva, Marcelo e11513<br />

Pajares, B. 4630<br />

Pajares, Bella 10616<br />

Pal, Sumanta Kumar 2538,<br />

4504, 4536, e15084,<br />

e15114, e15178, e16563<br />

Pal, Tuya 1585<br />

Palacios, Amalia 3571<br />

Palacios, Gemma 10512<br />

Palacka, Patrik TPS4677,<br />

e15028<br />

Palaia, Thomas e15504<br />

Palanichamy, Kamalakannan<br />

e19040<br />

Palapattu, Ganesh S.<br />

TPS4678<br />

769s


Palassini, Elena 10026,<br />

10053, 10065<br />

Palazzo, Juan 3621<br />

Palefsky, Joel 4030<br />

Palepu, Prakruthi R. 1535,<br />

7586, 9032, 10522<br />

Palesandro, Erica 10066,<br />

e20512<br />

Palescandolo, Emanuele<br />

2020, 5011<br />

Palese, Michael 4623<br />

Palesh, Oxana 9051, 9113,<br />

e16564<br />

Paley, Carole S. 8012,<br />

e16537<br />

Paliwal, Prashni TPS6634<br />

Palka, Magda e11527,<br />

e12015, e17502, e17503,<br />

e17515<br />

Pall, Georg e14719<br />

Palla, Shana L. 8536, 9127<br />

Pallares, Cinta e18057,<br />

e18130<br />

Paller, Channing Judith 3009,<br />

4559<br />

Palli, Swetha e16542<br />

Pallis, Athanasios G. 7081,<br />

7564<br />

Pallisgaard, Niels 5052<br />

Pallotta Guadalupe, Maria<br />

10074<br />

Pallotti, Maria Caterina<br />

10087<br />

Palma, Alejandra 9063<br />

Palma, David 6130, 7009<br />

Palma, Giuseppe e11052,<br />

e13539<br />

Palmbos, Phillip Lee 4591<br />

Palmer, Daniel H. TPS4150,<br />

e14625<br />

Palmer, Frank L. 5562<br />

Palmer, Gary A. 1015, 3533,<br />

7529, 10559, 10590<br />

Palmer, J. Lynn 9002, 9023,<br />

9025, 9062, 9063, 9096,<br />

e19643<br />

Palmer, Joycelynne 6547,<br />

8089, e18542<br />

Palmer, Steven C 9145<br />

Palmerini, Emanuela 10022,<br />

10026<br />

Palmero, Edenir Inez 1522<br />

Palmero, Ramon 7542,<br />

e18137<br />

Palmieri, Diane 505<br />

Palmieri, Giovannella e15034,<br />

e19038<br />

Palmieri, Giuseppe 8581,<br />

e14094, e18021<br />

Palmisano, Joseph N 6097<br />

Palomares, Melanie R. 1562,<br />

e15114<br />

Palomba, Grazia 8581,<br />

e14094<br />

Palomba, Maria Lia 6609,<br />

8054<br />

Palomo Colli, Miguel Angel<br />

e20005<br />

Palos, Guadalupe R. e19625,<br />

e19629<br />

Palozzo, Angelo Claudio<br />

e15072<br />

Palti, Yoram e14616<br />

Palumbo, Antonio Pierangelo<br />

TPS8112, TPS8113,<br />

TPS8114<br />

Palumbo, Michael Ostaric<br />

9557<br />

Palumbo, Raffaella 589<br />

Pamidighantam, Suresh 9055,<br />

e12501<br />

Pampaloni, Miguel H 8580,<br />

e13592<br />

Pan, Chong-xian 4607<br />

Pan, Edward 2099<br />

Pan, Hongming 7519^<br />

Pan, Jian-ji 5535, 5538<br />

Pan, Jianmin e16545<br />

Pan, Linda S. 2589,<br />

TPS8111, 8547, e17558<br />

Pan, Qiong 10042<br />

Pan, Qiulu 8596<br />

Pan, Summer 8078, 10051,<br />

e18546, e20509<br />

Pan, Xueliang Jeff 609<br />

Pan, Yumei 6038<br />

Pan, Zhiying 5504^<br />

Panageas, Katherine 2006,<br />

2518, 8575, 8598<br />

Panagiotarakou, Meropi<br />

e17557<br />

Panasci, Lawrence C. 1099<br />

Pancholi, Minjal J 7033<br />

Pande, Amitkumar U. 4006<br />

Pandey, Apurva e21105<br />

Pandey, Romy e14642<br />

Pandha, Hardev S. 4653,<br />

TPS4692^, e15517<br />

Pandit-Taskar, Neeta<br />

TPS9595<br />

Pandite, Lini 4605, e15059,<br />

e15085, e15087<br />

Pandya, Deep 10516<br />

Pandya, Kishan J. 7018<br />

Pandya, Naimish B. 6028,<br />

6060, 6075<br />

Pandya, Shuchi Sumant 2566<br />

Pang, Angela 1064, 1575,<br />

e19523<br />

Pang, Herbert CRA9013,<br />

e13589, e21038<br />

Pang, See-Tong e15064<br />

Pang, Vincent 1597<br />

Pang, Zuoliang e18013<br />

Pani, Giuseppe e14711<br />

Panka, David J 8516<br />

Pankow, Wulf 9033<br />

Panneerselvam, Ashok 4014,<br />

e15047, e15064<br />

Panni, Stefano 4097<br />

Panopoulos, Chris G. e11519<br />

Panos, Rebecca 10576<br />

Panse, Jens Peter e19610<br />

Pant, Alok e15523<br />

Pant, Rajive e20508<br />

Pant, Shubham 3008, 3041,<br />

3067, 3097, e17012<br />

Pantaleo, Maria A. 10087<br />

Pantazatos, Dionysios e21116<br />

Pantel, Klaus 1090,<br />

TPS1146, 10540, 10599<br />

Panting, Lisa 8593<br />

Pantos, Christos e11073<br />

Pantuck, Allan J. e13045<br />

Panu, Lori e16009<br />

Panzone, Filomena e19634<br />

Pao, William 6038, 7094,<br />

7532, 7568, 7589<br />

Paolicchi, Elisa 10611<br />

Paolini, Jolanda 4644<br />

Papa, Anselmo e11039,<br />

e11514, e12510, e19620<br />

Papa, Sophie 3100<br />

Papachristou, Irini 3044,<br />

TPS3115, 7603<br />

Papadimitrakopoulou, Vassiliki<br />

5524<br />

Papadimitriou, Christos A.<br />

592, 5033<br />

Papadimitriou, Konstantinos<br />

9090<br />

Papadopoulos, Iordanis<br />

e21027<br />

Papadopoulos, Kyriakos P.<br />

2512, 2555, 3001, 3019,<br />

3029, TPS3110, e13025,<br />

e13077, e13599, e13602,<br />

e13604<br />

Papadopoulos, Nicholas E.<br />

8514, e19009, e19014,<br />

e19039<br />

Papadopoulou, Eirini e21087,<br />

e21141<br />

Papagikos, Michael A. 5503,<br />

5515<br />

Papaiah Susheela, Sridhar<br />

e19619<br />

Papaioannou, Alexandra<br />

e14001<br />

Papakostidi, Aristoula e21098<br />

Papalini, Francisco e21025<br />

Papamichail, Michael 625,<br />

e15125<br />

Papanikolaou, Alexandros<br />

5033<br />

Papanikolaou, Xen<strong>of</strong>on 8041,<br />

e18548<br />

Papayannidis, Cristina 6531<br />

Pape, Ulrich-Frank 4128<br />

Papin, Jason A 8597<br />

Papini, Enrico 1604<br />

Pappa, Vassiliki e21027<br />

Pappagallo, Giovanni L. 7577,<br />

e15160, e18026<br />

Pappas, Theodore N. e14724<br />

Pappo, Alberto S. 8504,<br />

9509<br />

Pappou, Efi 625, 2508,<br />

e15125<br />

Papsdorf, Kirstin 2000<br />

Paquet, Agnes 603, 608, 614<br />

Paquette, Ronald 6503, 6624<br />

Parafioriti, Antonina 10022<br />

Paraiso, Isbou e21014,<br />

e21046<br />

Parambi, Ron J. 5594,<br />

e16059<br />

Paramio, Jesus 4584<br />

Parashar, Deepak e14100<br />

Parchment, Ralph E. 3031<br />

Pardanani, Animesh Dev<br />

6614, TPS6639, TPS6641<br />

Pardee, Timothy S. 6524^<br />

Pardo, Beatriz 5068, e15575<br />

Pardo, Nuria 4579, e12504<br />

Pardo, Wandaly Ibis e18547<br />

Pardoll, Drew M. CRA2509,<br />

2510, 5506, e13559<br />

Paré, Laia e14052, e14104,<br />

e18057<br />

Parekh, Hiral D. e17010<br />

Parent, Teresa e17547,<br />

e17550<br />

Parente, Barbara e18111<br />

Parente, Phillip e15152<br />

Parganiha, Arti e14006<br />

Parham, David 9509, 9535<br />

Parham, Laura R 10532<br />

Parhar, Tarnjit 3610<br />

Paridaens, Robert 1020<br />

Parienti, Jean-Jacques<br />

TPS7112<br />

Parikh, Kunal e14656<br />

Parikh, Nila 3561<br />

Parikh, Omi CRA4502<br />

Parikh, Rohan e14627<br />

Parikh, Unnati e14632<br />

Parise, Carol 1582, 1606<br />

Parise, Robert A 2553<br />

Parise, Sara e16026<br />

Park, Bernard J. 7024<br />

Park, Byeong-Woo 2542<br />

Park, Byung-Kiu 9579<br />

Park, Chan Hee 3606<br />

Park, Cho Hyung e14558<br />

Park, Chul-Kee 2041<br />

Park, Daniel e15171<br />

Park, Deric M. 3037<br />

Park, Elyse R. 6048, 6106,<br />

9101<br />

Park, Hee Sook 1003,<br />

e14688, e19554<br />

Park, Hyeon Jin 9579<br />

Park, Ilwoo 4660<br />

Park, In Hae 1003<br />

Park, In Ja 3556<br />

Park, In-Ae e11532<br />

Park, Inkeun 10085, e14543,<br />

e15080<br />

Park, Jae Hong 6532<br />

Park, Jae-Gahb 3624<br />

Park, Jangchul e17510,<br />

e18145<br />

Park, Jenny e21159<br />

Park, Jeong-Yeol e15559<br />

Park, Jin Hyun 7566<br />

Park, Jinha Mark 1035<br />

Park, Jinny e14137, e14685<br />

Park, Jong Ho e17523<br />

Park, Joon Oh 4011, 4064<br />

Park, Joong-Won 4033, 4103<br />

Park, Joonoh e13094<br />

Park, Julie R. 9533, 9534<br />

Park, Jun Seong e18519<br />

Park, Jun Won e21043<br />

Park, Keon Uk e14570<br />

Park, Keon-Woo 4064<br />

Park, Keunchil 7004, 7104,<br />

7549, 7557, TPS7614,<br />

e16003, e18093, e18148<br />

Park, Kwonoh 5586, e15080<br />

Park, Kyong Hwa 2542,<br />

e12029<br />

Park, Kyu Hyun e14081<br />

Park, Kyu Joo 3624<br />

Park, Kyung 4649<br />

Park, Kyung-Mi 3076,<br />

e13092, e14572<br />

Park, Lee Chun e13094,<br />

e16003, e18148<br />

Park, Min Keun e14114<br />

Park, Min-Jung 2041<br />

Park, Noh Jin 2587<br />

Park, Rachel S. 4577, 4580,<br />

4585<br />

Park, Sang Min 9066<br />

Park, Se Hoon 4064, e13094<br />

Park, Seong Yong e16534<br />

Park, Seongjoon e15080<br />

Park, Seung Woo e19538<br />

Park, Seung-Il 4093, 7510,<br />

e14573<br />

Park, Silvia 644, 7104,<br />

e13094, e16003, e18148<br />

Park, Sook Ryun 4028<br />

Park, Sophie 6523, 6534,<br />

8026<br />

Park, Steven I. TPS8109,<br />

e13074<br />

Park, Sun Jin e14128<br />

Park, Sung Kyu e14688,<br />

e19554<br />

Park, Wungki e14169,<br />

e14170<br />

770s Numerals refer to abstract number


Park, Yeon Hee 644, 1003,<br />

2542<br />

Park, Yeon Ho e14685<br />

Park, Yo An e13092<br />

Park, Young Soo e14543<br />

Park, Young Suk 3531, 4064,<br />

e13044, e13094, e14072<br />

Park, Young-Hun 6564<br />

Park, Young-Iee 4028,<br />

e14572<br />

Parker, Alex 3533<br />

Parker, Bethany H. e14078<br />

Parker, Charla A. 594<br />

Parker, Chris LBA4512, 4551<br />

Parker, Douglas 8543<br />

Parker, Imelda 581, e13520<br />

Parker, Joel S 10500<br />

Parker, Lynn 5095<br />

Parker, Matthew 9518<br />

Parker, Pablo Miguel 8089<br />

Parker, Patricia A. 6054,<br />

9031<br />

Parker, Susan 2510<br />

Parli, Teresa 8020<br />

Parlour, Louise 5527<br />

Parmakhtiar, Basmina 8524<br />

Parmar, Mahesh 4509, 6091<br />

Parmar, Simrit 8102<br />

Parmar, Vani 1108, e11552<br />

Parmentier, Gilles 10562<br />

Parmiani, Giorgio 8513, 8529<br />

Parmigiani, Giovanni 2005<br />

Parnaby, Adam e15198<br />

Parnes, Howard L. 3043,<br />

4569<br />

Parnis, Francis 7079^<br />

Parno, Jeff 3016, 7030<br />

Parot, Xavier 4129<br />

Parpia, Sameer LBA1031<br />

Parra, Juan Francisco 4089<br />

Parras, Manuela e11517<br />

Parrella, Paola e21092<br />

Parrilla, Lucia e15033<br />

Parsons, Henrique Afonseca<br />

9063, e13534<br />

Parsons, Michael W. 1076,<br />

2018<br />

Parsons, Ramon 10516<br />

Parsons, Stephen H 8082<br />

<strong>Part</strong>han, Anju e11077<br />

<strong>Part</strong>hasarathy, Mala e13121<br />

<strong>Part</strong>hasarathy, Ramya 3087<br />

<strong>Part</strong>ridge, Ann H. 1100,<br />

6025, 9008, 9071, 9084,<br />

9100, e16515<br />

Parulekar, Wendy 500, 4509,<br />

TPS5600<br />

Parvathaneni, Upendra 5503,<br />

5515, 9008<br />

Parvin, Jeffrey e19053<br />

Paschold, Eugene Henry 7584<br />

Pasciuti, Giulia e11039,<br />

e11514, e19620<br />

Pascoletti, Gaetano 1044<br />

Pascual, Tomas 642,<br />

e15033, e15552<br />

Pasello, Giulia 7082<br />

Pasero, Christine e15155<br />

Pasetka, Mark e15180<br />

Pashos, Chris L 6122, 8037,<br />

e18556, e18561<br />

Pasini, Felice e13019<br />

Pasion, Kristine A. e13574<br />

Paskett, Electra D. CRA9013<br />

Pasqual, Nicolas 10562<br />

Pasquier, Florence e11012<br />

Pasquini, Enzo LBA4001<br />

Pass, Harvey e17528,<br />

e21012<br />

Numerals refer to abstract number<br />

Passalacqua, Rodolfo 4097,<br />

9120<br />

Passamonti, Francesco<br />

6514^, 6625^, 6626^,<br />

TPS6639<br />

Passardi, Alessandro 4017<br />

Passaro, Antonio e18065<br />

Passioukov, Alexandre 5522,<br />

7544<br />

Pastan, Ira 2503^, 7030<br />

Pastina, Pierpaolo e13098<br />

Pastrana, Miguel Angel<br />

e11035<br />

Paszat, Lawrence Frank 1123,<br />

6027, 6134<br />

Paszkiewicz-Kozik, Ewa<br />

e18512<br />

Patal, Payal 6629<br />

Patchett, Matthew 6595<br />

Patel, Akshal e11544<br />

Patel, Amit S. e16527<br />

Patel, Anand 8072<br />

Patel, Bijal e17524<br />

Patel, Devalben 1535, 7586,<br />

10522<br />

Patel, Jai Narendra e13109<br />

Patel, Jay 9507<br />

Patel, Jyoti D. LBA4, 3083<br />

Patel, Kiran 3102, 3528,<br />

LBA8509, 8510, e19046<br />

Patel, Krina K. 8093<br />

Patel, Manali I. 6050<br />

Patel, Manish R. 3631,<br />

TPS8115<br />

Patel, Mital 2076, 2092,<br />

2093, 2096, 2100<br />

Patel, Monaliben e14175<br />

Patel, Nayana e15176<br />

Patel, Neema e17004<br />

Patel, Oneel e15117<br />

Patel, Pooja Nandwani<br />

e15505<br />

Patel, Poulam M 8517<br />

Patel, Premal H. TPS2616,<br />

3021, e13114<br />

Patel, Priti 8086, e18569<br />

Patel, Purvi D. 5562<br />

Patel, Sapna Pradyuman<br />

8514, e19009, e19014,<br />

e19039<br />

Patel, Shejal B e19551<br />

Patel, Shreyaskumar 10071,<br />

TPS10101, e19520<br />

Patel, Snehal G. 5545^,<br />

5562<br />

Patel, Tejal Amar 2015<br />

Patel, Uday Bharat e14097<br />

Patel, Vihas 9086<br />

Patel, Vipul e15171<br />

Patel, Yash R. e15179<br />

Patel-Donnelly, Dipti 3069,<br />

8098<br />

Paterlini-Brechot, Patrizia<br />

7591<br />

Paterson, Daniel 1057<br />

Pathak, Ashutosh Kumar<br />

TPS2613<br />

Pathak, Prathamesh 570<br />

Pather, Selvin TPS9153<br />

Pathy, Sushmita 5104<br />

Pati, H P e17000<br />

Patil, Chirag 2080, 2087<br />

Patil, Dattatraya Hari e14692<br />

Patil, Poonam e14710,<br />

e15516<br />

Patil, Ritesh 625, 2508,<br />

2529<br />

Patil, Shekhar e11511,<br />

e12030, e14721, e18041<br />

Patil, Sujata 557, 575, 1006,<br />

1118, 4532, 6001, 9104,<br />

10618<br />

Patil, Vijay 5585, e16028,<br />

e17500<br />

Patnaik, Amita 2512, 2555,<br />

3001, 3019, TPS3110,<br />

e13025, e13576, e13599,<br />

e13600^, e13602, e13604<br />

Patnaik, Mrinal M. 6614<br />

Patrick, Patricia e15504<br />

Patrick-Miller, Linda J. 9106,<br />

e15173<br />

Patrikidou, Anna 10044<br />

Patruno, Rosa e21113,<br />

e21134<br />

Patt, Debra A. 6109<br />

Patt, Richard H. TPS4152,<br />

e14566<br />

Pattabiraman, Nagarajan<br />

e13545<br />

Pattali, Shinoj e14186,<br />

e14590<br />

Pattaras, John e15006<br />

Patterson, Christopher J<br />

8042, 8106, 8107,<br />

e18575<br />

Patti, Marco G. 10090<br />

Patton, J. Scott e13581<br />

Paty, Philip 3577, 3583<br />

Patyna, Shem 4118<br />

Patzak, Ingrid 2504<br />

Paucar, Daniel 8598<br />

Pauels, Hans-Gerd e18532<br />

Paul, Devchand 547<br />

Paul, Diane B. 9047<br />

Paul, Doru 6123<br />

Paul, MJ e19562<br />

Pauligk, Claudia 4080, 4083,<br />

4090, 10552, e14557,<br />

e21063<br />

Paulk, M. Elizabeth e16500<br />

Paulli, Marco e13103<br />

Paulsau, Antonello e15194<br />

Paulson, Guillermo e15571<br />

Paulus, Jessica e14005<br />

Pautier, Patricia 5028, 5083,<br />

10042, 10098<br />

Pavel, Marianne E. 4014,<br />

4122, 4128, e14564<br />

Pavese, Ida e18065<br />

Pavey, Emily S 3617<br />

Pavia, Orestes Antonio 1060,<br />

e11015, e18547<br />

Pavia-Jimenez, Andrea<br />

TPS4678<br />

Paviot, Alexandre 5565<br />

Pavlakis, Nick 3515, 7079^,<br />

e19642<br />

Pavlicek, Adam 3509<br />

Pavlick, Anna C. 2589,<br />

8503^, 8508, 8550, 8557,<br />

8565, 8567, 8589,<br />

e13598, e19003<br />

Pavlidis, Nicholas 573, 592,<br />

10575<br />

Pavlov, Dmitri 6512<br />

Pavlovitz, Brian 6572<br />

Pawaskar, Dipti K e13006<br />

Pawitan, Yudi 4561<br />

Pawlik, Timothy M. 3616<br />

Pawlik, Timothy M 3596,<br />

4055, 4114<br />

Pawlinski, Rafal e14505<br />

Pawloski, Pamala A. e14160<br />

Pawlowski, Ryszard 10561<br />

Paydas, Semra 6550, e19024<br />

Payne Ondracek, Rochelle<br />

e15203, e15204<br />

Payrard, Sandrine 8057<br />

Paz, Isaac Benjamin 1035<br />

Paz, Maria fe e21158<br />

Paz-Ares, Luis G. 7542,<br />

TPS7615, e13605, e18100<br />

Paz-Ares, Luis 2042, 3062,<br />

LBA7507, 7544, TPS7616,<br />

TPS9149^, e16037<br />

Paz-Querubin, Emma R<br />

e14124, e18516<br />

Paz-Querubin,, Emma e19638<br />

Pazart, Lionel e19623<br />

Pazdur, Richard 3074<br />

Pazzola, Antonio e14094<br />

Peach, James TPS10633^<br />

Peachey, Lynnda LBA4001<br />

Peachey, Matt 9004<br />

Peacock, Nancy Walker 1018,<br />

4127<br />

Peacock, Stuart 549^<br />

Pearlberg, Joseph 7556<br />

Pearlstone, David B. 1132<br />

Pearse, Maria TPS4690<br />

Pearse, Roger N 8036<br />

Pearson, Alexander T. 4573<br />

Pearson, Andrew DJ 9542,<br />

9583<br />

Pearson, Richard 8520<br />

Pearson, Sarah R LBA4001<br />

Pecchio, Silvia 627<br />

Pechacova, Zdenka e14631<br />

Peck, Rahul e18095<br />

Peck-Radosavljevic, Markus<br />

e14681<br />

Pecora, Andrew e18507,<br />

e19641<br />

Pecorari, Giancarlo 5546,<br />

5548<br />

Pecorelli, Sergio L. e15549<br />

Pectasides, Dimitrios G. 573,<br />

5033, e21027<br />

Pectasides, Eirini 5576<br />

Pedani, Fulvia 5548, e11524<br />

Peddareddigari, Vijay Gopal<br />

Reddy 3000, 3023<br />

Peddi, Prakash e16009<br />

Peddi, Prashanth 8536<br />

Pedersen, Hans Christian<br />

e21129<br />

Pedersen, Joanna Vitfell<br />

10038<br />

Pedersen, Kasper 632, 2536<br />

Pedersen, Lars Moeller 8074<br />

Pedrazzoli, Paolo e13103<br />

Pedulla, Lillian Vitale e19637<br />

Peer, Avivit 4564, 4619,<br />

e15058<br />

Peereboom, David M. 2018,<br />

2076, 2092, 2100,<br />

TPS2107<br />

Peeters, Dieter 10504<br />

Peeters, Justine 10545,<br />

10597, e21013<br />

Peeters, Marc 3535, 3559,<br />

3581, 4050, e13051<br />

Peeters, Olivier 8559<br />

Pegoraro, Cristina e13019<br />

Pegourie, Brigitte 8014<br />

Pegram, Mark D. LBA1, 608<br />

Peguet, Elodie e14148<br />

Pei, Jianming 4605<br />

Pei, Jun e12013, e13528,<br />

e18088<br />

Peila, Elena e11524<br />

Peintinger, Florentina 515<br />

Peiper, Matthias e11556<br />

Peipp, Matthias e18565<br />

Peiretti, Michele 5090<br />

Peiro, Rafael e16025<br />

771s


Peisker, Uwe 10600<br />

Peker, Deniz e18503,<br />

e18506, e21123<br />

Pekle, Karen 8036<br />

Pelayo, Eva e11028<br />

Pelayo, Miguel Ricardo 5564<br />

Pelegrin, Andre 5069<br />

Peligros, Isabel 4089<br />

Pelizon, Christina H. TPS653,<br />

TPS654^<br />

Pella, Nicoletta 3612<br />

Pellacani, Giovanni 8578<br />

Pellegatta, Serena 2083<br />

Pellegrino, Antonio e18048<br />

Pellegrino, Arianna e18065<br />

Pellegrino, Renata e16041<br />

Peller, Patrick J. 2031<br />

Pelles-Avraham, Sharon 5078<br />

Pelling, Katy TPS651<br />

Pelloski, Christopher E. 9544<br />

Pelmus, Manuela TPS1139<br />

Pelt, Gabi W. van e14151<br />

Peltonen, Nina e15564<br />

Pelusio, Adina e13044<br />

Pelzer, Uwe 4044, e14533<br />

Pemberton, Kristine 3048<br />

Pemmaraju, Naveen 6544,<br />

6578, 6592, 6597<br />

Pena, Carol Elaine e15086<br />

Peña, Jose Maria 3601,<br />

e14093, e14144<br />

Pena, Omar 7068<br />

Pena-Llopis, Samuel 4616<br />

Penalver, Juan-Carlos 7011<br />

Penault-Llorca, Frederique<br />

Madeleine 543, 568^,<br />

1011<br />

Penault-Llorca, Frederique<br />

1051<br />

Pendergast, Jane F 6034<br />

Pendharkar, Dinesh e16057<br />

Pendse, Deepa 4581^,<br />

e15022<br />

Pendurti, Gopichand e18537<br />

Penel, Nicolas 2540, 10005,<br />

10006, 10007, 10010,<br />

10013, 10014, 10018,<br />

10020, 10023, 10027,<br />

10035, 10042, 10056,<br />

TPS10102^<br />

Peng, Feng e13538, e18033<br />

Peng, Guangbin 2554, 5533,<br />

7002<br />

Peng, Shiwen 5506<br />

Peng, Xianghong e13110<br />

Peng, Yingwei 1583<br />

Peng, Zhuochun 4561<br />

Pengo, Stefano 10022<br />

Penn, Dolly 6104<br />

Pennacchioli, Elisabetta<br />

e19035, e19036<br />

Pennathur, Arjun 7074<br />

Pennell, Nathan A. 2081,<br />

7010, e17563, e18046,<br />

e18085<br />

Pennella, Eduardo J. LBA4<br />

Pennella, Eduardo 5533,<br />

e16053<br />

Penning, Trevor 4520<br />

Pennington, Kenneth L. 4043<br />

Pennock, Gregory K.<br />

TPS10099, e13088<br />

Pensabene, Matilde 1554<br />

Penson, Richard T. 5012,<br />

5029<br />

Pentheroudakis, George E.<br />

592, 5033, 10575<br />

Pentland, Alice P 9027<br />

Pentlow, Keith S. 5509^<br />

Pentsova, Elena 2008<br />

Pentz, Rebecca D. e13026<br />

Penuel, Elicia M. e13048<br />

Pénzváltó, Zsófia e21005<br />

Peock, Susan 1545<br />

Peoples, George Earl 625,<br />

2508, 2529<br />

Pepe, Carmela e18061,<br />

e18098, e19626<br />

Peppercorn, Jeffrey M. 6095,<br />

6128<br />

Peppone, Luke Joseph 9009,<br />

9010, 9014, 9028, 9141,<br />

9142<br />

Pera, Miguel 3586<br />

Peralta, Sergi e18030<br />

Percy, Andrew 4543, 4635<br />

Perdikouri, Eleni Isidora<br />

e18098<br />

Perdona, Gleici da Silva<br />

Castro e19628<br />

Perdue, Nikole 2563<br />

Perea, Rachelle 3032<br />

Perea, S<strong>of</strong>ia 602, e11006<br />

Pereda, Teresa e14116<br />

Pereira, Allan Andresson Lima<br />

e11014<br />

Pereira, Augusto Akikubo<br />

Rodrigues 643, e14680,<br />

e14728, e18078<br />

Pereira, Bruno dos Santos<br />

Vilhena e14027<br />

Pereira, Deolinda LBA5002^,<br />

e15532<br />

Pereira, Jose Rodrigues 7503,<br />

7506, 7512, e18078<br />

Pereira, Verónica 3536,<br />

e13540<br />

Perel, Yves 9517<br />

Perello, Antonia e11036<br />

Perelman, Marina e17509<br />

Perelman, Michael Sidney<br />

e19544, e19567, e19578<br />

Perentesis, John Peter 9541,<br />

9581, e20007<br />

Perepu, Usha S. 8061<br />

Perera, Nicole 1535, 7586<br />

Peretz, Tamar 10034<br />

Perey, Lucien e11048<br />

Perez Callejo, David e11527,<br />

e12015, e17502, e17515<br />

Perez Garcia, Jose Ignacio<br />

10555<br />

Perez Ramirez, Sara e12003<br />

Perez Regadera, Jose Fermin<br />

e15033<br />

Pérez Segura, Pedro 2045<br />

Perez Verdera, Palmira<br />

e11021<br />

Pérez Villanueva, Heynar<br />

e20005<br />

Perez, Cesar Augusto e17564<br />

Perez, Crystal 2512<br />

Perez, Diego 602, e11535,<br />

e14116, e21088<br />

Perez, Edith A. 605, TPS653,<br />

LBA1000, CRA1002, 1005,<br />

1021, 1083, TPS1140^,<br />

2015, e13501<br />

Perez, Eloy 9554<br />

Perez, Francisco Javier<br />

e11545, e15144, e17545<br />

Perez, Francisco Jose e11517<br />

Perez, Iratxe TPS10101<br />

Perez, Javier e18011<br />

Perez, Jose Ricardo TPS6640<br />

Perez, Kimberly 4098<br />

Perez, Marta 9558<br />

Perez, Omar D. 2101<br />

Perez, Quionia e15149,<br />

e18044<br />

Perez, Solene 10562<br />

Perez, Sonia A. 625, 2508,<br />

e15125<br />

Perez, Thomas B e21060<br />

Perez-Carrion, Ramon 1111,<br />

e11074, e11081<br />

Perez-Cruz, Pedro Emilio<br />

9049, e19528, e19584<br />

Perez-Cruz, Pedro Emilio<br />

9023<br />

Perez-Fidalgo, Jose Alejandro<br />

1074, TPS3637, e11523,<br />

e14589<br />

Perez-Garcia, Jose Manuel<br />

509, 566, 619, 3003,<br />

3014<br />

Perez-Gonzalez, Nuria 5520,<br />

e15094, e18069, e18106,<br />

e21015<br />

Perez-Gracia, Jose Luis<br />

TPS4674, 8572, e15041,<br />

e18008<br />

Perez-Moreno, Pablo Diego<br />

TPS7612<br />

Perez-Rivas, L.G. 4630<br />

Pérez-Ruiz, Elisabeth e14115,<br />

e14116<br />

Perez-Soler, Roman 7573,<br />

e18113<br />

Pérez-Valderrama, Begoña<br />

TPS4674, e15149<br />

Perez-Villa, L. 4630, e14115<br />

Peria, Fernanda Maris<br />

e12512, e19628<br />

Pericay Pijaume, Carles<br />

e14097<br />

Pericay, Carles 3571, 3586,<br />

TPS3637, 4079, e14041<br />

Pericleous, Marinos e14696,<br />

e17543<br />

Perin, Alessandra e12037,<br />

e15160<br />

Perin, Tiziana 10554<br />

Perini, Rodolfo F. e13592<br />

Perisanidis, Beata e14095,<br />

e14099<br />

Perkins, Geraldine 10525<br />

Perkins, Sherrie L 9501<br />

Perkins, Susan M. 4534,<br />

e14683<br />

Perkkio, Mikko 9590<br />

Perlewitz, Kelly Shea 10011^<br />

Perlman, Elizabeth 9534<br />

Perlroth, Daniella 4666,<br />

e15186<br />

Perng, Reury-Perng e18132<br />

Pernod, Gilles 1580<br />

Pernot, Simon 4087<br />

Perol, David 4614, 4625,<br />

7574, 10006, 10092,<br />

10095, 10562, e16560<br />

Perol, Maurice 7574, e16560<br />

Peron, Julien 601, e14730<br />

Perona, Rosario 3601,<br />

e14093, e14144<br />

Peronto, Erica 6524^<br />

Peros, George e21027<br />

Perotti, Antonella 3080<br />

Perou, Charles M. 1008,<br />

1027, 1524, 10500<br />

Perraud, Yves e15109<br />

Perrin, Allison e14525<br />

Perrin, Paul e15182<br />

Perrino, Matteo 2580, 7061,<br />

7082, e21139<br />

Perrone, Federica 10016<br />

Perrone, Francesco TPS4151,<br />

LBA5003, e18021<br />

Perrone, Giuseppe e18021<br />

Perrone, Tania e19612<br />

Perrot, Delphine 10014<br />

Perruccio, Katia e21096<br />

Perry, James R. TPS2104<br />

Perry, Matthew 4640,<br />

e15100<br />

Persico, Justin e11548<br />

Persigehl, Thorsten 2541<br />

Persky, Daniel Oscar<br />

TPS8109, e18534, e18536<br />

Persky, Mark 5581<br />

Person, Bernadette e14148<br />

Personeni, Nicola e14672<br />

Pertschuk, Daniel 2101<br />

Pertschy, Danielle e15540<br />

Pervez, Nadeem e15540<br />

Pesavento, Tony e16554<br />

Pescarmona, Edoardo 1050<br />

Pesce, Catherine 557<br />

Pesce, Veronica e14598<br />

Peschaud, Frederique 8540<br />

Pesek, Milos e18062<br />

Pesonen, Sari e13035<br />

Pestell, Richard G. e13545<br />

Pestova, Ekaterina e21060,<br />

e21111<br />

Petain, Aurélie e13036<br />

Petekkaya, Ibrahim e11020,<br />

e11080, e11092, e11509<br />

Petenko, Natalia N. 2524<br />

Petermann-Meyer, Andrea<br />

e19610<br />

Peters, Ann-Marie e17558<br />

Peters, Christopher A. 6031,<br />

TPS9150<br />

Peters, Godefridus J 10611<br />

Peters, John 4611<br />

Peters, Katherine B. 2027,<br />

2074^, 2090^, 2094^,<br />

2095^<br />

Peters, Lester J. 5571<br />

Peters, Martin 5005<br />

Petersen, Gloria M. e14594<br />

Petersen, Judith A. CRA6009<br />

Petersen, Peter Meidahl<br />

e18527<br />

Petersen, Volker 4065<br />

Peterson, Amy C. TPS668,<br />

TPS4695<br />

Peterson, Gilbert e13571<br />

Peterson, Lanell TPS3109<br />

Peterson, Lindsay e14684<br />

Peterson, Patrick 1068, 7075<br />

Peterson, Scott 3024<br />

Peterson, Susan K. e13026<br />

Petillo, David 4624<br />

Petillon, Marie-Odile 8014<br />

Petit, Robert G. 5106,<br />

e15546<br />

Petit, Thierry TPS646,<br />

TPS653, 1051, e11012,<br />

e11542, e15535<br />

Petito, Emily 3020<br />

Petoka, Renana e14731<br />

Petrakova, Katarina 559,<br />

e11047<br />

Petrella, Teresa M. e13049<br />

Petrelli, Fausto e19612<br />

Petrelli, Nicholas J. TPS1615<br />

Petri, Roberto 3612<br />

Petricoin, Emanuel e14151,<br />

e15549<br />

Petridou, Eleni e20002<br />

Petrik, David W. 6071<br />

Petrini, Mario 8006<br />

Petro, Daniel P. 3037, 3049<br />

772s Numerals refer to abstract number


Petro, Robin M. 6127<br />

Petrocelli, Teresa e13107<br />

Petr<strong>of</strong>f, Brian K. 520<br />

Petroianu, Andy e12018<br />

Petroni, Gina R. 3047, 8530<br />

Petropoulos, Christos J. 603,<br />

608, 614<br />

Petros, John e15187<br />

Petrosiute, Agne 9520<br />

Petrova, Tatyana e13059<br />

Petrow, Peter e15524,<br />

e15535<br />

Petru, Edgar LBA5000, 5031<br />

Petruzelka, Lubos B. e11047,<br />

e14631<br />

Petry, Christoph 542<br />

Petry, Vanessa e11513<br />

Petrylak, Daniel Peter 4522,<br />

4525, 4656, 4662,<br />

TPS4675^, TPS4693<br />

Pettaway, Curtis A. 9031<br />

Pettengell, Ruth 8056<br />

Petty, Nathan 8041, e18548<br />

Petty, Russell LBA4001<br />

Peuser, Bettina TPS3639<br />

Peuvrel, Lucie e19019<br />

Peylan-Ramu, Nili 10034<br />

Peynircioglu, Bora TPS4148<br />

Peyrade, Frederic 5516^,<br />

5521, 6633, e16044,<br />

e16051, e16055<br />

Peyrat, Patrice e14730<br />

Peyro Saint Paul, Helene P.<br />

TPS667<br />

Peyton, James D. 7035<br />

Peyton, Michael e18077<br />

Pezaro, Carmel Jo 4553,<br />

e15134, e15136<br />

Pezet, Denis 10505<br />

Pezzutto, Antonio e14140,<br />

e15053, e18552<br />

Pfeffer, Raphael M. e14143<br />

Pfeffer, Ulrich e21065<br />

Pfeiffer, Per 3538, 3548,<br />

e14053, e14517, e14633<br />

Pfeiffer, Sebastian 8004<br />

Pfeiler, Georg 514<br />

Pfister, David A. e15195<br />

Pfister, David G. 5509^,<br />

5514, 5537, 5545^, 6001<br />

Pfister, Thomas D. 3031<br />

Pfisterer, Katharina 5572<br />

Pfreundschuh, Michael 1523,<br />

6510, 6584, 8004, 8022,<br />

8024, 8025<br />

Phalen, Adrien e15164<br />

Pham, Deborah e15577<br />

Pham, Huyen e13531<br />

Pham, Minh Thy e16523<br />

Pham, Tam 584, TPS10631<br />

Pham, Thang V e18094<br />

Phan, Alexandria T. 4085,<br />

4641, 9002<br />

Phan, Alexandria 4639<br />

Phan, Jack 5561, 5589<br />

Phelan, Vanessa H 5560<br />

Phelps, Andrea C. 9116,<br />

9132<br />

Phelps, Andrea Catherine<br />

9102<br />

Phelps, Michael E. 10012<br />

Philip, Jennifer e16562<br />

Philip, Philip Agop 3579,<br />

4019, e14163, e14519,<br />

e14609, e14610<br />

Philips, George 4592<br />

Philips, Santosh 525<br />

Phillipe, Colucci 6619^<br />

Numerals refer to abstract number<br />

Phillips, Callista Maria<br />

e14001<br />

Phillips, Christine L. 9581<br />

Phillips, Claire TPS2104<br />

Phillips, Jennifer 6584<br />

Phillips, Joanna J 2026<br />

Phillips, Jonathan 4060<br />

Phillips, Kelly-Anne TPS1136,<br />

1502, 9022<br />

Phillips, Larry 3087<br />

Phillips, Luann 9547,<br />

e18522<br />

Phillips, Martin 5062^<br />

Phillips, Paula 6108<br />

Phillips, Penny 3572<br />

Phillips, Teresa A. 520<br />

Phillips, Tycel Jovelle 9085<br />

Phipps, Amanda I. 3526<br />

Phoenix, Timothy 9518<br />

Phuan, Crystal e14155<br />

Phung, Yen e14118<br />

Phuphanich, Surasak 2080,<br />

2084, 2087, TPS2107<br />

Piacentini, Federico 631<br />

Piana, Raimondo 10055<br />

Pianetti, Gerson Antonio<br />

e13008<br />

Pianowski, Luiz F e19552<br />

Piao, Yongzhe 4092, e14575<br />

Piao, Yuji 2011<br />

Piato, Jose R. e11513<br />

Piazik, Breanne Lee 6013<br />

Piazza, Dario e14676<br />

Picaud, Francois 601<br />

Piccaluga, Pier Paolo e13569<br />

Piccart-Gebhart, Martine J.<br />

507, 512, 540, 551, 559,<br />

596, 604, TPS649, 1022,<br />

3559, e13605, e21010<br />

Picci, Piero 10022, 10079<br />

Picciotti, Giovanna 9098<br />

Piccirillo, Maria Carmela<br />

e14130<br />

Piccolini, Ezio 7084<br />

Pichinuk, Edward e13018<br />

Pichler, Josef 2034<br />

Pichler, Martin e14066,<br />

e18525<br />

Pichon, Audrey e15145<br />

Pichon, Christophe e21138<br />

Pickering, Curtis R. 5524<br />

Pickett-Gies, Cheryl Ann 529<br />

Picone, Antonio e18039<br />

Picone, Giuseppe e12512<br />

Picozzi, Vincent J. 4038<br />

Picus, Joel 3047, 4118,<br />

4571, 10088<br />

Piderman, Katherine M. 1610<br />

Piedmonte, Marion 1519<br />

Piekarz, Richard 2553, 7103,<br />

9144, e19633<br />

Pienkowski, Tadeusz 9046,<br />

e11553<br />

Pier, Thomas e13537<br />

Pierantoni, Chiara e21070<br />

Pierce, Aisling 622, 1042<br />

Pierce, Sherry 6544, 6578,<br />

6597, 6603, 6607<br />

Pierga, Jean-Yves TPS646,<br />

10562<br />

Pieri, Gabriella 5513<br />

Pierobon, Mariaelena e14151,<br />

e15549<br />

Pierotti, Marco TPS9155<br />

Pierpoint, Ann 3043<br />

Pierre, Andrew 9032<br />

Pierre, Bonnette e18102<br />

Pierron, Gaëlle 9510<br />

Pierson, Namali e17012<br />

Pietanza, Maria Catherine<br />

7014, 7052, 7527,<br />

e17558, e19647<br />

Pietra, Claudio e19577<br />

Pietrabissa, Andrea e13103<br />

Pietragalla, Antonella 5059<br />

Pietrantonio, Filippo 7581,<br />

e21118<br />

Pietri, Elisabetta e11054<br />

Pietrobon, Ricardo e14544,<br />

e14562<br />

Piette, Francois e18505<br />

Pietzner, Klaus 5080,<br />

e15531<br />

Pignata, Sandro LBA5003,<br />

5017^<br />

Pignochino, Ymera 10066<br />

Pignon, Jean-Pierre 3063,<br />

3507, 4032, 7007, 10556<br />

Piha-Paul, Sarina Anne 3106,<br />

3107^, 8551, 10510,<br />

10607, e13020, e13595<br />

Pike, Ian e21091<br />

Pike, Laura 6527, e13541<br />

Pikul, Frank e16513<br />

Pilcher, James 4640<br />

Pilegaard, Han Kristian 7001<br />

Pilegaard, Hans e21110<br />

Pileri, Stefano A e13569<br />

Pileri, Stefano 8006<br />

Pilger, Adak e19597<br />

Pilgrim, Charles Henry Caldow<br />

TPS3643<br />

Pili, Roberto 4550, 4619,<br />

4665, TPS4687, 10609,<br />

e15042, e15058<br />

Pilla, Lorenzo 8513, 8529<br />

Pillai, Raji 10584<br />

Pillai, Rathi Narayana 10625<br />

Pillay, Sada 2042<br />

Pilling, Amanda B. 7504<br />

Pilon-Thomas, Shari 2511<br />

Pilotti, Silvana 10026<br />

Pilotto, Sara 630<br />

Pilz, Korinna 3018^<br />

Pimentel, Richard 9561<br />

Pin, Elisa e14151<br />

Pina, Filomena e14673<br />

Pinato, David James 2608,<br />

3022, e13033<br />

Pinchev, Mila 1578<br />

Pinder, Mary Colleen 7065,<br />

e17559<br />

Pinder, Sarah TPS665<br />

Pineda, Estela 3536<br />

Pineda, Rosa 1120<br />

Pingpank, James F. e14184<br />

Pinhal, Maria Aparecida Silva<br />

10568, e13016, e21151<br />

Pinheiro, Laura C 6001,<br />

6101<br />

Pinho, Marco C 2009, 2012,<br />

2025, 2059, 3036<br />

Pinilla-Ibarz, Javier 6503,<br />

6568, 6575, 6588, 6605,<br />

6608<br />

Pink, Daniel 10032^<br />

Pink, Desmond B. e21090<br />

Pinker-Domenig, Katja 10570<br />

Pinna, Antonio Daniele 4110<br />

Pinney, Emmett e21051<br />

Pino, Maria Simona e14661<br />

Pinotti, Graziella LBA4001<br />

Pinski, Jacek K. e15153<br />

Pinson, Stephane 10049<br />

Pinter, Matthias e14681<br />

Pinter-Brown, Lauren C.<br />

8028, 8060<br />

Pintilie, Melania 5105, 9131,<br />

e14535<br />

Pinto Marin, Alvaro TPS4672<br />

Pinto, Antonio 8066, 8083<br />

Pinto, Carmine e14040,<br />

e14042, e14113, e18021,<br />

e19592<br />

Pinto, Flavio Augusto Ismael<br />

643, e14680, e18078<br />

Pinto, Joseph A. 1024, 2097<br />

Pinto, Joseph e11066,<br />

e11518<br />

Pinto, Navin R. 9516<br />

Pinto, Peter A. TPS4680,<br />

TPS4701<br />

Pinton, Sandra 4661<br />

Piperdi, Bilal e18113<br />

Piperno-Neumann, Sophie<br />

8532, 8546, 10005,<br />

10006, 10023, 10056<br />

Piperopoulou, Fani<br />

Athanasiadou e20002<br />

Pippen, John E. 547<br />

Piras, Alessandra 9006<br />

Pircher, Tony J 584,<br />

TPS10631<br />

Pires Da Silva, Ines Esteves<br />

Domingues 8571<br />

Pirker, Robert 7007<br />

Pirl, William F. 6004, 6048,<br />

6062, 6076, 6106, 9016,<br />

9030, 9101<br />

Piro, Eugenio e18564<br />

Pirolli, Melissa 1581<br />

Pirollo, Melanie 6021<br />

Pirooz, Nicholas 8010<br />

Pirron, Ulrich TPS6643^<br />

Pirtoli, Luigi e13098<br />

Pirvulescu, Cristina 541<br />

Pirzkall, Andrea 2567, 2568,<br />

5575, e13048<br />

Pisa, Federica Edith 3612<br />

Pisanello, Anna 2057<br />

Pisano, Carmela LBA5003<br />

Pisansky, Thomas Michael<br />

4595<br />

Piscopo, Giovanna e11052,<br />

e13539<br />

Pishvaian, Michael J. 2590,<br />

3095, TPS3638, e14009,<br />

e14569, e14658<br />

Pisle, Stephen T 4671<br />

Pistamaltzian, Nikolaos<br />

Fragkiskos e11519,<br />

e15125<br />

Pistelli, Mirco e14663<br />

Pisters, Katherine 9072,<br />

e18018<br />

Pisters, Louis L. 9029<br />

Pisters, Peter W. T. 10088<br />

Pistola, Lorenza e18101,<br />

e21094<br />

Pitcher, Brandy 6015<br />

Pitini, Vincenzo e18039<br />

Pitot, Henry Clement 2510<br />

Pitou, Celine 3001<br />

Pitsiava, Dimitra e14567<br />

Pitt, Michael 4555<br />

Pittaluga, Stefania 8051<br />

Pittman, Kenneth B. 7512,<br />

e17544, e19513<br />

Pitz, Marshall W. 2051,<br />

e17518<br />

Piulats Rodriguez, Jose Maria<br />

LBA4518<br />

Piva, Eleonora 627<br />

Pivot, Xavier B. 568^, 597^,<br />

1567, 1568<br />

Piwnica-Worms, David 503<br />

773s


Piwnica-Worms, Helen 3047<br />

Pizzo, Fabrizio TPS9155<br />

Pizzolitto, Stefano 3612<br />

Plakovic, Mary K. 9094<br />

Plamondon, Louis 1055<br />

Plana, Maria e18030<br />

Planchard, David 7531,<br />

7557, 10556<br />

Plancher, Corine 8546,<br />

e15535<br />

Planci, Kezban Nur e14698<br />

Plante, Marie 5001<br />

Plastino, Francesca e14156<br />

Platero, Suso J. e17541<br />

Platica, Ovidiu e12016<br />

Platt, Jonathan J e14054<br />

Platten, Michael 2000<br />

Platz, Timothy Alan e14712<br />

Platzek, Ivan 10068<br />

Plaza, Carlos 4040<br />

Plazaola, Arrate 1001,<br />

10500, 10512, 10595,<br />

10616<br />

Plescia, Janet e13504<br />

Plesner, Torben 8019<br />

Pless, Miklos e18012<br />

Plessis, Virginie TPS9154<br />

Plestina, Stjepko 9097<br />

Pleyer, Uwe 2054<br />

Plimack, Elizabeth R.<br />

TPS2618, 4526, TPS4679,<br />

e19580<br />

Ploeen, John e14134<br />

Ploquin, Anne 2540<br />

Plotkin, Scott Randall 6136<br />

Plotti, Francesco 5093,<br />

5094, e15547, e15550,<br />

e15551, e15553<br />

Pluard, Timothy J. 534, 3047<br />

Plummer, John 2604,<br />

TPS9153<br />

Plummer, Ruth 2552, 3000,<br />

3048, 3051, 3052, 3100<br />

Plunkett, William 6528<br />

Pluschnig, Ursula 10570<br />

Pluzanski, Adam e12011<br />

Poch, Michael Adam e15203,<br />

e15204<br />

Pochettino, Paolo 10055<br />

Podda, Maurizio 8505<br />

Poddubnaya, I. e20501<br />

Poddubnaya, Irina e11528<br />

Podolski, Paula e21126<br />

Podratz, Karl C. 5004<br />

Poeschel, Viola 8022, 8024,<br />

8025<br />

Poestlberger, Sabine 514<br />

Pogge von Strandmann, Elke<br />

8079<br />

Poggi, Rosalba 2057, 2061<br />

Pognan, Francois 9562<br />

Pogoda, Katarzyna e11553<br />

Pohl, Gerhardt 7101<br />

Pohlman, Brad L. 8055,<br />

8081<br />

Poillucci, Victoria 2074^<br />

Pointreau, Yoann 5505<br />

Poire, Xavier 6582<br />

Poiree, Maryline e20006<br />

Poissonnet, Gilles 5521,<br />

e16044, e16055<br />

Poklepovic, Andrew Stewart<br />

e21058<br />

Poku, K<strong>of</strong>i 639<br />

Polascik, Thomas 4670<br />

Polaskova, Pavlina 2009,<br />

2012, 2025<br />

Polcari, Anthony e16544<br />

Polders, Larissa 8532<br />

Poli, Rossana 4097<br />

Polik<strong>of</strong>f, Jonathan TPS657,<br />

LBA1000, 4550<br />

Polite, Blase N. 2561, 6137,<br />

e16509<br />

Polivka, Marc 2023<br />

Polivka, Valentine 10027,<br />

e16036<br />

Pollack, Jonathan e14037<br />

Pollak, Michael N. 519, 538,<br />

561, e15118<br />

Pollard, Stephanie e15054<br />

Pollett, Aaron 5026<br />

Polli, Anna 7533, 7599,<br />

7600<br />

Pollmanns, Anke 552<br />

Pollock, Brad H. e16566<br />

Pollock, Raphael E. 10000<br />

Polo, Valentina 7082<br />

Polsky, David 8574, e19003<br />

Polterauer, Stephan 5111<br />

Polycarpe, Emmanuel<br />

Mohandas e19500<br />

Polychronis, Athanasios 583<br />

Polychronopoulou, Sophia<br />

e20002<br />

Polyzos, Aristidis 7564<br />

Polyzos, Nikolaos P. 5066<br />

Poma, Allen 9077<br />

Pomel, Christophe 568^<br />

Pomerantz, Mark M 4543,<br />

4635<br />

Pommier, Pascal e16036<br />

Pommier, Yves 3031<br />

Pompei, Luciano e12514<br />

Pompili, Alfredo 2085<br />

Pompili, Cecilia e21070<br />

Pompilus, Farrah e12515<br />

Pon, Vickie e15154<br />

Ponce, Mario e18109<br />

Pond, Gregory Russell 4522,<br />

4644, 5587, 9021,<br />

e21083<br />

Pondenis, Holly e21145<br />

Poniewierski, Marek S. 6129,<br />

9135<br />

Ponniah, Sathibalan 625,<br />

2508, 2529<br />

Pons, Francesc e15007<br />

Pontes Valero, Maria Jose<br />

10013<br />

Ponti, Antonio 4110<br />

Ponto, Sarah Nicks 6020<br />

Ponzone, Riccardo 627<br />

Pook, David William e15056<br />

Pool, Mark 7067, e18117<br />

Poole, Christopher John<br />

TPS654^, TPS665, 5001,<br />

5046, e11062<br />

Poole, Karen 6091, e16007<br />

Poon, Darren M. C. e15110<br />

Poon, Donald 6100, 10051,<br />

e20509<br />

Poon, Jensen T.C. e14032<br />

Poon, LM 6540<br />

Poon, Patricia 5511<br />

Poon, Ronnie Tung Ping<br />

e14545<br />

Poon, Ronnie Tung Ping<br />

4103, TPS4147, TPS4149<br />

Poon, Ronnie Tung-Ping 4108<br />

Poonamallee, Uday e21026<br />

Poortmans, Philip M. 8039<br />

Pop, Marius e15535<br />

Pop, Simona e13017<br />

Popa, Mihaela A. 2528<br />

Popa, Xitlally 2527<br />

Popalzai, Muhammad Jawad<br />

e14704, e18574<br />

Popat, Sanjaykumar 7081<br />

Popat, Uday R. 6540, 6546,<br />

8040, 8102<br />

Pope, Janice 549^, 582<br />

Pope, Lorna 3022, 10525<br />

Popiel, Brenten e14012<br />

Poplawska, Lidia e18512<br />

Popovic, Kosta e19062<br />

Popovici, Vlad Calin 3509,<br />

3511, 3575<br />

Popplewell, Leslie 6545,<br />

6582<br />

Porat, Yaara e14616<br />

Porcu, Luca 4629<br />

Porcu, Pierluigi e18508<br />

Porfiri, Emilio e15091<br />

Porneuf, Marc e14148<br />

Porntepkasemson, Apirudee<br />

e19608<br />

Porrata, Luis F. 8600<br />

Porrati, Paola 2083<br />

Porro, Maria Grazia 2543<br />

Porsok, Stefan e19020<br />

Porta, Camillo 4006,<br />

CRA4502, 4541, 4627,<br />

TPS4683<br />

Porta, Rut 7542, e18137,<br />

e18143, e19542<br />

Portales, Fabienne 3633<br />

Portell, Craig Anthony 8071<br />

Porter, Aaron T. 6034<br />

Porter, Christopher R. 4669,<br />

e15207<br />

Porter, David L. e18540<br />

Porter, Jennifer 7525, 7588<br />

Porter, Ryan Frederick 7032,<br />

7107, 7108<br />

Portera, Chia C. 3543, 3552<br />

Portier, Bryce P. 620<br />

Portnow, Jana 3108<br />

Portuesi, Rosa Alba Valentina<br />

5094, e15550, e15553<br />

Portwine, Carol 9556<br />

Posada, Juan G. 10037<br />

Posadas, Edwin M. 4586,<br />

4654<br />

Posner, John 3100<br />

Posner, Marshall R. 5501,<br />

5582, e16060<br />

Posner, Mitchell 10090<br />

Possinger, Kurt e14140<br />

Post, Sean M. 10506<br />

Postelnick, Michael J 2532<br />

Postigo, Antonio e21035<br />

Poston, Graeme John 3508,<br />

3613<br />

Postorino, Maurizio e18564<br />

Postow, Michael Andrew<br />

2518, 8515, 8549, 8575<br />

Potenza, Enrico e18026<br />

Pothuri, Bhavana 5016<br />

Potkul, Ronald e13121<br />

Potler, Hannah 1026<br />

Potosky, Arnold L. 4651,<br />

6006, 9130<br />

Pott, Dirk 4065<br />

Potter, Beth e21066<br />

Potter, Kristin 9571<br />

Potter, Nicholas 3580<br />

Potter, Vanessa 7013, 7583<br />

Potts, James 9529<br />

Potvin, Kylea R. 1036<br />

Potz, Brittany 557<br />

Pouessel, Damien LBA4567^<br />

Poulot, Virginie e18075,<br />

e18089<br />

Poupon, Marie-France e15161<br />

Pourtsidis, Apostolos e20002<br />

Poveda, Andres LBA5002^,<br />

5068, 10079, e15575<br />

Powazniak, Yanina e14109<br />

Powderly, John 4505<br />

Powell, Alex 10067<br />

Powell, Arfon e14110<br />

Powell, Bayard L. 6524^,<br />

6526<br />

Powell, C. Bethan 5022<br />

Powell, Eric 4041<br />

Powell, Erin Diana e14073<br />

Powell, Isaac 1560<br />

Powell, Matthew A. 5013,<br />

5101<br />

Power, Derek Gerard 1559,<br />

2062, e14666, e14687,<br />

e15078, e16506, e19059<br />

Power, Derek G e11569<br />

Powers, David e16513<br />

Powers, Jacquelyn 1506<br />

Powers, Jay P e13580<br />

Powers, Jean 7093<br />

Powers, John Joseph 6575<br />

Powers, Penny 10509<br />

Powles, Thomas 4529, 4611<br />

Pozdnyakova, Victoria e19049<br />

Pozo-Kaderman, Christina<br />

e19646<br />

Prabhash, Kumar 5585,<br />

e13068, e13127, e16021,<br />

e16028, e17500<br />

Prabhu Das, Irene 6046<br />

Prabhu, Aruna Laxman 1108,<br />

e11552<br />

Prabhu, Roshan Sudhir 2047<br />

Prabhu, Sujit S. 2019<br />

Prabhudesai, Shilpa A.<br />

e11043, e12030<br />

Prachand, Vivek N. 10090<br />

Pracht, Marc e19037<br />

Prado, Yolanda e11042<br />

Prados, Michael 2026<br />

Prady, Catherine 1036<br />

Praet, Michel TPS4140<br />

Prager, Gerald e15083<br />

Prakash, Deepa Meda e21132<br />

Prakash, Gaurav e18518<br />

Pramana, Setia 4561<br />

Prashanth, Gothlu<br />

Ramachandra e14675,<br />

e14721<br />

Prashanth, Jayanna e11511<br />

Prasnikar, Nicole 4083, 4090<br />

Prat, Aleix TPS656, 1008,<br />

1524, 10500<br />

Prati, Veronica e15073,<br />

e15133<br />

Pratschke, Johann e14719<br />

Prausová, Jana 3505<br />

Prayer, Daniela 2053<br />

Prayson, Richard 2014<br />

Prearo, Fernando Mauro Lima<br />

e19537<br />

Prébet, Thomas 6523<br />

Precivale, Maristela 7506<br />

Preet, Mohan e15097,<br />

e18504<br />

Prendergast, Brendan M<br />

e14555<br />

Prentice, Ross L 1501<br />

Presant, Cary A. e11562,<br />

e13003<br />

Prescott, Stephen 4<br />

Presley, Carolyn J. 1030<br />

Presley, Chris e17529<br />

Press, Michael F. 4010<br />

Press, Oliver W. 8001<br />

Pressler, Heather M 4671<br />

Preston, Megan e15585<br />

774s Numerals refer to abstract number


Pretet, Jean-Luc 10507<br />

Preusser, Matthias 2024,<br />

2053, 2071, e21033<br />

Price Hiller, Julie A. e14073<br />

Price, Alicia e21045<br />

Price, Allan 7081<br />

Price, Douglas K. 3043<br />

Price, Gregory 7101<br />

Price, Karen N. 504, 9022<br />

Price, Samantha e18509<br />

Price, Timothy Jay 3504,<br />

3515, 3534, 3535, 3572,<br />

3578, e13007, e19513<br />

Priebe, Anna M. e15568<br />

Priebe, Waldemar e18557<br />

Priedane, Eugenia e14061<br />

Priego, Victor M. 1018<br />

Priest, John R. 9521, 9522<br />

Prieto, Veronica E e14734<br />

Prieto, Victor 8537<br />

Prigerson, Holly Gwen 6012,<br />

9015, 9037, 9102, 9146,<br />

e16500<br />

Prim, Nathalie 10507<br />

Primrose, John Neil 3508<br />

Prince, Jenny 3022<br />

Prince, Mark E e16014<br />

Princet, Isabelle e19555<br />

Prinsen, Hetty 9070<br />

Prinzler, Judith 523, 1023<br />

Prior, Celia e15041<br />

Prior, John 10033<br />

Priou, Franck LBA4567^,<br />

4583<br />

Pritchard, Kathleen I. 500,<br />

512, 524, 538, 539, 540,<br />

551, 559, 561, 596, 604,<br />

6049, e19545<br />

Pritchard-Jones, Kathy 9510<br />

Privett, Melissa C. 5014<br />

Probst, Stephan 4083, 4090<br />

Procopio, Giuseppe 4627,<br />

4629<br />

Proctor, Janie e15054<br />

Proia, David A 3090<br />

Prokai, Laszlo e13557<br />

Pronzato, Paolo 579, 7577,<br />

e21065<br />

Proost, Johannes H 4528<br />

Propert-Lewis, Cerys 10039<br />

Prost, Jean-Francois 5069<br />

Protheroe, Andrew 4599<br />

Protsenko, Svetlana e13059<br />

Protter, Andrew A 564, 3072<br />

Proux, Laetitia 3547<br />

Provan, Pamela 550<br />

Provencher, Louise 634,<br />

LBA1000, 1025, 9138<br />

Provencio, Mariano 1531,<br />

6633, 7540, 8062,<br />

e11035, e11527, e12015,<br />

e17503, e18011, e18521<br />

Provencio-Pulla, Mariano<br />

e17502, e17515<br />

Proverbs-Singh, Tracey<br />

e19621, e21016<br />

Prudkin, Ludmila 7041<br />

Prudkin, Ludmilla 509<br />

Pruneri, Giancarlo 519, 546<br />

Pruvot, Francois-Rene 3506<br />

Pryma, Daniel A. e13592<br />

Przybyl, Joanna 9536, 10538<br />

Przybylo, Jennifer A. e15022<br />

Pshak, Thomas J. e15176<br />

Psyrri, Amanda 573, 5574,<br />

5576<br />

Ptaszynski, Konrad 10538<br />

Pu, Xia 7608<br />

Numerals refer to abstract number<br />

Puccetti, Maurizio 10601,<br />

e18045<br />

Puchades Roman, Iciar<br />

e14097<br />

Puchalski, Thomas 2583,<br />

3059<br />

Puchtler, Gerhard 3588<br />

Puduvalli, Vinay K. 2003,<br />

2022, 2029^, 2030, 2036,<br />

2064<br />

Puente, Javier TPS4674,<br />

10564, e15041<br />

Puertolas, Teresa 10512,<br />

10595<br />

Puggina, Anna e18010<br />

Puglisi, Fabio 1044, e13058,<br />

e13070, e15091<br />

Puglisi, Martina 3004<br />

Puhalla, Shannon 1038,<br />

3049<br />

Puhlmann, Markus 4606,<br />

5038<br />

Pujade-Lauraine, Eric<br />

LBA5000, LBA5002^,<br />

5018, TPS5112, 9079<br />

Pujol, Eduardo e18015<br />

Pujol, Jean-Louis 7013,<br />

LBA7507<br />

Pulido, Marina e14023<br />

Pulignano, Giovanni 645<br />

Pulitzer, Melissa 8515<br />

Pulivarthi, Kalyan 9127,<br />

e19599, e19643<br />

Pulvirenti, Alfredo 1007<br />

Puma, Francesco e21094<br />

Pun, Yat Wa 7011<br />

Pundalika, Mohan e19619<br />

Punglia, Rinaa S. 6022<br />

Pungliya, Manish e18041<br />

Punnoose, Elizabeth e21122<br />

Punt, Cornelis J. A. e13111<br />

Puntoni, Matteo 519<br />

Pupic, Gordana e21136<br />

Pupo, Alexandra e14673<br />

Purcell, Fay M. e14582<br />

Puri, Tarun 2072<br />

Purim, Ofer e14067, e14571<br />

Purohit, Om Pra-Kash 511<br />

Purvis, Joseph D. TPS4134<br />

Pushpalatha,<br />

Kameshwarachari e15518<br />

Pusic, Andrea 9042, 9043<br />

Pustowka, Annette 10031<br />

Pusztai, Lajos 515, 545, 581,<br />

612, 1012, 1047, 10559,<br />

10613<br />

Putnam, Joe B. 4062, 7008<br />

Putt, Mary e18507<br />

Puttabasavaiah, Suneetha<br />

1557<br />

Puttawibul, Puttisak 551,<br />

e14639<br />

Putter, Hein 516, 517, 526<br />

Puyraveau, Marc 7057<br />

Puzanov, Igor 3102, 4117,<br />

4506, 4545, 8501, 8510,<br />

8519<br />

Pyle, Doug e16505<br />

Pyman, Jan 5073<br />

Pyo, Hongryull 7004<br />

Pytel, Peter 9534<br />

Pytynia, Kristen B. 5528<br />

Pöttler, Marina e15083<br />

Q<br />

Qaqish, Bahjat F. 6093<br />

Qazilbash, Muzaffar H. 6546<br />

Qazilbash, Muzaffar 8040,<br />

8102<br />

Qi, Huanpeng e13524<br />

Qi, Jing 2541, e13064<br />

Qi, Lin 4628<br />

Qi, Yingwei 7555, 7605,<br />

e13052<br />

Qi, Yuan 545<br />

Qi, Zhengyun 4612<br />

Qian, Dapeng e21131<br />

Qian, Jiang 2013, 3532,<br />

7512<br />

Qian, Mark 3077<br />

Qian, Meng 8574, e19003<br />

Qian, Wendi 4121<br />

Qian, Xiaoping 4068,<br />

e14509, e21034<br />

Qian, Ximei e21161<br />

Qian, Yi e15172<br />

Qiang, Zhou e14648<br />

Qiao, Wei 2011, 3598, 9072,<br />

10021<br />

Qin, Jing e14575, e17512<br />

Qin, Li-Xuan 10002, 10003,<br />

10004<br />

Qin, Na e18142<br />

Qin, Qin 3532<br />

Qin, Rui 1557, 4047, 4099,<br />

5025, 5077, e15132<br />

Qin, Shukui TPS649,<br />

LBA4537, 7559, 8554,<br />

e18100<br />

Qin, Xinyu 4092, e14575<br />

Qin, Yangda 5558<br />

Qiu, Ji Qing 10543<br />

Qiu, Meng 4638<br />

Qiu, Miao-zhen e14655<br />

Qiu, Xiaoping 1535, 7586,<br />

10522<br />

Qiu, Xin 1535, 6005, 7586<br />

Qu, Angela Q. 4522, 4525<br />

Qu, Jinrong TPS2619<br />

Qu, Kevin Z. 8596<br />

Qu, Kevin 5510<br />

Qu, Tao e14502, e14668<br />

Qu, Xiaoyu e15207<br />

Qu, Zengqiang 4008<br />

Quaas, Alexander 2504<br />

Quadrini, Silvia e14082,<br />

e18065, e18072<br />

Quadt, Cornelia 508, e13605<br />

Quaglia, Alberto e21091<br />

Quah, Thuan Chong e20003<br />

Qualls, Clifford 1558<br />

Quan, Lan-Ping e14502<br />

Quant, Eudocia Carmen 2009<br />

Quaranta, Annamaria e11547<br />

Quaresma, Luisa e14673<br />

Quarshie, William O. 7037<br />

Qubaiah, Osama M. e17010<br />

Queenan, Maria B e19016<br />

Queirolo, Paola 8511, 8513,<br />

8517, 8529, 8573<br />

Quek, Marcus Lee e15008,<br />

e15014<br />

Quek, Richard Hong Hui<br />

1593, 8078, 10051,<br />

e18515, e18546, e20509<br />

Quellhorst, George J e21011<br />

Quenet, Francois 3633<br />

Quer, Miquel 5590, 5595<br />

Queralt, Bernardo 4079,<br />

e14120<br />

Queralt, Cristina 7540,<br />

10596<br />

Quereshy, Fayez A. e14032<br />

Quereux, Gaelle e19019<br />

Querleu, Denis 5083<br />

Quero, C. 4630<br />

Querzoli, Patrizia 10554<br />

Quesada, Jose e15036<br />

Quesenberry, Peter J.<br />

e21116, e21157<br />

Quevedo, Paloma e11517<br />

Quiao, Rui Fang e15191<br />

Quick, Donald 8073<br />

Quigley, Jane 1581<br />

Quijano, Yolanda 602, 4040<br />

Quillin, John 1563<br />

Quimbo, Ralph A. e16542<br />

Quindós Varela, Maria<br />

e15076, e15077, e15079<br />

Quinn, Cecily 1042<br />

Quinn, David I. 4, 4506,<br />

4511, 4547, 4563, 4663,<br />

10503, e13084, e13531,<br />

e15005, e15153<br />

Quinn, Gwendolyn P. e21155<br />

Quinn, Michael TPS5116<br />

Quinn, Raven 7035, 7567,<br />

7587, 10530, e21023<br />

Quinn, Ryan e13555<br />

Quinn, Ryann 8036<br />

Quinn, Stephen 2604,<br />

TPS9153<br />

Quinn, Tim e21085<br />

Quintana, Ana 5590<br />

Quintana, Maria Jesus 1120,<br />

5595<br />

Quintanal, Nelson 2515<br />

Quintanilla-Fend, Leticia<br />

8526<br />

Quintas-Cardama, Alfonso<br />

6527, 6587, 6589, 10506<br />

Quinten, Chantal 6002,<br />

6090, 7607<br />

Quintero, Guillermo e18070<br />

Quintyne, Keith Ian e16551<br />

Quiring, John e14501<br />

Quirino, Michela e16553<br />

Quiroz, Anteo 8580<br />

Quoix, Elisabeth A. TPS7111,<br />

TPS7610<br />

Quoix, Elisabeth 7057,<br />

10507<br />

Quon, Doris 3015, 10036<br />

Quon, Harry e16062<br />

Quon, Jennifer L. e15150<br />

Quon, Peter 6553<br />

Qureshi, S<strong>of</strong>ia 8102<br />

Qureshi, Zaina P. e19639<br />

Quyyumi, Arshed e21045<br />

Qvortrup, Camilla 3538,<br />

e14053<br />

R<br />

R, Nalini Kilara e11536<br />

Raab, Marc 8020<br />

Raab, Rachel Elizabeth<br />

e11550<br />

Raad, Roy e20004<br />

Raba, Katharina e21017,<br />

e21020<br />

Rabah, Raja 9578<br />

Rabascio, Cristina 2083<br />

Rabault, Bertrand e13605<br />

Rabbe, Nathalie e18075<br />

Rabbitt, Jane E 2026<br />

Rabe, Kari G. 6571<br />

Raben, Adam 5530<br />

Raben, David 7018<br />

Rabeneck, Linda e12034<br />

Raber, Martin N. 4130, 4131<br />

Rabey, Jonathan e15042<br />

Rabin, Michael S. 7553,<br />

7588, 10592, e18018<br />

Rabin, Tatiana e14143<br />

Rabinovitch, Rachel TPS657,<br />

TPS9150<br />

775s


Rabinowits, Guilherme 5501,<br />

5582, 9086<br />

Rabossi, Guillermo e14109<br />

Raby, Samuel 10530,<br />

e21023<br />

Raca, Gordana 6562<br />

Racadot, Severine e14730,<br />

e16036<br />

Rack, Brigitte Kathrin 554,<br />

1072, 1081, 1090, 1600^,<br />

1602, 10540, 10599<br />

Radebach, Katrina 5523<br />

Radecka, Barbara 505, 603<br />

Rademaker, Alfred 610,<br />

3083, 6623<br />

Rader, Janet Sue 5101<br />

Rader, Michael E. 4642,<br />

e15172<br />

Radford, John A. 8056<br />

Radhakrishnan, Deepa 1508<br />

Radhakrishnan, Kavita 6537<br />

Radheshyam, Nayak e11027<br />

Radhi, Saba e21105<br />

Radich, Jerald P. TPS6636<br />

Radinsky, Robert 3580<br />

Radjabi, Amir Reza e15576<br />

Radona, Marietta 3007<br />

Radosa, Marc P. e15566<br />

Radosavljevic, Davorin<br />

e12020<br />

Radova, Lenka 1087<br />

Radovich, Milan 7032, 7107,<br />

7108<br />

Radu, Aurelian e21138<br />

Radulovic, Sinisa e12020<br />

Radvanska, Eva 9592<br />

Radvanyi, Laszlo George<br />

8514, e19014<br />

Radwan, Rachel R e15565<br />

Radzikowska, Elzbieta 4632<br />

Rae, Frances TPS10633^<br />

Rae, James M. 525<br />

Raefsky, Eric 1086<br />

Raemaekers, John M. M.<br />

8039<br />

Raetz, Elizabeth A. 9507,<br />

CRA9508, 9543, 9548<br />

Raez, Luis E. 5500, 5503,<br />

e13032, e17564<br />

Raffel, Glen 6569<br />

Raffeld, Mark 2503^,<br />

TPS7609, e18568<br />

Rafferty, Stephen W 4671,<br />

e15167<br />

Rafferty, Teresa 535<br />

Raffy, Olivier 1574, TPS7111<br />

Rafi, Rezvan 1009<br />

Rafii, Saeed 5046, e11062<br />

Raftopoulos, Harry e19635<br />

Ragab, Anan 3050<br />

Raghav, Kanwal Pratap Singh<br />

3544<br />

Raghavan, Derek 4<br />

Raghavendra, Akshara 1124<br />

Raghavendra, Rao M. 1093,<br />

e11043, e11511, e12030<br />

Ragulin, Yury 7558<br />

Ragulin-Coyne, Elizaveta<br />

e14691<br />

Ragusa, Mark e21094,<br />

e21096<br />

Raha, Paromita 3058<br />

Rahal, Arslane Skander<br />

10044<br />

Rahal, Chahinez 10044<br />

Rahal, Chahineze 10062<br />

Rahatli, Samed e11011,<br />

e11023, e11026, e11045,<br />

e11515, e14174, e15572,<br />

e15573<br />

Rahili, Amine 5075<br />

Rahma, Osama E. 2577,<br />

2578<br />

Rahman, Mahbubur e14008<br />

Rahnenfuehrer, Joerg 10551<br />

Rai, Hema e21114<br />

Rai, Shesh e16545, e21144<br />

Railey, Elda 6053<br />

Raimondo, Luca 5546, 5548<br />

Raimundo, Karina 6583<br />

Raina, Vinod 6593, e11013,<br />

e17505, e17511, e18518,<br />

e18573<br />

Raines, Ronald T. TPS3113<br />

Rainfray, Muriel 9012<br />

Raisch, Dennis W 2532,<br />

4063<br />

Raiti, Concetta e13019<br />

Raizer, Jeffrey J. 2006, 2035<br />

Raj, Ganesh 4616, TPS4678<br />

Raj, Renju V. e16503<br />

Raja, Madras A. e20508<br />

Raja, Rajiv 5575<br />

Rajagopalan, Prabhu 3012,<br />

3019, 3046, 4050, 4103<br />

Rajan, Arun 7033, 7103,<br />

TPS7609<br />

Rajan, Sabrina 544<br />

Rajan, Sudhan e19619<br />

Rajangam, Kanya 8035<br />

Rajapaksa, Ranjani 2513<br />

Rajaram, Veena e20001<br />

Rajdev, Lakshmi e14019<br />

Rajec, Jan TPS4677, e15028<br />

Rajguru, Saurabh e14554<br />

Rajkovic, Aleksandar 1501<br />

Rajkumar, Krishnappa<br />

e14675<br />

Rajkumar, S. Vincent<br />

TPS8116<br />

Rajkumar, Vincent 8096,<br />

8105<br />

Rajpar, Laetitia 10556<br />

Raju, Robert N. TPS3111<br />

Raju, Savitha e15565<br />

Rajurkar, Swapnil Padmakar<br />

e11562, e13003<br />

Raker, Richard K e13066<br />

Rakovitch, Eileen 1123<br />

Rakus, John Frank e19018<br />

Rakyan, Vardhman 10550<br />

Ram, Prahlad T 8530<br />

Ramacci, Carole TPS10634<br />

Ramadori, Giuliano e13039,<br />

e14707, e21128<br />

Ramaekers, Ryan C. 9110<br />

Ramaiya, Nikhil H. 7553<br />

Ramakant, Pooja e19562<br />

Ramakrishna, Satvik 7054<br />

Ramalingam, Ashok TPS9150<br />

Ramalingam, Lekshmi<br />

e14184<br />

Ramalingam, Suresh S. 2576,<br />

3049, 3094, 7048, 7059,<br />

7512, 7516, TPS7613^,<br />

TPS7615, 10625, e21045<br />

Ramalingam, Suresh S 5560,<br />

5570, 7097<br />

Ramamoorthy, Sonia e14069<br />

Ramamurty, S e13522<br />

Raman, Siva P 4045<br />

Ramanathan, Michelle Leana<br />

e14092<br />

Ramanathan, Muthalagu 6569<br />

Ramanathan, Ramesh K.<br />

2579, 3019, 3033, 3037,<br />

3060, 4013, 4019,<br />

e13576<br />

Ramani, Rupal 9034, 9109,<br />

9123<br />

Ramchandren, Rod 8028<br />

Ramée, Jean François<br />

LBA3500^, e14148<br />

Ramfidis, Vassilios e17557<br />

Ramies, David A. 535, 4007,<br />

4504, 5547, 7514, 8531<br />

Ramirez Morales, Rafael<br />

e14563<br />

Ramirez, Amelie G. e16566<br />

Ramirez, Arturo e21148<br />

Ramirez, Jacqueline 2611<br />

Ramirez, Jose Luis 10596<br />

Ramirez, Mildred M. 6116<br />

Ramirez, Nicolas e11565<br />

Ramirez, Robert A. 7072^,<br />

e17531^<br />

Ramirez-Fort, Marigdalia K.<br />

e19624<br />

Ramirez-Morales, Rebeca<br />

e12027<br />

Ramlau, Rodryg 3579,<br />

LBA7000, 7090, 9117<br />

Ramnath, Nithya 2602,<br />

e17535, e18118<br />

Ramnauth, Hemwattie<br />

e19004<br />

Ramon y Cajal, Santiago 566,<br />

3014, 7041<br />

Ramon y Cajal, Teresa 1120,<br />

10555, e18057<br />

Ramondetta, Lois M. 9134<br />

Ramondino, Stefano 5096<br />

Ramos Vazquez, Manuel<br />

10616, e15067, e15076,<br />

e15077<br />

Ramos, Alexis 10502<br />

Ramos, Carlos Almeida 2558<br />

Ramos, Clarissa CA e15562<br />

Ramos, Rafael 10079<br />

Ramos, Teresa e14147<br />

Ramos-Lamboy, Valmarie<br />

e11015<br />

Rampello, Elvira e13019<br />

Rampersaud, Edward N.<br />

e13045<br />

Rampias, Theodoros 5574<br />

Ramsey, Sara 3041, 3097<br />

Ramsey, Scott David 6007,<br />

6077, e14068, e14141,<br />

e14146, e15175, e15183,<br />

e16524, e18097<br />

Ramsey, William Jay 2571,<br />

e19008<br />

Ramskold, Daniel 10539<br />

Rana, Durgesh N e15011<br />

Rana, Vishal 8096<br />

Ranasinghe, Weranja Kalana<br />

Bodhisiri e15117<br />

Ranchicchio, Prabha e18074<br />

Randolph, Sophia 8520<br />

Rane, R. C. 3089^<br />

Ranganathan, Anjana 7077,<br />

7092<br />

Ranganathan, Gayatri 6553<br />

Ranganathan, Sulabha 531<br />

Rangel, Claudia e18028<br />

Ranger-Moore, Jim 4041<br />

Rani, Sweta 617<br />

Ranieri, Girolamo e21113,<br />

e21134<br />

Ranki, Annamari 8076<br />

Ranki, Tuuli e13035<br />

Rankin, Cathryn J 4113<br />

Ransom, David e13007,<br />

e16004<br />

Ranson, Malcolm 3000,<br />

3004, 3030, 3048, 3051<br />

Rao, Archana C. e11089<br />

Rao, Chandra 7092, 10533<br />

Rao, Ganesh 2019<br />

Rao, Gautam G. e16546<br />

Rao, Manoj e15014<br />

Rao, Nalini K 1093, e11027,<br />

e11511, e12030<br />

Rao, Nikhil G 5564<br />

Rao, Rekha 10549, e13568<br />

Rao, Shantha 512<br />

Rao, Shyam S. 5537, 5545^<br />

Raoelfils, Ines 543<br />

Raoul, Jean Luc LBA4003<br />

Raoul, Jean-Luc 4118<br />

Rapalino, Otto 2059<br />

Rapetti, Simonetta e18103<br />

Rapisardi, Stefania e15544<br />

Rapke, Tal e15152<br />

Raponi, Mitch 4041<br />

Rapoport, Aaron e19565<br />

Rappold, Erica 596<br />

Raptis, Anastasios 6563<br />

Raptis, George e21032<br />

Rasalan, Teresa 2518<br />

Rasche, Herbert 6610<br />

Rasco, Drew Warren 2512,<br />

3001, 3019, TPS3110,<br />

e13025, e13602, e18076<br />

Rashid, Asif e14662<br />

Rashtian, Afshin TPS9150<br />

Rasinski, Kenneth 9586<br />

Rask, Sara e14001<br />

Raskin, Leon e12026<br />

Rasmussan, Karen 3630<br />

Rasmussen, Erik 5072<br />

Raso, Maria G 7023<br />

Raspagliesi, Francesco<br />

LBA5003, 5096<br />

Rastelli, Francesca 9098<br />

Rastogi, Priya LBA506,<br />

LBA1000, 1025, TPS1142<br />

Rastogi, Shishir 9580<br />

Ratain, Mark J. 2533, 2611,<br />

6117, e13106<br />

Ratanatharathorn, Voravit<br />

6549<br />

Rath, G K e15518<br />

Rath, Goura Kishore e11082<br />

Rath, Goura Kishor 2072<br />

Rath, Michelle Genevieve<br />

1547<br />

Rath, Niharika e14632<br />

Rathbone, Dustin 10002<br />

Rathkopf, Dana E. 4513,<br />

LBA4518, 4548, TPS4697<br />

Rathmell, Kimryn 4617,<br />

TPS4686<br />

Rathore, Bharti e13124<br />

Rathore, Ritesh e13124<br />

Ratner, Elena 5099, e13125,<br />

e15526, e15581<br />

Ratner, Lee 4030, 8005<br />

Ratti, Margherita 4097<br />

Rau, Kun-Ming 4103<br />

Raubitschek, Andrew Antony<br />

639, 6545<br />

Rauch, Michael 2007<br />

Rauch, Philippe 10517<br />

Rauckhorst, Myrna 2526,<br />

TPS4701<br />

Rauh, Jacqueline e14138<br />

Raunig, David e13082<br />

Rauscher, Garth e16531<br />

776s Numerals refer to abstract number


Raut, Chandrajit P. 1547,<br />

5090, 10017<br />

Rauthan, Amit e14710,<br />

e15516<br />

Rauw, Jennifer 2572<br />

Ravaggi, Antonella e15549<br />

Ravandi, Farhad 6501, 6528,<br />

6544, 6558, 6576, 6587,<br />

6589, 6592, 6597, 6603,<br />

6607, TPS6637<br />

Ravaud, Alain 4583, 4606,<br />

9012<br />

Ravdin, Peter e14684<br />

Rave-Fraenk, Margret e13039,<br />

e21128<br />

Ravel, Patrice e13578<br />

Ravi, Vinod 10071, e19520<br />

Ravic, Miroslav 8079<br />

Ravindranath, Yaddanapudi<br />

9578<br />

Ravn, Jesper 7001<br />

Rawal, Sudhir e15101<br />

Rawl, Susan e16557<br />

Ray Coquard, Isabelle 636,<br />

10062<br />

Ray, Anna M. e15205<br />

Ray, Saurabh 570, e12515,<br />

e13038, e16571<br />

Ray, Ujjal Kanti e13030<br />

Ray, Vani e19030<br />

Ray-Coquard, Isabelle Laure<br />

LBA5000, 10027, 10095<br />

Ray-Coquard, Isabelle 1095,<br />

2583, LBA5002^, 5018,<br />

5067, 10005, 10006,<br />

10007, 10010, 10014,<br />

10020, 10056, 10092,<br />

10562<br />

Raymond, Eric 2554, 2557,<br />

4118, e13594, e14553,<br />

e14660<br />

Raynaud, Jean-Pierre e15145<br />

Raynov, Julian e14501<br />

Rayson, Daniel TPS658,<br />

1036<br />

Raza, Azra 3081, 6624<br />

Razak, Albiruni R.A. 3082,<br />

3101, e13001<br />

Razak, Albiruni e19616<br />

Razavi, Pedram 1556<br />

Razis, Evangelia 573, e15040<br />

Razzaque, Saqib e14715<br />

Razzini, Giorgia e11071<br />

Razzouk-Cadet, Micheline<br />

8077, e15574<br />

Re, Alessandro 8046, 8058<br />

Rea, Daniel 516, 517, 530,<br />

TPS665, e11062<br />

Rea, Delphine 6503<br />

Rea, Domenica e11052,<br />

e13539<br />

Rea, Silvio e11041<br />

Read, Nancy E 5530<br />

Read, William L. 3055<br />

Ready, Neal e13032, e17558<br />

Reagan, John Leonard<br />

e21116<br />

Real, Francisco X e12033<br />

Reaman, Gregory H. 9543,<br />

9587<br />

Reamer, Margaret 7071<br />

Reames, Bradley Norman<br />

6020<br />

Reardon, David A. 2027,<br />

2090^, 2094^, 2095^,<br />

TPS2103<br />

Reber, Howard A. e14582<br />

Rebersek, Martina e14083<br />

Numerals refer to abstract number<br />

Rebollo, Joseba 3064,<br />

e16000<br />

Reboredo, Margarita e14595,<br />

e21002<br />

Rebours, Vinciane 4133<br />

Reburiano, Eunicia 8089<br />

Recchia, Francesco e11041<br />

Reche, Encarni 10082<br />

Rechis, Ruth 9136, e19503<br />

Recht, Abram 6022, 7040,<br />

7054, 7055<br />

Recinos, Violette 2076, 2100<br />

Recio, Manuel 1111<br />

Reck, Martin 2573^, 7013,<br />

7030, TPS7113, LBA7507,<br />

7536, TPS7611, e13113<br />

Reckamp, Karen L. 3023,<br />

3108, 7518, 9068<br />

Reckova, Maria TPS4677,<br />

e15028<br />

Reddick, Cathy TPS1148,<br />

TPS4147<br />

Reddy Goteke, Vamshi<br />

Krishna 6554, e18544<br />

Reddy, Akhila Sunkepally<br />

9002, e19599<br />

Reddy, Indraneel e11097,<br />

e12004<br />

Reddy, Manjula 2583<br />

Reddy, Pavan S. 9000, 9142<br />

Reddy, Prem-veer e15174<br />

Reddy, Sangeetha Meda 1561<br />

Reddy, Suresh K. 9002,<br />

e19528, e19584<br />

Reddy, Vijay Anand Palkonda<br />

9568<br />

Reddy, Virgil 6622<br />

Redfern, Charles H. TPS4697<br />

Redman, Bruce G. 8504,<br />

8525<br />

Redman, Bruce TPS2613,<br />

4546, 8587<br />

Redman, Mary Weber 7517,<br />

7552, 7570<br />

Redner, Robert 6563<br />

Redon, Christophe E. 2553,<br />

3031<br />

Redondo, Andres 10052,<br />

e13085<br />

Redondo, Maximino e14116<br />

Redwan, Bassam 10080<br />

Reece, Donna Ellen 8020,<br />

TPS8112, TPS8113<br />

Reed, Angela D. e15179<br />

Reed, Carolyn E 4062<br />

Reed, Damon R. 10003,<br />

10048, 10070, 10084<br />

Reed, Galen 4660<br />

Reed, Mark E. 5014<br />

Reed, Michael 7054, 7055,<br />

e17560<br />

Reed, Nicholas 4121, 4122,<br />

LBA5000<br />

Reed, Robert 1529<br />

Reed, Shelby D. 1525<br />

Reeder, Craig B. 8010, 8034,<br />

8043, 8053<br />

Reeder, Kimberley Alex<br />

e15549<br />

Reeder-Hayes, Katherine<br />

Elizabeth 6017, 6029,<br />

6096, 6104<br />

Reedijk, Michael 1019, 5019<br />

Reeler, Anne 580<br />

Rees, Miranda 567<br />

Rees, Robert 1013<br />

Rees, Rowland e15100<br />

Reese, Emily S e15108<br />

Reese, Jane TPS1616<br />

Reeve, Bryce B. 6002, 6107,<br />

7607, 9047, 9144,<br />

e19633<br />

Reeves, Leslie e13581<br />

Refaat, Tamer 1069<br />

Regan, David H. e19615<br />

Regan, Eileen 4027, 4125<br />

Regan, Kelly E. 9584<br />

Regan, Meredith M. 504,<br />

e15170<br />

Regazzi, Ashley Marie 4581^,<br />

e15022<br />

Rege, Jessicca TPS1145<br />

Regine, William 4041<br />

Regitnig, Peter 515<br />

Regnier-Rosencher, Elodie<br />

8591<br />

Rego, Juliana Florinda De<br />

Mendonga e12513<br />

Reguart, Noemi 7540<br />

Rehman, Farah 3050<br />

Rehrauer, William M. e13537<br />

Reich, Elsa 5078<br />

Reich, Richard 5564<br />

Reichardt, Annette 10032^<br />

Reichardt, Peter LBA10008,<br />

10010, 10032^<br />

Reiche, Jana 3519^<br />

Reichel, Carol e19510<br />

Reichle, Albrecht 6500^,<br />

6610<br />

Reid, Alison Helen 4553<br />

Reid, Gregory K. 3039<br />

Reid, Joel M. 9581, e13086<br />

Reid, Julia E. 7023<br />

Reid, Tony R. e14069,<br />

e14078, e14079<br />

Reid, Tony 4055<br />

Reidy, Diane Lauren 4047,<br />

4057, 9105, e14112<br />

Reifenberger, Guido 2000,<br />

2067<br />

Reig, Beatriu 1126<br />

Reig, Óscar 3536<br />

Reijneveld, Jaap 2071, 6090<br />

Reilly, Patrick J e13038<br />

Reimer, Daniel 5080<br />

Reimer, Toralf 5080<br />

Reina, Juan J e14671<br />

Reiner, Anne S. 2006<br />

Reiner, Johnathan e17543<br />

Reiner-Concin, Angelika<br />

e14087<br />

Reiners, Katrin S. 8079<br />

Reinert, Anne 3006^, 3058<br />

Reinhard, Todd 8528<br />

Reinhardt, Carsten 2522,<br />

3530<br />

Reinholz, Monica Madden<br />

1083<br />

Reinke, Denise K. TPS10100<br />

Reinke, Petra 8030<br />

Reinthaller, Alexander<br />

LBA5000, 5031, 5080,<br />

5111<br />

Reiriz, Andre B. 606<br />

Reis, Isildinha M. 595<br />

Reiser, Marcel 1533, 8022,<br />

8024, 8025, 8031, 10526,<br />

CRA10529<br />

Reiser, Vadim 1564<br />

Reisman, Arlene 7596<br />

Reisman, David 10522<br />

Reitter, Sonja e18525<br />

Reitz, Richard E 5510<br />

Rejeb, Narmyn e13594<br />

Rekhtman, Natasha 7505,<br />

10560<br />

Reklaitis, Gintaras V e18555<br />

Rekman, Janelle e14154<br />

Relander, Thomas 10024<br />

Rembak-Szynkiewicz, Justyna<br />

e21049<br />

Remenar, Eva 5516^<br />

Remillard, Stephen P. 10029<br />

Remy, Herve 9560<br />

Ren, Chao e14653<br />

Ren, Chen 3017, 3081<br />

Ren, Guo-xin 5593<br />

Ren, Henning 5524<br />

Ren, Li e14000<br />

Ren, Min 8594<br />

Ren, Shengxiang 7520,<br />

7535, 10527, e18123<br />

Ren, Xiu Bao LBA4537, 8554<br />

Ren, Zhenggang 4008<br />

Renard, Vincent 4622<br />

Renbarger, Jamie L. 2601<br />

Rendleman, Justin 8557<br />

Renehan, Andrew e14015<br />

Rengan, Ramesh 6070,<br />

7050, 7098, e17534,<br />

e18032<br />

Rengucci, Claudia 10601<br />

Reni, Michele 4017<br />

Renne, Salvatore Lorenzo<br />

4120<br />

Rennert, Gad 1578<br />

Rennert, Hedy S 1578<br />

Renninger, Markus e19021<br />

Renouf, Daniel John 3527,<br />

6018, e14048, e14588<br />

Renouvel, Florence e11551<br />

Renschler, Markus F. 7590<br />

Renschler, Markus Frederic<br />

7592, TPS7618<br />

Rensvold, Diane M 3028<br />

Renton, Corrinne 9021<br />

Resci, Federica e17006,<br />

e18563, e18566<br />

Rescigno, Pasquale e15034<br />

Reske, Tom e11030<br />

Resteghini, Carlo 5555<br />

Rettenmaier, Christopher<br />

e15500<br />

Rettenmaier, Nicholas<br />

e15500<br />

Retter, Avi S. 3075<br />

Rettig, Klaus 9114<br />

Rettig, Matthew 4665<br />

Retz, Margitta 4530, 4590,<br />

e15195<br />

Reu, Frederic J. 8035<br />

Reuben, James M. 594,<br />

1047, 8091, 9002, 9072,<br />

9081, 9083, e13582<br />

Reungwetwattana, Thanyanan<br />

1565<br />

Reusch, Uwe 8059, e18532<br />

Reuss, Alexander LBA5002^,<br />

5005<br />

Reutemann, Patricia e19025<br />

Reuter, Christoph W. e15190<br />

Reveiz, Ludovic e12039<br />

Revnic, Julia e19544<br />

Rey, Annie 5028, 5083,<br />

10098<br />

Rey, Augustin A. TPS5112<br />

Rey, Jean-Baptiste 1095,<br />

5067<br />

Rey, Severino 3064<br />

Reychler, Herve 5519<br />

Reyderman, Larisa 2552,<br />

e18063^<br />

Reyes, Carolina M. 6007<br />

Reyes, Jose 639<br />

Reyes, Ruben 10605,<br />

e13568<br />

777s


Reyes-Rivera, Irmarie<br />

CRA3503<br />

Reyna Asuncion, Bernadette<br />

7597<br />

Reyners, An K.L 5017^<br />

Reynes, Gaspar 2045<br />

Reyno, Leonard M. 4568<br />

Reynolds, Alison e13570<br />

Reynolds, C. Patrick 8073,<br />

e15103<br />

Reynolds, Craig H. LBA4,<br />

e18063^<br />

Reynolds, Jana e11069<br />

Reynolds, Joe TPS663<br />

Reynolds, John 6505^<br />

Reynolds, Kerry 9136<br />

Reynolds, Marleta e20001<br />

Reynoso, Jose e14700<br />

Reyt, Emile e16013<br />

Rezaei, Mansour 6556<br />

Rezai, Mahdi 554<br />

Rezvani, Katy 10550<br />

Rha, Sun Young TPS4142,<br />

e12029, e14081<br />

Rha, Sun Young 4077, 4536,<br />

4538, 4544^, TPS4683,<br />

e14580, e14652, e15064<br />

Rhee, Ina Park e13000,<br />

e13113<br />

Rhee, Jiyoung 4028, e14580<br />

Rhees, Brian e20507<br />

Rhiem, Kerstin 3044<br />

Rhoads, Kim F 6010<br />

Rhode, Peter e13088,<br />

e15010<br />

Rhodes, Dan e14164<br />

Rhondali, Wadih 9049, 9080,<br />

e19603, e19613<br />

Ribalta, Teresa 2045<br />

Ribas, Antoni 8502^, 8503^,<br />

8504, 8518, 8521, 8525,<br />

8527, 8567, TPS8603,<br />

e13045<br />

Ribeiro, Adriana Regina G.<br />

e11058, e14680, e14728<br />

Ribeiro, Heber Salvador de<br />

Castro e14728<br />

Ribeiro, Karina Braga 9523<br />

Ribeiro, Maria Leonor 6044<br />

Ribeiro, Maria-Jose 10625<br />

Ribeiro, Thiago Neto e12018<br />

Ribelles, Nuria 10500<br />

Ribera, Josep-Maria 8005,<br />

8058<br />

Ribero, Dario 4110<br />

Ribi, Karin 9022, e19648,<br />

9045, 9059<br />

Ribrag, Vincent 2539, 8057,<br />

8068<br />

Ricarte-Filho, Julio Cezar M.<br />

5509^<br />

Riccardi, Alberto 589<br />

Riccardi, Riccardo 9517<br />

Ricci, Angela M 6537<br />

Ricci, Claudio e14647<br />

Ricci, Fabio e11514,<br />

e19002, e19620<br />

Ricci, Jean-Francois 539<br />

Ricci, Marianna 10615,<br />

e18045<br />

Ricci, Rossana 10615<br />

Ricci, Sergio LBA4001<br />

Ricci, Veronica e13098<br />

Ricci, Vincenzo 3540, 3549<br />

Ricciardi, Giuseppina R. R.<br />

e11056<br />

Ricciardi, Roberto 5093,<br />

5094, e15547, e15550,<br />

e15551, e15553<br />

Riccio, Gennaro e13539<br />

Rice, Audie 8087<br />

Rice, Carol e16538<br />

Rice, David C. 4069, 4078,<br />

4086<br />

Rice, Thomas W. e14611,<br />

e14665<br />

Ricevuto, Enrico e14010,<br />

e15200<br />

Rich, Barrie S. 8586<br />

Rich, Elizabeth Shima 6552,<br />

e16503<br />

Rich, Tara e18046<br />

RiChard, Jamie e15021<br />

Richard, Marie-Aleth 8555,<br />

8568<br />

Richard, Sandrine e19640<br />

Richard, Scott Daniel e15578<br />

Richardet, Eduardo arnoldo<br />

e11501<br />

Richardet, Eduardo e11044,<br />

e18114, e18125<br />

Richardet, Martin Eduardo<br />

e11044, e11501, e18125<br />

Richards, Catherine A. 6118<br />

Richards, Donald A. LBA4,<br />

2516, TPS3111, LBA3501,<br />

9117, e14501, e18047<br />

Richards, JoAnne S e15583<br />

Richards, Jon M. 8508,<br />

e13088<br />

Richards, Kristen 8537<br />

Richards, Paul D. 1017<br />

Richardson, Andrea 576<br />

Richardson, Blakely S. 1603<br />

Richardson, Debra L. e15501<br />

Richardson, E. 561<br />

Richardson, Gary Edward<br />

e19514<br />

Richardson, Jaime 2087<br />

Richardson, Paul Gerard Guy<br />

8012, 8016, 8017, 8019,<br />

8020, 8033, 8043, 8095,<br />

TPS8112, TPS8113,<br />

e18572^<br />

Richardson, Stacie e18139<br />

Richardson, Steve 569, 571,<br />

6098, e11002<br />

Richardson, William 4041<br />

Riche, Christian e14148<br />

Richel, Dirk 4094<br />

Richer, Jennifer K. 564,<br />

TPS668<br />

Richert-Boe, Kathryn E.<br />

e12024, e15197, e18107,<br />

e19630<br />

Richhariya, Akshara e12024,<br />

e15197, e18107, e19514,<br />

e19630<br />

Richie, Jerome P. 4521<br />

Richie, Mary Ann 6558, 6585<br />

Richly, Heike 2086, e13605<br />

Richman, Joni TPS3638,<br />

e14501<br />

Richter, Barbara 5005<br />

Richter, Rolf 5031, 5034,<br />

5071, e15531, e19597<br />

Richter, Stephan 10068<br />

Richter, Sue 1555, 3082,<br />

e15017, e15020<br />

Ricigliano, Mark 3066<br />

Rick, Oliver e15026<br />

Ricke, Jens TPS4148,<br />

e14630<br />

Ricker, Charite Nicolette<br />

1124<br />

Ricker, Justin L. 3532, 7512,<br />

e13583<br />

Rickmann, Mariana e21035<br />

Ricordeau, Philippe e14065<br />

Riddell, Angela M TPS4143<br />

Rider, Alex e16570<br />

Rider, David N. 5077<br />

Ridge, John A. 10064<br />

Ridgway, Paul F 10050<br />

Ridner, Sheila H e16038,<br />

e19534, e19559<br />

Ridolfi, Kimberly 555, 563,<br />

6063<br />

Ridolfi, Ruggero 8513, 8529<br />

Ridwelski, Karsten 4072,<br />

4095^<br />

Riechelmann, Rachel Pimenta<br />

e14702<br />

Riechelmann, Rachel 6055<br />

Riedel, Richard F. 10010<br />

Rief, Winfried e11040^<br />

Rieke, Damian 5517<br />

Riely, Gregory J. 3083, 7014,<br />

7033, 7052, 7527, 7531,<br />

7532, 7533, 7580, 7599,<br />

7600<br />

Riera-Knorrenschild, Jorge<br />

e14152<br />

Riesco Martinez, Maria C.<br />

e14171<br />

Riesco, M. Carmen e14058<br />

Riess, Hanno 2554, 4020,<br />

4044, 4050, TPS4135,<br />

8030, e14533<br />

Riess, Jonathan 10574<br />

Riester, Markus 4577, 4585<br />

Riethdorf, Sabine 1090,<br />

10599<br />

Rietschel, Petra 10003<br />

Rifa, Julio e11036<br />

Rifai, Aya e17015<br />

Rifkin, Robert M. 8037,<br />

8064, e18556<br />

Riga, Anne 4109<br />

Rigacci, Luigi 8006<br />

Rigas, James R. 7022, 7541,<br />

e14514<br />

Rigatos, Sotirios 583<br />

Riggs, Heather Dawn e16557<br />

Righini, Christian e16044,<br />

e16051, e16055<br />

Rigo, Raul 3096<br />

Riisnaes, Ruth 10525,<br />

e13601<br />

Rijavec, Erika 7577, 7581,<br />

e21065<br />

Riker, Adam Irwin e19008<br />

Riley, Sarah 5501<br />

Rimassa, Lorenza 4006,<br />

e21139<br />

Rimawi, Mothaffar F. 606,<br />

TPS654^<br />

Rimely, Endre 2077<br />

Rimes, Sue A 6116<br />

Rimm, David 517, 573,<br />

1121, 5576, e21106<br />

Rimmer, Yvonne L. 4531<br />

Rimner, Andreas 7043<br />

Rimsza, Lisa M. 8001,<br />

e18534, e18536<br />

Rinaldi, Giulia e11039,<br />

e11514, e19620<br />

Rinaldi, Yves LBA3500^<br />

Rinard, Katherine e15103<br />

Rinck, Jose A. e15122,<br />

e18078<br />

Rincon, Raul 5520, e15094,<br />

e18069, e18106, e21015<br />

Rinderknecht, Jeannine<br />

Desiree e19017<br />

Ring, Andrej 10059<br />

Ringash, Jolie 5571,<br />

TPS5600, 6002, 7607<br />

Ringel, Ralf 640<br />

Ringert, Rolf-Hermann<br />

e15156, e15158<br />

Ringom, Rune e13518<br />

Rini, Brian I. TPS2614,<br />

4503, 4536, 4538, 4546,<br />

4609, TPS4679, TPS4684,<br />

e15095<br />

Rino, Yasushi 4070<br />

Rio Frio, Thomas e14059<br />

Rios, Maria 10056, 10092,<br />

10095<br />

Rios, Mary Beth 6501<br />

Rios, Paola e12039<br />

Rios, Sandra 1041<br />

Rios-Perez, Jorge Arturo<br />

1085, TPS1138<br />

Riou, Francois 9074<br />

Ripaldi, Mimmo 6603<br />

Ripamonti, Carla 9005<br />

Ripoche, Veronique 7554<br />

Rippin, Gerd 2573^<br />

Riquelme, Alejandro e21104<br />

Riquet, Marc 7024<br />

Risch, Harvey TPS1614<br />

Rischin, Danny TPS5116,<br />

5571, e13054, e15541<br />

Risendal, Matthew e18550<br />

Rishi, Arun K 9578<br />

Risk, Janet 5543<br />

Riska, Henrik 7001<br />

Riska, Shaun M e15132<br />

Riss, Joseph e13045<br />

Risse, Marie-Laure TPS4696^<br />

Ristagno, Ross e14714<br />

Risueno, Noelia e14097<br />

Ritch, Paul S. e14613<br />

Ritchie, Christine 6080<br />

Ritchie, Ellen K. TPS6637<br />

Ritenour, Chad e15006<br />

Ritgen, Matthias 6500^<br />

Ritter, Barbara 2504<br />

Ritter, Gerd e11563<br />

Rittweger, Karen 3578,<br />

TPS7616<br />

Rivarola, Edgardo G. J. 9038,<br />

e11037, e15507, e15508<br />

Rivas Ruiz, Francisco e14116<br />

Rivas, Carlos e19569<br />

Rivelli, Thomas Giollo e16568<br />

Rivera, Daniel e12027<br />

Rivera, Gabriel 8103<br />

Rivera, Lorna e16500<br />

Rivera, Ragene Ruth 547<br />

Rivera-Rodriguez, Noridza<br />

e18547<br />

Rivero, Elisabeth e11035<br />

Rivero, Gustavo e17018<br />

Riviere, Alain TPS7112, 7575<br />

Riviere, Isabelle TPS2619,<br />

TPS4700<br />

Rivoire, Michel e14730<br />

Rivoltini, Licia 5555, 8529<br />

Rix, Peter J. 4548, TPS4697<br />

Rixe, Olivier TPS2103,<br />

TPS3116, 7556, e16017<br />

Rizack, Tina 1045, 5100<br />

Rizel, Shulamith 548<br />

Rizk, Nabil P. 4061<br />

Rizvi, Naiyer A. TPS2615,<br />

TPS2621, 7014, 7052,<br />

7527, 7578<br />

Rizvi, Syed 4081<br />

Rizzi, Anna e14018<br />

Rizzi, Rita 6584<br />

Rizzieri, David 6525,<br />

TPS6636<br />

778s Numerals refer to abstract number


Rizzo, Pietro e18048<br />

Rizzo, Virginia e15010<br />

Ro, Jungsil 1003, 2542<br />

Roa, Wilson TPS2104<br />

Roach, Jim 4056<br />

Robak, Tadeusz 8018^<br />

Robb, Caroline M. e21018<br />

Robbins, Anthony 6011<br />

Robbins, Edward T e17531^<br />

Robbins, Jared R. 5088<br />

Robbins, Joan M. 2101<br />

Robbins, Samuel G e17531^<br />

Robert, Caroline 1566, 8501,<br />

8502^, LBA8509, 8512,<br />

8591, 8601, e19022<br />

Robert, Francisco 2610,<br />

3034, 7502<br />

Robert, Jacques e14023<br />

Robert, Rhonda J. 9096<br />

Roberts, Catherine H. e18550<br />

Roberts, Christopher e15209<br />

Roberts, Heidi 10522<br />

Roberts, Ian SD 4599<br />

Roberts, Kenneth B 1030,<br />

4651<br />

Roberts, Lewis R. 4033<br />

Robertson, Cary N. 4670,<br />

e15194<br />

Robertson, Chris 1509, 1510<br />

Robertson, Jane D e13541<br />

Robertson, John M. 3545<br />

Robertson, Philmore 9547,<br />

e18522<br />

Robidoux, Andre LBA506,<br />

616, LBA1000, 1025,<br />

TPS1139<br />

Robin, Marie 6534, 6542<br />

Robin, Yves Marie 10018<br />

Robinet, Gilles TPS7112,<br />

e16560<br />

Robins, Deborah A 2516<br />

Robins, Harlan 6631<br />

Robins-Browne, Kate 9124<br />

Robinson, Austin e18549<br />

Robinson, Bruce 5591<br />

Robinson, David 4668<br />

Robinson, Eliezer e16505<br />

Robinson, Giles W. 9518<br />

Robinson, Kim e21013<br />

Robinson, Lary 7024<br />

Robinson, Max 5543<br />

Robinson, Patricia A. e16531<br />

Robison, David J e18541<br />

Robison, Katina 5100<br />

Robison, Leslie L. 6030,<br />

9505, CRA9513, 9514,<br />

9526, 9528, 9531<br />

Robles Irizarry, Lizbeth 2076,<br />

2093, 2100<br />

Robles, Robert Leon TPS6636<br />

Roboz, Gail J. 6577<br />

Robson, Mark E. 10097<br />

Robson, Matthew 3088^<br />

Robson, Tammy 516, 517<br />

Roca, Enrique Luis e14521,<br />

e14598<br />

Roca, Fernanda e15570<br />

Rocca, Andrea e11054<br />

Rocchi, Palma e15155<br />

Rocco, James W. e16059<br />

Rocha Lima, Caio Max S.<br />

4049<br />

Rocha, Eduardo 7095<br />

Rocha, Rafael Malagoli 643<br />

Rochard, Sophie e14148<br />

Roche, Henri Hubert 1011,<br />

10076<br />

Roche, Henri 543<br />

Roche, Sandra 2536<br />

Numerals refer to abstract number<br />

Rochet, Nicole M. 8600<br />

Rochlitz, Christoph 9059<br />

Rock, Colleen e13047<br />

Rock, Edwin P. TPS4134<br />

Rock, Hans 2079<br />

Rocque, Gabrielle Betty 9039<br />

Roda Perez, Desamparados<br />

2567, 2568, 3021,<br />

e13048, e14589<br />

Roddie, Huw 8015<br />

Rodeghier, Mark e14697<br />

Rodell, Timothy C. TPS3638,<br />

e14501<br />

Rodenhuis, Sjoerd 10013<br />

Roder, Heinrich e18096,<br />

e18100<br />

Roder, Joanna e18096,<br />

e18100<br />

Rodgers, Kristen e18147<br />

Rodier, Jean-Francois e11542<br />

Rodig, Scott J. 3053<br />

Rodin, Gary 9003, e16567<br />

Rodkey, Travis e13516<br />

Rodler, Eve T. 10011^,<br />

TPS10103<br />

Rodolakis, Alexandros 5033<br />

Rodolfo, Monica 5555<br />

Rodon Ahnert, Jordi 508,<br />

509, 2042, 3005, 3014<br />

Rodon, Jordi 566<br />

Rodon, Philippe 8014<br />

Rodrigo, Esteban 10082<br />

Rodrigues, Ana Maria Santos<br />

e12018<br />

Rodrigues, George 6130,<br />

7009<br />

Rodrigues, Helena 5567<br />

Rodrigues, Heloisa Veasey<br />

e13534<br />

Rodrigues, Isabelle 2070<br />

Rodrigues, Kristen 8512<br />

Rodrigues, Manuel Jorge 601<br />

Rodrigues, Newton Augusto<br />

Ferreira 643, e18078<br />

Rodriguez de Antona, Cristina<br />

4620, 10546, e15041<br />

Rodriguez de la Borbolla,<br />

Maria e11075<br />

Rodriguez Garcia, S<strong>of</strong>ia<br />

e15033<br />

Rodriguez Garrote, Mercedes<br />

e16029<br />

Rodriguez Lopez, Carmela<br />

e14733, e19505<br />

Rodriguez Paniagua, Jose<br />

Manuel 7011<br />

Rodriguez Villalva, Silvia<br />

e16025<br />

Rodriguez, Cesar 1001<br />

Rodriguez, Cle<strong>of</strong>e e15001<br />

Rodriguez, Cristina P. 7029,<br />

e14611, e14665, e16045,<br />

e18046<br />

Rodriguez, Cynthia e11061<br />

Rodriguez, Delvys 1531<br />

Rodriguez, Estelamari 7049<br />

Rodriguez, Isabel e11517<br />

Rodriguez, Luis 8596<br />

Rodriguez, Margarita 3066<br />

Rodriguez, Maria Alma 8067,<br />

e16574, e18530, e19625,<br />

e19629<br />

Rodriguez, Maria Carmen<br />

e19576<br />

Rodriguez, Melissa 8036<br />

Rodriguez, Miro e12041<br />

Rodriguez, Paulo 6580<br />

Rodriguez, Pedro Camilo<br />

2527<br />

Rodriguez, Rufo 4089<br />

Rodriguez, Vivian 1563<br />

Rodriguez-Antona, Cristina<br />

9073<br />

Rodriguez-Bigas, Miguel A.<br />

3556, 4060<br />

Rodriguez-Freixinos, Victor<br />

5068<br />

Rodriguez-Galindo, Carlos<br />

9523, 9539, 9545<br />

Rodriguez-Lescure, Alvaro<br />

1001, 1008<br />

Rodriguez-Marquez, Cristina<br />

1111<br />

Rodriguez-Moreno, Juan<br />

Francisco e15111<br />

Rodriguez-Morera, Anna<br />

e19542<br />

Rodriguez-Pascual, Jesus<br />

4040, e15111<br />

Rodríguez-Rigueiro, Teresa<br />

e14595, e21002<br />

Rodrigueza, Wendi V.<br />

TPS3110<br />

Rodriquenz, Mariagrazia<br />

e14156<br />

Roecken, Cristoph 4107<br />

Roeland, Eric e14069,<br />

e14078, e14079<br />

Roengvoraphoj, Monic<br />

e13567<br />

Roepman, Paul 3510, 10545<br />

Roesel, Siegfried 556<br />

Roesler, Rafael e13546<br />

Roessler, Bernhard 3530<br />

Roeth, Alexander 2007, 8031<br />

Roewert-Huber, Joachim<br />

8535, e19022<br />

R<strong>of</strong>f, Kate TPS4145<br />

Rogak, Lauren J. 9047,<br />

9104, 9144, e19633,<br />

e19647<br />

Rogan, Jane TPS10633^<br />

Rogerio, Jaqueline Willemann<br />

4608, 8028<br />

Rogers, Anna R 8543<br />

Rogers, Anna e19506<br />

Rogers, Gary S e11548<br />

Rogers, Jane 4060<br />

Rogers, John S. 7546<br />

Rogers, Karl e11562, e13003<br />

Rogers, Lisa R. TPS1616<br />

Roggenkamp, Betty 1553,<br />

6120<br />

Roggero, Enrico 4661<br />

Roggin, Kevin K. 10090<br />

Rogowski, Sabine 2040<br />

Rogowski, Wojciech e21099<br />

Roh, Jae Kyung e14081,<br />

e14084<br />

Roh, Jae Kyung 3531<br />

Roh, Sang Young e14558,<br />

e19614<br />

Rohan, Thomas e14005<br />

Roig, Jose Vicente 3586<br />

Roiter, Ignazio 1604<br />

Roitman, Janna 6114<br />

Rojas Llimpe, Fabiola Lorena<br />

e14040<br />

Rojas, Jose Ernesto e15036<br />

Rojas, Luis 7107, 7108,<br />

9571<br />

Rojas, Maria TPS4146<br />

Rojat-Habib, Marie-Christine<br />

8555<br />

Rojo, Federico 4580, 4582,<br />

4585, 5520, 10616,<br />

e14147, e15094, e18069,<br />

e18106, e21015<br />

Rojo, Sandra Viviana e14521<br />

Rokitta, Denis 3044<br />

Roland, Joseph e15582<br />

Rolet, Fanny 10505<br />

Rolfe, Lindsey 4041, e13028<br />

Rolfo, Christian Diego 4587,<br />

7058, 10594, e12012<br />

Rolland, Alain e19058<br />

Rolland, Frederic LBA4567^<br />

Rolle, Cleo E e17549<br />

Rolli, Luigi e17546<br />

Rollot, Florence TPS667<br />

Rom, Joachim 1096<br />

Rom, William e21012<br />

Romagosa, Cle<strong>of</strong>e e21021<br />

Romaguera, Jorge Enrique<br />

8067<br />

Romain, Ahmed Jerome 9074<br />

Roman, Laslo 8539<br />

Roman, Lynda e13084<br />

Roman, Orvelin e13581<br />

Roman, Ruth Ann 2521,<br />

8549, 8598<br />

Romaniuk, Chris 8564<br />

Romano, Anna 8573, e19034<br />

Romano, Carmen e14130<br />

Romano, Emanuela e19050<br />

Romano, Giampiero e18048<br />

Romano, Karen S. 3582<br />

Romano, Olivier e14560<br />

Romeder, Franz 4074<br />

Romejko-Jarosinska, Joanna<br />

e18512<br />

Romera, Alejandro 10564<br />

Romero, Atocha e14147<br />

Romero, Ignacio 5068,<br />

e15575<br />

Romieu, Gilles 532<br />

Romigh, Todd 1508<br />

Romkes, Marjorie 8528<br />

Rommel, Christian 626<br />

Rommel, Denis 5519<br />

Ron, Ilan-Gil 535<br />

Roncalli, Massimo 7061,<br />

7585, e14672, e18104<br />

Roncella, Silvio e17533<br />

Rondena, Roberta 9005<br />

Rondini, Ermanno LBA4001<br />

Rondon, Gabriela 6529,<br />

6540, 8102<br />

Rong, Tie-hua e18121<br />

Ronnett, Brigitte M. 1519<br />

Ronot, Maxime e14553<br />

Ronzoni, Monica 3518<br />

Rooney, Cliona M. 2558<br />

Root, Dana 2587<br />

Root, Elizabeth J. 1547<br />

Roothumnong, Ekkapong<br />

e14051<br />

Roque, Dana Marie e15526<br />

Roque, Dario R. 5100<br />

Rosa, Roberta e13509<br />

Rosales, Beatriz e19636<br />

Rosati, Gerado LBA4001<br />

Rosati, Gerardo e14040<br />

Rosati, Kayla 1045, 4098<br />

Rosbrook, Brad 4546<br />

Roschewski, Mark J. 8088,<br />

8104, e18568<br />

Roscoe, Joseph 9113, 9141,<br />

e16056<br />

Rose, Brent Shane 6121<br />

Rose, Inga 5523<br />

Rose, Jeffrey Scott e14178<br />

Rose, Jim 8060<br />

Rose, Mathieu e15193,<br />

e15198<br />

Rose, Michal G. 6033<br />

Rose, Shelonitda e13598<br />

779s


Rose, Steven C. 3590<br />

Rosell, Rafael 3093, 4068,<br />

4579, 7011, 7095, 7522,<br />

7531, 7540, 7542,<br />

TPS7613^, 10596,<br />

e14509, e14521, e18114,<br />

e18130, e18131, e18137,<br />

e18143<br />

Roselli, Mario TPS4151<br />

Rosello Keranen, Susana<br />

1074, e14589<br />

Rosemurgy, Alexander 4055,<br />

TPS4134, e14501<br />

Rosen, Alexander 6129<br />

Rosen, Alyx C. e19052,<br />

e19624<br />

Rosen, Barry 5107<br />

Rosen, Bruce R. 2059<br />

Rosen, David I e13086<br />

Rosen, Fred R 1611<br />

Rosen, Lane R e17524<br />

Rosen, Lee S. 3032, 3034,<br />

3042^, 3631, e13000,<br />

e13598<br />

Rosen, Mark Alan 4596<br />

Rosen, Neal 10618<br />

Rosen, Peter J. 3010<br />

Rosen, Steven T. 610,<br />

TPS1134, 2532, 4063,<br />

e18523<br />

Rosenbaum, Cara Ann 2561<br />

Rosenbaum, Eli 4564<br />

Rosenbaum, Paul R. 6000<br />

Rosenberg, Anne Louise<br />

e11505<br />

Rosenberg, Jonathan E.<br />

TPS2618, 4522, 4525,<br />

4543, 4577, 4580, 4582,<br />

4585, 4592, 4635,<br />

e15007<br />

Rosenberg, Joshua Daniel<br />

e21061<br />

Rosenberg, Martin e19635<br />

Rosenberg, Per e13095^,<br />

e13097^, e13108^<br />

Rosenberg, Robert 3510,<br />

TPS10632<br />

Rosenberg, Shoshana 9100<br />

Rosenberg, Steven A. 8576,<br />

e14179<br />

Rosenblat, Todd L. 6609,<br />

6613<br />

Rosenblatt, Joseph David<br />

8027, e13079<br />

Rosenfeld, Nitzan 544<br />

Rosenko, Liudmila e19049<br />

Rosenman, Julian e17547,<br />

e17550<br />

Rosenow, Joshua M. 2035<br />

Rosenshein, Neil B. 5055<br />

Rosenstock, Julio 1509, 1510<br />

Rosenthal, David Ira 5530,<br />

5561, 5589<br />

Rosenthal, Mark 4531,<br />

e13054<br />

Rosenthal, Seth A e21026<br />

Rosenzweig, Margaret Quinn<br />

1085, 1589<br />

Roses, Daniel F. e11010<br />

Rosina Duarte, Eva 9063<br />

Rosing, Hilde 2550, e13028<br />

Rosmorduc, Olivier 4032,<br />

TPS4148<br />

Rosner, Gary L. 4559<br />

Rosovsky, Rachel Pam<br />

Greenerger 7010<br />

Ross, Douglas T. 516<br />

Ross, Eric A 1502, 4526<br />

Ross, Fiona 8015<br />

Ross, Gillian 10060<br />

Ross, Graham 533^, 597^,<br />

598^, e11055^<br />

Ross, Helen J. 7029, 7555<br />

Ross, Jeffrey S. 3533, 7510,<br />

10590, e17541<br />

Ross, Joseph 1595<br />

Ross, Mikel TPS3112<br />

Ross, Richard N. 6000<br />

Ross, Robert W. 3105<br />

Ross, Robert W 4516, 4577,<br />

4582<br />

Ross, Ronald K 4575<br />

Ross, Sharona B. e14501<br />

Rosselli Del Turco, Marco<br />

1500<br />

Rossello, Rosalba e11056<br />

Rosser, Charles Joel e15010<br />

Rossetti, James M. 6620<br />

Rossetti, Maria Grazia<br />

e13098<br />

Rossetto, Ciro 5513<br />

Rossi, Adriana C. 8036<br />

Rossi, Cristina 10593<br />

Rossi, David 4084<br />

Rossi, Edmund A e13005<br />

Rossi, Elisa e21018<br />

Rossi, Emma C e15561<br />

Rossi, Gabriela R. 2571,<br />

e19008<br />

Rossi, Giorgia 9054, e19532,<br />

e19533<br />

Rossi, Giuseppe 8006<br />

Rossi, Jean-Francois 8018^,<br />

8020<br />

Rossi, John 10545<br />

Rossi, Lorena e14082<br />

Rossi, Luigi e11039,<br />

e11514, e12510, e19620<br />

Rossi, Maura e13569<br />

Rossi, Peter John e15006<br />

Rossoni, Gilda 7076, 7581<br />

Rosti, Giovanni e13019<br />

Rota, Luigina e14018<br />

Rota, Selene 623<br />

Roth, Arnaud 3509, 3511,<br />

3575, 3595, TPS4140,<br />

10033<br />

Roth, Daniela 10593<br />

Roth, Diane 3019<br />

Roth, Jack A. 7043<br />

Roth, Joshua A. 560<br />

Roth, Laura 2503^<br />

Roth, Patrick 2007, 2055<br />

Roth, Stephan L. e11556<br />

Rothbard, Marianna TPS669,<br />

TPS1614<br />

Rothe, Achim 8079<br />

Rothenberg, Stephen M.<br />

4566, e16059<br />

Rothenberger, David A. 6016<br />

Rothhammer-Hampl, Tanja<br />

4034<br />

Rotmensz, Nicole 1049,<br />

1115, 1500<br />

Rotoloni, Christina e14689<br />

Rottey, Sylvie 4622, 5504^,<br />

e13594, e15115<br />

Rouanet, Philippe 568^,<br />

3633<br />

Roubec, Jaromir e18062<br />

Roughley, Adam e16570<br />

Rougier, Philippe R 3562,<br />

3579<br />

Rougier, Philippe 3508,<br />

3547, TPS3634, 4087<br />

Roupret, Morgan e15105<br />

Rouquette, Isabelle 10507<br />

Rourke, Sean 9021<br />

Rousseau, Raphael F.<br />

TPS9596, TPS9597^<br />

Rousseau, Vanessa 4032,<br />

10556<br />

Roussel, Claire 1568<br />

Roussel, Helene e13578<br />

Roussel, Murielle 8009, 8014<br />

Routt Barnes, Sheri e19577<br />

Rouyer, Magali e14168<br />

Rouyer, Marie TPS10634<br />

Rouzier, Roman 568^<br />

Rovati, Bianca 4017, e13103<br />

Rove, Kyle O. e15176<br />

Rovere, Giulia e18103<br />

Rovere, Rodrigo e12035<br />

Roveta, Annalisa 7082, 7084<br />

Rovithi, Maria e18094<br />

Rowbottom, Jacqui 5001<br />

Rowe, Julie e14684<br />

Rowe, Richard 6113<br />

Rowell, Jessica Lauren<br />

e15187<br />

Rowland, Christine 6113<br />

Rowland, Julia Howe 6006,<br />

9130<br />

Rowland, Kendrith M. 7073<br />

Rowland, Suzanne e14015<br />

Rowley, Steve 1021<br />

Roxburgh, Campbell SD<br />

3611, e14054, e14092,<br />

e14117<br />

Roy, Amit TPS2622, 8541<br />

Roy, Josee-Anne TPS1139<br />

Roy, Larry 2612<br />

Roy, Rajasree TPS5601<br />

ROY, Rakesh - e19517<br />

Roy, Siddharth 10515<br />

Roy, Vivek 8033<br />

Royce, Melanie 1558<br />

Royce, Trevor Joseph 4658,<br />

6032, 6037<br />

Royer-Joo, Stephanie 3543,<br />

3552<br />

Royer-Pokora, Brigitte 3550<br />

Roz, Elena e14042, e18021<br />

Rozanec, Natalie 6005<br />

Rozmovits, Linda e11000<br />

Rozmus, Cathy L. 9096<br />

Rozzini, Renzo e14018<br />

Rua, Oliver e15036<br />

Ruan, Libin e14523<br />

Ruane, Thomas e16552<br />

Ruatta, Fiorella e15073,<br />

e15133<br />

Rubanov, Oleg 4023<br />

Rubenstein, Jonathan B.<br />

e18502<br />

Rubenstein, Marvin e13513,<br />

e13532, e15119<br />

Rubie, Herve 9517, 10076<br />

Rubin, Eric H. e13599<br />

Rubin, Mark A. 4649<br />

Rubin, Mark S. 7587, 8556,<br />

10530, e21023<br />

Rubin, Peter 1016<br />

Rubin, Stephen D. 531, 610<br />

Rubini, Michele e14041<br />

Rubino, Barbara 4588<br />

Rubino, Corrado 8581<br />

Rubino, Luca 7585, e14672,<br />

e18104, e21139<br />

Rubinsak, James R. LBA4<br />

Rubinstein, Daniel B. e13018<br />

Rubinstein, Larry 2606,<br />

3529, 10603<br />

Rubinstein, Paul G. 1611,<br />

8085, e14590<br />

Rubio Salvador(2), Ana Rosa<br />

e11543, e19596<br />

Rubio, Belen e18015<br />

Rubio, Gustavo 5520,<br />

e15094, e18069, e18106,<br />

e21015<br />

Rubio, Itziar e11525,<br />

e17522, e18031, e21009<br />

Rubio, Maria Jesus 5031<br />

Rubio, Marie Therese 6534<br />

Rubio-Viqueira, Belen 1531<br />

Ruby, Jeannine Alberts 3563<br />

Ruch, Joshua Michael<br />

e14519, e14578<br />

Rucinska, Monika e19561<br />

Ruckdeschel, John C. 7029<br />

Ruckser, Reinhard e14087<br />

Ruco, Luigi e14042<br />

Ruda, Roberta 2037, 2075<br />

Rudas, Margaretha 542,<br />

10570<br />

Rudd, Robin 7562<br />

Ruddy, Kathryn Jean 9071,<br />

9100<br />

Rudek, Michelle A. TPS3117<br />

Rudengren, Cecilia 9054<br />

Rudenko, Elena G. 9060<br />

Rudich, Steven e14714<br />

Rudin, Charles M. 1573,<br />

3024, 7532, 7589,<br />

e18147<br />

Rudin, Claudius 8015<br />

Rudman, Sarah Maria 4555<br />

Rudnick, Jeremy 2080, 2084,<br />

2087<br />

Rudnicka, Halina 10582<br />

Rudzinski, Erin R. 9535<br />

Ruebe, Christian 8022<br />

Rueda, Antonio 8062,<br />

e14116<br />

Ruediger, Thomas 1023<br />

Ruehle, Kathleen e19565<br />

Ruehlman, Peter 4127<br />

Ruel, Christopher 3108,<br />

e15114<br />

Ruf, Peter 2584<br />

Ruff, Paul 3505<br />

Ruffion, Alain e15182<br />

Rugge, Massimo e18130<br />

Ruggeri, Enzo Maria e18065<br />

Ruggeri, Enzo e12514,<br />

e14082<br />

Ruggeri, Maria Grazia e19595<br />

Ruggieri, Eustachio e21134<br />

Ruggieri, Pietro 10022<br />

Ruggiero, Alessandro 10091<br />

Rugo, Hope S. 512, 539,<br />

540, 551, 559, 599,<br />

CRA1002, 1006,<br />

TPS1140^, 1526, 3058,<br />

9107, e11096<br />

Ruhstaller, Thomas 9059<br />

Ruiz Borrego, Manuel 1001,<br />

e11081<br />

Ruiz de Lobera, Abigail<br />

e11525, e21009<br />

Ruiz de Porras, Vicenç 9129<br />

Ruiz Sanchez, Daneli e15525<br />

Ruiz, Amparo 1001, 1008,<br />

1059<br />

Ruiz, Ana L. 7049<br />

Ruiz, Irune 4089<br />

Ruiz, Marco A e11030<br />

Ruiz, Wilson e12041<br />

Ruiz, Yelitza e14102<br />

Ruiz-Garcia, Ana e18093<br />

Ruiz-Opazo, Nelson e13574<br />

Ruiz-Sauri, Amparo e15575<br />

Ruiz-Valdepeñas, Andrea<br />

e11527, e12015, e17502,<br />

e17503, e17515<br />

780s Numerals refer to abstract number


Rummel, Mathias J. 3<br />

Rumyantzeva, E e11528<br />

Rundle, Twana e19588<br />

Runnebaum, Ingo B. e15566<br />

Ruocco, Raffaella 1554<br />

Ruperez Blanco, Ana Belen<br />

e12003, e14058<br />

Rupin, Matthieu TPS648<br />

Ruppert, Amy S. e18508<br />

Ruppert, Anne-Marie 10507<br />

Rusakov, Igor e15047<br />

Rusch, Valerie W. 1541,<br />

4061, 7014, 7066<br />

Ruscito, Ilary 5070<br />

Rush, Janet 6530<br />

Rushing, Daniel A. 9571,<br />

10010<br />

Rusinek, Dagmara e14159<br />

Russell, Christy Ann 1065<br />

Russell, Greg B. 6127<br />

Russell, Karen Bullock<br />

e13589<br />

Russell, Ludivine e19625,<br />

e19629<br />

Russell, Prudence e17539<br />

Russell, Stephen J. 8096,<br />

8105, TPS8116<br />

Russell, Valerie 2521<br />

Russell, Wesley Alan e18502<br />

Russo, Antonio e11056,<br />

e14042, e18021<br />

Russo, Domenico 6531<br />

Russo, Filippo 8066, 8083,<br />

e20512<br />

Russo, Leila 1022<br />

Russo, Lucianna e14711,<br />

e15160<br />

Russo, Moises e14002<br />

Russo, Stefania 1044,<br />

e13058, e13070<br />

Russo, Suzanne M. e17547,<br />

e17550<br />

Rustin, Gordon J. S. 3048<br />

Ruszniewski, Philippe 4014,<br />

4071, 4133, e14660<br />

Rutgers, Emiel J. 1022<br />

Ruth, Karen 1544, 6061<br />

Ruther, Nancy R. 6074<br />

Rutherford, Thomas J. 5099,<br />

e13125, e15526, e15581<br />

Rutkowski, Piotr LBA8500^,<br />

LBA8509, 8548,<br />

LBA10008, 10033, 10096,<br />

10538, e19022<br />

Rutledge, James e11562,<br />

e13003<br />

Rutstein, Mark D. TPS3634,<br />

TPS4675^, 5012<br />

Ruud, Christopher O. e11069<br />

Ruvalcaba Limon, Eva<br />

e15527<br />

Ruxer, Robert L. 3069<br />

Ruzich, Janet C. 9028<br />

Ruzzo, Annamaria 3518,<br />

3540<br />

Ryan, Aoife M 1559<br />

Ryan, Charles J. 3058,<br />

LBA4518, 4549, 4664,<br />

4667, e15137<br />

Ryan, Christine 3047<br />

Ryan, Christopher W. 4586,<br />

10011^, TPS10103<br />

Ryan, David P. 3528, 3594,<br />

4021, 4027, 4112, 4125,<br />

e14615<br />

Ryan, Julie L. 9027<br />

Ryan, Paula D. 1009,<br />

e11085<br />

Rybak, Christina 1544<br />

Numerals refer to abstract number<br />

Rybicki, Lisa A. 1076, 8071,<br />

e14611, e14665<br />

Rybka, Witold B. 8090<br />

Rydall, Anne 9003<br />

Rydstrom, Karin 8074<br />

Rymeski, Beth A e20007<br />

Rymkiewicz, Grzegorz e18512<br />

Ryoo, Baek-Yeol 4082,<br />

10085, 10093, e13092,<br />

e13104, e14543, e14617<br />

Ryoo, Hun-Mo e14570,<br />

e19538<br />

Rüsch<strong>of</strong>f, Josef 3600,<br />

e14161, e14162, e14167<br />

Rytting, Michael E. 6501<br />

Ryu, Jeong Seon e13065<br />

Ryu, Keun Won 4028<br />

Ryu, Min-Hee 4082, 10085,<br />

10093, e13092, e13104,<br />

e14543, e14580<br />

Rzyman, Witold 10561<br />

Rödel, Claus e14161,<br />

e14162<br />

Rödel, Franz e14161,<br />

e14162<br />

S<br />

S K, Raghunath e15101<br />

S, Nirmala Srikantaiah<br />

e11536<br />

S, Ramesh Bilimagga 1093,<br />

e11511, e11536, e12030,<br />

e14721<br />

Sa Cunha, Antonio e14168<br />

Sá, Lilian Andrade e19628<br />

Saab, Raya Hamad e20004<br />

Saad, Alain 8026<br />

Saad, Everardo D. 3557,<br />

e13055<br />

Saad, Fred 4510, 4519^,<br />

4558, e15202, e19514<br />

Saad-Hossne, Rogerio e14173<br />

Saada, Esma 5521, 10062<br />

Saadeh, Charles e21105<br />

Saarelainen, Sami Kristian<br />

e15564<br />

Saavedra-Perez, David<br />

e18028<br />

Saba, Hanna M. 7005<br />

Saba, Nabil F. 2576, 5560,<br />

5570, 7048<br />

Sabaawy, Hatem E. e13543<br />

Sabado, Rachel Lubong 2589<br />

Sabariz, Luis 10595<br />

Sabate, Josep Maria 1120<br />

Sabatino, Susan A 6052<br />

Sabatti, Chiara 10513<br />

Sabbatini, Armando e21070<br />

Sabbatini, Paul 5041<br />

Sabbatini, Roberto TPS4683<br />

Sabeh, Farideh 8099<br />

Sabel, Michael 2000, 2067<br />

Sabet, Amir e14565<br />

Sabet, Salwa Farouk e21055<br />

Sabichi, Anita Lyn 5592<br />

Sabin, Pilar 8062<br />

Sabir, Noreen 6612<br />

Sablin, Marie-Paule e13017<br />

Sabl<strong>of</strong>f, Bradley e14547<br />

Sabo, Roy T. e18550<br />

Sabourin, Jean-Christophe<br />

5565, e14010, e14129<br />

Sabzevari, Helen TPS2617<br />

Saccaro, Steven J. 9061<br />

Sacchi, Stefano 8006<br />

Sacco, Cosimo LBA5003,<br />

e15160<br />

Sacco, Rosario e21113<br />

Sacconi, Andrea 3521<br />

Sachdev, Deepali e13590<br />

Sachdev, Jasgit C. TPS1137<br />

Sachdev, Pallavi 5518,<br />

e19055<br />

Sachdeva, Kush 6021<br />

Sacher, Adrian G. 7606<br />

Sachs, Karen L e21060,<br />

e21111<br />

Sacks, Natalie 2562<br />

Sada, Yvonne 3542<br />

Sadaf, Tabinda e16043<br />

Sadahiro, Sotaro 3607<br />

Sadak, Karim Thomas 9587<br />

Saddekni, Souheil e13087<br />

Sadeghi, Sarmad 1514<br />

Sadek, Ibrahim 6573<br />

Sadelain, Michel TPS2619,<br />

TPS4700<br />

Sadetsky, Natalia e19060<br />

Sadhashiv, Santhosh 6620<br />

Sadighi, Zsila Sousan 2022<br />

Sadikov, Aleksander e16505<br />

Sadim, Maureen 1561<br />

Sadis, Seth e14164<br />

Sadoun, Alain 5069<br />

Sadowinski Pine, Stanislaw<br />

e20005<br />

Sadrzadeh, Hossein 6606,<br />

6617, 6618, 6629<br />

Saeb-Parsy, Kasra e15011<br />

Saeed, Anwaar Mohammed<br />

e21085<br />

Saeed, Kamran e16043<br />

Saeed, Sadiya 10525<br />

Saeger, Sally TPS2622<br />

Saeki, Hiroshi e14033,<br />

e14601<br />

Saeki, Sho e13072, e18059<br />

Saeki, Toshiaki TPS659<br />

Saenz, Alberto e15144<br />

Saetaew, Manit e13023<br />

Saez, M.I. 4630, TPS4672<br />

Safferman, Allan Zachary<br />

7598<br />

Saffery, Claire LBA4000<br />

Safgren, Stephanie L. e13086<br />

Safina, Sufia 531<br />

Safont, M. José 3571, 3586,<br />

e14097<br />

Safra, Tamar 5078<br />

Safran, Howard 4049, 4098,<br />

9117<br />

Safwat, Mohab W. e15039,<br />

e15046<br />

Safwat, Reham e16002<br />

Sagalowsky, Arthur I. 4616,<br />

TPS4678, e15192<br />

Sagan, Christine 543,<br />

e21117<br />

Sagara, Yasuaki 588<br />

Sagasser, Jaqueline e13100^<br />

Sagatys, Elizabeth e18503,<br />

e18506<br />

Saggese, Mariangela 8066<br />

Saggio, Jennifer 9540<br />

Saghatchian, Mahasti e11019<br />

Sagiv, Eyal 1507<br />

Saglam, Sezer e14628<br />

Saglio, Giuseppe 6506,<br />

6509^<br />

Saha, Debabrata 10624<br />

Saha, Sukamal 3619,<br />

e14045, e14046, e15554<br />

Sahai, Vaibhav e14515<br />

Sahani, Dushyant e14615<br />

Sahar, Sumrin 6612<br />

Sahasrabudhe, Deepak M.<br />

e15132<br />

Sahebi, Firoozeh 6545, 8038,<br />

8089<br />

Sahin, Aysegul A. 1130<br />

Sahin, Berksoy e11088<br />

Sahin, Ugur e11020,<br />

e11092, e11509<br />

Sahmoud, Tarek 512, 540,<br />

551, 559, TPS648, 4081<br />

Sahmoun, Abe E e21125<br />

Sahoo, Tarini Prasad<br />

LBA7507<br />

Sahovic, Entezam A. 6620<br />

Sai, Yang 3038, 3040<br />

Saiag, Philippe 1566, 8540,<br />

8591, 8601<br />

Saiagh, Soraya e19019<br />

Said, Jonathan W. e13045<br />

Said, Rabih 10607<br />

Saida, Isamu e14622<br />

Saida, Yoshihisa 3569<br />

Saif, Wasif M. 3522, 3523,<br />

3525<br />

Saigal, Kunal 595, e16001<br />

Saigí, Eugeni 4079<br />

Saigusa, Susumu e14029,<br />

e14039, e14541, e21053<br />

Saijo, Nagahiro 6112, 7003,<br />

7017, 7028<br />

Saijo, Yasuo 7561<br />

Saikia, Junmi e15084<br />

Sailer, Scott Lee 9537<br />

Sailors, Mary H 6551, 8091,<br />

9083, 9140<br />

Saini, Ashima e11084<br />

Saini, Kamal V.S. e21010<br />

Saint-Martin, Jean-Richard<br />

1055, e13549<br />

Saint-Pierre, Christine 634<br />

Saintigny, Pierre 5524<br />

Sainz de la Cuesta, Ricardo<br />

1111<br />

Sainz Jaspeado, Miguel 3096<br />

Sairi, Alisa 10051, e20509<br />

Sait, Sheila N.J. 6565<br />

Saita, Naoki e18059<br />

Saito, Akihisa 5084<br />

Saito, Hideyuki e13072<br />

Saito, Hiroshi 7521<br />

Saito, Kanako e19546<br />

Saito, Kayoko e21108<br />

Saito, Kazutaka e15071<br />

Saito, Mari 1106<br />

Saito, Naoko e19611<br />

Saito, Norio 3524, 3569<br />

Saito, Ryota 7086, 10569,<br />

10604, e18004, e18014,<br />

e18017<br />

Saito, Tatsuro e18064<br />

Saito, Yuki 565<br />

Saiura, Akio 3625, e14506<br />

Saji, Shigetoyo 3607<br />

Sajjad, Monique 1585<br />

Sakaguchi, Masahiro e18126<br />

Sakai, Asao e17540<br />

Sakai, Hiroshi 7012, 7028,<br />

7515, 10542<br />

Sakai, Kazuko 10569<br />

Sakairi, Yuichi 7046<br />

Sakakibara, Tomohiro 7086,<br />

7565<br />

Sakamaki, Hisashi 6533<br />

Sakamoto, Junichi 1106,<br />

3607, TPS3642, e14123<br />

Sakamoto, Kazuhiro e14139<br />

Sakamoto, Michiie 4104<br />

Sakamoto, Susumu e18127<br />

Sakamoto, Yasuo e13553<br />

Sakamoto, Yoshihiro e14506<br />

Sakar, Burak e14628<br />

781s


Sakata, Shinya e18059<br />

Sakata, Yuh 3593, e14062<br />

Sakatani, Toshio 10569,<br />

10604, e18004, e18014,<br />

e18017<br />

Sakellis, Chris 8552<br />

Sakihama, Hideyasu 10578<br />

Sakr, Amr Yehia 5557<br />

Sakr, Bachir Joseph 1034,<br />

1045, 5040<br />

Sakr, Rita 10618<br />

Saks, Sam TPS10100<br />

Sakurada, Mutsumi e14139<br />

Sakurai, Toshihiko e15057,<br />

e15088<br />

Sakuramoto, Shinichi 4012<br />

Sakuyama, Naoki e14004<br />

Sala, Nuria 1588<br />

Salajka, Frantisek e18062<br />

Salamon, Hugh 5074<br />

Salani, Ritu e15585<br />

Salanova, Ruben e14109<br />

Salas, Sébastien 10042<br />

Salat, Christoph e13095^,<br />

e13097^, e13108^,<br />

e21067<br />

Salati, Michele e21070<br />

Salaun, Pierre-Yves TPS646<br />

Salazar, Maria Teresa e12033<br />

Salazar, Ramon 3510, 3571,<br />

4122, TPS10632<br />

Salcedo, Maite 7041<br />

Saleh, Mansoor N. LBA3501,<br />

TPS3640, TPS4697<br />

Salek, Tomas TPS4677,<br />

e15028, e19020<br />

Salem, Ahmed Abdel-Fattah<br />

e15024<br />

Salem, Ayman Maher e21055<br />

Salem, Jonas Hauben e14101<br />

Salem, Marwa 552<br />

Salem, Mohamed E. 2081,<br />

5534, e14609<br />

Salem, Naji e15155, e19539<br />

Saleri, Jessica 9120<br />

Salerno May, Kilian e14659,<br />

e14712, e19041<br />

Sales De Gauzy, Jerome<br />

10076<br />

Sales, Elizabeth 5029<br />

Salesi, Nello e19501<br />

Saletan, Stephen 4014<br />

Salgado, Roberto 507,<br />

e21010<br />

Salgia, Ravi 7508, 7533,<br />

7596, 7599, 7600,<br />

e17549<br />

Salhi, Yacine e15007<br />

Salice, Placida e18026<br />

Salido, Marta 4580<br />

Salim, Muhammad 1036<br />

Salimi, Ghobad 6556<br />

Salinas, Claudia A. e15205<br />

Salkeni, Mohamad Adham<br />

TPS3116, e16017<br />

Sallan, Stephen E. 9530<br />

Salles, Gilles A. 2539, 8021,<br />

8068<br />

Sallman, David A e18520<br />

Salloum, Ramzi George 4647,<br />

e18144<br />

Salman, Reem 1113,<br />

e19601, e21147<br />

Salmasi, Amirali H e15214<br />

Salmen, Jessica 1600^<br />

Salmon, Asher 10034<br />

Salmon, Isabelle e19026<br />

Saltarelli, Francesca e17006,<br />

e18563, e18566<br />

Saltel, Pierre 9080<br />

Saltman, Benjamin TPS5601,<br />

e16026<br />

Saltz, Leonard 3563, 3583,<br />

6073, 9094, e14112<br />

Salud, Antonia 3586<br />

Salud, Antonieta e14041,<br />

e14097<br />

Salva, Silvia 2515<br />

Salvador B<strong>of</strong>ill, Francisco<br />

Javier e11081<br />

Salvador B<strong>of</strong>ill, Javier 10608<br />

Salvador, Christopher G. 6553<br />

Salvador, Javier e11075,<br />

e11535, e21088<br />

Salvador, Wenimar e17547<br />

Salvadori, Barbara 629<br />

Salvagni, Stefania 4006<br />

Salvajoli, Joao Victor e15509,<br />

e16046<br />

Salvat, Jacques 9011<br />

Salvati, Mark e16053<br />

Salvatierra, Alejandro e14521<br />

Salvatore, Lisa 3518, 3540,<br />

3546, 3549, 3555, 3585,<br />

3597<br />

Salvesen, Helga B 5010<br />

Salvini, Jessica 3521,<br />

e21018, e21095<br />

Salvini, Piermario 623<br />

Salvioni, Roberto 4507, 4629<br />

Salvucci, Marzia 6531<br />

Salwen, Helen 9534<br />

Salz, Talya 6073<br />

Salzberg, Marc Oliver e18012<br />

Salzer, Wanda L. CRA9508<br />

Samadi, David B. e15164<br />

Samalin, Emmanuelle 3633,<br />

4087<br />

Samanez Figari, César<br />

e18524<br />

Samaniego, Felipe 8067<br />

Samant, Meghna 528<br />

Samant, Sandeep 5500<br />

Samaras, Panagiotis 3595<br />

Sambataro, Daniela e15544,<br />

e18026<br />

Samlowski, Wolfram E. 8527,<br />

10614<br />

Samoilova, Olga S 8018^<br />

Samonigg, Hellmut 514,<br />

TPS3641^, 10501,<br />

e14066<br />

Sampaio, Carlos e11087<br />

Sampath, Sagus TPS5602<br />

Samper, Esther e21035<br />

Sampson, John Howard<br />

2090^, 2094^, 2095^,<br />

TPS2103<br />

Samrao, Damanzoopinder<br />

5091<br />

Sams, Katherine 9108<br />

Samsa, Julia e18046,<br />

e18085<br />

Samson, Benoit LBA3500^,<br />

3572<br />

Samson, Solene e14065<br />

Samson, Susan e11096<br />

Samstein, Benjamin 4101<br />

Samuel, Leslie M. TPS3637<br />

Samuel, Thomas A. 8531<br />

Samuels, Judith 7049<br />

Samuels, Michael 7049,<br />

e16001<br />

Samuelson, Megan I e15580<br />

Samura, Osamu 5084<br />

San Juan del Moral(1),<br />

Alberto e11543, e19596<br />

San-Miguel, Jesùs F. 8095,<br />

TPS8112, TPS8113,<br />

e18572^<br />

Sanabria, Sandra 2568,<br />

e13048<br />

Sanborn, Rachel E. 7546,<br />

e13550<br />

Sanchez Rovira, Pedro 1059,<br />

10616<br />

Sanchez Torres, Jose Miguel<br />

e18011<br />

Sanchez Torres, Jose Miguel<br />

e18055<br />

Sánchez, Antonio 3618,<br />

e18011, e18521<br />

Sanchez, Belen 4579<br />

Sanchez, Bertha E. e15174<br />

Sanchez, Eric e18549<br />

Sanchez, Jose Angel e19515<br />

Sanchez, Jose Javier 4068,<br />

4579, 10594, e14521,<br />

e18130, e18131, e18137,<br />

e18143<br />

Sanchez, Jose Luis e19576<br />

Sanchez, Jose Miguel 7011,<br />

7540<br />

Sanchez, Lara 9073<br />

Sanchez, Lorena e15111<br />

Sanchez-Avila, Carmen 1570<br />

Sanchez-Hernandez, Alfredo<br />

e15149, e17545, e18011,<br />

e18049, e18053<br />

Sanchez-Lara, Karla e19594<br />

Sanchez-Muñoz, Alfonso<br />

10616<br />

Sánchez-Ollé, Gessamí 509,<br />

566, 619, 3014<br />

Sanchez-Pla, Alex 10511<br />

Sánchez-Rovira, Pedro 1011,<br />

10608<br />

Sanchez-Simon, Raquel<br />

10500<br />

Sanchez-Tillo, Esther e21035<br />

Sancho, Aintzane e11525,<br />

e17522, e18031, e21009<br />

Sancho, Blanca 642<br />

Sancho, Francesc e14552<br />

Sancho, P e19023<br />

Sanda, Martin G. 4521<br />

Sanda, Takaomi e13586<br />

Sandadi, Samith 2586<br />

Sandanayaka, Vincent 1055,<br />

e15200<br />

Sandberg, Rickard 10539<br />

Sandecki, Anita TPS4687<br />

Sanden, Mats Olot 10528,<br />

10575, e21008<br />

Sandene, Erin K. TPS3638<br />

Sander, Chris 4527<br />

Sander, Cindy 8533, 8547<br />

Sanders, Heather 8596<br />

Sanders, Joyce 562<br />

Sanders, Karen 4509<br />

Sanders, Lea 1084<br />

Sanders, Melinda 510, 1024<br />

Sanders, Sheri e21152<br />

Sandhu, Gurdarshan Singh<br />

4594<br />

Sandhu, Shahneen Kaur<br />

4553, e13601, e15134,<br />

e15136<br />

Sandhu, Sonia 8085<br />

Sandin, Rickard 6092,<br />

e16530<br />

Sandler, Alan 7094,<br />

TPS7609<br />

Sandler, Andrew TPS4673<br />

Sandor, George 9529<br />

Sandor, Victor TPS6643^<br />

Sandoval-Sus, Jose D e18109<br />

Sandoval-Tan, Jennifer 7519^<br />

Sands, Colleen B. 6095,<br />

6128<br />

Sands, Robert e15193<br />

Sanfilippo, Nicholas e16052<br />

Sanfilippo, Roberta 10053,<br />

e19549<br />

Sanford, Tiffany 3630<br />

Sanft, Tara Beth 9007,<br />

e16511<br />

Sang, Jim 3090<br />

Sangai, Takafumi 1054,<br />

e14662<br />

Sangale, Zaina 7023<br />

Sangalli, Claudia 10065<br />

Sangha, Randeep e13011<br />

Sangiolo, Dario 10066<br />

Sangro, Bruno TPS4148,<br />

e14654<br />

Sanhes, Laurence 8026<br />

Sanjay, Gupta e14664<br />

Sanjuan, Xavier 10052<br />

Sankhala, Kamalesh 10036<br />

Sanmamed, Miguel F. 8572<br />

Sanmartin, Elena 7058,<br />

7060, 10594<br />

Sanmugarajah, Jasotha<br />

e19598<br />

Sanna, Stefano e17546<br />

Sano, Fumiho e17517<br />

Sano, Go e18127<br />

Sano, Takeshi 4012, 10541<br />

Sanpajit, Theeranun 4108<br />

Santaballa, Ana 5068,<br />

e11081, e15575<br />

Santacruz, Mayra e15571<br />

Santagata, Sandro 2020<br />

Santamaria-Galicia, Julieta<br />

e12027<br />

Santamarina Cainzos, Isabel<br />

e14595, e15076, e15077,<br />

e15079<br />

Santana, Iuri amorim 2038<br />

Santana, M. Christina 6097<br />

Santana, Rosane O e16046<br />

Santana-Davila, Rafael 1089,<br />

e17555<br />

Santander Lobera, Carmen<br />

TPS4672<br />

Santarpia, Libero 1012<br />

Santarpia, Mariacarmela<br />

7522, 7542, e18039<br />

Santegoets, Saskia 2562<br />

Santel, A. e13597<br />

Santella, Regina 4101<br />

Santi, Patricia e19552<br />

Santiago Crespo, JA e19590<br />

Santiago, Karen J. e11015,<br />

e18547<br />

Santiesteban, Eduardo Rafael<br />

2527<br />

Santillo, Barbara 1500<br />

Santin, Alessandro 5099,<br />

e13078, e13125, e15526,<br />

e15581<br />

Santinami, Mario 8513,<br />

8529, 8559<br />

Santinelli, Alfredo e21070<br />

Santini, Daniele 4627, 9005,<br />

10063, e14135<br />

Santini, Donatella e14647<br />

Santini, Fernando Costa<br />

e11014<br />

Santomé, Lucía e15067,<br />

e15076, e15077<br />

Santoni-Rugiu, Eric 10617<br />

782s Numerals refer to abstract number


Santoro, Armando 615, 623,<br />

2580, 3007, LBA4001,<br />

4006, 4115, 4117, 4545,<br />

7061, 7082, 7085, 7585,<br />

TPS10101, e13095^,<br />

e13097^, e14672,<br />

e18100, e18104, e21139<br />

Santoro, Joseph e15138<br />

Santos Borges, Giuliano<br />

e12035<br />

Santos Cores, Jesus e14097<br />

Santos Ortega, Manuel<br />

e16025<br />

Santos, Carlos F. 2528<br />

Santos, Cristina 9090<br />

Santos, Edgardo S. e13032,<br />

e16001, e17564, e18109<br />

Santos, Elizabeth S. 643,<br />

e18078<br />

Santos, Elizabeth Santana<br />

e14680<br />

Santos, Erika Maria Monteiro<br />

e14119<br />

Santos, Evandro Airton Sordi<br />

dos e12512<br />

Santos, Fábio Nasser e11058<br />

Santos, José Sebastião<br />

e14599<br />

Santos, Marcos Antonio 3063<br />

Sanyal, Arun e14581<br />

Sanz, Ignacio 1111<br />

Sanz, Julian 10564<br />

Saphner, Thomas James 605<br />

Sapino, Anna 10554<br />

Sapolsky, Jeffrey 2503^<br />

Saponara, Maristella 10087<br />

Sappington, Sandra 6132<br />

Sapunar, Francisco J.<br />

TPS4140<br />

Saracino, Valeria e18048<br />

Saragiotto, Daniel F. e14702<br />

Saralli, Erminio e19002<br />

Saran, Frank TPS9596,<br />

10060<br />

Sarangarajan, Rangaprasad<br />

3015, e14593<br />

Sarangi, Manas ranjan 7102<br />

Sarantopoulos, John 3060,<br />

3073, 3078, 4603,<br />

e15116<br />

Sardesai, Sagar D 6565<br />

Sargent, Daniel J. 2004,<br />

3502, 3514, 3526, 3564,<br />

3576, 3617, 10602<br />

Saric, Nera e21126<br />

Sarici, Saim Furkan e11020<br />

Sarina, Barbara 6541<br />

Sarkar, Fazlul H. 1560,<br />

e21057<br />

Sarkar, Sudipa 10558<br />

Sarkaria, Jann Nagina 2031,<br />

2032, 7073, e14507<br />

Sarker, Debashis 3022<br />

Sarkisian, Saro 8533<br />

Sarma, Deesha 1577<br />

Sarma, Syam e14141,<br />

e14146, e15183, e18097<br />

Sarnaik, Amod 2511<br />

Sarno, Francesca 3068<br />

Sarno, Maria Anna 1554<br />

Sarno, Mark J. e15113<br />

Sarobba, Maria Giuseppa<br />

LBA7501<br />

Sarr, Celine 2543<br />

Sarta, Catherine e16058<br />

Sartelet, Hervé 9563, 9570<br />

Sarti, Manuela 4661<br />

Sarti, Samanta e11054<br />

Sarto, Gloria 1501<br />

Numerals refer to abstract number<br />

Sartor, A. Oliver LBA4512,<br />

4513, 4515, 4551, 4652,<br />

TPS4696^, 10623,<br />

e15130<br />

Sartore-Bianchi, Andrea 3570<br />

Sartorelli, Alan e13125<br />

Sartorius, Ute 3562, 3603,<br />

e14043<br />

Sarvadikar, Ajit 4553<br />

Sasada, Shinji e18036,<br />

e18042<br />

Sasada, Tetsuro e13046,<br />

e13050<br />

Sasajima, Yuko 5085<br />

Sasaki, Clarence 5529, 5532<br />

Sasaki, Fumiaki 9577<br />

Sasaki, Hidefumi 7510, 7532<br />

Sasaki, Jiichiro 7045,<br />

e13072, e18059<br />

Sasaki, Katsuhiro 8094<br />

Sasaki, Koji 8040<br />

Sasaki, Masaoki 585<br />

Sasaki, Miyuki e18134<br />

Sasaki, Naoki 5037, e15579<br />

Sasaki, Ryohei e16034<br />

Sasaki, Shinichi e18019<br />

Sasaki, Takaaki e18135<br />

Sasaki, Toru 10557<br />

Sasaki, Yasutsuna 3084,<br />

3088^<br />

Sasako, Mitsuru 4012<br />

Sasane, Medha 10094,<br />

e16537, e20511<br />

Sasano, Hironobu e11530<br />

Sasatomi, Teruo e14077<br />

Saseen, Joseph e16541<br />

Saskin, Refik 1123<br />

Sassa, Naoto e15068<br />

Sasse, Andre Deeke e14150<br />

Sassi, Salem 2530<br />

Sassolas, Bruno 1566, 8601<br />

Sastre, Javier CRA3503,<br />

3571, 4119, e14147<br />

Sastre, Xavier e15524,<br />

e15535<br />

Satchithananda, Keshthra<br />

e21137<br />

Satele, Daniel 6133<br />

Sathaiah, Magesh e14184<br />

Sathya, Abhinay 6130<br />

Sathya, Jinka 4509<br />

Sathyanarayanan, Sriram<br />

3531, 3587, 4054<br />

Sato, Atsushi 4035<br />

Sato, Ayuko 7080<br />

Sato, Fumiaki e21031<br />

Sato, Kaori e19586<br />

Sato, Kazuhiko TPS659,<br />

e11007<br />

Sato, Keiko e19557<br />

Sato, Keita e18127<br />

Sato, Kenichi 7046<br />

Sato, Koichi e14139<br />

Sato, Masayo e16537<br />

Sato, Ryo e18059<br />

Sato, Takami 8560, e19016<br />

Sato, Takashi e18058<br />

Sato, Tomomi e11060<br />

Sato, Toshihiko 3524, 3607<br />

Sato, Tsuyoshi e14004<br />

Satoh, Hiroyuki 9569<br />

Satoh, Toyomi 5006<br />

Satomi, Ryousuke e18058<br />

Satou, Mototaka e15065<br />

Satouchi, Miyako 7003,<br />

7070, 7521, e18060,<br />

e18134<br />

Satpute, Shailesh R. 4534<br />

Satram-Hoang, Sacha 6007<br />

Satre, Jose Manuel 4587<br />

Sattler, Christopher A. 4009<br />

Satwani, Prakash 6537<br />

Sauer, Annette 6566<br />

Sauerland, Maria Cristina<br />

6610<br />

Saunders, Ed 1545<br />

Saunders, Mark P. e14015<br />

Saura, Cristina 508, 509,<br />

566, 619, 3014, 3021,<br />

10511<br />

Saura, Ryuichi e17020<br />

Saura, Salvador e18015<br />

Saussele, Susanne 6510<br />

Sausville, Edward A. 6611<br />

Sauter, Craig Steven 2008,<br />

2089<br />

Sauter, Guido 10501<br />

Sauter, Matthias 4632<br />

Sautiere, Jean-Loup 568^<br />

Sauve, Anthony e13545<br />

Sava, Teodoro e15160<br />

Savage, Dionne T 4055<br />

Savage, Kerry J. 8060<br />

Savage, Sharon H 5563<br />

Savarino, Graziana e21065<br />

Savas, Burhan e14070<br />

Savas, Sevtap 7586<br />

Savignoni, Alexia 9538<br />

Savin, Michael A. 3069<br />

Savin, Michael 3590<br />

Savini, Agnese e21070<br />

Savion, Naftaly 2556,<br />

e13080<br />

Savoldo, Barbara 2558<br />

Savona, Michael R. TPS6636<br />

Savona, Steven R. TPS5601<br />

Savulsky, Claudio Isaac 2552<br />

Savva-Bordalo, Joana e15532<br />

Savvoukidis, Theodoros<br />

e14707<br />

Sawa, Toshiyuki 7017<br />

Sawada, Satoshi e14500<br />

Sawada, Takashi 3079<br />

Sawaki, Akira e14510<br />

Sawaya, Raymond 2019<br />

Sawitzki, Birgit e15053<br />

Sawyer, Emma 1545<br />

Sawyer, Michael B. 3504<br />

Sax, Cornelia 2071<br />

Saxena, Ashish e18029<br />

Saxena, P.U. Prakash 5526<br />

Saxena, Renu e17000<br />

Saxman, Scott 7502<br />

Saxton, Jerrold P. e14611,<br />

e14665<br />

Saya, Hideyuki 635<br />

Sayar, Hamid TPS6637<br />

Saydam, Serdar e11507<br />

Sayer, Herbert G e15026<br />

Saylor, Philip James 4566<br />

Sbar, Eric TPS2615<br />

Sbenghe, Maria Magdalena<br />

e19016<br />

Sbidian, Emilie 5554<br />

Scafati, Chiara e18023,<br />

e18101<br />

Scagliotti, Giorgio V. 3579,<br />

7549, 7551, 7599<br />

Scagliotti, Giorgio TPS7109,<br />

TPS7619<br />

Scala, Stefania e11052,<br />

e13539<br />

Scaletta, Giuseppe 5093,<br />

5094, e15547, e15550,<br />

e15551, e15553<br />

Scambia, Giovanni LBA5003,<br />

5059, e13095^, e13097^<br />

Scanni, Alberto LBA7501<br />

Scapulatempo Neto,<br />

Cristovam e14599<br />

Scardina, Angela 3020<br />

Scardino, Peter T. e15196<br />

Scarlattei, Maura e18096<br />

Scarpa, Aldo 4076, e14042,<br />

e14625<br />

Scarpelli, Giovanni e19038<br />

Scarpi, Emanuela 1099,<br />

10615, e11054, e17546<br />

Scartozzi, Mario e14040,<br />

e14086, e14561, e14663,<br />

e15074<br />

Scelzi, Elvira e19595<br />

Scepura, Barbara 7033<br />

Schaapveld, Michael 8039<br />

Schache, Andrew 5543<br />

Schacherer, Christopher W<br />

8536<br />

Schachter, Jacob 8517<br />

Schackert, Hans K. 3550<br />

Schad, Arno TPS4140<br />

Schadendorf, Dirk 8501,<br />

8505, LBA8509, 8511,<br />

8518, 8526, 8532<br />

Schaefer, Claus 4072<br />

Schaefer, Fritz Werner 1084,<br />

e13503<br />

Schaefer, Inga-Marie e21128<br />

Schaefer-Hesterberg, Gregor<br />

8535<br />

Schafhausen, Philippe 5516^,<br />

6511<br />

Schairer, Catherine 1521<br />

Schalke, Berthold 7105<br />

Schall, Thomas J. 2043,<br />

e13580<br />

Schallier, Denis C. C. 4622<br />

Schally, Andrew V e15153<br />

Schalper, Kurt 573<br />

Schapira, Lidia 1122, 9071,<br />

9100<br />

Scharl, Anton J. 2560<br />

Schatlo, Bawarjan 2068<br />

Schattner, Mark TPS4144<br />

Schaub, Betty 9564<br />

Schaub, Richard 2500^<br />

Schauer, Dominic e14099<br />

Schauer, Matthias e14527<br />

Schechter, Geraldine P. 8103<br />

Scheel, Birgit 2573^<br />

Scheeren, Ferenc 2514<br />

Scheetz, Janet S. e17002<br />

Scheff, Ronald J. e18029<br />

Scheffler, Matthias 3044,<br />

TPS3115, 7603, 10526<br />

Scheffold, Christian 4513<br />

Schefter, Tracey E. 4062<br />

Scheier, Benjamin 7526<br />

Scheithauer, Bernd W. 2008b<br />

Scheithauer, Werner<br />

LBA3500^, e14681<br />

Schell, Michael J. 2544,<br />

2559, e21155<br />

Schellenberg, Devin e14537<br />

Schellens, Jan H. M. 2540,<br />

2550, 2596, e13028,<br />

e13596<br />

Schellens, Jan HM e13594,<br />

e13598<br />

Scheller, Herbert 5051<br />

Schellhammer, Paul F. 4,<br />

4571, 4648<br />

Schelman, William R. 3101,<br />

3103, e13601, e14554<br />

Schem, Christian 1084,<br />

e13503<br />

Schenk, Marcus 4539<br />

Schenk, Nora TPS6640<br />

783s


Schenkein, David P. 6606<br />

Schepp, Wolfgang 4072<br />

Scher, Howard I. 4513,<br />

4516, 4517, LBA4518,<br />

4519^, 4548, 4558, 4562,<br />

TPS4689, TPS4697,<br />

TPS4700<br />

Scherer, Stefan TPS3635^,<br />

10581<br />

Scherer-Rath, Michael<br />

e19560<br />

Scherle, Peggy A 2500^<br />

Schernhammer, Eva S 1556<br />

Scherpereel, Arnaud<br />

TPS7112, e18006, e18066<br />

Schetelig, Johannes 6584<br />

Scheulen, Max E. 2086,<br />

8532, 10032^<br />

Schiano De Colella,<br />

Jean-Marc 8026<br />

Schiantarelli, Clara 8046<br />

Schiavo, Roberta 3570<br />

Schiavon, Gaia 10063,<br />

10091, e14135<br />

Schiavone, Paola e11001,<br />

e11547<br />

Schiff, Bradley A. e16058<br />

Schiff, David 2006, 2031,<br />

TPS2107<br />

Schiff, Susan 6035<br />

Schiffer, Charles Alan 6506<br />

Schiffman, Joshua David<br />

9525, 9561<br />

Schild, Steven E. 6108,<br />

7073, 7605<br />

Schilder, Jeanne e15561<br />

Schilder, Russell J. 3029<br />

Schilder, Russell 5021<br />

Schildgen, Oliver e18000<br />

Schildgen, Verena e18000<br />

Schildhaus, Hans-Ulrich 1533<br />

Schiller, Gary J. TPS6637<br />

Schiller, Joan H. 7516, 7589<br />

Schilling, Joerg Peter e19540<br />

Schilsky, Richard L. e13106<br />

Schimanski, Carl Christoph<br />

TPS3641^<br />

Schimmoller, Frauke 8531<br />

Schimp, Veronica L. 5012<br />

Schindlbeck, Christian 1600^<br />

Schindler, Fernanda e19552<br />

Schink, Julian C. 1553,<br />

5039, 6120, e15523<br />

Schinzari, Giovanni e16553<br />

Schipper, Joerg e16033<br />

Schipper, Matthew J e14603<br />

Schirm, Sabine 3590<br />

Schirmacher, Peter e18016^<br />

Schirripa, Marta 3518, 3540,<br />

3546, 3549, 3555, 3585,<br />

3597, e13057, e14040<br />

Schirrmacher-Memmel, Silke<br />

4539<br />

Schlaak, Max 3044, e19031<br />

Schlag, Peter M. 3508<br />

Schlag, Rudolf 4065<br />

Schlattmann, Peter TPS4138<br />

Schlegel, Mariette 10596<br />

Schlegel, Uwe S. 2040, 8031<br />

Schleicher, Jan e15026<br />

Schlesinger, Andreas 1533,<br />

10526, CRA10529<br />

Schlichting, Michael 3562,<br />

3574, 3591, e14043<br />

Schlom, Jeffrey 2526, 2577,<br />

TPS2617, TPS3638,<br />

TPS4701<br />

Schloms, Anna 9094<br />

Schlossman, Robert L. 8012<br />

Schlumberger, Martin 5508,<br />

5518, 5569, 5591<br />

Schmalenberg, Harald 4080,<br />

4083, 4090, e21063<br />

Schmalfeldt, Barbara 5001,<br />

5005, e13100^<br />

Schmandt, Rosemarie e13516<br />

Schmeler, Kathleen M. 5027<br />

Schmelz, Hans U. 4530<br />

Schmelzel, Mark e16513<br />

Schmid, Jasmin e12031<br />

Schmid, Martina e19032<br />

Schmid, Peter 624<br />

Schmid, Roland M. 4034<br />

Schmid-Bindert, Gerald 7554<br />

Schmidinger, Manuela<br />

e15083, e15090, e19502<br />

Schmidt, Brian L. e16052<br />

Schmidt, C. Max e21048,<br />

e21121<br />

Schmidt, Emmett V. 3531<br />

Schmidt, Henrik 8545<br />

Schmidt, Joachim 10080<br />

Schmidt, Lindsay A. 2602<br />

Schmidt, Manuel 4636,<br />

e14152<br />

Schmidt, Marcus 542, 556,<br />

640, 1077, 10551<br />

Schmidt, Mary Lou 9533<br />

Schmidt, Rodney 5515<br />

Schmidt-Hieber, Martin<br />

e18552<br />

Schmidt-Wolf, Ingo GH 4636<br />

Schmiegel, Wolff H. 3550<br />

Schmier, Johann Wilhelm<br />

10040<br />

Schmitt, Anita 6567<br />

Schmitt, Michael 6567<br />

Schmitt, Nicola 6527<br />

Schmitt, Thomas 10040,<br />

10041^<br />

Schmitt-Bormann, Beate<br />

TPS4139<br />

Schmitz, Arndt 10536,<br />

10537, e21017<br />

Schmitz, Joerg e15044<br />

Schmitz, Kathryn TPS669<br />

Schmitz, Norbert 6543,<br />

8004, 8022, 8024, 8025<br />

Schmitz, Sandra 5519<br />

Schmitz, Shu-Fang 9059<br />

Schmitz, Stephan H. 1533,<br />

10526, CRA10529<br />

Schmoll, Hans-Joachim 3522,<br />

3523, 3525, 3578,<br />

e14152, e19579<br />

Schmucker-Von Koch, Jospeh<br />

6002, 7607<br />

Schmulewitz, Nathan e14714<br />

Schmutzler, Andreas Gerd<br />

1084<br />

Schnabel, Catherine A. 561,<br />

10530, 10588, e21019<br />

Schnadig, Ian D. 3024,<br />

e15089<br />

Schneebaum, Schlomo 8534<br />

Schneeweiss, Andreas 554,<br />

1059, 1077, 1090, 1096,<br />

TPS1146, 1600^, 1602,<br />

10540, e13095^, e13097^<br />

Schneider, Anneliese 4034<br />

Schneider, Bryan J. 7546<br />

Schneider, Bryan P. 6025,<br />

7032, 7107, 7108<br />

Schneider, Charles e18122<br />

Schneider, Coursen 4517<br />

Schneider, Guenther 4107<br />

Schneider, Jeffrey Gary<br />

e21114<br />

Schneider, Lars 8505<br />

Schneider, Robert e11064,<br />

e11529<br />

Schneider, Vanessa 6567<br />

Schneider-Kappus, Wolfgang<br />

4065<br />

Schneiderman, Rosa S.<br />

e14616<br />

Schnell, Patrick 7598<br />

Schnell, Roland 1533, 3044,<br />

10526<br />

Schneller, Folker 2573^<br />

Schnittger, Susanne 6510,<br />

6610<br />

Schoen, Michael TPS3641^<br />

Schoenegg, Winfried 640,<br />

1077<br />

Schoenewolf, Nicola L 8563<br />

Schoengen, Alfred 8535<br />

Schoettle, Glenn P e17531^<br />

Sch<strong>of</strong>fski, Patrick 535, 1020,<br />

2540, 4622, 5508,<br />

LBA10008, 10009, 10013,<br />

10091<br />

Sch<strong>of</strong>ield, Penelope 6111<br />

Sch<strong>of</strong>ield, Penny e18068<br />

Scholl, Suzy Marie Elisabeth<br />

e15535<br />

Scholz, Christian Winfried<br />

8031<br />

Scholz, Christoph 1072,<br />

1600^, 1602<br />

Scholz, Hans-Jorg e15112<br />

Schoor, Oliver 2522, 3530<br />

Schorb, Elisabeth 2040<br />

Schore, Reuven J. 9543<br />

Schorge, John O. 5029,<br />

e15545, e15568<br />

Schori, Clara e16055<br />

Schorno, Kevin e13568<br />

Schott, Roland e18005<br />

Schott, Sarah 1090<br />

Schotzinger, Robert J 4671,<br />

e15167<br />

Schouten, Viviane 6535^<br />

Schpero, William Lewis 6013<br />

Schrader, Iris 554, 1077<br />

Schrader, Mark 4530<br />

Schrag, Deborah 3537, 6003,<br />

9047, e19586<br />

Schraml, Peter 2055<br />

Schreeder, Marshall T.<br />

6518^, 8032<br />

Schreier, Jörg 1600^<br />

Schriner, Megan 9110<br />

Schroder, Henrik 9590<br />

Schroeder, Brock e21019<br />

Schroeder, Jan 640<br />

Schroeder, Sebastian Edoardo<br />

Artur 8575<br />

Schroeder, Wendy e21152<br />

Schroeder, Willibald 5005<br />

Schroers, Roland 8031<br />

Schr<strong>of</strong>, Inga 5051<br />

Schr<strong>of</strong>f, Matthias 4636,<br />

e14152<br />

Schrope, Beth e14708<br />

Schroth, Gary P. 10539<br />

Schroyens, Wilfried A. 2506<br />

Schrump, David S. 7103<br />

Schubert, Erin K 1088,<br />

TPS3109<br />

Schubert, Joerg 8022<br />

Schubert, Jörg 6543<br />

Schuchert, Matthew J. 7071,<br />

7074<br />

Schuchter, Lynn Mara 3102,<br />

8503^, 8534, 8567<br />

Schueller, Katharina 10540<br />

Schueneman, Aaron e14585<br />

Schuette, Kerstin TPS4148<br />

Schuette, Wolfgang TPS7109,<br />

7554, e18016^<br />

Schuetz, Florian 1090, 1096<br />

Schuetze, Scott 10003,<br />

10028<br />

Schuh, Anna 6584<br />

Schuhmacher, Christoph<br />

4095^<br />

Schuler, Gerold e19032<br />

Schuler, Markus Kajo 10068<br />

Schuler, Martin H. 2086,<br />

LBA7500, 7557, 7558,<br />

10032^, 10046, e13605<br />

Schuler-Thurner, Beatrice<br />

e19032<br />

Schulick, Richard D. 3596,<br />

e13559<br />

Schully, Sheri D 3567<br />

Schulman, Kevin A. 1525,<br />

6047, 6051<br />

Schulmann, Karsten 3550,<br />

4083<br />

Schulte, John e19058<br />

Schulte, Nathan 7534<br />

Schulte, Wendy e21090<br />

Schultes, Birgit Corinna 4056<br />

Schultheis, Beate e13597<br />

Schultz, Christopher J. 2001<br />

Schultz, Johan 6557<br />

Schultz, Kris Ann<br />

Pinekenstein 9521, 9522<br />

Schulz, Christoph 4072<br />

Schulz, Holger CRA10529,<br />

e13100^<br />

Schulz, Stephanie e14619<br />

Schulz, Ulrike 1084<br />

Schulze, Mathias 3588<br />

Schumacher, Claudia 1077<br />

Schumann, Christian 7531<br />

Schupp, Clayton 6050<br />

Schuster, Stephen J. e18529,<br />

e18540<br />

Schuster, Tibor 10012<br />

Schusterbauer, Claudia 3010<br />

Schutt, Tessa e15010<br />

Schutz, Fabio A. B. 4577,<br />

4580<br />

Schutz, Fabio Augusto Barros<br />

4543, 4582, 4585, 4635,<br />

e15007<br />

Schuur, Eric e21022<br />

Schwaab, Thomas e15039,<br />

e15042, e15046, e15204<br />

Schwachula, Tim 5053<br />

Schwarb, Heike 10033<br />

Schwarer, Anthony P. 6505^<br />

Schwartz, Ann G. 6038,<br />

7037, 7063, 7593<br />

Schwartz, Antonia e21128<br />

Schwartz, Benjamin M 5062^<br />

Schwartz, Brian E. 4006,<br />

4117, 4545, 8519<br />

Schwartz, Cindy L. 9503,<br />

9524<br />

Schwartz, David L. TPS5601,<br />

e16026<br />

Schwartz, Edward L e18113<br />

Schwartz, Erich 10574<br />

Schwartz, Gary K. 8549,<br />

8575, 8598, 10002,<br />

10003, 10004, 10019,<br />

TPS10099, TPS10103,<br />

e13588, e13600^, e14586<br />

Schwartz, Gary e21125<br />

Schwartz, Jonathan D.<br />

TPS4675^, 5012<br />

784s Numerals refer to abstract number


Schwartz, Kendra L. 6023,<br />

e15147<br />

Schwartz, Kenneth A. 2056<br />

Schwartz, Lawrence H. 2541,<br />

4507, e13064, e19647<br />

Schwartz, Marc D 1608<br />

Schwartz, Myron E. 4033,<br />

e14729, e14732<br />

Schwartz, Peter E. TPS1614,<br />

5099, e13125, e15526,<br />

e15581<br />

Schwartzberg, Alison e21077<br />

Schwartzberg, Lee Steven<br />

1016, TPS1137, 2569,<br />

3085, 7002, 7502,<br />

e19511, e21077<br />

Schwarz, Donald E. 10037<br />

Schwarz, Luis Jesus 2097,<br />

e11518, e15036<br />

Schwed, Daniel 7077<br />

Schwed-Lustgarten, Daniel<br />

7092<br />

Schwedler, Kathrin 556<br />

Schweizer, Charles 7030<br />

Schwentner, Christian e19021<br />

Schwentner, Lukas 1078,<br />

1082<br />

Schwiep, Frances 10094,<br />

e20511<br />

Schwock, Joerg e14535<br />

Schäfer, Henning S e13515<br />

Schäfer, Reinhold e21005<br />

Schöder, Heiko 4517<br />

Schöffski, Patrick 3018^,<br />

10030, 10047<br />

Schönemann, Constanze<br />

e15053<br />

Schønnemann, Katrine<br />

Rahbek e14517, e14633<br />

Sciacca, Dorotea e18002<br />

Sciamanna, Chris 7055<br />

Sciandivasci, Angela 546,<br />

1049, 1115<br />

Sciot, Raf 9536, 10030<br />

Sclafani, Francesco 615<br />

Scoazec, Jean-Yves 4129<br />

Scola, Michael A. 10530,<br />

e21023<br />

Scorilas, Andreas 5574,<br />

10606<br />

Scott, Alastair 3516<br />

Scott, Amie 9042, 9043<br />

Scott, Andrew Mark 8547,<br />

e13054, e15056<br />

Scott, Brian J. 2052<br />

Scott, Charles B. e19606<br />

Scott, Clare L. 5073<br />

Scott, Jeffrey A. e16540,<br />

e16552<br />

Scott, Richard W 9119<br />

Scott, Sasinya N. 4604<br />

Scotte, Florian e19640<br />

Scotto, Luigi e13569<br />

Scriboni, Sonia 9098<br />

Sculier, Jean Paul e18006<br />

Scurr, Michelle R. 10039,<br />

10060, 10086<br />

Scurr, Michelle 10038<br />

Seah, Davinia Shien 6089<br />

Seal, Brian S. e14013,<br />

e14016, e14035, e14056,<br />

e14106, e14141, e14146,<br />

e14522, e14626, e14638,<br />

e15146, e15148, e15175,<br />

e15183, e18097<br />

Sears, Alan K. 625, 2508,<br />

2529<br />

Seay, Thomas E. TPS657<br />

Numerals refer to abstract number<br />

Sebag-Montefiore, David<br />

4004, 4029<br />

Sebastian, Lucille T 7079^<br />

Sebastian, Martin 2573^<br />

Sebastian, Nikhil 9544<br />

Sebban, Catherine 8068<br />

Sebe, Philippe e15105<br />

Sebesta, Christian e14087<br />

Sebio, Ana e14104<br />

Sebrow, Dov e15164<br />

Sebti, Rani e19641<br />

Seckl, Michael 2608, e13033<br />

Secord, Angeles Alvarez 5101<br />

Secreto, Charla e13517<br />

Sedda, Tito e14094<br />

Sederias, Joana 2051<br />

Sedlacek, Scot M. 547<br />

Sedlmaier, Benedikt 5512<br />

Sedova, Michaela 4081<br />

See, Lionel e18528<br />

See, Siewju 2066<br />

Seedhouse, Claire 1014<br />

Seeger, Grant R. 9001<br />

Seeh<strong>of</strong>er, Daniel TPS3641^<br />

Seery, Virginia Jean 8516<br />

Seetalarom, Kasan TPS7614<br />

Seetharam, Raviraja e14019<br />

Seetharam, Shobha 3059<br />

Seetharamu, Nagashree<br />

e16052, e18139<br />

Seftel, Matthew D. 8002<br />

Segal, Michal e14586<br />

Segal, Neil Howard 4057,<br />

9105<br />

Segalla, J.G.M. 5017^, 7512<br />

Segati, Romana e13019<br />

Segawa, Yoshihiko 7042,<br />

7560<br />

Segelov, Eva e13054<br />

Seggewiss-Bernhardt, Ruth<br />

2504<br />

Seghers, Amelie Clementine<br />

e19026, e19027<br />

Segnan, Nereo 1500<br />

Segni, Jessica 6123<br />

Seguí-Palmer, Miguel Angel<br />

1001, 10500, 10616<br />

Seguin, Carole TPS1139<br />

Segura Gonzalez, Eduardo<br />

Raul e20513<br />

Segura, Miguel F. 8550,<br />

8565<br />

Sehl, Mary Elizabeth e11003<br />

Sehn, Laurie Helen 8049<br />

Sehouli, Jalid 5031, 5034,<br />

5044, 5047, 5053, 5058,<br />

5064, 5070, 5071, 5080,<br />

5087, e13095^, e13097^,<br />

e13100^, e13108^,<br />

e15520, e15531, e19558,<br />

e19597<br />

Sehovic, Marina 6100<br />

Seibers, Nicholas e13041<br />

Seidel, Christoph 4631<br />

Seidensticker, Max e14630<br />

Seider, Michael J. TPS9150<br />

Seidman, Andrew David<br />

1016, TPS1145<br />

Seif, Alix Eden 1504<br />

Seifert, Michael 514<br />

Seijo, Luis Miguel e18008<br />

Seimetz, Diane 2584<br />

Seisler, Drew K. 9075<br />

Seiter, Karen 6520<br />

Seitz, Gerhard 10554<br />

Seitz, Jean Francois 4087,<br />

4091<br />

Seitz, Jean-François 4036<br />

Seitz, Robert S. 516<br />

Seiwert, Tanguy Y. 5500,<br />

5517<br />

Seke, Mihalj LBA4512, 4551<br />

Sekeres, Mikkael A. 6521,<br />

6522<br />

Seki, Nobuhiko 1530,<br />

e18099<br />

Sekijima, Masaru e21031<br />

Sekine, Ikuo 7070<br />

Sekulic, Aleksandar 8579<br />

Selbach, Johannes 640<br />

Selby, Brian 4519^<br />

Selby, George Basil e17010<br />

Selby, Peter John 6091<br />

Selby, Rick e14679<br />

Selchuk, Vladimir Yur’evich<br />

e19005<br />

Selcukbiricik, Fatih e21143<br />

Selder, Sonja 1092<br />

Seligman, Mark e11042<br />

Selinger, Cristina 504<br />

Selistre, Simone Geiger<br />

10593<br />

Sella, Avishay 4564, 4619,<br />

e15058<br />

Sella, Tal 1507, 1564<br />

Sellami, Dalila B. 3099<br />

Selle, Frédéric 5017^, 5018,<br />

5067<br />

Sellecchia, Rita 5591<br />

Selleslag, Dominik L. D.<br />

TPS6637<br />

Selva, Julio Cesar 2515<br />

Selvarajah, Shamini 4585<br />

Semba, Hiroshi e13072<br />

Semenova, Anna Igorevna<br />

e13059<br />

Semiglazova, Tatyana 9046<br />

Semlani, Neha TPS3112<br />

Semmes, Oliver John e15113<br />

Sempoux, Christine e14044,<br />

e14060<br />

Semrad, Thomas John 3566<br />

Sen, Fatma e14628<br />

Sen, Urmi e11522<br />

Sena, Susana e15570<br />

Senan, Suresh 7009<br />

Senapedis, William 1055,<br />

e13549<br />

Sencan, Orhan 3565<br />

Sendrup, Sarah Louise 2565<br />

Sendur, Mehmet Ali Nahit<br />

e11009, e11033, e11093,<br />

e13031<br />

Senellart, Helene e13010<br />

Sener Dede, Didem e11011<br />

Sener, Nur 638<br />

Senff, Tina 10552<br />

Seng Yue, Corinne 6619^<br />

Seng, Sonia Maria e21016<br />

Sengar, Manju e17003<br />

Sengelov, Lisa TPS4696^<br />

Sengeløv, Lisa e15115<br />

Sengupta, Sandip 1123<br />

Sengupta, Saubhik e13030<br />

Senju, Hiroaki 7569<br />

Sensi, Elisa 3521, 3585<br />

Senzer, Neil N. 3024, 3029,<br />

LBA3501, e13040, e14546<br />

Seo, Youngho e21026<br />

Seoane, J, e15552<br />

Seoane, Joan 2042, 10511<br />

Seol, Miee 6564<br />

Seon, Ben K. 3034, 3042^<br />

Seow, Hsien 6039<br />

Sepkowitz, Kent 6101<br />

Sepucha, Karen 6025<br />

Sepulveda, Antonia 4111<br />

Sepúlveda, Juan Manuel<br />

2042<br />

Sepulveda, Juan 4584, 4620,<br />

e15149<br />

Sequist, Lecia V. 7010,<br />

LBA7500, 7509, 7524,<br />

7525, 7532, 7543, 7546,<br />

7547, 7556<br />

Serafini, Aldo N. e13592<br />

Serdengecti, Suheyla e21143<br />

Sereni, Maria Isabella e15549<br />

Sereno, Maria 1570, 9073,<br />

e11028, e18015<br />

Sergeev, Urii e15029<br />

Sergi, Concetta e18026<br />

Serke, Monika Heidi 1533,<br />

TPS7109, 10526<br />

Serke, Monika 7575,<br />

CRA10529<br />

Seronde, Audrey e14129<br />

Serpe, Roberto e19634<br />

Serpico, Danila 3014<br />

Serra, Javier 3586<br />

Serra, Luigi 10601<br />

Serra, Olbia e18030<br />

Serra, Patrizia 10601, 10615<br />

Serra, Stefano 4109,<br />

e14524, e14535, e14592<br />

Serrano, Carole 9560<br />

Serrano, Davide 519, 1500<br />

Serrano, Gloria 5520,<br />

e14147, e15094, e18069,<br />

e18106, e21015<br />

Serrano, M. Jose 10608<br />

Serrano, Sergio Vicente<br />

e14599<br />

Serrano, Silvia J e18114<br />

Sertoli, Mario Roberto 8576<br />

Seruga, Bostjan 558, e16505<br />

Servent, Véronique e11012,<br />

e11070<br />

Servitja, Sonia e11545<br />

Servois, Vincent 8546<br />

Serwatowski, Piotr 7090<br />

Sesikeran, Nanditha 9055<br />

Sessa, Cristiana 3003, 3048,<br />

3080, TPS5112<br />

Seth, Tulika e17000<br />

Sethi, Seema 1560, e21057<br />

Seto, Takashi 3045, 7012,<br />

7521, 7602<br />

Sette, Giovanni e13512<br />

Settle, Stephen H 8584<br />

Settleman, Jeff 5575<br />

Setton, Jeremy 5537<br />

Seufferlein, Thomas<br />

TPS1612, TPS3636, 4034,<br />

e14564<br />

Sevestre, Marie-Antoinette<br />

1580<br />

Sevilla, Isabel 4119, e14115,<br />

e14671<br />

Sevin, Emmanuel CRA4502,<br />

5018, 9079<br />

Sevinc, Alper e19024<br />

Sexton, Rachel 3078<br />

Seymour, Lesley 2547, 7007,<br />

7093, 7511, 10602,<br />

e18079<br />

Seymour, Matthew T.<br />

TPS4143, 6091, e16007<br />

Seynaeve, Caroline M. 516,<br />

517, 526, 1080, 10504,<br />

10518<br />

Sferruzza, Anthony 5510,<br />

8596<br />

Sfiniadakis, Ioannis e21098<br />

Sgarbossa, Gigliola e19595<br />

Sgroi, Dennis 561<br />

785s


Sha, Dan 3564<br />

Sha, Huifang e13528<br />

Shaaban, Hamid e12025<br />

Shacham, Sharon 1055,<br />

4634, e13549, e13586,<br />

e15200<br />

Shada, Amber L 8530, 8597<br />

Shadduck, Richard K. 6620<br />

Shadman, Mazyar 1527<br />

Shaeffer, David e14632<br />

Shafa, Bob B. 2101<br />

Shafey, Mona 6548<br />

Shaffer, Thomas 6028, 6060,<br />

6075<br />

Shah, Amit 1128<br />

Shah, Bijal D. 6568, 6608,<br />

e18503, e18506, e21123<br />

Shah, Bijal Dinesh e18509<br />

Shah, Binay Kumar e17005<br />

Shah, Dhruvil R e19001,<br />

e19013<br />

Shah, Gaurav D. 4632,<br />

10619<br />

Shah, Jatin J. 8037, 8093,<br />

e18556<br />

Shah, Jatin P. 5545^, 5562<br />

Shah, Kajal e13068<br />

Shah, Katherine Sanvidge<br />

e18553<br />

Shah, Krunal Jitendrakumar<br />

2530, 3030<br />

Shah, Manali e13531<br />

Shah, Manan 8036<br />

Shah, Manish A. 4061,<br />

e14586<br />

Shah, Manisha H. 4047,<br />

4126, 5508, 5518, 5591,<br />

e14542, e14551<br />

Shah, Mazhar Ali e16043<br />

Shah, Mira e15512<br />

Shah, Monjri 5036<br />

Shah, Neil P. 6504, 6506<br />

Shah, Nina 6546, 6551,<br />

8040, 8102, 9081, 9082<br />

Shah, <strong>Part</strong>h K. e15124<br />

Shah, Payal D. 1506<br />

Shah, Preeti 10025<br />

Shah, Raj J. 4055<br />

Shah, Raj e14632<br />

Shah, Riyaz N.H. LBA7500,<br />

7583<br />

Shah, Ruchita R. 1028, 1130<br />

Shah, Shetal N. TPS4684<br />

Shah, Shilpa Rashmikant<br />

e15213<br />

Shah, Trushil e17021<br />

Shah, Vallabh 1558<br />

Shaha, Ashok R. 5545^<br />

Shahabi, Vafa 8593,<br />

TPS8603<br />

Shahan, Mark N. TPS3113<br />

Shaheen, Mahmoud e20508<br />

Shaheen, Montaser F. 3009<br />

Shahid, Tanweer e18043<br />

Shahidi, Mehdi LBA7500<br />

Shahinian, Vahakn B. e15147<br />

Shaik, Mohammed saifullah<br />

3619, e14046<br />

Shaikh, Arif 6040<br />

Shaikh, Asif 3572<br />

Shak, Steven 552, 1005,<br />

1021, 3512<br />

Shakes, LaShaina e13588<br />

Shalgi, Ruth 2556, e13080<br />

Shamash, Jonathan 4529,<br />

4531<br />

Shamberger, Robert C. 9537<br />

Shames, David S. 5575<br />

Shammo, Jamile M. 6577<br />

Shamonki, Jamie 1105<br />

Shan, Minghua LBA4512,<br />

4551, e18100<br />

Shan, Wen Wen 7020<br />

Shanafelt, Tait D. 6571,<br />

e13517<br />

Shanahan, Marie e19588<br />

Shang, Aijing TPS9596<br />

Shang, Shulian 8550, 8557<br />

Shang, Yuqin 628<br />

Shani, Adi e14143<br />

Shankar, Anjali e11097,<br />

e12004<br />

Shankar, Lalitha 10602<br />

Shankaran, Veena 6077,<br />

e14068<br />

Shannon, Keith A. e13599<br />

Shannon, Kevin 6582<br />

Shao, Guoliang e14605<br />

Shao, Minhua e13528<br />

Shao, Qian e13524, e14735<br />

Shao, Yongzhao 8550, 8557,<br />

8574, e19003<br />

Shao, Zhimin 531, e13036<br />

Shapira, Iuliana 1046<br />

Shapira-Frommer, Ronnie<br />

5022<br />

Shapiro, Charles L. 609,<br />

1007, CRA9013<br />

Shapiro, Daniela e15210<br />

Shapiro, Ge<strong>of</strong>frey 2566,<br />

3028, 3039, 3053, 3062,<br />

TPS3118, 7508, 8531,<br />

e13598<br />

Shapiro, Irina M 1057<br />

Shapiro, Jeremy David 3504,<br />

3515, 3572, TPS4692^,<br />

e15047<br />

Shapiro, Lauren Quinn 5537<br />

Shapiro, Marc A. e18046<br />

Shapiro, Richard L. 8550,<br />

8557, 8565, e19003<br />

Shapiro, Richard 8589<br />

Shapiro, Roman 6043<br />

Sharabi-Nov, Adi e14025<br />

Sharan, Krishna 5526<br />

Sharawat, Surender Kumar<br />

9549, 9551, 9580<br />

Sharif, Saima 3512<br />

Sharifi, Nima 4645<br />

Sharkey, Robert M. 3091<br />

Sharma, Anand K. 5503<br />

Sharma, Atul 5540, 6593,<br />

e18518, e18573<br />

Sharma, Dayanand 2072,<br />

e15518<br />

Sharma, Dhruv B 6003<br />

Sharma, Hari 6600<br />

Sharma, Kamal 6616,<br />

e18500<br />

Sharma, Kamalesh 4645<br />

Sharma, Manish e13106<br />

Sharma, Mehar C e17505,<br />

e18518<br />

Sharma, Nancy e14600,<br />

e14620, e15143<br />

Sharma, Navesh K 3590<br />

Sharma, Neelesh e17563<br />

Sharma, Om Prakash e16012<br />

Sharma, Padmanee 2520^,<br />

TPS4673, TPS4691,<br />

e15092<br />

Sharma, Poonam e17549<br />

Sharma, Priyanka 9000<br />

Sharma, Puza P. e11067<br />

Sharma, Raj Govind e12014<br />

Sharma, Rameshwaram<br />

e16012<br />

Sharma, Ricky A e13587<br />

Sharma, Rohini e21093<br />

Sharma, Shantanu e16012<br />

Sharma, Shelly 9527<br />

Sharma, Sonam e18032<br />

Sharma, Sunil 3007, 3057,<br />

7029, 10549, 10605,<br />

e13548, e14090, e14177,<br />

e15115<br />

Sharma, Vipra e16565<br />

Sharma, Vivek R. e21072,<br />

e21144, e21149<br />

Sharman, Jeff Porter 3069,<br />

6122, 6507, 6518^, 8032<br />

Sharp, Linda 521, 2599,<br />

6036, e13096, e16514<br />

Sharpe, Dunbar 2589<br />

Sharpe, Rowena TPS10633^<br />

Sharples, Katrina e15054<br />

Sharvashidze, Ineza O.<br />

e11086<br />

Shasha, Daniel e15138<br />

Shashidhar, H P e12030<br />

Shashidhara, P e11511<br />

Shastry, Mythili 618<br />

Shaughnessy, John D.<br />

e18548<br />

Shaw, Alice Tsang 3007,<br />

7010, 7503, 7508, 7533,<br />

7589, 7596, 7599, 7600<br />

Shaw, Ashley 4121<br />

Shaw, Edward G. 2008b,<br />

2047, 9108<br />

Shaw, Emily TPS10633^<br />

Shaw, James Edward e19551<br />

Shaw, Mary e19647<br />

Shaw, Richard 5543, e16042<br />

Shaw, Robin J. e12027<br />

Shaw, Sally 1035<br />

Shawver, Laura K. e15555<br />

Shaya, Fadia T. e14522,<br />

e14626, e14638<br />

Shazer, Ronald L. TPS8113<br />

Shea, S<strong>of</strong>ia M 8530<br />

Shea, Thomas C. e13074<br />

Sheahan, Kieran e13570,<br />

e21024<br />

Shechter, Sharon 1055,<br />

e15200<br />

Shedden, Kerby A e17530<br />

Sheehan, Christine E 3533<br />

Sheehan, Jonas e11544<br />

Sheehan, Margaret e14687<br />

Sheehy, Niall 8552<br />

Sheehy, Patricia S 8042,<br />

8107, e18575<br />

Sheehy, Patricia 8043<br />

Sheehy, Patrick Frank 2569<br />

Sheeran, Lisa 9124<br />

Sheets, Nathan Christopher<br />

e15184<br />

Shehata, Christina 518<br />

Sheikh, Arif 5050<br />

Sheikh, Nadeem A. 2563,<br />

4650<br />

Sheikh, S. 3089^<br />

Sheikh-Salah, Moktar 6095,<br />

6128<br />

Sheinfeld, Joel 4532<br />

Shekar, Mamatha e21159<br />

Sheldon, John M. e14632<br />

Sheldon, Lisa Kennedy<br />

e19515<br />

Shelkey, Julia 7040, e15121<br />

Shell, Scott A. e21120<br />

Shellenberger, Thomas D.<br />

e16030<br />

Shelton, Joseph e15006<br />

Shen, Chan e14550<br />

Shen, Changyu e21099<br />

Shen, Dong 503, e12503<br />

Shen, Hong e14026<br />

Shen, Jie 4068, e14509,<br />

e21034<br />

Shen, John P. e14069<br />

Shen, Li TPS4687<br />

Shen, Liji TPS4696^<br />

Shen, Lin 3040, 4081,<br />

e14122, e14604<br />

Shen, Ronglai 5545^<br />

Shen, Steven S. 4586,<br />

e15003<br />

Shen, Wei 1068, 7075,<br />

e18138<br />

Shen, Xian 2532, 4063<br />

Shen, Xiaodong 535, 4007,<br />

7514, 8531<br />

Shen, Yi e18013<br />

Shen, Yufeng 10516<br />

Shen, Yun TPS2614,<br />

TPS2615<br />

Shen, Zhenbin 4092, e14575<br />

Sheng, Xi Nan 8506, 8561,<br />

8562, e15051, e15052<br />

Shenkier, Tamara Nina 549^,<br />

582<br />

Shenolikar, Rahul 6060<br />

Shenouda, George TPS5600<br />

Shenoy, Aditya e18068<br />

Shepard, Harold Michael<br />

2579<br />

Shepard, Nancy 9107<br />

Shepherd, Frances A. 1535,<br />

1583, TPS2615, LBA7000,<br />

7007, 7093, 7511, 7586,<br />

10522, e18079<br />

Shepherd, Lois E. 500, 501,<br />

524, 538, 561, 1008,<br />

e11005<br />

Shepherd, Scott e14110<br />

Shepherd, Stacie Peacock<br />

TPS1138, 3049, 3054<br />

Sheppard, Karen E 8520<br />

Sher, David J 5579, 5582<br />

Sher, David 7067<br />

Sher, Tamara G. 6117<br />

Sherer, Stefan 10512, 10595<br />

Sheridan, Elizabeth 4553<br />

Sherman, Christopher G.<br />

4613<br />

Sherman, David H. e12026<br />

Sherman, Eric Jeffrey 5509^,<br />

5514, 5537, 5545^<br />

Sherman, Laurie Jill LBA8509<br />

Sherman, Marion E. 1598<br />

Sherman, Mark E 1519,<br />

10603<br />

Sherman, Matthew L. 607,<br />

e15170, e19609<br />

Sherman, Morris 4033<br />

Sherman, Stephen L. e16510<br />

Sherman, Steven A e14028,<br />

e14030<br />

Sherman, Steven I. TPS3117,<br />

5508, 5518, 5547, 5591<br />

Sherman, William H. e14708<br />

Shermock, Kenneth M.<br />

e15175, e15183<br />

Sherrill, Gary Bradley e13109<br />

Sherwood, Anna 6631<br />

Sherwood, James 1548<br />

Shet, Tanuja 1108<br />

Shete, Jitendra e16002<br />

Sheth, Pulin 1124<br />

Shethia, Maithili A 4037<br />

Shetty, Aditya V. 4615<br />

Shetty, P. 3089^<br />

Shi, Hongliang 2597<br />

Shi, Lehua 4008<br />

786s Numerals refer to abstract number


Shi, Li e18073<br />

Shi, Lizheng 6600, e15130<br />

SHI, Meiqi 7548<br />

Shi, Michael 2543, TPS4147,<br />

TPS4683<br />

SHI, Peipei 3001<br />

Shi, Qian 3526, 3564, 3576,<br />

3617, 4009, 4075<br />

Shi, Rong e15529<br />

Shi, Runhua e14667,<br />

e16009<br />

Shi, Shan Rong 4575<br />

Shi, Shu-zhen e14668<br />

Shi, Ting 2014<br />

Shi, Weiji 5537<br />

Shi, Wenyin e19507<br />

Shi, Xiaojin 3045<br />

Shi, Yi e18116<br />

Shi, Yuanjun TPS4693<br />

Shi, Yuankai 1534, 7533,<br />

7559, 7596, e14606<br />

Shia, Jinru 3583<br />

Shiau, Chung-Wai e14516<br />

Shibata, Atsushi e15098<br />

Shibata, Henry R. LBA506<br />

Shibata, Kazuhiko 7521<br />

Shibata, Stephen 2538,<br />

3108, e14182, e14586<br />

Shibata, Taro 5006, 7003,<br />

7017, 7028<br />

Shibayama, Takuo 7560<br />

Shibuta, Kenji 588<br />

Shibuya, Kazutoshi e18127<br />

Shibuya, Masahiko e18099<br />

Shichijo, Shigeki e13046,<br />

e13050<br />

Shields, Anthony Frank<br />

e14163, e14519, e14609<br />

Shields, Carol L 8560<br />

Shields, Jerry A 8560<br />

Shields, Lisa B.E. 2058<br />

Shields, Mario A. e14515<br />

Shiels, Meredith S. 6070<br />

Shien, Kazuhiko e17517<br />

Shifrin, Alexander L. 1540<br />

Shiga, Kiyoto e16020<br />

Shigaki, Hironobu 10523<br />

Shigematsu, Yoshiki 10585<br />

Shigeoka, Yasushi TPS659,<br />

10073<br />

Shih, Jen-Fu e18132<br />

Shih, Kent C. 3006^, 4027,<br />

4127, 7567, 7587, 8556<br />

Shih, Le-Ming e14538<br />

Shih, Tiffany Ting-Fang 1079<br />

Shih, Ya-Chen T. e14549,<br />

e14550<br />

Shiiba, Kenichi e14011<br />

Shiina, Shuichiro e14622<br />

Shilkrut, Mark e14603<br />

Shim, Byoung Yong e14072<br />

Shim, Hyun-Jeong e14649<br />

Shim, Hyunsuk 3055<br />

Shim, Joomee e15140<br />

Shima, James e21159<br />

Shimabukuro, Kelly Anne<br />

e14069, e14078, e14079<br />

Shimada, Andrea Kazumi<br />

e11014<br />

Shimada, Hiroyuki 9533,<br />

9534, 9564<br />

Shimada, Kazuaki e14506<br />

Shimada, Ken e14123<br />

Shimada, Masaaki e14596<br />

Shimada, Mitsuo 3558<br />

Shimada, Muneaki 5103<br />

Shimada, Takanobu e16034<br />

Shimada, Temiko e18060,<br />

e18134<br />

Numerals refer to abstract number<br />

Shimada, Yasuhiro 3524,<br />

3569, e14126<br />

Shimamoto, Takashi 8029<br />

Shimamura, Hiromune<br />

e14011<br />

Shimamura, Takeshi e14047<br />

Shimizu, Chikako 10519,<br />

e11065, e19535<br />

Shimizu, Eiji 7521<br />

Shimizu, Junichi e17510,<br />

e18145<br />

Shimizu, Kazuo 1112<br />

Shimizu, Kazuyuki 8097<br />

Shimizu, Kimihiro 7012<br />

Shimizu, Seiichi 8094<br />

Shimizu, Shigeki e18126<br />

Shimizu, Toshiki e18081<br />

Shimizu, Toshio e13025<br />

Shimizu, Yasuhiro e14540<br />

Shimizu, Yasushi e17520<br />

Shimko, Sara 6616, e18500<br />

Shimodaira, Hideki 4002,<br />

e19611<br />

Shimokawa, Mototsugu 3558<br />

Shimokawa, Tsuneo e18099<br />

Shimokuni, Tatsushi 10578<br />

Shimoyama, Tatsu TPS3642<br />

Shimozaki, Shingo 10075<br />

Shin Ogawa, Luisa 3086<br />

Shin, Dong Bok TPS4142,<br />

e14137, e14685<br />

Shin, Dong Moon 2576,<br />

3094, e13110<br />

Shin, Dong M 5560, 5570,<br />

e21161<br />

Shin, Dongbok e14572<br />

Shin, Eun Soon 7089,<br />

e13065<br />

Shin, Hee Jung e11508<br />

Shin, Hee-Chul 1056, 1133,<br />

e11532, e21160<br />

Shin, Hyun Soo e18501<br />

Shin, Im-Hee 3606<br />

Shin, Jae-Gook 3592<br />

Shin, James e18147<br />

Shin, Ji Hoon 4093<br />

Shin, Jung-Young e18050<br />

Shin, Sang Joon 3606,<br />

e14072, e14081, e14084<br />

Shin, Soyoung TPS2621<br />

Shin, Sung Kwan e14559<br />

Shinagawa, Atsushi 8094<br />

Shinde, Arvind Manohar 1035<br />

Shingoji, Masato e21054<br />

Shinkai, Tetsu 6112, 7017,<br />

7028, 7549, 7576,<br />

e18099<br />

Shinkura, Nobuhiko e21031<br />

Shinoda, Kazunobu 9569<br />

Shinohara, Nobuo e15068,<br />

e15071, e15098, e15127<br />

Shinohara, Takeshi e19583<br />

Shinozaki, Eiji 10541,<br />

e14038, e14145<br />

Shintani, Satoru 5584<br />

Shio, Yutaka e13015<br />

Shiokawa, Rie 3022<br />

Shiono, Masatoshi e19611<br />

Shioyama, Yoshiyuki 7045<br />

Shiozawa, Toshihiro e18064<br />

Shipe-Spotloe, Janice 8533,<br />

8547<br />

Shipley, Dianna 7100, 7587<br />

Shipley, Janet 9510<br />

Shirabe, Ken e14536,<br />

e14602<br />

Shirafuji, Naoki 9053<br />

Shirai, Takao e13029<br />

Shirai, Toshiharu 10075,<br />

e20505<br />

Shirane, Masatoshi e18064<br />

Shirar, Kristen Leigh 7107<br />

Shirar, Kristen L 7106, 7108<br />

Shirasaki, Ryosuke 9053<br />

Shiroki, Ryoichi e15068<br />

Shirouzu, Kazuo e13046,<br />

e13050, e14077<br />

Shiroyama, Takayuki e18141<br />

Shirvani, Shervin Mohajer<br />

7062<br />

Shitara, Kohei 4067, e14500<br />

Shivakumar, Swamy e14675<br />

Shivakumar, Swarna e21132<br />

Shivapurkar, Narayan e14009<br />

Shmueli, Einat 1507<br />

Shockley, Nancy 8560<br />

Shockro, Laura 9084<br />

Shoemaker, J. Samantha<br />

6060, 6075<br />

Shohet, Jason 9534<br />

Shoji, Tadahiro 5103<br />

Shoni, Melina 5011<br />

Shook, Stephanie TPS9150<br />

Shore, Neal D. 4510, 4642,<br />

4662, TPS4698^<br />

Shore, Neal TPS4697<br />

Shores, Carol G. 5539<br />

Shouery, Marwan 9042,<br />

e19647<br />

Shouldis, Jennfier 7077<br />

Shourbaji, Rania Ayman<br />

5531, 5559, 5581,<br />

e15138<br />

Shovlin, Margaret 8051<br />

Showalter, Timothy Norman<br />

e11505, e15012, e15163,<br />

e19507<br />

Shows, Joseph Ryan e14667<br />

Shpall, Elizabeth J. 6529<br />

Shparyk, Yaroslav e15037<br />

Shrager, Joseph B 7008<br />

Shreenivas, Aditya V. e16060<br />

Shrivastav, S. 3089^<br />

Shrivastava, Shyam K<br />

TPS5116<br />

Shr<strong>of</strong>f, Rachna T. 4051,<br />

4054<br />

Shtalrid, Mordechai e19529<br />

Shtivelband, Mikhail 7512<br />

Shu, Hui-Kuo George 3055<br />

Shu, Jihong e16011<br />

Shu, Sei e13502<br />

Shu, Yongqian 9017<br />

Shuai, Yongli 8533<br />

Shubhakar, Poornima 3009<br />

Shuda, Masahiro 8577<br />

Shukla, N. K e11013,<br />

e17505<br />

Shukuya, Takehito 7088,<br />

e21007<br />

Shulkes, Arthur e15117<br />

Shulman, Katherina 1578<br />

Shulman, Lawrence N. 1021,<br />

e19503<br />

Shumak, Rene e12034<br />

Shuman, Andrew G 9121<br />

Shumway, Nathan M. 625,<br />

2508, 2529, 9058<br />

Shuping, Jo e21066<br />

Shurell, Elizabeth 10000<br />

Shurin, Galina e21001<br />

Shurin, Michael e21001<br />

Shuryak, Igor e15162,<br />

e17562<br />

Shustov, Andrei R. 8070<br />

Shvidel, Lev e19529<br />

Shyr, Yu 6038<br />

Si, Lu 8506, 8561, 8562,<br />

e15051, e15052<br />

SI, Yongfeng 5558<br />

Siakavellas, Spiros e21098<br />

Sica, Gabriel 7097<br />

Sicchieri, Renata D 1075<br />

Sicklick, Jason Keith e14069<br />

Siddiqi, Rabia 9565<br />

Siddiqi, Tanya 6547<br />

Siddique, Mohammad Hasan<br />

6612<br />

Siddiqui, Bilal Karim 7107<br />

Siddiqui, Javed e15205<br />

Siddiqui, Jehan 3504<br />

Sideras, Kostandinos 1083,<br />

10509<br />

Sideri, Mario e19036<br />

Sidhom, Mark TPS4690<br />

Sidhu, Gurinder Singh<br />

e15097, e18504<br />

Sidhu, Roger 3535<br />

Sidoni, Angelo e21094<br />

Sieber, Markus TPS3639<br />

Sieber, Oliver 3623<br />

Sieber, Paul 4510, 4550<br />

Siebert, Fred W e21111<br />

Siedlecki, Janusz A 8548,<br />

10538, e19022<br />

Siefker-Radtke, Arlene O.<br />

4506, 4523<br />

Siegal, Gene P. e13087<br />

Siegel, Abby B. 4101<br />

Siegel, Cara e14112<br />

Siegel, David Samuel DiCapua<br />

8016, 8035<br />

Siegel, Petra 8535<br />

Siegel, Robert D. 9047,<br />

e16532<br />

Siegel, Robert S. e11084<br />

Siegel, Samantha 9578<br />

Siegfried, Anne 8518<br />

Siegfried, Jill 7071, 7074,<br />

e21012<br />

Siegmann, Katja C. 1067<br />

Siemionow, Vlodek 1076<br />

Siena, Salvatore 3502, 3570,<br />

3581, TPS3637, 5513,<br />

7550, TPS9155, e13095^,<br />

e13097^<br />

Sierocinski, Thomas 4565<br />

Sierra, Enrique 3586<br />

Sierra, Marta Isabel e18044<br />

Sieuwerts, Anieta M 10504<br />

Sifuentes, Erika e12027<br />

Siggillino, Annamaria<br />

e18023, e18101, e21094<br />

Signorelli, Carlo e18065<br />

Signoretti, Sabina 4543,<br />

4582, 4585, 4635<br />

Signorovitch, James E. 4612,<br />

e14621<br />

Sigurdson, Elin R. 3605,<br />

6061<br />

Sikora, Katarzyna Otylia 3511<br />

Sikov, William M. 1034,<br />

1045, 10508<br />

Silasi, Dan-Arin e15526<br />

Silber, Jeffrey H. 6000<br />

Silberstein, Peter T. e14641,<br />

e16554<br />

Silcocks, Paul TPS8605<br />

Silginer, Manuela 2055<br />

Silletta, Marianna 10063<br />

Silliman, Rebecca A e11003<br />

Silling, Gerda 6610, 8004<br />

Silva, Alejandro J. e15584<br />

Silva, Antonio P. 508<br />

Silva, Eric TPS6637<br />

Silva, Gualdino e14673<br />

787s


Silva, Jose D. 5567<br />

Silva, Milton Jose B e14680,<br />

e14728<br />

Silva, Rosa Rita e15074<br />

Silvain, Christine 3520<br />

Silver, Bruce A. 8032<br />

Silver, Joel 6555, e18543<br />

Silver, Richard T. 6624<br />

Silverman, Craig I. 5530<br />

Silverman, Lewis B. 9530<br />

Silverman, Michael H.<br />

e14731<br />

Silverman, Paul e15212<br />

Silverman, Paula TPS655<br />

Silvestri, Alessandra e14151<br />

Silvestri, Rosangela e11071<br />

Silvestris, Nicola 4627<br />

Sima, Camelia S. 1541,<br />

7014, 7052, 7066, 7527,<br />

7580, e19647<br />

Simcock, Richard 5527<br />

Simeone, Diane M. 4591<br />

Simeone, Ester 8513, 8529,<br />

8573, e19034<br />

Simeonova, Anna 5596^<br />

Simes, John 3504, 3515,<br />

3534, 3572, 5081,<br />

e13007<br />

Simi, Paolo e13057<br />

Simko, Jeffrey 2564<br />

Simkova, Iveta 9592<br />

Simmonds, Peter 511<br />

Simmons, Christine E. 6084,<br />

9111<br />

Simms, Lorinda 7502<br />

Simon, George R. 2544,<br />

2559, TPS7619, e18112<br />

Simon, Helene e11019<br />

Simon, Iris 3510, e21013<br />

Simon, Jordi Rovira TPS9154<br />

Simon, Laurence e11542<br />

Simon, Marike Anna e19610<br />

Simon, Matthias 2000<br />

Simon, Michael S. e14005<br />

Simon, Noah e15005<br />

Simon, Richard 2578<br />

Simon, Ronald 10501<br />

Simon, Sergio D. TPS661,<br />

e12021<br />

Simon, Wolfgang 1072, 1081<br />

Simoncini, Edda 645<br />

Simone, Charles B. 7098,<br />

e17534<br />

Simone, Nicole L. e11505<br />

Simonelli, Matteo 2580,<br />

4117, e21139<br />

Simonoska, Rusana 5536<br />

Simonson, Rachel 8516<br />

Simpkins, Fiona 5065<br />

Simpson, Dayna e13028<br />

Simpson, Jennifer 6545,<br />

e18542<br />

Simpson, Pamela S. e18122<br />

Sims, Robert Brownell 2564,<br />

4648, 4650, TPS4673<br />

Sin, VC 5511<br />

Sina, Sokol e14647<br />

Sinclair, Natalie Faye 1045,<br />

10508<br />

Singal, Amit G e14165<br />

Singavi, Arun e18507,<br />

e19641<br />

Singer, Barak e14731<br />

Singer, Christian F. 514,<br />

1537, 10501<br />

Singer, Eric A. TPS4680<br />

Singer, Samuel 10001,<br />

10002, 10015, 10019<br />

Singer, Susanne e19610<br />

Singh Riyat, Malkit e20504<br />

Singh, Abhijai e14186<br />

Singh, Akshita e11552<br />

Singh, Amar e17511<br />

Singh, Arun D 8071<br />

Singh, Arun S. 3015<br />

Singh, Baljit e11064<br />

Singh, Balraj e21107<br />

Singh, Dharam Pal e16012<br />

Singh, Harpreet 2522, 3530<br />

Singh, Harvinder 9103<br />

Singh, Hitesh e11069<br />

Singh, J. K. 3089^<br />

Singh, Jasmeet Chadha<br />

e11529<br />

Singh, Jaspreet e15012<br />

Singh, Jaswinder TPS3640,<br />

e14632<br />

Singh, Juswinder 8032<br />

Singh, Madhu Sudan e16562<br />

Singh, Mamta 9544<br />

Singh, Mandeep 4106<br />

Singh, Navneet e18009<br />

Singh, Nishith K. 2526,<br />

TPS2617, TPS4701<br />

Singh, Onkar 2598<br />

Singh, Pankaj R 628<br />

Singh, Preet Paul 6614<br />

Singh, Rajkumar Bikramjit<br />

5104<br />

Singh, Randeep 9559<br />

Singh, Sharat 4011, 10535,<br />

e14584, e21017<br />

Singh, Shivendra TPS8606<br />

Singh, Simron 5502<br />

Singh, T. Trevor e15554<br />

Singh, T.Trevor 3619<br />

Singh, Veena 10588, e21019<br />

Singh, Zeba e18576<br />

Singhal, Anil 8020,<br />

TPS8112, TPS8113,<br />

TPS8114<br />

Singhal, Jyotsana e13557<br />

Singhal, Nimit 4531,<br />

e17544, e19513<br />

Singhal, Sandeep K. e21010<br />

Singhal, Seema 6623, 8035<br />

Singhal, Sharad S. e13557<br />

Sinha, Ishan e19030<br />

Sini, Claudio 7577, e21065<br />

Sini, MariaCristina 8581<br />

Sinibaldi, Victoria J. 4564,<br />

4619, e15058, e15188<br />

Sinicrope, Frank A. 3514,<br />

3564, 3617, 4009, 4075<br />

Sinicrope, Pamela S 3567<br />

Sinn, Bruno Valentin 522,<br />

523, 4020<br />

Sinn, Hans-Peter 1096<br />

Sinn, Marianne 4020, 4044,<br />

e14533<br />

Siolos, Spyros 5574<br />

Sipkins, Dorothy 6562<br />

Sipos, Laszlo 2<br />

Siqueira, Sheila Aparecida<br />

Coelho e15562<br />

Siracusano, Licia 7061,<br />

7585, e18104<br />

Siraj, Muhammad Ahsan<br />

e21138<br />

Sire, Christian 5505<br />

Sirera, Rafael 7058, 7060,<br />

10594<br />

Siritanaratkul, Noppadol 8097<br />

Siriwardana, Melany e18505<br />

Sirtaine, Nicolas 507<br />

Sirulnik, L. Andres 6514^,<br />

6625^, 6626^, TPS6642^<br />

Sirvent, Nicolas e20006<br />

Sisani, Michele e15185<br />

Sismondi, Piero 627<br />

Sisti, Valerio 4084<br />

Sit, Laura 9104, e19633<br />

Sitarik, Mark A. 6109<br />

Sittampalam, Kesavan 10051,<br />

e13061, e20509<br />

Siu, Lillian L. 3027, 3077,<br />

3082, 3101, 3504, 3572,<br />

5587, TPS5600, e13001,<br />

e13107, e13603<br />

Siu, Lillian e19616<br />

Siu, Steven 4105<br />

Sivanand, Sharanya 4616<br />

Sivapalasuntharam, Asha<br />

e18068<br />

Sivarajan, Kulumani M<br />

TPS3635^<br />

Siveke, Jens T 4115<br />

Sixtova, Dimka e18062<br />

Siziopikou, Kalliopi P.<br />

TPS657, TPS1134<br />

Sizoo, Eefje 2071<br />

Sjoquist, Katrin Marie<br />

TPS3643, TPS4137, 5081<br />

Skaar, Todd C. 525<br />

Skaarup, Jesika e11517<br />

Skailes, Geraldine 7562<br />

Skapek, Stephen 1576, 9535<br />

Skarlos, Dimosthenis V. 573<br />

Skarlos, Dimosthenis-Vasilios<br />

e15040<br />

Skeel, Roland T. e14098<br />

Skiadaresis, Julia 9076<br />

Skibber, John Michael 3556<br />

Skikne, Barry 6522<br />

Skiles, Jodi L 2601<br />

Skinner, Donald G 4575<br />

Skinner, Eila C. 4571, 4576,<br />

4588, 4589<br />

Skinner, Eila C 4575, 4578<br />

Skinner, William Kempton<br />

e16058<br />

Skitzki, Jospeh J. e14172,<br />

e18537, e18539, e19041<br />

Skladowski, Krzyszt<strong>of</strong> A.<br />

5504^<br />

Sklar, Charles A 9514<br />

Sklar, Larry A. TPS5602<br />

Sklarin, Nancy T. 10514<br />

Skokan, Margaret 7061,<br />

7534, 10580<br />

Skol, Andrew e15086<br />

Skolnik, Jeffrey e13599<br />

Skoog, Lambert 4561<br />

Skovlund, Eva 3548<br />

Skrickova, Jana e18062<br />

Skrzypski, Marcin Tomasz<br />

10561<br />

Skuse, David e19581<br />

Slack, James L. 8053<br />

Slaets, Leen 1022<br />

Slager, Susan L 6571<br />

Slamon, Dennis J. 8592<br />

Slater, Sarah LBA4000<br />

Slaughter, James e15582<br />

Slavkovic, Vesna e13066<br />

Sleckman, Bethany G. 4022<br />

Sledge, George W. 505, 605,<br />

1005, 1027, 6025, 6053,<br />

9020, e21099<br />

Sleer, Leanne Sophia 2517<br />

Sleigtholm, Richard 6561<br />

Sleijfer, Stefan 2593, 2596,<br />

10091, 10504<br />

Slepushkina, Nadejda e21011<br />

Slichenmyer, William J. 4501<br />

Slingluff, Craig L. 8530,<br />

8597, e19028, e19062<br />

Sliwkowski, Mark X. 5575<br />

Sloan, Andrew E. TPS1616,<br />

TPS2107<br />

Sloan, Brittany e13032<br />

Sloan, David e16035<br />

Sloan, Jeff A. 1557, 1610,<br />

3617, 6108, 6133, 9001,<br />

9047, e11051, e16572<br />

Sloan, Philip 5543<br />

Sloane, Robert e15141<br />

Slomovitz, Brian M. 5027<br />

Slone, Jeremy 9558<br />

Slotman, Ben J. 7009<br />

Slovin, Susan F. 4548,<br />

TPS4700<br />

Sluiter, Wim J. 10581<br />

Small, Alexander C. 4623,<br />

TPS4676, 6065, e21032<br />

Small, Art e11077<br />

Small, David I. e18061,<br />

e18098<br />

Small, David e19626<br />

Small, Eric Jay 4, 2564,<br />

4513, 4515, LBA4518,<br />

4592, 4593, 4655, 4660,<br />

4664, 4667, e15137<br />

Small, Isabele Avila 7506,<br />

e14027<br />

Small, William 1069, 4049,<br />

TPS5116<br />

Smalley, Robert J e15565<br />

Smallridge, Robert C. 5544<br />

Smart, James 8566<br />

Smeland, Sigbjorn 10081<br />

Smetzer, Leslie e13025<br />

Smid, Marcel 10504<br />

Smirnov, Denis A e21146<br />

Smit, Egbert F 7009, 7543,<br />

7607, e18094<br />

Smith, Alan D. e14532<br />

Smith, B. Douglas 6629<br />

Smith, Barbara L. 1122<br />

Smith, Benjamin D. 6124,<br />

7062<br />

Smith, Brian 3056, 8061<br />

Smith, Cardinale B. 6065,<br />

7064<br />

Smith, Carly J 9511<br />

Smith, Carmen J. 6034<br />

Smith, Carrie L. e15557<br />

Smith, Catrin Tudur 8523<br />

Smith, Christopher G 3516<br />

Smith, Clare e21147<br />

Smith, Cornelia e14689<br />

Smith, Daryn W. 1560,<br />

e15081, e17528<br />

Smith, Daryn 5534<br />

Smith, David A. TPS1143,<br />

2516, 3023, TPS3111,<br />

e18047, e18063^<br />

Smith, David C. CRA2509,<br />

3025, 3077, 4505, 4506,<br />

4513, e13603<br />

Smith, David M 6137<br />

Smith, David LBA4000,<br />

TPS4143, e14587<br />

Smith, Deborah A. 610,<br />

e13596<br />

Smith, Denis 3506, 4036,<br />

4118, e14168<br />

Smith, Dennis M. e21048,<br />

e21121<br />

Smith, Dennis Wesley e19509<br />

Smith, Donald 3090<br />

Smith, Donna M. e13121<br />

Smith, Ellen M. Lavoie<br />

CRA9013<br />

Smith, Emma-Louise e19550<br />

Smith, Frederick P. 7587<br />

788s Numerals refer to abstract number


Smith, Garrett TPS4145<br />

Smith, Gary L 2606<br />

Smith, Heind 4081<br />

Smith, Ian C. 7503, e13599<br />

Smith, Ian Edward 596, 604<br />

Smith, Jennifer A e19571<br />

Smith, Jillian K 4059<br />

Smith, Judith A. 5041<br />

Smith, Julia Anne e11010,<br />

e11529<br />

Smith, Ken R 9525, 9561<br />

Smith, Kristin e20001<br />

Smith, L. Mary 9576<br />

Smith, Larry R e19058<br />

Smith, Lauren A 8574<br />

Smith, Lauren B 9121<br />

Smith, Lon S. 2555, e13025,<br />

e13602, e13604<br />

Smith, Lon 2512<br />

Smith, Lynette 9501, e17549<br />

Smith, Mary Lou 6053<br />

Smith, Matthew Raymond<br />

4510, 4513, LBA4518,<br />

4566, 4642, 4644, 6045<br />

Smith, Milton T e14714<br />

Smith, Mitchell R. 8081<br />

Smith, Mitchell Reed<br />

e13079, e19580<br />

Smith, Myles JF 10050<br />

Smith, Nick e14587<br />

Smith, Pamela Ann e13536<br />

Smith, Paul D. 7503, e13599<br />

Smith, Paul 1581<br />

Smith, Richard V. e16058<br />

Smith, Robin e19644<br />

Smith, Scott E. 6582,<br />

e13079<br />

Smith, Sonali M. 8060,<br />

8072, 8075<br />

Smith, Stephen James<br />

e14503<br />

Smith, Susan C. 6516^<br />

Smith, Thomas J. 9075,<br />

e19564^<br />

Smith, Thomas J e18144<br />

Smith, Timothy 6017, 6029<br />

Smith, Webb A 9526<br />

Smith-Machnow, Victoria<br />

TPS3641^<br />

Smith-Marrone, Stephanie<br />

5514<br />

Smithers, Bernard Mark<br />

TPS4145, 8559<br />

Smithers, Mark TPS4141,<br />

8534<br />

Smits, Evelien L. 2506<br />

Smits, Evelien e13051<br />

Smitt, Melanie 532<br />

Smolej, Lukas 6584<br />

Smolenski, Kathy N. 2018<br />

Smolkin, Mark E 8530<br />

Smoll, Nicolas Roydon 2068<br />

Smorenburg, Carolien H.<br />

1080<br />

Smorodinsky, Nechama<br />

e13018<br />

Smylie, Michael 8539<br />

Smyrk, Thomas C. 3514<br />

Smyth, Elizabeth C TPS4143<br />

Smyth, Emily Nash 1068,<br />

7101, e18138<br />

Smyth, Fiona Elizabeth 8547,<br />

e15056<br />

Smyth, Lillian e19601<br />

Snedecor, Sonya J. e15189<br />

Sng, Natasha J 7523<br />

Snider, Jessica N. TPS1137<br />

Sninsky, John J. 10586<br />

Numerals refer to abstract number<br />

Snitcovsky, Igor M. L. e21040<br />

Snook, Adam e14619<br />

Snowdon, Jaime 5032<br />

Snuderl, Matija 2010<br />

Snyder, Carrie 1538<br />

Snyder, Claire Frances 6026,<br />

6028, 6080, 9103<br />

Snyder, David 1592<br />

So, Ping Fai Peter TPS7614<br />

So, Sam e14037<br />

So, Tomoko 10585<br />

Soares, Fernando Augusto<br />

1522<br />

Soares, Fernando A e20506<br />

Sobecks, Ronald M. 8055<br />

Sobol, Hagay e12502<br />

Sobol, Joseph TPS9147<br />

Sobol, Robert W. TPS1138<br />

Sobrero, Alberto F. 3502,<br />

3535, 4042<br />

Socci, Nicholas D 4527<br />

Socinski, Mark A. LBA4,<br />

LBA7000, 7513, 7590,<br />

7592, TPS7618<br />

Soe, Aye Min e13099<br />

Soehrman Brossard, Sophia<br />

5522<br />

Soejima, Kenzo e18058<br />

Soejima, Yuji e14536,<br />

e14602<br />

Soeling, Ulrike 554<br />

Soerensen, Bente Thornfeldt<br />

7001<br />

Soerensen, Flemming Brandt<br />

3573<br />

Soergel, Fritz 3044<br />

Soetekouw, Patricia M.M.B.<br />

e13028, e15115<br />

S<strong>of</strong>fietti, Riccardo 2037,<br />

2075<br />

Soh, I Peng Thomas 1064,<br />

e13002, e19523<br />

Soh, Junichi e17517<br />

Soh, Lay-Tin 10051, e20509<br />

Sohal, Davendra 4111,<br />

e13602<br />

Sohji, Sunao e15212<br />

Sohl, Jessica 6089<br />

Sohn, Byeong Seok 10093<br />

Sohn, Christ<strong>of</strong> 1090, 1096<br />

Sohn, Claudine 6633<br />

Sohn, Joo Hyuk 1003, 2542<br />

Sohn, Sang Kyun 3554,<br />

6536, 6538, e14074,<br />

e18533<br />

Soini, Barbara e14711<br />

Sokol, Lubomir 2099, 6608,<br />

8084, e18503, e18506,<br />

e18509, e21123<br />

Sokol, Lubosh 6568<br />

Solak, Mustafa e11020,<br />

e11092<br />

Solassol, Jerome 10507<br />

Solberg, Arne 4508<br />

Soldi, Raffaella 3057,<br />

e14090<br />

Solé, Claudio J. e14002<br />

Sole, Claudio Vicente e14002<br />

Solé, Juan M. e14002<br />

Solé, Sebastián e14002<br />

Solé, Vicente e14002<br />

Soler, Christine e20006<br />

Soler, Claude e15109,<br />

e21056<br />

Solima, Eugenio 5096<br />

Soliman, Hatem Hussein<br />

1585, 2501, TPS2620<br />

Soliman, Pamela T. 1513,<br />

5027<br />

Solin, Lawrence J. 1005<br />

Solis, Luisa 10506<br />

Solis-Trapala, Ivonne 5527<br />

Solit, David B. 4527, 5030<br />

Solleza, Franco 10061<br />

Solomayer, Erich 10599<br />

Solomayer, Erich-Franz 1090,<br />

TPS1146, 1523<br />

Solomon, Benjamin J. 7508,<br />

7511, 7596, 7600, 7601,<br />

e13054<br />

Solomon, Benjamin Maurice<br />

6571<br />

Solomon, James A e19048^<br />

Solomon, Maria Teresa 2515<br />

Som, Peter M. 5578<br />

Somaiah, Neeta TPS10099,<br />

e18112, e19580<br />

Somani, Naresh 3011<br />

Somashekar, Mohan Kumar<br />

e11536<br />

Somashekhar, S. P. e14710,<br />

e15516<br />

Sombroek, Cherita 8039<br />

Somer, Robert A. CRA1002,<br />

e12008<br />

Somers, Elizabeth Brooke<br />

e21028<br />

Somlo, George 1010, 1035,<br />

1070, 1592, 3108, 8035,<br />

8038, 10545, 10597,<br />

e21013<br />

Sommer, Harald Leo 554,<br />

1072, 1081, 10540,<br />

e15520<br />

Sommerfeld, Alex e14612<br />

Sommerfeld, James 9544<br />

Sommerfeldt, Silvia e14099<br />

Somorat, Bhattacharjee<br />

e11027, e14721<br />

Son, Byung Ho e11504,<br />

e11508<br />

Son, Crystal 6101<br />

Son, Jeewoong 3076, e14572<br />

Son, Seyoung 4093<br />

Sondak, Vernon K. 8504,<br />

8521, 8525, 8527, 10048<br />

Sondel, Paul M. e19047<br />

Sonderfan, Andrew J 3086<br />

Sone, Takashi e17540<br />

Sonet, Anne 8065<br />

Song, Cheryn e15080<br />

Song, Eun-Kee e14580<br />

Song, Gina 5050, e13063<br />

Song, Ho June 4093<br />

Song, Ho Young 4093<br />

Song, Hong-Suk e14570<br />

Song, Ik Chan e14529<br />

Song, Kyo Young e14558<br />

Song, Meijuan e14643<br />

Song, Paul Y. 3015, e14593<br />

Song, Si Yeol 5586, 7004<br />

Song, Tianqiang 4008<br />

Song, Weifeng e14508<br />

Song, Xiangqun 7091<br />

Song, Xinni 587<br />

Song, Yan e14668<br />

Song, Yang 10010<br />

Song, Ying e11095<br />

Song, Yiyi e12013<br />

Song, Yong 7559, e18082,<br />

e18116<br />

Song, Yong-Mao e14024<br />

Sonis, Stephen T. 9024,<br />

9119<br />

Sonja, Betschart e19648<br />

Sonke, Gabe S. 5001<br />

Sonnemans, Hetty 1129<br />

Sonnenblick, Amir 10034<br />

Sonneveld, Pieter 8095<br />

Sonoo, Hiroshi 1119<br />

Sonpavde, Guru 4522, 4524,<br />

4525, 4586, 4603, 4623,<br />

4643, 4644, TPS4675^,<br />

TPS4676, TPS4678, 6065,<br />

e15002, e15003, e21016<br />

Soo, Ross A. 2551, 3002,<br />

7512, e13002, e13061<br />

Sood, Anil 5015<br />

Sood, Suman Lata e14578<br />

Soon, Yu Yang 2016, 5553<br />

Soong, Richie Chuan Teck<br />

2551<br />

Soori, Gamini S. 1557, 3545,<br />

6133<br />

Soraya, Sami sahnoun<br />

e11034<br />

Sorbye, Halfdan 3508, 3538,<br />

3548, 4015, e14053<br />

Sorensen, A. Gregory 2009,<br />

2012, 2025, 3036<br />

Sorensen, Flemming Brandt<br />

3513<br />

Sorensen, Gregory A<br />

TPS10635<br />

Sorensen, Jens Benn 7001,<br />

10617<br />

Sorenson, Leslie 9019<br />

Sorescu, George P. e14591<br />

Soria Rivas, Ainara e16029<br />

Soria, A e19023<br />

Soria, Claudine e21156<br />

Soria, Irene e13085<br />

Soria, Jean-Charles 2539,<br />

2540, 3000, 3012, 3021,<br />

3026, 3063, 3080, 3104,<br />

4540, 4614, 4618, 5522,<br />

7007, 7529, 10556,<br />

e13118<br />

Soria, Jeannette e21156<br />

Soriano, Carmen e19576<br />

Soriano, Jordi e14120<br />

Soriano, V. e19023<br />

Sorio, Roberto LBA5002^,<br />

LBA5003<br />

Sorlini, Cristina 7544<br />

Sos, Martin TPS3115, 7532,<br />

7603<br />

Sosa, Aarón 3536<br />

Sosinska-Mielcarek, Katarzyna<br />

505<br />

Soslow, Robert A 5043<br />

Sosman, Jeffrey Alan 3102,<br />

4545, 8502^, 8503^,<br />

8507, 8504, 8510, 8519<br />

Sotiriou, Christos 507, 515,<br />

e21010<br />

Soto Parra, Hector J. e18096<br />

Soto-parra, Hector e18026<br />

Sotomayor, Eduardo M. 2099,<br />

6568, 6575, 6608, 8084,<br />

e18503, e18506, e18509,<br />

e21123<br />

Sottile, Roberta 1044,<br />

e13058, e13070<br />

Sottotetti, Federico 589<br />

Souadka, Abdelilah e14133<br />

Souadka, Amine e14133<br />

Soubeyran, Pierre-Louis 2023,<br />

9012<br />

Souhami, Luis 2008b<br />

Soulie, Michel LBA4567^<br />

Soulieres, Denis e18136,<br />

e19591<br />

Soulos, Pamela R. 1030,<br />

4651, e15150<br />

Soupos, Nikolaos 5033<br />

789s


Sousa, Carlos Eduardo Pires<br />

643<br />

Sousa, Susana e15532<br />

Soussain, Carole 2023<br />

South, Natalie L. 9575<br />

Southerland, Cindy 2074^<br />

Southey, Melissa C 1502<br />

Souza Crovador, Camila<br />

e19528, e19584, e19602<br />

Souza, Cristiano de padua<br />

e14173<br />

Souza, Sulene Cunha 606<br />

Sovak, Mika A. 5054<br />

Soveri, Leena-Maija 4015<br />

Soverini, Simona 6531<br />

Sˇ pacek, Jirí 5001<br />

Spacek, Martin 6584<br />

Spada, Daniele 4084<br />

Spadola, Giuseppe e19035,<br />

e19036<br />

Spaeth, Dominique 568^<br />

Spaeth-Schwalbe, Ernst<br />

TPS3639<br />

Spafford, Michael TPS5602<br />

Spaggiari, Lorenzo 7023<br />

Spaggiari, Paola e14672<br />

Spagnoli, Giulio C e21126<br />

Spain, Pamela 6046<br />

Span, Paul e13111<br />

Spaner, David 10622<br />

Spangenberger, Holger<br />

e13508<br />

Spannberger, Thomas 2053<br />

Spano, Jean-Philippe 641,<br />

1095, 5067, 6110, 10078,<br />

e11551, e18505<br />

Sparagana, Steven 10619<br />

Sparano, Joseph A. 1005,<br />

1010, 1021, 4030, 8005<br />

Sparano, Joseph 1027, 9020<br />

Sparreboom, Alex 2549<br />

Spasic, Jelena e12020<br />

Spatti, Gian Battista 5096<br />

Spatz, Alan e18061<br />

Speaker, Stephanie 8036<br />

Specht, Jennifer M. 1088,<br />

TPS3109, e13596<br />

Specht, Lena e18527<br />

Specht, Michelle Connolly<br />

1122<br />

Speck, Rebecca M e19531<br />

Specterman, Sergio 10074<br />

Spector, Mathew E e16014<br />

Spector, Nelson 6505^<br />

Spector, Yael 10575, e21008<br />

Speer, Tod e16010<br />

Speer, Vanessa e11556<br />

Speers, Caroline 3527, 6014,<br />

6018, e14048, e14176<br />

Speiser, Daniel E e19050<br />

Speiser, Paul 5071<br />

Spelda, Stanislav e18062<br />

Spelman, Amy 9029<br />

Spencer, Elysia Sophia 4669,<br />

e15207<br />

Spencer, Jennifer e19527<br />

Spencer, Kathy 9115<br />

Spencer, Shawn D. 4671<br />

Spencer, Shawn D 3042^<br />

Spencer, Stuart e13541<br />

Spengler, Werner 7572<br />

Speranza, Giovanna 3031,<br />

3087, 3529<br />

Sperber, Fanny 1564<br />

Sperduti, Isabella 4076,<br />

e12514, e14082, e14661,<br />

e18065, e18072<br />

Sperinde, Jeff 603, 608, 614<br />

Speyer, James L. 5016,<br />

e11064, e11529<br />

Speyer, Mark e19588<br />

Speyer, Sue TPS3638,<br />

e14501<br />

Spicer, Darcy V. 1065<br />

Spicer, James F. 3022,<br />

3100, 5522, 7544<br />

Spicka, Ivan 8018^<br />

Spickh<strong>of</strong>f, Frank 1600^<br />

Spiegel, David 9051, e14006<br />

Spiegelmann, Roberto 2078<br />

Spiekermann, Karsten 6510<br />

Spielberger, Ricardo Tomas<br />

8089<br />

Spier, Catherine M 8001<br />

Spigel, David R. LBA4,<br />

CRA2509, TPS2615, 7035,<br />

7100, TPS7113, 7509,<br />

7549, 7567, 7587,<br />

TPS7616, 10530, e13076,<br />

e13113, e21023<br />

Spillane, Donna e14111<br />

Spillane, Susan 2599<br />

Spina, Michele 8005, 8046,<br />

8058<br />

Spindler, Karen-Lise Garm<br />

3573<br />

Spinelli, Gian Paolo e11039,<br />

e11514, e12510, e19620<br />

Spinelli, Tulla e19530<br />

Spinnato, Francesca e18002<br />

Spira, Alexander I. LBA7000,<br />

e18063^<br />

Spittler, Aaron John 7107,<br />

7108<br />

Spitzer, Thomas R. 6618<br />

Spivey, Tara L 8576<br />

Spleiss, Olivia 8503^<br />

Spoelstra, Nicole S. 564<br />

Sporchia, Andrea 3570<br />

Sportelli, Peter LBA3501<br />

Sposto, Richard 1065,<br />

e19506<br />

Spradlin, Natalie M. 6068<br />

Spraggs, Colin F. 10532,<br />

e15059<br />

Sprangers, Miriam 6002<br />

Spranklin, Laura Jane e17506<br />

Spratlin, Jennifer L. 3013<br />

Sprauten, Mette 4637<br />

Sprecher, Emmet 10558<br />

Sprenger, Thilo 3600,<br />

e14161, e14162, e14167<br />

Spriggs, David R. 2586, 5043<br />

Springett, Gregory M. 4043<br />

Springfield, Sanya 6086<br />

Sprod, Lisa 9009, 9010,<br />

9028, 9141, 9142<br />

Sprott, Kam M. 1091,<br />

e21146<br />

Spruyt, Odette 2604<br />

Spry, Nigel TPS4145,<br />

TPS4690<br />

Spunt, Sheri L. 9527<br />

Squarcina, Paola 5555<br />

Squiban, Patrick J. e13009,<br />

e13010<br />

Squire, Jeremy 2051<br />

Squires, Jennifer 2545<br />

Squires, Matthew TPS1148,<br />

6514^, 6625^<br />

Sreenivas, Vishnu kumar<br />

9549, 9551<br />

Sreenivasappa, Shylandra B.<br />

e14590<br />

Sridhar, Kala e15017<br />

Sridhar, P S e11027, e14675<br />

Sridhar, Papaiah Susheela<br />

e14721<br />

Sridhar, Srikala S. 2572,<br />

4522, 4525, e15020,<br />

e15177^<br />

Srimuninnimit, Vichien 551,<br />

7519^, e11022, e15064,<br />

e20514<br />

Srinath, B S e11511,<br />

e12030<br />

Srinivas, Nuggehally R<br />

e13548<br />

Srinivas, Sandy 4506, 4610,<br />

e15005, e15154<br />

Srinivas, Shanthi e18516,<br />

e19638<br />

Srinivasan, Alagiri e15510<br />

Srinivasan, Ramaprasad<br />

TPS4680<br />

Srinivasan, Sankar 1534<br />

Srinivasan, Shankar 8016,<br />

8037<br />

Sriuranpong, Virote 7519^,<br />

TPS7614, e20514<br />

Srivastava, Deo Kumar 9527,<br />

9531<br />

Srivastava, Deokumar 9526<br />

Srivastava, Geetika 8096<br />

Srivastava, Kirti e19516<br />

Srivastava, Shiv e15207<br />

Srkalovic, Gordan 7512<br />

Srour, Samer Ali e17012<br />

Srovnal, Josef e14556<br />

St. Cyr, Brianna 10575<br />

St. Germain, Diane C. 6086<br />

Staak Junior, Marcos Cesar<br />

e12035<br />

Staals, Eric L. 10022<br />

Staar, Susanne 5512, 7001<br />

Stabler, Stacy Marie e19566<br />

Stacchiotti, Silvia 10006,<br />

10022, 10026, 10053,<br />

10065<br />

Stack, Edward C. 4577,<br />

4580, 4582, 4585<br />

Staddon, Arthur P. 10009,<br />

10010, 10067<br />

Stadler, Rudolf 8076<br />

Stadler, Walter Michael 2526,<br />

TPS2613, 4022, 4522,<br />

4550, 4617, 4655, 4667,<br />

8075, 10028<br />

Stadtmauer, Edward Allen<br />

e18540<br />

Staebler, Annette e15577<br />

Staehler, Michael D. 4539,<br />

e15044<br />

Staehler, Sandra S. 9571<br />

Staffurth, John 4558<br />

Stafkey-Mailey, Dana 9143<br />

Stahl, Phillip R. e15168<br />

Staib, Peter 6610<br />

Stalbovskaya, Viktoriya 6514^<br />

Staley, Charles A. 3605,<br />

e14169, e14170<br />

Stallard, Nigel TPS665<br />

Stallings, Holley e16532<br />

Stalmeier, Peep FM 9036<br />

Stam, Anita 2562<br />

Stamatopoulou, S<strong>of</strong>ia 10606,<br />

e11073<br />

Stamatoulas, Aspasia 6523<br />

Stamatovic, Ljiljana 9046<br />

Stambler, Nancy 4662<br />

Stambolic, Vuk 1019<br />

Stambrook, Peter e16017<br />

Stampfer, Martha 1012<br />

Stampino, Corrado Gallo<br />

2595, e13606<br />

Stan, Radu V. e13567<br />

Stancu, Alexandru e13093<br />

Stanelle, Eric J. 8586<br />

Stanford, Marianne 2588<br />

Stang, Andreas 580<br />

Stanganelli, Ignazio 8581<br />

Stanhope, Lucy J 1502<br />

Stapleford, Liza Jane 9122<br />

Staren, Edgar Donald 5049<br />

Stark, Daniel P. 4531<br />

Starling, Gary 8087<br />

Starlinger, Patrick e14095,<br />

e14099<br />

Starmans, Maud HW e17539<br />

Starodub, Alexander 4603,<br />

TPS4676<br />

Staroslawka, Elzbieta TPS649<br />

Starr, Mark e21038<br />

Stash, Diane e15116<br />

Stasi, Irene 629<br />

Stassi, Giorgio e21095<br />

Staszewski, Harry e18554<br />

Staszkiewicz, Rafal 603<br />

Stathis, Anastasios 3003,<br />

3082<br />

Stathopoulos, George 583<br />

Stati, Valeria e11039,<br />

e11514, e12510, e19620<br />

Stattin, Pär 4668<br />

Stauch, Kathrin 4628<br />

Stauch, Martina 3519^,<br />

3588, 3<br />

Stauder, Heribert 4072<br />

Staudinger, Matthias e18565<br />

Stavraka, Chara 2608, 3022,<br />

e13033<br />

Stayner, Lee-Anne 3082<br />

Stea, Baldassarre TPS2107<br />

Steadman, Kenneth 2590<br />

Stearns, Valerie T. e21121<br />

Stearns, Vered 518, 525,<br />

1006, 1128, 9103, 10509<br />

Stebbing, Justin 3546, 3597,<br />

4088, 9103, e11018,<br />

e21137<br />

Steeg, Patricia Schriver 505<br />

Steeghs, Neeltje 2596,<br />

e13605<br />

Steegmann, Juan Luis 6506<br />

Steegmann, Juan Luis 6505^<br />

Steel, Jason C e21064<br />

Steel, Malcolme 3629<br />

Steensma, David P. 6103,<br />

6621<br />

Steer, Christopher B.<br />

LBA5000<br />

Stefan, Holdenrieder e21067<br />

Stefanek, Michael Edward<br />

6006<br />

Stefani, Laetitia 9079<br />

Stefani, Micaela 586<br />

Stefanick, Marcia L 1501<br />

Stefanos, Mona 6508<br />

Stefanski, Jolanta TPS2619,<br />

TPS4700<br />

Steffensen, Karina Dahl 5052<br />

Steg, Adam D. 5036<br />

Stega, Jeanetta e14729,<br />

e14732, e15576<br />

Stegelmann, Frank 6510<br />

Steger, Guenther G. 514,<br />

e21033<br />

Stegmaier, Petra e16033<br />

Stehman, Frederick B.<br />

e15561<br />

Steidl, Stefan 6574<br />

Steidle, Christopher P<br />

e15199^<br />

Stein, Andrew Marc 2543<br />

790s Numerals refer to abstract number


Stein, Anette 8505<br />

Stein, Anthony Selwyn 1592<br />

Stein, Barbara 2506, e13051<br />

Stein, John Peter 4575<br />

Stein, Lincoln D. e13107<br />

Stein, Mark N. 2526, 2539,<br />

TPS3112<br />

Stein, Rob TPS665<br />

Stein, Sandy 10584<br />

Stein, Stacey e14534<br />

Stein, Stacy e11529<br />

Stein, Wilfred Donald e13122<br />

Steinberg, Joyce Leta<br />

TPS668, 8587<br />

Steinberg, Michael S. 1017<br />

Steinberg, Seth M. 505,<br />

2528, 2577, 3043, 4569,<br />

5020, 8051, e18568<br />

Steinbrecher, Jill Elise 4548,<br />

TPS4697<br />

Steiner, Aline e15109,<br />

e21056<br />

Steiner, Gabriel e15044<br />

Steiner, Mitchell S. e19582,<br />

e19585<br />

Steiner, Thomas e15044<br />

Steinger, Brunhilde e16520<br />

Steinhauer, David e17529<br />

Steinhauer, Hjalmar B. 8024<br />

Steinh<strong>of</strong>f, Ann-Katrin 1067<br />

Steinke, Verena 3550<br />

Steinmetz, Kristina e14557<br />

Steinstraesser, Lars 10059<br />

Steis, Ronald G. TPS2103<br />

Stella, Mattia e14730<br />

Stella, Philip J. TPS657,<br />

1025, 7605, e16552,<br />

e18118<br />

Stelljes, Matthias 6500^,<br />

6610<br />

Stelma, Foekje 9070<br />

Stemkowski, Stephen e18560<br />

Stemmer, Salomon M. 548,<br />

1092, 2556, e13080,<br />

e14731<br />

Stemmer, Salomon TPS661<br />

Stemmer-Rachamimov, Anat<br />

2010<br />

Stempel, Michelle 557,<br />

1118, e11531<br />

Stenehjem, David D. 6583<br />

Stenholm, Linnea 10552<br />

Stenning, Sally P TPS4143<br />

Stenzl, Arnulf e15195,<br />

e19021<br />

Stepanek, Vanda M.T. 10510<br />

Stepanski, Edward J. e15061<br />

Stephens, Craig 4563, 7582,<br />

7594, e12007<br />

Stephens, Deborah Marie<br />

e18508<br />

Stephens, Peter 3052<br />

Stephens, Philip J 1015<br />

Stephens, Philip 3533, 4649,<br />

7510, 7529, 10524,<br />

10559, 10590, e17541,<br />

e19004<br />

Stephens, Sonya e17544<br />

Stephens, Talitha e15162<br />

Stephenson, Andrew J. 4652<br />

Stephenson, Joe 4593, 4662,<br />

4665<br />

Stephenson, Patricia 7508<br />

Stephenson, Robert A 2564<br />

Sterchele, James A. 6553,<br />

e13081<br />

Stergiopoulos, Spyros e21027<br />

Stern, Howard 5575, e13048<br />

Numerals refer to abstract number<br />

Stern, Lee e14028, e14030,<br />

e14627, e15043<br />

Stern, Ralph H. e12026<br />

Sternberg, Cora N. 4501,<br />

4519^, 4522, 4525, 4570,<br />

TPS4683, e15003, e15059<br />

Sterry, Wolfram 8535<br />

Stetler-Stevenson, Maryalice<br />

2503^, 8051, 8088,<br />

e18568<br />

Steuer, Conor E 8101<br />

Steurer, Stefan 10501<br />

Steven, Neil Matthew 2583<br />

Steven, Neil TPS8605<br />

Stevens, Craig William 7065,<br />

e17559<br />

Stevens, Glen 2076, 2100<br />

Stevens, Sandy e16533<br />

Stevenson, James 7077,<br />

7092, 7098, e18032<br />

Stevenson, Kristen 9530<br />

Stevenson, Laura TPS4143<br />

Stewart, A. Keith 8010,<br />

8013, 8035, 8053<br />

Stewart, Alex e21012,<br />

e21142<br />

Stewart, Andrew 6046, 6056,<br />

6088<br />

Stewart, Clinton F. e13584<br />

Stewart, Douglas Allan 6548<br />

Stewart, Elizabeth e13584<br />

Stewart, Frances P. 2563<br />

Stewart, John H. 6127<br />

Stewart, Larissa 8537<br />

Stewart, Rena e21045<br />

Stewart, Robert 4633<br />

Stewart, Tyler 1132<br />

Stiede, Kathryn 8032<br />

Stieglitz, Heather e16500<br />

Stieler, Jens 4020, 4044,<br />

4050, e14533<br />

Stien, Kathy J. e16572<br />

Stiff, Patrick J. 4022,<br />

e13121<br />

Stilidi, Ivan S. e20501<br />

Stilwell, Jackie L. e21148<br />

Stimpson, Sarah Jane 3080<br />

Stinchcombe, Tom 7513,<br />

TPS7619<br />

Stineman, Margaret G<br />

e19531<br />

Stingl, Julia TPS1612<br />

Stinson, Jennifer 9556<br />

Stintzing, Sebastian 3519^,<br />

3541, 3588, TPS3639,<br />

4072<br />

Stirewalt, Derek L. 2523<br />

Stitzenberg, Karyn Beth 6104<br />

Stobiecka, Ewa e14159,<br />

e21049<br />

Stock, Richard e15164<br />

Stock, Wendy 6562, 6582,<br />

e13120^<br />

Stocken, Deborah TPS4150<br />

Stocker, Susan e14642<br />

Stockerl-Goldstein, Keith<br />

8011<br />

Stockler, Martin R. 4531,<br />

TPS4681, TPS5116,<br />

7079^, 7511, 9087, 9126,<br />

e16501, e19642<br />

Stockman, Paul 6527<br />

Stoeckle, Eberhard 10096<br />

Stoecklein, Nikolas H.<br />

e21017, e21020<br />

Stoecklein, Nikolas e14527<br />

Stoeger, Herbert 10501<br />

Stoehlmacher-Williams, Jan<br />

2573^, 4090, 5504^,<br />

10552<br />

Stoelben, Erich 1533,<br />

TPS7109, 7603, 10526,<br />

CRA10529, e18000<br />

Stoemmer, Peter E. e14076,<br />

e15013<br />

Stoenescu, Adela 5007<br />

Stoermer, Kati M e16508<br />

Stoetzer, Oliver J. e21067<br />

St<strong>of</strong>fels, Ingo e19031<br />

Stoger, Herbert 514<br />

Stokes, Maurice 1113<br />

Stokes, William A. 4658,<br />

6032<br />

Stokes, William Allen 6037<br />

Stokoe, Christopher T. 547<br />

Stoller, Ronald G. TPS1138,<br />

3037, 3054<br />

Stoller, Ronald 3049<br />

Stone, Nelson e15164<br />

Stone, Rebecca Lynn 5027<br />

Stone, Richard M. 6103,<br />

6526, 6606, 6617, 6621,<br />

e13586<br />

Stopeck, Alison e16528<br />

Stopfer, Jill 1506<br />

Stoppa, Anne-Marie 1580,<br />

e19539<br />

Storino, Claudio e15521<br />

Stork, Linda C. 9546,<br />

TPS9594<br />

Stork, Lisette 3510, 10545<br />

Stork-Sloots, Lisette 577,<br />

10554, TPS10632,<br />

e21013<br />

Storm, Courtney D. 6095,<br />

6128<br />

Storniolo, Anna Maria 525,<br />

6053, 10509<br />

Storoz, Cristina e15202<br />

Stott, Will e16045<br />

Stotz, Michael e14066<br />

Stouthard, Jacqueline 10504<br />

Stovall, Marilyn 9505,<br />

CRA9513, 9514, 9528<br />

Stoyianni, Aikaterini 10575<br />

Stradella, Agostina 5595,<br />

e14104, e15159<br />

Strahs, Andrew Louis 4132,<br />

4501, e13041, e15082<br />

Straley, Kimberly 6606<br />

Strasser, Florian e19648<br />

Stratmann, Antje e21017,<br />

e21020<br />

Straubinger, Robert M<br />

e13006<br />

Straughn, John Michael<br />

e13087<br />

Straus, David J. 8054<br />

Strauss, Gary M. 1532, 4545,<br />

10579<br />

Strauss, Hans-Georg 5007,<br />

5031<br />

Strauss, James Fredric 10037<br />

Strauss, Lewis C. TPS4134<br />

Strauss, Marcie e16542<br />

Strausz, Janos 7557<br />

Strauß, Arne e15156,<br />

e15158<br />

Stravodimos, Kostas e15040<br />

Stravodimou, Athina e11048,<br />

e17007<br />

Street, Thomas 6122, 8037,<br />

e18556<br />

Streicher, Howard 2501<br />

Streit, Michael R. W. 8501<br />

Streitenberger, Patricia 10074<br />

Stremitzer, Stefan 3613,<br />

e14095<br />

Strenger, Rochelle 1045<br />

Strevel, Elizabeth Laureen<br />

5019, 5025<br />

Stricker, Ingo 10059<br />

Stricker, Thomas 5517, 9534<br />

Strickland, Andrew H.<br />

TPS3637<br />

Strickland, Stephen Anthony<br />

TPS6637<br />

Strickler, John H. 3039<br />

Striefler, Jana Kaethe 4020,<br />

e14533<br />

Strik, Herwig Matthias 2079<br />

Strimpakos, Alexios S<br />

e17508, e17557<br />

Strina, Carla e11546<br />

Stringer, Bradley 3077<br />

Strippoli, Antonia e14156<br />

Strobel, Deike e19032<br />

Stroebel, Philipp 7105<br />

Stroh, Mark 3009<br />

Strola, Giuliana 2600<br />

Strom, Brian L. 1503<br />

Strom, Charles m 2587<br />

Strong, Laura E. TPS3113<br />

Strope, Seth A. 4594<br />

Strosberg, Jonathan R. 2534,<br />

2612, 4047, 4099, 4125,<br />

4126, e14542, e14551,<br />

e14579, e14610, e14627<br />

Strother, Douglas R. 9533<br />

Strother, Robert Matthew<br />

5107<br />

Stroyakovsky, Daniel 5017^<br />

Struble, Erika J. 9058<br />

Strudel, Martina e11039,<br />

e11514, e12510, e19620<br />

Struemper, Herbert 5065<br />

Strumberg, Dirk e13597<br />

Strutz, Juergen 5512<br />

Strychor, Sandra 3049<br />

Stryker, Scott 1581, 4659,<br />

e15147, e15169<br />

Stuart, August 6616, e18500<br />

Stuart, Bruce C. 6028, 6060,<br />

6075<br />

Stuart, Joshua M. 503<br />

Stuart, Robert K. TPS6637<br />

Studeny, Matus e15037<br />

Studer, Cristina e19648<br />

Stuebs, Patrick e14630<br />

Stuermer, Andrea 7105<br />

Stukel, Therese A. 6027<br />

Stump, Tammy K 6021<br />

Stupp, Roger 2001, 2034<br />

Sturgeon, Jeremy F. G. 4535,<br />

e14020<br />

Sturgis, Erich M. 2558<br />

Stuurman, Frederik 2550,<br />

e13028<br />

Styczen, Hanna 3600<br />

Styler, Michael e15522<br />

Styles, Amy e11076<br />

Stylianou, Spyros e13525<br />

Ståhl, Birgitta 7539<br />

Stöckle, Michael 4590,<br />

e15195<br />

Su, ChunXia 7535, 10527<br />

Su, Ih-jen e11068<br />

Su, Jian 7039, e18090<br />

Su, Jie 9032<br />

Su, Pei-Fang 7589<br />

Su, Qiaojuan e21159<br />

Su, Wayne 6553<br />

Su, Wen-Hui 5109<br />

Su, Wu-Chou 4007, LBA7500<br />

791s


Su, Xinying 4124<br />

Su, Yu-Chieh e16008<br />

Su, Zhaohui e11051<br />

Suárez, Ana 602, e11006,<br />

e11517<br />

Suarez, Cristina TPS4679,<br />

e15001, e15041, e21021<br />

Suarez, Eloah Rabello 10568,<br />

e13016<br />

Suarez, Javier 3586<br />

Suarez-Almazor, Maria 8536,<br />

e19631<br />

Suarez-Garcia, Ines e11028<br />

Subbiah, Ishwaria Mohan<br />

e13020<br />

Subbiah, Shanmuga e14641<br />

Subbiah, Vivek 10021,<br />

e13020<br />

Subiza, Jose Luis 10564,<br />

e13085<br />

Subklewe, Marion 8030<br />

Subramaniam, Deepa Suresh<br />

2590, 3095<br />

Subramanian, Somasundaram<br />

e19005<br />

Subramonia-Iyer, Subramony<br />

e21133, e21154<br />

Subtil, Fabien 4091<br />

Subudhi, Sumit Kumar 4516<br />

Suciu, Stefan 8532<br />

Suckstorff, Ivonne 2584<br />

Sud, Shelly 6083<br />

Suda, Margaret 5579<br />

Sudaka, Anne 5521<br />

Suder, Aneta 3022<br />

Sudhakar, S e14675<br />

Sudhakar, Tummala 6546<br />

Sudo, Kentaro 4046<br />

Suehara, Yoshiyuki 7578<br />

Suenaga, Mitsukuni 10541,<br />

e14145<br />

Suetsugu, Atsushi e14014,<br />

e21051<br />

Sufliarsky, Jozef TPS649<br />

Sugar, Elizabeth 4045,<br />

e14585<br />

Sugar, Linda M. 4613<br />

Sugawara, Shunichi 7086,<br />

7515, 7565, 7571,<br />

e18129, e19521<br />

Sugi, Kazurou e17517<br />

Sugi, Naoko Hata e13511<br />

Sugie, Tomoharu e21004<br />

Sugihara, Eiji 635<br />

Sugihara, Kenichi e14091,<br />

e14127<br />

Sugimori, Kazuya 4035<br />

Sugimoto, Hiroyuki e18081<br />

Sugimoto, Kiichi e14139<br />

Sugimoto, Mikio e15127<br />

Sugimoto, Naotoshi 4002<br />

Sugino, Keishi e18127<br />

Sugio, Kenji TPS7110<br />

Sugita, Hir<strong>of</strong>umi 7066<br />

Sugiyama, Eri 7044<br />

Sugiyama, Shunsuke e19611<br />

Sugiyama, Tomohide e13029<br />

Sugiyama, Toru 5003, 5103<br />

Suh, Cheol Won e18501<br />

Suh, Cheolwon 7004, 8023,<br />

e13104, e18052<br />

Suh, Dong-Hoon e15503<br />

Suh, Eugene 9586<br />

Suh, John H. 2018, 2076,<br />

2092, 2100<br />

Suh, K. Stephen e18507<br />

Suhail, Ali M. e21039<br />

Suhaimi, Nur-Afidah Binte<br />

3589<br />

Sujoy, Victoria 595<br />

Sukari, Ammar 2081, 5534,<br />

7593, e16019<br />

Sukeepaisarnjaroen, Wattana<br />

4108<br />

Sukegawa, Gen e15038<br />

Suki, Dima 2019<br />

Sukor, Sumainizah 622<br />

Sukov, William R. 4009<br />

Sukumar, Saraswati 518,<br />

e13559<br />

Sukumaran, Shawgi e19550<br />

Sukumaran, Sujita e13500,<br />

e13577, e21029<br />

Sukumaran, Sunitha e17555<br />

Suleiman, Yaman 4534,<br />

7032, 7107, 7108<br />

Sulkes, Aaron 548, e14571<br />

Sullivan, Adam 9116<br />

Sullivan, Amy 6038<br />

Sullivan, Clare e16515<br />

Sullivan, Daniel 2501, 2534,<br />

TPS2620, 3077, 5020,<br />

8013, e13603<br />

Sullivan, Frank e15126<br />

Sullivan, Irene e19646<br />

Sullivan, Ivana 5590, 5595,<br />

7041, e15159, e18057<br />

Sullivan, Kristen 6122, 8037,<br />

e18556<br />

Sullivan, Laura A 5029<br />

Sullivan, Maryann e14125,<br />

e19638<br />

Sullivan, Peggy S 10588<br />

Sullivan, Ryan J. 8516, 8569<br />

Sullivan, Sean D e14141,<br />

e14146, e15175, e15183,<br />

e18097<br />

Sullivan, Timothy J. 2043,<br />

e13580<br />

Sulman, Erik P. 2001, 2002,<br />

8584<br />

Sultan, Sadaf e13039,<br />

e21128<br />

Sultana, Rebeka 1014<br />

Suman, Saurav e15181<br />

Suman, Vera J. 503<br />

Suman, VeraJean 534, 5544,<br />

e13086<br />

Sumi, Minako 7070, 9574<br />

Sumino, Hiroshi 1103<br />

Summer, Stephen J. 9575<br />

Summers, Deborah 8560<br />

Summers, Yvonne J. e18095<br />

Sumner, Dyana K. 7052,<br />

e19647<br />

Sumpter, Katherine Anne<br />

TPS4143<br />

Sumstand, Darin 2546<br />

Sun Myint, Arthur e14149<br />

Sun, Can-Lan 1592, 9512<br />

Sun, Canlan 8038<br />

Sun, Charlotte C. 1518,<br />

6116, 9134, e13026<br />

Sun, Dandan 8597<br />

Sun, Di 9548<br />

Sun, Guigin e15563<br />

Sun, Guihua 10545<br />

Sun, Haibo 7034<br />

Sun, Han e18076<br />

Sun, Hong 9059<br />

Sun, Hongliu e14098<br />

Sun, Jian 5593<br />

Sun, Jiayuan e17551<br />

Sun, Jong-Mu 7004, 7104,<br />

e18148<br />

Sun, Julia e15540<br />

Sun, Kai e15183<br />

Sun, Lihong 10563<br />

Sun, Luhong e18557<br />

Sun, Mai e15560<br />

Sun, Peng 3528, 8510<br />

Sun, Peter 1609, e12042,<br />

e15096<br />

Sun, Qiang TPS649<br />

Sun, Shiliang e14620<br />

Sun, Si e11567, e11568<br />

Sun, Sophie 549^, 7020<br />

Sun, Virginia 9068, e16563<br />

Sun, Weihong 1585<br />

Sun, Weijing 3522, 3523,<br />

3525, 4077, 4111<br />

Sun, Weimin 2587<br />

Sun, Wenyong e21080<br />

Sun, William 6624<br />

Sun, Xia 4068<br />

Sun, Xiaojiang e17527<br />

Sun, Xindong 4073<br />

Sun, Xiu 614<br />

Sun, Yan 7519^, 7559<br />

Sun, Yihong 4092, e14575<br />

Sun, Yong-kun e14668<br />

Sun, Yongkun e14606<br />

Sun, Yu-Nien 5072<br />

Sun, Yuanjue 9017<br />

Sun, Yuanlu 5538<br />

Sun, Yuliang 626, e13579<br />

Sun, Zhuoxin 504<br />

Sunaga, Noriaki e18037<br />

Sunakawa, Yu 3088^<br />

Sunami, Eiji e14142<br />

Sundaram, Shankar e15116<br />

Sundby Hall, Kirsten 10081<br />

Sunderkötter, Cord 8505<br />

Sundstrom, Stein 7001<br />

Sundstrøm, Stein 7027<br />

Suneja, Gita 6070<br />

Suner, Ali 638<br />

Sung, Max W. 4043<br />

Sung, Myung-Whun 5552<br />

Sunkavalli, Venkata Ratnam<br />

9055<br />

Sunkavalli, Venkataratnam<br />

e12501<br />

Sunpaweravong, Patrapim<br />

e14639, e20514<br />

Sunpaweravong, Somkiat<br />

e14639<br />

Sunwoo, John 2514<br />

Sunyach, Marie-Pierre 10042<br />

Supko, Jeffrey G. 4112<br />

Suppapanya, Nittaya e12001<br />

Suppiah, Kathiresan e18122<br />

Suraokar, Milind B. 7078<br />

Surapaneni, Aparna e15097<br />

Surapaneni, Rakesh 1127,<br />

e12008<br />

Sure, Ulrich 2086<br />

Sureda, Anna 8002<br />

Suri, Anuj 5063<br />

Susanne, Jungwirth 2024<br />

Suschak, Jessica 1091<br />

Suskan, Emine e20000<br />

Sussman, Jeffrey J. e14714<br />

Sussman, Jonathan 6035,<br />

6039<br />

Sutaria, Jaini e17021<br />

Suter, Thomas 532<br />

Sutherland, Heather J 8018^<br />

Sutherland, Robert L. 504,<br />

TPS1136<br />

Sutphen, Rebecca 1608<br />

Sutradhar, Rinku 6039<br />

Suttle, Ben B 2593<br />

Sutton, John E. e14514<br />

Sutton, Steve e21155<br />

Suva, Larry J. e21039<br />

Suvannasankha, Attaya<br />

e18555<br />

Suvorov, Alexander 8018^<br />

Suwa, Yusuke e14050<br />

Suzuki, Akihiro 4069, 4078,<br />

4086, e14547, e14669<br />

Suzuki, Haruko 7021<br />

Suzuki, Hidekazu e18036,<br />

e18042<br />

Suzuki, Hidenori e16032<br />

Suzuki, Hiroyuki e13015<br />

Suzuki, Kenji 7012, 10548<br />

Suzuki, Kenshi 8097<br />

Suzuki, Kousuke e14722<br />

Suzuki, Osamu e15533<br />

Suzuki, Ritsuro 6533<br />

Suzuki, Tatsuya 8029<br />

Suzuki, Toshiro 7086<br />

Suzuki, Yasuhiro 565<br />

Suzuki, Yasukuni e14510<br />

Suzumura, Tomohiro e17538<br />

Svabova, Veronika e19020<br />

Svane, Inge Marie 2565,<br />

2574<br />

Svegliati Baroni, Gianluca<br />

e14561<br />

Svensson, Liz TPS5598<br />

Svetlovska, Daniela TPS4677,<br />

e15027<br />

Svoboda, Jakub e18529,<br />

e18540<br />

Svoboda, Marek 1087<br />

Swaby, Ramona F. 605,<br />

6025, e11085<br />

Swain, Sandra M. LBA506,<br />

533^, 597^, 598^,<br />

LBA1000, 1025, TPS1142,<br />

10603, e11055^, e16543<br />

Swaisland, Helen 3048, 3051<br />

Swallow, Carol Jane 10050<br />

Swallow, Elyse e14621<br />

Swami, Nadia 9003, e16567<br />

Swami, Umang 3093<br />

Swann, R. Suzanne<br />

LBA8500^, 8501<br />

Swanson, Gregory P. 4509<br />

Swanson, Kenneth D. e21078<br />

Swanton, Charles TPS650<br />

Swart, Rachel Elizabeth 529<br />

Swatkowski, Pamela L<br />

e21111<br />

Sweeney, Christopher 4513,<br />

4555, 4603, TPS4694<br />

Sweeney, HelenAnne 1598<br />

Sweeney, John P. e15078<br />

Sweeney, Thomas J. 9115<br />

Sweep, Fred C.G.J. e13111<br />

Sweet, Joan e15118<br />

Sweet, Kendra Lynn 6568,<br />

6575, 6588, 6608, 8084<br />

Sweetenham, John W. 8032,<br />

8055, 8071<br />

Sweis, Randy F. 4573<br />

Swensen, Ron E. 5082^<br />

Swern, Arlene S 6122,<br />

e18556<br />

Swieboda-Sadlej, Anna 4042<br />

Swift, Lonnie e13587<br />

Swift, Regina A. 8098,<br />

e18549, e18558<br />

Swinton, Nelda e19626<br />

Swisher, Elizabeth M. 5073<br />

Swisher, Stephen 4069,<br />

4078, 4086, 7023, 7043,<br />

7047, 7062, e14547,<br />

e14548<br />

Switaj, Tomasz 8548<br />

Switzky, Julie C. 8501<br />

792s Numerals refer to abstract number


Sycova-Mila, Zuzana<br />

TPS4677, e15028<br />

Sydes, Matthew Robert 4509,<br />

10081<br />

Syed, Fayaz ahmed e12501<br />

Syed, Nel<strong>of</strong>er e18567<br />

Syh, Jarron e17524<br />

Syh, Joseph e17524<br />

Sülberg, Heiko e14162<br />

Sym, Sun Jin e14137,<br />

e14685<br />

Symanowski, James Thomas<br />

e15210<br />

Symmans, William Fraser<br />

515, 545, 1012, 10559<br />

Symon, Zvi e14143<br />

Synold, Timothy W. 3078,<br />

3108, e15114<br />

Syrigos, Konstantinos N.<br />

7554<br />

Syrigos, Kostas N e17508,<br />

e17557<br />

Syto, Mary 6577<br />

Szabatura, Audrea e16515<br />

Szabo, Eva 7033, 7103<br />

Szabo, Miklos e13102<br />

Szabo, Peter e21140<br />

Szabo, Stephen 569, 571,<br />

e11002, e14106, e14141,<br />

e14146, e15183, e18097,<br />

e19615<br />

Szallasi, Zoltan 10571<br />

Szarka, Christine E. e19519<br />

Szasz, Attila Marcell e21005<br />

Szcylik, Cezary TPS4683<br />

Szczudlo, Tomasz K. 6505^<br />

Szczylik, Cezary TPS4135,<br />

4501<br />

Sze, Ming 6111, e16507<br />

Szerlip, Nicholas 2081<br />

Sziraki Kiss, Valeria e21140<br />

Szlosarek, Peter Wojciech<br />

e18567<br />

Sznol, Mario CRA2509,<br />

TPS2613, 4505, 8507,<br />

8508<br />

Szostakiewicz, Barbara 505<br />

Szubert, Alexander 8015<br />

Szumera-Cieckiewicz, Anna<br />

10538<br />

Szyldergemajn, Sergio A.<br />

2539<br />

Szymonifka, Jackie 4027,<br />

8041, 9585, 10058<br />

Sørensen, Henrik T<strong>of</strong>t 1579,<br />

e12023<br />

T<br />

Ta, Matthew e16523<br />

Taback, Bret 1107<br />

Tabarroki, Ali 6521<br />

Tabata, Masahiro 2607,<br />

7042, 7560, 7576,<br />

e18027<br />

Tabata, Toshiharu e19508<br />

Tabata, Tsutomu e14029<br />

Tabatabai, Ghazaleh 2055,<br />

2067, TPS2105, 8526<br />

Tabayashi, Koichi e19508<br />

Tabbara, Imad A. e11084<br />

Tabei, Toshio 613<br />

Taber, Angela Marie 4098<br />

Numerals refer to abstract number<br />

Tabernero, Josep 509, 566,<br />

619, 2567, 2568, 3003,<br />

3014, 3021, 3059, 3062,<br />

3502, 3505, 3510, 3531,<br />

3539, 3574, 3578, 3579,<br />

3587, 3614, TPS3634,<br />

TPS3637, 4053, 7041,<br />

10511, e13048, e13114,<br />

e13605<br />

Tabouret, Emeline 2060,<br />

e19539<br />

Tachibana, Isao e18056,<br />

e18141<br />

Tachikawa, Ryo 7528, 10610<br />

Tacyildiz, Nurdan e20000<br />

Tada, Hirohito TPS7110,<br />

7521<br />

Taddei, Gian Luigi e14042,<br />

e18021<br />

Taffurelli, Giovanni e14647<br />

Taft, David R. 3081<br />

Tagawa, Scott T. 4649<br />

Tagawa, Tomoko 1035<br />

Taghian, Alphonse G. 1122<br />

Taghizadeh, Nilo<strong>of</strong>ar 1502<br />

Tagieva, Naila e18005<br />

Tagliaferri, Pierosandro<br />

e13098, e14577<br />

Taha, Said 2048<br />

Tahar, Zoubir e15142<br />

Tahara, Makoto 3079, 5504^,<br />

5542, TPS5598<br />

Tahir, Ammara H 6612<br />

Tai, Ming-Hui 6075<br />

Tai, Tak Hing P. e16572<br />

Tai, Wai Meng David 3589,<br />

5551, 5580, e14155,<br />

e18546<br />

Tai, Wei-Tien e14516<br />

Tai, Yu-Chong 4663, e15153<br />

Tai, Yu-Zu 8106<br />

Taïeb, Julien 3574, 4018,<br />

4053, 4087, 9026^<br />

Taieb, Sophie 4618<br />

Taillandier, Luc 2023<br />

Taillibert, Sophie TPS662,<br />

TPS2106<br />

Taioli, Emanuela 1046<br />

Taira, Kaoru e14518<br />

Taira, Koichi e18036,<br />

e18042<br />

Taira, Tetsuhiko e18064<br />

Tait, Diana M. e14088<br />

Tait, Nancy 9103<br />

Tajika, Masahiro e14540<br />

Tajiri, Tatsuro 9577<br />

Tak, Won-Young 4103<br />

Takabatake, Daisuke<br />

TPS1147<br />

Takada, Jun 10073<br />

Takada, Minoru 7000, 7521,<br />

10577, e18128<br />

Takaesu, Yotaro 10557<br />

Takagi, Keigo e18127<br />

Takaha, Natsuki e15055,<br />

e15099<br />

Takahara, Masatoshi e19521<br />

Takahari, Daisuke e14500<br />

Takahashi, Atsushi 10520,<br />

e13014, e15068<br />

Takahashi, Hideaki 4046<br />

Takahashi, Hidekazu e19611<br />

Takahashi, Kazuhiro 10519<br />

Takahashi, Kazuhisa e18019<br />

Takahashi, Maiko e11060<br />

Takahashi, Masato 1119,<br />

TPS1147<br />

Takahashi, Masayuki e15068,<br />

e15098<br />

Takahashi, Masazumi e14624<br />

Takahashi, Michinaga e14011<br />

Takahashi, Naoki 8023,<br />

e14126<br />

Takahashi, Naoto 8023<br />

Takahashi, Norihiko 10578<br />

Takahashi, Ryo e18007,<br />

e18056<br />

Takahashi, Ryuji e13046,<br />

e13050<br />

Takahashi, Shin e19611<br />

Takahashi, Shunji e15038,<br />

e19512<br />

Takahashi, Takao 3558,<br />

TPS3642<br />

Takahashi, Tiago Kenji<br />

e21040<br />

Takahashi, Toshiaki 7088,<br />

e21007<br />

Takai, Yujiro e18127<br />

Takamochi, Kazuya 10548<br />

Takamori, Hiroshi e21084<br />

Takamura, Kei 7561, 7571,<br />

e18135<br />

Takano, Atsushi e13552,<br />

e13565<br />

Takano, Masashi 5006, 5037,<br />

e15579<br />

Takano, Toshimi TPS659<br />

Takao, Shintaro TPS1141<br />

Takasa, Akiyuki e19583<br />

Takashima, Atsuo 4031,<br />

e14510<br />

Takata, Ryo 10520<br />

Takayama, Hitoshi e15065,<br />

e15069<br />

Takayama, Tatsuya e15071<br />

Takayanagi, Hiroyuki e11007<br />

Takebe, Naoko 3078, 10004,<br />

10005, e13106<br />

Takechi, Hirokazu e11560<br />

Takeda, Koji 6112, 7003,<br />

7017, 7028, TPS7110,<br />

7549, e18036, e18042<br />

Takeda, Masayuki e18060<br />

Takeda, Shugaku 4604<br />

Takeda, Yuichiro 7017<br />

Takeda, Yutaka e14518<br />

Takehara, Kazuhiro 5084<br />

Takei, Hiroyuki 613, 1106<br />

Takemoto, Mitsuhiro 7045,<br />

e15533<br />

Takemoto, Shuji 5085<br />

Takemura, Shigekazu e14518<br />

Takemura, Shinichi e14011<br />

Takeoka, Hiroaki 3045<br />

Takeshima, Nobuhiro 5103<br />

Takeshita, Jumpei 7528,<br />

10610, e18025<br />

Taketa, Takashi 4069, 4078,<br />

4086, e14547<br />

Taketomi, Akinobu 10578<br />

Takeuchi, Akihiko 10075,<br />

e20505<br />

Takeuchi, Hideya e14722<br />

Takeuchi, Hiroya e14045<br />

Takeuchi, Kengo e18017<br />

Takeuchi, Masahiro 7012,<br />

7515<br />

Takeuchi, Yoshiko e18007,<br />

e18056<br />

Takeyoshi, Izumi 613<br />

Takigawa, Nagio 2607, 7042,<br />

7045, 7560, 7576,<br />

e18027<br />

Takiguchi, Nobuhiro 3524<br />

Takiguchi, Yuichi 7046,<br />

7515, 10542<br />

Takii, Yasumasa 3524<br />

Takimoto, Chris H.M. 3078<br />

Takiuchi, Hiroya 3088^,<br />

e14510<br />

Takizawa, Ken 5103<br />

Takuwa, Teruhisa 7031,<br />

7080, 10585<br />

Takvorian, Anna e15053<br />

Talalay, Paul TPS8606<br />

Talamantes, Tatjana e13557<br />

Talamini, Renato 8058<br />

Talamo, Giampaolo 8090<br />

Talamonti, Mark S. 4049<br />

Talamonti, Mark e14642<br />

Talano, Julie 6537<br />

Talavera, Pablo e20513<br />

Talbot, Denis Charles 4121,<br />

7081<br />

Talcott, James Austin 6107<br />

Tallam, Celestine 2601<br />

Tallman, Martin S. 6532,<br />

6601<br />

Talluri, Jyothsna 7102<br />

Talluto, Craig Charles<br />

TPS2616<br />

Talpaz, Moshe 6503, 6624,<br />

8017<br />

Talwar, Sumit 6114<br />

Tam, Alda e14664<br />

Tam, Vincent Channing<br />

e14073, e16558<br />

Tamagawa, Hiroshi TPS3642<br />

Tamagawa, Koji 3607<br />

Tamai, Hideyuki e14622<br />

Tamaki, Kentaro e11530<br />

Tamaki, Nobumitsu e11530<br />

Tamandl, Dietmar e14095,<br />

e14099<br />

Tamari, Roni 8005<br />

Tamaru, Satoshi e19546<br />

Tambaro, Francesco Paolo<br />

6517, 6603<br />

Tambo, Yuichi e17540<br />

Tamboli, Pheroze 4624<br />

Tamborini, Elena 10026<br />

Tamburelli, Mercedes e15570<br />

Tamimi, Rulla M. 9071,<br />

9100<br />

Tamiya, Motohiro e18036,<br />

e18042<br />

Tamm, Eric P. 4051<br />

Tamura, Kenji 10519,<br />

e11065, e19535<br />

Tamura, Takao e14510<br />

Tamura, Tomohide 6112,<br />

7003, 7017, 7028, 7070,<br />

7602<br />

Tan, Angelina D. 6108<br />

Tan, Antoinette R. 611,<br />

1025, 2534, 3054,<br />

TPS3112, e13596<br />

Tan, Benjamin R. 2525,<br />

e15116<br />

Tan, Bo e14645<br />

Tan, Charmaine e14155<br />

793s


Tan, Chee Seng 1064,<br />

e19523<br />

Tan, Chun-Wen e14026<br />

Tan, Cong 3551<br />

Tan, Daniel S. W. 5551,<br />

e16024<br />

Tan, Daniel Shao-Weng 3007,<br />

3098<br />

Tan, Daryl C.L. 8097,<br />

e18528, e18546<br />

Tan, Eng Huat 5580, 7575<br />

Tan, Eng-Huat 2598, 3098,<br />

5551, e16024<br />

Tan, Eugene 2603, TPS4147<br />

Tan, Francis 4106<br />

Tan, Glaiza L.A. e13574<br />

Tan, Iain B. 3589, 8078<br />

Tan, Iain BeeHuat e14155<br />

Tan, Jonathan 7604<br />

Tan, Kai e14523<br />

Tan, King L. 8003<br />

Tan, Leonard 1593, 8078,<br />

e18515<br />

Tan, Mann Hong 10051,<br />

e20509<br />

Tan, Min-Han 3589, 4536,<br />

4538<br />

Tan, Ming Tony 7546, 9103<br />

Tan, Puay Hoon 5551, 5580<br />

Tan, Sing-Huang 2551<br />

Tan, Soek Siam 4106<br />

Tan, Soo Yong 1593, e18515<br />

Tan, Soo Yong 8078,<br />

e18528, e18546<br />

Tan, Thuan Tong 3098,<br />

e18546<br />

Tan, Wah Siew 3589<br />

Tan, Wei 6565, 6602,<br />

e13006, e14659, e17535<br />

Tan, Weiwei 2610, 7508<br />

Tan, Wen-Lee 2551<br />

Tan, Winston e13501<br />

Tan, Wu Meng e18546<br />

Tan, Yongqiang 2541<br />

Tan, Yuying 1063<br />

Tana, Silvia 5555<br />

Tanaka, Akiko e19521<br />

Tanaka, Fumihiro 7080,<br />

10585<br />

Tanaka, Hidenori e17538<br />

Tanaka, Hiroshi e11065<br />

Tanaka, Junji 6533<br />

Tanaka, Kaoru e18060<br />

Tanaka, Keiji 8094<br />

Tanaka, Koji e14029,<br />

e14039, e14541, e21053<br />

Tanaka, Kosuke 7528, 10610<br />

Tanaka, Kuniya e14047,<br />

e14050<br />

Tanaka, Maki 588<br />

Tanaka, Masao 4035, e14506<br />

Tanaka, Toshiki e17517<br />

Tanaka, Tsutomu e14540<br />

Tanaka, Yoshihiro e13014<br />

Tanaka, Yoshimasa e21004<br />

Tanaka, Yoshinobu 8029<br />

Tanasanvimon, Suebpong<br />

e19645<br />

Tanca, Francesca Maria 4627<br />

Tanco, Kimberson e19603,<br />

e19613<br />

Tanda, Francesco e14094<br />

Tandon, Vickram 10586<br />

Tandstad, Torgrim 4508<br />

Taneja, Charu e12024,<br />

e15197, e18107, e19630<br />

Tang, Bi Xia 8506, 8562,<br />

e15051, e15052<br />

Tang, Cha-Mei 6581, e21079<br />

Tang, Colin TPS4690<br />

Tang, Gong LBA506,<br />

LBA1000<br />

Tang, Jie LBA4537, 7551<br />

Tang, Laura H. e14586<br />

Tang, Lichen e11567,<br />

e11568<br />

Tang, Patricia 3013, 6082,<br />

e14073, e21083<br />

Tang, Rui 2535, 2594, 3009,<br />

4005<br />

Tang, Sujie 1124<br />

Tang, Tiffany 8078, e18515<br />

Tang, Ya-Ling e14629<br />

Tang, Yiyun 7598, 7600<br />

Tang, Yu e14511<br />

Tang, Yuen Fong e14545<br />

Tang, Zhaoqing 4092,<br />

e14575<br />

Tang, Zuojian 9507<br />

Tangen, Catherine M. 4,<br />

4511, 4547, 4571, 4663,<br />

10503<br />

Tangirala, Muralikrishna<br />

4666, e15186<br />

Tangoku, Akira e11560<br />

Tani, Kenzaburo e13014<br />

Tani, Tohru e11564<br />

Taniere, Philippe e13507,<br />

e13554<br />

Tanigawa, Go e15065,<br />

e15069<br />

Taniguchi, Cullen M 10629<br />

Taniguchi, Hirokazu e14126<br />

Taniguchi, Naoyuki e17520<br />

Taniguchi, Shuichi 6533<br />

Tanimoto, Azusa 10569,<br />

10604, e18004, e18014,<br />

e18017<br />

Tanimoto, Mitsune 2607,<br />

7042, 7045, 7560, 7576,<br />

e18027<br />

Tanino, Hirokazu 1119,<br />

TPS1141<br />

Tanioka, Maki 5045, e19548<br />

Taniyama, Kiyomi 5084<br />

Taniyama, Tomoko e19535<br />

Tanizawa, Yutaka e14635<br />

Tannapfel, Andrea 10059<br />

Tanner, Lanier 10004<br />

Tannir, Bayane TPS4149<br />

Tannir, Nizar M. 4533, 4609,<br />

4615, 4624, 9002, 9029,<br />

e15092<br />

Tannock, Ian 6043, 9003,<br />

9021, e13001<br />

Tanvetyanon, Tawee 2559,<br />

5564, 7024, 7065,<br />

e17559<br />

Tanwandee, Tawesak<br />

TPS4149, e13061<br />

Tanzawa, Hideki 10542<br />

Tanzawa, Yoshikazu 10075<br />

Tao, Hiroyuki e17517<br />

Tao, Jianguo e21123<br />

Tao, Li e14648<br />

Tao, Min 8554<br />

Tao, Miriam 1593, 8078,<br />

e18515, e18546<br />

Tap, William D. 10002,<br />

10003, 10004, 10012,<br />

10019, TPS10099,<br />

TPS10100, TPS10103<br />

Taphoorn, Martin J. B. 2,<br />

6002, 6090, 7607<br />

Tapia, Gonzalo e14058<br />

Taplin, Mary-Ellen 4519^,<br />

4520, 4521, 4665,<br />

TPS4695<br />

Taran, Florin-Andrei 1090,<br />

e21003<br />

Taran, Tanya 512, 551, 559,<br />

TPS648<br />

Tarantini, Luigi 645<br />

Tarass<strong>of</strong>f, Peter G. 6559,<br />

6627, 6632<br />

Tarazi, Jamal Christo 4546<br />

Tarazona, Noelia e14589<br />

Tarbell, Nancy 9537, 9585<br />

Tarco, Emily 1054<br />

Tarella, Corrado 8030<br />

Targarona, Eduardo e14104<br />

Tarhan, Cagla e15572,<br />

e15573<br />

Tarhini, Ahmad A. 8522,<br />

8528, 8533, 8547,<br />

TPS8606<br />

Tarifa, Antonio e21086<br />

Tarleton, Kenneth 9025<br />

Tarnai, Robert 6501<br />

Tarnawski, Rafal e14159,<br />

e21049<br />

Taron, Miquel 4068, 4579,<br />

7007, 7011, 7522, 7540,<br />

7542, 10596, e14509,<br />

e14521, e18130, e18131,<br />

e18137, e18143<br />

Tarpgaard, Line Schmidt<br />

3548<br />

Tarpin, Carole e19539<br />

Tarpinian, Karineh 9103<br />

Tarrant, Teresa K. e13063<br />

Tarraran, Loredana 10066<br />

Tarrío, Nuria e14595<br />

Tartarini, Roberta 10611<br />

Tartour, Eric 5554<br />

Tashakkor, Amir Yashar 6085<br />

Tashiro, Haruko 9053<br />

Tasian, Sarah Kathleen 9506<br />

Taskinen, Minna 8074<br />

Tasneem, Asba 6047, 6051<br />

Tassone, Pierfrancesco<br />

e14577<br />

Tatangelo, Fabiana e14130<br />

Tatarian, Talar e11084<br />

Tateishi, Souichirou 1103<br />

Tateishi, Ukihide 4626, 8023<br />

Tatersall, Martin 10006<br />

Tateyama, Miki 3593<br />

Tatla, Raman e16547<br />

Tatli, Ali Murat e14070<br />

Tatsumi, Mitsuaki 8023<br />

Tatsuta, Noriaki 3086<br />

Tattersall, Martin HN 9087,<br />

9126<br />

Tatzreiter, Georg e14087<br />

Taub, Robert N. e13064,<br />

e13066, e13067<br />

Taube, Janis M. CRA2509,<br />

5506<br />

Tauber Jakab, Katalin e21140<br />

Tauber, Rudolf 5047<br />

Tauchert, Felix 4080<br />

Taunton, Jack 8544<br />

Tavakkoli, Fatemeh e13075<br />

Tavares, Renato TPS6640<br />

Tavassoli, Fattaneh 1109<br />

Tawashi, Manal e13602,<br />

e13604<br />

Tawbi, Hussein Abdul-Hassan<br />

TPS1138, 3049, 3054<br />

Tawbi, Hussein 8528, 8533,<br />

TPS8606, TPS10103<br />

Tay, Cheong Kiat Julian<br />

e19523<br />

Tay, Kevin 1593, 8078,<br />

10051, e18515, e18546,<br />

e20509<br />

Tay, Miah Hiang 4103<br />

Tayi, Ravi e19544, e19567,<br />

e19578<br />

Taylor, Barry S. 4527<br />

Taylor, Clare TPS2619,<br />

TPS4700<br />

Taylor, Gretchen Elizabeth<br />

4055<br />

Taylor, Ian 7530, TPS7615<br />

Taylor, Julie 9048<br />

Taylor, Marianne 549^<br />

Taylor, N Jane 1066<br />

Taylor, Paul 7013, 7583,<br />

e18095<br />

Taylor, Sally e19588<br />

Taylor, Sara Kristina 549^<br />

Taylor, Sarah A. e13551<br />

Taylor, Sarah H 7608,<br />

e15181<br />

Taylor, Stephanie 2081,<br />

e14609<br />

Taylor-Stokes, Gavin e16570<br />

Tazbirkova, Andrea e13128<br />

Tchakov, Ilian 3048, 3051,<br />

5001<br />

Te Loo, Dunja Maroeslea<br />

W.M. 9550, 10077<br />

Tea, Muy-Kheng Maria 1537<br />

Teachey, David T. 9506<br />

Teal, Christine B. e11084<br />

Tebbutt, Niall Christopher<br />

3515, 3534, 3614,<br />

TPS3643<br />

Tecimer, Coskun e14698<br />

Tedder, Thomas 2514<br />

Tee, Goh Yeow 6512, e18528<br />

Tefera, Eshetu e16543<br />

Tefferi, Ayalew 6614,<br />

TPS6639, TPS6641<br />

Tegze, Bálint e21005<br />

Teh, Bin Tean 4624, 8078,<br />

e18515<br />

Tehfe, Mustapha Ali e18136<br />

Tehfe, Mustapha e17507,<br />

e18120<br />

Teijeira, Lucia e21086<br />

Teira, Pierre 9563, 9570<br />

Teishima, Jun e15068<br />

Teitelbaum, April H. e17017<br />

Teitelbaum, April e16539<br />

Teitelbaum, Ursina R. 4111,<br />

e13602<br />

Teixeira, Adriana e12040<br />

Teixeira, Manoel Jacobsen<br />

2038<br />

Tejani, Mohamedtaki<br />

Abdulaziz 10064<br />

Tejeda, Nicole e21071<br />

Tejerina, Eva 10547<br />

Tejiram, Shawn 10070<br />

Tejpar, Sabine 3509, 3511,<br />

3531, 3575, 3587<br />

Tejura, Bindu TPS4693<br />

Tejwani, Sheela 3078<br />

Tekautz, Tanya M. 2076,<br />

2100<br />

Teker, Fatih e14107<br />

Tekin, Deniz e20000<br />

Tekin, Salim Basol e14526<br />

Tekkis, Paris P e14132<br />

Teknos, Theodoros Nicholas<br />

e16014<br />

Teleni, Laisa Vicki e19550<br />

Telera, Stefano Maria 2085<br />

Teletaeva, Gulfiya Midhatovna<br />

e13059<br />

Telfer, Margaret C. e18534<br />

Tello, Sebrina A TPS4678<br />

Teltsch, Dana e13081<br />

794s Numerals refer to abstract number


Temam, Stéphane 5505,<br />

5522<br />

Tembhare, Prashant 8088,<br />

e18568<br />

Temel, Jennifer S. 6004,<br />

6048, 6062, 6106, 7010,<br />

7099, 9004, 9030, 9101<br />

Temiz, Suleyman e14618<br />

Temkin, Sarah Madhu<br />

e16546<br />

Tempelh<strong>of</strong>f, G.-F. e13100^<br />

Tempero, Margaret A. 3105,<br />

4048<br />

Temple, Graham e15037<br />

Temple, Larissa K. F. 3583,<br />

6073<br />

Templeton, Arnoud 9059<br />

Tenen, Daniel G. 7523<br />

Tenen, Daniel G e13505<br />

Teng, Gaojun e14605<br />

Teng, Yun 6000<br />

Tenglin, Richard Charles<br />

1083<br />

Tennant, Lucinda 2571,<br />

e19008<br />

Tennevet, Isabelle 5505<br />

Teo, Melissa 10051, e20509<br />

Teo, MinYuen 2062, e14705,<br />

e15093, e17561, e19059<br />

Teo, Yi Ling e13023<br />

Te<strong>of</strong>ilovici, Florentina 3086,<br />

3094, TPS7613^<br />

Teoh, Gerrard 8097<br />

Teomete, Mehmet e16015<br />

Teot, Lisa A. 10081<br />

Tepperberg, James H e21111<br />

Ter Voert, E. e13111<br />

Terachi, Toshiro e15071<br />

Terada, Mizuho 565<br />

Teragni, Cristina 589<br />

Terai, Hideki e18058<br />

Terajima, Hiroaki e14518<br />

Teramukai, Satoshi 588<br />

Terao, Mayako 565<br />

Terashima, Masaaki e18060<br />

Terashima, Masanori 4012,<br />

e14635<br />

Terauchi, Fumitoshi 5003,<br />

10557<br />

Terauchi, Takashi 8023<br />

Terebelo, Howard R. 8037,<br />

e18556<br />

Terlizzo, Monica 3613<br />

Terpos, Evangelos 8095<br />

Terracciano, Luigi 7061<br />

Terranova, Corrado 5093,<br />

e15547, e15550<br />

Terrasa, Josefa e12012<br />

Terrell, Deirdra e17001<br />

Terrenato, Irene 1050<br />

Terriat, Beatrice 1580<br />

Terrier, Philippe 9536,<br />

10018, 10044<br />

Terry, Mary Beth 1502<br />

Tertian, Gerard 6523<br />

Terui, Yasuhito 10541<br />

Tesch, Hans 554, 1023<br />

Teschendorf, Christian<br />

TPS3639<br />

Tesfaye, Anteneh A. 3619,<br />

7102, e14046, e15554<br />

Tessen, Hans Werner 6566<br />

Testa, Enrica 4084<br />

Testa, Isabella 4629<br />

Testa, Joseph 4605, 7083<br />

Testelin, Sylvie e16013<br />

Tester, William J. 7051<br />

Numerals refer to abstract number<br />

Testori, Alessandro 8502^,<br />

8513, 8529, 8532, 8534,<br />

e19035, e19036<br />

Teten, Rachel Threet 1608<br />

Tetteh, Ernestina e13599<br />

Tetterton, Jill 2074^<br />

Teule, Alex 4119<br />

Teulon-Navarro, Isabelle 5069<br />

Tevaarwerk, Amye 555, 563,<br />

3101, 6063<br />

Tew, William P. 5056, 5057,<br />

5108, 9034, 9109, 9123,<br />

e18133<br />

Tewari, Krishnansu Sujata<br />

1519<br />

Thakar, Alok 5540<br />

Thaker, Premal H. 5013,<br />

5101, 9134<br />

Thakkar, Jigisha P. e12036<br />

Thakral, Charu e18535<br />

Thakur, Archana e13089<br />

Thaler, Howard T. e19566<br />

Thaler, Josef 4074<br />

Thalgott, Mark K. 4590<br />

Thall, Peter F. e14548<br />

Thalmann, George 4661<br />

Tham, Chee Kian 2066,<br />

4100^<br />

Tham, Ivan Weng Keong<br />

2016, 5553<br />

Thanvilay, Dominic- Vidaroun<br />

9563<br />

Thariani, Rahber e16524<br />

Thariat, Juliette 10035,<br />

10042, e16013<br />

Tharkar, Shweta 10614<br />

Tharnish, Mark 9110<br />

Thatcher, Nick LBA7000,<br />

TPS7612<br />

Thaw, Sunn Sunn Htet<br />

e21125<br />

Thayer, Mathew 9555<br />

Theaker, Jeffrey 8566<br />

Theaux, Ricardo Alejandro<br />

e21025<br />

Theeler, Brett James 2022<br />

Thein, Mya Sanda e13042<br />

Thelen, Paul e15156,<br />

e15158<br />

Theocharis, Stamatios<br />

e21098<br />

Theodore, Christine<br />

LBA4567^, TPS4696^<br />

Theodoulou, Maria 10514<br />

Theoleyre, Sandrine e21117<br />

Thépot, Sylvain 6523<br />

Therasse, Patrick e13061<br />

Theriault, Richard L. 599,<br />

6116<br />

Thery, Jean Christophe 1095,<br />

5067<br />

Theuer, Charles P. 3034,<br />

3042^, e21038<br />

Thézenas, Simon 3633<br />

Thibodeau, Stephen N. 3514<br />

Thiébaut, Anne e14065<br />

Thieblemont, Catherine 8021,<br />

8048<br />

Thiel, Eckhard 2007, 2054,<br />

8031<br />

Thiel, Falk 5007<br />

Thielemans, Kris 2507<br />

Thienprayoon, Rachel e19536<br />

Thierry, Alain R. 10505<br />

Thiery- Vuillemin, Antoine<br />

TPS4696^<br />

Thies, Svenja 2055<br />

Thiessen, Brian 2051<br />

Thigpen, J. Tate 1519<br />

Thiis-Evensen, Espen 4015<br />

Thijssen, Bas 2550<br />

Thippeswamy, Ravi Shankar<br />

9559<br />

Thirlwell, Michael P. 1025<br />

Thirman, Michael J. 6562<br />

Thirot-Bidault, Anne 4018<br />

Thiruvenkatam, Radhika 9518<br />

Thng, Choon Hua 1064,<br />

4100^<br />

Tholander, Bengt 5017^<br />

Thomas, Anish 7033, 7103,<br />

TPS7609<br />

Thomas, Charles R. 5596^,<br />

7018, e14555<br />

Thomas, David e13585<br />

Thomas, Deborah A. 6501,<br />

6578, 6585, 6592<br />

Thomas, George TPS3116,<br />

9541<br />

Thomas, Gerry 1094<br />

Thomas, Gillian TPS5116<br />

Thomas, Hala TPS3116<br />

Thomas, J TPS1144<br />

Thomas, James P. e14613<br />

Thomas, Jay 9068<br />

Thomas, John 2066<br />

Thomas, Luc 8512, 8578,<br />

8591<br />

Thomas, Michael 2573^,<br />

LBA7507, 7599, e18016^,<br />

e18100<br />

Thomas, Pascal e16560<br />

Thomas, Sachdev P.<br />

TPS3635^, 4022, 7073,<br />

7555, e18556<br />

Thomas, Sachdev P 8037,<br />

8075<br />

Thomas, Scott 3058<br />

Thomas, Sheeba K. 8091,<br />

8093, 9082, 9140<br />

Thomas, Sheila e16542<br />

Thomas, Tarita O. e18523<br />

Thomas, Xavier G. 6559<br />

Thomassin, Jeanne e15155<br />

Thomeer, Michiel 7013<br />

Thomes-Pepin, Jessica<br />

e15561<br />

Thompson, Alastair Mark<br />

9022<br />

Thompson, Carrie A. 9057<br />

Thompson, Cheryl L 1569<br />

Thompson, Dana Shelton<br />

3033<br />

Thompson, David e17012<br />

Thompson, Dawn e15162<br />

Thompson, Emilda 4553,<br />

e15136<br />

Thompson, Ian Murchie 4,<br />

4511, 4547, 4571, 4663,<br />

10503<br />

Thompson, James E 6565,<br />

6602<br />

Thompson, Jennifer TPS4681<br />

Thompson, John A. 3077,<br />

8504, e13088, e13603<br />

Thompson, John F 8534,<br />

8559<br />

Thompson, John TPS8116<br />

Thompson, Joyce LBA4001<br />

Thompson, Lisa A. e16541<br />

Thompson, Lisa A e16550<br />

Thompson, Lora 7065,<br />

e17559<br />

Thompson, Michael A.<br />

e16561<br />

Thompson, R. Houston 4657<br />

Thompson, Simon TPS9148<br />

Thompson, Stephen F 4666,<br />

e14028, e14030, e15186<br />

Thomson, Damien B. 4531<br />

Thomson, Erin 593<br />

Thomssen, Christoph 552,<br />

580<br />

Thongprasert, Sumitra 1534,<br />

7519^, e13061, e18063^<br />

thor Straten, Per 2565, 2574<br />

Thor, Ann D. e21099<br />

Thorat, Mangesh A. 505<br />

Thorn, Molly 6114<br />

Thornton, Bob 10573<br />

Thornton, Claire 9539<br />

Thorpe, Villette 3025<br />

Thorsen, Cristina e16504<br />

Thota, Narender Kumar 6554,<br />

e18544, e18545<br />

Thota, Ramya e14641<br />

Thota, Swapna e15023<br />

Thotakura, Vijaya 5582<br />

Thottakam, Bensita M.V<br />

e15011<br />

Threatt, Stevie 2094^, 2095^<br />

Throngprasert, Sumitra<br />

TPS4147<br />

Thu, Minn Minn Myint 5551,<br />

5580<br />

Thuer<strong>of</strong>f, Stefan e15194<br />

Thulkar, Sanjay 5104<br />

Thumar, Jaykumar<br />

Ranchodbhai e14534<br />

Thunberg, Allen L TPS9152<br />

Thurlimann, Beat J. K. 9022,<br />

9059<br />

Thurm, Craig 10528, e21008<br />

Thurm, Holger C. TPS7616<br />

Thurn, Kenneth Ted 3058<br />

Thuss-Patience, Peter C.<br />

TPS4138<br />

Thway, Khin 9510, 10038,<br />

10525, e13601<br />

Thwin, Malar e13099<br />

Thyparambil, Sheeno e21068<br />

Thyss, Antoine 6633, 8077,<br />

10042, 10062<br />

Tiab, Mourad 8009, 8014<br />

Tian, Erming e18548<br />

Tian, Gary G. TPS10099<br />

Tian, Ming e12013<br />

Tian, Qiang e14153<br />

Tian, Song e21011<br />

Tian, Wei e13526<br />

Tian, Ying 3535<br />

Tiangco, Beatrice 7549<br />

Tibau Martorell, Ariadna<br />

1120, 10555<br />

Tibau, Ariadna e14552<br />

Tibbetts, Randal S. e13537<br />

Tica, Stefani 6623<br />

Tidwell, Michael 6611<br />

Tie, Jeanne e16516<br />

Tiedemann, Rodger Edwin<br />

8053<br />

Tiemann, Katharina e13503<br />

Tiemsim, Maricel Cordero<br />

1064<br />

Tier, Katherine 8566<br />

Tiersten, Amy 5016, e11522,<br />

e11529<br />

Tiezzi, Daniel G. 1075<br />

Tiftik, Nalan e15104<br />

Tijani, Lukman Adeoju<br />

e21105<br />

Tijssen, Cees 2<br />

Tilburt, Jon C. 4657, 6126<br />

Tiley, Stephen e11550,<br />

e17547<br />

Tilgen, Wolfgang 8505<br />

795s


Tillmanns, Todd D. 5014,<br />

e15514<br />

Tilly, Herve 8021, 8057,<br />

8068<br />

Timcheva, Constanta TPS661,<br />

9046<br />

Timmer, Wolfgang e19530,<br />

e19532, e19533<br />

Timotheadou, Eleni 5033,<br />

e15040<br />

Tims, David e21146<br />

Timsit, Yoav E 9562<br />

Tin, Aung Soe e20003<br />

Tin, Sanda e13582<br />

Tinh<strong>of</strong>er, Inge e16018<br />

Tinquaut, Fabien 9079<br />

Tinsley, Sara 6588<br />

Tinterri, Corrado 10554<br />

Tinton, Nicolas e14044<br />

Tirabosco, Roberto 10063<br />

Tirado Gomez, Laura Leticia<br />

e15527<br />

Tirapelli, Daniela P. C.<br />

e12512<br />

Tirapelli, Daniela Pretti da<br />

Cunha e19628<br />

Tirelli, Umberto 8046, 8058<br />

Tiribelli, Mario 6531<br />

Tirier, Christian 4065<br />

Tirona, Kattleya M 5587<br />

Tirrell, Stephen 3077<br />

Tiseo, Marcello e18096<br />

Tishler, Roy B. 5501, 5579,<br />

5582, e16060<br />

Tisol, William e17555<br />

Titarova, Yulia Y 9060<br />

Titov, Dmitry e15029<br />

Titus, Mark Anton 4556<br />

Tiu, Ramon Velasquez 6521<br />

Tiutiunnik, Pavel e14717<br />

Tiwari, Aradhana e21150<br />

Tjalma, Wiebren 1129<br />

Tjulandin, Sergei TPS661,<br />

e14623, e15025, e15029<br />

Tkaczuk, Katherine Hanna<br />

1039, 9103, 10572<br />

To, Ka Fai e14085<br />

Tobeña, Maria e14041,<br />

e14104<br />

Tobermann, Anja e17516<br />

Tobimatsu, Kazutoshi 4046<br />

Tobinai, Kensei 8023, 8029,<br />

8050<br />

Toblli, Jorge E e19569<br />

Tobros, Kimberly 6005<br />

Tocchetti, Carlo G. e11052,<br />

e13539<br />

Tochon-Danguy, Henri<br />

e15056<br />

Tod, Michel e15182<br />

Todaro, Matilde e21095<br />

Todd, Jonathan 6032<br />

Todd, Knox H. 6125<br />

Todenhoefer, Tilman e19021<br />

Todor<strong>of</strong>f, Karen e14118<br />

Toepelt, Karin 7603<br />

T<strong>of</strong>anetti, Francesca Romana<br />

e18101, e21094<br />

T<strong>of</strong>fart, Anne-Claire 7529<br />

Toh, Chee-Keong e15064<br />

Toh, Han Chong 4100^<br />

Toh, Uhi e13046, e13050<br />

Toh, Yasushi e14601<br />

Tohnai, Iwai 5556<br />

Tohyama, Osamu e13511<br />

Toi, Masakazu TPS648,<br />

e21004, e21031, e21052<br />

Toi, Yukihiro e19521<br />

Toita, Takafumi 5086,<br />

e15533<br />

Toiyama, Daisuke e15055<br />

Toiyama, Yuji e14029,<br />

e14039, e14541, e21053<br />

Tokes-Fuzesi, Margit e21140<br />

Tokuda, Yutaka 540, 565<br />

Tokumura, Carolina e12041<br />

Tokunaga, Masanori e14635<br />

Tokunaga, Shinya 4002,<br />

e18036, e18042<br />

Tokunaga, Shoji 3558<br />

Tokura, Akemi 5086<br />

Tolaney, Sara M. 1006,<br />

1026, 9084<br />

Tolaney, Sara 535, 3053<br />

Tolba, Khaled A. e16001<br />

Tolcher, Anthony W. 2512,<br />

2555, 3001, 3019, 3025,<br />

TPS3110, e13025,<br />

e13077, e13599, e13602,<br />

e13604<br />

Toledo, Carolina 2515<br />

Toledo, Frederico G.S. 3019<br />

Tolentino, Krysteena e13526<br />

Tolia, Maria e21098<br />

Tollali, Terje 7027<br />

Tollenaar, Rob A e14151<br />

Toller, Isabella 2575<br />

Toloui, Helen Nicole 10525<br />

Toloza, Eric M. 10070<br />

Tolstov, Yanis 8577<br />

Tom, Ashlyn 4045<br />

Tomao, Federica e11039,<br />

e11514, e19620<br />

Tomao, Silverio e11039,<br />

e11514, e12510, e19620<br />

Tomasello, Chiara e18039<br />

Tomasello, Gianluca 4097<br />

Tomasevic, Zoran e18531,<br />

e21030<br />

Tomasevic, Zorica 505,<br />

e18531, e21030<br />

Tomassetti, Paola 4014,<br />

e14647<br />

Tomaszewski, Garin e14690<br />

Tomaszewski, Joseph E.<br />

3031, 10603<br />

Tomatis, Mariano 4110<br />

Tombal, Bertrand 4510, 4652<br />

Tomblyn, Michael B. 2099,<br />

TPS9150<br />

Tomczak, Piotr 4501, 4606<br />

Tome, Yasunori 10072<br />

Tomeczko, Janusz 7090<br />

Tomei, Sara 8576<br />

Tomezzoli, Anna 4076<br />

Tomida, Mikio 10542<br />

Tomii, Keisuke 7528, 10610<br />

Tomilo, Mark e14164<br />

Tomimatsu, Nozomi 10624<br />

Tominaga, Shigeru e18019<br />

Tominaga, Shusei e13060<br />

Tomioka, Hideo e16034<br />

Tomirotti, Maurizio LBA7501<br />

Tomita, Dianne 1062,<br />

e19511<br />

Tomita, Naohiro 3524, 3607<br />

Tomita, Yoshihiko e15057,<br />

e15068, e15071, e15088,<br />

e15098<br />

Tomizawa, Yoshio 7017,<br />

e18037<br />

Tomonaga, Nanae 7569<br />

Toner, Aidan e15126<br />

Toner, Guy C. 4531<br />

Tonev, Anton e21100<br />

Tong, Joanna e14085<br />

Tong, Shan 639<br />

Tong, Wilson P. e18140<br />

Tonini, Giuseppe 3518,<br />

4627, 10063, e14135<br />

Tonkin, Katia Sonia e16528<br />

Tonkin, Katia e15540<br />

Tono, Yasutaka e19546<br />

Tonolla, Sabina 8563<br />

Tonorezos, Emily S. 9552,<br />

9586<br />

Tontchev, Anton e21100<br />

Toomey, Kathleen 8037,<br />

e18556<br />

Toor, Amir A. e18550<br />

Topalian, Suzanne Louise<br />

CRA2509, 2510, 5506,<br />

8507<br />

Topham, Allan 3621<br />

Topilow, Arthur 1540<br />

Topliss, Duncan 5518<br />

Topp, Max S. 8079<br />

Topp, Max 6500^<br />

Topp, Monique 5073<br />

Topp, Stefan A. e21020<br />

Toppo, Laura 4097<br />

Toptan, Tuna 8577<br />

Torbenson, Michael S. 3616<br />

Torchia, James 2513,<br />

e13555<br />

Torday, Laszlo 2522, 3530<br />

Toretsky, Jeffrey 9575<br />

Torigian, Drew A. 7092<br />

Torii, Yoshitaro e18081<br />

Torp, Kari 1504<br />

Torralvo, Helena Fragata<br />

e12021<br />

Torrea, Natalia e14057<br />

Torrecabota, Joan e11036<br />

Torregrosa, Maria Dolores<br />

e11074<br />

Torregroza, Maria Patricia<br />

e14714<br />

Torrejon, Davis Yuri 619<br />

Torrents, A. e13540<br />

Torres, Alejandra e15178<br />

Torres, Barbara 10082<br />

Torres, Esperanza e14115<br />

Torres, Harrys A. e17016<br />

Torres, Karen E. e15124<br />

Torres, Luis M. e19544,<br />

e19567<br />

Torres, Michelle 5076<br />

Torres, Mylin Ann 9122<br />

Torres-Galea, Patricia<br />

e14076, e15013<br />

Torres-Torres, Blanca 1041<br />

Torres-Vigil, Isabel 9025<br />

Torresani, Michele TPS9155<br />

Torri, Valter TPS7109,<br />

e19034<br />

Torricelli, Francesca e18074<br />

Torrisi, Rosalba 623<br />

Torrubia, M S<strong>of</strong>ia 10555<br />

Torsello, Angela e14661<br />

Torta, Riccardo e11524<br />

Tortora, Giampaolo 630,<br />

4076, 4541, e13509,<br />

e14661<br />

Torzilli, Guido 4110<br />

Toscano, Michele 1608<br />

Toschi, Luca 7061, 7585,<br />

e18104<br />

Tosello, Celia e11087<br />

Tosetto, Miriam e13570,<br />

e21024<br />

Toshima, Takeo e14536,<br />

e14602<br />

Tosi, Diego 5069<br />

Tosi, Mario e14010<br />

Toso, John F 5065<br />

Tosolini, Alessandra e13598<br />

Tosti, Giulio e19035, e19036<br />

Toth, Jennifer 7054, 7055,<br />

e17560<br />

Totoonchian, Farhoud 6556<br />

Touboul, Chantal 1566<br />

Touboul, Emmanuel e11551<br />

Tougeron, David 3520<br />

Toulgoat, Frederique 2082<br />

Toumpanakis, Christos<br />

e14696, e17543, e19581<br />

Tountas, Nikolaos e21027<br />

Tourani, Jean Marc 3520<br />

Tourani, Jean-Marc e19555,<br />

e19640<br />

Tournat, Helene e13578<br />

Tournigand, Christophe<br />

2537^, LBA3500^,<br />

e13095^, e13097^,<br />

e19623, e19640<br />

Tourovskaia, Anna e17529<br />

Tousseyn, Thomas 8030<br />

Towers, Russell 10618<br />

Towne, Penny 4041<br />

Townsend, Amanda Rose<br />

3534<br />

Townsley, Carol A. 3013,<br />

e19522<br />

Toy, Elizabeth W. 7562<br />

Toyoda, Masanori e16034<br />

Toyokawa, Hideyoshi e14518<br />

Toyooka, Shinichi 7521,<br />

e17517<br />

Toyota, Brian e13123<br />

TR, Arul Ponni e11536<br />

Trabulsi, Edouard John 4526,<br />

e15012<br />

Tracy, Michael 1009<br />

Traeger, Lara 6106, 9030<br />

Traina, Fabiola 6521<br />

Traina, Tiffany A. TPS668,<br />

1006, TPS1143, TPS1145,<br />

10514<br />

Trakarnsanga, Atthaphorn<br />

3583<br />

Tralongo, Paolo e18002<br />

Tramontano, Angela 6004<br />

Tran, Ben 3082, 4058, 4535,<br />

e13107<br />

Tran, Benjamin 2044<br />

Tran, Hai T. 3598, 5592,<br />

e15075, e15085, e15087<br />

Tran, Huyen 7051<br />

Tran, Namphuong 4521,<br />

4556<br />

Tran, Sinhan 8547, e17558<br />

Trappe, Ralf Ulrich 8030<br />

Trappey, Alfred F 625<br />

Trapsa, Daniela 3013<br />

Trarbach, Tanja 4032<br />

Trassard, Martine 10018<br />

Traut, Alexander 5051<br />

Travers, Debbie 6119<br />

Travis, Lois B. 1536<br />

Travis, William D 7014, 7052<br />

Traynor, Anne M. 5544,<br />

e13537, e16010<br />

Treat, Joseph 7002<br />

Tredan, Olivier TPS662,<br />

1051, 9079, 10562<br />

Tredaniel, Jean TPS7111<br />

Trefzer, Uwe 8501, 8505,<br />

LBA8509, 8518, 8559<br />

Trehu, Elizabeth G. 3105<br />

Treiber, Gerhard 4107<br />

Treisman, Jonathan S e19030<br />

Tremblay, Lise LBA7000<br />

Tremblay-Lemay, Rosemarie<br />

634<br />

796s Numerals refer to abstract number


Trent, Jonathan C. 10088,<br />

e13519, e19520<br />

Treon, Steven P. 8042, 8043,<br />

8106, 8107, e18575<br />

Trepel, Jane B. 3043, 3087,<br />

4569, TPS4701, 7033<br />

Trepel, Martin e15168<br />

Tresca, Patricia TPS662<br />

Tresch, Emmanuelle e14560<br />

Tresguerres, Maria e18044<br />

Trevarthen, David R. 5566<br />

Trevino, Jose Gilberto<br />

TPS4136<br />

Trevino, Kelly Marie 9015<br />

Trevino, Lara Anne 9010,<br />

e16056<br />

Trevisan, Elisa 2037, 2075<br />

Trevisan, Felipe A. e12512<br />

Trevisani, Franco 4115<br />

Tribius, Silke 5512<br />

Tribolo, Antonella e11524<br />

Triche, Timothy J. 4565,<br />

9535, 9564<br />

Tricomi, Marco TPS9155,<br />

10053<br />

Trieu, Vuong N. e15542<br />

Trifan, Ovidiu C. 3615, 7584<br />

Trifanov, Dmitriy e20503<br />

Trifanov, Vladimir e20503<br />

Trifilio, Steven M 4063, 6623<br />

Trigo Perez, Jose Manuel<br />

4630, LBA7000<br />

Triller, Raoul 8578<br />

Trillsch, Fabian 5071<br />

Trimble, Edward Lloyd<br />

TPS5116<br />

Trinh, Long 6587, 6598<br />

Trinkaus, Kathryn 2525<br />

Trinkaus, Martina 6084<br />

Trinkaus, Mateya Elizabeth<br />

e19642<br />

Triolo, Renza e21102<br />

Tripathy, Debu 1065, 1124,<br />

1526, 9144, e19506<br />

Tripodo, Claudio 8083<br />

Trippa, Lorenzo 2005<br />

Tripsas, Christina 8042,<br />

8106, 8107, e18575<br />

Triscott, Joanna e13123<br />

Tritschler, Isabel 2055<br />

Trivanovic, Dragan 9097<br />

Trivedi, Digisha e17009<br />

Trivedi, Meghna S. e18529<br />

Troalen, Frederic 5028<br />

Trocchi, Pietro 580<br />

Troicki, Filip e14157<br />

Trojan, Jorg 4006, 4083<br />

Trojan, Lutz 4539<br />

Trojanec, Radek 1087<br />

Trojanowski, Tomasz 603<br />

Trojniak, Marta Paulina<br />

e15072<br />

Tromp, Brenda J. 2583<br />

Tron, Victor 5032<br />

Troncone, Giancarlo e13509,<br />

e14010, e14042, e18021,<br />

e19038<br />

Troncoso, Patricia 4521,<br />

4556<br />

Trop, Isabelle 616<br />

Trope, Claes 5008<br />

Troppan, Katharina e18525<br />

Trosman, Julia R. 1553,<br />

6120, e16531<br />

Trotman, Judith 9126<br />

Trotti, Andy 5564, 5571<br />

Trouilloud, Isabelle 4018<br />

Troutman, Sarah M 4671<br />

Trova, Vittorio e14094<br />

Numerals refer to abstract number<br />

Troxel, Andrea B. 7092<br />

Truant, Stephanie 3506<br />

Trucco, Matteo Maria 10025<br />

Trucza, Eva e21140<br />

Trudeau, Maureen E. 531,<br />

e11000, e11050<br />

Trudel, Geralyn C. TPS6634<br />

Trudel, Geralyn Carol e15151<br />

Trudel, Suzanne 8013<br />

True, Lawrence D. 4520,<br />

4521<br />

Truemper, Lorenz 6543<br />

Trufelli, Damila 5588<br />

Truini, Mauro e21065<br />

Truman, Matt 7519^<br />

Trump, Donald L. e18118<br />

Trumper, Lorenz H. 8052<br />

Trumpp, Andreas 1090<br />

Trunzer, Kerstin 8503^<br />

Truong, Minh Tam e16059<br />

Truong, Mylene e19645<br />

Truong, Thach-Giao 3058,<br />

4048<br />

Trusk, Patricia B e21120<br />

Tryakin, Alexey e14623,<br />

e15025, e15029<br />

Tryfonopoulos, Dimitrios<br />

e11519, e13520<br />

Trümper, Lorenz 8004<br />

Tryon, Pamela e15114<br />

Trypsianis, Grigorios e17553<br />

Tsai, Chun-Ming 1534,<br />

LBA7500, TPS7614,<br />

e18132<br />

Tsai, Kenneth Yee 8537<br />

Tsai, Ni-Chun 6545, 6547<br />

Tsai, Yi-Fang 1079<br />

Tsang, Janice TPS651,<br />

TPS661<br />

Tsang, Kwong Yok TPS4701<br />

Tsangaris, Theodore N. 1128<br />

Tsao, Anne S. 7078, 7083<br />

Tsao, Che-Kai 4623,<br />

TPS4676, 6065, e21016<br />

Tsao, Claire 8020<br />

Tsao, Ming Sound 1036,<br />

1535, 2051, 7007, 7586,<br />

10522<br />

Tsao, Philip S e21153<br />

Tsao-Wei, Denice D 4575<br />

Tsao-Wei, Denice e15153<br />

Tsapralis, Nikos 10606,<br />

e11073<br />

Tschaika, Marina 4636,<br />

e14152<br />

Tschirschmann, Miriam 5080<br />

Tschoepe, Inga 3104,<br />

e13010<br />

Tseng, Jennifer F. 4059,<br />

e14691<br />

Tseng, Ling-Ming TPS651,<br />

1079<br />

Tsiambas, Evangelos e21098<br />

Tsianakas, Athanasios e19031<br />

Tsiatas, Marinos 5033<br />

Tsigani, Theodora 4123<br />

Tsimafeyeu, Ilya 3070<br />

Tsimberidou, Apostolia Maria<br />

3106, 3107^, 8551,<br />

10510, 10607, e13020,<br />

e13534, e13595, e19004<br />

Tsolaki, Eleftheria 592<br />

Tsottles, Nancy e16061<br />

Tsoukalas, Grecory 10606,<br />

e11073<br />

Tsoukalas, Nikolaos 10606,<br />

e11073, e21098<br />

Tsuboi, Masahiro TPS7110<br />

Tsubota, Noriaki 7080<br />

Tsuchida, Masanori 7012<br />

Tsuchihara, Katsuya 1552,<br />

e14038<br />

Tsuchihashi, Zenta 8593<br />

Tsuchiya, Hiroyuki 10072,<br />

10075, e20505<br />

Tsuchiya, Noriko e19583<br />

Tsuda, Hiroshi 5003<br />

Tsuda, Hitoshi TPS1147,<br />

5085, 10519<br />

Tsuda, Masahiro 4002<br />

Tsugawa, Koichiro 590<br />

Tsui, Dana 544<br />

Tsuji, Akihito 3558, e14510<br />

Tsuji, Wakako e21052<br />

Tsuji, Yasushi e14062<br />

Tsujimura, Akira e15065,<br />

e15069<br />

Tsujimura, Tohru 7080<br />

Tsujino, Kayoko e15533<br />

Tsujino, Kazuyuki 7000,<br />

e18128<br />

Tsujitani, Shunichi e14033<br />

Tsukamoto, Kazuhiro 7569<br />

Tsukamoto, Tadashi e14518<br />

Tsukamoto, Taiji e15048<br />

Tsukasaki, Kunihiro 8050<br />

Tsumura, Takehiko 4002<br />

Tsunoda, Takuya e13014,<br />

e13015<br />

Tsurusawa, Masahito 9574<br />

Tsurutani, Junji TPS659<br />

Tsutani, Yasuhiro 7031<br />

Tsuya, Asuka 7088, e21007<br />

Tsuyuki, Shigeru 588<br />

Tsuzuki, Tomoyuki e14596<br />

Tsvirkun, Viktor e14717<br />

Tsymzhitova, Natalia e20501<br />

Tu, Dongsheng 524, 3504,<br />

3515, 3572<br />

Tu, Eugene e21047<br />

Tu, Shi-Ming 4533<br />

Tu, Wen-yong 5593<br />

Tubbs, Raymond R. 620,<br />

e18085<br />

Tubiana-Mathieu, Nicole<br />

1051, LBA3500^<br />

Tucci, Marco 10063<br />

Tuck, Alan 1123<br />

Tuck, David P. 10558<br />

Tucker, Eric e21122<br />

Tucker, Margaret A. 1603<br />

Tucker, Thomas 5095<br />

Tucker-Seeley, Reginald D<br />

6012<br />

Tuda, Banri 565<br />

Tudur-Smith, Catrin TPS8605<br />

Tuerk, Ingolf e15123<br />

Tufa, Gemechu 580<br />

Tuff, Alice TPS10633^<br />

Tufman, Amanda TPS7617<br />

Tumedei, Maria Maddalena<br />

10601<br />

Tumolo, Salvatore 3547<br />

Tumulo, Salvatore e14711<br />

Tun, Nay Min e13099,<br />

e18024<br />

Tunariu, Nina 2530, 3080,<br />

10038, e15134<br />

Tuncer, Ersin e11083<br />

Tung, Nadine M. 1009<br />

Tung, Stewart Yuk 4105,<br />

5511<br />

Tunkyi, Adrian e21078<br />

Tuominen, Rainer 10521<br />

Turak, Esra Ermis e14670<br />

Tural, Deniz e21143<br />

Turan, Mehmet Ozhan 5055<br />

Turbiez, Isabelle TPS662<br />

Turbiglio, Marco 10055<br />

Turcich, Michelle e13106<br />

Turco, Diana L. 1549<br />

Turcott, Jenny 7068, e19594<br />

Turcotte, Simon e14179<br />

Turhal, Serdar 9050,<br />

e14526, e16015<br />

Turik, Michael 2516<br />

Turin, Ilaria e13103<br />

Turk, Mary 577<br />

Turkbey, Ismail B. TPS4701<br />

Turker, Ibrahim e11540<br />

Turki, Hajer 4087<br />

Turley, Helen 1066<br />

Turna, Hande e19024,<br />

e21143<br />

Turnbull, Barry e18575<br />

Turner, Christopher D. 6503<br />

Turner, Crystal 9019<br />

Turner, Evadne Jane 7604<br />

Turner, Gareth D 4599<br />

Turner, Nancy A 9562<br />

Turner, Robert e21047<br />

Turner, Sandra TPS4690<br />

Turnwald, Brian 6109,<br />

e16536<br />

Turpin, Anthony e11500<br />

Turriziani, Adriana e16553<br />

Turtiaien, Tarita TPS10633^<br />

Turturro, Francesco 8093<br />

Tuscano, Joseph M. e13573<br />

Tuthill, Ralph 8504<br />

Tutrone, Ronald TPS4697<br />

Tutt, Andrew 10586<br />

Tuttle, Robert Michael 5509^<br />

Tuveson, David A. 4056<br />

Tveit, Kjell Magne 3548<br />

Tvsvgk, Tilak 9580<br />

Twardowski, Przemyslaw<br />

3108, 4506, 4511, 4547,<br />

4663, 10503, e15084,<br />

e15114, e15178<br />

Twelves, Christopher<br />

TPS1140^, 2540, 2552,<br />

3522, 3523, 3525,<br />

e13083<br />

Twist, Clare 9533<br />

Tyagi, Seema e17000<br />

Tye, Lesley 7508<br />

Tykodi, Scott S. 2510,<br />

e21148<br />

Tyler, Allison Janine e15208<br />

Tyler, Carl V 1598<br />

Tyler, Douglas S. 10563,<br />

e14724<br />

Tymrakiewicz, Malgorzata<br />

1545<br />

Tyner, Jeffrey 9555<br />

Tyree, Seth D 5539<br />

Tyroller, Karin LBA7000<br />

Tysarowski, Andrzej 8548<br />

Tyson, Leslie B. e19647<br />

Tyszkiewicz, Tomasz e14159<br />

Tzabari, Moran 2556, e13080<br />

Tzanis, Kimon e15040<br />

Tzen, Kai-Yuan e14576<br />

Tzovaras, Alexandros 10606,<br />

e11073, e21098<br />

U<br />

Uberti, Joseph P. 6549<br />

Uccellini, Lorenzo 8576<br />

Ucci, Giovanni e19592<br />

Uchida, Junji e18025<br />

Uchida, Keiichi e14029,<br />

e14541, e21053<br />

Uchida, Toshiki 8023<br />

Uchiyama, Hideaki e14602<br />

Uchiyama, Tetsuyuki e14011<br />

797s


Uchiyama, Tomotaka e21108<br />

Udovitsa, Dmitry TPS651<br />

Ueda, Eiji e15098<br />

Ueda, Shinya 4002<br />

Ueda, Takashi e15055<br />

Ueda, Takeshi 4503<br />

Uehara, Kanou e11530<br />

Uehara, Masahiro TPS659<br />

Uehara, Takeshi 4111<br />

Uemura, Hiroji 4626<br />

Uemura, Hirotsugu e15048,<br />

e15098<br />

Uemura, Motohide e15065,<br />

e15069<br />

Ueno, Hideki 3625, 4035,<br />

e14506<br />

Ueno, Makoto 4031<br />

Ueno, Masashi 3625, e14145<br />

Ueno, Naoto T. 594, 1047,<br />

1102<br />

Ueno, Takayuki e21031,<br />

e21052<br />

Ueoka, Hiroshi 7042<br />

Uesugi, Mai e13511<br />

Uetake, Hiroyuki e14127<br />

Uges, Donald R.A. 4528<br />

Uharek, Lutz 6543<br />

Uhm, Joon H. 2031<br />

Uike, Naokuni 8023<br />

Ujiie, Hideki 10542<br />

Ukimura, Osamu e15060,<br />

e15212<br />

Ulasov, Ilya 2014<br />

Ulaszek, Jodie 6535^<br />

Ulcickas Yood, Marianne<br />

1526, 1590, 1591<br />

Ule, Ulla Jo 9061<br />

Uleer, Christoph 552<br />

Ulhoa-Cintra, Alice e21036<br />

Ulivi, Paola e18045<br />

Ulivieri, Alessandra e19033<br />

Ullmann, Martin TPS2622<br />

Ulloa, Adolph e11549<br />

Ulloa, Victor e12041<br />

Ullrich, Stephen 8537<br />

Ulmer, Hans Ulrich 1602<br />

Ulrich, Jens e19031<br />

Umeda, Tomoko e11564<br />

Umekawa, Kanako e17538<br />

Umemura, Shigeki 1552,<br />

7044, e17513, e17526,<br />

e17554, e18054, e18064<br />

Umesh, Anita e21159<br />

Unal, Emel e20000<br />

Uncu, Dogan e13031,<br />

e14526<br />

Underhill, Craig 3572,<br />

TPS4137<br />

Underwood, Willie e15046,<br />

e15204<br />

Undevia, Samir D. 10003,<br />

10028<br />

Unger, Jackie e11550,<br />

e17008<br />

Unger, Joseph M CRA6009,<br />

7570, 8001, 9018<br />

Unger, Pamela D e15191<br />

Unno, Michiaki e14011<br />

Untch, Brian R. e14724<br />

Untch, Michael 523, 541,<br />

1114<br />

Upadhyay, Sunil 7562, 7583<br />

Upadhyaya, Gargi H. 6591<br />

Upanal, Nazeerahamad N.<br />

e14166<br />

Ura, Takashi 4002, e14123,<br />

e14500<br />

Urakami, Shinji e15038<br />

Uram, Jennifer e14585<br />

Uramoto, Hidetaka 10585<br />

Urata, Yoshiko e18060<br />

Urba, Susan e16014<br />

Urba, Walter John 8508<br />

Urbach, David 6027<br />

Urbain, Remi 5069<br />

Urbanova, Dagmar 9592<br />

Urbanowitz, Gladys TPS4683<br />

Urbauer, Diana 1520, 6054,<br />

9134<br />

Urbine, Terry Francis e17529<br />

Urbini, Benedetta 2057<br />

Urbini, Milena 10087<br />

Uren, Aykut 9575<br />

Urken, Mark 5581<br />

Urun, Yuksel 1572, 1596<br />

Urva, Shweta Ramesh 3099<br />

Urzua, Lorena e12027,<br />

e15527<br />

Usakova, Vanda e15027,<br />

e15028<br />

Usari, Tiziana 2610<br />

Uselman, Ryan R. e13590<br />

Usharani, Malur R. 1093<br />

Ushijima, Kimio 5006, 5085<br />

Uslu, Ruchan 3565<br />

Usmani, Saad Zafar 8041,<br />

TPS8116, e18548, e18576<br />

Usó, Marta 7058, 7060,<br />

10594<br />

Ustoyev, Yelena 10002,<br />

10004<br />

Ustuner, Zeki e21050<br />

Usui, Kazuhiro 7086, 7561,<br />

7565<br />

Usui, Noriko 8050<br />

Usul Afsar, Cigdem e13523<br />

Uthe, Regina 610<br />

Utiger, Urs e19648<br />

Utikal, Jochen LBA8509<br />

Utkan, Gungor 1572, 1596<br />

Utsumi, Tomoki e18126<br />

Uttenreuther-Fischer, Martina<br />

Maria 606, TPS649,<br />

TPS651, 3104<br />

Uyeji, Jennifer 8596<br />

Uyeturk, Ummugul e11540<br />

Uygun, Kazim e14618<br />

Uysal Sonmez, Ozlem e11540<br />

Uysal, Mukremin e14070<br />

Uyterlinde, Wilma 7019<br />

Uzan, Catherine 5083, 10098<br />

Uzunaslan, Didem 10514<br />

Uzzan, Bernard e18003,<br />

e18020<br />

Uzzo, Robert G. 4500, 4526<br />

V<br />

V, Murali Subramanian<br />

e11536<br />

Vaccarella, Salvatore e14042<br />

Vaccaro, Lisa e16507<br />

Vaccaro, Vanja 630, 4541,<br />

e14661<br />

Vachani, Anil 7595<br />

Vachani, Carolyn 6135<br />

Vadell, Catalina 1531<br />

Vadhan-Raj, Saroj 4037,<br />

9002, e19520<br />

Vaena, Daniel A. e15010,<br />

e15143<br />

Vaessen, Christophe e15105<br />

Vago-Ady, Nora TPS662,<br />

8546<br />

Vahanian, Nicholas N. 2501,<br />

2571, 4049, e19008<br />

Vahdat, Linda T. 1010<br />

Vaidya, Bhavesh 7092<br />

Vaillant, Brian D. 2002<br />

Vaishampayan, Ulka N. 4,<br />

4506, 4525, 4536, 4538,<br />

e15081<br />

Vaishnaw, Akshay 3062<br />

Vaissiere, Nathalie 7001<br />

Vajpayee, Neerja 6572,<br />

e18535<br />

Vakar-Lopez, Funda e15207<br />

Vakiani, Efsevia TPS4144<br />

Valachis, Antonis 5066<br />

Valadares, Adriana D e16046<br />

Valdes, Frances 3108<br />

Valdez, Riccardo 8053<br />

Valdiviezo, Natalia e15033<br />

Valduga, Francesco e14711<br />

Valea, Fidel A. 5101<br />

Valencia, Alfonso 3068<br />

Valent, Alexander 10556<br />

Valent, Francesca 1044,<br />

e13058, e13070<br />

Valenti, Marco 4572<br />

Valentine, Erin 3587<br />

Valentini, Antonella 2057<br />

Valentino, Joseph e16035<br />

Valeria, Visconte 6521<br />

Valerio, Maria Rosaria e11056<br />

Valero, Vicente 515, 527,<br />

594, 1029, 1104, 10613<br />

Valgus, John 6042<br />

Valigi, Paolo e21096<br />

Vallabhaneni, Geetha D.<br />

7048, e14169, e14170<br />

Valladares Ayerbes, Manuel<br />

e15079<br />

Valladares-Ayerbes, Manuel<br />

e14595, e21002<br />

Valle, Juan W. 4118, 4121,<br />

4122, e14625, e21037<br />

Vallee, Audrey e21117<br />

Vallejo, Carlos Teodoro<br />

e11021<br />

Vallejos Sologuren, Carlos<br />

e18524<br />

Vallejos, Carlos S. e11518,<br />

e15036<br />

Vallejos, Carlos 2097<br />

Valleriani, Mario e19002<br />

Vallier, A-L TPS660,<br />

TPS1144<br />

Vallone, Marcy TPS3640<br />

Valluri, Satish e14013,<br />

e14016, e14106, e14141,<br />

e14146, e15183<br />

Valmadre, Giuseppe TPS7109<br />

Valo, Isabelle 10018<br />

Valori, Vanna Maria e21092<br />

Valota, Olga e15070<br />

Valsecchi, Matias Emanuel<br />

8560, e19016<br />

Valsecchi, Matias E 536<br />

Valteau Couanet, Dominique<br />

9560, 9572<br />

Valverde, Claudia María<br />

10052, e21021<br />

Valverde, Ivana 4579<br />

Valvo, Francesca e21118<br />

Vamvakas, Lampros 7564<br />

van ’t Veer, Laura J. 1022<br />

Van Akkooi, Alexander<br />

Christopher Jonathan 8535,<br />

e19022<br />

Van Allen, Eliezer Mendel<br />

10502<br />

Van Alstine, Lindsay Joy 4532<br />

Van Arkel, Cornelis 4539<br />

van Berge Henegouwen, Mark<br />

I. 4094<br />

van Berkel, Cees e19617<br />

van Besien, Koen 6535^,<br />

e13120^<br />

Van Binst, An 2050<br />

Van Brunt, Kate e16528<br />

Van Brussel, Thomas 1020<br />

van Coevorden, Frits 10096<br />

Van Cutsem, Eric 3502,<br />

CRA3503, 3505, 3509,<br />

3532, 3539, 3562, 3574,<br />

3578, 3579, 3581, 3591,<br />

3602, 4007, 4081, 4118,<br />

TPS4134<br />

van de Vaart, Paul 7019<br />

Van de Velde, Ann 2506<br />

van De Velde, Cock JH 10518<br />

Van De Velde, Cornelis J. H.<br />

516, 517, 526<br />

Van De Velde, Helgi 2583,<br />

8018^<br />

Van de ven, Carmella<br />

e13022, e21150<br />

Van Den Abbeele, Annick D.<br />

510<br />

van den Akker, Jeroen 1022<br />

Van Den Bent, Martin J. 2<br />

Van Den Brink, Marcel R. M.<br />

6539<br />

van den Eertwegh, Alfons JM<br />

2562, TPS4689<br />

Van den Eynde, Marc 3559,<br />

e14044, e14060<br />

van den Heuvel, Michel M<br />

7019<br />

van den H<strong>of</strong>f, Joerg 10068<br />

Van Den Neste, Eric 8046<br />

Van Den Wyngaert, Tim 1129<br />

van der Elst, A 7009<br />

Van Der Graaf, Winette T.A.<br />

2593, 3035, 5061, 9036,<br />

10009<br />

Van Der Hoeven, Dharini<br />

e13516<br />

Van Der Hoeven, Jacobus<br />

TPS10632<br />

van der Holt, Bronno 10091<br />

van der Meulen, Egbert<br />

e15199^<br />

van der Noll, Ruud e13596<br />

van der Straaten, Tahar 526<br />

van der Werf-Langenberg,<br />

Mirte e16526<br />

Van Deventer, Hendrik<br />

e13074<br />

van Diest, Paul J. 562<br />

van Doorn, Leni 4132<br />

Van Driessche, Ann 2506<br />

Van Es, Suzanne C. 10581<br />

Van Ewijk, Reyn 1082<br />

van Goethem, Mireille 1129<br />

van Haaften, Gijs e13515<br />

Van Hazel, Guy A. 3515<br />

Van Hazel, Guy 3505, 4118<br />

van Herk-Sukel, Myrthe<br />

e16526<br />

Van Herpen, Carla 2593,<br />

5061, 8511, 9070<br />

Van Hummelen, Paul 2020<br />

Van Imh<strong>of</strong>f, Gustaaf Willem<br />

8039<br />

van Ingen, Hein 3026<br />

Van Laarhoven, Hanneke<br />

W.M. 3035, 9070, e13111,<br />

e19560<br />

Van Laere, Steven J. 1047<br />

Van Laethem, Jean Luc 3559,<br />

4006<br />

Van Laethem, Jean-Luc 2583,<br />

4050, 4053, TPS4135<br />

798s Numerals refer to abstract number


Van Le, Linda 1519, 2591,<br />

5050, e13063<br />

van Leeuwen, Flora E. 8039<br />

van Lessen, Antje e15053<br />

Van Loan, Marta 4547<br />

Van Loon, Katherine 4038<br />

Van Looy, Thomas 10030<br />

van Marck, Veerle 1129<br />

Van Meerbeeck, Jan P. 7081<br />

Van Nuffel, An M. T. 2507<br />

van Oort, Inge M e15112<br />

Van Poppel, Hendrik e13061<br />

Van Puyvelde, Katrien 9035<br />

Van Remortel, Piet e13073<br />

Van Rhee, Frits 8041,<br />

e18548, e18576<br />

Van Riet, Ivan 2507<br />

van Schaik, Ron H. N. 526<br />

Van T Veer, Laura 10597<br />

Van Tendeloo, Viggo F. 2506<br />

Van Tendeloo, Viggo e13051<br />

van Tets, W F 7009<br />

Van Tine, Brian Andrew<br />

TPS10099, TPS10103<br />

van Tinteren, Harm 1080,<br />

e13028<br />

Van Veldhuizen, Peter J.<br />

3078, 4511, 4663, 10503,<br />

e13535, e13551<br />

Van Veldhuizen, Peter 4510,<br />

e15115<br />

Van Vlieberghe, Hans 4006<br />

Van Vugt, Vincent 2052<br />

Van Wert, Michael 9132<br />

van’t Veer, Laura 3065<br />

VanCriekinge, Mark E. 4660<br />

Vandam, Jacquez e14679<br />

Vander Velde, Nancy S. 6600<br />

Vanderburg, Sky 6102<br />

Vandermeer, Lisa 10620<br />

Vanderschaeghe, Simon 8532<br />

Vanderwalde, Ari M. 5502<br />

VanderWalde, Noam Avraham<br />

5539<br />

Vanderweele, Tyler J. 9116,<br />

9132<br />

Vandewalker, Kristen MN<br />

e21026<br />

Vandoni, Irene 8046<br />

VanDraanen, Leah e15180<br />

Vanhoefer, Udo J. e13100^<br />

Vank, C. e13597<br />

Vankayala, Hema M. e15081<br />

VanLaeken, Nancy 9043<br />

Vanlemmens, Laurence 1051,<br />

e11012<br />

Vanmali, Vaibhav 1108,<br />

e11552<br />

Vanmany, Sengdao e21111<br />

Vannier, Jean-Pierre e21156<br />

Vannier, Michael 4022<br />

Vannucchi, Alessandro M.<br />

6514^, 6626^, TPS6641,<br />

TPS6642^, TPS6643^<br />

Vannucci, Jacopo e21094<br />

Vanorle, Katrien 9035<br />

Vansteenkiste, Johan F. 3007,<br />

6058, 7013, e18100<br />

Vansteenkiste, Johan 7503<br />

Vaporciyan, Ara A. 4086,<br />

7043<br />

Vaquer, Jose 2515<br />

Vaquero, Eva Cristina e21035<br />

Varadan, Vinay 10508<br />

Varadarajan, Prakash 5541<br />

Varadhachary, Gauri R. 4051,<br />

4054, 4130, 4131, 10584<br />

Vardakis, Nikolaos e18105<br />

Vardhanabhuti, Saran 1539<br />

Numerals refer to abstract number<br />

Vardy, Janette L. 4644, 9021<br />

Varela Ferreiro, Silvia<br />

e18040, e18146<br />

Varela Ponte, Rafael e14733,<br />

e19505<br />

Varela, Silvia e18070<br />

Varella-Garcia, Marileila<br />

7061, 7504, 7508, 7534,<br />

7552, 7589, 10580,<br />

e14071, e21018<br />

Varga, Andrea 3012, e13118<br />

Vargas, Carlos A. e12039<br />

Varghese, Anna M. 7052,<br />

7527, 7580<br />

Vargo, Dennis e13041<br />

Vari, Sabrina e14661<br />

Varin, Rémi e21156<br />

Varlet, Pascale TPS9596<br />

Varlotto, John M. 7040,<br />

7054, 7055, e17560<br />

Varma, Ankur 527<br />

Varricchio, Clara 1500<br />

Varughese, Joyce e13078<br />

Vasconcelos, Ines 5053,<br />

5070<br />

Vashi, Pankaj G e14012<br />

Vasile, Enrico 10611<br />

Vasovic, Suzana 9046<br />

Vassal, Gilles TPS9596<br />

Vassel, Svetlana 6123<br />

Vassilaki, Ismini 8588<br />

Vassilakopoulou, Maria 573,<br />

5576<br />

Vassilev, Lyubomir T e13600^<br />

Vassilopoulou-Sellin, Rena<br />

4641<br />

Vastola, Francesca e13019<br />

Vatandoust, Sina e19513<br />

Vaughan, E. Darracott e15196<br />

Vaughn, David J. 1503,<br />

1539, 4522, 4596, 9145<br />

Vauthey, Jean-Nicolas 3598,<br />

4116<br />

Vavala, Tiziana TPS7619,<br />

e18103<br />

Vay, Christian e21020<br />

Vaz, M. Angeles e21104<br />

Vazquez Estevez, Sergio<br />

TPS4672, e12012,<br />

e18040, e18055, e18146<br />

Vazquez Osorio, Jose Lorenzo<br />

e11028<br />

Vazquez Sequeiros, Enrique<br />

e12033<br />

Vazquez, Adrienne Marie<br />

e17564<br />

Vazquez, Francisca 10534,<br />

e14733<br />

Vazquez, Jesus e13105<br />

Vazquez-Estevez, Sergio<br />

e15067, e15076, e15077,<br />

e17545, e18070<br />

Vecchiarelli, Silvia e14647<br />

Veccia, Antonello e14711,<br />

e15160<br />

Vecht, Charles J 2<br />

Vedrine, Lionel 1580<br />

Veenstra, David L e16524<br />

Vega, Estela e15033, e15552<br />

Vega, Iranzazu 4079<br />

Vega-Saenz de Miera, Eleazar<br />

8544, 8565<br />

Vehling-Kaiser, Ursula 3519^,<br />

3588, TPS3639, 4023,<br />

4072<br />

Veit-Friedrich, Iris 9573<br />

Velasco, Roser 9090<br />

Velcheti, Vamsidhar e21106<br />

Velculescu, Victor E. 3068<br />

Velehorschi, Wiorika 7056<br />

Velez, Michel e17564<br />

Vella, Alessandro 1500<br />

Vella, Laura J 8542<br />

Vellayappan, Balamurugan A<br />

5553<br />

Vellos, Ted e15014<br />

Velten, Michel 1584, e11542<br />

Veltri, Enzo e19501<br />

Vemuganti, Geeta K 9568<br />

Vendel, Yvette e11012<br />

Vendrely, Veronique 4091<br />

Venegas, Diego 2531,<br />

e12041, e12043<br />

Vengco, Isabelita 2589<br />

Venhaus, Ralph Rudolph<br />

2589, TPS8111, 8547,<br />

e17558<br />

Venkannagari, Sreedhar<br />

10549<br />

Venkatakrishnan, Karthik<br />

2597<br />

Venkataraman, Ganesh 4056<br />

Venkatramani, Rajkumar 9564<br />

Venna, Suraj S. e19000,<br />

e19025<br />

Venner, Peter 4<br />

Vennin, Philippe e11500,<br />

e12502<br />

Venook, Alan Paul 3105,<br />

3528, 4102, e14581,<br />

e19046<br />

Venselaar, Hanka 9550<br />

Ventrucci, Cristiano 3061<br />

Venturi, Claudia 6531<br />

Venturini, Filippo 3570<br />

Venturini, Marco 1073,<br />

e19592<br />

Venturino, Paola 7577<br />

Venugopal, Parameswaran<br />

6552, e16503<br />

Vera Badillo, Francisco Emilio<br />

6043<br />

Vera, Ruth 3586, e14097,<br />

e14120, e21086<br />

Verdail, Francys C 7008<br />

Verdeguer, Amparo 10082<br />

Verduyn, Cora e19010<br />

Veremeychuk, T. e11528<br />

Vergamini, Lucas B. 9523<br />

Vergara, Ismael A. 4565<br />

Vergara, Lori 8073<br />

Verger, Eugenia 2045<br />

Vergidis, Joanna 3560<br />

Vergnenegre, Alain 7522,<br />

7540, 7542, 7574,<br />

e16560, e18130<br />

Vergo, Maxwell Thomas 4102<br />

Vergote, Ignace B. LBA5000,<br />

LBA5002^, 5034, 5038,<br />

5052, 5058, 5071, 5072,<br />

5087, TPS5112, e13095^,<br />

e13097^, e13108^<br />

Verheij, Marcel 7019<br />

Verheul, Henk M.W. e18094<br />

Verhoef, Gregor E.G. 8030<br />

Verhoef, Gregor 8065<br />

Verma, Ashok Kumar e16023<br />

Verma, Sunil LBA1, e16547,<br />

e16558<br />

Verma, Udit N. 3605<br />

Verman, Radha e21061<br />

Vermeij, Joanna 4622<br />

Vermeulen, Jessica 2583<br />

Vermorken, Jan Baptist 562,<br />

5504^, 5516^, TPS5599,<br />

8532<br />

Vermund, Sten H. 9558<br />

Verna, Elizabeth 4101<br />

Vernick, Jerome 1540<br />

Veronese, Maria Luisa 8517<br />

Veronese, Silvio LBA7501<br />

Veronesi, Umberto 1500<br />

Verrecchia, Francesco<br />

e19035, e19036<br />

Verri, Cristian 4572<br />

Verrico, Monica e11039,<br />

e11514, e12510, e19620<br />

Verrielle, Veronique 543<br />

Verrill, Clare 4599<br />

Verschraegen, Claire F. 3009<br />

Verschuur, Arnauld 9517<br />

Verslype, Chris 4007,<br />

TPS4148<br />

Verso, Melina e19612<br />

Verstegen, Naomi E 7009<br />

Verstovsek, Srdan 6587,<br />

6624, TPS6642^,<br />

TPS6643^<br />

Vertakova-Krakovska, Bibiana<br />

e15028<br />

Vervenne, Walter 4539<br />

Vervliet, Johan 1129<br />

Verweij, Jaap 2540, 2593,<br />

10013, 10054, 10091<br />

Verzoni, Elena 4629<br />

Vescio, Robert A. 8098,<br />

e18558<br />

Vesole, David H. 8011<br />

Vessella, Robert 4520,<br />

e15113<br />

Vestermark, Lene Weber<br />

4015, e14517<br />

Vetto, John T. 8504<br />

Vettori, Josiane Cheli e19628<br />

Vey, Norbert 6523, TPS6637,<br />

e18526<br />

Veys, Isabelle e21010<br />

Viada, Carmen 2527<br />

Viale, Agnes 4527<br />

Viale, Giuseppe 504, 1022,<br />

4120<br />

Viana, Benjamin e14158<br />

Viana, Luciano S. e14599<br />

Viardot, Andreas 6500^,<br />

8025<br />

Viau, Philippe e15574<br />

Vicario, Giovanni e19595<br />

Vicente, Angeles e11024<br />

Vicente, David 8062<br />

Vicente, Vicente 7095,<br />

e11024<br />

Vichaya, Elisabeth G. 6551,<br />

9082, 9118, 9140<br />

Vicioso, Luis 10500, e14115<br />

Vickers, Andrew J. 4668<br />

Vickers, Michael M. e14073<br />

Vickers, Selwyn M. 6016<br />

Victor, Charles e16547<br />

Victor, Joel 4098<br />

Victoria, Iván 3536<br />

Victorino, Ana Paula Ornelas<br />

e14101<br />

Vidal Boixader, Laura<br />

e13540, e15575<br />

Vidal, Jose e18053<br />

Vidal, Laura 5068<br />

Vidal, Liath 548<br />

Vidal, Maria 509, 566, 619<br />

Vidal, Michel e13056<br />

Vidal, Silvia e15527<br />

Vidal, Yolanda 10534,<br />

e14733, e19505<br />

Vidal-de-Miguel, Guillermo<br />

1570<br />

vidal-Millan, Silvia 6599<br />

Vidaurre, Tatiana e11518<br />

Vidaurreta, Javier e15507<br />

799s


Videtic, Gregory M.M.<br />

e14611, e14665<br />

Vidiri, Antonello 2085<br />

Vieillefond, Annick 4583<br />

Vieira, Fernando Meton 7506,<br />

e14101<br />

Vieira, Nivaldo Farias e12512,<br />

e19628<br />

Vieira, Thibault e18075,<br />

e18102<br />

Vieito, Maria 10534, e14733,<br />

e19505<br />

Viens, Patrice TPS1613<br />

Vierling, John 4116, e19631<br />

Vieser, Eva 2504<br />

Vigani, Antonella e17533<br />

Viganò, Maria Grazia 7076,<br />

7085<br />

Vigil, C. E. 2097<br />

Vigil, Carlos Enrique 6602<br />

Vigil, Carlos E 6565<br />

Vigna, Leonardo e19033<br />

Vigna, Paolo Della e14607<br />

Vigneau, Fawn D. 7037<br />

Vigneron, Daniel B. 4660<br />

Vignetti, Marco 6531<br />

Vignot, Stéphane 641, 6110,<br />

7529, e11019<br />

Vigouroux, Stéphane<br />

6534<br />

Vigouroux, Stephane 6542<br />

Viguier, Jérôme 1567, 1568,<br />

e14129<br />

Vij, Brinder e19566<br />

Vij, Ravi 8011, 8016, 8020,<br />

8035, TPS8112, TPS8114,<br />

e18551<br />

Vijarnsorn, Chodchanok 9529<br />

Viktil, Ellen e14637<br />

Vila, Luis 5590<br />

Vilardell, Felip 4089<br />

Vilardell, Loreto 1588<br />

Vilas Boas, Viviane e11059<br />

Vilcot, Laurence 10044<br />

Vilgrain, Valérie 4618,<br />

e14553<br />

Villa Guzman, José Carlos<br />

TPS4674<br />

Villa, Diego 549^<br />

Villa, Eugenio 4017<br />

Villà, Salvador e12504<br />

Villablanca, Judith 9533<br />

Villacampa, Felipe 4584,<br />

4620<br />

Villaflor, Victoria Meucci 5500<br />

Villalona-Calero, Miguel Angel<br />

5020<br />

Villalva, Claire 3520<br />

Villamil, Beatriz Perez 10564<br />

Villani, Gina M. e13099<br />

Villano, John L. 2073,<br />

e12036<br />

Villanueva Silva, Maria Jose<br />

10608, e11535, e18040,<br />

e18146<br />

Villanueva, Cristian 1011,<br />

5504^, e11019<br />

Villanueva, Geraldine e19594<br />

Villanueva, Noemi e18044<br />

Villanueva, Rafael e18030<br />

Villanueva, Ronald 1540<br />

Villareal, Reina 1558<br />

Villarin, Eligie 584<br />

Villarreal-Garza, Cynthia<br />

Mayte 1607, e11016,<br />

e12027<br />

Villasboas, Jose Caetano 595,<br />

608<br />

Villasboas, Tercia Reis<br />

e12021<br />

Villatoro, J Carlos 5595<br />

Villatoro, R. 4630, e14116<br />

Villeda, Virgilio Alexander<br />

e11031<br />

Villella, Jeannine A e15504<br />

Villines, Gary e18576<br />

Vilmar, Adam 10617<br />

Vin, Harina 8537<br />

Vinas, Elaine e19646<br />

Vincent, Andrew D. 562,<br />

7019<br />

Vincent, Linda V. e11096<br />

Vincent, Mark David e21120<br />

Vincent, Tiffaney 9506<br />

Vincent-Salomon, Anne 601<br />

Vincenzi, Bruno 4627,<br />

10063, e14135<br />

Viner, Jaye TPS2618,<br />

TPS2621, e18073<br />

Vinholes, Jeferson J.F. 606<br />

Vinik, Aaron 4118<br />

Vinks, A.A. 9541<br />

Vinogradova, Liliya Igorevna<br />

e15543<br />

Vinolas, Nuria 1531, e12000,<br />

e12012, e18011<br />

Viny, Aaron D 10021<br />

Vinyals, Juan M 10052<br />

Violante, Assunta e19002<br />

Viqueira, Andrea e15111<br />

Virchow, Isabel 2086<br />

Viret, Frédéric LBA3500^,<br />

4032<br />

Virgo, Katherine S. 1100,<br />

1608, 6006, 6008, 6079,<br />

6081, e15528, e19593<br />

Virk, Navneet 4563<br />

Virk, Shakeel 3515<br />

Virzì, Vladimir 4627<br />

Visa, Laura 4079, e21035<br />

Viscusi, Johnny e13598<br />

Vishnubhatla, Sreenivas<br />

5104, 9580<br />

Vishnubhatla, Sreeniwas 6593<br />

Viskov, Christian e13117<br />

Visser, Otto 8039<br />

Visseren Grul, Carla M. 7554<br />

Visseren-Grul, Carla LBA7507<br />

Vistisen, Kirsten 3538<br />

Visvanathan, Kala 521<br />

Visvikis, Antonios e15040<br />

Viswanathan, Chitra e19645<br />

Viswanathan, Shankar e16058<br />

Vital, Anne 2023<br />

Vitale, Alessandro e14563<br />

Vitale, Antonella 6531<br />

Vitalini, Cristina e19549<br />

Vitek, Pavel e14631<br />

Viterbo, Rosalia 4526<br />

Viteri Ramirez, Santiago<br />

3093, 7540, e18131,<br />

e18137, e18143<br />

Vito, Courtney 1035, 1117<br />

Vitolins, Mara 9108<br />

Vitolo, Umberto 8006<br />

Viudez, Antonio e14057,<br />

e14120, e21086<br />

Vivancos, Ana 509, 3014,<br />

10511<br />

Vivenza, Daniela 2600<br />

Viviers, Louis TPS9596<br />

Vlachos, George 5033<br />

Vladimirova, Liubov Yu<br />

e11558, e20503<br />

Vladislav, Ioan Tudor 7106<br />

Vlahovic, Gordana 3077,<br />

e13603<br />

Vletter - Bogaartz, Judith M.<br />

526<br />

Vnencak-Jones, Cindy L 7532<br />

Vo, Lien e14164<br />

Vocke, Cathy TPS4680<br />

Voduc, David 1008<br />

Voegeli, Anne-Claire 10507<br />

Voehringer, Matthias<br />

TPS3641^<br />

Voest, Emile E. 2540, 2550,<br />

2596, e13028, e13596<br />

Vogel, Ulrich 1067<br />

Vogelbaum, Michael A. 2018,<br />

2076, 2100, 2101<br />

Vogelius, Ivan Richter e18527<br />

Vogelzang, Nicholas J. 4,<br />

4511, LBA4512, 4513,<br />

4525, 4547, 4551, 4558,<br />

4662, 4663, 4665,<br />

TPS4675^, TPS4683,<br />

TPS4686, 7514, 10503,<br />

e15063, e15089, e15210<br />

Vogl, Steven E. e11005<br />

Vogl, Thomas J. e14131<br />

Vogl, Ursula Maria e15090<br />

Vogt, Andreas 8577<br />

Vogt, Luc 9012<br />

Vogt, Ulf 10627<br />

Vohra, Nasreen A. 10070<br />

Voi, Maurizio e13101<br />

Voillat, Laurent 8026<br />

Voit, Christiane A. 8535,<br />

e19022<br />

Vokes, Everett E. 5500,<br />

5517, 7513<br />

Volandes, Angelo E. 9004,<br />

9105<br />

Volchenboum, Samuel Louis<br />

9583, 9584, e20008<br />

Vollmer, Laura 8577<br />

Volm, Matthew e11529<br />

Voloschin, Alfredo Daniel<br />

3055<br />

Volovat, Constantin D. 9125<br />

Volpe, Silvia e21095<br />

Voltolini, Luca 7024<br />

Volzone, Francesco 8066,<br />

8083<br />

vom Dorp, Frank 4590<br />

von Abel, Ekkehard 10540<br />

von Blomberg, Mary E 2562<br />

von Boehmer, Lotta 2573^<br />

Von Briel, Thomas 3595<br />

Von Broen, Ingo TPS4146<br />

Von Euler, Mikael e13035<br />

Von Euw, Erika Maria 8592<br />

von Gruenhagen, Ulrich 3<br />

Von Gruenigen, Vivian E.<br />

TPS1614<br />

von H<strong>of</strong>e, Eric e15125<br />

Von H<strong>of</strong>f, Daniel D. 2516,<br />

3033, 4013<br />

von Kalle, Thekla 9573<br />

von Knebel Doeberitz, Magnus<br />

3550<br />

von Mehren, Margaret<br />

LBA10008, 10013, 10064,<br />

10088<br />

Von Minckwitz, Gunter 522,<br />

523, 541, 556, 1023<br />

Von Moos, Roger CRA3503,<br />

3595, e19648, 9059,<br />

10033, e13605, e18012,<br />

e19514<br />

Von Pawel, Joachim 7536<br />

Von Roenn, Jamie H. 610,<br />

TPS1134<br />

von Rohr, Lukas 9059<br />

Von Weikersthal, Ludwig<br />

Fischer 4023, 4539<br />

von Wichert, Goetz<br />

TPS3641^, 4032, 4034<br />

Vona, Karen L e19041<br />

Vonk, Judith M. 1502<br />

vonMehren, Margaret e19580<br />

VonMerveldt, Dendra<br />

TPS4678<br />

Voon, Pei Jye e13002,<br />

e19523<br />

Voon, Pei-Jye 1064<br />

Voorhees, Peter Michael 6526<br />

Vora, Lalit 1035<br />

Vora, Nilesh e14075<br />

Vora, Sujay A. 6108<br />

Vose, Julie 8047, 8060,<br />

8063<br />

Vosganian, Gregory Sumpao<br />

e21061<br />

Voss, Martin Henner 4532,<br />

4604<br />

Voss, Stephan D 9500<br />

Vouis, Jan e19532<br />

Voutsadakis, Ioannis e17007<br />

Vozzhaev, Alexander V. 9060<br />

Vrbanec, Damir TPS661,<br />

9046<br />

Vredenburgh, James J. 2027,<br />

TPS2103<br />

Vredenburgh, James J 2074^,<br />

2090^, 2094^, 2095^<br />

Vreeland, Timothy J 625,<br />

2508, 2529<br />

Vrelust, Inge 2506<br />

Vrignaud, Patricia e15161<br />

Vrooman, Lynda M. 9530<br />

Vuento, Maarit e15564<br />

Vugrin, Davor e12044<br />

Vukelja, Svetislava J. 547,<br />

1017<br />

Vukovic, Vojislav M.<br />

TPS7613^<br />

Vulsteke, Christ<strong>of</strong> 1020,<br />

3018^<br />

Vultaggio, Giuseppe e19595<br />

Vuong, Te e14020<br />

Vutukuru, Suresh TPS6640<br />

Vuylsteke, Peter 5001,<br />

e14060<br />

Vyas, Rakesh e15505<br />

Vyas, Shachi 2521<br />

Vyzula, Rostislav e11072,<br />

e18062<br />

Vågberg, Jan 6557<br />

W<br />

Waalen, Jill e21061<br />

Waanders, Esme 9550<br />

Wachsman, William 4030<br />

Wactawski-Wende, Jean 1501<br />

Waczulikova, Iveta e19020<br />

Wada, Noriaki 585, 1106,<br />

TPS1147<br />

Wada, Randal K. 6537<br />

Wada, Satoshi e13559<br />

Wada, Yasuhiro 10519<br />

Wada, Yumiko 3086, 3090<br />

Waddell, Tom Samuel<br />

LBA4000<br />

Wade, James Lloyd 9018,<br />

e15115<br />

Wade, Mark 3057, e13548,<br />

e14177<br />

Wadehra, Madhuri 1052<br />

Wadghiri, Youssef Zaim<br />

e19015<br />

800s Numerals refer to abstract number


Wadleigh, Martha 6526,<br />

6621<br />

Wadlow, Raymond Couric<br />

4021<br />

Wadsley, Jonathan LBA4000,<br />

6113<br />

Wadsworth, Trad 5570<br />

Wagenius, Gunnar e14531<br />

Wagenseller, Aubrey G 8530,<br />

8597<br />

Wages, David e18047<br />

Wagie, Terry 9019<br />

Wagle, Nikhil 5011, 10502<br />

Wagner, Andrea 3502<br />

Wagner, Andrew J. 1547,<br />

10029, e13600^<br />

Wagner, Jeffrey D. 8534<br />

Wagner, Lars M. 9541<br />

Wagner, Ludwig 2024<br />

Wagner, Lynne I. 605, 5566,<br />

6076, 8007, 9016, 9020,<br />

9106, 9532, e15173<br />

Wagner, Manfred e17536<br />

Wagner, Margaret 2516<br />

Wagner, Michael 10061<br />

Wagner, Steffen 556<br />

Wagner, Stephanie Glacy<br />

9571<br />

Wagner, Susanne 7023<br />

Wagner, Teresa M. U. 1537<br />

Wagner, Volker Jean<br />

TPS8604, e19043,<br />

e19044, e19045, e19060<br />

Wagner, Walter 4530<br />

Wagner, Wolfgang e13091<br />

Wagner-Johnston, Nina D.<br />

6518^<br />

Waguespack, Steven 4641<br />

Waheed, Sarah 8041,<br />

e18548, e18576<br />

Wahid, Fadilah Abdul e18528<br />

Wahl, Richard L. 10509,<br />

e13592<br />

Wai, Daniel 9564<br />

Waikins, Kate e13542<br />

Wainberg, Zev A. 3552<br />

Wainer, Zoe e17539<br />

Wainman, Nancy 1036<br />

Wakatsuki, Takeru 3518,<br />

3540, 3546, 3549, 3597,<br />

3604, 3615, 3622, 4026,<br />

4088, 4096, e11018,<br />

e14031, e14034, e14052<br />

Wakayama, Akihiko 5086<br />

Wakefield, Matthew 5073<br />

Wakelee, Heather A. 6050,<br />

7541, 10574<br />

Wakuda, Kazushige 7088<br />

Waldeck, Kelly 8520<br />

Waldman, Frederic 2587,<br />

5510, 8596, 10586<br />

Waldman, Scott A e14619<br />

Waldron, John N TPS5600,<br />

10522<br />

Waldschmidt, Dirk 4023<br />

Waldstrøm, Marianne 5052<br />

Walecki, Jerzy TPS4148<br />

Walewski, Jan Andrzej<br />

e18512<br />

Walker, Andrew e21107<br />

Walker, Dana Marie 1504<br />

Walker, Gregg 1608<br />

Walker, Luke N. 3024<br />

Walker, Mark S. 5014,<br />

e15061, e21077<br />

Walker, Michael 10576,<br />

10584<br />

Walker, Paul R. e11550,<br />

e17008, e17547, e17550<br />

Numerals refer to abstract number<br />

Walker, Steven M. 10553<br />

Walker-Corkery, Elizabeth<br />

9004<br />

Walkiewicz, Marzena e17539<br />

Walko, Christine Marie<br />

e13074, e13109<br />

Wall, Gregory e21159<br />

Wall, Louise e13026<br />

Wallace, Andrew e18095<br />

Wallace, Audrey S e17525<br />

Wallace, Elaine Margaret<br />

e14592<br />

Wallace, James Alfred<br />

TPS3640, 4022<br />

Wallace, Michael J. 515,<br />

4116, e14664<br />

Wallace, Paul K 6565<br />

Wallace, Robert B 6034,<br />

e14005<br />

Wallace, Sarah G. 5503,<br />

5515<br />

Wallenstein, Gudrun e15037<br />

Waller, Anna 6119<br />

Waller, Edmund K. 2576,<br />

e21045<br />

Wallin, Bruce 7030<br />

Wallin, Johan 3005<br />

Wallis, John W. 503<br />

Wallis, Matthew 544<br />

Wallwiener, Diethelm 1067,<br />

5042, e11040^, e15577,<br />

e21003<br />

Wallwiener, Markus 1067,<br />

1090, e21003<br />

Walpole, Euan Thomas 3508,<br />

TPS4145, e19550<br />

Walsh, Bridget 10509<br />

Walsh, Cathy 9589<br />

Walsh, Elaine e12038<br />

Walsh, Garrett L 4086<br />

Walsh, Mark Doughty 5050<br />

Walsh, Naomi 632, 633, 637,<br />

e13520<br />

Walsh, Wendy 1036, 2051,<br />

5010, 7093<br />

Walsh, William Vincent 6569<br />

Walshe, Janice Maria e17561<br />

Walshe, Margaret B. e12031<br />

Walston, Steve e19040<br />

Walter, Clare e16538<br />

Walter, Kim 5575<br />

Walter, Roland B. 1527<br />

Walter, Steffen 2522, 3530<br />

Walter, Thomas 4071, 4109,<br />

e14524, e14592<br />

Walterhouse, David 9509<br />

Walters, Christy Lynn 5060<br />

Walters, Ian B. 3504, 3572,<br />

7584<br />

Walters, Kelly Kaiser 7067,<br />

e18117, e21069<br />

Walters, Matthew J. 2043,<br />

e13580<br />

Walther, Philip John 4670<br />

Walz, Jochen e15155<br />

Wampfler, Jason A. 1610<br />

Wan, Wen e21058<br />

Wandel, Simon 8511<br />

Wanders, Jantien 2552<br />

Wands, Jack 4098<br />

Wanebo, Harold J. 5576,<br />

e13561<br />

Wang, Alice M. 10586<br />

Wang, Andrew 4658, 6037<br />

Wang, Bailiang 10559<br />

Wang, Benjamin e18063^<br />

Wang, Biyun 10567, e11567,<br />

e11568<br />

Wang, Bushi 7557, 7558<br />

Wang, Cathy 10622, e18549<br />

Wang, Changli 7520<br />

Wang, Chen-Chi e16006<br />

Wang, Cheng-Ping e16050<br />

Wang, Chenguang e13545<br />

Wang, Ching-Ping e16006<br />

Wang, Christopher Gene<br />

7072^, e17531^<br />

Wang, Chun-Wei e16050<br />

Wang, Chunting e13538<br />

Wang, Cong 4092, e14575<br />

Wang, Dan 5091<br />

Wang, David X. 5503<br />

Wang, Ding 2013, 2018,<br />

3078<br />

Wang, Dong 7559<br />

Wang, Emily 10545<br />

Wang, Ena 8576, 10565<br />

Wang, Eric 3590<br />

Wang, Erjian 2595, e13606<br />

Wang, Eunice S. 6526, 6565,<br />

6602<br />

Wang, Fan e13584<br />

Wang, Fang-zheng e13558<br />

Wang, Fen 2013<br />

Wang, Fuli e14643<br />

Wang, Haibin 7036<br />

Wang, Hao 4055, 5506,<br />

e14509, e16061, e21034<br />

Wang, Hong Qiang 2087<br />

Wang, Hong e21161<br />

Wang, Hongkun 2590, 3095<br />

Wang, Hongshan 4092,<br />

e14575<br />

Wang, Hongwei 5543,<br />

e15193<br />

Wang, Hsiu-Po e14576<br />

Wang, Huamin 4130, 4131<br />

Wang, Huei 4642<br />

Wang, Hui 4008<br />

Wang, Hung-Ming e16050<br />

Wang, J-J e14699<br />

Wang, James 3039, e13602,<br />

e13604<br />

Wang, Jennifer e19014<br />

Wang, Jia Hua TPS4149<br />

Wang, Jian hua 4073<br />

Wang, Jian-guo e14505<br />

Wang, Jian-qiu e13558<br />

Wang, Jian-Wei e14024<br />

Wang, Jiang e14714, e16017<br />

Wang, Jianhua e14605,<br />

e14645<br />

Wang, Jianmin 9518<br />

Wang, Jiayu e11025, e11538<br />

Wang, Jie 7520, 7537, 7545,<br />

e12503, e18022, e18035<br />

Wang, Jin e14648, e18033<br />

Wang, Jin-Wan e14502,<br />

e14668<br />

Wang, Jing 7096, 10612<br />

Wang, Jingyuan 4656<br />

Wang, Jinhua 9507<br />

Wang, Jinwan LBA4537<br />

Wang, Jue e15015<br />

Wang, Jun e17504, e21103<br />

Wang, Kai 10524, e17541<br />

Wang, Lan-Ying e16024<br />

Wang, Le e15522<br />

Wang, Lei Ping e11568<br />

Wang, Lidong 4591<br />

Wang, Lihua 3031<br />

Wang, Lin-Pei e14629<br />

Wang, Lina 4563<br />

Wang, Linda C 8552<br />

Wang, Ling Zhi 4100^<br />

Wang, Ling 6625^<br />

Wang, Ling-Zhi 2551,<br />

e13002<br />

Wang, Lisa 3082, 4092,<br />

5019, 8585, e14575<br />

Wang, Lisu 8593<br />

Wang, Liwei e14508, e14539<br />

Wang, Liya e14734<br />

Wang, Lu 7505, 7578,<br />

e14548<br />

Wang, Luhua e14511<br />

Wang, Lulu 2098<br />

Wang, Lv hua 4073<br />

Wang, Maoqiang e14605<br />

Wang, Meihua 2001, 2008b,<br />

2047<br />

Wang, Michael 8017, 8035,<br />

8093, e18557<br />

Wang, Min 6000<br />

Wang, Mingjun 4643<br />

Wang, Peng 3037<br />

Wang, Ping 1068, 7075,<br />

e18013, e18138<br />

Wang, Qian 9008<br />

Wang, Qiang 7536, e14617<br />

Wang, Qin e14583<br />

Wang, Qing 604<br />

Wang, Qiong J. e14179<br />

Wang, Qun e18013<br />

Wang, Ren-sheng 5535,<br />

e16054<br />

Wang, Rong 1595, 6628<br />

Wang, Rongfu F 4643<br />

Wang, Rui-Yu TPS6635<br />

Wang, S. L. 4644<br />

Wang, Sa 6501<br />

Wang, San Ming 1538<br />

Wang, Shuhuai e11095<br />

Wang, Si-Yu 7034<br />

Wang, Song e19043,<br />

e19044, e19045<br />

Wang, Ting Ting 1064<br />

Wang, Trent P e14124,<br />

e14125, e14727, e19639<br />

Wang, Wei e11521, e13530<br />

Wang, Wei-Lien 10071<br />

Wang, Weidong 5597<br />

Wang, Weigang TPS4675^<br />

Wang, Weili e17530<br />

Wang, Wen e14645<br />

Wang, Wenqing e14511<br />

Wang, Wenquan TPS1145<br />

Wang, Wenshi 2511<br />

Wang, Xiaoen e14700<br />

Wang, Xia<strong>of</strong>ei F. 7513<br />

Wang, Xicheng e14604<br />

Wang, Xin Shelley 6551,<br />

8091, 9072, 9081, 9082,<br />

9083, 9099, 9112, 9118,<br />

9140<br />

Wang, Xiuqiang 2587<br />

Wang, Xiuyan TPS2619,<br />

TPS4700<br />

Wang, Xu e21161<br />

Wang, Xuan 4008<br />

Wang, Xue e13564<br />

Wang, Xuefei 4092, e14575<br />

Wang, Xuemei 4054, 9072,<br />

e15075, e15092<br />

Wang, Xufang 4612, e14525,<br />

e14621, e14627<br />

Wang, Yan 3040<br />

Wang, Yanna 4092, e14575<br />

Wang, Yanxia e14645<br />

Wang, Yicheng 4643<br />

Wang, Ying 1047<br />

Wang, Yipeng 1058, e14164<br />

Wang, Yixin 10504<br />

Wang, Yongsheng e18051<br />

Wang, Yongyu TPS4147<br />

Wang, Yu e13580, e16039<br />

Wang, Yu-Chieh 10539<br />

801s


Wang, Yuan e15124<br />

Wang, Yunfei 547<br />

Wang, Yuyan 7537<br />

Wang, Zeng e17512<br />

Wang, Zhaohui e14118<br />

Wang, Zhen 620<br />

Wang, Zhensheng e14512<br />

Wang, Zhibo e14169,<br />

e14170<br />

Wang, Zhihong Joanne 9578<br />

Wang, Zhijie 7537, 7545,<br />

e18022, e18035<br />

Wang, Zhiqiang e13021<br />

Wang, Zhixiao e16521<br />

Wang, Zhong Hua 10567,<br />

e11568<br />

Wang, Zuoheng 5529, 5532<br />

Wang-Gillam, Andrea e14584<br />

Wani, Sachin e14584<br />

Wapnir, Irene e11032<br />

Ward, James Edward e16509<br />

Ward, John Francis e15194<br />

Ward, Kevin 5570, e14608<br />

Ward, Malcolm e21091<br />

Ward, Patrick J. 7567<br />

Ward, Tim H e15141<br />

Warde, Padraig Richard 4509,<br />

4535<br />

Wardelmann, Eva 10031<br />

Wardley, Andrew M. TPS660<br />

Wariabharaj, Darshan<br />

TPS6638<br />

Warlick, Christopher e15211<br />

Warloe, Trond e14637<br />

Warneke, Carla L 10607<br />

Warner, Andrew 7009<br />

Warner, Douglas 5072<br />

Warner, Ellen 1542, 1555,<br />

e12034, e19545<br />

Warner, Richard e21109<br />

Warnick, Ronald TPS3116<br />

Warr, Julia e15180<br />

Warren, Graham Walter 1529,<br />

e14659, e14712<br />

Warren, Robert S. 3517<br />

Warsch, Sean e16001<br />

Warsi, Ghulam 8012, 8028<br />

Wartenberg, Markus e19502<br />

Wasada, Izumi 8050<br />

Wasan, Harpreet S 3516,<br />

e21093<br />

Washam, Charity L. e21039<br />

Washington, Sierra 5107<br />

Washio, Marie e14004<br />

Wasielewska, Teresa TPS2619<br />

Wason, James 4531<br />

Wassberg, Cecilia 7539<br />

Wassermann, Johanna 601,<br />

e14553<br />

Wassmann, Karl 4516<br />

Watanabe, Hideki e13511<br />

Watanabe, Hiroyoshi 9574<br />

Watanabe, Jun e14047,<br />

e14050<br />

Watanabe, Kana 7565<br />

Watanabe, Kazuteru e14047,<br />

e14050<br />

Watanabe, Kenichiro 9577<br />

Watanabe, Koshiro 7012,<br />

e18099, e19556<br />

Watanabe, Masahiko 3071,<br />

4026, 4088, e14091<br />

Watanabe, Masayuki 10523,<br />

e13553<br />

Watanabe, Reiko e21007<br />

Watanabe, Tomoo e14004<br />

Watanabe, Toru 10554,<br />

e19557<br />

Watanabe, Toshiaki e14142<br />

Watanabe, Yasuyoshi 10519<br />

Waterhouse, Anna 511<br />

Waterhouse, David Michael<br />

7035, 7100, 7567<br />

Waterhouse, Jim e14006<br />

Waterkamp, Daniel 3614<br />

Waterman, Gabriel N. e18549<br />

Waters, Justin S. LBA4000<br />

Waterston, Ashita Marie<br />

TPS3637<br />

Watkin, Nicholas 4640,<br />

e15100<br />

Watkins, Brynmor A 9119<br />

Watkins, David J. 3531, 3587<br />

Watkins, Stanley P. 10509<br />

Watkinson, Anthony TPS4150<br />

Watson, Mark 503, 3047<br />

Watson, Melanie 8101<br />

Watson, William 633<br />

Watt, Christopher D. 4111<br />

Watt, Stuart e13107<br />

Wattel, Eric 6523<br />

Waxman, Ian TPS2614<br />

Waxman, Irving 10090<br />

Wayne, Jennifer L. 8081<br />

Wazaefi, Yanal 8578<br />

Wazer, David E. e11548<br />

Wear, Sandra Marija 8011<br />

Weaver, Amy 5004, 5076,<br />

8590<br />

Weaver, David T. 1057, 1091<br />

Weaver, JoEllen 577<br />

Weaver, Valerie e11096<br />

Webb, Christopher e13066<br />

Webber, Kate 9044<br />

Weber Hauge, Petra e14531<br />

Weber, Beatrice E. 1051,<br />

9079<br />

Weber, Béatrice LBA5002^<br />

Weber, Damien C. 2068<br />

Weber, Denis TPS6642^<br />

Weber, Dirk 2543<br />

Weber, Donna M. 8012, 8093<br />

Weber, Hans-Jochen e18572^<br />

Weber, Jeffrey S. 2511,<br />

3102, 8508, 8510, 8582,<br />

8587, e13088, e15010<br />

Weber, Jeffrey 8503^<br />

Weber, Jill M. 2612, 4125,<br />

e14579, e14715<br />

Weber, Karsten E. 542<br />

Weber, Michael J 8530<br />

Weber, Randal S. e16030<br />

Weberpals, Johanne Ingrid<br />

5019, 5032, e15537<br />

Webster, Edvelyn 10535<br />

Webster, Jennifer A e14160<br />

Webster, Kevin P. 5544<br />

Webster, Marc A. e21083<br />

Webster, Scott e21129<br />

Wechsler, Janine e21014,<br />

e21046<br />

Wedding, Ulrich 9114<br />

Wee, Sandra K 6074<br />

Weekes, Colin D. 3017,<br />

4050, 4052, e13006<br />

Weeks, Jane C. 599, 1125<br />

Weems, Garry Alan 4574<br />

Wege, Henning 4115<br />

Wegener, William A. 4043<br />

Wegner, Brett 8054<br />

Wei, Andrew 6527<br />

Wei, Jia 4068, e14509,<br />

e21034<br />

Wei, Jun 2011<br />

Wei, Li 3055<br />

Wei, Min e13531<br />

Wei, Ning e13004<br />

Wei, Qi 9029<br />

Wei, Sen e17514<br />

Wei, Weidong e11091<br />

Wei, Ye e14000<br />

Wei, Yuquan e13538<br />

Wei, Zhigang 7545<br />

Weibel, Kelcy C 8061<br />

Weichenthal, Michael 8505<br />

Weichselbaum, Ralph R.<br />

e11512<br />

Weickhardt, Andrew James<br />

3534, 7504, 7526<br />

Weide, Benjamin 8526<br />

Weideman, Prue C 1502<br />

Weidler, Jodi Marie 603, 608,<br />

614<br />

Weidmann, Eckhart 3<br />

Weigang-Koehler, Karin 2554<br />

Weigel, Brenda 9500, 9540,<br />

9541, 9581<br />

Weigel, Marion T. e13503<br />

Weikert, Steffen 4636,<br />

e15044<br />

Weil, Marcie K. 3031, 3087,<br />

3529<br />

Weil, Robert J. 2014, 2018,<br />

2076, 2100<br />

Weil, Susan 5062^<br />

Wein, Alan J. e15196<br />

Wein, Axel 4072<br />

Weinberg, Benjamin e14658<br />

Weinberg, Jeffrey 2019<br />

Weinberg, Lori Elizabeth<br />

5089<br />

Weinberg, Uri TPS2106,<br />

e14616<br />

Weinberg, Vivian K. 2564,<br />

3517, 4593, 4660,<br />

e15137<br />

Weiner, Bryan 6104<br />

Weiner, Louis M. 2590,<br />

TPS3638, e14569<br />

Weiner, Michael e16557<br />

Weingart, Saul e16515,<br />

e16559<br />

Weinhold, Kent 2585<br />

Weinreich, David M. 5072<br />

Weinreich, David Michael<br />

4606<br />

Weinrich, Scott 3509, 3511<br />

Weinschenk, Toni 2522,<br />

3530<br />

Weinstein, Howard J. 9502<br />

Weinstein, John N. 7096<br />

Weinstock, Chana 1039,<br />

10572<br />

Weintraub, Sheri 5596^<br />

Weir, Barbara 4585<br />

Weir, Genevieve 2588<br />

Weis, Joachim 6002, 7607<br />

Weis, John R. 3057, e14177<br />

Weiselberg, Lora Rose 1046<br />

Weiser, Martin R. 3583<br />

Weiskopf, Kipp 2514<br />

Weismann-Brenner, Alina<br />

e14067<br />

Weiss Smith, Sheila e16519,<br />

e16537<br />

Weiss, Alan 4006<br />

Weiss, Ge<strong>of</strong>frey R. 8530<br />

Weiss, Glen J. 2516, 2534,<br />

3019, 3033, 3046, 3060,<br />

3062, e19048^<br />

Weiss, Jared e16062<br />

Weiss, Lawrence M. e21019<br />

Weiss, Mark Adam 6122,<br />

6609<br />

Weiss, Matthias 6054,<br />

e16549<br />

Weiss, Robert H 4634<br />

Weiss, Rudolf 8005<br />

Weiss, Stephen 8099<br />

Weiss, Talia 9123<br />

Weissfeld, Joel L. e21012<br />

Weissler, Mark C 5539, 5577<br />

Weissman, Irving L 2523<br />

Weitbruch, Delphine e11542<br />

Weith, Ekaterina 4636,<br />

e14152<br />

Weitman, Steven D. e16566<br />

Weitzel, Jeffrey N. 1010,<br />

CRA1505<br />

Welch, Mary Roberta 2089<br />

Welch, Stephen 3013, 5019,<br />

6082, e19616<br />

Welcher, Rosanne e21129<br />

Weldon, Christine B. 1553,<br />

6120, e16531<br />

Welin, Staffan 4015<br />

Weller, Michael 2000, 2007,<br />

2055, 2067, TPS2105<br />

Wells, Karen 1590, 1591<br />

Wells, Nancy L. e16038,<br />

e19600<br />

Wells, Robert J. 9096<br />

Wells, Samuel A. e13122<br />

Wells, Susanne I e16017<br />

Wellstein, Anton e14009,<br />

e14569<br />

Welsh, James 4069, 4078,<br />

4086, 7062, 7096,<br />

e14547, e14669<br />

Welslau, Manfred LBA1, 640,<br />

3, e13100^<br />

Welz, Stephan e16033<br />

Wen, Hongxiu 1538<br />

Wen, Patrick Y. 2005, 2009,<br />

2012, 2025<br />

Wen, Sijin 4556, 6516^,<br />

e13506, e13595, e15181<br />

Wen, Wenhsiang e20507<br />

Wen, Yu-Ye 10598<br />

Wen, Yvonne TPS3112<br />

Wen, Zou Bing e14648<br />

Wendt, Thomas e16033<br />

Wendtner, Clemens Martin<br />

10031, 10054<br />

Weng, David Edward 3029<br />

Weng, Peggy 4010<br />

Weng, Wen-Kai 2514<br />

Weng, Xiaoduan e18136<br />

Wenham, Robert M TPS3114,<br />

5025, e13601<br />

Wenk, Kurt S e19025<br />

Wenners, Antonia Sophie<br />

1084, e13503<br />

Wenzel, Lari B. 6132<br />

Wenzel, Ralph 6136<br />

Wernecke, Klaus Dieter 5512<br />

Werner, Dominique e14557<br />

Werner, Jochen 5512<br />

Werner, Lillian 4577, 4580,<br />

4582, 4585, 6617, 6618<br />

Werner, Lynn 7092<br />

Werner, Theresa Louise<br />

e13548, e13596<br />

Werner-Wasik, Maria 2001,<br />

e19507<br />

Weroha, Saravut John 5077<br />

Wertz, Marie 2060<br />

Werutsky, Gustavo e11087<br />

Wesenberg, Finn 9590<br />

Wesley, Johnna D. 2563<br />

Wessalowski, Ruediger 10054<br />

Wesseling, Jelle 562, 10554<br />

Wessels, Judith A. M. 10518<br />

Wessels, Judith A.M. 526<br />

West, Dennis P. 2532, 4063,<br />

6623, e18571<br />

802s Numerals refer to abstract number


West, Howard Jack 7517,<br />

7552<br />

West, James K. 4059<br />

West, William W e17549<br />

Westbroek, David 1131<br />

Westeel, Virginie 7057,<br />

TPS7111, 7574, e16560,<br />

e18089<br />

Westermann, A.M. e13095^,<br />

e13097^<br />

Westermann, Jörg e15053<br />

Westin, Shannon Neville<br />

1513<br />

Westlin, William 8032<br />

Weston, Brian 4069, 4078,<br />

4086, e14547<br />

Westphal, Manfred 2033,<br />

2067<br />

Westra, William H. 5506,<br />

5566<br />

Wetzel, Antje e19540<br />

Wetzler, Meir 6513, 6520,<br />

6565, 6596, 6602, 6604<br />

Wex, Thomas 4107<br />

Wexler, Leonard H. 9503,<br />

9545<br />

Weyker, Paul e13066<br />

Weyman, Elizabeth A. e16059<br />

Weynand, Birgit 5519<br />

Whalen, John 6553<br />

Whalen, Michael e15019,<br />

e15021<br />

Whalen, Micheal e15018<br />

Wheater, Matthew 8566<br />

Wheatley-Price, Paul 6083,<br />

e19527<br />

Wheeler, Chris 2084<br />

Wheeler, Christopher Jay<br />

2087<br />

Wheeler, Helen e12507<br />

Wheeler, Stephanie B. 6017,<br />

6029, 6096, 6104<br />

Wheeler, Thomas M. 4643<br />

Wheelock, Alyse 9107<br />

Whelan, Jeremy 6115,<br />

10063, 10081<br />

Whelan, Timothy Joseph 500,<br />

LBA1031, 6035, 9020<br />

Wheler, Jennifer J. 3106,<br />

3107^, 8551, 10510,<br />

10607, e13020, e13506,<br />

e13534, e13595, e19004<br />

Whisnant, John 3060<br />

Whisnant, Mindy 6054<br />

White, Alison Marie e16004<br />

White, Carol B 6053<br />

White, Darrell 8020,<br />

TPS8112, TPS8114<br />

White, Emily 1527<br />

White, Jeff 4531<br />

White, Kevin P. 5517<br />

White, Nicole MA 4633<br />

White, Rebekah Ruth<br />

e14544, e14724<br />

White, Shane e17539<br />

White, Therese 2528<br />

White-Koning, Melanie 2549<br />

Whitehead, Robert P. 4019<br />

Whiteside, Theresa 8528<br />

Whiting, Samuel H. e14501<br />

Whitlow, Ann B. 5050<br />

Whitman, Eric D. e19057<br />

Whitmarsh, Karen A e18068<br />

Whitmore, James Boyd 4648,<br />

4650, e15120<br />

Whittaker, Sadie 1062<br />

Whittaker, Sean 8076<br />

Whittemore, Alice S 1502<br />

Numerals refer to abstract number<br />

Whittington, Richard 3605,<br />

4030<br />

Whitworth, Jenny M. 5060<br />

Whorf, Robert C. 8556<br />

Wiater, Katarzyna 10538<br />

Wichmann, Gunnar e16016<br />

Wick, Jo e13551<br />

Wick, Michael Jude e13025<br />

Wick, Wolfgang 2000, 2034<br />

Wickart-Johansson, Gun<br />

e14531<br />

Wickerham, D. Lawrence<br />

TPS1615<br />

Wickham, Thomas J. TPS663<br />

Wickrema, Amittha 6535^,<br />

e13120^<br />

Widemann, Brigitte C. 6530<br />

Widhalm, Georg 2053, 2071<br />

Widmark, Anders LBA4512,<br />

4551<br />

Wiebe, Karsten 10080<br />

Wiebe, Lauren Allison e14613<br />

Wiechert, Andrew e15545<br />

Wiechno, Pawel J. LBA4512,<br />

4551<br />

Wieczorek, Agnieszka Anna<br />

e18552<br />

Wieczorek, Stefan 1523<br />

Wiedenmann, Bertram 4122,<br />

4128, e14564<br />

Wiederholt, Peggy A e16010<br />

Wiegand, Phillip e16053<br />

Wiegand, Volker 2573^<br />

Wiegel, Thomas e15199^,<br />

e16033<br />

Wieland, Scott 10036<br />

Wierda, William G. 6516^,<br />

6517, 6584, 6598, 6603<br />

Wiernekowski, Jennifer 6035<br />

Wiernik, Peter H. 6595<br />

Wierzbowska, Agnieszka<br />

6559, 6632<br />

Wiese, David e14045,<br />

e14046, e15554<br />

Wiesner, Jason B e21026<br />

Wiesneth, Markus 6567<br />

Wiestner, Adrian 6581<br />

Wiezorek, Jeffrey S. 3581<br />

Wigginton, Jon CRA2509,<br />

2510, 2521, TPS2613,<br />

4505, 7509, 8507<br />

Wigler, Devin e19586<br />

Wihl, Jessica 5052<br />

Wiita, Elisee e13518<br />

Wikstrom, Katarina 10553<br />

Wiktor, Anne E. 4009<br />

Wilbaux, Melanie e15182<br />

Wilcken, Nicholas 550, 9087<br />

Wilczynski, Sharon 1035<br />

Wild, Aaron Tyler 4045, 4055<br />

Wildes, Tanya Marya e18551<br />

Wildiers, Hans 529, 1020,<br />

9035<br />

Wilding, George 4, 3101,<br />

3103, e13601<br />

Wilding, Greg 3582<br />

Wilding, Gregory E. 4654,<br />

e15204<br />

Wiley, Tim e19577<br />

Wilfong, Lalan S. e14501,<br />

e16536<br />

Wilgenh<strong>of</strong>, S<strong>of</strong>ie 2507,<br />

e19026, e19027<br />

Wilhelm, Christian 8004<br />

Wilhelm, Francois 3017,<br />

3081<br />

Wilhelm, Sibylla 8022<br />

Wilhelm, Stefan 6566<br />

Wilke, Hansjochen TPS4139<br />

Wilke, Lee G 1107<br />

Wilkerson, Julia 2548,<br />

e13122, e14064<br />

Wilkerson, Matthew D 7006<br />

Wilkinson, Kathy e16561<br />

Wilkinson, Neal W. e14659<br />

Wilkinson, Rosemary 1545<br />

Wilky, Breelyn A. 10025<br />

Willemen, Yannick e13051<br />

Willems, Luc e18006<br />

Willerding, Gregor 2054<br />

Willett, Christopher e14724<br />

Willey, Jie S. 594<br />

William, Basem M. 8047<br />

William, William 7047<br />

Williams, Andrew E e14160<br />

Williams, Cathy 8018^<br />

Williams, Charles C. 2559,<br />

7065, e17559<br />

Williams, Christopher Kwesi<br />

Oladipupo e17013<br />

Williams, Daphne 8541<br />

Williams, Denise E 2543<br />

Williams, Farah J 2608<br />

Williams, Gretchen M 9521,<br />

9522<br />

Williams, Jacqueline P 9027<br />

Williams, James Andrew<br />

2595, e13606<br />

Williams, Janet L. e19589<br />

Williams, Jennifer 1055<br />

Williams, Julie C e14505<br />

Williams, Lisa Simone 596,<br />

604<br />

Williams, Loretta A. 6551,<br />

8091, 9081, 9082, 9083,<br />

9140, e19589<br />

Williams, Mark e19062<br />

Williams, Melanie A. 6070<br />

Williams, Michael E. 8007<br />

Williams, Michael V. 4531<br />

Williams, Noelle S. 4616<br />

Williams, P. Mickey 3529,<br />

5020<br />

Williams, Paul e14164<br />

Williams, Richard Fred e17550<br />

Williams, Richard Thomas<br />

3581<br />

Williams, Sarah Jane 5046<br />

Williams, Scott G. TPS4690<br />

Williams, Stephanie A. e11084<br />

Williams, Steve e21012,<br />

e21142<br />

Williams-Elson, Irene e15010<br />

Williamson, Dan 9510<br />

Williamson, Elizabeth A.<br />

TPS5602<br />

Williamson, Stephen K.<br />

e13535, e13551<br />

Williamson, Todd E. e14106<br />

Williford, Susan Kidwell 9108<br />

Willinek, Winfried e14565<br />

Willis, Carl Rogaston e11562,<br />

e13003<br />

Willis, Nick e13522<br />

Willis, Scooter 1027, e21099<br />

Willman, Cheryl L. 6502,<br />

9506<br />

Willson, Anna 2015<br />

Wilmink, Johanna 4094<br />

Wilner, Keith D. 7508, 7596,<br />

7598, 7599, 7600<br />

Wilsdon, John B. e13522<br />

Wilson, Alex e15011<br />

Wilson, Andrew J e15582<br />

Wilson, Bethany Cox e21000<br />

Wilson, Carmen Louise 9528<br />

Wilson, Charles e14100<br />

Wilson, Dale e15554<br />

Wilson, David O 7071, 7074<br />

Wilson, David e14061<br />

Wilson, Don 3610<br />

Wilson, J. Frank 6052<br />

Wilson, James Matthew e17519<br />

Wilson, John 599<br />

Wilson, Kate 3534, TPS4145,<br />

e13007<br />

Wilson, Keith e16017<br />

Wilson, Leslie e15154,<br />

e19571<br />

Wilson, Manda e19647<br />

Wilson, Matthew W. e13544<br />

Wilson, Nicole Maire e18553<br />

Wilson, Peter M.D. 3584,<br />

3622, e14034<br />

Wilson, Peter 4529<br />

Wilson, Richard H. 3516<br />

Wilson, Richard K. 503, 9518<br />

Wilson, Tim 5575<br />

Wilson, Timothy G. e15084,<br />

e15178<br />

Wilson, William A. e16035<br />

Wilson, Wyndham Hopkins<br />

2503^, 2528, 8005, 8051<br />

Wilt, Marc e11542<br />

Wiltshire, Kirsty TPS4690<br />

Wiltshire, Robin 4542<br />

Wimberger, Pauline LBA5002^,<br />

5007, 5031, 5042, 5044,<br />

e13095^, e13097^,<br />

e13100^, e13108^<br />

Winch, Chad TPS2104<br />

Winchester, David J. 1033,<br />

1101, 1116, 6040<br />

Winchester, David Porter<br />

1033, 1101, 1116, 6040<br />

Windemuth-Kiesselbach,<br />

Christiane 8052<br />

Winder, Thomas 3584,<br />

e14031<br />

Winderlich, Mark 6574<br />

Winer, Eric P. 510, 535,<br />

CRA1002, 1026, 6015,<br />

9084, e21010<br />

Winer, Eric S. 9117<br />

Winer, Eric TPS658, 1009,<br />

6089, 9071, 9100, 10558,<br />

e16515<br />

Winfree, Katherine B. 7075,<br />

e18138<br />

Wingate, Patti 4098<br />

Wingmo, Inga-lill 4561<br />

Winick, Naomi Joan<br />

CRA9508, 9543, 9548,<br />

e19536<br />

Winkelman, J. e13588<br />

Winkelmann, Cornelia 4023<br />

Winkler, Annette 9059<br />

Winkler, Peter 9573<br />

Winquist, Eric 4535, 5504^,<br />

TPS5600, e15177^,<br />

e19616<br />

Winrich, Barbara 4021<br />

Winslow, John William 603,<br />

608<br />

Winter, Christian 4601<br />

Winter, Eva 4530<br />

Winter, Kathryn A. 4041<br />

Winterhalder, Ralph C. 3595,<br />

9059<br />

Winterle, Natalie e14554<br />

Winton, Elliott F. 6624<br />

Winzer, Harald e15566<br />

Wirapati, Pratyaksha 3509,<br />

9510<br />

Wirth, Lori J. 5501, 5508,<br />

5579, 5594, e16059<br />

Wirth, Manfred e15195<br />

803s


Wischnewsky, Manfred 1078<br />

Wischnik, Arthur 1072, 1081<br />

Wise-Draper, Trisha e16017<br />

Wisinski, Kari Braun 555,<br />

563, 6063<br />

Wislez, Marie TPS7111,<br />

e18075, e18089, e18102<br />

Wisnivesky, Juan P. 4623,<br />

6065, 7064, e15513<br />

Wissel, Paul Stephen 4111<br />

Wistuba, Ignacio I. 7023,<br />

7078, 7597, 10506<br />

Witek, Matthew e14619<br />

Witkiewicz, Agnieszka e19016<br />

Witkowski, Elan R. 4059,<br />

e14691<br />

Witowski, Steven 8032<br />

Witt, Lisa A e21000<br />

Witta, Samir E. e13101,<br />

e18077<br />

Witte, Maria 8526<br />

Witteles, Ronald 4610<br />

Witten, Matthew R. e21114<br />

Witter, Merle 8527<br />

Witteveen, Petronella<br />

LBA5002^<br />

Witthuhn, Ralf 4601<br />

Wittig, Burghardt 4636,<br />

e14152<br />

Wittlinger, Michael 5512<br />

Witzig, Thomas E. 8043,<br />

8053, TPS8118<br />

Wloch, Mary K e19058<br />

Wo, Jennifer Yon-Li 4021,<br />

e14615<br />

Woda, Bruce 6569<br />

Woditschka, Stephan 505<br />

Woehrer, Adelheid 2024,<br />

2053<br />

Woelber, Linn Lena 5007,<br />

5034, 5058<br />

Woell, Ewald 4074, 4095^<br />

Woerns, Marcus A 4115<br />

Wogu, Adane 6129, 9135<br />

Wohnrath, Durval R. e14599<br />

Woike, Michael 4631<br />

Wojcicki, Anne 10097<br />

Wojciechowski, Tomasz<br />

e15500<br />

Wojtowicz-Praga, Slawomir<br />

7573<br />

Wojtukiewicz, Marek TPS649<br />

Wolanski, Andrew 3053<br />

Wolanskyj, Alexandra P. 6614<br />

Wolchok, Jedd D. 2518,<br />

2521, 8508, 8512, 8515,<br />

8539, 8549, 8575, 8583,<br />

8598, TPS8603<br />

Wold, Gwyn e13028<br />

Wolden, Suzanne L. 5537,<br />

CRA9513<br />

Wolf, Gregory T. e16014<br />

Wolf, Hans-Heinrich e19579<br />

Wolf, Holly J 10580<br />

Wolf, Judith 5027<br />

Wolf, Juergen 1533, 3044,<br />

TPS3115, 7603,<br />

CRA10529<br />

Wolf, Jurgen 10526<br />

Wolf, Martin 2504<br />

Wolf, Max 8002<br />

Wolfe, Joanne 9076<br />

Wolfer, Anita e11048<br />

Wolff, Antonio C. 1128, 6026<br />

Wolff, Greg G. e12024,<br />

e15197, e18107, e19630<br />

Wolff, Hendrik A. 3600<br />

Wolff, Robert A. 4038, 4051,<br />

4054, 4060<br />

Wolff, Steven N. 6038<br />

Wolfgang, Christopher Lee<br />

3596, 4045, 4055<br />

Wolfgarten, Matthias Kim<br />

1077<br />

Wolfman, Judith 518<br />

Wolfson, Julie Anna 9512<br />

Wolin, Edward M. 4014,<br />

e14610<br />

Wolkeinstein, Pierre 10049<br />

Woll, Penella J. 7562<br />

Woll, Penella 6113<br />

Wollins, Dana CRA1505<br />

Wollner, Mira 9052<br />

Wollschlaeger, Kerstin 5005<br />

Wollschleger, Dennis J<br />

e19615<br />

Wolmark, Norman LBA506,<br />

538, 611, TPS657,<br />

LBA1000, 1025, TPS1142,<br />

TPS1615, 3512, 3545<br />

Wolpin, Brian M. e19586<br />

Wolpin, Seth 9008<br />

Wolter, Pascal 4622<br />

Wolters, Regine 1078<br />

Womack, Chris 4124<br />

Womer, Richard 9503, 9537<br />

Won, Hye Sung e14634<br />

Won, Jong-Ho e14688,<br />

e19554<br />

Won, Minhee 2047<br />

Wong, Andrea LA 1064,<br />

2551, 3002<br />

Wong, Bob 9137<br />

Wong, Chung-Kwun Amy<br />

6005<br />

Wong, Eric e18541, e21078<br />

Wong, F. Lennie 9505<br />

Wong, Hai Ming e12006<br />

Wong, Helen 8564, e15128,<br />

e15135, e18068<br />

Wong, Hilda e14545<br />

Wong, Hing C. e13088,<br />

e15010<br />

Wong, Jan H. e11550<br />

Wong, Janice C. 6087<br />

Wong, Karina 584, TPS10631<br />

Wong, Kit Man 10522<br />

Wong, Kwong-Kwok e15583<br />

Wong, Lilly 3006^<br />

Wong, Linda 582<br />

Wong, Lucas 3078<br />

Wong, Michael K.K. 3042^,<br />

4608, 4612<br />

Wong, Nicole TPS3643,<br />

TPS4137<br />

Wong, Rachel e14088<br />

Wong, Rebecca TPS4141,<br />

9131, 10522<br />

Wong, Richard J. 5545^<br />

Wong, Sandra L. 6020<br />

Wong, SC Cesar e14085<br />

Wong, Shirley S. 5038<br />

Wong, Siu-Fun 9018, e16523<br />

Wong, Stephen 2098<br />

Wong, Stuart J. 5583,<br />

e16053<br />

Wong, Terence e13116<br />

Wong, William 6108<br />

Wong, Winston 6098, e16540<br />

Wong, Yu-Ning 1544, 4522,<br />

4525, 4526, 6021, 6061,<br />

e14021, e19580<br />

Woo, Henry TPS4690<br />

Woo, Hyun Young e14566<br />

Woo, Jacky e13500, e13562,<br />

e13575, e13577, e21028,<br />

e21058<br />

Wood, Bradford J 2545,<br />

TPS4701<br />

Wood, Charlotte 9543<br />

Wood, Christopher G. 4500,<br />

9029, e15075, e15092<br />

Wood, Karen TPS4682<br />

Wood, Laura S. 4609,<br />

TPS4684, e15095<br />

Wood, Lauren V. 10589<br />

Wood, Leslie 3073<br />

Wood, Lori 4536, 4538<br />

Wood, Marie CRA1505<br />

Wood, Matthew D. e14514<br />

Wood, Patricia Ann 2543<br />

Wood, William C. 1005<br />

Woodman, Ciaran e13507<br />

Woodman, Richard Charles<br />

6509^<br />

Woodman, Richard John<br />

e19550<br />

Woodman, Scott Eric e19009<br />

Woodman, Scott e19039<br />

Woodruff, Teresa K. e20001<br />

Woods, Ryan 582, 3527,<br />

6014<br />

Woodward, Wendy A. 594,<br />

1047, 1102, e13582<br />

Woodward, William J. 10011^<br />

Woopen, Hannah e15531<br />

Wooten, Kathleen 10572<br />

Wooten, Michael e14030<br />

Worden, Francis P. e16014<br />

Worden, Rebekah 3083<br />

Worman, Melissa 1536<br />

Wormann, Bernhard J. 6610<br />

Wormanns, Dag e13547<br />

Worni, Mathias e14544,<br />

e14562<br />

Worth, Laura L 6540<br />

Wotherspoon, Andrew<br />

LBA4000, e14088<br />

Woulfe, Bernie e16551<br />

Woyach, Jennifer Ann 6508<br />

Wozniak, Agnieszka 10030<br />

Wozniak, Antoinette J. 2081,<br />

7018, 7029, 7083, 7517,<br />

7552, 7593, e17528<br />

Wozniak, Timothy F. 3545,<br />

e18122<br />

Wrangle, John e18147<br />

Wreschner, Daniel H. e13018<br />

Wright, Alan e21152<br />

Wright, Alexi A. 5011,<br />

e16500<br />

Wright, Brian 4013, 10630,<br />

e15209<br />

Wright, Frances e19545<br />

Wright, Gavin e16501,<br />

e17539<br />

Wright, Jason D. 5013, 5101,<br />

6078<br />

Wright, Jason Dennis 6118<br />

Wright, Jean L. 595<br />

Wright, Jim R. TPS5600<br />

Wright, John Joseph 2553,<br />

TPS3112, 4569<br />

Wright, Jonathan 511<br />

Wrin, Joe 3534<br />

Wroblewski, David 8553<br />

Wroblewski, Kristen 9586<br />

Wroblewski, Susan TPS661<br />

Wroclawska-Swacha, Monika<br />

M e18572^<br />

Wronski, Samantha 6553<br />

Wu, Abraham Jing-Ching<br />

4061<br />

Wu, Benjamin 529, 5038<br />

Wu, Bor-Wen e17558<br />

Wu, Cassandra TPS8118<br />

Wu, Changping 9017<br />

Wu, Chengyu e14700<br />

Wu, Christina Sing-Ying 609,<br />

e14178, e14658<br />

Wu, Colleen 10629<br />

Wu, Eijean e19506<br />

Wu, Eric Q. 1037, 6594,<br />

6600, 10094, e11076,<br />

e16571, e20511<br />

Wu, Fang e16054<br />

Wu, Fei 3551<br />

Wu, Frank S. LBA8509<br />

Wu, Han-chieh e11068<br />

Wu, Hillary H. TPS3111<br />

Wu, Hong-Gyun 5552<br />

Wu, Hui 626, e13579<br />

Wu, Jianrong 9502, e13584<br />

Wu, Jie e14701<br />

Wu, Jing 2029^, 2091,<br />

e12010<br />

Wu, Jingyang 2534<br />

Wu, Jonn 7020<br />

Wu, Kehua 2611, e13106<br />

Wu, Lin 10596, e13600^<br />

Wu, Meina 7537, 7545,<br />

e18035<br />

Wu, Minghui e13564<br />

Wu, Nian 10004<br />

Wu, Pei-Fang 1079<br />

Wu, Ping 8015<br />

Wu, Qi e18033<br />

Wu, Qian 5530<br />

Wu, Quinfei e19647<br />

Wu, Shao-xiong e18121<br />

Wu, Shenhong 9088, 9092,<br />

e13591, e15140, e18571,<br />

e19624<br />

Wu, Shi Xiu e18013<br />

Wu, Terry e17524<br />

Wu, Timothy 3105<br />

Wu, Tsung-Teh 4009<br />

Wu, Wei e14701<br />

Wu, Weiguo e13562,<br />

e13575, e21058<br />

Wu, Wen Shuo e18132<br />

Wu, Wenting 2004, 2031<br />

Wu, Xiao-cheng 6052<br />

Wu, Xiaoyuan e14645<br />

Wu, Xifeng 7608, e14153<br />

Wu, Xiwei 10545<br />

Wu, Yi Long 7039, 7519^,<br />

7520, 7533, 7559,<br />

e13510, e18035, e18090<br />

Wu, Yuehan TPS5602<br />

Wu, Yun 1054, 1130, 10559<br />

Wu, Zheng e14569<br />

Wucherpfennig, Kai 2502<br />

Wuchter, Patrick 10040<br />

Wuendisch, Thomas 4600<br />

Wuerflein, Dieter e17536<br />

Wujcik, Debra 9137<br />

Wulf, Gerald 6543, 8004<br />

Wunderlich, John R e14179<br />

Wustner, Jason 3016<br />

Wuthrick, Evan John 5530,<br />

e19507<br />

Wyatt, Gwen TPS9150<br />

Wyen, Christoph 8005, 8046,<br />

8058<br />

Wyld, David 3572, 9087<br />

Wyld, Peter J. 9542<br />

Wymenga, Machteld 1080<br />

Wynendaele, Wim 4622<br />

Wynes, Murry W. 7552, 7597<br />

Wyngaard, Peter e14164<br />

Wypij, Jackie M. e21145<br />

Wyrwicz, Lucjan 10561<br />

Wyss, Annah 6119<br />

Wöckel, Achim 1078, 1082<br />

804s Numerals refer to abstract number


X<br />

Xanthakis, Ioannis 5033<br />

Xanthopoulos, Eric 7050,<br />

7098, e17534, e18032<br />

Xavier, Patricia e11087<br />

Xenidis, Nikolaos e14567<br />

Xi, Liqiang TPS7609<br />

Xia, Chang 3056<br />

Xia, Lin e14539<br />

Xia, Menghang 10025<br />

Xian, Shuang e13538<br />

Xiao, Han 5514<br />

Xiao, Hugh 2597<br />

Xiao, Lianchun 4069, 4116,<br />

4615, e14547, e14669<br />

Xiao, Lin e15117<br />

Xiao, Shaowen 10045<br />

Xiaoyan, Chen e13036<br />

Xiaoyu, Shawn 5503, 5515<br />

Xiazhen, Yu e14153<br />

Xie, Conghua 5535<br />

Xie, Hao 2092<br />

Xie, Hong 8018^<br />

Xie, Jiansong 3580<br />

Xie, Li e14509, e21034<br />

Xie, Sherlly 4559<br />

Xie, Tao 3509<br />

Xie, Wanling 4521, 4538<br />

Xie, Xian-Jin 4616, e14165,<br />

e15066<br />

Xie, Xiaoming e11091<br />

Xie, Yang e14153<br />

Xie, Zhi e13510<br />

Xin, Song 8554<br />

Xin, Yan 2567<br />

Xing, Heming e19055<br />

Xing, Jun e13529<br />

Xing, Ligang e14643<br />

Xing, Pu-Yuan e14502<br />

Xiong, Jian ping 7548<br />

Xiong, Liwen 1551, e12010,<br />

e17551<br />

Xiong, Shigang e15153<br />

Xiong, Shunbin 10506<br />

Xiros, Nikolaos e21027<br />

Xiu, Joanne 5523<br />

Xiu, Qingyu 7520<br />

Xiu, Xia e14511<br />

Xu, Binghe TPS649, TPS1135,<br />

e11025, e11537, e11538<br />

Xu, Chun-fang e15059<br />

Xu, Cindy 559<br />

Xu, Fei e13021<br />

Xu, Feng 4638<br />

Xu, Jian-Ming 3040,<br />

LBA10008<br />

Xu, Jianmin e14000<br />

Xu, Jiasen e18142<br />

Xu, Jishu 1515<br />

Xu, Lian 8042, 8106, 8107<br />

Xu, Liang Guo e14071<br />

Xu, Lu 3025<br />

Xu, Min e11520, e20001<br />

Xu, Na 532<br />

Xu, Ning-Zhi e14502<br />

Xu, Nong 7559<br />

Xu, Peng 5597<br />

Xu, Ping e11061, e15177^<br />

Xu, Qing 9017<br />

Xu, Qinghua 3551<br />

Xu, Rui-hua e14653, e14655<br />

Xu, Sha<strong>of</strong>a e18013<br />

Xu, Shidong e18013<br />

Xu, Shuichan 3006^<br />

Xu, Wei Xu e13004<br />

Xu, Wei 1535, 6005, 7586,<br />

9021, 9032<br />

Xu, Xiao wei 8561, 8562<br />

Xu, Xunhai 10516<br />

Numerals refer to abstract number<br />

Xu, Yang e14538<br />

Xu, Yanqing 9557<br />

Xu, Yaping e14678, e17527<br />

Xu, Ye 3551<br />

Xu, Yihuan 5012<br />

Xu, Ying e14549, e14550<br />

Xu, Yingxin e14061<br />

Xu, Yong e14648, e18033<br />

Xu, Younong 2585<br />

Xu, Zijie S. 6093<br />

Xuan, Lixue e11538<br />

Xue, Cong 5538<br />

Xue, Hong-Ling TPS4149,<br />

e15047, e15064<br />

Xue, Liquan 3090<br />

Xue, Mei 553<br />

Xue, Xiaonan 8005<br />

Xue, Zhengyu e15059<br />

Xyrafas, Alexandros e18105<br />

Y<br />

Ya-Qin, Li e16017<br />

Yachnin, Jeffrey TPS4696^,<br />

e15115<br />

Yaegashi, Nobuo 5087<br />

Yagata, Hiroshi 590<br />

Yagmurdur, Mahmut Can<br />

e11023, e11026, e11515<br />

Yahalom, Joachim 2006<br />

Yakoub-Agha, Ibrahim 6534,<br />

6542<br />

Yakushev, Vadim A. 9060<br />

Yalcin, Bulent TPS4148<br />

Yalcin, Selim e11011,<br />

e11023, e11026, e11045,<br />

e11515, e14174, e15572,<br />

e15573<br />

Yalcin, Suayib 3565, e20514<br />

Yalçintas Arslan, Ülkü 638,<br />

e11540<br />

Yale, Steven e16549<br />

Yam, David e15180<br />

Yamada, Yasuhide e14123,<br />

e14126, e14510<br />

Yamagishi, Atsushi e15057<br />

Yamaguchi, Akito e15127<br />

Yamaguchi, Hironori e14530<br />

Yamaguchi, Hiroyuki 7569<br />

Yamaguchi, Ken D. 5074<br />

Yamaguchi, Kensei 4002,<br />

e14510<br />

Yamaguchi, Motoko 8050<br />

Yamaguchi, Nise Hitomi<br />

e16505<br />

Yamaguchi, Ou e19583<br />

Yamaguchi, Risa e11017<br />

Yamaguchi, Satoshi 5103<br />

Yamaguchi, Shigeki 3524,<br />

3607, e14091<br />

Yamaguchi, Takayoshi e14622<br />

Yamaguchi, Taketo 4031,<br />

4035<br />

Yamaguchi, Takuhiro 1530<br />

Yamaguchi, Tatsuro e14123<br />

Yamaguchi, Tomohiro 4046<br />

Yamaguchi, Toshiharu 3625,<br />

10541, e14145<br />

Yamaguchi, Yoko e17554,<br />

e18064<br />

Yamamichi, Noboru 1119<br />

Yamamoto, Chiriko 3625<br />

Yamamoto, Hiromasa e17517<br />

Yamamoto, Hiroyuki e14635<br />

Yamamoto, Junji 3625<br />

Yamamoto, Kuniharu e14011<br />

Yamamoto, Makoto e18135<br />

Yamamoto, Naohito 1119<br />

Yamamoto, Naoto TPS1147<br />

Yamamoto, Nobuyuki 6112,<br />

7003, 7017, 7028, 7088,<br />

LBA7500, 7551, 7602,<br />

e18093, e21007<br />

Yamamoto, Noriko 10541<br />

Yamamoto, Norio 10075,<br />

e20505<br />

Yamamoto, Satomi 7000,<br />

e18128<br />

Yamamoto, Seiichiro 3569,<br />

e14091<br />

Yamamoto, Shinya e15038<br />

Yamamoto, Suguru e18007,<br />

e18056, e18141<br />

Yamamoto, Tadashi 9053<br />

Yamamoto, Yutaka 588,<br />

e11017<br />

Yaman, Sebnem e11009,<br />

e11033, e11093, e13031<br />

Yamana, Hideaki e14077<br />

Yamanaka, Kentaro e21059<br />

Yamanaka, Takeharu TPS659,<br />

e14038<br />

Yamane, Yuki 1552, e18064<br />

Yamao, Kenji 4031, 4035,<br />

e14540<br />

Yamaoka, Toshimitsu e13029<br />

Yamashita, Hideomi e15533<br />

Yamashita, Hiroharu e14530<br />

Yamashita, Keishi 3071,<br />

3084, 3088^<br />

Yamashita, Koji 1112<br />

Yamashita, Motohiro 7012<br />

Yamashita, Yoshiki e19546<br />

Yamashita-Kashima, Yoriko<br />

e13502<br />

Yamauchi, Hideko 590,<br />

10048, e13037<br />

Yamauchi, Yoshikane 7038<br />

Yamazaki, Hiroko e13076<br />

Yamazaki, Kentaro e14038,<br />

e14123<br />

Yamoah, Kosj e15163<br />

Yan, Feng-qin e13558<br />

Yan, Haolin e16054<br />

Yan, Houling e19606,<br />

e19639<br />

Yan, Jingsheng e15066<br />

Yan, Kun e14701<br />

Yan, Li e13599<br />

Yan, Max 504<br />

Yan, Xiaowei (Sherry) e15536<br />

Yan, Yibing TPS2616<br />

Yanagi, Hidenori e14541<br />

Yanagisawa, Satoru e19508<br />

Yanagitani, Noriko 10569,<br />

10604, e18004, e18014,<br />

e18017<br />

Yancey, Mary Ann 8088,<br />

8104, e18568<br />

Yang, Allen S. 8073<br />

Yang, Anthony D e19001,<br />

e19013<br />

Yang, Arvin 8508<br />

Yang, Connie L 9591<br />

Yang, Dan e13519<br />

Yang, David T e13537<br />

Yang, Di 6056<br />

Yang, DongQing e21116<br />

Yang, Dongyun 3518, 3540,<br />

3546, 3549, 3584, 3597,<br />

3604, 3615, 3622, 4026,<br />

4088, 4096, 4563, e11018,<br />

e14031, e14034, e14052<br />

Yang, Eddy Shih-Hsin 621<br />

Yang, Fan e17504, e21103<br />

Yang, Feng e14651<br />

Yang, Guang 8042, 8106,<br />

8107, e14153<br />

Yang, Heidi 6600<br />

Yang, Hengxuan 4106<br />

Yang, Hongbo 4612<br />

Yang, Huibin 4591<br />

Yang, Huiyi e18142<br />

Yang, Hwai-I e17014<br />

Yang, James Chih-Hsin<br />

LBA7500, 7557, 7558<br />

Yang, Jennifer e13549<br />

Yang, Jianliang e14606<br />

Yang, Jianyong e14605<br />

Yang, Jijin e14605<br />

Yang, Jinji 7039, e18090<br />

Yang, Jui-Chen 4608<br />

Yang, Li 3040, 3551<br />

Yang, Lin e14668, e18098<br />

Yang, Ling TPS4146<br />

Yang, Lixin 1070<br />

Yang, Lu 7537, 7545,<br />

e18022, e18035<br />

Yang, Pan-Chyr 1534, 7533,<br />

e13061<br />

Yang, Ping 1610<br />

Yang, Qun-ying e12503<br />

Yang, Renjie e14605<br />

Yang, Sheng e14606<br />

Yang, Sherry X. 10603<br />

Yang, Tong e17514<br />

Yang, Tsai-Shen 4007<br />

Yang, Wei Tse 594<br />

Yang, Wei e14701<br />

Yang, Weizhu 4008<br />

Yang, Woo Ick 3606<br />

Yang, Xiaopin e14523<br />

Yang, Ximing J 605<br />

Yang, Xinjie e18142<br />

Yang, Xinqun 5038<br />

Yang, Xuening 7039<br />

Yang, Xuezhong e14658<br />

Yang, Yang 5097<br />

Yang, Yongping 4008<br />

Yang, Yu 3077<br />

Yang, Yue e18013<br />

Yankelevich, Maxim e13089<br />

Yann-Alexandre, Vano 601<br />

Yannopoulou, A 583<br />

Yano, Michihiro 9574, 9577<br />

Yano, Shuya 1063<br />

Yano, Tokujiro e13090<br />

Yanovich, Saul e19565<br />

Yao, Bin 7549, TPS8604<br />

Yao, James C. 4014, 4126,<br />

e14542, e14549, e14550,<br />

e14551, e14662<br />

Yao, Jianchao 4562<br />

Yao, Katharine 577, 1033,<br />

1101, 1116, 6040<br />

Yao, Lie e14651<br />

Yao, Masahiro 4626<br />

Yao, Xiaopan 6033, e14534,<br />

e21106<br />

Yao, Yang 9017<br />

Yao, Yanwen e18082<br />

Yap, Jeffrey T. 510<br />

Yap, Mei Ling 5105<br />

Yap, Timothy Anthony 2530,<br />

5022, 10525<br />

Yap, Yoon Sim 9041, e13023<br />

Yarde, Erin 3047<br />

Yardley, Denise Aysel 567,<br />

618, 1018, 1086, 1526<br />

Yarlagadda, Naveen e14659<br />

Yarlett, Daniel 6072<br />

Yarney, Joel e15163<br />

Yarnold, Paul R 2532<br />

Yaron, Yifah 5508<br />

Yasenchak, Christopher A.<br />

535, 3069, 7514<br />

Yashiro, Hideki 7038<br />

805s


Yasuda, Hiromi e14029<br />

Yasuda, Hiroyuki 7523<br />

Yasuda, Makoto 5003<br />

Yasuda, Yosuke e15038<br />

Yasui, Hir<strong>of</strong>umi 4002, 4082,<br />

e14510<br />

Yasui, Yutaka 6030<br />

Yasunaga, Yutaka e15065,<br />

e15069<br />

Yatabe, Yasushi 7521,<br />

e18145<br />

Yau, Christina 10586<br />

Yau, Cindy Y. F. 3013<br />

Yau, Stephen 5511, 7549<br />

Yau, Thomas Cheung 4108,<br />

e14545<br />

Yau, Tsz Kok 5511<br />

Yavuz, Gulsan e20000<br />

Yaya, Ricardo 3618, 10547<br />

Yazbek, Gabriel e19640<br />

Ychou, Marc 3502, 3506,<br />

3633, 4087, 4091, 10505<br />

Ye, Chen Yang 3040<br />

Ye, Dingwei LBA4537, 4628<br />

Ye, Fei 8544, e21014,<br />

e21046<br />

Ye, Josephine 3086<br />

Ye, Ming 4073, 7520<br />

Ye, Sheng e18517<br />

Ye, Sheng-Long 4008<br />

Ye, Wei-Wu e17512<br />

Ye, Xiaobu e17552<br />

Ye, Xun 3551<br />

Ye, Yang e13595<br />

Ye, Zhishen 4510<br />

Yeap, Beow Yong 4021,<br />

7579, 9585, 10058,<br />

10592<br />

Yeap, Shang Heng 550<br />

Yeazel, Mark W. 6030<br />

Yee, Cecilia e15147<br />

Yee, Douglas e13590<br />

Yee, John 7020<br />

Yee, Lorrin 3059<br />

Yeend, Susan e19513<br />

Yeganegi, Homa 8011<br />

Yeh, Benjamin M. 4102<br />

Yeh, Dah-Cherng 1079<br />

Yeh, Jen Jen e14505<br />

Yeh, Kun-Huei 4081<br />

Yeh, Su-Peng e17014<br />

Yelamanchili, Radhika e14001<br />

Yelensky, Roman 1015,<br />

3533, 4649, 7510, 7529,<br />

10524, 10559, 10590,<br />

e17541, e19004<br />

Yellapragada, Sarvari e17018<br />

Yellin, Ori 8098, e18558<br />

Yen, Chia-Jui 4103, 4108,<br />

e14617<br />

Yen, Katharine 6606<br />

Yen, Yun 1070, 10545,<br />

e15178<br />

Yendamuri, Saikrishna S.<br />

e14712, e17535<br />

Yenidunya, Gulsah e21143<br />

Yennurajalingam, Sriram<br />

9002, 9023, 9063, 9065,<br />

9127, e19528, e19599,<br />

e19602<br />

Yeo, Wee-Lee 7523<br />

Yeo, Winnie 4103, e14528<br />

Yepes, Ethel 2589<br />

Yerasuri, Durga e14163<br />

Yermilov, Irina 6057<br />

Yerushalmi, Rinat 548<br />

Yetisyigit, Tarkan e14526<br />

Yeung, Alex 4593<br />

Yeung, Annie e12017<br />

Yeung, Choh L 9511<br />

Yeung, S. J. 6125<br />

Yi, Anthony 2080<br />

Yi, Jing 5054<br />

Yi, Min 1028, 1130<br />

Yi, Nengjun 1561<br />

Yi, Sandy TPS6636<br />

Yildirim, Asif e15122<br />

Yildiz, Ferah e15572,<br />

e15573<br />

Yildiz, Ozcan e21143<br />

Yildiz, Ramazan 638<br />

Yilmaz, Bahiddin e14107<br />

Yilmaz, Emrullah 10612<br />

Yilmaz, Mette K. N. 3548,<br />

e14517<br />

Yilmaz, Ugur 3565, e20514<br />

Yim, Barbara 9085<br />

Yim, John H 1035, 1117<br />

Yim, Vincent 10003<br />

Yim, Yeun Mi e11051,<br />

e14068, e16539<br />

Yin, Wei e13041<br />

Ying, Weiwen 3086<br />

Ying, Wendy e19514<br />

Yip, Desmond e13007,<br />

e14703<br />

Yip, George W e21041<br />

Yip, Sonia TPS4145<br />

Yip, Stephen 3628<br />

Yock, Torunn I. 9585<br />

Yoda, Satoshi e18058<br />

Yoh, Kiyotaka 1552, 7044,<br />

e17513, e17526, e17554,<br />

e18054, e18064<br />

Yohji, Itoh 1534<br />

Yokoi, Kohei 7021<br />

Yokoi, Takashi e18081<br />

Yokosuka, Osamu e14622<br />

Yokota, Soichiro e18025<br />

Yokoyama, Akira 6112, 7003,<br />

7017, 7028, 7515,<br />

e19556<br />

Yokoyama, Ryohei 9574<br />

Yokoyama, Takuma e19556<br />

Yom, Cha Kyong 1056<br />

Yonas, Bekuretsion 580<br />

Yoneda, Kazue 7080, 10585<br />

Yonemori, Kan 10519,<br />

e19535<br />

Yonese, Junji e15038<br />

Yoneyama, Kimiyasu 585<br />

Yong, Candice e15148<br />

Yong, Kol Jia e13505<br />

Yong, Kwee e18572^<br />

Yong, Wei-Peng 2551, 3002,<br />

4100^, e13002<br />

Yong, William H. 2101<br />

Yong-Hing, Charlotte Jane<br />

e14565<br />

Yoo, Changhoon 10085,<br />

e11508<br />

Yoo, George H. 5534,<br />

e16019<br />

Yoo, Simon 8579<br />

Yook, Jeong Hwan 10093<br />

Yoon, Chan Seok 1053<br />

Yoon, Dok Hyun 4093, 5586,<br />

e13104<br />

Yoon, Harry H. 4009, 4075<br />

Yoon, Hwi Joong e14128<br />

Yoon, Hyungeun e18126<br />

Yoon, Jeong Lim 1105,<br />

e19029<br />

Yoon, Jong Hyung 9579<br />

Yoon, Jung-Hwan 4103<br />

Yoon, Ki Tae e14566<br />

Yoon, Kyong-Ah e18119<br />

Yoon, Sam S. 10058<br />

Yoon, Sang Min e18501<br />

Yoon, Sung-Soo e18572^<br />

Yoong, Jaclyn 9101<br />

York, Sergernne 6501<br />

Yoshida, Atsushi 590,<br />

e13037<br />

Yoshida, Chikashi 8094<br />

Yoshida, Kazuhiro 3558,<br />

3607<br />

Yoshida, Kimihide e17510,<br />

e18145<br />

Yoshida, Koji e13014,<br />

e13015, e13029, e13037<br />

Yoshida, Kozue e19611<br />

Yoshida, Masahiro e19583<br />

Yoshida, Motoki 3088^<br />

Yoshida, Shigetoshi 7046<br />

Yoshida, Takahiro e15065<br />

Yoshida, Takashi 613<br />

Yoshida, Tatsuya e18054<br />

Yoshida, Tsukihisa e13090<br />

Yoshida, Yoshio e15539<br />

Yoshii, Akihiro e18037<br />

Yoshikawa, Hiroyuki 5006<br />

Yoshikawa, Takaki 4070<br />

Yoshikawa, Takayuki e21007<br />

Yoshimori, Kouzou e19556<br />

Yoshimura, Kiyoshi e13559<br />

Yoshimura, Naruo 7017,<br />

e17538<br />

Yoshino, Ichiro 7046<br />

Yoshino, Kunitoshi e16032<br />

Yoshino, Reiko e18037<br />

Yoshino, Takayuki 3079,<br />

3502, e14038<br />

Yoshino, Tomoko e21007<br />

Yoshino, Yuki e19611<br />

Yoshioka, Hiroshige 6112,<br />

7521, 7528, e18025,<br />

e18134<br />

Yoshioka, Masakata e18019<br />

Yoshioka, Toshiaki e15065,<br />

e15069<br />

Yoshioka, Yasuko e18019<br />

Yoshiya, Shohei e14536,<br />

e14602<br />

Yoshizawa, Carl N. 549^,<br />

1021<br />

Yoshizawa, Carl 1005<br />

Yoshizumi, Tomoharu<br />

e14536, e14602<br />

Yost, Kathleen J. 1610<br />

Yothers, Greg 3512, 3522,<br />

3523, 3525, 3545<br />

You, Benoit 5110, 8068,<br />

e15182<br />

You, Jee Man 1133, e21160<br />

You, John J. 6049<br />

You, Si Young e14558<br />

You, Y. Nancy 3556, 4060<br />

You, Zhiying e13087<br />

Younan, Rami 9138<br />

Younas, Mariam e18510<br />

Younes, Anas 8067, 8079<br />

Younes, Mamoun e15003<br />

Young, Anne Mary 2530<br />

Young, Brandon F 1027<br />

Young, Brian 6108<br />

Young, Debra R. 2021<br />

Young, Elliana J. 527, 10613<br />

Young, Lillian L 4575<br />

Young, Mary Elizabeth 1128<br />

Young, Nancy 6103<br />

Young, Richard J. 5571,<br />

8520<br />

Young, Robyn R. TPS1137<br />

Young, Ruth T. e14503<br />

Young, Tracy e11562<br />

Youngson, Bruce 1123<br />

Younos, Ibrahim 2511<br />

Yousef, George M. 4613, 4633<br />

Yousif, Nasser Ghaly e11046<br />

Yousif, Saif e11053<br />

Youssef, Youssef M. 4633<br />

Youssoufian, Hagop TPS1143,<br />

e13040<br />

Yovine, Alejandro Javier<br />

TPS1148<br />

Yovino, Susannah G. 2017<br />

Ysebeart, Loic 8048<br />

Yu Min Lee, Ursula Joan 3515<br />

Yu, Ba<strong>of</strong>a e14716<br />

Yu, Bin 8582<br />

Yu, Chang Sik 3568, 10093<br />

Yu, Changhong 4552, 4652,<br />

5562, e15070<br />

Yu, Chih-Chia e16008<br />

Yu, Chong-Jen 7519^,<br />

TPS7614<br />

Yu, Choon Geok 4106<br />

Yu, Ding e18033<br />

Yu, Elaine e11051, e14068,<br />

e16539<br />

Yu, Evan Y. 4506, 4513,<br />

4514, 4557, 4654,<br />

e15206<br />

Yu, Gang e18142<br />

Yu, Hao 7548<br />

Yu, Helena Alexandra 3083,<br />

7052, 7527, 7580<br />

Yu, James B. 1030, 4651,<br />

6019, e15150<br />

Yu, Jiaye e21011<br />

Yu, Jie-Kai e14026<br />

Yu, Jinming 4073, e21115<br />

Yu, John 2080, 2084, 2087<br />

Yu, Kenneth H. 4057, 9105<br />

Yu, Kim Wai 1035<br />

Yu, Kwok Hung e15110<br />

Yu, Lei e17542<br />

Yu, Lixia 4068, e14509,<br />

e21034<br />

Yu, Mandi 1536<br />

Yu, Margaret K. TPS666<br />

Yu, Menggang e16010,<br />

e21016<br />

Yu, Mimi C 4575<br />

Yu, Minshu 2545<br />

Yu, Qitao 5538<br />

Yu, Ren 6122<br />

Yu, Rong e11068<br />

Yu, Seung Shin e19574<br />

Yu, Shi Ying 5097, e18100<br />

Yu, Simon e14528<br />

Yu, Wei TPS7616<br />

Yu, Xiang-Qing TPS2618<br />

Yu, Xinhua 7025, 7069<br />

Yu, Xinmin e17512<br />

Yu, Xinshuang e14643<br />

Yu, Xujie 6522<br />

Yu, Yanhong e15563<br />

Yu, Yuefei e21105<br />

Yu, Zheng e13121<br />

Yuan, Constance 8088,<br />

e18568<br />

Yuan, Dongmei e18082,<br />

e18116<br />

Yuan, Jianda 2518, 2521,<br />

8575, 8583<br />

Yuan, Jinyu e15070<br />

Yuan, Ma Dai e14648<br />

Yuan, Peng TPS1135,<br />

e11025, e11538<br />

Yuan, Shuanghu e21115<br />

Yuan, Shuqiang e14674<br />

Yuan, Ying e14024, e14026<br />

Yuan, Zheng 2516<br />

Yuasa, Miyuki 3002<br />

806s Numerals refer to abstract number


Yuasa, Takeshi 4536, 4538,<br />

e15038, e15098<br />

Yuce, Deniz 9050<br />

Yucel, Birsen e11083,<br />

e16005, e19575<br />

Yucel, Idris e14107<br />

Yue, Binglin 7065, e17559<br />

Yue, Guang H. 1076<br />

Yue, Huibin 8579, 8585<br />

Yuen, Eunice 3008<br />

Yuen, Kwok Keung 5511<br />

Yuki, Satoshi 3593, e14038,<br />

e14062<br />

Yukisawa, Seigo e14677<br />

Yule, Murray 9542<br />

Yumuk, Fulden e16015<br />

Yun, Hwan Jung e14529<br />

Yun, Jeong H 536<br />

Yun, Jina e14688, e19554<br />

Yun, Lingsong e14003<br />

Yun, Mi Jin e14559<br />

Yun, Rui 8087<br />

Yun, Ya-Lin 6038<br />

Yun, Young Ho 9066<br />

Yunes Ancona, Alejandro 5502<br />

Yung, W. K. Alfred 2003,<br />

2029^, 2030, 2036, 2064<br />

Yunokawa, Mayu 10519,<br />

e11065, e19535<br />

Yunus, Furhan 5500, 5503<br />

Yusuf, Anny 2062<br />

Yvonnet, Vanessa e15161<br />

Z<br />

Zaanan, Aziz 4018<br />

Zabel, Brian e21071<br />

Zabel, Frauke 554<br />

Zabel, Maciej 10537<br />

Zabkowska, Katarzyna e21099<br />

Zaborek, Piotr<br />

Zacarias Föhrding, Luisa<br />

9046<br />

Zaccarelli, Eleonora<br />

e21020<br />

e11039,<br />

e11514, e12510, e19620<br />

Zach, Leor 2078, e14143<br />

Zachee, Pierre TPS6640<br />

Zadnik, Vesna 558<br />

Zafar, Faiza 8036<br />

Zafar, Syed Farhan e14692<br />

Zafar, Yousuf 6102<br />

Zaffaroni, Nadia 4507<br />

Zagaya, Ciara 4022<br />

Zage, Peter E. 9534<br />

Zagel, Miriam e17516<br />

Zager, Jonathan S. 10048<br />

Zagonel, Vittorina 586, 2049,<br />

3555, 3585, 4115,<br />

e12037, e14040, e14563<br />

Zagst, Patricia D. 3029,<br />

e14690<br />

Zahaf, Katia 7591<br />

Zahlmann, Gudrun TPS9596<br />

Zahnow, Cynthia A. e18147<br />

Zahurak, Marianna TPS3117<br />

Zaia, John 8089, e18542<br />

Zaidi, Aisha Salman e17019<br />

Zaidi, Ali H e14689<br />

Zaidi, Bushra 8583<br />

Zain, Jasmine M. e13079<br />

Zairi, Fahed 2070<br />

Zaja-Milatovic, Snjezana 6038<br />

Zajac-Kaye, Maria TPS4136<br />

Zajchowski, Deborah A. 5074,<br />

e15555<br />

Zajda, Katarzyna e12011<br />

Zakari, Ahmed 4127<br />

Zakeri, Kaveh 6121<br />

Zakharova, Tatiana e15025,<br />

e15029<br />

Numerals refer to abstract number<br />

Zaki, Bassem I. e14514<br />

Zaki, Mohamed H. 8016<br />

Zaknoen, Sara L. 6577, 7546<br />

Zakotnik, Branko 558<br />

Zakowski, Maureen Frances<br />

10560<br />

Zakrzewski, Sandra e13121<br />

Zalcberg, John Raymond<br />

3504, 3515, 3572,<br />

TPS4141, TPS4145<br />

Zalcberg-Renault, Ilana<br />

e14101<br />

Zalcman, Gerard 3000, 7057,<br />

TPS7112, 7574, e16560,<br />

e18089<br />

Zalewski, Pawel e15017<br />

Zalis, Mariano e14101<br />

Zalles, Carola M. 518, 520<br />

Zalupski, Mark M. 4039,<br />

e14519<br />

Zamagni, Claudio 532, e13057<br />

Zamagni, Elena e21006<br />

Zaman, Khalil 9059, e11048<br />

Zamba, Gideon e14620<br />

Zambetti, Milvia 545<br />

Zamboni, Beth A. 5050<br />

Zamboni, William 2591,<br />

5050, e13063<br />

Zambrana Tevar, Francisco<br />

1570, 9073, e11028<br />

Zambrowicz, Brian 4085,<br />

e14564<br />

Zamechek, Leah B. e21082<br />

Zamek-Gliszczynski, Maciej<br />

3008<br />

Zamora, Esther 7041<br />

Zamora, Pilar e11081<br />

Zamora, Victor e14598<br />

Zampa, Germano 4572,<br />

e14082<br />

Zampino, M. Giulia e19036<br />

Zamudio Osuna, Michelle<br />

e15525<br />

Zanaboni, Flavia 5096<br />

Zanconati, Fabrizio 10554<br />

Zander, Dani 7054<br />

Zander, Thomas 1533,<br />

TPS3115, 7603, 10526,<br />

CRA10529<br />

Zandstra, Fran e19625,<br />

e19629<br />

Zanetta, Sylvie LBA4567^,<br />

5505, e13009<br />

Zang, Dae Young TPS4142,<br />

e14572, e14580<br />

Zang, Qingqiao e13004<br />

Zanghì, Mariangela e11056<br />

Zangos, Stephan e14131<br />

Zaniboni, Alberto 1073,<br />

3555, e14018<br />

Zanichelli, Maria Aparecida<br />

6509^<br />

Zannella, Vanessa e15118<br />

Zanoni, Vanessa e11546<br />

Zanotti, Kristine M. 5089<br />

Zanotti, Laura e15549<br />

Zanus, Giacomo e14563<br />

Zanzonico, Pat TPS9595<br />

Zaorsky, Nicholas George<br />

e19507<br />

Zapas, John L. LBA1000<br />

Zapka, Jane 6086<br />

Zappa, Magaly 4133, e14660<br />

Zappala, Stephen M e15196<br />

Zarba, Juan Jose e14521<br />

Zaren, Howard e16561<br />

Zaritskey, Andrey 6512<br />

Zarotsky, Victoria e17017<br />

Zarour, Hassane M. TPS8606<br />

Zastrow, Leonhard 5064,<br />

e19558<br />

Zastrow, Stefan e15195<br />

Zatloukal, Petr LBA7000,<br />

7575, e18062<br />

Zaucha, Jan M. 8030, 8077<br />

Zauderer, Marjorie Glass 1541<br />

Zaugg, Kathrin 3595<br />

Zaun, Silke e11040^,<br />

e18016^<br />

Zavadil, Jiri 8544, 8565,<br />

9507, e19018<br />

Zavaglia, Katia 629<br />

Zavala, Diego G e18028<br />

Zavala, Jose D 8075<br />

Zavala, Laura 9034, 9109,<br />

9123<br />

Zaveleva, Elina 3070<br />

Zaveri, Shabber e14710,<br />

e15516<br />

Zavos, Apostolos 5066<br />

Zawam, Husam e18001<br />

Zayas, Myriam 9565<br />

Zazo, Sandra 5520, e14147,<br />

e15094, e18069, e18106,<br />

e21015<br />

Zazpe, Cruz e14120, e21086<br />

Zazulina, Victoria 7503<br />

Zbieranowski, Ingrid J. 4613<br />

Zbuk, Kevin M. e14001<br />

Zbynek, Bortlicek e11072,<br />

e18062<br />

Zdzienicki, Marcin 8532,<br />

8548<br />

Zebary, Abdlsattar 8588<br />

Zebracka, Jadwiga e14159<br />

Zechmeister, Jenna R.<br />

e15548<br />

Zee, Benny C.Y. 4105, 5511,<br />

e14706<br />

Zee, Ying Kiat 2551, 3002,<br />

4100^<br />

Zeger, Erik L. e14157<br />

Zeger, Gary 7582, 7594,<br />

e12007<br />

Zeh, Herbert e14184<br />

Zehr, Pamela e15010<br />

Zeidan, Amer e16537<br />

Zeillinger, Robert 5034,<br />

5058, 5071<br />

Zeimet, Alain G 5034, 5058<br />

Zeis, Matthias 8004<br />

Zeitouni, Nathalie e19041<br />

Zeitzer, Jamie 9051<br />

Zelayas, Pedro Guillermo<br />

e15556<br />

Zeldenrust, Steven R. 8096,<br />

8105, TPS8116<br />

Zelenetz, Andrew David 8054,<br />

8069, e18530<br />

Zelenka, Peter e14087<br />

Zelko, Frank 9532<br />

Zellars, Richard C. 1128<br />

Zellmann, Klaus 3588<br />

Zemanova, Milada e14631,<br />

e18062<br />

Zemoura, Leila e18102<br />

Zen, Yoh e21091<br />

Zeng, Xiangqun 628<br />

Zeng, Xiao e21011<br />

Zeng, Xuemei e14689<br />

Zeng, Zhihong TPS6635<br />

Zengerling, Friedemann 4530<br />

Zengin, Nurullah 3565,<br />

e11011, e11033, e11093,<br />

e13031<br />

Zenka, Roman M e15132<br />

Zenz, Thorsten 6519<br />

Zerche, Peter 580<br />

Zeringue, Angelique 4594<br />

Zernovak, Oleg TPS6638<br />

Zeuli, Massimo e14661<br />

Zevon, Michael 1529<br />

Zeynalova, Samira 8024, 8025<br />

Zha, Yuanyuan 2561, 2582<br />

Zhai, Yirui e14511<br />

Zhan, Ping 4124<br />

Zhang, Alicia 5502<br />

Zhang, Baohui 9102<br />

Zhang, Bin 3010, TPS8109<br />

Zhang, Chen-ping 5593<br />

Zhang, Chong 555, 563,<br />

6063, e13537, e16010<br />

Zhang, David Y. 8544<br />

Zhang, Deng e21133<br />

Zhang, Gu e21080<br />

Zhang, Helong 7559<br />

Zhang, Hong 1029, 1104,<br />

e19631<br />

Zhang, Hong-Tu e14502<br />

Zhang, Hua 3546, 3597,<br />

e11018<br />

Zhang, Hui 7530, 7590,<br />

7592, TPS7618, e18142<br />

Zhang, Jian 3038, e11567,<br />

e11568<br />

Zhang, Jiandong e14643<br />

Zhang, Jianjun 7047<br />

Zhang, Jingbo 4542<br />

Zhang, Jinghui 9518<br />

Zhang, Jingsong e13564<br />

Zhang, Joshua 4132, 4501,<br />

e15082<br />

Zhang, Jun e17547<br />

Zhang, Kathy 3581<br />

Zhang, Lan-jun e18121<br />

Zhang, Lei e13069, e14700<br />

Zhang, LI 5524, 5538, 7091,<br />

7519^, 7520, 7548, 7559,<br />

e13021, e13036<br />

Zhang, Liang e18557<br />

Zhang, Ling 6588, e18115,<br />

e18503, e18506, e21123<br />

Zhang, Lu 6567<br />

Zhang, Nan e21051<br />

Zhang, Ning 600, 9091<br />

Zhang, Penglie e13580<br />

Zhang, Peter e11068<br />

Zhang, Pin e11025, e11538<br />

Zhang, Qian Nan 8561<br />

Zhang, Qian e13516<br />

Zhang, Qiang 5530, 5583<br />

Zhang, Qing 5553, e18548<br />

Zhang, Quanwu e15193<br />

Zhang, Qunling e11568<br />

Zhang, Shanwen 10045<br />

Zhang, Sheng e15132<br />

Zhang, Shucai 7520, 7559,<br />

e18142<br />

Zhang, Suzhan e14024<br />

Zhang, Tao e18033, e19603,<br />

e19613<br />

Zhang, Tong e13107<br />

Zhang, Wendy TPS4692^<br />

Zhang, Wu 3518, 3540,<br />

3546, 3549, 3584, 3597,<br />

3604, 3615, 3622, 4026,<br />

4088, 4096, e11018,<br />

e14031, e14034, e14052<br />

Zhang, Xi 8596, e18033<br />

Zhang, Xiang-Hua e18050<br />

Zhang, Xianglan e18092<br />

Zhang, Xiao 5032<br />

Zhang, Xiaodong e14604<br />

Zhang, Xiaohua 9017<br />

Zhang, XiaoJing 4628<br />

Zhang, Xiaotian e14604<br />

Zhang, Xiaotun 4669<br />

807s


Zhang, Xinmin e16022<br />

Zhang, Xu Dong 8553<br />

Zhang, Xu-Chao e13510,<br />

e18035<br />

Zhang, Xuchao 7039, e18090<br />

Zhang, Ya 515<br />

Zhang, Yanwei e17551,<br />

e18038<br />

Zhang, Yi 561, 10588,<br />

e21019<br />

Zhang, Yi-Fang e13510<br />

Zhang, Ying e21111<br />

Zhang, Yingjian 10567<br />

Zhang, Yingjie e13524,<br />

e13529<br />

Zhang, Yingzi e21080<br />

Zhang, Yiping 3031, 7091,<br />

7520, 7548, 7559,<br />

e17512<br />

Zhang, Yixiang 10029<br />

Zhang, Yong TPS1148,<br />

TPS4147, e14700, e16054<br />

Zhang, Yongping 10567<br />

Zhang, Yu e14648<br />

Zhang, Yufeng 600<br />

Zhang, Yuji e15132<br />

Zhang, Yujian e15124<br />

Zhang, Zhe 10509<br />

Zhang, Zheng TPS5114,<br />

TPS10635<br />

Zhang, Zhi-Yong e18077<br />

Zhang, Zhi-yuan 5593<br />

Zhang, Zhigang 5537, 8054<br />

Zhang, Zhong-guo e14036<br />

Zhao, Binsheng 2541,<br />

e13064, e19647<br />

Zhao, Chong 5535, e13021<br />

Zhao, Dave e16060<br />

Zhao, Fei 10566<br />

Zhao, Fengmin 6076, 9016,<br />

9099, 9106, 9112,<br />

e15173<br />

Zhao, Hong 2098, 4616<br />

Zhao, Hongyun e13021<br />

Zhao, Hui 7036, e21103<br />

Zhao, Huijun 10620<br />

Zhao, Jinxiu e16561<br />

Zhao, Liping e13021<br />

Zhao, Lujun e14511<br />

Zhao, Ming 2044, e14700<br />

Zhao, Naiqing 4092, e14575<br />

Zhao, Ni 5577<br />

Zhao, Qiang e14662<br />

Zhao, Weiqiang 5566<br />

Zhao, Xiaoying e16039<br />

Zhao, Yingdong 10565<br />

Zhao, Yu e18087<br />

Zhao, Zhongyun e19043,<br />

e19044, e19045, e19060<br />

Zhenfu, Fu 5568<br />

Zheng, Chunlei e11568<br />

Zheng, Di e18077<br />

Zheng, Hao W. 9543<br />

Zheng, Lei 4045, e14585<br />

Zheng, Leizhen 9017<br />

Zheng, Min 1540<br />

Zheng, Ping TPS657<br />

Zheng, Rongsheng 9017<br />

Zheng, Shu e14026<br />

Zheng, Wang fang 5568<br />

Zheng, Wei 9017<br />

Zheng, Yong-Qing e17512,<br />

e18142<br />

Zheng, Yuanda e17527<br />

Zheng, Zhao-xu e14668<br />

Zhi, Jianguo e13600^<br />

Zhi, Xin e15116<br />

Zhi, Xiuyi 7520<br />

Zhimin, Shao e11567<br />

Zhong, Dafang e13036<br />

Zhong, Fengming e14124,<br />

e14125, e14727<br />

Zhong, Hua e21001<br />

Zhong, Lai-ping 5593<br />

Zhong, Lixian e15154<br />

Zhong, Liz 3098<br />

Zhong, Mei e15563<br />

Zhong, Runbo e21001<br />

Zhong, Wenzhao 7039,<br />

e18090<br />

Zhong, Xiaoyin 2021<br />

Zhong, Yunshi e14000<br />

Zhong, Zhandong Don 5038<br />

Zhou, Ai-Ping 4628, e14668<br />

Zhou, Caicun 7519^, 7520,<br />

7535, 7557, 7559, 7600,<br />

10527, e18100, e18123<br />

Zhou, Chen e14568<br />

Zhou, Cong e21037<br />

Zhou, Dan 3086<br />

Zhou, Daohong e18576<br />

Zhou, Fei e14539<br />

Zhou, Gang 2513<br />

Zhou, He 4056<br />

Zhou, Hong 2503^<br />

Zhou, Jenny e11549<br />

Zhou, Jianying 7559<br />

Zhou, Jun e14122, e14604<br />

Zhou, Lin e14648, e18086<br />

Zhou, Lisa Ya 10574<br />

Zhou, Min 10567<br />

Zhou, Qian 3594<br />

Zhou, Qin 5030, 5108<br />

Zhou, Qing 7039<br />

Zhou, Qinghua e17514<br />

Zhou, Songwen 7520<br />

Zhou, Tianni 9504<br />

Zhou, Weiping 4008<br />

Zhou, Xi Kathy 10514<br />

Zhou, Xian C 10509<br />

Zhou, Xiangdong 2013<br />

Zhou, Xiao 4037, e19520<br />

Zhou, Xia<strong>of</strong>ei 2597, 5021<br />

Zhou, Xiaolei 6626^<br />

Zhou, Yang TPS669,<br />

TPS1614<br />

Zhou, Yangsheng 8042,<br />

8106, 8107<br />

Zhou, Yi e14502<br />

Zhou, Yunping e18560<br />

Zhou, Zheng-Yi 1037<br />

Zhou, Zhiwei e14674<br />

Zhou, Zu li e21103<br />

Zhu, Andrew X. 4021, 4112,<br />

TPS4146, e14615<br />

Zhu, Dexiang e14000<br />

Zhu, Fang 1517<br />

Zhu, Guang ying 4073<br />

Zhu, Guopei e16039<br />

Zhu, Han-guang 5593<br />

Zhu, Hong-Xia e14502<br />

Zhu, Jay-Jiguang TPS2107<br />

Zhu, Jian-fei e18121<br />

Zhu, Jiang e14648<br />

Zhu, Jin TPS2616, 3021<br />

Zhu, Jingqiang 5550<br />

Zhu, Jinxian e16054<br />

Zhu, Joy 2579<br />

Zhu, Jun TPS8118<br />

Zhu, Kangshun 4008<br />

Zhu, Ling e17009<br />

Zhu, Liting 501, e11005<br />

Zhu, Min 2535, 2594<br />

Zhu, Pei Hong 10622<br />

Zhu, Peixuan 6581, e21079<br />

Zhu, Qian 3002<br />

Zhu, Qingfeng e14538<br />

Zhu, Shu chai 4073<br />

Zhu, Xiaolei e15140<br />

Zhu, Yanrong 6016<br />

Zhu, Yongxue e16039<br />

Zhu, Yuan e14049<br />

Zhu, Yue 1039<br />

Zhu, Yunzhong 7519^<br />

Zhu, Yuping e14049<br />

Zhuang, Sen Hong e13122<br />

Zhuang, Tingting 3095<br />

Zhuang, Xusheng e14523<br />

Zhuang, Zhixiang 9017<br />

Zhukovsky, Donna S. 9096,<br />

9134, e19528, e19584<br />

Zhukovsky, Eugene 8059,<br />

e18532<br />

Zhuo, Minglei 7537, 7545,<br />

e18022, e18035<br />

Ziani, Leslie e19046<br />

Zibert, John Robert e19056<br />

Zichi, Dom e21142<br />

Zidan, Abdel-Aziz 9565<br />

Zidan, Jamal e14025<br />

Zidan, Marwan 9578<br />

Ziebarth, Angela 5036<br />

Ziegelmann, Matthew e15211<br />

Ziegenfuss, Jeanette Y. 1565,<br />

4657<br />

Ziegler, Mark A. 4055<br />

Ziegler, Regina G 1521<br />

Zielinski, Christoph 2053,<br />

TPS7113, e15083,<br />

e15090, e21033<br />

Zielonka, Tania e18507<br />

Ziemendorff, Gabriele 10540<br />

Ziepert, Marita 8022, 8025<br />

Zighelboim, Israel 5013,<br />

5101<br />

Zijlstra, Andries e21090<br />

Zijlstra, Jose M 8039<br />

Zikos, Efstathios 6002, 6090,<br />

7607<br />

Zilembo, Nicoletta 7550,<br />

7581<br />

Zimmerman, Pamela 6108<br />

Zimmerman, Todd M. 8034<br />

Zimmermann, Annamaria<br />

LBA7507, 7554<br />

Zimmermann, Benedikt 4565<br />

Zimmermann, Camilla 9003,<br />

e16567<br />

Zimmermann, Heiner 8030<br />

Zimmermann, Ute 8540<br />

Zimmermann, Uwe 4539<br />

Zingaretti, Costantino 4084<br />

Zingone, Adriana 8088,<br />

8104, e18568<br />

Zinner, Ralph 10607<br />

Zinser, Juan Wolfgang e11016<br />

Zintl, Patrik 10013<br />

Zinzani, Pier Luigi 8052,<br />

e13569<br />

Ziparo, Vincenzo e17006<br />

Zippelius, Alfred 2573^<br />

Ziske, Carsten 6058<br />

Zissimopoulos, Athanasios<br />

e17553<br />

Zitt, Matthias e14719<br />

Ziv, Ravit e13018<br />

Zivanovic, Maureen 8056<br />

Zivanovic, Oliver 5090<br />

Zivi, Andrea 4553<br />

Ziviani, Luciene Fabres<br />

e12021<br />

Zlotecki, Robert TPS4136<br />

Zo, Jae Ill e16534, e21043<br />

Zogopoulos, George 4058<br />

Zok, Jolanta e21099<br />

Zola, Fabio Eduardo e12512<br />

Zoli, Wainer 10601, e18045<br />

Zomorodi, Meghan e19644<br />

Zonder, Helene B. e19644<br />

Zonder, Jeffrey A. 6549,<br />

8013, 8020<br />

Zongo, Nayi 641<br />

Zonnenberg, B. A. 4632,<br />

10619<br />

Zoratto, Federica e11039,<br />

e11514, e12510, e19620<br />

Zorludemir, Suzan e11088<br />

Zorman, Darko 558<br />

Zornosa, Carrie C. 4126,<br />

e14542, e14551, e18018<br />

Zorza, Grégoire e13036<br />

Zorzi, Alexandra Patricia 9591<br />

Zorzi, Jane 10509<br />

Zou, Li qun e18514<br />

Zou, Qing 4628<br />

Zou, Shuangmei e14511<br />

Zou, Yiyu e18113<br />

Zou, Zhengyun 4068,<br />

e14509, e21034<br />

Zouvani, Ioanna e15030<br />

Zovato, Stefania 10063<br />

Zrounba, Philippe e16036<br />

Zsiros, Emese 5039<br />

Zu, Maoheng e13004<br />

Zubel, Angela 3003, 8511<br />

Zuber, Markus e14045<br />

Zubkus, John D. 7100<br />

Zubritsky, Lindsey 607,<br />

e14520, e15170<br />

Zucali, Paolo A. 4545<br />

Zucali, Paolo Andrea 2580,<br />

7082, e21139<br />

Zuckerman, Ilene H. 6028,<br />

6060, 6075<br />

Zudaire, Mari-a I. e18008<br />

Zugazagoitia, Jon e20513<br />

Zugmaier, Gerhard 6500^<br />

Zuhlke, Kimberly A. e15205<br />

Zukin, Mauro 7506, 7511<br />

Zulfikar, Bulent 9567<br />

Zulfiqar, Muhammad e14176<br />

Zullig, Leah L. 5539, 6102<br />

Zullo, Marzio Angelo 5094,<br />

e15551<br />

Zuluaga Toro, Tania<br />

TPS4136, e14612<br />

Zulueta, Javier e18008<br />

Zumbo, Paul 9507<br />

Zuo, Zhixiang 5517<br />

Zupo, Simonetta e18021<br />

Zuradelli, Monica 615, 623<br />

Zureik, Mahmoud 1574<br />

Zureikat, Amer H e14184<br />

Zurita, Amado J. 4533, 4556,<br />

e15075, e15085, e15087<br />

Zustovich, Fable 2049,<br />

e12037, e15160<br />

Zvirinska, Lenka e13112<br />

Zwaan, Michel 9517,<br />

TPS9594<br />

Zwahlen, Daniel Rudolf 3595<br />

Zwart, Nynke 4528<br />

Zwarthoed, Colette 8077,<br />

e15574<br />

Zwas, Tzila S. e17509<br />

Zwenger, Ariel e11021<br />

Zwick, Carsten 8022, 8024<br />

Zwingers, Thomas 1072,<br />

1600^, 1602<br />

Zwischenberger, Joseph<br />

Bertram e14562<br />

Zyczynski, Teresa Maria<br />

e15165<br />

Zylberberg, Ricardo e17548<br />

Zylis, Dimosthenis 10606<br />

808s Numerals refer to abstract number


Support for this program is funded through

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!